+ All Categories
Home > Documents > SYNTHETIC PEPTIDE LIGAND MIMETICS AND TUMOR...

SYNTHETIC PEPTIDE LIGAND MIMETICS AND TUMOR...

Date post: 25-Apr-2018
Category:
Upload: leanh
View: 215 times
Download: 3 times
Share this document with a friend
143
Synthetic Peptide Ligand Mimetics and Tumor Cell Motility Item Type text; Electronic Dissertation Authors Sroka, Thomas Charles Publisher The University of Arizona. Rights Copyright © is held by the author. Digital access to this material is made possible by the University Libraries, University of Arizona. Further transmission, reproduction or presentation (such as public display or performance) of protected items is prohibited except with permission of the author. Download date 09/06/2018 10:56:08 Link to Item http://hdl.handle.net/10150/194831
Transcript

Synthetic Peptide Ligand Mimetics and Tumor Cell Motility

Item Type text; Electronic Dissertation

Authors Sroka, Thomas Charles

Publisher The University of Arizona.

Rights Copyright © is held by the author. Digital access to this materialis made possible by the University Libraries, University of Arizona.Further transmission, reproduction or presentation (such aspublic display or performance) of protected items is prohibitedexcept with permission of the author.

Download date 09/06/2018 10:56:08

Link to Item http://hdl.handle.net/10150/194831

SYNTHETIC PEPTIDE LIGAND MIMETICS AND TUMOR CELL MOTILITY

by

Thomas Charles Sroka

_______________________________

A Dissertation Submitted to the Faculty of the

GRADUATE INTERDISCIPLINARY PROGRAM IN CANCER BIOLOGY

In Partial Fulfillment of the Requirements For the Degree of

DOCTOR OF PHILOSOPHY

In the Graduate College

THE UNIVERISTY OF ARIZONA

2005

2

THE UNIVERSITY OF ARIZONA GRADUATE COLLEGE

As members of the Dissertation Committee, we certify that we have read the dissertation prepared by Thomas Charles Sroka entitled Synthetic Peptide Ligand Mimetics and Tumor Cell Motility and recommend that it be accepted as fulfilling the dissertation requirement for the Degree of Doctor of Philosophy _______________________________________________________________________ Date: (10/21/05)Anne E. Cress, Ph.D. _______________________________________________________________________ Date: (10/21/05)Roger L. Miesfeld, Ph.D. _______________________________________________________________________ Date: (10/21/05)Scott E. Klewer, M.D.

Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation to the Graduate College. I hereby certify that I have read this dissertation prepared under my direction and recommend that it be accepted as fulfilling the dissertation requirement.

________________________________________________ Date: (10/21/05)Dissertation Director: Anne E. Cress, Ph.D.

3

STATEMENT BY AUTHOR

This dissertation has been submitted in partial fulfillment of requirements for an advanced degree at The University of Arizona and is deposited in the University Library to be made available to borrowers under rules of the Library.

Brief quotations from this dissertation are allowable without special permission, provided that accurate acknowledgement of source is made. Requests for permission for extended quotation from or reproduction of this manuscript in whole or in part may be granted by the head of the major department of the Dean of the Graduate College when in his or her judgment the proposed use of the material is in the interests of scholarship. In all other instances, however, permission must be obtained from the author.

SIGNED: Thomas Charles Sroka

4

ACKNOWLEDGEMENTS

In working toward this dissertation the most valuable knowledge gained was an

understanding that research is a collaborative effort. The work presented here would not

be possible without many minds and helping hands. I am eternally grateful to members

of the Cress laboratory, fellow students in the Cancer Biology Interdisciplinary Program,

members of Core Services, and my dissertation committee members.

To my mentor, Anne Cress, thank you. Thank you for your patience, excitement,

and encouragement that have enriched my development as a scientist. With your

guidance I now feel prepared to meet the challenges of Science and challenge my goals.

I am grateful for your honest, critical review of my work and cherish our discussions,

both science and non-science related. I feel lucky to have been a part of your laboratory.

Finally I would like to thank my family, my friends, and my Isis. Thank you for

your support and patience as I travel through an extended career-training path. None of

this work would have been possible without you.

5

DEDICATION

I dedicate this work to anyone that has been a victim, witness, or survivor of

cancer. May it serve as a step toward new therapies, new hope.

6

TABLE OF CONTENTS LIST OF FIGURES………………………………………………………………….9 LIST OF TABLES…………………………………………………………………..12 ABSTRACT…………………………………………………………………………. 13

I. INTRODUCTION……………………………………………………………….15 Cancer Metastasis……………………………………………………………………..15 Prostate Cancer………………………………………………………………………..16 Integrins and Cell Motility…………………………………………………………….19 Protrusive and Contractile Contacts…………………………………………………...21 Focal Adhesion Proteins: The Role of Focal Adhesion Kinase and the Extracellular Signal-Regulated Protein Kinase/Mitogen-Activated Protein Kinase Pathway in Cell Migration………………………………………………………………………24 Isolation of Tumor Cell Adhesion Peptides Using a One-Bead-One-Compound Combinatorial Library……………………………………………………………...27 II. MATERIALS AND METHODS…………………………………………...31 Affinity Precipitation with Peptides…………………………………………………..31 Antibodies and Reagents……………………………………………………………...32 Biotinylation of Cell Surface Proteins………………………………………………...33 Cell Lines and Culture Conditions……………………………………………………33 Cell Motility Assays…………………………………………………………………..34 Cell Adhesion Assay and Peptide Inhibition of Adhesion…………………………….37 Cell Lysis and Western-Blot…………………………………………………………..37 Immunoprecipitation…………………………………………………………………..38 Epithelial and Stromal Cell Adhesion Assay………………………………………….39 FACS Analysis and Confocal Microscopy……………………………………………40 In Vivo Actin Distribution Assay……………………………………………………...40 Immunofluorescence…………………………………………………………………..41 Synthetic Peptides, Fibronectin, Laminin-1, Laminin-5, and Collagen IV…………...42 Laminin-5 Coating Conditions for Motility and Signaling Experiments……………..43 III. IDENTIFICATION AND CHARACTERIZATION OF TUMOR CELL ADHESION PEPTIDES………………………………………………44 Introduction……………………………………………………………………………44 Results…………………………………………………………………………………46 RZ-3, HYD-1 and AG-73 Bind to Prostate Tumor Cell Surfaces………………….46 DU-H Prostate Tumor Cells Adhere to Immobilized Peptide and ECM Proteins…49

7

TABLE OF CONTENTS – CONTINUED-

Tumor Cell Adhesion to Immobilized Peptides is Integrin Dependent…………….51 Inhibition of DU-H Cell Attachment to ECM Proteins…………………………….53 Inhibition of SCC-25 and HFF Co-Culture Interaction by HYD-1 Peptide………..56 Adhesion to Immobilized HYD1 Prevents Cell Spreading and Adhesion Dependent Phosphorylation……………………………………………………...56 Discussion……………………………………………………………………………..61

IV. SYNTHETIC D-AMINO ACID PEPTIDE INHIBITS TUMOR CELL MOTILITY ON LAMININ-5………………………………………...64 Introduction……………………………………………………………………………64 Results…………………………………………………………………………………66 HYD1 Blocks Laminin-5 Dependent Migration and Invasion……………………..66 HYD1 Induces Actin Cytoskeletal Remodeling and Cortactin Mediated Actin Dynamics at the Cell Membrane…………………………………………………72 HYD1 interacts with α6 and α3 Integrins and Activates Integrin-Associated Signaling…………………………………………………………………………75 HYD1 Enhances Signaling on Laminin-5 Resulting in Transient Activation of ERK………………………………………………………………………………77 Discussion……………………………………………………………………………..81

V. IDENTIFICATION OF THE MINIMAL ACTIVE ELEMENT OF THE SYNTHETIC D-AMINO ACID PEPTIDE HYD1 (kikmviswkg)………………………………………………………………………85 Introduction……………………………………………………………………………85 Results…………………………………………………………………………………86 The Minimal Element that Mediates Cell Ahesion to Immobilized HYD1 is Xkmvixw…………………………………………………………………………86 Analysis of the Minimal Element of HYD1 Necessary for Activation of ERK…...91 Analysis of the Minimal Element of HYD1 Necessary to Block Cell Migration on Laminin-5……………………………………………………………………..94 Discussion……………………………………………………………………………..97

8

TABLE OF CONTENTS – CONTINUED –

VI. PRELIMINARY DATA: EFFECT OF THE SYNTHETIC D-AMINO ACID PEPTIDE, HYD1 (kikmviswkg), ON INTEGRIN LATERAL-ASSOCIATIONS……………………………....100 Introduction……………………………………………………………………….....100 Results…………………………………………………………………………….....102 HYD1 Interacts with CD151/Integrin Complexes………………………………..102 HYD1 Increases CD81 Association with the α6 Integrin………………………...102 HYD1 Alters the Lateral-Associations of α6 and α3-Integrins and the Tetraspanin CD81……………………………………………………………………………105 Discussion……………………………………………………………………………105

VII. CONCLUDING STATEMENTS………………………………………..111

REFERENCES……………………………………………………………………..120

9

LIST OF FIGURES Figure 1: Schematic of two-dimensional cell migration……………………………….22

Figure 2: Schematic of integrin-activated focal adhesion signaling……………………25

Figure 3: Isolation of tumor cell adhesion peptides using a one-bead-one-compound

combinatorial library……………………………………………………………………..30

Figure 4: Fluorescence-activated cell sorting analysis of biotinylated peptides bound to

DU-H prostate carcinoma cells…………………………………………………………..47

Figure 5: Confocal microscopy of the peptides RZ-3 and HYD1 binding to the surface

of human prostate tumor cells……………………………………………………………48

Figure 6: Comparison of human prostate tumor cell adhesion to immobilized peptides or

ECM proteins…………………………………………………………………………….50

Figure 7: Effect of antibodies and reagents on DU-H cell adhesion to peptides and

extracellular matrix proteins……………………………………………………………..52

Figure 8: Inhibition of DU145 cell adhesion to immobilized β1 integrin antibody by

peptides…………………………………………………………………………………..54

Figure 9: Inhibition of DU-H cell adhesion to extracellular matrix proteins

by peptides……………………………………………………………………………….55

Figure 10: Inhibition of SCC-25 cell adhesion to HFF monolayers by peptides………57

Figure 11: Adhesion to immobilized HYD1 prevents cell spreading…………………..59

Figure 12: Suppression of adhesion dependent phosphorylation by immobilized

HYD1…………………………………………………………………………………….60

Figure 13: HYD1 blocks cell migration on laminin-5………………………………….67

10 LIST OF FIGURES – CONTINUED – Figure 14: Soluble HYD1 does not effect cell spreading on immobilized laminin-5…70

Figure 15: HYD1 inhibits laminin-5 dependent cellular invasion……………………..71

Figure 16: HYD1 induces cytoskeletal reorganization on laminin-5…………………..73

Figure 17: HYD1 induces phosphorylation of cortactin and alters membrane actin

dynamics…………………………………………………………………………………74

Figure 18: HYD1 interacts with α6 and α3 integrins…………………………………..76

Figure 19: HYD1 activates integrin-associated signaling……………………………...78

Figure 20: HYD1 temporally enhances activation of FAK and phosphorylation of MEK

on laminin-5……………………………………………………………………………...79

Figure 21: HYD1 enhances Laminin-5 signaling and transiently activates ERK……...80

Figure 22: Cell adhesion to immobilized HYD1 is mediated by N- and C-terminal

regions of the peptide…………………………………………………………………….87

Figure 23: The minimal sequence mediating cell adhesion to immobilized HYD1 is

ikmviswk…………………………………………………………………………………89

Figure 24: The minimal element mediating cell adhesion to immobilized HYD1 is

xkmvixw…………………………………………………………………………………90

Figure 25: xikmviswkx is the minimal element of HYD1 required to activate ERK…..92

Figure 26: HYD1 induced ERK activation is mediated by N- and C-terminal regions of

the peptide………………………………………………………………………………..93

Figure 27: xikmviswxx is the minimal element necessary to block cell migration on

laminin-5…………………………………………………………………………………95

11 LIST OF FIGURES – CONTINUED – Figure 28: HYD1 inhibition of cell migration on laminin-5 is mediated by N- and C-

terminal regions of the peptide…………………………………………………………..96

Figure 29: HYD1 interacts with CD151/Integrin complexes…………………………103

Figure 30: HYD1 does not change the amount of CD151 associated with α6 or α3-

containing integrins……………………………………………………………………..104

Figure 31: HYD1 increases the amount of CD81 associated with α6-integrins and

decreases the amount of CD81 associated with α3-integrins…………………………..106

Figure 32: HYD1 induces α6 integrin clustering……………………………………..107

Figure 33: HYD1 alters the lateral-associations of α6 and α3-containing integrins and

the tetraspanin CD81……………………………………………………………………108

Figure 34: Potential mechanisms for the anti-migratory activity of HYD1…………..116

12 LIST OF TABLES Table 1: Alanine-substituted and N- and C-terminal truncated peptides of HYD1 with

their cell adhesion, cell signaling, and anti-migratory activity…………………………..98

13 ABSTRACT

Human tumor cell progression and metastasis is partially dependent on the ability

of tumor cells to adhere to the proteins of the extracellular matrix and migrate to distant

locations. Using a combinatorial screening approach, six novel D-amino acid containing

peptides were identified and analyzed for their ability to adhere to human prostate tumor

cells, support tumor cell adhesion and inhibit tumor cell adhesion to ECM proteins. Two

peptides, RZ-3 (kmviywkag) and HYD1 (kikmviswkg) bound to tumor cell surfaces. A

scrambled peptide derivative of HYD1, HYDS (wiksmkivkg) is not active. As

immobilized ligands, RZ-3 and HYD1 can support prostate tumor cell adhesion. Prostate

tumor cell adhesion to immobilized RZ-3 and HYD1 is integrin dependent. Soluble RZ-3

and HYD1 inhibits tumor cell adhesion to extracellular matrix proteins in a concentration

dependent manner. These results indicate that RZ-3 and HYD1 are biologically active D-

amino acid containing peptides that can support tumor cell adhesion and can inhibit

tumor cell adhesion to immobilized extracellular matrix proteins.

Cell migration is dependent on adhesive interactions with the extracelluar matrix.

These interactions induce signaling and cytoskeletal responses necessary for migration.

HYD1 completely blocks random haptotactic migration and inhibits invasion of prostate

carcinoma cells on laminin-5. This effect is adhesion independent and reversible. The

inhibition of migration by HYD1 involves a dramatic remodeling of the actin

cytoskeleton resulting in increased stress fiber formation and actin colocalization with

cortactin at the cell membrane. HYD1 interacts with α6 and α3 integrin subunits and

elevates laminin-5 dependent intracellular signals including focal adhesion kinase,

14 mitogen activated protein kinase kinase, and extracellular signal-regulated kinase. The

scrambled derivative of HYD1, HYDS, does not interact with the α6 or α3 integrin

subunits and is not biologically active. The minimal element for bioactivity of HYD1

was determined using alanine-substituted analogs of HYD1 and N- and C-terminal

deletion mutants of HYD1. The minimal element necessary to block cell migration on

laminin-5 and activate cell signaling through ERK is xikmviswxx. Taken together, these

results indicate that HYD1 is a biologically active integrin-targeting peptide that

reversibly inhibits tumor cell migration on laminin-5 and uncouples phosphotyrosine

signaling from cytoskeletal dependent migration.

15 I. INTRODUCTION

Cancer Metastasis

Metastasis is the result of the spread of cancer from the primary tumor site to a

distant site or organ. The process of metastasis consists of several steps in order for a

tumor to achieve successful growth at a distant site (Reviewed in [1-5]). First, a tumor

must be able to support its metabolic needs during progressive growth through

angiogenesis [6]. After vascularization, tumor cells may escape into the blood supply

through these new vessels or by impeding into vessels in the vicinity. In order to escape

into the vasculature or lymphatics, tumor cells must also invade through normal

surrounding tissue and vascular endothelium. Once in the blood supply they must

survive in the circulation and arrest in capillary beds at distant organs. Extravasation into

a distant site requires proper adhesive interactions as well as the ability to invade into

normal tissue. Finally, these tumor cells must be adaptive to progressively grow in a new

environment. The multiple events necessary for the development of metastasis

foreshadow its molecular complexity.

Considering cancer mortality, most of the deaths are a direct cause of the

consequences of metastatic tumors rather than the primary tumor itself [1,7]. There is an

intense need for new therapeutics to target the processes involved in metastasis given that

cancer diagnosis often occurs at a relatively late stage in cancer progression. In order to

develop novel therapeutics, further research is needed to identify the critical molecular

components involved in metastasis. Although metastasis is a complicated in vivo

16 process, it is valuable to initially examine the molecular players at the cellular level in

vitro. In general, study of the machinery responsible for cellular motility can lead to the

important discovery of key molecular players that regulate cell migration and/or invasion.

Some of these molecules may prove to be appropriate anti-metastatic targets.

Prostate Cancer

Adenocarcinoma of the prostate is the second most common cause of cancer

deaths in the United States with a projection of 30,350 deaths in 2005 [8,9]. New

diagnoses in 2005 will reach 232,090, making this type of cancer the most common non-

cutaneous malignancy in American men [8,9]. The problem of prostate cancer also exists

outside America. According to the World Health Organization, there were 679,023 new

diagnoses and 221,002 deaths due to prostate cancer worldwide in 2002 [10].

Undoubtedly, the high incidence of prostate cancer is directly related to improved

prostate cancer screening such as the use of prostate specific antigen (PSA) as a

molecular marker for cancer progression and recurrence [11]. In addition, improved

prostate screening and detection may have had a direct effect on the mortality rates given

that in the early 1990s there were approximately 40,000 deaths per year; this figure

dropped to 29,000 in 2004 [8]. Nevertheless, a staggering figure from the American

Cancer Society reveals that the five-year survival rate for localized or regionally detected

prostate cancer is nearly 100%. However, the five-year survival rate drops to 33.5% for

prostate cancer at a distant stage at the time of diagnosis [8]. Distant stage is defined as

cancer that has spread to parts of the body remote from the primary tumor, either by

17 direct extension or by metastasis to distant organs. Therefore, it is clear that this type of

cancer is in need of new therapies to target and ablate its ability to metastasize.

The clinical history of prostate cancer progression indicates that it is an indolent

disease. Without initial treatment, prostate cancer diagnosed at an early clinical stage

will cause relatively few deaths within the first 10-15 years following diagnosis [12-14].

This is most likely related to a small rate of progression to metatstatic disease during that

period. However, a study has reported that additional follow-up at 15-20 years post-

diagnosis significantly changes disease-specific survival [15]. This particular cohort

reports a 3-fold increase in the prostate specific mortality rate compared with the first 15

years of follow-up. Therefore, early stage prostate cancer has a large period of latency

before metastatic conversion.

The indolent nature of prostate cancer has important implications regarding

therapy. First, it is not a highly proliferative cancer and therefore will not be sensitive to

cytotoxic agents used in other proliferative tumors. Second, as previously stated, survival

rates are linked to a loss of organ-confined disease and development of distant metastasis.

Therefore, non-cytotoxic agents that modify the mechanisms of cell invasion and motility

can be useful at preventing the spread of localized disease. Identifying key molecules

involved in prostate cancer invasion and metastasis should be a priority for novel

therapeutics.

Much work has been focused on characterization of both genetic and protein

changes that take place during prostate cancer progression. Chromosomal instability is a

hallmark of cancer and accordingly many genetic alterations have been discovered and

18 described for prostate cancer. All of these changes are out of the scope of this

dissertation. Although not exhaustive, the major findings can be found in several reviews

[16-19]. Alterations at the genetic and protein level act as molecular clues to the etiology

of prostate cancer as well as providing a foundation to understand its progression to

metastatic disease.

Integrins are cell adhesion molecules that play a crucial role in not only cell

adhesion, but also dictate multiple aspects of cell behavior including cell motility and

cellular metastasis [20-22]. Changes that occur in integrin expression during prostate

cancer progression seem to be early events [16,23,24]. Complete loss of specific alpha

and beta subunits have been described and include α5, α4, αv, β1C, and β4 [23,25,26].

Coupled with these findings, conservation of the integrins α6β1 and α3β1 have been

documented [23,24], making these laminin receptors critical molecular components and

targets encompassing prostate cancer metastasis. The role of alpha-6 integrins has been

solidified by the findings that α6β1 maintains expression in prostate micrometastases

[27]. Teleologically, the retention of these receptors correlates with the finding that

prostate tumors often invade out of the capsule along nerve-endings and exit into the

laminin-rich perineural space [28-31]. Taken together, these findings warrant further

study of the biology and functional role of α6 and α3-containing integrins in prostate

cancer progression. Specifically, how do these integrins functionally regulate tumor cell

invasion and metastasis?

19 Integrins and Cell Motility

Integrins are evolutionarily conserved heterodimeric cell surface molecules [32-

34]. Structurally, they contain an alpha and beta subunit, each consisting of a large

extracellular domain, a single-pass transmembrane domain, and a short cytoplasmic tail

with the exception of the β4 subunit, which has a large cytoplasmic domain [32,35]. To

date there are 18 distinct α and 8 distinct β subunits that pair in a restrictive manner to

give about 24 different integrins that have individual ligand binding specificities and

nonredundant functions given their phenotypes in knockout mice [32,35,36]. The main

function of integrins is to mediate and regulate cell adhesion to extracellular matrix

proteins. In addition to cell adhesion, integrins affect many aspects of cell behavior

including cell survival, proliferation, motility, and cytoskeletal reorganization. These

roles in cell biology mediate the impact of integrins on biological processes including

hemostasis, immunity, inflammation, and cancer metastasis [32-34]. The ability to act as

signal transducers is central to their multitude of functions. Their effect on cell signaling

is directly related to the ability to form mechanical links from the extracellular matrix

(ECM) to the cytoskeleton. Through these links integrins support the formation of

membrane proximal adhesion complexes that contain a vast array of different proteins

that are involved in cytoskeletal structure and signaling [32,37,38].

Given the complexities of their function, integrin activation must be tightly

regulated and much work has focused on uncovering the mechanisms of integrin

activation. Integrin adhesion and activation is rapidly regulated by changes in affinity for

extracellular matrix ligands [33]. Mutational studies and the crystal structure of the αvβ3

20 [39] suggests that affinity modulation is linked through a conformational change that

spans the integrin protein. In support of this, it has been demonstrated that a conserved

GFFKR sequence in the cytoplasmic domain of the α subunit is a negative regulator of

activation [40]. A critical affinity-independent mechanism of activation is integrin

clustering [33,34]. When integrins bind to the ECM they become clustered in the plane

of the membrane and recruit cytoskeletal and signaling elements that promote the

formation and reorganization of actin filaments. This process is thought to be dependent

on the action of the Rho family of GTPases and lateral associating proteins that interact

with integrins such as caveolin-1 or tetraspanin molecules [34,41,42]. Importantly, this

process of activation can be induced by binding to ECM proteins, termed “outside-in”

signaling or by response to soluble or internal processes, termed “inside-out” signaling.

Both result in the adhesive and signaling responses that elicit integrin function [32]. Cell migration is a complex process coordinating the actions of many different

cellular players. The adhesive complexes formed from integrin ligation and activation

serve as signaling regulators of migratory processes as well as traction points over which

the body of the cell moves [43]. Work has shown both in vitro and in vivo that integrins

influence the ability of a malignant cell to invade and metastasize [44]. As an example,

use of a anti-α6 antibody co-injected into nude mice with invasive human melanoma

cells will reduce metastasis of the xenograft [45].

21 Protrusive and Contractile Contacts

The process of migration is complicated, requiring the dynamics of many

processes. In general, cell migration is a reiterative process that requires extensive

cortical actin reorganization including lamellipodia and filopodia extension [43,46,47],

integrin dependent adhesion leading to the formation of focal complexes, and maturation

of focal complexes into focal adhesions [46,48]. These adhesions then form a platform

for traction over which the cell body can move [43]. With the description just given, two

types of contacts can be described that regulate this process, protrusive and contractile.

