15.02.2013 Views

2001 - Center for Advanced Biotechnology and Medicine - Rutgers ...

2001 - Center for Advanced Biotechnology and Medicine - Rutgers ...

2001 - Center for Advanced Biotechnology and Medicine - Rutgers ...

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.

C A B M<br />

<strong>Center</strong> <strong>for</strong><br />

<strong>Advanced</strong> <strong>Biotechnology</strong><br />

<strong>and</strong> <strong>Medicine</strong><br />

http://cabm.umdnj.edu<br />

2002 Report<br />

An <strong>Advanced</strong> Technology <strong>Center</strong><br />

of<br />

The New Jersey Commission on Science <strong>and</strong> Technology<br />

Jointly Administered<br />

By<br />

<strong>Rutgers</strong>, The State University of New Jersey<br />

<strong>and</strong> by<br />

The University of <strong>Medicine</strong> <strong>and</strong> Dentistry of New Jersey


CONTENTS<br />

Director’s Overview 3<br />

Research Programs <strong>and</strong> Laboratories<br />

Cell & Developmental Biology 6<br />

Molecular Neurobiology Cory Abate-Shen 7<br />

Molecular Chronobiology Isaac Edery 9<br />

Tumor Virology Céline Gélinas 11<br />

Growth & Differentiation Control Fang Liu 13<br />

Protein Targeting Peter Lobel 15<br />

Developmental Neurogenetics James Millonig 17<br />

Mammalian Embryogenesis Michael Shen 19<br />

Viral Trans<strong>for</strong>mation Eileen White 21<br />

Molecular Neurodevelopment Mengqing Xiang 24<br />

Structural Biology 25<br />

Biomolecular Crystallography Edward Arnold 26<br />

Protein NMR Spectroscopy Gaetano Montelione 31<br />

Protein Microchemistry Stanley Stein 33<br />

Protein Crystallography Ann Stock 34<br />

Molecular Genetics 36<br />

Protein Engineering Stephen Anderson 37<br />

Viral Pathogenesis Arnold Rabson 39<br />

Molecular Virology Aaron Shatkin 41<br />

Education, Training <strong>and</strong> Technology Transfer 43<br />

Lectures <strong>and</strong> Seminars 44<br />

CABM Lecture Series 45<br />

Annual Retreat 46<br />

15 th Annual Symposium 48<br />

Undergraduate, Graduate Students <strong>and</strong> Postdoctoral Fellows 52<br />

Patents 56<br />

Fiscal In<strong>for</strong>mation 57<br />

Grants <strong>and</strong> Contracts<br />

__________________________________________________________________________________<br />

<strong>Center</strong> <strong>for</strong> <strong>Advanced</strong> <strong>Biotechnology</strong> <strong>and</strong> <strong>Medicine</strong><br />

2


Director’s Overview<br />

The community of research scientists at CABM is always looking <strong>for</strong>ward to new<br />

experimental findings that will promote our mission – advancing basic knowledge<br />

in the life sciences to improve human health. On this occasion, the completion of<br />

our 15 th year, it is appropriate <strong>and</strong> reassuring to reflect on some of our recent<br />

accomplishments in research, teaching <strong>and</strong> service.<br />

CABM in its short lifetime has already become an important contributor to the<br />

biosciences revolution. As described in the following pages, CABM scientists<br />

continued to report the results of their latest research, designed to underst<strong>and</strong><br />

better <strong>and</strong> ultimately prevent AIDS, cancer, neurological disorders <strong>and</strong> other lifethreatening<br />

diseases. During the past year nearly 100 peer-reviewed publications<br />

from CABM laboratories have appeared in high impact international journals<br />

including Development, Genes <strong>and</strong> Development, EMBO Journal, Molecular Cell,<br />

Molecular <strong>and</strong> Cellular Biology, Nature, Nature Structural Biology, Proceedings<br />

of the National Academy of Sciences <strong>and</strong> Science.<br />

Generous support <strong>for</strong> this important work was provided by competitive awards<br />

from numerous public <strong>and</strong> private sources. We are grateful to the National<br />

Institutes of Health (NIH), U.S. Army Medical Research & Materiel Comm<strong>and</strong>,<br />

National Science Foundation (NSF), Veterans Administration, N.J. Commission<br />

on Science <strong>and</strong> Technology R&D Excellence Program, N.J. Commission on<br />

Cancer Research <strong>and</strong> Governor’s Council on Autism; the Howard Hughes<br />

Medical Institute (HHMI), American Association <strong>for</strong> Cancer, American Heart<br />

Association, Leukemia <strong>and</strong> Lymphoma Society, March of Dimes, Kimmel<br />

Foundation <strong>for</strong> Cancer Research <strong>and</strong> Sinsheimer Fund; the Ara Parseghian<br />

Medical Research, Burroughs Wellcome, AHEPA Cancer Research, Cure <strong>for</strong><br />

Lymphoma, Emerald, Janssen Research, National Ataxia, <strong>and</strong> Whitehall<br />

Foundations; <strong>and</strong> Janssen-Cilag, Johnson & Johnson, Roche, Genzyme,<br />

GeneFormatics, Human Genome Sciences, Merck, Aventis <strong>and</strong> Genencor. In<br />

addition to two NIH MERIT awards <strong>and</strong> two HHMI investigators, CABM is home<br />

to the NIH-funded Northeast Structural Genomics Consortium.<br />

Besides their strong commitment to classroom teaching in courses <strong>for</strong><br />

undergraduate, graduate <strong>and</strong> medical students, CABM researchers trained <strong>and</strong><br />

mentored many postdoctoral fellows <strong>and</strong> students in their laboratories in<br />

preparation <strong>for</strong> positions of scientific leadership in academia <strong>and</strong> industry. The<br />

__________________________________________________________________________________<br />

<strong>Center</strong> <strong>for</strong> <strong>Advanced</strong> <strong>Biotechnology</strong> <strong>and</strong> <strong>Medicine</strong><br />

3


Director’s Overview<br />

Annual Retreat <strong>and</strong> CABM Lecture Series have again this year provided<br />

opportunities <strong>for</strong> students to present <strong>and</strong> defend their work <strong>and</strong> to hear <strong>and</strong> interact<br />

with exciting speakers visiting from other leading educational institutions. The<br />

15 th Annual CABM Symposium, “Structural Genomics in Pharmaceutical<br />

Design”, was sponsored by 18 organizations <strong>and</strong> enjoyed by nearly 250<br />

participants. The meeting fostered productive collaborations <strong>and</strong> licensing of<br />

intellectual property <strong>for</strong> industrial development, adding to CABM’s economic<br />

impact in New Jersey <strong>and</strong> across the nation.<br />

CABM investigators served on various advisory, faculty search, departmental <strong>and</strong><br />

other committees at <strong>Rutgers</strong> University <strong>and</strong> UMDNJ-Robert Wood Johnson<br />

Medical School. To foster translational aspects of our mission, several also have<br />

appointments <strong>and</strong> leadership roles in clinical departments <strong>and</strong> in the Cancer<br />

Institute of New Jersey, e.g. as Associate Director <strong>for</strong> Basic Research <strong>and</strong><br />

Scientific Director of the Gallo Prostate Cancer <strong>Center</strong>. At the national <strong>and</strong><br />

international levels, CABM faculty participate in meetings as invited speakers,<br />

serve on many journal editorial boards <strong>and</strong> meeting organizational committees,<br />

several grant review panels of the NIH <strong>and</strong> NSF, the National Cancer Institute<br />

Board of Counselors, the HHMI Cell Biology Review Panel <strong>and</strong> advisory boards<br />

of other government agencies, professional societies, foundations <strong>and</strong> companies.<br />

This has been a challenging <strong>and</strong> productive year, with CABM scientists working<br />

at the frontiers of structural biology, molecular genetics <strong>and</strong> cell <strong>and</strong><br />

developmental biology, using <strong>and</strong> creating novel systems to translate genomic<br />

sequences into protein function <strong>and</strong> gaining in<strong>for</strong>mation <strong>and</strong> insights that are<br />

having positive impacts on human health. After 1-1/2 decades of outst<strong>and</strong>ing<br />

progress, 2002 promises exciting, new scientific advances at CABM.<br />

Aaron J. Shatkin, Professor <strong>and</strong> Director<br />

Shatkin@cabm.rutgers.edu<br />

http://cabm.umdnj.edu<br />

__________________________________________________________________________________<br />

<strong>Center</strong> <strong>for</strong> <strong>Advanced</strong> <strong>Biotechnology</strong> <strong>and</strong> <strong>Medicine</strong><br />

4


Research Programs <strong>and</strong> Laboratories<br />

__________________________________________________________________________________<br />

<strong>Center</strong> <strong>for</strong> <strong>Advanced</strong> <strong>Biotechnology</strong> <strong>and</strong> <strong>Medicine</strong><br />

5


Cell & Developmental Biology<br />

Laboratory Faculty Director<br />

Molecular Neurobiology Cory Abate-Shen 7<br />

Molecular Chronobiology Isaac Edery 9<br />

Tumor Virology Céline Gélinas 11<br />

Growth & Differentiation Control Fang Liu 13<br />

Protein Targeting Peter Lobel 15<br />

Developmental Neurogenetics James Millonig 17<br />

Mammalian Embryogenesis Michael Shen 19<br />

Viral Trans<strong>for</strong>mation Eileen White 21<br />

Molecular Neurodevelopment Mengqing Xiang 24<br />

__________________________________________________________________________________<br />

<strong>Center</strong> <strong>for</strong> <strong>Advanced</strong> <strong>Biotechnology</strong> <strong>and</strong> <strong>Medicine</strong><br />

6


Cory Abate-Shen, Ph.D.<br />

Resident Faculty Member, CABM; Professor, UMDNJ Robert Wood Johnson Medical<br />

School, Department of Neuroscience <strong>and</strong> Cell Biology <strong>and</strong> Department of <strong>Medicine</strong>,<br />

Scientific Director of the Dean <strong>and</strong> Betty Gallo Prostate Cancer Institute, Cancer<br />

Institute of NJ; Head, Division of Developmental <strong>Medicine</strong> <strong>and</strong> Research-Department of<br />

<strong>Medicine</strong><br />

Molecular Neurobiology Laboratory<br />

Dr. Cory Abate-Shen joined CABM in August 1991 after spending three years as a postdoctoral <strong>and</strong> research fellow in Tom Curran’s laboratory<br />

at the Roche Institute of Molecular Biology. Her work at Roche contributed to characterization of the DNA binding <strong>and</strong> transcriptional properties<br />

of the regulatory proteins Fos <strong>and</strong> Jun. Dr. Abate-Shen received her Ph.D. from Cornell University Medical College where she was awarded the<br />

Vincent du Vigneaud Award <strong>for</strong> Excellence in Graduate Research. She is a recipient of a Sinsheimer Scholar Award <strong>and</strong> an NSF Young<br />

Investigator Award <strong>and</strong> received a career recognition award from The American Society <strong>for</strong> Cell Biology. Dr. Abate-Shen is a member of the<br />

NIH Study Section on Cell Development <strong>and</strong> Function. She was recently appointed head of a new division in the Department of <strong>Medicine</strong>.<br />

My long-term interests are to underst<strong>and</strong> the molecular mechanisms underlying homeobox gene<br />

function during embryogenesis <strong>and</strong> how aberrant homeobox function contributes to oncogenesis.<br />

A major focus of my recent work has been the development of animal models to elucidate the<br />

roles of particular homeobox genes in development, oncogenesis <strong>and</strong> embryogenesis. We have<br />

found that homeobox genes Msx1 <strong>and</strong> Pax3 are co-expressed in migrating limb muscle<br />

precursors, which are committed myoblasts that do not express myogenic differentiation genes<br />

such as MyoD. Msx1 negatively regulates differentiation of migrating limb muscle precursor<br />

cells through its ability to antagonize the myogenic activity of Pax3. Forced misexpression of<br />

Msx1 in cell culture <strong>and</strong> in chicken embryos inhibits MyoD expression as well as muscle<br />

differentiation. Conversely, <strong>for</strong>ced misexpression of Pax3 activates MyoD expression <strong>and</strong><br />

myogenesis, while this effect is counteracted by Msx1. Moreover, the Msx1 protein product<br />

interacts with Pax3, thereby inhibiting the DNA binding activity of Pax3 as well as its ability to<br />

transcriptionally activate MyoD regulatory elements. A major focus of our current research is the<br />

elucidation of additional protein partners that <strong>for</strong>m functional associations with Msx proteins in<br />

vivo. Analysis of a functional role <strong>for</strong> Msx in breast <strong>and</strong> other carcinomas, as well as the<br />

identification of downstream targets <strong>for</strong> Msx, represent another important focus of our current<br />

studies. Our Msx1 transgenic mice provide an excellent resource to pursue these studies.<br />

Prostate cancer is the most commonly diagnosed neoplasm, <strong>and</strong> ranks second to lung cancer as<br />

the leading cause of cancer death in American men. To investigate early stages of initiation <strong>and</strong><br />

progressionof prostate cancer, we have developed a mouse model using the Nkx3.1 homeobox<br />

gene, which is expressed specifically in the developing <strong>and</strong> adult prostate, <strong>and</strong> whose targeted<br />

disruption results in defects of prostate differentiation <strong>and</strong> function. Notably, human NKX3.1<br />

maps to chromosomal region 8p21, which is deleted in ~60-80% of prostate tumors. Moreover,<br />

loss of 8p21 represents an early event in carcinogenesis since it is also deleted in prostatic<br />

intraepithelial neoplasia (PIN), the major precursor of prostate carcinoma in humans. We have<br />

found that Nkx3.1 mutant mice display lesions that resemble human PIN with respect to their<br />

histological appearance as well as the molecular changes that occur in early stages of<br />

__________________________________________________________________________________<br />

<strong>Center</strong> <strong>for</strong> <strong>Advanced</strong> <strong>Biotechnology</strong> <strong>and</strong> <strong>Medicine</strong><br />

7


__________________________________________________________________________________<br />

<strong>Center</strong> <strong>for</strong> <strong>Advanced</strong> <strong>Biotechnology</strong> <strong>and</strong> <strong>Medicine</strong><br />

Cory Abate<br />

carcinogenesis, such as loss of the basal cell layer of the prostate epithelium. Based on our<br />

findings we have proposed that interactions between tissue-specific regulators, such as Nkx3.1,<br />

<strong>and</strong> broad-spectrum tumor suppressors, such as Pten, underlie the distinct phenotypes of different<br />

cancers. By utilizing mouse models, we are beginning to assemble a molecular pathway <strong>for</strong><br />

prostate carcinogenesis that includes loss of NKX3.1 as an initiating event followed by loss of<br />

PTEN as a later event leading to progression. Future goals include the identification of additional<br />

molecular alterations that characterize early stage prostate carcinogenesis, further elucidation of<br />

a molecular pathway <strong>for</strong> prostate carcinogenesis through the generation of new mutant mouse<br />

models, <strong>and</strong> the application of these mutant mouse models <strong>for</strong> the design of new therapeutics.<br />

Publications:<br />

Bendall, A. J. <strong>and</strong> Abate-Shen, C. (2000). Roles <strong>for</strong> MSX <strong>and</strong> DLX homeoproteins in vertebrate<br />

development. Gene. 247:17-31.<br />

Abate-Shen, C. <strong>and</strong> Shen, M. M. (2000). Molecular genetics of prostate cancer. Genes Dev.<br />

14:2410-2434.<br />

Hu, G., Price, S., Shen, M.M. <strong>and</strong> Abate-Shen, C. (<strong>2001</strong>). Msx homeobox genes inhibit<br />

differentiation by upregulation of Cyclin D1. Development 128:2373-2384.<br />

Pizette, S., Abate-Shen, C. <strong>and</strong> Nisw<strong>and</strong>er, L. (<strong>2001</strong>). BMP mediates vertebrate limb growth <strong>and</strong><br />

dorsal-ventral patterning by differential use of two homeodomain proteins. Development<br />

128:4463-4474.<br />

Hovde, S., Abate-Shen, C. <strong>and</strong> Geiger, J.H. (<strong>2001</strong>). Crystal structure of the Msx-1<br />

homeodomain/DNA complex. Biochemistry 40: 12013-21.<br />

Cunha, G. R., Donjacour, A. A., Hayward, S.W., Thomson, A. A., Marker, P. C., Abate-Shen,<br />

C., Shen, M. M. <strong>and</strong> Dahiya, R. (2002). Cellular <strong>and</strong> molecular biology of prostatic development.<br />

In: Prostate Cancer: Principles <strong>and</strong> Practice. Kantoff, P. W., Carroll, P. D'Amico, A. (eds.),<br />

Lippincott, Williams <strong>and</strong> Wilkins, Philadelphia, pp. 16-28.<br />

Kim, M., Desai, R., Cardiff, N., Bhatia-Gaur, R., Banach-Petrosky, W., Parsons, R., Shen, M. <strong>and</strong><br />

Abate-Shen, C. Cooperativity of tissue-specific <strong>and</strong> broad-spectrum tumor suppressor genes in a<br />

mouse model of prostate carcinogenesis. PNAS. In Press.<br />

Stein S. <strong>and</strong> Abate-Shen C. Homeobox Genes <strong>and</strong> Cancer. In: Schwab M (ed.) Encyclopedic<br />

Reference of Cancer. Springer-Verlag, Berlin Heidelberg New York. In Press.<br />

8


Isaac Edery, Ph.D.<br />

Resident Faculty Member, CABM; Associate Professor, <strong>Rutgers</strong> University, Department<br />

of Molecular Biology <strong>and</strong> Biochemistry<br />

Molecular Chronobiology Laboratory<br />

Dr. Isaac Edery completed his doctoral studies in biochemistry as a Royal Canadian Cancer Research Fellow under Dr. Nahum Sonenberg at<br />

McGill University in Montreal. His Ph.D. research focused on the role of the eukaryotic mRNA cap structure during protein synthesis <strong>and</strong><br />

precursor mRNA splicing. Subsequently, he was in the laboratory of Dr. Michael Rosbash at Br<strong>and</strong>eis University where he pursued<br />

postdoctoral studies aimed at underst<strong>and</strong>ing the time-keeping mechanism underlying biological clocks. He joined CABM in 1993, <strong>and</strong> his<br />

research is supported by NIH. Dr. Edery is a member of the editorial board of Chronobiology International.<br />

A wide range of organisms from bacteria to humans exhibit daily or circadian rhythms in<br />

physiological <strong>and</strong> behavioral phenomena that are controlled by endogenous pacemakers or<br />

clocks. Circadian clocks are precisely synchronized to the 24-hr solar day by the daily lightdark<br />

<strong>and</strong> temperature cycles, enabling organisms to per<strong>for</strong>m activities at advantageous times<br />

<strong>and</strong> manifest appropriate seasonal responses. We use the fruitfly Drosophila as our model<br />

system to underst<strong>and</strong> the cellular <strong>and</strong> biochemical bases underlying clock function.<br />

The isolation of "clock genes" has provided significant insights into the molecular<br />

underpinnings governing circadian rhythms. A common theme in clocks from bacteria to<br />

humans is that at the "heart" of these pacemakers lie transcriptional-translational feedback<br />

loops. The best characterized animal model system <strong>for</strong> a circadian clock is Drosophila<br />

melanogaster, where four clock proteins termed PERIOD (PER),TIMELESS (TIM), dCLOCK<br />

