20.12.2012 Views

FR AB - Science Reference

FR AB - Science Reference

FR AB - Science Reference

SHOW MORE
SHOW LESS

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

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

VOLUME 11, ISSUE 4, DECEMBER 2000<br />

CONTAINS <strong>AB</strong>STRACTS <strong>FR</strong>OM <strong>AB</strong>RF 2001 MEETING<br />

JBTtgaacagatgatcaggtgaaatcacc<br />

gtctgaaatcacgtgaaatcacgtgaa<br />

THE aaatcaccaggtgaaaaatcacgtga<br />

caggtgaaagctaacagaggggggt<br />

OFFICIAL gacagtgctcggagaaattacaatac<br />

JOURNAL cgaggagaaatcctttcaatactcgag<br />

OF gagaaacttgacaatactagaaacttg<br />

THE acaatacagaaacttgacaatacaga<br />

Association aacctgaaaaatcaatcaggtgaaaa<br />

of atcaatcaggtgaaatcaatcaggtga<br />

Biomolecular aatcacatcaggtgaaatcacatcagg<br />

tgaaatcacatcaggtgagctaacgg<br />

Resource ggggttgacagtgctcgagaaatgttg<br />

Facilities acaatagaggagaaatttgaaataga<br />

ggagaaacttgaaatatcgaggagaa<br />

acttgatactcgaggagaaacttgac<br />

actcgaggagaaactgaatcacaga<br />

gatcaggtgaatcacagatgatcagg<br />

gatcacaaatgatcaggtgaatcacaa<br />

atgatcaggtgaaatcacaaatgatca<br />

ggtgaaatcacgaatgcaggtgaaag<br />

ctaacaaaggggggttgacagtgctc<br />

JOURNAL OF BIOMOLECULAR TECHNIQUES


RF <strong>AB</strong><br />

JBT<br />

JOURNAL OF BIOMOLECULAR TECHNIQUES<br />

Clive A. Slaughter<br />

St. Jude Children’s Research Hospital<br />

332 North Lauderdale Street<br />

Memphis, TN 38105-2794<br />

Tel: (901) 495-4844 Fax: (901) 495-2945<br />

email: clive.slaughter@stjude.org<br />

Ruth Angeletti<br />

Albert Einstein College<br />

of Medicine<br />

Kalyan R. Anumula<br />

SmithKline Beecham<br />

Pharmaceutical<br />

Lynda Bonewald<br />

University of Texas<br />

Health <strong>Science</strong> Center<br />

at San Antonio<br />

Gerald Carlson<br />

University of Missouri<br />

Steven Carr<br />

SmithKline Beecham<br />

Pharmaceutical<br />

Dan Davison<br />

Bristol-Myers Squibb<br />

EDITOR-IN-CHIEF<br />

Clayton W. Naeve<br />

St. Jude Children’s Research Hospital<br />

332 North Lauderdale Street<br />

Memphis, TN 38105-2794<br />

Tel: (901) 495-3861 Fax: (901) 495-2945<br />

email: clayton.naeve@stjude.org<br />

ASSOCIATE EDITORS<br />

EDITORIAL BOARD<br />

Karen De Jongh<br />

Zymogenetics<br />

Greg Grant<br />

Washington University<br />

at St. Louis<br />

George Grills<br />

Albert Einstein College<br />

of Medicine<br />

Susan Hardin<br />

University of Houston<br />

Preston Hensley<br />

Pfizer, Inc.<br />

Harold Hills<br />

Iowa State University<br />

Paul Morrison<br />

Dana-Farber Cancer<br />

Institute<br />

COPPERPLATE PRESS, INC.<br />

85 East Stewart Avenue<br />

Lansdowne, Pennsylvania 19050<br />

Tel: (610) 284-5257 Fax: (610) 284-5258<br />

email: mconnors@copperplatepress.com<br />

http://www.copperplatepress.com<br />

David Speicher<br />

The Wistar Institute<br />

3601 Spruce Street<br />

Philadelphia, PA 19104-4268<br />

Tel: (215) 898-3972 Fax: (215) 898-0664<br />

email: speicher@wistar.upenn.edu<br />

Ron Niece<br />

Research Resources and<br />

Technologies<br />

Margaret Robertson<br />

University of Utah<br />

Richard Simpson<br />

Ludwig Institute for<br />

Cancer Research<br />

Laurey Steinke<br />

University of Nebraska<br />

Medical Center<br />

John Stewart<br />

University of Colorado<br />

Dan Strydom<br />

BioNebraska, Inc.<br />

John R. Yates<br />

University of Washington<br />

ASSOCIATION OF BIOMOLECULAR RESOURCE FACILITIES


JBT<br />

JOURNAL OF BIOMOLECULAR TECHNIQUES<br />

VOLUME 11, ISSUE 4, DECEMBER 2000<br />

METHODS & REVIEWS<br />

CONTENTS<br />

Implementation of Automation in a Small-Scale DNA Sequencing Core Facility 151<br />

Jennifer L. Ware, Laurie Moran, Ching-Lun Lin, and Barton Slatko<br />

RESEARCH GROUP REPORTS<br />

Strategies for the Synthesis of Labeled Peptides 155<br />

Lisa Bibbs, Nicholas P. Ambulos, Steven A. Kates, Ashok Khatri,<br />

Katalin F. Medzihradszky, George Ösapay, and Susan T. Weintraub<br />

REPORT<br />

Research Equipment and Resource Requirements of NIH-Supported Investigators: 166<br />

An Assessment of Current Conditions and Recommendations for<br />

Future Funding and Programs<br />

Tamara R. Zemlo, Howard H. Garrison, David Lester, Mustafa Lokhandwala,<br />

Fred Naider, Stephen White, and David W. Speicher<br />

BOOK REVIEWS<br />

Amino Acid Analysis Protocols, edited by Catherine Cooper, 177<br />

Nicolle Packer, and Keith Williams<br />

Reviewed by Alan Mahrenholz<br />

— continued —<br />

The Journal of Biomolecular Techniques (ISSN 1524-0215) is published quarterly by The Association of Biomolecular<br />

Resource Facilities, 1201 Don Diego Avenue, Santa Fe, NM 87505. Copyright © 2000 by The Association of Biomolecular<br />

Resource Facilities. All rights reserved. Periodicals postage paid at Bethesda, Maryland, and at additional mailing<br />

offices.<br />

Address for subscription information, orders, or changes of address: The Association of Biomolecular Resource<br />

Facilities, 1201 Don Diego Avenue, Santa Fe, NM 87505; Tel: (505) 983-8102; Fax: (505) 989-1073.<br />

Annual subscription rates: Members of the Association of Biomolecular Resource Facilities receive a subscription as<br />

part of their membership fee. Other subscribers are charged as follows. Within the United States: $75.00 individual,<br />

$100.00 institution. (All other countries add $21.00 for air freight shipping—delivery within 7 to 21 days, worldwide.)<br />

International subscriptions must be prepaid. Please add $4.00 to all rates above for handling. Single copies, when available,<br />

may be ordered from the publisher. Single copies: $25.00. Prices subject to change without notice. Copies will be<br />

replaced without charge if the publisher receives a request within 90 days of the mailing date, both U.S. and worldwide.<br />

POSTMASTER: Send address changes to The Association of Biomolecular Resource Facilities, 1201 Don Diego Avenue,<br />

Santa Fe, NM 87505.


CONTENTS<br />

NEWS & EVENTS<br />

Message From the Executive Board 179<br />

<strong>AB</strong>RF/ASBMB Joint Symposium 180<br />

ARTICLE WATCH<br />

Amino Acid Composition and Sequence Analysis 181<br />

Carbohydrates and Glycoproteins 181<br />

Phospholipids 181<br />

Nucleic Acids 182<br />

Mass Spectrometry 182<br />

Proteins—Purification and Characterization 183<br />

Microarrays 184<br />

Bioinformatics 184<br />

UPCOMING EVENTS<br />

Dates to Remember 185<br />

<strong>AB</strong>RF 2001: THE NEW BIOLOGY: TECHNOLOGIES FOR<br />

RESOLVING MACROMOLECULAR COMMUNICATIONS<br />

Abstracts 188<br />

Author Index 233<br />

INDEX TO VOLUME 11, 2000<br />

Author Index 237<br />

Subject Index 237<br />

The Association of Biomolecular Resource Facilities cannot be held responsible for errors or for any consequences arising<br />

from the use of the information contained in this journal. The appearance of advertising in this journal does not<br />

constitute an endorsement or approval by The Association of Biomolecular Resource Facilities for the quality or value<br />

of the product advertised or the claims made for it by its manufacturer.<br />

PERMISSION TO PHOTOCOPY ARTICLES: This publication is protected by copyright. Permission to photocopy must<br />

be secured in writing from Clayton Naeve, Editor-in-Chief, St. Jude Children’s Research Hospital, 332 North Lauderdale<br />

Street, Memphis, TN 38105-2794; Tel: (901) 495-3861; Fax: (901) 495-2945.


CORPORATE SPONSORSHIP<br />

<strong>AB</strong>RF corporate sponsors provide valuable financial support to the organization; however, they do not in any way<br />

influence the selection of articles for or the content of the Journal. All articles published in the Journal undergo a<br />

peer-review process to determine merit and accuracy before being accepted for publication.<br />

<strong>AB</strong>RF CORPORATE<br />

BENEFACTORS<br />

Applied Biosystems<br />

Bruker Daltonics, Inc.<br />

Genomic Solutions, Inc.<br />

Micromass<br />

Millipore Corporation<br />

ThermoQuest Corporation<br />

Waters Corporation<br />

<strong>AB</strong>RF CORPORATE PATRON<br />

SciQuest.com<br />

Corporations interested in supporting <strong>AB</strong>RF should contact Laurey Steinke, Protein Structure Core Facility, UNMC,<br />

Omaha, NE 68198-4525; Tel: (402) 559-6647; Fax: (402) 559-6650; email: lsteinke@molbio.unmc.edu<br />

MISSION STATEMENT<br />

<strong>FR</strong>OM THE BY-LAWS OF THE ASSOCIATION OF<br />

BIOMOLECULAR RESOURCE FACILITIES<br />

The Association of Biomolecular Resource<br />

Facilities has been organized for the following<br />

purposes:<br />

• To promote and support resource facilities,<br />

research laboratories, and individual<br />

researchers regarding operation,<br />

research, and development in the areas<br />

of methods, techniques, and instrumentation<br />

relevant to the analysis and synthesis<br />

of biomolecules.<br />

• To provide mechanisms for the selfevaluation<br />

and improvement of procedural<br />

and operational accuracy, precision,<br />

and efficiency in resource facilities<br />

and research laboratories.<br />

• To provide a mechanism for the education<br />

of resource facility and research<br />

laboratory staff, users, administrators,<br />

and interested members of the scientific<br />

community.<br />

<strong>AB</strong>RF CORPORATE SPONSORS<br />

Affymetrix, Inc.<br />

Agilent Technologies<br />

Amersham Pharmacia Biotech, Inc.<br />

AnaSpec, Inc.<br />

Beckman Coulter, Inc.<br />

Biacore, Inc.<br />

Bio-Rad Laboratories, Inc.<br />

BioWhittaker Molecular Applications<br />

Edge BioSystems<br />

EM <strong>Science</strong><br />

GeneMachines<br />

Genentech, Inc.<br />

Geospiza, Inc.<br />

Glen Research Corporation<br />

<strong>AB</strong>RF ELECTRONIC COMMUNICATIONS<br />

EMAIL AND WEB ADDRESSES FOR CONTACTING<br />

<strong>AB</strong>RF AND ACCESSING <strong>AB</strong>RF INFORMATION<br />

ELECTRONIC DISCUSSION GROUP<br />

abrf-request@aecom.yu.edu (Subscription)<br />

abrf@aecom.yu.edu (Correspondence)<br />

ombudsmn@aecom.yu.edu (Ombudsman)<br />

ELECTRONIC DISCUSSION GROUP<br />

ARCHIVES<br />

http://www.abrf.org/archives<br />

<strong>AB</strong>RF HOME PAGE<br />

http://www.abrf.org<br />

Hitachi Instruments, Inc.<br />

LC Packings, Inc.<br />

Michrom BioResources, Inc.<br />

Prolinx, Inc.<br />

Promega Corporation<br />

QIAGEN<br />

Rainin Instrument Company<br />

Sigma-Aldrich<br />

SynPep Corporation<br />

Transgenomic, Inc.<br />

Vydac/The Separations Group<br />

Wyatt Technology Corporation<br />

ZymoGenetics, Inc.<br />

JOURNAL OF BIOMOLECULAR<br />

TECHNIQUES<br />

http://www.abrf.org/JBT/JBT.html


ASSOCIATION OF BIOMOLECULAR RESOURCE FACILITIES<br />

RESEARCH GROUP MEMBERS<br />

Amino Acid Analysis<br />

Peter Hunziker (chair) David Chin Len Packman (EB liaison)<br />

Michail Alterman Reed Harris Karen West<br />

Carbohydrate Analysis<br />

Adriana Manzi (chair) Roberta Merkle Monica Palcic<br />

DNA Sequence<br />

Dina Leviten (chair) John Hawes Mark Lively (EB liaison)<br />

Duane Bartley (ad hoc) Tim Hunter Theodore Thannhauser<br />

George Grills (ad hoc) Emily Jackson-Machelski James Van Ee (ad hoc)<br />

Lawrence Hall Kevin Lee Knudtson<br />

Fragment Analysis<br />

Doug Bintzler (chair) Duane Bartley Caprice Rosato<br />

Pamela Scott Adams Laura Kasch Laurey Steinke (EB liaison)<br />

Linda Wood Ballard Lynn Petukhova Catherine E. Terrell<br />

Yongde Bao<br />

Microarray<br />

Chandi Griffin (co-chair) Jeffrey Delrow Aldo Massimi<br />

George Grills (co-chair) Kevin Knudtson Laurey Steinke (EB liaison)<br />

Yongde Bao (ad hoc) Kathryn Lilley James Van Ee (ad hoc)<br />

Roger Bumgarner<br />

Molecular Interactions<br />

Preston Hensley (chair) Ed Eisenstein Fred Schwarz<br />

Karen De Jongh (EB liaison) David Myszka Steven Swanson<br />

Mike Doyle<br />

Nucleic Acids<br />

Martha Gunthorpe (chair) Kathryn Lilley Theodore Thannhauser<br />

Scottie Adams (ad hoc) Mark Lively (EB liaison) (ad hoc)<br />

Jay Fox Kathy Mills Anthony Yeung<br />

Brian Holloway Stephen Scaringe<br />

Peptide Synthesis<br />

Nicolas Ambulos (chair) George Osapay Arpad Somogyi<br />

Lynda Bonewald (EB liaison) Henriette Remmer George Tsaprailis<br />

Steve Kates<br />

Protein Analysis<br />

Terry Lee (co-chair) David Arnott Rich Johnson<br />

Linda Siconolfi-Baez John Crabb Len Packman (EB liaison)<br />

(co-chair) Bryan Dunbar<br />

Quality and Compliance<br />

Elizabeth Fowler (chair) Michael Cohrs Ronald Niece (EB liaison)<br />

Ruth Angeletti (ad hoc) John Dougherty Nadine Ritter<br />

Eleanor Canova-Davis Timothy Hayes Alan Smith<br />

COMMITTEE MEMBERS<br />

Corporate Relations<br />

Barbara Merrill (chair) Elizabeth Fowler Len Packman<br />

Karen De Jongh (EB liaison) Preston Hensley Ray Paxton<br />

Joe Fernandez<br />

Membership<br />

John Crabb (chair) Marc Lemaitre Daniel J. Strydom<br />

Lowell Ericsson Ronald Niece (EB liaison) Satya Yadov<br />

Peter Hunziker<br />

Nominations<br />

Ruth Angeletti (chair) Lynda Bonewald (EB liaison) Paul Morrison<br />

<strong>Science</strong> Policy<br />

Ruth Angeletti (chair) Lynda Bonewald (ad hoc) David Speicher<br />

Thomas Anderson A. L. Burlingame (EB liaison)<br />

Publications<br />

Ralph A. Bradshaw (chair) Clayton Naeve (ad hoc) Al Smith<br />

Lynda Bonewald (EB liaison) Clive Slaughter (ad hoc) David Speicher (ad hoc)<br />

Ben Dunn<br />

Survey<br />

Katheryn Resing (chair) Charles Nicolet John Stults (EB liaison)<br />

Janice Bleibaum Gautam Sarath Satya Yadav<br />

Debra McMillen (ad hoc)<br />

<strong>AB</strong>RF Award<br />

Greg Grant (chair) Lynda Bonewald (EB liaison)<br />

Web Page Redesign<br />

Theodore Thannhauser Paul Morrison Laurey Steinke<br />

(chair) Len Packman James Van Ee<br />

Finance and Investments<br />

Karen De Jongh (chair)<br />

EXECUTIVE BOARD<br />

RF <strong>AB</strong><br />

Research•Technology<br />

Communication•Education<br />

PRESIDENT<br />

Lynda F. Bonewald<br />

SA-Med-Endocrine Core Facility<br />

University of Texas Health <strong>Science</strong> Center<br />

7703 Floyd Curl Drive<br />

San Antonio, TX 78284<br />

Tel: (210) 567-6666 Fax: (210) 567-6693<br />

email: bonewald@uthscsa.edu<br />

A. L. Burlingame<br />

Department of Pharmaceutical Chemistry<br />

University of California San Francisco<br />

San Francisco, CA 94143-0446<br />

Tel: (415) 476-5641 Fax: (415) 476-0688<br />

email: alb@itsa.ucsf.edu<br />

TREASURER<br />

Karen De Jongh<br />

Zymogenetics, Inc.<br />

1201 Eastlake Avenue East<br />

Seattle, WA 98102<br />

Tel: (206) 442-6659 Fax: (206) 442-6608<br />

email: dejonghk@zgi.com<br />

Mark O. Lively<br />

Department of Biochemistry<br />

Bowman Gray School of Medicine<br />

Medical Center Boulevard<br />

Winston-Salem, NC 27157-3001<br />

Tel: (336) 716-2969 Fax: (336) 716-7200<br />

email: mlively@wfubmc.edu<br />

Ronald Niece<br />

Research Resources & Technologies<br />

58 Whitman Court<br />

Irvine, CA 92612<br />

Tel: (949) 856-0553 Fax: (949) 856-0564<br />

email: RLNiece@aol.com<br />

Len C. Packman<br />

Department of Biochemistry<br />

Cambridge University<br />

80 Tennis Court Road<br />

Cambridge, UK<br />

Tel: 44-122-333-3639 Fax: 44-122-376-6002<br />

email: lcp2@mole.bio.cam.ac.uk<br />

Laurey Steinke<br />

University of Nebraska Medical Center<br />

Protein Structure Core Facility<br />

600 South 42nd Street, Box 984525<br />

Omaha, NE 68798-4525<br />

Tel: (402) 559-6647 Fax: (402) 559-6650<br />

email: lsteinke@molbio.unmc.edu<br />

John T. Stults<br />

1 DNA Way, #63<br />

Genentech, Inc.<br />

460 Point San Bruno Boulevard<br />

South San Francisco, CA 94080<br />

Tel: (650) 225-1203 Fax: (650) 225-5945<br />

email: jts@gene.com<br />

<strong>AB</strong>RF BUSINESS OFFICE<br />

1201 Don Diego Avenue<br />

Santa Fe, NM 87505<br />

Tel: (505) 983-8102 Fax: (505) 989-1073<br />

email: abrf@abrf.org


Implementation of Automation<br />

in a Small-Scale DNA<br />

Sequencing Core Facility<br />

Jennifer L. Ware, Laurie Moran,<br />

Ching-Lun Lin, and Barton Slatko<br />

New England Biolabs, Inc., Beverly, MA<br />

METHODS<br />

REVIEWS<br />

New England Biolabs (NEB) sequencing core facility provides<br />

automated sequencing services to support various<br />

company-wide projects in house, but on a very small scale of<br />

about 1000 to1500 reactions per month. A procedure has<br />

been implemented at the NEB core sequencing facility to<br />

integrate simplified methods and robotics to provide a more<br />

efficient small-scale process. This has been done using a<br />

Beckman Biomek 2000 robot combined with an MJ DNA<br />

Engine, 96-well plate cycler (PTC-200), <strong>AB</strong> 373 and 377<br />

sequencers, BMA Singel gels, and several other materials that<br />

help reduce the time required for otherwise lengthy procedures<br />

in a cost-efficient manner. Protocols have also been<br />

developed for efficient sequencing of a variety of templates<br />

submitted to the NEB core facility. (J Biomol Tech 2000;11:<br />

151–154)<br />

KEY WORDS: automation, robotics, sequencing template<br />

cleanup, small-scale DNA sequencing.<br />

ADDRESS CORRESPONDENCE AND REPRINT REQUESTS TO: Jennifer L.<br />

Ware, New England Biolabs, Inc., 32 Tozer Road, Beverly, MA<br />

01915 (email: ware@neb.com).<br />

&<br />

Journal of Biomolecular Techniques<br />

11:151–154 © 2000 <strong>AB</strong>RF<br />

RF <strong>AB</strong><br />

Genome sequencing and analysis technology<br />

has advanced in a manner that allows companies<br />

to generate enormous amounts of data<br />

per day, via automated processes, in an effort to complete<br />

genome projects and use that information as a<br />

source of possible disease treatment and control.<br />

Unfortunately, smaller core facilities, which have less<br />

of a need to produce such high-throughput data, are<br />

not the primary targets of application of these<br />

advanced technologies and are in need of simplified<br />

methods. The core sequencing laboratory at New<br />

England Biolabs, Inc. (NEB, Beverly, MA) has combined<br />

recent advances in sequencing technology to<br />

efficiently simplify the process from beginning to end.<br />

The sequencing core facility provides additional measures<br />

of quality control of appropriate DNA products<br />

for sale at NEB, such as sequencing primers and other<br />

oligonucleotides, cloning vectors by sequencing reaction<br />

assay, and vector and phage display products to<br />

ensure correct sequence. For our in-house research<br />

program, sequences are required from vector and<br />

overexpression constructs of clones, cDNA, polymerase<br />

chain reaction (PCR), phage display, and other<br />

sequencing templates, including our involvement in<br />

the Filarial Genome Project (http://www.neb.com/<br />

fgn/filgen1.html). Among those templates submitted<br />

for analysis are plasmids, M13 derivatives, � phage,<br />

PCR products, cosmids, and bacterial artificial chromosomes<br />

(BACs), each of which requires a different<br />

amount of template and/or primer per reaction and<br />

sometimes different sequencing reaction conditions as<br />

well.<br />

ELECTRONIC SUBMISSION<br />

To make this process efficient from beginning to end,<br />

researchers start by submitting an electronic form to<br />

the core facility in which key information necessary to<br />

perform the sequencing is provided. The form is accessible<br />

on the DNA Sequencing Core Facility intranet<br />

JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000 151


J. L. WARE ET AL.<br />

site (on a Unix server) through the user’s desktop<br />

Macintosh or PC. The intranet site provides the home<br />

base for submission and receipt of sequence information<br />

to and from the core facility. By selecting the<br />

“submit your template to DNASEQ lab” icon, the<br />

researcher can fill out an html form in which they<br />

describe the project, identify the type of template<br />

being submitted, and list any special instructions concerning<br />

the reaction (eg, “please include DMSO; high<br />

G�C content”). It is necessary that the instructions to<br />

the sequencing core be precise. We require both the<br />

templates and primers be submitted at the proper<br />

concentrations for the reaction conditions. The core<br />

facility maintains a stock of common “standard”<br />

primers such as T3, T7, SP6 promoter primers, and<br />

pUC-based forward and reverse primers, and the<br />

researcher need not provide them. The form also asks<br />

the submitter about the method of purification of the<br />

template to enable the core facility to track statistics of<br />

success and failure with different methods and to<br />

identify any failure correlations. In addition, the form<br />

requires some substantiation of DNA concentration to<br />

ensure that the submitter has indeed verified template<br />

concentration. One major class of reaction failures<br />

is the result of too much or too little DNA. A second<br />

class is suboptimal (or no) sequence data, usually<br />

attributable to template impurity or primer design<br />

issues. We consider a failure any sequence that does<br />

not give a reasonable amount of sequence (eg, 500–<br />

600 “good” nucleotide base calls for double-stranded<br />

plasmid DNA templates).<br />

After the form is submitted to and received by the<br />

core sequencing facility, a message to the sender<br />

thanks them for their submission. If the form is not<br />

completely filled out, it will not be submitted, and an<br />

error message appears to the submitter identifying<br />

the problem and asking them to “try again.”<br />

We chose a Unix-based server as the site for submission<br />

and return of sequence files because of its<br />

faster speed and higher data-handling capabilities,<br />

especially when downloading sequences to the end<br />

user. We also run CAP (Columbia Appletalk Package<br />

for Unix) on our Web server so the sequence data<br />

from the sequencers can be uploaded easily. The submission–data<br />

porting script was written at NEB and<br />

provides a rapid and simple way to handle data electronically.<br />

After the submission form is sent, the researcher<br />

physically submits the template and primer in a small<br />

plastic bag with their name on it by placing it in a<br />

box marked “TEMPLATES IN” in a small freezer in the<br />

core facility. Template and oligonucleotide are submitted<br />

in pairs—for every template submitted there<br />

must be a companion primer tube, and vice versa.<br />

The sequencing reactions are then performed accord-<br />

152 JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000<br />

ing to the instructions on the submission sheet using<br />

the corresponding primers and templates submitted<br />

to the core facility. After the sequence data are<br />

obtained, and retracked and recalled if necessary, a<br />

copy of the raw data is directly ported through the<br />

server back to a folder automatically created at the<br />

DNA Sequencing Core Facility intranet site. Users<br />

simultaneously receive an email message, delivered<br />

to the email address they submitted on the form,<br />

stating, “Your DNA sequence data is ready.” By<br />

returning to the intranet site and choosing the “pick<br />

up your sequence” icon, the researcher can go<br />

directly to the server to download the sequences<br />

to his or her own computer for analysis. Sequenced<br />

templates and primers can then be picked up by the<br />

user from the core facility freezer “TEMPLATES OUT”<br />

box.<br />

The NEB core sequencing facility uses the Applied<br />

Biosystems (Foster City, CA) <strong>AB</strong> 373 and 377<br />

automated sequencers to process approximately 84 to<br />

96 dye terminator samples per day. Dye terminator<br />

sequencing chemistry is used to accommodate the<br />

variety of template types and vector constructs being<br />

sequenced and to maximize the number of primers<br />

that can be used.<br />

ROBOTICS AND REACTION<br />

PREPARATION<br />

The Biomek 2000 Robot (Beckman Instruments, Palo<br />

Alto, CA) is used to assemble the components of sequencing<br />

reactions. Using adaptors for 1.5-mL centrifuge<br />

tubes, the robot can build any number of desired<br />

reactions so that in-house customers can submit<br />

in standard-sized tubes. The reaction components are<br />

transferred directly into a 96-well tray on the MJ cycler<br />

with a Power Bonnet heated lid (MJ Research, Watertown,<br />

MA) attached to the robot and thermally cycled<br />

immediately on addition of all reagents, as specified<br />

by a preset program.<br />

The robot has limitations that affect volume per<br />

tube, namely, a lack of ability to withdraw small<br />

volumes of reagent. It is necessary to provide at least<br />

10 �L of reagent if 5 �L is to be withdrawn by the<br />

robot. Similarly, for 1 �L to be withdrawn, 5 �L must<br />

be present. For our sequencing protocol, doublestranded<br />

and single-stranded DNA templates must be<br />

at 100 ng/�L (5 �L is used, thus 10 �L must be submitted),<br />

and PCR products must be at 8 ng/�L (5 �L<br />

is used, thus 10 �L must be submitted). We require<br />

oligonucleotide concentrations to be 3.2 pmol/�L for<br />

double-stranded DNA, and 5 �L must be provided per<br />

primer tube although only 1 �L is added per reaction.<br />

For single-stranded DNA, primers are submitted at


0.8 pmol/�L with 5 �L per primer tube; again, only<br />

1 �L is added per reaction.<br />

To use the robot to assemble the components of<br />

these reactions, a simple program was composed to<br />

transfer samples between the robot benchtop platform<br />

and the 96-well plate on the connected MJ cycler. A<br />

script is available from Beckman, as well as from MJ<br />

Research, that allows the cycler to close the heated lid<br />

and start cycling on reaction setup completion.<br />

We have found that reactions containing one half<br />

of the manufacturer’s prescribed quantity of Amplitaq<br />

DNA Polymerase FS premix (<strong>AB</strong> PRISM Dye Terminator<br />

Cycle Sequencing Ready Reaction Kit using 4 �L<br />

premix per reaction with the <strong>AB</strong> recommended<br />

amount of DNA template and primer), yielding a total<br />

volume of 10 �L, works better and is more costefficient<br />

than the standard <strong>AB</strong> protocol that includes<br />

8 �L premix in each reaction, yielding a total volume<br />

of 20 �L. 1<br />

� PHAGE/COSMID/BAC<br />

SEQUENCING METHODS<br />

Protocols have been designed and optimized for<br />

sequencing reactions other than standard doublestranded<br />

or single-stranded DNA templates. These<br />

sequences are done manually and cycled on an MWG<br />

cycler (Biotech, High Point, NC), but can be done on<br />

the robot with the appropriate programming.<br />

With Amplitaq FS terminator chemistry, BAC sequencing<br />

requires a large-scale reaction for maximum<br />

output of at least 500 bases per reaction and minimum<br />

background. Sixteen microliters of premix is combined<br />

with 1 �g of DNA template and 50 pmol primer<br />

and dH2O to give a total volume of 40 �L. 2<br />

Cosmid and � phage reactions are performed<br />

using 500 ng DNA (the standard amount of DNA used<br />

in double-stranded and single-stranded reactions), but<br />

with 12.8 pmol of primer (approximately 4 times the<br />

standard 3.2 pmol amount) and 8 �L premix to provide<br />

a total reaction volume of 20 �L.<br />

For reaction product purification, Centrisep spin<br />

columns (Princeton Separations, Adelphia, NJ) are<br />

used on an individual basis for the reactions performed<br />

manually, as described in the product manual.<br />

A 96-well system (Edge Biosystems, Gaithersburg,<br />

MD) is used for product purification of the robotprepared<br />

automated sequencing reactions performed<br />

in the 96-well cycler. Product purification is also done<br />

with the assistance of the robot. A simple program<br />

was written to transfer the cycled sequencing reactions<br />

directly onto the center of each well of a 96-well<br />

separation plate with gel bed of hydrated sephadex<br />

mixture. After all reaction mixtures are transferred to<br />

AUTOMATION IN SMALL-SCALE DNA SEQUENCING FACILITY<br />

this separation plate, the plate is centrifuged at 800<br />

rpm in a Beckman GS-15 microtiter tray centrifuge<br />

with a collection tray underneath it to collect the purified<br />

sequencing sample. This tray is then placed in a<br />

Jouan R6-1010 vacuum centrifuge (Jouan, Winchester,<br />

VA) to dry the samples. The dried samples, as well as<br />

those dried samples prepared via Centrisep columns,<br />

are then resuspended in 2-�L Accutrac dye (3 �L for<br />

the <strong>AB</strong> 373 sequencer; Commonwealth BioTechnologies,<br />

Richmond, VA), which provides a unique dye<br />

marker for each lane on the gel image regardless of<br />

whether or not the reaction has failed. This reduces<br />

software errors in lane tracking. After the samples are<br />

resuspended in dye, they are vortexed and centrifuged<br />

at maximum speed for 30 seconds and then<br />

heat denatured at 90�C for 2 minutes. The samples are<br />

then loaded onto the sequencers in a staggered manner<br />

in which the odd samples are loaded first and<br />

subjected to electrophoreses for 10 minutes, and then<br />

the even samples are loaded.<br />

GELS<br />

Biowhitaker prepackaged Singel gels (Long Ranger<br />

Singel packs for <strong>AB</strong> 373 and 377; BMA, Rockland, ME)<br />

are used as a simple, efficient alternative to making<br />

our own gel solutions. It takes 12 minutes to prepare<br />

the gels for pouring. Using mild alkanox detergent<br />

and isopropanol to clean the glass plates, the gel solution<br />

is placed in a side-arm flask for pouring onto the<br />

plates using an Owl Scientific Otter sequencing gel<br />

caster (Owl Separation Systems, Portsmouth, NH).<br />

A period of 2 to 2.5 hours is necessary to allow the<br />

gels to completely polymerize; other steps in the<br />

process are performed during this period.<br />

DATA ANALYSIS<br />

The core facility currently processes 1000 to 1500<br />

sequence reactions per month, with about 60 facility<br />

users submitting templates for sequencing. Data from<br />

each sequence run is immediately backed up on a<br />

gigabyte tape drive and copied onto a CD for permanent<br />

storage. In addition, a summary of the sequencing<br />

results is transferred to tape for short-term storage.<br />

Gel image files are kept for 3 days to enable retracking<br />

or for use of alternative base-calling programs, if<br />

necessary.<br />

When novice users first submit templates for<br />

sequencing, they are given a written set of instructions<br />

concerning template preparation and quantitation.<br />

They are also instructed in how to configure their<br />

Netscape Navigator (Netscape Communications Corp.,<br />

Mountain View, CA) to download sequences from the<br />

JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000 153


J. L. WARE ET AL.<br />

intranet site. Netscape Navigator must be configured<br />

using a sequence trace editor to view the sequence<br />

chromatogram files generated by the <strong>AB</strong> sequencers.<br />

They are also shown how to use the analysis programs.<br />

This information is also present on the intranet<br />

site, along with information on sequencing chemistry<br />

and helpful hints on reading sequences. Numerous<br />

copies of computer programs for analysis are mounted<br />

on individual Macintosh or Windows-based PC computers<br />

with access by limited user-number licenses. In<br />

addition, each user is given a copy of EditView for<br />

Macintosh users (available by FTP from the <strong>AB</strong> Web<br />

site) or Chromas for PC users (Techlysium, PTY LTD,<br />

Helensvale, Australia) to do simple editing and text<br />

file saving. The core facility provides support and<br />

training for certain computer programs for assembling<br />

sequencing projects, such as Autoassembler (<strong>AB</strong>),<br />

Sequencher (Gene Codes, Ann Arbor, MI), and GCG<br />

Programs (Pharmacopeia, Princeton, NJ). Other Macand<br />

PC-based programs are also in common use.<br />

CONCLUSIONS<br />

Despite the focus on high-throughput automated<br />

sequencing, many laboratories, such as NEB, have a<br />

154 JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000<br />

medium-throughput level. From the submission/data<br />

porting script to the template cleanup, we have<br />

designed simplified methods to make each step more<br />

efficient and less time consuming for a small-scale<br />

core facility. Using this system has enabled us to efficiently<br />

perform DNA sequencing service, generally<br />

achieving a 2-day turnaround between submission<br />

and return of sequence information.<br />

ACKNOWLEDGMENTS<br />

This work was supported by New England Biolabs, Inc.<br />

(NEB). We sincerely thank the computer group at NEB for<br />

their essential aid in the core facility. All programs mentioned<br />

in this article can be obtained from Beckman, MJ<br />

Research, or NEB.<br />

REFERENCES<br />

1. <strong>AB</strong>I PRISM Dye Terminator Cycle Sequencing Ready<br />

Reaction Kit with Amplitaq DNA Polymerase FS Protocol,<br />

revision A. Foster City, CA: Applied Biosystems,<br />

August 1995.<br />

2. Boysen C, Simon M, Hood L. Fluorescence-based sequencing<br />

directly from bacterial and P1-derived artificial<br />

chromosomes. BioTechniques 1997;23:978–982.


RESEARCH GROUP<br />

REPORTS<br />

Strategies for the Synthesis<br />

of Labeled Peptides<br />

Lisa Bibbs, a Nicholas P. Ambulos, b<br />

Steven A. Kates, c Ashok Khatri, d<br />

Katalin F. Medzihradszky, e<br />

George Ösapay, f and<br />

Susan T. Weintraub g<br />

a The Scripps Research Institute, La Jolla, CA;<br />

b University of Maryland at Baltimore,<br />

Baltimore, MD; c PerSeptive Biosystems, Inc.,<br />

Farmingham, MA; d Massachusetts General<br />

Hospital, Boston, MA; e University of California,<br />

San Francisco, CA; f University of California,<br />

Irvine, CA; g University of Texas Health <strong>Science</strong><br />

Center at San Antonio, San Antonio, TX<br />

Labeled peptides synthesized by core facilities are frequently<br />

used by researchers for following trafficking of a peptide, for<br />

binding studies, to determine substrate specificity, and for<br />

receptor cross-linking studies.The membership of the Association<br />

of Biomolecular Resource Facilities was asked to participate<br />

in a study focusing on synthesis of a biotin-labeled<br />

peptide, and it was suggested that a new strategy, using Rink<br />

amide 4-methylbenzhydrylamine resin coupled with Fmoc-<br />

Lys(Dde)-OH, be used.This strategy can be used for addition<br />

of a variety of labels other than biotin and should prove useful<br />

to core facilities. Comparison of the new strategy to<br />

other strategies was performed. Biotin labeling has long been<br />

ADDRESS CORRESPONDENCE AND REPRINT REQUESTS TO: Lisa Bibbs,<br />

The Scripps Research Institute, Core Facility, SP-3, La Jolla, CA<br />

92037 (email: bibbs@scripps.edu).<br />

assumed to be routine and specific. Despite the assumed<br />

routine nature of synthesizing biotinylated peptides, 9 of the<br />

34 samples submitted did not contain any of the correct<br />

product. Although synthesis using Fmoc-Lys(Dde)-OH plus<br />

biotin generally gave the highest yields, other approaches<br />

also yielded a high percentage of the correct product.Therefore,<br />

the various strategies are generally comparable. The<br />

major advantage of this new approach is that other labels<br />

such as fluorescein, dansyl groups, methyl coumarin, and<br />

potentially fluorophores and quenchers used for fluorescence<br />

resonance energy transfer (<strong>FR</strong>ET) can be directly<br />

incorporated into peptides. (J Biomol Tech 2000;11:155–165)<br />

KEY WORDS: biotin, labeled peptides, synthesis.<br />

Journal of Biomolecular Techniques<br />

11:155–165 © 2000 <strong>AB</strong>RF<br />

RF <strong>AB</strong><br />

Two approaches can be used to generate labeled<br />

peptides: the peptide can be synthesized<br />

using labeled amino acids, or the label can be<br />

added after peptide synthesis has been completed.<br />

When a peptide is labeled after synthesis, there occasionally<br />

are problems with the location of the label,<br />

depending on the composition of the peptide. The<br />

use of labeled amino acids during synthesis usually<br />

insures the correct positioning of the label.<br />

The <strong>AB</strong>RF Peptide Synthesis Research Group<br />

(PSRG) of the Association of Biomolecular Resource<br />

Facilities (<strong>AB</strong>RF) conducts annual studies to help<br />

member laboratories assess their peptide synthesis<br />

capabilities. 1 Concurrently, through careful design and<br />

synthesis of the test sequence, these studies also serve<br />

as an avenue to introduce new techniques to member<br />

laboratories. This study focused on the synthesis of<br />

side-chain–labeled peptides—specifically, a peptide<br />

with a C-terminal biotin-labeled lysine. Participating<br />

laboratories were asked to construct the following<br />

peptide:<br />

H-Ala-Glu-Lys-Gly-Lys-Leu-Arg-Phe-Lys(biotin)-NH 2<br />

Although use of Fmoc-Lys(biotin)-OH was an<br />

obvious choice for direct assembly of this sequence,<br />

JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000 155


L. BIBBS ET AL.<br />

one goal of the study was to acquaint member laboratories<br />

with a flexible strategy that could be used for<br />

introduction of a variety of different labels other than<br />

biotin. To make this endeavor especially valuable for<br />

core laboratories, it was suggested that the new<br />

approach, based on utilization of a side-chain protected<br />

lysine residue orthogonal to Fmoc/t-Bu that<br />

employs Rink amide MBHA resin coupled with Fmoc-<br />

Lys(Dde)-OH be used. 2 In this approach, first the<br />

peptide is synthesized, then the label is added for<br />

coupling to the Lys, followed by cleavage and deprotection.<br />

Participating laboratories were asked to submit<br />

the requested peptide without purification. The<br />

PSRG members used amino acid analysis, capillary<br />

electrophoresis, reverse-phase high-performance liquid<br />

chromatography (HPLC), matrix-assisted laser desorption/ionization<br />

time-of-flight mass spectrometry<br />

(MALDI-TOF/MS), and electrospray ionization mass<br />

spectrometry (ESI/MS) to analyze all peptides submitted<br />

from participating laboratories in addition to a<br />

test peptide synthesized by the PSRG in preparation<br />

for the study. In this report, the results from analysis<br />

of the PSRG test peptide and submitted peptides are<br />

presented, and the method used by the PSRG for synthesis<br />

of the test peptide is described.<br />

METHODS<br />

Synthesis of the <strong>Reference</strong> Peptide<br />

For testing by the PSRG, H-Ala-Glu-Gly-Lys-Leu-Arg-<br />

Phe-Lys(biotin)-NH 2 was synthesized on an <strong>AB</strong> 431A<br />

synthesizer (Applied Biosystems, Foster City, CA).<br />

Rink amide MBHA resin was coupled with Fmoc-<br />

Lys(Dde)-OH using HBTU/HOBt/DIEA activation. 3 All<br />

remaining residues except the N-terminal Ala were<br />

incorporated as Fmoc-amino acids with the same activation<br />

chemistry, using Pbf for Arg, Boc for Lys, and<br />

OtBu for Glu as the side-chain protecting groups. The<br />

N-terminal Ala was incorporated as t-Boc-Ala. After<br />

completion of the synthesis, the resin was washed<br />

twice with N,N-dimethylformamide (DMF). The Dde<br />

side-chain protection was removed by treatment with<br />

2% hydrazine (Sigma, St. Louis, MO) in DMF (7 mL/0.1<br />

mmole of the protected peptide resin; 2 times, 5 minutes<br />

each). The resin was then washed 3 times with<br />

DMF followed by 2 washes with DMF:dimethyl sulfoxide<br />

(DMSO; 1:1, v/v). A 10-fold molar excess of<br />

biotin (Aldrich, Milwaukee, WI) was dissolved in 5 mL<br />

of DMF:DMSO (1:1). It was necessary to warm the<br />

mixture and vortex it for several minutes to dissolve<br />

the biotin completely. The biotin solution was then<br />

treated with 2.1 mL of 0.45-M HBTU/HOBt in DMF followed<br />

by 0.3 mL of diisopropylethylamine (DIEA).<br />

The activated biotin solution was added to the resin,<br />

156 JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000<br />

and the mixture was stirred overnight. The resin was<br />

washed with DMF:DMSO (1:1; 3 times) followed by<br />

dichloromethane:methanol (1:1; 2 times). After the<br />

resin was thoroughly dried, the peptide was cleaved<br />

and deprotected with Reagent-K.<br />

Amino Acid Analysis<br />

Crude peptide samples were hydrolyzed for 24 hours<br />

at 110�C using vapor-phase hydrolysis in 6-N HCl containing<br />

2% saturated phenol/water. The samples were<br />

then analyzed by Waters AccQ*Tag chemistry using a<br />

Waters Alliance HPLC system (Milford, MA) equipped<br />

with a fluorescence detector.<br />

High-Performance Liquid Chromatography<br />

Analyses were conducted on a Waters HPLC system<br />

consisting of two model 600 solvent delivery systems,<br />

a Wisp model 712 automatic injector, a model 490 programmable<br />

wavelength ultraviolet detector, and a DEC<br />

model 860 Networking Computer to control system<br />

operation and collect data. HPLC conditions were as<br />

follows: column, Delta Pak C18 (Waters), 3.9 � 150<br />

mm, 5 �m, 100 Å; buffer A, 0.1% trifluoroacetic acid<br />

(TFA) in water; buffer B, 0.1% TFA in acetonitrile; linear<br />

gradient, 5% B to 60% B in 45 minutes; flow rate,<br />

1.0 mL/minute; detector wavelengths, 220 and 279 nm.<br />

Capillary Electrophoresis<br />

Peptides were analyzed on a PACE-MDQ CE system<br />

(Beckman Instruments, Palo Alto, CA) using an aminecoated<br />

capillary and acetate buffer, pH 4.5. The capillary<br />

was preconditioned with an amine regenerator<br />

and a buffer rinse. Samples were applied by a 5-second<br />

pressure injection (2 psi) and separated at 30 kV<br />

for 10 minutes using reversed polarity.<br />

Electrospray Ionization Mass Spectrometry<br />

ESI/MS analyses were conducted on a ThermoFinnigan<br />

LCQ ion trap mass spectrometer (San Jose, CA).<br />

Samples were dissolved in 1 mL of water and diluted<br />

at least 100-fold in 70% aqueous acetonitrile/0.5%<br />

acetic acid. Peptide samples were introduced into the<br />

electrospray interface by flow injection at a rate of<br />

5 �L/minute. MS conditions were as follows: capillary<br />

temperature, 200�C; spray voltage, 5 kV; capillary voltage,<br />

30 V; tube lens offset, 18 V; sheath gas, 30 L/min.<br />

For MS n , the relative collision energy was 35%. Spectral<br />

averaging for 10 scans was used prior to data<br />

acquisition.


Matrix-Assisted Laser Desorption Ionization<br />

Time-of-Flight Mass Spectrometry<br />

One microliter of a sample solution of approximately<br />

10 to 50 pmole/�L was combined with 1 �L of 4hydroxy-alpha-cyano-cinnamic<br />

acid matrix solution<br />

(Hewlett-Packard, Palo Alto, CA), and the mixture<br />

was deposited on a stainless steel target (2 spots/sample).<br />

MALDI-TOF analyses were performed on either<br />

a Voyager Elite or a Voyager DE-STR mass spectrometer<br />

(Applied Biosystems) in reflectron mode with<br />

delayed extraction. A two-point external calibration<br />

based on angiotensin II (m/z 1046.54, MH � ) and<br />

adrenocorticotropic hormone (ACTH) 18 to 39 Clip<br />

peptide (m/z 2465.2, MH � ) was used for the molecular<br />

mass determinations. Post-source decay (PSD) experiments<br />

were performed on the Voyager DE-STR.<br />

Data were acquired in 10 consecutive frames, lowering<br />

the reflectron voltage by 25% at each step. Air was<br />

used as collision gas while focusing on the low mass<br />

region (m/z � 200). The data were smoothed to yield<br />

mostly average masses. ACTH-Clip peptide was used<br />

for PSD calibration.<br />

Sequence Analysis<br />

Selected peptides were sequenced by Edman degradation<br />

using an Applied Biosystems model 494 protein<br />

sequencer and an Applied Biosystems model<br />

120A HPLC analyzer fitted with a reverse-phase (RP)<br />

HPLC column.<br />

RESULTS AND DISCUSSION<br />

Analysis of the Test Peptide<br />

The integrity of the reference peptide was confirmed<br />

by amino acid analysis, capillary electrophoresis,<br />

Edman degradation, RP-HPLC, MALDI-TOF/MS, and<br />

STRATEGIES FOR SYNTHESIS OF L<strong>AB</strong>ELED PEPTIDES<br />

FIGURE 1<br />

Elution position of PTH-Lys(biotin) by<br />

Edman sequence analysis. The Lys(biotin)<br />

is well resolved from other amino acids<br />

and can easily be identified.<br />

ESI/MS. All analyses gave the anticipated results. As<br />

shown in Figure 1, PTH-Lys(biotin) was well resolved<br />

from the other amino acids during Edman degradation<br />

analysis.<br />

The ESI mass spectrum of the test peptide is<br />

shown in Figure 2. Three charge states of the peptide<br />

were readily detected: m/z 1173.7, [M�H] � , m/z<br />

587.5, [M�2H] 2� , and m/z 392.1, [M�3H] 3� . The tandem<br />

mass spectrometry (MS/MS) spectrum obtained<br />

by collision-induced dissociation of the 2� ion confirmed<br />

the expected sequence (Fig. 3).<br />

Analysis of Submitted Peptides<br />

Thirty-four peptides were submitted in crude form by<br />

participating laboratories. Eight different methods<br />

were used for the synthesis. The best crude material<br />

was found to be 96% pure by RP-HPLC. A summary<br />

of the analytical data for the submitted peptides is<br />

presented in Table 1. The following results were obtained:<br />

1. Nine of the peptides were constructed using a<br />

prelabeled Lys(biotin), yielding from 0% to 88%<br />

correct product by HPLC. In three of these peptides,<br />

the Lys(biotin) was not efficiently coupled.<br />

2. Eleven laboratories used Fmoc-Lys(Dde)-OH (as<br />

did the PSRG). Three of these submitted samples<br />

contained no correct product. One contained the<br />

Lys(biotin) but was missing the Arg. The other<br />

two samples contained two biotins. The attachment<br />

of the extra biotin was the result of the fact<br />

that Boc-Ala-OH was not employed as the N-terminal<br />

residue. These last two laboratories also<br />

used modified biotins.<br />

3. Nine laboratories used Fmoc-Lys(Mtt)-OH. Of<br />

these samples, one had no correct product and<br />

was missing the biotin, and another contained<br />

only low levels of the requested peptide. For this<br />

synthesis, the Mtt group must be selectively<br />

JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000 157


L. BIBBS ET AL.<br />

FIGURE 2<br />

FIGURE 3<br />

Electrospray ionization mass spectrum of the test peptide,AEGKLRFK(biotin).Three charge states of the peptide<br />

are readily detected, verifying a molecular mass of 1173 daltons.<br />

Tandem mass spectrum obtained by collision-induced dissociation of [M�2H] 2�(m/z 587.5) for the test peptide.The<br />

fragmentation pattern confirms the expected sequence.<br />

158 JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000


T<strong>AB</strong>LE 1<br />

Summary of Results from <strong>AB</strong>RF Peptide Synthesis Research Group Study<br />

STRATEGIES FOR SYNTHESIS OF L<strong>AB</strong>ELED PEPTIDES<br />

Percentage<br />

Mass Spectrometry Data<br />

Correct Product Major Product<br />

Other Lys<br />

RP- Correct Compo- Protec- Cleavage<br />

Samplea HPLC CE Mass Identity nents tion Biotin Chemistry Instrument Reagent<br />

1340A 88.9 85.9 Yes Aloc D-Biotin Fmoc/HBTU Rainin<br />

Symphony<br />

Modified K<br />

4761 12.5 31 Yes Multiple Aloc EZ-link<br />

NHS 9050<br />

Biotin<br />

Fmoc Milligen R<br />

0715 86.4 88 Yes � Lys Biotin Fmoc/PyBOP/ Shimadzu R<br />

(biotin) HOBt Pssm-8<br />

1231 82.2 81.7 Yes � Glu, Biotin Fmoc/HBTU/ Rainin Modified B<br />

� Ala,<br />

� Lys<br />

(biotin)<br />

HOBt Symphony<br />

1354 54.9 76.8 Yes � Lys Biotin Fmoc/HBTU/ <strong>AB</strong>I 431 Modified B<br />

(biotin) HOBt<br />

4612 28.5 58.9 Yes peptide Biotin Fmoc/HBTU/ <strong>AB</strong>I 431A B<br />

�16,<br />

� Lys<br />

(biotin)<br />

HOBt<br />

8105 0 0 No � Lys Glu-to- Biotin Fmoc/HBTU/ <strong>AB</strong>I 433A Modified K<br />

(biotin)<br />

and Gluto-Gln<br />

Gln HOBt<br />

8398A 73.8 80.1 Yes Biotin Fmoc/HATU Rainin<br />

Symphony<br />

B<br />

8583 11.7 12.7 No � Lys Biotin Fmoc/HBTU/ <strong>AB</strong>I 433A K<br />

(biotin) HOBt<br />

MPCF 76.6 83.8 Yes � Pmc Biotin Fmoc/HBTU Rainin<br />

Symphony<br />

Modified B<br />

8398B 59.3 81.7 Yes (Biotin � Extra Biotin Fmoc/HATU Rainin B<br />

caproate) Lys(biotin caproate Symphony<br />

caproate) resin<br />

2958 50.4 47 Yes � 91 Amu, Boc-Lys D-Biotin Boc/HBTU/ <strong>AB</strong> 430 5 mL HF/<br />

possible (2ClZ) HOBt 500 �L<br />

bzl on Glu Anisole<br />

0690 61.2 84 Yes (Biotin � Pmc Dde (�)Biotin Fmoc/HBTU/ <strong>AB</strong> 433A Reagent-K<br />

caproate) HOBt<br />

2311 0 0 No � Arg Dde Biotin Fmoc/HBTU/<br />

HOBt<br />

<strong>AB</strong> 430 Reagent-K<br />

2913 52.9 71.1 Yes � Pbf Dde D-Biotin Fmoc/HBTU/ <strong>AB</strong> 431A Modified<br />

HOBt Reagent-B<br />

4005 0 0 No 2 Biotin 1902 Dde Biotin-- Fmoc/PyBOP AMS 422 Modified<br />

caproates b daltons c amido- Reagent-B<br />

caproate<br />

NHS ester<br />

4209D 61.7 65.6 Yes Dde (�)Biotin Fmoc/HBTU ACT Modified<br />

396MBS Reagent-B<br />

(continued)<br />

JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000 159


L. BIBBS ET AL.<br />

T<strong>AB</strong>LE 1<br />

Summary of Results from <strong>AB</strong>RF Peptide Synthesis Research Group Study (continued)<br />

Percentage<br />

Mass Spectrometry Data<br />

Correct Product Major Product<br />

Other Lys<br />

RP- Correct Compo- Protec- Cleavage<br />

Samplea HPLC CE Mass Identity nents tion Biotin Chemistry Instrument Reagent<br />

IRBJ 0 0 No 2 Biotinsb Dde Biotin-N- Fmoc/HBTU ACT Modified<br />

hydroxysuccinimide<br />

MPS 396 Reagent-B<br />

1068 84.3 87.2 Yes � Pbf Dde NHS-LC- Fmoc/PyBOP Perseptive Reagent-K<br />

Biotin Pioneer<br />

1374 82.8 83.2 Yes � Ala Dde Succin- Fmoc/HBTU/ <strong>AB</strong> 431 Reagent-R<br />

imidyl<br />

D-Biotin<br />

HOBt<br />

5130 72.9 82.9 Yes Dde D-Biotin Fmoc/HOBt/<br />

DCC<br />

<strong>AB</strong> 431A Reagent-K<br />

5557 96.1 90 Yes Dde D-Biotin Fmoc/HBTU/<br />

HOBt<br />

<strong>AB</strong> 433 Reagent-K<br />

9818 92.1 92.5 Yes Dde D-Biotin Fmoc/HBTU/ <strong>AB</strong> 432 Modified<br />

HOBt Reagent-B<br />

9453 81.6 81.5 Yes Peptide Fmoc (�)Biotin Boc <strong>AB</strong> 430 90:10<br />

� 90 HF:Anisole<br />

1340T 96 90 Yes MeOtrt D-Biotin Fmoc/HBTU Rainin Modified<br />

Symphony Reagent-K<br />

0775 85.1 84 Yes � Biotinb Mtt (�)Biotin Fmoc/TBTU Rainin Modified<br />

Symphony Reagent-B<br />

1524 33.7 63.4 Yes (Biotin � Biotin Mtt Sulfo-NHS- Fmoc/TBTU/ Milligen Reagent-K<br />

caproate) caproate LC-Biotin HOBt 9050<br />

1609 74.1 66.1 Yes � Biotinb Mtt D-Biotin Fmoc/HBTU/<br />

HOBt<br />

<strong>AB</strong> 432 Reagent-R<br />

1633 60 55.8 Yes � Biotin Mtt EZ-link Fmoc/HOBt/ <strong>AB</strong> 431A Reagent-K<br />

NHS<br />

Biotin<br />

DCC<br />

3845 0 0 No � Biotin Mtt NHS-LC- Fmoc Opfp/ Milligen Modified<br />

Biotin HOBt 9050 Plus Reagent-B<br />

4209M 7 6.8 No � Biotin Mtt (�)Biotin Fmoc/HBTU ACT Modified<br />

396MBS Reagent-B<br />

7298 81.2 70.8 Yes � Biotinb Mtt D-Biotin Fmoc/HBTU/<br />

HOBt<br />

<strong>AB</strong> 433 Reagent-K<br />

7299 89.9 80.8 Yes Mtt D-Biotin Fmoc/HBTU/<br />

HOBt<br />

<strong>AB</strong> 433 Reagent-K<br />

7347 78 83.5 Yes � Biotinb Mtt (�)Biotin Fmoc/HBTU Perseptive<br />

Pioneer<br />

Reagent-B<br />

aThe samples are grouped according to Lys protection and then listed in numerical order.<br />

bDid not use Boc-Ala-OH.<br />

cThe extra mass is on the Lys(biotin) by tandem mass spectrometry.<br />

�, with and without; CE, capillary electrophoresis; RP-HPLC, reverse-phase high-performance liquid chromatography.<br />

160 JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000


FIGURE 4<br />

removed using dilute TFA or acetic acid/trifluoroethanol/methylene<br />

chloride (AcOH/TFE/DCM). 4<br />

4. Two laboratories used Fmoc-Lys(Aloc)-OH. One<br />

of these samples contained multiple components,<br />

with the product of interest at only 12.5%. For this<br />

synthesis, the Aloc group must be selectively<br />

removed using Pd(0),Bu 3 SnH, 5 or Pd(0), HOAc,<br />

N-methylmorpholine in chloroform. 6<br />

5. Two laboratories constructed their peptides using<br />

t-Boc chemistry. In both of these samples, the<br />

FIGURE 5<br />

STRATEGIES FOR SYNTHESIS OF L<strong>AB</strong>ELED PEPTIDES<br />

Electrospray ionization mass spectrum of a submitted sample of AEGKLRFK(biotin) in which there was a caproyl<br />

linker between biotin and the C-terminal Lys.The three charge states detected for this peptide indicated a molecular<br />

mass of 1286 daltons.<br />

product of interest was the major component by<br />

HPLC.<br />

Overall, nine of the samples did not have the product<br />

of interest as the major component by HPLC.<br />

Two types of biotin were used by the participating<br />

laboratories: biotin and biotin caproate (b-c). The<br />

caproate arm increased the mass of the correct peptide<br />

to 1286 daltons (Fig. 4) and caused an extra peak<br />

in the amino acid analysis corresponding to the<br />

Amino acid analysis of a peptide constructed with a caproyl linker to biotin. An extra peak was observed at<br />

28.6 minutes between Met and Lys.<br />

JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000 161


L. BIBBS ET AL.<br />

FIGURE 6<br />

Elution position of PTH-Lys biotin caproate(Lys(BC))<br />

by Edman sequence analysis.<br />

Lys(BC) was not readily resolved from<br />

PTH-Tyr using the gradient conditions<br />

described in Methods.<br />

derivatized caproate. (Fig. 5) The retention time of<br />

PTH-Lys(b-c) during Edman degradation analysis was<br />

within 0.1 minute of that of PTH-Tyr and therefore<br />

was not well resolved (Fig. 6).<br />

ESI/MS was a valuable tool in the analysis of<br />

these crude products. In peptide 8105, ESI/MS and<br />

subsequent ESI/MS/MS analysis rapidly identified a<br />

Glu-to-Gln substitution in the peptide in addition to a<br />

significant quantity of a truncated product (Figs. 7<br />

and 8). Analysis of this peptide illustrates the fact that<br />

tandem mass spectrometry can readily identify an<br />

amino acid substitution that would be undetected by<br />

amino acid analysis.<br />

FIGURE 7<br />

162 JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000<br />

Peptide 8398B was synthesized using a prelabeled<br />

resin. ESI/MS analysis indicated that, in addition<br />

to the expected biotin caproate peptide, there was an<br />

additional product which presumably corresponds to<br />

the peptide plus an extra Lys(b-c) (Fig. 9). This was<br />

verified by subsequent MS/MS analysis (Fig. 10). The<br />

mono-Lys(b-c) peptide constituted 17.5% of the sample<br />

by RP-HPLC.<br />

It is interesting to note that a variety of different<br />

cleavage cocktails were developed in the 1990s.<br />

These reagents contain different scavengers in TFA to<br />

reduce side reactions of the reactive cations generated<br />

from cleaved protecting groups. Reagent-K<br />

Electrospray ionization mass spectrum of sample 8105 containing a Glu-to-Gln substitution in the requested<br />

biotin-labeled peptide (A) in addition to a substantial quantity of a truncated product (B).


