12.07.2015 Views

Conference Program - ABRF 2011 - Association of Biomolecular ...

Conference Program - ABRF 2011 - Association of Biomolecular ...

Conference Program - ABRF 2011 - Association of Biomolecular ...

SHOW MORE
SHOW LESS
  • No tags were found...

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

Meeting SponsorsThe <strong>Association</strong> <strong>of</strong> <strong>Biomolecular</strong> Resource Facilities (<strong>ABRF</strong>) and the <strong>ABRF</strong> <strong>2011</strong> Organizing Committee wouldlike to extend their gratitude for the generous support received from the following organizations:Meeting SponsorsNetworking Night at the RanchCyber Café Monday Munch & Mingle Meeting BagAward Sponsors<strong>ABRF</strong> Annual Award for Outstanding Contributionsto <strong>Biomolecular</strong> Technologies<strong>ABRF</strong> Poster AwardsMedia Sponsors<strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine • 5


Hotel FloorplanHotel Floorplan6 • <strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine


Registration ServicesRegistration ServicesTicketed EventsFriday through TuesdayLevel 2, Ballroom Level Registration DeskOn–site registration will be open during the following hours:Friday, February 18 ........................................4:00 pm – 6:00 pmSaturday, February 19 ................................... 7:00 am – 6:00 pmSunday, February 20 ...................................... 7:00 am – 6:00 pmMonday, February 21 .................................... 7:00 am – 6:00 pmTuesday, February 22 .................................... 7:00 am – 6:00 pmMeeting RegistrationFull meeting registration includes: food functions served by<strong>ABRF</strong> <strong>2011</strong>, access into the Exhibit Hall, Receptions, ScientificSessions, Workshops, and Research Group Presentation Sessions.Note: All attendees are welcome to attend the Research GroupPresentation Sessions.Activities for which you have purchased tickets or guest ticketsrequire the actual ticket for entrance. Tickets are provided at thetime <strong>of</strong> check-in. Please be sure to check that you received ticketsfor all purchased items. Additional tickets can be purchased atthe On-site Registration Desk.Networking Night at the Ranch (Sponsored by )Guest Ticket ..................................................................................$50Exhibit Hall ONLY RegistrationExhibit Hall Only Registration ..................................................$35An Exhibit Hall Only Registration is available for those whowish to visit the Exhibition only. Exhibit Hall Only Registrationallows access into the exhibits, as well as all Research GroupPresentation Sessions.Registration ServicesOn-site FeesMember ....................................................................................... $510Non-Member ............................................................................ $635Student ........................................................................................ $195Membership + Meeting Registration ................................. $610ReceiptsRegistered participants will receive a receipt/confirmation viaemail. Participants who register on-site will receive a paperreceipt, if requested.Satellite Educational WorkshopsMember ....................................................................................... $325Non-Member ............................................................................ $425Please see page 28 for additional information about eachWorkshop listed below. Satellite Educational Workshops are notincluded in the regular meeting registration. You must purchasea separate registration to attend a specific Satellite EducationalWorkshop. Session Organizers will be checking badges for allSatellite Educational Workshops.• (SW1) Protein Purification for Mass Spectrometry• (SW2) An Introduction to Metabolomics• (SW3) Next Generation Sequencing Considerations ForCore Pr<strong>of</strong>essionals• (SW4) Lean Management in Core FacilitiesBadgesBadges should be worn at all <strong>of</strong>ficial functions <strong>of</strong> the meeting.Badge checkers will be stationed throughout the hotel. Onlythose with full meeting registrations will be allowed into sessions.If you forget or lose your badge, you may obtain a second badgeat registration with pro<strong>of</strong> <strong>of</strong> registration.<strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine • 7


Meals & RefreshmentsMeals& RefreshmentsMeeting registration includes all meals, receptions, andrefreshments provided by <strong>ABRF</strong> <strong>2011</strong>, as well as one ticket tothe Networking Night at the Ranch (sponsored by ).Opening ReceptionSaturdayLevel 1, Exhibit Ballroom Foyer7:00 pm – 9:00 pmContinental BreakfastSunday through TuesdayLevel 1, Exhibit Ballroom Foyer7:00 am – 7:45 amMorning Refreshment BreakSunday through TuesdayLevel 2, Grand Oaks Ballroom, Exhibit Hall10:30 am – 11:00 amMunch & Mingle (Lunch)Sunday through TuesdayLevel 2, Grand Oaks Ballroom, Exhibit Hall12:00 pm – 1:30 pmAfternoon Refreshment BreakSunday through MondayLevel 2, Grand Oaks Ballroom, Exhibit Hall4:15 pm – 4:45 pmTuesdayLevel 2, Grand Oaks Ballroom N/S Foyer4:15 pm – 4:45 pmNetworking Night at the Ranch (sponsored by )SundayOffsite: Leon Springs Dance Hall7:30 pm – 10:30 pmShuttles to Leon Springs Dance Hall will be provided to all <strong>ABRF</strong><strong>2011</strong> Networking Night attendees. Shuttles will begin loadingpromptly at 7:00 pm to depart from the Exhibit Level Lobby.Shuttles to return to the JW Marriott San Antonio Hill CountryResort will also be provided. Please review the shuttle schedulefor further information.Closing ReceptionTuesdayJW Marriott San Antonio Hill Country Resort,Grand Ballroom Terrace7:00 pm – 9:00 pmClose out <strong>ABRF</strong> <strong>2011</strong> with the rest <strong>of</strong> your colleagues at the JWMarriott’s Grand Ballroom Terrace. Reminisce about the week’sevents with fantastic cocktails and hors d’oeuvres.8 • <strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine


Presenter InformationPresenter InformationSpeaker Ready RoomSaturday through TuesdayLevel 2, Ballroom Level Registration DeskALL Presenters must check-in at the Speaker Ready Room at least2 hours prior to their scheduled presentation. Even if you havesubmitted your presentation in advance and have no changes,you must check and confirm that the presentation is correct.The hours <strong>of</strong> operation <strong>of</strong> the Speaker Ready Room are asfollows:Saturday, February 19 ................................... 7:00 am – 4:00 pmSunday, February 20 ...................................... 7:00 am – 4:00 pmMonday, February 21 .................................... 7:00 am – 4:00 pmTuesday, February 22 .................................... 7:00 am – 4:00 pmYou may also edit your presentation at this time. When youare finished reviewing your presentation and verify that it isready, the AV personnel will queue your presentation ontothe networked conference computers. The file will then betransferred to the computer network for presentation in thescheduled room.Meeting ManagementDonna Johnson, Strategic Account DirectorLauren Deaton, Meeting ManagerChris Cherkis, Exhibits & Sponsorship ManagerChezka Solon, <strong>Program</strong> & Registration Manager<strong>ABRF</strong> 2010c/o Courtesy Associates2025 M Street, NWSuite 800Washington, DC 20036Phone: +1 202-973-8670Fax: +1 202-331-0111Email: abrf@courtesyassoc.comPoster PresentationsSunday through TuesdayOver 140 posters will be on display all day Sunday throughTuesday. The Poster Presenters should be available for questionsat their display boards during the specific session times. PosterSession times are as follows:Poster Session I (Odd Numbered Posters)Sunday, February 20Level 2, Grand Oaks Ballroom, Exhibit Hall & Foyer10:30 am – 12:00 pmPoster Session II (Even Numbered Posters)Monday, February 21Level 2, Grand Oaks Ballroom, Exhibit Hall & Foyer10:30 am – 12:00 pmPoster Session III (All Posters)Tuesday, February 22Level 2, Grand Oaks Ballroom, Exhibit Hall & Foyer10:30 am – 12:00 pmOdd numbered posters will be presented during Poster SessionI. Even numbered posters will be presented during Poster SessionII. Please see page 60 for the full listing <strong>of</strong> poster presentations.Poster Session III on Tuesday, February 22 is a free period forattendees to review all posters.All posters should be set up at least one hour prior to theopening <strong>of</strong> the Exhibit Hall on Sunday, February 20. Posterpresenters have been given a special pass to allow them accessinto the Exhibit Hall during these hours. If you are missing thispass, please visit the Registration Desk to receive the access pass.All posters must be removed from the Exhibit Hall between thehours <strong>of</strong> 2:00 pm – 3:00 pm on Tuesday, February 22. Do notleave your presentation on the poster board. Posters remainingafter 3:00 pm on Tuesday will be discarded.10 • <strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine


www.454.comSequence-Enrichment SolutionGS Junior andSequence CapturePerform targeted resequencing <strong>of</strong> any genomic region in your own labby combining two new Roche technologies: NimbleGen EZ ChoiceSequence Capture custom probes and the GS Junior Instrument.Use an easy, scalable workflow for sequence enrichment —Choose a simple workflow featuring gel-free sequencing librarypreparation, single-tube capture with stable DNA probes, and easyhybridization setup.Reduce sequencing costs — Multiplex sequencing samples withmolecular barcodes to maximize data output per run.To learn more, visit www.454.com Figure 1: The GS Junior Sequence Capture Protocol.1. The genomic DNA sample is fragmented and polished.2. Linkers are ligated to the fragments.3. The sample is hybridized to a library <strong>of</strong> DNA capture probes.4. Unbound fragments are washed away.5. The captured genomic DNA (bound to beads) is used directly as PCR template.6. The amplified, enriched sample is ready for high-throughput sequencing withthe GS Junior Instrument.For life science research only.Not for use in diagnostic procedures.454, 454 SEQUENCING, NIMBLEGEN, and GS JUNIOR are trademarks <strong>of</strong> Roche.© <strong>2011</strong> Roche Diagnostics. All rights reserved.Roche Diagnostics CorporationRoche Applied ScienceIndianapolis, Indiana


Awards<strong>ABRF</strong> Annual Award for Outstanding Contributions to <strong>Biomolecular</strong> TechnologiesPresentation & LectureSponsored by Agilent TechnologiesMonday, February 21Level 1, Exhibit Ballroom C3:15 pm – 4:15 pmAwardsThe <strong>ABRF</strong> Executive Board is pleased to announce that therecipient <strong>of</strong> the <strong>2011</strong> <strong>ABRF</strong> Annual Award for OutstandingContributions to <strong>Biomolecular</strong> Technologies is Sir Alec JohnJeffreys.Biological research is driven by technology.As new instruments and chemistries areconceived and implemented, new frontiersare created and new possibilities emerge.Technology, from the invention <strong>of</strong> thelight microscope to the deciphering <strong>of</strong>the Human Genome, opens doors intothe unknown that otherwise remain firmlysealed. The <strong>ABRF</strong> Award recognizes those pioneers responsiblefor developing these powerful new tools that serve as thefoundation <strong>of</strong> the modern biological research enterprise. The<strong>ABRF</strong> Award is sponsored by Agilent Technologies and ispresented at the annual <strong>ABRF</strong> meeting.About Sir Alec John JeffreysPr<strong>of</strong>. Sir Alec Jeffreys studied biochemistry and genetics atMerton College, Oxford. He then began an EMBO PostdoctoralFellowship at the University <strong>of</strong> Amsterdam where, with Dr.Richard Flavell, he was one <strong>of</strong> the first to discover split genes. Hemoved in 1977 to the Department <strong>of</strong> Genetics at the University<strong>of</strong> Leicester where he currently holds the positions <strong>of</strong> Pr<strong>of</strong>essor<strong>of</strong> Genetics and Royal Society Wolfson Research Pr<strong>of</strong>essor.Sir Alec’s research at Leicester has focused on exploring humanDNA variation and the mutation processes that create thisdiversity. He was one <strong>of</strong> the first to discover inherited variationin human DNA, then went on to invent DNA fingerprinting,showing how it could be used to resolve issues <strong>of</strong> identityPast <strong>ABRF</strong> Award Winners1994 Frederick Sanger1995 Klaus Bieman1996 David Lipman1997 Lloyd Smith1998 Bruce Merrifield1999 Marvin Caruthers2000 Leroy Hood2001 Csaba Horváth2002 John Fenn2003 Franz Hillenkamp and Michael Karas2004 Edwin Southern2005 Stephen Fodor2006 Roger Tsien*2007 Don Hunt2008 Ruedi Abersold2009 Mathias Uhlen2010 Patrick O. Brown*also won the 2008 Nobel Prize in Chemistryand kinship. His current work concentrates on developingnew approaches to analyzing variation and mutation in humanchromosomes.Sir Alec’s work has received widespread recognition, includinghis election to the Royal Society in 1986 and a Knighthood forservices to genetics in 1994. Other awards include the Louis-Jeantet Prize for Medicine (2004), the Lasker Award (2005) andthe Heineken Prize (2006). He was also one <strong>of</strong> the four finalistsfor the Millennium Prize in 2008.12 • <strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine


<strong>ABRF</strong> Outstanding Scientist/Technologist Travel AwardsRecipients <strong>of</strong> the <strong>ABRF</strong> Travel Awards are those <strong>ABRF</strong> members who have made outstanding contributions in their institutional corefacilities; have developed new biotechnologies with applications in core facilities; and/or have been active in <strong>ABRF</strong> activities.The <strong>2011</strong> awardees are:Pamela “Scottie” AdamsTrudeau InstituteSaranac Lake, NY, United StatesTaslimarif SaiyedCentre for Cellular and Molecular Platforms,NCBS-TIFRBangalore, IndiaElke Küster-SchöckProteomics and Genomics Coordinator,Microscopy AssociateCIAN (Cell Imaging and Analysis Network),McGill UniversityMontreal, QC, CanadaLaurence SandbergLoma Lind UniversityLoma Linda, CA, United StatesAwardsAnoja PereraStowers Institute for Medical ResearchKansas City, MO, United StatesToni WhistlerChronic Viral Diseases BranchCenters for Disease Control and PreventionAtlanta, GA, United StatesMichelle CiliaUnited States Department <strong>of</strong> Agriculture,Robert W. Holley Center for Agricultureand HealthIthaca, NY, United States<strong>ABRF</strong> <strong>2011</strong> Travel Award Selection Committee:Michelle M. Detwiler (Chair), Roswell Park CancerInstituteDebbie Adam, University <strong>of</strong> British ColumbiaDr. Richard T. Pon, University <strong>of</strong> CalgaryDr. Satya P. Yadav, Cleveland Clinic FoundationResearch Institute<strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine • 13


<strong>ABRF</strong> Robert A. Welch Outstanding ResearchGroup / Committee Member <strong>of</strong> the Year AwardThe <strong>ABRF</strong> Robert A. Welch OutstandingResearch Group/Committee Member <strong>of</strong>the Year Award recognizes the significantcontributions <strong>of</strong> members <strong>of</strong> the <strong>ABRF</strong>Research Groups and Committees. The<strong>2011</strong> award will be presented to JeffKowalak.The Journal <strong>of</strong> <strong>Biomolecular</strong> Techniques ( JBT)AwardThis annual award recognizes the best research article publishedeach year in the Journal <strong>of</strong> <strong>Biomolecular</strong> Techniques, the <strong>ABRF</strong>society publication. This year’s JBT award recipient is:The Comparison <strong>of</strong> Different Pre- and Post-Analysis Filters for Determination <strong>of</strong> Exon-LevelAlternative Splicing Events Using AffymetrixArraysAwards<strong>ABRF</strong> Lifetime Membership AwardThis award is given by the Executive Board to members withlongstanding contributions to the success <strong>of</strong> <strong>ABRF</strong> and whohave been keystones to its missions. The <strong>2011</strong> Award will bepresented to Lowell Ericsson.<strong>ABRF</strong> Poster AwardsSponsored by Waters CorporationWaters Corporation sponsors prizes for the four best researchposters presented at <strong>ABRF</strong> <strong>2011</strong>. Eligible posters focus onthe latest scientific research results enabled by advanced lifesciences technologies, methods and s<strong>of</strong>tware tools that facilitateapplications, and the latest technology developments in thebiotechnology field. This year <strong>ABRF</strong> Education Committeereviewed all poster abstracts submitted to the meeting andselected 11 semifinalists for award consideration.Toni Whistler, Cheng-Feng Chiang, Jin-Mann Lin,William Lonergan, and William C. ReevesChronic Viral Diseases Branch, Division <strong>of</strong> Viral andRickettsial Diseases, Centers for Disease Control andPrevention, Atlanta, GeorgiaPublished in the Journal <strong>of</strong> <strong>Biomolecular</strong> Techniques, 2010 April;21(1)FASEB MARC (Minority Access to ResearchCareers) Travel AwardsThe FASEB MARC <strong>Program</strong> sponsors <strong>ABRF</strong> <strong>2011</strong> Travel Awardsto help support the participation <strong>of</strong> full-time faculty/mentors,postdoctoral fellows, and students from minority institutionsand historically black colleges and universities.The <strong>2011</strong> Faculty/Mentors and Students Travel awardeesare:This year’s semifinalists are:• Mark Turner, Proteome S<strong>of</strong>tware, Inc. (Poster 137)• Christine Vogel, New York University (Poster 143)• Jenny Chuu, Life Technologies (Poster 149)• Jin Jen, Mayo Clinic (Poster 161)• Shripa Patel, Stanford University (Poster 169)• Ilene Magpiong, Mount Holyoke College (Poster 193)• Anand Narayanan, West Virginia University (Poster 197)• J. Will Thompson, Duke Institute for Genome Sciences andPolicy (Poster 199)• Stefan Duhr, University Munich, Schellingstrasse (Poster201)• Keith Fadgen, Waters Corporation (Poster 227)• Emily Freeman, Harvard School <strong>of</strong> Public Health (Poster231)The semifinalists are invited to give a short (5 minute) presentation<strong>of</strong> their research poster in front <strong>of</strong> a panel <strong>of</strong> judges on Sunday,February 20. The authors <strong>of</strong> the four top-scoring posters willpresent their work on Tuesday, February 22 during the WatersCorporation-sponsored Poster Award Session, 11:00 am – 12:00pm, Level 2, Grand Oaks Ballroom, Exhibit Hall, Demo Stage.Semifinalists are designated by ** next to their poster numberwithin the Poster Session Abstract Listing.• Dr. Natalia Vinas (Faculty), Jackson State University• Christina Bernard (Undergraduate Student), Jackson StateUniversity• Charnia Hall (Undergraduate Student), Jackson StateUniversityThe recipients <strong>of</strong> the <strong>2011</strong> Poster Presenter Travel Awardare:• Ilene Magpiong, Mount Holyoke College• Thurman Young, Langston University14 • <strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine


The most accuratenext-gen sequencingtechnology available.Every Illumina sequencer is powered by TruSeq—the technology that delivers the most accuratehuman genome at any coverage. TruSeqproduces the highest yield <strong>of</strong> error-free reads.The most bases over Q30. The greatest number<strong>of</strong> peer-reviewed publications—more than 1,400in the past four years.That’s Tru data quality.Get the pro<strong>of</strong>. Go towww.illumina.com/TruSeqHiSeq 2000HiSeq 1000 HiScan SQGenome Analyzer IIx MiSeq Visit us at <strong>ABRF</strong> Booth #515Illumina Technology WorkshopSunday, February 20, <strong>2011</strong>12:00 p.m. – 1:30 p.m.Grand Oaks Ballroom: Rooms P-Q (Level 2)Complimentary lunch will be provided.


<strong>Program</strong>-at-a-Glance<strong>Program</strong>-at-a-GlanceTIME SATURDAY, FEBRUARY 19 SUNDAY, FEBRUARY 20 MONDAY, FEBRUARY 21 TUESDAY, FEBRUARY 227:00 am – 6:00 pmRegistration Open Registration OpenRegistration OpenRegistration Open7:00 am – 7:45 amContinential BreakfastContinential BreakfastContinential Breakfast7:45 am – 8:00 am8:00 am – 8:50 amSatellite EducationalWorkshops(8:00 am – 4:30 pm)(SW1) Protein Purificationfor Mass SpectrometryOpening Remarks Outstanding Scientist/Technologist Award Journal <strong>of</strong> <strong>Biomolecular</strong> Techniques AwardPlenary Session: Unlocking Biomarker Discovery:Unbiased Human Proteomics at High Scale,Sensitivity, and Accuracy — Larry Gold, Chairman,CEO <strong>of</strong> SomaLogicPlenary Session: En Route to the Era <strong>of</strong> GenomicMedicine — Eric Green, Director, National HumanGenome Research InstitutePlenary Session: Development by Selventa <strong>of</strong> aTherapeutic Diagnostic TM Stratification Biomarkerfor Drug Response in Ulcerative Colitis — David deGraaf, President & CEO, Selventa9:00 am – 10:30 am10:00 am – 6:30 pm10:30 am – 12:00 pm10:30 am – 11:30 am12:00 pm – 1:30 pm1:30 pm – 3:00 pm3:00 pm – 4:15 pm4:15 pm – 4:45 pm(SW2) An Introduction toMetabolomics(SW3) Next GenerationSequencingConsiderations for CorePr<strong>of</strong>essionals(SW4) Lean Managementin Core FacilitiesConcurrent Scientific SessionsS1 - Improving Human HealthS2 - Integration <strong>of</strong> Bioinformatics for Genomicsand Proteomics DataS3 - Single-cell Environmental GenomicsPoster Session I (Odd Numbered Posters)C<strong>of</strong>fee and Tea in Exhibit Hall (10:30 am – 11:00 am)Career Fair Presentation (11:30 am – 11:45 am)Meet the Speaker: Larry GoldLunch in Exhibit HallVendor Presentation in Meeting RoomsConcurrent Research Group PresentationsR1 - Joint Session: iPRG & sPRGR2 - Antibody Technology (ARG)R3 - Joint Session: DSRG & GVRGC<strong>of</strong>fee and Tea in Exhibit HallConcurrent Scientific SessionsS4 - EpigeneticsS5 - High Throughput Genome CentersS6 - Deep Mining <strong>of</strong> Complex Protein MixturesPoster Session II (Even Numbered Posters)C<strong>of</strong>fee and Tea in Exhibit HallMeet the Speaker: Eric GreenLunch in Exhibit Hall (sponsored by )Demo Stage Presentations in Exhibit HallVendor Presentation in Meeting RoomsConcurrent Research Group PresentationsR4 - Joint Session: PSRG & gPRGR5 - Joint Session: MARG & NARGW7 - Cellular 3D ImagingC<strong>of</strong>fee and Tea in Exhibit HallConcurrent Scientific SessionsS7 - NGS Technologies on the HorizonS9 - Proteomics StandardizationExhibit Hall Open Exhibit Hall Open Exhibit Hall Open (10:00 am – 2:00 pm)Poster Session IIIStatus <strong>of</strong> the NCRR (10:30 am – 11:00 am)Waters Poster Awards (11:00 am – 12:00 pm)C<strong>of</strong>fee and Tea in Exhibit HallMeet the Speaker: David de GraafLunch in Exhibit HallDemo Stage Presentations in Exhibit HallVendor Presentation in Meeting RoomsConcurrent Research Group PresentationsR6 - Joint Session: PERG & MIRGR7 - Light Microscopy (LMRG)R8 - Joint Session: PRG & MRGFriday, February 18th – Registration Open (4:00 – 6:00 pm)Concurrent WorkshopsW1 - The Diagnostics Core FacilityW2 - Database Searching in ProteomicsW3 - Protein Turnover<strong>ABRF</strong> Award Lecture: Sir Alec John Jeffreys(sponsored by Agilent Technologies)3:15 pm – 4:15 pmConcurrent WorkshopsW11 - Business <strong>of</strong> Running a Core FacilityW12 - Microarrays: Still AliveW15b - ETD Workshop Seminar (Part II)C<strong>of</strong>fee and Tea in Exhibit Hall4:45 pm – 6:00 pm6:00 pm –7:00 pmOpening Plenary Session:Raju KucherlapatiPaul C. Cabot Pr<strong>of</strong>essor,Harvard Medical SchoolDepartment <strong>of</strong> Genetics(6:00 pm – 7:00 pm)Concurrent WorkshopsW4 - Protein Expression at Industrial ScaleW5 - Proteomics Tips & TricksW6 - Capillary Electrophoresis<strong>ABRF</strong> Affiliates and Chapters Open Mic Session(6:00 pm – 6:45 pm)Concurrent WorkshopsW8 - Production <strong>of</strong> Functional ProteinsW9 - Therapeutic AntibodiesW10 - Drug Targets from Network MiningCAC Hosted <strong>ABRF</strong>-Vendor Interface MeetingConcurrent WorkshopsW13 - Institutional Core ManagementW14 - NGS S<strong>of</strong>tware for Data ManagementW15b - ETD Workshop Seminar (Part II) (continued)<strong>ABRF</strong> Members Meeting7:00 pm – 11:00 pmOpening Reception7:00 pm – 9:00 pmNetworking Night at the Ranch(sponsored by )7:30 pm – 10:30 pmVendor Presentations in Meeting Rooms(7:30 pm – 9:00 pm)CAN-CC Meeting(7:00 pm – 8:00 pm)Closing Reception7:00 pm – 9:00 pmW15a - ETD Workshop Seminar (Part I)(8:00 pm – 11:00 pm)16 • <strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine


Daily <strong>Program</strong>Friday, February 184:00 pm – 6:00 pm Registration Open — Level 2, Ballroom Level Registration DeskSaturday, February 197:00 am – 6:00 pm Registration Open — Level 2, Ballroom Level Registration Desk7:00 am – 8:00 am Continental Breakfast — Level 2, Grand Oaks Ballroom P/Q8:00 am – 4:30 pm Satellite Educational WorkshopsSee page 28 for more detailed information.(SW1) Protein Purification for Mass SpectrometryLevel 2, Grand Oaks Ballroom R(SW2) An Introduction to MetabolomicsLevel 2, Grand Oaks Ballroom S(SW3) Next Generation Sequencing Considerations for Core Pr<strong>of</strong>essionalsLevel 2, Grand Oaks Ballroom NDaily <strong>Program</strong>(SW4) Lean Management in Core FacilitiesLevel 2, Grand Oaks Ballroom O6:00 pm – 7:00 pm Opening Plenary Session — Level 1, Exhibit Ballroom CPersonalized Medicine: Opportunities and ChallengesRaju Kucherlapati, Paul C. Cabot Pr<strong>of</strong>essor, Harvard Medical School Department <strong>of</strong> Genetics and Pr<strong>of</strong>essor, Department <strong>of</strong>Medicine, Brigham and Women’s Hospital7:00 pm – 9:00 pm Opening Reception — Level 1, Exhibit Ballroom FoyerSunday, February 207:00 am – 6:00 pm Registration Open — Level 2, Ballroom Level Registration Desk7:00 am – 7:45 am Continental Breakfast — Level 1, Exhibit Ballroom Foyer7:45 am – 8:50 am Plenary Session — Level 1, Exhibit Ballroom CUnlocking Biomarker Discovery: Unbiased Human Proteomics at High Scale, Sensitivity, and AccuracyLarry Gold, Chairman, Founder, & CEO <strong>of</strong> SomaLogic, Inc.9:00 am – 10:30 am Concurrent Scientific Sessions(S1) Improving Human Health from the Ground UpLevel 2, Grand Oaks Ballroom R/SSession Organizer: Theodore W. Thannhauser, United States Department <strong>of</strong> Agriculture/Agricultural Research Service(S1-1) Vaccine and Therapeutic Protein Manufacture in PlantsTerence E. Ryan, iBio, Inc.(S1-2) Improving Human Nutrition from the Ground Up: Linking Agriculture to Human HealthRoss M. Welch, Department <strong>of</strong> Food Science, Cornell University<strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine • 17


Sunday, February 20 (Continued)(S1-3) A Food Systems Approach to Address Poor Nutritional Health in Both Advanced and DevelopingEconomiesRobin D. Graham, Flinders University <strong>of</strong> South Australia(S2) Integration <strong>of</strong> Bioinformatics for Genomics and Proteomics DataLevel 2, Grand Oaks Ballroom P/QSession Organizer: Cathy Wu, Center for Bioinformatics & Computational Biology, University <strong>of</strong> Delaware(S2-1) Integrative Bioinformatics for Genomics and ProteomicsCathy Wu, Center for Bioinformatics & Computational Biology, University <strong>of</strong> Delaware(S2-2) Integrated Bioinformatics for MS-Based ProteomicsEric Deutsch, Institute for Systems Biology(S2-3) Skate Genome Project: Cyber-Enabled Bioinformatics CollaborationJames Vincent, University <strong>of</strong> Vermont(S3) Single-Cell Environmental GenomicsLevel 2, Grand Oaks Ballroom N/OSession Organizer: Rob Knight, University <strong>of</strong> ColoradoDaily <strong>Program</strong>(S3-1) Early Elements <strong>of</strong> a Pipeline for Single Bacterial Cell GenomicsDave Relman, Departments <strong>of</strong> Microbiology & Immunology, and <strong>of</strong> Medicine, Stanford University(S3-2) Assembling Complete Genomes from Complex MixturesFolker Meyer, Argonne National Laboratory, Mathematics and Computer Science Division(S3-3) Dining in with Trillions <strong>of</strong> Fascinating Friends: Exploring our Human Gut Microbiome in Healthand DiseaseJeff Gordon, Center for Genome Sciences and Systems Biology, Washington University School <strong>of</strong> Medicine10:00 am – 6:30 pm Exhibit Hall Open — Level 2, Grand Oaks Ballroom10:30 am – 11:00 am Morning Refreshment Break — Level 2, Grand Oaks Ballroom, Exhibit Hall10:30 am – 12:00 pm Poster Session I (Odd Numbered Posters) — Level 2, Grand Oaks Ballroom, Exhibit Hall10:30 am – 11:30 am Meet the Speaker — Level 2, Grand Oaks Ballroom, Exhibit Hall, Demo StageLarry Gold, Chairman, Founder, & CEO <strong>of</strong> SomaLogic, Inc.11:30 am – 11:45 am Career Fair Presentation — Level 2, Grand Oaks Ballroom, Exhibit Hall, Demo Stage12:00 pm – 1:30 pm Munch & Mingle — Level 2, Grand Oaks Ballroom, Exhibit Hall12:00 pm – 1:30 pm Vendor PresentationsSee page 120 for detailed information.1:30 pm – 3:00 pm Concurrent Research Group Presentations(R1) Joint Session: Proteome Informatics Research Group (iPRG) & Proteomics Standards Research Group(sPRG)Level 2, Grand Oaks Ballroom R/S(R1a) iPRG <strong>2011</strong>: A Study on the Identification <strong>of</strong> Electron Transfer Dissociation (ETD) Mass SpectraSession Organizer & Presenter: Lennart Martens, Ghent University(R1b) <strong>ABRF</strong>-sPRG<strong>2011</strong> Study: Development <strong>of</strong> a Comprehensive Standard for Analysis <strong>of</strong> Post-Translational ModificationsSession Organizer & Presenter: Alexander R. Ivanov, Harvard School <strong>of</strong> Public Health18 • <strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine


(R2) Antibody Technology Research Group (ARG): Antibodies — Moving Closer to Personalized Therapeuticsand DiagnosticsLevel 2, Grand Oaks Ballroom P/QSession Organizers: Robert Carnahan, Vanderbilt University Medical Center, and Frances Weis-Garcia, Memorial Sloan-Kettering Cancer CenterPresenters: Brian Curtis, Blood Center <strong>of</strong> Wisconsin, John Harlan, Abbott Laboratories, and Robert Umek, Meso ScaleDiscovery(R3) Joint Session: Genomic Variation Research Group (GVRG) & DNA Sequencing Research Group (DSRG)Level 2, Grand Oaks Ballroom N/O(R3a): Evaluation <strong>of</strong> DNA Whole Genome Amplification Technologies for GenotypingSession Organizers & Presenters: Casey Dagnall and Amy Hutchinson, Core Genotyping Facility, SAIC-Frederick,Inc., NCI-Frederick(R3b): DNA Sequencing Research Group (DSRG)Session Organizer: Deborah Grove, Pennsylvania State University(R3b-1) Comparison <strong>of</strong> Custom Target Enrichment Methods; Agilent vs. NimblegenAnoja Perera, Stowers Institute for Medical Research Molecular Biology Facility(R3b-2) A Methodology Study for Metagenomics Using Next Generation SequencersSushmita Singh, BioMedical Genomics Center, University <strong>of</strong> Minnesota3:00 pm – 4:15 pm Concurrent Workshop Sessions(W1) The Diagnostics Core Facility: Harvesting the Promise <strong>of</strong> Personalized MedicineLevel 2, Grand Oaks Ballroom R/SSession Organizer: Katia Sol-Church, Nemours Biomedical Research, A.I. duPont Hospital for ChildrenDaily <strong>Program</strong>(W1-1) Win on Sunday, Sell on Monday: From the Exome Sequencing <strong>of</strong> One Boy to the Delivery <strong>of</strong>Clinical DiagnosticsMichael R. Tschannen, Human and Molecular Genetics Center, Department <strong>of</strong> Physiology, Medical College <strong>of</strong>Wisconsin(W1-2) Chromosomal Microarray Analysis in the ClinicLisa D. White, Microarray Core, Baylor College <strong>of</strong> Medicine(W1-3) Whole Genome Sequencing in the Clinical LaboratoryTina Hambuch, Illumina Clinical Services Laboratory, Illumina, Inc.(W2) Spectral and Sequence Database Searching in ProteomicsLevel 2, Grand Oaks Ballroom P/QSession Organizer: Lennart Martens, Ghent University(W2-1) ETD Performance and Complementarity to Other Fragmentation Methods for ProteomicAnalysisRobert Chalkley, University <strong>of</strong> California San Francisco(W2-2) Discovery, Identification and Localization <strong>of</strong> Post-Translational ModificationsNuno Bandeira, Center for Computational Mass Spectrometry, Department <strong>of</strong> Computer Science and EngineeringSkaggs School <strong>of</strong> Pharmacy and Pharmaceutical Sciences, University <strong>of</strong> California San Diego(W2-3) Building and Using MS/MS Spectral Libraries for Peptide Identifications in ProteomicsHenry Lam, The Hong Kong University <strong>of</strong> Science and Technology(W3) Quantifying Protein Turnover by In Vivo Metabolic LabelingLevel 2, Grand Oaks Ballroom N/OSession Organizer: Kieran Geoghegan, Pfizer, Inc.<strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine • 19


Sunday, February 20 (Continued)(W3-1) Stable Isotope Tracers Applied to Measuring Rates <strong>of</strong> Protein Synthesis and Breakdown inMuscle: Principles and ApplicationsRobert R. Wolfe, University <strong>of</strong> Arkansas Medical Sciences(W3-2) In Vivo Stable Isotope Labeling for Quantifying Amyloid-Beta Kinetics in Alzheimer’s Disease:Is it All in our Head?Kevin E. Yarasheski, Biomedical Mass Spectrometry Research Laboratory, Washington University School <strong>of</strong> Medicine4:15 pm – 4:45 pm Afternoon Refreshment Break — Level 2, Grand Oaks Ballroom, Exhibit Hall4:45 pm – 6:00 pm Concurrent Workshop Sessions(W4) Insights for Expression <strong>of</strong> Recombinant Proteins for Drug Target ValidationLevel 2, Grand Oaks Ballroom R/SSession Organizer: Francis Rajamohan, Pfizer, Inc.(W4-1) New Developments in BacMam VectorsFrederick M. Boyce, Massachusetts General Hospital, Department <strong>of</strong> Neurology(W4-2) Strategies for Optimized High-Throughput Cloning, Expression, and Purification <strong>of</strong>Recombinant ProteinsRebecca Page, Brown University, Department <strong>of</strong> Molecular Biology, Cell Biology and Biochemistry, Center forGenetics, Genomics and ProteomicsDaily <strong>Program</strong>(W5) Proteomics Tips and Tricks: From Discovery to Protein-Protein InteractionsLevel 2, Grand Oaks Ballroom P/QSession Organizer: Michelle Cilia, United States Department <strong>of</strong> Agriculture/Agricultural Research Service, Robert W. HolleyCenter for Agriculture and Health(W5-1) Proteomics Tips and Tricks: From Discovery to Protein-Protein InteractionsMichelle Cilia, United States Department <strong>of</strong> Agriculture/Agricultural Research Service, Robert W. Holley Center forAgriculture and Health(W5-2) Protein Interactions and Topologies in CellsJames Bruce, University <strong>of</strong> Washington, Department <strong>of</strong> Genome Sciences(W5-3) The Virus-Host Interface: Exploring Dynamic Protein Interactions via Targeted ProteomicsIleana Cristea, Princeton University, Department <strong>of</strong> Molecular Biology(W5-4) Towards the Development <strong>of</strong> Proteomics Workflows for the Analysis <strong>of</strong> Samples Derived fromRefractory Plant TissuesTheodore W. Thannhauser, United States Department <strong>of</strong> Agriculture/Agricultural Research Service(W6) Current State and Future <strong>of</strong> Capillary Electrophoresis and Sanger SequencingLevel 2, Grand Oaks Ballroom N/OSession Organizer: Jan Kieleczawa, Pfizer, Inc.Presenters: Debbie Adam, NAPS Unit, Michael Smith Laboratories, University <strong>of</strong> British Columbia, Jan Kieleczawa, Pfizer,Inc., Peter Schweitzer, Cornell University, Eric Vennemeyer, LifeTechnologies, and Michael Zianni, Plant-Microbe GenomicsFacility, Ohio State University6:00 pm – 6:45 pm <strong>ABRF</strong> Affiliates and Chapters Open Mic Session — Level 2, Grand Oaks Ballroom N/O7:30 pm – 10:30 pm Networking Night — Offsite: Leon Springs Dance Hall*(Sponsored by )*Buses will begin boarding at 7:00 pm from the Level 1, Exhibit Hall Lobby. Buses will run continuously between 7:00 pmand 10:30 pm.20 • <strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine


Monday, February 217:00 am – 6:00 pm Registration Open — Level 2, Ballroom Level Registration Desk7:00 am – 7:45 am Continental Breakfast — Level 1, Exhibit Ballroom Foyer7:45 am – 8:50 am Plenary Session — Level 1, Exhibit Ballroom CEn Route to the Era <strong>of</strong> Genomic MedicineEric Green, Director, National Human Genome Research Institute9:00 am – 10:30 am Concurrent Scientific Sessions(S4) EpigeneticsLevel 2, Grand Oaks Ballroom R/SSession Organizer: Benjamin A. Garcia, Princeton University(S4-1) Next Generation Quantitative Proteomic Tools for Analyzing Histone ModificationsBenjamin A. Garcia, Princeton University(S4-2) Chromatin Dynamics in Melanoma: A Role for MacroH2AAvnish Kapoor, Department <strong>of</strong> Oncological Sciences, Mount Sinai School <strong>of</strong> Medicine(S4-3) Using Protein Domain Microarrays to Read the Histone CodeMark Bedford, Department <strong>of</strong> Carcinogenesis, Science Park, Research Division, The University <strong>of</strong> Texas MD AndersonCancer Center(S5) High-Throughput Genome CentersLevel 2, Grand Oaks Ballroom P/QSession Organizer: George Grills, Cornell UniversityDaily <strong>Program</strong>(S5-1) Overview <strong>of</strong> the Illumina Sequencing Platform at the Broad InstituteKristen Connolly, Process & Technology Development, Genome Sequencing Platform, The Broad Institute <strong>of</strong> MIT andHarvard(S5-2) Science and Technology at a High-Throughput Genome CenterLucinda Fulton, The Genome Center at Washington University School <strong>of</strong> Medicine(S5-3) High-Throughput Next Generation Sequencing Methods and ApplicationsDonna Muzny, Baylor College <strong>of</strong> Medicine, Human Genome Sequencing Center(S6) Strategies for Deep Mining <strong>of</strong> Complex Protein MixturesLevel 2, Grand Oaks Ballroom N/OSession Organizer: David Speicher, The Wistar Institute(S6-1) Coverage and Recovery <strong>of</strong> Upstream Protein Fractionation Methods in LC-MS/MS WorkflowsLeonard J. Foster, Centre for High-Throughput Biology, The University <strong>of</strong> British Columbia(S6-2) Improving the Comprehensiveness <strong>of</strong> Large-Scale Proteomics Experiments Using AdvancedComputational Tools and Accurate Multiple Hypothesis Testing StatisticsMichael J. MacCoss, Department <strong>of</strong> Genome Sciences, University <strong>of</strong> Washington(S6-3) In-Depth Analysis <strong>of</strong> Human and Mouse Plasma Using 3-D and 4-D Fractionation StrategiesDavid Speicher, The Wistar Institute10:00 am – 6:30 pm Exhibit Hall Open — Level 2, Grand Oaks Ballroom10:30 am – 11:00 am Morning Refreshment Break — Level 2, Grand Oaks Ballroom, Exhibit Hall10:30 am – 12:00 pm Poster Session II — Level 2, Grand Oaks Ballroom, Exhibit Hall10:30 am – 11:30 am Meet the Speaker — Level 2, Grand Oaks Ballroom, Exhibit Hall, Demo StageEric Green, Director, National Human Genome Research Institute12:00 pm – 1:30 pm Munch & Mingle — Level 2, Grand Oaks Ballroom, Exhibit Hall (Sponsored by )<strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine • 21


Monday, February 21 (Continued)12:00 pm – 1:30 pm Vendor and Demo Stage PresentationsSee pages 120 and 126 for detailed information.1:30 pm – 3:00 pm Concurrent Research Group Presentations(R4) Joint Session: Protein Sequencing Research Group (PSRG) & Glycoprotein Research Group (gPRG)Level 2, Grand Oaks Ballroom R/S(R4a) PSRG <strong>2011</strong> Study: Sensitivity Assessment for Terminal Sequencing Techniques Using an UnknownProteinSession Organizer: Jim Walters, Sigma AldrichPresenters: Kwasi Mawuenyega, Washington University School <strong>of</strong> Medicine, Wendy Sandoval, Genentech, Inc., SteveSmith, University <strong>of</strong> Texas, and Jim Walters, Sigma Aldrich(R4b) gPRG: Toward Consensus on Glycan Analysis: Reliable Methods and ReproducibilitySession Organizer & Presenter: Joseph Zaia, Boston University(R5) Joint Session: Nucleic Acids Research Group (NARG) & Microarray Research Group (MARG)Level 2, Grand Oaks Ballroom P/QDaily <strong>Program</strong>(R5a) Determining miRNA Expression Levels in Degraded RNA Samples Using Real-Time RT-qPCR andMicroarray TechnologiesSession Organizer: Sridar Chittur, State University <strong>of</strong> New York at AlbanyPresenters: Sridar Chittur, State University <strong>of</strong> New York at Albany, and Scott Tighe, University <strong>of</strong> Vermont(R5b) Microarray Research Group Projects, 2010-11Session Organizer: Don A. Baldwin, Molecular Pr<strong>of</strong>iling Facility, University <strong>of</strong> PennsylvaniaPresenters: Nadereh Jafari, Northwestern University, and Natalia Reyero-Vinas, Jackson State University(W7) Cellular 3D Imaging*Level 2, Grand Oaks Ballroom N/OSession Organizer: Richard Cole, Wadsworth Center, New York State Department <strong>of</strong> Health(W7-1) 3D Cellular Imaging: Beyond the Simple Imaging ParadigmRichard Cole, Wadsworth Center, New York State Department <strong>of</strong> Health(W7-2) Basics <strong>of</strong> Colocalization AnalysesJudith Lacoste, Cell Imaging and Analysis Network, Department <strong>of</strong> Biology, McGill University*A Leica confocal microscope, provided by Leica Microsystems, will be featured in this session in Grand Oaks B.Leica Scanner — Dedicated to advancing biomolecular core facilitiesThe <strong>ABRF</strong> will host a Leica SP5 II confocal equipped with a tandem scanner. This Confocal covers a broad range <strong>of</strong>requirements in confocal imaging — with the full array <strong>of</strong> scan speeds at highest resolution. With its high-efficiency SPdetection (five channels simultaneously) and the optional AOBS (Acousto-Optical Beam Splitter), the system deliversbright, noise-free images with minimal photo damage at high speed. Additionally, there will be available the latestAutoquant’s advanced image deconvolution and 3D visualization s<strong>of</strong>tware for life science researchers.Imaging sessions include:• Co-registration & deconvolution “hands-on”• Confocal microscope & deconvolution s<strong>of</strong>tware on-site• Opportunity to image and analyze “your” sample (physical or digital file from your scope)• Performance testing: point spread functions, spectral calibration & beyond• Core management session/round tables• Networking for imaging core personnel3:15 pm – 4:15 pm <strong>ABRF</strong> Award Lecture: Sir Alec John Jeffreys — Level 1, Exhibit Ballroom C(Sponsored by Agilent Technologies.)22 • <strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine


4:15 pm – 4:45 pm Afternoon Refreshment Break — Level 2, Grand Oaks Ballroom, Exhibit Hall4:45 pm – 6:00 pm Concurrent Workshop Sessions(W8) Successful Production <strong>of</strong> Functional ProteinsLevel 2, Grand Oaks Ballroom R/SSession Organizer: Jeff Culp, Pfizer, Inc.(W8-1) Successful Protein ProductionJeff Culp, Pfizer, Inc.(W8-2) Overcoming Problems in Protein Expression and PurificationBill Gillette, SAIC-Frederick, Inc., National Cancer Institute at Frederick(W8-3) When Proteins Misbehave, Try Adding a Little PressureRobert M. Petrovich, National Institute <strong>of</strong> Environmental Health Sciences Laboratory <strong>of</strong> Structural Biology(W9) Therapeutic Antibodies: Over-Hyped “Magic Bullet”, or Under-Explored Technology?Level 2, Grand Oaks Ballroom P/QSession Organizer: Dan L. Crimmins, Washington University School <strong>of</strong> MedicinePresenters: Dan L. Crimmins, Washington University School <strong>of</strong> Medicine, John Harlan, Abbott Laboratories, and FrancesWeis-Garcia, Memorial Sloan-Kettering Cancer Center(W10) Identification <strong>of</strong> Mechanism-Based Biomarkers and Drug Targets Using Pathway AnalysisLevel 2, Grand Oaks Ballroom N/OSession Organizer: Alexander Kel, geneXplain GmbH(W10-1) GeneXplain — Identification <strong>of</strong> Causal Biomarkers and Drug Targets in PersonalizedCancer PathwaysAlexander Kel, geneXplain GmbHDaily <strong>Program</strong>(W10-2) Pathway Analysis in Expression ProteomicsRoman Zubarev, Chemistry I Division, Department <strong>of</strong> Medical Biochemistry and Biophysics, Karolinska Institute(W10-3) Metabolic Biomarkers, Metabolic Networks, and Pathway AnalysisVladimir Tolstikov, University <strong>of</strong> California Davis Genome Center6:00 pm – 7:00 pm CAC Hosted <strong>ABRF</strong>-Vendor Interface Meeting — Level 1, Exhibit Ballroom C7:00 pm – 8:00 pm Core Administrators Network – Coordinating Committee (CAN-CC) Meeting — Level 1, Exhibit Ballroom C7:30 pm – 9:00 pm Vendor PresentationsSee page 120 for detailed information.8:00 pm – 11:00 pm (W15a) ETD Workshop Seminar Series PART I: Manual Interpretation <strong>of</strong> Electron Transfer Dissociation (ETD)Mass Spectra <strong>of</strong> Peptides* — Level 2, Grand Oaks Ballroom N/OPresenter: Donald F. Hunt, Departments <strong>of</strong> Chemistry and Pathology, University <strong>of</strong> VirginiaTuesday, February 22*Attendees must be registered for <strong>ABRF</strong> <strong>2011</strong> to attend the workshop and must also complete the ETD Workshop SeminarForm. Please visit the Registration Desk to sign up for the session.7:00 am – 6:00 pm Registration Open — Level 2, Ballroom Level Registration Desk7:00 am – 7:45 am Continental Breakfast — Level 1, Exhibit Ballroom Foyer7:45 am – 8:50 am Plenary Session — Level 1, Exhibit Ballroom CDevelopment by Selventa <strong>of</strong> a Therapeutic Diagnosticin Ulcerative ColitisDavid de Graaf, President & CEO, SelventaTMStratification Biomarker for Drug Response<strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine • 23


Tuesday, February 22 (Continued)9:00 am – 10:30 am Concurrent Scientific Sessions(S7) Next Generation Sequencing Technologies on the HorizonLevel 2, Grand Oaks Ballroom R/SSession Organizer: Katia Sol-Church, Nemours Biomedical Research, A.I. duPont Hospital for Children(S7-1) Towards Optical DNA Sequencing Using Nanopore ArraysAmit Meller, Department <strong>of</strong> Biomedical Engineering & Department <strong>of</strong> Physics, Boston University(S7-2) Single Molecule Real-Time DNA Sequencing on the Surface <strong>of</strong> a Quantum-Dot NanocrystalPeter B. Vander Horn, Genetic Systems, Life Technologies(S7-3) Sequencing with Semiconductor ChipsGlenn Powell, Ion Torrent(S9) Proteomics StandardizationLevel 2, Grand Oaks Ballroom N/OSession Organizer: David B. Friedman, Proteomics Laboratory Mass Spectrometry Research Center, Vanderbilt University(S9-1) A Multi-Laboratory Study Assessing Robustness and Reproducibility <strong>of</strong> Plasma ReferenceSample for Benchmarking LC-MS Platform PerformanceJuan-Pablo Albar, ProteoRed, National Center for Biotechnology-CSICDaily <strong>Program</strong>(S9-2) Clinical Proteomic Technologies for Cancer: NIH-Funded Measurement ScienceChristopher Kinsinger, National Cancer Institute, Center for Strategic Scientific Initiatives, Office <strong>of</strong> Cancer ClinicalProteomics Research(S9-3) Why Reproducible Outcomes are Essential in Proteomic Research and WhyStandardization <strong>of</strong> Processes is Essential for Achieving ReproducibilityAndy Borthwick, Nonlinear Dynamics Limited10:00 am – 2:00 pm Exhibit Hall Open — Level 2, Grand Oaks Ballroom10:30 am – 11:00 am Morning Refreshment Break — Level 2, Grand Oaks Ballroom, Exhibit Hall10:30 am – 12:00 pm Poster Session III — Level 2, Grand Oaks Ballroom, Exhibit Hall10:30 am – 11:00 am Status <strong>of</strong> the NCRR — Level 2, Grand Oaks Ballroom P/QPresenter: Mark O. Lively, Wake Forest University School <strong>of</strong> Medicine11:00 am – 12:00 pm Poster Awards (Sponsored by Waters Corporation) — Level 2, Grand Oaks Ballroom, Exhibit Hall, Demo Stage10:30 am – 11:30 am Meet the Speaker — Level 2, Grand Oaks Ballroom, Exhibit HallDavid de Graaf, President & CEO, Selventa12:00 pm – 1:30 pm Munch & Mingle — Level 2, Grand Oaks Ballroom, Exhibit Hall12:00 pm – 1:30 pm Vendor and Demo Stage PresentationsSee pages 120 and 126 for detailed information.1:30 pm – 3:00 pm Concurrent Research Group Presentations(R6) Joint Session: Protein Expression Research Group (PERG) & Molecular Interactions Research Group (MIRG)Level 2, Grand Oaks Ballroom R/S(R6a) PERG Research Group Presentation: Refolding StudySession Organizer: Cynthia Kinsland, Cornell University(R6b) Conclusions from the MIRG 2010 Benchmark Study: Molecular Interactions in a ThreeComponent System and Presentation <strong>of</strong> <strong>2011</strong> Survey Results on Label-Free TechnologiesSession Organizer: Aaron Yamiuk, Bristol-Myers SquibbPresenters: Satya Yadav, Cleveland Clinic Foundation, and Aaron Yamiuk, Bristol-Myers Squibb24 • <strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine


(R7) Light Microscopy Research Group (LMRG)Level 2, Grand Oaks Ballroom P/QSession Organizer: Robert Stack, Wadsworth Center, New York State Department <strong>of</strong> Health(R7-1) Point Spread Functions, Spectral Calibration, and BeyondRobert Stack, Wadsworth Center, New York State Department <strong>of</strong> Health(R7-2) Deconvolution: Core Concepts, Algorithms, and Advanced IssuesBrian Northan, MediaCybernetics(R8) Joint Session: Proteomics Research Group (PRG) & Metabolomics Research Group (MRG)Level 2, Grand Oaks Ballroom N/O(R8a) PRG-<strong>2011</strong>: Defining the Interaction Between Users and Suppliers <strong>of</strong> Proteomics ServicesSession Organizer: David Hawke, The University <strong>of</strong> Texas MD Anderson Cancer Center(R8b) Metabolomics Research Group <strong>2011</strong> StudySession Organizer: William R. Wik<strong>of</strong>f, University <strong>of</strong> California DavisPresenters: John M. Asara, Beth Israel Deaconess Medical Center, Vladimir Tolstikov and William R. Wik<strong>of</strong>f,University <strong>of</strong> California Davis3:00 pm – 4:15 pm Concurrent Workshop Sessions(W11) The Business <strong>of</strong> Running a Core FacilityLevel 2, Grand Oaks Ballroom R/SSession Organizer: Nicholas Ambulos, University <strong>of</strong> Maryland School <strong>of</strong> MedicinePresenters: Nicholas Ambulos, University <strong>of</strong> Maryland School <strong>of</strong> Medicine, and Steve Bobin, Dartmouth School <strong>of</strong> Medicine(W12) Microarrays: The Reports <strong>of</strong> My Death Have Been Greatly ExaggeratedLevel 2, Grand Oaks Ballroom P/QSession Organizer: Don Baldwin, University <strong>of</strong> Pennsylvania, Penn Molecular Pr<strong>of</strong>iling FacilityDaily <strong>Program</strong>(W12-1) Chip or Seq: Helping Clients ChooseDon Baldwin, University <strong>of</strong> Pennsylvania, Penn Molecular Pr<strong>of</strong>iling Facility, and Kevin Knudtson, University <strong>of</strong> Iowa(W12-2) Microarray Analysis <strong>of</strong> Fluorescence Activated Cell Sorter-Derived Cells: Creating Harmonybetween TechnologiesScott Tighe, University <strong>of</strong> Vermont(W12-3) Microarray Futures: Don’t Decommission Your Scanners Just YetSeth Crosby, Department <strong>of</strong> Genetics, Washington University School <strong>of</strong> Medicine(W15b) ETD Workshop Seminar Series PART II: Manual Interpretation <strong>of</strong> Electron Transfer Dissociation (ETD)Mass Spectra <strong>of</strong> Peptides*Level 2, Grand Oaks Ballroom N/OPresenter: Donald F. Hunt, Departments <strong>of</strong> Chemistry and Pathology, University <strong>of</strong> Virginia*Attendees must be registered for <strong>ABRF</strong> <strong>2011</strong> to attend the workshop and must also complete the ETD Workshop SeminarForm. Please visit the Registration Desk to sign up for the session.4:15 pm – 4:45 pm Afternoon Refreshment Break — Level 2, Grand Oaks Ballroom N/S Foyer4:45 pm – 6:00 pm Concurrent Workshop Sessions(W13) Institutional Core ManagementLevel 2, Grand Oaks Ballroom R/SSession Organizers: Susan Meyn, Vanderbilt University Medical Center, and Paula Turpen, University <strong>of</strong> Nebraska MedicalCenterPresenter: Pam Alexander, Morehouse School <strong>of</strong> Medicine, Julie Auger, University <strong>of</strong> California San Francisco, GregoryFarber, National Center for Research Resources, and Sheenah Mische, New York University Langone Medical Center<strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine • 25


Tuesday, February 22 (Continued)(W14) Next Generation Sequencing S<strong>of</strong>tware for Data Management, Analysis, and VisualizationLevel 2, Grand Oaks Ballroom P/QSession Organizer: Kip Bodi, Tufts University School <strong>of</strong> Medicine(W14-1) Tools for Next Generation Sequencing Data AnalysisKip Bodi, Tufts University School <strong>of</strong> Medicine(W14-2) GenomeView: Visualizing the Next Generation <strong>of</strong> DataThomas Abeel, Broad Institute <strong>of</strong> MIT and Harvard, and VIB Department <strong>of</strong> Plant Systems Biology, Ghent University(W14-3) Galaxy Next Generation Sequencing Functionality from Sample Tracking to SNP CallingGreg Von Kuster, Pennsylvania State University(W15b) ETD Workshop Seminar Series PART II: Manual Interpretation <strong>of</strong> Electron Transfer Dissociation (ETD)Mass Spectra <strong>of</strong> Peptides (continued)Level 2, Grand Oaks Ballroom N/OPresenter: Donald F. Hunt, Departments <strong>of</strong> Chemistry and Pathology, University <strong>of</strong> Virginia*Attendees must be registered for <strong>ABRF</strong> <strong>2011</strong> to attend the workshop and must also complete the ETD Workshop SeminarForm. Please visit the Registration Desk to sign up for the session.Daily <strong>Program</strong>6:10 pm – 7:00 pm <strong>ABRF</strong> Member’s Meeting — Level 1, Exhibit Ballroom C7:00 pm – 9:00 pm Closing Reception — JW Marriott San Antonio Hill Country Resort, Grand Ballroom Terrace26 • <strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine


The Future Is EmergingTake an integrated view <strong>of</strong> biology, with tools from Agilent. A new frontier is emerging. Advances in technologyhave given researchers the ability to study complex biological systems beyond individual components—and Agilentis committed to helping scientists harness this integrated approach. Agilent supports the range <strong>of</strong> omics platforms,with genomic, transcriptomic, proteomic, and metabolomic applications, enabling a multi-faceted view into biologicalprocesses. Informatics tools provide analysis for each <strong>of</strong> the omics, and <strong>of</strong>fer integrated data sets across the biologicalsystem. Agilent is also collaborating with scientists to overcome the challenges <strong>of</strong> this emerging discipline, helpingfi nd answers to some <strong>of</strong> our most complex questions. Get the full picture at www.agilent.com/lifesciences/biology.© Agilent Technologies, Inc. 2010


Satellite Educational Workshop Sponsors(SW1) Protein Purification for Mass Spectrometry(SW2) An Introduction to Metabolomics(SW3) Next Generation Sequencing Considerations for Core Pr<strong>of</strong>essionalsSatellite EducationalWorkshops(SW4) Lean Management in Core Facilities28 • <strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine


Satellite Educational WorkshopsAll Satellite Educational Workshops will be held on Saturday, February 19 at the JW Marriott San Antonio Hill Country Resort.Attendees must register separately for all Satellite Educational Workshops. To register, please visit the Registration Desk on Level 2.For further information and for individual workshop schedules, please review the proceeding pages.SW1: Protein Purification for Mass Spectrometry8:00 am – 4:00 pm Level 2, Grand Oaks Ballroom ROrganizers & Presenters: Robert Carnahan, and W. Hayes McDonald, Vanderbilt University Medical CenterWorkshop Description: The workshop is intended as a practical guide for investigators wishing to purify proteins and proteincomplexes for mass spectrometry based analysis. It should be useful for individuals wishing to perform such purifications as well asfor mass spectrometrists who will be analyzing these types <strong>of</strong> samples. We will explore various considerations and possibilities indesigning and executing protein purifications intended for mass spectrometry and then focus on antibody-based affinity purifications.While there will not be time to take workshop participants through all <strong>of</strong> the hands-on aspects, our intent is to guide them throughdesigning and troubleshooting the various stages <strong>of</strong> the process. We will then utilize data generated from an actual hands-on course,and allow the participants to see and interpret some real data that they have “watched” being generated.Workshop Agenda7:00 am – 6:00 pm Registration Open — Level 2, Ballroom Level Registration Desk7:00 am – 8:00 am Continental Breakfast — Level 2, Grand Oaks Ballroom P/Q8:00 am – 9:00 am Affinity Purification for Mass SpectrometryPresenter: Robert Carnahan, Vanderbilt University Medical Center9:00 am – 10:00 am Mass Spectrometry Analysis <strong>of</strong> Protein ComplexesPresenter: W. Hayes McDonald, Vanderbilt University Medical Center10:00 am – 10:30 am Morning Refreshment Break — Level 2, Grand Oaks Ballroom P/Q10:30 am – 12:00 pm Virtual “Hands-On” WorkshopPresenters: W. Hayes McDonald and Robert Carnahan, Vanderbilt University Medical CenterReagent generation and QCBiological OptimizationSample Preparation and ProcessingData Analysis12:00 pm – 1:00 pm Lunch — Level 2, Grand Oaks Ballroom P/Q1:00 pm – 2:30 pm Exploration <strong>of</strong> Experimental Design Considerations and their ImplicationsPresenters: W. Hayes McDonald and Robert Carnahan, Vanderbilt University Medical Center2:30 pm – 3:00 pm Afternoon Refreshment Break — Level 2, Grand Oaks Ballroom P/Q3:00 pm – 4:00 pm Alternative ApproachesPresenters: W. Hayes McDonald and Robert Carnahan, Vanderbilt University Medical CenterSatellite EducationalWorkshops<strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine • 29


SW2: An Introduction to Metabolomics8:00 am – 4:30 pm Level 2, Grand Oaks Ballroom SOrganizer: William R. Wik<strong>of</strong>f, University <strong>of</strong> California DavisPresenters: William R. Wik<strong>of</strong>f, University <strong>of</strong> California Davis, and Pavel Aronov, Stanford UniversityWorkshop Description: Metabolomics can be defined as the systems-level investigation <strong>of</strong> small molecules and metabolites inbiological cells, tissues, and organisms. The course will provide the scientist with an overview <strong>of</strong> the field, with an emphasis on practical,mass spectrometry-based approaches, presented in a clear scientific framework. The entire process <strong>of</strong> metabolomics, from studydesign, sample preparation and extraction, chromatography, mass spectrometry, data processing and analysis will be presented.Topics will include: types <strong>of</strong> detectors and their relative merits for specific metabolomics applications, including quadrupole, triplequad, ion trap, TOF, QTOF, and FT-ICR. Ionization sources. Application <strong>of</strong> LCMS and GCMS, including a comparison <strong>of</strong> observablecompounds in these techniques. Brief coverage <strong>of</strong> specialized instruments: GC-QQQ, GC-TOF, chemical ionization. Methods forsample extraction. Targeted versus untargeted methods. Peak integration, data alignment, and s<strong>of</strong>tware. Data analysis approaches willbe discussed, including uni- and multi-variate statistics, with an emphasis on selecting straightforward approaches appropriate for agiven problem. Basic statistics for metabolomics: types <strong>of</strong> t-tests, multiple testing correction, False Discovery Rate, basic multivariatemethods. Approaches to biomarker discovery; metabolomics in drug discovery. Review and discussion <strong>of</strong> selected publications fromthe literature (case studies). Some familiarity with mass spectrometry and analytical chemistry is preferred, but background will beprovided.Workshop AgendaSatellite EducationalWorkshops7:00 am – 6:00 pm Registration Open — Level 2, Ballroom Level Registration Desk7:00 am – 8:00 am Continental Breakfast — Level 2, Grand Oaks Ballroom P/Q8:00 am – 8:30 am Introduction and Overview with Historical BackgroundPresenter: William Wik<strong>of</strong>f, University <strong>of</strong> California Davis8:30 am – 10:00 am Instrumentation for Metabolomics: Mass Spectrometry, Mass Analyzers, Resolution and Mass Accuracy.GC/MS: Separation, Mass Spec, and LibrariesPresenter: Pavel Aronov, Stanford University10:00 am – 10:30 am Morning Refreshment Break — Level 2, Grand Oaks Ballroom P/Q10:30 am – 12:00 pm Sample Preparation: Liquid Chromatography, Columns and MethodsPresenter: William Wik<strong>of</strong>f, University <strong>of</strong> California Davis12:00 pm – 1:00 pm Lunch — Level 2, Grand Oaks Ballroom P/Q1:00 pm – 2:00 pm Data Processing: MZmine, XCMS, Databases. Compound IdentificationPresenter: Pavel Aronov, Stanford University2:00 pm – 2:30 pm Statistics Part IPresenter: William Wik<strong>of</strong>f, University <strong>of</strong> California Davis2:30 pm – 3:00 pm Afternoon Refreshment Break — Level 2, Grand Oaks Ballroom P/Q3:00 pm – 3:30 pm Statistics Part IIPresenter: William Wik<strong>of</strong>f, University <strong>of</strong> California Davis3:30 pm – 4:30 pm Metabolomics Case Studies, Summary and DiscussionPresenter: William Wik<strong>of</strong>f, University <strong>of</strong> California Davis30 • <strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine


SW3: Next Generation Sequencing Considerations For Core Pr<strong>of</strong>essionals8:00 am – 4:30 pm Level 2, Grand Oaks Ballroom NOrganizer: Kevin L. Knudtson, University <strong>of</strong> IowaPresenters: Deborah Grove, Pennsylvania State University, Anoja G. Perera, Stowers Institute for Medical Research, and Peter Schweitzer,Cornell UniversityWorkshop Description: This workshop is intended for the laboratory pr<strong>of</strong>essional looking to enhance the quality <strong>of</strong> their recentlyestablished Next Generation Sequencing (NGS) services. It is not intended for the seasoned NGS service provider. The organization<strong>of</strong> the workshop will be similar to the workflow a core pr<strong>of</strong>essional expert might take when working with an investigator on theirnext generation sequencing project. The early sessions will focus on platform choice, experimental design, and sample preparationconsiderations. The latter sessions will focus on the performance <strong>of</strong> specific applications.Workshop Agenda7:00 am – 6:00 pm Registration Open — Level 2, Ballroom Level Registration Desk7:00 am – 8:00 am Continental Breakfast — Level 2, Grand Oaks Ballroom P/Q8:00 am – 8:30 am Overview and First ContactPresenter: Kevin Knudtson, University <strong>of</strong> Iowa8:30 am – 9:15 am Providing Next Gen Services Using the GS FLXPresenter: Kevin Knudtson, University <strong>of</strong> Iowa9:15 am – 10:00 am Providing Next Gen Services using the GAIIxPresenter: Anoja Perera, Stowers Institute for Medical Research10:00 am – 10:30 am Morning Refreshment Break — Level 2, Grand Oaks Ballroom P/Q10:30 am – 11:15 am Providing Next Gen Services using the HiSeq2000Presenter: Peter Schweitzer, Cornell University11:15 am – 12:00 pm Providing Next Gen Services using the SOLiDPresenter: Deborah Grove, Pennsylvania State University12:00 pm – 1:00 pm Lunch — Level 2, Grand Oaks Ballroom P/Q1:00 pm – 1:30 pm Agilent Presentation1:30 pm – 2:30 pm Specialized Protocols for Use with Next GenPresenters: Anoja Perera, Stowers Institute for Medical Research, and Peter Schweitzer, Cornell University2:30 pm – 3:00 pm Afternoon Refreshment Break — Level 2, Grand Oaks Ballroom P/Q3:00 pm – 3:45 pm Data Management /LIMSPresenters: Deborah Grove, Pennsylvania State University, Kevin Knudtson, University <strong>of</strong> Iowa, Anoja Perera, StowersInstitute for Medical Research, and Peter Schweitzer, Cornell University3:45 pm – 4:30 pm Panel DiscussionPanelists: Deborah Grove, Pennsylvania State University, Kevin Knudtson, University <strong>of</strong> Iowa, Anoja Perera, Stowers Institutefor Medical Research, and Peter Schweitzer, Cornell UniversitySatellite EducationalWorkshops<strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine • 31