(CLK) <strong>and</strong> CYCLE (CYC) function in a negative transcriptional autoregulatory loop. CLK <strong>and</strong><br />

CYC are members of the basic-helix-loop-helix (bHLH)/PAS (PER-ARNT-SIM) superfamily<br />

of transcription factors <strong>and</strong> are required <strong>for</strong> the daily stimulation of per <strong>and</strong> tim expression. PER<br />

<strong>and</strong> TIM <strong>for</strong>m a complex in the cytoplasm that enters the nucleus in a temporally gated manner<br />

where they bind the CLK-CYC heterodimer, blocking its DNA binding activity. In the absence<br />

of denovo synthesis, the concentrations of PER <strong>and</strong> TIM in the nucleus decrease below<br />

threshold levels, relieving autoinhibition <strong>and</strong> enabling the next round of per <strong>and</strong> tim transcript<br />

accumulation. Posttranscriptional mechanisms play an important role because they introduce<br />

"biochemical time constraints" that stretch the transcriptional feedback loop to ~24 hr <strong>and</strong> also<br />

allow it to respond to external stimuli. For example, light evokes the rapid degradation of TIM,<br />

the primary clock-specific photoresponse resetting the oscillatory mechanism. A blue-light<br />

photoreceptor called CRYPTOCHROME (CRY) has been implicated in transducing photic<br />

signals to TIM. Furthermore, the cytoplasmic phosphorylation of PER by the kinase DOUBLE-<br />

TIME (DBT) renders PER unstable. Cytoplasmic PER is stabilized by interacting with TIM<br />

which ensures that the accumulation <strong>and</strong> nuclear entry of the PER-TIM complex is a slow<br />

process, creating a time-window <strong>for</strong> daily increases in the levels of per <strong>and</strong> tim transcripts.<br />

Our studies are geared towards isolating all the components that comprise a circadian<br />

timekeeping device, characterizing their interactions within the various feedback loops <strong>and</strong><br />

__________________________________________________________________________________<br />

<strong>Center</strong> <strong>for</strong> <strong>Advanced</strong> <strong>Biotechnology</strong> <strong>and</strong> <strong>Medicine</strong><br />

9


__________________________________________________________________________________<br />

<strong>Center</strong> <strong>for</strong> <strong>Advanced</strong> <strong>Biotechnology</strong> <strong>and</strong> <strong>Medicine</strong><br />

Isaac Edery<br />

underst<strong>and</strong>ing how the daily changes in visible light modulate the oscillatory mechanism. In<br />

addition, we recently showed that the splicing efficiency of an intron in the 3' untranslated region of<br />

per is thermosensitive. At lower temperatures this intron is spliced more efficiently leading to higher<br />

levels of per mRNA <strong>and</strong> mainly daytime activity. By regulating the splicing efficiency of this intron<br />

flies are active during the warmer daytime hours during autumn/winter days typical of temperate<br />

zones <strong>and</strong> avoid the midday sun by manifesting mainly nocturnal activity during the warmer<br />

spring/summer seasons. We are continuing these studies to underst<strong>and</strong> how a clock helps measure<br />

calendar time by adapting to seasonal changes in temperature <strong>and</strong> daylength.<br />

Given the similarities in the circadian timing mechanisms operating in Drosophila <strong>and</strong> mammals, we<br />

anticipate that our findings will continue to provide important insights into disorders associated with<br />

clock malfunction in humans, including manic-depression, seasonal affective disorders (SAD or<br />

winter depression), chronic sleep problems in the elderly <strong>and</strong> symptoms associated with transmeridian<br />

flight ("jet-lag") <strong>and</strong> shift-work.<br />

Publications:<br />

Bae, K., Lee, C., Hardin, P.H. <strong>and</strong> Edery, I. (2000). dCLOCK is present in limiting amounts<br />

<strong>and</strong> likely mediates daily interactions between the dCLOCK-CYC transcription factor <strong>and</strong> the<br />

PER-TIM complex. J. of Neurosci. 20, 1746-1753.<br />

Edery, I. (2000). Circadian rhythms in a nutshell. Physiol. Genomics 3, 59-74.<br />

Kim, E.Y., Bae, K., Ng, F.S., Glossop, N.R.J., Hardin, P.E. <strong>and</strong> Edery, I. Drosophila CLOCK<br />

protein is under posttranscriptional control <strong>and</strong> influences light-induced activity. Neuron. In<br />

press.<br />

10


Céline Gélinas, Ph.D.<br />

Resident Faculty Member, CABM; Professor, UMDNJ-Robert Wood Johnson Medical<br />

School, Department of Biochemistry<br />

Tumor Virology Laboratory<br />

Dr. Céline Gélinas came to CABM in September 1988 from the University of Wisconsin where she conducted postdoctoral studies with Nobel<br />

laureate Dr. Howard M. Temin. She earned her Ph.D.at the Université de Sherbrooke in her native country, Canada, <strong>and</strong> received a number of<br />

honors including the Jean-Marie Beauregard Award <strong>for</strong> Academic <strong>and</strong> Research Excellence <strong>and</strong> the National Cancer Institute of Canada King<br />

George V Postdoctoral Fellowship. Her work is currently funded by the NIH <strong>and</strong> The Cure <strong>for</strong> Lymphoma Foundation.<br />

Our laboratory has a long-st<strong>and</strong>ing interest in the role of the Rel/NF-κB proteins in the onset <strong>and</strong><br />

progression of hematopoietic <strong>and</strong> solid tumors. Proteins in the Rel/NF-κB-family of transcription<br />

factors play fundamental roles in immune <strong>and</strong> inflammatory responses, <strong>and</strong> are implicated in the<br />

control of cell proliferation, the inhibition of apoptosis <strong>and</strong> in oncogenesis. Experimental<br />

evidence linking deregulated Rel/NF-κB activity to human cancer has emerged in recent years,<br />

consistent with the acute oncogenicity of the Rel/NF-κB oncoprotein v-Rel in inducing fatal<br />

leukemia/lymphomas in animal models. Aberrant Rel/NF-κB activity is found in various human<br />

leukemias, lymphomas, myelomas <strong>and</strong> Hodgkin’s disease. Chromosomal amplification,<br />

rearrangement, overexpression <strong>and</strong>/or constitutive activation of the rel <strong>and</strong> nf-κB genes are also<br />

observed in breast, colon, lung, ovarian <strong>and</strong> prostate cancer. The viral <strong>and</strong> cellular Rel proteins<br />

offer a powerful system to study how the Rel/NF-κB factors function in normal lymphoid cells<br />

<strong>and</strong> to underst<strong>and</strong> how their aberrant activities lead to cancer. Our research focuses on the<br />

transcriptional activity <strong>and</strong> regulation of the Rel/NF-κB factors, on their role in cell growth,<br />

apoptosis <strong>and</strong> oncogenesis, <strong>and</strong> on the cellular genes that they control.<br />

In prior studies, we demonstrated that the transcriptional <strong>and</strong> anti-apoptotic activities of the v-Rel<br />

oncoprotein were necessary <strong>for</strong> its oncogenic function. Our analyses characterizing Rel-mediated<br />

effects on cell survival <strong>and</strong> neoplasia have recently demonstrated the ability of a subset of<br />

mammalian cellular Rel/NF-κB factors to malignantly trans<strong>for</strong>m primary lymphoid cells.<br />

Detailed analyses of the determinants required <strong>for</strong> this activity are starting to provide important<br />

insights into the mechanisms involved in tumors associated with aberrant Rel/NF-kB function.<br />

Our recent experiments suggest that the specificity of target gene activation is likely to play a<br />

critical role in the oncogenic conversion of cellular Rel/NF-kB factors. Our ongoing functional<br />

characterization of the Rel/NF-kB target genes Bfl-1/A1 <strong>and</strong> TAPIR <strong>and</strong> the analysis of their<br />

transcriptional regulation by Rel/NF-κB offers new avenues to characterize the Rel/NF-kB<br />

signaling pathway <strong>and</strong> its role in cell survival. This comprehensive approach will help to clarify<br />

the mechanism by which Rel/NF-κB proteins function in the immune system <strong>and</strong> in oncogenesis.<br />

In addition, since Rel/NF-κB activity has been implicated in various disease conditions, the<br />

systematic analysis of Rel/NF-κB regulation <strong>and</strong> relevant Rel/NF-κB target genes may also<br />

provide important insights into novel approaches <strong>for</strong> therapeutic intervention.<br />

__________________________________________________________________________________<br />

<strong>Center</strong> <strong>for</strong> <strong>Advanced</strong> <strong>Biotechnology</strong> <strong>and</strong> <strong>Medicine</strong><br />

11


Publications:<br />

__________________________________________________________________________________<br />

<strong>Center</strong> <strong>for</strong> <strong>Advanced</strong> <strong>Biotechnology</strong> <strong>and</strong> <strong>Medicine</strong><br />

Céline Gélinas<br />

Luque, I., Zong, W.-X., Chen, C. <strong>and</strong> Gélinas, C. (2000). N-terminal determinants of IκBα<br />

necessary <strong>for</strong> the cytoplasmic regulation of c-Rel. Oncogene 19: 1239-1244.<br />

Chen C., Edelstein, L.C. <strong>and</strong> Gélinas, C. (2000). Rel/NF-κB directly activates expression of the<br />

apoptosis inhibitor Bcl-xL. Mol. Cell. Biol. 20: 2687-2695.<br />

Ravi, R., Bedi, G.C., Engstrom, L.W., Zeng, Q., Mookerjee, B., Gélinas, C., Fuchs, E.J. <strong>and</strong><br />

Bedi, A. (<strong>2001</strong>) Regulation of death receptor expression <strong>and</strong> TRAIL/Apo2L-induced apoptosis<br />

by NF-κB. Nature Cell Biol. 3: 409-416.<br />

Shah, N., Thomas, T.J., Lewis, J.S., Klinge, C.M., Shirahata, A., Gélinas, C. <strong>and</strong> Thomas, T.<br />

(<strong>2001</strong>). Regulation of estrogenic <strong>and</strong> nuclear factor κB functions by polyamines <strong>and</strong> their role in<br />

polyamine analog-induced apoptosis of breast cancer cells. Oncogene 20: 1715-1729.<br />

12


Fang Liu, Ph.D.<br />

Resident Faculty Member, CABM; Assistant Professor, <strong>Rutgers</strong> University, Susan<br />

Lehman Cullman Laboratory <strong>for</strong> Cancer Research, Department of Chemical Biology,<br />

Ernest Mario School of Pharmacy<br />

Growth <strong>and</strong> Differentiation Control Laboratory<br />

Dr. Fang Liu obtained her B.S. in biochemistry from Beijing University <strong>and</strong> Ph.D. in biochemistry from Harvard University working with Dr.<br />

Michael R. Green. She conducted postdoctoral research with Dr. Joan Massagué at Memorial Sloan-Kettering Cancer <strong>Center</strong> <strong>and</strong> joined CABM<br />

in 1998. Dr. Liu has received awards from the American Association <strong>for</strong> Cancer Research-National Foundation <strong>for</strong> Cancer Research, the<br />

Pharmaceutical Research <strong>and</strong> Manufacturers of America Foundation, the Burroughs Wellcome Fund, <strong>and</strong> the Sidney Kimmel Foundation <strong>for</strong><br />

Cancer Research. She also obtained fellowships from the K.C. Wong Education Foundation <strong>and</strong> the Jane Coffin Childs Memorial Fund <strong>for</strong><br />

Medical Research.<br />

Inhibition of SMAD Transcriptional Activity by Cyclin-Dependent Kinase<br />

Phosphorylation: TGF-ß is the most relevant physiological inhibitor of cell proliferation of a<br />

wide variety of cell types. During the earliest stage of tumorigenesis, the ability of TGF-ß to<br />

inhibit cell growth enables it to act as a potent tumor suppressor. TGF-ß inhibits cell<br />

proliferation by causing cell cycle arrest at the G1 phase. SMAD proteins are c<strong>and</strong>idate tumor<br />

suppressors <strong>and</strong> can mediate the TGF-ß growth-inhibitory effects by regulating the expression of<br />

several cell cycle-related genes. We have recently discovered that certain SMAD proteins are<br />

major substrates <strong>for</strong> cyclin-dependent kinases (CDKs). We expect that our findings will have a<br />

major impact in the cancer research field <strong>and</strong> may be useful <strong>for</strong> therapeutic applications.<br />

TGF-ß-Inducible Gene Regulation: SMAD proteins can mediate trans<strong>for</strong>ming growth factor-ß<br />

(TGF-ß) inducible transcriptional responses. In an ef<strong>for</strong>t to search <strong>for</strong> TGF-ß/SMAD inducible<br />

genes, we identified Smad7, which is rapidly upregulated by TGF-ß <strong>and</strong> can antagonize TGF-ß<br />

signaling by binding to <strong>and</strong> inhibiting the TGF-ß receptor function. We found that TGF-ß can<br />

stabilize Smad7 mRNA as well as activate Smad7 transcription. The Smad7 promoter is the first<br />

TGF-ß responsive promoter identified in vertebrates that contains the 8 base pair palindromic<br />

SMAD binding element (SBE), an optimal binding site <strong>for</strong> SMAD. We are using Smad7<br />

promoter as a model system to elucidate the molecular mechanisms of TGF-ß-inducible gene<br />

regulation. We have also identified SMAD interreacting proteins that may regulate SMAD<br />

transcription activity <strong>and</strong> are in the process of characterizing these interacting proteins.<br />

Characterization of SMAD4 as a pancreatic tumor suppressor: SMAD4 is a pancreatic<br />

tumor suppressor. More than 50% of pancreatic cancers bear mutations in the SMAD4 locus<br />

(~30% homozygous deletions <strong>and</strong> ~22-25% mutations with loss of heterozygosity). The<br />

mechanism of how SMAD4 functions as a pancreatic tumor suppressor is not completely<br />

defined. Our work is directed towards addressing this important issue.<br />

__________________________________________________________________________________<br />

<strong>Center</strong> <strong>for</strong> <strong>Advanced</strong> <strong>Biotechnology</strong> <strong>and</strong> <strong>Medicine</strong><br />

13


Publications:<br />

__________________________________________________________________________________<br />

<strong>Center</strong> <strong>for</strong> <strong>Advanced</strong> <strong>Biotechnology</strong> <strong>and</strong> <strong>Medicine</strong><br />

Fang Liu<br />

Denissova, N. G., Pouponnot, C., Long, J., He, D. <strong>and</strong> Liu, F. (2000). Trans<strong>for</strong>ming growth<br />

factor ß-inducible independent binding of SMAD to the Smad7 promoter.<br />

Proc. Natl. Acad. Sci. USA, 12:6397-6402.<br />

Liu, F. (<strong>2001</strong>). SMAD4/DPC4 <strong>and</strong> pancreatic cancer survival. Clin Cancer Res. 7:3853-3856.<br />

14


Peter Lobel, Ph.D.<br />

Resident Faculty Member, CABM; Professor, UMDNJ-Robert Wood Johnson Medical<br />

School, Department of Pharmacology<br />

Protein Targeting Laboratory<br />

Dr. Peter Lobel trained at Columbia University <strong>and</strong> Washington University in St. Louis <strong>and</strong> joined CABM in 1989. He is currently conducting<br />

research on lysosomes <strong>and</strong> associated human hereditary metabolic diseases. This work recently led to the identification of a disease gene that<br />

causes a fatal childhood neurodegenerative disorder. At CABM he was the first faculty member of UMDNJ to be named a Searle Scholar, a<br />

prestigious award given to newly appointed faculty members who show outst<strong>and</strong>ing promise in biomedical research.<br />

Our laboratory has developed new methods <strong>for</strong> disease discovery <strong>and</strong> identified the molecular<br />

bases <strong>for</strong> three fatal neurodegenerative disorders based on our research on lysosomal enzyme<br />

targeting. Lysosomes are membrane-bound, acidic organelles that are found in all eukaryotic<br />

cells. They contain a variety of different proteases, glycosidases, lipases, phosphatases, nucleases<br />

<strong>and</strong> other hydrolytic enzymes, most of which are delivered to the lysosome by the mannose 6phosphate<br />

targeting system. In this pathway, lysosomal enzymes are recognized as different from<br />

other glycoproteins <strong>and</strong> are selectively phosphorylated on mannose residues. The mannose 6phosphate<br />

serves as a recognition marker that allows the enzymes to bind mannose 6-phosphate<br />

receptor which ferry the lysosomal enzyme to the lysosome. In the lysosome, the enzymes<br />

function in concert to break down complex biological macromolecules into simple components.<br />

The importance of these enzymes is underscored by the identification of over thirty lysosomal<br />

storage disorders (e.g., Tay Sach's disease) where loss of a single lysosomal enzyme leads to<br />

severe health problems including neurodegeneration, progressive mental retardation, <strong>and</strong> early<br />

death. Our approach to identify the molecular basis <strong>for</strong> unsolved lysosomal storage disorders is<br />

based on our ability to use mannose 6-phosphate receptor derivatives to visualize <strong>and</strong> purify<br />

mannose 6-phosphate containing lysosomal enzymes. For instance, we can fractionate proteins in<br />

normal <strong>and</strong> disease specimens by 2-dimensional gel eletecrophoresis <strong>and</strong> then, in a manner<br />

analogous to Western blotting, use a radiolabeled mannose 6-phosphate receptor derivative to<br />

selectively visualize phosphorylated lysosomal enzymes. This allows us to compare the spectrum<br />

of lysosomal enzymes present in normal <strong>and</strong> disease specimens. If the disease specimen lacks a<br />

given lysosomal protein, this may be responsible <strong>for</strong> disease. To investigate this, we purify <strong>and</strong><br />

sequence the normal protein, clone the corresponding gene, <strong>and</strong> examine patients <strong>for</strong> mutations<br />

associated with disease. In this manner, we found that a fatal childhood neurodegenerative disease<br />

called LINCL (late infantile neuronal ceroid lipofuscinosis) is caused by mutations in a gene<br />

encoding a previously undiscovered lysosomal protease.<br />

After we identified the gene <strong>and</strong> determined the function of corresponding protein, we developed<br />

rapid biochemical <strong>and</strong> DNA-based assays <strong>for</strong> definitive pre-<strong>and</strong> postnatal diagnosis <strong>and</strong> carrier<br />

screening. This allows <strong>for</strong> genetic counseling to prevent further occurrence of the disease.<br />

However, in the absence of universal carrier testing, new cases will continue to arise so it is<br />

important to develop effective therapies that can halt <strong>and</strong> reverse disease progression. To this end,<br />

__________________________________________________________________________________<br />

<strong>Center</strong> <strong>for</strong> <strong>Advanced</strong> <strong>Biotechnology</strong> <strong>and</strong> <strong>Medicine</strong><br />

15


__________________________________________________________________<br />

Peter Lobel<br />

we have produced recombinant enzyme in a <strong>for</strong>m that can be taken up by affected cells in culture<br />

to correct the primary defect. We are also working to develop a LINCL mouse model that should<br />

allow detailed studies of disease pathophysiology <strong>and</strong> evaluation of potential therapeutics<br />

strategies.<br />

Another research program in the laboratory is to identify the spectrum of lysosomal enzymes<br />

encoded by the human genome. This research is particularly timely given the current ef<strong>for</strong>t<br />

towards determining the complete sequence of the human genome. Our approach is to purify<br />

mannose 6-phosphorylated proteins <strong>and</strong> then analyze each protein by peptide mapping, mass<br />

spectrometry, <strong>and</strong> chemical sequencing. This in<strong>for</strong>mation is used to search sequence databases to<br />

determine if a given protein corresponds to a known lysosomal enzyme or if it represents a<br />

previously unidentified species. We recently used this approach to determine the molecular basis<br />

