09.06.2020 Views

2005 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.

Van Andel Research Institute<br />

<strong>Scientific</strong> <strong>Report</strong> <strong>2005</strong>


®<br />

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

Phone (616) 234-5000; Fax (616) 234-5001; Web site: www.vai.org<br />

Cover photograph of the Van Andel Institute building, Grand Rapids, Michigan


Van Andel Research Institute<br />

<strong>Scientific</strong> <strong>Report</strong><br />

<strong>2005</strong>


Title page photo: Podosomes in transformed cells<br />

This image shows podosomes in NIH3T3 mouse fibroblasts transformed with activated Src tyrosine<br />

kinase. Podosomes are the rounded structures at the tips of many of the cell extensions. They are rich in<br />

filamentous actin, which has been stained green with a conjugated phalloidin dye. The cells are co-stained<br />

with an antibody that recognizes the adhesion protein CrkL (red; or where co-localized with actin and its<br />

phalloidin stain, yellow). The protein CrkL is involved in integrin-induced cell adhesion and migration.<br />

The study of these proteins (Src, F-actin, CrkL, integrins) in these and other tumorigenic cell types may<br />

shed light on the mechanisms of podosome-mediated cancer cell metastasis and invasion.<br />

(Eduardo F. Azucena, Jr., Darren Seals, James Resau, and Sara Courtneidge)<br />

Published June <strong>2005</strong><br />

©<strong>2005</strong> by the Van Andel Institute<br />

All rights reserved<br />

Van Andel Institute<br />

333 Bostwick Avenue, N.E.<br />

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


Contents<br />

Director’s Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3<br />

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

Cell Structure and Signal Integration . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9<br />

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

Antibody Technology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 13<br />

Brian Cao, M.D.<br />

Mass Spectrometry and Proteomics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16<br />

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

Signal Regulation and Cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 18<br />

Sara A. Courtneidge, Ph.D.<br />

Cancer and Developmental Cell Biology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20<br />

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

Vivarium and Transgenics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 24<br />

Bryn Eagleson, A.A., RLATG<br />

Bioinformatics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 26<br />

Kyle Furge, Ph.D.<br />

Cancer Immunodiagnostics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 28<br />

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

Molecular Medicine and Virology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 31<br />

Sheri L. Holmen, Ph.D.<br />

Integrin Signaling and Tumorigenesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 34<br />

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

Analytical, Cellular, and Molecular Microscopy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 38<br />

and<br />

Microarray Technology and Molecular Diagnostics<br />

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

Germline Modification . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 42<br />

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

Cancer Genetics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 45<br />

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

iii


Molecular Oncology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 49<br />

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

Tumor Metastasis and Angiogenesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 53<br />

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

Chromosome Replication . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 58<br />

Michael Weinreich, Ph.D.<br />

Cell Signaling and Carcinogenesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 60<br />

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

Structural Sciences . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 63<br />

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

Mammalian Developmental Genetics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 66<br />

Nian Zhang, Ph.D.<br />

Daniel Nathans Memorial Award . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .71<br />

Postdoctoral Fellowship Program . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .75<br />

Student Programs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .79<br />

Han-Mo Koo Memorial Seminar Series . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .85<br />

Organization . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .91<br />

Recent VARI Photos . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .99<br />

iv


Director’s Introduction


Director’s Introduction<br />

We are now in our<br />

fifth year since the<br />

opening of the Van<br />

Andel Institute. The<br />

Institute was created<br />

because of the<br />

generosity of Jay Van<br />

Andel and his wife<br />

Betty, both of whom<br />

George F. Vande Woude<br />

passed away in 2004.<br />

While deeply saddened by their loss,<br />

we continue our endeavor to fulfill their vision of<br />

a center for research and education excellence in<br />

the heart of Grand Rapids.<br />

I know Jay was able to see the beginning of<br />

what a great contribution he made to society and<br />

the betterment of human health. Sharing his and<br />

Betty’s vision, we have begun to expand our<br />

research beyond cancer biology into the field of<br />

neurological disorders. David and Carol Van<br />

Andel, in Jay’s honor, have created an endowed<br />

chair dedicated to Parkinson disease (PD)<br />

research. The search for a leading scientist to<br />

take this chair is now underway.<br />

Already, Jim Resau and Bin Teh have<br />

initiated a collaboration with Australian<br />

scientists Alan Mackay-Sim and Peter Silburn at<br />

Queensland University in Australia to better<br />

understand Parkinson disease and how it<br />

develops. Alan is a developmental biologist who<br />

has initiated some exciting discoveries using<br />

adult olfactory stem cells. Peter is a neurologist<br />

who studies and treats PD patients. Together<br />

with these new collaborators, we will increase<br />

our understanding of the function, growth, and<br />

death of human nerve cells as models for PD. In<br />

addition, these approaches will be used in the<br />

study of Alzheimer disease pathophysiology.<br />

Our scientists have also partnered with the St.<br />

Mary’s Hauenstein Parkinson Center—named in<br />

honor of lead donor (and VAI trustee) Ralph<br />

Hauenstein—to determine the interactions of the<br />

environment and genes associated with PD. We<br />

will learn how these genes correlate with disease<br />

progression and drug response.<br />

We are pleased to announce that, in<br />

collaboration with the Van Andel Education<br />

Institute, the Research Institute is developing a<br />

graduate school with a program leading to the<br />

Ph.D. degree in cellular and molecular genetics,<br />

with an emphasis on translation. We expect the<br />

graduate program to contribute to the vitality and<br />

creativity of our successful research programs<br />

and to also address the nation’s need for<br />

expertise in the life sciences and biotechnology.<br />

We have received a charter from the state of<br />

Michigan and have appointed a Board of<br />

Directors for the school. Our goal is to have our<br />

first students on board in September 2006.<br />

Personnel<br />

A special occasion occurred this past year<br />

with our first promotion review. We are very<br />

proud to announce the promotion of Bin Tean Teh<br />

to Distinguished <strong>Scientific</strong> Investigator, our<br />

highest appointment level. Bin has made major<br />

contributions to the understanding of kidney,<br />

nasopharyngeal, and endocrine cancer. In recognition<br />

of his efforts, Bin was recently appointed<br />

to the Medical Advisory Board of the Kidney<br />

Cancer Association. We are also proud to<br />

announce the appointment of Rick Hay as a<br />

Senior <strong>Scientific</strong> Investigator. Rick will establish<br />

the Laboratory of Animal Imaging. Our congratulations<br />

to both scientists!<br />

Sara Courtneidge has taken a position at the<br />

Burnham Institute and is relocating her<br />

laboratory in early summer <strong>2005</strong>. Sara and I<br />

have been friends and colleagues for over 20<br />

years. I was delighted when she joined VARI<br />

and I am grateful for all her contributions to the<br />

Institute, especially her efforts in helping to<br />

establish the graduate program with MSU and<br />

the VARI postdoctoral advisory committee. The<br />

Burnham Institute is very fortunate to have<br />

recruited her, where she will be reunited with her<br />

many West Coast friends. I am sorry to see her<br />

leave, but I wish her great success and look<br />

forward to our continued scientific interactions.<br />

Publications and Competitive Funding<br />

As of April 1, <strong>2005</strong>, there have been 190<br />

peer-reviewed articles published by VARI<br />

investigators. In December 2004, a VARI article<br />

was featured on the cover of the Journal of Bone<br />

3


and Mineral Research; in January <strong>2005</strong>, we had a<br />

featured article in Molecular Cell; and in<br />

February, a VARI article in Cancer Cell was the<br />

source of the issue’s cover photo. Our<br />

investigators have also demonstrated their<br />

competitive research abilities in terms of receiving<br />

grants for funding of their work. In fiscal year<br />

2004, extramural funding for VARI dramatically<br />

increased over that in 2003. Seventeen of our<br />

scientists and six of our postdoctoral fellows<br />

received funding from 42 grants.<br />

Our new major awards have come from a<br />

variety of sources. The National Institutes of<br />

Health (NIH) National Cancer Institute (NCI)<br />

made two awards to Nick Duesbery. The first<br />

was an R01 grant for studying MEK signaling in<br />

sarcoma growth and vascularization. The second<br />

grant, an R21, was for investigating the antitumor<br />

effects of an anthrax toxin moiety (which<br />

we have termed tumor lethal factor, or “TLF”) on<br />

Kaposi sarcoma. Nick’s lab has been developing<br />

TLF as a potential cancer therapeutic.<br />

Brian Haab was awarded an R21 NCI grant for a<br />

two-year project entitled “Longitudinal Cancer-<br />

Specific Serum Protein Signatures.” This project<br />

seeks to develop protein microarray methods for<br />

detecting and diagnosing prostate cancer by<br />

examining changes over time in several cancerrelated<br />

proteins in serum. And, Art Alberts<br />

received an R21 award from the NIH to exploit a<br />

discovery in his lab with a long-term goal of<br />

developing novel anti-cancer therapies.<br />

The American Cancer Society awarded two<br />

Research Scholar Grants to our researchers.<br />

One grant, to Art Alberts, is for a four-year<br />

project to study Diaphanous-related formins in<br />

myelodysplasia. Art has been studying the role<br />

of formins in cancer. A second American Cancer<br />

Society grant was awarded to Michael Weinreich.<br />

Michael is identifying small-molecule inhibitors<br />

of Cdc7 kinase for study of its regulation in DNA<br />

replication, with a long-term goal of identifying<br />

novel targets for cancer diagnosis and therapy.<br />

Two major grants have also been received<br />

from the Michigan Technology Tri-Corridor<br />

(MTTC). One award went to Rick Hay for the<br />

development of novel agents for nuclear imaging<br />

and therapy of Met-expressing human tumors.<br />

The project is a collaboration among scientists at<br />

VARI, Michigan State University, the Department<br />

of Veterans Affairs Healthcare System in Ann<br />

Arbor, ApoLife, Inc., and the National Cancer<br />

Institute. A second grant was awarded to Bin Teh<br />

for the development of the “RenoChip,”<br />

a diagnostic and prognostic tool for use against<br />

kidney cancer. In addition, the Department of<br />

Defense awarded Eric Xu a grant for a three-year<br />

study of the structure and function of the<br />

androgen receptor in prostate cancer. Eric’s lab<br />

aims to make progress in understanding the<br />

androgen dependence (or independence) of<br />

prostate cancer.<br />

Funding from other sources in the past year<br />

has included Brian Haab’s grant from DHHS/<br />

NCI via the University of Michigan for a project<br />

entitled “Accelerated Cancer Biomarker<br />

Discovery.” This project is being undertaken by<br />

a consortium of laboratories and focuses on the<br />

development and application of new proteomics<br />

technologies for cancer biomarker discovery.<br />

Bin Teh has received a grant from the<br />

Schregardus Family Foundation for a project on<br />

renal cell carcinoma (RCC) in which his lab will<br />

be studying the prognostic value of genes for<br />

improving the clinical management of RCC<br />

patients. Bin is also the recipient of a grant from<br />

the Gerber Foundation for gene expression<br />

profiling in newborns with congenital chromosomal<br />

abnormalities.<br />

We are also proud that three of our<br />

postdoctoral fellows have received awards.<br />

Carrie Graveel (Vande Woude lab) and Kate<br />

Eisenmann (Alberts lab) have received National<br />

Research Service Awards from the NIH, while<br />

Jennifer Bromberg-White (Webb lab) received a<br />

fellowship award from the Multiple Myeloma<br />

Research Foundation.<br />

Looking to the Future<br />

We now look to expanding not only our<br />

research goals but the Institute itself. On May<br />

17th we celebrated our fifth anniversary, and our<br />

CEO, David Van Andel, announced that in 2006<br />

we will begin the second construction phase of<br />

our Institute. The new building, a model of<br />

which is displayed in the Cook-Hauenstein Hall,<br />

will join and mirror our current exceptional<br />

facility, but it will provide two-and-a-half times<br />

the existing laboratory space, or an additional<br />

150,000 square feet.<br />

4


As we plan and begin our expansion, we are<br />

part of the unprecedented growth in the health<br />

industry that Grand Rapids is experiencing.<br />

The area of Grand Rapids in which the Institute is<br />

situated has been appropriately renamed “Medical<br />

Hill.” Our neighbor across Bostwick Avenue,<br />

Spectrum Health, has celebrated the opening of<br />

the new Fred and Lena Meijer Heart Center.<br />

Furthermore, the hospital will soon break ground<br />

for the construction of a new cancer center as well<br />

as a new pediatric hospital. In another project, our<br />

own Rick Hay serves as the chairman of a<br />

combined VARI and Grand Valley State<br />

University group that is planning a good<br />

manufacturing practices (GMP) facility. This<br />

facility is being established with state and federal<br />

funding, and it will produce small quantities of<br />

clinical-grade biological products that can be<br />

tested in patients. Finally, there is the strong<br />

possibility of Michigan State University’s College<br />

of Human Medicine relocating to Grand Rapids,<br />

which would certainly be a major event in the<br />

development of the biomedical community here.<br />

Overall, with the new hospital facilities at<br />

Spectrum Health and St. Mary’s, Grand Valley<br />

State University’s strength in health sciences, and<br />

our own research program and future expansion,<br />

we will witness in the next decade exciting and<br />

dramatic developments in biomedical research,<br />

scientific discovery, and health care delivery<br />

taking place in Grand Rapids. We look forward<br />

to these many exciting changes and to the<br />

formation of a center of excellence in biomedical<br />

disciplines in western Michigan.<br />

5


Van Andel Research Institute<br />

Laboratory <strong>Report</strong>s


<strong>2005</strong> VARI<br />

<strong>Scientific</strong> Retreat<br />

8


Laboratory of Cell Structure and Signal Integration<br />

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

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

California, San Diego, in 1993, where he studied with James Feramisco. From 1994<br />

to 1997, he served as a postdoctoral fellow in Richard Treisman’s laboratory at the<br />

Imperial Cancer Research Fund in London, England. From 1997 through 1999, he<br />

was an Assistant Research Biochemist in the laboratory of Frank McCormick at the<br />

Cancer Research Institute, University of California, San Francisco. Dr. Alberts joined<br />

VARI as a <strong>Scientific</strong> Investigator in January 2000.<br />

Staff<br />

Art Alberts, Ph.D.<br />

Jun Peng, M.D.<br />

Yunju Chen, Ph.D.<br />

Kathryn Eisenmann, Ph.D.<br />

Holly Holman, Ph.D.<br />

Susan Kitchen, B.S.<br />

Laboratory Members<br />

Students<br />

Aaron DeWard, B.S.<br />

Yaojian Liu, B.S.<br />

Katharine Collins<br />

Visiting Scientists<br />

Stephen Matheson, Ph.D.<br />

Brad Wallar, Ph.D.<br />

Research Interests<br />

T<br />

he actin cytoskeleton is a dynamic,<br />

tightly regulated protein network that<br />

plays a crucial role in mediating<br />

diverse cellular processes including cell division,<br />

migration, endocytosis, vesicle trafficking, and<br />

cell shape. The research focus of the lab is the<br />

genetics and molecular biology of the Rho<br />

family of small GTPases and their effectors,<br />

which together control multiple aspects of<br />

cytoskeletal dynamics. The guiding hypothesis<br />

of the laboratory is that cytoskeletal dynamics<br />

defines the what, where, and how of signal<br />

transduction pathways, control responses to<br />

growth factors, and other extracellular cues, and<br />

that defects in these tightly controlled dynamics<br />

can contribute to cancer pathophysiology.<br />

Support for this hypothesis is observed in human<br />

cancers that carry mutations in genes encoding<br />

regulators of Rho GTPase activity. Ultimately,<br />

our goal is to exploit our understanding of the<br />

mechanics of GTPase-effector relationships in<br />

order to develop anti-cancer therapeutics.<br />

PAK1 negatively regulates the activity of the<br />

Rho exchange factor NET1<br />

Rho GTPases act as molecular switches in<br />

cells, alternating between on and off states while<br />

bound to GTP and GDP nucleotides, respectively.<br />

The activated, GTP-bound proteins preferentially<br />

interact with numerous autoregulated downstream<br />

effector proteins. Rho GTPases are activated by<br />

Rho guanine nucleotide exchange factors (Rho<br />

GEFs), one of which is the neuroepithelioma<br />

transforming gene 1 (NET1). Recently it was<br />

demonstrated that wild-type NET1 is localized in<br />

the nucleus and that truncation of the amino<br />

terminus results in relocalization of a fraction of<br />

the NET1 to the cytoplasm. This is at least<br />

partially due to the elimination of two putative<br />

nuclear localization signals within the amino<br />

terminus. Thus, NET1 activity is regulated at least<br />

in part through subcellular localization.<br />

Rho family members can modulate the<br />

activity of other Rho proteins. The protein kinase<br />

PAK1 down-regulates the activity of the RhoAspecific<br />

GEF NET1. Specifically, PAK1<br />

phosphorylates NET1 on three sites in vitro:<br />

serines 152, 153, and 538. Replacement of<br />

serines 152 and 153 with glutamate residues<br />

reduces the activity of NET1 as an exchange<br />

factor in vitro, as well as its ability to stimulate<br />

actin stress fiber formation in cells. Using a<br />

phospho-specific antibody, PAK1 can be shown<br />

to phosphorylate NET1 on serine 152 in cells, and<br />

Rac1, which activates PAK1, stimulates serine<br />

152 phosphorylation in a PAK1-dependent<br />

manner. Furthermore, coexpression of<br />

constitutively active PAK1 inhibits NET1<br />

stimulation of actin polymerization only when<br />

serines 152 and 153 are present. These<br />

observations provide a novel mechanism for the<br />

control of RhoA activity.<br />

9


Signal transduction and spatially controlled<br />

assembly of F-actin networks<br />

One well-characterized actin nucleator, the<br />

Arp2/3 complex, induces the formation of<br />

branched actin filaments. Arp2/3 works by<br />

complexing with G-actin or by binding to the side<br />

of preexisting filaments. Its nucleation and<br />

filament-binding activity is tightly regulated by<br />

interactions with nucleation-promoting factors<br />

(NPFs), the most prominent being the WASp/Scar<br />

family. WASp is an autoregulated molecular<br />

switch controlled by yet other switch-like proteins<br />

such as Cdc42, a Rho family small GTPase. Thus,<br />

NPFs integrate signals controlling growth<br />

factor–stimulated actin nucleation and branching.<br />

Cdc42-activated WASp induction of Arp2/3<br />

activity is shown schematically in Fig. 1A.<br />

The mammalian Diaphanous-related formins<br />

Formins are a highly conserved family of<br />

proteins implicated in a diverse array of cellular<br />

functions including the cytoskeletal remodeling<br />

events necessary for cytokinesis, bud formation<br />

in yeast, establishment of cell/organelle polarity,<br />

and endocytosis. Formins have the ability to<br />

stabilize microtubules, which (like F-actin) are<br />

assembled by tightly controlled cycles of<br />

polymerization and depolymerization.<br />

The mammalian Diaphanous-related formin<br />

(mDia) proteins are a subfamily of formins that<br />

share a loosely conserved Rho GTPase-binding<br />

domain (GBD) in the amino terminus and a<br />

highly conserved Diaphanous-autoregulatory<br />

domain (DAD) in the carboxy terminus.<br />

The GBDs can interact with their internal DAD<br />

partners in vitro, leading to the autoregulation<br />

model depicted in Fig. 1B. The model shows that<br />

while the formin proteins dimerize through their<br />

FH2 domains, it is the GBD-DAD interaction that<br />

is the linchpin of autoregulation. GTP-bound<br />

Rho can interact with the GBD and interrupt the<br />

autoinhibited conformation, leading to nucleation<br />

and elongation of nonbranched actin filaments.<br />

It has been unclear whether GTPase binding<br />

simply activates the mDia proteins’ ability to<br />

nucleate actin, or provides other signals that<br />

direct subcellular targeting and recruitment of<br />

mDia-associated proteins. Another question is,<br />

do mDia proteins activated in specific cellular<br />

contexts (i.e., on vesicles or at sites of adhesion)<br />

associate with or work in parallel with other<br />

modifiers of actin polymerization to generate<br />

site-specific F-actin networks? We are studying<br />

such questions using FRET technology.<br />

Site-specific interactions between Rho<br />

GTPases and mDia proteins<br />

Fluorescence resonance energy transfer<br />

(FRET) is a powerful technique that allows us to<br />

assay protein-protein interactions in cells by<br />

using two fluorophores, in this case, cyan<br />

fluorescent protein (CFP) and yellow fluorescent<br />

protein (YFP). When fused to the GTPase and<br />

excited at the appropriate wavelength, CFP acts<br />

as a fluorescent donor which then excites YFP,<br />

which is fused to a Drf protein (Fig. 1B). FRET<br />

Figure 1. Drfs are actin nucleators whose activity is regulated through interactions with small<br />

GTPases. A) A model for formin (mDia1-3) and WASp collaborating in cells with activated Cdc42, in which<br />

Cdc42 interacts with mDia2 in cells at specific sites associated with membrane protrusions. In this model,<br />

activated WASp nucleates branched filaments from the side of mDia2 nucleated “mother” filaments;<br />

alternatively, mDia2 binds to and processively elongates filaments after nucleation by Arp2/3. B) GTP-bound<br />

Rho GTPase binding disrupts intramolecular interactions between the GBD and DAD of a DRF. If the GTPase<br />

and GBD are linked to fluorophores (ECFP and EYFP), the proximity of the two can be determined by FRET.<br />

10


occurs only when the donor/acceptor pair is in<br />

close proximity (less than 30 Å) .<br />

Fusion proteins are expressed following<br />

microinjection of their expression plasmids into<br />

cells, which are then fixed 4 h later. We have<br />

shown that YFP-mDia2 is expressed with CFP-<br />

Cdc42, primarily at the leading cell edge (or<br />

cortex) and at the microtubule-organizing center.<br />

In other experiments, we have shown that this<br />

interaction depends upon the integrity of the<br />

CRIB motif within the mDia2 GBD. CRIB<br />

motifs are necessary for binding to the GTPase.<br />

Our observations indicate that one particular<br />

GTPase-formin pair, Cdc42 and mDia2, may<br />

have a role in remodeling actin at the cell edge.<br />

What this pair contributes to actin or microtubule<br />

dynamics at the MTOC, however, remains an<br />

open question. We speculate that the pair may<br />

participate in microtubule regulation at the minus<br />

(–) end of the tubules, which (like actin) are<br />

assembled and disassembled in a polarized<br />

fashion. Other GTPase-formin pairs may be<br />

working at the plus end to direct them to focal<br />

adhesions or other sites. From collaborative<br />

efforts with the Gundersen lab, it has been shown<br />

that mDia1 and mDia2 can complex with the<br />

MT(+)-end binding proteins APC and EB1.<br />

In contrast to the speculative role of Cdc42<br />

and mDia2, RhoB is known to have a role in<br />

endocytic or vesicular trafficking, and it interacts<br />

with mDia2 on endosomes (Fig. 2). This result<br />

is consistent with our discovery of both mDia1<br />

and mDia2 on endosomes. The expression of<br />

either activated RhoB or deregulated versions of<br />

mDia1, mDia2, and mDia3 blocks the movement<br />

of vesicles and increases their number. One<br />

interpretation is that expression of RhoB or<br />

deregulated mDia1–3 triggers an inappropriate<br />

transition from fast microtubule-dependent<br />

transport to actin-dependent transport.<br />

Formins as anti-cancer drug targets<br />

A dynamic cytoskeleton is required for tumor<br />

cell growth. Drugs that stabilize the cytoskeleton<br />

are emerging as effective anti-cancer<br />

therapeutics. For example, Taxol binds directly<br />

to the components that comprise the microtubule<br />

cytoskeleton and blocks their dynamics. A<br />

cyclopeptide derived from a sea sponge,<br />

jasplakinolide, has similar effects on actin.<br />

Figure 2. RhoB<br />

interacts with mDia2<br />

on vesicles. CFP-fused<br />

activated RhoB-G14V<br />

and YFP-fused mDia2<br />

were co-injected into<br />

cells and FRET was<br />

assessed 4 h later. A<br />

FRET signal is observed<br />

between activated RhoB<br />

and mDia2 upon<br />

endosomes. These data<br />

indicate that individual<br />

GTPase-formin pairs<br />

would appear to be<br />

functionally distinct,<br />

promoting the formation<br />

of different actin<br />

structures at different<br />

sites. Since both RhoB<br />

and Cdc42 have been<br />

implicated in endocytosis<br />

and vesicle trafficking, it<br />

is possible that both<br />

GTPases use mDia2<br />

sequentially or in parallel<br />

as effectors to transport<br />

cargo within cells.<br />

Previously, we found that a peptide derived<br />

from the DAD region of mDia proteins, when<br />

expressed in cells, stabilized both the actin and<br />

microtubule cytoskeletons. Because the<br />

mechanism of DAD action is unique—it binds to<br />

cellular formins and disrupts their normal<br />

autoregulatory mechanism—DAD represents a<br />

novel class of anti-tumor drugs.<br />

Because DAD is unable to enter cells as a<br />

drug, we have begun searching for functional<br />

analogs of DAD that could have similar properties<br />

under a drug-development program funded by the<br />

National Cancer Institute. We hypothesize that by<br />

deregulating specific mDia molecules in tumor<br />

cells, we can arrest dynamic remodeling of the<br />

cytoskeleton, a process required for cell motility<br />

and cytokinesis. We will characterize the<br />

structural and functional requirements for DADinduced<br />

cell death and develop a high-throughput<br />

screen for the identification of novel molecules<br />

that can deregulate mDia proteins and kill tumor<br />

cells. Our objectives are to initiate a drug<br />

discovery program by validating mDia proteins as<br />

molecular targets in cancer and to determine the<br />

physical requirements for DAD interactions with<br />

the mDia GBD.<br />

11


External Collaborators<br />

Philippe Chavrier, Institut Curie, Paris, France<br />

Jeff Frost, University of Houston, Texas<br />

Gregg Gundersen, Columbia University, New York<br />

George Prendergast, Lankenau Institute, Wynnewood, Pennsylvania<br />

Kathy Siminovitch, University of Toronto, Canada<br />

Recent Publications<br />

Alberts, A.S., H. Qin, H.S. Carr, and J.A. Frost. In press. PAK1 negatively regulates the activity of<br />

the Rho exchange factor NET1. Journal of Biological Chemistry.<br />

Eisenmann, K.M., J. Peng, B.J. Wallar, and A.S. Alberts. In press. Rho GTPase-formin pairs in<br />

cytoskeletal remodeling. In Signaling Networks in Cell Shape and Motility, London, U.K.:<br />

Novartis Foundation.<br />

Wen, Ying, Christina H. Eng, Jan Schmoranzer, Noemi Cabrera-Poch, Edward J.S. Morris, Michael<br />

Chen, Bradley J. Wallar, Arthur S. Alberts, and Gregg G. Gundersen. 2004. EB1 and APC bind<br />

to mDia to stabilize microtubules downstream of Rho and promote cell migration. Nature Cell<br />

Biology 6(9): 820–830.<br />

Left to right: Liu, Holman, DeWard, Chen, Peng, Collins, Alberts, Kitchen, Eisenmann<br />

12


Laboratory of Antibody Technology<br />

Brian Cao, M.D.<br />

Dr. Cao obtained his M.D. from Beijing Medical University, People’s Republic of<br />

China, in 1986. On receiving a CDC fellowship award, he was a visiting scientist at<br />

the National Center for Infectious Diseases, Centers for Disease Control and<br />

Prevention (1991–1994). He next served as a postdoctoral fellow at Harvard<br />

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

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

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

Cancer Institute–Frederick Cancer Research and Development Center, Maryland.<br />

