<<

DEFINING THE ROLE OF HYPOXIA AND HIFs IN VASCULAR CELLS

by

ANNA CLARK HENRY BORTON

Submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy

Department of Pathology

CASE WESTERN RESERVE UNIVERSITY

August, 2018

CASE WESTERN RESERVE UNIVERSITY

SCHOOL OF GRADUATE STUDIES

We hereby approve the thesis/dissertation of

Anna Clark Henry Borton

candidate for the PhD Degree*.

Committee Chair

Nicholas P. Ziats, Ph.D.

Committee Members

Diana L. Ramirez-Bergeron, Ph.D.

Aaron Proweller, M.D., Ph.D.

George R. Dubyak, Ph.D.

Clive R. Hamlin, Ph.D.

Date of Defense

June 25, 2018

*We also certify that written approval has been obtained

for any proprietary material contained therein.

2

Table of Contents

List of Tables……………………………………………………………………. 7

List of Figures…………………………………………………………………… 8

Acknowledgements…………………………………………………………….. 10

List of Abbreviations……………………………………………………………. 12

Abstract………………………………………………………………………….. 16

Chapter 1: Introduction…………………………………………………...... 18

Vascular Smooth Muscle ……………………………………………… 18

Structure of the vascular wall.....……………………………… 18

Diverse developmental origins of VSMCs……………………. 20

Vascular smooth muscle contractile function………………... 22

Phenotypic plasticity in VSMCs……………………………….. 27

Hypoxia-Inducible Factors……………………………………………... 29

Oxygen dependent regulation of HIFs………………………... 30

Alternative HIF stabilization pathways………………………... 32

HIF function in the vasculature………………………………... 33

HIF in pulmonary VSMCs……………………………………… 33

HIF in VSMCs of the systemic vasculature………………….. 35

Peripheral Vascular Disease…………………………………………... 37

Epidemiology and disease burden……………………………. 38

Clinical presentation and diagnosis…………………………… 38

Pathogenesis and vascular compensation…………………... 40

Therapeutic interventions……………………………………… 44

3

HIF in VSMCs of the peripheral vasculature………………………… 45

A murine model of PVD: HLI…………………………………... 45

HIFs in HLI model………………………………………………. 46

HIF as a therapeutic……………………………………………. 48

Summary and Hypothesis……………………………………………… 48

Chapter 2: Aryl Hydrocarbon Receptor Nuclear Translocator in

Vascular Smooth Muscle Cells is Required for Optimal Peripheral

Perfusion Recovery…………………………………………………………… 50

Authors…………………………………………………………………… 50

Summary………………………………………………………………… 51

Introduction……………………………………………………………… 53

Methods………………………………………………………………….. 56

Results…………………………………………………………………… 66

Generation of smooth muscle specific Arnt knockout mouse

model…………………………………………………………….. 66

ArntSMKO mice show impaired flow recovery in HLI

model…………………………………………………………….. 68

Limb ischemia stimulates collateral remodeling in ArntSMKO

and Arntlox/lox mice………………………………………………. 70

Early angiogenesis does not depend on VSMC

Arnt in HLI……………………………………………………….. 73

ArntSMKO mice show increased hypoxia and damage in

gastrocnemius in response to ischemia……………………… 81

4

Altered vascular smooth muscle morphology in ArntSMKO

mice………………………………………………………………. 84

Loss of ARNT alters VSMC phenotype………………………. 87

Discussion……………………………………………………………….. 91

Chapter 3 Loss of Vascular Smooth Aryl Hydrocarbon

Receptor Nuclear Translocator Impairs …………….. 97

Authors…………………………………………………………………… 97

Summary………………………………………………………………… 98

Introduction……………………………………………………………… 99

Methods………………………………………………………………….. 101

Results…………………………………………………………………… 105

Transcriptional expression of contractile elements are

reduced in VSMCs lacking ARNT…………………………….. 105

Large vessel structure and thickness is not

affected in ArntSMKO…………………………………………….. 107

Loss of Arnt in VSMCs impairs aortic vasoconstriction…….. 107

Loss of Arnt in VSMCs does not affect aortic vasorelaxation 109

Discussion……………………………………………………………….. 112

Conclusions……………………………………………………………… 115

Chapter 4: Discussion and Closing …………………………………...... 116

Discussion……………………………………………………………….. 116

Molecular dysregulation………………………………………... 116

5

Implications for other vascular pathologies………………….. 123

Potential for translation to human disease…………………… 129

Closing………………………….………………………………………... 135

Future Directions………………………………………………………... 136

References……………………………………………………………………... 139

6

List of Tables

Supplemental Table 1: qPCR primers………………………………………... 64

7

List of Figures

Figure 1.1. Layers of the vascular wall……………………………………….. 19

Figure 1.2. VSMCs and ECs of the vascular wall………………………..…. 21

Figure 1.3. VSMC components………………………………… 23

Figure 1.4. Regulation of VSMC contraction and relaxation……………….. 25

Figure 1.5. Canonical HIF regulation…………………………………………. 31

Figure 1.6. Vascular responses to flow limitation…………………………… 43

Figure 1.7. Hind limb ischemia: a murine model of PAD………………...... 47

Figure 2.1. Characterization of ArntSMKO mice……………………………….. 67

Figure 2.2. Bulk perfusion and functional recovery is reduced in ArntSMKO mice following femoral ligation…………………………………………. 69

Figure 2.3. VSMC ARNT is not required for ischemia induced collateralization………………………………………………………………….. 71

Supplemental Figure 2.1. Collateral vessel lumen cross sectional area vs limb perfusion…………………………………………………………………… 72

Supplemental Figure 2.2. Proximal HLI model………………………………. 75

Figure 2.4. Histological assessment of capillary number and perfusion status in gastrocnemius muscle (GC)………………………………………… 76

Supplemental Figure 2.3. Capillary density and smooth muscle cell colocalization in gastrocnemius (GC) at day 7………………………………. 78

Supplemental Figure 2.4. Regional assessment of capillary density and perfusion status in gastrocnemius muscle (GC)…………………………….. 79

8

Figure 2.5. Ischemic regeneration and vessel integrity are impaired in ArntSMKO mice…………………………………………………. 83

Figure 2.6. Smooth muscle morphology and perimural wrapping of small …………………………………………………………………………. 85

Supplemental Figure 2.5. Additional images of arterioles in skeletal muscle……………………………………………………………………………. 86

Figure 2.7. Transcriptional expression of proliferation and migration regulators in response to hypoxia…………………………………………….. 89

Figure 2.8. Assessment of VSMC phenotype in vitro………………………. 90

Figure 3.1. Contractile gene expression is reduced in ArntSMKO VSMCs…. 106

Figure 3.2. Large vessel structure unchanged in ArntSMKO………………… 108

Figure 3.3. ArntSMKO vessels have impaired vasoconstriction……………… 110

Figure 3.4. Vasorelaxation is intact in ArntSMKO……………………………... 111

Figure 4.1. Effects of hypoxia exposure on human iliac artery VSMCs…... 130

Figure 4.2. Effects of hypoxia exposure on human iliac VSMCs…….. 133

Figure 4.3. Iliac vein smooth muscle cells are more susceptible to H2O2 induced apoptosis………………………………………………………………. 134

9

Acknowledgements

Numerous people have helped me reach this milestone, and for each of their

contributions, I am tremendously grateful.

It has been an outstanding privilege to train under Diana Ramirez-Bergeron. Her

enthusiasm and love of science are positively infectious, as is the commitment to

excellence, scientific rigor, and collaborative spirit she instills in her trainees. Her

mentorship inspired me to step outside my academic comfort zone, encouraged

me through challenges, and helped me develop skills I will use for the rest of my

career.

My thesis project, spanned the expertise of the Ramirez and Proweller

laboratories. I am incredibly grateful to Aaron Proweller for sharing his scientific

guidance, technical expertise, and perspective as a physician scientist.

Enormous thanks also to my thesis committee members, Nicholas Ziats, George

Dubyak, and Clive Hamlin for all their scientific insight and advice.

Science does not happen in a vacuum: I am grateful to have had the opportunity to work with and learn from all the past and present members of the Ramirez and

Proweller laboratories.

I would also like to thank Mukesh Jain, Xudong Liao, Lalitha Nayak, Yuan Lu,

Andrei Maiseyeu, Anne Hamik, Alex Huang, Rongli Zhang, Bryan Benson, Lee

Neilson, Iulia Barbur, Stephanie Lapping, Amer Alaiti, and Keiki Sugi for their technical assistance, professional and scientific advice, goodwill, and encouragement.

10

I have been fortunate to have a wonderful community of peers including the MSTP entering classes of 2011 and 2012 and my CVRI graduate student compatriots

Nelson, David, and Liyan.

I am grateful to the MSTP, our director Cliff Harding, the co-directors, and the administrative team for the opportunity to chase the dream of a career as a physician scientist. Thanks also to the CVRI and the Departments of Pathology and Medicine and their administrators for being my academic home during the

PhD.

Overwhelming thank yous are also due to my friends and extended family for graciously listening to the minutia of experimental success and failure, and cheering me through the celebratory moments and the frustrations.

I am so grateful to my family for fostering curiosity and dedication and for their love, support, and encouragement. Finally, a tremendous thank you to my wonderful husband Peter. He has been there every step of the way as my greatest supporter and the baker of thesis committee appeasing banana bread.

11

List of Abbreviations

ABI – Ankle-brachial index

ACC – American College of Cardiology Foundation

Ach –

AHA – American Association

AHR – Aryl hydrocarbon receptor

ANG1 – Angiopoietin

AngII – Angiotensin II

ARNT; HIF-1β – Aryl hydrocarbon receptor nuclear translocator

Arntlox/lox – Animals and cells with floxed ARNT gene

ArntSMKO – Smooth muscle specific ARNT deletion model

AT1 – Angiotensin receptor 1

BSA – Bovine serum albumin

Ca – Calcium

CaCm – Calcium

CLI – Critical limb ischemia

Cnn1 – Calponin

DMOG – Dimethyloxalylglycine

DN- HIF-1α – Dominant negative HIF-1α

12

EC – Endothelial cell

EDHF – -derived hyperpolarizing factors

EEL – External elastic lamina

eNOS – Endothelial synthase

ET-1 – Endothelin receptor 1

FGF2 – Fibroblast growth factor 2

FIH – Factor inhibiting HIF

GC – Gastrocnemius

Glut1 – Glucose transporter 1

HIFs – Hypoxia-inducible factors

HLI – Hind limb ischemia

HRE – Hypoxia response element

HXK2 – Hexokinase 2

IEL – Internal elastic lamina

IP – Intraperitoneal

IV – Intravenous

KLF4 – Krüppel-like factor 4

KO – Knockout

13

MAPK – Mitogen activated protein kinase

MEJ – Myoendothelial junctions

MLC; MYL9 – Regulatory light chain

MLCK – Myosin light chain kinase

MLCP – Myosin light chain phosphatase

MMPs – Matrix metalloproteinases

O2 –

OCT4 – Octamer-binding transcription factor 4

PAD – Peripheral arterial disease

PAH – Pulmonary artery

PAI-1 – Plasminogen activator inhibitor-1

PAS – Per-ARNT-Sim

PASMC – Pulmonary artery smooth muscle cells

PDGF-BB – Platelet derived growth factor BB

PE – Phenylephrine

PGI2 – Prostaglandin I2

PHD – Prolyl hydroxylase domain containing enzymes

PVD – Peripheral vascular disease

14

ROCK – Rho-associated protein kinase

ROS – Reactive oxygen species

Sca-1 – Stem cells antigen-1

SMA; Acta2 – Alpha smooth muscle

SMM – Smooth muscle myosin

SMMHC; MYH11 – Smooth muscle myosin heavy chain

SNP – Sodium nitroprusside

SRF – Serum response factor

THBS-1 – Thrombospondin 1

Thbs2 – Thrombospondin 2

Timp1 – TIMP metallopeptidase inhibitor 1

TNF-α – Tumor necrosis factor alpha

VEGFA – Vascular endothelial growth factor A

VEGFR-1 – Vascular endothelial growth factor receptor 1

VHL – von hippel-lindau protein

VSMCs – Vascular smooth muscle cells

WT – Wild type

15

Defining the Role of Hypoxia and HIFs in Vascular Smooth Muscle Cells

Abstract

by

ANNA CLARK HENRY BORTON

Vascular diseases are major causes of morbidity and mortality. Vascular smooth muscle cells (VSMCs), as integral constituents of the vascular wall, provide structural support and dynamically alter vessel shape and structure in response to environmental stimuli. While the role of their endothelial neighbors has been extensively examined in facilitation of ischemic responses and vascular remodeling, less is known about VSMCs. Hypoxia-inducible factors (HIFs) are master regulators of cellular responses to O2 tension. These heterodimeric transcription factors drive endothelial vascularization and are putative regulators of VSMC driven vascular remodeling in pulmonary artery hypertension. The studies presented in this dissertation examine the role of canonical HIF activity in modulating VSMC phenotype, vasoreactivity, and responses to peripheral ischemia. Mice with smooth muscle specific deletion of aryl hydrocarbon receptor nuclear translocator (ARNT; HIF-1β) (ArntSMKO), required for HIF transcriptional activity, feature impaired peripheral perfusion recovery following femoral artery ligation. Vessels from ArntSMKO mice display altered vasomotor function, increased hypoxic microvascular permeability, and disrupted arteriolar VSMC morphology.

Aberrant behavior and transcriptional profile in isolated ArntSMKO VSMCs indicate

16 phenotypic switching. Collectively, the data support a role for ARNT regulating

VSMC phenotype, essential for vasoreactivity and perfusion recovery in peripheral ischemia. As such, ARNT provides a therapeutic target for modulating VSMC phenotypic change, a feature of many vascular diseases.

17

Chapter 1: Introduction

In vertebrates, the vasculature is critical to sustaining life. Every cell in the body

resides no more than two hundred microns away from the nearest vessel1. At a

very simple level, blood vessels are conduits for the provision of nutrients and

oxygen and the removal of waste, but they also aid in endocrine signal delivery

and provide a network for the rapid transit of migratory cells from to tissue.

Structure of the vascular wall

The vessel wall is composed of three layers, the intima, media, and adventitia

(Figure 1.1) 2.The innermost layer lining the vessel lumen is the intima. It is composed of a single layer of endothelial cells (ECs) that interact directly with blood and provide selectively permeable barrier function. Just deep to the intima, the internal elastic lamina marks the inner boundary of the tunica media. The media, much thicker than the intima especially in , is formed by smooth muscle cells and encircling the vessel in circumferential layers outward to the external elastic lamina. Vascular smooth muscle cells (VSMCs), the subject of this dissertation, provide structural support to the vessel, regulate the lumen diameter, and are integral to vessel physiology. Outside the external elastic lamina the tunica adventitia is composed primarily of connective tissue 3. In large

vessels, the adventitia may contain smaller neurovascular bundles. The vessels

within these bundles, namely vasovasorum, supply nutrients to the outer layers of the vessel wall too far from the lumen to be adequately supplied by diffusion 3. The

relative positions of the mural layers can be observed in Figure 1.2, a cross section

18

Figure 1.1 Layers of the vessel wall. The vascular wall can be divided into three distinct layers, the intima, the media, and the adventitia.

19

of popliteal artery, where the CD31+ endothelial cells of the intima are visible in

green and the α-smooth muscle actin+ (SMA) VSMCs of the medial layer are identified in red. Differences in tunica media thickness in the artery and vein are also apparent in this micrograph.

Diverse developmental origins of VSMCs

VSMCs can be seen wrapped around nearly every larger than

. While they are typically referred to as a singular tissue type, the

developmental origins of these cells are diverse 4. Indeed, in the vertebrate

, VSMCs are derived from at least 7 unique non-overlapping sources 5. The

majority of VSMCs differentiate from various mesodermal fields, though the

ectodermal neural crest population also participates. The contributors to the

are an excellent example of this diversity. The root of the aorta, where it exits the

left , is formed from cells of the second heart field 6. Neighboring VSMCs

in the ascending aortic arch and pulmonic trunk are derived from neural crest cells

having migrated toward the developing heart 7, 8. As the aorta begins its descent

through the thorax, individual mesodermal that also form each vertebral

segment, contribute aortic VSMCs at their corresponding vertebral levels 9, 10.

Continuing distally to the abdominal aorta and vessels of the lower limbs reveals

VSMCs differentiated from the splanchnopleuric layer of the lateral plate

mesoderm 11. In total, VSMCs that wrap the aorta have at least 4 separate origins,

counting the somites collectively. Vessels and the VSMCs that invest them also

develop alongside the they perfuse. This pattern can be seen in pulmonary

20

Figure 1.2 VSMCs and ECs of the vascular wall. Micrograph of murine popliteal artery and vein cross section. CD31+ ECs of the tunica intima are labeled in green and SMA+ VSMCs of the tunica media are visible in red. The artery (right) features a thicker VSMC layer than the vein (left) supporting the open lumen.

21

VSMCs that differentiate from lung mesoderm, and in coronary VSMCs contributed by the pro epicardium 12,13, 14. The maintenance of VSMCs in the vascular wall is also supported by local populations. A population of resident progenitors, dubbed the medial side population, has been described within the tunica media 15 while a separate population of stem cells antigen-1 (Sca-1)+ cells in the adventitia can also differentiate into VSMCs when stimulated with PDGF-BB 16. These Sca-1+ cells are not committed along the VSMC lineage and can become endothelial cells in the presence of VEGFA 16. The diverse origins of VSMCs contribute to their tissue specific responses in vivo and in vitro, yet their expression of similar makers and overall function within the vessel wall underlie their classification as a single tissue type.

Vascular smooth muscle contractile function

A unifying characteristic of VSMCs is their contractile ability. Force generation, as in other muscle cells, is achieved by cyclical cross bridging of actin and myosin

(Figure 1.3) 17. In vascular smooth muscle, the primary myofilament interaction occurs between SMA and smooth muscle myosin (SMM). SMA is abundant in

VSMCs accounting for ~40% of total cellular protein 18. SMM is a type two myosin consisting of two intertwined heavy chains (SMMHC) and four light chains: two regulatory light chains (20kDa; MYL9, MLC herein) and two essential light chains

(17kDa). The structure of SMM is similar to in striated muscle 19.The two sets of light chains associate with the neck region of the heavy chains. Unlike other muscle cells, SMA and SMM are not arranged in . Instead,

22

Figure 1.3. VSMC myofilament components. Actin and Myosin interact to generate contractile force in VSMCs. These interactions are physically regulated by MLC, calponin, , and caldesmon, among other proteins.

23

the number and structure of the myofilaments are dynamic 20. Indeed, studies show

increases in polymerized actin in response to stimulation by contractile agonist and

that short term inhibition of actin polymerization reduces contractile force 21, 22.

Regulation of contraction centers on the phosphorylation status of the regulatory

MLC as determined by opposing regulatory enzymes, myosin light chain kinase

(MLCK) and myosin light chain phosphatase (MLCP) 23. Phosphorylation of MLC

by MCLK releases the myosin head to interact with actin and increase contraction,

while dephosphorylation by MLCP promotes relaxation. Many upstream factors

and second messengers further regulate the function of these enzymes (Figure

1.4).

Depolarization of the cell opens L type calcium channels increasing the

concentration of cytosolic free calcium (Ca) though influx of extracellular Ca and

release of intracellular Ca reservoirs in the 24. Increased

calcium activates calcium calmodulin (CaCm), which subsequently activates

MCLK. MLCK in turn phosphorylates MLC promoting constriction. In addition to

direct neuromodulation of the membrane potential, increased intraluminal tension

can stimulate myogenic vasoconstriction as initially described by Bayliss in the

early 1900s 25. L type calcium channels also participate as mediators of myogenic tone 26. Contraction can also be stimulated by receptor dependent vasoconstrictors

which activate G protein coupled receptors and, via signaling cascades, converge

to increase activity of MLCK and/or decrease activity of MLCP 24, 27.

24

Figure 1.4. Regulation of VSMC contraction and relaxation. Signaling, including from a variety of G protein coupled receptors and ion channels, converges on enzymes, namely MLCK and MLCP, which regulate phosphorylation and de-phosphorylation of MLC to control myosin activity and thus contraction. Pathways colored green promote contraction while pathways colored red inhibit contraction and promote relaxation.

25

Angiotensin II (AngII), a key factor in renin-angiotensin mediated hypertension,

28 binds to the angiotensin receptor 1 (AT1) on the cell surface . AT1, a Gq coupled receptor increases intracellular IP3 and Rho-associated protein kinase (ROCK).

IP3 facilitates calcium release from the sarcoplasmic reticulum thus increasing

MLCK activity through CaCm. ROCK in contrast inhibits MLCP activity to further increase the population of phosphorylated MLC and intensify contraction. Other vasoconstrictors that act through Gq coupled receptors include and phenylephrine (PE) which bind α1-adrenergic receptors, endothelin which binds endothelin ETA receptor, vasopressin which binds vasopressin 1 receptor, and serotonin which binds 5-HT2 receptor. Adrenergic stimulation also acts synergistically through α2-adrenergic receptors, which are Gi G-protein coupled receptors and whose activity decreases cAMP to further disinhibit MLCK.