<strong>for</strong> Niemann Pick type C2 disease, a fatal cholesterol storage disorder. In addition to their roles in<br />

human inherited diseases, alterations in the lysosomal system have been implicated in a variety of<br />

disease processes such as tumor invasion <strong>and</strong> metastasis in cancer, tissue destruction in arthritis,<br />

<strong>and</strong> early changes associated with Alzheimer’s disease. Once we develop the tools to visualize<br />

<strong>and</strong> characterize the players, our ultimate goal will be to underst<strong>and</strong> the role that lysosomal<br />

proteins play in these widespread pathological processes.<br />

Publications:<br />

Naureckiene, S., Sleat, D.E., Lackl<strong>and</strong>, H., Fensom, A., Vanier, M.T., Wattiaux, R., Jadot, M. <strong>and</strong><br />

Lobel, P. (2000). Indentification of HE1 as the second gene of Niemann-Pick C disease. Science<br />

290:2227-2229.<br />

Sohar, I., Lin, L. <strong>and</strong> Lobel, P. (2000). Enzyme-based diagnosis of classical late infantile<br />

neuronal ceroid lipofuscinosis: comparison of tripeptidyl peptidase I <strong>and</strong> pepstatin-insensitive<br />

protease assays. Clin. Chem. 46:1005-1008.<br />

Lin, L., Sohar, I., Lackl<strong>and</strong>, H. <strong>and</strong> Lobel, P. (<strong>2001</strong>). The human CLN2 protein/tripeptidylpeptidase<br />

I is a serine protease that autoactivates at acidic pH. J. Biol. Chem. 276: 2249-2255.<br />

Sleat, D.E., Sohar, I., Gin, R.M. <strong>and</strong> Lobel, P. (<strong>2001</strong>). Aminoglycoside-mediated suppression of<br />

nonsense mutations in late infantile neuronal ceroid lipofuscinosis. Eur. J. Paed. Neurol. 5A:45-<br />

50.<br />

Lin, L. <strong>and</strong> Lobel, P. (<strong>2001</strong>). Production <strong>and</strong> characterization of recombinant human CLN2<br />

protein <strong>for</strong> enzyme replacement therapy in late infantile neuronal ceroid lipofuscinosis. Biochem.<br />

J. 357:49-55.<br />

Lin, L. <strong>and</strong> P. Lobel. (<strong>2001</strong>). Expression <strong>and</strong> analysis of CLN2 variants in CHO cells: Q100r<br />

represents a polymorphism, <strong>and</strong> G389E <strong>and</strong> R447H represent loss-of-function mutations. Hum.<br />

Mutat. 18:165.<br />

__________________________________________________________________________________<br />

<strong>Center</strong> <strong>for</strong> <strong>Advanced</strong> <strong>Biotechnology</strong> <strong>and</strong> <strong>Medicine</strong><br />

16


James H. Millonig, Ph.D.<br />

Resident Faculty Member, CABM; Assistant Professor, UMDNJ-Robert Wood Johnson<br />

Medical School, Department of Neuroscience <strong>and</strong> Cell Biology<br />

Developmental Neurogenetics Laboratory<br />

Dr. James H. Millonig came to CABM in September 1999 from the Rockefeller University where he was a postdoctoral fellow in the laboratory of<br />

Dr. Mary E. Hatten. His postdoctoral research combined neurobiology <strong>and</strong> mouse genetics to characterize <strong>and</strong> clone the dreher mouse locus. He<br />

did his doctoral research at Princeton University with Dr. Shirley M. Tilghman. Dr. Millonig is a recipient of the March of Dimes Basil O’Connor<br />

Starter Research Award <strong>and</strong> grants from the Whitehall Foundation, the National Ataxia Foundation <strong>and</strong> N.J. Governor’s Council on Autism.<br />

My laboratory studies the development of the dorsal CNS by combining mouse molecular<br />

genetics <strong>and</strong> neuroanatomical approaches. The dorsal CNS includes the cerebrum, hippocampus,<br />

cerebellum <strong>and</strong> sensory neurons of the spinal cord. These structures play an important role in<br />

cognition, learning, memory, motor movement <strong>and</strong> sensation. During development, millions of<br />

different types of neurons are generated which are involved in controlling these neural functions.<br />

Our ultimate goal is to underst<strong>and</strong> the molecular pathways that lead to the generation of different<br />

types of dorsal neurons in two simple dorsal anatomical structures, the spinal cord <strong>and</strong><br />

cerebellum.<br />

The entire dorsal CNS arises from the lateral neural plate very early in development. At this<br />

stage, the epidermal ectoderm is adjacent to the lateral neural plate <strong>and</strong> dorsalizes it through the<br />

action of secreted proteins. This results in the delamination of the neuroectoderm from the<br />

epidermis <strong>and</strong> the eventual folding of the neural plate into a neural tube. Failure of this to occur<br />

results in spina bifida, a common human disorder that has an incidence of 1:1000.<br />

Once the neural tube <strong>for</strong>ms, another transitory signaling center called the roof plate that is situated<br />

on the dorsal midline is believed to instruct neighboring cells to a particular lineage again through<br />

the action of secreted factors. As a postdoctoral fellow at The Rockefeller University, we<br />

identified the first mouse mutation to lack a roof plate. It is a spontaneous mouse mutation called<br />

dreher (dr). Our positional cloning of the locus determined that a gene called Lmx1a is<br />

responsible <strong>for</strong> the loss of roof plate in dr/dr embryos (Millonig et al, 2000).<br />

Expression analysis of Lmx1a indicated that it was expressed just in the roof plate. Dr can then<br />

be used as a mouse model to determine the role of the roof plate during dorsal CNS development.<br />

Any perturbation to dorsal neurons in dr/dr embryos must be due to loss of roof plate signaling.<br />

Our phenotypic analysis has determined that the roof plate is necessary to generate the normal<br />

complement of dorsal neurons <strong>and</strong> is required even after their commitment <strong>for</strong> their initial steps of<br />

differentiation, neuronal migration <strong>and</strong> axon extension (Millonig, unpublished results).<br />

We are now investigating the possible cellular mechanisms that result in the decrease of dorsal<br />

neurons in dr/dr embryos. There are at least three possibilities: decrease in the rate of proliferation<br />

of progenitors, increased cell death or progenitors spend a longer time in the cell<br />

__________________________________________________________________________________<br />

<strong>Center</strong> <strong>for</strong> <strong>Advanced</strong> <strong>Biotechnology</strong> <strong>and</strong> <strong>Medicine</strong><br />

17


__________________________________________________________________________________<br />

<strong>Center</strong> <strong>for</strong> <strong>Advanced</strong> <strong>Biotechnology</strong> <strong>and</strong> <strong>Medicine</strong><br />

James H. Millonig<br />

cycle. Preliminary data suggests that the last could be true, indicating that the roof plate<br />

coordinates the withdrawal of cells from the cell cycle (Tanya Borusk, unpublished results).<br />

Splotch (Sp), another mouse mutant, exhibits a similar cell cycle withdrawal phenotype, providing<br />

us with two mutations that together should lead to insights into the molecular mechanisms that<br />

control dorsal neuronal generation.<br />

Another spontaneous mouse mutation, vacuolated lens (vl), appears to have the opposite<br />

phenotype to dr, too much roof plate. Vl/vl embryos have an exp<strong>and</strong>ed dorsal midline <strong>and</strong> exhibit<br />

phenotypes consistent with too much roof plate, such as an increase in the number of dorsal<br />

neurons <strong>and</strong> overgrowth of the dorsal vertebrae that lie above the roof plate. In dr, this part of the<br />

vertebrae is missing. Vl maps near the dr locus but sequencing of Lmx1a from vl/vl embryos<br />

indicates that Lmx1a is not responsible <strong>for</strong> the mutation (Jigar Desai, unpublished results). We<br />

are now in the process of positionally cloning the locus.<br />

We have also been focusing on identifying genes that are downstream of roof plate signaling. To<br />

this end, we have identified transcription factors that are expressed in nested domains in the<br />

developing cerebellum that appear to be restricted to particular cellular populations (Max<br />

Tischfield, unpublished results). Phenotypic analysis of dr embryos <strong>and</strong> in vitro explants are<br />

being used to determine if roof plate signaling is necessary <strong>and</strong> sufficient <strong>for</strong> the expression of<br />

these dorsal markers. The function of these genes is being tested through a combination of mouse<br />

genetics <strong>and</strong> the <strong>for</strong>ced misexpression in the cerebellum during chick embryogenesis. Through<br />

these different approaches the pathways involved in dorsal CNS development will be ascertained.<br />

Publications:<br />

Millonig, J.H., Millen, K.J <strong>and</strong> Hatten, M.E. (2000) The Mouse Dreher gene (Lmx1a) Controls<br />

Formation of the Roof Plate Formation in Vertebrate CNS. Nature. 403:764-768.<br />

18


Michael M. Shen, Ph.D.<br />

Resident Faculty Member, CABM; Associate Professor, UMDNJ-Robert Wood Johnson<br />

Medical School, Department of Pediatrics<br />

Mammalian Embryogenesis Laboratory<br />

Dr. Michael Shen completed his doctoral work in genetics under Dr. Jonathan Hodgkin at the MRC laboratory of Molecular Biology in<br />

Cambridge, Engl<strong>and</strong>. He then per<strong>for</strong>med postdoctoral work at Harvard Medical School under Dr. Philip Leder be<strong>for</strong>e joining CABM in 1994.<br />

He is a <strong>for</strong>mer recipient of a Leukemia Society of America Special Fellowship, a Howard Hughes Medical Institute Post-doctoral Fellowship, a<br />

Jane Coffin Childs Post-doctoral Fellowship, <strong>and</strong> a National Science Foundation Graduate Fellowship. Dr. Shen is a member of the NIH Study<br />

Section on Cell Development <strong>and</strong> Function <strong>and</strong> of the National Cancer Institute Steering Committee <strong>for</strong> the Mouse Models of Human Cancer<br />

Consortium.<br />

The research in our laboratory is directed towards underst<strong>and</strong>ing the molecular mechanisms<br />

involved in (i) <strong>for</strong>mation of the embryonic anterior-posterior <strong>and</strong> left-right axes in the mouse<br />

embryo, (ii) signal transduction by EGF-CFC proteins <strong>and</strong> the TGF-beta-related factor Nodal,<br />

<strong>and</strong> (iii) prostate development <strong>and</strong> carcinogenesis.<br />

To address the first two areas, we are pursuing studies of the recently identified EGF-CFC gene<br />

family, whose members encode novel extracellular proteins. We have used gene targeting<br />

approaches in mice to demonstrate that EGF-CFC genes play essential roles in embryonic axis<br />

<strong>for</strong>mation, namely that Cripto is required <strong>for</strong> correct orientation of the anterior-posterior (A-P)<br />

axis, while Cryptic is necessary <strong>for</strong> determination of the left-right (L-R) axis. Thus, targeted<br />

disruption <strong>for</strong> Cripto results in a 90 degree mis-orientation of the A-P axis, indicating that Cripto<br />

is essential <strong>for</strong> an axial rotation that converts a proximal-distal asymmetry into an orthogonal A-<br />

P axis during pre-gastrulation stages. In contrast, null mutation <strong>for</strong> Cryptic results in L-R<br />

laterality defects, including r<strong>and</strong>omized abdominal situs, pulmonary right isomerism, <strong>and</strong> severe<br />

cardiac defects, as well as lack of expression of regulatory genes in the L-R pathway, suggesting<br />

that Cryptic is essential <strong>for</strong> a key step in L-R axis specification. Our results also support an<br />

interaction of EGF-CFC proteins with the TGF-beta-related factor Nodal <strong>and</strong> with activin<br />

receptors, which we are currently examining using biochemical <strong>and</strong> cell culture approaches.<br />

In another major area of interest, we are investigating prostate development <strong>and</strong> cancer (in<br />

collaboration with Dr. Cory Abate-Shen's lab). We are focusing on the role of the Nkx3.1<br />

homeobox gene, which displays restricted expression in the embryonic <strong>and</strong> adult mouse prostate.<br />

We have demonstrated that homozygous Nkx3.1-/- mice created by gene targeting display<br />

progressive defects in prostate histology with advancing age, resulting in epithelial hyperplasia<br />

<strong>and</strong> dysplasia. These lesions in aged mutant mice resemble human prostatic intraepithelial<br />

neoplasia (PIN), which is believed to represent the precursor to prostate carcinoma. These<br />

observations are noteworthy given the mapping of human NKX3.1 to the minimal deleted<br />

interval of chromosome 8p21, which is lost as an early <strong>and</strong> frequent event in human prostate<br />

cancer. Our findings show that Nkx3.1 plays a critical role in the normal morphogenesis <strong>and</strong><br />

function of the prostate, <strong>and</strong> suggest that it may represent a prostate-specific tumor suppressor<br />

gene.<br />

__________________________________________________________________________________<br />

<strong>Center</strong> <strong>for</strong> <strong>Advanced</strong> <strong>Biotechnology</strong> <strong>and</strong> <strong>Medicine</strong><br />

19


Publications:<br />

__________________________________________________________________________________<br />

<strong>Center</strong> <strong>for</strong> <strong>Advanced</strong> <strong>Biotechnology</strong> <strong>and</strong> <strong>Medicine</strong><br />

Michael M. Shen<br />

Yan, Y.-T., Stein, S. M., Ding, J., Shen, M. M. <strong>and</strong> Abate-Shen, C. (2000). A novel PF/PN<br />

motif inhibits nuclear localization <strong>and</strong> DNA binding activity of the ESX1 homeoprotein. Mol.<br />

Cell. Biol. 20:661-671.<br />

Mudgett, J. S., Ding, J., Guh-Siesel, L., Chartrain, N. A., Yang, L., Gopal, S. <strong>and</strong> Shen, M. M.<br />

(2000). Essential role <strong>for</strong> p38a mitogen-activated protein kinase in placental angiogenesis. Proc.<br />

Natl. Acad. Sci. USA 97:10454-10459.<br />

Bam<strong>for</strong>d, R. N., Roessler, E., Burdine, R. D., Saplakoglu, U., de la Cruz, J., Splitt, M.,<br />

Goodship, J. A., Towbin, J., Bowers, P., Ferrero, G. B., Marino, B., Schier, A. F., Shen, M. M.,<br />

Muenke, M. <strong>and</strong> Casey, B. (2000) . Loss-of-function mutations in the EGF-CFC gene Cfc1 are<br />

associated with human left-right laterality defects. Nature Genet. 26:365-369.<br />

Hu, G., Lee, H., Price, S. M., Shen, M. M. <strong>and</strong> Abate-Shen, C. (<strong>2001</strong>) . Msx homeobox genes<br />

inhibit differentiation through up-regulation of Cyclin D1. Development 128:2373-2384.<br />

Kimura, C., Shen, M. M., Takeda, N., Aizawa, S. <strong>and</strong> Matsuo, I. (<strong>2001</strong>) . Complementary<br />

functions of Otx2 <strong>and</strong> Cripto in initial patterning of mouse epiblast. Dev. Biol. 128:2373-384.<br />

Review articles (peer-reviewed):<br />

Schier, A. F. <strong>and</strong> Shen, M. M. (2000) . Nodal signalling in vertebrate development. Nature<br />

403:385-389.<br />

Shen, M. M. <strong>and</strong> Schier, A. F. (2000). The EGF-CFC gene family in vertebrate development.<br />

Trends Genet. 16:303-309.<br />

Abate-Shen, C. <strong>and</strong> Shen, M. M. (2000). Molecular genetics of prostate cancer. Genes Dev.<br />

14:2410-2434.<br />

Book chapters/technical articles/reviews:<br />

Shen, M. M. (<strong>2001</strong>). Identification of differentially expressed genes in mouse development<br />

using differential display <strong>and</strong> in situ hybridization. Methods 24:15-27.<br />

Cunha, G. R., Donjacour, A. A., Hayward, S. W., Thomson, A. A., Marker, P. C., Abate-Shen,<br />

C., Shen, M. M. <strong>and</strong> Dahiya, R. (2002). Cellular <strong>and</strong> molecular biology of prostatic<br />

development. In: Prostate Cancer: Principles <strong>and</strong> Practice. Eds. Kantoff, P. W., Carroll, P.,<br />

D'Amico, A. V. (Lippincott, Williams <strong>and</strong> Wilkins, Philadelphia), pp. 16-28.<br />

20


Eileen White, Ph.D.<br />

Associate Investigator, Howard Hughes Medical Institute; Resident Faculty Member,<br />

CABM; Professor, <strong>Rutgers</strong>, Department of Molecular Biology <strong>and</strong> Biochemistry<br />

Viral Trans<strong>for</strong>mation Laboratory<br />

Dr. Eileen White transferred her research program to CABM in July 1990 from Cold Spring Harbor Laboratory where she was a staff<br />

investigator. She conducted postdoctoral research at the Cold Spring Harbor laboratory as a Damon Runyon – Walter Winchell Fellow, working<br />

with Dr. Bruce Stillman. Her work is currently funded by a MERIT Award from NIH <strong>and</strong> Howard Hughes Medical Institute. Dr. White is a<br />

member of the National Cancer Institute Board of Scientific Counselors.<br />

The goal of our research program is to examine viral oncogenes <strong>and</strong> mechanisms of mammalian<br />

cell growth. Specifically, the White lab is working to determine the mechanisms by which<br />

oncogenes of the DNA tumor virus adenovirus stimulate cell proliferation <strong>and</strong> inhibit cell death<br />

which leads to malignant trans<strong>for</strong>mation.<br />

We study the mechanisms by which human DNA tumor viruses deregulate cell cycle <strong>and</strong><br />

apoptosis (programmed cell death) control in the development of cancer <strong>and</strong> during<br />

productive viral infection. Underst<strong>and</strong>ing pathways that control cell proliferation <strong>and</strong> cell death<br />

will lead to better treatments <strong>for</strong> cancer <strong>and</strong> viral infections.<br />

Description of Research<br />

The DNA tumor virus adenovirus infects human cells, recruits them into a proliferative state,<br />

<strong>and</strong> borrows elements of the host cell transcription, translation, <strong>and</strong> DNA replication machinery<br />

to reproduce viral proteins <strong>and</strong> DNA. In rodent cells which are semipermissive <strong>for</strong> adenovirus<br />

infection, cell growth is deregulated but virus replication is ineffective. As the viral infection<br />

does not progress to completion, the deregulation of cell growth control produces trans<strong>for</strong>mation.<br />

The viral genes required <strong>for</strong> oncogenic trans<strong>for</strong>mation are the E1A <strong>and</strong> E1B oncogenes.<br />

