26.11.2012 Views

Annual Report 2011 - Center for Advanced Biotechnology and ...

Annual Report 2011 - Center for Advanced Biotechnology and ...

Annual Report 2011 - Center for Advanced Biotechnology and ...

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.

CENTER FOR ADVANCED<br />

BIOTECHNOLOGY AND MEDICINE<br />

<strong>Annual</strong> <strong>Report</strong> <strong>2011</strong>


CONTENTS<br />

Director’s Overview 3<br />

Research Programs <strong>and</strong> Laboratories 6<br />

Cell & Developmental Biology 7<br />

Molecular Chronobiology Isaac Edery 8<br />

Tumor Virology Céline Gélinas 11<br />

Bacterial Physiology <strong>and</strong> Protein Engineering Masayori Inouye 15<br />

Growth <strong>and</strong> Differentiation Control Fang Liu 19<br />

Protein Targeting Peter Lobel 22<br />

Developmental Neurogenetics James Millonig 25<br />

Viral Pathogenesis Arnold Rabson 29<br />

Molecular Virology Aaron Shatkin 32<br />

Molecular Neurodevelopment Mengqing Xiang 35<br />

Structural Biology 39<br />

Protein Engineering Stephen Anderson 40<br />

Biomolecular Crystallography Eddy Arnold 42<br />

Structural Biology <strong>and</strong> Bioin<strong>for</strong>matics Helen Berman 47<br />

Structural Microbiology Joseph Marcotrigiano 51<br />

Structural Bioin<strong>for</strong>matics Gaetano Montelione 54<br />

Protein Design <strong>and</strong> Evolution Vikas N<strong>and</strong>a 62<br />

Protein Crystallography Ann Stock 65<br />

Education, Training <strong>and</strong> Technology Transfer 68<br />

Lectures <strong>and</strong> Seminars 69<br />

CABM Lecture Series 70<br />

<strong>Annual</strong> Retreat 71<br />

25 th <strong>Annual</strong> Symposium 73<br />

Undergraduate Students 76<br />

Administrative <strong>and</strong> Support Staff 77<br />

Patents 78<br />

Scientific Advisory Board 81<br />

Current Members 82<br />

Past Members 84<br />

Fiscal In<strong>for</strong>mation 86<br />

Grants <strong>and</strong> Contracts 87<br />

2


Director’s Overview<br />

CABM—25 <strong>and</strong> counting.<br />

CABM experienced a momentous <strong>2011</strong>. It was remarkable as our 25 th anniversary year. Exciting<br />

events to celebrate this milestone included a festive dinner attended by nearly 200 <strong>for</strong>mer students<br />

<strong>and</strong> postdoctoral fellows who returned to CABM from around the country, Europe <strong>and</strong> Asia. This<br />

enjoyable reunion was held on October 20 th . Announced at this event was the establishment of the<br />

Aaron J. Shatkin Endowed Lectureship which will start in the spring of 2012 <strong>and</strong> the naming of the<br />

CABM south atrium as the Shatkin Atrium – two overwhelming <strong>and</strong> highly appreciated honors.<br />

The CABM <strong>Annual</strong> Symposium was held on the next day. The meeting attracted more than 450<br />

participants <strong>and</strong> featured lectures by current <strong>and</strong> <strong>for</strong>mer CABM Scientific Advisory Board members<br />

Bruce Alberts, Wayne Hendrickson, Robert Roeder <strong>and</strong> Phillip Sharp. In addition, several CABM<br />

alumni from academia <strong>and</strong> industry gave talks, <strong>and</strong> many others described recent their recent work<br />

in poster presentations.<br />

Continuing the CABM Mission<br />

The year was also noteworthy <strong>for</strong> the continued commitment to excellence of CABM faculty <strong>and</strong><br />

students. Their research success made possible the vigorous pursuit of our mission--the<br />

advancement of basic knowledge to improve human health. CABM research programs were<br />

supported by NIH <strong>and</strong> several additional federal <strong>and</strong> state granting agencies as well as other public<br />

<strong>and</strong> private sources. The resulting research advanced our underst<strong>and</strong>ing of cancer, infectious<br />

diseases <strong>and</strong> hereditary <strong>and</strong> developmental disorders. As described in the individual research<br />

summaries, CABM researchers are collaborating with clinical investigators at The Cancer Institute<br />

of New Jersey to elicit new targets <strong>and</strong> to develop therapies <strong>for</strong> non-Hodgkins lymphomas <strong>and</strong><br />

metastatic breast cancers as well as to design novel proteins <strong>for</strong> therapeutic use in cancer <strong>and</strong> other<br />

devastating diseases. Several CABM groups have gained new insights <strong>and</strong> found clever approaches<br />

to answer the health challenges of infections by antibiotic resistant bacteria, HIV/AIDS, hepatitis<br />

<strong>and</strong> influenza viruses. Other groups have used model systems to define a temperature dependent<br />

molecular mechanism <strong>for</strong> regulating biological clocks, uncover an autism susceptibility gene <strong>and</strong><br />

decipher the differentiation cascade of retinal cells essential <strong>for</strong> vision. Another CABM laboratory<br />

has discovered the molecular basis of Batten Disease, a lethal childhood hereditary disorder that<br />

may become reversible <strong>and</strong> even curable based on this finding <strong>and</strong> developing technology licensed<br />

to a biotechnology company.<br />

Fostering the Growth of Structural Biology<br />

Another stellar event in <strong>2011</strong> has been the completion by Rutgers University of an impressive<br />

<strong>Center</strong> <strong>for</strong> Integrative Proteomics Research. The new building is physically connected to CABM<br />

<strong>and</strong> will accommodate computational research <strong>and</strong> training activities including the Biomaps<br />

Institute, high field NMR groups, the CABM Mass Spectrometry core facility, a cryoEM laboratory<br />

<strong>and</strong> the international Protein Data Bank (PDB). CABM had the pleasure of hosting the PDB group<br />

this year while the proteomics building was under construction. This proved to be an enjoyable<br />

arrangement that led to productive interactions <strong>and</strong> mutually beneficial collaborations, notably<br />

between PDB <strong>and</strong> the Northeast Structural Genomics Consortium, a multi-institutional center led<br />

by a CABM group <strong>and</strong> sponsored by NIH as part of the Protein Structural Initiative (PSI).<br />

3


Teaching in the Classroom <strong>and</strong> Laboratory<br />

All CABM faculty participate actively in the teaching of students <strong>and</strong> postdoctoral fellows both in<br />

the lecture hall <strong>and</strong> at the laboratory bench. Trainees include undergraduates, graduate students<br />

from many different MS <strong>and</strong> PhD programs <strong>and</strong> medical students. The NIH <strong>Biotechnology</strong><br />

Program has been an excellent source of training <strong>and</strong> support <strong>for</strong> PhD students from various<br />

disciplines <strong>for</strong> more than 20 years. In recognition of its success, the training program was recently<br />

renewed <strong>for</strong> another five years. Outreach education programs also introduce scientific research to<br />

highly motivated high school students at local schools through SMART Team <strong>and</strong> Biolinks<br />

programs. In addition, CABM hosts a Summer Undergraduate Research Experience (SURE) that is<br />

now in its 8th year. In <strong>2011</strong> SURE students from Rutgers <strong>and</strong> several other leading universities<br />

participated in lectures <strong>and</strong> lab projects mentored by CABM graduate students <strong>and</strong> postdoctoral<br />

fellows—providing opportunities <strong>for</strong> both budding <strong>and</strong> committed scientists to interact. Other<br />

outreach programs include a seminar series that brings visiting scientists to talk about their work<br />

<strong>and</strong> to meet with trainees <strong>and</strong> the annual CABM Symposium each year on a topic of special interest.<br />

The next Symposium, our 26 th , will be held in October, 2012.<br />

Awards <strong>and</strong> Service<br />

CABM faculty have received significant national honors <strong>for</strong> their scientific contributions <strong>and</strong><br />

service. These include elected fellowship in AAAS, the American Academy of Microbiology, the<br />

American Academy of Arts & Sciences <strong>and</strong> the US National Academy of Sciences. Drs. Stock <strong>and</strong><br />

Arnold are recipients of MERIT awards from NIH, <strong>and</strong> Drs. Gelinas, Millonig <strong>and</strong> Rabson are NIH<br />

study section members. Dr. Stock recently completed service as Chair of the Board of Scientific<br />

Counselors <strong>for</strong> NIH-NIDCR <strong>and</strong> is completing a long-held position as HHMI investigator. After<br />

completing service on the American Society of Biochemistry & Molecular Biology Council, she<br />

was appointed this year to the Society’s Education & Professional Development <strong>and</strong> Finance<br />

Committees. Dr. Montelione is advisor to the NSF Committee of Visitors. Many faculty serve on<br />

other national committees <strong>and</strong> in journal editorial positions. At the local level, Dr. Millonig recently<br />

joined Drs. Gelinas <strong>and</strong> Stock as Master Educators. In addition, Dr. Gelinas serves as Associate<br />

Dean <strong>for</strong> Research, <strong>and</strong> Dr. Millonig leads the RWJMS/Rutgers/Princeton joint MD/PhD program.<br />

This past year Dr. Rabson was appointed director of the Child Health Institute of New Jersey.<br />

Financial Support<br />

Advancing science through research requires strong support from many sources, administrative as<br />

well as financial. CABM is jointly administered by the University of Medicine <strong>and</strong> Dentistry of<br />

New Jersey-Robert Wood Johnson Medical School <strong>and</strong> Rutgers, the State University of New Jersey.<br />

We thank the Universities, in particular <strong>for</strong> providing a wonderfully functional building <strong>for</strong> doing<br />

science. CABM also benefits from the advice, wisdom <strong>and</strong> guidance freely given by a Scientific<br />

Advisory Board composed of leaders from academia <strong>and</strong> industry. We acknowledge with gratitude<br />

their help <strong>and</strong> support on many issues. Despite difficult economic times, CABM faculty were<br />

successful again in obtaining grants <strong>and</strong> contracts—nearly $18 million in <strong>2011</strong>. Since the founding<br />

of CABM, a total of ~$300 million has been awarded, mostly <strong>for</strong> research but also <strong>for</strong> education<br />

<strong>and</strong> service functions that have had positive impacts on the two universities <strong>and</strong> stimulated the NJ<br />

economy. We are indebted to the numerous public agencies, private foundations, companies <strong>and</strong><br />

individuals <strong>for</strong> their generous <strong>and</strong> essential financial assistance in our pursuit of excellence at<br />

CABM <strong>for</strong> the past 25 years. The sources of federal funding notably include the National Institutes<br />

4


of Health, National Science Foundation, the Department of Defense <strong>and</strong>, at the state level, The NJ<br />

Commission on Spinal Cord Research, NJ Commission on Cancer Research, <strong>and</strong> the Governor’s<br />

Council <strong>for</strong> Medical Research <strong>and</strong> Treatment of Autism. The Howard Hughes Medical Institute has<br />

been supportive of CABM <strong>for</strong> nearly 20 years, <strong>and</strong> the Ara Parseghian Medical Research<br />

Foundation <strong>and</strong> Batten Disease Support <strong>and</strong> Research Association provided important help. CABM<br />

is very grateful to numerous companies including Johnson & Johnson, Hoffmann-LaRoche, Sanofi-<br />

Aventis, Pfizer, Takara Bio Inc., Cisco Inc., Nexomics Biosciences Inc., BioMarin Pharmaceuticals,<br />

Amicus Therapeutics, LifeGas, CIL, GenScript, Macrogen, New Engl<strong>and</strong> BioLabs, Taxis<br />

Pharmaceuticals Inc., Rigaku <strong>and</strong> Genewiz. Without this combined financial help, CABM could<br />

not have maintained its commitment to excellence in advancing basic knowledge in the life sciences<br />

to improve human health. We are most grateful.<br />

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

shatkin@cabm.rutgers.edu<br />

http://www.cabm.rutgers.edu<br />

5


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

Laboratories<br />

6


Cell & Developmental Biology<br />

Laboratory Faculty Director<br />

Molecular Chronobiology Isaac Edery 8<br />

Tumor Virology Céline Gélinas 11<br />

Bacterial Physiology <strong>and</strong> Protein Engineering Masayori Inouye 15<br />

Growth <strong>and</strong> Differentiation Control Fang Liu 19<br />

Protein Targeting Peter Lobel 22<br />

Developmental Neurogenetics James Millonig 25<br />

Viral Pathogenesis Arnold Rabson 29<br />

Molecular Virology Aaron Shatkin 32<br />

Molecular Neurodevelopment Mengqing Xiang 35<br />

7


Dr. Isaac Edery<br />

CABM Resident Faculty Member<br />

Professor<br />

Department of Molecular Biology<br />

<strong>and</strong> Biochemistry<br />

Rutgers University<br />

Dr. Rudra Dubey<br />

Asst. Research Professor<br />

Dr. Weihuan Cao<br />

Research Assistant<br />

Dr. Kwang Huei Low<br />

Research Assistant<br />

Evrim Yildirim<br />

Graduate Student<br />

Chhaya Dharia<br />

Lab Researcher<br />

Molecular Chronobiology Laboratory<br />

Dr. Isaac Edery completed his doctoral studies in biochemistry as a Royal<br />

Canadian Cancer Research Fellow under Dr. Nahum Sonenberg at McGill<br />

University in Montreal. His Ph.D. research focused on the role of the<br />

eukaryotic mRNA cap structure during protein synthesis <strong>and</strong> precursor<br />

mRNA splicing. Subsequently, he was in the laboratory of Dr. Michael<br />

Rosbash at Br<strong>and</strong>eis University where he pursued postdoctoral studies<br />

aimed at underst<strong>and</strong>ing the time-keeping mechanism underlying circadian<br />

clocks. He joined CABM in 1993, <strong>and</strong> his research is supported by NIH. Dr.<br />

Edery is a member of the editorial board of Chronobiology International <strong>and</strong><br />

Journal of Biological Rhythms.<br />

The main focus of the Edery lab is to underst<strong>and</strong> the biochemical <strong>and</strong> cellular bases<br />

underlying circadian (~24 hr) rhythms, using Drosophila as a model system. Our<br />

strength lies in applying biochemical strategies that are integrated with genetic,<br />

ecological <strong>and</strong> evolutionary perspectives to underst<strong>and</strong> how circadian clocks<br />

function <strong>and</strong> regulate animal physiology <strong>and</strong> behavior. Circadian rhythms are driven<br />

by cellular clocks <strong>and</strong> enable organisms to anticipate daily <strong>and</strong> seasonal changes in<br />

environmental conditions, ensuring activities occur at optimal times in the day <strong>and</strong><br />

appropriate seasonal responses are elicited. Underst<strong>and</strong>ing how clocks function is<br />

highly relevant <strong>for</strong> human health <strong>and</strong> well-being. Indeed, clock malfunctions in<br />

humans have been implicated in many diseases, including manic-depression, cancer,<br />

sleep problems <strong>and</strong> metabolic syndromes. Our lab is investigating the core clock<br />

mechanism, identifying novel clock proteins <strong>and</strong> how they interact to build a<br />

biological time-keeping device that is synchronized to local time. In a second major<br />

ef<strong>for</strong>t, we are determining the role of clocks in seasonal adaptation, which has led to<br />

novel ecological <strong>and</strong> evolutionary implications. These studies have also broadened<br />

our research scope by revealing mechanisms that regulate sleep <strong>and</strong> arousal. In<br />

addition, we are investigating how environmental stimuli, most notably temperature,<br />

controls global gene expression. Below is a brief summary of some of our ongoing<br />

research <strong>and</strong> future directions<br />

Mechanisms underlying circadian clocks: The central clock mechanism is a<br />

dynamically changing multi-subunit biochemical oscillator based on a small set of<br />

interacting core “clock” proteins <strong>and</strong> auxiliary factors that as a unit generates a selfperpetuating<br />

daily transcriptional feedback loop that also drives global cyclical gene<br />

expression, which underlies many of the daily rhythms manifested by organisms. In<br />

addition, to the transcriptional architecture, numerous post-translational regulatory<br />

pathways, most notably reversible phosphorylation, regulate various aspects of clock<br />

8


protein metabolism/activity, such that the biochemical oscillator has an endogenous<br />

period of ~24 hr. A conserved feature of animal clocks is that the key clock protein<br />

termed PERIOD (PER) undergoes daily rhythms in phosphorylation that are central<br />

to normal clock progression in animals.<br />

We showed that phosphorylated PER is recognized by the F-box protein, SLIMB<br />

(homolog of β-TrCP) <strong>and</strong> targeted to the 26S proteasome <strong>for</strong> rapid degradation (Ko et<br />

al., 2002, Nature), a key event in stimulating the next round of daily gene expression.<br />

We used mass spectrometry <strong>and</strong> phospho-specific antibodies to identify the critical<br />

phosphorylation events that promote the binding of SLIMB to PER (Chiu et al., 2008,<br />

Genes & Development). Converting key phospho-sites to Ala or Asp (phosphomimetic)<br />

led to decreases or increases in the pace of the clock, respectively (Chiu et<br />

al., 2008, Genes & Development). In more recent work we identified a novel clock<br />

kinase, NEMO, <strong>and</strong> showed that different phospho-clusters have distinct functions<br />

(Chiu et al., <strong>2011</strong>, Cell). Ongoing work is aimed at identifying the roles of different<br />

phospho-clusters on PER <strong>and</strong> the relevant kinases <strong>and</strong> phosphatases. We are also<br />

applying these techniques to other clock proteins, <strong>and</strong> investigating the effects of other<br />

post-translational modifications in clock function, such as SUMOylation <strong>and</strong><br />

acetylation. Moreover, PER proteins not only function as the principle biochemical<br />

timer but also link to gene expression by acting as transcriptional repressors, whereby<br />

they “deliver” factors such as chromatin remodeling factors to central clock<br />

transcription factors, yielding cyclical gene expression <strong>and</strong> hence rhythmic physiology<br />

<strong>and</strong> behavior. We are undertaking proteomic strategies to identify native clock<br />

protein complexes <strong>and</strong> how they change throughout a daily cycle. In related work we<br />

are investigating the roles of micro RNAs (miRNAs) in clock function.<br />

Seasonal <strong>and</strong> thermal adaptation: Many animals exhibit a bimodal distribution of<br />

activity, with ‘morning’ <strong>and</strong> ‘evening’ bouts of activity that are separated by a midday<br />

dip in activity or ‘siesta’. Ambient temperature is a key environmental modality<br />

regulating the daily distribution of activity in animals. In D. melanogaster, as<br />

temperatures rise there is less midday activity <strong>and</strong> the two bouts of activity are<br />

increasingly shifted into the cooler nighttime hours, almost certainly an adaptive<br />

response that minimizes the detrimental effects of the hot midday sun (Majercak et al.,<br />

1999, Neuron). We showed that the temperature-dependent splicing of the 3'-terminal<br />

intron (termed dmpi8) from the D. melanogaster per RNA is a major ‘thermosensor’<br />

that adjusts the distribution of daily wake-sleep cycles, eliciting seasonably<br />

appropriate responses. In more recent work we showed that this mechanism does not<br />

operate in several Drosophila species with more restricted <strong>and</strong> ancestral locations in<br />

equatorial Africa wherein temperature <strong>and</strong> daylength do not show large seasonal<br />

variations (Low et al., 2008, Neuron). We investigated the molecular basis <strong>for</strong> the<br />

species-specific splicing phenotypes <strong>and</strong> found that multiple suboptimal splicing<br />

signals on dmpi8 underlie the thermosensitivity (Low et al., 2008, Neuron).<br />

Presumably, higher temperatures progressively destabilize interactions between the<br />

non-consensus 5’ splice site (ss) <strong>and</strong> the U1 snRNP, the initial step in the splicing<br />

reaction. Ongoing work is aimed at underst<strong>and</strong>ing how temperature regulates global<br />

gene expression. In addition, these studies are revealing new mechanisms that<br />

regulate sleep <strong>and</strong> arousal.<br />

9


Publications (2010-<strong>2011</strong>):<br />

Ko, H.W., Chiu, J., Vanselow, J.T., Kramer, A. <strong>and</strong> Edery, I. A hierarchical<br />

phosphorylation cascade that regulates the timing of PERIOD nuclear entry reveals novel<br />

roles <strong>for</strong> proline-directed kinases <strong>and</strong> GSK-3β/SGG in circadian clocks. J. Neurosci.<br />

2010 30: 12664-75.<br />

Chiu, J.C., Low, K.H., Pike, D.H., Yildirim, E. <strong>and</strong> Edery, I. Assaying locomotor activity<br />

to study circadian rhythms <strong>and</strong> sleep parameters in Drosophila. J. Visualized Expts.<br />

2010 43.<br />

Sun, W.C., Jeong, E.H., Jeong, H.J., Ko, H.W., Edery, I. <strong>and</strong> Kim, E.Y. Two distinct<br />

modes of PERIOD recruitment onto dCLOCK reveal a novel role <strong>for</strong> TIMELESS in<br />

circadian transcription J. Neurosci. 2010 43: 14458-69.<br />

Edery, I. Circadian rhythms. Temperatures to communicate by. Science 2010 330: 329-<br />

30.<br />

Chiu, J.C. Ko, H.W. <strong>and</strong> Edery, I. NEMO/NLK phosphorylates PERIOD to initiate a<br />

time-delay phosphorylation circuit that sets circadian clock speed. Cell <strong>2011</strong> 145: 357-<br />

70.<br />

Edery, I. A master CLOCK hard at work brings rhythm to the transcriptome. Genes &<br />

Dev. <strong>2011</strong> 25 (in press).<br />

Edery, I. A morning induced phosphorylation-based repressor times evening gene<br />

expression: a new way <strong>for</strong> circadian clocks to use an old trick. Mol. Cell. <strong>2011</strong> (in press).<br />

10


Dr. Céline Gélinas<br />

CABM Resident Faculty Member<br />

Professor<br />

Department of Biochemistry<br />

Associate Dean <strong>for</strong> Research<br />

UMDNJ-Robert Wood Johnson<br />

Medical School<br />

Dr. Sheng Ge<br />

Research Associate<br />

Dr. Poonam Molli<br />

Research Associate<br />

Dr. Sophie Richard-<br />

Bail<br />

Research Associate<br />

Dr. Anitha Suram<br />

Research Associate<br />

Na Yang<br />

Graduate Student<br />

Tumor Virology Laboratory<br />

Dr. Céline Gélinas came to CABM in September 1988 from the University of<br />

Wisconsin, where she conducted postdoctoral studies with Nobel laureate Dr.<br />

Howard M. Temin. She earned her Ph.D. at the Université de Sherbrooke in<br />

her native country, Canada, <strong>and</strong> received a number of honors including the<br />

Jean-Marie Beauregard Award <strong>for</strong> Academic <strong>and</strong> Research Excellence, the<br />

National Cancer Institute of Canada King George V Silver Jubilee<br />

Postdoctoral Fellowship, a Medical Research Council of Canada Postdoctoral<br />

Fellowship <strong>and</strong> a Basil O’Connor Starter Scholar Research Award. Her<br />

research is funded by the NIH, the Leukemia <strong>and</strong> Lymphoma Society <strong>and</strong> the<br />

Department of Defense Breast Cancer Research Program. Dr. Gélinas served<br />

<strong>for</strong> several years as a member of the NIH Experimental Virology Study Section<br />

<strong>and</strong> the Virology B Study Section. In 2006, she was elected to the UMDNJ<br />

Stuart D. Cook Master Educators Guild. Dr. Gelinas was appointed Associate<br />

Dean <strong>for</strong> Research <strong>for</strong> RWJMS in 2008. She was elected to Fellowship in the<br />

American Academy of Microbiology in 2010.<br />

Our laboratory focuses on the Rel/NF-kB signaling pathway <strong>and</strong> its role in cell<br />

survival <strong>and</strong> cancer. The Rel/NF-kB transcription factors are key to normal immune<br />

<strong>and</strong> inflammatory responses <strong>and</strong> play critical roles in tumor cell survival,<br />

pathogenesis <strong>and</strong> chemoresistance. They are thus important targets <strong>for</strong> therapy.<br />

Rel/NF-kB is constitutively activated in many human cancers, <strong>and</strong> the Rel proteins<br />

are implicated in leukemia/lymphomagenesis <strong>and</strong> in breast cancer. However, the<br />

detailed mechanism is not fully understood. Our studies aim at underst<strong>and</strong>ing how<br />

changes in NF-kB’s transcriptional activity <strong>and</strong> regulation contribute to<br />

carcinogenesis <strong>and</strong> tumor cell resistance to anti-cancer treatment.<br />

We published a study demonstrating that defective ubiquitin-proteasome mediated<br />

turnover of the NF-kB-regulated apoptosis inhibitor Bfl-1 (Bcl2A1, A1) can<br />

predispose to lymphoma (Fan et al. 2010). Ubiquitination-resistant mutants of Bfl-1<br />

showed decreased turnover <strong>and</strong> greatly accelerated tumor <strong>for</strong>mation in a mouse<br />

model of leukemia/lymphoma. We also showed that these tumors also displayed<br />

upregulation <strong>and</strong> activation of tyrosine kinase Lck along with activation of the IKK,<br />

Akt <strong>and</strong> Erk signaling pathways, which are key mediators in cancer. Coexpression of<br />

Bfl-1 <strong>and</strong> constitutively active Lck promoted tumor <strong>for</strong>mation, whereas Lck<br />

knockdown in tumor-derived cells suppressed leukemia/lymphomagenesis. Since<br />

Bfl-1 is overexpressed in many therapy-resistant leukemia <strong>and</strong> lymphomas <strong>and</strong> is<br />

necessary <strong>for</strong> tumor cell maintenance <strong>and</strong> chemoresistance, these data suggest that<br />

ubiquitination is an important tumor suppression mechanism to regulate Bfl-1<br />

function. This also raises the possibility that mutations in bfl-1 or in the signaling<br />

pathways that control its ubiquitination may predispose to cancer. This work was<br />

conducted together with our colleagues, Drs. Eileen White (CINJ <strong>and</strong> Rutgers<br />

Univ.), Yacov Ron (RWJMS) <strong>and</strong> David Weissmann (RWJMS, RWJUH).<br />

11


The findings above suggested that accelerating Bfl-1 turnover could perhaps be<br />

used to improve tumor cell response to anti-cancer therapy. We pursued our<br />

investigation of kinase-specific inhibitors that can accelerate Bfl-1 turnover <strong>and</strong><br />

showed that some of them could significantly sensitize drug-resistant human<br />

leukemia <strong>and</strong> lymphoma cell lines to anti-cancer agents. Ongoing studies are<br />

evaluating the effectiveness of combination treatment in vivo in mouse xenograft<br />

models <strong>and</strong> in patient-derived specimens ex vivo together with our Drs. Roger<br />

Strair, Daniel Medina, Lauri Goodell <strong>and</strong> Susan Goodin (CINJ).<br />

Our ef<strong>for</strong>ts looking into the mechanisms that regulate Bfl-1 turnover <strong>and</strong> function<br />

revealed that while Bfl-1 is regulated by ubiquitination, it is not a substrate <strong>for</strong><br />

sumoylation. This indicated that ubiquitination is the primary mode of posttranslational<br />

regulation to control its half-life. We identified specific mutations in<br />

putative Bfl-1 phosphorylation sites that markedly alter its turnover <strong>and</strong> have been<br />

investigating if these are substrates <strong>for</strong> phosphorylation. We also identified a<br />

naturally occurring gene mutation in human lymphoma cells that alters turnover<br />

of apoptosis inhibitor Bfl-1 <strong>and</strong> chemoresistance, <strong>and</strong> have optimized conditions<br />

to purify Bfl-1 binding partners with the ultimate goal to identify factors that<br />

control its turnover.<br />

Collaborative studies with the group of Dr. Gaetano Montelione (CABM)<br />

elucidated the structure of Bfl-1∆C (residues 1-151) bound to a Noxa BH3<br />

peptide, refined to 2.25 Å resolution. This provided additional key insights into<br />

the structure of the helix-binding cleft of Bfl-1 <strong>and</strong> helped to underst<strong>and</strong> how Bfl-<br />

1 selectively associates with some proapoptotic Bcl-2 family members <strong>and</strong> what<br />

dictates its affinity <strong>and</strong> specificity <strong>for</strong> Noxa compared to other Bcl-2 family<br />

members. A manuscript is in preparation (Guan et al. In preparation).<br />

We also pursued our studies on the role of CAPER in breast cancer <strong>and</strong> its<br />

functional interaction with estrogen receptor <strong>and</strong> NF-kB. Constitutive Rel/NF-kB<br />

activity is inversely correlated with expression of estrogen receptor alpha (ERα)<br />

<strong>and</strong> is key to breast cancer cell survival, invasion <strong>and</strong> resistance to chemotherapy.<br />

Our studies uncovered a new role <strong>for</strong> CAPER in breast cancer cell invasion. In<br />

vivo studies using a mouse model of breast cancer metastasis are ongoing to<br />

elucidate its role in breast cancer progression <strong>and</strong> metastasis. Since CAPER was<br />

previously implicated both in transcriptional regulation as well as in alternative<br />

splicing, we used Affymetrix exon arrays to probe <strong>for</strong> possible effects on global<br />

gene expression in order to better underst<strong>and</strong> the mechanisms by which it affects<br />

invasion. Preliminary results suggest interesting effects of CAPER at both levels.<br />