Vasorelaxation is regulated by NO, prostaglandin (PGI2), and other factors collectively known as endothelium-derived hyperpolarizing factors (EDHF). NO in the smooth muscle increases cGMP activating MLCP to dephosphorylate MLC thus decreasing contractile force. The source of NO is thought to be endothelial cells where stimulation of muscarinic receptors by acetylcholine activates endothelial nitric oxide synthase (eNOS) to produce NO 29, 30. Vasorelaxation can also be achieved by activation of Gs coupled receptors that increase cAMP to inhibit MLCK. This pathway is activated by prostaglandin binding to IP, epinephrine binding to β2 adrenergic receptors, and adenosine binding to A2. The primary

26

relaxing substance differs by artery size, with NO as the primary mediator of large

artery relaxation and EHDF modalities more common in smaller vessels 31, 32.

Phenotypic plasticity in VSMCs

Mature VSMCs in the vascular wall typically exhibit a contractile phenotype and

rarely divide; however, they retain a considerable amount of phenotypic plasticity.

When stressed these cells can take on a more synthetic phenotype with increased

proliferation, migration, and secreted protein production. Markers of VSMC

contractile phenotype include many contractile apparatus proteins the most

prominent of which are, SMA33, 34, SMMHC35, calponin36, 37, and SM22α36, 38.

Correspondingly, a shift away from contractile phenotype is marked by reduced

expression of those same myofilaments and others 39. Presence of a synthetic

phenotype in VSMCs is a feature in many pathologies, including pulmonary

hypertension, systemic hypertension, , graft stenosis, aneurysm,

and dissection 39, 40.

Through the study of these pathologies and VSMCs in culture, multiple factors

have been identified as initiators or mediators of phenotypic changes. Indeed,

VSMC phenotype as identified by expression of contractile marker genes can be

influenced by mechanical forces, contractile agonists, extracellular matrix components such as laminin, elastin, and , neuronal factors, reactive oxygen species, oxygen homeostasis, endothelial-VSMC interactions, and growth factors including, PDGF-BB, thrombin, TGF-B1, and FGF-2 39. PDGF-BB is a

27

particularly good example of a driver of synthetic phenotype as it down regulates

expression of nearly every VSMC marker gene and promotes increased

proliferation and migration of VSMCs 41-45.

In searching for a master regulator of VSMC phenotype, Wang et. al. identified myocardin, which is exclusively expressed in VSMCs and cardiomyocytes 46. At the molecular level, it is a transcriptional co-activator that cooperates with the ubiquitously expressed transcription factor serum response factor (SRF) to promote VSMC development and contractile phenotype. CArG elements which allow for SRF binding and thus regulation have been identified in the promoter regions of many genes that mark smooth muscle identity including: SMA,

SMMHC, SM22α, calponin, smoothelin, caldesmon, and telokin among others 47.

Indeed, inhibition of myocardin through use of a dominant negative myocardin or by siRNA mediated suppression decreased the expression of VSMC marker genes

48, 49. However, not all marker genes are activated by myocardin, suggesting that other regulatory pathways also contribute to phenotypic determination 50.

Many well-known transcription factors have been implicated as contributors to

VSMC phenotype modulation. Krüppel-like factor 4 (KLF4) and octamer-binding

transcription factor 4 (OCT4), key regulators of embryonic stem cell differentiation,

are among that group. OCT4 expression is increased in synthetic VSMCs within

atherosclerotic plaques, and cultures of VSMCs with lentiviral overexpression of

OCT4 feature increased migration though proliferation is reduced 51. KLF4 levels

28

are also increased in VSMCs associated with atherosclerotic plaques and vascular

injury; however the effects of its induction are less clear. One study demonstrated

reduced proliferation attributable to KLF4 expression 52 while several more identify

KLF4 as a driver of synthetic phenotypes with the ability to repress myocardin

activity and contractile gene expression 53, 54. Yes-associated protein 1 (YAP1) of the Hippo-YAP/TAZ pathway is also increased by PDGF-BB stimulation and promotes increased proliferation 55. This is particularly interesting as YAP is known to be responsive to mechanical forces in ECs, specifically having increased expression in areas of disturbed blood flow 56. The Notch family, well known for

mediators of direct cell to cell signaling and cell fate decision making, has been

implicated in promoting both the contractile and synthetic phenotypes. Direct

action by intracellular Notch in cooperation with myocardin/SRF promotes SMA

expression while increased expression of Notch target genes, HEY1 and HEY2, support a synthetic phenotype either by inhibiting myocardin/SRF binding or through an independent pathway 57-59. Furthermore the receptor for mitogen

PDGF-BB, namely PDGFR-β, is also an independent Notch target 60. Finally,

hypoxia driven activation of hypoxia-inducible factors (HIFs) play a role in VSMC

phenotypic modulation in the pulmonary circulation and aorta, and as they are the

topic of this dissertation project are discussed at length below.

Hypoxia-Inducible Factors

Tissues require oxygen (O2) for survival. Hypoxia occurs when the O2 supply is

below the tissue demand. Hypoxia-inducible factors are activated by low oxygen

29

and are master regulators of cellular responses to hypoxia. HIF-1 was initially

discovered as a driver of hypoxia induced erythropoietin expression and has since

been shown to be involved in hypoxia driven gene expression changes in multiple

cell types 61-63. In the subsequent decade, additional family members HIF-2 and

HIF-3 were also identified 64, 65.

Oxygen dependent regulation of HIFs

HIFs are heterodimers formed by the complexation of a HIF-α subunit with aryl

hydrocarbon nuclear translocator (ARNT; HIF-1β). Both the HIF-α subunits and

ARNT are basic helix-loop-helix proteins that contain Per-ARNT-Sim (PAS)

domains. ARNT and HIF-1α are ubiquitously expressed while HIF-2α and -3α have

more tissue restricted expression patterns 66. HIF-α subunits are the limiting factors of HIF activity and are rarely detectable in normoxia due to post translational regulation illustrated in Figure 1.5 and reviewed by Giaccia et al 67. Under normoxic

conditions HIF-1α is hydroxylated at prolyl residues Pro 402 and Pro 564 by

oxygen dependent prolyl hydroxylase domain containing enzymes (PHDs), most

prominently PHD2. Hydroxylated HIF-αs are ubiquitinated through a von Hippel-

Lindau protein (VHL) dependent pathway and targeted to the proteasome for

degradation. HIF-1’s transcriptional activity can also be inhibited through

hydroxylation of an asparagine residue on the C-terminal transactivation domain

of HIF-1α by an oxygen dependent asparagine hydroxylase enzyme also known

as factor inhibiting HIF (FIH) that prevents complex formation with transcriptional

binding partners in the nucleus 68. Under hypoxic conditions, PHD enzymes are

30

Figure 1.5. Canonical HIF regulation. In normoxia, HIF-α is hydroxylated and degraded. In hypoxia, PHD is inhibited and HIF-α accumulates in the cell. It binds ARNT to form HIF and complexes with CBP/P300 to alter the transcription of a variety of genes regulated by HREs including those involved with vascular responses.

31

inhibited and the HIF-1α subunit is stabilized in the non-hydroxylated form allowing

it to accumulate in the cell and dimerize with ARNT. In the nucleus, HIF-1

complexes with CBP/ P300 and binds to hypoxia response elements (HRE) found

in the regulatory elements of genes. HIF-1 individually has been implicated in the

transcriptional regulation of hundreds of genes.

Alternative HIF stabilization pathways

In addition to hypoxic HIF-1α accumulation, there is evidence that HIF-1α plays a

role in mediating the intracellular effects of growth factors and mechanical

stimulation even in normoxic conditions. Cytokines implicated in the regulation of

cell growth including tumor necrosis factor alpha (TNF-α), PDGF, AngII, thrombin,

endothelin-1 (ET-1), and fibroblast growth factor 2 (FGF2) have been shown to

increase HIF-1α protein levels in VSMCs independent of the oxygen status. Major

signaling pathways play a role in stabilization, as evidenced by thrombin, PDGF-

BB, AngII and others signaling through mitogen activated protein kinase (MAPK)

and increased reactive oxygen species (ROS) to trigger HIF-1α accumulation. ET-

1 may rely on changes in calcium in addition to the previously listed pathways 69.

Many of the growth factors implicated are also known to be regulated by HRE containing promoters. Thus the activation of the HIF-1 pathway can establish a positive feedback system in which growth factors activate HIF-1 inducing growth factor production and release that in turn further increases HIF-1α stabilization.

Experiments conducted by Black et al. support this pattern of regulation in the

FGF2/HIF axis 70. Cyclic stretch has also been shown to stabilize HIF-1α through

32

MAPK signaling 71. The diversity of mechanisms involving HIF-1 effects supports

its importance in regulating cellular function and response to environmental

changes.

HIF function in the vasculature

HIF-1 is essential for vascular development. As grow they become too

large to receive oxygen by diffusion and the developing

ultimately alleviates this relative hypoxia. Correspondingly, global Hif-1α or Arnt

knockouts die in utero by 11 days gestation and display malformations of the heart

and vasculature and decreased erythropoiesis 72-74. Genes regulated by HREs include many that have been implicated in vascular growth and remodeling including those that contribute to endothelial cell proliferation and migration, circulating progenitor cell homing, and differentiation, and vascular smooth muscle proliferation and migration 75.

HIF in pulmonary VSMCs

The importance of HIF in the pulmonary smooth muscle has been demonstrated

in a mouse model of chronic hypoxia where mice are placed in conditions of 10%

O2 in inhaled air to induce pulmonary artery hypertension (PAH). Smooth muscle

specific Hif-1α knockouts experienced less pulmonary vascular remodeling and

hypertension than their wild type (WT) littermates when treated with low levels of

76, 77 inhaled O2 . Furthermore, pharmacologic HIF-1 pathway inhibition has

reduced pathologic PAH remodeling in the same model 78. Examining isolated

33

VSMCs, Pak and colleagues reviewed the available studies of pulmonary artery

smooth muscle cells (PASMCs) in hypoxia in 2007 and found disagreement

regarding whether hypoxia increases (12 publications) or decreases (12

publications) proliferation of PASMCs 79. Debate is also ongoing regarding whether hypoxia is a direct stimulus or augments other stimuli 79. Variations between studies may be explained in part by variations in level of hypoxia, source of particular isolates selected, and serum levels in media 79. The role of HIF-1, downstream of hypoxia in the proliferative phenotype, is equally murky. For example, Schultz et. al. reported in 2006 that increases in HIF-1α promoted a proliferative response after stimulation by hypoxia and growth factors, by demonstrating knockdown of HIF-1α decreased proliferation of PASMCs 80. In

contrast, the same author reported in 2009 that HIF negatively impacted

proliferation, when increased HIF-1α levels following PHD inhibition decreased

proliferation that was rescued by HIF-1α depletion 81. While there may be debate about the effects of HIF-1, there is consistency that HIF-1 levels are increased in

the lung by hypoxia. In ex-vivo testing this increase is seen in as little as thirty

minutes of exposure but returns to baseline after 4 hours of exposure and is even

less than baseline by 16 hours of exposure 82. This apparent periodicity of HIF-1

exposure may contribute to the inconsistencies in observed responses in vitro. The

downstream effects of HIF-1 observed in PASMCs are diverse and consistent with the many HIF-1 target genes. HIF-1 is also required for thrombin induced proliferation 83. In addition to hypoxic regulation of growth factors, HIF-1 has effects on ion channels and cellular pH that contribute to the vascular pathology 78, 84. HIF-

34

1 activity in the pulmonary system may have effects in the systemic vasculature

through inducing expression of angiotensin converting enzymes that increase

levels of AngII 85.

HIF in VSMCs of the systemic vasculature

Differences in the response to hypoxia on the level of gene regulation emerge

when comparing systemic and pulmonary VSMCs 86. Consideration of the systemic vascular network includes both arteries and . The normoxic proliferation and apoptosis characteristics differ between arterial and venous

VSMCs87. Saphenous vein VSMCs have reduced migration toward PDGF compared to mammary artery VSMCs due to differences in receptor expression88.

There is additional evidence that aortic and umbilical vein VSMCs express different levels of vascular endothelial growth factor receptor 2 (VEGFR-2) 89. The role of

these receptors in proliferation and migration and their status as HIF-1 targets make it important to examine arteries and veins independently. As with the pulmonary system, the proliferative response of systemic arterial and venous

VSMCs to hypoxia is under debate. HIF involvement is confirmed by HIF-1α knock down resulting in decreased proliferation, decreased migration, and increased apoptosis 90, 91. Downstream HIF-1 targets including Thrombospondin 1 (THBS-

1), hexokinase 2 (HXK2), plasminogen activator inhibitor-1 (PAI-1), matrix

metalloproteinases (MMPs), and adrenergic receptors have all been proposed as

important in systemic VSMC physiology and response to hypoxic stimuli. Hypoxic

coronary artery SMCs feature increased migration and increased expression of

35

THBS-1 and its receptors, integrin-β1,-β3, CD36, which were mediated by HIF-1 activity and inhibited by receptor inhibition 90. In the functional regulation of THBS-

1, HIF-1 is also essential for the protein’s extracellular release 90. In the aorta, HIF-

1 has been implicated in the expression of PAI-1, I, and multiple matrix

metalloproteinases, including MMP-9,-2,-7 and -1, that contribute to the pathologic vascular remodeling in aneurysm and hypertension92, 93. Aortic vasoreactivity is also regulated by hypoxia through control of expression of α1badrenergic receptors, again differentiating it from the vena cava that does not experience such regulation94. HIF-1 is also implicated in mitochondrial regulation through HXK2.

Induction of HXK2 by HIF-1 triggers mitochondrial hyperpolarization resulting in apoptotic responses 91. In addition to direct effects of HIF-1 regulated genes, there is also evidence that HIF-1 action promotes autocrine signaling. In coronary artery smooth muscle cells, hypoxic accumulation of HIF-1α promotes expression of vascular endothelial growth factor A (VEGFA) and vascular endothelial growth factor receptor-1 (VEGFR-1) 95. Intrinsic increases in both receptor and ligand promote proliferation through autocrine stimulation of VEGFR-1 by VEGFA, inhibited with a VEGF neutralizing antibody95. The role of HIF-1 in systemic vascular remodeling in the vascular wall as a whole was examined in a model of

HIF-1 inhibition by local infusion of a dominant negative HIF-1α (DN- HIF-1α) adenovirus. Decreased carotid remodeling and neointimal formation in response to ligation was seen in mice related with the DN-HIF-1α compared to WT 91. This

implicates HIF-1 in the proliferative remodeling response, but does not help to

36

clarify which cells of the vascular wall are affected by the inhibition, and thus

responsible for the phenotypes.

In smooth muscle specific studies, the interplay between AngII and HIF-1α has

been a focus of several studies published on VSMC Hif-1α knockout in the

systemic circulation. Previously, AngII has been shown to increase cell proliferation and migration96. AngII stabilizes HIF-1α through induction of PI3k,

MAPK, and ROS signaling pathways 69. However the regulation is not unidirectional, as VSMC specific Hif-1α knockouts express higher levels of ATR1

indicating HIF dependent regulation of the angiotensin pathway 97. Phenotypically,

AngII induced aortic remodeling was decreased in Hif-1α knockouts, which in WT

animals occurs independently of vasoconstriction 93. HIF-1α has also been shown

to regulate . In one study a VSMC specific Hif-1α knockout

increased systolic, diastolic, and mean arterial pressure ultimately resulting in

systemic hypertension independent of cardiac function, but this finding was not

replicated in a subsequent study 93, 97. The role of VSMC HIF-1 mediated vessel

remodeling and contractility in the periphery is unknown.

Peripheral Vascular Disease

Peripheral vascular disease is an umbrella classification that describes both

arterial and venous pathologies. Peripheral arterial disease (PAD), only slightly

smaller in scope, includes an array of non-coronary arterial syndromes caused by

the altered structure and function of the arteries that supply the brain, visceral

37

organs, and the limbs. I will focus here on lower-extremity PAD, where occlusive

lesions limit perfusion to distal tissues in the limb.

Epidemiology and disease burden

PAD is a major cause of morbidity affecting 8-12 million patients in the United

States and more than 200 million worldwide 98, 99. Prevalence increases with age from 5.28% in the 45-49 year old population to 29% in those over 70 98, 99. The

distribution between men and women is nearly equal, though the presentation

differs between genders 100. Risk factors for PAD are similar to those of other

cardiovascular diseases, and include smoking, diabetes, hypertension, and hypercholesterolemia 98. In fact, smoking has a higher correlation with PAD than with coronary disease 101, 102. Consequences of PAD can include pain, mobility

limitation, tissue damage, and in severe cases can necessitate amputation of the

limb. PAD also correlates with increased mortality, such that the likelihood of death

over a 10 year period is increased 6 fold in a patient newly diagnosed with PAD

compared to a patient without PAD 103.

Clinical presentation and diagnosis

Symptomatic PAD classically presents with lower extremity pain exacerbated by

exercise and relieved by rest; however, symptoms vary substantially. Indeed,

intermittent claudication, defined as aching pain, cramping, weakness, numbness,

or heaviness of the leg, only affects 10-35% of patients. 40-50% of patients present

with atypical leg pain involving the calf, thigh, or buttock, and an additional 20-50%

38

are asymptomatic 104, 105. Overall, men are more likely to present with claudication

(50%) than women (25%) 100. The most severe cases, categorized as critical limb

ischemia (CLI), affect 1-2% of patients 104, 106. Findings in CLI include pain, aching or burning at rest and many also present with tissue loss characterized by non- healing ulcers, necrosis, or gangrene that can threaten the limb. Overall, any indication of exertional limitation in lower extremity muscles, history of walking impairment particularly with claudication, presence of a poorly healing or non- healing wound on the legs or feet, or pain at rest localized to the lower leg or foot particularly if it varies with upright or recumbent position should raise suspicion for

PAD 106. Physical exam findings for any PAD patient may also include diminished

pulses, changes such as cool temperature to touch and pallor or rubor, loss

of hair, brittle nails, muscle atrophy, and/or bruits auscultated in the distal aorta,

iliacs or femoral arteries 107, 108.

Severity of PAD can be classified by the Fontaine staging system which stratifies

patients based on symptom severity 109. A stage 1 patient is asymptomatic. Stage

2 includes those with intermittent claudication subdivided into IIa where

claudication only presents after more than 200m of walking and IIb where

claudication arises in less than 200m of walking. In stage III, patients have pain at

rest and/or overnight. Finally, stage IV, the most severe, is assigned when necrosis

or gangrene is involved. The level of arterial obstruction can also be assessed and

stratified non-invasively using the ankle-brachial index (ABI) which is a ratio of the

highest systolic pressure of the dorsalis pedis and posterior tibial arteries and the

systolic brachial artery pressure 110. The American Heart Association (AHA) and

39

American College of Cardiology Foundation (ACC) defines an ABI of ≤0.9 as

abnormal with 0.71-0.90 as mild obstruction, 0.41-0.70 as moderate obstruction,

and 0.00-0.40 as severe obstruction. Above 0.9, 0.9-0.99 is borderline, 1.0-1.4 is

normal, and >1.40 as indicative of noncompressible, stiff vessels 104, 111. The AHA

and ACC recommends that all patients ≥65 years of age be screened for PAD with

an ABI assessment 105. At diagnosis, angiography, through conventional contrast

radiography (the gold standard), ultrasound, computed tomography, or magnetic

resonance, can provide visualization of location and degree of vessel occlusion

and pressure gradients across lesions 112.

Pathogenesis and vascular compensation

The most common etiology of PAD is atherosclerosis, characterized by subintimal accumulation of lipid and cholesterol that form lesion which encroach on the vessel lumen limiting blood flow to distal tissue. These lesions are most commonly found in the femoral and popliteal arteries, but can also occur in the abdominal aorta, iliac arteries, and distal vessels including tibial and fibular arteries 113. The vasculature responds to flow limitations and ischemic sequelae through arteriogenic and angiogenic processes promoting restored perfusion.

Arteriogenesis

Arteriogenesis, defined by dilation and arterialization of a smaller pre-existing vessel, allows for increased blood flow. Adaptive arteriogenesis, often called collateral remodeling, describes the lumen diameter enlargement of smaller

40

collateral vessels to compensate for blockage of a larger vessel while de novo

arteriogenesis refers to the arterialization of a newly formed capillary 114. Collateral

vessels connect artery to artery or to arteriole in a redundant circuit. If flow through the primary conduit vessel is restricted, collaterals provides a pathway for blood to be detoured around the occlusion and renter the vascular tree at a distal point (Figure 1.6). In the context of peripheral ischemia, small diameter collateral vessels at baseline, engorge shortly after blockage 114. Over the subsequent days

to weeks a remodeling process occurs thickening the vessel wall and further

improving their carrying capacity. The drivers of this process remain the subject of

debate. Shear stress from rapidly increased blood flow has been implicated in

collateral remodeling 115-117. However, shear stress is not the only mediator, as evidenced by collateral growth in ischemia without the presence of a pressure gradient 118, 119. A proposed pathway relies on NO such that activation of eNOS by

shear stress on the endothelium or another means, triggers vessel dilation. The

increased vessel diameter corresponds to increased circumferential wall stress

promoting VSMC proliferation 115. NO appears to be critical to the process as demonstrated by eNOS global KO mice having reduced arteriogenesis 120. Cells outside the vessel wall have also been identified as contributors. Macrophages

particularly appear to play an important role through secretion of vasoactive

cytokines including FGFs and VEGFA 121, 122. HIF has also been identified as a

regulator of these outcomes with HIF stabilization through PHD2 deletion in

macrophages increasing arteriogenesis in mouse models of PAD 123.