Dr. Cao joined VARI as a Special Program Investigator in June 1999.<br />

Staff<br />

Ping Zhao, M.S.<br />

Tessa Grabinski, B.S.<br />

Laboratory Members<br />

Visiting Scientist<br />

Mei Guo, M.S.<br />

Students<br />

Yong-jun Jiao<br />

Xin Wang<br />

Jin Zhu<br />

Research Interests<br />

H<br />

epatocyte<br />

growth factor/scatter factor<br />

(HGF/SF) is a multifunctional<br />

heterodimeric protein produced by<br />

mesenchymal cells and is an effector of cells<br />

expressing the tyrosine kinase receptor Met. Met,<br />

the protein product of the c-met protooncogene, is<br />

from the same family as epidermal growth factor<br />

(EGF) receptors. The activation of Met by<br />

HGF/SF affects downstream signaling pathways<br />

(including other protein kinases) responsible for<br />

cellular differentiation, motility, proliferation,<br />

organogenesis, angiogenesis, and apoptosis.<br />

Aberrant expression of the Met-HGF/SF<br />

receptor-ligand complex—resulting either from<br />

mutations in the complex or in conjunction with<br />

mutations in other oncogenes—is associated with<br />

an invasive/metastatic phenotype in most solid<br />

human tumors. Met-HGF/SF and downstream<br />

kinases are therefore attractive targets for new<br />

anti-cancer agents for clinical diagnosis,<br />

prognosis, and treatment.<br />

The aberrant expression of the Met receptor<br />

kinase by two-thirds of localized prostate cancers,<br />

and apparently by all osseous metastases,<br />

suggests that Met provides a strong mechanism of<br />

selection for metastatic development. We have<br />

generated and characterized several anti-Met<br />

murine monoclonal antibodies (mAbs) that have<br />

high affinity for and specifically recognize Met<br />

extracellular domains in their native<br />

conformation. In collaborative studies, we are<br />

using two of these radiolabeled anti-Met mAbs,<br />

designated Met3 and Met5, to study mouse<br />

xenograft and orthotopic models of localized and<br />

metastatic prostate cancer via clinical nuclear<br />

imaging. Moreover, we will soon be testing these<br />

two radiolabeled mAbs on dog spontaneous<br />

prostate cancer and bone metastasis models.<br />

In collaboration with the Nanjing Medical<br />

University of China, we have initiated a project to<br />

construct a phage-display antibody fragment<br />

library. This technique involves the construction<br />

and use of animal/human, immunized/naïve Fab<br />

and scFv antibody gene repertoires by phage<br />

display. The ability to co-select antibodies and<br />

their genes allows the isolation of high-affinity,<br />

antigen-specific mAbs derived from either<br />

immunized animals or non-immunized humans. A<br />

number of procedures for selecting such antibodies<br />

from recombinant libraries have been described,<br />

and some useful antibodies have been produced<br />

with this approach. Over the past two years, we<br />

have closely followed the development of this<br />

technology for producing novel recombinant<br />

antibody-like molecules. We have constructed a<br />

human naïve Fab library with the diversity of 2 ×<br />

10 9 and have screened out some mAb fragments<br />

against tumor marker proteins. In particular, we<br />

have selected from this library and characterized<br />

one specific anti-Met Fab fragment, designated as<br />

Fab-Met-1, using a subtractive whole-cell panning<br />

approach (Figs. 1 and 2).<br />

We have also established the technology of a<br />

phage-display peptide library for mAb epitope<br />

13


A<br />

B<br />

mapping. A random peptide library<br />

is constructed by genetically fusing<br />

oligonucleotides to the coding<br />

sequence of a coat protein of<br />

bacteriophage, resulting in display<br />

of the fused polypeptide on the<br />

surface of the virion. Phage display<br />

has been used to create a physical<br />

linkage between a vast library of<br />

random peptide sequences and the<br />

DNA encoding each sequence,<br />

allowing rapid identification of<br />

peptide ligands for a variety of<br />

target molecules such as antibodies.<br />

A library of phage is exposed to a<br />

plate coated with mAb. Unbound<br />

phages are washed away, and<br />

specifically bound phages are eluted<br />

by lowering the pH. The eluted pool of phage is<br />

amplified, and the process is repeated for two more<br />

rounds. Individual clones are isolated, screened by<br />

ELISA, and sequenced. We have successfully<br />

epitope-mapped a variety of important mAbs<br />

including anti-HGF/SF, anti-Met, and anti-anthrax<br />

lethal factor. We are now exploring the use of this<br />

technology on protein-protein interactions; one<br />

example is the mapping of the HGF/SF-Met<br />

binding site in an in vitro system, and several<br />

interesting peptides have been selected from the<br />

library as being potential Met antagonists.<br />

Functioning as an antibody production core<br />

facility at the Van Andel Research Institute, this<br />

lab has extensive capabilities in the generation,<br />

characterization, scaled-up production, and<br />

purification of mAbs using comprehensive<br />

cutting-edge technologies. The technologies and<br />

services available in the core include animal<br />

immunization and antigen preparation; peptide<br />

design; DNA immunization (Gene-gun<br />

technology); immunization of a wide range of<br />

antibody-producing models (including mice,<br />

rats, rabbits, human cells, and transgenic or<br />

knock-out mice); and in vitro immunization.<br />

Other services we provide include the generation<br />

of hybridomas from spleen cells of immunized<br />

mice, rats, and rabbits; hybridoma expansion and<br />

subcloning; cryopreservation of hybridomas<br />

secreting mAbs; monoclonal antibody isotyping;<br />

ELSIA screening of hybridoma supernatants;<br />

monoclonal antibody characterization by<br />

immunoprecipitation, Western blot, immunohistochemistry,<br />

immunofluorescence staining,<br />

FACS, or in vitro bioassays; production of bulk<br />

quantities of mAbs using high-density cell culture;<br />

purification of mAbs using FPLC affinity columns;<br />

generation of bi-specific mAbs by secondary<br />

fusion; conjugation of mAbs to detection enzymes<br />

(biotin/streptavidin, fluorescence reporters, etc.);<br />

and the development of detection methods/kits<br />

such as sandwich ELISA. Over the past few years,<br />

this facility has generated more than 200 different<br />

mAbs, 10 of which have been licensed to<br />

commercial companies. We have also contracted<br />

services to local biotechnology companies to<br />

generate, characterize, produce, and purify mAbs<br />

for their research/diagnostic kit development.<br />

S114<br />

Figure 1. A) SDS–PAGE of Fab-<br />

Met-1 fragment purified by affinity<br />

chromatography. Lane 1, standard<br />

molecular weight markers; lane 2,<br />

purified Fab fragment under reducing<br />

conditions. The concentration of the<br />

running gel was 12%.<br />

B) Immunoprecipitation analysis of<br />

Fab-Met-1. Met from cell extracts<br />

was immunoprecipitated with purified<br />

Fab-Met-1 and detected by western<br />

blot analysis. Lane 1 is S114 cell<br />

lysate immunoprecipitated with C-28<br />

(rabbit anti-human Met polyclonal<br />

antibody). Lanes 2–5 are cell lysates<br />

immunoprecipitated with Fab-Met-1:<br />

lane 2, S-114 (Met+); lane 3, MKN45<br />

(Met+); lane 4, M14 cell (Met–); and<br />

lane 5, NIH3T3 (Met–).<br />

M14<br />

Figure 2. The binding affinity of a selected Fab<br />

fragment (Fab-Met-1) was tested by FACS analysis.<br />

Two cell lines, S114 (Met+) and M14 (Met–), were<br />

incubated with purified Fab-Met-1. Bound Fab was<br />

detected by staining the cells with secondary goat<br />

anti-human Fab–FITC conjugate and analyzing by<br />

FACS (black lines). Green lines indicate staining<br />

with the secondary antibody only.<br />

14


External Collaborators<br />

Zhen-qing Feng, Nanjing Medical University, China<br />

Milton Gross, Department of Veterans Affairs Medical Center/University of Michigan, Ann Arbor<br />

Xiao-hong Guan, Nanjing Medical University, China<br />

Beatrice Knudsen, Fred Hutchinson Cancer Research Center, Seattle, Washington<br />

Yi Ren, Cancer Center, Cleveland Clinic Foundation, Cleveland, Ohio<br />

Kang-lin Wan, Chinese Centers for Disease Control and Prevention, Beijing, China<br />

David Waters, Gerald P. Murphy Cancer Foundation, Seattle, Washington<br />

David Wenkert, Michigan State University, East Lansing<br />

Wei-cheng You, Beijing Institute for Cancer Research, China<br />

Recent Publications<br />

Jiao, Y., P. Zhao, J. Zhu, T. Grabinski, Z. Feng, X. Guan, R.S. Skinner, M.D. Gross, Y. Su,<br />

G.F. Vande Woude, R.V. Hay, and B. Cao. In press. Construction of human naïve Fab<br />

library and characterization of anti-Met Fab fragment generated from the library.<br />

Molecular Biotechnology.<br />

Zhang, Yu-Wen, Yanli Su, Nathan Lanning, Margaret Gustafson, Nariyoshi Shinomiya, Ping Zhao,<br />

Brian Cao, Galia Tsarfaty, Ling-Mei Wang, Rick Hay, and George F. Vande Woude. <strong>2005</strong>.<br />

Enhanced growth of human Met-expressing xenografts in a new strain of immunocompromised<br />

mice transgenic for human hepatocyte growth factor/scatter factor. Oncogene 24(1): 101–106.<br />

Tan, Min-Han, Carl Morrison, Pengfei Wang, Ximing Yang, Carola J. Haven, Chun Zhang, Ping<br />

Zhao, Maria S. Tretiakova, Eeva Korpi-Hyovalti, John R. Burgess, Khee Chee Soo, Wei-Keat<br />

Cheah, Brian Cao, James Resau, Hans Morreau, and Bin Tean Teh. 2004. Loss of parafibromin<br />

immunoreactivity is a distinguishing feature of parathyroid carcinoma. Clinical Cancer<br />

Research 10(19): 6629–6637.<br />

Zhao, Ping, Xudong Liang, Jessica Kalbfleisch, Han-Mo Koo, and Brian Cao. 2003. Neutralizing<br />

monoclonal antibody against anthrax lethal factor inhibits intoxication in a mouse model.<br />

Human Antibodies 12(4): 129–135.<br />

From left to right: Zhu, Wang, Cao, Ferrell, Grabinski, Zhao<br />

15


Laboratory of Mass Spectrometry and Proteomics<br />

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

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

joining VARI he was employed at Pharmacia in Kalamazoo, Michigan, for nearly 15<br />

years. As a member of a biotechnology development unit, he was group leader for<br />

a protein characterization core laboratory. More recently as a research scientist in<br />

discovery research, he was principal in the establishment and application of a stateof-the-art<br />

proteomics laboratory for drug discovery. Mr. Cavey joined VARI as a<br />

Special Program Investigator in July 2002.<br />

Staff<br />

Paula Davidson, M.S.<br />

Laboratory Members<br />

Student<br />

Wendy Johnson<br />

Research Interests<br />

The Mass Spectrometry and Proteomics<br />

program works with many of the<br />

research labs at the Institute, using stateof-the-art<br />

mass spectrometers in combination with<br />

analytical protein separation and purification<br />

methods to help answer a wide range of biological<br />

questions. Using mass spectrometry data and<br />

database search software, proteins can be<br />

identified and characterized with unprecedented<br />

sensitivity and throughput. Since proteomics is a<br />

relatively new scientific discipline, many of the<br />

analytical techniques are rapidly changing;<br />

therefore our mission involves using established<br />

protocols, improving them, and developing new<br />

approaches to expand the scope of biological<br />

challenges being addressed.<br />

Protein-protein interactions<br />

Analyzing samples representing different<br />

cellular conditions or disease states is a step toward<br />

understanding the role of a protein with an<br />

unknown function or understanding the regulatory<br />

mechanism of several proteins in a given pathway.<br />

In this approach, a known protein is affinitypurified<br />

from a nondenatured sample. The purified<br />

protein and its binding partners are separated using<br />

two-dimensional (2D) electrophoresis gels or<br />

SDS-PAGE. After staining, the proteins are cut<br />

from the gel, enzymatically digested into peptides,<br />

and then analyzed by nanoscale high-pressure<br />

liquid chromatography on line with a mass<br />

spectrometer (LC-MS). The mass spectrometer<br />

fragments the peptides and the resulting spectra are<br />

used to search protein or translated DNA<br />

databases. Identifications are made using the<br />

amino acid sequences derived from the mass<br />

spectrometry data. We have optimized all aspects<br />

of this analysis for sample recovery yields and<br />

high-sensitivity protein identification.<br />

Recently, we have been evaluating newly<br />

developed software that allows us to eliminate the<br />

electrophoresis separation step from these analyses,<br />

giving the potential to identify more proteins from<br />

complex mixtures. With this software, affinitypurified<br />

protein complexes are compared to a<br />

control sample via peptide differential display. The<br />

proteins are digested into peptides in solution rather<br />

than from gels and are analyzed by LC-MS.<br />

Peptides unique to the experimental sample relative<br />

to the control are used to identify proteins that are<br />

part of a protein complex.<br />

Protein characterization<br />

Our laboratory also characterizes proteins and<br />

their post-translational modifications. Purified<br />

proteins are analyzed by protein electrospray to<br />

confirm the average protein molecular weight<br />

before proceeding to labor-intensive studies such<br />

as protein crystallization.<br />

Mapping the post-translational modifications<br />

of proteins such as phosphorylation is an important<br />

undertaking in cancer research. Phosphorylation<br />

regulates many protein pathways, several of which<br />

could serve as potential drug targets for cancer<br />

therapy. In recent years, mass spectrometry has<br />

emerged as a primary tool that helps investigators<br />

determine exactly which amino acids of a protein<br />

are modified. This undertaking is complicated by<br />

many factors, but principally by the fact that<br />

pathway regulation can occur when only 0.01% of<br />

the molecules of a given protein are<br />

16


phosphorylated. Thus, we are dealing with an<br />

extremely small number of molecules, in addition<br />

to the fact that the purification of phosphopeptides<br />

is always difficult. Our lab collaborates with<br />

investigators to map protein phosphorylation using<br />

techniques including immobilized metal affinity<br />

purification following esterification; immunoaffinity<br />

purification of phosphoproteins and<br />

peptides; and phosphorylation-specific mass<br />

spectrometry detection.<br />

Protein expression<br />

As mass spectrometry instruments and protein<br />

separation methods develop, we hope to identify<br />

and quantitate all the proteins expressed in a given<br />

cell or tissue, as a means of evaluating all of the<br />

physiological processes occurring within. This<br />

approach, termed systems biology, aims at<br />

understanding how all proteins interact to affect a<br />

biological outcome. Traditionally this approach<br />

has used 2D gel electrophoresis, image analysis of<br />

stained proteins, and identification of proteins<br />

from gels using mass spectrometry. Due to the<br />

labor-intensive nature of 2D gels and the<br />

underrepresentation of some classes of proteins<br />

(such as membrane proteins), proteomics has been<br />

moving toward solution-based separations and<br />

direct mass spectrometry. Our first approach is to<br />

digest all proteins into peptides and label their<br />

C-terminus with 18 O water to effect a mass shift.<br />

Experimental and control samples are then mixed<br />

and separated by multidimensional high-pressure<br />

liquid chromatography using strong-cation ion<br />

exchange and reverse-phase separation modes.<br />

Peptides that are differentially expressed in<br />

experimental and control samples according to<br />

their 16 O/ 18 O ratio are identified using mass<br />

spectrometry and database searching.<br />

We intend to apply this or other mass<br />

spectrometry–based approaches in the discovery<br />

of biomarkers for early cancer detection, for morespecific<br />

diagnosis, and for more-accurate<br />

prognosis following drug treatment.<br />

External Collaborators<br />

Greg Fraley, Hope College, Holland, Michigan<br />

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

Brett Phinney, Michigan State University, East Lansing<br />

Recent Publications<br />

Li, Yong, Mihwa Choi, Greg Cavey, Jennifer Daugherty, Kelly Suino, Amanda Kovach, Nathan C.<br />

Bingham, Steven A. Kliewer, and H. Eric Xu. <strong>2005</strong>. Crystallographic identification and functional<br />

characterization of phospholipids as ligands for the orphan nuclear receptor steroidogenic factor-1.<br />

Molecular Cell 17(4): 491–502.<br />

From left to right: Davidson, Johnson, Cavey<br />

17


Laboratory of Signal Regulation and Cancer<br />

Sara A. Courtneidge, Ph.D.<br />

Dr. Courtneidge completed her Ph.D. at the National Institute for Medical Research<br />

in London. She began her career in the basic sciences in 1978 as a postdoctoral<br />

fellow in the laboratory of J. Michael Bishop at the University of California School of<br />

Medicine. She later joined her alma mater as a member of the scientific staff. In<br />

1985 Dr. Courtneidge joined the European Molecular Biology Laboratory as group<br />

leader and in 1991 was appointed a senior scientist with tenure. She joined Sugen<br />

in 1994 as Vice President of Research, later becoming Senior Vice President of<br />

Research and then Chief Scientist. Dr. Courtneidge joined VARI in January 2001<br />

as a Distinguished <strong>Scientific</strong> Investigator.<br />

Staff<br />

Eduardo Azucena, Ph.D.<br />

Paul Bromann, Ph.D.<br />

Hasan Korkaya, Ph.D.<br />

Laboratory Members<br />

Ian Pass, Ph.D.<br />

Darren Seals, Ph.D.<br />

Laila Al-Duwaisan<br />

Research Interests<br />

Our laboratory wants to understand at the<br />

molecular level how proliferation is<br />

controlled in normal cells and by what<br />

mechanisms these controls are subverted in tumor<br />

cells. We largely focus on a family of oncogenic<br />

tyrosine kinases, the Src family. The prototype of<br />

the family, vSrc, originally discovered as the<br />

transforming protein of Rous sarcoma virus, is a<br />

mutated and activated version of a normal cellular<br />

gene product, cSrc. The activity of all members of<br />

the Src family is normally under strict control;<br />

however the enzymes are frequently activated or<br />

overexpressed, or both, in human tumors.<br />

In normal cells, Src family kinases (SFKs) have<br />

been implicated in signaling from many types of<br />

receptors, including receptor tyrosine kinases,<br />

integrin receptors, and G protein–coupled<br />

receptors. Signals generated by SFKs are thought<br />

to play roles in cell cycle entry, cytoskeletal<br />

rearrangements, cell migration, and cell division.<br />

In tumor cells, Src may play a role in growth<br />

factor–independent proliferation or in invasiveness.<br />

Furthermore, some evidence points to a role for<br />

SFKs in angiogenesis. Some of the current projects<br />

in the laboratory are outlined below.<br />

The role of the Src substrate<br />

Tks5/Fish in tumorigenesis<br />

Tks5/Fish is an adaptor protein which has<br />

five SH3 domains and a phox homology (PX)<br />

domain. Tks5/Fish is tyrosine-phosphorylated in<br />

Src-transformed fibroblasts (suggesting that it is a<br />

target of Src in vivo) and in normal cells after<br />

treatment with any of several growth factors.<br />

We recently found that in Src-transformed cells,<br />

Tks5/Fish is localized to specialized regions of<br />

the plasma membrane called podosomes<br />

(sometimes referred to as invadopodia).<br />

These actin-rich protrusions from the plasma<br />

membrane are sites of matrix invasion and<br />

locomotion. The PX domain of Tks5/Fish<br />

associates with phosphatidylinositol 3-phosphate<br />

and phosphatidylinositol 3,4-bisphosphate, and it<br />

is required for targeting Tks5/Fish to podosomes.<br />

The fifth SH3 domain of Tks5/Fish mediates its<br />

association with members of the ADAMs family of<br />

membrane metalloproteases, which in Srctransformed<br />

cells are also localized to podosomes.<br />

We have begun to dissect the role of Tks5/Fish in<br />

transformation. Src-transformed cells with<br />

reduced Tks5/Fish levels no longer form<br />

podosomes and are poorly invasive. We detected<br />

Tks5/Fish expression in podosomes in invasive<br />

human cancer cell lines, as well as in tissue<br />

samples from human breast cancer and melanoma.<br />

Tks5/Fish expression was also required for<br />

invasion of human cancer cells. Furthermore, we<br />

have developed an assay to generate podosomes<br />

upon expression of Tks5/Fish, which will allow us<br />

to dissect the requirements for podosome<br />

formation in more detail. We are also investigating<br />

the potential of both Tks5/Fish and its binding<br />

proteins as markers of invasive disease and as<br />

potential therapeutic targets.<br />

18


The role of Src family kinases in<br />

mitogenic signaling pathways<br />

We have previously shown that Src family<br />

kinases are required for both Myc induction and<br />

DNA synthesis in response to platelet-derived<br />

growth factor (PDGF) stimulation of NIH3T3<br />

fibroblasts. We have also previously identified and<br />

characterized a small-molecule inhibitor of Src<br />

family kinases called SU6656. We wanted to<br />

address whether there is a discrete SFK-specific<br />

pathway leading to enhanced gene expression, or<br />

whether SFKs act to generally enhance PDGFstimulated<br />

gene expression. To do this, we treated<br />

quiescent NIH3T3 cells with PDGF in the<br />

presence or absence of SU6656 and analyzed<br />

global patterns of gene expression. We determined<br />

that a discrete set of immediate early genes was<br />

induced by PDGF and inhibited by SU6656.<br />

We further determined that SFKs did not stimulate<br />

the rate of transcription of these genes, but rather<br />

promoted mRNA stabilization. We are currently<br />

exploring how SFKs signal gene expression by<br />

enhancing mRNA stability.<br />

Breast cancer<br />

Increased Src activity can be demonstrated in<br />

the majority of breast cancers, both estrogendependent<br />

and estrogen-independent, yet the role<br />

of Src in breast tumorigenesis has not been fully<br />

established. We have been characterizing the role<br />

of Src in estrogen-stimulated signal transduction<br />

pathways in breast cancer cell lines. We have<br />

shown that Src family kinase activity is required<br />

for estrogen to stimulate mitogenesis in MCF7<br />

cells. Furthermore, inhibition of Src prevents<br />

estrogen stimulation both of Myc and of MAP<br />

kinase activity. We are currently dissecting which<br />

Src signaling pathways are necessary for estrogenstimulated<br />

growth, as well as how Src activity<br />

results in the activation of MAP kinase and in the<br />

production of Myc.<br />

Recent Publications<br />

Bromann, Paul A., Hasan Korkaya, Craig P. Webb, Jeremy Miller, Tammy L. Calvin, and Sara A.<br />

Courtneidge. <strong>2005</strong>. Platelet-derived growth factor stimulates Src-dependent mRNA stabilization of<br />

specific early genes in fibroblasts. Journal of Biological Chemistry 280(11): 10253–10263.<br />

Seals, Darren F., Eduardo F. Azucena, Jr., Ian Pass, Lia Tesfay, Rebecca Gordon, Melissa Woodrow, James<br />

H. Resau, and Sara A. Courtneidge. <strong>2005</strong>. The adaptor protein Tks5/Fish is required for podosome<br />

formation and function, and for the protease-driven invasion of cancer cells. Cancer Cell 7(2):<br />

155–165.<br />

Bromann, Paul A., Hasan Korkaya, and Sara A. Courtneidge. 2004. The interplay between Src family<br />

kinases and receptor tyrosine kinases. Oncogene 23(48): 7957–7968.<br />

From left to right: Azucena, Seals, Pass, Al-Duwaisan, Bromann<br />

19


Laboratory of Cancer and Developmental Cell Biology<br />

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

Dr. Duesbery received both his M.Sc. (1990) and Ph.D. (1996) degrees in zoology<br />

from the University of Toronto, Canada, under the supervision of Yoshio Masui.<br />

Before his appointment as a <strong>Scientific</strong> Investigator at VARI in April 1999, he was a<br />

postdoctoral fellow in the laboratory of George Vande Woude in the Molecular<br />

Oncology Section of the Advanced BioScience Laboratories–Basic Research<br />

Program at the National Cancer Institute–Frederick Cancer Research and<br />

Development Center, Maryland.<br />

Staff<br />

Paul Spilotro, M.D.<br />

Philippe Depeille, Ph.D.<br />

Hilary Wagner, M.S.<br />

John Young, M.S.<br />

Elissa Boguslawski<br />

Laboratory Members<br />

Students<br />

Chia-Shia Lee<br />

Lisa Orcasitas<br />

Visiting Scientist<br />

Gustavo Nacheli, M.D.<br />

Research Interests<br />

The overall goal of the Laboratory of<br />

Cancer and Developmental Cell Biology<br />

is to further the study of mitogenactivated<br />

protein kinase kinase (MEK) signaling<br />

in health and disease. Currently, work performed<br />

in the lab is organized into three projects to<br />

explore 1) MEK signaling and tumor biology,<br />

2) the therapeutic potential of anthrax lethal toxin<br />

(LeTx), and 3) molecular mechanisms of<br />

LeTx action.<br />

MEK signaling and tumor biology<br />

Many malignant sarcomas, such as<br />

angiosarcomas, are refractory to currently<br />

available treatments. However, sarcomas possess<br />

unique vascular properties which indicate they<br />

may be more responsive to therapeutic agents that<br />

target endothelial function. MEKs have been<br />

demonstrated to play an essential role in the<br />

growth and vascularization of carcinomas.<br />

We hypothesize that signaling through multiple<br />

MEK pathways is also essential for growth and<br />

vascularization of sarcomas. The objective of this<br />

research is to define the role of MEK signaling in<br />

the growth and vascularization of human sarcoma<br />

and to determine whether inhibition of multiple<br />

MEKs by agents such as anthrax lethal toxin, a<br />

proteolytic inhibitor of MEKs, may form the basis<br />

of a novel therapeutic approach to the treatment of<br />

human sarcoma. In 2004 we successfully<br />

obtained funding from the National Institutes of<br />

Health for this project.<br />

The therapeutic potential of<br />

anthrax lethal toxin<br />

Data from the National Cancer Institute’s<br />

Anti-Neoplastic Drug Screen indicates that several<br />

tumor types, notably melanomas and colorectal<br />

adenocarcinomas, are sensitive to LeTx.<br />

In addition, we have noted that angio-proliferative<br />

tumors are also very sensitive to LeTx treatment.<br />

Consequently we have undertaken a systematic<br />

evaluation of the effects of LeTx upon human<br />

tumor-derived melanomas, colorectal adenocarcinomas,<br />

and Kaposi’s sarcoma. The goal of<br />

this project is to develop novel therapeutic agents<br />

that may be efficacious in the treatment of human<br />

malignancies. In 2004 we successfully obtained<br />

funding from the National Institutes of Health to<br />

evaluate the therapeutic potential of LeTx in the<br />

treatment of Kaposi’s sarcoma.<br />

Molecular mechanisms of LeTx action<br />

The lethal effects of Bacillus anthracis have<br />

been attributed to an exotoxin that it produces.<br />

This exotoxin is composed of three proteins:<br />

protective antigen (PA), edema factor (EF), and<br />

lethal factor (LF). EF is an adenylate cyclase and<br />

together with PA forms a toxin referred to as<br />

edema toxin. LF is a Zn 2+ -metalloprotease which<br />

together with PA forms a toxin referred to as lethal<br />

toxin. LeTx is the dominant virulence factor<br />

produced by B. anthracis and is the major cause of<br />

death in infected animals. The goal of this project<br />

is to develop a detailed molecular understanding of<br />

20


the LF/MEK interaction that will facilitate the<br />

development of therapeutic agents for anthrax. In<br />

2004, we identified a cluster of surface-exposed<br />

residues of LF that are distal to the catalytic site<br />

and are essential for its catalytic activity (Fig. 1).<br />

Details of this study were published in the Journal<br />

of Biological Chemistry.<br />

Staff notes<br />

Sherrie Boone, who has served as a technician<br />

in the lab since 2001, has left us. She was replaced<br />

in February 2004 by John Young, an M.Sc.<br />

graduate from the University of Oregon. John has<br />

initiated studies of LeTx and Kaposi’s sarcoma<br />

and has played a significant role in our<br />

identification of novel regions of LF that are<br />

required for its activity. Xudong Liang, a<br />

postdoctoral fellow who joined us in 2002, has<br />

moved on to a new position at the University of<br />

Minnesota. In his place we welcome Paul<br />

Spilotro, who joined us in August. Paul is<br />

currently evaluating MEK signaling in human<br />

fibrosarcoma. Elissa Boguslawski and Lisa<br />

Orcasitas joined our team in September. Elissa<br />

will serve as our vivarium technician in charge of<br />

murine studies, including xenografts. Lisa is a<br />

Bridges to the Baccalaureate student and is<br />

currently making sarcoma tissue microarrays so<br />

that she can evaluate MEK signaling in human<br />

tumor samples. In the summer of 2004, the lab<br />

was joined by Mia Hemmes, an undergraduate<br />

student from Michigan State University, who<br />

undertook a cytogenetic analysis of a primary<br />

human sarcoma-derived cell line. As well, we<br />

hosted Ricky Gonzalez and Lynda Gladding, two<br />

Grand Rapids area high school students interested<br />

in careers in biological research. Ricky and Lynda<br />

evaluated the sensitivity of murine endothelial<br />

cells to LeTx.<br />

Figure 1. A surface plot of anthrax LF highlighting mutagenized residues. A space-filled surface plot of LF<br />

was generated using Protein Explorer freeware. Residues identified as being critical for LF activity are colored<br />

yellow (K294), green (L293), red (L514), purple (N516), and orange (R491). Residues found to play a neutral or<br />

marginal role in LF activity are white. The NH 2 -terminus of MEK is indicated in black. A magnified image of this<br />

region shows that the critical residues are organized side-by-side in a focused band (KLLNR), which lies at one end<br />

of the catalytic groove.<br />

21


External Collaborators<br />

Jean-François Bodart, Université des Sciences et Technologies de Lille, France.<br />