This is consistent with CAPER mutation data <strong>and</strong> with immunofluorescence data<br />

showing co-localization of CAPER with splicing factor SC35. These experiments<br />

are being conducted in collaboration with the group of Dr. Guna Rajagopal<br />

(CINJ).<br />

Collaborative work emanating from the laboratories of Drs. Daniel Medina <strong>and</strong><br />

Roger Strair (CINJ), together with Drs. John Glod (CINJ), Arnold Rabson<br />

12


(CHINJ, CINJ <strong>and</strong> CABM) <strong>and</strong> Lauri Goodell (RWJUH) characterized the effects<br />

of mesenchymal stromal cell (MSC) on the survival <strong>and</strong> drug resistance of<br />

primary mantle cell lymphoma (MCL) cells, an aggressive subtype of non-<br />

Hodgkin’s lymphoma. Studies revealed an important role <strong>for</strong> the canonical <strong>and</strong><br />

non-canonical NF-kB pathways <strong>and</strong> expression of cytokine BAFF by MSC in the<br />

long term survival of MSC. A manuscript was submitted (Medina et al.<br />

Submitted).<br />

Finally, we pursued our collaborative work with Dr. Eileen White (CINJ) on the<br />

role of autophagy in tumorigenesis <strong>and</strong> the role of NF-kB in this context. These<br />

studies uncovered that activated Ras, commonly found in human tumors, requires<br />

autophagy to maintain oxidative metabolism <strong>and</strong> tumorigenesis. This suggests<br />

that therapeutic strategies to inhibit autophagy could perhaps be useful <strong>for</strong> the<br />

treatment of cancers in which Ras is activated. This work was published (Guo et al.<br />

<strong>2011</strong>). Ongoing studies focus on underst<strong>and</strong>ing how p62 modulates autophagy <strong>and</strong><br />

tumorigenesis <strong>and</strong> the role of NF-kB in this context, expressed <strong>and</strong> purified active<br />

fragment was crystallized <strong>and</strong> the structure determined by X-ray crystallography.<br />

Seven related con<strong>for</strong>mational states were obtained in the crystal. Position<br />

differences of the oligonucleotide/oligosaccharide (OB) binding fold lid domain<br />

over the conserved GTP binding site in the seven structures provided snapshots of<br />

the opening <strong>and</strong> closing of the active site cleft via a swivel motion. While the<br />

GTP binding site is structurally <strong>and</strong> evolutionarily conserved, the overall GTase<br />

mechanism in mammalian <strong>and</strong> yeast systems differs somewhat. Experiments are<br />

underway to crystallize complexes of human GTase with CTD <strong>and</strong> RNA as well<br />

as GTP. Protein engineering is being applied in an ef<strong>for</strong>t to crystallize the full<br />

length human CE.<br />

CAPER co-localizes<br />

with splicing factor<br />

SC35 in breast<br />

cancer cells, as seen<br />

by<br />

immunofluorescence<br />

<strong>and</strong> confocal<br />

microscopy.<br />

13


Publications:<br />

Fan, G., Simmons, M.J., Ge, S., Dutta-Simmons, J., Kucharczak, J..F, Ron, Y.,<br />

Weissmann, D., Chen, C.C., Mukherjee, C., White, E. <strong>and</strong> Gélinas, C. Defective<br />

ubiquitin-mediated degradation of anti-apoptotic Bfl-1 predisposes to lymphoma.<br />

Blood 2010 115: 3559-3569.<br />

Guo, J.Y., Chen, H-Y, Mathew, R., Fan, J., Strohecker, A.M., Kamphorst, J.J.,<br />

Chen, G., Lemmons, J.M.S., Karantza, V., Coller, H.A., DiPaola, R.S., Gélinas,<br />

C., Rabinowitz, J.D. <strong>and</strong> White, E.) Activated Ras requires autophagy to maintain<br />

oxidative metabolism <strong>and</strong> tumorigenesis. Genes & Dev. <strong>2011</strong> 25: 460-470.<br />

14


Dr. Masayori Inouye<br />

CABM Resident Faculty Member<br />

Distinguished Professor<br />

Department of Biochemistry<br />

UMDNJ-Robert Wood Johnson<br />

Medical School<br />

Dr. Sangita Phadtere<br />

Adjunct Associate Professor<br />

Yojiro Ishida<br />

Research Associate<br />

Hiroshi Kobayashi<br />

Research Associate<br />

Dr. Lili Mao<br />

Research Associate<br />

Dr. Yoshihiro<br />

Yamaguchi<br />

Research Associate<br />

Dr. Kuen-Phon Wu<br />

Postdoctoral Associate<br />

Dr. Hisako Masuda<br />

Postdoctoral Fellow<br />

Yu-Jen Chen<br />

Graduate Student<br />

Chun-Yi Lin<br />

Graduate Student<br />

Bacterial Physiology <strong>and</strong> Protein Engineering Laboratory<br />

Dr. Masayori Inouye joined CABM in October 2009. He is well-known <strong>for</strong><br />

his discovery of antisense-RNA in 1984, the function of the signal peptide <strong>for</strong><br />

secretion, biogenesis of outer membrane proteins, propeptide-mediated<br />

protein folding <strong>and</strong> molecular biology of histidine kinases. Currently he is<br />

working on the characterization of the toxin-antitoxin (TA) systems from E.<br />

coli <strong>and</strong> human pathogens such as Mycobacterium tuberculosis <strong>and</strong><br />

Staphylococcus aureus. The discovery of a sequence (ACA)-specific<br />

endoribonuclease from the E. coli TA systems led his laboratory to develop a<br />

unique protein production system converting E. coli cells to a single-protein<br />

production (SPP) bioreactor. This system allows one to label only a protein<br />

of interest in the cells with a specific isotope(s), an amino acid(s) or a toxic<br />

non-natural amino acid analogue(s) in a high yield. His laboratory also<br />

does research on the mechanism of ribosome stalling during protein<br />

synthesis. In May <strong>2011</strong>, he received the Edward J. Ill Excellence in Medicine<br />

Award <strong>for</strong> Outst<strong>and</strong>ing Medical Research Scientist. Dr. Inouye is a member<br />

of the American Academy of Arts & Sciences.<br />

Toxin-antitoxin (TA) systems; their roles in bacterial physiology <strong>and</strong> the<br />

development of novel antibiotics<br />

Almost all bacteria including human pathogens contain suicide or toxin genes which<br />

are induced under stress conditions leading to cell growth arrest <strong>and</strong> eventual cell<br />

death in a way similar to apoptosis or programmed cell death in higher systems.<br />

Escherichia coli contains at least 35 TA systems <strong>and</strong> their toxins are highly diverse,<br />

targeting DNA, mRNA, protein <strong>and</strong> cell wall synthesis. The importance of these TA<br />

systems in medical science has not been fully appreciated until recently. The Inouye<br />

laboratory works to decipher the cellular targets of these toxins <strong>and</strong> the molecular<br />

mechanisms of toxin functions.<br />

Promoter<br />

ATP-dependent<br />

proteases<br />

Antitoxin<br />

Toxin<br />

Cellular targets<br />

Bacterial Toxin-Antitoxin (Apoptosis) System<br />

Protein<br />

DNA<br />

15


mRNA interferases<br />

mRNA interferases are encoded by one of the TA systems. MazF from E. coli is<br />

the first mRNA interferase discovered in Dr. Inouye’s laboratory <strong>and</strong> functions as<br />

a sequence-specific (ACA) endoribonuclease. Its induction in E. coli cells results<br />

in growth arrest <strong>and</strong> eventual cell death. The Inouye group also observed that<br />

MazF induction in mammalian cells effectively causes Bak (a pro-apoptotic<br />

protein)-dependent programmed cell death. The application of bacterial mRNA<br />

interferases <strong>for</strong> mammalian cell growth regulation is currently being explored to<br />

develop an effective, novel method <strong>for</strong> cancer treatment <strong>and</strong> HIV eradication. In<br />

addition to E. coli MazF, the laboratory has identified a large number of mRNA<br />

interferases having different RNA cleavage specificity, including one from a<br />

highly halophilic archaeon that cleaves a specific seven base sequence. mRNA<br />

interference using highly sequence-specific mRNA interferases instead of<br />

antisense RNA or RNAi may be a novel <strong>and</strong> exciting way to regulate gene<br />

expression.<br />

Single protein production in living cells<br />

Expression of MazF, an ACA-specific mRNA interferase, results in nearly<br />

complete degradation of cellular mRNAs, leading to almost complete inhibition<br />

of protein synthesis. Intriguingly, MazF-induced cells are still fully capable of<br />

producing a protein at a high level if the mRNA <strong>for</strong> that protein is engineered to<br />

have no ACA sequences without altering its amino acid sequence. There<strong>for</strong>e, it is<br />

possible to convert E. coli cells into a bioreactor producing a single protein of<br />

interest. This “single-protein production” (SPP) system allows NMR structural<br />

studies of proteins without purification, which makes this system especially useful<br />

<strong>for</strong> structural studies of membrane proteins. In addition, the SPP system provides<br />

a unique opportunity to produce proteins in which all the residues of a specific<br />

amino acid in a protein are replaced with toxic non-natural amino acid analogues<br />

to create proteins of novel structures <strong>and</strong> functions.<br />

Publications:<br />

Awano, N., Rajagopal, V., Arbing, M., Patel, S., Hunt, J., Inouye, M., Phadtare, S.<br />

Escherichia coli RNase R has dual activities, helicase <strong>and</strong> ribonuclease. J.<br />

Bacteriol. 2010 192(5):1344-52.<br />

Peng, Y.Y., Yoshizumi, A., Danon, S.J., Glattauer, V., Prokopenko, O.,<br />

Mirochinitchenko, O., Yu, Z., Inouye, M., Werkmeister, J.A., Brodsky, B.,<br />

Ramshaw, J.A. A Streptococcus pyogenes derived collagen-like protein as a noncytotoxic<br />

<strong>and</strong> non-immunogenic cross-linkable biomaterial. Biomaterials 2010<br />

31(10):2755-61.<br />

Jung-Ho Park<br />

Graduate Student<br />

Narumi Tokunaga<br />

Graduate Student<br />

Zhuoxin Yu<br />

Graduate Student<br />

Dr. Qian Tan<br />

Research Teaching Spec.<br />

Chun Ying Xu<br />

Research Teaching Spec.<br />

Dr. Yoshihiro<br />

Yamaguchi<br />

Research Teaching Spec.<br />

Dr. Yong Long Zhang<br />

Research Teaching Spec.<br />

Xushen Chen<br />

Lab Technician<br />

16


Xu, Z., Mirochnitchenko, O., Xu, C., Yoshizumi, A., Brodsky, B., Inouye, M.<br />

Noncollagenous region of the streptococcal collagen-like protein is a trimerization domain<br />

that supports refolding of adjacent homologous <strong>and</strong> heterologous collagenous domains.<br />

Protein Sci. 2010 19(4):775-85.<br />

Hwang, J., <strong>and</strong> Inouye, M. Interaction of an essential E. coli GTPase, Der with 50S<br />

ribosome via the KH-like domain. J. Bacterial. 2010 192(8):2277-83.<br />

Schneider, W.M., Tang, Y., Vaiphei, S.T., Mao, L., Maglaqui, M., Inouye, M., Roth, M.J.,<br />

Montelione, G.T. Efficient condensed-phase production of perdeuterated soluble <strong>and</strong><br />

membrane proteins. J. Struct. Funct Genomics 2010 11(2):143-54.<br />

Hwang, J. <strong>and</strong> Inouye, M. A bacterial GAP-like protein, YihI, regulating the GTPase of<br />

Der, an essential GTP-binding protein in Escherichia coli. J. Mol. Biol. 2010 399 (5):759-<br />

72.<br />

Vaiphei, S. T., Mao, L., Shimazu, T., Park, J.H. <strong>and</strong> Inouye, M. The Use of Amino Acids<br />

as Inducers <strong>for</strong> High Level Protein expression in the Single-Protein Production (SPP)<br />

System. Appl. Environ. Microbiol. 2010 76 (18):6063-8.<br />

Love, J., Mancia, F., Shapiro, L., Punta, M., Rost, B., Girvin, M., Wang, D.N., Zhou, M.,<br />

Hunt, J.F., Szyperski, T., Gouaux, E., Mackinnon, R., McDermott, A., Honig, B., Inouye,<br />

M., Montelione, G., Hendrickson, W.A. The New York Consortium on Membrane Protein<br />

Structure (NYCOMPS): a high-throughput plat<strong>for</strong>m <strong>for</strong> structural genomics of integral<br />

membrane proteins. J. Struct. Funct. Genomics. 2010 11(3):191-9.<br />

Arbing, M.A., H<strong>and</strong>elman, S.K., Kuzin, A.P., Verdon, G., Wang, C., Su, M., Rothenbacher,<br />

F.P., Abashidze, M., Liu, M., Hurley, J.M., Xia, R., Acton, T., Inouye, M., Montelione,<br />

G.T., Woychik, N.A., Hung, J.F. Crystal structures of Phd-Doc, HigA, <strong>and</strong> YeeU establish<br />

multiple evolutionary links between microbial growth-regulating toxin-antitoxin systems.<br />

Structure. 2010 18(8):996-1010.<br />

Tang, Y., Schneider, W.M., Shen, Y., Raman, S., Inouye, M., Baker, D., Roth, M.J.,<br />

Montelione, G.T. Fully automated high-quality NMR structure determination of small<br />

(2)H-enriched proteins. J. Struct. Funct. Genomics 2010 11(4):223-32.<br />

Mao, L., Stathopulos, P.B., Ikura, M., Inouye, M. Secretion of human superoxide<br />

dismutase in Escherichia coli using the condensed single-protein-production (cSPP)<br />

System. Protein Sci. 2010 19 (12):2330-5.<br />

Zhu, L., Sharp, J.D., Kobayashi, H., Woychik, N.A., Inouye, M. Noncognate<br />

Mycobacterium tuberculosis toxin-antitoxins can physically <strong>and</strong> functionally interact. J.<br />

Biol. Chem. 2010 285(51):39732-8.<br />

Vaiphei, S.T., Tang, Y., Montelione, G.T., Inouye, M. The Use of the Condensed Single<br />

Protein Production System <strong>for</strong> Isotope-Labeled Outer Membrane Proteins, OmpA<br />

17


<strong>and</strong> OmpX in E. coli. Mol. Biotechnol. 2010 47(3):205-10.<br />

Chono, H., Matsumoto, K., Tsuda, H., Saito, N., Lee, K., Kim, S., Shibata, H.,<br />

Ageyama, N., Terao, K., Yasutomi, Y., Mineno, J., Kim, S., Inouye M. <strong>and</strong> Kato I.<br />

Acquisition of HIV-1 Resistance in T Lymphocytes Using an ACA-Specific E.<br />

coli mRNA interferase. Hum. Gene. Ther. <strong>2011</strong> 22(1):35-43.<br />

Tan, Q., Awano, N., Inouye, M. YeeV is an Escherichia coli Toxin that Inhibits<br />

Cell Division by Targeting the Cytoskeleton Proteins, FtsZ <strong>and</strong> MreB. Mol. Micro.<br />

<strong>2011</strong> 79(1):109-18.<br />

Mao, L., Inouye, K., Tao, Y., Montelione, G.T., McDermott, A.E. <strong>and</strong> Inouye, M.<br />

Suppression of phospholipid biosynthesis by cerulenin in the condensed Single-<br />

Protein-Production (cSPP) system. J. Biomol. NMR. <strong>2011</strong> 49(2):131-7.<br />

Zhang, Y.L. <strong>and</strong> Inouye, M. RatA (YfjG), an Escherichia coli toxin, inhibits 70S<br />

ribosome association to block translation initiation. Mol. Microbiol. <strong>2011</strong><br />

79(6):1418-29.<br />

Yu, Z., Brodsky, B., Inouye, M. Dissecting a bacterial collagen domain from<br />

Streptococcus pyogenes: sequence <strong>and</strong> length-dependent variations in triple helix<br />

stability <strong>and</strong> folding. J. Biol. Chem. <strong>2011</strong> 286(21):18960-8.<br />

Park, J.H., Yamaguchi, Y., Inouye, M. Bacillus subtilis MazF-bs (EndoA) is a<br />

UACAU-specific mRNA interferase. FEBS Lett. <strong>2011</strong> 585(15):2526-32.<br />

Yamaguchi, Y. <strong>and</strong> Inouye, M. Regulation of growth <strong>and</strong> death in Escherichia<br />

coli by toxin-antitoxin systems. Nat. Rev. Microbiol. <strong>2011</strong> 9(11):779-90.<br />

Yamaguchi, Y., Park, J.H. <strong>and</strong> Inouye, M. Toxin-antitoxin systems in bacteria <strong>and</strong><br />

archaea. Annu. Rev. Genet. <strong>2011</strong> 45:61-79.<br />

18


Dr. Fang Liu<br />

CABM Resident Faculty Member<br />

Associate Professor<br />

Department of Chemical Biology<br />

Ernest Mario School of Pharmacy<br />

Susan Lehman Cullman Laboratory<br />

<strong>for</strong> Cancer Research<br />

Rutgers University<br />

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

Dr. Fang Liu obtained her B.S. in biochemistry from Beijing University <strong>and</strong><br />

Ph.D. in biochemistry from Harvard University working with Dr. Michael R.<br />

Green. She conducted postdoctoral research with Dr. Joan Massagué at<br />

Memorial Sloan-Kettering Cancer <strong>Center</strong> <strong>and</strong> joined CABM in 1998. Dr.<br />

Liu has received awards from the American Association <strong>for</strong> Cancer<br />

Research-National Foundation <strong>for</strong> Cancer Research, the Pharmaceutical<br />

Research <strong>and</strong> Manufacturers of America Foundation, the Burroughs<br />

Wellcome Fund, <strong>and</strong> the Sidney Kimmel Foundation <strong>for</strong> Cancer Research.<br />

She also obtained fellowships from the K.C. Wong Education Foundation<br />

<strong>and</strong> the Jane Coffin Childs Memorial Fund <strong>for</strong> Medical Research.<br />

We study TGF-ß/Smad signal transduction, transcriptional regulation, cell cycle<br />

control <strong>and</strong> their roles in cancer.<br />

TGF-ß regulates a wide variety of biological activities. It induces potent growth<br />

inhibitory responses in normal cells, but promotes migration <strong>and</strong> invasion of cancer<br />

cells. Smads mediate the TGF-ß responses. TGF-ß binding to the cell surface<br />

receptors leads to the phosphorylation of Smad2/3 in their carboxyl-terminus as well<br />

as in the proline-rich linker region. The serine/threonine phosphorylation sites in the<br />

linker region are followed by the proline residue. Pin1, a peptidyl-prolyl cis/trans<br />

isomerase, recognizes phosphorylated serine/threonine-proline (pS/T-P) motifs. We<br />

show that Smad2/3 interacts with Pin1 in a TGF-ß-dependent manner. We further<br />

show that the phosphorylated threonine 179-proline motif in the Smad3 linker region<br />

is the major binding site <strong>for</strong> Pin1. Although EGF also induces phosphorylation of<br />

threonine 179 <strong>and</strong> other residues in the Smad3 linker region the same as TGF-ß,<br />

Pin1 is unable to bind to the EGF-stimulated Smad3. Further analysis suggests that<br />

phosphorylation of Smad3 in the carboxyl-terminus is necessary <strong>for</strong> the interaction<br />

with Pin1. Depletion of Pin1 by shRNA does not affect significantly TGF-ß-induced<br />

growth-inhibitory responses <strong>and</strong> a number of TGF-ß/Smad target genes analyzed. In<br />

contrast, knockdown of Pin1 in human PC3 prostate cancer cells strongly inhibited<br />

TGF-ß-mediated migration <strong>and</strong> invasion. Accordingly, TGF-ß induction of Ncadherin,<br />

which plays an important role in migration <strong>and</strong> invasion, is markedly<br />

reduced when Pin1 is depleted in PC3 cells. The catalytic activity of Pin1 is essential<br />

<strong>for</strong> TGF-ß-induced migration <strong>and</strong> invasion (see Figure). Since Pin1 is overexpressed<br />

in many cancers, our findings highlight the importance of Pin1 in TGF-ß-induced<br />

migration <strong>and</strong> invasion of cancer cells.<br />

19


Smad3 plays an important role in inhibiting cell proliferation <strong>and</strong> promoting<br />

apoptosis. Human Smad3 is localized near a hot spot mutation area <strong>for</strong> breast<br />

cancer <strong>and</strong> <strong>for</strong> several other types of cancers. We have found that Smad3 levels are<br />

reduced in human primary breast cancer samples <strong>and</strong> in human breast cancer cell<br />

lines. Importantly, Smad3 expression is high in normal breast stem cells but low in<br />

breast cancer stem cells. In addition, our preliminary studies indicate that 7,12dimethylbenz-[a]-anthracene<br />

(DMBA), a chemical carcinogen, induces mammary<br />

tumor <strong>for</strong>mation at a much higher frequency in Smad3 -/- or Smad3 +/- mice than in<br />

Smad3 +/+ mice. Thus, we hypothesize that Smad3 has important tumor suppressor<br />

function <strong>for</strong> breast cancer.<br />

Breast cancer stem cells are CD44 + CD24 -/low . Although CD44 lacks its own<br />

signaling domain, it associates with <strong>and</strong> co-stimulates signaling by a number of<br />

growth factor receptors, such as Her2 <strong>and</strong> EGF receptor 1. CD44 is overexpressed<br />

in the vast majority of breast cancers. CD44 is not just a marker <strong>for</strong> breast cancer<br />

stem cells. It is necessary <strong>for</strong> breast tumor initiation, tumor growth, <strong>and</strong> metastasis.<br />

Knockdown of CD44 greatly reduces tumor-initiating frequency, tumor weight,<br />

<strong>and</strong> colony <strong>for</strong>mation in soft agar assay. Disruption of tumor cell surface CD44<br />

function induces apoptosis in metastatic mammary carcinoma cells in vivo. A<br />

predominant function of CD44 is to promote growth <strong>and</strong> to inhibit apoptosis. The<br />

growth-inhibitory <strong>and</strong> tumor-suppressive functions of p53 depend on its repression<br />

of CD44 expression. Our preliminary studies have discovered that Smad3 potently<br />

represses CD44 transcription in mammary epithelial cells. Thus, we further<br />

hypothesize that Smad3 repression of CD44 expression is essential <strong>for</strong> its tumor<br />

suppressor activity, <strong>and</strong> that Smad3 <strong>and</strong> p53 cooperate to inhibit breast<br />

tumorigenesis. Our research is directed towards testing these hypotheses. Our<br />

findings will be highly significant <strong>for</strong> preventing <strong>and</strong> treating breast cancer.<br />

20


Publications:<br />

Matsuura, I., Chiang, K.-N., Lai, C.-Y., He, D., Wang, G., Ramkumar, R. Uchida, T.,<br />

Ryo, A., Lu, K. P., <strong>and</strong> Liu, F. (2010)Pin1 promotes TGF-ß-induced migration <strong>and</strong><br />

invasion.J. Biol. Chem. 285, 1854-1864.<br />

Sasseville, M., Ritter, L. J., Nguyen, T., Liu, F., Mottershead, D. G., Russell, D. L., <strong>and</strong><br />

Gilchrist, R. B. (2010).Growth differentiation factor 9 signaling requires ERK1/2<br />

activity in mouse granulosa <strong>and</strong> cumulus cells.J. Cell Sci. 123, 3166-3176.<br />

Chang, X., Liu, F., Wang, X., Lin, A., Zhou, H., <strong>and</strong> Su, B. (<strong>2011</strong>) The kinases MEKK2<br />

<strong>and</strong> MEKK3 regulate trans<strong>for</strong>ming growth factor-ß-mediated helper T cell<br />

differentiation. Immunity 34, 201-212.<br />

Liu, F. (<strong>2011</strong>) Inhibition of Smad3 activity by cyclin D-CDK4 <strong>and</strong> cyclin E-CDK2 in<br />

breast cancer cells. Cell Cycle 10, 186.<br />

So, J. Y., Lee, H. J., Smolarek, A. K., Paul, S., Maehr, H., Uskokovic, M., Zheng, X.,<br />

Conney, A., Cai, L., Liu, F., <strong>and</strong> Suh, N. (<strong>2011</strong>) A novel Gemini vitamin D analog<br />

represses the expression of a stem cell marker CD44 in breast cancer. Mol. Pharmacol.<br />

79, 360-367.<br />

Cohen-Solal K. A., Merrigan K. T., Chan, J. L., Goydos, J. S., Chen, W., Foran, D. J.,<br />

Liu, F., Lasfar, A., <strong>and</strong> Reiss, M. (<strong>2011</strong>) Constitutive Smad linker phosphorylation in<br />

melanoma: a mechanism of resistance to trans<strong>for</strong>ming growth factor-β-mediated growth<br />

inhibition. Pigment Cell Melanoma Res. 24, 512-524.<br />

Dhar-Mascareno, M., Belishov, I., Liu, F., <strong>and</strong> Mascareno, E. J. (<strong>2011</strong>) Hexim-1<br />

modulates <strong>and</strong>rogen receptor <strong>and</strong> the TGFβ signaling during the progression of prostate<br />

cancer. The Prostate in press<br />

21


Dr. Peter Lobel<br />

CABM Resident Faculty Member<br />

Professor<br />

Department of Pharmacology<br />

UMDNJ-Robert Wood Johnson<br />

Medical School<br />

Dr. David Sleat<br />

Research Associate Professor<br />

Dr. Haiyan Zheng<br />

Principal Research Associate<br />

Dr. Yu Meng<br />

Research Associate<br />

Dr. Istvan Sohar<br />

Research Associate<br />

Ling Huang<br />

Graduate Student<br />

Su Xu<br />

Graduate Fellow<br />

Mukarram El-Banna<br />

Research Teaching Spec.<br />

Dr. Meiqian Qian<br />

Research Teaching Spec.<br />

Protein Targeting Laboratory<br />

Dr. Peter Lobel trained at Columbia University <strong>and</strong> Washington University<br />

in St. Louis <strong>and</strong> joined CABM in 1989. He is currently conducting research<br />

on lysosomes <strong>and</strong> associated human hereditary metabolic diseases. This<br />

work resulted in identification of three disease genes that cause fatal<br />

neurodegenerative disorders. He was a Searle Scholar <strong>and</strong> received the<br />

excellence in research award from the UMDNJ Foundation. Dr. Lobel is the<br />

director of the CABM Mass Spectrometry Core Facility.<br />

.<br />

Our laboratory has developed new methods <strong>for</strong> disease discovery that evolved from<br />

our research on lysosomal enzyme targeting. Lysosomes are membrane-bound,<br />

acidic organelles that are found in all eukaryotic cells. They contain a variety of<br />

different proteases, glycosidases, lipases, phosphatases, nucleases <strong>and</strong> other<br />

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<br />

different from other glycoproteins <strong>and</strong> are selectively phosphorylated on mannose<br />

residues. The mannose 6-phosphate serves as a recognition marker that allows the<br />

enzymes to bind mannose 6-phosphate receptor which ferries the lysosomal enzyme<br />

to the lysosome. In the lysosome, the enzymes function in concert to break down<br />

complex biological macromolecules into simple components. The importance of<br />

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

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<br />

<strong>and</strong> early death.<br />

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

disorders is based on our ability to use mannose 6-phosphate receptor derivatives to<br />

visualize <strong>and</strong> purify mannose 6-phosphate containing lysosomal enzymes. Once we<br />

discover the cause of a given disease, we conduct further research to underst<strong>and</strong> the<br />

underlying system <strong>and</strong> potentially to develop therapeutics. We also investigate the<br />

basic mechanism <strong>for</strong> targeting of lysosomal proteins <strong>and</strong> the composition of the<br />

lysosome.<br />

22


In the prior year, we have made considerable progress investigating late infantile<br />

neuronal ceroid lipofuscinosis (LINCL), a recessive neurodenerative disease of<br />

childhood that is due to deficiencies in the lysosomal protease tripeptidyl-peptidase 1<br />

(TPP1). In one study, we per<strong>for</strong>med mass spectrometric analyses on storage material<br />

<strong>and</strong> lysosomal fractions from an LINCL mouse model. This revealed several<br />

c<strong>and</strong>idates <strong>for</strong> proteins stored in brain. In depth analysis of the most prominent of<br />

these, glial fibrillary acidic protein, revealed that this protein represented a<br />

contaminant that adventitiously associated with storage material <strong>and</strong> lysosomes during<br />

the isolation procedure. We also used the mouse model to explore potential<br />

therapeutic approaches <strong>and</strong> found that intrathecal administration of recombinant<br />

human TPP1 resulted in significant delivery of enzyme to the brain. Importantly, this<br />

treatment prolonged lifespan <strong>and</strong> improved decreased disease symptoms. Finally, in<br />

collaboration with scientists at the University of Missouri <strong>and</strong> BioMarin Therapeutics,<br />

as a step towards the clinic we conducted pilot enzyme replacement studies using an<br />

