83
Analysis of Munc18c and Syntaxin4 Function During Tumour Cell Invasion by David Michael Martynowicz A Thesis presented to The University of Guelph In partial fulfilment of requirements for the degree of Master of Science in Molecular and Cellular Biology Guelph, Ontario, Canada © David Michael Martynowicz, December, 2015

Analysis of Munc18c and Syntaxin4 Function During Tumour

  • Upload
    others

  • View
    1

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Analysis of Munc18c and Syntaxin4 Function During Tumour

Analysis of Munc18c and Syntaxin4 Function During Tumour Cell Invasion

by

David Michael Martynowicz

A Thesis presented to

The University of Guelph

In partial fulfilment of requirements for the degree of

Master of Science in

Molecular and Cellular Biology

Guelph, Ontario, Canada

© David Michael Martynowicz, December, 2015

Page 2: Analysis of Munc18c and Syntaxin4 Function During Tumour

ABSTRACT

ANALYSIS OF MUNC18C AND SYNTAXIN4 FUNCTION DURING TUMOUR CELL INVASION

David Martynowicz Advisor: University of Guelph, 2015 Dr. M. G. Coppolino

Tumour cell invasion through the ECM (extracellular matrix) involves the precise

localization of proteins required for ECM proteolysis and cell migration. Membrane

trafficking events, mediated by SNAREs (Soluble NSF Attachment Protein Receptors),

have been implicated in these cellular processes. Previous studies on SNAREs indicate

that Syntaxin4 is involved in the formation of invadopodia (specialized degradative

structures formed during tumour cell invasion) in MDA-MB-231 cells; however, it

remains unclear how Syntaxin4 function is regulated during tumour cell invasion.

Munc18c is a known regulator of Syntaxin4 activity, and a potential role for Munc18c in

invadopodium-based ECM degradation and cell migration has been identified. Here,

biochemical and microscopic analyses revealed an association between Munc18c and

Syntaxin4. Munc18c knockdown perturbed invadopodium formation and cell migration.

Expression of a truncated form of Syntaxin4 designed to perturb Syntaxin4-Munc18c

interactions had similar effects. These results suggest Munc18c facilitates Syntaxin4

function during tumour cell invasion.

Page 3: Analysis of Munc18c and Syntaxin4 Function During Tumour

iii

Table of Contents

Acknowledgments ..............................................................................................................v

Declaration of Work Performed ..................................................................................... vi

List of Abbreviations ...................................................................................................... vii

List of Figures ..................................................................................................................... x

1.0 – Introduction ...............................................................................................................1

1.1 – Overview ................................................................................................................1

1.2 – Tumour Cell Invasion ...........................................................................................3

1.3 – Cellular contact and adhesion with the extracellular matrix ...........................3

1.4 – Cell Migration .......................................................................................................7

1.5 – Extracellular matrix proteolysis ..........................................................................7

1.6 – Invadopodia Formation ......................................................................................10

1.7 – Intracellular membrane trafficking ..................................................................13

1.8 – SNARE structure and function .........................................................................14

1.9 – SNARE-mediated trafficking during cell migration and invasion .................18

1.10 – Regulation of SNARE Complex Formation ...................................................19

1.11 – Munc18c Function as a Sec1/Munc18 protein ................................................21

1.12 – Experimental Objectives ..................................................................................24

2.0 – Materials and Methods ...........................................................................................26

2.1 – Materials ..............................................................................................................26

2.1.1: Reagents ...........................................................................................................262.1.2: cDNA constructs ..............................................................................................262.1.3: Cell Culture ......................................................................................................272.1.4: Transfections ....................................................................................................28

2.2 – Methods ................................................................................................................28

2.2.1: Creation of Stable Cell Lines ...........................................................................282.2.2: Cell Migration Assay .......................................................................................28

Page 4: Analysis of Munc18c and Syntaxin4 Function During Tumour

iv

2.2.3: Invadopodium Formation Assay ......................................................................292.2.4: Immunoprecipitation ........................................................................................292.2.5: Immunoblotting ...............................................................................................302.2.6: Immunofluoresence Microscopy .....................................................................302.2.7: Statistical Analysis ...........................................................................................31

3.0 Results .........................................................................................................................32

3.1 Syntaxin4 associates with Munc18c in MDA-MB-231 cells ...............................32

3.2 Syntaxin4 and Munc18c association is enhanced during invadopodium formation ......................................................................................................................34

3.3 Analysis of Munc18c localization relative to invadopodium components ........34

3.4 Munc18c knockdown impairs invadopodium formation ...................................36

3.5 Expression of N-terminal Syntaxin4-GFP impairs invadopodium formation .39

3.6 Munc18c knockdown and expression of N-terminal Syntaxin4-GFP impair cell migration .......................................................................................................................43

3.7 Creation of stable cell lines expressing either Munc18c, Syntaxin4-FL-GFP or N-terminal Syntaxin4-GFP .........................................................................................43

3.8 Evaluating invadopodium formation and cell migration by stable cell lines ...49

4.0 Discussion ...................................................................................................................52

5.0 Summary .....................................................................................................................61

6.0 References ...................................................................................................................64

Page 5: Analysis of Munc18c and Syntaxin4 Function During Tumour

v

Acknowledgments I would like to thank my supervisor, Dr. Marc Coppolino for his support during

my studies and providing me with the tools and experience that facilitated my growth as a

molecular and cellular biologist. I would also like to thank my advisory committee, Dr.

Nina Jones and Dr. Dick Mosser for their additional support during the entire process.

In no particular order, special thanks goes to the following people for their

influence throughout my entire experience in MCB: Maria Anillo, Vadika Mishra, Erin

Specker, Sean White, Matt Clarke, Dr. Steffen Graether, Evan Mallette, Kevin Bosse,

Agata Zienowicz, Charles Wroblewski, Reema Deol, Kestral Danzmann, Rachael

MacNeilly, Jennifer Butler, Allie Davidson, Tijana Matovic, Michael Brodzikowski, Wei

Wu, Rebecca Rumney, Jeff Madge, John Atkinson, Andrew Jenkins, Dr. Vladimir

Bamm, Dr. George Harauz, Sara Timpano, Richard Preiss, Angus Ross, Kristina

O’Hanley, Jessica Wong, Ranko Savic, Jordan Meyers, Marina Atalla, Greer Wallen,

Shruti Patel, Anna Quach, Katie MacKenzie, Megan Brasher, Joban Dhanoa, Pam

Loughran, Dr. Rod Merrill, Dr. John Dawson, Julia Ebeling, Dan Krska, Adam Schmidt,

Tom Keeling, Jordan Willis, Ava Keyvani Chahi, Claire Martin, Olivia Anderson,

Brianna Guild, Roman Kondra, Megan Massey, Adam Rocker, Kayla Heney, Dr. Karla

Williams, Stephanie Brunelle, Madison Turner, Rob Taylor-Reid, Amanda Poole, Kelly

Boddington, Dr. Kenrick Vassall, Dr. Miguel de Avila, Paula Russell, Hussam Alsarraf,

Jaspreet Kaur, Kim Kirby, Dr. Peihua Lu, Haidun Lu, Matiyo Ojehomon, Mackenzie

Charter, Candace Mehaffey, Brass Taps management, and the Second cup ladies.

Last but not least, to my parents and family, I could not have done this without

you – the most important tools to succeed were provided by you.

Page 6: Analysis of Munc18c and Syntaxin4 Function During Tumour

vi

Declaration of Work Performed Together, Ranko Savic and Megan Brasher performed ligations and subsequently

verified the creation of the following expression constructs: N-terminal Syntaxin4-GFP

and untagged human Munc18c. Dr. Marc Coppolino created the G418 kill-curve

necessary for the selection and maintenance of the stable cell lines described in this

thesis. The author performed all other experiments.

Page 7: Analysis of Munc18c and Syntaxin4 Function During Tumour

vii

List of Abbreviations α-SNAP N-ethylmaleimide-sensitive factor attachment protein alpha

ARP Actin-related protein BCS Bovine Calf Serum BSA Bovine Serum Albumin CD44 Cluster of differentiation 44 cDNA Complementary deoxyribonucleic acid CHO Chinese Hamster Ovary DMEM Dulbecco’s modified Eagle’s medium DNA Deoxyribonucleic acid DOC Deoxycholate ECM Extracellular Matrix EDTA Ethylenediaminetetraacetic acid EGFP Enhanced Green Fluorescent Protein F-actin Filamentous actin FBS Fetal Bovine Serum FL Full Length G418 Geneticin GFP Green Fluorescent Protein GLUT4 Glucose transporter type 4 GTPase Guanosine triphosphate hydrolase Habc Domain with anti-parallel α-helices labelled a, b, and c HEK-293 Human embryonic kidney cells 293

Page 8: Analysis of Munc18c and Syntaxin4 Function During Tumour

viii

HRP Horseradish peroxidase IgG Immunoglobulin type G IP Immunoprecipitation kDa kiloDaltons M Mouse miRNA Micro ribonucleic acid MMP Matrix Metalloprotease MT1-MMP Membrane Type 1 – Matrix Metalloprotease Munc18 Mammalian uncoordinated-18 NA Numerical Aperture NSF N-ethylmaleimide sensitive fusion protein NP40 Nonylphenoxypolyethoxylethanol N-terminal Amino terminal N-WASP Neuronal Wiskott-Aldrich syndrome protein ORF Open reading frame PAGE Polyacrylamide gel electrophoresis PBS Phosphate buffered saline PCR Polymerase chain reaction PFA Paraformaldehyde PLL Poly-L-Lysine PVDF Polyvinylidene fluoride Rb Rabbit Rab Ras-related in brain

Page 9: Analysis of Munc18c and Syntaxin4 Function During Tumour

ix

Ras Rat sarcoma RNA Ribonucleic acid RNAi Ribonucleic acid interference SDS Sodium dodecyl sulfate Sec1 Protein transport protein SEC1 S.E.M. Standard error of the mean STX4 Syntaxin 4 siRNA Short interfering ribonucleic acid SNAP Synaptosomal-associated protein

SNARE Soluble N-ethylmaleimide-sensitive factor attachment protein receptor

TBST Tris buffered saline with Tween-20 added Tris 2-Amino-2(hydroxymethyl)-1,3-propanediol TX-100 Triton X-100 VAMP Vesicle-associated membrane protein

Page 10: Analysis of Munc18c and Syntaxin4 Function During Tumour

x

List of Figures

Figure 1: Schematic representation of metastasis ...........................................................4

Figure 2: Schematic diagram of cell adhesion .................................................................6

Figure 3: Schematic diagram of two-dimensional cell migration ..................................8

Figure 4: Simplified model of invadopodium formation ..............................................12

Figure 5: Overview of membrane trafficking pathways ..............................................15

Figure 6: SNARE-mediated membrane fusion .............................................................17

Figure 7: Different Syntaxin binding modes of SM proteins .......................................22

Figure 8: Analysis of Syntaxin4 and Munc18c association in MDA-MB-231 cells. ...33

Figure 9: Syntaxin4 and Munc18c association increases during invadopodium formation. .................................................................................................................35

Figure 10: Subcellular distribution of Munc18c during the formation of invadopodia ..............................................................................................................37

Figure 11: RNAi-mediated knockdown of Munc18c ....................................................38

Figure 12: RNAi-mediated knockdown of Munc18c impairs invadopodium formation. .................................................................................................................40

Figure 13: Segmental architecture of N-terminal Syntaxin4-GFP. .............................41

Figure 14: N-terminal Syntaxin4-GFP impairs invadopodium formation. ................42

Figure 15: N-terminal Syntaxin4-GFP does not alter co-immunoprecipitation of Syntaxin4 with Munc18c. ........................................................................................44

Figure 16: Munc18c knockdown or overexpression of N-terminal Syntaxin4 impairs cell migration. ...........................................................................................................45

Figure 17: Expression of GFP-Munc18c in HEK-293 cells. .........................................46

Figure 18: Expression of Munc18c, GFP-Syntaxin4-full length or GFP-Syntaxin4-N-terminus in stable cell lines. ....................................................................................48

Figure 19: Analysis of invadopodium formation by stable cell lines. .........................50

Figure 20: Quantification of invadopodium formation and cell migration by stable cell lines. ....................................................................................................................51

Page 11: Analysis of Munc18c and Syntaxin4 Function During Tumour

xi

Figure 21: Proposed model of Munc18c function during tumour cell invasion .........63

Page 12: Analysis of Munc18c and Syntaxin4 Function During Tumour

1

1.0 – Introduction 1.1 – Overview

Cellular invasion and migration are fundamental to the homeostasis of

multicellular organisms. They are integral parts of many physiological processes

including embryogenesis and the maintenance of tissue architecture. During embryonic

development, cells must remodel the ECM and migrate to their intended destination in

order to form specialized tissues and organs (Moissoglu & Schwartz, 2012). During

vertebrate organogenesis, movement through and breakdown of the ECM by epithelial

cells of the lung and kidney are important for the expansion of these tissues (Andrew &

Ewald, 2010; Lu, Sternlicht, & Werb, 2006). Cellular invasion also plays an important

role in wound healing and immune cell surveillance. Mesenchymal stem cells must

migrate to the damaged area in order to fill in a wound, and subsequently leukocytes, in

response to external stimuli, extravasate through the vascular basement membrane to

mitigate foreign material within the wounded area (Huttenlocher & Horwitz, 2011).

Similar cellular processes that mediate cellular invasion in normal physiological

processes are also attributed to the progression of pathological disorders such as cancer

(Friedl & Alexander, 2011). Metastatic spread of cancer is mediated, in part, by tumour

cell invasion through the ECM. Tumour cell invasion through the ECM barrier can be

defined as a multistep process comprising cell adhesion, ECM proteolysis and cell

migration (Bravo-Cordero, Hodgson, & Condeelis, 2012). To move through the ECM,

cells first utilize surface receptors to engage with ECM components. Cells also secrete

proteases to degrade the ECM and ultimately migrate through it. The molecular

mechanisms that regulate the function of these processes are currently an important area

Page 13: Analysis of Munc18c and Syntaxin4 Function During Tumour

2

of research due to their significance to cancer management and treatment. An important

area of study that has advanced our understanding of the molecular machinery used by

cells during tumour cell invasion is invadopodia. These subcellular structures are actin-

driven cell membrane protrusions utilizing both cell surface ECM receptors and

proteolytic enzymes for their function and formation. Invadopodia have been studied in

cancer cells in a variety of microenvironments in vitro (Artym et al., 2015; Tolde, Rösel,

Veselý, Folk, & Brábek, 2010), and evidence from in vivo studies supports their

relevance and role in the dissemination of tumour cell populations (Clark et al., 2008;

Leong et al., 2014; Lohmer, Kelley, Hagedorn, & Sherwood, 2014).

Membrane trafficking of proteins to invadopodia is required for their formation

and function during tumour cell invasion. The molecular mechanisms controlling the

trafficking of these proteins are an active area of study. Key players of intracellular

membrane trafficking events are SNAREs. These proteins function to localize and fuse

vesicles with target membranes. Work has started to highlight an important role for

SNARE-mediated trafficking in invadopodium formation and tumour cell invasion;

however, the mechanism that controls SNARE function in this context is not well

understood. Research into this area may lead to a better understanding of the formation of

invadopodia and the mechanisms by which cells can become invasive. Furthermore, by

defining the mechanism of SNARE regulation in this process, the development of

therapies that could reduce ECM degradation and invasion by tumour cells may be

possible.

Page 14: Analysis of Munc18c and Syntaxin4 Function During Tumour

3

1.2 – Tumour Cell Invasion

A simple model of metastasis describes the escape of a tumour cell from a

primary tumour and movement through the ECM, ultimately leading to its dissemination

to distant sites within the body (Beaty & Condeelis, 2014). The degradation of the ECM

by invading carcinoma cells is a good example of this (Hanahan & Weinberg, 2011).

Carcinomas restricted to the epithelium may be considered benign. However, the

acquisition of an invasive phenotype by cells of the tumour would lead to the breaching

of the basement membrane, and individual or groups of cancer cells may then begin to

invade underlying ECM and nearby lymphatic or vascular vessels (Yamaguchi, 2012)

(Figure 1). Once these cells arrive within the lumen of these vessels, they can travel

within the blood or lymph to other areas of the body and potentially establish metastases.