FIGURE 8<br />

FIGURE 9<br />

Tandem mass spectrum obtained by collision-induced dissociation of [M�2H] 2� (m/z 587.0) for the biotincontaining<br />

peptide in sample 8105 indicating the Glu-to-Gln substitution.<br />

Electrospray ionization mass spectrum of sample 8398B indicating the presence of peptides containing one (A)<br />

and two (B) residues of biotin caproate.<br />

JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000 163


L. BIBBS ET AL.<br />

FIGURE 10<br />

Tandem mass spectrum obtained by collision-induced dissociation of [M�2H] 2� (m/z 877.5) for the (biotin<br />

caproate) 2 -containing peptide in sample 8398B.The fragmentation pattern verifies the presence of two adjacent<br />

biotin caproates.<br />

(TFA-phenol-H 2 O-thioanisole-1,2-ethandithiol<br />

82.5:5:5:5:2.5), 7 Reagent-B (TFA-phenol-H 2 O-triisopropylsilane<br />

88:5:5:2), 8 and Reagent-R (TFA thioanisole-1,2-ethanedithiol-anisole<br />

(90:5:3:2) 9 are the<br />

most commonly used. Reagent-B is becoming more<br />

popular because it provides clean, highly efficient<br />

cleavage and does not contain any malodorous thiol<br />

components. Forty-one percent of the respondents<br />

used cleavage Reagent-B for this study.<br />

The results of this study showed that use of either<br />

a prelabeled amino acid or resin did not guarantee a<br />

good product; 37% of the peptides constructed using<br />

prelabeled starting materials were failures. It is also<br />

clear that when an Fmoc-Lys(Dde)-OH derivative is<br />

employed to introduce a label into a peptide, a Boc<br />

amino acid must be utilized for the N-terminus because<br />

the 2% hydrazine 3 employed for selective deprotection<br />

of the Dde group will also remove all Fmoc groups<br />

on the peptide. Finally, for characterization of synthetic<br />

peptides, MALDI-TOF/MS and ESI/MS were extremely<br />

valuable for qualitative, but not quantitative, analysis.<br />

164 JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000<br />

Considerable potential exists for the use of the<br />

Rink amide MBHA resin coupled with the Fmoc-<br />

Lys(Dde)-OH approach for the incorporation of various<br />

labels into peptides. One new and exciting application<br />

would be in the synthesis of peptides for <strong>FR</strong>ET.<br />

<strong>FR</strong>ET is based on the ability of molecules called<br />

quenchers to block fluorescence when in close proximity<br />

to fluorescent dyes. <strong>FR</strong>ET has been used for the<br />

identification of novel proteases. 10 Incorporating a<br />

fluorophore and a quencher during peptide synthesis<br />

would avoid labeling and purification problems in<br />

peptides used for future <strong>FR</strong>ET studies. It is hoped<br />

that core facilities will be able to make use of this<br />

strategy.<br />

ACKNOWLEDGMENTS<br />

We thank all the <strong>AB</strong>RF core facilities that participated in this<br />

study and Lynda Bonewald, our <strong>AB</strong>RF Executive Board liaison,<br />

for her valuable advice.


REFERENCES<br />

1. Angeletti RH, Bonewald LF, Fields G. Six-year study<br />

of peptide synthesis. Methods Enzymol 1997;289:697–<br />

717.<br />

2. Blackburn C, Kates SA. Solid-phase synthesis of cyclic<br />

homodetic peptides. Methods Enzymol 1997;289:175–<br />

197.<br />

3. Bycroft BW, Chan WC, Chhabra SR, Hone ND. A novel<br />

lysine protecting procedure for continuous flow solid<br />

phase synthesis of branched peptides. J Chem Soc Chem<br />

Commun 1993;778–779.<br />

4. Aletras A, Barlos K, Gatos D, Koutsogianni S, Mamos P.<br />

Preparation of the very acid-sensitive Fmoc-Lys(Mtt)-<br />

OH. Application in the synthesis of side-chain to sidechain<br />

cyclic peptides and oligolysine cores suitable for<br />

the solid-phase assembly of MAPs and TASPs Int J Pept<br />

Protein Res 1995;45:488–496.<br />

5. Loffet A, Zhang HX. Allyl-based groups for side-chain<br />

protection of amino-acids. Int J Pept Protein Res 1993;<br />

42:346–351.<br />

STRATEGIES FOR SYNTHESIS OF L<strong>AB</strong>ELED PEPTIDES<br />

6. Kates SA, Daniels SB, Albericio F. Automated allyl<br />

cleavage for continuous-flow synthesis of cyclic and<br />

branched peptides. Anal Biochem 1993;212:303–310.<br />

7. King DS, Fields CG, Fields GB. A cleavage method<br />

which minimizes side reactions following Fmoc solid<br />

phase peptide synthesis. Int J Pept Protein Res 1990;36:<br />

255–266.<br />

8. Sole NA, Barany G. Optimization of solid-phase synthesis<br />

of [Ala 8]-dynorphin A. J Org Chem 1992;57:5399–<br />

5403.<br />

9. Albericio F, Kneib-Cordonier N, Biancalana S, Gera L,<br />

Masada RI, Hudson D, Barany G. Preparation and application<br />

of the 5-(4-(9-fluorenylmethyloxycarbonyl)aminomethyl-3,5-dimethoxyphenoxy)valeric<br />

acid (PAL) handle<br />

for the solid phase synthesis of C-terminal peptide<br />

amides under mild conditions. J Org Chem 1990;55:<br />

3730–3743.<br />

10. Olsen MJ, Stephens D, Griffiths D, Daugherty P, Georgiou<br />

G, Iverson BL. Function-based isolation of novel<br />

enzymes from a large library. Nat Biotechnol 2000;18:<br />

1071–1074.<br />

JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000 165


Research Equipment and<br />

Resource Requirements of<br />

NIH-Supported Investigators:<br />

An Assessment of<br />

Current Conditions and<br />

Recommendations for<br />

Future Funding<br />

and Programs<br />

Tamara R. Zemlo, PhD, MPH;<br />

Howard H. Garrison, PhD;<br />

David Lester, PhD; Mustafa<br />

Lokhandwala, PhD; Fred Naider,<br />

PhD; Stephen White, PhD;<br />

and David W. Speicher, PhD<br />

The Federation of American Societies for<br />

Experimental Biology, Bethesda, MD<br />

Reprinted with permission from FASEB © 2000.<br />

ADDRESS CORRESPONDENCE AND REPRINT REQUESTS TO: The Federation<br />

of American Societies for Experimental Biology, Office of<br />

Public Affairs, 9650 Rockville Pike, Bethesda, MD 20814-3998<br />

(www.faseb.org/opar).<br />

166 JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000<br />

REPORT<br />

EXECUTIVE SUMMARY<br />

Journal of Biomolecular Techniques<br />

11:166–176<br />

RF <strong>AB</strong><br />

In response to a growing level of concern over<br />

unmet needs for advanced instrumentation, the <strong>Science</strong><br />

Policy Committee of the Federation of American<br />

Societies for Experimental Biology (FASEB) decided<br />

to undertake a systematic look at the way in which<br />

current programs were meeting researchers’ needs<br />

for major equipment. In November of 1999, a questionnaire<br />

was mailed to 1,000 recipients of R01<br />

awards from the National Institutes of Health (NIH)<br />

to determine the researchers’ views about the following<br />

issues: degree of dependence on instrumentation;<br />

current funding arrangements; access to<br />

equipment; and adequacy of funding for instrumentation,<br />

particularly the Shared Instrumentation Grants<br />

(SIG) program; and priorities for future federal funding.<br />

508 scientists, representing 51 percent of the<br />

sample, returned completed surveys. Survey respondents<br />

were similar to all NIH R01 grantees in terms of<br />

institutional affiliations, experience as principal investigators<br />

and laboratory budgets.<br />

There was strong agreement about the importance<br />

of advanced instrumentation for NIH-funded<br />

research. Most of the survey respondents (84 percent)<br />

responded that, “shared equipment and core facilities<br />

at my institution are extremely important for my<br />

research.” However, a substantial number of scientists<br />

reported difficulty in obtaining adequate access to<br />

these critical technologies. Almost half of the respondents<br />

(48 percent) felt that their institutions were not<br />

able to implement new, shared research instrumentation<br />

and resources in a timely manner. Only a small<br />

percentage of the respondents (5 percent) were<br />

strongly supportive of the view that their institutions<br />

were able to adopt new technologies in a timely<br />

manner.<br />

A sizeable portion of the survey respondents also<br />

questioned the adequacy of current funding for


shared research equipment. When asked if current<br />

funding for the SIG Program was adequate, 42 percent<br />

of the respondents responded that it was not.<br />

A smaller fraction of the respondents (26 percent)<br />

felt that the current level of support for the SIG budget<br />

was adequate. Survey respondents expressed<br />

even more dissatisfaction over the funds available for<br />

shared equipment not covered by the SIG program<br />

(instruments costing less than $100,000 involving multiple<br />

users). In this case, 55 percent of the respondents<br />

did not believe that “the level of funding support<br />

from NIH was adequate” and only 17 percent of<br />

the researchers supported this statement. Consistent<br />

with these findings, the survey respondents placed a<br />

high priority on having NIH increase the level of funding<br />

for research equipment. A majority of them (65<br />

percent) indicated that increased funding for equipment<br />

and equipment-related categories should be one<br />

of the top priorities in an expanding federal research<br />

budget. In contrast, only 13 percent of the respondents<br />

disagreed with this position. Consistent with this perception,<br />

survey respondents also placed a very high<br />

priority on increased funding for new specialized research<br />

equipment within their own research laboratories.<br />

For shared equipment and resources, respondents<br />

indicated that NIH should invest more funds in<br />

establishing new resource facilities employing emerging<br />

technologies.<br />

The overall perception of the survey respondents<br />

was that equipment and equipment-related needs<br />

were unmet and represented a serious problem in the<br />

extramural research community. Data from the survey<br />

indicates that NIH should invest additional funds to<br />

meet these needs.<br />

RECOMMENDATIONS<br />

• FASEB recommends that NIH increase its level of<br />

support for shared equipment costing $100,000 or<br />

more to $150 million per year for FY2001 with<br />

appropriate incremental increases thereafter.<br />

• FASEB recommends that NIH increase its level of<br />

support within the context of existing grant mechanisms<br />

such as R01s and P01s for equipment costing<br />

less than $100,000 to $50 million per year for<br />

FY2001.<br />

These amounts actually reflect the minimum<br />

funding requirements that exist in the biomedical<br />

research community because the survey results were<br />

extrapolated to only the NIH R01-population and do<br />

not capture the needs of investigators supported by<br />

other NIH funding mechanisms or non-NIH funded<br />

scientists.<br />

INSTRUMENTATION NEEDS<br />

FASEB also proposes that an expanded SIG program<br />

be improved by:<br />

• Decreasing the time from receipt of application to<br />

award from the current approximately one year to<br />

six months.<br />

• Increasing the number of review cycles from one<br />

to three per year.<br />

• Raising the caps to at least $1 million to authorize<br />

the purchase of more expensive equipment and<br />

to compensate for inflation pressures.<br />

• Allowing applicants to bundle two or more unrelated<br />

pieces of equipment that together cost more<br />

than $100,000.<br />

• Providing support for a maintenance agreement<br />

for up to three years if included in the instrument<br />

purchase price.<br />

• Permitting the establishment of facilities using<br />

components rather than strictly commercial instruments.<br />

• Using standing rather than ad hoc study sections<br />

for reviewing SIG grant proposals so that consistency<br />

in the review process is maintained.<br />

• Selecting peer reviewers with adequate expertise<br />

in emerging technologies.<br />

THE SHARED INSTRUMENTATION<br />

GRANTS PROGRAM<br />

For almost twenty years, the Shared Instrumentation<br />

Grant (SIG) program has allowed scientists to obtain<br />

equipment and equipment-related items that would<br />

be too expensive to purchase with research project<br />

grant funding. In existence since 1982 and administered<br />

by the National Center for Research Resources<br />

(NCRR), the SIG program has provided researchers<br />

with grants ranging between $100,000 to $500,000 to<br />

procure state-of-the-art instrumentation. Instruments<br />

purchased with SIG funds must be shared by at least<br />

three National Institutes of Health (NIH)-supported<br />

researchers.<br />

The budgets of the SIG Program have not kept<br />

pace with the expansion of biomedical research in the<br />

1990s. SIG awards totaled $32.5 million per year in<br />

1990 and 1991, with no appreciable growth in program<br />

funds since 1985. In 1992, SIG funds were reduced<br />

to $8.8 million and did not rise above $10 million<br />

per year until 1996. Only in 1999 did the SIG<br />

budget return to the 1990 level. But the 1999 budget<br />

for SIG grants ($34.2)—after adjusting for inflation—<br />

had less purchasing power than the 1990 budget. In<br />

2000, the SIG budget rose to $43.1 million. But when<br />

expressed in 1990 dollars ($30.3 million), it is still<br />

below the purchasing power of the program in 1990.<br />

JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000 167


FEDERATION OF AMERICAN SOCIETIES FOR EXPERIMENTAL BIOLOGY<br />

Moreover, during this same period the number of<br />

research project grants funded by NIH grew from<br />

23,177 in 1990 to a projected 32,942 in 2000 (42 percent),<br />

while the research project grant budget increased<br />

from $4,640 million to $7,103 million in 1990<br />

dollars (53.1 percent). 1<br />

THE ORIGIN OF THE STUDY<br />

Over the past several years, the leadership of the Federation<br />

of American Societies for Experimental Biology<br />

(FASEB) and its member societies have been<br />

hearing reports of unmet instrumentation needs.<br />

These concerns have been raised in society council<br />

meetings, in letters from concerned scientists and at<br />

consensus conferences. The shared instrumentation<br />

issue has been reviewed in nearly every FASEB Federal<br />

Funding Consensus Conference from 1993–1999.<br />

In the face of these persistent but informal indicators<br />

of a more general problem, the Instrumentation and<br />

Infrastructure Subcommittee of the <strong>Science</strong> Policy<br />

Committee (David Lester, PhD; Mustafa Lokhandwala,<br />

PhD; Fred Naider, PhD; Stephen White, PhD; and<br />

David Speicher, PhD, chair) decided to conduct a<br />

detailed investigation into this issue. Representatives<br />

of the committee met with Judy Vaitukaitis, MD,<br />

Director of the NCRR, who provided an overview of<br />

NCRR initiatives and data on the Center’s programs.<br />

The committee also collected available information<br />

on instrumentation assessments conducted by the<br />

National <strong>Science</strong> Foundation (NSF) and co-sponsored<br />

by the NIH. 2<br />

After reviewing the existing data, the committee<br />

concluded that there was a shortage of current information<br />

on investigators’ instrumentation needs. The<br />

last NSF/NIH instrumentation survey, published in<br />

1994, was based on the views of institutional administrators<br />

and did not necessarily reflect the views of<br />

individual practicing scientists. Another instrumentation<br />

study, an NCCR-contracted overview of the SIG<br />

program, was conducted using data from 1993. 3 Due<br />

to the relative paucity of current data, the committee<br />

decided to take a more systematic look at the way in<br />

which current programs were meeting researchers’<br />

needs for advanced equipment.<br />

To derive an objective standard of “need,” the<br />

committee chose to focus on a major group of scientists<br />

whose work had already been deemed meritorious:<br />

recipients of NIH R01 awards. A series of questions<br />

was developed to determine the researchers’<br />

views about the importance of instrumentation, current<br />

funding arrangements, access to equipment, and<br />

adequacy of funding for instrumentation and priorities<br />

for future federal funding.<br />

168 JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000<br />

This report contains the results of the survey of<br />

R01 funded investigators. Under the direction of<br />

David W. Speicher, of the Wistar Institute in Philadelphia,<br />

PA, and with the collaboration of Tamara R.<br />

Zemlo and Howard H. Garrison of the FASEB Office<br />

of Public Affairs, the Instrumentation and Infrastructure<br />

Subcommittee developed the survey report. The<br />

FASEB <strong>Science</strong> Policy Committee reviewed the final<br />

report. On June 6th the FASEB Public Affairs Executive<br />

Committee on behalf of the FASEB Board of<br />

Directors accepted it.<br />

SURVEY METHODS<br />

FASEB obtained a file listing the names and addresses<br />

of all FY 1998 recipients of research project grants<br />

from NIH. 4 From this file containing the records for<br />

21,145 R01s awarded to 14,745 investigators, a random<br />

sample of 1,000 principal investigators was selected.<br />

In November of 1999, each member of the<br />

sample was sent a short letter announcing the study. 5<br />

Two weeks later the NIH-funded investigators<br />

received a copy of the survey questionnaire asking<br />

them for their views on the adequacy of federal support<br />

for instrumentation, the availability of shared<br />

resources, and their recommendations regarding federal<br />

funding priorities for equipment.<br />

Completed surveys were returned by 508 scientists,<br />

51 percent of the sample when the survey closed<br />

in February of 2000. Some members of the sample<br />

(e.g., behavioral scientists, epidemiologists, and some<br />

clinical researchers) may have not returned questionnaires<br />

because their research did not make extensive<br />

use of instrumentation. To address the possibility of<br />

selection bias in survey responses, characteristics of<br />

the survey respondents were compared to those of<br />

the entire grantee population and/or the survey nonrespondents<br />

on three dimensions: experience as head<br />

of a laboratory, institutional affiliation and laboratory<br />

budgets. On all three measures, no evidence of selection<br />

bias was found. 6<br />

RESULTS<br />

Importance of Shared Equipment<br />

Most of the survey respondents (61 percent) strongly<br />

agreed with the statement “shared equipment and<br />

core facilities at my institution are extremely important<br />

for my research” (Fig. 1). Another 23 percent agreed<br />

with this statement for a combined total of nearly<br />

84 percent. This overwhelming appreciation of the<br />

importance of shared equipment is very notable and


provides a striking endorsement of the role of advanced<br />

instrumentation in NIH-funded research.<br />

Access to Equipment<br />

While NIH-funded investigators reported that shared<br />

equipment was very important to their research programs,<br />

large numbers encountered difficulties obtaining<br />

access to such instrumentation. Only one half of<br />

the survey respondents felt that their institutions adequately<br />

supported shared research resources such as<br />

common use equipment and specialized core facilities<br />

(12 percent strongly agreed and 38 percent agreed)<br />

FIGURE 2<br />

My institution adequately supports shared<br />

research resources.<br />

FIGURE 1<br />

INSTRUMENTATION NEEDS<br />

Shared equipment and core facilities are<br />

essential to my research.<br />

(Fig. 2). One third of the respondents indicated that<br />

their institutions have not adequately supported<br />

shared research resources (12 percent strongly disagreed<br />

and 21 percent disagreed with the survey<br />

question).<br />

A large fraction of the researchers polled (48 percent)<br />

felt that their institutions were not able to<br />

implement new shared research technologies and<br />

required associate equipment in a timely manner (13<br />

percent strongly disagreed and 35 percent disagreed)<br />

(Fig. 3). Only 5 percent of the respondents<br />

strongly agreed and 24 percent agreed with a<br />

statement indicating that their institutions were able<br />

to adopt new technologies in a timely fashion.<br />

JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000 169


FEDERATION OF AMERICAN SOCIETIES FOR EXPERIMENTAL BIOLOGY<br />

FIGURE 3<br />

New shared resource technologies are<br />

implemented in a timely manner at my<br />

institution.<br />

Funding for Instrumentation<br />

Substantial numbers of survey respondents expressed<br />

concern about the adequacy of current funding for<br />

research equipment. When asked if current funding<br />

and management of the NIH SIG program was adequate,<br />

10 percent strongly disagreed and another<br />

32 percent of the respondents disagreed (Fig. 4).<br />

Just over one-quarter of the respondents felt that the<br />

SIG program was adequately funded and managed<br />

(4 percent strongly agreed and 22 percent agreed).<br />

The survey respondents expressed even more<br />

dissatisfaction over the funds available for shared<br />

equipment costing less than $100,000. In this case,<br />

55 percent of the respondents disputed the statement<br />

that the level of support from NIH was adequate:<br />

170 JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000<br />

17 percent strongly disagreed and nearly two in<br />

five respondents—38 percent—disagreed (Fig. 5).<br />

Only 17 percent of the researchers polled strongly<br />

agreed or agreed with the statement that NIH funding<br />

for this equipment category was adequate. Results<br />

were very similar for a question about support<br />

for equipment used within the researchers’ own laboratory<br />

costing less than $100,000. Over half of the<br />

respondents (52 percent) rejected the statement that<br />

the level of support from NIH for smaller equipment<br />

for individual laboratories was adequate (Fig. 6). Of<br />

these respondents, 17 percent strongly disagreed<br />

and 35 percent disagreed. A much smaller fraction,<br />

32 percent, reported that funding in this area was<br />

adequate (28 percent strongly agreed and 4 percent<br />

agreed).<br />

FIGURE 4<br />

NIH’s current level of support and management<br />

of the SIG program is adequate.


Survey Respondents’ Priorities for<br />

Federal Funding<br />

When asked if increased funding for equipment and<br />

equipment-related categories should be one of the top<br />

priorities in an expanding federal research budget, 24<br />

percent strongly agreed and another 41 percent of<br />

the sample agreed with the statement (Fig. 7). Only<br />

13 percent of the respondents dissented from this<br />

position. Consistent with the perception that increased<br />

equipment funding should be a major priority, a high<br />

percentage of survey respondents indicated that there<br />

should be increased funding for new specialized<br />

research equipment 7 within their own research laboratories<br />

(Fig. 8). Following this, they identified having<br />

adequate funds to either replace or purchase new<br />

FIGURE 6<br />

NIH’s current level of support for<br />

equipment in my own laboratory costing<br />

$5–$100K is adequate.<br />

FIGURE 5<br />

INSTRUMENTATION NEEDS<br />

NIH’s current level of support for shared<br />

equipment �$100K is adequate.<br />

routine use equipment 8 as the next highest set of<br />

priorities for their own laboratories. For shared equipment<br />

and resources, respondents indicated that the<br />

most important priority for government funding<br />

would be to establish new resource facilities employing<br />

emerging technologies (Fig. 9). The next most<br />

important categories respondents believed should<br />

merit support were purchasing new specialized<br />

shared equipment and providing adequate technical<br />

and/or maintenance support for shared resources.<br />

Survey respondents were asked to specify the<br />

shared equipment and equipment-related items they<br />

would need to carryout their research programs<br />

between 2000 and 2002 (Fig. 10). For items costing<br />

less than $100,000, PCR systems were identified as the<br />

greatest unmet need, and for items costing $100,000<br />

JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000 171


FEDERATION OF AMERICAN SOCIETIES FOR EXPERIMENTAL BIOLOGY<br />

FIGURE 7<br />

Increased federal funding for equipment<br />

should be a top priority.<br />

or more imaging technologies such as ultrasound,<br />

MRI and CT were identified as the greatest unmet<br />

need. To estimate the total cost of the annual shared<br />

equipment needs of NIH investigators, the sum of the<br />

cost for all the items survey respondents identified as<br />

necessary for their future research was extrapolated to<br />

the entire R01 population. The responses were divided<br />

into two categories: those items costing less<br />

than $100,000 and those items costing $100,000 or<br />

more. From information provided by the National<br />

Center for Research Resources on the average number<br />

of users for shared instrumentation in the SIG program,<br />

the number of users for each equipment category<br />

was estimated. 9 From these approximations and<br />

the survey data, FASEB calculates that approximately<br />

$75 million annually would be needed to meet the<br />

equipment and equipment-related needs for those<br />

172 JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000<br />

items costing less than $100,000, and $150 million<br />

annually would be needed for those items costing<br />

$100,000 or more for the next three years.<br />

CONCLUSIONS<br />

The overwhelming majority of the respondents to the<br />

survey of R01 grantees indicated that shared research<br />

resources are important elements of their research<br />

programs. Yet nearly one half of the respondents<br />

(48 percent) indicated that their institutions were<br />

unable to meet their shared equipment needs in a<br />

timely manner. Nearly two-thirds of the respondents<br />

reported that increased funding for shared equipment<br />

should be a top priority in an expanding research<br />

budget.<br />

FIGURE 8<br />

Choices for government-supported<br />

equipment spending within my laboratory.


FIGURE 9<br />

FIGURE 10<br />

Choices for government-supported equipment spending for shared resources.<br />

Survey Respondents’ Shared Equipment Needs:<br />

Top Ten Categories Ranked by Cost a<br />

Rank Order b


FEDERATION OF AMERICAN SOCIETIES FOR EXPERIMENTAL BIOLOGY<br />

These views described above were consistent<br />

across subsets of the survey sample, reflecting general<br />

rather than isolated sentiments. Comparisons across<br />

categories of seniority (number of years in charge of<br />

a laboratory), primary research institution (medical<br />

school, university, etc.), research budget, percentage<br />

of funding from NIH, total cost of equipment or percentage<br />

of equipment budget from NIH revealed very<br />

few statistically significant differences of opinions.<br />

The perception that instrumentation needs were<br />

unmet and represented a serious problem was widespread<br />

and pervasive in the extramural research community.<br />

It was not a phenomenon limited to newer<br />

investigators, scientists with smaller grants, or researchers<br />

with modest equipment budgets.<br />

FASEB RECOMMENDATIONS<br />

FASEB recommends that NIH significantly increase<br />

resources to meet the equipment and equipmentrelated<br />

needs of the researchers it funds. While it<br />

believes NIH’s foremost obligation is still to support<br />

investigator-initiated research, FASEB recognizes that<br />

having access to the appropriate equipment is essential<br />

for conducting investigator-initiated research.<br />

These research tools include routine-use items like<br />

incubators and centrifuges and specialized equipment<br />

like micro array instruments and x-ray diffractometers.<br />

Bioinformatic hardware and software packages are<br />

also important to fund since they interface multiple<br />

high-tech pieces of equipment and store and process<br />

large volumes of data. To catalyze the pace of discovery<br />

in biomedicine, NIH should provide greater<br />

opportunities for SIG program funding. This critical<br />

infusion of support will maximize the rate of return<br />

on our investment in scientific research.<br />

• FASEB recommends that NIH increase their level<br />

of support for shared equipment costing $100,000<br />

or more to $150 million10 per year for FY2001<br />

with appropriate incremental increases thereafter<br />

• FASEB recommends that NIH increase its level of<br />

support within the context of existing grant mechanisms<br />

such as R01s and P01s for equipment costing<br />

less than $100,000 to $50 million11 per year<br />

for FY2001.<br />

In this era of enhanced support for biomedical<br />

research, FASEB recommends that NIH equipmentfunding<br />

mechanisms be substantially expanded to<br />

provide for a broader range of new technologies in<br />

order to make the most of promising research opportunities.<br />

A greatly expanded and more responsive SIG<br />

program would allow scientists to procure necessary<br />

equipment in a timelier manner. To help achieve<br />

174 JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000<br />

these objectives, FASEB proposes that the program be<br />

improved by:<br />

• Decreasing the time from receipt of application to<br />

award from the current approximately one year to<br />

six months.<br />

• Increasing the number of review cycles from one<br />

to three per year.<br />

• Raising the caps to at least $1 million to authorize<br />

the purchase of more expensive equipment and<br />

to compensate for inflation pressures.<br />

• Allowing applicants to bundle two or more unrelated<br />

pieces of equipment that together cost more<br />

than $100,000.<br />

• Providing support for a maintenance agreement<br />

for up to three years if included in the instrument<br />

purchase price.<br />

• Permitting the establishment of facilities using<br />

components rather than strictly commercial instruments.<br />

• Using standing rather than ad hoc study sections<br />

for reviewing SIG grant proposals so that consistency<br />

in the review process is maintained.<br />

• Selecting peer reviewers with adequate expertise<br />

in emerging technologies.<br />

ACKNOWLEDGMENTS<br />

The FASEB Office of Public Affairs wishes to thank the<br />

members of the Instrumentation and Infrastructure Subcommittee<br />

of the <strong>Science</strong> Policy Committee for the valuable<br />

time they devoted to this report.<br />

The FASEB <strong>Science</strong> Policy Committee<br />

David L. Brautigan, Ph.D., Center for Cell Signaling, University<br />

of Virginia. (FASEB Vice President for <strong>Science</strong><br />

Policy)<br />

Joseph R. Haywood, Ph.D. Professor, Department of Pharmacology,<br />

University of Texas Health <strong>Science</strong> Center.<br />

(American Physiological Society)<br />

Frederick Grinnell, Ph.D. Professor, Department of Cell<br />

Biology and Neuroscience, University of Texas Southwestern<br />

Medical Center. (American Society for Biochemistry<br />

and Molecular Biology)<br />

*Mustafa F. Lokhandwala, Ph.D. Dean and Professor,<br />

Department of Pharmacology, University of Houston<br />

College of Pharmacy. (American Society for Pharmacology<br />

and Experimental Therapeutics)<br />

Richard G. Lynch, M.D. Professor and Head, Department<br />

of Pathology, University of Iowa College of Medicine.<br />

(American Society for Investigative Pathology)<br />

Patsy M. Brannon, Ph.D. Dean, College of Human Ecology,<br />

Cornell University. (American Society for Nutritional<br />

<strong>Science</strong>)<br />

Jeffrey A. Frelinger, Ph.D. Kenan Professor and Chairman,<br />

University of North Carolina, Chapel Hill. (The<br />

American Association of Immunologists)<br />

Samuel C. Silverstein, M.D. Professor and Chairman,<br />

Department of Physiology and Cell Biophysics, Colum-


FIGURE 11<br />

Distribution of NIH Research Project Grants by Institution, 1998<br />

bia University College of Physicians and Surgeons.<br />

(American Society for Cell Biology)<br />

*Stephen H. White, Ph.D. Professor, Department of Physiology<br />

and Biophysics, University of California–Irvine.<br />

(Biophysical Society)<br />

*David S. Lester, Ph.D. Pharmacologist, Food and Drug<br />

Administration. (American Association of Anatomists)<br />

Tony E. Hugli, Ph.D. Professor, Department of Immunology,<br />

Scripps Research Institute. (The Protein Society)<br />

Nicola C. Partridge, Ph.D. Professor, Department of Pharmacology<br />

and Physiological <strong>Science</strong>, St. Louis University<br />

School of Medicine. (The American Society for<br />

Bone and Mineral Research)<br />

Edward J. Benz, M.D. Department of Medicine, Johns Hopkins<br />

University. (American Society for Clinical Investigation)<br />

Henry M. Kronenberg, M.D. Professor of Medicine and<br />

Chief, Endocrine Unit, Massachusetts General Hospital.<br />

(The Endocrine Society)<br />

Philip R. Reilly, M.D., J.D., President/CEO, Eunice<br />

Kennedy Shriver Center, Waltham, Mass. (The American<br />

Society of Human Genetics)<br />

Associate Members (non-voting)<br />

Margaret S. Saha, Ph.D. Professor, Department of Biology,<br />

College of William and Mary. (Society for Developmental<br />

Biology)<br />

*Fred R. Naider, Ph.D. Professor, Department of Chemistry,<br />

College of Staten Island, CUNY. (American Peptide<br />

Society)<br />

*David W. Speicher, Ph.D. Professor, The Wistar Institute,<br />

University of Pennsylvania. (<strong>AB</strong>RF)<br />

Barry R. Zirkin, Ph.D. Department of Biochemistry, Division<br />

of Reproductive Biology, Johns Hopkins University<br />

School of Hygiene and Public Health. (Society for<br />

the Study of Reproduction)<br />

Thomas B. Knudsen, Ph.D. Professor, Department of Anatomy,<br />

Jefferson Medical College. (Teratology Society)<br />

Ex Officio (non-voting)<br />

David G. Kaufman, M.D., Ph.D. Professor, Department of<br />

Pathology and Laboratory Medicine, University of<br />

North Carolina, Chapel Hill. (FASEB President)<br />

INSTRUMENTATION NEEDS<br />

1998 NIH Research Project Grants FASEB Survey<br />

Institution Number Percentage Dollar, K Percentage Percentage<br />

Medical School 14,898 54.3% 4,141,424 54.3% 52%<br />

University 7,661 27.9% 2,129,645 27.9% 37%<br />

For-Profit 103 0.4% 28,632 0.4% 1.1%<br />

Hospital 2,157 7.9% 599,614 7.9% 2.4%<br />

Nonprofit 2,641 9.6% 734,159 9.6% 7.5%<br />

Totals 27,460 100.0% 7,633,474 100.0% 100%<br />

Distribution of NIH research project grants by institution, 1998.<br />

Sue P. Duckles, Ph.D. Professor and Associate Dean, Department<br />

of Pharmacology, University of California–<br />

Irvine College of Medicine. (FASEB Vice-President Elect<br />

for <strong>Science</strong> Policy)<br />

Mary J.C. Hendrix, Ph.D. Professor and Head, Department<br />

of Anatomy and Cell Biology, University of<br />

Iowa—College of Medicine. (FASEB President Elect)<br />

Sidney H. Golub, Ph.D. (FASEB Executive Director)<br />

*Members of the Instrumentation and Infrastructure Subcommittee.<br />

ENDNOTES<br />

1. NIH Competing and Noncompeting Research Project<br />

Grants by Type at http://silk.nih.gov/public/cbz2zoz.<br />

@www.trends99.rpgtype.fy9099.html and FY2001 President’s<br />

Budget Press Briefing at http://www4.od.nih.gov/<br />

ofm/budget/fy2001Pressbriefing.htm<br />

2. The National Survey of Academic Research Instruments<br />

and Instrumentation Needs (Instrumentation Survey) is<br />

a congressionally mandated program that collects data<br />

concerning scientific research instruments and the academic<br />

departments and facilities in which they are located.<br />

3. Evaluation of NIH Shared Instrumentation Grant Program:<br />

Reports From Users, published in 1996.<br />

4. The authors would like to thank Bob Moore of the NIH<br />

Office of Extramural Research for assistance in obtaining<br />

information on NIH R01 recipients.<br />

5. Bohne Silber and colleagues at Silber & Associates,<br />

Clarksville, MD conducted the survey and data tabulation.<br />

6. The respondents’ primary professional appointment<br />

and their annual direct cost laboratory budgets corresponded<br />

closely with the profile of the NIH R01 population.<br />

Just over one-half of the survey respondents<br />

(52%) reported that their primary appointment was in<br />

a medical school. A nearly identical fraction of all<br />

research project grants from the NIH (54%) were made<br />

to investigators with medical school affiliations (Fig.<br />

11). There is a slight over-representation of universitybased<br />

researchers in the survey respondent sample,<br />

but in all other categories the distribution of survey<br />

JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000 175


FEDERATION OF AMERICAN SOCIETIES FOR EXPERIMENTAL BIOLOGY<br />

respondents is very similar to that of all NIH grantees.<br />

The over-representation of university-based researchers<br />

in the survey sample may reflect the fact that this group<br />

was selected only from the population of R01 grantees<br />

rather than recipients of all types of research project<br />

grants.<br />

The median direct cost laboratory budget for the survey<br />

respondents is $300K. By taking the median direct<br />

cost of all competing R01 awards in FY 1999, which<br />

was $171K and multiplying it by 1.43 (the average<br />

number of R01s per laboratory) and dividing it by 77%<br />

(the average percentage of the direct cost budget from<br />

NIH), one can obtain an estimate of the direct cost laboratory<br />

budget for NIH R01-supported investigators.<br />

This amount is $319K, which compares reasonably<br />

with the $300K amount from the survey.<br />

It was also possible to compare the survey respondents<br />

and non-respondents in terms of the length of<br />

time a particular R01 award was retained. Each NIH<br />

grant is assigned a unique identification number, part<br />

of which consists of a “year of support” field. This field<br />

indicates the number of continuous years of support<br />

for that grant. For survey respondents, the distribution<br />

was 54% for R01s held for less than five years, 25% for<br />

R01s held between five and ten years, and 21% for<br />

R01s held for greater than 10 years. For the nonrespondents,<br />

these percentages were 54%, 28% and<br />

18%, respectively. [If an investigator had multiple R01<br />

grants, only the longest-held grant was used in the<br />

analysis.]<br />

7. Specialized equipment refers to instruments found in<br />

limited numbers in a department or entire institution.<br />

Examples are biosensors, cell sorters, mass spectrometers,<br />

NMR instruments, confocal microscopes, biomedical<br />

imagers, DNA sequencers, X-ray diffractometers,<br />

analytical ultracentrifuges, microarray instruments and<br />

bioinfomatics hardware/software.<br />

176 JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000<br />

8. Routine use equipment is common research equipment<br />

items typically found in most labs and/or departmental<br />

common use rooms. Examples are tissue culture hoods<br />

and incubators, preparative centrifuges, UV spectrophotometers,<br />

scintillation counters and freezers.<br />

9. From the Evaluation of NIH Shared Instrumentation<br />

Grant Program: Reports From Users (NCRR), the average<br />

number of major users per instrument is 5.2. A<br />

major user is defined as a Public Health Service (PHS)supported<br />

investigator who accounts for a significant<br />

fraction of instrument use time. The average number of<br />

minor users per instrument is 9.0. A minor user may be<br />

either PHS-sponsored investigators whose individual<br />

share does not constitute a significant percentage of<br />

average use time, or investigators whose instrument<br />

research is not supported by PHS. Because most of the<br />

instruments costing $100,000 or more will likely be<br />

shared, the committee assumed that the number of<br />

users should be a minimum of five and a maximum of<br />

fourteen (five major users and nine minor users). However,<br />

it is unlikely that all of the minor users would be<br />

R01 recipients. Therefore, the committee estimated ten<br />

users (five major users and five minor users) for instruments<br />

costing $100,000 or more. For instruments costing<br />

less than $100,00, some instruments would be<br />

shared and some would not be, so the committee estimated<br />

that the average number of users is approximately<br />

five.<br />

10. Since funding for high-end equipment (instrumentation<br />

costing more than $100,000) is extremely limited,<br />

FASEB recommends that NIH provide funding for the<br />

total estimated need of the research community: $150<br />

million.<br />

11. For equipment costing less than $100,000 (for which<br />

there are other sources of support) FASEB recommends<br />

that NIH fund two-thirds of the $75 million estimated<br />

need: $50 million.


BOOK REVIEWS<br />

Amino Acid Analysis Protocols. Edited by Catherine Cooper,<br />

Nicolle Packer, and Keith Williams. Methods in<br />

Molecular Biology series number 159.Totowa, NJ: Humana<br />

Press, September 2000. Hardcover, 280 pp, $84.50<br />

US.<br />

This volume is what the title states; it is intended<br />

as a benchtop reference that provides recipes and<br />

procedures for amino acid analysis. An introductory<br />

chapter by Margaret Tyler provides guidance to the<br />

topics found among the 17 other chapters of the<br />

book. Of these, several focus on standard amino acid<br />

analysis strategies commonly found in core facilities:<br />

postcolumn ninhydrin and AQC (AccuTag). The former,<br />

by Macchi and colleagues, is focused on modifications<br />

that occur (at low levels) during protein<br />

expression in a pharmaceutical setting. Here the ninhydrin<br />

system is well suited to resolving minor components<br />

(e.g., norleucine, hydroxylysine, amino sugars,<br />

and carboxmethylCys) amidst the high levels<br />

analyzed. One useful feature of this article is a clear<br />

illustration of typical (Excel) spreadsheet calculations<br />

employed to obtain protein amino acid compositions.<br />

The article on AQC by S. A. Cohen is clear and helpful,<br />

particularly in providing the buffer recipes for the<br />

analysis using Waters’ proprietary column. Shindo and<br />

colleagues describe an application of AQC analysis to<br />

identify blotted protein using the ExPASy Web Site.<br />

The authors correctly emphasize the need for good<br />

hygiene in sample handling and provide diagrams for<br />

an isolation box and their version of a hydrolysis<br />

vessel. In several places, the reader is reminded that<br />

desalting protein samples is important for accurate<br />

compositional analysis. To address this issue, there is<br />

a brief contribution from Zhang and Denslow that<br />

details simple desalting protocols.<br />

Phenylisothiocyanate (PITC) as detection reagent<br />

is discussed in comparison with new reagents and in<br />

the analysis of complex biologic matrices. Woo presents<br />

a comparison between butyl-isothiocyanate–<br />

(BITC-) and benzyl-isothiocyanate– (BZITC-) derivatized<br />

amino acids and the familiar PITC approach.<br />

Journal of Biomolecular Techniques<br />

11:177–178 © 2000 <strong>AB</strong>RF<br />

RF <strong>AB</strong><br />

The primary motive was to shorten analysis time and<br />

increase convenience by finding a volatile reagent. In<br />

contrast to PTC chromatography, both BITC and<br />

BZITC compounds provide separation of cysteine<br />

and cystine. Although BITC possessed the desired<br />

volatility, resolution on-column suffered (i.e., benzylthiocarbamyl<br />

[BTC]-Asn and BTC-Ser co-eluted)<br />

and sample stability was only about 8 hours. The<br />

BTC derivatives appeared to provide superior resolution<br />

in the same system relative to PTC compounds,<br />

but the volatility of the reagent was similar to PITC.<br />

It would be surprising indeed if laboratories abandoned<br />

the established PITC reagent in favor of a new<br />

reagent to gain incremental advantages, particularly<br />

because the authors admit that analysis of cysteine<br />

and cystine is undependable without prior derivatization<br />

of thiols to stabilize them. Stark and Johansson<br />

discuss PITC analysis in the context of lipid-containing<br />

samples.<br />

Two chapters provide examples of electrochemical<br />

detection. One of these, by Jandik and colleagues,<br />

competently discusses integrated pulsed amperometric<br />

detection (IPAD) coupled to ion-exchange chromatrography,<br />

which is widely used for carbohydrate<br />

analysis. The balance of the chapters discuss techniques<br />

that diverge from the norm, either in instrumentation<br />

or sample source. Instrumentation and detection<br />

strategies include a quick, universal approach<br />

using Marfey’s reagent, the amperometric methods<br />

noted previously, capillary electrophoresis detection,<br />

precolumn and postcolumn OPA, and elegant flame<br />

photometry/gas chromatography techniques (used for<br />

analysis of O-phospho-amino acids and thiol-amino<br />

acids). Among this last group of chapters, analysis of<br />

blood plasma, foods, and modified amino acids are<br />

the predominent subjects. The final chapter on protein<br />

glycation products introduces mass spectrometry<br />

as the detection device and provides a concise introduction<br />

to this side reaction.<br />

Although the chapters present disparate approaches<br />

to amino acid analysis, editorial consistency<br />

is maintained through the use of extensive footnotes<br />

that, in many cases, provide useful general information.<br />

JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000 177


BOOK REVIEWS<br />

It may be in these details that the book will serve the<br />

widest audience by suggesting specific reagents and<br />

protocols. Generally, more specialized topics have<br />

been emphasized in this volume; readers seeking background<br />

or introductory information will find copious<br />

references to earlier literature. Conversely, those con-<br />

178 JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000<br />

cerned with problematic amino acid analyses, such as<br />

complex biologic samples or posttranslational modifications,<br />

will want to mine this volume for helpful<br />

insights and sample-handling tips.<br />

Reviewed by Alan Mahrenholz


MESSAGE <strong>FR</strong>OM THE<br />

EXECUTIVE BOARD<br />

<strong>AB</strong>RF Annual Meetings<br />

<strong>AB</strong>RF2001, to be held February 24 through 27 in San<br />

Diego, California, is fast approaching. Paul Tempst<br />

and David Schwartz have put together an exciting<br />

program. Plenary speakers include Ronald Evans,<br />

Roger Brent, Andrew Marks, Allan Wolffe, and Roger<br />

Kornberg. Considerable interest has been expressed<br />

in John Quakenbush’s pre-meeting course on microarray<br />

analyses. The <strong>AB</strong>RF Award Recipient this year is<br />

Csabo Horvath. He is being recognized for his accomplishments<br />

in the development of High-Pressure<br />

Liquid Chromatography, which is essential for many<br />

of our technologies. Mark your calendars for <strong>AB</strong>RF<br />

2002, to be held March 9 through 12 in Austin, Texas.<br />

Plans are being made to incorporate proteomics as a<br />

major theme.<br />

Research Group Studies<br />

The Research Group Guidelines are being revised for<br />

presentation at the Research Group Chairs–Executive<br />

Board meeting at <strong>AB</strong>RF 2002. The following is a suggested<br />

timeline:<br />

January<br />

Recruit new members for the committee,<br />

Executive Board approval of new members<br />

<strong>AB</strong>RF Meeting<br />

Old and prospective new members meet at<br />

the conference<br />

Poster presentation<br />

Final manuscript due<br />

March<br />

Elect new chair for group.<br />

June 1<br />

New research project defined and submitted to<br />

the Executive Board (may be a 2-year study,<br />

but must be in proposal)<br />

NEWS<br />

EVENTS<br />

June/July<br />

Project details finalized<br />

September 1<br />

Study disseminated<br />

November 1<br />

Data in<br />

November–January<br />

Data analyzed and manuscript drafted; poster<br />

should be derived from manuscript<br />

It has also been suggested that the following<br />

points be included: that the September issue of JBT be<br />

dedicated to Research Group studies, that members<br />

be recruited based on writing skills as well as technical<br />

skills, that studies be designed for introduction of<br />

new techniques and assessment of a laboratory performance,<br />

and that more challenging studies be<br />

designed for those who have the appropriate facilities<br />

and equipment. The focus of the latter group would<br />

be on cutting-edge technology. The Executive Board<br />

welcomes additional comments and suggestions for<br />

the Research Group Guidelines.<br />

JBT and HighWire<br />

&<br />

Ralph Bradshaw, chair of the Publications Committee,<br />

has been instrumental in promoting JBT online<br />

through HighWire. HighWire was recommended<br />

because of their vast stable of journals, including the<br />

Journal of Biological Chemistry, among others. <strong>AB</strong>RF<br />

members will be able to access HighWire through a<br />

hyperlink on our Web Site. <strong>AB</strong>RF members will also<br />

be able to view abstracts or contents of all other High-<br />

Wire journals. Members will use a password for<br />

access to the articles in JBT. JBT articles will be available<br />

online prior to print publication, allowing faster<br />

dissemination of information.<br />

New Editor-in-Chief Sought for JBT<br />

Journal of Biomolecular Techniques<br />

11:179–180 © 2000 <strong>AB</strong>RF<br />

RF <strong>AB</strong><br />

Clayton Naeve has announced his resignation as<br />

Editor-in-Chief of JBT. Clayton’s enthusiasm and drive<br />

have been responsible for the establishment of JBT as<br />

a new vehicle for the growth of knowledge in the<br />

JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000 179


NEWS & EVENTS<br />

field of biotechnology and as a mouthpiece for <strong>AB</strong>RF<br />

in the larger scientific community. He will be keenly<br />

missed by all associated with the Journal. Both Lynda<br />

Bonewald, retiring President of <strong>AB</strong>RF, and Mark<br />

Lively, incoming President, have expressed their gratitude<br />

to Clayton for services to the Society. The Publications<br />

Committee, chaired by Ralph Bradshaw, is<br />

seeking nominations from the membership of <strong>AB</strong>RF<br />

for a new Editor-in-Chief, and asks that these be sent<br />

in writing to Ralph at rablab@uci.edu.<br />

Dave Speicher will join Clive Slaughter as the<br />

newest Associate Editor for JBT. He and Clive will be<br />

reorganizing the review process and restructuring the<br />

editorial board. <strong>AB</strong>RF members can support JBT by<br />

submitting their best work to our society’s journal.<br />

Often, articles published in the field of biomolecular<br />

techniques present “results” but not detailed methods.<br />

<strong>AB</strong>RF members are encouraged to publish articles<br />

providing detailed techniques in JBT to be referenced<br />

in other articles. JBT will also publish Research Group<br />

reports targeted to <strong>AB</strong>RF members and including current<br />

and future technologies.<br />

<strong>AB</strong>RF Web Page<br />

Ted Thannhauser, chair of the Web Site Committee,<br />

has presented a plan to secure bids from commercial<br />

companies to update our Web Site. A list of desired<br />

features for the redesigned <strong>AB</strong>RF Web Page includes<br />

the following: the Web Site should serve as a window<br />

to other resources; Research Groups should be able to<br />

do online updates; there should be hyperlinks to corporate<br />

sponsors’ sites; and laboratories should be able<br />

to do online updates of their entries in the “yellow<br />

pages.” The option of banner advertising to generate<br />

revenue will be explored.<br />

New Board Members<br />

It is with regret that we bid farewell to John Stults and<br />

Karen DeJongh as they rotate off of the Executive<br />

Board as of January 1, 2001. John has provided insight<br />

and wisdom concerning <strong>AB</strong>RF operations in his usual<br />

quiet yet effective manner. Karen has been absolutely<br />

invaluable as treasurer of the organization and leaves<br />

this position with <strong>AB</strong>RF in good financial shape. Laurey<br />

Steinke will assume the Treasurer’s position, and<br />

Mark Lively will become President. We are looking<br />

180 JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000<br />

forward to the participation of our newest board<br />

members, Ted Thannhauser and Preston Hensley.<br />

<strong>AB</strong>RF/ASBMB JOINT SYMPOSIUM<br />

The <strong>AB</strong>RF/American Society of Biochemistry and Molecular<br />

Biology (ASBMB) Joint Symposium will be<br />

presented at the 2001 ASBMB Annual and Satellite<br />

Meetings held in conjunction with Experimental Biology<br />

2001, to be held March 31 through April 4 in<br />

Orlando, Florida. This is a continuing series sponsored<br />

by both <strong>AB</strong>RF and ASBMB. The goal of these<br />

joint symposia is to bring emerging technologies to<br />

biochemists and molecular biologists, who can then<br />

use the new techniques in their research programs.<br />

“High-Throughput Genomic Technologies: Decisions<br />

Based on <strong>Science</strong> and Reality” is scheduled for<br />

9:45 A.M. on Wednesday, April 4. Michael R. Sussman,<br />

Director of University of Wisconsin Biotechnology<br />

Center, and Ronald L. Niece, Research Resources and<br />

Technologies, organized the symposium, which covers<br />

several different applications of developing technologies.<br />

The symposium includes an introduction<br />

highlighting the role of resource laboratories in contemporary<br />

research and illustrating how the instrumentation<br />

and experience of the core laboratory can<br />

expedite research programs. Dr. Sussman will discuss<br />

progress in the development of saturation<br />

reverse genetics using knockout plants and will<br />

describe a maskless array synthesizer for producing<br />

high-density DNA oligonucleotide arrays “on the fly.”<br />

Hundreds of thousands of lines can be screened for<br />

knockout plants for any gene of interest. Combinatorial<br />

chemistry comes to the laboratory benchtop in<br />

the form of high-density oligonucleotide arrays. Dr.<br />

Smith addresses the problem of complexity in mass<br />

spectral data. His laboratory’s approach is to reduce<br />

complex spectra by controlling the charge state of the<br />

ion to a small number. Using electrospray ionization<br />

and an orthogonal time-of-flight mass spectrometer,<br />

the aerosol is exposed to a bipolar ionizing gas. In<br />

mixtures of proteins or nucleic acids, spectra are simplified<br />

and chemical noise minimized.