SW4: Lean Management in Core Facilities8:00 am – 4:30 pm Level 2, Grand Oaks Ballroom OOrganizer: Tim C. Hunter, Vermont Cancer CenterPresenters: Belynda Hicks, SAIC-Frederick, National Cancer Institute at Frederick, Nicholas Ambulos, University <strong>of</strong> Maryland School <strong>of</strong>Medicine, Val Scott, The Jackson Laboratory, Rand Haley, Huron Consulting Group, Wayne Collins, Agilent Technologies, and Tim C.Hunter, University <strong>of</strong> VermontWorkshop Description: This workshop is intended for core directors, managers, and facility personnel that are interested in learningnew and creative approaches to core management during lean budget times. Sessions will focus on ways to reduce or maintain costswhile sustaining the activity necessary to continue meeting the demands <strong>of</strong> the scientific user base. Topics include: balancing yourcore budget, non-compensatory approaches to acknowledge high performing staff and foster pr<strong>of</strong>essional development, negotiatingservice contracts, money saving tips, and decision making strategies for in-house or outsourcing analyses.Workshop AgendaSatellite EducationalWorkshops7:00 am – 6:00 pm Registration Open — Level 2, Ballroom Level Registration Desk7:00 am – 8:00 am Continental Breakfast — Level 2, Grand Oaks Ballroom P/Q8:00 am – 8:05 am Introductions and Workshop Overview8:05 am – 9:00 am Cost Savings via Cost Avoidance: Quality Management for the Core LaboratoryPresenter: Belynda Hicks, SAIC-Frederick, National Cancer Institute at Frederick9:00 am – 10:00 am Novel Institutional Approaches to Managing Core Facilitiesduring Difficult Economic TimesPresenter: Rand Haley, Huron Consulting Group10:00 am – 10:30 am Morning Refreshment Break — Level 2, Grand Oaks Ballroom P/Q10:30 am – 11:30 am Building Synergistic Relationships with Institutional Leadership that Pays OffPresenter: Nicholas Ambulos, University <strong>of</strong> Maryland School <strong>of</strong> Medicine11:30 am – 12:00 pm Non-compensatory Approaches to Acknowledging High Performing Staffand Fostering Pr<strong>of</strong>essional DevelopmentPresenter: Tim C. Hunter, University <strong>of</strong> Vermont12:00 pm – 1:00 pm Lunch — Level 2, Grand Oaks Ballroom P/Q1:00 pm – 2:00 pm How to Develop and Balance your Core BudgetPresenter: Val Scott, The Jackson Laboratory2:00 pm – 2:30 pm Can I Afford that Service Contract? Getting the Most for the LeastPresenter: Tim C. Hunter, University <strong>of</strong> Vermont2:30 pm – 3:00 pm Afternoon Refreshment Break — Level 2, Grand Oaks Ballroom P/Q3:00 pm – 3:30 pm <strong>Association</strong> <strong>of</strong> Laboratory Managers: Promoting Excellence in Laboratory ManagementPresenter: Wayne Collins, Agilent Technologies3:30 pm – 4:30 pm Interactive Roundtable Discussion on Lean Management in Core FacilitiesPanelists: Belynda Hicks, SAIC-Frederick, National Cancer Institute at Frederick, Nicholas Ambulos, University <strong>of</strong> MarylandSchool <strong>of</strong> Medicine, Val Scott, The Jackson Laboratory, Rand Haley, Huron Consulting Group, Wayne Collins, AgilentTechnologies, and Tim C. Hunter, University <strong>of</strong> Vermont32 • <strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine


NotesNotes<strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine • 33


Plenary Session AbstractsPlenary SessionAbstracts(PS1) Personalized Medicine: Opportunitiesand ChallengesR. KucherlapatiHarvard Medical School Department <strong>of</strong> Genetics andPr<strong>of</strong>essor, Department <strong>of</strong> Medicine, Brigham and Women’sHospital, Boston, MA, United StatesThe completion <strong>of</strong> the sequencing <strong>of</strong> the human genome has augureda new era in genetics. The genome program not only provided uswith the complete sequence but also lead to a revolution in manytechnologies that are critical to biology and medicine. The cost <strong>of</strong>DNA sequencing has gone down significantly and is expected to godown further. This technology is enabling us to examine genetic andgenomic differences among and between individuals at a depth thathas not been possible before. The application <strong>of</strong> genetic and genomicknowledge to assess disease risk, to accurately diagnose disease and tohelp with determining the appropriate therapy is called personalizedmedicine. Our understanding about human genetic variation and howthis influences risk <strong>of</strong> many complex disorders is increasing at a rapidpace. Assessing individual risk may help prevent or postpone the onset<strong>of</strong> human diseases. The genetics <strong>of</strong> human disease is complex and theuse <strong>of</strong> genetic information may help us better diagnose disease that inturn may alter management <strong>of</strong> the disease. In the case <strong>of</strong> cancer, somaticgenetic changes are helping stratify patients and helping choose themost appropriate drug or therapy to which the patients is most likelyto respond. All <strong>of</strong> these changes are bringing in a new era whereimplementation <strong>of</strong> the principles <strong>of</strong> personalized medicine wouldresult in better health for our population at an affordable cost.(PS2) Unlocking Biomarker Discovery: UnbiasedHuman Proteomics at High Scale, Sensitivity,and AccuracyL. GoldSomaLogic, Inc., Boulder, CO, United StatesUnbiased human proteomics should be used for biomarker discovery,given that one’s capacity to deduce (from the literature) sensiblebiomarkers for disease is severely constrained by our limitedknowledge <strong>of</strong> human biology. Empiricism will rule the field for yearsto come. We have worked for more than a decade on building aproteomics platform that allows such empiricism. The platform utilizesa special class <strong>of</strong> aptamers, called SOMAmers that have enhancedaffinities and specificities toward their target proteins. SOMAmerperformance is sufficient to allow our platform to operate without pairs<strong>of</strong> affinity reagents for each target protein — in other words, we do notuse “sandwiches.” Today we measure more than 1,000 human proteinsfrom human samples (plasma, serum, tissue extracts, or other matrices),using only a small volume <strong>of</strong> sample. The measurements have limits<strong>of</strong> detection similar to those <strong>of</strong> good ELISA’s, and low CV’s (around5%). We anticipate additional content in the near future — as newcontent is added to the platform, performance appears to get better.To date we have analyzed more than 10,000 samples from individuals,and studied a number <strong>of</strong> diseases. In most cases we have found novel(and unexpected) biomarkers, which, when used together in a smallpanel, <strong>of</strong>fer hopes for accurate diagnostic information for patients andphysicians. The first diagnostic products will enter the market soon.(PS3) En Route to the Era <strong>of</strong> Genomic MedicineE.D. GreenNational Human Genome Research Institute, Bethesda, MD,United StatesThe Human Genome Project s completion <strong>of</strong> the human genomesequence in 2003 was a landmark scientific achievement <strong>of</strong> historicsignificance. It also signified a critical transition for the field <strong>of</strong> genomics,as the new foundation <strong>of</strong> genomic knowledge started to be used inpowerful ways by researchers and clinicians to tackle increasinglycomplex problems in biomedicine. To exploit the opportunitiesprovided by the human genome sequence and to ensure the productivegrowth <strong>of</strong> genomics as one <strong>of</strong> the most vital biomedical disciplines <strong>of</strong>the 21st century, the National Human Genome Research Institute(NHGRI) is pursuing a broad vision for genomics research beyond theHuman Genome Project. This vision includes facilitating and supportingthe highest-priority research areas that interconnect genomics tobiology, to health, and to society. Current efforts in genomics researchare focused on using genomic data, technologies, and insights toacquire a deeper understanding <strong>of</strong> biology and to uncover the geneticbasis <strong>of</strong> human disease. Some <strong>of</strong> the most pr<strong>of</strong>ound advances arebeing catalyzed by revolutionary new DNA sequencing technologies;these methods are already producing prodigious amounts <strong>of</strong> DNAsequence data, including from large numbers <strong>of</strong> individual patients.Such a capability, coupled with better associations between geneticdiseases and specific regions <strong>of</strong> the human genome, are acceleratingour understanding <strong>of</strong> the genetic basis for complex genetic disordersand for drug response. Together, these developments will usher in theera <strong>of</strong> genomic medicine.(PS4) Selventa Development <strong>of</strong> a TherapeuticDiagnostic TM Stratification Biomarker for DrugResponse in Ulcerative ColitisD. de GraafSelventa, Cambridge, MA, United StatesUsing a gene expression data set from Ulcerative Colitis patientstreated with Infliximab with objective response criteria as input dataand our proprietary Selventa analytics, we have developed a diseaseclassifier which identifies alternative molecular mechanisms driving thedisease beyond TNF-alpha, the target <strong>of</strong> Infliximab. This TherapeuticDiagnostic TM classifier identifies at least 3 more strong, independentdisease drivers, including IL-6, and provides the opportunity to stratifypatient a priori by molecular disease mechanism and associatedtreatment(s). In the case <strong>of</strong> IL6 we provide supporting evidence in arelated disease that indicates the complementary nature <strong>of</strong> the TNFalphaand IL-6 disease driving mechanisms, as a partial validation <strong>of</strong> theapproach. This approach does not rely on response data to developclassifiers and is easily generalized to deliver patient stratification forany specific disease population in Early Discovery with an optimizedproject portfolio as a result.34 • <strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine


VISIT BOOTH 601 TO LEARN MORE!genetic research mattersdemand accuracyIntroducing the NEW 5500 Series SOLiD SequencersDiscover what comes from accuracy. The new, totally redesigned 5500 Series SOLiD Sequencers—now every lab can access a translational research platform with 99.99% accuracy.• Accurate detection <strong>of</strong> somatic mutations or rare genetic variants• Rapid and cost effective sample processing with flexible micr<strong>of</strong>luidics FlowChip• Simple adoption into your lab with streamlined data analysis and a robust, easy to use platformThere’s a lot riding on the quality <strong>of</strong> your results.That’s why you need the 5500 Series SOLiD Sequencer in your lab.go to www.appliedbiosystems.com/solid5500DOWNLOAD THE FREE MOBILE APP AT HTTP://GETTAG.MOBI scan the barcodeto instantly access more information about the 5500 Series SOLiD SequencersLife Technologies <strong>of</strong>fers a breadth <strong>of</strong> products DNA | RNA | protein | cell culture | instrumentsFOR RESEARCH USE ONLY. NOT INTENDED FOR ANY ANIMAL OR HUMAN THERAPEUTIC OR DIAGNOSTIC USE.© 2010 Life Technologies Corporation. All rights reserved. The trademarks mentioned herein are the property <strong>of</strong> Life Technologies Corporation or their respective owners, unless otherwise noted.


Scientific Session AbstractsScientific SessionAbstracts(S1) Improving Human Health from the GroundUpT.W. ThannhauserUnited States Department <strong>of</strong> Agriculture/AgriculturalResearch Service, Ithaca, NY, United StatesThe focus <strong>of</strong> this scientific session will be the essential link betweenagriculture and human health that is grounded in biotechnology.Advances in medicine, in particular personalized medicine, willultimately demand equal and outstanding advances in agriculturalbiotechnologies to occur in parallel. Such advances promise tomake highly nutritious food accessible and affordable to most <strong>of</strong> theworld’s population. Speakers will describe the ways in which humansrely on agriculture, and increasingly, on agricultural technologies, tomeet their basic nutritional needs. Direct applications <strong>of</strong> agriculturalbiotechnologies for crop improvement, agricultural diversification andthe production <strong>of</strong> vaccines and other protein therapeutics will alsobe presented. Members <strong>of</strong> the <strong>ABRF</strong> can help to meet the growingdemand <strong>of</strong> agricultural research scientists for analytical advances andthroughput via the application <strong>of</strong> state-<strong>of</strong>-the-art biotechnologies toagricultural research.(S1-1) Vaccine and Therapeutic Protein Manufacturein PlantsT.E. RyaniBio, Inc., Newark, DE, United StatesThe need to contain costs, as well as the desire to expand patient accessto biotherapeutics has increased interest in non-traditional methods<strong>of</strong> recombinant protein expression. The production <strong>of</strong> vaccines andbiotherapeutic proteins in whole plants holds the promise <strong>of</strong> dramaticallylowering the capital and operating costs for the manufacture <strong>of</strong> lifesavingdrugs, and can also be used in developing nations that lack asophisticated drug manufacturing infrastructure. A transient expressionsystem in whole plants using a combination Agrobacterium/viral genevector (iBio Launch TM ) has been developed to manufacture vaccinesand biotherapeutics in a variety <strong>of</strong> plant species (including Nicotianabenthamiana), and has been successfully scaled to pilot plant levels<strong>of</strong> plant biomass (50kg). The speed <strong>of</strong> protein production in thissystem makes it highly effective for personalized vaccines or vaccinesthat counter pandemic threats. Influenza vaccines for H 5N 1and H 1N 1isolates generated in whole plants using this system have been shownto raise neutralizing antibodies in animal challenge models, and are nowin human clinical trials.(S1-2) Improving Human Nutrition from the GroundUp: Linking Agriculture to Human HealthR.M. WelchDepartment <strong>of</strong> Food Science, Cornell University, Ithaca, NY,United StatesMalnutrition is the leading cause <strong>of</strong> death globally. Both overt nutrientdeficiencies and diet-related chronic diseases account for over 20million deaths a year. The causes <strong>of</strong> malnutrition are complex but arerooted in dysfunctional food systems dependent on agricultural systemsthat have never had an explicit goal <strong>of</strong> improving human health. Thesedeaths are preventable. Linking agricultural systems to human healthcould provide sustainable solutions to malnutrition. Various agriculturaltools can be used to improve the health and felicity <strong>of</strong> people afflictedwith malnutrition. Bi<strong>of</strong>ortification is one tool that is currently beingemployed to address micronutrient malnutrition among resourcepoorfamilies in the developing world. Fertilizers provide another toolthat has been used successfully to address selenium, iodine and zincdeficiencies in several nations. There are numerous other “<strong>of</strong>f the shelf”agricultural tools that could be used to improve the nutrient output<strong>of</strong> farming systems and improve the health all people dependent onagricultural systems for their sustenance. These include: designingcropping systems to maximize nutrient output, using agronomicpractices to improve the nutritional and health promoting quality <strong>of</strong>food crops, re-diversifying cropping systems, and genetically modifyingcrops to be more nutritious and healthy. This can only be accomplishedif explicit links are made between the agriculture, nutrition and healthcommunities. Further, government policies should be reoriented toreflect the important roles that agriculture plays in the health <strong>of</strong> allpeople. We need to closely link agriculture to health if we want to findsustainable solutions to malnutrition globally.(S1-3) A Food Systems Approach to AddressPoor Nutritional Health in Both Advanced andDeveloping EconomiesR.D. GrahamFlinders University <strong>of</strong> South Australia, Adelaide, SouthAustraliaSince the retreat <strong>of</strong> the oceans, the land surface <strong>of</strong> planet Earth has forplant growth become widely deficient in nitrogen and phosphorus andto a smaller extent, potassium and sulphur. A century ago, agriculturewas focused on adding these nutrients to solve production problems<strong>of</strong> otherwise good soils exhausted from many hundreds <strong>of</strong> years<strong>of</strong> cultivation and <strong>of</strong> new, more marginal lands being brought intoproduction. The use <strong>of</strong> mineral fertilisers such as Chilean saltpetre,superphosphate and potash, together with the other then-knownessential minerals, calcium and magnesium, brought production upto expectations, but to experienced eyes, anomalous results hinted atlimitations to production as yet unknown. The micronutrients, as theybecame to be known, contributed greatly to the 20th century cropproductivity. It is estimated that <strong>of</strong> the agricultural soils <strong>of</strong> the world,49% were low in zinc, 31% low in boron, 15% low in molybdenum, 14%low in manganese, 10% low in copper and just 3% deficient in iron. Thepost war explosion in the human population put great stress on our36 • <strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine


food systems in the 1950s but the Green Revolution (1960-1980) morethan doubled world food production and mass starvation was avoided.Instead, there was in the 1980s and 90s a massive rise in micronutrientdeficiencies, especially in subsistence farming systems, which are stillunresolved today. It appears that the Green Revolution emphasison cereal production at the expense <strong>of</strong> pulses and other nutrientrichfoods is the cause, and new food systems capable <strong>of</strong> addressingall human nutritional needs are being developed, a multifactorialchallenge. Dealing with the complex nutritional requirements <strong>of</strong> newand effective food systems for the future is demanding new analyticalcapability to support plant breeders and agronomists in the field.(S2) Integration <strong>of</strong> Bioinformatics for Genomicsand Proteomics Data(S2-1) Integrative Bioinformatics for Genomics andProteomicsC.H. WuCenter for Bioinformatics and Computational Biology,University <strong>of</strong> Delaware, Newark, DE, United StatesSystems integration is becoming the driving force for 21st centurybiology. Researchers are systematically tackling gene functions andcomplex regulatory processes by studying organisms at different levels<strong>of</strong> organization, from genomes and transcriptomes to proteomesand interactomes. To fully realize the value <strong>of</strong> such high-throughputdata requires advanced bioinformatics for integration, mining,comparative analysis, and functional interpretation. We are developinga bioinformatics research infrastructure that links data mining withtext mining and network analysis in the systems biology contextfor biological network discovery. The system features include: (i)integration <strong>of</strong> over 100 molecular and omics databases, along with gene/protein ID mapping from disparate data sources; (ii) data mining andtext mining capabilities for literature-based knowledge extraction; and(iii) interoperability with ontologies to capture functional properties<strong>of</strong> proteins and complexes. The system further connects with a dataanalysis pipeline for next-generation sequencing, linking genomics datato functional annotation. The integrative approach will reveal hiddeninterrelationships among the various components <strong>of</strong> the biologicalsystems, allowing researchers to ask complex biological questions andgain better understanding <strong>of</strong> biological and disease processes, therebyfacilitating target discovery.(S2-2) Integrated Bioinformatics for MS-BasedProteomicsE. DeutschInstitute for Systems Biology, Seattle, WA, United StatesA typical tandem mass spectrometry (MS/MS) proteomics workflowinvolves a series <strong>of</strong> steps including format conversion, spectrumidentification, peptide validation, protein inference, quantification,interpretation, and public repository deposition. This talk will providean overview <strong>of</strong> the proteomic bioinformatics resources developed atthe Institute for Systems Biology, covering the Trans-Proteomic Pipeline(TPP) and related tools, the PeptideAtlas public repository, and theemerging SRMAtlas resource. The TPP provides an easily-installablesuite <strong>of</strong> tools to enable users to perform nearly all steps in an MS/MSanalysis workflow. PeptideAtlas is a multi-species public compendium<strong>of</strong> peptide and protein identifications assembled from a large number<strong>of</strong> uniformly processed MS/MS experiments, along with tools touse the information in a variety <strong>of</strong> ways. SRMAtlas is a resource thatenables the design <strong>of</strong> selected reaction monitoring (SRM) experimentsbased on information from several different sources. In addition, theinterface <strong>of</strong> these resources with community standardization andcooperation efforts such as the Proteomics Standards Initiative and theProteomeXchange Consortium will be presented.(S2-3) Skate Genome Project: Cyber-EnabledBioinformatics CollaborationJ. VincentUniversity <strong>of</strong> Vermont, Burlington, VT, United StatesThe Skate Genome Project, a pilot project <strong>of</strong> the North EastCyberinfrastructure Consortium, aims to produce a draft genomesequence <strong>of</strong> Leucoraja erinacea, the Little Skate. The pilot projectwas designed to also develop expertise in large scale collaborationsacross the NECC region. An overview <strong>of</strong> the bioinformatics andinfrastructure challenges faced during the first year <strong>of</strong> the project willbe presented. Results to date and lessons learned from the perspective<strong>of</strong> a bioinformatics core will be highlighted.(S3) Single-Cell Environmental Genomics(S3-1) Early Elements <strong>of</strong> a Pipeline for SingleBacterial Cell GenomicsD.A. RelmanDepartments <strong>of</strong> Microbiology & Immunology, and <strong>of</strong>Medicine, Stanford University, Stanford, CA, United StatesA critical aspect <strong>of</strong> microbial community analysis is to understandthe contributions <strong>of</strong> individual cells to overall community structureand function. Together with Steve Quake and Paul Blainey from theDepartment <strong>of</strong> Bioengineering, we have built a micr<strong>of</strong>luidics device andplatform that permits sequence-based identification <strong>of</strong> single bacterialcells using fluorescence in situ hybridization, single cell sorting usinglaser tweezers, and subsequent on-chip whole genome amplification(WGA). Current trapping performance enables the sorting <strong>of</strong> up to 60cells per hour, and complete processing <strong>of</strong> 30 - 46 cells, 0 – 16 negativecontrols, and two positive controls per day on a routine basis. Eachdevice enables 48 simultaneous single-cell WGA reactions, which canbe monitored in real-time with SYBR green fluorescence. In conjunctionwith other developments described in this Session, we envision a nearfuturecapability for microbial community genomic analysis at a level <strong>of</strong>resolution not previously imagined.(S3-2) Assembling Complete Genomes fromComplex MixturesF. MeyerArgonne National Laboratory, Mathematics and ComputerScience Division, Argonne, IL, United StatesCharacterizing genomes <strong>of</strong> un-culturable microbes (most specieson earth) requires new approaches for genome assembly fromenvironmental or biomedical samples that <strong>of</strong>ten contain manyhundreds or thousands <strong>of</strong> species. Here we show we can reconstructthe complete genome (Candidatus Sulfuricurvum sp) via short-readmetagenomics and novel approaches for assembly based on simpleScientific SessionAbstracts<strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine • 37


Scientific SessionAbstractsstatistical principles. While previous examples <strong>of</strong> complete genomesequences from metagenomes stem from samples <strong>of</strong> very limitedcomplexity (>10 OTUs), this sequence was obtained from a complexmix <strong>of</strong> over 300 OTUs. The reason traditional genome assemblyapproaches fail are varying abundance levels and strain variation andwe show a simple approach to overcome those hurdles. This novelapproach will allow the assembly <strong>of</strong> genomes and via metabolic modelsderived from the genomic sequences, hopefully the cultivation <strong>of</strong> keyspecies from diverse environments/enrichment cultures.(S3-3) Dining in with Trillions <strong>of</strong> Fascinating Friends:Exploring Our Human Gut Microbiome in Healthand DiseaseJ.I. GordonCenter for Genome Sciences and Systems Biology,Washington University School <strong>of</strong> Medicine, St. Louis, MO,United StatesOur genetic landscape is a summation <strong>of</strong> the genes embedded inour human genome and in the genomes <strong>of</strong> our microbial symbionts(the microbiome). Similarly, our metabolic features (metabotypes)are an amalgamation <strong>of</strong> human and microbial traits. Therefore,understanding <strong>of</strong> the range <strong>of</strong> human genetic and metabolic diversitymeans that we must characterize our microbiomes, which containat least several hundred-fold more genes than our human genome,as well as the factors that influence the properties <strong>of</strong> our microbialcommunities (microbiota). The results should provide an additionalperspective about contemporary human biology as we assesshow our changing lifestyles, cultural norms, socioeconomic status,and biosphere are influencing our microbial ecology and healthstatus. I will discuss the results <strong>of</strong> our group’s ongoing metagenomicstudies <strong>of</strong> the interrelationships between diet and the structure anddynamic operations <strong>of</strong> the human gut microbiome. We believe thatunderstanding these interrelationships is important for advancing ourappreciation <strong>of</strong> the nutritional value <strong>of</strong> food ingredients, for creatingnew nutritional guidelines for humans at various stages <strong>of</strong> their lifespan,and for developing new ways to deliberately manipulate the properties<strong>of</strong> the gut microbiota to prevent or treat various diseases. We havedeveloped a translational medicine pipeline that involves metagenomicanalyses <strong>of</strong> the gut microbial communities <strong>of</strong> adult mono- and dizygotictwins living in the USA who are lean, or concordant or discordant forobesity, and twins aged 0-3 years living in developing countries whodevelop normally, or who become malnourished and are treated witha ready-to-use therapeutic food (RUTF). Intact fecal communities fromthese individuals, or ‘personal’ culture collections that capture themajority <strong>of</strong> bacterial diversity in their microbiota, are then transplantedinto germ-free mice, which are fed the diets <strong>of</strong> the human donors, orsystematically manipulated derivatives <strong>of</strong> these diets. The impact <strong>of</strong>diet and microbiota on these humanized mice, including the degreeto which the human donor’s physiologic/metabolic phenotypes can betransmitted to gnotobiotic animals via microbiota transplants, are thenstudied using a variety <strong>of</strong> methods.(S4) Epigenetics(S4-1) Next Generation Quantitative ProteomicTools for Analyzing Histone ModificationsB.A. GarciaPrinceton University, Princeton, NJ, United StatesHistones are small proteins that package DNA into chromosomes,and a large number <strong>of</strong> studies have showed that several singlepost-translational modification sites on the histones are associatedwith both gene activation and silencing. Nevertheless, what type<strong>of</strong> effect distinct combinations <strong>of</strong> simultaneously occuring histonemodifications (Histone Codes or patterns) have upon cellular eventsis poorly understood. The main reason for this lack <strong>of</strong> knowledge isthat robust high-throughput methods for quantitative characterizationor even qualitative identification <strong>of</strong> combinatorial Histone Codesby any standard biological, immunological or physical techniquedo not exist. We plan to specifically address this deficiency bydeveloping novel mass spectrometry based proteomic methods andaccompanying bioinformatics to quantitatively characterize molecularlevel descriptions <strong>of</strong> combinatorial Histone Codes, and apply thesemethods to study how these dynamic Histone Codes influence geneexpression under different biological conditions. Here we presentinitial proteomics data that describes: (i) high-throughput comparison<strong>of</strong> histone modifications from multiple cellular states (ii) developingmass spectrometry methods for quantitative tracking <strong>of</strong> combinatorialHistone Codes (iii) monitoring in vivo Histone Code dynamics, and(iv) investigating the role <strong>of</strong> Histone Code interpreting proteins inrecognizing distinct Histone Codes. Ultimately, we will work towardsthe goal <strong>of</strong> taking any defined part <strong>of</strong> the genome and accuratelyquantifying the Histone Codes, detecting all the non-histone proteinsthat reside on these distinct pieces <strong>of</strong> chromatin, and then mappingthis proteomic data back to specific genomic locations, therefore takinga proteomic snapshot <strong>of</strong> what that chromosome landscape looks likeduring any nuclear event. These studies in combination with biologicalexperiments will help provide a systems biology outlook on geneexpression that will lay down the basic scientific foundation to advanceseveral applications, such as stem cell reprogramming and cancerprogression.38 • <strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine


(S4-2) Chromatin Dynamics in Melanoma: A Role forMacroH2AA. Kapoor 1,2 , M. Goldberg 1,2 , L. Cumberland 1,2 *,K. Ratnakumar 1,2 *, M. Segura 4,6 , P. Emanuel 2,3 ,S. Menendez 4,6 , C. Vardabasso 1,2 , G. LeRoy 7 , C. Vidal 2,3 †,D. Polsky 4,5,6 , I. Osman 5,6 , B. Garcia 7 , E. Hernando 4,6 ,E. Bernstein 1,21Department <strong>of</strong> Oncological Sciences, Mount Sinai School<strong>of</strong> Medicine, New York, NY, United States; 2 Department<strong>of</strong> Dermatology, Mount Sinai School <strong>of</strong> Medicine, NewYork, NY, United States; 3 Department <strong>of</strong> Pathology, MountSinai School <strong>of</strong> Medicine, New York, NY, United States;4Department <strong>of</strong> Pathology, New York University LangoneMedical Center, New York, NY, United States; 5 Department<strong>of</strong> Dermatology, New York University Langone MedicalCenter, New York, NY, United States; 6 InterdisciplinaryMelanoma Cooperative Group, New York UniversityLangone Medical Center, New York, NY, United States;7Department <strong>of</strong> Molecular Biology, Princeton University,Schultz Laboratory, Princeton, NJ, United States; †PresentAddress: Department <strong>of</strong> Dermatology, Saint Louis UniversitySchool <strong>of</strong> Medicine, St. Louis, MO, United States; *Theseauthors contributed equally to this work.Cancer is a disease consisting <strong>of</strong> both genetic and epigenetic changes.Although increasing evidence demonstrates that tumour progressionentails chromatin-mediated changes such as DNA methylation, therole <strong>of</strong> histone variants in cancer initiation and progression currentlyremains unclear. Histone variants replace conventional histones withinthe nucleosome and confer unique biological functions to chromatin.Using well characterized, paired series <strong>of</strong> murine and human melanomacells lines, we probed the epigenetic pr<strong>of</strong>ile <strong>of</strong> melanoma. Analysis<strong>of</strong> histones from both series using multiplexed quantitative massspectrometryrevealed changes in several histone posttranslationalmodifications and histone variants. The loss <strong>of</strong> mH2A is<strong>of</strong>orms,histone variants generally associated with condensed chromatin andfine-tuning <strong>of</strong> developmental gene expression programs is positivelycorrelated with increasing malignant phenotype <strong>of</strong> melanoma cellsin culture and human tissue samples. Knockdown <strong>of</strong> mH2A is<strong>of</strong>ormsin melanoma cells <strong>of</strong> low malignancy results in significantly increasedproliferation and migration in vitro and growth and metastasis invivo. Restored expression <strong>of</strong> mH2A is<strong>of</strong>orms rescues these malignantphenotypes in vitro and in vivo. We demonstrate that the tumourpromotingfunction <strong>of</strong> mH2A loss is mediated, at least in part, throughdirect transcriptional upregulation <strong>of</strong> CDK8. Suppression <strong>of</strong> CDK8, acolorectal cancer oncogene inhibits proliferation <strong>of</strong> melanoma cells,and knockdown <strong>of</strong> CDK8 in cells depleted <strong>of</strong> mH2A suppresses theproliferative advantage induced by mH2A loss. Moreover, a significantinverse correlation between mH2A and CDK8 expression levels existsin melanoma patient samples. Taken together, our results demonstratethat mH2A is a critical component <strong>of</strong> chromatin that suppresses thedevelopment <strong>of</strong> malignant melanoma, a highly intractable cutaneousneoplasm.(S4-3) Using Protein Domain Microarrays to Read theHistone CodeM.T. BedfordMD Anderson Cancer Center, Department <strong>of</strong> MolecularCarcinogenesis, Smithville, TX, United StatesFor cells to survive, differentiate, and grow, information has to betransferred from the cell surface to the nucleus. This process is referredto as signal transduction. A hallmark <strong>of</strong> cancer is the deregulation <strong>of</strong>signal transduction pathways. Signaling events in eukaryotic cells involvethe assembly and disassembly <strong>of</strong> large protein-protein complexes.These diverse associations are mediated through interactions <strong>of</strong>a limited number <strong>of</strong> modular signaling units or protein-domains.Protein interactions involving domains are <strong>of</strong>ten regulated by posttranslationalmodification (PTM – like phosphorylation, methylationand acetylation) <strong>of</strong> the smaller protein motif within the ligand. Wehave developed a chip-size protein microarray that harbors a display<strong>of</strong> over 300 modular protein-interacting domains including SH2, SH3,PDZ, FHA, 14-3-3, WW, Chromo, Tudor, PHD and MBT domains. Inthe emerging proteomic era, it is becoming easier to identify proteinsusing tryptic digestion followed by mass spectrometric approaches.These same methods also detect sites <strong>of</strong> posttranslational modificationon proteins. Many <strong>of</strong> these posttranslational modifications likelygenerate docking sites for protein modules. We have developedprotein-domain microarray technology to help identify proteins thatcan interact with motifs that are either methylated or phosphorylated.This high-throughput approach facilitates the rapid identification<strong>of</strong> protein-protein interactions in vitro. Further in vivo studies areneeded to confirm that these interactions do indeed occur in biologicalsystems. Protein domains are cloned into a GST expression vector, andrecombinant protein is produced in bacteria. These fusion proteinsare then arrayed onto nitrocellulose coated glass slides using a robot.These slides are probed with biotinylated peptides that are preconjugatedto streptavidin-Cy3. The peptides used in this experimentare synthesized as 15 mers, and both the modified and unmodifiedforms <strong>of</strong> the peptides are tested on the array. In this manner, we canidentify novel methyl- and phospho-dependent interactions. We havebuilt three types <strong>of</strong> arrays: (1) A phospho-tyrosine reader harbors 70SH2 domains and 5 PTB domains (total = 75 domains). (2) A phosphothreonine/serinereader that harbors 7 14-3-3 domains, 5 FHAdomains, 15 BRCT domains and a WW domain (total = 28 domains).(3) An epigenetic reading array that harbors methyl and acetyl readers.This array is composed <strong>of</strong> 50 tudor domains, 22 bromo domains, 36PHD domains, 17 MBT domains, 11 WD40 domains, 9 SANT domains,28 chromo domains, 15 PWWP domains, 5 BRK domains, 5 CWdomains, and 9 Ank repeats (total = 207 domains). More and moreposttranslational modifications are being discovered on proteins. Theroles <strong>of</strong> many <strong>of</strong> these methylation and phosphorylation events <strong>of</strong>tenremain obscure. This approach provides an easy way for a researcherto identify potential binding partners for their favorite proteins.These arrays thus <strong>of</strong>fer researchers tools to get at “mechanism”. Onceinvestigators know that they are working with a clearly functional PTM,they can proceed with confidence to generate modification specificantibodies and interrogate the signaling pathway that is engaged bythe identified PTM-driven protein-protein interaction.Scientific SessionAbstracts<strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine • 39


Scientific SessionAbstracts(S5) High-Throughput Genome Centers(S5-1) Overview <strong>of</strong> the Illumina Sequencing Platformat the Broad InstituteK. ConnollyProcess & Technology Development, Genome SequencingPlatform, The Broad Institute, Cambridge, MA, United StatesThe constant increase in quality and quantity <strong>of</strong> Next-Generationsequencing data necessitates a parallel growth in sample preparationand a scalable tracking system. The Broad Institute’s Illumina SequencingPlatform handles a variety <strong>of</strong> applications and comprises Illumina’s latesthardware, s<strong>of</strong>tware and kit releases, and a high-throughput samplepreparation process. With our automated sample preparation and QCprocesses, we have been able to meet our increased capacity goals <strong>of</strong>up to 3,840 libraries per week, and have reduced our rework rate to5% through attaining target cluster densities with high reproducibility.We continue to work closely with Illumina to develop the sequencingtechnology, using data to drive process improvements and exploringmethods to improve GC bias. Maximizing platform-wide efficiency ispossible through the implementation and continuous development <strong>of</strong>tools for process quality control and centralized communication, suchas our real-time run monitoring dashboard and JIRA tracking system.We were able to convert rapidly from GAIIxs to HiSeq2000s byestablishing and using an enterprise Knowledge Management system,with which we can efficiently accumulate and disseminate a changingknowledge base. All <strong>of</strong> these improvements are applicable to both theGA and HiSeq platforms.(S5-2) Science and Technology at a HighThroughput Genome CenterL. Fulton, R. Wilson, The Genome Center ProductionGroupThe Genome Center at Washington University School <strong>of</strong>Medicine, St. Louis, MO, United StatesThe Genome Center (GC) at Washington University School <strong>of</strong> Medicinehas developed a state <strong>of</strong> the art genomics facility. Our scientists workon a variety <strong>of</strong> cutting edge projects with researchers from aroundthe world. These collaborative research projects lead to cutting edgeadvances in the field <strong>of</strong> genomics. The structural organization at theGC reflects these efforts and is centered around six major scientificareas: Transcriptome Sequencing, Genome Assembly, Whole GenomeSequencing, Human Microbiome, Human Genetics, and TargetedResequencing. These specific scientific areas are supported by onecentral data production pipeline. Attributes <strong>of</strong> this pipeline includedetailed sample screening protocols, sample barcoding capabilitiesthat allow for a broad range <strong>of</strong> sample cohorts, multiplatform dataproduction, and the ability to select from more than one method <strong>of</strong>sequencing strategies. All <strong>of</strong> this is supported by one centralized LIMSgroup dedicated to maintaining and developing the data productioncapabilities. The technology development group investigates newtechniques and instrumentation prior to any changes in the main dataproduction pipeline. Only robust protocols and instrumentation areallowed into the data production pipeline. This strategy allows TheGenome Center to run a base data production pipeline while constantlyinfusing high quality advances. Sequence data for each project is sentinto an advanced analysis pipeline built to conduct a multitude <strong>of</strong>assessments. When needed, validation (a second sequence event) canbe used to confirm variants detected by the analysis s<strong>of</strong>tware.(S5-3) High-Throughput Next GenerationSequencing Methods and ApplicationsD. Muzny 1 , M. Wang 1 , I. Newsham 1 , Y.Q. Wu 1 , H. Dinh 1 ,C. Kovar 1 , J. Santibanez 1 , A. Sabo 1 , J. Reid 1 , M. Bainbridge 1 ,E. Boerwinkle 2 , T. Albert 3 , R. Gibbs 11Baylor College <strong>of</strong> Medicine, Human Genome SequencingCenter, Houston, TX, United States; 2 University <strong>of</strong> TexasHealth Science Center at Houston, School <strong>of</strong> Public Health,Houston, TX, United States; 3 Roche NimbleGen, Inc.,Madison, WI, United StatesSecond Generation high-throughput sequencing technologies haverevolutionized the genome sequencing applications and will ultimatelyhave great impact on personalized medicine. The increase in capacity<strong>of</strong> both the AB/Life Technologies SOLiD 4.0 and Illumina HiSeqinstrumentation and the ability <strong>of</strong> the platforms to multiplex sampleshas led to process innovations impacting many ongoing projectsat the HGSC. Applications have ranged from regional and wholeexome capture sequencing to the use <strong>of</strong> whole genome shotgun fordeep coverage and determining structural rearrangements. Internaladvancements have complemented the higher capacity instrumentationthrough the implementation <strong>of</strong> library automation, low DNA inputsamples, capture hybridization multiplexing and application <strong>of</strong> readmapping tools such as BFAST and BWA. Development <strong>of</strong> sample intakeprocedures, LIMS tracking and defined reporting metrics has enabledNexGen sequencing pipelines that can effectively deliver targetedand whole genome shotgun data for thousands <strong>of</strong> samples. Thesetechnical advancements to the pipeline have allowed us to achievea rate <strong>of</strong> ~1500 libraries/captures per month. To date the center hascompleted over 5000 exome and regional capture libraries for TheCancer Genome Atlas (TCGA), NIMH Autism, Cohorts for Heart andAging Research in Genomic Epidemiology (CHARGE-S) and 1000Genomes Project. Development <strong>of</strong> these applications and methods willbe discussed along with key data metrics, process management andpipeline organization.(S6) Strategies for Deep Mining <strong>of</strong> ComplexProtein Mixtures(S6-1) Coverage and Recovery <strong>of</strong> Upstream ProteinFractionation Methods in LC-MS/MS WorkflowsL.J. FosterCentre for High-Throughput Biology, The University <strong>of</strong> BritishColumbia, Vancouver, BC, CanadaThe proteome <strong>of</strong> any cell or even any subcellular fraction remainstoo complex for complete analysis by one dimension <strong>of</strong> liquidchromatography-tandem mass spectrometry (LC-MS/MS). Hence, toachieve greater depth <strong>of</strong> coverage for a proteome <strong>of</strong> interest, mostgroups routinely subfractionate the sample prior to LC-MS/MS sothat the material entering LC-MS/MS is less complex than the originalsample. Protein and/or peptide fractionation methods that biochemistshave used for decades, such as strong cation exchange chromatography(SCX), isoelectric focusing (IEF) and SDS-PAGE, are the most commonprefractionation methods used currently. There has, as yet, been nocomprehensive, controlled evaluation <strong>of</strong> the relative merits <strong>of</strong> thevarious methods, although some binary comparisons have been made.We will discuss the most popular methods for fractionating samples atboth the protein and peptide level, demonstrating quantitatively which40 • <strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine


are the best methods for optimal recovery and proteome coverage.A novel approach for fractionating samples at the level <strong>of</strong> proteincomplexes will also be discussed.(S6-2) Improving the Comprehensiveness <strong>of</strong> Large-Scale Proteomics Experiments Using AdvancedComputational Tools and Accurate MultipleHypothesis Testing StatisticsM.J. MacCoss, J. Egertson, B. Frewen, L. Käll, W. NobleDepartment <strong>of</strong> Genome Sciences, University <strong>of</strong> Washington,Seattle, WA, United StatesMass spectrometry based technology for the analysis <strong>of</strong> complexprotein mixtures has improved at an amazing rate. With each newinstrument release, mass spectrometers have become more sensitiveand have faster MS/MS data acquisition speeds. Furthermore,instruments are continuously improving the dynamic range, massaccuracy, and resolution <strong>of</strong> the resulting mass spectrometry data. All<strong>of</strong> these developments have increased the number <strong>of</strong> peptides thatcan be identified and quantified without extending the overall analysistime. While the technological hardware advances that are required toincrease the number <strong>of</strong> peptide identifications by 50% with a constantanalysis time is monumental, we have been able to demonstrate thatincrease in performance without increasing the analysis time at all. Toaccomplish this, we have made use <strong>of</strong> improved database searchingalgorithms, spectrum library searching, use <strong>of</strong> chromatographicretention time, powerful machine learning tools, accurate multiplehypothesis testing statistics, and many more. Strategies will bediscussed on how to increase the comprehensiveness <strong>of</strong> any datasetusing improved data analysis strategies.(S6-3) In-Depth Analysis <strong>of</strong> Human and MousePlasma Using 3-D And 4-D Fractionation StrategiesD.W. SpeicherThe Wistar Institute, Philadelphia, PA, United StatesIn-depth pr<strong>of</strong>iling <strong>of</strong> plasma proteomes can potentially identify noveldisease biomarkers. But few biomarkers identified by proteomicapproaches have advanced to early-stage clinical testing because they<strong>of</strong>ten are not sufficiently disease specific. Major challenges in plasmaproteome analysis include the very wide dynamic range <strong>of</strong> proteinconcentrations, the high protein complexity, and the substantialheterogeneity <strong>of</strong> most protein concentrations in the normal humanpopulation. Because most disease-specific biomarkers are present inblood at very low concentrations, extensive fractionation is requiredprior to LC-MS/MS analysis. In general, more fractionation will result ingreater depth <strong>of</strong> analysis, but there is a point <strong>of</strong> diminishing return foreach fractionation method and throughput decreases as the number<strong>of</strong> LC-MS/MS runs per proteome increases. A common feature <strong>of</strong> mostcurrent plasma pr<strong>of</strong>iling methods is to first immunodeplete as manyhigh abundance plasma proteins as possible, followed by extensiveprotein fractionation <strong>of</strong> the depleted plasma prior to trypsin digestionand LC-MS/MS. In addition, reliable quantitative comparisons areneeded for most types <strong>of</strong> studies. While all quantitative methods havestrengths and weaknesses, label-free quantitative comparison <strong>of</strong> LC-MSsignals is increasing in popularity and seems adequately reproduciblefor most studies. Our laboratory commonly uses two alternative plasmaproteome analysis strategies. One powerful approach utilizes a 3-Dprotein/peptide pr<strong>of</strong>iling method consisting <strong>of</strong> depleting 20 abundantproteins followed by 1-D SDS PAGE, fractionation <strong>of</strong> the gel lane into20 to 60 fractions and LC-MS/MS analysis. Proteins can be quantitativelycompared using label-free analysis <strong>of</strong> ion current patterns from the MSfull scans. An even greater depth <strong>of</strong> analysis can be achieved using a4-D protein/peptide pr<strong>of</strong>iling strategy utilizing microscale solutionisoelectr<strong>of</strong>ocusing <strong>of</strong> proteins prior the SDS gel in the 3-D scheme,although throughput is substantially reduced.(S7) Next Generation Sequencing Technologieson the Horizon(S7-1) Towards Optical DNA Sequencing UsingNanopore ArraysA. MellerDepartment <strong>of</strong> Biomedical Engineering, Boston University,Boston, MA, United StatesNext generation DNA sequencing methods that utilize nanometersizepores have been subject <strong>of</strong> numerous studies in past years. One<strong>of</strong> the compelling features <strong>of</strong> the nanopore technique lies in its abilityto electrophoretically focus long DNA strands towards the pore areaand thread the molecules inside the pore in a highly efficient manner.Thus extremely small copy numbers <strong>of</strong> the target DNA are requiredfor analyses†, circumventing the need for costly and time-consumingtarget amplification. One <strong>of</strong> the major bottlenecks for the realization<strong>of</strong> a viable nanopore-based DNA sequencing has been the ability tosimultaneously read the electrical signals from hundreds to thousands<strong>of</strong> nanopores densely fabricated on sub-millimeter size silicon chip. Toaddress this issue we develop an extremely high throughput singlemoleculeDNA sequencing technique, which employs optical, widefieldreadout from DNA molecules electrically mobilized throughthe nanopores. Our method consists <strong>of</strong> two steps: First, target DNAmolecules are converted according to pre-determined code, which isrecognized by molecular beacons with four types <strong>of</strong> fluorophores (eachuniquely corresponding to one <strong>of</strong> the four DNA bases). Solid-statenanopores are then used to sequentially strip <strong>of</strong>f the beacons, leadingto a series <strong>of</strong> photon bursts that can be detected with a custom mademicroscope. Notably the method circumvents the use <strong>of</strong> enzymes in thereadout stage, and is thus not affected by their limited processivity andlifetime. Here we demonstrate the feasibility <strong>of</strong> this method using a twocolor model system, and show for the first time, individual nucleotiderecognition from multiple nanopores simultaneously‡, allowingstraightforward parallelization <strong>of</strong> our system to nanopore arrays. †.Wanunu, M., W. Morrison, Y. Rabin, A. Y. Grosberg, and A. Meller. 2010.Electrostatic Focusing <strong>of</strong> Unlabeled DNA into Nanoscale Pores using aSalt Gradient. Nature Nanotechnology 5:160-165. ‡. McNally, B., A.Singer, Z. Yu, Y. Sun, Z. Weng, and A. Meller. 2010. Optical Recognition<strong>of</strong> Converted DNA Nucleotides for Single-Molecule DNA SequencingUsing Nanopore Arrays. Nano Letters 10:2237-2244.Scientific SessionAbstracts<strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine • 41


Scientific SessionAbstracts(S7-2) Single Molecule Real-Time DNA Sequencingon the Surface <strong>of</strong> a Quantum-Dot NanocrystalP.B. Vander HornGenetic Systems, Life Technologies, Carlsbad, CA, UnitedStatesA single molecule, long read-length, real-time sequencingby-synthesistechnology has been developed by buildinga sequencer directly on the surface <strong>of</strong> a ~ 10 nm quantumdotnanocrystal. Fluorescence resonance energy-transfertechnology (FRET) is utilized for DNA sequence detection, inwhich signals from the quantum-dot labeled DNA polymeraseplus 4 DNA-base-specific acceptor dyes are simultaneouslydetected. Precisely engineered sequencing-grade Qdot TMnanocrystals are smaller than current commercially availablematerials (to increase FRET signals), and have an exctinctioncoefficient ~100X greater than organic-dyes, allowing for verylow levels <strong>of</strong> excitation power to be used while sequencing,Acting as the FRET donor, the Qdot TM -polymerase generates acorrelated “photon-dip” for every inserted based (termed the“quantum-correlation-signal”), allowing for more accurate basecalling.Because the sequencer is not physically bound to anysolid substrate, it can be exchanged (like a reagent) during midsequenceruns, effectively replacing damaged non-functioningpolymerases mid-reaction. Each exchange cycle lengthens theeffective read-length <strong>of</strong> the sequencer. In this manner, theread-length can be continuously extended without “gaps”.Expanding upon this flexibility, after sequencing a particularlength <strong>of</strong> DNA, the newly synthesized strand can be selectivelyremoved. The original genomic DNA strand is then re-primed,Qdot TM -polymerase sequencers are rebound, and the identicalgenomic DNA strand can be sequenced again, greatly increasingthe net accuracy and not requiring circularization <strong>of</strong> genomictemplates. In combining these features, the desired accuracyand read-length can be “tuned” by adjusting the number <strong>of</strong>reagent exchange cycles. Because each sequencing reaction canbe completed in minutes, multiple exchange experiments canbe performed per sequencing hour. These Qdot TM -polymerasesequencers can also bind to ultra-long DNA segments (>10kb)at multiple positions along the length <strong>of</strong> the DNA andsequence while moving “horizontally” (parallel to TIRF field),enabling the possibility <strong>of</strong> “ordered-reads” for long-phasedhaplotype sequencing. Examples <strong>of</strong> real-time sequencing <strong>of</strong>homopolymeric, patterned, and complex templates will beshown.(S9) Proteomics StandardizationD.B. FriedmanProteomics Laboratory Mass Spectrometry Research Center,Vanderbilt University, Nashville, TN, United StatesThe needs and issues related to establishing inter-laboratorystandardization in quantitative proteomics will be highlighted bypresentations from three international initiatives. By establishing theneed for standardization, each group will also highlight the tremendousamount <strong>of</strong> instrument and experimental variation that is also measuredwhen trying to determine real biological changes. The Spanish-basedProteoRed consortium addresses multiple proteomics platforms,ranging from MS-based to gel-based. The NIH CPTAC network isfocused on MS-based studies, and the UK-based FixingProteomicsinitiative covers mostly gel-based methods. Results from thesestudies will demonstrate complement and overlap in technology andapproaches used between these groups, all motivated by the universalgoal <strong>of</strong> standardizing these complex technologies across laboratories.(S9-1) A Multi-Laboratory Study AssessingRobustness and Reproducibility <strong>of</strong> PlasmaReference Sample for Benchmarking LC-MS PlatformPerformanceJ.P. Albar 5 , A. Campos 1,4 , E. Oliveira 1,4 ,S. Martínez-Bartolomé 3,4 , F. Canals 2,41Barcelona Science Park, Barcelona, Spain; 2 Vall d’HebronUniversity Hospital Research Institute, Barcelona, Spain;3Centro Nacional de Biotecnologia-CSIC, Madrid, Spain;4ProteoRed Consortium, Spanish National Institute <strong>of</strong>Proteomics, Madrid, Spain; 5 ProteoRed, National Center forBiotechnology-CSIC, Madrid, SpainAn increasingly common request for proteomics core facilities isdetermining qualitative and quantitative differences among clinicalsamples such as plasma, CSF, or urine. One <strong>of</strong> the missions <strong>of</strong> theSpanish Network <strong>of</strong> Proteomics Facilities (ProteoRed-ISCIII) is to assistits proteomics core facilities in evaluating their capabilities to performqualitative and quantitative proteomics analysis. This year, in an attemptto represent a realistic experiment scenario that might be requested toa proteomics core facility, we provided a moderately complex plasmastandard reference sample to be used for routine QC monitoring <strong>of</strong>laboratory instrumentation. The ProteoRed Plasma Reference (PPR)sample is a subset <strong>of</strong> highly abundant well-characterized human plasmaproteins with a number <strong>of</strong> is<strong>of</strong>orms, in addition to 4 spiked-in proteins,altogether distributed over 5 orders <strong>of</strong> magnitude in concentration.The PPR sample was recently stress tested in the latest ProteoRedMulticenter Experiment (PME6) that counted with the participation<strong>of</strong> 17 proteomics facilities using a wide range <strong>of</strong> LC-MS platforms. Werequested the sample be analyzed in a single LC-MS run in experimentaltriplicate (3 different digestions). Evaluation <strong>of</strong> the results submitted bythe study participants revealed moderate discrepancies at the peptideidentification level, and poor overlap at the protein identificationlevel. In an attempt to identify the source <strong>of</strong> such irreproducibility,raw data <strong>of</strong> 8 laboratories (24 LC-MS runs) were reanalyzed centrallyusing a standardized data analysis pipeline, which included proteininference using ProteinProphet s<strong>of</strong>tware. We found that the majority<strong>of</strong> protein identification discrepancies across submitted reports <strong>of</strong>these 8 laboratories were due to inconsistencies on how data analysisand computational tools group and/or infer proteins. Immunoglobulin42 • <strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine


variable chain identifications were particularly conflicting throughoutidentification lists, even in the centralized analysis. Using a series <strong>of</strong>LC-MS performance metrics, we benchmarked the performance <strong>of</strong> 8LC-MS instruments (Orbitraps) and identified system components thatvary the most across laboratories.(S9-2) Clinical Proteomic Technologies for Cancer:NIH-Funded Measurement ScienceC.R. Kinsinger, E. Boja, T. Hiltke, M. Mesri, A. Rahbar,R. Rivers, H. RodriguezNational Cancer Institute, Center for Strategic ScientificInitiatives, Office <strong>of</strong> Cancer Clinical Proteomic Research,Bethesda, MD, United StatesIn 2006, the National Cancer Institute (NCI) launched the ClinicalProteomic Technologies for Cancer initiative (CPTC). The overallmission <strong>of</strong> this initiative was to foster the building <strong>of</strong> an integratedfoundation <strong>of</strong> proteomic technologies, data, analysis systems, andreagents and reference materials to systematically advance theapplication <strong>of</strong> protein science to accelerate discovery and clinicalresearch in cancer. Specifically, the CPTC was charged to addressissues <strong>of</strong> variability and irreproducibility in proteomic measurements.During the past five years, CPTC investigators have focused onassessing proteomic platforms involving mass spectrometry. Interlaboratorystudies have addressed variability in both unbiased andtargeted mass spectrometric methods. These studies have producedreference materials and data, performance metrics, standard operatingprocedures, and guidance for the community on the current ability<strong>of</strong> mass spectrometry for proteomics. Outputs from the technologyassessment aspects <strong>of</strong> CPTC have leveraged additional developments.First, CPTC inter-laboratory studies provided a basis for engaging theFDA on the metrological requirements for approval in vitro diagnosticmultivariateindex assays. Second, the NCI developed a follow-onfunding opportunity that applies the technology pipeline developedin the first phase <strong>of</strong> CPTC.(S9-3) Why Reproducible Outcomes are Essentialin Proteomic Research and Why Standardisation <strong>of</strong>Processes is Essential for Achieving ReproducibilityA. Borthwick, W. DracupNonlinear Dynamics, Newcastle upon Tyne, United KingdomIn both 2D electrophoresis and LC-MS proteomic analysis we are dealingwith highly complex samples, and there are many complex processesinvolved which in turn can be affected by a host <strong>of</strong> parameters and issuessuch as reagent batches, column performance, even the temperature <strong>of</strong>the lab. This complexity means that it can be very difficult to generatethe same results from the same samples in different labs and even inthe same lab at different times. This in turn makes it very difficult forlabs to build upon published results, a fundamental principle <strong>of</strong> thescientific method. Quality Control (QC), based on the use <strong>of</strong> standardsto monitor levels <strong>of</strong> technical variation in industrial processes isfundamental in the output <strong>of</strong> a reproducible product. We argue thatbecause proteomic analysis is significantly more challenging than mostindustrial processes, employing standards and the standardisation <strong>of</strong>processes in proteomics experiments is key to arriving at reproducibleoutcomes. Focusing mainly on 2D electrophoresis and to some extenton LC-MS we examine the importance <strong>of</strong> standardisation and howsuch standards may be applied to Proteomic research in order t<strong>of</strong>acilitate reproducible discoveries. Using studies carried out with singleand multi-users from within and between different laboratories, wedescribe our experiences <strong>of</strong> achieving standardisation using Standardsamples to provide feedback on the reproducibility <strong>of</strong> each stage andas well as the complete proteomic workflow.Scientific SessionAbstracts<strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine • 43


Workshop Session AbstractsWorkshop SessionAbstracts(W1) The Diagnostics Core Facility: Harvestingthe Promise <strong>of</strong> Personalized Medicine(W1-1) Win on Sunday, Sell on Monday: From theExome Sequencing <strong>of</strong> One Boy to the Delivery <strong>of</strong>Clinical DiagnosticsM.R. TschannenHuman and Molecular Genetics Center, Department <strong>of</strong>Physiology, Medical College <strong>of</strong> Wisconsin, Milwaukee, WI,United StatesFor several years, there have been discussions about using bothSanger and whole genome sequencing in clinical practice. In late2009, the Medical College <strong>of</strong> Wisconsin initiated the infrastructureto streamline the delivery <strong>of</strong> current and emerging DNA technologiesinto state-<strong>of</strong>-the-art molecular diagnostics. The online publication <strong>of</strong>our initial case in Genetics <strong>of</strong> Medicine in late 2010 further intensifiedour efforts in this endeavor. However, being relatively new to thefield <strong>of</strong> NextGen sequencing, we began with the addition <strong>of</strong> Sangerdiagnostic sequencing to our already successful research core, whichat that point had been in operation for almost ten years. This was agreat undertaking, as typically, independent research laboratoriesperforming cutting-edge science lack the financial resources andbreadth <strong>of</strong> experience to launch their custom product or applicationto the diagnostic industry. An independent research laboratory is ableto resolve these shortages by partnering with a core laboratory staffedwith diagnostic expertise. Due to our lack <strong>of</strong> diagnostic experience, wequickly aligned the research core to a consortium <strong>of</strong> individuals withclinical experience to allow us to benefit from established diagnosticfacilities on campus. Difficulties faced at the onset <strong>of</strong> diagnostic startupwere many, including large issues such as accreditation program (CAPvs. CLIA), SOP generation and validation, competency and pr<strong>of</strong>iciencytesting, and reimbursement, as well as smaller problems like semiannualpipette calibration, temperature monitoring, and inventorycontrol. The purpose <strong>of</strong> this talk is to give insight into efficient ways toresolve these problems, both large and small, and transform a decadeor more <strong>of</strong> research expertise into a viable diagnostic laboratory.(W1-2) Chromosomal Microarray Analysis in theClinicL.D. WhiteMicroarray Core, Baylor College <strong>of</strong> Medicine, Houston, TX,United StatesCurrent Array Comparative Genomic Hybridization (aCGH) orChromosomal Microarray Analysis (CMA) performed at BaylorCollege <strong>of</strong> Medicine utilizes the latest microarray technology to detectunbalanced chromosome abnormalities associated with over 210 clinicaldisorders with exon by exon coverage <strong>of</strong> over 1,700 genes. We haveperformed approximately 35,000 postnatal CMA tests. Additionally,inclusive <strong>of</strong> our reported experience with 300 prenatal cases (PMID19012303), we now have CMA results on approximately 800 clinicalprenatal samples. This talk will cover the utility CMA for chromosomalabnormality detection in the clinical lab as well as development anddeployment <strong>of</strong> clinical tests from the core lab perspective.(W1-3) Whole Genome Sequencing in the ClinicalLaboratoryT. Hambuch, M. Laurent, B. Sickler, A. Liao, P. Cotter,S. Jain, Y. Lyan, J. Bernd, J.O. Daniel, P. Poggio, M. Ross,D. BentleyIllumina Clinical Services Laboratory, San Diego, CA, UnitedStatesThe advent <strong>of</strong> routine whole genome sequencing creates an opportunityto provide an accurate, comprehensive and cost-effective catalogue <strong>of</strong>germline variation for an individual. The process <strong>of</strong> sequencing anddelivering genomes for individual use must be driven by clinical andeducational opportunities balanced by addressing ethical concerns.Using guidelines issued from pr<strong>of</strong>essional and accrediting agenciesas well as an independent ethics board, we developed and launchedindividual genome sequencing (IGS) as a physician-led service. Aphysician orders the sequence and obtains informed consent from theindividual; the sample is sequenced within a CLIA certified laboratoryand after a series <strong>of</strong> quality checks the sequence is returned to thephysician for communication back to the individual. The sequencingplatform and process were validated for accuracy and precision, andaccredited following review by a College <strong>of</strong> American Pathologist(CAP) inspection team. Each individual genome is sequenced at >30fold coverage using paired-end reads <strong>of</strong> 100 base pairs. Resultingsequence information is provided for >93% <strong>of</strong> the NCBI36 genome, theremainder being mostly recently duplicated repeats where ambiguousread alignment is not permitted in our ELAND analysis. On average, wedetect over 3 million SNPs most <strong>of</strong> which are previously documentedin dbSNP129. The overall accuracy <strong>of</strong> our base calling is measured as>99.99% and the accuracy for SNP calling is >99.7% based on multiplemethodlogical assessments. The aim <strong>of</strong> the Illumina Clinical ServicesLaboratory is to make Individual Genome Sequencing accurate,accessible and clinically relevant for physicians and patients through afully accredited process. Here we have established baseline processes,tools, and policies to maximize the benefit to patients and minimizepotential misuse. Additionally, our ongoing efforts to develop clinicallyrelevant interpretation tools for physicians are described in a separateabstract (see M. Ross). Individual Genome Sequencing has the capacityto replace current genetic testing with a near-complete description <strong>of</strong>the sequence <strong>of</strong> an individual. Considering this potential, it is essentialto engage policy makers and ethicists so that appropriate policiesare developed around information access and use <strong>of</strong> whole genomeinformation.44 • <strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine


(W2) Spectral and Sequence DatabaseSearching in ProteomicsL. MartensGhent University, Ghent, BelgiumThis session will present an overview <strong>of</strong> more advanced methods tomatch acquired MS/MS spectra to peptides, including strategies todetect (unexpected) protein modifications, and the use <strong>of</strong> spectrallibraries for peptide identification by spectrum-to-spectrum matching.Introduced by some <strong>of</strong> the world’s foremost experts in these proteomicsinformatics challenges, this session is meant to provide a pragmaticintroduction to the topics for interested researchers, thus making thesemethods directly adoptable in the lab.(W2-1) ETD Performance and Complementarityto Other Fragmentation Methods for ProteomicAnalysisR. ChalkleyUniversity <strong>of</strong> California San Francisco, San Francisco, CA,United StatesRadical-driven fragmentation approaches present an alternative to thewell-established collisional cleavage approaches that have dominatedproteomic research up until now. With the recent availability <strong>of</strong>electron transfer dissociation (ETD) in commercial quadrupole iontrap and hybrid instruments, this technology is now accessible to manyresearchers. It has been described as a complementary approachto CID, and decision tree approaches have been employed where achoice between CID or ETD is made depending on the precursor m/zand charge. In this presentation I will discuss the performance <strong>of</strong> ETD forpeptide identification, drawing heavily on data acquired as part <strong>of</strong> the<strong>2011</strong> iPRG study ‘Identification <strong>of</strong> Electron Transfer Dissocation (ETD)Mass Spectra’. Results will be compared to analyses <strong>of</strong> the same sampleusing CID and HCD, decision tree approaches and the differencein measuring data in the ion trap versus in the orbitrap detector. Acomparison <strong>of</strong> search engine performance will be presented andmethods for improving database search engine analysis <strong>of</strong> ETD datawill also be discussed.(W2-2) Discovery, Identification and Localization <strong>of</strong>Post-Translational ModificationsN. BandeiraCenter for Computational Mass Spectrometry, Department<strong>of</strong> Computer Science and Engineering Skaggs School <strong>of</strong>Pharmacy and Pharmaceutical Sciences University <strong>of</strong>California, San Diego, CA, United StatesMass spectrometry based analysis <strong>of</strong> post-translational modificationscommonly report thousands <strong>of</strong> modified-peptide identificationsaccompanied by both precisely and ambiguously localizedmodification sites. Since these identifications <strong>of</strong>ten motivate extensivefollow up studies, the confident identification <strong>of</strong> the peptide andaccurate localization <strong>of</strong> the modification site(s) remains one <strong>of</strong> themajor challenges in computational proteomics. As revealed by the2010 iPRG study on identification <strong>of</strong> phosphopeptides and localization<strong>of</strong> phosphorylation sites, participants only attempted to call themodification sites for less than 2 out <strong>of</strong> every 3 identified spectraand actually disagreed on over 20% <strong>of</strong> all cases where at least twoparticipants called a modification site. In this talk we will cover currentand novel methods for identification <strong>of</strong> post-translationally modifiedpeptides and automated determination <strong>of</strong> site localization confidencescores and false discovery rates.(W2-3) Building and Using MS/MS Spectral Librariesfor Peptide Identifications in ProteomicsH. LamThe Hong Kong University <strong>of</strong> Science and Technology, ClearWater Bay, Hong Kong, ChinaSpectral library searching is an emerging approach in peptideidentifications from tandem mass spectra, a critical step in proteomicdata analysis. In this approach, a spectral library is first meticulouslycompiled from a large collection <strong>of</strong> previously observed and identifiedpeptide MS/MS spectra. An unknown spectrum is then identified bycomparing it to all the candidates in the spectral library for the mostsimilar match. Thanks to the reduction <strong>of</strong> search space to only thepreviously discovered peptides, and the use <strong>of</strong> real, experimentallyobserved reference spectra for more precise spectral matching, thisapproach is considerably faster and more sensitive than the popularalternative <strong>of</strong> sequence searching. This talk will explain the basicprinciples <strong>of</strong> spectral library building and searching, describe itsadvantages and limitations, and provide a starting point for researchersinterested in adopting this new approach in their data analysis. It willalso discuss the future outlook on the evolution and utility <strong>of</strong> spectrallibraries in the field <strong>of</strong> proteomics.(W3) Quantifying Protein Turnover by In VivoMetabolic Labeling(W3-1) Stable Isotope Tracers Applied to MeasuringRates <strong>of</strong> Protein Synthesis and Breakdown in Muscle:Principles and ApplicationsR.R. WolfeUniversity <strong>of</strong> Arkansas for Medical Sciences, Little Rock, AR,United StatesMuscle is in a constant state <strong>of</strong> turnover, meaning that it is continuouslysynthesized and broken down. The balance between the rates <strong>of</strong>synthesis and breakdown determines if an individual is gaining orloosing muscle mass. It is therefore <strong>of</strong> interest in a variety <strong>of</strong> physiologicalcircumstances to quantify the rates <strong>of</strong> muscle protein synthesis andbreakdown. Tracer methodology using both radioactive and stableisotopes has been used in a wide variety <strong>of</strong> kinetic studies, includingmeasurement <strong>of</strong> synthetic and breakdown rates <strong>of</strong> various compounds.Stable isotopes are particularly suited for the study <strong>of</strong> muscle proteinmetabolism, as multiple amino acid tracers can be used simultaneously,and multiple labels can be used with any individual amino acid,including labeling the nitrogen with 15N. Two basic approaches canbe used to measure muscle protein synthesis. The direct incorporation<strong>of</strong> a labeled amino acid is the most conventional. This techniqueinvolves infusion or injection <strong>of</strong> tracer and measurement <strong>of</strong> subsequentincorporation into muscle protein over time. The measurement <strong>of</strong> theprecursor enrichment, which is usually taken to be the free intracellularpool <strong>of</strong> the tracer amino acid, is necessary to calculate the actual rate<strong>of</strong> synthesis. Alternatively, the rate <strong>of</strong> muscle protein synthesis can bederived from the rate <strong>of</strong> uptake <strong>of</strong> an amino acid from blood. In thisWorkshop SessionAbstracts<strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine • 45