Invasion into the ECM by carcinoma cells also releases growth and survival factors that

have been sequestered by the ECM (Talmadge & Fidler, 2010). Taken together, tumour

cell invasion involves alterations in the molecular mechanisms that control cell adhesion,

proteolytic degradation of the ECM, and cell migration.

1.3 – Cellular contact and adhesion with the extracellular matrix

The ECM is an important interlocking mesh composed of fibrous proteins and

glycoproteins acting as a supportive scaffold and physical barrier to maintain tissue

organization (Ridley et al., 2003). Examples of ECM components include: collagen,

fibronectin and vitronectin (Oskarsson, 2013). In multicellular organisms, contact

between the cell and ECM provides a crucial biochemical bridge between the

extracellular and intracellular environments allowing modulation of cellular behaviour.

Cells make contact with the ECM through a variety of specialized structures that mediate

Page 15: Analysis of Munc18c and Syntaxin4 Function During Tumour

4

Figure 1: Schematic representation of metastasis In the epithelium, cells of a primary tumour develop an invasive phenotype and invade the basement membrane with the aid of invadopodia. Invasive cells can then migrate through the stromal tissue to intravasate a nearby blood vessel, travel to secondary sites, and potentially form metastases.

Invadopodia) Invasion)

Primary)Tumour)

Basement))membrane)

Blood)vessel)

Distant)Metastasis)

Stroma)

Page 16: Analysis of Munc18c and Syntaxin4 Function During Tumour

5

homeostasis by conferring anchorage, motility, and signal transduction (Adams, 2002).

Anchorage of the cell to the ECM can be provided by mechanically supportive contacts

such as hemidesmosomes, which link the cytoskeleton of epithelial cells to the basement

membrane through the engagement of specialized adhesion receptors (Borradori &

Sonnenberg, 1999). Contractile contacts, such as focal adhesions, mediate cellular

movement by utilizing the cells actomyosin motor system to provide force against the

rigid ECM via cytoskeletal remodelling (Zamir et al., 2000). Protrusive contacts, such as

filopodia, are actin-containing extensions of the plasma membrane at the leading edge of

migrating cells, which facilitate extension into the ECM (Mattila & Lappalainen, 2008).

Collectively, cell-matrix contacts are dynamic and specialized structures that allow

sampling of the environment around the cell body.

Cell adhesion and interaction with the ECM is primarily mediated through the

integrin family of receptors (Berrier & Yamada, 2007). Integrins are transmembrane

glycoproteins, which perform their function as heterodimers of α and β subunits. Each

subunit comes together to bind ECM substrates located in the pericellular environment

and this transmits intracellular signals through the cytoplasmic domains. Briefly, cell

adhesion begins with the binding of integrins to ECM substrates resulting in the

recruitment of other integrins and signalling and scaffolding proteins that mediate

cytoskeletal rearrangements. This focal accumulation of proteins is termed a focal

adhesion, and as the cell spreads, more focal adhesions take shape, leading to a fully

spread and adherent cell (Figure 2).

Page 17: Analysis of Munc18c and Syntaxin4 Function During Tumour

6

Figure 2: Schematic diagram of cell adhesion Adhesion to the ECM is mediated by integrins. Upon binding to an ECM substrate, integrins recruit cytoskeletal, scaffolding and signalling proteins. Focal sites of attachment form, which can mature into focal adhesions. Focal adhesions are stabilized by F-actin fibres, leading to a fully spread and adherent cell.

Cell$in$suspension$

Adhesion$ Spreading$

F3ac5n$ Integrin$ Extracellular$Matrix$

Page 18: Analysis of Munc18c and Syntaxin4 Function During Tumour

7

1.4 – Cell Migration Cell migration is a multistep process that occurs in response to external stimuli

and involves the repetition of the following steps: 1) membrane extension at the leading

edge, 2) cell adhesion to the ECM, 3) contraction of the cell body, and 4) detachment at

the cell rear (Ridley et al., 2003). Initially, membrane protrusions such as lamellipodia at

the leading edge of the cell take shape and sample the environment beyond the cell body

(Figure 3). Regulators of actin polymerization promote the formation of these

protrusions. Once an actin containing protrusion has been produced, adhesive contacts

must form to prevent retraction of the newly formed membrane protrusion. This is

facilitated by focal adhesions, which ultimately link the actin cytoskeleton to the ECM.

The next step in cell migration involves the contraction of the cell body. This is primarily

mediated through actomyosin-based forces directed against the ECM. Concomitantly,

adhesions that were initially produced at the leading edge reach the rear of the cell as the

cell body translocates. These older adhesions disassemble in response to actomyosin

contraction or protease-directed degradation. Taken together, actomyosin contraction

and adhesion disassembly at the rear promotes cell body translocation.

1.5 – Extracellular matrix proteolysis

Dissolution of the ECM is an important prerequisite for tumour cell invasion

(Hanahan & Weinberg, 2011). This process has been shown to be largely dependent on

the engagement of MMPs with ECM substrates (Egeblad & Werb, 2002; Hojilla, Wood,

& Khokha, 2008). Furthermore, invadopodia rely on MMPs to facilitate their maturation

(Clark & Weaver, 2008; Linder, 2007). The human MMP family of proteins comprises

23 members that are zinc dependent-endopeptidases with broad substrate specificity

Page 19: Analysis of Munc18c and Syntaxin4 Function During Tumour

8

Figure 3: Schematic diagram of two-dimensional cell migration Cells adherent to ECM establish focal adhesions. Migration begins with membrane protrusions such as lamellipodia that extend in the direction of movement. New focal adhesions form at the leading edge and actomyosin contraction causes the cell body to translocate while older focal adhesions at the cell rear disassemble.

Focal&Adhesion&

New&Adhesion&

Lamellipodium&

Extension&

Adhesion&

De8adhesion&

Direc:on&of&Movement&

Cell&body&movement&

Page 20: Analysis of Munc18c and Syntaxin4 Function During Tumour

9

(Jackson, Nebert, & Vasiliou, 2010). Two further divisions categorize these proteins as

soluble or membrane-anchored. The membrane-anchored MMPs contain either a

transmembrane domain or a glycosylphosphatidylinositol anchor and the soluble MMPs

are secreted by cells into the ECM environment (Llano et al., 1999; Sohail et al., 2008;

Takino, Sato, Shinagawa, & Seiki, 1995). Both types are translated as inactive zymogens,

therefore requiring cleavage of their intrinsic pro-domains to be proteolytically active.

The activity of MMPs has been shown to be closely linked to the invasiveness of several

aggressive cancers such as melanoma, breast, ovarian, and colorectal (Decock et al.,

2008; Seftor et al., 2001; Sodek, Ringuette, & Brown, 2007; Zucker et al., 1999). Tumour

progression in these cancers has also been shown to correlate specifically with the

enzymatic activity of MT1-MMP (Sabeh, Shimizu-Hirota, & Weiss, 2009). Moreover,

invadopodium formation has been shown to be mediated by MT1-MMP (Artym, Zhang,

Seillier-Moiseiwitsch, Yamada, & Mueller, 2006). Over-expression of MT1-MMP in

moderately invasive melanoma cells was shown to increase matrix degradation and

invadopodium activity (Revach & Geiger, 2014). Other studies have demonstrated that

over-expression of MT1-MMP in different cancer cell types promoted invasion using in

vitro and in vivo models (Hotary et al., 2003; Sabeh, 2004). In addition, the knockdown

of MT1-MMP in invasive breast cancer cell lines reduced the ability of cells to invade

ECM substrates in vitro (Jiang et al., 2006). MT1-MMP is important to the degradation

of ECM components utilized by invasive carcinomas, and much evidence points to a role

for MT1-MMP in invadopodium-based tumour cell invasion.

Page 21: Analysis of Munc18c and Syntaxin4 Function During Tumour

10

1.6 – Invadopodia Formation

Cellular degradation and migration through the ECM is important for cellular

processes involving tissue remodelling, including wound healing, embryogenesis, and

immune cell surveillance (Daley, Peters, & Larsen, 2008; Lu, Takai, Weaver, & Werb,

2011; Vicente-Manzanares, 2005). Actin-driven invadosomes are protrusive contacts that

facilitate focal ECM degradation and cell motility through the use of proteolytic enzymes

(Linder, Wiesner, & Himmel, 2011; Murphy & Courtneidge, 2011). Podosomes refer to

these structures in normal cells, such as macrophages and dendritic cells (Burns, 2001;

Linder, Hufner, Wintergerst, & Aepfelbacher, 2000), whereas in cancerous cells they are

referred to as invadopodia (Gimona, Buccione, Courtneidge, & Linder, 2008). The partial

degradation of ECM components is crucial for tumour cell invasion and it is the

employment of invadopodia with their localized proteolytic machinery that helps to

initiate metastasis (Blouw, Seals, Pass, Díaz, & Courtneidge, 2008; Clark et al., 2008).

In 1989, the term ‘invadopodia’ was first used to describe the phenomenon of

overlapping microfilament-containing membrane protrusions and localized sites of

substratum degradation in Rous sarcoma virus transformed fibroblasts (Chen, 1989).

More accurately, these sub-cellular structures are now described to be located

predominantly on the ventral surface and situated below the nucleus of an invasive

carcinoma cell (Artym et al., 2015). Furthermore, these protrusions can be observed in

vitro, using confocal microscopy techniques, and have been observed to overlap with

sites of proteolytic degradation and F-actin punctae when aggressive cancer cells are

cultured on various ECM substrates, such as fluorescently conjugated gelatin or

fibronectin (Hoshino et al., 2013; Oser et al., 2009).

Page 22: Analysis of Munc18c and Syntaxin4 Function During Tumour

11

The multitude of proteins that converge at invadopodia is indicative of a complex

and highly regulated means of cellular invasion. Research into these sub-cellular

structures has discerned 4 main subgroups of proteins involved in their formation and

activity. The first group consists of cell motility and protrusion-inducing machinery,

which includes regulators of F-actin polymerization and branching. Examples of these are

ARP2/3, N-WASP, cofilin, cortactin, and dynamin (Baldassarre, 2002; Oser et al., 2009;

Yamaguchi et al., 2005). Adhesion proteins constitute the second group, categorized

based on the maintenance of cell-ECM contact. These adhesion molecules are

exemplified by integrins and CD44 (Kajita et al., 2001; Wolf et al., 2007).The third group

consists of signalling proteins from the tyrosine kinase family and Ras-related GTPases,

which ultimately regulate membrane trafficking and dynamics (Neel et al., 2012; Tehrani,

Tomasevic, Weed, Sakowicz, & Cooper, 2007). The final group is made up of the

proteolytic machinery that facilitates ECM degradation, including members of the MMP

family, separase, and urokinase-type plasminogen activators system (Artym, 2002;

Monsky et al., 1994; Revach & Geiger, 2014).

During the formation of invadopodia (Figure 4), cells adhere to the ECM through

interactions with ventral surface integrins. Concomitantly, growth factors such as

epidermal growth factor and platelet derived growth factor trigger a signalling cascade

that ultimately leads to the recruitment of actin modifying proteins to sites of forming

invadopodia (Mader et al., 2011; Yamaguchi et al., 2005). Next, cortactin associates with

N-WASP and the ARP2/3 complex to mediate F-actin assembly and branching, resulting

in a protrusion of the plasma membrane, driven by the force of actin filament formation.

Subsequently, membrane trafficking of MT1-MMP to invadopodia facilitates membrane

Page 23: Analysis of Munc18c and Syntaxin4 Function During Tumour

12

Figure 4: Simplified model of invadopodium formation Cells initially establish adhesions through integrins and stimulate signalling pathways through growth factor receptor tyrosine kinases (RTKs). F-actin formation and branching by N-WASP and cortactin establishes protrusions of the cell membrane into the extracellular matrix. Trafficking of MT1-MMP facilitates invadopodium maturation through degradation of extracellular matrix components. F-actin core disassembly begins through changes to actin modifying proteins, initiating invadopodia disassembly and subsequent cellular migration through the degraded extracellular matrix.

Ini$a$on' Assembly' Matura$on' Disassembly'

Cortac$n'

N7WASP'

F7ac$n'

MT17MMP'

RTK'

Integrin'

Page 24: Analysis of Munc18c and Syntaxin4 Function During Tumour

13

protrusion into the basement membrane as the invadopodium matures (Clark, Whigham,

Yarbrough, & Weaver, 2007; Steffen et al., 2008). MT1-MMP is theorized to invoke a

positive, feed forward mechanism through the release of extracellular growth factors

from the ECM (Díaz, Yuen, Iizuka, Higashiyama, & Courtneidge, 2013) and the

activation of MMP-2, which can also activate growth factor receptors (Dean et al., 2007;

Sato et al., 1994). Mature invadopodia are established once ECM degradation has

occurred and are relatively long-lived structures persisting for hours. Their disassembly

has been suggested to begin with the clearance of F-actin and cortactin leaving behind an

MT1-MMP containing invadopodium that is still capable of localized ECM degradation

(Artym et al., 2006).

1.7 – Intracellular membrane trafficking

Eukaryotic plasma membranes provide a physical barrier between a cell and its

extracellular environment, and regulate the transport of molecules to and from this

environment in order to maintain homeostasis inside the cell. Membranes are also

important inside the cell: each organelle within the cell (for example, the Golgi complex)

is surrounded by a lipid bilayer, which, similar to the plasma membrane, is involved in

regulation of transport into and out of each organelle.

Cell migration and invasion require the precise localization of adhesion and

signalling proteins. Membrane trafficking coordinates the transport and localization of

these proteins from different membrane compartments. The transport of vesicles between

membranes can be broadly defined (Figure 5). In summary, vesicle formation involves

the budding off of vesicles from the donor membrane. Vesicle movement is the

transportation of vesicles along microtubules or actin filaments through the action of

Page 25: Analysis of Munc18c and Syntaxin4 Function During Tumour

14

motor proteins. Vesicle tethering and docking mediates the correct targeting of vesicles

and brings donor and target membranes in close proximity. Ultimately, this leads to the

process by which lipid bilayers merge, therefore delivering the contents of the vesicle to

the target compartment or extracellular environment. SNAREs are proteins that mediate

membrane fusion events and the purpose of the studies described in this thesis is to

examine the regulation of SNAREs during cellular invasion.

1.8 – SNARE structure and function

Thirty-six SNAREs have been identified in humans (Jahn & Scheller, 2006).

SNAREs assemble into complexes containing a 4-helix bundle formed by their SNARE

motifs. Initially, SNARE proteins were classified according to the membrane they reside

in, the target or vesicle. Elucidation of the 3D structures of SNARE complexes

subsequently revealed the presence of a central ionic layer in the α-helix bundle

containing three glutamine (Q) residues and one arginine (R). This led to the

reclassification of SNARE proteins as either R- or Q-SNAREs depending on which

residue they contribute to the ionic layer (Kloepper, Kienle, & Fasshauer, 2007). The 4-

helix bundle of the SNARE complex contains one member of each of the four SNARE

subtypes, referred to as R, Qa, Qb, and Qc.

Within cells, SNAREs can be functionally described under three general protein

types. Syntaxins, a family of Q-SNARE proteins, are found to be localized to different

membrane compartments and function in several trafficking pathways (Teng, Wang, &

Tang, 2001). membrane compartments and function in several trafficking pathways For

example, Syntaxin4 is predominantly found at the plasma membrane in many cell types

(Band et al., 2002), and in adipocytes it is involved in the fusion of GLUT4 storage

Page 26: Analysis of Munc18c and Syntaxin4 Function During Tumour

15

Figure 5: Overview of membrane trafficking pathways Intracellular trafficking allows transport of cargo contained in vesicles to other membranous compartments within the cell. The biosynthetic pathway shuttles material synthesized in the ER to the Golgi complex for processing and is subsequently secreted to the plasma membrane. Recycling begins with endocytosis at the plasma membrane and trafficking to an early endosomal compartment. Some endocytosed material is then delivered to late endosomes and then lysosomes for degradation. Another endocytic route brings material back to the plasma membrane from the early endosome directly or through a recycling endosome.