Underst<strong>and</strong>ing how the products of these viral oncogenes alter cell growth control pathways is<br />

important <strong>for</strong> establishing how cancer develops.<br />

Regulation of programmed cell death (apoptosis) by E1A <strong>and</strong> E1B is necessary <strong>for</strong> sustaining a<br />

productive infection in human cells <strong>and</strong> is an integral part of the trans<strong>for</strong>mation process in rodent<br />

cells. The E1A proteins are responsible <strong>for</strong> initiating a proliferative response, <strong>and</strong> an indirect<br />

consequence of this required function of E1A is the induction of apoptosis. The E1B gene<br />

encodes overlapping, redundant functions to suppress apoptosis, the 19K <strong>and</strong> 55K proteins.<br />

Thus, E1A expression <strong>and</strong> subsequent growth deregulation can occur unimpeded by cell death in<br />

the presence of E1B expression. Without inhibition of apoptosis by E1B, trans<strong>for</strong>mation of<br />

rodent cells is rare, <strong>and</strong> premature death of the host cell impairs virus yield in productively<br />

infected human cells. The human bcl-2 proto-oncogene, which is known to inhibit apoptosis in<br />

other systems, will also suppress apoptosis induced by E1A expression <strong>and</strong> cooperate with E1A<br />

to trans<strong>for</strong>m primary rodent cells. Thus, inhibition of apoptosis is an important step in the<br />

progression of the trans<strong>for</strong>med state.<br />

__________________________________________________________________________________<br />

<strong>Center</strong> <strong>for</strong> <strong>Advanced</strong> <strong>Biotechnology</strong> <strong>and</strong> <strong>Medicine</strong><br />

21


__________________________________________________________________________________<br />

<strong>Center</strong> <strong>for</strong> <strong>Advanced</strong> <strong>Biotechnology</strong> <strong>and</strong> <strong>Medicine</strong><br />

Eileen White<br />

There is substantial evidence that the product of the p53 tumor suppressor gene mediates the<br />

induction of apoptosis by E1A <strong>and</strong> that the E1B <strong>and</strong> bcl-2 gene products function by disabling<br />

p53. Expression of the E1B 19K or bcl-2 genes will prevent p53-dependent apoptosis by<br />

modifying p53 function indirectly, whereas the E1B 55K protein acts by a direct interaction with<br />

p53. bcl-2 may be the cellular equivalent of the E1B 19K gene, with its oncogenic <strong>and</strong> antiapoptotic<br />

activity, in all or in part, attributed to bypassing the function of p53.<br />

One of the most important, recently identified events in humans <strong>and</strong> other life <strong>for</strong>ms is<br />

programmed cell death or apoptosis. Failure of this process can result in excessive cell growth<br />

as in cancer or abnormal development; inappropriate induction of programmed cell death is<br />

responsible <strong>for</strong> many other diseases including neurodegenerative disorders. By studying the<br />

oncogenes of the DNA tumor virus, adenovirus, we have determined that malignant<br />

trans<strong>for</strong>mation requires (i) stimulation of proliferation by viral E1A proteins (ii) inhibition by<br />

E1B proteins of the apoptosis that results from the inappropriate stimulation, (iii) inhibition by<br />

E1B proteins of the apoptosis that results from the inappropriate E1A induction of apoptosis is<br />

mediated through the cellular p53 tumor suppressor, <strong>and</strong> E1B products prevent p53-dependent<br />

cell death both directly (55 Kd protein) <strong>and</strong> indirectly (19 Kd protein).<br />

Results<br />

Our recent studies have demonstrated that E1B 19 Kd protein is the equivalent of the cellular<br />

product of the bcl-2 gene. Both have oncogenic <strong>and</strong> anti-apoptotic activity, all or in part,<br />

attributed to bypassing the function of the p53 tumor suppressor. We have found that the E1B 19<br />

Kd protein blocks apoptosis by interacting with <strong>and</strong> inhibiting the p53-inducible <strong>and</strong> death<br />

promoting Bax protein. An E1B 19 Kd interacting human protein called Nbk/Bik has been<br />

identified, shown to contain a BH3 domain, <strong>and</strong> demonstrated to induce apoptosis.<br />

E1A promotes apoptosis by interacting with <strong>and</strong> inhibiting negative regulators of cell cycle<br />

control. Binding of E1A to, <strong>and</strong> inhibition of, the transcriptional coadaptor p300 promotes<br />

accumulation of the p53 tumor suppressor protein which induces apoptosis. By inhibiting p300,<br />

E1A prevents the transcriptional activation of mdm-2, the product of which interacts with <strong>and</strong><br />

promotes the degradation of p53. Thus the E1A-p300 interaction disables the negative feedback<br />

loop to control p53 levels, which left unrestrained, causes apoptosis rather than growth arrest.<br />

The E1B 19K protein functions analogously to Bcl-2 to inhibit apoptosis by E1A, p53, <strong>and</strong><br />

multiple other stimuli. The E1B 19K protein functions by at least two independent mechanisms<br />

to inhibit apoptosis. First, the E1B 19K protein binds to the pro-apoptotic Bax protein to prevent<br />

loss of mitochondrial membrane potential, caspase activation, <strong>and</strong> apoptosis. Second, the E1B<br />

19K protein inhibits caspase interaction by interfering with the function of adaptor molecules<br />

such as FADD <strong>and</strong> Ced-4 that interact with <strong>and</strong> activate caspases. By inhibiting FADDdependent<br />

activation of the caspase FLICE, the E1B 19K protein can disable both the TNF-α<br />

<strong>and</strong> Fas-mediated death signaling pathways which play an important role in immune surveillance<br />

against virus infection <strong>and</strong> cancer. The E1B 19K protein binds to Ced-4, <strong>and</strong> presumably<br />

mammalian Ced-4 homologues, <strong>and</strong> thereby prevents caspase activation. Thus, the study of the<br />

22


__________________________________________________________________________________<br />

<strong>Center</strong> <strong>for</strong> <strong>Advanced</strong> <strong>Biotechnology</strong> <strong>and</strong> <strong>Medicine</strong><br />

Eileen White<br />

mechanism of regulation of apoptosis by the adenovirus trans<strong>for</strong>ming proteins has revealed<br />

important regulatory steps in death signaling pathways.<br />

Our findings have been reported in several original research reports <strong>and</strong> review articles <strong>and</strong> at<br />

numerous international symposia.<br />

Future Directions<br />

The current aim is to determine the molecular basis by which apoptosis is regulated by E1A, p53,<br />

E1B <strong>and</strong> Bcl-2. Establishing how the trans<strong>for</strong>ming genes of DNA tumor viruses subvert p53<br />

function will provide insight into the cause <strong>and</strong> prevention of cancer <strong>and</strong> lead to new strategies in<br />

anticancer therapy.<br />

Publications:<br />

Perez, D. <strong>and</strong> White, E. (2000). TNF-α Signals apoptosis through a Bid-dependent<br />

con<strong>for</strong>mational change in Bax which is inhibited by E1B 19K. Mol. Cell 6:53-63.<br />

Thomas, A., Giesler, T. <strong>and</strong> White, E. (2000). p53 mediates Bcl-2 phosphorylation <strong>and</strong><br />

apoptosis via activation of the Cdc42/JNK1 pathway. Oncogene 19:5259-5269.<br />

Degenhardt, K., Perez, D. <strong>and</strong> White, E. (2000). Pathways used by adenovirus E1B 19K to<br />

inhibit apoptosis in programmed cell death in animals <strong>and</strong> plants. Editors: J. Bryant, S. Hughes,<br />

J. Garl<strong>and</strong>; Bios Scientific Publishers LTD, 241-251.<br />

Sundararajan, R., Cuconati, A., Nelson, D. <strong>and</strong> White, E. (<strong>2001</strong>). Tumor Necrosis Factor-α<br />

induces Bax-Bak interaction <strong>and</strong> apoptosis which is inhibited by adenovirus E1B 19K. J. Biol.<br />

Chem. 276:45120-45127.<br />

White, E. (<strong>2001</strong>). Regulation of the cell cycle <strong>and</strong> apoptosis by the oncogenes of adenovirus.<br />

Oncogene 20:7836-7846.<br />

Sundararajan, R. <strong>and</strong> White, E. (<strong>2001</strong>). E1B 19K Blocks Bax oligomerization <strong>and</strong> tumor<br />

necrosis factor alpha-mediated apoptosis. J. Virol. 75: 7506-7516.<br />

Shen, Y. <strong>and</strong> White, E. (<strong>2001</strong>). p53-Dependent apoptosis pathways. Adv. Cancer Res. 82: 55-<br />

84.<br />

Henry, H., Thomas, A., Shen, Y. <strong>and</strong> White, E. (<strong>2001</strong>). Regulation of the mitochondrial<br />

checkpoint in p53-mediated apoptosis confers resistance to cell death. Oncogene 21:748-760.<br />

23


Mengqing Xiang, Ph.D.<br />

Resident Faculty Member, CABM; Assistant Professor, UMDNJ-Robert Wood Johnson<br />

Medical School, Department of Pediatrics<br />

Molecular Neurodevelopment Laboratory<br />

Dr. Mengqing Xiang came to CABM in September 1996 from the Johns Hopkins University School of <strong>Medicine</strong> where he conducted<br />

postdoctoral studies with Dr. Jeremy Nathans. He earned his Ph.D. at the University of Texas M.D. Anderson Cancer <strong>Center</strong> <strong>and</strong> has received a<br />

number of honors including a China-U.S. Government Graduate Study Fellowship, a Howard Hughes Medical Institute Postdoctoral Fellowship,<br />

a Basil O’Connor Starter Scholar Research Award, <strong>and</strong> a Sinsheimer Scholar Award. His work is currently supported by NIH <strong>and</strong> the<br />

Alex<strong>and</strong>rine <strong>and</strong> Alex<strong>and</strong>er L. Sinsheimer Fund.<br />

Our laboratory investigates the molecular mechanisms that govern the determination <strong>and</strong><br />

differentiation of highly specialized sensory cells <strong>and</strong> neurons. We employ molecular genetic<br />

approaches to identify <strong>and</strong> study transcription factors that are required <strong>for</strong> programming<br />

development of the retina, inner ear, <strong>and</strong> somatosensory ganglia. A major focus of our work is to<br />

develop animal models to study roles of transcription factor genes in normal sensorineural<br />

development, as well as to elucidate how mutations in these genes cause sensorineural disorders<br />

such as blindness <strong>and</strong> deafness. Current projects in my laboratory are centered on studies of<br />

biological roles of two small families of transcription factor genes – Brn3 POU domain genes<br />

<strong>and</strong> Barhl homeobox genes. During the past year we made significant progress in three projects.<br />

First, our studies have shown that Brn3a plays a key role in the control of soma size, target field<br />

innervation, <strong>and</strong> axon pathfinding of inner ear sensory neurons. Second, our work has identified<br />

an important transcriptional cascade involved in the control of retinal ganglion cell development.<br />

Finally, we have generated knockout mice to demonstrate that Barhl1 is specifically required <strong>for</strong><br />

the maintenance of cochlear outer hair cells.<br />

Publications:<br />

Liu, W., Khare, S. L., Liang, X., Peters, M. A., Liu, X., Cepko, C. L. <strong>and</strong> Xiang, M. (2000). All<br />

Brn3 genes can promote retinal ganglion cell differentiation in the chick. Development<br />

127:3237-3247.<br />

Liu, W., Mo, Z. <strong>and</strong> Xiang, M. (<strong>2001</strong>). The Ath5 proneural genes function upstream of Brn3<br />

POU domain transcription factor genes to promote retinal ganglion cell development. Proc. Natl.<br />

Acad. Sci. USA. 98:1649-1654.<br />

Huang, E. J., Liu, W., Fritzsch, B., Bianchi, L. M., Reichardt, L. F. <strong>and</strong> Xiang, M. (<strong>2001</strong>).<br />

Brn3a is a transcriptional regulator of soma size, target field innervation, <strong>and</strong> axon pathfinding of<br />

inner ear sensory neurons. Development 128:2421-2432.<br />

__________________________________________________________________________________<br />

<strong>Center</strong> <strong>for</strong> <strong>Advanced</strong> <strong>Biotechnology</strong> <strong>and</strong> <strong>Medicine</strong><br />

24


Laboratory Faculty Director<br />

Structural Biology<br />

Biomolecular Crystallography Edward Arnold 26<br />

Protein NMR Spectroscopy Gaetano Montelione 31<br />

Protein Microchemistry Stanley Stein 33<br />

Protein Crystallography Ann Stock 34<br />

__________________________________________________________________________________<br />

<strong>Center</strong> <strong>for</strong> <strong>Advanced</strong> <strong>Biotechnology</strong> <strong>and</strong> <strong>Medicine</strong><br />

25


Edward Arnold, Ph.D.<br />

Resident Faculty Member, CABM; Professor, <strong>Rutgers</strong>, Department of Chemistry <strong>and</strong><br />

Chemical Biology, Adjunct Professor, UMDNJ-RWJMS, Department of Molecular<br />

Genetics <strong>and</strong> Microbiology<br />

Biomolecular Crystallography Laboratory<br />

Dr. Arnold obtained his Ph.D. in organic chemistry with Professor Jon Clardy at Cornell University in 1982. From 1982 to 1987, he did<br />

postdoctoral work with Professor Michael G. Rossmann at Purdue University <strong>and</strong> was a central member of the team that solved the structure of a<br />

human common cold virus by X-ray crystallography. Among the awards <strong>and</strong> fellowships Arnold has received are a National Science Foundation<br />

Predoctoral Fellowship, Damon Runyon–Walter Winchell <strong>and</strong> National Institutes of Health Postdoctoral Fellowships, an Alfred P. Sloan<br />

Research Fellowship, a Johnson <strong>and</strong> Johnson Focused Giving Award, <strong>and</strong> a Board of Trustees Award <strong>for</strong> Excellence in Research at <strong>Rutgers</strong>. Dr.<br />

Arnold is the director of a multi-center NIH Program Project. He received an NIH MERIT Award in 1999 <strong>and</strong> was elected a Fellow of the<br />

American Association <strong>for</strong> the Advancement of Science in <strong>2001</strong>. The laboratory is supported by grants from NIH <strong>and</strong> industrial collaborators, <strong>and</strong><br />

by research fellowships.<br />

Many of the underlying biological <strong>and</strong> chemical processes of life are being detailed at the<br />

molecular level, providing unprecedented opportunities <strong>for</strong> the development of novel approaches<br />

to the cure <strong>and</strong> prevention of human disease. A broad base of advances in chemistry, biology,<br />

<strong>and</strong> medicine has led to an exciting era in which knowledge of the intricate structure of life’s<br />

machinery can help to accelerate the development of new small molecule drugs <strong>and</strong> biomaterials<br />

such as engineered viral vaccines. Drs. Eddy Arnold <strong>and</strong> Gail Ferst<strong>and</strong>ig Arnold <strong>and</strong> their<br />

colleagues are working to develop <strong>and</strong> apply structure-based drug <strong>and</strong> vaccine designs <strong>for</strong> the<br />

treatment <strong>and</strong> prevention of serious human diseases. In pursuit of these goals, their laboratory<br />

takes advantage of cutting-edge research tools, including X-ray crystallography, molecular<br />

biology, virology, protein biochemistry, <strong>and</strong> macromolecular engineering.<br />

The approaches being developed in the Arnold laboratory are applicable to a wide array of<br />

human health problems, ranging from infectious diseases to cancer <strong>and</strong> diseases caused by<br />

hereditary genetic defects. Much of the Arnold lab’s research ef<strong>for</strong>t to date has focused on the<br />

development of drugs <strong>and</strong> vaccines <strong>for</strong> the treatment <strong>and</strong> prevention of AIDS. Examples of the<br />

results of these studies include: 1) collaborative development of drugs <strong>for</strong> the treatment of AIDS,<br />

some of which appear to be more effective than treatments in current use; <strong>and</strong> 2) production of<br />

AIDS vaccine c<strong>and</strong>idates that have elicited protective immune responses against HIV.<br />

Dr. Eddy Arnold <strong>and</strong> coworkers study the structure <strong>and</strong> function of reverse transcriptase (RT), an<br />

essential component of the AIDS virus <strong>and</strong> the target of many of the most widely used anti-AIDS<br />

drugs. Using the powerful techniques of X-ray crystallography, his team has solved the threedimensional<br />

structures of HIV-1 RT in complex with a variety of antiviral drugs <strong>and</strong> small<br />

segments of the HIV genome. These studies have revealed the workings of an intricate <strong>and</strong><br />

fascinating biological machine in atomic detail <strong>and</strong> have yielded numerous novel insights into<br />

polymerase structure-function relationships, detailed mechanisms of drug resistance, <strong>and</strong><br />

structure-based design of RT inhibitors. Synthesis of this in<strong>for</strong>mation has led to the development<br />

of a number of inhibitors that show great promise as potential treatments <strong>for</strong> AIDS.<br />

__________________________________________________________________________________<br />

<strong>Center</strong> <strong>for</strong> <strong>Advanced</strong> <strong>Biotechnology</strong> <strong>and</strong> <strong>Medicine</strong><br />

26


__________________________________________________________________________________<br />

<strong>Center</strong> <strong>for</strong> <strong>Advanced</strong> <strong>Biotechnology</strong> <strong>and</strong> <strong>Medicine</strong><br />

Edward Arnold<br />

Drug development <strong>and</strong> structural studies of a molecule as complex as HIV RT require immense<br />

<strong>and</strong> highly coordinated resources. Dr. Eddy Arnold has been <strong>for</strong>tunate to have highly successful<br />

collaborations with the groups of Dr. Stephen Hughes (NIH NCI, Frederick, MD) <strong>and</strong> Dr. Paul<br />

Janssen (<strong>Center</strong> <strong>for</strong> Molecular Design, Janssen Research Foundation, Belgium), <strong>and</strong> generous<br />

access to synchrotron X-radiation sources (CHESS, APS, <strong>and</strong> BNLS). Hughes <strong>and</strong> his group<br />

have contributed expertise in protein engineering, production, <strong>and</strong> biochemistry at every stage of<br />

the RT project. Janssen <strong>and</strong> his coworkers (including chemists at Janssen Research Foundation,<br />

Springhouse, PA) have synthesized hundreds of molecules from a number of chemical families<br />

in search of optimal drug c<strong>and</strong>idates, eventually leading to the discovery of agents effective<br />

against isolates of HIV containing drug-resistance mutations that can cause the currently<br />

available drugs to fail. Crystallographic work from the Arnold <strong>and</strong> Hughes laboratories has<br />

allowed precise visualization of how potential anti-HIV drug c<strong>and</strong>idates latch onto RT, their<br />

molecular target. Janssen <strong>and</strong> colleagues at the <strong>Center</strong> <strong>for</strong> Molecular Design have used this<br />

structural in<strong>for</strong>mation to guide the design <strong>and</strong> synthesis of new molecules with improved<br />

properties. Following further evaluation against drug-resistant variants, scientists at Tibotec-<br />

Virco (Mechelen, Belgium) have coordinated testing of two of the molecules with highly<br />

promising results in Phase II clinical trials. Some of these anti-HIV compounds are inexpensive<br />

to make, potently <strong>and</strong> broadly effective, non-toxic, <strong>and</strong> simple to administer. These molecules<br />

have the potential to be widely accessible <strong>for</strong> treating AIDS in underdeveloped nations.<br />

Vaccines have proven to be the most effective tools <strong>for</strong> worldwide control of infectious diseases.<br />

