09.06.2020 Views

2007 Scientific Report

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

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

VARI | <strong>2007</strong><br />

Van Andel Research Institute<br />

<strong>Scientific</strong> <strong>Report</strong> <strong>2007</strong>


Van Andel Research Institute <strong>Scientific</strong> <strong>Report</strong> <strong>2007</strong><br />

Cover photo: The glass sculpture “Life”, by Dale Chihuly,<br />

in the Van Andel Institute lobby.<br />

Photo by David Nadziejka.<br />

Van Andel Research Institute<br />

333 Bostwick Avenue, N.E., Grand Rapids, Michigan 49503<br />

Phone 616.234.5000 Fax 616.234.5001 www.vai.org


VARI | <strong>2007</strong><br />

Van Andel Research Institute <strong>Scientific</strong> <strong>Report</strong> <strong>2007</strong>


Van Andel Research Institute | <strong>Scientific</strong> <strong>Report</strong><br />

ii<br />

Published June <strong>2007</strong>.<br />

Copyright <strong>2007</strong> by the Van Andel Institute; all rights reserved.<br />

Van Andel Institute, 333 Bostwick Avenue, N.E.,<br />

Grand Rapids, Michigan 49503, U.S.A.


VARI | <strong>2007</strong><br />

Table of Contents<br />

Director’s Introduction 1<br />

George F. Vande Woude, Ph.D.<br />

Laboratory <strong>Report</strong>s 5<br />

Arthur S. Alberts, Ph.D.<br />

Cell Structure and Signal Integration 6<br />

Brian Cao, M.D.<br />

Antibody Technology 10<br />

Gregory S. Cavey, B.S.<br />

Mass Spectrometry and Proteomics 13<br />

Nicholas S. Duesbery, Ph.D.<br />

Cancer and Developmental Cell Biology 18<br />

Bryn Eagleson, B.S., RLATG<br />

Vivarium and Transgenics 21<br />

Kyle A. Furge, Ph.D.<br />

Computational Biology 23<br />

Brian B. Haab, Ph.D.<br />

Cancer Immunodiagnostics 26<br />

Rick Hay, Ph.D., M.D., F.A.H.A.<br />

Noninvasive Imaging and Radiation Biology<br />

Office of Translational Programs 31<br />

Jeffrey P. MacKeigan, Ph.D.<br />

Systems Biology 35<br />

Cindy K. Miranti, Ph.D.<br />

Integrin Signaling and Tumorigenesis 39<br />

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

Division of Quantitative Sciences<br />

Analytical, Cellular, and Molecular Microscopy<br />

Microarray Technology<br />

Molecular Epidemiology 46<br />

Pamela J. Swiatek, Ph.D., M.B.A.<br />

Germline Modification and Cytogenetics 51<br />

Bin T. Teh, M.D., Ph.D.<br />

Cancer Genetics 55<br />

Steven J. Triezenberg, Ph.D.<br />

Transcriptional Regulation 62<br />

George F. Vande Woude, Ph.D.<br />

Molecular Oncology 66<br />

Craig P. Webb, Ph.D.<br />

Program for Translational Medicine<br />

Tumor Metastasis and Angiogenesis 70<br />

Michael Weinreich, Ph.D.<br />

Chromosome Replication 74<br />

Bart O. Williams, Ph.D.<br />

Cell Signaling and Carcinogenesis 78<br />

H. Eric Xu, Ph.D.<br />

Structural Sciences 84<br />

iii


Van Andel Research Institute | <strong>Scientific</strong> <strong>Report</strong><br />

2006 Van Andel Research Institute Symposium 88<br />

Winning the War against Cancer: From Genomics to Bedside and Back<br />

Daniel Nathans Memorial Award 94<br />

Tony Hunter, Ph.D., and Tony Pawson, Ph.D.<br />

Postdoctoral Fellowship Program 96<br />

List of Fellows<br />

Student Programs 98<br />

Grand Rapids Area Pre-College Engineering Program<br />

Summer Student Internship Program<br />

Han-Mo Koo Memorial Seminar Series 102<br />

2006 | <strong>2007</strong> Seminars<br />

iv<br />

Van Andel Research Institute Organization 107<br />

Boards<br />

Office of the Director<br />

VAI Administrative Organization


VARI | <strong>2007</strong><br />

Director’s Introduction<br />

1


Van Andel Research Institute | <strong>Scientific</strong> <strong>Report</strong><br />

George F. Vande Woude<br />

Director’s Introduction<br />

2<br />

Now in our seventh year at a site in downtown Grand Rapids recently dubbed Medical Mile, the Van Andel Institute is embarking<br />

on a new phase in its growth as a leading center for biomedical research. An event for which we have been patiently<br />

waiting took place on April 12, <strong>2007</strong>, when groundbreaking ceremonies officially marked the beginning of construction for<br />

the Phase II expansion of the Van Andel Institute. Undeterred by April snow showers, Dave Van Andel got things rolling<br />

by maneuvering a GPS-directed John Deere bulldozer to break ground, to the cheers of employees and honored<br />

guests who watched from indoors via live video. For the next two years, we will see our Phase II laboratory emerge,<br />

fulfilling the plans to increase our research capacity to accommodate 400 new scientists. You can check out the construction at<br />

http://www.vai.org/About/Facilities/PhaseII.aspx.<br />

In addition to our own project, we are witnessing all around us phenomenal growth in the Medical Mile community. Already<br />

established south of us is the St. Mary’s Lacks Cancer Center. To our east and north, construction is underway for Spectrum<br />

Health’s new Lemmen-Holton Cancer Pavilion and the Helen DeVos Children’s Hospital. Just adjacent to our campus to the west<br />

and north, the Secchia Center that is being built will be headquarters to the College of Human of Medicine (CHM) of Michigan<br />

State University (MSU). Second-year CHM students will begin study in Grand Rapids in 2008, while a first-year class of 100<br />

students is scheduled to begin in 2010, leading to a full enrollment of about 400 students.<br />

It is hard to match all this excitement, but we have many accomplishments to our credit, and no doubt this has been a key factor<br />

stimulating Medical Mile and the growth of the biomedical enterprise. We are all very proud of what is happening.<br />

Personnel<br />

It is my pleasure to report that Jim Resau has been promoted to the rank of Distinguished <strong>Scientific</strong> Investigator. Jim has provided<br />

the impetus in developing VARI’s imaging core, and his efforts have led to novel imaging approaches and to many successful<br />

collaborations of benefit to our Institute. Jim has also contributed a strong interest and much help with the Van Andel Education<br />

Institute (VAEI) educational programs, including the graduate school, the Grand Rapids Area Pre-College Engineering Program,<br />

and other student programs of VAEI. He serves as VARI’s Deputy Director for Special Programs and Director of the Quantitative<br />

Sciences Division.<br />

Congratulations also to Eric Xu on his promotion to Distinguished <strong>Scientific</strong> Investigator. Eric has made significant scientific<br />

contributions to defining the structures of nuclear receptor proteins, including the peroxisome proliferator–activated receptors<br />

(PPARs) and the “orphan” nuclear receptors for which the ligand and function are unknown. The importance and excellence of<br />

his work is reflected in his success with NIH grants.


VARI | <strong>2007</strong><br />

In addition, four of VARI’s original investigators have been promoted to the rank of Senior <strong>Scientific</strong> Investigator: Art Alberts, Brian<br />

Cao, Nick Duesbery, and Bart Williams.<br />

Art’s studies on Diaphanous-related formins and the DAD peptide have developed new insights into the assembly of cell structures<br />

and the possibility of new approaches to cancer therapy. He has recently played a key role in establishing VARI’s flow<br />

cytometry facility.<br />

Brian Cao was recognized for the development of VARI’s state-of-the-art antibody technology lab. He has produced novel<br />

antibodies for several VARI research programs, developed and improved his lab’s capabilities to meet research needs, and<br />

further serves as director of the Michigan Antibody Technology Core of the Core Technology Alliance.<br />

Nick Duesbery’s work with anthrax lethal toxin has shown that the lethal factor component of the toxin is a metalloprotease that<br />

cleaves MAPK kinases. His lab’s work has increased our understanding of how anthrax toxin works and has also shown that the<br />

two-component moiety called “lethal toxin” inhibits the growth of some tumors. In addition to directing his lab, Nick also serves<br />

as VARI’s Deputy Director for Research Operations.<br />

Bart Williams has pursued the regulation and function of Wnt signaling as it affects various key cellular processes. The breadth<br />

of Wnt’s effects has led him from an initial interest in Wnt’s effects in tumorigenesis to the recognition of the role of Wnt in bone<br />

development and disease. Bart has also been a major contributor to the development of VARI’s mouse models and to the<br />

inception of the VAI graduate school.<br />

3<br />

We congratulate each of these researchers, and we look forward to their continued valuable contributions toward the Institute’s<br />

goals.<br />

We are pleased to announce the recruitment in 2006 of two exceptional principal investigators (PIs). Jeff MacKeigan, Ph.D., was<br />

recruited from Novartis and has established the Laboratory of Systems Biology. Jeff is interested in phosphatases and kinases,<br />

how they are regulated, and what signaling pathways they affect. He also brings platform screening technology to our program<br />

and has stimulated collaborations with our PIs to use RNAi screens as a genetic tool to understand gene function.<br />

Steve Triezenberg, Ph.D., was recruited from MSU and he wears two hats. In addition to Steve’s studies of herpes virus transcription<br />

in his newly established Laboratory of Transcriptional Regulation, he is also founding Dean of our new graduate program,<br />

established by VAEI. To Steve’s great credit, VAEI’s graduate school has an inaugural class of students that will arrive to begin<br />

studies in August <strong>2007</strong>. The Ph.D. program, like most of the research at VARI, will focus on the molecular, cellular, and genetic<br />

biology of human disease with a pronounced emphasis on translational research. The graduate school will foster the effective<br />

transition of students into professional scientists through a unique curriculum employing problem-based learning methods and<br />

through workshops to develop the cognate skills of grant and manuscript preparation, financial management, small-group leadership,<br />

and career planning.<br />

Programs<br />

On June 1, 2006, the Program for Translational Medicine was established under Craig Webb’s direction. This program will push<br />

forward our emphasis on moving our research findings into clinical practice and will help to develop “personalized medicine”<br />

founded on molecular-based, individual diagnosis and treatment. Craig’s staff will be developing strategies for data collection,<br />

integration, and analysis using the XB-BioIntegration Suite (formerly Xenobase), and Craig will work closely with the Office of<br />

Translational Programs, directed by Rick Hay, to help achieve VARI’s translational aims.


Van Andel Research Institute | <strong>Scientific</strong> <strong>Report</strong><br />

The Institute’s entire animal care and use program was evaluated in March <strong>2007</strong> by the Association of Assessment and<br />

Accreditation of Laboratory Animal Care, as part of our application for accreditation. AAALAC standards go beyond<br />

governmental regulations, and meeting their standards symbolizes quality, promotes scientific validity, demonstrates<br />

accountability, and shows commitment to humane animal care. The preliminary results of the review were very favorable, and<br />

we anticipate receiving approval and our formal accreditation in a timely manner. Our thanks to Pam Swiatek and Bryn Eagleson<br />

and their staffs, as well as to all the others involved in preparing for this evaluation; they did a great job in getting us ready.<br />

Grants<br />

In 2006, Eric Xu received his second R01 from NIH for a five-year study of “Structure and Function of Steroid Hormone Receptors”.<br />

Also, Brian Haab received his second R21 grant for “Defining Secreted Glycan Alterations in Pancreatic Cancer”.<br />

Rick Hay received a state appropriation from the MEDC Michigan Strategic Fund for “Creation of a Good Manufacturing Practices<br />

(GMP) Facility”. The project support runs from October 2006 through December <strong>2007</strong>.<br />

Steve Triezenberg’s graduate student, Sebla Kutluay, received VARI’s first predoctoral grant, for two years from the American<br />

Heart Association. Sponsored funding by commercial firms for specific research areas was received by the laboratory of Bin Teh<br />

and by my own lab. Other funding was received by various labs from the Breast Cancer Research Foundation, American Cancer<br />

Society, and as subgrants through collaborations with other research organizations.<br />

4<br />

Collaborations<br />

In late 2006, we established a collaboration for medical education and research between MSU and VARI. The new CHM medical<br />

school will establish an innovative molecular medicine curriculum with research in areas including cancer and neurobiology and<br />

an emphasis on translational research. The medical school faculty will have laboratory space in our Phase II building upon its<br />

completion in 2009, and the school intends to be fully operational in 2010. We anticipate that unique and fruitful collaborations<br />

will result from the proximity of the MSU and VARI scientists, and we foresee benefits accruing not only to both institutions, but<br />

more importantly to the patients afflicted by the diseases we study.<br />

Also in 2006, our joint effort with Spectrum Health has created the Center for Molecular Medicine, which offers molecular-based<br />

diagnostics to physicians. Further, a multi-member alliance under the name “ClinXus” offers a venue for novel biomarker-based<br />

clinical trials and for future biomarker drug development collaborations with pharmaceutical and biotech firms.<br />

In February <strong>2007</strong>, we signed a groundbreaking agreement with the National Cancer Center, Singapore (NCCS) to establish a joint<br />

translational research program in Singapore. The program will be directed by Bin Teh and will focus on the biological basis for<br />

different drug responses in Asian versus non-Asian patients having specific cancers.<br />

When we opened our doors in 2000, our commitment to basic sciences and translation was considered new and innovative.<br />

Lately, as I travel to other world-class academic institutions, it is clear that everyone has the same burning desire to turn discovery<br />

into application. This means that our success will be contingent not only on having the right scientific expertise, but also upon the<br />

growth of an ideal medical environment and a very supportive community. I know Grand Rapids has the “right stuff” and is poised<br />

to become a leading biomedical center in the next decade. It is an honor to be a part of such an exciting endeavor.


VARI | <strong>2007</strong><br />

Laboratory <strong>Report</strong>s<br />

5


Van Andel Research Institute | <strong>Scientific</strong> <strong>Report</strong><br />

Arthur S. Alberts, Ph.D.<br />

Laboratory of Cell Structure and Signal Integration<br />

6<br />

In 1993, Dr. Alberts received his Ph.D. in physiology and pharmacology at the University of California,<br />

San Diego, where he studied with James Feramisco. From 1994 to 1997, he served as a postdoctoral<br />

fellow in Richard Treisman’s laboratory at the Imperial Cancer Research Fund in London, England. From<br />

1997 through 1999, he was an Assistant Research Biochemist in the laboratory of Frank McCormick<br />

at the Cancer Research Institute, University of California, San Francisco. Dr. Alberts joined VARI as a<br />

<strong>Scientific</strong> Investigator in January 2000 and was promoted to Senior <strong>Scientific</strong> Investigator in 2006.<br />

Staff Students Visiting Scientists<br />

Laboratory Staff<br />

Students<br />

Visiting Scientists<br />

Jun Peng, M.D.<br />

Kathryn Eisenmann, Ph.D.<br />

Holly Holman, Ph.D.<br />

Richard A. West, M.S.<br />

Susan Kitchen, B.S.<br />

Aaron DeWard, B.S.<br />

Dagmar Hildebrand, B.S.<br />

Stephen Matheson, Ph.D.<br />

Brad Wallar, Ph.D.


VARI | <strong>2007</strong><br />

Research Interests<br />

Research in the Laboratory of Cell Structure and Signal Integration focuses on the molecular machinery responsible for the<br />

reorganization of the cell’s architecture during division and directed migration. Of particular interest is how defects in the<br />

machinery drive the progression to malignancy. The goal is to identify key control steps that are altered in disease states and<br />

exploit that knowledge to improve diagnostic and prognostic capabilities. We have been targeting key points in the cytoskeletal<br />

control system to devise novel targets for molecular therapy.<br />

The cytoskeleton comprises microfilaments, microtubules, and intermediate filaments. Each of these structures is a polymer<br />

whose assembly from individual monomer subunits is controlled by accessory proteins. While the term “cytoskeleton” implies a<br />

static or rigid structure within cells, the various filamentous structures are actually highly dynamic. Microfilaments, for example,<br />

are made of polymerized actin; these filaments rapidly polymerize, bundle, bend, depolymerize, or are severed so as to assume<br />

different shapes within the cell to fulfill a given function. In some cases, individual strands are woven into networks and contract<br />

against each other so that cells can attach to extracellular substrates and crawl along them. For example, actin/microfilament<br />

remodeling is crucial in the immune cells’ role to search for and destroy invading pathogens. Cancer cells use such remodeling<br />

to migrate from primary tumors (often located at an innocuous site) to a secondary site. At the secondary site, tumor cells grow<br />

and damage adjacent tissue, often leading to the eventual death of the patient. This process is called metastasis, and to date<br />

there are few, if any, effective anti-cancer therapies that block it. Thus, there is an important need to identify mechanisms that can<br />

be effectively targeted to block the spread of tumor cells throughout the body.<br />

The Rho family of small GTPases controls critical steps in cytoskeletal remodeling. The GTPases are triggered by signals dictated<br />

by activated growth or adhesion receptors and, in turn, bind to “effectors” that govern the machinery assembling the cytoskeleton.<br />

Some of these effector proteins directly participate in cytoskeletal remodeling. One fundamentally important set of GTPase<br />

effectors is the mammalian Diaphanous-related (mDia) formins.<br />

7<br />

Formins nucleate, processively elongate, and (in some cases) bundle filamentous actin (F-actin) through conserved formin<br />

homology-2 (FH2) domains. mDia proteins participate in many cytoskeletal remodeling events including cytokinesis, vesicle trafficking,<br />

and filopodia assembly while acting as effectors for Rho small GTPases. Rho proteins govern mDia proteins by regulating<br />

an intramolecular autoregulatory mechanism. GTPases binding to the mDia amino-terminal GTPase-binding domain (GBD)<br />

sterically hinder the adjacent Dia-inhibitory domain (DID) interaction with the carboxyl-terminal Dia-autoregulatory (DAD) domain<br />

(Fig. 1). The release of DAD allows the adjacent FH2 domain to then nucleate and elongate nonbranched actin filaments.<br />

Figure 1.<br />

Figure 1. mDia proteins are autoregulated nucleators<br />

of actin. Autoinhibition of mDia is mediated by interaction<br />

between the DID and DAD domains. Activated GTP-bound<br />

Rho proteins bind to the GBD where they interfere with DAD<br />

binding to DID. Then the free FH2 domains, which also function<br />

as dimerization interfaces, can nucleate actin monomers and<br />

processively elongate actin filaments. Tagged fusion proteins<br />

(CFP-Rho GTPase and YFP-mDia) are used in fluorescence<br />

resonance energy transfer (FRET) to monitor the sites of<br />

protein-protein interactions. Excitation of CFP by a specific<br />

wavelength of light results in emitted light at a wavelength that<br />

excites YFP, but YFP excitation occurs only if the proteins are<br />

close enough to approximate direct binding. This approach is<br />

used to generate the data shown in Fig. 2.


Van Andel Research Institute | <strong>Scientific</strong> <strong>Report</strong><br />

The cytoskeleton not only provides the impetus for cell movement, but it also allows the internal architecture to be organized into<br />

different compartments having specific functions in the cellular responses to growth factors. Rho GTPases and the dynamic<br />

assembly and disassembly of actin filaments have been shown to have crucial roles in both the internalization and trafficking of<br />

growth factor receptors. While all three mammalian Diaphanous-related formins (mDia1, mDia2, and mDia3) have been localized<br />

on endosomes, their roles in actin nucleation, filament elongation, and/or bundling remains poorly understood in the context of<br />

intracellular trafficking.<br />

In a recent publication in Experimental Cell Research, we reported the functional relationship between RhoB, a GTPase known<br />

to associate with both early and late endosomes, and the formin mDia2. We were able to show that 1) RhoB and mDia2 interact<br />

on endosomes, as seen in Fig. 2 using the FRET approach; 2) GTPase activity—the ability to hydrolyze GTP to GDP—is required<br />

for the ability of RhoB to govern endosome dynamics; and 3) the actin dynamics controlled by RhoB and mDia2 is necessary for<br />

vesicle trafficking. These studies further suggested that Rho GTPases significantly influence the activity of mDia family formins in<br />

driving cellular membrane remodeling through the regulation of actin dynamics.<br />

8<br />

In another recent study, in the journal Current Biology, we reported how Diaphanous-interacting protein (DIP) binds to and<br />

regulates the activity of the formin mDia2 and its ability to assemble filopodia. Filopodia are small finger-like projections<br />

comprising several bundled nonbranched actin filaments emanating from the leading edge of migrating cells and essentially<br />

acting as sensors for directed cell movement. We investigated an interaction occurring between a conserved leucine-rich<br />

region (LRR) in DIP and the mDia FH2 domain. While DIP has been shown to interact with and stimulate N-WASp-dependent<br />

branched filament assembly via Arp2/3, it interfered with mDia2-dependent filament assembly and bundling.<br />

Figure 2.<br />

Figure 2. RhoB and mDia2 interact on a subset of vesicles bearing internalized EGF. CFP-RhoB and YFP-mDia2 interact on vesicles<br />

bearing internalized Texas Red–labeled epidermal growth factor. Cells expressing the two FRET probes (4 h after injection) were<br />

incubated with fluorescent EGF for 5 min prior to fixation. RhoB-mDia2 FRET occurs on a subset of vesicles (FRET is false-colored<br />

green, with Texas Red–EGF shown in red).


VARI | <strong>2007</strong><br />

Surprisingly, DIP had no effect on the highly related mDia1. Consistent with a role for mDia2 as a Cdc42 effector, DIP both blocked<br />

the formation of filopodia and induced non-apoptotic membrane blebbing, a physiological process involved in both cytokinesis<br />

and amoeboid cell movement. DIP-induced blebbing occurred independently of Arp2/3 activity. Figure 3 shows the result of<br />

microinjection of DIP LRR into a mouse embryo fibroblast in which a critical subunit of Arp2/3 has been knocked down by siRNA.<br />

The experiment reveals a pivotal role for DIP in the control of nonbranched versus branched actin filament assembly mediated,<br />

respectively, by Diaphanous-related formins and by activators of Arp2/3. The ability of DIP to trigger blebbing also suggests a<br />

role for mDia2 in the assembly of actin filaments at the cell cortex necessary for the maintenance of plasma membrane integrity.<br />

Future experiments will address how DIP regulates mDia2 in directed cell movement and during cell division.<br />

Figure 3.<br />

Figure 3. DIP LRR–induced plasma membrane blebbing<br />

does not require Arp2/3 activity. This mouse embryo<br />

fibroblast, which expresses siRNA directed against<br />

Arp3, was injected with 0.1 μM recombinant DIP LRR<br />

protein along with Texas Red dextran as a marker.<br />

9<br />

External Collaborators<br />

Harry Higgs, Dartmouth Medical School,<br />

Hanover, New Hampshire<br />

Recent Publications<br />

From left: Peng, Holman, DeWard, Eisenmann, Kitchen, Alberts, Hildebrand<br />

Eisenmann, Kathryn M., Elizabeth S. Harris, Susan M. Kitchen, Holly A. Holman, Henry N. Higgs, and Arthur S. Alberts. <strong>2007</strong>.<br />

Dia-interacting protein modulates formin-mediated actin assembly at the cell cortex. Current Biology 17(7): 579–591.<br />

Wallar, Bradley J., Aaron D. DeWard, James H. Resau, and Arthur S. Alberts. <strong>2007</strong>. RhoB and the mammalian Diaphanousrelated<br />

formin mDia2 in endosome trafficking. Experimental Cell Research 313(3): 560–571.


Van Andel Research Institute | <strong>Scientific</strong> <strong>Report</strong><br />

Brian Cao, M.D.<br />

Laboratory of Antibody Technology<br />

10<br />

Dr. Cao obtained his M.D. from Peking University Medical Center, People’s Republic of China, in 1986.<br />

On receiving a CDC fellowship award, he was a visiting scientist at the National Center for Infectious<br />

Diseases, Centers for Disease Control and Prevention in Atlanta (1991–1994). He next served as a<br />

postdoctoral fellow at Harvard (1994–1995) and at Yale (1995–1996). From 1996 to 1999, Dr. Cao was<br />

a Scientist Associate in charge of the Monoclonal Antibody Production Laboratory at the Advanced<br />

BioScience Laboratories–Basic Research Program at the National Cancer Institute, Frederick Cancer<br />

Research and Development Center, Maryland. Dr. Cao joined VARI as a Special Program Investigator in<br />

June 1999, and he was promoted to Senior <strong>Scientific</strong> Investigator in July 2006.<br />

Staff<br />

Laboratory Staff<br />

Ping Zhao, M.S.<br />

Tessa Grabinski, B.S.<br />

Students<br />

Students<br />

Xin Wang<br />

Ning Xu<br />

Aixia Zhang<br />

Jin Zhu<br />

Visiting Scientists


VARI | <strong>2007</strong><br />

Research Interests<br />

Antibodies are primary tools of biomedical science. In basic research, the characterization and analysis of almost any molecule<br />

involves the production of specific monoclonal or polyclonal antibodies that react with it. Antibodies are also widely used in<br />

diagnostic applications for clinical medicine. ELISA and radioimmunoassay systems are antibody-based. Analysis of cells<br />

and tissues in pathology laboratories includes the use of antibodies on tissue sections and in flow cytometry analyses. Further,<br />

antibodies are making rapid inroads into medical therapeutics, driven by technological evolution from chimeric and humanized to<br />

fully human antibodies. The therapeutic antibody market has the potential to reach $30 billion by 2010.<br />

Our Antibody Technology laboratory has developed several technologies over the last few years: 1) state-of-the-art monoclonal<br />

antibody (mAb) production and characterization, followed by scaled-up production and purification; 2) antibody-binding-site<br />

epitope mapping using a phage-display peptide library; 3) a human-antibody-fragment phage-display library and screening<br />

of specific fragments from the library; and 4) characterization of these human antibody fragments and conjugation with<br />

chemotherapeutics to generate immuno-chemotherapeutic reagents for preclinical studies.<br />

In collaboration with Nanjing Medical University, China, we constructed our own human naïve Fab fragment phage-display library,<br />

with a diversity of 2 × 10 9 , in late 2004. In 2005, we screened out several Fab fragments from the library that specifically recognize<br />

HGF/SF, Met, and EGFR. By modifying and improving biopanning strategies, we have selected Fab fragments that recognize<br />

the Met and EGFR extracellular domains in native conformation with reasonable affinity and, importantly, with the internalization<br />

property that makes these Fabs attractive as conjugate reagents for immuno-chemotherapy or immuno-radiation therapy against<br />

cancer. In the past year, we have conjugated anti-EGFR human Fab to paclitaxel (Taxol) as an immuno-chemotherapy agent and<br />

investigated its in vitro anti-tumor efficacy on A431 epidermoid carcinoma cells using cell proliferation inhibition and apoptosis<br />

assays. The Fab-Taxol conjugate inhibited A431 cell proliferation at low concentrations and in a dose-responsive manner; more<br />

than 70% inhibition was observed at 52 pM. Furthermore, almost 100% of the cells underwent apoptosis after treatment with<br />

Fab-Taxol at 26 pM for 48 hours. The in vitro anti-tumor efficacy is four- to fivefold more potent than Taxol alone. We are modifying<br />

the Taxol conjugation conditions and working with other drug conjugations to investigate their in vivo anti-tumor efficacy in<br />

xenograft and orthotopic animal models.<br />

11


Van Andel Research Institute | <strong>Scientific</strong> <strong>Report</strong><br />

Functioning as an antibody production core facility, this lab has extensive capabilities. Our technologies and services<br />

include antigen preparation and animal immunization; peptide design and coupling to protein carriers; DNA immunization<br />

(gene-gun technology); immunization with living or fixed cells; conventional antigen/adjuvant preparation; immunizing a wide<br />

range of antibody-producing models (including mice, rats, rabbits, human cells, and transgenic or knock-out mice); and in vitro<br />

immunization. Our work also includes the generation of hybridomas from spleen cells of immunized mice, rats, and<br />

rabbits; hybridoma expansion and subcloning; cryopreservation of hybridomas secreting mAbs; isotyping of mAbs; ELISA<br />

screening of hybridoma supernatants; mAb characterization by immunoprecipitation, Western blot, immunohistochemistry,<br />

immunofluorescence staining, FACS, and in vitro bioassays; generation of bi-specific mAbs by secondary fusion; conjugation<br />

of mAbs to enzymes, biotin/streptavidin, or fluorescent reporters; and development of detection methods/kits such as sandwich<br />

ELISA. We also contract services to biotechnology companies, producing and purifying mAbs for their research and for<br />

diagnostic kit development.<br />

The Michigan Core Technology Alliance (CTA), funded by the state government, was created in 2001. The Antibody Technology<br />

Core at VARI and the Hybridoma Core at the University of Michigan in Ann Arbor joined together to form the Michigan Antibody<br />

Technology Core (MATC) and became the seventh core of CTA in March 2005. Our goals are to provide state-of-the-art antibody<br />

technologies and services to research scientists; to generate, characterize, produce, and purify a wide variety of monoclonal<br />

antibodies; to make human antibody fragments and humanize murine mAbs for clinical diagnostic/therapeutic applications; and<br />

to advance biomedical research and development. The Antibody Technology Lab at VARI serves as the core’s hub, and Dr. Brian<br />

Cao is the director of MATC.<br />

12<br />

From left: Gu, Zhang, Xu, Zhao, Nelson, Grabinski, Cao<br />

Recent Publications<br />

Wang, X., J. Zhu, P. Zhao, Y. Jiao, N. Xu, T. Grabinski, C. Liu, C.K. Miranti, T. Fu, and B. Cao. In press. In vitro efficacy of immunochemotherapy<br />

with anti-EGFR human Fab-Taxol conjugate on A431 epidermoid carcinoma cells. Cancer Biology & Therapy.<br />

Zhang, Y.-W., B. Staal, Y. Su, P. Swiatek, P. Zhao, B. Cao, J. Resau, R. Sigler, R. Bronson, and G.F. Vande Woude. <strong>2007</strong>. Evidence<br />

that MIG-6 is a tumor-suppressor gene. Oncogene 26(2): 269–276.<br />

Tsarfaty, Galia, Gideon Y. Stein, Sharon Moshitch-Moshkovitz, Dafna W. Kaufman, Brian Cao, James H. Resau, George F. Vande<br />

Woude, and Ilan Tsarfaty. 2006. HGF/SF increases tumor blood volume: a novel tool for the in vivo functional molecular imaging<br />

of Met. Neoplasia 8(5): 344–352.