AMINO ACID COMPOSITION AND<br />

SEQUENCE ANALYSIS<br />

Sechi S, Chait BT.A method to define the carboxy terminal<br />

of proteins. Anal Chem 2000;72:3374–3378.<br />

Proteins are first digested with lysylendopeptidease.<br />

Anydrotrypsin, a catalytically inert derivative of<br />

trypsin in which serine-195 is converted to dehydroalanine,<br />

is used to bind peptides containing a<br />

lysine (or arginine) residue at their C-termini. The<br />

peptide derived from the C-terminus of the intact protein,<br />

lacking a C-terminal lysine or arginine, is not<br />

retained by anhydrotrypsin and is collected for subsequent<br />

analysis. Matrix-assisted laser desorption/ionization<br />

(MALDI) is used to identify the peptide, and<br />

tandem mass spectrometry is used to sequence it.<br />

CARBOHYDRATES AND<br />

GLYCOPROTEINS<br />

Charlwood J, Skehel JM, Camilleri P.Analysis of N-linked<br />

oligosaccharides released from glycoproteins separated<br />

by two-dimensional gel electrophoresis. Anal Biochem<br />

2000;284:49–59.<br />

Protocols are described for analysis of N-linked<br />

carbohydrates on glycoproteins that have been sub-<br />

ARTICLE<br />

WATCH<br />

This column highlights recently published articles that are of interest to the<br />

readership of this publication. We encourage <strong>AB</strong>RF members to forward<br />

information on articles they feel are important and useful to Clive A. Slaughter,<br />

St. Jude Children’s Research Hospital, 332 North Lauderdale Street, Memphis,<br />

TN 38105-2794; Tel: (901) 495-4844; Fax: (901) 495-2945; email: clive.slaughter@<br />

stjude.org; or to any member of the editorial board. Article summaries reflect the<br />

reviewers’ opinions and not necessarily those of the Association.<br />

jected to two-dimensional gel electrophoresis. Glycans<br />

are released from the proteins by digestion with<br />

protein N-deglycosidase F (PNGase F), derivatized<br />

with 3-acetamido-6-aminoacridine, and analyzed by<br />

matrix-assisted laser desorption/ionization (MALDI)–<br />

time-of-flight mass spectrometry. Enzymic release of<br />

the glycan is demonstrated using in-gel digestion of<br />

the excised gel spot, with or without prior tryptic<br />

digestion, and also using glycoproteins electroblotted<br />

to polyvinylidene difluoride membranes.<br />

PHOSPHOLIPIDS<br />

Journal of Biomolecular Techniques<br />

11:181–184 © 2000 <strong>AB</strong>RF<br />

RF <strong>AB</strong><br />

Taguchi R, Hayakawa J,Takeguchi Y, Ishida M.Two-dimensional<br />

analysis of phospholipids by capillary liquid chromatography/electrospray<br />

ionization mass spectrometry.<br />

J Mass Spectrom 2000;35:953–966.<br />

A strategy for analysis of phospholipids extracted<br />

from cultured cells is described. The strategy uses<br />

capillary liquid chromatography combined with tandem<br />

mass spectrometry in a quadrupole mass analyzer.<br />

More than 500 species of phospholipids are<br />

included by the method. Positive molecular ions,<br />

negative molecular ions, positive fragment ions, and<br />

negative fragment ions are monitored, consecutively,<br />

in 11-second cycles. Positive molecular ions indicate<br />

choline-containing phospholipids such as phosphatidylcholine,<br />

sphingomyelin, lysophosphatidylcholine,<br />

and phosphatidylethanolamine. Negative<br />

JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000 181


ARTICLE WATCH<br />

molecular ions indicate acidic phospholipids such as<br />

phosphatidylinositol. Positive fragment ions indicate<br />

diradylglycerol and derivatives of 1-alkyl and 2alkenyl<br />

cyclic phosphatidic acid from phosphatidylethanolamine<br />

and phosphorylcholine from cholinecontaining<br />

phospholipids. Negative fragment ions<br />

indicate ions from the acyl fatty acid and phosphorylcholine,<br />

phosphorylethanolamine, and cyclic inositol<br />

phosphate.<br />

NUCLEIC ACIDS<br />

Premstaller A, Oberacher H, Huber CG. High-performance<br />

liquid chromatography-electrospray ionization<br />

mass spectrometry of single- and double-stranded<br />

nucleic acids using monolithic capillary columns. Anal<br />

Chem 2000;72:4386–4393.<br />

High-performance liquid chromatography (HPLC)<br />

columns are most commonly filled with microparticulate<br />

stationary phases, but these suffer from the relatively<br />

large void volumes between the packed particles<br />

and the slow diffusion of solutes into and out of<br />

the stagnant mobile phase present in the pores of the<br />

separation medium. These problems are alleviated by<br />

using stationary phases consisting of a continuous<br />

rod of rigid, porous polymer with no interstitial volume<br />

and only internal porosity. This report uses such<br />

“monolithic” stationary phases, packed into capillary<br />

columns, to improve the separation of oligonucleotides.<br />

A mobile phase using 100-mM triethylammonium<br />

acetate as an ion pairing reagent is employed,<br />

and elution is performed with a gradient of acetonitrile.<br />

The method is adapted for online electrospray<br />

mass spectrometry by using 25-mM triethylammonium<br />

bicarbonate as the ion pairing reagent, with only a<br />

small reduction in chromatographic performance.<br />

Acetonitrile is supplied postcolumn to enhance the<br />

elecrospray process. Good-quality mass spectra of<br />

femtomole amounts or 3-mers to 80-mers are presented<br />

showing little cation adduction. Doublestranded<br />

DNA fragments from 51 to 587 base pairs<br />

are separated and detected.<br />

Xu G, Enderle H, Liebich H, Lu P. Study of normal and<br />

modified nucleosides in serum by RP-HPLC. Chromatographia<br />

2000;52:152–158.<br />

A high-performance liquid chromatography<br />

(HPLC) method is employed for separating nucleoside<br />

mixtures containing both modified and unmodified<br />

components that are present in serum and urine of<br />

182 JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000<br />

normal individuals and cancer patients. The method<br />

uses a KH 2 PO 4 buffer (pH 4.65), and elution is performed<br />

with a 45-minute period of a gradient of<br />

methanol. The nucleosides are detected by their absorption<br />

at 260 and 280 nm.<br />

Puapaiboon U, Jai-nhuknan J, Cowan JA. Rapid and direct<br />

sequencing of double-stranded DNA using exonuclease<br />

III and MALDI-TOF MS. Anal Chem 2000;72:3338–3341.<br />

A strategy for sequencing double-stranded DNA<br />

by matrix-assisted laser desorption/ionization (MALDI)<br />

mass spectrometry is reported. The strategy uses<br />

exonuclease III to degrade phosphodiester bonds<br />

from the 3�-end of the double-stranded DNA. The<br />

enzyme requires the addition of appropriate divalent<br />

cations. A cation exchange column is then used in<br />

conjunction with an ammonium citrate additive to<br />

reduce formation of alkali salt adducts.<br />

MASS SPECTROMETRY<br />

Timperman AT,Aebersold R. Peptide electroextraction<br />

for direct coupling of n-gel digests with capillary LC-<br />

MS/MS for protein identification and sequencing. Anal<br />

Chem 2000;72:4115–4121.<br />

An electrophoretic method for extracting proteins<br />

from in-gel digests is described for use in mass spectrometric<br />

protein identification. A small-scale cell is<br />

fabricated for electroextraction, and peptides are<br />

trapped on a strong cation-exchange resin housed in<br />

a fused silica capillary tube. Immobilized in this way,<br />

they can be stored for several days without loss. The<br />

design of the system is suitable for direct interfacing<br />

with capillary liquid chromatography in tandem liquid<br />

chromatography/mass spectrometry analyses. Significant<br />

enhancements in sensitivity are realized for<br />

digests of proteins at the low nanogram level (ie, the<br />

limits of sensitivity of silver staining).<br />

Doucette A, Craft D, Li L. Protein concentration and<br />

enzyme digestion on microbeads for MALDI-TOF peptide<br />

mass mapping of proteins from dilute solutions.<br />

Anal Chem 2000;72:3355–3362.<br />

A method for digesting proteins available in solutions<br />

of low concentration is presented for the acquisition<br />

of peptide mass fingerprint data. The protein<br />

is first concentrated by adsorption to POROS R2<br />

reverse-phase polymeric beads (Applied Biosystems,<br />

Foster City, CA). The beads may then be washed to


emove contaminants. The bound proteins are subsequently<br />

digested with trypsin. The beads retain peptides<br />

during digestion and may be deposited directly<br />

on a target for matrix assisted laser desorption/ionization<br />

(MALDI). On-target washing may also be performed<br />

to remove substances inhibiting ionization.<br />

The method provides high-quality peptide mass fingerprint<br />

data for protein samples of only 100 nM.<br />

PROTEINS—PURIFICATION<br />

AND CHARACTERIZATION<br />

Zuo X, Speicher DW. Quantitative evaluation of protein<br />

recoveries in two-dimensional electrophoresis with<br />

immobilized pH gradients. Electrophoresis 2000;21:3035–<br />

3047.<br />

Protein recoveries are systematically evaluated at<br />

the various steps of separation by two-dimensional<br />

gel electrophoresis using immobilized pH gradients<br />

(IPGs) for the isoelectric focusing stage. The Multiphor<br />

and IPGphor systems from Amersham Pharmacia<br />

Biotech (Uppsala, Sweden) are used for the separation<br />

of metabolically radiolabeled proteins from<br />

Escherichia coli. With the Multiphor system, in which<br />

sample application to the IPG strips and subsequent<br />

electrofocusing are performed in separate vessels,<br />

sample application from cups gives better protein<br />

recoveries than sample loading by IPG rehydration. In<br />

the IPGphor system, in which sample loading and<br />

electrofocusing are performed in the same vessel, the<br />

rehydration method gives better recoveries. In all<br />

cases, carrier ampholytes in the sample buffer increase<br />

protein recoveries. Thiourea does not affect<br />

protein recoveries, but does improve protein resolution<br />

during electrofocusing.<br />

Zuo X, Speicher DW. A method for global analysis of<br />

complex proteomes using sample prefractionation by<br />

solution isoelectofocusing prior to two-dimensional<br />

electrophoresis. Anal Biochem 2000;284:266–278.<br />

Increasing the protein load on immobilized pH<br />

gradient (IPG) strips in an attempt to improve the<br />

number of visible spots resolved by two-dimensional<br />

electrophoresis is limited by the tendency of proteins<br />

to aggregate or precipitate when high protein loads<br />

are used, causing smearing in the isoelectric focusing<br />

dimension. The use of solution-phase isoelectric focusing<br />

to minimize such smearing is discussed, and an<br />

isoelectric focusing apparatus for prefractionating proteins<br />

for this purpose is described.<br />

ARTICLE WATCH<br />

Gygi SP, Corthals GL, Zhang Y, Rochon Y, Aebersold R.<br />

Evaluation of two-dimensional gel electrophoresis-based<br />

proteome analysis technology. Proc Natl Acad Sci USA<br />

2000;97:9390–9395<br />

It is widely assumed that two-dimensional gel<br />

electrophoresis, coupled with mass spectrometric<br />

identification of spots, enables a broad coverage of<br />

the proteome. Consistent with this view is the observation<br />

that the number of spots observed by twodimensional<br />

electrophoresis approaches the number<br />

of proteins predicted for a given organism or tissue.<br />

This study examines an arbitrarily selected section<br />

from a narrow pH range (4.9–5.7) two-dimensional<br />

display of yeast proteins in which more than 1500<br />

spots are visualized by silver staining. Fifty spots were<br />

digested with trypsin and analyzed by liquid chromatography<br />

combined with tandem mass spectrometry,<br />

and a total of 39 different proteins were identified<br />

from a region where the yeast genome is predicted to<br />

encode 57 gene products. However, these represent<br />

only the proteins predicted on the basis of codon<br />

usage to be the most abundant. Furthermore, only 14<br />

of the proteins identified were predicted to appear in<br />

the analyzed section. Additionally, single genes produced<br />

multiple spots, and the products of up to six<br />

genes were found to make up a single spot, presenting<br />

problems for both quantitative protein expression<br />

comparisons and database-matching studies. It is concluded<br />

that the method is unsuited for the analysis of<br />

low-abundance proteins and that statements about<br />

the feasibility and straightforwardness of proteome<br />

analysis based on the two-dimensional electrophoresis<br />

mass spectrometry strategy should be rethought.<br />

[This article has also received comment by Smith RD.<br />

Probing proteomics—seeing the whole picture. Nat<br />

Biotechnol 2000;18:1041–1042.]<br />

Ghaemmaghami S, Fitzgerald MC, Oas TG. A quantitative,<br />

high-throughput screen for protein stability. Proc<br />

Natl Acad Sci USA 2000;97:8296–8301<br />

The use of mass spectrometric measurements of<br />

hydrogen/deuterium exchange to compare the rates of<br />

denaturation of different but related proteins is<br />

described. Matrix-assisted laser desorption/ionization<br />

(MALDI) is employed to measure the rates of exchange<br />

of amide hydrogen atoms with deuterium in<br />

solution as a function of denaturant concentration to<br />

provide a measure of protein stability. The method<br />

permits large numbers of protein samples to be<br />

screened in a short time, does not demand that the<br />

proteins of interest are pure or available in large quantities,<br />

and is forgiving of the presence of denaturants.<br />

Mutants of the � repressor are studied to illustrate the<br />

JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000 183


ARTICLE WATCH<br />

utility of the method, and the stabilization of maltosebinding<br />

protein on binding to maltose is also demonstrated.<br />

MICROARRAYS<br />

Lee MT, Kuo FC,Whitmore GA, Sklar J. Importance of<br />

replication in microarray gene expression studies: statistical<br />

methods and evidence from repetitive cDNA<br />

hybridizations. Proc Natl Acad Sci USA 2000;97:9834–<br />

9839<br />

This study investigates the inherent variability in<br />

gene expression data and assess the extent to which<br />

replication in an experiment produces more consistent<br />

and reliable findings. The results show that any<br />

single microarray output is subject to substantial variability,<br />

even though, by design, variability as a result<br />

of multiple preparations of probe, arrays on different<br />

slides, or arrays generated at different times is not<br />

admitted. A single output yields numerous misclassifications.<br />

Replications are not consistent and therefore<br />

produce different lists of expressed genes. Modeling<br />

the random variation in gene expression indicates<br />

that the probability that mRNA in the sample tissue<br />

184 JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000<br />

either fails to be represented as probe or fails to<br />

hybridize to the cDNAs on the slide may be as large<br />

as 5% (false-negatives). The probability that ghost<br />

genes are expressed may be as large as 10% (falsepositives).<br />

When microarray data from several replicates<br />

are combined, however, a more accurate picture<br />

of gene expression is produced. It is recommended<br />

that at least three replicates be included in the design<br />

of experiments using cDNA microarrays.<br />

BIOINFORMATICS<br />

Fenyö D. Identifying the proteome: software tools. Curr<br />

Opin Biotechnol 2000;11:391–395.<br />

This article reviews the various software tools that<br />

are available on the internet for searching protein<br />

sequence databases using mass spectral information.<br />

Both peptide mapping data listing the mass values of<br />

proteolytic peptides and fragment ion data derived by<br />

collisional dissociation of peptides to yield sequenceinformative<br />

ions are included. Attention is paid to the<br />

methods used by the various tools to rank protein<br />

candidates and the methods by which the quality of<br />

identifications are assessed.


2001<br />

DATES TO<br />

REMEMBER<br />

Jan 13–18 HPCE 2001, 14th International Symposium<br />

on Microscale Separations and Analysis,<br />

Boston, MA. Contact: Joan Oefner, Symposium<br />

Manager, Rhema Association Management,<br />

156 South Spruce Avenue, Suite 207A, South<br />

San Francisco, CA 94080-4556; Tel: (650)<br />

876-0792; Fax: (650) 876-0793 (email:<br />

joefner@casss.org).<br />

Jan 29–30 IBC’s Process Validation for Biologicals,<br />

San Diego, CA. Contact: Jim Prudhomme, Lifesciences<br />

Marketing Division, IBC USA Conferences,<br />

Inc., 1 Research Drive, Suite 400A,<br />

Southborough, MA 01581. Tel: (508) 616-5550<br />

X205; Fax: (508) 616-5533 (email:<br />

jprudhomme@ibcusa.com; URL:<br />

http://www.ibcusa.com/2532).<br />

Feb 8–9 IBC’s 9th Annual Conference on Alzheimer’s,<br />

Atlanta, GA. Contact: Julie Anderson,<br />

Marketing Manager—Life <strong>Science</strong>s, IBC USA<br />

Conferences, Inc., 1 Research Drive, Suite 400A,<br />

Westborough, MA 01581. Tel: (508) 616-5550<br />

X423; Fax: (508) 616-5533 (email:<br />

janderson@ibcusa.com; URL:<br />

www.ibcusa.com/2579).<br />

UPCOMING<br />

EVENTS<br />

<strong>AB</strong>RF members and Corporate Sponsors are encouraged to make announcements<br />

about pertinent meetings, workshops, etc. that are beneficial to our<br />

members and open to the public. Send items to be listed to Daniel J.<br />

Strydom, BioNebraska, Inc., 3820 NW 46th St., Lincoln, NE 68524-1637 (Tel:<br />

402-470-2100; Fax: 402-470-2345; Email: strydom@inetnebr.com).<br />

Journal of Biomolecular Techniques<br />

11:185–186 © 2000 <strong>AB</strong>RF<br />

RF <strong>AB</strong><br />

Feb 15–16 IBC’s Chemo * Bio Informatics,<br />

San Diego, CA. Contact: Jim Prudhomme,<br />

Lifesciences Marketing Division, IBC USA Conferences,<br />

Inc., 1 Research Drive, Suite 400A,<br />

Southborough, MA 01581. Tel: (508) 616-5550<br />

X205; Fax: (508) 616-5533 (email:<br />

jprodhomme@ibcusa.com; URL:<br />

http://www.ibcusa.com/2621).<br />

Feb 20–23 WCBP 2001, 5th Symposium on the<br />

Interface of Regulatory and Analytical <strong>Science</strong>s<br />

for Biotechnology Products, Washington, DC.<br />

Contact: Joan Oefner, Symposium Manager,<br />

Rhema Association Management, 156 South<br />

Spruce Avenue, Suite 207A, South San Francisco,<br />

CA 94080-4556; Tel: (650) 876-0792;<br />

Fax: (650) 876-0793 (email: joefner@casss.org;<br />

URL: http://www.casss.org/wcbp).<br />

Feb 24–27 <strong>AB</strong>RF 2001: The New Biology: Technologies<br />

for Resolving Macromolecular Communications,<br />

San Diego, CA. Contact: <strong>AB</strong>RF Meeting<br />

Management, FASEB, Office of Scientific<br />

Meetings, 9650 Rockville Pike, Bethesda, MD<br />

20814. Tel: (301) 530-7010; Fax: (301)<br />

530-7014 (email: marcella@faseb.org; URL:<br />

http://www.faseb.org/meetings/abrf2001/).<br />

Feb 26–28 IBC’s Enzyme Technologies 2001,<br />

San Diego, CA. Contact: Jim Prudhomme,<br />

Lifesciences Marketing Division, IBC USA<br />

Conferences, Inc., 1 Research Drive, Suite 400A,<br />

Southborough, MA 01581. Tel: (508) 616-5550<br />

X205; Fax: (508) 616-5533 (email:<br />

JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000 185


UPCOMING EVENTS<br />

jprodhomme@ibcusa.com; URL:<br />

http://www.ibcusa.com/2573).<br />

Mar 6–8 IBC’s Formulation Strategies for Biopharmaceuticals,<br />

San Diego, CA. Contact: Julie<br />

Anderson, Marketing Manager—Life <strong>Science</strong>s,<br />

IBC USA Conferences, Inc., 1 Research Drive,<br />

Suite 400A, Westborough, MA 01581. Tel:<br />

(508) 616-5550 X423; Fax: (508) 616-5533<br />

(email: janderson@ibcusa.com; URL:<br />

http://www.ibcusa.com/2624).<br />

Mar 12–16 IBC’s ScreenTech 2001, San Diego, CA.<br />

Contact: Jim Prudhomme, Lifesciences Marketing<br />

Division, IBC USA Conferences, Inc.,<br />

1 Research Drive, Suite 400A, Southborough,<br />

MA 01581. Tel: (508) 616-5550 X205; Fax: (508)<br />

186 JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000<br />

616-5533 (email: jprodhomme@ibcusa.com;<br />

URL: http://www.ibcusa.com/screentech).<br />

Apr 4–6 Conservation Genetics Symposium,<br />

New York, NY. Contact: Fiona Brady, Center<br />

for Biodiversity and Conservation, American<br />

Museum of Natural History, Central Park West<br />

at 79th Street, New York, NY 10024; Tel:<br />

(212) 769-5742; Fax: (212) 769-5292<br />

(email: brady@amnh.org; URL:<br />

http://research.amnh.org/biodiversity/<br />

conservation-genetics.html).<br />

Jun 9–14 17th American Peptide Symposium,<br />

San Diego, CA. Contact: Doris Rice. Tel:<br />

(858) 455-4752 (email: drice@tpims.org;<br />

URL: http://www.5z.com/aps/).


<strong>AB</strong>RF 2001<br />

THE NEW BIOLOGY<br />

Technologies for Resolving<br />

Macromolecular Communications<br />

An International Symposium Sponsored by<br />

The Association of Biomolecular Resource Facilities<br />

Town & Country Resort<br />

San Diego, California<br />

February 24–27, 2001<br />

RF <strong>AB</strong><br />

Research•Technology<br />

Communication•Education<br />

<strong>AB</strong>STRACTS


<strong>AB</strong>RF 2001 <strong>AB</strong>STRACTS<br />

P1-S<br />

Compatibility of the RapXtract Dye Terminator Removal Kit with<br />

purification of sequencing reactions for the applied biosystems<br />

Prism 3100 Capillary Sequencer.<br />

A.L. Springer, L.R. Booth, K.A. Hughes, R.J. Kaiser, D.A. Spicer;<br />

Prolinx, Inc., 22322 20th Avenue SE, Bothell, WA 98021<br />

Successful automated DNA sequence analysis requires that sequencing reactions<br />

be purified of excess unincorporated dye terminators. Common methods<br />

for purifying sequencing reactions involve several steps and require<br />

processes that are not easily automated. Prolinx, ® Inc. has developed the<br />

unique RapXtract Dye Terminator Removal Kit for this application that is<br />

rapid, easy to perform, yields high quality product and does not require modified<br />

primers. The RapXtract kit uses a magnetic particle format to remove<br />

dye terminator contaminants by mass action. The protocol has three steps:<br />

1. Remove storage buffer from particles, 2. Add sequencing reactions and<br />

mix, and 3. Remove purified extension products. The <strong>AB</strong>I PRISM 3100<br />

Genetic Analyzer is a 16-capillary instrument that utilizes technology from<br />

other <strong>AB</strong>I capillary sequencers in a medium-throughput format. RapXtractpurified<br />

samples analyzed on the <strong>AB</strong>I PRISM 3100 yielded sequences with<br />

good Phred quality scores and signal strength. The RapXtract kit has been<br />

optimized to minimize the use of expensive sequencing reagents and can be<br />

performed on reactions containing as little as 1 �l of Big Dye Reaction Mix<br />

and as little as 5 �l total volume. The RapXtract kit has been used successfully<br />

with double-stranded plasmid, PCR product or single-stranded DNA<br />

templates. RapXtract purification can also be used for dye primer reactions.<br />

The ease of use and versatility of the RapXtract kit make it a good choice for<br />

manual or automated purification of sequencing reactions.<br />

P3-T<br />

Improved signal strength on <strong>AB</strong>I Prism 377 DNA Sequencer and<br />

310 Genetic Analyzer by using a new sequencing run module.<br />

S-M. Chen, H. Zielke, G. Amparo, S. Spurgeon; Applied Biosystems,<br />

850 Lincoln Centre Dr., Foster City, CA 94404<br />

In many sequencing labs DNA sample template quality varies greatly due to<br />

the diverse sources of these samples and the different DNA preparation<br />

methods used. This variable DNA template quality results in variable sequencing<br />

data quality. Many low quality data are associated with low quality<br />

or quantity of DNA templates. This frequently results in low signal strength<br />

and low signal to noise ratio. A new sequencing module was developed for<br />

the <strong>AB</strong>I PRISM 377 and 310 systems that will generate greater signal strength<br />

during DATA collection. With the use of this module in data collection we<br />

were able to improve signal to noise ratio and were able to improve success<br />

rate in data analysis.<br />

POSTER <strong>AB</strong>STRACTS<br />

188 JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000<br />

P2-M<br />

Serial analysis of gene expression using the Applied Biosystems<br />

capillary electrophoresis platforms.<br />

K.M. Gunning, B. Nutter, A. Swei, J. Zon; Applied Biosystems,<br />

850 Lincoln Centre Dr., Foster City, CA 94404<br />

In today’s genomic environment, the draft completion of the human genome<br />

has allowed researchers access to a wealth of information with respect to<br />

gene identity and gene expression profiles. Scientists have developed novel<br />

ways in which to elucidate this information and SAGE is deemed one of the<br />

more comprehensive methods available for rapid, detailed analysis of large<br />

numbers of cellular transcripts. To further enhance the rapidity at which one<br />

can determine this global gene expression profile, one needs more efficient<br />

and precise methods for analyzing these transcripts and for assessing their<br />

abundance. Automated capillary electrophoresis provides a platform whereby<br />

the sequence of these many transcripts can be determined with speed and<br />

precision. The <strong>AB</strong>I PRISM. ® 3700 and the <strong>AB</strong>I PRISM. ® 3100, the most recent<br />

addition to the CE family of platforms, provide the SAGE researcher with a<br />

means by which they can determine quickly and reliably, the transcript<br />

information necessary to build these gene expression profiles. These platforms<br />

partnered with the uniformity of signal strength and length of read of<br />

BigDye Terminators, are the labor saving answer to large scale transcript<br />

analysis using the SAGE method. We will demonstrate the ease of use and<br />

flexibility of these platforms as a preferred method for the sequence analysis<br />

of ditags in the SAGE process.<br />

P4-S<br />

Two steps cycle-sequencing improves base ambiguities and signal<br />

dropouts in certain BigDye DNA sequencing reactions.<br />

L. Wen; San Diego State Univ., 5500 Campanile Drive, San Diego,<br />

CA 92182-4614<br />

The use of automated fluorescent DNA sequencer systems and PCR-based<br />

DNA sequencing methods play an important role in the actual effort to<br />

improve the efficiency of large-scale DNA analysis. While dideoxy-terminators<br />

labeled with energy-transfer dyes (BigDye; PE/<strong>AB</strong>I) provide the most<br />

versatile method of automated DNA sequencing, premature terminations<br />

results in a substantially reduced reading length of the DNA sequence. Premature<br />

terminations are usually evidenced by base ambiguities and are often<br />

accompanied by diminished signal intensity after that point. I studied a twostep<br />

protocol for Taq cycle sequencing using the <strong>AB</strong>I BigDye terminator for<br />

reducing premature terminations in DNA sequences. I demonstrated that<br />

combining the annealing step with the extension step at one temperature<br />

(60�C) reduces premature terminations in DNA sequences that regularly contain<br />

premature terminations when the three temperature steps is used. This<br />

procedure can be significantly improved sequence quality in our core facility.<br />

Sequence results from initial studies are still forthcoming and will be presented.


P5-M<br />

Using high speed data collection on the <strong>AB</strong>I Prism 377XL<br />

to improve core facility DNA sequencing throughput.<br />

F. Lach, J. Medalle, M. Randesi, B. Imai; Rockefeller Univ.,<br />

1230 York Avenue, Box 105, New York, NY 10021<br />

The variety of samples subjected to a core facility presents a challenge when<br />

it comes to achieving high throughput DNA sequencing. Any given run can<br />

exhibit different degrees of success based on template type and size, template<br />

base content and arrangement, primer design, and overall sample quality.<br />

Although the <strong>AB</strong>I Prism 3700 DNA Analyzer excels in high throughput<br />

capabilities, the instrument still requires stricter sample management that is<br />

harder to achieve in the core facility environment. The <strong>AB</strong>I Prism 377XL continues<br />

to be an important resource since it requires less sample stringency<br />

than the 3700. Modification of the 377XL module parameters and new polyacrylamide<br />

premixes give the potential for increased sample throughput<br />

with minimal loss of resolution. This study will investigate the conditions<br />

required to obtain quality data using the 4X high speed data collection module<br />

on the 377XL. A data comparison will be made between the 2X standard<br />

speed and 4X high speed modules using various samples and polyacrylamide<br />

premixes.<br />

P7-S<br />

Automated transfer of DNA sequencing files to an ftp site.<br />

K.M. Ivanetich, R. Taylor, W. Yan, F. Sandifer, D. Wolber;<br />

UCSF, Box 0541, San Francisco, CA 94143<br />

The Organize Samples program has been developed to automate the renaming,<br />

consolidation, and transfer of sequencing data from Applied Biosystems<br />

373, 377 and 3700 DNA sequencers directly onto the facility’s ftp site. The<br />

program accesses (1) information in the Applied Biosystems sequencer’s<br />

analysis file, (2) the Applied Biosystems Tool Kit, (3) a file containing a list<br />

of user’s names paired with their laboratory’s principal investigator’s names,<br />

and (4) the compression program DropStuffit. Using the above, the program<br />

compresses all sequence files for one user into one document, names the<br />

document with the user’s name and the date, and places the compressed file<br />

into the appropriate laboratory’s folder on the facility’s ftp server. The laboratory’s<br />

folders are password protected. Users can access their laboratory’s<br />

folder, download the file, and uncompress it with a single click. The result<br />

is a set of uncompressed files, with one file for each sequence analysis file.<br />

The uncompressed files are named with the run and lane numbers or coordinate<br />

and capillary numbers plus the user’s template and primer names. Separate<br />

sequence text files are handled equivalently by Organize Samples, and<br />

are differentiated by a “.seq” suffix. Users can access the ftp site and download<br />

their DNA sequencing data from their own computer 24 hours/day–<br />

7 days/week.<br />

POSTER <strong>AB</strong>STRACTS<br />

<strong>AB</strong>RF 2001 <strong>AB</strong>STRACTS<br />

P6-T<br />

Update on the continuation of the DNA Sequencing Research Group<br />

2000 Study: an evaluation of methods used to sequence and isolate<br />

bacterial artificial chromosomes.<br />

T. Thannhauser1, L.S. Hall2, J. Hawes3, T. Hunter4, E. Jackson-Machelski5, K. Knudtson6, D. Leviten7, M.A. Robertson8; 1Cornell Univ., Rm 149<br />

Biotechnology Bldg, Ithaca, New York 14853-2703, 2Albert Einstein<br />

Col. of Med., 1695 Poplar Street, NewYork, NY 10464, 3Indiana Univ.<br />

Sch. of Med., 4Univ. of Vermont, 5Washington Univ. Sch. of Med.,<br />

6Univ. of Iowa, 7ICOS Corp., 8Univ. of Utah<br />

Recent emphasis on BACs as a primary source of template has created significant<br />

challenges for those who operate DNA sequencing facilities. Due to<br />

their large size (80–350 KB) BACs behave differently in sequencing reactions<br />

than do plasmid clones. To achieve optimal performance, standard sequencing<br />

reactions have to be modified. Moreover, due to their low copy number<br />

it is difficult to isolate enough BAC template using standard miniprep protocols.<br />

Using data submitted to part1 of the original DSRG 2000 BAC study,<br />

the DSRG extracted a recommended protocol for BAC sequencing and performed<br />

carefully controlled experiments in an internal study to test the efficiency<br />

and reliability of the protocol. This protocol was made available<br />

along with a standard BAC template and primer for testing in the community.<br />

Part 1 of the study examines the effect of a DSRG recommended sequencing<br />

protocol on the quality of the results obtained on a standard BAC template<br />

sent to participating laboratories. The sequencing results are analyzed<br />

for quality and length of read on the wide variety of instrumention used by<br />

the DNA sequencing community. The success rate of the standard protocol<br />

is compared to the internal study results and to the original results where<br />

applicable.<br />

Part 2 of the study continues to assess and evaluate the quality of BAC isolation<br />

methods commonly used in DNA sequencing facilities with the goal<br />

of providing a recommended isolation protocol. The isolated BAC DNA is<br />

sequenced using the DSRG recommended protocol. Results will be analyzed<br />

for quality and length of read on standard instrumentation to determine the<br />

success of the different BAC isolation procedures.<br />

P8-M<br />

Sequencing performance of the <strong>AB</strong>I Prism 3100 Genetic Analyzer.<br />

P.A. Baybayan, B. Johnson, K. Roy, R. Pingue, Q. Liwei; Applied<br />

Biosystems, 850 Lincoln Centre Drive, Foster City, CA 94404<br />

The new <strong>AB</strong>I Prism 3100 Genetic Analyzer is the latest addition to the suite<br />

of capillary electrophoresis platforms offered by Applied Biosystems. This 16capillary<br />

electrophoresis platform employs complete automation designed to<br />

meet the demands of mid to high-throughput sequencing and genotyping<br />

laboratories.<br />

The <strong>AB</strong>I Prism 3100 Genetic Analyzer offers flexibility by providing options<br />

for a variety of applications. In de novo sequencing where throughput, consistency<br />

and efficiency are important, several arrays (including 36 cm, 50 cm,<br />

and 80 cm) are available. These arrays require different run parameters and<br />

therefore, perform differently. The user will have the option to an array<br />

depending on their requirements (readlength and throughput). In comparative<br />

sequencing where accuracy and sensitivity are required for the detection<br />

and identification of polymorphisms, a 36-cm array is sufficient. Here, we will<br />

present the performance (readlength, accuracy, consistency) of the <strong>AB</strong>I Prism<br />

3100 Genetic Analyzer in the area of de novo and comparative sequencing<br />

and demonstrate how this instrument is the platform to best accommodate<br />

to your research needs.<br />

JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000 189


<strong>AB</strong>RF 2001 <strong>AB</strong>STRACTS<br />

P9-T<br />

A comparison of alternative methods for sequencing through<br />

difficult templates.<br />

D.A. Bintzler, Y. Song; Univ. of Cincinnati, 231 Bethesda Avenue,<br />

Cincinnati, OH 45267-0524<br />

Template DNA that has a seventy percent or greater content of guanosine<br />

and cytosine bases (GC rich) has been a potential problem for automated fluorescent<br />

DNA sequencing. Two well-known strategies for improving the base<br />

read length of the GC rich template include the addition of dimethyl sulfoxide<br />

(DMSO) or betaine to the PCR amplification of the template. Applied<br />

Biosystems Incorporated (<strong>AB</strong>I) has offered a third method with dGTP big dye<br />

chemistry. The dGTP chemistry has been significantly effective for extending<br />

through templates that contain a chain of ten or more guanosine bases.<br />

The dRhodamine chemistry (<strong>AB</strong>I) has also been effective for some problematic<br />

templates. However, no single method has been effective for all problematic<br />

templates in this facility. Therefore, research has continued to find an<br />

alternative method. A PCR additive available from Qiagen, Q buffer, was<br />

introduced as a potential method for improving the base read length of the<br />

GC rich template. Preliminary studies showed that Q buffer had improved<br />

PCR extension through difficult templates that were not successfully<br />

sequenced with the other methods previously mentioned. However, a thorough<br />

investigation of the Q buffer compared to DMSO, betaine, dRhodamine<br />

and dGTP big dye chemistry was required. The results of the investigation<br />

are reported here.<br />

P11-M<br />

Integration of new sequencing technologies into the W.M. Keck<br />

Biotechnology/HHMI Biopolymer Lab. at Yale.<br />

K.M. Hager; Yale Univ., 295 Congress Ave., New Haven, CT 06536<br />

In the past year, we have successfully integrated the Applied Biosystems 3700<br />

DNA Sequencer into our DNA sequencing service. This machine utilizes 96<br />

capillaries and a proprietary polymer rather than polyacrylamide slab gels to<br />

separate fluorescently-labeled DNA termination fragments. Because of 3700’s<br />

unattended robotic sample loading from as many as four sample plates per<br />

operator interaction, we have increased the lab’s overall sequencing throughput<br />

by 25% with no increase in staff and lowered the average sample turnaround<br />

time. In addition, the 3700’s automation has allowed us to initiate<br />

Genescan and SNAPSHOT single-nucleotide polymorphism (SNP) detection<br />

assays as new services. At present, about 75% of our total DNA sequencing<br />

samples are run on the 3700 with the remainder run on one 377 (slab gel).<br />

About 2–5% of the samples run on the 3700 give a poor separation and these<br />

samples are rerun on the 377. Such poor separations are due to either<br />

improper loading of the separation polymer into the capillary or overloading<br />

of the capillary with excessive amounts of DNA. On the vast majority of<br />

runs, our pgem standards yield 650–750 bases of sequence data at �99%<br />

accuracy (POP-6 polymer).<br />

Recently, we have begun an evaluation of Pyrosequencing, a non-electrophoretic<br />

DNA sequencing technology using an enzyme coupled assay<br />

where light is ultimately generated by luciferase from PPI released following<br />

nucleotide incorporation (<strong>Science</strong> 281, pp. 363–364 (1998)). Our initial applications<br />

for Pyrosequencing will include sequence tag (20–40 base) determination<br />

and SNP detection.<br />

POSTER <strong>AB</strong>STRACTS<br />

190 JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000<br />

P10-S<br />

Fragment and sequence analysis in Portugal: a survey of facilities<br />

and applications.<br />

L.M. Souto; Univ. de Aveiro, Portugal, Departamento de Biologia,<br />

Universidade de Aveiro, Aveiro, Aveiro 3810 Portugal<br />

Since 1992, with the first unit of Automated Sequencers starting to work in<br />

Portugal, a significant improvement in the number of equipments and institutions<br />

dealing with sequencing and fragment analisys has been registered.<br />

The leading groups are the forensic institutes which deal with a heavy routine<br />

work, namely in paternity cases. These groups are using very standardised<br />

techniques and markers, in accordance to the EDNAP (European DNA<br />

Profiling Group) and the Portuguese and Spanish Working Group of the ISFH<br />

(GEPISFH).<br />

Recently, the universities as well as some private research and diagnostic labs<br />

began to use DNA Sequencers, opening the spectrum of applications.<br />

We present in this poster the state of the art of Automated Sequence and<br />

Fragment Analysis in Portugal and its relationship with other iberian and<br />

latin-american countries, with a special emphasis onto the forensic genetics<br />

field.<br />

P12-T<br />

Investigation of the entire spectrum of genes induced by staphylococcal<br />

enterotoxin B in human lymphoid cells using differential<br />

display-PCR.<br />

C.A. Mendis, C. Sanchez, R. Das, M. Jett; Walter Reed Army Inst. of Res.,<br />

503 Forney Drive, Silver Spring, MD 20910<br />

Since its introduction in 1991, Differential Display has evolved into a powerful<br />

and efficient technique in analyzing differentially expressed genes in<br />

various cells under altered conditions. Here we used differential display not<br />

only to screen the entire gene population in human lymphoid cells induced<br />

by SEB but also to understand the specificity of the expression pattern of<br />

altered genes by comparing it to the expression by Cholera Toxin (CT). In<br />

order to approach our goal in a systematic manner as well as to increase the<br />

efficiency of cDNA product recovery we used a set of arbitrary and anchored<br />

primers to further subdivide the gene population. Out of the 750 altered<br />

genes, at least 200 genes showed a SEB specific expression pattern when<br />

compared to CT exposure. The altered genes were then subjected to a high<br />

throughput cloning procedure and sequenced. Out of the 250 genes that<br />

were sequenced, 150 genes matched with available sequences in the Gene<br />

Bank and EMBL databases. Unique to SEB are genes involved in ancillary<br />

functions as adhesion molecules, regulators of vascular tone, wound healing,<br />

inflammation, heat shock, cell death and T-cell proliferation. Altered genes<br />

that matched with known sequences were verified by northern blots, RT-PCR<br />

or real time PCR. Genes that were specific for SEB were then placed on glass<br />

chips for use with other cell lines or to understand the gene expression profile<br />

of other toxins. In the quest for finding a set of genes that can be used<br />

as surrogate markers, differential display has shown to be a proven technique<br />

as it has the capability to look at the whole gene spectrum irrespective of<br />

wide use or availability of individual genes.


P13-S<br />

Microsatellite analysis using fluorescent PCR primers synthesized<br />

in tandem.<br />

S.L. Wegener, G.J. Wiebe, S. Yu, R.T. Pon; Univ. of Calgary,<br />

3330 Hospital Drive N.W., Calgary, <strong>AB</strong> T2N 4N1, Canada<br />

A new procedure for solid-phase synthesis of multiple oligonucleotides<br />

linked end-to-end (tandem synthesis) on the same solid-phase support has<br />

recently been developed in our laboratory. Upon cleavage and deprotection,<br />

the oligonucleotides are released from each other yielding a mixture of two<br />

or more sequences in the same solution. This procedure has practical benefits<br />

for applications requiring large sets of oligonucleotides, especially PCR<br />

amplification, since the number of individual oligonucleotide syntheses to be<br />

set-up and the number of individual oligonucleotide samples handled is<br />

reduced by half. Fluorescently labelled primer pairs for automated genotyping<br />

are also possible by incorporating a fluorescent dye phosphoramidite<br />

onto the 5�-end of the terminal oligonucleotide. PCR primer pairs with FAM,<br />

HEX, and TET fluorescent labels have been prepared by tandem synthesis<br />

and used to amplify known microsatellite markers from (C57BL/6 � NOD)<br />

mouse genomic DNA. Automated microsatellite analysis using GeneScan<br />

and an Applied Biosystems Prisim 377 DNA sequencer was then performed.<br />

P15-T<br />

Quality control in a core oligonucleotide synthesis facility.<br />

R.R. Muhlhauser, C.G. Miller, A. Yeung; Fox Chase Cancer Ctr.,<br />

7701 Burholme Avenue, Philadelphia, PA 19111<br />

Our facility makes about 400 oligonucleotides each month. For the past 16<br />

years, we have done quality control on each oligonucleotide prior to delivery.<br />

Our users do not want the quality of oligonucleotides to ever be a variable<br />

for troubleshooting in their experiments. Even a failure rate of 1% in<br />

oligonucleotide synthesis would have caused four experiments to fail each<br />

month. Such a low rate of synthesis failure is difficult to isolate in a statistical<br />

approach of quality control by random sampling. Analyzing every<br />

oligonucleotide also allows us to catch instrument and reagent problems<br />

more quickly. By analyzing each oligonucleotide immediately after deprotection,<br />

we reject 1 to 3% of the oligonucleotides we make that do not meet<br />

our quality standard. These oligonucleotides are resynthesized without delay.<br />

The oligonucleotide analysis method we use is anion-exchange at pH 12.5,<br />

using the Mono Q system on an FPLC (Yeung, A. T. and Miller, C. G. Anal.<br />

Biochem. 187:6675, 1990), assisted by a home-made autosampler and autoinjector.<br />

At that pH, both n-1’s and deprotection problems are visible. We<br />

highly recommend this practice because it does not add appreciable expense<br />

or effort to the cost of the oligonucleotide, but provides the facility personnel<br />

with assurance in the quality of their products.<br />

POSTER <strong>AB</strong>STRACTS<br />

<strong>AB</strong>RF 2001 <strong>AB</strong>STRACTS<br />

P14-M<br />

Economical usage of value-added phosphoramidites.<br />

J.B. Hobbs; Univ. of British Columbia, #237-6174 University Boulevard,<br />

Vancouver, BC V6T 1Z3, Canada<br />

A method is described for economical use of value-added (dye, biotin, etc.)<br />

phosphoramidites. The method was developed on an Applied Biosystems<br />

380B synthesizer. The amidite is placed in a glass autosampler vial which is<br />

located in a plastic jacket inside a 10 ml glass vial. The amidite is dissolved<br />

in situ using a user-variable microdilution adapted from a bottle-change procedure.<br />

Typically 5 mg. of amidite are dissolved in 100 microlitres of acetonitrile.<br />

The amidite is used in conjunction with Applied Biosystems LV<br />

columns and synthesis cycles adapted for use with LV columns which use a<br />

similar “multiple-coupling” approach to that seen in “LV cycles” on later (392,<br />

394, etc.) Applied Biosystems synthesizer models. These cycles are also<br />

described. Good tagging efficiency can be obtained for 40 nmole or 200<br />

nmole scales using 5 mg. of amidite in this way, and the wastage of large<br />

quantities of expensive amidites is avoided. While the methods are applicable<br />

to later model synthesizers, the relatively modest flow rates, gas pressures<br />

and line volumes of the 380B render it a good platform for processes of this<br />

type.<br />

P16-S<br />

Incorporation of pseudouridine and 4-thio-uridine into RNA<br />

oligonucleotides using 5�silyl-2�-ACE-orthoester chemistry.<br />

S.A. Scaringe1, D. Kitchen2, J. Qui2; 1Dharmacon Res. Inc.,<br />

3200 Valmont Road #5, Boulder, CO 80301, 2Dharmacon Res., Inc.,<br />

Boulder<br />

The ability to incorporate a wide range of modified ribonucleotides into RNA<br />

oligos is an essential requirement for RNA chemical synthesis methodologies.<br />

Many modified bases have already been incorporated into RNA oligos using<br />

5�-Silyl-2�-ACE-orthoester chemistry. We report here the incorporation of<br />

two more important modified bases, pseudouridine and 4-thio-uridine.<br />

Pseudouridine was converted from the nucleoside to the protected nucleoside<br />

phosphoramidite in five steps in 45% overall yield. During synthesis the<br />

amidite coupled in 99% stepwise yields. The high yields for both the amidite<br />

synthesis and the oligonucleotide coupling have allowed pseudouridine to<br />

be incorporated into RNA on a routine basis. For example, a series of RNA<br />

oligonucleotides containing 1–3 pseudouridines were synthesized to study<br />

the 1920 loop region of E. coli 23S RNA.<br />

4-Thio-uridine was incorporated into RNA oligos using the convertible nucleoside<br />

approach. The protected 4-triazole-uridine phosphoramidite was synthesized<br />

from uridine in six steps in 32% yield. The amidite coupled in 99%<br />

yields. Following oligo synthesis the support bound oligonucleotide was<br />

treated with thioacetic acid, buffered to pH 8.0, for 6 hours at 55�C. The support<br />

was washed and then treated with 10% DBU methanol at room temperature<br />

for 24 hours. The oligonucleotide was desalted and 2�hydroxyl<br />

protecting groups were removed using a pH 3.8 aqueous buffer for 30 minutes<br />

at 55�C. The resulting oligonucleotides contained 98% 4-thio-uridine at<br />

the desired position as assayed by anion exchange HPLC analysis. This is significantly<br />

higher than the results using cyanoethyl protection of 4-thiouridine.<br />

All oligonucleotides containing 4-thio-uridine clearly exhibited the<br />

characteristic 330 nm absorption peak. 4-Thio-uridine is now being routinely<br />

incorporated in RNA oligonucleotides for use in several collaborative<br />

studies.<br />

JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000 191


<strong>AB</strong>RF 2001 <strong>AB</strong>STRACTS<br />

P17-M<br />

New methods for the synthesis of multiple oligonucleotide<br />

sequences either singly or linked end-to-end in tandem.<br />

R.T. Pon1, S. Yu1, Y.S. Sanghvi2; 1Univ. of Calgary, 3350 Hospital Dr. NW,<br />

Calgary, <strong>AB</strong> T2N 4N1 Canada, 2Isis Pharmaceut.<br />

Solid-phase oligonucleotide synthesis requires attachment of the 3�-terminal<br />

nucleoside to the support, usually through a 3�-ester linkage. Phosphoramidite<br />

methods for performing this attachment through a phosphate linkage<br />

on “Universal” supports have been developed. However, these methods<br />

introduce a terminal 3�-phosphate residue which is difficult to remove. We<br />

have developed new linker phosphoramidite reagents which eliminate this<br />

problem by incorporating a cleavable 3�-ester linkage within the linker arm.<br />

Thus, only products with 3�-OH ends are produced and no extra conditions<br />

for 3�-dephosphorylation are required. These new reagents provide an alternative<br />

to our previous method for automated nucleoside attachment using<br />

HBTU and DMAP (Pon and Yu, Synlett 1999, 1778). The linker phosphoramidites<br />

are especially suited to high-throughput synthesis in 96-well plates<br />

since inexpensive, underivatized CPG can be used as a “Universal” support.<br />

Nucleoside attachment to the 5�-OH group of a prior sequence is also possible.<br />

This allows multiple oligonucleotides linked end-to end in tandem to<br />

be prepared in a single synthesis. The linked oligonucleotides are released<br />

from each other upon cleavage from the support and used together as a mixture.<br />

Sequences prepared in tandem can be PCR primers, forward and<br />

reverse sequencing primers, or duplex DNA fragments. The total number of<br />

bases is only limited by the support’s pore size. We have prepared strings of<br />

oligonucleotides up to 96 bases long (4 � 24 base primers) and combinations<br />

of up to ten shorter sequences.<br />

P19-S<br />

Production of antigen specific MHC class I tetramers as a core<br />

facility service.<br />

P.S. Adams, T.B. Miller, M.J. Dobrzanski, R.J. Hogan, D.L. Woodland;<br />

Trudeau Inst., 100 Algonquin Avenue, Saranac Lake, NY 12983<br />

Major Histocompatibility Complex (MHC) Class I tetramers are composed of<br />

a tetrameric complex of the MHC Class I molecule and a specific antigenic<br />

peptide. The Class I heavy chain has been modified to contain a BirA<br />

sequence that allows biotinylation and subsequent tetramerization of the<br />

complex with a fluorescently tagged strepavidin. These tetramers can be used<br />

to identify antigen specific CD8� T cells involved in immune responses using<br />

flow cytometry. The Molecular Biology Core Facility prepares these tetramers<br />

as a core facility service to aid the researchers at the Trudeau Institute. One<br />

project involves the characterization of the underlying mechanisms and<br />

immunoregulatory roles of tumor antigen-reactive Tc1 and Tc2 effector cell<br />

sub-populations, which can provide insight into the modification and/or<br />

development of novel immunotherapeutic approaches to disseminated malignancies.<br />

Another project involving viral immunity has demonstrated that a<br />

substantial population of antigen specific CD8� T cells can be found not<br />

only in the secondary lymphoid organs, but also in the peripheral tissues after<br />

resolution of a primary respiratory virus infection. These data have significant<br />

implications for vaccines that induce systemic rather than local immunity. The<br />

production procedure and examples of the usefulness of MHC Class I<br />

tetramer technology will be presented.<br />

POSTER <strong>AB</strong>STRACTS<br />

192 JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000<br />

P18-T<br />

Performance of high-speed oligonucleotide synthesizer in 96-well<br />

format.<br />

D. Luk, A. Tuyet-Doan, S. Hall, J. Koh, R. Guettler; GeneMachines,<br />

935 Washington St., San Carlos, CA 94070<br />

Oligonucleotides are essential elements of genomic research. The Poly-<br />

PlexTM oligonucleotide synthesizer makes 96 different oligos in standard 96well<br />

format, which is amenable to downstream high-throughput processing<br />

and handling. Operator time is minimal and synthesis time is less than 3<br />

hours for a 96-well plate of 20-mers. By eliminating flushing of reagent lines<br />

through a parallel dispensing technology, optimal synthesis time and reagent<br />

consumption are achieved. PolyPlex’s low reagent consumption generates<br />

oligos quickly, at low costs and small scales as low as 10 nmol. Synthesis<br />

costs, including all consumables, are less than $0.10 per base. The PolyPlex<br />

synthesis chamber provides an inert-gas environment where synthesis<br />

progress can be monitored by using full-plate trityl collection after any base<br />

addition. PolyPlex utilizes fully licensed chemistry and generates oligos with<br />

greater than 98% coupling efficiency. Versatile, easy-to-use software provides<br />

powerful synthesis control along with extreme simplicity.<br />

P20-M<br />

Advantages of directionally immobilized 6xHis-tagged proteins<br />

for capture assays.<br />

K. Steinert, F. Schäfer, S. Wahle, A. Hoffer, P. Söhnlein, J. Ribbe; QIAGEN<br />

GmbH, Max-Volmer-Strasse 4, Hilden, Nordrhein-Westfalen 40724,<br />

Germany<br />

Assay signals and reproducibility can be greatly enhanced by immobilizing<br />

the assay components in a directed manner. For example, to set up an interaction<br />

assay it is essential that the immobilization of the capture molecule<br />

occurs in active conformation to allow binding of the interaction partner. Similarly,<br />

oriented binding in immunoassays results in optimal accessibility of<br />

antigenic regions and therefore increased signal strengths. Directed immobilization<br />

of assay components can easily be achieved by using 6xHis-tagged<br />

recombinant proteins and immobilizing them via their 6xHis tags. The use of<br />

this interaction is well established for efficient purification of 6xHis-tagged<br />

proteins, but is also advantageous for immobilization of recombinant proteins<br />

in highly sensitive and reproducible protein-based assays. In addition, relying<br />

on the 6xHis-Ni-NTA interaction for immobilization of assay components<br />

simplifies assay development, because immobilization conditions no<br />

longer have to be optimized for each individual protein, but rely on the same<br />

biochemical characteristics of the 6xHis-Ni-NTA interaction for each protein.<br />

As an alternative to Ni-NTA, monoclonal antibodies recognizing the 6xHis tag<br />

can be used for highly specific and oriented immobilization of 6xHis-tagged<br />

proteins. The effect of specifically oriented binding on assay results is demonstrated<br />

by comparison of ELISA experiments employing proteins passively<br />

adsorbed to polystyrene plates and proteins immobilized via their 6xHis tags,<br />

as well as by a study on the interaction of two chaperones and development<br />

of a protease assay.


P21-T<br />

Versalinx chemical affinity tools for purification in protein analysis.<br />

J.P. Wiley, M.D. Ferguson, A. Gall, K.A. Hughes, G. Li, K.P. Lund,<br />

M.L. Stolowitz; Prolinx, Inc., Bothell, WA, 22322 20th Ave. SE, Bothell,<br />

WA 98072-8341<br />

Fractionating complex biological mixtures is one of the challenges in proteome<br />

analysis. MALDI-TOF (matrix assisted laser desorption ionization time<br />

of flight) mass spectrometry analysis, for instance, has a high mass accuracy<br />

but requires a relatively pure sample for positive structural identification. Prolinx<br />

® Inc. has developed Versalinx Chemical Affinity Tools, and has demonstrated<br />

success using this technology to isolate and purify proteins of interest<br />

from complex mixtures. Versalinx Chemical Affinity Tools are based upon<br />

the interaction of phenyldiboronic acid (PDBA) and salicylhydroxamic acid<br />

(SHA) to form a complex that is reversible under certain conditions. Immobilization<br />

of PDBA-modified proteins can occur on a variety of SHA-modified<br />

surfaces. The system is stable over a wide pH range and is compatible with<br />

a number of denaturants, detergents and organic solvents. With Versalinx<br />

Chemical Affinity Tools, purification of biological molecules is rapid and<br />

reproducible.<br />

P23-M<br />

Detection of residual isopropyl-1-thio-�-D-galactopyranoside<br />

in proteins by HPLC with pulsed amperometric detection.<br />

X. Ji, L. Couch, A. Pennetti, K. Venkat, J. Mozdzanowski; SmithKline<br />

Beecham Pharmaceut., 709 Swedeland Road, King of Prussia, Pa 19406<br />

Isopropyl-1-thio-�-D-galactopyranoside (IPTG) is an inducer of the lac<br />

operon and may be used in fermentation processes for the production of protein<br />

biopharmaceuticals in E. coli. Use of this compound in the fermentation<br />

process creates the need for the determination of residual IPTG in the final<br />

product. Chemical and optical properties of IPTG (e.g. absorbance in the UV<br />

region) do not permit sensitive detection using traditional techniques. Hemiacetal<br />

group of galactose is substituted by a 2-thiopropyl group and thus<br />

eliminates an option for simple derivatization reactions leading to fluorescent<br />

derivatives. IPTG is a sulfur-containing galactoside, and therefore, properties<br />

of sulfur and sugar may be utilized for electrochemical detection. With this<br />

mode of detection, residual IPTG can be detected in the presence of high<br />

concentrations of protein. This required development of a modified HPLC<br />

separation method permitting the use of high concentrations of acetonitrile<br />

to remove protein from the column during a wash step. HPLC conditions<br />

developed for the separation are free from interference with protein and a<br />

so-called “oxygen dip” typically observed with the gold electrode. The waveform<br />

developed by William LaCourse for the Dionex electrochemical detector<br />

(ED-40) with the gold electrode can be used for the determination of<br />

IPTG at a concentration as low as 0.05 �M (12 ng/mL).<br />

<strong>Reference</strong>:<br />

William LaCourse, Determination of Isopropyl-�-D-thiogalactoside (IPTG) by<br />

HPLC-Pulsed Electrochemical Detection. Pittcon-2000 presentation.<br />

POSTER <strong>AB</strong>STRACTS<br />

<strong>AB</strong>RF 2001 <strong>AB</strong>STRACTS<br />

P22-S<br />

Fully automated 96-well protein purification and magnetic beadbased<br />

assay using 6xHis-Ni-NTA technology.<br />

K. Steinert, H. Lubenow, S. Wahle, R. Stoll, J. Ribbe; QIAGEN GmbH,<br />

Max-Volmer-Strasse 4, Hilden, Nordrhein-Westfalen 40724 Germany<br />

The purification of proteins from expression libraries requires a method that<br />

performs reliably regardless of the characteristics of the proteins to be purified.<br />

Using Ni-NTA for purification of 6xHis-tagged recombinant proteins provides<br />

a one-step purification method that is robust and meets the challenge<br />

presented by the need to purify thousands of proteins with differing structure<br />

and characteristics. To allow this method to be performed in highthroughput<br />

applications, automated protocols were developed which run on<br />

BioRobot systems.<br />

Ready-to-run protocols covering a wide range of protein assay and purification<br />

applications are available. They rely on 6xHis-Ni-NTA technology based<br />

on the high affinity and specificity of nickel ions immobilized on matrices<br />

bearing nitrilotriacetic acid (NTA) for recombinant biomolecules with a tag<br />

of six consecutive histidine residues (6xHis tag).<br />

Ni-NTA Magnetic Agarose Beads allow flexible interaction or diagnostic<br />

assays with structurally active immobilized 6xHis-tagged proteins as well as<br />

micro-scale purification of up to 15 �g of 6xHis-tagged protein. The purification<br />

and assay protocols based on Ni-NTA Magnetic Agarose Beads may<br />

even be carried out directly in series.<br />

If much larger amounts of purified proteins are needed, the Ni-NTA Superflow<br />

96 BioRobot Protocol provides a convenient high-throughput method<br />

for purification of approximately 100 �g of 6xHis-tagged protein per well in<br />

a 96-well format. Examples of purification and assay applications using both<br />

methods are shown and data on reproducibility and cross-contamination free<br />

processing are given.<br />

P24-T<br />

Construction of a tagging system for subcellular localization of<br />

proteins encoded by newly discovered open reading frames.<br />

W-J. Syu; Natl. Yang Ming Univ., 155 Sec 2, Li-Long St., Pai-Tao,<br />

Taipei, Taiwan 112, Taiwan<br />

We have previously characterized a monoclonal antibody (SC1D7) that is<br />

directing to maltose-binding protein (MBP) of Escherichia coli and other<br />

closely related enteric bacteria. SC1D7 does not cross-react with proteins in<br />

eucaryotes and appears to be a highly specific tool in immunochemical<br />

analyses. To better map the epitope, we took advantage of an available plasmid<br />

pMAL-c2 that encodes E. coli MBP-coding sequence and constructed<br />

plasmids to express MBP fragments. A construct containing the N-terminal<br />

portion of MBP does not react with SC1D7 whereas a second construct<br />

expressing glutathione-S-transferase fused with the C-terminal half of MBP<br />

does react with SC1D7. To precisely define the epitope, random peptides displayed<br />

on M13 were used to react with SC1D7. Sequences of reactive peptides<br />

were aligned, and a consensus sequence of XDXRIPX was deduced.<br />

This sequence matches to MBP with an amino acid stretch of KDPRIAA. To<br />

consolidate the mapping result, a sequence encoding this epitope was<br />

inserted into an expression vector and the resulted recombinant protein did<br />

react with SC1D7. Thereafter, this epitope was incorporated into an eucaryotic<br />

expression plasmid that contains a previously defined hepatitis delta<br />

virus epitope for protein tagging. The so constructed two-epitope-tagging<br />

vector is useful in various molecular analyses. We demonstrated its usage in<br />

localization of a bacterial virulence factor in host cells. This vector should be<br />

applicable for high throughput characterization of new open reading frames<br />

found in genome sequencing.<br />

JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000 193


<strong>AB</strong>RF 2001 <strong>AB</strong>STRACTS<br />

P25-S<br />

Truncated midkine (tMK): its molecular cloning and expression in<br />

Escherichia coli, and the detection of native tMK in Wilms’ tumor<br />

by an anti-tMK monoclonal antibody.<br />

S. Paul1, W. Dansithong1, T. Mitsumoto1, Y. Asano1, M. Kato2, M. Kato2, T. shinozawa1; 1Gunma Univ., Japan, 1-5-1 Tenjin-cho, Kiryu, Gunma<br />

376-8515 Japan, 2Tottori Univ., Japan<br />

Midkine (MK) is a heparin binding growth factor identified as a product of<br />

a retinoic acid-responsive gene; it is frequently expressed at high levels in<br />

many human carcinomas. Although the expression of the mRNA encoding<br />

truncated MK (tMK) in unique human cancer cells has been reported, the<br />

tMK polypeptide itself has not yet been identified. In order to clarify the biological<br />

role of tMK, recombinant tMK was expressed in Escherichia coli and<br />

purified. Purified recombinant tMK showed the same extent of proliferative<br />

activity for the Wilms’ tumor (G401) cells as full length human MK. A mouse<br />

hybridoma cell line producing an IgG2b monoclonal antibody (mab) against<br />

this purified recombinant tMK was also established. This anti-tMK mab<br />

(MiStMK-V3) did not crossreact with synthetic full length (or c-half) human<br />

MK. A native tMK, showing the same apparent molecular mass as the recombinant<br />

tMK in SDS-PAGE, was identified in G401 cells using this mab. These<br />

results suggest that the structure of the recombinant tMK is same as that of<br />

native tMK. The expression of tMK in Wilms’ tumor patient specimens was<br />

also detected in an immunohistochemical study using the anti-tMK mab. The<br />

usefulness of this mab (MiStMK-V3) for the detection of tMK in Wilms’ tumor<br />

was demonstrated.<br />

P27-T<br />

Performance evaluation of a novel application for comparative<br />

DNA sequencing analysis and mutation detection.<br />

C.A. Kosman, L. Johnston-Dow, H. Breu, G. Chappell, S. Kumar,<br />

B. Iasnopolski, B. Kshirsagar, P.A. Suri, R. Paul, G. Mason, D. Siu,<br />

E. Sword, M. Schoppe, V. Bawge; Applied Biosystems, 850 Lincoln Centre<br />

Drive, Foster City, CA 94404<br />

Various software tools are available for the analysis and interpretation of denovo<br />

DNA sequencing data. While such applications work well for sequencing<br />

for discovery many of the underlying assumptions in these tools are not<br />

valid when applied to comparative sequencing data.<br />

We have developed a new practical application for comparative DNA<br />

sequencing analysis. The focus of this new application is the discovery or<br />

identification of sequence variants in a dataset of sequence derived from a<br />

single locus in various individuals. This tool allows for the rapid and accurate<br />

analysis and alignment of multiple sequence comparisons containing<br />

mixed base positions against a reference sequence. While NT-based, this software<br />

is compatible with data generated from all of Applied Biosystems<br />

sequencing platforms (<strong>AB</strong>I PRISM ® 373, 377, 310, 3100 and 3700).<br />

This application uses procedures and algorithms better suited to the unique<br />

nature of comparative sequencing data. Specifically, the prior knowledge of<br />

a reference sequence available in many comparative sequencing projects is<br />

used to streamline and automate portions of the analysis. The software performs<br />

sequence analysis and aligns the analyzed sequence to a reference<br />

sequence. It also aligns imported sequences that contain variants. It will then<br />

analyze the compared sequences, provide protein translation and report the<br />

analysis in a convenient format.<br />

Here, we present the results from the performance evaluation of this new<br />

software tool on several types of comparative sequencing datasets. For these<br />

analyses, this new application produced results concordant with those previously<br />

obtained using standard DNA analysis tools while providing substantial<br />

performance advantages over these tools. In all cases the analyses<br />

were completed in significantly less time and required less manual manipulation<br />

of the data. The innovative approaches employed in this tool result in<br />

less user intervention needed to achieve a better level of mutation detection.<br />

POSTER <strong>AB</strong>STRACTS<br />

194 JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000<br />

P26-M<br />

Information management systems for molecular biology.<br />

T.M. Smith, D. Campbell, R. Connelly, A. Leonard, S.G. Porter, J. Slagel;<br />

Geospiza, Inc., Seattle, 2442 NW Market St. #344, Seattle, WA 98107<br />

Technological advances in molecular biology have made it possible for laboratories<br />

to generate unprecedented amounts of data. DNA sequencing, for<br />

example, has seen an increase in throughput of over 400-fold in recent<br />

years. The accelerated production of data has produced a corresponding<br />

need for better data management.<br />

The increased need for data management is partly due to institutional<br />

changes. Many institutions have been able to economize by establishing core<br />

facilities which offer laboratory services to researchers on a per sample or<br />

project basis. Core facilities are cost-effective because they provide several<br />

researchers with access to expensive equipment and skilled personnel.<br />

Because these core facilities handle a variety of data types and a large number<br />

of samples from multiple sources, their information management needs<br />

provide a worthwhile model for laboratory information management systems<br />

(LIMS).<br />

LIMS are collections of software, communication devices, and computers that<br />

acquire, store, analyze, and present data and information about laboratory<br />

samples and their processing. LIMS centralize data storage, automate data<br />

analysis, and provide quality assurance reports for process monitoring. The<br />

central component of a LIMS is a database with software interfaces for entering,<br />

viewing, and processing information.<br />

The level of complexity needed in a LIMS depends on factors such as the<br />

mission of the laboratory, the number of samples to be processed, analysis<br />

requirements, and workflow complexity. LIMS development presents many<br />

challenges and may take years to complete. Most labs have no need for a<br />

custom LIMS and lack the wherewithal to create one, and thus will benefit<br />

from commercial LIMS<br />

The general features of LIMS will be presented along with case studies of<br />

Geospiza’s Finch-Suite LIMS solution illustrating how a commercially available<br />

software system can be used to meet the information management<br />

needs of DNA sequencing facilities.<br />

P28-S<br />

A Web interface for BioLIMS System—data and user management<br />

for sequencing core facility.<br />

L. Liu, L. Roinishvili; Univ. of Illinois, Urbana, 330 ERML,<br />

1201 W. Gregory Dr., Urbana, IL 61801<br />

Sequencing core facility serves many customers. Samples in a sequencing<br />

core facility are very heterogeneous although the volume of samples from<br />

each customer is relatively small. Data and user management can be a challenge<br />

task. The BioLIMS database system from PE Bioinformatics provides a<br />

smooth data flow from sequencer to database. However, it really lacks of<br />

friendly user interface for both core facility and customers. To provide an<br />

easy management system for the core facility at the W. M. Keck Center for<br />

Comparative and Functional Genomics, we implemented a web interface for<br />

BioLIMS system. This interface serves two purposes in the core facility. One<br />

is for core facility staff to manage customers and their data. The other is for<br />

customers to view and retrieve their own data from the web. In customer<br />

management, we allow super user and sub user relationship. Super user will<br />

have access to all the data of his or her sub users. In data management, a customer<br />

can only view and retrieve his or her own data unless others give him<br />

or her privilege to view their data. The most convenient feature is web<br />

based. Customers can access their data from anywhere at anytime.