LINCL dog model.<br />

We are also continuing our investigation of the lysosomal proteome. Following the<br />

principles of de Duve <strong>and</strong> colleagues that lead to the discovery of the lysosome, we<br />

analyzed rat liver subcellular fractions using mass spectrometry to identify <strong>and</strong><br />

determine the distribution of different proteins. This led to the identification of<br />

numerous c<strong>and</strong>idate lysosomal proteins <strong>and</strong> provided further evidence that ABCB6 is<br />

lysosomal <strong>and</strong> that superoxide dismutase 1 has a mixed cytoplasmic-lysosomal<br />

localization.<br />

Percent survival<br />

100<br />

50<br />

0<br />

1.2 mg/animal<br />

0.38 mg/animal<br />

0.12 mg/animal<br />

0.038 mg/animal<br />

vehicle<br />

0 30 60 90 120 150 180<br />

Time (days)<br />

Acute intrathecal administration of recombinant human TPP1 extends lifespan of a<br />

LINCL mouse model. Animals were administered the indicated dose of TPP1 at the<br />

age of 28-30 days <strong>and</strong> then followed <strong>for</strong> survival.<br />

Jennifer Wiseman<br />

Research Teaching Spec.<br />

Caifeng Zhao<br />

Research Teaching Spec.<br />

23


Publications:<br />

Della Valle, M. C., Sleat, D. E., Zheng, H., Moore, D. F., Jadot, M. <strong>and</strong> Lobel, P.<br />

Classification of subcellular location by comparative proteomic analysis of native<br />

<strong>and</strong> density-shifted lysosomes. Mol.Cell. Proteomics. <strong>2011</strong> 10, M110 006403.<br />

Qian, Y., Lee, I., Lee, W. S., Qian, M., Kudo, M., Canfield, W. M., Lobel, P. <strong>and</strong><br />

Kornfeld, S. Functions of the alpha, beta, <strong>and</strong> gamma subunits of UDP-<br />

GlcNAc:lysosomal enzyme N-acetylglucosamine-1-phosphotransferase. J. Biol.<br />

Chem. 2010 285: 3360-3370.<br />

Vuillemenot, B. R., Katz, M. L., Coates, J. R., Kennedy, D., Tiger, P., Kanazono, S.,<br />

Lobel, P., Sohar, I., Xu, S., Cahayag, R., Keve, S., Koren, E., Bunting, S., Tsuruda,<br />

L. S. <strong>and</strong> O'Neill, C. A. Intrathecal tripeptidyl-peptidase 1 reduces lysosomal storage<br />

in a canine model of late infantile neuronal ceroid lipofuscinosis. Mol. Genet. Metab.<br />

<strong>2011</strong> 104: 325-337.<br />

Xu, S., Sleat, D. E., Jadot, M. <strong>and</strong> Lobel, P. Glial fibrillary acidic protein is elevated<br />

in the lysosomal storage disease classical late-infantile neuronal ceroid<br />

lipofuscinosis, but is not a component of the storage material. Biochem. J. 2010 428:<br />

355-362.<br />

Xu, S., Wang, L., El-Banna, M., Sohar, I., Sleat, D. E. <strong>and</strong> Lobel, P. Large-volume<br />

intrathecal enzyme delivery increases survival of a mouse model of late infantile<br />

neuronal ceroid lipofuscinosis. Mol Ther. <strong>2011</strong> 19: 1842-1848<br />

24


Dr. James Millonig<br />

CABM Resident Faculty Member<br />

Associate Professor<br />

Department of Neuroscience <strong>and</strong><br />

Cell Biology<br />

UMDNJ-Robert Wood Johnson<br />

Medical School<br />

Adjunct Assistant Professor<br />

Department of Genetics<br />

Rutgers University<br />

Dr. Paul Matteson<br />

Research Associate<br />

Anna Dulencin<br />

Graduate Fellow<br />

Myka Ababon<br />

Graduate Student<br />

Ji Yeon Choi<br />

Graduate Student<br />

Silky Kamdar<br />

Graduate Student<br />

Bo Li<br />

Graduate Student<br />

Mai Soliman<br />

Graduate Student<br />

Developmental Neurogenetics Laboratory<br />

Dr. James H. Millonig came to CABM in September 1999 from the Rockefeller<br />

University where he was a postdoctoral fellow in the laboratory of Dr. Mary<br />

E. Hatten. His postdoctoral research combined neurobiology <strong>and</strong> mouse<br />

genetics to characterize <strong>and</strong> clone the dreher mouse locus. He did his doctoral<br />

research at Princeton University with Dr. Shirley M. Tilghman. Dr. Millonig<br />

is a recipient of the March of Dimes Basil O’Connor Starter Research Award<br />

<strong>and</strong> grants from the National Institutes of Health, Department of Defense,<br />

National Alliance <strong>for</strong> Research on Schizophrenia <strong>and</strong> Depression, Autism<br />

Speaks, National Alliance <strong>for</strong> Autism Research, N.J. Governor’s Council on<br />

Autism <strong>and</strong> New Jersey Commission on Spinal Cord Research. He is director<br />

of the RU/Princeton/RWJMS M.D./Ph.D. program.<br />

Autism Spectrum Disorder (ASD)<br />

Individuals diagnosed with ASD exhibit deficiencies in communication <strong>and</strong> reciprocal<br />

social interactions that are accompanied by rigid or repetitive interests <strong>and</strong> behaviors.<br />

ASD is considered to be a neurodevelopmental disorder that has a polygenic basis.<br />

Increased risk <strong>for</strong> the disorder is believed to be due to multiple genes interacting with each<br />

other as well as environmental factors.<br />

Risk <strong>for</strong> ASD is likely due to both genetic <strong>and</strong> non-genetic environmental factors, with<br />

epigenetic regulation of genes providing a possible interface between genetic <strong>and</strong><br />

environmental factors. Our previous research has focused on the homeobox transcription<br />

factor, ENGRAILED 2 (EN2). The common alleles (underlined) of two intronic EN2 SNPs,<br />

rs1861972 (A/G) <strong>and</strong> rs1861973 (C/T), are significantly associated with ASD (518<br />

families, P=.00000035). Subsequent association, LD mapping, <strong>and</strong> re-sequencing<br />

identified the associated rs1861972-rs1861973 as the most suitable c<strong>and</strong>idate to test <strong>for</strong><br />

function.<br />

Function was then tested in primary cultures of cerebellar granule cells since endogenous<br />

En2 is expressed at high levels in these cells. Luciferase assays demonstrated that the A-C<br />

haplotype results in an ~250% increase in expression. Additional analysis determined that<br />

both associated alleles are sufficient <strong>and</strong> necessary <strong>for</strong> this activator function. EMSAs<br />

revealed that nuclear factors specifically bind the A-C haplotype. An unbiased proteomic<br />

screen identified two transcription factors, Cux1 <strong>and</strong> NfiB, that bind significantly better to<br />

the A-C haplotype. Knock-down, over-expression, ChIP <strong>and</strong> EMSA supershift analysis<br />

demonstrated that both transcription factors bind the A-C haplotype at the same time <strong>and</strong><br />

are required <strong>for</strong> activator function. These studies determined that the ASD-associated A-C<br />

haplotype functions as a transcriptional activator.<br />

25


We then investigated if EN2 levels are increased in individuals with autism. Ninety<br />

cerebellar samples were acquired <strong>and</strong> Taqman QRTPCR measured normalized EN2<br />

mRNA levels. A significant increase was observed in affected individuals with an A-C<br />

haplotype compared to controls (74% increase, P=0.0005). Thus the A-C haplotype is<br />

also correlated with increased EN2 levels in individuals with ASD.<br />

Because ASD is a neurodevelopmental disorder, we generated transgenic mice. EN2 was<br />

replaced with a DsRed, reporter <strong>and</strong> 25kb of evolutionarily conserved sequence was<br />

used to drive expression of the transgene. 6 A-C <strong>and</strong> 8 G-T lines were generated.<br />

QRTPCR measured Ds-Red levels <strong>for</strong> all the lines at 5 developmental time points (E9.5,<br />

E12.5, E17.5, P6 <strong>and</strong> adult). At all stages the A-C haplotype results in increased<br />

expression (P


We have mapped 5 different vl modifier loci on these different genetic<br />

backgrounds (Modifiers of vacuolated lens, Modvl 1-5, LOD 3.7-5.0)(Matteson<br />

et al., 2008; Korstanje et al., 2008).<br />

To identify the genes in the Modvl 95% CIs that may contribute to these<br />

modifying effects, transcripts co-expressed with Gpr161 during development<br />

were identified by bioin<strong>for</strong>matic analysis. Ingenuity Pathway Analysis<br />

indicates the Wnt <strong>and</strong> EMT pathways are over-represented (P


Publications:<br />

Figure 1. A) Luc assays demonstrate 50% increase in A-C haplotype<br />

after 1 day in vitro (left), <strong>and</strong> ~250% increase after 3 days in vitro in P6<br />

mouse granule cells (right). B) Luc assays demonstrate that a 200bp<br />

fragment encompassing both SNPs is sufficient <strong>for</strong> activator function<br />

(left), while analysis of the rare haplotypes (A-T <strong>and</strong> G-C) determined<br />

that both associated alleles are necessary (right) <strong>for</strong> function. C)<br />

EMSAs with P6 mouse granule cell nuclear extract demonstrate<br />

nuclear factors (arrow) bind better to A-C than G-T haplotype. D)<br />

Knockdown of both Cux1 <strong>and</strong> NfIB demonstrate that both factors are<br />

required <strong>for</strong> endogenous EN2 expression in HEK-293T cells (n=3).<br />

Choi J, Matteson PG, Millonig JH. <strong>2011</strong> Cut-like homeobox1 <strong>and</strong> Nuclear factor I/B<br />

mediate ENGRAILED2 Autism Spectrum Disorder-associated haplotype function. Human<br />

Molecular Genetics. EPub 12/12/11<br />

Choi J, Kamdar S, Rahman T, Matteson PG, Millonig JH. (<strong>2011</strong>). ENGRAILED 2 (EN2)<br />

genetic <strong>and</strong> functional analysis. Autism Spectrum Disorders - From Genes to Environment,<br />

edited by<br />

Tim Williams<br />

Deng Q, Andersson E, Hedlund E, Alekseenko Z, Coppola E, Panman L, Millonig JH,<br />

Brunet J-F, Ericson J. Specific <strong>and</strong> Integrated Roles of Lmx1a, Lmx1b <strong>and</strong> Phox2a in<br />

Ventral Midbrain. Development <strong>2011</strong> 138:3399-408<br />

28


Dr. Arnold Rabson<br />

Director<br />

Child Health Institute of New Jersey<br />

Professor<br />

Department of Molecular Genetics,<br />

Microbiology <strong>and</strong> Immunology<br />

Department of Pathology <strong>and</strong><br />

Laboratory Medicine<br />

Department of Pediatrics<br />

UMDNJ-Robert Wood Johnson<br />

Medical School<br />

Dr. Celine Granier<br />

Postdoctoral Fellow<br />

Dr. Hsin-Ching Lin<br />

Research Scientist<br />

Viral Pathogenesis Laboratory<br />

Dr. Rabson’s laboratory studies the molecular basis of cancer <strong>and</strong> human<br />

retroviral infections. His recent research activities are focused in two major<br />

areas: 1) the mechanisms responsible <strong>for</strong> the pathogenesis of retroviral<br />

infections, particularly infection with the human T-cell leukemia virus<br />

(HTLV-1), <strong>and</strong> 2) the roles of cellular transcriptional regulation in<br />

development <strong>and</strong> in human cancer.<br />

It is estimated that over 20 million people are infected by HTLV-1, the first<br />

identified human retrovirus, <strong>and</strong> 2-5% of them are likely to develop a serious<br />

HTLV-1-associated disease. Dr. Rabson is studying the differential mechanisms<br />

by which HTLV-1 causes an aggressive <strong>and</strong> fatal T cell leukemia/lymphoma<br />

(Adult T-Cell Leukemia, ATL) in some infected individuals <strong>and</strong> a series of<br />

immunological disorders including a neurological disease of the spinal cord<br />

(HTLV-associated Myelopathy, HAM/TSP) in other infected people. These<br />

disorders occur in only a minority of patients, years after initial infection. This<br />

suggests that there are important interactions between the virus <strong>and</strong> the host that<br />

determine its pathogenicity. Furthermore, a strong host immune response against<br />

HTLV-1 gene products favors the establishment of latent infection in vivo.<br />

Nonetheless, expression of HTLV-1 gene products, particularly the Tax<br />

transactivator, is required <strong>for</strong> disease development. The Rabson laboratory has<br />

identified <strong>and</strong> characterized the mechanisms by which the expression of HTLV-1<br />

can be activated in infected human T-lymphocytes leading ultimately to disease<br />

pathogenesis. They have shown that stimulation through the T-cell receptor can<br />

potently induce HTLV-1 gene expression, including the expression of Tax,<br />

leading to T cell immortalization. They have recently confirmed this in a mouse<br />

model of HTLV-1 latency <strong>and</strong> gene expression. T cell receptor stimulation of<br />

CD4+ cells in a Tax transgenic mouse leads to increased T cell proliferation <strong>and</strong><br />

long-lived survival (>13 months). Thus, activation of latently infected T cells in<br />

HTLV-1-infected individuals may induce expression of Tax, ultimately leading to<br />

proliferation of subsets of cells, based on specific T cell receptor-lig<strong>and</strong><br />

interactions. This could explain the progressive polyclonal to oligoclonal<br />

proliferation to ultimately monoclonal proliferation of infected T-cells that<br />

characterizes HTLV-1-associated diseases.<br />

29


Another important feature of HTLV-1-infected T cells expressing the Tax<br />

transactivator is the production of multiple cytokines. The Rabson laboratory has<br />

shown that Tax-expressing CD4+ T cells fail to properly “bias”, i.e. fail to<br />

differentiate into specific T helper subtypes, such as Th1, Th2, Th17 <strong>and</strong> Treg cells.<br />

Instead, the Tax-expressing cells express cytokines characteristic of all of these<br />

different cells, suggesting a “confused” phenotype that may in turn lead to immune<br />

dysregulation with the potential <strong>for</strong> both opportunistic infections <strong>and</strong> autoimmunelike<br />

diseases.<br />

The second major area of study in Dr. Rabson’s laboratory continues to be the<br />

roles of transcriptional regulation in the pathogenesis of human cancer. In<br />

collaboration with Drs. Ruth Steward (Waksman Institute), Dale Schaar (CINJ),<br />

<strong>and</strong> Hatem Sabaawy (CINJ), the Rabson lab is investigating the functions of<br />

PDCD2, a highly conserved nuclear <strong>and</strong> cytoplasmic protein, the mutation of<br />

which disrupts hematopoiesis in Drosophila (Drs. Steward <strong>and</strong> Minakhina). The<br />

Rabson lab has shown high levels of expression of this enigmatic protein in very<br />

early developing embryos <strong>and</strong> is currently per<strong>for</strong>ming mouse genetic experiments<br />

<strong>and</strong> transcriptional studies to determine its function. Dr. Rabson has also<br />

continued collaborations with Dr. Roger Strair at CINJ on the potential utility of<br />

NF-κB inhibition in leukemia therapy. A clinical trial, examining the effects of<br />

NF-κB inhibition, as part of induction chemotherapy, on the molecular phenotype<br />

of Acute Myeloid Leukemia is in the late stages of development <strong>and</strong> is a follow-up<br />

to a published trial showing the feasibility of inhibiting NF-κB in patients.<br />

Differentiated mouse embryonic stem cells<br />

30


Publications:<br />

Zheng, X., Cui, X.X., Gao, Z., Zhao, Y., Lin, Y., Shi, W.J., Huang, M.T., Liu, Y.,<br />

Rabson, A., Reddy, B., Yang, C.S., <strong>and</strong> Conney, A.H. Atorvastatin <strong>and</strong> celecoxib in<br />

combination inhibits the progression of <strong>and</strong>rogen-dependent LNCaP xenograft prostate<br />

tumors to <strong>and</strong>rogen independence. Cancer Prev. Res. 2010 3:114-24.<br />

Rabson, A.B. Trisomy 21 leukemias: Finding the hits that matter. Oncogene 2010<br />

29:6099-101.<br />

Swaims, A.Y., Khani, F, Zhang, Y., Roberts, A.I., Devadas, S., Shi, Y.*, <strong>and</strong> Rabson,<br />

A.B.* Immune activation induces immortalization of HTLV-1 LTR-Tax transgenic CD4 +<br />

T-cells. Blood 2010 116(16):2994-3003.<br />

Chen, Y., Rabson, A.B., <strong>and</strong> Gorski, D.H. MEOX2 regulates nuclear factor-κB activity in<br />

vascular endothelial cells through interactions with p65 <strong>and</strong> IκBβ. Cardiovasc Res. 2010<br />

87:723-31.<br />

Chen, Y., B<strong>and</strong>a, M., Speyer, C.L., Smith, J.S., Rabson, A.B. <strong>and</strong> Gorski, D.H.<br />

Regulation of the expression <strong>and</strong> activity of the antiangiogenic homeobox gene,<br />

GAX/MEOX2, by ZEB2 <strong>and</strong> microRNA-221. Mol. Cell Biol. 2010 30:3902-13.<br />

Fu, J., Qu, Z., Yan, P., Ishikawa, C., Mori, N., Aqeilan, R.I., Rabson, A.B. <strong>and</strong> Xiao, G.<br />

The tumor suppressor gene WWOX links the canonical <strong>and</strong> non-canonical NF-κB<br />

pathways in HTLV-1 Tax-mediated tumorigenesis. Blood <strong>2011</strong> 117:1652-61.<br />

De Lorenzo, M.S., Baljinnyam, E., Vatner, DE, Abarzua, P., Vatner S.F., <strong>and</strong> Rabson,<br />

A.B. Caloric Restriction reduces mammary tumor growth <strong>and</strong> metastasis. Carcinogenesis<br />

<strong>2011</strong> 32:1381-7.<br />

31


Dr. Aaron J. Shatkin<br />

Director of CABM<br />

Professor<br />

Department of Molecular<br />

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

Immunology<br />

UMDNJ-Robert Wood Johnson<br />

Medical School<br />

University Professor<br />

of Molecular Biology<br />

Rutgers University<br />

Dr. Weihua Qiu<br />

Research Associate<br />

Molecular Virology Laboratory<br />

Dr. Aaron J. Shatkin, a member of the National Academy of Sciences, has held<br />

research positions at the National Institutes of Health, The Salk Institute, <strong>and</strong> the<br />

Roche Institute of Molecular Biology. He has taught at Georgetown University<br />

Medical School, Cold Spring Harbor Laboratory, The Rockefeller University,<br />

UMDNJ-Newark Medical School, University of Puerto Rico, Princeton University <strong>and</strong><br />

other institutions. Shatkin was editor of the Journal of Virology from 1973-1977,<br />

founding editor-in-chief of Molecular <strong>and</strong> Cellular Biology from 1980-1990 <strong>and</strong> is<br />

currently editor of Advances in Virus Research. He has also served on the advisory<br />

boards of a number of organizations. In 1989 he was recognized by New Jersey<br />

Monthly magazine with a New Jersey Pride Award <strong>for</strong> his contributions to the State’s<br />

economic development. In 1991 the State of N.J. awarded Shatkin the Thomas Alva<br />

Edison Science Award, <strong>and</strong> he was selected as one of New Jersey’s top ten scientists<br />

by N.J. Business magazine. He was elected Fellow of the American Academy of Arts<br />

<strong>and</strong> Sciences in 1997 <strong>and</strong> the American Association <strong>for</strong> the Advancement of Science in<br />

1999. Shatkin received the 1977 National Academy of Sciences Molecular Biology<br />

Award, the 2003 Association of American Medical Colleges Award <strong>for</strong> Distinguished<br />

Research in the Biomedical Sciences, the Edward J. Ill Outst<strong>and</strong>ing Medical Research<br />

Scientist Award <strong>and</strong> from Robert Wood Johnson Medical School the R.W. Schlesinger<br />

Basic Science Mentoring Award in 2009 <strong>and</strong> the Honorary Alumnus Award in <strong>2011</strong>.<br />

mRNA 5'-capping<br />

One of the earliest steps in the cascade of events necessary <strong>for</strong> mRNA <strong>for</strong>mation <strong>and</strong><br />

function is the addition of a 5'-terminal m7GpppN "cap" to nascent RNA Polymerase<br />

II transcripts. This structural hallmark is present on most eukaryotic cellular as well<br />

as viral mR2NAs <strong>and</strong> is essential <strong>for</strong> viability. The presence of a cap enhances several<br />

downstream events in cellular gene expression including RNA stability, splicing of<br />

pre-mRNAs in the nucleus <strong>and</strong> initiation of protein synthesis in the cytoplasm. These<br />

important effects have fostered many studies that defined the enzymatic mechanisms<br />

of capping. We have cloned <strong>and</strong> sequenced the mouse <strong>and</strong> human capping enzymes<br />

(CEs, ~98% identical) <strong>and</strong> mapped the human protein to 6q16, a region implicated in<br />

tumor suppression. The 597-amino acid, 68kD mammalian polypeptides consist of<br />

two functional domains --N-terminal RNA 5' triphosphatase (RTase) <strong>and</strong> C-terminal<br />

guanylyltransferase (GTase). Mutational, biochemical <strong>and</strong> genetic analyses<br />

demonstrated that the GTase active site is a lysine in the sequence 294 Lys-X-Asp-<br />

Gly 297, one of several highly conserved motifs characteristic of a<br />

nucleotidyltransferase superfamily of proteins that includes other cellular <strong>and</strong> viral<br />

CEs. A haploid deletion strain of S. cerevisiae missing the guanylyltransferase<br />

enzyme was complemented <strong>for</strong> growth by the mouse wild type cDNA clone despite<br />

only ~25% sequence identity. However, a mouse clone containing alanine in place of<br />

lysine in the KXDG motif did not complement. The results demonstrated the<br />

functional conservation of CEs from yeast to mammals.<br />

We found that mammalian capping enzyme binds via its GTase region to the<br />

hyperphosphorylated C-terminal domain (CTD) of the largest subunit of RNA<br />

32


polymerase II, facilitating the selective capping of pre-mRNAs. Similarly, the full<br />

length <strong>and</strong> C-terminal domain of CE were<br />

localized to the nucleus in transfected cells <strong>and</strong> also bound poly (U) in vitro,<br />

suggesting that the C-terminal domain of CE can bind nascent transcript 5' termini<br />

<strong>for</strong> capping directly. The CE N-terminal RNA 5'-triphosphatase (amino acids 1-<br />

237) contains the sequence VHCTHGFNRTG which corresponds to the<br />

conserved active-site motif in protein tyrosine phosphatases (PTPs). Mutational<br />

analyses identified the Cys <strong>and</strong> Arg residues in this motif <strong>and</strong> an upstream<br />

aspartate as required <strong>for</strong> triphosphatase activity. These <strong>and</strong> other results indicate<br />

that removal of phosphate from RNA 5' ends <strong>and</strong> from modified tyrosine residues<br />

in proteins occurs by a similar mechanism.<br />

We also cloned <strong>and</strong> characterized the third essential enzyme <strong>for</strong> mRNA 5'capping,<br />

human mRNA (guanine-7-) methyltransferase (MTase). It mapped to<br />

18p11.22-p11.23, a region encoding brain transcripts that have been suggested as<br />

positional c<strong>and</strong>idates <strong>for</strong> susceptibility to bipolar disorder. Sequence alignment of<br />

the 476-amino acid MTase protein within the corresponding yeast, C. elegans <strong>and</strong><br />

Drosophila enzymes demonstrated several required, conserved motifs including<br />

one <strong>for</strong> binding S-adenosylmethionine. MTase bound to human CE <strong>and</strong> also<br />

<strong>for</strong>med ternary complexes with the elongating <strong>for</strong>m of RNA polymerase II. To<br />

identify other proteins that interact with CEs, we used a yeast two-hybrid system<br />

to screen a human fetal brain cDNA library with full length human CE <strong>and</strong><br />

isolated transcription elongation factor SPT5. It bound to CE <strong>and</strong> stimulated RNA<br />

guanylylation but not the triphosphatase step of capping. Purified,<br />

hyperphosphorylated CTD similarly stimulated RNA guanylylation in vitro, but<br />

the effects of P-CTD <strong>and</strong> SPT5 were not additive, suggesting a common or<br />

overlapping binding site on CE. By using two-hybrid, GST-pulldown <strong>and</strong> coimmunoprecipitation<br />

approaches, we also found that MTase interacts with the<br />

nuclear transporter, importin-α (Impα). MTase selectively bound <strong>and</strong> methylated<br />

RNA containing 5'-terminal GpppG, <strong>and</strong> both activities were stimulated severalfold<br />

by Impα. MTase/RNA/Impα complexes were dissociated by addition of<br />

Impβ which also blocked Impα stimulation of RNA cap methylation. RanGTP but<br />

not RanGDP prevented these effects of Impβ. The results suggested that, in<br />

addition to a linkage between capping <strong>and</strong> transcription, mRNA biogenesis <strong>and</strong><br />

nucleocytoplasmic transport are functionally connected.<br />

A general model of capping: RNA Polymerase II containing hypophosphorylated<br />

CTD initiates transcription, produces 20-25 nucleotide 5'-triphosphorylated<br />

transcripts <strong>and</strong> pauses with SPT5 bound as part of a large transcription complex.<br />

Serine 5 residues in the heptad repeats that comprise the CTD <strong>and</strong> SPT5 are<br />

phosphorylated by transcription factor TFIIH, changing the CTD con<strong>for</strong>mation to<br />

allow CE binding. The 5’ end of the ~25 nucleotide nascent transcript is capped<br />

as it becomes exposed at the surface of the polymerase complex, stimulated by<br />

SPT5 as well as by the hyperphosphorylated CTD (P-CTD). MTase binds to CE<br />

(mammals) or to P-CTD (yeast). Impα stimulates MTase binding <strong>and</strong> N7 guanine<br />

methylation. After phosphorylation of SPT5 <strong>and</strong> RNA Polymerase II on CTD<br />

33


serine 2 residues by pTEF-b, polymerase bound complexes of factor DSIF <strong>and</strong><br />

negative elongation factor (NELF) dissociate <strong>and</strong> processive elongation ensues. In<br />

an ef<strong>for</strong>t to decipher how the critically important human GTase works, we<br />

determined that the minimum enzymatically active domain resides in CE residues<br />

229-567. The<br />

expressed <strong>and</strong> purified active fragment was crystallized <strong>and</strong> the structure<br />

determined by X-ray crystallography. Seven related con<strong>for</strong>mational states were<br />

obtained in the crystal. Position differences of the oligonucleotide/oligosaccharide<br />

(OB) binding fold lid domain over the conserved GTP binding site in the seven<br />

structures provided snapshots of the opening <strong>and</strong> closing of the active site cleft<br />

via a swivel motion. While the GTP binding site is structurally <strong>and</strong> evolutionarily<br />

conserved, the overall GTase mechanism in mammalian <strong>and</strong> yeast systems differs<br />

somewhat. Experiments are underway to crystallize complexes of human GTase<br />

with CTD <strong>and</strong> RNA as well as GTP. Protein engineering is being applied in an<br />

ef<strong>for</strong>t to crystallize the full length human CE.<br />

Publications:<br />

Structure of the GTase domain of<br />

human CE consisting of the base,<br />

hinge <strong>and</strong> flexible lid. The<br />

predicted single-str<strong>and</strong>ed RNA<br />

binding track, marked by sulfate<br />

ions incorporated from the<br />

crystallization solution, lies in a<br />

positively charged cleft below the<br />

lid <strong>and</strong> close to modeled GMP with<br />

phosphoamide linkage to K294 of<br />

the conserved KXDG active site<br />

sequence near the center of the<br />

structure. Also shown is the<br />

predicted CTD binding site<br />

modeled based on the structure of a<br />

mouse homolog complex (Ghosh et<br />

al. <strong>2011</strong> Mol. Cell 43: 299-310).<br />

Blue to red correlates with<br />

increasing flexibility.<br />

Chu, C., Das, K., Tyminski, J.R., Bauman, J.D., Guan, R., Qiu, W., Montelione,<br />

G.T., Arnold, E., <strong>and</strong> Shatkin, A.J. Structure of the guanylyltransferase domain of<br />

human mRNA capping enzyme. Proc. Natl. Acad. Sci. USA. <strong>2011</strong> 108:10104-8.<br />

Topisirovic, I., Svitkin, Y., Sonenberg, N., <strong>and</strong> Shatkin, A.J. Cap <strong>and</strong> cap-binding<br />

proteins in the control of gene expression. Wiley Interdiscip Rev RNA. <strong>2011</strong><br />

2(2):277-98.<br />

34


Dr. Mengqing Xiang<br />

CABM Resident Faculty Member<br />

Professor<br />

Department of Pediatrics<br />

UMDNJ-Robert Wood Johnson<br />

Medical School<br />

Dr. Kangxin Jin<br />

Postdoctoral Associate<br />

Dr. Shengguo Li<br />

Research Teaching Spec.<br />

Dr. Huijun Luo<br />

Research Teaching Spec.<br />

Dr. Kamana Misra<br />

Research Teaching Spec.<br />

Min Zou<br />

Graduate Student<br />

Molecular Neurodevelopment Laboratory<br />

Dr. Mengqing Xiang came to CABM in September 1996 from the Johns<br />

Hopkins University School of Medicine where he conducted postdoctoral<br />

studies with Dr. Jeremy Nathans. He earned his Ph.D. at the University of<br />

Texas M.D. Anderson Cancer <strong>Center</strong> <strong>and</strong> has received a number of honors<br />

including a China-U.S. Government Graduate Study Fellowship, a Howard<br />

Hughes Medical Institute Postdoctoral Fellowship, a Basil O’Connor Starter<br />

Scholar Research Award <strong>and</strong> a Sinsheimer Scholar Award. In 2003 he<br />

received the Award in Auditory Science from the National Organization <strong>for</strong><br />