Art Frankel, Wake Forest University, Winston-Salem, North Carolina<br />

Silvio Gutkind, National Institute of Dental and Craniofacial Research, Bethesda, Maryland<br />

Stephen Leppla, National Institute of Allergies and Infectious Diseases, Bethesda, Maryland<br />

Recent Publications<br />

Bodart, J.-F., and N.S. Duesbery. In press. Xenopus tropicalis oocytes: more than just a beautiful genome.<br />

In Cell Biology and Signal Transduction, J. Liu, ed. Humana Press.<br />

Singh, Yogendra, Xudong Liang, and Nicholas S. Duesbery. <strong>2005</strong>. Pathogenesis of Bacillus anthracis:<br />

the role of anthrax toxins. In Microbial Toxins: Molecular and Cellular Biology, T. Proft, ed.<br />

Norwich, U.K.: Horizon <strong>Scientific</strong>, pp. 285–312.<br />

Liang, Xudong, John J. Young, Sherrie A. Boone, David S. Waugh, and Nicholas S. Duesbery. 2004.<br />

Involvement of domain II in toxicity of anthrax lethal factor. Journal of Biological Chemistry<br />

279(50): 52473–52478.<br />

From left to right, standing: Lee, Duesbery, Depeille, Young, Cumbo-Nacheli, Spilotro<br />

seated: Holman, Boguslawski, Wagner<br />

22


Microinjection of embryonic stem cells<br />

In these photos, technicians are microinjecting embryonic stem cells into mouse blastocysts for gene targeting studies.<br />

(Photos by Julie Koeman)<br />

23


Vivarium and Laboratory of Transgenics<br />

Bryn Eagleson, A.A., RLATG<br />

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

Bionetics at the National Cancer Institute’s Frederick Cancer Research and<br />

Development Center (NCI-FCRDC) in Maryland. In 1983, she joined the Johnson &<br />

Johnson Biotechnology Center in San Diego, California. In 1988, she returned to the<br />

NCI-FCRDC, where she continued to develop her skills in transgenic technology and<br />

managed the transgenic mouse colony. During this time Ms. Eagleson attended<br />

Frederick Community College and Hood College in Frederick, Maryland. In 1999,<br />

she joined VARI as the Vivarium Director and Transgenics Special Program Manager.<br />

Managerial staff<br />

Jason Martin, RLATG<br />

Laboratory Members<br />

Technical staff<br />

Dawna Dylewski, B.S.<br />

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

Elissa Boguslawksi, RALAT<br />

Jamie Bondsfield<br />

Sylvia Marinelli<br />

Vivarium staff<br />

Lisa DeCamp, B.S.<br />

Laura Sixburry, B.S.<br />

Crystal Brady<br />

Kathy Geil<br />

Jarred Grams<br />

Tina Schumaker<br />

Research Interests<br />

T<br />

he<br />

goal of the vivarium and the<br />

transgenics laboratory is to develop,<br />

provide, and support high-quality mouse<br />

modeling services for the Van Andel Research<br />

Institute investigators, Michigan Technology Tri-<br />

Corridor collaborators, and the greater research<br />

community. We use two Topaz Technologies<br />

software products, Granite and Scion, for<br />

integrated management of the vivarium finances,<br />

the mouse breeding colony, and the Institutional<br />

Animal Care and Use Committee (IACUC)<br />

protocols and records. Imaging equipment, such<br />

as the PIXImus mouse densitometer and the<br />

Acuson Sequoia 512 ultrasound machine, is<br />

available for noninvasive imaging of mice.<br />

VetScan blood chemistry and hematology<br />

analyzers are now available for blood analysis.<br />

Also provided by the vivarium technical staff are<br />

an extensive xenograft model development and<br />

analysis service, rederivation, surgery, dissection,<br />

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

Transgenics<br />

Fertilized eggs contain two pronuclei, one that<br />

is derived from the egg and contains the maternal<br />

genetic material and one derived from the sperm<br />

that contains the paternal genetic material.<br />

As development proceeds, these two pronuclei<br />

fuse, the genetic material mixes, and the cell<br />

proceeds to divide and develop into an embryo.<br />

Transgenic mice are produced by injecting small<br />

quantities of foreign DNA (the transgene) into a<br />

pronucleus of a one-cell fertilized egg.<br />

DNA microinjected into a pronucleus randomly<br />

integrates into the mouse genome and will<br />

theoretically be present in every cell of the<br />

resulting organism. Expression of the transgene is<br />

controlled by elements called promoters that are<br />

genetically engineered into the transgenic DNA.<br />

Depending on the selection of the promoter, the<br />

transgene can be expressed in every cell of the<br />

mouse or in specific cell populations such as<br />

neurons, skin cells, or blood cells.<br />

Temporal expression of the transgene during<br />

development can also be controlled by genetic<br />

engineering. These transgenic mice are excellent<br />

models for studying the expression and function of<br />

the transgene in vivo.<br />

24


Standing from left to right: Bondsfield, Sixbury, Grams, DeCamp, Brady, Guikema, Martin<br />

seated from left to right: Schumaker, Dylewski, Marinelli, Eagleson, Boguslawski<br />

25


Bioinformatics Special Program<br />

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

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

of Medicine in 2000. Prior to obtaining his degree, he worked as a software<br />

engineer at YSI, Inc., where he wrote operating systems for embedded computer<br />

devices. Dr. Furge did his postdoctoral work in the laboratory of Dr. George Vande<br />

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

Laboratory Members<br />

Staff<br />

Karl Dykema, B.A.<br />

Research Interests<br />

Ashigh-throughput biotechnologies such<br />

as DNA sequencing, gene expression<br />

microarrays, and genotyping become<br />

more available to researchers, analysis of the data<br />

produced by these technologies becomes<br />

increasingly difficult. Disciplines such as<br />

bioinformatics and computational biology have<br />

recently emerged to help develop methods that<br />

assist in the storage, distribution, integration, and<br />

analysis of these data sets. The bioinformatics<br />

program at VARI is currently focused on using<br />

computational approaches to understand how<br />

cancer cells differ from normal cells at the<br />

molecular level. In addition, VARI is also part of<br />

the overall bioinformatics effort in the state of<br />

Michigan through the Michigan Center for<br />

Biological Information.<br />

Laboratory members from the bioinformatics<br />

program have worked on a wide variety of projects<br />

to further the research efforts at VARI in 2004.<br />

Recently, we constructed a program to identify<br />

short sequences within genes that are likely to be<br />

responsive to siRNA interference. siRNAs are<br />

short, double-stranded DNA sequences that when<br />

introduced into living cells bind to the RNA<br />

produced from a gene of interest and inhibit<br />

expression of the gene. As such, the introduction<br />

of siRNAs is becoming a more widely used<br />

technique to examine the role of individual genes<br />

in both cancerous and noncancerous cells.<br />

The program we developed contained a new<br />

algorithm, developed by one of the VARI<br />

investigators, to identify potential sites within<br />

genes that would be sensitive to siRNAs. In<br />

another project, we assisted the Microarray<br />

Technology Laboratory in the placement of quality<br />

control markers on gene expression microarrays.<br />

These microarrays contain more than 20,000<br />

unique DNA fragments that are robotically placed<br />

on a small glass slide. In order to ensure the DNA<br />

fragments are placed correctly, the quality control<br />

markers are robotically placed on the arrays in a<br />

very specific pattern as the arrays are constructed.<br />

As each array is produced, a quick visual<br />

inspection of the pattern of quality control markers<br />

can be used to verify that all the DNA fragments<br />

were placed on the arrays correctly.<br />

In addition to assisting other VARI research<br />

labs, our group has a special focus on<br />

understanding how cytogenetic abnormalities<br />

influence cancer development and progression.<br />

Many cancer types, including liver and kidney<br />

cancers, are associated with defined sets of DNA<br />

gains and losses. For example, the majority of<br />

hepatocellular carcinomas contain extra copies of<br />

chromosome 1p and lack copies of chromosome<br />

4q. In contrast, the majority of clear cell renal cell<br />

carcinomas contain an extra copy of chromosome<br />

5q and lack a copy of chromosome 3p.<br />

Interestingly, we and other groups have noticed<br />

that transcription is dramatically disrupted within<br />

regions of DNA copy number change. We are<br />

currently developing and testing a number of<br />

different algorithms to identify these disrupted<br />

regions using gene expression data. In addition,<br />

we are developing methods to identify key<br />

regulatory genes that lie within the abnormal<br />

region and may be involved in tumor development<br />

and/or progression.<br />

26


External Collaborators<br />

Xin Chen, Stanford University, Stanford, California<br />

Recent Publications<br />

Yang, X.J., M.-H. Tan, H.L. Kim, J.A. Ditlev, M.W. Betten, C.E. Png, E.J. Kort, K. Futami, K.J. Dykema,<br />

K.A. Furge, M. Takahashi, H. Kanayama, P.H. Tan, B.S. Teh, C. Luan, et al. In press. A molecular<br />

classification of papillary renal cell carcinoma. Cancer Research.<br />

Dykema, K.J., and K.A. Furge. 2004. Diminished transcription of chromosome 4q is inversely related to<br />

local spread of hepatocellular carcinoma. Genes, Chromosomes and Cancer 41(4): 390–394.<br />

Furge, Kyle A., Kerry A. Lucas, Masayuki Takahashi, Jun Sugimura, Eric J. Kort, Hiro-omi Kanayama,<br />

Susumu Kagawa, Philip Hoekstra, John Curry, Ximing J. Yang, and Bin T. Teh. 2004. Robust<br />

classification of renal cell carcinoma based on gene expression data and predicted cytogenetic<br />

profiles. Cancer Research 64(12): 4117–4121.<br />

Haven, C.J., V.M. Howell, P.H.C. Eilers, R. Dunne, M. Takahashi, M. van Puijenbroek, K. Furge, J. Kievit,<br />

M.-H. Tan, G.J. Fleuren, B.G. Robinson, L.W. Delbridge, J. Philips, A.E. Nelson, U. Krause, et al.<br />

2004. Gene expression of parathyroid tumors: molecular subclassification and identification of the<br />

potential malignant phenotype. Cancer Research 64(20): 7405–7411.<br />

Lindvall, Charlotta, Kyle Furge, Magnus Björkholm, Xiang Guo, Brian Haab, Elisabeth Blennow, Magnus<br />

Nordenskjöld, and Bin Tean Teh. 2004. Combined genetic and transcriptional profiling of acute<br />

myeloid leukemia with normal and complex karyotypes. Haematologica 89(9): 1072–1081.<br />

Sugimura, Jun, Richard S. Foster, Oscar W. Cummings, Eric J. Kort, Masayuki Takahashi, Todd T. Lavery,<br />

Kyle A. Furge, Lawrence H. Einhorn, and Bin Tean Teh. 2004. Gene expression profiling of earlyand<br />

late-relapse nonseminomatous germ cell tumor and primitive neuroectodermal tumor of the testis.<br />

Clinical Cancer Research 10(7): 2368–2378.<br />

Tan, Min-Han, Craig G. Rogers, Jeffrey T. Cooper, Jonathon A. Ditlev, Thomas J. Maatman, Ximing Yang,<br />

Kyle A. Furge, and Bin Tean Teh. 2004. Gene expression profiling of renal cell carcinoma. Clinical<br />

Cancer Research 10(18): 6315S–6321S.<br />

Yang, Ximing J., Jun Sugimura, Maria S. Tretiakova, Kyle Furge, Gregory Zagaja, Mitchell Sokoloff,<br />

Michael Pins, Raymond Bergan, David J. Grignon, Walter M. Stadler, Nicholas J. Vogelzang, and Bin<br />

Tean Teh. 2004. Gene expression profiling of renal medullary carcinoma: potential clinical relevance.<br />

Cancer 100(5): 976–985.<br />

From left to right: Furge, Dykema<br />

27


Laboratory of Cancer Immunodiagnostics<br />

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

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

Berkeley in 1998. He then served as a postdoctoral fellow in the laboratory of<br />

Patrick Brown in the Department of Biochemistry at Stanford University. Dr. Haab<br />

joined VARI as a Special Program Investigator in May 2000.<br />

Staff<br />

Songming Chen, Ph.D.<br />

Michael Shafer, Ph.D.<br />

Sara Forrester, B.S.<br />

Darren Hamelinck, B.S.<br />

Randall Orchekowski, B.S.<br />

Laboratory Members<br />

Students<br />

Thomas LaRoche<br />

Richard Schildhouse<br />

Research Interests<br />

Many cancers are difficult to detect at<br />

early stages and are often diagnosed too<br />

late to allow curative treatment. Earlier<br />

and more accurate detection of cancer could lead<br />

to better outcomes for many patients. A greater<br />

knowledge of the molecular changes associated<br />

with the development of cancer could lead to a<br />

better understanding of disease mechanisms and<br />

improved diagnostic tests. The Haab laboratory<br />

is developing novel experimental approaches to<br />

gather such molecular information and to use it<br />

for the diagnosis of cancer. Our goal is that these<br />

studies will produce measurable benefits to<br />

cancer patients.<br />

We are taking a variety of approaches to<br />

identify changes in blood protein composition that<br />

define cancer and that could be diagnostically<br />

useful. Microarray methods have features that are<br />

particularly useful for this research. We have been<br />

developing several related antibody and protein<br />

microarray methods—such as two-color<br />

competition assays, sandwich assays, glycan<br />

detection, and antigen detection of antibodies—to<br />

analyze serum samples from cancer patients and<br />

controls. With these methods (Fig. 1), we can<br />

efficiently probe the binding to many different<br />

antibodies and proteins and explore the use of<br />

multiple measurements for classifying samples.<br />

Previous technological developments that are now<br />

in routine use include a high-sensitivity detection<br />

method (two-color rolling-circle amplification)<br />

and a new method for isolating multiple<br />

microarrays on a single slide, allowing highthroughput<br />

sample processing. Our research also<br />

makes use of gel and chromatographic separations<br />

and mass spectrometry to provide complementary<br />

experimental information.<br />

An ongoing study of protein profiles from the<br />

sera of pancreatic cancer patients and controls (in<br />

collaboration with Randall Brand and George<br />

Vande Woude) shows the value of antibody<br />

microarrays for diagnostics research. Protein<br />

profiles from antibody microarrays targeting a<br />

wide variety of proteins—such as those previously<br />

associated with cancer, involved in biological<br />

systems altered by cancer, or having elevated levels<br />

in the tumor environment—are revealing many<br />

proteins at either higher or lower abundances in the<br />

cancer patients. Some of the proteins were not<br />

known to be associated with pancreatic cancer and<br />

may contribute to improved early detection of the<br />

disease. The serum samples can be classified as<br />

from cancer or control patients using the antibody<br />

measurements. The accuracy of the classifications<br />

was greatly improved with multiple measurements<br />

in combination (relative to using individual<br />

measurements); we need to further develop this<br />

approach for cancer diagnostics.<br />

A recent modification to this technology<br />

measures alterations in the glycosylation state of<br />

the proteins. Glycosylation—the attachment of<br />

specific carbohydrate structures to proteins—<br />

plays a major role in determining protein function,<br />

and glycosylation alterations have been associated<br />

with the development and progression of cancer.<br />

The ability to conveniently measure changes in<br />

specific carbohydrates on different proteins could<br />

be valuable in identifying the changes most<br />

28


associated with cancer and thus of use for<br />

diagnostics. We have methods for detecting<br />

changes in glycosylation on proteins captured by<br />

antibody microarrays (see Fig. 1C) and are using<br />

those methods to profile the glycosylation<br />

alterations on serum proteins from cancer patients<br />

and control subjects.<br />

We use protein microarrays (see Fig. 1D) to<br />

gather additional information about changes<br />

occurring in the blood of cancer patients. Some<br />

tumor proteins elicit the production of antibodies<br />

targeting those proteins. The identification and<br />

measurement of tumor-recognizing antibodies<br />

could provide information about molecular<br />

alterations in the tumor and be valuable in cancer<br />

detection. The protein microarray is an ideal<br />

screening tool for that purpose. In collaboration<br />

with Gilbert Omenn and Samir Hanash,<br />

microarrays of tumor-derived proteins from<br />

cancer cell lines are probed with sera from cancer<br />

patients to identify proteins recognized by the<br />

patients’ antibodies. An ongoing study of serum<br />

from prostate cancer patients (in collaboration<br />

with Alan Partin) has identified several proteins<br />

that could be involved in an immune response.<br />

We are characterizing the nature of the responses<br />

and the proteins involved.<br />

The above methods may be applied in a novel<br />

way to further their effectiveness. We have begun<br />

to develop and improve existing diagnostic markers<br />

through the use of longitudinal measurements<br />

(serial measurements over time). In a collaboration<br />

with William Catalona, Robert Vessella, and Ziding<br />

Feng, we are looking at changes over time in the<br />

concentrations of several serum proteins leading up<br />

to disease recurrence in prostate cancer patients.<br />

We hypothesize that the use of individualized<br />

thresholds defining abnormal protein levels,<br />

defined by each person’s history of measurements,<br />

will yield improved diagnostic accuracy over the<br />

use of single, population-wide thresholds. Our<br />

hypothesis has been supported in some individual<br />

demonstrations, and we now have an experimental<br />

system for systematically exploring it for a large<br />

number of proteins and many patients.<br />

Figure 1. Antibody and protein microarray<br />

formats. A) Two-color competition assay. Two pools<br />

of proteins, respectively labeled with biotin and<br />

digoxigenin tags, are mixed and co-incubated on<br />

antibody microarrays. The relative amount of binding<br />

to each antibody from the two pools is determined<br />

through detection of the biotin and digoxigenin tags.<br />

B) Sandwich assay. After incubation of a protein<br />

sample on an antibody microarray, the amount of<br />

protein binding to each antibody is measured using a<br />

second antibody that targets the captured proteins.<br />

C) Glycan detection. A pool of digoxigenin-labeled<br />

proteins is incubated on antibody microarrays, and the<br />

level of protein binding at each antibody is determined<br />

by detection of the digoxigenin tag. The amount of a<br />

particular glycan on the captured proteins is detected<br />

using a biotin-labeled protein that specifically binds to<br />

that glycan. D) Protein array detection of antibodies.<br />

Serum samples are incubated on arrays containing a<br />

variety of tumor-derived proteins. Antibodies in the<br />

serum samples that recognize and bind to the<br />

proteins are detected using a secondary antibody that<br />

binds to human antibodies.<br />

We are seeking to apply the methods<br />

described above in ways that will have a positive<br />

result in terms of cancer care. The incorporation<br />

of mass spectrometry methods, through<br />

collaborations with Greg Cavey at VARI and<br />

members of the Michigan Proteome Consortium,<br />

will provide more opportunities for discovery.<br />

The further testing of the value of these tools is<br />

being pursued through local clinical oncology<br />

programs. The access to clinical practice is<br />

especially valuable for translating the<br />

development and discovery in our laboratory into<br />

benefits for cancer patients.<br />

External Collaborators<br />

Phil Andrews, Gilbert Omenn, and Diane Simeone, University of Michigan, Ann Arbor<br />

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

E. Brian Butler and Bin S. Teh, Baylor College of Medicine, Houston, Texas<br />

William Catalona, John Grayhack, and Anthony Schaeffer, Northwestern University, Evanston, Illinois<br />

29


Jose Costa and Paul Lizardi, Yale University School of Medicine, New Haven, Connecticut<br />

Deborah Dillon, Brigham and Women’s Hospital, Boston, Massachusetts<br />

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

Jorge Marrero, University of Michigan Hospital, Ann Arbor<br />

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

Peter Schirmacher, University of Cologne, Germany<br />

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

Cornelius Verweij, University of Amsterdam, The Netherlands<br />

Recent Publications<br />

Hamelinck, D., H. Zhou, L. Li, Z. Feng, C. Verweij, D. Dillon, J. Costa, and B.B. Haab. In press.<br />

“Optimized normalization for antibody microarrays and the identification of serum protein<br />

alterations associated with pancreatic cancer.” Molecular & Cellular Proteomics.<br />

Haab, B.B., and P.M. Lizardi. In press. “RCA-enhanced protein detection arrays.” Methods in<br />

Molecular Biology.<br />

Breuhahn, Kai, Sebastian Vreden, Ramsi Haddad, Susanne Beckebaum, Dirk Stippel, Peer Flemming,<br />

Tanja Nussbaum, Wolfgang H. Caselmann, Brian B. Haab, and Peter Schirmacher. 2004.<br />

Molecular profiling of human hepatocellular carcinoma defines mutually exclusive interferon<br />

regulation and insulin-like growth factor II overexpression. Cancer Research 64(17): 6058–6064.<br />

Konwinski, R., R. Haddad, J.A. Chun, S. Klenow, S. Larson, B.B. Haab, and L.L. Furge. 2004.<br />

Oltipraz, 3H-1,2-dithiole-3-thione and sulforaphane induce overlapping and protective antioxidant<br />

responses in murine microglial cells. Toxicology Letters 153(3): 343–355.<br />

Lindvall, Charlotta, Kyle Furge, Magnus Björkholm, Xiang Guo, Brian Haab, Elisabeth Blennow,<br />

Magnus Nordenskjöld, and Bin Tean Teh. 2004. Combined genetic and transcriptional profiling<br />

of acute myeloid leukemia with normal and complex karyotypes. Haematologica 89(9):<br />

1072–1081.<br />

Qiu, Ji, Juan Madoz-Gurpide, David E. Misek, Rork Kuick, Dean E. Brenner, George Michailidis,<br />

Brian B. Haab, Gilbert S. Omenn, and Sam Hanash. 2004. Development of natural protein<br />

microarrays for diagnosing cancer based on an antibody response to tumor antigens. Journal of<br />

Proteome Research 3(2): 261–267.<br />

Zhou, Heping, Kerri Bouwman, Mark Schotanus, Cornelius Verweij, Jorge A. Marrero, Deborah<br />

Dillon, Jose Costa, Paul Lizaardi, and Brian B. Haab. 2004. Two-color, rolling-circle<br />

amplification on antibody microarrays for sensitive, multiplexed serum-protein measurements.<br />

Genome Biology 5(4): R28.<br />

From left to right: Haab, Hamelinck, Orchekowski, Chen, Shafer, Forrester<br />

30


Molecular Medicine and Virology Group<br />

Sheri L. Holmen, Ph.D.<br />

Dr. Holmen received her M.S. in biomedical science from Western Michigan University<br />

in 1995 and her Ph.D. in tumor biology from the Mayo Clinic College of Medicine in<br />

2000. She did her postdoctoral work at VARI in the laboratory of Bart Williams from<br />

2000 to 2003 and became a Junior Investigator at VARI in December 2003.<br />

Laboratory Members<br />

Staff<br />

Marleah Russo<br />

Research Interests<br />

T<br />

he<br />

primary focus of the Molecular<br />

Medicine and Virology Group is to<br />

identify molecules that can be effective<br />

targets for cancer therapy, with the goal of<br />

developing better therapies with fewer side effects.<br />

The sequencing of the human genome has yielded<br />

a wealth of biological data, but we still know<br />

relatively little about which genes are causally<br />

associated with tumor development and which are<br />

only markers of cancer. Because of the high cost<br />

of developing new therapies, it is important that we<br />

identify which genetic changes can be<br />

productively targeted. We are concentrating our<br />

initial efforts on melanoma and glioblastoma,<br />

tumors which demonstrate constitutive activation<br />

of Ras signaling.<br />

The RCAS system<br />

We use a series of replication-competent<br />

retroviral vectors based on the SR-A strain of Rous<br />

sarcoma virus (RSV), a member of the avian<br />

leukosis virus (ALV) family, to study the roles of<br />

different genes in tumor initiation and progression.<br />

RSV is the only known naturally occurring,<br />

replication-competent retrovirus that carries an<br />

oncogene, src. In the RCAS vectors, the region<br />

encoding src (which is dispensable for viral<br />

replication) has been replaced by a synthetic DNA<br />

linker. Foreign genes inserted into this linker are<br />

expressed from the viral LTR promoter via a<br />

subgenomic splice site (just as src is in RSV).<br />

RCAN vectors differ from RCAS vectors in that<br />

they lack the src splice acceptor; the gene of<br />

interest is inserted along with an internal promoter.<br />

Higher-titer viruses subsequently have been<br />

generated by replacing the RSV SR-A pol gene<br />

with the pol gene of the Bryan strain of RSV.<br />

These vectors are termed RCASBP or RCANBP.<br />

The ability of these vectors to infect non-avian<br />

cells relies on expression of the corresponding<br />

receptor on the cell surface. The viral receptor is<br />

typically introduced into mammalian cells (or<br />

mice) via an inducible and/or tissue-specific<br />

transgene. Therefore, this system allows for tissueand<br />

cell-specific targeted infection of mammalian<br />

cells through ectopic expression of the viral<br />

receptor. Alternatively, when targeted infection of<br />

mammalian cells is not required (e.g., in cell<br />

culture), infection can be achieved through the use<br />

of non-avian envelopes, such as the amphotropic<br />

envelope from murine leukemia virus. The<br />

receptor for this envelope is endogenously<br />

expressed on almost all mammalian cells.<br />

We have used the RCASBP/RCANBP family<br />

of retroviral vectors extensively in both cultured<br />

cells and live animals for studies of viral<br />

replication and of cancer modeling in mice. Most<br />

of these studies have analyzed gain-of-function<br />

phenotypes by delivering and overexpressing a<br />

particular gene of interest. Recently, we<br />

engineered the RCANBP vector to reduce the<br />

expression of specific genes through the delivery<br />

of short hairpin RNA sequences. We also<br />

engineered this vector to control the expression of<br />

the inserted gene using the tetracycline (tet)-<br />

regulated system. Sequences inserted into this<br />

region are transcribed from a tet-responsive<br />

element and not the viral LTR. This virus allows<br />

inserted genes to be turned on and off in order to<br />

determine if expression of the gene is required for<br />

tumor initiation, maintenance, and progression.<br />

The ability to turn off gene expression will help<br />

determine if that gene is a good target for therapy.<br />

31


Melanoma<br />

Melanoma is the most rapidly increasing<br />

malignancy among young people in the United<br />

States. If detected early, the disease is easily<br />

treated, but once the disease has metastasized it<br />

has a high mortality rate. Current systemic<br />

therapy for advanced, metastatic melanoma<br />

includes dacarbazine (DTIC) chemotherapy,<br />

either alone or in combination with other agents,<br />

and biological therapy using recombinant<br />

interferon-α (IFN-α) and/or interleukin-2 (IL-2).<br />

However, except on rare occasions, none of these<br />

treatments has produced long-term control of the<br />

disease, and cytokine therapy is associated with<br />

significant toxicities.<br />

We have developed a mouse model of<br />

melanoma based on the avian RCAS/TVA system.<br />

In this model, the retroviral receptor TVA is<br />

expressed under the control of the tyrosinaserelated<br />

protein 2 (TRP2) promoter, which is<br />

expressed in melanocytes. In replicating<br />

mammalian cells that express TVA, the viral vector<br />

is capable of stably integrating into the DNA and<br />

expressing the experimental gene at high levels,<br />

but the virus is replication-defective because viral<br />

RNA and proteins are inefficiently produced.<br />

Therefore, the viral vectors cannot spread in the<br />

target animals; in addition, since little viral<br />

envelope protein is produced, there is no<br />

interference to superinfection. Theoretically, there<br />

is no limit to the number of experimental genes<br />

that can be introduced into a TVA-expressing<br />

mammalian cell. The ability of these cells to be<br />

infected by multiple viruses allows efficient<br />

modeling of melanoma, because multiple<br />

oncogenic alterations can be introduced into the<br />

same cell or animal without the costs associated<br />

with mating multiple strains of transgenic mice.<br />

Activated ras oncogenes, which turn on<br />

mitogen-activated protein kinase (MAPK)<br />

signaling, are detected in approximately 20% of<br />

human melanomas. Recently, activating mutations<br />

in the B-raf gene, which also activate MAPK<br />

signaling, have been found in more than 65% of<br />

malignant melanomas. With mutually exclusive<br />

mutations in ras and B-raf, the MAPK signaling<br />

pathway is constitutively activated in over 85% of<br />

cases of malignant melanoma, indicating its<br />

importance. Overexpression of activated H-ras<br />

(G12V) specifically in melanocytes of Ink4adeficient<br />

mice results in the development of<br />

multiple spontaneous cutaneous melanomas.<br />

However, unlike in the human disease, these<br />

tumors fail to metastasize. A conditional Ink4a/Arf<br />

knockout allele, Ink4a-lox, has been introduced<br />

into the germline of FVB/N mice. The lox sites<br />

flank exons 2 and 3 of this locus such that Cremediated<br />

excision eliminates both p16 INK4A and<br />

p19 ARF . We have recently obtained two<br />

homozygous Ink4a-lox/lox breeding pairs, which<br />

will be crossed to our TRP2-TVA transgenic mice.<br />

We plan to use the mice to study the role of<br />

different genes in melanoma initiation,<br />

maintenance, and progression.<br />

Glioblastoma<br />

Glioblastoma multiforme (GBM) is the most<br />

common and aggressive primary brain tumor. It is<br />

also the most fatal: mean survival is less than one<br />

year from the time of diagnosis, with less than 10%<br />

survival after two years. Despite major<br />

improvements in imaging, radiation, and surgery,<br />

the prognosis for patients with this disease has not<br />

changed in the last 20 years. Recently, genes that<br />

are differentially expressed in tumor tissue relative<br />

to normal brain tissue have been found. However,<br />

those that can be productively targeted for<br />

therapeutic intervention in human patients remain<br />

to be identified.<br />

A mouse model of human GBM based on the<br />

avian RCAS/TVA system has been developed by<br />

Eric Holland. In this model, the retroviral receptor<br />

TVA is expressed under the control of the Nestin<br />

promoter, which is active in neural and glial<br />

progenitors. Intracranial infection of Nestin-TVA<br />

mice with RCASBP(A)-Akt and RCASBP(A)-<br />

KRas induces glioblastomas that are histologically<br />

similar to human GBM. We have generated an<br />

RCANBP(A)TRE-KRas virus in which expression<br />

of K-Ras can be controlled post-delivery. We are<br />

using the Nestin-TVA model to test the function of<br />

this virus in vivo. We plan to use these vectors to<br />

determine if K-Ras expression is required for<br />

tumor maintenance in this system and to identify<br />

which genes are appropriate targets for therapy.<br />

Antiviral strategies<br />

A second, related focus of our group is the<br />

identification of effective antiviral strategies using<br />

the RCASBP/RCANBP family of retroviral<br />

vectors as a model system. Viral diseases pose a<br />

32


major risk to the food supply and to animal<br />

welfare, especially in today’s high-intensity animal<br />

agriculture. Many viruses are highly<br />

communicable and are capable of rapid mutation<br />

to escape immune surveillance; few effective antiviral<br />

drugs are available. Over the past several<br />

years, we have developed strategies aimed at<br />

conferring dominant resistance to viral pathogens<br />

in chickens. These strategies have focused on<br />

manipulating viral (“pathogen-derived resistance”)<br />

and/or cellular genes (“host-derived resistance”) to<br />

express new proteins capable of disrupting the<br />

viral life cycle. The results have provided valuable<br />

information on the biology of avian viruses, as<br />

well as having the potential for practical<br />

application. We are now working to adapt the new<br />

RNA interference (RNAi) technology to the<br />

development of new anti-viral strategies for two<br />

important chicken pathogens, ALV and Marek’s<br />

Disease virus (MDV). These two targets have<br />

been chosen because both are economically<br />

important and because they have distinctly<br />

different infectious cycles that provide different<br />

challenges for RNAi.<br />

External Collaborators<br />

Jerry Dodgson, Michigan State University, East Lansing<br />

Henry Hunt and Huanmin Zhang, Avian Disease and Oncology Laboratory, East Lansing, Michigan<br />