P29-M<br />

Protein/DNA technology DSL version 2000 is a set of shareware<br />

protocols for sample and data management.<br />

J. Medalle, M. Randesi, B. Imai; Rockefeller Univ., 1230 York Ave.,<br />

Box 105, New York, NY 10021<br />

Physical sample and data management are major concerns for a DNA<br />

sequencing core facility. Especially one with a small staff and limited space<br />

are affected the most. Samples can overtake the limited freezer space. Paper<br />

trail can overfill office and lab spaces. Both can cause a quick turnover of<br />

DNA sequencing staff. DNA Sequencing Lab’s (DSL) version 2000 objectives<br />

are to alleviate repetitive computer tasks for DNA Sequencing staff and eliminate<br />

physical paper trail. DSL 2000 is a set of protocols for sample and data<br />

management. Its major components are comprised of online submittal forms<br />

at http://protein13-pc.rockefeller.edu/, scripts from Apple Script, and standardized<br />

forms and macros from Microsoft Excel 98, and Outlook Express 5.<br />

The DSL funnels the online sample submissions into sample statements for<br />

each investigator. This is a file containing sample IDs, names, account numbers,<br />

primer info, template info, etc. DSL also transforms the web submissions<br />

into logs for prep and for slab or capillary sequencing reactions. These logs<br />

direct the staff on how to process incoming samples. The set of protocols<br />

streamlines the sample flow from the investigator to the machine as well as<br />

the data flow from machine back to the investigator. DSL’s protocols are<br />

“hands on” to help the staff create e-mails for investigator notifications and<br />

“a click of a button” to compress and to transfer investigator’s sample statements<br />

or data files into their web account. DSL 2000 is a freeware application<br />

and can be integrated into other small core facilities with a limited budget.<br />

P31-S<br />

The analysis of complex tryptic peptide mixtures by multidimensional<br />

LC-MS/MS on a hybrid quadrupole orthogonal<br />

acceleration time-of flight (Q-TOF) mass spectrometer.<br />

A. Millar1, C. Hughes1, T. Andresson2, T. Hemesath2, J.I. Langridge1; 1Micromass UK Ltd., Floats Road, Wythenshawe, Manchester M23 9LZ,<br />

United Kingdom, 2deCODE genetics Inc., Reykjavik<br />

Advances in both HPLC and mass spectrometry instrumentation have allowed<br />

the analysis of protein complexes which have not been separated on a two<br />

dimensional gel. These experiments involve separation of the complex digest<br />

mixture by microcapillary liquid chromatography connected to an instrument<br />

capable of data directed switching between the MS and MS/MS modes. Protein<br />

identification is then achieved via databank searching of the ESI-MS/MS,<br />

providing qualitative information on the proteins that are present. Hundreds<br />

of MS/MS spectra can be acquired in a fully automated fashion, resulting in<br />

the identification of significant numbers of proteins, including low copy<br />

number proteins, from a single LC-MS/MS experiment1. If, however, a complex protein mixture is to be investigated then a fractionation<br />

step prior to separation of the peptides on the basis of their hydrophobicity<br />

is advantageous. We have, therefore, adopted a 2D LC-MS/MS approach<br />

using a capillary LC system (CapLC) operating at nanoliter per min<br />

flow rates coupled to a Q-Tof 2 mass spectrometer. By placing a strong cation<br />

exchange (SCX) cartridge followed by a C18 trap cartridge it is possible to<br />

pre-fractionate the peptides before separation on an analytical C18 column.<br />

After loading the sample, discreet fractions are sequentially eluted from the<br />

cation exchange cartridge using a salt step gradient; the eluted peptides are<br />

then retained on the trapping C18 cartridge whilst they are desalted. Finally<br />

the peptides are eluted from the C18 pre-column, at 200 nL/min, onto a 75<br />

�M ID � 10 cm Waters Symmetry analytical column for separation and elution<br />

into the mass spectrometer.<br />

This analytical approach will be discussed with examples where this methodology<br />

has been used for the analysis of standard protein mixtures and for the<br />

analysis of cell lysates and sub-cellular fractions.<br />

1. Yates et al., Nature Biotechnology (1999);17, (7), 676–682.<br />

POSTER <strong>AB</strong>STRACTS<br />

<strong>AB</strong>RF 2001 <strong>AB</strong>STRACTS<br />

P30-T<br />

A laboratory information management system for a small<br />

DNA sequencing core facility.<br />

M.J. Miller; NCI, NIH, Bldg 37, Rm 3C28, MSC 4255, Bethesda,<br />

MD 20817-4255<br />

The DNA Sequencing MiniCore facility of the NCI’s Division of Basic <strong>Science</strong>s<br />

currently services over 300 investigators. The facility is designed to support<br />

small scale, short-term sequencing. In the past year we ran over 25,000 samples<br />

with an average turnover time of less than one day. While this represents<br />

a 60% increase in samples over the year before, it is still a relatively<br />

modest operation.<br />

Keeping all these users and their data organized, as well as minimizing the<br />

amount of paperwork involved is a major concern. Although several Laboratory<br />

Information Management (LIM) System software packages exist, they<br />

are often too expensive or too inflexible for a small operation such as ours.<br />

I describe here a LIM system built using components of the Microsoft Office<br />

software package. Users enter sample information data into an Internet form<br />

and this data is automatically entered into the database after inspection by<br />

the MiniCore staff. Programs built into the database determine which sample<br />

sets can be run together on the same gel, and what parameters (such as<br />

which virtual filter and which dye/primer set settings) should be used with<br />

each sample. Samples are assigned to a particular gel and the “sample sheet”<br />

for that gel is automatically generated by the database. By keeping track of<br />

when samples are submitted, how many samples there are, and how many<br />

basepairs of read the user requests, the database aids the facility’s operators<br />

in determining sample priority. An email message is automatically sent when<br />

samples have been processed and data deposited in the user’s data-destination<br />

directory. The database also keeps track of charges and automatically<br />

sends this data to the NIH’s accounting system. The facility thus runs in a virtually<br />

“paperless” environment. There are no hardcopy forms to fill out. All<br />

information is maintained by the computer system.<br />

P32-M<br />

Identification of in vivo phosphorylation sites in<br />

Drosophila armadillo by tandem mass spectrometry.<br />

C.S. Raska, R.M. Pope, D. Rubenstein; Univ. of North Carolina at<br />

Chapel Hill, 4 Casabelle Ct, Durham, NC 27713<br />

Phosphorylation is one of the most important reversible modifications of<br />

eukaryotic proteins. Often, proteins which are associated with uncontrolled<br />

cell growth, and ultimately cancer show an anomaly in their phosphorylation/dephosphorylation<br />

pathways. In humans, a protein, �-catenin, plays a<br />

central role in the development, organization, and regulation of epithelial tissues.<br />

Aberrant regulation of �-catenin is associated with malignancies. For<br />

example, alterations in �-catenin gene structure have been identified in colorectal<br />

and breast carcinoma, and in a large number of melanoma cell lines.<br />

Specifically, �-catenin signaling function is constitutively active in many<br />

melanomas due to mutations that remove phosphoresidues at the amino terminus<br />

of the protein. Armadillo, a protein found in Drosophila melanogaster,<br />

is 71% identical to �-catenin at the amino acid level. �-catenin from Drosophila<br />

and from human keratinocytes binds to armadillo and �-catenin,<br />

respectively, and this binding has been found to be influenced by phosphorylation.<br />

Thus, we have been using armadillo as a model system to study<br />

�-catenin regulation. To further define the regulatory function of phosphorylation,<br />

we are using mass spectrometry to map post-translationally modified<br />

amino acids in armadillo. Armadillo was isolated by immuno-affinity and<br />

ion exchange chromatography. 1-D SDS-PAGE, and in-gel tryptic digestion<br />

were performed to produce a mass spectral fingerprint on a triple quadrupole<br />

instrument using nanoelectrospray. MS scans confirmed peptides matching<br />

both armadillo and �-catenin from one gel spot. Neutral loss scans<br />

identified potentially phosphorylated peptides. MS/MS generated enough<br />

sequence coverage to specifically identify phosphorylated residues. We note<br />

that phosphoresidues are located within the region where binding sites for<br />

cadherin, dTCF, and dAPC are located. We plan mutation studies to further<br />

elucidate the role of phosphorylation in these systems.<br />

JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000 195


<strong>AB</strong>RF 2001 <strong>AB</strong>STRACTS<br />

P33-T<br />

A targeted approach for more sensitive and accurate protein<br />

identification.<br />

D.K. Leung, M.J. Horn; BioMolecular Technol., Inc., 525F Del Rey Ave.,<br />

Sunnyvale, CA 94086<br />

Selective isolation of peptides containing low abundance amino acids such<br />

as tryptophan, offers a direct way to simplify protein digest mixtures prior to<br />

mass spectrometric analysis. This approach provides benefits for both in-gel<br />

digests and digests of protein mixtures.<br />

To this end a solid phase reagent capable of selectively binding to tryptophan<br />

has been developed. This solid support material binds covalently to tryptophan<br />

or tryptophan containing peptides, allowing for their selective separation<br />

from mixtures of peptides and proteins. Bound peptides can be released<br />

from the solid-support by a reducing reagent to give analytical samples for<br />

further analysis (LC, MS, etc.). The specificity of the binding chemistry coupled<br />

with mass spectrometric analysis by MALDI affords improved accuracy<br />

in data base search and in protein identification.<br />

<strong>Reference</strong>s<br />

1. Shechter, Y., Rubinstein, M. and Patchornik, A. (1976), Biochemical and<br />

Biophysical Research Comm. {4} (70), 1257.<br />

2. Early, S. L., Magil, S. G., Novak, C. and Horn, M. J. (1989), Techniques in<br />

Protein Chemistry, Academic Press, Inc., 439.<br />

3. McEldoon, W., and Horn, M.J. (2000), 14th Protein Society Symposium,<br />

{August}.<br />

P35-M<br />

Guanylation of lysines as a means to enhance confidence of protein<br />

identification in proteomics.<br />

S. Krishnan1, M. Lin2, K. Waddell1; 1Applied Biosystems, Foster City, CA,<br />

500 Old Connecticut Path, Framingham, MA 01701, 2Applied Biosystems,<br />

Framingham, MA<br />

The completion of several genomes, including that of the human, has shifted<br />

the emphasis to determining the proteome counterpart of biological systems.<br />

The current approach for such a proteomic analysis is the mass fingerprinting<br />

of tryptic digests of proteins (generated by ingel digest of 1 or 2-D gel<br />

spots or of chromatographic fractions) using MALDI-Tof mass spectrometry<br />

followed by database searching of the masses so obtained. It has recently<br />

been noticed that the arginine containing peptides ionize better under MALDI<br />

conditions compared to lysine containing peptides. We present here an<br />

approach to use this as an advantage in enhancing confidence of the database<br />

search matches by modifying the lysines to homoarginines. The increase<br />

in confidence is achieved due to increase in the number of peptides seen in<br />

the mass spectrum resulting from the guanylation of lysine e-amino group<br />

(addition of 42 Da) and also the fact that such a modification indicates the<br />

lysine containing peptides in the tryptic digest mix. Tryptic digests of BSA,<br />

Enolase, alpha lactalbumin, and cytochrome C were generated by routine<br />

procedures and mixed in various combinations. The digest mixtures were<br />

than analyzed by MALDI-Tof mass spectrometry followed by database searching<br />

of the peptide masses for identification of proteins. A portion of the digest<br />

mixture was then reacted with O-methyl isourea sulfate to convert the lysines<br />

to homoarginines. The modified digest mixtures were analyzed by MALDI-<br />

Tof mass spectrometry followed by database searching. The analysis before<br />

and after the modification helped identify the lysine containing peptides in<br />

the digest and hence a more comprehensive identification of the protein mixture.<br />

POSTER <strong>AB</strong>STRACTS<br />

196 JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000<br />

P34-S<br />

Approaches to validating SEQUEST database search results.<br />

R.E. Moore, M.K. Young, T.D. Lee; Beckman Res. Inst., City of Hope,<br />

1450 E. Duarte Rd., Duarte, CA 91010<br />

Tandem mass spectrometry and database searching is a very powerful<br />

approach to protein identification. Unfortunately, the wealth of data that<br />

makes the technique so powerful also requires extensive analyst time for data<br />

reduction and validation of search results. When analyzing complex samples,<br />

it can easily take more time to validate and reduce the data than it did to generate<br />

it. This time requirement is a serious problem because the technique<br />

is otherwise well suited to high throughput analysis.<br />

Two aspects of result validation are analyzed: validation of search results for<br />

individual tandem mass spectra and validation of composite results for sets<br />

of individual spectral matches. Statistical analysis and experiments with falsified<br />

databases demonstrate that there is a significant chance of generating<br />

multiple incorrect matches to the same protein when examining large data<br />

sets. This makes it imperative to validate the individual spectral matches, as<br />

simple reliance on multiplicity of matches to a single protein does not guarantee<br />

a correct result.<br />

Several approaches to validating individual spectral matches were tried and<br />

compared to the existing standard of manually comparing actual and predicted<br />

spectra. The most successful approach was to search each spectrum<br />

twice, once using tryptic specificity and once using no enzyme specificity,<br />

and correlate the results. When both searches generate the same result, that<br />

result is almost always validated by manual examination. When the non-specific<br />

search generated a higher cross-correlation score the result of the search<br />

using tryptic cleavage was almost never manually validated. Surprisingly,<br />

there were a large number of searches in which the search using tryptic<br />

cleavage resulted in a higher cross-correlation score than the search using<br />

non-specific cleavage. This third group yielded a moderate number of spectra<br />

that were manually validated.<br />

P36-T<br />

Automated analysis of single nucleotide polymorphism using mass<br />

spectrometry.<br />

M.S. Minkoff1, P. Ross1, L. Hall1, R. Jones2, D. Ledman1, L. Haff1; 1Applied<br />

Biosystems, 500 Old Connecticut Path, Framingham, MA 01701, 2Inst. for<br />

Child Hlth.<br />

Single nucleotide polymorphisms (SNPs) are increasingly employed as<br />

genetic markers for associated studies of many inherited diseases and traits.<br />

It is apparent that approximately 3 million putative SNPs will be found in the<br />

Human Genome. In order to tackle the task of validating the significance of<br />

any of these putative SNPs the chemistry and analytical platforms must meet<br />

demanding throughput and cost requirements. The SNP assay developed to<br />

meet these criteria incorporates the use of simple, inexpensive primers and<br />

unlabeled nucleotides for single base extensions, thus keeping down the cost<br />

of analysis. Multiplexing the analysis of SNPs enhances affordability of genotyping<br />

studies as well as significantly improving throughput. The use of<br />

high throughput automated Mass Spectrometry with integrated data acquisition,<br />

genotyping, sample handling software package simplifies and speedsup<br />

the task. In addition the top-level software application package tracks<br />

sample information throughout the workflow and stores all information in a<br />

central database. Data in support of multiplexed, automated sample handling,<br />

acquisition, analysis and reporting is presented.


P37-S<br />

Data-directed real-time instrument feedback and control:<br />

creation of dynamic peptide include and exclude lists from<br />

on-the-fly databank searching.<br />

A. Millar, J.B. Hoyes, R.A. Carruthers, C. Jones, A. Millar, C. Hughes,<br />

S. Leicester, J.I. Langridge; Micromass UK Ltd., Floats Road, Wythenshawe,<br />

Manchester M23 9LZ, United Kingdom<br />

An alternative approach to 2D gel electrophoresis for the qualitative analysis<br />

of complex protein mixtures is the use of tryptic digestion followed by<br />

electrospray LC-MS/MS. This approach has been shown to increase the<br />

dynamic range of protein identification and identify low copy number proteins.<br />

However there is often a large degree of redundant sequence information<br />

acquired, as in theory one peptide MS/MS spectrum is sufficient to<br />

identify a protein from a sequence databank. If a protein identification is<br />

obtained from a databank search of an MS/MS spectrum, excluding the rest<br />

of the theoretical tryptic peptides during an LC-MS/MS experiment may<br />

allow deeper “mining” into the protein complex being studied.<br />

We have introduced a new search engine capable of matching a tryptic peptide<br />

from the Swissprot/TrEMBL databank to an MS/MS spectrum in one second.<br />

Based upon these results, we are able to generate dynamic include or<br />

exclude lists, based upon the theoretical tryptic peptides from the identified<br />

protein. These can be passed to the acquisition software of our Q-Tof mass<br />

spectrometer in real time. Thus, we are able to automatically steer the Q-Tof<br />

during acquisition to select and switch to the MS/MS mode only on those<br />

peaks that meet the modified selection criteria. Therefore precursor ions that<br />

belong to a protein already identified during acquisition can be avoided. This<br />

exclusion list is based upon m/z, charge state and a user defined mass tolerance.<br />

Mass measurement on the Q-Tof 2 mass spectrometer is typically better<br />

than 10ppm and therefore a tight mass tolerance can be selected, making<br />

the exclude list extremely specific.<br />

To illustrate this methodology we present examples where Q-Tof data acquisition<br />

has been directed based upon the results from a databank search. This<br />

data will be compared and contrasted to data acquired in the normal automated<br />

LC-MS/MS mode.<br />

P39-T<br />

Automated software for identification and relative quantification<br />

of differentially expressed proteins from isotope coded<br />

affinity tag LC-MS data.<br />

A. Millar, P. Young, R. O’Malley, A. Millar, J.I. Langridge; Micromass UK<br />

Ltd., Floats Road, Wythenshawe, Manchester M23 9LZ, United Kingdom<br />

Whilst LC-MS/MS has been utilised for the identification of proteins from<br />

complexes and cell lysates (qualitative proteomics), the quantitative study of<br />

gene expression using differential display has until recently been the preserve<br />

of a 2D gel based proteomic experiment. However, recently a great deal of<br />

interest has been generated on the use of isotope coded affinity tags (ICAT)<br />

1 for the quantitative study of gene expression at the proteome level.<br />

The technique is based upon chemically modifying the cysteine residues of<br />

proteins isolated from cells in two different states with light and heavy isotopically<br />

labeled reagents. The two cell states are then combined, digested<br />

with trypsin and the cysteine containing peptides preferentially selected by<br />

binding to an avidin column, prior to analysis by mass spectrometry. The eluent<br />

from this column is then analysed by capillary LC ESI-MS/MS. Interrogation<br />

of the eluting peptides by tandem mass spectrometry and databank<br />

searching results in the identification of the associated protein.<br />

We describe how ICAT data analysis has been automated within a software<br />

environment. The MS and MSMS data acquired using the QTof instrument are<br />

processed and analysed using a new algorithm which recognises related isotope<br />

clusters and quantifies their relative intensities. Based on a user defined<br />

ratio threshold the software will automatically carry out an LC-MS/MS experiment<br />

and databank search in a client-server mode and provide a report of<br />

the identified proteins and their expression ratio in the two cell states.<br />

1. Gygi et al. Nature Biotechnology (1999) 17, 994–999.<br />

POSTER <strong>AB</strong>STRACTS<br />

<strong>AB</strong>RF 2001 <strong>AB</strong>STRACTS<br />

P38-M<br />

Routine identification of large (�5 kDa) peptides derived from<br />

biological sources using an ESI-quadrupol-TOF mass spectrometer.<br />

M. Kellmann, S. Neitz, M. Juergens; BioVisioN GmbH & Co KG,<br />

Hannover, Feodor-Lynen-Str. 5, Hannover, Lower Saxony 30625, Germany<br />

Investigating the peptide portion of a proteom results in a multitude of peptides/small<br />

proteins in the mass range from 5 to 15 kDa [1]. We developed a<br />

routine procedure for direct identification exploiting the relatively high parent<br />

and daughter ion resolution (�7500) and mass accuracy (�50 ppm) of<br />

a hybrid ESI-quadrupol-time-of-flight mass spectrometer. Data processing<br />

was made with automated charge state deconvolution based on Zhang and<br />

Marshall´s Zscore algorithm [2] and a deisotoping algorithm included in the<br />

Voyager 5.0 software from Applied Biosystems. Database searching was performed<br />

by the MASCOT search engine [3].<br />

We will illustrate that this procedure will lead to a fast and reliable identification<br />

of several large peptides in the mass range from 5 to 15 kDa derived<br />

from blood filtrate.<br />

[1] Kellmann, M, et al., Proceedings of the 48th Conference on Mass Spectrometry<br />

and Allied Topics, 2000, Long Beach<br />

[2] Zhang, Z and Marshall, AG. A universal algorithm for fast and automated<br />

charge state deconvolution of electrospray mass-to-charge ratio spectra. J Am<br />

Soc Mass Spectrom 9(3): 225–33 (1998).<br />

[3] Perkins, DN, Pappin, DJ, Creasy, DM and Cottrell, JS, Probability-based<br />

protein identification by searching sequence databases using mass spectrometry<br />

data. Electrophoresis 20(18) 3551–67 (1999).<br />

P40-S<br />

New method for preparation of DNA for MALDI-MS analysis.<br />

I.P. Smirnov, L.A. Haff, P.L. Ross, L.R. Hall, D.W. Ledman;<br />

Applied Biosystems, 500 Old Connecticut Path, Framingham,<br />

MA 01701<br />

In recent years, matrix assisted laser desorption ionization mass spectrometry<br />

(MALDI MS) became one of the main analytical tools for short DNA fragments<br />

and synthetic oligonucleotides. The application of mass spectrometry<br />

for genetic analysis (such as SNP typing) offers, compared to other methods,<br />

higher sensitivity, higher accuracy and lower costs by removing the necessity<br />

for fluorescent- labeled primers. Still, the susceptibility of MALDI-MS to<br />

the presence of salts of alkali metals requires careful and often tedious<br />

desalting procedures which complicates and slows the throughput of the MS<br />

based methods. To overcome that obstacle we have developed a novel<br />

approach to sample preparation. The idea is to extract DNA out of the solution<br />

on a solid surface coated with an anion exchange polymer (e.g. polyethyleneimine).<br />

The observed binding is strong enough to remove and concentrate<br />

DNAs from buffer solutions and hold them on the plate through<br />

extensive washing procedures if necessary. The properties of anion exchange<br />

films make possible to perform purification, desalting and concentration in<br />

a single fast step. After the DNA is immobilized on the surface and supernatant<br />

solution is removed, subsequent addition of MALDI matrix releases<br />

material from the surface, which co-crystallizes with matrix. The MS analysis<br />

can then be performed directly from that support. Such surfaces may be<br />

obtained upon glass, plastic, or metal MALDI plates. Analysis of oligonucleotides<br />

and complex multiplexed SNP typing reactions (Sequazyme-Pin-<br />

Point) performed by this method shows greatly improved sensitivity and<br />

excellent resolution for wide range of DNA lengths, together with high tolerance<br />

to various buffers components, such as alkali metals and surfactants.<br />

The importance and role of different factors have been studied, such as composition<br />

of ion exchange film and compatibility of the MALDI matrix in<br />

order to obtain the best performance.<br />

JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000 197


<strong>AB</strong>RF 2001 <strong>AB</strong>STRACTS<br />

P41-M<br />

Review of gel- and column-based separation technologies for<br />

the identification of yeast proteins using MS/MS.<br />

V.C. Wasinger, G.L. Corthals; Garvan Inst. of Med. Res., 384 Victoria St.,<br />

Sydney, NSW 2010, Australia<br />

Proteome analysis involves the identification and quantitation of expressed<br />

proteins by a given cell type, tissue or organism and most commonly involves<br />

2-dimensional electrophoresis (2-DE) followed by mass spectrometry (MS)<br />

for protein separation and identification. While 2-DE is unequalled in its ability<br />

to separate and resolve thousands of proteins in complex solutions, we<br />

have recently shown that many proteins of biological significance lay beyond<br />

the detection limits of 2-DE. The application of 2-D gels becomes ineffective<br />

when proteins co-migrate to the same grid coordinates, merge to become<br />

one large spot or are not displayed because their concentration is simply too<br />

low for visualisation and subsequent biochemical MS analysis.<br />

Two important biological aspects prevent us from fully exploiting the power<br />

of the 2-DE technology: 1) Firstly, protein expression exists for up to 12<br />

orders of magnitude (in serum). This broad range of expression critically limits<br />

the current 2-DE approach, as at best only 5 orders of magnitude difference<br />

in expression can be displayed; 2) “functional” proteins operate in association<br />

with other proteins; such that research must be directed toward the<br />

analysis of multiply interacting proteins or functional modules that regulate<br />

complex biological networks and pathways.<br />

Alternative fractionation methods such as chromatography exist and allow<br />

proteins/peptides to be separated based on similar (charge, size) or alternative<br />

(affinity, hydrophobicity) properties to 2-DE. Used in series with MS,<br />

these technologies have the potential to fractionate large numbers of proteins<br />

and peptides to enable the comprehensive, comparative and relational analysis<br />

of proteins. Yeast is an ideal choice for this study as both genome, proteome<br />

and transcriptome data is available for evaluation.<br />

A comparison of the separation potential based on protein numbers, protein<br />

classes and properties are described using 2-D gel electrophoresis, size<br />

exclusion and cation exchange chromatography followed by protein identification<br />

using ESI-MS/MS.<br />

P43-S<br />

Characterization of differential protein expression between wild<br />

type and Rce1 knockout mouse embryonic fibroblasts using<br />

2-D SDS PAGE and MALDI TOF mass spectrometry.<br />

S.C. Hall1, D. Smith-Beckerman2, M. Lobo2, S.G. Young3; 1Applied<br />

Biosystems, 850 Lincoln Centre Drive, Foster City, CA 94404,<br />

2San Francisco State Univ., 3UCSF<br />

Ras proteins play an important role in transmitting growth signals from membrane<br />

receptors to the nucleus, triggering the transcription of genes involved<br />

in cellular proliferation. Ras proteins are membrane bound, guanine<br />

nucleotide-binding proteins with GTP-ase activity. Many human cancers contain<br />

mutationally activated Ras proteins that transmit growth signals in an<br />

uncontrolled manner, triggering neoplastic transformation and uncontrolled<br />

cellular proliferation. The proper intracellular location of the Ras proteins<br />

depends on a series of posttranslational modifications: isoprenylation of a Cterminal<br />

cysteine, endoproteolytic release of the carboxyl-terminal three<br />

amino acids, and methyl esterification of the carboxyl-terminal isoprenylcysteine.<br />

Each of these processing steps represents a potential target for cancer<br />

therapy. The endoproteolytic cleavage step is carried out by the product<br />

of the Rce1 gene. Recently, the Rce1 gene was inactivated in mice, completely<br />

blocking the endoproteolytic processing of the Ras proteins. We used<br />

2D-PAGE to assess differences in protein expression in primary embryonic<br />

fibroblasts from wild-type and Rce1 knockout mice. First, we wanted to<br />

detect substrates for Rce1. Altered electrophoretic mobility of individual protein<br />

spots on the gel indicated the possibility of aberrant post-translational<br />

processing. Second, we wanted to detect proteins whose expression level<br />

was affected by the Rce1 knockout mutation. MALDI TOF mass spectrometry<br />

was used to generate peptide mass fingerprints to identify several proteins<br />

having identical molecular weight, pI, and relative abundance in both wildtype<br />

and Rce1 knockout mice. Additional confirmation of the identities of<br />

these proteins was obtained by performing post-source decay analysis on<br />

selected tryptic peptides. It was important to identify these “marker” proteins<br />

as they will be used as migration reference points when comparing future<br />

2D-gel separations. Furthermore, they permitted optimization of protocols for<br />

the MS analysis of Ras proteins from wild-type and Rce1 knockout fibroblasts.<br />

POSTER <strong>AB</strong>STRACTS<br />

198 JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000<br />

P42-T<br />

Quantitative analysis of tumor antigens by mass spectrometry.<br />

A. Kishiyama, D. Arnott; Genentech, 1 DNA Way, South San Francisco,<br />

CA 94080<br />

Both genomics and proteomics are useful for comparing gene expression<br />

patterns. The discovery of receptors overexpressed on tumor cells has lead<br />

to effective cancer therapies, for example. Techniques such as DNA arrays<br />

can be used to measure mRNA levels with great sensitivity and speed. More<br />

detailed information can be obtained through 2D gels, western blotting, and<br />

immunohistochemistry, but these protein-based approaches are time-consuming,<br />

prone to biases, or require generating antibodies. A method has<br />

therefore been developed, called the mass western experiment in analogy to<br />

the western blot, to better bridge genomics and proteomics.<br />

In this experiment, a variation on the ICAT experiment described by Gygi et<br />

al. (Nature Biotech. 1999 v.17 p.994), specific proteins (such as those found<br />

to be of interest from DNA array experiments) are detected and compared<br />

between samples. Proteins extracted from two samples are labeled with a<br />

custom ICAT reagent. The samples are mixed, digested, and the labeled peptides<br />

collected. LC-MS/MS is performed on an ion trap instrument; anticipated<br />

tryptic peptides from the protein of interest are continuously subjected to<br />

CID. Heavy and light ICAT-labeled peptides are simultaneously trapped and<br />

fragmented. Both identification and quantitation is thus obtained in one<br />

experiment.<br />

This approach has been validated by the comparison of cell lines expressing<br />

known tumor antigens in different amounts. Overexpression of the<br />

receptor Her-2 in breast cancer cell lines was shown, and by factors in<br />

agreement with other measurements. Other potential tumor antigens have<br />

likewise been detected.<br />

P44-M<br />

Intelligent data acquisition and automated sample analysis via<br />

orthogonal MALDI- QqTOF, a new tool for protein identification.<br />

C.M. Lock; MDS-Sciex, 71 Four Valley Drive, Concord, Ontario<br />

L4K 4V8, Canada<br />

The application of a novel UV-MALDI ionisation source coupled to an<br />

Applied Biosystems/MDS-Sciex QSTAR Pulsar QqTof mass spectrometer for<br />

protein sequencing and identification is described. The coupling of these two<br />

devices enables collision induced dissociation spectra of singly charged<br />

MADLI ions to be generated, with all the associated QqTof benefits of high<br />

mass accuracy and resolution. The inherent pulsed nature of the o-MALDI<br />

source is converted into a pseudo continuous beam of ions by collisional<br />

cooling in the Q0 region. The o-MALDI source is completely decoupled from<br />

and has no influence on the orthogonal Tof analyser.<br />

High mass accuracy and resolution is thus maintained simultaneously over<br />

the full mass range when switching between MS and MS/MS modes as<br />

opposed to conventional MALDI post source decay experiments.<br />

The application of the technique to the analysis of low femtomole unseparated<br />

protein tryptic digests is demonstrated using an automated data acquisition<br />

approach. The software developed enables the intelligent acquisition<br />

of data from sample plates with minimal user intervention.<br />

The high speed data acquisition capabilities of the o-MALDI source in combination<br />

with the high performance of the QqTof offers unique possibilities<br />

for rapid identification of proteins. Rapid analysis and identification of proteins<br />

via a peptide-mass fingerprinting approach and MS/MS sequence information<br />

will be shown.


P45-T<br />

Identification of changes in protein expression in Deinoccocus<br />

radiodurans using isotope-coded affinity tags.<br />

E.A. Panisko1, T.P. Conrads1, T. Veenstra1, L. Pasa-tolic1, G.A. Anderson1, R. Aebersold2, R.D. Smith1; 1Pacific Northwest Natl. Lab., P.O. Box 999,<br />

K8-98, Richland, WA 99352, 2Univ. of Washington<br />

The isotope-coded affinity tag (ICAT) strategy was used to identify the<br />

effects of irradiation on relative protein abundances in the highly radioresistant<br />

organism, Deinococcus radiodurans (D. radiodurans). The ICAT strategy<br />

uses two distinct isotopic versions of a cysteine-specific reagent; a “light”<br />

version and a “heavy” version in which eight hydrogen atoms are substituted<br />

for deuterium atoms. Proteome samples extracted from the organisms just<br />

prior to, as well as 30 min. and 3 hours after treatment with ionizing radiation<br />

were labeled with either the heavy or light isotopic version of the<br />

ICAT reagent. After mixing the samples in various combinations the cysteine<br />

containing polypeptides (Cys-polypeptides) were extracted using immobilized<br />

avidin chromatography. The Cys-polypeptides were initially identified<br />

by capillary reverse phase liquid chromatography (LC) using a<br />

conventional mass spectrometer (MS) operating in the tandem mass spectrometry<br />

(MS/MS) mode. The masses of the identified peptides were subsequently<br />

identified in a capillary LC separation coupled on-line with Fourier<br />

transform ion cyclotron resonance (FTICR) MS. Changes in relative protein<br />

expression were measured using the results obtained by LC/FTICR. The<br />

results show that most proteins undergo a decrease in expression when D.<br />

radiodurans is subjected to ionizing radiation.<br />

P47-M<br />

Use of stable isotope amino acid labels to simplify MS/MS peptide<br />

analysis.<br />

S.J. Berger, S-W. Lee, Y. Shen, G.A. Anderson, R.D. Smith; Pacific<br />

Northwest Natl. Lab., 902 Battelle Blvd. Box 999 MS: K8-98,<br />

Richland, WA 99352<br />

Tandem mass spectrometry (MS/MS) is a primary analysis tool for the identification<br />

of peptides from proteolytic sample digests. When coupled with<br />

online separations, one or more peptides from a single 2D-gel spot digest can<br />

uniquely identify a given protein, or more complex digests can yield a global<br />

view of the proteome. The identification of individual fragment ions from a<br />

parent peptide is indirect, and the resulting peptide identification is strictly<br />

correlative. The ability to add sequence and composition constraints to<br />

assign peptide identity should significantly narrow the possible assignments<br />

of ions, and simplify interpretation of MS/MS spectra. Here we describe an<br />

application of stable isotope labeling in conjunction with MS/MS analysis of<br />

proteolytic digests that permit significant improvements assigning ions in the<br />

resultant MS/MS spectrum. Auxotrophic cells grown in parallel in the presence<br />

of a labeled or unlabeled amino acid are combined prior to cell disruption.<br />

Peptides derived from proteolytic digests generate pairs of peaks<br />

separated by a known mass difference during an initial LC/MS scan. These<br />

identified peptide pairs are isolated, and analyzed by LC/MS/MS in subsequent<br />

scans. When using a protease that cuts adjacent to the labeled amino<br />

acid, the paired/unpaired daughter ion pattern permits simplified identification<br />

of the parent peptide.<br />

This work was supported by OBER (U.S. DOE) and PNNL Laboratory<br />

Directed R&D. Battelle Memorial Institute operates PNNL for the U.S. DOE<br />

under Contract DE-AC06-76RLO 1830.<br />

POSTER <strong>AB</strong>STRACTS<br />

<strong>AB</strong>RF 2001 <strong>AB</strong>STRACTS<br />

P46-S<br />

Quantification of neurosteroids using NCI GC/MS.<br />

R.L. Fitzgerald; VA Med. Ctr., UCSD, VAMC-113, 3350 La Jolla Village Dr.,<br />

San Diego, CA 92161<br />

Electron capture negative chemical ionization (NCI) is one of the most sensitive<br />

ionization techniques available and is especially well suited for quantitative<br />

analysis of target compounds in biological extracts. For the analysis<br />

of small molecules, NCI is often several orders of magnitude more sensitive<br />

than traditional techniques such as electron ionization or positive chemical<br />

ionization. There are several important prerequisites for performing quantitative<br />

analysis using NCI, including an electronegative functional group, stable<br />

isotopic internal standards, and good method validation. We synthesized<br />

deuterium labeled analogs of neurosteroids and developed a NCI GC/MS<br />

method for quantification of neurosteroids in biological samples using isotope<br />

dilution.<br />

Neurosteroids have distinct neurotransmitter mediated effects and consequently<br />

it is important to be able to identify and quantify individual compounds.<br />

Previously, the determination of neurosteroids in biological matrices<br />

involved complicated purification protocols or did not use appropriate<br />

internal standards. We added deuterium-labeled internal standards to brain<br />

(100 mg of cortex homogenate) or plasma (300 �L). Samples were homogenized<br />

in methanol, centrifuged and diluted to contain 5% methanol and then<br />

applied to C-18 columns. After washing the column with methanol/water<br />

(50/50), steroids were eluted with methanol. Following evaporation, steroids<br />

were converted to pentafluorobenzyl oxime/trimethylsilyl ether derivatives.<br />

The extracts were analyzed using SIM. The present method allows simultaneous<br />

quantification of pg amounts (100 pg in 300 �L of plasma and 250 pg<br />

in 100 mg of brain tissue) of neurosteroids and will be helpful in elucidating<br />

the role of neurosteroids in health and disease.<br />

P48-T<br />

A comprehensive proteomic analysis of human cilia using<br />

nanoscale capillary LC/MS/MS.<br />

M. Moyer1, K. Blackburn1, W. Burkhart1, A. Moseley1, L. Ostrowski2, R. Boucher2; 1Glaxo Wellcome, 5 Moore Drive, Research Triangle Park,<br />

NC 27709, 2Univ. of North Carolina<br />

Ciliated cells play an integral role in the defense mechanisms of the respiratory<br />

system. By the coordinated beating of their cilia they provide the force<br />

necessary to clear potentially harmful material from the airways. In order to<br />

better understand the protein composition of cilia, human cilia were subjected<br />

to a comprehensive proteomic analysis. Cilia were isolated from cultures<br />

of airway epithelial cells and component proteins separated by 1D or<br />

2D gel electrophoresis. Bands or spots were excised, subjected to in-gel proteolytic<br />

digestion, and component proteins identified by nanoscale capillary<br />

LC/MS/MS. Alternatively, proteolytic digests of intact cilia were analyzed<br />

directly by nanoscale capillary LC/MS/MS with or without the use of cysteinespecific<br />

affinity tags. Data will be presented on proteins identified as components<br />

of cilia as well as details of the analytical methodologies.<br />

JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000 199


<strong>AB</strong>RF 2001 <strong>AB</strong>STRACTS<br />

P49-S<br />

4-(4-Hydroxystyryl)pyridine, a rationally designed negative ion<br />

matrix for MALDI-TOF mass spectrometry.<br />

J.C. Lopez, P.A. Liddell, J.W. Lippert, D.C. Brune; Arizona State Univ.,<br />

PO Box 871604, Tempe, AZ 85287-1604<br />

To date, most matrices for MALDI-TOF mass spectrometry have been discovered<br />

by trial and error experiments, and through broad investigation of<br />

compounds chemically related to known matrix molecules. The most widely<br />

used matrices have been used to analyze proteins and other analytes in the<br />

positive ion mode. We chose to work with negative ion matrices because this<br />

area has received less attention, and because we occasionally analyze samples<br />

for which a good negative ion matrix is required. In this investigation,<br />

we synthesized and studied 4-(4-hydroxystyryl)pyridine. This compound<br />

resembles those derived from 4-hydroxycinnamic acid in having an electronwithdrawing<br />

moiety (in this case a pyridyl group) separated from a 4-hydroxyphenyl<br />

moiety by a vinyl group. In the electronically excited state, charge<br />

density delocalized across the vinyl group onto the pyridyl group will make<br />

the latter more strongly basic, favoring abstraction of protons from both the<br />

analyte and other matrix molecules. Preliminary experiments on Fmoc and<br />

other modified amino acids have shown that this matrix works as well as<br />

3-aminoquinoline, one of the best negative ion matrices reported so far. Further<br />

comparisons involving structurally related matrix molecules are in progress<br />

and will be reported. A characteristic for negative ion matrices is a shift<br />

to longer wavelengths under acidic conditions.<br />

P51-T<br />

Advances in quantitation and identification of proteins using<br />

isotope-coded affinity tags and LC/MS/MS using informationdependent<br />

acquisition.<br />

T.A. Settineri, C.L. Hunter, L. Nuwaysir, A. Patel, A. Tomaney, B. Halpern;<br />

Applied Biosystems, 850 Lincoln Ctre. Dr., Foster City, CA 94404<br />

Recently, Aebersold et al. introduced a method in which proteins are labeled<br />

with an isotope-coded affinity tag (ICAT), digested, and the resulting labeled<br />

peptides separated and analyzed by mass spectrometry. The advantage to<br />

this technique is that the ICAT approach precludes the use of 2D gels while<br />

allowing complete automation of the entire process. In addition, the ICAT<br />

technique is generally applicable and can be applied to the global analysis<br />

of protein expression. In this study we investigate the ICAT approach using<br />

an API-QSTAR (hybrid quadrupole time-of-flight) mass spectrometer fitted<br />

with a nano-electrospray source, ICAT reagents synthesized in-house, and<br />

novel data reduction algorithms to both quantitate protein expression and<br />

identify proteins from MS/MS data acquired by using information dependent<br />

acquisition (IDA)<br />

This approach will be demonstrated for the analysis of proteins arising from<br />

various cell lines including E. coli and Arabidopsis treated in different ways.<br />

After treatment with ICAT reagent and purification, samples are analyzed by<br />

LC/MS/MS using a QSTAR mass spectrometer using information dependent<br />

acquisition (IDA). Two new algorithms developed to identify and quantitate<br />

ICAT expression pairs are applied to the MS scans while a third new algorithm<br />

is applied to the MS/MS data to identify proteins. The first two algorithms<br />

cluster the data based on ICAT fragments, and optimally collapse adjacent<br />

spectra for maximum signal to noise. The third algorithm then identifies<br />

the protein using the MS/MS fragment ion data to perform a database search.<br />

POSTER <strong>AB</strong>STRACTS<br />

200 JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000<br />

P50-M<br />

LC-MS of oligonucleotides using polystyrene supports and<br />

API-TOF mass spectrometry.<br />

D.H. Hawke1, J.S. Nelson2, R. Vinayak2, W. Xiao3, P.J. Oefner3; 1Applied<br />

Biosystems, 850 Lincoln Ctr. Dr., Foster City, CA 94404, 2Applied<br />

Biosystems, 3Stanford Genome Res. Ctr.<br />

Oligonucleotides may be analyzed by either Maldi or electrospray methods<br />

after suitable desalting of samples. Maldi methods are convenient and offer<br />

high-speed analysis, while electrospray methods can be directly coupled to<br />

HPLC separations. A classical application for these methods is oligonucleotide<br />

synthesis, in this case there is usually enough material available that<br />

sensitivity is not a major problem. LCMS (1) is advantagous if UV detection<br />

is required, and desalting is effectively performed on-line. A newer area is<br />

that of genotyping. In this application separation may be required prior to<br />

analysis and current ion-pairing methods have remarkable separating power<br />

for these molecules (2). We have coupled this separation technology to a<br />

high-resolution API-TOF MS system (Mariner) to give both high resolution<br />

separations and high mass accuracy measurements of oligonucleotides and<br />

labeled oligos.<br />

1. Huber C.G., Krajete A., Anal. Chem. (1999), 71:3730–3739.<br />

2. Underhill, P.A., L. Jin, A.A. Lin, S.Q. Mehdi, T. Jenkins, D. Vollrath, R.W.<br />

Davis, L.L. Cavalli-Sforza and P.J. Oefner. Genome Res. (1997), 7:996–1005.<br />

P52-S<br />

High-throughput SNP genotyping by MALDI-TOF MS.<br />

M. Kostrzewa1, T. Fröhlich1, T. Wenzel1, C. Franke1, W. Pusch2, K-O. Kräuter2, Y. Stalgies2; 1Bruker Saxonia Analytik GmbH, Permoserstrasse<br />

15, Leipzig, Saxonia D-04318 Germany, 2Bruker Daltonik GmbH,<br />

Bremen<br />

One of the great challenges of the upcoming postgenomic era is the determination<br />

of sequence variations, in particular single nucleotide polymorphisms<br />

(SNPs). These polymorphisms are believed to have an enormous<br />

impact in diagnosis of diseases, improvement of drugs, and forensic analysis<br />

in future. In contrast to the increasing demand for SNP genotyping, there<br />

is still a lack of highly reliable high-throughput SNP typing techniques.<br />

Matrix-assisted laser desorption/ionization time-of-flight mass spectrometry<br />

(MALDI-TOF MS) is a powerful advanced technology capable of the accurate<br />

and cost-effective typing of many thousands of SNPs per day. The “GOOD<br />

assay”, a novel sample preparation procedure for MALDI-TOF typing of<br />

SNPs was recently presented (Sauer et al., Nucleic Acids Research, methods<br />

online, 2000, 28, e13). This preparation uses sensitivity enhancing, chemical<br />

modifications of the allele specific products. Main advantage of the GOOD<br />

assay over other SNP genotyping preparation methods with MALDI detection<br />

is that this protocol does not require solid phase purification of the products<br />

prior MALDI analysis. SNP typing starts with a PCR encompassing a region<br />

containing a known SNP. Thereafter dNTPs are digested with shrimp alkaline<br />

phosphatase. In the following primer extension reaction using a charge tag<br />

containing extension primer, a conditioned set of ddNTPs and a DNA polymerase<br />

of allele specific products are generated. The 5� part of the extension<br />

primer is removed by an exonuclease. By alkylation the backbone phosphate<br />

groups are neutralised. This results in small singly charged product molecules<br />

which are transferred onto non-protonating matrix on the MALDI target.<br />

Mass spectra of 384 samples were acquired in less than one hour. Allele calling<br />

was performed automatically by the novel genotools SNP manager software,<br />

online during spectra acquisition or offline. The resulting genotypes are<br />

stored in a table in the ASCII format together with a quality asse.


P53-M<br />

Nano-LC ion-trap mass spectrometry for the investigation of<br />

low-fmol protein amounts.<br />

U. Schweiger-Hufnagel1, A. Schneider1, A. Ingendoh1, S. Liedtke2, R. van Soest3; 1Bruker Daltonik GmbH, Bremen, Fahrenheitstr. 4,<br />

Bremen, Bremen 28359 Germany, 2Bai GmbH, 3LC Packings NL<br />

Ion trap mass spectrometry is a powerful tool for the proteomic research<br />

since it is predestined for the connection to a liquid chromatography system,<br />

which allows the simple fragmentation of all separated peptides in one<br />

experiment. However, the limiting factor for the protein analysis so far was<br />

the chromatography, which, due to the high inner column diameter and high<br />

flow rates reduced the detection limit of the mass spectrometer. To improve<br />

this, we connected a nano-LC system to the ion trap mass spectrometer to<br />

investigate the applicability of this setup for the proteomic research.<br />

The peptides were separated on a 75 �m ID RP column at a flow rate of 160<br />

nl/min. To achieve the LC-MS coupling fused-silica spray needles in combination<br />

with interfaces that support the use of peek based HPLC fittings (New<br />

Objective, USA) were used. The ion trap mass spectrometer was equipped<br />

with an online-nanospray device.<br />

Using this setup, a detection limit in the low fmol range is achieved (about<br />

1/100 of the detection limit for a capillary HPLC column). Thus, this powerful<br />

technique can be applied to those protein amounts which are provided<br />

by the proteome researcher (e.g. obtained from 2-D gels). Therefore, the<br />

nano-LC ion trap mass spectrometer meets the sensitivity requirements of<br />

proteomic research, and it enables the use of the ion trap for diverse analytical<br />

questions. Those include protein identification, the analysis of protein<br />

mixtures, the analysis of post-translational modifications and the sequence<br />

analysis.<br />

P55-S<br />

ESI-ion trap mass spectrometry for the analysis of PCR products.<br />

S. Hahner, A. Schneider, A. Ingendoh; Bruker Daltonik GmbH, Bremen,<br />

Fahrenheitstr. 4, Bremen, HB 28359 Germany<br />

The polymerase chain reaction (PCR) is an important tool used in many<br />

applications in molecular biology for the generation and amplification of specific<br />

DNA sequences from very small sample material. PCR products are commonly<br />

analyzed by means of gel electrophoresis. Major disadvantage of this<br />

technique is that the size of a PCR product is determined by comparison of<br />

the relative electrophoretic mobility in the gel matrix to an internal size<br />

standard. In addition, a labelling of the analyte is required for detection.<br />

Mass spectrometry (MS) has been proven to be a useful technique for the<br />

determination of both the length and sequence variations of PCR products<br />

based on the accurate determination of molecular weights. So far, PCR products<br />

have been analyzed using either matrix-assisted laser desorption-<br />

(MALDI) or electrospray ionization- (ESI) MS. The performance of both techniques<br />

is less labour- and time consuming compared to gel electrophoretic<br />

techniques. However, the application of MS is mainly limited due to the tendency<br />

of the DNA molecules for the formation of multiple salt adducts which<br />

complicate the data.<br />

The development of a purification approach based on the adsorption of PCR<br />

products on magnetic particles has been found to be efficient for purification<br />

of PCR products prior to MS. In addition, this approach is amenable to full<br />

automated handling. The application of the magnetic bead purification technique<br />

in combination with ESI ion trap MS yields mass spectra with a resolution<br />

and accuracy sufficient to assign single nucleotide sequence deviation<br />

in PCR products up to a size of 90 bp.<br />

POSTER <strong>AB</strong>STRACTS<br />

<strong>AB</strong>RF 2001 <strong>AB</strong>STRACTS<br />

P54-T<br />

Characterization of the proteome of Oryza sativa.<br />

P.A. Haynes, A. Koller, N. Andon, J. Wei, J.R. Yates, III; Novartis Agr.<br />

Discovery Inst., San Diego, 3115 Merryfield Row, San Diego, CA 92121<br />

As part of our ongoing efforts to enhance our understanding of plant biology<br />

in cereal crops, we have undertaken the characterization of the proteome<br />

of the major tissues and subcellular fractions of Oryza sativa (Rice). The tissues<br />

analyzed include leaf, root and seed. These were chosen because,<br />

despite certain similarities, each tissue performs very different functions in the<br />

plant life cycle and interacts with unique environmental factors. Further fractionation<br />

of leaf tissue has allowed the analysis of organelles including<br />

nuclei, chloroplasts and mitochondria, and subcellular fractions including<br />

cytosol and membrane preparations. Further fractionation of seed has allowed<br />

the analysis of endosperm and embryo fractions. Protein extracts<br />

were prepared from each sample, and proteins were visualized by two<br />

dimensional electrophoresis using immobilized pH gradient–SDS-PAGE. Proteins<br />

were then excised from the gel and digested in-gel with trypsin. Peptide<br />

mixtures were separated and analyzed using liquid chromatography–<br />

tandem mass spectrometry, and peptides were identified using computer<br />

database searching of uninterpreted fragment ion mass spectra.<br />

We have identified several hundred proteins by searching our data against<br />

publicly available sequence data from various plant species. These include<br />

proteins common to several tissues as well as those which are unique to a<br />

particular tissue. This data represents the most comprehensive picture to date<br />

of the proteome of tissues and organelles from cereals, and in particular rice<br />

plants. The knowledge of the identity and distribution of expressed proteins<br />

will add value to our genomic sequencing efforts, and will be used to provide<br />

a baseline for future studies of which proteins are up- or down-regulated<br />

in response to environmental stress.<br />

P56-M<br />

Hydrophilic anchors enable high throughput protein identification<br />

in the subfemtomole range.<br />

P. Hufnagel, M. Schürenberg, C. Köster, K-O. Kräuter; Bruker Daltonik<br />

GmbH, Bremen, Fahrenheitstr. 4, Bremen, Bremen D-28359, Germany<br />

Research strategies in the Proteomics field rely on both high sensitivity and<br />

high throughput spectra acquisition. In addition, there is a strong need for<br />

convenient analysis tools that allow high throughput processing, database<br />

searches and the assessment of a high number of search results.<br />

Hydrophobically coated targets equipped with small hydrophilic anchors<br />

have shown 10–100-fold higher sensitivity for MALDI TOF MS than conventional<br />

steel targets. A droplet (0.5–3 �l) containing matrix and analyte is<br />

applied onto a hydrophilic anchor and shrinks down to the anchor’s size,<br />

thereby restricting the formation of matrix crystals to an exactly defined<br />

area. This drying process increases the concentration of the analyte by a factor<br />

that depends on the droplet’s and on the anchor’s size. The fact that the<br />

matrix crystals are restricted to the area of the anchor is a big advantage of<br />

anchor targets when it comes to automation.<br />

Using an eight-channel pipetting robot for sample preparation (approx. 2 sec/<br />

sample) arrays of hydrophilic anchors (as well as standard metal targets) were<br />

loaded with protein digests of varying sample amounts. The combination of<br />

automatic spectra acquisition with batchwise database searching led to an<br />

overview of the sensitivity increase (a factor of approx. 100) achieved by the<br />

anchor technology. The database search results have been automatically<br />

assessed by a fuzzy-control-based algorithm.<br />

Here, high throughput capabilities of preparation, acquisition and analysis<br />

have been used to compare the sensitivity of standard sample preparation<br />

with the hydrophilic anchor technology on a statistical basis. Furthermore,<br />

this analysis serves as a model for the application of these fundamental<br />

techniques for the fast achievement and analysis of search results coming<br />

from proteomic research tasks.<br />

JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000 201


<strong>AB</strong>RF 2001 <strong>AB</strong>STRACTS<br />

P57-T<br />

Optimization of a procedure for labeling proteins with isotopecoded<br />

affinity tags.<br />

K. Parker, S. Pillai, S. Daniels, W. Stanick, R. Lotti, B. Purkayastha,<br />

T. Nadler; Applied Biosystems, 500 Old Connecticut Path, Framingham,<br />

MA 01701<br />

The use of the isotope coded affinity tag (ICAT), developed by the laboratory<br />

of R. Aebersold (Nat. Biotechnol. 1999 Oct 17(10):994–9), allows one<br />

to compare the relative protein abundance between two samples by mass<br />

spectrometry. The reagent used is essentially cysteine reactive biotinylation<br />

reagent into which stable isotopes have been incorporated. One of the two<br />

samples is treated with the light reagent while the other sample is treated<br />

with the same reagent containing 8 deuterium atoms such that the samples<br />

are chemically equivalent but differ only in mass. In this way two peptides<br />

may be monitored in the mass spectrometer and quantified relative to one<br />

another by peak intensity.<br />

A number of imporant questions concerning the use of this technique are<br />

addressed in this presentation. The reagent has been extensively characterized<br />

and the recommended protocol for its use has been optimized for ease<br />

of use and robustness. Experiments were carried out to determine the purity,<br />

stability, and reaction kinetics of the reagent. Additional experiments were<br />

performed to optimize the derivatization protocol, including measurements<br />

of the degree of alkylation obtained, determining the optimal reduction<br />

reagent, how to remove excess reagent, and measurements of stability,<br />

capacity, carryover, and recovery of biotinylated peptides from monomeric<br />

avidin columns, Finally, the reproducibility of quantitative measurements in<br />

the mass spectrometer was addressed and will be presented.<br />

P59-M<br />

Quantitative proteomic and mRNA expression analysis of<br />

S. cerevisiae via metabolic labeling and multidimensional<br />

protein identification technology.<br />

M.P. Washburn1, G. Oshiro2, D. Wolters1, D. Schieltz1, C. Deciu1, E. Winzeler2, J.R. Yates, III1; 1Novartis Agr. Discovery Inst., 3115 Merryfield<br />

Row Suite 100, San Diego, CA 92121, 2Genomics Inst. of Novartis Fndn.,<br />

San Diego<br />

Proteomic technologies are being developed to determine large-scale<br />

changes in protein expression levels. Several quantitative proteomic methods<br />

have recently been published where the protein expression levels are compared<br />

between two different growth conditions by metabolic isotopic labeling<br />

of proteins or post-growth isotopic labeling of proteins in a sample. To<br />

fully understand how growth conditions effect biological systems, both protein<br />

and mRNA expression levels must be analyzed. We have applied metabolic<br />

labeling strategies to S. cerevisiae in order to determine the changes in<br />

protein and mRNA expression levels. S. cerevisiae was grown in 15N enriched<br />

minimal media and compared to S. cerevisiae grown in rich media. Each sample<br />

was analyzed via mRNA expression array and multidimensional protein<br />

identification technology. By combining these methods we were able to correlate<br />

the changes in protein and mRNA expression levels of several hundred<br />

gene products.<br />

POSTER <strong>AB</strong>STRACTS<br />

202 JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000<br />

P58-S<br />

Rapid genotyping using spectrometrically monitored selections.<br />

J. Stoerker1, J.D. Mayo2, C.N. Tetzlaff3, D.A. Sarracino4, C. Richert3; 1Bruker Daltonics, 15 Fortune Drive, Billerica, MA 01821, 2Tufts Univ.,<br />

3Univ. of Konstanz, 4Variagenics, Inc.<br />

Presented here is an inexpensive and rapid method for genotyping single<br />

nucleotide polymorphisms (SNPs) on PCR products using spectrometrically<br />

monitored selection from probe libraries. We have previously reported nuclease<br />

selections of modified oligonucleotides with increased RNA or DNA targets.<br />

This methodology relies on the selection of short hybridization probes<br />

complementary to the target sequence with a single-strand specific nuclease<br />

that does not attack duplexes. Conditions are described where the matched<br />

duplex is profoundly better protected from enzymatic digestion than single<br />

base mismatches. Semi-quantitative MALDI-TOF MS is used to identify the<br />

selected probes after a 10-minute digestion reaction with snake venom phosphodiesterase.<br />

We have examined sequences from exon 10 of the cystic<br />

fibrosis transmembrane regulatory (CFTR) gene, using a probe library representing<br />

a number of alleles, including: native, I506S, 1651 benign polymorphism,<br />

I506S/1651 dual polymorphism, and dF508, and dI507. Probe<br />

libraries composed of 8-mers and 9-mers annealing to PCR products have<br />

been used in our experiments. DNA from blood or cheekbrush samples was<br />

used from 10 genomic sources. The method discriminates both alleles of a<br />

heterozygote pair in a single experiment, and can discriminate alleles from<br />

multiple loci in the same sample.<br />

P60-T<br />

A software algorithm for automated quantitation of ICAT labeled<br />

peptides analyzed by mass spectrometry is described.<br />

J. Eng, J. Ranish, R. Aebersold; Inst. for Systems Biol., 4225 Roosevelt<br />

Way NE, #200, Seattle, WA 98105<br />

We describe a software program which automatically generates quantitation<br />

ratios of Isotope Coded Affinity Tag (ICAT) labeled peptides analyzed in an<br />

LC-MS/MS experiment performed on an LCQ ion trap mass spectrometer. The<br />

peptides are initially identified via a sequence database search. Quantitation<br />

of each ICAT-labeled peptide pair is then performed by integrating over the<br />

peptide’s light and heavy elution profiles. An interactive, graphical program<br />

displays the light and heavy elution traces and allows the researcher to<br />

adjust the area of integration as needed. This program should enable and<br />

facilitate large scale, quantitative proteomics studies.