Protrusive contacts are membrane extensions formed by cells to examine the surrounding

environment. Cellular protrusions at the leading-edge of the cell form adhesive contacts

with the extracellular matrix and polarize the cell for migration [37]. These contacts can

be any type of extension ranging from very defined, localized extensions such as

filopodia or microspikes, to very broad protrusions such as lamellipodia or membrane

ruffles. Contractile contacts are those involved in stable, prolonged attachments such as

those displayed by focal adhesions [37]. These types of contacts apply tension to the cell,

showing that the adhesions formed are stable and worthy of use as a migration surface

[49]. Figure 1 is an overview of the important events necessary for two-dimensional cell

migration. Dysregulation of the migration machinery during cancer progression may

create hypermotile tumor cells that have an increased capacity to invade and metastasize.

22

Figure 1. Schematic of two-dimensional cell migration. This figure illustrates important components at both the leading edge and the retracting end of a migrating cell on a 2-dimensional, rigid surface.

23

Assembly of protrusive contacts are influenced by two members of the Rho

family of GTPases. Specifically, Rac1 and Cdc42 act as signal transducers to form

lamellipodia and filopodia respectively [43,47,48,50]. Activation of these GTPases can

take place from “outside-in” or “inside-out” signaling and are reliant upon guanine

nucleotide exchange factors for their activation [47]. There is accumulating evidence

suggesting that tips of lamellipodia and filopodia serve as docking sites to localize factors

that contribute to actin polymerization required for cell motility. A good example is

highlighted by a study illustrating that the amount of vasodilator-stimulated

phosophoprotein (VASP) in lamellipodium tips increased directly with protrusion rate

[51]. The molecular players involved in initiating actin polymerization are starting to be

characterized and the Arp 2/3 complex has been strongly implicated in nucleating and

structuring actin networks [52,53]. In addition, it has been shown that a member of the

Wiskott-Aldrich syndrome protein family, Scar/Wave, is integral in activating Arp 2/3 in

lamellipodium formation [54]. Further work is warranted to determine the additional

players that mediate actin polymerization at the tips of protrusive contacts. The small GTPase Rho is a major player in contractile contacts through its role in

regulating the organization of actin stress fibers. Additionally, Rho activation directly

correlates to the formation of focal adhesions [43,48]. As described above, contractile

contacts are important in cell motility by providing traction over which the cell moves.

The molecular mechanism in how Rho regulates this process is well characterized. Rho

can activate Rho kinase, which phosphorylates and inhibits myosin light chain

phosphatase [55]. This allows myosin light chain to be in a highly phosphorylated, pro-

24 contractile state, contributing to integrin clustering and stress fiber formation. This is one

characterized mechanism, however, given the multiplicity of Rho targets, there are

undoubtedly many more molecular players regulating this process.

Focal Adhesion Proteins: The Role of Focal Adhesion Kinase and the Extracellular

Signal-Regulated Protein Kinase/Mitogen-Activated Protein Kinase Pathway in Cell

Migration

As previously mentioned, integrin activation results in recruitment of many

structural and signaling proteins to the cell membrane at the site of integrin/ECM contact

[56]. These protein complexes organize into focal adhesions that physically link

integrins to the actin cytoskeleton and are capable of integrating external cues into

cellular signals that regulate cell behavior. The dynamic regulation in the formation and

disassembly of focal adhesions are crucial to cell motility, [56] and often the components

of the adhesion complex directly regulate its dynamic. Because of the abundance of

proteins involved, this section will focus on the role of focal adhesion kinase (FAK) and

the mitogen-activated protein kinase pathway in cell migration events. Figure 2 is a

schematic that displays a few important cytoskeletal and signaling proteins incorporated

into focal adhesions and their downstream effects on motility. A prominent biochemical change that occurs with integrin clustering is tyrosine

phosphorylation of several proteins, including FAK [57]. FAK is a 125 kD protein that is

a member of non-receptor protein kinases. Integrin clustering leads to rapid recruitment

25

Figure 2. Schematic of integrin-acitvated focal adhesion signaling. Upon interaction with the extracellular matrix (ECM), integrins become activated and recruit multiple structural and signaling proteins to the cell membrane. These proteins compose the structural integrity of the adhesion site and serve as signal-transducers to modulate many aspects of cell behavior including motility. This figure displays a few of the proteins linked to cell migration discussed in the introduction. PP2 and Y-27632 are chemical inhibitors of Src and ROCK respectively that were used in this study.

26 of FAK to focal contact sites [46]. FAK has several phosphorylation sites. In particular

phosphorylation on tyrosine 397, an autophosphorylation site, correlates with increased

kinase activity [58], thus providing a marker for FAK activation. FAK is implicated in

migratory processes by several lines of evidence. First, FAK deficient fibroblasts migrate

poorly in response to haptotactic or chemotactic cues while forming prominent focal

adhesions by vinculin staining [46,59,60]. In addition, FAK overexpression increases

fibronectin stimulated motility only if tyrosine 397 site is kept intact [61]. Finally, FAK

tyrosine phosphorylation has been shown to be critical for cell spreading and migration of

endothelial cells on fibronectin [62]. These data indicate that FAK has both signaling

and focal adhesion remodeling functions that are crucial for cell migratory processes.

Like FAK, the extracellular signal-regulated kinase/mitogen-activated protein

kinase pathway (ERK/MAPK) has a role in cell migration through both signaling and

focal adhesion remodeling activities [56]. It has been shown that the ERK/MAPK

activity stimulates myosin light chain kinase (MLCK) activation, which phosphorylates

myosin light chain (MLC). Phosphorylation of MLCs initiates myosin dimerization and

engagement with the actin cytoskeleton producing cellular contraction [63,64]. In

addition, activation of the ERK/MAPK pathway, in cooperation with Src, may negatively

regulate the Rho-ROCK-LIMK pathway and thus alters actin dynamics [65]. The focal

adhesion remodeling activities are thought to be directed by interplay with the protease

calpain 2 [56]. Specifically, chemotactic-induced detachment from the matrix during cell

motility required calpain 2 at the cell membrane. The proper localization of this protease

was dependent on ERK/MAPK signaling [66], and furthermore, ERK can directly

27 regulate calpain activity through phosphorylation [67]. Finally, calpain activity is

attenuated by inhibition of kinase components of the MAPK pathway [68]. Taken

together, the ERK/MAPK pathway influences cell motility through its ability to regulate

cytoskeletal dynamics and focal adhesion proteolysis.

Isolation of Tumor Cell Adhesion Peptides Using a One-Bead-One-Compound

Combinatorial Library

The retention of α6β1 and α3β1 integrins in prostate cancer progression [23,24]

foreshadows their importance in prostate cancer metastasis, marking them as potential

therapeutic targets. The field of combinatorial peptide chemistry has enabled scientists to

create peptide ligand mimetics to multiple protein targets including kinases and cell

surface receptors [reviewed in [69]]. Two particularly useful methods to isolate

candidate peptide ligands for cell surface receptors are phage-display and the one-bead-

one-compound (OBOC) approach. Isolation and characterization of these peptides not

only creates potential inhibitors, it also fosters increased understanding of protein biology

and function. This section will briefly compare the phage-display method to the OBOC

approach and highlight the methodology behind the one-bead-one-compound

combinatorial library for the development of tumor cell adhesion peptides.

Combinatorial peptide chemistry allows the screening for peptide ligands among

billions of random peptides created through a variety of approaches. The one-bead-one-

compound combinatorial library method [70,71] and the phage-display peptide library

approach [72-75] have been successfully used to identify peptide ligands for cell surface

28 molecules. Specifically, the phage display method and the OBOC approach has

identified peptides that interact with the surface idiotypes of B-cell lymphoma and human

chronic lymphocytic lymphoma [76-78]. Both of these approaches have also been

efficient at identifying ligands that interact with integrins [79-84]. Although each

technique is effective, they each have specific advantages and disadvantages.

Phage-display is a biological library method that manipulates filamentous phage

to express peptides on specific surface proteins such as the viral pIII coat [85]. Random

deoxyoligonuleotides dictating peptide size and orientation are inserted directly into the

pIII gene creating a distinct peptide on the surface of individual phage [85]. Panning for

peptides with specificity to a cell surface protein can be performed using purified cell

surface receptors [86] or live cells [87,88]. This technique is advantageous because up to

109 peptides can be screened per library while there is no restriction on the size of peptide

displayed.

The one-bead-one-compound combinatorial method uses a solid phase technique

and a “split-mix” synthesis approach to create a peptide library [70,89,90]. In essence,

individual peptides are synthesized on polyethylene glycol-grafted polystyrene beads that

yield approximately 1013 copies of the same peptide per bead [70,71]. Like the phage

display method, OBOC libraries can identify cell surface ligands using purified receptors

[71] or intact cells [81]. Some specific advantages of using the OBOC approach

compared to phage display are that peptides from the OBOC method are spatially

separable, meaning that multiple motifs can easily be identified from isolation and

characterization of specific beads. Most importantly, the OBOC method is not limited to

29 the use of L-amino acids as in phage display. D-amino acids, unnatural amino acids or

even nonamino acid moieties can be used in OBOC libraries. Peptides created for

example, from D-amino acids are much more resistant to proteolysis in vivo [91,92] and

are therefore more desirable for therapeutic purposes.

We have used the OBOC peptide library method to identify peptide sequences

that support tumor cell adhesion via the alpha-6 integrin [81,82]. The method of

selection was based on functional cell adhesion of live tumor cells to peptide-containing

beads. Briefly, DU-145H cells, a prostate tumor cell line that expresses high levels of α6

[93], was applied to OBOC libraries and analyzed under a dissecting microscope. Beads

with cells attached were retrieved, and those beads were rescreened with the same tumor

cell line in the presence of a functional-blocking antibody to α6, GoH3. The beads that

no longer bound to tumor cells in this condition contained candidate α6-specific adhesion

peptides. Those beads were isolated, retested for adhesion without the antibody, and

sequenced. Figure 3 displays the strategy for the selection of a cell adhesion peptide

using the OBOC method. This method is particularly powerful because it does not

require purification of the receptor and thus allows the appropriate post-translational

modifications. In addition, by using live cells in this scheme, peptides can be identified

that interact with conformationally sensitive surface molecules. In principle, this method

allows the identification of specific cell adhesion peptides for any cell surface receptor

for which a function-blocking reagent is available.

30

Figure 3. Isolation of tumor cell adhesion peptides using a one-bead-one-compound combinatorial library. This selection scheme was used to isolate and characterize peptides that interact with α6 integrins. The function-blocking reagent was GoH3, a function-blocking antibody to α6.

31 II. MATERIALS AND METHODS

Affinity Precipitation with Peptides

1.2 x108 DU-145H cells were harvested with 2mM EDTA in PBS. Cells were

washed in AP buffer containing 25 mM HEPES pH 8.0, 100 mM KCl, 1 mM MgCL, 1

mM CaCl, 20% glycerol, 1 mM PMSF, 1 µg/ml leupeptin and aprotinin. Cells were lysed

using a dounce homogenizer then centrifuged for 5 minutes at 15,000 g at 4oC in an

Eppendorf microcentrifuge. Supernatants were further centrifuged for 15 minutes at

96,000 g at 4oC in a Sorvall RC M150GX ultracentrifuge. Membrane fraction pellets

were suspended in AP buffer containing 0.2% NP-40 and analyzed for protein

concentration using the BCA protein assay kit (Pierce, Rockford, IL). 500 µg of

biotinylated peptide was preloaded on to 30 µl of UltraLink Immobilized NeutrAvidin

Plus beads (Pierce, Rockford, IL) in a buffer containing 0.5mM KCl, 0.3mM KH2PO4,

27.6mM NaCl, and 1.6mM Na2HPO4 pH 7.4 for 1 hour at room temperature. The

resulting beads were washed twice with AP buffer and incubated with 30µg of the

membrane fraction adjusted to a total volume of 500 µl for 18 hours at 4oC. NeutrAvidin

beads were washed 3 times with AP buffer containing 0.2% NP-40. All samples were

suspended in 2x laemmli buffer and analyzed by SDS-PAGE electrophoresis.

32 Antibodies and Reagents

GoH3, a rat anti-α6 functional-blocking antibody was purchased from Serotech

Inc. (Raleigh, NC). The anti-α3 functional-blocking antibody, P1B5, was purchased

from Chemicon (Temecula, CA). AIIB2, a rat anti-β1 functional-blocking antibody, was

purified from its hybridoma (Developmental Studies Hybridoma Bank, University of

Iowa). Integrin antibodies used for western blotting: Anti-α6, AA6A rabbit polyclonal,

was generated and purified by Bethyl Laboratories Inc. (Montgomery, TX). Anti-α3,

AB1920, was purchased from Chemicon (Temecula, CA.). Antibodies to FAK, Tyr-397

and 4.47, in addition to phospho-Mek (serine 298) and cortactin (clone 4F11) were

purchased from Upstate (Lake Placid, NY). MEK1 antibody was obtained from BD

Transduction Laboratories (San Diego, CA). Map Kinase antibodies, phospho-p44/42

(Thr202/Tyr204) and total p44/42, were purchased from Cell Signaling Technology Inc.

(Beverly, MA). CD151 monoclonal antibody, 5C11, was a gift from Dr. Martin Hemler

(Dana-Farber Cancer Institute and Department of Pathology, Harvard Medical School,

Boston, MA.). CD81 monoclonal antibody, JS64, was purchased from Immunotech

(France). Inhibitors for Src (PP2) and ROCK (Y-27632) were purchased from

Calbiochem (San Diego, CA). Anti-mouse and anti-rabbit secondary antibodies

conjugated to horseradish peroxidase for immunoblotting were obtained from Chemicon

(Temecula, CA). Anti-mouse secondary antibody conjugated to Alexa Fluor 568 and

phalloidin conjugated to Alexa Fluor 488, used in immunofluorescence, were obtained

through Molecular Probes (Eugene, OR).

33

The mouse monoclonal antibodies used for the adhesion blocking studies were as

follows: P4C10, anti-β1 integrin; P1E6, anti-α2 integrin; P1B5, anti-α3 integrin; and

P1D6, anti-α5 integrin (all from Life Technologies Inc.). Mouse preimmune IgG was

purchased from ICN Biomedicals Inc. (Aurora, OH). The β1 antibody used for in the

adhesion assay was TS2/16 obtained from the American Type Culture Collection.

Biotinylation of Cell Surface Proteins

Cells were adhered to laminin-5 coated tissue culture plates for 1 hour prior to

biotinylation. Cells were washed twice in HBSM (20mM Hepes, pH 7.5, 150mM NaCl,

and 5mM MgCl2 ) followed by biotinylation with 0.2 mg/ml sulfo-NHS-LC biotin

(Pierce Chemical Co) in HBSM for 1 hour at room temperature. Cells were rinsed three

times with HBSM prior to lysis.

Cell Lines and Culture Conditions

All cell lines were incubated at 37o C in a humidified atmosphere of 95% air and

5% CO2. The human prostate carcinoma cell lines, PC3N and DU-145H, were grown in

Iscove’s Modified Dulbecco’s Medium (Gibco BRL, Gaithersburg, MD.) plus 10% fetal

bovine serum (Gibco BRL). All medium was supplemented with penicillin/streptomycin,

100 units/ml (Gibco BRL). Serum-Free medium was supplemented with 0.1% Bovine

Serum Albumin (Sigma, St. Louis, MO.). PC3N cells are a variant of the human PC3

prostate carcinoma cell line [94] . DU-145H (DU-H) cells are DU-145 cells, a prostate

carcinoma cell line originating from a brain metastasis, selected for high expression of α6

34 integrin [93]. DU-145 and PC3 cell lines are available from ATCC (Rockville, MD).

HaCaT cells [95] were obtained from Dr. Norbert E. Fusenig (German Cancer Research

Center, University of Heidelberg, Heidelberg, Germany). The human oral squamous cell

carcinoma line, SCC-25, was obtained from American Type Culture Collection

(Rockville, MD). Human foreskin fibroblasts (HFF) were obtained as previously

described [96]. SCC-25 and HFF cells were maintained in Ham's F:12/DMEM

purchased from Gibco BRL (Grand Island, NY) supplemented with 10% fetal bovine

serum from Gemini-Bio-Products (Calabasas, CA), 0.1% hydrocortisone and penicillin

(100units/ml)/streptomycin (100units/ml).

Cell Motility Assays

Both time-lapse video microscopy and modified Boyden chamber transwell

assays were performed. Time-lapse video microscopy is offered as a core service

through the Southwest Environmental Health Services Center Cellular Imaging Core.

Time-lapse video microscopy was used to analyze random haptotaxis of PC3N cells on

laminin-5. The effect of the peptides or pharmacological inhibitors was observed with

differential interference contrast optics on an inverted Olympus IMT2 microscope

(Olympus America, Melville NY). The microscope was equipped with a BiopTechs

Delta T live Cell system (BiopTechs, Butler PA) with a humidified 5/95% CO2/Air

atmosphere and was maintained at 37oC. Images were obtained with a grayscale CCD

camera (ORCA-100, Hamamatsu, Japan) and analyzed using Compix imaging software

(SimplePCI 4.0, Compix Imaging, Cranberry Township PA). 70,000 cells were plated

35 for 1 hour in serum-free conditions on laminin-5 coated Delta T dishes (0.15 mm,

BiopTechs, Butler PA). Coating was done as described above. Soluble peptides or

chemical inhibitors in serum-free media at the stated concentrations were added to the

adhered cells and video was started at approximately 30 min post peptide or inhibitor

treatment. For peptide reversibility experiments, cells were adhered and treated as above

but after 30 minutes of treatment with peptide the cells were washed 1 time with PBS and

serum-free media or soluble laminin-5 was added back. Videos were four hours in length

with a frame capture every 5 minutes. The speed of each cell was calculated manually

with the Compix software by measuring the distance traveled by the cell nuclei over the 4

hour time period. The average rate of migration (15 cells/experiment) in microns was

calculated. Each experiment was performed at least three times.

Analysis of invasion was performed using a modified Boyden chamber assay.

Falcon polyethylene terephthalate (PET) track-etched membrane 8µm pore inserts

(Becton Dickinson Labware, Franklin Lakes NJ) were coated on the underside with

laminin-5 (HaCaT conditioned media) for two hours at room tempurature and were

washed 1 time with HEPES buffer (20mM HEPES, 130mM NaCl, 5mM KCl, 0.8mM

MgCl2, 1mM CaCl2, pH 7.4). Membranes were blocked with 1% BSA in PBS for 30

minutes at room temperature and were washed 1 time with HEPES buffer. 50,000 cells

were seeded in the upper chamber in serum free media and allowed to attach for 1 hour

followed by the indicated treatments with peptide or chemical inhibitor. Peptides and

inhibitors were in serum-free media and in both upper and lower chambers. For

experiments with integrin blocking antibodies, cells were pretreated in suspension with

36 the indicated antibody(s) for 15 min before attachment. Cells were allowed to invade for

18 hours at 37o C and cells remaining in the upper well were removed with a cotton swab

while cells on the lower surface were fixed with methanol and stained with crystal violet.

The number of cells on the lower surface of the insert was quantified by solubilizing the

dye in 0.1 M sodium citrate and reading the absorbance at 562 nm. Triplicate

determinations were done with each treatment and each experiment was performed at

least 3 times.

The scratch assay was performed with PC3N cells grown to confluency on

laminin-5 coated square coverslips in IMDM (Gibco BRL, MD, USA) plus 10% fetal

bovine serum (FBS). The source of laminin-5 was serum-free conditioned medium from

HaCaT cells. A scratch was made diagonally across the square coverslip with a

plastic cell scraper (Fisherbrand Cat. # 08-773-2). The coverslips were then

rinsed in medium and placed in fresh medium containing 75 µg/ml peptide and 1% FBS

for 12 hours at 37oC. The cells were fixed with chilled methanol for 10 min

followed by 10 dips in chilled acetone. On drying, the cells were then stained

with 0.5 µg/ml DAPI for 10 min. The coverslips were washed in PBS, post-fixed

in Ethanol for 4 min and mounted using Prolong Antifade (Molecular Probes, Or,

USA). The slides were visualized on a Zeiss Axiovert microscope. Pictures were

taken using Axiocam camera at 10 X magnification. Quantification was done using

Scion Image.

37 Cell Adhesion Assay and Peptide Inhibition of Adhesion.

Ten µg of ligand or 20 µg of neuralite avidin (Molecular Probes, Inc.) was

dissolved in 1 ml of distilled water and 100 µl of solution was added to each well of a

tissue culture 96 multiwell plate (Falcon, Franklin Lakes, NJ). The solution of either

ECM protein or neuralite avidin was allowed to dry overnight. Use of neuralite avidin

ensures maximum binding of the biotinylated peptide to the surface, avoiding the

variability of peptide coating. In the β1 antibody adhesion assay, the TS2/16 antibody

(2µg/ml) was coated for 18 hours in the HEPES buffer at 4C. The wells for all substrates

were then blocked with 100 µl of 1% bovine serum albumin (BSA) for 1 hour. The wells

were washed with HEPES buffer and varying amounts of peptide added per well. After

one hour of peptide incubation, wells were washed with IDMEM without serum and

suspended DU-H cells (5 x 104) in serum free IDMEM were added in each well. The

cells were allowed to adhere for 60 minutes at 37C. The wells were washed three times

with HEPES buffer and fixed with 2.5% formaldehyde in PBS. The cells were then

stained with 0.5% crystal violet in 20% (v/v) methanol/water and viewed under a

microscope. The amount of bound cells was estimated by solubilizing the dye using

0.1M sodium citrate and reading the absorbance at 570nm. Triplicate determinations

were done at each data point.

Cell Lysis and Western-Blot

For cell signaling experiments involving FAK, MEK and ERK, cells were

incubated in serum-free media overnight and harvested with 5mM EDTA in PBS for 10

38 minutes. Cells were washed in serum-free media and added to laminin-5 coated plates or

BSA coated plates for 1 hour at 37o C. Cells were then treated with peptide in serum-free

media for stated times at 37o C. Two minutes before lysis, 0.5mM sodium orthovanadate

was added into the media. Cells were lysed for 5 minutes in a modified RIPA lysis buffer

containing 40mM Tris, pH 7.5, 150mM NaCl, 1% Triton X-100, 6mM EDTA, 100mM

NaF, 1mM sodium orthovanadate, 10mM sodium pyrophosphate, 1mM PMSF, and

1µg/ml leupeptin and aprotinin. Lysates were processed for SDS-PAGE after adjusting

for equal loading by using the BCA protein assay kit (Rockford IL). Proteins resolved in

the gel were electrotransferred to Millipore Immobilon-P polyvinylidene fluoride (PVDF)

membrane (Millipore, Bedford MA). Blots were developed using chemiluminescence

(ECL Western Blotting Detection System, Amersham, Arlington Heights, IL) and band

densities were analyzed by densitometry using NIH Image software (Scion). For

stripping, membranes were incubated in stripping buffer (62.5mM Tris, pH 6.75, 2%

SDS, and 100mM beta-mercaptoethanol) at 50o C for 30 minutes followed by washing 5

times in TBS.

Immunoprecipitation

All immunoprecipitations, unless specified, contained 200 µg total protein lysate.

Immunoprecipitation of cortactin per reaction contained, 200µg total protein lysate, 35 µl

of protein G sepharose and 3 µg of antibody. The final volume was adjusted to 400 µl of

RIPA lysis buffer described above. The mixture was rotated at 4o C for 18 hours and the

complexes were washed 3 times with cold lysis buffer. For co-immunoprecipitation

39 studies, cells were lysed in HBSM ( 20mM Hepes, pH 7.5, 150mM NaCl, and 5mM

MgCl2 ) supplemented with 1% Brij 96 (or 97). These complexes were washed 4 times

with cold lysis buffer following rotation at 4o C for 18 hours. Immunoprecipitation of

CD151 was performed on the supernatants from affinity precipitations described above.