VARI | <strong>2007</strong><br />

Gregory S. Cavey, B.S.<br />

Laboratory of Mass Spectrometry and Proteomics<br />

Mr. Cavey received his B.S. degree from Michigan State University in 1990. Prior to joining VARI he was<br />

employed at Pharmacia in Kalamazoo, Michigan, for nearly 15 years. As a member of a biotechnology<br />

development unit, he was group leader for a protein characterization core laboratory. More recently as a<br />

research scientist, he was principal in the establishment and application of a state-of-the-art proteomics<br />

laboratory for drug discovery. Mr. Cavey joined VARI as a Special Program Investigator in July 2002.<br />

13<br />

Staff<br />

Laboratory Staff<br />

Paula Davidson, M.S.<br />

Joan Krilich, B.S.<br />

Students<br />

Visiting Scientists


Van Andel Research Institute | <strong>Scientific</strong> <strong>Report</strong><br />

Research Interests<br />

The Mass Spectrometry and Proteomics laboratory provides protein identification analysis and protein molecular weight<br />

determination as core services. Nanogram amounts of protein in SDS-PAGE gels or in solution are digested into peptides and<br />

analyzed by HPLC with on-line electrospray mass spectrometry. Peptides are fragmented in the mass spectrometer to generate<br />

amino acid sequence data that is used to identify proteins by searching protein and DNA databases. Submicrogram amounts<br />

of intact proteins are analyzed by nanoscale liquid chromatography–mass spectrometry (LC-MS) to determine their average<br />

molecular weight; this work is performed using a variety of HPLC columns to optimize recovery and provide reliable results.<br />

These core services are provided to both VARI investigators and external clients. Research in the lab focuses on improving<br />

existing services and developing new methods based on the needs of VARI investigators. Our three main areas of interest are<br />

intact-protein molecular weight determination, phosphopeptide analysis, and protein expression profiling using LC-MS.<br />

Protein LC-MS<br />

14<br />

We use protein LC-MS to confirm correct expression and purification of recombinant proteins from bacteria. The average<br />

molecular weight of a protein is experimentally determined and compared with the calculated weight from the expected amino<br />

acid sequence. Proteins of 50 kDa and larger are analyzed with mass accuracy often better than 0.01%, or ±1 Da per 10 kDa.<br />

Unlike with conventional SDS-PAGE, protein truncation and modifications such as oxidation or acetylation can be accurately<br />

characterized using protein LC-MS. This information is essential when protein reagents are used for labor-intensive and costly<br />

protocols such as x-ray crystallography, antibody production, or drug screening. We have a dedicated LC-MS instrument with<br />

optimized HPLC separation and comprehensive data processing for analyzing complex mixtures of proteins. For proteins that<br />

degrade during purification, we can alter the use of protease inhibitors or minimize degradation through site-directed mutagenesis<br />

of susceptible amino acids. We are also exploring the use of this equipment for biomarker discovery of intact proteins. The goal<br />

is to provide relative quantitation of proteins in disease cell culture models, tumor tissue, and cancer patient body fluids.<br />

Protein phosphorylation analysis<br />

Mapping post-translational modifications of proteins such as phosphorylation is an important yet difficult undertaking in cancer<br />

research. Phosphorylation regulates many protein pathways that could serve as potential drug targets in cancer therapy. In recent<br />

years, mass spectrometry has emerged as a primary tool in determining site-specific phosphorylation and relative quantitation.<br />

Phosphorylation analysis is complicated by many factors, but principally by the low-stoichiometry modifications that may regulate<br />

pathways: we are sometimes dealing with 0.01% or less of phosphorylated protein among a large excess of a nonphosphorylated<br />

counterpart. Our lab collaborates with investigators to map protein phosphorylation using techniques including multiple<br />

enzyme digestion, titanium dioxide phosphopeptide enrichment, and phosphorylation-specific mass spectrometry detection.<br />

Although trypsin is often the enzyme of choice for digesting proteins into peptides for identification, additional enzymes such as<br />

Lys-C, Staph V8, chymotrypsin, thermolysin, or elastase may also be employed. Multiple enzyme digests and titanium dioxide<br />

enrichment are used in combination with precursor ion scanning for –79 m/z on a Waters Q-Tof Premier mass spectrometer.


VARI | <strong>2007</strong><br />

We have developed a robust negative-ion-mode method using nanoscale HPLC that provides specific detection of phosphopeptides<br />

below 20 fmol in the presence of 2 pmol of nonphosphorylated protein. Once detected in the negative mode, phosphopeptides<br />

are sequenced in a subsequent LC-MS analysis in the positive ion mode using accurate mass parent ion selection, a narrow<br />

retention time window, and collision energy ramping. This approach has provided a reliable and sensitive means of analyzing<br />

phosphoproteins in our laboratory. Our current focus is on applying this label-free method to studies requiring relative quantitation<br />

of phosphorylation events.<br />

Protein expression/biomarker discovery<br />

As mass spectrometry instruments and protein separation methods develop, proteomics techniques allow researchers to identify<br />

and quantitate protein samples of increasing complexity. The ultimate goal is to catalog all proteins expressed in a given<br />

cell or tissue as a means of evaluating dynamic physiological events and understanding how all proteins interact to affect a<br />

biological outcome. Traditionally this goal has been approached using 2D gel electrophoresis, image analysis of stained proteins,<br />

and identification of proteins from gels using mass spectrometry. Because of the labor-intensive nature of 2D gels and the<br />

underrepresentation of some protein classes (such as membrane proteins), proteomics has been moving toward solution-based<br />

separations and direct mass spectrometry analysis. Our laboratory recently purchased and installed a Waters Corporation Protein<br />

Expression System for non-gel-based, label-free protein expression analysis. This system represents a paradigm shift in the<br />

field of proteomics, because it provides both quantitative and qualitative data on complex mixtures of proteins in a single LC-MS<br />

analysis. Proteins are enzymatically digested using trypsin and, without any chemical or isotopic labeling, the resulting peptides<br />

are analyzed by LC-MS. The combination of molecular mass and LC retention time establishes a signature for each peptide and<br />

allows comparison across samples. The mass spectrometer signal intensity of each peptide is used for quantitation. Qualitative<br />

protein identification data is obtained by fragmenting all peptides eluting into the mass spectrometer, a feature unique to the<br />

Waters instrument. VARI is one of an elite group of institutions that have this powerful new technology. This system will be used to<br />

map protein pathways under a systems biology approach and to discover potential biomarkers for early detection and diagnosis<br />

in cancer and other diseases.<br />

15<br />

External Collaborators<br />

Gary Gibson, Henry Ford Hospital, Detroit, Michigan<br />

Michael Hollingsworth, Eppley Cancer Center, University of Nebraska, Omaha<br />

Waters Corporation<br />

Core Technology Alliance (CTA)<br />

This laboratory participates in the CTA as a<br />

member of the Michigan Proteomics Consortium.<br />

From left: Davidson, Cavey, Krilich


Van Andel Research Institute | <strong>Scientific</strong> <strong>Report</strong><br />

16<br />

Cells prepared by Miles Qian and Daisuke Matsuda<br />

of the Teh laboratory.<br />

Image by Kristin VendenBeldt of the Resau laboratory.


VARI | <strong>2007</strong><br />

Murine lymph node/vascular tissue.<br />

17<br />

Murine lymph node/vascular tissues stained by immunohistochemisty and photographed using the CRI Nuance camera. Green, pericyte cell marker;<br />

red, CD34 blood vessel marker; blue, nuclear/DNA marker.


Van Andel Research Institute | <strong>Scientific</strong> <strong>Report</strong><br />

Nicholas S. Duesbery, Ph.D.<br />

Laboratory of Cancer and Developmental Cell Biology<br />

18<br />

Dr. Duesbery received a B.Sc. (Hon.) in biology (1987) from Queen’s University, Canada, and both his<br />

M.Sc. (1990) and Ph.D. (1996) degrees in zoology from the University of Toronto, Canada, under the<br />

supervision of Yoshio Masui. Before his appointment as a <strong>Scientific</strong> Investigator at VARI in April 1999,<br />

he was a postdoctoral fellow in the laboratory of George Vande Woude in the Molecular Oncology<br />

Section of the Advanced BioScience Laboratories–Basic Research Program at the National Cancer<br />

Institute, Frederick Cancer Research and Development Center, Maryland. Dr. Duesbery was promoted<br />

to Senior <strong>Scientific</strong> Investigator and appointed Deputy Director for Research Operations in 2006.<br />

Staff<br />

Students<br />

Jennifer Bromberg-White, Ph.D.<br />

Philippe Depeille, Ph.D.<br />

Yan Ding, Ph.D.<br />

John Young, M.S.<br />

Jaclyn Lynem, B.S.<br />

Elissa Boguslawski<br />

Laura Holman<br />

Students<br />

Visiting Scientists<br />

Chih-Shia Lee, M.S.<br />

Naomi Asantewa-Sechereh<br />

Lisa Orcasitas


VARI | <strong>2007</strong><br />

Research Interests<br />

Many malignant sarcomas such as fibrosarcomas are refractory to available treatments. However, sarcomas possess unique<br />

vascular properties which indicate they may be more responsive to therapeutic agents that target endothelial function.<br />

Mitogen-activated protein kinase kinases (MKKs) have been shown to play an essential role in the growth and vascularization of<br />

carcinomas, and we hypothesize that signaling through multiple MKK pathways is also essential for sarcomas. The objective of<br />

our research is to define the role of MKK signaling in the growth and vascularization of human sarcomas and to determine whether<br />

inhibition of multiple MKKs by agents such as anthrax lethal toxin (LeTx), a proteolytic inhibitor of MKKs, can form the basis of a<br />

novel and innovative approach to the treatment of human sarcoma.<br />

In the past year we have made substantial progress in achieving this objective. Yan Ding, a postdoctoral fellow in the lab, and<br />

Lisa Orcasitas have shown that MKKs are active in fibrosarcoma and that LeTx can inhibit the in vitro tumorigenic potential<br />

of cells derived from human fibrosarcoma. The anti-tumoral properties of LeTx probably stem from its ability to substantially<br />

decrease the release of many growth factors, notably the pro-angiogenic vascular endothelial growth factor (VEGF). In vivo, LeTx<br />

caused a substantial decrease in both tumor volume and mean vascular density of fibrosarcoma xenografts. These changes also<br />

correlated with a decreased level of pro-angiogenic factors, including VEGF. Dr. Ding also found that the ability of LeTx to<br />

decrease the release of VEGF was not limited to fibrosarcoma, but was observed in cell lines derived from various sarcomas<br />

including malignant fibrous histiocytoma and leiomyosarcoma. These results are consistent with the hypothesis that MKK<br />

signaling is required for the growth and vascularization of fibrosarcoma both in vitro and in vivo, and this probably is also true of<br />

other types of soft-tissue sarcomas.<br />

Similarly, using an endothelial model of Kaposi sarcoma, Philippe Depeille, another postdoctoral fellow, and Elissa Boguslawski<br />

showed that in vitro, LeTx 1) decreases proliferation, 2) inhibits tumorigenesis, and 3) dramatically reduces the secretion<br />

of angioproliferative cytokines such as VEGF. Furthermore, in vivo, systemic treatment with LeTx inhibits tumor growth and<br />

vascularization. These findings support the importance of MKK pathways in the release of angioproliferative cytokines that<br />

promote tumor growth and vascularization. Our data suggest that inhibition of MKK signaling may be an effective therapeutic<br />

strategy for the treatment of Kaposi sarcoma.<br />

19<br />

In collaboration with Bart Williams’ lab, John Young, our senior technician, and Jennifer Bromberg-White, a postdoctoral fellow,<br />

investigated the mechanism of anthrax toxin entry into cells. Together they showed that mice or cells lacking LRP6, or a related<br />

protein called LRP5, are still susceptible to anthrax toxin. The discovery that anthrax toxin can enter cells without the help of LRP6<br />

presents a significant challenge to the published models of anthrax toxin function. These findings will help focus the efforts of<br />

scientists working on new ways to treat anthrax.<br />

In collaboration with Arthur Frankel, director of the Scott & White Cancer Research Institute in Texas, we have also tested the<br />

therapeutic potential of LeTx in the treatment of malignant melanoma. Progress to date indicates that melanoma is particularly<br />

sensitive to MKK inhibition. This is likely due in part to the fact that more than 80% of melanoma tumors harbor somatic mutations<br />

that cause constitutive activation of the MKK1 and MKK2 signaling pathways, though indirect evidence suggests that other MKK<br />

pathways also play a role in melanoma progression. Chih-Shia Lee is performing a detailed study of the individual contributions<br />

of MKK pathways to melanoma survival. Jaclyn Lynem and Naomi Asantewa-Sechereh are investigating the molecular basis<br />

of LF inactivation of MKK. We are currently performing preclinical studies to evaluate the potential of LeTx as a therapeutic for<br />

malignant melanoma.


Van Andel Research Institute | <strong>Scientific</strong> <strong>Report</strong><br />

20<br />

From left: Asantewa-Sechereh, Orcasitas, Lynem, Boguslawski, Lee,<br />

Bromberg-White, Duesbery, Holman, Young, Depeille<br />

Recent Publications<br />

Young, J.J., J.L. Bromberg-White, C.R. Zylstra, J. Church, E. Boguslawski, J. Resau, B.O. Williams, and N. Duesbery. In press.<br />

LRP5 and LRP6 are not required for protective antigen-mediated internalization or lethality of anthrax lethal toxin. PLoS Pathogen.<br />

Depeille, P.E., Y. Ding, J.L. Bromberg-White, and N.S. Duesbery. <strong>2007</strong>. MKK signaling and vascularization. Oncogene 26(9):<br />

1290–1296.<br />

Abi-Habib, Ralph J., Ravibhushan Singh, Stephen H. Leppla, John J. Greene, Yan Ding, Bree Berghuis, Nicholas S. Duesbery,<br />

and Arthur E. Frankel. 2006. Systemic anthrax lethal toxin therapy produces regressions of subcutaneous human melanoma<br />

tumors in athymic nude mice. Clinical Cancer Research 12(24): 7437–7443.<br />

Bodart, Jean-François L., and Nicholas S. Duesbery. 2006. Xenopus tropicalis oocytes: more than just a beautiful genome.<br />

In Xenopus Protocols: Cell Biology and Signal Transduction, X. Johné Liu, ed. Methods in Molecular Biology series, Vol. 322.<br />

Totowa, N.J.: Humana Press, pp. 43–53.


VARI | <strong>2007</strong><br />

Bryn Eagleson, B.S., RLATG<br />

Vivarium and Laboratory of Transgenics<br />

Bryn Eagleson began her career in laboratory animal services in 1981 with Litton Bionetics at the<br />

National Cancer Institute’s Frederick Cancer Research and Development Center (NCI–Frederick) in<br />

Maryland. In 1983, she joined the Johnson & Johnson Biotechnology Center in San Diego, California.<br />

In 1988, she returned to the NCI–Frederick, where she continued to develop her skills in transgenic<br />

technology and managed the transgenic mouse colony. In 1999, she joined VARI as the Vivarium Director<br />

and Transgenics Special Program Manager.<br />

21<br />

Technical Staff<br />

Lisa DeCamp, B.S.<br />

Laboratory Staff<br />

Dawna Dylewski, B.S.<br />

Audra Guikema, B.S., L.V.T.<br />

Kellie Jilbert, B.S., A.S.<br />

Jamie Bondsfield, A.S.<br />

Elissa Boguslawski, RALAT<br />

Students<br />

Animal Caretaker Staff<br />

Sylvia Marinelli, Team leader<br />

Angie Rogers, B.S.<br />

Crystal Brady<br />

Jarred Grams<br />

Janelle Post<br />

Tina Schumaker<br />

Michael Shearer<br />

Bobbie Vitt<br />

Visiting Scientists


Van Andel Research Institute | <strong>Scientific</strong> <strong>Report</strong><br />

Research Interests<br />

The goal of the vivarium and the transgenics laboratory is to develop, provide, and support high-quality mouse modeling services<br />

for the Van Andel Research Institute investigators, Michigan Technology Tri-Corridor collaborators, and the greater research<br />

community. We use two Topaz Technologies software products, Granite and Scion, for integrated management of the vivarium<br />

finances, the mouse breeding colony, and the Institutional Animal Care and Use Committee (IACUC) protocols and records.<br />

Imaging equipment, such as the PIXImus mouse densitometer and the ACUSON Sequoia 512 ultrasound machine, is available<br />

for noninvasive imaging of mice. VetScan blood chemistry and hematology analyzers are now available for blood analysis.<br />

Also provided by the vivarium technical staff are an extensive xenograft model development and analysis service, rederivation,<br />

surgery, dissection, necropsy, breeding, and health-status monitoring.<br />

Transgenics<br />

22<br />

Fertilized eggs contain two pronuclei, one that is derived from the egg and contains the maternal genetic material and one derived<br />

from the sperm that contains the paternal genetic material. As development proceeds, these two pronuclei fuse, the genetic<br />

material mixes, and the cell proceeds to divide and develop into an embryo. Transgenic mice are produced by injecting small<br />

quantities of foreign DNA (the transgene) into a pronucleus of a one-cell fertilized egg. DNA microinjected into a pronucleus<br />

randomly integrates into the mouse genome and will theoretically be present in every cell of the resulting organism. Expression<br />

of the transgene is controlled by elements called promoters that are genetically engineered into the transgenic DNA. Depending<br />

on the selection of the promoter, the transgene can be expressed in every cell of the mouse or in specific cell populations such as<br />

neurons, skin cells, or blood cells. Temporal expression of the transgene during development can also be controlled by genetic<br />

engineering. These transgenic mice are excellent models for studying the expression and function of the transgene in vivo.<br />

From left: Bondsfield, Eagleson, Jason, Guikema, Shearer, Marinelli, Vitt, Post, Jilbert, Dylewski, Brady,<br />

Schumaker, Rogers, Boguslawski, Grams, DeCamp


VARI | <strong>2007</strong><br />

Kyle A. Furge, Ph.D.<br />

Laboratory of Computational Biology<br />

Dr. Furge received his Ph.D. in biochemistry from the Vanderbilt University School of Medicine in<br />

2000. Prior to obtaining his degree, he worked as a software engineer at YSI, Inc., where he wrote<br />

operating systems for embedded computer devices. Dr. Furge did his postdoctoral work in the<br />

laboratory of George Vande Woude. He became a Bioinformatics Scientist at VARI in June of 2001 and<br />

a <strong>Scientific</strong> Investigator in May of 2005.<br />

23<br />

Staff<br />

Laboratory Staff<br />

Karl Dykema, B.A.


Van Andel Research Institute | <strong>Scientific</strong> <strong>Report</strong><br />

Research Interests<br />

As high-throughput technologies such as DNA sequencing, gene and protein expression profiling, DNA copy number analysis,<br />

and single nucleotide polymorphism genotyping become more available to researchers, extracting the most significant<br />

biological information from the large amount of data produced by these technologies becomes increasingly difficult.<br />

Computational disciplines such as bioinformatics and computational biology have emerged to develop methods that assist<br />

in the storage, distribution, integration, and analysis of these large data sets. The Computational Biology laboratory at VARI<br />

currently focuses on using mathematical and computer science approaches to analyze and integrate complex data sets in order<br />

to develop a better understanding of how cancer cells differ from normal cells at the molecular level. In addition, members of the<br />

lab provide assistance in data analysis and other computational projects on a collaborative and/or fee-for-service basis.<br />

In the past year the laboratory has taken part in many projects to further the research efforts at VARI. We have worked closely<br />

with the Laboratory of Mass Spectrometry and Proteomics in developing computational infrastructure to support new protein<br />

profiling instrumentation and analysis. We have contributed to several gene expression microarray analysis projects ranging<br />

from mechanisms of oncogene transformation to the identification of genes that are associated with drug sensitivity. We also<br />

work closely with the Laboratory of Cancer Genetics in the development of gene expression–based models for diagnosis and<br />

prognosis of renal cell carcinoma. Moreover, we and other groups have demonstrated that several types of biological information,<br />

in addition to relative transcript abundance, can be derived from high-density gene expression profiling data. Taking advantage<br />

of this additional information can lead to the rapid development of plausible computational models of disease development and<br />

progression.<br />

24<br />

Changes in DNA copy number result in dramatic changes in gene expression within the abnormal region and are detectable<br />

through examination of the population of mRNAs generated from the genes that map to each chromosome. Additionally,<br />

activation of certain oncogenes or inactivation of certain tumor suppressor genes can produce context-independent gene<br />

signatures that can be detected in a gene expression profile. For example, genes that are up-regulated by overexpression of<br />

RAS in breast epithelial cells also tend to be overexpressed in other samples containing activated RAS signaling, such as lung<br />

tumors that contain activating RAS mutations. We have invested a reasonable portion of the past several years developing and<br />

evaluating computational methods to predict deregulated signal transduction pathways and chromosomal abnormalities using<br />

gene expression data. We have worked closely with the Laboratory of Cancer Genetics on computational models to describe the<br />

development and progression of renal cell carcinoma. An example of the successful application of this analytic approach is in<br />

the examination of gene expression profiling data derived from papillary renal cell carcinoma (RCC).


VARI | <strong>2007</strong><br />

Computational analysis of gene expression data derived from papillary RCC revealed that a transcriptional signature indicative of<br />

MYC pathway activation was present in high-grade papillary RCC, but not other high-grade RCCs. Predictions of chromosomal<br />

gains and losses were also generated from the gene expression data, and it was demonstrated that the presence of the MYC<br />

signature was coincident with a predicted amplification of chromosome 8q. Because the c-MYC gene maps to chromosome<br />

8q, a computational model was developed such that amplification of chromosome 8q occurs in the high-grade papillary tumors,<br />

which leads to c-MYC overexpression and activation of the MYC pathway. The importance of MYC activation was confirmed<br />

by both pharmacological and siRNA inhibition of active MYC signaling in a cell line model of high-grade papillary RCC. These<br />

results highlight the effectiveness of using gene expression profiling data to build integrative computational models of tumor<br />

development and progression.<br />

25<br />

From left: Dykema, Furge<br />

Recent Publications<br />

Furge, Kyle A., Jindong Chen, Julie Koeman, Pamela Swiatek, Karl Dykema, Kseniji Lucin, Richard Kahnoski, Ximing J. Yang, and<br />

Bin Tean Teh. <strong>2007</strong>. Detection of DNA copy number changes and oncogenic signaling abnormalities from gene expression data<br />

reveals MYC activation in high-grade papillary renal cell carcinoma. Cancer Research 67(7): 3171–3176.<br />

Furge, K.A., M.H. Tan, K. Dykema, E. Kort, W. Stadler, X. Yao, M. Zhou, and B.T. Teh. <strong>2007</strong>. Identification of deregulated oncogenic<br />

pathways in renal cell carcinoma: an integrated oncogenomic approach based on gene expression profiling. Oncogene 26(9):<br />

1346–1350.<br />

Furge, Kyle A., Eric J. Kort, Ximing J. Yang, Walter M. Stadler, Hyung Kim, and Bin Tean Teh. 2006. Gene expression profiling in<br />

kidney cancer: combining differential expression and chromosomal and pathway analyses. Clinical Genitourinary Cancer 5(3):<br />

227–231.<br />

Yang, Ximing J., Jun Sugimura, Kristian T. Schafernak, Maria S. Tretiakova, Misop Han, Nicholas J. Vogelzang, Kyle Furge, and<br />

Bin Tean Teh. 2006. Classification of renal neoplasms based on molecular signatures. Journal of Urology 175(6): 2302–2306.


Van Andel Research Institute | <strong>Scientific</strong> <strong>Report</strong><br />

Brian B. Haab, Ph.D.<br />

Laboratory of Cancer Immunodiagnostics<br />

26<br />

Dr. Haab obtained his Ph.D. in chemistry from the University of California at Berkeley in 1998. He then<br />

served as a postdoctoral fellow in the laboratory of Patrick Brown in the Department of Biochemistry at<br />

Stanford University. Dr. Haab joined VARI as a Special Program Investigator in May 2000 and became a<br />

<strong>Scientific</strong> Investigator in 2004.<br />

Staff<br />

Laboratory Staff<br />

Songming Chen, Ph.D.<br />

Michael Shafer, Ph.D.<br />

Yi-Mi Wu, Ph.D.<br />

Derek Bergsma, B.S.<br />

Sara Forrester, B.S.<br />

Thomas LaRoche, B.S.<br />

Tingting Yue, B.S.<br />

Alex Turner<br />

Students<br />

Krysta Collins<br />

Jennifer Lunger<br />

Devin Mistry<br />

Visiting Scientist<br />

Rasmus Lundquist


VARI | <strong>2007</strong><br />

Research Interests<br />

Members of the Haab laboratory identify protein and carbohydrate abnormalities in the blood of cancer patients and investigate<br />

the significance and potential clinical usefulness of those abnormalities. We develop novel experimental methods to facilitate this<br />

work, and we collaborate with both clinicians and basic scientists to pursue research on pancreatic and prostate cancers.<br />

Low-volume, high-throughput antibody and protein arrays<br />

We have developed the ability to probe multiple proteins or carbohydrate structures using low sample volumes, which provides<br />

a powerful tool for identifying and measuring protein and carbohydrate abnormalities in cancer. Antibody and protein arrays<br />

immobilized on the surface of a microscope slide are the key to such a capability. A biological sample such as blood serum can<br />

be incubated on an array to investigate interactions between the immobilized molecules and the proteins or antibodies in the<br />

sample. Those interactions can be probed to obtain information such as protein abundance, glycosylation level, or protein-protein<br />

interaction level.<br />

The routine use of these tools was made possible by the development of a practical method for processing multiple arrays on<br />

a microscope slide (Fig. 1). A stamp imprints a wax pattern onto the surface of a slide, creating hydrophobic partitions that<br />

segregate various samples. Distinct stamp designs can be used to form differing sizes and numbers of partitions. A design<br />

that imprints 48 arrays on one slide requires only 6 μl of sample per array, with each array composed of 144 distinct spots of<br />

immobilized molecules. Such a design enables the efficient processing of many samples or testing of many conditions in parallel,<br />

as demonstrated in the projects described below. The device for creating these slides is commercially available from The Gel<br />

Company, San Francisco.<br />

27<br />

Figure 1A. Figure 1B. Figure 1C.<br />

Figure 1. High-throughput sample processing using a novel slide partitioning method. A) Wax is imprinted onto a microscope slide to<br />

form borders around multiple arrays. Wax is melted by the hotplate under the bath, and a slide is inserted upside-down into the holder.<br />

Bringing the lever forward raises a stamp out of the wax bath to touch the slide, imprinting the design onto the slide. Two stamps are<br />

shown in front of the machine. B) Loading samples onto a slide containing 48 arrays. The arrays are spaced by 4.5 mm, which is<br />

compatible with the 9 mm spacing of standard multichannel pipettes. C) Samples loaded onto slides containing 12 (top), 48 (middle),<br />

and 192 (bottom) arrays (96 samples loaded).