P61-S<br />

Genopure: a novel magnetic bead DNA purification system for<br />

MALDI-TOF MS analysis.<br />

M. Kostrzewa1, J. Bimmler2, I. Thomas2, T. Wenzel1, E. Nordhoff3, H. Rauth3, T. Fröhlich1; 1Bruker Saxonia Analytik GmbH, Permoserstrasse<br />

15, Leipzig, Saxonia D-04318 Germany, 2Bruker Saxonia Analytik<br />

GmbH, Leipzig, 3Max Planck Inst. of Molec. Genet., Berlin<br />

MALDI-TOF mass spectrometry has a high potential for high throughput DNA<br />

analyses because of its accuracy, speed, automation capabilities, and costeffectiveness.<br />

Unfortunately, molecular biological reactions are performed in<br />

the presence of high amounts of salts and detergents which form adducts<br />

with DNA or interfere with matrix crystallization, respectively. Therefore,<br />

effective DNA purification is indispensable for successful and valid MALDI-<br />

TOF MS analysis. We present a novel magnetic DNA purification system<br />

which gives excellent results in subsequent mass spectrometric measurement.<br />

Small double stranded PCR products were precipitated on paramagnetic<br />

beads using an appropriate binding solution. The binding of DNA to the particles<br />

did not require any labeling of primers or dNTPs. After several washing<br />

steps with buffers containing either ethanol or isopropanol, DNA was<br />

eluted and measured with 3-HPA matrix in a MALDI-TOF mass spectrometer.<br />

Residual PCR primers were removed while PCR products down to about<br />

50 bp in size were recovered with high yield. Quality of the acquired spectra<br />

was superior to that of alternative purification protocols.<br />

Alternatively, eluted PCR-Products were used for subsequent reactions like<br />

primer extension or restriction enzyme digest. Primer extension products or<br />

very small restriction enzyme digest products (about 20 bp) were again purified<br />

by the magnetic bead system to MALDI quality with high recovery using<br />

a dedicated binding buffer. The combination of both purification procedures<br />

enabled the genotyping of SNPs by performing PCR, purification of the<br />

PCR product, primer extension or digest, and clean-up of the allele specific<br />

products in one reaction tube. High quality spectra could be acquired for several<br />

model systems allowing the unambiguous determination of genotypes.<br />

The bead handling can easily be automated on standard pipetting robots<br />

thereby enabling the processing of thousands of samples per day.<br />

P63-T<br />

Bioinformatics assessment of mass spectrometric chemical<br />

derivatisation techniques for proteome database searching.<br />

K.S. Sidhu1, P.J. Lester1, S.J. Gaskell1, O.V. Wolkenhauer1, S.G. Oliver2, F. Brancia1, A.G. Sullivan1, P. Sangvanich1, S.J. Hubbard1; 1UMIST, UK,<br />

PO Box 88, Manchester, Lancashire M60 1QD, United Kingdom,<br />

2Manchester Univ.<br />

Identification of proteins from the mass spectra of peptide fragments generated<br />

by proteolytic fission and subsequent database searching is one of the<br />

most powerful techniques in proteome science. Using computer simulation,<br />

we have studied how various chemical derivatisation techniques developed<br />

in the Michael Barber Centre for Mass Spectrometry at UMIST can improve<br />

the efficiency of protein identification from mass spectrometric data. The<br />

impact of these different derivatization strategies, which promote and stabilise<br />

certain fragmentation pathways yielding additional database search<br />

information has been assessed. For example, by reliably promoting fragmentation<br />

at aspartic acid residues after homoarginine derivatisation, 85% of<br />

yeast proteins can be unambiguously identified from a single peptide with a<br />

measured mass accuracy of 500 ppm by using the associated fragment ion<br />

data. We present results here for 3 alternative techniques, both for single proteins<br />

and for simple mixtures. Additional data have also been generated to<br />

compare these “partial sequencing” methods with high accuracy mass spectrometry<br />

where peptide mass accuracy can be achieved at 50 ppm and<br />

beyond. Interestingly, for protein mixture analysis, the inclusion of limited<br />

sequence information for the peptides can compensate and exceed the<br />

search efficiency available via high accuracy searches of around 20 ppm, suggesting<br />

the particular use of this experimental approach for the simple protein<br />

mixtures that are routinely obtained from 2D-gels.<br />

POSTER <strong>AB</strong>STRACTS<br />

<strong>AB</strong>RF 2001 <strong>AB</strong>STRACTS<br />

P62-M<br />

Industrial scale high throughput proteomics.<br />

J. Brown, D. Gostick, P. Young, J. Langridge; Micromass UK Ltd,<br />

Floats Road, Wythenshawe, Manchester M23 9LZ, United Kingdom<br />

With the completion of the Human Genome sequence MALDI-TOF-MS is<br />

increasingly becoming an established method for identification of proteins<br />

separated by 2D gel electrophoresis. Mono-isotopic peptide mass fingerprinting<br />

(PMF) has been previously shown to be amenable to full automation<br />

encompassing the process of acquisition, data processing and databank<br />

searching under full software control.<br />

Until now the throughput of MALDI-TOF-MS for proteomics has been limited<br />

to several hundred samples in a working day and this represents approximately<br />

5–10% of the total proteins resolved by a large format 2D gel. To<br />

reduce the number of proteins to be identified the 2D gels are imaged and<br />

analysed to determine differences in expression levels within a set of gels.<br />

Although much of the image processing is semi-automated the comparison<br />

is labour intensive as manual pattern matching has a role in the gel alignments<br />

(land marking). Increased MS sample throughput allows the possibility<br />

of identifying every protein spot in a 2D gel within a day. This could eliminate<br />

the potentially erroneous step of human gel image alignment, whereby<br />

land marking could be achieved using the MS data.<br />

Increased sample throughput requires greater capacity and robust unattended<br />

instrument operation. In this poster we describe an integrated robotic<br />

multiple plate loader that allows overnight unattended MS operation. Other<br />

improvements include an increased laser repetition rate that allows the data<br />

capture rate to increase four fold. Sample tracking, data archiving and data<br />

reporting are essential attributes of this new technology and these aspects are<br />

outlined in the present<br />

P64-S<br />

Automated high throughput protein identification on a hybrid<br />

quadrupole orthogonal acceleration time-of-flight mass<br />

spectrometer coupled with a MALDI ion source.<br />

A. Millar, R. Tyldesley, J.I. Langridge, J.B. Hoyes, Y. Philip, R. O’Malley,<br />

R. Bateman; Micromass UK Ltd., Floats Road, Wythenshawe, Manchester<br />

M23 9LZ, United Kingdom<br />

The mass spectrometry technique providing the highest throughput in terms<br />

of samples per hour, is currently MALDI-TOF-MS. This technique provides a<br />

peptide mass fingerprint of the protein digests and allows rapid and accurate<br />

identification of the parent protein by comparison to a databank. However,<br />

under some circumstances, for example if the number of peptides detected<br />

is small or if the sequence coverage is poor, it is advantageous to be able to<br />

include even a short piece of sequence information to provide added specificity.<br />

In a conventional MALDI-TOF-MS instrument post source decay (PSD)<br />

can be used to try and generate sequence information, however this<br />

approach is notoriously unreliable in producing good quality MS/MS data.<br />

One reason for this is that the peptide does not undergo fragmentation in a<br />

controlled environment such as a gas cell.<br />

An alternative approach is to couple a MALDI ion source to a hybrid quadrupole<br />

orthogonal acceleration time-of-flight (Q-Tof) mass spectrometer, which<br />

results in predictable fragmentation. In contrast to a conventional MALDI-<br />

TOF-MS instrument the resolution and mass measurement accuracy of the<br />

data is comparable between the MS and MS/MS modes. This allows superior<br />

data acquisition in the MS-MS mode compared to other MALDI-TOF systems.<br />

In this paper we demonstrate the application of the MALDI Q-TOF instrument<br />

for high throughput proteomics. A number of modifications have been<br />

made to optimise the system for high throughput proteomics, these include<br />

a MALDI source with a high-density target plate and software that has been<br />

developed for automated data acquisition in both the MS mode and the MS<br />

to MS/MS switching mode. Dedicated processing software has been developed<br />

to fully automate the post acquisition and databank searching. This software<br />

has been optimised to consider the unique nature of the data acquired<br />

from this configuration of instrument.<br />

JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000 203


<strong>AB</strong>RF 2001 <strong>AB</strong>STRACTS<br />

P65-M<br />

Single scan PSD-MALDI-TOF analysis at high speed and sensitivity.<br />

D. Suckau, A. La Rotta, A. Holle, P. Hufnagel, D. Suckau; Bruker Daltonik<br />

GmbH, Bremen, Fahrenheitstr. 4, Bremen 28359, Germany<br />

The typical approach to Proteomics measurements involves the MALDI-TOF<br />

fingerprint analysis of protein digests, which allows the identification of a significant<br />

fraction of samples. PSD as it is available on a reflector TOF instrument<br />

is in our hands a very sensitive technique (1–10 fmol level) and is automated;<br />

therefore it principally is the ideal technique to continue MS/MS<br />

analysis from the same prepared sample. However, it suffers from the fact<br />

that each PSD spectrum is pasted together from a set of ca. 10 voltage segments,<br />

each consisting of 100–200 laser shots. Sample consumption is such<br />

that a maximum of 2–3 peptides can be submitted to MS/MS analysis and the<br />

process is time consuming. Here we report on the usability of single scan<br />

PSD spectra from digests for protein identification.<br />

The acquisition of the entire fragment spectrum has been achieved for a<br />

MALDI reflector TOF mass spectrometer by introducing a “potential lift”, a<br />

precursor selection device consisting of a pulsed HV cell with acceleration<br />

grid. An HV poten-tial is applied to the lift cell at the moment the precursor<br />

ion and its PSD fragment ions have entered it. By the subsequent acceleration,<br />

the energies of the fragment ions are compressed into the operating<br />

window of the reflector, which allows analysing them in a single spectrum.<br />

With this device LIFT-PSD spectra from proteolytic digest mixture were<br />

obtained and evaluated with respect to sensitivity, mass accuracy, speed of<br />

analysis and number of PSD spectra per sample. It potential use in Proteomics<br />

is discussed.<br />

P67-S<br />

Proteomics approach to investigate the toxicity of diclofenac.<br />

L.J. Alward, O.V. Nemirovskiy, G.S. Cavey, J.E. Carlson, W.R. Mathews,<br />

J.A. Ware; Pharmacia, 301 Henrietta Street, Kalamazoo, MI 49007<br />

Diclofenac, a nonselective inhibitor of cyclooxygenase, is a widely used<br />

non-steroidal anti-inflammatory drug. It has been shown to produce<br />

hemolytic anemia in sensitive individuals. A proteomics approach was used<br />

to determine the mechanism of diclofenac-induced hemolytic anemia in the<br />

mouse. A flexible, high performance proteomics laboratory was assembled<br />

using commercially available instrumentation. The system used was validated<br />

with protein standards and included robotic spot excision from 2D gels, automated<br />

in-gel digestion, robotic sample preparation, automated MALDI-Tof<br />

analysis, automated nanoLC-MS/MS analysis, and automated database searching.<br />

Diclofenac was administered to mice by gavage. Liver, plasma, and RBC<br />

proteins were separated using 1D and 2D gels, and diclofenac-protein<br />

adducts were visualized by either radiolabel or immunoblot. Proteins of<br />

interest were excised, reduced, alkylated, and trypsin digested. Successful<br />

protein identification was accomplished via MALDI-Tof MS peptide mass fingerprint<br />

analysis using 20% of digest sample. Nanoscale LC-MS/MS provided<br />

comprehensive identification using 50% of digest sample. The results<br />

obtained by nano-LC-MS/MS analyses were in a good agreement with those<br />

obtained by MALDI-Tof. Many proteins were identified including fibrinogen<br />

alpha and beta, hemoglobin beta and delta chains, and RBC anion exchanger<br />

(AE1). Abnormalities in AE1 have been shown to accelerate membrane<br />

destruction. Preliminary data from these experiments is promising and may<br />

help explain diclofenac toxicity.<br />

POSTER <strong>AB</strong>STRACTS<br />

204 JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000<br />

P66-T<br />

Quantitative proteomic analysis using a MALDI quadrupole<br />

time-of-flight mass spectrometer.<br />

T.J. Griffin1, S.P. Gygi2, B. Rist3, H. Zhou1, H. Lee1, A. Loboda4, A. Jilkine5, W. Ens6, K.G. Standing6, R. Aebersold1; 1Inst. for Systems Biol., Seattle,<br />

4225 Roosevelt Way N., Seattle, WA 98105, 2Harvard Med. Sch., 3BioVisioN<br />

GmbH and Co. KG, Hannover, 4MDS Sciex, Concord, Canada, 5Univ. of<br />

Manitoba, 6Univ. of Manitoba<br />

We describe an approach to the quantitative analysis of complex protein mixtures<br />

using a MALDI quadrupole time-of-flight (MALDI QqTOF) mass spectrometer<br />

and isotope coded affinity tag (ICAT) reagents. Proteins in mixtures<br />

are first labeled on cysteinyl residues using an ICAT reagent, the proteins are<br />

enzymatically digested, and the labeled peptides are purified using a multidimensional<br />

separation procedure, with the last step being the elution of the<br />

labeled peptides from a microcapillary reverse-phase liquid chromatography<br />

column directly onto a MALDI sample target. After addition of matrix, the<br />

sample spots are analyzed using a MALDI QqTOF mass spectrometer, by first<br />

obtaining a mass spectrum of the peptides in each sample spot in order to<br />

quantify the ratio of abundance of pairs of isotopically tagged peptides, followed<br />

by tandem mass spectrometric analysis to ascertain the sequence of<br />

selected peptides for protein identification. The effectiveness of this approach<br />

is demonstrated in the quantification and identification of peptides from a<br />

control mixture of proteins of known relative concentrations, and also in the<br />

comparative analysis of protein expression in Saccharomyces cerevisiae<br />

grown on two different carbon sources. Additionally, we have demonstrated<br />

the utility of this approach for the quantitative analysis of isotopically labeled<br />

phosphorylated proteins, and we are currently working on automating the<br />

sample preparation procedures for high-throughput applications.<br />

P68-M<br />

Selective separation of methionine-containing peptides<br />

combined with mass spectrometry: a high accuracy technique<br />

for identification of proteins.<br />

W.L. McEldoon, M.J. Horn; BioMolecular Technol., Inc.,<br />

525F Del Rey Ave., Sunnyvale, CA 94085<br />

Although MALDI-TOF MS is a widely accepted technique for identification of<br />

proteins and peptides in proteomic and genomic analyses, rapid and accurate<br />

identification of a large number of proteins is still a great challenge. Previously,<br />

we reported an easy method, which uses a novel water-insoluble<br />

reagent that selectively separates methionine-containing peptides from peptide<br />

mixtures. This method, combined with MALDI-TOF analysis, demonstrates<br />

a significantly improved accuracy in the identification of proteins. In<br />

the present study, we present a total methodology for the separation and<br />

identification of methionine-containing peptides generated from 2D gel<br />

digestion mixtures. The methodology provides a “one-pot” method for sample<br />

desalting and concentration after the isolation process, giving cleaned-up<br />

samples ready for multiple MALDI or other analytical analyses.


P69-T<br />

Efficient detergent and Coomassie blue dye removal from peptide<br />

samples prior to MALDI-TOF MS.<br />

W. Kopaciewicz, E. Kellard; Millipore, 17 Cherry Hill Drive, Danvers,<br />

MA 01923<br />

Advancements in Mass Spectrometry (MS) instruments over the last 5 years<br />

have reduced the amount of sample required and expanded the molecular<br />

weight analysis range. As such, MS is an increasingly valuable tool for the<br />

analysis of biomolecules in areas of proteomics and functional genomics.<br />

However, the presence of salts, detergents and dyes can decrease macromolecular<br />

ionization and/or obscure mass peaks, thus decreasing instrument<br />

performance. As such, numerous methods and devices have been<br />

developed for sample preparation prior to MS analysis.<br />

Millipore ZipTip sample preparation devices containing C18 and C4 media<br />

reversed phase media have been demonstrated to be effective for the concentration<br />

and desalting of microliter volumes of peptide, protein and<br />

oligonucleotide samples prior to MALDI-TOF MS. Although reversed phase<br />

chromatography meets the vast majority of the sample preparation needs,<br />

there are cases where it isn’t totally effective. This is especially evident when<br />

the contaminants have similar hydrophobic character as the solutes and thus<br />

co-purify.<br />

Alternatively, a subset of normal phase chromatography termed hydrophilic<br />

interaction chromatography has been shown to be an efficient sample preparation<br />

technique for certain biomolecules in those cases where reversed<br />

phase struggles (1,2,3). The method utilizes a highly hydrophilic adsorbent<br />

that binds sample out of a high organic mobile phase (e.g. 90% acetonitrile).<br />

After washing with this solvent, bound molecules are then eluted by decreasing<br />

the organic content (e.g. 50% acetonitrile). In this poster, we demonstrate<br />

the use of a ZipTip with hydrophilic interaction media (ZipTipHPL). The<br />

device was very effective for the removal of detergents and coomassie blue<br />

dye from sub-picomole quantities of peptides. The resulting mass spectra<br />

demonstrated good signal strength and fidelity of peptide capture.<br />

1. Alpert, A.J., J. Chromatogr. 499 (1990) p.177<br />

2. Zhu, B.Y., et al., J. Chromatogr. 548 (1991) p.13<br />

3. Scherer, J.P., et al., Anal. Biochem. 215 (1993) p.292<br />

P71-M<br />

Factors affecting rapid sample processing for protein capture<br />

chips analyzed by MS.<br />

J. Chan1, P.T. Jedrzejewski1, P. Wagner1, S. Nock2; 1Zyomyx, Inc,<br />

3911 Trust Way, Hayward, CA 94545, 2Zyomyx, Inc.<br />

Robust, sensitive, and comprehensive methodologies for systemic approaches<br />

to proteome analysis analogous to genomics methods (e.g., gene chips) are<br />

not available. Current methodologies (e.g., 2D-PAGE and multi-dimensional<br />

chromatographic methods) suffer from fundamental limitations. In order to<br />

overcome these limitations, we as well as others have focused on the development<br />

of alternative methods (e.g., protein capture chips) for proteomics.<br />

Protein capture chips, consisting of arrayed capture molecules (e.g. protein,<br />

DNA, or small molecule), allow for rapid and comprehensive micropurification<br />

and analysis of proteins. Protein chips may be readily interrogated by<br />

mass spectrometric analysis. Data on the post-translational modifications<br />

(PTM), protein identification, and quantitation may be obtained with a single<br />

detection scheme without labeling of analytes.<br />

We have been successful in microfabricating protein capture arrays, the<br />

accompanying microfluidics devices, and implementing MS analysis. We<br />

have investigated various parameters which have an effect on the utility and<br />

efficacy of the overall system for proteomics. In this presentation, we will<br />

demonstrate data on parameters affected by the type of capture molecules,<br />

digest efficiency (e.g., time), and sensitivity of analysis (e.g., LC-MS conditions).<br />

POSTER <strong>AB</strong>STRACTS<br />

<strong>AB</strong>RF 2001 <strong>AB</strong>STRACTS<br />

P70-S<br />

Improved proteome coverage through the use of affinity tags<br />

with nanoscale capillary LC/MS/MS.<br />

K. Blackburn1, W. Burkhart2, R. Davis2, M. Moyer2, A. Moseley2; 1Glaxo Wellcome, 5 Moore Drive, PO Box 13398, Research Triangle Park,<br />

NC 27709, 2Glaxo Wellcome<br />

Because of the inherent difficulties associated with 2D gel electrophoresis<br />

(low throughput, bias against certain classes of proteins, poor peptide recovery<br />

from in-gel digests, etc.), numerous groups have explored ways to analyze<br />

complex protein mixtures directly by mass spectrometry, avoiding the<br />

gel separation altogether. In proteomic experiments where qualitative and/or<br />

quantitative data is required for complex biological samples such as protein<br />

complexes, organelles, or tissue samples, the number of proteins potentially<br />

encountered may range from tens to thousands. The number of proteolytic<br />

peptides generated from digests of such complex protein mixtures precludes<br />

“complete” proteome coverage by any 1D-LC/MS/MS analysis. Following on<br />

the work of Aebersold and Patterson, we have investigated the use of cysteine-modifying<br />

“affinity” tags with avidin affinity chromatography for the<br />

simplification of complex peptide mixtures prior to nanoscale capillary<br />

LC/MS/MS analysis. This approach reduces the number of peptides presented<br />

to the mass spectrometer to a more reasonable number, thus allowing<br />

improved proteome coverage. Data will be presented on improved sample<br />

handling procedures for labeling and digestion steps as well as qualitative<br />

and quantitative applications to complex biological samples.<br />

P72-T<br />

High throughput in-gel peptide digestion and microscale sample<br />

preparation for MALDI-MS analysis of the resulting peptide<br />

mass fingerprint.<br />

M.G. Pluskal1, A.M. Pitt2; 1Proteome Systems, Inc., 14 Gill St.,<br />

Woburn, MA 01801, 2Millipore Corp.<br />

In-gel peptide digestion has become a widely used technique for characterizing<br />

proteins resolved by two dimensional gel electrophoresis. Peptides<br />

generated from gel pieces are frequently contaminated with detergent and<br />

salts. Prior to MALDI-MS analysis, these contaminants are removed using<br />

microscale C18 sample preparation columns. In this poster, data will be presented<br />

to demonstrate the application of a solvent resistant Multiscreen 96<br />

well plate with an optimized low peptide binding membrane and ZipTip C18<br />

micropipet based sample preparation. Recoveries of peptides (Mz range of<br />

1000 to 5000 Dalton) derived from standard protein protease digests, were<br />

estimated at various stages of the analytical process. An optimized protocol<br />

has been established and all the reagents and consumables have been packaged<br />

in a ready to use commercial kit. Data will be presented to show application<br />

of this technology package to accelerate the throughput of protein<br />

characterization by protease fragmentation.<br />

JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000 205


<strong>AB</strong>RF 2001 <strong>AB</strong>STRACTS<br />

P73-S<br />

Rapid and high throughput proteomics by protein capture chips<br />

combined with MS analysis.<br />

J. Chan1, P.T. Jedrzejewski1, P. Zwahlen2, P. Indermuhle2, P. Wagner1, S. Nock3; 1Zyomyx, Inc, 3911 Trust Way, Hayward, CA 94545,<br />

2Zyomyx, Inc., Hayward, CA, 3Zyomyx, Inc.<br />

The study of the proteome is a critical step towards the function determination<br />

of each gene since proteins are the agents of life’s work. However,<br />

robust, sensitive, and comprehensive methodologies for systemic approaches<br />

to proteome analysis similar to genomics methods (e.g., gene arrays) are not<br />

available. Current methodologies such as 2D-PAGE and multi-dimensional<br />

chromatographic methods (e.g., LC-LC, LC-CE) suffer from fundamental limitations.<br />

In order to overcome these limitations, we as well as others have<br />

focused on the development of alternative methods (e.g., protein capture<br />

chips) for proteomics.<br />

Consisting of arrayed capture molecules, protein chips allow for rapid and<br />

comprehensive micropurification and analysis of proteins. Protein chips may<br />

be readily analyzed by optical methods; however, detailed data may be<br />

obtained when arrays are interrogated by MS. Data on the post-translational<br />

modifications (PTM), protein identification (especially important when complexes<br />

are isolated), and quantitation may be obtained with a single detection<br />

scheme without labeling of analytes. In this presentation, we will show<br />

our efforts in this area.<br />

We have been successful in microfabricating protein capture arrays, the accompanying<br />

microfluidics devices, and implementing MS analysis. These mircrofluidic<br />

devices offer a number of advantages (e.g., high density elements,<br />

integration of operations). We have devised a microfluidic device capable of<br />

performing a number of sample preparation operations prior to MS analysis.<br />

In this presentation, we will demonstrate the functionality and utility of these<br />

devices through the identification and characterization of proteins captured<br />

from mixtures. Various assay types will be demonstrated (e.g., protein-protein,<br />

small molecule-protein, protein-DNA). The overall system performance characteristics<br />

will be presented: low femtomole sensitivity, high dynamic range,<br />

and selectivity. The utility of rapid protein identification through database<br />

searching and PTM characterization will be highlighted in various applications.<br />

P75-T<br />

Rapid protein identification of blocked proteins using dilute acid<br />

cleavage and automated Edman sequencing.<br />

S. Wong, A. Kishiyama, V. Pham, W.J. Henzel; Genentech<br />

We have developed a method for rapidly cleaving and identifying proteins<br />

electroblotted onto polyvinylidene difluoride (PVDF) membranes. Cleavage<br />

is performed with 10% acetic acid in 7 M guanidine chloride at pH 2.5 for<br />

1 hour at 90�C, resulting in fragmentation primarily at aspartyl-prolyl bonds.<br />

Peptides resulting from non Asp-Pro cleavage are N-terminally blocked by<br />

reaction with orthophthalaldehyde (OPA) prior to automated Edman degradation.<br />

Reaction with OPA after cleavage blocks all amino acids containing<br />

primary amino groups. Only peptides containing an N-terminal amino acid<br />

with a secondary amino group (proline) will be available for reaction with<br />

the Edman reagent. The sequences obtained are used for protein database<br />

searching. Using this approach, proteins that are found to be N-terminally<br />

blocked can be removed from the sequencer, cleaved with acetic acid,<br />

blocked with OPA and reapplied to the sequencer. The protein can then be<br />

identified from a database search using the sequence mixture obtained.<br />

POSTER <strong>AB</strong>STRACTS<br />

206 JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000<br />

P74-M<br />

Reflector in-source-decay MALDI TOF MS: a powerful tool for<br />

N-terminal sequence characterization of proteins.<br />

D. Suckau, A. Resemann, M. Witt; Bruker Daltonik GmbH, Bremen,<br />

Fahrenheitstr. 4, Bremen 28359, Germany<br />

MS-characterization of proteins can simply be initiated by intact molecular<br />

weight determinations. If the experimental value doesn’t agree with the<br />

anticipated structure, a closer investigation of the termini is a must—besides<br />

modifications. In particular N-terminal modifications impose a significant<br />

analytical problem, since Edman sequencing fails and MS/MS of digested<br />

peptides generates a tremendous amount of (useless) information to address<br />

this question and might even not contain the N-terminal peptide.<br />

We introduce ISD measurements on a standard reflector MALDI-TOF instrument<br />

to obtain near-N-terminal sequence information (residues �10–30).<br />

C-type MS/MS fragment ions generated from intact proteins of up to 60 kDa<br />

using this approach allow to read the sequence with a �0.04 Da mass accuracy.<br />

This sequence information can directly be used for sequence database<br />

searches. This information was sufficient to characterize the N-terminus, i.e.,<br />

to identify the fusion protein system and the unexpected N-terminal Metmethylation,<br />

which was confirmed by additional PSD and fingerprint analysis.<br />

ISD seem to be an ideal technique to characterize the N-terminus of proteins<br />

and in particual of N-terminally blocked proteins, provided they are available<br />

on the 10 pmol level and not contaminated by other proteins in a mixture.<br />

P76-S<br />

Quantitation of gel separated proteins at the low picomole level.<br />

S.W. Yuen, L.R. Zieske, K-L. Hsi, T.L. Schlabach; Applied Biosystems,<br />

850 Lincoln Centre Drive, Foster City, CA 94404<br />

Comparative proteomics depends on the ability to quantify changes in cellular<br />

protein levels between the control state and the abnormal or treated<br />

state. The most widely accepted method for protein quantitation is amino<br />

acid analysis, but this method performs poorly at low picomole levels and<br />

below, which are most common in proteomics studies.<br />

Edman chemistry is a nearly stoichometric method for removing amino acid<br />

residues from a protein with typical repetitive yields of greater than 90%. The<br />

problem is the initial yield is not stoichometric and shows variability that is<br />

dependent on protein sequence.<br />

Cleavage of an unknown protein into numerous fragments reduces the<br />

impact of variability in the initial yield of any one fragment and removes<br />

much of variability associated with sequence dependence. We examine both<br />

chemical and enzymatic digestion techniques for fragmentation.<br />

We report in this presentation on the relative and absolute accuracy of protein<br />

sequencing for determining protein concentration. Both solution and gel<br />

isolated proteins will be studied and the results compared with sequence<br />

analysis of intact proteins.


P77-M<br />

The utilities of N-terminal sequencing in the post-genomic era.<br />

L.R. Zieske1, S.W. Yuen1, T.A. Settineri1, D. Hawke1, C. Bloch2; 1Applied Biosystems, 850 Lincoln Centre Drive, Foster City, CA 94404,<br />

2Embrapa-Cenargen, Brazil<br />

Mass spectrometry continues to develop and improve methods and procedures<br />

to make it the primary tool to identify proteins for “proteome analysis”.<br />

The high mass accuracy and sensitivity of instruments such as hyphenated<br />

quadrupole TOF mass spectrometers has made low level sequencing of<br />

peptides and proteins possible. Yet even with these advancements in MS the<br />

need for chemical sequencing still exists!<br />

MS sequencing is very powerful, but exact location of the fragmentation patterns<br />

are not predictable. Thus creating difficulty in assigning the exact N-termini.<br />

The need for exact N-terminal sequence information is especially<br />

important in determining mRNA editing and/or determining the true open<br />

reading frames when EST database searching. Edman chemistry is the one<br />

sure way of determining this type of information. Complementary to this is<br />

the use of “multiple peptide sequencing” which simultaneously provides multiple<br />

amino acid information per residue cycle. Thus providing very high<br />

quality sequence information with which to interrogate the error prone databases<br />

now used for searching. In addition, chemical sequencing provides the<br />

necessary information to determine quality assurance of cloned products;<br />

again making sure no frame shifts occurred in the processing. Another complementary<br />

need for chemical sequencing is in assisting the assignment of the<br />

correct ion series patterns being generated even when relatively complete<br />

fragmentation data is recorded. Chemical sequencing also identifies difficult<br />

to discern or unusual amino acids easily such as I/L and/or hydroxyproline.<br />

In this work we will show some examples of how N-terminal analysis was<br />

used in concert with mass spectrometry to unravel protein/peptide structural<br />

information. In addition, we will show demonstration of “multiple peptide<br />

sequencing” in inspecting the various databases available.<br />

P79-S<br />

Microsatellite analysis using fluorescent PCR primers synthesized<br />

in tandem.<br />

J. Fernandez, M. Kirchner, J. Brito, T-J. Daley, G. Dolios, B. Imai;<br />

Rockefeller Univ., 1230 York Ave., New York, NY 11021<br />

SDS-PAGE is typically the final purification step for isolation of small amounts<br />

of proteins for further chemical characterization. Proteins are digested in-gel<br />

with trypsin and the resultant peptides analyzed by 1) MALDI-TOF mass<br />

spectrometry for tentative protein identification using database search programs<br />

such as ProFound (http://prowl.rockefeller.edu) and/or 2) capillary<br />

HPLC followed by Edman sequence analysis to obtain definitive primary<br />

sequence information. While this strategy works well for Coomassie stained<br />

proteins it poses a problem for the more sensitive silver staining method. A<br />

technique has been published for protein identification using MALDI-TOF<br />

mass spectrometric analysis after reversal of the silver stain (Gharahdaghi et<br />

al., Electrophoresis 1999 20, 601–605). We have employed this technique for<br />

preparation of proteins for internal Edman sequence analysis. Because of the<br />

usually low amount of silver stained proteins, peptides need to be isolated<br />

by capillary HPLC and collection techniques need to minimize peptide loss.<br />

Silver stained gels, MALDI-TOF mass spectra, capillary HPLC, and Edman<br />

sequence data will be presented as well as guidelines for sample handling.<br />

POSTER <strong>AB</strong>STRACTS<br />

<strong>AB</strong>RF 2001 <strong>AB</strong>STRACTS<br />

P78-T<br />

Increasing the capacity of a commercial Edman sequencer:<br />

a protein auto sampler.<br />

W. Shillinglaw, S. Wong, T. Moreno, W.J. Henzel; Genentech<br />

We have designed an implemented a high throughput autosampler to<br />

increase the capacity and speed of protein sequencing. The autosampler<br />

attaches to a standard <strong>AB</strong>I Procise sequencer, enabling a single separate sample<br />

cartridge to now hold up to six separate samples. The autosampler is<br />

used in combination with faster Edman cycles and a rapid 12 minute PTH<br />

separation to significantly increase the speed of sample analysis. The reaction<br />

cartridges on the autosampler are composed of disposable Teflon tubing,<br />

allowing for a significantly cleaner background when compared to the<br />

reusable glass cartridge blocks, which are standard on the <strong>AB</strong>I sequencers.<br />

The lower background reduces the ambiguity of identifying the amino acids<br />

in the first few cycles, which is often a problem. A low cost program logic<br />

controller which is connected through an external relay to the protein<br />

sequencer controls the autosampler.<br />

P80-M<br />

Amino acid analysis 2000: a collaborative study from the<br />

<strong>AB</strong>RF AAA Research Group.<br />

M.A. Alterman1, P. Hunziker2, K. West3, R. Harris4, D. Chin5; 1Univ. of<br />

Kansas, 6038 Malott Hall, Lawrence, KS 66045, 2Univ. of Zurich,<br />

3Cleveland Clin. Fndn., 4Genentech, 5Univ. of Missouri<br />

The AAA Research Group of the <strong>AB</strong>RF periodically provides member laboratories<br />

with test samples in order to assist members in maintaining and<br />

improving the quality of AAA and assess the reliability of AAA. The latest<br />

study consists of two parts: Internet-based survey of equipment and technique<br />

currently used for AAA, and experimental data from this year’s test<br />

sample. The experimental part of 2000 study focused on quantitation and<br />

identification of proteins. The participating laboratories received solutions of<br />

4 pure proteins for the determination of their concentration by AAA as well<br />

as by a colorimetric method (BCA, Bradford, or similar). Data obtained will<br />

be used to examine the use of AAA for determining of the exact amount of<br />

protein in the sample and the use of compositional data to identify the protein.<br />

The relative precision and accuracy of the colorimetric assays will be<br />

compared with the AAA values. The comparison of the results of this study<br />

with the results from previous studies will show changes and improvements<br />

in the practice of AAA.<br />

JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000 207


<strong>AB</strong>RF 2001 <strong>AB</strong>STRACTS<br />

P81-T<br />

How to obtain reproducible gradient capillary/nano LC for ultrahigh<br />

sensitivity MS detection of biological macromolecus with<br />

and without flow splitting.<br />

Y-H. Jou, C. Wu, C. Wu, Y.W. Hong, F.J. Yang; Micro-Tech Scientific,<br />

Sunnyvale, CA, 140 South Wolfe Road, Sunnyvale, CA 94086<br />

The applications of gradient capillary column LC for biological macromolecular<br />

analysis and drug discovery research have increased significantly in<br />

recent years. As a result of this increasing need for sub-picomole detection,<br />

capillary column LC with eletro-spray and nano-spray MS is increasingly<br />

important.<br />

Flow stream splitting technique introduced in 1983 by van der Wal and Yang<br />

(1) has been utilized by many users for rapid evaluation and realization of<br />

the advantages of capillary LC and capillary LC-MS. However, the flow stream<br />

splitting technique has suffered from inherent poor retention time and gradient<br />

slope reproducibility when the following condition(s) occurred: high<br />

pressure, high split ratios, large sample amount of viscose sample solvent is<br />

injected, column inlet flow restriction change, or splitter flow restriction<br />

change.<br />

Splitless flow capillary LC allows the same performance and ease of validation<br />

for routine work as the conventional 4.6 mm id. gradient HPLC. However,<br />

it requires each pump to deliver accurate/reproducible flow rates at<br />

sub-�l/min. It also requires high pressure mixer(s) with small volume for fast<br />

gradient generation and minimum solvent gradient delay time from the mixer<br />

to the column inlet.<br />

This poster will discuss design concepts of a new reciprocating pump that<br />

delivers sub-�l/min flow rates for routine capillary/Nano LC applications.<br />

Performance of the system in terms of long-term retention time reproducibility<br />

for flow rates from 0.01 to 50 �l/min will be compared to a flow<br />

stream splitting system. Practical considerations in terms of gradient regeneration,<br />

system liquid end volume, mixer volume, mixing noise, and sample<br />

clean up, desalt, and concentrating will also be discussed.<br />

1. Sj. van der Wal and F. J. Yang, J. Resolut. Chromatogr. Chromatogr. Commun.<br />

6, 216 (1983).<br />

P83-M<br />

An automated sequence assignment and multiple database<br />

searching.<br />

T. Sasagawa, Y. Matsumoto, M. Kojima, Y. Mizuno; Toray Res. Ctr.,<br />

1111 Tebiro, Kamakura, Kanagawa 2488-8555, Japan<br />

Mass spectrometry and Edman degradation are complementary to each other<br />

and are indispensable techniques in proteomics in which systematic analysis<br />

of a large number of expressed proteins is required. In order to facilitate<br />

the interpretation of these data, automated data analysis and multiple database<br />

search programs linked together are developed. PepMs is for de novo<br />

sequencing based on a new algorithm. The sequence information can be<br />

obtained even if a few gaps are present. Post-translationally modified amino<br />

acid and ambiguous Edman sequence data can be used to aid the sequence<br />

determination.<br />

Seq is for the automated interpretation of Edman sequence data and for multiple<br />

database searching. By correcting raw data with extraction efficiency of<br />

PTH amino acids and with lag due to incomplete cleavage, unambiguous<br />

sequence assignment is possible. The assigned data are automatically subjected<br />

to database searching. A minor second sequence can be also determined.<br />

The program also has a routine to generate theoretically possible<br />

amino acid sequences based on observed PTH amino acids, when equal<br />

amount of multiple sequences are observed. Using the generated pairs of<br />

sequences, the correct pair of the sequence can be found by multiple database<br />

searching. The result is confirmed mass spectrometry and de novo<br />

sequence routine.<br />

POSTER <strong>AB</strong>STRACTS<br />

208 JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000<br />

P82-S<br />

Proteomics: identification of low abundance proteins<br />

by MDLC/MALDI-TOF analysis.<br />

M. Meys, S. Krishnan, K.C. Parker, M. Lin, M.D. Lynch, R. Carberry,<br />

K. Waddell; Applied Biosystems, Foster City, CA, 500 Old Connecticut<br />

Path, Framingham, MA 01701<br />

The study of proteome components commonly involves the separation of a<br />

protein extract on a 2-D gel followed by the analysis of the protein spots.<br />

This analysis is currently being done by proteolytic digestion of the spots followed<br />

by mass spectrometric techniques. Although this approach has proven<br />

to be useful, it has several limitations. The main limitations are the inability<br />

to analyze low abundance proteins, proteins of extreme pI’s and molecular<br />

weights. We present here a method that can overcome these limitations by<br />

eliminating the 2-D gel separation step. The process involves performing a<br />

multidimensional chromatographic separation of the protein extract followed<br />

by proteolytic digestion and MALDI-TOF mass spectrometric analysis of the<br />

digests. The removal of the gel step in the process offers accessibility to all<br />

classes of proteins, makes the process amenable for automation and enhances<br />

speed of analysis. The use of chromatography also improves peptide<br />

recovery. In addition, the use of the chromatographic separation enables the<br />

removal of the highly abundant proteins from the extract and aids in the<br />

identification of the low abundance proteins. In the present study Escherichia<br />

coli is used as a model to validate this approach. The crude extract was<br />

fractionated over a cation exchange column followed by further fractionation<br />

on a reverse phase column. The fractions from the reverse phase column<br />

were then digested with trypsin and subjected to MALDI-TOF mass spectrometric<br />

analysis. Using this approach we identified several low abundance<br />

proteins such as maltodextrin phosphorylase, leucine aminopeptidase, phosphate<br />

starvation inducible protein precursor. These proteins to our knowledge<br />

have not been located on 2-D gels of E coli indicating the validity and<br />

usefulness of the approach.<br />

P84-T<br />

Amino acid specific effects on C-terminal sequencing efficiency:<br />

comparison of activation chemistries.<br />

D.R. Dupont1, S.W. Yuen1, R.L. Noble1, K.S. Graham2; 1Applied<br />

Biosystems, 850 Lincoln Centre Drive, Foster City, CA 94404,<br />

2Beckman Res. Inst., City of Hope<br />

Efficient initial activation of the C-terminus of a protein or peptide is essential<br />

to successful C-terminal sequencing. To facilitate the evaluation of activation<br />

chemistries, we have synthesized sets of peptides that contain each of<br />

the genetically coded amino acids (except proline) in three sequence positions,<br />

at the C-terminus, penultimate to the C-terminus and third from the<br />

C-terminus. The peptides are sequenced in groups of four or five to limit the<br />

number of sequencing runs necessary to evaluate the behavior of each of the<br />

amino acids in each sequence position. Included in each group are several<br />

amino acids with reactive side chains together with amino acids with unreactive<br />

side chains. The peptides are covalently attached to modified PVDF<br />

membrane to minimize the effect of sample loss due to washout.<br />

In the ATH method, the C-terminus is reacted with acetic anhydride to form<br />

an oxazolone, which is then reacted with tetrabutylammonium thiocyanate<br />

in the presence of TFA vapor to form a C-terminal thiohydantoin. An alternative<br />

approach is the use of a reagent that can form thiohydantoin without<br />

first forming oxazolone, which should minimize the potential for side reactions<br />

and improve the sequencing initial yield. In the C-terminal sequencing<br />

chemistry developed at the Beckman Research Institute, one such reagent,<br />

diphenylphosphoroisothiocyanatidate (DPP-ITC), is used for activation and<br />

thiohydantoin formation. Several years ago we presented a preliminary comparison<br />

of these activation chemistries using several model proteins. Here we<br />

compare the activation chemistries for all the amino acids (except proline)<br />

using the peptide sets and outline the effects of incorporating DPP-ITC activation<br />

into the ATH chemistry. In addition, we compare the efficiency of the<br />

activation chemistries on a variety of proteins at different sample amounts.


P85-S<br />

Rapid oligosaccharide mapping using fluorescent<br />

anthranilic acid detection.<br />

S.T. Dhume, S.A. Batz; SmithKline Beecham, 709 Swedeland Rd,<br />

Mailcode UW2960, King of Prussia, PA 19406<br />

Oligosaccharide mapping and characterization methods based on fluorescent<br />

Anthranilic acid (AA, 2-aminobenzoic acid) labeling affords high resolution<br />

and high sensitivity detection of glycans (1). The AA tagging method offers<br />

a significant improvement over other methods and is being rapidly adopted<br />

in the area of glycoprotein analysis.<br />

The oligosaccharide mapping method starting with an N-linked glycoprotein<br />

requires an overnight enzymatic digestion, 1 hour reaction with AA, a purification<br />

step followed by a 2 hour chromatographic run. It is the intent of this<br />

work to allow rapid detection of oligosaccharides while retaining the quality<br />

and reproducibility of the original method.<br />

The enzymatic release of oligosaccharides with the amounts of substrate and<br />

enzyme used is essentially complete in 3 hours. The glycan profiles obtained<br />

with PNGase F incubations between 30 min and 72 hours are similar except<br />

for the lower peak intensities at shorter (�2.5 hours) incubation times.<br />

The purification step to remove excess AA may be omitted. The mapping is<br />

directly applicable to fetuin, a highly sialylated glycoprotein but needed gradient<br />

changes for neutral glycan species to retain resolution, tailing and<br />

quality of maps as those from the original method.<br />

Oligosaccharide mapping was carried out on a short (2.1 mm � 15 cm) polymeric<br />

amine-bonded column allowing reduction in the time of analysis to<br />

about an hour. Initial results with reverse phase chromatography are promising<br />

and also allow 1 hour runs per sample.<br />

The modifications may be used alone or in any combination. Without consideration<br />

of the time for enzyme incubation, the total time saved is more<br />

than 50%. In addition, 80% of HPLC solvent consumption is avoided. The<br />

reproducibility and universal applicability of this method for other oligosaccharides<br />

will be discussed in detail.<br />

1. Anumula, K. R. and Dhume, S. T. (1998) Glycobiology, 8, 685–694.<br />

P87-T<br />

FMN is covalently attached to a specified threonine residue<br />

via phosphate group in the NqrB and NqrC subunits of<br />

Na�-translocating NADH-quinone reductase from<br />

Vibrio alginolyticus.<br />

M. Maeda1, M. Hayashi2, Y. Nakayama2, M. Yasui2, K. Furuishi3, T. Unemoto2; 1Applied Biosystems, Framingham, MA, 2Chiba Univ.,<br />

3Applied Biosystems, 4-5-4 Hatchobori, Chuo-ku, Tokyo 104-0032, Japan<br />

Na �-translocating NADH-quinone reductase (NQR) from Vibrio alginolyticus<br />

is composed of six subunits (NqrA to NqrF). We previously demonstrated that<br />

both NqrB and NqrC subunits contain a flavin cofactor covalently attached<br />

to a threonine residue. Fluorescent peptide fragments derived from the NqrB<br />

and NqrC subunits were applied to a matrix-assisted laser desorption time of<br />

flight (MALDI-TOF) mass spectrometer and covalently attached flavin was<br />

identified to be FMN in both subunits. From post-source decay (PSD) fragmentation<br />

analysis, it was concluded that FMN is attached via phosphate<br />

group to Thr-235 in the NqrB and to Thr-223 in the NqrC subunits. The ester<br />

binding of FMN to the threonine residue reported here is a new type of flavin<br />

attachment to polypeptide.<br />

POSTER <strong>AB</strong>STRACTS<br />

<strong>AB</strong>RF 2001 <strong>AB</strong>STRACTS<br />

P86-M<br />

GlycoSuiteDB—a database of glycan structures.<br />

C.A. Cooper, M.J. Harrison, M.R. Wilkins, N.H. Packer; Proteome Systems<br />

Ltd., North Ryde, Australia, 1/35-41 Waterloo Rd, North Ryde, NSW 2113,<br />

Australia<br />

GlycoSuiteDB is a relational database that contains information from the scientific<br />

literature on glycoprotein derived glycan structures, their biological<br />

sources, the literature references used to obtain the information, and the<br />

methods used to determine each glycan structure. The main aims in the construction<br />

of GlycoSuiteDB are to present a consistent, up-to-date and reliable<br />

source of information. The database provides an essential resource for the<br />

glycobiologist and the protein chemist.<br />

GlycoSuiteDB is available on the web at http://www.glycosuite.com. The<br />

web site allows the user to search the database using a combination of composition,<br />

monoisotopic or average mass, protein name, SWISS-PROT/TrEMBL<br />

accession number, species, biological system, tissue or cell type. There are<br />

at present no restrictions on the use of GlycoSuiteDB by non-profit organisations<br />

as long as its content is not modified in any way. Usage by and for<br />

commercial entities will require a licence after the initial free trial period on<br />

the web. Full conditions of use will be made available on the web site and<br />

through GeneBio (www.genebio.com), the exclusive worldwide distributor<br />

of GlycoSuiteDB.<br />

P88-S<br />

Disulfide bridge determination of SETI-IIa, a squash<br />

trypsin inhibitor.<br />

V.M. Faca, L.J. Greene; FMRP-USP, Av. Bandeirantes, 3900, Ribeirao Preto,<br />

Sao Paulo 14049-900 Brazil<br />

The squash trypsin inhibitor SETI-IIa, one of the smallest strong inhibitors<br />

described in the literature, contains 31 aminoacids residues of which 6 are<br />

cysteine residues. Its amino acid sequence is: EDRKCPKILMRCKRDSDCLAKC<br />

TCQESGYCG. It forms a compact tridimensional structure maintained by<br />

three disulfide bonds. Due to its small size, this inhibitor can be synthesized,<br />

using Fmoc chemistry. The reoxidation step which requires formation of the<br />

correct disulfide bonds is a critical step in obtaining the active inhibitor and<br />

knowledge of the correct disulfide pairing is required. We describe a procedure<br />

to determine the disulfide pairing of SETI-IIa using small amounts of<br />

protein. About 50 nmol (175 �g) of native SETI was submitted to digestion<br />

with thermolysin for 72 hours at 45�C in a 0.5 M MES buffer pH 6.5. The fragments<br />

obtained were submitted to RP-HPLC in a C18 column (4.6 � 220 mm)<br />

in a TFA/Acetonitrile elution system. The major fragments were collected an<br />

identified by amino acid composition and sequencing by Edman degradation.<br />

The disulfide bridges obtained from native SETI-IIa and their yields were:<br />

Cys1–Cys4 (50%), Cys2–Cys5 (10%) and Cys3–Cys6 (38%). This procedure<br />

will permit us to characterize refolded synthetic analogs of SETI-IIA<br />

Supported by FAPESP. Faça, V.M. has a FAPESP pre-doctoral fellowship.<br />

JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000 209


<strong>AB</strong>RF 2001 <strong>AB</strong>STRACTS<br />

P89-M<br />

Identification of posttranslational modifications using<br />

MALDI-TOF-TOF.<br />

J.P. DeGnore, J. Campbell, P. Juhasz, J. Peltier, M. Vestal; Applied<br />

Biosystems, 500 Old Connecticut Path, Framingham, MA 01701<br />

To realize the full potential of proteomics, large numbers of proteins must be<br />

rapidly identified, usually separated by 2D gels. To investigate the function<br />

of these identified proteins it is important to determine the potential posttranslational<br />

modifications. It has been estimated that 80% of mammalian<br />

cells contain posttranslational modifications (1) and up to one-third of proteins<br />

expressed in a typical mammalian cell are thought to be phosphorylated<br />

(2). Identification of the site and type of modification in a protein provides<br />

insight into the activity and regulation of the posttranslationally modified protein.<br />

The location of posttranslational modifications within a peptide fragment<br />

can be determined from mass shifts of the fragment ions in tandem mass<br />

spectrum. Often the expression of posttranslationally modified proteins is<br />

low, requiring sensitive detection. Tandem mass spectrometry has become<br />

the method of choice for identification of posttranslational modifications<br />

due to it sensitivity and speed. The new MALDI-TOF-TOF mass spectrometer<br />

allows both rapid protein identification and the determination of the precise<br />

site of posttranslational modifications. For example, signature ions in the<br />

low mass range can serve as a tool to screen for phosphopeptides in a high<br />

throughput experiment. Typical fragmentation patterns will be presented for<br />

posttranslational modifications including phosphorylation. A number of processing<br />

tools developed to allow quality peak lists to be derived from the raw<br />

mass spectra will be presented.<br />

(1) Kornfield, R.; Kornfield, S., Annu. Rev. Biochem. 1985, 54, 631–664.<br />

(2) Hubbard, M.J.; Cohen, P. Trends Biochem. Sci. 1993, 18, 172–177.<br />

P91-S<br />

The application of a novel precursor ion discovery method for<br />

the characterisation of phosphoproteins.<br />

J.I. Langridge1, J.B. Hoyes1, R.H. Bateman1, R.A. Carruthers1, C. Jones1, A. Millar1, O.N. Jensen2; 1Micromass UK Ltd., Floats Road, Wythenshawe,<br />

Manchester M23 9LZ, United Kingdom, 2Univ.of Southern Denmark,<br />

Odense Univ.<br />

The huge increase in genomic sequence information available, combined<br />

with the increased sensitivity and selectivity provided by mass spectrometry,<br />

has allowed large-scale protein identification. However the analysis of the<br />

post translational modifications present on the identified proteins is a more<br />

challenging problem.<br />

In this paper we describe a method that allows specific post translationally<br />

modified peptides to be identified and sequenced during the course of an<br />

HPLC experiment on the Q-Tof mass spectrometer. During the HPLC run the<br />

instrument is switched alternately at one-second intervals between low and<br />

high collision energy with argon in the collision cell. The quadrupole, MS1<br />

operates in the rf only mode allowing the full mass range to be passed to the<br />

Tof. The first data set at low energy (4 eV) shows only the normal pseudo<br />

molecular ions. The second at higher energy contains all their fragments.<br />

Wherever a product ion of interest occurs in the high-energy data all its possible<br />

precursors are present in the corresponding low energy data. The mass<br />

spectrometer may then switch to MS/MS mode selecting the potential precursors<br />

in turn to reveal the true parent. In addition to creating a mass list of<br />

precursors from the low energy spectra the computer may also make a list<br />

of neutral losses from these precursors. The exact mass capability of orthogonal<br />

Tof’s increase the specificity of the neutral loss particularly in the case<br />

of a mass deficient loss such as is observed with phosphopeptides. Appearance<br />

of the neutral loss causes the spectrometer to switch to MS/MS mode<br />

to acquire additional sequence information and confirmation of the neutral<br />

loss.<br />

Examples of this methodology and its application to protein phosphorylation<br />

will be presented.<br />

POSTER <strong>AB</strong>STRACTS<br />

210 JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000<br />

P90-T<br />

Immunoprecipitation as a method to enrich phosphoproteins<br />

destined for 2-D gel analysis.<br />

T.R. Shephard1, A. Ducret2; 1Concordia Univ., Canada, 1455 de Maisonneuve<br />

West, Montreal, Quebec H3G 1M8, Canada, 2Merck Frosst<br />

Our lab is interested in the construction of 2D phosphoprotein maps for<br />

determining the substrates of protein tyrosine phosphatase-1B (PTP-1B), a<br />

protein believed to play a role in the down regulation of the insulin receptor.<br />

The construction of 2D phosphoprotein maps is made difficult by the fact<br />

that 1) phosphoproteins are intermixed with all cellular proteins, complicating<br />

gel analysis, and 2) phosphoproteins often show relatively low abundance,<br />

which can result in important proteins being overlooked.<br />

To overcome the difficulties listed above, we attempted to enrich the phosphoprotein<br />

content of our cell lysates [obtained from control and insulin stimulated<br />

immortalized fibroblast cell lines derived from PTP-1B �/� and PTP-<br />

1B �/� mice (Elchebly, M. et al. 1999 <strong>Science</strong>. 283, 1544–1548)] through<br />

immunoprecipitation procedures before proceeding with our 2D gel analysis.<br />

This was accomplished by pre-clearing total cell lysates and then<br />

immunoprecipitating under native conditions with a mix of i) monoclonal<br />

4G10 antibody coupled to agarose beads and ii) biotin-conjugated RC20 antibody,<br />

as previously described by Lodish et al. (1999 Proc. Nat. Acad. Sci. USA<br />

97, 179–184). This poster will illustrate the 1D and 2D gel analysis of tyrosine<br />

phosphorylated proteins immunoprecipitated under a variety of conditions<br />

from PTP-1B �/� and PTP-1B �/� cell lines.<br />

P92-M<br />

Protein modifications in diabetics and individuals taking aspirin<br />

studied by mass spectrometric and functional assays of fibrinogen.<br />

A.H. Henschen-Edman; Univ. of California, Irvine, Biological <strong>Science</strong>s 2,<br />

Irvine, CA 92697-3900<br />

It is a well-known fact that proteins are modified posttranslationally both<br />

intracellularly and extracellularly. Certain protein modifications may be specifically<br />

caused by diseases or drugs. In the case of the human blood plasma<br />

protein fibrinogen, a central protein in blood coagulation, both structural and<br />

functional properties are altered by glucose in diabetics and by aspirin in<br />

individuals on that kind of medication. For the structure analysis fibrinogen<br />

was S-pyridylethylated, cleaved with cyanogen bromide and the resulting 36<br />

fragments partly separated by gel filtration chromatography; the pools were<br />

then analyzed by MALDI-TOF mass spectrometry. Glucose and aspirin modify<br />

amino groups by glycation and acetylation respectively, thereby adding<br />

the corresponding 162 and 42 amu to the original mass of the fragment.<br />

Under in vitro conditions extensive modifications and dose-dependent<br />

increases in the degrees of modification were observed. Additional evidence<br />

for the reaction with glucose was obtained by sequence analysis of tryptic<br />

fragments retained by a boronate affinity column; several predicted lysines<br />

were missing, as they were present in the glycated form. For the function<br />

analysis fibrinogen was clotted with thrombin and the proteolytic cleavage<br />

as well as the polymerization-dependent increase in absorbance monitored.<br />

The effect of glycation and acetylation on the proteolysis was marginal, but<br />

both caused an extensive and dose-dependent decrease in the polymerization<br />

reaction. These effects are expected to be of pathophysiological relevance.