Briefly, the supernatant was added to an immunoprecipitation reaction containing 1 µl of

5C11 antibody and 30 µl of protein G sepharose. The mixture was rotated for 18 hours at

4oC. Additional 5C11 immunoprecipitations were carried out with 1 µl of 5C11, 20 µg

membrane fraction and 30 µl protein G beads adjusted to a total volume of 500 µl with

the standard rotated for 18 hours at 4oC. Protein G sepharose beads were washed 3 times

with RIPA buffer. All samples were suspended in 2x laemmli buffer and analyzed by

SDS-PAGE electrophoresis. When western blot analysis was for α6, non-reducing

laemmli buffer was used.

Epithelial and Stromal Cell Adhesion Assay.

HFF cells were used at 3 x 105 cells/well in 6-well plates in serum free medium

and allowed to attach at 37C. After 24 hours the medium was removed, the well was

washed once with PBS and 3 x 105 SCC-25 cells were placed on top of the HFF

monolayer. The SCC-25 cells were added using 1ml of serum free medium containing

either 40ug/ml HYD-1, 40ug/ml HYDS-1 or no treatment. The number of SCC-25

unable to attach were recovered by a PBS wash and counted every hour for three hours

using a hemocytometer. The determinations were done in triplicate.

40 FACS Analysis and Confocal Microscopy.

The peptides were synthesized with a biotin at the amino terminus. The peptides

were then allowed to bind to neuralite avidin conjugated with Bodipy (Molecular Probes,

Eugene, OR) and the tetrameric peptide-avidin complexes were incubated with the cells.

The non-specific binding was determined using the neuralite avidin-Bodipy alone

incubated with the cells. Suspended DU-H cells (1x106) in 1 ml of IDMEM without

serum were incubated with 10 µg of peptide and 10 µg Bodipy conjugated to neuralite

avidin at 4oC in the dark for 30 minutes. The cells were washed several times with

IDMEM without serum and then analyzed for fluorescence on a FACStarPlus (Becton

Dickenson) or examined using a Laser Scanning Confocal Microscope (LSM 10, Zeiss ).

Digital images were collected on each Z-series using identical contrast and brightness

settings.

In Vivo Actin Distribution Assay

The distribution of globular actin and filamentous actin was analyzed using an F-

actin/G-actin in vivo assay kit (Cytoskeleton Inc., Denver CO). Briefly, after peptide

treatment, cells were lysed in a cell lysis and F-actin stabilization buffer and

homogenized using 27 gauge needles. In order to isolate cellular filamentous actin, cell

lysates were centrifuged at 100,000 g for 60 minutes at 37o C and the supernatants (G-

actin) were immediately separated from the pellets (F-actin). The pellets were

resuspended in the same volume of dH2O as the supernatants and were incubated on ice

for 60 minutes. Cytochalasin D (2µM) was added to the resuspended pellets to promote

41 dissociation of filamentous-actin in the pellets into actin monomers. The pellet was then

further fractioned to insoluble and soluble forms by centrifugation at 14,000 rpm for 1

minute. The insoluble portion containing the cell membrane was solubilized in RIPA

buffer. Equal amounts (2µg of protein for each sample) of each fraction were subjected

to SDS-PAGE and analyzed by Western Blot with an anti-actin antibody provided in the

kit.

Immunofluorescence

Coverslips were coated with laminin-5 as stated in materials and methods and

washed with PBS before plating the cells. Cells were plated subconfluently and allowed

to adhere for one hour before treatments. Peptide was added in serum-free media for 30

minutes before fixation. Coverslips were dipped in PBS and fixed in PBS containing

3.2% formaldehyde (Ted Pella Inc. Irvine CA) for 5 minutes. The coverslips were rinsed

in water before incubation with 50 mM NH4Cl in PBS for 5 minutes. Following a wash

in PBS for 5 minutes cells were treated with 0.2% Triton X-100 in PBS for 5 minutes.

Fixation and washing were performed at room temperature. Cells were washed again in

PBS and blocked with 1% BSA for 30 min at room temperature before incubating with

the primary antibody (cortactin 1:100) and phallodin (1:40) for 1 hour at room

temperature. Staining for alpha-6 (J1B5 1:50) and CD81 (JS64 1:50) followed the same

protocol. Secondary antibody was diluted at 1:200 for 30 minutes at room temperature.

The coverslips were mounted with Anti-Fade (Molecular Probes, Eugene OR) and

observed at 40X with an inverted Zeiss Axiophot (Carl Zeiss, Gottingen, Germany)

42 equipped with an Axiocam camera. Captured images were analyzed with Photoshop 7.0

software (Adobe Systems Inc., San Jose CA).

Synthetic Peptides, Fibronectin, Laminin-1, Laminin-5, and Collagen IV

The RZ series of peptides was found using the “one-bead one-compound”

combinatorial library method and a D-isomer Octyl-mer peptide library as previously

described by us [81]. The amino acid sequences of the D-amino acid peptides are RZ-3,

kmviywkag; RZ-4, kggrhykfg; RZ-6, arkfkglig; RZ-12, yiknrkhhg. The HYD-1 peptide

(kikmviswkg) was generated by overlapping the positive adhesion peptides and

postulating a hybrid sequence. The HYDS-1 scrambled peptide (wiksmkivkg) was

generated by random ordering of the HYD-1 peptide sequence. AG73

(RKRLQVQLSIRT) is a previously characterized L-amino acid peptide from the G-

domain of the laminin α1 chain [97]. The six peptides were synthesized with a biotin at

the amino terminus and purified by Molecular Resources Core Facility, Colorado State

University, Dept. of Biochemistry, Ft. Collins, Colorado. Peptides were also synthesized

and purified by High-Performance-Liquid-Chromatography to greater than 90% by

Global Peptide Service (Fort Collins CO). The human fibronectin and mouse collagen

IV was obtained from Gibco BRL, Inc. The laminin-1 was obtained from Becton

Dickinson Corp., Franklin Lakes, NJ. and is derived from the Engelbreth-Holm-Swarm

(EHS) mouse tumor. The laminin-5 was obtained from the matrix of HaCaT cells[95]

(Dr. Norbert Fusenig, University of Heidelberg).

43

Laminin-5 Coating Conditions for Motility and Signaling Experiments

Laminin-5 was obtained from conditioned media of HaCaT cells. Briefly, sub-

confluent HaCaT cells were incubated with serum-free media overnight. The media was

collected and filtered through a 0.2µm acetate filter. For laminin-5 coating of tissue

culture plastic, coverslips, and time-lapse video plates, conditioned media was added to

the surface for 2 hours at room temperature and washed 1 time with PBS (2.7mM KCl,

1.5mM KH2PO4, 138mM NaCl, 8.1mM Na2HPO4, pH 7.4) before use. For signaling

experiments, plates were blocked with 1% BSA for 30 minutes at room temperature.

44 III. IDENTIFICATION AND CHARACTERIZATION OF TUMOR

CELL ADHESION PEPTIDES

Introduction

Tumor cell adhesion to the extracellular matrix (ECM) within tissues greatly

influences a malignant cell’s ability to invade and metastasize to outlying tissues,

reviewed in [98]. Further, the survival of the metastasized tumor cell depends in part

upon the activity of the ECM receptors [99-104]. Cell adhesion and the accompanying

intermediate filaments in tumor cells are also an important factor in resistance to the

killing effects of several chemotherapeutic drugs. Fibronectin mediated adhesion of

myeloma cells confers a survival advantage in a phenomenon known as cell adhesion

mediated drug resistance (CAM-DR) [105-108]. The pivotal role of adhesion

modulation in both tumor cell metastasis and their survival prompted us to characterize

tumor cell adhesion peptides for their ability to alter cell adhesion to the ECM.

The proteins of the ECM consist of type I and IV collagens, laminins, heparin

sulfate proteoglycan, fibronectin, and other noncollagenous glycoproteins [109]. Cell

adhesion to the laminins, fibronectin, and collagens is mediated in part by a group of

heterodimeric transmembrane proteins called integrins, which are composed of a non-

covalently associated α- and β- subunit that define the integrin ligand specificity [20].

The integrins α6β1, α3β1, and α6β4 are laminin receptors [110,111]. These integrins in

particular are associated with epithelial tumor progression in prostate, breast, colon,

pancreatic carcinomas, head and neck tumors, and melanoma [23,112-115]. Prostate

45 micrometastases continue to express the α6 integrin whereas other cell adhesion

molecule expression is suppressed [27].

Previous attempts to block adhesion of cells to the ECM (i.e. laminin-1, 5 or

collagen) have been accomplished using integrin specific, function blocking monoclonal

antibodies or peptide mimics of the natural ECM protein [116-118]. For example, human

melanoma cells co-injected into nude mice with an anti-α6 antibody have a lowered

ability to metastasize [119]. Similarly, the α1β1 and α2β1 integrins are collagen

receptors and antibodies to these integrins reduced invasion of basement membranes

[120]. Biologically active adhesion peptides derived from specific regions of the ECM

protein laminin-1 have had reported effects on a variety of biological processes including

invasion, metastasis, migration and matrix metalloproteinase expression [97,121-131].

Our previous work identified several L-amino acid containing peptide candidates

as anti-adhesive agents by selecting peptides based upon the ability of the prostate tumor

cells to bind to the immobilized peptide [81]. Further screening with a D-amino acid

library yielded several new peptide candidates. Adhesion and biological properties of

these peptides are analyzed here, a hybrid peptide proposed and further tested. The

identification of D-amino acid peptides for the interruption of cell adhesion to ECM

proteins or dermal fibroblasts should be useful in the increased killing of distant

metastases, as well as providing insight into regulation of integrin-ligand interactions.

46 Results

RZ-3, HYD-1 and AG-73 Bind to Prostate Tumor Cell Surfaces

The RZ-3, RZ-4, RZ-6, RZ-12, HYD-1 peptides and the previously reported AG-

73 peptide [97] were tested for their ability to bind to DU-H cells by FACS analysis. In

Figure 4 (top panel), the mean peak of the fluorescence (MPF) for each peptide is shown

and indicates that Bodipy neuralite avidin alone resulted in a barely detectable amount of

non-specific binding. The RZ-4, RZ-6, and RZ-12 peptides all demonstrated slight

activity above the background of Bodipy neuralite avidin alone. The RZ-3, HYD-1 and

AG-73 peptides exhibited significantly higher amounts of cell binding as compared to

Bodipy neuralite avidin alone. Use of the RZ-3 peptide resulted in a mean peak of

fluorescence (MPF) of 350; HYD-1 resulted in a MPF of 1200. The previously reported

AG-73 peptide resulted in a MPF of 500. The scrambled peptide, HYDS-1, had a MPF

similar to the non-specific binding detected by the bodipy neuralite avidin alone

indicating that the scrambled peptide did not bind to the cells. The fluorescence

histogram for each binding peptide is shown in the remaining panels of Figure 4. All

three peptides result in a normal distribution pattern of cell binding, indicating that there

are populations of cells which are low and high binders of the peptides. The AG-73

peptide had the broadest distribution pattern, followed by RZ-3 and HYD-1 respectively.

Of the three peptides, the HYD-1 peptide displays the most uniform distribution. The

distribution of both RZ-3 and HYD-1 peptides was also examined in live DU-H prostate

tumor cells by confocal microscopy. The data in Figure 5 indicate that the peptides

remained on the surface of the cells during the 30-minute incubation period.

47

Figure 4. Fluorescence-activated cell sorting (FACS) analysis of biotinylated peptides bound to DU-H prostate carcinoma cells. The cells were incubated with BODIPY-peptide complexes for 30 minutes and the amount of peptide bound was estimated using FACS analysis. The mean peak of fluorescence (MPF) for each peptide from the resulting histograms are shown in the top panel. The representative histograms for the AG-73, HYD-1, HYDS-1 and the RZ-3 peptides are shown in the remaining panels.

48

Figure 5. Confocal microscopy of the peptides RZ-3 and HYD1 binding to surface of human prostate tumor cells. The DU-H cells were incubated with BODIPY-RZ-3 (top panel,) or BODIPY-HYD-1 middle panel) peptide or BODIPY alone (bottom panel) for 30 minutes and the peptide (green) was observed using confocal microscopy on the live cells. The nuclei are stained with propidium iodide (red). The bar is 25 microns.

49 The confocal image also indicates that all of the cells were decorated with the peptide.

DU-H Prostate Tumor Cells Adhere to Immobilized Peptide and ECM Proteins

Using the three peptides that displayed the most cell-binding activity, RZ-3,

HYD-1 and AG-73, the biological activities of the peptides were further tested. The

ability of DU-H cells to adhere to the peptides immobilized on a plastic surface was

compared to the ability of the cells to adhere to several immobilized natural ECM

ligands, such as fibronectin, laminin-1 and collagen-1 (Figure 6).

All three peptides supported cell adhesion within the 60-minute incubation period

(Figure 6). The HYD-1 peptide promoted the most adhesion, with concentrations as low

as 2µg/well supporting cell attachment. Maximal cell adhesion to HYD-1 occurred at

10µg/well. The scrambled peptide, HYDS-1, did not support cell adhesion at any

concentration tested (i.e.10-100µg/well). The RZ-3 peptide required 10-50µg/well for

tumor cell adhesion. A comparison of the peptides indicates that 50% cell binding

occurred using 1.5µg, 20µg or 30µg per well for HYD-1, AG-73 and RZ-3 respectively.

Mixtures of the peptides were not synergistic for promoting adhesion (data not shown).

All three ECM proteins are sufficient ligands for DU-H adhesion, and can support

adhesion in a similar manner at concentrations as low as 0.1µg/well. A comparison of

the ECM proteins indicates that 50% cell binding occurred using 0.1µg, 0.6µg and 1.5µg

of fibronectin, laminin-1 and collagen IV respectively. Taken together, a comparison of

the ability of the peptides and the ECM proteins to support adhesion indicates that the

50

Figure 6. Comparison of human prostate tumor cell adhesion to immobilized peptides or ECM proteins. The DU-H cells were allowed to attach to HYD1 (open circles), AG73 (open triangle), RZ3 (closed triangle), or HYDS (open square) peptide coated 96-well microtiter plates (top panel) or fibronectin (closed square), laminin-1 (open circle), or collagen IV (closed triangle) protein coated 96-well microtiter plates (bottom panel). The number of cells attached after 60 minutes was determined by absorbance at 570 nm. Data are the mean triplicates. Error bars = SD.

51

HYD-1 peptide compares favorably to the natural ligands.

Tumor Cell Adhesion to Immobilized Peptides is Integrin Dependent

Next, we tested if cell adhesion to the peptides or the natural ligands was

dependent upon cations or whether the interaction could be blocked by integrin function

blocking antibodies. Cell attachment to laminin-1 and fibronectin was inhibited by 3mM

EDTA as expected (Figure 7), indicating that these interactions are conducted through an

interaction that requires cations [132]. In contrast, cell attachment to RZ-3, and HYD-1

was not affected by 3mM EDTA, suggesting that the adhesive properties occur through

an integrin interaction which does not require cations. This is consistent with a peptide

interaction occurring independent of the calcium binding motif. The calcium binding

motifs of the α integrin subunit lie on a surface of the integrin far from the ligand contact

sites [132]. Cell attachment to laminin-1 and fibronectin was not affected by the

addition of 10 ug/ml heparin suggesting that the cell adhesion to the immobilized matrix

proteins was not due to the positive charge interactions with the cell surface. Similarly,

attachment to RZ-3, and HYD-1 was unaffected by adding heparin (data not shown).

Cell attachment to fibronectin was inhibited by the anti-α5 and the anti-β1 antibody,

consistent with its known function as a ligand for the α5β1 integrin. Cell attachment to

laminin-1 was inhibited by anti-α3, 6 and anti-β1 integrin antibodies, consistent with the

fact that adhesion to laminin-1 is mediated by both the α3, 6 and β1 integrins. Cell

attachment to laminin-5 was partially inhibited by the α3, 6 antibodies and unaffected by

52

Figure 7. Effect of antibodies and reagents on DU-H cell adhesion to peptides and extracellular matrix proteins. The DU-H cells were allowed to attach to Fibronectin (1µg/well), Laminin-1 (1µg/well), Collagen IV (1µg/well), HYD-1 (10µg/well), or RZ-3 (50µg/well) coated plates in the presence of 10µg/ml IgG, 3mM EDTA; anti-α 2; anti-α5; anti α3 and 6; anti α2 and 5 integrin antibodies or anti-β1 integrin antibody, as indicated. After 60 minutes of incubation at 37oC, the number of attached cells was determined by absorbance at 570nm. Data are expressed as a mean of triplicate results; error bars are the standard deviation. The percent of control is the absorbance value of cells with the function-blocking antibody divided by the absorbance value of cells without the function-blocking antibody X 100.

53 the β1 blocking antibody. A combination of the α2, 3, 5, and 6 integrin blocking

antibodies partially inhibited adhesion to immobilized HYD-1 or RZ-3 peptides.

Although cell adhesion to HYD-1 was not inhibited by β1 blocking antibodies, HYD-1

peptide can inhibit cell adhesion to the β1 integrin specific activating antibody TS2/16

(Figure 8). Taken together, the data suggest that cell adhesion to the immobilized RZ-3

and HYD-1 are integrin dependent.

Inhibition of DU-H Cell Attachment to ECM Proteins

The next test of the biological properties of the peptides was to determine

whether the soluble peptides could inhibit DU-H adhesion to the immobilized ECM

proteins. The DU-H cell attachment to immobilized laminin-1 and fibronectin was

decreased in a concentration-dependent manner when HYD-1 or RZ-3 was added to the

wells (Figure 9). The RZ-3 peptide was the most effective peptide for inhibiting

attachment to all ECM proteins, including laminin-5, as judged by comparing the

concentration of peptide required to inhibit 50% of the cell adhesion (IC50) to the

respective ECM protein. The previously reported AG-73 peptide inhibited cell adhesion

to fibronectin, requiring approximately 9ug/well and did not inhibit cell adhesion to

laminin-1 or collagen IV. The HYD-1 peptide inhibited cell adhesion to laminin-1,

fibronectin and collagen IV in a similar manner. The HYD-1 peptide inhibition of cell

adhesion showed a threshold response in that greater than 2µg/well of peptide was

required before an inhibition of cell adhesion was detected. The scrambled HYDS-1

peptide was not able to inhibit adhesion

54

Figure 8. Inhibition of DU145 cell adhesion to immobilized ββββ1 integrin antibody by peptides. The cells were allowed to attach to the immobilized β1 integrin antibody called TS2/16 (2µg/well) in the presence of increasing concentrations of soluble peptide (HYD-1, RZ-3, HYDS-1 or AG-73) using serum free medium. After one hour at 37oC, the number of attached cells was determined by absorbance at 570nm. Data are expressed as a mean of triplicate results; error bars are the standard deviation.

55

Figure 9. Inhibition of DU-H cell adhesion to extracellular matrix proteins by peptides. The cells were allowed to attach to laminin-1 (closed triangle, 1µg/well), fibronectin (open circle, 1µg/well), laminin-5 (closed circle, 1µg/well) or collagen IV (open triangle, 1µg/well) coated 96 well microtiter plates in the presence of various amounts the RZ-3, HYD-1 or HYDS-1 peptides using serum free media. After a 60 min incubation at 37oC, the number of attached cells was determined by absorbance at 570nm. Data are expressed as a mean of triplicate results. The percent of control is the absorbance value of cells with the peptide divided by the absorbance value of the cells without the peptide X 100.

56 to any ECM protein. A comparison of the IC50 values of the peptides reveals that the

most potent inhibitor for attachment to all three ECM proteins is RZ-3.

Inhibition of SCC-25 and HFF Co-Culture Interaction by HYD-1 Peptide

The biological relevance of the adhesion blocking ability of the HYD-1 peptide

was tested using an epithelial-fibroblast co-culture model system. Epithelial stromal

interactions are increasingly recognized as determinants of tumor progression. The

adhesion of SCC-25 cells to a HFF monolayer is a time dependent process that was

maximal within one hour of incubation at 37oC (Figure 10). The HYD-1 peptide

inhibited approximately half of the SCC-25 cells from attaching to the HFF monolayer.

The inhibition of SCC-25 adhesion was maintained over the three-hour period of the

assay. The scrambled form of the peptide, HYDS-1, was ineffective in altering adhesion.

Adhesion to Immobilized HYD1 Prevents Cell Spreading and Adhesion Dependent

Phosphorylation

Cell spreading is a critical event that normally follows cell adhesion to the

extracellular matrix. Cell adhesion is coupled to integrin activation, initiating multiple

signaling pathways [133,134]. In general, cell spreading occurs from extensive actin

cytoskeletal remodeling through actions of the Rho Family of small GTPases [133].

Several focal adhesion proteins play a regulatory role in cell spreading events such as

focal adhesion kinase (FAK), paxillin, and p130CAS [135]. In addition, phosphorylation

57

Figure 10. Inhibition of SCC-25 cell adhesion to HFF monolayers by peptides. Approximately 3 x 105 SCC-25 cells were plated onto HFF monolayers in the absence (circle) or presence of the HYD-1 (square), HYDS-1 (triangle) peptides. After the indicated times of incubation, the number of SCC-25 cells unable to attach was determined using a hemocytometer. The number of HFF cells unable to attach was also determined in the presence of the HYD-1 peptide (diamond). A minimum of three determinations were done at each time point. The percent adhesion is the number of SCC-25 cells adhering to the HFF monolayer divided by the number of SCC-25 cells added to the wells X 100.

58 of mitogen activated protein kinase kinase (MEK) at a specific residue, serine 298, is an

adhesion dependent phosphorylation regulated by p21-activated kinase (PAK) [136].

Using immobilized peptide as a ligand, we found that HYD1 is an adhesion

agonist by mediating integrin-dependent tumor cell adhesion (Figures 6-8). To determine

if HYD1 is a sufficient ligand to initiate cell spreading, we quantified the amount of cell

spreading and average area occupied by the cells after adhesion to immobilized HYD1 or

laminin-5 after 1 hour. As expected, adhesion to laminin-5 initiates cell spreading in

nearly 100% of the cells within a microscopic field (Figure 11A). Interestingly, adhesion

to HYD1 prevented cell spreading even in the presence of EGF (10ng/ml) (Figure 11B)

suggesting that adhesion to immobilized HYD1 uncouples integrin-dependent adhesion

from cell spreading.

Since cell spreading events are tightly regulated by intracellular signals [133,135],

we examined two integrin-proximal cellular signals that occurred during adhesion to

HYD1. Adhesion to HYD1 induced phosphorylation of FAK on tyrosine 397 and MEK

on serine 298 after thirty minutes of attachment (Figure 12A and B). The magnitude of

the response was quite different when compared to cells attached to laminin-5 over the

same time course (Figure 12A and B). Laminin-5 induced more activation of FAK at

both 10 and 30 minutes post-adhesion while inducing significantly more phosphorylation

of MEK at 10 minutes post-adhesion. Adhesion to HYDS was not able to induce a

comparable response with either signal. Taken together these data suggest that adhesion

to immobilized HYD1 uncouples cellular adhesion from cell spreading in part by

suppressing adhesion dependent phosphorylation events.

59

Figure 11. Adhesion to immobilized HYD1 prevents cell spreading. Spreading of PC3N cells was determined using differential interference contrast optics on an inverted Olympus IMT2 microscope. A) PC3N cell adhesion on a native ECM ligand, laminin-5 (HaCaT conditioned media), after 1 hour. B) PC3N cell adhesion on HYD1 (150µg) + EGF 10ng/ml after 1 hour. C) Cell spreading was quantified with Compix Imaging software. Error bars are standard error from the mean area spread of 15 cells/experiment.

60

Figure 12. Suppression of adhesion dependent phosphorylation by immobilized HYD1. Phosphorylation of focal adhesion kinase (FAK) on tyrosine 397 (A) and the mitogen-activated protein kinase MEK1 on serine 298 (B) was analyzed by western blot using phosphospecific antibodies. Densitometry analysis of percent phosphorylation is shown. PC3N cells were serum-starved overnight and attached in serum-free conditions to the indicated ligands for 10 and 30 minutes. Peptides were coated at a concentration of 150µg/plate. HYDS is a scrambled peptide derivative of HYD1.

61 Discussion

Using a previously described method to isolate candidate peptides for biological

activity with the α6 integrin, we report the existence of two bioactive D-amino acid

containing peptides called RZ-3, and HYD-1. These peptides attach to the surfaces of live

tumor cells, can themselves support cell attachment, can inhibit attachment to ECM

proteins such as fibronectin, laminin-1, laminin-5 and collagen IV and can inhibit

epithelial stromal adhesion interactions.