Workshop SessionAbstractscase incorporation <strong>of</strong> the amino acid into protein must be its onlymetabolic fate within the muscle. The basic principle <strong>of</strong> measuringmuscle protein breakdown is to determine the rate at which intracellularamino acid tracer is diluted by the appearance <strong>of</strong> unlabeled amino acidthat is not coming from the plasma. There are different approachesto accomplishing this measurement, and the choice <strong>of</strong> the optimaldepends <strong>of</strong> the method used to measure protein synthesis. Ideally,methods to measure synthesis and breakdown are compatible (i.e., thesame units) to enable calculation <strong>of</strong> the balance between synthesis andbreakdown to determine if there is a net gain or loss in muscle protein.(W3-2) In Vivo Stable Isotope Labeling forQuantifying Amyloid-beta Kinetics in Alzheimer’sDisease: Is it All in our Head?K.E. YarasheskiBiomedical Mass Spectrometry Research Laboratory,Washington University School <strong>of</strong> Medicine, St. Louis, MO,United StatesMass spectrometry has revolutionized the manner in which we identify,characterize, and quantify proteins. In combination with in vivo stableisotope labeling strategies, mass spectrometry-based analyses canprovide valuable information about human amino acid and proteinkinetics, protein production (synthesis), and clearance (proteolysis)rates. Dysregulated or imbalanced protein synthesis and degradationrates is the basis for many clinical disorders. These protein kineticrates can be quantified in vivo and serve to identify potential targetsfor novel drug therapies. Our group uses an intravenous infusion <strong>of</strong>13C6-Leu, cerebral spinal fluid (CSF) sampling, affinity isolation <strong>of</strong>relevant proteins, and tandem mass spectrometry to quantify 13C6-Leu incorporation and removal rates from CSF amyloid-beta andapolipoproteins in Alzheimer’s disease patients. This approach hasidentified slower amyloid-beta clearance rates as a primary lesion thatmay explain an accumulation <strong>of</strong> amyloid plaques in Alzheimer’s disease.(W4) Insights for Expression <strong>of</strong> RecombinantProteins for Drug Target Validation(W4-2) Strategies for Optimized High-ThroughputCloning, Expression and Purification <strong>of</strong> RecombinantProteins in E. coliR. PageBrown University, Department <strong>of</strong> Molecular Biology, CellBiology and Biochemistry, Center for Genetics, Genomicsand Proteomics, Providence, RI, United StatesThe arrival <strong>of</strong> structural genomics, in addition to large-scale effortsinitiated in pharmaceutical companies, have resulted in the development<strong>of</strong> numerous new methods and strategies to minimize the time requiredto optimize the cloning, expression and purification <strong>of</strong> novel proteindrug targets. Here, I will present both best practices and unusual (‘lastditch’) methods that are used to successfully express and purify bothprokaryotic and eukaryotic proteins in E. coli. Recent developments inprotein expression, including co-expression with protein partners andbacterial chaperones will be presented. In addition, a comparison <strong>of</strong>the best solubility enhancing tags, and methods for subsequent tagremoval, will be presented. Finally, I will also describe two case studieswhich required highly tailored expression protocols for the production<strong>of</strong> a bacterial toxin and eukaryotic phosphatase. In summary, the focus<strong>of</strong> this lecture will be to provide practical information that researchersactively involved in protein purification can readily implement intotheir own workflows. This work was supported in part by a medicalresearch grant from the American Cancer Society (RSG-08-067-01-LIB)and an NSF-CAREER award (MCB 0952550).(W5) Proteomics Tips and Tricks: FromDiscovery to Protein-Protein Interactions(W5-1) Proteomics Tips and Tricks: From Discoveryto Protein-Protein InteractionsM. CiliaUnited States Department <strong>of</strong> Agriculture/AgriculturalResearch Service, Robert W. Holley Center for Agricultureand Health, Cornell University, Ithaca, NY, United StatesProteins are the functional constituents <strong>of</strong> cells. The diverse physicalproperties <strong>of</strong> proteins enable them to be multifunctional and also posechallenges to analyzing their abundance, subcellular localization, andpotential interacting partners using proteomics. Some samples areespecially recalcitrant and pose additional challenges in proteomicanalysis due to the unanticipated modification <strong>of</strong> proteins. Thesechallenges extend from initial protein extraction to analysis usingmass spectrometry. Broadly, the focus <strong>of</strong> workshop will be to discussthe benefits and limitations <strong>of</strong> using proteomics to study proteinidentification, protein quantification, protein structure, and proteinproteininteractions. Examples will be drawn from different biologicaldisciplines, including cell biology, host-pathogen biology, anddevelopmental biology to demonstrate how scientists are solving theproblems posed by their discovery methods and biological systems.The workshop will include presentations from each panelist followedby an open discussion and question and answer session.(W5-2) Protein Interactions and Topologies in CellsJ. BruceUniversity <strong>of</strong> Washington, Department <strong>of</strong> Genome Sciences,Seattle, WA, United StatesLife on earth has evolved to utilize proteins as functional molecules dueto the wide diversity <strong>of</strong> structures and physical properties this class <strong>of</strong>molecules can exhibit. However, the divergent properties that criticallysupport life also pose fundamental challenges in all efforts to measurethe proteome. As a result, most proteomics experiments only samplea small subset <strong>of</strong> expressed molecules and typically appear biasedtowards cytoplasmic proteins. Furthermore, detection <strong>of</strong> proteinproteininteractions is even more challenging. For example, most largescalemethods such as the yeast two-hybrid, tandem affinity purification,and co-IP methods are predicated on the production/maintenance<strong>of</strong> native protein structures and co-localization <strong>of</strong> native interactingpartners. These two requirements can result in failure to identify manybone fide interactions and in false discovery because many interactions46 • <strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine


may not survive cell lysis steps and non-relevant interactions can formin cell lysates during sample preparation. Significant progress onmany <strong>of</strong> these issues has been achieved in recent years, including theuse <strong>of</strong> cryogenic cell lysis techniques. Alternatively, covalent linkage<strong>of</strong> interacting proteins within cells as accomplished with chemicalcross-linking has long held potential for protein interaction studies.If successful, chemical cross-linking approaches mitigate the needto maintain native interactions and structures during subsequentproteome sample preparation and may <strong>of</strong>fer unique insight on proteininteractions and structures present within cells. However, the analysis <strong>of</strong>cross-linked peptides presents a different set <strong>of</strong> challenges related todynamic range and detection specificity. This presentation will highlightadvancements in technology, informatics and sample preparation stepswe have pursued to enable cross-linked peptide measurements from invivo cross-linking experiments and will illustrate the unique informationthat can be derived from this approach.(W5-3) The Virus-Host Interface: Exploring DynamicProtein Interactions via Targeted ProteomicsI. CristeaPrinceton University, Department <strong>of</strong> Molecular Biology,Princeton, NJ, United StatesDynamic protein interactions carry out the majority <strong>of</strong> the processeswithin a cell, including cellular responses to environmental stimuliand pathogens. Isolation and characterization <strong>of</strong> protein complexescan provide invaluable insights into their biological functions. Thedevelopment <strong>of</strong> approaches that can access stable and transientinteractions is invaluable for numerous fields <strong>of</strong> study, including that<strong>of</strong> temporal and spatial virus-host protein interactions. Viruses haveco-evolved with their hosts, developing remarkable mechanisms forsubverting cellular processes for their own benefit. The study <strong>of</strong> virushostinteractions has therefore emerged as a driving force in infectiousdisease research. Despite these efforts, the protein interactomeremains in large part uncharted, and our knowledge <strong>of</strong> mechanismscontrolling the outcome <strong>of</strong> an infection is limited. Modern proteomicstechniques are currently emerging as powerful tools, bringing a newperspective to the field <strong>of</strong> virology. This presentation will describe theintegration <strong>of</strong> targeted proteomics with genetic, molecular biology,and bioinformatics techniques for studying dynamic virus-host proteinassociations. Strategies for isolating protein complexes, quantifyinginfection-triggered changes in interactions, and assessing interactionspecificity will be presented. In studies <strong>of</strong> human cytomegalovirus(HCMV) infection, we discovered parallel processes occurring at distinctcellular sites during the assembly <strong>of</strong> infectious virions. Additionally, wehave observed that certain viral proteins recruit chromatin-remodelingenzymes, such as histone deacetylases, indicating a possible mean <strong>of</strong>controlling virus or host gene expression. In addition to revealing theirfunctional roles during infection, our studies provided insights intothe regulation <strong>of</strong> these enzymes outside the context <strong>of</strong> infection. Acombinatorial proteomics approach, incorporating CID, HCD and ETDpeptide fragmentation using a nLC LTQ Orbitrap Velos-ETD, identified17 in vivo phosphorylation sites on HDAC5. Functional phosphomutantscreening and live cell imaging allowed the characterization <strong>of</strong> novel siteswithin functional domains, and identified a previously unrecognizedregulatory point <strong>of</strong> its nuclear import.(W5-4) Towards the Development <strong>of</strong> ProteomicsWorkflows for the Analysis <strong>of</strong> Samples Derived fromRefractory Plant TissuesT.W. ThannhauserUnited States Department <strong>of</strong> Agriculture/AgriculturalResearch Service, Ithaca, NY, United StatesCarrying out proteomic analyses in plant tissues involves dealing witha number <strong>of</strong> specialized challenges that can make protein extractionand quantification significantly more difficult than in other organisms.In addition to having relatively low protein concentrations, plant tissuesare <strong>of</strong>ten rich in proteases, protease inhibitors and other materials thatimpede protein analysis. These compounds include lipids, tannins,polysaccharides, and a large variety <strong>of</strong> secondary metabolites. Theextent <strong>of</strong> the problems encountered is dependent on tissue typestudied. Two major research thrusts in our lab involve detaileddevelopmental and time-course studies on staple crops and fruits. Anunderstanding <strong>of</strong> the proteins involved in the development <strong>of</strong> fruits,seeds, tubers and other plant organs will enhance our ability to controlthe agronomically-important traits <strong>of</strong> these crops including stabilityin storage, disease resistance, and vitamin and mineral content. Tworecent studies that have posed unique challenges for proteome analysisinclude 1) comparing protein expression in red ripe tomato fruit to thatin mature green tomato fruit and 2) studying the effects <strong>of</strong> storage onpotato tubers. This talk will focus on the problems encountered in theserecalcitrant tissue types and our efforts to provide plant scientists witha broader array <strong>of</strong> extraction and chemical modification protocols thanis represented by those that constitute the existing paradigm. Effortsto move away from narrowly defined prescriptive methods and movetowards results-based methods will also be discussed.(W6) Current State and Future <strong>of</strong> CapillaryElectrophoresis and Sanger SequencingJ. Kieleczawa 1 , D. Adam 2 , P. Schweitzer 3 , E. Vennemeyer 4 ,M. Zianni 51Pfizer, Inc., Cambridge, MA, United States; 2 NAPS Unit,Michael Smith Laboratories, University <strong>of</strong> British Columbia,Vancouver, BC, Canada; 3 DNA Sequencing and GenotypingLab, Cornell University, Ithaca, NY, United States;4LifeTechnologies, Carlsbad, CA, United States; 5 Plant-Microbe Genomics Facility, Ohio State University, Columbus,OH, United StatesCapillary Electrophoresis remains a widely used, <strong>of</strong>ten irreplaceable,sequencing technology continuing to experience increasing demands.We believe that the speed, simplicity and high quality <strong>of</strong> reads for lowvolumesequencing needs will maintain the viability <strong>of</strong> this technologyfor many years to come. Although the main thrust <strong>of</strong> developmentin the sequencing space is now in NGS area, there are still stepswhich can be improved or adjusted as other methods mature. In thissession we propose to cover: The effect <strong>of</strong> long-term storage <strong>of</strong> DNAunder different conditions (temp/buffers) on sequencing, spectralcharacteristics, integrity and transformation efficiency; A Description<strong>of</strong> the Apollo system; and the Future <strong>of</strong> Capillary Electrophoresis andSanger sequencing. In addition to the featured talks, there will be aquestion & answer component in which participants are encouraged tobring up any issues relevant to this topic.Workshop SessionAbstracts<strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine • 47


Workshop SessionAbstracts(W7) Cellular 3D Imaging(W7-1) 3D Cellular Imaging: Beyond the SimpleImaging ParadigmR. ColeWadsworth Center, New York State Department <strong>of</strong> Health,Albany, NY, United StatesThe modern microscope has advanced so far beyond what Leeuwenhoekinvented, it’s doubtful he would even recognize it as a microscope.Modern microscopes, or more acutely, imaging workstations, arean integrated compilation <strong>of</strong> optoelectronic components. It is nowpossible to produce much more than simple images using thesesystems. Quantitation <strong>of</strong> various metrics are now possible with theseadvanced instruments. With the advent <strong>of</strong> fluorescently-taggedproteins/organelles, it is possible to actually quantify sub-cellularamounts <strong>of</strong> these transgenically produced or exogenously labeledmacromolecules/complexes. Spectral un-mixing <strong>of</strong> overlapping signalsfrom within a single image (fluorescent and absorbent) has blurreddistinction between imaging and spectroscopy. It is now possible toget chemical data from techniques such as unmixing in addition tomore conventional modes such as polarized light microscopy. Theability to determine if two macromolecules are precisely co-localized(< 100 Å apart) is now routinely accomplished in both living and fixedpreparations with Fluorescence Resonance Energy Transfer (FRET). Thelatest technological advances has been achieving resolution beyondAbbe’s predicted limits. This has been accomplished both throughhardware and s<strong>of</strong>tware improvements, such as Stimulated EmissionDepletion (STED), and Structured Illumination Microscopy (SIM) toname a couple.(W7-2) Basics <strong>of</strong> Colocalization AnalysesJ. LacosteCell Imaging and Analysis Network, Department <strong>of</strong> Biology,McGill University, Montreal, QU, CanadaFor many fluorescence microscopy-based projects, colocalizationanalyses are <strong>of</strong>ten the main objectives <strong>of</strong> the study, and detecting“yellow” is considered a pro<strong>of</strong> <strong>of</strong> colocalization. However, the “yellow”statement is essentially a qualitative one which opens the door tomore quantitative approaches. This presentation will provide aquick overview <strong>of</strong> colocalization analysis methods. First the requiredpreliminary steps and words <strong>of</strong> caution will be discussed. Secondly,intensity correlation coefficient -based methods (both the traditionaland more modern ones) will be presented, and object-based analysismethods will next be introduced. Lastly, it is important to rememberthat colocalization studies are dependent on quantitative microscopyand as such requires careful considerations.(W8) Successful Production <strong>of</strong> FunctionalProteins(W8-1) Successful Protein ProductionJ. CulpPfizer, Inc., Groton, CT, United StatesSuccessful production <strong>of</strong> functional proteins is more than animmunoreactive band on a Western blot. Availability <strong>of</strong> multipleexpression vectors make accessible a variety <strong>of</strong> expression systems andparallel expression approaches can speed results and increase chance<strong>of</strong> success. The next hurdle is isolation <strong>of</strong> the protein target in sufficientamounts and with sufficient purity to support subsequent experimentalwork. Occasionally, protein refolding is the only method available toachieve the desired protein. Finally, sufficient characterization <strong>of</strong>the purified protein is required to verify that the protein is in thebiologically relevant state. Often, the protein is heterogeneous withpost-translational modifications and may be aggregated or in multipleoligomeric complexes.(W8-2) Overcoming Problems in Protein Expressionand PurificationB. GilletteSAIC-Frederick, Inc., National Cancer Institute at Frederick,Frederick, MD, United StatesWe use a parallel, micro-scale protein purification approach to screenpurification outcomes for multiple expression constructs from multipleexpression systems. Method development and optimization for leadcandidates are then performed on the same platform. Scale-up fromthe micro-scale format (10-160 microliter column volumes available)to bench-top scale (5-100 milliliter column volume) has provenpredictive in terms <strong>of</strong> qualitative and quantitative results. The increasein throughput allows us to follow a “purify first” approach that, whilestill providing data on expression levels and solubility, also producesvaluable purification information within the same time-frame as atraditional solubility assessment approach.(W8-3) When Proteins Misbehave, Try Adding aLittle PressureR.M. PetrovichNational Institute <strong>of</strong> Environmental Health SciencesLaboratory <strong>of</strong> Structural Biology, Research Triangle Park,NC, United StatesThe most economical way to produce the mg quantities <strong>of</strong> purifiedprotein required by many studies still requires expression in E. coli.Unfortunately, between 50% and 70% the time, a given protein willnot express properly in bacteria. Of those that are correctly folded, apercentage <strong>of</strong> them will either lose activity during purification or coldstorage once purified. To overcome these obstacles we have begunusing high hydrostatic pressure refolding (Bar<strong>of</strong>old) which allows us torefold protein at high concentration (1 mg/ml or higher) without addingand removing chaotropic agents.48 • <strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine


(W9) Therapeutic Antibodies: Over-Hyped“Magic Bullet”, or Under-Explored Technology?D.L. Crimmins 1 , J.E. Harlan 2 , F. Weis-Garcia 31Washington University School <strong>of</strong> Medicine, Saint Louis, MO,United States; 2 Abbott Laboratories, Abbott Park, IL, UnitedStates, 3 Memorial Sloan-Kettering Cancer Center, New YorkCity, NY, United StatesBringing the vision that monoclonal antibodies (mAbs) could bemedicinal “magic bullets” into practice has been a long and challengingjourney which began in 1975 when Kohler and Milstein made it possibleto generate these homogenous, highly specific homing devices. In thelast 35 years, many hurdles have slowed the manifestation <strong>of</strong> this dream,including: target location; host species, specificity, and subclass <strong>of</strong> themAb; immunogenicity <strong>of</strong> murine mAbs; mAb pharmacokinetics (i.e.hepatic and renal clearance); and mAb form (i.e. natural, conjugatedto a toxin or radionuclide, humanized murine mAbs, scFvs). The workinvolved in overcoming these factors explains why few mAb’s have beenmade it to the clinic in more than 3 decades later. Nonetheless, over20 mAbs have been FDA-approved for human use since 1994. Sincethe total global therapeutic monoclonal antibody market increasedfrom $40 billion in 2009 to $45 billion in 2010, pursuit <strong>of</strong> these “magicbullets” is unlikely to wane anytime soon. This workshop is a roundtableformat what where we will collectively discuss what we have learned onthis long road <strong>of</strong> bringing mAbs to the clinic and try and look forwardto where we are going.(W10) Identification <strong>of</strong> Mechanism-BasedBiomarkers and Drug Targets Using PathwayAnalysis(W10-1) GeneXplain — Identification <strong>of</strong> CausalBiomarkers and Drug Targets in Personalized CancerPathwaysA. Kel 1 , F. Kolpakov 2 , V. Poroikov 3 , G. Selivanova 41geneXplain GmbH, Wolfenbuettel, Germany; 2 Institute<strong>of</strong> Systems Biology Ltd., Novosibirsk, Russia; 3 Institute <strong>of</strong>Biomedical Chemistry <strong>of</strong> Russian Academy <strong>of</strong> MedicalSciences, Moscow, Russia; 4 Microbiology and Tumor BiologyCenter (MTC), Karolinska Institutet, Stockholm, SwedenWe have developed a geneXplain TM platform (www.genexplain.com)for causal interpretation <strong>of</strong> data coming from microarray, proteomics,miRNA and ChIP-chip/seq experiments. GeneXplain TM applies aunique upstream analysis approach based on implementation <strong>of</strong>machine learning and graph topological analysis algorithms in orderto identify causality keynodes in the network <strong>of</strong> gene regulation andsignal transduction and combines it with full genome sequence analysisand chemoinformatics methods for drug discovery. The power <strong>of</strong> thisapproach is that we are trying to identify causal biomarkers — thosewhich are more than just correlating with disease or treatment outcomebut which are parts <strong>of</strong> the disease mechanism, which may differ inpatient cohorts. Such personalized networks are analyzed in order t<strong>of</strong>ind key nodes — most important nodes triggering the disease. Thesekeynodes and genes directly influenced by them are considered aspromising biomarkers which can discriminate patients into cohortsfrom the disease mechanism point <strong>of</strong> view. In the current study, weanalyzed a large scale gene expression and ChIP-seq data from astudy <strong>of</strong> a breast cancer samples treated with antineoplastic agentsincluding the novel drug compounds — RITA and Nutlin, targeting p53and Mdm2. We analyzed promoters <strong>of</strong> downregulated pro-survivalgenes and identified combinations <strong>of</strong> transcription factors involvedin their regulation. Topological modeling <strong>of</strong> the signal transductionnetwork upstream <strong>of</strong> these transcription factors revealed key-nodes— potent master-regulators <strong>of</strong> the cell survival program that preventefficient apoptosis <strong>of</strong> cancer cells. We considered these key-nodeproteins (e.g. PI3K subunits) as causal biomarkers as well as prospectivetargets for novel anticancer drug combinations. We applied acheminformatics computer tool PASS to these targets and identifiedtwo novel prospective antineoplastic chemical compounds which wereexperimentally validated in a cellular assay confirming their synergisticpotential in highly selective triggering <strong>of</strong> apoptosis <strong>of</strong> cancer cells.(W10-2) Pathway Analysis in Expression ProteomicsR.A. ZubarevChemistry I Division, Department <strong>of</strong> Medical Biochemistryand Biophysics, Karolinska Institutet, Stockholm, SwedenProteomics studies have revealed unexpected plasticity and dynamicnature <strong>of</strong> the human proteome. The paradigm that the time evolution<strong>of</strong> a biological system can be described by abundance variation <strong>of</strong>relatively few “regulated” proteins has been shuttered, being replacedby the growing understanding that the whole proteome is regulated,and no protein remains unaffected when the system undergoestransition from one state to another. This finding underlines theimportance <strong>of</strong> systems biology analysis <strong>of</strong> expression proteomics data.Systems biology shifts the analytical focus from thousands <strong>of</strong> proteins tohundreds <strong>of</strong> signaling pathways, thus reducing the number <strong>of</strong> entities tobe analyzed. Application <strong>of</strong> these methods required the development<strong>of</strong> novel systems biology tools, such as the pathway search engine (PSE[1-3]). These tools can only be effective when they are quantitative, i.e.predict not only the activated pathway, but also the relative degree <strong>of</strong> itsactivation. Introducing the quantitative aspect in systems biology is one<strong>of</strong> the greatest challenges this field is facing today, since the final goal<strong>of</strong> pathway analysis, which is the creation <strong>of</strong> a quantitative predictingmodel <strong>of</strong> the biological process under investigation. 1. Zubarev, R. A.;Nielsen, M. L.; Savitski, M. M.; Kel-Margoulis, O.; Wingender, E.; Kel,A. Identification <strong>of</strong> dominant signaling pathways from proteomicsexpression data, J. Proteomics, 2008, 1, 89-96. 2. Stahl, S.; Fung, Y.M.E.;Adams, C. M.; Lengqvist, J.; Mörk, B.; Stenerlöw, B.; Lewensohn, R.;Lehtiö, J.; Zubarev, R. A.; Viktorsson, K. Proteomics and Pathway AnalysisIdentifies JNK-signaling as Critical for High-LET Radiation-inducedApoptosis in Non-Small Lung Cancer Cells, Mol. Cell Proteomics, 2009,8, 1117-1129. 3. Marin-Vicente, C.; Zubarev, R. A. Search engine forproteomics, Fact or Fiction? G.I.T. Lab J, 2009, 11-12, 10-11.(W10-3) Metabolic Biomarkers, MetabolicNetworks, and Pathway AnalysisV. TolstikovUniversity <strong>of</strong> California Davis Genome Center, Davis, CA,United StatesWe apply well established MS-based Metabolomics platform towardsthe detection, characterization and identification <strong>of</strong> small moleculebiomarkers. Data acquisition is performed with the assistance <strong>of</strong> GC/TOF/MS line and LC/MS line. Sophisticated data mining approach usingdifferent algorithms deals with electron impact (EI) generated data as wellas data generated with the unit resolution, high resolution and ultrahighWorkshop SessionAbstracts<strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine • 49


Workshop SessionAbstractsresolution hybrid instruments having API sources. These competitiveapproaches deliver exciting results on selection <strong>of</strong> the predictors’ paneland potential metabolic biomarkers discovery. We here report onefficiency <strong>of</strong> Metabolic Biomarkers Discovery Project as a platform forcases study: Kidney diseases (RCC and PKD); Pancreatic Cancer (PDAC);Human Embryonic Stem Cells functional characterization. However,Metabolic Networks simulation and Metabolic Pathway Analysis,which could be essential tools for Metabolic Biomarkers validation andconnections with underlying biochemical processes are still difficultand approached not systematically, but rather case wise. Challengesand developments in this field will be further presented and discussed.(W11) The Business <strong>of</strong> Running a Core FacilityN.P. Ambulos, Jr. 1 , S.A. Bobin 21University <strong>of</strong> Maryland School <strong>of</strong> Medicine, Baltimore, MD,United States; 2 Dartmouth School <strong>of</strong> Medicine, Hanover, NH,United StatesThe success and sustainability <strong>of</strong> a Core Facility has been dependenton factors which include (1) the ability to maintain state-<strong>of</strong>-thearttechnology, (2) recruiting and retaining talented technical staff,(3) a strong financial management plan, and (4) strong Institutionalsupport from both end users and the Administration. Historically,most core facilities enjoyed generous subsidies from their InstitutionalAdministration, which have helped defray costs for end users, purchasenew instrumentation, or to hire new staff. In light <strong>of</strong> the current economy,it is likely that many core facilities are now seeing these subsidiesdrastically cut, or even eliminated. In essence, core facilities are facedwith operating as a self-supporting ‘business’, and at the same time,ensure compliance with federal guidelines. This workshop will presentideas, both conventional and creative, that might provide core directorswith the tools necessary to continue to sustain a successful core facility.This will set the stage for an open discussion with the audience toencourage a free-flow <strong>of</strong> additional ideas that could benefit the futuresuccess <strong>of</strong> our cores.(W12) Microarrays: The Reports <strong>of</strong> My DeathHave Been Greatly Exaggerated(W12-1) Chip or Seq: Helping Clients ChooseD. BaldwinUniversity <strong>of</strong> Pennsylvania, Penn Molecular Pr<strong>of</strong>iling Facility,Philadelphia, PA, United StatesThe Penn Molecular Pr<strong>of</strong>iling Facility provides services using microarrayand deep sequencing platforms for a variety <strong>of</strong> applications. Drawingfrom experiences helping prospective clients to choose one approachor the other, and sometimes both, this portion <strong>of</strong> the workshop willaddress some <strong>of</strong> the factors to consider when thinking about a newproject. The budgeting process will be discussed, with particular focuson the use <strong>of</strong> interactive cost calculators.(W12-2) Microarray Analysis <strong>of</strong> FluorescenceActivated Cell Sorter-Derived Cells: CreatingHarmony Between TechnologiesS. TigheUniversity <strong>of</strong> Vermont, Advanced Genome Technology Core,Burlington, VT, United StatesAlthough microarray technology is well-established in both theresearch and clinical fields, it continues to evolve into new areas thatrequire new methods for the successful isolations <strong>of</strong> nucleic acid fromnon-traditional sources. Because RNA specifically is a labile molecule,special procedures and considerations must be implemented to avoiddegradation from methods such as fluorescence activated cell sorting(FACS) and laser capture microdissection (LCM) to name a few. Thispresentation will discuss specific methodologies to maximize the success<strong>of</strong> nucleic acid recovery from these approaches including instrumentpreparation, extraction methods, and the use <strong>of</strong> special reagents todeal with problematic samples.(W12-3) Microarray Futures: Don’t DecommissionYour Scanners Just YetS.D. CrosbyDepartment <strong>of</strong> Genetics, Washington University School <strong>of</strong>Medicine, St. Louis, MO, United StatesThe first attempt to wind down the Washington University MicroarrayFacility was made by Genome Sequencing Center in 2008. It wasclear to most <strong>of</strong> us involved in the attempt that, with the advent <strong>of</strong>NGS, microarrays were headed the way <strong>of</strong> differential display PCR(remember that!?). The attempt failed, and 2010 was the busiest yearyet for our microarray facility. Sequence remains a bit too expensiveand complex for many to make the leap. In addition, because <strong>of</strong> therarity <strong>of</strong> variants and the modest number <strong>of</strong> bases required to identifya locus, most sequence data is superfluous. Beyond that, as the actualcost <strong>of</strong> sequence continues to fall, so do the price <strong>of</strong> arrays, while thedensity <strong>of</strong> elements <strong>of</strong> the latter rises. It seems that until the advent <strong>of</strong>reagentless sequencing, it is unlikely that the actual cost <strong>of</strong> sequencingwill be less than the cost <strong>of</strong> an array that covers the same loci. Whilesequencing technology will remain an important tool for discovery,over time most exons and biologically relevant variants will be capturedin toto on low cost arrays. Hospital or contract labs will (indeed arealready) use small, cheap arrays that capture disease relevant genes andvariants.50 • <strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine


(W13) Institutional Core ManagementS. Meyn 1 , P. Turpen 2 , G.K. Farber 3 , S. Mische 4 ,P. Alexander 5 , J. Auger 61Vanderbilt University Medical Center, Nashville, TN, UnitedStates; 2 University <strong>of</strong> Nebraska Medical Center, Omaha,NE, United States; 3 National Center for Research Resources,Bethesda, MD, United States; 4 New York University LangoneMedical Center, New York, NY, United States; 5 MorehouseSchool <strong>of</strong> Medicine, Atlanta, GA, United States; 6 University <strong>of</strong>California San Francisco, San Francisco, CA, United StatesThis workshop session will focus on issues related to InstitutionalCore Management, in response to the national conversation evolvingaround research core facility issues and management. The workshopwill be formatted as an experts’ panel; each participant currentlyplays an important role in supporting and developing research coreresources at an institutional level. Some <strong>of</strong> the topics to be discussedinclude: (1) Core Consolidation — one size fits all? (2) Bottom-up vs.top-down management, advantages and disadvantages <strong>of</strong> centrallymanaged cores. (3) Performance metrics and impacts on pr<strong>of</strong>essionaldevelopment, core infrastructure support and improved operations.(4) Impacts <strong>of</strong> NIH-NCRR programs on improving access to researchresources, including core facilities. We also plan to highlight the newCore Administrators Network Coordinating (CAN). In responseto an emerging trend to centralize the oversight <strong>of</strong> research corefacilities, <strong>ABRF</strong> has fostered development <strong>of</strong> this network and anew committee: the Core Administrators Network-CoordinatingCommittee (CAN-CC). The committee seeks input and participationfrom scientists, administrators and others with an interest in issuesrelated to the administration <strong>of</strong> research core facilities which, by thenature <strong>of</strong> their service role, must interface with multiple constituencieswithin a research enterprise. Today many institutions have establishedadministrative positions designed to assist core facilities withmanagement <strong>of</strong> economic, regulatory and performance issues. In orderto facilitate greater interaction between and among core scientistsand administrators, the mission <strong>of</strong> the CAN-CC is to contribute to thecommon interests <strong>of</strong> core administrators, and promote interactions withcore scientists in a collegial and productive manner. The specific goals<strong>of</strong> the Core Administrators Network Coordinating Committee (CAN-CC) are: to identify and reach out to our target community; provideopportunities for networking; and assess goals for program focus anddevelopment.(W14) Next Generation Sequencing S<strong>of</strong>twarefor Data Management, Analysis, andVisualization(W14-1) Tools for Next Generation Sequencing DataAnalysisK. BodiTufts University School <strong>of</strong> Medicine, Tufts University CoreFacility, Boston, MA, United StatesAs NGS technology continues to improve, the amount <strong>of</strong> data generatedper run grows exponentially. Unfortunately, the primary bottleneckin NGS studies is still bioinformatics analysis. Not all researchershave access to a bioinformatics core or dedicated bioinformatician.Additionally, much <strong>of</strong> the s<strong>of</strong>tware for NGS analyses is written to runin a Unix / Linux environment. Researchers unfamiliar with the Unixcommand line may be unable to use these tools, or face a steep learningcurve in trying to do so. Commercial packages exist, such as the CLCGenomics Workbench, DNANexus, and GenomeQuest. However,these commercial packages <strong>of</strong>ten incorporate proprietary algorithmsto perform data analysis and may be costly. Galaxy provides a solutionto this problem by incorporating popular open-source and communitylinux command line tools into an easy to use web-based environment.After sequence data has been uploaded and mapped, there are avariety <strong>of</strong> workflows for NGS analyses that use open-source tools. Thisincludes peak-calling analyses for ChIP-Seq (MACS, GeneTrack indexer,Peak predictor), RNA-Seq (Tophat, Cufflinks), and finding smallinsertions, deletions, and SNPs using SAMtools. Any researcher canapply a workflow to his NGS data and retrieve results, without having tointeract with a command line. Additionally, since Galaxy is cloud-based,expensive computing hardware for performing analyses is not needed.In this presentation we will provide an overview <strong>of</strong> two popular opensourceRNA-Seq analysis tools, Tophat and Cufflinks, and demonstratehow they can be used in Galaxy.(W14-2) GenomeView: Visualizing the Next-Generation <strong>of</strong> DataT. AbeelBroad Institute <strong>of</strong> MIT and Harvard, Cambridge, MA, UnitedStates, and VIB Department <strong>of</strong> Plant Systems Biology, GhentUniversity, Ghent, BelgiumDue to recent advances in sequencing technologies, billions <strong>of</strong> nucleotidesequences are now produced on a daily basis. A major challenge isto visualize these data, including both whole genome sequence andtranscriptome data, for further downstream analyses. Visualization is<strong>of</strong>ten overlooked and undervalued, but it is an extremely valuable toexplore your data on several levels. A first area where visualization shinesis at the early stages <strong>of</strong> data analysis to perform sanity checks on yourdata. Eye-balling your data in a visually pleasing way is the best way toget a good feel on what came out <strong>of</strong> your experiments. Once you havea good idea <strong>of</strong> what is in your data a good visual representation can beused to generate new hypotheses and to fine-tune analysis parameters.The appropriate image <strong>of</strong>ten makes the solution obvious and as such, itreally makes it easier to develop algorithms. The ability to interactivelyexplore gives you insights in large-scale data sets and definitelyaugments our ability to reason about complex data. To this end, wepresent GenomeView, a stand-alone sequence browser specificallydesigned to visualize and manipulate a multitude <strong>of</strong> genomics data.GenomeView enables users to dynamically browse high volumes <strong>of</strong>aligned short read data, with dynamic navigation and semantic zooming,from the whole genome level to the single nucleotide. At the same time,the tool enables visualization <strong>of</strong> whole genome alignments <strong>of</strong> dozens <strong>of</strong>genomes relative to a reference sequence. GenomeView is unique in itscapability to interactively handle huge data sets consisting <strong>of</strong> dozens<strong>of</strong> aligned genomes, thousands <strong>of</strong> annotation features and millions <strong>of</strong>mapped short reads both as viewer and editor. GenomeView is freelyavailable for academic use as an open source s<strong>of</strong>tware package athttp://genomeview.org.Workshop SessionAbstracts<strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine • 51


(W14-3) Galaxy Next Generation SequencingFunctionality from Sample Tracking to SNP CallingG. Von Kuster 1 , The Galaxy Team 1,21Center for Comparative Genomics and Bioinformatics,Pennsylvania State University, University Park, PA, UnitedStates; 2 Departments <strong>of</strong> Biology and Mathematics &Computer Science, Emory University, Atlanta, GA, UnitedStatesA new generation <strong>of</strong> DNA sequencing technologies has enabled a variety<strong>of</strong> novel genome-scale experimental techniques. What is perhapsmost unique about this recent data explosion is that it is distributed— relatively inexpensive instruments allow any lab or institution toproduce enormous amounts <strong>of</strong> data. Yet the infrastructure upstreamand downstream <strong>of</strong> sequencing instruments is largely undeveloped.In addition to the instrument cost labs, core facilities and sequencingservice providers are forced to earspend thousands on commercialLIMS systems and sequence analysis packages, which are in-turn basedon tools from the public domain. Galaxy provides a robust open-sourcealternative. It’s lightweight sample tracking system is aimed at helpingsmall labs and core facilities managing requests for sequencing runs. Itallows one to track the entire “life-cycle” <strong>of</strong> sequencing request fromthe initial sample to the resulting dataset. Once the run is completethe user can apply a variety <strong>of</strong> NGS tools including format converters,mappers, ChIP-seq and transcriptome utilities. Results <strong>of</strong> these analysescan be visualized, shared, and published. In this presentation we willdemonstrate sample tracking functionality from the moment <strong>of</strong> samplesubmission to the sequencing facility, through the sequencing run, untilthe sample becomes a dataset and can be analyzed with a variety <strong>of</strong>NGS tools.(W15) ETD Workshop Seminar Series: ManualInterpretation <strong>of</strong> Electron Transfer Dissociation(ETD) Mass Spectra <strong>of</strong> PeptidesD.F. HuntDepartments <strong>of</strong> Chemistry and Pathology, University <strong>of</strong>Virginia, Charlottesville, VA, United StatesElectron transfer dissociation mass spectrometry is a break-thoughtechnology for sequencing post-translationally modified peptides.In this technique, radical anions <strong>of</strong> polyaromatic hydrocarbons(fluoranthene and azulene) are employed to transfer electrons tothe carbonyl group in the polyamide backbone <strong>of</strong> multiply chargedpeptides generated by electrospray ionization. Capture <strong>of</strong> an electroninto the peptide backbone reduces the positive charge on the ion byone, and forms a carbonyl radical anion that then abstracts a protonfrom a nearby protonated amino group. The resulting carbonyl radicaltriggers cleavage <strong>of</strong> the adjacent nitrogen-carbon bond to producefragments <strong>of</strong> type c’ and z•. The purpose <strong>of</strong> the workshop is toprovide instruction on how to manually interpret peptide ETD massspectra. Following a lengthy tutorial about ion structures, fragmentationpathways, predictable changes in fragment ion isotope patterns, etc.,we will outline a general approach for the manual interpretation <strong>of</strong>peptide ETD spectra, solve the sequence <strong>of</strong> several post-translationallymodified peptides, assign homework spectra, and reconvene on thesecond day <strong>of</strong> the workshop to go over the homework problems. Apacket <strong>of</strong> lecture notes and handouts will be provided. Attendeesshould bring an inexpensive calculator, pad <strong>of</strong> paper, and a small ruler.Workshop SessionAbstracts52 • <strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine


NotesNotes<strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine • 53


Research Group Presentation AbstractsResearch GroupPresentation Abstracts(R1) Joint Session: Proteome InformaticsResearch Group (iPRG) & Proteomics StandardsResearch Group (sPRG)(R1a) iPRG <strong>2011</strong>: A Study on the Identification <strong>of</strong>Electron Transfer Dissociation (ETD) Mass SpectraL. Martens 10 , M. Askenazi 1 , N. Bandeira 2 , R.J. Chalkley 3 ,K.R. Clauser 4 , E. Deutsch 5 , H.H.N. Lam 6 , W.H. McDonald 7 ,T. Neubert 8 , P.A. Rudnick 91Dana-Farber Cancer Institute, Boston, MA, United States;2University <strong>of</strong> California San Diego, La Jolla, CA, UnitedStates; 3 University <strong>of</strong> California San Francisco, San Francisco,CA, United States; 4 The Broad Institute <strong>of</strong> MIT and Harvard,Cambridge, MA, United States; 5 Institute for SystemsBiology, Seattle, WA, United States; 6 University <strong>of</strong> Scienceand Technology, Hong Kong, China; 7 Vanderbilt UniversitySchool <strong>of</strong> Medicine, Nashville, TN, United States; 8 NewYork University School <strong>of</strong> Medicine, New York, NY, UnitedStates; 9 National Institute <strong>of</strong> Standards and Technology,Gaithersburg, MD, United States; 10 Ghent University and VIB,Ghent, BelgiumThe field <strong>of</strong> mass spectrometry based proteomics has seen several keyinnovations over the last several years, including novel experimentalmethods, new instruments, and unique fragmentation strategies. Thelatter, in the form <strong>of</strong> electron capture dissociation (ECD) and electrontransfer dissociation (ETD) have captured the imaginations <strong>of</strong> manyresearchers, expanding their ability to identify and analyze peptidesand proteins. However, since ECD/ETD spectra differ substantial frommore traditional collision induced dissociation (CID) spectra in boththeir prominent ion series as well as their preferred bond-breakingcharacteristics, the (automatic) interpretation <strong>of</strong> ECD/ETD spectrarequires novel algorithm optimizations. Efficient identification <strong>of</strong> ECD/ETD spectra thus remains an active and exciting field <strong>of</strong> proteomicsinformatics research. In this work, the <strong>ABRF</strong> Proteome InformaticsResearch Group (iPRG) presents the results <strong>of</strong> a collaborative studyfocusing on the analysis <strong>of</strong> an LC-MS/MS dataset from a yeast lysatedigested with Lys-C and enriched for highly charged peptides usingstrong cation exchange fractionation. The data derived from one fractionanalyzed exclusively by ETD was distributed to participants for analysisin several equivalent formats, along with a standardized sequencedatabase derived from the UniProtKB/Swiss-Prot yeast complement,a decoy version <strong>of</strong> this database, and an applicable spectral library.Participants were free to use any and all methods available to them toidentify this fraction, and results were to be submitted using an Exceltemplate. All participant identities were subsequently anonymized, anda survey was used to collect information about participant experienceand s<strong>of</strong>tware tools used to produce the submitted analysis. This uniformcollection <strong>of</strong> data has allowed a thorough comparison <strong>of</strong> participantresults. A summary, including a comparison <strong>of</strong> results submitted bymembers <strong>of</strong> the iPRG, will be presented.(R1b) <strong>ABRF</strong>-sPRG<strong>2011</strong> Study: Development <strong>of</strong>a Comprehensive Standard for Analysis <strong>of</strong> Post-Translational ModificationsA. Ivanov 1 , C. Colangelo 2 , C.P. Dufresne 3 , J.G. Farmar 4 ,D.B. Friedman 5 , C. Kinsinger 6 , K.S. Lilley 7 , K. Mechtler 8 ,B.S. Phinney 9 , S.A. Shaffer 10 , S.T. Weintraub 111Harvard University School <strong>of</strong> Public Health, Cambridge,MA, United States; 2 Yale University, New Haven, CT, UnitedStates; 3 Thermo Fisher Scientific, West Palm Beach, FL,United States; 4 University <strong>of</strong> Virginia, Charlottesville, VA,United States; 5 Vanderbilt University, Nashville, TN, UnitedStates; 6 National Cancer Institute, Bethesda, MD, UnitedStates; 7 University <strong>of</strong> Cambridge, Cambridge, UnitedKingdom; 8 Research Institute <strong>of</strong> Molecular Pathology,Vienna, Austria; 9 Proteomics Core, University <strong>of</strong> CaliforniaDavis Genome Center, Davis, CA, United States; 10 University<strong>of</strong> Massachusetts Medical School, Worcester, MA, UnitedStates; 11 University <strong>of</strong> Texas Health Science Center at SanAntonio, San Antonio, TX, United StatesThe Proteomics Standards Research Group (sPRG) has initiated a studythat focuses on development <strong>of</strong> a standard that can be used in bothassessment <strong>of</strong> a laboratory’s ability to detect an array <strong>of</strong> post-translationalmodifications in a complex proteomic sample and development <strong>of</strong>new approaches for characterization <strong>of</strong> post-translationally modifiedproteins. The sample that has been generated for the first stage <strong>of</strong> thisstudy contains a mixture <strong>of</strong> more than seventy synthetic peptides with avariety <strong>of</strong> modifications, in a tryptic digest <strong>of</strong> six proteins. Modificationsrepresented in the sample include acetylation, methylation, nitration,phosphorylation, and sulfation. The individual proteins were purified,digested and analyzed prior to formulation <strong>of</strong> the sample. Thesynthetic peptides were each analyzed and then mixed with the digests;the mixture was aliquoted and lyophilized in sufficient quantities thatwould permit evaluation by several analytical approaches. The samplewas fully characterized by members <strong>of</strong> the sPRG. It is planned that in<strong>2011</strong>, the sample will be distributed to requesting laboratories for alarger study focusing on PTM characterization. The sPRG will presentresults obtained at different stages <strong>of</strong> preparation and characterization<strong>of</strong> the sample. Approaches for analysis <strong>of</strong> complex proteomic samplescontaining various post-translationally modified proteins will also bediscussed. The sPRG is planning to open the study for sample requestsat the <strong>ABRF</strong> <strong>2011</strong> conference. From the responses returned by theparticipating laboratories, the sPRG will subsequently report on theeffectiveness <strong>of</strong> the approaches used for characterization <strong>of</strong> posttranslationalmodifications in protein digests.54 • <strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine


(R2) Antibodies: Moving Closer to PersonalizedTherapeutics and DiagnosticsB.R. Curtis 1 , R. Umeck 2 , J.E. Harlan 31Blood Center <strong>of</strong> Whisonsin, Milwaukee, WI, United States;2Meso Scale Discovery, Gaithersburg, MD, United States;3Abbott Laboratories, Abbott Park, IL, United StatesThe possibilities <strong>of</strong> personalized medicine are largely rooted insuccessfully exploiting the ever-growing genome and proteome-wideunderstanding <strong>of</strong> human biology. Translating this knowledge base intothe clinic requires, among other things, being able measure the patient’sgenomic and proteomic disease signature in a routine manner. Thus,expanded focus on assays and platforms capable <strong>of</strong> measuring thesespecific signatures within tens, hundreds or even thousands <strong>of</strong> complexpatient samples simultaneously is critical. Antibody-based assays andplatforms have become a centerpiece <strong>of</strong> many technologies pushingthese boundaries because <strong>of</strong> the antibody’s intrinsic specificity, diverserecognition capacity and ease with which they can be produced.Furthermore, these same characteristics have made them an equallypromising prospect for therapeutic approaches. This seminar trifectawill highlight the pivotal role antibodies have in multiplex biomarkerassay development and therapeutic venues. Specifically: 1. BrianCurtis will discuss a robust multiplex assay platform to detect humanplatelet auto-antibodies and platelet antigen polymorphisms bycombining bead array technologies with flow cytometry. Such asimple, yet high-throughput platform would facilitate routine testingfor platelet disorders like neonatal alloimmune thrombocytopenia,multi-platelet transfusion refractoriness, and post-transfusion purpura;2. Robert Umeck will summarize the challenges in developingantibody-based multiplex assays and how Meso Scale Discoveryutilizes an electrochemiluminescence detection system combinedwith a patterned multi-array format to generate multiplex assays fornearly any application. Several actual examples will be presented todemonstrate how this approach could support personalized medicine;3. John Harlan will introduce and discuss the evolution <strong>of</strong> therapeuticantibodies. To illustrate where the field is venturing, he will highlightAbbott Laboratory’s new bi-specific antibody format.(R3) Joint Session: Genomic Variation ResearchGroup (GVRG) & DNA Sequencing ResearchGroup (DSRG)(R3a) Evaluation <strong>of</strong> DNA Whole GenomeAmplification Technologies for GenotypingA. Hutchinson 1 , C. Dagnall 1 , C. Nicolet 2 , H. Escobar 3 ,S. Blake 4 , B. Sanderson 5 , B. Kingham 6 , K. Jonscher 71Core Genotyping Facility, SAIC-Frederick, Inc., NCI-Frederick, Frederick, MD, United States; 2 Data ProductionFacility, USC Epigenome Center, Los Angeles, CA, UnitedStates; 3 DNA Sequencing Operations, Eur<strong>of</strong>ins MWGOperon, Huntsville, AL, United States; 4 DNA Core Facility,University <strong>of</strong> Missouri, Columbia, MO, United States; 5 LifeTechnologies, Austin, TX, United States; 6 Director, DNASequencing & Genotyping Center, University <strong>of</strong> Delaware,Newark, DE, United States; 7 Director, SBCF and TATCProteomics, University <strong>of</strong> Colorado Denver, Denver, CO,United StatesThe evolution <strong>of</strong> genomic technologies is occurring rapidly and <strong>of</strong>tenrequires large amounts <strong>of</strong> source DNA. There is also an expandeddesire to analyze smaller numbers <strong>of</strong> cells for higher resolution studiesas well as to take advantage <strong>of</strong> large numbers <strong>of</strong> archived samples (e.g.FFPE, serum, etc.). To provide enough material for the newest genomictechnologies, whole genome amplification (WGA) has reemerged as animportant and necessary technique. With some new WGA productson the market, we have evaluated the quantity and quality <strong>of</strong> WGAproducts generated as well their performance on some <strong>of</strong> the currentgenomic applications. The GVRG has completed a benchmarkingstudy evaluation <strong>of</strong> 6 commercially available WGA kits using severalgenotyping assays. Utilizing 6 samples, the different WGA kits weretested following the manufacturer’s protocols. The samples includedCoriell DNA from a trio <strong>of</strong> CEPH individuals, 2 FFPE DNAs, bothfrom the same individual (1 newly extracted and the other extracted2 years ago), and 1 sample from the Coriell trio that was fragmentedprior to amplification for a total <strong>of</strong> 6 samples tested. The WGA kitsincluded those from Sigma, NuGen, Qiagen and GE Life Sciences. The6 samples were amplified as blind duplicates at several GVRG memberlab sites resulting in a total <strong>of</strong> 78 amplified products. Quality metricswere performed on the 78 amplified products using several differenttechniques in order to provide measurements on yield, fragment size,and concentration. Several widely used genotyping methods wereused to evaluate the amplification products, including Illumina HumanOmni1 Quad Beadchip, TaqMan copy number and SNP genotypingassays, and STR genotyping. Data will be presented on genotypingconcordance and loss <strong>of</strong> heterozygosity checks across the differentplatforms where possible.Research GroupPresentation Abstracts<strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine • 55


Research GroupPresentation Abstracts(R3b) DNA Sequencing Research Group (DSRG)(R3b-1) Comparison <strong>of</strong> Custom Target EnrichmentMethods; Agilent vs. NimblegenA. Perera 4 , K. Bodi 1 , P.S. Adams 2 , D. Bintzler 3 , K. Dewar 5 ,D.S. Grove 6 , J. Kieleczawa 7 , R.H. Lyons 8 , T. Neubert 9 ,A.C. Noll 1 , S. Singh 10 , R. Steen 11 , M. Zianni 121Tufts University, Boston, MA, United States; 2 TrudeauInstitute, Saranac Lake, NY, United States; 3 DNA Analysis,Inc., Cincinnati, OH, United States; 4 Stowers Institute, KansasCity, MO, United States; 5 McGill University, Montreal, QC,Canada; 6 Pennsylvania State University, University Park,PA, United States; 7 Pfizer Research, Cambridge, MA, UnitedStates; 8 University <strong>of</strong> Michigan, Ann Arbor, MI, UnitedStates; 9 New York University, New York, NY, United States;10University <strong>of</strong> Minnesota, Minneapolis, MN, United States;11Harvard Medical School, Cambridge, MA, United States;12Ohio State University, Columbus, OH, United StatesOver the last four years, we witnessed the tremendous advances inNext Generation Sequencing (NGS) that have dramatically decreasedthe cost <strong>of</strong> whole genome sequencing. However, the cost <strong>of</strong> sequencinglarger genomes is still significant. In addition and depending on thegoal <strong>of</strong> study, whole genome sequencing creates a large amount <strong>of</strong>additional/auxiliary data that complicates data analysis. There areseveral commercial methods available for isolating subsets <strong>of</strong> genomesthat greatly enhance the efficiency <strong>of</strong> NGS by allowing researchersto focus on their regions <strong>of</strong> interest. For the 2009-11 DSRG study,we compared products from two leading companies; Agilent andNimblegen that <strong>of</strong>fer custom enrichment methods. Both companiesobtained the same genomic DNA stock and performed DNA captureon the same specified regions. Following capture, the Illumina GenomeAnalyzer IIx system was used, in two different laboratories, to generatethe sequence data. We present our data comparing in terms <strong>of</strong> cost,quality, reproducibility and most importantly completeness and depth<strong>of</strong> coverage. Acknowledgements: We would like to thank Agilent,Illumina and Nimblegen for all their support in making this studypossible.(R3b-2) A Methodology Study for MetagenomicsUsing Next Generation SequencersS. Singh 1 , D. Grove 21Biomedical Genomics Center, University <strong>of</strong> Minnesota,Saint Paul, MN, United States; 2 Genomics Core Facility, HuckInstitutes for Life Sciences, Pennsylvania State University,University Park, PA, United StatesMetagenomics is one <strong>of</strong> several genomics applications, which hasbenefited immensely from the high throughput and cost efficacy <strong>of</strong>Next generation sequencers. And although hundreds <strong>of</strong> studies onmetagenome analysis have been published over the past few years,the methodology for conducting them is still very much evolving.In this DSRG study we will evaluate the influence <strong>of</strong> various samplepreparation methods, specifically DNA extraction and amplificationapproaches, on data output along with a comparative analysis <strong>of</strong> Nextgeneration sequencing platforms. We will study the effect <strong>of</strong> thesedifferent experimental and technical strategies on determination <strong>of</strong>sample biodiversity.(R4) Joint Session: Protein Sequencing ResearchGroup (PSRG) & Glycoprotein Research Group(gPRG)(R4a) PSRG <strong>2011</strong> Study Results: SensitivityAssessment <strong>of</strong> Terminal Sequencing TechniquesUsing an Unknown ProteinJ.J. Walters 1 , W. Sandoval 2 , K. Mawuenyega 3 ,J.S. Smith 4 , H. Remmer 5 , B. Xiang 6 , D. Suckau 7 , V. Katta 2 ,P. Hunziker 81Sigma-Aldrich, St. Louis, MO, United States; 2 Genentech,Inc., South San Francisco, CA, United States, 3 WashingtonUniversity School <strong>of</strong> Medicine, St. Louis, MO, United States,4University <strong>of</strong> Texas Medical Branch, Galveston, TX, UnitedStates, 5 University <strong>of</strong> Michigan, Ann Arbor, MI, UnitedStated, 6 Monsanto Company, St. Louis, MO, United States,7Bruker Daltonics, Bremen, Germany, 8 University <strong>of</strong> Zurich,Zurich, SwitzerlandEstablishing the N-terminal sequence <strong>of</strong> intact proteins plays a criticalrole in biochemistry and potential drug development. N-terminalsequence analysis is necessary for quality control <strong>of</strong> protein biologics,for determining sites <strong>of</strong> signal peptide cleavage events, as a first stepin elucidating the sequences <strong>of</strong> genes from uncommon species andfor the characterization <strong>of</strong> monoclonal antibodies. Automated Edmandegradation has been the method <strong>of</strong> choice for these analyses. However,alternate methods for N-terminal sequence analysis have emerged. Therecent PSRG studies have established that Edman sequencing and massspectrometry based techniques have varied strengths and weaknessesdepending on several experimental factors and both play an importantrole in terminal sequencing. With this complimentary role realized,the <strong>2011</strong> PSRG study attempts to evaluate the sensitivity limits <strong>of</strong> thevarious sequencing techniques. The PSRG distributed three samplesets <strong>of</strong> 3 tubes each, varying by sample format (lyophilized, gel sliceor membrane piece). Each set <strong>of</strong> three samples contains the samerecombinant protein with increasing amounts <strong>of</strong> material. The sequence<strong>of</strong> this protein is not listed in any database. Participants could requestany one, two, or all three sample sets. Including PSRG committee, a total<strong>of</strong> 38 participants requested 74 sample sets. The participants wereasked to determine as many amino acids from both termini by theirmethod <strong>of</strong> choice, and were encouraged to try multiple methods forsequence elucidation. Study participants were directed to a websiteto anonymously upload sequences and supporting data. Our analysisfocuses on the length and accuracy <strong>of</strong> the sequence calls reported bythe participants, and how that compares with decreasing amounts <strong>of</strong>protein and the type <strong>of</strong> sample format analyzed. A comparison <strong>of</strong> theresults obtained by Edman chemistry and by alternative technologiesas well as information on the type <strong>of</strong> instruments and protocols isreported.56 • <strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine


(R4b) gPRG: Toward Consensus on Glycan Analysis:Reliable Methods and ReproducibilityJ. Zaia 1 , D. Kolarich 2 , R. Orlando 31Boston University, Boston, MA, United States; 2 Max PlanckInstitute <strong>of</strong> Colloids and Interfaces, Berlin, Germany; 3 TheUniversity <strong>of</strong> Georgia Carbohydrate Research Center,Atlanta, GA, United StatesThe field has undertaken over the past few years a series <strong>of</strong> studies toevaluate methods for glycan analysis. There are several methods thatare widely used at this time for characterization <strong>of</strong> glycoprotein glycansthat appear to be generally reliable. These include (1) reductiveamination followed by reversed phase chromatography, (2) reductiveamination with matrix assisted laser desorption/ionization (MALDI)mass spectrometry (MS), (3) reductive amination with electrosprayliquid chromatography-MS, and (4) permethylation with tandem MS.The <strong>ABRF</strong> glycoprotein research group (gPRG) is planning a study inwhich participant laboratories carry out glycan analysis according toprotocols representing best practice in the field. The results will bepresented at the 2012 <strong>ABRF</strong> meeting. The results will be useful fordemonstrating the reproducibility <strong>of</strong> glycan analysis using comparableprotocols. They will also highlight the extent to which different methodsbias the glycan analysis results obtained. This workshop will summarizethe history and use <strong>of</strong> the glycan analysis methods proposed for the2012 study. There will be opportunity for discussion and comment onthese methods.(R5) Joint Session: Nucleic Acids Research Group(NARG) & MicroArray Research Group (MARG)(R5a) Determining miRNA Expression Levels inDegraded RNA Samples Using Real-Time RT-qPCRand Microarray TechnologiesS. Chittur 1 , S. Tighe 2 , J. Holbrook 3 , V. Nadella 4 ,R. Carmical 5 , K. Sol-Church 3 , A.T. Yueng 61State University <strong>of</strong> New York at Albany, Albany, NY, UnitedStates; 2 University <strong>of</strong> Vermont, Burlington, VT, United States;3Nemours/A.I. duPont Hospital for Children, Wilmington, DE,United States; 4 Ohio University, Athens, OH, United States;5The University <strong>of</strong> Texas Medical Branch, Galveston, TX ,United States; 6 Fox Chase Cancer Center, Philadelphia, PA,United StatesThe Nucleic Acid Research Group (NARG) has previously conductedstudies evaluating the impact <strong>of</strong> RNA integrity and priming strategieson cDNA synthesis and real-time RT-qPCR. The results <strong>of</strong> last year’s fieldstudy as it relates to degraded RNA will be presented. In continuation<strong>of</strong> the RNA integrity theme, this year’s study was designed to evaluatethe impact <strong>of</strong> RNA integrity on the analysis <strong>of</strong> miRNA expressionusing real-time RT-qPCR. Target section was based on data obtainedby the Microarray Research Group (MARG) and other published datafrom next gen sequencing. These 9 miRNAs represent three groups<strong>of</strong> miRNA that are expressed at low, medium or high levels in the FirstChoice human brain reference RNA sample. Two popular RT primingstrategies tested in this study include the Megaplex miRNA TaqManassay (ABI) and the RT2 miRNA qPCR assay (Qiagen/SA Biosciences).The basis for the ABI assay design is a target-specific stem-loopstructure and reverse-transcription primer, while the Qiagen designcombines poly(A) tailing and a universal reverse transcription in onecDNA synthesis reaction. For this study, the human brain referenceRNA was subject to controlled degradation using RNase A to RIN (RNAIntegrity Number) values <strong>of</strong> 7 (good), 4 (moderately degraded), and2 (severely degraded).These templates were then used to assess bothRT methods. In addition to this real-time RT-qPCR data, the same RNAtemplates were further analyzed using universal poly(A) tailing andhybridization to Affymetrix miRNA GeneChips. This talk will provideinsights into RT priming strategies for miRNA and contrast the qPCRresults obtained using different technologies.(R5b) Microarray Research Group Projects, 2010-11D. Baldwin 1 , N.G. Reyero-Vinas 2 , N. Jafari 31Penn Molecular Pr<strong>of</strong>iling Facility, University <strong>of</strong> Pennsylvania,Philadelphia, PA, United States; 2 Jackson State University,Jackson, MS, United States; 3 Northwestern University,Evanston, IL, United StatesMembers <strong>of</strong> the MARG will discuss our research projects: Comparison<strong>of</strong> microarray and deep sequencing platforms for microRNA pr<strong>of</strong>iling,Performance <strong>of</strong> a synthetic human microRNA reference panel,Participation in the SEQC Sequencing Quality Control consortium, andRNA-Seq pr<strong>of</strong>iling <strong>of</strong> environmental samples exposed to the Gulf oilspill.(R6) Joint Session: Protein Expression ResearchGroup (PERG) & Molecular InteractionsResearch Group (MIRG)(R6a) PERG Research Group Presentation: RefoldingStudyC. KinslandCornell University, Ithaca, NY, United StatesCurrently, the overwhelming majority <strong>of</strong> protein purification projectsstart with a recombinant protein expressed in a suitable host. For arange <strong>of</strong> reasons, E. coli is the predominant expression host yet a largepercentage <strong>of</strong> proteins expressed therein are found in an insolubleform called inclusion bodies. Inclusion bodies have the advantage <strong>of</strong>consisting <strong>of</strong> relatively homogeneous protein, which can simplify thepurification process. However, this leaves the challenge <strong>of</strong> solubilizingand refolding the protein into its native and biologically active structure.The conditions for efficient refolding are particular for each protein andthere are a wide range <strong>of</strong> methods to choose from. For this and otherreasons, many researchers are hesitant to pursue a refolding strategyto obtain a target protein. An overview <strong>of</strong> refolding methods andstrategies will be presented along with a description <strong>of</strong> the upcomingProtein Expression Research Group (PERG) refolding study.Research GroupPresentation Abstracts<strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine • 57


Research GroupPresentation Abstracts(R6b) Conclusions from the MIRG 2010 BenchmarkStudy: Molecular Interactions in a Three ComponentSystem and Presentation <strong>of</strong> <strong>2011</strong> Survey Results onLabel-Free TechnologiesA.P. Yamniuk 1 , S.P. Yadav 5 , S. Bergqvist 2 , M.L. Doyle 1 ,E. Eisenstein 3 , M.K. Robinson 4 , T. Neubert 61Bristol-Myers Squibb, Princeton, NJ, United States; 2 Pfizer, LaJolla, CA, United States; 3 University Maryland BiotechnologyInstitute, Baltimore, MA, United States; 4 Fox Chase CancerCenter, Philadelphia, PA, United States; 5 Cleveland ClinicFoundation, Cleveland, OH, United States; 6 New YorkUniversity School <strong>of</strong> Medicine, New York, NY, United StatesCharacterizing the assembly <strong>of</strong> multi-protein complexes and thecompetition between multiple protein ligands for a given target arecommon challenges faced by core facilities. The MIRG2010 Benchmarkstudy was designed to assess participants’ ability to correctly describethe interactions between two protein ligands and their target proteinusing primarily biosensor technologies such as surface plasmonresonance. Participants were provided with microgram quantities <strong>of</strong>three proteins (A, B and C) and asked to determine if a ternary A-B-Ccomplex can form, or if ligands B and C bind competitively to proteinA. This presentation will summarize the conclusions from the 2010Benchmark Study, and provide perspective on the potential for futureapplication <strong>of</strong> this system as a reference standard for quantitativecharacterization <strong>of</strong> protein-protein interactions using biosensortechnologies. The field <strong>of</strong> label-free biophysical technologies likesurface plasmon resonance (SPR) and isothermal calorimetry (ITC) arebecoming indispensable in translational research and in the discoveryphase <strong>of</strong> biotherapeutics. Investigators are much more aware about thedevelopments in biomolecular interaction analysis using SPR and ITCand usefulness <strong>of</strong> these technologies in designing better drugs basedon biomolecules and vaccines. The Molecular Interaction ResearchGroup (MIRG) <strong>of</strong> <strong>ABRF</strong> has conducted an on-line survey to capture therecent explosive developments in these technologies. The survey wastargeted to both academia and pharmaceutical industry and the surveydata will be presented during the meeting.(R7) Light Microscopy Research Group (LMRG)(R7-1) Point Spread Functions, Spectral Calibration,and BeyondR. Stack, R. ColeWadsworth Center, New York State Department <strong>of</strong> Health,Albany, NY, United StatesModern light microscopes are highly evolved opto-electronicmechanicaldevices. Establishing instrument performance is crucial inensuring that reliable and accurate images can be acquired. Imagers, aswell as granting agencies, need to be confident that data collected willbe uniform and quantifiable both temporally and from instrument toinstrument. Last year, in phase-one <strong>of</strong> our world-wide research studyon instrument performance, we successfully concentrated our effortson three image-based tests: long and short term stability <strong>of</strong> illuminationsources, uniformity <strong>of</strong> field illumination, and co-registration acrossvarious wavelengths. A manuscript summarizing the phase-one studyhas been submitted to and accepted by Microscopy & Microanalysis,one <strong>of</strong> the highest rated imaging journals. In the coming year, phasetwo<strong>of</strong> our instrument performance study will focus on determining thefollowing: the point spread function <strong>of</strong> an imaging system, the system’sspectral separation ability and the spectral calibration and resolution<strong>of</strong> the detection system. As with the phase-one study, the goal <strong>of</strong> thisstudy will be neither to compare the performance <strong>of</strong> different brands<strong>of</strong> instruments, nor to ascertain which brand had better performancein a given area. Instead, the goal <strong>of</strong> our proposed phase-two study iscontinue to focus on determining the current state <strong>of</strong> modern imagingsystems through straightforward, efficient and robust tests. These testswill aid imagers in the early detection <strong>of</strong> system problems. Moreover,these tests will continue to help define relative standards that will assistboth core personnel and imagers in maintaining their instruments inoptimal operating conditions.(R7-2) Deconvolution: Core Concepts, Algorithmsand Advanced IssuesB. Northan, N. BeaversMedia Cybernetics, Guilderland, NY, United StatesDeconvolution is a computational technique used to remove blurfrom images. In this presentation image formation in the microscopewill be reviewed and it will be explained what deconvolution doesand why deconvolution is needed as a post acquisition processingstep. The concept <strong>of</strong> Point Spread Function (PSF) will be introduced.Several approaches to deconvolution and deblurring exist from simplesubtractive methods to complex iterative approaches. The majorapproaches will be examined. It will be explained why in the presence<strong>of</strong> noise a statistical approach is required. Blind deconvolution will beintroduced. The concept <strong>of</strong> Point Spread Function will be examinedin detail. The effect <strong>of</strong> microscope modality, lens parameters, andspecimen parameters on PSF shape will be discussed. Theoretical PSFcalculation and PSF measurement using beads will be reviewed withexamples. It will be explained why theoretical and measured PSFs candiffer from the true system PSF. Blind deconvolution will be furtherexamined as a tool to deal with the uncertainty <strong>of</strong> the true form <strong>of</strong>the PSF. Practical guidelines for data acquisition will be reviewed. Therelationship between sampling rate and quality <strong>of</strong> deconvolutionresults will be explained.58 • <strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine


(R8) Joint Session: Proteomics Research Group(PRG) & Metabolomics Research Group (MRG)(R8a) PRG-<strong>2011</strong>: Defining the Interaction betweenUsers and Suppliers <strong>of</strong> Proteomics ServicesD. Hawke 1 , T.M. Andacht 2 , M.K. Bunger 3 , C. Bystrom 4 ,L. Dangott 5 , H. Molina 6 , R.L. Moritz 7 , R.E. Settlage 8 ,C.W. Turck 91University <strong>of</strong> Texas MD Anderson Cancer Center, Houston,TX, United States; 2 Centers for Disease Control andPrevention, Atlanta, GA, United States; 3 RTI International,Research Triangle Park, NC, United States; 4 QuestDiagnostics, San Juan Capistrano, CA, United States;5Texas A&M University, College Station, TX, United States;6Center for Genome Regulation, Barcelona, Spain; 7 Institutefor Systems Biology, Seattle, WA, United States; 8 VirginiaBioinformatics Institute, Blacksburg, VA, United States; 9 MaxPlanck Institute <strong>of</strong> Psychiatry, Munich, GermanyOver the last ten years the Proteomics Research Group (PRG) hasundertaken technical studies that have covered a wide range <strong>of</strong>issues unique to the rapidly developing field <strong>of</strong> proteomics and haveincluded a range <strong>of</strong> qualitative and quantitative experiments. The PRGstudies have resulted in a great deal <strong>of</strong> attention not only within the<strong>ABRF</strong> community but also outside as is evident from numerous articlesdealing with proteomics methods, procedures and standardization.As the field continues to develop, the diversity <strong>of</strong> instrumentationand laboratory workflows have grown in tandem. Therefore, in thePRG<strong>2011</strong> study it seemed especially useful to perform a survey tohelp the PRG define future studies based on the current blend <strong>of</strong>sample types and technologies and obtain a view <strong>of</strong> emerging trends.A survey was created to ascertain three main insights into core facilityfunction: 1) How labs interact with their clients, 2) The capacity <strong>of</strong> labsto meet the demands <strong>of</strong> their clients, and 3) The blend <strong>of</strong> experimentaltechniques <strong>of</strong>fered to and requested by clients. Survey questions weredesigned to obtain information from both users <strong>of</strong> core facilities andthe directors and personnel <strong>of</strong> core facilities. Questions covered suchtopics as the type and age <strong>of</strong> instruments in use, how data is analyzedand presented to client, sources <strong>of</strong> funding, and emerging proteomicstrends. Results are compiled en masse and presented without regardto institution. Early results reveal that about 2/3 <strong>of</strong> the responders arenot <strong>ABRF</strong> members, and at least one lab still has an operational massspectrometer that was acquired in 1990!(R8b) Metabolomics Research Group <strong>2011</strong> StudyW.R. Wik<strong>of</strong>f 1 , J.M. Asara 3 , V.V. Tolstikov 1 , P. Aronov 2 ,B. Kesler 4 , V. Shulaev 5 , C.W. Turck 61University <strong>of</strong> California Davis, Davis, CA, United States;2Stanford University, Stanford, CA, United States; 3 BethIsrael Deaconess Medical Center, Boston, MA, United States;4Thermo Fisher Scientific, Redwood City, CA, United States;5University <strong>of</strong> North Texas, Denton, TX, United States; 6 MaxPlanck Institute <strong>of</strong> Psychiatry, Munich, GermanyThe <strong>ABRF</strong> Metabolomics Research Group (MRG) was formed in2009 and aims to educate research scientists and resource facilitiesin the analytical approaches and management <strong>of</strong> data resulting fromcomprehensive metabolite studies and to promote the science andstandardization <strong>of</strong> metabolomic analyses for a variety <strong>of</strong> applications.Last year the MRG conducted a ‘Survey Study’ on the current use <strong>of</strong>metabolomics technologies and procedures in core facilities. This yearthe MRG is organizing a ‘Research Study’ involving a spiked plasmasample. The study sample consists <strong>of</strong> a human bi<strong>of</strong>luid as the matrix,replicating a typical small scale metabolomics pilot experiment thateither a core or research laboratory would perform. The sampleconsists <strong>of</strong> two groups <strong>of</strong> normal human plasma (NIST plasma ‘StandardReference Material’) with spiked in compounds. There are threebiological replicates in each group (n = 3 design) with different levels<strong>of</strong> spiked compounds differentiating the two groups. Participants areasked to determine statistical significance, fold change, and identifycompounds that differ significantly between groups A and B. The designreflects issues encountered in an actual metabolomics experimentconducted with human or animal specimens. The study is compatiblewith many methodological approaches in metabolomics, including,but not limited to LC/MS, GC/MS, NMR, as well as other methods. Aswith any metabolomics pr<strong>of</strong>iling experiment, the best results wouldbe expected using a combination <strong>of</strong> approaches. The study is the first<strong>of</strong> its kind in the field <strong>of</strong> metabolomics and is expected to produceimportant information on the strengths and limitations <strong>of</strong> the variousplatforms and technologies that are commonly used for comprehensivemetabolite analyses.Research GroupPresentation Abstracts<strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine • 59


Poster Session Abstracts**<strong>ABRF</strong> Poster Award Semifinalists102 Designing an Institutional Web-basedCore Facility Management SystemPoster Abstracts101 Center for Genome Research andBiocomputing at Oregon StateUniversityC. Rosato, M. Dasenko, A. Girard, M. Peterson,C. Sullivan, J.C. CarringtonCenter for Genome Research & Biocomputing,Oregon State University, Corvallis, OR, UnitedStatesThe Center for Genome Research and Biocomputing (CGRB) CoreLab at Oregon State University provides services for fee in genomictechnologies (DNA sequencing, DNA genotyping (fragment analysis))and in functional genomic technologies (microarray). We manage aZeiss LSM510Meta confocal microscope as a multi-user instrument;training is required and assistance is available, both for fee. Sequencingis provided both for traditional Sanger sequencing on an AB 3730 andultra high throughput sequencing on the Illumina Genome AnalyzerGAIIX. With the most recent s<strong>of</strong>tware and reagent upgrades, 2 x150bp runs are now supported with up to 40 million reads per lanepassing filter. DNA genotyping (fragment analysis) runs on the AB3730. Our microarray services include Affymetrix and NimbleGenplatforms, and sample labeling, hybridization and scanning are<strong>of</strong>fered. We purchased ArrayStar microarray analysis s<strong>of</strong>tware, whichis accessible to our researchers through our Biocomputing clusternetwork. The Agilent BioAnalyzer service <strong>of</strong>fers High Sensitivity DNAand RNA applications. Our Biocomputing capabilities have expanded.This past year we reorganized the computing resources into a cloudarchitecture. GENOME Cloud is at 507 Processors, 1.5TB Memoryand 300+TB Storage. We process around 20,000 jobs / day on thecloud. We have increased access to our computational resources,decreased analysis turn-around time, and we have added new toolsto monitor computational resources. The Biocomputing changes toour High Throughput Sequencing service include removing the IPARunit from the network and simplifying data pathways both for datamanagement and researcher analysis capabilities. The CGRB Core Labalso maintains multi-user instruments available to researchers. Theseinclude a Zeiss LSM510Meta confocal microscope, an AB 7500 FASTqPCR instrument includes High Resolution Melting (HRM) capability, aStorm 820 Phosphoimager, a Nanodrop, an Invitrogen Qubit, a GenetixQ-Pix colony picker, an Axon Genepix 4200A microarray scanner and afluorescent plate reader.A. Hagen 1 , D. Tabarini 2 , S. Clisham 3 , D. John 31iLab Solutions, LCC, Cambridge, MA, UnitedStates; 2 Memorial Sloan-Kettering Cancer Center,New York, NY, United States; 3 Institute for SystemsBiology, Seattle, WA, United States; 4 Dana-FarberCancer Institute, Boston, MA, United StatesThe authors and their four institutions collaborated to (i) identify the keychallenges to core facility management; (ii) identify the requirementsfor an effective core facility management system; (iii) design, test anddeploy such a system. Through a series <strong>of</strong> interviews with all participantsin the core work flow (customers, core staff, administrators), the teamidentified a number <strong>of</strong> key challenges, including: (i) difficulty forresearchers in identifying available services; (ii) inconsistent processesfor requesting services; (iii) inadequate controls for approving servicerequests; (iv) inefficient processes for tracking and communicating aboutproject processes; (v) time-consuming billing practices; (vi) incompleterevenue capture; (vii) manual reporting processes. The team identifiedthe following requirements for a system to address these challenges:(i) ability to support a broad range <strong>of</strong> core business practices suchas complex quote generation and project management; calendaring/equipment reservation management; sample tracking; complex forms;and import <strong>of</strong> usage data from hardware; (ii) ability to <strong>of</strong>fer servicesfor both internal and external customers, including flexible pricingand <strong>of</strong>f-site access; (iii) ability to interact with institutional financialsystems (e.g. SAP, PeopleS<strong>of</strong>t, Lawson, SunGard Banner) and identifymanagement systems (e.g. Micros<strong>of</strong>t Active Directory, LDAP, and otherSAML 2.0-compliant services). The team developed and deployed thissystem across the collaborative partners, as well as other major researchinstitutions.103 RI-INBRE Centralized Research CoreFacilityA. Ahmed, N. NousUniversity <strong>of</strong> Rhode Island, Kingston, RI, UnitedStatesThe RI-INBRE Centralized Research Core Facility inaugurated in July2003, is being supported by the Rhode Island IDeA Network <strong>of</strong>Biomedical Research Excellence (RI-INBRE) grant from NCRR /NIHand by the participating institutions that include: University <strong>of</strong> RhodeIsland, Brown University, Rhode Island College, Providence College,Salve Regina University, and Roger Williams University. This facility islocated in the College <strong>of</strong> Pharmacy at the University <strong>of</strong> Rhode Island’sKingston campus. It is equipped with instrumentation in biomedical,pharmaceutical and biotechnological research. The Core Facility isproviding access to research instrumentation and training supportto RI-INBRE participants as well as other scientists affiliated withacademic institutions and the private sector throughout the state <strong>of</strong>Rhode Island. Instrument reservation and scheduling are available,through the core facility’s website (wwww.uri.edu/inbre/corelab).All new users are supported with operator assisted access to theequipment. Independent access to the equipment is also available to60 • <strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine


all trained users. In addition, full service access via sample submissionis provided particularly in ICP-MS, LC/MS/MS and N-terminal proteinsequencing. Technical staff members manage the laboratory and areavailable to assist in the operation <strong>of</strong> various instruments and to analyzesamples on a fee-for-service basis. (Supported by NIH-NCRR Grant #1P20RR16457)104 Implementation <strong>of</strong> QualityManagement in Core ServiceLaboratoriesB. Hicks, T. Creavalle, J. Dickens, K. Haque,C. Raley, M.W. SmithGenetics and Genomics Group, AdvancedTechnology <strong>Program</strong>, SAIC-Frederick, NationalCancer Institute at Frederick, Frederick, MD, UnitedStatesThe Genetics and Genomics group <strong>of</strong> the Advanced Technology<strong>Program</strong> <strong>of</strong> SAIC-Frederick exists to bring innovative genomic expertise,tools and analysis to NCI and the scientific community. The SequencingFacility (SF) provides next generation short read (Illumina) sequencingcapacity to investigators using a streamlined production approach.The Laboratory <strong>of</strong> Molecular Technology (LMT) <strong>of</strong>fers a wide range<strong>of</strong> genomics core services including microarray expression analysis,miRNA analysis, long read (Roche) next generation sequencing,transgenic genotyping, Sanger sequencing, and clinical mutationdetection services to investigators from across the NIH. SF and LMT areworking together to bring online the third generation Pacific BioscienceSMRT sequencing platform. As the technology supporting thisgenomic research becomes more complex, the need for basic qualityprocesses within all aspects <strong>of</strong> the core service groups becomes critical.The Quality Management groupworks alongside members <strong>of</strong> theselabs to establish or improve processes supporting operations control(equipment, reagent and materials management), process improvement(reengineering/optimization, automation, acceptance criteria for newtechnologies and tech transfer), and quality assurance and customersupport (controlled documentation/SOPs, training, service deficienciesand continual improvement efforts). Implementation and expansion <strong>of</strong>quality programs within unregulated environments demonstrates SAIC-Frederick’s dedication to providing the highest quality products andservices to the NIH community.105 Cornell University Life Sciences CoreLaboratories CenterG. Grills, J. VanEe, P. Schweitzer, S. Zhang,R. Williams, J. Pillardy, Q. Sun, W. Wang, Y. Li,D. Betel, T. Stelick, J. Spisak, L. Cote, R. Cameron,H. Wroblewski, B. Hover, L. Zhang, J. Mosher,Y. Xin, G. Westby, J. Busuttil, S. Monni,R. Sherwood, A.C. Ptak, W. Chen, J. McCardle,C. Bayles, J. Dela Cruz, M. Riccio, R. Bukowski,L. Ponnala, C. Myers, H. Singh, M. Howard,J. Flaherty, A. Manocchia, E. Dodge, K. Smith,C. Aquadro, A. Melnick, T. Brenna, W. Zipfel,A. Clark, A. Siepel, L. Carr, J.K.C. RoseCornell University, Ithaca, NY, United StatesThe Cornell University Life Sciences Core Laboratories Center (CLC)provides an array <strong>of</strong> genomics, proteomics, imaging and informaticsshared research resources and services to the university communityand to outside investigators. The CLC includes fee-for-service research,technology testing and development, and educational components.The Center has seven core facilities, including genomics (DNAsequencing, genotyping, and microarrays), epigenomics, proteomicsand mass spectrometry, microscopy and imaging, bioinformatics, bio-IT, and advanced technology assessment. The CLC is part <strong>of</strong> a NewYork State designated Center for Advanced Technology in Life ScienceEnterprise. The mission <strong>of</strong> the CLC is to promote research in the lifesciences with advanced technologies in a shared resource environment.Use <strong>of</strong> the CLC resources and services is steadily increasing due to thegrowth in the number and types <strong>of</strong> cores in the center, to the expansion<strong>of</strong> existing services and the implementation <strong>of</strong> new core technologies,and to the coordinated integration and synergy <strong>of</strong> services betweenthe CLC cores. Multidisciplinary support for multi-functional instrumentplatforms is implemented by integrated operations <strong>of</strong> the CLC corefacilities. Investigators are <strong>of</strong>fered coordinated project consultationswith the directors and staff <strong>of</strong> all relevant cores during the design,data production and analysis phases <strong>of</strong> their projects. The CLC isinvolved in establishing and supporting multidisciplinary researchprojects that involve both intercampus initiatives and multi-institutionalcollaborations. With a concentration <strong>of</strong> advanced instrumentation andexpertise in their applications, the CLC is a key resource for life sciencesresearch.106 Searchable Core Facility Database:Building Resource BridgesB. Fleming, T. HunterVermont Genetics Network, University <strong>of</strong> Vermont,Burlington, VT, United StatesThe VGN Searchable Core Facility Database (http://vgn.uvm.edu/corefacilities) is a directory <strong>of</strong> Core Facilities primarily focused on NorthAmerica but with entries from around the world. It is a tool intendedto foster collaboration and assist cores in growing their user base andproviding networking opportunities. It is populated with Core Facilitiesthat have voluntarily listed themselves and would like to be contactedby researchers and other core facilities for potential collaborations.Benefits: Allows researchers to locate resources needed for their studies;Provides a channel for facilities to collaborate; and Facilitates cores toreach financial sustainability. Researchers are able to perform searchesonline by service <strong>of</strong>ferings, location, association, and key phrases to finda facility that will best meet their needs. Information listed for individualcores include: short description <strong>of</strong> core, contact name, email, address,services <strong>of</strong>fered, hyperlink to website, equipment, and date <strong>of</strong> lastrevision <strong>of</strong> information. The data can be exported to an excel readableXML file. The database currently lists 292 cores, representing 39 statesplus DC, 104 institutions, and 10 associations.107 Dartmouth Genomics SharedResourcesJ. Hamilton, H. Trask, W. Taylor, C. TomlinsonDartmouth Medical School, Norris Cotton CancerCenter, Hanover, NH, United StatesIn order to carry out an accurate diagnosis, prognosis, and/ortherapeutic assessment for a disease; high-throughput approaches toexamine the whole genome and transcriptome are now a necessity formodern research. Furthermore, to more fully understand the underlyingcauses <strong>of</strong> disease, high-throughput genomics are required to examinePoster Abstracts<strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine • 61


Poster Abstractsglobal gene expression, regulation, and interactions. To meet theseand the future needs <strong>of</strong> the research community at Dartmouth, thelatest technologies in deep sequencing and microarrays are <strong>of</strong>fered. Inaddition, we <strong>of</strong>fer the following services: Specialized, expensive, highendinstrumentation; Expert staffing; Cost-effective for individual labs;Competitive pricing and services with outside sources; On site NorrisCotton Cancer Center facility; Free experimental design consultations;Competitive fee for service charges for all high-throughput approaches;and Close proximity to Biostatistics and Bioinformatics shared resources.108 A Collaborative Life Cycle Process forthe Bioinformatics Core FacilityS. Lin, W.A. KibbeNorthwestern University, Evanston, IL, United StatesThe operation <strong>of</strong> the bioinformatics core facility is constantly challengedby increasing data volume, emerging technologies, and limited budget.We discuss a Collaborative Life Cycle (CLC) process as a businessmanagement model for this challenging environment. Unlike thetraditional involvement at the last stage <strong>of</strong> data analysis, the CLC processengages the bioinformatics core facility throughout the project with thePis and the wet lab core facilities. Various tasks <strong>of</strong> the bioinformaticscore during the project’s life cycle include: 1) Planning -study designand statistical power analysis with the PIs 2) Experiment-core wet labsample tracking and data management 3) Data analysis -data qualitycontrol and interpretation in the biological context. Collaborationsthroughout the project life cycle are critical because the multiple-phaseprocess involves numbers <strong>of</strong> pr<strong>of</strong>essional disciplines with many skills,tools, and procedures. The CLC process will help the bioinformaticscore facility align the multiple goals, adjust the expectations, mitigatethe potential risks, and improve the end results.109 Promoting Diversity in the CoreFacilityM. PersonThe University <strong>of</strong> Texas at Austin, Austin, TX,United StatesDiversity is encouraged to counter historical bias against a variety<strong>of</strong> groups and has emerged as a positive attribute <strong>of</strong> the workplace.However, among pressing economic and scientific outcome pressures,issues not tied to measurable rewards are <strong>of</strong>ten overlooked in the corefacility. This poster will present an overview <strong>of</strong> diversity issues andstrategies relevant to core facilities. The variety <strong>of</strong> types <strong>of</strong> diversityis discussed, including sex, race, ethnicity, nationality, socioeconomicbackground, and sexual orientation. Institutional efforts to encouragediversity are described with regards to organizational mandates andimplementation at the level <strong>of</strong> core facilities. Contrast between writtenpolicies and real world practices are noted. A valuable tool availableto the core facility director is self-examination, actively counteringbarriers that unconscious bias and learned prejudice have created.Systematic analysis can identify areas <strong>of</strong> diversity strength and weaknessin the facility. Strategies for increasing diversity are presented for thehiring process. Practices in the workplace that accommodate diversepopulations and foster career development are discussed. Finally,identifying rewards for diversity promotion encourages consistentprogress in the face <strong>of</strong> competing demands.110 A Web Based Tool for EfficientManagement and Use <strong>of</strong> ResearchCore FacilitiesP. Turpen, L. Wilkie, L. MillerUniversity <strong>of</strong> Nebraska Medical Center, Omaha,NE, United StatesThe need to efficiently and effectively manage research resourcesis an area <strong>of</strong> growing concern to both grantor agencies and granteeinstitutions. Central administration <strong>of</strong> core facilities is one way toaddress some <strong>of</strong> those concerns, but managing the needs <strong>of</strong> disparatecores can be a challenge. All research core facilities, irrespective <strong>of</strong>their emphasis, need to be able to contract for services, bill for thoseservices and provide documentation <strong>of</strong> facility use. While commercialsystems are a welcome recent development, most concentrate onsample management data, LIMS, with invoicing and utilization metricsas secondary functions. The shortcoming in these types <strong>of</strong> <strong>of</strong>ferings isthat they do not readily capture the institution’s or grantor agencies’needs for use reporting without considerable customization andcorresponding associated costs. We have chosen to create a web basedtool that can facilitate the scheduling/contracting <strong>of</strong> services, invoicingand fee collection for those services, and address the complex reportingneeds <strong>of</strong> the institution.111 XSQ: A New Binary Output Format <strong>of</strong>5500/5500XL SystemsC. Yang, J. Zhang, D. Thomas, P. SuriLife Technologies, Carlsbad, CA, United StatesThe XSQ (eXtensible SeQuence) format was designed to accommodatedifferent run types (standard SOLiD sequencing and exact chemistrycall (ECC) runs), simplify indexing samples workflow and support newdata types such as ECC in the new 5500 sequencing instrument; thesedata cannot be stored in any existing formats (e.g. csfasta & QV.qual) asthere are more than one call per position. Other problems with existingformats are the file size, I/O demand, and lack <strong>of</strong> pairing <strong>of</strong> reads. TheXSQ format uses information packing to reduce the file size by 60%,resulting in reduced storage needs and reduced transfer times. It alsohas integrated pairing <strong>of</strong> reads, so mapping and pairing can happentogether much more quickly, rather than having the two reads <strong>of</strong> a pairmapped separately and then merged subsequently. The new XSQ fileallows users to perform very efficient indexing reassignment and suchreassignment only introduces minimal impact to downstream analysisinstead <strong>of</strong> reanalyzing all indexing samples. The hierarchical data formatalso provides a basic level <strong>of</strong> partitioning and indexing within the file sothat subsets <strong>of</strong> the data can be retrieved without reading through theentire file.112 Micros<strong>of</strong>t Biology Initiative: .NETBioinformatics Platform and ToolsB. Diaz AcostaMicros<strong>of</strong>t Biology Initiative: .NET BioinformaticsPlatform and ToolsThe Micros<strong>of</strong>t Biology Initiative (MBI) is an effort in Micros<strong>of</strong>t Researchto bring new technology and tools to the area <strong>of</strong> bioinformatics andbiology. This initiative is comprised <strong>of</strong> two primary components,the Micros<strong>of</strong>t Biology Foundation (MBF) and the Micros<strong>of</strong>t BiologyTools (MBT). MBF is a language-neutral bioinformatics toolkit built62 • <strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine


as an extension to the Micros<strong>of</strong>t .NET Framework—initially aimed atthe area <strong>of</strong> Genomics research. Currently, it implements a range <strong>of</strong>parsers for common bioinformatics file formats; a range <strong>of</strong> algorithmsfor manipulating DNA, RNA, and protein sequences; and a set <strong>of</strong>connectors to biological web services such as NCBI BLAST. MBFis available under an open source license, and executables, sourcecode, demo applications, documentation and training materials arefreely downloadable from http://research.micros<strong>of</strong>t.com/bio. MBT is acollection <strong>of</strong> tools that enable biology and bioinformatics researchersto be more productive in making scientific discoveries113 NERLSCD: A Model for RegionalNetworking <strong>of</strong> Life Sciences CoreDirectorsG.S. Grills 1 , M. Detwiler 2 , T. Thannhauser 3 ,T. Hunter 4 , P.S. Adams 5 , S.A. Bobin 6 ,K. Sol-Church 7 , P. Spatrick 8 , S. Perkins 81Cornell University, Ithaca, NY, United States;2Roswell Park Cancer Institute, Buffalo, NY, UnitedStates; 3 United States Department <strong>of</strong> Agriculture/Agricultural Research Service, Beltsville, MD, UnitedStates; 4 University <strong>of</strong> Vermont, Burlington, VT,United States; 5 Trudeau Institute, Saranac Lake,NY, United States; 6 Dartmouth Medical School,Hanover, NH, United States; 7 Nemours COBRECenter for Pediatric Research, Wilmington, DE,United States; 8 University <strong>of</strong> Massachusetts MedicalSchool, Worcester, MA, United StatesThe Northeast Regional Life Sciences Core Directors (NERLSCD)annual meeting is a forum that provides an exceptional opportunityfor life sciences core facility directors and managers to network withcolleagues, to learn about biotechnology advances and applications,and to discuss the challenges and results <strong>of</strong> regional implementation <strong>of</strong>shared research resources. The NERLSCD meeting was established in2006 as a grass roots effort by core directors. The meeting was held lastyear at the University <strong>of</strong> Massachusetts Medical School in Worcester,MA. The sixth annual NERLSCD meeting will be held at CornellUniversity in Ithaca, NY, Nov. 9-11, <strong>2011</strong>. The goal <strong>of</strong> the meetingis to facilitate regional networking and sharing <strong>of</strong> resources and tohelp reduce regional duplication <strong>of</strong> costly biotechnology researchinfrastructure. The meeting provides a structured yet informal settingfor addressing the operational and technical challenges <strong>of</strong> life sciencescore laboratories. There are plenary sessions, workshops and discussionforums on financial and management issues facing biotechnology corelaboratories. There are technical workshops on genomics, functionalgenomics, proteomics, imaging, bioinformatics, bio-IT, and othertechnologies. A core facility poster session <strong>of</strong>fers an opportunity forlearning about regional life sciences shared resources and services.The number and diversity <strong>of</strong> attendees reflects the broad appeal andusefulness <strong>of</strong> this meeting for core directors at many institutions in theregion. The meeting supports a diverse array <strong>of</strong> core directors andmanagers who play a crucial role in facilitating advances in knowledgeand understanding in a wide variety <strong>of</strong> life sciences research areas. Thisregional networking meeting for life sciences core facility directorscan serve as an example for the organization, funding, structure, andcontent <strong>of</strong> regional meetings for core facility directors in other regions<strong>of</strong> the United States and in other countries.114 <strong>ABRF</strong> Affiliates and ChaptersS.F. Jennings 1 , G. Grills 2 , M. Detwiler 31University <strong>of</strong> Arkansas at Little Rock, Little Rock,AR, United States; 2 Cornell University, Ithaca,NY, United States; 3 Roswell Park Cancer Institute,Buffalo, NY, United StatesThe mission <strong>of</strong> the <strong>ABRF</strong> is to advance life sciences core facilities andbiotechnology laboratories through research, communication, andeducation. To facilitate this mission, the <strong>ABRF</strong> has implemented <strong>ABRF</strong>Affiliates and Chapters and the <strong>ABRF</strong> Affiliates and Chapters Committeefor the following purposes: a) To encourage the establishment, supportthe operations, and facilitate the coordination <strong>of</strong> new regional andspecial interest groups that have goals related to those <strong>of</strong> the <strong>ABRF</strong> insupport <strong>of</strong> life sciences shared resources; b) To establish partnershipsand collaborate with other existing organizations that have goals relatedto those <strong>of</strong> the <strong>ABRF</strong> in support <strong>of</strong> life sciences shared resources; c)To promote the technologies, research support and administration <strong>of</strong>biomolecular resource facilities; d) To promote the development andapplications <strong>of</strong> biotechnologies as shared research resources and t<strong>of</strong>acilitate the advancement <strong>of</strong> life sciences research; e) To play a leadershiprole in networking core laboratories, researchers, and students,matching those with similar and complementary interests and skills; e)To enhance communication on the regional, national and internationallevel regarding <strong>ABRF</strong> activities; to enhance the visibility <strong>of</strong> the <strong>ABRF</strong>in the scientific community; to educate the scientific community aboutthe value <strong>of</strong> the <strong>ABRF</strong>; and to broaden the number and diversity <strong>of</strong>core laboratories and biotechnology laboratories that take advantage<strong>of</strong> the <strong>ABRF</strong> research group studies and <strong>ABRF</strong> membership networkingopportunities; and f) To enhance the visibility <strong>of</strong> the <strong>ABRF</strong> with fundingagencies that support the development, acquisition and application<strong>of</strong> core facility shared research resources. <strong>ABRF</strong> Chapters are specialinterest groups which may be formed based on common interestsand/or geographical boundaries and support grassroots networks <strong>of</strong>individuals who wish to help advance <strong>ABRF</strong> goals and promote themission <strong>of</strong> shared resource facilities and biomolecular resources. <strong>ABRF</strong>Affiliates are special interest organizations that are autonomous fromthe <strong>ABRF</strong>, have common and complementary interests with the <strong>ABRF</strong>,and have the goal <strong>of</strong> developing a collaborative relationship with the<strong>ABRF</strong>. Please join us for the <strong>ABRF</strong> Affiliates and Chapters Open MicSession from 6:00 pm to 6:45 pm on Sunday.115 The Midwest <strong>Association</strong> <strong>of</strong> CoreDirectorsW. Hendrickson 1 , P. Hockberger 21University <strong>of</strong> Illinois, Chicago, IL, United States;2Northwestern University, Evanston, IL, UnitedStatesThe Midwest <strong>Association</strong> <strong>of</strong> Core Directors (MWACD) was organized in2010 by a group <strong>of</strong> scientists at 6 different institutions to foster closerinteractions among directors and managers <strong>of</strong> core facilities throughoutthe Central Plains. The organization shares the same goals as <strong>ABRF</strong>,and it has applied to become a chapter <strong>of</strong> <strong>ABRF</strong>. The MWACD differsfrom <strong>ABRF</strong> only in that its focus is on regional matters rather than onissues <strong>of</strong> national concern. Towards this goal, the first annual meeting<strong>of</strong> the MWACD took place on October 21-23, 2010, at the CrownePlaza Hotel in Chicago. The goal <strong>of</strong> the meeting was to provide anopportunity for networking among core directors and managers, toenable interactions with colleagues, sharing <strong>of</strong> technical advice, andPoster Abstracts<strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine • 63


Poster Abstractsdiscussions <strong>of</strong> continuing challenges associated with the operation <strong>of</strong>shared research resources and technologies. Keynote presentationswere delivered by leaders <strong>of</strong> NIH-NCRR and FASEB, and there werepanel discussions on networking, bioinformatics, and informationmanagement systems. There was a poster session, vendor exhibits, andbreakout sessions on 8 different core-related topics. The meeting wasattended by 120 researchers and supported by 17 corporate and notfor-pr<strong>of</strong>itorganizations.116 Automated Isolation <strong>of</strong> Genomic DNAfrom Large Volumes <strong>of</strong> Whole BloodE. Vincent 1 , S. Krueger 1 , J. Kennedy 2 , S. Lee 2 ,C. Helt 1 , A. Bonk 2 , C. Cowan 11Promega Corporation, Madison, WI, UnitedStates; 2 Hamilton Robotics, Reno, NV, United StatesA key source for genomic DNA(gDNA) is blood drawn into a standard10mL Vacutainer® tube. The Promega ReliaPrep Large Volume HTgDNA Isolation System integrated on the Hamilton MICROLAB®STARplus liquid handling workstation provides a unique anddependable system for isolating genomic DNA from large volumes(3 mL–10 mL) <strong>of</strong> blood. The novel chemistry and instrumentationresolve many challenges encountered when processing large-volumesamples in a high-throughput format such as: loss <strong>of</strong> sample pelletsduring decanting <strong>of</strong> fluids, transport <strong>of</strong> full 50 mL tubes to variouslocations on a liquid handling robot, and manual re-suspension <strong>of</strong>final DNA pellets. Liquid handler resource constraints were removedby creation<strong>of</strong> a new accessory, the ReliaPrep HSM 32 LV instrument,which provides heating, shaking and magnetization <strong>of</strong> samples atone deck position. The combination <strong>of</strong> this device, the MICROLABSTARplus workstation and the ReliaPrep Large Volume HT gDNAIsolation System allows automated recovery <strong>of</strong> pure gDNA from up to96 ten milliliter blood samples within 8 hours. We present verificationstudies demonstrating automated system performance. Comparisonsbetween the ReliaPrep Large Volume HT gDNA Isolation System anda standard precipitation-based method were made for duplicate bloodsamples from multiple donors. Yield, purity, and integrity <strong>of</strong> extractedgDNAwere assessed using UV absorbance spectroscopyand gelelectrophoresis. Genomic DNA yields from normal 10 mL whole bloodsamples were 200 400 µg (depending on white blood cell count) in aneluted volume <strong>of</strong> 1mL. Recovered DNA exhibited good purity withA260/A280 ratios greater than 1.7 and A260/A230 ratios between 1.8and 2.2. Isolated DNA was suitable for storage and was used in manydownstream analysis applications. Results <strong>of</strong> genomic DNA purificationfrom frozen (hemolysed) blood samples and blood collected usingcommon anticoagulants (EDTA, heparin, citrate)are also comparedtodemonstrate the efficacy <strong>of</strong> the new system.117 Overview <strong>of</strong> the Agilent TechnologiesSureSelect TM Target EnrichmentSystemJ. Ong 1 , A. Giuffre 1 , S. Joshi 1 , H. Ravi 1 ,C. Pabón-Peña 2 , B. Novak 2 , M. Visitacion 2 ,M. Hamady 2 , F. Useche 2 , B. Arezi 3 , B. Buehler 3 ,E. Lin 2 , S. Hunt 2 , D. Roberts 2 , S. Happe 1 ,E. Leproust 21Agilent Technologies, Cedar Creek, TX, UnitedStates; 2 Agilent Technologies, Santa Clara, CA,United States; 3 Agilent Technologies, La Jolla, CA,United StatesNext-generation DNA sequencing has revolutionized the discovery<strong>of</strong> rare polymorphisms, structural variants, and novel transcripts. Tomeet the demand for fast, cost-effective, and accurate genome analysismethods from small scale studies to large sample cohorts, AgilentTechnologies has developed the SureSelectTM Target EnrichmentSystem. Available for the Illumina, SOLiD, and 454 NGS sequencingplatforms, SureSelect is a highly robust, customizable, and scalablesystem that focuses analyses on specific genomic loci by in-solutionhybrid capture. In addition, Agilent has introduced SureSelect XTfor Illumina and SOLiD, which combines gDNA prep, library prep,and SureSelect Target Enrichment reagents in one complete kit. BothSureSelect and SureSelect XT demonstrate high performance, asmeasured by capture efficiency, uniformity, reproducibility, and SNPdetection. We highlight the utility <strong>of</strong> the SureSelect system across awide range <strong>of</strong> target sizes and genome complexity using pre-designedcatalog libraries targeting cancer gene sets, sequences encoding thekinome, and both human and mouse All Exon content. In addition,user-defined custom content can be easily developed using theAgilent eArray s<strong>of</strong>tware with candidate variant coordinates as input.User-defined content can be manufactured on-demand as a customSureSelect kit, or combined with pre-defined Agilent catalog contentusing the Plus option. We propose a novel approach for variantdiscovery - using SureSelect catalog designs to uncover candidatevariants, followed by the design <strong>of</strong> smaller focused custom libraries forSNP validation and region pr<strong>of</strong>iling. By pooling many samples togetherper lane or slide, SureSelect multiplexing kits for Illumina and SOLiDenable validation across large sample cohorts with substantial costsavings. Accurate post target enrichment pooling is facilitated by theAgilent Bioanalyzer and QPCR NGS Library Quantification kits whichensure equal representation across samples. Further efficiencies arerealized using the Bravo Automated Liquid Handling Platform to meetthe need for parallel preparation <strong>of</strong> multiplexed libraries.118 Gene Synthesis: A Cost-EffectiveAlternative to Traditional MolecularCloningA. Liao, M. Schwartz, H. Lo, J. Zhou, P. YangGENEWIZ, Inc., South Plainfield, NJ, United StatesGene synthesis is the process <strong>of</strong> synthesizing a gene in vitro without theneed for initial template. Contrary to what many researchers’ beliefs,commercial gene synthesis service is quickly evolving to become a costeffectivealternative to traditional cloning and other molecular biologyprocedures. The main reasons include: 1) Time savings: Traditionalcloning involves a multi-step process that includes cloning strategydesign, primer synthesis, PCR, gel extraction, bacteria transformation,and other complex steps. This process requires considerable amount <strong>of</strong>64 • <strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine


time and human resource that gene synthesis does not. 2) Cost savings:In most cases, it costs less to order a synthetic gene than it does to orderoligos, cloning kits, and DNA sequencing services. 3) Enhanced DNAperformance: Gene synthesis allows for codon optimization whichhas been proven to increase the efficiency <strong>of</strong> protein expression. 4)Convenience: Without the need for a physical template and withoutdesign restrictions associated with the traditional cloning process, aresearcher can get a gene <strong>of</strong> his/her choice by simply supplying thenucleotide sequence or amino acid sequence. GENEWIZ is a globalCRO that provides a wide range <strong>of</strong> DNA services, including genesynthesis. GENEWIZ’s gene synthesis service features a 2-3 weekturnaround and expert technical and project management support.This informational poster will present case studies <strong>of</strong> how GENEWIZ’sgene synthesis service benefited researchers who had previously reliedon traditional molecular cloning for plasmid construction.119 Nucleotide-Level Variant Analysis <strong>of</strong>Next-Generation Sequencing DataUsing a Cloud-Based Data AnalysisPipelineB. Ganter, G. Asimenos, A. SundquistDNAnexus, Palo Alto, CA, United StatesTo demonstrate the flexibility <strong>of</strong> a cloud-based solution for analyzingdisparate sets <strong>of</strong> next-generation sequencing data, we looked at carefullychosen samples across different populations from the 1,000 GenomesProject (www.1000genomes.org) and conducted an extensive analysison two Chinese populations, the “Chinese in Beijing” (CHB) and the“Chinese in metropolitan Denver” (CHD), each consisting <strong>of</strong> 28 exomes.Each dataset was uploaded into the system using raw data files acquiredfrom the 1,000 Genomes Project. Using these data and a cloud-baseddata analysis pipeline, we performed a nucleotide-level variant analysiscombined with a population allele frequency analysis across all samplesfor the two populations . To identify alleles that are significantlydifferent across the two populations, a Pearson’s chi-square test wasapplied, which resulted in a total <strong>of</strong> 1.5 Mio SNPs, <strong>of</strong> which 84 werenonsynonymous with a p-value <strong>of</strong> less than 0.01. Interestingly, the genesassociated with nonsynonymous variants <strong>of</strong> the Chinese in metropolitanDenver population were enriched for biological annotations such asendocrine system disorder, metabolic disease, cardiac fibrosis, andinflammation (includes ZNF264, RPS6KA2, ROBO2, CRK, MUSK, CBL,CRK, and others). Furthermore, genes usually associated with liverinjury were also identified for this population, suggesting the liver isexposed to toxic agents more so in this population compared to theCHB population. The observed genomic differences in these twodifferent Chinese populations living in different parts <strong>of</strong> the world hinttowards a potential link between nutrition and different diseases (e.g.heart disease or metabolic diseases). Using this analysis as a case study,we will demonstrate how a scalable computational infrastructure canprovide researchers and sequencing service providers alike, a costeffectiveand secure cloud-based computing platform as a powerfuland collaborative technology solution for large scale sequence dataanalysis and management.120 Genome Technology Center at theNYU Langone Medical Center: NewSupport for Clinical and TranslationalScienceJ. Zavadil, S. MischeNew York University Langone Medical Center, NewYork, NY, United StatesTo significantly enhance support for clinical and translational researchwithin the framework <strong>of</strong> its CTSI, the NYU Langone Medical Centerconsolidated the Microarray and DNA Sequencing Cores into a newGenome Technology Center, a shared resource overseen by the Officefor Collaborative Science. The GTC’s team <strong>of</strong> 4 technical personneland one faculty level director assists >120 NYULMC laboratories intheir basic, clinical and translational research. The Sequencing Unitoperates 2 Illumina GAIIs, and a HiSeq sequencer will be added in Q1<strong>2011</strong>. The GAII capacity is applied to research applications (ChIP-seq,small-RNA-seq and RIP-seq) and to identification <strong>of</strong> disease-relatedgenome-level structure changes and correlates (e.g. RNA-seq <strong>of</strong> cancertranscriptomes). GTC also has a Roche GS FLX System (454) used forde novo sequencing <strong>of</strong> microbial species and for amplicon sequencingin clinical genetics, patient microbiome diversity, etc. The MicroarrayUnit operates Affymetrix GeneChip system and high-capacity QPCR(ABI 7900HT) with automated plate setup and loading for gene andmicroRNA pr<strong>of</strong>iling and for SNP genotyping in clinical genetics. TheGTC cooperates closely with the newly established Center for HealthInformatics and Bioinformatics (CHIBI) supported by the NIH/NCRRCTSA Award. CHIBI provides an HPC facility for sequencing andmicroarray data storage and <strong>of</strong>fers a full range <strong>of</strong> informatics services.The GTC is committed to regional and nationwide collaborations withother Cores. GTC participates in the activities <strong>of</strong> the Genomic Analysisand Technology Excellence (GATE) Working Group <strong>of</strong> the Academy forMedical Development and Collaboration (AMDeC), particularly in thesections <strong>of</strong> Core Facility Directors, Funding Strategy and Bioinformatics.It also contributes to the AMDeC Facilities Instrumentation ResourcesServices Technologies (FIRST), a real-time online database <strong>of</strong>biomedical research technology and resources available in the NewYork City area and throughout Northeastern US. Key services <strong>of</strong> theGTC are <strong>of</strong>fered to external clients.121 Integrated Core Facility Support andOptimization <strong>of</strong> Next GenerationSequencing TechnologiesG. Grills, P. Schweitzer, Q. Sun, J. Pillardy,W. Wang, T. Stelick, R. Bukowski, L. Ponnala ,J. VanEeCornell University, Ithaca, NY, United StatesNew DNA sequencing technologies present an exceptional opportunityfor novel and creative applications with the potential for breakthroughdiscoveries. To support such research efforts, the Cornell UniversityLife Sciences Core Laboratories Center has implemented the IlluminaHiSeq 2000 and the Roche 454 GS FLX platforms as academic corefacility shared research resources. We have established sample handlingmethods, LIMS tools and BioHPC informatics analysis pipelines insupport <strong>of</strong> these new technologies. Our genomics core laboratory,in collaboration with our epigenomics core and bioinformatics core,provides sample preparation and data generation services andboth project consultation and analysis support for a wide range <strong>of</strong>Poster Abstracts<strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine • 65


Poster Abstractspossible applications, including de novo or reference based genomeassembly, detection <strong>of</strong> genetic variation, transcriptome sequencing,small RNA pr<strong>of</strong>iling, and genome-wide epigenomic measurements <strong>of</strong>methylation and protein-nucleic acid interactions. Implementation <strong>of</strong>next generation sequencing platforms as shared resources with multidisciplinarycore facility support enables cost effective access andbroad based use <strong>of</strong> these technologies.122 Better Outcomes for Our Patients:Moving Science Forward in the<strong>Biomolecular</strong> Core Laboratory at theNemours Center <strong>of</strong> Pediatric ResearchJ. Holbrook, D.L. Stabley, E.C. Hitchens, P. Geller,K. Sol-ChurchNemours/A.I. duPont Hospital for Children,Wilmington, DE, United StatesNemours Biomedical Research has a long-standing commitment toscholarly and scientific endeavors directed towards improving thediagnosis and treatment <strong>of</strong> pediatric medical conditions. Establishedin 2004 with the support <strong>of</strong> a COBRE grant from the NIH s NationalCenter for Research Resources, the Nemours Center for PediatricResearch provides clinical and translational researchers with the tools aswell as training they need to perform their research. The <strong>Biomolecular</strong>Core Laboratory (BCL), located at the Alfred I. duPont Hospital forChildren in Wilmington, Delaware, is supported by the Center <strong>of</strong>Pediatric Research. We provide a unique opportunity for Nemoursclinicians, research staff, and affiliates at University <strong>of</strong> Delaware andThomas Jefferson University to develop competitive research projectsin genetics as well as provide essential services in molecular biologyand genomics. The mission <strong>of</strong> the BCL is to facilitate, through state-<strong>of</strong>the-artsservices and stewardship, discoveries that begin at a molecularlevel and move rapidly from patient-oriented research to the bedside.Working in close collaboration with those engaged in research activities,our goal is to enable fast, effective and productive top quality servicesas well as to match BCL s strengths with our clients needs. This posterhighlights the services the <strong>Biomolecular</strong> Core lab <strong>of</strong>fers, includingmolecular biology training, a listing <strong>of</strong> our instrumentation, and corebusinesses including DNA sequencing, genotyping, gene expressionand methylation analysis. We will also highlight our newly establishedmicroarray service utilizing the Affymetrix Microarray platform. Thedevelopment <strong>of</strong> this vital core allowed investigators, who previouslyused extramural services and technologies, to move those projects inhouse, thus saving both time and money for our clients.123 Critical Reagent AnalyticalCharacterization <strong>Program</strong> atGenentechS. Chamberlain, C. Williams, A. Meier, C. Lu,P. MotchnikGenentech, Inc., South San Francisco, CA, UnitedStatesAt Genentech, Critical Reagents are internally prepared, recombinantlyproducedproteins used in GLP/GMP assays in support <strong>of</strong> regulatoryfilings. These proteins are typically extracellular domain constructions,growth factors, antibodies and novel hybrid constructions producedin CHO transient or stable cell lines, E. coli or Baculovirus. Genentech’sCritical Reagent System (CritRS) is used to document the process<strong>of</strong> ordering, manufacturing, characterization, and delivering acritical reagent. The CritRS was put in place in 2004 at Genentechto ensure quality, consistency, and traceability <strong>of</strong> critical reagentpreparations. Critical reagent analytical characterization is performedusing four platform assays to assess the molecule’s identity andpurity/heterogeneity. Identity is confirmed using Edman N-terminalsequencing and MALDI-TOF peptide mass fingerprinting. Purity/heterogeneity is assessed by size exclusion chromatography (SEC)and SDS-PAGE. The four platform assays provide sufficient identityand purity information for most critical reagents. However, in somecases additional characterization methods have to be used due tounexpected results, such as observed N-terminal sequence is differentfrom expected sequence, more than one N-terminal sequences arepresent, SDS PAGE shows unexpected band pattern, and SEC can notseparate the high and low molecular species <strong>of</strong> the reagent. In thisposter we’ll present typical analytical characterization results as well assome unusual findings.124 Progress towards Becoming a CLIA-Certified Core FacilityM. McMillan 1 , M. Gray 1 , M. Sandoval 1 ,S. McCann 1 , S. Gorda 21University <strong>of</strong> Southern California, Norris CancerCenter/Keck School <strong>of</strong> Medicine, Los Angeles, CA,United States; 2 University <strong>of</strong> Southern California ,College <strong>of</strong> Letters, Arts and Sciences, Los Angeles.CA, United StatesThe USC Norris DNA Core Facility (formerly known as the MicrochemicalCore) was created in 1984. It is equipped with an AB3730 DNAsequencer and an AB3900 oligonucleotide synthesizer. It is affiliatedto the Molecular Genomics Core in the Epigenome Center. Our goal isto establish the DNA Core Facility as a CLIA-certified laboratory. Therationale is that for Norris Cancer Center clinicians to use our DNAsequencing data in the diagnosis, treatment and prognosis <strong>of</strong> cancerpatients, the facility must be CLIA (Clinical Laboratory ImprovementAmendments)-certified. We will present our progress towards this goalincluding qualified personnel, identifying genes <strong>of</strong> medical interest,the establishment <strong>of</strong> appropriate records and the purchase <strong>of</strong> CLIAcompliantequipment. We plan to file for CAP (College <strong>of</strong> AmericanPathologists) accreditation and apply for a Clinical Laboratory License(State) as well as submitting a CLIA application (Federal) to theCalifornia Department <strong>of</strong> Health Services.125 Trudeau Institute Molecular BiologyCore FacilityP. Adams, A.M. Moquin, J.J. H<strong>of</strong>fmanTrudeau Institute, Saranac Lake, NY, United StatesCentralization and merging <strong>of</strong> core facilities is a favorite topic in thecurrent economy. The Molecular Biology Core Facility (MBCF) at theTrudeau Institute has provided a variety <strong>of</strong> services to the investigatorsat the institute while recovering the majority <strong>of</strong> its expenses since itsinception 12 years ago. The MBCF survives by <strong>of</strong>fering an assortment<strong>of</strong> services that are specifically tailored to the Institute missionand by personalizing each service to the individual investigator.Shared resource equipment and work areas optimize space andinstrumentation. Chargeback fees are designed to recover costsaccording to OMB Circular A-21. The MBCF recovers ~ 80% <strong>of</strong> costs.Services provided include DNA sequencing, spectratyping <strong>of</strong> the T-cellrepertoire (fragment analysis), production, purification and labeling66 • <strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine


<strong>of</strong> Major Histocompatibility (MHC) Class I and Class II tetramers forFluorescent Activated Cell Sorter (FACS) analysis, RNA and Proteinanalysis using “Lab on a Chip” technology, real-time PCR measurement<strong>of</strong> gene expression and viral loads, knock-out mouse and Mycoplasmascreening, DNA haptenation and recombinant protein expression/purification for antibody detection, monoclonal and polyclonalantibody production and in vivo immunization. A variety <strong>of</strong> simpleservices such as primer design, primer ordering, stock primers, peptideordering and Taq production save the investigators time, effort andmoney. Education and training are provided for all techniques andfor using shared MBCF instrumentation, such as spectrophotometers,real-time PCR equipment, tissue prep and image capture/analysisequipment. Not only does the MBCF provide the service, the personnelassist in the planning and analysis to maximize proper usage <strong>of</strong> thetechnique. Services not available in the MBCF are outsourced to otherCore facilities in the area to be sure the quality <strong>of</strong> service is the highest.The MBCF at Trudeau Institute survives through versatility, imaginationand customization.126 Molecular Resource Facility, UMDNJ-New Jersey Medical SchoolR. Donnelly, S. Kuppasani, K. DhawanUniversity <strong>of</strong> Medicine & Dentistry <strong>of</strong> New Jersey,New Jersey Medical School, Newark, NJ, UnitedStatesThe Molecular Resource Facility <strong>of</strong> the New Jersey Medical School wasestablished in April 1995 to enhance the resources available to theresearch community within the medical school. It serves to provideservices to the research community and as a source <strong>of</strong> informationon molecular techniques and research strategies involving molecularbiology. Our facility has developed through the years to be an “allinclusive” nucleic acid facility with limited protein analysis capabilities.We provide services in many areas including DNA sequencing, proteinsequencing, peptide synthesis, qPCR and others. In 2009 we began<strong>of</strong>fering High Throughput DNA Sequencing on the SOLiD® instrument.Our services are available to any laboratory requiring these services.127 United States Department <strong>of</strong>Agriculture/Agricultural ResearchService (USDA-ARS) Eastern RegionalResearch Center Core TechnologiesD. Needleman, A. Nuñez, G. Strahan, D.S. Soroka,W. DamertUnited States Department <strong>of</strong> Agriculture/Agricultural Research Service (USDA-ARS) EasternRegional Research Center Core Technologies,Wyndmoor, PA, United StatesUnited States Department <strong>of</strong> Agriculture. The CT unit is comprised<strong>of</strong> four research related components: genetic analysis, proteomicsbiopolymersmass spectrometry, electron microscopy, and magneticresonance spectroscopy (NMR). In addition, the Research Data Systems,the information pipeline <strong>of</strong> the CT, provides the means to facilitate datadistribution to researchers, stakeholders, and the general public. Theavailability <strong>of</strong> integrated resource laboratories assures pr<strong>of</strong>essional anddependable support to the goals <strong>of</strong> the ARS community.128 NYULMC Office <strong>of</strong> CollaborativeScience Cores - Enabling PersonalizedMedicine through TranslationalTesearchC. Curchoe, T. Winner, J. Salcedo, S. Mische,D. LevyNew York University, Langone Medical Center,New York, NY, United StatesThe New York University Langone Medical Center (NYULMC) hascommitted $15 million to ensure that researchers have access to cuttingedge enabling technologies. The Office <strong>of</strong> Collaborative Scienceenables access to expertise and technology through the centralizedadministration <strong>of</strong> Shared Resource Centers. In partnership with theNYU Cancer Institute, Center for AIDS Research and Clinical andTranslational Science Institute, we strive to increase collaboration amongclinical, translational and basic scientists to support novel science andto classify and treat diseases not just by their phenotype, but also bytheir molecular pr<strong>of</strong>iles. Here we highlight several Core Resource Labsthat support translational research at NYULMC. Human biospecimensare essential validating mechanistic insights into biological processesgleaned from cell lines, animal models <strong>of</strong> disease, and epidemiologicalstudies. The availability <strong>of</strong> well-annotated human biospecimensis a critical link between basic science and translational research.The NYULMC Tissue Acquisition and Biorepository Core providesinvestigators with freshly acquired frozen, normal, and diseased tissueat the time <strong>of</strong> surgery, as well as pathology-archived, formalin fixed,paraffin-embedded (FFPE) specimens, from appropriately consentedpatients. The Immunohistochemistry and Histopathology Coresclosely interface with the Tissue Biorepository, providing the meansfor clinical and basic research collaborations and the microscopicanalysis <strong>of</strong> clinical research specimens. The Genome TechnologyCenters sequencing and microarray units are dedicated to RNA-seq<strong>of</strong> cancer patient transciptomes, array expression pr<strong>of</strong>iling and SNPgenotyping, enabling translation research as well as the preparation <strong>of</strong>nucleic acids for molecular pr<strong>of</strong>iling, mutational analysis and microRNAscreens. The RNAi Core provides an integrated, state-<strong>of</strong>-the-art, RNAinterference (RNAi) based high-throughput screening (HTS) and<strong>of</strong>fers siRNA/dsRNA screening libraries for the cross-species functionalcharacterization <strong>of</strong> whole genomes in a systematic, comprehensive, andcost effective manner.Poster AbstractsThe Core Technologies (CT) unit, located at the Eastern RegionalResearch Center (ERRC), is a centralized resource <strong>of</strong> specializedinstrumentation and technologies. Its objective is to providesupplementary research data processing, interpretation, analysis andconsultation for a broad range <strong>of</strong> research programs approved by theAgricultural Research Service (ARS), the in-house research arm <strong>of</strong> the<strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine • 67


Poster Abstracts129 Bravo Automated Liquid HandlingPlatform for SureSelect TargetEnrichmentA. Giuffre 1 , M. Visitacion 2 , J. Karbowski 3 ,B. Novak 2 , H. Ravi 1 , J. Ong 1 , S. Joshi 1 ,C. Pabón-Peña 2 , E. LeProust 2 , D. Roberts 2 ,S. Happe 11Agilent Technologies, Genomics, Cedar Creek, TX,United States; 2 Agilent Technologies, GenomicsDivision, Santa Clara, CA, United States; 3 AgilentTechnologies, Automation Solutions Division, SantaClara, CA, United StatesAgilent’s SureSelect Target Enrichment products have met the need fora robust and cost-effective approach for systematic resequencing <strong>of</strong>candidate regions in the human genome and other species. SureSelectis an in-solution method <strong>of</strong> targeting only the user defined regions<strong>of</strong> interest <strong>of</strong> a genome to identify genetic variants and mutationsassociated with disease.With the advent <strong>of</strong> SureSelect Target EnrichmentMultiplexing kits, the ability to achieve targeted enrichment on multiplesamples in a single sequencing lane is maximized, saving time and moneywithout sacrificing performance. However, the handling <strong>of</strong> numeroussamples can be cumbersome and the possibility for the introduction <strong>of</strong>sample processing errors greatly increases. Agilent has expanded theSureSelect Target Enrichment System to allow for higher throughput <strong>of</strong>numerous samples with the Bravo Automated Liquid Handling Platform.The SureSelect method selects user defined target regions <strong>of</strong> interestfrom SureSelect XT prepped genomic DNA libraries by hybridizationto in-solution biotinylated cRNA probes; these enriched libraries arethen compatible for sequencing using the llumina GAII and HiSeqplatforms. The Agilent automation solution for SureSelect enablesprocessing <strong>of</strong> up to 96 samples per run in much less time than canbe obtained manually. Our SureSelect automation workflow includesSureSelect XT genomic DNA library construction, hybridization setup,and automated capture and wash steps. Each <strong>of</strong> these steps can berun with minimal user intervention and provide more uniformity thanmanual methods. We demonstrate here the throughput potential andconsistency in performance across entire 96-well plates <strong>of</strong> multiplexedsamples. In summary, SureSelect Automation solution provides an easyto use, automated gDNA library preparation and target enrichmentsystem that is a cost-effective approach to analyzing discrete genomicregions with unprecedented depth, accuracy and throughput.130 Modification <strong>of</strong> the Transplex WTA2Amplification Product for NextGeneration SequencingK. Heuermann 1 , B. Ward 1 , D. Fenoglio 21Biotech R&D, Sigma Aldrich Corporation, St. Louis,MO, United States; 2 Biotech Marketing, Sigma-Aldrich Corporation, St. Louis, MO, United StatesTransplex Whole Transcriptome Amplification (WTA2)ä exponentiallyamplifies RNA producing a double-stranded cDNA library whileprecisely maintaining differential levels <strong>of</strong> individual transcripts in testand reference samples. Though originally designed to amplify nanogramquantities <strong>of</strong> RNA, Transplex WTA2 has been shown to be exceedinglyeffective for amplification from damaged RNA template (FFPE and lasercaptured tissue samples) and single-cell input quantities (picograms).The efficacy <strong>of</strong> Transplex WTA2 amplification for downstreamapplications, primarily qPCR and expression microarray analysis, iswell-documented. It follows that the utilization <strong>of</strong> next-generationsequencing for gene expression research and diagnostics would bewell served by Transplex amplification <strong>of</strong> RNA isolated from samples <strong>of</strong>severely restricted quantity or quality. Strategies for the integration <strong>of</strong>Transplex WTA2 with next-generation sequencing are examined, withparticular emphasis on elimination <strong>of</strong> the characteristic fixed primersequence associated with each amplicon in the amplification library.Removal <strong>of</strong> these sites will allow direct entry <strong>of</strong> the resulting productinto the sequencing workflow. Methods under consideration will enablethe WTA2 amplicon to feed into the current sample prep protocols forthe Illumina GA and GAII, SoLiD 5500/5500xl, and Roche-454 GS FLX/Junior platforms.131 Robotic Scripts, Methods, Reagents,and Devices for High ThroughputAutomated Production <strong>of</strong> NextGeneration Sequencing DNAFragment LibrariesJ. Bishop, M. Allen, K. Poulter, W. Zhang,M. Landers, D. Mandelman, B. Laubert, A. Harris,R. BennettLife Technologies, Carlsbad, CA, United StatesRecent technological advances have greatly increased both the speedand throughput <strong>of</strong> genome and transcriptome sequencing. The pace<strong>of</strong> sequencing has further increased with target-enrichment andlibrary barcoding techniques that allow multiplexing <strong>of</strong> sequencingruns. However, current manual methods for creating libraries do notscale well, limiting the practical investigation <strong>of</strong> large numbers <strong>of</strong>samples. To ease the library-creation bottleneck, we describe here aset <strong>of</strong> protocols, robotics scripts, bulk reagents and instrumentationdeveloped to automate the production <strong>of</strong> up to 96 DNA fragmentsequencing libraries at once. Current protocols for creating sequencinglibraries are lengthy, laborious, and not amenable to automation.We describe here new magnetic bead based methods that producelibraries with yield, purity, and size-selection comparable or superiorto current column and gel-based protocols. Using these bead-basedmethods we developed a unique combination <strong>of</strong> optimized adaptorconcentrations and clean-up techniques which increase the yield <strong>of</strong>libraries from small amounts <strong>of</strong> DNA by several fold. We also describerobotic scripts for producing 1-96 libraries simultaneously on twocommonly used robotic platforms. These protocols accept 10-8000 ng<strong>of</strong> sheared DNA, calculate and dilute barcode adaptors for each libraryas necessary, automate all intermediate processing steps, and deliverpurified libraries ready for amplification <strong>of</strong>f-instrument. An additionalscript for post-PCR purification <strong>of</strong> the libraries is also provided. Wefurther describe the Library BuilderTM System, comprising a benchtopdevice and kits <strong>of</strong> plastic tips, tubes and sealed cartridges prefilledwith reagents necessary for producing 1 to 13 DNA fragment librarieseither with or without size selection. Our analysis <strong>of</strong> sequencing datashows that libraries produced by all <strong>of</strong> the above methods are free <strong>of</strong>excess adaptors which interfere with quantitation, are unbiased, <strong>of</strong> highcomplexity, and free from cross-contamination. The protocols hereinare provided to the community for use or customization.68 • <strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine


132 Improvements in SOLiD TM WholeTranscriptome Library PreparationWorkflow to Enable Low Input RNAAmountsC. San Jose HinahonLife Technologies, Austin, TX, United StatesThe SOLiD TM Total RNA-Seq (STaR-Seq) kit from Life Technologiesprovides a complete workflow for generating directional, randomprimed, whole transcriptome libraries from total RNA and fractionatedRNA. Poly(A) selected RNA is commonly used for expression pr<strong>of</strong>ilingusing short read sequencing on the SOLiD TM instrument. The inputrequirements in the current STaR-Seq protocol is 100-500ng poly(A)selected RNA, input amounts well suited for studies using cell cultureor large tissues. However, in circumstances where the available sampleis limited, such as in the case <strong>of</strong> clinically derived material or the needto focus the analysis on specific cell populations, there is <strong>of</strong>ten too littleRNA available to perform the cDNA library analysis. Improvements tothe STaR-Seq workflow have been developed and validated pushingthe starting input to 5ng <strong>of</strong> poly(A) selected RNA, while still maintaininghigh concordance with the current method. Improvements includeoptimizations in: 1. Reducing RNA fragmentation time to increase RNAfragments within the suitable size range; 2. Altering purification methodsafter RNA fragmentation to minimize sample loss; 3. Adoption <strong>of</strong> abead purification and size selection method to reduce sample loss dueto gel size selection; 4. Modifying PCR conditions to maximize cDNAlibrary yields without biasing expression pr<strong>of</strong>iles. These improvementsenable STaR-Seq to be a viable option for samples with small quantities<strong>of</strong> total RNA. As the field <strong>of</strong> next generation sequencing advances, it isnecessary to meet the need for decreased sample requirements, whilemaintaining strand specificity, and accuracy.133 RNA-Seq Analysis with NextGENeS<strong>of</strong>twareJ. McGuigan, Y. You, C. LiuS<strong>of</strong>tGenetics, LLC, State College, PA, United StatesRNA sequencing is a powerful tool for interrogating the entiretranscriptome at once. It allows identification <strong>of</strong> novel is<strong>of</strong>ormsincludinggene fusions and alternative splicing- and expression levelanalysis across several orders <strong>of</strong> magnitude. NextGENe’s new RNA-seqapplication can be used to analyze data from any <strong>of</strong> the three main2nd generation sequencing systems including Roche GS FLXTM andFLX Titanium, Illumina Genome Analyzers, and Applied BiosystemsSOLiDTM Systems. Variant detection, expression level analysis, andis<strong>of</strong>orm identification are all performed in one analysis. The tool employsa hybrid approach, combining the use <strong>of</strong> an exon junction referencesequence with the de novo detection <strong>of</strong> exons. This allows discovery <strong>of</strong>previously un-annotated genes in addition to highly accurate alignmentto known transcripts. All detected transcripts are reported in additionto the information provided by NextGENe’s variant and expressionreports. Results are shown in the NextGENe Viewer which provides ahigh level <strong>of</strong> visualization for review and editing.134 GeneMarker® Genotyping S<strong>of</strong>tware:Tools to Increase the Statistical Power<strong>of</strong> DNA Fragment AnalysisC.S. Johathan Liu, D. Hulce, X. Li, T. Snyder-LeibyS<strong>of</strong>tGenetics, LLC, State College, PA, United StatesThe discriminatory power <strong>of</strong> post-genotyping analyses, such askinship or clustering analysis, is dependent on the amount <strong>of</strong> geneticinformation obtained from the DNA fragment/genotyping analysis. Thenumber <strong>of</strong> microsatellite loci amplified in one multiplex is limited by thenumber <strong>of</strong> dyes and overlapping loci boundaries; requiring researchersto amplify replicate samples with 2 or more multiplexes in orderto obtain a genotype for 12-15 loci. AFLP is another method that islimited by the number <strong>of</strong> dyes, <strong>of</strong>ten requiring multiple amplifications<strong>of</strong> replicate samples to obtain more complete results. Traditionally,researchers export the genotyping results into a spread sheet, manuallycombine the results for each individual and then import into a thirds<strong>of</strong>tware package for post-genotyping analysis. GeneMarker is highlyaccurate, user-friendly genotyping s<strong>of</strong>tware that allows all <strong>of</strong> these stepsto be done in one s<strong>of</strong>tware package, avoiding potential errors fromdata transfer to different programs and decreasing the amount <strong>of</strong> timeneeded to process the results. The Merge Project tool automaticallycombines the results from replicate samples processed with differentprimer sets. Replicate animal (diploid) DNA samples were amplifiedwith three different multiplexes, each multiplex provided informationon 4 -6 loci. The kinship analysis using the merged results provideda 1017 increase in statistical power with a range <strong>of</strong> 108 when 5 lociwere used versus 1025 when 15 loci were used to determine potentialrelationship levels with identity by descent calculations. These samesample sets were used in clustering analysis to diagram dendrograms.The dendrogram based on a single multiplex resulted in three branchesat a given Euclidian distance. In comparison, the dendrogram that wasconstructed using the merged results had eight branches at the sameEuclidian distance.135 Hotspot-Mutation Analysis <strong>of</strong> theEGFR/KRAS/BRAF Pathway UsingMutation Surveyor® S<strong>of</strong>twareJ. Liu, D. Hulce, K. LeVan, N. ShouyongS<strong>of</strong>tGenetics, LLC, State College, PA, United StatesHotspot-mutation analysis <strong>of</strong> the EGFR/KRAS/BRAF pathway (orother clinically relevant pathway) can quickly genotype patients ascandidates who may respond favorably to specific drug treatmentsand therapies or into other groups where treatment options arelimited and less favorable. Sanger sequencing analysis using MutationSurveyor s<strong>of</strong>tware provides high-throughput, high-sensitivity variationdetection. Increased efficiency can be achieved using flexible andcustomizable reporting-sequencing results can be organized bypatient identifiers, variation type (reported or unreported, pathogenicor benign or drug sensitive), by gene/exon/amplicon, or qualitymetrics, and other options. GenBank sequence files from NCBI forEGFR exons 18, 19, 20, and 21; KRAS exons 2 and 3; and BRAF exon15 were edited to contain reported variations. These reportedvariations included polymorphisms from dbSNP (downloaded withthe GenBank file), pathogenic and drug-sensitivity variations for EGFR(obtained from http://www.egfr.org/), activating mutations for KRAS,and constitutive mutations for BRAF. Bidirectional sequencing datafor twelve, simulated (mutations obtained from sequencing reportsin the scientific literature), patients were developed and comparedPoster Abstracts<strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine • 69


distribution. Additionally, we show that the controls can be used toevaluate performance <strong>of</strong> various normalization and scaling methodsfor RNA-Seq count data. This analysis suggests that traditional scalingmethods based on the total number <strong>of</strong> reads result in a significant loss<strong>of</strong> accuracy and more robust methods are <strong>of</strong>ten required. This analysisreveals the importance <strong>of</strong> using standardized reagents and controls as aroutine part <strong>of</strong> the RNA sequencing workflow.139 S<strong>of</strong>tware Systems for ClinicalResearchT. Smith 1 , N.E. Olson 1 , D. Smith 2 , C. Mason 31Geospiza, Inc. Seattle, WA, United States; 2 MayoClinic, Rochester, MN, United States; 3 Weil CornellMedical College, New York, NY, United StatesBy the end <strong>of</strong> <strong>2011</strong> we will likely know the DNA sequences for 30,000human genomes. However, to truly understand how the variationbetween these genomes affect phenotype at a molecular level, futureresearch projects need to analyze these genomes in conjunction withdata from multiple ultra-high throughput assays obtained from largesample populations. In cancer research, for example, studies thatexamine 1000s <strong>of</strong> specific tumors in 1000s <strong>of</strong> patients are neededto fully characterize the more than 10,000 types and subtypes <strong>of</strong>cancer and develop diagnostic biomarkers. These studies will usehigh throughput DNA sequencing to characterize tumor genomes andtheir transcriptomes. Sequencing results will be validated with nonsequencingtechnologies and putative biomarkers will be examined inlarge populations using rapid targeted assay approaches. Geospiza istransforming the above scenario from vision into reality in several ways.The Company’s GeneSifter platform utilizes scalable data managementtechnologies based on open-source HDF5 and BioHDF technologiesto capture, integrate, and mine raw data and analysis results from DNA,RNA, and other high-throughput assays. Analysis results are integratedand linked to multiple repositories <strong>of</strong> information that includevariation, expression, pathway, and ontology databases to enablediscovery process and support verification assays. Using this platformand RNA-Sequencing and Genomic DNA sequencing from matchedtumor/normal samples, we were able to characterize differential geneexpression, differential splicing, allele specific expression, RNA editing,somatic mutations and genomic rearrangements as well as validatethese observations in a set <strong>of</strong> patients with oral and other cancers.140 Hybrid-Core Computing for High-Throughput BioinformaticsG. VacekConvey Computer Corporation, Richardson, TX,United StatesAdvanced architectures can deliver dramatically increased throughputfor genomics and proteomics applications, reducing time-tocompletionin some cases from days to minutes. One such architecture,hybrid-core computing, marries a traditional x86 environment witha reconfigurable coprocessor, based on field programmable gatearray (FPGA) technology. Application-specific instructions executedby the coprocessor appear as extensions to the x86 instruction setarchitecture. This integrated approach provides users familiar C, C++and FORTRAN development environments without the complexity <strong>of</strong>non-standard dialects or programming models. Thus the performance<strong>of</strong> application-specific hardware is achievable with the familiarprogrammability and deployment <strong>of</strong> a commodity server. In addition tohigher throughput, increased performance can fundamentally improveresearch quality by allowing more accurate, previously impracticalapproaches. This presentation will discuss the suitability <strong>of</strong> hybridcoreservers’ advanced architecture and compiler technology forsequence alignment and assembly applications. For example, the Smith-Waterman alignment algorithm is 172 times faster on Convey’s HC-1than the best s<strong>of</strong>tware implementation on a commodity server. Suchperformance speeds research, while reducing energy consumption,floor space, and management effort. Most bioinformatics applicationsare similarly well suited for this architecture because they have lowdata interdependence, which greatly increases performance throughhardware parallelism. Furthermore, small data type operations (fournucleotides can be represented in two bits) make more efficient use <strong>of</strong>logic gates than the data types dictated by conventional programmingmodels. Bioinformatics applications that have random access patternsto large memory spaces, such as graph-based algorithms, experiencememory performance limitations on cache-based x86 servers. Convey’shighly parallel memory subsystem allows application-specific logic tosimultaneously accesses 8192 individual words in memory, significantlyincreasing effective memory bandwidth over cache-based memorysystems. Many algorithms, such as Velvet and other de Bruijn graphbased, short-read, de novo assemblers, greatly benefit from this type<strong>of</strong> memory architecture.141 Implementation <strong>of</strong> GeneSifterImproves Workflow Managementwithin a Core FacilityA. McCary, J. Boland, J. Bacior, V. LonsberryNational Cancer Institute, Bethesda, MD, UnitedStatesAs throughput within sequencing facilities increases, LaboratoryInformation Management Systems (LIMS) will become necessary toallow laboratory managers and staff to organize workflows, samplesand sample data in a repeatable and reliable manner. At the NCI’s CoreGenotyping Facility, we have successfully implemented Geospiza’sGeneSifter TM Lab Edition, a commercially available LIMS, for use with ourRoche 454 GS FLX sequencer processes. Our current setup consists <strong>of</strong>seven different sample preparation techniques, (six target enrichmentmethods and one whole genome method), which have been dividedinto a series <strong>of</strong> modular workflows that are arranged in the properorder for each process. Each workflow is then further broken down intospecific protocol steps, which are used as guides for the techniciansduring protocol performance. Additionally, mastermix preparations,reagent lot numbers, file uploads and other pertinent information areinput into the system at all relevant protocol steps. Each sample’s datais stored on our internal GeneSifter TM server, with the ability to recalland report it at a later date. Therefore, by developing an extensiveand interconnected setup <strong>of</strong> workflows in GeneSifter TM , the NCI’s CoreGenotyping Facility has successfully increased reliability in sampletracking while decreasing the need for paper- or Excel-base loggingmethods. This project has been funded in whole or part with federalfunds from the NCI, NIH under contract HHSN261200800001E.Poster Abstracts<strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine • 71


