261
The Role of STXBP5, VAMP8, and SNAP23 in Endothelial Exocytosis by Qiuyu Zhu Submitted in Partial Fulfillment of the Requirements for the Degree Doctor of Philosophy Supervised by Professor Charles J. Lowenstein Department of Pharmacology and Physiology School of Medicine and Dentistry University of Rochester Rochester, New York 2015

The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

  • Upload
    others

  • View
    0

  • Download
    0

Embed Size (px)

Citation preview

Page 1: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

The Role of STXBP5, VAMP8, and SNAP23 in Endothelial Exocytosis

by

Qiuyu Zhu

Submitted in Partial Fulfillment of the

Requirements for the Degree

Doctor of Philosophy

Supervised by Professor Charles J. Lowenstein

Department of Pharmacology and Physiology

School of Medicine and Dentistry

University of Rochester

Rochester, New York

2015

Page 2: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

ii

Biographical Sketch

The author was born in Tengzhou, Shandong Province, People’s Republic of China. He

attended Xi’an Jiaotong University in 2003. He completed the Seven-Year Program of

Clinical Medicine (equivalent to MD) with his internship and residency in Internal

Medicine, and he graduated with a BS/MS dual degree in Clinical Medicine in 2010. He

began his doctoral studies in Physiology at the University of Rochester in 2010. He

earned the Master of Science in Physiology in 2012. He was awarded the Howard

Hughes Medical Institute "Med-into-Grad" Fellowship in Cardiovascular Science from

2010 to 2012 and received cardiovascular-oriented clinical and translational research

training. He was awarded an American Heart Association (AHA) Predoctoral Fellowship

from 2013 to 2015. He received the 2014 Travel Award for Young Investigators from the

AHA’s Council on Arteriosclerosis, Thrombosis, and Vascular Biology (ATVB), as well

as the Travel Funds for PhD Students awarded by the University Dean of Graduate

Studies in 2014, and he presented part of this thesis work at the Sol Sherry Distinguished

Lecture in Thrombosis at AHA’s Scientific Sessions 2014. He pursued his research in

endothelial exocytosis and thrombosis under the direction of Charles J Lowenstein, MD.

The following publications are a result of work conducted during this doctoral study:

1. Nadtochiy SM, Zhu Q, Urciuoli W, Rafikov R, Black SM, and Brookes PS.

Nitroalkenes confer acute cardioprotection via adenine nucleotide translocase 1. The

Journal of Biological Chemistry. 2012;287(5):3573-80.

Page 3: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

iii

2. Zhu Q, Yamakuchi M, Ture S, de la Luz Garcia-Hernandez M, Ko KA, Modjeski

KL, LoMonaco MB, Johnson AD, O'Donnell CJ, Takai Y, Morrell CN, and

Lowenstein CJ. Syntaxin-binding protein STXBP5 inhibits endothelial exocytosis and

promotes platelet secretion. The Journal of Clinical Investigation.

2014;124(10):4503-16.

3. Zhu Q, Bao C, Ture S, Ferlito M, Morrell CN, Yamakuchi M, Lowenstein CJ.

VAMP8 Mediates Endothelial Granule Exocytosis. Blood. (in revision for

publication)

4. Zhu Q, Yamakuchi M, Lowenstein CJ. SNAP23 Regulates Endothelial Exocytosis.

PLOS One (in review for publication)

Published Abstracts:

1. Zhu Q, Yamakuchi M, Ture S, de la Luz Garcia-Hernandez M, Ko KA, Modjeski

KL, LoMonaco MB, Johnson AD, O'Donnell CJ, Takai Y, Morrell CN, and

Lowenstein CJ. STXBP5 Regulates Endothelial Exocytosis, Platelet Secretion and

Thrombosis. American Heart Association 2014 Scientific Sessions

2. Zhu Q, Bao C, Ture S, Ferlito M, Morrell CN, Yamakuchi M, and Lowenstein CJ.

VAMP8 Mediates Endothelial Granule Exocytosis. Arteriosclerosis, Thrombosis, and

Vascular Biology 2014 Scientific Sessions

Page 4: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

iv

Acknowledgments

I would like to thank my mentor Dr. Charles J Lowenstein. During the past five years, he

has not only been an inspiring role model, but also more importantly, as an original

thinker who imparted wisdom and unlimited intellectual stimulation to my doctoral study.

His mentoring has prepared me well to move forward in my career, which I appreciate

more than I can express. His word and deed will continue to illuminate and encourage my

future pursuit towards the great scientific unknown.

I have benefited tremendously from the invaluable guidance and support from my thesis

committee members Drs. Robert Dirksen, Ingrid Sarelius, and Craig Morrell, as well as

our collaborators Drs. Christopher O’Donnell (NIH, NHLBI, and Harvard Medical

School), Andrew Johnson (NIH and NHLBI), and Sidney Whiteheart (University of

Kentucky). I want to thank Professor James Palis for chairing my thesis defense

committee.

I want to thank all the Lowenstein lab members, previous and current, for making my

personal ecosystem equally productive and fun. This includes an enormous debt of

gratitude to Dr. Munekazu Yamakuchi, who systematically taught me the methodology in

vascular biology, and imbued me with an exceptional standard of scientific rigor and

work ethic. Dr. Maria de la Luz Garcia-Hernandez offered substantial help to my project,

as well as constant support as a bench-side mentor. Michael LoMonaco gave me valuable

technical insights and made sure that everything “under the hood” always runs smoothly.

Page 5: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

v

Previous lab members Drs. Kenji Matsushita, Shusuke Yagi, Takashi Ito, Amit

Dhamoon, and Vijay Vanchinathan, showed me by example how scientists would study,

think, and work.

I have been blessed to work with the most extraordinary and dynamic colleagues in the

Morrell lab, who enriched my life in science and contributed to the body of this work. I

want to extend my thanks to perceptive consultant Dr. Craig N. Morrell, who guided me

through many critical steps of my study. Sara Ture assisted in technically challenging in

vivo experiments, and her exemplary work taught me many lessons in physiology. I want

to thank Dr. Scott Cameron, Dr. Angela Aggrey, David Field, and Lesley Chapman for

offering valuable suggestions and support, and for sharing frustrations and successes in

research. Thank you, Kristina Modjeski, for your advice and friendship.

I am indebted to the Department of Pharmacology and Physiology, and to the Aab CVRI

for the environments I have been privileged to work in, and for offering me many great

exemplars and teachers. This includes Dr. Joseph Miano, for his advice, guidance, and

stimulating discussions. This also includes Dr. Burns Blaxall and Dr. Paul Brookes with

whom I have apprenticed, and who encouraged my passions in science.

Thanks also to the CVRI microsurgical core and Dr. Kyung Ae Ko, whose

meticulousness and exceptional surgical skills helped address many challenging

questions.

Page 6: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

vi

I want to thanks Linda Lipani and Sandra Morgan for their administrative effort and

dedication.

Last but not least, I want to thank all my friends and my family for their unending support

to my scientific aspirations. Most specially, I want to thank my wife, soul-mate and

intellectual partner, Cindy Yingya Zhou, for her devotion, inspiration, and selfless love.

Page 7: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

vii

Abstract

Thromboembolic diseases are major causes of morbidity and mortality. Endothelial

exocytosis plays a critical role in thrombosis. The exocytic machinery that regulates

granule secretion from endothelial cells (EC) is not completely defined. We hypothesized

that specific members of the soluble N-ethylmaleimide sensitive factor attachment

protein receptor (SNARE) superfamily and their regulatory partners mediate endothelial

granule exocytosis and thrombosis.

EC exocytosis releases von Willebrand factor (VWF), a major factor in thrombogenesis.

Recent genome-wide association studies (GWAS) identified syntaxin-binding protein 5

(STXBP5) as a candidate gene linked to changes in plasma VWF levels. We found

STXBP5 is expressed in human ECs and interacts with SNARE proteins. STXBP5

inhibits endothelial exocytosis in vitro. Stxbp5 KO mice had higher plasma VWF levels

and more P-selectin translocation than wild-type mice, suggesting STXBP5 inhibits

endothelial exocytosis in vivo. However, Stxbp5 KO mice also displayed impaired

hemostasis and thrombosis. Platelets from Stxbp5 KO mice had defects in secretion and

activation. Therefore, STXBP5 is a novel SNARE regulatory partner that inhibits

endothelial exocytosis, but promotes platelet secretion and thrombosis.

In secretory cells, a ternary complex that mediates exocytosis is comprised of three

SNARE molecules, including isoforms of the syntaxin (STX), vesicle-associated

membrane protein (VAMP), and synaptosomal-associated protein (SNAP) families.

Page 8: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

viii

Human ECs express proteins of the SNARE superfamily, but the exact identity of EC

SNAREs that regulate endothelial exocytosis has been unclear. We explored the SNARE

molecules required for endothelial exocytosis. We found VAMP8 co-localized with

Weibel-Palade bodies, the major endothelial granules. VAMP8 interacted with

components of the exocytic machinery. Knock down of VAMP8 expression inhibited

endothelial exocytosis. Vamp8 KO mice had decreased endothelial exocytosis. These data

suggest that VAMP8 plays a critical role in endothelial exocytosis.

In addition, we identified SNAP23 as the predominant endothelial SNAP isoform that

mediates endothelial exocytosis. SNAP23 was localized to the plasma membrane.

Knockdown of SNAP23 decreased endothelial exocytosis. SNAP23 also interacted with

the endothelial exocytic machinery. These data suggest that SNAP23 is another key

component of the endothelial SNARE machinery.

Taken together, the present work identified the role of STXBP5, VAMP8, and SNAP23

in EC exocytosis. Given the importance of EC exocytosis to vascular thrombosis and

inflammation, the insights gained from these studies may lead to novel targets for the

controlling of thromboembolic diseases.

Page 9: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

ix

Contributors and Funding Sources

This work was supported by a dissertation committee consisting of Professor Charles J.

Lowenstein (advisor) of the Department of Medicine, Professors Robert T. Dirksen and

Ingrid H. Sarelius of the Department of Pharmacology and Physiology, and Professor

Craig N. Morrell of the Department of Medicine, Aab Cardiovascular Research Institute.

The defense committee was chaired by Professor James Palis of the Department of

Pediatrics, Hematology and Oncology.

All work for the dissertation was completed by the author except for:

Chapter 2: GWAS data was analyzed in collaboration with Drs. Andrew D. Johnson and

Christopher J. O’Donnell (NHLBI and NHLBI’s Framingham Heart Study, Framingham,

MA). The initial expression and RNAi studies in HUVEC was performed by Dr.

Munekazu Yamakuchi (Figure 2-1A and Figure 2-2A-C). Mammalian expression

plasmid pMEX neo-3×FLAG-tomosyn (8)-ATG was provided by Dr. David James

(Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia), and FLAG-

STXBP5-N436S was designed and generated by Dr. Munekazu Yamakuchi. XS-VWF

protein was a generous gift of Dr. Sriram Neelamegham (Department of Chemical and

Biological Engineering, State University of New York at Buffalo, Buffalo, NY). Drs.

Yoshimi Takai (Department of Biochemistry and Molecular Biology, Kobe University

Graduate School of Medicine, Kobe, Japan) and Jun Miyoshi (Osaka Medical Center for

Cancer and Cardiovascular Disease, Osaka University, Osaka, Japan) generated and

Page 10: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

x

provided the Stxbp5 KO mice. Bone-marrow transplantation was performed under the

direction of Dr. Maria de la Luz Garcia-Hernandez. Dr. Kyung Ae Ko performed FeCl3-

induced carotid thrombosis. Kristina L. Modjeski conducted analysis of ex vivo platelet

studies.

Chapter 3: Clare Bao performed in vitro studies depicted in Figure 3-2 A-C, Figure 3-3

B, Figure 3-4 B, D-E, and Figure 3-5 right panel.

In addition, the in vivo mesenteric thrombosis model, platelet rolling, leukocyte rolling,

and microsphere rolling experiments were all performed by Sara Ture. Michael B.

LoMonaco provided substantial technical assistance and conducted some of the VWF

exocytosis assays.

This work was supported by NIH/NHLBI R21 HL108372, R01 HL074061, R01

HL78635 (to Charles J. Lowenstein), by the HHMI "Med-into-Grad" Fellowship in

Cardiovascular Sciences at the Aab Cardiovascular Research Institute, University of

Rochester (to Charles J. Lowenstein and Qiuyu Zhu), and by American Heart Association

grants 0835446N (to Munekazu Yamakuchi) and 13PRE17050105 (to Qiuyu Zhu).

Page 11: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

xi

Table of Contents

Biographical Sketch ............................................................................................................ ii

Acknowledgments.............................................................................................................. iv

Abstract ............................................................................................................................. vii

Contributors and Funding Sources..................................................................................... ix

Table of Contents ............................................................................................................... xi

List of Tables .................................................................................................................... xv

List of Figures .................................................................................................................. xvi

List of Schemes ................................................................................................................ xix

List of Abbreviations ........................................................................................................ xx

Introduction ....................................................................................................... 1

1.1 Venous thromboembolism: morbidity and mortality ............................................... 2

1.2 VTE pathophysiology: the good, the bad, and the ugly ........................................... 6

1.2.1 Endothelial cells: the good ............................................................................ 6

1.2.2 Platelets: the bad ........................................................................................... 8

1.2.3 Hypercoagulation, stasis, and beyond: the ugly.......................................... 11

1.3 Exocytosis and the SNARE hypothesis ................................................................. 15

1.3.2 Components of SNAREs ............................................................................ 17

Page 12: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

xii

1.3.3 SNARE regulators ...................................................................................... 23

1.4 Endothelial exocytosis ........................................................................................... 28

1.4.1 Endothelial granules.................................................................................... 28

1.4.2 Major compounds released by endothelial exocytosis................................ 31

1.4.3 Types of VWF release ................................................................................ 39

1.4.4 Regulators of endothelial exocytosis .......................................................... 42

1.5 Thesis overview ..................................................................................................... 44

Dual Role of STXBP5 in Endothelial Exocytosis, Thrombosis, and Platelet

Secretion ........................................................................................................................... 46

2.1 Introduction ............................................................................................................ 47

2.1.1 Novel genetic variants are associated with human VWF levels ................. 47

2.1.2 STXBP5: A novel regulator of exocytosis.................................................. 47

2.1.3 STXBP5 SNP and plasma VWF .................................................................. 49

2.2 Results .................................................................................................................... 54

2.2.1 STXBP5 is expressed in human ECs and murine tissues ........................... 54

2.2.2 STXBP5 inhibits endothelial exocytosis in vitro ........................................ 55

2.2.3 Genetic variation in STXBP5 affects VWF exocytosis in vitro ................. 56

2.2.4 STXBP5 does not co-localize with endothelial WPBs ............................... 57

2.2.5 STXBP5 interacts with endothelial exocytic machinery ............................ 57

Page 13: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

xiii

2.2.6 STXBP5 inhibits endothelial exocytosis in vivo ........................................ 60

2.2.7 STXBP5 affects hemostasis and thrombosis .............................................. 62

2.2.8 STXBP5 promotes platelet secretion and activation .................................. 63

2.3 Discussion .............................................................................................................. 96

VAMP8 Mediates Endothelial Exocytosis .................................................... 105

3.1 Introduction .......................................................................................................... 106

3.2 Results .................................................................................................................. 108

3.2.1 Endothelial cells express a distinct set of SNAREs that mediate granule

exocytosis ................................................................................................................ 108

3.2.2 VAMP8 and VAMP3 co-localize with VWF ........................................... 108

3.2.3 VAMP8 and VAMP3 interact with the SNAREs STX4 and SNAP23 .... 109

3.2.4 VAMP8 and VAMP3 mediates endothelial exocytosis in vitro ............... 110

3.2.5 VAMP8 mediates endothelial exocytosis in vivo ..................................... 111

3.3 Discussion ............................................................................................................ 126

SNAP23 Regulates Endothelial Exocytosis .................................................. 131

4.1 Introduction .......................................................................................................... 132

4.2 Results .................................................................................................................. 133

4.2.1 SNAP23 is expressed in human ECs and murine tissues ......................... 133

4.2.2 SNAP23 regulates endothelial exocytosis ................................................ 133

Page 14: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

xiv

4.2.3 SNAP23 is primarily localized on cell membrane in EC ......................... 134

4.2.4 SNAP23 interacts with endothelial exocytic machinery .......................... 135

4.3 Discussion ............................................................................................................ 145

Conclusions and Perspectives ....................................................................... 149

5.1 Summary .............................................................................................................. 150

5.2 Working model for endothelial exocytosis .......................................................... 152

5.3 Future directions .................................................................................................. 156

5.3.1 STXBP5 SNP and VWF ............................................................................ 156

5.3.2 STXBP5 in ECs and in platelets ............................................................... 158

5.4 Significance and perspectives .............................................................................. 161

Materials and Methods .................................................................................. 163

References ....................................................................................................................... 181

Page 15: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

xv

List of Tables

Table 1 Description of STXBP5 SNPs associated with plasma VWF levels. ................... 51

Table 2 List of antibodies ............................................................................................... 177

Table 3 Primers used for SYBR Green RT-qPCR .......................................................... 180

Page 16: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

xvi

List of Figures

Figure 1-1 Model of VWF-platelet interaction in vascular injury. ................................... 38

Figure 2-1 STXBP5 expression and transcript variants. ................................................... 65

Figure 2-2 STXBP5 inhibits endothelial exocytosis in vitro. ........................................... 67

Figure 2-3 STXBP5 and constitutive release of VWF...................................................... 69

Figure 2-4 STXBP5 does not affect HUVEC content of VWF. ....................................... 70

Figure 2-5 Knockdown of STXBP5 does not affect the number of WPB granules per cell.

........................................................................................................................................... 71

Figure 2-6 Knockdown of STXBP5 does not affect the size of WPB granules. .............. 72

Figure 2-7 Knockdown of STXBP5 does not affect the shape of WPB granules. ........... 73

Figure 2-8 STXBP5 inhibits HUVEC release of P-selectin. ............................................ 75

Figure 2-9 STXBP5 SNP and release of VWF. ................................................................ 76

Figure 2-10 Particles containing STXBP5 are not co-localized with endothelial granules

containing VWF. ............................................................................................................... 77

Page 17: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

xvii

Figure 2-11 STXBP5 co-sediments, co-localizes, and co-precipitates with STX4 and

SYT1. ................................................................................................................................ 78

Figure 2-12 STXBP5 does not co-localize with markers for ER, Golgi, lysosomes, or

endosomes. ........................................................................................................................ 82

Figure 2-13 STXBP5 does not interact with NSF and Munc family members. ............... 83

Figure 2-14 STXBP5 does not co-localize with SNAP23, VAMP8, or VAMP3. ........... 84

Figure 2-15 STXBP5 does not co-localize with Caveolin-1, Munc18-3, Munc18-2, or

NSF. .................................................................................................................................. 86

Figure 2-16 STXBP5 does not co-localize with Rab27, Myosin5a or MyRIP. ................ 87

Figure 2-17 Stxbp5 affects plasma VWF in mice. ............................................................ 88

Figure 2-18 Stxbp5 inhibits endothelial exocytosis in mice. ............................................ 89

Figure 2-19 Stxbp5 increases thrombosis in mice. ........................................................... 92

Figure 2-20 Stxbp5 regulates platelet secretion and activation. ....................................... 93

Figure 2-21 Stxbp5 in platelets increases thrombosis in mice. ......................................... 95

Figure 3-1 Expression of SNAREs in endothelial cells. ................................................. 113

Page 18: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

xviii

Figure 3-2 Subcellular localization of VWF and SNAREs in endothelial cells. ............ 115

Figure 3-3 Interaction of SNAREs in endothelial cells. ................................................. 116

Figure 3-4 VAMP8 and VAMP3 mediate endothelial exocytosis.................................. 118

Figure 3-5 Knockdown of VAMP3 or VAMP8 does not affect endothelial granule

number or VWF content. ................................................................................................ 120

Figure 3-6 Knockdown of VAMP3 or VAMP8 does not affect WPB morphology. ...... 122

Figure 3-7 Vamp8 mediates endothelial exocytosis and leukocyte rolling in vivo. ....... 124

Figure 4-1 SNAP23 is expressed in human endothelial cells and murine tissues. ......... 138

Figure 4-2 SNAP23 is important for endothelial exocytosis. ......................................... 140

Figure 4-3 Subcellular localization of SNAP23 in endothelial cells. ............................. 142

Figure 4-4 SNAP23 interacts with endothelial exocytic machinery. .............................. 144

Page 19: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

xix

List of Schemes

Scheme 1-1 A common pathophysiology of VTE and atherothrombosis. ......................... 5

Scheme 1-2 Virchow's triad. ............................................................................................. 14

Scheme 1-3 Topologic model of neuronal SNARE complex. .......................................... 20

Scheme 1-4 Transition states of SNARE complex and membrane fusion. ...................... 21

Scheme 2-1 Schemes of STXBP5 and Lgl family member structures. ............................ 52

Scheme 2-2 STXBP5 differentially influences exocytosis in platelets and endothelial

cells. ................................................................................................................................ 104

Scheme 5-1 Working model of endothelial exocytosis regulation. ................................ 155

Page 20: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

xx

List of Abbreviations

ADAMTS13 ADAM metallopeptidase with thrombospondin type 1 motif, 13

ADP adenosine diphosphate

ANOVA analysis of variance

APC activated protein C

AT antithrombin

ATP adenosine triphosphate

ATPase triphosphatase

cAMP 3'-5'-cyclic adenosine monophosphate

Cas9 CRISPR-associated 9

CHARGE Cohorts for Heart and Aging Research in Genomic Epidemiology

CRISPR clustered regularly interspaced short palindromic repeats

DVT deep vein thrombosis

EC endothelial cell

EGF epidermal growth factor

ENCODE the Encyclopedia of DNA Elements

ER endoplasmic reticulum

ET-1 endothelin-1

GABA gamma-aminobutyric acid

GMP-140 granule membrane protein 140

GP glycoprotein

GPCR G-protein coupled receptors

Page 21: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

xxi

GST glutathione S-transferase

GWAS genome-wide association studies

HAEC human aortic endothelial cell

HBMEC human brain microvascular endothelial cell

HCT hematocrit

HDMVEC human dermal microvascular endothelial cell

HUS hemolytic uremic syndrome

HUVEC human umbilical vein endothelial cell

ICAM1 intercellular adhesion molecule 1

IL-8 interleukin-8

LD linkage disequilibrium

LFA-1 lymphocyte function-associated antigen-1

Lgl lethal giant larvae

Mac-1 macrophage-1 antigen

MAF minor allele frequency

MFI median fluorescence intensity

MP micro particle

NEM N-ethylmaleimide

NETs neutrophil extracellular traps

NO nitric oxide

NSF N-ethylmaleimide-sensitive factor

PADGEM platelet activation-dependent granule to external membrane protein

PAF platelet activating factor

Page 22: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

xxii

PAI plasminogen activator inhibitor

PDGF platelet-derived growth factor

PE pulmonary embolism

PF4 platelet factor 4

PGI2 prostacyclin

PKA protein kinase A

PKC protein kinase C

proVWF VWF pro-polypeptide

PSGL-1 P-selectin glycoprotein ligand-1

PTS post-thrombotic syndrome

ROS reactive oxygen species

SDS sodium dodecyl sulfate

SM protein Sec1/Munc18 protein

SNAP (1) soluble NSF attachment protein

(2) synaptosomal-associated protein

SNAP23 synaptosomal-associated protein, 23-KD

SNAP25 synaptosomal-associated protein, 25-KD

SNARE soluble N-ethylmaleimide sensitive factor attachment protein receptor

SNP single nucleotide polymorphism

SNV single nucleotide variant

STX syntaxin

STXBP5 syntaxin-binding protein 5

SYT synaptotagmin

Page 23: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

xxiii

TALEN transcription activator-like effector nuclease

TF tissue factor (thromboplastin)

TFPI tissue factor pathway inhibitor

TGF-β1 transforming growth factor β1

t-PA tissue plasminogen activator

t-SNARE target membrane-SNARE

TTP thrombotic thrombocytopenic purpura

TXA2 thromboxane A2

VAMP vesicle-associated membrane protein

VCAM1 vascular cell adhesion molecule 1

VEGF vascular endothelial growth factor

VLD VAMP-like domain

VSMC vascular smooth muscle cell

v-SNARE vesicle-SNARE

VTE venous thromboembolism

VWD von Willebrand disease

VWF von Willebrand factor

WPB Weibel-Palade body

ZFN zinc finger nuclease

Page 24: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

1

Introduction

Page 25: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

2

1.1 Venous thromboembolism: morbidity and mortality

Venous thromboembolism (VTE) is a disease caused by the formation of thombi in deep

veins, including deep vein thrombosis (DVT) and pulmonary embolism (PE).

VTE is a major cause of morbidity (1, 2). The annual incidence of VTE is 0.1% - 0.27%,

affecting 300,000-600,000 individuals in the United States each year; 5% of the general

population will have at least one occurrence of VTE in their lifetime (3-5). Typical

symptoms of DVT include pain, swelling, and ulcer of extremities, which severely limit

patients’ physical activity. Severe forms of DVT, such as phlegmasia cerulea dolens,

result in massive thrombotic occlusion of deep veins, ischemia and gangrene, which can

be limb-threatening. PE is caused by the dislodgement of a venous clot that travels to

and clogs vasculature in the lung. It is estimated that 20% of patients with PE die on the

first day of occurrence (6). Debilitating long-term complications of VTE which harm

patients' quality of life include post-thrombotic syndrome (PTS) in 23 - 60% of patients

following DVT (7), and pulmonary hypertension in 1% - 4% of PE patients (8, 9).

The mortality from VTE is high. Before the implementation of anticoagulant therapy,

VTE was often fatal (up to 30% mortality in the first month) (10). VTE is the second

leading cause of death in cancer patients (11). VTE is the third leading cardiovascular

disease after acute coronary syndrome and stroke (12). Autopsy data shows PE is the

third most common cause of death in hospitalized patients in the United States (13), with

nearly one quarter of cases presenting as sudden death (5). However, because of the

Page 26: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

3

difficulty in prediction and detection, VTE is under diagnosed, and the true prevalence is

probably even higher.

Anticoagulant therapy has enormously reduced the morbidity and mortality from VTE.

However, current treatment for VTE remains inadequate. Anticoagulant therapy increases

the risk of bleeding which has now emerged as a cause of death in about 25% of VTE

patients (14, 15). Anticoagulant therapy is also associated with other side effects

including thrombocytopenia, osteoporosis (heparin), and skin necrosis (coumarin) (16).

Moreover, the long-term prognosis of VTE remains dismal despite continued

anticoagulant therapy, primarily due to the high recurrence rate both during and after

anticoagulation therapy (approximately 7% at 6 months, and up to 40% in the first 10

years) (17, 18), which often necessitates indefinite anticoagulant treatment.

Risk factors for VTE are divided into acquired and hereditary causes. Well-established

risk factors include aging, immobilization, pregnancy, cancer, surgery or trauma, obesity,

diabetes mellitus, hypertension, hyperlipidemia, cigarette smoking, oral contraceptives,

and previous VTE (5, 19). In addition, genetic predisposition has been linked to VTE

(20). VTE appears more frequently in black and white populations than in East Asians or

Hispanics. Inherited rick factors include factor V Leiden, prothrombin G20210A

mutation, protein C or S deficiency, activated protein C (APC) resistance, antithrombin

(AT) deficiency, plasma von Willebrand factor (VWF), and elevated levels coagulation

factors VIII, IX, or XI (5, 19, 21).

Page 27: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

4

Recently, a new paradigm has been proposed to consider VTE as part of a “pan-

cardiovascular syndrome” that comprised of coronary artery diseases, cerebral vascular

diseases, and peripheral vascular diseases (22). Studies show patients with VTE have

higher risk of arterial thrombosis than those without VTE (23-25). VTE shares a common

pathophysiology with atherothrombosis: both are predisposed by common risk factors,

and share mechanistic pathways in common such as endothelial injury, inflammation, and

hypercoagulability (Scheme 1-1).

Appreciation of VTE pathophysiology may eventually furnish new therapeutic targets.

Identifying the risk factors for VTE and unravelling the details of its molecular pathways

not only provides a mechanistic framework for the prevention and treatment VTE, but

also helps our understanding of the scourge of arterial thrombotic diseases, such as

coronary artery diseases and stroke, which are of growing worldwide importance.

Page 28: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

5

Scheme 1-1 A common pathophysiology of VTE and atherothrombosis.

Adapted from Piazza et al. Circulation. 2010;121(19):2146-50 (22).

Page 29: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

6

1.2 VTE pathophysiology: the good, the bad, and the ugly

The pathophysiology of VTE is a multifactorial process that includes interactions

between endothelial dysfunction and platelet activation and aggregation, which together

set up a cascade of pro-thrombotic events (1).

1.2.1 Endothelial cells: the good

Endothelial cells (ECs) line the inner surface of vessel wall, maintaining the integrity of

the vasculature. An adult contains about 1×1013 ECs forming a nearly 1-kilogram “organ”

(26). Under normal conditions, EC preserves vascular metabolic homeostasis and patency

through multiple mechanisms:

1) Barrier function. EC acts as an antithombotic blood-tissue barrier separating the

endothelial matrix from blood and prevents the activation of coagulation (1, 27).

Potent pro-thrombotic proteins such as tissue factor (TF; thromboplastin) and

collagen in the subendothelial matrix initiate physiologic hemostasis when the

endothelial barrier is breached (28, 29).

2) Synthesis and release of anti-thrombotic factors. EC inhibit thrombosis by

producing nitric oxide (NO) (30, 31), prostacyclin (PGI2) (32), CD39 (33), and other

anti-thrombotic factors (34-36). EC inhibits the generation and activation of thrombin

by expressing tissue factor pathway inhibitor (TFPI) (37, 38). When pathologic

insults injure the vessel wall, EC decreases production of anti-thrombotic factors and

Page 30: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

7

increases production and release of pro-thrombotic factors such as VWF,

thromboxane A2 (TXA2), and expands binding sites for coagulation factors which

facilitate thrombus formation (34). The loss of balance between anti-thrombotic

activity and pro-thrombotic activity characterizes EC dysfunction (36, 39), portending

an elevated risk of cardiovascular events such as acute coronary syndrome and

sudden death (40-42).

3) Regulation of hemodynamics. EC regulates vascular tone by balancing the

production of vasoconstrictors and vasodilators. EC produces vasoconstrictors such as

endothelin-1 (ET-1), the most potent known vasoconstrictor, and platelet activating

factor (PAF). EC also produces labile vasodilators such as NO (30, 43-46) and PGI2

(34, 47).

4) Fibrinolysis. EC promotes fibrinolysis and maintains blood fluidity by a surface-

connected fibrinolytic system. EC produces and secretes tissue plasminogen activator

(t-PA) (48) and its receptors (49), and plasminogen activator inhibitors (PAIs) (50).

5) Regulation of vascular inflammation. EC produce adhesion molecules and

cytokines that participate and regulate the inflammatory process (51, 52).

Cardiovascular risk factors and clinical syndromes including aging, obesity, smoking,

hypertension, and hypercholesterolemia, are partly related to endothelial dysfunction

(53). Conversely, interventions that reduce cardiovascular risk factors improve

endothelial function and restore vascular homeostasis. Therefore, endothelium has been

proposed as a “barometer” for cardiovascular risk (54). Pathological insults to the

Page 31: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

8

vasculature disrupt the anti-thrombotic function of EC and set up the stage for a sequence

of pro-thrombotic events discussed below.

1.2.2 Platelets: the bad

Platelets are the second most abundant cells in human blood (150 - 400 × 109/L) after red

blood cells. They are formed by an elaborate membranous system in their precursor

megakaryocytes (the demarcation membrane system), and shed as cytoplasmic fragments

from megakaryocytes. Therefore, platelets are anucleate and have long been considered

as fragments of cells. Advances in electron microscopy technology have revealed the

elaborate internal structure of platelets. Platelets are essential for physiological

hemostasis and thrombosis. In addition, they also mediate angiogenesis, wound repair,

cancer, and infection (55-58). Roles for platelets as important immune and inflammatory

cells have recently been recognized (59-64).

Circulating, quiescent platelets normally do not adhere to the vessel wall because of the

antithrombotic blood-tissue interface maintained by ECs. However, circulating platelets

rapidly respond to vascular damage (e.g. hemorrhage or ruptured arterial plaque) by

forming platelet plugs (thrombus). Defects of platelet number (thrombocytopenia) or

function can lead to a severe bleeding diathesis. The function of platelets in mediating

hemostasis and thrombosis depends on multiple properties of platelets:

Page 32: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

9

1) Platelet surface receptors. Platelet glycoprotein Ib (GPIb) constitutes the GPIb-V-

IX membrane receptor complex that binds to VWF, which mediates the first step in

platelet adhesion to injured vasculature. Platelet GPIb and GPVI binds to collagen in

the sub-endothelial matrix. Collagen binding to GPIb or GPVI activates a cascade of

signals inside a platelet that include elevations in intracellular Ca2+ levels, arachidonic

acid metabolism by cyclooxygenase isoforms, and release of TXA2 (65-67). Platelet

GPIIb/IIIa (also known as integrin αIIbβ3) is a platelet receptor for VWF, fibrinogen

and fibronectin (68-70). GPIIb/IIIa on the surface of a resting platelet does not

interact with ligands, but activation of the platelet leads to a conformational change in

GPIIb/IIIa enabling it to bind to VWF and fibronectin.

2) Platelet metabolites. Platelets contain enzymes that metabolize arachidonic acid into

vasoactive substances including TXA2 that promotes thrombosis.

3) Platelet granules. Platelets have three major types of granules containing over 300

vasoactive substances that modulate hemostasis, thrombosis, and vascular

inflammation (71, 72):

a. α granules, the most abundant granules, contain peptides and proteins,

including pro-coagulation proteins such as factor V, VWF, fibronectin and

fibrinogen, growth factors such as platelet-derived growth factor (PDGF),

epidermal growth factor (EGF), chemokines such as platelet factor 4 (PF4)

and transforming growth factor β1 (TGF-β1), and adhesion molecules such as

P-selectin. These proteins constitute the bulk of the platelet secretome.

Page 33: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

10

b. δ granules or dense granules contain small molecules such as polyphosphates,

serotonin, and calcium, which are important signaling molecule and potent

platelet-activators.

c. Lysosomes contain proteolytic enzymes such as β-hexosaminidase, cathepsin

D and E, and other enzymes. There are only a few lysosomes per platelet.

Lysosomes are considered important for clot- and wound-remodeling (73, 74).

How do platelets mediate thrombus formation? This function is carried out by a complex

process that transforms quiescent platelets into a platelet plug. This process is artificially

separated into three stages: adhesion, activation, and aggregation. First, injury to the

vessel wall exposes the sub-endothelial matrix. Injured endothelium release VWF;

platelet surface receptors recognize VWF and sub-endothelial collagen: the GPIb-V-IX

complex binds to VWF; and the GPVI receptor binds to collagen. Receptor binding not

only anchors platelets to the injury site, but also triggers a cascade of intraplatelet signals

that are essential to activation and aggregation (65-67). Second, platelet activation occurs

right after platelet receptor binding to VWF, collagen, or TF. Ligand interaction with

these platelet G-protein coupled receptors (GPCR) activates a cascade of intracellular

signals including an increase in intracellular Ca2+ which in turn leads to an array of

downstream effects: GPIIb/IIIa activation, granule secretion, and platelet spreading.

GPIIb/IIIa activation initiates outside-in and inside-out signaling and increases

production of pro-thrombotic factors such as TXA2; secretion of granule contents

enhances local platelet aggregation and attracts more circulating platelets to the injury

site; and morphological changes generate forces to plug and contract the wound. Finally,

Page 34: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

11

platelets aggregate with other platelets as a result of GPIIb/IIIa activation. Activated

platelets also interact with coagulation factors to propagate the coagulation cascade,

which generates thrombin and fibrin. These three stages facilitate and overlap with each

other. The result is hemostasis and thrombosis.

1.2.3 Hypercoagulation, stasis, and beyond: the ugly

Virchow identified a triad of factors that contribute to VTE: abnormalities in blood flow,

injury of the vessel wall, and blood hypercoagulability (Scheme 1-2) (75, 76). Many

thromboembolic diseases involve one or more factors of this triad. Regardless of the

dispute about its origin (77), Virchow's triad integrates the basic roles of three

coordinated components in thrombosis:

1) Endothelial injury lays the foundation for platelet adhesion and coagulation.

Endothelial cells can be activated by numerous stimuli into a pro-thrombotic and pro-

inflammatory phenotype. EC release a plethora of pro-thrombotic substances by

exocytosis to trigger platelet adhesion and activation. The exposed sub-endothelial

matrix anchors VWF and platelets, while exposed TF activates the coagulation

cascade. Activated ECs synthesize and release PAF in large quantities, which triggers

inflammation and thrombosis. P-selectin recruits leukocytes that participate in

inflammation by producing reactive oxygen species (ROS) and proteases, further

boosting thrombosis (78).

Page 35: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

12

2) Static flow induced by immobility, compression or heart failure increases the

thrombotic risk. Normal laminar flow maintains EC morphology and function and

diminish local VWF, thrombin and fibrin to prevent platelet adhesion and thrombosis.

Stasis increases the risk of local pro-thrombotic factor accumulation and fibrin

formation and polymerization (79).

3) Blood hypercoagulability can result from the antiphospholipid syndrome, cancer and

oral contraceptives. High levels of circulating pro-thrombotic factors, such as

circulating ECs, VWF, thrombomodulin, and D-dimers promote blood cell aggregates

(78). Increase of blood coaguability is also seen when there is a higher than normal

hematocrit (HCT) or erythrocytes (polycythemia vera) (80, 81), higher than normal

number of leukocytes (81), higher than normal number of platelets (thrombocytosis),

and their inappropriate activation (80, 82, 83), which all increase the risk of clotting.

Recently studies have shown thrombus formation can be triggered by discoid platelets

on undisrupted ECs indirectly through local production of ROS, adding yet another

mechanism by which elevated blood hypercoagulability (thrombocytosis) can

increase thrombosis risk (84).

However, Virchow’s triad is not the complete picture of blood clot formation. The

mechanisms responsible for VTE are manifold. In reality, in a great proportion of

patients, factors from Virchow’s concept are not detectable. Additional elements that are

important to thrombosis have been identified, including ABO blood groups, fibrinolysis,

neutrophil extracellular traps (NETs) (85), and infection (86) to name just a few. For

instance, procoagulant circulating micro particles (MPs), membrane fragments shed by

Page 36: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

13

blood cells and the vasculature, are newly regarded as a risk factor for thrombosis (87,

88). Activated platelets and ECs are regarded as the main sources of MPs. TF-positive

MPs may explain the increased rates of VTE in cancer patients (87).

Page 37: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

14

Scheme 1-2 Virchow's triad.