Van Andel Research Institute | <strong>Scientific</strong> <strong>Report</strong><br />

Glycans in pancreatic cancer<br />

One of the major interests of the lab is characterizing and studying the changes in carbohydrate structures (glycans) on particular<br />

proteins from pancreatic cancer patients. A novel technique developed in our laboratory enables the measurement of specific<br />

glycans on multiple proteins in biological samples (Fig. 2A, B). We use lectins—proteins that bind specific glycan structures—as<br />

well as glycan-binding antibodies to probe the levels of particular glycans on the proteins captured on the antibody arrays.<br />

Several types of lectins, each with its own carbohydrate binding specificity, can be used to identify the carbohydrate structures<br />

associated with each protein. We can analyze many different patient samples or cell culture conditions, looking at associations<br />

between glycan levels and disease states or at the effects of certain perturbations on glycan structures. This method is in<br />

development for commercial use by GenTel Biosciences (Madison, WI).<br />

Mucins are long-chain, heavily glycosylated proteins on epithelial cell surfaces that have roles in cell protection, interaction with<br />

the extracellular space, and regulation of extracellular signaling. Screening studies in collaboration with Randall Brand and Diane<br />

Simeone have revealed a variety of glycan alterations on mucin molecules from pancreatic cancer patients (a representative<br />

example is shown in Fig. 2C). Altered carbohydrates on mucins can affect critical processes in cancer such as cell migration<br />

or extracellular signaling to the immune system. We are characterizing the glycan structural variation on mucins secreted from<br />

cancer cells and other cells, and we are using cell culture systems to study the origins and effects of those variations. We are<br />

pursuing hypotheses about the effects of extracellular stress from an inflammatory tumor environment on mucin carbohydrate<br />

structures and the resulting interactions of those structures with inflammatory proteins and host cells.<br />

Figure 2A.<br />

28<br />

Figure 2B.<br />

Figure 2C.<br />

Figure 2. Complementary antibody array formats for protein and glycan detection. A) Sandwich assay with fluorescence detection<br />

to measure protein abundance. B) Antibody-lectin assay. The biotinylated lectin binds to glycans on the proteins captured by the<br />

immobilized antibodies. The antibodies are first chemically derivatized to prevent lectin binding to the glycans of the immobilized<br />

capture antibodies. C) Detecting protein and glycan variation in cancer and control sera. Sandwich detection of the MUC1 and CEA<br />

proteins showed similar levels in serum samples from a cancer patient and a control subject (left images). The anti-CA19-9 antibody,<br />

which targets a glycan structure, detected a significant glycan increase on MUC1 and CEA in the cancer serum (right images).


VARI | <strong>2007</strong><br />

Cancer biomarkers<br />

Improved methods of detecting and diagnosing cancer could significantly improve outcomes for many patients. We are seeking<br />

to identify and validate protein biomarkers that could form the basis of clinical cancer diagnostics. The antibody-based assays<br />

that we are using are valuable for this work because they are very reproducible, inexpensive, and high-throughput. In addition,<br />

the use of miniaturized arrays of antibodies allows us to efficiently test many antibodies and samples and to rapidly develop new<br />

assays. We are applying these capabilities in novel approaches to biomarker discovery and validation.<br />

Mouse models of cancer may provide a good resource for biomarker discovery because the genetic and experimental variation<br />

between samples can be closely controlled, thus making the identification of abnormal protein levels easier than with human<br />

clinical specimens. Mass spectrometry studies performed by other members of an NCI-sponsored consortium have identified<br />

candidate biomarkers in mouse models of ovarian and pancreatic carcinomas. Using newly generated antibodies that target<br />

those proteins, we are developing assays to determine the levels of these candidate biomarkers in the mouse models and to<br />

assess their diagnostic value for human cancer. Low-volume methods are crucial for these studies because only a small sample<br />

is available from each mouse. These studies could establish a new paradigm for biomarker discovery and validation.<br />

Longitudinal biomarkers<br />

An NCI-sponsored project in our laboratory focuses on the hypothesis that the diagnostic performance of particular biomarkers<br />

can be improved by using measurements collected on multiple occasions (longitudinal measurements) rather than at just a<br />

single point in time. By looking at changes over time, it may be possible to more accurately distinguish abnormal levels in a<br />

given individual, since that person’s normal level could be used as a reference point. In a collaboration with Robert Vessella<br />

and William Catalona, we are investigating this question for the detection of prostate cancer recurrence. By using various<br />

formats of antibody arrays, we can explore different data types and multiple proteins, which we hope will establish the extent of<br />

diagnostic improvement using longitudinal information. Another collaborator, Ziding Feng, is developing the statistical methods<br />

for analyzing the data, which may have value for other applications of this approach.<br />

29<br />

Tumor-reactive antibodies<br />

We and others have investigated measurements of tumor-reactive antibodies as biomarkers. Certain tumor proteins elicit<br />

an antibody-based immune response in a high percentage of cancer patients. In collaboration with Samir Hanash, Gilbert<br />

Omenn, and others, we have further developed the experimental methods for identifying tumor-reactive antibodies using protein<br />

arrays. We are applying this method to the detection of prostate cancer and prostate cancer recurrence. The changes in the<br />

tumor-reactive antibodies are being assessed using the longitudinal approach described above, which may improve the<br />

diagnostic performance of those biomarkers and give insight into the role of immune response in determining the likelihood of<br />

cancer recurrence.<br />

Pancreatic cancer biomarkers<br />

Other biomarker studies in our lab are focused on pancreatic cancer in collaboration with Anna Lokshin, Michael Hollingsworth,<br />

and others in the Early Detection Research Network (EDRN), which is an NCI-sponsored consortium dedicated to discovering<br />

and validating cancer biomarkers. We use the glycan and protein detection technologies described above to identify and study<br />

biomarkers for the early detection or more accurate diagnosis of pancreatic cancer. We have shown that, in certain cases,<br />

the measurement of a glycan on a protein is more accurate for detecting cancer than the measurement of the protein alone in<br />

traditional antibody assays. We are now seeking to define which protein and glycan alterations have the highest diagnostic and<br />

prognostic significance.


Van Andel Research Institute | <strong>Scientific</strong> <strong>Report</strong><br />

External Collaborators<br />

Philip Andrews, University of Michigan, Ann Arbor<br />

Randall Brand, Evanston Northwestern Healthcare, Evanston, Illinois<br />

William Catalona, Northwestern University, Evanston, Illinois<br />

Ziding Feng, Fred Hutchinson Cancer Research Center, Seattle, Washington<br />

Irwin Goldstein, University of Michigan, Ann Arbor<br />

Samir Hanash, Fred Hutchinson Cancer Research Center, Seattle, Washington<br />

Michael A. Hollingsworth, University of Nebraska, Omaha<br />

Anna Lokshin, University of Pittsburgh, Pennsylvania<br />

Gilbert Omenn, University of Michigan, Ann Arbor<br />

Alan Partin, Johns Hopkins University, Baltimore, Maryland<br />

Diane Simeone, University of Michigan, Ann Arbor<br />

Robert Vessella, University of Washington, Seattle<br />

Recent Publications<br />

From left: Forrester, Porter, Nelson, Haab, Bergsma,<br />

Collins, Lundquist, Chen, Yue, Wu, Turner<br />

30<br />

Chen, S., and B.B. Haab. In press. Antibody microarrays for protein and glycan detection. In Clinical Proteomics, Wiley-VCH.<br />

Chen, S., T. LaRoche, D. Hamelinck, D. Bergsma, D. Brenner, D. Simeone, R.E. Brand, and B.B. Haab. In press. Multiplexed<br />

analysis of glycan variation on native proteins captured by antibody microarrays. Nature Methods.<br />

Forrester, S., J. Qiu, L. Mangold, A.W. Partin, D. Misek, B. Phinney, D. Whitten, P. Andrews, E. Diamandis, G.S. Omenn, S. Hanash,<br />

and B.B. Haab. In press. An experimental strategy for quantitative analysis of the humoral immune response to prostate cancer<br />

antigens using natural protein microarrays. Proteomics.<br />

Omenn, Gilbert S., Raji Menon, Marcin Adamski, Thomas Blackwell, Brian B. Haab, and Weimin Gao, and David J. States. <strong>2007</strong>.<br />

The human plasma proteome. In Proteomics of Human Body Fluids: Principles, Methods, and Applications, V. Thongboonkerd,<br />

ed. Totowa, N.J.: Humana Press.<br />

Shafer, Michael W., Leslie Mangold, Alam W. Partin, and Brian B. Haab. <strong>2007</strong>. Antibody array profiling reveals serum TSP-1 as<br />

a marker to distinguish benign from malignant prostatic disease. The Prostate 67: 255–267.<br />

Haab, B.B. 2006. Applications of antibody array platforms. Current Opinion in Biotechnology 17(4): 415–421.<br />

Haab, B.B. 2006. Using array-based competitive and noncompetitive immunoassays. In American Association of Cancer<br />

Research Annual Meeting Education Book, Phildelphia: American Association of Cancer Research.<br />

Haab, Brian B., Amanda G. Paulovich, N. Leigh Anderson, Adam M. Clark, Gregory J. Downing, Henning Hermjakob, Joshua<br />

LaBaer, and Mathias Uhlen. 2006. A reagent resource to identify proteins and peptides of interest for the cancer community: a<br />

workshop report. Molecular & Cellular Proteomics 5(10): 1996–<strong>2007</strong>.<br />

Hung, Kenneth E., Alvin T. Kho, David Sarracino, Larissa Georgeon Richard, Bryan Krastins, Sara Forrester, Brian B. Haab, Isaac<br />

S. Kohane, and Raju Kucherlapati. 2006. Mass spectrometry–based study of the plasma proteome in a mouse intestinal tumor<br />

model. Journal of Proteome Research 5(8): 1866–1878.


VARI | <strong>2007</strong><br />

Rick Hay, Ph.D., M.D., F.A.H.A.<br />

Laboratory of Noninvasive Imaging and Radiation Biology<br />

Dr. Hay earned a Ph.D. in pathology (1977) and an M.D. (1978) at the University of Chicago and the<br />

Pritzker School of Medicine. He became a resident in anatomic pathology and then a postdoctoral<br />

research fellow in the University of Chicago Hospitals and Clinics. Following a postdoctoral fellowship at<br />

the Biocenter/University of Basel (Switzerland), he returned to the University of Chicago as an Assistant<br />

Professor in the Department of Pathology and Associate Director of the Section of Autopsy Pathology<br />

from 1984 to 1992. He moved to the University of Michigan Medical Center in 1992 as a clinical fellow<br />

in the Division of Nuclear Medicine and became Chief Fellow in 1993. From 1994 to 1997 he was a staff<br />

physician, and from 1995 to 1997 the Medical Director in the Department of Nuclear Medicine at St. John<br />

Hospital and Medical Center in Detroit. He joined VARI in 2001 as a Senior <strong>Scientific</strong> Investigator. In<br />

2002 he was named Assistant to the Director for Clinical Programs, and in 2003 was appointed Deputy<br />

Director for Clinical Programs.<br />

31<br />

Staff<br />

Laboratory Staff<br />

Visiting Scientist<br />

Students<br />

Visiting Scientists<br />

Physician-in-training<br />

Troy Giambernardi, Ph.D.<br />

Kim Hardy, M.A., RT(R), RDMS<br />

Yue Guo, B.S.<br />

Joel Strehl, B.S.<br />

Catherine Walker, B.S.<br />

Nigel Crompton, Ph.D., D.Sc.<br />

Matthew Steensma, M.D.<br />

Laboratory Staff<br />

Students<br />

Students<br />

Consultants<br />

Visiting Scientists<br />

Elianna Bootzin<br />

Natalie Kent<br />

Sara Kunz<br />

Jose Toro<br />

Rebecca Trierweiler<br />

Helayne Sherman, M.D., Ph.D., F.A.C.C.<br />

Milton Gross, M.D., F.A.C.N.P.


Van Andel Research Institute | <strong>Scientific</strong> <strong>Report</strong><br />

Research Interests<br />

In July 2005, the Laboratory of Noninvasive Imaging and Radiation Biology originated as an outgrowth and expansion of activities<br />

of the Laboratory of Molecular Oncology. This lab is devoted to both noninvasive imaging (i.e., the generation and analysis of<br />

images or depictions of structure and selected functions in living organisms without surgically or mechanically penetrating a body<br />

cavity) and radiation biology (which involves analysis of the consequences of external and internal radiation exposure in living<br />

organisms).<br />

The lab’s work follows three common themes:<br />

• Development and use of laboratory models that address medical imaging and radiation exposure problems.<br />

• Advancement of technology in imaging and radiation biology, including novel agents, probes, and reporters;<br />

new strategies for tackling research problems; and new instrumentation.<br />

• Pursuit of two-way translation between the laboratory and the clinical setting, i.e., using examples of human<br />

disease to design and improve laboratory model systems for study, as well as moving new discoveries from<br />

the laboratory benchtop to the patient’s bedside.<br />

32<br />

We depend heavily upon access to sophisticated instruments and equipment, including nuclear imaging cameras; planar and<br />

tomographic X-ray units; clinical and research ultrasonography units; fluorescence detection systems; and cell and organism<br />

irradiation capability. Because of the equipment- and expertise-intensive nature of our projects, we could not succeed without<br />

the help of our valued collaborators. During this past year we have acquired two new state-of-the-art noninvasive imaging<br />

instruments: a Vevo 770 high-resolution micro-ultrasound imaging system (VisualSonics) and a nanoSPECT/CT imaging unit<br />

(BioScan), and we have continued to pursue research projects in radiation biology, nuclear medicine, and multimodality imaging.<br />

One established research project continues work begun by Nigel Crompton at the Paul Scherrer Institute in Switzerland, now<br />

performed in collaboration with the radiation oncology service at Saint Mary’s Health Care. This project seeks to predict the<br />

sensitivity of a patient’s normal tissues to irradiation that is being administered for treatment of a serious condition such as<br />

cancer. For this project a sample of the patient’s blood is drawn before radiation therapy. The blood sample is then irradiated<br />

(outside the patient) under precise conditions of exposure, treated with fluorescent molecules that detect certain types of blood<br />

cells (lymphocytes), and then analyzed by fluorescence-activated cell sorting (FACS) for evidence of lymphocyte death. In<br />

Switzerland, Dr. Crompton established a close correlation between lymphocyte death and a patient’s normal tissue tolerance to<br />

irradiation. We are now determining whether western Michigan patients respond similarly, as well as investigating the effects of<br />

patient age, gender, and administered radiation dose on the apoptotic response.


VARI | <strong>2007</strong><br />

A new radiation biology project this year, in collaboration with Weiwen Deng and Aly Mageed of DeVos Children’s Hospital,<br />

investigates a new approach for treating graft-versus-host disease in mice undergoing bone marrow transplantation, with planned<br />

extension to human patients in the near future.<br />

Our major established project in nuclear medicine continues work initiated by Dr. Hay and colleagues while he was a member of<br />

the Laboratory of Molecular Oncology. Since 2001 we have been evaluating radioactive antibodies and smaller molecules that<br />

attach to the Met receptor tyrosine kinase, collectively designated Met-avid radiopharmaceuticals (MARPs). Met plays a key<br />

role in causing cancers to become more aggressive, so that they spread to nearby tissues (invasion) and/or travel through the<br />

bloodstream or lymph channels to distant organs (metastasis). We previously showed that both large and small MARPs are useful<br />

for nuclear imaging of Met-expressing human tumors (xenografts) grown under the skin of immunodeficient mice. During the past<br />

year, in collaboration with our colleagues at VARI and with our outside collaborators at DVAHS, ApoLife, and MSU, we have been<br />

evaluating new ways of complexing radioactive atoms with MARPs for improved ease of use and future clinical applications.<br />

In 2006 we began a multimodality noninvasive imaging program for evaluating the growth, Met expression, and response to<br />

therapy of aggressive human tumor xenografts grown orthotopically in immunodeficient mice. Employing a combination of<br />

high-resolution ultrasound with and without contrast agents, planar and tomographic nuclear imaging, and CT imaging, we are<br />

now acquiring data for tumors of the brain, pancreas, adrenals, and bone.<br />

External Collaborators<br />

33<br />

Our lab depends critically on intramural and extramural collaborations to address our research themes. Our extramural<br />

collaborators include scientists and physicians at the Department of Veterans Affairs Healthcare System in Ann Arbor; the<br />

University of Michigan in Ann Arbor; Michigan State University in East Lansing; ApoLife, Inc., in Detroit; Henry Ford<br />

Hospital in Detroit; West Michigan Heart, P.C., in Grand Rapids; DeVos Children’s Hospital in Grand Rapids; St. Mary’s<br />

Health Care in Grand Rapids; Fred Hutchinson Cancer Research Center in Seattle; the Gerald P. Murphy Foundation in<br />

West Lafayette, Indiana; the National Cancer Institute in Bethesda, Maryland; the University of Illinois in Champaign-Urbana;<br />

and VisualSonics, Inc., in Toronto.<br />

Recent Publications<br />

Meng, L.J., N.H. Clinthorne, S. Skinner, R.V. Hay, and M. Gross. 2006. Design and feasibility study of a single photon emission<br />

microscope system for small animal I-125 imaging. IEEE Transactions on Nuclear Science 53(3): 1168–1178.<br />

Hay, R.V., and M.D. Gross. 2006. Scintigraphic imaging of the adrenals and neuroectodermal tumors. In Nuclear Medicine, 2nd<br />

edition, R.E. Henkin, D. Bova, G.L. Dillehay, S.M. Keresh, J.R. Halama, R.H. Wagner, and A.M. Zimmer, eds. Philadelphia: Mosby<br />

Elsevier, pp. 820–844.


Van Andel Research Institute | <strong>Scientific</strong> <strong>Report</strong><br />

Office of Translational Programs<br />

Since July 2005 the Office of Translational Programs (OTP) has been the administrative home base for activities overseen by the<br />

Deputy Director for Clinical Programs. The role of OTP is to promote and facilitate collaborative programs involving the Van Andel<br />

Research Institute and other institutions in the realm of translational medicine.<br />

OTP accomplishments during our second year of formal operation include the following.<br />

• Serving as the administrative home for the GMP facility. With funding from the state of Michigan and the federal Health<br />

Resources and Services Administration, VARI and Grand Valley State University have partnered to build and operate<br />

Grand River Aseptic Pharmaceutical Packaging (GR-APP), a Good Manufacturing Practices facility that will package<br />

pharmaceuticals for early-phase clinical trials commissioned by academic and commercial investigators, primarily in<br />

Michigan and the Midwest. As of this writing, construction of GR-APP is nearing completion, and we expect operations<br />

to begin by autumn of <strong>2007</strong>.<br />

• Serving as the administrative home for the West Michigan Chapter of the Michigan Cancer Consortium (MCC).<br />

As an active member of the MCC, VARI is committed to participating in statewide programs to reduce the burden of<br />

cancer in Michigan. In 2005, we and other regional MCC members launched an initiative to develop community-based<br />

programs more relevant to western Michigan. Our first project, designated “C-Works!”, will provide cancer screening<br />

and follow-up services to uninsured working women in Kent County.<br />

34<br />

• Organizing and hosting meetings. In October 2006, OTP hosted the Great Lakes Regional Meeting of the American<br />

Cancer Society at VARI (Troy Giambernardi, Conference Chair) and assisted with preparations for the fall meeting<br />

of the Central Chapter-Society of Nuclear Medicine in Traverse City (Rick Hay, Conference Co-Chair). In November<br />

2006, OTP assisted with local arrangements for the annual meeting of the Michigan Cancer Consortium at DeVos Hall.<br />

• Promoting new interinstitutional collaborations and providing resources for funding proposals. OTP provides a broad<br />

range of administrative assistance, logistical support, grant preparation expertise, meeting venues, and seed funding<br />

for new interinstitutional collaborations seeking extramural funding from state, federal, or private sources. During this<br />

past year we helped secure state funding for ClinXus, a west Michigan–based consortium for conducting innovative<br />

clinical trials, and we are awaiting the outcomes of recent collaborative proposals submitted to NIH and to two private<br />

foundations.<br />

• Coordinating research rotations for physicians-in-training. In collaboration with the Grand Rapids Medical Education<br />

and Research Consortium (MERC), we schedule each first-year general surgery resident to spend one month working<br />

in a designated research laboratory at VARI. This program has been well received by both residents and VARI<br />

investigators. Custom-tailored rotations of variable duration at VARI can be arranged for other physicians-in-training.<br />

Staff<br />

Rick Hay, Ph.D., M.D., F.A.H.A.<br />

Troy Carrigan<br />

Jean Chastain


VARI | <strong>2007</strong><br />

Jeffrey P. MacKeigan, Ph.D.<br />

Laboratory of Systems Biology<br />

Dr. MacKeigan received his Ph.D. in microbiology and immunology at the University of North Carolina<br />

Lineberger Comprehensive Cancer Center in 2002. He then served as a postdoctoral fellow in the<br />

laboratory of John Blenis in the Department of Cell Biology at Harvard Medical School. In 2004, he joined<br />

Novartis Institutes for Biomedical Research in Cambridge, Massachusetts, as an investigator and project<br />

leader in the Molecular and Developmental Pathways expertise platform. Dr. MacKeigan joined VARI in<br />

June 2006 as a <strong>Scientific</strong> Investigator.<br />

35<br />

Staff<br />

Students<br />

Laboratory Staff Students Visiting Scientists<br />

Brendan Looyenga, Ph.D.<br />

Christina Ludema, B.S.<br />

Natalie Wolters, B.S.<br />

Katie Sian, B.S.<br />

Geoff Kraker


Van Andel Research Institute | <strong>Scientific</strong> <strong>Report</strong><br />

Research Interests<br />

The primary focus of the Systems Biology laboratory is identifying and understanding the genes and signaling pathways that<br />

when mutated contribute to the pathophysiology of cancer. We take advantage of RNA interference (RNAi) and novel proteomic<br />

approaches to identify the enzymes that control cell growth, cell proliferation, and cell survival. For example, after screening the<br />

human genome for more than 600 kinases and 200 phosphatases—called the “kinome” and “phosphatome”, respectively—that<br />

act with chemotherapeutic agents in controlling apoptosis, we identified 73 kinases and 72 phosphatases whose roles in cell<br />

survival were previously unrecognized. We are asking several questions. How are these novel survival enzymes regulated at the<br />

molecular level? What signaling pathway(s) do they regulate? Does changing the number of enzyme molecules present inhibit<br />

waves of compensatory changes at the cellular level (system-level changes)? What are the system-level changes after reduction<br />

or loss of each gene?<br />

Identification of kinases that regulate cell survival<br />

36<br />

We have performed RNAi screens in the presence of apoptosis-inducing chemotherapeutic agents (Taxol, cisplatin, and etoposide)<br />

and identified a group of kinases whose loss of function sensitizes cells to undergo cell death, the most interesting of these being<br />

PINK1 (PTEN-induced kinase 1). PINK1 was originally shown to be up-regulated by the tumor suppressor PTEN. Although<br />

PINK1 does not fall into a particular kinase subfamily, it has a known role in maintaining mitochondrial membrane potential. Other<br />

work has recently shown inherited mutations at chromosomal location 1p36 in familial Parkinson disease, and the two mutated<br />

genes that map to this region are PINK1 (PARK6) and DJ-1 (PARK7). Both genes are responsible for early-onset autosomal<br />

recessive parkinsonism. We had previously noted that DJ-1 is overexpressed in non–small cell lung carcinoma and that<br />

its down-regulation enhances apoptosis. Further, Parkinson disease–causing mutations in LRRK2 (PARK8), which are dominantly<br />

inherited gain-of-function mutations, sensitize neurons to cell death, and a significant fraction of the LRRK2 population is<br />

associated with the mitochondria. We are currently investigating whether the molecular mechanisms of PINK1 and LRRK2 in<br />

cancer and in Parkinson disease are linked.


VARI | <strong>2007</strong><br />

Identification of phosphatases that regulate chemoresistance<br />

Our research has shown that a large percentage of phosphatases and their regulatory subunits contribute to cell survival. This<br />

is a previously unrecognized general role for phosphatases as negative regulators of apoptosis, and it is important because<br />

phosphatases may no longer be simply viewed as enzymes that oppose the action of kinases. This research also identified<br />

a number of phosphatases whose loss of function results in chemoresistance, implicating these proteins as potential tumor<br />

suppressors. In our RNAi study, 5% of all phosphatases were shown to act in this way; an example is MK-STYX. Down-regulation<br />

of MK-STYX resulted in dramatic cellular resistance to cisplatin-, Taxol- or etoposide-induced cell death, which is consistent<br />

with up-regulated survival signals in these cells (Fig. 1). Also, MK-STYX is located at 7q11.23, a chromosome region mutated<br />

in colon cancer. MK-STYX is similar to MKP-1, which inactivates MAPKs; MK-STYX, however, is predicted to be a catalytically<br />

inactive phosphatase. Our observations suggest that MK-STYX acts against cell survival by sequestering pro-survival signaling<br />

components in a way analogous to the “substrate-trapping” effects of catalytically inactive phosphatases.<br />

Figure 1A.<br />

Figure 1B.<br />

37<br />

Figure 1. Identification of MK-STYX as a potential tumor suppressor<br />

phosphatase. Cells were transfected with control siRNA or<br />

MK-STYX siRNA for 48 h and then were treated for an additional<br />

24 h with solvent control (–) or 50 μM cisplatin (+). Cell viability was<br />

visualized by A) crystal violet stain and B) cleavage of full-length<br />

PARP measured by western blot analysis.


Van Andel Research Institute | <strong>Scientific</strong> <strong>Report</strong><br />

Graded MAPK signaling and switch-like c-Fos induction<br />

We also take a systems biology approach to understanding two key molecular pathways, Ras/MAPK and PI3K/mTOR.<br />

In the Ras/MAPK pathway, growth factors activate the small G protein Ras, which recruits Raf to the plasma membrane where<br />

it is activated and phosphorylates MEK1/2, which in turn phosphorylates ERK1/2-MAPKs. Activated ERK1/2 phosphorylates<br />

additional kinases (such as RSK) and specific transcription factors (such as c-Fos and Elk-1) that are important in cellular<br />

proliferation, differentiation, and survival.<br />

One project in the lab involved the question of whether the evolutionarily conserved MAPK pathway exhibits a switch-like or a<br />

graded response in mammalian cells. Ultrasensitive switch-like responses control cell-fate decisions in many biological settings,<br />

and the regulation of kinase activity is one way in which such behavior can be initiated. Signaling molecules switch between<br />

two discontinuous, stable states with no intermediate; this is referred to as a bistable response (Fig. 2, top panel). Given the<br />

irreversible, all-or-none nature of many cell behaviors, including cell cycle control and apoptosis, significant effort has been<br />

focused on identifying the cellular mechanisms underlying bistability. Our research and that of others has provided solid evidence<br />

for graded MAPK signaling in mammalian cells (Fig. 2, lower panel); that is, as agonist concentration increases, single-cell kinase<br />

activity increases proportionally. Yet we have also found that the proliferative response to growth factor stimulation is switch-like,<br />

demonstrating that the ultrasensitive step in the MAPK pathway occurs at the level of MAPK nuclear concentration and switch-like<br />

c-Fos induction. Although c-Fos induction and cell cycle entry in mammalian cells is switch-like, graded MAPK activation could<br />

have an important role in cell survival, since many MAPK targets regulating cell survival are in the cytoplasm.<br />

38<br />

Figure 2.<br />

Figure 2. Total cell population MAPK measurements.<br />

Single cells exhibiting a bistable (all-or-none) response or graded<br />

response (linear).<br />

From left: Wolters, Ludema, Kraker, Looyenga, MacKeigan, Nelson, Sian


VARI | <strong>2007</strong><br />

Cindy K. Miranti, Ph.D.<br />

Laboratory of Integrin Signaling and Tumorigenesis<br />

Dr. Miranti received her M.S. in microbiology from Colorado State University in 1982 and her Ph.D. in<br />

biochemistry from Harvard Medical School in 1995. She was a postdoctoral fellow in the laboratory<br />

of Joan Brugge at ARIAD Pharmaceuticals, Cambridge, Massachusetts, from 1995 to 1997 and in the<br />

Department of Cell Biology at Harvard Medical School from 1997 to 2000. Dr. Miranti joined VARI as a<br />

<strong>Scientific</strong> Investigator in January 2000. She is also an Adjunct Assistant Professor in the Department of<br />

Physiology at Michigan State University.<br />

39<br />

Staff<br />

Laboratory Staff<br />

Mathew Edick, Ph.D.<br />

Suganthi Sridhar, Ph.D.<br />

Kristin Saari, M.S.<br />

Lia Tesfay, M.S.<br />

Laura Lamb, B.S.<br />

Veronique Schulz, B.S.<br />

Susan Spotts, B.S.<br />

Students<br />

Students<br />

Eric Graf<br />

Gary Rajah<br />

Visiting Scientists


Van Andel Research Institute | <strong>Scientific</strong> <strong>Report</strong><br />

Research Interests<br />

Our laboratory is interested in the mechanisms by which integrin receptors, interacting with the extracellular matrix (ECM), regulate<br />

cell processes involved in the development and progression of cancer. Using tissue culture models, biochemistry, molecular<br />

genetics, and mouse models, we are defining the cellular and molecular events involved in integrin-dependent adhesion and<br />

downstream signaling that are important for prostate tumorigenesis and metastasis.<br />

Integrins are transmembrane proteins that serve as receptors for ECM proteins. By interacting with the ECM, integrins stimulate<br />

intracellular signaling transduction pathways to regulate cell shape, proliferation, migration, survival, gene expression, and<br />

differentiation. Integrins do not act autonomously, but “crosstalk” with receptor tyrosine kinases (RTKs) to regulate many of these<br />

cellular processes. Studies in our lab indicate that integrin-mediated adhesion to ECM proteins activates epidermal growth factor<br />

receptors EGFR/ErbB2 and the HGF/SF receptor c-Met. Integrin-mediated activation of these RTKs is ligand-independent and<br />

required for the activation of a subset of intracellular signaling molecules in response to cell adhesion.<br />

The prostate gland and cancer<br />

40<br />

Tumors that develop from cells of epithelial origin, i.e., carcinomas, represent the largest tumor burden in the United States.<br />

Prostate cancer is the most frequently diagnosed cancer in American men and the second leading cause of cancer death in<br />

men. Patients who at the time of diagnosis have androgen-dependent and organ-confined prostate cancer are relatively easy<br />

to cure through radical prostatectomy or localized radiotherapy, but patients with aggressive and metastatic disease have fewer<br />

options. Androgen ablation can significantly reduce the tumor burden in the latter patients, but the potential for relapse and the<br />

development of androgen-independent cancer is high. Currently there are no effective treatments for patients who reach this<br />

stage of disease.<br />

In the human prostate gland, α3β1 and α6β4 integrins on epithelial cells bind to the ECM protein laminin 5 in the basement<br />

membrane. In tumor cells, however, the α3 and β4 integrin subunits disappear—as does laminin 5—and the tumor cells express<br />

primarily α6β1 and adhere to a basement membrane containing laminin 10. There is also an increase in expression of the RTKs<br />

EGFR and c-Met in the tumor cells. Two fundamental questions are whether the changes in integrin and matrix interactions that<br />

occur in tumor cells are required for or help to drive the survival of tumor cells, and whether crosstalk with RTKs is important.