Poster Abstracts142 Comparison <strong>of</strong> Data EnvelopmentAnalysis Models to Identify PotentialCancer Biomarker Genes for ColonCancerP. Diaz-Candelas, M.E. Sanchez-Pena, C.E. Isaza,M. Cabrera-RiosBio IE Lab at University <strong>of</strong> Puerto Rico, Mayagüez,Puerto RicoThe identification <strong>of</strong> cancer biomarkers is critical to characterize,detect and understand the illness. In our research group, a novelmethod based on multiple criteria optimization has been proposedto detect potential cancer biomarker genes through the analysis <strong>of</strong>microarray data. More precisely, the multiple criteria optimizationproblem is approached through a technique called Data EnvelopmentAnalysis (DEA). Given a number <strong>of</strong> genes characterized by at least twoperformance measures, DEA is able to find those genes that are thebest compromises among these measures. When using microarray dataand quantifiers <strong>of</strong> significant differences <strong>of</strong> gene expression betweenhealthy tissues and cancer tissues, those genes identified through DEAhave been shown to have a high potential to be biomarkers. DEA isknown to be efficient due to its structure based on linear optimization.Our studies have also shown that DEA can be very effective for thestated purpose. There are, however, several different DEA formulationsthat can be used. In this work, a comparison <strong>of</strong> these formulations ispresented in terms <strong>of</strong> number and quality <strong>of</strong> the selection <strong>of</strong> genes fortwo publicly available colon cancer microarray databases. The resultswill provide some light on the different solutions that can be arrivedto in multiple criteria optimization using DEA, to then establish what ismost adequate to find cancer biomarkers.**143 Comparing Protein and mRNAAbundances to Protein ExpressionRegulationC. Vogel 1,2 , J.M. Laurent 2 , T. Kwon 2 , S.A. Craig 2 ,D.R. Boutz 2 , H.K. Huse 2 , K. Nozue 3 , H. Walia 3 ,M. Whiteley 2 , P.C. Ronald 3 , S. Abreu Rde 4 , D. Ko 4 ,S.Y. Le 5 , B.A. Shapiro 5 , S.C. Burns 4 , D. Sandhu 4 ,L.O. Penalva 4 , E.M. Marcotte 21New York University, New York, NY, United States;2University <strong>of</strong> Texas at Austin, Austin, TX, UnitedStates; 3 UC Davis, CA, United States; 4 University<strong>of</strong> Texas at San Antonio, San Antonio, TX, UnitedStates; 5 NCI-Frederick, Frederick, MD, United StatesTranscription, mRNA decay, translation and protein degradationare essential processes during eukaryotic gene expression, but theirrelative global contributions to steady-state protein concentrations inmulti-cellular eukaryotes are largely unknown. Using measurements<strong>of</strong> absolute protein and mRNA abundances in cellular lysate from thehuman Daoy medulloblastoma cell line, we quantitatively evaluate theimpact <strong>of</strong> mRNA concentration and sequence features implicated intranslation and protein degradation on protein expression. Sequencefeatures related to translation and protein degradation have an impactsimilar to that <strong>of</strong> mRNA abundance, and their combined contributionexplains two-thirds <strong>of</strong> protein abundance variation. mRNA sequencelengths, amino-acid properties, upstream open reading framesand secondary structures in the 5’ untranslated region (UTR) werethe strongest individual correlates <strong>of</strong> protein concentrations. In acombined model, characteristics <strong>of</strong> the coding region and the 3’UTRexplained a larger proportion <strong>of</strong> protein abundance variation thancharacteristics <strong>of</strong> the 5’UTR. Further, we used data from human and sixother organisms (bacteria, yeast, worm, fly, and plant) and establishedthat steady-state abundances <strong>of</strong> proteins show significantly highercorrelation across these diverse phylogenetic taxa than the abundances<strong>of</strong> their corresponding mRNAs (p=0.0008, paired Wilcoxon). Thesedata suggest strong selective pressure to maintain protein abundancesduring evolution, even when mRNA abundances diverge. The absoluteprotein and mRNA concentration measurements for >1000 humangenes and for other organisms represent one <strong>of</strong> the largest datasetscurrently available, and reveal both general trends and specificexamples <strong>of</strong> post-transcriptional regulation.144 The Application <strong>of</strong> Targetedand Exome Sequencing for theIdentification <strong>of</strong> Spontaneous andInduced Mutations in MiceD. Hinerfeld, E. Antoniou, S. Daigle, Y. Ding,W. Zhang, L. Reinholdt, M. Barter, L. RoweThe Jackson Laboratory, Bar Harbor, ME, UnitedStatesThe Jackson Laboratory has established a large collection <strong>of</strong>spontaneous and N-ethyl-N-nitrosourea (ENU) induced mousemutants with a wide variety <strong>of</strong> medically relevant phenotypes. Whilespontaneous mutations can be quite complex, including singlenucleotide polymorphisms (SNPs), transposon insertions, deletions, orinversions, ENU induced mutations are typically SNPs. The traditionalmethod <strong>of</strong> identifying the causative mutation through genetic mappingand Sanger sequencing <strong>of</strong> candidate genes has been effective but is timeconsuming, requires large populations <strong>of</strong> mice and can be expensive.In addition, it is particularly challenging when the mutation is not ina coding sequence. With a size <strong>of</strong> over 3 GB, it is still too expensiveto sequence the genomes <strong>of</strong> the many mutant strains <strong>of</strong> interest. Thecombination <strong>of</strong> array capture for targeted resequencing and/or exomecapture followed by high-throughput sequencing on the Illumina GAIIXhas greatly accelerated the pace at which mutations have be identified.In deciding what approach should be employed to identify a specificmutation, a number <strong>of</strong> factors must be considered; 1) Is the mutationspontaneous or ENU induced; 2) Have traditional mapping approachesbeen exploited, and if not, should they be; 3) If there is mapping data,what is the size <strong>of</strong> the genetic interval; 4) What data analysis tools will berequired; This approach has led to the identification <strong>of</strong> many mutationsthat result in disease-relevant phenotypes including crani<strong>of</strong>acialdisorders, neurodegeneration, neuromuscular dysfunctions, cholesterolbiosysthesis and reproduction.145 Single-Cell Copy Number AnalysisUsing the Illumina Genome AnalyzerE. Kamberov, J. Langmore, T. Kurihara,J. M’Mwirichia, T. Tesmer, D. OldfieldRubicon Genomics, Ann Arbor, MI, United StatesCNV, SNP, and mutation analyses <strong>of</strong> singles cells are important tocharacterize cancer, stem, and embryo cells. We used PicoPlex WholeGenome Amplification kits to sequence single human cancer cells,single-copy mouse chromosomes, and single bacterial cells using the72 • <strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine


Illumina Genome Analyzer. PicoPlex is a 1-tube, 3-hr, 4-step methodto convert a single cell into cluster-station ready DNA. High qualitysequences with very little background were obtained (0.7% mismatchrates, 90+% mapped reads, 98% concordance betweenSureSelect re-sequencing results and previously determined genotypeis observed. Lastly, we introduce the SureSelect XT kit for preparation<strong>of</strong> samples for multiplex sequencing using the Illumina GAII or HiSeq.The SureSelect Multiplexing kit provides the ability to combinetargeted enrichment with multiplexing, thus maximizing the number <strong>of</strong>samples that can be sequenced at one time, providing optimum timeand cost savings without sacrificing performance.148 Comparison <strong>of</strong> SYBR Enzymesand Standards in Illumina LibraryQuantificationK. Thai, S.S. LevineMIT BioMicro Center, Massachusetts Institute <strong>of</strong>Technology, Cambridge, MA, United StatesPoster AbstractsAccurate quantification <strong>of</strong> libraries generated for Illumina nextgenerationsequencing is critical to prevent under- and overclustering<strong>of</strong> the flowcell. Success <strong>of</strong> qPCR library quantifications depend onthe accuracy and reproducibility <strong>of</strong> standards used in addition to theability <strong>of</strong> DNA polymerase to efficiently amplify all adaptor-flankedlibraries. We compared the (i) Roche SYBR Green and (ii) KAPASYBR Fast polymerases and the PhiX standards and KAPA pre-dilutedstandards to determine the relative importance <strong>of</strong> these two factorsin library quantification by qPCR. We evaluated the success <strong>of</strong> eachmethod by comparing variation in the number <strong>of</strong> reads and absolute<strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine • 73


Poster Abstractsyield generated from sequencing. Our results indicate that the DNApolymerase from the KAPA SYBR Fast kit is better suited to libraryquantification applications while the Roche SYBR Green kit showedsignificant variability, possibly related to sample type or insert size.No differences were observed between the two types <strong>of</strong> standardsused, making DNA polymerase the determining factor in the successfulamplification <strong>of</strong> each library.**149 A New Sequencing Primer andWorkflow Increase 5’ Resolution andThroughput on HLA SequencingJ. Chuu 1 , S.C. Hung 1 , S. Berosik 1 , M. Wenz 1 ,S. Schneider 1 , P. Ma 1 , D. Berchanskiy 2 , D. Dinauer 21Life Technologies, Foster City, CA, United States;2Life Technologies, Brown Deer, WI, United StatesHigh quality and high accuracy are the hallmarks <strong>of</strong> Sanger resequencingprojects. We have developed a new sequencing primer andworkflow that improves 5’ sequence resolution, increases throughput,and reduces hands-on time. The novel sequencing primer chemistryproduces high quality bases from base 1 on POP-7TM polymer thatpreviously only could be resolved on the slower POP-6TM polymer.The new primer chemistry and workflow also eliminates the needfor a separate PCR clean-up step. These improvements reduce theentire workflow from PCR to finished sequence data to under 5hours, compared to 8 hours for the standard workflow. We used ourenhanced sequencing primer and workflow to investigate feasibilityon Human Leukocyte Antigen (HLA) polymorphisms on twelve DNAsamples by using the Invitrogen SeCore® HLA-DRB1 primer set andGroup Specific Sequencing Primers. Sequencing reactions generatedwith the traditional sequencing primer and with the new sequencingprimer were electrophoresed on Applied Biosystems 3500xl GeneticAnalyzer using POP-7TM polymer. For each sequencing primer, wecompared 5’ resolution and basecalling accuracy and quality. Onaverage the traditional primers produced high quality readablebases by base 25 after the sequencing primer while the new primersproduced high quality bases by base 5, and by base 1 in many cases.Because <strong>of</strong> improved resolution, basecalling accuracy was increased.This simplified process without a separate PCR clean-up step reducedthe overall workflow time by 40%. For HLA genes, obtaining readablesequence within 5 bases <strong>of</strong> the primer <strong>of</strong>fers improved polymorphismdetection and more efficient use <strong>of</strong> allele specific sequencing primersfor heterozygous ambiguity resolution. In conclusion, the novel primerchemistry and workflow generates data superior in quality relative toother currently used solutions and <strong>of</strong>fers significant time savings as well.Specific applications <strong>of</strong> this product are under development and notintended for clinical use.150 ScriptSeq RNA-Seq LibraryPreparation Method: A SimplifiedWork-Flow for Directional NGS RNA-Seq Library Preparation with Whole-Transcript RepresentationA. Khanna 1 , R. Sooknanan 2 , J. Hitchen 2 , A. Radek 21Epicentre BIOtechnologies, Madison, WI, UnitedStates; 2 RiboTherapeutics Inc., Saint Laurent, QC,CanadaRNA sequencing is an emerging revolutionary tool for wholetranscriptomeanalysis that provides information about the structure<strong>of</strong> transcripts and their expression levels. Current methods for makingsequencer-specific di-tagged DNA fragment libraries for RNA-Seqtypically comprise preparing rRNA-depleted RNAand either (i) RNAfragmentation, 5’ and 3’ adaptor-ligation, size selection, cDNA synthesis,and multiple clean-up steps; or (ii) cDNA synthesis followed by cDNAfragmentation, end-polishing, 5’ and 3’ adaptor-ligation, size selectionand multiple clean-up steps. These methods are generally long andrequire significant hands-on time. We describe a novel protocol thatutilizes a unique Terminal-Tagging technology that simplifies thepreparation <strong>of</strong> directional RNA-Seq libraries from rRNA-depleted orpoly(A)-enriched RNAin about 3 hours, without the need for adaptorligation, cDNA nebulization or gel purification. The di-tagged cDNAfragments that are generated from this simple, single tube protocolare compatible with different NGS sequencing platforms. Apart fromits simplicity, another major strength <strong>of</strong> this RNA-seq protocol is itsability to determine the polarity <strong>of</strong> the RNA transcripts, which is criticalfor the annotation <strong>of</strong> novel genes. Sequencing results obtained fromlibraries prepared by using RiboZeroTM rRNA-depletion method andScriptSeq mRNA-Seq Library Preparation Method show excellentdirectionality with less than 2 % <strong>of</strong> the sequence reads that map torRNA sequences (28S, 18S, 5.8S and 5S). This reduction in rRNAsequence reads improves sequence depth and coverage, and increasesthe percentage <strong>of</strong> uniquely mapped reads. Further, there is a highcorrelation (R2=0.9235) between differentially expressed transcriptsfound in the ScriptSeq RNA-Seq libraries and the MAQC QPCR panel<strong>of</strong> genes.151 The Agilent Technologies’SureSelect TM All Exon ProductPortfolio: High Performance TargetEnrichment System for Human andMouse Exome Sequencing on Illuminaand SOLiD PlatformsH. Ravi, A. Giuffre, C. Pabón-Peña, B. Novak,S. Joshi, J. Ong , M. Visitacion, M. Hamady,F. Useche, J. Eberle, S. Hunt, S. Happe, D. Roberts,E. LeproustAgilent Technologies, Stratagene Products Division,Cedar Creek, TX, United StatesThe dramatic increase in throughput <strong>of</strong> sequencing data from nextgenerationsequencing platforms has enabled scientists to study thegenome with unprecedented depth and accuracy. Nevertheless, routinegenetic screens in large numbers <strong>of</strong> individuals continue to remain costprohibitivethrough these approaches. Agilent Technologies’ SureSelectplatform for targeted exome capture, combined with massively parallelsequencing, provides a more affordable method to gain novel insightsinto the genetic causes <strong>of</strong> inherited disorders. In addition, identification<strong>of</strong> both common and rare polymorphisms implicated in complexdiseases like cancer is greatly facilitated by selectively sequencing theprotein-coding regions <strong>of</strong> the genome. In collaboration with the Broadand Sanger Institutes, Agilent Technologies has continued to expandthe number <strong>of</strong> SureSelect target enrichment catalog products in orderto enable a more comprehensive view <strong>of</strong> the protein-coding regionsin humans and model organisms. We discuss the SureSelectHuman AllExon v2 (44Mb) and SureSelectHuman All Exon 50Mb designs. We alsointroduce the SureSelectMouse All Exon target enrichment system,which improves the ability to study genetic variation between strains ingreater detail, and significantly increases the efficiency <strong>of</strong> screening forcausative mutations in N-ethyl-N-nitrosourea (ENU)-mutagenized mice.We demonstrate high performance with respect to capture efficiency,74 • <strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine


uniformity, reproducibility <strong>of</strong> enrichment, and ability to detect SNPs,insertion/deletions, and CNVs across Illumina (Genome Analyzer IIxand HiSeq2000) and SOLiD platforms. We highlight the utility <strong>of</strong> theSureSelect All Exon product portfolio for a wide variety <strong>of</strong> applicationsprimarily due to the high specificity and excellent cross-platformsequence coverage. SureSelect All Exon designs also provide a meansfor standardization, consistency <strong>of</strong> performance, and reliability acrossmultiple laboratories.152 PCR-Free Nextera Di-TaggedDNA Library Preparation for NGSApplicationsC. Kinross, H. Grunenwald, B. Baas, I. Goryshin,N. Caruccio, M. MaffittEpicentre Biotechnologies, Madison, WI, UnitedStatesThe Nextera TM technology for generating libraries <strong>of</strong> di-tagged DNAfragments is rapidly becoming the preferred method for massivelyparallel DNA sequencing. Despite rapid advances in sequencinginstrument throughput, classic library preparation by step-wise ligation<strong>of</strong> adaptors is a time-intensive and throughput-limiting bottleneck.PCR amplification <strong>of</strong> libraries prior to cluster generation is also amajor concern because <strong>of</strong> its possibility to reduce library complexity,particularly in regions <strong>of</strong> extreme G+C content (high or low), therebyproducing uneven genome coverage and confounding mapping andassembly. In this study, we describe novel modifications <strong>of</strong> the Nextera TMlibrary preparation system to address such library preparation biasby eliminating PCR amplification. Sequencer-ready libraries can beobtained from as little as 200 ng <strong>of</strong> genomic DNA in 3 hours with90-minutes <strong>of</strong> hands-on time. Deep sequencing <strong>of</strong> genomic librariesindicates that this system reduces coverage bias and GC bias, as well asimproves library diversity.153 Semiconductor Sequencing for LifeJ. Myers, J. RothbergIon Torrent, South San Francisco, CA, United StatesIon Torrent has invented the first device-a new semiconductor chipcapable<strong>of</strong> directly translating chemical signals into digital information.The first application <strong>of</strong> this technology is sequencing DNA. The deviceleverages decades <strong>of</strong> semiconductor technology advances, and in justa few years has brought the entire design, fabrication and supply chaininfrastructure <strong>of</strong> that industry-a trillion dollar investment-to bear on thechallenge <strong>of</strong> sequencing. The result is Ion semiconductor sequencing,the first commercial sequencing technology that does not use light, andas a result delivers unprecedented speed, scalability and low cost. All<strong>of</strong> these benefits are a result <strong>of</strong> applying a technology that is massivelyscalable, as proven by Moore’s Law, to a task that has traditionally usedoptics-based solutions, which work in a linear fashion: increasing capacityrequires increasing the number <strong>of</strong> signals that must be read resultingin longer run times, higher capital costs and ever more sophisticatedoptics. By contrast, Ion Torrent semiconductor technology can provideincreases in chip capacity without impacting capital costs or runtime.Ion Torrent sequencing uses only natural (label-free) reagents andtakes place in Ion semiconductor microchips that contain sensors whichhave been fabricated as individual electronic detectors, allowing onesequence read per sensor. We will show how the technology has scaledin just a few months from ~1 million sensors in the first-generationIon 314 chips to ~7 million sensors in the second-generation Ion 316chips-all while maintaining the same 1- to 2-hour runtime. We will alsodemonstrate that Ion semiconductor sequencing provides exceptionalaccuracy, long read length and scalability on a single, affordable benchtopsequencing platform.154 A Comparison <strong>of</strong> Post-DNASequencing Dye-Terminator RemovalProtocolsM. Zianni, A. McCoyThe Ohio State University, Columbus, OH, UnitedStatesCapillary electrophoresis, a method for separation <strong>of</strong> ions based upontheir size to charge ratio, remains in high demand for DNA sequencing.In the process <strong>of</strong> dideoxynucleotide terminator sequencing,unincorporated nucleotides and other contaminants remaining in thereaction mixture can cause multiple issues in the electropherograms,such as unincorporated dye peaks, missed base calls, decreased signalstrength or a complete lack <strong>of</strong> data as a result <strong>of</strong> blocked capillaries. Avariety <strong>of</strong> dye-terminator removal protocols exists to clean and purifythe sequencing reaction extension products. To determine their qualityand reproducibility, six protocols were tested with one large volumecontrol reaction aliquoted into a 96-well PCR plate. The protocolsincluded ethanol precipitation, gel filtration, and 4 solid phasereversible immobilization procedures with 1 utilizing the surface <strong>of</strong> aplate and the other 3 utilizing magnetic beads. The cleaned and purifiedsequencing reactions were processed on the 3730 DNA Analyzer(Applied Biosystems), and the contiguous read lengths, QV20+ scores,and signal strengths <strong>of</strong> the resulting sequences were analyzed withSequence Scanner v1.0 (Applied Biosystems). Based on the results fromtwo replicate rounds <strong>of</strong> testing, the gel-filtration protocol provided thelongest contiguous read lengths and highest QV20+ scores.155 Simplified Reagents and Workflowsfor Robust Sample Preparation <strong>of</strong>DNA, mRNA, and Small RNAF.J. Stewart, C.L. Hendrickson, L.M. Apone,D.B. Munafo, C.R. MeyerNew England Biolabs, Inc., Ipswich, MA, UnitedStatesAs yields <strong>of</strong> data generated by the Illumina, SOLiD, and 454sequencing platforms increase, NGS users have transitioned fromperforming multiple sequencing runs per sample to multiple samplesper sequencing run. As a result, the bottleneck in sequencing labs hastransitioned from data generation to sample preparation, necessitatingthe development <strong>of</strong> streamlined library construction workflows. Wehave developed a series <strong>of</strong> reagents to facilitate the easy preparation<strong>of</strong> numerous samples in parallel, compatible with both manual andautomated pipelines. These reagents reduce the amount <strong>of</strong> laborrequired, minimize error in reaction set up, and increase the stability <strong>of</strong>enzymes used in the construction <strong>of</strong> libraries for DNA, mRNA and SmallRNA sequencing. As a result, these reagents enable the development<strong>of</strong> robust workflows for both individual and high throughput samplepreparation.Poster Abstracts<strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine • 75


156 Comparison <strong>of</strong> Custom TargetEnrichment Methods: Agilent vs.NimblegenK. Bodi 1 , P.S. Adams 2 , D. Bintzler 3 , A. Perera 4 ,K. Dewar 5 , D.S. Grove 6 , J. Kieleczawa 7 , R.H. Lyons 8 ,T. Neubert 9 , A. C. Noll 1 , S. Singh 10 , R. Steen 11 ,M. Zianni 121Tufts University, Boston, MA, United States;2Trudeau Institute, Saranac Lake, NY, UnitedStates; 3 DNA Analysis, Inc., Cincinnati, OH, UnitedStates; 4 Stowers Institute, Kansas City, MO, UnitedStates; 5 McGill University, Montreal, QC, Canada;6Pennsylvania State University, University Park,PA, United States; 7 Pfizer Research, Cambridge,MA, United States; 8 University <strong>of</strong> Michigan, AnnArbor, MI, United States; 9 New York University, NewYork, NY, United States; 10 University <strong>of</strong> Minnesota,Minneapolis, MN, United States; 11 Harvard MedicalSchool, Cambridge, MA, United States; 12 OhioState University, Columbus, OH, United StatesOver the last four years, we witnessed the tremendous advances inNext Generation Sequencing (NGS) that have dramatically decreasedthe cost <strong>of</strong> whole genome sequencing. However, the cost <strong>of</strong> sequencinglarger genomes is still significant. In addition and depending on thegoal <strong>of</strong> study, whole genome sequencing creates a large amount <strong>of</strong>additional/auxiliary data that complicates data analysis. There areseveral commercial methods available for isolating subsets <strong>of</strong> genomesthat greatly enhance the efficiency <strong>of</strong> NGS by allowing researchersto focus on their regions <strong>of</strong> interest. For the 2009-11 DSRG study,we compared products from two leading companies; Agilent andNimblegen, that <strong>of</strong>fer custom enrichment methods. Both companiesobtained the same genomic DNA stock and performed DNA captureon the same specified regions. Following capture, the Illumina GenomeAnalyzer IIx system was used, in two different laboratories, to generatethe sequence data. We present our data comparing in terms <strong>of</strong> cost,quality, reproducibility and most importantly completeness and depth<strong>of</strong> coverage. Acknowledgements: We would like to thank Agilent,Illumina and Nimblegen for all their support in making this studypossible.157 A Methodology Study forMetagenomics Using Next GenerationSequencersD. Grove 1 , I. Albert 1 , D. Bintzler 2 , K. Bodi 3 ,M. Bruns 1 , K. Dewar 4 , G. Gloor 5 , T. Johnson 6 ,J. Kieleczawa 7 , R.H. Lyons 8 , T. Neubert 9 ,A.G. Perera 10 , S. Singh 6 , R. Steen 11 , M. Zianni 121Penn State University, University Park, PA, UnitedStates; 2 DNA Analysis, Inc., Cincinnati, OH, UnitedStates; 3 Tufts University, Boston, MA, UnitedStates, 4 McGill University, Montreal, QC, Canada;5University <strong>of</strong> Western Ontario, London, ON,Canada; 6 University <strong>of</strong> Minnesota, Minneapolis,MN, United States; 7 Pfizer Research, Cambridge,MA, United States; 8 University <strong>of</strong> Michigan, AnnArbor, MI, United States; 9 New York University, NewYork, NY, United States; 10 Stowers Institute, KansasCity, MO, United States; 11 Harvard Medical School,Cambridge, MA, United States; 12 Ohio StateUniversity, Columbus, OH, United StatesMetagenomics is one <strong>of</strong> several genomics applications, which hasbenefited immensely from the high throughput and cost efficacy <strong>of</strong>Next Generation sequencers. And although hundreds <strong>of</strong> studies onmetagenome analysis have been published over the past few years,the methodology for conducting them is still very much evolving.In this DSRG study we will evaluate the influence <strong>of</strong> various samplepreparation methods, specifically DNA extraction and amplificationapproaches, on data output along with a comparative analysis <strong>of</strong> NextGeneration sequencing platforms. We will study the effect <strong>of</strong> thesedifferent experimental and technical strategies on determination <strong>of</strong>sample biodiversity.158 What Your Blood Has to Say:Amplifying Blood RNA for theAffymetrix GeneChip® PlatformN. Supunpong Hernandez, T. Barta, C. Willis,R. Conrad, P. Whitley, K. BramlettLife Technologies, Austin, TX, United StatesPoster AbstractsGene Expression measurements from human blood RNA havebecome an increasingly important research area <strong>of</strong> focus. A reliableand consistent workflow to obtain RNA measurements from bloodwould serve to open the clinical arena to gene expression studies aspotential diagnostic indicators. The challenge lies in isolating highquality RNA from human whole blood at room temperature withoutcompromising gene expression pr<strong>of</strong>iles. To address this challenge, wereport a workflow using newly developed MagMax TM RNA isolation kitsfor Tempus TM stabilized blood and PAXgene® stabilized blood. Thesetwo stabilization methods are currently the most widely used methodsfor blood collection and stabilization in clinics in the United States.High quality RNA generated from these two RNA isolation processeswas amplified using the Ambion MessageAmp TM Premier RNAAmplification Kit and hybridized to Affymetrix GeneChip® microarrays.This platform and experimental tools are used, in combination, todemonstrate high quality gene expression pr<strong>of</strong>iles from human bloodRNA. The reported experimental design includes human whole bloodobtained from two donors collected and stabilized in either Tempus TMPAXgene® tubes. RNA was isolated using new MagMax TM RNA isolation76 • <strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine


kits for Tempus TM and PAXgene® stabilized blood. Isolated total RNAwas then processed through the GLOBINclear TM -Human kit and thenamplified with the MessageAmp TM Premier kit creating a library forhybridization to the Affymetrix Human U133A 2.0 expression arrays.Affymetrix GeneChip® microarray analysis showed parameters withinnormal limits for expressed genes. Reported results<strong>of</strong> this controlledexperiment, support a validated gene expression workflow for bloodon Affymetrix expression arrays using a combination <strong>of</strong> commercial kitsfor RNA purification from stabilized blood and library amplificationoptimized for hybridization and analysis on expression microarrays.159 Performance Comparison <strong>of</strong> FourMethods Utilized for the Purification<strong>of</strong> Enzymatically-Digested,Fluorescently-Labeled PCR FragmentsGenerated During T-RFLP AnalysisM. Zianni, J. Panescu, P. KumarThe Ohio State University, Columbus, OH, UnitedStatesThe fluorescently-labeled Terminal Restriction Fragment LengthPolymorphism (T-RFLP) assay based on amplified ribosomal DNAfrom bacteria is an inexpensive, widely accessible, effective and wellestablishedmolecular technique for the identification and comparativequantification <strong>of</strong> bacterial species in metagenomics. In order t<strong>of</strong>acilitate the detection <strong>of</strong> a large proportion <strong>of</strong> species in a givensample, it is necessary to maximize the recovery <strong>of</strong> the fluorescentlylabeled,restriction enzyme-digested PCR fragments generatedduring the process. The post-digestion purification method used t<strong>of</strong>acilitate fragment separation is a critical step in the retention <strong>of</strong> DNAfragments. Four methods for post-digestion purification were testedin an effort to characterize their integrity, effectiveness, ease <strong>of</strong> useand potential biases: solid phase reversible immobilization (AMPure TMand CleanSEQ TM ), contaminant capture (BigDye® XTerminator TM ),and dialysis (Millipore TM Nitrocellulose Membranes). Samples werecollected from four different oral sites in each <strong>of</strong> 16 patients. Eachsample was divided equally and the DNA was isolated with two distinctmethods. PCR reactions were carried out with two paired universalprimers for the 16S gene that were labeled with VIC and FAM, purifiedwith AMPure TM and digested separately with HhaI and MspI restrictionenzymes. From each <strong>of</strong> the digestions, identical aliquots were purifiedwith the four different methods. The DNA fragments were analyzedon the Applied BioSystems TM 3730 DNA Analyzer using identicalconditions, followed by data analysis with GeneMapper® v4.0. Basedon preliminary data analysis, CleanSEQ TM is the superior purificationmethod because it resulted in the most numerous peaks recovered persample with a wide distribution <strong>of</strong> sizes from approximately 50 to 1200bp.160 Electrochemical Simulation<strong>of</strong> Covalent DNA AdductFormation Monitored with LiquidChromatography/Mass SpectrometryJ. Powers 2 , S. Plattner 1 , R. Erb 1 , F. Pitterl 1 ,J.P. Chervet 2 , H. Oberacher 11Institute <strong>of</strong> Legal Medicine, Innsbruck MedicalUniversity, Innsbruck, Austria; 2 Antec, Palm Bay, FL,United StatesDNA adduct is a piece <strong>of</strong> DNA covalently bond to chemicals. Adductsactivate repair processes and, unless repaired prior to replication, maylead to nucleotide substitutions, deletions, and other rearrangements.As alterations <strong>of</strong> the genetic material can cause severe diseasesincluding cancer, inflammation, and neurodegenerative disorders,tests on mutagenicity/genotoxicity have become an integral part <strong>of</strong> riskassessment during the development <strong>of</strong> any new chemical. A number <strong>of</strong> invitro tests using different cell lines and in vivo tests mainly using rodentsare available to identify hazardous compounds. The majority <strong>of</strong> thesetests are time-consuming, labor-intensive and hardly automatable. So insearch for alternative methods, the usefulness <strong>of</strong> electrochemistry (EC)-liquid chromatography (LC)-mass spectrometry (MS) was evaluated.Generally, EC represents a powerful tool to study in vitro biologicaloxidation processes <strong>of</strong> a number <strong>of</strong> different compounds, includingdrugs, peptides and proteins. Here, we used EC to initiate adductformation. The obtained reaction products were separated by LC anddetected by MS. Tandem MS experiments were used for structuralconfirmation. In a pro<strong>of</strong> <strong>of</strong> principle study acetaminophen was selectedas model compound. Covalent adduct formation was observed forelectrochemical activated mixtures <strong>of</strong> acetaminophen and guanosine.Mechanistic studies revealed that adduct formation will only start atelectrochemical potentials sufficiently high to initiate oxidation <strong>of</strong> bothacetaminophen and guanosine. The stringent necessity <strong>of</strong> coactivationsheds a new light on the mechanism <strong>of</strong> adduct formation, becauseaccording to the generally accepted theory activation <strong>of</strong> the adductformingagent should be sufficient. Chromatographic separationenabled the differentiation <strong>of</strong> four isomeric forms. Their connection toacetaminophen was proven in dose-response experiments. EC/LC/MSrepresents a fast, simple, convenient and functional tool to study DNAadduct formation which has great potential to complement the existingbattery <strong>of</strong> mutagenicity/genotoxicity tests.**161 Quantitative miRNA ExpressionAnalysis Using Fluidigm Micr<strong>of</strong>luidicsDynamic ArraysJ. Jen, J. Sung Jang, V.A. Simon, R.M. Feddersen,F. Rakhshan, D.A. Schultz, M.A. Zschunke,W.L. Lingle, C.P. Kolbert.Mayo Clinic Micorarray Shared Resource andBiospecimens Accessioning Processing SharedResource, Rochester, MN, United StatesPoster AbstractsMicroRNA (miRNA) is a small non-coding RNA that can regulate geneexpression in both plants and animals. Studies showed that miRNAs playa critical role in human cancer by targeting messenger RNAs that arepositive or negative regulators <strong>of</strong> cell proliferation and apoptosis. Here,we evaluated miRNA expression in formalin fixed, paraffin embedded(FFPE) samples and fresh frozen (FF) samples using a high throughputqPCR-based micr<strong>of</strong>luidic dynamic array technology (Fluidigm). Wecompared the results to hybridization-based microarray platforms<strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine • 77


Poster Abstractsusing the same samples. We obtained a highly correlated Ct valuesbetween multiplex and single-plex RT reactions using standard qPCRassays for miRNA expression. For the same samples, the micr<strong>of</strong>luidictechnology (Fluidigm 48.48 dynamic array systems) resulted in a leftshifttowards lower Ct values compared to those observed by standardTaqMan (ABI 7900HT, mean difference, 3.79). In addition, as little as10ng total RNA was sufficient to reproducibly detect up to 96 miRNAsat a wide range <strong>of</strong> expression values using a single 96-multiplexingRT reaction in either FFPE or FF samples. Comparison <strong>of</strong> miRNAsexpression values measured by micr<strong>of</strong>luidic technology with thoseobtained by other array and Next Generation sequencing platformsshowed positive concordance using the same samples but revealedsignificant differences for a large fraction <strong>of</strong> miRNA targets. The qPCRarraybased micr<strong>of</strong>luidic technology can be used in conjunction withmultiplexed RT reactions for miRNA gene expression pr<strong>of</strong>iling. Thisapproach is highly reproducible and the results correlate closely withthe existing singleplex qPCR platform while achievingmuch higherthroughput atlower sample input and reagent usage. It is a rapid, costeffective, customizable array platform for miRNA expression pr<strong>of</strong>ilingand validation. However, comparison <strong>of</strong> miRNA expression usingdifferent platforms requires caution and the use <strong>of</strong> multiple platforms.162 Apollo 200 Fully Integrated DNAHID System: Multi Channel Results inUnder 2 HoursS. Jovanuvich, O. El-Sissi, H. Franklin, B. Nielsen,S. Pagano, R. Belcinski, G. BogdanIntegneX, Inc, Pleasanton, CA, United StatesThe ideal solution to meet the requirements <strong>of</strong> modern humanidentification is an automated, DNA-based hu man identificationsystem that processes samples rap idly and at low cost. IntegenX Inc.will describe the Apollo 200 DNA HID System. The Apollo200 isthe first fully automated sample-to-answer system for STR based HID.The system is based on integration <strong>of</strong> the company’s proprietary andpatented technologies as well as its rapid in-house micr<strong>of</strong>luidic chipprototyping. Reagents in disposable cartridges are loaded on thesystem with up to four buccal swab samples, the sample processing isinitiat ed and a CODIS compatible pr<strong>of</strong>ile is ready in less than two hourswith no further user interaction. The Apollo 200 integrates all <strong>of</strong> thesample handling steps starting from buccal swab(s) or blood, throughcell lysis, DNA extraction, amplification, separation, and detection.Further, the system uses rapid PCR chemistry, on-board capillaryelectrophoresis, and integrated la ser induced fluorescence detection.Initial data from testing conducted by IntegenX and customers will beshown. **Apollo 200 is a trademark <strong>of</strong> IntegenX, Inc.163 Transcriptome Analysis Using Next-Generation Sequencing TechnologyK. Bramlett, K. Lea, L. Qu, P. Whitley,J. Schageman, J. GuLife Technologies, Carlsbad, CA, United StatesHigh throughput RNA sequencing (RNA-Seq) is becoming increasinglyutilized as the technology <strong>of</strong> choice to detect and quantify known andnovel transcripts. Multiple next-generation sequencing (NGS) platformsare available that enable transcriptome pr<strong>of</strong>iling through RNA-Seqworkflows. Demonstrations <strong>of</strong> the power <strong>of</strong> RNA-Seq to pr<strong>of</strong>ile thewell annotated transcriptome and also identify novel transcribedregions, gene fusions, and even identify novel classes <strong>of</strong> RNA arerapidly increasing in the field <strong>of</strong> RNA research. Our aim has been todevelop library preparation methods and tools that aid in the reliablegeneration <strong>of</strong> libraries for next generation sequencing from total RNA.Reported here are results from the development <strong>of</strong> the Ambion®RNA-Seq Library Construction kit optimized for sequencing on theIllumina® next generation sequencing instruments. We show resultsfrom two protocols utilizing the same reagents that allow generation<strong>of</strong> RNA-Seq libraries targeting either the small RNA fraction <strong>of</strong> totalRNA, or the whole transcriptome which includes transcripts larger than100 base pairs. Results are reported from Illumina® Genome AnalyzerII sequencing <strong>of</strong> both small RNA and transcriptome libraries with afocus on mapping to the miRBase and RefSeq references respectively.We also demonstrate the use <strong>of</strong> External RNA Control Consortium(ERCC) transcripts as spike-in controls for transcriptome librariesthat aid in quality control <strong>of</strong> the library generation procedure andaid in downstream data analysis. The library construction technologyembedded in the Ambion® RNA-Seq Library Construction kit enablesresearchers to analyze the transcriptome <strong>of</strong> their research samples ina precise, sensitive and robust manner while maintaining informationregarding the genomic DNA strand to which the RNA transcript mapsutilizing the Illumina® Genome Analyzer II sequencing platform. Theworkflow and results reported here demonstrate new commerciallyavailable options for library construction enabling small RNA andtranscriptome pr<strong>of</strong>iling and novel discovery using next-generationsequencing technology.164 A Case Study: Molecular Pr<strong>of</strong>iling <strong>of</strong>Breast Cancer from Formalin-Fixed,Archival Material — Gene ExpressionPr<strong>of</strong>iles from FFPE Samples withImproved RNA DecrosslinkingTechnologyR. Jaggi 1 , S. Quabius 2 , G. Krupp 31University Bern, Bern, Switzerland; 2 UKSHUniversity Klinik, Kiel, Germany; 3 AmpTec GmbH,Hamburg, GermanyWe have developed a novel demodifaction/decrosslinking protocolfor RNA recovery from archival (FFPE) material. The resulting FFPERNA quality is superior to RNA obtained with other commercialFFPE RNA isolation kits: larger RNAs can be recovered, and RTqPCRdata demonstrate less variability and lower Cq values. ThisFFPE RNA is suitable for differential gene expression measurementby qPCR, high concordance with parallel RNA samples from freshfrozentissues was observed. Prognosis <strong>of</strong> breast cancer is determinedby clinicopathological and molecular factors. We developed andvalidated molecular scores reflecting the hormone status (ER, PGR,HER2 scores) and the proliferation status (PRO score) <strong>of</strong> breast cancercells. The scores can be combined to an overall RISK score. Molecularscores are independent prognostic parameters, they were validated inpostmenopausal patients with estrogen receptor positive breast cancer.Multivariate analysis revealed that PRO and RISK scores outperformconventional parameters (histological grading and Ki-67 labelingindex). Molecular scores are based on routine pathological material,testing can be implemented easily into routine diagnosis.78 • <strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine


165 Benchmarking miRNA ExpressionLevels in Degraded RNA SamplesUsing Real-Time RT-qPCR andMicroarray TechnologiesJ. Holbrook 5 , S.V. Chittur 1 , S. Tighe 2 , V. Nadell 3 ,R. Carmica 4 , K. Sol-Church 5 , A.T. Yeung 61State University <strong>of</strong> New York at Albany, Albany,NY, United States; 2 University <strong>of</strong> Vermont,Burlington, VT, United States; 3 Ohio University,Athens, OH, United States; 4 The University <strong>of</strong>Texas Medical Branch, Galveston, TX, UnitedStates; 5 Nemours/A.I. duPont Hospital for Children,Wilmington, DE, United States; 6 Fox Chase CancerCenter, Philadelphia, PA, United StatesThe Nucleic Acid Research Group (NARG) has conducted experimentsto determine the impact <strong>of</strong> RNA integrity and priming strategies oncDNA synthesis and Real-Time RT-qPCR. As a continuation <strong>of</strong> the RNAintegrity theme, this year’s study was expanded to evaluate the impact<strong>of</strong> RNA integrity on priming strategies for the analysis <strong>of</strong> nine miRNAtargets using Real-Time RT-qPCR. The nine targets were selectedbased on data obtained by the Microarray Research Group (MARG)and represent groups <strong>of</strong> miRNAs that are expressed at low, medium, orhigh levels in the First Choice human brain reference RNA sample. Thetwo RT-qPCR priming strategies tested in this study include the miRNATaqMan assay (Megaplex) <strong>of</strong> ABI and the RT2 miRNA qPCR assay <strong>of</strong>Qiagen/SA Biosciences. The basis for the ABI assay design is a targetspecificstem-loop structure and reverse-transcription primer, whilethe Qiagen design combines poly (A) tailing and a universal reversetranscription in one cDNA synthesis reaction. For this study to assessboth RT methods, samples that were used as templates were humanbrain reference RNA that has been subjected to controlled degradationusing RNase A to RIN (RNA Integrity Number) values <strong>of</strong> 7 (good), 4(moderately degraded), and 2 (severely degraded). In addition tothis Real-Time RT-qPCR data, the same RNA templates were furtheranalyzed using universal poly (A) tailing followed by hybridizationto Affymetrix miRNA GeneChips. We present some insights into RTpriming strategies for miRNA and contrasts qPCR results obtained usingdifferent technologies.168 MicroRNA Analysis Using RNAExtracted from Matched Formalin-Fixed Paraffin-Embedded (FFPE) andFresh Frozen Samples on SOLiD TMSystemK. Lea, J. Gu, E. Zeringer, S. Heater, J. Schageman,C. Mueller, K. BramlettLife Technologies, Carlsbad, CA, United StatesArchived formalin-fixed paraffin-embedded (FFPE) specimensrepresent excellent resources for biomarker discovery, but it has beena major challenge to study gene expression in these samples due tomRNA degradation and modification during fixation and processing.MicroRNAs (miRNAs) regulate gene expression at post-transcriptionallevel and are considered as important regulators <strong>of</strong> cancer progression.Next generation sequencing technologies such as SOLiD TM providean ideal method for measuring the abundance <strong>of</strong> miRNA moleculesin different cancer stages and provide insightful information ontumorigenesis. However, currently there is no good method tosystematically study miRNA expression in FFPE samples on nextgeneration sequencing platforms. We have designed and developeda ligation-based miRNA detection method to capture small RNAsequences in FFPE samples and convert them into templates suitablefor sequencing on the SOLiD TM System. Total RNA was isolated frommatched lung adenocarcinoma FFPE and snap frozen tissues using anAmbion RecoverAll TM kit. A PureLink TM miRNA Isolation kit was usedto enrich the small RNA fraction in these total RNA samples. Librarypreparation using a SOLiD TM Total RNA-Seq kit with modified protocolwas performed on the enriched RNA followed by sequencing onSOLiD TM system. Our results show that small RNA extracted from FFPEsamples was successfully converted to small RNA libraries. Very similarmapping statistics were obtained from matched FFPE and fresh-frozensamples after SOLiD TM sequencing. A good correlation <strong>of</strong> miRNAexpression pattern was also observed. This suggests that miRNAmolecules are less affected by sample degradation and RNA-proteincrosslink. This study provides a foundation for miRNA expressionanalysis on SOLiD TM system using FFPE samples in cancer and otherdiseases.**169 Structural Insights into theMechanism <strong>of</strong> microRNA ModulatedViral TranslationS. Patel 2 , E.A. Pham 1 , P. Pang 1 , M.A. Winters 1 ,M. Eckart 2 , J.S. Glenn 1,31Department <strong>of</strong> Medicine, Stanford UniversitySchool <strong>of</strong> Medicine, Stanford, CA, United States;2Protein and Nucleic Acid Core Facility, StanfordUniversity School <strong>of</strong> Medicine, Stanford, CA, UnitedStates; 3 Palo Alto Veterans Administration MedicalCenter, Palo Alto, CA, United StatesMicroRNAs (miRNAs) are small non-coding regulatory RNAs that controla vast array <strong>of</strong> cellular processes by repressing mRNA translation. LiverexpressedmiR-122 is a miRNA that has been co-opted by hepatitis Cvirus (HCV) to enhance viral translation. Recently, miR-122 antagomirtherapy in non-human primates has been shown to suppress HCVviremia; this pro<strong>of</strong>-<strong>of</strong>-concept study demonstrates the considerablepotential <strong>of</strong> this novel antiviral strategy. The mechanism by which miR-122 modulates HCV translation, however, is unclear. To examine thestructural changes that miR-122 exerts on the HCV internal ribosomalentry site (IRES), we developed an advanced Selective 2’-HydroxylAcylation analyzed by Primer Extension (SHAPE) method <strong>of</strong> analyzingRNA architecture. SHAPE determines RNA secondary structure atsingle-nucleotide resolution, with an accuracy far superior to othermapping methods. Using the above strategy, we show that binding <strong>of</strong>miR-122 to one <strong>of</strong> its target sites within the 5’ UTR <strong>of</strong> HCV induces aconformational shift in the HCV IRES at the distant AUG translation startsite. Surprisingly, binding <strong>of</strong> miR-122 to its second target site in HCVis mediated by a number <strong>of</strong> non-canonical base-pairings. Mutation <strong>of</strong>the 3’ half <strong>of</strong> miR-122 (tail) disrupted these non-canonical interactionsand its ability to induce a conformational shift at the AUG start site.We also observed that, in vitro, the first miR-122 target site in HCVis part <strong>of</strong> a putative triple-strand RNA motif. These results providethe first demonstration that the tail <strong>of</strong> this liver-encoded miRNA candirectly alter the RNA conformation <strong>of</strong> the HCV IRES, and therebyprovide new insights into the mechanism by which miR-122 influencesviral translation. We also show that the 5’ UTR <strong>of</strong> HCV contains a triplexstructure important for viral translation.Poster Abstracts<strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine • 79


Poster Abstracts170 Development <strong>of</strong> ERCC RNA Spike-InControl MixesL. Qu, A. Lemire, K. Lea, D. Batten, S. Jian Gu,P. Whitley, K. BramlettLife technology, Carlsbad, CA, United StatesThe National Institute <strong>of</strong> Standards and Technology (NIST) hostedExternal RNA Control Consortium (ERCC) has been working since2003 to generate a common set <strong>of</strong> RNA controls that can be used ingene expression measurements. These controls have been designedto mimic natural eukaryotic mRNA sequences with the ability to beused across multiple measurement platforms. These controls open-upthe possibilities for technical evaluation <strong>of</strong> multiple gene expressionsystems including real-time quantitative PCR, one-color and two-colormicroarray systems, next generation sequencing platforms, as well asserve as an in-process quality control check for library amplificationprocesses. The long standing history <strong>of</strong> Ambion&reg; as the RNACompany creates a great opportunity to formulate the ERCCs controlsinto useful mixes that can be spiked into RNA after isolation. Each mixcontains 92 <strong>of</strong> the ERCC controls in a mixture that spans over 6 logs <strong>of</strong>dynamic range. The 92 transcripts in each mix are further divided into4 sub-pools that can be utilized to evaluate fold-change measurements<strong>of</strong> gene expression between the two mixes. TaqMan® Gene Expressionassays targeting the 92 transcripts are utilized to monitor progressthrough the transcriptome library preparation steps required tocreate a next generation sequencing (NGS) library from RNA. Wedemonstrated the use <strong>of</strong> TaqMan® assays to inform a researcher<strong>of</strong> library quality prior to continuing with an expensive and timeconsumingnext generation sequencing experiment. These controlsare platform agnostic and provide informative data for multiple librarygeneration methods targeting various gene expression measurementplatforms from RNA. ERCC RNA spike-in control mixes from theNIST traceable ERCC plasmids has great potential in the hands <strong>of</strong> ourcustomers to open-up new capabilities in understanding variabilityin RNA preparation, RNA library preparation, determining detectionlimits <strong>of</strong> measurement systems, and informing downstream analysis. Assuch, we report here the first in a potential line <strong>of</strong> RNA control productsleveraging the certified plasmid sequences from NIST.171 Effects <strong>of</strong> Particle Porosity on theSeparation <strong>of</strong> Larger MoleculesR. Freeman, D. DiFeo, H. Jurgen Wirth, A. GooleySGE Analytical Science, Austin, TX, United StatesThe pore structure <strong>of</strong> a chromatographic stationary phase accountsfor the vast majority <strong>of</strong> the surface area responsible for the separation.The pore diameter influences the overall surface area and with it thecapacity <strong>of</strong> the column but also limits the size <strong>of</strong> the analyte the columncan or should be used for. In adsorption chromatography the limitingeffect <strong>of</strong> the pore diameter is further enhanced by adsorbed analytemolecules partially blocking the pore structure. In the analysis <strong>of</strong> largemolecules pore diffusion becomes a crucial parameter in the efficiency<strong>of</strong> the column. A number <strong>of</strong> models deal with hindered mass transfer inporous systems. Effects <strong>of</strong> particle porosity on the separation <strong>of</strong> largermolecules are discussed and examples for the separation <strong>of</strong> small,medium and large analytes on various pore size stationary phases aregiven. Pore size is shown to be an important parameter when analyzinglarger molecules. By selecting the right pore size for a task the capacityand the mass transfer trade-<strong>of</strong>f can be optimized to achieve the bestpossible separation. For tryptic digests a 200 Å stationary phase issuperior to a 300 Å due to its two-fold increase in surface area while1000 Å are required for the separation <strong>of</strong> proteins.172 Biological Application <strong>of</strong> ConstrainedPeptidesE. Murage, M. Castro, H. FazeliBiosynthesis, Inc., Lewisville, TX, United StatesPeptides are attractive therapeutic agents for many diseases. However,poor cell permeability, short half life in vivo due to rapid enzymedegradation has long been a major drawback in their clinical application.In order to overcome this, modification <strong>of</strong> peptides backbone hasbeen <strong>of</strong> most interest. In particular, synthetic cyclic peptides are beinginvestigated as potential drug candidates due to their improved cellpermeability, enzyme stability, high receptor affinity and selectivity. Forinstance, hydrocarbon stapled peptides has been shown to bind andinhibit the NOTCH1 transcription factor. NOTCH proteins have beenshown to play a pivotal role in cellular differentiation, proliferationand apoptosis. Mutations in NOTCH1 have been linked with diseaseslike T-cell acute lymphoblastic leukemia.1 Thus constrained peptideassemblies would provide a good opportunity to explore the proteinproteininteractions due to the increased binding affinity comparedto the linear counterparts. As a result <strong>of</strong> the attractive biologicalproperties <strong>of</strong>fered by these modified peptides there is a growingdemand for constrained peptides in the current drug discoveryresearch. To support the unmet need for custom made constrainedpeptides, Biosynthesis Inc. is now <strong>of</strong>fering stapled peptides and lactambridge constrained peptides. 1. Raymond E. M., Melanie C., Tina N. D.,Cristina Del Bianco, Jon C. A., Stephen C. B., Andrew L. K., D. Gary G.,Gregory L. V., James E. B., Nature 2009, 462, 182-188.173 The University <strong>of</strong> Texas at Austin— Protein and Metabolite AnalysisFacilityM. Gadush, H. Lo, F. Geigerman, M. Mercado,M. PersonUniversity <strong>of</strong> Texas at Austin, Austin, TX, UnitedStatesThe Protein and Metabolite Analysis Facility at the University <strong>of</strong> Texasat Austin is a joint effort <strong>of</strong> the College <strong>of</strong> Pharmacy, Center for Researchon Environmental Disease (CRED), and the Institute for Cellular andMolecular Biology (ICMB). Services and collaborative research are<strong>of</strong>fered for the detection, characterization and quantification <strong>of</strong>biomolecules. The Facility’s goals are to provide sensitive proteinidentification and modification analyses, to provide custom peptidesyntheses, to <strong>of</strong>fer services for the identification and quantification <strong>of</strong>metabolites, nutrients and xenobiotics, to implement novel analyticalmethods, to improve the sensitivity <strong>of</strong> existing analyses, to provideconsultation on the selection and implementation <strong>of</strong> analytical methods,to <strong>of</strong>fer training in the usage and applications <strong>of</strong> the instrumentation,and to provide technical expertise in support <strong>of</strong> individual researchgoals. The ICMB portion <strong>of</strong> the Core contains an ABI Procise 492 cLCprotein sequencer, a Protein Technologies Inc. Symphony peptidesynthesizer, two Bio-rad Du<strong>of</strong>lows and a GE Heathcare AKTA proteinpurification systems, two Beckman System Gold HPLC systems, aBerthold Technologies Mithras luminescence and fluorescence detector,an Invitrogen gel electrophoresis set-up, an Art Robbins Instruments80 • <strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine


Phoenix crystallography robot and a LC-MALDI-TOF/TOF (an ABI 4700with a LC Packings Ultimate Nano-LC system with a Probot spottingrobot). In the College <strong>of</strong> Pharmacy, the Core has an Applied Biosystems4000 Q-trap LC MS/MS system with ESI, APCI and nanospray sourcescoupled with a Shimadzu LC-20AD HPLC system, ThermoFinnigan LCQion trap mass spectrometer with ESI, APCI and microspray interfacescombined with a Michrom Magic 2002 HPLC system, a ThermoFinniganTrace MS GC-quadropole with EI positive, negative CI and selected ionmonitoring (SIM), an ABI Voyager-DE Pro MALDI-TOF and a Bio-radBioplex 200 fluorescent microbead array system.174 Microscopy and Imaging Facility,Cornell UniversityR. Williams, C. Bayles, J. Dela Cruz, M. Riccio,R. Doran, W. ZipfelCornell University, Ithaca, NY, United StatesThe Microscopy and Imaging Facility <strong>of</strong> the Cornell University LifeSciences Core Laboratories Center (CLC) provides an array <strong>of</strong> sharedresearch resources and services relating to optical microscopy,fluorescence, and whole animal imaging. The mission <strong>of</strong> the facility isto provide cutting edge technologies and high quality services that willsignificantly contribute to life sciences research, training and educationprograms.175 Utilization <strong>of</strong> Flow Cytometry inPersonalized MedicineP. LopezNew York University, New York, NY, United StatesFlow cytometry is a multiparametric data-gathering tool with anenormous, well-established array <strong>of</strong> applications for intact cells,organelles, functionalized beads, lysed cell contents or supernatants.Multiparametric analysis using state-<strong>of</strong>-the-art cytometers has thepotential to yield a distribution <strong>of</strong> cellular phenotypes with highdimensionality (up to 20 parameters per cell simultaneously) specificto an individual disease state or system-wide perturbation. Purifiedpopulations <strong>of</strong> atypical and/or cells representative <strong>of</strong> the disease statecan be collected as the first step towards biomolecular characterization.Flow cytometers may be outfitted with multiwell-plate accessories toprovide single-cell capture <strong>of</strong> specified cells with correlation to eachcell’s individual phenotype (indexed sorting), or rapid sampling frommicrotiter plates (plug-flow). An exciting new cytometric innovationmay allow determining the full signature <strong>of</strong> biomarkers from individualcells . The massively multiparametric mass cytometer analyzer makesuse <strong>of</strong> the high sensitivity and resolution <strong>of</strong> mass spectroscopy torecord the elemental composition as well as metal-tagged biomarkerdistribution <strong>of</strong> individual cells at a rate <strong>of</strong> up to 1000 cells per second.176 Cell Imaging and Analysis Network(CIAN) - Multi-Platform Resourcesand Services<strong>of</strong> six scientific platforms: Cell Imaging (confocal and fluorescencemicroscopy; walk-up), Proteomics (2-D, DiGE and fluorescent proteinanalysis; walk-up), Automation and High throughput screening(Pinning robot and liquid handler; full service), Protein expression andantibody production (in collaboration with local animal facilities; fullservice), Genomics (real-time PCR; walk-up), and Data storage/analysis(cluster, server and workstations). Users get in-depth consultationfor proposed projects, and can obtain training in any <strong>of</strong> the walk-upaspects <strong>of</strong> the facility, or take advantage <strong>of</strong> the full-service platforms.CIAN is designed to facilitate training, enhance interactions, as well asshare and maintain resources and expertise.177 Danforth Center: Proteomics & MassSpectrometry Core FacilityL. Hicks, S. Alvarez, B. Zhang, Z. Liu, H. Wang,J. FazlicDanforth Center, St. Louis, MO, United StatesThe Proteomics & Mass Spectrometry Facility at the Donald DanforthPlant Science Center (http://www.danforthcenter.org/pmsf/) isequipped with state-<strong>of</strong>-the-art technologies for the detailed study <strong>of</strong>a wide range <strong>of</strong> biomolecules. The facility provides both full- and selfservicecapabilities to both internal and external clients at competitiverates. The facility <strong>of</strong>fers fast, high quality specialized analytical servicesincluding: protein extractions, liquid chromatographic separations; highresolution 1D/2D gel electrophoresis; gel image analysis and proteinexpression analysis; high-throughput protein spot excision; in-solutionand in-gel protein digestion; high-throughput protein identification;accurate protein molecular weight analysis; protein covalent/noncovalentcomplex analysis; biomolecule interactions (surface plasmonresonance); small molecule separation/structure determination; andprotein post-translational modification analysis. Major instrumentationincludes: LTQ Orbitrap Velos (Thermo Scientific), QSTAR XL Q-TOF,two 4000QTRAPs, two 6520 Q-TOFs (Agilent), 5975C GC-MS(Agilent), TriVersa NanoMate (Advion), Chip Cube (Agilent), two 1200HPLCs (Agilent), 3 nan<strong>of</strong>low HPLCs (LC Packings/Eksigent), SystemGold HPLC (Beckman Coulter), two Shimadzu HPLCs (Shimadzu),UPLC (Waters), Biacore2000,3100 OFFGEL fractionator (Agilent), 1Dand high resolution2D gel electrophoresis systems (BioRad/AmershamBiosciences), Typhoon 9410 (Amersham Biosciences), GelPix (Genetix),and MultiProbe II (Perkin-Elmer). Protein intact mass, identification andcharacterization are a few <strong>of</strong> the many applications that the facilityperforms. For proteomics applications,two Q-TOFs andthe LTQOrbitrap Velos instruments are well-suited for analyzing both smallpeptides and large proteins. The LTQ Orbitrap Velos can be set upwith the TriVersa to automate direct infusion <strong>of</strong> samples to performexact mass measurements for molecular formula determination orfacilitate targeted analysis <strong>of</strong> modifications. The LTQ Orbitrap Velosand the 6520 Q-TOF are also used for online LC-based quantitativeproteomics (iTRAQ and label-free). The 4000 QTRAP systems serveas powerful instruments for targetedmetabolite analyses (e.g. planthormones, pABA, etc.), and the GCMS and one LC-QTOF are used formetabolomics pr<strong>of</strong>iling initiatives.Poster AbstractsE. Küster-Schöck, J. Lacoste, G. Lesage,S. Bunnell, H. HanMcGill University, Montreal, QC, CanadaThe Cell Imaging and Analysis Network (CIAN) provides servicesand tools to researchers in the field <strong>of</strong> cell biology from within oroutside Montreal’s McGill University community. CIAN is composed<strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine • 81


178 Proteomics and Mass SpectrometryApplications in Biomedical ResearchC. Diaz, M. Chow, R. Zheng, C. Silva-Sanchez,J. Koh, S. ChenUniversity <strong>of</strong> Florida ICBR Proteomics Facility,Gainesville, FL, United States<strong>Biomolecular</strong> interaction analysis (Surface Plasmon Resonance – Biacore).Each <strong>of</strong> our services is staffed and supported by highly experiencedand dedicated scientists. Beyond merely making available facilities andservices, the PAN facility also enables methods development, and newapplications development, designed to meet the needs <strong>of</strong> the researchcommunity requiring the services. We will present research exampleswhere the PAN facility played a significant role in the application <strong>of</strong>these technologies to basic science projects.Poster AbstractsProteomics and mass spectrometry have provided unprecedentedtools for fast, accurate, high throughput biomolecular separation andcharacterization, which are indispensable towards understandingthe biological and medical systems. Studying at the protein levelallows researchers to investigate how proteins, their dynamics andmodifications affect cellular processes and how cellular processes andthe environment affect proteins. The mission <strong>of</strong> our facility is to provideexcellent service and training in proteomics and mass spectrometryto UF scientists and students. Here we present our capabilities inproteomics and other analytical services. The tools include a gel-based2D-DIGE (Two Dimentional Difference Gel Electrophoresis) and gelfreeiTRAQ (Isobaric Tags for Relative and Absolute Quantitation).Along with our capacity <strong>of</strong> separating thousands <strong>of</strong> proteins andcharacterizing differential protein expression, we have a suite <strong>of</strong> state<strong>of</strong>-the-artmass spectrometers available for biomedical sciences andadvanced technology research, including a tandem time-<strong>of</strong>-flight (4700Proteomics Analyzer, AB), quadrupole/time-<strong>of</strong>-flight (QSTAR XL, AB),and hybrid quadrupole-linear ion-trap (4000 QTRAP, AB). Theseinstruments are mainly used for protein identification, posttranslationalmodification characterization and protein expression analysis (e.g., MassWestern). Our facility is also set up to provide Edman de novo N-terminalprotein sequence analysis and Biacore biomolecule interaction analysis.We are fully set up to synthesize and purify peptides and have a goodtrack record with this service as well. Proteomics and mass spectrometryare useful in large-scale suvey <strong>of</strong> proteome for hypothesis generation aswell as in detailed analysis <strong>of</strong> target proteins for hypothesis testing. Ourservices also include accurate molecular weight analysis, MRM-basedprotein screening and targeted metabolite pr<strong>of</strong>iling. To ensure successand maximize productivity, the facility <strong>of</strong>fers education, consultation,data processing and reporting, and support <strong>of</strong> grant application.179 The Protein and Nucleic Acid (PAN)Facility at Stanford UniversityS. Patel, M. Eckart, N. Kosovilka, A. Sanchez,Y. Tran, P. Walker, R. Winant, E. ZuoBeckman Center, Stanford University, CA, UnitedStatesThe Protein and Nucleic Acid (PAN) Facility (http://pan.stanford.edu) atStanford University’s Beckman Center is a multifaceted biotechnologyfee-for-service laboratory providing services to the Stanford scientificcommunity, other non-pr<strong>of</strong>it and biopharmaceutical organizations. TheFacility’s mission is to be adaptable and responsive to the changingneeds <strong>of</strong> biomedical research by providing basic science investigatorscontinued access to key tools and applications in an efficient and costeffective manner. The Facility <strong>of</strong>fers a diverse array <strong>of</strong> instrumentationand technical capabilities in Molecular Genetics and Protein Analytics.In Molecular Genetics the services include Gene Expression andGenotypring (Affymetrix microarray), quantitative methylation andmutation analysis (PyroMark), qPCR, DNA sequencing and fragmentanalysis, Nucleic Acid QC (Agilent Bioanalyzer) and Oligonucleotidesynthesis. For protein characterization the laboratory <strong>of</strong>fers ProteinSequencing, Peptide Synthesis, Protein Identification (MALDI) and180 University <strong>of</strong> Nebraska MedicalCenter Mass Spectrometry andProteomics Core FacilityM. Wojtkiewicz, P. CiborowskiUniversity <strong>of</strong> Nebraska Medical Center, Omaha,NE, United StatesThe UNMC Mass Spectrometry and Proteomics Core Facility <strong>of</strong>fers abroad range <strong>of</strong> services, such as ESI and MALDI protein identificationusing Mascot and Sequest Algorithms, iTRAQ-based quantitativeproteomics, MRM protein quantitation, phosphoproteomics pr<strong>of</strong>ilingand molecular weight determination for proteins, peptides and smallmolecules. The facility is equipped with LTQ Orbitrap ETD, LTQ Velos,4800 MALDI TOF-TOF, 4000 Q TRAP, all with supporting nano-LCsystems. Although the majority <strong>of</strong> users are investigators from UNMC,we also provide services for other outside academics and corporations.For further information, visit our website: www.unmc.edu/mspcf.181 UCLA Molecular InstrumentationCenter- Proteomics and MassSpectrometry FacilitiesM. Sondej, W. Yan, R.J. Alvarado, G.A. Khitrov,G. Czerwieniec, J. StrouseUniversity <strong>of</strong> California at Los Angeles, Los Angeles,CA, United StatesThe UCLA Molecular Instrumentation Center (MIC) is a state-<strong>of</strong>-theartcampus-wide facility dedicated to enabling the use <strong>of</strong> moderninstrumentation in molecular characterizations (www.mic.ucla.edu). TheUCLA Molecular Instrumentation Center is housed within and managedthrough the Department <strong>of</strong> Chemistry and Biochemistry and is composed<strong>of</strong> five divisions: J.D. McCullough Laboratory <strong>of</strong> X-ray Crystallography,Magnetic Resonance Facility, Materials Characterization lab, W. M.Keck Proteomics Center and Mass Spectrometry (MS) Laboratory.The MIC operates as an open access center where qualified users areencouraged to perform their own sample analysis under the trainingand guidance <strong>of</strong> the MIC personnel and is available to researchers atUCLA, other academic institutions and commercial enterprises. TheUCLA Proteomics Center and Mass Spectrometry Laboratories havefive Ph.D. level staff memberswho are experts in sample preparation,2-D gel and other electrophoresis techniques, bioinformatics, andmass spectrometry. The equipment for the UCLA Proteomics Centerincludes Bio-Rad electrophoresis cells for running 1- and 2-D gels; Bio-Rad Fx Fluorescence Imager and GS-800 Densitometer for imaging;DIGILAB Genomic Solutions ProPicII spotcutter; Thermo LTQ FT MSwith Eksigent NanoLC-2D HPLC; Thermo LTQ Orbitrap XL MS withEksigent NanoLC-2D HPLC and a Bruker SolariX-hybrid Qq-FTMSequipped with a 15 Tesla Magnet System. Our Bioinformatics resourcecenter hosts a number <strong>of</strong> qualitative and quantitative s<strong>of</strong>tware for 2-Dgel and mass spectrometry data analysis and hardware such as a linuxcluster and servers for MS database searching and data storage.The MS82 • <strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine


Laboratory is located next door and works closely with the ProteomicsCenter. Their mass spectrometers includes Applied Biosystems-MDSSciex 4000 Q Trap with Autosampler; Applied Biosystems Q-Star EliteQuad-TOF Hybrid LC/MS/MS system; Applied Biosystems Voyager-DE STR MALDI-TOF; Thermo Finnigan LCQ Deca Ion Trap MS withAutosampler and PDA; Agilent 6890-5975 GC-MS with Autosamplerand Waters LCT Premier with ACQUITY UPLC and Autosampler.184 New MAbPac Phases for MonoclonalAntibody (MAb) Variant AnalysisG. Gendeh, S. Rao, Y.X. Hou, X. Liu, Y. Agroskin,C. PohlDionex Corporation, Sunnyvale, CA, United States182 Microscale Thermophoresis:Interactions <strong>of</strong> Proteins, SmallMolecules, Nucleic Acids, and VesiclesS. Duhr, P. BaaskeNanoTemper Technologies GmbH, Munich,GermanyThis work gives an overview on a new Technology for the measurement<strong>of</strong> biomolecule interaction that is termed Microscale Thermophoresis(MST). The term Microscale Thermophoresis refers to the directedmovement <strong>of</strong> molecules in optically generated microscopic temperaturegradients. This thermophoretic movement is determined by the entropy<strong>of</strong> the hydration shell around the molecules. Almost all interactionsbetween molecules and virtually any biochemical process related toa change in size, stability and conformation <strong>of</strong> molecules alters thishydration shell and can be quantified. Such changes allow quantification<strong>of</strong> binding affinities <strong>of</strong> proteins, nucleic acids and small molecules aswell as measurement <strong>of</strong> enzymatic activities with MST. In additionalso functional studies <strong>of</strong> small molecule inhibitors are possible. Themicroscopic temperature gradient is generated by an IR-Laser, whichis strongly absorbed by water. The readout method <strong>of</strong> the interactionanalysis is based on fluorescence: intrinsic fluorescence <strong>of</strong> proteins canbe used as well as proteins expressed with GFP/YFP/RFP and also dyelabeled biomolecules. In this presentation we will describe the technicaldetails and the benefits <strong>of</strong> the Microscale Thermophoresis technologyplatform. We will show examples for interaction measurements rangingfrom protein -ribosome, protein -protein, small molecule -receptorbinding to studies where the interactions between receptor containingvesicles and proteins are analyzed.183 Reduction <strong>of</strong> Sample Carryover inProteomics LC-MS ExperimentsG. Gendeh, M. Karsten, E.J. Sneekes, R. SwartDionex Corporation, Sunnyvale, CA, United StatesRelevant biomarkers are <strong>of</strong>ten present at concentrations near or belowthe detection limit <strong>of</strong> current analytical methods. Despite this challenge,several biomarker candidates have been identified and moved into thevalidation phase. The increase in sensitivity <strong>of</strong> analytical methods andmass spectrometry, in particular, over the past years is the reason forthis accomplishment. However, a sensitivity increase alone is insufficientto accurately identify potential biomarkers; carryover reduction is alsoimportant to ensure a marker is actually present in the sample beinganalyzed. Therefore, reduction <strong>of</strong> carryover has received increasedattention from the proteomics community.MAbs generally exhibit complex heterogeneity including glycosylation,oxidation, phosphorylation, amino-terminal modifications, incompleteprocessing <strong>of</strong> the C-terminus, and asparagine deamidation. Thesevariations in composition could impact their efficacy, stability, and safety.Monitoring and reporting such variations in therapeutic proteins isrequired by the FDA and other regulatory agencies. Two new MAbPac TMphases were developed to meet these needs. The MAbPac SCX-10 is anewly designed strong cation-exchange column for the characterization<strong>of</strong> heterogeneity <strong>of</strong> MAbs. This is a complementary addition to theexisting ProPac® WCX-10 column that provides high resolution andorthogonal selectivity for MAb charge variant analysis. The MAbPacSCX stationary phase is based on nonporous, highly cross-linked styrenictype polymeric media with a proprietary hydrophilic coating. Sulfonicacid functionality is added through controlled radical polymerizationgrafting. These particles exhibit a wide range <strong>of</strong> pH stability with highselectivity and minimal band spreading. The MAbPac SEC-1 is a newsize-exclusion chromatography (SEC) column specifically developed forcharacterization <strong>of</strong> monoclonal antibody (MAb) aggregates, enzymedigested fragments, and other size-based separation applications. TheMAbPac SEC column is based on high-purity, spherical, porous (300Å), 5 μm silica covalently modified with a proprietary diol hydrophiliclayer. This stationary phase can handle both high- and low- salt eluentsas well as mass spectrometry compatible eluents. The MAbPac columnis packed into a nonmetallic, biocompatible PEEK TM column housingto eliminate metal contamination from the column hardware that cancompromise MAb separations. The stationary phase is designed tominimize undesired nonspecific interactions between proteins and thestationary phase. Various applications with relevant comparisons alongwith a demonstration <strong>of</strong> the ruggedness <strong>of</strong> these new phases are shownin this poster.185 The VGN Proteomics Module: ATransferable Laboratory Module forUndergraduatesJ. MurrayVermont Genetics Network, University <strong>of</strong> Vermont,Burlington VT, United StatesThe Vermont Genetics Network (VGN) Outreach Core’s mission isto bring cutting-edge technology and knowledge to undergraduatesat colleges throughout the state <strong>of</strong> Vermont. The VGN ProteomicsOutreach project initiated in the fall <strong>of</strong> 2009 exposes undergraduatesin the state <strong>of</strong> Vermont to proteomics technology using handsonlaboratory experiences. We provide all teaching materials,laboratory materials and if necessary equipment for colleges withinthe state to run the module. All materials become the property <strong>of</strong> therecipient institution upon completion <strong>of</strong> the laboratory module. Theundergraduate students learn about this cutting edge technology andgain new skills that we believe will help them with their future scientificcareers. In this transferable module, students learn how proteinexpression in yeast is changed after exposure to oxidative stress oran environmental toxin. Total protein is then harvested and preparedfor 2D gel analysis. Proteins with differential expression are isolatedfrom the 2D gel and prepared for Mass Spectrometry at the UVMPoster Abstracts<strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine • 83


Poster AbstractsProteomics Core Facility. The data is processed and students examinetheir results and use bioinformatics tools to further understand thebiological implications <strong>of</strong> the results. The students than present theirfindings, describe specific proteins that showed differential expression.A hypothesis is presented explaining the biological relevance <strong>of</strong> theprotein expression change along with a plan for testing this hypothesis.This work was sponsored by Grant Number P20 RR16462, from theIDeA Networks <strong>of</strong> Biomedical Research Excellence (INBRE) <strong>Program</strong> <strong>of</strong>the National Center for Research Resources (NCRR), a component <strong>of</strong>the National Institutes <strong>of</strong> Health (NIH).186 Quantitative Western Blotting withAmersham ECL PrimeM. Winkvist, S. Grimsby, K. Söderquist,A. MarcussonGE Healthcare Bio-Sciences AB, Uppsala, SwedenWestern blotting is a well established technique used to study proteinsfrom a wide variety <strong>of</strong> sources. The technique is used throughout thelife sciences from basic research to medical diagnostic applications.Western Blot is at best considered as semi-quantitative and hencelimited to studies involving large protein differences.Here wedemonstrate the use <strong>of</strong> a new ECL TM reagent, Amersham TM ECL Primein a number <strong>of</strong> typical Western Blotting applications. The resultsdemonstrate that, Amersham ECL Prime can be used for detection <strong>of</strong>low abundant proteins, that signals are very stable over time and covera broad dynamic range. These features make Amersham ECL Primehighly suitable for accurate quantitative analysis.187 New Approaches to QuantitativeWestern BlottingM. Winkvist, Å. Hagner-McWhirter,K. Söderquist, S. GrimsbyGE Healthcare Bio-Sciences AB, Uppsala, SwedenFluorescent detection in Western blotting <strong>of</strong>fers high sensitivity, broaddynamic range and stability <strong>of</strong> signals. This makes it highly suitable forquantitative Western blotting. Here we show how fluorescent Westernblotting can be used for simultaneously detection <strong>of</strong> up to threedifferent proteins on the same blot at the same time and for detection<strong>of</strong> proteins <strong>of</strong> the same molecular weight without stripping and reprobing.We also demonstrate how fluorescent Western blotting with3 layer probing can be used to increase sensitivity and thereby enablesdetection <strong>of</strong> very low abundant proteins. Finally we demonstrate thatit is possible to compare the total protein amount to the target proteinby using Deep Purple TM protein staining prior to ECLTM Plex TM Westernblotting.188 Rubicon PicoPlex-NGS Kits Availablefor Sequencing Single Cells Using theIllumina Genome AnalyzerJ. Langmore, T. Kurihara, E. Kamberov,J. M’Mwirichia, T. Tesmer, D. OldfieldRubicon Genomics, Ann Arbor, MI, United StatesRubicon Genomics has released its PicoPlex-NGS kits to prepare singlecells for NGS analysis on the Illumina Genome Analyzer. These kitsenable single eukaryotic or prokaryotic cells to be lysed, DNA extractedand amplified, and adapted for paired-end sequencing in a 1-tube,3-hr, 4-step process. Although the read coverage is poor in a single lane,the reproducibility <strong>of</strong> the reads allows single cells to be compared forSNP and CNV genotype, mutations. The same amplified samples canbe used for PCR and microarray analysis, including genome-wide SNPgenotyping, mutation, and copy number analysis. PRC amplification ortarget enrichment can be used for high accuracy and coverage singlecellgenomic analysis.189 PRG-<strong>2011</strong>: Defining the InteractionBetween Users and Suppliers <strong>of</strong>Proteomics Services/FacilitiesD.H. Hawke 1 , T.M. Andacht 2 , M.K. Bunger 3 ,C.E. Bystrom 4 , L.J. Dangott 5 , H. Molina 6 ,R.L. Moritz 7 , R.E. Settlage 8 , C.W. Turck 91University <strong>of</strong> Texas MD Anderson Cancer Center,Houston, TX, United States; 2 Centers for DiseaseControl and Prevention, Atlanta, GA, UnitedStates; 3 RTI International, Research TrianglePark, NC, United States; 4 Quest Diagnostics, SanJuan Capistrano, CA, United States; 5 Texas A&MUniversity, College Station, TX, United States;6Center for Genome Regulation, Barcelona,Spain; 7 Institute for Systems Biology, Seattle, WA,United States; 8 Virginia Bioinformatics Institute,Blacksburg, VA, United States; 9 Max Planck Institute<strong>of</strong> Psychiatry, Munich, GermanyOver the last ten years the Proteomics Research Group (PRG) hasundertaken technical studies that have covered a wide range <strong>of</strong> issuesunique to the rapidly developing field <strong>of</strong> proteomics. These studieshaveincluded a range <strong>of</strong> qualitative and quantitative experiments. The PRGstudies have resulted in a great deal <strong>of</strong> attention not only within the<strong>ABRF</strong> community but also outside as is evident from numerous articlesdealing with proteomics methods, procedures and standardization.As the field continues to develop, the diversity <strong>of</strong> instrumentationand laboratory workflows have grown in tandem. Therefore, for thePRG<strong>2011</strong> study it seemed especially useful to perform a survey tohelp define future studies based on the current blend <strong>of</strong> sample typesand technologies and obtain a view <strong>of</strong> emerging trends. A survey wascreated to ascertain three main insights into core facility function: 1)How labs interact with their clients, 2) The capacity <strong>of</strong> labs to meet thedemands <strong>of</strong> their clients, and 3) The blend <strong>of</strong> experimental techniques<strong>of</strong>fered to and requested by clients. Survey questions were designed toobtain information from both users <strong>of</strong> core facilities and the directorsand personnel <strong>of</strong> core facilities. Questions covered such topics as thetype and age <strong>of</strong> instruments in use, how data is analyzed and presentedto client, sources <strong>of</strong> funding, and emerging proteomics trends. Resultsare compiled en masse and presented without regard to institution.190 GlycoMaster — S<strong>of</strong>tware forGlycopeptide Identification withCombined ETD and CID/HCD SpectraP. Shan, L. XinBioinformatics Solutions Inc., Waterloo, ON,CanadaObjective: To automate the data analysis for the identification <strong>of</strong>glycopeptides with combined ETD and CID/HCD fragmentation. Theinputs for GlycoMaster s<strong>of</strong>tware include the raw mass spectrometry84 • <strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine


data <strong>of</strong> a Thermo Orbitrap instrument and the list <strong>of</strong> proteins inthe sample. The list <strong>of</strong> proteins can be identified from the samemass spectrometry data by using a database search method. Thes<strong>of</strong>tware uses the signature ions <strong>of</strong> simple sugars in the HCD spectrato identify the HCD-ETD spectrum pairs <strong>of</strong> glycopeptides. Thenthe ETD spectrum in each pair is used to identify the glycopeptidesequence, the glycosylation site and glycan mass. The output <strong>of</strong>the s<strong>of</strong>tware is an excel file that contains information about theidentified glycopeptides, including glycan composition, mass error,glycan components, glycosylation site and glycopeptide sequence.A score is associated with each identification result that can be usedto sort the identifications according to confidence. GlycoMaster wastested with an ETD-CID dataset containing 3561 MS and 2738 MS/MS spectra from tryptic digest <strong>of</strong> reduced and alkylated 12 standardprotein mixture containing 3 glycoproteins ( human serotransferrin,chicken ovalabumin and ovomucoid) from Sigma. Glycopeptides wereenriched on cellulose column or by using ZIC-HILIC spin columns fromEMD. Purified glycopeptides were analyzed by LTQ Orbitrap VelosETD. PEAKSTM Studio Suite was used for spectral preprocessing andprotein identification. GlycoMaster reported 161 identified HCD-ETDspectrum pairs <strong>of</strong> glycopeptides. 95 <strong>of</strong> 161 were manually validated. 68glycopeptides were reported by GlycoMaster, and 40 glycopeptideswere validated.191 Development <strong>of</strong> an AutomatedMethod for Antibody Purificationand AnalysisG. Gendeh, W. Decrop, R. SwartDionex Corporation, Sunnyvale, CA, United StatesThe screening and analysis <strong>of</strong> monoclonal antibodies can be anextremely time consuming task, especially as the many <strong>of</strong> the workflowsused today require many manual steps. This situation is fully remediedby the technology <strong>of</strong>fered by the MAb Analysis platform from Dionex- a single platform can screen a large number <strong>of</strong> MAbs and, in a secondstep, provide detailed analytical information, including charge variantanalysis and aggregate assessment. This unique technology allowsdrug discovery laboratories to develop MAb therapeutics much morequickly than before, and bring these important new therapeutics tomarket in a shorter time frame.192 Fast QC <strong>of</strong> Intact MonoclonalAntibodies Using MALDI-Top-DownSequencingA. Resemann, R. Paape, L. Vorwerg, D. SuckauBruker Daltonics, Bremen, GermanyAntibodies are playing an important role in drug development in thelast years. The function <strong>of</strong> therapeutic monoclonal antibodies (mAbs) isdirectly depending on their structure including terminal modificationslike C-terminal lysine excision and N-terminal pyroglutamylation <strong>of</strong>the heavy chain. Here, we describe a method for very fast and simplevalidation <strong>of</strong> the terminal sequences <strong>of</strong> heavy and light chain <strong>of</strong> intactmonoclonal antibodies using MALDI-Top-Down-Sequencing (MALDI-TDS) using in-source decay (ISD). Intact mAb (IgG) in a concentration<strong>of</strong> 1 mg to 5 mg/ml was directly mixed with matrix solution without priorreduction and alkylation or separation <strong>of</strong> the different chains. We used1,5-diaminonaphtalene (DAN) as matrix because <strong>of</strong> its reductive andISD enhancing properties. The samples were mixed with matrix solutionand dried at ambient air on the MALDI sample plate. In a secondexperiment, the same samples were prepared using sDHB (super DHB),which is known to be a good ISD matrix but without reduction capacity.The samples in sDHB didn’t generate any ISD fragmentation due to theintact disulfide crosslinks in IgG. In DAN, the same samples generatedrich ISD spectra containing fragments from both the heavy and the lightchain <strong>of</strong> the antibodies. The ISD spectra permitted reading throughthe cysteine residues in the sequences implicating the disulfide bridgeswere reduced by DAN. Up to 60 residues <strong>of</strong> the different chains <strong>of</strong>various mAbs were matched and validated, respectively. A dedicateds<strong>of</strong>tware tool allowed a very fast interpretation <strong>of</strong> the spectra. Overallthe method takes only a few minutes from sample preparation,spectrum acquisition and processing to the result in form <strong>of</strong> sequenceannotations in the ISD spectra.**193 Developing a Redox-Sensitive RedFluorescent Protein BiosensorI. Magpiong 1 , N. Koon 2 , S.M. Yei 2 , A.J. Risenmay 2 ,K. Kallio 2 , S.J. Remington 21Mount Holyoke College, South Hadley, MA,United States; 2 University <strong>of</strong> Oregon, Portland, OR,United StatesRedox environments are <strong>of</strong> particular interest, especially in themitochondria with its highly reducing environment and its role as thecentral processing unit <strong>of</strong> apoptosis. Monitoring <strong>of</strong> mitochondrial redoxenvironments is crucial to the study <strong>of</strong> apoptotic disorders. Reporting<strong>of</strong> the thiol/disulfide status in live cells was made possible with thedevelopment <strong>of</strong> redox-sensitive green fluorescent protein (roGFP). Weaim to develop a red version redox-sensitive fluorescent protein (roRFP).Expanding the array <strong>of</strong> redox-sensitive proteins with a red version willenable simultaneous visualization <strong>of</strong> multiple reducing intracellularcompartments. mKeima is a monomeric red fluorescent protein thatabsorbs light maximally at 440nm and emits red light at 620nm. Thislarge Stokes shift is dramatically decreased in acidic environments. Byfollowing protocol similar to that used in the development <strong>of</strong> roGFP,surface residues at key positions were changed to cysteines andrandom mutagenesis was performed on varying excitation species <strong>of</strong>mKeima. Mutants were screened and a ratiometric variant <strong>of</strong> mKeimawas identified (roRFP2) which exhibits changes in its spectral propertiesas a result <strong>of</strong> changes in the thiol/disulfide equilibrium. Preliminaryfluorescence spectroscopy measurements <strong>of</strong> roRFP2 indicate a highlyreducing redox potential <strong>of</strong> -330mV indicating it may be a usefulprobe in reducing subcellular compartments such as mitochondria orin the cytoplasm. By employing vector recombination <strong>of</strong> shuttle vectorPYX142, we successfully targeted roRFP2 in vivo to the mitochondriaand cytoplasm <strong>of</strong> Saccharomyces cerevisiae. Expression <strong>of</strong> roRFP2 wasvisualized using fluorescence microscopy. Thus, through mutagenesisand residue substitution we successfully created a red version redoxsensitivebiosensor that tested effectively as a ratiometric indicatorand expressed in the mitochondria and cytoplasm <strong>of</strong> S. cerevisiae.Moreover, the redox potential <strong>of</strong> roRFP2 is significantly more negativethan the widely used roGFPs.Poster Abstracts<strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine • 85


Poster Abstracts194 PeaksDB: New S<strong>of</strong>tware forSubstantially Improved PeptideIdentification from Orbitrap ETDMass SpectrometryB. Ma 1 , J. Zhang 2 , L. Xin 2 , B. Shan 2 , W. Chen 21University <strong>of</strong> Waterloo, Waterloo, ON, Canada;2Bioinformatics Solutions, Inc., Waterloo, ON,CanadaObjective: To substantially improve the peptide identification sensitivityand accuracy from the Orbitrap ETD data with computational methods.Method: The algorithm takes full advantage <strong>of</strong> the characteristics<strong>of</strong> the Orbitrap ETD data, including: (1) high mass resolution <strong>of</strong> theprecursor ions, and (2) the distributions <strong>of</strong> different fragment iontypes in the MS/MS scans. For the first characteristic, a pre-search stepis conducted to determine the precursor mass error distribution. Thisdoes not only make the precursor mass more accurate by a s<strong>of</strong>twarerecalibration, but also allows the use <strong>of</strong> the mass error as an importantfeature in the peptide-spectrum matching score function. For thesecond characteristic, the frequencies <strong>of</strong> different fragment ion typesat different precursor charge states are statistically learned, and usedin the score calculation. Moreover, the precursor-related ions in theMS/MS spectra are removed. Additionally, the score function makesuse <strong>of</strong> the similarity between a database peptide and the de novosequencing result. Result: PeaksDB was compared against three othersearch engines: MSGF-DB, Mascot, and ZCore. The same shuffled decoydatabase was appended to the target database and searched togetherto estimate the false discovery rate (FDR) <strong>of</strong> each individual engine. Thesame search parameters were used for all engines except that MSGF-DB does not support variable PTMs. If no variable PTM is allowed, thenumbers <strong>of</strong> identified peptides <strong>of</strong> different engines at 1% FDR are:PeaksDB (2356) > MSGF-DB (2147) > Mascot (1459) > ZCore (1030).If a few common PTMs are allowed, the numbers change to PeaksDB(3501) > Mascot (2677) > MSGF-DB (2147) > ZCore(1125). Conclusion:PeaksDB substantially improved the sensitivity and accuracy <strong>of</strong> peptideidentifications on Orbitrap ETD data. At 1% false discovery rate,PeaksDB identified 1.3 to 1.6 times as many peptides as Mascot 2.3.195 IDSieve: Protein IdentificationUsing Peptide pI Filtering <strong>of</strong> MS/MSData for Improved Confidence inIdentificationsN.R. Garge 1 , K.D. West 1 , X. Zhang 1 , J.L. Bundy 2 ,J.L. Stephenson, Jr. 2 , B.J. Cargile 3 , M.K. Bunger 11RTI International, Research Triangle Park, NC,United States; 2 Centers for Disease Control andPrevention, Atlanta, GA, United States; 3 SapientProteomics, Morrisville, NC, United StatesThe main challenge <strong>of</strong> tandem mass spectrometry based proteomicanalysis is to correctly match the tandem mass spectra produced to thecorrect peptides. However, the large number <strong>of</strong> protein sequences ina database increases the chances <strong>of</strong> a false positive identification forany given peptide match. Here we present an automated algorithmcalled IDSieve that utilizes target-decoy database search strategy incombination with pI filtering to allow greater confidence for peptideidentifications. IDSieve considers the SEQUEST parameters Xcorrand ΔCn to assign statistical confidence (false discovery rates) tothe peptide matches. The distribution <strong>of</strong> predicted pI values forpeptide spectrum matches (PSMs) is considered separately for eachimmobilized pH gradient isoelectric focusing fraction, and matcheswith pI values within 1.5 times inter-quartile range (within pI range)are analyzed independently <strong>of</strong> matches outside the pI ranges. Wetested the performance <strong>of</strong> IDSieve and Peptide/Protein Prophet on theSEQUEST outputs from 60 immobilized pH gradient isoelectric focusingfractions derived from mouse intestinal epithelial cell protein extracts.Our results demonstrated that IDSieve produced 1355 more peptidespectrum matches (or 330 more peptides) than Peptide Prophet usingcomparable false positive rate cut<strong>of</strong>fs. Therefore, combining pI filteringwith the appropriate statistical significance measurements allows for ahigher number <strong>of</strong> protein identifications without adversely affectingthe false positive rate. We further tested the performance <strong>of</strong> pI filteringusing ID Sieve when samples were prefractionated using either pHrange 3.5-4.5 or 3-10, and either 24cm or 7cm IPG strips.196 Proteomics-Based Molecular Modelfor Prediction <strong>of</strong> HeterogenicTreatment Response and Discovery<strong>of</strong> New Cell Survival Mechanisms <strong>of</strong>Basal-Like Breast CancerT. Hemström, Y. Lyutvinskiy, R. ZubarevKarolinska Institutet, Stockholm, SwedenWe aim to build a proteomics-based model for prediction <strong>of</strong> drugresponse and discovery <strong>of</strong> new signaling-related mechanismsexplaining the heterogeneity in treatment response observed in thegroup <strong>of</strong> basal-like breast cancer (BLBC), which is a group <strong>of</strong> breastcancer associated with a relatively short survival <strong>of</strong> patients. A panel<strong>of</strong> cell lines <strong>of</strong> BLBC origin will be subjected to conventional as well asmolecular pathway specific anti-cancer drugs followed by MTT-assayviability assessment. The range <strong>of</strong> drug concentrations will cover themaximal plasma drug concentration <strong>of</strong> patients. Focus will be on celllines exhibiting a high or low sensitivity to treatment. Drug responses <strong>of</strong>such cell lines will be further examined in terms <strong>of</strong> cell cycle distributionand cell death levels and based on these assessments relevant drugconcentrations and exposure times will be chosen. Analysis <strong>of</strong> proteinexpression in cells exposed to relevant treatment conditions will beperformed by LC-MS/MS. By way <strong>of</strong> key node analysis <strong>of</strong> proteinexpression data cellular signaling involved in sensitivity or resistance todrugs will be hypothesized and validated by means <strong>of</strong> anti body-basedtechniques, experimental modulation <strong>of</strong> signaling and analysis <strong>of</strong> cellcycle and cell death. Thereby we will add two more dimensions, whichare the drug response-related changes in activity <strong>of</strong> signaling pathwaysand functional morphological changes, such as apoptotic cell death, togene expression-based models <strong>of</strong> treatment response. Drug responsedata in terms <strong>of</strong> viability, protein expression, and cellular signaling willbe integrated in a computerized model. By focusing on a well definedsubgroup <strong>of</strong> breast cancer exhibiting clinical heterogeneity in anticancerdrug response we have a reasonable good chance to developa fine-tuned model for prediction <strong>of</strong> treatment response and identifycurrently unknown drug resistance mechanisms, which may involve newpossible predictive markers and drug targets.86 • <strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine


**197 Multi-Functional SuperparamagneticIron Oxide Particles as CancerTherapeutic AgentsA. Narayanan 1 , P.M. Gannett 1 , J.A. Barr 1 ,R.L. Carroll 2 , S.L. Yedlapalli 21Robert C. Byrd Health Sciences Center,Department <strong>of</strong> Basic Pharmaceutical Sciences,West Virginia University, Morgantown, WV,United States; 2 C. Eugene Bennett, Department <strong>of</strong>Chemistry, West Virginia University, Morgantown,WV, United StatesSuperparamagnetic iron-oxide nanoparticles (SPIONs) are magneticnanoparticles may be useful for early detection and treatment <strong>of</strong>cancers. SPIONs provide therapeutic drug-loading capabilities andtargeting specificity through the use <strong>of</strong> antibodies or receptor specifictags. These versatile particles are prime candidates for improvingcurrent means to the treatment <strong>of</strong> cancer and potentially otherdiseases. This multi-disciplinary project includes various milestonesand multiple aims: i) identify and optimize SPION design parametersthat optimize aqueous stability and maximize amphiphilic character,ii) evaluate SPION drug storage and release characteristics, andiii) demonstrate binding and entry into targeted cells. The specificaim <strong>of</strong> this project is the coupling <strong>of</strong> cell-specific targeting agents toSPIONs. An antisense oligonucleotide against Survivin mRNA wassynthesized by solid-phase DNA synthesis with an amino-terminatedlinker on the 3’ end (5’CCCAGCCTTCCAGCTCCTTG-(CH2)6-3’NH2).SPIONs were prepared and then coated with a copolymer containingsurface carboxylic acid groups (-COOH). The carboxylic acid groupswere activated by treatment with N-((3-dimethylamino)-propyl)-Nethylcarbodiimide and coupled to oligonucleotides via the –NH2terminus to the –COOH groups resulting in the formation <strong>of</strong> an amidelinkage between the –COOH groups <strong>of</strong> the SPION and the –NH2 <strong>of</strong>the antisense oligonucleotide. Circular dichroism (CD) studies wereperformed to quantify/optimize coupling and to demonstrate antisenseSurvivin duplex formation was not inhibited by the presence <strong>of</strong> theSPION. CD results were correlated with agarose gel electrophoresisdata and demonstrated oligonucleotides coupling to the SPION andthat the SPION did not significantly alter duplex formation. Futurestudies will target cellular absorption and antisense binding to SurvivinmRNA using confocal microscopy (Supported, in part, from NIHGM081348 grant and the WVU Research Corporation).198 Ultrahigh-Performance Nano LC-MS/MS Analysis <strong>of</strong> Complex ProteomicSamplesG. Gendeh, E.J. Sneekes, B. de Haan, R. SwartDionex Corporation, Sunnyvale, CA, United StatesDetermination <strong>of</strong> the proteome and identification <strong>of</strong> biomarkers arerequired to monitor dynamic changes in living organisms and predictthe onset <strong>of</strong> an illness. One popular method to tackle contemporaryproteomic samples is called shotgun proteomics, in which proteins aredigested, the resulting peptides are separated by high-performanceliquid chromatography (HPLC), and identification is performed withtandem mass spectrometry. Digestion <strong>of</strong> proteins typically leadsto a very large number <strong>of</strong> peptides. For example, digestion <strong>of</strong> a celllysate easily generates 500,000 peptides. The separation <strong>of</strong> thesehighly complex peptide samples is one <strong>of</strong> the major challenges inanalytical chemistry. The main strategy to improve the efficiency <strong>of</strong>packed columns is either to increase column length or to decreasethe size <strong>of</strong> the stationary phase particles. However, to operate thesecolumns effectively, the LC conditions need to be adjusted accordingly.Naturally, the on-line coupling to MS systems has to be taken intoaccount in the optimization process. Here, the authors report on theperformance <strong>of</strong> nano LC columns operating at ultrahigh pressure.The effects <strong>of</strong> column parameters (particle size and column length)and LC conditions (gradient time, flow rate, column temperature)were investigated with reversed-phase (RP) gradient nano LC. HighresolutionLC-MS separations <strong>of</strong> complex proteomic peptide samplesare demonstrated by combining long columns with 2 &mu;m particlesand long gradients. The effects <strong>of</strong> LC parameters on performance andthe influence on peptide identification are discussed.**199 Comprehensive Biomarker DiscoveryPlatform Reveals Qualified MRMAssay for Prediction <strong>of</strong> INF/RibavirinTreatment Response in Hepatitis CPatientsJ.W. Thompson 1 , L.G. Dubois 1 , K. Patel 2 ,J.E. Lucas 1 , J. McCarthy 1 , J.G. McHutchison 2 ,M.A. Moseley 11Duke Institute for Genome Sciences and Policy,Durham, NC, United States; 2 Duke Clinical ResearchInstitute, Durham, NC, United StatesThe current standard <strong>of</strong> care for chronic Heptatitis C (CHC) results insustained viral response in only half <strong>of</strong> patients and also has significantside effects. Improved pre-treatment clinical predictors <strong>of</strong> responseare needed to individualize treatment and select individuals mostappropriate for new therapy. We have identified 10 peptides to 6proteins which can differentiate non-responders from responders withhigh accuracy based on pre-treatment samples. Serum samples wererandomized, immunodepleted (MARS 14), and digested with trypsinprior to LC-MSE or LC-MRM analysis. LC-MSE analyses were performedon a Waters nanoAcquity LC and QToF Premier, while MRM analyseswere performed on nanoAcquity and Xevo TQ. Rosetta Elucidator® wasused to quantify all LC-MSE data. PLGS v2.4 was used to make peptideidentifications. MRM method generation and sample quantitationwas performed with Skyline v0.6. The response signature was initiallyidentified from the analysis <strong>of</strong> 96 samples (Duke Hepatology ClinicalResearch database) by LC-MSE on a QToF mass spectrometer, usingsparse latent factor regression analysis. This yielded a group <strong>of</strong> almost400 candidate peptides which were part <strong>of</strong> 3 metaprotein predictors;these peptides were then curated based on set selection criteria togive 86 target peptides for MRM analysis. MRM analysis <strong>of</strong> the original96 samples yielded a final group <strong>of</strong> 10 peptides which maintainedBonferroni-corrected statistical significance for predicting treatmentresponse. The assay has been qualified by a blinded analysis <strong>of</strong> an allcomersclinical trial sample set (PEDS-C, NIH, n=51) using the MRMmethod, which yielded an AUROC <strong>of</strong> 0.91 with a sensitivity <strong>of</strong> 0.828and specificity <strong>of</strong> 0.786. The analysis <strong>of</strong> an additional CHC clinical trialcohort (Chariot, n = 243) is ongoing. This presentation will utilize CHCtreatment response as a case study on the critical analytical, statistical,and clinical cohort requirements to successfully perform biomarkerdiscovery and verification.Poster Abstracts<strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine • 87


Poster Abstracts200 Discovery <strong>of</strong> Yersinia pestis andYersiniophage Peptide Targets forUse in Multiple Reaction MonitoringMethodsL. Luna 1 , S. Bearden 21Division <strong>of</strong> Vector-Borne Infectious Diseases,Centers for Disease Control and Prevention,Fort Collins, CO, United States; 2 Toxicology andEnvironmental Research and Consulting, The DowChemical Company, Midland, MI, United StatesAims: We present the discovery <strong>of</strong> functional yersiniophage andYersinia pestis (Y. pestis) peptide targets for monitoring bacteriophagebasedamplification processes and species-specific identification <strong>of</strong>Yersinia pestis, the etiological agent <strong>of</strong> plague. Methods: For proteinconfirmation and peptide target discovery, ultra performance liquidchromatography and hybrid tandem mass spectrometry was utilizedto identify proteolytic cleaved peptides generated from digestion <strong>of</strong>whole phage and bacterial lysates. Peptide discovery was accomplishedwith a UPLC NanoAcquity interfaced into a Q-TOF Premier (WatersCorporation) for full scan and MS/MS experiments. Targeteddevelopment utilized the MRM initiated detection and sequencing(MIDAS) workflow design s<strong>of</strong>tware (Applied Biosystems) to create insilico MRM transitions which optimized collision energies for trypsincleaved peptides from the proteins <strong>of</strong> interest. The analytical columnutilized was an UPLC bridged ethyl hybrid (BEH) 1.0 x 50 mm reversephase C18 (1.7-µm particle size) interfaced into a 4000 Q TRAP forMRM triggered MSMS. Results: We screened yersiniophage phiA1122,R, V, and Y proteins for conserved peptide targets for utilization inroutine mass spectrometry monitoring <strong>of</strong> the phage amplificationprocess. We also identified murine toxin and F1 antigen peptide targetsthat are unique to Yersinia pestis, which upon lysis, can be utilized forconfirmatory identification <strong>of</strong> Y. pestis. Conclusions: Major and minorcapsid protein peptides will serve to enhance quantifiable metricmeasurements <strong>of</strong> the amplification process for rapid yersiniophagediagnostic capabilities and concomitant determination <strong>of</strong> the presence<strong>of</strong> viable Y. pestis bacilli. Moreover, the targeted murine toxin peptidedirectly confirmed the presence <strong>of</strong> Y. pestis at 26°C and can be appliedas an additional tool for zoonosis surveillance capabilities in a clinicallaboratory setting. Significance and Impact <strong>of</strong> Study: Applicationsutilizing these peptide targets identified in this work may be beneficialin emergency response situations requiring pre**201 Protein-Binding Assays inBiological Liquids Using MicroscaleThermophoresisS. Duhr 1 , P. Baaske 1 , C. Wienken 2 ,M.J. Willemsen 2 , D. Braun 21NanoTemper Technologies GmbH, and Centerfor Nanoscience, University Munich, Munich,Germany; 2 University Munich, Physics Department,Munich, GermanyProtein interactions inside the human body are expected to differ fromthe situation in vitro. This is crucial when investigating protein functionsor developing new drugs. In this study, we present a sample-efficient,free-solution method, termed Microscale Thermophoresis (MST), thatis capable <strong>of</strong> analysing interactions <strong>of</strong> proteins or small molecules inbiological liquids such as blood serum or cell lysate. The technique isbased on the thermophoresis <strong>of</strong> molecules, which provides informationabout molecule size, charge and hydration shell. We validated themethod using immunologically relevant systems including humaninterferon gamma and the interaction <strong>of</strong> calmodulin with calcium. Theaffinity <strong>of</strong> the small-molecule inhibitor quercetin to its kinase PKA wasdetermined in buffer and human serum, revealing a 400-fold reducedaffinity in serum. This information about the influence <strong>of</strong> the biologicalmatrix may allow to make more reliable conclusions on proteinfunctionality, will facilitate more efficient drug development, and mayallow for sensitive diagnostics in complex biological samples.202 2-D DIGE Analysis <strong>of</strong> MulticellularTumor Spheroids in Evaluation <strong>of</strong>Breast Cancer TreatmentV. Ruddat 1 , M. Winkvist 2 , S. Grimsby 2 ,A. Monazzam 3 , K. Nyamekye 3 ,Å. Hagner-McWhirter 21GE Healthcare, Piscatway, NJ, United States; 2 GEHealthcare Bio-Sciences AB, Uppsala, Sweden;3Uppsala Applied Science Lab, GE Healthcare,Uppsala, SwedenMany cancers can be diagnosed using positron emission tomography(PET) and PET can also be used to monitor how effective varioustreatments are in individual patients. Tumor spheroids are cancer cellsgrown on agar coated dishes forming a 3D structure. They are widelyused in preclinical cancer research, where the multicellular tumorspheroid model is considered biologically and physiologically similarto in vivo grown tumors. In this study we have used Two-dimensionalDifference Gel Electrophoresis (2-D DIGE) analysis to gain more insightin differences in protein expression as a result <strong>of</strong> drug treatment<strong>of</strong> multicellular tumor spheroids. 2-D DIGE can be used to identifypossible new biomarkers for development <strong>of</strong> new PET tracers and drugtargets. Combining 2-D DIGE and PET results can be used for improvingdiagnosis and treatment.203 N-Glycosylation <strong>of</strong> AntibodiesCharacterized by Mass Spectrometry:An Integrated S<strong>of</strong>tware ApproachU. Schweiger-Hufnagel, A. Asperger,A. Resemann, D. SuckauBruker Daltonics, Bremen, GermanyAntibodies represent one <strong>of</strong> the most important classes <strong>of</strong>glycoproteins playing a central role in the immune response <strong>of</strong> livingorganisms. Furthermore, there is a growing interest in recombinantantibodies as potential biotherapeutic agents. The analysis <strong>of</strong> theN-glycosylation pattern present on antibodies is challenging due to itsheterogeneous structure. The glycan pr<strong>of</strong>ile is highly dependent onthe process by which a recombinant glycoprotein is generated, suchas host organism and growth conditions. Changes to the glycosylationpattern can significantly alter biological function. To characterize theN-glycosylation pattern <strong>of</strong> a recombinant antibody, a bottom-upapproach was pursued. Tryptic digests <strong>of</strong> antibody samples wereseparated by nano-LC and analyzed by MALDI mass spectrometry. Anintegrated s<strong>of</strong>tware approach allowed a detailed characterization <strong>of</strong>the glycosylation pattern and visualization <strong>of</strong> the relevant mass spectra.LC-MALDI-TOF/TOF analysis <strong>of</strong> the digested antibody provided,in addition to the nearly complete sequence coverage <strong>of</strong> the nonglycosylatedpeptides, a detail-rich picture <strong>of</strong> the highly complex88 • <strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine


pattern <strong>of</strong> N-linked glycans in form <strong>of</strong> the respective N-glycopeptides.Targeted analysis <strong>of</strong> potential glycopeptides significantly increased theinformation content. Integrated glycoprotein analysis s<strong>of</strong>tware tools(ProteinScape 2.2) allowed identification <strong>of</strong> glycan modifications andinteractive result validation. In this process s<strong>of</strong>tware facilitated theinitial characterization <strong>of</strong> the antibody as well as the subsequent qualitycontrol tasks.204 Magnetic ZIC-HILIC BeadsEnrichment for Neutral and AcidicGlycopeptidesA. Resemann 1 , J. Wohlgemuth 2 , S. Andrecht 2 ,A. Schneider 1 , U. Schweiger-Hufnagel 1 , D. Suckau 11Bruker Daltonics, Bremen, Germany; 2 Merck KGaA,Darmastadt, GermanyGlycosylation is the most abundant protein posttranslational modificationand is involved in many relevant biological processes and crucial to theunderstanding <strong>of</strong> many diseases. In depth analysis <strong>of</strong> glycosylationsites is difficult, however, as glycopeptides exhibit a significant microheterogeneity at glycosylation sites. In addition, ion suppressioneffects require selective methods for glycopeptide enrichment. Massspectrometric analysis <strong>of</strong> glycopeptides is challenging because boththe peptide as well as the glycan moiety have to be elucidated for a fullstructural understanding. We used Fetuin, Asialo-Fetuin and Alpha-1-Acidglycoprotein to equally representing sialylated and non-sialylatedglycosylic structures. In addition, monoclonal antibodies were analyzedas a dedicated example for pharmaceutical QC. Proteins were digestedwith trypsin and glycopeptides were enriched using a dedicated ZIC-HILIC glycocapture beads in combination with an optimized buffersystem (EMD Chemicals Inc.). The glycopeptides were analyzed usingESI ion trap MS for glycopr<strong>of</strong>iling and MALDI-TOF/TOF-MS for indepth characterization <strong>of</strong> the glycopeptides. For database searches,an integrated s<strong>of</strong>tware approach was used: protein searches <strong>of</strong> theglycopeptide MS/MS spectra were performed for obtaining the aminoacid sequence <strong>of</strong> the glycopeptide, and searches in glycan databasesbased on the same glycopeptide MS/MS spectra were carried outto complete the characterization <strong>of</strong> N-linked glycopeptides. In thecurrent study, two important features <strong>of</strong> glycoprotein analysis areshown: (1) The employed integrated s<strong>of</strong>tware approach allowed theglycan identification in a similar way as peptide identification. Theimportant step <strong>of</strong> interactive result validation was facilitated by a suite<strong>of</strong> dedicated data and result viewers. (2) Compared to MS analysis <strong>of</strong>native glycoprotein digests, the enriched samples allowed to detectmore glycopeptides and permitted the acquisition <strong>of</strong> higher qualityMS/MS spectra. For MALDI-TOF/TOF-MS analysis, linear positive ionmode detection <strong>of</strong> precursor ions proved to be highly suitable for theanalysis <strong>of</strong> even multi-sialylated glycopeptides.205 Fragment Analysis <strong>of</strong> CarbohydratesFollowing Capillary ElectrophoresisT. Snyder-Leiby, D. Hulce, F. Li, X. Li, C.S. LiuS<strong>of</strong>tGenetics, LLC, State College, PA, United StatesDepending on the macromolecule size and configuration, migrationrates through capillary electrophoresis vary greatly. Internal sizestandards for capillary electrophoresis <strong>of</strong> the same macromolecule maynot be readily available. The Macromolecule Tool in GeneMarker&reg;aids with analysis <strong>of</strong> macromolecule fragments without an internallane size standard. Methods included importing raw data files to thes<strong>of</strong>tware and physically identifying reference peaks in the samplesknown to have the same size. The program uses this information tocalibrate from one capillary to another. Characteristics <strong>of</strong> the aligneddata (such as relative size, peak height, peak area, peak ratios) wereexported in an excel sheet. Ninety six raw data files from 4 dye capillaryelectrophoresis were analyzed. Peak height, height ratio, area, arearatio, and relative sizes were determined for all samples. These valuescan be used to determine characteristics such as number and relativesize <strong>of</strong> degradation products or other macromolecules, such as DNAbinding carbohydrates commonly functioning in gene regulation.206 Analysis <strong>of</strong> Complex Oligosaccharidesfrom Glycopeptides andGlycoproteins Using MSn Spectra andOligosaccharides Spectral LibraryF. XiangShimadzu Biotech, Pleasanton, CA, United StatesGlycosylation is a common post-translational modification to cellsurface and extra cellular matrix proteins as well as to lipids. Unlikeproteins and nucleic acids that are linear polymers <strong>of</strong> amino acidsand nucleotides respectively, with linkages at only one position,carbohydrates can adopt complex branched structures with individualmonomeric units linked at one <strong>of</strong> several sites. A detailed analysis<strong>of</strong> complex carbohydrate structures has been explored with massspectrometric techniques, and still presents challenge for the analyst.This presentation will describe a systematic approach <strong>of</strong> carbohydratesand glycoconjugates analysis with MSn techniques. Oligosaccharides,cleaved from glycoproteins (by hydrazinolysis or enzymatically), werecharacterized using a hybrid MALDI Ion Trap / TOF mass spectrometer.High mannose, biantennary and triantennary oligosaccharides wereanalyzed using MS, NS2, MS3 and MS4 modes. Intact oligosaccharideswere analyzed in MS mode using a cooling gas to prevent fragmentation.Individual precursor ions were isolated in the trap, subjected t<strong>of</strong>ragmentation with Argon, to provide MS2 data. Product ions wereselected for further fragmentation, which was achieved by increasingthe energy for collisionally induced dissociation. In MS mode, the singlycharged sodium adduct form <strong>of</strong> these molecules was detected, whichis typical <strong>of</strong> the analysis on conventional MALDI mass spectrometers.In MS2 mode, high mannose oligosaccharides readily lost the coreN-acetylglucosamine residues, whilst MS3 and MS4 modes were usedto sequentially fragment the product ion corresponding to the residualbranched mannose oligomer. In MS2 mode analysis <strong>of</strong> biantennaryand triantennary structures also fragmented losing disaccharide units,such as the galactose - N - acetylglucosamine units that define eachantennary branch, or core fucosylated - N - acetylglucosamine units.MS3 <strong>of</strong> selected MS2 products ions could be used to differentiatefragments generated from either the reducing or non-reducing ends.Cross-ring cleavages were also observed during fragmentation in MS2,and the relevant product ions could be used to differentiate branchedstructures by further fragmentation in MS3 mode. Many tandem massspectrometric experiments have been revealing that oligosaccharidesmight have characteristic signal intensity pr<strong>of</strong>iles, depending on theglycosidic linkage and branching structures. In addition to the MSncapability <strong>of</strong> the platform, there is a library <strong>of</strong> observational mass spectraacquired from structurally defined oligosaccharides. The presentationwill show the enhancement <strong>of</strong> carbohydrate identification by utilizingcomparison procedure <strong>of</strong> the signal intensity pr<strong>of</strong>iles <strong>of</strong> MSn spectrabetween the analyte and structurally defined oligosaccharides in thelibrary.Poster Abstracts<strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine • 89


Poster Abstracts207 A Unique Workflow for GlycoproteinCharacterization from SamplePreparation to MS/MS SpectralInterpretationJ. Albanese 1 , R. Lee 21AB SCIEX, Foster City, CA, United States; 2 ProZyme,Inc., Hayward, CA, United StatesProtein glycosylation is a complex dynamic post-translationalmodification, which is used by an organism to regulate a number <strong>of</strong>important functions. Variable composition, linkage, branching andanomericity <strong>of</strong> the constituent monosaccharides in combination withthe general heterogeneity due to the indirect, non-template control<strong>of</strong> their biosynthesis are the basis <strong>of</strong> the structural complexity <strong>of</strong>glycoprotein glycans. This poster presents a complete workflow forglycoprotein characterization comprising sample preparation forglycan release, glycan separation using graphitized carbon separationcoupled to MALDI spotting, automated MS and MS/MS analysisMALDI-TOF analysis and spectral interpretation with SimGlycan®S<strong>of</strong>tware. The workflow has been first optimized using commerciallyavailable immunoglobulin G from human serum and a glycan library<strong>of</strong> defined bisecting and none-bisecting glycan structures. The IgGsample contains both different protein sequence is<strong>of</strong>orms (IgG1-3) anddifferent glycan is<strong>of</strong>orms. The results demonstrated that, the RGSPS kitwith InstantABTM labeling provides a fast and robust glycan releaseand labeling method including sample clean up in less than 2 hoursprior to mass spectrometric analysis. The combination <strong>of</strong> the LC MALDIWorkflow with the nan<strong>of</strong>low hypercarb column enables better glycanseparation and therefore more specificity for isomeric glycans structurescharacterization. High energy CID MS/MS spectral comparison <strong>of</strong> thereleased and/or labeled glycans with the control samples provides agreater number <strong>of</strong> MS/MS fragments than electrospray CID MS/MSspectra or MALDI post source decay (PSD) spectra for more detailedinformation. Next, this robust workflow will be tested and expandedon therapeutically interesting antibodies. Studying the heterogeneity<strong>of</strong> glycosylation patterns obtained from cell culture influences can helpunderstand efficacy, binding affinity, specificity and pharmacokineticproperties.208 Analysis <strong>of</strong> Intact Proteins inBiotechnologyR. Freeman, D. DiFeo, H.J. Wirth, A. GooleySGE Analytical Science, Austin, TX, United StatesThe demand for separation techniques for intact proteins is increasingwith the introduction <strong>of</strong> a new generation <strong>of</strong> high resolution massspectrometers which are able to measure the mass <strong>of</strong> small to mediumsize proteins very accurately. Liquid chromatography is a valuable toolfor separating these proteins prior to the MS analysis. Intact proteinchromatography is most commonly used in a top-down approach inproteomics or to determine expression levels during recombinantprotein synthesis. The size <strong>of</strong> the protein molecule results in verylow diffusion coefficients and therefore slow mass transfer in and out<strong>of</strong> the pore system. A sufficiently large pore diameter is required tominimize the effects <strong>of</strong> restricted pore diffusion. We show examples <strong>of</strong>the separation <strong>of</strong> intact proteins on a column packed with 3µm C8 silicawith 1000Å pore size. The molecular weight <strong>of</strong> the protein examplesreach from ribosomal proteins (


advantage <strong>of</strong> using two separate 15 cm columns rather than one 30cm column is that sample matrix components that are not retained onthe first column (e.g. salts) can be directed to waste, instead <strong>of</strong> enteringthe nanospray source/mass spectrometer. We have investigated theeffect <strong>of</strong> increased resolution through doubling column length on thenumber <strong>of</strong> peptides/proteins identified in a digested cytosolic fraction<strong>of</strong> a human cell lysate using various gradient lengths. An ABSCIEXTripleTOF 5600 MS was used with an Information DependentAcquisition method consisting <strong>of</strong> a TOF MS survey scan at >30 000resolution, followed by 20 MS/MS in a second at >15 000 resolution.All data was processed using ProteinPilot S<strong>of</strong>tware 4.0 with integratedfalse discovery rate (FDR) analysis. 1 Remco van Soest*, David W.Neyer; Jia Eng Siow, and Phil Paul, Eksigent Technologies, Dublin, CA;poster ASMS 2008: “Improving resolution in nanoLC separations forproteomics using ultra high pressures”.211 Using 200 µm ID cHiPLC Columnsfor Increased Sample Throughput inPeptide QuantitationR. van Soest 1 , N. Hebert 1 , D.W. Neyer 1 ,J.B. Young 21Eksigent Technologies, Dublin, CA, USA; 2 ABSCIEX, Concord, ON, CanadaNan<strong>of</strong>low liquid chromatography coupled with nano-electrospray(nanoLC-MS) is the method <strong>of</strong> choice for sensitive peptide and proteinanalysis. We recently reported on a new micr<strong>of</strong>luidic platform (cHiPLCnan<strong>of</strong>lex)for nanoLC-MS applications. In addition to deliveringeasy-to-use, dead-volume-free connections to micr<strong>of</strong>luidic devices,the platform’s cHiPLC columns provide extreme column-to-columnseparation reproducibility. The column ID <strong>of</strong> choice for best sensitivityis 75 µm, using a flow rate <strong>of</strong> 200 nl/min. While providing excellentsensitivity, the low flow rate reduces sample throughput because<strong>of</strong> gradient delay in the nanoLC system itself, the autosampler valveand sample loop and the connecting tubing between injection valve,column and nanospray source. One way to address this is to use a largerID column at proportionally higher flow rate. While this will reducesensitivity when the injection volume is kept equal, delay times can begreatly reduced. In this presentation we will show data demonstratingthe throughput increase when using 200 µm vs. 75 µm ID cHiPLCcolumns while reporting as well on the sensitivity trade-<strong>of</strong>f, and theeffect there<strong>of</strong> on peptide quantitation.212 Flow Rate, Concentration andSaturation: Investigating theFundamentals <strong>of</strong> Sample-TrappedColumn InjectionC. Marshall-Waggett, H. Svobodova, A. Berg,G. ValaskovicNew Objective, Inc., Woburn, MA, United StatesSample trapped column injection is an injection strategy commonlyemployed in nanobore LC/MS based analysis <strong>of</strong> complex peptidemixtures. The approach <strong>of</strong> sample trapped column injection providesseveral inherent advantages. By effectively desalting and concentratingsamples on-line, sample traps improve analytical column longevityand throughput. Additionally, the ability to load samples onto a trapcolumn at a much higher flow rate than is feasible for a typical 75umID nanobore analytical column is a particularly attractive feature <strong>of</strong> thisapproach. Duty cycle can be significantly improved when the sampleloading flow rate is decoupled from the gradient flow rate. Here weinvestigate the role sample trap column injection has on analyteretention and overall chromatographic performance. Using a direct flownano-LC pump with the ability to deliver flow rates ranging from 50 nl/min to 20 ul/min coupled to an autosampler and commercially availablepeptide standards and protein digests, we evaluated the relationshipbetween flow rate, analyte concentration and analyte composition todetermine the effect on chromatographic performance.214 Spatial Proteomics: A New LC-MS/MS Tissue Imaging WorkflowProviding Protein Identities and TheirDistribution in TissueC. Lübbert, M. Schürenberg, M.Becker, R. Paape,D. SuckauBruker Daltonics, Bremen, GermanyMALDI-Imaging <strong>of</strong> proteins in tissue sections has established itself as apowerful new approach to biomarker discovery and histopathologicalresearch in recent years. However, the lack <strong>of</strong> direct identificationstrategies continues to be an obstacle preventing its broader use inProteomics studies. Initial studies that utilized in situ digestion followedby MALDI-MS/MS analysis typically provided 5-50 peptide IDs <strong>of</strong> only1-5 high abundant proteins. Here we introduce a novel proteomicstechnology that combines the spatial information with the routineidentification <strong>of</strong> proteins from tissue sections. Highly resolved proteindigests are generated by applying trypsin onto two subsequent tissuesections by supersonic nebulization. One <strong>of</strong> the sections is then analyzedby MALDI imaging mass spectrometry at up to 50 µm spatial resolutionyielding a list <strong>of</strong> 200-2000 peptide signals per image. Peptides areextracted from the other sections entire surface and submitted toroutine LC-MS/MS analysis, in our case using MALDI-TOF/TOF. Theidentified peptide list is then filtered by the peaklist from the image.All matching peaks in the image can then be assigned to a protein andthe co-localization <strong>of</strong> 2 or more tryptic peptides confirm their proteinassociation. We analyzed rat brain and testis/epididymis using the newSpatial Proteomics approach typically yielding peptide distributions atthe 50-100 µm level. In brain, more than 100 peptides were identifiedand more than 20 proteins localized without the need for MS/MSanalysis directly from the tissue. The intensity, co-localization <strong>of</strong> 2 ormore peptides and the degeneracy <strong>of</strong> molecular weight <strong>of</strong> peptideto-proteinmapping were used as primary validation tools beyondsignificant mascot scores from peptide identification. As an extension<strong>of</strong> the established top-down imaging strategy, this bottom-up SpatialProteomics approach may facilitate the identification and simultaneouslocalization <strong>of</strong> a much greater number proteins than it was previouslypossible.215 Increasing the Sample LoadingCapacity for Peptide Analysis by LC-MS/MS Using 150 µm ID Packed TipColumnsA. Berg, H. Svobodova, C. Marshall-Waggett,G. ValaskovicNew Objective, Inc., Woburn, MA, United StatesA predominant workflow for qualitative proteomics has been “GeLC-MS,” a combination <strong>of</strong> 1- (or 2-D) gel electrophoresis with reverse-phasenan<strong>of</strong>low liquid chromatography mass spectrometry (nLC-MS/MS). Thelimited protein quantity isolated from a single gel band coupled withPoster Abstracts<strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine • 91


Poster Abstractscolumn loading capacity maximums necessitate the use <strong>of</strong> 75 µm IDpacked columns for optimal sensitivity. However, limitations on sampleinjection volume, gradient and flow characteristics, and excessive delayvolume hinder throughput. Novel methods for fractionating complexbiological samples with higher loading capacities and more efficientrecovery, such as novel solution phase tube-gel fractionation andothers, demand a column format which maximizes on the extendeddynamic range <strong>of</strong> these emerging techniques. Packed tip columns witha larger ID (150 µm to 200 µm) facilitate higher sample loading capacityand enable higher flow rates for improved cycle time while maintainingthe optimal sensitivity realized in the nanobore packed tip colummformat. Using peptide standards, single protein digests and whole yeastdigests improvements in cycle time and sample loading capacity using150 µm ID packed tip columns are demonstrated.216 Comparing Global and Targeted Lipidand Fatty Acid Shotgun Pr<strong>of</strong>iling <strong>of</strong>Brain Tissue Extracts by NanoESI-InfusionJ. Albanese 1 , B. Simons 2 , G. Impey 2 , E. Duchoslav 2 ,K. Koisten 3 , K. Ekroos 31AB SCIEX, Foster City, CA, United States; 2 ABSCIEX, Concord, ON, Canada; 3 Zora Biosciences,Espoo, FinlandThe growth in lipidomics research is uncovering a need for completeand comprehensive workflows for identifying and quantifying lipidspecies from biological extracts. In choosing analytical methodsfor lipidomics, different yet complementary mass spectrometryapproaches can provide a more complete and comprehensive dataset leading to a detailed characterization <strong>of</strong> lipid molecular speciesfrom complex extracts. A preliminary strategy carried out as global“shotgun” tandem mass spectrometry by direct infusion electrosprayionization or LC-MS/MS analysis, uses information dependant MS/MSscanning in both polarities for unbiased lipid pr<strong>of</strong>iling. The secondapproach involves multiple lipid-class-specific precursor ion andneutral loss scanning whose resulting spectra can be used directly toidentify and characterize lipids and fatty acids in tissue-derived lipidextracts. The multiple precursor ion scanning (MPIS) methods havebeen published and recently reviewed by Ekroos et al, describing theadvantages <strong>of</strong> targeted MPIS techniques for generating comprehensivelipid arrays from small sample volumes. Fully characterizing these lipidcomponents by high quality MS/MS for fatty acid chain length anddouble bond positioning is a critical step for understanding theirbiological implications in cell signaling and lipid-initiated diseaseprogression. Taking advantage <strong>of</strong> the speed, selectivity, and sensitivity<strong>of</strong> hybrid triple quadrupole technology, whole lipid extracts fromrat brain tissue can be analyzed by direct nanoESI infusion for indepth glycerophospholipid pr<strong>of</strong>iling - achieving both qualitative andquantitative data (with the use <strong>of</strong> synthetic lipid internal standards) invery fast analysis times. Lipid species identification and quantitationis carried out using LipidViewTM S<strong>of</strong>tware enabling post acquisitionprocessing <strong>of</strong> precursor ion, neutral loss, MRM, and MS/MS data via lipiddatabase searching and accurate peak integration. We present robusttargeted and global workflows for the identification and quantitation <strong>of</strong>glycerphospholipids in total lipid extracts from rat brain tissue.217 Detection <strong>of</strong> p53 Phosphorylationand Oligomerization Using ProximityLigation AssayS.M. Chen, K. Huwiler, R. Bruinsma, B. Marks,B. Schweitzer, T. Settineri, M. Shannon, D. RuffApplied Biosystems, part <strong>of</strong> Life Technologies,Foster City, CA, United StatesThe tumor suppressor protein p53 is one <strong>of</strong> the most studiedarchitects <strong>of</strong> transcription control in cells. Recent reports detailcrucial tertiary structural requirements for p53 influence intranscription regulation complexes. These findings indicate thetetrameric assembly <strong>of</strong> p53 protein is essential for stabilizationand localization <strong>of</strong> the protein. This event is critical for DNA bindingand transcriptional activation. Post-translational modifications (PTM)have been linked to activation <strong>of</strong> p53 function in response to genotoxicstress. In particular, phosphorylation <strong>of</strong> the p53 Ser-15 is a critical eventin the genotoxic pathway cascade. We investigate the interconnectionbetween this PTM and p53 higher order structure formation. Weemploy a novel antibody binding assay scheme to elucidate details<strong>of</strong> the multimeric status <strong>of</strong> p53 in both recombinant p53 proteinexperiments and in cells undergoing genotoxic responses. Thisapproach utilizes the Proximity Ligation Assay (PLA) to simultaneouslyquery the role <strong>of</strong> Ser-15 phosphorylation and the multimeric state <strong>of</strong>p53. By use <strong>of</strong> a phospho-Ser-15 specific monoclonal antibody PLAprobe, we quantify proximity <strong>of</strong> two individual phospho-Ser-15 motifsin experimental samples. Our results demonstrate that phospho-Ser15promotes multimerization <strong>of</strong> p53 protein both in vitro and in cellsresponding to genotoxic stress. Further studies using PLA to explore thep53 protein-protein interactome should reveal a more complete view<strong>of</strong> the downstream interplay facilitated by the tetrameric conformation.218 Investigating the Robustness <strong>of</strong> a NewMicr<strong>of</strong>luidic DeviceC. Hughes, J. Langridge, T. McKennaWaters Corporation MS Technologies Centre,Manchester, United KingdomSuccessful assembly <strong>of</strong> the components used in nanoscale LC requiressome level <strong>of</strong> expertise in order produce good chromatographywith minimal band broadening.Micr<strong>of</strong>luidic devices, wherechromatographic separation is performed on a chip mounted close toa MS inlet, enable significant reduction in dead volumes and enableoptimal chromatography to be achieved by the novice user. Here, wedemonstrate the robustness and reproducibility <strong>of</strong> a Waters Nanotilecoupled to a new Time <strong>of</strong> Flight MS. We made 35 injections <strong>of</strong> an E.Coli tryptic digest, containing 50fmol spikes <strong>of</strong> four proteins, onto ananoAcquity UPLC system coupled to a Trizaic source.The Trizaicconsists <strong>of</strong> a ceramic tile containing a trapping column, analyticalcolumn and emitter. Trapping flow rate was set to 8uL/min for 1.8minsand the analytical reversed phase separation was performed at a flowrate <strong>of</strong> 450nL/min, changing the acetonitrile composition from 3 to40% over 90minutes, with 30minute re-equilibration. Emitter eluentwas analysed using a ToF MS operating in MSe mode. In this mode, theMS performed alternate low and elevated energy scans, the low energyproviding precursor ion information and the elevated energy providingfragment ion information. Data was processed using ProteinLynx GlobalServer where the ion information was collated using retention timeinformation in order to associate precursors and fragments. Data wasthen searched using a non-redundant database and the internal spike92 • <strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine


allowed absolute quantitation. Comparing chromatograms from variousstages <strong>of</strong> the 70-hour experiment shows excellent reproducibility. Theaverage number <strong>of</strong> E. Coli proteins identified was 389±17, in which325 replicated in 20 out <strong>of</strong> the 35 injections. Moreover, the absolutequantitation measurements using the ADH spike for identified proteinswith >3 peptides is 285±7ng and the quantitation measurement for one<strong>of</strong> the other protein spikes, Enolase, shows a CV <strong>of</strong> 7%.220 Identification <strong>of</strong> Various Pink-RedPigments Formed by Reacting VariousAmino Acids with Onion (allium cepaL.) Thiosulfinates Using HPLC withDiode Array Detector and TandemMass Spectrometry219 Effective Semi-Automated Extraction<strong>of</strong> Intact Mitochondria from SolidTissues Using Gentle MechanicalHomogenization and Pressure CyclingTechnologyV. S. Gross 1 , G. Carlson 1 , E. Freeman 2 ,A.R. Ivanov 2 , A. Lazarev 11Pressure BioSciences, Inc., South Easton, MA,United States; 2 HSPH Proteomics Resource, HarvardSchool <strong>of</strong> Public Health, Boston, MA, United StatesImpaired mitochondrial function has been linked to many diseases,such as stroke, heart disease, cancer, Type II diabetes and Parkinson’sdisease. Mitochondria-enriched preparations are needed forproteomic and metabolomic studies that may provide crucial insightsinto tissue-specific mitochondrial function and dysfunction, andanswer fundamental questions <strong>of</strong> cell energetic and oxidative stress.Mitochondria extractions from whole tissue samples are typicallyperformed using Potter-Elvehjem homogenizers or similar laborintensivemanual disruption methods that require extensive operatorexperience, and <strong>of</strong>ten result in damage to fragile organelles and highsample-to-sample variability. Here we describe a semi-automatedmethod that uses a novel gentle mechanical homogenizer (The PCTShredder) and hydrostatic pressure to release intact mitochondriafrom fresh rat tissues with minimal hands-on time. Pressure CyclingTechnology (PCT)-based tissue homogenization is conducted undercontrolled thermodynamic conditions (time, temperature and pressure)leading to more reproducible results. The quality <strong>of</strong> mitochondriapreparations was characterized by electron microscopy, 2D PAGE andrespiration assays. Our data demonstrate that mitochondria extractedby the PCT sample preparation system (PCT-SPS) are intact, functional,and exhibit a protein pattern comparable to control samples isolatedusing a conventional Potter-Elvehjem homogenizer. The resultingmitochondria-enriched samples were also subjected to trypsindigestion followed by nanoLC-MS/MS analysis on an LTQ-Orbitrap.Proteomic and pathway pr<strong>of</strong>iles <strong>of</strong> mitochondria samples preparedusing the novel extraction technique were compared to those extractedusing a conventional manual method to demonstrate the purity <strong>of</strong>mitochondrial preparations extracted using the novel PCT-SPS method.Y. Rezenom 1 , E. Lee 2 , K. Yoo 2 , D. Russell 1 , B. Patil 21Laboratory for Biological Mass Spectrometry,Department <strong>of</strong> Chemistry, Texas A&M University,College Station, TX, United States; 2 Vegetable& Fruit Improvement Center, Department <strong>of</strong>Horticultural Science, Texas A&M University,College Station, TX, United StatesDuring the processing <strong>of</strong> onion, pink-red colored pigments are <strong>of</strong>tenformed. The process is believed to be a multistep process includingenzymatic and non enzymatic reactions. In order to investigate thisprocess, we developed a reaction system, where pink-red pigments(‘pinking’) can be formed by reacting amino acids with onionthiosulfinate formed by reacting an isolated garlic alliinase and (+)-S-1-propenyl-L-cysteine sulfoxide (1-PeCSO) in the natural onion juice. Theunknown pink-red pigments formed during this process were separatedand detected using a high-performance liquid chromatography (HPLC)and a diode array detector (DAD) at 515 nm. Fractions collected fromthis separation were further analyzed using liquid chromatography(LC) and tandem mass spectrometry. Similar head group structure, two3,4-dimethyl pyrrolyl rings that were cross linked by allyl group, wasdetermined for all conjugate-pigments formed from different aminoacids based on the accurate and tandem mass spectrometry. However,the tail group attached to the N-terminal <strong>of</strong> pyrrole ring differed foreach pink-red pigment depending on the amino acid used. In addition,in most cases more than one pink compound were identified for thesame amino acid used. We presumed that the complexity <strong>of</strong> the pinkredpigments was due to the involvement <strong>of</strong> 21 natural amino acids andother derivatives <strong>of</strong> the products. Finally, we suggest that the pinkingprocess in crushed onion is very similar to the greening process incrushed garlic, emphasizing that thiosulfinates from flavor precursorsand free amino acids are absolutely necessary during the discoloration.221 Improvements in Data-DependentAcquisition for Enhanced ProteinIdentificationC. Miller, N. Kitagawa, W. Tang, J. Roark,J. Satulovsky, P. PerkinsAgilent Technologies, Palo Alto, CA, United StatesPoster AbstractsComprehensive proteome analysis can be very challenging due tocomplexity and range <strong>of</strong> protein concentrations. Techniques such as2D LC, pI-based fractionation and gel electrophoresis are typicallyemployed to increase separation efficiency as a strategy for obtainingmore peptide MS/MS spectra and thus increasing the number <strong>of</strong> proteinsidentified. In data-dependent mode, efficient and comprehensiveprotein identification depends on several conditions during theacquisition, among them: the number <strong>of</strong> precursors examined per unittime, selection <strong>of</strong> the most promising precursors for fragmentation,and application <strong>of</strong> the appropriate fragmentation conditions to yieldthe highest quality product ion spectrum during acquisition. This workdescribes changes to the precursor selection and fragmentation stepsto select the precursors most likely to produce good quality peptideMS/MS spectra, and to increase the quality <strong>of</strong> those spectra.<strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine • 93