Page 38: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

15

1.3 Exocytosis and the SNARE hypothesis

Exocytosis is the first response shared by many cells in the thrombogenic pathway. EC

responds to injury by exocytosis. Platelet activation depends on granule secretion (74).

Activated leukocytes release pro-inflammatory enzymes by exocytosis (89). Enhanced

understanding of the mechanisms that regulate exocytosis is critical for improving the

prevention and treatment of thromboembolic diseases.

Exocytosis is the regulated release of granule contents by intracellular vesicle fusion with

the plasma membrane. Exocytosis is an intracellular protein transport mechanism that

exists in all eukaryotic cells, and is conserved from yeast to human (90, 91). Protein

trafficking maintains the internal structure of a cell, and allows it to communicate with

adjacent cells and its environment. Endothelial cells, leukocytes, and platelets all

participate in the pathology of VTE by exocytosis.

By 1970, the seminal work by George Palade (1974 Nobel laureate) and others had

already made it evident that secretory proteins are transported by a series of vesicles from

the endoplasmic reticulum (ER) to the Golgi apparatus to the cell surface where they are

released to the extracellular space (92). The fusion of vesicles with a target membrane

was obviously done with extremely high specificity, to ensure that the correct cargos is

always delivered to its correct destination. However, many questions remain. One such

question is: How does each type of vesicle fuse with the right target membrane at the

right time?

Page 39: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

16

Satisfactory answers to this question were not available until the last two decades. In the

1970s, the prevailing belief held that the anatomical arrangement of the endomembrane

systems dictated the trafficking specificity of cargos. One example is the close proximity

between the ER and the Golgi, where vesicles budded from ER appear to be force-fed

into the Golgi, but not into other intracellular organelles. Thus, it was the anatomical

proximity of membrane compartments that appeared vital to vesicle targeting specificity.

However, Rothman et al. proposed a simple hypothesis that specific membrane fusion

was governed by the intrinsic chemical specificity of membranes, not by the intricate

patterns of anatomy. Rothman and colleagues proved that membrane transport can be

reconstituted in a cell-free membrane-fusion assay (93); targeted, but not non-specific

membrane fusion, was faithfully replicated using a minimum set of proteins (94-96).

Using this assay, it was discovered that the membrane fusion machinery consists of three

components later to be called SNAREs (soluble N-ethylmaleimide-sensitive factor (NSF)

attachment protein receptors) proteins (97).

Based on this evidence, Rothman and others proposed the SNARE hypothesis to explain

vesicle fusion at the right place and the right time: SNARE proteins are vesicle and target

membrane markers; specific membrane fusion is mediated by SNARE proteins on vesicle

and target membranes; and distinct SNAREs on vesicles and distinct SNAREs on target

membranes determined the membrane fusion specificity (97). The SNARE hypothesis

was supported by many studies. One strong piece of evidence is that the neurotoxins

tetanus toxin or botulinus toxin block exocytosis by specifically degrading SNARE

proteins (98-100).

Page 40: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

17

The work by James E. Rothman, Randy W. Schekman and Thomas C. Südhof jointly

won them the 2013 Nobel Prize in Physiology or Medicine “for their discoveries of

machinery regulating vesicle traffic, a major transport system in our cells” (101). Their

work provided a unifying conceptual framework for vesicle transport and membrane

fusion that integrated many proteins into an evolutionarily-conserved molecular machine.

SNAREs has been confirmed as a universal machinery that mediates membrane fusion in

a variety of cell types (102-113).

1.3.2 Components of SNAREs

SNAREs are membrane proteins. The SNARE hypothesis proposes that SNARE

machinery consists of three components: a SNARE on the vesicle (v-SNARE) binds

specifically to two cognate SNAREs on the target membrane (t-SNAREs), forming a

SNARE complex. The specificity of a transport vesicle for its target membrane is

mediated by the specific interaction between one unique v-SNARE and two unique t-

SNAREs. Much of our understanding of the SNARE machinery comes from presynaptic

neurons (114, 115), the v-SNARE on presynaptic vesicles is vesicle-associated

membrane protein 2 (VAMP2), whereas the t-SNAREs on presynaptic membrane are

synaptosomal-associated protein, 25-KD (SNAP25), and syntaxin 1 (STX1).

The crystal structure of a SNARE complex reveals that each SNARE protein possess at

least one α-helix. The SNARE complex is formed by the parallel arrangement of these α-

helices into a highly twisted four-helix bundle (116) (Scheme 1-3). The center of this

Page 41: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

18

four-helix coiled-coil contains conserved leucine-zipper-like layers, with hydrophobic

residues buried inside. These hydrophobic residues consist of one arginine (R) and three

glutamine (Q) residues, contributed from each of the four helices. Therefore, SNARE

motifs used to be classified as R-SNAREs (from v-SNAREs) and Qa-, Qb-, Qc-SNAREs

(form t-SNAREs). These residues are highly conserved and sensitive to mutations (117-

120).

Vesicle fusion is mediated by the combined action of many four-helix SNARE

complexes (Scheme 1-4). Unfolded SNAREs can spontaneously assemble into four-helix

trans-SNARE complex (SNAREpin), a process called “priming”, which brings vesicles

into a “docked” position (121). The assembly is thermodynamically favorable, resistant

to sodium dodecyl sulfate (SDS) (122) and heat (123) treatment. Multiple SNAREpins

aggregate at the vesicle-target membrane interface, exerting an inward force that pulls

membrane into close proximity; yet spontaneous fusion is prevented by “clamping”

proteins such as complexin. Upon Ca2+ stimulation, the “clamps” are removed by specific

Ca2+ sensors. The energy from multiple SNAREpins eventually overwhelms the

resistance from the membranes and drives the membrane to fuse (124, 125). SNAREpins

drive fusion cooperatively; upon membrane fusion they transforms into a low-energy,

fully zippered cis-SNARE (126).

Disassembly of the SNARE complex is important for the vesicle transport cycle. Initial

experiments with cell-free membrane assays by Glick et al found that membrane

transport was blocked by N-ethylmaleimide (NEM), a sulfhydryl alkylating reagent

Page 42: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

19

(127). NEM treatment resulted in accumulation of fused vesicles, suggesting some

regulator sensitive to NEM is required to recycle the membrane fusion machinery (128).

This soluble, N-ethylmaleimide-sensitive protein was soon purified based on its ability to

restore membrane transport following NEM treatment and was named “N-

ethylmaleimide-sensitive factor” (NSF) (128). NSF is a well conserved cytosolic

triphosphatase (ATPase). NSF hydrolysis of adenosine triphosphate (ATP) is required for

SNARE disassembly and recycling. NSF binds to soluble NSF attachment protein

(SNAP, different from synapsomal-associated protein) which in turn interacts with the

SNARE complex (129). SNARE proteins were named because they interact with SNAP

(thus SNARE stands for soluble NSF attachment protein receptors). SNARE, SNAP, and

NSF form a complex on the cell membrane which sediments at 20S (97). Unfolding of

substrate SNARE proteins is coupled to NSF hydrolysis of ATP via a “spring-loaded”

mechanism (130-132).

Page 43: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

20

Scheme 1-3 Topologic model of neuronal SNARE complex.

Shown is the four-helix coiled-coil bundle contributed by v-SNARE VAMP2 (blue), and

t-SNAREs syntaxin 1 (red) and SNAP25 (green). Transmembrane domains of syntaxin 1

and SNAP25 are shown in yellow. SNAP25 has no transmembrane domain; instead,

SNAP25 is anchored to the target membrane by a loose unstructured loop connecting its

two helices.

Adapted from Sutton et al. Nature. 1998;395(6700):347-53(116).

Page 44: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

21

Scheme 1-4 Transition states of SNARE complex and membrane fusion.

(A) Tans-SNARE complex. One helix on the vesicle membrane bundles with three

cognate helices on the target membrane to form a fourth-helix trans-SNARE complex.

This assembled fourth-helix bundle “zippers” towards its transmembrane anchors, but is

not completely zippered, generating an inward force that brings the vesicle membrane

into close proximity to the target membrane.

(B) Cis-SNARE complex. Eventually the inward force pulled membranes to fuse, and the

SNAREs are in parallel alignment, a low-energy state called a cis-SNARE complex.

Page 45: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

22

(C) Membrane transition in SNARE-mediated fusion, including (a) separated

membranes, (b) fusion stalk formation, (c) membrane bilayer contact, and (d) fusion pore.

Adapted from Sudhof et al. Science. 2009;323(5913):474-7 (133) and Jahn et al. Annu

Rev Biochem. 1999;68(863-911) (134).

Page 46: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

23

1.3.3 SNARE regulators

The SNARE machinery ensures specificity of membrane transport: the correct cargo is

delivered to the correct location. Membrane fusion specificity is encoded in SNARE

proteins per se as an intrinsic physico-chemical property (135), which can be faithfully

reconstituted using isolated SNARE proteins and liposomes (136). However, it has been

well-documented that vesicles inside cells traffic and fuse in a highly efficient and

choreographed manner, with both temporal and spatial precision. How was it achieved?

Cell-free liposome fusion assay demonstrates SNARE proteins alone cannot achieve the

same degree of speed and temporal precision in membrane fusion (137, 138). Despite its

simplicity, SNARE machinery requires additional regulators, and its controlling

mechanism proved to be a rather complicated system and only began to be appreciated.

In neurons and other cell types, several SNARE regulatory proteins have been identified.

1.3.3.2 Synaptotagmin and complexin

The SNARE machinery relies on two families of proteins, synaptotagmin (SYT) and its

cofactor complexin, as “triggers” to achieve calcium-dependent time precision (139).

Synaptotagmins are a membrane protein family characterized by an N-terminal

transmembrane region, a variable central linker, and two C-terminal C2 domains (C2A

and C2B) which are autonomous Ca2+-binding modules (140). The best characterized

member, SYT1, is a synaptic vesicle Ca2+-binding protein. Ca2+ binding to C2 domains of

Page 47: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

24

SYT1 enhances its affinity to membrane phospholipids and SNAREs and triggers fast

vesicle release. SYT1 is required and only required for Ca2+-triggered fast

neurotransmitter release. Knock-out of SYT1 in mice impairs Ca2+-triggered neurovesicle

release. SYTs are believed to be universal Ca2+-sensors for triggered-exocytosis in many

different forms, and are evolutionarily conserved. Mammals express 16 synaptotagmins.

Different SYT homologs are expressed in different cell types, with different affinities for

Ca2+, which may explain the varied Ca2+-sensing kinetics in different vesicle transport

pathways (141-146).

However, SYTs do not act alone. Complexin is an essential co-factor of SYT1.

Complexin is a soluble protein originally discovered as a partner for trans-SNARE

complex and was subsequently named “complexin” (147). Similar to SNARE proteins,

complexin contains an α-helix that fits into the groove formed by the four-helix bundle in

the SNARE complex (148). Complexin is also evolutionarily conserved, but its role is

paradoxical. On the one hand, complexin inhibits SNARE-mediated fusion, as evidenced

in vitro and ex vivo (149-152). On the other hand, complexin knockout neurons also

exhibit similar phenotype as SYT1-null cells: loss of rapid and synchronous exocytosis,

suggesting it is required for Ca2+-triggered release (153). In addition, complexin null

mice exhibit motor and cognitive function deficits similar to those seen in Huntington's

disease, suggesting it is required for normal neurological function (154).

SYT and complexin act in concert to trigger Ca2+-induced synaptic exocytosis (139) and

both are essential for this process. But the detailed mechanism has not been completely

Page 48: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

25

elucidated. The current hypothesis is built on the crystal structure of the complexin-

SNARE complex (148). The major part of complexin is an α-helix, which competes with

synaptotagmin for binding to the same site at assembled SNARE complexes: a unique

groove in the four-helix bundle formed by syntaxin and VAMP. Complexin “clamping”

of the SNARE complex reinforced vesicle docking and poised them for simultaneous,

Ca2+-triggered synchronous release. Ca2+ binding to synaptotagmin displaces the

clamping of complexin and frees SNARE complex, allowing assembled SNARE

complex to zipper freely. Thus complexin clamps the SNARE complex before

membrane fusion occurs, and synaptotagmin releases the complexin clamp and permits

membrane fusion to occur. This model, however, does not explain why complexin is

counter-intuitively required for Ca2+-induced exocytosis and how knockout of complexin

abolishes Ca2+-triggered synchronous release (139). Novel mechanisms have been

proposed to complement this evolving model (155).

1.3.3.3 Munc18 and Munc13

High efficiency of SNARE-mediated fusion in cells is likely conferred by Munc18 and

Munc13 proteins that prepare the core SNARE engine for assembly.

Munc18 is a member of the Sec1/Munc18 (SM) proteins that are conserved from yeast to

mammals. Unc18 (C. elegans) and Sec1 (yeast) are absolutely essential for C. elegans

movements and yeast secretion (156). Similarly, Munc18 is essential for synaptic vesicle

release in mammals. The first-reported and best characterized mammalian Munc18

Page 49: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

26

member, Munc18-1 (also called STXBP1, Munc18a or neuronal Sec1) is a neural-

specific protein. Munc18-1 deficiency in cells abolishes synaptic membrane fusion (157).

Knockout of Munc18-1 alone in mice results in complete loss of neurotransmitter

secretion, rendering the brain “synaptically silent” (158), a phenotype that even exceeds

the knockout of a SNARE protein (159); in humans, Munc18-1 mutations cause early

infantile epileptic encephalopathy (160) called “STXBP1 encephalopathy” (161-164).

These results suggest Munc18-1 is not a by-stander but an essential component and

central actor in the membrane fusion machinery. Expression of Munc18 homologs has

been detected in various tissues, supporting it as a common component of vesicle fusion

machinary in many cell types (165, 166).

Exactly how Munc18-1 participates in membrane fusion is still unclear (133). Munc18-1

may promotes vesicle fusion by cooperating with SNAREs. In a reconstituted lipid

bilayer system, Munc18-1 strongly accelerated the fusion reaction through direct contact

with both v- and t-SNAREs (167). Crystal structure reveals Munc18-1 forms a complex

with STX1A in neurons (168). Munc18-1 guides syntaxin as a cheparone towards

productive SNARE complex formation, thus facilitates vesicle docking (157, 169).

Munc18-1 may also facilitate the topological arrangements of SNAREpins, clasp

SNAREpins circumferentially at the fusion interface, prevent their diffusion. These

functions collectively enhance the efficiency of SNARE-mediated fusion (133, 170).

Munc13 is a presynaptic member of the CATCHR protein family (members involved in

vesicle tethering) and is homologous to Unc13 of C. elegans. Munc13 is required for

Page 50: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

27

certain types of synaptic transmission (171, 172). Glutamatergic neurons from Munc13-1

null mice show arrest of synaptic-vesicle cycles and loss of transmitter release in

response to stimulation (172). Munc13 function together with Munc18-1 to cooperate the

assembly of SNARE complexes (173, 174). Munc13 may release syntaxin from the clasp

of Munc18-1, accelerating its transition from the closed syntaxin-Munc18 complex to the

SNARE complex (173). Munc13 lowers the energy barrier for synaptic vesicle fusion

(175). Moreover, NSF disassembly of the SNARE complex requires Munc18-1 and

Munc13 (174). Interestingly, unlike its absolutely-required participation in glutamatergic

synaptic secretion (171), Munc13 is not essential for dense-core vesicle release, but only

controls dense-core vesicle localization and release efficiency (176). Similarly, synaptic

release from some GABA (gamma-aminobutyric acid)-releasing neurons are not affected

by depletion of Munc13-1 (172). Therefore, Munc13 orchestrates membrane fusion with

the SNAREs complex in a transmitter- or cell-specific manner.

Page 51: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

28

1.4 Endothelial exocytosis

EC maintain the integrity of the vasculature. Due to its privileged location at the interface

between blood and organs, EC constantly senses changes in the blood and in the

subendothelial matrix, and actively modulates its interaction with other cells (including

blood cells, vascular smooth muscle cells and pericytes) or the blood flow. In response to

injury, EC undergo exocytosis, releasing numerous hemostatic and inflammatory

mediators into the blood steam that mediate interaction with platelets and leukocytes

(177-179).

1.4.1 Endothelial granules

In 1964, Ewald Weibel and George Palade discovered a remarkable cytoplasmic

component in rat and human arterial endothelia by electron microscopy (180). They

described the prominent morphologic features of this new organelle:

“A hitherto unknown rod-shaped cytoplasmic component which consists of a bundle of

fine tubules, enveloped by a tightly fitted membrane, was regularly found in endothelial

cells of small arteries in various organs in rat and man. It is about 0. 1 μ thick, measures

up to 3 μ in length, and contains several small tubules, ~ 150 A thick, embedded in a

dense matrix, and disposed parallel to the long axis of the rod.” (Weibel ER, and Palade

GE. J Cell Biol. 1964;23(101-112))

Page 52: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

29

Despite the discrepancy in their distribution, these organelles were ubiquitously found in

the endothelia:

“… cytoplasmic components were found with great regularity in endothelial cells of

small branches of the pulmonary artery in rats. Much less frequently, rod-shaped bodies

were also found in the endothelium of alveolar capillaries.” (Weibel ER, and Palade GE.

J Cell Biol. 1964;23(101-112))

As Weibel and Palade concluded in their report in 1964, “the nature and significance of

these cytoplasmic components are yet unknown” (180). But they did notice that “the rod-

shaped body described in this article has been observed with regularity in numerous

vascular endothelia of the rat, man, and Amblystoma. This indicates that it must be a

structure of some functional significance which for the moment remains obscure” (180).

Thanks to the EC isolation and culture technique developed in the 1970s (181, 182), these

rod-shaped structure with numerous internal bundles were further studied in cultured ECs

(183). These granules, now termed Weibel-Palade bodies (WPBs), turned out to be a

specific marker of EC. In 1982, Wagner et al. found WPBs are storage organelles for

VWF (184). In 1989, P-selectin that mediates leukocytes rolling was found on the

membrane of WPBs (185, 186). With these and other components in WPB discovered,

one major structural basis for endothelium interactions with blood cells was gradually

delineated. WPBs essentially equip EC with a rapid means of response to stimulation and

vascular injury. A variety of stimuli, including hypoxia, physical trauma, or inflammatory

mediators, trigger EC to release the contents of WPBs (178).

Page 53: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

30

WPBs are not the only type of endothelial granules that undergo regulated release. A few

other compounds have been reported to form specialized granules distinct from WPBs in

ECs, and undergo regulated release in response to stimuli. These compounds include

protein S, multimerin, t-PA (outside of WPBs), TFPI, and certain types of cytokines,

including CCL2 and CXCL1 (187-190). Some of them are important mediators of

thrombosis and inflammation, but the processing and release of these granules are poorly

understood. Perhaps these distinct populations of granules allow the same cell to respond

to diverse stimuli in a more controlled manner (190).

Page 54: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

31

1.4.2 Major compounds released by endothelial exocytosis

Today, a growing list of components have been found to be present in WPBs,

participating in a broad range of biological processes including hemostasis, thrombosis,

inflammation, vasoconstriction and angiogenesis (191).

Perhaps the potent vasoactive properties of WPB contents determines that they must be

contained in such granules, and only released in a highly-regulated manner: exocytosis.

The compounds stored in WPBs make the granule “the perfect first aid kit after an insult

to the vasculature” (192).

1.4.2.1 Von Willebrand factor

VWF is synthesized exclusively by EC and by megakaryocytes (193, 194). Plasma VWF

is predominantly contributed by ECs (195).

VWF is the major and defining constituent of WPBs (184, 196). Expression of VWF

propolypeptide in a variety of nonendothelial cells results in rod-shaped granules that

closely resemble WPBs, suggesting VWF itself drives the formation of WPB structure

(197). The internal bundles in WPBs described by Weibel and Palade in 1964 were in

fact densely-packed VWF multimers (184). Once released into the blood stream by

exocytosis, these densely-packed VWF multimers are unfurled by local shear forces,

exposing its A1 domain which binds to platelet GPIb receptor, A3 domain which anchors

to collagen in the subendothelial matrix, and C1 domain which binds to activated platelet

Page 55: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

32

GPIIbIIa. The unfurled VWF multimer can be cleaved by the protease called

ADAMTS13 (ADAM metallopeptidase with thrombospondin type 1 motif, 13) on its A2

domain (198), rendering smaller, non-thrombogenic VWF fragments that were

commonly found in the plasma (199, 200). The biosynthesis, secretion, and clearance of

VWF have been review recently in elegant detail (201).

The functional importance of VWF is demonstrated by the bleeding diathesis caused by

the quantitative or qualitative deficiency of VWF: von Willebrand disease (VWD). VWD

was first reported in 1926 by an internist from Finland, Erik von Willebrand (202) in

several members of a family. The proband, a 13-year-old girl, bled to death at her fourth

menstruation. Erik von Willebrand traveled to the girl’s hometown to study the disease in

depth. He mapped the family pedigree and found that 23 of the 66 family members had

bleeding problems. The pedigree and symptoms were both distinct from typical

hemophilia. Erik von Willebrand called the disease “hereditary pseudo hemophilia”. The

molecular basis of VWD was discovered by Southern blot in 1987 (203). This plasma

glycoprotein involved in the disease was then named for Dr. von Willebrand, together

with the bleeding disorder itself. VWD was found to be the most common inherited

bleeding disorder, with a prevalence of up to 1% (204).

VWF has two vital functions: it mediates platelet adhesion to the vessel wall during

primary hemostasis, and is the chaperone of factor VIII (205). Therefore, VWF is

essential for hemostasis and thrombosis (Figure 1-1). Recently, VWF was found to

participate in the formation of NETs (85). VWF binds to histone in vitro (206), and VWF

Page 56: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

33

is associated with NETs in vivo (207, 208). These studies suggest VWF may have yet

undiscovered roles in immune response and thrombogenesis (86).

Plasma VWF levels are affected by both non-genetic and genetic factors. Non-genetic

determinants of plasma VWF include well-established risk factors for endothelial

dysfunction such as smoking and diabetes, as well as other less studied factors such as

aging (209, 210), exercise (211) and alcohol (212). Plasma VWF level is mainly

genetically determined. VWF levels vary with race and sex (213, 214), and its heritability

estimates from 0.31 to 0.75 (215, 216). Genetic defects are the major cause of

quantitative or qualitative VWF abnormalities in VWD individuals. While in severe

VWF patients, VWF gene mutations are common (217, 218), in milder cases the genetic

determinants are more variable and complex. Moreover, plasma VWF levels are highly

variable even in VWD individuals with identical VWF gene mutations (217), and their

clinical manifestation can be quite heterogeneous (217-219). Studies have shown that

plasma VWF level is subject to incomplete penetrance and variations in other genes. For

instance, plasma VWF level is influenced by the blood group gene ABO, with 25% lower

VWF level in O group than in non-O groups (220). The protein carring A, B and H blood

group antigens is also found on VWF A2 domain, close to the ADAMTS13 cleavage site.

The modification by ABO blood group antigen on VWF molecule directly influences its

cleavage by ADAMTS13, resulting in varied plasma VWF levels in different blood

groups (221, 222). Polymorphisms in CLEC4M gene affect the clearance of VWF, and

contribute to the variation in plasma VWF levels (223).

Page 57: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

34

Plasma VWF levels are strongly correlated with thrombotic risk, especially VTE. For

patients with unusually large VWF multimers, as seen in hemolytic uremic syndrome

(HUS) and thrombotic thrombocytopenic purpura (TTP) resulted from ADAMTS13

mutation, there is an increased risk of platelet thrombi formation in small vessels where

shear rates is high (224-227). Increased VWF levels are strongly associated with risk for

coronary heart disease especially acute coronary syndrome (228-230). Clinical studies

show plasma level of VWF is significantly higher in VTE patients (231), and higher

VWF level independently increases VTE risk by up to 27% (232). Plasma VWF is also

frequently used as an indicator of endothelial dysfunction (233-235). Although VWF is

regarded as a novel therapeutic target for thromboembolic diseases (236, 237), regulation

of its release is still not well understood.

1.4.2.2 P-selectin

In 1824, Dutrochet first reported the adherence of white blood cells to the vessel wall and

their emigration to tissues (238). Classic works by Wagner, Cohnheim, Metchnikoff,

Addison, and the Clarks (75, 239-241) described the process in more details. In the early

20th century, Fahraeus (242) and Mogilnicki (243) provided quantitative account of

leukocyte rolling on the vessels. However, the mechanism and its significance were

largely unknown.

In 1972, Atherton and Born developed the mouse mesentery model (recapitulated and

refined in this thesis) to quantitatively investigate the adhesion of circulating leucocytes

Page 58: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

35

to blood vessel walls in response to a variety of stimuli, and noticed that rolling

leucocytes were increased after the application of E. coli culture filtrate, suggesting the

phenomenon may implicate a defensive mechanism against infection (240). Atherton and

Born hypothesized that some “adhesive substance” existed on the interface between

leukocytes and the vessel wall.

It was not until 25 years later that Bonfanti and McEver found that P-selectin is a

component of endothelial WPBs, and Bevilacqua et al. found that P-selectin released

from WPBs was the “adhesive substance” that recruited leukocytes (244). In mice that

have defective WPB formation (VWF knockout), the recruitment of leukocytes onto the

blood vessel is essentially abolished, resulting in inflammatory response defects similar

to P-selectin knockout mice (245). P-selectin-mediated leukocyte rolling on EC is now

regarded as a hallmark of inflammation, the self-defense mechanism of the vessels

against harmful stimuli such as infection.

P-selectin (also called CD62P, platelet activation-dependent granule to external

membrane protein (PADGEM), or granule membrane protein 140 (GMP-140)) is a

selectin family membrane protein expressed in ECs (exclusively in WPBs) and in platelet

α granules. P-selectin rapidly externalizes onto the EC surface upon stimulation.

Leukocyte rolling along the vessel lumen is mediated by the interaction between P-

selectin and its ligand PSGL-1 (P-selectin glycoprotein ligand-1) expressed on most

leukocytes. P-selectin display on the EC surface is often transient (< 2min) and is readily

recycled into the cytoplasm and back to the WPBs (246). Therefore, the initial “rolling”

Page 59: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

36

of leukocytes on P-selectin is labile and often transient. Firm adhesion and transmigration

of leukocytes involve additional adhesion molecules on ECs (e.g., intercellular adhesion

molecule 1(ICAM1) and vascular cell adhesion molecule 1 (VCAM1)), and β2 integrins

on leukocytes such as CD11a/CD18 (lymphocyte function-associated antigen-1, LFA-1)

and CD11b/CD18 (macrophage-1 antigen, Mac-1) (75).

1.4.2.3 Interleukin-8 (IL-8)

Activated ECs release IL-8, a chemotactic cytokine that stimulates neutrophil

phagocytosis and migration towards inflammatory sites (247). Expression and release of

IL-8 is contingent upon stimulation. Unstimulated EC does not store IL-8 in WPB,

whereas stimulation with IL-1β turns on its production and release from WPBs (247).

Intriguingly, IL-8 is present in the WPBs even after stimulation is removed, suggesting

storage of IL-8 in WPB may serve as the endothelial “memory” of inflammation,

enabling EC to rapidly respond to the next insult without de novo protein synthesis (248).

1.4.2.4 Other constituents in WPBs

Other important constituents in WPBs include tPA, a major initiator of fibrinolysis (249);

ET-1, a vasoconstrictor enabling EC to regulate hemodynamics (250-252); CD63 which

mediates cell-cell interaction (253); and angiopoietin-2, which sensitize ECs to

inflammation and induces angiogenesis (254, 255). Endothelial granules also contain

other pro-inflammatory and pro-thrombotic mediators that activate inflammation and

thrombosis in response to vascular injury (178, 191, 256). Together, release of these

Page 60: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

37

compounds trigger a cascade of events that modulates hemostasis, thrombosis,

inflammation, hemodynamics, and angiogenesis (178, 257, 258) . Endothelial exocytosis

is thus a novel therapeutic target for thrombotic and inflammatory diseases (229, 230,

259).

Page 61: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

38

Figure 1-1 Model of VWF-platelet interaction in vascular injury.

Adapted from Mannucci PM. New Engl J Med. 2004;351(7):683-94.(260)

Page 62: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

39

1.4.3 Types of VWF release

Current studies suggest the major components of WPBs, VWF, is released from the ECs

via one of the following three pathways: unstimulated, WPB-dependent “basal” release;

unstimulated, WPB-independent “constitutive” release; or stimulated WPB exocytosis.

Co-existence of multiply secretion pathways in the same cell for the same cargo is not

uncommon. For instance, in neurons, in addition to action potential-evoked exocytosis,

two additional forms of neurotransmitter release exist: asynchronous release and

spontaneous “mini” release (261). Diversified vesicle release pathways enable cells to

more flexibly adjust to the changing environment with precise and optimized response.

In the 1990s, only two types of endothelial VWF secretion was proposed: constitutive

and stimulated secretion (262, 263). It was initially estimated that over 90% of VWF in

cultured EC would be secreted constitutively, with less than 10% secreted by the

regulated exocytosis (264). However, later studies revealed that high molecular weight

VWF (the majority of endothelial VWF content) is released almost exclusively by the

regulated pathway, and constitutive secretion of VWF (mostly immature VWF

fragments) was negligible (265). More recent studies suggest that in the absence of

stimuli, the majority of resting VWF release (approximate 80%) is from WPBs (termed

“basal secretion” to distinguish from “constitutive section”) (266); WPB-independent

“constitutive” release is only significant when the biogenesis machinery of WPB is

compromised, for example, by NH4Cl treatment or AP-1/clathrin coat depletion (266,

267). G protein-dependent signaling may differentially regulate basal and evoked VWF

Page 63: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

40

secretion (268). Other mechanisms that regulate these different secretion pathways

remains to be discovered.

1.4.3.2 Basal release

The majority (80%) of unstimulated VWF release is though this route (266).

Study by Giblin et al. demonstrates in cultured ECs, most VWF is not sorted into the

constitutive secretory pathway, but into a post-Golgi compartment; part of it can be

subsequently secreted without stimulation. Since the only known post-Golgi secretory

organelle that contains VWF is WPBs, it was postulated that the majority of sorted VWF

ends in WPBs; WPBs then either spontaneously release their cargo, or undergo regulated

exocytosis in the presence of secretagogues (266). The mechanism that determines the

latter two different fates of WPBs is unknown. However, quantitative analysis suggests

although basal release accounts for 80% of unstimulated VWF release, the majority of

intracellular VWF content (high molecular multimers) is released by stimulated

exocytosis that far exceeds the amount by unstimulated VWF release (265).

WPBs that undergo basal secretion might differ from those secreted by exocytosis. These

WPBs are released shortly after they are formed, which are immature in their membrane

and VWF content (269, 270), whereas WPBs in exocytosis release almost all mature,

large multimers. It is postulated that the post-Golgi secretory organelles in basal secretion

are an immature form of WPBs (266).

Page 64: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

41

1.4.3.3 Constitutive release

The hallmark of constitutive secretion is the lack of post-Golgi materials (263).

Consistent with the prediction, VWF released from this pathway is VWF pro-polypeptide

(proVWF), an unprocessed precursor of mature VWF. This pathway accounts for only an

insignificant amount of unstimulated VWF release (265), but becomes significant when

the machinery of basal release pathway or WPB biogenesis is impaired (266, 267).

1.4.3.4 Regulated exocytosis

Endothelial exocytosis can be triggered by a variety of stimulations, including physical

damage (hypoxia, radiation, stretch, temperature, and trauma), chemical stimulation

(ATP, adenosine diphosphate (ADP), epinephrine, histamine, serotonin, and

leukotrienes), peptides (thrombin, vascular endothelial growth factor (VEGF),

complements), and lipids (oxidized low-density lipoprotein, sphingosine-1 phosphate,

and ceramide) (178, 271, 272).

These agonists induce WPB exocytosis by increasing the intracellular concentration of

two different second messengers: Ca2+ or 3'-5'-cyclic adenosine monophosphate (cAMP)

(271). Most stimuli induce exocytosis through raising intracellular Ca2+ (including

thrombin and histamine). Calmodulin mediates this signaling pathway (273, 274). A few

secretagogues, such as serotonin, epinephrine and vasopressin, trigger endothelial

exocytosis by increasing cAMP, with protein kinase A (PKA) as the effector (275, 276).

Purine nucleotides stimulation of EC involves both Ca2+ and cAMP (277). It is

Page 65: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

42

noteworthy that agonists effecting through these two different signaling pathway also

induce distinct changes on cytoskeleton remodeling and EC barrier function (278). Ca2+-

raising agonists diminish endothelial cell barrier function (279, 280), whereas cAMP-

raising agonists promote barrier function of EC (281-283).

1.4.4 Regulators of endothelial exocytosis

Much understanding of the SNARE mechanism in controlling exocytosis comes from

studies of yeast and neurons (131, 284-288). The exocytic machinery that drives vesicle

trafficking and membrane fusion in EC is similar to that found in neurons and yeast (90,

114, 134, 289, 290). V-SNARE molecules on endothelial granule surface interact with

specific t-SNAREs on the plasma membrane surface, forming a SNARE complex that

bridges the two membranes and directs vesicle fusion with the plasma membrane (291,

292).

Recent studies have defined some of the endothelial proteins that control the vesicle

secretory pathway in EC. Members of the SNARE family were found in human ECs,

including STX, VAMP, and SNAP homologs (102, 108, 109), although the exact identity

and function of endothelial SNAREs remains unclear. One exception is perhaps STX4,

which is required for endothelial exocytosis (108, 109).Various small GTPases and their

effectors such as Rab 3A, Rab3B, Rab3D, Rab11, Rab15, Rab27a, Rab27b, Rab 33a,

Rab37, RalA, and Rap1 play an important role in WPB maturation and exocytosis (102,

108, 178, 257, 293-305). MyRIP (Slac2c) serves as a bridge between Rab27a and myosin

Page 66: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

43

Va/MyosinVIIa to control the location of endothelial granules (269, 296, 298, 305, 306).

Slp4a (granuphilin) binds to Rab8, Rab27a, Rab3A-D, and syntaxins, and promotes VWF

release (177, 296, 307). Munc13-4 was found in human EC and co-localizes with WPBs,

presumably acting as an effector of Rab27A (305). NO and thioredoxin regulate

exocytosis by chemically modifying NSF (108, 308). An assortment of secretagogues or

pathologic stimuli can stimulate granule secretion from human ECs through discrete

signaling pathways (190, 309-315). Members of the synaptotagmin family function as a

conserved sensor for Ca2+ and triggers regulated exocytosis in most cell types (139).

Unidentified factors relating to autophagy also affect exocytosis (316).

However, the detailed mechanisms controlling the endothelial SNARE machinery remain

partially unknown. Specifically, critical SNARE regulators in endothelial cell remains to

be identified, and the exact mechanism of how these regulators are coupled to the

SNARE machinery is to be characterized.

Page 67: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

44

1.5 Thesis overview

Understanding regulation of exocytosis in ECs will address critical questions in the

pathogenesis of VTE. To elucidate the molecular mechanism of endothelial exocytosis,

the primary question is to identify and characterize the endothelial SNARE protein

members and their regulators. The thesis will approach this important question from the

following perspectives:

First, high-throughput platforms such as GWAS have discovered novel potential genetic

“hot spots” for VTE. These potential genetic determinants remain to be validated and

characterized, among which important regulatory molecules of endothelial exocytosis,

such as SNARE regulatory partners, may be identified and characterized.

Second, few endothelial SNARE candidates have been investigated in detail to date. The

identities of key endothelial SNAREs, especially VAMP homologs and SNAP homologs,

are unclear.

Therefore, we hypothesized that specific members of the SNARE superfamily and

their regulatory partners mediate endothelial exocytosis and thrombosis.

Chapter 2 harvests GWAS data to identify a novel protein STXBP5 that is associated

with human plasma VWF levels, and explores its role as a regulator of endothelial and

platelet exocytosis, as well as hemostasis and thrombosis.

Page 68: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

45

Chapter 3 identifies the v-SNARE that mediates WPB release, and finds that VAMP8 is

an important endothelial v-SNARE.

Chapter 4 identifies the endothelial SNAP isoform that mediates WPB release: SNAP23.

The work of this thesis not only defined the role of the SNARE components and one

important SNARE regulator in endothelial exocytosis, but also expanded the potential

pathways to regulate endothelial function, hemostasis and thrombosis that will provide

novel perspectives for the prevention and treatment of VTE.

Page 69: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

46

Dual Role of STXBP5 in Endothelial

Exocytosis, Thrombosis, and Platelet Secretion

Page 70: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

47

2.1 Introduction

2.1.1 Novel genetic variants are associated with human VWF levels

The pathophysiology of VTE is complex, and much remains to be discovered about

cellular pathways that contribute to thrombosis. In Chapter 2, we used genetic

approaches to discover novel pathways that regulate thrombosis.

Recently, GWAS by the Cohorts for Heart and Aging Research in Genomic

Epidemiology (CHARGE) Consortium identified novel genetic variants that are

associated with altered plasma VWF levels in humans (317-319). These genetic loci

might indicate novel genes whose products regulate endothelial exocytosis. One genetic

locus with the highest significance is located within the gene that encodes syntaxin-

binding protein 5 (STXBP5). This association was strikingly reaffirmed by studies on

venous thrombosis in humans (320). The prominent relationship between human plasma

VWF alterations and genetic variation of STXBP5 prompted us to investigate the

potential role of STXBP5 in endothelial exocytosis.

2.1.2 STXBP5: A novel regulator of exocytosis

STXBP5 was first discovered by Fujita et al. as a protein interacting with syntaxin1A in

neurons and was named tomosyn (“tomo” means friend in Japanese; tomosyn means

“friend” of “syn”taxin) (321). It is enriched in rat brain and is also found in the heart,

Page 71: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

48

spleen, lung, liver, skeletal muscle, kidney, and testis (321, 322). The gene encoding

STXBP5 (originally called tomosyn and later called tomosyn-1) is located on mouse

chromosome 10 and human chromosome 6. Murine Stxbp5 gene has at least 3 transcript

variants: Stxbp5-b, Stxbp5-m, and Stxbp5-s. Human STXBP5 gene has at least 5 predicted

transcript variants: STXBP5-1, STXBP5-2, STXBP5-X1, STXBP5-X2, and STXBP5–X3.

The gene Stxbp5L (originally called tomosyn-2) is located on mouse chromosome 16 and

human chromosome 3. Murine Stxbp5L has at least 4 transcript variants: Stxbp5L-xb,

Stxbp5L-b, Stxbp5L-m and Stxbp5L-s – all with distinct distribution patterns (322, 323).

STXBP5 belongs to the Lgl (lethal giant larvae) family which is conserved from yeast to

human. Lgl family members include Lgl, Sro7, Sro77, and STXBP5. Their common

feature are N-terminal WD40 repeats composed of β-propeller structures, which often

serve as scaffolds for protein-complex assembly (324)(Scheme 2-1). Lgl family members

have been involved in vesicle trafficking. Lgl, Sro7, and STXBP5 directly interact with

SNARE protein (321, 325-327).