The inhibition of tumor cell adhesion to a number of ECM proteins was observed

with both peptides. In addition, the integrin blocking antibodies only partially inhibited

the adhesion of the cells to the peptides. Taken together, these data suggest that the

peptides can affect adhesion processes that are both integrin dependent and independent.

This fact may have significance for both understanding cross-talking pathways of cell

adhesion to the ECM and interrupting adhesion dependent biological events. The broad

nature of the inhibition could prove useful in disabling the varied adhesion events that are

activated within human tumor cell populations. It should be noted that these peptides do

not show homology to any known proteins in the SwissProt database from a BLASTP

search (data not shown).

The potential use of the peptides could include the prevention of tumor cells

adhesion in vivo. Preliminary experiments utilizing a mouse human tumor xenograft

model system [137] suggests that the peptides themselves are not toxic to animals (data

not shown). The alteration of tumor cell adhesion could be of benefit for preventing

metastasis as well as increasing the killing of tumor cells that are refractory to treatment.

62

The peptides may be particularly useful in human tumors with a wild type p53

function since clustering of integrin in tumor cells containing wt p53 can trigger

apoptosis [104]. Our preliminary experiments indicate that the altering of attachment can

sensitize the tumor cells to the killing effects of ionizing radiation (data not shown). We

are currently testing both the efficacy of the peptides to inhibit tumor cell adhesion in

vivo and the potential for synergistic lethality with DNA damaging agents in vivo.

It has been demonstrated that fibronectin-mediated adhesion confers a survival

advantage for myeloma cells acutely exposed to cytotoxic drugs in a phenomenon known

as cell adhesion mediated drug resistance (CAM-DR)[108,138]. Myeloma cells within

the bone marrow are adherent to fibronectin and are thought to serve as a reservoir of

tumor cells that are difficult to eradicate. The anti-adhesive peptides HYD-1 and RZ-3

may be lead candidates to overcome CAM-DR.

The role of the ECM as a cell survival factor has been demonstrated in both

normal cell types and fibroblasts. The ECM is critical for normal survival signals and the

loss of the ECM will result in normal cell death in a process termed anoikis [139-142].

However, during transformation, normal cells down regulate many of their adhesive

interactions and display anchorage independent cell growth. Although the majority of the

adhesion events are suppressed during transformation, selected adhesion events are

preserved. For example, in prostate cancer progression, the majority of the integrin ECM

receptors and the E-cadherin class of adhesion molecules are suppressed [143] while the

laminin receptors and N-cadherin remain expressed [23,94]. The preserved adhesion

63 events may be important targets for inactivating the known survival advantage of

invasive or metastatic tumor cells.

Finally, the cell adhesion peptides characterized in this study have both agonist

and antagonistic properties when compared to native extracellular matrix ligands. Both

HYD1 and RZ3 were active as adhesion mimetics by both supporting tumor cell adhesion

when immobilized and inhibiting tumor cell adhesion to immobilized extracellular matrix

proteins. However, adhesion to HYD1 was not sufficient to produce cell spreading. In

addition, compared to laminin-5, adhesion to HYD1 did not initiate similar adhesion-

dependent signals. Therefore, HYD1 uncouples cell adhesion and cell spreading in part

by preventing the appropriate adhesion-dependent phosphorylation events. The ability of

HYD1 to interrupt the normal response following cell adhesion may prove useful in

developing agents to block cell invasion and metastasis.

64 IV. SYNTHETIC D-AMINO ACID PEPTIDE INHIBITS TUMOR

CELL MOTILITY ON LAMININ-5

Introduction

Cell migration is a complex process integral to normal biological events such as

wound healing and inflammatory responses as well as the pathological circumstances of

tumor invasion and metastasis. The motile nature of all cell types depends upon the

actions of many different molecular components [144]. Central to this process are the

signaling and cytoskeletal responses elicited by the interactions of integrins with the

extracellular matrix (ECM). The adhesive complexes formed from integrin ligation and

activation regulate intracellular signaling events that dictate the cytoskeletal

reorganization necessary for cell movement [56]. Several signaling pathways have been

shown to be important for cell movement and specific pathways may have crucial roles

depending on the extracellular environment [56,145-147]. In addition, tumors associated

with an invasive and migratory phenotype may favor a certain integrin repertoire

[93,148,149], displaying a pivotal role of specific integrin/ECM interactions that favor

tumor metastasis.

It is well established that the ECM can induce integrin dependent cell spreading

and migration by activation of specific signaling programs that regulate focal adhesion

and cytoskeletal dynamics [56,144]. Interestingly, specific integrin/ECM pairs have been

shown to differentially modulate the activities of these programs [147,150] suggesting

that integrin and ECM composition will dictate the signaling response and phenotype.

65 Laminin-5 dependent cell spreading and migratory activities, for example, has been

linked to the activities of focal adhesion kinase (FAK), phosphoinositide 3-OH kinase

(PI3-K), p21-activated kinase, and the mitogen-activated protein kinase pathway

[147,151,152]. Inhibition of these pathways using small molecules can ultimately block

a migratory phenotype. However, given the multitude of components in these pathways

and their redundancy in function, targeted dysregulation of integrin/ligand activity may

prove to be a more potent method to inhibit motility.

Integrins are evolutionarily conserved heterodimeric cell surface molecules. To

date there are 18 distinct α and 8 distinct β subunits that pair in a restrictive manner to

give about 24 different integrins that have individual ligand binding specificities [134].

The primary ligands for integrins are proteins of the ECM that consist of type I and IV

collagens, fibronectin, laminins, heparin sulfate proteoglycan, and other noncollagenous

glycoproteins [109]. The integrins α6β1, α6β4, and α3β1 are laminin receptors,

[110,111] and these integrin pairs are associated with the progression of many epithelial

tumors [23,112,115]. In particular, the α6 subunit is continually expressed during

prostate cancer progression and found in micrometastases [23,27,149]. Previous studies

have shown that biologically active peptides developed from defined regions within

laminin chains can have profound effects on biological events including cell migration

and metastasis [97,123,125,127,153-156]. These findings prompted us to develop α6-

binding cell adhesion peptides.

Our previous work [81,82] has identified human tumor cell adhesion peptides

capable of binding prostate carcinoma cells expressing the α6 integrin. We characterized

66 two D-amino acid peptides, HYD1 (kikmviswkg) and RZ3 (kmviywkag), as cell

adhesion peptides based on their ability to both support tumor cell adhesion themselves

and inhibit tumor cell adhesion to immobilized ECM proteins [82]. In the present study,

we examine the effect of these peptides on laminin-5 dependent haptotaxis. HYD1

causes dramatic cytoskeletal reorganization in prostate tumor cells adhered to laminin-5,

resulting in a loss of cell migration. HYD1 interacts with both α6 and α3 integrin

complexes and induces signaling through FAK, MEK, and ERK. These data show that

HYD1 is a novel synthetic peptide that disconnects pro-migration phosphorylation

signals from cytoskeletal dependent migration.

Results

HYD1 Blocks Laminin-5 Dependent Migration and Invasion

Effect of the peptides, HYD1 and RZ3, on laminin-5 mediated cell migration and

invasion was investigated. Video microscopy was used to measure random haptotaxis

and a modified Boyden chamber assay was used to assess the effects on invasion toward

laminin-5. In order to analyze the effect of the peptides independent of disrupting

adhesive contacts with the ligand, cells were allowed to adhere for 1 hour prior to

treatment with peptides in serum-free media. Treatment with HYD1 completely blocked

random haptotaxis on laminin-5 that was quantified for a four-hour period (Figure 13A

and B). A scrambled variant of HYD1, HYDS, was inactive. The activity of HYD1 was

a post-ligand-occupancy event; there was no loss of adhesion and the cells remain spread

over the course of the video (4 hours). In addition, membrane surfaces were observed to

67

Figure 13. HYD1 blocks cell migration on laminin-5. (A) PC3N cells were placed on laminin-5 for 1 hour followed by the addition of serum-free media or serum-free media containing HYD1 (75µg/ml). Videos were started approximately 30 minutes post-treatment. Time-lapse images were taken at start of video T=0, T=15 (15 minutes), and T=30 (30 minutes). White arrowhead in T=0 frame marks the cell followed during analysis at T=30. Scale bar = 100µm.

68

Figure 13 Continued. (B) Cells were added as described in A and treated with 75µg/ml of the peptides HYD1, RZ3, AG73, or HYDS in serum-free media. The Src inhibitor, PP2, was used at a concentration of 10µM. Videos were started 30 minutes post-treatment and images were taken every 5 minutes for 4 hours. (C) The effect of HYD1 is reversible. Cells were added as described in A. Cells were treated with HYD1 for the entire course of the video (4 hours). Alternatively, cells were treated for 30 minutes, the peptide was washed-out, and the cells were exposed to serum-free media alone or serum-free media containing soluble laminin-5 and allowed to recover over the 4-hour period. Migration rates were quantified as described in Experimental Procedures. Error bars are standard error from the mean migration rate of 15 cells/experiment.

69 remain active after peptide treatment and cell division occurred normally. Cells remained

non-motile after cell division and reattachment. The effect of HYD1 was reversible

following peptide wash-out (Figure 13C). Adding soluble laminin-5 after peptide wash-

out enabled a faster recovery of migration over a four-hour period as compared to adding

serum-free media, indicating that the cells retain the capacity to respond to a pro-

migratory stimulus. The inhibitory activity for HYD1 was comparable quantitatively to

PP2, a Src family kinase inhibitor previously published to inhibit migration of prostate

carcinoma cell lines [157] (Figure 13B). The morphology of cells treated with HYD1 as

compared to PP2 suggest a distinct mechanism of action. HYD1 treated cells remain

spread and have active membrane surfaces compared to the non-spread, rounded

morphology of cells treated with PP2 (Figure 14). RZ3 reduced the rate of random

haptotaxis (Figure 13B), however its inability to remain soluble may contribute to the

activity since many cells were trapped in an insoluble peptide web (data not shown). In

addition, AG73, a cell adhesion peptide derived from the alpha globular domain of

laminin-1 [97] had no migration-blocking activity in this assay.

HYD1 also was functional in inhibiting invasion toward immobilized laminin-5.

The activity of HYD1 to inhibit invasion was concentration dependent with

approximately 60% inhibition at a concentration of 250µg/ml (Figure 15A). The

scrambled variant, HYDS, was inactive at all concentrations tested. The HYD1 peptide

was able to inhibit invasion with similar potency as other known inhibitors of invasion

such as the Rho-kinase inhibitor, Y-27632 [158], as well as integrin-function blocking

70

Figure 14. Soluble HYD1 does not effect cell spreading on immobilized laminin-5 Spreading of PC3N cells was determined using differential interference contrast optics on an inverted Olympus IMT2 microscope. Following 1 hour adhesion to laminin-5 in serum-free conditions cells were treated with serum-free media alone (SFM), HYD1 (75mg/ml), a Src inhibitor, PP2 (10mM), or a Rho-kinase inhibitor, Y-27632 (25mM) in serum-free media.

71

Figure 15. HYD1 inhibits laminin-5 dependent cellular invasion. (A) Concentration-dependent inhibition of PC3N cell invasion toward laminin-5. Cells were added into the upper chamber of laminin-5 coated transwells for 1hour prior to treatment with 75, 100, 250, or 500 (µg/ml) HYD1 or HYDS in serum-free media as described in Experimental Procedures. (B) Comparison of HYD1 activity to other known inhibitors of invasion. Cells were added as in A and followed by treatment with 250µg/ml HYD1 or HYDS in serum-free media. The Rho-kinase inhibitor, Y-27632, was used at a concentration of 25µM in serum-free media. Integrin function-blocking antibodies P1B5 (α3), GoH3 (α6), and AIIB2 (β1) were used at a concentration of 10µg/ml and were incubated with cells for 15 minutes at room temperature prior to adding the cells into transwells. Error bars are standard deviation from the mean.

72 antibodies to α6, α3, and β1 (Figure 15B). These data indicate that HYD1 can block

random haptotaxis on laminin-5 and inhibit cellular invasion toward laminin-5.

HYD1 Induces Actin Cytoskeletal Remodeling and Cortactin Mediated Actin

Dynamics at the Cell Membrane

A critical component of cell motility dynamics is the regulation of the actin

cytoskeleton [159,160]. Given the immediate loss of motility after HYD1 treatment we

investigated the concurrent changes in actin organization. Cells treated for 30 minutes

with HYD1 displayed increased filamentous actin and an accumulation of actin at the cell

membrane with associated microspikes (Figure 16A and B-asterisk). This robust change

in the cytoskeletal architecture is not seen with treatment of the cells with scrambled

peptide, HYDS. Interestingly, although HYD1 generated the formation of stress fibers

(Figure 16B-arrows), the peptide did not increase Rho activity (data not shown)

suggesting Rho-independent signaling events are responsible for the phenotype.

Cortactin is a cortical actin binding protein that localizes at sites of dynamic

membrane activity [161]. Given the accumulation of actin and actin microspikes at the

cell membrane following HYD1 treatment, we hypothesized that this phenotype may be

mediated through cortactin. We show that cortactin is involved in HYD1 induced

accumulation of filamentous actin at the cell membrane by its colocalization with actin at

the cell membrane in cells treated with HYD1 for 30 minutes (Figure 16B Inset). In

addition, HYD1 induced an increase in tyrosine phosphorylation of cortactin over time

(Figure 17A). This is consistent with a concomitant increase in filamentous actin in the

73

Figure 16. HYD1 induces cytoskeletal reorganization on laminin-5. (A) HYD1 treatment causes global actin reorganization. PC3N cells were placed on laminin-5 coated coverslips for 1 hour followed by addition of HYD1 or HYDS (75µg/ml) in serum-free media for 30 minutes. Coverslips were fixed and stained for actin (green) and cortactin (red) as described in Materials and Methods. Asterisks designate points of microspike formation at the cell membrane. Closed arrowhead marks the cell enlarged in B. (B) HYD1 induces colocalization of cortactin with actin at the cell membrane. The cells marked in A were enlarged using Photoshop 7.0 software. The insets were created by using the auto level function in Photoshop on the boxed region of the cell membrane. Asterisks designate points of microspike formation at the cell membrane. Closed arrows designate areas containing stress fibers.

74

Figure 17. HYD1 induces phosphorylation of cortactin and alters membrane actin dynamics. (A) HYD1 induces phosphorylation of cortactin. PC3N cells were placed on laminin-5 for 1 hour followed by treatment with HYD1 or HYDS (75µg/ml) in serum-free media for 5, 10, 15, or 30 minutes before lysis. The phosphorylation state of cortactin was analyzed after immunoprecipitation with the phosphotyrosine antibody 4G10. (B) HYD1 induces F-actin assembly at the cell membrane. PC3N cells were added as described in A and treated with HYD1 or HYDS for 30 minutes. The distribution of F-actin in the membrane compared to the globular pool of actin was analyzed as described in Experimental Procedures. (C) Desitometry plot of results shown in B.

75 cell membrane fraction following treatment with HYD1 for 30 minutes (Figure 17B, C).

These data show that HYD1 stimulates cytoskeletal reorganization mediated in part by

signaling through cortactin.

HYD1 Interacts with αααα6 and αααα3 Integrins and Activates Integrin-Associated

Signaling

Given that cell migration on laminin-5 is mediated by α6 and α3 integrins

[152,162-165], we determined if either peptide interacted with α6 or α3 integrin

subunits. To define receptor specificity, each peptide was incubated with membrane

preparations from DU-145H prostate carcinoma cells and the resulting precipitate was

analyzed for integrin content. HYD1 interacted with complexes containing full-length

α6 and α3 integrins (Fig. 18A and B). RZ3 interacted with α3 integrin to a greater extent

than α6 integrin as judged by the amount of integrin retrieved. HYD1 was superior to

RZ3 in retrieving the α6 and α3 subunits. The scrambled derivative, HYDS, does not

interact with either alpha integrin subunit (Fig. 18A and B).

Upon engagement with the ECM, integrins are signal transduction receptors with

the capability of activating many intracellular protein kinases and influencing a wide

array of cellular processes [134]. One kinase linked to integrin activation and cell

motility is focal adhesion kinase (FAK), and phosphorylation of FAK on tyrosine 397 is

indicative of FAK activation [58,166]. Phosphorylation of mitogen activated protein

kinase kinase (MEK) on serine 298 is an adhesion dependent phosphorylation regulated

by p21-activated kinase (PAK) [136]. Using these two signals as biochemical markers

76

Figure 18. HYD1 interacts with αααα6 and αααα3 integrins. Affinity precipitation reactions were performed with HYD1, RZ3, or HYDS peptides on DU-145H cell membrane fractions as described in Experimental Procedures. (A) The precipitates were analyzed for α6 or (B) α3 integrin by SDS-PAGE electrophoresis followed by a western blot with AA6A antibody (α6 specific) or Antibody 1920 (α3 specific). ML (membrane lysate) was used to indicate the starting material.

77 for integrin activation, we determined if HYD1 or RZ3 could act as signaling ligands to

PC3N cells attached to BSA-coated plates. After 30 minutes of exposure, HYD1 induced

both activation of FAK and phosphorylation of MEK on serine 298 (Figure 19A Lane 2

and B Lane 2). RZ3 and the scrambled derivative, HYDS, and AG73 showed no activity

compared to cells treated with serum-free media alone.

HYD1 Enhances Signaling on Laminin-5 Resulting in Transient Activation of ERK

Cellular haptotaxis involves the dynamic regulation of signaling and cytoskeletal

components. Ultimately, signals elicited from integrin interaction with the ECM drive

the dynamic adhesive and cytoskeletal remodeling necessary for movement [56]. The

anti-migratory activity of HYD1 coupled with its ability to reorganize the cytoskeleton

prompted us to analyze the immediate effect on laminin-5 dependent pro-migratory

signals. As mentioned previously, cell migration on laminin-5 is coupled to the activity

of several kinases including FAK and p21-activated kinase (PAK) [147,152]. After 5

minutes of treatment, HYD1 increased the amount of activated FAK and elevated

phosphorylation of MEK on serine 298, which is mediated by PAK [136], compared to

baseline levels generated by haptotaxis on laminin-5 (Figure 20A and B Lanes 1 and 2).

Both of these signals had the greatest difference above baseline at 15 minutes post-

treatment (Figure 20A and B Lanes 3 and 4). At this time-point RZ3 had similar activity,

albeit less than HYD1, while AG73 did not show an effect (Figure 21A and B). Given

that PAK phosphorylation of MEK on serine 298 primes the MAPK pathway for

activation, [136,167] we investigated the activation of ERK in response to the peptides.

78

Figure 19. HYD1 activates integrin-associated signaling. PC3N cells were attached to BSA coated plates in the presence of serum-free media for 1 hour followed by the addition of peptides (75µg/ml) in serum-free media or serum-free media (SFM) alone for 30 minutes. Cell lysates were analyzed for phosphorylation of FAK Tyr-397 (A) and phosphorylation of MEK Serine 298 (B). Blots were stripped and reprobed for total FAK and MEK and the amount of activity was normalized by densitometry.

79

Figure 20. HYD1 temporally enhances activation of FAK and phosphorylation of MEK on laminin-5. PC3N cells were placed on laminin-5 coated tissue culture plates in the presence of serum-free media for 1 hour followed by the addition of HYD1 (75µg/ml) in serum-free media or serum-free media (SFM) alone for 5, 15, and 30 minutes. (A) Effect of HYD1 on FAK activation (Tyr-397). (B) Effect of HYD1 on MEK phosphorylation (Serine 298). Blots were stripped and reprobed for total FAK and MEK and the amount of activity was normalized by densitometry.

80

Figure 21. HYD1 enhances laminin-5 signaling and transiently activates ERK. PC3N cells were placed on laminin-5 coated tissue culture plates in the presence of serum-free media for 1 hour followed by the addition of peptides (75µg/ml) in serum-free media or serum-free media (SFM) alone for 15 minutes. Cell lysates were analyzed for phosphorylation of FAK Tyr-397 (A), phosphorylation of MEK Serine 298 (B), and phosphorylation of ERK Thr-202/Tyr-204 (C). (D) Time course of ERK activation following treatment with 75µg/ml of HYD1 or HYDS. Blots were stripped and reprobed for total FAK, MEK, ERK and the amount of activity was normalized by densitometry.

81 HYD1 induced activation of ERK (Figure 21C) that was maximal at 10 min post-

treatment and returned to baseline levels after 30 minutes (Figure 21D). Taken together

these data demonstrate that HYD1 disrupts the normal pro-migratory signaling dynamic

on laminin-5 and results in a transient activation of ERK.

Discussion

We have identified a D-amino acid peptide, HYD1 (kikmviswkg), which inhibits

tumor cell migration on laminin-5. HYD1 interacts with both α6 and α3-containing

integrin complexes. Thus, HYD1 interacts with the integrins α6β1, α6β4, and α3β1.

These are all receptors for laminin-5 [168] and although α3β1 has been documented as

the primary integrin receptor involved in mediating migration on laminin-5 by use of

function-blocking antibodies to α3 [152,162], evidence also suggests that α6 integrins

play a major regulatory role in motility on laminin-5 [163-165]. The receptor specificity

mediating the direct effects on motility, signaling, and the cytoskeleton will require

further investigation.

It is well known that integrin function-blocking antibodies or RGD-based

peptiomimetics can inhibit cell invasion by blockage of integrin/ECM interaction. HYD1

is novel in that its activity is not dependent on interrupting integrin/laminin-5

interactions. This suggests that the effects are induced through binding to available

integrin complexes on the surface rather than integrin complexes pre-engaged with the

ECM. Alternatively, our previous study [82] suggested that HYD1 interaction was

independent of the calcium-binding motif. Therefore, the peptide may effect both non-

82 engaged and pre-engaged integrin complexes. The post-ligand occupancy activity of

HYD1 is reinforced by the fact that cells remained attached and spread on laminin-5 after

peptide treatment. The inhibitory effect of the peptide was reversible by removing the

peptide and adding soluble laminin-5. This suggests that the migration-blocking activity

is not a lethal event and can be rescued by the normal ligand.

Several peptide sequences that bind α6β1 have been isolated from the laminin

α1 chain [97,169] as well as a peptide from CCN1, [170] a protein that is involved in

angiogenesis. Screening of a phage display library [171] has also isolated α6β1 peptide

ligand mimetics. These peptides do not reveal a consensus binding motif and do not

overlap with the sequence of HYD1. In addition, since HYD1 is a D-amino acid peptide

it might be structurally distinct from other L-amino acid ligand mimetics and therefore

not a good candidate for comparative purposes. The diversity of the ligand sequences for

α6β1 may allude to the fact that integrins are involved in a multitude of biological

processes and specific ligands could interact with distinct binding sites on integrins to

give the appropriate biological response.

Studies have shown that migration rates of cells are dependent on the organization

of the actin cytoskeleton and the presence of long, centrally distributed stress fibers

correlates with reduced migration rates [150,151,172]. HYD1 generates this type of

cytoskeletal morphology within 30 minutes, producing a global increase in filamentous

actin both at the membrane and throughout the cytoplasm. Cortactin, a regulator of

membrane actin dynamics, is also shown to be tyrosine phosphorylated in response to the

peptide and colocalized at the cell membrane with filamentous actin. Although the full

83 significance of cortactin phosphorylation has yet to be determined, tyrosine

phosphorylation is partially mediated by Src in response to cell matrix interactions [173].

This suggests that the cytoskeletal remodeling events at the cell membrane produced by

HYD1 are activated in part by integrin mediated signaling through cortactin.

Interestingly, HYD1 induced cytoskeletal reorganization occurred independent of

increased RhoA activity (data not shown), however, RhoA effectors can be modulated by

other signaling pathways such as the MAPK pathway [56,64]. Therefore, HYD1 may

elicit changes in cytoskeletal architecture directly through RhoA effectors rather than

increasing RhoA activity. Alternatively, it has been shown in keratinocytes that ablation

of α3β1 created a cytoskeletal reorganization event that resulted in enhanced stress fiber

formation [174]. The interaction of HYD1 with α6 and α3 integrins is consistent with

the idea that the cytoskeletal effects could be a consequence of inhibiting integrin

function independent of modulating actin dynamics through cellular signals.