41

Angiogenesis

Angiogenesis in the adult is an adaptive process through which new vessels are formed at the capillary level. These new vessels can replaced damaged vessels, provide alternative pathways if the capillary network is blocked, or expand the network to supply growing tissue. In the setting of ischemia, hypoxic tissue initiates an angiogenic cascade by secreting and array of cytokines, most notably VEGFA.

Indeed, hypoxia increases VEGF expression up to 30 fold within a matter of

minutes 124. This milieu of VEGF and other angiogenic cytokines stimulate

endothelial cells in the existing vasculature to initiate vessel sprouting 124, 125. In each sprout, a single endothelial cell acquires tip cell identity and neighboring cells follow behind forming the stalk 125. The newly formed vessel tube then elongates along the cytokine gradient toward the ischemic tissue 126. Further support by a

mural layer of pericytes or VSMCs marks vessel maturation and enhances vessel

stability. Vessels with associated pericytes persist even after reoxygenation of the

tissue, while those without regress 127. HIFs are important drivers of angiogenesis in hypoxia, and are well known to increase VEGF expression 128-130. Indeed, loss of ARNT disrupts angiogenesis 73. VEGFA is necessary for effective angiogenesis

but not sufficient to complete the process alone 131 as VEGFA treatment in isolation

also detrimentally increases vascular permeability. Angiopoietin signaling through

Tie2 receptor helps to balance these effects as such co-expression of the ANG1 and VEGF result in increased vessel numbers without excessive permeability 131,

132. When effective, vessel growth restores blood flow, alleviating hypoxia and thus

inhibiting further angiogenic stimulation.

42

Figure 1.6. Vascular responses to flow limitation. Obstruction of a main artery generates hypoxia in the distal tissue. The vasculature responds through (A) arteriogenesis and (B) angiogenesis to alleviate the ischemia and hypoxia. Arteriogenesis is characterized by lumen diameter enlargement and remodeling of existing vessels and collaterals to detour blood around the obstructed segment. Angiogenesis involves the growth of new vessels at the capillary level to expand local delivery and reperfuse ischemic tissue.

43

Therapeutic interventions

Treatment of PAD can include modifying underlying systemic disease, addressing risk factors for progression, and direct mechanical intervention to restore blood flow. Many factors that contribute to PAD progression are also risk factors for other cardiovascular diseases thus systemic intervention can not only impact PAD outcomes but also improve or prevent disease in other vascular beds. Such systemic interventions include smoking cessation, dietary adjustment, exercise therapy, lipid lowering, hypertension management, blood sugar control in diabetic patients, and antiplatelet therapy 104, 105. With focus on the affected area, proper foot care can prevent or treat wounds that are slow to heal or non-healing in the context of PAD. Symptomatic relief of claudication can be achieved with pharmacologic therapy to promote and inhibit platelet aggregation and exercise therapy, which in some cases can have improvements comparable to stent revascularization 133, 134.

In severe presentations such as critical limb ischemia (CLI), direct and rapid revascularization can limit tissue damage and prevent limb loss. Endovascular interventions can include plain balloon angioplasty, drug-coated balloons, bare- metal or drug-eluting stents, covered stents, and plaque removal 104-106, 135.

These interventions have better outcomes for aortoiliac disease than those of the smaller vessels and for lesions that are stenotic rather than occlusive 104, 136.

Open surgical interventions can also be beneficial allowing for direct plaque removal by endarterectomy. Bypass grafting using autologous veins, typically

44

saphenous, or prosthetic polymer grafts provide a new conduit for blood flow

circumventing the lesion. Duration of graft patency varies by both graft type and

location of bypass placement 105, 137.

HIF in VSMCs of the peripheral vasculature

While headway is being made in understanding the regulation of pulmonary and

aortic VSMC responses to hypoxic stress, the findings may not extend to the

peripheral vasculature. In rabbits, aortic and carotid artery VSMCs have greater

capacity for PDGF-BB induced proliferation and migration than femoral artery

VSMCs 138. This may be explained in part by increased PDGF induction of MAPK in aortic VSMCs 138. Thus, studies of the periphery specifically are necessary to shed light on the functions of VSMCs in PAD.

Model of PVD: HLI

Flow limiting lesions in PAD most frequently localize to the iliac and femoral

arteries. Mouse models of PAD, referred to as hind limb ischemia (HLI), are

generated by ligature occlusion of vessels at locations corresponding to the sites

of human disease 113, 139, 140. Occlusions are made in the iliac, femoral, or superficial femoral arteries generating ischemia in the distal tissue. Selection of occlusion site determines severity of ischemia and also affects the ease of study of angiogenic and arteriogenic processes 139, 141. Ligation of the superficial femoral artery occurs distal to the branching of the deep femoral artery (Figure 1.7), leaving intact the collaterals fed by the deep femoral artery and allowing for assessment

45

of arteriogenic remodeling of those vessels in addition to angiogenic

vascularization of the gastrocnemius. Occlusion of the femoral artery or iliac artery

above the branching of the collaterals, generates a more profound ischemia and

limits the contributions of the deep femoral artery collaterals to reperfusion.

HIFs in HLI model

A considerable body of literature has implicated the involvement of HIF-1 in the

response to peripheral ischemia. Levels of stabilized HIF-1α affect the response

to femoral artery ligation. Global Hif+/- mice show decreased reperfusion following

ligation 142. Non-specific HIF-1 activity is known to promote angiogenesis and arteriogenesis. Indeed, rabbits injected with a mutant, constitutively stabilized,

HIF-1α in the ischemic thigh after artery ligation showed increased angiogenesis and arteriogenesis 143, 144. Systemic treatment with dimethyloxalylglycine (DMOG)

to inhibit hydroxylation and thus stabilize HIF-1α resulted in increased

angiogenesis in the hind limbs of mice following femoral artery ligation 145.

Arteriogenesis was not examined in this model. With regard to the cells of the vascular wall, a reliance on HIFs has been shown in endothelial knockouts of HIF-

2α that display decreased reperfusion compared to wild type controls following induction of hind limb ischemia 146. Likewise, a recent paper demonstrated that

HIF-1α deletion in VSMCs reduced perfusion in a severe model of PAD, but did not examine mechanism for this deficit 147. HIFs are clearly important factors in promoting reperfusion responses to peripheral ischemia and a promising therapeutic target worthy of further study.

46

Figure 1.7. Hind limb ischemia: a murine model of PAD. Permanent ligatures occlude the superficial femoral artery proximal to the popliteal bifurcation and distal to the branching of the deep femoral artery to model large peripheral vessel blockage.

47

HIF as a therapeutic

In the last decade, gene therapy employing HIF-1α has been trialed in patients with CLI or intermittent claudication 148. Injections of stabilized HIF-1α in replication-deficient adenoviral vectors into ischemic limbs proved safe in phase 1; however, later stage testing in patients with intermittent claudication found no benefit over placebo in claudication onset time, ABI, or quality-of-life measurements 149, 150. This lack of response may in part be explained by lack of increased HIF-1 activity on a functional level as no change was seen in expression of HIF target, VEGF, or in EC progenitor recruitment 149.

Summary and Hypothesis

Although a variety of factors present during an ischemic episode may influence the accumulation of HIF-1α, the best recognized and singularly important is hypoxia.

HIF target genes have been implicated in regulation of vascular responses to peripheral ischemia through observations of global or endothelial deficiencies of

HIF-1α. VSMCs are an integral part of the mature vascular wall, yet their role in regulating these responses is not well understood. Hypoxia influences VSMC phenotype and contributes to remodeling of the pulmonary and central systemic vasculature, but its impact may be highly dependent on the specifics of the vessel type, the microenvironment, and pathology. This dissertation centers on the hypothesis that HIFs are essential regulators of smooth muscle cell activation and phenotype in the systemic vasculature. The following studies will evaluate the consequences of loss of canonical HIF activity in VSMCs on vascular remodeling,

48 including angiogenic and arteriogenic responses to peripheral ischemia, and on vasomotor function. The results obtained will offer important insights on HIF’s mechanistic regulation of VSMCs’ contributions to vascular responses challenging the paradigm that endothelial cells are the sole regulators of vascular function.

49

Chapter 2: Aryl Hydrocarbon Receptor Nuclear Translocator in Vascular

Smooth Muscle Cells is Required for Optimal Peripheral Perfusion

Recovery

Author List:

Anna Henry Borton, Bryan L. Benson, Lee E. Neilson, Ashley Saunders, M. Amer

Alaiti, Alex Y. Huang, Mukesh K. Jain, Aaron Proweller, Diana L. Ramirez-

Bergeron

Affiliations:

From the Department of Pathology (A.H.B., B.L.B., A.Y.H.), Case Cardiovascular

Research Institute (A.H.B., A.S.,M.A.A., M.K.J., A.P., D.L.R.-B.), Division of

Pediatric Hematology-Oncology, Department of Pediatrics (A.Y.H.), and Case

Comprehensive Cancer Center (A.Y.H.) at Case Western Reserve University

School of Medicine Cleveland OH 44106, USA, and Harrington Heart & Vascular

Institute (A.H.B., A.S., M.A.A., M.K.J., A.P., D.L.R.-B.), Neurological Institute

(L.E.N.), Angie Fowler Adolescent and Young Adult Cancer Institute and University

Hospitals Rainbow Babies and Children's Hospital (A.Y.H.) at University Hospitals,

Cleveland, OH 44106, USA.

Portions of this chapter are published in Journal of the American Heart Association

2018; 7. DOI:10.1161/jaha.118.009205

50

Summary

Background- Limb ischemia resulting from peripheral vascular disease (PVD) is a common cause of morbidity. Vessel occlusion limits blood flow creating a hypoxic environment that damages distal tissue requiring therapeutic revascularization.

Hypoxia-inducible factors (HIFs) are key transcriptional regulators of hypoxic vascular responses including angiogenesis and arteriogenesis. Despite vascular smooth muscle cells’ (VSMCs) importance in vessel integrity, little is known about their functional responses to hypoxia in PVD. This study investigated the role of

VSMC HIF in mediating peripheral ischemic responses.

Methods and Results- We utilized ArntSMKO mice with smooth muscle specific deletion of Aryl Hydrocarbon Receptor Nuclear Translocator (ARNT, HIF-1β), required for HIF transcriptional activity, in a femoral artery ligation model of PVD.

ArntSMKO mice exhibit impaired perfusion recovery despite normal collateral vessel dilation and angiogenic capillary responses. Decreased blood flow manifests in extensive tissue damage and hypoxia in ligated limbs of ArntSMKO mice.

Furthermore, loss of ARNT changes the proliferation, migration, and transcriptional profile of cultured VSMCs. ArntSMKO mice display disrupted VSMC morphology and wrapping around arterioles, and increased vascular permeability linked to decreased local blood flow.

Conclusions- Our data demonstrate that traditional vascular remodeling responses are insufficient to provide robust peripheral tissue reperfusion in

ArntSMKO mice. In all, this study highlights HIF responses to hypoxia in arteriole

51

VSMCs critical for the phenotypic and functional stability of vessels that aid in the recovery of blood flow in ischemic peripheral tissues.

52

Introduction

Limb ischemia is a sequela of peripheral vascular disease, a major cause of

morbidity affecting millions of people worldwide 98. Arterial occlusion limits blood

flow creating a hypoxic environment and damaging tissue, which in severe cases

can necessitate amputation. Vascular responses to episodes of ischemia attempt

to restore perfusion through arteriogenic processes of redirecting blood flow

through extant collateral circulation and angiogenic processes initiating de novo

growth of vessels to regenerate the downstream vascular network 151-153. While the role of endothelial cells (ECs) in initiating and effectuating these responses has been extensively examined, far less is known about the contributions and regulators of neighboring vascular smooth muscle cells (VSMCs).

In response to various physiological stresses, VSMC undergo phenotypic switching, permitting them to proliferate and migrate, contributing to the remodeling of the vascular wall in (PAH), systemic hypertension, and atherosclerosis among others 154, 155. For many of these conditions hypoxia is an

important pathologic trigger. Hypoxia-inducible factors (HIFs) are heterodimeric

transcription factors, composed of HIF-α and aryl hydrocarbon receptor nuclear

translocator (ARNT, HIF-β) subunits, essential for cellular responses to hypoxia 67,

72, 73, 156-159 . In the presence of oxygen (O2), HIF-1α and -2α are hydroxylated and

targeted for proteasomal degradation. Hypoxic conditions inhibit the hydroxylation

of α- subunits which dimerize with ubiquitously expressed ARNT to form active

HIF-1 or HIF-2 functioning as master regulators of oxygen homeostasis by binding to and activating gene promoters containing hypoxia response elements 159. While

53

HIF-2’s expression and activity are more restricted, HIF-1 has been implicated in

the transcriptional regulation of hundreds of genes including many involved in

vascular growth and remodeling 75, 160, 161.

Hypoxia and HIFs have been studied extensively in VSMCs of the pulmonary

vasculature. In the context of hypoxia driven PAH, HIF-1 is essential for VSMC

proliferation in vivo 76-78. However, the pulmonary vasculature differs from the

systemic vasculature in hemodynamic forces and O2 status suggesting that these

results may not be directly translatable to other arterial beds. Studies of the role of

HIF in VSMCs outside of the pulmonary circulation are limited and have primarily

focused on examining the interplay between HIF-1 and ANGII signaling in vascular

remodeling of the aorta 93, 97.

A considerable body of literature demonstrates the involvement of HIF in response

to peripheral ischemia. Human patients with critical limb ischemia exhibit changes

in HIF-1α protein levels and vascular density 162. Increased levels of stabilized HIF-

1α are also seen in mouse models of hind limb ischemia (HLI) induced by femoral

artery ligation 142. While global Hif-1α+/- mice show decreased blood reperfusion following ligation, injection of stabilized HIF-1α in ischemic muscle promotes angiogenesis and arteriogenesis thereby increasing reperfusion 142-145. Specific to vascular cells, EC responses are known to be dependent on HIFs, with EC-specific knockouts of HIF-2α displaying decreased blood flow following induction of hind limb ischemia due to aberrant arteriogenic and angiogenic responses 146. Most

recently, VSMC HIF-1α knockout mice were characterized with decreased limb

reperfusion following femoral artery ligation due to the enhanced production of

54

thrombospondin-2 (THBS-2), known to inhibit angiogenesis 147. However, while

HIFs’ regenerative vascular properties have been well described in the non- targeted and EC contexts, the specific mechanisms of revascularization dependent on HIF driven VSMC responses to peripheral ischemia are not well described.

The following studies tested the hypothesis that endogenous HIF in VSMCs contributes to peripheral perfusion recovery. Canonical HIF transcriptional activity was ablated in VSMCs in vivo through the tissue specific deletion of ARNT, the required HIF-β subunit. Our findings reveal impaired reperfusion following femoral artery ligation despite signs of arteriogenesis and angiogenesis. Evidence of disrupted VSMC organization at the arteriolar level implicate VSMCs as central to this deficit. Moreover, this study finds hypoxia’s capacity to regulate the molecular markers of VSMC structure, homeostasis, proliferation, and migration is ARNT- dependent. Our findings underscore the essential role of HIF-dependent hypoxia driven responses by VSMCs to achieve optimal peripheral perfusion recovery.

55

Methods

The data, analytic methods, and study materials will be made available upon

reasonable request to other researchers for purposes of reproducing the results or

replicating the procedure.

Mouse generation

Previously described SM22α-Cre+/- and Arntlox/lox mice were crossed and

maintained in a C57Bl/6j background 163-167. The progeny of SM22-Cre+/- Arnt+/lox

x Arntlox/lox were utilized for experiments. Arnt deletion was verified by protein and mRNA levels in aortic VSMCs isolated and cultured as described below. Sex matched and litter matched, or if unavailable, aged matched Cre- Arntlox/lox or Cre-

Arntwt/lox mice were selected as controls. All animal studies were performed with

the approval of the Case Western Reserve University Institutional Animal Care and

Use Committee.

Echocardiography

For transthoracic echocardiography, mice were anaesthetized by inhalation of 1%

isoflurane with O2. All data were recorded and analyzed by the VEVO 770 High

Resolution Imaging System (Fujifilm Visual Sonics Inc.) and the RMV-707B 30

MHz probe. In M-mode short-axis images, ejection fraction, and fractional

shortening were measured at the papillary muscle level. Measurements were

acquired at heart rates above 500 bpm.

56

HLI model

HLI procedure, tissue harvest for frozen preparation, and pigment perfusion were

performed as previously described with minor modifications139. Briefly, 8-12 week old mice, assigned a number for blinding purposes, were anesthetized by intraperitoneal (IP) injection of ketamine (80mg/kg; IP) and xylazine (7mg/kg; IP).

Surgical depth of anesthesia was verified by toe pinch. Following hair removal, pre-operative perfusion assessment, and preparation of the surgical field, an incision was made in the left inner thigh. Blunt dissection was used to visualize and separate the femoral artery from the neighboring vein and nerve. Two ligatures of 7-0 braided silk suture were placed on the superficial femoral artery distal to the branching of the deep femoral artery and proximal to the popliteal bifurcation, and the intervening artery was cut. Alternatively, a more severe model of HLI was achieved by placing both ligatures proximal to the branching of the deep femoral artery. The epigastric artery was dissected and cauterized (Bovie). Wound clips

(7.5mm, Michel) and 6-0 polypropylene suture were used to close the incision.

Post-operative analgesia with buprenorphine (0.06 mg/kg; IP; every 12 hours) was administered for 3 days. Male mice were utilized for these studies due to previously described improved HLI recovery in male vs female mice168, allowing more

dynamic range and increasing power to detect recovery deficits.

To assess perfusion, mice were anesthetized as described above and placed on

37°C heating pad for 5 minutes prior to scanning. Infrared laser Doppler scans of

the foot pad were performed at 4ms/pix in triplicate on a MOORLDI2-IR (Moore

Instruments Ltd). Mean blood flow on flux images was assessed by moorLDI v5.0

57

software package. Perfusions heat maps for visual display are presented with

palate limits of 0 and 2000.

Where indicated, 594-conjugated lycopersicon esculentum lectin (100μl, Dylight,

DL-1177) was administered via the jugular vein and allowed to circulate for 5

minutes at 7 days following HLI. For vascular permeability assessments, 2mg of

2,000 kDa FITC-Dextran (Sigma, FD2000S) was administered via tail vein, 2 hours

before euthanasia. In anesthetized mice, the chest was opened and the left

ventricle was cannulated and infused with vasodilation solution (10U/ml heparin

[Sigma], 100μM adenosine [Sigma], 100μM papaverine hydrochloride [Sigma],

0.05% wt/vol bovine serum albumin [BSA, fraction V, Fisher] in Ca+ Mg+ DPBS

[Invitrogen]) at 10ml/min followed by warm 10% neutral buffered formalin, or

pigmented fixation solution (8g gouache [Winsor & Newton, 470] in 50ml 4%

paraformaldehyde). Some mice were injected IP with the hypoxic marker

pimonidazole [1-[(2-hydroxy-3-piperidinyl)propyl]-2-nitroimidazole] hydrochloride

(60mg/kg, HPI) 30 minutes before euthanasia to visualize hypoxic regions.

Collateral assessment

Limbs perfused with pigmented fixation solution were removed at the hip. After removing the skin, limbs were dehydrated to 100% methanol and the tissues cleared with 1:1 benzyl alcohol: benzyl benzoate. Vessels were imaged with Leica

MZ 16 FA at 10x, without a filter, and 65x, with a GFP filter for improved contrast.

Average diameter was quantified in Image J as the dividend of the vessel profile

58 area and the vessel length from tiled 65x images segmented in Photoshop

(Adobe).

Histology and immunohistochemistry

Gastrocnemius (GC) muscles for histological assessment were harvested, cryoprotected in 15% sucrose then 30% sucrose overnight, embedded in OCT compound (Tissue Tek), frozen and stored at -80°C. 8-μm serial transverse sections of GC muscle were obtained with a cryostat (Leica CM 1850 UV) and post-fixed in 2% paraformaldehyde. For damage assessment, sections were stained with hematoxylin & eosin and imaged with Leica DM 2000LED (40x objective). For capillary quantification, sections were blocked with 2% BSA and 5% normal goat serum in PBS and subsequently incubated with anti-CD31 (1:50, BD-

Pharmingen, 550274) followed by goat anti-rat IgG-488 (1:200, Invitrogen, A-

11006) and, where indicated, Cy3-conjugated SMA (1:400, Sigma, C6198). To detect areas of hypoxia, 0.1% tritonX-100 was added to the blocking buffer above followed by FITC-conjugated mouse anti-pimonidazole (1:200, Hypoxyprobe).

Tissues were counterstained with DAPI in mounting medium (Vector). Images were captured on a Leica DMI 6000 B with a 10x objective. For myocyte enumeration, GC sections were labeled with 5μg/ml wheat germ agglutinin,

Oregon green-488 (Invitrogen, W7024) in PBS. Damage and hypoxic areas were quantified as a percentage of GC cross sectional area in image J. CD31+ capillaries, Lectin+ vessels, SMA+ VSMCs, and skeletal myocytes were

59

enumerated in blinded raw images collected at the same exposure. For each limb,

6, 301um2 regions of 2 GC sections were quantified.