All STXBP5 isoforms possess an N-terminal domain that contains WD40 repeats, a

variable tail domain, and a C-terminal domain that includes an R-SNARE-like motif

homologous to VAMP (VAMP-like domain, VLD) (322, 323, 328) (Scheme 2-1). In

neurons, VLD mediates the interaction of STXBP5 with syntaxin1A as a competitor to

VAMP2, and blocks formation of the heterotrimeric SNARE complex composed of

syntaxin1A, VAMP2, and SNAP25 (329, 330). STXBP5 inhibits neuron release of

neurotransmitters and endocrine cell secretion of insulin or other vesicles (331-337).

Page 72: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

49

However, the role of STXBP5 in the vasculature has never been studied, and the relation

of STXBP5 with thrombosis has not been explored.

2.1.3 STXBP5 SNP and plasma VWF

The CHARGE Consortium identified genetic variations within the STXBP5 locus that

associate with plasma VWF, factor VII, and factor VIII levels (Table 1). The single

nucleotide polymorphism (SNP) with the highest genome-wide significance level (P =

6.9 × 10−22) for VWF plasma levels in the meta-analysis is rs9390459 (hg19

chr6:g.147680359G>A), a synonymous variation in the coding region for gene STXBP5

(317). Therefore, we attempted to prioritize nonsynonymous STXBP5 variants in high

linkage disequilibrium (LD) with this candidate SNP. The top non-synonymous variation,

rs1039084 (hg19 chr6:g.147635413A>G), encodes STXBP5 asparagine (N) to serine (S)

substitution at the 436 residue (hereinafter STXBP5-N436S), and is in high LD with

rs9390459 (R2 = 0.87, D’ = 0.97 from International HapMap Project; R2 = 0.93, D’ =

0.97 from 1000 Genomes Project, data from: http://www.broadinstitute.org/mpg/snap/)

(338). This N436S substitution encoded by rs1039084 is in one of the WD40 repeat

domains of STXBP5. The minor allele of rs1039084 is associated with decreased VWF

levels, higher bleeding score and decreased venous thrombosis in humans (319, 339,

340).

The CHARGE Consortium data yields two important pieces of information. First, a

cluster of SNPs within a single genetic loci identifies a candidate gene. For example,

Page 73: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

50

many SNPs associated with altered VWF levels cluster within the STXBP5 gene.

Second, an individual SNP within a gene identifies a critical functional element within a

gene product. For example, rs1039084 alters STXBP5 residue 436 from Asn to Ser,

which suggests that this N436 is an important functional amino acid of STXBP5. Given

the association between STXBP5 genetic variations and plasma VWF levels, its structural

similarity with SNAREs, and its role in regulating neurotransmitter release, we

hypothesized that STXBP5 regulates endothelial cell exocytosis.

Page 74: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

51

Table 1 Description of STXBP5 SNPs associated with plasma VWF levels.

refSNP Location

(hg19) Variant MAF1

Protein2

Position Residue change

rs9390459

chr6:147680359 G>A 0.4958 815 Leu > Leu

rs1039084

chr6:147635413 A>G 0.4571 436 Asp > Ser

1. MAF, minor allele frequency. Source: 1000 Genomes

2. RefSeq: NP_001121187.1

Page 75: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

52

Scheme 2-1 Schemes of STXBP5 and Lgl family member structures.

(A) STXBP5 is composed of an N-terminal WD40 repeat domain, a C-terminal VAMP-

like domain, and a tail domain in between. The VAMP-like domain of STXBP5 enable it

to bind to the coiled-coil SNARE-domains such as one on syntaxin1.

(B) Structural resemblance of Lgl family proteins. The Lgl family proteins share

structural characteristics in N-terminal WD40 repeats composed of β-propellers. The tail

Page 76: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

53

domain and the C-terminal VAMP-like domain are absent in Lgl. P indicates

phosphorylation sites.

Adapted from Yamamoto et al. Presynaptic Terminals. Springer Japan; 2015:129-40

(341).

Page 77: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

54

2.2 Results

2.2.1 STXBP5 is expressed in human ECs and murine tissues

We first defined the expression of STXBP5 in human cells and in murine tissue. We

performed immunoblotting for STXBP5 on cultured human aortic endothelial cells

(HAEC), human umbilical vein endothelial cells (HUVEC), and human dermal

microvascular endothelial cells (HDMVEC). STXBP5 is expressed as a 130 kDa protein

in all three human endothelial cell types (Figure 2-1A) (321). Stxbp5 mRNA is expressed

in murine tissues, including lung, spleen, and aorta, as measured by qPCR (Figure 2-1B).

Stxbp5 mRNA is also found in murine brain, supporting studies identifying a role for

Stxbp5 in neurovesicle release (321, 330).

We next characterized the STXBP5 isoforms expressed by human tissues and cells. We

performed RT-PCR on RNA from HUVEC, human brain, human platelets, and HEK293

cells using primers flanking the splice regions. We identified 5 transcript variants of

STXBP5, all of which have been predicted but not previously characterized: STXBP5-1

(NM_139244.4), STXBP5-2 (NM_001127715.2), STXBP5-X1 (XR_245502.1), STXBP5-

X2 (XR_245503.1), and STXBP5-X3 (XR_245504.1) (Figure 2-1C). To quantitate the

relative abundance of each splice variant, we also performed qPCR. The STXBP5

transcript variant expressed at highest levels by endothelial cells is STXBP5-1, followed

by STXBP5-2, and other transcript variants are expressed at lower levels (Figure 2-1D).

The STXBP5 splice variant profile in endothelial cells resembles the profile in platelets

Page 78: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

55

(Figure 2-1D). However, human brain expresses one predominant isoform, STXBP5-1,

and very little of the other variants were detected in brain (Figure 2-1D). Other studies

confirm that the same isoform STXBP5-1 is expressed in mammalian brain (321, 322).

2.2.2 STXBP5 inhibits endothelial exocytosis in vitro

We next explored the role of STXBP5 in endothelial exocytosis. We knocked down

expression of endogenous STXBP5 by RNA interference in HAEC, HUVEC, and

HDMVEC, stimulated the cells with the physiological agonist histamine, and then we

measured the amount of VWF released into the media using an ELISA. siRNA directed

against STXBP5 decreased expression of STXBP5 in HAEC, HUVEC, and HDMVEC

(Figure 2-2A-C, upper panel). Knockdown of STXBP5 expression does not affect the

constitutive secretion of VWF under resting conditions (Figure 2-2A-C, white bars).

However, knockdown of STXBP5 expression significantly increases histamine-induced

VWF release into media in all three different EC types (Figure 2-2A-C, black bars).

Similarly, knockdown of STXBP5 in HUVEC increases VWF release induced by ATP

(Figure 2-2D), and by calcium ionophore A23187 (Figure 2-2E), but did not change

VWF release at resting condition.

To further characterize the effect of STXBP5 on constitutive release of VWF, we

measured VWF levels in the media of HUVEC transfected with siControl of siSTXBP5

over an extended period of 12 hours, and found no change in constitutive release (Figure

2-3). STXBP5 does not affect total cellular VWF content (Figure 2-4).

Page 79: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

56

We also tested the idea that STXBP5 affects VWF release by regulating granule number

and morphology. We counted the number of WPB granules in HUVEC transfected with

siControl or with siSTXBP5, and found that STXBP5 does not affect the number of

WPBs per cell (Figure 2-5). Analysis of the size of individual WPB revealed that

STXBP5 does not affect the size of these granules (Figure 2-6). Finally, we measured the

morphology of WPB by quantifying the aspect ratio of individual WPB granules:

STXBP5 does not affect the aspect ratio distribution of granules (Figure 2-7).

We also explored the effect of STXBP5 upon externalization of P-selectin, another

component of WPB translocated by endothelial exocytosis (186). Knockdown of

STXBP5 increases the endothelial externalization of P-selectin upon histamine

stimulation, as measured by HL-60 cell adherence (Figure 2-8).

Taken together, these results suggest that STXBP5 inhibits endothelial exocytosis.

2.2.3 Genetic variation in STXBP5 affects VWF exocytosis in vitro

We also tested the effect of genetic variation upon STXBP5 function. We knocked down

STXBP5 in endothelial cells and then rescued STXBP5 expression with either STXBP5

(WT) or STXBP5 (N436S), a variant of STXBP5 associated with altered VWF levels in

humans (319). STXBP5 (N436S) inhibits VWF secretion more effectively than wild-type

STXBP5 (Figure 2-9).

Page 80: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

57

2.2.4 STXBP5 does not co-localize with endothelial WPBs

Since genetic variants in STXBP5 are associated with alterations in VWF levels, we next

defined the subcellular location of STXBP5 in relation to endothelial granules which

contain VWF as well as other pro-inflammatory and pro-thrombotic mediators. We found

that STXBP5 is located in the cytoplasm, primarily in a punctate pattern (Figure 2-10).

Immunostaining for VWF revealed the typical rod-shaped morphology of WPB (Figure

2-7 and Figure 2-10). Using confocal microscopy, we found that STXBP5 is minimally

co-localized with VWF (Figure 2-10). The morphology of the particles containing

STXBP5 is different from the WPB granules containing VWF, and the overlap is minor

(Figure 2-10). We searched for subcellular location of STXBP5: confocal microscopy did

not reveal a definitive co-location with markers for the ER, Golgi, lysosome, or

endosome compartments (Figure 2-12).

2.2.5 STXBP5 interacts with endothelial exocytic machinery

We next searched for links between STXBP5 and the exocytic machinery in endothelial

cells. Prior studies showed that STXBP5 can inhibit neurotransmitter release from

neurons and insulin release from pancreatic beta-cells by forming complexes with the

SNARE proteins, including syntaxin isoforms and SNAP isoforms (321, 342-344). We

and others have previously shown that endothelial SNARE molecules are likely STX4,

VAMP3, VAMP8, and SNAP23; some of them play an important role in regulating VWF

exocytosis (102, 108, 109, 345). Accordingly, we searched for an interaction between

Page 81: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

58

STXBP5 and endothelial SNARE molecules. We first used sucrose density gradient

ultracentrifugation to specify the cellular components that co-sediment with STXBP5.

We loaded HUVEC lysates on top of a 5%-40% discontinuous sucrose density gradient,

performed ultracentrifugation, and analyzed fractions by immunoblotting. We found that

STXBP5 and STX4 partially co-sediment, suggesting these two proteins might be able to

form a complex in HUVEC, and might interact directly or indirectly with each other

(Figure 2-11A). However, STXBP5 does not co-sediment with others SNAREs that are

involved in endothelial exocytosis, such as SNAP23 (Figure 2-11A).

To further characterize the interaction between STXBP5 and STX4, we

immunoprecipitated HUVEC lysates with antibody to STXBP5 and immunoblotted the

precipitants with antibody to STX4. STXBP5 and STX4 co-precipitate (Figure 2-11B).

However, STXBP5 does not co-precipitate with STX11 or SNAP23 (Figure 2-11B).

We then searched for co-localization of STXBP5 and STX4 by immunofluorescence

staining of HUVEC. Confocal microscopy showed STXBP5 and STX4 are both

distributed in a punctate pattern, and a subset of STXBP5 co-localizes with STX4

(Pearson’s coefficient = 0.80 ± 0.03, Figure 2-11C). Co-localization of STXBP5 and

STX4 was mainly found in the cytoplasm (Figure 2-11C, upper panel). In a subset of

cells, a fraction of STX4 also co-localizes with a relatively sparser fluorescent punctation

of STXBP5 on the cell membrane (Figure 2-11C, lower panel). Taken together, these

data show that STXBP5 interacts with a complex containing STX4 in endothelial cells.

Page 82: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

59

Given the pivotal role that STX4 plays in endothelial vesicle trafficking (108, 109), their

interaction may underline the inhibitory function of STXBP5 in endothelial exocytosis.

We also searched for an association between STXBP5 and SYT1. STXBP5 and SYT1

partially co-sediment (Figure 2-11A). STXBP5 and SYT1 co-precipitate, especially in

cells that have been stimulated with histamine or A23187 (Figure 2-11B). We also

searched for co-localization of STXBP5 and SYT1 by immunofluorescence staining of

HUVEC. In resting HUVEC, STXBP5 and SYT1 are both distributed in a punctate

pattern, and a subset of STXBP5 co-localizes with SYT1 (Pearson’s coefficient = 0.70 ±

0.08, Figure 2-11D). Furthermore, both STXBP5 and SYT1 co-localize to plasma

membranes of HUVEC after stimulation (Pearson’s coefficient = 0.69 ± 0.08, Figure

2-11D, lower row). These data reinforce observations that STXBP5 interacts with SYT1

in neurons (321, 346).

We also searched for interactions of STXBP5 and other molecules that regulate vesicle

trafficking in endothelial cells. STXBP5 does not co-precipitate with STX11 or SNAP23

(Figure 2-11B), or with NSF, Munc18-2, or Munc18-3 (Figure 2-13). Furthermore,

STXBP5 fails to co-localize significantly with other SNARE molecules, such as

SNAP23, VAMP8, or VAMP3 (Figure 2-14). STXBP5 also does not co-localize with

caveolin 1, Munc18-2, Munc18-3, and NSF (Figure 2-15). Finally, STXBP5 does not co-

localize with Rab27a, Myosin 5a, or MyRIP (Figure 2-16).

Page 83: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

60

Taken together, our data suggest that STXBP5 interacts with a complex containing STX4

and SYT1 in endothelial cells.

2.2.6 STXBP5 inhibits endothelial exocytosis in vivo

In order to test whether or not STXBP5 regulates endothelial exocytosis in vivo, we

compared the plasma level of VWF in mice that were Stxbp5 deficient (Stxbp5 KO) and

in wild-type (Stxbp5 WT) mice. VWF levels are higher in Stxbp5 KO mice than in Stxbp5

WT mice (Figure 2-17). These data match our in vitro data showing that VWF release is

greater from endothelial cells with knockdown of STXBP5 compared to control cells

(Figure 2-2). In addition, this difference in VWF levels is not due to the ADAMTS13

enzyme that cleaves VWF in the blood, since ADAMTS13 activity is unchanged in

Stxbp5 WT mice and Stxbp5 KO mice (Figure 2-17B).

To explore the physiological relevance of STXBP5, we measured the effect of STXBP5

upon platelet interactions with endothelial cells in vivo. Platelet rolling or adherence to

the vessel walls is mediated by VWF released by exocytosis (347). We hypothesized that

deficiency of STXBP5 would permit an increase in WPB exocytosis, resulting in an

increase in platelet adherence to venule walls. Anesthetized Stxbp5 KO mice or Stxbp5

WT mice were transfused with fluorescent antibodies to label platelets. The mesentery

was externalized, superfused with 10 µM Ca2+ ionophore A23187 to stimulate WPB

exocytosis, and intravital microscopy was used to record interactions of fluorescently

labeled platelets with mesenteric venules. Platelets that remained static over multiple

Page 84: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

61

frames or were captured by the endothelium in consecutive frames and then released in a

stop-and-go fashion were classified as adherent platelets.

In Stxbp5 WT mice, A23187 rapidly induces platelet adhesion to the venule wall (Figure

2-18A). In Stxbp5 KO mice, A23187-induced platelet interaction with the venule wall

was significantly increased (Figure 2-18A). Quantification of adherent platelets showed

that STXBP5 deficiency significantly increased A23187-induced platelet interactions

with the venule walls, but not the interaction in the resting condition (Figure 2-18B). This

increase in platelet adherence to the venule would be predicted if STXBP5 deficiency led

to increased exocytosis of WPBs and greater release of VWF.

Finally we used an assay that directly measures endothelial exocytosis, so that we would

eliminate the possible effects of STXBP5 upon platelets. Mice were perfused with

fluorescently labeled microspheres conjugated with antibody to P-selectin, and adhesion

to resting and A23187-stimulated mesenteric venules was imaged by intravital

microscopy. In Stxbp5 WT mice, A23187 treatment increases microsphere adherence to

venules (Figure 2-18 C-D). However, in Stxbp5 KO mice, microsphere adherence to

venules increases even more after A23187 stimulation (Figure 2-18 C-D). This suggests

that STXBP5 limits endothelial display of P-selectin in vivo. Taken together, these data

show that STXBP5 inhibits endothelial exocytosis in vivo.

Page 85: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

62

2.2.7 STXBP5 affects hemostasis and thrombosis

We proceeded to test the effect of STXBP5 upon thrombosis. We expected that mice

deficient in Stxbp5 would have increased thrombosis, since they have increased plasma

VWF levels and platelet-endothelium interaction upon stimulation. We first measured the

time for hemostasis in a murine tail bleeding model. Contrary to our expectations, mice

lacking Stxbp5 displayed prolonged bleeding compared to wild-type mice (Figure

2-19A). We next measured the time for thrombosis in a murine mesenteric thrombosis

model. Stxbp5 KO mice had severely delayed time to formation of thrombus and time to

vessel occlusion, compared to Stxbp5 WT mice (Figure 2-19B). We then measured the

time for flow cessation in a murine carotid artery thrombosis model. Although carotid

artery flow ceases approximately 8 min after arterial injury in the wild-type mice, carotid

arteries remain patent for more than 30 min after arterial injury in the Stxbp5 KO mice

(Figure 2-19C). Taken together, these data suggest that mice lacking Stxbp5 have a

defect in thrombosis.

These results were surprising: mice lacking Stxbp5 have elevated plasma levels of VWF,

increased endothelial exocytosis, and increased platelet rolling along the vessel wall – but

also have decreased thrombosis. One explanation for this apparent paradox is that

STXBP5 also regulates platelet secretion.

Page 86: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

63

2.2.8 STXBP5 promotes platelet secretion and activation

To test the hypothesis that Stxbp5 promotes platelet secretion, we first discovered that

murine platelets express Stxbp5 protein (Figure 2-20A). These data match our data

showing that human platelets express mRNA for STXBP5 isoforms (Figure 2-1 C-D).

We then explored the role of Stxbp5 in platelet activation and secretion, using platelets

from wild-type and Stxbp5 KO mice. To study platelet exocytosis of alpha-granules, we

measured externalization of P-selectin by flow cytometery. To study platelet secretion of

dense granules, we measured release of ATP by a luciferase based assay. To study

platelet activation, we measured activation of the integrin GPIIbIIIa by flow cytometery.

Platelets from Stxbp5 KO mice have defective P-selectin externalization, ATP release,

and GPIIbIIIa activation (Figure 2-20B). These data suggest that Stxbp5 facilitates

platelet exocytosis and activation.

To confirm these data, we analyzed wild-type or Stxbp5 KO mice that had received bone

marrow transplantation of either genotypes. We measured plasma VWF levels in Stxbp5

WT recipient mice receiving bone marrow from Stxbp5 WT donors or Stxbp5 KO donors,

and in Stxbp5 KO recipient mice receiving bone marrow from either donors. Plasma

levels of VWF were not changed by bone marrow transplantation (Figure 2-21A).

However, bleeding times increased in mice receiving Stxbp5 KO bone marrow, and

decreased in mice receiving Stxbp5 WT bone marrow (Figure 2-21B). Furthermore,

Stxbp5 WT mice that received Stxbp5 KO bone marrow failed to form an occlusive

thrombus in a carotid artery thrombosis model, but the non-occlusive phenotype of

Page 87: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

64

Stxbp5 KO recipients were completely abolished by transplantation of Stxbp5 WT bone

marrow (Figure 2-21C). Taken together, these data show that Stxbp5 in endothelial cells

determines the plasma VWF level, but the thrombosis phenotype is determined by Stxbp5

in bone marrow cells (such as platelets), not by the host cells (such as endothelial cells).

Page 88: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

65

Figure 2-1 STXBP5 expression and transcript variants.

(A) STXBP5 expression in human endothelial cells. Lysates of cultured human aortic

endothelial cells (HAEC), human umbilical vein endothelial cells (HUVEC), and human

dermal microvascular endothelial cells (HDMVEC) were probed by immunoblotting with

Page 89: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

66

antibody to STXBP5 (top). Blotting to β-actin was used as a loading control (bottom).

Representative of 3 separate experiments.

(B) Stxbp5 expression in murine tissue. RNA was isolated from Stxbp5 WT mouse

tissues, and Stxbp5 mRNA expression was measured by qPCR and normalized to mRNA

level in brain (n=3 mice ± S.D).

(C) Human STXBP5 transcript variants. STXBP5 transcript variants were detected by RT-

PCR using primers flanking splice region in human brain, HUVEC, human platelets, and

HEK293 cells. The products were separated by agarose gel, sequenced, and compared

with NCBI Reference Sequences. The length of each PCR product were: 370 bp

(STXBP5-X1), 322 bp (STXBP5-X2), 307 bp (STXBP5-2), 262 bp (STXBP5-X3), and 199

bp (STXBP5-1). The similarities between PCR products and NCBI entries were 99%

(STXBP5-X1), 95% (STXBP5-X2), 100% (STXBP5-2), 100% (STXBP5-X3), and 100%

(STXBP5-1).

(D) Relative abundance of STXBP5 transcript variants in human brain, HUVEC, and

human platelets were measured by qPCR using variant-specific Taqman probes, with

brain STXBP5-1 set as 100% (n = 3 ± S.D).

Page 90: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

67

Figure 2-2 STXBP5 inhibits endothelial exocytosis in vitro.

HUVEC was transfected with siRNA against STXBP5 (siSTXBP5) or control siRNA

(siControl), stimulated with an agonist, and the amount of VWF released into the media

at resting condition and after 30-min stimulation was measured by an ELISA (n=3 S.D.

Page 91: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

68

* P < 0.05 for agonist treatment vs. control). Immunoblots at the top show siRNA

knockdown of STXBP5 expression in EC, with endogenous actin as loading control.

Shown are noncontiguous parts from the same gel.

(A-C) Knockdown of STXBP5 increases histamine-induced release of VWF, but has no

effect upon constitutive VWF release in (A) HAEC, (B) HUVEC, and (C) HMVDEC.

(D-E) Knockdown of STXBP5 in HUVEC increases VWF release induced by (D) ATP,

and by (E) calcium ionophore A23187.

Page 92: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

69

Figure 2-3 STXBP5 and constitutive release of VWF.

Constitutive release of VWF was measured in 3-h intervals over 12 h in resting HUVEC

transfected with control siRNA or STXBP5 siRNA for 72 hours (n = 3 ± S.D. No

significant difference between siControl and siSTXBP5 was detected over the time

course).

C u ltu re t im e

VW

F r

ele

as

e(m

U/m

l)

3 h 6 h 9 h 1 2 h

0

1

2

3

s iC o n tro l

s iS T X B P 5

Page 93: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

70

Figure 2-4 STXBP5 does not affect HUVEC content of VWF.

Total protein content in lysates of HUVEC transfected with siControl or siSTXBP5 was

measured and normalized by bicinchoninic acid assay, and lysates contain 1 μg total

protein were measured VWF content by ELISA (n = 4 ± S.D. No significant difference

was detected).

Page 94: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

71

Figure 2-5 Knockdown of STXBP5 does not affect the number of WPB granules per

cell.

HUVEC were transfected with siSTXBP5 or siControl, or sham transfected, and the

number of WPB was counted by ImagePro Plus software (n = 3 replicate plates ± S.D.;

granules were counted in over 240 cells per plate. No significant difference among

groups were detected by one-way analysis of variance (ANOVA). Scale bar = 50 μm).

Page 95: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

72

Figure 2-6 Knockdown of STXBP5 does not affect the size of WPB granules.

HUVEC were transfected with siSTXBP5 or siControl, and the size of individual WPB

was measured by ImagePro Plus (n = 3 replicate plates ± S.D.; granules were counted in

over 240 cells per plate. No significant difference between siControl and siSTXBP5 in all

groups).

P>0.05 for all groups

In d iv id u a l W P B S iz e (p ix e l2)

% T

ota

l W

PB

Po

pu

lati

on

3 - 8 8 - 1 3 1 3 - 1 8 1 8 - 2 3 2 3 - 2 8 2 8 - 3 3 3 3 - 3 8 3 8 - 4 3 4 3 - 4 8 > = 4 8

0

5

1 0

1 5

2 0

6 5

7 0

7 5S iC o n tro l

S iSTXBP5

Page 96: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

73

Figure 2-7 Knockdown of STXBP5 does not affect the shape of WPB granules.

HUVEC was transfected with siSTXBP5 or siControl. The aspect ratio of individual

WPB was measured by ImagePro Plus, and its distribution was plotted as histogram (n =

3 replicate plates ± S.D.; granules were counted in over 240 cells per plate. No significant

difference between siControl and siSTXBP5). Shown at the lower panel is one

representative image of WPB and one example of measurement.

Page 97: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

74

Page 98: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

75

Figure 2-8 STXBP5 inhibits HUVEC release of P-selectin.

(A) Knockdown of STXBP5 in HUVEC increases P-selectin externalization as measured

by HL-60 cell adherence after 10 μM histamine stimulation. HL-60 cells were labeled

with calcein AM to allow visualization by a fluorescent microscope. HUVEC treated

with an antibody to P-selectin (anti-CD62) serve as a negative control. Shown are

representative bright-field and corresponding fluorescent images from multiple fields

from 3 wells per transfection per treatment (10 × water immersion lens; scale bar = 150

μm).

(B) Knockdown of STXBP5 in HUVEC increases P-selectin externalization as measured

by HL-60 cell adherence. Cells treated with antibody to P-selectin (Anti-CD62) serve as

negative control (n = 3 ± S.D. * P < 0.05 vs siControl).

Page 99: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

76

Figure 2-9 STXBP5 SNP and release of VWF.

HUVEC were transfected with siSTXBP5 to knock down endogenous STXBP5

expression, and some co-transfected with an expression vector for STXBP5(WT) or

STXBP5(N436S). Release of VWF was measured after treatment with media or

histamine (n = 4 ± S.D. N.S. Not significant. * multiplicity adjusted P <0.05 by two-way

ANOVA followed by the Tukey post test).

Page 100: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

77

Figure 2-10 Particles containing STXBP5 are not co-localized with endothelial

granules containing VWF.

(A) HUVECs were imaged by confocal microscopy after staining with antibodies to

STXBP5 (green), VWF (red), and DNA (blue) (Scale bar = 20 μm; Objective: oil 40 ×;

confocal z-resolution: 0.40 µm).

(B) Enlargement of insets in (A). STXBP5 is minimally co-localized with WPBs (Scale

bar = 5 μm; Objective: oil 40 ×; confocal z-resolution: 0.40 µm). Representative of more

than 3 separate experiments.

Page 101: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

78

Figure 2-11 STXBP5 co-sediments, co-localizes, and co-precipitates with STX4 and

SYT1.

Page 102: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

79

(A) STXBP5 co-sediments with STX4 and SYT1 by sucrose density gradient

fractionation. HUVEC lysate was ultracentrifuged through discontinuous 5%-40%

sucrose gradient, and the fractions were analyzed by SDS-PAGE (T, total proteins of the

lysate before fractionation; P, pellet after fractionation). Representative of 3 separate

experiments.

(B) STXBP5 co-precipitates with STX4 and SYT1. HUVEC lysate treated with media or

histamine or A23187 were immunoprecipitated with antibody to STXBP5 or mouse IgG1;

precipitants were probed with antibody to SYT1 or STX4 or STX11 or SNAP23.

Representative of 3 similar experiments.

(C) STXBP5 co-localizes with STX4 by confocal microscopy. HUVEC were stained for

STXBP5 (green), STX4 (red), and DNA (blue). STXBP5 partially co-localizes with

STX4. Limited cell membrane localization of STX4 and STXBP5 is detectible in a subset

of cells shown in the lower panel (Upper panel: scale bar = 20 μm; Objective: oil 40 ×;

confocal z-resolution: 0.40 µm; lower panel: scale bar = 30 μm; Objective: oil 60 ×;

confocal z-resolution: 0.32 µm). Pearson’s coefficient (green vs red) = 0.80 ± 0.03.

(D) STXBP5 co-localizes with SYT1. Resting and stimulated HUVEC were stained for

STXBP5 (green), SYT1 (red), and DNA (blue). STXBP5 partially co-localizes with

SYT1, and both STXBP5 and SYT1 partially translocate to the membrane after

stimulation. (Scale bar = 20 μm; Objective: oil 60 ×; confocal z-resolution: 0.32 µm).

Page 103: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

80

Pearson’s coefficient (green vs red) =0.70 ± 0.08 for resting cells; 0.69 ± 0.08 for

histamine-stimulated cells.

Page 104: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

81

Page 105: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

82

Figure 2-12 STXBP5 does not co-localize with markers for ER, Golgi, lysosomes, or

endosomes.

Confocal microscopy was used to localize STXBP5, markers for endoplasmic reticulum,

Golgi apparatus, lysosome, or early endosome. STXBP5 does not co-localize with these

organelle markers. Scale bar = 20 μm.

Page 106: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

83

Figure 2-13 STXBP5 does not interact with NSF and Munc family members.

Lysates of resting or stimulated HUVEC were precipitated with antibody to STXBP5 or

IgG, and precipitants were immunoblotted with antibody to NSF or Munc18-2 or

Munc18-3.

Page 107: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

84

Figure 2-14 STXBP5 does not co-localize with SNAP23, VAMP8, or VAMP3.

Confocal microscopy was used to localize STXBP5 (green), SNAP23, VAMP8, VAMP3

(red), and DNA (blue) in HUVEC. STXBP5 does not co-localize with these SNAREs.

Scale bar = 20 μm.

Page 108: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

85

Page 109: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

86

Figure 2-15 STXBP5 does not co-localize with Caveolin-1, Munc18-3, Munc18-2, or

NSF.

Confocal microscopy was used to localize STXBP5 (green), Caveolin-1, Munc18-3,

Munc18-2, NSF (red), and DNA (blue) in HUVEC. STXBP5 does not co-localize with

these proteins in HUVEC. Scale bar = 20 μm.

Page 110: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

87

Figure 2-16 STXBP5 does not co-localize with Rab27, Myosin5a or MyRIP.

Confocal microscopy was used to localize STXBP5 (green), Rab27, Myosin5a, MyRIP

(red), and DNA (blue) in HUVEC. STXBP5 does not co-localize with these proteins in

HUVEC. Scale bar = 20 μm.

Page 111: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

88

Figure 2-17 Stxbp5 affects plasma VWF in mice.

(A) Plasma levels of VWF are higher in Stxbp5 KO mice than in WT mice. Plasma VWF

levels were measured by an ELISA from retro-orbital bleeding (n = 8~12 S.D.; * P <

0.05 vs. WT). (B) Plasma ADAMTS13 activity is similar in Stxbp5 KO and Stxbp5 WT

mice. ADAMTS13 activity in mouse plasma was measured by a fluorescent assay and

expressed as percentage of pooled WT plasma.

Page 112: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

89

Figure 2-18 Stxbp5 inhibits endothelial exocytosis in mice.

(A) Intravital microscopy of platelet-EC interactions in vivo. Platelets were fluorescently

labeled in Stxbp5 WT and Stxbp5 KO mice. 10 µM ionophore A23187 was superfused

onto the mesenteric venule after 120s baseline recording. Platelet adhesion to the

Page 113: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

90

mesenteric venule was continuously recorded by intravital microscopy. Shown are

representative images at baseline (90s) and after A23187 stimulation (345s and 600s).

Magnification 20×.

(B) Stxbp5 deficiency increases platelet-EC interactions in vivo. Platelet adhesion in (C)

was calculated by averaging the number of platelets that remained static over at least two

consecutive frames (frame rate: approximately 31 frames/second). Quantification was

expressed as adherent platelets per minute in an area of 51200 pixel2 (n = 6 mice per

genotype S.D. * P < 0.05 vs. Stxbp5 WT).

(C) Stxbp5 deficiency increases microsphere-EC interactions in vivo. Mice were perfused

with fluorescent microspheres conjugated with antibody to P-selectin, and adhesion of

microspheres was imaged 2 min before and 8 min after A23187 superfusion on

mesenteric venule. Green lines mark borders of vessel within which adherent

microspheres were counted. Magnification 20×.

(D) Quantification of adherent microspheres in (E) (n = 3-4 mice per genotype S.D. * P

< 0.05 vs. Stxbp5 WT).

Page 114: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

91

Page 115: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

92

Figure 2-19 Stxbp5 increases thrombosis in mice.

(A) Stxbp5 regulates bleeding time in mice. Tail bleeding times in WT and Stxbp5 KO

mice were measured after distal 5 mm tail amputation (n = 10-12 ± S.D. *P < 0.01 by

Kolmogorov-Smirnov test).

(B) Stxbp5 regulates mesenteric thrombosis in mice. Intravital microscopy was used to

measure thrombosis of mesenteric arterioles in WT and Stxbp5 KO mice, including the

time to form (left) a 50-pixel diameter thrombus, and (right) full occlusion (n = 3 - 6 ±

S.D. *P < 0.05 vs Stxbp5 WT by Kolmogorov-Smirnov test).

(C) Stxbp5 regulates carotid thrombosis in mice. Carotid arterial flow was measured by a

Doppler flow probe after FeCl3 injury, and flow after FeCl3 wash-off was plotted as

percentage of baseline flow before injury. Stxbp5 KO mice failed to form vessel

occlusion (n = 7 ± S.D. *P<0.05 by repeated measures two-way ANOVA).

Page 116: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

93

Figure 2-20 Stxbp5 regulates platelet secretion and activation.

(A) Stxbp5 expression in platelets from Stxbp5 WT and Stxbp5 KO mice as measured by

immunoblotting. Gapdh was used as loading control.

(B) Stxbp5 promotes platelet secretion. Platelets were isolated from Stxbp5 WT and

Stxbp5 KO mice, and stimulated with PBS (resting) or thrombin at indicated

concentrations. Median fluorescence intensity (MFI) changes as measured by flow

cytometry following P-selectin externalization (left), integrin GPIIbIIIa activation

(middle), and bioluminescence change following ATP release (right) were measured (n =

3 mice per treatment ± S.D. *P < 0.05 vs. Stxbp5 WT).

Page 117: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

94

Page 118: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

95

Figure 2-21 Stxbp5 in platelets increases thrombosis in mice.

(A) Transplantation of bone marrow does not affect plasma VWF levels. Bone marrow

from Stxbp5 WT or Stxbp5 KO mice were transplanted into lethally-irradiated Stxbp5 WT

or Stxbp5 KO recipients, respectively. Plasma VWF levels were measured 6 weeks after

transplantation (n = 5 – 9 ± S.D. NS, not significant).

(B) Transplantation of Stxbp5 KO bone marrow increases Stxbp5 WT tail bleeding time,

whereas transplantation of Stxbp5 WT bone marrow decreases Stxbp5 KO tail bleeding

time. Bone marrow from Stxbp5 WT or Stxbp5 KO mice were transplanted into lethally-

irradiated Stxbp5 WT or Stxbp5 KO recipients. Tail bleeding time was measured 6 weeks

after transplantation (n = 6 – 9 ± S.D. *P < 0.05 by Kolmogorov-Smirnov test).

(C) Transplantation of Stxbp5 KO bone marrow decreases carotid arterial thrombosis in

Stxbp5 WT recipients, whereas transplantation of Stxbp5 WT bone marrow increases

thrombosis in Stxbp5 KO recipients. Bone marrow from Stxbp5 WT or Stxbp5 KO mice

were transplanted into lethally irradiated Stxbp5 WT or Stxbp5 KO recipients. Carotid

arterial flow was measured 6 weeks after transplantation with FeCl3 injury (n = 4 – 7 ±

S.D. * multiplicity adjusted P < 0.05 by repeated measures two-way ANOVA with Tukey

multiple comparisons).

Page 119: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

96

2.3 Discussion

The major finding of Chapter 2 is that STXBP5 regulates endothelial and platelet

exocytosis. STXBP5 interacts with components of the exocytic machinery and inhibits

release of VWF in endothelial cells and in mice. STXBP5 also promotes platelet

secretion and thrombosis in mice. Thus, our current study provides strong functional

evidence for the regulatory role on circulating VWF and thrombosis by a candidate gene

identified by GWAS (317-320, 339).

We find that STXBP5 inhibits endothelial exocytosis. Others have found that STXBP5

inhibits secretion from other cell types. STXBP5 was originally identified as a protein

(called tomosyn) that inhibits neurotransmitter release from neurons (321). Subsequent

studies showed that STXBP5 decreased exocytosis in yeast, C. elegans synapses,

pancreatic β-cells and neurosecretory cells (329, 331-337). We found that STXBP5

inhibits endothelial exocytosis of granules. Cells with decreased STXBP5 have increased

VWF release into the media and increased cell membrane display of P-selectin; and mice

lacking Stxbp5 have elevated plasma VWF levels and greater display of P-selectin

(Figure 2-2, Figure 2-8, and Figure 2-18).

STXBP5 might regulate endothelial exocytosis through several potential mechanisms.

One proposed model is that STXBP5 blocks formation of a ternary SNARE complex by

competing with VAMP isoforms to interact with SNAP and syntaxin isoforms. For

example, in neurons the VAMP-like domain in the carboxy-terminus of STXBP5 forms a

Page 120: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

97

SNARE complex-like structure with SNAP25 and syntaxin 1A. This STXBP5-SNARE

complex is stable: VAMP2 cannot displace STXBP5, resulting in inhibition of SNARE

complex assembly (328-330, 348). Alternatively, another proposal is that STXBP5

enhances oligomerization of the ternary SNARE complexes, locking the complexes into a

structure that cannot proceed with membrane fusion. For example, in neurons the N-

terminal WD40 repeat domain of STXBP5 functions as a scaffold that enhances

oligomerization of pre-assembled SNARE complexes and reduces vesicle priming and

turnover (332). In addition to these STXBP5-SNARE interaction models, Sro7, the yeast

STXBP5 homolog, acts as an allosteric regulator of exocytosis. The tail domain of Sro7

interacts with the Rab GTPase Sec4, raising the possibility that STXBP5 might be able to

interact with some other factors that control exocytosis (324, 349). Our observation that

STXBP5 interacts with a complex containing STX4 (Figure 2-11) favors the STXBP5-

SNARE interaction model. STX4 is a t-SNARE that binds to cognate v-SNAREs. The

inhibition of VWF exocytosis is probably a consequence of displacement of the

endothelial v-SNARE by STXBP5, forming a “dead-end” complex and sequestering the

formation of productive SNARE complexes.

STXBP5 can interact with either syntaxin or SNAP isoforms, as reported previously

(321, 325, 329, 350). However, our data seem to favor the STXBP5-syntaxin binary

interaction over the STXBP5-syntaxin-SNAP ternary interaction or STXBP5-SNAP

interaction in ECs, since co-localization and co-IP studies failed to detect an interaction

between STXBP5 and SNAP23. Indeed, syntaxin has been shown to be responsible for

STXBP’s inhibition of exocytosis and subcellular localization (350). The VLD of

Page 121: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

98

STXBP5 interacts with syntaxins in various cell types (321, 328-332, 335, 337, 350-353).