Recycling))Endosome)

Recycling)

Lysosome)

Late))Endosome)

Endocytosis)

Early)Endosome)

Trans4Golgi)network)

Golgi)

Endoplasmic)Re9culum)

Nucleus)

Page 27: Analysis of Munc18c and Syntaxin4 Function During Tumour

16

vesicles with the plasma membrane in response to insulin signalling (Tellam, Macaulay,

McIntosh, & Hewish, 1997). The cytoplasmic portion of Syntaxin4 contains a conserved

SNARE motif, and a regulatory Habc domain consisting of three anti-parallel helices at its

N-terminus (Ungar & Hughson, 2003). The Habc domain is able to regulate SNARE

complex formation by conferring an open or closed conformation. The open

conformation allows Syntaxin4 to interact with cognate partners. The closed

conformation prevents SNARE complex formation (Dulubova et al., 2007). The second

type of SNAREs, synaptosome-associated proteins (SNAP), has two Q-SNARE domains.

For example, the ubiquitously expressed, SNAP23 interacts with Syntaxin4 during

GLUT4 vesicle fusion with the plasma membrane in adipocytes (Widberg, 2003).

Separated by a flexible linker region, these proteins are membrane anchored by post-

translational palmitoylation of cysteine residues in this linker region (Puri & Roche,

2006). Finally, vesicle of associated membrane proteins (VAMP) are members of the R-

SNARE family, and are comprised of a C-terminal transmembrane domain, a SNARE

motif, and a variable N-terminal domain. (Jahn & Scheller, 2006). Multiple VAMP

isoforms have been identified, with roles in a wide range of cellular trafficking pathways.

SNARE complex formation can occur when a vesicle membrane is apposed to a

target membrane (Figure 6). Once the SNAREs are in contact with each other, they form

a trans-SNARE complex when SNARE motifs form a coiled-coil. The energy released

from this complex formation helps overcome the energy barrier to merge the two

membranes, promoting membrane fusion and leaving a cis-SNARE complex (Hong,

2005). The disassembly of cis-SNARE complexes does not occur spontaneously and is

regulated by the cytosolic proteins NSF (N-ethylmaleimide sensitive fusion protein) and

Page 28: Analysis of Munc18c and Syntaxin4 Function During Tumour

17

Figure 6: SNARE-mediated membrane fusion SNARE complex assembly and vesicle docking occurs as the SNARE motif of an R-SNARE (blue) forms a complex with two other SNARE motifs from a SNAP family member (yellow) and one from Syntaxin (red). Trans-complex formation results from the zippering of the four SNARE motifs. Membrane fusion results in a cis-complex.

Target'Membrane'

Target'+'Vesicle'Membrane'

trans2complex'

cis2complex'

Vesicle'

Page 29: Analysis of Munc18c and Syntaxin4 Function During Tumour

18

α-SNAP (soluble NSF attachment protein alpha). The adaptor protein, α-SNAP, binds to

the cis-SNARE complex and recruits NSF, which through its ATPase activity separates

the complex. The SNARE proteins are recycled through the retrograde transport of R-

SNAREs and separation of Q-SNAREs.

1.9 – SNARE-mediated trafficking during cell migration and invasion

Studies of SNARE-mediated trafficking and integrin localization have shown the

relevance of SNARE function during cell migration and cell spreading. Expression of a

dominant negative form of NSF (E329Q-NSF) has been observed to impair disassembly

of SNARE complexes (Coppolino et al., 2001), thereby reducing the amount of free

SNAREs available to mediate membrane fusion. Expression of this construct impaired

cell migration in CHO cells (Tayeb et al., 2005). It was then determined that inhibition of

specific SNARE-mediated trafficking pathways perturbed trafficking of α5β1 and

impeded cell spreading, but not cell adhesion, when CHO cells were seeded on

fibronectin (Skalski & Coppolino, 2005).

SNARE-mediated membrane trafficking is also important for the delivery of

MMPs to the plasma membrane and during invadopodium formation. Previous work has

demonstrated that in the invasive cell line, HT-1080, inhibition of Syntaxin13, SNAP23

and VAMP3 perturbed the trafficking of MT1-MMP and the secretion of MMP2 and

MMP9, but did not alter cell migration (Kean et al., 2009). Other studies have shown the

importance of SNARE-mediate trafficking of MT1-MMP to the plasma membrane in

invasive carcinoma cells (Miyata et al., 2004; Steffen et al., 2008). Using the MDA-MB-

231 cell line as a model system for the study of invadopodia, SNAREs have been

identified to have a role in invadopodium formation (Williams & Coppolino, 2014). The

Page 30: Analysis of Munc18c and Syntaxin4 Function During Tumour

19

SNARE complex responsible for the delivery of MT1-MMP to invadopodia was

elucidated to be dependent on the Q-SNAREs SNAP23 and Syntaxin4, and the R-

SNARE VAMP7 (Williams, McNeilly, & Coppolino, 2014). Moreover, inhibition of

these SNAREs perturbed invadopodium formation. Taken together, specific SNARE-

mediated trafficking pathways have been identified which are important for the

localization of key proteins involved in cellular invasion.

1.10 – Regulation of SNARE Complex Formation

Although it is known that SNAREs are central mediators of membrane fusion,

how the formation of SNARE complexes is regulated during cell invasion in not

completely understood. The control of SNARE complex formation has been shown to

involve regulatory non-SNARE proteins and post-translational modifications (Hong,

2005; Tomes, 2015). Both of these factors have been implemented in the restriction of

SNARE assembly, catalysis of SNARE complex assembly, or the maintenance of

SNAREs in an active or inactive conformation. Some of the ways that SNARE complex

formation can be regulated by these factors are briefly described below.

The regulation of SNARE complex formation can be controlled by the

phosphorylation of SNAREs. For example, SNAP23 is phosphorylated by SNAK kinase,

resulting in a lack of association with Syntaxin4 at the plasma membrane (Cabaniols,

Ravichandran, & Roche, 1999). SNAP23 has also been shown to be phosphorylated by

protein kinase C, resulting in reduced binding to Syntaxin4 (Polgár, Lane, Chung, Houng,

& Reed, 2003). Syntaxin4 can be serine/threonine phosphorylated in vitro by casein

kinase 2 (Risinger & Bennett, 2002), protein kinase C (Chung, Polgár, & Reed, 2000),

and protein kinase A (Foster et al., 1998) and these phosphorylation events reduce

Page 31: Analysis of Munc18c and Syntaxin4 Function During Tumour

20

binding to cognate SNAREs. Phosphorylation of Syntaxin4 by Rab3d-kinase was also

found to decrease binding to SNAP23 (Pombo et al., 2001). Previous work has made the

observation that serine/threonine phosphorylation of Syntaxin4 was decreased during

invadopodium formation, suggesting that Syntaxin4 dephosphorylation was regulated

during trafficking of MT1-MMP to the invadopodial plasma membrane (Williams et al.,

2014). Taken together, phosphorylation of SNARE proteins is a means of regulating

cognate SNARE binding and therefore subsequent vesicle-target membrane fusion.

SNARE complex formation can also be regulated through interactions with non-

SNARE proteins. For example, the neuronal protein Snapin complexes with SNAP25 in

order to enhance binding to cognate SNAREs and therefore facilitate exocytosis (Pan,

Tian, & Sheng, 2009). Other neuronal proteins, synaptophysin and tomosyn, also regulate

vesicle fusion by controlling SNARE interactions. Synaptophysin binds to the R-SNARE

synaptobrevin preventing any binding with SNAP25, and tomosyn binds with SNAP25 to

prevent synaptobrevin binding (Yelamanchili et al., 2005). Another neuronal protein,

Munc18a, has been shown to bind to isolated Syntaxin1 in the closed conformation as

well as its cognate SNARE trans-complex (Dulubova et al., 2007; Schollmeier, Krause,

Kreye, Malsam, & Söllner, 2011). This protein is hypothesized to regulate membrane

fusion in neuronal cells either by inhibiting Syntaxin1 from forming complexes or by

enhancing membrane fusion – its exact function remains unclear. It has also been shown

that in pancreatic beta cells gelsolin binds Syntaxin4, and this interaction prevents

SNARE complex formation. Perturbation of this interaction by glucose uptake facilitates

insulin exocytosis (Kalwat, Wiseman, Luo, Wang, & Thurmond, 2012).

Page 32: Analysis of Munc18c and Syntaxin4 Function During Tumour

21

1.11 – Munc18c Function as a Sec1/Munc18 protein

Sec1/Munc18 (SM) proteins were first identified through a genetic screen of

uncoordinated mutants of the nematode Caenorhabditis elegans (Brenner, 1974) and

orthologs in mammals were subsequently identified, defining three isoforms: Munc18-a, -

b and –c (Hata, Slaughter, & Südhof, 1993). SM proteins are 60-70 kDa proteins, found

both in the cytosol and associated with membranes via interaction with their cognate

SNAREs. Studies in neuronal tissue identified Syntaxin as a major binding partner of SM

proteins (Pevsner, Hsu, & Scheller, 1994). Structural data shows that the overall fold of

SM proteins is highly conserved between different species, supporting the notion of a

common function (Bracher & Weissenhorn, 2002; Hu, Latham, Gee, James, & Martin,

2007; Misura, Scheller, & Weis, 2000). The structure includes three domains that fold

together to form a large cavity on one side, and a groove on the other. SM proteins have

been shown to interact with syntaxin by at least one of three ways (Figure 7): through

binding to the closed conformation, the N-terminal peptide domain, and the 4-helical

SNARE complex (Südhof & Rothman, 2009; Yu et al., 2013).

SM proteins show a similar loss-of-function phenotype as that of SNAREs and

are essential for every pathway of intracellular vesicle fusion (Burgoyne et al., 2009; Carr

& Rizo, 2010; Toonen & Verhage, 2007). SM proteins have previously been viewed as

regulators of SNARE function, but current findings suggest that SM proteins also

cooperate with SNARE complexes to mediate membrane fusion. Munc18a is able to bind

to closed Syntaxin1, as well as the trans-SNARE complex (Dulubova et al., 2007;

Schollmeier et al., 2011). The same observation has been made with Munc18c and its

high affinity binding partner, Syntaxin4 (Yu et al., 2013). Munc18a and Munc18c are not

Page 33: Analysis of Munc18c and Syntaxin4 Function During Tumour

22

Figure 7: Different Syntaxin binding modes of SM proteins SM proteins (purple) have been shown to interact with their cognate syntaxin (red) by three ways: full-length closed conformation of the syntaxin, the trans-SNARE complex, and the N-terminal domain of Syntaxin.

Full$length* SNARE$complex* N$terminal*

NH3+*

NH3+*

NH3+*

Habc*

SNARE$mo<f*

Page 34: Analysis of Munc18c and Syntaxin4 Function During Tumour

23

functionally interchangeable, suggesting there is a conserved mechanism of action

between isoforms though differences lie in cognate SNARE recognition. As a result, the

current model for SM protein function is that the Q-SNARE Syntaxin adopts a closed

conformation and is sequestered from cognate SNAREs by an SM protein (Baker et al.,

2015). When the vesicle comes into the proximity of the target membrane, the SM

protein-bound Q-SNARE assembles with the R-SNARE and then the other Q-SNAREs.

The SM protein then repositions to enhance trans-SNARE complex formation, therefore

promoting membrane fusion.

Munc18c has broad tissue distribution (McIntosh, 1995) and has been implicated

in several exocytic pathways. It has been found to influence secretion in neutrophils

(Brochetta et al., 2008), exocytosis in platelets (Schraw et al., 2004), and the sustained

phase of insulin secretion in adipocytes (Oh & Thurmond, 2009). Physiologically,

Munc18c has been shown to be crucial for the exocytosis of the glucose transporter,

GLUT4, during glucose homeostasis (Jewell et al., 2011). In response to insulin

signalling, GLUT4 storage vesicle fusion requires Syntaxin4 and SNAP23 as the Q-

SNAREs, VAMP2 as the R-SNARE, and Munc18c as the cognate SM protein (Brandie

et al., 2008). Homozygous knockout of Munc18c is embryonic lethal in mice; however,

heterozygous mice are viable, exhibiting a large decrease in insulin-stimulated GLUT4

plasma membrane integration compared to wild-type mice (Oh, Spurlin, Pessin, &

Thurmond, 2005). Furthermore, disruption of the interaction between endogenous

Munc18c and Syntaxin4 was also found to cause a decrease in the fusion of GLUT4-

containing vesicles with the plasma membrane and thus glucose uptake (Thurmond,

Kanzaki, Khan, & Pessin, 2000).

Page 35: Analysis of Munc18c and Syntaxin4 Function During Tumour

24

1.12 – Experimental Objectives

The observed functional importance of SNARE complex regulation on membrane

trafficking events in other contexts suggests that it may be contributing to membrane

trafficking events that support tumour cell invasion. Recent research has contributed to a

model wherein cellular invasion and invadopodium formation are dependent on the

SNARE-mediated trafficking of key proteins that facilitate invasion through the ECM

(Kean et al., 2009; Williams et al., 2014; Williams & Coppolino, 2014). Syntaxin4 has

been identified as one of the SNARE proteins responsible for the trafficking of MT1-

MMP to the plasma membrane (Miyata et al., 2004), including sites of invadopodium

formation in MDA-MB-231 cells. The ubiquitous expression of Munc18c in tissues

suggests that it may be involved in regulating Syntaxin4 in cells forming invadopodia.

The purpose of this study is to characterize the relationship between Syntaxin4 and

Munc18c during tumour cell invasion in MDA-MB-231 cells. The hypothesis of this

study is thus: Munc18c function facilitates invasion of the ECM by MDA-MB-231 cells.

To elucidate the function of Munc18c, the aims of this thesis are as follows:

Aim 1: Characterize the interaction between Syntaxin4 and Munc18c during cellular

invasion.

This will be achieved by using co-immunoprecipitation experiments to monitor

the association between endogenous Munc18c and Syntaxin4. Confocal microscopy will

be used to assess the localization of these proteins during invadopodium formation.

Aim 2: Examine the role of Munc18c during cellular invasion.

Inhibition of Munc18c function will be carried out using RNAi-mediated

knockdown and through the expression of a truncated version of Syntaxin4. Truncated

Page 36: Analysis of Munc18c and Syntaxin4 Function During Tumour

25

Syntaxin4 is predicted to act as a competitive inhibitor for Munc18c and Syntaxin4

interactions. Munc18c overexpression will be used to assess the effect of excess

Munc18c within cells. Using these approaches, the role of Syntaxin4-Munc18c

interactions in cell migration, invasion and invadopodium formation will be examined.

Page 37: Analysis of Munc18c and Syntaxin4 Function During Tumour

26

2.0 – Materials and Methods 2.1 – Materials 2.1.1: Reagents Reagents and chemicals were purchased from either Fisher-Scientific Ltd.

(Nepean, ON) or Sigma-Aldrich Co. (St. Louis, MO, USA), unless otherwise indicated.

Primary antibodies were purchased from the following suppliers: Rabbit anti-MMP14,

mouse anti-MMP14, rabbit anti-GFP, rabbit anti-Munc18c (Abcam: ab3644, ab78738,

ab290, ab175238); mouse anti-Munc18c (Santa Cruz Biotechnology: sc-373813); mouse

anti-Syntaxin4, (BD Biosciences: 610439); mouse anti-actin (Pierce: MA5-15739);

mouse anti-β1 integrin (Developmental Hybridoma Studies Bank: P4C10). All

fluorescently labelled secondary antibodies, Hoechst 33342, and AlexaFluor647-

conjugated phalloidin were purchased from Life Technologies (Mississauga, ON). HRP-

conjugated secondary antibodies were purchased from Bio-Rad (Mississauga, ON). Anti-

fade fluorescent mounting medium was obtained from DAKO, Inc. (Burlington, ON).