VARI | <strong>2007</strong><br />

Integrins and RTKs in prostate epithelial cell survival<br />

How integrin engagement of various ECMs regulates survival pathways in normal and tumor cells is poorly understood.<br />

We recently initiated studies to determine how adhesion to matrix regulates cell survival in normal epithelial cells. We<br />

have shown that integrin-induced activation of EGFR in normal primary prostate epithelial cells is required for survival on their<br />

endogenous matrix, laminin 5. The ability of EGFR to support integrin-mediated cell survival on laminin 5 is mediated through<br />

α3β1 integrin and requires signaling downstream to Erk. Surprisingly, we found that the death induced by inhibition of EGFR<br />

in normal primary prostate cells is not mediated through or dependent on classical caspase-mediated apoptosis. The presence<br />

of an autophagic survival pathway (Fig. 1), regulated by adhesion to matrix, prevents the induction of caspases when EGFR is<br />

inhibited. Suppression of autophagy is sufficient to induce caspase activation and apoptosis in laminin 5–adherent primary<br />

prostate epithelial cells. Thus, adhesion of normal cells to matrix regulates survival through at least two mechanisms, crosstalk<br />

with EGFR and Erk and the maintenance of an autophagic survival pathway (Fig. 2).<br />

Figure 1.<br />

Figure 2.<br />

Normal<br />

Autophagic<br />

41<br />

Figure 1. Induction of autophagy in primary prostate epithelial cells<br />

as shown by punctate staining of the autophagic LC3 protein using<br />

fluorescence microscopy.<br />

Figure 2. Laminin-mediated survival pathways in primary prostate<br />

epithelial cells.<br />

Interestingly, both of these pathways are absent in at least one metastatic prostate cancer cell line, PC3. Accordingly,<br />

integrin-mediated survival of PC3 cells does not depend on EGFR or Erk, but is instead dependent on PI-3K. The PI-3K pathway is<br />

inhibitory to autophagy. We are currently testing additional prostate tumor cells lines to determine if this switch in matrix-mediated<br />

survival pathways is found in all prostate cancers.<br />

Our next step is to determine how integrins regulate survival through autophagy. Since loss of autophagy results in activation<br />

of caspases and classical apoptosis, we have been searching for signaling pathways whose inhibition also results in caspase<br />

activation. We have tentatively identified two important molecules, the RTK c-Met and the anti-apoptotic protein Bcl-XL. Inhibition<br />

of either molecule leads to caspase-induced cell death, indicating that they may be involved in regulating integrin-mediated<br />

autophagy. Future studies in our lab will be aimed at deciphering this pathway.


Van Andel Research Institute | <strong>Scientific</strong> <strong>Report</strong><br />

The androgen receptor in integrin-mediated survival<br />

All primary and metastatic prostate cancers express the intracellular steroid receptor for androgen, AR. In the normal gland, the<br />

AR-expressing cells do not interact with the ECM in the basement membrane; however, all AR-expressing tumor cells do adhere<br />

to the ECM in the basement membrane. In normal cells, AR expression suppresses growth and promotes differentiation, but in<br />

tumor cells AR expression promotes cell growth and is required for cell survival. The mechanisms that lead to the switch from<br />

growth inhibition and differentiation to growth promotion and survival are unknown. Our hypothesis is that adhesion to the ECM<br />

by the tumor cells is responsible for driving the switch in AR function.<br />

When prostate tumor cells are placed in culture, they lose expression of AR. The reason for this is not clear, but it may have to<br />

do with loss of the appropriate ECM-containing basement membrane. When we introduce AR into prostate tumor cells, it actually<br />

suppresses their growth and induces cell death. However, if we place the AR-expressing tumor cells on laminin (the ECM found<br />

in tumors), these cells no longer die. The mechanisms responsible for this change in survival are unknown. Preliminary studies<br />

indicate that there are changes in integrin expression upon expression of AR that may enhance specific signaling pathways<br />

when those integrins bind to matrix. We are currently determining which cell survival pathways are activated by AR upon integrin<br />

engagement.<br />

CD82 and integrin signaling in prostate cancer metastasis<br />

42<br />

Death from prostate cancer is due to the development of metastatic disease, which is difficult to control and occurs by mechanisms<br />

that are not understood. One approach we are taking is to characterize genes that are specifically associated with metastatic<br />

prostate cancer. CD82/KAI1 is a metastasis suppressor gene whose expression is specifically lost in metastatic cancer but not<br />

in primary tumors. Interestingly, CD82/KAI1 is known to associate with both integrins and RTKs. Our goal has been to determine<br />

how loss of CD82/KAI1 expression promotes metastasis.<br />

We have found that reexpression of CD82/KAI1 in metastatic tumor cells suppresses laminin-specific migration and invasion<br />

via suppression of both integrin- and ligand-induced activation of the RTK c-Met. Interestingly, c-Met is often overexpressed<br />

in metastatic prostate cancer. Thus, CD82/KAI1 normally acts to regulate signaling through c-Met such that upon CD82 loss in<br />

tumor cells, signaling through c-Met is increased, leading to increased invasion. We are currently determining the mechanism<br />

by which CD82/KAI1 down-regulates c-Met signaling. Our studies indicate that c-Met and CD82 do not directly interact, and<br />

CD82 may act to suppress c-Met signaling indirectly by dispersing c-Met aggregates present on metastatic tumor cells. We<br />

have developed mutants of CD82 in order to determine which part of the CD82 molecule is required for suppression of<br />

c-Met activity. Also, we have determined that reexpression of CD82 in tumor cells induces a physical association between CD82<br />

and a related family member, CD9. We are determining whether this association is important for suppressing c-Met activity.<br />

We have also initiated mouse studies to demonstrate the importance of CD82 in regulating metastasis in vivo. Using orthotopic<br />

injection of wild-type or CD82-expressing metastatic prostate tumor cells directly into the prostate, we found that CD82 also<br />

suppresses metastasis in vivo. We are continuing these studies to determine if CD82’s ability to specifically affect c-Met is<br />

responsible for metastasis suppression. In addition, we are generating mice in which CD82 expression is specifically lost in<br />

the epithelial cells of the prostate gland. This approach will allow us to determine if CD82 is important for the normal biology of<br />

prostate epithelial cells in vivo. Furthermore, we will be able to determine if loss of CD82 in the mouse prostate gland will lead to<br />

an increased ability to produce metastatic prostate cancer.<br />

.


VARI | <strong>2007</strong><br />

External Collaborators<br />

Beatrice Knudsen, Fred Hutchinson Cancer Research Center, Seattle, Washington<br />

Senthil Muthuswamy, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York<br />

Ilan Tsarfaty, Tel Aviv University, Israel<br />

Valera Vasioukin, Fred Hutchinson Cancer Research Center, Seattle, Washington<br />

Xin Zhang, University of Tennessee, Memphis<br />

43<br />

From left: Lamb, Saari, Spotts, Rajah, Tesfay, Graf, Schulz, Miranti<br />

Recent Publications<br />

Edick, M.J., Tesfay, L., Lamb, L.E., Knudsen, B.S., and Miranti, C.K. In press. Inhibition of integrin-mediated crosstalk with<br />

EGFR/Erk or Src signaling pathways in autophagic prostate epithelial cells induces caspase-independent death. Molecular<br />

Biology of the Cell.<br />

Sridhar, S.C., and C.K. Miranti. In press. Tumor metastasis suppressor KAI1/CD82 is a tetraspanin. In Contemporary Cancer<br />

Research: Metastasis, C. Rinker-Schaeffer, M. Sokoloff, and D. Yamada, eds.<br />

Wang, X. , J. Zhu, P. Zhao, Y. Jiao, N. Xu, T. Grabinski, C. Liu, C.K. Miranti, T. Fu, and B. Cao. In press. In vitro efficacy of immunochemotherapy<br />

with anti-EGFR human Fab-Taxol conjugate on A431 epidermoid carcinoma cells. Cancer Biology & Therapy.<br />

Knudsen, Beatrice S., and Cindy K. Miranti. 2006. Impact of cell adhesion changes on proliferation and survival during prostate<br />

cancer development and progression. Journal of Cellular Biochemistry 99(2): 345–361.


Van Andel Research Institute | <strong>Scientific</strong> <strong>Report</strong><br />

44<br />

Image courtesy of Qian Xie, Yue Guo, Rick Hay, and<br />

George Vande Woude, Van Andel Research Institute;<br />

Helayne Sherman, West Michigan Heart, P.C.; and Ai Lockard, VisualSonics, Inc.


VARI | <strong>2007</strong><br />

Evaluating the blood supply of cancer with ultrasound.<br />

45<br />

Human glioblastoma (brain cancer) cells were grown as a tumor beneath the skin of a laboratory mouse. Small bubbles about the size of individual blood<br />

cells were then injected into the mouse’s bloodstream. During the next few minutes the tumor was imaged by ultrasound, using a device similar to sonar.<br />

Echoes from the bubbles, shown in green, depict complex branching patterns of tiny blood vessels growing around and within the tumor to supply it with<br />

nutrients and oxygen. We are using the ultrasound technique to monitor how new types of anticancer medicine change the abundance and branching<br />

of tumor blood vessels in mice, with the hope of applying this technology in the near future to patients with cancer.


Van Andel Research Institute | <strong>Scientific</strong> <strong>Report</strong><br />

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

Division of Quantitative Sciences<br />

Laboratory of Analytical, Cellular, and Molecular Microscopy<br />

Laboratory of Microarray Technology<br />

Laboratory of Molecular Epidemiology<br />

46<br />

Dr. Resau received his Ph.D. from the University of Maryland School of Medicine in 1985. He has<br />

been involved in clinical and basic science imaging and pathology-related research since 1972.<br />

Between 1968 and 1994, he was in the U.S. Army (active duty and reserve assignments) and served in<br />

Vietnam. From 1985 until 1992, Dr. Resau was a tenured faculty member at the University of Maryland<br />

School of Medicine, Department of Pathology. Dr. Resau was the Director of the Analytical, Cellular and<br />

Molecular Microscopy Laboratory in the Advanced BioScience Laboratories–Basic Research Program<br />

at the National Cancer Institute, Frederick Cancer Research and Development Center, Maryland, from<br />

1992 to 1999. He joined VARI as a Special Program Senior <strong>Scientific</strong> Investigator in June 1999 and<br />

in 2003 was promoted to Deputy Director. In 2004, Dr. Resau assumed as well the direction of the<br />

Laboratory of Microarray Technology to consolidate the imaging and quantification of clinical samples<br />

in a CLIA-type research laboratory program. In 2005, Dr. Resau was made the Division Director of the<br />

quantitative laboratories (pathology-histology, microarray, proteomics, epidemiology, and bioinformatics),<br />

and in 2006 he was promoted to Distinguished <strong>Scientific</strong> Investigator.<br />

Staff<br />

Laboratory Staff<br />

Eric Kort, M.D.<br />

Brendan Looyenga, Ph.D.<br />

Bree Berghuis, B.S., HTL<br />

(ASCP), QIHC<br />

Eric Hudson, B.S.<br />

Paul Norton, B.S.<br />

Ken Olinger, B.S.<br />

David Satterthwaite, B.S.<br />

Kristin VandenBeldt, B.S.<br />

JC Goolsby<br />

Students<br />

Pete Haak, B.S.<br />

Alicia Coleman<br />

Kate Jackson<br />

Wei Luo, B.A.<br />

Nick Miltgen<br />

Kara Myslivec<br />

Sara Ramirez<br />

Jourdan Stuart<br />

Mohan Thapa, M.S.<br />

Huong Tran<br />

Grant Van Eerden<br />

Visiting Scientist<br />

Yair Andegeko


VARI | <strong>2007</strong><br />

Research Interests<br />

The Division of Quantitative Sciences includes the laboratories of Analytical, Cellular, and Molecular Microscopy (ACMM), the<br />

Laboratory of Microarray Technology (LMT), the Laboratory of Computational Biology, the Laboratory of Molecular Epidemiology,<br />

and the Laboratory of Mass Spectrometry and Proteomics. The Division’s laboratories use objective measures to define pathophysiologic<br />

events and processes. For example, the LMT measures the expression of genes relative to a control or a standard.<br />

When pathology and tissue organization is combined with expression, one can better determine not only what the change is but<br />

also possible causation, treatment targets, and effects of treatment. The Molecular Epidemiology laboratory builds objective data<br />

and pathology correlations to infer causation and prognosis.<br />

The ACMM laboratory has programs in pathology, histology, and imaging to describe and visualize changes in cell, tissue, or<br />

organ structure. Our imaging instruments allow us to visualize cells and their components with striking clarity, allowing researchers<br />

to determine where in a cell specific molecules are located. We also use a laser for microdissection of cells from a sample. The<br />

laboratory provides paraffin-block (SPIN program) and frozen-section (TAS program) staining of tissues. An archive of pathology<br />

tissues in the paraffin blocks (Van Andel Tissue Repository; VATR) is being accumulated with the cooperation of local hospitals,<br />

and the data on the samples is being converted to computerized files. The lab also carries out research that will improve our<br />

ability to quantify images, so that we will be able to not only state that a particular protein is present in an image, but also answer<br />

the questions of how much is there and with what other molecules is it co-localized? We are able to image using either fluorescent<br />

(e.g., FITC, GFP) or chromatic agents (e.g., DAB, H&E) and separate the components using our confocal, Nuance, or Maestro<br />

instruments.<br />

47<br />

The Laboratory of Microarray Technology provides gene expression analysis using cDNA microarrays. High-throughput robotics<br />

are used to maintain and process cDNA clone sets for the human, mouse, rat, and canine genomes. The clones are used to<br />

produce both cDNA and spotted oligonucleotide microarrays that are evaluated using strict quality control and quality assurance<br />

criteria developed using the Clinical Laboratory Improvement Amendments (CLIA) as a model. These criteria allow the<br />

laboratory to function in a manner consistent with fully accredited clinical laboratories. In 2006 we produced and used 790<br />

cDNA microarrays, and we also produced 112 custom protein microarrays. In addition, the laboratory has expanded its services<br />

to include Agilent and Operon commercial oligonucleotide microarrays. The use of these products will remove much of the<br />

internal quality control and quality assurance burden, and they will also facilitate the requirement to perform array comparative<br />

genomic hybridization, chromatin immunoprecipitation (chip-on-chip), and splice variant analysis.<br />

Hauenstein Parkinson’s Center<br />

Throughout 2006 we have continued our collaboration with the Hauenstein Parkinson’s Center to collect patient blood samples<br />

and controls from 114 individuals. Mutations in the parkin gene in a series of families with more than one generation affected by<br />

Parkinson disease are being investigated by DNA sequence analysis and will be correlated to gene expression data obtained<br />

from microarray analysis.


Van Andel Research Institute | <strong>Scientific</strong> <strong>Report</strong><br />

Blood spot arrays<br />

State laws in the U.S.A. mandate that blood be drawn from all newborn infants to screen for a variety of health-threatening<br />

conditions. The assays consume only a small portion of the blood samples, which are collected on filter paper (“Guthrie”) cards.<br />

Many states archive the leftover cards, often in unrefrigerated storage. Pete Haak and Eric Kort have successfully isolated<br />

mRNA from archived unfrozen neonatal blood spots obtained as long as nine years ago. Using both quantitative RT-PCR and<br />

multiplex gene expression analysis with cDNA arrays, we can detect RNA from hundreds to thousands of genes in these samples.<br />

Furthermore, we have shown through use of freshly spotted blood cards that the genes detected approximate those found in<br />

whole blood and purified buffy coat. These preliminary experiments demonstrate the feasibility of detecting and identifying RNA<br />

amplified from unfrozen stored neonatal blood spots. The application of high-throughput assays to the analysis of these widely<br />

available samples may be a valuable resource for the study of perinatal markers and determinants of subsequent disease<br />

development. The coming year will see this technology applied to the study of cerebral palsy and neuroblastoma.<br />

Mouse models of Parkinson disease<br />

48<br />

As part of the VAI initiative into Parkinson disease, we have begun to generate novel rodent models of dopaminergic cell loss in<br />

the brain in collaboration with Bart Williams. One of the key tools for these studies is the transgenic dopamine-transporter/cre<br />

(DAT-cre) mouse line, which specifically expresses the cre recombinase in dopaminergic neurons of the brain. In combination<br />

with other transgenic and knock-out mouse lines, the DAT-cre mice will allow us to address the response of such neurons to toxic<br />

stimuli in the context of specific gene deletions and additions. Several of the ongoing and future projects based on the DAT-cre<br />

mouse model are briefly described below.<br />

• Imaging and isolation of primary dopaminergic neurons from mouse brain. We have performed a genetic cross<br />

between the DAT-cre strain and ROSA26 reporter strain to generate mice that specifically express the LacZ reporter<br />

gene in dopaminergic neurons. The DAT-cre/ROSA26 mice will permit us to visualize and quantify live dopaminergic<br />

neurons in vivo. With these mice we will assess the effect of cytotoxic agents (e.g., MMTP, rotenone, or<br />

6-hydroxydopamine) on the number of dopaminergic cells, and more importantly, assess the ability of mice to recover<br />

from these insults. These studies will provide insight into the regenerative capacity of the brain when dopaminergic<br />

neurons are lost or injured. The DAT-cre/ROSA26 mice will also provide a source of highly pure dopaminergic neurons<br />

for in vitro studies. Dopaminergic neurons from these mice will be isolated from brain tissue treated with DDAOgalactoside<br />

and will be identified from the cellular population by fluorescence-activated cell sorting in the VAI flow<br />

cytometry core facility.<br />

• Dopaminergic cell regeneration as a function of age. The relationship between age and the likelihood of developing<br />

Parkinson disease is well established, though the causal nature of this relationship is unclear. One hypothesis is that the<br />

capacity of the brain to regenerate damaged neurons decreases with age, consistent with a gradual loss of brain stem<br />

cells that give rise to new dopaminergic neurons. To test this hypothesis in a mammalian system, we are planning a<br />

genetic cross between DAT-cre and pu TK mice, the latter specifically expressing herpes simplex virus thymidine<br />

kinase (hsvTK) in cells that contain cre recombinase. Cells expressing hsvTK are sensitive to the antiviral compound<br />

ganciglovir (G418) and undergo programmed cell death after systemic treatment. Using the DAT-cre/pu TK model,<br />

we will eliminate dopaminergic neurons at various ages (3, 6, 9, and 12 months) and assess the regenerative potential<br />

of these mice using behavioral and histological parameters. These studies will indicate both the absolute and relative<br />

capacities of the mammalian brain to regenerate dopaminergic neurons as a function of age, thereby providing<br />

information about the value of therapies intended to stimulate the endogenous regenerative capacity of the brain in<br />

Parkinson disease patients.


VARI | <strong>2007</strong><br />

• Effect of hypoxia-inducible factor signaling on dopaminergic cell survival. Dopaminergic neurons are exquisitely<br />

sensitive to oxidative stress, which is defined by an increase in toxic reactive oxygen species. Reactive oxygen<br />

species lead to cell death by direct mechanisms, such as damage to important cellular biomolecules, and indirect<br />

ones, such as the induction of cell death pathways. The latter effect may be offset by cell survival pathways, which<br />

increase thethreshold signal intensity required to induce cell death. Because both chemically induced and idiopathic<br />

Parkinson disease are characterized by increased oxidative stress in dopaminergic neurons, therapies that increase<br />

cell survival pathways in these neurons may be broadly applicable as a treatment to decrease cell death in patients.<br />

The PI-3-kinase (PI3K)/Akt pathway is a highly conserved cell survival pathway operating in virtually all mammalian cell types.<br />

This pathway is tightly regulated by the phosphatase PTEN, which directly opposes the kinase activity of PI3K. We have crossed<br />

DAT-cre mice to mice with a conditionally inactivated allele for PTEN (PTEN flox/flox ). Expression of the cre recombinase in these<br />

mice leads to a genetic deletion of PTEN, thereby increasing Akt activity. DAT-cre/PTEN flox/flox mice and their wild-type littermates<br />

will be treated with the neurotoxin MPTP, which induces high levels of oxidative stress in dopaminergic neurons. We will compare<br />

the mice using behavioral and histological parameters to determine whether increased Akt activity leads to greater cell survival<br />

after an oxidative stress insult.<br />

Educational highlights<br />

This year we had one student from GRAPCEP, two students from the MSU-CVM program, and a guest student from Bath University<br />

in the United Kingdom. Our GRAPCEP mentorship program continues to be funded by Pfizer for a seventh year. Dr. Resau is<br />

a member of the graduate school committee that established the VAEI Graduate School, which will increase our research and<br />

educational opportunities.<br />

49<br />

From left, back row: Goolsby, Satterthwaite, Norton, Resau, Haak, Hudson;<br />

front row: Kort, Luo, VandenBeldt, Berghuis, Jason, Ramirez, Looyenga


Van Andel Research Institute | <strong>Scientific</strong> <strong>Report</strong><br />

Recent Publications<br />

Baldus, S.E., E.J. Kort, P. Schirmacher, H.P. Dienes, and J.H. Resau. In press. Quantification of MET and hepatocyte growth<br />

factor/scatter factor expression in colorectal adenomas, carcinomas, and non-neoplastic epithelia by quantitative laser scanning<br />

microscopy. International Journal of Oncology.<br />

Kort, E.J., M.R. Moore, E.A. Hudson, B. Leeser, G.M. Yeruhalmi, R. Leibowitz-Amit, G. Tsarfaty, I. Tsarfaty, S. Moshkovitz, and J.H.<br />

Resau. In press. Use of organ explant and cell culture. In Mechanisms of Carcinogenesis, Hans Kaiser, ed. Dordrecht, The<br />

Netherlands: Kluwer Academic.<br />

Lindemann, K.K., J. Resau, J. Nährig, E. Kort, B. Leeser, K. Anneke, A. Welk, J. Schäfer, G.F. Vande Woude, E. Lengyel, and N.<br />

Harbeck. In press. Differential expression of c-Met, its ligand HGF/SF, and HER2/neu in DCIS and adjacent normal breast tissue.<br />

Histopathology.<br />

Whitwam, T., M.W. VanBrocklin, M.E. Russo, P.T. Haak, D. Bilgili, J.H. Resau, H.-M. Koo, and S.L. Holmen. In press. Differential<br />

oncogenic potential of activated RAS isoforms in melanocytes. Oncogene.<br />

Young, J.J., J.L. Bromberg-White, C.R. Zylstra, J. Church, E. Boguslawski, J. Resau, B.O. Williams, and N. Duesbery. In press. LRP5<br />

and LRP6 are not required for protective antigen–mediated internalization or lethality of anthrax lethal toxin. PLoS Pathogen.<br />

50<br />

Bruxvoort, Katia J., Holli M. Charbonneau, Troy A. Giambernardi, James C. Goolsby, Chao-Nan Qian, Cassandra R. Zylstra,<br />

Daniel R. Robinson, Pradip Roy-Burman, Aubie K. Shaw, Bree D. Buckner-Berghuis, Robert E. Sigler, James H. Resau, Ruth<br />

Sullivan, Wade Bushman, and Bart O. Williams. <strong>2007</strong>. Inactivation of Apc in the mouse prostate causes prostate carcinoma.<br />

Cancer Research 67(6): 2490–2496.<br />

Qian, Chao-Nan, James H. Resau, and Bin Tean Teh. <strong>2007</strong>. Prospects for vasculature reorganization in sentinel lymph nodes.<br />

Cell Cycle 6(5): 514–517.<br />

Wallar, Bradley J., Aaron D. DeWard, James H. Resau, and Arthur S. Alberts. <strong>2007</strong>. RhoB and the mammalian Diaphanousrelated<br />

formin mDia2 in endosome trafficking. Experimental Cell Research 313(3): 560–571.<br />

Zhang, Y.-W., B. Staal, Y. Su, P. Swiatek, P. Zhao, B. Cao, J. Resau, R. Sigler, R. Bronson, and G.F. Vande Woude. <strong>2007</strong>. Evidence<br />

that MIG-6 is a tumor-suppressor gene. Oncogene 26(2): 269–276.<br />

Moshitch-Moshkovitz, Sharon, Galia Tsarfaty, Dafna W. Kaufman, Gideon Y. Stein, Keren Shichrur, Eddy Solomon, Robert H. Sigler,<br />

James H. Resau, George F. Vande Woude, and Ilan Tsarfaty. 2006. In vivo direct molecular imaging of early tumorigenesis and<br />

malignant progression induced by transgenic expression of GFP-Met. Neoplasia 8(5): 353–363.<br />

Qian, Chao-Nan, Bree Berghuis, Galia Tsarfaty, MaryBeth Bruch, Eric J. Kort, Jon Ditlev, Ilan Tsarfaty, Eric Hudson, David G.<br />

Jackson, David Petillo, Jindong Chen, James H. Resau, and Bin Tean Teh. 2006. Preparing the “soil”: the primary tumor induces<br />

vasculature reorganization in the sentinel lymph node before the arrival of metastatic cancer cells. Cancer Research 66(21):<br />

10365–10376.<br />

Tsarfaty, Galia, Gideon Y. Stein, Sharon Moshitch-Moshkovitz, Dafna W. Kaufman, Brian Cao, James H. Resau, George F. Vande<br />

Woude, and Ilan Tsarfaty. 2006. HGF/SF increases tumor blood volume: a novel tool for the in vivo functional molecular imaging<br />

of Met. Neoplasia 8(5): 344–352.<br />

Yao, Xin, Chao-Nan Qian, Zhong-Fa Zhang, Min-Han Tan, Eric J. Kort, James H. Resau, and Bin Tean Teh. 2006. Two distinct<br />

types of blood vessels in clear cell renal cell carcinoma have contrasting prognostic implications. Clinical Cancer Research<br />

13(1): 161–169.<br />

.