In PC12 cells, overexpressed STXBP5 showed a primarily cytosolic localization similar

to endothelial cells, but overexpression of syntaxin directs approximately 55% of

cytosolic STXBP5 to the plasma membrane (350). In contrast, whether a binary

interaction between STXBP5 and an SNAP isoform directs exocytosis has been

controversial. For instance, glutathione S-transferase (GST)-syntaxin-1a could bind to

STXBP5 overexpressed in COS7 cells, but GST-SNAP25 could not bind to STXBP5

under the same condition (321). Other studies, however, demonstrate STXBP5 interacts

with SNAP homologs in yeas (325), adipocytes (354), and PC12 cells (331), but whether

this interaction is formed indirectly via a ternary complex containing syntaxin is

unknown. Although we cannot exclude the existence of such a ternary complex in EC,

our data agrees with a recent study showing STXBP5 interacts mainly with syntaxin, and

STXBP5-SNAP interaction is not abundant and relies on the presence of syntaxin (350).

Another possible mechanism for STXBP5 inhibition of exocytosis is through its

interaction with a calcium sensor, SYT1. SYT1 regulates SNARE-mediated vesicle

fusion by bridging membrane fusion through a calcium-dependent mechanism (355-358).

SYT1 has been shown to be recruited to pseudogranules containing heterologously

expressed VWF (359). Others have shown that STXBP5 interacts with SYT1 through

WD40 repeat domain in the amino-terminus of STXBP5, blocking the ability of SYT1 to

mediate calcium-dependent neurotransmitter release (346). We also found that STXBP5

interacts with SYT1 with high affinity after histamine stimulation (Figure 2-11). This

raises the possibility that calcium triggers exocytosis in part by relieving the inhibitory

Page 122: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

99

effects of STXBP5, either by displacing STXBP5 from t-SNAREs or by promoting an

interaction between STXBP5 and SYT1.

Our data raise some interesting issues. First, our data suggest STXBP5 inhibits secretory

release of VWF but not constitutive release of VWF (Figure 2-2). Recent studies suggest

that endothelial cells release VWF through three distinct pathways: stimulated secretion,

unstimulated basal release, and unstimulated constitutive release (266). However, the

proteins that mediate unstimulated release have not been defined, and it is unknown

which proteins regulate both the unstimulated release pathway and the stimulated

pathway. Our data suggest that STXBP5 acts upon one or more proteins in the stimulated

secretory pathway. For example, our data show that STXBP5 interacts with STX4.

Furthermore, others have shown that in neurons, STXBP5 interacts with SYT1 whose

regulatory role in exocytosis depends on calcium (346).

One striking finding of our study is that STXBP5 has opposing effects on exocytosis

from endothelial cells and platelets: STXBP5 inhibits endothelial exocytosis but activates

platelet exocytosis. Platelets from Stxbp5 KO mice have impaired secretion of alpha-

granules as measured by P-selectin externalization, defective exocytosis of dense

granules as measured by ATP release, and blunted activation as measured by GPIIbIIIA

conformational changes (Figure 2-20B). Furthermore, mice lacking Stxbp5 display severe

defects in bleeding time, mesenteric arteriole thrombosis, and carotid artery thrombosis.

Bone marrow transplant experiments suggest that this hemostasis defect is due to bone

Page 123: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

100

marrow-derived cells, presumably platelets, lacking STXBP5, not endothelial cells

lacking STXBP5 (Figure 2-21).

Why does STXBP5 promote platelet exocytosis but suppress endothelial exocytosis? We

considered the idea that platelets and endothelial cells express different isoforms of

STXBP5, but our PCR studies show that both cell types express predicted STXBP5

transcript variants in similar relative abundance (Figure 2-1D). Another possibility is that

STXBP5 interacts with different SNARE members in different cell types. Although

STXBP5 interacts with STX4 and SYT1 in endothelial cells (Figure 2-11), STXBP5 does

not significantly interact with STX4 or SYT1 in human platelets (data not shown), or

with STX4 in rat brain (321, 322). Others have reported divergent effects of STXBP5 in

differing cell systems. For example, STXBP5 inhibits neurotransmitter release from

neurons (331-337). But STXBP5 actually increases the readily releasable pool of vesicles

in PC12 cells and in rat β-cell line INS-1E; this stimulation of exocytosis depends in part

on the phosphorylation of STXBP5 (336, 353). Detailed studies of molecular

mechanisms are needed to reconcile the dual stimulatory and inhibitory role of STXBP5

in different cell types.

Mice lacking Stxbp5 have increased plasma levels of VWF but decreased thrombosis.

Why? One possible explanation is that STXBP5 has opposing effects upon endothelial

and platelet exocytosis. In Stxbp5 null mice, the increased plasma VWF is caused by the

absence of endothelial STXBP5 that normally limits endothelial release of VWF. Since

the major source of VWF in mouse plasma is from endothelial cells, not from platelets

Page 124: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

101

(195), mice lacking Stxbp5 in both endothelial cells and platelets demonstrate increased

plasma VWF. Our data with bone marrow transplanted mice supports this conclusion,

since plasma VWF levels are unchanged by bone marrow from Stxbp5 KO donors

(Figure 2-21). In Stxbp5 null mice, the decreased thrombosis is caused by the absence of

platelet STXBP5 that normally boosts platelet activation. The enhanced plasma VWF is

not sufficient to overcome the diminished platelet activation, and the net result of

STXBP5 absence from both endothelial cells and platelets is defective thrombosis.

Human GWAS show that multiple genetic variants associated with altered VWF levels

lie within the STXBP5 allele (317-319, 340). One SNP highly correlated with altered

VWF levels in human, rs1039084, is located in exon 23 of STXBP5 (319). This non-

synonymous SNP rs1039084 changes 1307 A to G in STXBP5, changing codon 436 from

Asn to Ser (N436S). Human studies have found that this mutation STXBP5 (N436S) is

associated with lower levels of VWF in the plasma (319).

We found that this STXBP5 (N436S) mutant decreases endothelial exocytosis more than

STXBP5-WT (Figure 2-9). Residue N436 lies within the second N-terminal WD40 repeat

domain of STXBP5. WD40 repeats can form scaffolds which assemble protein

complexes, and it has been previously shown that the STXBP5 WD40 repeat domain

interacts with SNAREs STX1 and SNAP25 in neurons (332). How might the STXBP5

(N436S) mutation enhance inhibition of exocytosis? Prior studies show that the integrity

of the WD40 domain is critical for STXBP5’s inhibition of exocytosis (331). PC12 cells

expressing mutant rat m-STXBP5 lacking two fragments (aa 537–578 and aa 897–917) in

Page 125: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

102

the WD40 domain lost the inhibition of exocytosis compared with cells expressing the

wild type m-STXBP5 (335). These mutants also showed altered binding and diffusion

kinetics of STXBP5 on the PC12 cell membrane compared to wild-type m-STXBP5,

possibly via altered interactions with syntaxin or with other unknown membrane proteins

(350). In addition, using a liposome co-flotation assay, both the N-terminal domain and

C-terminal domain (VLD) of STXBP5 are required for the binding to the syntaxin

monomer, suggesting features unique to the full-length STXBP5. It is possible that the

mutation STXBP5 (N436S) alters the function of its WD40 domain and enhances the

affinity of STXBP5 for STX4 or SYT1 in endothelial cells, blocking the formation of a

ternary SNARE complex or blunting the Ca2+ triggering of membrane fusion. Another

possibility is that the mutation decreases the conformational changes that might permit

STXBP5 to release SNAREs to form the ternary SNARE complex. Or perhaps the

STXBP5 (N436S) mutation serves as a gain-of-function mutation that mediates the

interaction of STXBP5 with additional partners.

Genetic variation within STXBP5 is linked to the risk of venous thromboembolic events

in human subjects (320). The effect of the mutation STXBP5 (N436S) upon platelet

activation is unknown. We found that the mutation STXBP5 (N436S) enhances the

function of STXBP5 in endothelial cells, with enhanced suppression of exocytosis

(Figure 2-9). We would expect that the mutation would also enhance the function of

STXBP5 in platelets, with further promotion of platelet activation. However, in human

studies, the SNP rs1039084 for STXBP5 (N436S) is associated with a decreased

incidence of venous thromboembolic disease (320). It is possible that different partners

Page 126: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

103

interacting with STXBP5 in platelets and endothelial cells determines the effect of the

N436S mutation. Further studies are needed to determine the effect of mutations upon

STXBP5 function in platelets.

In conclusion, our data identify a key role for STXBP5 as a regulator in endothelial

exocytosis and thrombosis. STXBP5 inhibits stimulated endothelial release of VWF and

translocation of P-selectin in vitro. STXBP5 co-localizes with endothelial vesicles and

forms complexes with endothelial SNARE STX4. Moreover, Stxbp5 deficiency in mice

significantly increases plasma VWF levels and P-selectin translocation. However,

STXBP5 also promotes platelet secretion, and mice lacking Stxbp5 have increased

bleeding (Scheme 2-2). Our studies are relevant to humans, since human GWAS data

associates STXBP5 genetic variants with plasma VWF levels and thombosis. Further

characterization of the regulatory functions of STXBP5 in exocytosis may lead to novel

insights into vascular diseases such as VTE and VWD.

Page 127: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

104

Scheme 2-2 STXBP5 differentially influences exocytosis in platelets and endothelial

cells.

Adapted from Lillicrap D. The Journal of Clinical Investigation. 2014;124(10):4231-3.

(360).

Page 128: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

105

VAMP8 Mediates Endothelial Exocytosis

Page 129: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

106

3.1 Introduction

The ternary SNARE complex is typically comprised of one member from each of the

following families: syntaxin (t-SNARE), SNAP (t-SNARE) and VAMP (v-SNARE).

Unique combinations of three SNARE molecules play a role in directing granules to

specific target membranes.

The molecular components of the ternary SNARE machinery has been intensively

studied in neurons and many neuroendocrine and immune cells. Neurotransmission is

regulated in part by VAMP2 on synaptic vesicles interacting with STX1 and SNAP25 on

the pre-synaptic plasma membrane (361). Compound exocytosis in mast cells is mediated

in part by VAMP8 on secretory granules in conjunction with SNAP23 and STX4(362-

366). The three SNAREs controlling alpha-granule exocytosis from platelets include

STX11(367), VAMP8 on alpha-granule membranes (368-372), and SNAP23 that

distributes predominantly on platelet membranes as well as on membranes of granules

and the platelet open canalicular system (367-369, 373-378).

The exact identities of endothelial v-SNAREs are ambiguous and poorly understood. A

prior study found VAMP3 and VAMP8 associated with endothelial WPBs (102).

However, the function of these VAMPs was not fully characterized. More importantly,

the importance of endothelial v-SNARE in regulating exocytosis and thrombosis in vivo

is unknown.

Page 130: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

107

We hypothesized that endothelial exocytosis is regulated by unique v-SNARE molecules.

Chapter 3 shows that VAMP8, in addition to VAMP3, is a key SNARE in endothelial

cells that drives granule exocytosis and affects plasma VWF and thrombosis.

Page 131: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

108

3.2 Results

3.2.1 Endothelial cells express a distinct set of SNAREs that mediate

granule exocytosis

We hypothesized that a distinct subset of SNAREs would mediate endothelial exocytosis.

We searched for endothelial expression of SNAREs by immunoblotting endothelial cell

lysates, and compared it to the known expression of SNAREs in brain, platelets, PC12 or

HeLa cells (as positive controls) or absence of SNAREs in human vascular smooth

muscle cells (VSMC, as a negative control). We found that HUVEC express certain

SNAREs such as STX4, SNAP23, VAMP3 and VAMP8 (Figure 3-1A-B), but have

undetectable level of other SNAREs such as STX1, SNAP25, VAMP1, and VAMP2.

Intriguingly, different endothelial cell types (such as HUVEC, HAEC, and human brain

microvascular endothelial cell (HBMEC)) express different levels of VAMP3 and

VAMP8 (Figure 3-1C). We focused our attention on four of these SNAREs, VAMP3,

VAMP8, STX4 and SNAP23, since they can interact with each other in other biological

systems (365, 379, 380).

3.2.2 VAMP8 and VAMP3 co-localize with VWF

We next performed confocal microscopy to see which SNARE is associated with

endothelial granules containing VWF, the WPBs (180). We found that VWF is expressed

in a granular pattern throughout endothelial cells (Figure 3-2A-D, green). Neither STX4

Page 132: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

109

nor SNAP23 co-localizes with VWF (Figure 3-2A-B, red). However, VAMP8 and

VAMP3 are both expressed in a granular pattern that partially overlaps with VWF

(Figure 3-2C-D, red and merge). To quantify the extent of SNARE co-localization with

VWF, we analyzed the confocal images using FV10-ASW 4.0 software (Olympus). The

Pearson’s correlation coefficient for SNAP23 and VWF is only 0.21 ± 0.04, and for

STX4 and VWF is only 0.15 ± 0.02. However, the Pearson’s correlation for VAMP8 and

VWF is 0.49 ± 0.05, for VAMP3 and VWF is 0.40 ± 0.03. Thus the confocal microscopy

data suggests that VAMP3 and VAMP8 co-localize with VWF in endothelial cells.

Although VAMP3 and VAMP8 co-localize with WPBs respectively, these two SNAREs

do not seem to co-localize with each other, as the granular structures that stain positive

for each molecule are distributed in distinct cellular compartments and are minimally

overlapped with a Pearson’s correlation coefficient of 0.27 ± 0.02 (Figure 3-2E).

3.2.3 VAMP8 and VAMP3 interact with the SNAREs STX4 and

SNAP23

SNAREs drive membrane fusion by forming a complex of three interacting SNAREs.

We next sought to determine whether or not VAMP8 or VAMP3 can interact with STX4

or SNAP23, which are target membrane SNAREs expressed in endothelial cells.

Immunoprecipitation of VAMP8 followed by immunoblotting of precipitates reveals that

VAMP8 interacts with STX4 and SNAP23 in endothelial cells (Figure 3-3A). Similar

interaction were detected between VAMP3 and STX4, or VAMP3 and SNAP23 (Figure

3-3B). Conversely, immunoprecipitation of STX4 revealed comparable interactions

Page 133: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

110

among STX4 with SNAP23, VAMP3 or VAMP8 (Figure 3-3C). These results suggest

that VAMP8 and VAMP3 interact with SNAREs STX4 and SNAP23 in endothelial cells.

3.2.4 VAMP8 and VAMP3 mediates endothelial exocytosis in vitro

To examine the role of VAMP8 and VAMP3 in exocytosis, we knocked down VAMP8

or VAMP3 expression by transfecting HUVEC with control siRNA or siRNA directed

against VAMP8 or VAMP3 (Figure 3-4A). Knockdown of VAMP8 does not affect

expression of other SNARE components or the thrombin receptor (Figure 3-4B). We then

stimulated these transfected HUVEC with histamine or thrombin and measured VWF

release into culture media. Histamine or thrombin increase VWF release in control

siRNA-treated cells, as expected (Figure 3-4 C, E, siControl). However, knockdown of

VAMP3 or VAMP8 decreases VWF exocytosis, and knockdown of both VAMP3 and

VAMP8 further decreases exocytosis (Figure 3-4 C, E). Importantly, knockdown of

VAMP3 or VAMP8 does not affect WPB granule number per cell (Figure 3-5), VWF

protein expression (Figure 3-5), and individual WPB size or shape (Figure 3-6).

Since siRNA directed against VAMP8 might have off-target effects, silencing genes

other than VAMP8, we next performed a rescue experiment. We used siRNA to

knockdown VAMP8 in endothelial cells, and then over-expressed VAMP8 in these cells

(The vector encoding VAMP8 lacks the siRNA target sequence, so the siRNA for

VAMP8 targets endogenous VAMP8 but not exogenous VAMP8). Endothelial cells

normally express VAMP8, siRNA knocks down VAMP8, and transfection of a VAMP8

Page 134: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

111

vector restores VAMP8 expression (Figure 3-4 D). We then treated these cells with

medium or thrombin and measured exocytosis. Thrombin activates exocytosis in control

transfected cells (Figure 3-4 E, left). Silencing VAMP8 blocks exocytosis (Figure 3-4 E,

middle). Over-expression of VAMP8 restores exocytosis (Figure 3-4 E, right). These

experiments support the hypothesis that VAMP8 mediates endothelial exocytosis.

3.2.5 VAMP8 mediates endothelial exocytosis in vivo

To examine the role of VAMP8 in exocytosis in mice, we examined wild-type mice

(Vamp8 WT) and in Vamp8 null mice (Vamp8 KO). Vamp8 is expressed at highest levels

in lung, pancreas, and spleen; and Vamp8 is expressed at lower levels in the liver and

kidney (Figure 3-7A). Plasma levels of VWF are higher in Vamp8 WT mice than in

Vamp8 KO mice (Figure 3-7B). We then measured exocytosis in vivo using intravital

microscopy. We injected Rhodamine 6G into mice to label their leukocytes, and then

performed intravital microscopy of mesenteric venules to measure leukocyte-endothelial

interactions. At baseline, there are fewer rolling leukocytes in Vamp8 KO mice than in

Vamp8 WT mice (Figure 3-7C-D). After stimulation of exocytosis by superfusing

A23187, an ionophore that induces endothelial exocytosis by directly causing

intracellular calcium surge, the number of rolling leukocyte increases, but the rolling cell

increase is much greater in the wild-type mice than in the knockout mice (Figure 3-7C-

D). To further investigate whether Vamp8 KO mice have defects in VWF exocytosis, we

gave mice subcutaneous epinephrine injection to induce VWF secretion, and collected

blood before and 30 min after epinephrine challenge. In Vamp8 WT mice, VWF levels

Page 135: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

112

increase significantly in response to epinephrine injection (Figure 3-7E; fold increase of

1.50 ± 0.42, mean ± S.D.) However, Vamp8 KO mice fail to respond significantly to

epinephrine stimulation (Figure 3-7E; fold increase of 1.24 ± 0.36, mean ± S.D.) This

lack of VWF release after epinephrine stimulation is consistent with our in vitro

observation (Figure 3-4 C). Finally, bleeding times are prolonged in Vamp8 KO mice

compared to Vamp8 WT mice (Figure 3-7F).

Page 136: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

113

Figure 3-1 Expression of SNAREs in endothelial cells.

(A) Differential expression of VAMP8 and VAMP3 in different human endothelial cells

assessed by immunoblotting.

(B) SNAREs expression in mouse brain and HUVEC measured by immunoblotting.

(C) VAMP8 expression in lysates of HUVEC, human platelets, HeLa cells, PC12 cells,

or human VSMC measured by immunoblotting.

Page 137: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

114

Page 138: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

115

Figure 3-2 Subcellular localization of VWF and SNAREs in endothelial cells.

HUVEC was stained with antibodies to VWF and various SNAREs and imaged with

confocal microscopy. Shown are representative images of green (Alexa Fluor 488, left

column), red and blue (Alexa Fluor 594 or Cy3, DAPI, middle column), and merged

(right column) stains highlighting co-localization of the antibody-binding sites (yellow

signals). Images were taken on an Olympus IX81 confocal microscope at room

temperature. Original magnification ×60. The confocal images were analyzed by FV10-

ASW 4.0 software (Olympus). The stained intracellular antigens are:

(A) VWF and Syntaxin 4;

(B) VWF and SNAP23;

(C) VWF and VAMP8;

(D) VWF and VAMP3;

(E) VAMP8 and VAMP3.

VWF co-localizes with VAMP8 with calculated Pearson's coefficient of 0.49 ± 0.05 (C).

VWF co-localizes with VAMP3 with calculated Pearson's coefficient of 0.40 ± 0.03 (D).

Page 139: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

116

Figure 3-3 Interaction of SNAREs in endothelial cells.

HUVEC lysates were immunoprecipitated (IP) with antibodies to VAMP8, VAMP3,

STX1, or STX4, and immunoblotted (IB) with antibodies to SNARE proteins. Input

represents 2% of total cell lysate; equal amount of lysate were used for all IP lanes. Co-

immunoprecipitation was detected for

(A) VAMP8 with STX4 and SNAP23;

(B) VAMP3 with STX4 and SNAP23; and

(C) STX4 with SNAP23, VAMP3, and VAMP8.

Page 140: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

117

Page 141: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

118

Figure 3-4 VAMP8 and VAMP3 mediate endothelial exocytosis.

HUVEC were transfected with control siRNA (siControl) or siRNA for VAMP3

(siVAMP3) or VAMP8 (siVAMP8), and treated with 10 uM histamine or 1 U/mL

thrombin. The release of VWF into media was measured by ELISA.

(A) Immunoblots show siRNA knockdown of VAMP3 and VAMP8 expression.

(B) Knockdown of VAMP8 does not affect expression of a subset of endothelial SNARE

molecules or the thrombin receptor PAR1, as shown by immunoblotting.

(C) Measurements of VWF release in siRNA-treated cells (n = 4-6 ± S.D. *P < .05 vs.

siControl with histamine). Knockdown of VAMP3, VAMP8, or both, decreases

exocytosis.

(D) HUVEC was transfected with siRNA for endogenous VAMP8 or control siRNA, and

then co-transfected with a vector expressing c-myc tagged VAMP8 (resistant to siRNA).

Immunoblots show siRNA knockdown of endogenous VAMP8 expression (middle lane)

and rescue of VAMP8 expression after knockdown (right lane).

(E) The transfected cells in (C) were treated with thrombin, and the release of VWF was

measured. Knockdown of VAMP8 decreases exocytosis and rescue of VAMP8 restores

endothelial exocytosis (n = 6 ± S.D. *P = 0.004 vs. siControl with thrombin; P = 0.14

between siControl with thrombin vs siVAMP8 + VAMP8-vector with thrombin,

suggesting no significant difference between control cells and VAMP8-rescued cells).

Page 142: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

119

Page 143: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

120

Figure 3-5 Knockdown of VAMP3 or VAMP8 does not affect endothelial granule

number or VWF content.

(A) Endothelial granules with VWF in HUVEC transfected with siControl, siVAMP3, or

siVAMP8. Shown are stacked confocal images over the entire HUVEC monolayer,

representative of > 5 dishes, with VWF stained with an anti-VWF antibody (green) and

DNA stained with DAPI (blue). Scale bar = 50um.

(B) Knockdown of VAMP3 or VAMP8 does not affect WPB number per cell. HUVEC

was transfected with siControl, siVAMP3, or siVAMP8, cultured for 72 hs, followed by

immunofluorescence staining of WPBs using an anti-VWF antibody. The number of

WPBs were counted over stacks of confocal images that cover the HUVEC monolayer in

multiple culture dishes (siControl, n = 6 dishes ± S.D., 345 cells; siVAMP3, 5 dishes ±

S.D., 347 cells; siVAMP8, 6 dishes ± S.D., 390 cells. NS, not significant.).

(C) HUVEC was transfected with siControl, siVAMP3, siVAMP8, or siVAMP3 and

siVAMP8, cultured for 72 hs, followed by cell lysis. After protein normalization with

BCA assay for each sample, 7.6 ug total cell lysate in 100 ul volume was measured for

VWF content by an ELISA. (n = 4-6 ± S.D. NS, not significant vs. No Treatment).

Page 144: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

121

Page 145: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

122

Figure 3-6 Knockdown of VAMP3 or VAMP8 does not affect WPB morphology.

(A) The size of WPB granules is not changed by VAMP3 or VAMP8 knockdown.

HUVEC were transfected with siControl, siVAMP3, or siVAMP8, imaged using a

confocal microscope as in Figure S3, and the size of individual WPB was measured over

z-stacks of optical sections by ImagePro Plus (n = 5-6 replicate plates ± S.D.; granules

were counted in over 300 cells per plate. No significant difference among siControl,

siVAMP3, and siVAMP8 by one-way ANOVA).

(B) Knockdown of VAMP3 or VAMP8 does not affect the shape of WPB granules.

HUVEC were transfected with with siControl, siVAMP3, or siVAMP8, imaged using a

confocal microscope as in Figure S3. Optical sections were captured to generate z-stacks

over the entire HUVEC monolayer. The aspect ratio of individual WPB was measured by

ImagePro Plus, and its distribution was plotted as histogram (n = 5-6 replicate plates ±

S.D.; granules were counted in over 300 cells per plate. No significant difference among

siControl, siVAMP3, and siVAMP8 by one-way ANOVA).

Page 146: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

123

Page 147: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

124

Figure 3-7 Vamp8 mediates endothelial exocytosis and leukocyte rolling in vivo.

(A) Expression of Vamp8 in mice. Immunoblot of Vamp8 in wild-type mice (Vamp8 WT,

top) and Vamp8 knockout mice (Vamp8 KO, bottom) shows Vamp8 expression in lung,

pancreas, and spleen, and lower levels of Vamp8 expression in liver and kidney.

(B) Vamp8 regulates plasma VWF levels in mice. VWF was measured in the plasma of

wild-type mice and Vamp8 null mice. VWF levels are higher in wild-type mice than in

Vamp8 KO mice (n = 11 - 21 ± S.D. *P < .05).

(C) VAMP8 mediates endothelial interactions with leukocytes. Intravital microscopy

was used to measure leukocyte rolling along mesenteric venules in non-treated mice for 2

min and for 8 min after stimulation with the calcium ionophore A23187. Video were

captured with an electron-multiplying CCD video camera (QUANTEM, 512SC) and

were analyzed using Image-Pro Analyzer 6.2 software (Media Cybernetics). Original

magnification ×20. Shown are epresentative videomicrographs of wild-type and Vamp8

KO mice before and after treatment with A23187.

(D) Quantitation of leukocyte rolling from data above (n = 6 ± S.D. *P < .05 for WT vs.

KO). The average number of rolling leukocytes per unit area (512 pixel × 100 pixel)

were counted with Image-Pro Analyzer 6.2 software (Media Cybernetics) for

quantification. Vamp8 mediates endothelial interactions with leukocytes, and absence of

Vamp8 decreases endothelial-leukocyte interactions.

Page 148: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

125

(E) Plasma VWF levels in wild-type (n = 13) and Vamp8 KO mice (n=15) before and

after epinephrine injection. Values are expressed relative to the level of VWF in the

pooled plasma of 10 wild-type mice and 10 Vamp8 KO mice, respectively. *P < .05 for

WT epinephrine vs. baseline. NS, not significant.

(F) Vamp8 regulates bleeding time. The tail bleeding time in wild-type and Vamp8 KO

mice was measured. Vamp8 KO mice have a prolonged bleeding time compared to wild-

type mice (n = 9 - 14 ± S.D. *P < .05).

Page 149: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

126

3.3 Discussion

Chapter 3 identifies VAMP8 as a critical component of the machinery that controls

exocytosis of endothelial granules. Endothelial cells express VAMP8, VAMP8 co-

localizes with endothelial granules, and VAMP8 interacts with additional SNAREs in

endothelial cells. Finally, knockdown of VAMP8 inhibits exocytosis of endothelial cells

in vitro; and mice lacking Vamp8 have impaired secretion of VWF in vivo.

Although endothelial cells express a variety of SNAREs that mediate vesicle trafficking,

the set of three SNAREs that regulate endothelial granule fusion with the plasma

membrane is not full characterized, except for that STX4 has been shown to play a role in

endothelial exocytosis (108, 109). Endothelial cells express the SNAP isoform SNAP23

which is likely one of the three components of the SNARE complex (110-113). Others

have shown that endothelial cells express VAMP family members (102-107). But the

precise identity of the v-SNARE that is associated with endothelial granules and mediates

endothelial exocytosis is unclear.

VAMP8 regulates endothelial exocytosis in vitro and in vivo

Our data show that VAMP8 mediates endothelial exocytosis in vitro. VAMP8 is

associated with endothelial granules, and VAMP8 interacts with other endothelial

SNAREs. Knockdown of VAMP8 decreases endothelial release of VWF, and rescue of

VAMP8 expression increases VWF secretion. Furthermore, our in vivo data support a

Page 150: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

127

role for VAMP8 in mediating exocytosis. Mice lacking Vamp8 have lower levels of

plasma VWF and epinephrine-induced VWF release, and since most plasma VWF and

epinephrine-induced VWF release is from endothelial cells rather than platelets (195),

these data support the idea that Vamp8 mediates endothelial exocytosis in vivo.

Furthermore, mice lacking Vamp8 have a hemostasis defect, supporting the work of

others who show that endothelial derived VWF is a major contributor to hemostasis

(195). Finally, mice lacking Vamp8 have decreased leukocyte trafficking, further

supporting the central role of VAMP8 in regulating endothelial exocytosis.

Although our study supports a major role for VAMP8 regulation of endothelial

exocytosis in vitro and in vivo, one prior study suggests that VAMP8 does not regulate

endothelial exocytosis (102). But several aspects of this prior study are not as robust as

our current data. First, the prior study does not trigger exocytosis with a physiological

agonist applied to intact cells; instead the prior study adds exogenous calcium to

permeabilized cells. Second, the prior study does not demonstrate that VAMP8

expression or function is impaired in experimental assays before measuring exocytosis.

Finally, the prior study does not employ in vivo studies of VAMP8. In contrast, our data

use intact endothelial cells, genetic silencing techniques, and knockout mice in vivo to

show that VAMP8 mediates exocytosis and controls VWF levels.

VAMP8 in Platelet Exocytosis

Page 151: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

128

Our studies also support the work of others which show that VAMP8 plays a prominent

role in regulating secretion of alpha-granules, platelet granules that resemble endothelial

WPB. Platelets express VAMP8 (370-372, 381, 382), VAMP8 is associated with alpha-

granules (382), platelets from mice lacking VAMP8 secrete less VWF and externalize

less P-selectin than platelets from wild-type mice (371), and mice lacking VAMP8 have

delayed thrombus formation(372). In addition, humans with genetic variation in the gene

encoding VAMP8 can have increased platelet reactivity and an increased rate of arterial

thrombosis (381, 383). However, it is unclear if thrombosis in this subset of patient is

due to abnormalities in endothelial or platelet secretion (384) .

VAMP3 and Endothelial Granule Trafficking

Our data show that VAMP3 regulates endothelial exocytosis in vitro. VAMP3 co-

localizes with granules, interacts with SNAREs, and is required for secretion of VWF.

Our data support the findings of one prior study showing VAMP3 regulates endothelial

exocytosis in vitro (102). However, the function of VAMP3 in vesicle trafficking remains

controversial, since some studies show VAMP3 regulates exocytosis and others show

VAMP3 regulates endocytosis and vesicle recycling. VAMP3 may play one or several

roles in endothelial granule trafficking. VAMP3 might play a role in granule biogenesis

by transporting VWF and other cargo between ER and Golgi or between Golgi and

granule; but this possibility is not supported by our data or data of others (since we find

that endothelial cells contain normal numbers of granules and normal levels of VWF

even after VAMP3 knockdown). VAMP3 might mediate granule exocytosis by

Page 152: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

129

functioning as a vesicle SNARE on WPBs and interacting with target membranes; data

from our studies and from other studies support this idea in vitro (for example, some in

vitro studies show that VAMP3 controls exocytosis of vesicles from epithelial cells,

macrophages, beta-cells, platelets, oligodendrocytes, and endothelial cells) (102, 370,

385-392). But studies of VAMP3 knockout mice contradict this idea (for example,

platelets from VAMP3 knockout mice have normal alpha-granule release and beta-cells

from VAMP3 knockout mice have normal insulin secretion) (387, 393, 394). VAMP3

might control endocytosis of granule components from the plasma membrane to early

endosomes, retrieving SNAREs and other proteins for further cycles of exocytosis; some

data support this pathway (for example, VAMP3 mediates recycling of integrin-

containing vesicles from plasma membrane to the trans-Golgi network in human cancer

cells)(395, 396).VAMP3 might also modulate vesicle recycling from an early endosome

compartment to the trans-Golgi network; several studies support this suggestion (for

example, VAMP3 mediates transport of the mannose 6-phosphate receptor from

endosome to Golgi)(397). Finally, VAMP3 might regulate VWF trafficking by

controlling endothelial autophagy (316, 398, 399).

Model of Endothelial Granule Release

Our data and the findings of others support a model of exocytosis specific for endothelial

cells. The G-protein Rab27a along with MyRIP and myosin Va targets granules to the

plasma membrane (269, 296, 298, 305, 306, 400).VAMP8 on the granule membrane

interacts with two t-SNAREs on the inner surface of the plasma membrane, potentially

Page 153: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

130

STX4 and SNAP23. This interaction of the three SNAREs brings the granule and plasma

membrane into apposition. However, complexins may bind to the three SNAREs,

blocking membrane fusion.(356, 401) When an agonist such as thrombin activates the

endothelial cell, intracellular calcium currents rise, synaptotagmin releases the complexin

clamp, the SNARE complex drives fusion of the granule and plasma membrane, and the

contents of the granule are released (356, 400-402) NSF then disassembles the ternary

SNARE complex, and the individual SNARE components are recycled (108, 257, 300-

304, 403).

Page 154: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

131

SNAP23 Regulates Endothelial Exocytosis

Page 155: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

132

4.1 Introduction

Among the three types of SNARE molecules that regulate endothelial exocytosis, little is

known about the precise identity of the SNAP homolog. SNAP25 is present almost

exclusively in the brain. In endothelial cells, SNAP25 is not detectable, suggesting that a

homolog of SNAP25 mediates the endothelial SNARE complex (108, 178, 295).

SNAP23, a ubiquitously-expressed homolog of SNAP25, shares 59% identity to

SNAP25. SNAP23 can regulate exocytosis in several distinct cell types. SNAP23 is

localized to the plasma membrane in adipocytes and interacts with multiple syntaxin

isoforms (syntaxin 2, 3, 4, and 5) (404). SNAP23 regulates GLUT4 translocation,

neuroendocrine cell exocytosis, and mast cell degranulation (404-408), suggesting

SNAP23 appears to fulfill the function of SNAP25 in non-neuronal tissues in forming

SNARE complex. SNAP23 has been found in human endothelial cells (102, 108).

SNAP23 interacts with Cav-1 and plays an important role in endothelial caveolae

transcytosis (110). However, studies of the role of SNAP23 in endothelial exocytosis are

limited: partial knockdown of SNAP23 led to a non-significant decrease in exocytosis

(102). Therefore, in an effort to resolve the ambiguities surrounding the function of

SNAP23, we explore the role of SNAP23 in endothelial exocytosis.

Page 156: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

133

4.2 Results

4.2.1 SNAP23 is expressed in human ECs and murine tissues

We first searched for endothelial expression of SNAP isoforms. Using microarray

hybridization techniques, we found HUVEC express RNA for SNAP23, SNAP25,

SNAP29, SNAP47, and SNAP91 (Figure 4-1A). RNA expression of these homologs

were confirmed by RT-qPCR (Figure 4-1B). Transcription of these SNAP homologs in

HUVEC was further confirmed using ENCODE RNA-Seq data (Figure 4-1C). The

SNAP homolog expressed at highest levels in endothelial cells is SNAP23 (Figure 4-1 A-

B).

We next characterized the expression of SNAP23 in human endothelial cells and murine

tissues by Western blot. We found SNAP23 is expressed in different human endothelial

cell types, including HBMEC, HAEC, and HUVEC (Figure 4-1D). SNAP23 protein is

expressed in murine lung, brain, aorta, pancreas and liver (Figure 4-1E). Our results

extend previous studies showing SNAP23 mRNA is ubiquitously expressed, although its

tissue abundance appears different in mouse and human (409, 410).

4.2.2 SNAP23 regulates endothelial exocytosis

We next explored the role of SNAP23 in endothelial exocytosis. VWF is the major

component of endothelial granules that is released by endothelial exocytosis (184). We

Page 157: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

134

knocked down the expression of endogenous SNAP23 in HDMVEC and HUVEC,

stimulated the cells to trigger exocytosis, and then measured the amount of VWF released

into the media by an ELISA. Expression of SNAP23 was significantly reduced by siRNA

(Figure 4-2A). The expression of other SNARE proteins in endothelial cells were not

affected by siRNA against SNAP23, including STX4, VAMP3, and VAMP8 (Figure

4-2A). The total intracellular VWF content was also unaffected by SNAP23 knockdown

(Figure 4-2B). We found that knockdown of SNAP23 significantly reduced VWF

exocytosis induced by physiological agonists histamine and thrombin, as well as by the

Ca2+ ionophore A23187 (Figure 4-2C). Knockdown of SNAP23 decreases exocytosis

between by approximately 29% in HDMVEC to 58% in HUVEC (Figure 4-2C). Taken

together, these results suggest SNAP23 is important for Ca2+-dependent endothelial

exocytosis.

4.2.3 SNAP23 is primarily localized on cell membrane in EC

We then studied the subcellular localization of SNAP23 in endothelial cells by confocal

microscopy. We focused on the location of SNAP23 relative to the endothelial granules

called WPBs that contain VWF as well as other pro-thrombotic and pro-inflammatory

compounds. SNAP23 is primarily localized to the plasma membrane (Figure 4-3A).

VWF is localized to the typical cigar-shaped WPB granules (Figure 4-3A). There is no

significant overlap between SNAP23 and VWF staining (Figure 4-3A).

Page 158: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

135

Most SNAP23 is localized to the plasma membrane, and a minor portion of SNAP23 is

also found in the cytoplasm, consistent with previous studies (411-413).

Immunofluorescent staining on cells cultured at sub-confluent and confluent conditions

showed the subcellular distribution of SNAP23 seems to be dependent on cell

confluence. Fully confluent cells have prominent cell membrane staining of SNAP23 and

less cytoplasmic SNAP23, whereas cells at sub-confluent condition showed more

SNAP23 in the cortical region and the cytoplasm (Figure 4-3A). To confirm this

observation, we cultured HUVEC at sub-confluent and confluent conditions and isolated

cytosol and membrane fractions, and immunoblotted fractions for SNAP23. SNAP23 is

mostly found on the membrane fraction of subconfluent cells and confluent cells (Figure

4-3B). A significant amount of SNAP23 was also detected in the cytosol at sub-

confluent conditions but less SNAP23 in the cytosol of confluent cells (Figure 4-3B).

Taken together, these data suggest SNAP23 is primarily localized on the plasma

membrane in endothelial cells, and its cytosolic distribution depends on cell confluence.