Recent Publications<br />

Ai, Minrong, Sheri L. Holmen, Wim van Hul, Bart O. Williams, and Matthew W. Warman. <strong>2005</strong>.<br />

Reduced affinity to and inhibition by DKK1 form a common mechanism by which high bone<br />

mass–associated missense mutations in LRP5 affect canonical Wnt signaling. Molecular and<br />

Cellular Biology 25(12): 4946–4955.<br />

Holmen, Sheri L., Scott A. Robertson, Cassandra R. Zylstra, and Bart O. Williams. <strong>2005</strong>.<br />

Wnt-independent activation of ß-catenin mediated by a Dkk-1-Frizzled 5 fusion protein.<br />

Biochemical and Biophysical Research Communications 328(2): 533–539.<br />

Holmen, Sheri L., Cassandra R. Zylstra, Aditi Mukherjee, Robert Sigler, Marie-Claude Faugere,<br />

Mary Bouxsein, Lianfu Deng, Thomas Clemens, and Bart O. Williams. <strong>2005</strong>. Essential role of<br />

ß-catenin in postnatal bone acquisition. Journal of Biological Chemistry 280(22): 21162–21168.<br />

Sanchez-Perez, Luis, Timothy Kottke, Rosa Maria Diaz, Atique Ahmed, Jill Thompson, Heung<br />

Chong, Alan Melcher, Sheri Holmen, Gregory Daniels, and Richard G. Vile. <strong>2005</strong>. Potent<br />

selection of antigen loss variants of B16 melanoma following inflammatory killing of<br />

melanocytes in vivo. Cancer Research 65(5): 2009–2017.<br />

Bromberg-White, Jennifer L., Craig P. Webb, Veronique<br />

S. Patacsil, Cindy K. Miranti, Bart O. Williams, and<br />

Sheri L. Holmen. 2004. Delivery of short hairpin<br />

RNA sequences by using a replication-competent<br />

avian retroviral vector. Journal of Virology 78(9):<br />

4914–4916.<br />

Holmen, Sheri L., Troy A. Giambernardi, Cassandra R.<br />

Zylstra, Bree D. Buckner-Berghuis, James H. Resau,<br />

J. Fred Hess, Vaida Glatt, Mary L. Bouxsein,<br />

Minrong Ai, Matthew L. Warman, and Bart O.<br />

Williams. 2004. Decreased BMD and limb<br />

deformities in mice carrying mutations in both Lrp5<br />

and Lrp6. Journal of Bone and Mineral Research<br />

19(12): 2033–2040.<br />

From left to right: Russo, Holmen<br />

33


Laboratory of Integrin Signaling and Tumorigenesis<br />

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

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

and her Ph.D. in biochemistry from Harvard Medical School in 1995. She was a<br />

postdoctoral fellow in the laboratory of Dr. Joan Brugge at ARIAD Pharmaceuticals,<br />

Cambridge, Mass., from 1995 to 1997 and in the Department of Cell Biology at<br />

Harvard Medical School from 1997 to 2000. Dr. Miranti joined VARI as a <strong>Scientific</strong><br />

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

Department of Physiology at Michigan State University.<br />

Laboratory Members<br />

Staff<br />

Suganthi Chinnaswamy, Ph.D.<br />

Mathew Edick, Ph.D.<br />

Robert Long, B.A.<br />

Veronique V. Schulz, B.S.<br />

Student<br />

Erik Freiter<br />

Research Interests<br />

Our laboratory is interested in<br />

understanding the mechanisms by which<br />

integrin receptors, interacting with the<br />

extracellular matrix, regulate cell processes<br />

involved in the development of cancer. Using<br />

tissue culture models, biochemistry, molecular<br />

genetics, and mouse models, we are defining the<br />

cellular and molecular events of integrindependent<br />

adhesion and downstream signaling that<br />

are important in melanoma and prostate<br />

tumorigenesis and metastasis.<br />

Integrins are transmembrane proteins that<br />

serve as receptors for extracellular matrix (ECM)<br />

proteins. By interacting with the ECM, integrins<br />

stimulate intracellular signaling transduction<br />

pathways that regulate cell shape, proliferation,<br />

migration, survival, gene expression, and<br />

differentiation. Integrins do not act autonomously;<br />

they are involved in “crosstalk” with receptor<br />

tyrosine kinases (RTKs) to regulate many cellular<br />

processes. Studies in our lab, for example, indicate<br />

that integrin-mediated adhesion to ECM proteins<br />

activates the epidermal growth factor receptors<br />

EGFR and ErbB2 and the HGF/SF receptor Met.<br />

Integrin-mediated activation of these RTKs is<br />

ligand-independent and is required for activation<br />

of a subset of intracellular signaling molecules in<br />

response to cell adhesion.<br />

The prostate gland and cancer<br />

Tumors that develop in cells of epithelial origin,<br />

i.e., carcinomas, represent the largest tumor burden<br />

in the United States. Prostate cancer is the most<br />

frequently diagnosed cancer in U.S. men and the<br />

second leading cause of cancer death in men.<br />

Eighty percent of human prostate tumors arise in the<br />

peripheral zone of the gland and are primarily<br />

confined to the intermediate basal and secretory<br />

epithelial cells. Patients who at the time of diagnosis<br />

have androgen-dependent and organ-confined<br />

prostate cancer are relatively easy to cure through<br />

radical prostatectomy or localized radiotherapy.<br />

However, patients with aggressive and metastatic<br />

disease have fewer options. Androgen ablation can<br />

significantly reduce the tumor burden in these<br />

patients, but the potential for relapse and the<br />

development of androgen-independent cancer is<br />

high. Currently there are no effective treatments for<br />

patients who reach this stage of disease.<br />

In the human prostate secretory glands, basal<br />

epithelial cells form a contiguous layer adjacent to<br />

the basement membrane. Upon them rests a layer of<br />

secretory luminal cells, forming a stratified<br />

epithelium. The basal cells express a broad<br />

repertoire of integrins, including α2, α3, α6, β1, and<br />

β4. The secretory cells express primarily α6 and β1,<br />

with some α2. In vivo, basal cells secrete and<br />

organize a laminin 5–containing basement<br />

membrane that also contains collagens IV and VII<br />

and laminin 10. The basal cells bind to laminin 5<br />

and collegen VII through α6β4 to form<br />

hemidesmosomal complexes at the basal surface.<br />

Basal cells adhering to this matrix respond to growth<br />

factors secreted by the surrounding stroma,<br />

including EGF and HGF. They proliferate and give<br />

rise to the nonproliferating secretory cells through<br />

34


the generation of an intermediate, transiently<br />

amplifying cell population that has traits of both cell<br />

types. In primary prostate tumors, α6β4 integrin<br />

and its ligands, laminin 5 and collagen VII, are lost.<br />

The tumor cells, unlike normal secretory cells,<br />

develop the ability to adhere, via α2β1 and α6β1, to<br />

an altered basement membrane consisting of<br />

collagen IV and laminin 10. Thus, tumor cells have<br />

characteristics of both basal and secretory cells, but<br />

not all the properties of either.<br />

Whether the tumor cells are derived from the<br />

transient differentiating population of basal cells or<br />

from differentiated secretory cells has not been<br />

unequivocally determined, but it is clear that the<br />

way in which these cells interact with the ECM has<br />

been changed. If tumor cells are derived from<br />

basal cells, they are now interacting with an altered<br />

matrix and using different integrins to engage it. If<br />

they are derived from the secretory cells, the tumor<br />

cells are now engaging a matrix, which they did<br />

not do previously. A fundamental question in our<br />

lab is whether the changes in integrin/matrix<br />

interactions that occur in tumor cells are required<br />

for or help to drive the survival of tumor cells.<br />

The role of integrins and RTKs in<br />

prostate epithelial cell survival<br />

Increased cell survival due to resistance to cell<br />

death is a prerequisite for tumorigenesis. Several<br />

reports have suggested that the signaling pathways<br />

that regulate cell survival in normal prostate<br />

epithelial cells are different from those in prostate<br />

tumor cells. How integrin engagement of different<br />

ECMs regulates survival pathways is not known.<br />

We have recently found that integrin-induced<br />

activation of both EGFR and c-Met in primary<br />

prostate epithelial cells is required for cell survival<br />

in the absence of growth factors. Our goal is to<br />

determine how integrin activation of EGFR and<br />

c-Met regulate cell survival.<br />

We have previously shown that integrin<br />

activation of EGFR is required for integrinmediated<br />

induction of the Ras/Erk and PI3K/Akt<br />

signaling pathways. Recent studies indicate that<br />

integrin activation of Src depends on c-Met.<br />

Interestingly, inhibition of either Src or Ras/Erk<br />

signaling—but not PI3K/Akt signaling—induces<br />

cell death in primary prostate cells even when they<br />

are still adherent to matrix. Together these data<br />

indicate that Src signals generated by c-Met, as well<br />

as Ras/Erk signals generated by EGFR, are driving<br />

cell survival in primary cells (Fig. 1). We are<br />

currently exploring the downstream events that Src<br />

and Erk regulate to maintain cell survival on ECM.<br />

Figure 1. Met and EGFR both act independently<br />

to regulate integrin-mediated survival of primary<br />

prostate epithelial cells. Activation of Src and Erk<br />

through Met and EGFR, respectively, are proposed to<br />

be involved in integrin-mediated survival.<br />

We are also exploring the mechanisms by<br />

which integrins activate EGFR and ErbB2. This<br />

activation is ligand independent, requires only the<br />

cytoplasmic domain of EGFR, and stimulates the<br />

phosphorylation of only a subset of sites on EGFR.<br />

ErbB2 activation depends on EGFR, suggesting<br />

that integrins induce the formation of an<br />

EGFR/ErbB2 heterodimer. Integrin activation of<br />

Src or FAK is not required, but integrin activation<br />

of a phosphatase may be involved; we are currently<br />

investigating the role of several phosphatases.<br />

Integrin and RTK crosstalk in<br />

prostate cancer metastasis<br />

Death from prostate cancer is due to the<br />

development of metastatic disease, which is<br />

difficult to control. The mechanisms involved in<br />

progression to metastatic disease are not<br />

understood. Our approach is to characterize genes<br />

that are specifically associated with metastatic<br />

prostate cancer. CD82/KAI1 is a metastasis<br />

suppressor gene whose expression is specifically<br />

lost in metastatic cancer but not in primary tumors.<br />

CD82/KAI1 is known to associate with both<br />

integrins and RTKs. Our goal has been to<br />

determine how loss of CD82/KAI1 expression<br />

promotes metastasis by studying the role of<br />

CD82/KAI1 in integrin and RTK crosstalk.<br />

35


During prostate cancer progression there is a<br />

shift in the expression of laminin-specific integrins:<br />

β4 integrins are lost, and there is a concomitant<br />

increase in α6β1 and α3β1, which both interact<br />

with CD82/KAI1. We predict that the loss of<br />

CD82/KAI1 alters the function of α6β1 and α3β1.<br />

Using primary prostate epithelial cells, which<br />

express high levels of CD82/KAI1, as well as<br />

several prostate tumor cell lines that do not, we are<br />

exploring the role of CD82/KAI1 in regulating<br />

α6β1- and α3β1-mediated cell adhesion, migration,<br />

and integrin signaling. We have found that<br />

overexpression of CD82/KAI1 in tumor cells<br />

suppresses laminin-specific migration and invasion;<br />

integrin-induced EGFR and c-Met receptor<br />

activation; Src and Lyn activation; and activation of<br />

the Src/Lyn substrates Cas and FAK. Furthermore,<br />

integrin activation of Src is dependent on c-Met, and<br />

laminin-mediated invasion depends on both Src and<br />

c-Met. Together these data indicate that<br />

CD82/KAI1 normally acts to regulate integrin<br />

signaling to c-Met such that upon its loss of<br />

expression in tumor cells, signaling through c-Met<br />

to Src is increased, leading to increased motility and<br />

invasion (Fig. 2). We are currently determining the<br />

mechanism by which CD82/KAI1 down-regulates<br />

c-Met signaling. In reciprocal experiments, we are<br />

inhibiting the expression of CD82/KAI1 in primary<br />

cells using siRNA and mouse models.<br />

Integrin regulation of melanoma<br />

progression by PKC<br />

The incidence of melanoma has been steadily<br />

increasing over the last 10 years. If diagnosed at<br />

an early stage, melanoma is curable, but once it has<br />

become invasive, it progresses very rapidly and is<br />

virtually untreatable. Metastasis and invasion by<br />

tumor cells require the activity of integrins, and in<br />

melanoma, expression of the αvβ3 integrin is<br />

induced during the development of invasive<br />

disease. Therefore, an understanding of how the<br />

αvβ3 integrin functions to regulate invasion will<br />

help our understanding of melanoma metastasis.<br />

We have been focusing our attention on two<br />

signaling molecules, PKCα and Src, both of which<br />

are regulated by the αvβ3 integrin and whose<br />

activities are enhanced in metastatic melanoma.<br />

Adhesion of normal melanocytes to extracellular<br />

matrix induces the formation of focal adhesion<br />

complexes and actin stress fibers. However, in a<br />

highly invasive metastatic melanoma cell line,<br />

C8161.9, these structures are absent. We have<br />

shown that the levels of Src activity and PKCα<br />

protein are elevated in these cells, and<br />

overexpression of PKCα in immortalized normal<br />

melanocytes is sufficient to confer an invasive<br />

phenotype in vitro. We have found that the activity<br />

of Rac, a small GTPase that is required for the<br />

formation of lamellipodia, is elevated in these cells<br />

and that inhibition of PKCα blocks Rac activity.<br />

The activity of the small GTPase Rho, which is<br />

involved in stress fiber and focal adhesion<br />

formation, is negatively regulated by active Src.<br />

Thus PKCα, acting to enhance Rac, and active Src,<br />

acting to inhibit Rho, together drive the formation<br />

of lamellipodia and the dissolution of stress fibers<br />

and focal adhesions, leading to increased motility<br />

(Fig. 3). We are currently exploring how Src is<br />

activated in melanoma cells, as well as the effects<br />

of blocking Src and PKCα expression with siRNA<br />

on migration and invasion.<br />

Figure 2. CD82 reexpression in prostate tumor<br />

cells inhibits invasion by blocking integrinmediated<br />

signaling to Met and Src.<br />

Figure 3. PKCa and Src cooperate to enhance<br />

migration and invasion in melanoma cells by<br />

differentially targeting Rac and Rho.<br />

36


External Collaborators<br />

Beatrice Knudsen, Fred Hutchinson Cancer Research Center, Seattle, Washington<br />

Senthil Muthuswamy, Cold Spring Harbor Laboratory, New York<br />

Recent Publications<br />

Sridhar, S.C., and C.K. Miranti. In press. Tumor metastasis suppressor KAI1/CD82 is a tetraspanin.<br />

In Contemporary Cancer Research: Metastasis, C. Rinker-Schaeffer, M. Sokoloff, and D. Yamada,<br />

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

Bill, Heather M., Beatrice Knudsen, Sheri L. Moores, Senthil K. Muthuswamy, Vikram R. Rao, Joan S.<br />

Brugge, and Cindy K. Miranti. 2004. Epidermal growth factor receptor–dependent regulation of<br />

integrin-mediated signaling and cell cycle entry in epithelial cells. Molecular and Cellular Biology<br />

24(19): 8586–8599.<br />

Lee, Chong-Chou, Andrew J. Putnam, Cindy K. Miranti, Margaret Gustafson, Ling-Mei Wang, George F.<br />

Vande Woude, and Chong-Feng Gao. 2004. Overexpression of sprouty-2 inhibits HGF/SF-mediated<br />

cell growth, invasion, migration, and cytokinesis. Oncogene 23(30): 5193–5202.<br />

From left to right: Schulz, Edick, Miranti, Long, Freiter, Sridhar<br />

37


Laboratory of Analytical, Cellular, and Molecular Microscopy<br />

and<br />

Laboratory of Microarray Technology and Molecular Diagnostics<br />

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

Dr. Resau received his Ph.D. from the University of Maryland School of Medicine in<br />

1985. He has been involved in clinical and basic science imaging and pathologyrelated<br />

research since 1972. Between 1968 and 1994, he was in the U.S. Army<br />

(active duty and reserve) and served in Vietnam. From 1985 until 1992, Dr. Resau<br />

was a tenured faculty member at the University of Maryland School of Medicine,<br />

Department of Pathology. Dr. Resau was the Director of the Analytical, Cellular and<br />

Molecular Microscopy Laboratory in the Advanced BioScience Laboratories–Basic<br />

Research Program at the National Cancer Institute–Frederick Cancer Research and<br />

Development Center, Maryland, from 1992 to 1999. He joined VARI as a Special<br />

Program Senior <strong>Scientific</strong> Investigator in June 1999 and in 2003 was promoted to Deputy Director. In 2004, Dr.<br />

Resau assumed as well the direction of the Laboratory of Microarray Technology to consolidate the imaging and<br />

quantification of clinical samples in a CLIA-type research laboratory program.<br />

Staff<br />

Eric Kort, M.S.<br />

Bree Berghuis, B.S.,<br />

HTL (ASCP), QIHC<br />

Pete Haak, B.S.<br />

Eric Hudson, B.S.<br />

Paul Norton, B.S.<br />

J.C. Goolsby<br />

Laboratory Members<br />

Consulting Veterinarian<br />

Robert Sigler, D.V.M., Ph.D.<br />

Students<br />

Hien Dang<br />

Brandon Leeser<br />

Amy Percival<br />

Huang Tran<br />

Research Interests<br />

T<br />

he<br />

Laboratory of Analytical, Cellular,<br />

and Molecular Microscopy (ACMM)<br />

and the Laboratory of Microarray<br />

Technology and Molecular Diagnostics (MTMD)<br />

are organized and equipped to produce highresolution<br />

images, genomic arrays, and<br />

bioinformatics data that support the cellular and<br />

molecular biology programs of the Institute. Our<br />

laboratories collaborate with the investigators to<br />

improve the understanding, diagnosis, and<br />

characterization of disease, injury, and<br />

differentiation. Although we primarily study<br />

cancer, we work with investigators in and out of<br />

the Institute on a variety of diseases and<br />

processes. As examples, during the past year, we<br />

have used our microscopes to produce intravital<br />

images of the Met protein in living cells and<br />

animals; localized and quantified a unique<br />

transcriptional protein in human cells; worked<br />

closely with VARI’s Bart Williams to characterize<br />

the phenotypic changes in transgenic mice<br />

expressing mutations related to Wnt and the<br />

Lrp5/Lrp6 genes; began to image Art Alberts’<br />

small GTPase proteins in living cells; and<br />

collaborated in studies of the expression of c-Met<br />

in a series of primary human breast cancers.<br />

During 2004, we added two significant<br />

instruments to the ACMM laboratory. The first is<br />

the Aperio Scanscope, which enables us to<br />

digitize a 1 × 3-inch microscope slide in full<br />

color such that it can be analyzed, quantified, and<br />

shared throughout the digital network. The<br />

second is the Ventana automated immunostainer,<br />

which can carry out FISH, ISH, IHC, and<br />

array spotting in a programmed, specific, and<br />

accurate fashion. These two instruments have<br />

allowed us to increase our productivity, shorten<br />

our turnaround time, and improve the quality of<br />

our preparations without any increase in<br />

personnel. In the last calendar year, we<br />

processed 403 requests for histopathologic<br />

services that required 4,100 blocks and more<br />

than 31,600 glass slides. Using the Scanscope<br />

during December 2004, we scanned over 300<br />

cases in high resolution for inclusion in the<br />

VATR database. We anticipate generating digital<br />

Scanscope files for 3,000–4,000 cases in <strong>2005</strong><br />

that will be available on network servers. The<br />

MTMD laboratory is preparing gene expression<br />

data from cells and tissues and is correlating that<br />

with histology, tissue volume, and nuclear<br />

density to determine an effective and accurate<br />

38


screen for applications in molecular diagnostic<br />

assays. We have developed a QC and QA<br />

protocol for evaluating specimens for array<br />

analysis. During the past year we have prepared<br />

1,500 cDNA arrays for 20 collaborators from<br />

both in and outside the Institute.<br />

Our laboratories are primarily responsible for<br />

the archived clinical histopathology program<br />

called the Van Andel Tissue Repository (VATR).<br />

This program allows investigators to use existing<br />

human clinical samples to assess the expression<br />

of proteins. We also use these blocks to prepare a<br />

wide variety of tissue microarrays for research.<br />

We have increased the number of specimens in<br />

VATR to nearly 200,000 tissue samples. We are<br />

continuously entering data from the 200,000<br />

blocks and now have 54,473 reports that further<br />

explain and describe the archives. The reports are<br />

not directly linked to any personal identifiers or<br />

names and meet all HIPAA/CLIA regulations.<br />

During this year we have begun the process of<br />

imaging representative blocks from the cases and<br />

adding demographic data. The material from<br />

future years will have digital information on age,<br />

sex, and diagnosis and will be linked to image<br />

files. These samples will be used in cellular and<br />

molecular protocols approved by our Institutional<br />

Review Board. Since its inception, the VATR<br />

program has been used by 24 registered users<br />

who have submitted 534 requests for searches and<br />

101 subsequent tissue requests.<br />

In collaboration with Rick Hay, we have<br />

augmented the VATR program with freshly frozen<br />

tissues from the tissue acquisition program. This<br />

HIPAA-compliant program involves the active<br />

cooperation of patients, surgeons, and pathologists<br />

from area hospitals, and the surgical tissues<br />

collected are used primarily in cDNA and<br />

Affymetrix gene expression studies. This<br />

collection also provides the participating<br />

physicians with access to research collaborations,<br />

with the aim of facilitating the translation of<br />

research results into clinical practice. The goal of<br />

this project is to develop genetics-based<br />

diagnostic classification of human disease. There<br />

is a <strong>Scientific</strong> Advisory Board for this project<br />

comprising members of VARI and of the<br />

Spectrum Health pathology, surgical and medical<br />

oncology, and surgery departments.<br />

Original research within our labs focuses on<br />

quantification of images or arrays and the<br />

development of objective measurable data from<br />

images. A recent paper by Rozenblatt-Rosen et<br />

al. used a program written by Eric Kort for<br />

determining the co-localization of pixels that<br />

express unique fluorescent properties as well as<br />

the number of lumens or vessels in IHC-stained<br />

preparations. This is a direct extension of our<br />

Cytometry paper of 2003.<br />

We have recently obtained funding to<br />

advance our understanding of breast cancer in<br />

collaboration with Ilan Tsarfaty, George Vande<br />

Woude, and Craig Webb. We have begun<br />

molecular imaging of breast cancer and<br />

correlation of the findings with Affymetrix gene<br />

expression. Other collaborations within VARI<br />

involve Met and HGF/SF in cells and tissues; the<br />

location of gene-targeted proteins in rodents; and<br />

the evaluation of monoclonal antibodies as<br />

diagnostic reagents. We have been funded by the<br />

Michigan Technology Tri-Corridor to develop<br />

the commercialization of diagnostic gene<br />

expression (collaboration with Bin Teh), imaging<br />

of primary tumors (Rick Hay), and expression of<br />

mRNA in human sperm as an indicator of<br />

fertility or sterility (Stephen Krawetz). We are in<br />

the early stages of developing a program in<br />

neuropathology with a concentration on<br />

Parkinson’s disease, starting with the<br />

morphological and genomic characterization of<br />

human adult stem-cell populations of specific<br />

neurons.<br />

This year we have a student from Bath<br />

University in the U.K. The Bridges to the<br />

Baccalaureate program is a collaboration to<br />

support the recruitment of women and minorities<br />

into science careers; Dr. Resau is a coinvestigator<br />

and the VAI site coordinator for the<br />

program. In addition this year, we partnered with<br />

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

Program (GRAPCEP) to build a school within a<br />

school for science education and instruction in<br />

Creston High School of the Grand Rapids Public<br />

School system. Our GRAPCEP mentorship<br />

program continues to be funded by Pfizer for a<br />

fifth year. Seven high school students have<br />

trained in the laboratory and are now in<br />

baccalaureate programs.<br />

39


External Collaborators<br />

Eric Arnoys and John Ubels, Calvin College, Grand Rapids, Michigan<br />

Stephan Baldus, University of Cologne, Germany<br />

Lonson Barr, Marcos Dantus, and Matti Koeppel, Michigan State University, East Lansing<br />