VARI | <strong>2007</strong><br />

Pamela J. Swiatek, Ph.D., M.B.A.<br />

Laboratory of Germline Modification and Cytogenetics<br />

Dr. Swiatek received her M.S. (1984) and Ph.D. (1988) degrees in pathology from Indiana University.<br />

From 1988 to 1990, she was a postdoctoral fellow at the Tampa Bay Research Institute. From 1990<br />

to 1994, she was a postdoctoral fellow at the Roche Institute of Molecular Biology in the laboratory of<br />

Tom Gridley. From 1994 to 2000, Dr. Swiatek was a research scientist and Director of the Transgenic<br />

Core Facility at the Wadsworth Center in Albany, N.Y., and an Assistant Professor in the Department of<br />

Biomedical Sciences at the State University of New York at Albany. She joined VARI as a Special<br />

Program Investigator in August 2000. She has been the chair of the Institutional Animal Care and Use<br />

Committee since 2002 and is an Adjunct Assistant Professor in the College of Veterinary Medicine at<br />

Michigan State University. Dr. Swiatek received her M.B.A. in 2005 from Krannert School of Management<br />

at Purdue University. She was promoted to Senior <strong>Scientific</strong> Investigator in 2006.<br />

51<br />

Staff<br />

Laboratory Staff<br />

Students<br />

Visiting Scientists<br />

Sok Kean Khoo, Ph.D., Associate Laboratory Director<br />

Laura Ayotte, B.S.<br />

Julie Koeman, B.S., CLSp(CG)<br />

Kellie Sisson, B.S.<br />

Kaye Johnson, B.A.<br />

Diana Lewis


Van Andel Research Institute | <strong>Scientific</strong> <strong>Report</strong><br />

Research Interests<br />

The Germline Modification and Cytogenetics lab is a full-service lab that functions at the levels of service, research, and teaching<br />

to develop, analyze, and maintain mouse models of human disease. Our lab applies a business philosophy to core service<br />

offerings for both the VARI community and external entities. Our mission is to support mouse model and cytogenetics research<br />

with scientific innovation, customer satisfaction, and service excellence.<br />

Gene targeting<br />

Mouse models are produced using gene-targeting technology, a well-established, powerful method for inserting specific<br />

genetic changes into the mouse genome. The resulting mice can be used to study the effects of these changes in the complex<br />

biological environment of a living organism. The genetic changes can include the introduction of a gene into a specific site in<br />

the genome (gene “knock-in”) or the inactivation of a gene already in the genome (gene “knock-out”). Since these mutations are<br />

introduced into the reproductive cells known as the germline, they can be used to study the developmental aspects of gene<br />

function associated with inherited genetic diseases.<br />

52<br />

In addition to traditional gene-targeting technologies, the germline modification lab can produce mouse models in which the<br />

gene of interest is inactivated in a target organ or cell line instead of in the entire animal. These models, known as conditional<br />

knock-outs, are particularly useful in studying genes that, if missing, cause the mouse to die as an embryo. The lab also has<br />

the ability to produce mutant embryos that have a wild-type placenta using tetraploid embryo technology; this is useful when<br />

the gene-targeted mutation prevents implantation of the mouse embryo in the uterus. We also assist in the development of<br />

embryonic stem (ES) or fibroblast cell lines from mutant embryos, which allows for in vitro studies of the gene mutation.<br />

Our gene-targeting service encompasses three major procedures: DNA electroporation, clone expansion and cryopreservation,<br />

and microinjection. Gene targeting is initiated by mutating the genomic DNA of interest and inserting it into ES cells via<br />

electroporation. The mutated gene integrates into the genome and, by a process called homologous recombination, replaces one<br />

of the two wild-type copies of the gene in the ES cells. Clones are identified, isolated, and cryopreserved, and genomic DNA is<br />

extracted from each clone and delivered to the client for analysis. Correctly targeted ES cell clones are thawed, established into<br />

tissue culture, and cryopreserved in liquid nitrogen. Gene-targeting mutations are introduced by microinjection of the pluripotent<br />

ES cell clones into 3.5-day-old mouse embryos (blastocysts). These embryos, containing a mixture of wild-type and mutant ES<br />

cells, develop into mice called chimeras. The offspring of chimeras that inherit the mutated gene are heterozygotes possessing<br />

one copy of the mutated gene. The heterozygous mice are bred together to produce “knock-out mice” that completely lack the<br />

normal gene and have two copies of the mutant gene.<br />

Embryo/sperm cryopreservation<br />

We provide cryopreservation services for archiving and reconstituting valuable mouse strains. These cost-effective procedures<br />

decrease the need to continuously breed valuable mouse models, and they provide added insurance against the loss of custom<br />

mouse lines due to disease outbreak or a catastrophic event. Mouse embryos at various stages of development, as well as<br />

mouse sperm, can be cryopreserved and stored in liquid nitrogen; they can be thawed and used, respectively, by implantation<br />

into the oviducts of recipient mice or by in vitro fertilization of oocytes.


VARI | <strong>2007</strong><br />

Cytogenetics<br />

Our lab also directs the VARI cytogenetics core, which uses advanced molecular techniques to identify structural and numerical<br />

chromosomal aberrations in mouse, rat, and human cells. Tumor, fibroblast, blood, or ES cells can be grown in tissue culture,<br />

growth-arrested, fixed, and spread onto glass slides. Karyotyping of chromosomes using Leishman- or Giemsa-stained<br />

(G-banded) chromosomes is our basic service; spectral karyotyping (SKY) analysis of metaphase chromosome spreads in 24<br />

colors can aid in detecting subtle and complex chromosomal rearrangements. Fluorescence in situ hybridization (FISH) analysis,<br />

using indirectly or directly labeled bacterial artificial chromosome (BAC) or plasmid probes, can also be performed on metaphase<br />

spreads or on interphase nuclei derived from tissue touch preps or nondividing cells. Sequential staining of identical metaphase<br />

spreads using FISH and SKY can help identify the integration site of a randomly integrated transgene. Recently, FISH has been<br />

widely used to validate microarray data by confirming amplification/gain or deletion/loss of chromosomal regions of interest.<br />

Speed congenics<br />

Congenic mouse strain development traditionally involves a series of backcrosses, transferring a targeted mutation or genetic<br />

region of interest from a mixed genetic donor background to a defined genetic recipient background (usually an inbred strain).<br />

This process requires about ten generations (2.5 to 3 years) to attain 99.9% of the recipient’s genome. Since congenic mice have<br />

a more defined genetic background, phenotypic characteristics are less variable and the effects of modifier genes can be more<br />

pronounced.<br />

Speed congenics, also called marker-assisted breeding, uses DNA markers in a progressive breeding selection to accelerate the<br />

congenic process. For high-throughput genotyping, we use the state-of-the-art Sentrix BeadChip technology from Illumina, which<br />

contains 1,449 mouse single nucleotide polymorphisms (SNPs). These SNPs are strain-specific and cover the 10 most commonly<br />

used inbred mouse strains for optimal marker selection. The client provides the genomic DNA of male mice from the second,<br />

third, and fourth backcross generations for genotyping. The males having the highest percentage of the recipient’s genome from<br />

each generation are identified, and these mice are bred by the client. Using speed congenics, 99.9% of congenicity can be<br />

achieved in five generations (1 to 1.5 years).<br />

53<br />

Michigan Animal Model Consortium<br />

The VARI Germline Modification and Cytogenetics lab directs the Michigan Animal Model Consortium (MAMC), one of the ten<br />

Core Technology Alliance (CTA) collaborative core facilities located at the University of Michigan, Michigan State University,<br />

Wayne State University, Western Michigan University, Kalamazoo Valley Community College, Grand Valley State University, and<br />

VARI. The other facilities offer research services in proteomics, bioinformatics, structural biology, genomics, biological imaging,<br />

bioscience commercialization, high-throughput compound screening, good manufacturing practices, and antibody technology.<br />

The MAMC labs were developed with funding from the Michigan Economic<br />

Development Corporation and provide efficient mouse modeling services<br />

to researchers studying human diseases. MAMC’s long-term goal is to<br />

offer a comprehensive set of cutting-edge services that, through continuous<br />

enhancements and development, will define our organization as a single<br />

point-of-service site for animal models research. Centralized provision<br />

of services maximizes research productivity and decreases time to<br />

discovery and is in high demand by academia and pharmaceutical and<br />

biotechnology companies, which are increasingly looking to outsource to<br />

service centers. Through its well-organized service structure and staff<br />

of experts, MAMC supports the growth of the life science industry in<br />

Michigan, which is congruent with the CTA goals.<br />

From left, front row: Koeman, Sisson, Swiatek, Ayotte<br />

back row: Johnson, Lewis, Khoo


Van Andel Research Institute | <strong>Scientific</strong> <strong>Report</strong><br />

MAMC service offerings<br />

Animal model development<br />

• Mouse transgenics. Transgenic technology is used to produce genetically engineered mice expressing foreign genes<br />

and serving as models for human disease research. Microinjection delivers the foreign DNA into the pronucleus of a<br />

one-cell fertilized egg. This service is provided using various strains of laboratory mice, with production of three<br />

transgenic founder mice guaranteed from each procedure.<br />

• Gene targeting. By transfecting mouse embryonic stem cells with inactivating, homologous DNAs, target gene<br />

expression can be shut down. Genetically engineered mice are produced by microinjecting mutant stem cells into<br />

mouse embryos and breeding the progeny to mutant homozygosity. This service is provided using 129 or C57BL/6<br />

embryonic stem cells.<br />

• Xenotransplantation. Human cancer cells are injected into immunodeficient mice to produce human-derived<br />

tumors. Protocols are designed to test anti-tumor treatment regimens that can lead to prognostic, diagnostic, or<br />

therapeutic procedures for humans.<br />

Animal model analysis<br />

• Cytogenetics. Mouse and rat chromosomal abnormalities and genetic loci are visually observed using Giemsa stain,<br />

SKY, or FISH techniques.<br />

• Necropsy. Mice are dissected postmortem and tissues are fixed for histological analysis, with necropsy reports<br />

generated using voice-recognition software.<br />

54<br />

• Histology. Histological sections are prepared from mouse tissues using microtomes and cryostats; they are processed<br />

and stained using automated instruments and then are microscopically analyzed.<br />

• Veterinary pathology. A board-certified veterinary pathologist holding the D.V.M. and Ph.D. degrees provides<br />

expert microscopic analysis and project consultation.<br />

• DNA isolation. DNA is isolated from mouse tail biopsies using the AutogenPrep 960 instrument.<br />

Animal model maintenance and preservation<br />

• Mouse rederivation. All mouse strains entering the specific pathogen–free breeding facility are rederived to specific<br />

pathogen–free mouse status using embryo transfer techniques.<br />

• Animal technical services. Veterinary services such as injections, measurements, mating set-up, and tail biopsies are<br />

performed by the animal technician staff.<br />

• Contract breeding. Wild-type mouse strains and genetically engineered animal models are maintained for research<br />

purposes by breeding the strains in a specific pathogen–free environment.<br />

• Embryo/sperm cryopreservation. Genetically engineered mice are preserved for archival purposes, disease control,<br />

genetic stability, and economic efficiency using germplasm cryopreservation techniques.<br />

• Cancer model repository. Mouse cancer models of research interest are maintained through breeding strategies.<br />

Recent Publications<br />

Furge, Kyle A., Jindong Chen, Julie Koeman, Pamela Swiatek, Karl Dykema, Kseniji Lucin, Richard Kahnoski, Ximing J. Yang, and<br />

Bin Tean Teh. <strong>2007</strong>. Detection of DNA copy number changes and oncogenic signaling abnormalities from gene expression data<br />

reveals MYC activation in high-grade papillary renal cell carcinoma. Cancer Research 67(7): 3171–3176.<br />

Zhang, Y.-W., B. Staal, Y. Su, P. Swiatek, P. Zhao, B. Cao, J. Resau, R. Sigler, R. Bronson, and G.F. Vande Woude. <strong>2007</strong>. Evidence<br />

that MIG-6 is a tumor-suppressor gene. Oncogene 26(2): 269–276.<br />

Mukhopadhyay, Rita, Ye-Shih Ho, Pamela J. Swiatek, Barry P. Rosen, and Hiranmoy Bhattacharjee. 2006. Targeted disruption of<br />

the mouse Asnal gene results in embryonic lethality. FEBS Letters 580(16): 3889–3894.


VARI | <strong>2007</strong><br />

Bin T. Teh, M.D., Ph.D.<br />

Laboratory of Cancer Genetics<br />

Dr. Teh obtained his M.D. from the University of Queensland, Australia, in 1992, and his Ph.D. from<br />

the Karolinska Institute, Sweden, in 1997. Before joining the Van Andel Research Institute (VARI), he<br />

was an Associate Professor of medical genetics at the Karolinska Institute. Dr. Teh joined VARI as a<br />

Senior <strong>Scientific</strong> Investigator in January 2000. His research mainly focuses on kidney cancer, and he is<br />

currently on the Medical Advisory Board of the Kidney Cancer Association. Dr. Teh was promoted to<br />

Distinguished <strong>Scientific</strong> Investigator in 2005.<br />

55<br />

Staff<br />

Laboratory Staff<br />

Chao-Nan (Miles) Qian, M.D., Ph.D.<br />

Peng-Fei Wang, M.D., Ph.D.<br />

Xin Yao, M.D., Ph.D.<br />

Eric Kort, M.D.<br />

Daisuke Matsuda, M.D.<br />

Jindong Chen, Ph.D.<br />

Leslie Farber, Ph.D.<br />

Kunihiko Futami, Ph.D.<br />

Dan Huang, Ph.D.<br />

Sok Kean Khoo, Ph.D.<br />

Students<br />

Visiting Scientists<br />

Yan Li, Ph.D.<br />

Douglas Luccio-Camelo, Ph.D.<br />

David Petillo, Ph.D.<br />

Zhongfa (Jacob) Zhang, Ph.D.<br />

Stephanie Bender, M.S.<br />

Wangmei Luo, M.S.<br />

Mark Betten, B.S.<br />

Aaron Massie, B.S.<br />

Michael Westphal, B.S.<br />

Sabrina Noyes, B.S.


Van Andel Research Institute | <strong>Scientific</strong> <strong>Report</strong><br />

Research Interests<br />

Kidney cancer, or renal cell carcinoma (RCC), is the tenth most common cancer in the United States (35,000 new cases and<br />

more than 13,000 deaths a year). Its incidence has been increasing, a phenomenon that cannot be accounted for by the wider<br />

use of imaging procedures. We have established a comprehensive and integrated kidney research program, and our major<br />

research goals are 1) to identify the molecular signatures of different subtypes of kidney tumors, both hereditary and sporadic,<br />

and to understand how genes function and interact in giving rise to the tumors and their progression; 2) to identify and develop<br />

diagnostic and prognostic biomarkers for kidney cancer; 3) to identify and study novel and established molecular drug targets<br />

and their sensitivity and resistance; and 4) to develop animal models for drug testing and preclinical bioimaging.<br />

Our program to date has established a worldwide network of collaborators; a tissue bank containing fresh-frozen tumor pairs (over<br />

1,000 cases) and serum; and a gene expression profiling database of 500 tumors, with long-term clinical follow-up information for<br />

half of them. Our program includes positional cloning of hereditary RCC syndromes and functional studies of their related genes,<br />

microarray and bioinformatic analysis, generation of RCC mouse models, and more recently, molecular therapeutic studies.<br />

Hereditary RCC syndromes<br />

We are currently focusing on the cloning of the gene responsible for familial clear cell renal cell carcinoma, which is a separate<br />

entity from von Hippel-Lindau (VHL) and from familial RCC with a chromosome-3 translocation. These efforts involve the use of<br />

high-density, single-nucleotide-polymorphism (SNP) microarrays and correlation with our existing gene expression profiles.<br />

56


VARI | <strong>2007</strong><br />

Microarray gene expression profiling and bioinformatics<br />

High-density SNP genotyping has been performed on some of the specimens registered in our RCC expression database.<br />

We are currently focusing on analysis and data mining. Clinically, we continue to subclassify the tumors by correlation with<br />

clinicopathological information. One example is the study of the unclassified group of tumors for which the histological diagnosis<br />

is “unknown”. We have also identified a specific set of genes that can distinguish chromophobe (malignant) from oncocytoma<br />

(benign), two types that share a high degree of similarity in their expression profiles. Our database has proven to be very useful<br />

in RCC research, since we can obtain differential expression of any gene in seconds; this has led to numerous collaborations. We<br />

are currently combining SNP and expression data to identify novel RCC-related genes.<br />

Mouse models of kidney cancer and molecular therapeutic studies<br />

We have generated several kidney-specific conditional knock-outs including APC, PTEN, and VHL. The first two knock-outs<br />

give rise to renal cysts and tumors, whereas VHL remains neoplasia-free; double knock-outs are also being studied. We have<br />

successfully generated nine xenograft RCC models via subcapsular injection that have characteristic clinical features and<br />

outcomes. Tumors and serum have been harvested for a baseline data set. We are currently performing in vitro and in vivo<br />

studies on several new drugs for kidney cancer.<br />

Molecular and cellular studies<br />

We use numerous well-characterized kidney cancer cell lines to study the functions of novel kidney cancer–related genes by<br />

overexpressing or down-regulating the genes. In addition, we perform cell cycle, proliferation, and migration assays to assess<br />

the cellular effects of these genes. These studies are usually coupled with in vivo studies.<br />

57<br />

External Collaborators<br />

We have extensive collaborations with researchers and clinicians in the United States and overseas.<br />

From left: Zhang, Qian, Massie, Noyes, Westphal, Farber, Kort, Chen, Petillo, Matsuda, Teh


Van Andel Research Institute | <strong>Scientific</strong> <strong>Report</strong><br />

Recent Publications<br />

Al-sarraf, N., S. Mahmood, J.N. Reif, J. Hinrichsen, B.T. Teh, E. McGovern, P. De Meyts, K.J. O’Byrne, and S.G. Gray. In press.<br />

DOK4/IRS-5 expression is altered in clear cell renal cell carcinoma. International Journal of Cancer.<br />

Daly, A.F., J.-F. Vanbellinghen, S.K. Khoo, M.-L. Jaffrain-Rea, L.A. Naves, M.A. Guitelman, A. Murat, P. Emy, A.-P.<br />

Gimenez-Roqueplo, G. Tamburrano, G. Raverot, A. Barlier, W. De Herder, A. Penfornis, E. Ciccarelli, et al. In press. Aryl hydrocarbon<br />

receptor interacting protein gene mutations in familial isolated pituitary adenomas: analysis in 73 families. Journal of Clinical<br />

Endocrinology and Metabolism.<br />

Evans, Andrew J., Ryan C. Russell, Olga Roche, T. Nadine Burry, Jason E. Fish, Vinca W.K. Chow, William Y. Kim, Arthy Saravanan,<br />

Mindy A. Maynard, Michelle L. Gervais, Roxana I. Sufan, Andrew M. Roberts, Leigh A. Wilson, Mark Betten, Cindy Vandewalle, et<br />

al. <strong>2007</strong>. VHL promotes E2 box–dependent E-cadherin transcription by HIF-mediated regulation of SIP1 and Snail. Molecular<br />

and Cellular Biology 27(1): 157–169.<br />

Furge, Kyle A., Jindong Chen, Julie Koeman, Pamela Swiatek, Karl Dykema, Kseniji Lucin, Richard Kahnoski, Ximing J. Yang, and<br />

Bin Tean Teh. <strong>2007</strong>. Detection of DNA copy number changes and oncogenic signaling abnormalities from gene expression data<br />

reveals MYC activation in high-grade papillary renal cell carcinoma. Cancer Research 67(7): 3171–3176.<br />

58<br />

Furge, K.A., M.H. Tan, K. Dykema, E. Kort, W. Stadler, X. Yao, M. Zhou, and B.T. Teh. <strong>2007</strong>. Identification of deregulated oncogenic<br />

pathways in renal cell carcinoma: an integrated oncogenomic approach based on gene expression profiling. Oncogene 26(9):<br />

1346–1350.<br />

Gad, S., S.H. Lefèvre, S.K. Khoo, S. Giraud, A. Vieillefond, V. Vasiliu, S. Ferlicot, V. Molinié, Y. Denoux, N. Thiounn, Y. Chrétien,<br />

A. Méjean, M. Zerbib, G. Benoît, J.M. Hervé, G. Allègre, B. Bressac-de Paillerets, B.T. Teh, and S. Richard. <strong>2007</strong>. Mutations in<br />

BHD and TP53 genes, but not in HNF1β gene, in a large series of sporadic chromophobe renal cell carcinoma. British Journal<br />

of Cancer 96(2): 336–340.<br />

Greenman, Christopher, Philip Stephens, Raffaella Smith, Gillian L. Dalgliesh, Christopher Hunter, Graham Bignell, Helen Davies,<br />

Jon Teague, Adam Butler, Claire Stevens, Sarah Edkins, Sarah O’Meara, Imre Vastrik, Esther E. Schmidt, Tim Avis, et al. <strong>2007</strong>.<br />

Patterns of somatic mutation in human cancer genomes. Nature 446(7132): 153–158.<br />

Lin, Fan, Ping L. Zhang, Ximing J. Yang, Jianhui Shi, Tom Blasick, Won K. Han, Hanlin L. Wang, Steven S. Shen, Bin T. Teh, and<br />

Joseph V. Bonventre. <strong>2007</strong>. Human kidney injury molecule-1 (hKIM-1): a useful immunohistochemical marker for diagnosing<br />

renal cell carcinoma and ovarian clear cell carcinoma. American Journal of Surgical Pathology 31(3): 371–381.<br />

Qian, Chao-Nan, James H. Resau, and Bin Tean Teh. <strong>2007</strong>. Prospects for vasculature reorganization in sentinel lymph nodes.<br />

Cell Cycle 6(5): 514–517.<br />

Wang, Kim L., David M. Weinrach, Chunyan Luan, Misop Han, Fan Lin, Bin Teh, and Ximing J. Yang. <strong>2007</strong>. Renal papillary<br />

adenoma—a putative precursor of papillary renal cell carcinoma. Human Pathology 38(2): 239–246.<br />

Yang, X.J., M. Takahashi, K.T. Schafernak, M.S. Tretiakova, J. Sugimura, N.J. Vogelzang, and B.T. Teh. <strong>2007</strong>. Does “granular cell”<br />

renal cell carcinoma exist? Molecular and histopathological reclassification. Histopathology 50(5): 678–680.<br />

Adley, Brian P., Anita Gupta, Fan Lin, Chunyan Luan, Bin T. Teh, and Ximing J. Yang. 2006. Expression of kidney-specific<br />

cadherin in chromophobe renal cell carcinoma and renal oncocytoma. American Journal of Clinical Pathology 126(1): 79–85.


VARI | <strong>2007</strong><br />

Adley, Brian P., Veronica Papavero, Jun Sugimura, B.T. Teh, and Ximing J. Yang. 2006. Diagnostic value of cytokeratin 7 and<br />

parvalbumin in differentiating chromophobe renal cell carcinoma from renal oncocytoma. Analytical and Quantitative Cytology<br />

and Histology 28(4): 228–236.<br />

Furge, Kyle A., Eric J. Kort, Ximing J. Yang, Walter M. Stadler, Hyung Kim, and Bin Tean Teh. 2006. Gene expression profiling in<br />

kidney cancer: combining differential expression and chromosomal and pathway analyses. Clinical Genitourinary Cancer 5(3):<br />

227–231.<br />

Pimenta, Flávio J., Letícia F.G. Silveira, Gabriela C. Taveres, Andreza C. Silva, Paolla F. Perdigão, Wagner H. Castro, Marcus V.<br />

Gomez, Bin T. Teh, Luiz De Marco, and Ricardo S. Gomez. 2006. HRPT2 gene alterations in ossifying fibroma of the jaws. Oral<br />

Oncology 42(7): 735–739.<br />

Qian, Chao-Nan, Bree Berghuis, Galia Tsarfaty, MaryBeth Bruch, Eric J. Kort, Jon Ditlev, Ilan Tsarfaty, Eric Hudson, David G.<br />

Jackson, David Petillo, Jindong Chen, James H. Resau, and Bin Tean Teh. 2006. Preparing the “soil”: the primary tumor induces<br />

vasculature reorganization in the sentinel lymph node before the arrival of metastatic cancer cells. Cancer Research 66(21):<br />

10365–10376.<br />

Tretiakova, M., M. Turkyilmaz, T. Grushko, M. Kocherginsky, C. Rubin, B. Teh, and X.J. Yang. 2006. Topoisomerase IIα expression<br />

in Wilms’ tumour: gene alterations and immunoexpression. Journal of Clinical Pathology 59(12): 1272–1277.<br />

Yang, Ximing J., Jun Sugimura, Kristian T. Schafernak, Maria S. Tretiakova, Misop Han, Nicholas J. Vogelzang, Kyle Furge, and<br />

Bin Tean Teh. 2006. Classification of renal neoplasms based on molecular signatures. Journal of Urology 175(6): 2302–2306.<br />

59<br />

Yao, Xin, Chao-Nan Qian, Zhong-Fa Zhang, Min-Han Tan, Eric J. Kort, James H. Resau, and Bin Tean Teh. 2006. Two distinct<br />

types of blood vessels in clear cell renal cell carcinoma have contrasting prognostic implications. Clinical Cancer Research<br />

13(1): 161–169.<br />

Zhang, Chun, Dong Kong, Min-Han Tan, Donald L. Pappas, Jr., Peng-Fei Wang, Jindong Chen, Leslie Farber, Nian Zhang, Han-<br />

Mo Koo, Michael Weinreich, Bart O. Williams, and Bin Tean Teh. 2006. Parafibromin inhibits cancer cell growth and causes G1<br />

phase arrest. Biochemical and Biophysical Research Communications 350(1): 17–24.<br />

Zynger, Debra L., Nikolay D. Dimov, Chunyan Luan, Bin Tean Teh, and Ximing J. Yang. 2006. Glypican 3: a novel marker in<br />

testicular germ cell tumors. American Journal of Surgical Pathology 30(12): 1570–1575.


Van Andel Research Institute | <strong>Scientific</strong> <strong>Report</strong><br />

60<br />

Photo by Lia Tesfay of the Miranti lab<br />

and Jim Resau of the Resau lab.


VARI | <strong>2007</strong><br />

Normal mouse prostate tissue fixed and stained to visualize CD82 protein (brown).<br />

61<br />

CD82 is a metastasis suppressor protein. Its presence is seen in the groups of epithelial cells that surround the lumens in this normal prostate tissue.


Van Andel Research Institute | <strong>Scientific</strong> <strong>Report</strong><br />

Steven J. Triezenberg, Ph.D.<br />

Laboratory of Transcriptional Regulation<br />

62<br />

Dr. Triezenberg received his bachelor’s degree in biology and education at Calvin College in Grand<br />

Rapids, Michigan. His Ph.D. training in cell and molecular biology at the University of Michigan was<br />

followed by postdoctoral research in the laboratory of Steven L. McKnight at the Carnegie Institution of<br />

Washington. Dr. Triezenberg was a faculty member of the Department of Biochemistry and Molecular<br />

Biology at Michigan State University for more than 18 years, where he also served as Associate Director<br />

of the Graduate Program in Cell and Molecular Biology. Dr. Triezenberg was recruited to VAI to serve as<br />

the founding Dean of the Van Andel Institute Graduate School and as a <strong>Scientific</strong> Investigator in the Van<br />

Andel Research Institute, arriving in May 2006.<br />

Laboratory Staff<br />

Staff<br />

Martha Roemer, M.S.<br />

Student<br />

Sebla Kutluay, B.S.


VARI | <strong>2007</strong><br />

Research Interests<br />

The genetic information encoded in DNA must first be transcribed in the form of RNA before it can be translated into the proteins<br />

that do most of the work in a cell. Some genes must be expressed more or less constantly throughout the life of any eukaryotic<br />

cell. Others must be turned on (or turned off) in particular cells either at specific times or in response to a specific signal or event.<br />

Thus, regulation of gene expression is a key determinant of cell function. Our laboratory explores the mechanisms that regulate<br />

the first step in that flow, the process termed transcription.<br />

Over the past 20 years, my laboratory has used infection by herpes simplex virus as an experimental context for exploring the<br />

mechanisms of transcriptional activation. In the past 10 years, we have also asked similar questions in a very different biological<br />

context, the acclimation of plants to cold temperature.<br />

Transcriptional activation during herpes simplex virus infection<br />

Herpes simplex virus type 1 (HSV-1) causes the common cold sore or fever blister. The initial lytic or productive infection by<br />

HSV-1 results in obvious symptoms in the skin and mucosa, typically in or around the mouth. After the initial infection resolves,<br />

HSV-1 finds its way into nerve cells, where the virus can “hide” in a latent mode for long times—essentially for the lifetime of the<br />

host organism. Occasionally, some trigger event (such as emotional stress, damage to the nerve from a sunburn, or a root canal<br />

operation) will cause the virus to reactivate, producing new viruses in the nerve cell and sending those viruses back to the skin<br />

to cause a recurrence of the cold sore.<br />

The DNA genome of HSV-1 encodes approximately 80 different proteins. However, the virus does not have its own machinery for<br />

expressing those genes; instead, it must divert the gene expression machinery of the host cell. That process is triggered by a<br />

viral regulatory protein designated VP16, whose function it is to stimulate transcription of the first viral genes to be expressed in<br />

the infected cell (the immediate-early, or IE, genes).<br />

63<br />

VP16 recruitment of host cell transcription machinery<br />

The prevailing model for the mechanism of transcriptional activation is that a portion of an activating protein (such as VP16) called<br />

the activation domain (AD) can bind to the host cell RNA polymerase II or to its accessory proteins. In this manner, VP16 recruits<br />

or tethers accessory proteins to the genes that are to be activated. Over the years, several accessory proteins (also known as<br />

general transcription factors) have been implicated as potential targets for VP16. Of those, the evidence seems to point most<br />

directly at TFIID, a multi-protein complex that includes the TATA-binding protein (TBP). TBP itself can bind rather efficiently to<br />

the VP16 activation domain, and mutations in VP16 that disrupt transcriptional activation also disrupt the interaction with TBP.<br />

We have pursued the structure of the VP16-TBP interaction by methods including X-ray crystallography and nuclear magnetic<br />

resonance. We have also tested the hypothesis that VP16 can influence the orientation of TBP on the TATA-box DNA of a target<br />

gene promoter. This hypothesis, proposed by other laboratories, is based on the fact that both TBP and the TATA sequence to<br />

which it binds are quite symmetric, and yet TBP can effectively support transcription in only a single orientation. We developed<br />

a new quantitative method for assessing TBP orientation and using this method have now demonstrated that TBP binds in a<br />

well-oriented manner even in the absence of VP16. Moreover, on a TATA site engineered to be completely symmetric, to which<br />

TBP binds in both orientations, the VP16 activation domain has no significant influence. This work resolves a long-standing issue<br />

regarding TBP orientation and eliminates one hypothesis for the mechanism of transcriptional activation.