Laminin-5 dependent motility depends on the activity of multiple cell signaling

molecules including FAK, PAK, PI3K, and the MAP-kinase pathway, and studies have

shown that inhibition of these mediators can reduce migration [151,152,157]. It is

somewhat surprising then that HYD1 could block migration yet stimulate signaling

through these pathways. However, studies with disintegrins have displayed similar

phenomena. For example, it has been shown that contortrostatin, a snake venom

disintegrin that inhibits cell motility, induces the tyrosine phosphorylation of FAK,

p130CAS, and activation of ERK2 [175-178]. Disintegrins have also generated actin

cytoskeletal reorganization that was dependent on tyrosine kinase activity [179].

84 Although HYD1 does not contain a conserved disintegrin domain, the similarity of

activity is remarkable.

In summary, we have identified an integrin-targeting peptide, HYD1, that blocks

tumor cell migration on laminin-5. Loss of cell migration involved intense cytoskeletal

remodeling events mediated in part by cortactin. HYD1 interacts with full-length α6 and

α3 integrins and disrupts the pro-migratory signaling dynamic induced from laminin-5

haptotaxis. Therefore HYD1 uncouples the induction of pro-migratory signals with the

appropriate cytoskeletal response necessary for motility. The dramatic effect of HYD1

on cell migration and invasion prompts for further study to determine if it can efficiently

block invasion and metastasis in vivo. In addition, we are determining the minimal

element for activity in this peptide by deletion analysis. The minimal element of HYD1

may provide insight into novel compounds for drug development. HYD1 is novel

because it does not have to compete with the native ligand for activity, suggesting that

integrin functions can be regulated independently from adhesion to the extracellular

matrix. This study may provide new insights into development of antagonists of specific

integrin functions to target cancer metastasis.

85 V. IDENTIFICATION OF THE MINIMAL ACTIVE ELEMENT OF

THE SYNTHETIC D-AMINO ACID PEPTIDE HYD1 (kikmviswkg)

Introduction

Multiple studies have isolated biologically active peptides from defined regions

within laminin chains and documented profound effects on biological events including

cell migration and metastasis [97,123,125,127,153-156]. The discovery of RGD, the

tripeptide sequence found in many adhesive proteins such as fibronectin and vitronectin

[180,181], has led to several studies showing that this cell adhesion peptide has anti-

invasive and anti-metastatic effects both in vitro and in vivo [182,183]. Peptides derived

from the laminin β1 chain, YIGSR, and α5 chain, RLVSYNGIIFFLK, have also been

effective at blocking experimental metastasis [127,153,184]. HYD1, kikmviswkg, has

shown potent anti-migratory effects in vitro and warrants further study in experimental

models of metastasis. HYD1 is a linear peptide consisting of ten D-amino acids.

Because HYD1 is relatively large in size compared to other bioactive cell adhesion

peptides used in clinical studies, it is important to determine if the amino acid motif

contains a minimal element that mediates the biological activity of the peptide before

testing in mouse models. This will help in developing strategies to increase peptide

bioavailability and provide insight into the synthesis of novel compounds to target tumor

cell migration and metastasis.

Previous groups that isolated cell adhesion peptides from the laminin α1 chain

successfully determined the minimum active sequences of these peptides by using N-

86 terminal and C-terminal deletion mutants of the peptides [124]. To determine the

minimal element of HYD1, five N-terminal and five C-terminal deletion mutants were

synthesized. In addition, alanine substitution was performed throughout the sequence of

HYD1 to identify amino acids that are critical for activity. Three tests of bioactivity were

applied to determine the minimal element of HYD1: 1) Tumor cell adhesion. 2) ERK

activation. 3) Inhibition of tumor cell migration on laminin-5.

Results

The Minimal Element that Mediates Cell Adhesion to Immobilized HYD1 is

xkmvixw

Previously we demonstrated that HYD1, when immobilized, promoted cell

adhesion at concentrations as low as 2µg/well. Maximal cell adhesion to HYD1 occurred

at 10µg/well [82]. Since mutants of HYD1 have different molecular weights, equal

molar concentrations of each mutant was used to determine if adhesive activity was

retained. Cell adhesion to 50µM of immobilized HYD1, HYDS, N- and C-terminal

mutants, and alanine-substituted HYD1 was determined (Figure 22). Adhesion activity

of HYD1 was reduced following deletion of the second N-terminal lysine (Figure 22

Mutant 14). Additional deletion from the N-terminus did not entirely eliminate the

adhesive capacity of HYD1 suggesting that the five C-terminal amino acids retained

some adhesion-promoting activity. Deletion from the C-terminus reduced activity starting

with the loss of the C-terminal lysine (Figure 22 Mutant 18). Loss of additional C-

terminal residues (Figure 22 Mutants 19, 20, 21) completely abolished the adhesive

87

Figure 22. Cell adhesion to immobilized HYD1 is mediated by N- and C-terminal regions of the peptide. The DU-H cells were allowed to attach to HYD1, HYDS, deletion mutants (12-21), and alanine-substituted (2-11) peptides coated in 96-well microtiter plates. Each peptide was used at a concentration of 50µM. The number of cells attached after 90 minutes was determined by absorbance at 570 nm. Data are the mean triplicates. Error bars = SD.

88 activity of HYD1. The alanine-substituted mutants of HYD1 did not alter the adhesive

capacity of the peptide (Figure 22 Mutants 2-11).

Data from the deletion mutants suggested that both N- and C-terminal regions of

the peptide were bioactive. This initial screen indicated a minimal element of kmviswk.

To further explore crucial residues within this peptide, four additional truncated mutants

of HYD1 were synthesized. Adhesion activity of the peptides, ikmviswk- ikmvisw-

kmviswk- kmvisw, were determined over a concentration range of zero to four-hundred

micromolar (Figure 23). Two of these peptides, ikmviswk and ikmvisw, supported tumor

cell adhesion in a concentration dependent manner with maximal adhesion occurring at a

concentration of 100µM. The peptides kmviswk and kmvisw did not have adhesive

activity over the concentration range tested. These data suggest that ikmvisw is the

active adhesive sequence within HYD1 (kikmviswkg).

Although alanine-substitution mutants of full-length HYD1 did not reveal critical

amino acids (Figure 22), we postulated that alanine substitution with the smaller peptide

would be informative. Alanine-substitution mutants of ikmvisw were synthesized. Cell

adhesion to these mutant peptides revealed that the serine and N-terminal isoleucine were

not critical elements for adhesion to immobilized ikmvisw (Figure 24A). However,

substitution of alanine for any remaining amino acid within ikmvisw resulted in loss of

peptide activity. Interestingly, this minimal sequence is also a conserved element within

the sequence of RZ3 (Figure 24B). These data show that the minimal element of HYD1

that mediates cell adhesion is xkmvixw.

89

Figure 23. The minimal sequence mediating cell adhesion to immobilized HYD1 is ikmvisw. The DU-H cells were allowed to attach to peptides coated in 96-well microtiter plates. Four peptides, ikmviswk (closed circle)- ikmvisw (open circle)- kmviswk (closed triangle)- kmvisw (open triangle), were used over a concentration range of 0 to 400µM. The number of cells attached after 90 minutes was determined by absorbance at 570 nm. Data are the mean triplicates. Error bars = SD.

90

Figure 24. The minimal element mediating cell adhesion to immobilized HYD1 is xkmvixw. (A) The DU-H cells were allowed to attach to 100µM of peptide coated in 96-well microtiter plates. Alanine-substitution peptides were generated from the parent peptide (ikmvisw) that was derived from HYD1 (kikmviswkg). The number of cells attached after 90 minutes was determined by absorbance at 570 nm. Data are the mean triplicates. Error bars = SD. (B) Table comparing HYD1 sequence and type of amino acids with other cell adhesion peptides obtained with a combinatorial screening approach [82].

91 Analysis of the Minimal Element of HYD1 Necessary for Activation of ERK

HYD1, when introduced as a soluble ligand, completely blocks random hapotaxis

on laminin-5 (Figure 13). Enhanced cellular signaling was coupled to the loss of cell

motility (Figure 20 and 21). HYD1 induces activation of ERK that is maximal at 10 min

post-treatment (Figure 21D). Using this signal as an indicator for bioactivity of HYD1,

we tested the truncated and alanine-substituted mutants of HYD1 for their ability to

activate ERK. Each peptide was used at a concentration of 50µM. Activation of ERK at

10 min post-treatment was conserved with alanine-substitution only at the N-terminal

lysine and C-terminal glycine (Figure 25). All other alanine-substitution resulted in

partial or complete loss of ERK activation, suggesting that the interior residues,

ikmviswk, are critical.

Deletion analysis of HYD1 revealed that both N- and C-terminal regions are

involved in activation of ERK (Figure 26). Specifically, the N-terminal 5 amino acid

residues, kikvm, and the C-terminal 5 amino acid residues, iswkg, alone induced partial

activation of ERK. The N-terminal region, kikvm, induced a stronger signal than iswkg.

In addition, some of the truncated mutants were able to partially activate ERK (Figure

26), however the amount of activation was substantially reduced in comparison to the

full-length peptide. In addition, mixing the two terminal regions of HYD1, kikmv and

iswkg, enhanced ERK activation compared to the response of these two peptides alone

(data not shown). Taken together, these data show that both the N- and C-terminal

regions of HYD1 contain an element that can induce ERK activation while alanine-

substitution

92

Figure 25. xikmviswkx is the minimal element of HYD1 required to activate ERK. PC3N cells were placed on laminin-5 coated tissue culture plates in the presence of serum-free media for 1 hour followed by the addition of HYD1 (kikmviswkg), alanine-substitution mutants of HYD1, or HYDS (wiksmkivkg) at a concentration of 50µM in serum-free media for 10 minutes. Cell lysates were analyzed for phosphorylation of ERK Thr-202/Tyr-204. Blots were stripped and reprobed for total ERK.

93

Figure 26. HYD1 induced ERK activation is mediated by N- and C-terminal regions of the peptide. PC3N cells were placed on laminin-5 coated tissue culture plates in the presence of serum-free media for 1 hour followed by the addition of HYD1 (kikmviswkg), truncated mutants of HYD1, or HYDS (wiksmkivkg) at a concentration of 50µM in serum-free media for 10 minutes. Cell lysates were analyzed for phosphorylation of ERK Thr-202/Tyr-204. Blots were stripped and reprobed for total ERK.

94 analysis of HYD1 suggests that the critical amino acid residues mediating this response

are xikmviswkx.

Analysis of the Minimal Element of HYD1 Necessary to Block Cell Migration on

Laminin-5

HYD1 completely blocks random haptotaxis on laminin-5 (Figure 13). This

response to the peptide is independent of disrupting adhesion to laminin-5 since the cells

were adhered to the ligand before treatment with the peptide. A wound-healing assay

was used to investigate the minimal element of HYD1 necessary to inhibit cell migration.

Alanine-substitution analysis of HYD1 revealed that the N-terminal lysine and the final

two C-terminal residues, a lysine and glycine, were not critical amino acids (Figure 27).

All other alanine-substitution mutants resulted in loss of peptide activity. These data

suggest that the active sequence for blocking cell migration is xikmviswxx.

Analysis using the truncated mutants of HYD1 showed that minimal deletion

from either end of the peptide resulted in at least partial loss of activity (Figure 28).

Using the control peptide, HYDS, to indicate 100% cell migration into the scratch,

several peptides including, ikmviswkg- kmviswkg- kikmviswk, were effective at

blocking cell migration by approximately 50%. These data are consistent with the results

found in the adhesion and signaling assays in that both the N- and C-terminal regions of

HYD1 are responsible for its bioactivity. Taken together, these data show that the

minimal element necessary to block cell migration on laminin-5 is xikmviswxx while

95

Figure 27. xikmviswxx is the minimal element necessary to block cell migration on laminin-5. PC3N cells were grown to a monolayer on laminin-5. Cells were treated with HYD1 (kikmviswkg), alanine-substitution mutants of HYD1, or HYDS (wiksmkivkg) at a concentration of 50µM in the presence of 1% FBS for 12 hours following the scratch. Data represent the percent of cells in a microscopic field migrating into the scratch. Three fields were analyzed per sample. Error bars = SD.

96

Figure 28. HYD1 inhibition of cell migration on laminin-5 is mediated by N- and C-terminal regions of the peptide. PC3N cells were grown to a monolayer on laminin-5. Cells were treated with HYD1 (kikmviswkg), truncated mutants of HYD1, or HYDS (wiksmkivkg) at a concentration of 50µM in the presence of 1% FBS for 12 hours following the scratch. Data represent the percent of cells in a microscopic field migrating into the scratch. Three fields were analyzed per sample. Error bars = SD.

97 loss of amino acids from either end will attenuate activity.

Discussion

Our previous data has characterized the cell adhesion peptide, HYD1

(kikmviswkg), as a bioactive peptide. Specifically, when immobilized, HYD1 acts as an

adhesion agonist by supporting tumor cell adhesion [82]. In addition, when introduced in

soluble form to prostate tumor cells adhered to laminin-5, HYD1 completely blocked

random haptotaxis (Figure 13). Coupled to the loss of cell motility, HYD1 enhanced cell

signaling on laminin-5 (Figure 20) resulting in transient activation of ERK (Figure 21).

The objective of this study was to determine the minimal element within the linear 10

amino acid structure of HYD1 that mediates these biological activities. We utilized both

N- and C-terminal deletion mutants of HYD1 in addition to alanine substitution analysis

of the peptide. Table 1 summarizes the adhesion, signaling, and anti-migratory effects of

these peptides.

Analysis of the peptide mutants suggests the minimal element necessary to

support prostate tumor cell adhesion was ikmvisw, however, we found that deletion of

amino acids from both the N- and C-terminal regions of the peptide severely attenuated

its ability to activate ERK and block cell migration on laminin-5. Alanine-substitution

revealed that xikmviswxx was the minimal element necessary to induce ERK activation

and block cell migration. This is consistent with the idea that the bioactive regions of

HYD1 could be sensitive to structural changes produced with amino acid deletion when

98

Table 1. Alanine-substituted and N- and C-terminal truncated peptides of HYD1 with their cell adhesion, cell signaling, and anti-migratory activity. Biological activity was scored on the following subjective scale: + +, activity comparable to HYD1; +, activity apparent but weaker; -, no activity.

99 HYD1 is used in soluble form, such as with the signaling and migration assays.

Nevertheless, the interior motif ikmvisw was critical in each biological assay tested.

A challenge to the use of peptide-based drugs is their poor bioavailability

[185,186]. Modifications to increase bioavailability and potency have been applied to the

cell adhesion peptides RGD and YIGSR with some success. These modifications include

peptide-cyclization, D-amino acid substitution, and conjugation to water-soluble

polymeric modifiers [187-189]. Other groups have made chemical modifications or used

the peptide backbone as template to create pseudo-peptide analogs with increased

stability and activity [190,191]. HYD1 is a linear peptide consisting of ten D-amino

acids. These data suggest that HYD1 may have a specific secondary structure in solution

given that its bioactivity was lost following minimal deletions from both the N- and C-

terminal ends of the peptide. Techniques such as circular dichroism and NMR can be

used to study the conformation and secondary structure of peptides [192-195]; these

techniques would be useful in determining if HYD1 has a structure/activity relationship.

In summary, the minimal element for bioactivity of the cell adhesion peptide

HYD1 (kikmviswkg) was determined. When immobilized as an adhesive substrate the

minimal element is xkmvixw. In solution, the minimal element necessary to block cell

migration and activate cell signaling through ERK is xikmviswxx. Our current focus is

testing the bioavailability and activity of HYD1 in mouse models of metastasis. HYD1

structural information in combination with these data could provide a template for the

creation of a small molecule mimetic of HYD1 that may prove useful in the clinical

arena.

100 VI. PRELIMINARY DATA: EFFECT OF THE SYNTHETIC D-

AMINO ACID PEPTIDE, HYD1 (kikmviswkg), ON INTEGRIN

LATERAL-ASSOCIATIONS

Introduction

A particularly remarkable activity of HYD1 is the ability to block cell migration

on a native extracellular matrix protein, laminin-5. Data presented has shown that the

inhibition of migration by HYD1 involves a dramatic remodeling of the actin

cytoskeleton resulting in increased stress fiber formation and actin colocalization with

cortactin at the cell membrane. In addition, HYD1 causes an altered signaling response

on laminin-5 resulting in transient activation of ERK. In Chapter III it is suggested that

HYD1 acts through a post-ligand occupancy event on the integrin that initiates the

cascade of events leading to loss of cell motility. It is possible that HYD1 interacts with

a common binding partner of α6 and α3 on the cell surface and initiates a change in

integrin function indirectly by altering the assocation of this protein with α6 or α3.

Integrins interact directly with other transmembrane proteins such as tetraspanins and

members of the immunoglobulin superfamily of proteins [196]. Integrin lateral-

association with these proteins can dramatically influence integrin function including

adhesion, signaling, and motility [197-209]. This chapter will discuss preliminary data

exploring if HYD1 alters α6 and α3 integrin lateral-associations on the cell surface.

101

Several studies have shown functional lateral-associations with the tetraspanin

CD151 and integrins [202-204,207,208]. CD151 forms stable associations with α3β1

and α6-containing integrins [206,210,211]. Specifically, these studies have shown that

association of CD151 with these integrins contributes to cell motility events in vitro

[203,204] and this association also correlates to increased tumor cell motility in vivo

[202]. Although the exact mechanism of tetraspanin/integrin regulation of cell motility

remains unclear, it seems likely that tetraspanin/integrin association is important in

integrin-dependent signaling events that lead to cell migration [206,212].

Another potential tetraspanin important in integrin-mediated cell migration is

CD81. Like CD151, CD81 associates with α3 and α6-containing integrins [211] and has

been shown to contribute to integrin-dependent migration events in vitro [203,213]. In

addition, a member of the immunoglobulin superfamily of proteins, EWI2, associates

with the tetraspanins CD81 and CD9 [214,215] and can regulate α3β1 dependent

functions on laminin-5 [198]. These data show that integrin function can be regulated

through primary or secondary interactions on the cell surface.

Preliminary data has shown that HYD1 interacts with specific integrin/tetraspanin

complexes on the cell surface. PC3N cells on laminin-5 treated with HYD1 had an

increase in the amount of CD81 associated with α6, however it had no effect on the

amount of CD151 associated. Biotinylation studies revealed that HYD1 induces the

association of a 60kD protein with α6 and α3-containing integrins as well as the

tetraspanin CD81. These data suggest that HYD1 changes the cell surface associations

with α6 and α3-containing integrins.

102 Results

HYD1 Interacts with CD151/Integrin Complexes

Our previous data showed that HYD1 interacted with α6 and α3 integrin subunits

(Figure 29A and B). To define receptor specificity, the peptides were incubated with

membrane preparations from DU-145H prostate carcinoma cells and the resulting

precipitate was analyzed for integrin content. Since α6 and α3-containing integrins form

stable associations with the tetraspanin CD151, [206,210,211] we wanted to determine if

the peptide was interacting with integrin/CD151 complexes. HYD1 depleted membrane

preparations of CD151 associated with α3 and α6 integrins, (Figure 29C and D)

suggesting that HYD1 targets integrin/tetraspainin complexes.

HYD1 Increases CD81 Association with the αααα6 Integrin

As mentioned above, integrin association with specific tetraspanin proteins can

effect multiple aspects of integrin function including adhesion, signaling, and motility

[197-209]. Association with the tetraspanins CD151 and CD81 have specifically been

shown to influence cell motility [203,213]. Given that HYD1 interacts with

integrin/tetraspanin complexes, we hypothesized that HYD1 may alter

integrin/tetraspanin association and thus provide an initiating event to the blockade of cell

motility on laminin-5. PC3N cells on laminin-5 were treated with HYD1 or the

scrambled derivative, HYDS, for 10 and 30 minutes before cell lysis. The samples were

immunoprecipitated for CD151 and CD81 and analyzed for α6 and α3 integrin content.

103

Figure 29. HYD1 interacts with CD151/Integrin Complexes. Affinity precipitation reactions were performed with HYD1, RZ3, or HYDS peptides on DU-145H cell membrane fractions as described in Experimental Procedures. (A) The precipitates were analyzed for α6 or (B) α3 integrin by SDS-PAGE electrophoresis and western blot. ML (membrane lysate) was loaded as a control. Supernatants from the affinity precipitation reactions in A and B were then used in an immunoprecipitation reaction for the tetraspanin CD151 (C) and (D) and blotted for α6 and α3 respectively. ML (membrane lysate) was used for the control immunoprecipitation reactions.

104

Figure 30. HYD1 does not change the amount of CD151 associated with αααα6 or αααα3-containing integrins. PC3N cells were placed on laminin-5 coated tissue culture plates in the presence of serum-free media for 1 hour followed by the addition of the peptides, HYD1 or HYDS (75µg/ml), in serum-free media for 10 and 30 minutes. Samples were immunoprecipitated (Brij 96) for α6 (J1B5) and CD151 (5C11) overnight at 4oC. The precipitates were analyzed for α6 by western blot. The blot was stripped and reprobed for α3.

105

HYD1 did not change the amount of CD151 associated with either α6 or α3 compared to

the control peptide HYDS (Figure 30). However, after 30 minutes of treatment, HYD1

caused an increase in the amount of CD81 associated with α6 and decreased the amount

associated with α3 (Figure 31). HYD1 induced clustering of α6 on laminin-5 (Figure

32), however, this was only seen in 2-3 cells/microscopic field. In addition, α6 did not

colocalize with CD81 at these adhesion sites.

HYD1 Alters the Lateral-Associations of αααα6 and αααα3-Integrins and the Tetraspanin

CD81

To determine if HYD1 disrupted or induced other changes in integrin associations

on the cell surface, we biotinylated PC3N cells on laminin-5 that were treated with

HYD1 or the scrambled derivative, HYDS, for 30 minutes. The samples were

immunoprecipitated for α6 and α3 integrins in addition to the tetraspanin CD81. HYD1

treatment induced a protein at approximately 60kD to co-immunoprecipitate with α6 and

α3 integrins, as well as CD81 (Figure 33). These data suggest that HYD1 changes the

cell-surface protein associations of α6 and α3-containing integrins and the tetraspanin

CD81.

Discussion

Integrin lateral-associations with tetraspanins and other cell surface molecules can

influence integrin function. Specifically, the tetraspanins CD151 and CD81 have been

106

Figure 31. HYD1 increases the amount of CD81 associated with αααα6-integrins and decreases the amount of CD81 associated with αααα3-integrins. PC3N cells were placed on laminin-5 coated tissue culture plates in the presence of serum-free media for 1 hour followed by the addition of the peptides, HYD1 or HYDS (75µg/ml), in serum-free media or serum-free media alone for 30 minutes. Samples were immunoprecipitated (Brij 96) for CD81 (JS64) overnight at 4oC. The precipitates were analyzed for α6 and α3 by western blot. WCL = whole cell lysate.

107

Figure 32. HYD1 induces αααα6 integrin clustering. PC3N cells were placed on laminin-5 coated coverslips for 1 hour followed by addition of HYD1 or HYDS (75µg/ml) in serum-free media for 30 minutes. Coverslips were fixed and stained for CD81 (JS64) (green) and Alpha-6 Integrin (J1B5) (red) as described in Experimental Procedures.

108

Figure 33. HYD1 alters the lateral-associations of αααα6 and αααα3-containing integrins and the tetraspanin CD81. PC3N cells were adhered to laminin-5 for 1 hour followed by addition of HYD1 or HYDS (75µg/ml) in serum-free media for 30 minutes. Cells were biotinylated before immunoprecipitation for α6 (J1B5), α3 (P1B5), and CD81 (JS64) as described in materials and methods. HYD1 induced a protein at approximately 60kD to co-immunoprecipitate with α6, and α3 integrins as well as CD81.