P93-T<br />

Analytical strategies for the identification of phosphorylation<br />

and O-linked glycosylation sites in proteins.<br />

A. Furtos-Matei1, J. Kihlberg2, P. Thibault1; 1Natl. Res. Council, Canada,<br />

2Umea Univ.<br />

Glycosylation and phosphorylation are between the most common posttranslational<br />

modifications in proteins. Over the past decade, tandem mass<br />

spectrometry has become the method of choice for mapping attachment sites.<br />

In contrast with N-linked glycans, oligosacharides attached to ser/thr residues<br />

tend to fragment at the glycosidic bond on MS-MS to leave an unmodified<br />

peptide backbone, thereby making assignment of modification sites more difficult.<br />

A similar situation is also encountered for phosphopeptides analysis<br />

where the interpretation of positive ion MS-MS can be more complicated in<br />

view of fragment showing a loss of H 3 PO 4 moiety. In this work, synthetic glycopeptides<br />

and phosphopeptides were used to develop a method that allows<br />

simultaneous determination of phosphorylation and O-linked glycosylation<br />

sites at serine and threonine residues. The approach we propose is based on<br />

a ?-elimination reaction followed by a Michael type addition of a nucleophilic<br />

reagent, which is further used as a marker of the modification site. Among<br />

other reagents, the volatile triethylamine was selected to induce the elimination<br />

of both, phosphate groups and O-linked glycans. Several nucleophilic<br />

reagents have been tested and evaluated by mass spectrometry and capillary<br />

electrophoresis methods with respect to the chemical yield and the specificity<br />

toward the targeted functional groups. The relative reactivity of different<br />

attachment sites as a function of the amino acid nature and the type of modification<br />

is also discussed. This approach has been applied to enzymatic digestion<br />

mixtures of model glyco- and phosphoproteins.<br />

P95-M<br />

Structural properties of DsRed, a fluorescent protein from coral.<br />

L.A. Gross1, G.S. Baird1, D.A. Zacharias1, R.C. Hoffman1, K.K. Baldridge2, R.Y. Tsien1; 1UCSD, 9500 Gilman Dr., La Jolla, CA 92093-0647, 2San Diego<br />

Supercomputer Ctr.<br />

With the cloning of a red fluorescent protein from the Discosoma species of<br />

marine corals (Matz et al. 1999), an entire spectrum of colors are now available<br />

as gene reporters in cell microscopy. Similar to green fluorescent protein<br />

(GFP) derived from the jellyfish Aequorea, with which it shares only 30%<br />

homology, DsRed develops its fluorescence without the need for exogenous<br />

enzymes or co-factors. Studies are on-going to optimize its properties, especially<br />

for applications involving fluorescence resonance energy transfer<br />

(<strong>FR</strong>ET).<br />

DsRed is an oligomer of four �-barrels, as shown by analytical ultracentrifugation<br />

(Baird et al. 2000). Tandem mass spectrometry has shown that formation<br />

of the red chromophore involves a second step of oxidation, in comparison<br />

to the cyclization and oxidation which forms GFP (Gross et al. 2000).<br />

This extends the conjugation by adding an acylimine functional group at the<br />

2-position of the imidazolidinone ring of the GFP chromophore. Initially<br />

upon expression, DsRed emits at green wavelengths. The green color diminishes<br />

as the protein matures to its red fluorescent form, in a process taking<br />

3 to 4 days. However, the protein appears not to convert the total population<br />

of chromophores to the mature, red form. In the tetramer, <strong>FR</strong>ET occurs<br />

between immature green (as donors of resonance energy) and mature red<br />

emitters (as acceptors), accounting for the observed fluorescence spectra.<br />

Matz, M. V., Fradkov, A. F., Labas, Y. A., Savitsky, A. P., Zaraisky, A. G.,<br />

Markelov, M. L. & Lukyanov, S. A. (1999) Nature Biotechnology 17, 969–973.<br />

Baird, G. S., Zacharias, D. A. & Tsien, R. Y. (2000) Proc. Natl. Acad. Sci. USA<br />

97, 11984–89.<br />

Gross, L. A., Baird, G. S., Hoffman, R. C., Baldridge, K. K. & Tsien, R. Y.<br />

(2000) Proc. Natl. Acad. Sci. USA 97, 11990–95.<br />

POSTER <strong>AB</strong>STRACTS<br />

<strong>AB</strong>RF 2001 <strong>AB</strong>STRACTS<br />

P94-S<br />

Comparative phosphopeptide mapping from gel-derived proteins<br />

using a multidimensional mass spectrometry approach.<br />

R.S. Annan, M.J. Huddleston, F. Zappacosta, C.I. Susan, S.A. Carr;<br />

SmithKline Beecham Pharmaceut., Box 1539, King of Prussia, PA 19406<br />

Within the last ten years mass spectrometry has emerged as a key technology<br />

in the difficult task of mapping protein phosphorylation sites. Previously<br />

we have described a comprehensive multidimensional phosphopeptide mapping<br />

strategy which uses mass spectrometry to isolate and identify phosphorylated<br />

peptides. We have now significantly improved the sensitivity of<br />

this approach by incorporating capillary HPLC chromatography (0.18 mm<br />

i.d.) and micro-ionspray mass spectrometer. The current configuration allows<br />

us to detect and identify less than 200 fmol of phosphopeptides from SDS-<br />

PAGE derived proteins. Because the first dimension of the analysis utilizes an<br />

LC separation coupled to on-line phosphopeptide detection, it generates a<br />

phosphopeptide profile. In this report we will show how such a profile can<br />

be used for comparative purposes, providing a semi quantitative evaluation<br />

of how the phosphorylation state of a protein changes under a given set of<br />

differing conditions. The comparative profile makes it easier to identify and<br />

concentrate analysis efforts on those phosphorylation sites which are relevant<br />

to the changing conditions and ignore those which are not.<br />

P96-T<br />

Mass spectrometry analysis of the kinetics of in vivo rhodopsin<br />

phosphorylation.<br />

K.A. Lee, G.A. Niemi, J.B. Hurley; Univ. of Washington, Howard Hughes<br />

Medical Institute, Box 357370 Health <strong>Science</strong>s Building, Seattle, WA 98195<br />

Upon light stimulation, rhodopsin, the light-sensing protein in the rod cells<br />

of the retina, is phosphorylated at several sites as the first step in its deactivation.<br />

We developed a mass spectrometry-based method to determine the<br />

kinetics of this phosphorylation in vivo by quantitating the extent of phosphorylation<br />

at each site. After exposing a freshly dissected mouse retina to<br />

a flash of light, we use a rapid-quench device is used to quickly terminate<br />

phosphorylation and dephosphorylation reactions by homogenizing the<br />

retina in 8 M urea. The samples are washed and proteolyzed to release the<br />

carboxyl-terminus of rhodopsin, which contains all known phosphorylation<br />

sites. The resultant peptides are analyzed by LCMS. Synthetic monophosphorylated<br />

and unphosphorylated peptide standards were used to show<br />

that the mass spectral response to the rhodopsin peptides is linear over a<br />

range from 10 fmol to 100 pmol. The relative sensitivities of the mass spectrometer<br />

to unphosphorylated, singly, doubly, and triply phosphorylated<br />

peptides were determined using synthetic peptide standards. The resultant<br />

correction factors were used to determine the relative quantities of the peptides<br />

in the actual retinal samples. LCMS/MS was utilized to identify three primary<br />

sites of phosphorylation, S334, S338, and S343. Peptides monophosphorylated<br />

on S334 were separable from those monophosphorylated on<br />

S338 and S343 by reverse phase HPLC with our standard C18 column chromatography<br />

conditions. However, the S338 and S343 monophosphorylated<br />

peptides coeluted. We demonstrated that the relative amounts of each species<br />

in the single peak could be determined by monitoring the ratio of specific<br />

daughter ions characteristic of each peptide.<br />

JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000 211


<strong>AB</strong>RF 2001 <strong>AB</strong>STRACTS<br />

P97-S<br />

An improved procedure using immobilized metal-ion affinity<br />

chromatography for isolation and characterization of<br />

phosphopeptides from phosphoproteins.<br />

K-L. Hsi, D.R. Dupont, S.W. Yuen, C.A. Settinery; Applied Biosystems,<br />

Foster City, CA, 850 Lincoln Center Drive, Foster City, CA 94404<br />

It has become clear that phosphorylation is the most common and important<br />

reversible regulatory modification of proteins. Defining the sites of phosphorylation<br />

is necessarily important for the mechanism studies of cell regulation.<br />

However, methods to specifically isolate and characterize phosphopeptides<br />

remain challenged, especially for those proteins separated from<br />

1D/2D gel at low concentration. An attempt is thus made to improve the procedure<br />

based upon immobilized metal-ion affinity chromatography (IMAC)<br />

for working with sub-microgram amount of phosphoproteins.<br />

A protease digest of gel-separated phosphoprotein was treated with immobilized<br />

Fe��� on a chelating resin. The affinity bound phosphopeptides were<br />

then eluted from the resin with ammonium acetate buffer. We use a mini-size<br />

(250 �l) of cartridge to perform all the processes of isolation of phosphopeptides<br />

including washing of the resin, binding and elution of the phosphopeptides,<br />

instead of an open column as previously reported.<br />

This improved procedure provides some advantages over the previously<br />

reported open column method: 1. Fast, i.e. 30 minutes or less. 2. High washing<br />

efficiency, thus high recovery of pure phosphopeptides. 3. Sensitive<br />

working level, i.e. sub-microgram of gel separated phosphoproteins.<br />

Several phosphoproteins with different types of phosphorylation (Ser, Thr<br />

and Tyr) have been studied using the improved procedure. The effectiveness<br />

of the procedure was demonstrated by chemical sequencing and MS/MS<br />

analysis of MicroBlotter-collected phosphopeptides.<br />

P99-T<br />

Synthesis of lactam-bridged dipeptides mediated by<br />

aminoacylpyroglutamates.<br />

I. Rodionov, A. Chulin, V.T. Ivanov; Br. of Shemyakin-Ovchinnikov Inst.<br />

of Bioorganic Chem., 8 Academy Avenue, Pushchino, Moscow Region<br />

142290, Russian Federation<br />

Aminoacyl incorporation reaction (AI) discoverd about 40 years ago by Shemyakin<br />

et al. has been evaluated as a general synthetic route to variously<br />

constrained lactam-bridged dipeptides. The AI is effectively a ring enlargement,<br />

which involves intramolecular acylation of amino group (or different<br />

nucleophilic functions like HS- and HO-) by the activated cyclic diacylamino<br />

moiety via intermediate formation of bicyclic azacyclols. The most straightforward<br />

models for studying AI are aminoacylated pyroglutamates derived<br />

from diaminoacids<br />

R-Xaa-Glp-OR� (I), Xaa � Lys, Orn, Dab and Dpr,<br />

a class of unusual peptides, which remains unexplored as yet. We have studied<br />

3 different synthetic approaches to I:<br />

direct acylation of sodium derivative of Glp-OR�;<br />

pyrrolidone ring closure in the plain dipeptides R-Xaa-Glu(OX)-OR� promoted<br />

by bases and/or by activation of side chain carboxyl of Glu;<br />

selective oxidation of the related proline dipeptides Boc-Xaa-Pro-OR� by<br />

ruthenium tetroxide/sodium periodate.<br />

Scope and limitation of the above approaches will be discussed and exemplified<br />

by the synthesis of 26 protected dipeptides I. As expected, deprotection<br />

of amino or hydroxy functions followed by exposure to potassium carbonate<br />

buffer, pH 9.5 (water-acetonitrile) resulted in smooth AI for the<br />

majority of the synthesized dipeptides I. No oligomerization products were<br />

detected. In this way a number of bridged dipeptides (9–12 membered<br />

cycles) were obtained in 35–70% yield and characterized by NMR and MS<br />

data. NMR data suggested that in 4 cases stable azacyclols have been isolated.<br />

POSTER <strong>AB</strong>STRACTS<br />

212 JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000<br />

P98-M<br />

Postsynthesis labeling of agiotensin II peptide with d-rhodamine<br />

for fluorescence monitoring of receptor binding.<br />

S.P. Yadav, W-Z. Shen, Y. Luo, J. Zhang, S. Karnick; Lerner Res. Inst.,<br />

Cleveland, Mail Code NC10, 9500 Euclid Avenue, Cleveland, OH 44195<br />

Solid-phase peptide synthesis is now routinely used for drug discovery and<br />

immunological research and for studying the structure-function relationships<br />

of proteins. Protein-protein interactions play an important role in a wide variety<br />

of biochemical and physiological processes and generally involve large<br />

interfaces with many intermolecular contacts. In addition, protein-protein<br />

interaction may also occur through small surface binding epitopes such as in<br />

the case of human growth hormone-receptor binding and erythropietinreceptor<br />

complex formation. Therefore, the rational design of relatively small<br />

molecular size peptide activators/inhibitors for receptor surfaces has long<br />

been considered a formidable challenge. These findings convincingly open<br />

up the possibility that small peptides that mimic such small binding epitopes<br />

may help to block a large protein-protein interface. Usefulness of this hypothesis<br />

in drug design rationale remains to be further tested in different biological<br />

systems. Furthermore rationalization of general approach of inhibition of<br />

protein-protein interaction will of course have a tremendous impact on<br />

development of new therapeutic strategies for several human diseases. That<br />

is where fluorescent dye labeling of synthetic peptides becomes important.<br />

Studies on labeling of a peptide with fluorescent dyes in a manner without<br />

altering functional characteristics of the peptide are of considerable interest<br />

for studying the peptide-receptor interactions. Strategy for synthesis of an<br />

octa-mer angiotensin II by Fmoc chemistry and subsequent labeling of the<br />

cleaved peptide with Rhodamine dye are discussed in this report. The results<br />

of double labeling are presented here to show that the removal of excess free<br />

Rhodamine is critically important in confocal microscopy.<br />

P100-S<br />

The characterization of prostate specific antigen activating peptide.<br />

M. Pakkala1, P. Wu2, J. Leinonen2, U-H. Stenman2, J. Vepsäläinen1, A. Närvänen1; 1Univ. of Kuopio, P.O. Box 1627, Kuopio 70211, Finland,<br />

2Univ. Central Hosp., Helsinki<br />

Prostate specific antigen (PSA) is widely used as a marker of prostate cancer.<br />

PSA is a 30 kD serine protease and belongs to Kallikrein family. Peptides<br />

with specific binding properties to PSA were studied by using phage display<br />

peptide libraries. Four different PSA binding clones were isolated and characterized.<br />

One of the clones, containing 13 amino acids sequence with four<br />

cysteines, showed the highest affinity for PSA. This peptide (C4), as a fusion<br />

protein with glutathione-S-transferase (GST), increased the protease activity<br />

of PSA against a synthetic substrate (Wu et al. (2000), Eur. J. Biochem. 267,<br />

1–10)<br />

We have synthesized the active peptide C4 (CVAYCIEHHCWTC) and its modifications<br />

by using conventional solid phase peptide synthesis method (SPPS).<br />

Two cysteines (2. And 3.) were protected by using Acm protecting group,<br />

which remains uncleaved during the removal of the peptide from the resin.<br />

The final cyclization was made during the removal of the Acm-group from<br />

cysteines by Iodine treatment. The synthetic analog showed same activity to<br />

PSA than the original GST fusion protein where as the activity of the peptide<br />

with two Acm groups was less than 50% from the fully cyclized form. The<br />

acetylation of the ?-amino group did not affect to the activity. In addition we<br />

synthesized cyclic version with amide bond VAYCIEHHCWT instead of the<br />

cysteines 1. and 4. by using Allyl protected E(6) (Fmoc-L-Glu-OAll) as a starting<br />

amino acid. The final peptide was cyclic with one disulfide bond. This<br />

modification was totally inactive.<br />

The results suggest that the correct secondary structure of the peptide C-4<br />

plays an important role in increased activation of PSA. Structure analysis by<br />

NMR is under study. Based on the preliminary NMR results the prepared peptides<br />

are rather flexible and the structures are strongly dependent on the temperature.


P101-M<br />

Implementing surface plasmon resonance biosensors in<br />

drug discovery.<br />

D.G. Myszka; Univ. of Utah, 50 N. Medical Dr./School of Medicine Rm<br />

4A417, Salt Lake City, UT 84132<br />

Recent improvements in instrument hardware, experimental design, and<br />

data processing make it possible to utilize surface plasmon resonance (SPR)<br />

biosensor technology in the discovery and development of small-molecule<br />

drugs. The key features of SPR biosensors, real-time binding analysis and lack<br />

of labeling requirements, make this technology suitable for a wide range of<br />

applications. Current instruments have a throughput of �100–400 assays per<br />

day, providing a complement to high-throughput screening. The ability to<br />

collect kinetic data on compounds binding to therapeutic targets yields new<br />

information for lead optimization. Small-molecule analysis and emerging<br />

applications in the areas of ADME and proteomics have SPR biosensors<br />

poised to play a significant role in the pharmaceutical industry.<br />

P103-S<br />

Mapping of protein-protein interactions using mass spectrometry.<br />

D. Figeys, H. Duewel; MDS-Ocata, Toronto, 480 University Avenue<br />

Suite 401, Toronto, Ontario M5G 1V2, Canada<br />

Functional proteomics is becoming the next generation of large-scale proteomic<br />

approaches. It is based on the concept that the function of a protein<br />

is defined by its interactions. Therefore, large-scale approaches for the mapping<br />

of protein-protein interactions at the cellular level will be essential to<br />

the comprehensive understanding of the interactome, i.e. the protein-protein<br />

interactions related to a proteome. Here we will present an approach for the<br />

large-scale screening of the interactome using mass spectrometry. This technology<br />

identifies proteins involved in specific protein-protein interactions<br />

using protein identification by mass spectrometry. We will also discuss the<br />

incorporation in the process of novel technology, such as the ICATtm technology,<br />

for the rapid and relative quantitation of proteins by mass spectrometry.<br />

POSTER <strong>AB</strong>STRACTS<br />

<strong>AB</strong>RF 2001 <strong>AB</strong>STRACTS<br />

P102-T<br />

Study of the protein-DNA interaction responsible of the carbon<br />

catabolite repression in Lactobacillus casei.<br />

C.D. Esteban1, K. Mahr2, G. Pérez-Martínez1, W. Hillen2, F. Titgemeyer2; 1Agrochem. and Food Technol. Inst., Burjassot, Spain, Apartado de<br />

correos 73, Burjassot, Valencia 46100 Spain, 2Erlangen-Nürnberg Univ.<br />

In the industrially relevant lactic acid bacterium Lactobacillus casei the preferential<br />

utilization of carbon sources is controlled by the mechanism of carbon<br />

catabolite repression (CCR). As in other low-G�C gram-positive bacteria,<br />

CCR takes place through the binding of the transcriptional repressor,<br />

CcpA, to an operator called cre (catabolite responsive element). CcpA binding<br />

to cre sequences is enhanced by its correpressor HPr-ser46-P.<br />

It was our aim to characterize this protein-DNA interaction by Surface Plasmon<br />

Resonance (SPR). For this purpose CcpA was overexpressed and purified<br />

both with an N-terminal poly-histidine tag and without this tag. HPr was<br />

overexpressed, purified and in vitro phosphorylated. A synthetic biotinylated<br />

double stranded oligonucleotide containing the cre sequence present in the<br />

promoter of the lac operon of L. casei was immobilized on a streptavidin sensor<br />

chip. SPR experiments were performed flowing a range of CcpA (with<br />

and without his tag) concentrations over the chip in the presence and<br />

absence of saturating amounts of HPr-ser46-P. The experimental parameters<br />

for data acquisition were optimized and equilibrium and semiquantitative<br />

kinetic analysis allowed the calculation of KD and estimation of kinetic association<br />

and dissociation rate constants for protein-DNA interaction.<br />

The experimental parameters for the study of this protein-DNA interaction by<br />

SPR have been established. Binding of unmodified CcpA to the cre sequence<br />

was characterized by equilibrium and kinetic rate constants. The presence of<br />

the his tag was shown not to interfere (results with and without his tag are<br />

comparable). In conclusion, we have developed a powerful system to accurately<br />

characterize the intermolecular relationships of histidine tagged CcpA<br />

that could allow efficiently characterizing mutations in CcpA in the near<br />

future.<br />

P104-M<br />

Affinity chromatography and mass spectrometry in dissecting<br />

EGFr signaling interdicted by the quinazoline EGFr inhibitor<br />

OSI-774.<br />

J.D. Haley1, A. Thelemann1, H. Pan1, D. Fenyo2; 1OSI Pharmaceut. Inc.,<br />

106 Charles Lindbergh Blvd., Uniondale, NY 11553, 2Proteometrics LLC<br />

The blockade of EGFr signaling by the quinazoline tyrosine kinase inhibitor<br />

OSI-774 was investigated by direct affinity chromatography, reverse-phase<br />

chromatography and mass finger-printing. Phosphotyrosine complexes from<br />

OSI-774 and control treated HN5 squamous carcinoma cells were prepared<br />

by both Triton X-100 and RIPA lysis. Protein fractionation by SDS-PAGE was<br />

compared with capillary HPLC. Protein fractions were proteolytically digested<br />

with either trypsin, GluC or LysC, desalted by microC18 reverse phase tips<br />

and subjected to matrix assisted laser desorption- time of flight mass spectrometry.<br />

C4 chromatography greatly improved the signal strength and resolution<br />

of the peptide spectra obtained, when compared to direct MALDI MS<br />

of digested immunoaffinity fractions.<br />

Eighty-eight spectra were evaluated from five HN5 phosphotyrosine protein<br />

complex chromatography separations. Mass analysis was performed using a<br />

PerSeptive DE-Pro mass spectrometer using a-cyano-4-hydroxycinnamic acid<br />

or dihydrobenzoic acid matrices. Data were analyzed using RADARS, a SQL<br />

compliant database search engine allowing comparison across experiments<br />

of proteins identified under different experimental conditions (e.g. trypsin vs.<br />

GluC). The predominant protein identified was the epidermal growth factor<br />

receptor (EGFr) which was found in the major C4 HPLC protein fraction.<br />

Phosphorylation on both P1 and P2 C-terminal tyrosines was readily observed<br />

by mass spectrometry. A large number of well known and less defined<br />

proteins which either (1) contain phosphotyrosine or (2) form stable complexes<br />

with targets proteins were identified from multiple experiments. The<br />

Triton X100 lysis and affinity capture methodology allows the identification<br />

of known and novel protein complexes not detected by gel-based techniques.<br />

JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000 213


<strong>AB</strong>RF 2001 <strong>AB</strong>STRACTS<br />

P105-T<br />

Innovations in proteome analysis: biomolecular interaction<br />

analysis mass spectrometry.<br />

R.W. Nelson, D. Nedelkov; Intrinsic Bioprobes Inc., 625 S. Smith Rd.<br />

Suite 22, Tempe, AZ 85281<br />

The relative ease and fast pace by which the genomic data has been gathered<br />

stands out against the complexity of the proteome world and the<br />

involvedness required for protein characterization. Even though significant<br />

strides have been made lately in several protein characterization techniques,<br />

novel technologies and multiplexation of the existing ones are required to<br />

fully address the many sides of the proteome.<br />

Biomolecular Interaction Analysis Mass Spectrometry (BIA/MS) is a twodimensional<br />

chip-based analytical technique geared toward quantitative and<br />

qualitative detection and analysis of small volumes of biological samples. In<br />

the first (functional) dimension, BIA/MS takes a form of micro-scale planaraffinity<br />

chromatography performed on a sensor-active surface. Surface plasmon<br />

resonance (SPR) is used for detection of biorecognition events that<br />

occur at a sensor surface/solution interface between an immobilized biomolecule<br />

(the ligand) and its interacting partner (the analyte) present in the<br />

sample solution. For the second (structural) dimension, BIA/MS employs<br />

MALDI-TOF MS analysis. With minimal physical modifications and thorough<br />

application of MALDI matrix, SPR-active sensor surfaces are converted to<br />

amenable MALDI target. The ensuing mass spectrometry analysis serves the<br />

purpose of validating the SPR sensing data by providing the molecular mass<br />

of the retained analyte and it yields other qualitative information about the<br />

SPR-monitored interaction, such as identification of non-specific binding,<br />

binding of analyte variants/fragments and multi analyte binding. Presented<br />

here are results from the utilization of BIA/MS in detection of a number of<br />

biological markers found in complex biological fluids. Small volumes of<br />

human plasma and urine were analyzed for cystatin C, beta-2-microglobulin,<br />

urinary protein 1, retinol binding protein and transthyretin, exploring the<br />

effectiveness of BIA/MS in simultaneous detection of clinically related biomarkers.<br />

Issues such as limit of detection, recognition of protein complexes<br />

and delineation of non-specific binding were also explored.<br />

P107-M<br />

Miniature integrated surface plasmon resonance biosensor for<br />

characterization of protein-protein interactions.<br />

M.L. Stolowitz, G. Li, K.P. Lund, J.P. Wiley; Prolinx, Inc.,<br />

22322 20th Avenue SE, Bothell, WA 98021<br />

The utility of a miniature integrated surface plasmon resonance biosensor is<br />

described. The disposible device is approximately the size of a thumbnail and<br />

houses all of the optics and electronics needed to acquire surface plasmon<br />

resonance sensorgrams. The gold sensor surface has been modified so as to<br />

minimize nonspecific binding and utilizes Versalinx Chemical Affininty Tools<br />

to facilitate the immobilization of macromolecular targets for binding studies.<br />

The utility of the biosensor is demonstrated in conjunction with a prototype<br />

data acquisition interface and a simple orbital shaking device.<br />

POSTER <strong>AB</strong>STRACTS<br />

214 JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000<br />

P106-S<br />

Dissecting structure-function relationships in RNA/protein<br />

interaction using biocore.<br />

I.A. Laird-Offringa1, P.S. Katsamba1, D.G. Myszka2; 1Univ. of Southern<br />

California, 1441 Eastlake Ave., Los Angeles, CA 90089-9176, 2Univ. of Utah<br />

RNA-binding proteins play critical roles in gene expression and regulation at<br />

the post-transcriptional level. While much is known about the various naturally<br />

occurring RNA-binding motifs, and co-crystal structures of a number of<br />

RNA/protein complexes are available, very little is known about the dynamics<br />

of RNA/protein interactions. We have used the spliceosomal protein U1A<br />

and its RNA target in the U1 small nuclear RNA (U1hairpinII or U1hpII) as a<br />

model to study the kinetics of RNA/protein interaction. Using the previously<br />

solved structure of the U1A/U1hpII complex, we have engineered a series of<br />

mutants designed to probe the roles of electrostatics, hydrogen bonding, aromatic<br />

stacking, and RNA loop length, all of which have been implicated in<br />

formation of the U1A/U1hpII complex. The effects of these mutations on the<br />

binding dynamics were studied using BIACORE, which yielded high quality<br />

kinetic data about the interaction. We determined that neutralization of positive<br />

charges on the protein slows the association rate and reduces the deleterious<br />

effect of salt on complex formation. In contrast, removal of hydrogenbonding<br />

or stacking interactions within the RNA/protein interface, or<br />

reducing the size of the RNA loop, increases the dissociation rate. Our data<br />

support a mechanism of binding consisting of a rapid initial association<br />

based on electrostatic interactions and a subsequent locking step based on<br />

the hydrogen bonding and stacking interactions that occur during the<br />

induced fit of RNA and protein. Our results demonstrate the power of BIA-<br />

CORE to dissect the functional differences between structural features of two<br />

interacting macromolecules.<br />

P108-T<br />

The study of peptide-peptide interaction by ion-mobility MALDI.<br />

A.S. Woods1, J. Koomen2, M.A. Huestis1, K.J. Gillig2, D.H. Russell2, A.J. Schultz3, K. Fuhrer3, M. Gonin3; 1NIDA, NIH, 5500 Nathan Shock<br />

Drive, Baltimore, MD 21224, 2Texas A&M Univ., 3Ionwerks Inc.<br />

We showed in previous work that MALDI could be used to study peptidepeptide<br />

interactions. Matrices such as ATT [pH 5.4] do not disrupt non-covalent<br />

interactions, while more acidic matrices such as CHCA [pH 2.0] do. Our<br />

study found that one peptide had to have 2 adjacent Arg [RR] or an Arg-Lys-<br />

Arg [RKR] motif and the other had to have a minimum of two adjacent Glu<br />

[EE] or Asp [DD] in order to form a complex.<br />

In this work we used MALDI/mobility/TOF mass spectrometry to further<br />

study the formation of these non-covalent complexes [NCX]. The instrument<br />

consists of a short drift tube (the ion-mobility cell) in which is applied an<br />

electric field which causes the MALDI ions to drift through a Helium carrier<br />

gas (2 torr) into an orthogonal TOF mass spectrometer. Ion-mobility [IM] separates<br />

gas phase ions on the basis of their collision cross section-to-charge<br />

ratio, when combined with mass spectrometry it can be a powerful instrument<br />

for structural studies to determine the conformations of biomolecules.<br />

Dynorphin (a 17 a.a. peptide that contains an RR motif) and several other<br />

peptides containing two RR or RKR motifs formed NCX with acidic peptides<br />

(containing 2 or more adjacent Glu or Asp) and were detected by IM and<br />

TOF MS. We discuss the capabilities and limitations of this new instrument<br />

when applied to the gas phase conformational study of MALDI desorbed<br />

NCX, as well as the advantages of looking at peptide mixtures by IM in addition<br />

to MALDI-TOF MS


P109-S<br />

Protein molecular communications involving two or more<br />

different ligands studied by microcalorimetry.<br />

M.L. Doyle; Bristol-Myers Squibb Pharmaceut. Res. Inst., Mail Stop H13-07,<br />

206 Provinceline Rd., Princeton, NJ 08543-4000<br />

Biophysical methods are capable of characterizing the functional chemistry<br />

of protein-ligand interactions at a high, molecular level of resolution. Typically,<br />

biophysical studies have focused on interactions involving one protein<br />

with a single ligand. However, the biological roles of many proteins are likely<br />

to be regulated by interactions with multiple ligands. In this tutorial we<br />

describe how biophysical methods, particularly Isothermal Titration Calorimetry<br />

(ITC), can be used to begin to deconvolute the interdependency that<br />

different multiple ligands have on a protein’s function. Two examples will be<br />

presented. One case involves comparing the binding interactions of a TNF<br />

cytokine with four different members of the TNF receptor superfamily in<br />

order to predict which receptor may be most biologically relevant (1). The<br />

other example describes a case where two different small molecules regulate<br />

the binding of one another to an enzyme (2).<br />

<strong>Reference</strong>s<br />

1) Truneh, A., Sharma, S., Silverman, C., Khandekar, S., Reddy, M.P., Deen,<br />

K.C., Mclaughlin, M.M., Srinivasula, S.M., Livi, G.P., Marshall, L.A., Alnemri,<br />

E.S., Williams, W.V. and Doyle, M.L. (2000) J. Biol. Chem. 275, 23319–23325.<br />

2) Du, W., Liu, W-S., Payne, D.J. and Doyle, M.L. (2000) Biochemistry 39,<br />

6003–6011.<br />

P111-T<br />

Long-range fragment sizing on MegaBACE genotyping instrument.<br />

M. Minarik, K. Pirkola, M. Mahtani; Molecular Dynamics,<br />

928 E. Arques Ave., Sunnyvale, CA 94085<br />

Over the past decade, DNA fragment analysis by capillary electrophoresis has<br />

become a powerful alternative to classic sizing by slab gel electrophoresis.<br />

The most important applications include microsatellite genotyping, AFLP<br />

fingerprinting, differential display and SNP typing. Due largely to slab gel limitations,<br />

most current protocols are based on fragment sizing only 300–500<br />

bp. Extending this size range is desirable in order to accelerate throughput<br />

by increasing either the depth of microsatellite multiplexing, or the effectiveness<br />

of fingerprinting and differential display applications. Recently published<br />

results from long-read DNA sequencing suggest a possibility to separate<br />

DNA fragments over a large size range with high resolution (Zhou et al.,<br />

Anal Chem 2000). The key factors are proper composition of separation<br />

matrix and optimal selection of experimental parameters defining injection<br />

and separation conditions.<br />

In the present work, we have evaluated a sizing precision of large fragments<br />

up to several kilobases. Using optimized running conditions, we demonstrate<br />

routine sizing up to 1 kb with 2 base resolution in 120 minutes. For longer<br />

fragments, the sizing resolution was between 5 and 10 bases with analysis<br />

times under 3 hours. We present the influence of various experimental conditions<br />

on the peak resolution and its impact on the overall quality of sizing.<br />

POSTER <strong>AB</strong>STRACTS<br />

<strong>AB</strong>RF 2001 <strong>AB</strong>STRACTS<br />

P110-M<br />

Protein biochips: powerful new tools in proteomics.<br />

K.L. Witte1, F. Zaugg1, P. Indermuhle1, L. Ruiz-Taylor1, N. Tolani1, B. Muehlbauer1, P. Wagner1, S. Nock2; 1Zyomyx, Inc., 3911 Trust Way,<br />

Hayward, CA 94545, 2Zyomyx, Inc.<br />

With the imminent sequencing of the human genome there is a growing<br />

need for technologies, which can study the resulting wealth of gene products.<br />

While technological innovation has adapted the analysis of genetic<br />

material to a highly parallel and miniaturized format, the more delicate<br />

nature of protein structures has precluded the development of analogous<br />

devices. However, to achieve a fundamental understanding of biochemical<br />

pathways, high-throughput biology has to be expanded to protein analysis,<br />

proteomics and multi-target screening.<br />

We have developed high-density protein microarrays for quantification of<br />

multiple proteins in complex mixtures. These microarrays contain 10.000 individually<br />

addressable features in a 1 cm � 1cm area. The implementation<br />

of new surface chemistries allows the immobilization of exactly defined<br />

quantities of proteins on each spot while retaining the activity of the protein.<br />

Using this platform we have developed a multiplexed, microchip-based<br />

immunoassay to analyze expression levels of serum proteins. We demonstrate<br />

the binding of proteins of interest with very low non-specific binding<br />

from non-cognate proteins using fluorescence as readout. Detection limits on<br />

this microassay are equal to or lower than commercial ELISA tests and reduce<br />

the sample volume by many orders of magnitude.<br />

P112-S<br />

High throughput SNP genotyping on MegaBACE using multiple,<br />

time-spaced injections.<br />

A. Shuster1, D. Shen1, Y. Tsunoi1, M. Minarik1, K. Pirkola1, K. Jones1, T. Deldot1, R. Belcinski1, A. Mamone2, K. Dains1, M. Mahtani1; 1Molecular<br />

Dynamics, 928 E. Arques Ave., Sunnyvale, CA 94085, 2Molecular Dynamics<br />

We have developed a method for high throughput SNP genotyping on the<br />

MegaBACE 1000 capillary electrophoresis platform. The system takes<br />

advantage of the instrument’s flexibility to load 12 96-well sample plates over<br />

a period of about 30 minutes, using a proprietary method of repeated, timespaced<br />

injections. Samples are prepared in a single-tube SNuPe (Single<br />

Nucleotide Primer Extension) reaction which contains the four different fluorescently-labeled<br />

dideoxynucleotides (terminators), Thermosequenase enzyme<br />

and optimized reaction buffer. After single base extension and cleanup,<br />

SNP products are loaded onto the MegaBACE by automated electrokinetic<br />

injection. Multiple, pulsed injections of up to 12 different SNP marker plates<br />

are then loaded into the same polymer matrix, spaced at two minute electrophoresis<br />

intervals. Separation provides the advantage of excellent signalto-noise,<br />

predictable and characteristic marker genotype patterns, and good<br />

discrimination of negative controls over sample failures. 1300 SNP genotypes<br />

can be obtained with a turnaround time of less than 2 hours; we expect that<br />

further color and size multiplexing will greatly increase throughput. A characteristic<br />

injection marker is added to each sample prior to injection, acting<br />

both as internal control and also demarcating each injection from the next.<br />

A separate software package, SNP Profiler automatically processes the signal<br />

data and outputs the maximum likelihood SNP genotypes, and includes a<br />

user interface for editing and verification. The system can be completely<br />

switched between any three applications—high throughput sequencing, SNP<br />

typing or microsatellite genotyping—in less than five minutes.<br />

JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000 215


<strong>AB</strong>RF 2001 <strong>AB</strong>STRACTS<br />

P113-M<br />

An automatable magnetic particle-based kit for the removal of<br />

dye terminators.<br />

D.A. Spicer, K.A. Hughes, R.J. Kaiser, A.L. Springer; Prolinx, Inc.,<br />

22322 20th Avenue South East, Bothell, WA 98021<br />

Prolinx ® Inc. has developed the RapXtract dye terminator removal system<br />

that is fully automatable for high throughput purification of cycle sequencing<br />

reactions. The kit is based on a novel magnetic particle format allowing<br />

for rapid purification of up to 384 samples at one time, depending on the<br />

robotic platform. The kit provides reproducible, high-quality results without<br />

the need for modified primers. The RapXtract kit is easily automated, as it<br />

does not require centrifugation, vacuum filtration, or multiple wash steps.<br />

The kit has been automated on several platforms including the TECAN Genesis,<br />

and the Tomtec Quadra 384 model 320.<br />

P115-S<br />

Systematic validation and optimization of capillary electrophoresis<br />

for high performance genotyping and fragment analysis.<br />

D. Shen, M. Minarik, A. Shuster, K. Pirkola, S. Gopalan, K. Dains,<br />

M. Mahtani; Molecular Dynamics, 928 E. Arques Ave., Sunnyvale, CA 94085<br />

Capillary electrophoresis systems have quickly replaced traditional fluorescent<br />

slab gels for DNA analysis in many laboratory environments. The technology<br />

transition has been abrupt and, on the up-side, has excitingly<br />

advanced the Human Genome Project. On the down-side, the transition for<br />

many labs has occurred without a clear understanding of the strengths and<br />

weaknesses of the new technology, methods for troubleshooting, and experience<br />

in setting benchmarks. We have spent the past year validating and<br />

optimizing the capillary electrophoresis platform, the MegaBACE for high<br />

throughput genotyping and fragment analysis. In the development of the system,<br />

we have devised a set of metrics and guidelines that predict the system<br />

performance.<br />

We present data and benchmarks on variables including optical response,<br />

dynamic range, sizing precision and reproducibility, resolution, throughput,<br />

capillary and sample success rates, and other critical parameters of system<br />

performance. These metrics offer a better understanding of proper system<br />

function, and indices for perturbations of that function. We also provide data<br />

on the performance of the software and the effects of modifications to the<br />

workflow on the genotype outputs. In addition, we present a process control<br />

system (including software and chemistry) that can be used to evaluate<br />

the system performance or to test new methods and protocols before integrating<br />

them in a production environment. In collaboration with others, we<br />

will present data on the system’s performance for fragment analysis applications<br />

including microsatellite genome scanning, AFLP analysis, SNP genotyping,<br />

and differential display.<br />

POSTER <strong>AB</strong>STRACTS<br />

216 JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000<br />

P114-T<br />

Applications of an electric field-assisted capillary LC technique<br />

and instrumentation for biological macromolecules analysis.<br />

F.J. Yang, Y-H. Jou, C. Wu, C. Wu, Y.W. Hong; Micro-Tech Scientific,<br />

140 South Wolfe Road, Sunnyvale, CA 94086<br />

Recent rapid growth in the needs for biological macromolecules analysis and<br />

drug discovery research has increased significantly the utilization of capillary<br />

column separation techniques such as capillary column LC, capillary column<br />

electrophoresis, and capillary column electro-chromatography etc. The<br />

growth in the applications of capillary column separation techniques is also<br />

driven by the commercialization of electro-spray and nano-spray LC-MS<br />

interfacing techniques that allow mass spectra-analysis of biological macromolecules.<br />

Major advantages of capillary column separation techniques are:<br />

1). Improvement of detection limit by more than 2000 times in comparison<br />

to that possible using conventional 4.6 mm id columns. The smaller the column<br />

diameter, the lower the detection limits. It allows femto-mole detection<br />

of trace amount of biological samples.<br />

2). Direct interface to MS has greatly increased the applications of MS for biological<br />

sampler analysis.<br />

3). It reduces solvent consumption by more than 2000 times.<br />

4). 2D LC-MS can be easily configured for rapid sample clean up, de-salt,<br />

matrix elimination, and sample concentrating. It allows fast capillary LC-MS<br />

for high through put sample analysis.<br />

5). Capillary columns are common to capillary LC, CE, and CEC, it facilitates<br />

an unified methodology and instrumentation for harness the advantages of<br />

capillary column separation utilizing both differential electrophoretic migration<br />

of charge molecules and the differential mobility of the sample molecular<br />

zones carried by either electro-osmosis flow or hydrodynamic flow.<br />

This presentation will discuss the concept of an unified capillary column separation<br />

methodology and the design of the instrumentation that can perform conventional<br />

CE and CEC. It can also be utilized for both low pressure and high<br />

pressure gradient (pH, buffer strength, and organic composition gradient) CE<br />

and CEC applications. Examples for the applications of the unified capillary separation<br />

system in the electrical- field assisted high pressure capillary gradient LC<br />

applications for biological macromolecules analysis will also be presented.<br />

P116-M<br />

Long sequence readlengths with the MegaBACE 1000<br />

DNA Analysis System.<br />

J. Ellerbrock1, M. Reagin1, A. Mamone2, J. Nelson1, B. McArdle1, C. Fuller1; 1AP Biotech, 800 Centennial Ave., Piscataway, NJ 08855,<br />

2Molecular Dynamics<br />

MegaBACE 1000 DNA Analysis System is a high-throughput, fluorescencebased<br />

DNA sequencer that utilizes capillary electrophoresis with 96 capillaries<br />

operating in parallel. The system automates gel matrix replacement, sample<br />

injection, DNA separation, and data analysis with integrating software.<br />

The sensitivity of DYEnamic ET energy transfer terminators and the superior<br />

resolving power of linear polyacrylamide (LPA) separation matrix allow<br />

detection over a broader range of template amounts. Likewise, the robust<br />

performance of Thermo Sequenase II DNA polymerase improves success<br />

rates.<br />

The MegaBACE 1000 has the ability to sequence 96 samples in just 2 hours,<br />

perform up to 9 runs per day, and can be used in both high-throughput and<br />

core DNA analysis facilities. In high-throughput facilities it can produce as<br />

much as 500,000 raw sequence bases a day. We have developed protocols<br />

that allow reads of over 1000 bases ideally suited for final overnight runs (4–6<br />

hours) when such sustained high-throughput is not required. We will present<br />

a variety of examples and protocols to obtain optimal results on the system.<br />

Data will be presented showing long DNA readlengths on a variety of different<br />

templates (BAC, plasmid, M13, high GC, or PCR product) using the<br />

DYEnamic ET terminator cycle sequencing kit and the MegaBACE 1000 system.


P117-T<br />

New separation medium for the <strong>AB</strong>I Prism ® 3700 DNA Analyzer<br />

and <strong>AB</strong>I Prism ® 3100 Genetic Analyzer.<br />

B.F. Johnson1, K.O. Voss2, J.N. Tian2, J.A. Fisher2; 1Applied Biosystems,<br />

850 Lincoln Centre Drive, Foster City, CA 94404, 2Applied Biosystems,<br />

Foster City, CA<br />

A new separation medium for the <strong>AB</strong>I PRISM ® 3700 DNA Analyzer and <strong>AB</strong>I<br />

PRISM ® 3100 Genetic Analyzer has been developed. The new polymer formulation<br />

has a viscosity similar POP-5 and POP-6 formulations previously<br />

released for DNA separations on our capillary instruments and consequently<br />

has similar capillary filling times and sheath flow characteristics. High quality<br />

separations can still be obtained with bare capillaries with a minimum of<br />

100 separations before cleaning or replacement of the capillary array. Compared<br />

to POP-5 separation medium using the default run conditions for<br />

POP-5 on the <strong>AB</strong>I PRISM ® 3700 DNA Analyzer, the new separation medium<br />

results in a 20% decrease in electrophoretic separation time while increasing<br />

one-base peak resolution by 100 bases. On the <strong>AB</strong>I PRISM ® 3700 DNA Analyzer<br />

with a 50 cm array the new polymer formulation under these conditions<br />

results in an average length of read of 850 bases at 10 runs per day. On the<br />

<strong>AB</strong>I PRISM ® 3100 Genetic Analyzer the new polymer formulation results in<br />

an average length of read of 825 bases in a 50 cm array at 16 runs per day.<br />

Basecalling length of reads obtained from <strong>AB</strong>I PRISM ® Sequencing Analysis<br />

(1% error) and from TraceTuner 1.1 analysis software will be compared at different<br />

separation voltages, distances, and temperatures. Run parameters that<br />

yield the maximum length of read or maximum number of runs per day will<br />

be presented.<br />

P119-M<br />

Converting from slab gel to capillary electrophoresis:<br />

a user’s guide for genotyping.<br />

C.L. Brown, B.F. Johnson, C. Wike, K. Roy, A. Wheaton, Y. Wang,<br />

N. Caffo; Applied Biosystems, 850 Lincoln Centre Drive, MS 404-1,<br />

Foster City, CA 94404<br />

Capillary electrophoresis has rapidly become the platform of choice for<br />

human disease research, clinical diagnostics and population genetics studies<br />

due to its ease of operation, automation and increased throughput. Historically,<br />

most protocols were developed on slab gel instruments due to their<br />

flexibility, high throughput and consistency, yet required considerable cost<br />

and labor. The <strong>AB</strong>I PRISM ® 3100 Genetic Analyzer, a 16-capillary electrophoresis<br />

instrument, was introduced to match the throughput and reproducibility<br />

of slab gels while offering lower run costs, less human intervention<br />

and greater sensitivity. Laboratories considering the conversion from slab gel<br />

systems should be mindful of a few distinctions between these two platforms.<br />

Herein, we will discuss modified sample preparation, fragment mobility differences<br />

and suggestions for multiplexing schemes. We will also highlight<br />

enhancements to data analysis, which reduce existing rate limiting steps.<br />

POSTER <strong>AB</strong>STRACTS<br />

<strong>AB</strong>RF 2001 <strong>AB</strong>STRACTS<br />

P118-S<br />

Modifications to 96-well DNA preparation kits for the<br />

MegaBACE 1000.<br />

R. Dhulipala1, B. McArdle1, A. Mamone2; 1AP Biotech,<br />

800 Centennial Ave., Piscataway, NJ 08855, 2Molecular Dynamics<br />

With the introduction of high-throughput DNA sequencing instruments,<br />

methods for simultaneous preparation of template samples have come into<br />

common usage. Several groups have developed plasmid preparation methods<br />

enabling the user to easily decrease the time to prepare 96 DNA samples<br />

and thus increase sequencing throughput. The majority of these kits are<br />

based upon traditional alkaline lysis of bacterial culture yet differ in their<br />

method of purification of DNA. We have evaluated several of these methods<br />

for preparing templates for the MegaBACE 1000 capillary instrument. We find<br />

that results obtained with capillary sequencing instruments are dependent<br />

upon the quality and amount of the starting template. Hence, results might<br />

vary greatly due to the method of DNA preparation.<br />

We have found that most of the preparation methods and kits are configured<br />

to optimize DNA yield, not well-to-well consistency. Fortunately, the DNA<br />

yields are greater than necessary and simple modifications to the standard<br />

protocols can be made that improve consistency without reducing yields significantly.<br />

Each purification method is evaluated for amount and consistency<br />

of DNA yield, and sequencing performance using DYEnamic ET terminators<br />

and the MegaBACE 1000. In addition, we make recommendations<br />

for each method to achieve optimal success rates with the system.<br />

P120-T<br />

Sequencing reaction purification of BAC DNA using a combination<br />

of molecular weight cut-off membrane and a new grade of<br />

Sephadex G-50.<br />

R. Dhulipala, A. Kumar; AP Biotech, 800 Centennial Ave., Piscataway,<br />

NJ 08855<br />

The purification of DNA sequencing reactions by ethanol precipitation or gel<br />

filtration prior to the loading of automated fluorescent DNA sequencing<br />

instruments is a standard technique for the removal of unincorporated dye<br />

terminators. One standard gel filtration method that we have developed into<br />

a ready-to-use 96-well format with pre-swollen DNA Grade Sephadex<br />

G-50 is AutoSeq96 (please see our companion poster). AutoSeq96 purification<br />

is six times faster than ethanol precipitation, is easy to use, and minimizes<br />

handling because samples are purified directly into 96-well collection<br />

plates. However, one aspect of DNA sequencing reaction purification that has<br />

remained cumbersome for some types of samples containing large DNA<br />

constructs is the additional removal of template DNA.<br />

We have developed a new type of gel filtration method for purifying DNA<br />

sequencing reactions using the basic format of the AutoSeq96 plate but with<br />

the added benefit of template removal. Elimination of template DNA prior to<br />

electrophoresis can both increase the overall success rate of the reactions and<br />

improve read lengths for large plasmids and BACs. This new gel filtration<br />

method, called PureSeq96, has been optimized in 96-well format using a<br />

high molecular weight cut-off membrane (MWCOM) combined with preswollen<br />

low conductivity Capillary Electrophoresis Grade Sephadex G-50.<br />

The combination of the unique properties of the MWCOM and this new<br />

grade of G-50 are expected to improve sequencing success rates of both large<br />

construct plasmids and BACs using MegaBACE 1000. Described in this<br />

poster are the results of our studies and some of the development issues considered<br />

during the design of this gel filtration method for DNA sequencing<br />

reaction purification.<br />

JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000 217


<strong>AB</strong>RF 2001 <strong>AB</strong>STRACTS<br />

P121-S<br />

DYEnamic ET terminator reagents and protocols for the<br />

<strong>AB</strong>I 3700 DNA sequencer.<br />

C. Palaniappan1, L. Hosta1, M. Zahn1, G. Kaput1, J. Hockenberry1, M. Campion1, A. Brito1, B. McArdle1, A. Mamone2; 1AP Biotech,<br />

800 Centennial Ave., Piscataway, NJ 08855, 2Molecular Dynamics<br />

DYEnamic ET terminator sequencing products have long been available for<br />

the MegaBACE 1000 sequencing instrument and the <strong>AB</strong>I 310, 373, 377 instruments.<br />

The method offers several significant advantages for routine DNA<br />

sequencing. DYEnamic ET terminators afford high sensitivity sequencing<br />

through energy transfer technology. Thermo Sequenase II DNA polymerase<br />

has unprecedented tolerance to salt and contaminants that may be present<br />

in template preparations. Rapid and efficient incorporation of dITP by this<br />

enzyme allows short cycle times and robust, compression-free sequencing of<br />

GC rich templates.<br />

We have developed sequencing reagents incorporating ThermoSequenase II<br />

DNA polymerase and ET terminators for the <strong>AB</strong>I 3700 DNA sequencer. An<br />

optimized mobility file for DYEnamic ET terminator dyes, spectral matrix<br />

standards and protocols have been optimized for this instrument. Data will<br />

be presented that allow direct comparison of this chemistry on the <strong>AB</strong>I 3700<br />

DNA sequencer with other available chemistries and instruments.<br />

MegaBACE, DYEnamic and Sequenase are registered trademarks of Amersham<br />

Pharmacia Biotech.<br />

P123-T<br />

Whole mass determination of two-dimensional gel separated lens<br />

crystallins by electrospray ionization mass spectrometry.<br />

Y. Ueda, L.L. David; Oregon Hlth. Sci. Univ., 611 SW Campus Dr.,<br />

Portland, OR 97201<br />

Crystallins, the major proteins of the eye lens, have long half-lifes, and the<br />

accumulation of post-translational modifications can cause insolubilization<br />

and cloudiness in the lens, a condition called cataract. The identity of modified<br />

crystallins on two-dimensional electrophoresis (2-DE) gels can be determined<br />

by peptide mass finger printing, but the incomplete recovery of peptide<br />

fragment sometimes limits the determination of protein modification. The<br />

purpose of this study was to develop a method to efficiently elute modified<br />

crystallins from 2-DE gels so that their masses could be measured by LC-<br />

ESI-MS.<br />

Proteins from rat lens were separated by 2-DE and visualized by negative<br />

zinc stain. Excised spots were incubated in 0.1% SDS and then finely crushed<br />

to 20 mm particles by forcing them through a metal frit. Proteins were then<br />

allowed to diffuse into a non-detergent containing solution during sonication.<br />

The masses of eluted proteins were determined using a 1 mm C4 reversephase<br />

column (Vydac) to remove bound SDS and on-line ESI-MS. Using this<br />

method, we were able to determine the cleavage sites of either N-terminally<br />

or C-terminally truncated crystallins, and also identify phosphorylated species<br />

from single spots of moderately abundant proteins. The method was much<br />

simpler and resulted in higher yields compared to electroelution, and should<br />

be useful for analyzing a wide range of 2-DE separated proteins. We are currently<br />

testing whether �LC columns can be used to increase the sensitivity<br />

of the method.<br />

POSTER <strong>AB</strong>STRACTS<br />

218 JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000<br />

P122-M<br />

Development of a fifth-dye labeled short size standard for<br />

SNP interrogation.<br />

S.S. Kuo, G. Ayanoglu, D. Wei, C-a. Chang, R. Brown, A.B. Tomaney,<br />

P. Dong; Applied Biosystems, 850 Lincoln Centre Drive, Foster City,<br />

CA 94404<br />

We describe the development of a short size standard for analyzing small<br />

fragments on fluorescent detection systems. This standard contains 9 fragments<br />

ranging from 15 to 120 nucleotides, and is labeled with a fifth dye.<br />

During rigorous testing with various media on gel or capillary electrophoresis<br />

platforms, we are able to achieve excellent precision and curve-fitting<br />

cross-platform. The fifth dye is spectrally well resolved from other dyes.<br />

This standard is designed in particular to enable automated data analysis in<br />

methods for single nucleotide polymorphism (SNP) detection such as singlebase<br />

extension (SNE) assays. The presence of the size standard will minimize<br />

the need for manual data analysis especially in multiplexed electrophoresis<br />

assays. In our poster, we will demonstrate its utility in SNE assays. With the<br />

combination of different fragment lengths and four-color chemistry, the<br />

potential for multiplexing SNP loci exists for large-scale genotyping with minimal<br />

optimization.<br />

P124-S<br />

Protein separation optimization in large-format<br />

NuPAGE bis-tris gels.<br />

R.A. Bogoev, J.W. Amshey; Invitrogen Corp., San Diego,<br />

1600 Faraday Avenue, Carlsbad, CA 92008<br />

Large format gels are frequently employed to obtain better resolution of proteins<br />

that have similar apparent molecular weights in SDS-PAGE. We have<br />

quantitatively determined the resolution of proteins using the distance of separation<br />

and the band width to establish a relationship between the run distance<br />

and the resolution for NuPAGE ® bis-tris gels. Resolution increased in<br />

a nonlinear fashion. Increasing the run distance three-fold led to less than a<br />

2-fold increase in resolution. Increased run length did not improve the resolution<br />

of larger proteins with the 10% acrylamide concentration used.<br />

Increasing crosslinker concentration 3-fold led to an improvement in resolution<br />

for medium and small proteins but led to a decrease in resolution of<br />

larger proteins. Further increase did not improve the resolution for medium<br />

and small proteins and further reduced resolution for larger proteins. Over<br />

the range of 10–50�C, resolution was optimal at a temperature of 20 degrees.<br />

Decreasing the buffer concentration in the NuPAGE bis-tris gels provided a<br />

faster separation with no effect on resolution. For most applications, due to<br />

the relatively small increase in resolution with increasing run length, shorter<br />

gels may be preferable because they are more convenient for post-run processing.