4.2.4 SNAP23 interacts with endothelial exocytic machinery

Since SNAP23 is important for endothelial exocytosis, we next searched for a link

between SNAP23 and the endothelial exocytic machinery. In order to search for the

interaction partners of SNAP23, we first performed sucrose density gradient

fractionation. We separated HUVEC lysates through a 5% - 30% - 40% discontinuous

sucrose gradient, and probed 17 fractions for SNARE proteins involved in endothelial

exocytosis. SNAP23 co-sediments with STX4 in fractions 3 to 7 and 15 to P (Figure

Page 159: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

136

4-4A). SNAP23 partially co-sediments with VAMP3 and VAMP8 (Figure 4-4A). The

sucrose density gradient fractionation provided indirect evidence that SNAP23 may

interact with endothelial SNARE molecules. To confirm their interaction in a complex,

we immunoprecipitated HUVEC lysates with antibody to SNAP23 or isotype IgG, and

then probed precipitants for SNARE proteins. STX4, VAMP3, and VAMP8 were all

detectible in the precipitant, in resting and stimulated condition (Figure 4-4B). Taken

together, these data suggest SNAP23 interacts in a complex with components of the

endothelial exocytic machinery containing STX4, VAMP3, and VAMP8, both at resting

and stimulated conditions.

Page 160: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

137

Page 161: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

138

Figure 4-1 SNAP23 is expressed in human endothelial cells and murine tissues.

(A) Heat map of the gene expression values for SNAP homologs in 6 samples of HUVEC

by microarray. Relative expression values were normalized to that of GAPDH.

(B) Relative expression of SNAP homologs in 3 individual donors of HUVEC as assayed

by RT-qPCR. Expression was normalized as percentage of SNAP23 expression.

(C) ENCODE data on the UCSC genome browser depicting the expression of HUVEC

SNAP homologs as assayed by RNA-seq.

(D) SNAP23 is expressed in HBMEC, HAEC, and HUVEC as measured by Western

blot.

(E) SNAP23 is expressed in multiple murine tissues as measured by Western blot. Data

are represented as mean ± SD. See also Table 1.

Page 162: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

139

Page 163: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

140

Figure 4-2 SNAP23 is important for endothelial exocytosis.

(A) siRNA directed against SNAP23 (siSNAP23) knocks down SNAP23 protein levels

as measured by Western blot. The siRNA against SNAP23 has no effect on the

expression of other SNARE proteins including STX4, VAMP3, and VAMP8. GAPDH

was used as loading control.

(B) SNAP23 knockdown does not affect VWF expression in HUVEC. Total VWF

content was measured by an ELISA in control siRNA (siControl) and siSNAP23 treated

cells (n = 6; NS, non-significant).

(C) SNAP23 knockdown decreases endothelial exocytosis. HDMVEC and HUVEC were

treated with siControl or siSNAP23, stimulated with serum-free medium only (resting),

or 10 μM histamine, or 1 U/ml thrombin, or 10 μM Ca2+ ionophore A23187 for 30 min;

and then VWF released into the media was measured by an ELISA (n = 4 – 7; * P < 0.05

vs. siControl; NS, non-significant vs. siControl). Data are represented as mean ± SD.

Page 164: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

141

Page 165: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

142

Figure 4-3 Subcellular localization of SNAP23 in endothelial cells.

(A) Subcellular localization of SNAP23 in sub-confluent (upper panel) and confluent

(lower panel) HUVEC. Immunofluorescent staining was performed on HUVEC with

antibodies against SNAP23 (red) and VWF (green), DNA was stained with DAPI (blue),

and the cells were imaged by confocal microscopy (objective 60× oil, scale bar = 50 μm,

confocal z resolution = 0.32 μm).

(B) Western blot analysis of cell fractions from sub-confluent and confluent HUVEC

using markers for membrane (Caveolin-1) and cytosol (GAPDH). These fractions were

also probed for SNAP23. Whole cell lysates were used for total protein. SNAP23 is

depleted from cytosol fraction when reaching confluence.

Page 166: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

143

Page 167: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

144

Figure 4-4 SNAP23 interacts with endothelial exocytic machinery.

(A) SNAP23 co-sediments with STX4, VAMP3 and VAMP8 by sucrose density gradient

fractionation. HUVEC lysate was ultracentrifuged through a 5% - 40% discontinuous

sucrose gradient, and then the gradient was aliquoted into 17 fractions and analyzed by

SDS-PAGE (T, total proteins in the lysate; P, pellet after fractionation). β-actin was used

as control for fraction separation. Representative of 3 separate experiments.

(B) SNAP23 co-precipitates with STX4, VAMP3 and VAMP8. HUVEC stimulated with

serum-free medium only (Rest), or 10 μM histamine, or 10 μM Ca2+ ionophore A23187

for 30 min were immunoprecipitated with antibody to SNAP23 or isotype IgG. The

precipitants were probed with antibody to STX4, VAMP3 and VAMP8. Input represents

5% total protein. Representative of 3 similar experiments.

Page 168: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

145

4.3 Discussion

The identity of the SNAP homolog which functions as an endothelial t-SNARE is

unclear. In this study, we found that SNAP23 is the most highly expressed SNAP isoform

in different types of human endothelial cells; SNAP23 is localized to the endothelial cell

membrane; SNAP23 forms complexes with other endothelial SNARE molecules; and

most importantly, SNAP23 deficiency impairs endothelial exocytosis. These results

collectively suggest that SNAP23 plays a critical role in regulating endothelial cell

exocytosis.

Our work and the studies of others show that human endothelial cells express a

distinctive subset of SNARE molecules (102, 108, 178) Endothelial cells express

VAMP3 and VAMP8 of the v-SNARE family, STX4 of the syntaxin family, and several

SNAP isoforms including SNAP23. These results demonstrate that endothelial cells

contain family members of exocytic machinery also found in neurons and yeast. We also

found that a subset of endothelial SNAREs interact with each other: endothelial cells

contain SNARE complexes consisting of SNAP23, STX4, and VAMP3 or VAMP8

(Figure 4-4). This SNARE complex corresponds to SNARE complexes found in

neurons, composed of SNAP25, STX1a, and VAMP2 (133, 414). We also found that

SNAP23 co-sediments with STX4 and VAMP3 and VAMP8 (Figure 4-4A). Our work

supports the studies of others that show SNAP23 and STX4 form clusters in endothelial

cells, and the results of others showing SNAP25 and STX4 form clusters in

neuroendocrine cells (415, 416).

Page 169: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

146

In resting cells, SNAP23 interacts with STX4, VAMP3, and VAMP8 (Figure 4-4).

Stimulation of endothelial cells with histamine or calcium ionophore increases the

interaction of SNAP23 with STX4, VAMP3, and VAMP8 (Figure 4-4B). These data

support the idea that SNAP23 functions as one component of the ternary SNARE

complex in endothelial cells. Our work partially contrasts with the work of others (102),

who show that SNAP23 is localized to plasma membrane but has little effect on

endothelial exocytosis. One possible explanation for this discrepancy is that knockdown

of SNAP23 expression was incomplete in other studies (102).

Among the members of the SNAP family, only two isoforms have been regarded as

critical components in exocytosis, SNAP25 and SNAP23. SNAP25 is expressed in

neuronal and neuroendocrine tissues, whereas SNAP23 is ubiquitously expressed in non-

neuronal cells (409). Endothelial cells express lower levels of other SNAP isoforms

(Figure 4-1A-C).

We show that SNAP23 is localized to endothelial plasma membranes (Figure 4-3). Our

data suggest that SNAP23 plays a role similar to SNAP25 in neurons, serving as a t-

SNARE. However, a minor fraction of SNAP23 was also found in the cytosol (Figure

4-3), similar to previous studies (110). It has been previously confirmed that the plasma

membrane localization of SNAP family proteins depends on the palmitoylation of a

cysteine-rich domain (417). It is plausible that a sub-fraction of SNAP23 proteins is

detectable in the cytosol before palmitoylation occurs. Cytosolic levels of SNAP23 are

decreased in confluent cells (Figure 4-3B), suggesting that palmitoylation of SNAP23

Page 170: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

147

may be dynamically regulated and is not constitutive. In addition, SNAP homologs were

found in the cytoplasm as intracellular t-SNARE mediating intracellular membrane

fusion, such a possibility for SNAP23 cannot be ruled out (418).

Our data show SNAP23 is expressed in multiple murine tissues, confirming previous

observations (409, 410, 419). We and others found SNAP23 is most highly expressed in

the murine lung (Fig. 1E) (419). Why is SNAP23 highly expressed in the lung? One

explanation is SNAP23 is important for epithelial cell or pneumocyte exocytosis. Prior

studies have shown that regulated mucin secretion from airway epithelial cells requires

SNAP23 (420). Syntaxin 2 and SNAP23 are important for regulated surfactant secretion

from alveolar type II cells (421). SNAP23 is enriched in lipid rafts in type II alveolar

cells which serves as a functional platform for surfactant secretion (422). SNAP23 also

interacts with annexin A7 in a protein kinase C (PKC)-dependent manner which

facilitates membrane fusion during surfactant secretion (423). Another possibility is

SNAP23 is involved in the endothelial exocytosis of the lung vasculature. Still another

possibility is SNAP23 is important for the response of immune cells in the lung. SNAP23

is important for phagosome formation and maturation in macrophages (424), mast cell

degranulation (363, 405), neutrophil exocytosis (425), secretion of antibodies by human

plasma cells (426), and is found on the plasma membrane of several different human

inflammatory cells including eosinophils, basophils, neutrophils, and peripheral blood

mononuclear cells (427, 428). Interestingly, SNAP23 can be induced by various cytokines

as an immediate-early cytokine responsive gene in a murine myelomonocytic cell line.

Thus SNAP23 might be expressed by one or more cell types in the lung.

Page 171: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

148

In summary, in Chapter 4, we have shown that human endothelial cells express SNAP23,

SNAP23 interacts with other endothelial SNARE, and SNAP23 plays a crucial role in

endothelial exocytosis of VWF. This matches a model in which a three membered

SNARE complex forms before endothelial exocytosis, consisting of a VAMP on the

membrane of endothelial granules, along with STX4 and SNAP23 on the plasma

membrane (133, 361, 414).

Page 172: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

149

Conclusions and Perspectives

Page 173: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

150

5.1 Summary

EC mediates vascular injury response by exocytosis. WPBs are EC-specific granules for

exocytosis. EC exocytosis release VWF, a major initiator in thrombogenesis, and P-

selectin, which mediates vascular inflammation.

STXBP5 is a novel candidate gene linked to changes in plasma vWF levels by GWAS. In

Chapter 2, we found STXBP5 is expressed in ECs and interacts with SNARE proteins.

STXBP5 inhibits endothelial exocytosis in vitro and in vivo. However, despite higher

plasma VWF levels, Stxbp5 KO mice displayed impaired hemostasis and thrombosis, and

Stxbp5 KO platelets had severe defects in secretion and activation. These results suggest

STXBP5 inhibits endothelial exocytosis, but promotes platelet secretion and thrombosis.

STXBP5 is a novel SNARE regulatory partner with dual regulatory functions.

Human ECs express SNARE proteins, including STX, VAMP and SNAP homologs. But

the identities of endothelial SNAREs have been unclear. In Chapter 3, we found VAMP8,

in addition to VAMP3, plays a critical role in endothelial exocytosis. VAMP8 co-

localized with WPBs. VAMP8 interacted with other EC SNAREs. Knock down of

VAMP8 or VAMP3 expression inhibited endothelial exocytosis. More importantly,

Vamp8 KO mice had decreased endothelial exocytosis and hemostasis.

In Chapter 4, we found SNAP23 is another key component of the endothelial SNARE

machinery. We identified SNAP23 as the most abundant endothelial SNAP homolog that

Page 174: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

151

is important for endothelial exocytosis. SNAP23 was found on endothelial cell

membrane. Knockdown of SNAP23 decreased endothelial exocytosis. SNAP23 also

interacted with other important endothelial SNARE proteins.

In summary, the present work investigated the role of STXBP5, VAMP8, and SNAP23 in

endothelial exocytosis, provided functional relevance for a novel genetic risk factor

identified by GWAS, and improved our understanding on the regulatory mechanism of

endothelial exocytosis.

Page 175: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

152

5.2 Working model for endothelial exocytosis

Our current work identified a novel regulator of endothelial exocytosis, STXBP5, and

explored the functional implication of its genetic variants on plasma VWF levels and

thrombosis. Our work also identified important endothelial SNARE members and

characterized their role in endothelial exocytosis and thrombosis. These findings

provided a novel mechanistic view of the endothelial cell exocytic machinery.

We propose a working model that integrates the major elements examined by this project

(Scheme 5-1)

In this simplified model, the trafficking and fusion of WPBs is mediated by at least four

components. Chapter 3 and 4 have suggested that the components of the exocytic

machinery include VAMP8 or VAMP3 as the v-SNARE, and STX4 and SNAP23 as the

t-SNAREs. Cognate SNAREs interact to form a trans-SNARE complex. Formation of

this trans-SNARE complex tethers WPB to the endothelial cell membrane. Presumably,

intracellular Ca2+ elevation activates the Ca2+ sensor SYT1, which triggers the trans-

SNARE complex to change its conformation into a cis-SNARE complex, resulting in

membrane fusion and VWF release (Scheme 5-1A).

Chapter 2 identified a novel regulator of exocytosis, STXBP5. We propose that STXBP5

acts as a brake on the endothelial exocytic machinery through two potential mechanisms

(Scheme 5-1B-C):

Page 176: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

153

1) STXBP5 may interact with STX4, sequestering STX4 from the other SNAREs,

blocking formation of the trans-SNARE complex (Scheme 5-1B, marked as “1”).

2) STXBP5 may interact with the Ca2+ sensor SYT1, blunt Ca2+ signaling, resulting in

less Ca2+ sensitive vesicles and less VWF release (Scheme 5-1C, marked as “2”).

Either or both of these mechanisms are possible. Each of these steps will limit vesicle

fusion with the cell membrane and reduce VWF exocytosis.

Page 177: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

154

Page 178: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

155

Scheme 5-1 Working model of endothelial exocytosis regulation.

(A) The endothelial exocytic machinery is presumably composed of t-SNAREs and v-

SNARE. In this case, VAMP8 represents the v-SNARE whereas STX4 and SNAP23

represents the t-SNAREs. WPB docks to the cell membrane as a result of trans-SNARE

complexes formation. SYT1 triggers trans-SNARE “zippering” in response to

intracellular Ca2+, causing vesicle fusion and VWF exocytosis.

(B) STXBP5 inhibits endothelial exocytosis by interacting with STX4. STXBP5 binds to

STX4 and sequesters it, so that less t-SNARE is available for productive trans-SNARE

complex formation.

(C) STXBP5 inhibits endothelial exocytosis by interacting with SYT1. Occupancy of

SYT1 by STXBP5 blocks its Ca2+ sensing. The effect of Ca2+ to bind to SYT1 and trigger

exocytosis is therefore dampened.

Page 179: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

156

5.3 Future directions

Although our data demonstrated the importance of several novel regulators of endothelial

exocytosis, we have only begun to understand the intricate regulatory mechanisms in

endothelial cells. Many questions about the regulation of endothelial exocytosis remain to

be answered.

5.3.1 STXBP5 SNP and VWF

The most fascinating question is how genetic variation in STXBP5 affects STXBP5

function and plasma VWF levels.

The SNP with the highest genome-wide significance level for plasma VWF in the meta-

analysis of the CHARGE Consortium data is rs9390459, which is a synonymous

variation (317). The top non-synonymous variation, rs1039084 (encoding STXBP5-

N436S), is in high pairwise LD with rs9390459 (338). Is the top candidate rs9390459 a

proxy variation for rs1039084? Our data seem to support this prediction, because

although rs9390459 is synonymous, the STXBP5-N436S variant does seem to inhibit

VWF exocytosis more potently than wild-type STXBP5 (in this case, both the major and

minor allele of rs9390459 encode the same amino acid). Although the technical difficulty

in platelet transgenic studies prevented us from observing a phenotype in platelets using

the STXBP5-N436S mutant, our data (Figure 2-9) is consistent with the clinical data

showing the minor allele G of rs1039084 (corresponding to 436 serine) is associated with

Page 180: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

157

lower plasma VWF, decreased DVT risk, and higher bleeding score (319, 339). This

consistency strongly suggests genetic variation in STXBP5 is a novel risk factor for

plasma VWF level and thrombotic diseases, and warrants further mechanistic

investigation.

The asparagine to serine substitution at STXBP5 436 position may change its interaction

partners or subject it to additional post-translational modification. It has been shown that

STXBP5 is phosphorylated by PKA at serine-724 (located in the hyper-variable linker

region between the N-terminal WD40 repeats and the C-terminal VAMP-like domain),

which reduces its interaction with STX1 and increases SNARE complex formation (336).

The N436S substitution is located in one of the WD40 repeat domains. It is unknown

whether this position is involved in STXBP5 interaction with other proteins or is a

potential target for post-translational modification such as phosphorylation.

One approach to answer this question is to use genome editing techniques in cells and in

mice. The rapid developing field of genome-editing technology has provided

investigators precise and high-efficient tools, such as zinc finger nucleases (ZFNs),

transcription activator-like effector nucleases (TALENs), and clustered regularly

interspaced short palindromic repeats (CRISPR)/CRISPR-associated 9 (Cas9) (429, 430).

We have started to use the CRISPR/Cas9 system to introduce a single nucleotide variant

(SNV) into the genomic DNA encoding STXBP5 in human EC to see if changing one

nucleotide, such as rs1039084 (A>G), is able to produce a phenotypic change.

Considering human and mouse STXBP5 share 98% identity in protein sequence, we have

Page 181: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

158

also successfully generated mice containing the human SNP rs1039084 (Stxbp5-N437S

mice) using the CRISPR/Cas9 system (431, 432). We propose to characterize the

phenotype of endothelial exocytosis, platelet function, hemostasis and thrombosis in

these cell and mouse models. Moreover, the novel gene editing technology has also

enabled us to elucidate the potential effects of other genetic variants in STXBP5 as well

as in many other genetic loci.

5.3.2 STXBP5 in ECs and in platelets

Another intriguing question is how STXBP5 can display a dual positive and negative role

in exocytosis.

We initially hypothesized STXBP5 might produce different splice variants in ECs and

platelets, but qPCR shows the splice variant profiles were similar in ECs and platelets

(Figure 2-1D).

Another possible explanation is STXBP5 may have different interaction partners in ECs

and platelets. Ye et al. shows that in platelets STXBP5 interacts with STX11 and

SNAP23, and to a lesser extent with STX2 and STX4; STXBP5 also interacts with the

platelet cytoskeleton (433). Our preliminary co-immunoprecipitation experiment shows,

however, in HUVEC the interaction between STXBP5 and STX11 or SNAP23 were not

detectible (data not shown). It has been shown that STX11 is required for platelet

secretion (367), so one would expect that binding of STXBP5 may presumably block

Page 182: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

159

productive SNARE complex formation. However, it is conceivable that platelet syntaxin

homologs may display different binding affinities or kinetics for STXBP5, so that their

interaction does not lead to “dead-end” SNARE complex formation, but instead

facilitates the assembly of SNARE complexes. For example, STXBP5 perhaps binds to t-

SNAREs, ensure their proper localization, or orchestrates the arrangements of SNARE-

complex (with or without participation of platelet cytoskeleton). Since STXBP5 interacts

with both syntaxins and SNAP23 in platelets, it is also possible that STXBP5 may

enhance the efficiency of syntaxin-SNAP23 or SNARE-Munc18 complex clustering

(415, 416), which in neurons facilitate SNARE-mediated vesicle fusion.

An interesting observation is that the paradoxical regulation by STXBP5 in exocytosis

closely resembles that of complexin. In neurons, complexin facilitates Ca2+-triggered fast,

synchronized synaptic release, but inhibits spontaneous fusion. Unfortunately, although

several models for the role of complexin have been proposed, none has been

experimentally tested (434). One would speculate that STXBP5 and complexin to share

some common properties. In fact, these two protein do have quite a number of common

features. They are both conserved from yeast to human. They both carry a coiled α-helix

that binds to syntaxin (complexin helix inserts into the groove between VAMP and

syntaxin helices on trans-SNARE complex; STXBP5 binds to syntaxin and displaces

VAMP with its VAMP-like domain). They both interact with synaptotagmin 1 and

regulate Ca2+-triggered exocytosis.

Page 183: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

160

Unlike STXBP5, the crystal structure of complexin has been well documented (148). The

crystal structure shows complexin stabilizes the fully assembled SNARE complex, which

is essential for Ca2+-triggered fast, synchronized exocytosis. Complexin regulation of

fusion depends on its concentration (435). But how its concentration is controlled

remains unknown. Could STXBP5 behave just like complexin, where its concentration

dictates its inhibition or promotion of exocytosis? Or does it compete with complexin in

binding SNAREs and/or synaptotagmin? Given the significant similarities between

complexin and STXBP5, the above two possibilities are not mutually exclusive.

Note complexin only binds to assembled SNARE complex which is on the cell

membrane; in contrast STXBP5 is primarily located in the cytoplasm (Figure 2-10),

although a subset of STXBP5 is also found on the cell membrane via interaction with

syntaxin (Figure 2-11C) (350). Like other nonintegral membrane proteins, it is postulated

that STXBP5 is in equilibrium between cytosolic and membrane pools in certain cell

types. The synaptic protein unbinding rate of STXBP5 (0.139/s) (350) is slower than the

rate of complexin's off rate from the SNARE complex (0.3/s) (436, 437), suggesting a

higher affinity of STXBP5 with SNARE proteins. Could it be that STXBP5 acts as a

cytoplasmic (and to a lesser extent membranous) “buffer” for un-docked or recycled

SNARE protein or synaptotagmin, whereas complexin binds SNAREs more weakly and

only on the cell membrane, and it is the stochastic ratio of their relative abundance that

determines the vesicle fusion probability? Perhaps further studies on the STXBP5 domain

structure and functions, including its crystal structure, would eventually help answer

these questions.

Page 184: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

161

5.4 Significance and perspectives

Nearly a century has passed since Finnish physician Erik von Willebrand first described

the VWD case. Today our knowledge on the pathogenesis of hemorrhagic and thrombotic

disorders has greatly improved, but our diagnosis and therapy is still far from adequate

(238, 438).

VTE is a multifactorial disorder. However, a substantial proportion of VTE patients

present with no detectable acquired risk factors of thrombosis, suggesting that genetic

factors might be responsible for certain group of individuals. GWAS have revolutionized

the discovery of genomic factors associated with complex disease traits. Our study on the

role of STXBP5 served as a promising case which a new potentially targetable genetic

risk factor for VTE is identified by GWAS and is further validated by cell and animal

models. Our study on the role of VAMP8 and SNAP23, in addition, further

complemented the framework of the endothelial exocytosis machinery and how it can

affect hemostasis and thrombosis.

GWAS-based findings will continue to accelerate the discovery of translatable clinical

predictive or prognostic targets. Novel genome-editing techniques, such as

CRISPR/Cas9, offered remarkable technical possibilities to mine the mass of GWAS data

for additional trait associations, and to model and characterize genetic risk factors for

thromboembolic diseases. While the road to apply these techniques to treating diseases is

Page 185: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

162

long, our current work opened the door to further important research that will usher in

safer and more effective therapies for both bleeding and thrombotic diseases.

Page 186: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

163

Materials and Methods

Page 187: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

164

Antibodies and reagents. Primary and secondary antibodies used for this study are listed

in Table 2. Lipofectin reagent and Opti-MEM I media were purchased from Invitrogen.

Protease Inhibitor Cocktail (Complete Mini EDTA-free) was purchased from Roche.

Calcium ionophore A23187, FeCl3, histamine, thrombin, epinephrine and ATP were

purchased from Sigma-Aldrich. Thrombin was purchased from Enzyme Research

Laboratories. The cDNA for VAMP8 was cloned into a pCMV-Myc vector (Clontech).

Individual siRNA was from Ambion (Life Technologies). The sequence for siSTXBP5 is

5’-AGTGGGAACTCAGACTGGTGCTTTA-3’. The sequence for siVAMP3 is: 5’-

GCCACTGGCAGTAATCGAAGA-3’. The sequence for siVAMP8 is: 5'-

GAGGAAAUGAUCGUGUGCGGAACCU-3'. The siRNA sequence for siSNAP23 is

5’-GACACCAACAGAGAUCGUAUUGAUA-3’.

Cell culture and transfection. HUVEC, HAEC, HDMVEC, and endothelial cell culture

medium containing endothelial cell growth supplement (VascuLife EnGS) were obtained

from Lifeline Cell Technology. Endothelial cells were maintained collagen I coated

plates as described (439), and passages 3-6 were used for all in vitro studies. HL-60

promyelocytic cell line was purchased from American Type Culture Collection. Human

platelets were isolated from whole blood of healthy donors free of non-steroidal anti-

inflammatory drugs for at least 10 days as previously described (301). Transfection was

performed at 80-90% confluence using Lipofectin reagent following the manufacturer’s

protocol, with either 0.5 μg/ml plasmid DNA or 20 nM siRNA oligonucleotides for 5

hours. Cells were stimulated or imaged at least 72 h after transfection. Cell confluence

Page 188: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

165

was visually determined when cells were in contact and the entire culture surface had no

visible space among individual cells for at least 48 h.

VWF release assay. Confluent HUVEC, HAEC, or HDMVEC were stimulated with 10

μM histamine, or 10 μM A23187, or 10 μM ATP, or sham treatment (PBS) for 30 min at

37°C to stimulate VWF release. After stimulation of the cells, VWF concentration in the

cell media was quantified by a VWF ELISA kit (American Diagnostica Inc. and Sekisui

Diagnostics, LLC). In separated experiments, HUVEC transfected with control- or

STXBP5-siRNA were lysed without stimulation and intracellular VWF contents were

measured in equal amount of protein lysate. We found the antibody to VWF in this

ELISA cross-reacts with mouse VWF antigen. Separate measurements were performed in

diluted mouse plasma collected by retro-orbital bleeding from Stxbp5 WT mice and

Stxbp5 KO littermates.

Endothelial cell-leukocyte interaction. Adhesion of leukocytes to HUVEC was performed

as described (308, 314, 315). HUVEC were transfected with control siRNA or siRNA

against STXBP5 in a 12-well plate and were then cultured until 100% confluent. Prior to

the adhesion assay, HL-60 cells were labeled with 2 μM Calcein AM (Molecular Probes)

for 30 min at 37°C, washed, and suspended in serum-free media. Then 1×105 HL-60 in

500 μl serum-free media was added to each well in the presence or absence of 10 μM

histamine. The plate was kept at 4°C for 15 min, washed twice with HBSS, refilled with

media, and immediately imaged with an Olympus BX51 upright microscope with a 10×

Page 189: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

166

water immersion objective. Cells pre-incubated with an azide-free antibody raised against

full-length P-selectin (AK4) was used as negative control.

ADAMTS13 assay. We used an assay to measure ADAMTS13 in murine plasma (440).

Dr. Neelamegham (Department of Chemical and Biological Engineering, State

University of New York at Buffalo, Buffalo, NY 14260, USA) generously provided a

recombinant polypeptide, XS-VWF, which contains a truncated fragment of the VWF-A2

domain cleavable by ADAMTS13. This polypeptide is flanked by a CFP variant

Cerulean and an YFP variant Venus that exhibit fluorescence/Förster resonance energy

transfer (FRET) properties. ADAMTS13 cleavage of XS-VWF keeps the fluophores

apart, resulting in an increase of Cerulean emission and a decrease of FRET efficiency.

ADAMTS13 activity is quantified by the FRET ratio, defined as the ratio of Cerulean

emission versus Venus emission at 420 nm. We found mouse plasma efficiently cleaves

XS-VWF. In this assay, 15 μl WT or Stxbp5 KO mouse plasma were incubated with 10 μl

XS-VWF substrate in 100 μl cleavage buffer (50 mM Tris, pH 8.0 and 12.5 mM CaCl2)

in a 96-well plate for 1 h at 37°C. The FRET ratio was measured on a plate reader and

ADAMTS13 activity was calculated as percentage of pooled WT mouse plasma.

Western blotting. Western blotting was performed as described previously (108). In brief,

endothelial cells or platelets were lysed with Laemmli sample buffer (Bio-Rad), boiled

for 5 min at 95°C, resolved on 4-20% Mini-PROTEAN TGX Precast gels (Bio-Rad), and

transferred using a Trans-Blot SD semi-dry electrophoretic transfer unit (Bio-Rad) onto

nitrocellulose membranes. After 1-hour blocking with 5% non-fat milk in PBS containing

Page 190: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

167

0.05% Tween 20 at room temperature, the membranes were hybridized with primary

antibodies followed by HRP-conjugated secondary antibodies and enhanced

chemiluminescence detection using X-ray films.

Quantitative real-time PCR. Murine tissue was harvested immediately after transcardial

perfusion with diethylpyrocarbonate-treated PBS. Total RNA was isolated with Trizol

following the manufacture’s protocol (Invitrogen) and purified with lithium chloride

(Sigma-Aldrich). The A260/A280 ratio of all samples were between1.9-2.1 as measured

by spectrophotometry (NanoDrop; Thermo Scientific). cDNA was synthesized using an

iScript™ cDNA Synthesis kit (Bio-Rad). Quantitative real-time PCR was performed by

Taqman gene expression assay (Applied Biosystems) for 40 cycles on an iCycler thermal

cycler equipped with MyiQ PCR detection system (Bio-Rad). Three independent

experiments were performed for each organ, and in each experiment TaqMan

quantification was repeated in triplicate for each sample. Taqman probes were purchased

from Applied Biosystems. For each probe set, calibration curves were generated by 10-

fold serial dilutions of cDNA to ensure comparable PCR efficiency. Expression results

were calculated by ΔΔCT method and were normalized to the reference genes GAPDH or

Gapdh. SNAP homolog expression was quantified by ΔΔCt method using four reference

genes: B2M, GAPDH, HRPT1, and YWHAZ, and expressed as percentage relative to the

amount of SNAP23.

Identification of STXBP5 mRNA splice variants. Primers flanking the splice region of

human STXBP5 mRNAs were designed (5’-TCGCTGCAAATCTCCAACCT-3’ and 5’-

Page 191: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

168

GGACGAGTCCGTCTTTCGAG-3’). HUVEC, human platelets, and HEK293 cDNA

were synthesized as described above. Human adult brain cDNA was purchased from

Biochain. PCR was performed using 0.25 μg cDNA per reaction with the above primers.

The products were separated by agarose gel, sequenced, and compared with validated and

predicted NCBI Reference Sequences. Five NCBI sequences were found matching the

sequencing results: NM_001127715.2, NM_139244.4, XR_245502.1, XR_245503.1, and

XR_245504.1. The quantification of each splice variant were performed using custom

Taqman probes, each spans exon boundaries specific for one splice variant

(NM_001127715.2, exons 21-23; NM_139244.4, exons 19-23; XR_245502.1, exons 21-

22; XR_245503.1, exons 20-22; XR_245504.1, exons 19-22).

Site-directed mutagenesis. Mouse pMEX neo-3×FLAG-tomosyn (8)-ATG expression

plasmid was a kind gift from Dr. David James (Garvan Institute of Medical Research,

Darlinghurst, NSW 2010, Australia) (354). A point mutation corresponding to human

SNP rs1039084 (A→G) was introduced by site-directed mutagenesis with a QuikChange

Mutagenesis Kit (Agilent Technologies). Using the plasmid as template, PCR was

performed with the following primers for mutagenesis: 5’-

AAAGGAATGGCCCATCAGCGGAGGTAATTGGGGCTT-3’ and 5’-

AAGCCCCAATTACCTCCGCTGATGGGCCATTCCTTT-3’. PCR products were

treated by DpnI to digest template plasmid and then transformed into competent cells.

The mutated plasmid was extracted using EndoFree Plasmid Maxi Kit (Qiagen) and

mutation confirmed by DNA sequencing.

Page 192: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

169

Co-immunoprecipitation. HUVEC was lysed with coimmunoprecipitation buffer

containing 10 mM HEPES/NaOH, pH 7.4, 140 mM NaCl, 5 mM NaHCO3, 1.2 mM

NaH2PO4, 1 mM MgCl2, 10 mM glucose, 5 mM KCl, 1 mM EGTA, 1.3 mM CaCl2, and

1% (w/v) Triton X-100 supplemented with protease inhibitors cocktails on ice and

incubated for 60 min followed by centrifugation at 161,000 × g at 4 °C for 20 min. 25 μl

Protein A/G PLUS-Agarose was incubated with 2 μg antibodies or mouse IgG1 at 4 °C

overnight, washed, and then mixed with the pre-cleared cell lysate at 4 °C overnight. The

precipitants were washed with cold co-immunoprecipitation buffer 6 times and the bound

proteins were then solubilized in reducing Laemmli sample buffer at 95 °C. The elute

was resolved by SDS-PAGE followed by immunoblotting.

Confocal microscopy. HUVEC was cultured on collagen-coated 35-mm glass dishes

(MatTek). After removal of media, cells were rinsed twice with PBS and fixed

immediately with 4% PFA for 20 min. After permeabilization with 0.15% Triton X-100

for 10 min and blocking with 5% donkey serum for 1 hour at room temperature, the cells

were incubated with primary antibodies (1:100-1:330 dilution) overnight at 4 °C followed

by 1-hour incubation at room temperature with 1:2000 fluorescent secondary antibodies

and DAPI. Fluorescent microscopy was performed using an IX81 inverted confocal

microscope equipped with a high sensitivity digital camera (FV1000, Olympus). All

fluorescent images were generated using sequential line-scanning with identical settings

in each experiment at maximum z-resolution (0.40 μm for 40× objective, 0.32 μm for 60×

objective). Image analysis was carried out using FV10-ASW 3.0 (Olympus) and Image-

Pro Plus (Media Cybernetics).

Page 193: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

170

Sucrose density gradient ultracentrifugation. HUVEC were lysed on ice with 2 ml co-

immunoprecipitation buffer supplemented with protease inhibitors cocktails and

incubated for 1 hour at 4 °C followed by centrifugation at 161,000 × g for 20 min. The

supernatant was carefully loaded on top of a discontinuous sucrose density gradient of 1

ml 5% sucrose, 6 ml 30% sucrose, and 3 ml 40% sucrose in coimmunoprecipitation

buffer (from top to bottom) in a 14 ml PET thin walled tube (Thermo Scientific). The

sucrose density gradient was then ultracentrifuged at 166,880 × g for 20 hours at 4°C on a

Discovery 100s ultracentrifugation equipped with a SureSpin 630/17 swinging-bucket

rotor (Sorvall). After ultracentrifugation, 18 equal-volume aliquots were carefully

aspirated from top to bottom, boiled in equal-volume 2× sample loading buffer, and

analyzed by 7.5% SDS-PAGE.

Microarray. Total RNA was extracted from HUVEC from six different donors by

RNeasy mini kit (Qiagen) according to the manufacturer’s protocol. The gene expression

profiling was performed using Affymetrix GeneChip at the Genomics Research Center at

University of Rochester.

Transcriptional profile by ENCODE. The Feb 2009 GRCh37/hg19 Assembly was

searched for transcription levels of SNAP homologs. Transcriptional profiles were

visualized in UCSC Genome Browser with a customized ENCODE track for HUVEC.

Stxbp5 mice management. Stxbp5 KO mice on a background of 129Sv: C57BL6: DBA/2

= 2: 1: 1 were generated as previously described (332). PCR was used for genotyping

Page 194: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

171

with primers specific for a wild-type allele (5’-TTCTGCTCCCCGCTGCTCCTT-3’ and

5’-TCCCCGCTCCCTTCACCTTGC-3’) and a mutant allele (5’-

GGGCGCCCGGTTCTTTTTGTC-3’ and 5’-GCCATGATGGATACTTTCTCG-3’). The

PCR products for wild-type allele and mutant allele were 300 bp and 224 bp,

respectively. We used 4- to 8-week-old male Stxbp5 KO mice and Stxbp5 WT littermates

for all in vivo experiments except otherwise specified.

Platelet Isolation and activation. Murine blood was obtained by retro-orbital bleeding of

anesthetized animal into heparinized murine Tyrode's buffer (134 mM NaCl, 2.9 mM

KCl, 12 mM NaHCO3, 0.34 mM Na2HPO4, 20 mM HEPES, pH 7.0, 5 mM glucose,

0.35% bovine serum albumin) in Eppendorf tubes. The blood was then centrifuged to

yield platelet rich plasma which was then washed in new tubes containing Tyrode's buffer

with 1% PGE2 to prevent platelet activation. Platelets were then pelleted by 5 min 600 ×

g centrifugation at room temperature and the supernatant was discarded. The pelleted

platelets were gently resuspended in Tyrode's buffer and kept at room temperature for

further experiments within 2 hours. For platelet activation, diluted platelet suspension

was divided into 100 μl aliquots (3 per agonist per mouse), stimulated with 10 μl PBS or

thrombin for 10 min followed by staining with 2 μl CD62P-FITC and 4 μl JON/A-PE

antibody for 15 min, and immediately fixed with 100 ul 2% formalin. The fluorescence

intensity was measured on an Accuri C6 Flow Cytometer (BD Biosciences). The data

were analyzed with FlowJo software (Tree Star Inc.), using single-stained non-activated

WT platelets for fluorescence compensation. ATP release was measured in post-

stimulation supernatant using an ATP Bioluminescent Assay Kit (Sigma-Aldrich).

Page 195: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

172

Platelet adherence assay. Mice were anesthetized with a cocktail of ketamine/xylazine

(80/12 mg/kg) and saline (10 ml/kg) via an i.m. injection. Anesthetized mice were

injected with a DyLight488-antibody to GPIb beta subunit to label platelets without

apparent interference to platelet-EC binding. For platelet adherence assay, the mesentery

was exteriorized on a petri dish and then placed on an inverted fluorescent intravital

microscope (Nikon, ECLIPSE Ti) with a 37°C stage warmer. An area containing target

venule (120-150 µm in diameter) was selected by measuring the vessel diameter using

the measuring tool equipped on the microscope software. After 120s baseline recording,

the mesenteric venules were superfused with 10 µM Ca2+ ionophore A23187 to activate

endothelial cells and to induce platelet rolling. Images of platelets rolling in the

mesenteric venules were continuously recorded for a total of 10 min starting at the

baseline with an electron-multiplying CCD video camera (QUANTEM, 512SC) at the

highest frame rate (approximately 31 frames per second). Using Image-Pro Plus software,

platelet adherence was determined by counting the number of platelets that remained

static for at least two consecutive images, including captured-and-go platelets. For each

mouse, we quantified platelet rolling in five equally-distributed video segments (one

during baseline and four during stimulation), each 500 frames in length, and used the

average number of rolling platelets per unit time per unit area (512 pixel × 100 pixel) for

quantification.

In vivo P-selectin exocytosis assay. Yellow-green fluorescent 1 μm microspheres

(Invitrogen) were coupled to anti-mouse CD62P antibody. Anesthetized mice were

infused with 108 microspheres intravenously, and the mesentery externalized as

Page 196: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

173

described. Intravital microscopy was used to visualize fluorescent microspheres binding

to minimally stimulated (by surgical preparation per se) venules for 2 min followed by 8

min of stimulation by Ca2+ ionophore A23187. For each mouse, P-selectin exocytosis

was determined by counting the number of adherent microspheres on each frame of a

100-frame cropped video and averaged over resting and stimulation condition,

respectively. The adherent number of microspheres per unit area (512 pixel × 100 pixel)

was used for quantification.