Our laboratory’s vaccine development project, jointly directed by both of us, involves<br />

engineering a human common cold virus, rhinovirus, to display immunogenic segments from<br />

more dangerous pathogens <strong>for</strong> the purpose of developing vaccines against these pathogens. This<br />

work involves generating “combinatorial libraries” of chimeric human rhinoviruses using a<br />

technique called r<strong>and</strong>om systematic mutagenesis. Foreign sequences are linked to the HRV<br />

sequences via adapters of r<strong>and</strong>omized sequences <strong>and</strong> lengths, leading to a constellation of<br />

presentations. Large sets of such viruses presenting the <strong>for</strong>eign sequences in many<br />

con<strong>for</strong>mations are generated <strong>and</strong> then selected with appropriate antibodies aimed at the target<br />

pathogen, allowing <strong>for</strong> the isolation of vaccine c<strong>and</strong>idates with the most effectively reconstructed<br />

<strong>for</strong>eign segments. Our combinatorial approach to the vaccine problem is akin to buying many<br />

tickets <strong>for</strong> a lottery: the chances of winning the jackpot are increased by having more tickets.<br />

Chimeric rhinovirus constructs have been made that elicit antibodies (in guinea pigs) capable of<br />

potently neutralizing the AIDS virus in cell culture. Virus libraries incorporating immunogens<br />

from the HIV gp120 <strong>and</strong> gp41 envelope glycoproteins have been constructed. We are also<br />

working collaboratively with Professor John Taylor of the <strong>Rutgers</strong> Chemistry <strong>and</strong> Chemical<br />

Biology Department to probe the immunogenic determinants of an epitope from gp41 using<br />

synthetic peptides. In addition to looking <strong>for</strong> chimeric viruses <strong>and</strong> peptides capable of eliciting<br />

the most potent <strong>and</strong> broad immune responses possible, we are also interested in elucidating the<br />

molecular determinants of immunogenicity. Knowledge of the relationship between structure<br />

<strong>and</strong> function (i.e., neutralization) would give us the opportunity to develop better vaccine<br />

c<strong>and</strong>idates. The laboratory team is also using X-ray crystallography to analyze the structures of<br />

27


__________________________________________________________________________________<br />

<strong>Center</strong> <strong>for</strong> <strong>Advanced</strong> <strong>Biotechnology</strong> <strong>and</strong> <strong>Medicine</strong><br />

Edward Arnold<br />

some of the engineered viruses (<strong>and</strong> soon peptides), alone <strong>and</strong> in complex with anti-HIV<br />

antibodies. Ultimately we hope to identify three-dimensional correlates of immunogenicity, <strong>and</strong><br />

use this in<strong>for</strong>mation to develop a structural basis <strong>for</strong> design of more effective human vaccines.<br />

There is every reason to expect that a structure-based approach to vaccine development will<br />

become as important to vaccinology as has structure-based drug design to drug discovery <strong>and</strong><br />

development.<br />

In addition to working to develop novel vaccines <strong>and</strong> chemotherapeutic agents, the laboratory<br />

aims to gain greater insights into the basic molecular processes of living systems. Other projects<br />

currently being pursued in the lab include structural studies of: 1) the human mRNA capping<br />

enzyme <strong>and</strong> its associated factors (with Dr. Aaron Shatkin at CABM); 2) the TNF-like cytokine<br />

BLyS (B lymphocyte stimulator, with Yuling Li <strong>and</strong> colleagues at Human Genome Sciences); 3)<br />

<strong>and</strong> hantavirus proteins (with Dr. Colleen Jonssen at University of New Mexico). The structural<br />

group also collaborates with Dr. Gaetano Montelione (CABM) on crystallography of proteins<br />

targeted by the Northeast Structural Genomics Consortium.<br />

Publications:<br />

Sarafianos, S.G., Das, K., Tantillo, C., Clark, A.D. Jr., Ding, J., Whitcomb, J., Boyer, P.L.,<br />

Hughes, S.H. <strong>and</strong> Arnold, E. (<strong>2001</strong>). Crystal structure of HIV-1 reverse transcriptase in<br />

complex with a polypurine tract RNA:DNA. EMBO J. 20: 1449-1461.<br />

Boyer, P.L., Sarafianos, S.G., Arnold, E. <strong>and</strong> Hughes, S.H. (<strong>2001</strong>). Selective excision of AZT<br />

by drug-resistant human immunodeficiency virus reverse transcriptase. J. Virol. 75:4832-4842.<br />

Das, K, Xiong, X., Yang, H., Westl<strong>and</strong>, C.E., Gibbs, C.S., Sarafianos, S.G. <strong>and</strong> Arnold, E.<br />

(<strong>2001</strong>). Molecular modeling <strong>and</strong> biochemical characterization reveals the mechanism of<br />

hepatitis B virus polymerase resistance to lamivudine (3TC) <strong>and</strong> emtricitabine (FTC). J. Virol.<br />

75:4771-4779.<br />

Hsiou, Y., Ding, J., Das, K., Clark, A.D., Jr., Boyer, P.L., Lewi, P., Janssen, P.A.J., Kleim, J.P.<br />

Rösner, M., Hughes, S.H. <strong>and</strong> Arnold, E. (<strong>2001</strong>). The Lys103Asn mutation of HIV-1 RT: a<br />

novel mechanism of drug resistance. J. Mol. Biol. 309:437-445.<br />

Tachedjian, G., Orlova, M., Sarafianos, S.G., Arnold, E. <strong>and</strong> Goff, S.P. (<strong>2001</strong>). Nonnucleoside<br />

reverse transcriptase inhibitors are chemical inducers of dimerization of the human<br />

immunodeficiency type 1 reverse transcriptase. Proc. Natl. Acad. Sci. USA. 98:7188-7193.<br />

Boyer, P.L., Gao, H.-Q., Clark, P.K., Sarafianos, S.G., Arnold, E. <strong>and</strong> Hughes, S.H. (<strong>2001</strong>).<br />

YADD mutants of human immunodeficiency virus type 1 <strong>and</strong> Moloney murine leukemia virus<br />

28


__________________________________________________________________________________<br />

<strong>Center</strong> <strong>for</strong> <strong>Advanced</strong> <strong>Biotechnology</strong> <strong>and</strong> <strong>Medicine</strong><br />

Edward Arnold<br />

reverse transcriptase are resistant to lamivudine triphosphate (3TCTP) in vitro. J. Virol. 75:6321-<br />

6328.<br />

Rossmann, M.G. <strong>and</strong> Arnold, E. (<strong>2001</strong>). Editors of "International Tables <strong>for</strong> X-Ray<br />

Crystallography, Volume F: Crystallography of Biological Macromolecules.” Kluwer Academic<br />

Publishers, Dordrecht, 832 pages.<br />

Arnold, E. <strong>and</strong> Rossmann, M.G. (<strong>2001</strong>). Overview of this volume. In "International Tables <strong>for</strong><br />

X-Ray Crystallography, Volume F: Crystallography of Biological Macromolecules," M.G.<br />

Rossmann <strong>and</strong> E. Arnold, editors. Kluwer Academic Publishers, Dordrecht, pp. 1-3.<br />

Arnold, E. (<strong>2001</strong>). Gazing into the crystal ball: Perspectives <strong>for</strong> the future. In "International<br />

Tables <strong>for</strong> X-Ray Crystallography, Volume F: Crystallography of Biological Macromolecules,"<br />

M.G. Rossmann <strong>and</strong> E. Arnold, editors. Kluwer Academic Publishers, Dordrecht, p. 26.<br />

Arnold, E. <strong>and</strong> Rossmann, M.G. (<strong>2001</strong>). Noncrystallographic symmetry averaging of electron<br />

density <strong>for</strong> molecular-replacement phase refinement <strong>and</strong> extension. In "International Tables <strong>for</strong><br />

X-Ray Crystallography, Volume F: Crystallography of Biological Macromolecules," M.G.<br />

Rossmann <strong>and</strong> E. Arnold, editors. Kluwer Academic Publishers, Dordrecht, pp. 279-292<br />

Ding, J. <strong>and</strong> Arnold, E. (<strong>2001</strong>). Survey of programs <strong>for</strong> crystal structure determination <strong>and</strong><br />

analysis of macromolecules. In "International Tables <strong>for</strong> X-Ray Crystallography, Volume F:<br />

Crystallography of Biological Macromolecules," M.G. Rossmann <strong>and</strong> E. Arnold, editors.<br />

Kluwer Academic Publishers, Dordrecht, pp. 685-694.<br />

Rossmann, M.G. <strong>and</strong> Arnold, E. (<strong>2001</strong>). Patterson <strong>and</strong> molecular replacement techniques. In<br />

International Tables <strong>for</strong> Crystallography. Shmueli, U., ed. Vol. B: Reciprocal Space. Kluwer<br />

Academic Publishers, Dordrecht, 2 nd edition, pp. 235-263.<br />

Ludovici, D.W., Kukla, M.J., Grous, P.G., Krishnan, S., Andries, K., de Béthune, M.-P., Azijn,<br />

H., Pauwels, R., De Clercq, E., Arnold, E. <strong>and</strong> Janssen, P.A.J. (<strong>2001</strong>). Evolution of anti-HIV<br />

drug c<strong>and</strong>idates. Part 1: From ∝-Anilinophenylacetamide (∝-APA) to imidoyl thiourea (ITU).<br />

Bioorg. Med. Chem. Lett. 11:2225-2228.<br />

Ludovici, D.W., Kavash, R.W., Kukla, M.J., Ho, C.Y., Ye, H., De Corte, B.L., Andries, K., de<br />

Béthune, M.P., Azijn, H., Pauwels, R., Moereels, H.E.L., Heeres, J., Koymans, L.M.H., de<br />

Jonge, M.R., Van Aken, K.J.A., Daeyaert, F.F.D., Lewi, P., Das, K., Arnold, E. <strong>and</strong> Janssen,<br />

P.A.J. (<strong>2001</strong>). Evolution of Anti-HIV Drug C<strong>and</strong>idates Part 2: Diaryltriazine (DATA)<br />

Analogues. Bioorg. Med. Chem. Lett. 11:2229-2234.<br />

Ludovici, D.W., De Corte, B.L., Kukla, M.J., Ye, H., Ho, C.Y., Lichtenstein, M.A., Kavash,<br />

R.W., Andries, K., de Béthune, M.-P., Azijn, H., Pauwels, R., Lewi, P.J., Heeres, J., Koymans,<br />

L., de Jonge, M., Van Aken, K.J.A., Daeyaert, F.F.D., Das, K., Arnold, E. <strong>and</strong> Janssen, P.A.J.<br />

29


__________________________________________________________________________________<br />

<strong>Center</strong> <strong>for</strong> <strong>Advanced</strong> <strong>Biotechnology</strong> <strong>and</strong> <strong>Medicine</strong><br />

Edward Arnold<br />

(<strong>2001</strong>). Evolution of anti-HIV drug c<strong>and</strong>idates. Part 3: diarylpyrimidine (DAPY) analogues.<br />

Bioorg. Med. Chem. Lett. 11:2235-2239.<br />

Gao, H-Q., Sarafianos, S.G., Arnold, E. <strong>and</strong> Hughes, S.H. (<strong>2001</strong>). RNase H cleavage of the 5’<br />

end of the HIV-1 genome. J. Virol. 75:11874-11880.<br />

Das, K., Xiao, R., Wahlberg, E., Hsu, F., Arrowsmith, C.H., Montelione, G. <strong>and</strong> Arnold, E.<br />

(<strong>2001</strong>). X-ray crystal structure of MTH938 from Methanobacterium thermoautotrophicum at 2.2<br />

Å resolution reveals a novel tertiary protein fold. Proteins: Structure, Function <strong>and</strong> Genetics<br />

45:486-488.<br />

Fitzgibbon, J.E., Dicola, M.B., Arnold, E., Das, K., Sha, B.E., Pottage, J., Nahass, R., Gaur, S.<br />

<strong>and</strong> John Jr., J.F. HIV-1 reverse transcriptase mutations found in a drug-experienced patient<br />

confer reduced susceptibility to multiple nucleoside reverse transcriptase inhibitors. Antiviral<br />

Therapy. In press.<br />

Oren, D.A., Li, Y., Volovik, Y., Morris, T.S., Dharia, C., Das, K., Galperina, O., Gentz, R. <strong>and</strong><br />

Arnold, E. In the groove: structural basis of BlyS receptor recognition. Nature Structural<br />

Biology. In press.<br />

30


Gaetano Montelione, Ph.D.<br />

Resident Faculty Member, CABM; Professor, <strong>Rutgers</strong>, Department of Molecular Biology<br />

<strong>and</strong> Biochemistry; Adjunct Professor UMDNJ-RWJMS Department of Biochemistry<br />

Protein NMR Laboratory<br />

Dr. Gaetano Montelione did graduate studies in protein physical chemistry with Professor Harold Scheraga at Cornell University. He learned<br />

nuclear magnetic resonance spectroscopy at the Swiss Federal Technical Institute in Zürich where he worked with Dr. Kürt Wüthrich, the first<br />

researcher to solve a protein structure with this technique in 1985. Two years later Dr. Montelione solved the structure of epidermal growth<br />

factor. He has developed new NMR techniques <strong>for</strong> refining 3D structures of proteins <strong>and</strong> triple resonance experiments <strong>for</strong> making 1 H, 13 C, <strong>and</strong><br />

15 N resonance assignments in intermediate-sized proteins. His laboratory has determined 3D solution structures <strong>for</strong> epidermal growth factor,<br />

type-α trans<strong>for</strong>ming growth factor, RNA-binding proteins involved in cold-shock response, <strong>and</strong> immunoglobulin-binding proteins. He is a<br />

member of the NSF Molecular Biophysics Study Section. Dr. Montelione has received the Searle Scholar Award, the Dreyfus Teacher-Scholar<br />

Award, a Johnson <strong>and</strong> Johnson Research Discovery Award, the American Cyanamid Award in Physical Chemistry, the NSF Young Investigator<br />

Award, <strong>and</strong> the Michael <strong>and</strong> Kate Bárány Award of the Biophysical Society.<br />

Goals of our work involve developing high-throughput technologies suitable <strong>for</strong> determining<br />

many new protein structures from the human genome project using nuclear magnetic resonance<br />

spectroscopy (NMR) <strong>and</strong> X-ray crystallography. These structures provide important insights<br />

into the functions of novel gene products identified by genomic <strong>and</strong>/or bioin<strong>for</strong>matic analysis.<br />

The resulting knowledge of structure <strong>and</strong> biochemical function provides the basis <strong>for</strong><br />

collaboration with pharmaceutical companies to develop drugs useful in treating human diseases<br />

that are targeted to these newly discovered functions. The approach we are taking is<br />

opportunistic in the sense that only proteins which express well in bacterial expression systems<br />

are screened <strong>for</strong> their abilities to provide high quality NMR spectra or well-diffracting protein<br />

crystals. Those that provide good NMR or X-ray diffraction data are subjected to automated<br />

analysis methods <strong>for</strong> structure determination. The success of our approach relies on our abilities<br />

to identify, clone, express, <strong>and</strong> analyze hundreds of biologically-interesting proteins per year;<br />

only a fraction of the initial sequences chosen <strong>for</strong> cloning <strong>and</strong> analysis result in high-resolution<br />

3D structures. However, this “funnel” process can yield new functions <strong>for</strong> tens of new structures<br />

per year <strong>and</strong> can thus have tremendous scientific impact. A New Jersey Commission on Science<br />

<strong>and</strong> Technology Excellence Award has been made to a research team organized jointly by<br />

Montelione <strong>and</strong> Steve Anderson to pursue an Initiative in Structural Bioin<strong>for</strong>matics <strong>and</strong><br />

Genomics based on these ideas. Montelione is also director of the NIH-funded Northeast<br />

Structural Genomics Consortium, a multi-year $25 million inter-institutional pilot project in<br />

large scale structural proteomics.<br />

Publications:<br />

Sahasrabudhe, P.V., Xiao, R. <strong>and</strong> Montelione, G.T. (<strong>2001</strong>). Resonance assignments <strong>for</strong><br />

the N terminal domain from human RNA-binding protein with multiple splicing (RBP MS)<br />

J. Biomol. NMR 19: 285-286.<br />

__________________________________________________________________________________<br />

<strong>Center</strong> <strong>for</strong> <strong>Advanced</strong> <strong>Biotechnology</strong> <strong>and</strong> <strong>Medicine</strong><br />

31


Gaetano T. Montelione<br />

Moseley, H.N.B., Monleon, D. <strong>and</strong> Montelione, G.T. ( <strong>2001</strong>) Automatic determination of<br />

protein backbone resonance assignments from triple-resonance NMR data. Meth. Enzymol. 339:<br />

91-108.<br />

Bertone, P., Kluger, Y., Zheng, D.; Edwards, A.M., Arrowsmith, C.H., Montelione,<br />

G.T. <strong>and</strong> Gerstein, M. (<strong>2001</strong>). SPINE: An integrated tracking database <strong>and</strong> data mining<br />

approach <strong>for</strong> prioritizing feasible targets in high-throughput structural proteomics<br />

Nucleic Acids Res. 29: 2884-2898.<br />

Kulikowski, C. A., Muchnik, I., Yun, H. J., Dayanik, A. A., Zheng, D., Song, Y. <strong>and</strong><br />

Montelione, G. T. (<strong>2001</strong>). Protein structural domain parsing by consensus reasoning<br />

over multiple knowledge sources <strong>and</strong> methods. MedInfo 10: 965-969.<br />

Greenfield, N.J., Huang, Y., Palm, T., Swapna, G.V.T., Monleon, D., Montelione, G.T.<br />

<strong>and</strong> Hitchcock-DeGregori, S.E. (<strong>2001</strong>). Solution NMR structure <strong>and</strong> folding dynamics<br />

of the N-terminus of a rat non-muscle α-tropomyosin in an engineered chimeric protein.<br />

J. Mol. Biol. 312: 833 – 847.<br />

Das, K., Xiao, R., Wahlberg, E., Hsu, F., Arrowsmith, C.A., Montelione, G.T. <strong>and</strong><br />

Arnold, E. (<strong>2001</strong>). X-ray crystal structure of MTH938 from Methanobacterium<br />

thermoautotrophicum at 2.2 Å resolution reveals a novel tertiary protein fold. Proteins: Struct.<br />

Funct. Gen. 45: 486 – 488.<br />

Cassetti, M.C., Noah, D.L., Montelione, G.T. <strong>and</strong> Krug, R.M. (<strong>2001</strong>). Efficient translation<br />

of mRNAs in Influenza A virus-infected cells is independent of the viral 5’ untranslated region.<br />

Virology 289: 180-185.<br />

Montelione, G.T. (<strong>2001</strong>). Structural Genomics: An approach to the protein folding<br />

problem. Proc. Natl. Acad. Sci. U.S.A. 98: 13488 –13489.<br />

Sahasrabudhe, P.V., Chien, C.-Y. <strong>and</strong> Montelione, G.T. RNA binding motifs. Encyclopedia of<br />

Life Science. In press.<br />

Monleón, D., Colson, K., Moseley, H.N.B., Anklin, C., Oswald, R., Szyperski, T. <strong>and</strong><br />

Montelione, G.T. Rapid data collection <strong>and</strong> analysis of protein resonance assignments using<br />

AutoProc, AutoPeak, <strong>and</strong> AutoAssign Software. J. Struct. Funct. Genomics. In press.<br />

Swapna, G.V.T., Shukla, K., Huang, Y.J., Ke, H.; Xia, B., Inouye, M. <strong>and</strong> Montelione,<br />