Poster Abstracts222 UHR-Q-TOF Analysis Can AddressCommon Challenges in Targeted andUntargeted MetabolomicsA. Barsch 1 , G. Zurek 1 , D. Krug 2 , R. Müller 21Bruker Daltonics, Bremen, Germany; 2 Universitätdes Saarlandes, Saarbrücken, GermanyHere, we present an ESI-UHR-Q-TOF based analysis <strong>of</strong> myxobacterialsecondary metabolites, which permits to solve several challengesfrequently encountered in metabolite pr<strong>of</strong>iling studies. Myxobacteriaare promising producers <strong>of</strong> natural products exhibiting potentbiological activities, and several myxobacterial metabolites are currentlyunder investigation as potential leads for novel drugs. However, themyxobacteria are also a striking example for the divergence betweenthe genetic capacity for the production <strong>of</strong> secondary metabolitesand the number <strong>of</strong> compounds that could be characterised to date.Wildtype and mutant strains were analyzed concerning the productionpatterns <strong>of</strong> known metabolites and with regard to the discovery<strong>of</strong> new metabolites. Sample throughput: Since mass accuracy andresolution <strong>of</strong> TOF instruments are independent <strong>of</strong> the acquisition rate,they are perfectly suited for a coupling to UHPLC separations. Thesehyphenations enable a reduction <strong>of</strong> analysis time in combination witha high chromatographic resolution and therefore permit an increasedsample throughput. The UHR-TOF analysis revealed that an acquisitionrate <strong>of</strong> up to 20Hz did not compromise the achieved mass accuracy orresolution. Targeted and untargeted metabolite pr<strong>of</strong>iling: Acquisition <strong>of</strong>full scan accurate mass spectra enable the targeted screening for knowncompounds e.g. from the class <strong>of</strong> DKxanthenes based on very selectivehigh resolution EIC (hrEIC) traces with small mass windows <strong>of</strong> 1.0 - 0.5mDa. A comparison <strong>of</strong> several datasets following a “comprehensivefeature extraction” combined with a statistical analysis permits anuntargeted discovery <strong>of</strong> novel biomarkers using the same data filesas for the targeted analysis. Identification: Even a mass accuracy <strong>of</strong> 0.1ppm is not sufficient for an unambiguous formula identification for m/zvalues above 500. A combination <strong>of</strong> accurate mass data and isotopicpattern information in MS and MS/MS spectra can extend this m/z rangefor reliable formula suggestions. Examples for novel metabolites fromMyxobacteria will be shown.223 Metabolic Pr<strong>of</strong>iling <strong>of</strong> aCorynebacterium Glutamicum DeltaprpD2by GC-APCI High ResolutionQ-TOF AnalysisA. Barsch 1 , G. Zurek 1 , M. Persike 2 , J. Plassmeier 2 ,K. Niehaus 21Bruker Daltonik GmbH, Bremen, Germany;2Centrum für Biotechnologie, Universität Bielefeld,GermanyMetabolomics studies based on Gas chromatography -Massspectrometry (GC-MS) are well established and typically employelectron impact (EI) ionisation. Target compounds <strong>of</strong> interest canbe identified by comparison to commercial or public databases.Unfortunately, many possible biomarkers detected in metabolicpr<strong>of</strong>iling experiments cannot be identified due to the lack <strong>of</strong> referencespectra for a majority <strong>of</strong> biologically relevant compounds. Therefore,many possible biomarkers remain “unknowns” up till now. Hyphenatinggas chromatography with high resolution TOF-MS technology with s<strong>of</strong>tatmospheric pressure ionisation (APCI) can preserve the molecularion information and delivers accurate mass and isotopic patterninformation. This data enables a sum formula generation for knownand unknown target compounds.Additionally, optionally acquired MS/MS data can extend the capabilities for structural elucidation. Massaccuracy, resolution and isotopic fidelity are independent <strong>of</strong> the TOF-MS acquisition rate. Therefore, these instruments can be coupled to gaschromatography, which typically delivers narrow peak width requiringfast MS scan speeds. Corynebactrium glutamicum, a gram positive, nontoxicbacterium, is used in the industrial production <strong>of</strong> amino acids likelysine and glutamate. C. glutamicum can be grown on different carbonsources. Glucose is metabolised via glycolysis and the tricarboxylic acid(TCA) cycle, whereas propionate is catabolised through the methylcitricacid pathway. The prpD2 gene encodes a 2-methylcitrate dehydratasewhich is involved in the degradation <strong>of</strong> propionate. Metabolic pr<strong>of</strong>iling<strong>of</strong> Corynebacterium glutamicum delta-prpD2 extracts <strong>of</strong> cells grownon glucose or glucose and propionate analyzed by GC-APCI-TOF-MSrevealed several compounds elevated in cells grown on propionate.Identification <strong>of</strong> 2-methylcitric acid and alanine using accurate massand isotopic pattern information in MS and MS/MS spectra provided apro<strong>of</strong> <strong>of</strong> concept for the identification <strong>of</strong> target compounds using highresolution MS technology.224 Biomarker Discovery Using NewMetabolomics S<strong>of</strong>tware forAutomated Processing <strong>of</strong> HighResolution LC-MS DataD. Peake 1 , S. Hnatyshyn 2 ; M. Reily 2 ; P. Shipkova 2 ;T. McClure 1 ; M. Sanders 11Thermo Fisher Scientific, San Jose, CA, UnitedStates; 2 Bristol Myers Squibb, Princeton, NJ, UnitedStatesRobust biomarkers <strong>of</strong> target engagement and efficacy are required indifferent stages <strong>of</strong> drug discovery. Liquid chromatography coupledto high resolution mass spectrometry provides sensitivity, accuracyand wide dynamic range required for identification <strong>of</strong> endogenousmetabolites in biological matrices. LCMS is widely-used tool forbiomarker identification and validation. Typical high resolution LCMSpr<strong>of</strong>iles from biological samples may contain greater than a millionmass spectral peaks corresponding to several thousand endogenousmetabolites. Reduction <strong>of</strong> the total number <strong>of</strong> peaks, componentidentification and statistical comparison across sample groups remainsto be a difficult and time consuming challenge. Blood samples fromfourgroups <strong>of</strong> rats (male vs. female, fully satiated and food deprived)were analyzed using high resolution accurate mass (HRAM) LCMS.All samples were separated using a 15 minute reversed-phase C18LC gradient and analyzed in both positive and negative ion modes.Data was acquired using 15K resolution and 5ppm mass measurementaccuracy. The entire data set was analyzed using s<strong>of</strong>tware developedin collaboration between Bristol Meyers Squibb and Thermo FisherScientific to determine the metabolic effects <strong>of</strong> food deprivationon rats. Metabolomic LC-MS data files are extraordinarily complexand appropriate reduction <strong>of</strong> the number <strong>of</strong> spectral peaks viaidentification <strong>of</strong> related peaks and background removal is essential. Asingle component such as hippuric acid generates more than 20 relatedpeaks including isotopic clusters, adducts and dimers. Plasma andurine may contain 500-1500 unique quantifiable metabolites. Noisefiltering approaches including blank subtraction were used to reducethe number <strong>of</strong> irrelevant peaks. By grouping related signals such asisotopic peaks and alkali adducts, data processing was greatly simplifiedby reducing the total number <strong>of</strong> components by 10-fold. The s<strong>of</strong>twareprocesses 48 samples inunder 60minutes. Principle ComponentAnalysis showed substantial differences in endogenous metabolites94 • <strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine


levels between the animal groups. Annotation <strong>of</strong> components wasaccomplished via searching the ChemSpider database. Tentativeassignments made using accurate mass need further verification bycomparison with the retention time <strong>of</strong> authentic standards.225 On-Line Electrochemistry/MS - APowerful Tool for Rapid Prediction <strong>of</strong>Phase I and II Drug MetabolismJ. Powers 1 , J. Purkerson 1 , A. Kraj 2 , M. Eysberg 2 ,J.P. Chervet 21Antec, Palm Bay, FL, United States; 2 Antec,Zoeterwoude, The NetherlandsThe use <strong>of</strong> Electrochemsiry is a complementary approach to traditionalmethods such as in vivo (human, rodent) or in vitro (liver microsomes)metabolism studies, and delivers the oxidative metabolic fingerprint <strong>of</strong>a (drug) molecule in a very short time. The acquired mass spectra arepresented in simple 2 dimensional or more illustrative 3 dimensionalplots, so-called MS voltammograms. A MS voltammogram visualizesthe ion abundance versus m/z as a function <strong>of</strong> applied potential to theelectrochemical cell. With a MS voltammogram the optimal potentialcan be determined for electrochemical generation <strong>of</strong> the desiredmetabolite for further research, e.g., a phase II metabolism study (i.e.adduct formation). It is a quick method to identify reactive pathways<strong>of</strong> the compound <strong>of</strong> interest. Additionally, electrochemistry allows totrace the reactive metabolite conjugates with targets (e.g., proteins,glutathione) without matrix interactions in contrary to traditionalmethods. A dedicated s<strong>of</strong>tware program has been developed toautomate and simplify the MS voltammogram acquisition. The programcontrols the syringe pump, the potentiostat and triggers the acquisition<strong>of</strong> mass spectra at the designated cell potentials. The total acquisitiontime needed for recording <strong>of</strong> a full MS voltammogram can be as shortas 5 minutes. Amodiaquine an anti malaria agent was chosen as one <strong>of</strong>the model drugs to investigate oxidative metabolism using the on-lineEC/MS system with automated MS voltammogram acquisition The easyand fast Electrochemical conversion <strong>of</strong> Amodiaquine into its 4 majorphase I metabolites will be presented. In a second step Glutathione(GSH) is added to the electrochemically generated metabolites to formthe appropriate GSH-metabolite adducts, mimicking phase II reactions.All known adducts were successfully formed and identified with MS.Additionally, MS voltammograms <strong>of</strong> other drugs and xenobiotics (e.g.,acetaminophen, amiodarone, irinotecan) are presented. The datademonstrate that hyphenation <strong>of</strong> electrochemistry with electrospraymass spectrometry provides a versatile and user-friendly platformfor rapid and cost efficient screening <strong>of</strong> target compounds (drugs,xenobiotics, etc.) in phase I and phase II metabolomics studies.226 Facile Solid-Phase Synthesis <strong>of</strong>Peptide-p-Nitroanilide (pNA) AnalogContaining Conjugates Using a NovelWang or Rink Amide ResinX. Wang 1 , C. Po 2 , J. He 2 , A. Hong 21AnaSpec, Fremont, CA, United States; 2 EurogentecGroup, Fremont, CA, United StatesProteases play a key role in literally all biological processes, and are <strong>of</strong>great interest, especially to the pharmaceutical industry. Colorimetricbased Peptide-p-Nitroanilide conjugates (peptide-pNAs), withabsorbance at approximately 408 nm, have historically been and arestill widely used substrates for the study <strong>of</strong> protease activity. Thepreparation <strong>of</strong> peptide-pNA however, presents several technicalchallenges. Firstly, the amino group <strong>of</strong> pNA has a low nucleophilicproperty due to the electron-withdrawing effect <strong>of</strong> the nitro group.Secondly, the poor solubility <strong>of</strong> a p-nitroanilide intermediate and lastly,coupling in solution phase by DCC, azide or active ester, commonlyused techniques are not effective. Here we report the development<strong>of</strong> two novel supports for facile solid phase peptide syntheses,namely, Wang-resin and Rink Amide-resin conjugated with a pNAanalog, 5-amino-2-nitrobenzoic acid (Anb5,2). Based on a paper byHojo, et al. in which they described the introduction <strong>of</strong> Anb5,2 to ap-methylbenzhydrylamine (MB) resin; we successfully coupled Anb5,2to either Wang or Rink Amide resin using the TBTU method in thepresence <strong>of</strong> p-dimethylaminopyridine (DMAP). Anb5,2-Wang or RinkRmide resin is then coupled to a Fluorenylmethyloxycarbonyl (Fmoc)containing amino acid. Peptide synthesis can subsequently proceedusing Fmoc synthesis strategy. The use <strong>of</strong> this pNA analog containingresins circumvents the tehcnical difficulties stated above. These resinsalso greatly facilitates the synthesis <strong>of</strong> peptide-pNA-like chromogenicsubstrates for protease research.**227 Validation <strong>of</strong> an Ion MobilityHydrogen/Deuterium Exchange MassSpectrometry SystemK. Fadgen 2 , T. Wales 1 , M. Stapels 2 , M. Eggertson 1 ,J. Engen 21Northeastern University, Boston, MA, UnitedStates; 2 Waters Corporation, Milford, MA, UnitedStatesHydrogen/deuterium exchange mass spectrometry (HXMS) has provento be a useful analytical method for the study <strong>of</strong> protein dynamics andchanges to protein conformation. Rapid chromatographic separationsat 0°C must be utilized to preserve the deuterium label during LC/MS analysis. Unfortunately, fast, low temperature LC separations arenot very efficient and can cause spectral overlap for large proteins.The addition <strong>of</strong> a gas phase, ion mobility separation (IMS) into theHXMS workflow inserts an orthogonal separation that occurs on themillisecond timescale after the LC step without causing any detrimentalimpact on analysis <strong>of</strong> deuterium levels. An improved MS system hasrecently been developed that is capable <strong>of</strong> higher resolution gas-phasemobility separations. The improved MS platform was combined witha fully automated ultra performance liquid chromatography (UPLC)system developed for HXMS. To evaluate the effect <strong>of</strong> the new MSplatform on deuterium recovery, HXMS experiments were performedusing glycogen phosphorylase B as a model protein. Replicate studies<strong>of</strong> deuterium labeling reactions ranging from 10 seconds to 100minutes were evaluated. No significant deviation in deuterium levelswas observed when using either mobility (HDMSE) or non-mobility(MSE) separations and the standard deviation <strong>of</strong> deuterium uptakefor replicate analyses, for HDMSE and MSE separations were in goodagreement with one another. Using the newly developed protocol, anHXMS study <strong>of</strong> a monoclonal antibody was performed to comparedeuterium recovery using a 6 or 12 minute chromatographic separation.Even though chromatographic resolution was reduced with the fastergradient, HDMSE allowed several overlapping peptides to easily beinterrogated. In addition, the shorter analysis time improved deuteriumrecovery for the sample. The application <strong>of</strong> this additional level <strong>of</strong>separation will be essential in future studies <strong>of</strong> very large proteins inwhich chromatographic efficiency is expected to be suboptimal.Poster Abstracts<strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine • 95


228 Differential Expression <strong>of</strong> Proteins inLung Cancer Using Difference in GelElectrophoresis (DIGE)F. Masri 1 , P.Beckett 2 , K.S. Aulak 21University <strong>of</strong> Kalamoon, Faculty <strong>of</strong> Pharmacy,Syria; 2 Amersham Biosciences, Cleveland ClinicFoundation, Cleveland, OH, United Statesfocusing patterns between the two conditions is clearly visualized withthe Bioanalyzer protein assay. The combination <strong>of</strong> OFFGEL and lab-onchipprotein analysis allows separation and identification <strong>of</strong> is<strong>of</strong>ormsbased on pI and molecular weight. Protein is<strong>of</strong>orms differing only bya few kDa in apparent molecular weight were successfully separatedand enriched by OFFGEL electrophoresis for further downstreamanalysis by LC/MS-MS. The combination <strong>of</strong> OFFGEL, Bioanalyzer,and mass spectrometry is thus an efficient combination for detailedcharacterization <strong>of</strong> recombinant proteins such as mAb’s.Poster AbstractsBackground: Lung cancer remains the leading cause <strong>of</strong> cancer-relatedmortality worldwide. Early detection <strong>of</strong> lung cancer is problematic dueto the lack <strong>of</strong> a marker with high diagnosis sensitivity and specificity.The purpose <strong>of</strong> this study was to develop techniques to identify thedifferential expression protein pr<strong>of</strong>iles between tumor and tumorfree<strong>of</strong> lung cancer tissues. Methods: 2-dimensional differential ingelelectrophoresis (2D-DIGE) and matrix-assisted laser desorption/ionization time-<strong>of</strong>-flight mass spectrometry (MALDI-TOF-MS) wereused to analyze four samples <strong>of</strong> lung cancer tissue (3 replicates each).Results: From optimized 2DE image, A total <strong>of</strong> 2561 spots were detectedand 427 spots <strong>of</strong> these were differentially expressed (p


MuDPIT, and <strong>of</strong>f-gel electrophoresis (OGE) ahead <strong>of</strong> LC-MS/MS.We have evaluated an alternative method that does not requireadditional sample fractionation on additional equipment, meanwhileminimizing alterations introduced by analytical techniques. For bothwhole human cells and isolated mitochondria, the proposed methodinvolved exposure <strong>of</strong> the pelleted cells or organelles to sequencegradetrypsin in order to obtain peptides and proteins cleaved fromthe cell and organelle surfaces. The “stripped” cells and mitochondriawere also lysed and their contents digested. Additionally, total lysates<strong>of</strong> cells and mitochondria not exposed to trypsin ahead <strong>of</strong> lysis weregenerated and tryptically digested. The obtained proteomic pr<strong>of</strong>ilingresults were compared to those acquired using commonly employedfractionation techniques (OGE and SDS-PAGE). Resulting digests fromall protocols were analyzed using LC-MS/MS, and all were analyzedfor the proportion <strong>of</strong> unique and differentially enriched proteinsachieved with each. Our results showed that the inexpensive andsimplistic fractionation method allowed for similar or superior depth<strong>of</strong> proteomic pr<strong>of</strong>iling as well as a substantial increase in the number<strong>of</strong> unique identifications <strong>of</strong> membrane-bound and extracellular matrixrelated proteins. Additionally, the method provides information aboutspatial localization and differential abundance <strong>of</strong> proteome subsetsin the cell or organelle architectural compartments. These resultssuggest that a very straightforward approach for the preparation <strong>of</strong>cellular and organellar specimens prior to proteomic characterizationcan be reproducibly and reliably utilized by biological and medicallaboratories that have no previous analytical biochemistry expertise.232 Detecting Immune System ResponseProteins in a 500 Year-old IncaMummyA. Koller 6 , A. Corthals 1 , L. Davalos 2 ,D.W. Martin 3,4 , R. Rieger 3 , E.I. Chen 3,51City University <strong>of</strong> New York, John Jay College <strong>of</strong>Criminal Justice, Department <strong>of</strong> Sciences, New York,NY, United States; 2 Department <strong>of</strong> Ecology andEvolution and Consortium for Inter-DisciplinaryEnvironmental Research, State University <strong>of</strong> NewYork Stony Brook, Stony Brook, NY, United States;3Stony Brook Proteomics Center, State University<strong>of</strong> New York Stony Brook, Stony Brook, NY, UnitedStates; 4 Department <strong>of</strong> Medicine, Division <strong>of</strong>Hematology, State University <strong>of</strong> New York StonyBrook, Stony Brook, NY, United States; 5 StateUniversity <strong>of</strong> New York Stony Brook, Department <strong>of</strong>Pharmacological Sciences, Stony Brook, NY, UnitedStates; 6 State University <strong>of</strong> New York Stony Brook,Stony Brook, NY, United StatesDisease detection in ancient human samples currently relies ongenomic-based assays, which are error prone due to contamination andcannot distinguish between active and latent pathogenic infection. Onthe other hand, protein-based assays such as global protein pr<strong>of</strong>iling<strong>of</strong>fer complementary alternatives for the pathological diagnosis <strong>of</strong>archeological specimen. The discovery <strong>of</strong> three Inca mummies in 1998,perfectly preserved in the permafrost <strong>of</strong> the high Andes, allowed us toanalyze mummy samples by protein-based and genomic-based assay.A buccal swab from one <strong>of</strong> the 500 year old mummy was analyzed byshotgun proteomics to detect the protein pr<strong>of</strong>ile. Among the identifiedproteins, we found a signature <strong>of</strong> proteins indicating an immuneresponse to a bacterial infection at the time <strong>of</strong> the mummy’s death.Based on the external visible symptoms and the gamut <strong>of</strong> immuneresponse proteins obtained from the mouth swab, we suspectedthat the pulmonary infection was caused by Mycobacterium. PCRassay followed by direct sequencing <strong>of</strong> the PCR products confirmedthe presence <strong>of</strong> Mycobacterium sp. in the mouth swab. Until now,immunoassays have been the only way to detect an active immuneresponse and infer infection in historical samples, but these wereplagued by low specificity and sensitivity. However, we demonstratehere the feasibility <strong>of</strong> incorporating global protein pr<strong>of</strong>iling in thediagnosis <strong>of</strong> infection from archeological samples. Protein signaturesobtained from these samples could be extremely useful in determiningthe status <strong>of</strong> infection while genomic-based assays can be used todetect the identity <strong>of</strong> the pathogen.233 A New Technology for HighlyEfficient and Reproducible AlbuminDepletion from Whole SerumC. Bolcato, H. Anderson, M. Maust, G. Boyce,M.J. PowellProtea Biosciences, Morgantown, WV, United StatesOne <strong>of</strong> the major problems faced in the analysis <strong>of</strong> the human serumproteome is the broad dynamic range <strong>of</strong> its protein constituents.High abundance proteins, such as human serum albumin (HSA) andgamma-immunoglobulin (IgG), which together comprise 75% <strong>of</strong> totalserum protein content, inhibit the analysis <strong>of</strong> lower abundant proteins<strong>of</strong> interest. Affinity-based depletion strategies are <strong>of</strong>ten employed toselectively remove these high abundance proteins in order to enrichthe lower abundant proteins and facilitate their analysis. Traditionally,antibody-based schemes are used to produce affinity-binding ligandsfor HSA and IgG. Variability in the production <strong>of</strong> these antibodies leadsto a wide variety <strong>of</strong> specificities and selectivities. Thus, the efficienciesfor antibody-based depletion strategies for the selective removal<strong>of</strong> high abundance proteins in human serum can vary broadly, from70 to 95%, and can dramatically affect the reproducibility <strong>of</strong> humanserum proteome studies. Here, a new approach to affinity-based serumalbumin removal is presented. Instead <strong>of</strong> utilizing a traditional antibodybaseddepletion approach, the ProteaPrep albumin depletion captureligand is a recombinant bacterial protein that is highly purified forrobust, efficient removal <strong>of</strong> albumin from human serum samples. Therecombinant ligand technology produces binding constants withhuman serum albumin (Kd ~ 1 &times; 10-11M) that are significantlystronger than antibody-based methods, which have binding constantsthat range from Kd’s <strong>of</strong> 1 &times; 10-6 to 1 &times;10-8 M. The resultsshow that the functionalized beads bind and remove albumin fromserum in a highly efficient and rapid manner. Depletion efficiencies are>99% for the selective removal <strong>of</strong> HSA from serum samples in less than20 minutes. The recombinant bacterial protein ligands are shown to beeffective for the depletion <strong>of</strong> albumin from the sera <strong>of</strong> other species,including mouse, rat, pig, horse, cow, and monkey.234 Qualitative and QuantitativeCharacterization <strong>of</strong> Proteomes UsingIon Mobility Mass Spectrometry withData Independent AcquisitionM. Stapels, K. Fadgen, S. Geromanos, J. LangridgeWaters Corporation, Milford, MA, United StatesA central goal in a proteomics study is to fully characterize a sample bothqualitatively and quantitatively. A data-independent analysis yieldsreproducible fragmentation and peak area information for peptides inPoster Abstracts<strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine • 97


Poster Abstractsa complex mixture (1). The addition <strong>of</strong> ion mobility into this analysisinserts an orthogonal, gas-phase separation, occurring in the millisecondtimescale, which is poised nicely between chromatographic and TOFmass spectrometry timescales. Peptides and their correspondingfragment ions are aligned by chromatographic retention time in dataindependentanalyses. When ion mobility is used, peptides and theirfragment ions also share the same mobility drift time which dramaticallyimproves specificity. In this study, eukaryotic and prokaryotic sampleswere analyzed by one-dimensional (1D) and 2D chromatography usinghigh-low pH RP-RP separations, both with and without ion mobilityseparation. The number <strong>of</strong> proteins identified in a sample doubled byusing a five fraction 2D separation compared to 1D chromatography.Utilizing the mobility separation yielded an additional increase inthe number <strong>of</strong> proteins and peptides by at least 20%, without anyadditional instrument time. The amount <strong>of</strong> information obtained from asample depended on the amount loaded as well as the time dedicatedto analysis <strong>of</strong> the sample. For one prokaryotic sample, 552 proteinswere reproducibly identified with 1D chromatography without ionmobility from 0.75 ug <strong>of</strong> material. Loading 5 ug and performing 5fractions, along with ion mobility, yielded 1260 reproducible proteinidentifications. Stoichiometry between proteins was determined bycomparing the average intensity <strong>of</strong> the top three peptides to everyprotein to that <strong>of</strong> an internal standard (2). Agreement was foundbetween the experimental stoichiometric ratios and those found inliterature for many protein groups. 1. Geromanos, S.J., et. al. Proteomics.2009 (6):1683-95. 2. Silva, J.C., et. al. Mol Cell Proteomics. 2006 (1):144-56.235 Characterization <strong>of</strong> UnexpectedBy-Products in a Recombinant TAUProtein by Capillary LC Coupled toMALDI-Top-Down-Sequencing (LC-MALDI-TDS)A. Asperger 2 , B. Kovacech 1 , A. Kovac 1 , D. Suckau 21Institute <strong>of</strong> Neuroimmunology <strong>of</strong> Slovak Academy<strong>of</strong> Sciences, Bratislava, Slovakia; 2 Bruker Daltonics,Bremen, GermanyCompared to Edman sequencing, MALDI-TOFMS based top-downsequencing <strong>of</strong> proteins (MALDI-TDS) is much faster, cheaper, candeliver significantly longer sequence readout from both, protein N-and C-terminus, and has no limitations in case <strong>of</strong> terminally modifiedproteins. These features make MALDI-TDS an extremely appealingmethod for the QC analysis <strong>of</strong> recombinant protein products, forinstance biopharmaceuticals. Direct MALDI-TDS analysis requirespurified, homogeneous protein samples. However, coupling MALDI-TDS to upfront LC separation allows to analyze mixtures <strong>of</strong> proteins.Since recombinant protein products <strong>of</strong>ten contain unexpectedheterogeneities, LC-MALDI-TDS represents a promising methodto characterize such undesired by-products. We describe here thecharacterization by LC-MALDI-TDS <strong>of</strong> unexpected by-productsoccurring in a recombinant TAU protein. Initial MALDI-TOF analysis<strong>of</strong> the protein sample revealed at least two minor components beingdifferent from the expected protein mass. To separate these sampleconstituents, capillary LC was used. The LC fractions were depositedon a MALDI target pre-spotted with sDHB matrix. Subsequently,MALDI-ISD spectra <strong>of</strong> the LC separated compounds were acquiredusing a Bruker Ultraflex III MALDI-TOF/TOF. Analysis <strong>of</strong> the MALDI-TDS data was performed using Bruker’s BioTools s<strong>of</strong>tware. By means <strong>of</strong>LC-MALDI-TDS, one <strong>of</strong> the by-products, detected at 16.6kDa, turnedout to represent a truncated version <strong>of</strong> the expected TAU protein,containing the sequence range [1-161]. C-terminal ISD fragmentsyielded unambiguous evidence for the correct assignment <strong>of</strong> thetruncation site. Another unexpected by-product, being approx. 130Dalower in mass than the expected protein product, was lacking theN-terminal methionin residue. This was concluded from the assignment<strong>of</strong> the respective N-terminal fragments in the ISD spectrum. As shownabove, LC-MALDI-TDS turned out to be very efficient with regard toanalysis time and lab work required, especially when comparing themethod to alternative approaches like Edman sequencing or geLCworkflows, i.e. SDS-PAGE with subsequent LC-MS/MS analysis <strong>of</strong> thedigested proteins.236 Novel Proteomic Analysis <strong>of</strong>Esophageal InflammationK. Jonscher 1 , A. Kendrick 1 , S. Fillon 2 ,Z. Robinson 2 , J. Masterson 2 , S.J. Ackerman 3 ,G.T. Furuta 21Department <strong>of</strong> Anesthesiology, University <strong>of</strong>Colorado Denver, Aurora, CO, United States;2Department <strong>of</strong> Pediatrics, University <strong>of</strong> ColoradoDenver, United States; 3 Digestive Health Institute,University <strong>of</strong> Illinois at Chicago, Chicago, IL, UnitedStatesIntroduction: Esophageal inflammation associated with eosinophilicesophagitis (EoE) and gastroesophageal reflux disease (GERD) requireinvasive endoscopy and biopsy for diagnosis and testing efficacy <strong>of</strong>treatments. In the 1970’s, the Enterotest, a weighted gelatin capsulefilled with and attached to a 90 cm long nylon string, was introduced as aminimally invasive method to identify intestinal infections. Patients tapethe string end extruding from the capsule to their cheek and swallow thecapsule; the string subsequently deploys into the small intestine and thecapsule released. After incubation for 15 min to overnight the string ispulled back out <strong>of</strong> the mouth. Here we present pilot data demonstratingthat secretions adhering to the string may be recovered and analyzedusing shotgun proteomics. Methods: Strings from the distal esophagus<strong>of</strong> patients with GERD and normal controls (n=2/group) were cut into 2cm sections and proteins recovered by incubating strings in lysis buffer.Tandem mass spectrometry analysis using a high-capacity quadrupoleion trap was performed on trypsin-digested samples and data weresearched with X!Tandem through the LABKEY interface. Spectralcounting provided relative quantitation <strong>of</strong> proteins between samples.Results: After removal <strong>of</strong> is<strong>of</strong>orms, 34 proteins common to all stringsamples (2 each from 2 patients/subjects) were identified. Annexin A1,keratins 16, 4 and 13, and MUC5B were most highly upregulated inesophageal secretions from GERD subjects as compared with normalcontrols. In addition, functional clustering revealed that anti-apoptoticproteins and those involved in extracellular signaling were present inonly GERD and not normal controls. Conclusion: The Esophageal StringTest can be used to sample esophageal secretions in health and disease.Host response proteins are present in the esophageal lumen duringinflammation. This work was supported by R21-AI079925, ThrasherFoundation, CURED Foundation, Pappas Foundation and the ColoradoClinical Translational Scientific Institute (CCTSI) NIH/NCRR GrantNumber 5UL1RR026314-02.98 • <strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine


237 Multi-Site Assessment <strong>of</strong> ProteoRedPlasma Reference Sample forBenchmarking LC-MS PlatformPerformance238 In Depth Characterization <strong>of</strong> ProteinChanges Induced by GlucoseLimitation Using ComplementaryIonization TechniquesA. Campos 1 , K. Asanov 1 , E. Oliveira 1 , N. Colome 2 ,S. Martínez-Bartolomé 3 , J-P Albar 3 , F. Canals 2 ,ProteoRed ISCIII Consortium 41Proteomics Platform, Barcelona Science Park,Barcelona, Spain; 2 Proteomics Laboratory,Medical Oncology Research <strong>Program</strong>, Vall dHebron Institute <strong>of</strong> Oncology VHIO Vall d HebronUniversity Hospital, Barcelona, Spain; 3 CentroNacional de Biotecnologia-CSI, Madrid, Spain;4ProteoRed ISCIII Consortium, Madrid, SpainOne <strong>of</strong> the missions <strong>of</strong> the Spanish Proteomics Network (ProteoRedISCIII) is to assist its proteomics core facilities in evaluating theircapabilities to perform qualitative and quantitative proteomicsanalysis. In 2010, the ProteoRed’s Sample Collection and HandlingGroup designed a moderately complex plasma standard referencesample primarily to be used for routine quality assurance monitoring<strong>of</strong> laboratory instrumentation, as well as inter-laboratory performanceassessment, and development and validation <strong>of</strong> novel technologies.The ProteoRed Plasma Reference (PPR) sample is a subset <strong>of</strong> highlyabundant well-characterized human plasma proteins with a number<strong>of</strong> is<strong>of</strong>orms, in addition to 4 spiked-in proteins, altogether distributedover 5 orders <strong>of</strong> magnitude in concentration. The PPR sample wasrecently stress tested in the latest ProteoRed Multicenter Experiment(PME6) that counted with the participation <strong>of</strong> 17 proteomics facilitiesusing a wide range <strong>of</strong> LC-MS platforms. Although each laboratorywas allowed to use its own favorite methodology, we requested thesample be analyzed in a single LC-MS run in experimental triplicate(3 different digestions).Evaluation <strong>of</strong> the results submitted by thestudy participants revealed moderate discrepancies at the peptideidentification level, and poor overlap at the protein identificationlevel. In an attempt to identify the source <strong>of</strong> such irreproducibility,raw data <strong>of</strong> 8 laboratories (24 LC-MS runs) were reanalyzed centrallyusing a standardized data analysis pipeline, which included proteininference using ProteinProphet s<strong>of</strong>tware. We found that the majority<strong>of</strong> protein identification discrepancies across submitted reports <strong>of</strong>these 8 laboratories were due to inconsistencies on how data analystsand computational tools group and/or infer proteins. Immunoglobulinvariable chain identifications were particularly conflicting throughoutidentification lists, even in the centralized analysis. Using a series <strong>of</strong>LC-MS performance metrics, we benchmarked the performance <strong>of</strong> 8LC-MS instruments (Orbitraps) and identified system components thatvary the most across laboratories.B. Müller 1 , W. Jabs 2 , M. Behrens 2 , D. Suckau 2 ,C. Baessmann 2 , K. Niehaus 3 , T. Noll 11Institute <strong>of</strong> Cell Culture Technology, Faculty<strong>of</strong> Technology, Bielefeld University, Bielefeld,Germany; 2 Bruker Daltonik GmbH, Bremen,Germany; 3 Proteome and Metabolome Research,Faculty <strong>of</strong> Biology, Bielefeld University, Bielefeld,GermanyComprehensive quantification <strong>of</strong> specific changes in biological systemsin response to a certain treatment or perturbation is one <strong>of</strong> the mostimportant but also among the most challenging tasks in proteomics.Different kinds <strong>of</strong> separation techniques, mass spectrometer types,identification strategies, and quantification strategies can be appliedfor this purpose. Here, we investigate the benefits <strong>of</strong> combining theresults obtained by two main ionization techniques used in proteomics,ESI and MALDI. Results are derived with the aid <strong>of</strong> a bioinformaticsplatform, which integrates different proteomics techniques. Humancell cultures obtained from the periventricular region <strong>of</strong> the brainwere grown under normal and under glucose limitation conditions.To investigate the changes induced by the glucose limitation, stableisotope labeling with amino acids in cell culture (SILAC) was used.After labeling, samples were mixed and fractionated. 40&micro;g <strong>of</strong>the cytosolic protein fraction was separated by 1D-SDS PAGE, whichwas cut into 10 bands. Each band was digested and then analyzed withnanoscale liquid chromatography (nLC) coupled to an ESI-ion trapand a MALDI-TOF/TOF, respectively. The use <strong>of</strong> ESI-MS in additionto MALDI-MS resulted in a 46% increase in protein identificationand a 124% increase in peptide identification. Combining data <strong>of</strong>both ionization techniques, 664 proteins consisting <strong>of</strong> 4658 peptideswere identified. Peptides identified with ESI-MS are significantly morehydrophobic than peptides identified with MALDI-MS. An analysis<strong>of</strong> the amino acid composition <strong>of</strong> the identified peptides revealed asignificant bias <strong>of</strong> ESI-MS for aliphatic and hydroxyl-containing aminoacids while MALDI-MS favors peptides with aromatic, basic and amidegroup-containing amino acids. The results show, that the use <strong>of</strong> nLC-ESI-MS in addition to nLC-MALDI-MS greatly improves the number<strong>of</strong> identified and quantified proteins and peptides. The experimentrevealed little changes in the cytosolic proteome <strong>of</strong> cells growing underthe applied glucose limitation.239 Evaluation <strong>of</strong> Two SeparationTechniques, SCX and OFFGEL and<strong>of</strong> Two Fragmentation Methods,CAD and PQD, to Asses iTRAQQuantitation EfficiencyPoster AbstractsG. Grigorean 1 , A. DiFonzo 2 , P. S<strong>of</strong>fientini 2 ,T. Manousidou 21European Institute <strong>of</strong> Oncology, IFOM-IEOCampus, Milan Italy; 2 Cogentech, IFOM-IEOCampus, Milan, ItalyTo evaluate various sample preparation and analysis platforms forisobaric tags for iTRAQ experiments, we compared two separationand two fragmentation techniques: Strong Cation Exchange (SCX) with<strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine • 99


Poster Abstractsisoelectric point separation. In addition, we compared two types <strong>of</strong>mass analyzers: a quadrupole and a linear ion trap, the former withina quad-TOF hybrid mass spectrometer, and the later within an LTQ-FThybrid MS. This was done by comparing two fragmentation techniques,collisionally activated dissociation (CAD) with pulsed Q dissociation(PQD). Proteins from lysed HeLa cells were trypsinized and peptideswere iTRAQ labeled. Half <strong>of</strong> the labeled digest was separated via SCXinto20 fractions. The other half <strong>of</strong> digested peptides was separatedwith an OFFGEL isoelectric point fractionator into 24 fractions. Eachdried peptide fraction was desalted, and submitted to reverse-phaseC-18 HPLC on-line with the mass spectrometer. Quantitative dataanalysis for the LTQ-FT data was performed using Proteome Discoverer(Thermo Electron) and for the QStar data, PEAKS Studio (BioinformaticSolution Inc.). R s<strong>of</strong>tware was used for the statistical validation <strong>of</strong>results. The LTQ-FT data was also analyzed with the PEAKS s<strong>of</strong>twareto asses the viability <strong>of</strong> PEAKS as a universally applicable data analysispackage. Comparison between separation and dissociation was basedon a) number <strong>of</strong> proteins quantified b) reproducibility <strong>of</strong> technicalreplicates based on protein distribution c) standard deviation for dataobtained from the two dissociation techniques. Fractionation withOFFGEL increased the number <strong>of</strong> identified and quantified <strong>of</strong> proteinsby approximately 20%. Number <strong>of</strong> mass spectra which can be used forquantitation is higher from the QStar instrument implying that the order<strong>of</strong> magnitude increased sensitivity <strong>of</strong> the LTQ-FT relative to the quad-TOF is lost to the inefficiency <strong>of</strong> the PQD. Reproducibility <strong>of</strong> technicalreplicates from PQD is lower than that obtained with data from CAD.240 Quantification <strong>of</strong> Eicosanoid PathwayProteins in Human CerebrospinalFluid Using a Dual Pressure Linear IonTrap Mass SpectrometerR. Biringer 1 , J. Horner 1 , A. Fonteh 2 , S. Kauffman 2 ,A. Huhmer 1 , M. Harrington 21Thermo Fisher Scientific, San Jose, CA, UnitedStates; 2 Molecular Neurology <strong>Program</strong>, HuntingtonMedical Research Institutes, Pasadena, CA, UnitedStatesObjective: To develop a linear ion trap mass spectrometer-basedprotocol for quantifying eicosanoid pathway enzymes and to apply thisprotocol to monitor enzyme changes in episodic migraine. Methods:Peptides diagnostic for eicosanoid pathway enzymes were determinedfrom trypsin digests using standard LC/MSn methodologies.Corresponding stable isotope peptide analogs (SI-peptides) wereselected and synthesized. For each assay, a cocktail <strong>of</strong> all SI-peptidesis spiked into delipidated cerebrospinal fluid (CSF) from migrainestudy participants when well and sick. After trypsin digestion, each<strong>of</strong> the SI-peptides and the corresponding endogenous peptideswere simultaneously quantified using LC/stable isotope selectedreaction monitoring (SI-SRM) using a dual pressure linear ion trap massspectrometer. Results: Linear dynamic range for this assay spans nearlyfour orders <strong>of</strong> magnitude (0.5-4800 fmol). The LOQ is in the low to subfmolrange depending on the peptide, and corresponds to eicosanoidproteins at concentrations in the low ng/mL CSF range. Eicosanoidprotein concentrations exhibit both preparation and migraine statedependencies. Conclusions: We have developed a robust, targetedfull scan MS/MS linear ion-trap based quantitative assay (LC/SI-SRM) for eight enzymes <strong>of</strong> the eicosanoid pathway. Further, we havedemonstrated that this assay can be used to quantify changes in theexpression <strong>of</strong> eicosanoid proteins during episodic migraine.241 Optimization <strong>of</strong> Nanospray Voltageand Spray Stability: Impact onChromatographic Peak Area forAnalyte QuantitationG. Valaskovic 1 , M. Lee 2 , A. Berg 11New Objective, Inc., Woburn, MA, United States;2Milestone Development Services, Newton, PA,United StatesState-<strong>of</strong>-the art liquid chromatography-tandem mass spectrometry(LC-MS/MS) analysis uses a constant electrospray (ESI) voltage fordata acquisition. Modern qualitative and quantitative LC-MS/MSmethods depend on highly efficient gradient elution chromatography.The changing chemical composition <strong>of</strong> mobile phase during gradientelution results in an inherent disconnect with single point ESIvoltage optimization. A constant ESI voltage limits spray stability andcompromises chromatographic peak area quantification, limiting totalpeak area and increasing peak area relative standard deviation (RSD).We have adapted a previously reported closed-loop “orthogonal”feedback control system for qualitative proteomics† to one suitablefor targeted peptide quantification. A digitally controlled nanospraysource was modified to use the high-voltage output from an external,computer controlled power supply. A s<strong>of</strong>tware program controllingthe nanospray emitter stage position and the external power supplysupported a programmable spray compensation voltage duringchromatograpghic analysis. One microliter <strong>of</strong> a standard peptide(MRFA) was autosampler injected (CTC PAL) directly on column (NewObjective PicoFrit, 75 um x 10 cm, C18) to the source equipped linearion trap mass spectrometer (Thermo Scientific, LTQ). Repetitive oncolumninjections at different (fixed) target ESI voltage settings yieldsa apparent compound dependant response curve in which totalchromatographic peak area for selected ion currents reaches a maximumvalue. Spray stability and chromatographic quality was directlycorrelated with the observed spray mode. Altering the ESI voltage from2 kV to 3 kV resulted in a peak area increase <strong>of</strong> 38%. Qualitative visualcomparison <strong>of</strong> noise in the molecular ion signal (523.5 m/z) showed amarkedly higher level at 2.0kV. Fourier transform spectrum analysis <strong>of</strong>selected ion current, revealed a ten-fold increase in noise at 2 and 4Hz for the 2 kV acquisition. Spray instability results in increased noise<strong>of</strong> the reconstructed chromatogram, increasing the uncertainly <strong>of</strong> peakarea measurement. †Valaskovic, G.A, Murphy, J.P., Lee, M.S. “AutomatedOrthogonal Control System for Electrospray Ionization”, J. Am. Soc.Mass Spectrom., 2004, 15, 1201-1215.242 Development <strong>of</strong> a High-SensitivityTargeted MRM Assay for Peptide andProtein QuantificationT. McKenna, A. Bartlett, C. Hughes, J. LangridgeWater Corporation, Manchester, United KingdomDiscovery phase proteomics has generated numerous candidate proteinmarkers for a wide variety <strong>of</strong> biological processes and disease types.To assess the viability <strong>of</strong> these protein expression changes requires theanalysis <strong>of</strong> a larger number <strong>of</strong> samples, preferably in a targeted fashion,and hence the use <strong>of</strong> MRM has become routine. Specific peptides fromthe proteins <strong>of</strong> interest are targeted as surrogate markers for that protein,in a screening assay using a triple quadrupole mass spectrometeroperating in the multiple reaction monitoring (MRM) mode. The MRMmethod which is used to detect specific ions from target molecules hasthe capability to simultaneously quantify large numbers <strong>of</strong> proteins100 • <strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine


with good limits <strong>of</strong> quantification (LOQ) and linear dynamic range. Inthis mode <strong>of</strong> analysis the sensitivity and dynamic range are improvedand providing sufficient data points across a chromatographic peak arerecorded then quantitation is accurate (CV 5-10%). This high sensitivitycoupled with the specificity/selectivity afforded by MRM transitionsallows extensive panels <strong>of</strong> peptide biomarkers to be monitored ina single experiment from complex mixtures. We will describe thedevelopment and implementation <strong>of</strong> novel high-sensitivity MRM assaysfor large scale peptide quantification.243 High Sensitivity Protein QuantitationUsing a Triple Quadrupole with aDual Ion FunnelC. Miller, Y. Yang, K. WaddellAgilent Technologies, Palo Alto, CA, United StatesAssays that are both specific and quantitative for target proteins arecritical for preclinical validation <strong>of</strong> putative biomarkers. Such assays aretypically multiplexed, multiple reaction monitoring (MRM) analyseswhich can provide the high-throughput required. Sensitivity is a keyrequirement for such assays as protein biomarker concentrations maybe quite low in commonly used bi<strong>of</strong>luids such as serum and plasma.Improving the sensitivity <strong>of</strong> LC/MS can be achieved by using nan<strong>of</strong>lowLC, and by enhancing the sampling and transmission <strong>of</strong> ions in themass spectrometer. This study demonstrates the 5-10x sensitivity gainachieved for peptides using a triple quadrupole mass spectrometermodified with a dual ion funnel. The sensitivity achieved using amicr<strong>of</strong>luidic-based nan<strong>of</strong>low LC system compared to a standard LCsystem will be shown discussed.244 Development <strong>of</strong> a Label-FreeQuantitative PhosphoproteomicsPlatform Applicable to Non-CellCulture Biological MatricesE. Soderblom 1 , M.W. Foster 2 , J.W. Thompson 1 ,M.A. Moseley 11Duke Institue <strong>of</strong> Genome Science and Policy,Durham, NC, United States; 2 Duke UniversitySchool <strong>of</strong> Medicine, Durham, NC, United StatesThe most commonly employed LC-MS based quantitativephosphoproteomics experiments are performed using cultured celllineswith incorporated stable-isotope labeled amino acids. Althoughthese systems simplify experimental manipulation and providerelatively large amounts <strong>of</strong> soluble protein, they do not account forthe complexity <strong>of</strong> signal regulation from other factors or cell typespresent in the cell’s native physiological environment. Here wedescribe the general analytical and informatic methodologies usedfor the global quantitation <strong>of</strong> specific sites <strong>of</strong> phosphorylation fromclinically relevant matrices such as human tissue or biological fluidscompatible from a variety <strong>of</strong> different experimental designs. As anexemplar application <strong>of</strong> the methodology, a quantitative analysis <strong>of</strong>lipopolysaccharide challenged and ethyl nitrite protected mouse lungtissue was performed. Phosphopeptide enrichments from 600 ugaliquots <strong>of</strong> trypsin digested homogenized mouse lung tissue from fourunique treatment groups was accomplished using an optimized TiO2spin-column protocol and were subjected to triplicate 1D-LC-MS/MSanalysis on a Waters NanoAcquity UPLC coupled to a Thermo LTQOrbitrap XL. Following accurate mass and retention time alignmentacross all LC-MS runs performed within Rosetta Elucidator, a robustmean normalization <strong>of</strong> precursor ion intensities was applied and areaunder the curve quantitative measurements were made for all precursorions. Qualitative identifications were assigned to each precursor ionfollowing Mascot searches against a mus musculus database appendedwith reverse entries and scoring thresholds were adjusted to yield apeptide 1% FDR. Across all four treatment groups, a total <strong>of</strong> 778phosphopeptides were qualitatively identified. Analytical plus TiO2enrichment variation <strong>of</strong> pre-digested bovine alpha-casein spiked intoeach sample prior to enrichment yielded a phosphopeptide intensityvariation <strong>of</strong> < 20% RSD across approximately three orders <strong>of</strong> magnitude.In summary, these data demonstrate an effective and straight-forwardmethodology to study global changes in phosphorylation directly fromclinically relevant biological samples.245 Studying Whole-Body ProteinTurnover Using Stable-IsotopeLabeled MiceE. Chen 1,2 , A.A. Koller 2 , J. Relucio 1 , S. Nik 1 ,B. Dost 3 , H. Colognato 1 , N. Bandeira 31Department <strong>of</strong> Pharmacological Sciences, StonyBrook University, Stony Brook, NY, United States;2Stony Brook University, Proteomics Center, SchoolOf Medicine, NY, United States; 3 Department <strong>of</strong>Computer Science and Engineering, University <strong>of</strong>California, San Diego, CA, United StatesProtein turnover is a neglected dimension in functional genomicsstudies delineating the dynamic changes <strong>of</strong> protein regulation and linkstranscriptome, proteome, and potentially metabolome. It is known thatproteins are degraded and synthesized continuously at the cellularlevel. The balance <strong>of</strong> degradation and biosynthesis is tightly regulatedand has great implications in normal physiology, cellular regulation, andhuman diseases. Recent technological advances in high-resolution massspectrometry open up new opportunities to make proteome-wideprediction <strong>of</strong> protein turnover. However, studies <strong>of</strong> proteome turnoverin live animals require high efficiency <strong>of</strong> labeling using labeled aminoacids and bioinformatics to deal with highly complex mass spectradata. To unravel the dynamics <strong>of</strong> protein turnover in intact animals,we developed an in vivo pulse-chase strategy and bioinformatics tools(APCIE) to perform analysis <strong>of</strong> proteome turnover. We pulsed C57BL/6mice with 15N labeled amino acids through diet and chased 15N labeledyoung (3 weeks old) and old (4 months old) mice with unlabeled dietfor 2 weeks. Lungs from these two groups <strong>of</strong> mice were harvested andanalyzed by mass spectrometry-based shotgun proteomic techniques.Using APCIE, we determined protein degradation (loss <strong>of</strong> 15N labeledamino acids) and protein synthesis (incorporation <strong>of</strong> 14N aminoacids) <strong>of</strong> over 300 proteins commonly found in young and old miceand observed aged-dependent differences in proteome turnover. Asexpected, most proteins are turned over faster in young mice than oldmice reflected by massive loss <strong>of</strong> 15N amino acids and incorporation <strong>of</strong>14N amino acids. Our newly developed in vivo pulse-chase techniqueand algorithm can be used for global pr<strong>of</strong>iling <strong>of</strong> protein turnoverin intact animals to study complex mouse models mimicking humandiseases. Combining global pr<strong>of</strong>iling <strong>of</strong> protein turnover and proteinexpression will make a tremendous impact towards mechanism-driventherapeutics based on dynamic regulation <strong>of</strong> proteins and proteinnetworks.Poster Abstracts<strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine • 101


Poster Abstracts246 Purification <strong>of</strong> Bacterial APOA-1and Characterization <strong>of</strong> Novel Anti-Cancer Drug Delivery SystemT. Young 1 , A. Lacko 21North Carolina State University, Raleigh, NC,United States; 2 University <strong>of</strong> North Texas HealthScience Cente, Fort Worth, TX, United StatesAlthough chemotherapy regimens have proved effective in attackingcancer cells and tumors, side effects and drug resistance remain amajor concern during cancer therapy. The use <strong>of</strong> reconstituted highdensity lipoprotein (rHDL) nanoparticles has been investigated as adrug delivery system, including the transporting <strong>of</strong> small interferingribonucleic acid (siRNA). The use <strong>of</strong> rHDL nanoparticles has greatpotential, in this regard, due to their ability to specifically targetcancer cells via the HDL (SR-B1) receptor. The goal <strong>of</strong> these studieswas to improve the purification <strong>of</strong> Apolipoproteins A-1 (ApoA-1), amajor component <strong>of</strong> rHDL, and the preliminary characterization <strong>of</strong>the siRNA carrying rHDL nanoparticles. E.coli cells transfected withthe apo A-I gene was grown at 37OC, until an optical density <strong>of</strong> 0.6was reached. The cells were then induced with 0.5mM Isopropyl β-D-1-thiogalactopyranoside (IPTG) and centrifuged. Subsequently, thepellets were suspended in the lysate solution and loaded onto a Nickel-Sepharose column. Thereafter, rHDL nanoparticles using siRNA wereprepared. 160 mg per liter <strong>of</strong> purified ApoA1 was obtained. Particlemeasurements and the chemical composition <strong>of</strong> the particles are beinginvestigated. Further investigation regarding the efficiency <strong>of</strong> theincorporation <strong>of</strong> siRNA, the physical and chemical properties as wellas cytotoxicity <strong>of</strong> the particles will contribute to the assessment <strong>of</strong> theefficiency <strong>of</strong> these particles as a novel anti-cancer drug delivery system.247 Screening for Optimal PurificationConditions for Histidine-TaggedWater-Soluble and MembraneProteins Using Magnetic BeadsMethodsH. Hedlund, E. Mascher, G. RisbergGE Healthcare Life Sciences, Uppsala, SwedenThe development <strong>of</strong> methods for efficient expression screening <strong>of</strong>multiple numbers <strong>of</strong> recombinant proteins or optimization <strong>of</strong> thepurification conditions <strong>of</strong> proteins is essential to shorten the time fromgene to drug targets. The combination <strong>of</strong> para- magnetic beads andaffinity purification using IMAC resins (pre-charged with Ni2+-ions)for capture <strong>of</strong> histidine tagged proteins is an easy-to-use format anda powerful tool allowing for rapid, small-scale purification. We willdescribe two studies for optimization <strong>of</strong> purification conditions <strong>of</strong>histidine-tagged proteins. All experiments were done using Ni2+chargedHis Mag Sepharose Ni. In the first study optimal purificationconditions for a water-soluble histidine-tagged protein was investigatedat eight different buffer conditions and four different sample loads(varying from 25 % to 100 % <strong>of</strong> the total binding capacity). The studywas performed using a liquid handling workstation.A good balancebetween yield and purity was obtained at 40 mM imidazole and at asample load between 50 % and 100 % <strong>of</strong> the total binding capacity. Inthe second study screening for optimal solubilization and purificationconditions for an integral membrane protein was performed, usingseven detergents. The screening protocol involved a rapid purificationstep before analyses, evaluation and final choice <strong>of</strong> detergent(s).Obtained results from this screening gave reliable results that could alsobe fed into further scale-up experiments. We will present results fromWestern blotting, gel filtration and SDS-PAGE. The para-magnetic beadformat simplified and shortened the handling procedures through-outboth screening workflows.248 Production <strong>of</strong> Recombinant MouseFlower Protein in E. coli: Application<strong>of</strong> Mistic Fusion to Improving theExpression <strong>of</strong> Membrane ProteinsJ.L. Martinez-Torrecuadrada 1 , M. Marenchino 2 ,R. González 1 , J. López-Alonso 2 ,R. Campos-Olivas 2 , G. Roncador 3 , E. Moreno 41Proteomics Core Unit, Spanish National CancerCentre (CNIO), Madrid, Spain; 2 Spectroscopyand Nuclear Magnetic Resonance Unit. SpanishNational Cancer Centre (CNIO), Madrid, Spain;3Monoclonal Antibodies Core Unit, SpanishNational Cancer Centre (CNIO), Madrid, Spain;4Cell Competition Group, Spanish National CancerCentre (CNIO), Madrid, SpainThe structural and functional studies <strong>of</strong> membrane proteins havebeen greatly hindered due to difficulties in their over-expression andproduction. It is also <strong>of</strong>ten difficult to generate effective antibodies tomembrane proteins. In our laboratory, we have addressed the problem<strong>of</strong> producing high level amounts <strong>of</strong> membrane proteins in Escherichiacoli by use <strong>of</strong> Mistic, a Bacillus subtilis protein, as a fusion partner. Flower(Fwe) is a membrane protein that is conserved in animals and proposedto be a Ca2+ channel in neurons. It has been recently reported thatin Drosophila Flower is a component <strong>of</strong> the cell competition responsethat is required and sufficient to label cells as “winners” or “losers”,promoting the elimination <strong>of</strong> weaker cells from a growing populationin order to optimize tissue fitness. This process may have biomedicalimplications because imbalances in cell fitness appear during aging,cancer formation and metastasis. In this work, we employed themembrane protein Mistic to assist in the production <strong>of</strong> this protein.The construct 6xHis-Mistic – Flower carrying a TEV cleavage site wasefficiently overexpressed in E. coli. The IMAC-based purification andcleavage <strong>of</strong> the recombinant protein was achieved in the presence <strong>of</strong>the detergent lauryldimethylamino oxide (LDAO). Circular dichroismshowed a high content in helical structure as predicted from the aminoacid sequence by the program TMHMM. Also, the recombinant Mistic–Flower was used as antigen source to produce monoclonal antibodies inKO mice. The generated monoclonal antibodies were able to recognizeFlower protein by Western blot and immunohistochemistry, indicatingthat this recombinant protein retained the antigenicity <strong>of</strong> the nativeform. These antibodies will facilitate importantly further functionalstudies on Flower protein.102 • <strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine


249 Rapid Monoclonal Antibody GlycanPr<strong>of</strong>iling Using an IntegratedMicr<strong>of</strong>luidic-Based mAb-Glyco Chipand Quadrupole Tim-<strong>of</strong>-Flight MassSpectrometryC. Miller, N. Tang, K. WaddellAgilent Technologies, Palo Alto, CA, United StatesProtein biologics now represent a significant share <strong>of</strong> pharmaceuticalsales and future growth potential, particularly in an era <strong>of</strong> increasingpatent expirations. A range <strong>of</strong> analytical methods is required todetermine the purity, identity and integrity <strong>of</strong> protein biologics atmultiple points along the manufacturing process, from cell culture todownstream purification, product characterization and lot release.Characterization <strong>of</strong> glycans from antibodies is fundamentally importantin biotherapeutics design and disease progression and detection. Theability to characterize glycans rapidly has been limited by the samplepreparation steps and structural complexity <strong>of</strong> the glycoproteins.To address this problem, we have developed a micr<strong>of</strong>luidic chipthat performs rapid on-line cleavage <strong>of</strong> glycans from monoclonalantibodies, captures the released glycans and then separates themprior to nanospray ionization in the mass spectrometer. The entire runtime is 12 minutes. A glycan accurate mass database was establishedallowing quick assignment and identification <strong>of</strong> the glycans.250 Protein Cleavage, Disulfide BondsReduction, Metabolite Synthesis andMuch More Using Electrochemistry/MSJ. Powers 1 , J. Purkerson 1 , A. Kraj 2 , M. Eysberg 2 ,J.P. Chervet 21Antec, Palm Bay, FL, United States; 2 Antec,Zoeterwoude, The NetherlandsRecently, the scope <strong>of</strong> Electrochemistry (EC) upfront MS has beenextended from mimicking drug metabolism towards new applicationssuch as: protein/peptide cleavage, disulfide bonds reduction, covalentdrug-protein binding, etc. In this presentation we will show theapplication <strong>of</strong> on-line EC/MS as a powerful tool to simulate variousoxidation and reduction processes in life sciences. A specially designedµ-preparative electrochemical flow cell will be presented. The cellallows the synthesis <strong>of</strong> sufficient amounts <strong>of</strong> metabolites in a few minutesfor subsequent use as reference material (e.g. NMR or MS). Newscanning method was applied for oxidation <strong>of</strong> the highly concentratedsamples (mM range) to achieve high yield in the metabolites formation.Stable oxidation conditions were obtained without the need <strong>of</strong> anycell maintenance for a prolonged period <strong>of</strong> time. Electrochemistryup front MS can be applied for protein and peptide cleavage (as apromising new approach to enzymatic digestion). Electrochemicalcleavage <strong>of</strong> proteins and peptides occurs very specifically at C-terminal<strong>of</strong> the Tyrosine and Tryptophan peptide bonds. Examples <strong>of</strong> oxidativecleavage will be presented. Disulfide bonds are one <strong>of</strong> the mostimportant post-translational modifications for proteins. In this posterwe present the structural analysis <strong>of</strong> biologically active peptidesand proteins containing disulfide bonds (e.g., somatostatin, insulin,etc) using electrochemistry (EC) combined with mass spectrometry.Therefore the sample undergoes electrolytic disulfide cleavage in theelectrochemical flow cell followed by online MS analysis. Based on theintact protein mass and the resulting fragments and the MS/MS dataunambiguous assignment <strong>of</strong> the disulfide bonds becomes possible.All these applications illustrate the tremendous power and broadapplicability <strong>of</strong> electrochemistry as a tool to mimic nature’s Redoxreactions within a few seconds or minutes.251 Protein Sequencing Research Group(PSRG): Results <strong>of</strong> the PSRG <strong>2011</strong>Study: Sensitivity Assessment <strong>of</strong>Edman and Mass SpectrometricTerminal Sequencing <strong>of</strong> an UnknownProteinH. Remmer 1 , J.S. Smith 2 , W. Sandoval 3 , B. Xiang 4 ,K. Mawuenyega 5 , D. Suckau 6 , V. Katta 3 ,J.J. Walters 7 , P. Hunziker 81University <strong>of</strong> Michigan, Ann Arbor, MI, UnitedStates; 2 University <strong>of</strong> Texas Medical Branch,Galveston, TX, United States, 3 Genentech,Inc., South San Francisco, CA, United States,4Monsanto Company, St. Louis, MO, United States,5Washington University School <strong>of</strong> Medicine,St. Louis, MO, United States, 6 Bruker Daltonics,Bremen, Germany, 7 Sigma-Aldrich, St. Louis,MO, United States, 8 University <strong>of</strong> Zurich, Zurich,SwitzerlandEstablishing the N-terminal sequence <strong>of</strong> intact proteins plays a criticalrole in biochemistry and potential drug development. N-terminalsequence analysis is necessary for quality control <strong>of</strong> protein biologics,for determining sites <strong>of</strong> signal peptide cleavage events, as a first stepin elucidating the sequences <strong>of</strong> genes from uncommon species andfor the characterization <strong>of</strong> monoclonal antibodies. Automated Edmandegradation has been the method <strong>of</strong> choice for these analyses. However,alternate methods for N-terminal sequence analysis have emerged. Therecent PSRG studies have established that Edman sequencing and massspectrometry based techniques have varied strengths and weaknessesdepending on several experimental factors and both play an importantrole in terminal sequencing. With this complimentary role realized,the <strong>2011</strong> PSRG study attempts to evaluate the sensitivity limits <strong>of</strong> thevarious sequencing techniques. The PSRG distributed three samplesets <strong>of</strong> 3 tubes each, varying by sample format (lyophilized, gel sliceor membrane piece). Each set <strong>of</strong> three samples contains the samerecombinant protein with increasing amounts <strong>of</strong> material. The sequence<strong>of</strong> this protein is not listed in any database. Participants could requestany one, two, or all three sample sets. Including PSRG committee, a total<strong>of</strong> 38 participants requested 74 sample sets. The participants wereasked to determine as many amino acids from both termini by theirmethod <strong>of</strong> choice, and were encouraged to try multiple methods forsequence elucidation. Study participants were directed to a websiteto anonymously upload sequences and supporting data. Our analysisfocuses on the length and accuracy <strong>of</strong> the sequence calls reported bythe participants, and how that compares with decreasing amounts <strong>of</strong>protein and the type <strong>of</strong> sample format analyzed. A comparison <strong>of</strong> theresults obtained by Edman chemistry and by alternative technologiesas well as information on the type <strong>of</strong> instruments and protocols isreported.Poster Abstracts<strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine • 103