Mouse tail bleeding assay. Mouse tail bleeding time was measured as described (441).

After i.p. anesthetization with ketamine and xylazine (80/12 mg/kg), the distal 5mm of

the tails of the mice were amputated and immersed immediately in 37°C saline, and the

time to visual cessation of bleeding for 30 s or continuous bleeding to 20 mins maximal

duration, whichever occurs first, was recorded.

Mouse mesenteric and carotid thrombosis model. These models were performed as

described (442, 443). For the mesenteric thrombosis model, mice were anesthetized and

platelets labeled with DyLight488-antibody to GPIb beta. A target area containing

mesenteric arterioles (120-150 µm in diameter) was externalized for imaging. The

arteriole flow was recorded for 3 min at resting condition. Then 1 mm2 of Whatman

paper saturated with 7.5% FeCl3 solution was applied to the arteriole for 3 min and the

arteriole flow was continuously recorded for a total of 30 min. The time to form a small

thrombus (50-pixel diameter) and to full vessel occlusion were recorded. Recording was

terminated at the end of 30 min if no occlusion were observed. For the carotid thrombosis

Page 197: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

174

model, mice were sedated with 2.5% isoflurane and maintained anesthetized with 2%

isoflurane. The common carotid arteries were exposed for a baseline flow recording using

an MA1PRB Perivascular Flowprobe and a TS420 Flowmeter (Transonic Systems). Then

1×2 mm Whatmann paper soaked with 1.5 µl 7.5% FeCl3 solution was applied to the

ventral surface of the carotid upstream of the flowprobe for 3 min. Flow measurement

was resumed for a total of 30 min after FeCl3 wash-off. We define occlusion as the

absence of blood flow (0 ml/min) for 3 min.

Bone marrow transplantation. WT and Stxbp5 KO marrow donor mice were euthanized,

and femurs were isolated under sterile conditions. Bone marrow was harvested and then

aspirated repeatedly to prevent cell aggregates. Cell counts were manually performed and

107 cells were injected into each recipient mouse intravenously via the retroorbital plexus

on the same day as lethal irradiation of recipients. Stxbp5 WT 8 week old mice were used

as recipients and were lethally irradiated with an X-ray RS 2000 (Rad-sources) irradiator

delivering a single dose of 1100 rad. After marrow transplantation, mice were provided

with water supplemented with sulfatrim for 2 weeks, and allowed to reconstitute for 6

weeks prior to tail bleeding assay, blood collection, and carotid injury.

Vamp8 murine studies. Wild-type and Vamp8 KO mice were purchased from The

Jackson Laboratories (Bar Harbor, Maine). Plasma VWF was measured in platelet-poor

plasma collected into EDTA-tubes by an ELISA on 4-8 wk old mice. Bleeding time was

measured as described previously.(303) In brief, we measured the time to achieve

hemostasis after the distal 0.5 cm tip of a mouse tail (4-8 wk old) was amputated.

Page 198: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

175

Leukocyte rolling was measured as described previously.(301) In brief, 4-5 wk old mice

were anesthetized with a cocktail of ketamine/xylazine (80/12 mg/kg) and saline (10

mL/kg) via an intramuscular injection. Anesthetized mice were injected with Rhodamine

6G (Sigma-Aldrich) in order to label leukocytes. The mesentery was exteriorized, an area

containing target venules (120-150 µm in diameter) was selectedd the venules imaged by

a Nikon ECLIPSE Ti inverted fluorescent intravital microscope (Nikon Americas.

Melville, NY). After 120 s baseline recording, the mesenteric venules were superfused

with 10 µM Ca2+ ionophore A23187 (Sigma-Aldrich) in normal saline to activate

endothelial cells which induces leukocyte rolling. Images of leukocyte rolling in the

mesenteric venules were continuously recorded for a total of 10 min starting at the

baseline with a QUANTEM 512SC electron-multiplying CCD video camera

(Photometrics, Tucson, AZ). Using Image-Pro Analyzer 6.2 software, rolling leukocytes

were determined by counting the number of leukocytes that move asynchronously with

the blood flow or remained static over at least two consecutive frames. For each mouse,

we quantified leukocyte rolling in equally-divided video segments (5 during baseline and

20 during stimulation), and used the average number of rolling leukocytes per unit area

(512 pixel × 100 pixel) for quantification. The epinephrine challenge experiment was

performed by stimulating 8-12 wk old mice with subcutaneous injection of epinephrine

(0.5 mg/kg body weight). Blood was collected before and 30 min after challenge under

2.5% isoflurane by retro-orbital punch from left and right eye, respectively.

Statistical analyses. Results were expressed as mean ± SD except for the flow cytometry

fluorescence intensity represented as median ± SD. Significance between mean values

Page 199: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

176

was determined by the two-tailed student t test for comparison between two groups with

Gaussian distribution, and one-way ANOVA with Tukey multiple comparisons test for

comparison among three or more groups. For non-Gaussian distributed data including tail

bleeding time and mesenteric thrombosis measurement where a definite cut-off value was

assigned to some of the subjects, Kolmogorov-Smirnov test was used to compare two

groups. Responses affected by two factors were compared by two-way Tukey-corrected

ANOVA. For comparing carotid flow measurement among STXBP5 murine genotypes,

repeated measures two-way ANOVA with Tukey post test was used. Independent

variables with a value of P < .05 were considered as significant. For ANOVA post tests,

multiplicity adjusted P < .05 were considered as significant. The extent of protein co-

localization on confocal imaging was calculated using Pearson’s correlation coefficient

(444).

Study approval. All in vivo procedures and usage of mice were approved by the Division

of Laboratory Animal Medicine at the University of Rochester Medical Center (Protocol

numbers: UCAR 2010-005 and UCAR 2014-009).

Page 200: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

177

Table 2 List of antibodies

Antibody Name Species

Immunized Cat # Source

Anti-MYRIP antibody Goat ab10149 Abcam

Anti-Syntaxin 4 antibody Rabbit ab96382 Abcam

Anti-SNAP23 antibody Rabbit ab3340 Abcam

Anti-Von Willebrand Factor

antibody Rabbit ab6994 Abcam

Anti-VAMP1 antibody Rabbit ab3346 Abcam

SELP polyclonal antibody (B01P) Mouse H00006403-

B01P Abnova

FITC Anti-Mouse CD62P Rat 553744 BD Pharmingen

Purified Mouse Anti-GM130 Mouse 610822 BD Transduction

Laboratories

Purified Mouse Anti-Syntaxin 4 Mouse 610439 BD Transduction

Laboratories

Polyclonal Rabbit Anti-Caveolin Rabbit 610059 BD Transduction

Laboratories

Calnexin (C5C9) Rabbit 2679P Cell Signaling

EEA1 (C45B10) Rabbit 3288P Cell Signaling

LAMP1 (D2D11) XP® Rabbit 9091P Cell Signaling

Integrin alphaIIbbeta3 (GPIIb/IIIa,

CD41/CD61), clone JON/A Rat M023-2 Emfret

Anti - GPIbbeta derivative Rat X488 Emfret

Cy3-AffiniPure Bovine Anti-Goat

IgG (H+L) Bovine 805-165-180

Jackson

ImmunoResearch

Laboratories

Page 201: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

178

Alexa Fluor® 488 Goat Anti-

Rabbit IgG (H+L) Antibody,

highly cross-adsorbed

Goat A-11034 Molecular Probes

Alexa Fluor® 488 Goat Anti-

Mouse IgG (H+L) Antibody,

highly cross-adsorbed

Goat A-11029 Molecular Probes

Alexa Fluor® 594 Goat Anti-

Mouse IgG (H+L) Antibody,

highly cross-adsorbed

Goat A-11032 Molecular Probes

Alexa Fluor® 594 Goat Anti-

Rabbit IgG (H+L) Antibody,

highly cross-adsorbed

Goat A-11037 Molecular Probes

Alexa Fluor® 680 Donkey Anti-

Sheep IgG (H+L) Donkey A-21102 Molecular Probes

Rab27a Affinity Purified

Polyclonal Ab Sheep AF7245 R&D Systems

Human VAMP-8 Antibody Goat AF5354 R&D Systems

Goat IgG Horseradish Peroxidase-

conjugated Antibody Donkey HAF109 R&D Systems

SNAP 23 Antibody (H-50) Rabbit sc-50371 Santa Cruz

VAMP-1/2/3 Antibody (FL-118) Rabbit sc-13992 Santa Cruz

Tomosyn Antibody (15) Mouse sc-136105 Santa Cruz

Tomosyn Antibody (H-55) Rabbit sc-98350 Santa Cruz

SNAP25 Antibody (C-18) Goat sc-7538 Santa Cruz

VAMP2 Antibody (3E5) Mouse sc-69706 Santa Cruz

NSF Antibody (H-300) Rabbit sc-15339 Santa Cruz

Anti-Myosin Va (LF-18) antibody Rabbit M4812-.2ML SIGMA

Syntaxin 11 Rabbit 110 113 Synaptic Systems

GmbH

Page 202: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

179

SNAP23 Rabbit 111 203 Synaptic Systems

GmbH

VAMP8 Rabbit 104 303 Synaptic Systems

GmbH

Anti -Factor VIII Related Antigen

(VWF) Rabbit F0016-13 US Biologicals

Page 203: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

180

Table 3 Primers used for SYBR Green RT-qPCR

Gene RefSeq Forward (5'>3') Reverse (5'>3') Product

(bp)

SNAP23 NM_003825.3 ATGAGTCTCTGGAAAGTACG

AGG

CCACAGCATTTGTTGAGTTC

TG 190

SNAP25 NM_003081.3 TGTTGGATGAACAAGGAGA

ACAA CCGTCCTGATTATTGCCCCA 187

SNAP29 NM_004782.3 CCTGAACAGAATGGCACCCT TGGGGACAGGGTCTGTATCA 139

SNAP47 NM_053052.3 TGGAGGTGGCGGACAGATT AGGGTTCACAACTGGTCATG

G 129

SNAP91 NM_00124279

2.1 AGCCGGTCATGTTTGCACA

AGATCCGCTAATGGGTCCTT

T 139

B2M NM_004048.2 CCCAAGATAGTTAAGTGGG

ATCG

AGCAAGCAAGCAGAATTTG

GA 100

HPRT1 NM_000194.2 CCTGGCGTCGTGATTAGTGA

T

AGACGTTCAGTCCTGTCCAT

AA 131

YWHAZ NM_00113569

9.1

CCTGCATGAAGTCTGTAACT

GAG

GACCTACGGGCTCCTACAAC

A 100

GAPDH NM_002046.5 GGAGCGAGATCCCTCCAAA

AT

GGCTGTTGTCATACTTCTCA

TGG 197

Page 204: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

181

References

1. Furie B, and Furie BC. Mechanisms of thrombus formation. The New England

journal of medicine. 2008;359(9):938-49.

2. Wells PS, Forgie MA, and Rodger MA. Treatment of venous thromboembolism.

JAMA : the journal of the American Medical Association. 2014;311(7):717-28.

3. Carrier M, Le Gal G, Wells PS, and Rodger MA. Systematic Review: Case-

Fatality Rates of Recurrent Venous Thromboembolism and Major Bleeding Events

Among Patients Treated for Venous Thromboembolism. Ann Intern Med.

2010;152(9):578-+.

4. Tagalakis V, Patenaude V, Kahn SR, and Suissa S. Incidence of and Mortality

from Venous Thromboembolism in a Real-world Population: The Q-VTE Study Cohort.

Am J Med. 2013;126(9).

5. Beckman MG, Hooper WC, Critchley SE, and Ortel TL. Venous

Thromboembolism A Public Health Concern. Am J Prev Med. 2010;38(4):S495-S501.

6. Heit JA, Silverstein MD, Mohr DN, Petterson TM, O'Fallon WM, and Melton LJ.

Predictors of survival after deep vein thrombosis and pulmonary embolism - A

population-based, cohort study. Arch Intern Med. 1999;159(5):445-53.

7. Ashrani AA, and Heit JA. Incidence and cost burden of post-thrombotic

syndrome. J Thromb Thrombolys. 2009;28(4):465-76.

8. Pengo V, Lensing AWA, Prins MH, Marchiori A, Davidson BL, Tiozzo F,

Albanese P, Biasiolo A, Pegoraro C, Iliceto S, et al. Incidence of chronic thromboembolic

Page 205: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

182

pulmonary hypertension after pulmonary embolism. New Engl J Med.

2004;350(22):2257-64.

9. Kahn SR, and Ginsberg JS. Relationship between deep venous thrombosis and the

postthrombotic syndrome. Arch Intern Med. 2004;164(1):17-26.

10. Barritt DW, and Jordan SC. Anticoagulant Drugs in the Treatment of Pulmonary

Embolism - a Controlled Trial. Lancet. 1960;1(Jun18):1309-12.

11. Farge D, Debourdeau P, Beckers M, Baglin C, Bauersachs RM, Brenner B,

Brilhante D, Falanga A, Gerotzafias GT, Haim N, et al. International clinical practice

guidelines for the treatment and prophylaxis of venous thromboembolism in patients with

cancer. Journal of thrombosis and haemostasis : JTH. 2013;11(1):56-70.

12. Piazza G, and Goldhaber SZ. Venous Thromboembolism and Atherothrombosis

An Integrated Approach. Circulation. 2010;121(19):2146-50.

13. Lindblad B, Eriksson A, and Bergqvist D. Autopsy-Verified Pulmonary-

Embolism in a Surgical Department - Analysis of the Period from 1951 to 1988. Brit J

Surg. 1991;78(7):849-52.

14. Nieto JA, Solano R, Ruiz-Ribo MD, Ruiz-Gimenez N, Prandoni P, Kearon C,

Monreal M, and Investigators R. Fatal bleeding in patients receiving anticoagulant

therapy for venous thromboembolism: findings from the RIETE registry. J Thromb

Haemost. 2010;8(6):1216-22.

15. Davidson BL, Verheijen S, Lensing AW, Gebel M, Brighton TA, Lyons RM,

Rehm J, and Prins MH. Bleeding risk of patients with acute venous thromboembolism

taking nonsteroidal anti-inflammatory drugs or aspirin. JAMA internal medicine.

2014;174(6):947-53.

Page 206: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

183

16. Ginsberg JS. Management of venous thromboembolism. The New England

journal of medicine. 1996;335(24):1816-28.

17. Prandoni P, Lensing AWA, Piccioli A, Bernardi E, Simioni P, Girolami B,

Marchiori A, Sabbion P, Prins MH, Noventa F, et al. Recurrent venous thromboembolism

and bleeding complications during anticoagulant treatment in patients with cancer and

venous thrombosis. Blood. 2002;100(10):3484-8.

18. Rathbun S. The Surgeon General's Call to Action to Prevent Deep Vein

Thrombosis and Pulmonary Embolism. Circulation. 2009;119(15):E480-E2.

19. Anderson FA, Jr., and Spencer FA. Risk factors for venous thromboembolism.

Circulation. 2003;107(23 Suppl 1):I9-16.

20. Seligsohn U, and Lubetsky A. Genetic susceptibility to venous thrombosis. The

New England journal of medicine. 2001;344(16):1222-31.

21. Koster T, Blann AD, Briet E, Vandenbroucke JP, and Rosendaal FR. Role of

clotting factor VIII in effect of von Willebrand factor on occurrence of deep-vein

thrombosis. Lancet. 1995;345(8943):152-5.

22. Piazza G, and Goldhaber SZ. Venous thromboembolism and atherothrombosis: an

integrated approach. Circulation. 2010;121(19):2146-50.

23. Lind C, Flinterman LE, Enga KF, Severinsen MT, Kristensen SR, Braekkan SK,

Mathiesen EB, Njolstad I, Cannegieter SC, Overvad K, et al. Impact of incident venous

thromboembolism on risk of arterial thrombotic diseases. Circulation. 2014;129(8):855-

63.

Page 207: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

184

24. Di Minno MND, Tufano A, Ageno W, Prandoni P, and Di Minno G. Identifying

high-risk individuals for cardiovascular disease: similarities between venous and arterial

thrombosis in perspective. A 2011 update. Internal and emergency medicine.

2012;7(1):9-13.

25. Andrei MC, and Andercou A. Is there a Link Between Atherothrombosis and

Deep Venous Thrombosis? Maedica. 2014;9(1):94-7.

26. Sumpio BE, Riley JT, and Dardik A. Cells in focus: endothelial cell. Int J

Biochem Cell B. 2002;34(12):1508-12.

27. Morrissey JH, Macik BG, Neuenschwander PF, and Comp PC. Quantitation of

Activated Factor-Vii Levels in Plasma Using a Tissue Factor Mutant Selectively

Deficient in Promoting Factor-Vii Activation. Blood. 1993;81(3):734-44.

28. Butenas S. Tissue factor structure and function. Scientifica. 2012;2012(964862).

29. Farndale RW, Sixma JJ, Barnes MJ, and de Groot PG. The role of collagen in

thrombosis and hemostasis. Journal of thrombosis and haemostasis : JTH.

2004;2(4):561-73.

30. Ignarro LJ, Buga GM, Wood KS, Byrns RE, and Chaudhuri G. Endothelium-

derived relaxing factor produced and released from artery and vein is nitric oxide.

Proceedings of the National Academy of Sciences of the United States of America.

1987;84(24):9265-9.

31. Palmer RM, Ferrige AG, and Moncada S. Nitric oxide release accounts for the

biological activity of endothelium-derived relaxing factor. Nature. 1987;327(6122):524-

6.

Page 208: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

185

32. Marcus AJ, Broekman MJ, and Pinsky DJ. COX inhibitors and

thromboregulation. The New England journal of medicine. 2002;347(13):1025-6.

33. Marcus AJ, Broekman MJ, Drosopoulos JH, Olson KE, Islam N, Pinsky DJ, and

Levi R. Role of CD39 (NTPDase-1) in thromboregulation, cerebroprotection, and

cardioprotection. Seminars in thrombosis and hemostasis. 2005;31(2):234-46.

34. Cines DB, Pollak ES, Buck CA, Loscalzo J, Zimmerman GA, McEver RP, Pober

JS, Wick TM, Konkle BA, Schwartz BS, et al. Endothelial cells in physiology and in the

pathophysiology of vascular disorders. Blood. 1998;91(10):3527-61.

35. Rosenberg RD, and Rosenberg JS. Natural Anticoagulant Mechanisms. Journal of

Clinical Investigation. 1984;74(1):1-6.

36. Bonetti PO, Lerman LO, and Lerman A. Endothelial dysfunction: a marker of

atherosclerotic risk. Arterioscler Thromb Vasc Biol. 2003;23(2):168-75.

37. Broze GJ, Jr. Tissue factor pathway inhibitor. Thromb Haemost. 1995;74(1):90-3.

38. Broze GJ, and Girard TJ. Tissue factor pathway inhibitor: structure-function.

Front Biosci-Landmrk. 2012;17(262-80).

39. Lerman A, and Burnett JC, Jr. Intact and altered endothelium in regulation of

vasomotion. Circulation. 1992;86(6 Suppl):III12-9.

40. Lerman A, and Zeiher AM. Endothelial function: cardiac events. Circulation.

2005;111(3):363-8.

41. Horn IR, van den Berg BM, Moestrup SK, Pannekoek H, and van Zonneveld AJ.

Plasminogen activator inhibitor 1 contains a cryptic high affinity receptor binding site

Page 209: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

186

that is exposed upon complex formation with tissue-type plasminogen activator. Thromb

Haemost. 1998;80(5):822-8.

42. Cushman M, Lemaitre RN, Kuller LH, Psaty BM, Macy EM, Sharrett AR, and

Tracy RP. Fibrinolytic activation markers predict myocardial infarction in the elderly.

The Cardiovascular Health Study. Arterioscler Thromb Vasc Biol. 1999;19(3):493-8.

43. Furchgott RF, and Zawadzki JV. The obligatory role of endothelial cells in the

relaxation of arterial smooth muscle by acetylcholine. Nature. 1980;288(5789):373-6.

44. Martin W, Furchgott RF, Villani GM, and Jothianandan D. Depression of

contractile responses in rat aorta by spontaneously released endothelium-derived relaxing

factor. The Journal of pharmacology and experimental therapeutics. 1986;237(2):529-38.

45. Mitchell JA, Forstermann U, Warner TD, Pollock JS, Schmidt HH, Heller M, and

Murad F. Endothelial cells have a particulate enzyme system responsible for EDRF

formation: measurement by vascular relaxation. Biochemical and biophysical research

communications. 1991;176(3):1417-23.

46. Ignarro LJ, and Napoli C. Novel features of nitric oxide, endothelial nitric oxide

synthase, and atherosclerosis. Curr Diab Rep. 2005;5(1):17-23.

47. Forsberg EJ, Feuerstein G, Shohami E, and Pollard HB. Adenosine triphosphate

stimulates inositol phospholipid metabolism and prostacyclin formation in adrenal

medullary endothelial cells by means of P2-purinergic receptors. Proceedings of the

National Academy of Sciences of the United States of America. 1987;84(16):5630-4.

48. Levin EG, and Loskutoff DJ. Cultured bovine endothelial cells produce both

urokinase and tissue-type plasminogen activators. J Cell Biol. 1982;94(3):631-6.

Page 210: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

187

49. Hajjar KA. Homocysteine-induced modulation of tissue plasminogen activator

binding to its endothelial cell membrane receptor. The Journal of clinical investigation.

1993;91(6):2873-9.

50. Colucci M, Gesualdo L, Montemurro P, Cavallo LG, Conese M, Mascolo E,

Ranieri E, Di Paolo S, Schena FP, and Semeraro N. Cultured human mesangial cells

produce both type 1 and type 2 plasminogen activator inhibitors. Thromb Haemost.

1995;74(6):1516-20.

51. Libby P. Inflammation in atherosclerosis. Nature. 2002;420(6917):868-74.

52. Libby P, Ridker PM, and Maseri A. Inflammation and atherosclerosis.

Circulation. 2002;105(9):1135-43.

53. Widlansky ME, Gokce N, Keaney JF, Jr., and Vita JA. The clinical implications

of endothelial dysfunction. J Am Coll Cardiol. 2003;42(7):1149-60.

54. Vita JA, and Keaney JF, Jr. Endothelial function: a barometer for cardiovascular

risk? Circulation. 2002;106(6):640-2.

55. Pietramaggiori G, Scherer SS, Mathews JC, Gennaoui T, Lancerotto L, Ragno G,

Valeri CR, and Orgill DP. Quiescent platelets stimulate angiogenesis and diabetic wound

repair. The Journal of surgical research. 2010;160(1):169-77.

56. Assinger A. Platelets and infection - an emerging role of platelets in viral

infection. Frontiers in immunology. 2014;5(649).

57. Klinger MH, and Jelkmann W. Role of blood platelets in infection and

inflammation. Journal of interferon & cytokine research : the official journal of the

International Society for Interferon and Cytokine Research. 2002;22(9):913-22.

Page 211: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

188

58. Ware J, Corken A, and Khetpal R. Platelet function beyond hemostasis and

thrombosis. Current opinion in hematology. 2013;20(5):451-6.

59. Morrell CN, Aggrey AA, Chapman LM, and Modjeski KL. Emerging roles for

platelets as immune and inflammatory cells. Blood. 2014;123(18):2759-67.

60. Aggrey AA, Srivastava K, Ture S, Field DJ, and Morrell CN. Platelet Induction of

the Acute-Phase Response Is Protective in Murine Experimental Cerebral Malaria.

Journal of Immunology. 2013;190(9):4685-91.

61. Shi GF, Field DJ, Ko KA, Ture S, Srivastava K, Levy S, Kowalska MA, Poncz

M, Fowell DJ, and Morrell CN. Platelet factor 4 limits Th17 differentiation and cardiac

allograft rejection. Journal of Clinical Investigation. 2014;124(2):543-52.

62. Weyrich AS, and Zimmerman GA. Platelets: signaling cells in the immune

continuum. Trends in immunology. 2004;25(9):489-95.

63. Wagner DD, and Burger PC. Platelets in inflammation and thrombosis.

Arterioscler Thromb Vasc Biol. 2003;23(12):2131-7.

64. Iannacone M, Sitia G, Isogawa M, Marchese P, Castro MG, Lowenstein PR,

Chisari FV, Ruggeri ZM, and Guidotti LG. Platelets mediate cytotoxic T lymphocyte-

induced liver damage. Nature medicine. 2005;11(11):1167-9.

65. Kroll MH, Harris TS, Moake JL, Handin RI, and Schafer AI. von Willebrand

factor binding to platelet GpIb initiates signals for platelet activation. The Journal of

clinical investigation. 1991;88(5):1568-73.

66. Quinton TM, Ozdener F, Dangelmaier C, Daniel JL, and Kunapuli SP.

Glycoprotein VI-mediated platelet fibrinogen receptor activation occurs through calcium-

Page 212: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

189

sensitive and PKC-sensitive pathways without a requirement for secreted ADP. Blood.

2002;99(9):3228-34.

67. Wilner GD, Nossel HL, and LeRoy EC. Aggregation of platelets by collagen. The

Journal of clinical investigation. 1968;47(12):2616-21.

68. Ginsberg MH, Partridge A, and Shattil SJ. Integrin regulation. Current opinion in

cell biology. 2005;17(5):509-16.

69. Zou Z, Chen H, Schmaier AA, Hynes RO, and Kahn ML. Structure-function

analysis reveals discrete beta3 integrin inside-out and outside-in signaling pathways in

platelets. Blood. 2007;109(8):3284-90.

70. Kauskot A, and Hoylaerts MF. Platelet receptors. Handbook of experimental

pharmacology. 2012210):23-57.

71. Golebiewska EM, and Poole AW. Secrets of platelet exocytosis - what do we

really know about platelet secretion mechanisms? Br J Haematol. 2013.

72. Reed GL, Fitzgerald ML, and Polgar J. Molecular mechanisms of platelet

exocytosis: insights into the "secrete" life of thrombocytes. Blood. 2000;96(10):3334-42.

73. Bentfeld ME, and Bainton DF. Cytochemical-Localization of Lysosomal

Enzymes in Rat Megakaryocytes and Platelets. Journal of Clinical Investigation.

1975;56(6):1635-49.

74. Golebiewska EM, and Poole AW. Secrets of platelet exocytosis - what do we

really know about platelet secretion mechanisms? Brit J Haematol. 2014;165(2):204-16.

75. Wagner DD, and Frenette PS. The vessel wall and its interactions. Blood.

2008;111(11):5271-81.

Page 213: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

190

76. Mackman N. Triggers, targets and treatments for thrombosis. Nature.

2008;451(7181):914-8.

77. Bagot CN, and Arya R. Virchow and his triad: a question of attribution. Brit J

Haematol. 2008;143(2):180-90.

78. Barbui T, Finazzi G, and Falanga A. Myeloproliferative neoplasms and

thrombosis. Blood. 2013;122(13):2176-84.

79. Wolberg AS, Aleman MM, Leiderman K, and Machlus KR. Procoagulant activity

in hemostasis and thrombosis: Virchow's triad revisited. Anesthesia and analgesia.

2012;114(2):275-85.

80. Marchioli R, Finazzi G, Specchia G, Cacciola R, Cavazzina R, Cilloni D, De

Stefano V, Elli E, Iurlo A, Latagliata R, et al. Cardiovascular events and intensity of

treatment in polycythemia vera. The New England journal of medicine. 2013;368(1):22-

33.

81. Pearson TC. The risk of thrombosis in essential thrombocythemia and

polycythemia vera. Seminars in oncology. 2002;29(3 Suppl 10):16-21.

82. Adams BD, Baker R, Lopez JA, and Spencer S. Myeloproliferative disorders and

the hyperviscosity syndrome. Hematology/oncology clinics of North America.

2010;24(3):585-602.

83. Schafer AI. Thrombocytosis. The New England journal of medicine.

2004;350(12):1211-9.

84. Nishimura S, Manabe I, Nagasaki M, Kakuta S, Iwakura Y, Takayama N,

Ooehara J, Otsu M, Kamiya A, Petrich BG, et al. In vivo imaging visualizes discoid

Page 214: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

191

platelet aggregations without endothelium disruption and implicates contribution of

inflammatory cytokine and integrin signaling. Blood. 2012;119(8):e45-56.

85. Martinod K, and Wagner DD. Thrombosis: tangled up in NETs. Blood.

2014;123(18):2768-76.

86. Engelmann B, and Massberg S. Thrombosis as an intravascular effector of innate

immunity. Nat Rev Immunol. 2013;13(1):34-45.

87. Geddings JE, and Mackman N. Tumor-derived tissue factor-positive

microparticles and venous thrombosis in cancer patients. Blood. 2013;122(11):1873-80.

88. Owen BAL, Xue A, Heit JA, and Owen WG. Procoagulant activity, but not

number, of microparticles increases with age and in individuals after a single venous

thromboembolism. Thrombosis research. 2011;127(1):39-46.

89. Lipshits RU, and Belozorov AP. Exocytosis of Peripheral-Blood Leukocyte

Enzymes under the Effect of Aseptic Inflammation. Fiziol Zh Sssr. 1982;28(3):369-70.

90. Rothman JE. Mechanisms of intracellular protein transport. Nature.

1994;372(6501):55-63.

91. Bennett MK, and Scheller RH. The molecular machinery for secretion is

conserved from yeast to neurons. Proceedings of the National Academy of Sciences of the

United States of America. 1993;90(7):2559-63.

92. Palade G. Intracellular aspects of the process of protein synthesis. Science.

1975;189(4206):867.

93. Fries E, and Rothman JE. Transient Activity of Golgi-Like Membranes as Donors

of Vesicular Stomatitis Viral Glycoprotein Invitro. J Cell Biol. 1981;90(3):697-704.

Page 215: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

192

94. Balch WE, Dunphy WG, Braell WA, and Rothman JE. Reconstitution of the

Transport of Protein between Successive Compartments of the Golgi Measured by the

Coupled Incorporation of N-Acetylglucosamine. Cell. 1984;39(2):405-16.

95. Balch WE, Glick BS, and Rothman JE. Sequential Intermediates in the Pathway

of Intercompartmental Transport in a Cell-Free System. Cell. 1984;39(3):525-36.

96. Braell WA, Balch WE, Dobbertin DC, and Rothman JE. The Glycoprotein That Is

Transported between Successive Compartments of the Golgi in a Cell-Free System

Resides in Stacks of Cisternae. Cell. 1984;39(3):511-24.

97. Sollner T, Whiteheart SW, Brunner M, Erdjument-Bromage H, Geromanos S,

Tempst P, and Rothman JE. SNAP receptors implicated in vesicle targeting and fusion.

Nature. 1993;362(6418):318-24.

98. McMahon HT, Ushkaryov YA, Edelmann L, Link E, Binz T, Niemann H, Jahn R,

and Sudhof TC. Cellubrevin is a ubiquitous tetanus-toxin substrate homologous to a

putative synaptic vesicle fusion protein. Nature. 1993;364(6435):346-9.

99. Blasi J, Chapman ER, Link E, Binz T, Yamasaki S, De Camilli P, Sudhof TC,

Niemann H, and Jahn R. Botulinum neurotoxin A selectively cleaves the synaptic protein

SNAP-25. Nature. 1993;365(6442):160-3.

100. Hayashi T, McMahon H, Yamasaki S, Binz T, Hata Y, Sudhof TC, and Niemann

H. Synaptic vesicle membrane fusion complex: action of clostridial neurotoxins on

assembly. The EMBO journal. 1994;13(21):5051-61.

101. Friedrich MJ. 2013 Nobel Prize recognizes work of scientists who illuminated

molecular transport system of cells. JAMA : the journal of the American Medical

Association. 2013;310(19):2027-9.

Page 216: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

193

102. Pulido IR, Jahn R, and Gerke V. VAMP3 is associated with endothelial weibel-

palade bodies and participates in their Ca(2+)-dependent exocytosis. Biochimica et

biophysica acta. 2011;1813(5):1038-44.

103. Han WQ, Xia M, Zhang C, Zhang F, Xu M, Li NJ, and Li PL. SNARE-mediated

rapid lysosome fusion in membrane raft clustering and dysfunction of bovine coronary

arterial endothelium. American journal of physiology Heart and circulatory physiology.

2011;301(5):H2028-37.

104. Randriamboavonjy V, Schrader J, Busse R, and Fleming I. Insulin induces the

release of vasodilator compounds from platelets by a nitric oxide-G kinase-VAMP-3-

dependent pathway. The Journal of experimental medicine. 2004;199(3):347-56.

105. McIntosh DP, and Schnitzer JE. Caveolae require intact VAMP for targeted

transport in vascular endothelium. The American journal of physiology. 1999;277(6 Pt

2):H2222-32.

106. Isenmann S, Khew-Goodall Y, Gamble J, Vadas M, and Wattenberg BW. A

splice-isoform of vesicle-associated membrane protein-1 (VAMP-1) contains a

mitochondrial targeting signal. Mol Biol Cell. 1998;9(7):1649-60.

107. Schnitzer JE, Liu J, and Oh P. Endothelial caveolae have the molecular transport

machinery for vesicle budding, docking, and fusion including VAMP, NSF, SNAP,

annexins, and GTPases. The Journal of biological chemistry. 1995;270(24):14399-404.

108. Matsushita K, Morrell CN, Cambien B, Yang SX, Yamakuchi M, Bao C, Hara

MR, Quick RA, Cao W, O'Rourke B, et al. Nitric oxide regulates exocytosis by S-

nitrosylation of N-ethylmaleimide-sensitive factor. Cell. 2003;115(2):139-50.

Page 217: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

194

109. Fu J, Naren AP, Gao X, Ahmmed GU, and Malik AB. 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. 2005;280(5):3178-84.

110. Predescu SA, Predescu DN, Shimizu K, Klein IK, and Malik AB. Cholesterol-

dependent syntaxin-4 and SNAP-23 clustering regulates caveolar fusion with the

endothelial plasma membrane. The Journal of biological chemistry. 2005;280(44):37130-

8.

111. Predescu SA, Predescu DN, Timblin BK, Stan RV, and Malik AB. Intersectin

regulates fission and internalization of caveolae in endothelial cells. Mol Biol Cell.

2003;14(12):4997-5010.

112. Sehgal PB, Mukhopadhyay S, Xu F, Patel K, and Shah M. Dysfunction of Golgi

tethers, SNAREs, and SNAPs in monocrotaline-induced pulmonary hypertension.

American journal of physiology Lung cellular and molecular physiology.

2007;292(6):L1526-42.

113. Sprenger RR, Fontijn RD, van Marle J, Pannekoek H, and Horrevoets AJ. Spatial

segregation of transport and signalling functions between human endothelial caveolae

and lipid raft proteomes. The Biochemical journal. 2006;400(3):401-10.

114. Sudhof TC. The synaptic vesicle cycle: a cascade of protein-protein interactions.

Nature. 1995;375(6533):645-53.

115. Scheller RH. Membrane trafficking in the presynaptic nerve terminal. Neuron.

1995;14(5):893-7.

Page 218: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

195

116. Sutton RB, Fasshauer D, Jahn R, and Brunger AT. Crystal structure of a SNARE

complex involved in synaptic exocytosis at 2.4 A resolution. Nature.

1998;395(6700):347-53.

117. Weimbs T, Mostov K, Low SH, and Hofmann R. A model for structural similarity

between different SNARE complexes based on sequence relationships. Trends in cell

biology. 1998;8(7):260-2.

118. Hao JC, Salem N, Peng XR, Kelly RB, and Bennett MK. Effect of mutations in

vesicle-associated membrane protein (VAMP) on the assembly of multimeric protein

complexes. J Neurosci. 1997;17(5):1596-603.

119. Saifee O, Wei LP, and Nonet ML. The Caenorhabditis elegans unc-64 locus

encodes a syntaxin that interacts genetically with synaptobrevin. Molecular biology of the

cell. 1998;9(6):1235-52.

120. Fasshauer D, Bruns D, Shen B, Jahn R, and Brunger AT. A structural change

occurs upon binding of syntaxin to SNAP-25. Journal of Biological Chemistry.

1997;272(7):4582-90.

121. Goda Y. SNAREs and regulated vesicle exocytosis. Proceedings of the National

Academy of Sciences of the United States of America. 1997;94(3):769-72.

122. Hayashi T, Mcmahon H, Yamasaki S, Binz T, Hata Y, Sudhof TC, and Niemann

H. Synaptic Vesicle Membrane-Fusion Complex - Action of Clostridial Neurotoxins on

Assembly. Embo Journal. 1994;13(21):5051-61.

123. Yang B, Gonzalez L, Prekeris R, Steegmaier M, Advani RJ, and Scheller RH.

SNARE interactions are not selective - Implications for membrane fusion specificity.

Journal of Biological Chemistry. 1999;274(9):5649-53.

Page 219: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

196

124. Hanson PI, Roth R, Morisaki H, Jahn R, and Heuser JE. Structure and

conformational changes in NSF and its membrane receptor complexes visualized by

quick-freeze/deep-etch electron microscopy. Cell. 1997;90(3):523-35.

125. Weber T, Zemelman BV, McNew JA, Westermann B, Gmachl M, Parlati F,

Sollner TH, and Rothman JE. SNAREpins: minimal machinery for membrane fusion.

Cell. 1998;92(6):759-72.

126. Hua Y, and Scheller RH. Three SNARE complexes cooperate to mediate

membrane fusion. Proceedings of the National Academy of Sciences of the United States

of America. 2001;98(14):8065-70.

127. Glick BS, and Rothman JE. Possible role for fatty acyl-coenzyme A in

intracellular protein transport. Nature. 1987;326(6110):309-12.

128. Block MR, Glick BS, Wilcox CA, Wieland FT, and Rothman JE. Purification of

an N-ethylmaleimide-sensitive protein catalyzing vesicular transport. Proceedings of the

National Academy of Sciences of the United States of America. 1988;85(21):7852-6.

129. Clary DO, Griff IC, and Rothman JE. SNAPs, a family of NSF attachment

proteins involved in intracellular membrane fusion in animals and yeast. Cell.

1990;61(4):709-21.

130. Rothman JE. The protein machinery of vesicle budding and fusion. Protein

science : a publication of the Protein Society. 1996;5(2):185-94.

131. Sollner T, Bennett MK, Whiteheart SW, Scheller RH, and Rothman JE. A protein

assembly-disassembly pathway in vitro that may correspond to sequential steps of

synaptic vesicle docking, activation, and fusion. Cell. 1993;75(3):409-18.

Page 220: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

197

132. Ryu JK, Min D, Rah SH, Kim SJ, Park Y, Kim H, Hyeon C, Kim HM, Jahn R,

and Yoon TY. Spring-loaded unraveling of a single SNARE complex by NSF in one

round of ATP turnover. Science. 2015;347(6229):1485-9.