Hearing Research Foundation. His work is currently supported by NIH.<br />

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

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

variety of molecular genetic approaches to identify <strong>and</strong> study transcription <strong>and</strong> other<br />

regulatory factors that are required <strong>for</strong> programming development of the retina, inner<br />

ear, spinal cord, <strong>and</strong> other CNS areas. A major focus of our work is to develop<br />

animal models to study the roles of these regulatory genes during normal<br />

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

cause sensorineural disorders such as blindness <strong>and</strong> deafness.<br />

Generation of a transgenic line <strong>for</strong> studying V2 neuronal lineages <strong>and</strong> functions<br />

in the spinal cord. During spinal neurogenesis, the p2 progenitor domain generates<br />

at least three subclasses of interneurons named V2a, V2b <strong>and</strong> V2c, which are crucial<br />

components of the locomotor central pattern generator. We have previously shown<br />

that the winged-helix/<strong>for</strong>khead transcription factor Foxn4 is expressed in a subset of<br />

p2 progenitors <strong>and</strong> required <strong>for</strong> specifying V2b interneurons. To determine the cell<br />

lineages derived from Foxn4-expressing progenitors, we generated a Foxn4-Cre<br />

BAC transgenic mouse line that drives Cre recombinase expression, mimicking<br />

endogenous Foxn4 expression pattern in the developing spinal cord. We used this<br />

transgenic line to map neuronal lineages derived from Foxn4-expressing progenitors<br />

<strong>and</strong> found that they gave rise to all neurons of the V2a, V2b as well as the newly<br />

identified V2c lineages. These data suggest that Foxn4 may be transiently expressed<br />

by all p2 progenitors <strong>and</strong> that the Foxn4-Cre line may serve as a useful genetic tool<br />

not only <strong>for</strong> lineage analysis but also <strong>for</strong> functional studies of genes <strong>and</strong> neurons<br />

involved in locomotion.<br />

35


Diverse developmental roles of Foxn4. In our previous studies, we have shown that<br />

Foxn4 is transiently expressed in a subset of progenitors during retinogenesis <strong>and</strong><br />

spinal neurogenesis. Targeted inactivation <strong>and</strong> gain-of-function analyses have<br />

revealed an essential function of Foxn4 in the generation of amacrine <strong>and</strong> horizontal<br />

cells during retinal development as well as in specifying the V2b interneurons during<br />

spinal cord development. In collaboration with Drs. Stavros Malas (University of<br />

Cyprus) <strong>and</strong> William Richardson (University College London), we have now used<br />

genetic fate mapping <strong>and</strong> loss-of-function analysis to show that the transcription<br />

factor Sox1 is expressed in <strong>and</strong> required <strong>for</strong> a third type of p2-derived interneuron,<br />

which we named V2c. These are close relatives of V2b interneurons, <strong>and</strong>, in the<br />

absence of Sox1, they switch to the V2b fate. The absence of Foxn4 results in a<br />

complete loss of Sox1-expressing V2c neurons, indicating that Foxn4 is necessary <strong>for</strong><br />

specifying not only V2b but also V2c interneuron subtypes. In another collaboration<br />

with Dr. Jeffrey Goldberg (University of Miami, Miller School of Medicine), we<br />

observed a developmental delay in the distribution of RGC (retinal ganglion cell)<br />

projections to the superior colliculus. Moreover, RGC axons failed to penetrate into<br />

the retinorecipient layers in Foxn4 mutant mice. Foxn4 was not expressed by RGCs<br />

<strong>and</strong> was undetectable in the superior colliculus itself. Thus, Foxn4 may be indirectly<br />

required <strong>for</strong> RGC distal axon patterning. Aside from its neural expression, we have<br />

also shown that during murine lung development Foxn4 is expressed in proximal<br />

airways by a subpopulation of postmitotic epithelial cells which are distinct from<br />

basal <strong>and</strong> ciliated cells. Foxn4 inactivation causes dilated alveoli, thinned alveolar<br />

walls <strong>and</strong> reduced septa in the distal lung but no overt gross alterations in proximal<br />

airways, suggesting that Foxn4 may have a non-cell-autonomous role critical <strong>for</strong><br />

alveologenesis during lung development. Together, our data suggest that Foxn4 may<br />

play multiple cell-autonomous <strong>and</strong> non-cell-autonomous roles during epithelial cell<br />

development.<br />

Brn3a regulates dorsal root ganglion sensory neuron specification <strong>and</strong> axonal<br />

projection into the spinal cord. The sensory neurons of the dorsal root ganglia<br />

(DRG) must project accurately to their central targets to convey proprioceptive,<br />

nociceptive <strong>and</strong> mechanoreceptive in<strong>for</strong>mation to the spinal cord. How these different<br />

sensory modalities <strong>and</strong> central connectivities are specified <strong>and</strong> coordinated still<br />

remains unclear. Given the expression of the POU homeodomain transcription factors<br />

Brn3a <strong>and</strong> Brn3b in DRG <strong>and</strong> spinal cord sensory neurons, we analyzed subtype<br />

specification of DRG <strong>and</strong> spinal cord sensory neurons as well as DRG central<br />

projections in Brn3a <strong>and</strong> Brn3b single <strong>and</strong> double mutant mice. Inactivation of either<br />

or both genes causes no gross abnormalities in early spinal cord neurogenesis;<br />

however, in Brn3a single <strong>and</strong> Brn3a;Brn3b double mutant mice, sensory afferent<br />

axons from the DRG fail to <strong>for</strong>m normal trajectories in the spinal cord. The TrkA +<br />

cutaneous nociceptive afferents stay outside the dorsal horn <strong>and</strong> fail to extend into the<br />

spinal cord, while the projections of TrkC + proprioceptive afferents into the ventral<br />

horn are also impaired. Moreover, Brn3a mutant DRGs are defective in sensory<br />

neuron specification, as marked by the excessive generation of TrkB + <strong>and</strong> TrkC +<br />

neurons as well as TrkA + /TrkB + <strong>and</strong> TrkA + /TrkC + double positive cells at early<br />

embryonic stages. At later stages in the mutant, TrkB + , TrkC + <strong>and</strong> parvalbumin +<br />

36


neurons diminish while there is a significant increase of GRP + <strong>and</strong> c-ret + neurons.<br />

In addition, Brn3a mutant DRGs display a dramatic down-regulation of Runx1<br />

expression, suggesting that the regulation of DRG sensory neuron specification by<br />

Brn3a is mediated in part by Runx1. Our data together demonstrate a critical role <strong>for</strong><br />

Brn3a in generating DRG sensory neuron diversity <strong>and</strong> regulating sensory afferent<br />

projections to the central targets.<br />

Altered TrkA + <strong>and</strong> TrkC + central afferent projection patterns in Brn3a mutant<br />

spinal cords at E12.5. The bundles of TrkA + <strong>and</strong> TrkC + afferent fibers are detected<br />

by immunostaining from lateral to medial regions of the dorsal horn edge in Brn3a +/-<br />

spinal cords at E12.5. In Brn3a -/- spinal cords, TrkA + <strong>and</strong> TrkC + fibers are narrowly<br />

located in a much smaller region, <strong>and</strong> extensively overlap with each other.<br />

Publications:<br />

Jin, K., Jiang, H., Mo, Z., <strong>and</strong> Xiang, M. Early B-cell factors are required <strong>for</strong> specifying<br />

multiple retinal cell types <strong>and</strong> subtypes from postmitotic precursors. J. Neurosci. 2010<br />

30:11902-11916.<br />

Panayi, H., Or<strong>for</strong>d, M., Panayiotou, E., Genethliou, N., Mean, R., Lapathitis, G., Li, S.,<br />

Xiang, M., Kessaris, N., Richardson, W., <strong>and</strong> Malas, S. SOX1 is required <strong>for</strong> the<br />

specification of a novel p2-derived interneuron subtype in the mouse ventral spinal cord. J.<br />

Neurosci. 2010 30:12274-12280.<br />

Li, S., Misra, K., <strong>and</strong> Xiang, M. A Cre transgenic line <strong>for</strong> studying V2 neuronal lineages<br />

<strong>and</strong> functions in the spinal cord. Genesis 2010 48:667–672.<br />

Li, S. <strong>and</strong> Xiang, M. Foxn4 influences alveologenesis during lung development. Dev.<br />

Dyn. <strong>2011</strong> 240:1512-1517.<br />

37


Kunzevitzky, N. J., Almeida, M. V., Duan, Y., Li, S., Xiang, M., <strong>and</strong> Goldberg, J. L.<br />

Foxn4 is required <strong>for</strong> retinal ganglion cell distal axon patterning. Mol. Cell. Neurosci.<br />

<strong>2011</strong> 46:731-741.<br />

Jin, K. <strong>and</strong> Xiang, M. Ebf1 deficiency causes increase of Müller cells in the retina <strong>and</strong><br />

abnormal topographic projection at the optic chiasm. Biochem. Biophys. Res. Commun.<br />

<strong>2011</strong> 414:539-544.<br />

Xiang, M., Jiang, H., Jin, K., <strong>and</strong> Qiu, F. Molecular control of retinal ganglion cell<br />

specification <strong>and</strong> differentiation. In Glaucoma - Basic <strong>and</strong> Clinical Concepts (R. Shimon,<br />

ed.),<strong>2011</strong> pp. 65-84, InTech, Rijeka.<br />

Jin, K. <strong>and</strong> Xiang, M. In vitro explant culture <strong>and</strong> related protocols <strong>for</strong> the study of<br />

mouse retinal development. In Methods in Molecular Biology. Springer, New York, NY.<br />

<strong>2011</strong> (in press).<br />

Pöschl, J., Lorenz, A., von Bueren, A.O., Peraud, A., Li, S., Tonn, J.-C., Herms, J.,<br />

Xiang, M., Rutkowski, S., Kretzschmar, H.A., <strong>and</strong> Schüller, U. Expression of BARHL1<br />

in medulloblastoma is associated with prolonged survival in mice <strong>and</strong> humans.<br />

Oncogene <strong>2011</strong>(in press).<br />

Ding, Q., Joshi. P. S., Xie, Z.-H., Xiang, M., <strong>and</strong> Gan, L. BARHL2 regulates the<br />

ipsilateral/contralateral subtype divergence in postmitotic dI1 neurons of the developing<br />

spinal cord. Proc. Natl. Acad. Sci. USA <strong>2011</strong> (in press).<br />

38


Laboratory Faculty Director<br />

Structural Biology<br />

Protein Engineering Stephen Anderson 40<br />

Biomolecular Crystallography Edward Arnold 42<br />

Structural Biology <strong>and</strong> Bioin<strong>for</strong>matics Helen Berman 47<br />

Structural Microbiology Joseph Marcotrigiano 51<br />

Structural Bioin<strong>for</strong>matics Gaetano Montelione 54<br />

Protein Design <strong>and</strong> Evolution Vikas N<strong>and</strong>a 62<br />

Protein Crystallography Ann Stock 65<br />

39


Dr. Stephen Anderson<br />

CABM Resident Faculty Member<br />

Associate Professor<br />

Department of Molecular Biology<br />

<strong>and</strong> Biochemistry<br />

Rutgers University<br />

Yi-Wen Chiang<br />

Lab Researcher<br />

Protein Engineering Laboratory<br />

Dr. Stephen Anderson conducted his postdoctoral research under Nobel<br />

laureate Dr. Frederick Sanger at the MRC Laboratory of Molecular Biology<br />

in Cambridge, Engl<strong>and</strong>. That project involved the sequencing of human <strong>and</strong><br />

bovine mitochondrial genomes. He went on to a position at the Cali<strong>for</strong>niabased<br />

biotechnology start-up company Genentech. Anderson has held<br />

research or teaching positions at Harvard University, the MRC Laboratory<br />

of Molecular Biology, Genentech, Inc., University of Cali<strong>for</strong>nia, San<br />

Francisco, <strong>and</strong> Rutgers University. While at Genentech he was responsible<br />

<strong>for</strong> all specialty chemical projects <strong>and</strong> second generation tissue plasminogen<br />

activator research.<br />

In <strong>2011</strong> the lab embarked on a three-year $2.8M NIH-funded project to express a<br />

representative set of antigens derived from human transcription factors. The<br />

collaborators <strong>and</strong> co-PIs on this project are Profs. Gaetano T. Montelione <strong>and</strong> Joseph<br />

Marcotrigiano, both of CABM <strong>and</strong> Rutgers, <strong>and</strong> Prof. Cheryl Arrowsmith of the<br />

Ontario Cancer Research <strong>Center</strong> <strong>and</strong> the University of Toronto. Late in <strong>2011</strong>, NIH<br />

identified two “affinity capture reagent” consortia <strong>for</strong> our antigen production group<br />

to partner with: one, headed by Prof. Anthony Kossiakoff at the University of<br />

Chicago <strong>and</strong> including groups at UCSF <strong>and</strong> University of Toronto, that will be<br />

making Fabs <strong>and</strong> antibodies using high throughput phage display technology; <strong>and</strong><br />

one based at Johns Hopkins University, headed by Prof. Jef Boike, that has invented<br />

a low-cost, high throughput method of isolating <strong>and</strong> screening traditional<br />

monoclonal antibodies.<br />

NIH’s overarching goal is to eventually produce renewable cognate affinity capture<br />

reagents (including antibodies) directed against every human protein, an ef<strong>for</strong>t that<br />

has been unofficially dubbed the “Human Anti-Proteome Project”. NIH wants to<br />

make such reagents available to the research community as powerful tools <strong>for</strong><br />

studying human biology. A description of this initiative is at<br />

http://commonfund.nih.gov/proteincapture/index.aspx. The ef<strong>for</strong>t that we are<br />

undertaking, namely producing human transcription factor antigens <strong>for</strong> affinity<br />

capture reagent production, is a pilot project designed to kick-start this overall ef<strong>for</strong>t.<br />

The first envisioned application <strong>for</strong> the anti-TF antibodies is to enable chromatin<br />

immunoprecipitation (ChIP) studies aimed at obtaining a complete map of potential<br />

transcription start sites in the human genome – much of this work is being carried<br />

out by the NIH-funded ENCODE Consortium.<br />

40


Human transcription factors are typically complex, multidomain proteins, <strong>and</strong> in<br />

terms of numbers of unique genes represent roughly 10% of the entire human<br />

proteome. Our goal is to express native, folded, individual domains from these<br />

different transcription factors with the hope that optimal specificity <strong>and</strong> affinity may<br />

be obtained by raising affinity capture reagents agains 3D epitopes. In the first year<br />

the Rutgers group has designed single-domain expression constructs using advanced<br />

“domain parsing” software developed by Dr. Janet Huang <strong>and</strong> Prof. Gaetano T.<br />

Montelione (see figure below). Together with Dr. Thomas Acton <strong>and</strong> Prof.<br />

Montelione, we have also developed a new series of high-efficiency E. coli vectors<br />

using “transcript optimized expression-enhancement technology” (“TOEET” – patent<br />

applied <strong>for</strong>). These vectors enable unprecedented levels of expression <strong>and</strong> solubility<br />

to be achieved. In <strong>2011</strong> the group cloned domains from nearly 1400 separate human<br />

transcription factors in >2700 distinct constructs <strong>and</strong> expression-tested approximately<br />

80% of these at small scale. We are now moving on to scale-up <strong>and</strong> production of<br />

multi-milligram quantities of purified antigen <strong>for</strong> our affinity capture reagent partners.<br />

In December, <strong>2011</strong>, we shipped our first batch of purified antigens.<br />

Fig. 1. Example of a portion of the output of the DisMeta server <strong>and</strong> how it can<br />

be used to analyze transcription factor sequences <strong>for</strong> structural in<strong>for</strong>mation.<br />

Shown schematically is a portion of the sequence of a human transiption factor, IF –<br />

16 (γ-interferon-inducible protein 16), analyzed by the DisMeta server.<br />

In this segment of the sequence the server predicts two regions that<br />

appear to have a complex, folded, tertiary structure separated by a<br />

region of polypeptide chain that is predicted to be relatively disordered.<br />

Three-dimentional structures <strong>for</strong> domains represented by the two<br />

ordered regions shown were determined by the NESG. The leftmost<br />

structure (PDB ID 2oq0) is a HIN-200/IF120x domain (InterPro ID<br />

IPR004021), <strong>and</strong> the rightmost structure (PDB ID 3b6y) is a pyrin<br />

domain (InterPro ID IPR004020) (Liao et al, unpulished). This example<br />

illustrates how the DisMeta server can be used to scan TF protein<br />

sequences to find the regions encoding domains that are likely to have<br />

3D epitopes.<br />

41


Dr. Eddy Arnold<br />

CABM Resident Faculty Member<br />

Board of Governors Professor<br />

Department of Chemistry <strong>and</strong><br />

Chemical Biology<br />

Rutgers University<br />

Adjunct Professor<br />

Department of Molecular Genetics<br />

Microbiology <strong>and</strong> Immunology<br />

UMDNJ-RWJMS<br />

Dr. Kalyan Das<br />

Research Professor<br />

Dr. Joseph Bauman<br />

Assistant Research Professor<br />

Dr. Daniel Himmel<br />

Research Associate<br />

Dr. Sergio Martinez<br />

Research Associate<br />

Dr. Matthew Miller<br />

Research Associate<br />

Dr. Steven Tuske<br />

Research Associate<br />

Dr. Mary Ho<br />

Postdoctoral Associate<br />

Dr. William Ho<br />

Postdoctoral Associate<br />

Biomolecular Crystallography Laboratory<br />

Dr. Eddy Arnold obtained his Ph.D. in organic chemistry with Professor Jon Clardy<br />

at Cornell University in 1982. From 1982 to 1987, he was a postdoctoral researcher<br />

with Professor Michael G. Rossmann at Purdue University <strong>and</strong> was a central member<br />

of the team that solved the structure of a human common cold virus by X-ray<br />

crystallography. Among the awards <strong>and</strong> fellowships Arnold has received are a<br />

National Science Foundation Predoctoral Fellowship, Damon Runyon–Walter<br />

Winchell <strong>and</strong> National Institutes of Health Postdoctoral Fellowships, an Alfred P.<br />

Sloan Research Fellowship, a Johnson <strong>and</strong> Johnson Focused Giving Award, <strong>and</strong> a<br />

Board of Trustees Award <strong>for</strong> Excellence in Research at Rutgers. He received an NIH<br />

MERIT Award in 1999 <strong>and</strong> a second consecutive one again in 2009. He was elected a<br />

Fellow of the American Association <strong>for</strong> the Advancement of Science in 2001 <strong>and</strong> a<br />

Fellow of the American Academy of Microbiology in 2006. In 2010 Dr. Arnold was<br />

named a Board of Governors Professor at Rutgers University. His laboratory is<br />

supported by grants from NIH <strong>and</strong> industrial collaborators, <strong>and</strong> by research<br />

fellowships. Dr. Arnold is involved in numerous international collaborations aimed<br />

at developing drugs <strong>for</strong> treatment <strong>and</strong> prevention of global infectious diseases,<br />

including HIV/AIDS, flu, <strong>and</strong> tuberculosis. His team has contributed to the discovery<br />

of two drugs used to treat HIV infection.<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<br />

approaches to the treatment, cure <strong>and</strong> prevention of human disease. A broad base of<br />

advances in chemistry, biology, <strong>and</strong> medicine has led to an exciting era in which<br />

knowledge of the intricate structure of life’s machinery can help to accelerate the<br />

development of new small molecule drugs <strong>and</strong> biomaterials such as engineered viral<br />

vaccines. Drs. Eddy Arnold <strong>and</strong> his colleagues are working to underst<strong>and</strong> molecular<br />

mechanisms of drug resistance <strong>and</strong> apply structure-based drug design <strong>for</strong> the treatment of<br />

serious human diseases. In pursuit of these goals, the laboratory uses research tools from<br />

diverse fields, including X-ray crystallography, molecular biology, virology, protein<br />

biochemistry, <strong>and</strong> macromolecular engineering. Eddy’s team of very experienced <strong>and</strong><br />

gifted coworkers, many of whom have been in the lab 10 years or longer, is the driving<br />

<strong>for</strong>ce behind the continuing progress.<br />

Since its establishment at CABM in 1987, our laboratory has studied the structure <strong>and</strong><br />

function of reverse transcriptase (RT), an essential component of the AIDS virus <strong>and</strong> the<br />

target of most widely used anti-AIDS drugs. Using the powerful techniques of X-ray<br />

crystallography, we have solved the three-dimensional structures of HIV-1 RT in complex<br />

with a variety of antiviral drugs <strong>and</strong> model segments of the HIV genome. These studies<br />

have revealed the workings of an intricate <strong>and</strong> fascinating biological machine in<br />

42


atomic detail <strong>and</strong> have yielded numerous novel insights into polymerase structure-function<br />

relationships, detailed mechanisms of drug inhibition <strong>and</strong> resistance, <strong>and</strong> structure-based<br />

design of RT inhibitors. The team has solved a variety of crystal structures representing<br />

multiple functional states of HIV-1 RT. These structures include HIV-1 RT in complex with a<br />

double-str<strong>and</strong>ed DNA template-primer, HIV-1 RT complexes with RNA:DNA templateprimers,<br />

structures of RT with AZTMP-terminated primer representing pre-translocation <strong>and</strong><br />

post-translocation complexes, <strong>and</strong> ternary complexes of wild-type <strong>and</strong> drug-resistant RT with<br />

DNA, <strong>and</strong> AZT-triphosphate/tenofovir-diphosphate. We have also determined the structures<br />

of numerous non-nucleoside inhibitors with wild-type <strong>and</strong> drug resistant HIV-1 RT, <strong>and</strong> the<br />

structural in<strong>for</strong>mation was used in the design of two recently approved non-nucleoside drugs.<br />

Also, we have obtained structures of RT:RNase H inhibitor <strong>and</strong> RT:DNA:AZTppppA (an<br />

ATP-mediated AZT excision product) complexes (Tu et al., 2010).<br />

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

immense <strong>and</strong> highly coordinated resources. The Arnold group has been <strong>for</strong>tunate to have<br />

successful long-term collaborations with the groups of Stephen Hughes (NIH NCI-Frederick),<br />

Roger Jones (Rutgers), Michael Parniak (U. of Pittsburgh), <strong>and</strong> Ronald Levy (Rutgers). The<br />

group also benefits from generous access to synchrotron X-radiation sources (CHESS, APS,<br />

<strong>and</strong> BNLS). Hughes <strong>and</strong> his coworkers have contributed expertise in protein engineering,<br />

production, <strong>and</strong> biochemistry at every stage of the RT project since its inception.<br />

Through collaboration with the late Dr. Paul Janssen we participated in a structure-based drug<br />

design ef<strong>for</strong>t that resulted in the discovery <strong>and</strong> development of non-nucleoside inhibitors<br />

(diarylpyrimidine, or DAPY analogs) with high potency against all known drug-resistant<br />

variants of HIV-1 RT. Crystallographic work from the Arnold <strong>and</strong> Hughes laboratories<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 successfully<br />

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

improved potency against wild-type <strong>and</strong> drug-resistant HIV-1 strains. Scientists at Tibotec, a<br />

subsidiary of Johnson & Johnson, tested the compounds <strong>for</strong> antiviral activity against wild-type<br />

<strong>and</strong> resistant HIV-1, <strong>and</strong> have led the clinical trials.<br />

The DAPY compounds are simple, inexpensive to make <strong>and</strong> have near ideal pharmacological<br />

properties. Etravirine (TMC125/Intelence) was approved <strong>for</strong> treatment of HIV infection by<br />

the FDA in 2008, <strong>and</strong> rilpivirine (TMC278) was approved as Edurant in May <strong>2011</strong>. In what<br />

may be unprecedented <strong>for</strong> a new best-in-class drug, Johnson & Johnson is permitting<br />

rilpivirine to be available in generic <strong>for</strong>m immediately in developing nations; this will make<br />

the drug available to millions of people. The prototypical DAPY compound,<br />

TMC120/dapivirine, is now being developed as a microbicide <strong>for</strong> blocking sexual<br />

transmission of HIV-1. A broader outcome of this study is a drug design concept <strong>for</strong><br />

overcoming drug resistance; the strategic flexibility that permits the DAPY compounds to<br />

“wiggle” <strong>and</strong> “jiggle” in a binding pocket to accommodate mutations apparently accounts <strong>for</strong><br />

their potency against a wide range of drug-resistant variants. Through a systematic protein<br />

engineering ef<strong>for</strong>t we obtained high-resolution crystals of HIV-1 RT <strong>and</strong> demonstrated that<br />

strategic flexibility of rilpivirine was responsible <strong>for</strong> its resilience against drug-resistant RT<br />

variants. Recent ef<strong>for</strong>ts include using the high-<br />

Dr. Ramaswamy<br />

S. Vijayan<br />

Postdoctoral Associate<br />

Dr. Angela<br />

McKoy<br />

Postdoctoral Fellow<br />

Dabyaben Patel<br />

Graduate Student<br />

Joseph Thomas<br />

Eck<br />

Graduate Fellow<br />

Arthur Clark<br />

Laboratory Researcher<br />

43


esolution HIV-1 RT crystals <strong>for</strong> drug-like fragment screening. A number of novel<br />

allosteric sites <strong>for</strong> inhibition of both polymerase <strong>and</strong> RNase H activity have been<br />

identified from the fragment screening ef<strong>for</strong>t.<br />

In addition to working to study HIV-1 RT <strong>and</strong> to develop chemotherapeutic agents, the<br />

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

Other projects currently being pursued in the lab include structural studies of: 1) bacterial<br />

RNA polymerase holoenzyme complexes with inhibitors <strong>and</strong> substrates in collaboration<br />

with Dr. Richard Ebright at Rutgers University; 2) the human mRNA capping enzyme <strong>and</strong><br />

its associated factors with Dr. Aaron Shatkin at CABM, <strong>and</strong> 3) influenza virus proteins<br />

including the polymerase from 2009 H1N1 p<strong>and</strong>emic strain.<br />

Figure from Das et al., 2012 showing nevirapine bound in the presence of DNA. The study<br />

confirms that nonnucleoside drugs displace the primer terminus from the polymerase active<br />

site. This structure, which had been elusive <strong>for</strong> decades, represents one of the key missing<br />

pieces in the HIV-1 RT structure/function puzzle.<br />

44


Figure from Das et al., <strong>2011</strong> showing how the nonnucleoside inhibitor TSAO has a<br />

shape that resembles a dragon when bound to HIV-1 RT.<br />

Publications:<br />

Das, K. J.M. Aramini, L.-C. Ma, R.M. Krug, <strong>and</strong> E. Arnold. Recent advances in structural<br />

studies of influenza A proteins: insights into antiviral drug targets. Nat. Struct. Mol. Biol.<br />

2010 17:530-538.<br />

Lapelosa, M., E. Gallicchio, G.F. Arnold, E. Arnold, <strong>and</strong> R.M. Levy. Antigenic<br />

characteristics of rhinovirus chimeras designed in silico <strong>for</strong> enhanced presentation of HIV-<br />

1 gp41 epitopes. J. Mol. Biol. 2010 397:752-766.<br />

Tu, X., K. Das, Q. Han, J.D. Bauman, A.D. Clark, Jr., X. Hou, Y.V. Frenkel, B.L. Gaffney,<br />

R.A. Jones, P.L. Boyer, S.H. Hughes, S.G. Sarafianos, <strong>and</strong> E. Arnold. Structural basis of<br />

HIV-1 resistance to AZT. Nat. Struct. Mol. Biol. 2010 17:1202-1209. (This publication<br />

was the focus of a commentary by Scott, W.A. <strong>2011</strong>. Structures of Reverse Transcriptase<br />

Pre- <strong>and</strong> Post-Excision Complexes Shed New Light on HIV-1 AZT Resistance. Viruses<br />

3:20-25.)<br />

Das, K., J.D. Bauman, A.S. Rim, C. Dharia, A.D. Clark, Jr., M.J. Camarasa, J. Balzarini,<br />

<strong>and</strong> E. Arnold. Crystal structure of tert-butyldimethylsilyl-spiroaminooxathioledioxidethymine<br />

(TSAO-T) in complex with HIV-1 reverse transcriptase (RT) redefines the elastic<br />

limits of the non-nucleoside inhibitor-binding pocket. J. Med. Chem. <strong>2011</strong> 54:2727-2737.<br />

Chu, C., Das, K., J.R. Tyminski, J.D. Bauman, R. Guan, W. Qiu, G.T. Montelione, E.<br />

Arnold, <strong>and</strong> A.J. Shatkin. Structure of the guanylyltransferase domain of human capping<br />

enzyme. Proc. Natl. Acad. Sci. <strong>2011</strong> 108:10104-10108.<br />

45


Chung, S., D.M. Himmel, J.K. Jiang, K. Wojtak, J.D. Bauman, J.W. Rausch, J.A. Wilson,<br />