P125-M<br />

A high sensitivity silver stain without glutaraldehyde sensitization<br />

for protein gels used in mass spectrometry.<br />

S.E. Whitney, A. Bautista, J.W. Amshey; Invitrogen Corp., San Diego,<br />

1600 Faraday, San Diego, CA 92008<br />

Conventional sensitive silver stains use glutaraldehyde as a sensitizer to<br />

enhance silver deposition in the protein zone. This sensitizer reacts with basic<br />

amino acids (i.e. Lysine) which are target sites for trypsin cleavage, and can<br />

result in reduced yields of peptides when the protein is processed for identification<br />

by mass spectrometry. A silver stain kit (SilverQuest) has been<br />

developed which provides high sensitivity and good peptide recovery when<br />

stained gel bands are used for protein identification in mass spectrometry.<br />

Following staining and isolation of a protein band, the silver is removed by<br />

oxidation with potassium ferricyanide and complexation of Ag � with sodium<br />

thiosulfate. At the 5 picomole level of BSA oxidation of metallic silver resulted<br />

in an increase in the number of identified peptides from 11 to 32, compared<br />

to 17 with a conventional glutaraldehyde-sensitized stain. Percent coverage<br />

rose from 22 to 41% of the sequence. For L-glutamate dehydrogenase we<br />

observed 24 peptides and 48% sequence coverage while 31 peptides and<br />

34% coverage was found for Phosphorylase B. In addition, a rapid and sensitive<br />

procedure employing the use of a microwave has been developed<br />

which allows for sensitive staining in �30 min. The typical sensitivity of this<br />

new silver stain was found to be 0.2 ng for BSA, 0.4 ng for Ovalbumin, 0.3<br />

ng for Soybean trypsin inhibitor and 0.4 ng for a-Lactalbumin.<br />

P127-S<br />

Identification of endosperm proteins by peptide mass mapping:<br />

patterns of accumulation during wheat grain development.<br />

W.H. Vensel, W.J. Hurkman, C.K. Tanaka; USDA, 800 Buchanan Street,<br />

Albany, CA 94710<br />

As part of a project with the goal of decreasing the impact of environmental<br />

conditions on wheat productivity and quality, we are using proteomics to<br />

identify key events and metabolic pathways in wheat grain development.<br />

Wheat (Triticum aestivum L. cv. Butte 86) plants were grown in a climatecontrolled<br />

greenhouse that had an average daily maximum daytime temperature<br />

of 25�C and nighttime temperature of 17�C. Water and fertilizer were<br />

supplied by drip irrigation. Developing heads were tagged at anthesis. Grain<br />

was harvested at pre-selected time points and endosperm collected. Salt soluble<br />

proteins were extracted and separated by 2-D gel electrophoresis. Gel<br />

patterns for the different developmental stages were characterized using<br />

computer-based image analysis. As expected, a number of proteins increase,<br />

decrease, and were newly synthesized during grain development. Interestingly,<br />

basic proteins increased significantly throughout the developmental<br />

time course. Proteins were identified using Western blots, Edman degradation,<br />

and MALDI mass spectrometry. These proteins are involved in storage<br />

protein biosynthesis, protein folding, glycolysis, starch biosynthesis, and pest<br />

protection.<br />

POSTER <strong>AB</strong>STRACTS<br />

<strong>AB</strong>RF 2001 <strong>AB</strong>STRACTS<br />

P126-T<br />

Proteomics of brown spider venom.<br />

M.V. Sousa1, C.A.O. Ricart1, W. Fontes1, K.C. Barbaro2, L. Machado1, R.B. Cunha1; 1Univ. of Brasilia, Campus Universitario, Brasilia,<br />

DF 70910-900, Brazil, 2Butatan Inst.<br />

Brown spiders are found in several regions of the world. In Brazil, the<br />

brown spider Loxosceles gaucho is very common, and have caused severe<br />

accidents due to a potent dermonecrotic and lethal venom. Little is known<br />

about the composition of the venom proteome. Proteomic analysis was<br />

employed to identify some proteins in the venom in order to gain new<br />

insights on its mechanism of action.<br />

Venon was separated by two-dimensional electrocphoresis using IPG gels in<br />

the first dimension and SDS-PAGE for the second dimension. The resulting<br />

gels were scanned and analysed via the ImageMaster software. Sixty protein<br />

spots were detected. Most of the spots were found in the 25–35 kDa range.<br />

Proteins such as insectotoxins, dermonecrotic toxin and annexin IV were<br />

identified by either mass spectrometry or chemical sequencing. The dermonecrotic<br />

toxin was purified by gel filtration, anion exchange FPLC and RP-<br />

HPLC. At least four isoforms of the toxin exist in the venom. The construction<br />

of a venom gland cDNA bank is under way.<br />

P128-M<br />

Proteomics: facts, fictions and applications.<br />

S.J. Cordwell; Macquarie Univ., Australia, Level 4, Building F7B,<br />

Macquarie University, New South Wales 2109, Australia<br />

Proteomics as a science is dependent on finding molecules of significance<br />

from amongst highly complex biological mixtures. Technically, however,<br />

the protein ‘pool’ from which information can be gathered is dramatically<br />

lower than that predicted by genome sequencing. This is due to limiting factors<br />

defined by protein abundance, hydrophobicity, mass and pI, in association<br />

with two-dimensional gel electrophoresis. Furthermore, the variable<br />

nature of gene expression in vivo means that predicting the ‘functional’ proteome<br />

under a given set of conditions is dependent upon a thorough knowledge<br />

of biochemical pathways encoded by a genome and influenced by the<br />

surrounding environment. We have now begun to define the physical boundaries<br />

determined by these parameters to predict whether proteins will be visible<br />

using 2-D gel ‘arrays’ consisting of cellular pre-fractionation via relative<br />

protein solubility or cellular location, in combination with narrow-range<br />

immobilised pH gradients. The latter allows lower abundance proteins to be<br />

characterised utilising the sensitivity of mass spectrometry. Furthermore, we<br />

can now predict whether proteins are expressed via knowledge of physical<br />

characteristics within given detection parameters and upon the presence or<br />

absence of pathway components determined via nearest-neighbour predictions.<br />

Such predictions are based upon defining each translated gene within<br />

four theoretical sets. These interacting sets provide the real ‘pool’ from which<br />

2-D electrophoresis can be expected to provide utility, while proteins outside<br />

the ‘pool’ must be annotated via alternative means. Such predictions of biologically<br />

significant molecules that remain outside the scope of 2-D electrophoresis<br />

then allow the researcher to target single proteins or sets of proteins<br />

utilising complementary technologies including peptide labelling or<br />

monoclonal antibodies. Here we present the advantages and disadvantages<br />

of 2-DGE-MS proteomics in comparison and in complement to alternative<br />

technologies and describe the application of high-throughput technologies<br />

for biological experimentation in a variety of microorganisms.<br />

JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000 219


<strong>AB</strong>RF 2001 <strong>AB</strong>STRACTS<br />

P129-T<br />

Isolation and identification of rat liver proteins using ultracentrifugation<br />

with Nycodenz and 1D/2D-SDS-PAGE.<br />

K. Murayama1, T. Fujimura1, M. Morita2, N. Shindo1; 1Juntendo Univ. Sch.<br />

of Med., 2-1-1, Hongo, Bunkyo-ku, Tokyo, Tokyo 113-8421, Japan, 2Hitachi<br />

Koki Co., Ltd.<br />

The use of 2D-SDS-PAGE as a clinical molecular scanner of various tissues<br />

and physiological fluid samples has proved useful. However, each organ contains<br />

more than 4,000 proteins and accordingly, it is almost impossible to<br />

study the functional role of these proteins unless separated. Our ultimate goal<br />

is to use 2D-SDS-PAGE as a clinical molecular scanner to define each<br />

organelle in various organs.<br />

In this study, we report the isolation of rat liver organelles by density gradient<br />

centrifugation using Nycodenz. Nycodenz solution at 10, 20, or 30% concentration,<br />

containing 0.25 M sucrose (as an osmotic balancer), was added<br />

to each centrifuge tube and allowed to stand overnight at �20 to �80�. The<br />

solution was thawed at room temperature (�2 h), and analyzed to construct<br />

a density gradient curve. When used in a 5-ml tube, Nycodenz gradient densities<br />

from the top to the bottom without any centrifugation were 1.0334 to<br />

1.2188 at 10%, 1.0506 to 1.2878 at 20% and 1.0856 to 1.3199 at 30%. Liver<br />

homogenate (0.4 ml, 4 mg) was loaded on the Nycodenz gradient solution<br />

and centrifuged at 28,000 rpm for 20 min using a Hitachi ultracentrifuge<br />

CP100�-RPS40T-2. The mixture was fractionated by a fractionator, DGF-U, its<br />

absorbance measured at 360 nm with a spectrophotometer and density with<br />

an Abbe refract meter. Next, 5 �l of each fraction was applied onto 10% gel<br />

for 1D-SDS-PAGE, electrophoresis, and the gel was stained by silver nitrate.<br />

Another 1D-SDS-PAGE was erector-blotted to a PVDF membrane and the<br />

presence of organelles was confirmed using antibody of the marker protein<br />

for each organelle.<br />

P131-M<br />

Selective depletion of major serum proteins and fractionation prior<br />

to 2-dimensional differential gel electrophoresis.<br />

J.J. Cummings, E. Rohde, P.R. Griffin; Merck Res. Labs., RY800-B210, Rahway,<br />

NJ 07065<br />

Two dimensional differential gel electrophoresis (2DIGE) is a powerful technique<br />

for the study of protein expression in physiological fluids such as<br />

serum. However, the presence of a few major proteins interferes with the<br />

separation and detection of many low abundant, yet physiologically important<br />

proteins. Albumin (52%), IgG (20%), IgA (2.5%), IgM (1.6%), transferrin<br />

(3.6%) and �1-antitrypsin (1.6%) are the major constituents of serum in many<br />

species.<br />

Our objective was to selectively remove abundant proteins in a few sequential<br />

steps followed by the division of the depleted sera into multiple fraction<br />

based on their hydrophobicity (RP-HPLC) and/or charge (IEX-HPLC) prior to<br />

pre-electrophoresis fluorescent labeling.<br />

The effective removal of albumin was accomplished using Cibachron blue<br />

dye spin columns. Immunoglobulins (IgG, IgM) were removed by gel filtration<br />

over protein A and G columns. Affinity resins specific to transferrin and<br />

�1 antitrypsin were prepared in house and used for the depletion of the<br />

respective proteins. Chromatographic fractionation was carried out on largebore<br />

columns (4.6 � 100 mm). During all depletion and fractionation steps<br />

emphasis was placed on maximizing protein recovery. The protein concentration<br />

was monitored spectrophotometrically and effectiveness of depletion<br />

was assessed by SDS-PAGE.<br />

The separation of the depleted and fractionated sera by 2DIGE resulted in a<br />

significant increase in the dynamic range of the separation. Utilizing this<br />

approach proteins were detected in areas previously obscured by major<br />

serum constituents. Furthermore an increased number of proteins was<br />

observed. Combined with the power of differential protein mapping using<br />

fluorescent dyes the procedure has shown great utility in the search for differentially<br />

expressed proteins. We present examples of coupling 2DIGE with<br />

�LC-MS/MS and database searching for the identification of surrogate markers<br />

in sera.<br />

POSTER <strong>AB</strong>STRACTS<br />

220 JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000<br />

P130-S<br />

Rapid and simple single nanogram detection of glycoproteins<br />

in polyacrylamide gels and on electroblots.<br />

W.F. Patton, T.H. Steinberg, K.N. Berggren, K. Pretty On Top, C. Kemper,<br />

Z. Diwu, R.P. Haugland; Molecular Probes Inc., 4849 Pitchford Avenue,<br />

Eugene, OR 97402<br />

The fluorescent hydrazide, Pro-Q Emerald 300 dye, may be conjugated to<br />

glycoproteins by a periodic acid Schiff’s (PAS) mechanism. The glycols present<br />

in glycoproteins are initially oxidized to aldehydes using periodic acid.<br />

The dye then reacts with the aldehydes to generate a highly fluorescent conjugate.<br />

Reduction with sodium metabisulfite or sodium borohydride is not<br />

required to stabilize the conjugate. Though glycoprotein detection may be<br />

performed on transfer membranes, direct detection in gels avoids electroblotting<br />

and glycoproteins may be visualized 2–3 hours after electrophoresis.<br />

This is substantially more rapid than PAS labeling with digoxigenin<br />

hydrazide followed by detection with an anti-digoxigenin antibody conjugate<br />

of alkaline phosphatase, or PAS labeling with biotin hydrazide followed by<br />

detection with horseradish peroxidase or alkaline phosphatase conjugates of<br />

streptavidin, which require more than eight hours to complete. Pro-Q Emerald<br />

300 dye is spectrally compatible with SYPRO Ruby protein gel stain,<br />

allowing two-color detection of glycosylated and nonglycosylated proteins on<br />

the same gel or blot. Both fluorophores are excited with mid-range UV illumination.<br />

Pro-Q Emerald 300 dye maximally emits at 530 nm (green) while<br />

SYPRO Ruby dye maximally emits at 610 nm (red). As little as 300 pg of a1acid<br />

glycoprotein (40% carbohydrate) and 1 ng of avidin (10% carbohydrate)<br />

or glucose oxidase (12% carbohydrate) are detectable in gels after staining<br />

with Pro-Q Emerald 300 dye. Besides detecting glycoproteins, as little as 2–8<br />

ng of lipopolysaccharide is detectable in gels using Pro-Q Emerald 300 dye<br />

while 250–1000 ng is required for silver staining. Detection of glycoproteins<br />

may be achieved in 1-D or 2-D gels and on PVDF or nitrocellulose membranes.<br />

P132-T<br />

Rice tissue proteomics: towards a functional analysis of the<br />

rice genome.<br />

S. Komatsu, S. Shen, Z. Li, G. Yang, H. Konishi, M. Yoshikawa, R. Rakwal;<br />

Natl. Inst. of Agrobiol. Resources, 2-1-2 Kannondai, Tsukuba, Ibaraki<br />

305-8602, Japan<br />

The technique of proteome analysis with two-dimensional polyacrylamide<br />

gel electrophoresis (2D-PAGE) has the power to monitor global changes that<br />

occur in the protein expression of a tissue, an organism, and/or under<br />

stresses. In this study, proteins extracted from endosperm, embryo, root, callus,<br />

green shoot, etiolated shoot, leaf sheath and panicle of rice were separated<br />

by 2D-PAGE. The separated proteins were electroblotted onto a<br />

polyvinylidene difluoride membrane. The N-terminal amino-acid sequences<br />

of 117 out of 377 proteins were determined in this manner. N-terminal<br />

regions of the remaining proteins could not be sequenced and they were<br />

inferred to have a blocking group at N-terminus. Internal amino-acid<br />

sequences of 260 proteins were determined using the protein sequencer or<br />

matrix-assisted laser desorption/ionization time-of-flight mass spectrometry<br />

after enzyme digestion of proteins. Finally, a data-file of rice proteins was<br />

constructed, which included information on amino-acid sequence and<br />

sequence homology. Using this experimental approach, we could identify the<br />

major proteins involved in growth and development of rice. Some of these<br />

proteins, including a calcium-binding protein, which turned out to be carleticulin<br />

in rice, have functions in signal transduction pathway. The information<br />

thus obtained from amino-acid sequence of these proteins will be<br />

helpful in predicting the function of the proteins and for their molecular<br />

cloning in future experiments.<br />

This work was supported in part by a grant of Rice Genome Project PR-1201,<br />

MAFF, Japan.


P133-S<br />

Application of human cDNA microarray for the assessment of<br />

gene expression in mouse livers.<br />

G.S. Huang, M-Y. Hong, P-C. Yang; Pig Res. Inst., Taiwan, PO Box 23,<br />

Chunan, Miaoli, Taiwan, ROC 350, Taiwan<br />

A genomic survey for differentially expressed genes was performed to livers<br />

of ApoE deficient mice using human cDNA microarray containing approximately<br />

9,000 human cDNA clones. Due to the homology between mouse and<br />

human, hybridization was performed at lower stringency condition, 10 degrees<br />

below the regular hybridizing temperature. Gene expression profiles<br />

of livers corresponding to high levels of blood cholesterol were generated at<br />

genomic scale. Thirty-seven genes were randomly selected from a pool of<br />

differentially expressed genes and subjected to semi-quantitative RT-PCR, further<br />

confirmed the result from microarray hybridization. These included<br />

genes associated with atherosclerosis, and novel genes that implied novel<br />

pathways correlated to high levels of blood cholesterol. It is promising using<br />

human cDNA microarray, the most complete collection among all species, to<br />

study other mammalian systems with satisfying speed and accuracy.<br />

P135-T<br />

Meeting the demands for high throughput custom microarray<br />

production with the GeneMachines ® OmniGrid.<br />

K.S. Small, D. Henninger, G. Barragan, J. Dabrowiak, J. Fayet-Faber;<br />

GeneMachines, 935 Washington St., San Carlos, CA 94070<br />

As researchers in both independent labs and core facilities continue to<br />

approach the developing field of genomics with custom microarray production,<br />

the need for microarrayer instruments that maximize flexibility and<br />

throughput without compromising cost has become critical.<br />

The OmniGrid microarrayer offers a fully integrated and automated solution<br />

for creating custom DNA arrays. The OmniGrid is capable of printing 20,000<br />

spots/slide onto 100 slides using 48 pins in under 7 hours. Key components<br />

of the instrument’s flexibility include software features that allow random<br />

access of unique samples within a microtiter plate and inter-plate access<br />

among the 74 microtiter plates for production of fully customized microarrays.<br />

Data attesting to the instrument’s throughput, precision, and accuracy<br />

are discussed. Solutions to ensure microarray consistency during extended<br />

print runs through minimizing the effect of environmental variables are also<br />

explored.<br />

POSTER <strong>AB</strong>STRACTS<br />

<strong>AB</strong>RF 2001 <strong>AB</strong>STRACTS<br />

P134-M<br />

Planar waveguide microarrays for increased sensitivity in<br />

gene expression monitoring.<br />

W. Price1, A.P. Abel2, N.G. Scharer-Hernandez2, A.M. Zrolka1, A. Schafer1, H. Schwarz1; 1QIAGEN GmbH, 28159 Avenue Stanford, Valencia,<br />

CA 91355, 2Zeptosens AG<br />

The successful use of current microarray technology is impacted by its limited<br />

sensitivity, low signal to noise ratio and problems in reproducibility. In<br />

many bioanalytical applications—especially in toxicology, drug metabolism,<br />

and disease diagnostics—the amount of biological material that is available<br />

for an assay is limited. Reliable and robust methods for the quantitative<br />

detection of less than 1 �g of total RNA (equivalent to about 1 mg of tissue<br />

or 100,000 cells) are required in order to obtain valid experimental data. To<br />

fulfill these requirements currently available microarray detection approaches<br />

apply enzymatic target or signal amplification schemes, which bear among<br />

others the risk of biased amplification and non-linearity of the result. By combining<br />

Zeptosens’ planar wave guide (PWG) gene chip technology with<br />

QIAGEN’s expertise in sample preparation, extremely high levels of sensitivity<br />

and enhanced signal to noise ratios can be achieved. With this integrated<br />

approach, mRNA derived from the equivalent of 100 HeLa cells could<br />

be monitored without signal and/or target amplification.<br />

P136-S<br />

An investigation on reproducibility of GenChip expression<br />

methodology in UVa BMRF.<br />

B.P. Dragulev, P. Gallagher, Y. Bao, J.F. Fox; Univ. of Virginia,<br />

Jordan Hall, Box 441, Charlottesville, VA 22908<br />

A new service for microarray gene expression analysis based on the Affymetrix<br />

Gene Chip System is being developed in the Biomolecular Research<br />

Facility at University of Virginia. Concerns from investigators over the reproducibility<br />

of this high cost procedure have been raised that need to be<br />

addressed. In order to gain certain insight of the systematic performance of<br />

this system, we conducted a study focusing on reproducibility of the expression<br />

profiles obtained from repetition of one experiment under the same<br />

conditions. We used as a model VMM5 human melanoma cell line as the<br />

source of RNA. Five total RNA samples were extracted from five flasks of<br />

VMM5 cells grown under the same conditions. Labeled cRNAs were synthesized<br />

following Affymetrix procedure and hybridized to Affymetrix test chips.<br />

Statistical analyses were performed on the data based on differential intensity<br />

on both normal and log scales. The reliability of the data sets was assessed<br />

by SPSS Reliability Analysis software and other software. Another goal<br />

of this study was to compare the results produced from two different types<br />

of GeneChips, i.e., HuGene FL arrays and Human Genome U95A arrays in<br />

order to access the feasibility of cross-type comparison.<br />

JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000 221


<strong>AB</strong>RF 2001 <strong>AB</strong>STRACTS<br />

P137-M<br />

Dissecting structure-function relationships in RNA/protein<br />

interaction.<br />

P.S. Katsamba1, I.A. Laird-Offringa1, D.G. Myszka2; 1Univ. of Southern<br />

California, Norris Cancer Center/1441 Eastlake Ave., Los Angeles,<br />

CA 90089-9176, 2Univ. of Utah, 50 N. Medical Dr./School of Medicine<br />

Rm 4A417, Salt Lake City, UT 84132<br />

RNA-binding proteins play critical roles in gene expression and regulation at<br />

the post-transcriptional level. While much is known about the various naturally<br />

occurring RNA-binding motifs, and co-crystal structures of a number of<br />

RNA/protein complexes are available, very little is known about the dynamics<br />

of RNA/protein interactions. We have used the spliceosomal protein U1A<br />

and its RNA target in the U1 small nuclear RNA (U1hairpinII or U1hpII) as a<br />

model to study the kinetics of RNA/protein interaction. Using the previously<br />

solved structure of the U1A/U1hpII complex, we have engineered a series of<br />

mutants designed to probe the roles of electrostatics, hydrogen bonding, aromatic<br />

stacking, and RNA loop length, all of which have been implicated in<br />

formation of the U1A/U1hpII complex. The effects of these mutations on the<br />

binding dynamics were studied using BIACORE, which yielded high quality<br />

kinetic data about the interaction. We determined that neutralization of positive<br />

charges on the protein slows the association rate and reduces the deleterious<br />

effect of salt on complex formation. In contrast, removal of hydrogenbonding<br />

or stacking interactions within the RNA/protein interface, or<br />

reducing the size of the RNA loop, increases the dissociation rate. Our data<br />

support a mechanism of binding consisting of a rapid initial association<br />

based on electrostatic interactions and a subsequent locking step based on<br />

the hydrogen bonding and stacking interactions that occur during the<br />

induced fit of RNA and protein. Our results demonstrate the power of kinetics<br />

to dissect the functional differences between structural features of two interacting<br />

macromolecules.<br />

P139-S<br />

Automation for reliable RNA purification using<br />

silica-gel–membrane technology.<br />

F. Siegman, C. Schade, A. Wehren; QIAGEN Inc., 28159 Avenue Stanford,<br />

Valencia, CA 91355<br />

An automated system has been specially developed for laboratories requiring<br />

purification of high-quality RNA from animal and human cells for highthroughput<br />

gene-expression analysis, including microarray and real-time RT-<br />

PCR. This robotic workstation is designed to maximize reproducibility for all<br />

sample preparation steps (e.g., cell lysis, RNA purification, reaction setup),<br />

ensuring reliable results in subsequent analysis. CVs of less than 3% are consistently<br />

observed for quantitative TaqMan analysis when using this system<br />

for sample preparation. RNA purification using silica-gel–membrane technology<br />

allows recoveries of more than 90%, even with limiting starting material<br />

(e.g., as few as 10 cells). Purified RNA is free from enzyme-inhibiting<br />

impurities for excellent performance in the most sensitive quantitative applications.<br />

Optimized, ready-to-run protocols allow up to 192 samples to be<br />

purified in 90 minutes. The accurate liquid-handling system provides crosscontamination–free<br />

pipetting and small-volume liquid transfer, making it<br />

suitable for reaction setup and other liquid-handling tasks such as sample<br />

rearray. The easy-to-use operating system automatically tracks samples and<br />

records all process data generated in each run for complete documentation.<br />

Data is stored in standard formats and can be exchanged with other instruments,<br />

such as thermal cyclers or gridding robots, via network or disk.<br />

POSTER <strong>AB</strong>STRACTS<br />

222 JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000<br />

P138-T<br />

A rapid and efficient BAC DNA purification method for high<br />

throughput applications.<br />

C.M. Smith, A. Pryor; Princeton Separations, 920 HW 33, Building 7,<br />

Suite 6, Freehold, NJ 08724<br />

We have developed a method for the rapid and efficient purification of BAC<br />

DNA. Typical yields range from 0.5 to 1.0 �g from an overnight culture with<br />

Absorbance (OD600) � 5.0. BAC DNA was purified from an overnight culture<br />

of E. coli in 5 mL Terrific Broth (Cm 20 �g/mL). The culture was subject<br />

to gentle alkaline lysis, mixed with one volume of binding buffer, bound<br />

to a solid phase membrane, then eluted in a low salt buffer. Purified BAC<br />

DNA contains little or no chromosomal DNA or RNA visible after agarose gel<br />

electrophoresis and is suitable for downstream applications including restriction<br />

enzyme digestion and sequencing. A discussion of the protocol and<br />

accompanying data will be presented. The simplicity of the method makes<br />

it suitable for tailoring to a 96 well format providing a reliable method for<br />

high throughput applications<br />

P140-M<br />

Cotton biotechnology in China.<br />

B-H. Zhang; Cotton Res. Inst., Chinese Acad. of Agr. Sci., Baibi,<br />

Anyang, Henan 455112, People’s Republic of China<br />

During the past two decades, China has made great progress in cotton<br />

biotechnology and genetic engineering. Obtaining firstly regenerative plants<br />

from cotton anther and protoplast culture, and also obtaining regenerative<br />

plants from many domestic elite cotton varieties. After transgenic cotton carrying<br />

the insect-resistant (Bacillus thuringiensis: B.t.) gene was commercialized<br />

in 1996, at least ten Bt-cotton varieties were planted in China. In 1998,<br />

there were over 100,000 hectares of Bt-cotton were planted. Two kinds of<br />

bivalent insect-resistant transgenic cotton have been obtained. These new<br />

bivalent insect-resistant transgenic cotton carried two insecticidal genes, B.t.<br />

gene and CpTI gene, or pea lectin (P-Lec) gene and soybean Kunitz trypsin<br />

inhibitor (SKTI) gene respectively, and will be commercialized in 2000. Herbicide-resistant<br />

varieties for 2,4-D and Bromoxynil are under development<br />

and are expected to reach the market by 2001 or 2002. Disease-resistant<br />

transgenic cotton is under development and testing in lab and fields, and is<br />

expected to reach the market by 2000 or 2001. Fiber improvements, stress<br />

resistance, and male sterility and fertility for hybrid cotton are the next targets<br />

for cotton biotechnology. Several genes for fiber improvement and<br />

hybrid cotton are being tested in various laboratories. New genes for insect,<br />

herbicide and disease resistance are being sought.


P141-T<br />

Somatic embryony patterns and plant regeneration in cotton.<br />

B-H. Zhang; Cotton Res. Inst., Chinese Acad. of Agr. Sci., Baibi, Anyang,<br />

Henan 455112, People’s Republic of China<br />

Globular, heart, torpedo and cotyledon stages of development were observed<br />

in cotton somatic embryos, and only late torpedo and cotyledon stage somatic<br />

embryos could germinate into plantlets, the others could only produce<br />

advanentive embryos or adventive roots. Various developmental stage<br />

somatic embryos existed in cotton tissue cultue, and the number of the<br />

globular and heart-shaped stage was more than that of cotyledon embryos.<br />

The germination of cotton somatic embryos could be classified into three<br />

kinds and were affected by many factors such as hormons, basis medium et<br />

al. Resting phenomenon esists in cotton somatic embryos.<br />

P143-M<br />

Relationship between sexual maturation, serum leptin and<br />

sex hormone levels in obese and normal children.<br />

S.C. Hao, Y.C. Yuan; Harbin Med. Univ., China, 199 Dazhi Street,Harbin,<br />

Harbin, Heilongjiang 150001, People’s Republic of China<br />

In order to study the relationship between sexual maturation, serum leptin<br />

and sex hormone levels, the sexual maturation of 300 obese children and 300<br />

normal children not taking any medication or having evidence of endocrine<br />

or metabolic disease were questioned and their serum leptin and sex hormone<br />

levels were measured. Serum leptin levels were significantly increased<br />

(P � 0.01) in obese children compared with controls, while the sexual maturation<br />

of obese children was much earlier than that of controls (P � 0.01).<br />

Serum leptin levels were significantly increased (P � 0.01) in female compared<br />

with male, while the sexual maturation of female was much earlier<br />

than that of male (P � 0.01). There was significant negative correlation<br />

between testosterone and leptin in male (obese group: r � �0.83, P � 0.01;<br />

control group: r � 0.93, P � 0.01), while the estradiol was positive correlated<br />

with leptin in female (obese group: r � 0.84, P � 0.01; control group: r �<br />

0.95, P � 0.01). This study shows that leptin can promote sexual development<br />

in children and may be regulated by sex hormone. There are gender<br />

differences in the correlations between leptin and sex hormone, which may<br />

cause the differences of sexual maturation in male and female. This study<br />

also suggests that leptin play a more important role than sex hormones in the<br />

sexual development.<br />

POSTER <strong>AB</strong>STRACTS<br />

<strong>AB</strong>RF 2001 <strong>AB</strong>STRACTS<br />

P142-S<br />

The novel protein patterning method and its application<br />

for protein-protein interactions.<br />

B-G. Kim, C-S. Lee; Sch. of Chem. Engin., Seoul Natl. Univ., Seoul<br />

National University, Kwanak-Gu, Seoul, Seoul 151-742,<br />

Republic of Korea<br />

This research describes the new protein patterning method for the formation<br />

of highly defined two-dimensional protein arrays onto very hydrophobic thin<br />

film coated silicon substrate expressing protein functional activity at its patterned<br />

surface.<br />

In protein patterning process, Because very critical problem is non-specific<br />

protein binding onto undesirable region, We had tried a spin coated FluoroCarbon<br />

thin film, very hydrophobic surface property, which was expected<br />

to protect non-specific protein binding in our patterned silicon chip which<br />

was made from the lift-off process. From this simple method, we had obtained<br />

very high ordered protein patterns (180 �m, 300 �m, 500 �m diameter<br />

circle and 100 �m by 200 �m rectangular square) and also discarded<br />

nonspecific protein binding and blocking proteins which were generally<br />

used for protecting nonspecific binding such as bovine serum albumin(BSA)<br />

and skim milk proteins et al.<br />

Patterned micron-scale arrays were shown by fluorescence tagging proteins<br />

and had successfully obtained reliable quantitative data with confocal microscopy.<br />

And We also could create a versatile patterned protein surfaces for<br />

protein-protein interactions such as sandwich immuno-assay and indirect<br />

immuno-assay.<br />

From the these results, We have developed miniaturized assay systems showing<br />

the rapid, low sample volumes and high sensitivity.<br />

P144-T<br />

Experiences in implementing single nucleotide extension in a<br />

high throughput genotyping core facility.<br />

R. Scholl, L.W. Ballard; Univ. of Utah, 50 North Medical Dr. 4A430A,<br />

School of Medicine, Salt Lake City, UT 84132<br />

This poster will discuss some of our problems and successes in implementing<br />

single nucleotide extension (SNE) in a high throughput genotyping core<br />

facility as a method for DNA fragment analysis of SNP’s. We will discuss<br />

primer selection, PCR protocols, cleanup methods, quality control issues, and<br />

loading methods used. Product contamination and multiplexing of the samples<br />

will also be addressed.<br />

JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000 223


<strong>AB</strong>RF 2001 <strong>AB</strong>STRACTS<br />

POSTER AND RESEARCH GROUP <strong>AB</strong>STRACTS<br />

P145-S<br />

Effects of irradiation on the structure of proteins.<br />

K. Balamurugan, D-H. Chin; Natl. Changhua Univ. of Educ., Post Doctor,<br />

c/o Prof. Der-Hang Chin, Department of Chemistry, National Changhua<br />

University of Education, Changhua 50058, Taiwan<br />

In this study, we investigate the effects of irradiation on protein samples in<br />

optical spectrometers. The protein samples exposed to light in a spectrofluorometer<br />

are found to undergo in situ photoreactions. Continuous irradiation<br />

of proteins resulted in progressive decrease in the emission intensity. The<br />

light-induced changes are irreversible and appear to be oxygen-dependent.<br />

Continuous exposure to light renders the protein susceptible to cleavage. The<br />

secondary structural contents in proteins show appreciable decrease upon<br />

irradiation. In addition, the temperature-induced unfolding of proteins appear<br />

to be significantly influenced by the photoreactions caused due to irradiation.<br />

R1-M<br />

Fragment Analysis Research Group Study 2001: enhancing<br />

automated allele calling by tailing dinucleotide repeat markers<br />

to eliminate nontemplate driven nucleotide addition seen with<br />

Taq polymerase.<br />

D.A. Bintzler1, P.S. Adams2, L.W. Ballard3, Y. Bao4, D. Bartley5, L. Kasch6, L.O. Petukhova7, C. Rosato8, C.E. Terrell1; 1Univ. of Cincinnati,<br />

231 Bethesda Avenue, Cincinnati, OH 45267-0524, 2Trudeau Inst.,<br />

100 Algonquin Avenue, Saranac Lake, NY 12983, 3Univ. of Utah,<br />

4Univ. of Virginia, 5Johns Hopkins Univ., 6John Hopkins Univ.,<br />

7Rockefeller Univ., 8Oregon State Univ.<br />

Fragment analysis is a growing field in the genetic sciences. Some basics in<br />

methodology may provide useful tools to the many service facilities expanding<br />

to include fragment analysis. This tutorial will be an instruction based discussion<br />

covering basic methodology and challenges associated with fragment<br />

analysis using slab gel and capillary electrophoresis. Topics will include<br />

methods for producing clean PCR products through PCR optimization for single<br />

markers and multiplexing markers before and after PCR. Instrument<br />

related factors that may affect the quality of the data would be discussed for<br />

both slab gel and capillary electrophoresis platforms. Finally, a section covering<br />

analysis of the results will discuss improving accuracy through selecting<br />

correct size standard peaks and developing the best standard curve.<br />

Methods of calculation will be compared. Means to properly identify the correct<br />

fragment peak from the multiple peaks commonly seen during the fragment<br />

analysis run will be demonstrated. Participants in the tutorial will come<br />

away with methods designed to improve their capabilities to start, or continue<br />

Fragment Analysis in their laboratory.<br />

224 JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000<br />

P146-M<br />

Improving productivity in core facilities using the BioRobot 3000.<br />

F. Siegman, H. Kijenski, C. Schade, A. Wehren; QIAGEN Inc.,<br />

28159 Avenue Stanford, Valencia, CA 91355<br />

Core facilities providing molecular biology services often require automated<br />

systems to perform routine tasks. To meet the various demands faced by a<br />

core facility, these workstations should provide the flexibility to automate a<br />

wide variety of applications as well as the ability to exchange data with other<br />

laboratory instruments and data management systems. The BioRobot 3000 is<br />

a series of custom-designed workstations for automating routine tasks such<br />

as nucleic acid purification, protein purification and assay, and other liquidhandling<br />

tasks including reaction setup and sample rearray. Each workstation<br />

is tailor-made to meet individual application requirements. Different BioRobot<br />

3000 configurations designed for different applications will be described.<br />

A high-throughput sample purification system with 4 integrated vacuum<br />

manifolds provides rapid processing of 384 samples in a single run. Up to<br />

6 �g DNA (plasmids, cosmids, BACs, PACs, or P1 constructs) are purified per<br />

well in about 1 hour and 15 minutes using optimized protocols which minimize<br />

operator interaction and runtimes. Areas outside the BioRobot 3000<br />

worktable can be accessed using configurations with an extended arm. This<br />

arm allows the robotic handling system to move labware to external instruments,<br />

such as spectrophotometers and thermal cyclers. The operating system<br />

uses standard data formats to facilitate data exchange with these instruments<br />

as well as data management systems, allowing samples to be tracked<br />

as they move through various processing and analysis steps.<br />

R2-M<br />

PSRG 2001 Study: synthesis of difficult sequences.<br />

N.P. Ambulos, Jr. 1, L. Bonewald2, S. Kates3, G. Osapay4, H. Remmer5, A. Somogyi6, G. Tsaprailis6, S. Vigil-Cruz7; 1Univ. of Maryland Sch. of Med.,<br />

655 W. Baltimore St. BRB13-009, Baltimore, MD 21201, 2Univ. of Texas<br />

Hlth. Sci. Ctr., San Antonio, 3Consensus Pharmaceut., Inc., 4Univ. of<br />

California, Irvine, 5Univ. of Illinois, 6Univ. of Arizona, 7Univ. of<br />

Connecticut<br />

Particular peptide sequences are inherently a challenge to solid-phase synthetic<br />

methodologies as a result of concomitant side-reactions and/or conformational<br />

issues that may result in the failure to generate the desired product.<br />

Development of improved synthetic methodologies, reagents and amino<br />

acid derivatives are offering new solutions to the old problem of difficult<br />

sequences. This year’s <strong>AB</strong>RF Peptide Synthesis Research Group designed a<br />

study to evaluate how participating laboratories solve the challenge of correctly<br />

synthesizing these troublesome peptides. The laboratories were asked<br />

to prepare the following peptide that has several potential synthetic obstacles.<br />

H-Gln-Thr-Ser-Ser-Gly-Thr-Thr-Ser-Trp-Val-Thr-Ser-Arg-Arg-Asp-<br />

Gly-Ala-Gly-Ala-Gly-Pro-OH<br />

The peptide was synthesized and both crude and purified material was analyzed<br />

and characterized by the members of the Peptide Synthesis Research<br />

Group by HPLC, AAA, MALDI-MS and ESI-MS. Both crude and purified samples<br />

were requested from participating laboratories and were also characterized<br />

by HPLC, AAA, MALDI-MS and ESI-MS. Analysis of the samples also<br />

included a comparison of yield of correct product during purification. The<br />

results of these analyses will be presented.


R3-M<br />

A current profile of microarray laboratories: results of the<br />

2000–2001 <strong>AB</strong>RF Microarray Research Group Survey of Laboratories<br />

using microarray technologies.<br />

G.S. Grills1, C. Griffin2, A. Massimi1, K. Lilley3, K. Knudtson4, J. VanEe5; 1Albert Einstein Col. of Med., 1300 Morris Park Avenue, Bronx,<br />

NY 10461-1602, 2UCSF, 3Cambridge Univ., 4Univ. of Iowa,<br />

5Cornell Univ.<br />

The goal of this survey study was to build a current picture of the microarray<br />

analysis world. Microarray analysis is a fast developing field. New methods<br />

and instrumentation are being constantly introduced and the number of investigators<br />

using the technology is rapidly expanding. We focused on Affymetrix<br />

GeneChip and on cDNA, oligonucleotide and protein microarray<br />

spotting technologies. Data was requested from laboratories using these<br />

technologies by posting instructions for participation on microarray related<br />

electronic discussion groups. A web based survey form was used to collect<br />

information such as instrumentation, protocols, staffing, funding, and<br />

throughput. The survey was geared toward gathering information from academic,<br />

pharmaceutical, and commercial laboratories that offer microarray<br />

technologies as a shared resource. Individual laboratories that have these<br />

technologies were also invited to participate. This study is part of a continuing<br />

survey. The data from this survey was analyzed to build a current profile<br />

of microarray analysis laboratories.<br />

R5-S<br />

Preliminary results from the DNA Sequencing Research Group 2001<br />

Study: factors that affect the sequencing and detection of mixed<br />

base sequences in PCR products.<br />

M.A. Robertson1, L. Hall2, J. Hawes3, T. Hunter4, E. Jackson-Machelski5, K. Knudtson6, D. Leviten7; 1Univ. of Utah, 4A 438, School of Medicine,<br />

50 N. Medical Drive, Salt Lake City, UT 84132, 2Albert Einstein Col. of<br />

Med., 3Indiana Univ. Sch. of Med., 4Univ. of Vermont, 5Washington Univ.<br />

Sch. of Med., 6Univ. of Iowa, 7ICOS Corp.<br />

With the imminent release of the draft Human Genome Sequence, many laboratories<br />

are seeing increased demand for comparative sequencing. However,<br />

detection of mixed bases in a DNA population is a very demanding technique<br />

for many DNA Sequencing Core Laboratories. The DSRG will report on a<br />

study to test the ability of the <strong>AB</strong>RF DNA sequencing community to sequence<br />

PCR products containing several different kinds of base pair mutations, insertions<br />

and/or deletions. To limit variables, PCR products and sequencing<br />

primers were supplied along with a wild type reference sequence as a text<br />

file. Members were asked to sequence the PCR products using their choice<br />

of chemistry for mutation/mixed base detection. The set of PCR products<br />

contained mutations of varying difficulty. The PCR products were amplified<br />

from primers that contained universal forward and reverse primers to allow<br />

testing of both dye terminator and dye primer sequencing chemistries. Results<br />

were submitted as <strong>AB</strong>I chromatogram files with a short survey to gather relevant<br />

information. Analysis of the study will address:<br />

a) The ability of participants to be able to correctly identify the mutations.<br />

b) The accuracy of dye primer versus dye terminator sequencing for correct<br />

mutation detection.<br />

c) The ability to detect mutations from only one primer sequence.<br />

d) The differences in instrumentation in the abilty to detect mutations, e.g.,<br />

capillary vs slab gel instruments.<br />

e) The software/sequence alignment tools being used for mutation analysis.<br />

RESEARCH GROUP <strong>AB</strong>STRACTS<br />

<strong>AB</strong>RF 2001 <strong>AB</strong>STRACTS<br />

R4-T<br />

NARG 2000–2001 DNA Synthesis Study: Part I. Surveys of trends<br />

and strategies of DNA synthesis core facilities. Part II. Evaluation of<br />

oligonucleotide synthesis in core facilities utilizing homopolymers<br />

of A(20), G(20), C(20), T(20) and one heteropolymer.<br />

M.E. Gunthorpe1, J.F. Fox2, B.P. Holloway3, K.S. Lilley4, M. Lively5, K. Mills6, S.A. Scaringe7, T. Thannhauser8, A. Yeung9; 1Howard Hughes<br />

Med. Inst., UCSF, 533 Parnassus Ave. at 3rd Ave. Rm U436, San Francisco,<br />

CA 94143-0793, 2Univ. of Virginia, 3Ctrs. for Dis. Control and Prevent.,<br />

4Univ. of Cambridge, 5Wake Forest Univ. Sch. of Med., 6Millennium<br />

Pharmaceut. Inc., 7Dharmacon Res. Inc., 3200 Valmont Road #5,<br />

Boulder, CO 80301, 8Cornell Univ., 9Fox Chase Cancer Ctr.<br />

The Nucleic Acids Research Group (NARG) of the Association of Biomolecular<br />

Resource Facilities (<strong>AB</strong>RF) invited subscribers to the <strong>AB</strong>RF electronic discussion<br />

group to participate in our 2000–2001 study, which was accessed<br />

online at the web address listed below. Part I was a general survey regarding<br />

various aspects and issues pertaining to DNA synthesis core facilities,<br />

such as quality control, work philosophy and the future. Part II was an evaluation<br />

of oligonucleotide synthesis from core facilities by using a novel<br />

approach: Isolation of synthesis problems by analysis of homopolymer<br />

oligonucleotides. In this approach, the synthesis problems are magnified<br />

19�–20� depending on the chemical properties of the individual bases.<br />

Oligonulceotides submitted by participating labs will be analyzed by a range<br />

of analytical methods including HPLC, capillary electrophoresis, MALDI-TOF<br />

and ESI mass spectrometry to assess the quality of each product. The results<br />

of the evaluation of oligonucleotides from participants utilizing different<br />

instrumentation and protocols will be presented.<br />

Web address: http://www.abrf.org/ and choose “Nucleic Acids Research<br />

Group (NARG) 2000–2001 Study” under the category “Open Research Group<br />

Studies”.<br />

R6-S<br />

Dissecting structure-function relationships in RNA/protein<br />

interaction.<br />

I.A. Laird-Offringa1, D.G. Myszka2, P.S. Katsamba1; 1Univ. of Southern<br />

California, Norris Cancer Center, Room NOR 6420/1441 Eastlake Ave.,<br />

Los Angeles, CA 90089-9176, 2Univ. of Utah, 50 N. Medical Dr./School of<br />

Medicine Rm 4A417, Salt Lake City, Utah 84132<br />

RNA-binding proteins play critical roles in gene expression and regulation at<br />

the post-transcriptional level. While much is known about the various naturally<br />

occurring RNA-binding motifs, and co-crystal structures of a number of<br />

RNA/protein complexes are available, very little is known about the dynamics<br />

of RNA/protein interactions. We have used the spliceosomal protein U1A<br />

and its RNA target in the U1 small nuclear RNA (U1hairpinII or U1hpII) as a<br />

model to study the kinetics of RNA/protein interaction. Using the previously<br />

solved structure of the U1A/U1hpII complex, we have engineered a series of<br />

mutants designed to probe the roles of electrostatics, hydrogen bonding, aromatic<br />

stacking, and RNA loop length, all of which have been implicated in<br />

formation of the U1A/U1hpII complex. The effects of these mutations on the<br />

binding dynamics were studied using BIACORE, which yielded high quality<br />

kinetic data about the interaction. We determined that neutralization of positive<br />

charges on the protein slows the association rate and reduces the deleterious<br />

effect of salt on complex formation. In contrast, removal of hydrogenbonding<br />

or stacking interactions within the RNA/protein interface, or<br />

reducing the size of the RNA loop, increases the dissociation rate. Our data<br />

support a mechanism of binding consisting of a rapid initial association<br />

based on electrostatic interactions and a subsequent locking step based on<br />

the hydrogen bonding and stacking interactions that occur during the<br />

induced fit of RNA and protein. Our results demonstrate the power of kinetics<br />

to dissect the functional differences between structural features of two<br />

interacting macromolecules.<br />

JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000 225


<strong>AB</strong>RF 2001 <strong>AB</strong>STRACTS<br />

RESEARCH GROUP AND SPEAKER <strong>AB</strong>STRACTS<br />

R7-S<br />

On the development of standards for quantitative molecular<br />

interaction analysis: the interaction of barnase with its<br />

inhibitor barstar.<br />

E. Eisenstein; NIST, 9600 Gudelsky Drive, Rockville, MD 20850<br />

Virtually all biological processes rely on specific molecular interactions. Consequently,<br />

there is a growing need not only for useful tools for the analysis<br />

of molecular interactions, but also for standards to evaluate results with new<br />

experimental systems, and to assure comparability among different measurement<br />

techniques. Recently, the Molecular Interactions Research Group<br />

(MIRG) of <strong>AB</strong>RF focused on the interaction of barnase with barstar as a possible<br />

standard for reversible molecular interactions. Preliminary analyses<br />

were undertaken to clone, express, purify and characterize two barnase and<br />

barstar variants, and to measure the equilibrium constant for their association<br />

using three common experimental approaches: analytical ultracentrifugation,<br />

isothermal titration calorimetry and surface plasmon resonance using a<br />

BIAcore biosensor. Excellent agreement of the results obtained from the three<br />

techniques, as well as other experimental attributes of the proteins, suggests<br />

that the barnase-barstar system may be a useful standard for quantitative<br />

molecular interaction analysis.<br />

S2<br />

High throughput small molecule analysis using LC/MS and SFC/MS.<br />

M.J. Greig, W. Farrell, M. Ventura, J. Robinson, K.E. Milgram, K. Tivel,<br />

C. Aurigemma, X. Xiong, C. Pham, R. Lopez, A. Yanovsky; Pfizer—La Jolla,<br />

3550 General Atomics Ct., San Diego, CA 92121<br />

In our high-throughput discovery research, we use both commercial and custom<br />

mass spectrometry techniques to analyze tens of thousands of different<br />

compounds per month. Atmospheric pressure ionization sources have<br />

allowed the routine coupling of HPLC and recently supercritical fluid chromatography<br />

(SFC) to mass spectrometers for the analysis of a wide variety<br />

of small compounds. Electrospray ionization (ESI) is used not only for large<br />

biomolecules, but also for peptides, oligonucleotides, and a wide range of<br />

small polar molecules. Atmospheric pressure chemical ionization (APCI)<br />

extends the scope of mass spectrometry applications to include many small<br />

(�1000 daltons) polar and neutral molecules. The differences in these<br />

sources and their applicability to both LC/MS and SFC/MS will be described.<br />

The addition of quantitative parallel detectors such as nitrogen chemiluminescence<br />

detectors (NCD) and evaporative light scattering detectors (ELSD)<br />

will also be discussed. Finally, the benefits of SFC/MS in a high throughput<br />

environment will be discussed as well as strategies for high throughput data<br />

analysis.<br />

226 JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000<br />

S1<br />

Mass spectrometry of neurosteroids.<br />

R.L. Fitzgerald1, A. Alomary2, J.D. Rivera1, R.H. Purdy3, G.F. Koob4, M. Vallee4; 1VA Med. Ctr., UCSD, VAMC-113, 3350 La Jolla Village Dr.,<br />

San Diego, CA 92161, 2VA Med. Ctr., UCSD, Scripps Res. Inst.,<br />

3VA Med. Ctr., Scripps Res. Inst., 4Scripps Res. Inst.<br />

Electron capture negative chemical ionization (NCI) is one of the most sensitive<br />

ionization techniques available and is especially well suited for quantitative<br />

analysis of target compounds in biological extracts. For the analysis<br />

of small molecules, NCI is often several orders of magnitude more sensitive<br />

than traditional techniques such as electron ionization or positive chemical<br />

ionization. There are several important prerequisites for performing quantitative<br />

analysis using NCI, including an electronegative functional group, stable<br />

isotopic internal standards, and good method validation. We synthesized<br />

deuterium labeled analogs of neurosteroids and developed a NCI GC/MS<br />

method for quantification of neurosteroids in biological samples using isotope<br />

dilution.<br />

Neurosteroids have distinct neurotransmitter mediated effects and consequently<br />

it is important to be able to identify and quantify individual compounds.<br />

Previously, the determination of neurosteroids in biological matrices<br />

involved complicated purification protocols or did not use appropriate<br />

internal standards. We added deuterium-labeled internal standards to brain<br />

(100 mg of cortex homogenate) or plasma (300 uL). Samples were homogenized<br />

in methanol, centrifuged and diluted to contain 5% methanol and then<br />

applied to C-18 columns. After washing the column with methanol/water<br />

(50/50), steroids were eluted with methanol. Following evaporation, steroids<br />

were converted to pentafluorobenzyl oxime/trimethylsilyl ether derivatives.<br />

The extracts were analyzed using SIM. The present method allows simultaneous<br />

quantification of pg amounts (100 pg in 300 �L of plasma and 250 pg<br />

in 100 mg of brain tissue) of neurosteroids and will be helpful in elucidating<br />

the role of neurosteroids in health and disease.<br />

S3<br />

Algorithms and systems for high throughput structural biology.<br />

B.D. Randall1, T.A. Neubert2, H. Erdjument-Bromage3, S.A. Hefta4, R.S. Johnson5, J.T. Stults6; 1Dartmouth, 6211 Sudikoff Lab., Rm. 113,<br />

Dartmouth, Hanover, NH 03755-3510, 2New York Univ., 540 First Avenue,<br />

Lab 5-18, New York, NY 10016, 3Mem. Sloan-Kettering Cancer Ctr.,<br />

1275 York Avenue, New York, NY 10021, 4Bristol-Meyers Squibb,<br />

5Immunex Corp., 6Genentech, Inc.<br />

In the post-genomic era, key problems in molecular biology center on the<br />

determination and exploitation of three-dimensional protein structure and<br />

function. For example, modern drug design techniques use protein structure<br />

to understand how a drug can bind to an enzyme and inhibit its function.<br />

Large-scale structural and functional genomics will require high-throughput<br />

experimental techniques, coupled with sophisticated computer algorithms for<br />

data analysis and experiment planning. This talk will introduce techniques<br />

my lab is developing in two key areas: (1) data-directed computational protocols<br />

for high-throughput protein structure determination with nuclear magnetic<br />

resonance spectroscopy, and (2) experiment planning and data interpretation<br />

algorithms for reducing mass degeneracy in mass spectrometry for<br />

protein complex binding mode identification. These techniques promise to<br />

lead to fast, automated tools to aid the pursuit of deep structural and functional<br />

understanding of biopolymer interactions in systems of significant<br />

biochemical and pharmacological interest.


S4<br />

A crack in the egg: protein-protein interactions in failing hearts.<br />

A.R. Marks; Columbia Univ., 630 West 168th Street, New York, NY 10032<br />

Calcium (Ca 2�) ions are second messengers in numerous signaling pathways<br />

in all cell types. In the heart Ca 2� regulates muscle contraction, electrical signals<br />

that determine the cardiac rhythm, and probably plays a role in controlling<br />

cell growth. In the past decade elucidation of the molecular structure<br />

of the intracellular Ca 2� release channels on the sarcoplasmic and endoplasmic<br />

reticulum (SR/ER) has lead to an understanding of how these molecules<br />

regulate Ca 2� homeostasis in the heart. Consequently the role of<br />

these channels (ryanodine receptors, RyR, and inositol 1,4,5-trisphosphate<br />

receptors, IP3R) in cardiac pathophysiology is beginning to be understood.<br />

Intracellular Ca 2� release channels form a unique class of ion channels distinguished<br />

on the basis of structure size, and function. RyRs and IP3Rs have<br />

large cytoplasmic domains that are involved in the regulation of the channel<br />

pore located in the carboxy terminal 10% of the channel sequence. These<br />

enormous cytoplasmic domains serve as scaffolds for targeting proteins that<br />

bind kinases and phosphatases to the channels. Protein kinase A (PKA)<br />

phosphorylation of RyR2 dissociates the channel regulatory protein FKBP12.6<br />

and regulates the channel open probability (Po). We have defined a macromolecular<br />

complex comprised of RyR2, FKBP12.6, PKA, the protein phosphatases<br />

PP1 and PP2A, and an anchoring protein mAKAP. In failing human<br />

hearts RyR2 is PKA hyperphosphorylated resulting in defective channel function<br />

due to increased sensitivity to Ca 2�-induced activation resulting in<br />

impaired excitation-contraction (EC) coupling in failing hearts.<br />

S6<br />

VLiPs: a technique to study ligand binding to GPCRs in their<br />

natural environment.<br />

T.H. Jessen, N. Hunt; EVOTEC BioSystems AG, Hamburg, Schnackenburgallee<br />

114, Hamburg, Hamburg 22525, Germany<br />

Virus Like Particles (VLiPs) are generated by insect cells or mammalian cells<br />

through an engineering process of the retroviral export mechanism and the<br />

respective G-protein coupled receptor (GPCR). The retroviral gag protein<br />

interacts non-covalently but tightly and specifically with the C-terminus of the<br />

receptor of interest. The interaction leads to an enrichment of the receptor<br />

in its natural environment, the cell membrane, before budding of the VLiPs<br />

into the supernatant occurs.<br />

VLiPs carry up to 100 receptor molecules and turn out to be ideally suited<br />

for screening due to their robustness, long lived stability and physiological<br />

binding properties. Data obtained in binding and competition experiments<br />

are identical to data from literature. Currently, functional receptor screens are<br />

under investigation. The method allows to explore a number of other protein<br />

classes as well.<br />

SPEAKER <strong>AB</strong>STRACTS<br />

<strong>AB</strong>RF 2001 <strong>AB</strong>STRACTS<br />

S5<br />

Distinguishing agonist and antagonist binding to GPCRs using<br />

coupled plasmon-waveguide resonance analysis.<br />

G. Tollin, Z. Salamon, S. Cowell, V. Hruby; Univ. of Arizona,<br />

Biosciences West, Tucson, AZ 85721<br />

Structural changes accompanying the binding of ligands to the cloned human<br />

�-opioid receptor immobilized in a solid-supported lipid bilayer have been<br />

investigated using coupled plasmon-waveguide resonance spectroscopy.<br />

This highly sensitive new technique directly monitors mass density, conformation,<br />

and molecular orientation changes occurring within a single bilayer,<br />

and also allows direct determination of binding constants without structural<br />

modification of materials. Although both agonist binding and antagonist<br />

binding to the receptor cause increases in molecular ordering within the proteolipid<br />

membrane, only agonist binding induces an increase in thickness<br />

and molecular packing density of the membrane. This is a consequence of<br />

mass movements perpendicular to the plane of the bilayer occurring within<br />

the lipid and receptor components. These results are consistent with models<br />

of receptor function that involve changes in the orientation of transmembrane<br />

helices.<br />

S7<br />

A knockout for every gene and a chip for every purpose.<br />

M. Sussman; Univ. of Wisconsin, 425 Henry Mall, Madison, WI 53706<br />

We will report on progress in the development of two genomic technologiessaturation<br />

reverse genetics using a collection of insertionally mutagenized<br />

‘knockout’ Arabidopsis plants, and a maskless array synthesizer (MAS) for<br />

producing high density DNA oligonucleotide arrays ‘on the fly’. Our reverse<br />

genetic approach involves the rapid screening of several hundred thousand<br />

insertionally mutagenized Arabidopsis lines, for the isolation of knockout<br />

plants for any gene of interest. The bias of T-DNA for insertion at particular<br />

regions of the genome will be described and compared to published<br />

accounts of transposon bias. We have previously reported on a mathematical<br />

treatment required to isolate a knockout in every gene (Krysan et al.,<br />

1996), and we extend this analysis by taking into account our observations<br />

on the small but significant bias for the distributiion of T-DNA sites. Progress<br />

towards the establishment of a computer database for T-DNA insertions,<br />

using TAIL PCR to generate flanking sequences, will also be described. As a<br />

general tool for genotyping and for discerning the effects of a particular<br />

mutation on global genome expression, we have been testing existing and<br />

new technologies based on high density oligonucleotide arrays. Recent<br />

experiments utilizing a maskless array synthesizer (Singh-Gasson et al., 1999)<br />

which is capable of generating a half million different oligonucleotides on a<br />

2 cm squared glass surface, will be described. The MAS uses a digital<br />

micromirror device developed by Texas Instruments, to generate virtual<br />

masks for use in photolithography and is a powerful tool for bringing combinatorial<br />

chemistry to the benchtop in research laboratories. Krysan, P.J.,<br />

Young, J.C., Tax, F. and Sussman, M.R. 1996 Identification of T-DNA insertions<br />

within Arabidopsis genes involved in signal transduction and ion transport.<br />

Proc. Natl. Acad. Sci. 93:8145–8150. Singh-Gasson, S., Green, R.D,,<br />

Yue, Y., Nelson, C., Blattner, F., Sussman, M.R. and Cerrina, F. 1999. Maskless<br />

fabrication of light-directed oligonucleotide microarrays usig a digital<br />

micromirror array. Nature Biotechnology 17:974–978.<br />

JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000 227


<strong>AB</strong>RF 2001 <strong>AB</strong>STRACTS<br />

S8<br />

Assaying single cells to single organelles using mass spectrometry.<br />

J.V. Sweedler; Univ. of Illinois, Urbana, 600 S. Mathews Ave. 63-5,<br />

Urbana, IL 61801<br />

Understanding the interactions of relatively simple networks of neurons is<br />

hampered by a lack of knowledge of the full complement of neuropeptides<br />

involved in most neuronal systems. Using matrix-assisted laser desorption/ionization<br />

(MALDI) time-of-flight mass spectrometry, neuropeptides can<br />

be identified in single cells and even in individual neuronal processes. Mass<br />

spectrometric imaging methods are described that can provide spatial “maps”<br />

of the neuropeptides found in simple invertebrate networks, as well as identify<br />

new neuropeptides. Using these techniques, multiple novel neuropeptides<br />

have been discovered in the common neuronal model Aplysia californica.<br />

Approaches for direct de novo sequencing of peptides in single cells<br />

are demonstrated. A unique sampling protocol has been developed that<br />

allows the peptides in single attoliter to femtoliter volume vesicles to be measured<br />

using mass spectrometry. Using the atrial gland of Aplysia as a model,<br />

more than ten bioactive peptides are found in individual vesicles indicating<br />

the complexity of such hormonal signaling. Methods which combine capillary<br />

electrophoresis, fluorescence detection and mass spectrometry on the<br />

same sample are described.<br />

S10<br />

Chemical sensors for monitoring secretory and metabolic dynamics<br />

at single cells: application to pancreatic beta cells.<br />

R.T. Kennedy, W-j. Qian, M.G. Roper, L.S. Kauri, G.D. Dalgren;<br />

Univ. of Florida, PO Box 117200, Gainesville, FL 32611-7200<br />

Microscale electrochemical sensors that allow detection of insulin, glucose,<br />

and oxygen at single cells or small groups of cells have been developed. The<br />

insulin electrodes have been used to detect insulin secretion at the level of<br />

single exocytosis events while the glucose and oxygen electrodes have been<br />

used to monitor the dynamics of glucose and oxygen consumption. Such<br />

measurements, in combination with pharmacological probes and gene<br />

knock-outs, have been used to characterize secretory pathways and the<br />

interaction of metabolism with secretion. In one study, it was demonstrated<br />

that glyoclytic and respiratory oscillations occur in single islets of Langerhans<br />

and these oscillations require Ca 2� entry into the cell for proper feedback.<br />

Such oscillations provide a mechanism for oscillatory insulin secretion seen<br />

in vivo. In another study, using the single cell approach, we have shown that<br />

activation of insulin receptors leads to insulin secretion suggesting that positive<br />

feedback contributes to the mechanism for the first phase of insulin<br />

secretion. The signaling pathway by which insulin stimulates insulin secretion<br />

has been further studied revealing critical roles for IRS-1 and PI3-K in<br />

mediating insulin-stimulated insulin secretion. These studies have demonstrated<br />

that temporally resolved measurements at single cells or cell clusters<br />

are useful in evaluating mechanisms of signal transduction. The temporal<br />

measurements, especially of multiple key analytes, allow sequences of events<br />

to be evaluated and new mechansims to be uncovered.<br />

SPEAKER <strong>AB</strong>STRACTS<br />

228 JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000<br />

S9<br />

Profiling signal transduction networks in mammalian cells.<br />

N.L. Allbritton, C.E. Sims, G. Meredith; Univ. of California, Irvine,<br />

Medical <strong>Science</strong>s I, Rm D380, Irvine, CA 92697-4560<br />

A central goal of genomics and proteomics is to catalog the biological molecules<br />

present in different organisms and cell types under various conditions.<br />

A greater challenge for accurate and comprehensive characterization, however,<br />

lies in determining the activities and functional relationships of the biological<br />

molecules, particularly the enzymes, as they occur within the complex<br />

cellular networks that comprise biological systems. To accomplish this task,<br />

new technologies must be developed to measure multiple chemical species<br />

within intact intracellular networks. We have demonstrated a new method,<br />

the laser micropipet system, for the simultaneous measurement of the activation<br />

of key regulatory enzymes in small groups of cells, a single cell, or<br />

portions of a cell. This assay strategy should be broadly applicable to measurements<br />

of a broad range of enzymes, including kinases, phosphatases,<br />

proteases, and nucleases.<br />

S11<br />

Genetic analysis by mass spectrometry.<br />

L.M. Smith; Univ. of Wisconsin–Madison, 1101 University Ave.,<br />

Madison, WI 53706-1396<br />

In the last decade two powerful new tools for the mass spectrometric analysis<br />

of biomolecules have been developed, Matrix-Assisted Laser Desorption<br />

Mass Spectrometry (MALDI-MS), and Electrospray Ionization Mass Spectrometry<br />

(ESI-MS). The power of these methods lies in their ability to produce<br />

and mass analyze intact gas phase ions from very large molecules such as<br />

proteins and nucleic acids. The speed, accuracy, and sensitivity of the technologies<br />

make them well-suited to address a number of problems in genetic<br />

analysis, including the analysis of DNA sequence, genetic variations, and<br />

gene expression. Results in these areas will be presented, including recent<br />

work in which single nucleotide polymorphisms (SNPs) in genomic DNA<br />

may be analyzed without need for a prior PCR amplification step.