Arteriole imaging in spinotrapezius whole mount

Collection and visualization of vessels in the spinotrapezius has been previously

169 described . Briefly, following euthanasia with CO2 and left ventricular infusion of

warm PBS, spinotrapezius muscles were collected and fixed in 4%

paraformaldehyde in PBS for 20 minutes at room temperature. Tissues were

blocked with 2% BSA and 0.3% Triton-X 100 in PBS ON at 4°C. VSMCs were

identified by FITC-conjugated anti-SMA (1:400, Sigma, F3777). Confocal images

were taken on a Leica SP5 DMI 6000B using argon 488 nm and helium-neon 633

nm laser lines with a Leica 506192 HCX PL APO lambda blue 63x/1.4 oil objective

with 0.17 mm glass correction. For detection, 12-bit photomultiplier tubes were

used with Leica LAS AF acquisition software.

Vascular imaging in thick sections of GCs

Following post HLI tissue collection as described above, 400μm coronal sections

of GC were cut by vibratome (Leica VT 1200). Sections were blocked with 5%

normal goat serum, 2% BSA, and 0.3% Triton-X 100 in PBS. In permeability

studies, vessels were labeled with anti-CD31 (1:50, BD-Pharmingen, 550274)

followed by goat anti-rat IgG-647 (1:200, Invitrogen, A21247). VSMCs were

labeled with FITC-conjugated anti-SMA (1:400, Sigma, F3777). A Leica SP5

confocal microscope equipped with a 20x water immersion lens (Leica HCX-APO-

60

L, N.A. 1.0) and a tunable 16W Ti/Sapphire IR laser tuned to 800 nm (Chameleon

Coherent, Inc.) was used for two-photon laser scanning microscopy imaging using

non-descanned detectors set to capture Alexa Fluor 647 CD31 and/or FITC

(Dextran or SMA) fluorescence. For perfusion imaging, XYZ images with an XY

dimension of 775 x 775 µm were obtained at 512x512 pixels in 5 µm z stacks. For

SMA imaging of GC, XYZ images with an XY dimension of 310 x 310 µm were

obtained at 1024x1024 pixels in 1.95 µm z stacks.

Confocal and multiphoton image processing

High resolution confocal and multi photon images were deconvolved by Huygens

Professional 16.10 using Classic Maximum Likelihood Estimation at an estimated

signal-to-noise ratio of 5. Point spread functions were estimated a priori in Huygens

Professional using the wavelengths of excitation and emission light, position of the cover slip and orientation of the lens, lens immersion and specimen mounting media, and pinhole radius. These parameters were held identical within sets of images. Following restoration, data were imported to Imaris (BitPlane, Inc.) to

generate figures. Low resolution confocal images were not deconvolved, and a

median filter of 3x3 voxels was applied in Imaris. Due to inhomogeneities in SMA

staining intensity between and within tissues, brightness and contrast were

adjusted to facilitate comparison of smooth muscle cell architecture.

61

Cell culture

Isolation and culture of primary aortic VSMCs were performed as previously

described 164. Briefly, explants of thoracic aorta were divided longitudinally and plated lumen side down in 1mm2 pieces on 2 tissue culture dishes and covered

with glass cover slips. Plates were incubated in DMEM/F12 supplemented with

GlutaMAX-1 (Gibco, 10565-018) and 20% FBS (Atlanta or Gemini bio-products).

After 2 weeks of growth at 37°C and 5% CO2, explants and cover slips were

removed, and media was reduced to 10% FBS. For transcription analysis, the

following day, cells were starved in DMEM/F12+0.5% FBS for 16 hours, then

exposed to 2% O2 (hypoxia) or 21% O2 (normoxia) for 24 hours in

DMEM/F12+10% FBS.

Proliferation assays

20,000 cells per well of VSMC cultures were plated in 12- well plates. Following 16

hours starvation in serum free DMEM/F12, cells were exposed to hypoxia or

normoxia in DMEM/F12 +5% FBS. At 24 hours exposure, BrdU (5-bromo-2'-

deoxyuridine) was added to 10μM and incubated for an additional 8 hours in normoxia or hypoxia. Cells were fixed in 4% paraformaldehyde, followed by antigen retrieval with 1M HCl, and blocking with 0.75% BSA and 0.1% Triton-X

100. BrdU positive cells were labeled with Anti-BrdU (1.25ug/ml, BD biosciences,

B44) and counter stained with DAPI (Vectashield, H-1200). Assays were performed in triplicate. 6 representative images from each well were quantified.

62

Migration assay

Cells were plated at confluence and starved for 16 hours in serum free DMEM/F12.

Following scratch with 200- µl standard pipette tip, cultures were placed in normoxia or hypoxia in DMEM/F12+0.5% FBS+ 25ng/ml PDGF-BB (PeproTech,

315-18). Migration distance was quantified from phase contrast images taken at 0 and 6.5 hours and analyzed using NIH Image J software.

Real-time RT-PCR

Total RNA was isolated from cultured cells using TRIzol reagent (Ambion). RNA was reversed transcribed to cDNA with QuantiTect reverse transcription kit

(Qiagen, 205311). Relative expression was quantified in technical triplicates by real-time quantitative RT-PCR using the FastStart Universal SYBR Green Master

(ROX) Mix (Roche, 04913850001) on a StepOnePlus system (Applied

Biosystems). Target gene expression was analyzed using the 2-ΔΔCt method

(threshold values) with normalization to 18S ribosomal rRNA 170. Results are

reported as gene expression relative to Arntlox/lox in normoxia. Primers utilized for

target genes are presented in supplemental table 1.

Western blotting

Cell extraction buffer (Invitrogen, FNN0011) supplemented with cOmplete

protease inhibitor cocktail and PhosSTOP phosphatase inhibitor cocktail (Roche,

04693159001 and 04906845001) was used to collect protein either from samples

63

64

of GC muscle through bead homogenization (Qiagen TissueLyserII) or from

VSMCs in culture. Protein was quantified by BCA. 40μg of cell lysate or 100μg of

GC lysate in SDS sample buffer (Boston Bioproducts) were run on 8%

polyacrylamide gels in tris-gylcine-SDS running buffer. Semi dry transfer to nitrocellulose membrane was conducted in transfer buffer with 20% methanol.

Western blots of total protein isolates were probed overnight at 4°C with rabbit anti-

ARNT (1:800, Cell signaling, 5537S) diluted in Pierce protein-free T20 (TBS) blocking buffer (Thermo Scientific, 37571). To detect the protein bands, blots were washed and probed with secondary HRP linked, anti-rabbit IgG (1:1000 or 1:2000,

Cell Signaling, 7074S) and detected by enhanced chemiluminescence according to the manufacturer's instructions (Pierce, 32106). Membranes were stripped

(Thermo Scientific, 21059) and reprobed with β-actin rabbit antibody (1:5000, Cell

Signaling, 4967S).

Statistical analysis

Results are reported as the mean ± SEM. Statistical analyses were performed as

identified in each figure legend. Normality and homoscedasticity were evaluated

with Shapiro-Wilks test and F-test or Brown-Forsythe. Post testing utilized Tukey’s

multiple comparisons test for one-way and two-way ANOVAs, Bonferroni’s multiple

comparisons test for repeated measures two-way ANOVA, and two-stage linear

step-up procedure of Benjamini, Krieger and Yekutieli for Kruskal-Wallis test

(GraphPad). Significance was defined as p <0.05.

65

Results

Generation of smooth muscle specific Arnt knockout mouse model

To examine the role of HIF signaling in vascular smooth muscle, we generated a

model of Arnt deletion to disrupt HIF canonical transcriptional activity, and avoid compensatory changes in other HIF family members which can confound single α- subunit deletion models 171, 172. The smooth muscle specificity of this deletion was

accomplished by crossing well characterized SM22α-Cre mice 163-165 with Arntlox/lox

166. Primary aortic VSMCs isolated from SM22α-Cre+/-/Arntlox/lox mice (ArntSMKO)

show loss of Arnt expression at the mRNA and protein levels (Figure 2.1A, B).

Maintenance of ARNT expression in skeletal muscle was assessed in bulk tissue

samples of GC. Similar ARNT mRNA and protein levels were present in GCs of

ArntSMKO and Arntlox/lox mice (Figure 2.1C, D). ArntSMKO mice display no overt phenotypes with similar weight, appearance, and longevity to littermate controls

(Arntlox/lox) (Figure 2.1E and data not shown). Since SM22α-Cre has demonstrated

activity in cardiac tissue 163, cardiac function was assessed by echocardiography

173. No differences were seen in heart rate, ejection fraction, fractional shortening,

or left ventricle mass of ArntSMKO compared to Arntlox/lox littermate controls (Figure

2.1F-I). These results indicate that mice with tissue specific deletion of Arnt in the smooth muscle appear healthy overall without compromise of cardiac function.

66

Figure 2.1. Characterization of ArntSMKO mice. Isolated aortic VSMCs from ArntSMKO mice show loss of ARNT expression in (A) mRNA; n=5, and (B) total protein; n=3. Tissue samples from gastrocnemius muscle show no differences in bulk ARNT expression in (C) mRNA; n=5, and (D) total protein; n=4. (E) Bodyweights of male mice are similar in ArntSMKO; n=8, and Arntlox/lox littermates; n=11. Smooth muscle specific ARNT deletion does not compromise cardiac function assessed by (F) heart rate, (G) ejection fraction, (H) fractional shortening, and (I) left ventricular mass on echocardiogram; n=5, Unpaired 2-tailed t-test (A- F,H,I) with Welch’s correction (H), or Mann-Whitney test (G): *p<0.05.

67

ArntSMKO mice show impaired blood flow recovery in HLI model

While atherosclerotic obstruction of peripheral arteries ordinarily contributes to

PVD in humans, we employed inducible hindlimb ischemia (HLI), the most

common mouse model of PVD. The superficial femoral artery was surgically

ligated distal to the deep femoral artery restricting downstream blood flow to the

limb. Peripheral perfusion recovery in the foot pad was examined by infrared laser

Doppler at 3, 7, 14, and 21 days after ligation (Figure 2.2A). Results, expressed as

ratio of flow in ligated/ unligated limb, show decreased perfusion recovery in the

ligated limbs of ArntSMKO mice compared to Arntlox/lox littermate controls beginning

at day 7 with continued separation through day 21 after femoral artery ligation

(Figure 2.2B). Recovery of blood flow in ArntSMKO mice is not only slower, but also plateaus at a lower level than controls. By day 7 a gap in perfusion of nearly 20% emerges, closing by only an additional 5% over the subsequent 2 weeks.

Functional motor recovery, evaluated using a 0-3 point scale from full function of foot flexion and toe grasp to dragging of the ligated limb, was delayed in ArntSMKO

mice, though did not achieve statistical significance (Figure 2.2C). Collectively,

mice lacking ARNT in vascular smooth muscle demonstrate a significant and

persistent disruption in perfusion recovery after femoral artery ligation.

68

Figure 2.2. Bulk perfusion and functional recovery is reduced in ArntSMKO mice following femoral artery ligation. Ligations distal to the deep femoral artery branch point were performed in the left limbs of age- and gender- matched adult ArntSMKO and Arntlox/lox mice. (A) Representative images of perfusion measured using infrared laser Doppler scanning for ArntSMKO and Arntlox/lox mice over 21-day recovery. (B) Perfusion, reported as a ratio of ligated to unligated limb, was significantly lower in ArntSMKO compared to Arntlox/lox mice from days 7 through 21; n≥10, repeated-measures ANOVA *p<0.05 ArntSMKO vs. Arntlox/lox (C) Functional scoring on days 3 and 7 show impairment trend in functional recovery in ArntSMKO mice; n=11.

69

Limb ischemia stimulates collateral remodeling in ArntSMKO and Arntlox/lox mice

Appearance of perfusion deficits in the first week after ligation and previously described roles for smooth muscle in arteriogenic responses prompted assessment of collateral vessel dilation and remodeling. Luminal diameters of collateral blood vessels branching proximal to the ligation and traveling through the adductor muscles were visualized by pigment perfusion and subsequent tissue clearance which revealed similar collateral patterning and diameter in unligated limbs of Arntlox/lox and ArntSMKO mice (Figure 2.3A-C). Seven days post ligation, the time point with the largest perfusion deficit in ArntSMKO, we observed significant collateral vessel dilation in the ligated limbs of either mouse genotype (Figure 2.3A-

C). Surprisingly, despite lower levels of distal limb perfusion, collateral vessels in

ArntSMKO have significantly larger lumen diameters than those in control animals

(Figure 2.3C). Resistance to blood flow through a vessel is inversely proportional to the its cross-sectional area, indicating that collaterals in ArntSMKO have on average 2.5- fold lower resistance to flow than the analogous vessels in Arntlox/lox mice (Figure 2.3D). The collateral cross-sectional area is inversely correlated with perfusion of the limb; mice with the lowest perfusion had the largest diameter collaterals (Supplemental Figure 2.1). These results suggest that the observed deficit perfusion in ArntSMKO is not due to inadequate collateral vessel dilation.

70

Figure 2.3. VSMC ARNT is not required for ischemia induced collateralization. Collateral vessel responses in adductor muscles visualized with pigment perfusion angiography at day 7. (A) Lumen diameter enlargement visible in ligated compared to respective unligated limbs of ArntSMKO and Arntlox/lox mice. 10x images in top rows illustrate similar patterning; arrowheads indicate collaterals of interest. Higher magnification inset outlined by dashed rectangle. Scale bar, 1mm. 65x images in bottom rows captured through GFP filter for improved contrast. Scale bar, 250μm. (B) Tracings of skeletonized images across entire length of collaterals. Scale bar, 1mm. (C) Average collateral diameters show dilation in ligated limbs of both ArntSMKO and Arntlox/lox mice. Collaterals in ArntSMKO ligated limbs are significantly larger than in Arntlox/lox; n=3. (D) Cross sectional area calculations reflect approximation of flow resistance differences and projected flow rate. Measurements indicate 2.5-fold larger collateral vessels in ArntSMKO relative to Arntlox/lox mice; n=3, one-way ANOVA: *p<0.05 ArntSMKO vs. Arntlox/lox, ^p<0.05 ligated vs. unligated.

71

Supplemental Figure 2.1. Collateral vessel lumen cross sectional area vs limb perfusion. (A) An inverse correlation is present between ratio of cross sectional area of collateral vessel lumens in ligated/unligated limbs and foot pad perfusion, reported as LDI ratio ligated/unligated; R2=0.848; n=6.

72

Early capillary angiogenesis does not depend on VSMC Arnt in HLI

With sufficient collateralization observed, we next evaluated the angiogenic

response to induced ischemia in the downstream vascular beds. A more severe

ischemia ligation model eliminating collateralization of the deep femoral artery

showed decreased reperfusion in ArntSMKO mice implicating dysregulation of

smaller vessels in impaired perfusion recovery (Supplemental Figure 2.2). To

assess for differences in angiogenesis, gastrocnemius (GC) sections were

collected seven days post ligation. Capillary number was evaluated by staining for

CD31+ ECs, and the presence or absence of blood flow in each vessel was

assessed by intravenous (IV) lectin injection prior to tissue collection (Figure 2.4A).

At baseline, the unligated limbs of ArntSMKO and Arntlox/lox show equivalent capillary

densities and fractional perfusion (Figure 2.4B-D). At day 7, similar capillary and

perfused vessel densities in ligated limbs of Arntlox/lox and ArntSMKO mice were also

observed (Figure 2.4B-D). In the ligated limbs, total number of capillaries in a

301μm2 area are not significantly different between Arntlox/lox and ArntSMKO (Figure

2.4B). While there is a decrease in the number of CD31+ capillaries/myocyte in the

ligated limb of Arntlox/lox and ArntSMKO relative to their counterpart unligated limb, it

is only statistically significant in ArntSMKO mice (Figure 2.4C). In contrast, fractional

perfusion of capillaries is significantly decreased in the ligated relative to unligated

limbs in both ArntSMKO and Arntlox/lox mice (Figure 2.4D). Sections from unligated

and ligated limbs were stained with SMA to identify smooth muscle cells

associated with any identified CD31+ capillaries. Unligated and day 7 sections

exhibited little co-localization of SMA with capillaries (Supplemental Figure 2.3).

73

As the bulk of angiogenic responses are known to occur between days 7 and 28

post femoral artery ligation, capillary density was also measured at day 28 to

thoroughly assess for effects of Arnt deficiency on angiogenesis. Quantified by

average capillaries/301μm2, both Arntlox/lox and ArntSMKO have increased numbers

of capillaries in the day 28 ligated GCs relative to unligated GCs (Figure 2.4F); however, when capillaries per myocyte were quantified, the smaller myocytes of

ArntSMKO ligated GCs reveal a deficiency in late angiogenic responses. While

Arntlox/lox shows recovery in the number of capillaries per myocyte by day 28,

ArntSMKO recovery is limited with significantly reduced capillary to myocyte ratios in

ligated limbs (Figure 2.4E, G). No difference was apparent at day 28 in frequency

of SMA+ CD31+ vessels between ligated ArntSMKO and Arntlox/lox limbs (Figure 2.4E,

H). To comprehensively evaluate for variability in regional vascularization as recently shown by Schaad et. al. 174, we assessed vascularity in 6 distinct regions

across the GC muscle. Numbers of capillaries and perfused vessels between

ArntSMKO and Arntlox/lox were comparable in all 6 regions at days 7 and 28

(Supplemental Figure 2.4). In summary, while angiogenic deficiencies in ArntSMKO

may contribute to late (day 14-28) perfusion plateau, similar angiogenic patterns

and utilization of capillary beds are seen in both ArntSMKO and Arntlox/lox mice at day

7 following femoral artery ligation.

74

Supplemental Figure 2.2. Proximal HLI model. The left femoral artery was ligated proximal to the deep femoral artery branch point in age- and gender- matched adult ArntSMKO and Arntlox/lox mice. (A) Reduced perfusion, reported as a ratio of ligated/unligated limbs, was observed at days 3 and 14 in ArntSMKO mice; n=7, repeated-measures ANOVA: *p<0.05 ArntSMKO vs. Arntlox/lox.

75

Figure 2.4. Histological assessment of capillary number and perfusion status in gastrocnemius muscle (GC). (A) Representative micrographs of perfused vessels assessed by staining sections for CD31 (green) from GCs processed after mice were infused with endothelium binding IV lectin at day 7 following FAL (red). While, (B) the number of CD31+ capillaries per field at day 7 is not significantly different, ligated limbs of ArntSMKO and Arntlox/lox mice show decreased (C) CD31+ capillaries/myocyte, and (D) fraction of perfused, double positive, vessels compared to unligated limbs at day 7; n=4. No significant differences were seen between ArntSMKO vs. Arntlox/lox in any of the above metrics at day 7. (E) Representative images of immunostained sections for CD31 labeled vessels (green) and α-smooth muscle actin (SMA, smooth muscle cells, red) from day 28 post ligation GCs. (F) Numbers of CD31+ capillaries per field in day 28 ligated limbs of ArntSMKO and Arntlox/lox are similar to if not increased over unligated limbs; n=3. (G) Ligated limbs of ArntSMKO mice show decreased CD31+ capillaries/myocyte compared to ligated limbs of Arntlox/lox mice at day 28; n=3. Scale bars, 50μm. (H)

76

Day 28 ligated limbs of ArntSMKO and Arntlox/lox have similar numbers of CD31+ SMA+ vessels; n=3, One-way ANOVA (B), Kruskal-Wallis test (C,D,F,G), or unpaired 2-tailed t-test (H): *p<0.05 ArntSMKO vs. Arntlox/lox, ^p<0.05 ligated vs. unligated.

77

Supplemental Figure 2.3. Capillary density and smooth muscle cell colocalization in gastrocnemius (GC) at day 7. Representative images of immunostained sections for CD31 labeled vessels (green) and α-smooth muscle actin (SMA, smooth muscle cells, red) from (A) unligated and (B) day 7 post ligation GCs. No apparent difference in number of SMA+ vessels between ArntSMKO and Arntlox/lox in (A) unligated or (B) ligated day 7 limbs; n=3.

78

Supplemental Figure 2.4. Regional assessment of capillary density and perfusion status in gastrocnemius muscle (GC). (A) A diagram illustrates relative locations of evaluated regions in GC. (B) At day 7, assessment of CD31+ vessels shows similar capillary densities all regions ArntSMKO and Arntlox/lox GCs from both ligated and unligated limbs. Reductions in capillary density are seen in regions 1, 2, and

79

5 of ligated limbs relative to unligated while capillary density increases in region 3 of ligated limbs. (C) Comparable density of lectin+ vessels are also present in ArntSMKO and Arntlox/lox across the GC. Reductions in number of perfused vessels are seen in regions 1, 2, and 5 of ligated limbs. (D) Likewise, reductions in fraction of capillaries perfused are observed in regions 1, 2, 3, 5, and 6 of ligated limbs; n=4. (E) At day 28, GCs from ligated limbs of ArntSMKO and Arntlox/lox have similar CD31+ capillary densities; n=3, two-way ANOVA: ^p<0.05 ligated vs. unligated.