J.A. Beutler, C.J. Thomas, E. Arnold, <strong>and</strong> S.F. Le Grice. Synthesis, activity, <strong>and</strong> structural<br />

analysis of novel a-hydroxytropolone inhibitors of human immunodeficiency virus reverse<br />

transcriptase-associated ribonuclease H. J. Med. Chem. <strong>2011</strong> 54:4462-4473.<br />

Felts AK, K. Labarge K, J.D. Bauman, D.V. Patel, D.M. Himmel, E. Arnold, M.A. Parniak,<br />

R.M. Levy. Identification of alternative binding sites <strong>for</strong> inhibitors of HIV-1 ribonuclease H<br />

through comparative analysis of virtual enrichment studies. J Chem Inf Model. <strong>2011</strong><br />

51:1986-1998.<br />

Arnold, E., D.M. Himmel, <strong>and</strong> M.G. Rossmann. Co-editors of "International Tables <strong>for</strong> X-<br />

Ray Crystallography, Volume F: Crystallography of Biological Macromolecules, Second<br />

Edition.” <strong>2011</strong> Kluwer Academic Publisher, Dordrecht, xxix + 886 pages.<br />

Arnold, E., D.M. Himmel, <strong>and</strong> M.G. Rossmann. Overview of this volume. In "International<br />

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

Macromolecules, Second Edition" E. Arnold, D.M. Himmel, <strong>and</strong> M.G. Rossmann, editors.<br />

<strong>2011</strong> Kluwer Academic Publisher, Dordrecht.<br />

Arnold, E. Gazing into the crystal ball: perspectives <strong>for</strong> the future. In "International Tables<br />

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

Second Edition" E. Arnold, D.M. Himmel, <strong>and</strong> M.G. Rossmann, editors. <strong>2011</strong> Kluwer<br />

Academic Publisher, Dordrecht.<br />

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

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

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

Second Edition" E. Arnold, D.M. Himmel, <strong>and</strong> M.G. Rossmann, editors. <strong>2011</strong> Kluwer<br />

Academic Publisher, Dordrecht.<br />

Bauman, J.D., D. Patel, E. Arnold. Fragment screening <strong>and</strong> HIV therapeutics. Top. Curr.<br />

Chem. Oct 5. <strong>2011</strong> [Epub ahead of print]<br />

Das, K., S.E. Martinez, J.D. Bauman, E. Arnold. HIV 1 reverse transcriptase complex with<br />

DNA <strong>and</strong> nevirapine reveals nonnucleoside inhibition mechanism. Nat. Struct. Mol. Biol.,<br />

2012 in press.<br />

46


Dr. Helen M. Berman<br />

Board of Governors Professor<br />

Department of Chemistry <strong>and</strong><br />

Chemical Biology<br />

Interim Director<br />

Biological, Mathematical, <strong>and</strong><br />

Physical Sciences Interfaces<br />

(BioMaPS),<br />

Director<br />

RCSB Protein Data Bank<br />

Director<br />

PSI Structural Biology<br />

Knowledgebase<br />

Rutgers University<br />

Dr. Martha Quesada<br />

Research Professor<br />

Dr. John Westbrook<br />

Research Professor<br />

Dr. Feng Zukang<br />

Research Professor<br />

Dr. Catherine Lawson<br />

Associate Research Professor<br />

Dr. Shuchismita Dutta<br />

Assistant Research Professor<br />

Dr. Monica Sekharan<br />

Assistant Research Professor<br />

Dr. Jasmine Young<br />

Assistant Research Professor<br />

Structural Biology <strong>and</strong> Bioin<strong>for</strong>matics<br />

Dr. Berman's research area is structural biology <strong>and</strong> bioin<strong>for</strong>matics, with a<br />

special focus on protein-nucleic acid interactions. She is the founder of the<br />

Nucleic Acid Database, a repository of in<strong>for</strong>mation about the structures of<br />

nucleic acid-containing molecules; <strong>and</strong> is the co-founder <strong>and</strong> Director of the<br />

Protein Data Bank, the international repository of the structures of biological<br />

macromolecules. Professor Berman also leads the Protein Structure Initiative<br />

Structural Biology Knowledgebase. She is a Fellow of the American<br />

Association <strong>for</strong> the Advancement of Science <strong>and</strong> of the Biophysical Society,<br />

from which she received the Distinguished Service Award in 2000. A past<br />

president of the American Crystallographic Association, she is a recipient of<br />

the Buerger Award (2006) <strong>and</strong> a member of the inaugural class of ACA<br />

Fellows (<strong>2011</strong>). Dr. Berman received her A.B. in 1964 from Barnard College<br />

<strong>and</strong> a Ph.D. in 1967 from the University of Pittsburgh. She received the<br />

Department of Chemistry Alumni Award from the University of Pittsburgh in<br />

2010.<br />

The Protein Data Bank<br />

The Research Collaboratory <strong>for</strong> Structural Bioin<strong>for</strong>matics (RCSB) Protein Data Bank<br />

(PDB) supports scientific research <strong>and</strong> education worldwide by providing an essential<br />

resource of in<strong>for</strong>mation about biomolecular structures.<br />

The PDB archive is the single repository of in<strong>for</strong>mation about the 3D structures of<br />

large biological molecules, primarily proteins <strong>and</strong> nucleic acids. Scientists from<br />

around the world submit their data about their experiments to the PDB, which is then<br />

processed <strong>and</strong> made freely available in the PDB archive. The RCSB PDB, which is<br />

based at Rutgers <strong>and</strong> the University of Cali<strong>for</strong>nia, San Diego, manages the PDB<br />

archive as a member of the Worldwide PDB (wwPDB).<br />

The wwPDB organization (www.wwpdb.org) was <strong>for</strong>med to ensure that the PDB<br />

archive is <strong>and</strong> will be freely <strong>and</strong> publicly available to the global community. The<br />

wwPDB members (RCSB PDB, Protein Data Bank in Europe (PDBe), Protein Data<br />

Bank Japan (PDBj), <strong>and</strong> the BioMagResBank (BMRB)) host deposition, processing,<br />

<strong>and</strong> distribution centers <strong>for</strong> PDB data <strong>and</strong> collaborate on a variety of projects <strong>and</strong><br />

outreach ef<strong>for</strong>ts. As 'archive keeper' <strong>for</strong> the wwPDB, the RCSB PDB maintains the<br />

central repository of the PDB archive.<br />

The central activity of the wwPDB is the collection, validation, archiving <strong>and</strong><br />

distribution of high quality structural data to the scientific community on a timely<br />

basis. The systems developed by the RCSB PDB are reliable <strong>and</strong> stable to meet the<br />

challenges of high data rates <strong>and</strong> complex structures. As of November 8, <strong>2011</strong>, the<br />

PDB archive contains more than 77,000 entries.<br />

47


In <strong>2011</strong>, the wwPDB commemorated the PDB’s 40 th anniversary with a special<br />

symposium. The meeting was attended by 19 distinguished speakers,<br />

approximately 250 participants, <strong>and</strong> 93 poster presentations. 34 travel awards <strong>for</strong><br />

early career scientists were made using funds raised by the wwPDB. More<br />

in<strong>for</strong>mation about the meeting is available at<br />

http://meetings.cshl.edu/meetings/pdb40.shtml.<br />

The RCSB PDB’s website at www.pdb.org provides access to the PDB FTP<br />

archive as well as tools that help users search, analyze <strong>and</strong> visualize PDB<br />

structural data. In 2010, the RCSB PDB website had more than 5.6 million visits<br />

by users in 218 countries. On average, the website has more than 196,000 unique<br />

visitors each month. Additionally, 160,713,233 data files were downloaded from<br />

the FTP server in 2010.<br />

The website provides access to a relational database that integrates PDB data with<br />

related in<strong>for</strong>mation from external sources (such as journal abstracts, functional<br />

descriptors, sequence annotations, structure annotations, <strong>and</strong> taxonomy). Users<br />

can search <strong>for</strong> structures using simple searches (PDB ID, keyword, sequence, or<br />

author) <strong>and</strong> complex queries.<br />

PDB-101 is a unique view of the RCSB PDB that packages together the resources<br />

of interest to teachers, students, <strong>and</strong> the general public. Major topics include<br />

Molecule of the Month columns, a top-down exploration from high-level<br />

functional categories to PDB structures called Structural View of Biology, <strong>and</strong><br />

related Educational Resources <strong>and</strong> materials.<br />

The RCSB PDB group at Rutgers is involved with undergraduate <strong>and</strong> graduate<br />

education, <strong>and</strong> supports local K12 education with programs such as the protein<br />

modeling event at the New Jersey Science Olympiad, <strong>and</strong> K12 education<br />

throughout the world with online resources.<br />

Structural Genomics <strong>and</strong> the PSI Structural Biology Knowledgebase<br />

With the overarching goal of creating new knowledge about the interrelationships<br />

of sequence, structure, <strong>and</strong> function, the Protein Structure Initiative Structural<br />

Biology Knowledgebase (PSI SBKB, sbkb.org) is a free online resource that<br />

captures <strong>and</strong> highlights the structural <strong>and</strong> technical advances made by the PSI<br />

structural genomics ef<strong>for</strong>ts <strong>for</strong> use by the broader biological community. This<br />

in<strong>for</strong>mation is integrated with publicly available biological in<strong>for</strong>mation so that it<br />

can enable a better underst<strong>and</strong>ing of living systems <strong>and</strong> disease. Created in<br />

collaboration with the Nature Publishing Group, the Structural Biology<br />

Knowledgebase also provides a research library, highlights focused on new<br />

research advances <strong>and</strong> technical tips, the latest science news, <strong>and</strong> an events<br />

calendar to present a broader view of structural genomics <strong>and</strong> structural biology.<br />

Searches of the SBKB by sequence or PDB ID will yield a list of related protein<br />

structures from the Protein Data Bank, theoretical 3D models, associated<br />

Li Chen<br />

Research Associate<br />

Dr. Buvna<br />

Coimbatore-<br />

Narayanan<br />

Research Associate<br />

Dr. Luigi DiCostanzo<br />

Research Associate<br />

Dr. Margaret<br />

Gabanyi<br />

Research Associate<br />

Guanghua Gao<br />

Research Associate<br />

Saheli Ghosh<br />

Research Associate<br />

Dr. Sutapa Ghosh<br />

Research Associate<br />

Vladimir Guranovic<br />

Research Associate<br />

Dr. Brian Hudson<br />

Research Associate<br />

Aamir Jadoon<br />

Research Associate<br />

David Micallef<br />

Research Associate<br />

Dr. Ezra Peisach<br />

Research Associate<br />

Dr. Irina Persikova<br />

Research Associate<br />

Nomi Rom<br />

Research Associate<br />

Raul Sala<br />

Research Associate<br />

Raship Shah<br />

Research Associate<br />

48


Dr. Chenghua Shao<br />

Research Associate<br />

Dr. Lihua Tan<br />

Research Associate<br />

Dr. Yi-Ping Tao<br />

Research Associate<br />

Dr. Huanwang<br />

Yang<br />

Research Associate<br />

Christine Zardecki<br />

Research Associate<br />

Dr. Marina<br />

Zhuravleva<br />

Research Associate<br />

Kar Lun Chun<br />

Research Assistant<br />

Maria Voigt<br />

Research Assistant<br />

descriptions (annotations) from biological databases, as well as structural<br />

genomics protein target in<strong>for</strong>mation, experimental protocols, <strong>and</strong> the ability to<br />

order available DNA clones. Text searches will find technology reports <strong>and</strong><br />

publications that were created by the PSI’s high-throughput research ef<strong>for</strong>ts, as<br />

well as the monthly features <strong>and</strong> highlights from the website. Web tools that aid<br />

in bench top research, such as protein construct design, are also available. This<br />

in<strong>for</strong>mation can enable scientists by providing everything that is known about a<br />

protein sequence, including any experimental successes or failures, thus<br />

streamlining their own research ef<strong>for</strong>ts.<br />

The SBKB group also developed two entirely new resources <strong>for</strong> the Structural<br />

Genomics community. First, a new experimental data tracking resource,<br />

TargetTrack (sbkb.org/tt/), was created that merged two existing (<strong>and</strong> mostly<br />

redundant) <strong>and</strong> exp<strong>and</strong>ing to support new types of high-throughput <strong>and</strong><br />

biological research. Second, a “Metrics” resource was created to track the<br />

progress of the whole PSI program.<br />

The Unified Data Resource <strong>for</strong> Cryo Electron Microscopy<br />

The PDB archives large biological assemblies determined by cryo-electron<br />

microscopy (cryoEM), a maturing methodology in structural biology that<br />

bridges the gap between cell biology <strong>and</strong> the experimental techniques of X-ray<br />

crystallography <strong>and</strong> NMR. In addition to 3D density maps, cryoEM experiments<br />

often yield fitted coordinate models. Through an NIH/NIGMS-funded<br />

collaboration, a joint EM map <strong>and</strong> model deposition tool has been developed.<br />

The unified resource <strong>for</strong> deposition <strong>and</strong> retrieval network <strong>for</strong> cryoEM map,<br />

model, <strong>and</strong> associated metadata is available at EMDataBank.org. This work<br />

has been carried out in collaboration with the RCSB PDB at Rutgers, PDBe, <strong>and</strong><br />

the National <strong>Center</strong> <strong>for</strong> Macromolecular Imaging at Baylor College of Medicine.<br />

Publications:<br />

Dutta, S., Zardecki, C., Goodsell, D.S., <strong>and</strong> Berman, H.M. Promoting a<br />

Structural View of Biology <strong>for</strong> Varied Audiences: An Overview of RCSB PDB<br />

Resources <strong>and</strong> Experiences. Journal of Applied Crystallography. 2010 43:<br />

1224-1229.<br />

Lawson, C.L., Baker, M.L., Best, C., Bi, C., Dougherty, M., Feng, P., van<br />

Ginkel, G., Devkota, B., Lagerstedt, I., Ludtke, S.J., Newman, R.H., Oldfield,<br />

T.J., Rees, I., Sahni, G., Sala, R., Velankar, S., Warren, J., Westbrook, J.D.,<br />

Henrick, K., Kleywegt, G.J., Berman, H.M., Chiu, W. EMDataBank.org:<br />

unified data resource <strong>for</strong> CryoEM. Nucleic Acids Res. <strong>2011</strong> 39: D456-D464.<br />

49


Rose, P.W., Beran, B., Bi, C., Bluhm, W.F., Dimitropoulos, D., Goodsell, D.S.,<br />

Prlić, A., Quesada, M., Quinn, G.B., Westbrook, J.D., Young, J., Yukich, B.,<br />

Zardecki, C., Berman, H.M., Bourne, P.E., The RCSB Protein Data Bank:<br />

redesigned web site <strong>and</strong> web services. Nucleic Acids Res. <strong>2011</strong> 39: D392-D401.<br />

Bluhm, W.F., Beran, B., Bi, C., Dimitropoulos, D., Prlić, A., Quinn, G.B., Rose,<br />

P.W., Shah, C., Yukich, B., Berman, H.M., Bourne, P.E., Quality Assurance <strong>for</strong><br />

the Query <strong>and</strong> Distribution Systems of the RCSB Protein Data Bank. Database.<br />

<strong>2011</strong>.<br />

Rose, P.W., Bluhm, W.F., Beran, B., Bi, C., Dimitropoulos, D., Goodsell, D.S.,<br />

Prlić, A., Quinn, G.B., Yukich, B., Berman, H.M., Bourne, P.E. The RCSB<br />

Protein Data Bank: site functionality <strong>and</strong> bioin<strong>for</strong>matics use cases. NCI-Nature<br />

Pathway Interaction Database Bioin<strong>for</strong>matics Primer. <strong>2011</strong>.<br />

Gabanyi, M.J., Adams, P.D., Arnold, K., Bordoli, L., Carter, L.G., Flippen-<br />

Anderson, J., Gif<strong>for</strong>d, L., Haas, J., Kouranov, A., McLaughlin, W.A., Micallef,<br />

D.I., Minor, W., Shah, R., Schwede, T., Tao,YP.W., Westbrook, J.D.,<br />

Zimmerman, M., Berman, H.M. The Structural Biology Knowledgebase: A portal<br />

to protein structures, sequences, functions, <strong>and</strong> methods. Journal of Structural <strong>and</strong><br />

Functional Genomics <strong>2011</strong> 12:45-54.<br />

Beran, B., Bi, C., Bluhm, W.F., Dimitropoulos, D., Feng, Z., Goodsell, D.S., Prlic,<br />

A., Quinn, G., Rose, P., Westbrook, J., Yukich, B., Young, J., Zardecki, C.,<br />

Berman, H.M. The Evolution of the RCSB Protein Data Bank Website. WIREs<br />

Computational Molecular Science. <strong>2011</strong>.<br />

Berman, H.M. The Protein Data Bank: Evolution of a key resource in biology.<br />

Leadership in Science <strong>and</strong> Technology: A Reference H<strong>and</strong>book (William Sims<br />

Bainbridge, editor) SAGE Publications, Inc., <strong>2011</strong>.<br />

Kryshtafovych, A., Bartual, S., Bazan, J.F., Berman, H.M., Casteel, D.,<br />

Christodoulou, E., Everett, J., Hausmann, J., Heidebrecht, T., Hills, T., Hui, R.,<br />

Hunt, J., Jayaraman<strong>and</strong>, S., Joachimiak, A., Kennedy, M., Kim, C., Lingel, A.,<br />

Michalska, K., Montelione, G., Otero, J., Perrakis, O., Pizarro,J., vanRaaij,<br />

M.J., Ramelot, T., Rousseau, F., Wernimont, A., Young, J., Schwede, T. Target<br />

Highlights in CASP9: Experimental Target Structures <strong>for</strong> the Critical Assessment<br />

of Techniques <strong>for</strong> Protein Structure Prediction. PROTEINS: Structure, Function,<br />

<strong>and</strong> Bioin<strong>for</strong>matics, <strong>2011</strong> 79: 6–20.<br />

50


Dr. Joseph Marcotrigiano<br />

CABM Resident Faculty Member<br />

Assistant Professor<br />

Department of Chemistry <strong>and</strong><br />

Chemical Biology<br />

Rutgers University<br />

Dr. Abdul Khan<br />

Postdoctoral Assoc.<br />

Ankita Basant<br />

Graduate Student<br />

Fuguo Jiang<br />

Graduate Student<br />

Jillian Whidby<br />

Graduate Student<br />

Samantha Yost<br />

Graduate Student<br />

Structural Microbiology Laboratory<br />

Dr. Joseph Marcotrigiano obtained his B.A. from Rutgers with highest<br />

honors <strong>for</strong> research on the structure of HIV-1 RT <strong>and</strong> intergrase as a Henry<br />

Rutgers Undergraduate Scholar. He received the National Starch Award<br />

<strong>and</strong> the Bruce Garth Award <strong>for</strong> highest st<strong>and</strong>ing in the chemistry program<br />

<strong>and</strong> was selected <strong>for</strong> a graduate fellowship at Rockefeller University. He<br />

completed Ph.D. studies with Dr. Stephen Burley in 2000 <strong>and</strong> was awarded<br />

the inaugural David Rockefeller Jr. Alumni Fellowship. He conducted<br />

postdoctoral research as a Merck Fellow of the Life Sciences Research<br />

Foundation in the <strong>Center</strong> <strong>for</strong> the Study of Hepatitis C with Dr. Charles Rice<br />

at Rockefeller University. Dr. Marcotrigiano joined CABM in 2007.<br />

.<br />

Hepatitis C virus (HCV) continues to be a major public health problem. In most<br />

cases, HCV infection becomes chronic <strong>and</strong> can persist <strong>for</strong> decades, leading to<br />

cirrhosis, end-stage liver disease <strong>and</strong> hepatocellular carcinoma. Currently, 2% of the<br />

human population – approximately 123 million people – is infected with HCV. In<br />

fact, there are 3-4 times more individuals infected with HCV than HIV, making virus<br />

transmission a major public health concern. In the United States, HCV infection is<br />

the most common cause of liver transplantation <strong>and</strong> results in 10,000 to 20,000<br />

deaths a year. There is no vaccine, <strong>and</strong> current HCV therapy, pegylated interferonalpha<br />

in combination with ribavirin, leads to a sustained response in only 50% of<br />

genotype 1-infected patients, the prevalent genotype in the United States. The<br />

current HCV treatment stimulates the patient’s immune system to clear the virus, but<br />

numerous side effects cause many patients to prematurely stop treatment. Given the<br />

high prevalence of infection <strong>and</strong> poor response rate, inhibitors that specifically target<br />

HCV proteins with fewer side effects are desperately needed. In addition, an<br />

effective vaccine would greatly reduce the spread of the virus.<br />

Our laboratory studies how HCV enters a host cell <strong>and</strong> avoids the cellular innate<br />

immune response to infection. To elucidate these processes we employ a variety of<br />

structural, biophysical, biochemical <strong>and</strong> virological techniques. HCV is a member<br />

of the family Flaviviridae, which also includes Pestiviruses <strong>and</strong> Flaviviruses.<br />

51


The HCV virion consists of an enveloped nucleocapsid containing the viral genome, a<br />

single-str<strong>and</strong>ed, positive sense RNA that encodes a single open reading frame. Once<br />

the virus penetrates a permissive cell, the HCV genome is released into the cytosol<br />

where the viral RNA is translated in a cap-independent manner by an internal<br />

ribosome entry site (IRES) located within the 5’ nontranslated region (NTR).<br />

Translation generates a viral polyprotein that is proteolytically processed by cellular<br />

<strong>and</strong> viral encoded proteases into ten proteins. HCV is an enveloped virus with two<br />

glycoproteins (E1 <strong>and</strong> E2) that <strong>for</strong>m the outermost shell of the virion. These two<br />

proteins are thought to be involved in cell receptor binding, entry, membrane fusion<br />

<strong>and</strong> immune evasion. Both E1 <strong>and</strong> E2 are type I transmembrane proteins with an<br />

amino-terminal ectodomain <strong>and</strong> a carboxy-terminal membrane-associating region.<br />

The transmembrane regions are involved in ER retention <strong>and</strong> the <strong>for</strong>mation of<br />

noncovalent E1/E2 heterodimers. Since HCV is thought to bud into the ER, retention<br />

of the glycoproteins at the ER membrane ensures their placement on the virion<br />

particles. The E1 <strong>and</strong> E2 ectodomains are heavily glycosylated <strong>and</strong> contain several<br />

intramolecular disulfide bonds. E1 <strong>and</strong> E2 contain 4 <strong>and</strong> 11 predicted N-linked<br />

glycosylation sites, respectively, <strong>and</strong> many highly conserved cysteines.<br />

Intracellular double-str<strong>and</strong>ed RNA (dsRNA) is an important signal of virus replication.<br />

The host has developed mechanisms to detect viral dsRNA <strong>and</strong> initiate antiviral<br />

responses. Pathogen Recognition Receptors (PRR) are cellular proteins that sense the<br />

presence of pathogen associated molecular patterns (PAMPs) to induce an antiviral<br />

state. Retinoic acid inducible gene I (RIG-I) is one member of a family of PRR that<br />

resides within the cytoplasm of a cell <strong>and</strong> senses the presence of 5’ triphosphorylated<br />

dsRNA, an intermediate of virus replication. RIG-I encodes two caspase recruitment<br />

domains (CARD), a DExD/H box RNA helicase, <strong>and</strong> a repressor domain (RD). The<br />

RD blocks signaling by the CARD domains under normal growth conditions. To<br />

investigate the contributions of the individual domains of RIG-I to RNA binding, we<br />

determined the equilibrium dissociation constants (Kd) of the protein–RNA<br />

complexes. The tightest RNA affinity was observed with helicase-RD, whereas the<br />

full-length RIG-I, helicase domain <strong>and</strong> RD bind dsRNA with a 24-fold, 8,600-fold<br />

<strong>and</strong> 50-fold weaker affinity, respectively. Crystals of RIG-I helicase-RD in complex<br />

with ADP-BeF3 <strong>and</strong> 14 base-pair palindromic dsRNA were obtained <strong>and</strong> the structure<br />

was determined to 2.9Å resolution. RIG-I helicase-RD organizes into a ring around<br />

dsRNA, capping one end, while contacting both str<strong>and</strong>s using previously<br />

uncharacterized motifs to recognize dsRNA. Small-angle X-ray scattering, limited<br />

proteolysis <strong>and</strong> differential scanning fluorimetry indicate that RIG-I is in an extended<br />

<strong>and</strong> flexible con<strong>for</strong>mation that compacts upon binding RNA. These results provide a<br />

detailed view of the role of helicase in dsRNA recognition, the synergy between the<br />

RD <strong>and</strong> the helicase <strong>for</strong> RNA binding <strong>and</strong> the organization of full-length RIG-I bound<br />

to dsRNA. The results provide evidence of a con<strong>for</strong>mational change upon RNA<br />

binding.<br />

52


The long-term goals of our studies are to provide a structural <strong>and</strong> mechanistic<br />

underst<strong>and</strong>ing <strong>for</strong> how the HCV glycoproteins (E1 <strong>and</strong> E2) interact with each other<br />

<strong>and</strong> with cellular receptors. Also we are focused on how HCV evades the host<br />

antiviral response <strong>and</strong> how RIG-I discriminates viral from cellular RNAs.<br />

Publications:<br />

Structure of RIG-I helicase-<br />

RD bound to dsRNA <strong>and</strong><br />

ADP-BeF<br />

Jiang, F., Ramanathan, A., Miller, M.T., Tang, G.Q., Gale, M. Jr., Patel, S.S.,<br />

Marcotrigiano, J. Structural basis of RNA recognition <strong>and</strong> activation by innate<br />

immune receptor RIG-I. Nature <strong>2011</strong> 479(7373):423-7<br />

Whidby, J., Mateu, G., Scarborough, H., Demeler, B., Grakoui, A. <strong>and</strong><br />

Marcotrigiano, J.<br />

Blocking hepatitis C virus infection with recombinant <strong>for</strong>m of envelope protein 2<br />

ectodomain. J. Virol. 2009 83(21):11078-89.<br />

Saito, T., Owen, D.M., Jiang, F., Marcotrigiano, J., Gale, M. Jr. Innate immunity<br />

induced by composition-dependent RIG-I recognition of hepatitis C virus RNA.<br />

Nature 2008 454(7203):523-7<br />

53


Dr. Gaetano Montelione<br />

CABM Resident Faculty Member<br />

Jerome <strong>and</strong> Lorraine Aresty Chair in<br />

Cancer Biology Research<br />

Professor II<br />

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

Biochemistry<br />

Rutgers University<br />

Adjunct Professor<br />

Department of Biochemistry<br />

UMDNJ-RWJMS<br />

Director<br />

Northeast Structural Genomics<br />

Consortium<br />

Dr. Swapna Gurla<br />

Associate Research Professor<br />

Dr. Yuanpeng Huang<br />

Associate Research Professor<br />

Dr. Roberto Tejero<br />

Visiting Professor<br />

Dr. Thomas Acton<br />

Assistant Research Professor<br />

Dr. James Aramini<br />

Assistant Research Professor<br />

Dr. John Everett<br />

Assistant Research Professor<br />

Dr. Rongjin Guan<br />

Assistant Research Professor<br />

Dr. Gregory Kornhaber<br />

Assistant Research Professor<br />

Structural Bioin<strong>for</strong>matics Laboratory<br />

Dr. Gaetano Montelione did graduate studies in protein physical chemistry with<br />

Professor Harold Scheraga at Cornell University. He learned nuclear magnetic<br />

resonance spectroscopy at the Swiss Federal Technical Institute in Zürich where he<br />

worked with Nobel laureate Dr. Kürt Wüthrich, the first researcher to solve a protein<br />

structure with this technique in 1985. Two years later, Dr. Montelione solved the<br />

structure of epidermal growth factor. He has developed new NMR techniques <strong>for</strong><br />

refining 3D structures of proteins <strong>and</strong> triple resonance experiments <strong>for</strong> making 1 H,<br />

13 C, <strong>and</strong> 15 N resonance assignments in intermediate-sized proteins. He has served as<br />

a member of the NSF Molecular Biophysics Study Section, NSF Committee of Visitors,<br />

NSF Advisory Committee <strong>for</strong> Biological Sciences (BIO AC) <strong>and</strong> advisor to the World<br />

Wide Protein Data Bank (WW_PDB). Dr. Montelione has received the Searle<br />

Scholar Award, the Dreyfus Teacher-Scholar Award, a Johnson <strong>and</strong> Johnson<br />

Research Discovery Award, the American Cyanamid Award in Physical Chemistry,<br />

the NSF Young Investigator Award, <strong>and</strong> the Michael <strong>and</strong> Kate Bárány Award of the<br />

Biophysical Society. He is an elected Fellow of the American Association <strong>for</strong> the<br />

Advancement of Science.<br />

As director of the NIH-funded Northeast Structural Genomics Consortium of the<br />

NIGMS Protein Structure Initiative, Dr. Montelione leads an inter-institutional pilot<br />

project in large-scale structural proteomics <strong>and</strong> bioin<strong>for</strong>matics. Goals of our work<br />

involve developing high-throughput technologies suitable <strong>for</strong> determining many new<br />