Author IndexPlease note that only primary authors <strong>of</strong> poster presentations are listed in this index.AGKAuthor IndexAdams, P. - 125Ahmed, A. - 103Albanese, J. - 207, 216Asperger, A. - 235BBarsch, A. - 222, 223Berg, A. - 215Biringer, R. - 240Bishop, J. - 131Bodi, K. - 156Boland, J. - 146Bolcato, C. - 233Bramlett, K. - 163CCampos, A. - 237Chamberlain, S. - 123Chen, E. - 245Chen, S.M. - 217Chuu, J. - 149Curchoe, C. - 128DDiaz, C. - 178Diaz Acosta, B. - 112Diaz-Candelas, P. - 142Donnelly, R. - 126Duhr, S. - 182, 201EEckart, S.P. - 179FFadgen, K. - 227Fleming, B. - 106Freeman, R. - 171, 208Gadush, M. - 173Ganter, B. - 119Garge, N.R. - 195Gendeh, G. - 183, 184, 191, 198Giuffre, A. - 129Grigorean, G. - 239Grills, G. - 105, 113, 121Gross, V. - 219Grove, D. - 157HHagen, A. - 102Hamilton, J. - 107Hawke, D.H. - 189Hebert, N. - 210Hedlund, H. - 247Hemström, T. - 196Hendrickson, W. - 115Heuermann, K. - 130Hicks, B. - 104Hicks, L. - 177Hinerfeld, D. - 144Holbrook, J. - 122, 165Hughes, C. - 218JJaggi, R. - 164Jen, J. - 161Jennings, S.F. - 114Johnson, D. - 209Jonscher, K. - 236Joshi, S. - 147Jovanuvich, S. - 162Kain, S. - 136Kamberov, E. - 145Khanna, A. - 150Kinross, C. - 152Koller, A. - 232Küster-Schöck, E. - 176LLangmore, J. - 188Lea, K. - 168Liao, A. - 118Lin, S. - 108Liu, C.S. - 134Liu, J. - 135Lopez, P. - 175Lübbert, C. - 214Luna, L. - 200MMa, B. - 194Magpiong, I. - 193Marshall-Waggett, C. - 212Martinez-Torrecuadrada, J.L. - 248Masri, F. - 228McCary, A. - 141McGuigan, J. - 133McKenna, T. - 242McMillan, M. - 124Miller, C. - 221, 243, 249Müller, B. - 238Murage, E. - 172Murray, J. - 185Myers, J. - 153NNarayanan, A. - 197Needleman, D. - 127104 • <strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine


OOng, J. - 117PPatel, S. - 169Peake, D. - 224Person, M. - 109Powers, J. - 160, 225, 250QQu, L. - 170RRavi, H. - 151Remmer, H. - 251Resemann, A. - 192, 204, 230Rezenom, Y. - 220Rosato, C. - 101Ruddat, V. - 202SSalowsky, R. - 229San Jose Hinahon, C. - 132Schweiger-Hufnagel, U. - 203Shan, P. - 190Smith, T. - 139Snyder-Leiby, T. - 205Soderblom, E. - 244Sondej, M. - 181Stapels, M. - 234Stewart, F.J. - 155Supunpong Hernandez, N. - 158TThai, K. - 148Thompson, J.W. - 199Turner, M. - 137Turpen, P. - 110VVacek, G. - 140Valaskovic, G. - 241van Soest, R. - 211Vincent, E. - 116Vogel, C. - 143WWang, X. - 226Whitley, P. - 138Williams, R. - 174Winkvist, M. - 186, 187Wojtkiewicz, M. - 180XXiang, F. - 206YYang, C. - 111Young, T. - 246ZZavadil, J. - 120Zianni, M. - 154, 159Author Index<strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine • 105


<strong>ABRF</strong> <strong>2011</strong> Exhibits*All <strong>ABRF</strong> <strong>2011</strong> Meeting attendees are invited to visit the ExhibitHall featuring the leading experts in life sciences research andbiotechnology.Exhibit HoursSunday through TuesdayLevel 2, Grand Oaks BallroomSunday, February 20 ....................................10:00 am – 6:30 pmMonday, February 21 ..................................10:00 am – 6:30 pmTuesday, February 22 ..................................10:00 am – 2:00 pmThe purpose <strong>of</strong> the exhibition is to further the education <strong>of</strong>the scientists working in resource and research biotechnologylaboratories. The exhibitors will showcase instruments, productsor services for use in teaching, research, books or otherpublications in scientific fields <strong>of</strong> relevance.*Participation in the Exhibits <strong>Program</strong>, including attendance <strong>of</strong>Exhibitor Presentations, does not constitute an endorsement bythe <strong>Association</strong> <strong>of</strong> <strong>Biomolecular</strong> Resource Facilities (<strong>ABRF</strong>) <strong>of</strong> theclaims, products, or services <strong>of</strong>fered.Exhibitor Directory in Alphabetical OrderExhibitors confirmed as <strong>of</strong> January 29, <strong>2011</strong>.Exhibitor DirectoryAB SCIEX Booth 501110 Marsh DriveFoster City, CA 94404United StatesP +1 508-383-7640F +1 650-627-2601www.absciex.comAB SCIEX is a global leader in the development <strong>of</strong> life scienceanalytical technologies that help answer complex scientific challenges.The company provides mass spectrometry instrumentation,s<strong>of</strong>tware and services for scientific analysis, including coreand applied research.<strong>ABRF</strong> Booth 3179650 Rockville PikeBethesda, MD 20814United StatesP +1 301-634-7306F +1 301-634-7455abrf@abrf.orgwww.abrf.orgThe <strong>Association</strong> <strong>of</strong> <strong>Biomolecular</strong> Resource Facilities (<strong>ABRF</strong>) is aunique membership association comprised <strong>of</strong> approximately800 scientists working in resource and research biotechnologylaboratories. Our members represent over 140 internationalcore laboratories in government, academia, research, industryand commercial settings. <strong>ABRF</strong> promotes the education andcareer advancement <strong>of</strong> scientists through conferences, a quarterlyjournal, publication <strong>of</strong> <strong>ABRF</strong> Research Group studies, andconference travel awards. The <strong>ABRF</strong> Research Group studies aresponsored annually to help researchers assess and incorporatenew biotechnologies into their laboratories, and are world-renownedfor their practical and educational benefit.Advanced Analytical Booth 6022711 South Loop DriveAmes, IA 50010United StatesP +1 515-296-6600F +1 515-294-7141webinfo@aati-us.comwww.aati-us.comAdvanced Analytical is a leading innovator <strong>of</strong> analytical instrumentsthat improve laboratory throughput. The AdvanCE TMFS96 and AdvanCE TM FS12 are 96 and 12-channel parallel capillaryelectrophoresis fluorescent instruments for analyzing dsD-NA fragments and RNA. A specialized reagent kit is available forautomated, sensitive and rapid analysis <strong>of</strong> Next-Gen Sequencingfragments.Agilent Technologies Booth 3012850 Centerville RoadWilmington, DE 19808United StatesP +1 800-227-9770F +1 302-633-8944agilent_inquiries@agilent.comwww.agilent.com/chemAgilent is a leading supplier <strong>of</strong> life science instrumentation. LC,GC & mass specs, DNA microarrays and scanners, thermal cyclers,automation instruments, reagents and s<strong>of</strong>tware productsare used globally. Agilent has developed products and servicesutilized along the entire discovery value chain, from basic biologicalresearch through drug discovery and manufacturing.106 • <strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine


AnaSpec, Eurogentec Group Booth 41734801 Campus DriveFremont, CA 94555United StatesP +1 510-791-9560F +1 510-791-9573admin@anaspec.comwww.anaspec.comAs a subsidiary <strong>of</strong> Eurogentec, AnaSpec <strong>of</strong>fers expertise inpeptides, detection reagents, antibodies, assay kits, oligonucleotides,and qPCRs. AnaSpec carries a broad product line <strong>of</strong>biochemicals and reagents for basic research, high-throughputscreening and drug discovery.Bioinformatics SolutionsInc. (BSI) Booth 402470 Weber Street North, Suite 204Waterloo, Ontario N2L6J2CanadaP +1 519-885-8288F +1 519-885-9075info@bioinfor.comwww.bioinfor.comBSI produces life science s<strong>of</strong>tware to serve the needs <strong>of</strong> pharmaceutical,biotechnological and academic scientists; and tothe progression <strong>of</strong> drug discovery research. S<strong>of</strong>tware solutionsinclude: ZOOM for next-generation sequencing, PEAKS forproteomic mass spectrometry, RAPTOR for protein structureprediction and PatternHunter for general purpose homologysearching.Bioo Scientific Booth 7183913 Todd Lane, Suite 312Austin, TX 78744United StatesP +1 888-208-2246nextgen@biooscientific.comwww.biooscientific.comBioo Scientific provides innovative, custom solutions to corefacilities. These include DNA and RNA AIR TM Next-GenerationSequencing library preparation kits, which <strong>of</strong>fer increased sensitivity,flexibility and speed. We also <strong>of</strong>fer the ExoMir TM kit forthe isolation <strong>of</strong> RNA from exosomes and MaxDiscovery TM kits fortoxicity and cytotoxicity analysis.Bio-Synthesis, Inc. Booth 104612 East Main StreetLewisville, TX 75057United StatesP +1 972-420-8505F +1 972-420-0442biosyn@biosyn.comwww.biosyn.comBio-Synthesis provides synthetic peptides including libraries andarrays, Antibodies, PNA, Bioconjugates, Custom DNA synthesisand its analogs, Gene synthesis, PCR based detection kits, Cellline authentication, HLA typing and Analytical services. Withover two decades <strong>of</strong> experience producing custom productsfor both research and pharmaceutical use, BSI has developed areputation for both small and large scale synthesis using optimalprocesses that meet your specifications.Bruker Daltonics Inc. Booth 21440 Manning RoadBillerica, MA 01821United StatesP +1 978-663-3660F +1 978-667-5993www.bdal.comBruker is a leading provider <strong>of</strong> Separation and Mass Spectrometryinstruments for the Analytical Sciences. Our innovative andeasy to use product families (ESI-TOF, Ion Trap, FTMS, MALDI-TOF, GC, GCMS, ICP-MS) provide the highest performance,highest value systems for a wide range <strong>of</strong> small molecule andprotein analysis applications.Caliper Life Sciences Booth 30468 Elm StreetHopkinton, MA 01748United StatesP +1 508-435-9500www.caliperLS.comCaliper Life Sciences is a premier provider <strong>of</strong> cutting-edge technologiesenabling researchers in the life sciences industry tocreate life-saving and enhancing medicines and diagnostic testsmore quickly and efficiently. Caliper is aggressively innovatingnew technology to bridge the gap between in vitro assays andin vivo results and then translating those results into cures forhuman disease. Caliper’s portfolio <strong>of</strong> <strong>of</strong>ferings includes state-<strong>of</strong>the-artmicr<strong>of</strong>luidics, lab automation & liquid handling, opticalimaging technologies, and discovery & development outsourcingsolutions.Exhibitor Directory<strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine • 107


Exhibitor Directory — ContinuedExhibitor DirectoryCEM Corporation Booth 303P.O. Box 200, 3100 Smith Farm RoadMatthews, NC 28106United StatesP +1 704-821-7015F +1 704-821-8710info@cem.comwww.cem.comMake higher purity peptides faster with the Liberty MicrowavePeptide Synthesizer. This system has the fastest cycle times availablefor routine and difficult peptides, including peptides youcan’t access under conventional conditions. The Discover® Systemprovides improved sequence coverage and higher efficienciesfor your enzymatic digests in only 15 minutes.chemagen USA Booth 10067 Millbrook StreetSuite 522Worcester, MA 01606United StatesP +1 508-459-7727F +1 508-459-7548www.chemagen.comchemagic automation and reagents perform fast and reliablemagnetic bead-based DNA and RNA extractions from samplevolumes from 10 ul to 10 ml <strong>of</strong> blood, tissue, bacteria, food, PCRand sequencing products, etc. Advantages <strong>of</strong> this unique systemare fast processing, unmatched sample volume range and robustchemistry.Dionex Corporation Booth 2151228 Titan Way/P.O. Box 3603Sunnyvale, CA 94088-3603United StatesP +1 408-737-0700F +1 408-730-9403angie.jinks@dionex.comwww.dionex.comDionex has developed a large portfolio <strong>of</strong> powerful instrumentsand chemistries for separations <strong>of</strong> biomolecules, such asproteins, peptides, monoclonal antibodies, nucleic acids, andcarbohydrates that serve in diverse applications ranging frombiomarker discovery to drug R&D and QA/QC. Visit us at booth215 to learn more about ground-breaking life science solutionsfrom Dionex.DNAnexus Booth 404420 Florence StreetSuite 210Palo Alto, CA 94301United StatesP +1 650-204-1938info@dnanexus.comwww.DNAnexus.comDNAnexus provides solutions for both DNA sequencing centersgrowing their next-gen capacity, and the researchers workingwith next-gen sequence data. Our web-based platform solvesthe data management and analysis challenges common to bothwith a single, unified system. We support sequencing operationsand research organizations <strong>of</strong> any size, with absolutely no upfronthardware investment needed.eagle-i Consortium Booth 221One Kendall Square, Suite B6303Cambridge, MA 02139United StatesP+1 617-384-8787info@eagle-i.orgwww.eagle-i.orgThe eagle-i Consortium, made up <strong>of</strong> nine member institutionsacross the United States, is building a prototype <strong>of</strong> a nationalresearch resource discovery network. The eagle-i Network willconnect biomedical researchers with valuable resources, services,and technologies in a faster and more cost-effective way thanpreviously possible.EdgeBio Booth 300201 Perry ParkwaySuite 5Gaithersburg, MD 20877United StatesP +1 800-326-2685F +1 301-990-0881info@edgebio.comwww.edgebio.comEdgeBio <strong>of</strong>fers next-generation sequencing services. From experimentaldesign to bioinformatics, EdgeBio is the perfect solutionfor all your sequencing needs. EdgeBio also sells kits forsequencing reaction cleanup, PCR purification, and plasmid purification,as well as competent cells.108 • <strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine


FASEB MARC <strong>Program</strong> Booth 4169650 Rockville PikeBethesda, MD 20814United StatesP +1 301-634-7020F +1 301-634-7353marc@faseb.orgwww.faseb.org/marcFASEB MARC <strong>Program</strong> provides a variety <strong>of</strong> activities to supportthe training <strong>of</strong> underrepresented minority students, postdoctorates,faculty and scientists in the biomedical sciences. We <strong>of</strong>fertravel awards for scientific meetings, research conferences, andstudent summer research opportunity programs. We also sponsorCareer Development <strong>Program</strong>s including grantsmanship andleadership training seminars.Fluidigm Corporation Booth 6147000 Shoreline Court, Suite 100South San Francisco, CA 94080United StatesP +1 866-358-4354F +1 650-871-7152www.fluidigm.comFluidigm develops, manufactures and markets life-science systemsbased on integrated fluidic circuits (IFCs). This technologyfurthers research by minimizing costs and enhancing sensitivityfor applications such as single-cell gene expression pr<strong>of</strong>iling,high-throughput SNP genotyping, and next-generation sequencing.Fluidigm products are used for research only.GE Healthcare Booth 714800 Centennial AvenuePiscataway, NJ 08854United StatesP +1 800-526-3593F +1 877-295-8102cs-us@ge.comwww.gelifesciences.comGE Healthcare Life Sciences provides tools for drug discovery,biopharmaceutical manufacturing and cellular technologies, soresearch scientists and specialists around the world can be moreproductive, effective and creative. Our vision is to be the startto-finishbioprocessing solution provider, the partner <strong>of</strong> choicein cell and protein research, and the leader in life sciences services.Gene Codes Corporation Booth 220775 Technology DriveSuite 100AAnn Arbor, MI 48108United StatesP +1 734-769-7249F +1 734-769-7074gcinfo@genecodes.comwww.genecodes.comGene Codes is an international s<strong>of</strong>tware firm. Our DNA sequenceanalysis s<strong>of</strong>tware, Sequencher TM is the industry leader.Customer-driven product development, combined with the latestprogramming techniques, and rigorous quality control to developproducts <strong>of</strong> the utmost usability and relevance has beenthe key to our success.Genetix Booth 616120 Baytech DriveSan Jose, CA 95134United StatesP +1 408-719-6400F +1 408-719-6401bernette.jones@genetix.comwww.genetix.comGenetix provides unrivalled solutions for imaging and imageanalysis. The company’s imaging platforms <strong>of</strong>fer solutions topick microbial colonies in genomic studies, to screen and selectmammalian cell lines, monitor cell growth, evaluate cellular responseand quantify tissue biomarkers.GENEWIZ, Inc. Booth 316115 Corporate BoulevardSouth Plainfield, NJ 07080United StatesP +1 877-436-3949www.genewiz.comGENEWIZ, Inc. provides award-winning DNA sequencing, molecularbiology, and genomic services. We build codon-optimizedgenes through our powerful Gene Synthesis technologies,and provide high-quality plasmids from mini-to-giga scales usingour Plasmid Preparation services. With over10 years experiencein DNA sequencing, we are well-equipped to handle sequencingprojects <strong>of</strong> all complexities.Exhibitor Directory<strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine • 109


Exhibitor Directory — ContinuedGenomatix S<strong>of</strong>tware, Inc. Booth 2183025 Boardwalk, Suite 160Ann Arbor, MI 48108United StatesP +1 877-436-6628F +1 734-622-0477grant@genomatix-s<strong>of</strong>tware.comwww.genomatix-s<strong>of</strong>tware.comWith a focus on translating NGS data from sequence files tomeaningful biological results, Genomatix leverages it’s 12+ yearhistory <strong>of</strong> s<strong>of</strong>tware and data base development to bring youa state-<strong>of</strong>-the-art, comprehensive, integrated platform for theanalysis <strong>of</strong> your NGS data.Geospiza Booth 519100 West Harrison, North Tower, Suite 330Seattle, WA 98119United StatesP +1 206-633-4403F +1 206-633-4415info@geospiza.comwww.geospiza.comDesigned by biologists for biologists, Geospiza is the marketleading developer <strong>of</strong> GeneSifter® s<strong>of</strong>tware, providing integratedsolutions for Next Generation Sequencing analysis and LIMS.For over 10 years, Geopsiza has set the industry standard forhigh value, low cost, “out <strong>of</strong> the box” solutions serving the lifesciences market.Hamilton Robotics Booth 2234970 Energy WayReno, NV 89502United StatesP +1 800-548-5950F +1 775-858-3024roboticsales@hamiltoncompany.comwww.hamiltonrobotics.comHamilton Robotics is dedicated to the design and manufacture<strong>of</strong> automated liquid handling workstations. Key to our productsis our air displacement pipetting and monitoring technologyand s<strong>of</strong>tware controlling our systems. Our workstations ands<strong>of</strong>tware serve as a common high precision and flexible baseupon which to provide automated solutions.iLab Solutions, LLC Booth 700124 Lexington AvenueCambridge, MA 02138United StatesP +1 617-297-2805F +1 877-812-6477heather.evans@ilabsolutions.comwww.ilabsolutions.comLab Solutions provides core facility management s<strong>of</strong>tware tostreamline requests, track usage, and automate billing. The webbasedsolution supports the entire shared service work flow,from scheduling and request approvals through communication,billing and reporting. Integration available with financial systemsuch as SAP, PeopleS<strong>of</strong>t, Banner, and Lawson.Illumina Booth 5159885 Towne Centre DriveSan Diego, CA 92121United StatesP +1 858-202-4500F +1 858-202-4766info@illumina.comwww.illumina.comIllumina enables discovery by providing comprehensive DNAand RNA analysis solutions, including genotyping and copynumber analysis, gene expression, sequencing, and epigenetics:the cornerstones <strong>of</strong> genetic research. Our portfolio <strong>of</strong> tools integratesour sequencing and microarray technologies, as well aspowerful assay protocols to make possible a new scale <strong>of</strong> biologicalresearch.Intavis Booth 600945 West George StreetSuite 201Chicago, IL 60657United StatesP +1 773-572-5799www.intavis.comIntavis is an industry leader in manufacturing robotic instrumentsfor in situ hybridization, immunohistochemistry, peptidesynthesis, protein digestion, and MALDI spotting.Exhibitor Directory110 • <strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine


IntegenX, Inc. Booth 2015720 Stoneridge DriveSuite 300Pleasanton, CA 94588United StatesP +1 925-701-3400F +1 925-754-7373info@integenx.comwww.integenx.comIntegenX, Inc. produces automated sample processing systemsfor life sciences and applied sciences markets. The company’sproducts: The new Apollo 324 automates Next-Gen librarypreparation and the Apollo 100 automates sample processingfor Sanger cycle sequencing. Our products dramatically reduceprocess time and reagent expenses.Integrated DNA Technologies Booth 3051710 Commercial ParkCoralville, IA 52241United StatesP +1 800-328-2661custcare@idtdna.comwww.idtdna.comServing the areas <strong>of</strong> academic research, biotechnology, clinicaldiagnostics and pharmaceutical development, IDT is the largestmanufacturer <strong>of</strong> custom oligo products in the United States.From oligos in plates to gene knockdown products, from fluorescentDNA probes to gene synthesis, IDT <strong>of</strong>fers the highestquality, customer service and turnaround time.Kapa Biosystems, Inc. Booth 619600 West Cummings ParkSuite 2250Woburn, MA 01801United StatesP +1 781-497-2933F +1 781-497-2934sales@kapabiosystems.comwww.kapabiosystems.comKapa Biosystems, Inc. <strong>of</strong>fers a portfolio <strong>of</strong> best-in-class PCR reagentsengineered using our high-throughput molecular evolutionplatform. The novel DNA polymerases contained within ourkits confer dramatic improvements to performance that enableresults and applications not possible wtih wild-type enzymes.Kapa has engineered high performance reagents for real-timePCR, high fidelity PCR, next-generation DNA sequencing, fastPCR, crude sample PCR, robust PCR, and molecular diagnostics.LC Sciences Booth 3182575 West Bellfort Street, #270Houston, TX 77054United StatesP +1 713-664-7087F +1 713-664-8181info@lcsciences.comwww.lcsciences.comLC Sciences is a genomics and proteomics company <strong>of</strong>feringinnovative and quality products and services. We provideunique and customizable oligonucleotide and peptide microarrayproducts designed for nucleic acid and protein-pr<strong>of</strong>iling,biomarker-screening, drug screening, and development <strong>of</strong> diagnostic-devices.Leica Microsystems Booth 2172345 Waukegan RoadBannockburn, IL 60015United StatesP +1 800-248-0123F +1 847-236-3009info@leica-microsystems.comwww.leica-microsystems.comLeica Microsystems <strong>of</strong>fers the latest imaging technology. Withthe high sensitivity Leica TCS SP5, you can buy a basic confocalmicroscope today that is easily upgraded to keep pace withyour science. Other innovations: the Leica SCN400 slide scannerwith dynamic focus tracking for fast scanning while revealingmore details; and the Leica LMD6500 laser microdissectionsystem that uses gravity to collect specimens in a contact-freemanner.Life Technologies Booth 6015791 Van Allen WayCarlsbad, California 92008United StatesP +1 760-603-7200rose.mori@lifetech.comwww.lifetechnologies.comLife Technologies is a global biotechnology tools companydedicated to improving the human condition. Our customersdo their work across the biological spectrum, working to advancepersonalized medicine, regenerative science, moleculardiagnostics, agricultural and environmental research, and 21stcentury forensics.Exhibitor Directory<strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine • 111


Exhibitor Directory — ContinuedExhibitor DirectoryMassTech, Inc. Booth 1036992 Columbia Gateway Drive, #160Columbia, MD 21046United StatesP +1 443-539-1758F +1 443-539-1759www.apmaldi.comMassTech, Inc. manufactures AP/MALDI ion sources and conductson-going research and development in mass spectrometryrelated technology. MassTech’s AP/MALDI ion source is compatiblewith mass spectrometers from manufacturers such as: Agilent,Applied Biosystems/MDS SCIEX, ThermoFinnigan, Waters/Micromass, Bruker and JEOL, and is available for OEMs and directsales around the world.Matrix Science Booth 70464 Baker StreetLondon, W1U 7GBUnited KingdomP +1 800-716-6702F +1 800-716-6704info@matrixscience.comwww.matrixscience.comTake the guesswork out <strong>of</strong> protein identification with MascotServer. Get closer to your raw data with Mascot Distiller, givingdirect access to all popular file formats for peak picking, de novosequencing, quantitation, and more. Bring it all together withMascot Integra, a relational database system for proteomics.Michrom Bioresources Booth 6151945 Industrial DriveAuburn, California 95603United StatesP +1 530-888-6498F +1 530-888-8295michrommkt@michrom.comwww.michrom.comMichrom Bioresources is a premier supplier <strong>of</strong> LCMS instrumentation,accessories, and consumables for life science researchersaround the world. Michrom’s new Advance splitless nano-capillaryUHPLC and revolutionary “Plug and Play” CaptiveSprayionization source interface to most MS instruments, providingoptimum LCMS performance for qualitative and quantitativeproteomics applications.Morehouse School <strong>of</strong>Medicine/RCMI <strong>Program</strong> Booth 204720 Westview Drive, SWAtlanta, GA 30310United StatesP +1 404-752-1500www.msm.eduMorehouse School <strong>of</strong> Medicine (MSM) is a private, historicallyblack institution established in 1975. Major support for the biomedicalresearch infrastructure at MSM is received through theNIH/NCRR funded RCMI <strong>Program</strong>. The MSM/RCMI supportedcore facilities are: Cell/Tissue Imaging, Gene Pr<strong>of</strong>iling, AnalyticalChemistry/Protein Pr<strong>of</strong>iling and Biomedical Technology ServiceLaboratories.New England Biolabs, Inc. Booth 205240 County RoadIpswich, MA 01938United StatesP+1 800-632-5227info@neb.comwww.neb.comNew England Biolabs, Inc. leads the industry in the discoveryand production <strong>of</strong> enzymes for molecular biology applicationsincluding sample preparation for next-generation sequencing.NEB’s global reputation for manufacturing products <strong>of</strong> the highestquality coupled with best in class technical support makesNEB a first choice for customers demanding optimized reagentsfor advanced technologies.New Objective Booth 2032 Constitution WayWoburn, MA 01801United StatesP +1 781-933-9560F +1 781-933-9564sales@newobjective.comwww.newobjective.comNew Objective brings state-<strong>of</strong>-the-art systems to nano- ESI-MSand capillary LC/MS. In <strong>2011</strong>, New Objective introduces the revolutionaryand rugged PicoFrit Chip. This integrated column/high-voltage system provides high-throughput sample analysiswithout compromising sensitivity or flexibility. Our industryprovenPicoView® sources and PicoFrit columns continue to providehigh-performance and sensitivity in LC-MS.112 • <strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine


Nonlinear Dynamics Booth 1052530 Meridian Parkway, 3rd FloorDurham, NC 27713United StatesP +1 866-GELS-USAinfo@nonlinear.comwww.nonlinear.comNonlinear Dynamics’ range comprises analysis s<strong>of</strong>tware for label-freeLC-MS, 2D electrophoresis gels, and MALDI data as wellas tools for multivariate statistical analysis. Our unique analysisapproach has transformed proteomics research — it providesfaster more accurate quantitation and has enabled cross-lab reproducibility.Omega Bio-Tek, Inc. Booth 6051850-E Beaver Ridge CircleNorcross, GA 30071United StatesP +1 770-931-8400info@omegabiotek.comwww.omegabiotek.comOmega Bio-Tek manufactures a complete line <strong>of</strong> DNA|RNA isolationkits utilizing magnetic beads and silica filter technology forboth high throughput facilities and individual labs. Our novel nospin, high-throughput magnetic bead based plasmid DNA purification,PCR cleanup and DTR provides an affordability andselection that is unmatched.OpGen, Inc. Booth 603708 Quince Orchard Road, #160Gaithersburg, MD 20878United StatesP +1 301-869-9683www.opgen.comOpGen, Inc. provides the Argus Optical Mapping System forgenerating high-resolution, ordered, whole-genome restrictionmaps from single DNA molecules. Independent <strong>of</strong> sequenceinformation, Optical Maps enable a comprehensive view <strong>of</strong> genomicarchitecture. Use Optical Maps for comparative genomics,strain typing and sequence assembly.OriGene Technologies, Inc. Booth 4009620 Medical Center DriveSuite 200Rockville, MD 20850United StatesP +1 888 267 4436F +1 301-340-8606techsupport@origene.comwww.origene.comOriGene Technologies, Inc., is a gene centric life sciences companydedicated to support academic, pharmaceutical and biotechcompanies in their research <strong>of</strong> gene functions and drugdiscovery. OriGene develops proteins, antibodies, and othermolecular tools to allow researchers to analyze their data ona multiplex level. OriGene’s novel product line includes theworld’s largest cDNA and shRNA clone collections, over 5,000purified human proteins, high quality monoclonal antibodies(TrueMAB TM ), 100,000 highly validated human tissues, and proteinmicroarray products and services. OriGene also provides abroad range <strong>of</strong> antibody validation products including genomewidetagged antigen standards and extensive IHC slides derivedfrom our tissue collection.Peak Scientific, Inc. Booth 10119 Sterling RoadBoston, MA 01862United StatesP +1 866-647-1649F +1 978-608-9503infona@peakscientific.comwww.peakscientific.comOver the years our dynamic company has stimulated Peak designengineers into some remarkable developments in the manufacture<strong>of</strong> Laboratory Gas Generators. Smoother, quieter andmore efficient than anything on the market, our Generators <strong>of</strong>fersuperb technical performance in the lab and our world-class aftersales service comes as standard.Phenix Research Products Booth 51873 Ridgeway RoadCandler, NC 28715United StatesP +1 800-767-0665F +1 828-670-7020sales@phenixresearch.comwww.PhenixResearch.comPHENIX Research was founded in 1990 and is recognized as atechnical leader in providing outstanding laboratory productsat market beating prices. Our core competency and focus is toprovide value added solutions to life science researchers in University,Biotech and Government settings. We <strong>of</strong>fer outstandingcustomer service 8:00 am - 5:30 pm EST and a nationwide directtechnical sales force.Exhibitor Directory<strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine • 113


Exhibitor Directory — ContinuedExhibitor DirectoryPhotometrics Booth 5163440 East Britannia DriveTucson, AZ 85706United StatesP +1 520-889-9933F +1 520-573-1944info@photometrics.comwww.photometrics.comDeveloper <strong>of</strong> the new Evolve 128, Photometrics is the world’spremier designer and manufacturer <strong>of</strong> high-performance CCDand EMCCD cameras for life science research. Photometrics fordecades has led the industry with state-<strong>of</strong>-the-art imaging instrumentationfor the scientific community. Researchers globallyrely on Photometrics imaging solutions to meet their demandingrequirements.PolyLC Inc. Booth 2229151 Rumsey Road,Suite 175Columbia, MD 21045United StatesP +1 410-992-5400F +1 410-730-8340info@polylc.comwww.polylc.comUnique HPLC columns for life sciences. Featuring ERLIC! 1) Selectiveisolation and separation <strong>of</strong> tryptic phosphopeptides andglycopeptides; 2) Fractionation <strong>of</strong> tryptic peptides by isoelectricpoint in volatile solvents. Products for proteomics: PolySULFOE-THYL A for 2-D SCX-RPC separations; mixed-bed ion-exchangeand other columns for predigest fractionation <strong>of</strong> intact proteins,including histones and membrane proteins, metabolomics, andQC <strong>of</strong> biotech protein variants.Protea Biosciences, Inc. Booth 503955 Hartman Run RoadMorgantown, WV 26507United StatesP +1 877-776-8321F +1 304-292-7101www.proteabio.comWe are a diversified biotechnology company that applies novelbioanalytical technologies to the development <strong>of</strong> new pharmaceuticals,products and services. Our products and services empowerResearchers with new capabilities in their own laboratoryto improve the speed, quality and reproducibility <strong>of</strong> proteinmass spectrometry data obtained from their biological samples.Proteome S<strong>of</strong>tware Booth 5141340 SW Bertha BoulevardSuite #10Portland, OR 97219United StatesP +1 800-944-6027F +1 928-244-6024jana.lee@proteomes<strong>of</strong>tware.comwww.proeomes<strong>of</strong>tware.comProteome S<strong>of</strong>tware produces industry standard products forMS/MS-based proteomics. Scaffold identifies biologically importantresults by comparing data from Mascot, SEQUEST, ProteomeDiscoverer, IdentityE, SpectrumMill, Phenyx, OMSSA andX!Tandem. Scaffold Q+ adds iTRAQ and TMT quantification.MassQC monitors and troubleshoots LC-MS performance. ScaffoldPTM determines sites <strong>of</strong> phosphorylation and other modifications.Randox Pharma Services Booth 30255 Diamond RoadCrumlin C. Antrim, BT29 4QYUnited KingdomP +44 (0) 2894-422 413F +44 (0) 2894-452 912marketing@randox.comwww.randoxpharmaservices.comRandox Pharma Services <strong>of</strong>fers the most comprehensive range<strong>of</strong> biomarker solutions available to central labs, CRO’s and pharmaceuticalcompanies worldwide. Our award-winning, FDA approved,Biochip Array Technology is recognized as a ‘gold standard’in biomarker multiplexing for the analysis <strong>of</strong> proteins, genemutations, gene expression and SNP analysis.Research Scientific Services Booth 216P.O. Box 9188Gaithersburg, MD 20898United StatesP +1 301-977-9344F +1 301-977-9572jpeter@resci.orgwww.resci.comResearch Scientific Services (RSS) has provided pr<strong>of</strong>essionalservice on Waters and AB Sciex mass spectrometers and sellingrefurbished LC/MS/MS instruments for 20 years. Service isprovided by a nationwide network <strong>of</strong> highly experienced formerOEM engineers. Customer attentiveness and responsive serviceat a competitive price is why laboratories trust RSS service.114 • <strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine


Roche Applied Science Booth 5009115 Hague RoadIndianapolis, IN 46250United StatesP +1 800-262-1640www.roche-applied-science.comRoche Applied Science’s new GS Junior system uses 454’s ultrafastpyrosequencing technology to generate 400 to 500 bp sequencingread lengths for amplicon and transcriptome sequencingprojects. Ask about the newest version <strong>of</strong> our SequenceCapture sequence-enrichment technology, plus other new NimbleGenmicroarray products. Investigate real-time PCR with theLightCycler® 480 and LightCycler® 1536 Instruments, plus theaddition <strong>of</strong> rat and mouse targets to our preplated qPCR assays.Sage Science Booth 617500 Cummings CenterSuite 3150Beverly, MA 1915United StatesP +1 978-922-1832F +1 617-812-0540info@sagescience.comwww.sagescience.comSage Science manufactures the Pippin Prep TM DNA size selectionsystem. The Pippin Prep automates size selection <strong>of</strong> DNAfor next-gen sequencing library preparations, providing a laborsavingalternative to manual gel extraction and purification. Inaddition to time savings, one can achieve higher yields and eliminatelower molecular weight adapter contamination.SeqGen, Inc. Booth 6041725 Del Amo BoulevardTorrance, CA 90501United StatesP +1 877-377-3743F +1 800-790-4830info@seqgen.netwww.seqgen.netSeqGen is a full service provider <strong>of</strong> high quality, refurbished,DNA Sequencers, PCR Instrumentation, and Service Contracts.We maintain a large inventory <strong>of</strong> equipment/parts and havemany years <strong>of</strong> experience supporting Biotech laboratories. Ourexpert SeqGen Engineers conduct installations, warranty calls,service contracts, and paid day service calls.SeqWright, Inc. Booth 7022575 West Bellfort, Suite 2001Houston, TX 77054United StatesP +1 800-720-4363F +1 713-528-6232sales@seqwright.comwww.seqwright.comSeqWright, a CLIA certified and GLP compliant facility, providesFDA, Clinical, and Research-level Genomic services to the Pharmaceutical,Biotechnology and Academic communities. Specializingin Next Generation Genomics, Sequencing, ExpressionAnalysis and Molecular Biology services, SeqWright has earneda reputation for quality, technical expertise and a willingness toprovide client-specific solutions.Sigma Life Science Booth 5053050 Spruce StreetSt. Louis, MO 63103United StatesP +1 314-771-5765custserv@sial.comwww.wherebiobegins.comSIGMA® Life Science <strong>of</strong>fers a wide portfolio <strong>of</strong> innovative technologies,products and services spanning cell biology, protein assays,genomics, functional genomics, biomolecules, epigenetics,stem cell research and transgenic animal models. Through continuedinvestment in innovation and quality we are the leadingdestination for life science researchers to access deep biologicalinformation and market leading products that improve the quality<strong>of</strong> life.S<strong>of</strong>tGenetics Booth 110100 Oakwood AveSuite 350State College, PA 16803United StatesP +1 814-237-9340F +1 814-237-9343info@s<strong>of</strong>tgenetics.comwww.s<strong>of</strong>tgenetics.comFeaturing NextGENe® s<strong>of</strong>tware for analysis <strong>of</strong> all massively parallelsequencer data with newly included RNA-Seq module; enhancedcapabilities <strong>of</strong> functionalities added to GeneMarker®S<strong>of</strong>tware include carbohydrate analysis, new large sizing algorithms,kinship analysis <strong>of</strong> wild populations, genotype mergetool and project comparison tool. S<strong>of</strong>tGenetics is providingdemonstrations and 30-day trials.Exhibitor Directory<strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine • 115


Exhibitor Directory — ContinuedTecan Booth 4194022 Stirrup Creek DriveSuite 310Durham, NC 27703United StatesP+1 919-361-5200info@tecan.comwww.tecan.comTecan is a leading global provider <strong>of</strong> laboratory instruments andsolutions. The company specializes in the development, productionand distribution <strong>of</strong> instruments and automated workflowsolutions for laboratories in the life sciences sector. Its clients includepharmaceutical and biotechnology companies, universityresearch departments, forensic and diagnostic laboratories.Thermo Scientific Booth 200355 River Oaks ParkwaySan Jose, CA 95134United StatesP +1 800-532-4752analyze@therm<strong>of</strong>isher.comwww.thermoscientific.comThermo Scientific brand solutions leverage the market-leadingsample preparation products and ion trap mass spectrometersfor definitive quantitation <strong>of</strong> protein mixtures, greatly improvingprotein characterization, accelerating protein database searchesand reducing false identification rates.Vanderbilt UniversityMedical Center Booth 3193401 West End BuildingSuite 300Nashville, TN 37203-8375United StatesP +1 615-875-3363F +1 615-936-6530coresinfo@vanderbilt.eduwww.mc.vanderbilt.edu/CORESVICI Valco Instruments Booth 102P.O. Box 55603Houston, TX 77255United StatesP +1 800-347-8424F +1 713-956-3119sales_usa@vici.comwww.vici.comVICI Valco Instruments is a designer/ manufacturer <strong>of</strong> standard/custom valves and fittings for precision analytical, biomedical,and biocompatible instrumentation. Product variety includes;pneumatic and electric actuators, tubing, sampling loops, heatedenclosures, valve sequence and temperature controllers, gaspurifiers, GC detectors, and digital interfaces. Devices and instrumentationfor generation <strong>of</strong> calibration gas standards, containmenttraps and gas specific purifiers.Waters Corporation Booth 10834 Maple StreetMilford, MA 01757United StatesP +1 508-478-2000F +1 508-872-1990info@waters.comwww.waters.comWaters helps laboratory-dependent organizations by providingbreakthrough technologies and solutions. Pioneering a connectedportfolio <strong>of</strong> separation and analytical science, laboratoryinformatics and mass spectrometry, Waters provides the tools toimprove the quality <strong>of</strong> today’s science and explore the infinitepossibilities <strong>of</strong> tomorrow. Waters, The Science <strong>of</strong> What’s Possible.Vanderbilt University Medical Center (VUMC) is a comprehensivehealthcare facility. Vanderbilt’s mission is to advance healthand wellness through preeminent programs in patient care, education,and research. CORES (Core Ordering & Reporting EnterpriseSystem) is a web-based system developed by VUMC usedto process core billing and produce usage reporting.Exhibitor Directory116 • <strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine


Exhibit Hall FloorplanGrand Oaks Ballroom, Level 2Exhibit Hall Floorplan<strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine • 117


Exhibitor List in Alphabetical OrderExhibitors confirmed as <strong>of</strong> January 29, <strong>2011</strong>.M = <strong>ABRF</strong> Annual Meeting SponsorC = <strong>ABRF</strong> Corporate SponsorCompanyBoothAB SCIEX .................................................................................................501<strong>ABRF</strong> .........................................................................................................317Advanced Analytical ...........................................................................602Agilent Technologies M C ............................................................301AnaSpec, Eurogentec Group M C ..............................................417Bioinformatics Solutions Inc. (BSI)...................................................402Bioo S cientific ........................................................................................718Bio-Synthesis, Inc. .................................................................................104Bruker Daltonics Inc. ............................................................................214Caliper Life Sciences C ....................................................................304CEM C orporation .................................................................................303chemagen U SA ......................................................................................100Dionex Corporation ............................................................................215DNAnexus ..............................................................................................404eagle-i Consortium ..............................................................................221EdgeBio C ............................................................................................300FASEB MARC <strong>Program</strong> ........................................................................416Fluidigm Corporation .........................................................................614GE H ealthcare ........................................................................................714Gene Codes Corporation ..................................................................220Genetix ....................................................................................................616GENEWIZ, I nc. .......................................................................................316Genomatix S<strong>of</strong>tware, Inc ....................................................................218Geospiza .................................................................................................519Hamilton Robotics ...............................................................................223iLab Solutions, LLC ...............................................................................700Illumina M C ......................................................................................515Intavis .......................................................................................................600IntegenX, Inc. C ..................................................................................201Integrated DNA Technologies .........................................................305Kapa Biosystems, Inc. ..........................................................................619LC S ciences .............................................................................................318CompanyBoothLeica M icrosystems ..............................................................................217Life Technologies .................................................................................601MassTech, In c. ........................................................................................103Matrix Science M ................................................................................704Michrom B ioresources ........................................................................615Morehouse School <strong>of</strong> Medicine/RCMI <strong>Program</strong> ........................204New England Biolabs, Inc. ..................................................................205New O bjective ......................................................................................203Nonlinear D ynamics ............................................................................105Omega Bio-Tek .....................................................................................605OpGen, In c. ............................................................................................603OriGene Technologies, Inc. ..............................................................400Peak Scientific, Inc. ...............................................................................101Phenix Research Products ..................................................................518Photometrics ..........................................................................................516PolyLC Inc. ..............................................................................................222Protea Biosciences, Inc. .......................................................................503Proteome S <strong>of</strong>tware ..............................................................................514Randox Pharma Services ....................................................................302Research Scientific Services...............................................................216Roche Applied Science M C ........................................................500Sage Science ..........................................................................................617SeqGen, In c. ...........................................................................................604SeqWright, In c. ......................................................................................702Sigma Life Science ................................................................................505S<strong>of</strong>tGenetics...........................................................................................110Tecan ........................................................................................................419Thermo Scientific .................................................................................200Vanderbilt University Medical Center ..........................................319VICI Valco Instruments .......................................................................102Waters Corporation M C ..............................................................108Exhibitor List – Alpha118 • <strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine


Exhibitor List in Booth OrderExhibitors confirmed as <strong>of</strong> January 29, <strong>2011</strong>.M = <strong>ABRF</strong> Annual Meeting SponsorC = <strong>ABRF</strong> Corporate SponsorBooth Company100 .................. chemagen U SA101 .................. Peak Scientific, Inc.102 .................. VICI Valco Instruments103 .................. MassTech, In c.104 .................. Bio-Synthesis, I nc.105 .................. Nonlinear D ynamics108 .................. Waters C orporation M C110 .................. S<strong>of</strong>tGenetics200 .................. Thermo S cientific201 .................. IntegenX, In c. C203 .................. New O bjective204 .................. Morehouse School <strong>of</strong> Medicine/RCMI <strong>Program</strong>205 .................. New England Biolabs, Inc.214 .................. Bruker Daltonics Inc.215 .................. Dionex C orporation216 .................. Research Scientific Services217 .................. Leica M icrosystems218 .................. Genomatix S<strong>of</strong>tware, Inc.220 .................. Gene Codes Corporation221 .................. eagle-i C onsortium222 .................. PolyLC Inc.223 .................. Hamilton R obotics300 .................. EdgeBio C301 .................. Agilent Technologies M C302 .................. Randox Pharma Services303 .................. CEM C orporation304 .................. Caliper Life Sciences C305 .................. Integrated DNA Technologies316 .................. GENEWIZ, I nc.317 .................. <strong>ABRF</strong>318 .................. LC S ciences319 .................. Vanderbilt University Medical CenterBooth Company400 .................. OriGene Technologies, Inc.402 .................. Bioinformatics Solutions Inc. (BSI)404 .................. DNAnexus416 .................. FASEB MARC <strong>Program</strong>417 .................. AnaSpec, Eurogentec Group M C419 .................. Tecan500 .................. Roche Applied Science M C501 .................. AB S CIEX503 .................. Protea Biosciences, Inc.505 .................. Sigma Life Science514 .................. Proteome S <strong>of</strong>tware515 .................. Illumina M C516 .................. Photometrics518 .................. Phenix Research Products519 .................. Geospiza600 .................. Intavis601 .................. Life Technologies602 .................. Advanced A nalytical603 .................. OpGen, In c.604 .................. SeqGen, In c.605 .................. Omega B io-Tek614 .................. Fluidigm C orporation615 .................. Michrom B ioresources616 .................. Genetix617 .................. Sage Science619 .................. Kapa Biosystems, Inc.700 .................. iLab Solutions, LLC702 .................. SeqWright, Inc.704 .................. Matrix S cience M714 .................. GE H ealthcare718 .................. Bioo ScientificExhibitor List – Booth<strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine • 119


Vendor PresentationsSunday, February 20 — 12:00 pm – 1:30 pmAlyssum Room, Level 3Exhibit Booth: 214Moving Beyond ProteomicsSpeaker: Detlev Suckau, Bruker Daltonik GmbHIn current proteomics studies as well as biopharmaceutical developmentand characterization, post-translational modificationsobtain increasing attention. Amongst these, glycosylation isprominent but the lack <strong>of</strong> bioinformatics solutions slowed downthe analysis <strong>of</strong> glycoproteins in both, the biopharmaceutical industryas well as in proteomics studies.In this seminar, we introduce new solutions that include a newglycoprotein and glycoproteomics enabling bioinformatics platformand show examples from selected glycoprotein characterizationsincluding glycan and direct glycopeptide analysis usingBrukers leading ESI-MSn and MALDI platforms.Dedicated solutions for the development and quality control <strong>of</strong>Biologicals and Biosimilars will be described that show you howto rapidly characterize therapeutic proteins with the confidencethat is derived from the high specificity <strong>of</strong> Brukers ultra-highresolution QTOF maXis.As a further extension <strong>of</strong> classical protein characterization andvalidation workflows, we describe recent advances in Top-Downprotein characterization tools that will make your C- and N-terminaldefinitions specific and rapid. The ESI-ETD and MALDITop-Down Sequencing approaches will also forward your proteomicsresearch as they provide simultaneous access to PTManalysis, protein is<strong>of</strong>orm differentiation and proteolytic regulationevents that are not addressed with current technologies.Grand Oaks Ballroom,Rooms N&O, Level 2Exhibit Booth: 71412:00 pm – 12:30 pmAdvances in Sample Preparation <strong>of</strong> LowAbundant Proteins using Magnetic BeadsSpeaker: Helena Hedlund, Global Product Manager, GEHealthcare Life SciencesLow abundant proteins are <strong>of</strong>ten biologically important butdifficult to detect. Some proteins are present in a few copieswhereas others have a short transient presence. To be able toincrease the detection and identification rate, efficient samplepreparation methods are necessary. We have combined establishedaffinity chromatography methods with magnetic beadtechnology for the enrichment <strong>of</strong> such challenging proteins. Inthe first study changes in tyrosine phoshorylation in cancer cellsupon drug treatment was investigated. Immmonoprecipitationusing Protein G Mag Sepharose TM was used for enrichment <strong>of</strong>phosphorylated proteins prior to a 2-D DIGE experiment. Theeffect <strong>of</strong> the treatment will be presented as well as protein identificationfrom MS analysis.In the second study screening for optimal purification conditionsfor a number <strong>of</strong> integral membrane proteins was performed. Vitalparameters as choice <strong>of</strong> detergent wash and elution bufferconcentrations were evaluated. All experiments were done usingNi2+-charged His Mag Sepharose Ni. Results from screeningfor several detergents gave reliable results that could be fed int<strong>of</strong>urther scale-up experiments. The magnetic bead format simplifiedand shortened the handling procedures through-out thesetwo different workflows.Vendor Presentations12:30 pm – 1:00 pmQuantitative Western Blotting withAmersham TM ECL TM PrimeSpeaker: Maria Winkvist, Scientist, GE Healthcare LifeSciencesWestern blotting is a well established technique in life scienceresearch. Traditionally the technique has been restricted toqualitative protein analysis. However, development <strong>of</strong> refineddetection methods and reagents has opened up the possibilityto use the technique in quantitative analysis, for accurate monitoring<strong>of</strong> small changes in protein abundance and for detection<strong>of</strong> posttranslational modifications.120 • <strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine


This workshop will present ways to achieve quantitative Westernblotting by optimizing the imaging system and detection reagent.The impact <strong>of</strong> normalization, signal-to-noise, backgroundnoise and other experimental factors will also be discussed.1:00 pm – 1:30 pm2-D DIGE Analysis <strong>of</strong> Multicellular TumorSpheroids in Evaluation <strong>of</strong> Breast CancerTreatmentSpeaker: Viola Ruddat, Ph.D., Senior Applications Scientist,GE Healthcare Life SciencesMany cancers can be diagnosed using Positron Emission Tomography(PET) to visualize tumors. PET can also be used to monitorhow effective various treatments are in individual patientsby observing the decrease in tumor tissue. Tumor spheroids arecancer cells grown on agar coated dishes forming a 3D structure.They are widely used in preclinical cancer research, where themulticellular tumor spheroid model is considered biologicallyand physiologically similar to in vivo grown tumors.Grand Oaks Ballroom,Rooms P&Q, Level 2Exhibit Booth: 515Illumina’s New <strong>2011</strong> Portfolio andAnnouncementsIllumina’s broad portfolio <strong>of</strong> sequencing and array solutionsempowers scientists in labs <strong>of</strong> all sizes to take their researchfurther, faster. Please join us to hear the details about the recentannouncements <strong>of</strong> the terabase sequencing milestone, ourability to enable our customers to reach 600 Gb/run this spring,and our newest Platform, MiSeq.We’ll also describe our progressive research path for GWAS,our sequencing service without compromise and Eco, our ultraaccessibleqPCR system.In this workshop, we will present how we have used Two-dimensionalDifference Gel Electrophoresis (2-D DIGE) analysis to gainmore insight in changes <strong>of</strong> protein expression as a result <strong>of</strong> drugtreatment <strong>of</strong> multicellular tumor spheroids.Monday, February 21 — 12:00 pm – 1:30 pmGrand Oaks Ballroom,Rooms R&S, Level 2Exhibit Booth: 616Advanced Technologies for AntibodyDiscovery and Cell ScreeningSpeakers: Mark Truesdale, Gabe Longoria and AlisonGlaserThe use <strong>of</strong> antibodies in investigating and treating diseases isuniversal, however traditional methods for antibody developmentare time consuming and costly. The Genetix ClonePix TM FLsystem improves operational efficiency by reducing timelines,minimizing resource requirement and allows higher throughput<strong>of</strong> projects.Understanding disease mechanisms through the culture andstudy <strong>of</strong> relevant cell lines requires multiple technologies. Imagingsystems from Genetix are used to characterize cell growthand quantify the responses <strong>of</strong> cells to different treatments. Forrapid determination <strong>of</strong> cell growth in microwell plates, Clone-Select Imager TM provides objective, quantitative and consistentcharacterization <strong>of</strong> cell growth prior to investigation <strong>of</strong> cellularresponses to different treatments such as siRNA, small moleculeor antibody. To determine the responses <strong>of</strong> cells, CellReporterTM is used for fluorescent cell- and bead-based assays. Intuitiveworkflows provide flexibility to optimize and standardize assaysfrom image acquisition through to data analysis. Typical applicationsinclude cell-based assays for cell cycle analysis, apoptosisor monitoring protein translocation and bead-based homogeneousassays for quantifying cellular protein production.This seminar will present the latest technologies and applicationsfor screening cells, identification and isolation <strong>of</strong> clones,and data-rich cell based assays.Vendor Presentations<strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine • 121


Vendor Presentations — ContinuedMonday, February 21 — 12:00 pm – 1:30 pm (Continued)Vendor PresentationsGrand Oaks Ballroom,Rooms N&O, Level 2Exhibit Booth: 601From Discovery to TranslationalGenomics with Life Technologies’Comprehensive Sequencing SolutionsLife Technologies <strong>of</strong>fers the most comprehensive and complementaryportfolio <strong>of</strong> research sequencing solutions — fromthe market leading Capillary Electrophoresis solutions, throughthe recently launched Ion Torrent Personal Genome Machine,(PGM) to SOLiD TM systems — with the 5500 series SOLiD TM systemsbecoming available soon. The re-engineered 5500 seriesSOLiD TM system generates up to 30 gigabases <strong>of</strong> mapped dataper day and can be used in a myriad <strong>of</strong> research applications— including complete cancer genome re-sequencing, single celltranscriptome analysis, interrogation <strong>of</strong> methylation status, andde novo sequencing. The increasing throughput <strong>of</strong> the evolvingSOLiD TM platform and other next generation sequencingtechnologies now makes it possible to sequence a whole humangenome. As additional human genomes are sequenced, newbiological insights are obtained, and we begin to truly appreciatethe critical need for sequencing accuracy. Consensus accuracycan overcome some inherent error rates, however a flooris ultimately reached, thereby limiting identification <strong>of</strong> low frequencysomatic mutations or SNV’s when using low coveragetechniques for GWAS based projects. A new chemistry incorporatingerror-correction codes has been developed that enablesthe SOLiD TM system to achieve 99.99% accuracy within a singlesequencing read. This, and other technological improvements,will be discussed in the context <strong>of</strong> the emerging “TranslationalGenomics” era that we have recently entered.PostLight Sequencing with Semiconductor ChipsIon Torrent has invented the first device—a new semiconductorchip—capable <strong>of</strong> directly translating chemical signals intodigital information. The first application <strong>of</strong> this technology is sequencingDNA. The device leverages decades <strong>of</strong> semiconductortechnology advances, and in just a few years has brought theentire design, fabrication and supply chain infrastructure <strong>of</strong> thatindustry—a trillion dollar investment—to bear on the challenge<strong>of</strong> sequencing. The result is Ion semiconductor sequencing, thefirst commercial sequencing technologythat does not use light,and as a result delivers unprecedented speed, scalability andlow cost.Next Generation qPCR solutions from Life TechnologiesIn 2010, Life Technologies TM released the Applied Biosystems TMViia TM 7 Real-Time PCR System. This seventh generation systemcombines advances in optical and thermal cycling technologyto provide one the most powerful and flexible Real-Time PCRinstruments available on the market today. After a brief review<strong>of</strong> the Viia TM 7 Real-Time PCR System, we will explore the nextgeneration <strong>of</strong> qPCR systems.The OpenArray® Real-Time PCR platform employs 3072 wellreaction plates to increase throughput and to lower costs forqPCR. The throughput and cost efficiencies <strong>of</strong> the OpenArray®Real-Time PCR platform are enabling applications such as Next-Gen Sequencing library quantitation by digital PCR, gene expressionpathway analysis, high-throughput genotyping studies,and miRNA pr<strong>of</strong>iling. The OpenArray® Real-Time PCR platformdelivers TaqMan® sensitivity and specificity in miRNA pr<strong>of</strong>ilingwhile achieving cost savings, comparable to microarray systems.The throughput for miRNA pr<strong>of</strong>iling is up to 9 samples completedin a 2.5 hour run, and up to 36 samples per day.The next generation <strong>of</strong> Life Technologies’ TM qPCR systems alsoincludes a novel technology platform based upon a continuousflow liquid-bridge manifold. This will enable high throughputGenotyping and Gene Expression applications. The Stokes Bionanodroplet qPCR technology will be reviewed during this talk.Alyssum Room, Level 3Exhibit Booth: 500Amplicon Sequencing with GS FLX andGS Junior SystemsAccess Array TM System High-Throughput ResequencingSpeaker: Steven H<strong>of</strong>fman, Fluidigm CorporationFlexible Next-Generation Sequencing Approaches Enabledby the GS FLX and GS Junior SystemsSpeaker: Bruce Taillon, Ph.D., 454 Life SciencesFeaturing GS FLX Titanium series chemistry, the Genome SequencerFLX and GS Junior Systems <strong>of</strong>fer a powerful combination<strong>of</strong> long sequencing reads (400 to 500 base pairs) and dedicatedGS Amplicon Variant Analyzer (AVA) s<strong>of</strong>tware, enablingthe sequencing, alignment, and analysis <strong>of</strong> amplicons against areference sequence.122 • <strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine


Grand Oaks Ballroom,Rooms P&Q, Level 2Exhibit Booth: 505Controlling Experimental VariablesSpeaker: Jim Walters, Ph.D., Principal Investigator, AnalyticalResearch Department, Sigma Life ScienceThe enormous amount <strong>of</strong> data generated from complex proteomicanalyses with mass spectrometry is <strong>of</strong>ten overwhelming;requiring iterative analysis and optimization <strong>of</strong> a number <strong>of</strong> variables.To counteract the effects <strong>of</strong> potentially false data, a number<strong>of</strong> defined protein and peptide standards have been developedto allow for universal performance comparisons acrossplatforms and between labs. We will highlight our efforts in thedevelopment, commercialization, and utilization <strong>of</strong> proteomicsstandards. We will discuss our newly developed standards whichinclude both phosphopeptide standards as well as a syntheticprotein designed to fully assess standard LC-MS platforms.Begonia Room, Level 3Exhibit Booth: 108Recent Developments in WatersProteomics TechnologySpeaker: Martha Stapels, Principle Research Scientist,WatersWe will be discussing advances in ETD, Ion Mobility andseparations. Also featured will be a presentation by J. WillThompson from Duke University: “Absolute Quantitation Using2DLC-MSE and Species-Specific Correction to InvestigateChlamydia trachomatis Developmental Forms.”Monday, February 21 — 7:30 pm – 9:00 pmAlyssum Room, Level 3Exhibit Booth: 500Reviews <strong>of</strong> SeqCap EZ Exome v2.0Vendor Presentations<strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine • 123


Vendor Presentations — ContinuedTuesday, February 22 — 12:00 pm – 1:30 pmAlyssum Room, Level 3Exhibit Booth: 214The Latest Solutions for Metabolomicsand Small Molecule ResearchSpeaker: Aiko Barsch, Bruker Daltonik GmbHMetabolomics nowadays has several analytical challenges toovercome. The identification <strong>of</strong> metabolites is <strong>of</strong>ten defined asthe major bottleneck in metabolomics applications as well as ingeneral small molecule research.The confident generation <strong>of</strong> molecular formulae is the first stepin the identification <strong>of</strong> unknowns. Especially for m/z values above500, the number <strong>of</strong> ambiguous assignments increases exponentially.maXis TM Ultra high resolution Q-TOF accurate mass andisotopic pattern data from MS and MS/MS spectra significantlyextends the m/z range for reliable formula suggestions. The molecularformula can then be queried against public databasesto quickly match already known compounds. Hyphenating gaschromatography with s<strong>of</strong>t atmospheric pressure ionisation(APCI) to high resolution Q-TOF-MS technology can preservethe molecular ion information which enables the identification<strong>of</strong> unknowns in GC-MS data.One crucial advantage <strong>of</strong> applying maXis UHR-TOF in untargetedmetabolomic studies is the speed <strong>of</strong> analysis by couplingU-HPLC systems without compromising the performance interms <strong>of</strong> mass accuracy or resolution even at high acquisitionrates up to 20Hz. Overall, the sample throughput is increased,thus addressing the necessity <strong>of</strong> handling large sample numbersneeded for statistically relevant data.Grand Oaks Ballroom,Rooms N&O, Level 2Exhibit Booth: 501Breakthrough Solutions for Proteomics,Biomarker, and Lipidomics ResearchSpeakers: Doug Simmons and Jenny Albanese, AB SCIEXJoin us for lunch and learn about powerful new breakthrough innovationsthat can really boost your productivity in proteomics,biomarker, and lipidomics research!Begonia Room, Level 3Exhibit Booth: 108Recent Developments in WatersMetabolomics and Lipidomics BiomarkerTechnologiesSpeaker: Henry Shion, Principal Applications Scientist,WatersWe will be discussing innovations in metabolomic/lipidomicseparations with UPLC, comprehensive data collection with MSEand QuanTOF technology, and advanced data processing anddatabase searching.Vendor Presentations124 • <strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine


NotesNotes<strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine • 125


Demos (as <strong>of</strong> January 29, <strong>2011</strong>)Monday, February 21 — 12:00 pm – 12:15 pmExhibit Hall Demo StageMonday, February 21 — 12:30 pm – 12:45 pmExhibit Hall Demo StageExhibit Booth: 503DemosInnovations in protein massspectrometry from Protea Biosciences:New bioanalytical tools for proteinanalysis and characterizationSpeakers: Christopher A. Bolcato, Technical ServicesManager, and Haddon E. Goodman, Product MarketingManagerPresentation will include discussion around innovative technologiesand techniques in gel protein separation and recovery, highabundant protein depletion, and new surfactant technologydeveloped by Protea Biosciences. Several new products will bediscussed that aid in the preparation <strong>of</strong> protein samples for massspectrometry.Monday, February 21 — 12:15 pm – 12:30 pmExhibit Hall Demo StageExhibit Booth: 515Illumina’s Genotyping Portfolio Featuringa New FFPE SolutionThe revolutionary Infinium HD FFPE Restore kit has expandedIllumina’s broad portfolio <strong>of</strong> array solutions to enable genotyping<strong>of</strong> formalin-fixed, paraffin-embedded (FFPE) samples on theInfinium platform. Join us to hear more about this exciting optionavailable for the CyotSNP and OmniExpress arrays. We’ll alsodescribe the latest evolution <strong>of</strong> iSelect, the most flexible customgenotyping option available today, and give a brief update onthe Omni GWAS Roadmap.Comprehensive Genomic Tools fromClinical Research to TranslationalMedicine: Paving the Way forPersonalized MedicineSpeaker: Jessica Parra, Ph.D., Marketing Manager, GlobalField MarketingClinical researchers are making personalized medicine a realityby translating biomarker discoveries into clinical diagnostictests. Although many biomarker signatures have been identifiedin research labs, only a few microarray-based clinical diagnostictests have received clearance for commercialization. Clinical researchersface many challenges from the discovery process tovalidation and routine testing. Using a single platform from clinicalresearch discovery to clinical applications helps researchersto overcome the operational hurdles <strong>of</strong> commercialization. Affymetrixis paving the path to market with a toolkit that includesinstrumentation, arrays, and reagents for use in developing clinicaldiagnostic tests. The convenience <strong>of</strong> a cGMP-manufacturedkit helps to reduce rework and risk during the developmentprocess to fast-track commercializationMonday, February 21 — 1:00 pm – 1:15 pmExhibit Hall Demo StageExhibit Booth: 110NextGENe’s RNA-Seq Analysis toolSpeaker: Megan R. ManionNextGENe’s RNA-Seq Analysis tool, which includes a proprietaryalignment algorithm specifically designed for transcriptomedata, will be presented. Project set-up and results will be shown.Results, which include mutation detected, expression analysisand alternative splicing analysis, are displayed in the interactiveNextGENe Viewer which includes detailed annotation. Next-GENe is a standalone Windows-based program for the analysis<strong>of</strong> Next Generation Sequencing data and is compatible withdata from Illumina Genome Analyzers, Roche/454 GS FLX, FLXTitanium and Junior, the SOLiD System and Ion Torrent’s IonPersonal Genome Machine. In addition to RNA-Seq analysis,NextGENe can also be used for SNP/Indel detection, ChIP-Seq,miRNA analysis, de novo assembly, and metagenomic analyses.126 • <strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine


Tuesday, February 22 — 12:00 pm – 12:15 pmExhibit Hall Demo StageExhibit Booth: 515My samples. My study. MiSeq TM .Introducing Illumina’s personalsequencing system.The MiSeq personal sequencing system delivers the fastest timeto answer, a revolutionary workflow, and the widest breadth <strong>of</strong>sequencing applications, all in a compact and economical instrument.This session will provide an introduction to the instrumentand showcase the revolutionary workflow — come experience itfor yourself.Demos<strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine • 127


Membership Application<strong>Association</strong> <strong>of</strong> <strong>Biomolecular</strong> Resource Facilities9650 Rockville Pike, Bethesda, MD 20814-3998 (USA)Telephone: (301) 634-7306 Fax: (301) 634-7455 Email: abrf@abrf.orgContact InformationYour NameCompany/InstitutionAddressCity State Zip CountryPhoneEmailFaxReferring <strong>ABRF</strong> Member NameImportant! Your membership confirmation will be sent via e-mail, along with other <strong>ABRF</strong> communications.Membership OptionsPlease check one: Member, $100.00 Student Member*, $35.00*To qualify for Student membership you must be a full-time undergraduate, graduate or medical student. Your department chair orstudent advisor must complete the following information:I certify that the applicant is a full-time student inEmail address <strong>of</strong> Department Chair/Major AdvisorSignature <strong>of</strong> Department Chair or Major AdvisorAdditional InformationAre you part <strong>of</strong> a resource laboratory? Yes NoIf yes, please list your director/manager’s name:Facility Name:Payment MethodPlease return this form with your full payment to assure proper credit. <strong>ABRF</strong> Tax ID: 56-1659510. Check enclosed for full amount made payable to <strong>ABRF</strong>. (Check must be in U.S. dollars and drawn on U.S. Bank.) Credit card (full amount will be charged): VISA MasterCard American ExpressMembershipApplicationCredit Card Number Expiration Date / /Signature <strong>of</strong> CardholderSend this form with payment to: <strong>ABRF</strong>, 9650 Rockville Pike, Bethesda, MD 20850-3998 (USA)Fax: (301) 634-7455 (Faxed membership forms must include credit card information.)128 • <strong>ABRF</strong> <strong>2011</strong> — Technologies to Enable Personalized Medicine


MARCH 16-20, 2012 • DISNEY’S CONTEMPORARY RESORT • ORLANDO, FLORIDAMARK YOURCALENDARS


The Your partner <strong>of</strong> in choicefor translating genomicdiscoveries intoclinical toolsWith Partnering Affymetrix with solutions Affymetrix for for medical your medical genomics, genomics corelabs research can <strong>of</strong>fer gives clinical you access researchers to a broad a broad range range <strong>of</strong> tech-<strong>of</strong>technologies, expertise, and and experience to to help help turn turn yourtheir findings into into reliable reliable clinical clinical tools. toolsAffymetrix Take advantage <strong>of</strong>fers: <strong>of</strong> the expertise thatA platform Affymetrix for <strong>of</strong>fers all applications to help advance – Solutions your research: for everystep in the biomarker discovery, qualification, andvalidation A platform process for all applications – Solutions forevery step in the biomarker discovery, qualification,Extensive and validation scientific process validation – More than22,000 peer-reviewed papers published usingour Extensive technology scientific validation – More than22,000 peer-reviewed papers published usingProven our technology path to market – Multiple diagnosticproducts launched through the Powered byAffymetrix Proven path <strong>Program</strong> to market – Multiple diagnosticproducts launched through the Powered byThe Affymetrix first FDA-cleared <strong>Program</strong>microarray system –The GeneChip ® System 3000 Dx v2 for moleculardiagnostic Visit www.affymetrix.com/medicalgenomics applicationstolearn now published scientists have advanced theirVisit translational www.affymetrix.com/medicalgenomics research using Affymetrix technology. to learnhow published scientists have advanced their translationalresearch using Affymetrix technology.Unless otherwise noted, Affymetrix products are for research use only and are notfor use in diagnostic procedures.For research use only. Not for use in diagnostic procedures.©<strong>2011</strong>-12 Affymetrix, Inc. All rights reserved.©2010 Affymetrix, Inc. All rights reserved.

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!