109 shown to have crucial roles in cell motility through interaction with integrins

[203,204,213]. Here we explored if the anti-migratory effect of HYD1 on laminin-5 was

dependent on disrupting or altering α6 or α3 associations with these tetraspanins. HYD1

was shown to interact with integrin complexes containing CD151, however HYD1 did

not change the amount of CD151 associated with α6 or α3-containing integrins.

Interestingly, HYD1 did alter integrin association with the tetraspanin CD81. Peptide

treatment for 30 minutes induced increased CD81 association with α6 while reducing the

amount associated with α3. It is tempting to speculate that a shift in the amount of CD81

interacting with α6 or α3 could be an initiating step in the loss of cell motility induced by

HYD1.

There are several possibilities that could explain how a change in

integrin/tetraspainin interaction could have such a profound effect on cell motility. First,

tetraspanins have been shown to modulate integrin-dependent signaling events such as

focal adhesion kinase activation that lead to cell migration [206,212]. In addition,

integrin/tetraspanin association has been shown to recruit signaling molecules such as

phosphatidylinositol 4-kinase [204] and protein kinase C [209]. It is possible that an

increase in CD81 association with α6 recruits a signaling complex that initiates a

signaling cascade that functionally blocks cell motility. Although most data show that

CD81/integrin interaction is a pro-migratory association [203,213], it is possible that a

pro-migratory signaling complex is recruited to α3β1 while an inhibitory complex is

recruited to α6-containing integrins after association with CD81. This is consistent with

110 the data that HYD1 reduces association of CD81 with α3-containing integrins while

increasing its association with α6.

A second possibility is that HYD1 induces a change in the protein content of the

tetraspanin protein microdomain (tetraspanin-web) associated with the integrin. This

could, as stated above, recruit specific signaling complex that could alter integrin

function. A group recently reported that EWI2, a cell surface protein of the

immunoglobulin superfamily and a major CD81 and CD9 partner [214], can regulate

α3β1 functions on laminin-5 [198]. In this study, overexpression of EWI2 suppressed

cell migration on laminin-5. The association of EWI2 with α3β1 was mediated through

the tetraspanins CD81 and CD9. This is particularly intriguing due to the fact that HYD1

induced an additional protein of about 60kD to associate with α6, α3, and CD81. The

approximate molecular weight of EWI2 is 55-70kD. These data suggest that HYD1

induces a change in the tetraspanin-web associated with α6 and α3-containing integrins.

Whether the new associative protein is EWI2 and if this new association is coupled to the

loss of motility is to be determined.

111 VII. CONCLUDING STATEMENTS

Cancer is a national health concern for men and women worldwide. Although

methods for diagnosis and treatment have improved with technological innovations,

cancer mortality rates remain significant. Metastases, rather than the primary tumor,

accounts for most cancer related deaths [1,7]. There is an intense need for new

therapeutics that target the processes involved in metastasis given that cancer diagnosis

often occurs at a relatively late stage in the disease. In order to develop novel

therapeutics, research must focus on determining essential molecules involved in

metastasis. This dissertation focused on the development and biological properties of

syntheic cell adhesion peptides. Using a combinatorial method, peptides were selected to

target α6 integrins. These peptide ligand mimetics were used to explore integrin biology

of cell adhesion and migration. This study has produced a novel integrin-targeting

peptide, HYD1 (kikmviswkg) that is capable of blocking both tumor cell adhesion to

extracellular matrices and cell migration of tumor cells adhered to laminin-5. These in

vitro data provide rationale to determine the activity of this peptide in animal models of

metastasis.

The biological activity of HYD1 characterizes it as having both agonist and

antagonist effects on integrin functions. Since the combinatorial screen was based on

functional cell ahesion, it is logical that the peptide works as an adhesion agonist. Two

peptides, RZ3 and HYD1, could both support tumor cell adhesion and inhibit adhesion to

extracellular matrix proteins. In addition, as an integrin adhesion ligand, a true agonist

112 would promote other integrin-mediated functions such as signal transduction, leading to

cell spreading and motility. However, HYD1 uncoupled integrin-mediated cell adhesion

from cell spreading and migratory events. The most likely cause is due to the fact that

the peptide did not initiate the identical signaling program as the native extracellular

matrix. Therefore, when immobilized, HYD1 is not sufficient as agonist for all integrin

functions. One explanation could be that the immobilized peptide lacks the proper

avidity for integrin activation. Alternatively, HYD1 could initiate a signaling response

that antagonizes particular integrin functions. It would be interesting to test if cell

spreading and migration would occur with cell adhesion to multivalent forms of HYD1.

HYD1 had antagonistic effects when introduced in soluble form to cells adhered

to laminin-5. Specifically, HYD1 blocked tumor cell random haptotaxis and invasion.

The initiating event for this response remains unknown, however the effects on

cytoskeletal reorganization and cell signaling mirrors what has been described for

disintegin blockade of cell migration [175-179]. This suggests that HYD1 has a direct

effect on integrins. Further study is needed to solidify the mechanism for HYD1

inhibition of cell motility.

A couple of observations provide insight into the mechanism of action for HYD1.

First, the activity of HYD1 is a post-ligand occupancy event; the loss of cell motility

induced by HYD1 is independent of disrupting cell adhesion to the native ligand. Indeed,

following treatment with HYD1, cells remain adhered and retain a “spread” morphology.

This is consistent with the idea that the peptide does not compete for the ligand binding

sites with laminin-5, suggesting that the peptide interacts with integrins at a distinct site.

113 It is reinforced by the fact that, unlike adhesion to native ECM proteins such as laminin-5

and Fibronectin, tumor cell adhesion to immobized HYD1 is not affected by the presence

of EDTA.

Integrin function is intimately linked to the ability to modulate receptor structure

[33,216]. It has been documented that integrin affinity states are modulated whether the

receptor is in a ligand-bound, primed, or inactive confirmation [32,33,216-218]. A major

tool to study the link between receptor shape and activity is conformational-dependent

monoclonal antibodies. Antibodies have been identified that modulate the ability of

integrins to bind ligand or recognize epitopes that represent active confirmations of

integrins [219-221]. The mechanism of the former is thought to occur through

stabilization of ligand-bound or unbound integrin confirmations following antibody

exposure. It is tempting to speculate that HYD1 interaction with integrins causes a

conformational change that is integrated into an anti-migratory confirmation.

Conformational-dependent antibodies may prove useful to identify if HYD1 acitivty is

linked to integrin conformational shifts.

Chapter V. introduced the importance of integrin lateral-associations in mediating

integrin functions including adhesion, signaling, and motility [197-209]. An alternative

hypothesis should include that HYD1 disrupts or changes the protein milleu in the

integrin-associated tetraspanin web. Any such change could maintain integrin adhesion

while blocking cell motility through initiation of an inhibitory signal. This hypothesis

might also explain potential promiscuity of HYD1 in blocking cell migration on different

matrices. Preliminary data has shown that HYD1 is equally effective in blocking cell

114 motility of HT-1080 cells on Fibronectin (data not shown). This would seem confusing

since the peptides were screened for specificity to Laminin-receptors, however it is

possible that HYD1 interacts at a conserved site on alpha-subunits that is crucial in

mediating integrin lateral-associations. In this case, HYD1 would be expected to have

anti-migratory activity on a wide array of matrix proteins. This phenomenon could also

be explained by integrin cross-talk, meaning that HYD1 ligation of Laminin receptors

functionally regulates other integrin receptors through “inside-out” signaling.

A second observation is that although random haptotaxis is completely blocked,

cell surfaces remain active after treatment with HYD1. This is consistent with the data

that HYD1 induces actin cytoskeletal reorganization and cell signaling through focal

adhesion kinase and the extracellular signal-regulated protein kinase pathway. Focal

adhesion and actin dynamics are crucial regulators of cell motility [56], with integrins

serving as the molecular “feet” of a cell and the site of formation for adhesion complexes

[222]. Since membrane ruffling occurs yet existing adhesions seem to remain static,

HYD1 could influence the formation and/or disassembly of focal adhesions.

Focal adhesion dymanics is an essential part of cell locomotion by a reiterative

process involving the formation of new matrix adhesions at the leading edge coupled

with focal adhesion disassembly and retraction at the trailing end [56,222]. Observation

of cells treated with HYD1 suggests two possibilities. First, tumor cells treated with

HYD1 have active membrane surfaces, including membrane ruffling, yet the cells do not

seem to create viable adhesion sites from these protrusive events. This suggests a defect

in focal complex assembly at the leading edge or an inability of focal complexes to

115 mature into focal adhesions. Using live-cell imaging and flourescently labeled focal

adhesion components, a recent study has determined the rate constants for focal adhesion

disassembly [223]. Application of this technique would offer valuable insight as to

whether HYD1-treated cells disrupted the proper kinetics of adhesion assembly at the

leading edge. In addition, another group has characterized the adhesion protein zyxin as

a definitive marker for a mature focal adhesion [224]. Analysis of zyxin incorporation to

adhesions proximal to the leading edge would help determine if the defect was linked to

adhesion maturation.

Tumor cells treated with HYD1 do not seem to reorganize existing adhesions that

tether the cell body to the matrix. This suggests a loss in focal adhesion turnover

resulting in static adhesion sites. Focal ashesion disassembly is linked to the activity of

many signaling proteins and proteases [56]. The most direct approach to study the

regulation of mature focal adhesions is by applying the technology described above to

observe the kinetics of adhesion proteins with live-cell imaging. A significant change in

the half-life of mature adhesions, that is those distinct from the leading edge, would

suggest that HYD1 induces a defect in adhesion remodeling. Figure 34 is a schematic

representing the possible mechanisms for the antimigratory activity of HYD1.

Cancer therapeutics has entered an era of molecular targeting. New agents are

being developed to target specific molecular components important to tumor progression

and survival. Signal transduction pathways such as the extracellular signal-regulated

protein kinase/mitogein-activated protein kinase (ERK/MAPK) pathway have shown to

116

Figure 34. Potential mechanisms for the anti-migratory activity of HYD1. This model illustrates the hypotheses that HYD1 may influence integrin lateral-associations and/or focal adhesion dynamics to prevent cell migration.

117 be important for the pathogenesis of several cancers and inhibitors to specific molecules

in this pathway have been developed and are in clinical trials [225,226]. Other agents

have been developed for specific abnormalities. STI-571, an inhibitor of BCR-ABL

tyrosine kinase, is used to treat chronic myeloid leukemia [227,228]. Monoclonal

antibody therapy has also been employed with some success. Rituxan, an anti-CD-20

monoclonal antibody, has been approved for the treatment of B-cell lymphoma

[229,230]. Herceptin, a monoclonal antibody to the HER2-neu receptor has been

effective in treating certain types of breast cancer [231,232]. It is conceivable to imagine

that cancer therapy will evolve into individualized therapy, treatment regimens will be

designed based on the molecular characteristics of individual tumors.

Integrins are attractive as a therapeutic target because they are sentinel molecules

for a plethora of cellular functions critical to cancer progression [22,32]. This

characteristic is particularly appealing in regard to cancer metastasis because integrins

serve directly as a mediator for the adhesion events necessary in metastasis while also

integrating information from the extracellular environment into cellular signals that are

required for cell motility and survival at distant sites in the body. There are currently

several integrin inhibitors under investigation for cancer therapy. Antagonists to αvβ3

and αvβ5 have been developed as antibody and peptide-based inhibitors to target

angiogenesis and metastasis [233,234]. Vitaxin, a humanized αvβ3 antibody, is currently

in Phase II trials [235-238]. Cilengitide, a peptide inhibitor of αvβ3/αvβ5, is in Phase II

trial for advanced solid tumors and has shown promise for use in combination therapy

[239-242]. In addition, other αvβ3 and α5β1-blocking peptides have been documented

118 but have not yet entered clinical trials [243,244]. A humanized antibody has also been

developed for α5β1 integrin to target angiogenesis and is entering a Phase I trial

(www.pdl.com). Although the current integrin-based therapeutics are promising, they are

focused on αv and α5 integrin pairs. Antagonists to other integrins, specifically the

laminin receptors, may prove to be equally beneficial.

The integrins α6β1, α3β1, and α6β4 are laminin receptors [110,111]. These

integrins are associated with epithelial tumor progression in prostate, breast, colon,

pancreatic carcinomas, head and neck tumors, and melanoma [23,112-115,245].

Combinatorial screening, such as the one-bead-one-compound approach, has been

successful in identifying and characterizing cell adhesion peptides [84] with potential

antagonistic effects on the targeted cell surface receptor. An argument against the use of

peptide-based drugs is their poor bioavailability [185,186,234]. However, modifications

to increase bioavailability and potency have been applied to the cell adhesion peptides

RGD and YIGSR with some success [187-189]. In addition, using the peptide backbone

as a template, chemical modifications can create pseudo-peptide analogs with increased

stability and activity [190,191]. The studies shown in chapter IV should aid in the

modifcation of HYD1 into a small molecule mimetic, increasing its potential use as a

novel drug.

The work presented in this dissertation supports the use of synthetic cell adhesion

peptides as a means to study cell biology and develop novel therapeutics. The priority for

further development of HYD1 is determining in vivo activity. Both spontaneous and

experimental mouse models of metastasis should be employed to identify if the peptide

119 has preferential activity to early or late stages in metastasis [1,246]. After determining

therapeutic potential by these measures, studies of bioavailability will help clarify if

peptide modification is necessary before testing in the clinical arena.

120 REFERENCES

1. Chambers, A.F., Groom, A.C. and MacDonald, I.C. (2002) Dissemination and growth of cancer cells in metastatic sites. Nat Rev Cancer, 2, 563-72.

2. Fidler, I.J. (2003) The pathogenesis of cancer metastasis: the 'seed and soil'

hypothesis revisited. Nat Rev Cancer, 3, 453-8. 3. Fidler, I.J. (1999) Critical determinants of cancer metastasis: rationale for therapy.

Cancer Chemother Pharmacol, 43 Suppl, S3-10. 4. Chambers, A.F., Naumov, G.N., Varghese, H.J., Nadkarni, K.V., MacDonald, I.C.

and Groom, A.C. (2001) Critical steps in hematogenous metastasis: an overview. Surg Oncol Clin N Am, 10, 243-55, vii.

5. Woodhouse, E.C., Chuaqui, R.F. and Liotta, L.A. (1997) General mechanisms of

metastasis. Cancer, 80, 1529-37. 6. Folkman, J. (1992) The role of angiogenesis in tumor growth. Semin Cancer Biol,

3, 65-71. 7. Chambers, A.F., MacDonald, I.C., Schmidt, E.E., Morris, V.L. and Groom, A.C.

(2000) Clinical targets for anti-metastasis therapy. Advances in Cancer Research,79, 91-121.

8. Society, A.C. (2005) Cancer Facts and Figures 2005. American Cancer Society

Atlanta, Ga.

9. Routh, J.C. and Leibovich, B.C. (2005) Adenocarcinoma of the prostate: epidemiological trends, screening, diagnosis, and surgical management of

localized disease. Mayo Clin Proc, 80, 899-907. 10. Parkin, D.M., Bray, F., Ferlay, J. and Pisani, P. (2005) Global cancer statistics,

2002. CA Cancer J Clin, 55, 74-108. 11. Hankey, B.F., Feuer, E.J., Clegg, L.X., Hayes, R.B., Legler, J.M., Prorok, P.C.,

Ries, L.A., Merrill, R.M. and Kaplan, R.S. (1999) Cancer surveillance series: interpreting trends in prostate cancer--part I: Evidence of the effects of screening in recent prostate cancer incidence, mortality, and survival rates. J Natl Cancer Inst, 91, 1017-24.

121 12. Johansson, J.E., Holmberg, L., Johansson, S., Bergstrom, R. and Adami, H.O.

(1997) Fifteen-year survival in prostate cancer. A prospective, population-based study in Sweden. Jama, 277, 467-71.

13. Chodak, G.W., Thisted, R.A., Gerber, G.S., Johansson, J.E., Adolfsson, J., Jones,

G.W., Chisholm, G.D., Moskovitz, B., Livne, P.M. and Warner, J. (1994) Results of conservative management of clinically localized prostate cancer. N Engl J Med,330, 242-8.

14. Adolfsson, J., Steineck, G. and Hedlund, P.O. (1997) Deferred treatment of

clinically localized low-grade prostate cancer: actual 10-year and projected 15-year follow-up of the Karolinska series. Urology, 50, 722-6.

15. Johansson, J.E., Andren, O., Andersson, S.O., Dickman, P.W., Holmberg, L.,

Magnuson, A. and Adami, H.O. (2004) Natural history of early, localized prostate cancer. Jama, 291, 2713-9.

16. Banyard, J. and Zetter, B.R. (1998) The role of cell motility in prostate cancer.

Cancer Metastasis Rev, 17, 449-58. 17. Ozen, M. and Pathak, S. (2000) Genetic alterations in human prostate cancer: a

review of current literature. Anticancer Res, 20, 1905-12. 18. Pilat, M.J., Kamradt, J.M. and Pienta, K.J. (1998) Hormone resistance in prostate

cancer. Cancer Metastasis Rev, 17, 373-81. 19. Dong, J.T., Isaacs, W.B. and Isaacs, J.T. (1997) Molecular advances in prostate

cancer. Curr Opin Oncol, 9, 101-7. 20. Hynes, R.O. (1992) Integrins: versatility, modulation, and signaling in cell

adhesion. Cell, 69, 11-25. 21. Hood, J.D. and Cheresh, D.A. (2002) Role of integrins in cell invasion and

migration. Nat Rev Cancer, 2, 91-100. 22. Jin, H. and Varner, J. (2004) Integrins: roles in cancer development and as

treatment targets. Br J Cancer, 90, 561-5. 23. Cress, A.E., Rabinovitz, I., Zhu, W. and Nagle, R.B. (1995) The alpha 6 beta 1

and alpha 6 beta 4 integrins in human prostate cancer progression. Cancer Metastasis Rev, 14, 219-28.

122 24. Nagle, R.B., Hao, J., Knox, J.D., Dalkin, B.L., Clark, V. and Cress, A.E. (1995)

Expression of hemidesmosomal and extracellular matrix proteins by normal and malignant human prostate tissue. Am J Pathol, 146, 1498-507.

25. Fornaro, M., Tallini, G., Bofetiado, C.J., Bosari, S. and Languino, L.R. (1996)

Down-regulation of beta 1C integrin, an inhibitor of cell proliferation, in prostate carcinoma. Am J Pathol, 149, 765-73.

26. Perlino, E., Lovecchio, M., Vacca, R.A., Fornaro, M., Moro, L., Ditonno, P.,

Battaglia, M., Selvaggi, F.P., Mastropasqua, M.G., Bufo, P. and Languino, L.R. (2000) Regulation of mRNA and protein levels of beta1 integrin variants in human prostate carcinoma. Am J Pathol, 157, 1727-34.

27. Putz, E., Witter, K., Offner, S., Stosiek, P., Zippelius, A., Johnson, J., Zahn, R.,

Riethmuller, G. and Pantel, K. (1999) Phenotypic characteristics of cell lines derived from disseminated cancer cells in bone marrow of patients with solid epithelial tumors: establishment of working models for human micrometastases. Cancer Res, 59, 241-8.

28. Hassan, M.O. and Maksem, J. (1980) The prostatic perineural space and its

relation to tumor spread: an ultrastructural study. Am J Surg Pathol, 4, 143-8. 29. Villers, A., McNeal, J.E., Redwine, E.A., Freiha, F.S. and Stamey, T.A. (1989)

The role of perineural space invasion in the local spread of prostatic adenocarcinoma. J Urol, 142, 763-8.

30. Stephens, H., Bendayan, M. and Gisiger, V. (1985) Simultaneous labelling of

basal lamina components and acetylcholinesterase at the neuromuscular junction. Histochem J, 17, 1203-20.

31. Haraida, S., Nerlich, A.G., Bise, K., Wiest, I. and Schleicher, E. (1992)

Comparison of various basement membrane components in benign and malignant peripheral nerve tumours. Virchows Arch A Pathol Anat Histopathol, 421, 331-8.

32. Hynes, R. (2002) Integrins: bidirectional, allosteric signaling machines. Cell, 110,

673-87. 33. Liddington, R.C. (2002) Integrin Activation Takes Shape. Journal of Cell

Biology, 158, 833-839. 34. Giancotti, F.G. and Ruoslahti, E. (1999) Integrin signaling. Science, 285, 1028-

32.

123 35. Green, L.J., Mould, A.P. and Humphries, M.J. (1998) The integrin beta subunit.

International Journal of Biochemistry & Cell Biology, 30, 179-84. 36. Hemler, M. (1999) Integrin Guidebook. Oxford Publishing. 37. Adams, J.C. (2002) Regulation of protrusive and contractile cell-matrix contacts.

Journal of Cell Science, 115, 257-65. 38. Wehrle-Haller, B. (2002) The Inner Lives of Focal Adhesions. Trends in Cell

Biology, 12, 382-389. 39. Xiong, J.P., Stehle, T., Diefenbach, B., Zhang, R., Dunker, R., Scott, D.L.,

Joachimiak, A., Goodman, S.L. and Arnaout, M.A. (2001) Crystal structure of the extracellular segment of integrin alpha Vbeta3. [see comments.]. Science, 294,339-45.

40. O'Toole, T.E., Mandelman, D., Forsyth, J., Shattil, S.J., Plow, E.F. and Ginsberg,

M.H. (1991) Modulation of the affinity of integrin alpha IIb beta 3 (GPIIb-IIIa) by the cytoplasmic domain of alpha IIb. Science, 254, 845-7.

41. Hemler, M.E. (1998) Integrin associated proteins. Current Opinion in Cell

Biology, 10, 578-85. 42. Schwartz, M.A. (2001) Integrin signaling revisited. Trends in Cell Biology, 11,

466-70. 43. Horwitz, A.R. and Parsons, J.T. (1999) Cell migration--movin' on. [letter;

comment.]. Science, 286, 1102-3. 44. Boudreau, N. and Bissell, M.J. (1998) Extracellular matrix signaling: integration

of form and function in normal and malignant cells. Current Opinion in Cell Biology, 10, 640-6.

45. Ruiz, P., Dunon, D., Sonnenberg, A. and Imhof, B.A. (1993) Suppression of

mouse melanoma metastasis by EA-1, a monoclonal antibody specific for alpha 6 integrins. [erratum appears in Cell Adhes Commun 1993 Sep;1(2):following 190.]. Cell Adhesion & Communication, 1, 67-81.

46. Parsons, J.T., Martin, K.H., Slack, J.K., Taylor, J.M. and Weed, S.A. (2000) Focal

adhesion kinase: a regulator of focal adhesion dynamics and cell movement. Oncogene, 19, 5606-13.

47. Small, J.V. (2002) The lamellipodium: where motility begins. Trends in Cell

Biology, 12, 112-120.

124 48. Ridley, A.J. (2001) Rho family proteins: coordinating cell responses. Trends in

Cell Biology, 11, 471-7. 49. Zamir, E., Katz, M., Posen, Y., Erez, N., Yamada, K.M., Katz, B.Z., Lin, S., Lin,

D.C., Bershadsky, A., Kam, Z. and Geiger, B. (2000) Dynamics and segregation of cell-matrix adhesions in cultured fibroblasts. Nature Cell Biology, 2, 191-6.

50. Nobes, C.D. and Hall, A. (1995) Rho, rac, and cdc42 GTPases regulate the

assembly of multimolecular focal complexes associated with actin stress fibers, lamellipodia, and filopodia. Cell, 81, 53-62.

51. Rottner, K., Behrendt, B., Small, J.V. and Wehland, J. (1999) VASP dynamics

during lamellipodia protrusion. Nature Cell Biology, 1, 321-2. 52. Pantaloni, D., Boujemaa, R., Didry, D., Gounon, P. and Carlier, M.F. (2000) The

Arp2/3 complex branches filament barbed ends: functional antagonism with capping proteins. Nature Cell Biology, 2, 385-91.

53. Blanchoin, L., Amann, K.J., Higgs, H.N., Marchand, J.B., Kaiser, D.A. and

Pollard, T.D. (2000) Direct observation of dendritic actin filament networks nucleated by Arp2/3 complex and WASP/Scar proteins. Nature, 404, 1007-11.