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

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

insights into the functions of novel gene products identified by genomic <strong>and</strong>/or<br />

bioin<strong>for</strong>matic analysis. The resulting knowledge of structure <strong>and</strong> biochemical function<br />

provides the basis <strong>for</strong> collaborations with academic laboratories <strong>and</strong> pharmaceutical<br />

companies to develop drugs useful in treating human diseases that are targeted to these<br />

newly discovered functions. The approach we are taking is opportunistic in the sense<br />

that only proteins which express well in certain expression systems are screened <strong>for</strong><br />

their abilities to provide high quality NMR spectra or well-diffracting protein crystals.<br />

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<br />

abilities to identify, clone, express <strong>and</strong> analyze several hundred biologically interesting<br />

proteins per year; only a fraction of the initial sequences chosen <strong>for</strong> cloning <strong>and</strong><br />

analysis result in high-resolution 3D structures. However, this “funnel” process is<br />

yielding three-dimensional structures <strong>and</strong> new functions <strong>for</strong> some 200 proteins per year,<br />

<strong>and</strong> can thus have tremendous scientific impact. Research areas include networks of<br />

proteins associated with human cancer biology, protein complexes involved in<br />

influenza virus infection, innate immune response, <strong>and</strong> ubiquitination pathways.<br />

54


s<br />

Publications:<br />

Singarapu, K.K., Mills, J., Xiao, R., Acton, T., Punta, M., Fischer, M., Honig, B., Rost, B.,<br />

Montelione, G.T., Szyperski, T. Solution NMR structures of proteins VPA0419 from<br />

Vibrio parahaemolyticus <strong>and</strong> yiiS from Shigelle flexneri provide structural coverage from<br />

protein domain family PFAM 04175. PROTEINS: Struc. Funct. Bioin<strong>for</strong>matics 2010 78:<br />

779 - 784.<br />

Mao, L., Vaiphei. S.T., Shimazu, T., Schneider, W.M., Tang, Y., Mani, R., Roth, M.J.,<br />

Montelione, G.T., Inouye, M. The E. coli single protein production (cSPP) system <strong>for</strong><br />

production <strong>and</strong> structural analysis of membrane proteins. J. Struct. Funct. Genomics 2010<br />

11: 81-84.<br />

Rossi, P., Swapna, G.V.T., Huang, Y.J., Aramini, J.M., Anklin, C., Conover, K., Hamilton,<br />

K., Xiao, R., Acton, T.B., Ertekin, A., Everett, J.K., Montelione, G.T. A microscale protein<br />

NMR sample screening pipeline. J. Biomol. NMR 2010 46: 11 - 22.<br />

Raman, S., Huang, Y.J., Rossi, P., Aramini, J.M., Liu, G., Mao, B., Montelione, G.T.,<br />

Baker, D. Accurate automated protein NMR structure determination using unassigned<br />

NOESY data. J. Am. Chem. Soc. 2010 132: 202 - 207.<br />

Raman, S., Lange, O.F., Rossi, P., Tyka, M., Wang, X., Aramini, J., Liu, G., Ramelot, T.,<br />

Eletsky, A., Szyperski, T., Kennedy, M., Prestegard, J., Montelione, G.T., Baker, D. NMR<br />

structure determination <strong>for</strong> larger proteins using backbone-only data. Science 2010 327:<br />

1014 - 1018.<br />

Dr. Gaohua Liu<br />

Assistant Research Professor<br />

Dr. Li-Chung Ma<br />

Assistant Research Professor<br />

Dr. Paolo Rossi<br />

Assistant Research Professor<br />

Dr. Rong Xiao<br />

Assistant Research Professor<br />

Dr. Yuefeng Tang<br />

Research Associate<br />

Dr. Asli Ertekin<br />

Postdoctoral Associate<br />

Binchen Mao<br />

Graduate Student<br />

Philip Kostenbader<br />

System Administrator<br />

Saichiu Tong<br />

Research Programmer<br />

Dongyan Wang<br />

Research Teaching Spec.<br />

Hoi Wun Au<br />

Laboratory Researcher<br />

Colleen Ciccosanti<br />

Laboratory Researcher<br />

Kenith Conover<br />

Laboratory Researcher<br />

Keith Hamilton<br />

Laboratory Researcher<br />

Haleema Janjua<br />

Laboratory Researcher<br />

Eitan Kohan<br />

Laboratory Researcher<br />

Dong Yup Lee<br />

Laboratory Researcher<br />

55


Melissa Maglaqui<br />

Laboratory Researcher<br />

Lei Mao<br />

Laboratory Researcher<br />

Dayaben Patel<br />

Laboratory Researcher<br />

Seema Sahdev<br />

Laboratory Researcher<br />

Ritu Shastry<br />

Laboratory Researcher<br />

Huang Wang<br />

Laboratory Researcher<br />

Ari Sapin<br />

Technician<br />

Liu, G., Xiao, R., Wang, D., Huang, Y.J., Acton, T.B., Montelione, G.T. NMR structure Factin<br />

binding domain of Arg/Ab12 from Homo sapiens. PROTEINS: Struct. Funct.<br />

Bioin<strong>for</strong>matics 2010 78: 1326 - 1330.<br />

Montelione, G.T., Szyperski, T. Advances in protein NMR impacting drug discovery<br />

provided by the NIGMS Protein Structure Initiative. Curr. Opin. Drug Discovery 2010 13:<br />

335 - 349.<br />

Arragain, S., Garcia-Serres, R., Blondin, G., Douki, T., Clemancey, M., Latour, J-M.;<br />

Forouhar, F., Neely, H., Montelione, G.T., Hunt, J.F., Mulliez, E., Fontecave, M., Atta, M.<br />

Post-translational modification of ribosomal proteins: Structural <strong>and</strong> functional<br />

characterization of RimO from Thermotoga maritima, a radical S-adenosylmethionine<br />

methylthiotransferase. J. Biol. Chem. 2010 285: 5792 - 5801.<br />

Aramini, J.M., Tubbs, J.L., Kanugula, S., Rossi, P., Belote, R.L., Ciccosanti, C.T., Jiang, M.,<br />

Xiao, R., Soong, T., Rost, B., Acton, T.B., Everett, J.K., Pegg, A.E., Tainer, J.A.,<br />

Montelione, G.T. Structural basis of O6-alkylguanine recognition by a bacterial<br />

alkyltransferase-like DNA repair protein. J. Biol. Chem. 2010 285: 13736 - 13741.<br />

Arbing, M.A., H<strong>and</strong>elman, S.K., Kuzin, A.P., Verdon, G., Wang, C., Su, M., Rothenbacher,<br />

F.P., Abashidze, M., Liu, M., Hurley, J.M., Xiao, R., Acton, T., Inouye, M., Montelione,<br />

G.T., Woychik, N.A., Hunt, J.F. Crystal structures of Phd-Doc, HigA, <strong>and</strong> YeeU establish<br />

multiple evolutionary links between microbial growth-regulating toxin-antitoxin systems.<br />

Structure 2010 18: 996 - 1010.<br />

Price, W.N., H<strong>and</strong>elman, S.K., Everett, J., Tong, S.N., Bracic, A., Luff, J.D., Naumov, V.,<br />

Acton, T., Manor, P., Xiao, R., Rost, B., Montelione, G.T., Hunt, J.F. Large-scale<br />

experimental studies show unexpected amino acid effects on protein expression <strong>and</strong><br />

solubility in vivo in E. Coli. Microbial In<strong>for</strong>matics <strong>and</strong> Experimentation <strong>2011</strong> 1: 6 - 26.<br />

Nie, Y., Xiao, R., Xu, Y., Montelione, G.T. Novel anti-prelog stereospecific carbonyl<br />

reductases from C<strong>and</strong>ida parapsilosis <strong>for</strong> asymmetric reduction of prochiral ketones. Org.<br />

Biomol. Chem. <strong>2011</strong> 9: 4070 - 4078.<br />

Schneider, W.M., Tang, Y., Vaiphei, S.T., Mao, L., Maglaqui, M., Inouye, M., Roth, M.J.,<br />

Montelione, G.T. Efficient condensed-phase production of perdeuterated soluble <strong>and</strong><br />

membrane proteins. J. Struct. Funct. Genomics 2010 11: 143 - 154.<br />

Liu, G., Huang, Y.J., Xiao, R., Wang, D., Acton, T.B., Montelione, G.T. Solution NMR<br />

structure of the ARID domain of human AT-rich interactive domain-containing protein 3A:<br />

A human cancer protein interaction network target. PROTEINS: Struct Funct<br />

Bioin<strong>for</strong>matics 2010 78: 2170 - 2175.<br />

56


Zhao, L., Zhao, K., Hurst, R., Slater, M., Acton, T.B., Swapna, G.V.T., Shastry, R.,<br />

Kornhaber, G.J., Montelione, G.T. Engineering of a wheat germ expression system to<br />

provide compatibility with a high throughput pET-based cloning plat<strong>for</strong>m. J. Struct. Funct.<br />

Genomics 2010 11: 210 - 209.<br />

Lee, H-W., Wylie, G., Bonsal, S., Wang, X., Barb, A.W., Macnaughtan, M.A., Ertekin, A.,<br />

Montelione, G.T., Prestegard, J.H. Three-dimensional structures of the weakly associated<br />

protein homodimer SeR13 using RDCs <strong>and</strong> paramagnetic surface mapping. Protein Science<br />

2010 19: 1673 - 1685.<br />

Stark, J.L., Mercier, K.A., Mueller, G.A., Acton, T.B., Xiao, R., Montelione, G.T., Powers, R.<br />

Solution structure <strong>and</strong> function of YndB, an AHSA1 protein from Bacillus subtilis.<br />

PROTEINS: Struc. Funct. Bioin<strong>for</strong>matics 2010 78: 3328 - 3340.<br />

Tang, Y., Xiao, R.; Ciccosanti, C., Janjua, H., Lee, Y.L., Everett, J., Swapna, G.V.T., Acton,<br />

T.B., Rost, B., Montelione, G.T. Solution NMR structure of Lin0431 protein from Listeria<br />

innocua reveals high structural similarity with domain II of bacterial transcription<br />

antitermination protein NusG. PROTEINS: Struc. Funct. Bioin<strong>for</strong>matics 2010 78: 2563 -<br />

2568.<br />

Xiao, R., Anderson, S., Aramini, J.M., Belote, R., Buchwald, W., Ciccosanti, C., Conover, K.,<br />

Everett, J.K., Hamilton, K., Huang, Y.J., Janjua, H., Jiang, M., Kornhaber, G.J., Lee, D.Y.,<br />

Locke, J.Y., Ma, L.-C., Maglaqui, M., Mao, L., Mitra, S., Patel, D., Rossi, P., Sahdev, S.,<br />

Sharma, S., Shastry, R., Swapna, G.V.T., Tong, S.N., Wang, D., Wang, H., Zhao, L.,<br />

Montelione, G.T., Acton, T.B. The high-throughput protein sample production plat<strong>for</strong>m of<br />

the Northeast Structural Genomics Consortium. J. Struct. Biol. 2010 172: 21 - 33.<br />

Tang, Y., Schneider, W.M., Shen, Y., Raman, S., Inouye, M., Baker, D., Roth, M.J.,<br />

Montelione, G.T. Fully automated high quality NMR structure determination of small 2Henriched<br />

proteins. J. Struct. Funct. Genomics 2010 11: 223 - 232.<br />

Yang, Y., Ramelot, T.A., Cort, J.R., Wang, D., Ciccosanti, C., Hamilton, K., Nair, R., Rost,<br />

B., Acton, T.B., Xiao, R., Everett, J.K., Montelione, G.T., Kennedy, M. Solution NMR<br />

structure of photosystem II reaction center protein Psb28 from Synechocystis sp. strain PCC<br />

6803. PROTEINS: Struc. Funct. Bioin<strong>for</strong>matics <strong>2011</strong> 79: 340 - 344.<br />

Yang, Y., Ramelot, T.A., McCarrick, R., Ni, S., Feldmann, E., Cort, J.R., Wang, D.,<br />

Ciccosanti, C., Jiang, M., Janjua, H., Acton, T.B., Xiao, R., Everett, J.K., Montelione, G.T.,<br />

Kennedy, M. Combining NMR <strong>and</strong> EPR methods <strong>for</strong> homo-dimer protein structure<br />

determination. J. Am. Chem. Soc. 2010 132: 11910 - 11913.<br />

Love, J., Mancia, F., Shapiro, L., Punta, M., Rost, B., Girvin, M., Wang, D-N., Zhou, M.,<br />

Hunt, J.F., Szyperski, T., Gouaux, E., MacKinnon, R., McDermott, A., Honig, B., Inouye, M.,<br />

Montelione, G.T., Hendrickson, W.A. The New York Consortium on Membrane Structure<br />

(NYCOMPS): A high-throughput plat<strong>for</strong>m <strong>for</strong> structural genomics of integral membrane<br />

proteins. J. Struct. Funct. Genomics 2010 11: 191 - 199.<br />

57


Mani, R., Vorobiev, S., Swapna, G.V.T., Neely, H., Janjua, H., Ciccosanti, C., Xiao, R.,<br />

Acton, T.B., Everett, J.K., Hunt, J.F., Montelione, G.T. Solution NMR <strong>and</strong> X-ray crystal<br />

structures of membrane-associated lipoprotein-17 domain reveal a novel fold. J. Struct.<br />

Funct. Genomics <strong>2011</strong> 12: 27 - 32.<br />

Zhang, H., Constantine, R., Vorobiev, S., Chen, Y., Seetharaman, J., Huang, Y.J., Xiao, R.,<br />

Montelione, G.T., Gerstner, C.D., Davis, M.W., Inana, G., Whitby, F.G., Jorgensen, E.M.,<br />

Hill, C.P., Tong, L., Baehr, W. UNC119 is required <strong>for</strong> G protein trafficking in sensory<br />

neurons. Nature Neuroscience <strong>2011</strong> 14: 874 - 880.<br />

Aramini, J.M., Rossi, P., Cort, J., Ma, L-C., Xiao, R., Acton, T.B., Montelione, G.T.<br />

Solution NMR structure of the plasmid-encoded fimbriae regulatory protein PefI from<br />

Salmonella enterica serovar Typhimurium. PROTEINS: Struc. Funct. Bioin<strong>for</strong>matics <strong>2011</strong><br />

79: 335 - 339.<br />

Forouhar, F., Lew, S., Seetharaman, J., Xiao, R., Acton, T.B., Montelione, G.T., Tong, L.<br />

Crystal structures of bacterial biosynthetic arginine decarboxylases. Acta Cryst. F. 2010<br />

F66: 1562 – 1566.<br />

Montelione, G.T., Szyperski, T. IOS Press, Editors A.J. Dingley <strong>and</strong> S.M. Pascal.<br />

Advances in NMR-based structural genomics spectroscopy. Advances in BioNMR<br />

Spectroscopy 2010<br />

Vaiphei, S.T., Tang, Y., Montelione, G.T., Inouye, M. The use of the condensed single<br />

protein production (cSPP) system <strong>for</strong> isotope-labeled outer membrane proteins, OmpA <strong>and</strong><br />

OmpX in E. coli. Molecular <strong>Biotechnology</strong> <strong>2011</strong> 47: 205 – 210.<br />

You, L., Cho, E.J., Leavitt, J., Ma, L-C., Montelione, G.T., Anslyn, E.V., Krug, R.M.,<br />

Ellington, A., Robertus, J.D. Synthesis <strong>and</strong> evaluation of quinoxaline derivatives as<br />

potential NS1A protein inhibitors. Bioorg. Med. Chem. Lett. <strong>2011</strong> 21: 3007 - 3011.<br />

Schauder, C., Ma, L-C., Krug, R.M., Montelione, G.T., Guan, R. Crystal structure of iSH2<br />

domain of human phosphoinositide3-kinase p85β subunit reveals con<strong>for</strong>mational plasticity<br />

in the interhelical turn region. Acta Cryst. F. 2010 F66: 1567 - 1571.<br />

Vorobiev, S.M., Huang, Y.J., Seetharaman, J., Xiao, R., Acton, T.B., Montelione, G.T.,<br />

Tong, L. Structure <strong>and</strong> function of human retinoblastoma binding protein 9 (RBBP9).<br />

Protein Peptide Letts <strong>2011</strong> (e-pub ahead of print)<br />

58


Acton, T.B., Xiao, R., Anderson, S., Aramini, J.M., Buchwald, W., Ciccosanti, C., Conover,<br />

K., Everett, J.K., Hamilton, K., Huang, Y.J., Janjua, H., Kornhaber, G.J., Lau, J., Lee, D.Y.,<br />

Liu, G., Maglaqui, M., Ma, L-C., Mao, L., Patel, D., Rossi, P., Sahdev, S., Sharma, S., Shastry,<br />

R., Swapna, G.V.T., Tang, Y., Tong, S.N., Wang, D., Wang, H., Zhao, L., Montelione, G.T.<br />

Preparation of protein samples <strong>for</strong> NMR structure, function, <strong>and</strong> Small Molecule screening<br />

studies. Meth. Enzymology <strong>2011</strong> 493: 21 - 60.<br />

Mao, L., Inoue, K., Tao, Y., Montelione, G.T., McDermott, A.E., Inouye, M. J. Suppression of<br />

phospholipid biosynthesis by cerulenin in the condensed Single-Protein-Production (cSPP)<br />

system. Biomol. NMR <strong>2011</strong> 49: 131 - 137.<br />

Ramelot, T.A., Smola, M.J., Lee, H-W., Ciccosanti, C., Hamilton, K., Acton, T.B., Xiao, R.,<br />

Everett, J.K., Prestegard, J.H., Montelione, G.T., Kennedy, M.A. Solution NMR structure of<br />

4’-phosphopantetheine - GmACP3 from Geobacter metallireducens: a specialized acyl carrier<br />

protein with aty[ical structural features <strong>and</strong> a putative role in lipopolysaccharide biosynthesis.<br />

Biochemistry <strong>2011</strong> 50: 1442 - 1453.<br />

Yin, C., Aramini, J.M., Ma, L-C., Cort, J.R.; Swapna, G.V.T.; Krug, R.M.; Montelione, G.T.<br />

Backbone <strong>and</strong> Ile-δ1, Leu, Val methyl 1H, 13C <strong>and</strong> 15N NMR chemical shift assignments <strong>for</strong><br />

human interferon-stimulated gene 15 protein. J. Biomol. NMR Assignments <strong>2011</strong> 5: 215 -<br />

219.<br />

Karanicolas, J., Corn, J.E., Chen, I., Joachimiak, L.A., Dym, O., Peck, S.H., Albeck, S., Unger,<br />

T., Hu, W., Liu, G., Delbecq, S., Montelione, G.T., Spiegel, C.P., Liu, D.R., Baker, D. A de<br />

novo protein binding pair by computational design <strong>and</strong> directed evolution. Molecular Cell<br />

<strong>2011</strong> 42: 250 - 260.<br />

Grant, T., Luft, J.R., Wolfley, J.R., Tsuruta, H., Martel, A., Montelione, G.T., Snell, E. Small<br />

angle x-ray scattering as a complementary tool <strong>for</strong> high-throughput structural studies.<br />

Biopolymers <strong>2011</strong> 95: 517 - 530.<br />

Sgourakis, N.G., Lange, O.F., DiMaio, F., André, I., Fitzkee, N.C., Rossi, P., Montelione, G.T.,<br />

Bax, A., Baker, D. Determination of the structures of symmetric protein oligomers from NMR<br />

chemical shifts <strong>and</strong> residual dipolar couplings. J. Am. Chem. Soc. <strong>2011</strong> 133: 6288 - 6298.<br />

Barb, A.W., Cort, J.R., Seetharaman, J., Lew, S., Lee, H.W., Acton, T., Xiao, R., Kennedy,<br />

M.A., Tong, L., Montelione, G.T., Prestegard, J.H. Structures of domains I <strong>and</strong> IV from YbbR<br />

are representative of a widely distributed protein family. Protein Science <strong>2011</strong> 20: 396 - 405.<br />

59


Chu, C., Das, K., Tyminski, J.R., Bauman, J.D., Guan, R., Qiu, W., Montelione, G.T.,<br />

Arnold, E., Shatkin, A.J. Structure of the guanylyltransferase domain of human mRNA<br />

capping enzyme. Proc. Natl. Acad. Sci. U.S.A. <strong>2011</strong> 108: 10104 - 10108.<br />

Mao, B., Guan, R., Montelione, G.T. Improved technologies now routinely provide<br />

protein NMR structures useful <strong>for</strong> molecular replacement. Structure <strong>2011</strong> 19: 757 - 766.<br />

Aramini, J. M., Ma, L.-C., Zhou, L., Schauder, C. M., Hamilton, K., Amer, B. R., Mack, T.<br />

R., Lee, H.-W., Ciccosanti, C.T., Zhao, L., Xiao, R., Krug, R. M., Montelione, G. T.<br />

(<strong>2011</strong>) The dimer interface of the effector domain of non-structural protein 1 from<br />

influenza A virus: an interface with multiple functions. J. Biol. Chem. <strong>2011</strong> 286: 26050 -<br />

26060.<br />

Aramini, J.M., Rossi, P., Fischer, M., Xiao, R., Acton, T.B., Montelione, G.T. Solution<br />

NMR structure of VF0530 from Vibrio fischeri reveals a nucleic-acid binding function.<br />

PROTEINS: Struc. Funct. Bioin<strong>for</strong>matics <strong>2011</strong> 79: 2988 - 2991.<br />

Guan, R., Ma, L-C; Leonard, P.G., Amer, B.R., Sridharan, H., Zhao, C., Krug, R.M.,<br />

Montelione, G.T. Structural basis <strong>for</strong> the sequence-specific recognition of human ISG15<br />

by the NS1 protein of influenza B virus. Proc. Natl. Acad. Sci. U.S.A. <strong>2011</strong> 108: 13468 -<br />

13473.<br />

Eletsky, A., Ruyechan, W.T., Xiao, R., Acton, T.B., Montelione, G.T., Szyperski, T.<br />

Solution NMR structure of MED25(391-543) comprising the activator-interacting domain<br />

(ACID) of human mediator subunit 25. J. Struct. Funct. Genomics <strong>2011</strong> 12: 159 - 166.<br />

Forouhar, F., Saadat, N., Hussain, M., Seetharaman, J., Lee, I., Janjua, H., Xiao, R.,<br />

Shastry, R., Acton, T., Montelione, G.T., Tong, L. A large con<strong>for</strong>mational change in the<br />

putative ATP pyrophosphatase PF0828 induced by ATP binding. Acta Cryst. F <strong>2011</strong><br />

67:1323 - 1327.<br />

Lowery, J., Szul, T., Seetharaman, J., Xiaoying, J., Su, M., Forouhar, F., Xiao, R., Acton,<br />

T.B., Montelione, G.T., Lin, H., Wright, J.W., Lee, E., Holloway, Z.G., R<strong>and</strong>azzo, P.A.,<br />

Tong, L., Sztul, E. A novel C-terminal motif within the Sec7 domain of guanine<br />

nucleotide exchange factors regulates ARF binding <strong>and</strong> activation. J. Biol. Chem. <strong>2011</strong><br />

286: 36898 - 36906.<br />

Yang, Y., Ramelot, T.A., Cort, J.R., Wang, D., Ciccosanti, C., Jiang, M., Acton, T.B.,<br />

Xiao, R., Everett, J.K., Montelione, G.T., Kennedy, M. Solution NMR structure of<br />

Dsy0195 homodimer from Desulfitobacterium hafniense: First structure representative of<br />

the YapB domain family of proteins involved in spore coat assembly. J. Struct. Funct.<br />

Genomics <strong>2011</strong> 12: 175 - 179.<br />

60


Kryshtafovych, A., Bartual, S.G., Bazan, J.F., Berman, H., Casteel, D.E., Christodoulou, E.,<br />

Everett, J.K., Hausmann, J., Heidebrecht, T., Hills, T., Hui, R., Hunt, J.F., Tong, L.,<br />

Jayaraman, S., Joachimiak, A., Kennedy, M., Kim, C., Lingel, A., Michalska, K., Montelione,<br />

G.T., Otero, J.M., Perrakis, A., Pizarro, M.J., van Raaij, M.J., Ramelot, T.A., Rousseau, F.,<br />

Weraimont, A.K., Young, J., Schwede, T. Target highlights in CASP9: experimental target<br />

structures <strong>for</strong> the critical assessment of techniques from protein structure prediction.<br />

PROTEINS: Struc. Funct. Bioin<strong>for</strong>matics <strong>2011</strong> (e-pub ahead of print).<br />

Feldmann, E.A., Ramelot, T.A., Yang, Y., Xiao, R., Acton, T.B., Everett, J.K., Montelione,<br />

G.T., Kennedy, M.A. Solution NMR structure of Asl3597 from Nostoc sp. PCC7120, the first<br />

structure from protein domain family PF12095, adopts a novel fold. PROTEINS: Struc. Funct.<br />

Bioin<strong>for</strong>matics <strong>2011</strong> (e-pub ahead of print).<br />

Rosato, A., Aramini, J.M., Arrowsmith, C., Bagaria, A., Baker, D., Cavalli, A., Doreleijers,<br />

J.F., Eletsky, A., Giachetti, A., Guerry, P., Gutmanas, A., Güntert, P., He, Y., Herrmann, T.,<br />

Huang, Y.J., Jaravine, V., Jonker, H.R.A., Kennedy, M.A., Lange, O.F., Liu, G., Malliavan,<br />

T.E., Mani, R., Mao, B., Montelione, G.T., Nilges, M., Possi, P., van der Schot, G., Schwalbe,<br />

H., Szyperski, T.A., Vendruscolo, M., Vernon, R., Vranken, W.F., de Vries, S., Vuister, G.W.,<br />

Wu, B., Yang, Y., Bonvin, A.M.J.J. Blind testing of routine, fully automated determination of<br />

protein structures from NMR data. Nature Methods <strong>2011</strong> (in press).<br />

Bagaria, A., Jaravine, V., Huang, Y.P., Montelione, G.T., Guntert, P. Protein structure<br />

validation by generalized linear model root-mean-square deviation prediction Protein Science<br />

<strong>2011</strong> (in press).<br />

61


Dr. Vikas N<strong>and</strong>a<br />

CABM Resident Faculty Member<br />

Associate Professor<br />

Department of Biochemistry<br />

UMDNJ-Robert Wood Johnson<br />

Medical School<br />

Dr. Fei Xu<br />

Research Associate<br />

Dr. Agustina<br />

Rodriguez-Granillo<br />

Research Assistant<br />

Dr. James Stapleton<br />

Postdoctoral Fellow<br />

Dr. Isaac John Khan<br />

Postdoctoral Assoc.<br />

Dr. Avanish Parmar<br />

Postdoctoral Assoc.<br />

Sumana Giddu<br />

Research Teaching Spec.<br />

Teresita Silva<br />

Research Teaching Spec.<br />

Daniel Hsieh<br />

Graduate Student<br />

Protein Design <strong>and</strong> Evolution Laboratory<br />

Dr. Vikas N<strong>and</strong>a joined CABM in September of 2005 after studying as an<br />

NIH National Research Service Award postdoctoral fellow in the laboratory<br />

of Dr. William DeGrado at the Department of Biochemistry <strong>and</strong> Biophysics<br />

at the University of Pennsylvania School of Medicine. His research focused<br />

on studying the molecular basis of transmembrane interactions among<br />

integrins involved in regulation of platelet hemostasis. Prior to that, Dr.<br />

N<strong>and</strong>a received his doctorate in Biochemistry from Johns Hopkins<br />

University. His laboratory is supported by federal grants including a recent<br />

highly competitive NIH New Innovator Award.<br />

.<br />

Our group is interested in constructing new proteins <strong>for</strong> applications in biomedical<br />

research, nanotechnology <strong>and</strong> as tools <strong>for</strong> underst<strong>and</strong>ing how proteins fold <strong>and</strong><br />

evolve. Significant progress has been made in the last decade using sophisticated<br />

computer programs to design proteins with novel folds <strong>and</strong> functions. We maintain<br />

<strong>and</strong> develop the software package, protCAD (protein computer aided molecular<br />

design), which has been applied to protein design, structure prediction <strong>and</strong> docking<br />

of protein lig<strong>and</strong> complexes.<br />

Computational Design of an Extracellular Matrix<br />

The extracellular matrix (ECM) is a complex network of collagens, laminins,<br />

fibronectins <strong>and</strong> proteoglycans that provides a surface upon which cells can adhere,<br />

differentiate <strong>and</strong> proliferate. Defects in the ECM are the underlying cause of a wide<br />

spectrum of diseases. The ECM mediates endothelial cell polarity <strong>and</strong> under normal<br />

conditions can suppress pre-oncogenic transitions to a neoplastic state. We are<br />

constructing artificial, de novo collagen-based matrices using a hierarchic<br />

computational approach. These matrices will be physically characterized in the<br />

laboratory <strong>and</strong> used to probe the role of chemical <strong>and</strong> spatial organization in the<br />

ECM on the tumor <strong>for</strong>ming potential of adhered cells. Successfully designed<br />

matrices will be applied to engineering safer artificial tissues.<br />

Rational Design of Enhanced Peptide Therapeutics<br />

Peptides are an emergent <strong>and</strong> important class of therapeutics with over <strong>for</strong>ty<br />

compounds on the market <strong>and</strong> nearly 700 more in clinical or pre-clinical trials.<br />