G.T. Resonance assignments <strong>for</strong> cold-shock protein ribosome-binding factor A (RbfA) <strong>for</strong><br />

Escherichia coli. J. Biomol. NMR. In press.<br />

__________________________________________________________________________________<br />

<strong>Center</strong> <strong>for</strong> <strong>Advanced</strong> <strong>Biotechnology</strong> <strong>and</strong> <strong>Medicine</strong><br />

32


Stanley Stein, Ph.D.<br />

Resident Faculty Member, CABM; Adjunct Professor, UMDNJ-Robert Wood Johnson<br />

Medical School, Department of Genetics <strong>and</strong> Microbiology <strong>and</strong> Department of<br />

Neuroscience <strong>and</strong> Cell Biology; Graduate Faculty Member, <strong>Rutgers</strong> University,<br />

Department of Chemistry<br />

Protein Microchemistry Laboratory<br />

Dr. Stanley Stein has held research positions at Hoffmann-LaRoche <strong>and</strong> Schering Corporation where he participated in the discovery <strong>and</strong><br />

development of the prominent anti-viral <strong>and</strong> anti-cancer therapeutic protein, interferon. His interests in biological <strong>and</strong> analytical chemistry are<br />

directed toward both proteins <strong>and</strong> oligonucleotides. He has also contributed in several ways to academic-industrial technology transfer, including<br />

fee-<strong>for</strong>-service analyses, long-term research contracts <strong>and</strong> entrepreneurial enterprises.<br />

The Stein laboratory focuses on improving technologies <strong>for</strong> delivering therapeutic drugs. These<br />

include enhanced cell uptake of drugs against intracellular targets, preferential delivery to target<br />

cells such as to macrophages <strong>for</strong> treating AIDS <strong>and</strong> tuberculosis, sustained drug release over<br />

periods of weeks or months to overcome problems of patient noncompliance, oral delivery of<br />

protein/peptide drugs <strong>and</strong> oral delivery of water-insoluble small molecule drugs <strong>for</strong> cancer <strong>and</strong><br />

other life-threatening diseases.<br />

Publications:<br />

Qiu, B., Brunner, M., Zhang, G., Sigal, L. <strong>and</strong> Stein, S. (2000). Selection of continuous epitope<br />

sequences <strong>and</strong> their incorporation into polyethylene glycol-peptide conjugates <strong>for</strong> use in<br />

serodiagnostic immunoassays: Application to Lyme disease. Biopolymers (Peptide Science) 55:<br />

319-333.<br />

Tung, C.-H. <strong>and</strong> Stein, S. (2000). Preparation <strong>and</strong> applications of peptide-oligonucleotide<br />

conjugates. Bioconjugate Chem. 11: 605-618.<br />

Ramanathan, S., Pooyan, S., Stein, S., Prasad, P. D., Wang, J., Leibowitz, M. J., Ganapathy, V.<br />

<strong>and</strong> Sinko, S. (<strong>2001</strong>). Targeting the sodium dependent multivitamin transporter (SMVT) <strong>for</strong><br />

improving the oral absorption properties of a retro-inverso Tat nonapeptide. Pharmaceutical<br />

Res.18: 950-956.<br />

Ramanathan, S., Qiu, B., Pooyan, S., Zhang, G.,Leibowitz, M. J., Stein, S. <strong>and</strong> Sinko, P.<br />

(<strong>2001</strong>). Targeted PEG-based bioconjugates enhance the cellular<br />

uptake <strong>and</strong> transport of a HIV-1 TAT nonapeptide. J. Controlled Release 77: 199-213.<br />

__________________________________________________________________________________<br />

<strong>Center</strong> <strong>for</strong> <strong>Advanced</strong> <strong>Biotechnology</strong> <strong>and</strong> <strong>Medicine</strong><br />

33


Ann Stock, Ph.D.<br />

Associate Investigator, Howard Hughes Medical Institute; Resident Faculty Member,<br />

CABM; Professor, UMDNJ-Robert Wood Johnson Medical School, Department of<br />

Biochemistry<br />

Protein Crystallography Laboratory<br />

Dr. Ann Stock per<strong>for</strong>med graduate work on the biochemistry of signal transduction proteins with Professor Daniel E. Koshl<strong>and</strong>, Jr. at the<br />

University of Cali<strong>for</strong>nia at Berkeley. From 1987 to 1991 she pursued structural analysis of these proteins while a postdoctoral fellow with<br />

Professor Clarence Schutt at Princeton University <strong>and</strong> Professor Gregory Petsko at the Structural Biology Laboratory in the Rosenstiel <strong>Center</strong> at<br />

Br<strong>and</strong>eis University. Her CABM laboratory has solved structures of several signal transduction proteins, including receptor modification<br />

enzymes <strong>and</strong> members of the two-components family of signal transduction proteins. Stock has received National Science Foundation<br />

Predoctoral <strong>and</strong> Damon Runyon-Walter Winchell Postdoctoral Fellowships, a Lucille P. Markey Scholar Award in Biomedical Science, the NSF<br />

Young Investigator Award <strong>and</strong> a Sinsheimer Scholar Award. She is an associate investigator of the Howard Hughes Medical Institute.<br />

Research in the Stock laboratory focuses on structure/function studies of signal transduction<br />

proteins. Ef<strong>for</strong>t is concentrated on bacterial histidine protein kinases <strong>and</strong> response regulator<br />

proteins that mediate the majority bacterial signaling processes. These systems are important <strong>for</strong><br />

virulence in pathogenic organisms <strong>and</strong> are currently targets <strong>for</strong> development of new antibiotics in<br />

several pharmaceutical companies. During the past year the Stock laboratory determined the<br />

first structure <strong>for</strong> a full-length member of the OmpR family of transcription factors, the largest<br />

subfamily of response regulator proteins. The structure provides insight into the mechanism of<br />

regulation of the protein by phosphorylation <strong>and</strong> suggests a limit to the extent of mechanistic<br />

similarities between proteins of similar structure <strong>and</strong> function.<br />

Publications:<br />

Schlichting, I., Berendzen, J., Chu, K., Stock, A.M., Maves, S.A., Benson, D.E., Sweet, R.M.,<br />

Ringe, D., Petsko, G.A., <strong>and</strong> Sligar, S.G. (2000). The catalytic pathway of cytochrome P450cam<br />

at atomic resolution. Science 287: 1615-1622.<br />

Stock, A.M., Robinson, V.L. <strong>and</strong> Goudreau, P.N. (2000). Two-component signal transduction.<br />

Annu. Rev. Biochem. 69: 183-215.<br />

Buckler, D.R., An<strong>and</strong>, G.S. <strong>and</strong> Stock, A.M. (2000). Response regulator phosphorylation <strong>and</strong><br />

activation: a two-way street? Trends Microbiol. 8: 153-155.<br />

An<strong>and</strong>, G.S., Goudreau, P.N., Lewis, J.K. <strong>and</strong> Stock, A.M. (2000). Evidence <strong>for</strong><br />

phosphorylation-dependent con<strong>for</strong>mational changes in methylesterase CheB. Protein Sci. 9: 898-<br />

906.<br />

Buckler, D.R. <strong>and</strong> Stock, A.M. (2000). Synthesis of [ 32 P]phosphoramidate <strong>for</strong> use as a low<br />

molecular weight phosphodonor reagent. Anal. Biochem. 283: 222-227.<br />

__________________________________________________________________________________<br />

<strong>Center</strong> <strong>for</strong> <strong>Advanced</strong> <strong>Biotechnology</strong> <strong>and</strong> <strong>Medicine</strong><br />

34


__________________________________________________________________________________<br />

<strong>Center</strong> <strong>for</strong> <strong>Advanced</strong> <strong>Biotechnology</strong> <strong>and</strong> <strong>Medicine</strong><br />

Ann Stock<br />

Robinson, V.L., Buckler, D.R. <strong>and</strong> Stock, A.M. (2000). A tale of two components: a novel<br />

kinase <strong>and</strong> a regulatory switch. Nature Struct. Biol. 7: 628-633.<br />

Hughes, C.A., M<strong>and</strong>ell, J.G., An<strong>and</strong>, G.S., Stock, A.M. <strong>and</strong> Komives, E.A. (<strong>2001</strong>).<br />

Phosphorylation causes subtle changes in solvent accessibility at the interdomain interface of<br />

methylesterase CheB. J. Mol. Biol. 307: 967-976.<br />

West, A.H. <strong>and</strong> Stock, A.M. (<strong>2001</strong>). Histidine kinases <strong>and</strong> response regulator proteins in twocomponent<br />

signaling systems. Trends Biochem. Sci. 26: 369-376.<br />

Hughes, S.H. <strong>and</strong> Stock, A.M. (<strong>2001</strong>). Techniques in molecular biology <strong>and</strong> protein expression:<br />

Preparing recombinant proteins <strong>for</strong> X-ray crystallography. In International Tables <strong>for</strong><br />

Crystallography. Vol. F Crystallography of Biological Macromolecules, M.G. Rossmann <strong>and</strong> E.<br />

Arnold, eds., Kluwer Academic Publishers, Dordrecht. The Netherl<strong>and</strong>s. 65-80.<br />

Saxl, R.L., An<strong>and</strong>, G.S. <strong>and</strong> Stock, A.M. (<strong>2001</strong>). Synthesis <strong>and</strong> biochemical characterization of a<br />

phosphorylated analog of the response regulator CheB. Biochemistry 40: 12896-12903.<br />

35


Molecular Genetics<br />

___________________________________________________________________________________________________________<br />

Laboratory Faculty Director<br />

Protein Engineering Stephen Anderson 37<br />

Viral Pathogenesis Arnold Rabson 39<br />

Molecular Virology Aaron Shatkin 41<br />

__________________________________________________________________________________<br />

<strong>Center</strong> <strong>for</strong> <strong>Advanced</strong> <strong>Biotechnology</strong> <strong>and</strong> <strong>Medicine</strong><br />

36


Stephen Anderson, Ph.D.<br />

Resident Faculty Member, CABM; Associate Professor, <strong>Rutgers</strong> University, Department<br />

of Molecular Biology <strong>and</strong> Biochemistry<br />

Dr. Stephen Anderson conducted his postdoctoral research under Nobel laureate Dr. Frederick Sanger at the MRC Laboratory of Molecular<br />

Biology in Cambridge, Engl<strong>and</strong>. That project involved the sequencing of human <strong>and</strong> bovine mitochondrial genomes. He went on to a position at<br />

the Cali<strong>for</strong>nia-based biotechnology start-up company Genentech. Anderson has held research or teaching positions at Harvard University, the<br />

MRC Laboratory of Molecular Biology, Genentech, Inc., University of Cali<strong>for</strong>nia, San Francisco, <strong>and</strong> <strong>Rutgers</strong> University. While at Genentech<br />

he was responsible <strong>for</strong> all specialty chemical projects <strong>and</strong> second generation tissue plasminogen activator research. He is an associate editor of<br />

the journal SEQUENCE. Currently Dr. Anderson is on leave at GeneFormatics, Inc.<br />

Protein Engineering Laboratory<br />

Our laboratory is focusing on the study of protein folding <strong>and</strong> molecular recognition. We are<br />

particularly interested in the role these processes may play in the pathophysiology of<br />

Alzheimer’s disease. The Alzheimer’s β amyloid precursor protein (APP) is a large, membranebound<br />

glycoprotein that is expressed in the body from several different alternatively-spliced<br />

mRNAs. Contained within this precursor protein is the amyloid β peptide which has been<br />

strongly implicated as a causal agent in Alzheimer’s disease. Our laboratory is currently using a<br />

protein engineering strategy to dissect the structure <strong>and</strong> function of the APP molecule as well as<br />

its component domains. We are also seeking to identify <strong>and</strong> isolate other proteins in the body<br />

that bind to APP. We have discovered that amyloid β peptide fibrils bind <strong>and</strong> activate both<br />

tissue plasminogen activator <strong>and</strong> plasminogen in a manner remarkably similar to the known<br />

physiological cofactor, fibrin. This phenomenon may shed light on the involvement of amyloid<br />

peptide fibrils in cerebral amyloid angiopathy <strong>and</strong> hemorrhagic stroke.<br />

Recently the laboratory has been using a combination of protein expression <strong>and</strong> biophysical<br />

approaches to create a new structure-based functional genomics paradigm <strong>for</strong> analyzing<br />

“orphan” gene sequences. We have embarked on an ambitious, multidisciplinary, collaborative<br />

project to develop new methods <strong>for</strong> analyzing protein 3D structure <strong>and</strong> function. These methods,<br />

termed structural genomics, will be used to interpret the flood of novel gene sequence data being<br />

produced by the human genome project. The work on structural genomics, in collaboration with<br />

Professor Gaetano Montelione at CABM <strong>and</strong> other colleagues at <strong>Rutgers</strong>, UMDNJ-Robert Wood<br />

Johnson Medical School, CABM <strong>and</strong> HHMI, has picked up momentum during the last year.<br />

Significant progress has been made in underst<strong>and</strong>ing the structure <strong>and</strong> dimerization properties of<br />

the BRCT domain, which is found in the human breast cancer gene, BRCA1. We are also<br />

working on highly conserved bacterial gene products that are found in Mycobacterium<br />

tuberculosis <strong>and</strong> other human pathogens but not in humans themselves – underst<strong>and</strong>ing the<br />

structure <strong>and</strong> function of these may suggest strategies <strong>for</strong> developing novel antibiotics. Finally,<br />

we have embarked upon the scaled-up expression <strong>and</strong> biophysical characterization, in a pilotstudy<br />

mode, of a series of small orphan gene products from the roundworm, C. elegans.<br />

Our work on molecular recognition has centered on the well-characterized protease-protease<br />

inhibitor interaction. The specific model system we have employed is the interaction of avian<br />

ovomucoid third domains (OM3D) with members of the trypsin family of serine proteases. Our<br />

__________________________________________________________________________________<br />

<strong>Center</strong> <strong>for</strong> <strong>Advanced</strong> <strong>Biotechnology</strong> <strong>and</strong> <strong>Medicine</strong><br />

37


approach is the expression of recombinant turkey OM3D in E. coli <strong>and</strong> specific mutagenesis of<br />

reactive site residues. Mutant inhibitors are then tested <strong>for</strong> binding affinity with a panel of serine<br />

_____________________________________________________________<br />

Stephen Anderson<br />

proteases. Our goal is to provide a complete, canonical set of ovomucoid third domain (OM3D)<br />

variants with every amino acid at the reactive site mutagenized, individually, to every other<br />

amino acid. This project is virtually complete, with the last mutants currently being made. The<br />

work is being conducted in collaboration with Dr. Michael Laskowski Jr.’s laboratory at Purdue<br />

University <strong>and</strong> Dr. Michael James’ laboratory at Edmonton, Alberta. The resultant database will<br />

represent an unparalleled resource <strong>for</strong> drug design groups <strong>and</strong> other users interested in<br />

rationalizing the energetics of protein-protein interactions.<br />

We also have a long-term ef<strong>for</strong>t aimed at developing improved industrial methods <strong>for</strong><br />

synthesizing vitamin C. We are working on increasing the activity of a crucial enzyme in the<br />

pathway from glucose to 2-keto-L-gulonic acid [a precursor of vitamin C], 2,5-diketo-Dgluconic<br />

acid (2,5-DKG) reductase, using protein engineering techniques <strong>and</strong> extensive<br />

mutagenesis of the enzymes’ substrate <strong>and</strong> cofactor binding sites.<br />

Publications:<br />

Bateman, K. S., Anderson, S., Lu, W., Qasim, M. A., Laskowski Jr., M. L. <strong>and</strong> James, M. N. G.<br />

(2000). Deleterious effects of b-branched residues in the S1 specificity pocket of Streptomyces<br />

griseus proteinase B (SGPB), crystal structures of the turkey ovomucoid third domain variants<br />

Ile18I, Val18I, Thr18I, <strong>and</strong> Ser18I in complex with SGPB. Protein Sci., 9:83 - 94.<br />

Khurana, S., Powers, D. B., Anderson, S. <strong>and</strong> Blaber, M. (2000). Molecular dynamics of 2,5diketo-D-gluconic<br />

acid reductase A complexed with substrate. Proteins, Struct. Funct. & Genet.,<br />

39: 68-75.<br />

Bateman, K.S., Huang, K., Anderson, S., Lu,W., Qasim, M.A., Laskowski, M. Jr. <strong>and</strong> James,<br />

M.N. (<strong>2001</strong>). Contribution of peptide bonds to inhibitor-protease binding: crystal structures of<br />

the turkey ovomucoid third domain backbone variants OMTKY3-Pro18I <strong>and</strong> OMTKY3psi[COO]-leu18I<br />

in complex with Streptomuces griseus proteinase B (SGPB) <strong>and</strong> the structure<br />

of the free inhibitor, OMTKY-3-psi[CH2NH2+]-Asp19I. J. Mol. Biol. 305:839-49.<br />

__________________________________________________________________________________<br />

<strong>Center</strong> <strong>for</strong> <strong>Advanced</strong> <strong>Biotechnology</strong> <strong>and</strong> <strong>Medicine</strong><br />

38


Arnold B. Rabson, M.D.<br />

Resident Faculty Member, CABM; Professor, UMDNJ-Robert Wood Johnson Medical<br />

School, Department of Molecular Genetics <strong>and</strong> Microbiology; Adjunct Professor,<br />

UMDNJ-RWJMS, Department of Pathology<br />

Viral Pathogenesis Laboratory<br />

Dr. Arnold Rabson joined CABM in the summer of 1990. Previously, he per<strong>for</strong>med his residency in pathology at Brigham <strong>and</strong> Women’s<br />

Hospital in Boston. He was a medical staff fellow <strong>and</strong> a senior staff fellow in Malcolm Martin’s laboratory at the National Institute of Allergy<br />

<strong>and</strong> Infectious Diseases from 1981 to 1990. Dr. Rabson has served on a number of federal grant review boards, including an NIH Special Review<br />

Committee <strong>for</strong> AIDS. He is a member of the NIH Pathology B study section <strong>and</strong> has served as Chairman of several Special Emphasis Panels of<br />

the NIH <strong>Center</strong> <strong>for</strong> Scientific Review. He is on the editorial board of the Journal of Virology, Clinical Cancer Research <strong>and</strong> the Journal of<br />

Biomedical Science. Dr. Rabson is also the Associate Director <strong>for</strong> Basic Research of the Cancer Institute of New Jersey (CINJ) <strong>and</strong> directs the<br />

Transcriptional Regulation <strong>and</strong> Oncogenesis Program of CINJ.<br />

Dr. Rabson’s laboratory focuses on the molecular basis of cancer <strong>and</strong> human retroviruses. His<br />

research program encompasses studies of the viral <strong>and</strong> cellular mechanisms that regulate the<br />

expression of human retroviruses that cause cancer <strong>and</strong> AIDS, the human T-cell leukemia virus<br />

type 1 (HTLV-1) <strong>and</strong> the human immunodeficiency virus (HIV). A second aspect of the<br />

laboratory studies the roles of cellular transcription factors in the development <strong>and</strong> progression<br />

of human malignancies, including leukemia, lymphomas, <strong>and</strong> prostate cancer.<br />