54. Miki, H., Suetsugu, S. and Takenawa, T. (1998) WAVE, a novel WASP-family

protein involved in actin reorganization induced by Rac. EMBO Journal, 17,6932-41.

55. Kimura, K. (1996) Regulation of myosin phosphatase by Rho and Rho-associated

kinase (Rho-kinase). [see comments.]. Science, 273, 245-8. 56. Carragher, N.O. and Frame, M.C. (2004) Focal adhesion and actin dynamics: a

place where kinases and proteases meet to promote invasion. Trends Cell Biol, 14,241-9.

57. Guan, J.L., Trevithick, J.E. and Hynes, R.O. (1991) Fibronectin/integrin

interaction induces tyrosine phosphorylation of a 120-kDa protein. Cell Regulation, 2, 951-64.

58. Calalb, M.B., Polte, T.R. and Hanks, S.K. (1995) Tyrosine phosphorylation of

focal adhesion kinase at sites in the catalytic domain regulates kinase activity: a role for Src family kinases. Molecular & Cellular Biology, 15, 954-63.

59. Schlaepfer, D.D., Hauck, C.R. and Sieg, D.J. (1999) Signaling through focal

adhesion kinase. Progress in Biophysics & Molecular Biology, 71, 435-78.

125 60. Sieg, D.J., Hauck, C.R. and Schlaepfer, D.D. (1999) Required role of focal

adhesion kinase (FAK) for integrin-stimulated cell migration. Journal of Cell Science, 112, 2677-91.

61. Cary, L.A., Chang, J.F. and Guan, J.L. (1996) Stimulation of cell migration by

overexpression of focal adhesion kinase and its association with Src and Fyn. Journal of Cell Science, 109, 1787-94.

62. Gilmore, A.P. and Romer, L.H. (1996) Inhibition of focal adhesion kinase (FAK)

signaling in focal adhesions decreases cell motility and proliferation. Molecular Biology of the Cell, 7, 1209-24.

63. Klemke, R.L., Cai, S., Giannini, A.L., Gallagher, P.J., de Lanerolle, P. and

Cheresh, D.A. (1997) Regulation of cell motility by mitogen-activated protein kinase. J Cell Biol, 137, 481-92.

64. Fincham, V.J., James, M., Frame, M.C. and Winder, S.J. (2000) Active

ERK/MAP kinase is targeted to newly forming cell-matrix adhesions by integrin engagement and v-Src. Embo J, 19, 2911-23.

65. Pawlak, G. and Helfman, D.M. (2002) MEK mediates v-Src-induced disruption of

the actin cytoskeleton via inactivation of the Rho-ROCK-LIM kinase pathway. JBiol Chem, 277, 26927-33.

66. Glading, A., Chang, P., Lauffenburger, D.A. and Wells, A. (2000) Epidermal

growth factor receptor activation of calpain is required for fibroblast motility and occurs via an ERK/MAP kinase signaling pathway. J Biol Chem, 275, 2390-8.

67. Glading, A., Bodnar, R.J., Reynolds, I.J., Shiraha, H., Satish, L., Potter, D.A.,

Blair, H.C. and Wells, A. (2004) Epidermal growth factor activates m-calpain (calpain II), at least in part, by extracellular signal-regulated kinase-mediated phosphorylation. Mol Cell Biol, 24, 2499-512.

68. Cuevas, B.D., Abell, A.N., Witowsky, J.A., Yujiri, T., Johnson, N.L., Kesavan,

K., Ware, M., Jones, P.L., Weed, S.A., DeBiasi, R.L., Oka, Y., Tyler, K.L. and Johnson, G.L. (2003) MEKK1 regulates calpain-dependent proteolysis of focal adhesion proteins for rear-end detachment of migrating fibroblasts. Embo J, 22,3346-55.

69. Liu, R., Enstrom, A.M. and Lam, K.S. (2003) Combinatorial peptide library

methods for immunobiology research. Exp Hematol, 31, 11-30.

126 70. Lam, K.S., Salmon, S.E., Hersh, E.M., Hruby, V.J., Kazmierski, W.M. and

Knapp, R.J. (1991) A new type of synthetic peptide library for identifying ligand-binding activity. Nature, 354, 82-4.

71. Lam, K.S., Lebl, M. and Krchnak, V. (1997) The "One-Bead-One-Compound"

Combinatorial Library Method. Chem Rev, 97, 411-448. 72. Cwirla, S.E., Peters, E.A., Barrett, R.W. and Dower, W.J. (1990) Peptides on

phage: a vast library of peptides for identifying ligands. Proc Natl Acad Sci U S A, 87, 6378-82.

73. Devlin, J.J., Panganiban, L.C. and Devlin, P.E. (1990) Random peptide libraries:

a source of specific protein binding molecules. Science, 249, 404-6. 74. Rodi, D.J., Makowski, L. and Kay, B.K. (2002) One from column A and two

from column B: the benefits of phage display in molecular-recognition studies. Curr Opin Chem Biol, 6, 92-6.

75. Trepel, M., Arap, W. and Pasqualini, R. (2002) In vivo phage display and

vascular heterogeneity: implications for targeted medicine. Curr Opin Chem Biol,6, 399-404.

76. Buhl, L., Szecsi, P.B., Gisselo, G.G. and Schafer-Nielsen, C. (2002) Surface

immunoglobulin on B lymphocytes as a potential target for specific peptide ligands in chronic lymphocytic leukaemia. Br J Haematol, 116, 549-54.

77. Renschler, M.F., Bhatt, R.R., Dower, W.J. and Levy, R. (1994) Synthetic peptide

ligands of the antigen binding receptor induce programmed cell death in a human B-cell lymphoma. Proc Natl Acad Sci U S A, 91, 3623-7.

78. Lam, K.S., Lou, Q., Zhao, Z.G., Smith, J., Chen, M.L., Pleshko, E. and Salmon,

S.E. (1995) Idiotype specific peptides bind to the surface immunoglobulins of two murine B-cell lymphoma lines, inducing signal transduction. Biomed Pept Proteins Nucleic Acids, 1, 205-10.

79. Assa-Munt, N., Jia, X., Laakkonen, P. and Ruoslahti, E. (2001) Solution

structures and integrin binding activities of an RGD peptide with two isomers. Biochemistry, 40, 2373-8.

80. Koivunen, E., Wang, B. and Ruoslahti, E. (1995) Phage libraries displaying cyclic

peptides with different ring sizes: ligand specificities of the RGD-directed integrins. Biotechnology (N Y), 13, 265-70.

127 81. Pennington, M.E., Lam, K.S. and Cress, A.E. (1996) The use of a combinatorial

library method to isolate human tumor cell adhesion peptides. Mol Divers, 2, 19-28.

82. DeRoock, I.B., Pennington, M.E., Sroka, T.C., Lam, K.S., Bowden, G.T., Bair,

E.L. and Cress, A.E. (2001) Synthetic peptides inhibit adhesion of human tumor cells to extracellular matrix proteins. Cancer Res, 61, 3308-13.

83. Lau, d.h., Guo, l.l, Liu, r.w., Song, a.m., Shao, c.k., Lam, k.s. (2002) Identifying

peptide ligands for cell surface receptors using cell-growth-on-bead-assay and one-bead-one-compound combinatorial library. Biotechnology Letters, 24, 497-500.

84. Aina, O.H., Marik, J., Liu, R., Lau, D.H. and Lam, K.S. (2005) Identification of

novel targeting peptides for human ovarian cancer cells using "one-bead one-compound" combinatorial libraries. Mol Cancer Ther, 4, 806-13.

85. Parmley, S.F. and Smith, G.P. (1988) Antibody-selectable filamentous fd phage

vectors: affinity purification of target genes. Gene, 73, 305-18. 86. Fukuda, M.N., Ohyama, C., Lowitz, K., Matsuo, O., Pasqualini, R., Ruoslahti, E.

and Fukuda, M. (2000) A peptide mimic of E-selectin ligand inhibits sialyl Lewis X-dependent lung colonization of tumor cells. Cancer Res, 60, 450-6.

87. Romanov, V.I., Durand, D.B. and Petrenko, V.A. (2001) Phage display selection

of peptides that affect prostate carcinoma cells attachment and invasion. Prostate,47, 239-51.

88. Zhang, J., Spring, H. and Schwab, M. (2001) Neuroblastoma tumor cell-binding

peptides identified through random peptide phage display. Cancer Lett, 171, 153-64.

89. Houghten, R.A., Pinilla, C., Blondelle, S.E., Appel, J.R., Dooley, C.T. and

Cuervo, J.H. (1991) Generation and use of synthetic peptide combinatorial libraries for basic research and drug discovery. Nature, 354, 84-6.

90. Furka, A., Sebestyen, F., Asgedom, M. and Dibo, G. (1991) General method for

rapid synthesis of multicomponent peptide mixtures. Int J Pept Protein Res, 37,487-93.

91. Hamamoto, K., Kida, Y., Zhang, Y., Shimizu, T. and Kuwano, K. (2002)

Antimicrobial activity and stability to proteolysis of small linear cationic peptides with D-amino acid substitutions. Microbiol Immunol, 46, 741-9.

128 92. Van Regenmortel, M.H. and Muller, S. (1998) D-peptides as immunogens and

diagnostic reagents. Curr Opin Biotechnol, 9, 377-82. 93. Rabinovitz, I., Nagle, R.B. and Cress, A.E. (1995) Integrin alpha 6 expression in

human prostate carcinoma cells is associated with a migratory and invasive phenotype in vitro and in vivo. Clin Exp Metastasis, 13, 481-91.

94. Tran, N.L., Nagle, R.B., Cress, A.E. and Heimark, R.L. (1999) N-Cadherin

expression in human prostate carcinoma cell lines. An epithelial-mesenchymal transformation mediating adhesion withStromal cells. Am J Pathol, 155, 787-98.

95. Breitkreutz, D., Schoop, V.M., Mirancea, N., Baur, M., Stark, H.J. and Fusenig,

N.E. (1998) Epidermal differentiation and basement membrane formation by HaCaT cells in surface transplants. Eur J Cell Biol, 75, 273-86.

96. Borchers, A.H., Sanders, L.A., Powell, M.B. and Bowden, G.T. (1997)

Melanocyte mediated paracrine induction of extracellular matrix degrading proteases in squamous cell carcinoma cells. Exp Cell Res, 231, 61-5.

97. Nomizu, M., Kim, W.H., Yamamura, K., Utani, A., Song, S.Y., Otaka, A., Roller,

P.P., Kleinman, H.K. and Yamada, Y. (1995) Identification of cell binding sites in the laminin alpha 1 chain carboxyl-terminal globular domain by systematic screening of synthetic peptides. J Biol Chem, 270, 20583-90.

98. Boudreau, N. and Bissell, M.J. (1998) Extracellular matrix signaling: integration

of form and function in normal and malignant cells. Curr Opin Cell Biol, 10, 640-6.

99. Wewer, U.M., Shaw, L.M., Albrechtsen, R. and Mercurio, A.M. (1997) The

integrin alpha 6 beta 1 promotes the survival of metastatic human breast carcinoma cells in mice. Am J Pathol, 151, 1191-8.

100. Meerschaert, J., Vrtis, R.F., Shikama, Y., Sedgwick, J.B., Busse, W.W. and

Mosher, D.F. (1999) Engagement of alpha4beta7 integrins by monoclonal antibodies or ligands enhances survival of human eosinophils in vitro. J Immunol,163, 6217-27.

101. Sethi, T., Rintoul, R.C., Moore, S.M., MacKinnon, A.C., Salter, D., Choo, C.,

Chilvers, E.R., Dransfield, I., Donnelly, S.C., Strieter, R. and Haslett, C. (1999) Extracellular matrix proteins protect small cell lung cancer cells against apoptosis: a mechanism for small cell lung cancer growth and drug resistance in vivo. Nat Med, 5, 662-8.

129 102. Streuli, C.H. and Gilmore, A.P. (1999) Adhesion-mediated signaling in the

regulation of mammary epithelial cell survival. J Mammary Gland Biol Neoplasia, 4, 183-91.

103. Brizzi, M.F., Defilippi, P., Rosso, A., Venturino, M., Garbarino, G., Miyajima,

A., Silengo, L., Tarone, G. and Pegoraro, L. (1999) Integrin-mediated adhesion of endothelial cells induces JAK2 and STAT5A activation: role in the control of c-fos gene expression. Mol Biol Cell, 10, 3463-71.

104. Bachelder, R.E., Ribick, M.J., Marchetti, A., Falcioni, R., Soddu, S., Davis, K.R.

and Mercurio, A.M. (1999) p53 inhibits alpha 6 beta 4 integrin survival signaling by promoting the caspase 3-dependent cleavage of AKT/PKB. J Cell Biol, 147,1063-72.

105. Cress, A.E. and Dalton, W.S. (1996) Multiple drug resistance and intermediate

filaments. Cancer Metastasis Rev, 15, 499-506. 106. St Croix, B., Man, S. and Kerbel, R.S. (1998) Reversal of intrinsic and acquired

forms of drug resistance by hyaluronidase treatment of solid tumors. Cancer Lett,131, 35-44.

107. Kerbel, R.S., Rak, J., Kobayashi, H., Man, M.S., St Croix, B. and Graham, C.H.

(1994) Multicellular resistance: a new paradigm to explain aspects of acquired drug resistance of solid tumors. Cold Spring Harb Symp Quant Biol, 59, 661-72.

108. Damiano, J.S., Cress, A.E., Hazlehurst, L.A., Shtil, A.A. and Dalton, W.S. (1999)

Cell adhesion mediated drug resistance (CAM-DR): role of integrins and resistance to apoptosis in human myeloma cell lines. Blood, 93, 1658-67.

109. Hay, E.D. (1991) Cell Biology of the Extracelluar Matrix. Plenum Press, New

York. 110. Lee, E.C., Lotz, M.M., Steele, G.D., Jr. and Mercurio, A.M. (1992) The integrin

alpha 6 beta 4 is a laminin receptor. J Cell Biol, 117, 671-8. 111. Mercurio, A.M. and Shaw, L.M. (1991) Laminin binding proteins. Bioessays, 13,

469-73. 112. Friedrichs, K., Ruiz, P., Franke, F., Gille, I., Terpe, H.J. and Imhof, B.A. (1995)

High expression level of alpha 6 integrin in human breast carcinoma is correlated with reduced survival. Cancer Res, 55, 901-6.

113. Rabinovitz, I. and Mercurio, A.M. (1996) The integrin alpha 6 beta 4 and the

biology of carcinoma. Biochem Cell Biol, 74, 811-21.

130 114. Rosendahl, A., Neumann, K., Chaloupka, B., Rothmund, M. and Weinel, R.J.

(1993) Expression and distribution of VLA receptors in the pancreas: an immunohistochemical study. Pancreas, 8, 711-8.

115. Carey, T.E., Laurikainen, L., Ptok, A., Reinke, T., Linder, K., Nair, T.S. and

Marcelo, C. (1992) Culture conditions affect expression of the alpha 6 beta 4 integrin associated with aggressive behavior in head and neck cancer. Adv Exp Med Biol, 320, 69-79.

116. Weinel, R.J., Rosendahl, A., Neumann, K., Chaloupka, B., Erb, D., Rothmund,

M. and Santoso, S. (1992) Expression and function of VLA-alpha 2, -alpha 3, -alpha 5 and -alpha 6-integrin receptors in pancreatic carcinoma. Int J Cancer, 52,827-33.

117. Niessen, C.M., Hogervorst, F., Jaspars, L.H., de Melker, A.A., Delwel, G.O.,

Hulsman, E.H., Kuikman, I. and Sonnenberg, A. (1994) The alpha 6 beta 4 integrin is a receptor for both laminin and kalinin. Exp Cell Res, 211, 360-7.

118. Brown, J.C. and Goodman, S.L. (1991) Different cellular receptors for human

placental laminin and murine EHS laminin. FEBS Lett, 282, 5-8. 119. Ruiz, P., Dunon, D., Sonnenberg, A. and Imhof, B.A. (1993) Suppression of

mouse melanoma metastasis by EA-1, a monoclonal antibody specific for alpha 6 integrins. Cell Adhes Commun, 1, 67-81.

120. Lochter, A., Navre, M., Werb, Z. and Bissell, M.J. (1999) alpha1 and alpha2

integrins mediate invasive activity of mouse mammary carcinoma cells through regulation of stromelysin-1 expression. Mol Biol Cell, 10, 271-82.

121. Kibbey, M.C., Grant, D.S. and Kleinman, H.K. (1992) Role of the SIKVAV site

of laminin in promotion of angiogenesis and tumor growth: an in vivo Matrigel model. J Natl Cancer Inst, 84, 1633-8.

122. Grant, D.S., Kinsella, J.L., Fridman, R., Auerbach, R., Piasecki, B.A., Yamada,

Y., Zain, M. and Kleinman, H.K. (1992) Interaction of endothelial cells with a laminin A chain peptide (SIKVAV) in vitro and induction of angiogenic behavior in vivo. J Cell Physiol, 153, 614-25.

123. Bresalier, R.S., Schwartz, B., Kim, Y.S., Duh, Q.Y., Kleinman, H.K. and Sullam,

P.M. (1995) The laminin alpha 1 chain Ile-Lys-Val-Ala-Val (IKVAV)-containing peptide promotes liver colonization by human colon cancer cells. Cancer Res, 55,2476-80.

131 124. Nomizu, M., Kuratomi, Y., Song, S.Y., Ponce, M.L., Hoffman, M.P., Powell,

S.K., Miyoshi, K., Otaka, A., Kleinman, H.K. and Yamada, Y. (1997) Identification of cell binding sequences in mouse laminin gamma1 chain by systematic peptide screening. J Biol Chem, 272, 32198-205.

125. Skubitz, A.P., Letourneau, P.C., Wayner, E. and Furcht, L.T. (1991) Synthetic

peptides from the carboxy-terminal globular domain of the A chain of laminin: their ability to promote cell adhesion and neurite outgrowth, and interact with heparin and the beta 1 integrin subunit. J Cell Biol, 115, 1137-48.

126. Graf, J., Ogle, R.C., Robey, F.A., Sasaki, M., Martin, G.R., Yamada, Y. and

Kleinman, H.K. (1987) A pentapeptide from the laminin B1 chain mediates cell adhesion and binds the 67,000 laminin receptor. Biochemistry, 26, 6896-900.

127. Iwamoto, Y., Robey, F.A., Graf, J., Sasaki, M., Kleinman, H.K., Yamada, Y. and

Martin, G.R. (1987) YIGSR, a synthetic laminin pentapeptide, inhibits experimental metastasis formation. Science, 238, 1132-4.

128. Sakamoto, N., Iwahana, M., Tanaka, N.G. and Osada, Y. (1991) Inhibition of

angiogenesis and tumor growth by a synthetic laminin peptide, CDPGYIGSR-NH2. Cancer Res, 51, 903-6.

129. Skubitz, A.P., McCarthy, J.B., Zhao, Q., Yi, X.Y. and Furcht, L.T. (1990)

Definition of a sequence, RYVVLPR, within laminin peptide F-9 that mediates metastatic fibrosarcoma cell adhesion and spreading. Cancer Res, 50, 7612-22.

130. Kleinman, H.K., Graf, J., Iwamoto, Y., Sasaki, M., Schasteen, C.S., Yamada, Y.,

Martin, G.R. and Robey, F.A. (1989) Identification of a second active site in laminin for promotion of cell adhesion and migration and inhibition of in vivo melanoma lung colonization. Arch Biochem Biophys, 272, 39-45.

131. Charonis, A.S., Skubitz, A.P., Koliakos, G.G., Reger, L.A., Dege, J., Vogel,

A.M., Wohlhueter, R. and Furcht, L.T. (1988) A novel synthetic peptide from the B1 chain of laminin with heparin-binding and cell adhesion-promoting activities. J Cell Biol, 107, 1253-60.

132. Loftus, J.C. and Liddington, R.C. (1997) Cell adhesion in vascular biology. New

insights into integrin-ligand interaction. J Clin Invest, 99, 2302-6. 133. Arthur, W.T., Noren, N.K. and Burridge, K. (2002) Regulation of Rho family

GTPases by cell-cell and cell-matrix adhesion. Biol Res, 35, 239-46. 134. Hynes, R.O. (2002) Integrins: bidirectional, allosteric signaling machines. Cell,

110, 673-87.

132 135. Panetti, T.S. (2002) Tyrosine phosphorylation of paxillin, FAK, and p130CAS:

effects on cell spreading and migration. Front Biosci, 7, d143-50. 136. Slack-Davis, J.K., Eblen, S.T., Zecevic, M., Boerner, S.A., Tarcsafalvi, A., Diaz,

H.B., Marshall, M.S., Weber, M.J., Parsons, J.T. and Catling, A.D. (2003) PAK1 phosphorylation of MEK1 regulates fibronectin-stimulated MAPK activation. JCell Biol, 162, 281-91.

137. McCandless, J.R., Cress, A.E., Rabinovitz, I., Payne, C.M., Bowden G.T., Knox,

J.D., and Nagle R.B. (1997) A human xenograft model for testing early events of epithelial neoplastic invasion. Int J Oncol., 10, 279-285.

138. Hazlehurst, L.A., Foley, N.E., Gleason-Guzman, M.C., Hacker, M.P., Cress, A.E.,

Greenberger, L.W., De Jong, M.C. and Dalton, W.S. (1999) Multiple mechanisms confer drug resistance to mitoxantrone in the human 8226 myeloma cell line. Cancer Res, 59, 1021-8.

139. Ilic, D., Almeida, E.A., Schlaepfer, D.D., Dazin, P., Aizawa, S. and Damsky,

C.H. (1998) Extracellular matrix survival signals transduced by focal adhesion kinase suppress p53-mediated apoptosis. J Cell Biol, 143, 547-60.

140. Frisch, S.M. and Ruoslahti, E. (1997) Integrins and anoikis. Curr Opin Cell Biol,

9, 701-6. 141. Schwartz, M.A. (1997) Integrins, oncogenes, and anchorage independence. J Cell

Biol, 139, 575-8. 142. Ruoslahti, E. and Reed, J.C. (1994) Anchorage dependence, integrins, and

apoptosis. Cell, 77, 477-8. 143. MacGrogan, D. and Bookstein, R. (1997) Tumour suppressor genes in prostate

cancer. Semin Cancer Biol, 8, 11-9. 144. Lauffenburger, D.A. and Horwitz, A.F. (1996) Cell migration: a physically

integrated molecular process. Cell, 84, 359-69. 145. Sahai, E. and Marshall, C.J. (2003) Differing modes of tumour cell invasion have

distinct requirements for Rho/ROCK signalling and extracellular proteolysis. Nat Cell Biol, 5, 711-9.

146. Friedl, P. and Wolf, K. (2003) Tumour-cell invasion and migration: diversity and

escape mechanisms. Nat Rev Cancer, 3, 362-74.

133 147. Zhou, H. and Kramer, R.H. (2005) Integrin engagement differentially modulates

epithelial cell motility by RhoA/ROCK and PAK1. J Biol Chem, 280, 10624-35. 148. Guo, W. and Giancotti, F.G. (2004) Integrin signalling during tumour

progression. Nat Rev Mol Cell Biol, 5, 816-26. 149. Schmelz, M., Cress, A.E., Scott, K.M., Burger, F., Cui, H., Sallam, K., McDaniel,

K.M., Dalkin, B.L. and Nagle, R.B. (2002) Different phenotypes in human prostate cancer: alpha6 or alpha3 integrin in cell-extracellular adhesion sites. Neoplasia, 4, 243-54.

150. Gu, J., Sumida, Y., Sanzen, N. and Sekiguchi, K. (2001) Laminin-10/11 and

fibronectin differentially regulate integrin-dependent Rho and Rac activation via p130(Cas)-CrkII-DOCK180 pathway. J Biol Chem, 276, 27090-7.

151. Kariya, Y. and Miyazaki, K. (2004) The basement membrane protein laminin-5

acts as a soluble cell motility factor. Exp Cell Res, 297, 508-20. 152. Shang, M., Koshikawa, N., Schenk, S. and Quaranta, V. (2001) The LG3 module

of laminin-5 harbors a binding site for integrin alpha3beta1 that promotes cell adhesion, spreading, and migration. J Biol Chem, 276, 33045-53.