During the development of peptide drugs, D-enantiomers of amino acids are<br />

frequently incorporated to improve pharmacokinetic <strong>and</strong> pharmacodynamics<br />

62


properties by lowering susceptibility to proteolysis. Typically, such modifications are<br />

introduced in lead compounds by trial-<strong>and</strong>-error or combinatorial approaches.<br />

Our laboratory is developing components in protCAD to simulate the impact of nonnatural<br />

amino acids on structure <strong>and</strong> stability. Using fundamental principles of protein<br />

design, we will pursue the computational, structure-based development of peptides<br />

with variable chirality, broadly extending our capacity to create safe <strong>and</strong> potent<br />

therapeutics.<br />

Biochemical Basis of Food Allergies<br />

A crucial <strong>and</strong> unanswered question in the field of food allergy research is why certain<br />

proteins elicit an IgE mediated immune response, while others are tolerated. One<br />

compelling model is that non-allergens are more digestible, resulting in sufficient<br />

protein degradation in the stomach <strong>and</strong> intestine to render the remaining fragments<br />

immunologically inert. In this project, we develop a highly defined system <strong>for</strong><br />

exploring the relationship between digestibility <strong>and</strong> allergenicity using engineered<br />

variants of protein allergens from peanut <strong>and</strong> shrimp.<br />

Natural collagens in the connective<br />

tissue or the basement membrane<br />

assemble in a hierarchic fashion.<br />

Our peptides proceed from<br />

monomer to trimer to fiber, <strong>and</strong><br />

finally to a hydrogel. Hydrogels are<br />

versatile materials with applications<br />

in drug delivery, tissue scaffolds<br />

<strong>and</strong> enzyme immobilization. Insight<br />

gained from this study will improve<br />

molecular design of biomimetic<br />

materials. TOP PICTURE: Phase<br />

diagram of peptides CPB <strong>and</strong> CPC<br />

combinations showing the diversity<br />

of self-assembling structures<br />

<strong>for</strong>med. BOTTOM PICTURE:<br />

Electron Microscopy images of<br />

negatively stained CPB:CPC<br />

samples at various mixing ratios.<br />

Mihir Joshi<br />

Research Assistant<br />

Kenneth<br />

McGuinness<br />

Research Asstistant<br />

Yol<strong>and</strong>a Hom<br />

Technician<br />

63


Publications:<br />

Rodriguez-Granillo, A., Annavarapu, S., Zhang, L., Koder, R.L., N<strong>and</strong>a, V.<br />

Computational Design of Thermostabilizing D-Amino Acid Substitutions. J. Am. Chem.<br />

Soc. (in press)<br />

Xu, F., Zahid, S., Silva, T., N<strong>and</strong>a, V. Computational Design of a Collagen A:B:C-type<br />

Heterotrimer. J. Am. Chem. Soc. <strong>2011</strong> 183:15260-63.<br />

Hsieh, D., Davis, A., N<strong>and</strong>a, V. A Knowledge Based Potential Highlights Differences in<br />

Membrane a-helical <strong>and</strong> b-barrel Protein Insertion <strong>and</strong> Folding. Prot. Sci. (in press)<br />

Woronowicz, K., Sha, D., Frese, R.N., Sturgis, J.N., N<strong>and</strong>a, V., Niederman, R.A. The<br />

effects of protein crowding in bacterial photosynthetic membranes on the flow of quinone<br />

redox species between the photochemical reaction center <strong>and</strong> the ubiquinol-cytochrome c2<br />

oxidoreductase. Metallomics <strong>2011</strong> 3(8):765-74.<br />

N<strong>and</strong>a, V., Zahid, S., Xu, F., Levine, D. Computational Design of Intermolecular Stability<br />

<strong>and</strong> Specificity in Protein Self-Assembly. Methods Enzym. <strong>2011</strong> 487:575-93.<br />

Braun, P., Goldberg, E., Negron, C., von Jan, M., Xu, F., N<strong>and</strong>a, V., Koder, R.L., Noy, D.<br />

Design principles <strong>for</strong> chlorophyll-binding sites in helical proteins. Proteins. <strong>2011</strong><br />

79(2):463-76.<br />

Stapleton, J.A., Rodriguez-Granillo, A., N<strong>and</strong>a, V. Chapter 3.2-Artificial Enzymes (in<br />

Nanobiotechnology H<strong>and</strong>book). Taylor & Frances Group (in press)<br />

Rodriguez-Granillo, A., N<strong>and</strong>a, V. Designing new enzymes with molecular simulations.<br />

<strong>Biotechnology</strong> International <strong>2011</strong><br />

N<strong>and</strong>a, V., Koder ,R.L. Designing artificial enzymes by intuition <strong>and</strong> computation.<br />

Nature Chemistry 2010 2(1):15-24.<br />

Xu, F., Zhang, L., Koder, R.L., N<strong>and</strong>a, V. De novo self-assembling collagen heterotrimers<br />

using explicit positive <strong>and</strong> negative design. Biochemistry. 2010 49(11):2307-16.<br />

Grzyb, J., Xu, F., Weiner, L., Reijerse, E.J., Lubizt, W., N<strong>and</strong>a, V., Noy, D. De novo<br />

design of a non-natural fold <strong>for</strong> an iron-sulfur protein: alpha-helical coiled coil with fouriron<br />

four-sulfur cluster binding site in its central core. Biochim BIophys Acta 2010<br />

1797(3):406-13.<br />

64


Dr. Ann M. Stock<br />

Associate Director of CABM<br />

Investigator<br />

Howard Hughes Medical Institute<br />

Professor<br />

Department of Biochemistry<br />

UMDNJ-Robert Wood Johnson<br />

Medical School<br />

Dr. Rong Gao<br />

Research Associate<br />

Dr. Christopher<br />

Barbieri<br />

Postdoctoral Associate<br />

Dr. Paul Leonard<br />

Postdoctoral Associate<br />

Ian Bezar<br />

Graduate Fellow<br />

Hua Han<br />

Graduate Student<br />

Ti Wu<br />

Research Teaching Spec.<br />

Jizong Gao<br />

Research Specialist<br />

Protein Crystallography Laboratory<br />

Dr. Ann Stock per<strong>for</strong>med graduate work on the biochemistry of signal transduction<br />

proteins with Professor Daniel E. Koshl<strong>and</strong>, Jr. at the University of Cali<strong>for</strong>nia at<br />

Berkeley. From 1987 to 1991 she pursued structural analysis of these proteins<br />

while a postdoctoral fellow with Professor Clarence Schutt at Princeton University<br />

<strong>and</strong> Professor Gregory Petsko at the Structural Biology Laboratory in the<br />

Rosenstiel <strong>Center</strong> at Br<strong>and</strong>eis University. Stock has received National Science<br />

Foundation Predoctoral <strong>and</strong> Damon Runyon-Walter Winchell Postdoctoral<br />

Fellowships, a Lucille P. Markey Scholar Award in Biomedical Science, an NSF<br />

Young Investigator Award, a Sinsheimer Scholar Award <strong>and</strong> an NIH MERIT Award<br />

in 2002. She has received the Foundation of UMDNJ Excellence in Teaching<br />

Award, the Molecular Biosciences Graduate Association Educator of the Year<br />

Award <strong>and</strong> the designation of Master Educator at UMDNJ. In 2004 Stock was<br />

selected as an Outst<strong>and</strong>ing Scientist by the NJ Association <strong>for</strong> Biomedical Research.<br />

She was elected Fellow of the American Association <strong>for</strong> the Advancement of Science<br />

in 2006 <strong>and</strong> fellow of the American Academy <strong>for</strong> Microbiology in 2007. Stock was<br />

an investigator of the Howard Hughes Medical Institute from 1994-<strong>2011</strong>. She<br />

served as Chair of the Board of Scientific Counselors <strong>for</strong> the NIH-National Institute<br />

of Dental <strong>and</strong> Craniofacial Research in 2008-2009 <strong>and</strong> currently serves as an<br />

Editor of the Journal of Bacteriology. She served on the Council of the American<br />

Society <strong>for</strong> Biochemistry <strong>and</strong> Molecular Biology from 2008-<strong>2011</strong> <strong>and</strong> currently<br />

serves as a member of the ASBMB Education & Professional Development <strong>and</strong><br />

Finance Committees. Stock serves as an elected member of the RWJMS Faculty<br />

Council <strong>and</strong> the UMDNJ Faculty Senate.<br />

The research of the Stock laboratory focuses on structure/function studies of signal<br />

transduction proteins. Ef<strong>for</strong>t is concentrated on response regulator proteins that are<br />

the core of two-component systems, the most prevalent signal transduction pathways<br />

in bacteria. These systems are important <strong>for</strong> virulence in pathogenic organisms <strong>and</strong><br />

are targets <strong>for</strong> development of new antibiotics.<br />

The Stock laboratory has continued to focus ef<strong>for</strong>ts on characterizing representative<br />

members of the OmpR/PhoB transcription factor subfamily of response regulator<br />

proteins, but now from a systems biology perspective. Studies have shifted from in<br />

vitro analyses to in vivo analyses with the goal of being able to describe the<br />

parameters of protein levels, phosphorylation states, <strong>and</strong> gene expression activity<br />

during induction of the phosphate assimilation pathway, a model two-component<br />

system in E. coli. Tools have been developed <strong>and</strong> data are being accumulated that<br />

will hopefully allow the mathematical modeling of the mechanism of regulation in a<br />

representative two-component system.<br />

An additional research focus is characterization of two-component systems of<br />

Staphylococcus aureus, a re-emerging pathogen that is currently associated with<br />

more deaths annually in the United States than HIV1 (AIDS). New classes of drugs<br />

are needed to combat the economic <strong>and</strong> health burden of drug-resistant S. aureus<br />

strains (i.e. methicillin-resistant, MRSA). Several response regulators in<br />

Staphylococcus aureus play important roles in virulence <strong>and</strong> are potential drug<br />

targets. The Stock laboratory has focused ef<strong>for</strong>ts on two S. aureus transcription<br />

factors, AgrA <strong>and</strong> VraR. In 2008, the Stock laboratory reported the structure of the<br />

65


DNA-binding domain of AgrA, establishing a novel fold <strong>for</strong> the LytTR domain that<br />

regulates virulence factor expression in many pathogenic bacteria. Recent studies<br />

have focused on purification <strong>and</strong> characterization of full-length AgrA with an<br />

emphasis on underst<strong>and</strong>ing the role of the extreme C terminus that is altered by a<br />

phase-variation mechanism, inactivating AgrA at late stages of infection.<br />

WATERGATE LOGSY NMR experiments were used to identify drug-like fragments<br />

that bind to AgrA. All six lig<strong>and</strong>s identified bind to the same site, overlapping the<br />

surface required <strong>for</strong> binding. Three of the fragments have been shown to inhibit DNA<br />

binding <strong>and</strong> are being pursued <strong>for</strong> inhibitor development. The Stock laboratory has<br />

determined the structures of both inactive <strong>and</strong> activated full-length VraR, a<br />

vancomycin-resistance-associated regulator that plays a central role in maintaining<br />

the integrity of the cell wall peptidoglycan <strong>and</strong> in coordinating responses to cell wall<br />

damage. These structures represent the first inactive <strong>and</strong> active pair of full-length<br />

response regulator transcription factors. The structures establish the mechanism of<br />

activation of VraR via dimerization <strong>and</strong> reveal an unusual deep binding pocket with<br />

great potential <strong>for</strong> inhibitor development. Virtual docking analyses using the Zinc<br />

database of available compounds have identified many compounds predicted to bind<br />

to this pocket. In collaboration with Ed LaVoie (School of Pharmacy, Rutgers<br />

University) <strong>and</strong> John Kerrigan (CINJ), these compounds have been ranked <strong>and</strong> several<br />

have been selected as a starting point <strong>for</strong> inhibitor development.<br />

Dimer interface of active VraR<br />

receiver domain. The protruding<br />

Methionine 13 from one monomer<br />

fits into a deep binding cleft of the<br />

other monomer, a site that is<br />

being targeted <strong>for</strong> development of<br />

inhibitors as leads <strong>for</strong><br />

development of novel antibiotics.<br />

66


Publications:<br />

Barbieri, C.M., Mack, T.R., Robinson, V.L., Miller, M.T. <strong>and</strong> Stock, A.M. Regulation of<br />

response regulator autophosphorylation through interdomain contacts. J. Biol. Chem. 2010<br />

285: 32325-32335.<br />

Dixit, S.S., Jadot, M., Sohar, I., Sleat, D.E., Stock, A.M. <strong>and</strong> Lobel, P. Biochemical<br />

alterations resulting from the loss of either NPC1 or NPC2 suggest that these proteins<br />

underlie Niemann-Pick C disease function in a common lysosomal pathway. PLOS One<br />

<strong>2011</strong> in Press.<br />

67


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

Lectures <strong>and</strong> Seminars 69<br />

CABM Lecture Series 70<br />

<strong>Annual</strong> Retreat 71<br />

25 th <strong>Annual</strong> Symposium 73<br />

Undergraduate Students 76<br />

Administrative <strong>and</strong> Support Staff 77<br />

Patents 78<br />

Scientific Advisory Board 81<br />

Current Members 82<br />

Past Members 84<br />

Fiscal In<strong>for</strong>mation 86<br />

Grants <strong>and</strong> Contracts 87<br />

68


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

69


Lecture Series 2010-<strong>2011</strong><br />

Lectures will be held in CABM Seminar Room 010 unless otherwise noted<br />

October 19, 2010 – Waksman Auditorium<br />

(Pre-Registration Required)<br />

24th <strong>Annual</strong> CABM Symposium<br />

“Cancer – Progress & Prospects”<br />

Wednesday, November 17, 12:00<br />

Robert Lamb, HHMI, Northwestern University<br />

“Influenza Virus Budding: Who Needs an ESCRT When There Is M2 Protein”<br />

Wednesday, December 1, 12:00<br />

Alanna Schepartz, Yale University<br />

“Exploring Biology with Molecules that Nature Chose not to Synthesize”<br />

Wednesday, January 19, 2010,12:00<br />

Evgeny Nudler, New York University, School of Medicine<br />

“Trafficking <strong>and</strong> Cooperation in Gene Regulation <strong>and</strong> Genome Instability”<br />

Wednesday, February 2, 12:00<br />

Smita Patel, UMDNJ-Robert Wood Johnson Medical School<br />

“Mechanistic studies of a ring-shaped helicase that coordinates DNA replication”<br />

Wednesday, March 30, 12:00<br />

Charles Rice, The Rockefeller University<br />

“Hepatitis C virus: New insights into replication <strong>and</strong> control”<br />

Wednesday, April 13, 12:00<br />

Nahum Sonenberg, McGill University<br />

“Translational Control of Cancer”<br />

Supported by Sanofi Aventis<br />

70


CABM RETREAT, June 30, <strong>2011</strong><br />

Cook Campus <strong>Center</strong>, New Brunswick, NJ<br />

Program<br />

8:15 AM Registration, Poster Set-up <strong>and</strong> Continental Breakfast<br />

8:45 AM Opening Remarks – Aaron J. Shatkin<br />

9:00 AM Session I – Gene Expression –From Structure to Behavior <strong>and</strong> Steps In Between<br />

Chair: Janet Huang - Structural Bioin<strong>for</strong>matics <strong>and</strong> Proteomics Lab (Guy Montelione)<br />

Shuchismita Dutta – Protein Data Bank Lab (Helen Berman)<br />

“Molecular Anatomy Project: Promoting a Structural View of Biology”<br />

Lili Mao – Bacterial Physiology <strong>and</strong> Protein Engineering Lab (Masayori Inouye)<br />

“The Single Protein Production (SPP) system in E. coli”<br />

Hua Han – Protein Crystallography Lab (Ann Stock)<br />

“The Kinetics of phospho regulation by PhoBR two-component system in E. coli “<br />

Evrim Yildirim – Molecular Chronobiology Lab (Isaac Edery)<br />

“Phosphorylation of S826/828 on PER sets the pace of circadian clock”<br />

Weihua Qiu – Molecular Virology Lab (Aaron Shatkin)<br />

“Structure of the Guanylyltransferase Domain of Human mRNA Capping Enzyme”<br />

Stephen Anderson – Protein Engineering Lab (Stephen Anderson)<br />

“Production of human transcription factor immunogens”<br />

10:30 AM Coffee Break <strong>and</strong> Posters<br />

11:00 AM Session II – Model Systems to Study Growth <strong>and</strong> Development<br />

Chair: Su Xu – Protein Targeting Lab (Peter Lobel)<br />

Kenneth McGuinness – Protein Design <strong>and</strong> Evolution Lab (Vikas N<strong>and</strong>a)<br />

“Designing Collagen Self-Assembly using Hydrophobic Interactions”<br />

Céline Granier – Viral Pathogenesis Lab (Arnold Rabson)<br />

“PDCD2, a Novel Nuclear Body-associated Protein in Embryonic Stem Cells <strong>and</strong> Mouse<br />

Development”<br />

Bo Li – Developmental Neurogenetics Lab (Jim Millonig)<br />

“Vacuolated lens (vl): a multigenic mouse mutant model of Neural Tube Defects (NTDs)”<br />

71


12:30 PM Lunch <strong>and</strong> Break<br />

Kamana Misra – Molecular Neurodevelopment Lab (Meng Xiang)<br />

“To be or not to be" A neuron's perspective<br />

2:15 PM Posters <strong>and</strong> Coffee<br />

4:30 PM Close<br />

Session III – Underst<strong>and</strong>ing Disease at the Molecular Level as a Basis <strong>for</strong> Developing<br />

Novel Therapies<br />

Chair: Kamana Misra – Molecular Neurodevelopment Lab (Meng Xiang)<br />

Mary Fitzgerald Ho – Biomolecular Crystallography Lab (Eddy Arnold)<br />

“GE23077: A novel bacterial RNA polymerase inhibitor”<br />

Joseph Marcotrigiano – Structural Virology Lab (Joe Marcotrigiano)<br />

“The Structure of a Precursor from the Alphavirus Replication Complex sheds light on<br />

pathogenesis <strong>and</strong> mechanisms of polyprotein processing”<br />

Rongjin Guan – Structural Bioin<strong>for</strong>matics <strong>and</strong> Proteomics Lab (Guy Montelione)<br />

“Structural Basis <strong>for</strong> the Sequence-Specific Recognition of Human ISG15 by the NS1 Protein<br />

of Influenza B Virus”<br />

Yu Meng – Protein Targeting Lab (Peter Lobel)<br />

“Intravenous enzyme replacement therapy <strong>for</strong> late-infantile neuronal ceroid lipofuscinosis<br />

(LINCL)”<br />

Fang Liu – Growth <strong>and</strong> Differential Lab (Fang Liu)<br />

“Pin1 promotes TGF-beta-induced migration <strong>and</strong> invasion”<br />

Na Yang – Tumor Virology Lab (Céline Gélinas)<br />

“Underst<strong>and</strong>ing <strong>and</strong> targeting Bfl-1 turnover to overcome chemoresistance”<br />

72


25 th Anniversary CABM Symposium<br />

Friday, October 21, <strong>2011</strong><br />

Program<br />

8:15 am Registration & Continental Breakfast<br />

RWJMS Great Hall<br />

9:00 am Welcoming Remarks<br />

Aaron J. Shatkin, PhD<br />

Professor <strong>and</strong> Director, CABM<br />

Richard L. McCormick, PhD<br />

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

Session I: Chairperson: Joanna Chiu, PhD<br />

Assistant Professor, UC Davis<br />

9:10 am Phillip Sharp, PhD<br />

Institute Professor, MIT<br />

“The Cell Biology of Small Noncoding RNAs”<br />

9:50 am Ken Valenzano, PhD<br />

Vice President, Amicus Therapeutics<br />

“Pharmacological Chaperones Offer a Two-pronged Approach to Treat Lysosomal Storage<br />

Disorders: Fabry Disease as a Case Study”<br />

10:10 am Zhenyu Yue, PhD<br />

Associate Professor, Mt. Sinai Medical School<br />

“From LRRK2 Kinase to the Hope of Underst<strong>and</strong>ing <strong>and</strong> Treating Parkinson’s Disease”<br />

10:30 am Coffee Break<br />

Session II: Chairperson: Wei-xing Zong, PhD<br />

Assistant Professor, SUNY Stony Brook<br />

11:00 am Robert Roeder, PhD<br />

Arnold <strong>and</strong> Mabel Beckman Professor, Rockefeller University<br />

“Transcriptional Regulatory Mechanisms in Animal Cells”<br />

12:00 pm Scott Banta, PhD<br />

Associate Professor, Columbia University<br />

“Development of the Beta Roll Motif as a Novel Biomolecular Recognition Scaffold”<br />

74


12:20 pm Lunch Break<br />

RWJMS Great Hall<br />

1:00 pm Poster Session<br />

RWJMS Great Hall<br />

Session III: Chairperson: Jun Zhu, PhD<br />

Director, DNA Sequencing <strong>and</strong> Computational Biology Core, NIH-NHLBI<br />

2:00 pm Wayne Hendrickson, PhD<br />

University Professor, Columbia University; Investigator, HHMI<br />

“Structural Analysis of SLAC1-family Anion Channel Activity”<br />

2:40 pm Snezana Djordjevic, PhD<br />

Senior Lecture, University College London, UK<br />

“Small Molecule Inhibitors of the VEFG-Neuropilin Interaction”<br />

3:00 pm Hunter Moseley, PhD<br />

Assistant Professor, University of Louisville<br />

“Metabolic Modeling of Converging Metabolic Pathways: Analysis of Non-Steady State<br />

Stable Isotope-Resolved Metabolomics Data of UDP-GlcNAc <strong>and</strong> UDP-GalNAc”<br />

3:20 pm Coffee Break<br />

Session IV: Chairperson: Minjung Kim, PhD<br />

Assistant Professor, Moffitt Cancer <strong>Center</strong>, Tampa, FL<br />

3:50 pm Joseph Marcotrigiano, PhD<br />

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

“The Innate Immune Response to Viral RNA”<br />

4:20 pm Bruce Alberts, PhD<br />

Editor, Science; Professor Emeritus, UCSF<br />

“Stimulating Innovation in Scientific Research”<br />

5:00 pm Closing<br />

5:10 pm Reception<br />

RWJMS Great Hall<br />

75


Edery Lab<br />

Joseph Albistur<br />

George Ghanim (SURE)<br />

Yee Chen Low<br />

Neal Mehta<br />

Jash Vakil (SURE)<br />

Gelinas Lab<br />

Nikita Ekhelikar (SURE)<br />

Inouye Lab<br />

Yuliya Afinogenova<br />

Shantanu Baghel<br />

Neel Belani<br />

Sehrish Asmal<br />

Lobel Lab<br />

Mohamed Albana (SURE)<br />

Duo Hang<br />

Sharlina Keshava<br />

Aakash Panchal<br />

Dhruv Patel<br />

Dana Betts<br />

Tapan Patel<br />

Samantha Schlachter<br />

Tian Sun<br />

Central Lab Services<br />

Monika Kapedia<br />

Brendan Striano<br />

Millonig Lab<br />

Aziz Karakhanyan<br />

Samantha Kelly<br />

Omar Khan<br />

Keiry Rodriguez<br />

Sammy Taha (SURE)<br />

N<strong>and</strong>a Lab<br />

Alex<strong>and</strong>er Davis (SURE)<br />

UNDERGRADUATE STUDENTS<br />

Shatkin Lab<br />

Frank Zong (SURE)<br />

Stock Lab<br />

Christopher Burns<br />

Jose Valverde (SURE)<br />

Xiang Lab<br />

Benjamin Hanft (SURE)<br />

Montelione Lab<br />

Brendan Amer<br />

Hoi Wun Au<br />

Ashley Aya<br />

Alex<strong>and</strong>er Bendik<br />

Tina Bijlani<br />

Nicole Brunck<br />

Katherine Carrino-Bednarski<br />

Ya-Min Chi<br />

Rokhsareh Eyvazkhany<br />

Dhaval G<strong>and</strong>hi<br />

Emily Grasso<br />

Sultan Khawam<br />

Eitan Kohan<br />

Wylie Lopez<br />

Joseph Miller<br />

Ari Sapin<br />

Jaskamel Singh<br />

Chelsea Stahl<br />

Amy Suhotliv (SURE)<br />

Aparna Tatineni (SURE)<br />

Teddy Wohlbold<br />

Liu Lab<br />

Kavelbhai Patel<br />

Administration<br />

Ashley Redmond<br />

Daniel Yoon<br />

76


Administrative Assistants<br />

Darlene Bondoc<br />

Ellen Goodman<br />

Jane Kornhaber<br />

Janice Nappe<br />

Barbara Shaver<br />

Elaine Simpson<br />

Jane Kornhaber<br />

Business Office<br />

Lynnette Butler<br />

John Drudy<br />

Madeline Frances<br />

Sharon Pulz<br />

Kristin Rossi<br />

Diane Sutterlin<br />

ADMINISTRATIVE AND SUPPORT STAFF<br />

Central In<strong>for</strong>mation Technology<br />

Shelley Waltz<br />

Keith Williams<br />

Central Lab Services<br />

Ramon Dugenio<br />

Alba LaFi<strong>and</strong>ra<br />

Sally Marshall<br />

Harren Mercado<br />

Minoti Sinha<br />

Angela Sowa<br />

77


PATENTS<br />

P. Lobel, et al. “Methods of treating a deficiency of functional tripeptidyl peptidase I (CLN2) protein”.<br />

United States Patent 8,029,781<br />

P. Lobel, et al. “Human lysosomal protein <strong>and</strong> methods of its use”. United States Patent 7,745,166<br />

J.F. Hunt, W.M. Price, T.B. Acton, <strong>and</strong> G.T. Montelione. “Methods <strong>for</strong> altering polypeptide expression <strong>and</strong><br />

solubility”. PCT WO 2010/151593 A1 Filed Feb 9, <strong>2011</strong><br />

T. Acton, S. Anderson, Y. Huang, <strong>and</strong> G.T. Montelione. “System <strong>for</strong> high-level production of proteins <strong>and</strong><br />

protein domains”. PCT Filed Nov <strong>2011</strong><br />

Anderson, S., Lazarus, R.A., Hurle, M., Anderson, S. <strong>and</strong> Powers, D.B. "Enzymes <strong>for</strong> the production of 2-keto-<br />

L-gulonic acid". U. S. Patent No. 5,376,544<br />

Anderson, S. <strong>and</strong> Ryan, R. "Inhibitors of urokinase plasminogen activator". U. S. Patent No. 5,550,213<br />

Lazarus, R.A., Hurle, M., Anderson, S. <strong>and</strong> Powers, D.B. "Enzymes <strong>for</strong> the production of 2-keto-L-gulonic acid".<br />

U. S. Patent No. 5,583,025<br />

Anderson, S. "Methods <strong>for</strong> the prevention or treatment of vascular hemorrhaging <strong>and</strong> Alzheimer's disease". U. S.<br />

Patent No. 5,589,154<br />

Powers, D.B. <strong>and</strong> Anderson, S. "Mutants of 2,5-diketo-D-gluconic acid (2,5-DKG) reductase A". U. S. Patent No.<br />

5,795,761<br />

Lazarus, R.A., Hurle, M., Anderson, S. <strong>and</strong> Powers, D.B. "Enzymes <strong>for</strong> the production of 2-keto-L-gulonic acid".<br />

U. S. Patent No. 5,912,161<br />

Anderson, S. “Methods <strong>for</strong> identifying useful t-PA mutant derivatives <strong>for</strong> treatment of vascular hemorrhaging”.<br />

U. S. Patent No. 6,136,548<br />

Anderson, S. <strong>and</strong> Banta, S. “Design <strong>and</strong> production of mutant 2,5-diketo-D-gluconic acid reductase enzymes with<br />

altered cofactor dependency”. U. S. Patent No. 6,423,518<br />

Anderson, S. "Methods <strong>for</strong> the prevention or treatment of Alzheimer's disease". U. S. Patent No. 6,471,960<br />

Anderson, S. “Methods <strong>for</strong> identifying useful t-PA mutant derivatives <strong>for</strong> treatment of vascular hemorrhaging” U.<br />

S. Patent No. 6,136,548<br />

Anderson, S. <strong>and</strong> Banta, S. “Design <strong>and</strong> production of mutant 2,5-diketo-D-gluconic acid reductase enzymes with<br />

altered cofactor dependency”. U. S. Patent No. 6,423,518<br />

78


Lazarus, R.A., Hurle, M., Anderson, S. <strong>and</strong> Powers, D.B. "Enzymes <strong>for</strong> the production of 2-keto-L-gulonic acid".<br />

U. S. Patent No. 5,912,161<br />

Arnold, E., Kenyon, G.L., Stauber, M., Maurer, K., Eargle, D., Muscate, A., Leavitt, A., Roe, D.C., Ewing, T.J.A.,<br />

Skillman, A.G., Jr., Kuntz, I.D. <strong>and</strong> Young, M. “Naphthols useful in antiviral methods”. U.S. Patent No.<br />

6,140,368<br />

Arnold, E. <strong>and</strong> Ferst<strong>and</strong>ig Arnold, G. "Chimeric Rhinoviruses". U.S. Patent No. 5,714,374<br />