Nadia Harbeck, Ludwig-Maximilians-Universität, Munich, Germany<br />

Christine Hughes and O. Orit Rosen, Harvard University, Cambridge, Massachusetts<br />

Sylvia Kachalsky, Linkagene, Lod, Israel<br />

Iafa Keydar, Tel Aviv University, Israel<br />

Stephen Krawetz, Wayne State University, Detroit, Michigan<br />

Ernst Lengyel, University of Chicago, Illinois<br />

Maria Roberts, National Cancer Institute, Frederick, Maryland<br />

Rulong Shen, Ohio State University, Columbus<br />

Ilan Tsarfaty, Tel Aviv University, Israel<br />

Recent Publications<br />

Kort, E.J., M.R. Moore, E.A. Hudson, B. Leeser, G.M. Yeruhalmi, R. Leibowitz-Amit, G. Tsarfaty, I.<br />

Tsarfaty, S. Moshkovitz, and J.H. Resau. In press. Use of organ explant and cell culture. In<br />

Mechanisms of Carcinogenesis, Hans Kaiser, ed. Dordrecht, The Netherlands: Kluwer Academic.<br />

Lengyel, Ernst, Dieter Prechtel, James H. Resau, Katja Gauger, Anita Welk, Kristina Lindemann,<br />

Georgia Salanti, Thomas Richter, Beatrice Knudsen, George F. Vande Woude, and Nadia Harbeck.<br />

<strong>2005</strong>. c-Met overexpression in node-positive breast cancer identifies patients with poor clinical<br />

outcome independent of Her2/neu. International Journal of Cancer 113(4): 678–682.<br />

Rozenblatt-Rosen, Orit, Christina M. Hughes, Suraj J. Nannepaga, Kalai Selvi Shanmugam, Terry D.<br />

Copeland, Tad Guszczynski, James H. Resau, and Matthew Meyerson. <strong>2005</strong>. The parafibromin<br />

tumor suppressor protein is part of a human Paf1 complex. Molecular and Cellular Biology 25(2):<br />

612–620.<br />

Seals, Darren F., Eduardo F. Azucena, Jr., Ian Pass, Lia Tesfay, Rebecca Gordon, Melissa Woodrow,<br />

James H. Resau, and Sara A. Courtneidge. <strong>2005</strong>. The adaptor protein Tks5/Fish is required for<br />

podosome formation and function, and for the protease-driven invasion of cancer cells. Cancer<br />

Cell 7(2): 155–165.<br />

Akervall, Jan, Xiang Guo, Chao-Nan Qian, Jacqueline Schoumans, Brandon Leeser, Eric Kort,<br />

Andrew Cole, James Resau, Carol Bradford, Thomas Carey, Johan Wennerberg, Harald Anderson,<br />

Jan Tennvall, and Bin T. Teh. 2004. Genetic and expression profiles of squamous cell carcinoma<br />

of the head and neck correlate with cisplatin sensitivity and resistance in cell lines and patients.<br />

Clinical Cancer Research 10(24): 8204–8213.<br />

Birchenall-Roberts, Maria C., Tao Fu, Ok-sun Bang, Michael Dambach, James H. Resau, Cari L.<br />

Sadowski, Daniel C. Bertolette, Ho-Jae Lee, Seong-Jin Kim, and Francis W. Ruscetti. 2004.<br />

Tuberous sclerosis complex 2 gene product interacts with human SMAD proteins. Journal of<br />

Biological Chemistry 279(24): 25605–25613.<br />

Holmen, Sheri L., Troy A. Giambernardi, Cassandra R. Zylstra, Bree D. Buckner-Berghuis, James H.<br />

Resau, J. Fred Hess, Vaida Glatt, Mary L. Bouxsein, Minrong Ai, Matthew L. Warman, and Bart<br />

O. Williams. 2004. Decreased BMD and limb deformities in mice carrying mutations in both<br />

Lrp5 and Lrp6. Journal of Bone and Mineral Research 19(12): 2033–2040.<br />

Tan, Min-Han, Carl Morrison, Pengfei Wang, Ximing Yang, Carola J. Haven, Chun Zhang, Ping Zhao,<br />

Maria S. Tretiakova, Eeva Korpi-Hyovalti, John R. Burgess, Khee Chee Soo, Wei-Keat Cheah,<br />

Brian Cao, James Resau, Hans Morreau, and Bin Tean Teh. 2004. Loss of parafibromin<br />

immunoreactivity is a distinguishing feature of parathyroid carcinoma. Clinical Cancer Research<br />

10(19): 6629–663<br />

40


From left to right: Resau, Goolsby, Haak, Berghuis, Norton, Percival, Hudson, Ferrell<br />

41


Laboratory of Germline Modification<br />

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

Dr. Swiatek received her M.S. (1984) and Ph.D. (1988) degrees in pathology from<br />

Indiana University. From 1988 to 1990, she was a postdoctoral fellow at the Tampa<br />

Bay Research Institute. From 1990 to 1994, she was a postdoctoral fellow at the<br />

Roche Institute of Molecular Biology in the laboratory of Tom Gridley. From 1994 to<br />

2000, Dr. Swiatek was a Research Scientist and Director of the Transgenic Core<br />

Facility at the Wadsworth Center in Albany, N.Y., and an Assistant Professor in the<br />

Department of Biomedical Sciences at the State University of New York at Albany.<br />

She joined VARI as a Special Program Investigator in August 2000. She has been<br />

the chair of the Institutional Animal Care and Use Committee since 2002 and is an<br />

Adjunct Assistant Professor in the College of Veterinary Medicine at Michigan State<br />

University. Dr. Swiatek received her M.B.A. in <strong>2005</strong> from Krannert School of<br />

Management at Purdue University.<br />

Staff<br />

Julie Koeman, B.S.<br />

Kellie Sisson, B.S.<br />

Juraj Zahatnansky, B.S.<br />

Laboratory Members<br />

IACUC Coordinator<br />

Kaye Johnson, B.S.<br />

Research Interests<br />

The germline modification lab is a fullservice<br />

lab that functions at the levels of<br />

service, research, and teaching to<br />

develop, analyze, and maintain mouse models of<br />

human disease. Our lab applies a business<br />

philosophy to core service offerings and we focus<br />

on scientific innovation, customer satisfaction,<br />

and service excellence. Mouse models are<br />

produced using gene-targeting technology, a wellestablished,<br />

powerful method for inserting<br />

specific genetic changes into the mouse genome.<br />

The resulting mice can be used to study the effects<br />

of these changes in the complex biological<br />

environment of a living organism. The genetic<br />

changes can include the introduction of a gene<br />

into a specific site in the genome, (gene “knockin”)<br />

or the inactivation of a gene already in the<br />

genome (gene “knock-out”). Since these<br />

mutations are introduced into the reproductive<br />

cells known as the germline, they can be used to<br />

study the developmental aspects of gene function<br />

associated with inherited genetic diseases.<br />

In addition to traditional gene-targeting<br />

technologies, the germline modification lab can<br />

produce mouse models in which the gene of<br />

interest is inactivated in a target organ or cell line<br />

instead of in the entire animal. These types of<br />

mouse models, known as conditional knock-outs,<br />

are particularly useful in studying genes that, if<br />

missing, cause the mouse to die as an embryo.<br />

The lab also has the capability to produce mutant<br />

embryos that have a wild-type placenta using<br />

tetraploid embryo technology. This technique is<br />

useful when the gene-targeted mutation prevents<br />

implantation of the mouse embryo in the uterus.<br />

We also assist in the development of embryonic<br />

stem (ES) or fibroblast cell lines from mutant<br />

embryos, which allows for in vitro studies of the<br />

gene mutation.<br />

Our gene-targeting service encompasses<br />

three major procedures: DNA electroporation,<br />

clone expansion and cryopreservation, and<br />

microinjection. Gene targeting procedures are<br />

initiated by mutating the genomic DNA of interest<br />

and inserting it into ES cells using the<br />

electroporation technique. The mutated gene<br />

integrates into the genome of the ES cells and, by<br />

a process called homologous recombination,<br />

replaces one of the two wild-type copies of the<br />

gene in the cells. Clones are identified, isolated,<br />

and cryopreserved, and genomic DNA is extracted<br />

from each clone and delivered to the client for<br />

analysis. Correctly targeted ES cell clones are<br />

thawed, established into tissue culture, and<br />

cryopreserved in liquid nitrogen. Gene-targeting<br />

mutations are introduced into the mouse by<br />

microinjection of the pluripotent ES cell clones<br />

into 3.5-day-old mouse embryos (blastocysts).<br />

These embryos, containing a mixture of wild-type<br />

42


and mutant ES cells, develop into mice called<br />

chimeras. The offspring of chimeras that inherit<br />

the mutated gene are heterozygotes, because they<br />

possess one copy of the mutated gene. The<br />

heterozygous mice are bred together to produce<br />

mice that completely lack the normal gene. These<br />

homozygous mice have two copies of the mutant<br />

gene and are called knock-out mice.<br />

Once gene targeting mice are produced, our<br />

lab assists in developing breeding schemes and<br />

provides for complete analysis of the mutants.<br />

The efficiency of mutant mouse production and<br />

analysis is enhanced by the AutoGenprep 960, a<br />

robotic, high-throughput DNA isolation machine.<br />

Tail biopsies from genetically engineered mice are<br />

processed in a 96-well format and the DNA<br />

samples are delivered to the client for analysis. It<br />

is our future plan that the DNA analysis be fully<br />

automated, with samples moving directly from the<br />

AutoGenprep 960 to a high-throughput<br />

genotyping platform, eliminating the need for<br />

clients to perform this labor-intensive analysis.<br />

The germline modification lab also directs the<br />

VARI cytogenetics core, which offers a variety of<br />

custom services. Mouse, rat, and human cell lines<br />

derived from tumors, fibroblasts, blood, or ES<br />

cells can be grown in tissue culture, growtharrested,<br />

fixed, and spread onto glass slides.<br />

Karyotyping of chromosomes using Leishman- or<br />

Giemsa-stained (G-banded) chromosomes is our<br />

basic service. However, spectral karyotyping<br />

(SKY) analysis of metaphase chromosome<br />

spreads, using high-quality, 24-color, wholechromosome<br />

fluorescent paints, can aid in the<br />

detection of subtle and complex chromosomal<br />

rearrangements. Fluorescence in situ<br />

hybridization (FISH) analysis, using indirectly or<br />

directly labeled bacterial artificial chromosome<br />

(BAC) or plasmid probes, can also be performed<br />

on metaphase spreads or on interphase nuclei<br />

derived from tissue touch preps or nondividing<br />

cells. Sequential staining of identical metaphase<br />

spreads using FISH and SKY can assist in<br />

identifying the chromosome integration site of a<br />

randomly integrated transgene.<br />

Finally the germline modification lab<br />

provides cryopreservation services for archiving<br />

and reconstituting valuable mouse strains. These<br />

cost-effective procedures decrease the need to<br />

continuously breed valuable mouse models, and<br />

they provide added insurance against the loss of<br />

custom mouse lines due to disease outbreak or a<br />

catastrophic event. Eight-cell mouse embryos or<br />

mouse sperm can be cryopreserved and stored in<br />

liquid nitrogen; they can be reconstituted by<br />

implantation into the oviducts of recipient mice or<br />

by in vitro fertilization of oocytes, respectively.<br />

The VARI germline modification lab directs<br />

the Michigan Animal Model Consortium<br />

(MAMC) of the Core Technology Alliance (CTA)<br />

Corp. MAMC is one of six collaborative core<br />

facilities located at the University of Michigan,<br />

Michigan State University, Wayne State<br />

University, Kalamazoo Community College, and<br />

VARI, offering research services in proteomics,<br />

structural biology, genomics, bioinformatics,<br />

high-throughput compound screening, and animal<br />

modeling. These labs receive funding from the<br />

Michigan Economic Development Corporation to<br />

efficiently provide mouse modeling services to<br />

researchers studying human diseases and to<br />

promote the commercialization of the core<br />

services in order to stimulate the development of<br />

biomedical research in Michigan.<br />

The MAMC services are classified into three<br />

major categories: mouse model development;<br />

analysis; and maintenance and preservation.<br />

Model development services consist of gene<br />

targeting, transgenic, TVA transgenic, and<br />

xenotransplantation procedures. Analytical<br />

procedures are performed on the animal models to<br />

determine the nature and extent of any phenotypic<br />

consequences in the models and their correlation<br />

with human disease. Mouse analysis consists of<br />

histology, necropsy, veterinary pathology,<br />

cytogenetics, blood chemistry, blood hematology,<br />

and imaging. The DNA isolation service supports<br />

the genotyping analysis of the models, and the<br />

monoclonal antibody core supports the molecular<br />

analysis of the mutant mice and xenotransplantation<br />

models. Finally, the maintenance<br />

and preservation services include a mouse<br />

repository, mouse breeding services, mouse<br />

rederivation, and embryo/sperm cryopreservation.<br />

These services are described more completely on<br />

the MAMC website at .<br />

43


Recent Publications<br />

Robertson, Scott A., Jacqueline Schoumans, Brendan D. Looyenga, Jason A. Yuhas, Cassandra R.<br />

Zylstra, Julie M. Koeman, Pamela J. Swiatek, Bin T. Teh, and Bart O. Williams. <strong>2005</strong>. Spectral<br />

karyotyping of sarcomas and fibroblasts derived from Ink4a/Arf-deficient mice reveals chromosomal<br />

instability in vitro. International Journal of Oncology 26(3): 629–634.<br />

Wu, Lin, Jun Gu, Huadong Cui, Qing-Yu Zhang, Melissa Behr, Cheng Fang, Yan Weng, Kerri<br />

Kluetzman, Pamela J. Swiatek, Weizhu Yang, Laurence Kaminsky, and Xinxin Ding. <strong>2005</strong>.<br />

Transgenic mice with a hypomorphic NADPH-cytochrome P450 reductase gene: effects on<br />

development, reproduction, and microsomal cytochrome P450. Journal of Pharmacology and<br />

Experimental Therapeutics 312(1): 35–43.<br />

Graveel, Carrie, Yanli Su, Julie Koeman, Ling-Mei Wang, Lino Tessarollo, Michelle Fiscella, Carmen<br />

Birchmeier, Pamela Swiatek, Roderick Bronson, and George Vande Woude. 2004. Activating Met<br />

mutations produce unique tumor profiles in mice with selective duplication of the mutant allele.<br />

Proceedings of the National Academy of Sciences U.S.A. 101(49): 17198–17203.<br />

From left to right: Swiatck, Koeman, Zahatnansky, Sisson<br />

44


Laboratory of Cancer Genetics<br />

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

Dr. Teh obtained his M.D. from the University of Queensland, Australia, in 1992, and<br />

his Ph.D. from the Karolinska Institute, Sweden, in 1997. Before joining the Van<br />

Andel Research Institute (VARI), he was an Associate Professor of medical<br />

genetics at the Karolinska Institute. Dr. Teh joined VARI as a Senior <strong>Scientific</strong><br />

Investigator in January 2000. Dr. Teh’s research mainly focuses on kidney cancer,<br />

and he is currently on the Medical Advisory Board of the Kidney Cancer Association.<br />

He became VARI’s Deputy Director for Research Operations in the fall of 2003 and<br />

was promoted to Distinguished <strong>Scientific</strong> Investigator in <strong>2005</strong>.<br />

Staff<br />

Miles Chao-Nan Qian, M.D., Ph.D.<br />

Pengfei Wang, M.D., Ph.D.<br />

Xin Yao, M.D., Ph.D.<br />

Jindong Chen, Ph.D.<br />

Kunihiko Futami, Ph.D.<br />

Laboratory Members<br />

Sok Kean Khoo, Ph.D.<br />

Douglas Luccio-Camelo, Ph.D.<br />

David Petillo, Ph.D.<br />

Eric Kort, M.S.<br />

Stephanie Potter, M.S.<br />

Jeff Bates, B.S.<br />

Timothy Yaw Bediako, B.S.<br />

Mark Betten, B.S.<br />

Aaron Massie, B.S.<br />

Research Interests<br />

Kidney cancer, or renal cell carcinoma<br />

(RCC), is the tenth most common<br />

cancer in the United States (34,000 new<br />

cases and more than 12,000 deaths a year). Its<br />

incidence has been increasing, a phenomenon<br />

that cannot be accounted for by the wider use of<br />

imaging procedures. We have established a<br />

comprehensive and integrated kidney research<br />

program, and our major research goals are 1) to<br />

identify the molecular signatures of different<br />

subtypes of kidney tumors, both hereditary and<br />

sporadic, and to understand how these genes<br />

function and interact in giving rise to the tumors<br />

and their progression; 2) to identify and develop<br />

novel biomarkers and key drug targets; and 3) to<br />

generate animal models for drug testing and<br />

preclinical bioimaging.<br />

Our program to date has established a<br />

worldwide network of collaborators, a tissue bank<br />

containing fresh-frozen tumor pairs (over 700<br />

cases) and serum, and a gene expression profiling<br />

database of 500 tumors with long-term clinical<br />

follow-up information for half of them. Our<br />

program includes positional cloning of hereditary<br />

RCC syndromes and functional studies of their<br />

related genes, microarray and bioinformatic<br />

analysis, and generation of RCC mouse models.<br />

Hereditary RCC syndromes – positional<br />

cloning and functional studies<br />

Following the identification of the HRPT2,<br />

BHD, and two familial RCC breakpoint genes<br />

(NORE1 and LSAMP) by us and others, we are<br />

now focusing on functional studies of these genes,<br />

including generating mouse models that harbor<br />

mutations of these genes (see below). We<br />

established the role of HRPT2 in parathyroid<br />

carcinoma by mutation analysis and<br />

immunohistochemical staining. We have shown<br />

that its protein, parafibromin, accumulates<br />

predominantly in the nucleus, and this nuclear<br />

localization is essential to maintaining its antiproliferative<br />

function.<br />

Microarray gene expression profiling<br />

and bioinformatics<br />

Based on microarray profiling of 400 kidney<br />

tumors using both our own spotted arrays and<br />

Affymetrix microarrays, we have identified the<br />

molecular signatures for 1) different subtypes of<br />

kidney tumors; 2) the prognosis for clear cell RCC<br />

and papillary RCC; and 3) the prediction of drug<br />

response (immunotherapy). Our studies have been<br />

validated by RT-PCR and immunohistochemical<br />

staining. We have also been working closely with<br />

VARI’s Kyle Furge in analyzing our microarray<br />

data. Using a program developed by his team,<br />

comparative genomic microarray analysis<br />

(CGMA), we correlated gene expression profiles<br />

with the predicted chromosomal imbalances for<br />

different RCC histological subtypes, which may<br />

facilitate our efforts in identifying RCC-related<br />

genes from these chromosomal regions. More<br />

information and details can be found in several of<br />

our review articles and book chapters.<br />

45


Mouse models of kidney cancer<br />

In collaboration with VARI’s Bart Williams<br />

and Pam Swiatek, we have set up several mouse<br />

models for kidney tumors. These include<br />

conventional and conditional knock-outs for<br />

BHD, HRPT2, VHL, PTEN, and APC. For the<br />

latter three cases, we collaborated with Peter<br />

Igarashi of the University of Texas Southwestern<br />

Medical Center and obtained from him<br />

Ksp1.3/Cre transgenic mice expressing Cre<br />

recombinase exclusively in the kidney and<br />

developing GU tract, which can mediate<br />

epithelial-specific Cre/lox recombination in these<br />

tissues. We then crossed these mice with the<br />

APC-, VHL-, and PTEN-floxed mice. To date, we<br />

have completed the studies on APC conditional<br />

knock-outs, which give the phenotype of<br />

polycystic kidney disease and renal adenoma.<br />

We are currently characterizing the phenotypes of<br />

the other mice. In addition, we have established<br />

xenograft subcapsular models using five different<br />

RCC cell lines. The natural course of these<br />

models has been documented and studied by<br />

microarrary gene expression profiling.<br />

External Collaborators<br />

We have extensive collaborations with researchers and clinicians in the United States and overseas.<br />

Recent Publications<br />

Morris, M.R., D. Gentle, M. Abdulrahman, E.N. Maina, K. Gupta, R.E. Banks, M.S. Wiesener, T.<br />

Kishida, M. Yao, B. Teh, F. Latif, and E.R. Maher. In press. Tumor suppressor activity and<br />

epigenetic inactivation of hepatocyte growth factor activator inhibitor type 2 (HAI-2/SPINT2) in<br />

papillary and clear cell renal cell carcinoma. Cancer Research.<br />

Takahashi, M., X.J. Yang, S. McWhinney, N. Sano, C.H. Eng, S. Kagawa, B.T. Teh, and H. Kanayama.<br />

In press. cDNA microarray analysis assists in diagnosis of malignant intrarenal pheochromocytoma<br />

originally masquerading as a renal cell carcinoma. Journal of Medical Genetics.<br />

Tretiakova, M., M. Turkyilmaz, T. Grushko, M. Kocherginsky, C. Rubin, O. Olopade, B.T. Teh, and<br />

X.J. Yang. In press. Topoisomerase II[α] expression in Wilms tumors. Clinical Cancer Research.<br />

Wang, P., M.-H. Tan, C. Zhang, H. Morreau, and B.T. Teh. In press. HRPT2, a tumor suppressor gene<br />

for hyperparathyroidism-jaw tumor syndrome. Hormone and Metabolic Research.<br />

Yang, X.J., M.-H. Tan, H.L. Kim, J.A. Ditlev, M.W. Betten, C.E. Png, E.J. Kort, K. Futami, K.J.<br />

Dykema, K.A. Furge, M. Takahashi, H. Kanayama, P.H. Tan, B.S. Teh, C. Luan, et al. In press.<br />

A molecular classification of papillary renal cell carcinoma. Cancer Research.<br />

Cardinal, J., L. Bergman, N. Hayward, A. Sweet, J. Warner, L. Marks, D. Learoyd, T. Dwight, B.<br />

Robinson, M. Epstein, M. Smith, B.T. Teh, D. Cameron, and J. Prins. <strong>2005</strong>. A report of a national<br />

mutation testing service for the MEN1 gene: clinical presentations and implications for mutation<br />

testing. Journal of Medical Genetics 42(1): 69–74.<br />

Chuang, S.T., P. Chu, J. Sugimura, M.S. Tretiakova, V. Papavero, K. Wang, M.-H. Tan, F. Lin, B.T.<br />

Teh, and X.J. Yang. <strong>2005</strong>. Overexpression of glutathione S-transferase α in clear cell renal cell<br />

carcinoma. American Journal of Clinical Pathology 123(3): 421–429.<br />

Lee, Youn-Soo, Alexander O. Vortmeyer, Irina A. Lubensky, Timothy W.A. Vogel, Barbara Ikejiri,<br />

Sophie Ferlicot, Gérard Benoît, Sophie Giraud, Edward H. Oldfield, W. Marston Linehan, Bin T.<br />

Teh, Stéphane Richard, and Zhengping Zhuang. <strong>2005</strong>. Coexpression of erythropoietin and<br />

erythropoietin receptor in Von Hippel-Lindau disease–associated renal cysts and renal cell<br />

carcinoma. Clinical Cancer Research 11(3): 1059–1064.<br />

Qian, Chao-Nan, Jared Knol, Peter Igarashi, Fangmin Lin, Uko Zylstra, Bin Tean Teh, and Bart O.<br />

Williams. <strong>2005</strong>. Cystic renal neoplasia following conditional inactivation of Apc in mouse renal<br />

tubular epithelium. Journal of Biological Chemistry 280(5): 3938–3945.<br />

Robertson, Scott A., Jacqueline Schoumans, Brendan D. Looyenga, Jason A. Yuhas, Cassandra R.<br />

Zylstra, Julie M. Koeman, Pamela J. Swiatek, Bin T. Teh, and Bart O. Williams. <strong>2005</strong>. Spectral<br />

46


karyotyping of sarcomas and fibroblasts derived from Ink4a/Arf-deficient mice reveals<br />

chromosomal instability in vitro. International Journal of Oncology 26(3): 629–634.<br />

Rogers, C., M.-H. Tan, and B.T. Teh. <strong>2005</strong>. Gene expression profiling of renal cell carcinoma and<br />

clinical implications. Urology 65(2): 231–237.<br />

Schoumans, Jacqueline, Ann Nordgren, Claudia Ruivenkamp, Karen Brøndum-Nielsen, Bin Tean Teh,<br />

Göran Annéren, Eva Holmberg, Magnus Nordenskjöld, and Britt-Marie Anderlid. <strong>2005</strong>.<br />

Genome-wide screening using array-CGH does not reveal microdeletions/microduplications in<br />

children with Kabuki syndrome. European Journal of Human Genetics 13(2): 260–263.<br />

Takahashi, Masayuki, Veronica Papavero, Jason Yuhas, Eric Kort, Hiro-omi Kanayama, Susumu Kagawa,<br />

R.C. Baxter, Ximing J. Yang, Steven G. Gray, and Bin T. Teh. <strong>2005</strong>. Altered expression of members<br />

of the IGF-axis in clear cell renal cell carcinoma. International Journal of Oncology 26(4): 923–931.<br />

Akervall, Jan, Xiang Guo, Chao-Nan Qian, Jacqueline Schoumans, Brandon Leeser, Eric Kort,<br />

Andrew Cole, James Resau, Carol Bradford, Thomas Carey, Johan Wennerberg, Harald Anderson,<br />

Jan Tennvall, and Bin T. Teh. 2004. Genetic and expression profiles of squamous cell carcinoma<br />

of the head and neck correlate with cisplatin sensitivity and resistance in cell lines and patients.<br />

Clinical Cancer Research 10(24): 8204–8213.<br />

Atkins, Michael B., David E. Avigan, Ronald M. Bukowski, Richard W. Childs, Janice P. Dutcher, Tim<br />

G. Eisen, Robert A. Figlin, James H. Finke, Robert C. Flanigan, Daniel J. George, S. Nahum<br />

Goldberg, Michael S. Gordon, Othon Iliopoulos, William G. Kaelin, Jr., W. Marston Linehan, et<br />

al. 2004. Innovations and challenges in renal cancer: consensus statement. Clinical Cancer<br />

Research 10(18, Pt.2): 6277S–6281S.<br />

Calender, A., C. Morrison, P. Komminoth, J.Y. Scaoazec, K. Sweet, and B.T. Teh. 2004. Multiple<br />

endocrine neoplasia type 1. In Pathology and Genetics of Tumours of the Endocrine Organs,<br />

DeLellis, Heitz, Lloyd, and Eng, eds. WHO Classification of Tumours series, Vol. 8. Lyon,<br />

France: IARC Press, pp. 218–227.<br />

Furge, Kyle A., Kerry A. Lucas, Masayuki Takahashi, Jun Sugimura, Eric J. Kort, Hiro-omi<br />

Kanayama, Susumu Kagawa, Philip Hoekstra, John Curry, Ximing J. Yang, and Bin T. Teh. 2004.<br />

Robust classification of renal cell carcinoma based on gene expression data and predicted<br />

cytogenetic profiles. Cancer Research 64(12): 4117–4121.<br />

Haven, Carola J., Viive M. Howell, Paul H.C. Eilers, Robert Dunne, Masayuki Takahashi, Marjo van<br />

Puijenbroek, Kyle Furge, Job Kievit, Min-Han Tan, Gert Jan Fleuren, Bruce G. Robinson, Leigh<br />

W. Delbridge, Jeanette Philips, Anne E. Nelson, Ulf Krause, et al. 2004. Gene expression of<br />

parathyroid tumors: molecular subclassification and identification of the potential malignant<br />

phenotype. Cancer Research 64(20): 7405–7411.<br />

Lindvall, Charlotta, Kyle Furge, Magnus Björkholm, Xiang Guo, Brian Haab, Elisabeth Blennow,<br />

Magnus Nordenskjöld, and Bin Tean Teh. 2004. Combined genetic and transcriptional profiling of<br />

acute myeloid leukemia with normal and complex karyotypes. Haematologica 89(9): 1072–1081.<br />

Luccio-Camelo, Douglas C., Karina N. Une, Rafael E.S. Ferreira, Sok Kean Khoo, Radoslav Nickolov,<br />