2.1.2: cDNA constructs

The pEGFP-N1-Syntaxin4-FL construct was described previously (Williams et

al., 2014). The N-terminal sequence of Syntaxin4 was PCR amplified from pEGFP-N1-

Syntaxin4-FL, and the amplicon was cloned into pEGFP-N1 using XhoI and KpnI to

create the plasmid encoding N-terminal Syntaxin4-GFP. Mouse Munc18c with an N-

terminal V5-epitope tag in pDEST40 was a generous gift from William S. Trimble

(Hospital for Sick Children, Toronto). The untagged ORF was PCR amplified and the

amplicon was cloned into pEGFP-C1 using EcoRI and BamHI to create the plasmid

encoding GFP-Munc18c. Human Munc18c cDNA in pCMV-Sp6 was purchased from

Page 38: Analysis of Munc18c and Syntaxin4 Function During Tumour

27

Dharmacon (MHS6278-202759106). The ORF sequence was PCR amplified, and the

amplicon was cloned into pcDNA3.1(-) using XhoI and BamHI to create the plasmid

encoding untagged human Munc18c. The following oligonucleotides were used as

primers: Forward N-terminal Syntaxin4 (5’ –

TGACGGTAAATGGCCCGCCTGGCATTATG – 3’), Reverse N-terminal Syntaxin4

(5’ - TTTATCATTGGTACCGGGTGCACCACCAGCGCG – 3’), Forward human

Munc18c (5’ – TATTTATACTCGAGCGGGAAGATGGCGCCGC – 3’), Reverse

human Munc18c (5’ –

AATGTATTAGGATCCCATATTAGTAAGAATCTCTAAACCCTC – 3’), Forward

Mouse Munc18c (5’ – GTCTCGAAGCTTCGATGGCGCCGCCGGTATC – 3’), and

Reverse Mouse Munc18c (5’ – CGAACCGCGGATCCTCTAGATCAACCACTTTG –

3’).

Knockdown of Munc18c was performed using a commercially available pool of 3

siRNA duplexes targeting human Munc18c. Munc18c and control siRNA were purchased

from Santa Cruz Biotechnologies (SC-42312 and SC-37007).

2.1.3: Cell Culture

MDA-MB-231 and HEK-293 cells were cultured in DMEM supplemented with

10% BCS. Stable cell lines derived from MDA-MB-231 cells were cultured in selection

media comprising DMEM supplemented with 5% BCS, 1 mg/mL G418 (BioShop) and

Penicillin-Streptomycin (Life Technologies). Growth conditions were kept at 37°C with

humidity and a 5% CO2 atmosphere. Cells were lifted by using 5 mM EDTA/PBS, pH

7.4. For all experiments, cells were used between passage number 5 and 20. All cells

were passaged at most, 24 hours before each experiment.

Page 39: Analysis of Munc18c and Syntaxin4 Function During Tumour

28

2.1.4: Transfections

Cells were transfected using jetPRIME Polyplus (VWR International) as per the

manufacturer’s protocol. All transiently transfected constructs were expressed for a total

of 24 hours. Cells were transfected with 50 nM siRNA and underwent knockdown for 48

hours. Co-transfections were performed using the manufacturer’s recommended amount

of marker pEGFP-C1 plasmid in addition to 50 nM siRNA for a total of 48 hours.

2.2 – Methods 2.2.1: Creation of Stable Cell Lines

In a 10 cm tissue culture plate, cells were transfected with either pcDNA3.1(-)-

Munc18c, pEGFP-N1-full length Syntaxin4, or pEGFP-N1-Nterminal Syntaxin4. After

24 hours, transfected cells and non-transfected cells were lifted in selection media and

split at a ratio of 1:4 into one 15 cm plate. Once distinct colonies had formed and all cells

in the control non-transfected plate died, 18 separate colonies were lifted using a P200

pipet tip and seeded onto 24-well plates. Once confluent, each colony-derived population

of cells was split into a 6-well dish and western blot analysis was used to confirm

expression. The cell lines that indicated the highest level of expression were propagated.

2.2.2: Cell Migration Assay

Boyden transwell migration assays were performed as previously described

(Williams et al., 2014). Tissue culture inserts with an 8-µm pore diameter (Corning) in

24-well plates were coated with 20 µg/mL fibronectin/PBS on the bottom of the

membrane. Both transfected cells and stable cell lines were serum starved for 2 hours and

subsequently counted using a haemocytometer. In serum-free media, containing 0.1%

BSA and Penicillin-Streptomycin, 20,000 cells were added to the top chamber. Cells

Page 40: Analysis of Munc18c and Syntaxin4 Function During Tumour

29

were allowed to migrate for 6 hours towards the lower chamber containing the above

medium, supplemented with 10% FBS. The top and bottom of the membrane was fixed in

4% PFA/PBS for 20 minutes, washed with 150 mM glycine/PBS for 10 minutes, stained

with Hoechst, and mounted on coverslips. Ten fields of cells per membrane were

counted, using fluorescence microscopy. For transient transfections, the data is

represented as the number of transfected cells that migrated to the bottom of the

membrane divided by the number of transfected cells that remained on top. For stable

cell lines, the data is presented as the number of cells that migrated to the bottom of the

chamber divided by the number of parental MDA-MB-231 cells that migrated to the

bottom of the chamber.

2.2.3: Invadopodium Formation Assay Invadopodium formation was performed as previously described (Artym et al.,

2009). Glass coverslips were coated with 50 µg/mL PLL/PBS, followed by crosslinking

with 0.5% gluteraldehyde/PBS. Coverslips were then inverted onto 70 µL of AlexaFluor-

594 labelled gelatin. The coated coverslips were then incubated with 5 mg/mL

NaBH3/PBS and subsequently washed 10 times with PBS. Tissue culture plates were

coated similarly; the exception being plates were coated with 0.2% unlabelled

gelatin/PBS.

2.2.4: Immunoprecipitation

Antibody was coupled to 450 µg of Protein-G Dynabeads (Invitrogen) overnight

at 4°C in PBS/0.02%Tween on an end-over-end rotator. Cells were grown to 80%

confluency or seeded onto coated tissue culture plates at 60% confluency. Cells were

lysed in situ with cold lysis buffer comprising 1% NP40, 10% glycerol, 0.5% NaDOC,

Page 41: Analysis of Munc18c and Syntaxin4 Function During Tumour

30

137 mM NaCl, 20 mM Tris-HCl pH 8.0, 10 mM NaF, 10 mM Na2P4O7, 0.2 mM Na3VO4

and protease inhibitor cocktail. Lysate was incubated with antibody bound beads for 1

hour at 4°C on an end-over-end rotator, washed 3 times with cold PBS, and eluted with

2.5X SDS-PAGE loading buffer containing 700 mM β-mercaptoethanol. The resultant

beads-antibody-antigen complex was subsequently heated to 70°C for 15 minutes.

Proteins were separated using SDS-PAGE, and analyzed using Western immunoblotting.

2.2.5: Immunoblotting

Whole cell protein and immunoprecipitation samples were electrophoresed

through a polyacrylamide gel and transferred onto a PVDF membrane with a 0.45 µm

pore diameter (EMD Millipore). Membranes were blocked in either 5% skim milk

powder or 5% BSA in TBST and probed with primary antibody (diluted 1:1000 in

TBST). HRP-conjugated secondary antibodies (diluted 1:7500 in blocking solution) were

used to detect bound primary antibodies using an enhanced chemiluminesence kit (Bio-

Rad).

2.2.6: Immunofluoresence Microscopy Cells were either grown on glass coverslips overnight or seeded onto 0.2% gelatin

coated coverslips (as described under invadopodium formation). Cells were fixed in 4%

PFA/PBS for 20 minutes then washed in 150 mM glycine/PBS for 10 minutes at room

temperature or overnight at 4°C with gentle agitation. Cells were permeabilized in 0.1%

TX-100/PBS for 10 minutes and then blocked in 5% BSA prior to staining with primary

and secondary antibody. Coverslips were mounted onto glass microscope slides using

DAKO fluorescent mounting medium. Samples analyzed by confocal microscopy were

imaged through a 63X (NA 1.4) oil immersion lens using a Leica DM-IRE2 inverted

Page 42: Analysis of Munc18c and Syntaxin4 Function During Tumour

31

microscope with a Leica TCS SP2 scanning head (Leica, Heidelberg, Germany). Images

were captured using Leica confocal software. Phase-contrast and epifluorescence images

of HEK-293 cells were acquired using a Nikon Eclipse Ti-S inverted microscope through

a 40X (NA 0.6) lens (Melville, NY, USA). Images were captured using Nikon imaging

software. All images were processed and analyzed using ImageJ software (NIH,

Bethseda, MD, USA).

2.2.7: Statistical Analysis The mean of three independent experiments is shown (unless indicated

otherwise), where error bars represent the standard error of the mean. For all treatments,

the experimental group was compared to the respective control group by Student’s t-test,

where the statistical significance threshold was p=0.05. An asterisk in figures represented

a treatment that was significantly different from the control treatment (p<0.05). Microsoft

Excel was used to perform statistical analyses.

Page 43: Analysis of Munc18c and Syntaxin4 Function During Tumour

32

3.0 Results 3.1 Syntaxin4 associates with Munc18c in MDA-MB-231 cells Previous studies have shown that Syntaxin4 and Munc18c are strong interacting

partners and the latter plays an important role during Syntaxin4-mediated exocytosis

through this interaction (Latham et al., 2006; Yu et al., 2013). To test if Munc18c and

Syntaxin4 are associating in MDA-MB-231 cells, co-immunoprecipitation experiments

were utilized. Cells were lysed in situ and Munc18c was immunoprecipitated. Eluents

were subjected to SDS-PAGE and Western blot analyses using the same Munc18c

antibody for immunoprecipitation in addition to a Syntaxin4 antibody. The results

revealed successful immunoprecipitation of Munc18c, as indicated by the enrichment of

protein in the immunoprecipitation at the predicted molecular weight of ~67 kDa (Figure

8A). Moreover, there was a significant reduction of Munc18c in the output lane relative

to the input control. Successful co-immunoprecipitation of Syntaxin4 was also observed

in Munc18c immunoprecipitates compared to immunoprecipitation control samples.

To further evaluate the association of Munc18c and Syntaxin4, confocal

immunofluorescence microscopy was utilized to assess intracellular localization. Since

Syntaxin4 is a plasma membrane SNARE (Torres, Funk, Zegers, & Beest, 2011), images

of the ventral plasma membrane were analyzed in comparison to the midcell region of

cells grown on glass coverslips. Image analyses revealed that Munc18c and Syntaxin4

were predominantly localized at the ventral plasma membrane relative to the midcell

region (Figure 8B). At the ventral surface, strong co-localization was seen at the edges of

lamellipodia. Moreover, co-localization across the ventral focal plane was incomplete.

Page 44: Analysis of Munc18c and Syntaxin4 Function During Tumour

33

Figure 8: Analysis of Syntaxin4 and Munc18c association in MDA-MB-231 cells. Munc18c and Syntaxin4 association was analyzed via immunoprecipitation/SDS-PAGE/Western blot and confocal microscopy. (A) Munc18c immunoprecipitates were probed for Munc18c and Syntaxin4. (B) Cells were grown overnight on glass coverslips fixed, permeabilized, and stained for Rb α Munc18c and M α Syntaxin4. To visualize the ventral and midcell regions of cells, the confocal z-plane was changed and the observed signal was centered in the image. White arrows indicate areas of strong co-localization. Scale bar, 10 µm.

10% in

put

10% output

Beads +

Ab

Beads +

Lysa

te

Beads +

Lysa

te

+ IgG

IP

IP: M α Munc18c

IB: Syntaxin4

IB: M α Munc18c

A

B

Vent

ral

Mid

cell

Munc18c Syntaxin4 Overlay

75 –

35 –

Page 45: Analysis of Munc18c and Syntaxin4 Function During Tumour

34

3.2 Syntaxin4 and Munc18c association is enhanced during invadopodium formation

Syntaxin4 has been shown to play an important role in the trafficking of MT1-

MMP to sites of invadopodia formation in MDA-MB-231 cells (Williams et al., 2014).

Since Syntaxin4 and Munc18c appeared to be associating in MDA-MB-231 cells,

changes to this association were examined in the context of invadopodium formation.

Cells were seeded onto a non-ECM substrate (PLL) or an ECM substrate (gelatin) for 4

hours to induce invadopodium formation. Cells were lysed in situ and Munc18c was

immunoprecipitated. Eluents were subjected to SDS-PAGE and Western blot analyses. In

all treatments, to normalize the amount of Syntaxin4 associating with

immunoprecipitated Munc18c, a second antibody specific to Munc18c was used for

immunoblotting. An increase of about 20%, relative to unlifted cells, in the amount of

Syntaxin4 co-immunoprecipitated with Munc18c was observed when cells were seeded

onto PLL (Figure 9). An increase of about 50%, relative to unlifted cells, in the amount

of Syntaxin4 co-immunoprecipitated with Munc18c was observed during invadopodium

formation on gelatin.

3.3 Analysis of Munc18c localization relative to invadopodium components The increase in Syntaxin4 co-immunoprecipitated with Munc18c during

invadopodium formation suggested that there was an important role for the association of

Munc18c and Syntaxin4 during this process. To evaluate this role, confocal

immunofluorescence microscopy was utilized to assess the localization of Munc18c

relative to proteins typically associated with invadopodium formation and function. Cells

were seeded onto coverslips coated with a thin AlexaFluor-594-labelled gelatin matrix

and incubated for 4 hours prior to fixation and staining. Use of a thin fluorescently

Page 46: Analysis of Munc18c and Syntaxin4 Function During Tumour

35

Figure 9: Syntaxin4 and Munc18c association increases during invadopodium formation. Cells were seeded onto PLL and gelatin (IVF; invadopodium formation) coated plates for 4 hours, lysed and analyzed by immunoprecipitation/SDS-PAGE/Western blot. (A) Munc18c immunoprecitates were probed for Munc18c and Syntaxin4. (B) Quantification of the amount Syntaxin4 co-immunopreciptated with Munc18c normalized to unlifted cells. Means are from three independent experiments, +/- S.E.M. Asterisks denote values significantly different from control unlifted cells (p<0.05).

A

B

Lysa

te

Unlifted

PLL IV

F Bea

ds

+ Lys

ate

Beads

+ Ab

IP: M α Munc18c

IB: Rb α Munc18c

IB: Syntaxin4

0

20

40

60

80

100

120

140

160

180

Unli%ed PLL IVF

Boun

dSyntaxin4(%

ofC

ontrol)

**

Fig. 2 A-B

75 –

35 –

Page 47: Analysis of Munc18c and Syntaxin4 Function During Tumour

36

labelled gelatin matrix allowed detection of invadopodia-forming cells within a

heterogeneous population of cells, in addition to analysis of proteins that localize to focal

points of gelatin degradation. In parallel with Munc18c staining, cells were stained for F-

actin, β1 integrin, MT1-MMP, and Syntaxin4 (Figure 10). Confocal microscopy of the

ventral surface revealed that Munc18c was distributed across the focal plane. Black spots

of gelatin degradation that overlaid with F-actin, MT1-MMP and Syntaxin4 at the centre

had Munc18c localized to the edges of degradation. β1 integrin and Munc18c partly co-

localized to the edges of some black spots of gelatin degradation. Munc18c was also

observed to co-localize with F-actin, MT1-MMP, Syntaxin4, and β1 integrin in areas

other than focal degradation sites. However, co-localization of Munc18c with the above

proteins was not complete across the focal plane.

3.4 Munc18c knockdown impairs invadopodium formation Previous work has shown that knockdown of Syntaxin4 inhibits invadopodium

formation in MDA-MB-231 cells (Williams et al., 2014). Since Munc18c and Syntaxin4

are associating in these cells, we sought to test whether Munc18c knockdown produced a

similar phenotype. Cells were transfected with a pool of siRNA targeting human

Munc18c or a non-specific control siRNA. Optimal knockdown of Munc18c was seen

after 48 hours, as determined by SDS-PAGE and Western blot analyses (data not shown).