80

ArntSMKO mice show increased hypoxia and damage in gastrocnemius in

response to ischemia

To assess the anatomical consequences of perfusion deficit, GC muscle cross-

sections were stained with H&E. While ArntSMKO and Arntlox/lox unligated limbs

displayed normal skeletal muscle , striking differences were observed

between ligated limbs at day 7 (Figure 2.5A, B). Disruption of the muscular

structure, defined by centrally located nuclei and decreased myocyte diameter and

abundant infiltrating cells were prominent features in ArntSMKO while Arntlox/lox

tissues were relatively unaffected (Figure 2.5A). At day 28 post femoral artery

ligation, centralized nuclei and evidence of regenerating myocytes were still visible

in ArntSMKO GC sections (Figure 2.5A). Across the GC, these pathologic features extend over more than 60% of the ligated limbs of ArntSMKO mice compared to less

than 30% of the area of Arntlox/lox GC muscle (Figure 2.5B, C). Overall, the histology

reflects a maladaptive injury response consistent with chronically reduced blood

flow recovery in ArntSMKO mice.

At the tissue level, hypoxia identifies areas of limited perfusion; thus, to quantify the extent of hypoxic GC muscle following femoral artery ligation, pimonidazole intraperitoneal injection was used to label hypoxic tissues. Pimonidazole adducts, which identify tissue with <10mmHg O2, were neither found in GC sections from

unligated limbs of ArntSMKO and Arntlox/lox (Figure 2.5D), nor were they present in

Arntlox/lox sections at day 7 175, 176. However, large hypoxic pimonidazole+ areas

were present in ligated limbs of ArntSMKO mice 7 days post ligation (Figure 2.5D-

E). Together, significantly increased tissue damage and hypoxic areas

81

demonstrate that loss of VSMC ARNT leads to substantial skeletal muscle injury

following induced HLI.

Next, vascular integrity was examined by evaluating permeability to IV infused high

molecular weight FITC-Dextran. In both GCs of Arntlox/lox and the unligated limb of

ArntSMKO, Dextran was limited to CD31+ vascular structures in thick coronal sections imaged by multi-photon microscopy; however, extravascular Dextran infiltrates were diffusely present in GCs of ArntSMKO ligated limbs (Figure 2.5F).

Collectively, increased tissue damage, hypoxic areas, and vascular permeability suggest that compromised vasculature in ArntSMKO underlie impaired perfusion and

skeletal muscle injury following induced HLI.

82

Figure 2.5. Ischemic skeletal muscle regeneration and vessel integrity are impaired in ArntSMKO mice (A) Representative images of H&E stained GC sections show comparable muscle phenotype in unligated limbs. Day 7 assessment illustrates widespread atrophic myocytes and infiltrating cells in ArntSMKO while present only in small, well defined areas of Arntlox/lox GCs. Signs of damage and delayed regeneration, including centralized nuclei, persist to day 28 in ArntSMKO mice; n=3. Scale bars, 50μm. (B,C) Whole GC cross-sections show increased tissue damage in ArntSMKO compared to Arntlox/lox in ligated limbs at day 7; n=5. Scale bar, 500μm. Histological analysis (D) and quantification (E) of ArntSMKO ligated limbs show an increase in hypoxic regions identified by pimonidazole (Hypoxyprobe, green) staining at day 7 that is absent from unligated limbs of ArntSMKO and either Arntlox/lox limb, DAPI (blue, nuclei counter stain); n≥5. Scale bars: 50μm. (F) Representative multi-photon images of fluorescein-conjugated dextran (yellow) administered intravenously show increased permeability in ligated limbs of ArntSMKO mice at day 7. CD31+ vessels (purple); n=2. Scale bars: 100μm. Unpaired 2-tailed t-test (C) or Mann-Whitney test (E): *p<0.05 ArntSMKO vs. Arntlox/lox.

83

Altered vascular smooth muscle morphology in ArntSMKO mice

Despite substantial collateralization and similar capillary responses in the first week after ligation, the presence of significant and persistent perfusion deficits and tissue disruption point to a smooth muscle effect outside of these classically endothelial driven perfusion restoration mechanisms. In light of recent reports of aberrant arteriolar VSMC wrapping in dysfunctional vasculature 177, we examined

the cellular architecture of arterioles using whole mount confocal fluorescent

microscopy imaging of skeletal muscle samples. Visualization of α-SMA+ arterioles

revealed striking differences in VSMC morphology and conduit coverage in native

spinotrapezius from ArntSMKO versus Arntlox/lox mice (Figure 2.6A).

While the Arntlox/lox VSMC appear well organized and tightly wrapped with smooth coverage along the length of the vessel, the ArntSMKO VSMCs appeared more rounded, clearly delineated and detached (Figure 2.6A and Supplemental Figure

2.5). Similarly sized vessels were also examined deep in the GCs by multi-photon microscopy. Morphologic disruption of SMA+ VSMCs affected arterioles in ligated

and unligated limbs of ArntSMKO mice (Figure 2.6B). The aberrant perimural morphology of arterioles in ArntSMKO mice implicate a dysregulated VSMC

phenotype at the cellular level linked to impaired perfusion recovery.

84

Figure 2.6. Smooth muscle morphology and perimural wrapping of small arterioles. (A) Representative confocal images of SMA+ (cyan) VSMCs around small arterioles in spinotrapezius muscle illustrate disruption of organization and VSMC morphology in ArntSMKO mice; n=3. Top row scale bars: 60μm. Bottom row scale bars: 10μm. (B) Representative peripheral arterioles in GCs of ArntSMKO mice also display aberrant morphology and organization of SMA+ VSMCs visualized by multi-photon microscopy; n=3. Scale bars: 60μm, inserts 3x magnification.

85

Supplemental Figure 2.5. Additional images of arterioles in skeletal muscle. (A, B) Representative confocal images of SMA+ VSMCs around small arterioles in spinotrapezius muscle illustrate disruption of organization and VSMC morphology in ArntSMKO. (A) Scale bars: 60μm. (B) Scale bars: 10μm.

86

Loss of ARNT alters VSMC phenotype

To assess ARNT’s role in VSMC phenotype and responses to hypoxic stress in

ischemic limbs, cultured aortic VSMCs from ArntSMKO and Arntlox/lox mice were challenged with 24-hour hypoxic exposure at 2% oxygen 178, 179. Evaluation of transcripts revealed divergent changes in drivers of hypoxia induced phenotype modulation well described in pulmonary artery and aortic VSMCs (Figure 2.7) 80,

90, 92, 93, 180-182. Expression of HIF targets, vascular endothelial growth factor A

(Vegfa) and glucose transporter 1 (Glut1), whose levels rise in hypoxia and are

attenuated by ARNT deletion in ECs, responded as expected 167, 183-185. Hypoxia induced expression of Vegfa and Glut1 in Arntlox/lox VSMCs, and loss of ARNT

prevented induction of expression in hypoxia (Figure 2.7A, B). Transcriptional

changes were also observed in critical proliferative and migratory genes. ArntSMKO

VSMCs displayed reduced expression of serpin family E member 1 (Serpine1,

Pai1), fibroblast growth factor 2 (Fgf2), platelet derived growth factor receptor beta

(Pdgfrβ), and TIMP metallopeptidase inhibitor 1 (Timp1) mRNA levels relative to

hypoxic Arntlox/lox VSMCs (Figure 2.7C-E, H). Furthermore, differences in gene expression were observed under normoxia in ArntSMKO VSMCs including increases in matrix metalloproteinase 3 (Mmp3) and decreases in Vegfa, Glut1, Fgf2, Pdgfrβ, and thrombospondin-2 (Thbs2) relative to Arntlox/lox VSMCs (Figure 2.7A, B, D, E,

H, J), While hypoxic treatment of control VSMCs lead to reduced expression of

thrombospondin-1 and -2 (Thbs1 and 2), ArntSMKO VSMCs failed to mitigate Thbs2 reduction (Figure 2.7I, J). To examine the phenotypic consequences of transcriptional dysregulation, migration and proliferation were evaluated. Migration

87

assessed by 2-dimensional scratch assay in the presence of platelet derived

growth factor B (PdgfB) showed ArntSMKO cells are less migratory in hypoxia than

Arntlox/lox cultures (Figure 2.8A). Proliferation, quantified by percentage of BrdU+

cells, increased when Arntlox/lox cells were exposed to hypoxia for 24hours (Figure

2.8B). ArntSMKO VSMCs show no increased proliferation in hypoxia over normoxia,

but notably have greater percentage of replicating cells than Arntlox/lox under both conditions. Collectively, these findings suggest that gene expression differences

in hypoxia treated ArntSMKO VSMCs underlie cellular derangements responsible for vascular reperfusion impairment.

88

Figure 2.7. Transcriptional expression of proliferation and migration regulators in response to hypoxia. (A-J) mRNA from ArntSMKO and Arntlox/lox mouse aortic VSMC cultures following 24 hours hypoxia (2% O2) exposure were analyzed by qPCR, normalized to 18s rRNA, and compared to normoxic (21% O2) control samples. Transcriptional profile of VSMCs from ArntSMKO differs from control Arntlox/lox samples: (A) Vegfa, (B) Glut1, (C) Pai1 (Serpine1), (D) Fgf2, (E) Pdgfrβ, (F) Pdgfb, (G) Timp1, (H) Mmp3, (I) Thbs1, (J) Thbs2; n=3, one-way ANOVA: *p<0.05 ArntSMKO vs. Arntlox/lox, ^p<0.05 hypoxia vs. normoxia.

89

Figure 2.8. Assessment of VSMC phenotype in vitro. (A) Migration, detected in a 2-dimensional scratch assay, was measured after 6.5 hours in normoxic (21% O2) SMKO or hypoxic (2% O2) conditions. Arnt VSMCs show decreased migration in hypoxia compared to Arntlox/lox cultures; n=9. (B) Quantitative analyses of immunopositive BrdU cells reveal increased proliferation of ArntSMKO VSMCs in normoxia and hypoxia; n=3, one-way ANOVA: *p<0.05 ArntSMKO vs. Arntlox/lox, ^p<0.05 hypoxia vs. normoxia.

90

Discussion

The present study illustrates the importance of HIF orchestrated VSMC responses

in peripheral perfusion recovery in a model of induced HLI. Specifically, knockout

of ARNT in smooth muscle impairs perfusion restoration following femoral artery

ligation. While insufficient blood flow manifests in increased hypoxic tissue and

skeletal muscle damage in the ischemic limbs of ArntSMKO mice, interestingly there

were no signs of limitation in collateral dilation or of disruption of perfused capillary

density. However, loss of ARNT led to morphologic disorganization of VSMC

coverage of small arterioles and increased vascular permeability in ligated limbs.

Furthermore, the transcriptional dysregulation of multiple genes involved in VSMC

function affect proliferation and migration of isolated ArntSMKO cells. In all, this study

identifies hypoxia mediated responses in VSMCs critical to maintaining VSMC

phenotype, cellular organization around arterioles, and vessel integrity, and to

achieving optimal reperfusion of ischemic peripheral tissues.

Our observation of impaired limb recovery with diminished reperfusion is striking

in a mouse genetic model targeting smooth muscle cells. ECs have been

described as the primary regulators of blood flow and orchestrators of reperfusion;

however, the degree of impairment observed in ArntSMKO is more profound than that seen in global Hif-1α+/- mice 142, and on par with endothelial specific deletions of Hif-2α 146. Furthermore, our findings are supported by a recent report of

decreased reperfusion with Hif-1α deletion in VSMCs using a severe form of HLI

147. Our genetic model conditionally deleting VSMC-ARNT permits efficient study of all HIF- canonical transcriptional function, while HIF-α subunit activities involving

91

non-canonical binding partners remain undisrupted 186. In context, our results

demonstrate the importance of smooth muscle cell canonical HIF- dependent responses in perfusion recovery necessitated by regional peripheral ischemia.

Perfusion deficits in HLI models are often explained by impaired angiogenic and/or arteriogenic responses 187. Classically, hemodynamic changes prompt

arteriogenesis assessed by number and dilation of collaterals, while hypoxia drives

angiogenesis visualized by increased capillary density; however, local stabilization

of HIF-1α through introduction of a constitutively active variant or by inhibition of

its degradation pathway increases both angiogenic and arteriogenic responses 123,

144, 182, 188. The impacts on reperfusion mechanisms are not explicitly described in

Hif-1α+/- or tissue specific HIF-1α models, but endothelial specific deletion of Hif-

2α impairs both benchmark vascular remodeling processes 146. Lower capillary

density in day 28 ArntSMKO GCs help to explain the persisting perfusion limitations after recovery has plateaued. Yet, at day 7 when the perfusion deficit is largest, measurements of arteriogenesis and angiogenesis in response to HLI are similar in ArntSMKO and Arntlox/lox mice. Indeed, ligated limbs of ArntSMKO mice have larger

diameter collateral vessels at day 7 than their littermate controls. The inverse

correlation between collateral cross-sectional area and bulk perfusion in ArntSMKO

and Arntlox/lox limbs suggests that collateral vessel diameter is responsive to

changes in blood flow and compensating for elevated ischemia in mutant mice. In

a model that spares HIF signaling in the endothelium, traditionally viewed as the

primary driver of vascular ischemic responses, it may therefore be unremarkable

that these processes appear intact 189. Despite substantial arteriogenic and

92

angiogenic responses, these compensatory changes are insufficient to restore

perfusion to the affected tissue in the ArntSMKO mice. Whether due to greater extent of ischemic insult, injury, and/or impaired recovery, substantial areas of hypoxia remain at least 7 days post ligation and signs of tissue damage persist across the

GC muscle after 28 days of recovery. These findings suggest that established responses to ischemia through well-defined revascularization mechanisms do not adequately explain the perfusion deficit at day 7. Indeed, while increases in microvessel number have also been documented in affected peripheral tissues from patients with chronic limb ischemia, these responses also do not appear to be sufficient to alleviate ischemia162.

Perfusion, as measured by laser Doppler, can be described as a function of RBC concentration and flowrate 190. In the absence of differences in cardiovascular function, global volumetric flowrates should be comparable in ArntSMKO and

Arntlox/lox mice. Thus, the perfusion deficit can be attributed to local changes in

vascular flowrate in the hindlimb. Recent reports of pathologic vessel patterning in

regenerated vascular networks following HLI could clarify the ArntSMKO reperfusion

deficit. Arpino et al. implicated disordered vascular smooth muscle cell phenotype

and wrapping around small arterioles in limiting RBC transit through the

microvasculature of regenerating skeletal muscle 177. Similarly, in skeletal muscle

of ArntSMKO mice, we detect aberrant smooth muscle cell morphology and

investment of the small arterioles consistent with these novel reports. Furthermore,

the integrity of the microvasculature is compromised as evidenced by increased

permeability. Vascular leak affects pressure gradients and contributes to disruption

93

of local blood flow. Thus, while the hallmark revascularization measurements namely, collateral diameter, and capillary number and fractional perfusion appear normal, disruption in VSMC patterning in vessels of ArntSMKO mice likely

contributes to reduced reperfusion and muscle recovery following HLI.

In the pulmonary circulation, HIF-1 is a well described mediator of hypoxia

triggered vascular remodeling and VSMC phenotype modulation. Mouse models

of chronic hypoxia induced PAH with reduced HIF-1α activity through either global

haplodeficiency or SMC specific deletion demonstrate decreased vascular

remodeling 76, 191, 192. Mature VSMCs retain a remarkable amount of plasticity and

can exhibit a phenotypic spectrum ranging from principally contractile and rarely

dividing to highly synthetic, proliferative, and migratory 39. Early debate in the field regarding VSMC responses to hypoxia emerged in studies utilizing pulmonary artery SMC (PASMC) cultures 79. However, recent reports of PASMCs consistently

show HIF mediated hypoxic responses increase proliferation, survival, and

migration, and stimulate growth factor production including VEGFA 80, 180-182. HIF

targets including PDGFs and FGFs are key promoters of VSMC proliferation and

migration; their inhibition impairs proliferation of PASMCs in vitro and prevents

vascular remodeling in models of PAH193, 194. Expression of multiple HIF targets in

ArntSMKO VSMCs diverge from levels seen in control cells. Failure to maintain

expression levels of Pdgfrβ, Pdgfb, and Fgf2 indicates dysregulation of pathways

central to proliferative and migratory responses to hypoxia. Indeed, ArntSMKO cells do not show proliferation increase with hypoxic exposure over normoxia and are less migratory than Arntlox/lox in hypoxia.

94

While these effects of HIF dependent phenotypic modulation are varied in differing

oxygenation and hemodynamic environments outside of the pulmonary circulation,

several recent reports have identified HIF-1 as central to maintaining the structure

and function of the arterial wall 93, 97, 195. Downstream HIF-1 targets have been

identified as important in systemic VSMC physiology and response to hypoxic

stimuli 90, 92, 93. HIF targets, PDGF and THBS-2 regulate VSMC attachment to extracellular matrix; impaired expression of Pdgfrβ and Thbs2 in ArntSMKO help explain disrupted VSMC morphology observed around arterioles 196, 197. As

supporting cells in the vascular wall, it is well recognized that VSMCs are involved

in physiologic responses to mechanical and biochemical changes in blood vessels.

Changes in VSMC phenotype have consequences for vasoreactivity. Tissue

specific deletion of HIF-1α has been shown to increase contractility, a putative

marker of mature VSMC phenotype, in studies of aortic and pulmonary artery

VSMCs 77, 97, 198. The morphologic changes in VSMCs around small arterioles in

ArntSMKO mice taken together with the abnormal proliferative and migratory

behavior and the altered gene expression observed in isolated cells are consistent

with phenotypic dysregulation of VSMCs.

In summary, the present study indicates that loss of hypoxic signals in VSMCs

limits the ability of mice to recover from inducible hindlimb ischemia, a classic

model for PVD. We provide evidence that despite conventional compensatory

arteriogenic and angiogenic responses in the first week after ligation, dysregulated

smooth muscle cell function in ArntSMKO arterioles manifested by morphologic

disruption, aberrant expression of key phenotypic regulators, and altered

95

proliferation and migration is sufficient to compromise vascular integrity and

ultimately impair limb reperfusion. Our results therefore underscore a critical role

for VSMC HIF in peripheral perfusion recovery and reiterate the importance of

understanding the regulation of VSMC function in arteriolar vessels in supporting optimal blood flow.

Acknowledgements

We thank Alla Gomer, Keiki Sugi, Stephanie Lapping, and Alice Jo for their technical assistance.

Funding Sources

Funding for this work was provided by the NIH F30 HL127985 (AHB), RO1

HL128281 (AP & DLR-B), RO1 HL096597 (DLR-B), T32 HL105338 (AHB, MAA),

T32 GM7250 (AHB, BLB), and TL1 RR024991 (AHB, BLB), F31 NS096857 (BLB),

T32 NS077888 (BLB), R25 HL103152 (AS).

Conflict of Interest Disclosures

None.

96

Chapter 3: Loss of Vascular Smooth Muscle Cell Aryl Hydrocarbon

Receptor Nuclear Translocator Impairs Vasoconstriction

Authors:

Anna Henry Borton, Iulia Barbur, Alice Jo, Nicholas P Ziats, Aaron Proweller,

Diana L. Ramirez-Bergeron

Affiliations:

From the Department of Pathology (A.H.B., N.P.Z.), Case Cardiovascular

Research Institute (A.H.B., I.B., A.J., A.P., D.L.R.-B.) at Case Western Reserve

University School of Medicine Cleveland, OH, USA, and Harrington Heart &

Vascular Institute (A.H.B., I.B., A.J., A.P., D.L.R.-B.), at University Hospitals,

Cleveland, OH, USA.

97

Summary

Vasoreactivity mediated by vascular smooth muscle cells (VSMCs) is critical for

cardiovascular homeostasis. Mature VSMCs retain phenotypic plasticity, and can

shift under stress from contractile to synthetic states. We previously identified a

requirement for smooth muscle Aryl hydrocarbon receptor nuclear translocator

(ARNT; HIF-1β), in hypoxia-induced arterial reperfusion prompting an interrogation

of the consequences of smooth muscle specific loss of ARNT (ArntSMKO) on

vasomotor function. Isolated primary ArntSMKO VSMCs display reduced myofilament expression consistent with a shift away from a contractile phenotype.

Moreover, though morphologically indistinguishable from control arteries, ArntSMKO

arteries exhibit impaired vasoconstriction with preserved vasorelaxation assessed by ex-vivo wire myography. Taken together, our data implicate ARNT as a key regulator of the contractile phenotype in VSMCs and arterial vasoconstriction.

98

Introduction

Vascular reactivity is essential for maintaining stable blood pressure and

appropriate distribution of blood flow. Vascular smooth muscle cells (VSMCs)

residing in the tunica media of the arterial wall are responsible for constricting and

dilating the luminal diameter in response to physiologic and pathologic stimuli.

Alterations in vasoconstriction and contractile dynamics are features of many

pathologies, including hypertension, pulmonary hypertension, sepsis, and

ischemia199-202.

Hypoxia can both precipitate and be a sequela of alterations in blood flow.

Hypoxia-inducible factors (HIFs) are master regulators of oxygen homeostasis.