Van Andel Research Institute | <strong>Scientific</strong> <strong>Report</strong><br />

Chromatin-modifying coactivators in herpes virus infections<br />

Eukaryotic DNA is typically packaged as chromatin, in which the DNA is wrapped around “spools” of histone proteins, and<br />

these spools are then further arranged into higher-order structures. This packaging creates an impediment to transcription,<br />

during which RNA polymerase must separate the two strands of DNA. The impediment can be overcome with the help of<br />

chromatin-modifying coactivator proteins, some of which alter the histone proteins by post-translational modifications<br />

(e.g., acetylation or methylation) and others of which can slide or remove the histone proteins to permit access by RNA<br />

polymerase to the DNA.<br />

Experiments using the VP16 activation domain in artificial contexts (for example, in yeast genetic assays) have indicated that<br />

VP16 can recruit various coactivator proteins to target genes. However, the HSV-1 viral DNA is not packaged with histones in<br />

the infectious virion, and prior evidence suggested the viral DNA remained largely chromatin-free during infection. Therefore, we<br />

wondered whether VP16 would recruit these coactivators to viral IE genes, and if so whether those coactivators would be acting<br />

on histone proteins (which didn’t seem to be present) or on some other target. Our results have clearly indicated that VP16 can<br />

recruit certain coactivators to IE genes during lytic infection. We have also shown that at least some histone proteins do associate<br />

with viral DNA, although perhaps not to the same extent as with cellular DNA. We are currently exploring further which histones<br />

associate with viral DNA, how quickly they are put in place, the mechanisms used to put them in place, and what VP16 and other<br />

regulatory proteins might do to counteract the repressive effects of chromatin, which could be considered a molecular defense<br />

mechanism.<br />

64<br />

Can a curry spice block herpes infections?<br />

Curcumin, the bright yellow component of the curry spice turmeric, affects eukaryotic cells in several ways. Another laboratory<br />

has reported that curcumin could block the histone acetyltransferase activity of two coactivator proteins, p300 and CBP. Because<br />

we had shown that VP16 can recruit p300 and CBP to viral IE gene promoters, we tested whether curcumin, as an inhibitor of p300<br />

or CBP activity, would block viral IE gene expression and thus block HSV infection. Indeed, curcumin has dramatic effects on IE<br />

gene expression and substantial effects on virus infection (Fig. 1). We are now trying to determine whether that effect is indeed<br />

channeled through the p300 and CBP proteins or whether it arises from another of the biological activities of curcumin.<br />

Figure 1.<br />

- curcumin + curcumin<br />

Figure 1. HSV-1 infection of Vero cell monolayers.<br />

HSV-1 infection results in plaques or holes in a monolayer of<br />

cultured human cells (left). In the presence of curcumin (right),<br />

plaques are generally smaller and the cells within the plaques<br />

are not as completely obliterated. Photo by M. Roemer.


VARI | <strong>2007</strong><br />

Gene activation during cold acclimation in plants<br />

Although plants and their cells obviously have very different forms and functions than animals and their cells, the mechanisms used<br />

for expressing genetic information are quite similar. About ten years ago, we applied our emerging interest in chromatin-modifying<br />

coactivators to an interesting question in plant biology. Some plants, including the prominent experimental organism Arabidopsis,<br />

can sense low (but nonfreezing) temperature in a way that provides protection from subsequent freezing temperatures (Fig. 2).<br />

This process is known as cold acclimation. Michael Thomashow, an MSU plant scientist, has explored the genes expressed<br />

during this process, and we collaborated with his laboratory to explore the mechanisms involved. We have characterized one<br />

particular histone acetyltransferase, termed GCN5, and two of its accessory proteins, ADA2a and ADA2b. Mutations in the genes<br />

encoding these coactivator proteins result in diminished expression of cold-regulated genes. Moreover, histones located at these<br />

cold-regulated genes become more highly acetylated during initial stages of cold acclimation. We are now working to determine<br />

whether GCN5 and the ADA2 proteins are partially or fully responsible for this cold-induced acetylation. We are also collaborating<br />

with groups in Greece and Pennsylvania to explore the distinct biological activities of the two ADA2 proteins. This work may help<br />

us understand whether the mechanisms by which plants express their genes can be effectively modulated so as to protect crop<br />

plants from loss in yield or viability due to environmental stresses such as low temperature.<br />

Figure 2.<br />

Non-acclimated<br />

Acclimated<br />

Figure 2. Acclimation of Arabidopsis seedlings.<br />

Arabidopsis seedlings were grown on agar plates for three weeks<br />

at 20 °C. The plants in the right panel were chilled at 4 °C for two<br />

days. All plants were then subjected to subfreezing temperatures<br />

(–5 °C) for one day and then were returned to warm temperatures<br />

to recover. The acclimated plants remain healthy and green; the<br />

nonacclimated plants lose much of their color and die.<br />

Photo by K. Pavangadkar.<br />

65<br />

External Collaborators<br />

From left: Triezenberg, Roemer, Kutluay<br />

Kanchan Pavangadkar and Michael F. Thomashow, Michigan State University, East Lansing<br />

Amy S. Hark, Muhlenberg College, Allentown, Pennsylvania<br />

Kostas Vlachonasios, Aristotle University of Thessaloniki, Greece


Van Andel Research Institute | <strong>Scientific</strong> <strong>Report</strong><br />

George F. Vande Woude, Ph.D.<br />

Laboratory of Molecular Oncology<br />

66<br />

Dr. Vande Woude received his M.S. (1962) and Ph.D. (1964) from Rutgers University. From 1964–1972,<br />

he served first as a postdoctoral research associate, then as a research virologist for the U.S. Department<br />

of Agriculture at Plum Island Animal Disease Center. In 1972, he joined the National Cancer Institute as<br />

Head of the Human Tumor Studies and Virus Tumor Biochemistry sections and, in 1980, was appointed<br />

Chief of the Laboratory of Molecular Oncology. In 1983, he became Director of the Advanced Bioscience<br />

Laboratories–Basic Research Program at the National Cancer Institute’s Frederick Cancer Research and<br />

Development Center, a position he held until 1998. From 1995, Dr. Vande Woude first served as Special<br />

Advisor to the Director, and then as Director for the Division of Basic Sciences at the National Cancer<br />

Institute. In 1999, he was recruited to become the first Director of the Van Andel Research Institute.<br />

Staff<br />

Laboratory Staff<br />

Student<br />

Guest Researchers<br />

Yu-Wen Zhang, M.D., Ph.D.<br />

Chongfeng Gao, Ph.D.<br />

Carrie Graveel, Ph.D.<br />

Qian Xie, Ph.D.<br />

Dafna Kaufman, M.Sc.<br />

Matt VanBrocklin, M.S.<br />

Jack DeGroot, B.S.<br />

Betsy Haak, B.S.<br />

Liang Kang, B.S.<br />

Rachel Kuznar, B.S.<br />

Benjamin Staal, B.S.<br />

Ryan Thompson, B.S.<br />

Yanli Su, A.M.A.T.<br />

Angelique Berens<br />

David Wenkert, M.D.<br />

Yuehai Shen, Ph.D.<br />

Edwin Chen, B.S.


VARI | <strong>2007</strong><br />

Research Interests<br />

A mouse model of mutationally activated Met<br />

Signaling through Met and its ligand, HGF/SF, has been implicated in most types of human cancer. Compelling genetic evidence<br />

for the role of Met stems from the discovery that activating gain-of-function mutations are found in human kidney cancers and<br />

in other cancer types (http://www.vai.org/met/). To study how Met-activating mutations are involved in tumor development, we<br />

generated mice bearing mutations in the endogenous Met locus representative of both the inherited and the sporadic mutations<br />

found in human cancers. On a C57BL/6 background, the different mutant Met lines developed unique tumor profiles, including<br />

carcinomas, sarcomas, and lymphomas. We have found that the differences in tumor types and latency may be due to signaling<br />

differences triggered by the specific mutation in a tissue- or stem cell–specific pattern. Cytogenetic analysis of all tumor types<br />

shows frequent trisomy of the Met locus. Moreover, it is the mutant met allele that is amplified and likely to be required for tumor<br />

progression. When mutant Met was transferred to the FVB/N mouse background, these animals developed aggressive mammary<br />

tumors. Therefore, understanding the signaling specificity of these mutations is essential for developing successful cancer<br />

therapeutics. Our mutant mice provide a valuable model for testing Met inhibitors and for understanding the molecular events<br />

crucial for Met-mediated tumorigenesis.<br />

A novel mouse model for preclinical studies<br />

67<br />

We have generated a severe combined immune deficiency (SCID) mouse strain carrying a human HGF/SF transgene. This mouse<br />

provides a species-compatible ligand for propagating human tumor cells expressing human Met. The growth of Met-expressing<br />

human tumor xenografts can be significantly enhanced in this transgenic mouse relative to growth in nontransgenic hosts. This<br />

immunocompromised strain is vital for examining the role of Met in human tumor malignancy. We are developing metastasis<br />

models and generating orthotopic xenografts of human tumor cells. This model is being used for preclinical testing of drugs or<br />

compounds targeting the HGF/SF-Met complex and downstream signaling pathways.<br />

Understanding the “multiple personalities” of cancer cells<br />

Several years ago, we asked whether tumor cells can switch between proliferative and invasive phenotypes. We discovered<br />

that tumor cells can indeed switch, and they can do so rapidly; they may also express both proliferative and invasive features.<br />

We have established in vitro methods for selecting highly proliferative or highly invasive tumor cell populations that may mimic<br />

the in vivo process of clonal selection during tumor progression. We have determined that chromosomal instability correlates<br />

with the proliferative and invasive phenotypes. Using spectral karyotyping (SKY) and M-Fish, we observe significant changes in<br />

chromosome content with each phenotype, and the changes show remarkable concordance with changes in gene expression.<br />

Regional gene expression changes appear to favor the expression of specific genes appropriate for the invasive or proliferative<br />

phenotype. Moreover, the ratio of chromosomal changes closely parallels the ratio of gene expression in the chromosome. These<br />

results show that chromosome instability and the resulting heterogeneous chromosome composition provide the diversity in gene<br />

expression to allow tumor cell clonal evolution.


Van Andel Research Institute | <strong>Scientific</strong> <strong>Report</strong><br />

Examining how geldanamycin inhibits tumor cell invasion<br />

Our lab has been studying the mechanism by which geldanamycin (GA) inhibits urokinase activation of plasmin from plasminogen<br />

(uPA). Previously, we have shown that a subset of GA-related drug derivatives inhibits HGF/SF-induced activation of plasmin in<br />

canine MDCK cells. We found that such inhibition also occurs in several human glioblastoma tumor cell lines. Curiously, these<br />

GA drugs inhibit HGF/SF-induced uPA activation and block MDCK cell scattering and glioblastoma tumor cell invasion in vitro at<br />

concentrations below that required to exhibit a measurable effect on Met degradation through HSP90. This inhibition is observed<br />

only with HGF/SF-mediated activation and only when the magnitude of HGF/SF-uPA induction is 1.5 times basal uPA-plasmin<br />

activity.<br />

Inhibition of MAPK in melanoma<br />

68<br />

Extracellular signals activate mitogen-activated protein kinase (MAPK) cascades, potentiating biological activities such as cell<br />

proliferation, differentiation, and survival. Constitutive activation of MAPK signaling pathways is implicated in the development<br />

and progression of many human cancers, including melanoma. Mutually exclusive activating mutations in NRas or BRAF are<br />

found in about 85% of all melanomas, resulting in constitutive activation of the MAPK pathway (Ras-BRaf-MEK-Erk-Rsk). We<br />

have previously demonstrated that inhibition of this pathway with small-molecule MEK inhibitors selectively induces apoptosis in<br />

human melanoma cells but not in normal melanocytes both in vitro and in vivo. These results support the concept that the MAPK<br />

pathway represents a tumor-specific survival signaling pathway in melanoma cells and that targeting members of this pathway<br />

may be an effective therapeutic strategy. Understanding the mechanisms by which constitutive MAPK promotes survival and<br />

defining the minimal vital MAPK pathway components required for the development and progression of melanoma may have<br />

direct translational implications. Preliminary data suggest that MAPK activation actively suppresses several pro-apoptotic Bcl-2<br />

family members. We are currently using the specific small-molecule MEK inhibitor PD184352 together with molecular biological<br />

approaches to selectively modulate the expression and function of these molecules in order to validate and develop them as<br />

novel therapeutic targets for treating melanoma and other MAPK-associated cancers.<br />

External Collaborators<br />

Francesco DeMayo, Baylor College of Medicine, Houston, Texas<br />

Ermanno Gherardi, MRC Center, Cambridge, England<br />

Nadia Harbeck, Technische Universität, Munich, Germany<br />

Beatrice Knudsen, Fred Hutchinson Cancer Research Center, Seattle, Washington<br />

Ernest Lengyel, University of Chicago, Illinois<br />

Patricia LoRusso, Karmanos Cancer Institute, Detroit, Michigan<br />

Benedetta Peruzzi, National Cancer Institute, Bethesda, Maryland<br />

Alnawaz Rehemtulla, Brian Ross, and Richard Simon, University of Michigan, Ann Arbor<br />

Ilan Tsarfaty, Tel Aviv University, Israel<br />

Robert Wondergem, East Tennessee State University, Johnson City


VARI | <strong>2007</strong><br />

From left: Gao, Thompson, Xie, Graveel, Kaufman, Vande Woude, Staal, Bassett, Haak, Nelson, DeGroot, Su, Zhang<br />

Recent Publications<br />

Lindemann, K.K., J. Resau, J. Nährig, E. Kort, B. Leeser, K. Anneke, A. Welk, J. Schäfer, G.F. Vande Woude, E. Lengyel, and N.<br />

Harbeck. In press. Differential expression of c-Met, its ligand HGF/SF, and HER2/neu in DCIS and adjacent normal breast tissue.<br />

Histopathology.<br />

69<br />

Zhang, Y.W., and G.F. Vande Woude. In press. Mig-6, signal transduction, stress response, and cancer. Cell Cycle.<br />

Sawada, Kenjiro, A. Reza Radjabi, Nariyoshi Shinomiya, Emily Kistner, Hilary Kenny, Amy R. Becker, Muge A. Turkyilmaz, Ravi<br />

Salgia, S. Diane Yamada, George F. Vande Woude, Maria S. Tretiakova, and Ernst Lengyel. <strong>2007</strong>. c-Met overexpression is<br />

a prognostic factor in ovarian cancer and an effective target for inhibition of peritoneal dissemination and invasion. Cancer<br />

Research 67(4): 1670–1679.<br />

Zhang, Y.-W., B. Staal, Y. Su, P. Swiatek, P. Zhao, B. Cao, J. Resau, R. Sigler, R. Bronson, and G.F. Vande Woude. <strong>2007</strong>. Evidence<br />

that MIG-6 is a tumor-suppressor gene. Oncogene 26(2): 269–276.<br />

Gherardi, Ermanno, Sara Sandin, Maxim V. Petoukhov, John Finch, Mark E. Youles, Lars-Göran Öfverstedt, Ricardo N. Miguel,<br />

Tom L. Blundell, George F. Vande Woude, Ulf Skoglund, and Dmitri I. Svergun. 2006. Structural basis of hepatocyte growth<br />

factor/scatter factor and MET signalling. Proceedings of the National Academy of Sciences U.S.A. 103(11): 4046–4051.<br />

Lee, Jae-Ho, Chong Feng Gao, Chong Chou Lee, Myung Deok Kim, and George F. Vande Woude. 2006. An alternatively spliced<br />

form of Met receptor is tumorigenic. Experimental and Molecular Medicine 38(5): 565–573.<br />

Moshitch-Moshkovitz, Sharon, Galia Tsarfaty, Dafna W. Kaufman, Gideon Y. Stein, Keren Shichrur, Eddy Solomon, Robert H. Sigler,<br />

James H. Resau, George F. Vande Woude, and Ilan Tsarfaty. 2006. In vivo direct molecular imaging of early tumorigenesis and<br />

malignant progression induced by transgenic expression of GFP-Met. Neoplasia 8(5): 353–363.<br />

Tsarfaty, Galia, Gideon Y. Stein, Sharon Moshitch-Moshkovitz, Dafna W. Kaufman, Brian Cao, James H. Resau, George F. Vande<br />

Woude, and Ilan Tsarfaty. 2006. HGF/SF increases tumor blood volume: a novel tool for the in vivo functional molecular imaging<br />

of Met. Neoplasia 8(5): 344–352.


Van Andel Research Institute | <strong>Scientific</strong> <strong>Report</strong><br />

Craig P. Webb, Ph.D.<br />

Program for Translational Medicine<br />

Laboratory of Tumor Metastasis and Angiogenesis<br />

70<br />

Dr. Webb received his Ph.D. in cell biology from the University of East Anglia, England, in 1995. He then<br />

served as a postdoctoral fellow in the laboratory of George Vande Woude in the Molecular Oncology<br />

Section of the Advanced BioScience Laboratories–Basic Research Program at the National Cancer<br />

Institute, Frederick Cancer Research and Development Center, Maryland (1995–1999). Dr. Webb joined<br />

VARI as a <strong>Scientific</strong> Investigator in October 1999.<br />

Staff<br />

Laboratory Staff<br />

Students<br />

Visiting Scentists<br />

Visiting Scientists<br />

Student<br />

David Cherba, Ph.D.<br />

Jessica Hessler, PhD.<br />

Jeremy Miller, Ph.D.<br />

David Monsma, Ph.D.<br />

Emily Eugster, M.S.<br />

Sujata Srikanth, M.Phil.<br />

Dawna Dylewski, B.S.<br />

Brian Hillary, B.A.<br />

Marcy Ross, B.S.<br />

Stephanie Scott, B.S.<br />

Katherine Koehler<br />

Philip Grimley, M.D.<br />

David Reinhold, Ph.D.<br />

Guenther Tusch, Ph.D.<br />

Molly Dobb


VARI | <strong>2007</strong><br />

Research Interests<br />

The Program for Translational Medicine was launched on June 1, 2006. While maintaining a research effort focused on enhancing<br />

our understanding of the molecular basis of tumor metastasis, the program is also developing community capabilities around<br />

translational research and the future clinical applications of molecular-based medicine. These efforts are very much focused on<br />

the practical implementation of biomarkers for improving diagnostic and therapeutic strategies against chronic human diseases,<br />

including cancer. The program has recently launched a proof-of-concept personalized medicine initiative to identify novel<br />

treatment options for patients with late-stage cancer. The research portion of this community protocol includes the enhancement<br />

of computer-based predictive models, by overlaying knowledge of molecular networks and drug-target interactions to identify<br />

potential combination targeting strategies for late-stage disease. These predictions are evaluated for efficacy in xenograft models<br />

for each patient’s tumor. Our informatics system, XB-BioIntegration Suite (XB-BIS), has also been enhanced to permit reporting of<br />

molecular drug information back to the medical oncologists, who may use the information for treatment decisions. In the coming<br />

years, we plan on expanding our molecular profiling efforts to identify drugable targets within the cancer stem cell components of<br />

several malignancies and to improve our predictive modeling and reporting capabilities, with the hope of identifying the optimal<br />

combinational strategies for treating cancers using FDA-approved agents and/or drugs in the drug discovery pipeline.<br />

Our research typically begins with the analysis of human specimens. We aim to identify molecular correlates of important clinical<br />

phenotypes, such as the propensity to metastasize and drug resistance. The standardized collection of human specimens,<br />

along with information about the patient’s medical history, diagnosis, treatment, and response to therapy, represents a crucial<br />

component of our research. Identifying the molecular correlates of a given phenotype, whether nucleic acid or protein, often<br />

represents the first step in our translational pipeline. We have developed the essential workflow and integrated informatics that<br />

are required to manage and interpret complex data sets of longitudinal clinical/preclinical and molecular data across different<br />

experimental platforms. XB-BIS is now interfaced with the electronic medical records system of Spectrum Health, through the<br />

co-development of an IRB data exchange portal maintained by the Spectrum Health Research Department. This permits the<br />

transfer of de-identified medical record information from consenting patients into XB-BIS so that it can be combined with molecular<br />

data generated from the processing of the patient’s tissue, blood, or urine. In 2006, XB-BIS was commercialized and has been<br />

licensed by XB-TransMed Solutions (http://www.xbtransmed.com), who now provide professional support services related to the<br />

sales and support of the tool, while our laboratory maintains focus on XB-BIS research and development.<br />

71<br />

In the research lab, and increasingly within the Center for Molecular Medicine, we use various molecular technologies to generate<br />

the molecular data pertaining to a clinical or preclinical sample. XB-BIS permits the analysis of these data in conjunction with<br />

the clinical/preclinical information, and coupled with systems biology tools such as GeneGo’s MetaCore TM product or Ingenuity’s<br />

IPA suite, we identify potential diagnostic signatures that can predict clinically meaningful phenotypes. For example, using<br />

Affymetrix gene expression analysis, we have identified tumor profiles associated with metastatic outcome in colorectal cancer<br />

and with patient survival in mesothelioma. These signatures are now being validated within the CLIA/CAP-accredited Center for<br />

Molecular Medicine, a joint venture between VAI and Spectrum Health.<br />

Potential therapeutic intervention strategies are also identified and validated in the laboratory using a variety of approaches<br />

including RNA interference and/or existing therapeutic agents in the appropriate model systems. At this time, our focal diseases<br />

are pancreatic cancer and multiple myeloma. We have begun to identify potential new targets in these tumors and are using<br />

both inducible shRNA systems for gene knock-down and targeted nanoparticles to validate possible intervention strategies in<br />

mouse xenograft models developed and characterized within our laboratory. While our research is focused on discovering new<br />

diagnostic and therapeutic strategies for metastatic and refractory disease, the translational infrastructure we have developed<br />

can be applied to a broad spectrum of other diseases. The optimal therapeutic target is no longer the disease based on organ<br />

site, but rather the molecular networks driving the clinical phenotype within the disease and the individual.


Van Andel Research Institute | <strong>Scientific</strong> <strong>Report</strong><br />

Community initiatives<br />

As our research discoveries move closer to clinical application, we continue working to increase the readiness of the community<br />

to offer advances in molecular medicine. To translate our discoveries into human benefit, we must work in highly coordinated,<br />

multidisciplinary partnerships with community institutions. The synergistic goals are to benefit human health and promote Grand<br />

Rapids as a leader in translational medicine. The combination of powerful informatics, regulated diagnostics, and clinical trial<br />

coordination have aligned our collective community strengths with industrial demand and the FDA’s critical path intitiative.<br />

The following initiatives were successfully launched in 2006.<br />

• Innovative Clinical Research Alliance. Under the name “ClinXus”, this is a multi-institutional alliance that offers new<br />

biomarker-driven clinical trials to patients and physicians, and it will provide a single destination for pharmaceutical and<br />

biotech companies looking to carry out biomarker-drug co-development. In 2006, ClinXus obtained a $1.5 million state<br />

grant to accelerate the development of this community alliance. Partner institutions include VARI, Spectrum Health, Saint<br />

Mary’s Health Care, Jasper Clinic, Grand Valley Medical Specialists, and Grand Valley State University. Other members<br />

will join in <strong>2007</strong> as we continue to expand our collective capabilites for innovative clinical research. More information can<br />

be found at http://www.clinxus.com.<br />

72<br />

• The Center for Molecular Medicine. The CMM is a joint venture with Spectrum Health that is bringing cutting-edge,<br />

molecular-based diagnostic tests to physicians and their patients. It can offer a broad range of molecular services<br />

and has recently been certified by the College of American Pathology to run molecular diagnostic tests, including the<br />

Roche AmpliChip cytochrome p450 test indicating the correct dose for many prescription drugs. More information can<br />

be found at http://www.cmmdx.org.<br />

From left: Scott, Dobb, Hessler, Hillary, Koehler, Monsma, Cherba,<br />

Reinhold, Dylewski, Srikanth, Ross, Eugster, Webb


VARI | <strong>2007</strong><br />

External Collaborators<br />

Academic Surgical Associates, Grand Rapids, Michigan<br />

Barbara Ann Karmanos Institute, Detroit, Michigan<br />

Cancer & Hematology Centers of Western Michigan, P.C., Grand Rapids<br />

DeVos Children’s Hospital, Grand Rapids, Michigan<br />

Digestive Disease Institute, Grand Rapids, Michigan<br />

GeneGo, Inc., St. Joseph, Michigan<br />

Grand Valley Medical Specialists, Grand Rapids, Michigan<br />

Grand Valley State University, Grand Rapids, Michigan<br />

Henry Ford Hospital, Detroit, Michigan<br />

Jasper Clinical Research & Development, Inc., Kalamazoo, Michigan<br />

Johns Hopkins University, Baltimore, Maryland<br />

M.D. Anderson Cancer Center, Houston, Texas<br />

MMPC, Grand Rapids, Michigan<br />

New York University, New York City<br />

Oncology Care Associates, St. Joseph, Michigan<br />

Pfizer (Ann Arbor, Michigan; Saint Louis, Missouri; Groton, Connecticut)<br />

ProNAi Therapeutics, Kalamazoo, Michigan<br />

Saint Mary’s Health Care, Grand Rapids, Michigan<br />

Schering-Plough Research Institute, New Jersey<br />

Spectrum Health, Grand Rapids, Michigan<br />

TGEN, Phoenix, Arizona<br />

Uniformed Services University of the Health Sciences, Bethesda, Maryland<br />

University of Michigan, Ann Arbor<br />

University of California, San Francisco<br />

West Michigan Heart, Grand Rapids, Michigan<br />

73<br />

Recent Publications<br />

Kuick, Rork, David E. Misek, David J. Monsma, Craig P. Webb, Hong Wang, Kelli J. Peterson, Michael Pisano, Gilbert S. Omenn,<br />

and Samir M. Hanash. <strong>2007</strong>. Discovery of cancer biomarkers through the use of mouse models. Cancer Letters 249(1): 40–48.


Van Andel Research Institute | <strong>Scientific</strong> <strong>Report</strong><br />

Michael Weinreich, Ph.D.<br />

Laboratory of Chromosome Replication<br />

74<br />

Dr. Weinreich received his Ph.D. in biochemistry from the University of Wisconsin–Madison in 1993.<br />

He then was a postdoctoral fellow in the laboratory of Bruce Stillman, director of the Cold Spring<br />

Harbor Laboratory, New York, from 1993 to 2000. Dr. Weinreich joined VARI as a <strong>Scientific</strong> Investigator<br />

in March 2000.<br />

Staff<br />

Dorine Savreux, Ph.D.<br />

FuJung Chang, M.S.<br />

Amber Crampton, B.Sc.<br />

Carrie Gabrielse, B.S.<br />

Vickie Harkins, B.S.<br />

Students<br />

Ying-Chou Chen, M.S.<br />

Charles Miller, B.S.<br />

David Dornboss, Jr.<br />

Louise Haste<br />

Kate Leese


VARI | <strong>2007</strong><br />

Research Interests<br />

We are studying how cells accurately replicate their DNA, a process that begins at specific DNA sequences termed replication<br />

origins. There are approximately 400 replication origins in budding yeast and as many as 10,000 in human cells. Coordinating<br />

the activation of these origins for DNA synthesis during the cell cycle is a daunting task. We know that origins recruit many<br />

proteins prior to the DNA synthetic period (S-phase) that are required for the assembly and activation of replication forks. These<br />

proteins include Cdt1p, Cdc6p, and the origin recognition complex (ORC), which binds directly to origin DNA. Cdt1p, Cdc6p,<br />

and ORC cooperate to load the MCM DNA helicase at the origin in an ATP-dependent reaction. There are perhaps a score of<br />

additional proteins that assemble at the origin following MCM loading before DNA synthesis can begin. In our lab we are studying<br />

how Cdc6p activity is influenced by chromatin structure and ATP binding.<br />

We previously isolated genetic suppressors of a cdc6-4 temperature-sensitive (ts) mutant that inactivated the SIR2 gene. Sir2p is<br />

a histone H3 and H4 deacetylase, and therefore its loss leads to increased H3 and H4 acetylation within chromatin. Although loss<br />

of SIR2 allowed growth of the cdc6-4 strain at high temperature, we have found that Sir2p inhibits only specific origins. We have<br />

systematically identified multiple SIR2-regulated origins on chromosomes III and VI. Our studies so far indicate that these origins<br />

share a common organization including an inhibitory element through which Sir2p acts. Origins in Saccharomyces cerevisiae<br />

have a modular structure (Fig. 1) that includes an ORC binding site (A and B1 elements) and a loading site for the MCM helicase<br />

(B2 element). We have identified an inhibitory sequence in SIR2-regulated origins, termed the I S element, located downstream<br />

of B2. This element is responsible for Sir2p inhibition at these origins. Recent high-resolution mapping along chromosome<br />

III indicates that the I S element maps squarely within a positioned nucleosome. This nucleosome is directly adjacent to or<br />

overlaps the B2 element and therefore might influence MCM helicase loading. In support of this, we have found that<br />

excluding this nucleosome from the B2 element abolishes the activity of the I S element. Furthermore, the I S element acts in a<br />

distance-dependent manner, which is consistent with an effect through this positioned nucleosome.<br />

75<br />

Figure 1.<br />

Figure 1.<br />

S. cerevisiae origins have a modular structure consisting of an<br />

essential A element and important B elements. These elements<br />

direct binding of proteins in the “pre-replicative complex” (pre-RC)<br />

that forms during G1 phase. An inhibitory element (I S ) is present<br />

at some origins and likely interferes with pre-RC assembly.