Marcello D. Bronstein, Mario Vaisman, Bin Tean Teh, Lawrence A. Frohman, Berenice B.<br />

Mendonca, and Monica R. Gadelha. 2004. A meiotic recombination in a new isolated familial<br />

somatotropinoma kindred. European Journal of Endocrinology 150(5): 643–648.<br />

Marsh, Deborah J., Hans Morreau, and Bin T. Teh. 2004. HRPT2 and parathyroid cancer. Lancet<br />

Oncology 5(2): 78.<br />

Morrison, C., K. Sweet, and B.T. Teh. 2004. Hyperthyroidism-jaw tumor syndrome. In Pathology<br />

and Genetics of Tumours of the Endocrine Organs, DeLellis, Heitz, Lloyd, and Eng, eds. WHO<br />

Classification of Tumours series, Vol. 8. Lyon, France: IARC Press, pp. 228–237.<br />

Patton, Kurt T., Maria S. Tretiakova, Jorge L. Yao, Veronica Papavero, Lei Huo, Brian P. Adley, Guan<br />

Wu, Jiaoti Huang, Michael R. Pins, Bin T. Teh, and Ximing J. Yang. 2004. Expression of RON<br />

proto-oncogene in renal oncocytoma and chromophobe renal cell carcinoma. American Journal<br />

of Surgical Pathology 28(8): 1045–1050.<br />

47


Stephens, P., C. Hunter, G. Bignell, S. Edkins, H. Davies, J. Teague, C. Stevens, S. O’Meara, R. Smith,<br />

A. Parker, A. Barthorpe, M. Blow, L. Brackenbury, A. Butler, O. Clarke, et al. 2004. Lung cancer:<br />

intragenic ERBB2 kinase mutations in tumours. Nature 431(7008): 525–526.<br />

Sugimura, Jun, Richard S. Foster, Oscar W. Cummings, Eric J. Kort, Masayuki Takahashi, Todd T.<br />

Lavery, Kyle A. Furge, Lawrence H. Einhorn, and Bin Tean Teh. 2004. Gene expression profiling<br />

of early- and late-relapse nonseminomatous germ cell tumor and primitive neuroectodermal tumor<br />

of the testis. Clinical Cancer Research 10(7): 2368–2378.<br />

Sugimura, Jun, Ximing J. Yang, Maria S. Tretiakova, Masayuki Takahashi, Eric J. Kort, Barbara<br />

Fulton, Tomoaki Fujioka, Nicholas J. Vogelzang, and Bin Tean Teh. 2004. Gene expression<br />

profiling of mesoblastic nephroma and Wilms tumors—comparison and clinical implications.<br />

Urology 64(2): 362–368.<br />

Tan, Min-Han, Carl Morrison, Pengfei Wang, Ximing Yang, Carola J. Haven, Chun Zhang, Ping Zhao,<br />

Maria S. Tretiakova, Eeva Korpi-Hyovalti, John R. Burgess, Khee Chee Soo, Wei-Keat Cheah,<br />

Brian Cao, James Resau, Hans Morreau, and Bin Tean Teh. 2004. Loss of parafibromin<br />

immunoreactivity is a distinguishing feature of parathyroid carcinoma. Clinical Cancer Research<br />

10(19): 6629–6637.<br />

Tan, Min-Han, Craig G. Rogers, Jeffrey T. Cooper, Jonathon A. Ditlev, Thomas J. Maatman, Ximing<br />

Yang, Kyle A. Furge, and Bin Tean Teh. 2004. Gene expression profiling of renal cell carcinoma.<br />

Clinical Cancer Research 10(18): 6315S–6321S.<br />

Tan, M.-H., and B.T. Teh. 2004. Renal neoplasia in the hyperparathyroidism-jaw tumor syndrome.<br />

Current Molecular Medicine 4(8): 895–897.<br />

Teh, B.T. 2004. Gene expression profiling begins to fulfill promise in differential diagnosis and<br />

prognosis of renal cell carcinomas. Kidney Cancer Journal 2(3): 15–18.<br />

Standing, left to right: Qian, Wang, Massie, Petillo, Bates, Bediako, Chen<br />

seated, left to right: Betten, Khoo, Antio, Potter, Teh, Futami<br />

48


Laboratory of Molecular Oncology<br />

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

Dr. Vande Woude received his M.S. (1962) and Ph.D. (1964) from Rutgers<br />

University. From 1964–1972, he served first as a postdoctoral research associate,<br />

then as a research virologist for the U.S. Department of Agriculture at Plum Island<br />

Animal Disease Center. In 1972, he joined the National Cancer Institute as Head<br />

of the Human Tumor Studies and Virus Tumor Biochemistry sections and, in 1980,<br />

was appointed Chief of the Laboratory of Molecular Oncology. In 1983, he became<br />

Director of the Advanced Bioscience Laboratories–Basic Research Program at the<br />

National Cancer Institute’s Frederick Cancer Research and Development Center, a<br />

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 Institute.<br />

In 1999, he was recruited to the Directorship of the Van Andel Research Institute in Grand Rapids, Michigan.<br />

Staff<br />

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

Rick Hay, Ph.D., M.D.<br />

Yu-Wen Zhang, M.D., Ph.D.<br />

Chongfeng Gao, Ph.D.<br />

Carrie Graveel, Ph.D.<br />

Laboratory Members<br />

Sharon Moshkovitz, Ph.D.<br />

Qian Xie, Ph.D.<br />

Dafna Kaufman, M.Sc.<br />

Lia Tesfay, M.S.<br />

Matt VanBrocklin, M.S.<br />

Benjamin Staal, B.S.<br />

Yanli Su, A.M.A.T.<br />

Visiting Scientists<br />

Galia Tsarfaty, M.D.<br />

Ilan Tsarfaty, Ph.D.<br />

Student<br />

Jack DeGroot<br />

Research Interests<br />

Activating mutations in Met are found in<br />

human kidney and gastric cancers,<br />

providing compelling genetic evidence<br />

that Met is an important oncogene<br />

(http://www.vai.org/vari/metandcancer/). To study<br />

how activating mutations are involved in tumor<br />

development, we generated mice bearing Met<br />

with mutations representing both inherited and<br />

sporadic mutations found in human cancers. The<br />

different mutant Met lines developed unique<br />

tumor profiles including carcinomas, sarcomas,<br />

and lymphomas. Cytogenetic analysis of the<br />

tumors in Met mutant mice shows that in all cases<br />

amplification of the mutant met allele is observed.<br />

Selective chromosomal amplification is also<br />

found in patients with renal cancer, indicating<br />

that amplification of the mutant met allele may be<br />

required for tumor progression.<br />

The differences in tumor types and latency,<br />

depending on the mutation, may be due to<br />

signaling differences triggered by the specific<br />

mutation. Studies are underway to identify the<br />

signaling pathways selectively activated by these<br />

mutations. Understanding the signaling specificity<br />

of these mutations is essential to identifying and<br />

developing successful therapeutics. Our mutant<br />

mice provide a valuable model for testing Met<br />

inhibitors and for understanding the molecular<br />

events critical for Met-mediated tumorigenesis.<br />

Proliferation and invasion<br />

We are interested in how HGF/SF-induced<br />

proliferation and invasion contribute to tumor<br />

progression. We have established in vitro methods<br />

to select highly proliferative or invasive cell<br />

populations that may mimic the in vivo process of<br />

clonal selection during tumor progression. Our<br />

studies show that most tumor cells display both<br />

invasive and proliferative phenotypes and that they<br />

can reversibly change from invasive to proliferative<br />

phenotypes. We are currently studying the genetic<br />

and epigenetic factors that determine the different<br />

cellular responses.<br />

HGF/SF-Met-mediated tumor angiogenesis<br />

HGF/SF acts as an angiogenic switch by<br />

simultaneously up-regulating vascular endothelial<br />

growth factor (VEGF) and down-regulating<br />

thrombospondin 1 (TSP-1) expression in the same<br />

tumor cells, mediated through the MAPK<br />

pathway. In addition, HGF/SF also downregulates<br />

TSP-1 expression in normal human<br />

umbilical vascular endothelial cells, in which<br />

VEGF expression is undetectable. TSP-1 and<br />

inhibitors of VEGF (such as Avastin) have been<br />

shown to inhibit tumor angiogenesis and growth.<br />

Our data raise the question of whether TSP-1 in<br />

combination with VEGF inhibitors would<br />

enhance inhibition of HGF/SF-Met-mediated<br />

49


tumor angiogenesis and growth relative to each<br />

tested separately, and whether these inhibitors are<br />

better than those specifically directed against<br />

HGF/SF or Met receptor. We are testing these<br />

approaches.<br />

Immunocompromised transgenic<br />

mice with Met-expressing xenografts<br />

We have generated a severe combined<br />

immune deficiency (SCID) mouse strain carrying<br />

a human HGF/SF transgene. This mouse<br />

provides a species-compatible ligand for<br />

propagating human tumor cells expressing human<br />

Met receptors. The growth of Met-expressing<br />

human tumor xenografts can be significantly<br />

enhanced in this transgenic mouse relative to<br />

those in nontransgenic hosts. Our data strongly<br />

suggest that this immunocompromised strain will<br />

be a useful tool for investigating the role of Met<br />

in tumor malignancy. Currently, we are testing<br />

experimental metastases and orthotopic<br />

xenografts of human tumor cells. This model will<br />

also be used for preclinical testing of drugs or<br />

compounds targeting the HGF/SF-Met complex<br />

and downstream signaling pathways.<br />

Geldanamycin inhibits tumor cell<br />

invasion at femtomolar concentrations<br />

Our lab has been studying the mechanism of<br />

geldanamycin (GA) inhibition of urokinase<br />

activation of plasmin from plasminogen (uPA).<br />

Previously, we have shown that a subset of GA<br />

derivatives at femtomolar concentrations (fM-<br />

GAi) inhibit HGF/SF-induced activation of<br />

plasmin in canine MDCK cells. We have<br />

recently found that such inhibition also occurs in<br />

several human glioblastoma cell lines (DBTRG,<br />

U373, and SNB19) and in SK-LMS-1 human<br />

leiomyosarcoma cells. Curiously, fM-GAi drugs<br />

only inhibit HGF/SF-induced uPA activity, and<br />

only when the magnitude of HGF/SF-uPA<br />

induction is above 1.5 times basal uPA activity.<br />

These fM-GAi derivatives also block MDCK cell<br />

scattering and glioblastoma tumor cell invasion<br />

in vitro at concentrations well below those<br />

required to exhibit a measurable effect on Met<br />

expression. Other experiments using radicicol<br />

and macbecin II provide circumstantial evidence<br />

for a novel non-HSP90 molecular target that is<br />

involved in HGF/SF-mediated tumor cell<br />

invasion.<br />

Imaging of Met oncogene activation<br />

We have generated mice carrying a murine<br />

GFP-Met transgene that emits intense<br />

fluorescence to reveal where in the animal Met is<br />

expressed. Five founder mice were selected that<br />

had GFP-Met expression levels from high to<br />

moderate. All of the transgenic GFP-Met male<br />

mice—but no females—develop tumors in the<br />

preputial sebaceous glands. Moreover, mice<br />

expressing the highest levels of the GFP-Met<br />

transgene develop tumors earlier. However,<br />

tumors originating from the different founder<br />

lines had similar pathological phenotypes; the<br />

mice presented with cystic sebaceous tumors<br />

having a rapid growth rate. GFP-Met expression<br />

is always higher in the sebaceous gland tumors<br />

relative to normal skin, with tumors covering the<br />

spectrum of adenomas, adenocarcinomas, and<br />

angiosarcomas. Metastases developed in 71% of<br />

GFP-Met transgenic mice with adenocarcinoma<br />

and in 18% of mice with angiosarcoma. The<br />

metastases were found locally in the skin and in<br />

distant organs such as liver, lung, and kidney.<br />

Image analyses of unfixed frozen tumor<br />

metastases showed large numbers of cells<br />

overexpressing GFP-Met, and tissue arrays of<br />

these tumors revealed higher GFP-Met levels<br />

relative to the primary tumors.<br />

MAPK in melanoma<br />

Constitutive activation of MAPK signaling<br />

contributes to many human cancers, including<br />

melanoma, with activating mutations in Nras or<br />

BRAF found in 80% of the tumors. While most<br />

cancer cells exhibit a cytostatic response to<br />

disruption of MAPK signaling, melanomas show<br />

a cytotoxic response. The Koo laboratory<br />

showed that inhibition of MAPK signaling<br />

efficiently triggers cell death by apoptosis in<br />

human melanoma cells. Normal epidermal<br />

melanocytes, however, do not undergo apoptosis<br />

in response to MAPK inhibition, but arrest in the<br />

G1 phase of the cell cycle. Moreover, in vivo<br />

interference of MAPK signaling produces either<br />

significant or complete regression of human<br />

melanoma xenograft tumors in athymic nude<br />

mice. These results indicate that the MAPK<br />

pathway represents tumor-specific survival<br />

signaling in melanoma, and that inhibition of this<br />

pathway may be a therapeutic strategy.<br />

50


Nuclear oncology<br />

During the past year our laboratory has made<br />

progress in the area of nuclear oncology on two<br />

fronts: the continued development of full-length<br />

anti-Met monoclonal antibodies (mAbs) for<br />

radioimmunodiagnostic and therapeutic applications<br />

and the new development of genetically<br />

engineered Met-directed antibody fragments.<br />

We call our biological agents that recognize<br />

human Met “MetSeek” and we continue to<br />

evaluate two full-length mAbs, Met3 and Met5,<br />

in animal models of cancer. Met3 recognizes an<br />

epitope on the extracellular domain of human<br />

Met, but not on canine Met, whereas Met5<br />

recognizes an epitope common to human and<br />

canine Met. Experiments on the usefulness of<br />

radiolabeled Met3 for imaging human<br />

glioblastoma multiforme tumor xenografts<br />

(because of their exquisite sensitivity to<br />

geldanamycins) are in progress. We have a new<br />

collaboration with David Schteingart to study<br />

Met expression by human adrenocortical<br />

carcinomas and to evaluate MetSeek mAbs as<br />

diagnostic tools in this setting. Preliminary<br />

experiments (performed in collaboration with<br />

David Wenkert and Milton Gross) have shown<br />

that radiometal chelation (e.g., with Tc-99m<br />

pertechnetate) may be used instead of direct<br />

radioiodination for labeling MetSeek mAbs. In<br />

collaboration with David Waters, we have<br />

conducted a short-term analysis of nonradioactive<br />

Met5 in normal dogs. No clinical,<br />

hematological, or chemical evidence of toxicity<br />

has been found, and histopathological analysis of<br />

tissues harvested at necropsy is pending.<br />

We have launched a collaborative project to<br />

develop and evaluate genetically engineered<br />

Met-directed antibody fragments as alternatives<br />

to full-length MetSeek mAbs for diagnostic and<br />

therapeutic applications. Single-chain versions<br />

of Met3 and Met5 (scFv) are under construction<br />

at ApoLife, Inc., a biotechnology firm in Detroit,<br />

and we have shown that a new Met-directed<br />

human-Fab-expressing phage display product<br />

can be used to image autocrine human<br />

leiomyosarcoma (SK-LMS-1/HGF) tumor<br />

xenografts in nude mice.<br />

External Collaborators<br />

Milton Gross, Department of Veterans Affairs Healthcare System, Ann Arbor, Michigan<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 />

Alnawaz Rehemtulla, Brian Ross, and David Schteingart, University of Michigan, Ann Arbor<br />

Yuehai Shen and David Wenkert, Michigan State University, East Lansing<br />

Olga Volpert, Northwestern University, Evanston, Illinois<br />

David Waters, Gerald P. Murphy Cancer Foundation, West Lafayette, Indiana<br />

Robert Wondergem, East Tennessee State University, Johnson City<br />

Recent Publications<br />

Jiao, Y., P. Zhao, J. Zhu, T. Grabinski, Z. Feng, X. Guan, R.S. Skinner, M.D. Gross, Y. Su, G.F. Vande<br />

Woude, R.V. Hay, and B. Cao. In press. Construction of human naïve Fab library and<br />

characterization of anti-Met Fab fragment generated from the library. Molecular Biotechnology.<br />

Gao, Chong Feng, and George F. Vande Woude. <strong>2005</strong>. HGF/SF signaling in tumor progression. Cell<br />

Research 15(1): 49–51.<br />

Islam, Azharul, Yoko Sakamoto, Kazuhisa Kosaka, Satoshi Yoshitome, Isamu Sugimoto, Kazuo<br />

Yamada, Ellen Shibuya, George F. Vande Woude, and Eikichi Hashimoto. <strong>2005</strong>. The distinct<br />

stage-specific effects of 2-(p-amylcinnamoyl)amino-4-chlorobenzoic acid on the activation of<br />

MAP kinase and Cdc2 kinase in Xenopus oocyte maturation. Cellular Signalling 17(4): 507–523.<br />

Lengyel, Ernst, Dieter Prechtel, James H. Resau, Katja Gauger, Anita Welk, Kristina Lindemann,<br />

Georgia Salanti, Thomas Richter, Beatrice Knudsen, George F. Vande Woude, and Nadia Harbeck.<br />

51


<strong>2005</strong>. c-Met overexpression in node-positive breast cancer identifies patients with poor clinical<br />

outcome independent of Her2/neu. International Journal of Cancer 113(4): 678–682.<br />

Zhang, Yu-Wen, Yanli Su, Nathan Lanning, Margaret Gustafson, Nariyoshi Shinomiya, Ping Zhao,<br />

Brian Cao, Galia Tsarfaty, Ling-Mei Wang, Rick Hay, and George F. Vande Woude. <strong>2005</strong>.<br />

Enhanced growth of human Met-expressing xenografts in a new strain of immunocompromised<br />

mice transgenic for human hepatocyte growth factor/scatter factor. Oncogene 24(1): 101–106.<br />

Graveel, Carrie, Yanli Su, Julie Koeman, Ling-Mei Wang, Lino Tessarollo, Michelle Fiscella, Carmen<br />

Birchmeier, Pamela Swiatek, Roderick Bronson, and George Vande Woude. 2004. Activating Met<br />

mutations produce unique tumor profiles in mice with selective duplication of the mutant allele.<br />

Proceedings of the National Academy of Sciences U.S.A. 101(49): 17198–17203.<br />

Kelloff, Gary J., Robert C. Bast, Jr., Donald S. Coffey, Anthony V. D’Amico, Robert S. Kerbel, John<br />

W. Park, Raymond W. Ruddon, Gordon J.S. Rustin, Richard L. Schilsky, Caroline C. Sigman, and<br />

George F. Vande Woude. 2004. Biomarkers, surrogate end points, and the acceleration of drug<br />

development for cancer prevention and treatment: an update. Clinical Cancer Research 10(11):<br />

3881–3884.<br />

Lee, Chong-Chou, Andrew J. Putnam, Cindy K. Miranti, Margaret Gustafson, Ling-Mei Wang,<br />

George F. Vande Woude, and Chong-Feng Gao. 2004. Overexpression of sprouty-2 inhibits<br />

HGF/SF-mediated cell growth, invasion, migration, and cytokinesis. Oncogene 23(30):<br />

5193–5202.<br />

Shinomiya, Nariyoshi, Chong Feng Gao, Qian Xie, Margaret Gustafson, David J. Waters, Yu-Wen<br />

Zhang, and George F. Vande Woude. 2004. RNA interference reveals that ligand-independent<br />

Met activity is required for tumor cell signaling and survival. Cancer Research 64(21):<br />

7962–7970.<br />

Vande Woude, George F., Gary J. Kelloff, Raymond W. Ruddon, Han-Mo Koo, Caroline C. Sigman,<br />

J. Carl Barrett, Robert W. Day, Adam P. Dicker, Robert S. Kerbel, David R. Parkinson, and<br />

William J. Slichenmyer. 2004. Reanalysis of cancer drugs: old drugs, new tricks. Clinical<br />

Cancer Research 10(11): 3897–3907.<br />

Zhang, Yu-Wen, Carrie Graveel, Nariyoshi Shinomiya, and George F. Vande Woude. 2004.<br />

Met decoys: will cancer take the bait? Cancer Cell 6(1): 5–6.<br />

From left to right, back row: Zhang, Su, Gao, DeGroot, Staal<br />

middle row: Hay, Xie, Tesfay, Kaufman, Moshkovitz<br />

front row: Reed, Vande Woude, Graveel<br />

52


Laboratory of Tumor Metastasis and Angiogenesis<br />

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

Dr. Webb received his Ph.D. in cell biology from the University of East Anglia,<br />

England, in 1995. He then served as a postdoctoral fellow in the laboratory of<br />

George Vande Woude in the Molecular Oncology Section of the Advanced<br />

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

Institute–Frederick Cancer Research and Development Center, Maryland<br />

(1995–1999). Dr. Webb joined VARI as a <strong>Scientific</strong> Investigator in October 1999.<br />

Staff<br />

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

Jeremy Miller, Ph.D.<br />

David Monsma, Ph.D.<br />

Emily Eugster, M.S.<br />

Sujata Srikanth, M.Phil.<br />

Meghan Sheehan, B.S.<br />

Laboratory Members<br />

Visiting Scientist<br />

Gustavo Cumbo-Nacheli, M.D.<br />

Research Interests<br />

W<br />

e<br />

use model systems ranging from cellbased<br />

assays to preclinical animal<br />

models and clinical subjects to identify<br />

and validate biomarkers and therapeutic targets of<br />

aggressive cancer. The ability to navigate<br />

seamlessly across these diverse model systems<br />

(for example, comparing data from mouse<br />

models to that from human clinical trials) and<br />

between the various molecular platforms is<br />

essential in optimizing the translational research<br />

pipeline (Fig. 1).<br />

translate our investigational findings<br />

into clinical practice, initially in the<br />

areas of improved diagnostics and<br />

pharmacogenomics. Our discoveries<br />

of new, potentially “drugable”<br />

targets is being coupled with siRNA<br />

technologies to verify the functional<br />

validity of targets in preclinical<br />

models. Companion diagnostics are<br />

also being developed that could be<br />

used to assess drug efficacy in<br />

patients. We are beginning to partner<br />

with pharmaceutical companies to<br />

validate new therapeutic areas for<br />

existing drugs, as well as to test<br />

predictions of optimal drug<br />

combinations. At some point in the<br />

near future, accurate diagnosis of<br />

disease will naturally transition to<br />

appropriate treatment.<br />

Our goal is to efficiently<br />

Tumor metastasis<br />

Metastasis accounts for the majority of<br />

cancer-related mortalities. The active recruitment<br />

of tumor vasculature, termed angiogenesis,<br />

is integral to both tumor growth and metastasis.<br />

Our laboratory currently uses both in vitro and in<br />

vivo systems to study metastasis and angiogenesis.<br />

Using a variety of molecular technologies, we<br />

have identified genomic and proteomic correlates<br />

of metastatic disease that may be future<br />

biomarkers and/or molecular targets for diagnosis<br />

Figure 1. Overview of translational research. Commencing with<br />

human research subjects, we can readily transition through specimen<br />

and data collection, data analysis, preclinical models, and ultimately<br />

clinical trials.<br />

53


and treatment. Using our proprietary informatics<br />

system (described below), we have identified<br />

several genes that distinguish normal from<br />

abnormal colon tissue and predict the metastatic<br />

outcome of patients with colorectal cancer.<br />

The potential diagnostic applications of these data<br />

are currently being pursued in a larger cohort of<br />

patients. Our findings to date suggest we can<br />

accurately diagnose colon cancer in pathological<br />

samples and, moreover, predict the likelihood<br />

of metastatic relapse well in advance of<br />

clinical presentation.<br />

In addition, we are using laser capture<br />

microdissection in conjunction with genomic and<br />

proteomic technologies to identify key tumor-host<br />

interactions during metastatic progression, with<br />

emphasis on identifying the molecular factors that<br />

contribute to metastatic dormancy in the liver. A<br />

number of candidate genes are now being pursued<br />

as potential targets of future treatments. For this<br />

purpose, we have developed retroviral and<br />

lentiviral systems for delivering small hairpin RNA<br />

(shRNA) molecules that target candidate genes.<br />

We are knocking-out the expression of several<br />

potential mediators of the metastatic phenotype in<br />

human tumor cell lines and assessing the effects in<br />

orthotopic murine xenografts.<br />

Multiple myeloma<br />

Some 40,000 Americans are living with<br />

multiple myeloma, and deaths are over 11,000 per<br />

year, usually within three years of diagnosis.<br />

Treatment of this highly aggressive cancer is<br />

extremely limited. Without in-depth study into the<br />

molecular causes of multiple myeloma, near-term<br />

advances in diagnosis and treatment are unlikely.<br />

At the end of 2002, with the generous support<br />

of the McCarty Foundation and Ralph Hauenstein,<br />

we initiated the development of a dedicated<br />

multiple myeloma research laboratory (MMRL).<br />

Our specific goal is to use our unique integrated<br />

approach to identify optimal treatments for<br />

patients. We are collaborating with Keith Stewart<br />

at the Princess Margaret Hospital, Toronto, as well<br />

as with local hematologists, oncologists,<br />

pathologists, and other specialists. Through the<br />

collection of detailed clinical information such as<br />

treatment response (efficacy and toxicity), coupled<br />

with single nucleotide polymorphism, gene<br />

expression, and proteomic analysis of collected<br />

samples, we aim to identify molecular correlates of<br />

therapeutic response and disease progression.<br />

Systems biology<br />

XenoBase (patent pending) is a fully integrated<br />

genomic/proteomic/medical informatics system<br />

that includes analysis and annotation tools. Raw<br />

data from molecular analyses can be associated<br />

with specimens and subjects of interest, and<br />

comparative analysis can be performed on data<br />

across platforms and species. XenoBase allows for<br />

direct correlation between the subject, sample, and<br />

experimental parameters and the molecular data.<br />

Literature-based and gene ontology annotation<br />

software have been incorporated, along with<br />

specific metrics for biomarker and target discovery.<br />

Current therapeutics that specifically target<br />

molecular aberrations can also readily be identified<br />

(Fig. 2). Thus, XenoBase represents an integrated<br />

system for basic bimolecular research, clinical<br />

diagnostics, and new pharmacogenomic strategies<br />

for the future.<br />

XenoBase is written primarily in C#.NET and<br />

uses a rich Windows GUI for robust client<br />

interaction. All statistical algorithms are written<br />

in C++ and optimized for a Windows 32-bit<br />

operating system. This approach combines the<br />

speed of C++ with the easy maintenance of the<br />

.NET framework. The application itself is<br />

structured into four primary layers. The GUI layer<br />

allows for client interaction with the system. The<br />

Controller layer provides the bulk of the code and<br />

is responsible for controlling interactions between<br />

the GUI, the algorithms, and the database. The<br />

Algorithm components are part of the controller<br />

layer but are segmented to work independently<br />

and are optimized for performance using C++.<br />

Finally, the Database layer houses the database<br />

I/O code and “plumbing” necessary to keep the<br />

application database independent and allow the<br />

transactional objects to interact without the<br />

overhead of database-specific calls. The code<br />

itself uses a standard .NET exception handling<br />

framework and is documented internally using<br />

code-blocks and inline comments. The use of<br />

C#.NET allows for the automatic generation of<br />

both an object model and API documentation.<br />

The system was built with expandability in<br />

mind. The layered approach allows new user<br />

interfaces (Internet application, PDA, web<br />

54


Figure 2. Molecular<br />

networks associated<br />

with aggressive<br />

mesothelioma.<br />

This figure shows the<br />

interconnectivity<br />

between genes<br />

associated with rapidly<br />

progressing<br />

mesothelioma.<br />

Mapping genomic<br />

correlates of disease to<br />

highly curated<br />

molecular pathways can<br />

identify the underlying<br />

molecular mechanisms<br />

of the disease. This<br />

information could be<br />

used for diagnostic<br />

applications, as well as<br />

identification of key<br />

steps that may<br />

represent intervention<br />

points for treatment.<br />

Here, EGFR was<br />

identified as a key<br />

convergence point that<br />

appears hyperactivated<br />

in aggressive<br />

mesothelioma. Pathway<br />

mapping was generated<br />

using MetaCore<br />

(GeneGo, Inc., St.<br />

Joseph, MI). For more<br />

information on this tool,<br />

see http://genego.com).<br />

services, etc.) to be developed without<br />

redeveloping core functionality. The .NET<br />

framework provides ready access to development<br />

tools and plug-ins, speeding development of new<br />

features and allowing easy integration with<br />

virtually any software platform. The use of<br />

ADO.NET allows for seamless integration with a<br />

variety of databases, including Microsoft SQL<br />

Server, IBM DB/2, Oracle, and MySQL. Finally,<br />

the system makes use of a database-driven storage<br />

system and is database-independent. Its design<br />

includes scripts for database creation and baseline<br />

data setup and uses ODBC (OLEDB) for rapid<br />

execution and flexible database binding.<br />

External Collaborators<br />

James R. Baker, University of Michigan Health System, Ann Arbor, Michigan<br />

Lonson L. Barr, Michigan State University College of Osteopathic Medicine, Grand Rapids, Michigan<br />