This family of transcription factors are heterodimers formed when HIF-1α, -2α, or

-3α bind to a common β-subunit, aryl hydrocarbon nuclear translocator (HIF-1β,

ARNT) 67, 72, 73, 156-159. HIF-1α is widely expressed with HIF -2α and -3α more cell type restricted 75, 160, 161. In the presence of oxygen, the regulatory α-subunits are

hydroxylated and rapidly targeted for proteasomal degradation. In hypoxia, HIF

transcriptional activity is induced through the stabilization of the α-subunits and

their dimerization with constitutively expressed ARNT.

While investigating the role of HIFs in regulating the functional responses of

VSMCs in the systemic circulation, we reported that loss of ARNT led to decreased

reperfusion recovery with altered VSMC phenotype using a tissue specific

knockout mouse model 203. In vivo, VSMCs featured aberrant periarteriole

wrapping and morphology, while in vitro mutant cells displayed altered proliferation

and migration. Collectively these findings implicated ARNT as essential for

99 phenotype stability. VSMC phenotype is classically described as a spectrum between contractile and synthetic cell states39. Contractile cells are quiescent and rarely proliferate while synthetic cells proliferate and migrate with greater frequency. At baseline most cells are contractile, however stress or other pathologic triggers can prompt a phenotypic change toward a more synthetic state.

Our previous observations indicating a shift toward the synthetic phenotype in

VSMCs lacking ARNT prompted closer examination of contractile function. The following studies tested the hypothesis that loss of ARNT in VSMCs disrupts vasoreactivity. At the cellular level, ARNT deleted VSMCs display significant reductions in expression of contractile myofilaments. Vessels lacking VSMC ARNT feature impaired vasoconstriction with intact vasorelaxation and preserved dimensional structure of the vascular wall. Together, our findings describe an important role for VSMC canonical HIF activity in maintaining functional vasoreactivity.

100

Methods

Mouse model

All animal studies were carried out under the approval of the Case Western

Reserve University Institutional Animal Care and Use Committee. A smooth

muscle specific mouse genetic model of Arnt deletion was generated as previously

described and characterized. Briefly, SM22-Cre+/- Arnt+/lox male mice were crossed with Arntlox/lox females yielding SM22-Cre+/- Arntlox/lox experimental mice and Cre-

Arntlox/lox or Cre- Arntwt/lox controls. Pairs of experimental and control mice used for experiments were sex matched and age, if not litter, matched.

Cell culture

Cells were isolated and cultured as previously described. Briefly, thoracic were explanted, cleaned of , divided longitudinally into 2 tissue culture dishes, and plated in 1mm2 pieces, endothelial side down, under glass cover slips. Explants were incubated for 2 weeks in DMEM/F12 supplemented with

GlutaMAX-1 (Gibco, 10565-018) and 20% FBS (Atlanta or Gemini bio-products) at

37°C and 5% CO2. Thereafter, cover slips and explanted tissue pieces were

removed and FBS supplementation was reduced to 10%. Cells were starved for

16hours in DMEM/F12+0.5% FBS, then returned to DMEM/F12+10% FBS and

placed in hypoxia (2% O2, 5% CO2) or normoxia (atmospheric O2, 5% CO2) for 24

hours.

101

Real-time RT-PCR

At collection, cells were lysed and total RNA collected with Triazol reagent.

Following RNA isolation and quantification, cDNA was reverse transcribed from

500ng of RNA using the QuantiTect reverse transcription kit (Qiagen, 205311).

Relative expression was quantified by real-time quantitative RT-PCR in technical

triplicates on a StepOnePlus system (Applied Biosystems) using the FastStart

Universal SYBR Green Master (ROX) Mix (Roche, 04913850001). Target gene

expression was analyzed using the 2-ΔΔCt method (threshold values) with normalization to 18S rRNA 170and reported as gene expression relative to

normoxic Arntlox/lox sample. Primers for each target gene are as follows:

18s Forward: 5'-GAATTCCCAGTAAGTGCGGG-3', Reverse: 5'-

GGGCAGGGACTTAATCAACG-3'; Cnn1 Forward: 5'-

GGCCAAGACAAAAGGAAAC-3', Reverse: 5'-CCATCTGCAGTCCAATGATG-3';

Myh11 Forward: 5'-GGCTTCATTTGTTCCTTCCA-3', Reverse: 5'-

CGAGCGTCCATTTCTTCTTC-3'; Acta2 Forward: 5'-

TGTGCTGGACTCTGGAGAT-3', Reverse: 5'-GAAGGAATAGCCACGCTCA-3'.

Histology

Carotid arteries were collected from adult mice and fixed in 4% paraformaldehyde overnight. Tissues specimens were dehydrated to 100% ethanol followed by embedding in paraffin wax. 8μm sections affixed to slides were deparaffinized, rehydrated, and stained with hematoxylin & eosin or Verhoeff–Van Gieson elastin stain. Images were acquired with Leica DM 2000LED microscope (20x objective).

102

To quantify the average tunica media thickness, the areas enclosed by the external

elastic lamina (EEL) and internal elastic lamina (IEL) were manually outlined using

the free-hand selection tool (ImageJ 1.50, National Institutes of Health, USA). The

area was then used to calculate the radius of a circle of equivalent circumference.

In each image, the average radius of the IEL was subtracted from the average

radius of the EEL to obtain the average tunica media thickness.

Isometric tension measurement

Vasomotor function studies were performed as previously described 204. Briefly, thoracic aortas from 10-19 week mice were cleaned of adipose tissue and cut into

2mm rings. Two rings from the proximal thoracic aorta were suspended in an

isometric tension myograph (610M, Danish Myo Technology), in physiological

saline solution equilibrated with a gas mixture of 95% O2 and 5% CO2 at 37 °C.

All vessel segments were pretensed to 6mN and allowed to equilibrate. For depolarization-induced vasoconstriction studies, potassium chloride was added to a final concentration of 60mM. To assess agonist-based constriction, dose- response curves for phenylephrine (PE) and angiotensin II (AngII) over a range of

10-9 to 10-5 M were acquired in triplicate. Relaxation responses to acetylcholine

(ACh), and sodium nitroprusside (SNP) were assessed over a range of 10-9 to 10-

5 M in triplicate following a 10-2 M dose of PE. Data are presented as active tension in millinewtons for depolarization and agonist-induced contraction and as percentage of maximum PE-induced tension for ACh- and SNP-mediated relaxation.

103

Statistical analysis

Results are reported as mean ± standard error of the mean. Normality and homogeneity of variance were evaluated with Shapiro-Wilks test and F-test or

Brown-Forsythe. Statistical tests were performed as described in figures legends with post testing by Tukey’s multiple comparisons test for one-way ANOVAs, or

Bonferroni’s multiple comparisons test for repeated measures two-way ANOVA,

(GraphPad). Significance was defined as p <0.05.

104

Results

Transcriptional expression of contractile elements are reduced in VSMCs lacking ARNT

Phenotypic shifts in VSMCs are often marked by changes in expression of contractile genes 39. To determine whether ablation of canonical HIF activity impacted the contractile apparatus, tissue specific ARNT deletion in VSMCs was accomplished in a murine model, as previously described, by intercrossing a

SM22α driven CRE transgenic mouse line 163-165 with Arnt floxed mice 166

(Arntlox/lox) to generate SM22α-Cre Arntlox/lox (ArntSMKO). Primary aortic smooth

muscle cells were isolated from ArntSMKO and Arntlox/lox mice and mRNA expression was examined under normoxia (21% O2) and after 24- hour exposure to hypoxia

(2% O2). Results report expression normalized to 18srRNA and relative to

normoxic Arntlox/lox cell cultures. ArntSMKO VSMCs displayed reduced expression of alpha smooth muscle actin (Acta2), smooth muscle myosin heavy chain (Myh11), and Calponin (Cnn1) in normoxia with Myh11 and Cnn1 expression also significantly lower than Arntlox/lox cells in hypoxic conditions (Figure 3.1A-C).

Moreover, relative to normoxia, hypoxia exposure reduced expression of Acta2,

Myh11 and Cnn1 in controls but resulted in no further decline in transcript levels in

ArntSMKO cultures (Figure 3.1A-C). Collectively, absence of response to hypoxia and overall reduction in contractile genes indicated a shift away from contractile phenotype in ArntSMKO VSMCs.

105

Figure 3.1. Contractile gene expression is reduced in ArntSMKO VSMCs. mRNA from ArntSMKO and Arntlox/lox primary aortic VSMC cultures exposed to normoxia (21% O2) or hypoxia (2% O2) were analyzed by qPCR, normalized to 18s rRNA. Results are reported relative to expression in normoxic Arntlox/lox cells. (A) Alpha smooth muscle actin (Acta2), (B) smooth muscle myosin heavy chain (Myh11), and (C) Calponin (Cnn1) mRNA expression was reduced in SMKO VSMCs relative to Arntlox/lox in normoxia and hypoxia. Relative to normoxia Arntlox/lox VSMCs decreased expression of Acta2, Myh11, or Cnn1 expression under hypoxia, while ArntSMKO cells have similar levels in normoxia and hypoxia; n=3. One-way ANOVA: *p<0.05 ArntSMKO vs. Arntlox/lox, ^p<0.05 hypoxia vs. normoxia.

106

Large vessel structure and tunica media thickness is not affected in ArntSMKO

To assess for baseline changes in structure of the vessel wall, cross sections of

carotid artery were stained with Hematoxylin and Eosin (H&E) or Elastin as

displayed in Figures 3.2A and 3.2B, respectively. Vessels from Arntlox/lox and

ArntSMKO mice exhibit similar structure without disruption of the elastic lamina or

visible alterations in the intima or medial layers (Figure 3.2A, B). The average thickness of the tunica media composed of vascular smooth muscle cells was quantified and no difference was found between Arntlox/lox and ArntSMKO,

25.7±8.6μm and 22.9±6.7μm, respectively (Figure 3.2C). These data indicate

similar baseline vascular structure in large arteries of Arntlox/lox and ArntSMKO mice.

Loss of Arnt in VSMCs impairs aortic vasoconstriction

Consequences of ArntSMKO on constrictor function of aortic segments were determined ex vivo via wire myography. When constriction was induced through electromechanical coupling by depolarization with potassium chloride (KCl),

ArntSMKO aortas demonstrated significantly reduced force generation compared to

Arntlox/lox aortas (Figure 3.3A). Similarly, contractile responses to stimulation through pharmacomechanical coupling by receptor dependent agonists, phenylephrine (PE) and angiotensin II, were also significantly reduced in ArntSMKO

aortas (Figure 3.3B, C). Contractile force generation in response to PE was

reduced by more than 50% on average between 10-7-10-5M, with statistical

significance at 10-5M. Responses to AngII were significantly reduced at concentrations from 10-7-10-5M by an average of more than 60%. Collectively,

107

Figure 3.2. Large vessel structure unchanged in ArntSMKO. Histologic assessment by (A) H&E and (B) elastin staining and (C) quantification of tunica media thickness show similar structure of the vascular wall in cross sectional views of Arntlox/lox and ArntSMKO carotid arteries. Scale bars 50μm; n=8. Unpaired t-test.

108

reduced force generation to multiple contractile agonists indicates vasoconstriction deficits in ArntSMKO aortas.

Loss of Arnt in VSMCs does not affect aortic vasorelaxation

Impaired contraction can, in some cases, be explained by changes in structure or coordination that would also affect vasorelaxation. Endothelial dependent relaxation in response to acetylcholine (ACh) and smooth muscle intrinsic relaxation by sodium nitroprusside (SNP) were both evaluated in aortic segments ex vivo by wire myography and reported as percent of maximal constriction to PE

(Figure 3.4A, B). ArntSMKO and Arntlox/lox aortas showed similar relaxation responses to ACh or SNP. Maintenance of normal relaxation suggests functional endothelial responses and myo-endothelial coordination and implicates impaired smooth muscle rather than structural limitations as responsible for contractile deficits.

109

Figure 3.3. ArntSMKO vessels have impaired vasoconstriction. Assessed by ex vivo wire myography, ArntSMKO aortic rings show decreased contractile force generation when stimulated by depolarization with (A) potassium chloride (KCl); n=7. Dose dependent vasoconstriction in response to receptor dependent agonists (B) phenylephrine (PE); n=4, and (C) angiotensin II (AngII); n=3, were also significantly reduce in ArntSMKO aortas. (A) Unpaired t-test or (B, C) Repeated measures ANOVA: *p<0.05 ArntSMKO vs. Arntlox/lox.

110

Figure 3.4. Vasorelaxation is intact in ArntSMKO. Relaxation responses to (A) acetylcholine (ACh) and (B) sodium nitroprusside (SNP) are similar in ArntSMKO and Arntlox/lox aortic segments. Results from myography are reported as a percentage of maximal phenylephrine stimulated constriction; n=3. Repeated measures ANOVA.

111

Discussion

Our current study highlights the importance of canonical HIF regulation of VSMCs

in maintaining functional vasoconstriction. Isolated primary VSMCs from ArntSMKO

mice have reduced expression of myofilament components of the contractile

machinery consistent with previously described phenotypic alteration 203.

Moreover, deletion of ARNT in VSMCs impairs aortic contractile responses to multiple agonists without affecting tunica media thickness or vasorelaxation.

Vasoregulation by contraction and relaxation is a primary function of VSMCs.

Mature VSMCs are phenotypically contractile and quiescent; however, when stimulated, VSMCs display a surprising amount of phenotypic plasticity. Increased proliferation and migration as well as decreased expression of components of the contractile apparatus are well-described markers of a phenotypic shift 39. Hypoxia

through HIF is a known driver of synthetic phenotypes in VSMCs80. Likewise, in control aortic VSMCs we see reductions in Acta2, Myh11, and Cnn1 with 24- hour

exposure to hypoxia. ArntSMKO VSMCs not only fail to demonstrate similar reductions to hypoxia, but also have significantly decreased expression of Acta2,

Myh11, and Cnn1 in normoxia relative to Arntlox/lox. These findings are consistent

with our previous report of increased proliferation and altered transcriptional profile

in ArntSMKO VSMCs. Collectively, these findings underscore the classification of

ArntSMKO VSMCs as less phenotypically contractile and more synthetic than

Arntlox/lox control VSMCs regardless of oxygen tension.

As in other muscle cells, contractile force is generated in VSMCs by the cyclic formation of actin and myosin cross bridges. Mutations in either ACTA2 or MYH11

112

have been shown to impair vasoconstriction 205. Likewise, the reduction of

contractile force generation in ArntSMKO aortas in response to membrane

depolarization and to receptor dependent agonists occurs in the context of reduced

VSMC expression of contractile genes. Maintenance of the tunica media structure

and intact vasorelaxation further support that VSMC dysfunction at the contractile

level underlies impaired vasoconstriction rather than endothelial dysregulation or

structural limitations. In a similar pattern, systemic in vivo exposure to 48 hr of

hypoxia has been shown to decrease aortic VSMC contractility due to changes in

aortic smooth muscle function206. Consistent with these findings, we now report

decreases in contractile gene expression in control VSMCs exposed to hypoxia.

Vascular remodeling driven by hypoxia and HIF mediated responses in VSMCs

have been implicated in a number of pathologies. The muscularization of

pulmonary arteries in response to chronic hypoxia resulting in pulmonary artery

hypertension is well described. Here, HIF-1α global heterozygosity or tissue

specific homozygous deletion of HIF-1α in mature VSMCs inhibits vascular

remodeling 76. In the systemic circulation, VSMC HIF-1α is necessary to maintain mural structure and function in the aorta and mesenteric arteries and mediates aortic remodeling in response to AngII 93, 97, 195. While wall structure does not

appear to be affected in ArntSMKO carotid arteries, the small vessels of the peripheral circulation are modified. In our initial characterization we reported disrupted VSMC morphology and wrapping around 25 μm feeder vessels in skeletal muscle. Moreover, when ArntSMKO mice were subjected to ischemia induced by femoral artery ligation microvascular integrity was lost in the mutant

113

mice allowing for extravasation of high molecular weight dextran into the

perivascular space. Our findings of limited vasoconstrictor activity may hinder the

redistribution of blood flow and further contribute to impairment of perfusion

recovery in this model.

Interestingly, while evidence supports roles for both ARNT and HIF-1α subunits as

regulators of VSMC contractility, functional outcomes with deletion of either subunit

differ. Huang et al. reported increased vasoconstriction in response to AngII in

mesenteric vessels from VSMC-HIF-1α KO mice while responses to other

contractile agonists were unchanged 97. When challenged with hypoxia inhalation,

Kim et al. also found evidence of increased vasoconstriction in VSMC-HIF-1α KO

mice, which featured increased right systolic ventricular pressure and increased

phosphorylation of the regulatory myosin light chain77, 198.

The ablation of HIF transcriptional activity by ARNT deletion model differs from

HIF-1α deletion in several key areas. First, loss of ARNT eliminates the

transcriptional function of both HIF-1 and HIF-2. The loss of HIF-2 activity may

contribute to the divergent contractile behavior 207. Second, ARNT deletion also disrupts any non-HIF related complexes in which ARNT participates208, 209. Finally,

HIF-1α is also known to have an array of alternative binding partners186. Deletion

models that ablate HIF activity though the α-subunit could also affect these non-

canonical pathways. These subunit specific observations help clarify the

functionality of HIF in the vasculature. As such, potential therapeutics that target

HIF functionality for vasomotor modulation should consider subunit specificity.

114

Conclusions

Collectively, our data implicate ARNT and thus canonical HIF regulation in VSMCs

as necessary for optimal vasoconstrictor function. Phenotypic changes in ArntSMKO

VSMCs characterized by reduced expression of myofilaments impair vessel

contractility. Altered vasoreactivity can have consequences for blood flow

distribution and vessel homeostasis, supporting the consideration of ARNT (in

addition to HIF-1α) as a therapeutic target.

Sources of Funding

Funding for this work was provided by the NIH F30 HL127985 (AHB), RO1

HL128281 (AP & DLR-B), RO1 HL096597 (DLR-B), T32 HL105338 (AHB), T32

GM7250 (AHB), and TL1 RR024991 (AHB).

115

Chapter 4: Discussion and Conclusions

In the preceding chapters, we have demonstrated that loss of ARNT disrupts

VSMC phenotype, changing the morphologic appearance of VSMCs around small arterioles, reducing the expression of contractile phenotype markers in isolated cells in normoxia and hypoxia, and altering proliferation and migration.

Functionally, loss of VSMC ARNT impairs vasoconstriction to multiple agonists without affecting vasorelaxation. While displaying no baseline differences in vessel number, diameter, or wall thickness, faced with an ischemic challenge in the form of femoral artery ligation, ArntSMKO mice have impaired perfusion recovery and

increased hypoxia, tissue damage, and microvascular permeability. The

consequences of ARNT deletion provide insight into the regulation of VSMCs, yet

the direct mechanism by which loss of ARNT disrupts phenotype and contractility

at the molecular level remains to be delineated.

Determinants of contraction

Fundamentally, changes in cellular contraction are attributable to alterations in the

contractile apparatus or its regulators. Reduced levels of myofilament components

could limit the number of actin-myosin cross bridges, and thus decrease force

generation. Impaired vasoconstriction in ArntSMKO vessels occurs in the context of reduced mRNA expression of both SMA and SMMHC myofilaments in VSMCs isolated from these vessels. Additional support for this explanation would be bolstered by measurements of protein content in intact vessels; however, myofilament content does not thoroughly characterize the functional state of the

116

contractile apparatus. The organization of the actin and myosin network across the

cell is precise and dynamic 20. Upon stimulation with a contractile agonist, non-

filamentous actin polymerizes, through elongation or branch addition to existing

filaments or generation of new filaments, to increases the number of available locations for cross bridging 21, 210. Organization of actin alignment improves

contractility while depolymerization of actin filaments with cytochalasin D leads to

relaxation 22, 211, 212. Overall, actin polymerization or depolymerization can be quantified by changes in ratios of F (filamentous) to G (globular) actin and in the number and organization of stress fibers visualized microscopically. Assessing changes in myofilament quantity and organization could further characterize the cause of contractile deficits. At the regulatory level, RhoA and ROCK abundance and activity are also worthy of consideration. Not only does RhoA activity contribute to maintaining phosphorylated MLC, but also the pathway participates in the polymerization of actin 213, 214. Furthermore in developmental models,

RhoA/ROCK promote VSMC differentiation and expression of contractile genes downstream or HIF activity 214, 215. These alterations in contractile apparatus

structure and function could also contribute to the morphologic changes of VSMCs

associated with arterioles.

The contractile apparatus is tethered to the cell membrane and by extension to the

extracellular matrix at focal adhesions. Without this tethering, shortening of the contractile apparatus fails to generate force 216, 217. Focal adhesions are complex structures forming physical connections between transmembrane integrins and the

117

cytoskeleton20, 218. Integrins, and thus focal adhesions, are modulated by hypoxia

and HIF. For example, hypoxia increases integrin β1 expression in mouse ES cells

219 and HIF and ARNT regulate the expression of αvβ3 integrin in focal adhesions

of trophoblast stem cells 220. This link is far less studied in VSMCs. In bovine aortic and coronary VSMCs, HIF stabilization with cobalt chloride or constitutively active

HIF-1α decreased attachment of cells in culture to a variety of extracellular matrix materials 221. No changes were seen in focal adhesion number or location, but

focal adhesion kinase phosphorylation was decreased 221. Interestingly, unlike

other isolates these VSMCs did not show increased proliferation and had

decreased migration despite increased HIF activity, raising questions about the

broad applicability of these findings. Focal adhesions serve as points of integration

for matrix to cell connections such that alterations in the matrix affect VSMC

function. Indeed, VSMCs cultured on matrix with higher Young’s modulus

generated more force without changes in phenotypic markers 222. Not only does the matrix composition impact VSMC function, but VSMCs can also impact the matrix through regulation of matrix metalloproteinases and synthesis of matrix components 223-225. All told, visual indicators of morphologic detachment seen in

ArntSMKO arteriolar VSMCs could point to changes in cell-matrix interaction either

contributing to or as a result of phenotypic changes.