Van Andel Research Institute | <strong>Scientific</strong> <strong>Report</strong><br />

How might SIR2 inhibit DNA replication? We believe that this occurs through deacetylation of histone H4 K16. Sir2p deacetylates<br />

the histone H3 acetyl-lysine residues K9 and K14 as well as H4 K16. We found that an H4 mutation of K16 to Q that mimics<br />

the acetylated state suppresses the cdc6-4 and mcm2-1 ts mutations; H3 K9Q or K14Q mutations do not suppress these ts<br />

mutations. Taken together, our data suggest that a local nucleosome acetylated on H4 K16 facilitates MCM helicase loading and<br />

that a nucleosome impinging on B2, if it is deacetylated on K16, inhibits MCM loading. We would like to understand the molecular<br />

function of the I S element, which is presumably affecting this histone H4 modification. Based on the frequency of SIR2-regulated<br />

origins on chromosome III and VI, we expect that a significant number of origins (about 80 of 400) will be subject to this type of<br />

regulation.<br />

76<br />

The Cdc7p-Dbf4p kinase promotes DNA replication and assists in repair of certain DNA lesions. Cdc7p-Dbf4p is a two-subunit<br />

serine/threonine kinase required for initiating DNA replication, and it acts after assembly of the MCM helicase as diagramed in<br />

Fig 2. Cdc7p is the kinase subunit but it has no activity in the absence of the Dbf4p regulatory subunit. We have analyzed Dbf4p<br />

using a structure-function approach to determine the residues required for its essential role in DNA replication. We found that<br />

about 40% of the Dbf4p N-terminus is dispensable for its essential replication function, but that it encodes a conserved 100-<br />

amino-acid region with similarity to the BRCT motif. We have called this sequence the BRDF motif for BRCT and DBF4 similarity.<br />

The BRCT domain folds into a modular structure and is often found in proteins that participate in the DNA damage response. The<br />

BRCT domain likely binds to phosphoproteins and therefore allows regulated targeting to proteins modified by phosphorylation,<br />

as occurs following activation of the DNA damage checkpoint. Yeast dbf4 mutants altering this motif are sensitive to replication<br />

fork arrest, suggesting that the BRDF domain targets the kinase to stalled replication forks (Fig. 3). In support of this interpretation,<br />

we have performed domain-swapping experiments and identified a heterologous BRCT domain that will function in place of<br />

the Dbf4p BRDF domain. We are testing whether these two domains target Dbf4p to the same or similar substrates. It appears<br />

therefore, that the Dbf4p BRDF motif has a role in maintaining replication fork stability, likely through targeting Cdc7p kinase to<br />

non-origin sites. This is a separable activity to the essential role of Dbf4p in promoting the initiation of replication.<br />

Figure 2.<br />

Figure 2.<br />

DNA synthesis requires Cdc7p-Dbf4p kinase, which is thought to act on the pre-RC to<br />

promote Cdc45p and GINS binding. Assembly of a “pre-initiation complex” facilitates<br />

origin unwinding to give ssDNA.<br />

Figure 3.<br />

A.<br />

Figure 3.<br />

A) Schematic representation showing elements<br />

conserved among all Dbf4p orthologs.<br />

The N-terminal BRDF domain is dispensable<br />

for DNA replication. B) We propose this<br />

BRCT-like domain directs Cdc7p-Dbf4p kinase<br />

to stalled replication forks via recognition of<br />

a phosphorylated protein in the replisome.<br />

B.


VARI | <strong>2007</strong><br />

We are also studying the human Cdc7-Dbf4 protein kinase, called here HsCdc7-Dbf4. The HsCdc7 protein is up-regulated in<br />

about 50% of the NCI 60 tumor cell lines representing the most common forms of cancer in the USA. In contrast, HsCdc7 protein<br />

has very low abundance or is undetectable in normal cells and tissues. It may be that nondividing cells down-regulate HsCdc7<br />

expression. We have further determined by immunohistochemistry that HsCdc7 protein is highly expressed in some primary<br />

human tumors. Since HsCdc7 is an essential kinase required for DNA replication, its increased expression level in some tumors<br />

and tumor cell lines may reflect higher rates of cellular proliferation. Alternatively, since HsCdc7 is involved in other aspects<br />

of chromosome metabolism (e.g., DNA repair) and functions in the S-phase checkpoint, its increased expression may offer an<br />

advantage to tumor cells that have higher rates of chromosome instability.<br />

It was therefore interesting when we discovered several years ago that knockdown of HsCdc7 expression using RNAi results in<br />

an apoptotic response in some cancer cell lines but not in normal cells. We have been examining the molecular differences for<br />

this apoptotic response. Apoptosis occurs in cells lines that are either p53 wild type or phenotypically null for p53. There is good<br />

published evidence that in response to HsCdc7 depletion, wild-type cells undergo a G1 and G2/M arrest that is p53-dependent<br />

and protects against apoptosis. However, in some cancer cell lines, even in the absence of p53 function, HsCdc7 knockdown<br />

does not induce apoptosis, although these cells are otherwise competent to undergo the apoptotic program in response to various<br />

stimuli. Since HsCdc7 is required for DNA replication and apparently plays a role in other aspects of chromosome metabolism,<br />

we think that these findings have significance for inhibiting the growth and/or viability of certain types of tumor cells.<br />

77<br />

Recent Publications<br />

From left: Chen, Miller, Savreux, Weinreich, Crampton, Gabrielse, Chang, Haste<br />

Gabrielse, Carrie, Charles T. Miller, Kristopher H. McConnell, Aaron DeWard, Catherine A. Fox, and Michael Weinreich. 2006.<br />

A Dbf4p BRCA1 C-terminal-like domain required for the response to replication fork arrest in budding yeast. Genetics 173(2):<br />

541–555.<br />

Zhang, Chun, Dong Kong, Min-Han Tan, Donald L. Pappas, Jr., Peng-Fei Wang, Jindong Chen, Leslie Farber, Nian Zhang,<br />

Han-Mo Koo, Michael Weinreich, Bart O. Williams, and Bin Tean Teh. 2006. Parafibromin inhibits cancer cell growth and causes<br />

G1 phase arrest. Biochemical and Biophysical Research Communications 350(1): 17–24.


Van Andel Research Institute | <strong>Scientific</strong> <strong>Report</strong><br />

Bart O. Williams, Ph.D.<br />

Laboratory of Cell Signaling and Carcinogenesis<br />

78<br />

Dr. Williams received his Ph.D. in biology from Massachusetts Institute of Technology in 1996. For three<br />

years, he was a postdoctoral fellow at the National Institutes of Health in the laboratory of Harold<br />

Varmus, former Director of NIH. Dr. Williams joined VARI as a <strong>Scientific</strong> Investigator in July 1999 and was<br />

promoted to Senior <strong>Scientific</strong> Investigator in 2006.<br />

Staff<br />

Charlotta Lindvall, M.D., Ph.D.<br />

Dan Robinson, Ph.D.<br />

Cassandra Zylstra, B.S.<br />

Students<br />

Sarah Mange<br />

Amanda Field


VARI | <strong>2007</strong><br />

Research Interests<br />

Our laboratory is interested in understanding how alterations in the Wnt signaling pathway cause human disease. Specifically,<br />

we have focused our efforts on the functions of the Wnt co-receptors, Lrp5 and Lrp6. Wnt signaling is an evolutionarily conserved<br />

process that functions in the differentiation of most tissues within the body. Given its central role in growth and differentiation, it<br />

is not surprising that alterations in the pathway are among the most common events associated with human cancer. In addition,<br />

several other human diseases, including osteoporosis, have been linked to altered regulation of this pathway.<br />

We also work on understanding the role of Wnt signaling in bone formation. Our interest is not only from the perspective of normal<br />

bone development, but also in trying to understand whether aberrant Wnt signaling plays a role in the predisposition of some<br />

common tumor types (for example, prostate, breast, lung, and renal tumors) to metastasize to and grow in bone. The long-term<br />

goal of this work is to provide insights that could be used in developing strategies to lessen the morbidity and mortality associated<br />

with skeletal metastasis.<br />

Wnt signaling in normal bone development<br />

79<br />

Mutations in the Wnt receptor, Lrp5, have been causally linked to alterations in human bone development. We have characterized<br />

a mouse strain deficient for Lrp5 and shown that it recapitulates the low-bone-density phenotype seen in human patients deficient<br />

for Lrp5. We have furthered this study by showing that mice carrying mutations in both Lrp5 and the related Lrp6 protein have<br />

even more-severe defects in bone density.<br />

To test whether Lrp5 deficiency causes changes in bone density due to aberrant signaling through β-catenin, we created mice<br />

carrying an osteoblast-specific deletion of β-catenin (OC-cre;β-catenin-flox/flox mice). In collaboration with Tom Clemens of the<br />

University of Alabama at Birmingham, we found that alterations in Wnt/β-catenin signaling in osteoblasts lead to changes in the<br />

expression of RANKL and osteoprotegerin (OPG). Consistent with this, histomorphometric evaluation of bone in the mice with<br />

osteoblast-specific deletions of either Apc or β-catenin revealed significant alterations in osteoclastogenesis.<br />

We are currently addressing how other genetic alterations linked to Wnt/β-catenin signaling affect bone development and osteoblast<br />

function. We have generated mice with a conditional allele of Lrp6 that can be inactivated via cre-mediated recombination, and<br />

we will assess the role of Lrp6 in terminal osteoblast differentiation. We are also generating mice carrying a conditional deletion<br />

of Lrp5 in differentiated osteoblasts, and we will characterize their phenotype. Finally, we are working to determine what other<br />

signaling pathways in osteoblasts may impinge on β-catenin signaling to control osteoblast differentiation and function.<br />

General mechanisms of Wnt signaling<br />

There are many levels of regulating the reception of Wnt signals. The completion of the Human Genome Project has shown<br />

that there are 19 different genes encoding Wnt proteins, 9 encoding Frizzled proteins, and the genes encoding Lrp5 and Lrp6.<br />

In addition, there are several proteins that can inhibit Wnt signaling by binding to components of the receptor complex and<br />

interfering with normal signaling, including the Dickkopfs (Dkks) and the Frizzled-related proteins (FRPs). One of the long-term<br />

goals of our laboratory is to understand how specificity is generated for the different signaling pathways, with a specific focus on<br />

understanding the molecular functions of Lrp5 and Lrp6.


Van Andel Research Institute | <strong>Scientific</strong> <strong>Report</strong><br />

Wnt signaling in prostate development and cancer<br />

Two hallmarks of advanced prostate cancer are the development of skeletal osteoblastic metastasis and the ability of the tumor<br />

cells to become independent of androgen for survival. The association of Wnt signaling with bone growth, plus the fact that<br />

β-catenin can bind to the androgen receptor and make it more susceptible to activation with steroid hormones other than DHT,<br />

make Wnt signaling an attractive candidate for explaining some phenotypes associated with advanced prostate cancer. We<br />

have created mice with a prostate-specific deletion of the Apc gene. These mice develop fully penetrant prostate hyperplasia by<br />

four months of age, and these tumors progress to frank carcinomas by seven months. We have found that these tumors initially<br />

regress under androgen ablation but show signs of androgen-independent growth some months later.<br />

Wnt signaling in mammary development and cancer<br />

We are also addressing the relative roles of Lrp5 and Lrp6 in Wnt1-induced mammary carcinogenesis. A deficiency in Lrp5<br />

dramatically inhibits the development of mammary tumors in this context. A germline deficiency for Lrp5 or Lrp6 results in<br />

delayed mammary development. As Lrp5-deficient mice are viable and fertile, we have focused our initial efforts on these mice.<br />

In collaboration with Caroline Alexander’s laboratory, we have found dramatic reductions in the number of mammary progenitor<br />

cells in these mice. We are continuing to examine the mechanisms underlying this reduction.<br />

80<br />

VARI mutant mouse repository<br />

With support from the Van Andel Institute, my laboratory maintains a repository of mutant mouse strains to support the general<br />

development of animal models of human disease. We distribute these strains at a nominal cost to interested laboratories.<br />

External Collaborators<br />

Bone development<br />

Mary Bouxsein, Beth Israel Deaconness Medical Center, Boston, Massachusetts<br />

Thomas Clemens, University of Alabama–Birmingham<br />

Marie Claude Faugere, University of Kentucky, Lexington<br />

David Ornitz and Fanxin Long, Washington University, St. Louis, Missouri<br />

Matthew Warman, Harvard University, Boston, Massachusetts<br />

Prostate cancer<br />

Wade Bushman and Ruth Sullivan, University of Wisconsin–Madison<br />

Mammary development<br />

Caroline Alexander, University of Wisconsin–Madison<br />

Yi Li, Baylor Breast Center, Houston, Texas<br />

Jeffrey Rubin, National Cancer Institute, Bethesda, Maryland<br />

Mechanisms of Wnt signaling<br />

Kathleen Cho, University of Michigan, Ann Arbor<br />

Kang-Yell Choi, Yansei University, Seoul, South Korea<br />

Eric Fearon, University of Michigan, Ann Arbor<br />

Silvio Gutkind, National Institute of Dental and Craniofacial Research, Bethesda, Maryland<br />

Kun-Liang Guan, University of Michigan, Ann Arbor


VARI | <strong>2007</strong><br />

From left, standing: Williams, Robinson; seated: Zylstra, Lindvall<br />

Recent Publications<br />

Lindvall, C., W. Bu, B.O. Williams, and Y. Li. In press. Wnt signaling, stem cells, and the cellular origin of breast cancer.<br />

Stem Cell Reviews.<br />

Wu, R., N.D. Handrix, R. Kuick, Y. Zhai, D.R. Schwartz, A. Akyol, S. Hanash, D. Misek, H. Katabuchi, B.O. Williams, E.R. Fearon,<br />

and K.R. Cho. In press. Mouse model of human endometroid adenocarcinoma based on somatic defects in the Wnt β-catenin<br />

and PI3K/Pten signaling pathways. Cancer Cell.<br />

81<br />

Young, J.J., J.L. Bromberg-White, C.R. Zylstra, J. Church, E. Boguslawski, J. Resau, B.O. Williams, and N. Duesbery. In<br />

press. LRP5 and LRP6 are not required for protective antigen-mediated internalization or lethality of anthrax lethal toxin.<br />

PLoS Pathogens.<br />

Bruxvoort, Katia J., Holli M. Charbonneau, Troy A. Giambernardi, James C. Goolsby, Chao-Nan Qian, Cassandra R. Zylstra,<br />

Daniel R. Robinson, Pradip Roy-Burman, Aubie K. Shaw, Bree D. Buckner-Berghuis, Robert E. Sigler, James H. Resau, Ruth<br />

Sullivan, Wade Bushman, and Bart O. Williams. <strong>2007</strong>. Inactivation of Apc in the mouse prostate causes prostate carcinoma.<br />

Cancer Research 67(6): 2490–2496.<br />

Liu, X., K.M. Bruxvoort, Cassandra R. Zylstra, J. Liu, R. Cichowski, Marie-Claude Faugere, Mary L. Bouxsein, C. Wan, Bart O.<br />

Williams, and Thomas L. Clemens. <strong>2007</strong>. Lifelong accumulation of bone in mice lacking Pten in osteoblasts. Proceedings of the<br />

National Academy of Sciences U.S.A. 104(7): 2259–2264.<br />

Inoki, Ken, Hongjiao Ouyang, Tianqing Zhu, Charlotta Lindvall, Yian Wang, Xiaojie Zhang, Qian Yang, Christina Bennett, Yoku<br />

Harada, Kryn Stankunas, Cun-yu Wang, Xi He, Ormond A. MacDougald, Ming You, Bart O. Williams, and Kun-Liang Guan. 2006.<br />

TSC2 integrates Wnt and energy signals via a coordinated phosphorylation by AMPK and GSK3 to regulate cell growth. Cell<br />

126(5): 955–968.<br />

Lindvall, Charlotta, Nicole C. Evans, Cassandra R. Zylstra, Yi Li, Caroline M. Alexander, and Bart O. Williams. 2006. The Wnt<br />

signaling receptor LRP5 is required for mammary ductal stem cell activity and Wnt1-induced tumorigenesis. Journal of Biological<br />

Chemistry 281(46): 35081–35087.<br />

Zhang, Chun, Dong Kong, Min-Han Tan, Donald L. Pappas, Jr., Peng-Fei Wang, Jindong Chen, Leslie Farber, Nian Zhang,<br />

Han-Mo Koo, Michael Weinreich, Bart O. Williams, and Bin Tean Teh. 2006. Parafibromin inhibits cancer cell growth and causes<br />

G1 phase arrest. Biochemical and Biophysical Research Communications 350(1): 17–24.


Van Andel Research Institute | <strong>Scientific</strong> <strong>Report</strong><br />

82<br />

Photo taken by Veronique Schulz of the Mirant lab.


VARI | <strong>2007</strong><br />

Human melanoma cells.<br />

83<br />

Human melanoma cells were fixed and stained to show the nuclei (blue), actin stress fibers (red), and focal adhesions (green dots).


Van Andel Research Institute | <strong>Scientific</strong> <strong>Report</strong><br />

H. Eric Xu, Ph.D.<br />

Laboratory of Structural Sciences<br />

84<br />

Dr. Xu went to Duke University and the University of Texas Southwestern Medical Center, where he<br />

earned his Ph.D. in molecular biology and biochemistry. Following a postdoctoral fellowship with Carl<br />

Pabo at MIT, he moved to GlaxoWellcome in 1996 as a research investigator of nuclear receptor drug<br />

discovery. Dr. Xu joined VARI as a Senior <strong>Scientific</strong> Investigator in July 2002 and was promoted to<br />

Distinguished <strong>Scientific</strong> Investigator in March <strong>2007</strong>.<br />

Staff<br />

Laboratory Staff<br />

Jiyuan Ke, Ph.D.<br />

Schoen Kruse, Ph.D.<br />

Augie Pioszak, Ph.D.<br />

David Tolbert, Ph.D.<br />

Yong Xu, Ph.D.<br />

Students<br />

Chenghai Zhang, Ph.D.<br />

X. Edward Zhou, Ph.D.<br />

Jennifer Daugherty, B.S.<br />

Amanda Kovach, B.S.<br />

Kelly Powell, B.S.<br />

Visiting Scientist<br />

Visiting Scientists<br />

Ross Reynolds, Ph.D.


VARI | <strong>2007</strong><br />

Research Interests<br />

Our laboratory is employing multidisciplinary approaches to study the structures and functions of protein complexes that play key<br />

roles in major signaling pathways, and to use the resulting structural information to develop therapeutic agents for the treatment<br />

of human disease, including cancer and diabetes. Currently we are focusing on three families of proteins: nuclear hormone<br />

receptors, the Met tyrosine kinase receptor, and G protein–coupled receptors, because these proteins, beyond their fundamental<br />

roles in biology, are important drug targets for many human diseases.<br />

Nuclear hormone receptors<br />

The nuclear hormone receptors form a large family comprising ligand-regulated and DNA-binding transcriptional factors. The<br />

family includes receptors for classic steroid hormones such as estrogen, progesterone, androgens, and glucocorticoids, as well<br />

as receptors for peroxisome proliferator activators, vitamin D, vitamin A, and thyroid hormones. One distinguishing fact about<br />

these classic receptors is that they are among the most successful targets in the history of drug discovery: every receptor has<br />

one or more cognate synthetic ligands currently being used as medicines. The nuclear receptors also include a class of “orphan”<br />

receptors for which no ligand has been identified. In the last two years, we have developed the following projects centering on the<br />

structural biology of nuclear receptors.<br />

85<br />

Peroxisome proliferator–activated receptors<br />

The peroxisome proliferator–activated receptors (PPARα, δ, and γ) are key regulators of glucose and fatty acid homeostasis and<br />

as such are important therapeutic targets for treating cardiovascular disease, diabetes, and cancer. To understand the molecular<br />

basis of ligand-mediated signaling by PPARs, we have determined crystal structures of each PPAR’s ligand-binding domain<br />

(LBD) bound to diverse ligands including fatty acids, the lipid-lowering fibrate drugs, and a new generation of anti-diabetic drugs,<br />

the glitazones. We have also determined the crystal structures of these receptors bound to coactivators or co-repressors. We<br />

are developing this project into the structures of large PPAR fragment/DNA complexes.<br />

Human glucocorticoid and mineralocorticoid receptors<br />

The human glucocorticoid receptor (GR) and mineralocorticoid receptor (MR) are classic steroid hormone receptors that are key<br />

to a wide spectrum of human physiology including immune/inflammatory responses, metabolic homeostasis, and control of blood<br />

pressure. Both are well-established drug targets. GR ligands such as dexamethasone (Dex) and fluticasone propionate (FP) are<br />

used to treat asthma, leukemia, and autoimmune diseases; MR ligands such as spironolactone and eplerenone are used to treat<br />

hypertension and heart failure. However, the clinical use of these ligands is limited by undesirable side effects partly associated<br />

with their receptor cross-reactivity or low potency. Thus, the discovery of highly potent and more-selective ligands for GR and MR<br />

is an important goal of pharmaceutical research.<br />

We have determined a crystal structure of the GR LBD bound to dexamethasone and the MR LBD bound to corticosterone, both<br />

of which are in complex with a coactivator peptide motif. These structures provide a detailed basis for the specificity of hormone<br />

recognition and coactivator assembly by GR and MR. Currently we are studying receptor-ligand interactions by crystallizing GR<br />

and MR with various steroid or nonsteroid ligands. In collaboration with Brad Thompson and Raj Kumar at the University of Texas<br />

Medical Branch at Galveston, we are also extending our studies to the structure of a large GR fragment bound to DNA.


Van Andel Research Institute | <strong>Scientific</strong> <strong>Report</strong><br />

The human androgen receptor<br />

The androgen receptor (AR) is the central molecule in the development and progression of prostate cancer, and as such it serves<br />

as the molecular target of anti-androgen therapy. However, most prostate cancer patients develop resistance to such therapy,<br />

mainly due to mutations in this hormone receptor that alter its three-dimensional structure and allow AR to escape repression.<br />

The growth of prostate cancer cells that harbor a mutated AR is then no longer dependent on androgen, making anti-hormone<br />

therapy ineffective. This form of hormone-independent prostate cancer is highly aggressive and is responsible for most deaths<br />

from prostate cancer. The development of effective therapies requires a detailed understanding of the structure and functions<br />

of the central molecule, i.e., the androgen receptor and its interactions with hormones and co-regulators. In this project, we are<br />

aiming to determine the structures of the mutated AR proteins that alter the response to anti-hormone therapy. In collaboration<br />

with Donald MacDonnell at Duke University, we are working on the crystal structure of the full-length AR/DNA complex.<br />

Structural genomics of nuclear receptor ligand-binding domains<br />

86<br />

The LBDs of nuclear receptors contain key structural elements that mediate ligand-dependent regulation of these receptors,<br />

and as such, LBDs have been the focus of intense structural studies. There are only a few orphan nuclear receptors for which<br />

the LBD structure remains unsolved. In the past two years, we have focused on structural characterization of two orphan<br />

receptors: constitutive androstane receptor (CAR) and steroidogenic factor-1 (SF-1). The CAR structure reveals a compact LBD<br />

fold containing a small pocket that is only half the size of the pocket in PXR, a receptor closely related to CAR. The constitutive<br />

activity of CAR appears to be mediated by a novel linker helix between the C-terminal AF-2 helix and helix 10. On the other<br />

hand, SF-1 is regarded as a ligand-independent receptor, but its LBD structure reveals the presence of a phospholipid ligand in a<br />

surprisingly large pocket; its size is more than twice that of the pocket in the mouse LRH-1, a closely related receptor. The bound<br />

phospholipid is readily exchanged and modulates SF-1 interactions with coactivators. Mutations designed to reduce the size<br />

of the SF-1 pocket or to disrupt hydrogen bonds formed with the phospholipid abolish SF-1/coactivator interactions and reduce<br />

SF-1 transcriptional activity. These findings establish that SF-1 is a ligand-dependent receptor and suggest an unexpected link<br />

between nuclear receptors and phospholipid signaling pathways.<br />

The Met tyrosine kinase receptor<br />

MET is a tyrosine kinase receptor that is activated by hepatocyte growth factor/scatter factor (HGF/SF). Aberrant activation of<br />

the Met receptor has been linked to the development and metastasis of many types of solid tumors and has been correlated<br />

with poor clinical prognosis. HGF/SF has a modular structure with an N-terminal domain, four kringle domains, and an inactive<br />

serine protease domain. The structure of the N-terminal domain with a single kringle domain (NK1) has been determined. Less<br />

is known about the structure of the Met extracellular domain. The molecular basis of the MET receptor–HGF/SF interaction and<br />

the activation of MET signaling by this interaction remains poorly understood. In collaboration with George Vande Woude and<br />

Ermanno Gherardi, we are developing this project to solve the crystal structure of the Met receptor/HGF complex.


VARI | <strong>2007</strong><br />

G Protein–coupled receptors<br />

G protein–coupled receptors (GPCRs) form the largest family of receptors in the human genome; they are receptors for diverse<br />

signals carried by photons, ions, small chemicals, peptides, and hormones. These receptors account for over 40% of drug<br />

targets, but the structure of these receptors remains a challenge because they are seven-transmembrane receptors. Currently,<br />

there is only one reported GPCR structure, for an inactive form of bovine rhodopsin. Many important questions regarding GPCR<br />

ligand binding and activation remain unanswered. From our standpoint, GPCRs are similar to nuclear hormone receptors with<br />

respect to regulation by protein-ligand and protein-protein interactions. Due to their importance, we have decided to take on<br />

studies of the structural basis of ligand binding in, and activation of, GPCRs.<br />

External Collaborators<br />

Doug Engel, University of Michigan, Ann Arbor<br />

Ermanno Gherardi, University of Cambridge, UK<br />

Steve Kliewer, University of Texas Southwestern Medical Center, Dallas<br />

David Mangelsdorf, University of Texas Southwestern Medical Center, Dallas<br />

Donald MacDonnell, Duke University, Durham, North Carolina<br />

Stoney Simmons, National Institutes of Health, Bethesda, Maryland<br />

Scott Thacher, Orphagen Pharmaceuticals, San Diego, California<br />

Brad Thompson and Raj Kumar, University of Texas Medical Branch at Galveston<br />

Ming-Jer Tsai, Baylor College of Medicine, Houston, Texas<br />

87<br />

From left, standing: E. Xu, Daugherty, Tolbert, Kovach, Powell, Zhang, Zhou<br />

kneeling: Kruse, Pioszak, Y. Xu, Ke<br />

Recent Publications<br />

Choi, Mihwa, Antonio Moschetta, Angie L. Bookout, Li Peng, Michihisa Umetani, Sam R. Holmstrom, Kelly Suino-Powell, H. Eric<br />

Xu, James A. Richardson, Robert D. Gerard, David J. Mangelsdorf, and Steven A. Kliewer. 2006. Identification of a hormonal<br />

basis for gallbladder filling. Nature Medicine 12(11): 1253–1235.


Van Andel Research Institute | <strong>Scientific</strong> <strong>Report</strong><br />

88<br />

2006 Van Andel Research Institute Symposium


VARI | <strong>2007</strong><br />

Winning the War against Cancer:<br />

From Genomics to Bedside and Back<br />

In September 2006, the Van Andel Research Institute honored the lifetime achievements of George F. Vande Woude with a<br />

symposium titled “Winning the War against Cancer: From Genomics to Bedside and Back”. Organized by Nicholas Duesbery,<br />

Tony Hunter, and Bin Teh, the three-day symposium featured noted speakers, including three Nobel laureates; presentation of the<br />

Daniel Nathans Award; and a reception honoring Dr. Vande Woude. More than 250 scientists attended the meeting.<br />

89<br />

Symposium photos by Jindong Chen.