Krupey, J., Smith, A., Arnold, E. <strong>and</strong> Donnelly, R. "Method of Adsorbing Viruses from Fluid Compositions".<br />

U.S. Patent No. 5,658,779<br />

Arnold, E. <strong>and</strong> Ferst<strong>and</strong>ig Arnold, G. "Chimeric Rhinoviruses". U.S. Patent No. 5,541,100<br />

Edery, I., Altman, M. <strong>and</strong> Sonenberg, N. “Isolation of full-length cDNA clones <strong>and</strong> capped mRNA”. U.S. Patent<br />

No. 5219989<br />

Lobel, P. <strong>and</strong> Sleat, D. "Human lysosomal protein <strong>and</strong> methods of its use". US Patent No. 6,302,685 B1<br />

Lobel, P. <strong>and</strong> Sleat, D., “Human lysosomal protein <strong>and</strong> methods of its use”. US Patent No. 6,638,712. Submitted<br />

10/28/2003<br />

Millonig, J.H., Brzustowicz, L,M. <strong>and</strong> Gharani, N. “Genes as diagnostic tools <strong>for</strong> autism”. Provisional<br />

application submitted 7/1/03<br />

Montelione, G.T., Anderson, S. <strong>and</strong> Huang, Y. “Linking gene sequence to gene function by 3D protein structure<br />

determination”. EU Patent 99935746.0-2402. Issued in United Kingdom<br />

Stein, S. <strong>and</strong> Montelione, G.T. “Site specific 13C-enriched reagents <strong>for</strong> magnetic resonance imaging”. U.S.<br />

Patent No. 6210655<br />

Montelione, G. T. <strong>and</strong> Krug, R. “Process <strong>for</strong> designing inhibitors of influenza virus non-structural protein 1”.<br />

Filed November 13, 2003. PCT/US03/36292. Patent Pending.<br />

Montelione, G.T., Krug, R., Yin, C. <strong>and</strong> Ma, L. “Novel compositions <strong>and</strong> vaccines against influenza A <strong>and</strong><br />

influenza B infections”. Filed November 17, 2006. Patent Pending.<br />

Montelione, G. T, Arnold, E., Das, K., Ma, L., Xiao, R., Twu, K.Y., Rei-Lin, K. <strong>and</strong> Krug, R.M. “Influenza A<br />

virus vaccines <strong>and</strong> inhibitors”. Filed January 23, 2007. Patent Pending.<br />

Montelione, G.T., Das, K. <strong>and</strong> Arnold, E. “Ribosomal RNA methyltransferases RlmAI <strong>and</strong> RlmAII: Drug target<br />

validation <strong>and</strong> process <strong>for</strong> developing an inhibitor assay”. Submitted June 26, 2004; Priority date: June 27, 2003.<br />

PTC/US2004/020244. Patent Pending.<br />

79


Montelione, G.T., Ma, L. <strong>and</strong> Krug, R.M. “An assay <strong>for</strong> designing small molecule inhibitors of the life cycle of<br />

influenza (Flu) virus targeted to the viral non-structural protein 1”. Provisional Patent Application submitted<br />

March 2007.<br />

Suzuki, M., Schneider, W., Tang, Y., Roth, M., Inouye, M. <strong>and</strong> Montelione, G.T. “Processes using E. coli single<br />

protein production system (SPP) <strong>for</strong> deuterium/methyl enrichment of proteins, detergent screening of membrane<br />

proteins using NMR, <strong>and</strong> peptide bond labeling <strong>for</strong> high throughput NMR assignments”. Provisional Patent<br />

Application submitted March 2007.<br />

Xiao, R.; Nie, Y.; Xu, Y.; Montelione, G.T. “Three NADPH-dependant stereospecific reductase from C<strong>and</strong>ida<br />

parapsilosis <strong>and</strong> their application to chiral alcohol production.” Provisional Patent Application submitted June<br />

2009.<br />

Shatkin, A.J., Pillutla, R., Reinberg, D., Maldonado, E. <strong>and</strong> Yue, Z. “mRNA Capping enzymes <strong>and</strong> uses thereof.”<br />

U.S. Patent No. 631292631.<br />

80


Scientific Advisory Board<br />

81


Outside Academic Members<br />

Dr. Wayne A. Hendrickson<br />

Investigator, Howard Hughes Medical Institute<br />

Professor<br />

Department of Biochemistry & Molecular Biophysics<br />

Columbia University<br />

Dr. Robert G. Roeder<br />

Arnold <strong>and</strong> Mabel Beckman Professor<br />

Head, Laboratory of Biochemistry <strong>and</strong> Molecular Biology<br />

The Rockefeller University<br />

Dr. Thomas Eugene Shenk<br />

Elkins Professor<br />

Department of Molecular Biology<br />

Princeton University<br />

Industry Representatives<br />

Dr. Nader Fotouhi<br />

Vice President, Discovery Chemistry<br />

Roche<br />

Michael Dean Miller, Ph.D.<br />

Site Lead, Infectious Disease – HIV<br />

Merck Research Laboratories<br />

Garry Neil, M.D. (Chair)<br />

Corporate Vice President<br />

Corporate Office of Science & Technology<br />

Johnson & Johnson<br />

Dr. William S. Somers<br />

Assistant Vice President<br />

Global Biotherapeutic Technologies<br />

Pfizer<br />

Dr. Michael J. Tocci<br />

Director, Bridgewater Functional Genomics<br />

Sanofi<br />

CABM Scientific Advisory Board<br />

82


UMDNJ MEMBERS<br />

Robert S. DiPaola, M.D.<br />

Director, The Cancer Institute of New Jersey<br />

Associate Dean <strong>for</strong> Oncology Programs<br />

Professor of Medicine<br />

UMDNJ-Robert Wood Johnson Medical School<br />

Dr. Leroy F. Liu<br />

Professor <strong>and</strong> Chairman<br />

Department of Pharmacology<br />

Arnold B. Rabson, M.D.<br />

Professor <strong>and</strong> Director, Child Health Institute of New Jersey<br />

Interim Senior Associate Dean <strong>for</strong> Research<br />

UMDNJ-Robert Wood Johnson Medical School<br />

RUTGERS MEMBERS<br />

Dr. Kenneth J. Breslauer<br />

Linus C. Pauling Professor<br />

Dean of Biological Sciences<br />

Vice President of Health Science Partnerships<br />

Dr. Allan H. Conney<br />

William M. And Myrle W. Garber<br />

Professor of Cancer <strong>and</strong> Leukemia Research<br />

Dr. Joachim W. Messing<br />

University of Professor <strong>and</strong> Director<br />

Waksman Institute<br />

Martin L. Yarmush, M.D.<br />

Paul <strong>and</strong> Mary Monroe Professor of Science <strong>and</strong> Engineering<br />

Director, <strong>Center</strong> <strong>for</strong> Innovative Ventures of Emerging Technologies, Rutgers<br />

Director, <strong>Center</strong> <strong>for</strong> Engineering in Medicine, Massachusetts General Hospital<br />

83


Former CABM Scientific Advisory Board Members<br />

Alberts, Bruce, Prof., UCSF (Pres. National Academy of Sciences), '86<br />

Babiss, Lee, Vice President Pre-Clinical Res. & Dev. Hoffmann-La Roche Inc.'99-'06<br />

* Baltimore, David, Dir., Whitehead Inst., (Pres. Cal. Tech.), '86-'90<br />

Bayne, Marvin L., VP Discovery Technologies Merck, ’07-‘10<br />

Bolen, Joseph, Vice President, Hoechst Marin Roussel, '98<br />

Black, Ira, M.D., Prof. & Chairman, Dept. of Neurosci. & Cell Biol., RWJ/UMDNJ '91-'06, (deceased)<br />

Blumenthal, David, M.D., Exec. Dir., Ctr. <strong>for</strong> Health Policy & Mgmt. @ Harvard, '86-'88<br />

Burke, James, Chairman of the Board, J & J, '86-'89<br />

Denhardt, David, Prof. & Chairman, Rutgers, Dept. Biol. Sci., '88-'91<br />

Drews, Jürgen, M.D., Pres., Intl. R & D, Hoffmann-La Roche Inc., '93-'96<br />

Gage, L. Patrick, Pres., Wyeth-Ayerst, '01<br />

Gill, Davinder, Vice President <strong>and</strong> Head Global Biotherapeutic Technologies, Pfizer, Inc. ’10-‘11<br />

Gussin, Robert, Corporate V. P., Johnson <strong>and</strong> Johnson, '01<br />

Haber, Edgar, M.D., Pres., Bristol-Myers Squibb Pharma. Res. Inst., '90-'91(deceased)<br />

Hait, William, M.D., Ph.D., Prof., Associate Dean & Director, CINJ, '06-'07<br />

Harris, Edward, Jr., M.D., Prof. & Chairman, Dept. Med., RWJ/UMDNJ '86-'88 (deceased)<br />

Inouye, Masayori, Ph.D., Prof. of Biochemistry, UMDNJ-RWJMS 1986-2010<br />

Koblan, Kenneth V.P. <strong>and</strong> Site Head <strong>for</strong> Basic Research, Merck, '06-'10<br />

Lerner, Irwin, Pres. & CEO, Hoffmann-La Roche Inc., '86-'93<br />

Levine, Arnold, Prof. & Chairman, Dept. Biol. Princeton, (Pres. Rockefeller U.), '89-'93<br />

Loh, Dennis, Vice President Preclinical Res. & Dev. Hoffmann-La Roche Inc. '98<br />

Lowry, Stephen F., M.D., Prof <strong>and</strong> Chair Dept. of Surgery, UMDNJ-RWJMS ’07-’11 (deceased)<br />

Luck, David, M.D., Ph.D., Prof., Rockefeller Univ., '94-'97 (deceased)<br />

Mansour, Tareq S., C.S. Interim Head Wyeth Research, ’09-‘10<br />

* Merrifield, Robert, Prof., Rockefeller Univ., '91-'93 (deceased)<br />

Moliteus, Magnus, Pres., Pharmacia, '86-'88<br />

Morris, N. Ronald, M.D., Associate Dean & Professor, Dept. of Pharma, RWJ/UMDNJ, '86-'92<br />

* Nathans, Daniel, M.D., Sr. Invest., HHMI, Johns Hopkins Univ., (Pres., Johns Hopkins, deceased), '93-'96<br />

Olson, Wilma, Prof., Dept. of Chemistry, Rutgers,'86-'92<br />

Palmer, James, CSO <strong>and</strong> Pres., Bristol-Myers Squibb Company, '00-'04 (deceased)<br />

Pickett, Cecil, Executive V.P., Schering-Plough, '00-'06<br />

Pramer, David, Dir., Waksman Inst. '86-'88<br />

Reynolds, Richard C., M.D., Executive Vice President, Robert Wood Johnson Foundation,'88-' 91<br />

Rosenberg, Leon, M.D., Pres., Bristol-Myers Squibb, '91-'97<br />

Ruddon, Raymond W., M.D., Ph.D., Corp. V.P. Johnson & Johnson, '01<br />

Ringrose, Peter, Pres., Bristol-Myers Squibb, 2000<br />

Sabatini, David, M.D., Ph.D., Prof. & Chair, Dept. Cell Biol. NYU School of Med., '93-' 08<br />

S<strong>and</strong>ers, Charles, M.D., Vice Chairman, E.R. Squibb & Sons, '88-'91<br />

Scolnick, Edward M., M.D., President, Merck & Co. Inc. , '95 - '97<br />

Shapiro, Bennett, M.D., Executive V.P., Merck & Company, '01<br />

84


* Sharp, Phillip, Prof. & Dir., Ctr. <strong>for</strong> Cancer Res., MIT, '90-'93<br />

Sigal, Elliott, M.D., Ph.D., President <strong>and</strong> CSO, Bristol Myers Squibb, '05-'08<br />

Strader, Catherine, Exec. V.P. <strong>and</strong> CSO, Schering-Plough Research Institute, '06<br />

Tilghman, Shirley M., Prof., Princeton University (Pres. Princeton U.), '01<br />

Torphy, Theodore John, Corp. V.P. <strong>and</strong> CSO, Corp. Office of Science <strong>and</strong> Technology J&J, '03-'07<br />

Turner, Mervyn J., Senior V.P. Merck & Co., '01-'05<br />

Vagelos, P. Roy, M.D., Chairman & CEO, Merck & Co., '86-'94<br />

Walsh, Frank S., Ph.D., Senior V.P. <strong>and</strong> Head Wyeth Research, '02-'08<br />

Wilson, Robert, Vice Chairman, Board of Directors, J & J, '91-'95<br />

* Nobel Laureate<br />

85


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

86


CENTER FOR ADVANCED BIOTECHNOLOGY AND MEDICINE<br />

GRANTS AND CONTRACTS SUMMARY<br />

FY <strong>2011</strong>: JULY 1, 2010 - JUNE 30, <strong>2011</strong><br />

DIRECT COSTS INDIRECT COSTS TOTAL COSTS<br />

FEDERAL 11,356,398 4,219,231 15,575,629<br />

FOUNDATION 931,211 234,430 1,165,641<br />

INDUSTRY 374,064 51,380 425,444<br />

STATE 522,723 29,327 552,050<br />

UNIVERSITY 25,000 0 25,000<br />

TOTAL 13,209,396 4,534,368 17,743,764


Anderson, Stephen Rutgers<br />

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

FY <strong>2011</strong>: 7/01/2010 - 6/30/<strong>2011</strong><br />

Total Award<br />

Start - End<br />

FY <strong>2011</strong><br />

Direct Costs<br />

FY <strong>2011</strong><br />

Indirect Costs<br />

FY <strong>2011</strong><br />

Total Costs<br />

Federal 1 $2,812,622<br />

$564,435 $252,145 $816,580<br />

NIH<br />

Human Transcription Factor Immunogens: Generation of a<br />

Complete Set<br />

$2,812,622 9/23/10 8/31/13 $564,435 $252,145 $816,580<br />

Total - Anderson, Stephen 1 $2,812,622<br />

$564,435 $252,145 $816,580<br />

Arnold, Edward Rutgers<br />

Federal 3 $6,920,622<br />

$887,573 $485,078 $1,372,651<br />

NIH<br />

HIV RNase H Natural Product Inhibitors: Structural <strong>and</strong><br />

Computational Biology (University of Pittsburgh subcontract)<br />

Inhibitors of Bacterial RNA Polymerase: “Switch Region” (PI -<br />

Dr. Richard Ebright)<br />

HIV-1 Reverse Transcriptase Structure: Function, Inhibition, <strong>and</strong><br />

Resistance (MERIT Award)<br />

$2,005,345 3/5/07 2/28/12 $255,526 $141,119 $396,645<br />

$1,161,788 1/1/07 12/31/11 $142,430 $77,118 $219,548<br />

$3,753,489 2/1/09 1/31/14 $489,617 $266,841 $756,458<br />

Total - Arnold, Edward 3 $6,920,622<br />

$887,573 $485,078 $1,372,651<br />

Das, Kalyan Rutgers<br />

Federal 1 $421,242<br />

$126,958<br />

$68,605 $195,563


NIH<br />

Defining <strong>and</strong> Targeting the HIV Nucleoside-Computing RT<br />

Inhibitor Binding Site<br />

Total Award<br />

Start - End<br />

FY <strong>2011</strong><br />

Direct Costs<br />

FY <strong>2011</strong><br />

Indirect Costs<br />

FY <strong>2011</strong><br />

Total Costs<br />

$421,242 6/15/10 5/31/12 $126,958 $68,605 $195,563<br />

Total - Das, Kalyan 1 $421,242<br />

$126,958 $68,605 $195,563<br />

Edery, Isaac Rutgers<br />

Federal 2 $2,679,362<br />

$436,224 $231,271 $667,495<br />

NIH<br />

Clock Mechanism Underlying Drosophila Rhythmic Behavior $1,327,486 2/15/08 1/31/12 $217,474 $113,146 $330,620<br />

Seasonal Adaptation of a Circadian Clock $1,351,876 7/1/10 6/30/14 $218,750 $118,125 $336,875<br />

Total - Edery, Isaac 2 $2,679,362<br />

$436,224 $231,271 $667,495<br />

Gélinas, Celine UMDNJ<br />

Federal 1 $737,308<br />

$57,404<br />

$32,146 $89,550<br />

NIH<br />

Functional Analysis of BFL-1/A1 in Apoptosis <strong>and</strong> Oncogenesis $737,308 6/3/08 3/31/12 $57,404 $32,146 $89,550<br />

Foundation 1 $600,000<br />

$180,018<br />

$19,982 $200,000


Leukemia & Lymphoma Society<br />

BFL-1 as a Therapeutic Target <strong>and</strong> Prognostic Indicator in B-CLL<br />

<strong>and</strong> AML<br />

Total Award<br />

Start - End<br />

FY <strong>2011</strong><br />

Direct Costs<br />

FY <strong>2011</strong><br />

Indirect Costs<br />

FY <strong>2011</strong><br />

Total Costs<br />

$600,000 10/1/08 9/30/11 $180,018 $19,982 $200,000<br />

Total - Gélinas, Celine 2 $1,337,308<br />

$237,422 $52,128 $289,550<br />

Inouye, Masayori UMDNJ<br />

Federal 3 $3,008,717<br />

$576,258 $284,717 $860,975<br />

NIH<br />

Deciphering of the Toxin-Antitoxin Systems in E. coli $1,593,094 1/1/09 12/31/12 $250,674 $140,377 $391,051<br />

The Method <strong>for</strong> Determination of Membrane Protein Structures<br />

without Purification<br />

Deciphering of the Toxin-Antitoxin Systems in E. coli (ARRA<br />

Supplement)<br />

$1,240,223 8/1/08 7/31/12 $198,167 $110,973 $309,140<br />

$175,400 6/18/10 11/30/11 $127,417 $33,367 $160,784<br />

Industry 1 $5,200,000<br />

$208,333<br />

$41,667 $250,000<br />

Takara Bio Inc.<br />

Takara Bio Initiative <strong>for</strong> Innovative <strong>Biotechnology</strong> $5,200,000 10/1/05 9/30/10 $208,333 $41,667 $250,000<br />

Total - Inouye, Masayori 4 $8,208,717<br />

$784,591 $326,384 $1,110,975<br />

Liu, Fang Rutgers


Total Award<br />

Start - End<br />

FY <strong>2011</strong><br />

Direct Costs<br />

FY <strong>2011</strong><br />

Indirect Costs<br />

FY <strong>2011</strong><br />

Total Costs<br />

University 1 $25,000<br />

$25,000<br />

$0 $25,000<br />

Rutgers University<br />

Smad3 as a Tumor Suppressor <strong>for</strong> Breast Cancer (Busch<br />

Memorial Fund grant)<br />

$25,000 7/1/10 5/1/12 $25,000 $0 $25,000<br />

Total - Liu, Fang 1 $25,000<br />

$25,000<br />

$0 $25,000<br />

Lobel, Peter UMDNJ<br />

Federal 2 $2,788,578<br />

$521,881 $292,254 $814,135<br />

NIH<br />

Novel Lysosomal Enzyne Deficient in Batten Disease (ARRA<br />

Award)<br />

$858,000 7/17/09 6/30/11 $275,000 $154,000 $429,000<br />

Lysosomal Enzymes <strong>and</strong> Associated Human Genetic Diseases $1,930,578 4/1/08 3/31/13 $246,881 $138,254 $385,135<br />

Foundation 2 $153,000<br />

$113,318<br />

$8,182 $121,500<br />

Ara Parseghian Medical Research Foundation<br />

Molecular Characterization of Niemann Pick C2 Disease $90,000 7/1/10 6/30/11 $81,818 $8,182 $90,000<br />

Batten Disease Support & Research Association<br />

Intrathecal Enzyme Replacement Therapy <strong>for</strong> LINCL (Predoctoral<br />

Fellowship - Su Xu)<br />

$63,000 8/1/09 7/31/11 $31,500 $0 $31,500


Total Award<br />

Start - End<br />

FY <strong>2011</strong><br />

Direct Costs<br />

FY <strong>2011</strong><br />

Indirect Costs<br />

FY <strong>2011</strong><br />

Total Costs<br />

Total - Lobel, Peter 4 $2,941,578<br />

$635,199 $300,436 $935,635<br />

Marcotrigiano, Joseph Rutgers<br />

Federal 1 $1,523,219<br />

$208,333 $103,500 $311,833<br />

NIH<br />

Mechanistic Studies of HCV E2 $1,523,219 9/15/10 8/31/14 $208,333 $103,500 $311,833<br />

State 1 $243,900<br />

$106,130<br />

$11,145 $117,275<br />

NJ Commission on Cancer Research<br />

Defining Essential Regions of E2 <strong>for</strong> HCV Infection $243,900 12/1/09 11/30/11 $106,130 $11,145 $117,275<br />

Total - Marcotrigiano, Joseph 2 $1,767,119<br />

$314,463 $114,645 $429,108<br />

Meng, Yu UMDNJ<br />

Foundation 1 $120,000<br />

$36,667<br />

$0 $36,667<br />

Batten Disease Support & Research Association<br />

Evaluation of Peripheral Enzyme Replacement Therapy <strong>for</strong><br />

LINCL <strong>and</strong> Development of Inducible Transgenic TPPI Model<br />

(Postdoctoral Fellowship)<br />

$120,000 8/1/10 7/31/13 $36,667 $0 $36,667<br />

Total - Meng, Yu 1 $120,000<br />

$36,667<br />

$0 $36,667<br />

Millonig, James UMDNJ<br />

Federal 3 $3,511,051<br />

$286,527<br />

$154,191 $440,718


NIH<br />

Identification <strong>and</strong> Functional Assessment of Autism Susceptibility<br />

Genes<br />

Elucidating the Role of miRNA Dysregulation in Schizophrenia<br />

<strong>and</strong> Bipolar Disorder (Rutgers University subcontract)<br />

A Mouse Knock-in Model <strong>for</strong> ENGRAILED 2 Autism<br />

Susceptibility (Phased Innovation Award - R21/R33)<br />

Total Award<br />

Start - End<br />

FY <strong>2011</strong><br />

Direct Costs<br />

FY <strong>2011</strong><br />

Indirect Costs<br />

FY <strong>2011</strong><br />

Total Costs<br />

$2,147,788 9/30/05 7/31/10 $26,336 $14,205 $40,541<br />

$830,697 4/1/08 3/31/12 $135,191 $75,707 $210,898<br />

$532,566 5/5/08 4/30/11 $125,000 $64,279 $189,279<br />

State 2 $1,069,550<br />

$416,593<br />

$18,182 $434,775<br />

NJ Commission on Spinal Cord Research<br />

Orphan GPCR, Gpr161, <strong>and</strong> Apithelial-Mesenchymal Transition $600,000 6/15/10 6/30/13 $181,818 $18,182 $200,000<br />

NJ Governor’s Council <strong>for</strong> Medical Research <strong>and</strong> Treatment of Autism<br />

Genetics <strong>and</strong> Non-genetic ENGRAILED 2 ASD Risk Factors $469,550 6/28/10 6/27/12 $234,775 $0 $234,775<br />

Total - Millonig, James 5 $4,580,601<br />

$703,120 $172,373 $875,493<br />

Molli, Poonam UMDNJ<br />

Federal 1 $351,000<br />

$75,000<br />

$42,000 $117,000


Department of Defense<br />

Tumor suppressor role of CAPER alpha in ER alpha negative &<br />

Rel NF-kB positive breast cancer (Postdoctoral Fellowship)<br />

Total Award<br />

Start - End<br />

FY <strong>2011</strong><br />

Direct Costs<br />

FY <strong>2011</strong><br />

Indirect Costs<br />

FY <strong>2011</strong><br />

Total Costs<br />

$351,000 8/15/09 8/14/12 $75,000 $42,000 $117,000<br />

Total - Molli, Poonam 1 $351,000<br />

$75,000 $42,000 $117,000<br />

Montelione, Gaetano Rutgers<br />

Federal 3 $39,383,043<br />

$6,304,310 $1,571,103 $7,875,413<br />

NIH<br />

Targeting the NS1 Protein <strong>for</strong> the Development of Influenza Virus<br />

Antivirals (University of Texas subcontract)<br />

$1,451,279 8/17/07 7/31/12 $203,244 $109,828 $313,072<br />

Structural Genomics of Eukaryotic Domain Families $37,810,498 9/1/10 6/30/15 $6,066,300 $1,442,501 $7,508,801<br />

Membrane Protein Production using the Yeast SPP System<br />

(UMDNJ subcontract)<br />

$121,266 9/30/10 7/31/14 $34,766 $18,774 $53,540<br />

Industry 1 $80,228<br />

$17,821<br />

$9,713 $27,534<br />

Nexomics Biosciences, Inc.<br />

Protein Production <strong>and</strong> NMR Data Collection Services $80,228 7/1/08 6/30/11 $17,821 $9,713 $27,534<br />

Total - Montelione, Gaetano 4 $39,463,271<br />

$6,322,131 $1,580,816 $7,902,947<br />

N<strong>and</strong>a, Vikas UMDNJ


Total Award<br />

Start - End<br />

FY <strong>2011</strong><br />

Direct Costs<br />

FY <strong>2011</strong><br />

Indirect Costs<br />

FY <strong>2011</strong><br />

Total Costs<br />

Federal 4 $4,722,697<br />

$688,625 $378,163 $1,066,788<br />

National Science Foundation<br />

NIH<br />

Design of Programmable, Self-Assembling Collagen Biomaterials $404,997 9/1/09 8/31/12 $86,538 $48,461 $134,999<br />

Computational Design of a Synthetic Extracellular Matrix $2,340,000 9/30/09 8/31/14 $304,545 $170,545 $475,090<br />

Structure-Based Engineering of Allergens to Enhance<br />

Digestibility<br />

A Computational Approach to Developing Heterochiral Peptide<br />

Therapeutics<br />

$426,660 4/1/10 3/31/12 $130,875 $73,290 $204,165<br />

$1,551,040 9/1/10 8/31/15 $166,667 $85,867 $252,534<br />

Total - N<strong>and</strong>a, Vikas 4 $4,722,697<br />

$688,625 $378,163 $1,066,788<br />

Phadtare, Sangita UMDNJ<br />

Federal 1 $156,000<br />

$45,833<br />

$25,667 $71,500<br />

NIH<br />

Transcription Antitermination by OB-fold Family Proteins (ARRA<br />

Award)<br />

$156,000 6/1/09 5/31/11 $45,833 $25,667 $71,500<br />

Total - Phadtare, Sangita 1 $156,000<br />

$45,833 $25,667 $71,500<br />

Shatkin, Aaron UMDNJ


Total Award<br />

Start - End<br />

FY <strong>2011</strong><br />

Direct Costs<br />

FY <strong>2011</strong><br />

Indirect Costs<br />

FY <strong>2011</strong><br />

Total Costs<br />

Industry 1 $35,000<br />

$35,000<br />

$0 $35,000<br />

Pharmaceutical Companies<br />

CABM Symposium & Lecture Series $35,000 7/1/10 6/30/11 $35,000 $0 $35,000<br />

Total - Shatkin, Aaron 1 $35,000<br />

$35,000<br />

$0 $35,000<br />

Stock, Ann UMDNJ<br />

Federal 2 $1,919,216<br />

$265,579 $125,390 $390,969<br />

NIH<br />

Structure <strong>and</strong> Function of Response Regulator Proteins (MERIT<br />

Award)<br />

Structure <strong>and</strong> Function of Response Regulator Proteins (ARRA<br />

Supplement)<br />

$1,792,012 7/1/09 6/30/14 $225,720 $114,367 $340,087<br />

$127,204 9/30/09 12/31/10 $39,859 $11,023 $50,882<br />

Foundation 1 $13,990,822<br />

$601,208 $206,266 $807,474<br />

Howard Hughes Medical Institute<br />

Molecular Mechanisms of Signal Transduction $13,990,822 8/1/94 2/28/11 $601,208 $206,266 $807,474<br />

Total - Stock, Ann 3 $15,910,038<br />

$866,787 $331,656 $1,198,443<br />

Williams, Keith Rutgers<br />

Industry 1 $112,910<br />

$112,910<br />

$0 $112,910


Cisco, Inc.<br />

Infrastructure Improvements <strong>for</strong> Extracellular Matrix Research: A<br />

Computational Design (Equipment Grant)<br />

Total Award<br />

Start - End<br />

FY <strong>2011</strong><br />

Direct Costs<br />

FY <strong>2011</strong><br />

Indirect Costs<br />

FY <strong>2011</strong><br />

Total Costs<br />

$112,910 5/31/11 6/30/11 $112,910 $0 $112,910<br />

Total - Williams, Keith 1 $112,910<br />

$112,910<br />

$0 $112,910<br />

Xiang, Mengqing UMDNJ<br />

Federal 2 $2,669,332<br />

$311,458 $173,001 $484,459<br />

NIH<br />

Transcriptional Regulation of Retinal Development $1,156,132 12/1/07 11/30/10 $103,125 $56,334 $159,459<br />

Dll4 Gene Regulation <strong>and</strong> Function during Retinogenesis $1,513,200 9/1/10 8/31/14 $208,333 $116,667 $325,000<br />

Total - Xiang, Mengqing 2 $2,669,332<br />

$311,458 $173,001 $484,459<br />

CABM TOTAL $13,209,396<br />

$4,534,368 $17,743,764

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

Saved successfully!

Ooh no, something went wrong!