153. Hibino, S., Shibuya, M., Engbring, J.A., Mochizuki, M., Nomizu, M. and

Kleinman, H.K. (2004) Identification of an active site on the laminin alpha5 chain globular domain that binds to CD44 and inhibits malignancy. Cancer Res, 64,4810-6.

154. Ponce, M.L., Hibino, S., Lebioda, A.M., Mochizuki, M., Nomizu, M. and

Kleinman, H.K. (2003) Identification of a potent peptide antagonist to an active laminin-1 sequence that blocks angiogenesis and tumor growth. Cancer Res, 63,5060-4.

155. Nakahara, H., Mueller, S.C., Nomizu, M., Yamada, Y., Yeh, Y. and Chen, W.T.

(1998) Activation of beta1 integrin signaling stimulates tyrosine phosphorylation of p190RhoGAP and membrane-protrusive activities at invadopodia. J Biol Chem, 273, 9-12.

156. Song, S.Y., Nomizu, M., Yamada, Y. and Kleinman, H.K. (1997) Liver

metastasis formation by laminin-1 peptide (LQVQLSIR)-adhesion selected B16-F10 melanoma cells. Int J Cancer, 71, 436-41.

157. Slack, J.K., Adams, R.B., Rovin, J.D., Bissonette, E.A., Stoker, C.E. and Parsons,

J.T. (2001) Alterations in the focal adhesion kinase/Src signal transduction

134

pathway correlate with increased migratory capacity of prostate carcinoma cells. Oncogene, 20, 1152-63.

158. Somlyo, A.V., Bradshaw, D., Ramos, S., Murphy, C., Myers, C.E. and Somlyo,

A.P. (2000) Rho-kinase inhibitor retards migration and in vivo dissemination of human prostate cancer cells. Biochem Biophys Res Commun, 269, 652-9.

159. Mitchison, T.J. and Cramer, L.P. (1996) Actin-based cell motility and cell

locomotion. Cell, 84, 371-9. 160. Pollard, T.D. and Borisy, G.G. (2003) Cellular motility driven by assembly and

disassembly of actin filaments. Cell, 112, 453-65. 161. Weed, S.A. and Parsons, J.T. (2001) Cortactin: coupling membrane dynamics to

cortical actin assembly. Oncogene, 20, 6418-34. 162. Plopper, G.E., Domanico, S.Z., Cirulli, V., Kiosses, W.B. and Quaranta, V.

(1998) Migration of breast epithelial cells on Laminin-5: differential role of integrins in normal and transformed cell types. Breast Cancer Res Treat, 51, 57-69.

163. Li, X., Ma, Z., Zhu, J., Li, K., Wang, S., Bie, P. and Dong, J. (2004) Laminin-5

promotes cell motility by regulating the function of the integrin alpha6beta1 in pancreatic cancer. Carcinogenesis, 25, 2283.

164. Mercurio, A.M., Rabinovitz, I. and Shaw, L.M. (2001) The alpha 6 beta 4 integrin

and epithelial cell migration. Curr Opin Cell Biol, 13, 541-5. 165. Russell, A.J., Fincher, E.F., Millman, L., Smith, R., Vela, V., Waterman, E.A.,

Dey, C.N., Guide, S., Weaver, V.M. and Marinkovich, M.P. (2003) Alpha 6 beta 4 integrin regulates keratinocyte chemotaxis through differential GTPase activation and antagonism of alpha 3 beta 1 integrin. J Cell Sci, 116, 3543-56.

166. Parsons, J.T. (2003) Focal adhesion kinase: the first ten years. J Cell Sci, 116,

1409-16. 167. Slack-Davis, J.K. and Parsons, J.T. (2004) Emerging views of integrin signaling:

implications for prostate cancer. J Cell Biochem, 91, 41-6. 168. Colognato, H. and Yurchenco, P.D. (2000) Form and function: the laminin family

of heterotrimers. Dev Dyn, 218, 213-34.

135 169. Nakahara, H., Nomizu, M., Akiyama, S.K., Yamada, Y., Yeh, Y. and Chen, W.T.

(1996) A mechanism for regulation of melanoma invasion. Ligation of alpha6beta1 integrin by laminin G peptides. J Biol Chem, 271, 27221-4.

170. Leu, S.J., Liu, Y., Chen, N., Chen, C.C., Lam, S.C. and Lau, L.F. (2003)

Identification of a novel integrin alpha 6 beta 1 binding site in the angiogenic inducer CCN1 (CYR61). J Biol Chem, 278, 33801-8.

171. Murayama, O., Nishida, H. and Sekiguchi, K. (1996) Novel peptide ligands for

integrin alpha 6 beta 1 selected from a phage display library. J Biochem (Tokyo),120, 445-51.

172. Huttenlocher, A., Ginsberg, M.H. and Horwitz, A.F. (1996) Modulation of cell

migration by integrin-mediated cytoskeletal linkages and ligand-binding affinity. J Cell Biol, 134, 1551-62.

173. Vuori, K. and Ruoslahti, E. (1995) Tyrosine phosphorylation of p130Cas and

cortactin accompanies integrin-mediated cell adhesion to extracellular matrix. JBiol Chem, 270, 22259-62.

174. Hodivala-Dilke, K.M., DiPersio, C.M., Kreidberg, J.A. and Hynes, R.O. (1998)

Novel roles for alpha3beta1 integrin as a regulator of cytoskeletal assembly and as a trans-dominant inhibitor of integrin receptor function in mouse keratinocytes. JCell Biol, 142, 1357-69.

175. Ritter, M.R., Zhou, Q. and Markland, F.S., Jr. (2001) Contortrostatin, a

homodimeric disintegrin, actively disrupts focal adhesion and cytoskeletal structure and inhibits cell motility through a novel mechanism. Cell Commun Adhes, 8, 71-86.

176. Ritter, M.R. and Markland, F.S., Jr. (2000) Contortrostatin activates ERK2 and

tyrosine phosphorylation events via distinct pathways. Biochem Biophys Res Commun, 274, 142-8.

177. Schmitmeier, S., Markland, F.S., Ritter, M.R., Sawcer, D.E. and Chen, T.C.

(2003) Functional effect of contortrostatin, a snake venom disintegrin, on human glioma cell invasion in vitro. Cell Commun Adhes, 10, 1-16.

178. Ritter, M.R., Zhou, Q. and Markland, F.S., Jr. (2000) Contortrostatin, a snake

venom disintegrin, induces alphavbeta3-mediated tyrosine phosphorylation of CAS and FAK in tumor cells. J Cell Biochem, 79, 28-37.

179. Coelho, A.L., De Freitas, M.S., Mariano-Oliveira, A., Rapozo, D.C., Pinto, L.F.,

Niewiarowski, S., Zingali, R.B., Marcinkiewicz, C. and Barja-Fidalgo, C. (2004)

136

RGD- and MLD-disintegrins, jarastatin and EC3, activate integrin-mediated signaling modulating the human neutrophils chemotaxis, apoptosis and IL-8 gene expression. Exp Cell Res, 292, 371-84.

180. Pierschbacher, M.D. and Ruoslahti, E. (1984) Cell attachment activity of

fibronectin can be duplicated by small synthetic fragments of the molecule. Nature, 309, 30-3.

181. Ruoslahti, E. and Pierschbacher, M.D. (1987) New perspectives in cell adhesion:

RGD and integrins. Science, 238, 491-7. 182. Humphries, M.J., Olden, K. and Yamada, K.M. (1986) A synthetic peptide from

fibronectin inhibits experimental metastasis of murine melanoma cells. Science,233, 467-70.

183. Gehlsen, K.R., Argraves, W.S., Pierschbacher, M.D. and Ruoslahti, E. (1988)

Inhibition of in vitro tumor cell invasion by Arg-Gly-Asp-containing synthetic peptides. J Cell Biol, 106, 925-30.

184. Engbring, J.A. and Kleinman, H.K. (2003) The basement membrane matrix in

malignancy. J Pathol, 200, 465-70. 185. Mahato, R.I., Narang, A.S., Thoma, L. and Miller, D.D. (2003) Emerging trends

in oral delivery of peptide and protein drugs. Crit Rev Ther Drug Carrier Syst, 20,153-214.

186. Hamman, J.H., Enslin, G.M. and Kotze, A.F. (2005) Oral delivery of Peptide

drugs : barriers and developments. BioDrugs, 19, 165-77. 187. Kumagai, H., Tajima, M., Ueno, Y., Giga-Hama, Y. and Ohba, M. (1991) Effect

of cyclic RGD peptide on cell adhesion and tumor metastasis. Biochem Biophys Res Commun, 177, 74-82.

188. Kaneda, Y., Yamamoto, Y., Okada, N., Tsutsuml, Y., Nakagawa, S., Kakiuch,

M., Maeda, M., Kawasaki, K. and Mayumi, T. (1997) Antimetastatic effect of synthetic Glu-Ile-Leu-Asp-Val peptide derivatives containing D-amino acids. Anticancer Drugs, 8, 702-7.

189. Yamamoto, Y., Tsutsumi, Y. and Mayumi, T. (2002) Molecular design of

bioconjugated cell adhesion peptide with a water-soluble polymeric modifier for enhancement of antimetastatic effect. Curr Drug Targets, 3, 123-30.

190. Fujii, H., Nishikawa, N., Komazawa, H., Suzuki, M., Kojima, M., Itoh, I., Obata,

A., Ayukawa, K., Azuma, I. and Saiki, I. (1998) A new pseudo-peptide of Arg-

137

Gly-Asp (RGD) with inhibitory effect on tumor metastasis and enzymatic degradation of extracellular matrix. Clin Exp Metastasis, 16, 94-104.

191. Dechantsreiter, M.A., Planker, E., Matha, B., Lohof, E., Holzemann, G., Jonczyk,

A., Goodman, S.L. and Kessler, H. (1999) N-Methylated cyclic RGD peptides as highly active and selective alpha(V)beta(3) integrin antagonists. J Med Chem, 42,3033-40.

192. Greenfield, N.J. (1996) Methods to estimate the conformation of proteins and

polypeptides from circular dichroism data. Anal Biochem, 235, 1-10. 193. Kelly, S.M. and Price, N.C. (2000) The use of circular dichroism in the

investigation of protein structure and function. Curr Protein Pept Sci, 1, 349-84. 194. Klassen, R.B. and Opella, S.J. (1997) NMR studies of peptides and proteins

associated with membranes. Methods Mol Biol, 60, 271-97. 195. Reid, D.G., MacLachlan, L.K., Edwards, A.J., Hubbard, J.A. and Sweeney, P.J.

(1997) Introduction to the NMR of proteins. Methods Mol Biol, 60, 1-28. 196. Hemler, M.E. (1998) Integrin associated proteins. Curr Opin Cell Biol, 10, 578-

85. 197. Stipp, C.S., Kolesnikova, T.V. and Hemler, M.E. (2003) Functional domains in

tetraspanin proteins. Trends Biochem Sci, 28, 106-12. 198. Stipp, C.S., Kolesnikova, T.V. and Hemler, M.E. (2003) EWI-2 regulates

alpha3beta1 integrin-dependent cell functions on laminin-5. J Cell Biol, 163,1167-77.

199. Kolesnikova, T.V., Stipp, C.S., Rao, R.M., Lane, W.S., Luscinskas, F.W. and

Hemler, M.E. (2004) EWI-2 modulates lymphocyte integrin alpha4beta1 functions. Blood, 103, 3013-9.

200. Zhang, X.A., Bontrager, A.L., Stipp, C.S., Kraeft, S.K., Bazzoni, G., Chen, L.B.

and Hemler, M.E. (2001) Phosphorylation of a conserved integrin alpha 3 QPSXXE motif regulates signaling, motility, and cytoskeletal engagement. Mol Biol Cell, 12, 351-65.

201. Zhang, X.A., Lane, W.S., Charrin, S., Rubinstein, E. and Liu, L. (2003)

EWI2/PGRL associates with the metastasis suppressor KAI1/CD82 and inhibits the migration of prostate cancer cells. Cancer Res, 63, 2665-74.

138 202. Gesierich, S., Paret, C., Hildebrand, D., Weitz, J., Zgraggen, K., Schmitz-

Winnenthal, F.H., Horejsi, V., Yoshie, O., Herlyn, D., Ashman, L.K. and Zoller, M. (2005) Colocalization of the tetraspanins, CO-029 and CD151, with integrins in human pancreatic adenocarcinoma: impact on cell motility. Clin Cancer Res,11, 2840-52.

203. Stipp, C.S. and Hemler, M.E. (2000) Transmembrane-4-superfamily proteins

CD151 and CD81 associate with alpha 3 beta 1 integrin, and selectively contribute to alpha 3 beta 1-dependent neurite outgrowth. J Cell Sci, 113 ( Pt 11),1871-82.

204. Yauch, R.L., Berditchevski, F., Harler, M.B., Reichner, J. and Hemler, M.E.

(1998) Highly stoichiometric, stable, and specific association of integrin alpha3beta1 with CD151 provides a major link to phosphatidylinositol 4-kinase, and may regulate cell migration. Mol Biol Cell, 9, 2751-65.

205. Mazzocca, A., Carloni, V., Sciammetta, S., Cordella, C., Pantaleo, P., Caldini, A.,

Gentilini, P. and Pinzani, M. (2002) Expression of transmembrane 4 superfamily (TM4SF) proteins and their role in hepatic stellate cell motility and wound healing migration. J Hepatol, 37, 322-30.

206. Hemler, M.E. (2001) Specific tetraspanin functions. J Cell Biol, 155, 1103-7. 207. Lammerding, J., Kazarov, A.R., Huang, H., Lee, R.T. and Hemler, M.E. (2003)

Tetraspanin CD151 regulates alpha6beta1 integrin adhesion strengthening. Proc Natl Acad Sci U S A, 100, 7616-21.

208. Kazarov, A.R., Yang, X., Stipp, C.S., Sehgal, B. and Hemler, M.E. (2002) An

extracellular site on tetraspanin CD151 determines alpha 3 and alpha 6 integrin-dependent cellular morphology. J Cell Biol, 158, 1299-309.

209. Zhang, X.A., Bontrager, A.L. and Hemler, M.E. (2001) Transmembrane-4

superfamily proteins associate with activated protein kinase C (PKC) and link PKC to specific beta(1) integrins. J Biol Chem, 276, 25005-13.

210. Serru, V., Le Naour, F., Billard, M., Azorsa, D.O., Lanza, F., Boucheix, C. and

Rubinstein, E. (1999) Selective tetraspan-integrin complexes (CD81/alpha4beta1, CD151/alpha3beta1, CD151/alpha6beta1) under conditions disrupting tetraspan interactions. Biochem J, 340 ( Pt 1), 103-11.

211. Berditchevski, F., Zutter, M.M. and Hemler, M.E. (1996) Characterization of

novel complexes on the cell surface between integrins and proteins with 4 transmembrane domains (TM4 proteins). Mol Biol Cell, 7, 193-207.

139 212. Berditchevski, F. and Odintsova, E. (1999) Characterization of integrin-

tetraspanin adhesion complexes: role of tetraspanins in integrin signaling. J Cell Biol, 146, 477-92.

213. Domanico, S.Z., Pelletier, A.J., Havran, W.L. and Quaranta, V. (1997) Integrin

alpha 6A beta 1 induces CD81-dependent cell motility without engaging the extracellular matrix migration substrate. Mol Biol Cell, 8, 2253-65.

214. Stipp, C.S., Kolesnikova, T.V. and Hemler, M.E. (2001) EWI-2 is a major CD9

and CD81 partner and member of a novel Ig protein subfamily. J Biol Chem, 276,40545-54.

215. Clark, K.L., Zeng, Z., Langford, A.L., Bowen, S.M. and Todd, S.C. (2001) PGRL

is a major CD81-associated protein on lymphocytes and distinguishes a new family of cell surface proteins. J Immunol, 167, 5115-21.

216. Humphries, M.J. (2000) Integrin structure. Biochem Soc Trans, 28, 311-39. 217. Mould, A.P. (1996) Getting integrins into shape: recent insights into how integrin

activity is regulated by conformational changes. J Cell Sci, 109 ( Pt 11), 2613-8. 218. Shimaoka, M., Takagi, J. and Springer, T.A. (2002) Conformational regulation of

integrin structure and function. Annu Rev Biophys Biomol Struct, 31, 485-516. 219. Mould, A.P., Akiyama, S.K. and Humphries, M.J. (1996) The inhibitory anti-

beta1 integrin monoclonal antibody 13 recognizes an epitope that is attenuated by ligand occupancy. Evidence for allosteric inhibition of integrin function. J Biol Chem, 271, 20365-74.

220. Mould, A.P., Garratt, A.N., Askari, J.A., Akiyama, S.K. and Humphries, M.J.

(1995) Identification of a novel anti-integrin monoclonal antibody that recognises a ligand-induced binding site epitope on the beta 1 subunit. FEBS Lett, 363, 118-22.

221. Bazzoni, G., Shih, D.T., Buck, C.A. and Hemler, M.E. (1995) Monoclonal

antibody 9EG7 defines a novel beta 1 integrin epitope induced by soluble ligand and manganese, but inhibited by calcium. J Biol Chem, 270, 25570-7.

222. Ridley, A.J., Schwartz, M.A., Burridge, K., Firtel, R.A., Ginsberg, M.H., Borisy,

G., Parsons, J.T. and Horwitz, A.R. (2003) Cell migration: integrating signals from front to back. Science, 302, 1704-9.

140 223. Webb, D.J., Donais, K., Whitmore, L.A., Thomas, S.M., Turner, C.E., Parsons,

J.T. and Horwitz, A.F. (2004) FAK-Src signalling through paxillin, ERK and MLCK regulates adhesion disassembly. Nat Cell Biol, 6, 154-61.

224. Zaidel-Bar, R., Ballestrem, C., Kam, Z. and Geiger, B. (2003) Early molecular

events in the assembly of matrix adhesions at the leading edge of migrating cells. J Cell Sci, 116, 4605-13.

225. Rinehart, J., Adjei, A.A., Lorusso, P.M., Waterhouse, D., Hecht, J.R., Natale,

R.B., Hamid, O., Varterasian, M., Asbury, P., Kaldjian, E.P., Gulyas, S., Mitchell, D.Y., Herrera, R., Sebolt-Leopold, J.S. and Meyer, M.B. (2004) Multicenter phase II study of the oral MEK inhibitor, CI-1040, in patients with advanced non-small-cell lung, breast, colon, and pancreatic cancer. J Clin Oncol, 22, 4456-62.

226. Allen, L.F., Sebolt-Leopold, J. and Meyer, M.B. (2003) CI-1040 (PD184352), a

targeted signal transduction inhibitor of MEK (MAPKK). Semin Oncol, 30, 105-16.

227. Druker, B.J., Talpaz, M., Resta, D.J., Peng, B., Buchdunger, E., Ford, J.M.,

Lydon, N.B., Kantarjian, H., Capdeville, R., Ohno-Jones, S. and Sawyers, C.L. (2001) Efficacy and safety of a specific inhibitor of the BCR-ABL tyrosine kinase in chronic myeloid leukemia. N Engl J Med, 344, 1031-7.

228. Mauro, M.J. and Druker, B.J. (2001) STI571: targeting BCR-ABL as therapy for

CML. Oncologist, 6, 233-8. 229. White, C.A., Larocca, A. and Grillo-Lopez, A.J. (1999) Anti-CD20 monoclonal

antibodies as novel treatments for non-Hodgkin's lymphoma. Pharm. Sci. Technol. Today, 2, 95-101.

230. Grillo-Lopez, A.J., White, C.A., Varns, C., Shen, D., Wei, A., McClure, A. and

Dallaire, B.K. (1999) Overview of the clinical development of rituximab: first monoclonal antibody approved for the treatment of lymphoma. Semin Oncol, 26,66-73.

231. Dillman, R.O. (1999) Perceptions of Herceptin: a monoclonal antibody for the

treatment of breast cancer. Cancer Biother Radiopharm, 14, 5-10. 232. Graziano, C. (1998) HER-2 breast assay, linked to Herceptin, wins FDA's okay.

CAP Today, 12, 1, 14-6. 233. Kerr, J.S., Slee, A.M. and Mousa, S.A. (2002) The alpha v integrin antagonists as

novel anticancer agents: an update. Expert Opin Investig Drugs, 11, 1765-74.

141 234. Tucker, G.C. (2003) Alpha v integrin inhibitors and cancer therapy. Curr Opin

Investig Drugs, 4, 722-31. 235. Gutheil, J.C., Campbell, T.N., Pierce, P.R., Watkins, J.D., Huse, W.D., Bodkin,

D.J. and Cheresh, D.A. (2000) Targeted antiangiogenic therapy for cancer using Vitaxin: a humanized monoclonal antibody to the integrin alphavbeta3. Clin Cancer Res, 6, 3056-61.

236. Mikecz, K. (2000) Vitaxin applied molecular evolution. Curr Opin Investig

Drugs, 1, 199-203. 237. Patel, S.R., Jenkins, J., Papadopolous, N., Burgess, M.A., Plager, C., Gutterman,

J. and Benjamin, R.S. (2001) Pilot study of vitaxin--an angiogenesis inhibitor-in patients with advanced leiomyosarcomas. Cancer, 92, 1347-8.

238. Posey, J.A., Khazaeli, M.B., DelGrosso, A., Saleh, M.N., Lin, C.Y., Huse, W. and

LoBuglio, A.F. (2001) A pilot trial of Vitaxin, a humanized anti-vitronectin receptor (anti alpha v beta 3) antibody in patients with metastatic cancer. Cancer Biother Radiopharm, 16, 125-32.

239. Smith, J.W. (2003) Cilengitide Merck. Curr Opin Investig Drugs, 4, 741-5. 240. Eskens, F.A., Dumez, H., Hoekstra, R., Perschl, A., Brindley, C., Bottcher, S.,

Wynendaele, W., Drevs, J., Verweij, J. and van Oosterom, A.T. (2003) Phase I and pharmacokinetic study of continuous twice weekly intravenous administration of Cilengitide (EMD 121974), a novel inhibitor of the integrins alphavbeta3 and alphavbeta5 in patients with advanced solid tumours. Eur J Cancer, 39, 917-26.

241. Burke, P.A., DeNardo, S.J., Miers, L.A., Lamborn, K.R., Matzku, S. and

DeNardo, G.L. (2002) Cilengitide targeting of alpha(v)beta(3) integrin receptor synergizes with radioimmunotherapy to increase efficacy and apoptosis in breast cancer xenografts. Cancer Res, 62, 4263-72.

242. Raguse, J.D., Gath, H.J., Bier, J., Riess, H. and Oettle, H. (2004) Cilengitide

(EMD 121974) arrests the growth of a heavily pretreated highly vascularised head and neck tumour. Oral Oncol, 40, 228-30.

243. Reinmuth, N., Liu, W., Ahmad, S.A., Fan, F., Stoeltzing, O., Parikh, A.A.,

Bucana, C.D., Gallick, G.E., Nickols, M.A., Westlin, W.F. and Ellis, L.M. (2003) Alphavbeta3 integrin antagonist S247 decreases colon cancer metastasis and angiogenesis and improves survival in mice. Cancer Res, 63, 2079-87.

244. Stoeltzing, O., Liu, W., Reinmuth, N., Fan, F., Parry, G.C., Parikh, A.A.,

McCarty, M.F., Bucana, C.D., Mazar, A.P. and Ellis, L.M. (2003) Inhibition of

142

integrin alpha5beta1 function with a small peptide (ATN-161) plus continuous 5-FU infusion reduces colorectal liver metastases and improves survival in mice. Int J Cancer, 104, 496-503.

245. Weinel, R.J., Rosendahl, A., Pinschmidt, E., Kisker, O., Simon, B. and Santoso,

S. (1995) The alpha 6-integrin receptor in pancreatic carcinoma. Gastroenterology, 108, 523-32.

246. Welch, D.R. (1997) Technical considerations for studying cancer metastasis in

vivo. Clin Exp Metastasis, 15, 272-306.


Recommended