Van Andel Research Institute | <strong>Scientific</strong> <strong>Report</strong><br />

Invited Speakers<br />

Jerry Adams<br />

Walter & Eliza Hall Institute<br />

Tim Hunt<br />

Clare Hall Laboratories<br />

Tony Pawson<br />

Mount Sinai Hospital Research Institute<br />

James P. Allison<br />

Memorial Sloan-Kettering Cancer Center<br />

Tony Hunter<br />

Salk Institute for Biological Studies<br />

Bruce Ponder<br />

Cancer Research U.K<br />

Anton Berns<br />

Nederlands Kanker Institute<br />

Arnold Levine<br />

Institute for Advanced Study<br />

Martine Roussel<br />

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

J. Michael Bishop<br />

University of California, San Francisco<br />

David M. Livingston<br />

Dana-Farber Cancer Institute<br />

Janet Rowley<br />

University of Chicago<br />

Joan S. Brugge<br />

Harvard Medical School<br />

James L. Maller<br />

University of Colorado School of Medicine<br />

Joseph Schlessinger<br />

Yale University School of Medicine<br />

Suzanne Cory<br />

Walter & Eliza Hall Institute<br />

Paul A. Marks<br />

Memorial Sloan-Kettering Cancer Center<br />

Phillip A. Sharp<br />

Massachusetts Institute of Technology<br />

90<br />

Michael Dean<br />

National Cancer Institute–Frederick<br />

Frank McCormick<br />

University of California, San Francisco<br />

Louis Staudt<br />

National Cancer Institute<br />

Edward Harlow<br />

Harvard Medical School<br />

William Muller<br />

McGill University<br />

Craig Thompson<br />

Abramson Family Cancer Research Institute<br />

Stephen Hughes<br />

National Cancer Institute–Frederick<br />

Morag Park<br />

McGill University<br />

George Vande Woude<br />

Van Andel Research Institute<br />

Karen Vousden<br />

Beatson Institute for Cancer Research


VARI | <strong>2007</strong><br />

91


92<br />

Van Andel Research Institute | <strong>Scientific</strong> <strong>Report</strong>


VARI | <strong>2007</strong><br />

All of us who have had the privilege of working with George over the years join in appreciation<br />

and thanks for his constant interest, wisdom, insights, and humor.<br />

93<br />

Duesbery, N.S., and B.T. Teh. <strong>2007</strong>. Cancer: biology and therapeutics—a tribute to George Vande Woude.<br />

Oncogene 26(9): 1258–1259.<br />

Teh, B.T., and N. Duesbery. <strong>2007</strong>. A tribute to George F. Vande Woude, a man of character: 2006 <strong>Scientific</strong> Symposium<br />

“Winning the War against Cancer: From Genomics to Bedside and Back.” Cancer Research 67(6): 2394–2395.


Van Andel Research Institute | <strong>Scientific</strong> <strong>Report</strong><br />

94<br />

Daniel Nathans Memorial Award


VARI | <strong>2007</strong><br />

Daniel Nathans Memorial Award<br />

The Daniel Nathans Memorial Award was established in memory of Dr. Daniel Nathans, a distinguished member of our scientific<br />

community and a founding member of VARI’s Board of <strong>Scientific</strong> Advisors. We established this award to recognize individuals<br />

who emulate Dan and his contributions to biomedical and cancer research. It is our way of thanking and honoring him for his help<br />

and guidance in bringing Jay and Betty Van Andel’s dream to reality. The Daniel Nathans Memorial Award was announced at our<br />

inaugural symposium, “Cancer & Molecular Genetics in the Twenty-First Century”, in September 2000.<br />

95<br />

Previous Award Recipients<br />

2000 Richard D. Klausner, M.D.<br />

2001 Francis S. Collins, M.D., Ph.D.<br />

2002 Lawrence H. Einhorn, M.D.<br />

2003 Robert A. Weinberg, Ph.D.<br />

2004 Brian Druker, M.D.<br />

2005 Tony Hunter, Ph.D., and Tony Pawson, Ph.D.<br />

Tony Hunter, Ph.D.<br />

Tony Pawson, Ph.D.


Van Andel Research Institute | <strong>Scientific</strong> <strong>Report</strong><br />

96<br />

Postdoctoral Fellowship Program


VARI | <strong>2007</strong><br />

Postdoctoral Fellowship Program<br />

The Van Andel Research Institute provides postdoctoral training opportunities to Ph.D. scientists beginning their research careers.<br />

The fellowships help promising scientists advance their knowledge and research experience while at the same time supporting<br />

the research endeavors of VARI. The fellowships are funded in three ways: 1) by the laboratories to which the fellow is assigned;<br />

2) by the VARI Office of the Director; or 3) by outside agencies. Each fellow is assigned to a scientific investigator who oversees<br />

the progress and direction of research. Fellows who worked in VARI laboratories in 2006 and early <strong>2007</strong> are listed below.<br />

Jennifer Bromberg-White<br />

Dan Huang<br />

Michael Shafer<br />

Penn. State University College of Medicine, Hershey<br />

VARI mentor: Nick Duesbery<br />

Peking Union Medical College, China<br />

VARI mentor: Bin Teh<br />

Michigan State University, East Lansing<br />

VARI mentor: Brian Haab<br />

Philippe Depeille<br />

Schoen Kruse<br />

Suganthi Sridhar<br />

University of Montpellier, France<br />

VARI mentor: Nicholas Duesbery<br />

University of Colorado, Boulder<br />

VARI mentor: Eric Xu<br />

Southern Illinois University, Carbondale<br />

VARI mentor: Cindy Miranti<br />

Yan Ding<br />

Brendan Looyenga<br />

Peng Fei Wang<br />

Peking Union Medical College, China<br />

VARI mentor: Nicholas Duesbery<br />

University of Michigan, Ann Arbor<br />

VARI mentor: James Resau<br />

Fourth Military Medical University, China<br />

VARI mentor: Bin Teh<br />

Mathew Edick<br />

Douglas Luccio-Camelo<br />

Yi-Mi Wu<br />

University of Tennessee, Memphis<br />

VARI mentor: Cindy Miranti<br />

University of Brazil, Rio de Janeiro<br />

VARI mentor: Bin Teh<br />

National Tsin-Hua University, Taiwan<br />

VARI mentor: Brian Haab<br />

97<br />

Kathryn Eisenmann<br />

University of Minnesota, Minneapolis<br />

VARI mentor: Arthur Alberts<br />

Daisuke Matsuda<br />

Kitasato University, Japan<br />

VARI mentor: Bin Teh<br />

Yong Xu<br />

Shanghai Institute of Materia Medica, China<br />

VARI mentor: Eric Xu<br />

Leslie Farber<br />

George Washington University, Washington, D.C.<br />

VARI mentor: Bin Teh<br />

Augen Pioszak<br />

University of Michigan, Ann Arbor<br />

VARI mentor: Eric Xu<br />

Xin Yao<br />

Tianjin Medical University, China<br />

VARI mentor: Bin Teh<br />

Kunihiko Futami<br />

Tokyo University of Fisheries, Japan<br />

VARI mentor: Bin Teh<br />

Daniel Robinson<br />

University of California, Davis<br />

VARI mentor: Bart Williams<br />

Chenghai Zhang<br />

Virus Institute of the CDC, China<br />

VARI mentor: Eric Xu<br />

Quliang Gu<br />

Dorine Savreux<br />

Xiaoyin Zhou<br />

Sun Yat-sen University of Medicine, China<br />

VARI mentor: Brian Cao<br />

Virology University, France<br />

VARI mentor: Michael Weinreich<br />

University of Alabama – Birmingham<br />

VARI mentor: Eric Xu<br />

Carrie Graveel<br />

University of Wisconsin – Madison<br />

VARI mentor: George Vande Woude<br />

Jessica Hessler<br />

University of Michigan, Ann Arbor<br />

VARI mentor: Craig Webb<br />

Holly Holman<br />

University of Glasgow, U.K.<br />

VARI mentor: Arthur Alberts


Van Andel Research Institute | <strong>Scientific</strong> <strong>Report</strong><br />

98<br />

Student Programs


VARI | <strong>2007</strong><br />

Grand Rapids Area Pre-College Engineering Program<br />

The Grand Rapids Area Pre-College Engineering Program (GRAPCEP) is administered by Davenport University and jointly<br />

sponsored and funded by Pfizer, Inc., and VARI. The program is designed to provide selected high school students, who have<br />

plans to major in science or genetic engineering in college, the opportunity to work in a research laboratory. In addition to<br />

research methods, the students also learn workplace success skills such as teamwork and leadership. The three 2006 GRAPCEP<br />

students were<br />

Alicia Coleman (Resau/Duesbery)<br />

Creston High School<br />

Megan Spencer (Holmen)<br />

Creston High School<br />

Ware-Van Brunt (Webb)<br />

Creston High School<br />

99<br />

From left: Ware-Van Brunt, Coleman, Spencer


Van Andel Research Institute | <strong>Scientific</strong> <strong>Report</strong><br />

Summer Student Internship Program<br />

The VARI student internships were established to provide college students with an opportunity to work with professional researchers<br />

in their fields of interest, to use state-of-the-art equipment and technologies, and to learn valuable people and presentation skills.<br />

At the completion of the 10-week program, the students summarize their projects in an oral presentation.<br />

From January 2006 to March <strong>2007</strong>, VARI hosted 62 students from 23 colleges and universities in formal summer internships under<br />

the Frederik and Lena Meijer Student Internship Program and in other student positions during the year. An asterisk (*) indicates<br />

a Meijer student intern.<br />

100<br />

Andrews University, Berrien Springs, Michigan<br />

Christopher Armstrong* (Xu)<br />

Aquinas College, Grand Rapids, Michigan<br />

Krysta Collins (Haab)<br />

Natalie Kent (Hay)<br />

Sara Kunz (Hay)<br />

Rebecca Trierweiler (Hay)<br />

Calvin College, Grand Rapids, Michigan<br />

David Dornboss, Jr. (Weinreich)<br />

Jonathan Dudley (Vande Woude)<br />

Amanda Field* (Williams)<br />

Alysha Kett* (Vande Woude)<br />

Geoff Kraker (MacKeigan)<br />

Kate Leese (Weinreich)<br />

Sarah Mange (Williams)<br />

Devin Mistry (Haab)<br />

Jose Toro (Hay)<br />

Bill Wondergem (Teh)<br />

Case Western Reserve University, Cleveland<br />

Elianna Bootzin* (Hay)<br />

Central Michigan University, Mount Pleasant<br />

Sarah DeVos* (Teh)<br />

Franciscan University, Steubenville, Ohio<br />

Joan Krilich* (Cavey)<br />

Grand Rapids Community College, Michigan<br />

Wei Luo (Resau)<br />

Grand Valley State University, Allendale, Michigan<br />

Angelique Berens (Vande Woude)<br />

Eric Graf (Miranti)<br />

Nick Miltgen (Resau)<br />

Gary Rajah* (Miranti)<br />

Lisa Orcasitas (Duesbery)<br />

Sara Ramirez (Resau)<br />

Brittany Stropich* (Alberts)<br />

Indiana University, Bloomington<br />

Erin Jefferson* (Webb)<br />

Kalamazoo College, Kalamazoo, Michigan<br />

Adam Granger (Holmen)<br />

Marquette University, Milwaukee, Wisconsin<br />

Michael Avallone (Teh)<br />

Miami University, Oxford, Ohio<br />

Grant Van Eerden (Resau)<br />

Michigan State University, East Lansing<br />

David Achila (Xu/Weinreich)<br />

Ying-Chou Chen, M.S. (Weinreich)<br />

Michelle Dawes (Duesbery)<br />

Aaron DeWard (Alberts)<br />

Pete Haak, B.S. (Resau)<br />

Kate Jackson (Resau)<br />

Andrew Kraus (Vande Woude)<br />

Sebla Kutluay, B.S. (Triezenberg)<br />

Chih-Shia Lee, M.S. (Duesbery)<br />

Charles Miller (Weinreich)<br />

Kara Myslivec (Resau)<br />

Katie Sian, B.S. (MacKeigan)


VARI | <strong>2007</strong><br />

2006 summer intern students<br />

Nanjing Medical University, China<br />

Xin Wang (Cao)<br />

Ning Xu (Cao)<br />

Aixia Zhang (Cao)<br />

Jin Zhu (Cao)<br />

Northern Illinois University, Dekalb<br />

Mohan Thapa (Resau)<br />

Purdue University, West Lafayette, Indiana<br />

Brent Goodman* (Furge)<br />

University of Bath, United Kingdom<br />

Naomi Asantewa-Sechereh (Duesbery)<br />

Louise Haste (Weinreich)<br />

University of Illinois, Champaign-Urbana<br />

Huong Tran (Resau)<br />

University of Mannheim, Germany<br />

Dagmar Hildebrand (Alberts)<br />

Stefan Kutscheidt (Miranti)<br />

University of Michigan, Ann Arbor<br />

Katherine Koelzer* (Swiatek)<br />

Erin Lambers (Duesbery)<br />

Jennifer Lunger* (Haab)<br />

Renee VanderLaan* (Holmen)<br />

University of North Carolina, Chapel Hill<br />

Jourdan Stuart* (Resau)<br />

University of Notre Dame, South Bend, Indiana<br />

Kristin Buzzitta (Teh)<br />

Joe Church* (Duesbery)<br />

Margaret Condit (Teh)<br />

Western Michigan University, Kalamazoo<br />

Mallory Walters (Holmen)<br />

101


Van Andel Research Institute | <strong>Scientific</strong> <strong>Report</strong><br />

102<br />

Han-Mo Koo Memorial Seminar Series


VARI | <strong>2007</strong><br />

Han-Mo Koo Memorial Seminar Series<br />

This seminar series is dedicated to the memory of Dr. Han-Mo Koo, who was a VARI <strong>Scientific</strong> Investigator from 1999 until his<br />

passing in May of 2004.<br />

January 2006<br />

W. Michael Kuehl, National Cancer Institute<br />

“Molecular pathogenesis of multiple myeloma”<br />

Kenneth Bradley, University of California, Los Angeles<br />

“Anthrax lethal toxin”<br />

Valina L. Dawson, Johns Hopkins University<br />

“Life and death signaling by PAR in the brain”<br />

Ted M. Dawson, Johns Hopkins University<br />

“Genetic clues to the mysteries of Parkinson’s disease”<br />

Andy Futreal, Wellcome Trust Sanger Institute<br />

“Surveying somatic mutations in human cancer by targeted re-sequencing”<br />

103<br />

February<br />

Morag Park, McGill University, Montreal<br />

“The Met receptor tyrosine kinase: from tubes to tumorigenesis”<br />

Nicholas J. Vogelzang, Nevada Cancer Institute<br />

“Treatment options in metastatic renal cell carcinoma: an embarrassment of riches”<br />

March<br />

Teresa L. Burgess, Amgen, Inc.<br />

“Fully human monoclonal antibodies to hepatocyte growth factor”<br />

Kenneth L. van Golen, University of Michigan<br />

“Understanding the roles of Rho and Rac GTPases in prostate cancer bone metastasis”<br />

Thomas W. Glover, University of Michigan<br />

“Mechanisms and significance of chromosome fragile site instability in cancer”


Van Andel Research Institute | <strong>Scientific</strong> <strong>Report</strong><br />

April<br />

Stephen J. O’Brien, National Cancer Institute<br />

“Genetic architecture of complex diseases: lessons from AIDS”<br />

Richard Treisman, Cancer Research, U.K.<br />

“Regulation of the SRF transcription factor via cytoskeletal and MAP kinase signaling pathways”<br />

Dean Felsher, Stanford University<br />

“Molecular and cellular basis of oncogene addiction”<br />

May<br />

Partho Ghosh, University of California, San Diego<br />

“Met as a target for bacterial intracellular invasion”<br />

Ming-Jer Tsai, Baylor College of Medicine<br />

“Role of nuclear receptor co-regulator SRC-3/AIB1 in prostate cancer”<br />

104<br />

Laura S. Schmidt, National Cancer Institute–Frederick<br />

“Understanding the genetics of kidney cancer through familial renal cancer studies”<br />

David Drubin, University of California, Berkeley<br />

“Harnessing actin dynamics for endocytic trafficking”<br />

Thomas Clemens, University of Alabama at Birmingham<br />

“Oxygen sensing and osteogenesis”<br />

June<br />

Robert J. Motzer, Memorial Sloan-Kettering Cancer Center<br />

“Targeted therapy for metastatic renal cell carcinoma”<br />

Tom Blumenthal, University of Colorado<br />

“Widespread operons in the C. elegans genome: why and how”<br />

July<br />

Rudolf Jaenisch, Whitehead Institute and Massachusetts Institute of Technology<br />

“Nuclear cloning, stem cells, and pluripotency”


VARI | <strong>2007</strong><br />

August<br />

Douglas R. Green, St. Jude Children’s Research Hospital<br />

“p53, mitochondria, and apoptosis”<br />

Gregory S. Fraley, Hope College<br />

“Food, fat, and sex: how the brain integrates energetics and reproduction”<br />

September<br />

Stephen A. Krawetz, Wayne State University<br />

“Genome reprogramming and the paternal contribution at fertilization”<br />

Hilary Koprowski, Thomas Jefferson University<br />

“Rabies at the dawn of the 21st century”<br />

October<br />

Y. Eugene Chen, University of Michigan<br />

“Nitro-lipids and PPARs in metabolic syndrome”<br />

Jacques Pouyssegur, Institute of Signaling, University of Nice<br />

“Hypoxia signaling and cancer progression”<br />

105<br />

November<br />

Chuxia Deng, National Institute of Diabetes and Digestive and Kidney Diseases<br />

“BRCA1 and tumorigenesis in animal models”<br />

Dafna Bar-Sagi, New York University<br />

“RAS signaling: new trails in familiar territory”<br />

December<br />

Kun-Liang Guan, University of Michigan<br />

“Regulation and function of the TSC-mTOR pathway”<br />

January <strong>2007</strong><br />

Moses Lee, Hope College<br />

“Regulation of the topoisomerase IIα gene using polyamides that bind to the inverted CCAAT<br />

box present in the promoter”


Van Andel Research Institute | <strong>Scientific</strong> <strong>Report</strong><br />

February<br />

Raj Kumar, University of Texas Medical Branch<br />

“Structure and functions of the steroid receptors”<br />

David Kimelman, University of Washington<br />

“Tales of tails: the importance of Bmp signaling in embryogenesis”<br />

Arthur L. Haas, Louisiana State University<br />

“ISG15 and ubiquitin as antagonistic regulators of cell transformation”<br />

March<br />

S. Stoney Simons, Jr., National Institutes of Health<br />

“A systems biology approach to steroid hormone action: towards a quantitative understanding<br />

of whole cell responses to steroid hormones”<br />

John D. Shaughnessy, Jr., University of Arkansas for Medical Science<br />

“Using genomics to better understand the biology and clinical course of multiple myeloma”<br />

106<br />

Melanie H. Cobb, University of Texas Southwestern Medical Center<br />

“MAP kinase signaling in pancreatic beta cells”


VARI | <strong>2007</strong><br />

Van Andel Research Institute Organization<br />

107


Van Andel Research Institute | <strong>Scientific</strong> <strong>Report</strong><br />

David L. Van Andel,<br />

Chairman and CEO, Van Andel Institute<br />

VARI Board of Trustees<br />

David L. Van Andel, Chairman and CEO<br />

Fritz M. Rottman, Ph.D.<br />

James B. Wyngaarden, M.D.<br />

108<br />

Board of <strong>Scientific</strong> Advisors<br />

The Board of <strong>Scientific</strong> Advisors advises the CEO and the Board of Trustees, providing recommendations and suggestions regarding<br />

the overall goals and scientific direction of VARI. The members are<br />

Michael S. Brown, M.D., Chairman<br />

Richard Axel, M.D.<br />

Joseph L. Goldstein, M.D.<br />

Tony Hunter, Ph.D.<br />

Phillip A. Sharp, Ph.D.<br />

<strong>Scientific</strong> Advisory Board<br />

The <strong>Scientific</strong> Advisory Board advises the VARI Director, providing recommendations and suggestions specific to the ongoing<br />

research, especially in the areas of cancer, genomics, and genetics. It also coordinates and oversees the scientific review process<br />

for the Institute’s research programs. The members are<br />

Alan Bernstein, Ph.D.<br />

Joan Brugge, Ph.D.<br />

Webster Cavenee, Ph.D.<br />

Frank McCormick, Ph.D.<br />

Davor Solter, M.D., Ph.D.


VARI | <strong>2007</strong><br />

Office of the Director<br />

George F. Vande Woude, Ph.D.<br />

Director<br />

Deputy Director for Clinical Programs<br />

Rick Hay, Ph.D., M.D.<br />

Deputy Director for Special Programs<br />

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

Deputy Director for Research Operations<br />

Nicholas S. Duesbery, Ph.D.<br />

109<br />

Director for Research Administration<br />

Administrator to the Director<br />

Science Editor<br />

Roberta Jones<br />

Michelle Bassett<br />

David E. Nadziejka<br />

Administration Group<br />

From left, standing:<br />

Chastain, Lewis, Koehler, Noyes,<br />

Stougaard, Carrigan, Johnson, Resau;<br />

Seated:<br />

Holman, Jason, Nelson,<br />

Novakowski, Rappley


Van Andel Research Institute | <strong>Scientific</strong> <strong>Report</strong><br />

Van Andel Institute Administrative Organization<br />

The organizational units listed below provide administrative support to both the Van Andel Research Institute and the Van Andel<br />

Education Institute.<br />

110<br />

Executive<br />

Steven R. Heacock, Chief Administrative Officer and General Counsel<br />

R. Jack Frick, Chief Financial Officer<br />

Ann Schoen, Executive Assistant<br />

Communications and Development<br />

Joseph P. Gavan, Vice President<br />

Jaime Brookmeyer<br />

Sarah Friedman<br />

Stephanie Hehl<br />

Sarah Lamb<br />

Facilities<br />

Samuel Pinto, Manager<br />

Jason Dawes<br />

Ken De Young<br />

Christen Dingman<br />

Shelly King<br />

Richard Sal<br />

Richard Ulrich<br />

Pete VanConant<br />

Jeff Wilbourn<br />

Finance<br />

Timothy Myers, Controller<br />

Sandi Essenberg<br />

Stephanie Green<br />

Richard Herrick<br />

Keri Jackson<br />

Angela Lawrence<br />

Laura Lohr<br />

Heather Ly<br />

Susan Raymond<br />

Andrew Schmidt<br />

Jamie VanPortfleet<br />

Glassware and Media Services<br />

Richard M. Disbrow, CPM, Manager<br />

Bob Sadowski<br />

Marlene Sal<br />

Grants and Contracts<br />

Carolyn W. Witt, Director<br />

Anita Boven<br />

Nicole Higgins<br />

Sara O’Neal<br />

David Ross<br />

Human Resources<br />

Linda Zarzecki, Director<br />

Margie Hoving<br />

Pamela Murray<br />

Angela Plutschouw<br />

Information Technology<br />

Bryon Campbell, Ph.D., Chief Information Officer<br />

David Drolett, Manager<br />

Bill Baillod<br />

Tom Barney<br />

Phil Bott<br />

Nathan Bumstead<br />

Charles Grabinski<br />

Kenneth Hoekman<br />

Kimberlee Jeffries<br />

Jason Kotecki<br />

Theo Pretorius<br />

Thad Roelofs<br />

Russell Vander Mey<br />

Candy Wilkerson<br />

Investments Office<br />

Kathleen Vogelsang<br />

Ted Heilman<br />

Procurement Services<br />

Richard M. Disbrow, CPM, Manager<br />

Heather Frazee<br />

Chris Kutchinski<br />

Shannon Moore<br />

Amy Poplaski<br />

John Waldon<br />

Public Affairs<br />

John VanFossen<br />

Security<br />

Kevin Denhof, CPP, Chief<br />

Christen Dingman<br />

Sandra Folino<br />

Maria Straatsma<br />

Contract Support<br />

Mary Morgan, Librarian<br />

(Grand Valley State University)<br />

Jim Kidder, Safety Manager<br />

(Michigan State University)


VARI | <strong>2007</strong><br />

111


Van Andel Research Institute | <strong>Scientific</strong> <strong>Report</strong><br />

Van Andel Institute<br />

Van Andel Institute Board of Trustees<br />

David Van Andel, Chairman<br />

Peter C. Cook<br />

Ralph W. Hauenstein<br />

John C. Kennedy<br />

Board of <strong>Scientific</strong> Advisors<br />

Michael S. Brown, M.D., Chairman<br />

Richard Axel, M.D.<br />

Joseph L. Goldstein, M.D.<br />

Tony Hunter, Ph.D.<br />

Phillip A. Sharp, Ph.D.<br />

112<br />

Van Andel Research Institute<br />

Board of Trustees<br />

David Van Andel, Chairman<br />

Fritz M. Rottman, Ph.D.<br />

James B. Wyngaarden, M.D.<br />

Chief Executive Officer<br />

David Van Andel<br />

Van Andel Education Institute<br />

Board of Trustees<br />

David Van Andel, Chairman<br />

Donald W. Maine<br />

Gordon Van Harn, Ph.D.<br />

Gordon Van Wylen, Sc.D.<br />

Van Andel Research Institute<br />

Director<br />

George Vande Woude, Ph.D.<br />

Chief Administrative Officer<br />

and General Counsel<br />

Steven R. Heacock<br />

VP Communications<br />

and Development<br />

Joseph P. Gavan<br />

Van Andel Education Institute<br />

Director<br />

Gordon Van Harn, Ph.D.<br />

Chief Financial Officer<br />

R. Jack Frick


VARI | <strong>2007</strong><br />

Van Andel Research Institute<br />

DIRECTOR – George Vande Woude, Ph.D.<br />

Deputy Directors<br />

Clinical Programs Rick Hay, Ph.D., M.D.<br />

Special Programs James Resau, Ph.D.<br />

Research Operations Nick Duesbery, Ph.D.<br />

Director for Research Administration<br />

Roberta Jones<br />

SCIENTIFIC ADVISORY BOARD<br />

Alan Bernstein, Ph.D.<br />

Joan Brugge, Ph.D.<br />

Webster Cavenee, Ph.D.<br />

Frank McCormick, Ph.D.<br />

Davor Solter, Ph.D.<br />

BASIC SCIENCE<br />

SPECIAL PROGRAMS<br />

113<br />

Cancer Cell Biology<br />

Brian Haab, Ph.D.<br />

Cancer Immunodiagnostics<br />

George Vande Woude, Ph.D.<br />

Molecular Oncology<br />

Craig Webb, Ph.D.<br />

Tumor Metastasis & Angiogenesis<br />

Signal Transduction<br />

Art Alberts, Ph.D.<br />

Cell Structure & Signal Intergration<br />

Cindy Miranti, Ph.D.<br />

Integrin Signaling & Tumorigenesis<br />

DNA Replication & Repair<br />

Michael Weinreich, Ph.D.<br />

Chromosome Replication<br />

Animal Models<br />

Nicholas Duesbery, Ph.D.<br />

Cancer & Developmental Cell Biology<br />

Bart Williams, Ph.D.<br />

Cell Signaling & Carcinogenesis<br />

Cancer Genetics<br />

Bin Teh, M.D., Ph.D.<br />

Cancer Genetics<br />

Structural Biology<br />

Eric Xu, Ph.D.<br />

Structural Sciences<br />

Systems Biology<br />

Jeffrey MacKeigan, Ph.D.<br />

Systems Biology<br />

Brian Cao, M.D.<br />

Antibody Technology<br />

Pamela Swiatek, Ph.D., M.B.A.<br />

Germline Modification<br />

Bryn Eagleson, A.A.<br />

Transgenics and Vivarium<br />

Pamela Swiatek, Ph.D., M.B.A.<br />

Cytogenetics<br />

Bin Teh, M.D., Ph.D.<br />

Sequencing<br />

Art Alberts, Ph.D.<br />

Flow Cytometry<br />

Division of Quantitative Sciences<br />

James Resau, Ph.D.<br />

James Resau, Ph.D.<br />

Analytical, Cellular,<br />

& Molecular MIcroscopy<br />

James Resau, Ph.D.<br />

Microarray Technology<br />

Kyle Furge, Ph.D.<br />

Computational Biology<br />

Greg Cavey, B.S.<br />

Mass Spectrometry and<br />

Proteomics<br />

James Resau, Ph.D.<br />

Molecular Epidemiology<br />

Animal Imaging<br />

Rick Hay, Ph.D., M.D.<br />

Noninvasive Imaging<br />

& Radiation Biology<br />

Gene Regulation<br />

Steven Triezenberg, Ph.D.<br />

Transcriptional Regulation<br />

Dean of VAI Graduate School


The Van Andel Institute and/or its affiliated organizations (VARI and VAEI), through its responsible managers, recruits, hires, upgrades,<br />

trains, and promotes in all job titles without regard to race, color, religion, sex, national origin, age, height, weight, marital status,<br />

disability, pregnancy, or veteran status, except when an accommodation is unavailable or it is a bona fide occupational qualification.<br />

Printed by Spectrum Graphics, Inc.


333 Bostwick Avenue, N.E., Grand Rapids, Michigan 49503<br />

Phone 616.234.5000 Fax 616.234.5001 www.vai.org

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

Saved successfully!

Ooh no, something went wrong!