133. Sudhof TC, and Rothman JE. Membrane fusion: grappling with SNARE and SM

proteins. Science. 2009;323(5913):474-7.

134. Jahn R, and Sudhof TC. Membrane fusion and exocytosis. Annu Rev Biochem.

1999;68(863-911).

135. Paumet F, Rahimian V, and Rothman JE. The specificity of SNARE-dependent

fusion is encoded in the SNARE motif. Proceedings of the National Academy of Sciences

of the United States of America. 2004;101(10):3376-80.

136. McNew JA, Parlati F, Fukuda R, and Rothman JE. Compartmental specificity of

cellular membrane fusion encoded in SNARE proteins. Molecular biology of the cell.

2000;11(428a-a.

137. Fasshauer D, and Margittai M. A transient N-terminal interaction of SNAP-25

and syntaxin nucleates SNARE assembly. Journal of Biological Chemistry.

2004;279(9):7613-21.

138. Margittai M, Widengren J, Schweinberger E, Schroder GF, Felekyan S, Haustein

E, Konig M, Fasshauer D, Grubmuller H, Jahn R, et al. Single-molecule fluorescence

resonance energy transfer reveals a dynamic equilibrium between closed and open

conformations of syntaxin 1. Proceedings of the National Academy of Sciences of the

United States of America. 2003;100(26):15516-21.

139. Sudhof TC. Calcium control of neurotransmitter release. Cold Spring Harbor

perspectives in biology. 2012;4(1):a011353.

Page 221: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

198

140. Perin MS, Fried VA, Mignery GA, Jahn R, and Sudhof TC. Phospholipid binding

by a synaptic vesicle protein homologous to the regulatory region of protein kinase C.

Nature. 1990;345(6272):260-3.

141. Sudhof TC, and Jahn R. Proteins of Synaptic Vesicles Involved in Exocytosis and

Membrane Recycling. Neuron. 1991;6(5):665-77.

142. Perin MS, Fried VA, Mignery GA, Jahn R, and Sudhof TC. Phospholipid Binding

by a Synaptic Vesicle Protein Homologous to the Regulatory Region of Protein Kinase-

C. Nature. 1990;345(6272):260-3.

143. Brose N, Petrenko AG, Sudhof TC, and Jahn R. Synaptotagmin - a Calcium

Sensor on the Synaptic Vesicle Surface. Science. 1992;256(5059):1021-5.

144. Li C, Ullrich B, Zhang JZ, Anderson RGW, Brose N, and Sudhof TC. Ca2+-

Dependent and Ca2+-Independent Activities of Neural and Nonneural Synaptotagmins.

Nature. 1995;375(6532):594-9.

145. Sutton RB, Davletov BA, Berghuis AM, Sudhof TC, and Sprang SR. Structure of

the First C-2 Domain of Synaptotagmin .1. A Novel Ca2+/Phospholipid-Binding Fold.

Cell. 1995;80(6):929-38.

146. Chen YA, Scales SJ, and Scheller RH. Sequential SNARE assembly underlies

priming and triggering of exocytosis. Neuron. 2001;30(1):161-70.

147. McMahon HT, Missler M, Li C, and Sudhof TC. Complexins: cytosolic proteins

that regulate SNAP receptor function. Cell. 1995;83(1):111-9.

Page 222: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

199

148. Chen X, Tomchick DR, Kovrigin E, Arac D, Machius M, Sudhof TC, and Rizo J.

Three-dimensional structure of the complexin/SNARE complex. Neuron.

2002;33(3):397-409.

149. Tang J, Maximov A, Shin OH, Dai H, Rizo J, and Sudhof TC. A

complexin/synaptotagmin 1 switch controls fast synaptic vesicle exocytosis. Cell.

2006;126(6):1175-87.

150. Maximov A, Tang J, Yang X, Pang ZP, and Sudhof TC. Complexin controls the

force transfer from SNARE complexes to membranes in fusion. Science.

2009;323(5913):516-21.

151. Schaub JR, Lu XB, Doneske B, Shin YK, and Mcnew JA. Hemifusion arrest by

complexin is relieved by Ca2+-synaptotagmin I. Nat Struct Mol Biol. 2006;13(8):748-50.

152. Huntwork S, and Littleton JT. A complexin fusion clamp regulates spontaneous

neurotransmitter release and synaptic growth. Nature neuroscience. 2007;10(10):1235-7.

153. Reim K, Mansour M, Varoqueaux F, McMahon HT, Sudhof TC, Brose N, and

Rosenmund C. Complexins regulate a late step in Ca2+-dependent neurotransmitter

release. Cell. 2001;104(1):71-81.

154. Glynn D, Bortnick RA, and Morton AJ. Complexin II is essential for normal

neurological function in mice. Human molecular genetics. 2003;12(19):2431-48.

155. Trimbuch T, Xu J, Flaherty D, Tomchick DR, Rizo J, and Rosenmund C. Re-

examining how complexin inhibits neurotransmitter release. eLife. 2014;3(e02391).

156. Hata Y, Slaughter CA, and Sudhof TC. Synaptic Vesicle Fusion Complex

Contains Unc-18 Homolog Bound to Syntaxin. Nature. 1993;366(6453):347-51.

Page 223: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

200

157. Toonen RF, Wierda K, Sons MS, de Wit H, Cornelisse LN, Brussaard A, Plomp

JJ, and Verhage M. Munc18-1 expression levels control synapse recovery by regulating

readily releasable pool size. Proceedings of the National Academy of Sciences of the

United States of America. 2006;103(48):18332-7.

158. Verhage M, Maia AS, Plomp JJ, Brussaard AB, Heeroma JH, Vermeer H, Toonen

RF, Hammer RE, van den Berg TK, Missler M, et al. Synaptic assembly of the brain in

the absence of neurotransmitter secretion. Science. 2000;287(5454):864-9.

159. Schoch S, Deak F, Konigstorfer A, Mozhayeva M, Sara Y, Sudhof TC, and

Kavalali ET. SNARE function analyzed in synaptobrevin/VAMP knockout mice.

Science. 2001;294(5544):1117-22.

160. Saitsu H, Kato M, Mizuguchi T, Hamada K, Osaka H, Tohyama J, Uruno K,

Kumada S, Nishiyama K, Nishimura A, et al. De novo mutations in the gene encoding

STXBP1 (MUNC18-1) cause early infantile epileptic encephalopathy. Nature genetics.

2008;40(6):782-8.

161. Kim YO, Korff CM, Villaluz MMG, Suls A, Weckhuysen S, De Jonghe P, and

Scheffer IE. Head stereotypies in STXBP1 encephalopathy. Dev Med Child Neurol.

2013;55(8):769-72.

162. Mignot C, Moutard ML, Trouillard O, Gourfinkel-An I, Jacquette A, Arveiler B,

Morice-Picard F, Lacombe D, Chiron C, Ville D, et al. STXBP1-related encephalopathy

presenting as infantile spasms and generalized tremor in three patients. Epilepsia.

2011;52(10):1820-7.

163. Milh M, Villeneuve N, Chouchane M, Kaminska A, Laroche C, Barthez MA,

Gitiaux C, Bartoli C, Borges-Correia A, Cacciagli P, et al. Epileptic and nonepileptic

Page 224: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

201

features in patients with early onset epileptic encephalopathy and STXBP1 mutations.

Epilepsia. 2011;52(10):1828-34.

164. Saitsu H, Kato M, Okada I, Orii KE, Higuchi T, Hoshino H, Kubota M, Arai H,

Tagawa T, Kimura S, et al. STXBP1 mutations in early infantile epileptic encephalopathy

with suppression-burst pattern. Epilepsia. 2010;51(12):2397-405.

165. Swanson DA, Steel JM, and Valle D. Identification and characterization of the

human ortholog of rat STXBP1, a protein implicated in vesicle trafficking and

neurotransmitter release. Genomics. 1998;48(3):373-6.

166. van Breevoort D, Snijders AP, Hellen N, Weckhuysen S, van Hooren KWEM,

Eikenboom J, Valentijn K, Fernandez-Borja M, Ceulemans B, De Jonghe P, et al.

STXBP1 promotes Weibel-Palade body exocytosis through its interaction with the

Rab27A effector Slp4-a. Blood. 2014;123(20):3185-94.

167. Shen J, Tareste DC, Paumet F, Rothman JE, and Melia TJ. Selective activation of

cognate SNAREpins by Sec1/Munc18 proteins. Cell. 2007;128(1):183-95.

168. Misura KM, Scheller RH, and Weis WI. Three-dimensional structure of the

neuronal-Sec1-syntaxin 1a complex. Nature. 2000;404(6776):355-62.

169. Weimer RM, Richmond JE, Davis WS, Hadwiger G, Nonet ML, and Jorgensen

EM. Defects in synaptic vesicle docking in unc-18 mutants. Nature neuroscience.

2003;6(10):1023-30.

170. Rizo J, Chen X, and Arac D. Unraveling the mechanisms of synaptotagmin and

SNARE function in neurotransmitter release. Trends in cell biology. 2006;16(7):339-50.

Page 225: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

202

171. Varoqueaux F, Sigler A, Rhee JS, Brose N, Enk C, Reim K, and Rosenmund C.

Total arrest of spontaneous and evoked synaptic transmission but normal synaptogenesis

in the absence of Munc13-mediated vesicle priming. Proceedings of the National

Academy of Sciences of the United States of America. 2002;99(13):9037-42.

172. Augustin I, Rosenmund C, Sudhof TC, and Brose N. Munc13-1 is essential for

fusion competence of glutamatergic synaptic vesicles. Nature. 1999;400(6743):457-61.

173. Ma C, Li W, Xu Y, and Rizo J. Munc13 mediates the transition from the closed

syntaxin-Munc18 complex to the SNARE complex. Nat Struct Mol Biol. 2011;18(5):542-

9.

174. Ma C, Su L, Seven AB, Xu Y, and Rizo J. Reconstitution of the vital functions of

Munc18 and Munc13 in neurotransmitter release. Science. 2013;339(6118):421-5.

175. Basu J, Betz A, Brose N, and Rosenmund C. Munc13-1 C1 domain activation

lowers the energy barrier for synaptic vesicle fusion. J Neurosci. 2007;27(5):1200-10.

176. van de Bospoort R, Farina M, Schmitz SK, de Jong A, de Wit H, Verhage M, and

Toonen RF. Munc13 controls the location and efficiency of dense-core vesicle release in

neurons. J Cell Biol. 2012;199(6):883-91.

177. Nightingale T, and Cutler D. The secretion of von Willebrand factor from

endothelial cells; an increasingly complicated story. Journal of thrombosis and

haemostasis : JTH. 2013;11 Suppl 1(192-201).

178. Lowenstein CJ, Morrell CN, and Yamakuchi M. Regulation of Weibel-Palade

body exocytosis. Trends Cardiovasc Med. 2005;15(8):302-8.

Page 226: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

203

179. van Mourik JA, Romani de Wit T, and Voorberg J. Biogenesis and exocytosis of

Weibel-Palade bodies. Histochem Cell Biol. 2002;117(2):113-22.

180. Weibel ER, and Palade GE. New Cytoplasmic Components in Arterial

Endothelia. J Cell Biol. 1964;23(101-12).

181. Jaffe EA, Nachman RL, Becker CG, and Minick CR. Culture of human

endothelial cells derived from umbilical veins. Identification by morphologic and

immunologic criteria. The Journal of clinical investigation. 1973;52(11):2745-56.

182. Gimbrone MA, Jr., Cotran RS, and Folkman J. Human vascular endothelial cells

in culture. Growth and DNA synthesis. J Cell Biol. 1974;60(3):673-84.

183. Booyse FM, Quarfoot AJ, Bell S, Fass DN, Lewis JC, Mann KG, and Bowie EJ.

Cultured aortic endothelial cells from pigs with von Willebrand disease: in vitro model

for studying the molecular defect(s) of the disease. Proceedings of the National Academy

of Sciences of the United States of America. 1977;74(12):5702-6.

184. Wagner DD, Olmsted JB, and Marder VJ. Immunolocalization of von Willebrand

protein in Weibel-Palade bodies of human endothelial cells. J Cell Biol. 1982;95(1):355-

60.

185. Bonfanti R, Furie BC, Furie B, and Wagner DD. Padgem (Gmp140) Is a

Component of Weibel-Palade Bodies of Human-Endothelial Cells. Blood.

1989;73(5):1109-12.

186. Mcever RP, Beckstead JH, Moore KL, Marshallcarlson L, and Bainton DF. Gmp-

140, a Platelet Alpha-Granule Membrane-Protein, Is Also Synthesized by Vascular

Endothelial-Cells and Is Localized in Weibel-Palade Bodies. Journal of Clinical

Investigation. 1989;84(1):92-9.

Page 227: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

204

187. Stern D, Brett J, Harris K, and Nawroth P. Participation of endothelial cells in the

protein C-protein S anticoagulant pathway: the synthesis and release of protein S. J Cell

Biol. 1986;102(5):1971-8.

188. Hayward CP, Cramer EM, Song Z, Zheng S, Fung R, Masse JM, Stead RH, and

Podor TJ. Studies of multimerin in human endothelial cells. Blood. 1998;91(4):1304-17.

189. Lupu C, Lupu F, Dennehy U, Kakkar VV, and Scully MF. Thrombin induces the

redistribution and acute release of tissue factor pathway inhibitor from specific granules

within human endothelial cells in culture. Arterioscler Thromb Vasc Biol.

1995;15(11):2055-62.

190. Knipe L, Meli A, Hewlett L, Bierings R, Dempster J, Skehel P, Hannah MJ, and

Carter T. A revised model for the secretion of tPA and cytokines from cultured

endothelial cells. Blood. 2010;116(12):2183-91.

191. Rondaij MG, Bierings R, Kragt A, van Mourik JA, and Voorberg J. Dynamics

and plasticity of Weibel-Palade bodies in endothelial cells. Arterioscl Throm Vas.

2006;26(5):1002-7.

192. Weibel ER. Fifty years of Weibel-Palade bodies: the discovery and early history

of an enigmatic organelle of endothelial cells. Journal of thrombosis and haemostasis :

JTH. 2012;10(6):979-84.

193. Jaffe EA, Hoyer LW, and Nachman RL. Synthesis of von Willebrand factor by

cultured human endothelial cells. Proceedings of the National Academy of Sciences of the

United States of America. 1974;71(5):1906-9.

194. Nachman R, Levine R, and Jaffe EA. Synthesis of factor VIII antigen by cultured

guinea pig megakaryocytes. The Journal of clinical investigation. 1977;60(4):914-21.

Page 228: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

205

195. Kanaji S, Fahs SA, Shi Q, Haberichter SL, and Montgomery RR. Contribution of

platelet vs. endothelial VWF to platelet adhesion and hemostasis. Journal of thrombosis

and haemostasis : JTH. 2012;10(8):1646-52.

196. Reinders JH, Degroot PG, Gonsalves MD, Zandbergen J, Loesberg C, and

Vanmourik JA. Isolation of a Storage and Secretory Organelle Containing Von

Willebrand Protein from Cultured Human-Endothelial Cells. Biochimica et biophysica

acta. 1984;804(3):361-9.

197. Wagner DD, Saffaripour S, Bonfanti R, Sadler JE, Cramer EM, Chapman B, and

Mayadas TN. Induction of Specific Storage Organelles by Von-Willebrand Factor

Propolypeptide. Cell. 1991;64(2):403-13.

198. Furlan M, Robles R, and Lammle B. Partial purification and characterization of a

protease from human plasma cleaving von Willebrand factor to fragments produced by in

vivo proteolysis. Blood. 1996;87(10):4223-34.

199. Zheng X, Chung D, Takayama TK, Majerus EM, Sadler JE, and Fujikawa K.

Structure of von Willebrand factor-cleaving protease (ADAMTS13), a metalloprotease

involved in thrombotic thrombocytopenic purpura. The Journal of biological chemistry.

2001;276(44):41059-63.

200. Dong JF, Moake JL, Nolasco L, Bernardo A, Arceneaux W, Shrimpton CN,

Schade AJ, McIntire LV, Fujikawa K, and Lopez JA. ADAMTS-13 rapidly cleaves

newly secreted ultralarge von Willebrand factor multimers on the endothelial surface

under flowing conditions. Blood. 2002;100(12):4033-9.

201. Lenting PJ, Christophe OD, and Denis CV. von Willebrand factor biosynthesis,

secretion, and clearance: connecting the far ends. Blood. 2015;125(13):2019-28.

Page 229: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

206

202. Von Willebrand EA. Hereditary pseudohaemophilia. Haemophilia : the official

journal of the World Federation of Hemophilia. 1999;5(3):223-31; discussion 2.

203. Ngo KY, Glotz VT, Koziol JA, Lynch DC, Gitschier J, Ranieri P, Ciavarella N,

Ruggeri ZM, and Zimmerman TS. Homozygous and heterozygous deletions of the von

Willebrand factor gene in patients and carriers of severe von Willebrand disease.

Proceedings of the National Academy of Sciences of the United States of America.

1988;85(8):2753-7.

204. Rodeghiero F, Castaman G, and Dini E. Epidemiological investigation of the

prevalence of von Willebrand's disease. Blood. 1987;69(2):454-9.

205. Sadler JE. Biochemistry and genetics of von Willebrand factor. Annu Rev

Biochem. 1998;67(395-424).

206. Ward CM, Tetaz TJ, Andrews RK, and Berndt MC. Binding of the von

Willebrand Factor A1 domain to histone. Thrombosis research. 1997;86(6):469-77.

207. Brill A, Fuchs TA, Savchenko AS, Thomas GM, Martinod K, De Meyer SF,

Bhandari AA, and Wagner DD. Neutrophil extracellular traps promote deep vein

thrombosis in mice. J Thromb Haemost. 2012;10(1):136-44.

208. Fuchs TA, Brill A, Duerschmied D, Schatzberg D, Monestier M, Myers DD,

Wrobleski SK, Wakefield TW, Hartwig JH, and Wagner DD. Extracellular DNA traps

promote thrombosis. Proceedings of the National Academy of Sciences of the United

States of America. 2010;107(36):15880-5.

209. Kokame K, Sakata T, Kokubo Y, and Miyata T. von Willebrand factor-to-

ADAMTS13 ratio increases with age in a Japanese population. Journal of thrombosis

and haemostasis : JTH. 2011;9(7):1426-8.

Page 230: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

207

210. Aillaud MF, Pignol F, Alessi MC, Harle JR, Escande M, Mongin M, and Juhan-

Vague I. Increase in plasma concentration of plasminogen activator inhibitor, fibrinogen,

von Willebrand factor, factor VIII:C and in erythrocyte sedimentation rate with age.

Thromb Haemost. 1986;55(3):330-2.

211. Wannamethee SG, Lowe GD, Whincup PH, Rumley A, Walker M, and Lennon L.

Physical activity and hemostatic and inflammatory variables in elderly men. Circulation.

2002;105(15):1785-90.

212. Wannamethee SG, Lowe GD, Shaper G, Whincup PH, Rumley A, Walker M, and

Lennon L. The effects of different alcoholic drinks on lipids, insulin and haemostatic and

inflammatory markers in older men. Thromb Haemost. 2003;90(6):1080-7.

213. Blann AD. Associations of von Willebrand factor with age, sex and other risk

factors for atherosclerosis. Thromb Haemost. 1994;71(4):528-9.

214. Conlan MG, Folsom AR, Finch A, Davis CE, Sorlie P, Marcucci G, and Wu KK.

Associations of factor VIII and von Willebrand factor with age, race, sex, and risk factors

for atherosclerosis. The Atherosclerosis Risk in Communities (ARIC) Study. Thromb

Haemost. 1993;70(3):380-5.

215. Souto JC, Almasy L, Borrell M, Gari M, Martinez E, Mateo J, Stone WH,

Blangero J, and Fontcuberta J. Genetic determinants of hemostasis phenotypes in Spanish

families. Circulation. 2000;101(13):1546-51.

216. de Lange M, Snieder H, Ariens RA, Spector TD, and Grant PJ. The genetics of

haemostasis: a twin study. Lancet. 2001;357(9250):101-5.

217. Goodeve A, Eikenboom J, Castaman G, Rodeghiero F, Federici AB, Batlle J,

Meyer D, Mazurier C, Goudemand J, Schneppenheim R, et al. Phenotype and genotype

Page 231: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

208

of a cohort of families historically diagnosed with type 1 von Willebrand disease in the

European study, Molecular and Clinical Markers for the Diagnosis and Management of

Type 1 von Willebrand Disease (MCMDM-1VWD). Blood. 2007;109(1):112-21.

218. Robertson JD, Yenson PR, Rand ML, Blanchette VS, Carcao MD, Notley C,

Lillicrap D, and James PD. Expanded phenotype-genotype correlations in a pediatric

population with type 1 von Willebrand disease. Journal of thrombosis and haemostasis :

JTH. 2011;9(9):1752-60.

219. Eikenboom J, Van Marion V, Putter H, Goodeve A, Rodeghiero F, Castaman G,

Federici AB, Batlle J, Meyer D, Mazurier C, et al. Linkage analysis in families diagnosed

with type 1 von Willebrand disease in the European study, molecular and clinical markers

for the diagnosis and management of type 1 VWD. Journal of thrombosis and

haemostasis : JTH. 2006;4(4):774-82.

220. Gill JC, Endres-Brooks J, Bauer PJ, Marks WJ, Jr., and Montgomery RR. The

effect of ABO blood group on the diagnosis of von Willebrand disease. Blood.

1987;69(6):1691-5.

221. Jenkins PV, and O'Donnell JS. ABO blood group determines plasma von

Willebrand factor levels: a biologic function after all? Transfusion. 2006;46(10):1836-44.

222. Blann AD. Plasma von Willebrand factor, thrombosis, and the endothelium: the

first 30 years. Thromb Haemost. 2006;95(1):49-55.

223. Rydz N, Swystun LL, Notley C, Paterson AD, Riches JJ, Sponagle K, Boonyawat

B, Montgomery RR, James PD, and Lillicrap D. The C-type lectin receptor CLEC4M

binds, internalizes, and clears von Willebrand factor and contributes to the variation in

plasma von Willebrand factor levels. Blood. 2013;121(26):5228-37.

Page 232: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

209

224. Furlan M. Von Willebrand factor: molecular size and functional activity. Ann

Hematol. 1996;72(6):341-8.

225. Furlan M, Robles R, Galbusera M, Remuzzi G, Kyrle PA, Brenner B, Krause M,

Scharrer I, Aumann V, Mittler U, et al. von Willebrand factor-cleaving protease in

thrombotic thrombocytopenic purpura and the hemolytic-uremic syndrome. The New

England journal of medicine. 1998;339(22):1578-84.

226. Zheng XL. ADAMTS13 and von Willebrand Factor in Thrombotic

Thrombocytopenic Purpura. Annual review of medicine. 2015;66(211-25.

227. Sadler JE. Von Willebrand factor, ADAMTS13, and thrombotic

thrombocytopenic purpura. Blood. 2008;112(1):11-8.

228. Danesh J, Wheeler JG, Hirschfield GM, Eda S, Eiriksdottir G, Rumley A, Lowe

GD, Pepys MB, and Gudnason V. C-reactive protein and other circulating markers of

inflammation in the prediction of coronary heart disease. The New England journal of

medicine. 2004;350(14):1387-97.

229. Thompson SG, Kienast J, Pyke SD, Haverkate F, and van de Loo JC. Hemostatic

factors and the risk of myocardial infarction or sudden death in patients with angina

pectoris. European Concerted Action on Thrombosis and Disabilities Angina Pectoris

Study Group. The New England journal of medicine. 1995;332(10):635-41.

230. Spiel AO, Gilbert JC, and Jilma B. von Willebrand factor in cardiovascular

disease: focus on acute coronary syndromes. Circulation. 2008;117(11):1449-59.

231. Davies JA, Collins PW, Hathaway LS, and Bowen DJ. Effect of von Willebrand

factor Y/C1584 on in vivo protein level and function and interaction with ABO blood

group. Blood. 2007;109(7):2840-6.

Page 233: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

210

232. Mello TBT, Machado TFG, Montavao SAL, Ozello MC, and Annichino-

Bizzacchi JM. Assessing the Coagulation Factor Levels, Inherited Thrombophilia, and

ABO Blood Group on the Risk for Venous Thrombosis Among Brazilians. Clin Appl

Thromb-Hem. 2009;15(4):408-14.

233. Rubin DB, Wienerkronish JP, Murray JF, Green DR, Turner J, Luce JM,

Montgomery AB, Marks JD, and Matthay MA. Elevated Vonwillebrand-Factor Antigen

Is an Early Plasma Predictor of Acute Lung Injury in Nonpulmonary Sepsis Syndrome.

Journal of Clinical Investigation. 1990;86(2):474-80.

234. van Mourik JA, Boertjes R, Huisveld IA, Fijnvandraat K, Pajkrt D, van Genderen

PJJ, and Fijnheer R. von Willebrand factor propeptide in vascular disorders: A tool to

distinguish between acute and chronic endothelial cell perturbation. Blood.

1999;94(1):179-85.

235. Blann AD, Davis A, Miller JP, and McCollum CN. Von Willebrand factor and

soluble E-selectin in hyperlipidaemia: relationship to lipids and vascular disease.

American journal of hematology. 1997;55(1):15-23.

236. De Meyer SF, Stoll G, Wagner DD, and Kleinschnitz C. von Willebrand factor:

an emerging target in stroke therapy. Stroke; a journal of cerebral circulation.

2012;43(2):599-606.

237. Bonnefoy A, Vermylen J, and Hoylaerts MF. Inhibition of von Willebrand factor-

GPIb/IX/V interactions as a strategy to prevent arterial thrombosis. Expert review of

cardiovascular therapy. 2003;1(2):257-69.

238. Harlan JM. Leukocyte-endothelial interactions. Blood. 1985;65(3):513-25.

Page 234: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

211

239. Mayrovitz HN, Tuma RF, and Wiedeman MP. Leukocyte adherence in arterioles

following extravascular tissue trauma. Microvascular research. 1980;20(3):264-74.

240. Atherton A, and Born GV. Quantitative investigations of the adhesiveness of

circulating polymorphonuclear leucocytes to blood vessel walls. The Journal of

physiology. 1972;222(2):447-74.

241. R W. Erlauterungstaflen zur Physiologie und Entwicklungsgeschichte. Leopold

Voss, Leipzig. 1839.

242. Fahraeus R. The suspension stability of the blood. Physiological reviews.

1929;9(2):241-74.

243. Mogilnicki R, and Muczij T. The distribution of leukocytes in different parts of

the vascular system. Cr Soc Biol. 1931;108(603-4).

244. Bevilacqua MP, Pober JS, Wheeler ME, Cotran RS, and Gimbrone MA, Jr.

Interleukin 1 acts on cultured human vascular endothelium to increase the adhesion of

polymorphonuclear leukocytes, monocytes, and related leukocyte cell lines. The Journal

of clinical investigation. 1985;76(5):2003-11.

245. Denis CV, Andre P, Saffaripour S, and Wagner DD. Defect in regulated secretion

of P-selectin affects leukocyte recruitment in von Willebrand factor-deficient mice.

Proceedings of the National Academy of Sciences of the United States of America.

2001;98(7):4072-7.

246. Kameda H, Morita I, Handa M, Kaburaki J, Yoshida T, Mimori T, Murota S, and

Ikeda Y. Re-expression of functional P-selectin molecules on the endothelial cell surface

by repeated stimulation with thrombin. Br J Haematol. 1997;97(2):348-55.

Page 235: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

212

247. Utgaard JO, Jahnsen FL, Bakka A, Brandtzaeg P, and Haraldsen G. Rapid

secretion of prestored interleukin 8 from Weibel-Palade bodies of microvascular

endothelial cells. The Journal of experimental medicine. 1998;188(9):1751-6.

248. Wolff B, Burns AR, Middleton J, and Rot A. Endothelial cell "memory" of

inflammatory stimulation: human venular endothelial cells store interleukin 8 in Weibel-

Palade bodies. The Journal of experimental medicine. 1998;188(9):1757-62.

249. Wun TC, and Capuano A. Initiation and regulation of fibrinolysis in human

plasma at the plasminogen activator level. Blood. 1987;69(5):1354-62.

250. Schini VB, Hendrickson H, Heublein DM, Burnett JC, Jr., and Vanhoutte PM.

Thrombin enhances the release of endothelin from cultured porcine aortic endothelial

cells. European journal of pharmacology. 1989;165(2-3):333-4.

251. Emori T, Hirata Y, Ohta K, Shichiri M, and Marumo F. Secretory mechanism of

immunoreactive endothelin in cultured bovine endothelial cells. Biochemical and

biophysical research communications. 1989;160(1):93-100.

252. Russell FD, Skepper JN, and Davenport AP. Evidence using immunoelectron

microscopy for regulated and constitutive pathways in the transport and release of

endothelin. Journal of cardiovascular pharmacology. 1998;31(3):424-30.

253. Vischer UM, and Wagner DD. Cd63 Is a Component of Weibel-Palade Bodies of

Human Endothelial-Cells. Blood. 1993;82(4):1184-91.

254. Fiedler U, and Augustin HG. Angiopoietins: a link between angiogenesis and

inflammation. Trends in immunology. 2006;27(12):552-8.

Page 236: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

213

255. Fiedler U, Reiss Y, Scharpfenecker M, Grunow V, Koidl S, Thurston G, Gale

NW, Witzenrath M, Rosseau S, Suttorp N, et al. Angiopoietin-2 sensitizes endothelial

cells to TNF-alpha and has a crucial role in the induction of inflammation. Nature

medicine. 2006;12(2):235-9.

256. Wagner DD. The Weibel-Palade body: the storage granule for von Willebrand

factor and P-selectin. Thromb Haemost. 1993;70(1):105-10.

257. Calvert JW, Gundewar S, Yamakuchi M, Park PC, Baldwin WM, 3rd, Lefer DJ,

and Lowenstein CJ. Inhibition of N-ethylmaleimide-sensitive factor protects against

myocardial ischemia/reperfusion injury. Circulation research. 2007;101(12):1247-54.

258. Goodman DM, Burke AE, and Livingston EH. JAMA patient page. Bleeding

disorders. JAMA : the journal of the American Medical Association. 2012;308(14):1492.

259. Vischer UM. von Willebrand factor, endothelial dysfunction, and cardiovascular

disease. Journal of thrombosis and haemostasis : JTH. 2006;4(6):1186-93.

260. Mannucci PM. Drug therapy - Treatment of von Willebrand's disease. New Engl J

Med. 2004;351(7):683-94.

261. Pang ZPP, and Sudhof TC. Cell biology of Ca2+-triggered exocytosis. Current

opinion in cell biology. 2010;22(4):496-505.

262. Matsuuchi L, and Kelly RB. Constitutive and basal secretion from the endocrine

cell line, AtT-20. J Cell Biol. 1991;112(5):843-52.

263. Burgess TL, and Kelly RB. Constitutive and Regulated Secretion of Proteins.

Annu Rev Cell Biol. 1987;3(243-93).

Page 237: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

214

264. Sporn LA, Marder VJ, and Wagner DD. Inducible secretion of large, biologically

potent von Willebrand factor multimers. Cell. 1986;46(2):185-90.

265. Tsai HM, Nagel RL, Hatcher VB, Seaton AC, and Sussman, II. The high

molecular weight form of endothelial cell von Willebrand factor is released by the

regulated pathway. Br J Haematol. 1991;79(2):239-45.

266. Giblin JP, Hewlett LJ, and Hannah MJ. Basal secretion of von Willebrand factor

from human endothelial cells. Blood. 2008;112(4):957-64.

267. Lui-Roberts WW, Collinson LM, Hewlett LJ, Michaux G, and Cutler DF. An AP-

1/clathrin coat plays a novel and essential role in forming the Weibel-Palade bodies of

endothelial cells. J Cell Biol. 2005;170(4):627-36.

268. Rusu L, Andreeva A, Visintine DJ, Kim K, Vogel SM, Stojanovic-Terpo A,

Chernaya O, Liu G, Bakhshi FR, Haberichter SL, et al. G protein-dependent basal and

evoked endothelial cell vWF secretion. Blood. 2014;123(3):442-50.

269. Hannah MJ, Hume AN, Arribas M, Williams R, Hewlett LJ, Seabra MC, and

Cutler DF. Weibel-Palade bodies recruit Rab27 by a content-driven, maturation-

dependent mechanism that is independent of cell type. J Cell Sci. 2003;116(Pt 19):3939-

48.

270. Zenner HL, Collinson LM, Michaux G, and Cutler DF. High-pressure freezing

provides insights into Weibel-Palade body biogenesis. J Cell Sci. 2007;120(12):2117-25.

271. Rondaij MG, Bierings R, Kragt A, van Mourik JA, and Voorberg J. Dynamics

and plasticity of Weibel-Palade bodies in endothelial cells. Arterioscler Thromb Vasc

Biol. 2006;26(5):1002-7.

Page 238: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

215

272. Xiong Y, Hu ZQ, Han XF, Jiang BB, Zhang RL, Zhang XY, Lu Y, Geng CY, Li

W, He YL, et al. Hypertensive stretch regulates endothelial exocytosis of Weibel-Palade

bodies through VEGF receptor 2 signaling pathways. Cell Res. 2013;23(6):820-34.

273. Birch KA, Pober JS, Zavoico GB, Means AR, and Ewenstein BM.

Calcium/calmodulin transduces thrombin-stimulated secretion: studies in intact and

minimally permeabilized human umbilical vein endothelial cells. J Cell Biol.

1992;118(6):1501-10.

274. van den Eijnden-Schrauwen Y, Atsma DE, Lupu F, de Vries RE, Kooistra T, and

Emeis JJ. Involvement of calcium and G proteins in the acute release of tissue-type

plasminogen activator and von Willebrand factor from cultured human endothelial cells.

Arterioscler Thromb Vasc Biol. 1997;17(10):2177-87.

275. Vischer UM, and Wollheim CB. Epinephrine induces von Willebrand factor

release from cultured endothelial cells: involvement of cyclic AMP-dependent signalling

in exocytosis. Thromb Haemost. 1997;77(6):1182-8.

276. Kaufmann JE, Oksche A, Wollheim CB, Gunther G, Rosenthal W, and Vischer

UM. Vasopressin-induced von Willebrand factor secretion from endothelial cells

involves V2 receptors and cAMP. The Journal of clinical investigation. 2000;106(1):107-

16.

277. Vischer UM, and Wollheim CB. Purine nucleotides induce regulated secretion of

von Willebrand factor: Involvement of cytosolic Ca2+ and cyclic adenosine

monophosphate-dependent signaling in endothelial exocytosis. Blood. 1998;91(1):118-

27.

Page 239: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

216

278. Vischer UM, Barth H, and Wollheim CB. Regulated von Willebrand factor

secretion is associated with agonist-specific patterns of cytoskeletal remodeling in

cultured endothelial cells. Arterioscler Thromb Vasc Biol. 2000;20(3):883-91.

279. van Nieuw Amerongen GP, van Delft S, Vermeer MA, Collard JG, and van

Hinsbergh VW. Activation of RhoA by thrombin in endothelial hyperpermeability: role

of Rho kinase and protein tyrosine kinases. Circulation research. 2000;87(4):335-40.

280. Wojciak-Stothard B, Potempa S, Eichholtz T, and Ridley AJ. Rho and Rac but not

Cdc42 regulate endothelial cell permeability. J Cell Sci. 2001;114(Pt 7):1343-55.

281. Cullere X, Shaw SK, Andersson L, Hirahashi J, Luscinskas FW, and Mayadas

TN. Regulation of vascular endothelial barrier function by Epac, a cAMP-activated

exchange factor for Rap GTPase. Blood. 2005;105(5):1950-5.

282. Fukuhara S, Sakurai A, Sano H, Yamagishi A, Somekawa S, Takakura N, Saito

Y, Kangawa K, and Mochizuki N. Cyclic AMP potentiates vascular endothelial cadherin-

mediated cell-cell contact to enhance endothelial barrier function through an Epac-Rap1

signaling pathway. Molecular and cellular biology. 2005;25(1):136-46.

283. Kooistra MRH, Corada M, Dejana E, and Bos JL. Epac1 regulates integrity of

endothelial cell junctions through VE-cadherin. Febs Letters. 2005;579(22):4966-72.

284. Rothblatt JA, Deshaies RJ, Sanders SL, Daum G, and Schekman R. Multiple

Genes Are Required for Proper Insertion of Secretory Proteins into the Endoplasmic-

Reticulum in Yeast. J Cell Biol. 1989;109(6):2641-52.

285. Ferronovick S, Hansen W, Schauer I, and Schekman R. Genes Required for

Completion of Import of Proteins into the Endoplasmic-Reticulum in Yeast. J Cell Biol.

1984;98(1):44-53.

Page 240: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

217

286. Sudhof TC, Decamilli P, Niemann H, and Jahn R. Membrane-Fusion Machinery -

Insights from Synaptic Proteins. Cell. 1993;75(1):1-4.

287. Sudhof TC. The Synaptic Vesicle Cycle - a Cascade of Protein-Protein

Interactions. Nature. 1995;375(6533):645-53.

288. Wickner W, and Schekman R. Membrane fusion. Nat Struct Mol Biol.

2008;15(7):658-64.

289. Bajjalieh SM, and Scheller RH. Synaptic vesicle proteins and exocytosis. Adv

Second Messenger Phosphoprotein Res. 1994;29(59-79).

290. Jahn R, and Scheller RH. SNAREs--engines for membrane fusion. Nat Rev Mol

Cell Biol. 2006;7(9):631-43.

291. Sudhof TC. The synaptic vesicle cycle revisited. Neuron. 2000;28(2):317-20.

292. Martens S, and McMahon HT. Mechanisms of membrane fusion: disparate

players and common principles. Nat Rev Mol Cell Biol. 2008;9(7):543-56.

293. Jung JJ, Tiwari A, Inamdar SM, Thomas CP, Goel A, and Choudhury A.

Secretion of soluble vascular endothelial growth factor receptor 1 (sVEGFR1/sFlt1)

requires Arf1, Arf6, and Rab11 GTPases. PloS one. 2012;7(9):e44572.

294. van Hooren KW, van Agtmaal EL, Fernandez-Borja M, van Mourik JA, Voorberg

J, and Bierings R. The Epac-Rap1 signaling pathway controls cAMP-mediated exocytosis

of Weibel-Palade bodies in endothelial cells. The Journal of biological chemistry.

2012;287(29):24713-20.

Page 241: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

218

295. Yamakuchi M, Ferlito M, Morrell CN, Matsushita K, Fletcher CA, Cao W, and

Lowenstein CJ. Exocytosis of endothelial cells is regulated by N-ethylmaleimide-

sensitive factor. Methods in molecular biology. 2008;440(203-15).