Andrej Bugrim, GeneGo, Inc., St. Joseph, Michigan<br />

Alan D. Campbell, Spectrum Health Cancer Center, Grand Rapids, Michigan<br />

Sandra L. Cottingham, Pamela G. Kidd, Susan M. Mammina, and Timothy J. Pelkey, Spectrum Health,<br />

Pathology & Laboratory Medicine, Grand Rapids, Michigan<br />

Alan T. Davis, Michigan State University and Spectrum Health, Grand Rapids, Michigan<br />

Michael Dobbs, Anthony J. Foster, Pamela Grady, Thomas J. Monroe, Linda C. Pool, Deborah Ritz-<br />

Holland, Marcy Ross, Angela R. Tiberio, and Michael J. Warzynski, Spectrum Health, Grand<br />

Rapids, Michigan<br />

55


Timothy Fitzgerald, St. Mary’s Mercy Medical Center, Grand Rapids, Michigan<br />

Neal Goodwin, ProNAi, Kalamazoo, Michigan<br />

Jason Joseph, Order Streams Management, Inc., Grand Rapids, Michigan<br />

Donald G. Kim and Martin A. Luchtefeld, The Ferguson Clinic, Grand Rapids, Michigan<br />

David E. Langholz, Richard F. McNamara, and Timothy C. Vander Meulen, West Michigan Heart,<br />

Grand Rapids, Michigan<br />

Eric P. Lester, Oncology Care Associates, P.L.L.C., St. Joseph, Michigan<br />

Martin McMahon, University of California, San Francisco<br />

John B. O’Donnell, Grand Rapids Medical Education & Research Center, Grand Rapids, Michigan<br />

Gilbert S. Omenn, University of Michigan Medical School, Ann Arbor, Michigan<br />

Leon Oostendorp, Towers Surgeons, P.C., Grand Rapids, Michigan<br />

Timothy J. O’Rourke, Cancer & Hematology Centers of Western Michigan, P.C., Grand Rapids,<br />

Michigan<br />

Harvey I. Pass, Barbara Ann Karmanos Cancer Institute, Detroit, Michigan<br />

Keith Stewart, McLaughlin Centre for Molecular Medicine, Toronto, Canada<br />

Annette Thelen, Michigan State University, East Lansing<br />

Guenter Tusch, Grand Valley State University, Allendale, Michigan<br />

Recent Publications<br />

Creighton, C.J., J.L. Bromberg-White, D. Misek, D. Monsma, F. Brichory, R. Kuick, T.J. Giordano,<br />

W. Gao, G.S. Omenn, C.P. Webb, and S.M. Hanash. In press. Reciprocal tumor-host<br />

interactions revealed by gene expression profiling of lung adenocarcinoma xenografts.<br />

Molecular Cancer Therapeutics.<br />

Bromann, Paul A., Hasan Korkaya, Craig P. Webb, Jeremy Miller, Tammy L. Calvin, and Sara A.<br />

Courtneidge. <strong>2005</strong>. Platelet-derived growth factor stimulates Src-dependent mRNA<br />

stabilization of specific early genes in fibroblasts. Journal of Biological Chemistry 280(11):<br />

10253–10263.<br />

Webb, Craig P., and Harvey I. Pass. 2004. Translational research: from accurate diagnosis to<br />

appropriate treatment. Journal of Translational Medicine 2: 22 pp.<br />

From left to right: Bromberg-White, Monsma, Sheehan, Cumbo-Nacheli, Miller,<br />

Eugster, Srikanth, Webb<br />

56


High-throughput processing of antibody microarrays.<br />

Microscope slides containing 12 (A) or 48 (B) identical microarrays per slide were produced, and each<br />

microarray was incubated with a different sample. The insets show microarrays containing 288 spots (from A)<br />

and 96 spots (from B). The arrays were separated by a hydrophobic wax border that was imprinted onto the<br />

slide using a custom-built device (patent pending). The device uses an interchangable printing cartridge that<br />

can be designed to imprint any user-defined pattern onto a slide. The ability to process multiple samples on one<br />

slide increases the throughput and decreases the cost of the experiments and enables large-scale projects such as<br />

clinical biomarker studies.<br />

(Brian Haab)<br />

57


Laboratory of Chromosome Replication<br />

Michael Weinreich, Ph.D.<br />

Dr. Weinreich received his Ph.D. in biochemistry from the University of<br />

Wisconsin–Madison in 1993. He then was a postdoctoral fellow in the laboratory of<br />

Bruce Stillman, director of the Cold Spring Harbor Laboratory, New York, from 1993<br />

to 2000. Dr. Weinreich joined VARI as a <strong>Scientific</strong> Investigator in March 2000.<br />

Staff<br />

Carrie Gabrielse, B.S.<br />

Jeffrey Kasperski, B.S.<br />

Laboratory Members<br />

Jessica Lanning, B.S.<br />

Students<br />

Charles Miller, B.S.<br />

Amber Crampton<br />

Research Interests<br />

Figure 1. Model for the initiation of DNA replication.<br />

We are studying the initiation of<br />

chromosomal DNA replication in<br />

budding yeast and in human cells. The<br />

initiation of DNA synthesis occurs at multiple<br />

replication origins throughout the genome within<br />

S-phase, but each replication origin is activated<br />

only once per cell cycle. Initiation is precisely<br />

controlled because errors (such as initiating<br />

replication multiple times from a single origin)<br />

would cause genome amplification and instability.<br />

Since DNA replication is essential for cell division<br />

and genome instability is a property of many<br />

cancer cells, we are also investigating the aberrant<br />

regulation of initiation factors in cancer.<br />

Initiation occurs at very well defined<br />

sequences in the budding yeast, so we are using<br />

this organism as a genetic model. Initiation<br />

requires at least three steps (see Fig. 1). The first<br />

is origin marking by ORC, a six-subunit complex<br />

that recognizes conserved DNA sequence elements<br />

in all origins. The second step is the assembly of a<br />

large macromolecular complex called the prereplicative<br />

complex, or pre-RC. ORC, Cdc6p,<br />

Cdt1p, and the MCM complex are together<br />

required to form the pre-RC. Additional proteins<br />

associate with the origin during G1, and then this<br />

large complex of proteins is activated to form bidirectional<br />

replication forks by the Cdc7p-Dbf4p<br />

protein kinase. The process of origin activation is<br />

not understood.<br />

Does chromatin structure affect initiation?<br />

Cdc6p is a critical, limiting factor for assembly<br />

of the pre-RC. We have previously shown that<br />

Cdc6p interacts with ORC and that its essential<br />

activity requires a functional ATP-binding motif. A<br />

cdc6-4 mutant that alters the ATP binding domain is<br />

temperature sensitive (ts), and we have used this<br />

property to isolate suppressors of the cdc6-4 ts.<br />

Loss-of-function alleles within the silent<br />

information regulators (SIR2–4) suppress the cdc6-<br />

4 ts in varying degrees. Deletion of SIR2 gives the<br />

best suppression (Fig. 2). We have also shown that<br />

the loss of SIR2 suppresses several replication<br />

initiation mutants but that its function is originspecific.<br />

Sir2p is a histone<br />

deacetylase required for the<br />

formation of an altered<br />

(“silenced”) chromatin domain at<br />

several transcriptionally silent loci<br />

in the budding yeast. Therefore,<br />

one model to explain our finding<br />

proposes that histone acetylation<br />

stimulates pre-RC assembly. We<br />

are currently investigating the<br />

potential mechanism by which<br />

Sir2p inhibits Cdc6p function and<br />

also how the Sir2p origin<br />

specificity is achieved.<br />

58


CDC7 kinase is required for DNA<br />

replication and repair<br />

Another effort in the lab is to<br />

understand how the two-subunit<br />

Cdc7p-Dbf4p kinase triggers<br />

replication initiation. We have<br />

isolated mutants of the kinase that are<br />

proficient in DNA replication but<br />

defective in DNA repair. Dbf4p is<br />

phosphorylated following inhibition<br />

of DNA replication in a MEC1- and<br />

RAD53-dependent manner and this<br />

phosphorylation appears to inhibit<br />

Cdc7p-Dbf4p kinase activity. MEC1<br />

is a homologue of the human<br />

ATM/ATR (PI 3<br />

-related) kinases that<br />

are key regulators of the response to<br />

DNA damage. RAD53 is the homologue<br />

of the human Cds1/Chk2<br />

checkpoint kinase. Genetic data<br />

suggest that RAD53 is required for<br />

survival of our Cdc7p-Dbf4p kinase<br />

mutants and may promote an<br />

alternative function of this kinase,<br />

i.e., to aid recovery from DNA<br />

damage-induced replication arrest.<br />

Determining how the DNA damage<br />

checkpoint pathway alters the activity<br />

of Cdc7p-Dbf4p kinase or modulates<br />

DNA replication and repair is an<br />

ongoing and exciting area of research.<br />

A.<br />

B.<br />

WT<br />

cdc6-4<br />

cdc6-4 orc1∆N<br />

cdc6-4 sir1∆<br />

cdc6-4 sir2∆<br />

cdc6-4 sir3∆<br />

cdc6-4 sir4∆<br />

25˚C<br />

37˚C<br />

Figure 2. A) Representation of Cdc6p showing the conservation of eight<br />

motifs (II through SensII) within the AAA+ family of ATP binding proteins.<br />

A change of K114A within the “Walker A” ATP binding motif results in the<br />

cdc6-4 ts allele. B) Deletion of SIR2, SIR3, or SIR4 suppresses the<br />

cdc6-4 ts. Serial dilutions of wild-type, cdc6-4, and various cdc6-4<br />

double-mutant strains were spotted onto plates at low and high<br />

temperature to determine growth.<br />

From left to right, standing: Kasperski, Weinreich, Miller; seated: Crampton, Gabrielse<br />

59


Laboratory of Cell Signaling and Carcinogenesis<br />

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

Dr. Williams received his Ph.D. in biology from Massachusetts Institute of<br />

Technology in 1996. For three years, he was a postdoctoral fellow at the National<br />

Institutes of Health in the laboratory of Harold Varmus, former Director of NIH.<br />

Dr. Williams joined VARI as a <strong>Scientific</strong> Investigator in July 1999.<br />

Laboratory Members<br />

Staff<br />

Charlotta Lindvall-Weinreich, M.D., Ph.D.<br />

Troy A. Giambernardi, Ph.D.<br />

Katia Bruxvoort, B.S.<br />

Holli Charbonneau, B.S.<br />

Cassandra Zylstra, B.S.<br />

Students<br />

Nicole Evans<br />

Jose Toro<br />

Research Interests<br />

My laboratory is interested in<br />

understanding how alterations in the<br />

Wnt signaling pathway cause human<br />

disease. Wnt signaling is an evolutionarily<br />

conserved process that has been adapted to<br />

function in the differentiation of most tissues<br />

within the body. One of the long-term goals of<br />

our laboratory is to understand how specificity is<br />

generated for the different signaling pathways.<br />

Recently, we have focused on understanding<br />

the role of Wnt signaling in bone formation. We<br />

are interested not only in normal bone<br />

development, but also in whether aberrant Wnt<br />

signaling plays a role in the predisposition of<br />

some common tumor types (for example prostate,<br />

breast, lung, and renal tumors) to metastasize to<br />

and grow in the bone. The long-term goal is to<br />

provide insights that could be used in developing<br />

strategies to lessen the morbidity and mortality<br />

associated with skeletal metastasis.<br />

Wnt signaling in normal bone development<br />

Mutations in the Wnt receptor, Lrp5, have<br />

been causally linked to alterations in human bone<br />

development. We have characterized a mouse<br />

strain deficient in Lrp5 that recapitulates the lowbone-density<br />

phenotype seen in human patients<br />

having this deficiency. We have further shown<br />

that mice carrying mutations in both Lrp5 and<br />

the related Lrp6 protein have even more-severe<br />

defects in bone density.<br />

We also tested whether Lrp5 deficiency<br />

causes changes in bone density due to aberrant<br />

signaling through β-catenin (Fig. 1). To do this,<br />

we created mice carrying an osteoblast-specific<br />

deletion of β-catenin. These mice die within five<br />

weeks of birth due to profound deficiencies in<br />

bone development. A reciprocal experiment, in<br />

which mice missing the Apc gene specifically in<br />

osteoblasts (and therefore expressing elevated<br />

levels of β-catenin), was also performed. These<br />

mutant mice again died very early after birth, and<br />

Figure 1. Bone defects in mice carrying<br />

osteoblast-specific mutations in β-catenin and/or<br />

Apc. Top set of three: micro-CT of femurs from<br />

a) a 30-day-old wild-type mouse, b) a mouse with an<br />

osteoblast-specific deletion of β-catenin or c) a mouse<br />

lacking both Apc and β-catenin in osteoblasts. Lower<br />

two: micro-CT of femurs from d) 12-day-old mice<br />

either wild type or e) carrying an osteoblast-specific<br />

deletion of Apc. Note the presence of severe<br />

osteoporosis in mice lacking β-catenin (b) and<br />

osteopetrosis in those lacking Apc in osteoblasts (e).<br />

The image in (c) demonstrates that loss of β-catenin<br />

is epistatic to the loss of Apc in osteoblasts.<br />

60


they showed a dramatic overgrowth of bone to the<br />

point where very little marrow cavity was present.<br />

Our current work in this project is aimed at<br />

addressing the molecular mechanisms that<br />

underlie these phenotypes. We have isolated<br />

osteoblasts from these mice and are analyzing<br />

them in tissue culture to determine their abilities<br />

to produce and mineralize osteoid. Also, we are<br />

identifying potential downstream mediators of<br />

Wnt signaling in osteoblasts via microarraybased<br />

expression analysis. As a result of this<br />

work (done in collaboration with Tom Clemens),<br />

we found that alterations in Wnt/β-catenin<br />

signaling in osteoblasts led to changes in the<br />

expression of RANKL and osteoprotegerin<br />

(OPG). Consistent with this, histomorphometric<br />

evaluation of bone in the mice with osteoblastspecific<br />

deletions of either Apc or β-catenin<br />

revealed significant alterations in osteoclastogenesis.<br />

We are currently working to address<br />

how other genetic alterations linked to Wnt/βcatenin<br />

signaling affect bone development and<br />

osteoblast function.<br />

Wnt signaling in cancer<br />

Activation of the Wnt signaling pathway<br />

occurs in a significant percentage of prostate<br />

carcinomas. In some cases this is associated with<br />

activating mutations in the β-catenin genes, while<br />

in others there is a loss of APC. Two hallmarks<br />

of advanced prostate cancer are skeletal<br />

osteoblastic metastasis and the androgenindependent<br />

survival of tumor cells. The<br />

association of Wnt signaling with bone growth,<br />

plus the fact that β-catenin can bind to the<br />

androgen receptor and make it more susceptible<br />

to activation with steroid hormones other than<br />

dihydrotestosterone, make Wnt signaling an<br />

attractive candidate to explain some phenotypes<br />

associated with advanced prostate cancer.<br />

We have created mice with a prostatespecific<br />

deletion of the Apc gene. These mice<br />

develop fully penetrant prostate hyperplasia by<br />

four months of age, and these tumors progress to<br />

frank carcinomas by seven months. We are<br />

currently assessing whether these tumors are<br />

androgen-independent, and we are evaluating<br />

whether loss of Apc can synergize with other<br />

genetic alterations to produce higher-grade<br />

prostate carcinoma. It is our ultimate goal to test<br />

whether loss of activation of β-catenin signaling<br />

can lead to a mouse model of prostate cancer<br />

with skeletal metastases.<br />

We are also examining the roles of Lrp5 and<br />

Lrp6 in normal mammary development. We have<br />

found that mice lacking these genes have delays in<br />

normal mammary development. We are currently<br />

assessing the temporal and spatial organization of<br />

Lrp6 and Lrp5 expression during this process.<br />

We are also addressing the relative roles of<br />

Lrp6 and Lrp5 in Wnt1-induced mammary<br />

carcinogenesis. We have found that deficiency in<br />

Lrp5 dramatically inhibits the development of<br />

mammary tumors in this context. The tumors<br />

that do develop have altered morphology. We are<br />

testing the hypothesis that this inhibition of<br />

tumorigenesis is associated with changes in<br />

mammary stem cells.<br />

VARI mutant mouse repository<br />

With partial support from the Michigan<br />

Animal Models Consortium, my laboratory<br />

maintains a repository of mutant mouse strains to<br />

support the general development of animal<br />

models of human disease. The repository<br />

includes strains that can be used with the<br />

RCAS/TVA retroviral gene targeting system,<br />

which my laboratory has used to develop more<br />

efficient ways of accurately and efficiently<br />

modeling human diseases in mice.<br />

External Collaborators<br />

Caroline Alexander, University of Wisconsin–Madison<br />

Mary Bouxsein, Beth Israel Deaconess Medical Center, Boston, Massachusetts<br />

Thomas Clemens, University of Alabama–Birmingham<br />

Marie-Claude Faugere, University of Kentucky, Lexington<br />

Peter Igarashi, University of Texas–Southwestern Medical School, Dallas<br />

Michael T. Lewis and Yi Li, Baylor Breast Center, Houston, Texas<br />

Matthew Warman, Case Western Reserve University, Cleveland, Ohio<br />

61


Recent Publications<br />

Ai, Minrong, Sheri L. Holmen, Wim van Hul, Bart O. Williams, and Matthew W. Warman. <strong>2005</strong>.<br />

Reduced affinity to and inhibition by DKK1 form a common mechanism by which high bone<br />

mass–associated missense mutations in LRP5 affect canonical Wnt signaling. Molecular and<br />

Cellular Biology 25(12): 4946–4955.<br />

Holmen, Sheri L., Scott A. Robertson, Cassandra R. Zylstra, and Bart O. Williams. <strong>2005</strong>.<br />

Wnt-independent activation of ß-catenin mediated by a Dkk-1-Frizzled 5 fusion protein.<br />

Biochemical and Biophysical Research Communications 328(2): 533–539.<br />

Holmen, Sheri L., Cassandra R. Zylstra, Aditi Mukherjee, Robert Sigler, Marie-Claude Faugere,<br />

Mary Bouxsein, Lianfu Deng, Thomas Clemens, and Bart O. Williams. <strong>2005</strong>. Essential role of<br />

ß-catenin in postnatal bone acquisition. Journal of Biological Chemistry 280(22): 21162–21168.<br />

Qian, Chao-Nan, Jared Knol, Peter Igarashi, Fangmin Lin, Uko Zylstra, Bin Tean Teh, and Bart O.<br />

Williams. <strong>2005</strong>. Cystic renal neoplasia following conditional inactivation of Apc in mouse<br />

renal tubular epithelium. Journal of Biological Chemistry 280(5): 3938–3945.<br />

Robertson, Scott A., Jacqueline Schoumans, Brendan D. Looyenga, Jason A. Yuhas, Cassandra R.<br />

Zylstra, Julie M. Koeman, Pamela J. Swiatek, Bin T. Teh, and Bart O. Williams. <strong>2005</strong>. Spectral<br />

karyotyping of sarcomas and fibroblasts derived from Ink4a/Arf-deficient mice reveals<br />

chromosomal instability in vitro. International Journal of Oncology 26(3): 629–634.<br />

Holmen, Sheri L., Troy A. Giambernardi, Cassandra R. Zylstra, Bree D. Buckner-Berghuis, James<br />

H. Resau, J. Fred Hess, Vaida Glatt, Mary L. Bouxsein, Minrong Ai, Matthew L. Warman, and<br />

Bart O. Williams. 2004. Decreased BMD and limb deformities in mice carrying mutations in<br />

both Lrp5 and Lrp6. Journal of Bone and Mineral Research 19(12): 2033–2040.<br />

Spike, Benjamin T., Alexandra Dirlam, Benjamin C. Dibling, James Marvin, Bart O. Williams, Tyler<br />

Jacks, and Kay F. Macleod. 2004. The Rb tumor suppressor is required for stress<br />

erythropoiesis. EMBO Journal 23(21): 4319–4329.<br />

From left to right: Williams, Charbonneau, Toro, Zylstra, Giambernardi, Bruxvoort<br />

62


Laboratory of Structural Sciences<br />

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

Dr. Xu went to Duke University and the University of Texas Southwestern Medical<br />

Center, where he earned his Ph.D. in molecular biology and biochemistry.<br />

Following a postdoctoral fellowship with Carl Pabo at MIT, he moved to<br />

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.<br />

Staff<br />

Schoen Kruse, Ph.D.<br />

Yong Li, Ph.D.<br />

David Tolbert, Ph.D.<br />

X. Edward Zhou, Ph.D.<br />

Laboratory Members<br />

Jennifer Daughtery, B.S.<br />

Amanda Kovach, B.S.<br />

Kelly Suino, B.S.<br />

Tricia Velting, B.S.<br />

Visiting Scientist<br />

Ross Reynolds, Ph.D.<br />

Research Interests<br />

Our laboratory is using x-ray<br />

crystallography and molecular biology<br />

to study structures and functions of key<br />

protein complexes that are important in basic<br />

biology and in drug discovery relevant to human<br />

diseases such as cancers and diabetes. Currently<br />

we are focusing on nuclear hormone receptors<br />

and the Met tyrosine kinase receptor.<br />

The nuclear hormone receptors<br />

Nuclear hormone receptors form a large<br />

family of ligand-regulated and DNA-binding<br />

transcriptional factors, which include receptors<br />

for classic steroid hormones (such as estrogen,<br />

progesterone, androgens, and glucocorticoids),<br />

as well as receptors for peroxisome proliferator<br />

activators, vitamin D, vitamin A, and thyroid<br />

hormones. These classic receptors are among the<br />

most successful targets in the history of drug<br />

discovery. Almost every one has one or more<br />

synthetic ligands currently being used as<br />

medicines. In the last two years, we have<br />

developed the following projects centering on the<br />

structural biology of nuclear receptors.<br />

Peroxisome proliferator–activated receptors<br />

The peroxisome proliferator–activated<br />

receptors (PPARα, δ, and γ) are the key regulators<br />

of glucose and fatty acid homeostasis and as such<br />

are important therapeutic targets for cardiovascular<br />

disease, diabetes, and cancer.<br />

To understand the molecular basis of ligandmediated<br />

signaling, we have determined crystal<br />

structures of each PPAR’s ligand-binding domain<br />

(LBD) bound to diverse ligands including fatty<br />

acids, the lipid-lowering fibrate drugs, and the<br />

new anti-diabetic drugs called glitazones. We<br />

have also determined crystal structures of these<br />

receptors bound to co-activators or co-repressors.<br />

These structures have provided a framework for<br />

understanding the mechanisms of agonists and<br />

antagonists, as well as the mechanisms for<br />

recruitment of co-activators and co-repressors by<br />

nuclear receptors. We are now developing this<br />

project beyond the structures of the ligandbinding<br />

domains and into defining the structures<br />

of large PPAR fragment/DNA complexes.<br />

The human glucocorticoid receptor<br />

The human glucocorticoid receptor (GR) is a<br />

key regulator of energy metabolism and of<br />

homeostasis of the immune system. The GR is<br />

also a classic target of drug discovery due to its<br />

association with numerous pathological<br />

conditions. There are more than 10 GR ligands<br />

(including dexamethasone) that are currently<br />

used for treating such diverse medical conditions<br />

as asthma, allergy, autoimmune diseases, and<br />

cancer. At the molecular level, GR can function<br />

either as a transcriptional activator or repressor.<br />

Both functions are tightly regulated by small<br />

ligands that bind to the GR LBD. To explore the<br />

molecular mechanisms of GR ligand binding and<br />

signaling, we determined a crystal structure of<br />

the GR LBD bound to dexamethasone and a coactivator<br />

motif from TIF2. The structure<br />

revealed a novel LBD-LBD dimer interface, an<br />

unexpected charge clamp responsible for<br />

sequence-specific binding of co-activators, and a<br />

unique ligand-binding pocket that accounts for<br />

63


the specific recognition of diverse GR ligands.<br />

Currently we are studying receptor-ligand<br />

interactions by crystallizing GR with various<br />

steroid or nonsteroid molecules.<br />

The human androgen receptor<br />

The androgen receptor (AR) is the central<br />

molecule in the development and progression of<br />

prostate cancer, and as such it serves as the<br />

molecular target of anti-androgen therapy.<br />

However, the majority of prostate cancer patients<br />

develop resistance to anti-androgen therapy, mostly<br />

due to mutations in this hormone receptor that alter<br />

the three-dimensional structure of the receptor.<br />

These hormone-independent cells are highly<br />

aggressive and are responsible for most deaths from<br />

prostate cancer. In this project, we are aiming to<br />

determine the structures of the mutated AR proteins<br />

that alter the response to anti-hormone therapy. In<br />

collaboration with Donald MacDonnell, we are<br />

working on the crystal structure of the full-length<br />

AR/DNA complex.<br />

Structural genomics of nuclear receptor<br />

ligand binding domains<br />

The LBD of nuclear receptors contains key<br />

structural elements that mediate the receptors’<br />

ligand-dependent regulation, and as such, the<br />

LBD has been the focus of intense structural<br />

studies. There are only a few nuclear receptors for<br />

which the LBD structure remains unsolved. In the<br />

past two years, we have focused on structural<br />

characterization of two of these “orphan”<br />

receptors: constitutive androstane receptor (CAR)<br />

and steroidogenic factor-1 (SF-1). The CAR<br />

structure reveals a compact LBD fold that<br />

contains a small pocket only half the size of the<br />

pocket in PXR, a closely related receptor (Fig. 1).<br />

The constitutive activity of CAR appears to be<br />

mediated by a novel linker helix between the C-<br />

terminal AF-2 helix and helix 10.<br />

On the other hand, SF-1 is regarded as a<br />

ligand-independent receptor, but its LBD<br />

structure reveals the presence of a phospholipid<br />

ligand in a surprisingly large pocket, more than<br />

twice the size of the pocket in the mouse LRH-1,<br />

a closely related receptor (Fig. 2). The bound<br />

phospholipid is readily exchanged and modulates<br />

SF-1 interactions with co-activators. Mutations<br />

designed to reduce the size of the SF-1 pocket or<br />

to disrupt hydrogen bonds with the phospholipid<br />

Figure 1. Structural comparison of CAR vs. PXR.<br />

The ligand binding pockets are shown as the pink<br />

surface. The AF-2 helix of the receptors is in red and<br />

the co-activator helix is in purple.<br />

abolish SF-1/co-activator interactions and reduce<br />

SF-1 transcriptional activity. These findings<br />

establish that SF-1 is a ligand-dependent receptor<br />

and suggest an unexpected link between nuclear<br />

receptors and phospholipid signaling pathways.<br />

We can expect more surprises as structural work<br />

continues on the remaining orphan receptors.<br />

The Met tyrosine kinase receptor<br />

Met is a tyrosine kinase receptor that is<br />

activated by hepatocyte growth factor/scatter<br />

factor (HGF/SF). Aberrant activation of the Met<br />

receptor has been linked with the development<br />

and metastasis of many types of solid tumors and<br />

has been correlated with poor clinical prognosis.<br />

HGF/SF has a modular structure with an N-<br />

terminal domain, four kringle domains, and an<br />

inactive serine protease domain. The structure of<br />

the N-terminal domain with a single kringle<br />

domain (NK1) has been determined; less is<br />

known about the structure of the Met<br />

Figure 2. Ribbon representation of the SF-1/<br />

phospholipid/SHP complex in two views separated<br />

by 90˚. SF-1 is in red and the SHP ID1 motif is in<br />

yellow. The bound phospholipid ligand is shown in a<br />

space-filling representation, with carbon, oxygen,<br />

nitrogen, and phosphate depicted as green, red, blue,<br />

and purple, respectively.<br />

64


extracellular domain. The molecular basis of the<br />

Met-HGF/SF interaction and the activation of<br />

Met signaling by this interaction remain poorly<br />

understood. In collaboration with George Vande<br />

Woude and Ermanno Gherardi, we are<br />

developing this project to solve the crystal<br />

structure of the Met receptor/HGF complex.<br />

External Collaborators<br />

Doug Engel, University of Michigan, Ann Arbor<br />

Ermanno Gherardi, University of Cambridge, U.K.<br />

Steve Kliewer, University of Texas Southwestern Medical Center, Dallas<br />

Millard Lambert, GlaxoSmithKline Inc., Research Triangle Park, North Carolina<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, University of Texas Medical Branch at Galveston<br />