T1<br />

Oligonucleotide synthesis chemistry.<br />

R.T. Pon; Univ. of Calgary, 3350 Hospital Drive NW, Calgary,<br />

<strong>AB</strong> T2N 4N1, Canada<br />

Solid-phase oligonucleotide synthesis remains one of the most important<br />

technology platforms in the life sciences. It has been twenty years since the<br />

first automated DNA synthesizers were commercialized and phosphoramidite<br />

synthesis chemistry is well established. However, demand for increasing<br />

numbers and quantities of oligonucleotides continues to drive improvements<br />

to this technology. This presentation will provide an overview of DNA and<br />

RNA synthesis chemistry as it relates to a core facility operation. Alternative<br />

reagents for various synthetic steps will also be reviewed. This will include<br />

the Q-Linker and Linker Phosphoramidites reagents developed at the University<br />

of Calgary. These reagents have been used to improve synthesis productivity,<br />

recycle solid-phase supports, and perform tandem oligonucleotide<br />

synthesis. Such reagents may find future application in large scale or high<br />

throughput synthesis facilities.<br />

T3<br />

Practical aspects of DNA synthesizer operation.<br />

T.J. Demmitt; Biolytic Lab Performance, Inc., 39120 Argonaut Way, #229,<br />

Fremont, CA 94538<br />

Solid-phase oligonucleotide synthesis is typically performed via automated<br />

instrumentation. The quality of the product produced is related to many<br />

things one of which is the ability of the synthesis instrument to perform the<br />

chemistry consistently with quality. Maintenance, troubleshooting, repair<br />

and validation is key to keeping any instrumentation operating at peek efficiency.<br />

Maintaining these workhorse instruments has typically been entrusted<br />

to highly trained factory service engineers. The high level of expertise<br />

required to perform maintenance is defined at the instrument design stage,<br />

however it is entirely possible to perform many maintenance tasks with<br />

minimal electro-mechanical expertise. The focuses of this presentation is to<br />

show that a typical lab can perform much of its own service and thus minimize<br />

the need for high level maintenance engineering services.<br />

TUTORIAL <strong>AB</strong>STRACTS<br />

<strong>AB</strong>RF 2001 <strong>AB</strong>STRACTS<br />

T2<br />

Oligonucleotide synthesis quality control.<br />

A.T. Yeung, C.G. Miller, R.R. Muhlhauser; Fox Chase Cancer Ctr.,<br />

7701 Burholme Ave., Philadelphia, PA 19111<br />

Solid-phase oligonucleotide synthesis using automated DNA synthesizers<br />

and phosphoramidite synthesis chemistry produces high quality oligonucleotides<br />

most of the time. However, a variety of situations can compromise<br />

the quality of the DNA products. These situations include synthesizer malfunctions,<br />

reagent and column problems, incomplete reagent delivery and<br />

inadequate wash steps. The purpose of quality control is to prevent products<br />

of inferior quality from becoming variables in the users’ experiments. This<br />

presentation will discuss the wide range of oligonucleotide quality control<br />

methods in use in various laboratories. The semi-automated procedure in our<br />

facility, using Mono Q column anion-exchange on a FPLC unit, will be<br />

shown. A modification of the DNA synthesis procedure on the <strong>AB</strong>I model<br />

394 synthesizer to improve product quality will be discussed.<br />

T4<br />

Tutorial/practical aspects of DNA synthesizer operation:<br />

a look at high throughput DNA synthesizers.<br />

M.E. Gunthorpe; Howard Hughes Med. Inst., UCSF, 533 Parnassus Ave.<br />

at 3rd Ave., Rm U436, San Francisco, CA 94143-0793<br />

Tough competition from commercial companies require DNA synthesis core<br />

facilities to continuously provide excellent products and services. One possible<br />

solution to ideally meet the challenge is to purchase high-throughput<br />

instrumentation. In doing so, the facility would easily increase their capacity<br />

without additional personnel and would be able to reduce the base cost so<br />

that prices can be lowered to match those of the commercial market. The current<br />

choices for high-throughput DNA synthesizers provide a variety of features.<br />

A review of these instruments would help to delineate which choice<br />

would adequately match the facility’s unique needs. The features, advantages/disadvantages<br />

and viewpoints regarding these instruments will be discussed<br />

in an open format.<br />

JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000 229


<strong>AB</strong>RF 2001 <strong>AB</strong>STRACTS<br />

T5<br />

Technology resource grant opportunities.<br />

M.T. Marron1, M.A. Tingle1, N. Pearson2, M.J. Saunders3; 1NCRR,<br />

NIH, 6705 Rockledge Drive, Bethesda, MD 20892-7965, 2CSR, NIH,<br />

3Natl. Sci. Fndn.<br />

Federal funding is critical for equipping university-based Biomolecular<br />

Resource Facilities with state-of-the-art technologies. In this session, representatives<br />

from two Federal agencies, NSF and NIH, with day-to-day responsibility<br />

for administration of instrumentation grants, will discuss the review<br />

and funding of their various competitive grant programs.<br />

The tutorial session will be split into two parts. The first half of the session<br />

will provide an overview of the various instrumentation programs at NSF and<br />

NIH of interest to <strong>AB</strong>RF members. The second half of the session will focus<br />

on the preview processes at both agencies. Discussion will focus on practical<br />

tips on how to prepare a successful NSF and NIH proposal. Formal presentations<br />

will be followed by a question and answer period.<br />

T7<br />

Advances in DNA sequencing.<br />

L.S. Hall1, E. Thomas1, K. Lilley2, G. Grills1; 1Albert Einstein Col. of Med.,<br />

1695 Poplar Street, NewYork, NY 10464, 2Cambridge Univ.<br />

This tutorial will review past advances in DNA sequencing and present new<br />

techniques for sequencing difficult templates. Significant advances have<br />

taken place in DNA sequencing in the past four years by virtue of improved<br />

instrumentation, advances in the sequencing chemistries and alternative reaction<br />

formats for sequencing difficult clones. In particular, the replacement of<br />

Rhodamine terminators with Big Dye terminators increased the range of signal<br />

strengths compatible with good data and eliminated many of the peak<br />

height errors associated with rhodamine terminators; such as base calling<br />

ambiguities linked to the pattern of a small ‘G’ following an ‘A’.<br />

The recent introduction of dGTP terminators has improved the quality of data<br />

from templates with high GC content or regions of secondary structure.<br />

Data will be presented demonstrating the effectiveness of dGTP compared<br />

to Big Dye. In addition, the efficacy of betaine (NNN Trimethylglycine) versus<br />

DMSO will be discussed. New data will be presented demonstrating that<br />

a combination of Big dye and dGTP can sequence through regions of high<br />

G-C content without incurring compressions, a disadvantage of dGTP terminators<br />

by themselves.<br />

Finally, studies are in progress to evaluate the ability of different techniques<br />

to sequence through various types of difficult motifs by addition of commercial<br />

additives, such as the new GIBCO Rx Enhancer buffers. In general<br />

difficult motifs, such as repeats, induce stalling of Taq and loss of signal<br />

beyond the offending region. Depending upon the type of template, these<br />

buffers can restore the processivity of the enzyme and resolve regions of secondary<br />

structure. Results will be presented from studies that identify combinations<br />

of additives that work well with particular motifs.<br />

TUTORIAL <strong>AB</strong>STRACTS<br />

230 JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000<br />

T6<br />

Advances in DNA sequencing.<br />

M.A. Robertson; Univ. of Utah, 4A 438 School of Medicine,<br />

50 N. Medical Drive, Salt Lake City, UT 84132<br />

The Human Genome Project has been a major driving force for the development<br />

of new instrumentation and methodology for high throughput<br />

sequencing. During the last five years we have seen major advances both in<br />

instrumentation and sequencing chemistries. This tutorial will present user<br />

insight into one of the newer capillary instruments that promises to fit well<br />

into a core laboratory setting. The tutorial will also look at the changes in sequencing<br />

chemistry and present some solutions for difficult templates using<br />

these new chemistries. This tutorial is designed to be interactive. There will<br />

be a 30 minute question/answer session after the two main speakers, with a<br />

panel comprising the speakers and members of the DNA Sequencing Research<br />

Group. Please come armed with your most difficult sequencing problems<br />

and help spark a lively discussion session.<br />

T8<br />

Incorporating a 16 capillary electrophoresis DNA sequencer<br />

into a core facility.<br />

P.T. Morrison; Dana-Farber Cancer Inst., 44 Binney St., JFB216,<br />

Boston, MA 02115<br />

In the past few years the Human Genome Project (HGP) has driven innovation<br />

in DNA sequencing chemistry and instrumentation. For the most part this<br />

has been beneficial for all types of DNA sequencing projects both large and<br />

small. Recently manufacturers are now filling a void for instrumentation that<br />

is better suited for facilities that sequence less than 100,000 templates per<br />

year.<br />

The incorporation of an Applied Biosystems Model 3100 Genetic Analyzer<br />

into a core facility environment will be discussed.


T9<br />

More than one way to capture a phosphopeptide.<br />

H. Erdjument-Bromage; Mem. Sloan-Kettering Cancer Ctr.,<br />

1275 York Avenue, New York, NY 10021<br />

Despite the advances made during the past few years, mapping the phosphorylation<br />

sites on a protein often proves to be quite challenging. In this<br />

tutorial, several issues concerning sample preparation will be discussed,<br />

namely the choice of proteases and chromatographic media (both reversedphase<br />

and metal affinity). To illustrate the importance of sample preparation,<br />

“failed” mapping experiments will be used to highlight possible steps where<br />

the capturing may go wrong. The optimal approach to affinity capture is a<br />

combination of (1) one or more different proteases with careful planning of<br />

the “pre-affinity” buffer exchange steps, and (2) specific, micro-preparative<br />

capture of the phosphorylated fragments on immobilized metal ions (e.g.<br />

Ga 3�), followed by a desalting/concentration step. Critical examination of all<br />

sequential steps will be considered in turn.<br />

T11<br />

Affinity capture of DNA-binding proteins.<br />

M. Yaneva; Mem. Sloan-Kettering Cancer Ctr., 1275 York Ave. Box 137,<br />

New York, NY 10021<br />

Regulatory nuclear proteins and their complexes bind to cis-regulatory elements<br />

depending on the gene activities. These proteins can be isolated from<br />

cell nuclei and captured in vitro using specific DNA oligonucleotides (or their<br />

multimers) immobilized on solid supports (e.g. beads). The successful capture<br />

of these proteins depends on: 1) abundance of the protein in the cell;<br />

2) efficiency of protein extraction; 3) stability of the protein in extract; 4)<br />

maintenance of post-translational modification during extraction; 5) conditions<br />

used for in vitro DNA binding; 6) affinity of the protein for the isolated<br />

control element. The conditions for sample preparation and characterization<br />

as well as the optimization of the affinity capture on DNA beads will be discussed.<br />

Finally, the captured proteins are displayed on SDS PAGE and stained<br />

proteins bands are subjected to identification by MS.<br />

TUTORIAL <strong>AB</strong>STRACTS<br />

<strong>AB</strong>RF 2001 <strong>AB</strong>STRACTS<br />

T10<br />

Immunocapture of proteins—hints and tricks.<br />

R.M. Goetz; Mem. Sloan-Kettering Cancer Ctr., 1275 York Avenue,<br />

Box 137, New York, NY 10021<br />

Immunoprecipitation is a classical technique for capturing proteins from cell<br />

and tissue samples. Its sensitivity and specificity may be greater than those<br />

offered by any other technique, but critically depend on the experimental<br />

conditions applied. In this tutorial, various aspects of protein immunocapture<br />

will be discussed including the choice of sample buffer (detergents, chelators,<br />

enzyme inhibitors, reducing agents), the choice of antibody (monoclonal<br />

versus polyclonal; bivalent versus monovalent) and the choice of capture of<br />

formed immune complexes.<br />

T12<br />

Preparing and delivering an effective presentation.<br />

L.A. Steinke1, A. Smith2; 1Univ. of Nebraska Med. Ctr., Omaha,<br />

NE 68198-4525, 2Stanford Univ. Med Ctr.<br />

As scientists, we are often expected to present our data orally, accompanied<br />

by slides or a computer presentation. Many of us receive little training in public<br />

speaking, and see a microphone only at national meetings. Too often,<br />

important results are obscured by a poorly planned or poorly executed presentation.<br />

Sometimes, as speakers, we do not even know how to appropriately<br />

use the audio visual equipment that is available to us. Al Smith, Stanford<br />

University will present “How to get the message across—do’s, dont’s,<br />

and maybe’s”. The audiovisual department of the Town and Country will<br />

then present a short overview of the equipment found at most meeting<br />

places, and demonstrate its proper use. A professional speech coach will then<br />

cover “Organizing and Preparing a Scientific Presentation. Topics to be covered<br />

in this session will include overview of a presentation, organization and<br />

analysis of data, and the importance of a conclusion and a take home message.<br />

JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000 231


<strong>AB</strong>RF 2001 <strong>AB</strong>STRACTS<br />

T13<br />

Fragment Analysis Research Group Tutorial 2001: a practical<br />

discussion of fragment analysis: sample preparation,<br />

instrumentation and analysis of the results.<br />

D.A. Bintzler1, L.O. Petukhova2, C. Rosato3; 1Univ. of Cincinnati,<br />

271 Bethesda Avenue, Cincinnati, OH 45267-0524, 2Rockefeller Univ.,<br />

3Oregon State Univ.<br />

Fragment analysis is a growing field in the genetic sciences. Some basics in<br />

methodology may provide useful tools to the many service facilities expanding<br />

to include fragment analysis. This tutorial will be an instruction based discussion<br />

covering basic methodology and challenges associated with fragment<br />

analysis using slab gel and capillary electrophoresis. Topics will include<br />

methods for producing clean PCR products through PCR optimization for single<br />

markers and multiplexing markers before and after PCR. Instrument<br />

related factors that may affect the quality of the data would be discussed for<br />

both slab gel and capillary electrophoresis platforms. Finally, a section covering<br />

analysis of the results will discuss improving accuracy through selecting<br />

correct size standard peaks and developing the best standard curve.<br />

Methods of calculation will be compared. Means to properly identify the correct<br />

fragment peak from the multiple peaks commonly seen during the fragment<br />

analysis run will be demonstrated. Participants in the tutorial will come<br />

away with methods designed to improve their capabilities to start, or continue<br />

Fragment Analysis in their laboratory.<br />

TUTORIAL <strong>AB</strong>STRACTS<br />

232 JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000


Key to Abstract<br />

Numbering<br />

Prefixes:<br />

P = Poster<br />

R = Research Group<br />

S = Speaker<br />

T = Tutorial<br />

A<br />

Abel, A.P., P134-M<br />

Adams, P.S., P19-S, R1<br />

Aebersold, R., P45-T, P60-T, P66-T<br />

Allbritton, N.L., S9<br />

Alomary, A., S1<br />

Alterman, M.A., P80-M<br />

Alward, L.J., P67-S<br />

Ambulos, Jr., N.P., R2<br />

Amparo, G., P3-T<br />

Amshey, J.W., P124-S, P125-M<br />

Anderson, G.A., P45-T, P47-M<br />

Andon, N., P54-T<br />

Andresson, T., P31-S<br />

Annan, R.S., P94-S<br />

Arnott, D., P42-T<br />

Asano, Y., P25-S<br />

Aurigemma, C., S2<br />

Ayanoglu, G., P122-M<br />

B<br />

Baird, G.S., P95-M<br />

Balamurugan, K., P145-S<br />

Baldridge, K.K., P95-M<br />

Ballard, L.W., P144-T, R1<br />

Bao, Y., P136-S, R1<br />

Barbaro, K.C., P126-T<br />

Barragan, G., P135-T<br />

Bartley, D., R1<br />

Bateman, R., P64-S<br />

Bateman, R.H., P91-S<br />

Batz, S.A., P85-S<br />

Bautista, A., P125-M<br />

Bawge, V., P27-T<br />

Baybayan, P.A., P8-M<br />

Belcinski, R., P112-S<br />

Berger, S.J., P47-M<br />

Berggren, K.N., P130-S<br />

Bimmler, J., P61-S<br />

Bintzler, D.A., P9-T, R1, T13<br />

Blackburn, K., P48-T, P70-S<br />

Bloch, C., P77-M<br />

Bogoev, R.A., P124-S<br />

Bonewald, L., R2<br />

Booth, L.R., P1-S<br />

Boucher, R., P48-T<br />

Brancia, F., P63-T<br />

Breu, H., P27-T<br />

Brito, A., P121-S<br />

Brito, J., P79-S<br />

Brown, C.L., P119-M<br />

Brown, J., P62-M<br />

Brown, R., P122-M<br />

AUTHOR INDEX<br />

Brune, D.C., P49-S<br />

Burkhart, W., P48-T, P70-S<br />

C<br />

Caffo, N., P119-M<br />

Campbell, D., P26-M<br />

Campbell, J., P89-M<br />

Campion, M., P121-S<br />

Carberry, R., P82-S<br />

Carlson, J.E., P67-S<br />

Carr, S.A., P94-S<br />

Carruthers, R.A., P37-S, P91-S<br />

Cavey, G.S., P67-S<br />

Chan, J., P71-M, P73-S<br />

Chang, C-A., P122-M<br />

Chappell, G., P27-T<br />

Chen, S-M., P3-T<br />

Chin, D., P80-M<br />

Chin, D-H., P145-S<br />

Chulin, A., P99-T<br />

Connelly, R., P26-M<br />

Conrads, T.P., P45-T<br />

Cooper, C.A., P86-M<br />

Cordwell, S.J., P128-M<br />

Corthals, G.L., P41-M<br />

Couch, L., P23-M<br />

Cowell, S., S5<br />

Cummings, J.J., P131-M<br />

Cunha, R.B., P126-T<br />

D<br />

Dabrowiak, J., P135-T<br />

Dains, K., P112-S, P115-S<br />

Daley, T-J., P79-S<br />

Dalgren, G.D., S10<br />

Daniels, S., P57-T<br />

Dansithong, W., P25-S<br />

Das, R., P12-T<br />

David, L.L., P123-T<br />

Davis, R., P70-S<br />

Deciu, C., P59-M<br />

DeGnore, J.P., P89-M<br />

Deldot, T., P112-S<br />

Demmitt, T.J., T3<br />

Dhulipala, R., P118-S, P120-T<br />

Dhume, S.T., P85-S<br />

Diwu, Z., P130-S<br />

Dobrzanski, M.J., P19-S<br />

Dolios, G., P79-S<br />

Dong, P., P122-M<br />

Doyle, M.L., P109-S<br />

Dragulev, B.P., P136-S<br />

Ducret, A., P90-T<br />

Duewel, H., P103-S<br />

Dupont, D.R., P84-T, P97-S<br />

E<br />

Eisenstein, E., R7<br />

Ellerbrock, J., P116-M<br />

Eng, J., P60-T<br />

Ens, W., P66-T<br />

Erdjument-Bromage, H., S3, T9<br />

Esteban, C.D., P102-T<br />

F<br />

Faca, V.M., P88-S<br />

Farrell, W., S2<br />

Fayet-Faber, J., P135-T<br />

Fenyo, D., P104-M<br />

Ferguson, M.D., P21-T<br />

Fernandez, J., P79-S<br />

Figeys, D., P103-S<br />

Fisher, J.A., P117-T<br />

Fitzgerald, R.L., P46-S, S1<br />

Fontes, W., P126-T<br />

Fox, J.F., P136-S, R4<br />

Franke, C., P52-S<br />

Fröhlich, T., P52-S, P61-S<br />

Fuhrer, K., P108-T<br />

Fujimura, T., P129-T<br />

Fuller, C., P116-M<br />

Furtos-Matei, A., P93-T<br />

Furuishi, K., P87-T<br />

G<br />

Gall, A., P21-T<br />

Gallagher, P., P136-S<br />

Gaskell, S.J., P63-T<br />

Gillig, K.J., P108-T<br />

Goetz, R.M., T10<br />

Gonin, M., P108-T<br />

Gopalan, S., P115-S<br />

Gostick, D., P62-M<br />

Graham, K.S., P84-T<br />

Greene, L.J., P88-S<br />

Greig, M.J., S2<br />

Griffin, C., R3<br />

Griffin, P.R., P131-M<br />

Griffin, T.J., P66-T<br />

Grills, G., R3, T7<br />

Gross, L.A., P95-M<br />

Guettler, R., P18-T<br />

Gunning, K.M., P2-M<br />

Gunthorpe, M.E., R4, T4<br />

Gygi, S.P., P66-T<br />

<strong>AB</strong>RF 2001 <strong>AB</strong>STRACTS<br />

H<br />

Haff, L.A., P36-T, P40-S<br />

Hager, K.M., P11-M<br />

Hahner, S., P55-S<br />

Haley, J.D., P104-M<br />

Hall, L.R., P36-T, P40-S<br />

Hall, L.S., P6-T, R5, T7<br />

Hall, S., P18-T<br />

Hall, S.C., P43-S<br />

Halpern, B., P51-T<br />

Hao, S.C., P143-M<br />

Harris, R., P80-M<br />

Harrison, M.J., P86-M<br />

Haugland, R.P., P130-S<br />

Hawes, J., P6-T, R5<br />

Hawke, D.H., P77-M, P50-M<br />

Hayashi, M., P87-T<br />

Haynes, P.A., P54-T<br />

Hefta, S.A., S3<br />

Hemesath, T., P31-S<br />

Henninger, D., P135-T<br />

JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000 233


<strong>AB</strong>RF 2001 <strong>AB</strong>STRACTS<br />

Henschen-Edman, A.H., P92-M<br />

Henzel, W.J., P75-T, P78-T<br />

Hillen, W., P102-T<br />

Hobbs, J.B., P14-M<br />

Hockenberry, J., P121-S<br />

Hoffer, A., P20-M<br />

Hoffman, R.C., P95-M<br />

Hogan, R.J., P19-S<br />

Holle, A., P65-M<br />

Holloway, B.P., R4<br />

Hong, M-Y., P133-S<br />

Hong, Y.W., P114-T, P81-T<br />

Horn, M.J., P33-T, P68-M<br />

Hosta, L., P121-S<br />

Hoyes, J.B., P37-S, P64-S, P91-S<br />

Hruby, V., S5<br />

Hsi, K-L., P76-S, P97-S<br />

Huang, G.S., P133-S<br />

Hubbard, S.J., P63-T<br />

Huddleston, M.J., P94-S<br />

Huestis, M.A., P108-T<br />

Hufnagel, P., P56-M, P65-M<br />

Hughes, C., P31-S, P37-S<br />

Hughes, K.A., P1-S, P21-T, P113-M<br />

Hunt, N., S6<br />

Hunter, C.L., P51-T<br />

Hunter, T., P6-T, R5<br />

Hunziker, P., P80-M<br />

Hurkman, W.J., P127-S<br />

Hurley, J.B., P96-T<br />

I<br />

Iasnopolski, B., P27-T<br />

Imai, B., P29-M, P5-M, P79-S<br />

Indermuhle, P., P73-S, P110-M<br />

Ingendoh, A., P53-M, P55-S<br />

Ivanetich, K.M., P7-S<br />

Ivanov, V.T., P99-T<br />

J<br />

Jackson-Machelski, E., P6-T, R5<br />

Jedrzejewski, P.T., P71-M, P73-S<br />

Jensen, O.N., P91-S<br />

Jessen, T.H., S6<br />

Jett, M., P12-T<br />

Ji, X., P23-M<br />

Jilkine, A., P66-T<br />

Johnson, B., P8-M<br />

Johnson, B.F., P8-M, P117-T, P119-M<br />

Johnson, R.S., S3<br />

Johnston-Dow, L., P27-T<br />

Jones, C., P37-S, P91-S<br />

Jones, K., P112-S<br />

Jones, R., P36-T<br />

Jou, Y-H., P114-T, P81-T<br />

Juergens, M., P38-M<br />

Juhasz, P., P89-M<br />

K<br />

Kaiser, R.J., P113-M, P1-S<br />

Kaput, G., P121-S<br />

Karnick, S., P98-M<br />

Kasch, L., R1<br />

AUTHOR INDEX<br />

Kates, S., R2<br />

Kato, M., P25-S<br />

Kato, S., P25-S<br />

Katsamba, P.S., P106-S, P137-M, R6<br />

Kauri, L.S., S10<br />

Kellard, E., P69-T<br />

Kellmann, M., P38-M<br />

Kemper, C., P130-S<br />

Kennedy, R.T., S10<br />

Kihlberg, J., P93-T<br />

Kijenski, H., P146-M<br />

Kim, B-G., P142-S<br />

Kirchner, M., P79-S<br />

Kishiyama, A., P42-T, P75-T<br />

Kitchen, D., P16-S<br />

Knudtson, K., P6-T, R3, R5<br />

Koh, J., P18-T<br />

Kojima, M., P83-M<br />

Koller, A., P54-T<br />

Komatsu, S., P132-T<br />

Konishi, H., P132-T<br />

Koob, G.F., S1<br />

Koomen, J., P108-T<br />

Kopaciewicz, W., P69-T<br />

Kosman, C.A., P27-T<br />

Köster, C., P56-M<br />

Kostrzewa, M., P52-S, P61-S<br />

Kräuter, K-O., P52-S, P56-M<br />

Krishnan, S., P35-M, P82-S<br />

Kshirsagar, B., P27-T<br />

Kumar, A., P120-T<br />

Kumar, S., P27-T<br />

Kuo, S.S., P122-M<br />

L<br />

La Rotta, A., P65-M<br />

Lach, F., P5-M<br />

Laird-Offringa, I.A., P106-S, P137-M,<br />

R6<br />

Langridge, J., P62-M<br />

Langridge, J.I., P31-S, P37-S, P39-T,<br />

P62-M, P64-S, P91-S<br />

Ledman, D.W., P36-T, P40-S<br />

Lee, C-S., P142-S<br />

Lee, H., P66-T<br />

Lee, K.A., P96-T<br />

Lee, S-W., P47-M<br />

Lee, T.D., P34-S<br />

Leicester, S., P37-S<br />

Leinonen, J., P100-S<br />

Leonard, A., P26-M<br />

Lester, P.J., P63-T<br />

Leung, D.K., P33-T<br />

Leviten, D., P6-T, R5<br />

Li, G., P107-M, P21-T<br />

Li, Z., P132-T<br />

Liddell, P.A., P49-S<br />

Liedtke, S., P53-M<br />

Lilley, K.S., R3, R4, T7<br />

Lin, M., P35-M, P82-S<br />

Lippert, J.W., P49-S<br />

Liu, L., P28-S<br />

Lively, M., R4<br />

234 JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000<br />

Liwei, Q., P8-M<br />

Lobo, M., P43-S<br />

Loboda, A., P66-T<br />

Lock, C.M., P44-M<br />

Lopez, J.C., P49-S<br />

Lopez, R., S2<br />

Lotti, R., P57-T<br />

Lubenow, H., P22-S<br />

Luk, D., P18-T<br />

Lund, K.P., P107-M, P21-T<br />

Luo, Y., P98-M<br />

Lynch, M.D., P82-S<br />

M<br />

Machado, L., P126-T<br />

Maeda, M., P87-T<br />

Mahr, K., P102-T<br />

Mahtani, M., P111-T, P112-S,<br />

P115-S<br />

Mamone, A., P112-S, P116-M,<br />

P118-S, P121-S<br />

Marks, A.R., S4<br />

Marron, M.T., T5<br />

Mason, G., P27-T<br />

Massimi, A., R3<br />

Mathews, W.R., P67-S<br />

Matsumoto, Y., P83-M<br />

Mayo, J.D., P58-S<br />

McArdle, B., P116-M, P118-S, P121-S<br />

McEldoon, W.L., P68-M<br />

Medalle, J., P29-M, P5-M<br />

Mendis, C.A., P12-T<br />

Meredith, G., S9<br />

Meys, M., P82-S<br />

Milgram, K.E., S2<br />

Millar, A., P31-S, P37-S, P37-S, P39-T,<br />

P39-T, P64-S, P91-S<br />

Miller, C.G., P15-T, T2<br />

Miller, M.J., P30-T<br />

Miller, T.B., P19-S<br />

Mills, K., R4<br />

Minarik, M., P111-T, P112-S, P115-S<br />

Minkoff, M.S., P36-T<br />

Mitsumoto, T., P25-S<br />

Mizuno, Y., P83-M<br />

Moore, R.E., P34-S<br />

Moreno, T., P78-T<br />

Morita, M., P129-T<br />

Morrison, P.T., T8<br />

Moseley, A., P48-T, P70-S<br />

Moyer, M., P48-T, P70-S<br />

Mozdzanowski, J., P23-M<br />

Muehlbauer, B., P110-M<br />

Muhlhauser, R.R., P15-T, T2<br />

Murayama, K., P129-T<br />

Myszka, D.G., P101-M, P106-S,<br />

P137-M, R6<br />

N<br />

Nadler, T., P57-T<br />

Nakayama, Y., P87-T<br />

Närvänen, A., P100-S<br />

Nedelkov, D., P105-T


Neitz, S., P38-M<br />

Nelson, J., P116-M<br />

Nelson, J.S., P50-M<br />

Nelson, R.W., P105-T<br />

Nemirovskiy, O.V., P67-S<br />

Neubert, T.A., S3<br />

Niemi, G.A., P96-T<br />

Noble, R.L., P84-T<br />

Nock, S., P110-M, P71-M, P73-S<br />

Nordhoff, E., P61-S<br />

Nutter, B., P2-M<br />

Nuwaysir, L., P51-T<br />

O<br />

Oefner, P.J., P50-M<br />

Oliver, S.G., P63-T<br />

O’Malley, R., P39-T, P64-S<br />

Osapay, G., R2<br />

Oshiro, G., P59-M<br />

Ostrowski, L., P48-T<br />

P<br />

Packer, N.H., P86-M<br />

Pakkala, M., P100-S<br />

Palaniappan, C., P121-S<br />

Pan, H., P104-M<br />

Panisko, E.A., P45-T<br />

Parker, K.C., P57-T, P82-S<br />

Pasa-tolic, L., P45-T<br />

Patel, A., P51-T<br />

Patton, W.F., P130-S<br />

Paul, R., P27-T<br />

Paul, S., P25-S<br />

Pearson, N., T5<br />

Peltier, J., P89-M<br />

Pennetti, A., P23-M<br />

Pérez-Martínez, G., P102-T<br />

Petukhova, L.O., R1, T13<br />

Pham, C., S2<br />

Pham, V., P75-T<br />

Philip, Y., P64-S<br />

Pillai, S., P57-T<br />

Pingue, R., P8-M<br />

Pirkola, K., P111-T, P112-S, P115-S<br />

Pitt, A.M., P72-T<br />

Pluskal, M.G., P72-T<br />

Pon, R.T., P13-S, P17-M, T1<br />

Pope, R.M., P32-M<br />

Porter, S.G., P26-M<br />

Pretty On Top, K., P130-S<br />

Price, W., P134-M<br />

Pryor, A., P138-T<br />

Purdy, R.H., S1<br />

Purkayastha, B., P57-T<br />

Pusch, W., P52-S<br />

Q<br />

Qian, W-j., S10<br />

Qui, J., P16-S<br />

R<br />

Rakwal, R., P132-T<br />

Randall, B.D., S3<br />

AUTHOR INDEX<br />

Randesi, M., P29-M, P5-M<br />

Ranish, J., P60-T<br />

Raska, C.S., P32-M<br />

Rauth, H., P61-S<br />

Reagin, M., P116-M<br />

Remmer, H., R2<br />

Resemann, A., P74-M<br />

Ribbe, J., P20-M, P22-S<br />

Ricart, C.A.O., P126-T<br />

Richert, C., P58-S<br />

Rist, B., P66-T<br />

Rivera, J.D., S1<br />

Robertson, M.A., P6-T, R5, T6<br />

Robinson, J., S2<br />

Rodionov, I., P99-T<br />

Rohde, E., P131-M<br />

Roinishvili, L., P28-S<br />

Roper, M.G., S10<br />

Rosato, C., R1, T13<br />

Ross, P., P36-T<br />

Ross, P.L., P40-S<br />

Roy, K., P119-M, P8-M<br />

Rubenstein, D., P32-M<br />

Ruiz-Taylor, L., P110-M<br />

Russell, D.H., P108-T<br />

S<br />

Salamon, Z., S5<br />

Sanchez, C., P12-T<br />

Sandifer, F., P7-S<br />

Sanghvi, Y.S., P17-M<br />

Sangvanich, P., P63-T<br />

Sarracino, D.A., P58-S<br />

Sasagawa, T., P83-M<br />

Saunders, M.J., T5<br />

Scaringe, S.A., P16-S, R4<br />

Schade, C., P139-S, P146-M<br />

Schafer, A., P134-M<br />

Schäfer, F., P20-M<br />

Scharer-Hernandez, N.G., P134-M<br />

Schieltz, D., P59-M<br />

Schlabach, T.L., P76-S<br />

Schneider, A., P53-M, P55-S<br />

Scholl, R., P144-T<br />

Schoppe, M., P27-T<br />

Schultz, A.J., P108-T<br />

Schürenberg, M., P56-M<br />

Schwarz, H., P134-M<br />

Schweiger-Hufnagel, U., P53-M<br />

Settineri, T.A., P51-T, P77-M<br />

Settinery, C.A., P97-S<br />

Shen, D., P112-S, P115-S<br />

Shen, S., P132-T<br />

Shen, W-Z., P98-M<br />

Shen, Y., P47-M<br />

Shephard, T.R., P90-T<br />

Shillinglaw, W., P78-T<br />

Shindo, N., P129-T<br />

Shinozawa, T., P25-S<br />

Shuster, A., P112-S, P115-S<br />

Sidhu, K.S., P63-T<br />

Siegman, F., P139-S, P139-S,<br />

P146-M<br />

<strong>AB</strong>RF 2001 <strong>AB</strong>STRACTS<br />

Sims, C.E., S9<br />

Siu, D., P27-T<br />

Slagel, J., P26-M<br />

Small, K.S., P135-T<br />

Smirnov, I.P., P40-S<br />

Smith, A., T12<br />

Smith, C.M., P138-T<br />

Smith, L.M., S11<br />

Smith, R.D., P45-T, P47-M<br />

Smith, T.M., P26-M<br />

Smith-Beckerman, D., P43-S<br />

Söhnlein, P., P20-M<br />

Somogyi, A., R2<br />

Song, Y., P9-T<br />

Sousa, M.V., P126-T<br />

Souto, L.M., P10-S<br />

Spicer, D.A., P113-M, P1-S<br />

Springer, A.L., P113-M, P1-S<br />

Spurgeon, S., P3-T<br />

Stalgies, Y., P52-S<br />

Standing, K.G., P66-T<br />

Stanick, W., P57-T<br />

Steinberg, T.H., P130-S<br />

Steinert, K., P20-M, P22-S, P22-S<br />

Steinke, L.A., T12<br />

Stenman, U-H., P100-S<br />

Stoerker, J., P58-S<br />

Stoll, R., P22-S<br />

Stolowitz, M.L., P21-T, P107-M<br />

Stults, J.T., S3<br />

Suckau, D., P65-M, P65-M, P74-M<br />

Sullivan, A.G., P63-T<br />

Suri, P.A., P27-T<br />

Susan, C.I., P94-S<br />

Sussman, M., S7<br />

Sweedler, J.V., S8<br />

Swei, A., P2-M<br />

Sword, E., P27-T<br />

Syu, W-J., P24-T<br />

T<br />

Tanaka, C.K., P127-S<br />

Taylor, R., P7-S<br />

Terrell, C.E., R1<br />

Tetzlaff, C.N., P58-S<br />

Thannhauser, T., P6-T, R4<br />

Thelemann, A., P104-M<br />

Thibault, P., P93-T<br />

Thomas, E., T7<br />

Thomas, I., P61-S<br />

Tian, J.N., P117-T<br />

Tingle, M.A., T5<br />

Titgemeyer, F., P102-T<br />

Tivel, K., S2<br />

Tolani, N., P110-M<br />

Tollin, G., S5<br />

Tomaney, A., P51-T<br />

Tomaney, A.B., P122-M<br />

Tsaprailis, G., R2<br />

Tsien, R.Y., P95-M<br />

Tsunoi, Y., P112-S<br />

Tuyet-Doan, A., P18-T<br />

Tyldesley, R., P64-S<br />

JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000 235


<strong>AB</strong>RF 2001 <strong>AB</strong>STRACTS<br />

U<br />

Ueda, Y., P123-T<br />

Unemoto, T., P87-T<br />

Vallee, M., S1<br />

V<br />

van Soest, R., P53-M<br />

VanEe, J., R3<br />

Veenstra, T., P45-T<br />

Venkat, K., P23-M<br />

Vensel, W.H., P127-S<br />

Ventura, M., S2<br />

Vepsäläinen, J., P100-S<br />

Vestal, M., P89-M<br />

Vigil-Cruz, S., R2<br />

Vinayak, R., P50-M<br />

Voss, K.O., P117-T<br />

W<br />

Waddell, K., P35-M, P82-S<br />

Wagner, P., P71-M, P73-S, P110-M<br />

Wahle, S., P20-M, P22-S<br />

Wang, Y., P119-M<br />

Ware, J.A., P67-S<br />

Washburn, M.P., P59-M<br />

Wasinger, V.C., P41-M<br />

Wegener, S.L., P13-S<br />

Wehren, A., P139-S, P146-M<br />

Wei, D., P122-M<br />

Wei, J., P54-T<br />

AUTHOR INDEX<br />

Wen, L., P4-S<br />

Wenzel, T., P52-S, P61-S<br />

West, K., P80-M<br />

Wheaton, A., P119-M<br />

Whitney, S.E., P125-M<br />

Wiebe, G.J., P13-S<br />

Wike, C., P119-M<br />

Wiley, J.P., P21-T, P107-M<br />

Wilkins, M.R., P86-M<br />

Winzeler, E., P59-M<br />

Witt, M., P74-M<br />

Witte, K.L., P110-M<br />

Wolber, D., P7-S<br />

Wolkenhauer, O.V., P63-T<br />

Wolters, D., P59-M<br />

Wong, S., P75-T, P78-T<br />

Woodland, D.L., P19-S<br />

Woods, A.S., P108-T<br />

Wu, C., P81-T, P114-T<br />

Wu, D., P81-T, P114-T<br />

Wu, P., P100-S<br />

X<br />

Xiao, W., P50-M<br />

Xiong, X., S2<br />

Y<br />

Yadav, S.P., P98-M<br />

Yan, W., P7-S<br />

Yaneva, M., T11<br />

236 JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000<br />

Yang, F.J., P81-T, P114-T<br />

Yang, G., P132-T<br />

Yang, P-C., P133-S<br />

Yanovsky, A., S2<br />

Yasui, M., P87-T<br />

Yates, III, J.R., P54-T, P59-M<br />

Yeung, A., P15-T, R4<br />

Yeung, A.T., T2<br />

Yoshikawa, M., P132-T<br />

Young, M.K., P34-S<br />

Young, P., P39-T, P62-M<br />

Young, S.G., P43-S<br />

Yu, S., P13-S, P17-M<br />

Yuan, Y.C., P143-M<br />

Yuen, S.W., P76-S, P77-M, P84-T,<br />

P97-S<br />

Z<br />

Zacharias, D.A., P95-M<br />

Zahn, M., P121-S<br />

Zappacosta, F., P94-S<br />

Zaugg, F., P110-M<br />

Zhang, B-H., P140-M, P141-T<br />

Zhang, J., P98-M<br />

Zhou, H., P66-T<br />

Zielke, H., P3-T<br />

Zieske, L.R., P76-S, P77-M<br />

Zon, J., P2-M<br />

Zrolka, A.M., P134-M<br />

Zwahlen, P., P73-S


Admon, Arie, 92<br />

Bibbs, Lisa, 1<br />

Bintzler, Douglas A., 122<br />

Carr, Steve A., 92<br />

Croker, Curtis G., 135<br />

Davis, Gary, 92<br />

De Jongh, Karen, 92<br />

Denslow, Nancy, 1<br />

Devaney, Joseph M., 67<br />

Girard, James, 67<br />

Haqqi, Talat, 111<br />

Harper, Sandra, 74<br />

Hathaway, Gary M., 1<br />

Henzel, William J., 92<br />

<strong>AB</strong>RF-98SEQ, 92<br />

automated sample analysis, 135, 151<br />

biotechnology core laboratories, 1, 151<br />

capillary electrophoresis (CE), 67<br />

core facilities, biotechnology, 1, 151<br />

data-controlled analysis, 135<br />

desalting samples, 12<br />

differential display polymerase chain reaction<br />

(DD-PCR), 87<br />

digestion, in-gel, 74<br />

dimethyl sulfoxide (DMSO), 111<br />

DNA<br />

sequencing of at core facilities, 1<br />

sequencing telomeric, 111<br />

small-scale sequencing of, 151<br />

synthesis of at core facilities, 1, 122<br />

expert systems, 135<br />

genes, differentially expressed, 87<br />

genetic testing, 67<br />

JBT<br />

JOURNAL OF BIOMOLECULAR TECHNIQUES<br />

hemochromatosis, detection of, 67<br />

high-performance liquid chromatography (HPLC),<br />

purifying primers with, 87<br />

INDEX TO VOLUME 11, 2000<br />

AUTHOR INDEX<br />

Ivanetich, Kathryn M., 1<br />

Keen, Denise A., 135<br />

Kolbas, Olivera, 74<br />

Lane, William, 92<br />

Lee, Terry D., 135<br />

Liang, Qi, 67<br />

Lin, Ching-Lun, 151<br />

McMillen, Debra A., 1<br />

Moore, Roger E., 135<br />

Moran, Laurie, 151<br />

Naeve, Clayton, 1<br />

Niece, Ronald L., 1<br />

Pearcy, John O., 135<br />

Rohde, Michale, 92<br />

SUBJECT INDEX<br />

in-gel digestion, 74<br />

laser-induced fluorescence capillary electrophoresis<br />

(LIF-CE), 67<br />

liquid chromatography (LC), protein analysis using, 135<br />

mass analysis, core facilities capable of, 1<br />

mass spectrometry (MS)<br />

sequencing of at core facilities, 1<br />

core facility use of, 1<br />

desalting and concentrating samples for, 12<br />

peptide sequencing with, 92<br />

tandem mass (MS/MS), 92, 135<br />

matrix-assisted laser desorption and ionization (MALDI)<br />

mass spectrometry<br />

sequencing of at core facilities, 1<br />

core facilities using, 1<br />

desalting samples for, 12<br />

variations in peptide signals for, 75<br />

messenger RNA, amplification of, 87<br />

oligonucleotide synthesis, 87, 122<br />

Rusnak, Felicia, 1<br />

Simpson, John T., 67<br />

Slatko, Barton, 151<br />

Speicher, David W., 74<br />

Speicher, Kaye D., 74<br />

Stahl, Douglas C., 135<br />

Steinke, Laurey, 92<br />

Terrell, Catherine E., 122<br />

Thompson, Barry H., 67<br />

Tindall, Stephen, 1<br />

Ware, Jennifer L., 151<br />

Wen, Long, 87<br />

Yadav, Satya P., 111<br />

Zhao, Xiaolan, 111<br />

peptide adsorption, 74<br />

peptide, tryptic, 74<br />

polymerase chain reaction, differential display<br />

(DD-PCR), 87<br />

JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000 237


post-source decay (PSD), 92<br />

primers<br />

sequencing of at core facilities, 1<br />

HPLC purification of, 87<br />

DNA synthesis using, 122<br />

protein<br />

sequencing of at core facilities, 1<br />

automated analysis of, 135<br />

identification using in-gel digestion, 74<br />

sequencing and purification of at core facilities, 1, 92<br />

standards for consistency of recovery of, 77<br />

proteolytic digests, desalting of, 12<br />

proteome, analysis using in-gel digestion, 74<br />

238 JOURNAL OF BIOMOLECULAR TECHNIQUES, VOLUME 11, ISSUE 4, DECEMBER 2000<br />

repeat sequences, 111<br />

robotics, DNA sequencing using, 151<br />

single-nucleotide extension, 67<br />

single-nucleotide polymorphism (SNP), 67<br />

synthesis, oligonucleotide, 87<br />

telomeric DNA, 111<br />

tandem mass spectometry (MS/MS), 92, 135<br />

tryptic peptides, 75<br />

virtual instruments, 135<br />

INDEX TO VOLUME 11, 2000


JBT<br />

JOURNAL OF BIOMOLECULAR TECHNIQUES<br />

EDITORIAL POLICY AND CALL FOR ARTICLES<br />

The Journal of Biomolecular Techniques (JBT) is a peer-reviewed publication issued quarterly by<br />

the Association of Biomolecular Resource Facilities. It was established to promote the central role<br />

biotechnology plays in contemporary research activities, to disseminate information among biomolecular<br />

resource facilities, and to communicate the biotechnology research conducted by the<br />

Association’s Research Groups, Association Members, and interested investigators.<br />

The publication has an international audience and is intended for professionals engaged<br />

in biotechnology research and service. We welcome contributions from specialists in protein<br />

and nucleic acid chemistry, mass spectrometry, bioinformatics, biomolecular resource facilities<br />

operations and management, and related areas. To keep the publication focused on topics of<br />

interest, articles are often solicited by the Editorial Board. Direct submissions or inquiries about<br />

the suitability of proposed articles may be directed to the Editor, Clayton W. Naeve, Ph.D., St.<br />

Jude Children’s Research Hospital, Memphis, TN 38101; Tel: (901) 495-3861; Fax: (901) 495-<br />

2945; email: clayton.naeve@stjude.org<br />

JBT is published in English and features three types of articles: Methods & Reviews, Tips<br />

Articles, and Research Communications. The purpose and requirements of all three types<br />

of articles are discussed below in the “Instructions for Authors.” JBT is also published electronically<br />

on the Association’s Web site at http://www.abrf.org<br />

All material submitted for publication in the Journal<br />

should be sent to<br />

Clayton W. Naeve<br />

St. Jude Children’s Research Hospital<br />

332 North Lauderdale Street<br />

Memphis, TN 38105-2794<br />

Tel: (901) 495-3689<br />

Fax: (901) 495-2945<br />

email: Clayton.Naeve@stjude.org<br />

Queries can be addressed to the above address until<br />

notification of acceptance is received. Once the article<br />

is in production, address questions to<br />

Molly Connors<br />

Copperplate Press, Inc.<br />

85 East Stewart Avenue<br />

Lansdowne, PA 19050<br />

Tel: (610) 284-5257<br />

Fax: (610) 284-5258<br />

email: mconnors@copperplatepress.com<br />

INSTRUCTIONS FOR AUTHORS<br />

JOURNAL SECTIONS<br />

Please indicate to which section you wish to submit<br />

your manuscript. However, the publisher reserves the<br />

right to place articles in whichever sections the Editorial<br />

Board deems appropriate.<br />

Methods and Reviews: Methods and Reviews articles<br />

are communications reviewing and detailing existing<br />

or emerging technologies or providing comprehensive<br />

descriptions of bioanalytical techniques. Research<br />

articles are also welcomed.<br />

Tips: Tips articles are shorter communications focusing<br />

on bioanalytical techniques or providing detailed descriptions<br />

of new methodologies or new modifications<br />

to techniques that improve the performance of the<br />

method being discussed.<br />

Research Communications: Research communications<br />

articles are very short. They are intended to foster<br />

the rapid dissemination of new information and


INSTRUCTIONS FOR AUTHORS<br />

developments in bioanalytical methodology. They<br />

may consist of new techniques, improvements to<br />

existing methods, or solutions to common problems.<br />

Research Communications are submitted by email,<br />

receive expedited on-line review, and, on acceptance,<br />

are immediately posted on the <strong>AB</strong>RF Web Server.<br />

These articles are then printed in the next edition of<br />

the Journal. Detailed instructions for submitting Research<br />

Communications can be obtained from the<br />

<strong>AB</strong>RF Web site at http://www.abrf.org<br />

SUBMISSION REQUIREMENTS<br />

Please include a cover letter indicating the name,<br />

address, telephone and fax numbers, and email<br />

address of the individual to whom correspondence<br />

and proofs are to be addressed. Also indicate to<br />

which section of the Journal the manuscript is being<br />

submitted (see Journal Sections). Wrap the package<br />

well and use extra-strength envelopes if possible. All<br />

manuscripts will be acknowledged on receipt at the<br />

editorial office. After peer review is completed,<br />

authors will receive notification of acceptance or<br />

rejection, or articles may be returned for revision and<br />

resubmission.<br />

Submissions should contain the following materials.<br />

See respective sections below for details.<br />

• Cover letter<br />

• Four copies of manuscript, including title page,<br />

abstract, references, and figure legends<br />

• Manuscript on disk, labeled<br />

• Four copies of original art, labeled<br />

• Four copies of tables<br />

• Copyright release form<br />

All material submitted for publication, including<br />

solicited articles and supplements, is subject to peer<br />

review and revision and editorial review. Only previously<br />

unpublished material will be considered for<br />

publication. Material submitted to the Journal must<br />

not be under consideration for publication elsewhere.<br />

All accepted manuscripts become the property<br />

of the Journal and may not be reproduced or<br />

published without the written permission of the Publisher.<br />

Per United States Copyright Law of 1978, the<br />

transfer of copyright from author to publisher must be<br />

explicitly stated in writing to enable the publisher to<br />

ensure maximum dissemination of the author’s work,<br />

including, but not limited to, electronic formats. A<br />

copy of this agreement, executed and signed by all<br />

authors, is required before an article can be published.<br />

FORMAT FOR ARTICLES<br />

Four copies of the manuscript should be submitted in<br />

English. Include a disk, making certain that the version<br />

on the disk matches the hard copy. Use standard<br />

8 1 /2- � 11-inch paper and 1-inch margins. Do not use<br />

“erasable” bond. All copy, including figure legends,<br />

tables, and references, must be typed double-spaced<br />

and on one side of the sheet only. Include page numbers<br />

at the top of the page.<br />

The title page should include the title of the article,<br />

all authors’ names and affiliations, a complete<br />

description of any grants supporting the research,<br />

notation of any possible conflict of interest, a shortened<br />

version of the title to use as a running head (40<br />

characters maximum), and complete contact information<br />

for the corresponding author as outlined previously.<br />

The second page of the article should contain an<br />

abstract of approximately 200 words. This page<br />

should also contain three to five descriptive nouns or<br />

key words for indexing purposes.<br />

The body of the article should be placed next, followed<br />

in turn by the references, figure legends, and<br />

tables.<br />

STYLE<br />

Dorland’s Illustrated Medical Dictionary, 28th Edition,<br />

and Merriam Webster’s Collegiate Dictionary,<br />

10th Edition, should be used as standard references.<br />

Scientific (ie, generic) names of drugs should be<br />

used when possible. Copyrighted or trade names of<br />

drugs or devices should be capitalized, and the<br />

name of the manufacturer and the manufacturer’s<br />

location (city, state, and country) should be supplied<br />

in parentheses after first mention. It is not<br />

necessary to insert trademark or registration symbols.<br />

Units of measure should be expressed in the<br />

metric system. Temperatures should be expressed in<br />

degrees Celsius. In general, style should be patterned<br />

after the American Medical Association Manual<br />

of Style, 9th Edition.<br />

REFERENCES<br />

<strong>Reference</strong>s should be cited in order in the text by<br />

superscript number; citations should follow punctuation<br />

when possible. A corresponding list of references<br />

numbered in order of citation should appear, doublespaced,<br />

at the end of the manuscript. Unpublished<br />

data, personal communications, or articles submitted


ut not yet accepted for publication should not be<br />

numbered references but should be cited parenthetically<br />

in the text. (eg, Jones SA, personal communication,<br />

April 1998). If there are up to six authors, all<br />

should be listed; if there are more than 6 authors, the<br />

first three should be listed, followed by et al. Journal<br />

titles should be abbreviated as in Index Medicus. <strong>Reference</strong>s<br />

should conform the following style:<br />

Journal Article: Doe HT, Smith MA, Jones SA, et al.<br />

Subjective evaluations of proteases and mass<br />

spectrometry. N Engl J Med 1998;25:311–322.<br />

Book: Williams DW. Molecular Weight Markers, 2nd<br />

ed, vol 1. Philadelphia: WB Saunders, 1997:113–<br />

215.<br />

Chapter in Book: Brown PG, Miller FB. Equilibration<br />

of the first dimension. In Williams DW (ed):<br />

Molecular Weight Markers, 2nd ed, vol 1. Philadelphia:<br />

WB Saunders, 1997:216–228.<br />

ILLUSTRATIONS<br />

Four sets of camera-ready, high-quality illustrations<br />

should be provided. Label each piece of art on the<br />

back with the first author’s name, abbreviated title,<br />

figure number, and an arrow indicating the top of the<br />

piece. Any lettering on the art should be large enough<br />

to be legible after necessary reduction. Shading<br />

should be used sparingly in line art and should not be<br />

dark. For computer-generated graphics, provide highquality<br />

original laser output. Legends should be supplied<br />

on a separate typewritten sheet at the end of the<br />

manuscript.<br />

Black and white illustrations will be reproduced<br />

without charge. The cost of reproduction of color<br />

illustrations may be charged to the author; an estimate<br />

will be provided to the author for approval<br />

before the article enters production. There is no<br />

charge for converting color illustrations to black and<br />

white. The publisher reserves the right to establish a<br />

reasonable limit on the number of illustrations in<br />

each article.<br />

INSTRUCTIONS FOR ELECTRONIC<br />

MANUSCRIPT SUBMISSIONS<br />

The preferred storage medium is a 3 1 /2-inch disk in a<br />

PC format; however, files in a Macintosh-compatible<br />

format will also be accepted. Files should be submitted<br />

in a standard word-processing format such as<br />

INSTRUCTIONS FOR AUTHORS<br />

Microsoft Word or WordPerfect. Although we can convert<br />

from other word-processing formats, the vagaries<br />

of the conversion process may introduce errors, especially<br />

with special characters (eg, Greek or accented<br />

letters). Do not submit ASCII text files.<br />

Each submitted disk must be clearly labeled with<br />

the name of the first author, abbreviated article title,<br />

journal title, type of equipment used to generate the<br />

disk (PC or Mac), word-processing format (including<br />

version number), and filename used.<br />

It is imperative that the file submitted on disk be<br />

the final corrected version of the manuscript and<br />

agree with the final accepted version of the submitted<br />

paper manuscript. The disk submitted should contain<br />

only the final version of the manuscript. Delete all<br />

other material from the disk.<br />

Because we will typeset directly from the electronic<br />

version, it should not contain any extraneous<br />

formatting instructions. For example,<br />

Use hard carriage returns only at the end of paragraphs<br />

or display lines (eg, titles, heads).<br />

Do not use an extra hard return between paragraphs.<br />

Do not use tabs or extra spaces at the start of a<br />

paragraph or for list entries.<br />

Do not indent runover lines in references.<br />

Turn off hyphenation and justification.<br />

Do not specify page breaks or headers.<br />

Do not specify typeface.<br />

Take care to correctly enter “one” (1) and lower<br />

case “el” (l).<br />

Take care to correctly enter “zero” (0) and capital<br />

“oh” (O).<br />

Do not embed references in footnote functions of<br />

your word-processing program.<br />

Nonstandard characters (eg, Greek letters, math symbols)<br />

should be coded consistently throughout.<br />

Tables will be typeset from author-provided files<br />

and should be submitted in word-processing format;<br />

do not provide graphics files for tables. Illustrations<br />

will be shot from hard copy; do not include them on<br />

the disk. For computer-generated graphics, provide<br />

high-quality original laser output. Figure legends<br />

should be included at the end of the electronic manuscript<br />

file.<br />

REPRINTS<br />

All contributors will be offered the opportunity to<br />

purchase reprints of their articles when they receive<br />

proofs. Reprints are offered in 100-copy increments.


JOURNAL OF BIOMOLECULAR TECHNIQUES<br />

AUTHORSHIP RESPONSIBILITY, FINANCIAL DISCLOSURE, AND COPYRIGHT TRANSFER<br />

Manuscript Title: _________________________________________________________________________________________________<br />

Manuscript Authors: ______________________________________________________________________________________________<br />

Each author must read and sign the following statement. If necessary, photocopy this document and distribute to<br />

coauthors for their original ink signatures. Please compile all forms and include them with the manuscript.<br />

CONDITIONS OF SUBMISSION<br />

Retained Rights: Other than copyright, all proprietary rights (such as patent rights) are retained by the authors.<br />

Originality: The authors warrant that this submission is an original work. Neither this work nor a similar work has been<br />

published or will be submitted for publication elsewhere while under consideration by the Journal.<br />

Authorship Responsibility: The authors certify that they have participated sufficiently in the intellectual content, the<br />

analysis of data, if applicable, and the writing of this work to take public responsibility for it. They have reviewed the final<br />

version of the work, believe it represents valid work, and approve it for publication. Moreover, should the editors of the Journal<br />

request the data on which the work is based, they shall produce it.<br />

Disclaimer: The undersigned warrant that this work contains no libelous or unlawful statements and does not infringe<br />

on the rights or others. If excerpts (text or figures) from copyrighted works are included, written permission will be secured<br />

by the authors before submission, and credit will be properly acknowledged.<br />

Institutional Review Board/Animal Care Committee Approval: The undersigned authors certify that their institutions<br />

have approved the protocol for any investigation involving humans or animals and that all experimentation was conducted<br />

in conformity with ethical and humane principles of research.<br />

TRANSFER OF COPYRIGHT<br />

Authors’ Own Work: The undersigned authors hereby transfer, assign, or otherwise convey all copyright ownership<br />

worldwide, in all languages, and in all forms of media now or hereafter known, to the Association of Biomolecular Resource<br />

Facilities in the event that such work is accepted for publication in the Journal. To reproduce any figure or text from this article<br />

in future works of their own, the authors must obtain written permission. Such permission cannot be unreasonably withheld<br />

by the copyright holder.<br />

Government Employees:<br />

❏ If this article exists in the public domain because it was written as part of the official duties of the authors as employees<br />

of the U.S. Government, check this box.<br />

Note to Government Employees: If this work has been written in the course of employment by the U.S. Government, the<br />

box should be checked above and a copy of the relevant departmental statement of policy attached. A work prepared by a government<br />

employee as part of his/her official duties is called a “work of the U.S. Government” and is not subject to copyright. If<br />

the work is not prepared as part of the employee’s official duties, it may be copyrighted.<br />

Financial Disclosure: The undersigned authors certify that they have no commercial associations (eg, consultancies, stock<br />

ownership, equity interest, patent-licensing arrangements) that may pose a conflict of interests in connection with the submitted<br />

article, except as disclosed on a separate attachment. All funding sources supporting the work and all institutional or<br />

corporate affiliations of the authors should be acknowledged in the article.<br />

Signature Printed Name Date<br />

❏ Author’s own work ❏ Work for hire ❏ Government<br />

Signature Printed Name Date<br />

❏ Author’s own work ❏ Work for hire ❏ Government<br />

Signature Printed Name Date<br />

❏ Author’s own work ❏ Work for hire ❏ Government<br />

Signature Printed Name Date<br />

❏ Author’s own work ❏ Work for hire ❏ Government

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

Saved successfully!

Ooh no, something went wrong!