Relative to cells transfected with control siRNA, Munc18c levels were found to be

reduced by approximately 50% (Figure 11). Munc18c knockdown had no observed

effects on β1 integrin, Syntaxin4, and MT1-MMP protein levels. Invadopodium

formation was subsequently analyzed to evaluate the effect of reduced Munc18c

expression after 48 hours. Cells co-transfected with pEGFP-C1 and siRNA were

Page 48: Analysis of Munc18c and Syntaxin4 Function During Tumour

37

Figure 10: Subcellular distribution of Munc18c during the formation of invadopodia Cells were analyzed by confocal microscopy after being plated on AlexaFluor-594-labeled gelatin for 4 hours, fixed, permeabilized, and stained for markers of invadopodia as indicated: F-actin, β1 integrin, MT1-MMP, and Syntaxin4 (cyan; white arrow). Cells were also stained for Munc18c (green; orange arrow). Dark spots in the red field indicate sites of gelatin degradation corresponding to invadopodia. Scale bar, 10 µm.

Marker Munc18c Gelatin Overlay

F-ac

tinβ 1

inte

grin

MT1

-MM

PSy

ntax

in4

Fig. 3

Page 49: Analysis of Munc18c and Syntaxin4 Function During Tumour

38

Figure 11: RNAi-mediated knockdown of Munc18c Cells were transfected with siRNA targeting Munc18c or non-specific control siRNA. (A-B) Cells were lysed, and lysate was analyzed to assess knockdown of Munc18c via SDS-PAGE/Western blot. (C) Quantification of the amount of Munc18c protein present after knockdown normalized to actin. Means are representative of three independent experiments; +/- S.E.M. Asterisks denote values significantly different from control. (p<0.05).

Control siRNA

Munc18c siRNA

Munc18c siRNA

Control siRNA

IB: β1 integrin

IB: Munc18c

IB: Actin

IB: Syntaxin4

IB: MT1-MMP

IB: Actin

0

20

40

60

80

100

120

ControlsiRNA

Munc18csiRNA

Amou

ntofM

unc18c

(%ofcon

trol)

*

B

A

C

Fig. 3 A-C

75 –

135 –

48 –

35 –

50 –

50 –

37 –

Page 50: Analysis of Munc18c and Syntaxin4 Function During Tumour

39

incubated for 44 hours, and subsequently seeded onto coverslips coated with AlexaFluor-

594 labelled gelatin. After 4 hours of invadopodium formation, cells were fixed and

stained for F- actin or MT1-MMP (Figure 12). A GFP-positive cell that had F-actin

punctae overlying black spots of degradation was counted as a cell that was forming

invadopodia. Results indicated that knockdown of Munc18c reduced the number of GFP-

positive cells forming invadopodia by approximately 50% relative to control

transfections.

3.5 Expression of N-terminal Syntaxin4-GFP impairs invadopodium formation Previous work has shown that the N-terminal 29 amino acids of Syntaxin4

facilitates binding to Munc18c (Latham et al., 2006). In vitro pull-down experiments

showed that the presence of this small polypeptide reduced the association of Munc18c

and Syntaxin4, suggesting this N-terminal domain can act as a competitive inhibitor of

Munc18c and Syntaxin4 interactions. We therefore examined the effect of a GFP-tagged

N-terminal domain of Syntaxin4 (Figure ) on invadopodium formation. Cells transfected

with either N-terminal Syntaxin4-GFP or GFP alone were subjected to invadopodium

formation assays (Figure 13). Overexpression of N-terminal Syntaxin4-GFP reduced the

amount of cells forming invadopodia by approximately 80% relative to cells transfected

with control GFP.

To assess the biochemical effects of N-terminal Syntaxin4-GFP, co-

immunoprecipitation experiments were utilized to probe for changes in the amount of

Syntaxin4 associated with Munc18c. Cells were lysed in situ and eluents of Munc18c

immunoprecipitates from cells that were either non-transfected, GFP transfected, or N-

terminal Syntaxin4-GFP transfected were analyzed via SDS-PAGE and Western blot

Page 51: Analysis of Munc18c and Syntaxin4 Function During Tumour

40

Figure 12: RNAi-mediated knockdown of Munc18c impairs invadopodium formation. (A) Invadopodium-based degradation of AlexaFluor-594-gelatin by cells co-transfected with GFP and control siRNA or Munc18c siRNA Cells transfected for 44 hours, seeded onto gelatin for 4 hours and then fixed, permeabilized, stained for MT1-MMP and analyzed by confocal microscopy. (B) Cells with F-actin punctae overlying dark spots of gelatin degradation were counted as cells forming invadopodia. Percentages of cells forming invadopodia, normalized to control (GFP-transfected) cells, are presented as the mean +/- S.E.M. from four independent experiments in which 50 cells per sample were counted. Asterisks denote values significantly different from control (p<0.05). Bar, 10 µm.

Con

trol

si

RN

AM

unc1

8c

siR

NA

GFP MT1-MMP Gelatin Overlay

0

25

50

75

100

125

Control siRNA Munc18c siRNA

Inva

dopo

dium

For

mat

ion

(% o

f con

trol

)

B

A

*

Fig. 3 D-E

Page 52: Analysis of Munc18c and Syntaxin4 Function During Tumour

41

Figure 13: Segmental architecture of N-terminal Syntaxin4-GFP. The segmental architecture of Syntaxin4 (red) comprises: an N-terminal domain, Habc domain, SNARE motif, and membrane anchor. To create N-terminal Syntaxin4-GFP, the N-terminal domain (red stripes) of Syntaxin4 was fused to the N-terminus of GFP (green).

Habc SNAREmo-f MembraneAnchor

+H3N

COO-

COO-

+H3N

N-terminal

Syntaxin4

N-terminalSyntaxin4-GFP

34kDa

27kDaCOO-+H3N

GFP

GFP

31kDa

Page 53: Analysis of Munc18c and Syntaxin4 Function During Tumour

42

Figure 13: N-terminal Syntaxin4-GFP impairs invadopodium formation. (A) Invadopodia-based degradation of gelatin by cells transfected with GFP (control) and N-terminal Syntaxin4–GFP. Cells transfected for 20 hours, seeded onto gelatin for 4 hours, and then fixed, permeabilized, stained for F-actin and analyzed by confocal microscopy. (B) Cells with F-actin punctae overlying dark areas of gelatin degradation were counted as cells forming invadopodia. Percentages of cells forming invadopodia are presented as the mean +/- S.E.M. from four independent experiments, in which 50 cells per sample were counted. Asterisks denote values significantly different from control (p<0.05). Bar, 10 µm.

GFP F-actin Gelatin Overlay

Con

trol

STX4

N-te

rm

0

20

40

60

80

100

120

Control STX4 N-term

Inva

dopo

dium

For

mat

ion

(% o

f con

trol

)

*

A

B

Fig. 4

Page 54: Analysis of Munc18c and Syntaxin4 Function During Tumour

43

(Figure ). The amount of Syntaxin4 co-immunoprecipitated with Munc18c did not change

relative to control samples. No co-immunoprecipitation of GFP alone or N-terminal

Syntaxin4-GFP was detected.

3.6 Munc18c knockdown and expression of N-terminal Syntaxin4-GFP impair cell migration Since invadopodium formation was reduced by Munc18c knockdown or the expression

of N-terminal Syntaxin4-GFP, we examined the effects of these two treatments on cell

migration using transwell migration assays. Compared to control treatments, Munc18c

knockdown or expression of N-terminal Syntaxin4-GFP inhibited cell migration by

approximately 60% and 50%, respectively (Figure 14).

3.7 Creation of stable cell lines expressing either Munc18c, Syntaxin4-FL-GFP or N-terminal Syntaxin4-GFP Previous work has shown that the overexpression of Munc18c can inhibit vesicle

fusion events that require Syntaxin4 (Tamori et al., 1998). However, other work has also

shown that overexpression of Munc18c had no significant effect on Syntaxin4-mediated

exocytosis (Yu et al., 2013). To examine the effects of Munc18c overexpression in

MDA-MB-231 cells, transient transfections were first employed using GFP-tagged and

untagged mouse Munc18c constructs. Fluorescence microscopy and western blot

analyses showed that transient transfections yielded no expression of the aforementioned

Munc18c constructs in MDA-MB-231 cells (data not shown). To confirm that the lack of

expression was not due to a problem with the expression vector, the GFP-Munc18c

construct was transfected into HEK-293 cells. Fluorescence microscopy showed that

HEK-293 cells were expressing GFP-Munc18c (Figure 15).

Next, the creation of a cell line stably expressing untagged human Munc18c was

Page 55: Analysis of Munc18c and Syntaxin4 Function During Tumour

44

Figure 15: N-terminal Syntaxin4-GFP does not alter co-immunoprecipitation of Syntaxin4 with Munc18c. Cells were either untransfected (UT) or transfected with GFP or N-terminal Syntaxin4-GFP for 24 hours before lysing and analysis by immunoprecipitation/SDS-PAGE/Western blot. Munc18c immunoprecitates were first probed for Munc18c and Syntaxin4. Membranes were stripped and reprobed for GFP; two exposure times are shown to minimize antibody light chain over- saturation in the immunoprecipitation lanes.

IB: Syntaxin4

IB: Rb α Munc18c

Beads +

Ab

Beads +

Lysa

te

UT GFPGFP

UTN-term

N-term

Lysate IP: M α Munc18c

75 –

35 –

25 – IB: GFP

Page 56: Analysis of Munc18c and Syntaxin4 Function During Tumour

45

Figure 14: Munc18c knockdown or overexpression of N-terminal Syntaxin4 impairs cell migration. Transfected cells were lifted, counted and subjected to transwell migration assays. (A) Cells were transfected with GFP (control) or N-terminal Syntaxin4–GFP for 18 hours. (B) Cells were co-transfected with GFP and control siRNA or Munc18c siRNA for 38 hours. Means are +/- S.E.M from three independent experiments. Asterisks denote values significantly different from control (p<0.05).

0"

20"

40"

60"

80"

100"

120"

Control' STX4'N-term'

Cell'Migra4o

n'(%

'of'con

trol)'

*"

0"

20"

40"

60"

80"

100"

120"

Control'siRNA' Munc18c'siRNA'

Cell'Migra4o

n''(%

'of'con

trol)'

A

B

*

*

Fig. 5

Page 57: Analysis of Munc18c and Syntaxin4 Function During Tumour

46

Figure 15: Expression of GFP-Munc18c in HEK-293 cells. Cells were transfected and incubated for 20 hours prior to imaging with fluorescence microscopy. (A) Phase-contrast and GFP fluorescence overlay of HEK293 cells on tissue culture plate. (B) Zoom-in of GFP fluorescence. Scale bar, 10 µm.

A

B

Page 58: Analysis of Munc18c and Syntaxin4 Function During Tumour

47

undertaken to selectively pressure cells into expressing Munc18c. Cell lines

overexpressing full length Syntaxin4-GFP and N-terminal Syntaxin4-GFP were also

created for use in future experiments in addition to confirming previous results from

transient transfections. After selection and propagation of G418-resistant colonies, SDS-

PAGE and Western blot analyses were performed to assess expression of the proteins of

interest (Figure 16). Probing for Munc18c revealed no increase in Munc18c expression

relative to parental MDA-MB-231 cells. Probing for Syntaxin4 revealed protein bands at

the predicted molecular weights of ~63 kDa (full length Syntaxin4-GFP) and ~34 kDa

(endogenous Syntaxin4) in the full-length Syntaxin4-GFP cell line. A GFP antibody was

used to probe for N-terminal Syntaxin4-GFP, yielding a band at the predicted molecular

weight of ~31 kDa in the N-terminal Syntaxin4-GFP cell line. No observed effects on

MT1-MMP expression were seen in any of the cell lines.

Page 59: Analysis of Munc18c and Syntaxin4 Function During Tumour

48

Figure 16: Expression of Munc18c, GFP-Syntaxin4-full length or GFP-Syntaxin4-N-terminus in stable cell lines. Stable cell lines were generated to express untagged Munc18c, Syntaxin4-FL-GFP and N-terminal Syntaxin4 – GFP. Parental MDA-MB-231 cells were lysed in conjunction with stable cell lines and lysates were analyzed via SDS-PAGE/Western blot.

IB: Syntaxin4

IB: β1 integrin

IB: Munc18c

IB: Syntaxin4

IB: Actin

IB: GFP

135 –

75 –

63 –

50 –

37 –

31–

Paren

tal

Munc18c

STX4 FL

STX4 N-te

rm

Munc18c

STX4 FL

STX4 N-te

rm

WT

IB: Actin

IB: MT1-MMP

50–

50–

37–

Page 60: Analysis of Munc18c and Syntaxin4 Function During Tumour

49

3.8 Evaluating invadopodium formation and cell migration by stable cell lines The stable cell lines created were subjected to invadopodium formation assays

and transwell migration assays. Cell lines were seeded onto coverslips coated with

AlexaFluor-594-labelled gelatin and incubated for 4 hours prior to fixation and staining

for F-actin (Figure 17). Relative to parental MDA-MB-231 cells, no significant change in

invadopodium formation was seen for Munc18c and full-length Syntaxin4-GFP cell lines

(Figure 18). The N-terminal Syntaxin4-GFP cell line displayed approximately a 60%

decrease in invadopodium formation. Transwell migration assays revealed no significant

change for Munc18c and full-length Syntaxin4-GFP cell lines, but the N-terminal

Syntaxin4 cell line showed approximately a 60% decrease in cell migration.

Page 61: Analysis of Munc18c and Syntaxin4 Function During Tumour

50

Figure 17: Analysis of invadopodium formation by stable cell lines. Parental MDA-MB-231 cells or stable cell lines expressing untagged Munc18c, Syntaxin4-FL-GFP or N-terminal Syntaxin4–GFP were seeded onto AlexaFluor-594-labeled gelatin and incubated for 4 hours prior to fixation, staining for F-actin and analysis by confocal microscopy. Scale bar, 10 µm.

Mun

c18c

STX4

FL

STX4

Nte

rmPa

rent

al

F-actin Gelatin OverlayGFP

Page 62: Analysis of Munc18c and Syntaxin4 Function During Tumour

51

Figure 18: Quantification of invadopodium formation and cell migration by stable cell lines. (A) Cells with F-actin punctae overlying dark spots of gelatin degradation were counted as cells forming invadopodia. Percentages of cells forming invadopodia are presented as means +/- S.E.M. from three independent experiments in which 100 cells per sample were counted and normalized to parental MDA-MB-231 cells. (B) Stable cell lines were lifted, counted and subjected to transwell migration assays. Means +/- S.E.M. are presented from three independent experiments. Asterisks denote values significantly different from control (p<0.05).

0"

20"

40"

60"

80"

100"

120"

Munc18c' STX4FL' STX4Nterm'

Invado

podium

'Forma:

on'

(%'of'con

trol)'

0"

20"

40"

60"

80"

100"

120"

Munc18c' STX4FL' STX4Nterm'

Cell'Migra:o

n'(%

'of'con

trol)'

*

*

A

B

Page 63: Analysis of Munc18c and Syntaxin4 Function During Tumour

52

4.0 Discussion

The field of study pertaining to intracellular trafficking of proteins that are part of

the molecular machinery utilized by cells during tumour cell invasion has implications on

the establishment of cancer therapies. Previous work has shown the importance of

SNARE-mediated trafficking in all aspects of tumour cell invasion including:

invadopodium formation, cell migration, cell adhesion, and MMP-directed ECM

degradation. Although SNAREs have been identified to play an important role in

membrane trafficking events during the above cellular processes, there is no direct

knowledge of the role SNARE regulation has during tumour cell invasion. A recent study

has demonstrated an important role for the Q-SNARE, Syntaxin4, during tumour cell

invasion by MDA-MB-231 cells (Williams et al., 2014). Here, we demonstrate the

cognate SM protein of Syntaxin4, Munc18c, contributes to cellular migration and

invadopodium formation in these cells. These data may provide insight into the

mechanism of SNARE regulation during tumour cell invasion.