Disrupted transcriptional regulation

Much of the literature describing HIFs in VSMCs centers on the function of HIF-1α

and reports the consequences of HIF-1α deletion as a model of functional HIF

118

ablation. In our studies, VSMC-ARNT-KO phenocopied reports of VSMC-HIF-1α

deletion in impairing hindlimb reperfusion after femoral artery ligation 147. However,

the phenotypes of these deletion models diverge in studies of AngII induced

vasoconstriction where VSMC-Hif-1α- KO vessels are hypercontractile while

VSMC-Arnt-KO vessels are hypocontractile compared to controls 97. Deletion of

ARNT, the common binding partner of HIF-1α, -2α, and -3α, disrupts canonical

transcriptional activity for all members of the HIF family simultaneously while the

alpha subunits, and thus any non-canonical pathways, remain intact.

HIF-1 and HIF-2 are differentially expressed and, while sharing many target genes, also have their own unique transcriptional signatures and subset of targets. Thus, individual loss of either HIF-1 or HIF-2 may differ in consequence from their

simultaneous deletion. In VSMCs, HIF-1 is thought to be the primary driver of

hypoxic responses, as indicated by proliferation; however, loss of HIF-1α could

expose underlying contributions from HIF-2 207, 226. Indeed HIF-2 has been

specifically implicated in increased vasoconstriction in PAH, which is consistent

with reports of hypercontractility in VSMC-Hif-1α deletion 77, 207. The retained function of HIF-2 in models of Hif-1α deletion could thus help explain inconsistencies in phenotype between Arnt vs Hif-1α KO models.

Non-canonical subunit activity may also contribute to phenotypic divergence.

While canonical HIF-1 (HIF-1α/HIF-1β dimer) is a master regulator of cellular

responses, interactions of HIF-1α with other proteins have also been described 186.

119

Most of these interacting proteins serve to regulate the stability of HIF-1α, but

some, NOTCH, BCL2, NEMO, and STAT3 to name but a few, are independently

described cellular regulators 227-230. ARNT’s presence for or participation in these interactions is not routinely determined. If these interactions produce meaningful changes independent of ARNT, they could help to explain the divergent phenotypes in HIF-1α and ARNT knockout models. Along similar lines, the effects of ARNT dimerization with proteins other than the HIFα subunits should also be

considered 209. ARNT was initially described, and thus named, based on its dimerization with aryl hydrocarbon receptor (AHR) 231, 232. AHR is classically activated by environmental pollutants including dioxins, polycyclic aromatic hydrocarbons, and polychlorinated biphenyls; however, naturally occurring compounds including derivatives of tryptophan, biliverdin, bilirubin, prostaglandins, and some modified low-density have also been identified as AHR ligands in mouse hepatoma cell lines 233-236. AHR deletion in hematopoietic stem cells leads to hyper proliferation followed by marrow exhaustion 237-239, and

modulation of AHR activity in endothelial and immune cells has been described in

vascular pathologies including atherosclerosis and hypertension 233, 240, 241. While

reports in VSMCs are limited, there is some indication that AHR activity plays a

role in responses to high concentrations of a uremic toxin 242-244. The impact of

AHR activity under physiologic or ischemic conditions remains to be determined; however, it seems unlikely that loss of AHR/ARNT would outweigh the effects of lost HIF activity in an ARNT deletion model of ischemia pathologies.

120

Altered intercellular communication

While originally thought to be physically isolated from each other, ECs and VSMCs are in fact physically connected by cellular structures that reach through the internal elastic lamina termed myoendothelial junctions (MEJ) 245, 246. MEJs are known to allow communication between the cells via gap junctions and NO, provided locally via eNOS 247, 248. While their contributions are yet undefined, the presence of and ribosomes within the MEJ provides an opportunity for local protein synthesis that may also contribute to signaling 246.

Along an arterial tree, MEJs are present with greater frequency in smaller vessels

249, 250. Correspondingly, the disruption of VSMCs in ArntSMKO skeletal muscle in

localizes around small feeder arterioles, suggesting that MEJs may be

compromised by the aberrant VSMC morphology or that disrupted MEJs may

contribute to VSMC dysfunction. Indeed, PAI-1 is necessary for MEJ formation,

and ArntSMKO VSMCs have decreased Pai-1 mRNA compared to controls 251.

Communication through MEJs occurs both from ECs to VSMCs and vice versa.

Altered signaling from VSMCs to ECs could also underlie the increased vascular permeability, typically a sign of EC changes, in ligated limbs of ArntSMKO mice.

Intercellular signaling that requires physical cell to cell contact also characterizes the Notch family of transcription factors. Anchored within the plasma membranes of neighboring cells, interactions between Notch ligands (Delta-like 1, 3, and 4,

Jagged 1 and 2) and receptors (Notch 1-4) trigger the cleavage of the transcriptionally active intracellular domain of the Notch receptor by gamma

121

secretase 252. Suppression of Notch activity in VSMCs by introduction of a dominant negative cofactor, alters developmental blood vessel patterning impairs vasoconstriction and vasorelaxation, and compromises angiogenesis 164, 204, 253,

254. Notch has also been implicated in VSMC phenotype switching 58. Several reports show interaction between the Notch and HIF pathways 255-259. These reports describe interactions between HIF-1α or HIF-2α and the Notch intracellular domain, which would remain intact in our ARNT deletion model. In contrast, disruption of Notch signaling by physical separation of VSMCs and/or loss of function in regulatory pathways that require both HIF and Notch could contribute to the ArntSMKO phenotype.

Contributions of other potentially affected cell types

VSMCs wrap vessels from the aorta to small arterioles; however, the smallest vessels, namely pre-capillary arterioles, capillaries, and post capillary venules, are not in contact with VSMCs. Instead, the mural cell populations around these

vessels are provided by pericytes. These cells have been most extensively studied

in the blood brain barrier where loss of pericytes correlates with increased

permeability 260, 261. In the , VSMCs differentiate from cells with

pericytes markers 262. While SM22α is typically thought to be SMC restricted,

SM22α positive pericytes have been identified in the developing retina 263. The

SM22α-Cre driver we employ is not expressed identically with native SM22α 163;

however, closer examination of Cre activity in the pericyte population is warranted.

Given their role in maintaining endothelial monolayer integrity in the blood brain

122

barrier, pericyte dysregulation could contribute to the increased vascular

permeability seen in ligated limbs of ArntSMKO mice.

Cells of the myeloid lineage also warrant assessment for off target Cre activity.

Two SM22α-Cre lines from The Jackson Laboratory have evidence of Cre mediated deletion in neutrophils, monocytes, and macrophages 264. HIF activity in

macrophages can stimulate arteriogenic remodeling of collaterals in the hind limb

123. Thus, dysregulation of macrophage contributions could confound the

mechanistic interpretation of impaired perfusion phenotype. As stated above, the

expression of specific SM22α-Cre drivers can vary from native expression and

between different lines. Cre activity in blood cells was not examined in Lepore et.

al.’s initial characterization of the SM22α-Cre used in our studies. While worthy of

consideration, arteriogenesis is not the factor limiting perfusion recovery in

ArntSMKO mice subjected to HLI, thus reducing the likelihood that this could

compromise our findings.

Implications for other vascular pathologies

In the previous chapters, we examined the consequences of VSMC-ARNT-KO in

peripheral ischemia and aortic reactivity, but VSMC dysfunction contributes to

many other pathologies.

123

Pulmonary Artery Hypertension

Pulmonary artery hypertension (PAH), for example, has various distinct etiologies

all of which manifest in vascular remodeling and sustained vasoconstriction 78.

Several of these pathogenic conditions are associated with hypoxic environmental changes including chronic obstructive pulmonary disease and high altitude exposure. Furthermore, mouse models of exposure to less than atmospheric levels of O2 in inhaled air reveal pulmonary changes that phenotypically approximate

PAH 265. With such a strong hypoxic link, it is no surprise that HIFs have been

implicated in the pathogenesis of PAH. Global heterozygosity of either HIF-1α or

HIF-2α protects against hypoxia induced PAH in mice 191, 192, and the levels of HIF-

1α correspond with the extent of remodeling and severity of disease in rats 266.

Interestingly, the role of HIF-1α in VSMCs remains controversial, with pulmonary artery pressure reported to either increase or decrease with VSMC-HIF-1α deletion

198, 267. Accordingly, these findings would suggest that HIF-1α has a regulatory role

in promoting or limiting vasoconstriction and/or remodeling. As for the role of

ARNT, only the increased proliferation of ArntSMKO VSMCs would be consistent

with an increased remodeling phenotype, while decreased vasoconstriction in

ArntSMKO mice, reduced hypoxic migration of isolated ArntSMKO VSMCs, and the

preponderance of data supporting HIF activity as a driver of increased vascular

remodeling suggest that ArntSMKO mice would be protected from hypoxia induced

PAH. ArntSMKO mice subjected to models of induced PAH would be an important extension of our analysis relevant to clarifying mechanism of action.

124

Hypertension

Changes in vasoconstriction with hypoxia exposure are key pathologic findings in

PAH, and, as such, many of the pharmacologic interventions stimulate vasodilation

to relieve the increased vascular resistance 201. In the systemic circulation,

changes in vasoreactivity and peripheral resistance contribute to hypertension.

HIF-1α has also been implicated in regulating systemic vasoreactivity and thus

blood pressure. VSMC specific HIF-1α deletion increased systolic blood pressure

and vasoconstriction of mesenteric arteries stimulated by AngII 97. This is

consistent with the paradigm that hypoxia stimulates vasodilation in the systemic

vasculature, though interestingly, measurement of blood pressure by tail cuff,

rather than surgically placed aortic probe, reported no difference in systolic blood

pressure in either inducible or developmental models of VSMC HIF-1α deletion 267.

Our data demonstrate changes in systemic vasoreactivity with ARNT deletion, though notably, we report reduced rather than augmented vasoconstriction.

Arguably, our observations were made an elastic vessel, the aorta, rather than a resistance vessel, the mesenteric artery. Indeed conduit and resistance vessels have different structural compositions and physiologic functions 268. However, a

trend toward increased or decreased contractility in the aorta is often observed in

the resistance vessels 204, 269.

Aneurysm and dissection

In larger elastic vessels, alterations in VSMC phenotype are associated aneurysm and dissection 270-272. In Marfan syndrome, where a genetic mutation in fibrillin-1

125

underlies aneurysmal dilation of the vessel wall, aortic VSMCs display a

hypercontractile phenotype. Increased stress fibers, focal adhesions, and

exaggerated expression of contractile phenotype markers including SMA,

smoothelin, SM22α, and calponin-1 in addition to collagen I and myocardin were

identified in VSMCs from dilated vessel segments and in non-dilated areas, though

to a lesser degree 273. In contrast, shifts away from the contractile phenotype in

VSMCs are more common in aneurysm and dissection. Mutations in components of the VSMC contractile apparatus increase risk for both aortic aneurysm and dissection 271. Correspondingly, VSMCs isolated from human aortic aneurysms

had decreased staining for SM22α and SMA and increased matrix

metalloproteinase (MMP) -2 and -9 274, 275. In a mouse aneurysm model using

intraluminal elastase treatment, VSMCs associated with aortic dilations also had

decreased SM22 and SMA and increased MMPs 275. Moreover, VSMCs bordering

aortic dissections also featured decreased expression of contractile proteins and

markers and when isolated, have an increased proliferative phenotype 276, 277. HIF

has been implicated in the progression of aneurysm. Expression of HIF-1α and

MMPs are increased in the VSMCs of abdominal aortic aneurysms 92.

Correspondingly, chemical stabilization of HIF increased the incidence of

abdominal aortic aneurysm while global chemical inhibition of HIF activity

decreased aneurysm formation in a mouse model of AngII induced aortic

aneurysm 278. Seemingly in contrast, SMC specific HIF-1α knockout augmented

aneurysm formation and disruption of elastin fibers compared with controls when

challenged in vivo with β-aminopropionitrile and AngII 195. Yet studies by the same

126

group reported that VSMC Hif-1α knockout decreased AngII induced aortic

remodeling 93. In all, it would be reasonable to suggest that ArntSMKO mice may

exhibit an altered response in an aneurysm model. While the lack of consensus in

the HIF data complicates projections, synthetic phenotype and impaired

vasoconstriction in ArntSMKO would suggest that loss of ARNT may increase

susceptibility to aneurysm.

Atherosclerosis

Atherosclerotic disease affects the structure and composition of the vascular wall.

Not only does it underlie PAD, but also its stenotic and thrombotic consequences in coronary and carotid artery diseases can precipitate and respectively. While a complex disease involving many cell types, modulation of VSMC phenotype plays an important role in atherosclerotic lesion development and stability. Lesion formation is characterized by lipid accumulation in the sub intimal layer of the vessel wall. VSMCs of the tunica media proliferate and migrate into the intima forming a fibrous cap between the lipid pocket and the vessel lumen

279. The cells of this neointima display significant characteristics of synthetic VSMC phenotype including reduced expression of contractile proteins, SMMHC, SMA, and calponin, loss of myofilaments, alterations in contractility, increased DNA synthesis, and altered cell morphology 39, 280-287. VSMC migration into the fibrous cap is beneficial, as cap disruption is associated with plaque instability, which often precedes plaque rupture and thrombus formation 288, 289. VSMCs that remain within the tunica media are also affected by lipid accumulation. In the lipid core of the

127

plaque, VSMCs can take up LDL and become foam cells losing their VSMC

markers and acquiring monocyte markers 279. HIFs in VSMCs have been

implicated in the progression of atherosclerosis. Samples of lesions from human

carotid arteries show signs of hypoxia and stabilized HIF-1α localized to cells

bordering the necrotic core, including VSMCs 290, 291. HIF also appears to play a

role in lipid uptake by VSMCs. Hypoxia through HIF-1 activity increases expression

of low density receptor related protein 1 that allows for internalization

of aggregated LDL 292. The effects of VSMC HIF-1 inhibition on disease

progression are less clear. In a mouse model, VSMC specific knockout of Hif-1α in an ApoE null background reduced deposition of lipid and lesion size on a high fat diet 293. However, loss of the HIF target OCT4, a regulator of pluripotency in

embryonic stem cells, in VSMCs increased lesion size 51. In a non-targeted model,

IV injection of stabilized HIF-1α decreased lesion size in the ApoE null background

294. This lack of consensus once again makes predicting effects of VSMC-ARNT

difficult. The prevalent synthetic phenotype of ArntSMKO VSMCs could promote the

growth of larger plaques due to increased proliferation. Alternatively, the lack of

further changes in proliferation or migration of ArntSMKO VSMCs in response to

hypoxia, suggests they may not dramatically respond to the pathologic plaque

microenvironment, thus potentially resulting in decreased lesion size, consistent

with VSMC-Hif-1α-KO models.

128

Potential for translation to human disease

Arterial circulation

To determine the translatability of our findings to human disease we obtained

primary cultures of human iliac artery VSMCs from a single apparently healthy

donor through the National Institute on Aging. Exposure of these cells to hypoxia

(2% O2) for 24 hours altered transcriptional expression of phenotypic modulators.

Relative to normoxia and normalized to 18s, expression of HIF1A decreased in hypoxia while VEGFA increased consistent with increased HIF1 activity (Figure

4.1 A, B). Expression of PAI1 and THBS1 also increased, PAI1 significantly so, while no change was seen in PLAUR (Figure 4.1C-E). Changes in transcription of

Thbs1 and Pai1 were also seen in the murine aortic VSMCs (Chapter 2), though interestingly they were decreased rather than increased in response to hypoxia.

Some of these differing responses may be attributable to the diverse vascular origins of the VSMCs (aortic vs iliac), different durations in culture (direct isolation vs passage >25) and species specificity (mouse vs human). Functionally, 48 hours of hypoxia increased proliferation in human iliac artery VSMCs mirroring the proliferative response to hypoxia in murine VSMCs, although not sufficient for statistical significance (p < 0.07) (Figure 4.1F). Collectively, similar phenotypic responses to hypoxia, though not necessarily identical on a gene-by-gene basis inspire interest to further interrogate translatable correlations with human disease.

A more complete assessment of transcriptional and protein changes including

VSMC marker genes, examination of isolated VSMCs from additional individuals,

129

Figure 4.1. Effects of hypoxia exposure on human iliac artery VSMCs. Following 24 hour exposure to hypoxia (2% O2) human iliac artery VSMCs have decreased mRNA expression of (A) HIF1A, increased expression of (B)VEGFA, (C)PAI1, (E)THBS1, and no change in (D) PLAUR. (F) Proliferation measured by MTT assay was also increased by 48 hour hypoxia exposure though not to a statistically significant degree. n=3; 2 tailed t-test: ^p<0.05 hypoxia vs. normoxia.

130

and evaluation of cells with less duration in culture (<25 passages), would

considerably bolster findings and provide a framework for translation.

Venous circulation

Diseases of the peripheral vasculature are not limited to the arterial side of the

network. Venous pathologies, including varicosities and vein graft stenosis, are also thought to have VSMC alterations and hypoxic contributions295-300. Isolated

primary iliac vein VSMCs, from the same donor as the iliac artery cells, were also

exposed to 24 hours of hypoxia. They too had significant decreases in HIF1A

expression and increases in VEGFA indicative of HIF1 activity (Figure 4.2A, B).

Changes were also seen in PAI1, THBS1, and PLAUR, though interestingly all

were decreased relative to normoxia, PLAUR significantly so (Figure 4.2C-E).

Compared to increases seen in arterial cells, differential responses in venous

VSMCs could be attributable to the different baseline hemodynamic and oxygen

environment of the venous circulation. Venous cells also show increased

proliferation after 48hours in hypoxia relative to normoxia (p < 0.06) (Figure 4.2F).

While peripheral artery and vein VSMCs both show increased proliferation in

hypoxia, they differ in their ability to survive oxidative stress. When challenged with

600μM hydrogen peroxide in DMEM-F12 +5% FBS media for 24 hours, venous

VSMCs showed complete loss of cell viability by MTT assay in both normoxia and

hypoxia (Figure 4.3A). In contrast, arterial VSMCs under normoxia showed 30%

survival after the same stress, and further increased to 70% under hypoxic

conditions (Figure 4.3A). Interestingly, the loss of viability could be rescued in both

131

cell types under normoxia and hypoxia by culturing in EGM2-MV media rather than

DMEMF12 (Figure 4.3B). EGM2-MV is supplemented with hydrocortisone, ascorbic acid, and growth factors, namely VEGF, FGF-2, IGF-1, and EGF in addition to FBS. This suggests that in the right growth factor milieu, both iliac artery and vein VSMCs can survive high levels of oxidative stress. Taken together, this data demonstrates that iliac vein VSMCs also respond to hypoxia by increasing proliferation and altering transcription of phenotypic modulators, though in a pattern different from iliac artery VSMCs. Moreover, venous cells are more susceptible to oxidative stress when not in the presence of environmental enrichment than their arterial counterparts. Thus, venous VSMCs and their associated pathologies, would benefit from separate consideration in light of their deviation from arterial responses.

132

Figure 4.2. Effects of hypoxia exposure on human iliac vein VSMCs. mRNA collected from human iliac vein VSMCs following 24 hour exposure to normoxia or hypoxia (2%O2) and normalized to 18s, displayed decreased expression of (A) HIF1A, and increased expression of (B) VEGFA. Expression of (C) PAI1, (D) PLAUR, and (E) THBS1 were all reduced, PLAUR to a significant degree. (F) Exposure to hypoxia for 48 hours increases proliferation measured by MTT assay but not to a significant degree. n=3; 2 tailed t-test: ^p<0.05 hypoxia vs. normoxia.

133

Figure 4.3. Iliac vein smooth muscle cells are more susceptible to H2O2 induced apoptosis. ILA and ILV VSMC were treated with 600µM H2O2 in (a) DMEM-F12 +5% FBS or (b) EGM2-MV and incubated for 24hours in normoxia or hypoxia (2%O2). Cell numbers were measured using MTT assay and reported as ratios of optical density at 24 hours / time 0. (A) In minimal media, hypoxia increased the survival of ILA cells while no viable cells were detectable in venous cultures under either normoxia or hypoxia. (B) Culture in media enriched with growth factors removed the benefit of hypoxia in arterial cells and rescued venous cultures. n=3, one-way ANOVA: *p<0.05 arterial vs. venous VSMCs, ^p<0.05 hypoxia vs. normoxia.

134

Closing

HIFs are integrally connected master regulators of cellular responses to oxygen tension. Novel insights into the role of canonical HIF modulation shed light not only on the importance of HIFs in VSMCs but also on how crucial appropriate VSMC responses are to maintaining vascular function. These findings have implications for PAD, a major cause of morbidity, and moreover, for vascular diseases affecting

nearly every . Further examination of the interplay between cellular

participants, including VSMCs, ECs, and non-vascular populations, and the molecular mechanisms driving these responses would improve understanding of pathophysiology and thus therapeutic design. Harnessing HIFs’ regulatory role could allow for improved vessel regulation and tissue perfusion, ameliorating diseases that take the lives of millions worldwide.