Previous studies in Dr. Rabson's laboratory identified the frequent occurrence of molecular<br />

alterations in the gene encoding the NFKB2 transcription factor in the malignant cells of patients<br />

with cutaneous T-cell lymphoma (CTCL). Over the past year, Dr. Rabson’s laboratory has<br />

demonstrated that the tumor associated NF-κB-2 mutants also gain new gene regulatory<br />

functions. Regulated expression of these aberrant proteins can prevent T cell death <strong>and</strong> increase<br />

proliferation; thus the lymphoma associated rearrangements lead to both a loss of function <strong>and</strong> a<br />

gain of new transcriptional regulation. Dr. Rabson is now identifying the target genes of the<br />

tumor associated mutant proteins that may explain how this genetic alteration leads to T cell<br />

lymphomas.<br />

Dr. Rabson's laboratory has demonstrated inappropriate, constitutive, activation of NF-κB in a<br />

series of aggressive, tumorigenic, human prostate cancer cell lines <strong>and</strong> has shown inappropriate<br />

activation of NF-κB in focal areas of primary prostate cancer samples from patients with the<br />

disease. He has further shown that constitutive, inappropriate activation of NF-κB in the prostate<br />

cancer cells is due to activation of regulatory kinases such as the IkappaB kinases <strong>and</strong> the NF-κB<br />

kinase. He is continuing to study the consequences of NF-kB expression in prostate cancer cells.<br />

This work offers possible new approaches <strong>for</strong> therapy of the disease.<br />

__________________________________________________________________________________<br />

<strong>Center</strong> <strong>for</strong> <strong>Advanced</strong> <strong>Biotechnology</strong> <strong>and</strong> <strong>Medicine</strong><br />

39


Collaborative studies with Drs. Strair (CINJ) <strong>and</strong> White (CABM) are evaluating the potential<br />

utility of highly attenuated adenoviruses as cytotoxic agents <strong>for</strong> the therapy of different<br />

subsets of leukemias <strong>and</strong> lymphomas since mutated, highly attenuated adenoviruses may be<br />

__________________________________________________________________________________<br />

<strong>Center</strong> <strong>for</strong> <strong>Advanced</strong> <strong>Biotechnology</strong> <strong>and</strong> <strong>Medicine</strong><br />

Arnold B. Rabson<br />

useful in the therapy of this disease. In collaboration with Dr. R. Strair (CINJ) <strong>and</strong> Dr. A.<br />

Conney (<strong>Rutgers</strong> University), Dr. Rabson is studying the molecular mechanisms by which the<br />

phorbol ester, TPA, can induce differentiation of human leukemic cells. The collaborative<br />

studies at CABM have focused on the effects of TPA on NF-kB <strong>and</strong> on chromatin structure<br />

during the induction of differentiation.<br />

The Rabson laboratory has identified <strong>and</strong> characterized models of latent HTLV-1 infection <strong>and</strong><br />

demonstrated that immune activation stimuli can potently induce HTLV-1 gene expression. This<br />

suggests that stimulation of particular T cell clones through their T cell receptor could lead to<br />

enhanced HTLV-1 gene expression, resulting in increased T cell proliferation. This could<br />

explain the polyclonal to oligoclonal proliferation of infected T cells that characterizes HTLV-1associated<br />

diseases. Over the last year, Dr. Rabson’s laboratory has demonstrated that the<br />

HTLV-1 Tax protein is the target of the T cell activation stimuli. These results suggest that<br />

inhibition of T cell signaling might alter the progression of HTLV-1 infection.<br />

Publications:<br />

Rabson, A.B. <strong>and</strong> Lin, H.-C. (2000). NF-kappaB <strong>and</strong> HIV: Linking Viral <strong>and</strong> Immune<br />

Activation. Adv. Pharm. 48:161-207.<br />

Rabson, A.B., Padarathsingh, M. <strong>and</strong> Le Beau, M. (2000). Molecular Biology <strong>and</strong> Experimental<br />

Models <strong>for</strong> Hematologic Malignant Diseases: Workshop of the NIH Pathology B Study Section.<br />

Biochim. Biophys. Acta Reviews on Cancer, B R1-R7.<br />

Ke S, Rabson A.B., Germino J.F., Gallo M.A. <strong>and</strong> Tian Y. (<strong>2001</strong>). Mechanism of Suppression<br />

of Cytochrome P-450 1A1 Expression by Tumor Necrosis Factor-alpha <strong>and</strong> Lipopolysaccharide.<br />

J. Biol. Chem. 276:39638-39644, <strong>2001</strong>.<br />

Zheng, X., Chang, R.L., Cui, X.-X., Kelly, K.A., Strair, R., Suh, J., Han, Z.T., Rabson, A. <strong>and</strong><br />

Conney, A.H. (<strong>2001</strong>). Synergistic effects of clinically achievable concentrations of 12-Otetradecanoylphorbol-13-acetate<br />

in combination with all-trans retinoic acid, 1α, 25dihydroxyvitamin<br />

D 3 <strong>and</strong> sodium butyrate on differentiation of HL-60 cells. Oncol Res. 12:419-<br />

27.<br />

40


Aaron J. Shatkin, Ph.D.<br />

Director, CABM; University Professor of Molecular Biology at <strong>Rutgers</strong> University <strong>and</strong><br />

Professor, UMDNJ-Robert Wood Johnson Medical School, Department of Molecular<br />

Genetics <strong>and</strong> Microbiology<br />

Molecular Virology Laboratory<br />

Dr. Aaron J. Shatkin, a member of the National Academy of Sciences, has held research positions at the National Institutes of Health, The Salk<br />

Institute, <strong>and</strong> the Roche Institute of Molecular Biology. He has taught at, among other institutions, Georgetown University Medical School, Cold<br />

Spring Harbor Laboratory, The Rockefeller University, UMDNJ-Newark Medical School, University of Puerto Rico, <strong>and</strong> Princeton University.<br />

He was editor of the Journal of Virology from 1973-1977, founding editor-in-chief of Molecular <strong>and</strong> Cellular Biology from 1980-1990, <strong>and</strong> is<br />

currently an editor of Advances in Virus Research <strong>and</strong> a member of the editorial board of the Proceedings of the National Academy of Sciences.<br />

Shatkin serves on the advisory boards of a number of organizations including the Howard Hughes Medical Institute, <strong>and</strong> in 1989 he was<br />

recognized by New Jersey Monthly magazine with a New Jersey Pride Award <strong>for</strong> his contributions to the State’s economic development. In 1991<br />

the State of N.J. awarded Shatkin the Thomas Alva Edison Science Award. He was elected Fellow of the American Academy of Arts <strong>and</strong><br />

Sciences in 1997, <strong>and</strong> the American Association <strong>for</strong> the Advancement of Science in 1999.<br />

One of the earliest steps in the cascade of controls that regulate mRNA <strong>for</strong>mation <strong>and</strong> function is<br />

the addition of a 5'-terminal "cap." This structural hallmark is present on all eukaryotic cellular<br />

mRNAs <strong>and</strong> is essential <strong>for</strong> viability. The cap enhances several downstream events in gene<br />

expression including splicing of pre-mRNAs, initiation of protein synthesis <strong>and</strong> mRNA stability.<br />

Recently we have cloned <strong>and</strong> sequenced the mouse <strong>and</strong> human capping enzymes (CE) <strong>and</strong><br />

mapped the human protein to 6q16, a region implicated in tumor suppression. The mammalian<br />

CE are 597-aa polypeptides consisting of two functional domains, N-terminal RNA 5'<br />

triphosphatase (RT) <strong>and</strong> C-terminal guanylyltransferase (GT). Mutational analysis demonstrated<br />

that the GT active site lysine is present in one of several highly conserved motifs characteristic of<br />

a nucleotidyltransferase superfamily. A haploid strain of S. cerevisiae lacking GT was<br />

complemented <strong>for</strong> growth by the mouse wild type cDNA clone but not by a clone containing<br />

alanine in place of the active-site lysine. The results demonstrate the functional conservation of<br />

CE from yeast to mammals.<br />

We found that mammalian CE binds via its GT domain to the hyperphosphorylated C-terminal<br />

domain (P-CTD) of RNA polymerase II,accounting <strong>for</strong> the selective capping of pre-mRNAs. The<br />

CE N-terminal RT contains the sequenceVHCTHGFNRTG which corresponds to the conserved<br />

active-site motif in protein tyrosine phosphatases (PTPs). Mutational analyses indicate that<br />

removal of phosphate from RNA 5' ends <strong>and</strong> from protein tyrosines occurs by a similar<br />

mechanism. We have also cloned, mapped to 18p11.22-p11.23 <strong>and</strong> characterized the third<br />

essential enzyme <strong>for</strong> capping--mRNA (guanine-7-) methyltransferase (MT). Sequence alignment<br />

of the 476-aa human MT with the corresponding yeast, worm <strong>and</strong> fly enzymes demonstrated<br />

several required, conserved motifs including one <strong>for</strong> binding S-adenosylmethionine.<br />

Yeast two-hybrid screening with CE yielded transcription elongation factor SPT5 which<br />

stimulated GT, but the effects were not additive with SPT5, suggesting a common binding site<br />

__________________________________________________________________________________<br />

<strong>Center</strong> <strong>for</strong> <strong>Advanced</strong> <strong>Biotechnology</strong> <strong>and</strong> <strong>Medicine</strong><br />

41


on CE. We also found that MT interacts with the nuclear transporter, importin-alpha (Impa). MT<br />

selectively bound <strong>and</strong> methylated RNA containing 5'-GpppG, <strong>and</strong> both activities were stimulated<br />

Aaron J. Shatkin<br />

several-fold by Impa. MT/RNA/Impa complexes were dissociated by addition of Imp-beta<br />

(Impb) which also blocked Impa stimulation of RNA cap methylation. RanGTP but not RanGDP<br />

prevented these effects of Impb. The results suggest that, in addition to a linkage between<br />

capping <strong>and</strong> transcription, mRNA biogenesis <strong>and</strong> nucleocytoplasmic transport are functionally<br />

connected, a possibility we are exploring further.<br />

Publications:<br />

Shatkin, A.J. (2000). Reovirus translational control. Translational Control, Sonenberg, N.,<br />

Hershey, J.W.B., Mathews, M.B., eds. Cold Spring Harbor Laboratory Press, 2nd edition, 915-<br />

932.<br />

Furuichi, Y. <strong>and</strong> Shatkin, A.J. (2000). Viral <strong>and</strong> Cellular mRNA Capping: Past <strong>and</strong> prospects.<br />

Adv. in Vir. Res, 55:135-184.<br />

Shatkin, A.J. <strong>and</strong> Manley, J.L. (2000). The ends of the affair: Capping <strong>and</strong> polyadenylation.<br />

Nature Structural Biology. 7:838-842.<br />

Wen, Y. <strong>and</strong> Shatkin, A.J. (2000). Cap methyltransferase selective binding <strong>and</strong> methylation of<br />

GpppG-RNA are stimulated by importin-α. Genes <strong>and</strong> Development. 14:2944-2949.<br />

__________________________________________________________________________________<br />

<strong>Center</strong> <strong>for</strong> <strong>Advanced</strong> <strong>Biotechnology</strong> <strong>and</strong> <strong>Medicine</strong><br />

42


Education, Training &<br />

Technology Transfer<br />

___________________________________________________________________________________________________________<br />

__________________________________________________________________________________<br />

<strong>Center</strong> <strong>for</strong> <strong>Advanced</strong> <strong>Biotechnology</strong> <strong>and</strong> <strong>Medicine</strong><br />

43


Lectures <strong>and</strong> Seminars<br />

__________________________________________________________________________________<br />

<strong>Center</strong> <strong>for</strong> <strong>Advanced</strong> <strong>Biotechnology</strong> <strong>and</strong> <strong>Medicine</strong><br />

44


January 24<br />

James Manley, Biological Sciences, Columbia University<br />

“mRNA Processing <strong>and</strong> the Integration of Nuclear Events”<br />

February 14<br />

Heiner Westphal, NIH<br />

“How Mouse LIM Homeobox Genes Control Organogenesis”<br />

CABM Lecture Series<br />

March 29<br />

Rick Young, MIT<br />

“Lessons from Global Expression Profiling of Yeast <strong>and</strong> Human Cells”<br />

April 18<br />

Nadia Rosenthal, Harvard Medical School<br />

“Retinoic Acid <strong>and</strong> Heart Development”<br />

May 16<br />

Se-Jin Lee, Johns Hopkins University<br />

“Novel Growth <strong>and</strong> Differentiation Factors Related to TFG-β”<br />

September 19<br />

Jon Clardy, Chemistry <strong>and</strong> Chemical Biology, Cornell University<br />

“Natural Products <strong>and</strong> Natural Product Libraries”<br />

October 24 & 25<br />

15 th Annual CABM Symposium<br />

“Structural Genomics in Pharmaceutical Design”<br />

December 6<br />

Michael B. Kastan, Hematology-Oncology, St. Jude’s Children’s<br />

Research Hospital<br />

__________________________________________________________________________________<br />

<strong>Center</strong> <strong>for</strong> <strong>Advanced</strong> <strong>Biotechnology</strong> <strong>and</strong> <strong>Medicine</strong><br />

45


“DNA Damage Response Pathways in the Genesis <strong>and</strong> Treatment of Cancer”<br />

-- The CABM Lecture Series is supported in part by Aventis --<br />

ANNUAL CABM RETREAT, June 11 th , <strong>2001</strong><br />

FOREST LODGE, Warren, NJ<br />

Program<br />

8:15 AM Registration, Poster Set up <strong>and</strong> Continental Breakfast<br />

9:00 AM Opening Remarks – Aaron J. Shatkin<br />

Professor <strong>and</strong> Director, CABM<br />

9:10 AM Proteomics: Protein Structure, Engineering <strong>and</strong> Applications<br />

Chair: Hunter Moseley – Protein NMR Spectroscopy Lab (Guy Montelione)<br />

10:40 AM Coffee Break<br />

Tom Acton – Protein NMR Spectroscopy Lab (Guy Montelione)<br />

"The Northeast Structural Genomics Consortium, a Pilot Project Focusing on<br />

Eukaryotic Organisms"<br />

Scott Banta – Protein Engineering Lab (Stephen Anderson)<br />

“Alteration of the Cofactor Specificity of Corynebacterium<br />

2,5-diketo-D-gluconic Acid Reductase”<br />

Shahriar Pooyan – Protein Microchemistry Lab (Stanley Stein)<br />

“PEG Carrier <strong>for</strong> Drug Targeting”<br />

Li Lin – Protein Targeting Lab (Peter Lobel)<br />

“The Human CLN2 Protein/Tripeptidyl-peptidase I Is A Serine Protease”<br />

David Buckler – Protein Crystallography Lab (Ann Stock)<br />

“X-ray Crystal Structure of An OmpR Homolog from Thermotoga maritima"<br />

Deena Oren – Biomolecular Crystallography Lab (Eddy Arnold)<br />

“Determining the Structure of the Human B Lymphocyte Stimulator BlyS”<br />

11:00 AM Gene Expression <strong>and</strong> Cancer<br />

Chair: Leonard Edelstein – Tumor Virology Lab (Céline Gélinas)<br />

Priya Srinivasan – Molecular Virology Lab (Aaron J. Shatkin)<br />

__________________________________________________________________________________<br />

<strong>Center</strong> <strong>for</strong> <strong>Advanced</strong> <strong>Biotechnology</strong> <strong>and</strong> <strong>Medicine</strong><br />

46


“Analysis of C. elegans mRNA Capping Machinery by RNA Interference”<br />

Annual CABM Retreat<br />

Wen-Feng Chen – Molecular Chronobiology Lab (Isaac Edery)<br />

"Analysis of Locomotor Activity <strong>and</strong> Per mRNA Splicing in Flies from Different<br />

Latitudes"<br />

Paul Lizzul – Tumor Virology Lab (Céline Gélinas)<br />

“TAPIR is a Novel NF-κB-inducible Gene, Distinct from IκB, Which Suppresses<br />

NF-κB-mediated Transactivation in Response to TNFα <strong>and</strong> IL-1β Signaling”<br />

Junghan Suh – Viral Pathogenesis Lab (Arnold Rabson)<br />

"Mechanisms of NF-kB Activation in Prostate Cancer Cells"<br />

Isao Matsuura – Growth <strong>and</strong> Differentiation Control Lab (Fang Liu)<br />

“Phosphorylation of SMAD by Cyclin-dependent Kinases”<br />

Kurt Degenhardt – Viral Trans<strong>for</strong>mation Lab (Eileen White)<br />

“Adenovirus Induces DNA Fragmentation through a DFF-45/ICAD Dependent<br />

Mechanism That Is Inhibited by E1B 19K”<br />

12:30-3:30 PM Lunch <strong>and</strong> Free Time<br />

3:30 PM Poster Presentation (odd numbers)<br />

5:00 PM Mammalian Development<br />

Chair: Yu-Ting Yan – Mammalian Embryogenesis Lab (Michael Shen)<br />

Jixiang Ding – Mammalian Embryogenesis Lab (Michael Shen)<br />

“Eyes Wide Apart - Cripto <strong>and</strong> Midline Formation”<br />

Hansol Lee – Molecular Neurobiology Lab (Cory Abate-Shen)<br />

"Searching <strong>for</strong> the Regulatory Network of Msx1 Homeobox Protein"<br />

Shengguo Li – Molecular Neurodevelopment Lab (Mengqing Xiang)<br />

“Functional Analysis of the Barhl1 Homeobox Gene during Development of the<br />

Mouse Nervous System"<br />

Ahmed Usmani – Developmental Neurogenetics Lab (James Millonig)<br />

“Dorsal CNS Development in the Mutant Mouse Dreher”<br />

__________________________________________________________________________________<br />

<strong>Center</strong> <strong>for</strong> <strong>Advanced</strong> <strong>Biotechnology</strong> <strong>and</strong> <strong>Medicine</strong><br />

47


6:00 PM Poster Presentation (even numbers) <strong>and</strong> Buffet Reception<br />

15 th Annual CABM Symposium<br />

Structural Genomics in Pharmaceutical Design<br />

Princeton Forrestal Conference <strong>Center</strong><br />

Princeton, New Jersey<br />

October 24-25, <strong>2001</strong><br />

CABM Organizing Committee<br />

Edward Arnold Aaron J. Shatkin<br />

Gaetano Montelione Ann Stock<br />

Sponsors<br />

-Pfizer -Roche<br />

-Wyeth -Novartis<br />

-NJ Commission on Science -Bruker Biospin<br />

<strong>and</strong> Technology -Isotec Inc.<br />

-GeneFormatics, Inc. -Genentech, Inc.<br />

-Upstate <strong>Biotechnology</strong> -Martek<br />

-Merck Research Laboratories -Rigaku/MSC, Inc.<br />

-Structural GenomiX -Schering-Plough<br />

-3-Dimensional Research Institute<br />

Pharmaceuticals, Inc. -Bristol-Myers Squibb<br />

-Medarex<br />

Exhibitors<br />

-Bruker Daltonics, Inc. -ProteEx<br />

-GeneFormatics, Inc. -Rigaku/MSC, Inc.<br />

-Isotec Inc. -TKR Biotech Products<br />

-NJ Commission on Science -Upstate <strong>Biotechnology</strong><br />

__________________________________________________________________________________<br />

<strong>Center</strong> <strong>for</strong> <strong>Advanced</strong> <strong>Biotechnology</strong> <strong>and</strong> <strong>Medicine</strong><br />