SM proteins and their cognate Syntaxins form strong interactions. Furthermore,

both of these proteins have been shown to be important components of the membrane

fusion machinery (Carr & Rizo, 2010). Munc18c and Syntaxin4 have been well

documented to associate in cell lines such as 3T3-L1 adipocytes (Thurmond et al., 2000;

Yu et al., 2013), but their association has not been demonstrated in invasive cancer cell

lines. Thus, the first step in exploring a possible function for Munc18c during tumour cell

invasion was to determine if Syntaxin4 and Munc18c associate in MDA-MB-231 cells in

culture. Co-immunoprecipitation experiments indicated that Munc18c and Syntaxin4

associate. Furthermore, co-localization analysis by confocal microscopy suggested the

Page 64: Analysis of Munc18c and Syntaxin4 Function During Tumour

53

association observed by co-immunoprecipitation was occurring at the plasma membrane.

These findings are consistent with a role for Munc18c in regulating Syntaxin4-mediated

fusion events at the plasma membrane. Experiments showing the staining pattern of

Munc18c and Syntaxin4, compared to the localization of a plasma membrane marker can

be performed to confirm plasma membrane localization, for example using a construct

that encodes the acylation motif of Lyn kinase tagged with GFP (Teruel, Blanpied, Shen,

Augustine, & Meyer, 1999; Williams & Coppolino, 2014). Additionally, the detection of

Syntaxin4 and Munc18c in the mid-cell region, presumably around the nucleus, suggests

that these two proteins are also found on the endoplasmic reticulum. Future experiments

will have to assess the staining pattern of calnexin, an endoplasmic reticulum-specific

protein, relative to Syntaxin4 and Munc18c to confirm this possibility.

The evident association of Munc18c and Syntaxin4 prompted the evaluation of

changes to this association in the context of invadopodium formation. Cells seeded onto

PLL and gelatin both resulted in a significant increase in the amount of Syntaxin4

associating with Munc18c when compared to unlifted cells. The increase in Munc18c and

Syntaxin4 association was greater in cells seeded onto gelatin (invadopodia formation)

than those seeded onto PLL, a non-ECM substrate. The increased association when cells

were seeded onto PLL suggests that Munc18c and Syntaxin4 may have a putative role

during cell spreading and not during integrin-mediated cell attachment. Integrin-

mediated cell attachment, and the signalling pathways this stimulates are minimal when

cells are seeded onto PLL. Negatively charged cell surface proteins are proposed to

mediate the attachment to positively-charged PLL (Mazia, 1975). Future experiments will

need to evaluate the co-immunoprecipitates of Syntaxin4 with Munc18c from cells in

Page 65: Analysis of Munc18c and Syntaxin4 Function During Tumour

54

suspension, to confirm if their association is a result of cell-substratum interactions. It is

predicted that there will be less Syntaxin4 associating with Munc18c in suspended cells

relative to unlifted cells, because there will be no cell-substratum interactions to support

cell spreading or integrin-mediated signalling.

A previous study has demonstrated that serine/threonine phosphorylation of

Syntaxin4 in MDA-MB-231 cells was significantly reduced during invadopodium

formation, compared to cells seeded onto PLL (Williams et al., 2014). Another study has

shown that phosphorylation of Syntaxin4 and Munc18c inhibits their interaction (Fu,

Naren, Gao, Ahmmed, & Malik, 2005). Therefore, it is reasonable to speculate that

increased association of Syntaxin4 with Munc18c during invadopodium formation is a

result of changes in Syntaxin4 serine/threonine phosphorylation. Future experiments to

analyze the phosphorylation state of Munc18c-associated Syntaxin4 and Munc18c will

extend our understanding of the association between Munc18c and Syntaxin4 during

invadopodium formation in MDA-MB-231 cells.

The increased association of Munc18c and Syntaxin4 during invadopodium

formation prompted an analysis of the subcellular localization of Munc18c during this

process using confocal microscopy. Syntaxin4, MT1-MMP, and F-actin have been shown

to localize to the center of sites of invadopodium formation when invasive cell lines are

seeded onto an ECM substrate (Oser et al., 2009; Williams et al., 2014). β1 integrin has

been implicated in the initiation of invadopodia (Artym et al., 2015; Destaing et al., 2010;

Williams & Coppolino, 2014), in addition to their maturation, as exemplified by integrin

containing adhesion rings around focal sites of invadopodium degradation (Branch,

Hoshino, & Weaver, 2012). When analyzed under confocal microscopy, Munc18c was

Page 66: Analysis of Munc18c and Syntaxin4 Function During Tumour

55

distributed throughout the confocal plane representative of the ventral plasma membrane.

The location of the ventral plasma membrane was indicated by the strongest signal given

by invadopodium markers. Co-localization of Munc18c and Syntaxin4 observed in

unlifted cells was not consistent with localization observed during invadopodium

formation. It is plausible the observed co-localization of Munc18c and Syntaxin4 in

unlifted cells may be due to Munc18c priming Syntaxin4 for vesicle fusion events.

During invadopodium formation, the requirement for Syntaxin4-mediated vesicle fusion

is stimulated, resulting in changes to Munc18c localization relative to Syntaxin4.

Additionally, given the observed distribution of Munc18c at the cell periphery, it could

be suggested that Munc18c function is not exclusive to invadopodium formation, and

may therefore be involved in a variety of unidentified Syntaxin4-mediated exocytic

events within these cells. When analyzing invadopodia specifically, Munc18c was found

at some of the edges of sites of degradation showing stronger co-localization with β1

integrin. This suggests that Munc18c may be involved in regulating Syntaxin4-mediated

exocytic events that promote the maturation of invadopodia.

The potential role of Munc18c during invadopodium formation prompted an

analysis of Munc18c inhibition using RNAi-mediated knockdown to reduce Munc18c

expression. Previous studies have shown that the depletion or RNAi-mediated

knockdown of Munc18c perturbed Syntaxin4-mediated exocytic events, such as GLUT4

translocation (Jewell et al., 2011; Oh & Thurmond, 2009). Also, RNAi-mediated

knockdown of its cognate SNARE partner, Syntaxin4, in MDA-MB-231 cells was found

to significantly perturb invadopodium formation and cellular invasion, but not cell

migration (Williams et al., 2014). In the present study, RNAi-mediated knockdown of

Page 67: Analysis of Munc18c and Syntaxin4 Function During Tumour

56

Munc18c was observed to significantly inhibit both invadopodium formation and cell

migration, suggesting that the presence of Munc18c facilitates these processes. The

inhibition of cell migration is surprising, given the previous study that found no effect on

cell migration (Williams et al., 2014). Cell migration requires the precise localization of

adhesion complexes and polarized recycling of integrins (Bravo-Cordero et al., 2012). A

previous study showed that Munc18c stabilizes Syntaxin4 at the plasma membrane,

preventing disorganized intracellular localization of Syntaxin4 (Torres et al., 2011). This

study also showed that depletion of Syntaxin4 leads to the intracellular localization of

proteins such as β1 integrin. The phenotype observed by Williams et al., 2014 might

therefore be due to the cellular compensation for depleted Syntaxin4 by using an

alternative SNARE-mediated trafficking pathway during cell migration, but not during

invadopodium formation. Torres et al., 2011 also showed that Munc18c depletion did not

alter Syntaxin4 localization to the plasma membrane. Therefore, in the present study, the

available Syntaxin4 at the plasma membrane in MDA-MB-231 cells may require

Munc18c to mediate the membrane fusion of key proteins during invadopodium

formation and cell migration. Future experiments will need to verify the intracellular

localization of Syntaxin4 and β1 integrin during invadopodium formation in Munc18c

depleted cells. It is predicted that there will be no alterations in Syntaxin4 plasma

membrane localization and there will be an increased intracellular accumulation of β1

integrin. Taken together, the observations here support a model wherein Syntaxin4-

mediated exocytic machinery, and trafficking of proteins required for cell migration and

invadopodium formation, is dependent on Munc18c.

Page 68: Analysis of Munc18c and Syntaxin4 Function During Tumour

57

A second method to inhibit Munc18c utilized the N-terminal 29 amino acids of

Syntaxin4. The N-terminal domain of Syntaxin4 forms facilitates binding between

Munc18c and Syntaxin4 (Hu et al., 2007). A previous study demonstrated that the

presence of this small polypeptide inhibited recombinant Munc18c and Syntaxin4

interactions using pull-down experiments (Latham et al., 2006). Here, we expressed a

GFP-tagged version of this N-terminal domain and observed a significant decrease in

both invadopodium formation and cell migration. These results support a role for

Munc18c and Syntaxin4 interactions during invadopodium formation and cell migration.

Attempts to detect an interaction between Munc18c and N-terminal Syntaxin4-GFP were

unsuccessful. These results may be due to the difference in transfection efficiency

between N-terminal Syntaxin4-GFP and GFP. Since there was little N-terminal

Syntaxin4-GFP detected in the lysate, this could suggest the co-immunoprecipitation

experiments were not sensitive enough to detect N-terminal Syntaxin4-GFP. Moreover,

the immunoprecipitation conditions and/or the bulkiness of the GFP-tag may be

contributing to the lack of N-terminal Syntaxin4-GFP co-immunoprecipitation. Future

experiments should employ a FLAG-tagged N-terminal Syntaxin4 – the size and

molecular weight of a FLAG-tag is much smaller than GFP. It is predicted that a FLAG-

tag will allow for the co-immunoprecipitation of N-terminal Syntaxin4 with Munc18c

and will produce a similar level of impairment with respect to invadopodium formation

and cell migration.

Munc18c overexpression studies were attempted using a transiently transfected

expression construct containing a CMV promoter upstream of the Munc18c ORF. Initial

experiments were performed using the mouse Munc18c homolog N-terminally tagged

Page 69: Analysis of Munc18c and Syntaxin4 Function During Tumour

58

with GFP. The expression of GFP-Munc18c in MDA-MB-231 cells was not successful as

revealed by fluorescence microscopy. The same result was seen using untagged human

Munc18c. Interestingly, expression of GFP was observed when HEK-293 cells were

transfected with mouse GFP-Munc18c. Future experiments should employ western blot

analyses to determine if the observed GFP fluorescence in HEK-293 cells is from GFP-

Munc18c and not GFP where Munc18c is removed by proteases.

To selectively pressure MDA-MB-231 cells into overexpressing Munc18c, stable

cell lines that were transfected with the untagged Munc18c construct were created.

Despite having propagated cells that were resistant to antibiotic, no Munc18c

overexpression was observed. These observations suggest that Munc18c expression

might be regulated at the post-transcriptional (such as miRNA-mediated inhibition) or

post-translational level (such as proteasomal degradation) and future experiments will

need to be performed to explain these results. Interestingly, several studies have shown

that Syntaxin4 and Munc18c expression are interdependent (Kanda et al., 2005; Torres et

al., 2011). However, in the present study, no alteration in Syntaxin4 expression was seen

in the Munc18c depleted cells. It is plausible that the expression of Munc18c was not

depleted enough to see a significant reduction in Syntaxin4.

Stable cell lines derived from MDA-MB-231 cells were created to overexpress N-

terminal Syntaxin4-GFP and Syntaxin4-FL-GFP in order to confirm previous results seen

with transient transfections. Interestingly, the cell line expressing N-terminal-Syntaxin4-

GFP had impaired invadopodium formation and cell migration relative to control parental

MDA-MB-231 cells and Syntaxin4-FL-GFP expressing cells. Further experiments will

need to be performed to elucidate how Syntaxin4-N-terminal-GFP is influencing cell

Page 70: Analysis of Munc18c and Syntaxin4 Function During Tumour

59

migration and invadopodium formation. Using these stable cell lines, sucrose gradient

centrifugation could be used as a tool to assess the intracellular localization of proteins

involved in invadopodium formation and cell migration. This technique allows for the

detection of proteins accumulated at the plasma membrane and endosomal compartments.

For example, N-terminal Syntaxin4-GFP could be interfering with endogenous Munc18c

and Syntaxin4 function leading to the accumulation of integrins in an endosomal

compartment. The current model of SM protein function is that SM proteins interact with

their cognate Syntaxin and then reposition to impart specificity in SNARE complex

formation (Baker et al., 2015; Yu et al., 2013). Therefore, if N-terminal Syntaxin4-GFP is

interfering with Munc18c and Syntaxin4 interactions within MDA-MB-231 cells then

there may be less Syntaxin4 interaction with cognate SNARE partners. VAMP7,

SNAP23, and Syntaxin4 are required for the delivery of MT1-MMP to sites of

invadopodium formation. Thus, it is plausible that during invadopodium formation

Syntaxin4 immunoprecipitates from the cell line expressing N-terminal Syntaxin4-GFP

would contain less VAMP7 and SNAP23, relative to parental MDA-MB-231 cells,

resulting from N-terminal Syntaxin4-GFP inhibition of SNARE complex formation.

Studies over the past decade have made it clear that trafficking of MMPs and

adhesion molecules has an essential role in the remodelling of the extracellular matrix

during tumour cell invasion, facilitating tumour cell migration through interstitial tissues.

The picture that has emerged is that formation of invadopodia and subsequent matrix

degradation in invasive tumour cells rely on the coordination of cytoskeletal and specific

membrane trafficking events. Membrane trafficking mediated by SNAREs, exocyst

complexes and other components of the membrane fusion machinery have been observed

Page 71: Analysis of Munc18c and Syntaxin4 Function During Tumour

60

to function during tumour cell invasion (Poincloux, Lizárraga, & Chavrier, 2009).

Nonetheless, a direct role that regulators of SNARE function have on SNARE-mediated

trafficking during tumour cell invasion has yet to be defined. To our knowledge, this

study is the only direct evidence for a role in SNARE regulation during tumour cell

invasion. Previous studies have identified gelsolin, an actin modifying protein, as an

interaction partner for Syntaxin4 (Kalwat et al., 2012), and gelsolin has been identified

separately to have a role during invadopodium formation (Crowley, Smith, Fang,

Takizawa, & Luna, 2009). Whether or not the interaction between gelsolin and Syntaxin4

influences invadopodium formation and cell migration, however, is not known. Thus, the

current study furthers our knowledge of the SNARE-mediated mechanisms that

coordinate the precise localization of proteins during tumour cell invasion. Our results

indicate that regulation of Syntaxin4-mediated events by Munc18c contributes to the

formation of invadopodia and migration by MDA-MB-231 cells.

Page 72: Analysis of Munc18c and Syntaxin4 Function During Tumour

61

5.0 Summary The aim of this study was to analyze the function of Munc18c and Syntaxin4

during tumour cell invasion by MDA-MB-231 cells. Using co-immunoprecipitation

experiments and confocal microscopy, an association between Munc18c and Syntaxin4

was observed. This association was enhanced when cells were seeded onto non-ECM

(PLL) or ECM (gelatin) substrates. These results suggest that the association of Munc18c

with Syntaxin4 in these cells is not exclusive to cellular processes regulated by cell-ECM

interactions. ECM degradation assays and confocal microscopy indicated Munc18c could

be involved in a variety of Syntaxin4-mediated events within MDA-MB-231 cells,

including the maturation of invadopodia. Munc18c knockdown was shown to impair

invadopodium formation and cell migration. The expression of N-terminal Syntaxin4-

GFP, designed to impair Munc18c and Syntaxin4 interactions within cells, reduced both

invadopodium formation and cell migration. These results were confirmed in a stable cell

line expressing this protein domain of Syntaxin4. Disruption of the interaction between

Munc18c and Syntaxin4 by N-terminal Syntaxin4-GFP could not be demonstrated using

co-immunoprecipitation experiments, and future experiments will be employed to

determine if N-terminal Syntaxin4 fused with a smaller epitope tag is interacting with

Munc18c. Taken together, these data suggest Munc18c interacts with Syntaxin4 in MDA-

MB-231 cells and this interaction contributes to cell migration and invadopodium

formation. Additional biochemical experiments are needed to fully elucidate the effects

of Munc18c knockdown and N-terminal Syntaxin4 expression. Together, the findings

support a model for Munc18c-mediated tumour cell invasion wherein Munc18c regulates

the formation of Syntaxin4-containing SNARE complexes that contribute to the

Page 73: Analysis of Munc18c and Syntaxin4 Function During Tumour

62

localization of proteins involved in cell migration, and invadopodium formation (Figure

19). Ultimately, these data contribute to our understanding of possible regulatory

mechanisms that control SNARE function during tumour cell invasion. The information

provided in this thesis could potentially be used to design cancer therapies that inhibit

SNARE regulation by non-SNARE proteins within tumour cells, and by extension

metastasis.