Ming-Jer Tsai, Baylor College of Medicine, Houston, Texas<br />

Recent Publications<br />

Li, Y., M. Choi, K. Suino, A. Kovach, J. Daugherty, S.A. Kliewer, and H.E. Xu. In press. Structural<br />

and biochemical basis for selective repression of the orphan nuclear receptor LRH-1 by SHP.<br />

Proceedings of the National Academy of Sciences U.S.A.<br />

Li, Yong, Mihwa Choi, Greg Cavey, Jennifer Daugherty, Kelly Suino, Amanda Kovach, Nathan C.<br />

Bingham, Steven A. Kliewer, and H. Eric Xu. <strong>2005</strong>. Crystallographic identification and<br />

functional characterization of phospholipids as ligands for the orphan nuclear receptor<br />

steroidogenic factor-1. Molecular Cell 17(4): 491–502.<br />

Haffner, Curt D., James M. Lenhard, Aaron B. Miller, Darryl L. McDougals, Kate Dwornik, Olivia R.<br />

Ittoop, Robert T. Gampe, Jr., H. Eric Xu, Steve Blanchard, Valerie G. Montana, Tom G. Consler,<br />

Randy K. Bledsoe, Andrea Ayscue, and Dallas Croom. 2004. Structure-based design of potent<br />

retinoid X receptor α agonists. Journal of Medicinal Chemistry 47(8): 2010–2029.<br />

Suino, Kelly, Li Peng, Ross Reynolds, Yong Li, Ji-Young Cha, Joyce J. Repa, Steven A. Kliewer, and<br />

H. Eric Xu. 2004. The nuclear xenobiotic receptor CAR: structural determinants of constitutive<br />

activation and heterodimerization. Molecular Cell 16(6): 893–906.<br />

From left to right: Li, Zhou, Tolbert, Daugherty, Xu, Velting, Kruse, Suino, Kovach<br />

65


Laboratory of Mammalian Developmental Genetics<br />

Nian Zhang, Ph.D.<br />

Dr. Zhang received his M.S. in entomology from Southwest Agricultural University,<br />

People’s Republic of China, in 1985 and his Ph.D. in molecular biology from the<br />

University of Edinburgh, Scotland, in 1992. From 1992 to 1996, he was a<br />

postdoctoral fellow at the Roche Institute of Molecular Biology. He next served as a<br />

postdoctoral fellow (1996) and a Research Associate (1997–1999) in the laboratory<br />

of Tom Gridley in mammalian developmental genetics at the Jackson Laboratory, Bar<br />

Harbor, Maine. Dr. Zhang joined VARI as a <strong>Scientific</strong> Investigator in December 1999.<br />

Staff<br />

Wei Ma, Ph.D.<br />

Lisheng Zhang, Ph.D.<br />

Kate Groh, B.S.<br />

Liang Kang<br />

Laboratory Members<br />

Student<br />

William Bond<br />

Research Interests<br />

We are interested in understanding the<br />

cellular and molecular mechanisms<br />

underlying pattern formation during<br />

embryonic development. We previously cloned<br />

and targeted the mouse Lunatic fringe (Lfng) gene,<br />

which plays an important role in embryo<br />

segmentation. Mice homozygous for the Lfng<br />

mutation suffer from severe malformation of their<br />

axial skeleton as a result of irregular somite<br />

formation during embryonic development. Lfng<br />

encodes a secreted signaling molecule essential for<br />

regulating the Notch signaling pathway in mice.<br />

We showed that Lfng expression was in response to<br />

a biological clock that oscillated once during the<br />

formation of each segment, and the failure of the<br />

Lfng mutants in responding to this clock resulted in<br />

the abnormal segmentation phenotype. We want to<br />

understand how the rhythmic expression of Lfng is<br />

controlled. Our recent studies indicate that the<br />

cyclic expression of Lfng is controlled by a<br />

negative feedback loop. The signals transmitted<br />

through the loop are mediated by the components<br />

in the Notch signaling pathway. We found that<br />

Hes7 can directly bind to the N-boxes in the 5′-<br />

regulatory region of the Lfng gene and its own<br />

promoter and repress their transcription in vitro.<br />

Hes7 can also override the Notch-mediated<br />

activation of the Lfng promoter. However, our<br />

results suggest that Hes7 needs a co-factor to<br />

achieve this suppression in vivo. Our data suggests<br />

that when NOTCH1 is modified by LFNG, it<br />

becomes receptive to the signal from DLL3, thus<br />

activating its downstream target. This target<br />

interacts with Hes7 to turn down Hes7 and Lfng.<br />

When the level of HES7 is down, it relieves its<br />

repression on Lfng and its own promoter, and thus<br />

the next cycle begins. We have also demonstrated<br />

that the 3′-untranslated region (UTR) is important<br />

for the rapid degradation of Lfng mRNA, which<br />

ensures accurate oscillation.<br />

Germ cell development<br />

The second focus of our laboratory is on germ<br />

cell development, particularly the mechanisms that<br />

govern germ cell migration, survival,<br />

spermatogonial stem cell renewal, and<br />

differentiation, as well as their implications for<br />

human disease. It is unclear how spermatogonial<br />

stem cells are regenerated during the entire<br />

reproductive life in mammals. Previous studies on<br />

the nematode Caenorhabditis elegans have shown<br />

that the Notch/lin12-mediated signal transduction<br />

pathway is important if germ cells are to remain in<br />

an undifferentiated state. Mutations that<br />

compromise this pathway force germ cells to enter<br />

meiosis earlier than normal. A constitutively<br />

activated signal prevents germ cells from entering<br />

meiosis, resulting in overproliferation of germ<br />

cells, a phenotype called “germ cell tumor.” Given<br />

the fact that some members in the Notch signaling<br />

pathway are expressed in the testis, we speculate<br />

that Notch signaling may play a similar role in<br />

spermatogonial differentiation in mammals. We<br />

will further examine the role Notch signaling may<br />

play during spermatogenesis using transgenic<br />

animals and conditional gene targeting.<br />

66


We are also studying spontaneous mutations<br />

that cause sterility. In mice, primordial germ cells<br />

(PGCs) are differentiated from the epiblasts during<br />

an early embryonic stage. After their formation,<br />

they migrate through the dorsal mesentery and<br />

enter the genital ridge, where they collaborate with<br />

the somatic gonad cells to form the gonads. The<br />

PGCs proliferate during their migration. We are<br />

studying the spontaneous mutation atrichosis (at),<br />

which causes male and female sterility.<br />

Preliminary data suggest that this mutation affects<br />

fetal germ cell proliferation. We found that by 12.5<br />

dpc, there already are significantly fewer germ<br />

cells in the gonads of atrichosis embryos relative to<br />

the wild type. We have mapped the atrichosis<br />

mutation to a 270-kb region on chromosome 10.<br />

We are now screening the candidate genes by<br />

transgenic rescue and direct sequencing.<br />

Another mutation we are studying is in the sks<br />

gene; this mutation affects normal meiosis in both<br />

sexes. Our results indicate that sks is required for<br />

the metaphase/anaphase transition in meiosis I. In<br />

the sks mutant, homologous chromosomes fail to<br />

separate; therefore, meiosis is stopped at MI. We<br />

have demonstrated that this failure is due to the<br />

inability of the sks mutant to degrade securin in the<br />

primary spermatocytes and possibly in the oocytes.<br />

From left to right, back row: Kang, Ma, L. Zhang, Groh;<br />

front row: N. Zhang, Bond<br />

67


Double staining of protein and RNA in mouse testis<br />

This is a section of mouse testis stained with anti-MPM-2 antibody (green) and an RNA probe (red) to label the<br />

spermatocytes. This image is from studies of germ cell development and the implications for human disease.<br />

(Nian Zhang)<br />

68


Daniel Nathans Memorial Award


Daniel Nathans Memorial Award<br />

The Daniel Nathans Memorial Award was established in memory<br />

of Dr. Daniel Nathans, a distinguished member of our scientific<br />

community and a founding member of VARI’s Board of <strong>Scientific</strong><br />

Advisors. We established this award to recognize individuals who<br />

emulate Dan and his contributions to biomedical and cancer<br />

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<br />

reality. The Daniel Nathans Memorial Award was announced at<br />

our inaugural symposium, “Cancer & Molecular Genetics in the<br />

Twenty-First Century,” in September 2000.<br />

2004 Award to<br />

Dr. Brian Druker<br />

The 2004 recipient of the Daniel Nathans Memorial Award is Brian Druker, M.D., of the Oregon<br />

Health & Science University Cancer Institute and the Howard Hughes Medical Institute. Dr. Druker’s<br />

research focuses on translating knowledge of the molecular pathogenesis of cancer into specific<br />

therapies, and he is investigating the optimal use of molecularly targeted agents. Dr. Druker will visit<br />

Grand Rapids in September <strong>2005</strong> to deliver two lectures, one directed to a scientific audience and the<br />

second directed to the general public.<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 />

71


Postdoctoral Fellowship Program


Postdoctoral Fellowship Program<br />

The Van Andel Research Institute provides postdoctoral training opportunities to Ph.D. scientists<br />

beginning their research careers. The fellowships help promising scientists advance their knowledge<br />

and research experience while at the same time supporting the research endeavors of VARI. The<br />

fellowships are funded in three ways: 1) by the laboratories to which the fellow is assigned; 2) by the<br />

VARI Office of the Director; or 3) by outside agencies. Each fellow is assigned to a scientific<br />

investigator who oversees the progress and direction of research. Fellows who worked in VARI<br />

laboratories in 2004 and early <strong>2005</strong> are listed below.<br />

Eduardo Azucena<br />

Wayne State University, Detroit, Michigan<br />

VARI mentor: Sara Courtneidge<br />

Paul Bromann<br />

Northwestern University, Evanston, Illinois<br />

VARI mentor: Sara Courtneidge<br />

Jennifer Bromberg-White<br />

Pennsylvania State University College of<br />

Medicine, Hershey<br />

VARI mentor: Craig Webb<br />

Jun Chen<br />

West China University of Medical Sciences,<br />

Chengdu, China<br />

VARI mentor: Nian Zhang<br />

Yunju Chen<br />

University of Glasgow, U.K.<br />

VARI mentor: Arthur Alberts<br />

Philippe Depeille<br />

University of Montpellier, France<br />

VARI mentor: Nicholas Duesbery<br />

Mathew Edick<br />

University of Tennessee, Memphis<br />

VARI mentor: Cindy Miranti<br />

Kathryn Eisenmann<br />

University of Minnesota, Minneapolis<br />

VARI mentor: Arthur Alberts<br />

Kunihiko Futami<br />

Tokyo University of Fisheries, Japan<br />

VARI mentor: Bin Teh<br />

Chongfeng Gao<br />

Tokyo Medical and Dental University, Japan<br />

VARI mentor: George Vande Woude<br />

Troy Giambernardi<br />

University of Texas Health Science Center, San<br />

Antonio<br />

VARI mentor: Bart Williams<br />

Carrie Graveel<br />

University of Wisconsin – Madison<br />

VARI mentor: George Vande Woude<br />

Holly Holman<br />

University of Glasgow, U.K.<br />

VARI mentor: Arthur Alberts<br />

Hasan Korkaya<br />

International Center for Genetic Engineering<br />

and Biotechnology, New Delhi, India<br />

VARI mentor: Sara Courtneidge<br />

Schoen Kruse<br />

University of Colorado, Boulder<br />

VARI mentor: Eric Xu<br />

Xudong Liang<br />

Qinghai Medical University, Xining, China<br />

VARI mentor: Nicholas Duesbery<br />

Phumzile Loudidi<br />

Cambridge University, England<br />

VARI mentor: Eric Xu<br />

Wei Ma<br />

Chinese Academy of Science, Beijing<br />

VARI mentor: Nian Zhang<br />

75


Donald Pappas, Jr.<br />

Louisiana State University, Baton Rouge<br />

VARI mentor: Michael Weinreich<br />

Ian Pass<br />

University of Dundee, Scotland<br />

VARI mentor: Sara Courtneidge<br />

Michael Shafer<br />

Michigan State University, East Lansing<br />

VARI mentor: Brian Haab<br />

Muthu Shanmugam<br />

National University of Singapore, Singapore<br />

VARI mentor: Brian Haab<br />

Paul Spilotro<br />

St. George University, Grenada<br />

VARI mentor: Nicholas Duesbery<br />

Suganthi Sridhar<br />

Southern Illinois University, Carbondale<br />

VARI mentor: Cindy Miranti<br />

Jun Sugimura<br />

University of Morioka Medical School, Japan<br />

VARI mentor: Bin Teh<br />

Min-Han Tan<br />

National University of Singapore, Singapore<br />

VARI mentor: Bin Teh<br />

Rebecca Uzarski<br />

Michigan State University, East Lansing<br />

VARI mentor: Sara Courtneidge<br />

Pengfei Wang<br />

Fourth Military Medical University, Xian, China<br />

VARI mentor: Bin Teh<br />

Qian Xie<br />

Fudan University, Shanghai, China<br />

VARI mentor: George Vande Woude<br />

Chun Zhang<br />

Tokyo Medical and Dental University, Japan<br />

VARI mentor: Bin Teh<br />

Lisheng Zhang<br />

Chinese Academy of Science, Beijing<br />

VARI mentor: Nian Zhang<br />

Xiaoyin Zhou<br />

University of Alabama, Birmingham<br />

VARI mentor: Eric Xu<br />

76


Student Programs


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

The Grand Rapids Pre-College Engineering Program (GRAPCEP) is administered by Davenport<br />

College and jointly sponsored and funded by Pfizer, Inc., and VARI. The program is designed to<br />

provide selected high school students, who have plans to major in science or genetic engineering in<br />

college, the opportunity to work in a research laboratory. In addition to training in research methods,<br />

the students also learn workplace success skills such as teamwork and leadership. The three 2004<br />

GRAPCEP students were<br />

Lynda Gladding (Resau/Duesbery)<br />

Union High School<br />

Ricky Gonzales (Resau/Duesbery)<br />

Ottawa Hills High School<br />

Mehreteab Mengsteab (Xu)<br />

Creston High School<br />

79


Summer Student Internship Program<br />

The VARI student internships were established to provide college students with an opportunity<br />

to work with professional researchers in their fields of interest, to use state-of-the-art equipment and<br />

technologies, and to learn valuable people and presentation skills. At the completion of the 10-week<br />

program, the students summarize their projects in an oral presentation.<br />

From May 2004 to March <strong>2005</strong>, VARI hosted 44 students from 17 colleges and universities in<br />

formal summer internships under the Frederik and Lena Meijer Student Internship Program and in<br />

other student positions during the year. An asterisk (*) indicates a Meijer student intern.<br />

Aquinas College, Grand Rapids, Michigan<br />

Brent Goslin* (Weinreich)<br />

Calvin College, Grand Rapids, Michigan<br />

Arianne Folkema* (Williams)<br />

Jason Koning (Williams)<br />

Cornell University, Ithaca, New York<br />

Susan Kloet (Teh)<br />

Dalhousie University, Nova Scotia<br />

Jasmine Belanger* (Haab)<br />

Grand Rapids Community College, Michigan<br />

Jose Toro (Williams)<br />

Grand Valley State University,<br />

Allendale, Michigan<br />

Timothy Bearup* (Cao)<br />

Jack DeGroot (Vande Woude)<br />

Nicole Evans (Williams)<br />

Erik Freiter (Miranti)<br />

Lisa Orcasitas (Duesbery)<br />

Benjamin Staal* (Vande Woude)<br />

Neil Swanson (Duesbery)<br />

Kelli VanDussen (Weinreich)<br />

Tricia Velting* (Xu)<br />

Grove City College, Pennsylvania<br />

Sarah Feenstra (Vande Woude)<br />

Hope College, Holland, Michigan<br />

Marie Graves (Cavey)<br />

Wendy Johnson (Cavey)<br />

Tom LaRoche (Haab)<br />

Richard Schildhouse (Haab)<br />

Mary VerHeulen (Vande Woude)<br />

Michigan State University, East Lansing<br />

Aaron DeWard (Weinreich/Alberts)<br />

Stephanie Ellison* (Vande Woude)<br />

Mia Hemmes* (Duesbery)<br />

Jennifer Kaufman (Resau)<br />

Yaojian Liu (Alberts)<br />

Charles Miller (Weinreich)<br />

Michigan Technological University, Houghton<br />

Hien Dang (Resau)<br />

Nanjing Medical University, China<br />

Yong-jun Jiao (Cao)<br />

Jin Zhu (Cao)<br />

Xin Wang (Cao)<br />

University of Bath, United Kingdom<br />

William Bond (Zhang)<br />

Katharine Collins (Alberts)<br />

Amber Crampton (Weinreich)<br />

Victoria Hammond (Weinreich)<br />

Amy Percival (Resau)<br />

80


University of Detroit Mercy<br />

Brandon Leeser (Resau)<br />

University of Illinois at Urbana-Champaign<br />

Huang Tran (Resau)<br />

University of Michigan, Ann Arbor<br />

Benjamin Briggs* (Furge)<br />

Michael Wells* (Teh)<br />

Natalie Wolters* (Courtneidge)<br />

University of Richmond, Virginia<br />

Jeremiah NcNamara* (Webb)<br />

Western Michigan University, Kalamazoo<br />

Kenneth Olinger* (Resau)<br />

Nicole Repair* (Miranti)<br />

81


Han-Mo Koo Memorial Seminar Series


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><br />

Investigator from 1999 until his passing in May of 2004.<br />

2004<br />

January<br />

Karen Vousden, Beatson Institute for Cancer Research, Glasgow, Scotland<br />

“The p53 pathway as a therapeutic target”<br />

Patrick Brophy, University of Michigan, Ann Arbor<br />

“Ureteric budding: controlling two ends of the event”<br />

February<br />

Jeffrey Settleman, Harvard Medical School, Boston, Massachusetts<br />

“Rho GTPase signaling in development”<br />

Josef Prchal, Baylor College of Medicine, Houston, Texas<br />

“Tumors, hypoxia, and polycythemic disorders”<br />

March<br />

Robert Weinberg, Whitehead Institute for Biomedical Research, Cambridge, Massachusetts<br />

The Daniel Nathans Lecture: “Mechanisms of human tumor formation”<br />

“How cancer begins” (lay audience)<br />

Mike Caliguiri, Ohio State University, Columbus<br />

“Natural killer cells: biology and clinical implications”<br />

James Basilion, Massachusetts General Hospital, Charlestown, Massachusetts<br />

“Noninvasive imaging of gene expression: imaging multiple targets simultaneously”<br />

David Frank, Harvard University, Cambridge, Massachusetts<br />

“STAT signal transduction in the pathogenesis and treatment of cancer”<br />

April<br />

Ernst-Robert Lengyel, University of Chicago, Illinois<br />

“Regulation of proteolysis by adhesion receptors”<br />

Kathleen Siminovitch, Mount Sinai Hospital, Toronto, Canada<br />

“The Wiskott-Aldrich syndrome protein: forging the link between actin and T cell activation”<br />

Jose Cibelli, Michigan State University, East Lansing<br />

“Embryonic stem cells by parthenogenesis in mammals”<br />

85


May<br />

Francesco Marincola, National Institutes of Health, Bethesda, Maryland<br />

“Anti-cancer vaccines: from bench to bedside and from bedside to bench”<br />

Robert Gallo, University of Maryland, Baltimore<br />

“HIV in the third decade — some lessons from past experiences and future prospects”<br />

June<br />

Cynthia Wetmore, Mayo Clinic, Rochester, Minnesota<br />

“Implications of Ptc haploinsufficiency for the proliferation and cell fate of neuronal precursors”<br />

July<br />

Arnold Glazier, Drug Innovation and Design, Newton, Massachusetts<br />

“Pattern recognition tumor targeting”<br />

Renata Pasqualini, M.D. Anderson Cancer Center, Houston, Texas<br />

“Translating function protein interactions into targeted therapies for cancer and obesity”<br />

September<br />

Douglas Hanahan, University of California, San Francisco<br />

“Mechanisms of angiogenesis, and anti-angiogenic therapies in mouse models of cancer”<br />

Henry Higgs, Dartmouth Medical School, Hanover, New Hampshire<br />

“Comparative molecular physiology of mammalian formin proteins: potent actin assembly factors”<br />

October<br />

Janice P. Dutcher, Our Lady of Mercy Medical Center, New York<br />

“Clinical features and treatment of metastatic renal cell cancer”<br />

Robert L. Heinrikson, Proteos, Kalamazoo, Michigan<br />

“Innovation in design and production of proteins and peptides — the Proteos approach”<br />

Qing-Xiang Sang, Florida State University, Tallahassee, Florida<br />

“Metalloproteases and nonprotease factors in early stages of tumor invasion: new hypotheses on<br />

the mutated stem cells and cancer drug resistance”<br />

November<br />

John Carpten, Translational Genomics Research Institute, Phoenix, Arizona<br />

“Searching for prostate cancer in tumor suppressor genes”<br />

December<br />

Marcos Dantus, Michigan State University, East Lansing<br />

“The development of future biomedical applications using coherent laser control”<br />

Roland S. Annan, GlaxoSmithKline, King of Prussia, Pennsylvania<br />

“A qualitative and quantitative view of phosphorylation-dependent biological function”<br />

86


<strong>2005</strong><br />

January<br />

Ali Shilatifard, St. Louis University, Missouri<br />

“A COMPASS and a GPS in defining molecular machinery in histone modifications,<br />

transcriptional regulation, and human cancer: the coordinates of the genome”<br />

February<br />

Hsueh-Chia Chang, University of Notre Dame, Indiana<br />

“Microfluidic technologies for cancer detection and drug delivery”<br />

Paul A. Krieg, University of Arizona, Tucson<br />

“Growth factor regulation of vascular development”<br />

March<br />

Max Wicha, University of Michigan, Ann Arbor<br />

“Stem cells in normal human breast development and cancer”<br />

Judah Folkman, Harvard Medical School, Boston, Massachusetts<br />

“Platelet angiogenic profile as an early biomarker for cancer”<br />

Robert J. Amato, The Methodist Hospital, Houston, Texas<br />

“Therapeutic updates for the management of patients with renal cell carcinoma”<br />

Aaron M. Zorn, Cincinnati Children’s Hospital Research Foundation, Ohio<br />

“Sox17 and β-catenin signaling in development”<br />

April<br />

Martin E. Hemler, Harvard Medical School and Dana-Farber Cancer Institute, Boston,<br />

Massachusetts<br />

“Cell surface molecular networking: the role of tetraspanin-enriched microdomains”<br />

Alan Mackay-Sim, Griffith University, Australia<br />

“Adult stem cells from olfactory mucosa”<br />

Ravi Salgia, University of Chicago, Illinois<br />

“Role of c-Met in lung cancer”<br />

87


Organization


Van Andel Research Institute Boards<br />

VARI Board of Trustees<br />

David L. Van Andel, Chairman and CEO<br />

Christian Helmus, M.D.<br />

Fritz M. Rottman, Ph.D.<br />

James B. Wyngaarden, M.D.<br />

David L. Van Andel<br />

Board of <strong>Scientific</strong> Advisors<br />

The Board of <strong>Scientific</strong> Advisors advises the CEO and the Board of Trustees, providing<br />

recommendations and suggestions regarding the overall goals and scientific direction of VARI.<br />

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<br />

suggestions specific to the ongoing research, especially in the areas of cancer, genomics, and genetics.<br />

It also coordinates and oversees the scientific review process for the Institute’s research programs.<br />

The members are<br />

Alan Bernstein, Ph.D.<br />

Malcolm Brenner, M.D., Ph.D.<br />

Patrick O. Brown, M.D., Ph.D.<br />

Joan Brugge, Ph.D.<br />

Webster Cavenee, Ph.D.<br />

Frank McCormick, Ph.D.<br />

Davor Solter, M.D., Ph.D.<br />

Bruce Stillman, Ph.D.<br />

91


Van Andel Research Institute<br />

Office of the Director<br />

Director<br />

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

Deputy Director<br />

for Clinical Programs<br />

Deputy Director<br />

for Special Programs<br />

Rick Hay, M.D., Ph.D.<br />

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

Deputy Director<br />

for Research Operations<br />

Director for<br />

Research Administration<br />

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

Roberta Jones<br />

92


Van Andel Research Institute<br />

Administrator to<br />

the Director<br />

Science Editor<br />

Michelle Reed<br />

David E. Nadziejka<br />

Administration Group<br />

From left to right, back row: Dingman, Holman, Stougaard, Ferrell, Carrigan;<br />

front row: Antio, Koo, McGrail, Novakowski<br />

93


Van Andel Institute Administrative Organization<br />

The organizational units listed below provide administrative support to both the Van Andel<br />

Research Institute and the Van Andel Education Institute.<br />

Executive<br />

Steven R. Heacock, Chief Administrative<br />

Officer and General Counsel<br />

R. Jack Frick, Chief Financial Officer<br />

Ann Schoen, Executive Assistant<br />

Communications and Development<br />

Patrick Kelly, Vice President<br />

John Van Fossen<br />

Dianna Davidson<br />

Andrea Nielsen<br />

Margo Pratt<br />

Information Technology<br />

Bryon Campbell, Ph.D.,<br />

Chief Information Officer<br />

David Drolett, Manager<br />

Michael Roe, Manager<br />

Tom Barney<br />

Phil Bott<br />

Kenneth Hoekman<br />

Kimberlee Jeffries<br />

Theo Pretorius<br />

Russell Vander Mey<br />

Candy Wilkerson<br />

Human Resources<br />

Linda Zarzecki, Director<br />

Margie Hoving<br />

Pamela Murray<br />

Angela Plutschouw<br />

Grants and Contracts<br />

Carolyn W. Witt, Director<br />

Rob Junge<br />

Sara O’Neal<br />

David Ross<br />

Finance<br />

Timothy Myers, Controller<br />

Heather Ly, Supervisor<br />

Richard Herrick<br />

Keri Jackson<br />

Angela Lawrence<br />

Susan Raymond<br />

Kevin Tefelsky<br />

Jamie VanPortfleet<br />

Purchasing<br />

Richard Disbrow, Manager<br />

Chris Kutchinski<br />

Amy Poplaski<br />

John Waldon<br />

Facilities<br />

Samuel Pinto, Manager<br />

Jason Dawes<br />

Richard Sal<br />

Richard Ulrich<br />

Security<br />

Kevin Denhof, CPP, Chief<br />

Christen Dingman<br />

Sandra Folino<br />

Emily Young<br />

Glass Washing/ Media Preparation<br />

Heather Frazee<br />

Marlene Sal<br />

Contract Support<br />

Valeria Long, Librarian<br />

(Grand Valley State University)<br />

Jim Kidder, Safety Manager<br />

(Michigan State University)<br />

Raymond Rupp<br />

Patty Sund<br />

Al Troupe<br />

94


95


Van Andel Institute<br />

Van Andel Research Institute<br />

96


Recent VARI Photos


99


100


Back cover photo: Spectral karyotyping of a tumor cell line<br />

Fluorescence microscopy for spectral karyotyping (SKY) analysis of a human tumor cell line. The red light<br />

helps reduce fading in the fluorescent slides, which are light sensitive.<br />

(Julie Koeman)<br />

The Van Andel Institute and/or its affiliated organizations (VARI and VAEI), through its responsible managers,<br />

recruits, hires, upgrades, trains, and promotes in all job titles without regard to race, color, religion, sex,<br />

national origin, age, height, weight, marital status, disability, pregnancy, or veteran status, except where an<br />

accommodation is unavailable and/or it is a bone fide occupational qualification.<br />

Typesetting and printing by RiverRun Press, Parchment, Michigan.

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

Saved successfully!

Ooh no, something went wrong!