48


<strong>and</strong> Technology -VLI Research Inc./<br />

-NJ Economic Development Authority Silantes<br />

-Prosoft Software, Inc.<br />

15 th Annual CABM Symposium<br />

Wednesday, October 24, <strong>2001</strong><br />

Program<br />

8:00 AM Registration <strong>and</strong> Continental Breakfast<br />

8:45 AM Opening Remarks<br />

Aaron Shatkin, Director, <strong>Center</strong> <strong>for</strong> <strong>Advanced</strong> <strong>Biotechnology</strong> <strong>and</strong> <strong>Medicine</strong><br />

EXPERIMENTAL STRUCTURAL GENOMICS<br />

Chair: Edward Arnold, <strong>Rutgers</strong> University <strong>and</strong> CABM<br />

9:00 AM Vim Hol, University of Washington<br />

“Protein Structures from Tropical Parasites: A Drug Design Perspective”<br />

9:40 AM Gaetano Montelione, <strong>Rutgers</strong> University <strong>and</strong> CABM<br />

“Structural Proteomics of Eukaryotic Gene Families”<br />

10:20 AM Sean Buchanan, Structural GenomiX<br />

“New Bacterial Drug Targets from Structural Genomics”<br />

10:40 AM Refreshment Break<br />

INFORMATICS FOR STRUCTURAL GENOMICS<br />

Chair: Casimir Kulikowski, <strong>Rutgers</strong> University<br />

11:10 AM Helen Berman, <strong>Rutgers</strong> University <strong>and</strong> Protein Data Bank<br />

“The Protein Data Bank”<br />

11:50 AM Mark Gerstein, Yale University<br />

“Structural Genomics: Surveys of a Finite Parts List”<br />

PROTEIN PRODUCTION AND DRUG DESIGN<br />

Chair: Masayori Inouye, Robert Wood Johnson Medical School<br />

__________________________________________________________________________________<br />

<strong>Center</strong> <strong>for</strong> <strong>Advanced</strong> <strong>Biotechnology</strong> <strong>and</strong> <strong>Medicine</strong><br />

49


2:00 PM David Waugh, National Cancer Institute, Frederick<br />

“Generic Protein Expression <strong>and</strong> Purification Strategies <strong>for</strong> Structural<br />

Genomics”<br />

15 th Annual CABM Symposium<br />

2:40 PM Edward Arnold, <strong>Rutgers</strong> University <strong>and</strong> CABM<br />

“Optimization of Multiple Factors in Developing Potent HIV Reverse<br />

Transcriptase Inhibitors as Potential Anti-AIDS Drugs”<br />

3:40 PM Refreshment Break<br />

IDENTIFYING BIOCHEMICAL FUNCTIONS<br />

Chair: Gaetano Montelione, <strong>Rutgers</strong> University <strong>and</strong> CABM<br />

4:10 PM Jeffrey Skolnick, Dan<strong>for</strong>th Plant Science <strong>Center</strong><br />

“Prediction of Protein Structure <strong>and</strong> Function on a Genomic Scale”<br />

4:50 PM Ming-Ming Zhou, Mount Sinai School of <strong>Medicine</strong><br />

“Structure <strong>and</strong> Function of Protein Modular Domains”<br />

5:30 PM F. Raymond Salemme, 3-Dimensional Pharmaceuticals<br />

“Protein Thermal Stability Measurements <strong>for</strong> High-Throughput Drug<br />

Screening <strong>and</strong> Target Decryption”<br />

6:15 PM Reception<br />

Thursday, October 25, <strong>2001</strong><br />

8:00 AM Continental Breakfast<br />

HIGH-THROUGHPUT STRUCTURE DETERMINATION<br />

Chair: Ann Stock, Robert Wood Johnson Medical School <strong>and</strong> CABM<br />

9:00 AM Andrzej Joachimiak, Argonne National Laboratories<br />

“Structural Gemonics: from Genome Sequence to Proteome Structure”<br />

9:40 AM Stephen Burley, Rockefeller University<br />

“Structural Genomics of Sterol/Isoprenoid Biosynthesis”<br />

10:20 AM Clemens Anklin, Bruker NMR Instruments<br />

__________________________________________________________________________________<br />

<strong>Center</strong> <strong>for</strong> <strong>Advanced</strong> <strong>Biotechnology</strong> <strong>and</strong> <strong>Medicine</strong><br />

50


10:40 AM Refreshment Break<br />

“New Approaches to Biomolecular NMR Using Ultra High Field Systems<br />

<strong>and</strong> Cryoprobes”<br />

15 th Annual CABM Symposium<br />

STRUCTURAL BIOINFORMATICS<br />

Chair: Donna Bassolino, Bristol-Myers Squibb Pharmaceutical Research Institute<br />

11:10 AM Cyrus Chothia, Cambridge University<br />

“Protein Repertoires in Genomes: the Immunoglobulin Superfamily in<br />

Humans”<br />

11:50 AM Ronald Levy, <strong>Rutgers</strong> University<br />

“Recent Developments in Protein Fold Recognition, Determination, <strong>and</strong><br />

ab initio Prediction”<br />

12:30 PM Lunch<br />

NEW TARGETS FOR PHARMACEUTICAL DESIGN<br />

Chair: Stephen Anderson, GeneFormatics, Inc.<br />

2:00 PM Peter Lobel, Robert Wood Johnson Medical School <strong>and</strong> CABM<br />

“Lysosomal Proteomics <strong>and</strong> the Indentification of Human Disease Genes”<br />

2:40 PM Martin Rosenberg, GlaxoSmithKline<br />

“Microbial Genomics: Opportunities <strong>and</strong> Hurdles”<br />

3:20 PM John Chiplin, GeneFormatics, Inc.<br />

“Considerations <strong>for</strong> Target Selection <strong>and</strong> Drug Discovery in the<br />

Postgenomic Era”<br />

3:40 PM L. Patrick Gage, Wyeth Research<br />

“The Future of Molecular <strong>Medicine</strong>”<br />

Closing Remarks<br />

__________________________________________________________________________________<br />

<strong>Center</strong> <strong>for</strong> <strong>Advanced</strong> <strong>Biotechnology</strong> <strong>and</strong> <strong>Medicine</strong><br />

51


GRADUATE, UNDERGRADUATE STUDENTS AND<br />

POSTDOCTORAL FELLOWS INCLUDING<br />

COMPETITIVE FELLOWSHIP AWARDS<br />

CABM faculty members also participate in UMDNJ <strong>and</strong> <strong>Rutgers</strong> University graduate student rotation programs that<br />

place students in CABM labs each year in order to give them education <strong>and</strong> experience in a range of research<br />

methods <strong>and</strong> subjects.<br />

Graduate Students<br />

-Priti Bachhawat, Protein Crystallography<br />

-Scott Banta, Protein Engineering<br />

NIH Biotech Training Grant<br />

-Joseph Bauman, Biomolecular Crystallography<br />

-Annerban Bhattacharya, Protein NMR Spectroscopy<br />

-Wen-Feng Chen, Molecular Chronobiology<br />

-Natalia Denissova, Growth & Differentiation Control<br />

-Jui Dutta, Tumor Virology<br />

-Chaosu E, Mammalian Embryogenesis<br />

USAMRMC<br />

-Leonard Edelstein, Tumor Virology<br />

NIH Biotech Training Grant<br />

-John Everett, Protein NMR Spectroscopy<br />

NIH Biotech Training Grant<br />

-Jayita Guhaniyogi, Protein Crystallography<br />

-Gezhi Hu, Molecular Neurobiology<br />

American Heart Association<br />

-Hyuk Wan Ko, Molecular Chronobiology<br />

-Gregory Kornhaber, Protein NMR Spectroscopy<br />

-Lynn Lagos, Tumor Virology<br />

-Jung Eun Lee, Molecular Chronobiology<br />

-Wei Liu, Molecular Neurodevelopment<br />

-Paul F. Lizzul, Tumor Virology<br />

-Denise Perez, Viral Trans<strong>for</strong>mation<br />

NIH–NCI<br />

-Eduardo Perez, Protein Crystallography<br />

NIH Biochem Training Grant<br />

-Shahriar Pooyan, Protein Microchemistry<br />

-Yan Shen, Viral Trans<strong>for</strong>mation<br />

-Matthew Simmons, Tumor Virology<br />

NIH Biochem Training Gran<br />

- David Snyder, Protein NMR Spectroscopy<br />

NIH Biotech Training Grant<br />

__________________________________________________________________________________<br />

<strong>Center</strong> <strong>for</strong> <strong>Advanced</strong> <strong>Biotechnology</strong> <strong>and</strong> <strong>Medicine</strong><br />

52


-Priya Srinivasan, Molecular Virology<br />

-Stacey Stein, Molecular Neurobiology<br />

NJCCR<br />

-Junghan Suh, Viral Pathogenesis<br />

-Ramya Sundararajan, Viral Trans<strong>for</strong>mation<br />

-Alej<strong>and</strong>ro Toro, Protein Crystallography<br />

NIH Biophysics Training Grant<br />

-Paola Velasco, Biomolecular Crystallography<br />

-Gang Xiao, Protein Targeting<br />

-Cuifeng Yin, Protein NMR Spectroscopy<br />

-Guobao Zhang, Protein Microchemistry<br />

-Yujie Zhao, Viral Pathogenesis<br />

-Deyou Zheng, Protein NMR Spectroscopy<br />

Postdoctoral Students<br />

-Dr. Andrew Bendall, Molecular Neurobiology<br />

-Dr. David Buckler, Protein Crystallography<br />

-Dr. Rajula Bhatia-Gaur, Molecular Neurobiology<br />

Neuroscience & Cell Biology NIH Training Grant<br />

UMDNJ Foundation<br />

-Dr. Andrea Cuconati, Viral Trans<strong>for</strong>mation<br />

Howard Hughes Medical Institute<br />

-Dr. Kalyan Das, Biomolecular Crystallography<br />

-Dr. Kurt Degenhardt, Viral Trans<strong>for</strong>mation<br />

-Dr. Jianping Ding, Biomolecular Crystallography<br />

-Dr. Jixiang Ding, Mammalian Embryogenesis<br />

-Dr. Bonnie Lynn Dixon, Protein Engineering<br />

& Protein NMR Spectroscopy<br />

-Dr. Yongjun Fan, Tumor Virology<br />

-Dr. Kristin Gunsalus, Protein NMR Spectroscopy<br />

NIH Postdoctoral Fellowship<br />

-Dr. Daniel Himmel, Biomolecular Crystallography<br />

-Dr. Holly Henry, Viral Trans<strong>for</strong>mation<br />

Howard Hughes Medical Institute<br />

-Dr. Jiu-Zhen Jin, Protein Crystallography<br />

Howard Hughes Medical Institute<br />

-Dr. Natalia Khlebtsova, Protein Crystallography<br />

Howard Hughes Medical Institute<br />

-Dr. Eun Young Kim, Molecular Chronobiology<br />

-Dr. Minjung Kim, Molecular Neurobiology<br />

-Dr. Jerome Kucharczak, Tumor Virology<br />

-Dr. Hansol Lee, Molecular Neurobiology<br />

-Dr. Hsin-Ching Lin, Viral Pathogenesis<br />

Graduate <strong>and</strong> Postdoctoral Students<br />

__________________________________________________________________________________<br />

<strong>Center</strong> <strong>for</strong> <strong>Advanced</strong> <strong>Biotechnology</strong> <strong>and</strong> <strong>Medicine</strong><br />

53


-Dr. Li Lin, Protein Targeting<br />

-Dr. Heng-Ling Liou, Protein Targeting<br />

-Dr. Yuanpeng Huang, Protein NMR Spectroscopy<br />

-Dr. Jan-Jan Liu, Mammalian Embrogenesis<br />

-Dr. Jianyin Long, Growth & Differential Control<br />

-Dr. Xuejun Ma, Biomolecular Crystallography<br />

-Dr. Isao Matsuura, Growth & Differential Control<br />

-Dr. Daniel Monleon, Protein NMR Spectroscopy<br />

-Dr. Hunter Moseley, Protein NMR Spectroscopy<br />

-Dr. Deena Oren, Biomolecular Crystallography<br />

-Dr. Rajan Paranji, Protein NMR Spectroscopy<br />

-Dr. Beatrice Rayet, Tumor Virology<br />

Cure <strong>for</strong> Lymphoma Foundation Fellowship <strong>and</strong> UMDNJ Foundation<br />

-Dr. Victoria Robinson, Protein Crystallography<br />

Howard Hughes Medical Institute<br />

-Dr. Stefan Sarafianos, Biomolecular Crystallography<br />

-Dr. Anju Thomas, Viral Trans<strong>for</strong>mation<br />

-Dr. Steven Tuske, Biomolecular Crystallography<br />

NIH Postdoctoral Fellowship<br />

-Dr. Yingxia Wen, Molecular Virology<br />

-Dr. Yu-Ting Yan, Mammalian Embryogenesis<br />

Undergraduate Students<br />

-Alan Anschel, Viral Trans<strong>for</strong>mation<br />

Howard Hughes Medical Institute<br />

-Antonious Attallah, Protein Crustallography<br />

-Jisook Baek, Viral Trans<strong>for</strong>mation<br />

-Angelo Banaria, Lab Management<br />

-Marvin Bayro, Protein NMR Spectroscopy<br />

-Michael Chen, Protein Crystallography<br />

-Helen Chow, Protein NMR Spectroscopy<br />

-Bonnie Cooper, Protein NMR Spectroscopy<br />

-Gaurev Davé, Biomolecular Crystallography<br />

-Michelle DeFreese, Lab Management<br />

-Kate Drahos, Protein NMR Spectroscopy<br />

-Robert Faltas, Biomolecular Crystallography<br />

-Manju Goyal, Protein NMR Spectroscopy<br />

-April Graham, Molecular Neurobiology<br />

Biomedical Careers Fellowship<br />

-Michelle Hickey, Viral Pathogenesis<br />

-Hui Yun Ho, Administration<br />

-Lydia Hsu, Viral Pathogenesis<br />

Graduate <strong>and</strong> Postdoctoral Students<br />

__________________________________________________________________________________<br />

<strong>Center</strong> <strong>for</strong> <strong>Advanced</strong> <strong>Biotechnology</strong> <strong>and</strong> <strong>Medicine</strong><br />

54


-Hyunmi Kim, Molecular Virology<br />

-Rebecca Liu, Protein NMR Spectroscopy<br />

-Nicholas Lumia, Lab Management<br />

-Meera Mani, Protein Crystallography<br />

-Ram Mani, Protein NMR Spectroscopy<br />

Howard Hughes Medical Institute Fellowship<br />

<strong>Rutgers</strong> University Undergraduate Research Fellowship<br />

-Raehum Paik, Molecular Chronobiology<br />

-Hammad Rizvi, Mammalian Embryogenesis<br />

-Gurmukh Sahota, Protein NMR Spectroscopy<br />

-Beth Shafer, Molecular Virology<br />

-Rachel Siegel, Biomolecular Crystallography<br />

Howard Hughes Medical Institute Fellowship<br />

-Jennifer Staley, Administration<br />

-Max Tischfield, Developmental Neurogenetics<br />

-Melanie Vishnu, Lab Management<br />

-Deborah Weintraub, Protein Crystallography<br />

-Karlvin Wong, Molecular Neurobiology<br />

-Zeba Wunderlich, Protein NMR Spectroscopy<br />

Undergraduate Students<br />

__________________________________________________________________________________<br />

<strong>Center</strong> <strong>for</strong> <strong>Advanced</strong> <strong>Biotechnology</strong> <strong>and</strong> <strong>Medicine</strong><br />

55


PATENTS<br />

Abate-Shen, C., Shen, M. M. <strong>and</strong> Gridley, T. “Roles <strong>for</strong> Nkx3.1 in prostate development <strong>and</strong><br />

cancer”. U.S. Patent Application 09/756,151 (pending).<br />

Abate-Shen, C., Shen, M. M. <strong>and</strong> Kim, M. “Monoclonal antibody <strong>for</strong> NKX3.1 <strong>and</strong> method <strong>for</strong><br />

detecting same”. U.S. Patent Application 09/853,121 (pending).<br />

Arnold, E., Kenyon, G.L., Stauber, M., Maurer, K., Eargle, D., Muscate, A., Leavitt, A., Roe,<br />

D.C., Ewing, T.J.A., Skillman, A.G., Jr., Kuntz, I.D. <strong>and</strong> M. Young. “Naphthols useful in<br />

antiviral methods.” U.S. Patent #6,140, 368.<br />

Edery, I., Altman, M. <strong>and</strong> N. Sonenberg. “Isolation of full-length cDNA clones <strong>and</strong> capped<br />

mRNA”. U.S. Patent #5219989.<br />

Lobel, P. <strong>and</strong> Sleat, D “Human lysosomal protein <strong>and</strong> methods of its use” U.S.<br />

Patent #6,302,685.<br />

Shatkin, A.J., Pillutla, R., Reinberg, D., Maldonado, E., <strong>and</strong> Z. Yue. “mRNA capping enzymes<br />

<strong>and</strong> uses thereof”. U.S. Patent # 6,312,926 B1.<br />

Stein, S. “Site-specific 13C-enriched reagents <strong>for</strong> diagnostic medicine by magnetic resonance<br />

imaging”. U.S. Patent #6,210,655.<br />

Stein, S., Leibowitz, M. J. <strong>and</strong> P.J. Sinko. “Carrier <strong>for</strong> in vivo delivery of a therapeutic agent”.<br />

U.S. Patent #6,258,774.<br />

Strair, R., Rabson, A.B., Medina, D. <strong>and</strong> White E. "Cytotoxic agents <strong>for</strong> the selective killing of<br />

lymphoma <strong>and</strong> leukemia cells <strong>and</strong> methods of use thereof" PCT Application<br />

PCT/US99/09793.<br />

White, E., Degenhardt, K., Nelson, D. <strong>and</strong> Cuconati, A. "Trans<strong>for</strong>med epithelial baby mouse<br />

kidney cell lines (BMK) with targeted gene disruptions in Bax, Bak <strong>and</strong> Bax plus Bak”.<br />

Provisional Patent App. 60/323,392.<br />

Sabatini, P., Sherwood, A., Black, I. <strong>and</strong> White, E. "Immortalized <strong>and</strong> trans<strong>for</strong>med astrocytederived<br />

cells <strong>for</strong> treatment <strong>and</strong> diagnosis of brain disease" Utility App filed 1996.<br />

__________________________________________________________________________________<br />

<strong>Center</strong> <strong>for</strong> <strong>Advanced</strong> <strong>Biotechnology</strong> <strong>and</strong> <strong>Medicine</strong><br />

56


Thomas, A., White, E., Goyal, L. <strong>and</strong> Kasof, G. "Recombinant cell line <strong>and</strong> screening method<br />

<strong>for</strong> identifying agents which regulate apoptosis <strong>and</strong> tumor suppression" 2 provisionals filed <strong>and</strong><br />

US Utility filed 5/6/99 Application 09/674,876.<br />

Fiscal In<strong>for</strong>mation<br />

__________________________________________________________________________________<br />

<strong>Center</strong> <strong>for</strong> <strong>Advanced</strong> <strong>Biotechnology</strong> <strong>and</strong> <strong>Medicine</strong><br />

57

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

Saved successfully!

Ooh no, something went wrong!