Page 74: Analysis of Munc18c and Syntaxin4 Function During Tumour

63

Figure 19: Proposed model of Munc18c function during tumour cell invasion Munc18c and Syntaxin4 interactions impart Syntaxin4 stability and specificity in SNARE complex formation. This process is carried out in the following steps: [1] Munc18c (purple) is initially associated with Syntaxin4 (red). Vesicles that contain proteins facilitating cell migration and invadopodium formation are trafficked to specific sites of the plasma membrane. [2] Munc18c repositions to mediate trans-SNARE complex formation with the cognate R-SNARE (blue) and Qbc SNARE (yellow) and subsequent vesicle fusion (not shown) with target membrane. [3] Munc18c dissociates from the cis-SNARE complex. [4] NSF and α-SNAP separate the cis-SNARE complex.

1 32 4

Syntaxin4

R-SNARE

QbcSNARE

NH3+

NH3+

NH3+

+H3N

Munc18c

Page 75: Analysis of Munc18c and Syntaxin4 Function During Tumour

64

6.0 References Adams, J. C. (2002). Molecular organisation of cell-matrix contacts: essential

multiprotein assemblies in cell and tissue function. Expert Reviews in Molecular Medicine, 4, 1–24.

Andrew, D. J., & Ewald, A. J. (2010). Morphogenesis of epithelial tubes: Insights into tube formation, elongation, and elaboration. Developmental Biology, 341, 34–55.

Artym, V. V. (2002). Molecular proximity of seprase and the urokinase-type plasminogen activator receptor on malignant melanoma cell membranes: dependence on beta1 integrins and the cytoskeleton. Carcinogenesis, 23, 1593–1602.

Artym, V. V., Swatkoski, S., Matsumoto, K., Campbell, C. B., Petrie, R. J., Dimitriadis, E. K., et al. (2015). Dense fibrillar collagen is a potent inducer of invadopodia via a specific signaling network. The Journal of Cell Biology, 208, 331–350.

Artym, V. V., Zhang, Y., Seillier-Moiseiwitsch, F., Yamada, K. M., & Mueller, S. C. (2006). Dynamic Interactions of Cortactin and Membrane Type 1 Matrix Metalloproteinase at Invadopodia: Defining the Stages of Invadopodia Formation and Function. Cancer Research, 66, 3034–3043.

Baker, R. W., Jeffrey, P. D., Zick, M., Phillips, B. P., Wickner, W. T., & Hughson, F. M. (2015). A direct role for the Sec1/Munc18-family protein Vps33 as a template for SNARE assembly. Science, 349, 1111–1114.

Baldassarre, M. (2002). Dynamin Participates in Focal Extracellular Matrix Degradation by Invasive Cells. Molecular Biology of the Cell, 14, 1074–1084.

Band, A. M., Ali, H., Vartiainen, M. K., Welti, S., Lappalainen, P., Olkkonen, V. M., & Kuismanen, E. (2002). Endogenous plasma membrane t-SNARE syntaxin 4 is present in rab11 positive endosomal membranes and associates with cortical actin cytoskeleton. FEBS Letters, 531, 513–519.

Beaty, B. T., & Condeelis, J. (2014). Digging a little deeper: The stages of invadopodium formation and maturation. European Journal of Cell Biology, 93, 438–444.

Berrier, A. L., & Yamada, K. M. (2007). Cell-matrix adhesion. Journal of Cellular Physiology, 213, 565–573.

Blouw, B., Seals, D., Pass, I., Díaz, B., & Courtneidge, S. (2008). A role for the podosome/invadopodia scaffold protein Tks5 in tumor growth in vivo. European Journal of Cell Biology, 87, 555–567.

Borradori, L., & Sonnenberg, A. (1999). Structure and Function of Hemidesmosomes: More Than Simple Adhesion Complexes. Journal of Investigative Dermatology, 112, 411–418.

Bracher, A., & Weissenhorn, W. (2002). Structural basis for the Golgi membrane recruitment of Sly1p by Sed5p. The EMBO Journal, 21, 6114–6124.

Branch, K. M., Hoshino, D., & Weaver, A. M. (2012). Adhesion rings surround invadopodia and promote maturation. Biology Open, 1, 711–722.

Brandie, F. M., Aran, V., Verma, A., McNew, J. A., Bryant, N. J., & Gould, G. W. (2008). Negative Regulation of Syntaxin4/SNAP-23/VAMP2-Mediated Membrane Fusion by Munc18c In Vitro. PLoS ONE, 3, e4074.

Bravo-Cordero, J. J., Hodgson, L., & Condeelis, J. (2012). Directed cell invasion and migration during metastasis. Current Opinion in Cell Biology, 24, 277–283.

Brenner, S. (1974). The genetics of Caenorhabditis elegans. Genetics, 251–256.

Page 76: Analysis of Munc18c and Syntaxin4 Function During Tumour

65

Brochetta, C., Vita, F., Tiwari, N., Scandiuzzi, L., Soranzo, M. R., Guérin-Marchand, C., et al. (2008). Involvement of Munc18 isoforms in the regulation of granule exocytosis in neutrophils. Biochimica Et Biophysica Acta (BBA) - Molecular Cell Research, 1783, 1781–1791.

Burgoyne, R. D., Barclay, J. W., Ciufo, L. F., Graham, M. E., Handley, M. T. W., & Morgan, A. (2009). The Functions of Munc18-1 in Regulated Exocytosis. Annals of the New York Academy of Sciences, 1152, 76–86.

Burns, S. (2001). Configuration of human dendritic cell cytoskeleton by Rho GTPases, the WAS protein, and differentiation. Blood, 98, 1142–1149.

Cabaniols, J. P., Ravichandran, V., & Roche, P. A. (1999). Phosphorylation of SNAP-23 by the Novel Kinase SNAK Regulates t-SNARE Complex Assembly. Molecular Biology of the Cell, 10, 4033–4041.

Carr, C. M., & Rizo, J. (2010). At the junction of SNARE and SM protein function. Current Opinion in Cell Biology, 22, 488–495.

Chen, W.-T. (1989). Proteolytic activity of specialized surface protrusions formed at rosette contact sites of transformed cells. Journal of Experimental Zoology, 251, 167–185.

Chung, S. H., Polgár, J., & Reed, G. L. (2000). Protein Kinase C Phosphorylation of Syntaxin 4 in Thrombin-activated Human Platelets. The Journal of Biological Chemistry, 275, 25286–25291.

Clark, E. S., & Weaver, A. M. (2008). A new role for cortactin in invadopodia: regulation of protease secretion. European Journal of Cell Biology, 87, 581–590.

Clark, E. S., Brown, B., Whigham, A. S., Kochaishvili, A., Yarbrough, W. G., & Weaver, A. M. (2008). Aggressiveness of HNSCC tumors depends on expression levels of cortactin, a gene in the 11q13 amplicon. Oncogene, 28, 431–444.

Clark, E. S., Whigham, A. S., Yarbrough, W. G., & Weaver, A. M. (2007). Cortactin Is an Essential Regulator of Matrix Metalloproteinase Secretion and Extracellular Matrix Degradation in Invadopodia. Cancer Research, 67, 4227–4235.

Coppolino, M. G., Kong, C., Mohtashami, M., Schreiber, A. D., Brumell, J. H., Finlay, B. B., et al. (2001). Requirement for N-Ethylmaleimide-sensitive Factor Activity at Different Stages of Bacterial Invasion and Phagocytosis. The Journal of Biological Chemistry, 276, 4772–4780.

Crowley, J. L., Smith, T. C., Fang, Z., Takizawa, N., & Luna, E. J. (2009). Supervillin reorganizes the actin cytoskeleton and increases invadopodial efficiency. Molecular Biology of the Cell, 20, 948–962.

Daley, W. P., Peters, S. B., & Larsen, M. (2008). Extracellular matrix dynamics in development and regenerative medicine. Journal of Cell Science, 121, 255–264.

Dean, R. A., Butler, G. S., Hamma-Kourbali, Y., Delbe, J., Brigstock, D. R., Courty, J., & Overall, C. M. (2007). Identification of Candidate Angiogenic Inhibitors Processed by Matrix Metalloproteinase 2 (MMP-2) in Cell-Based Proteomic Screens: Disruption of Vascular Endothelial Growth Factor (VEGF)/Heparin Affin Regulatory Peptide (Pleiotrophin) and VEGF/Connective Tissue Growth Factor Angiogenic Inhibitory Complexes by MMP-2 Proteolysis. Molecular and Cellular Biology, 27, 8454–8465.

Page 77: Analysis of Munc18c and Syntaxin4 Function During Tumour

66

Decock, J., Hendrickx, W., Vanleeuw, U., Van Belle, V., Van Huffel, S., Christiaens, M.-R., et al. (2008). Plasma MMP1 and MMP8 expression in breast cancer: Protective role of MMP8 against lymph node metastasis. BMC Cancer, 8, 77.

Destaing, O., Planus, E., Bouvard, D., Oddou, C., Badowski, C., Bossy, V., et al. (2010). β1A integrin is a master regulator of invadosome organization and function. Molecular Biology of the Cell, 21, 4108–4119.

Díaz, B., Yuen, A., Iizuka, S., Higashiyama, S., & Courtneidge, S. A. (2013). Notch increases the shedding of HB-EGF by ADAM12 to potentiate invadopodia formation in hypoxia. The Journal of Cell Biology, 201, 279–292.

Dulubova, I., Khvotchev, M., Liu, S., Huryeva, I., Südhof, T. C., & Rizo, J. (2007). Munc18-1 binds directly to the neuronal SNARE complex. Proceedings of the National Academy of Sciences of the United States of America, 104, 2697–2702.

Egeblad, M., & Werb, Z. (2002). New functions for the matrix metalloproteinases in cancer progression. Nature Reviews Cancer, 2, 161–174.

Foster, L. J., Yeung, B., Mohtashami, M., Ross, K., Trimble, W. S., & Klip, A. (1998). Binary Interactions of the SNARE Proteins Syntaxin-4, SNAP23, and VAMP-2 and Their Regulation by Phosphorylation †. Biochemistry, 37, 11089–11096.

Friedl, P., & Alexander, S. (2011). Cancer Invasion and the Microenvironment: Plasticity and Reciprocity. Cell, 147, 992–1009.

Fu, J., Naren, A. P., Gao, X., Ahmmed, G. U., & Malik, A. B. (2005). Protease-activated receptor-1 activation of endothelial cells induces protein kinase Calpha-dependent phosphorylation of syntaxin 4 and Munc18c: role in signaling p-selectin expression. The Journal of Biological Chemistry, 280, 3178–3184.

Gimona, M., Buccione, R., Courtneidge, S. A., & Linder, S. (2008). Assembly and biological role of podosomes and invadopodia. Current Opinion in Cell Biology, 20, 235–241.

Hanahan, D., & Weinberg, R. A. (2011). Hallmarks of Cancer: The Next Generation. Cell, 144, 646–674.

Hata, Y., Slaughter, C. A., & Südhof, T. C. (1993). Synaptic vesicle fusion complex contains unc-18 homologue bound to syntaxin. Nature, 366, 347–351.

Hojilla, C. V., Wood, G. A., & Khokha, R. (2008). Metalloproteinases as common effectors of inflammation and extracellular matrix breakdown in breast cancer. Breast Cancer Res.

Hong, W. (2005). SNAREs and traffic. Biochimica Et Biophysica Acta, 1744, 120–144. Hoshino, D., Kirkbride, K. C., Costello, K., Clark, E. S., Sinha, S., Grega-Larson, N., et

al. (2013). Exosome Secretion Is Enhanced by Invadopodia and Drives Invasive Behavior. Cell Reports, 5, 1159–1168.

Hotary, K. B., Allen, E. D., Brooks, P. C., Datta, N. S., Long, M. W., & Weiss, S. J. (2003). Membrane Type I Matrix Metalloproteinase Usurps Tumor Growth Control Imposed by the Three-Dimensional Extracellular Matrix. Cell, 114, 33–45.

Hu, S. H., Latham, C. F., Gee, C. L., James, D. E., & Martin, J. L. (2007). Structure of the Munc18c/Syntaxin4 N-peptide complex defines universal features of the N-peptide binding mode of Sec1/Munc18 proteins. Proceedings of the National Academy of Sciences of the United States of America, 104, 8773–8778.

Huttenlocher, A., & Horwitz, A. R. (2011). Integrins in cell migration. Cold Spring Harbor Perspectives in Biology, 3, a005074–a005074.

Page 78: Analysis of Munc18c and Syntaxin4 Function During Tumour

67

Jackson, B. C., Nebert, D. W., & Vasiliou, V. (2010). Update of human and mouse matrix metalloproteinase families. Human Genomics, 4, 194.

Jahn, R., & Scheller, R. H. (2006). SNAREs — engines for membrane fusion. Nature Reviews. Molecular Cell Biology, 7, 631–643.

Jewell, J. L., Oh, E., Ramalingam, L., Kalwat, M. A., Tagliabracci, V. S., Tackett, L., et al. (2011). Munc18c phosphorylation by the insulin receptor links cell signaling directly to SNARE exocytosis. The Journal of Cell Biology, 193, 185–199.

Jiang, W., Davies, G., Martin, T., Parr, C., Watkins, G., Mason, M., & Mansel, R. (2006). Expression of membrane type-1 matrix metalloproteinase, MT1-MMP in human breast cancer and its impact on invasiveness of breast cancer cells. International Journal of Molecular Medicine. doi:10.3892/ijmm.17.4.583

Kajita, M., Itoh, Y., Chiba, T., Mori, H., Okada, A., Kinoh, H., & Seiki, M. (2001). Membrane-Type 1 Matrix Metalloproteinase Cleaves Cd44 and Promotes Cell Migration. The Journal of Cell Biology, 153, 893–904.

Kalwat, M. A., Wiseman, D. A., Luo, W., Wang, Z., & Thurmond, D. C. (2012). Gelsolin Associates with the N Terminus of Syntaxin 4 to Regulate Insulin Granule Exocytosis. Molecular Endocrinology, 26, 128–141.

Kanda, H., Tamori, Y., Shinoda, H., Yoshikawa, M., Sakaue, M., Udagawa, J., et al. (2005). Adipocytes from Munc18c-null mice show increased sensitivity to insulin-stimulated GLUT4 externalization. Journal of Clinical Investigation, 115, 291–301.

Kean, M. J., Williams, K. C., Skalski, M., Myers, D., Burtnik, A., Foster, D., & Coppolino, M. G. (2009). VAMP3, syntaxin-13 and SNAP23 are involved in secretion of matrix metalloproteinases, degradation of the extracellular matrix and cell invasion. Journal of Cell Science, 122, 4089–4098.

Kloepper, T. H., Kienle, C. N., & Fasshauer, D. (2007). An Elaborate Classification of SNARE Proteins Sheds Light on the Conservation of the Eukaryotic Endomembrane System. Molecular Biology of the Cell, 18, 3463–3471.

Latham, C. F., Lopez, J. A., Hu, S. H., Gee, C. L., Westbury, E., Blair, D. H., et al. (2006). Molecular Dissection of the Munc18c/Syntaxin4 Interaction: Implications for Regulation of Membrane Trafficking. Traffic, 7, 1408–1419.

Leong, H. S., Robertson, A. E., Stoletov, K., Leith, S. J., Chin, C. A., Chien, A. E., et al. (2014). Invadopodia Are Required for Cancer Cell Extravasation and Are a Therapeutic Target for Metastasis. Cell Reports, 8, 1558–1570.

Linder, S. (2007). The matrix corroded: podosomes and invadopodia in extracellular matrix degradation. Trends in Cell Biology, 17, 107–117.

Linder, S., Hufner, K., Wintergerst, U., & Aepfelbacher, M. (2000). Microtubule-dependent formation of podosomal adhesion structures in primary human macrophages. Journal of Cell Science, 113 Pt 23, 4165–4176.

Linder, S., Wiesner, C., & Himmel, M. (2011). Degrading Devices: Invadosomes in Proteolytic Cell Invasion. Annual Review of Cell and Developmental Biology, 27, 185–211.