296. Bierings R, Hellen N, Kiskin N, Knipe L, Fonseca AV, Patel B, Meli A, Rose M,

Hannah MJ, and Carter T. The interplay between the Rab27A effectors Slp4-a and

MyRIP controls hormone-evoked Weibel-Palade body exocytosis. Blood.

2012;120(13):2757-67.

297. Pulido IR, Nightingale TD, Darchen F, Seabra MC, Cutler DF, and Gerke V.

Myosin Va Acts in Concert with Rab27a and MyRIP to Regulate Acute Von-Willebrand

Factor Release from Endothelial Cells. Traffic. 2011;12(10):1371-82.

298. Nightingale TD, Pattni K, Hume AN, Seabra MC, and Cutler DF. Rab27a and

MyRIP regulate the amount and multimeric state of VWF released from endothelial cells.

Blood. 2009;113(20):5010-8.

299. Kim KS, Park JY, Jou I, and Park SM. Regulation of Weibel-Palade body

exocytosis by alpha-synuclein in endothelial cells. The Journal of biological chemistry.

2010;285(28):21416-25.

300. Lowenstein CJ, and Tsuda H. N-ethylmaleimide-sensitive factor: a redox sensor

in exocytosis. Biological chemistry. 2006;387(10-11):1377-83.

301. Morrell CN, Matsushita K, and Lowenstein CJ. A novel inhibitor of N-

ethylmaleimide-sensitive factor decreases leukocyte trafficking and peritonitis. The

Journal of pharmacology and experimental therapeutics. 2005;314(1):155-61.

Page 242: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

219

302. Matsushita K, Morrell CN, Mason RJ, Yamakuchi M, Khanday FA, Irani K, and

Lowenstein CJ. Hydrogen peroxide regulation of endothelial exocytosis by inhibition of

N-ethylmaleimide sensitive factor. J Cell Biol. 2005;170(1):73-9.

303. Matsushita K, Morrell CN, and Lowenstein CJ. A novel class of fusion

polypeptides inhibits exocytosis. Molecular pharmacology. 2005;67(4):1137-44.

304. Sollner TH, and Sequeira S. S-nitrosylation of NSF controls membrane

trafficking. Cell. 2003;115(2):127-9.

305. Zografou S, Basagiannis D, Papafotika A, Shirakawa R, Horiuchi H, Auerbach D,

Fukuda M, and Christoforidis S. A complete Rab screening reveals novel insights in

Weibel-Palade body exocytosis. J Cell Sci. 2012;125(Pt 20):4780-90.

306. Rojo Pulido I, Nightingale TD, Darchen F, Seabra MC, Cutler DF, and Gerke V.

Myosin Va acts in concert with Rab27a and MyRIP to regulate acute von-Willebrand

factor release from endothelial cells. Traffic. 2011;12(10):1371-82.

307. Fukuda M, Imai A, Nashida T, and Shimomura H. Slp4-a/granuphilin-a interacts

with syntaxin-2/3 in a Munc18-2-dependent manner. The Journal of biological chemistry.

2005;280(47):39175-84.

308. Ito T, Yamakuchi M, and Lowenstein CJ. Thioredoxin increases exocytosis by

denitrosylating N-ethylmaleimide-sensitive factor. The Journal of biological chemistry.

2011;286(13):11179-84.

309. Huang J, Motto DG, Bundle DR, and Sadler JE. Shiga toxin B subunits induce

VWF secretion by human endothelial cells and thrombotic microangiopathy in

ADAMTS13-deficient mice. Blood. 2010;116(18):3653-9.

Page 243: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

220

310. Liu F, Huang J, and Sadler JE. Shiga toxin (Stx)1B and Stx2B induce von

Willebrand factor secretion from human umbilical vein endothelial cells through different

signaling pathways. Blood. 2011;118(12):3392-8.

311. Into T, Kanno Y, Dohkan J, Nakashima M, Inomata M, Shibata K, Lowenstein

CJ, and Matsushita K. Pathogen recognition by Toll-like receptor 2 activates Weibel-

Palade body exocytosis in human aortic endothelial cells. The Journal of biological

chemistry. 2007;282(11):8134-41.

312. Bhatia R, Matsushita K, Yamakuchi M, Morrell CN, Cao W, and Lowenstein CJ.

Ceramide triggers Weibel-Palade body exocytosis. Circulation research. 2004;95(3):319-

24.

313. Matsushita K, Morrell CN, and Lowenstein CJ. Sphingosine 1-phosphate

activates Weibel-Palade body exocytosis. Proceedings of the National Academy of

Sciences of the United States of America. 2004;101(31):11483-7.

314. Jeong Y, Chaupin DF, Matsushita K, Yamakuchi M, Cameron SJ, Morrell CN,

and Lowenstein CJ. Aldosterone activates endothelial exocytosis. Proceedings of the

National Academy of Sciences of the United States of America. 2009;106(10):3782-7.

315. Yamakuchi M, Kirkiles-Smith NC, Ferlito M, Cameron SJ, Bao C, Fox-Talbot K,

Wasowska BA, Baldwin WM, 3rd, Pober JS, and Lowenstein CJ. Antibody to human

leukocyte antigen triggers endothelial exocytosis. Proceedings of the National Academy

of Sciences of the United States of America. 2007;104(4):1301-6.

316. Torisu T, Torisu K, Lee IH, Liu J, Malide D, Combs CA, Wu XS, Rovira, II,

Fergusson MM, Weigert R, et al. Autophagy regulates endothelial cell processing,

maturation and secretion of von Willebrand factor. Nature medicine. 2013;19(10):1281-7.

Page 244: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

221

317. Smith NL, Chen MH, Dehghan A, Strachan DP, Basu S, Soranzo N, Hayward C,

Rudan I, Sabater-Lleal M, Bis JC, et al. Novel associations of multiple genetic loci with

plasma levels of factor VII, factor VIII, and von Willebrand factor: The CHARGE

(Cohorts for Heart and Aging Research in Genome Epidemiology) Consortium.

Circulation. 2010;121(12):1382-92.

318. Antoni G, Oudot-Mellakh T, Dimitromanolakis A, Germain M, Cohen W, Wells

P, Lathrop M, Gagnon F, Morange PE, and Tregouet DA. Combined analysis of three

genome-wide association studies on vWF and FVIII plasma levels. BMC medical

genetics. 2011;12(102.

319. van Loon JE, Leebeek FW, Deckers JW, Dippel DW, Poldermans D, Strachan

DP, Tang W, O'Donnell CJ, Smith NL, and de Maat MP. Effect of genetic variations in

syntaxin-binding protein-5 and syntaxin-2 on von Willebrand factor concentration and

cardiovascular risk. Circulation Cardiovascular genetics. 2010;3(6):507-12.

320. Smith NL, Rice KM, Bovill EG, Cushman M, Bis JC, McKnight B, Lumley T,

Glazer NL, van Hylckama Vlieg A, Tang W, et al. Genetic variation associated with

plasma von Willebrand factor levels and the risk of incident venous thrombosis. Blood.

2011;117(22):6007-11.

321. Fujita Y, Shirataki H, Sakisaka T, Asakura T, Ohya T, Kotani H, Yokoyama S,

Nishioka H, Matsuura Y, Mizoguchi A, et al. Tomosyn: a syntaxin-1-binding protein that

forms a novel complex in the neurotransmitter release process. Neuron. 1998;20(5):905-

15.

322. Yokoyama S, Shirataki H, Sakisaka T, and Takai Y. Three splicing variants of

tomosyn and identification of their syntaxin-binding region. Biochemical and biophysical

research communications. 1999;256(1):218-22.

Page 245: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

222

323. Groffen AJ, Jacobsen L, Schut D, and Verhage M. Two distinct genes drive

expression of seven tomosyn isoforms in the mammalian brain, sharing a conserved

structure with a unique variable domain. Journal of neurochemistry. 2005;92(3):554-68.

324. Hattendorf DA, Andreeva A, Gangar A, Brennwald PJ, and Weis WI. Structure of

the yeast polarity protein Sro7 reveals a SNARE regulatory mechanism. Nature.

2007;446(7135):567-71.

325. Lehman K, Rossi G, Adamo JE, and Brennwald P. Yeast homologues of tomosyn

and lethal giant larvae function in exocytosis and are associated with the plasma

membrane SNARE, Sec9. J Cell Biol. 1999;146(1):125-40.

326. Musch A, Cohen D, Yeaman C, Nelson WJ, Rodriguez-Boulan E, and Brennwald

PJ. Mammalian homolog of Drosophila tumor suppressor lethal (2) giant larvae interacts

with basolateral exocytic machinery in Madin-Darby canine kidney cells. Mol Biol Cell.

2002;13(1):158-68.

327. Gangar A, Rossi G, Andreeva A, Hales R, and Brennwald P. Structurally

conserved interaction of Lgl family with SNAREs is critical to their cellular function.

Current biology : CB. 2005;15(12):1136-42.

328. Pobbati AV, Razeto A, Boddener M, Becker S, and Fasshauer D. Structural basis

for the inhibitory role of tomosyn in exocytosis. The Journal of biological chemistry.

2004;279(45):47192-200.

329. Hatsuzawa K, Lang T, Fasshauer D, Bruns D, and Jahn R. The R-SNARE motif

of tomosyn forms SNARE core complexes with syntaxin 1 and SNAP-25 and down-

regulates exocytosis. The Journal of biological chemistry. 2003;278(33):31159-66.

Page 246: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

223

330. Masuda ES, Huang BC, Fisher JM, Luo Y, and Scheller RH. Tomosyn binds t-

SNARE proteins via a VAMP-like coiled coil. Neuron. 1998;21(3):479-80.

331. Yizhar O, Lipstein N, Gladycheva SE, Matti U, Ernst SA, Rettig J, Stuenkel EL,

and Ashery U. Multiple functional domains are involved in tomosyn regulation of

exocytosis. Journal of neurochemistry. 2007;103(2):604-16.

332. Sakisaka T, Yamamoto Y, Mochida S, Nakamura M, Nishikawa K, Ishizaki H,

Okamoto-Tanaka M, Miyoshi J, Fujiyoshi Y, Manabe T, et al. Dual inhibition of SNARE

complex formation by tomosyn ensures controlled neurotransmitter release. J Cell Biol.

2008;183(2):323-37.

333. Yizhar O, Matti U, Melamed R, Hagalili Y, Bruns D, Rettig J, and Ashery U.

Tomosyn inhibits priming of large dense-core vesicles in a calcium-dependent manner.

Proceedings of the National Academy of Sciences of the United States of America.

2004;101(8):2578-83.

334. Yamamoto Y, Fujikura K, Sakaue M, Okimura K, Kobayashi Y, Nakamura T,

and Sakisaka T. The tail domain of tomosyn controls membrane fusion through tomosyn

displacement by VAMP2. Biochemical and biophysical research communications.

2010;399(1):24-30.

335. Williams AL, Bielopolski N, Meroz D, Lam AD, Passmore DR, Ben-Tal N, Ernst

SA, Ashery U, and Stuenkel EL. Structural and functional analysis of tomosyn identifies

domains important in exocytotic regulation. The Journal of biological chemistry.

2011;286(16):14542-53.

336. Baba T, Sakisaka T, Mochida S, and Takai Y. PKA-catalyzed phosphorylation of

tomosyn and its implication in Ca2+-dependent exocytosis of neurotransmitter. J Cell

Biol. 2005;170(7):1113-25.

Page 247: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

224

337. Gladycheva SE, Lam AD, Liu J, D'Andrea-Merrins M, Yizhar O, Lentz SI,

Ashery U, Ernst SA, and Stuenkel EL. Receptor-mediated regulation of tomosyn-

syntaxin 1A interactions in bovine adrenal chromaffin cells. The Journal of biological

chemistry. 2007;282(31):22887-99.

338. Johnson AD, Handsaker RE, Pulit SL, Nizzari MM, O'Donnell CJ, and de Bakker

PI. SNAP: a web-based tool for identification and annotation of proxy SNPs using

HapMap. Bioinformatics. 2008;24(24):2938-9.

339. van Loon JE, Sanders YV, de Wee EM, Kruip MJ, de Maat MP, and Leebeek

FW. Effect of genetic variation in STXBP5 and STX2 on von Willebrand factor and

bleeding phenotype in type 1 von Willebrand disease patients. PloS one.

2012;7(7):e40624.

340. Sanders YV, van der Bom JG, Isaacs A, Cnossen MH, de Maat MP, Laros-van

Gorkom BA, Fijnvandraat K, Meijer K, van Duijn CM, Mauser-Bunschoten EP, et al.

CLEC4M and STXBP5 gene variation contribute to von Willebrand factor level variation

in von Willebrand disease. Journal of thrombosis and haemostasis : JTH. 2015.

341. Yamamoto Y, and Sakisaka T. In: Mochida S ed. Presynaptic Terminals. Springer

Japan; 2015:129-40.

342. Hagedorn I, Vogtle T, and Nieswandt B. Arterial thrombus formation. Novel

mechanisms and targets. Hamostaseologie. 2010;30(3):127-35.

343. Stoll G, Kleinschnitz C, and Nieswandt B. Molecular mechanisms of thrombus

formation in ischemic stroke: novel insights and targets for treatment. Blood.

2008;112(9):3555-62.

Page 248: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

225

344. Falanga A, Marchetti M, Vignoli A, and Balducci D. Clotting mechanisms and

cancer: implications in thrombus formation and tumor progression. Clin Adv Hematol

Oncol. 2003;1(11):673-8.

345. Valentijn KM, Sadler JE, Valentijn JA, Voorberg J, and Eikenboom J. Functional

architecture of Weibel-Palade bodies. Blood. 2011;117(19):5033-43.

346. Yamamoto Y, Mochida S, Miyazaki N, Kawai K, Fujikura K, Kurooka T, Iwasaki

K, and Sakisaka T. Tomosyn inhibits synaptotagmin-1-mediated step of Ca2+-dependent

neurotransmitter release through its N-terminal WD40 repeats. The Journal of biological

chemistry. 2010;285(52):40943-55.

347. Savage B, Ginsberg MH, and Ruggeri ZM. Influence of fibrillar collagen

structure on the mechanisms of platelet thrombus formation under flow. Blood.

1999;94(8):2704-15.

348. McEwen JM, Madison JM, Dybbs M, and Kaplan JM. Antagonistic regulation of

synaptic vesicle priming by Tomosyn and UNC-13. Neuron. 2006;51(3):303-15.

349. Grosshans BL, Andreeva A, Gangar A, Niessen S, Yates JR, 3rd, Brennwald P,

and Novick P. The yeast lgl family member Sro7p is an effector of the secretory Rab

GTPase Sec4p. J Cell Biol. 2006;172(1):55-66.

350. Bielopolski N, Lam AD, Bar-On D, Sauer M, Stuenkel EL, and Ashery U.

Differential interaction of tomosyn with syntaxin and SNAP25 depends on domains in

the WD40 beta-propeller core and determines its inhibitory activity. The Journal of

biological chemistry. 2014;289(24):17087-99.

351. Yu H, Rathore SS, Gulbranson DR, and Shen J. The N- and C-terminal domains

of tomosyn play distinct roles in soluble N-ethylmaleimide-sensitive factor attachment

Page 249: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

226

protein receptor binding and fusion regulation. The Journal of biological chemistry.

2014;289(37):25571-80.

352. Bhatnagar S, Soni MS, Wrighton LS, Hebert AS, Zhou AS, Paul PK, Gregg T,

Rabaglia ME, Keller MP, Coon JJ, et al. Phosphorylation and degradation of tomosyn-2

de-represses insulin secretion. The Journal of biological chemistry. 2014;289(36):25276-

86.

353. Cheviet S, Bezzi P, Ivarsson R, Renstrom E, Viertl D, Kasas S, Catsicas S, and

Regazzi R. Tomosyn-1 is involved in a post-docking event required for pancreatic beta-

cell exocytosis. J Cell Sci. 2006;119(Pt 14):2912-20.

354. Widberg CH, Bryant NJ, Girotti M, Rea S, and James DE. Tomosyn interacts

with the t-SNAREs syntaxin4 and SNAP23 and plays a role in insulin-stimulated GLUT4

translocation. The Journal of biological chemistry. 2003;278(37):35093-101.

355. Schwab Y, Mouton J, Chasserot-Golaz S, Marty I, Maulet Y, and Jover E.

Calcium-dependent translocation of synaptotagmin to the plasma membrane in the

dendrites of developing neurones. Molecular Brain Research. 2001;96(1-2):1-13.

356. Tang J, Maximov A, Shin OH, Dai H, Rizo J, and Sudhof TC. A

complexin/synaptotagmin 1 switch controls fast synaptic vesicle exocytosis. Cell.

2006;126(6):1175-87.

357. Seven AB, Brewer KD, Shi L, Jiang QX, and Rizo J. Prevalent mechanism of

membrane bridging by synaptotagmin-1. Proceedings of the National Academy of

Sciences of the United States of America. 2013;110(34):E3243-E52.

Page 250: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

227

358. Xu JJ, Brewer KD, Perez-Castillejos R, and Rizo J. Subtle Interplay between

Synaptotagmin and Complexin Binding to the SNARE Complex. Journal of molecular

biology. 2013;425(18):3461-75.

359. Blagoveshchenskaya AD, Hannah MJ, Allen S, and Cutler DF. Selective and

signal-dependent recruitment of membrane proteins to secretory granules formed by

heterologously expressed von Willebrand factor. Molecular biology of the cell.

2002;13(5):1582-93.

360. Lillicrap D. Syntaxin-binding protein 5 exocytosis regulation: differential role in

endothelial cells and platelets. The Journal of clinical investigation. 2014;124(10):4231-

3.

361. Sudhof TC. The synaptic vesicle cycle. Annu Rev Neurosci. 2004;27(509-47.

362. Paumet F, Le Mao J, Martin S, Galli T, David B, Blank U, and Roa M. Soluble

NSF attachment protein receptors (SNAREs) in RBL-2H3 mast cells: functional role of

syntaxin 4 in exocytosis and identification of a vesicle-associated membrane protein 8-

containing secretory compartment. J Immunol. 2000;164(11):5850-7.

363. Guo Z, Turner C, and Castle D. Relocation of the t-SNARE SNAP-23 from

lamellipodia-like cell surface projections regulates compound exocytosis in mast cells.

Cell. 1998;94(4):537-48.

364. Lippert U, Ferrari DM, and Jahn R. Endobrevin/VAMP8 mediates exocytotic

release of hexosaminidase from rat basophilic leukaemia cells. FEBS Lett.

2007;581(18):3479-84.

365. Sander LE, Frank SP, Bolat S, Blank U, Galli T, Bigalke H, Bischoff SC, and

Lorentz A. Vesicle associated membrane protein (VAMP)-7 and VAMP-8, but not

Page 251: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

228

VAMP-2 or VAMP-3, are required for activation-induced degranulation of mature

human mast cells. Eur J Immunol. 2008;38(3):855-63.

366. Tiwari N, Wang CC, Brochetta C, Ke G, Vita F, Qi Z, Rivera J, Soranzo MR,

Zabucchi G, Hong W, et al. VAMP-8 segregates mast cell-preformed mediator

exocytosis from cytokine trafficking pathways. Blood. 2008;111(7):3665-74.

367. Ye S, Karim ZA, Al Hawas R, Pessin JE, Filipovich AH, and Whiteheart SW.

Syntaxin-11, but not syntaxin-2 or syntaxin-4, is required for platelet secretion. Blood.

2012;120(12):2484-92.

368. Flaumenhaft R, Croce K, Chen E, Furie B, and Furie BC. Proteins of the

exocytotic core complex mediate platelet alpha-granule secretion. Roles of vesicle-

associated membrane protein, SNAP-23, and syntaxin 4. The Journal of biological

chemistry. 1999;274(4):2492-501.

369. Chen D, Lemons PP, Schraw T, and Whiteheart SW. Molecular mechanisms of

platelet exocytosis: role of SNAP-23 and syntaxin 2 and 4 in lysosome release. Blood.

2000;96(5):1782-8.

370. Polgar J, Chung SH, and Reed GL. Vesicle-associated membrane protein 3

(VAMP-3) and VAMP-8 are present in human platelets and are required for granule

secretion. Blood. 2002;100(3):1081-3.

371. Ren Q, Barber HK, Crawford GL, Karim ZA, Zhao C, Choi W, Wang CC, Hong

W, and Whiteheart SW. Endobrevin/VAMP-8 is the primary v-SNARE for the platelet

release reaction. Mol Biol Cell. 2007;18(1):24-33.

Page 252: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

229

372. Graham GJ, Ren Q, Dilks JR, Blair P, Whiteheart SW, and Flaumenhaft R.

Endobrevin/VAMP-8-dependent dense granule release mediates thrombus formation in

vivo. Blood. 2009;114(5):1083-90.

373. Chen D, Bernstein AM, Lemons PP, and Whiteheart SW. Molecular mechanisms

of platelet exocytosis: role of SNAP-23 and syntaxin 2 in dense core granule release.

Blood. 2000;95(3):921-9.

374. Chung SH, Polgar J, and Reed GL. Protein kinase C phosphorylation of syntaxin

4 in thrombin-activated human platelets. The Journal of biological chemistry.

2000;275(33):25286-91.

375. Feng D, Crane K, Rozenvayn N, Dvorak AM, and Flaumenhaft R. Subcellular

distribution of 3 functional platelet SNARE proteins: human cellubrevin, SNAP-23, and

syntaxin 2. Blood. 2002;99(11):4006-14.

376. Flaumenhaft R, Rozenvayn N, Feng D, and Dvorak AM. SNAP-23 and syntaxin-

2 localize to the extracellular surface of the platelet plasma membrane. Blood.

2007;110(5):1492-501.

377. Polgar J, Lane WS, Chung SH, Houng AK, and Reed GL. Phosphorylation of

SNAP-23 in activated human platelets. The Journal of biological chemistry.

2003;278(45):44369-76.

378. Rutledge TW, and Whiteheart SW. SNAP-23 is a target for calpain cleavage in

activated platelets. The Journal of biological chemistry. 2002;277(40):37009-15.

379. Cosen-Binker LI, Binker MG, Wang CC, Hong W, and Gaisano HY. VAMP8 is

the v-SNARE that mediates basolateral exocytosis in a mouse model of alcoholic

pancreatitis. The Journal of clinical investigation. 2008;118(7):2535-51.

Page 253: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

230

380. Wang CC, Ng CP, Lu L, Atlashkin V, Zhang W, Seet LF, and Hong W. A role of

VAMP8/endobrevin in regulated exocytosis of pancreatic acinar cells. Developmental

cell. 2004;7(3):359-71.

381. Kondkar AA, Bray MS, Leal SM, Nagalla S, Liu DJ, Jin Y, Dong JF, Ren Q,

Whiteheart SW, Shaw C, et al. VAMP8/endobrevin is overexpressed in hyperreactive

human platelets: suggested role for platelet microRNA. J Thromb Haemost.

2010;8(2):369-78.

382. Peters CG, Michelson AD, and Flaumenhaft R. Granule exocytosis is required for

platelet spreading: differential sorting of alpha-granules expressing VAMP-7. Blood.

2012;120(1):199-206.

383. Shiffman D, Rowland CM, Louie JZ, Luke MM, Bare LA, Bolonick J, Young

BA, Catanese JJ, Stiggins CF, Pullinger CR, et al. Gene variants of VAMP8 and

HNRPUL1 are associated with early-onset myocardial infarction. Arterioscl Throm Vas.

2006;26(5):E44-E.

384. Gaussem P, Ishida BY, Fontana P, Pullinger CR, Khane JP, Aiach M, Bachelot-

Loza C, and Gandrille S. No influence of the VAMP8 rs1010 single nucleotide

polymorphism on platelet functions in vitro. Journal of cellular and molecular medicine.

2009;13(3):601-3.

385. Fields IC, Shteyn E, Pypaert M, Proux-Gillardeaux V, Kang RS, Galli T, and

Folsch H. v-SNARE cellubrevin is required for basolateral sorting of AP-1 B-dependent

cargo in polarized epithelial cells. J Cell Biol. 2007;177(3):477-88.

386. Kean MJ, Williams KC, Skalski M, Myers D, Burtnik A, Foster D, and Coppolino

MG. VAMP3, syntaxin-13 and SNAP23 are involved in secretion of matrix

Page 254: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

231

metalloproteinases, degradation of the extracellular matrix and cell invasion. J Cell Sci.

2009;122(22):4089-98.

387. Allen LA, Yang C, and Pessin JE. Rate and extent of phagocytosis in

macrophages lacking vamp3. Journal of leukocyte biology. 2002;72(1):217-21.

388. Veale KJ, Offenhauser C, Whittaker SP, Estrella RP, and Murray RZ. Recycling

Endosome Membrane Incorporation into the Leading Edge Regulates Lamellipodia

Formation and Macrophage Migration. Traffic. 2010;11(10):1370-9.

389. Veale KJ, Offenhauser C, Lei N, Stanley AC, Stow JL, and Murray RZ. VAMP3

regulates podosome organisation in macrophages and together with Stx4/SNAP23

mediates adhesion, cell spreading and persistent migration. Experimental cell research.

2011;317(13):1817-29.

390. Olson AL, Knight JB, and Pessin JE. Syntaxin 4, VAMP2, and/or

VAMP3/cellubrevin are functional target membrane and vesicle SNAP receptors for

insulin-stimulated GLUT4 translocation in adipocytes. Molecular and cellular biology.

1997;17(5):2425-35.

391. Regazzi R, Wollheim CB, Lang J, Theler JM, Rossetto O, Montecucco C, Sadoul

K, Weller U, Palmer M, and Thorens B. VAMP-2 and cellubrevin are expressed in

pancreatic beta-cells and are essential for Ca(2+)-but not for GTP gamma S-induced

insulin secretion. The EMBO journal. 1995;14(12):2723-30.

392. Feldmann A, Amphornrat J, Schonherr M, Winterstein C, Mobius W, Ruhwedel

T, Danglot L, Nave KA, Galli T, Bruns D, et al. Transport of the Major Myelin

Proteolipid Protein Is Directed by VAMP3 and VAMP7. J Neurosci. 2011;31(15):5659-

72.

Page 255: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

232

393. Yang C, Mora S, Ryder JW, Coker KJ, Hansen P, Allen LA, and Pessin JE.

VAMP3 null mice display normal constitutive, insulin- and exercise-regulated vesicle

trafficking. Molecular and cellular biology. 2001;21(5):1573-80.

394. Schraw TD, Rutledge TW, Crawford GL, Bernstein AM, Kalen AL, Pessin JE,

and Whiteheart SW. Granule stores from cellubrevin/VAMP-3 null mouse platelets

exhibit normal stimulus-induced release. Blood. 2003;102(5):1716-22.

395. Daro E, van der Sluijs P, Galli T, and Mellman I. Rab4 and cellubrevin define

different early endosome populations on the pathway of transferrin receptor recycling.

Proceedings of the National Academy of Sciences of the United States of America.

1996;93(18):9559-64.

396. Riggs KA, Hasan N, Humphrey D, Raleigh C, Nevitt C, Corbin D, and Hu C.

Regulation of integrin endocytic recycling and chemotactic cell migration by syntaxin 6

and VAMP3 interaction. J Cell Sci. 2012;125(Pt 16):3827-39.

397. Ganley IG, Espinosa E, and Pfeffer SR. A syntaxin 10-SNARE complex

distinguishes two distinct transport routes from endosomes to the trans-Golgi in human

cells. J Cell Biol. 2008;180(1):159-72.

398. Puri C, Renna M, Bento CF, Moreau K, and Rubinsztein DC. Diverse

Autophagosome Membrane Sources Coalesce in Recycling Endosomes. Cell.

2013;154(6):1285-99.

399. Puri C, Renna M, Bento CF, Moreau K, and Rubinsztein DC. ATG16L1 meets

ATG9 in recycling endosomes: additional roles for the plasma membrane and

endocytosis in autophagosome biogenesis. Autophagy. 2014;10(1):182-4.

Page 256: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

233

400. Nightingale TD, White IJ, Doyle EL, Turmaine M, Harrison-Lavoie KJ, Webb

KF, Cramer LP, and Cutler DF. Actomyosin II contractility expels von Willebrand factor

from Weibel-Palade bodies during exocytosis. J Cell Biol. 2011;194(4):613-29.

401. Giraudo CG, Eng WS, Melia TJ, and Rothman JE. A clamping mechanism

involved in SNARE-dependent exocytosis. Science. 2006;313(5787):676-80.

402. Melia TJ, Jr. Putting the clamps on membrane fusion: how complexin sets the

stage for calcium-mediated exocytosis. FEBS Lett. 2007;581(11):2131-9.

403. Yamakuchi M, Greer JJ, Cameron SJ, Matsushita K, Morrell CN, Talbot-Fox K,

Baldwin WM, 3rd, Lefer DJ, and Lowenstein CJ. HMG-CoA reductase inhibitors inhibit

endothelial exocytosis and decrease myocardial infarct size. Circulation research.

2005;96(11):1185-92.

404. Araki S, Tamori Y, Kawanishi M, Shinoda H, Masugi J, Mori H, Niki T,

Okazawa H, Kubota T, and Kasuga M. Inhibition of the binding of SNAP-23 to syntaxin

4 by Munc18c. Biochemical and biophysical research communications. 1997;234(1):257-

62.

405. Suzuki K, and Verma IM. Phosphorylation of SNAP-23 by IkappaB kinase 2

regulates mast cell degranulation. Cell. 2008;134(3):485-95.

406. Salaun C, Gould GW, and Chamberlain LH. The SNARE proteins SNAP-25 and

SNAP-23 display different affinities for lipid rafts in PC12 cells. Regulation by distinct

cysteine-rich domains. The Journal of biological chemistry. 2005;280(2):1236-40.

407. Salaun C, Gould GW, and Chamberlain LH. Lipid raft association of SNARE

proteins regulates exocytosis in PC12 cells. The Journal of biological chemistry.

2005;280(20):19449-53.

Page 257: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

234

408. Grant NJ, Hepp R, Krause W, Aunis D, Oehme P, and Langley K. Differential

expression of SNAP-25 isoforms and SNAP-23 in the adrenal gland. Journal of

neurochemistry. 1999;72(1):363-72.

409. Ravichandran V, Chawla A, and Roche PA. Identification of a novel syntaxin-

and synaptobrevin/VAMP-binding protein, SNAP-23, expressed in non-neuronal tissues.

The Journal of biological chemistry. 1996;271(23):13300-3.

410. Morikawa Y, Nishida H, Misawa K, Nosaka T, Miyajima A, Senba E, and

Kitamura T. Induction of synaptosomal-associated protein-23 kD (SNAP-23) by various

cytokines. Blood. 1998;92(1):129-35.

411. Chen D, and Whiteheart SW. Intracellular localization of SNAP-23 to endosomal

compartments. Biochemical and biophysical research communications. 1999;255(2):340-

6.

412. Reales E, Mora-Lopez F, Rivas V, Garcia-Poley A, Brieva JA, and Campos-Caro

A. Identification of soluble N-ethylmaleimide-sensitive factor attachment protein

receptor exocytotic machinery in human plasma cells: SNAP-23 is essential for antibody

secretion. J Immunol. 2005;175(10):6686-93.

413. Sprenger RR, Fontijn RD, van Marle J, Pannekoek H, and Horrevoets AJG.

Spatial segregation of transport and signalling functions between human endothelial

caveolae and lipid raft proteomes. Biochemical Journal. 2006;400(401-10.

414. Sudhof TC, and Rizo J. Synaptic vesicle exocytosis. Cold Spring Harbor

perspectives in biology. 2011;3(12).

415. Bar-On D, Wolter S, van de Linde S, Heilemann M, Nudelman G, Nachliel E,

Gutman M, Sauer M, and Ashery U. Super-resolution imaging reveals the internal

Page 258: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

235

architecture of nano-sized syntaxin clusters. The Journal of biological chemistry.

2012;287(32):27158-67.

416. Pertsinidis A, Mukherjee K, Sharma M, Pang ZP, Park SR, Zhang Y, Brunger

AT, Sudhof TC, and Chu S. Ultrahigh-resolution imaging reveals formation of neuronal

SNARE/Munc18 complexes in situ. Proceedings of the National Academy of Sciences of

the United States of America. 2013;110(30):E2812-20.

417. Pallavi B, and Nagaraj R. Palmitoylated peptides from the cysteine-rich domain

of SNAP-23 cause membrane fusion depending on peptide length, position of cysteines,

and extent of palmitoylation. The Journal of biological chemistry. 2003;278(15):12737-

44.

418. Fukuda R, McNew JA, Weber T, Parlati F, Engel T, Nickel W, Rothman JE, and

Sollner TH. Functional architecture of an intracellular membrane t-SNARE. Nature.

2000;407(6801):198-202.

419. Wong PP, Daneman N, Volchuk A, Lassam N, Wilson MC, Klip A, and Trimble

WS. Tissue distribution of SNAP-23 and its subcellular localization in 3T3-L1 cells.

Biochemical and biophysical research communications. 1997;230(1):64-8.

420. Adler KB, Tuvim MJ, and Dickey BF. Regulated mucin secretion from airway

epithelial cells. Frontiers in endocrinology. 2013;4(129).

421. Abonyo BO, Gou D, Wang P, Narasaraju T, Wang Z, and Liu L. Syntaxin 2 and

SNAP-23 are required for regulated surfactant secretion. Biochemistry.

2004;43(12):3499-506.

Page 259: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

236

422. Chintagari NR, Jin N, Wang P, Narasaraju TA, Chen J, and Liu L. Effect of

cholesterol depletion on exocytosis of alveolar type II cells. American journal of

respiratory cell and molecular biology. 2006;34(6):677-87.

423. Gerelsaikhan T, Vasa PK, and Chander A. Annexin A7 and SNAP23 interactions

in alveolar type II cells and in vitro: a role for Ca(2+) and PKC. Biochimica et biophysica

acta. 2012;1823(10):1796-806.

424. Sakurai C, Hashimoto H, Nakanishi H, Arai S, Wada Y, Sun-Wada GH, Wada I,

and Hatsuzawa K. SNAP-23 regulates phagosome formation and maturation in

macrophages. Mol Biol Cell. 2012;23(24):4849-63.

425. Martin-Martin B, Nabokina SM, Blasi J, Lazo PA, and Mollinedo F. Involvement

of SNAP-23 and syntaxin 6 in human neutrophil exocytosis. Blood. 2000;96(7):2574-83.

426. Gomez-Jaramillo L, Delgado-Perez L, Reales E, Mora-Lopez F, Mateos RM,

Garcia-Poley A, Brieva JA, and Campos-Caro A. Syntaxin-4 is implicated in the

secretion of antibodies by human plasma cells. Journal of leukocyte biology.

2014;95(2):305-12.

427. Logan MR, Lacy P, Bablitz B, and Moqbel R. Expression of eosinophil target

SNAREs as potential cognate receptors for vesicle-associated membrane protein-2 in

exocytosis. J Allergy Clin Immun. 2002;109(2):299-306.

428. Shukla A, Corydon TJ, Nielsen S, Hoffmann HJ, and Dahl R. Identification of

three new splice variants of the SNARE protein SNAP-23. Biochemical and biophysical

research communications. 2001;285(2):320-7.

Page 260: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

237

429. Gupta RM, and Musunuru K. Expanding the genetic editing tool kit: ZFNs,

TALENs, and CRISPR-Cas9. The Journal of clinical investigation. 2014;124(10):4154-

61.

430. Hsu PD, Lander ES, and Zhang F. Development and applications of CRISPR-

Cas9 for genome engineering. Cell. 2014;157(6):1262-78.

431. Wang H, Yang H, Shivalila CS, Dawlaty MM, Cheng AW, Zhang F, and Jaenisch

R. One-step generation of mice carrying mutations in multiple genes by CRISPR/Cas-

mediated genome engineering. Cell. 2013;153(4):910-8.

432. Yang H, Wang H, and Jaenisch R. Generating genetically modified mice using

CRISPR/Cas-mediated genome engineering. Nature protocols. 2014;9(8):1956-68.

433. Ye S, Huang Y, Joshi S, Zhang J, Yang F, Zhang G, Smyth SS, Li Z, Takai Y,

and Whiteheart SW. Platelet secretion and hemostasis require syntaxin-binding protein

STXBP5. The Journal of clinical investigation. 2014;124(10):4517-28.

434. Trimbuch T, Xu JJ, Flaherty D, Tomchick DR, Rizo J, and Rosenmund C. Re-

examining how Complexin Inhibits Neurotransmitter Release: SNARE complex Insertion

or Electrostatic Hindrance? eLife. 2014;3

435. Lai Y, Diao JJ, Cipriano DJ, Zhang YX, Pfuetzner RA, Padolina MS, and

Brunger AT. Complexin inhibits spontaneous release and synchronizes Ca2+-triggered

synaptic vesicle fusion by distinct mechanisms. eLife. 2014;3

436. Li Y, Augustine GJ, and Weninger K. Kinetics of complexin binding to the

SNARE complex: correcting single molecule FRET measurements for hidden events.

Biophysical journal. 2007;93(6):2178-87.

Page 261: The Role of STXBP5, VAMP8, and SNAP23 in Endothelial

238

437. Pabst S, Margittai M, Vainius D, Langen R, Jahn R, and Fasshauer D. Rapid and

selective binding to the synaptic SNARE complex suggests a modulatory role of

complexins in neuroexocytosis. The Journal of biological chemistry. 2002;277(10):7838-

48.

438. Lillicrap D. von Willebrand disease: advances in pathogenetic understanding,

diagnosis, and therapy. Blood. 2013;122(23):3735-40.

439. LoMonaco MB, and Lowenstein CJ. Enhanced assay of endothelial exocytosis

using extracellular matrix components. Anal Biochem. 2014;452(19-24).

440. Dayananda KM, Gogia S, and Neelamegham S. Escherichia coli-derived von

Willebrand factor-A2 domain fluorescence/Forster resonance energy transfer proteins

that quantify ADAMTS13 activity. Analytical biochemistry. 2011;410(2):206-13.

441. Morrell CN, Sun H, Ikeda M, Beique JC, Swaim AM, Mason E, Martin T,

Thompson LE, Gozen O, Ampagoomian D, et al. Glutamate mediates platelet activation

through the AMPA receptor. Journal of Experimental Medicine. 2008;205(3):575-84.

442. Owens AP, 3rd, Lu Y, Whinna HC, Gachet C, Fay WP, and Mackman N.

Towards a standardization of the murine ferric chloride-induced carotid arterial

thrombosis model. Journal of thrombosis and haemostasis : JTH. 2011;9(9):1862-3.

443. Eckly A, Hechler B, Freund M, Zerr M, Cazenave JP, Lanza F, Mangin PH, and

Gachet C. Mechanisms underlying FeCl3-induced arterial thrombosis. Journal of

thrombosis and haemostasis : JTH. 2011;9(4):779-89.

444. Manders E, Verbeek F, and Aten J. Measurement of co-localization of objects in

dual color confocal images. J Microsc. 1993;169(37).