Llano, E., Pendás, A. M., Freije, J. P., Nakano, A., Knäuper, V., Murphy, G., & López-Otin, C. (1999). Identification and characterization of human MT5-MMP, a new membrane-bound activator of progelatinase a overexpressed in brain tumors. Cancer Research, 59, 2570–2576.

Page 79: Analysis of Munc18c and Syntaxin4 Function During Tumour

68

Lohmer, L. L., Kelley, L. C., Hagedorn, E. J., & Sherwood, D. R. (2014). Invadopodia and basement membrane invasion in vivo. Cell Adhesion & Migration, 8, 246–255.

Lu, P., Sternlicht, M. D., & Werb, Z. (2006). Comparative Mechanisms of Branching Morphogenesis in Diverse Systems. Journal of Mammary Gland Biology and Neoplasia, 11, 213–228.

Lu, P., Takai, K., Weaver, V. M., & Werb, Z. (2011). Extracellular Matrix Degradation and Remodeling in Development and Disease. Cold Spring Harbor Perspectives in Biology, 3, a005058–a005058.

Mader, C. C., Oser, M., Magalhaes, M. A. O., Bravo-Cordero, J. J., Condeelis, J., Koleske, A. J., & Gil-Henn, H. (2011). An EGFR-Src-Arg-Cortactin Pathway Mediates Functional Maturation of Invadopodia and Breast Cancer Cell Invasion. Cancer Research, 71, 1730–1741.

Mattila, P. K., & Lappalainen, P. (2008). Filopodia: molecular architecture and cellular functions. Nature Reviews. Molecular Cell Biology, 9, 446–454.

Mazia, D. (1975). Adhesion of cells to surfaces coated with polylysine. Applications to electron microscopy. The Journal of Cell Biology, 66, 198–200.

McIntosh, S. (1995). Molecular Identification of Two Novel Munc-18 Isoforms Expressed in Non-neuronal Tissues. Journal of Biological Chemistry, 270, 5857–5863.

Misura, K. M. S., Scheller, R. H., & Weis, W. I. (2000). Three-dimensional structure of the neuronal-Sec1–syntaxin 1a complex. Nature, 404, 355–362.

Miyata, T., Ohnishi, H., Suzuki, J., Yoshikumi, Y., Ohno, H., Mashima, H., et al. (2004). Involvement of syntaxin 4 in the transport of membrane-type 1 matrix metalloproteinase to the plasma membrane in human gastric epithelial cells. Biochemical and Biophysical Research Communications, 323, 118–124.

Moissoglu, K., & Schwartz, M. A. (2012). Integrin signalling in directed cell migration. Biology of the Cell, 98, 547–555.

Monsky, W. L., Lin, C. Y., Aoyama, A., Kelly, T., Akiyama, S. K., Mueller, S. C., & Chen, W. T. (1994). A potential marker protease of invasiveness, seprase, is localized on invadopodia of human malignant melanoma cells. Cancer Research, 54, 5702–5710.

Murphy, D. A., & Courtneidge, S. A. (2011). The 'ins' and “outs” of podosomes and invadopodia: characteristics, formation and function. Nature Reviews. Molecular Cell Biology, 12, 413–426.

Neel, N. F., Rossman, K. L., Martin, T. D., Hayes, T. K., Yeh, J. J., & Der, C. J. (2012). The RalB Small GTPase Mediates Formation of Invadopodia through a GTPase-Activating Protein-Independent Function of the RalBP1/RLIP76 Effector. Molecular and Cellular Biology, 32, 1374–1386.

Oh, E., & Thurmond, D. C. (2009). Munc18c Depletion Selectively Impairs the Sustained Phase of Insulin Release. Diabetes, 58, 1165–1174.

Oh, E., Spurlin, B. A., Pessin, J. E., & Thurmond, D. C. (2005). Munc18c Heterozygous Knockout Mice Display Increased Susceptibility for Severe Glucose Intolerance. Diabetes, 54, 638–647.

Oser, M., Yamaguchi, H., Mader, C. C., Bravo-Cordero, J. J., Arias, M., Chen, X., et al. (2009). Cortactin regulates cofilin and N-WASp activities to control the stages of invadopodium assembly and maturation. The Journal of Cell Biology, 186, 571–587.

Page 80: Analysis of Munc18c and Syntaxin4 Function During Tumour

69

Oskarsson, T. (2013). Extracellular matrix components in breast cancer progression and metastasis. Breast (Edinburgh, Scotland), 22 Suppl 2, S66–72.

Pan, P.-Y., Tian, J.-H., & Sheng, Z.-H. (2009). Snapin Facilitates the Synchronization of Synaptic Vesicle Fusion. Neuron, 61, 412–424.

Pevsner, J., Hsu, S. C., & Scheller, R. H. (1994). n-Sec1: a neural-specific syntaxin-binding protein. Proceedings of the National Academy of Sciences of the United States of America, 91, 1445–1449.

Poincloux, R., Lizárraga, F., & Chavrier, P. (2009). Matrix invasion by tumour cells: a focus on MT1-MMP trafficking to invadopodia. Journal of Cell Science, 122, 3015–3024.

Polgár, J., Lane, W. S., Chung, S. H., Houng, A. K., & Reed, G. L. (2003). Phosphorylation of SNAP-23 in Activated Human Platelets. The Journal of Biological Chemistry, 278, 44369–44376.

Pombo, I., Martin-Verdeaux, S., Iannascoli, B., Le Mao, J., Deriano, L., Rivera, J., & Blank, U. (2001). IgE receptor type I-dependent regulation of a Rab3D-associated kinase: a possible link in the calcium-dependent assembly of SNARE complexes. The Journal of Biological Chemistry, 276, 42893–42900.

Puri, N., & Roche, P. A. (2006). Ternary SNARE Complexes Are Enriched in Lipid Rafts during Mast Cell Exocytosis. Traffic, 7, 1482–1494.

Revach, O. Y., & Geiger, B. (2014). The interplay between the proteolytic, invasive, and adhesive domains of invadopodia and their roles in cancer invasion. Cell Adhesion & Migration. doi:10.1073/pnas.94.15.7959

Ridley, A. J., Schwartz, M. A., Burridge, K., Firtel, R. A., Ginsberg, M. H., Borisy, G., et al. (2003). Cell migration: integrating signals from front to back. Science, 302, 1704–1709.

Risinger, C., & Bennett, M. K. (2002). Differential Phosphorylation of Syntaxin and Synaptosome-Associated Protein of 25 kDa (SNAP-25) Isoforms. Journal of Neurochemistry, 72, 614–624.

Sabeh, F. (2004). Tumor cell traffic through the extracellular matrix is controlled by the membrane-anchored collagenase MT1-MMP. The Journal of Cell Biology, 167, 769–781.

Sabeh, F., Shimizu-Hirota, R., & Weiss, S. J. (2009). Protease-dependent versus -independent cancer cell invasion programs: three-dimensional amoeboid movement revisited. The Journal of Cell Biology, 185, 11–19.

Sato, H., Takino, T., Okada, Y., Cao, J., Shinagawa, A., Yamamoto, E., & Seiki, M. (1994). A matrix metalloproteinase expressed on the surface of invasive tumour cells. Nature, 370, 61–65.

Schollmeier, Y., Krause, J. M., Kreye, S., Malsam, J., & Söllner, T. H. (2011). Resolving the Function of Distinct Munc18-1/SNARE Protein Interaction Modes in a Reconstituted Membrane Fusion Assay. The Journal of Biological Chemistry, 286, 30582–30590.

Schraw, T. D., Crawford, G. L., Ren, Q., Choi, W., Thurmond, D. C., Pessin, J., & Whiteheart, S. W. (2004). Platelets from Munc18c heterozygous mice exhibit normal stimulus-induced release. Thrombosis and Haemostasis, 92, 829–837.

Page 81: Analysis of Munc18c and Syntaxin4 Function During Tumour

70

Seftor, R. E., Seftor, E. A., Koshikawa, N., Meltzer, P. S., Gardner, L. M., Bilban, M., et al. (2001). Cooperative interactions of laminin 5 gamma2 chain, matrix metalloproteinase-2, and membrane type-1-matrix/metalloproteinase are required for mimicry of embryonic vasculogenesis by aggressive melanoma. Cancer Research, 61, 6322–6327.

Skalski, M., & Coppolino, M. G. (2005). SNARE-mediated trafficking of α5β1 integrin is required for spreading in CHO cells. Biochemical and Biophysical Research Communications, 335, 1199–1210.

Sodek, K. L., Ringuette, M. J., & Brown, T. J. (2007). MT1-MMP is the critical determinant of matrix degradation and invasion by ovarian cancer cells. British Journal of Cancer, 97, 358–367.

Sohail, A., Sun, Q., Zhao, H., Bernardo, M. M., Cho, J.-A., & Fridman, R. (2008). MT4-(MMP17) and MT6-MMP (MMP25), A unique set of membrane-anchored matrix metalloproteinases: properties and expression in cancer. Cancer Metastasis Reviews, 27, 289–302.

Söllner, T., Bennett, M. K., Whiteheart, S. W., Scheller, R. H., & Rothman, J. E. (1993a). A protein assembly-disassembly pathway in vitro that may correspond to sequential steps of synaptic vesicle docking, activation, and fusion. Cell, 75, 409–418.

Söllner, T., Whiteheart, S. W., Brunner, M., Erdjument-Bromage, H., Geromanos, S., Tempst, P., & Rothman, J. E. (1993b). SNAP receptors implicated in vesicle targeting and fusion. Nature, 362, 318–324.

Steffen, A., Le Dez, G., Poincloux, R., Recchi, C., Nassoy, P., Rottner, K., et al. (2008). MT1-MMP-Dependent Invasion Is Regulated by TI-VAMP/VAMP7. Current Biology, 18, 926–931.

Südhof, T. C., & Rothman, J. E. (2009). Membrane Fusion: Grappling with SNARE and SM Proteins. Science, 323, 474–477.

Takino, T., Sato, H., Shinagawa, A., & Seiki, M. (1995). Identification of the Second Membrane-type Matrix Metalloproteinase (MT-MMP-2) Gene from a Human Placenta cDNA Library: MT-MMPs FORM A UNIQUE MEMBRANE-TYPE SUBCLASS IN THE MMP FAMILY. The Journal of Biological Chemistry, 270, 23013–23020.

Talmadge, J. E., & Fidler, I. J. (2010). AACR centennial series: the biology of cancer metastasis: historical perspective. Cancer Research, 70, 5649–5669.

Tamori, Y., Kawanishi, M., Niki, T., Shinoda, H., Araki, S., Okazawa, H., & Kasuga, M. (1998). Inhibition of Insulin-induced GLUT4 Translocation by Munc18c through Interaction with Syntaxin4 in 3T3-L1 Adipocytes. The Journal of Biological Chemistry, 273, 19740–19746.

Tayeb, M. A., Skalski, M., Cha, M. C., Kean, M. J., Scaife, M., & Coppolino, M. G. (2005). Inhibition of SNARE-mediated membrane traffic impairs cell migration. Experimental Cell Research, 305, 63–73.

Tehrani, S., Tomasevic, N., Weed, S., Sakowicz, R., & Cooper, J. A. (2007). Src phosphorylation of cortactin enhances actin assembly. Proceedings of the National Academy of Sciences of the United States of America, 104, 11933–11938.

Tellam, J. T., Macaulay, S. L., McIntosh, S., & Hewish, D. R. (1997). Characterization of Munc-18c and Syntaxin-4 in 3T3-L1 Adipocytes. Journal of Biological …. doi:10.1074/jbc.272.10.6179

Page 82: Analysis of Munc18c and Syntaxin4 Function During Tumour

71

Teng, F., Wang, Y., & Tang, B. L. (2001). The syntaxins. Genome Biology, 2, reviews3012.1.

Teruel, M. N., Blanpied, T. A., Shen, K., Augustine, G. J., & Meyer, T. (1999). A versatile microporation technique for the transfection of cultured CNS neurons. Journal of Neuroscience Methods, 93, 37–48.

Thurmond, D. C., Kanzaki, M., Khan, A. H., & Pessin, J. E. (2000). Munc18c Function Is Required for Insulin-Stimulated Plasma Membrane Fusion of GLUT4 and Insulin-Responsive Amino Peptidase Storage Vesicles. Molecular and Cellular Biology, 20, 379–388.

Tolde, O., Rösel, D., Veselý, P., Folk, P., & Brábek, J. (2010). The structure of invadopodia in a complex 3D environment. European Journal of Cell Biology, 89, 674–680.

Tomes, C. N. (2015). The proteins of exocytosis: lessons from the sperm model. Biochemical Journal, 465, 359–370.

Toonen, R. F. G., & Verhage, M. (2007). Munc18-1 in secretion: lonely Munc joins SNARE team and takes control. Trends in Neurosciences, 30, 564–572.

Torres, J., Funk, H. M., Zegers, M. M. P., & Beest, ter, M. B. A. (2011). The syntaxin 4 N terminus regulates its basolateral targeting by munc18c-dependent and -independent mechanisms. The Journal of Biological Chemistry, 286, 10834–10846.

Ungar, D., & Hughson, F. M. (2003). SNARE protein structure and function. Annual Review of Cell and Developmental Biology, 19, 493–517.

Vicente-Manzanares, M. (2005). Cell migration at a glance. Journal of Cell Science, 118, 4917–4919.

Widberg, C. H. (2003). Tomosyn Interacts with the t-SNAREs Syntaxin4 and SNAP23 and Plays a Role in Insulin-stimulated GLUT4 Translocation. The Journal of Biological Chemistry, 278, 35093–35101.

Williams, K. C., & Coppolino, M. G. (2014). SNARE-dependent interaction of Src, EGFR and 1 integrin regulates invadopodia formation and tumor cell invasion. Journal of Cell Science, 127, 1712–1725.

Williams, K. C., McNeilly, R. E., & Coppolino, M. G. (2014). SNAP23, Syntaxin4, and vesicle-associated membrane protein 7 (VAMP7) mediate trafficking of membrane type 1-matrix metalloproteinase (MT1-MMP) during invadopodium formation and tumor cell invasion. Molecular Biology of the Cell, 25, 2061–2070.

Wolf, K., Wu, Y. I., Liu, Y., Geiger, J., Tam, E., Overall, C., et al. (2007). Multi-step pericellular proteolysis controls the transition from individual to collective cancer cell invasion. Nature Cell Biology, 9, 893–904.

Yamaguchi, H. (2012). Pathological roles of invadopodia in cancer invasion and metastasis. European Journal of Cell Biology, 91, 902–907.

Yamaguchi, H., Lorenz, M., Kempiak, S., Sarmiento, C., Coniglio, S., Symons, M., et al. (2005). Molecular mechanisms of invadopodium formation: the role of the N-WASP-Arp2/3 complex pathway and cofilin. The Journal of Cell Biology, 168, 441–452.

Yelamanchili, S. V., Reisinger, C., Becher, A., Sikorra, S., Bigalke, H., Binz, T., & Ahnert-Hilger, G. (2005). The C-terminal transmembrane region of synaptobrevin binds synaptophysin from adult synaptic vesicles. European Journal of Cell Biology, 84, 467–475.

Page 83: Analysis of Munc18c and Syntaxin4 Function During Tumour

72

Yu, H., Rathore, S. S., Lopez, J. A., Davis, E. M., James, D. E., Martin, J. L., & Shen, J. (2013). Comparative studies of Munc18c and Munc18-1 reveal conserved and divergent mechanisms of Sec1/Munc18 proteins. Proceedings of the National Academy of Sciences of the United States of America, 110, E3271–80.

Zamir, E., Katz, M., Posen, Y., Erez, N., Yamada, K. M., Katz, B. Z., et al. (2000). Dynamics and segregation of cell-matrix adhesions in cultured fibroblasts. Nature Cell Biology, 2, 191–196.

Zucker, S., Hymowitz, M., Conner, C., Zarrabi, H. M., Hurewitz, A. N., Matrisian, L., et al. (1999). Measurement of matrix metalloproteinases and tissue inhibitors of metalloproteinases in blood and tissues. Clinical and experimental applications. Annals of the New York Academy of Sciences, 878, 212–227.