135

Future Directions

While there are many potential avenues for continued investigation, the most

immediate focus of future studies would be to determine the molecular

underpinnings of impaired organization and contractility in ArntSMKO vessels and

the implications of these changes on vascular homeostasis. The data reported in

Chapter 3 demonstrate reduced mRNA expression of contractile genes in isolated

ArntSMKO VSMCs. Further assessment of myofilament abundance at the protein

level in these isolated cells and in intact vessels would provide valuable insights.

Alterations in myosin, actin, and/or their regulators may contribute to the observed

phenotype. Contractile regulation of myosin activity converges on the

phosphorylation and dephosphorylation of MLC; quantifying the relative

abundance of MLC-P in ArntSMKO and Arntlox/lox vessels after contractile agonist stimulation would assess for altered activity in regulatory pathways. Further upstream, relative abundance of MLCK, MLCP, and their regulators, including

CaCm and ROCK respectively, as well as quantification of their individual contributions through the use of enzyme specific inhibitors on contractile agonist stimulated vessels or even isolated cells, could help determine the pathways implicated in any altered MLC phosphorylation status. Abnormalities in VSMC actin could also result in impaired vasoconstriction. While challenging to assess in intact vessels, actin organization could be evaluated microscopically in isolated cells by labeling SMA. Studies would assess for disparities in actin structure at baseline and following agonist stimulation in ArntSMKO and Arntlox/lox VSMCs. In a similar

assay, the polymerization status of actin could also be determined though

136

comparisons of F/G actin ratios. The contributions of altered RhoA/ROCK activity

to any aberrant actin structure should also be assessed in the Arnt deficient

VSMCs. Evaluation of relative abundance of phosphorylated and total RhoA and

ROCK as well as responses to pathway inhibition by small molecule ROCK inhibitors or dominant negative RhoA would quantify the importance of the

RhoA/ROCK pathway to VSMC phenotype and function.

Additional studies of vessel function could also strengthen conclusions. Regulation of perfusion distribution and blood pressure principally occurs at the level of resistance vessels, thus additional myographic studies in the muscular arteries of the mesentery would more directly evaluate physiologically relevant vasoreactivity.

In light of altered peripheral perfusion, myographic assessment of vessels from the skeletal muscle vasculature would also be of interest. Given the requirement of

ARNT for the hypoxic mediated responses by HIFs, stabilization of HIF-alpha subunits with PHD inhibitors or sparging buffers with oxygen depleted gas mixtures, could provide novel insights to the intrinsic role of vasoreactivity by vessels in light of hypoxic conditions. These studies would be particularly interesting when comparing the responses of pulmonary arteries, which vasoconstrict in response to hypoxia, to arteries of the systemic vasculature, which feature hypoxic vasodilation. Meaningfully, the aberrant normoxic phenotype in

ArntSMKO VSMCs and vessels could be confounded during embryonic

development. Utilization of a SMC specific inducible Cre-recombinase driver such

as SMMHC-CreERT2 mouse line would allow comparison of phenotypes in

developmental vs adult deletion. Finally, alterations in vasoreactivity can impact

137 blood pressure homeostasis, thus catheter based analysis of intravascular blood pressure at baseline and in response to vasoconstrictors would assess for systemic in vivo sequela of altered vasoconstriction as a consequence of VSMC-

ARNT deletion.

138

References

1. Carmeliet P, Jain RK. Angiogenesis in cancer and other diseases. Nature.

2000;407:249-257

2. Mitchell RN. Blood vessels. In: Kumar V, Abbas AK, Aster JC, eds. Robbins

and cotran pathologic basis of disease. Philadelphia, PA:

Elsevier/Saunders; 2015:483-522.

3. Stenmark KR, Yeager ME, El Kasmi KC, Nozik-Grayck E, Gerasimovskaya

EV, Li M, Riddle SR, Frid MG. The adventitia: Essential regulator of vascular

wall structure and function. Annu Rev Physiol. 2013;75:23-47

4. Sinha S, Iyer D, Granata A. Embryonic origins of human vascular smooth

muscle cells: Implications for in vitro modeling and clinical application. Cell

Mol Life Sci. 2014;71:2271-2288

5. Majesky MW. Developmental basis of vascular smooth muscle diversity.

Arterioscler Thromb Vasc Biol. 2007;27:1248-1258

6. Waldo KL, Hutson MR, Ward CC, Zdanowicz M, Stadt HA, Kumiski D, Abu-

Issa R, Kirby ML. Secondary heart field contributes myocardium and

smooth muscle to the arterial pole of the developing heart. Dev Biol.

2005;281:78-90

7. Le Lievre CS, Le Douarin NM. Mesenchymal derivatives of the neural crest:

Analysis of chimaeric quail and chick embryos. J Embryol Exp Morphol.

1975;34:125-154

8. Kirby M, Gale T, Stewart D. Neural crest cells contribute to normal

aorticopulmonary septation. Science. 1983;220:1059-1061

139

9. Esner M, Meilhac SM, Relaix F, Nicolas J-F, Cossu G, Buckingham ME.

Smooth muscle of the dorsal aorta shares a common clonal origin with

skeletal muscle of the myotome. Development. 2006;133:737-749

10. Wasteson P, Johansson BR, Jukkola T, Breuer S, Akyurek LM, Partanen J,

Lindahl P. Developmental origin of smooth muscle cells in the descending

aorta in mice. Development. 2008;135:1823-1832

11. Murphy ME, Carlson EC. An ultrastructural study of developing extracellular

matrix in vitelline blood vessels of the early chick embryo. Am J Anat.

1978;151:345-375

12. Greif DM, Kumar M, Lighthouse JK, Hum J, An A, Ding L, Red-Horse K,

Espinoza FH, Olson L, Offermanns S, Krasnow MA. Radial construction of

an arterial wall. Dev Cell. 2012;23:482-493

13. Mikawa T, Gourdie RG. Pericardial mesoderm generates a population of

coronary smooth muscle cells migrating into the heart along with ingrowth

of the epicardial organ. Dev Biol. 1996;174:221-232

14. Mikawa T, Fischman DA. Retroviral analysis of cardiac morphogenesis:

Discontinuous formation of coronary vessels. Proceedings of the National

Academy of Sciences. 1992;89:9504-9508

15. Sainz J, Al Haj Zen A, Caligiuri G, Demerens C, Urbain D, Lemitre M, Lafont

A. Isolation of “side population” progenitor cells from healthy arteries of adult

mice. Arterioscler Thromb Vasc Biol. 2006;26:281-286

140

16. Hu Y, Zhang Z, Torsney E, Afzal AR, Davison F, Metzler B, Xu Q. Abundant

progenitor cells in the adventitia contribute to atherosclerosis of vein grafts

in apoe-deficient mice. J Clin Invest. 2004;113:1258-1265

17. Webb RC. Smooth and relaxation. Adv Physiol Educ.

2003;27:201-206

18. Fatigati V, Murphy RA. Actin and tropomyosin variants in smooth muscles.

Dependence on tissue type. J Biol Chem. 1984;259:14383-14388

19. Dominguez R, Freyzon Y, Trybus KM, Cohen C. Crystal structure of a

vertebrate smooth muscle myosin motor domain and its complex with the

essential light chain: Visualization of the pre-power stroke state. Cell.

1998;94:559-571

20. Yamin R, Morgan KG. Deciphering actin cytoskeletal function in the

contractile vascular smooth muscle cell. J Physiol. 2012;590:4145-4154

21. Rembold CM, Tejani AD, Ripley ML, Han S. Paxillin phosphorylation, actin

polymerization, noise temperature, and the sustained phase of swine

carotid artery contraction. Am J Physiol Cell Physiol. 2007;293:C993-1002

22. Cipolla MJ, Gokina NI, Osol G. Pressure-induced actin polymerization in

vascular smooth muscle as a mechanism underlying myogenic behavior.

FASEB J. 2002;16:72-76

23. Ogut O, Brozovich FV. Regulation of force in vascular smooth muscle. J

Mol Cell Cardiol. 2003;35:347-355

24. Brozovich FV, Nicholson CJ, Degen CV, Gao YZ, Aggarwal M, Morgan KG.

Mechanisms of vascular smooth muscle contraction and the basis for

141

pharmacologic treatment of smooth muscle disorders. Pharmacol Rev.

2016;68:476-532

25. Bayliss WM. On the local reactions of the arterial wall to changes of internal

pressure. The Journal of Physiology. 1902;28:220-231

26. Amberg GC, Navedo MF. Calcium dynamics in vascular smooth muscle.

Microcirculation. 2013;20:281-289

27. Klabunde RE. Cardiovascular physiology concepts. Baltimore [Md.]:

Lippincott Williams & Wilkins/Wolters Kluwer; 2012.

28. Mehta PK, Griendling KK. Angiotensin ii cell signaling: Physiological and

pathological effects in the cardiovascular system. Am J Physiol Cell Physiol.

2007;292:C82-97

29. Furchgott RF, Zawadzki JV. The obligatory role of endothelial cells in the

relaxation of arterial smooth muscle by acetylcholine. Nature.

1980;288:373-376

30. Rees DD, Palmer RM, Hodson HF, Moncada S. A specific inhibitor of nitric

oxide formation from l-arginine attenuates endothelium-dependent

relaxation. Br J Pharmacol. 1989;96:418-424

31. Godo S, Sawada A, Saito H, Ikeda S, Enkhjargal B, Suzuki K, Tanaka S,

Shimokawa H. Disruption of physiological balance between nitric oxide and

endothelium-dependent hyperpolarization impairs cardiovascular

homeostasis in mice. Arterioscler Thromb Vasc Biol. 2016;36:97-107

32. Shimokawa H, Yasutake H, Fujii K, Owada MK, Nakaike R, Fukumoto Y,

Takayanagi T, Nagao T, Egashira K, Fujishima M, Takeshita A. The

142

importance of the hyperpolarizing mechanism increases as the vessel size

decreases in endothelium-dependent relaxations in rat mesenteric

circulation. J Cardiovasc Pharmacol. 1996;28:703-711

33. Gabbiani G, Schmid E, Winter S, Chaponnier C, de Ckhastonay C,

Vandekerckhove J, Weber K, Franke WW. Vascular smooth muscle cells

differ from other smooth muscle cells: Predominance of vimentin filaments

and a specific alpha-type actin. Proc Natl Acad Sci U S A. 1981;78:298-302

34. Hungerford JE, Owens GK, Argraves WS, Little CD. Development of the

aortic vessel wall as defined by vascular smooth muscle and extracellular

matrix markers. Dev Biol. 1996;178:375-392

35. Miano JM, Cserjesi P, Ligon KL, Periasamy M, Olson EN. Smooth muscle

myosin heavy chain exclusively marks the smooth muscle lineage during

mouse embryogenesis. Circ Res. 1994;75:803-812

36. Duband JL, Gimona M, Scatena M, Sartore S, Small JV. Calponin and sm

22 as differentiation markers of smooth muscle: Spatiotemporal distribution

during avian . Differentiation. 1993;55:1-11

37. Miano JM, Carlson MJ, Spencer JA, Misra RP. Serum response factor-

dependent regulation of the smooth muscle calponin gene. J Biol Chem.

2000;275:9814-9822

38. Kim S, Ip HS, Lu MM, Clendenin C, Parmacek MS. A serum response

factor-dependent transcriptional regulatory program identifies distinct

smooth muscle cell sublineages. Mol Cell Biol. 1997;17:2266-2278

143

39. Owens GK, Kumar MS, Wamhoff BR. Molecular regulation of vascular

smooth muscle cell differentiation in development and disease. Physiol

Rev. 2004;84:767-801

40. Gomez D, Owens GK. Smooth muscle cell phenotypic switching in

atherosclerosis. Cardiovasc Res. 2012;95:156-164

41. Holycross BJ, Blank RS, Thompson MM, Peach MJ, Owens GK. Platelet-

derived growth factor-bb-induced suppression of smooth muscle cell

differentiation. Circ Res. 1992;71:1525-1532

42. Li X, Van Putten V, Zarinetchi F, Nicks ME, Thaler S, Heasley LE, Nemenoff

RA. Suppression of smooth-muscle alpha-actin expression by platelet-

derived growth factor in vascular smooth-muscle cells involves ras and

cytosolic phospholipase a2. Biochem J. 1997;327 ( Pt 3):709-716

43. Corjay MH, Thompson MM, Lynch KR, Owens GK. Differential effect of

platelet-derived growth factor- versus serum-induced growth on smooth

muscle alpha-actin and nonmuscle beta-actin mrna expression in cultured

rat aortic smooth muscle cells. J Biol Chem. 1989;264:10501-10506

44. Blank RS, Owens GK. Platelet-derived growth factor regulates actin isoform

expression and growth state in cultured rat aortic smooth muscle cells. J

Cell Physiol. 1990;142:635-642

45. Dandré F, Owens GK. Platelet-derived growth factor-bb and ets-1

transcription factor negatively regulate transcription of multiple smooth

muscle cell differentiation marker genes. American Journal of Physiology-

Heart and Circulatory Physiology. 2004;286:H2042-H2051

144

46. Wang D, Chang PS, Wang Z, Sutherland L, Richardson JA, Small E, Krieg

PA, Olson EN. Activation of cardiac gene expression by myocardin, a

transcriptional cofactor for serum response factor. Cell. 2001;105:851-862

47. Miano JM. Serum response factor: Toggling between disparate programs

of gene expression. J Mol Cell Cardiol. 2003;35:577-593

48. Yoshida T, Sinha S, Dandré F, Wamhoff BR, Hoofnagle MH, Kremer BE,

Wang D-Z, Olson EN, Owens GK. Myocardin is a key regulator of carg-

dependent transcription of multiple smooth muscle marker genes. Circ Res.

2003;92:856-864

49. Du KL, Ip HS, Li J, Chen M, Dandre F, Yu W, Lu MM, Owens GK, Parmacek

MS. Myocardin is a critical serum response factor cofactor in the

transcriptional program regulating smooth muscle cell differentiation. Mol

Cell Biol. 2003;23:2425-2437

50. Yoshida T, Kawai-Kowase K, Owens GK. Forced expression of myocardin

is not sufficient for induction of smooth muscle differentiation in

multipotential embryonic cells. Arterioscler Thromb Vasc Biol.

2004;24:1596-1601

51. Cherepanova OA, Gomez D, Shankman LS, Swiatlowska P, Williams J,

Sarmento OF, Alencar GF, Hess DL, Bevard MH, Greene ES, Murgai M,

Turner SD, Geng Y-J, Bekiranov S, Connelly JJ, Tomilin A, Owens GK.

Activation of the esc pluripotency factor oct4 in smooth muscle cells is

atheroprotective. Nat Med. 2016;22:657-665

145

52. Yoshida T, Kaestner KH, Owens GK. Conditional deletion of kruppel-like

factor 4 delays downregulation of smooth muscle cell differentiation

markers but accelerates neointimal formation following vascular injury. Circ

Res. 2008;102:1548-1557

53. Liu Y, Sinha S, McDonald OG, Shang Y, Hoofnagle MH, Owens GK.

Kruppel-like factor 4 abrogates myocardin-induced activation of smooth

muscle gene expression. J Biol Chem. 2005;280:9719-9727

54. Turner EC, Huang CL, Govindarajan K, Caplice NM. Identification of a klf4-

dependent upstream repressor region mediating transcriptional regulation

of the myocardin gene in human smooth muscle cells. Biochim Biophys

Acta. 2013;1829:1191-1201

55. Xie C, Guo Y, Zhu T, Zhang J, Ma PX, Chen YE. Yap1 protein regulates

vascular smooth muscle cell phenotypic switch by interaction with

myocardin. J Biol Chem. 2012;287:14598-14605

56. Wang L, Luo JY, Li B, Tian XY, Chen LJ, Huang Y, Liu J, Deng D, Lau CW,

Wan S, Ai D, Mak KK, Tong KK, Kwan KM, Wang N, Chiu JJ, Zhu Y, Huang

Y. Integrin-yap/taz-jnk cascade mediates atheroprotective effect of

unidirectional shear flow. Nature. 2016;540:579–582

57. Morrow D, Guha S, Sweeney C, Birney Y, Walshe T, O’Brien C, Walls D,

Redmond EM, Cahill PA. Notch and vascular smooth muscle cell

phenotype. Circ Res. 2008;103:1370-1382

146

58. Proweller A, Pear WS, Parmacek MS. Notch signaling represses

myocardin-induced smooth muscle cell differentiation. J Biol Chem.

2005;280:8994-9004

59. Tang Y, Urs S, Liaw L. Hairy-related transcription factors inhibit notch-

induced smooth muscle α-actin expression by interfering with notch

intracellular domain/cbf-1 complex interaction with the cbf-1–binding site.

Circ Res. 2008;102:661-668

60. Jin S, Hansson EM, Tikka S, Lanner F, Sahlgren C, Farnebo F, Baumann

M, Kalimo H, Lendahl U. Notch signaling regulates platelet-derived growth

factor receptor-β expression in vascular smooth muscle cells. Circ Res.

2008;102:1483-1491

61. Wang GL, Semenza GL. General involvement of hypoxia-inducible factor 1

in transcriptional response to hypoxia. Proc Natl Acad Sci U S A.

1993;90:4304-4308

62. Semenza GL, Wang GL. A nuclear factor induced by hypoxia via de novo

protein synthesis binds to the human erythropoietin gene enhancer at a site

required for transcriptional activation. Mol Cell Biol. 1992;12:5447-5454

63. Pugh CW, Tan CC, Jones RW, Ratcliffe PJ. Functional analysis of an

oxygen-regulated transcriptional enhancer lying 3' to the mouse

erythropoietin gene. Proc Natl Acad Sci U S A. 1991;88:10553-10557

64. Tian H, McKnight SL, Russell DW. Endothelial pas domain protein 1

(epas1), a transcription factor selectively expressed in endothelial cells.

Genes Dev. 1997;11:72-82

147

65. Gu YZ, Moran SM, Hogenesch JB, Wartman L, Bradfield CA. Molecular

characterization and chromosomal localization of a third alpha-class

hypoxia inducible factor subunit, hif3alpha. Gene Expr. 1998;7:205-213

66. Kallio PJ, Pongratz I, Gradin K, McGuire J, Poellinger L. Activation of

hypoxia-inducible factor 1alpha: Posttranscriptional regulation and

conformational change by recruitment of the arnt transcription factor. Proc

Natl Acad Sci U S A. 1997;94:5667-5672

67. Giaccia AJ, Simon MC, Johnson R. The biology of hypoxia: The role of

oxygen sensing in development, normal function, and disease. Genes Dev.

2004;18:2183-2194

68. Lando D, Peet DJ, Whelan DA, Gorman JJ, Whitelaw ML. Asparagine

hydroxylation of the hif transactivation domain a hypoxic switch. Science.

2002;295:858-861

69. Richard DE, Berra E, Pouyssegur J. Nonhypoxic pathway mediates the

induction of hypoxia-inducible factor 1alpha in vascular smooth muscle

cells. J Biol Chem. 2000;275:26765-26771

70. Black SM, DeVol JM, Wedgwood S. Regulation of fibroblast growth factor-

2 expression in pulmonary arterial smooth muscle cells involves increased

reactive oxygen species generation. American Journal of Physiology - Cell

Physiology. 2008;294:C345-C354

71. Chang H, Shyu KG, Wang BW, Kuan P. Regulation of hypoxia-inducible

factor-1alpha by cyclical mechanical stretch in rat vascular smooth muscle

cells. Clin Sci (Lond). 2003;105:447-456

148

72. Iyer NV, Kotch LE, Agani F, Leung SW, Laughner E, Wenger RH,

Gassmann M, Gearhart JD, Lawler AM, Yu AY, Semenza GL. Cellular and

developmental control of o2 homeostasis by hypoxia-inducible factor 1

alpha. Genes Dev. 1998;12:149-162

73. Maltepe E, Schmidt JV, Baunoch D, Bradfield CA, Simon MC. Abnormal

angiogenesis and responses to glucose and oxygen deprivation in mice

lacking the protein arnt. Nature. 1997;386:403-407

74. Kozak KR, Abbott B, Hankinson O. Arnt-deficient mice and placental

differentiation. Dev Biol. 1997;191:297-305

75. Andrikopoulou E, Zhang X, Sebastian R, Marti G, Liu L, Milner SM, Harmon

JW. Current insights into the role of hif-1 in cutaneous wound healing. Curr

Mol Med. 2011;11:218-235

76. Ball MK, Waypa GB, Mungai PT, Nielsen JM, Czech L, Dudley VJ, Beussink

L, Dettman RW, Berkelhamer SK, Steinhorn RH, Shah SJ, Schumacker PT.

Regulation of hypoxia-induced pulmonary hypertension by vascular smooth

muscle hypoxia-inducible factor-1alpha. Am J Respir Crit Care Med.

2014;189:314-324

77. Kim YM, Barnes EA, Alvira CM, Ying L, Reddy S, Cornfield DN. Hypoxia-

inducible factor-1alpha in pulmonary artery smooth muscle cells lowers

vascular tone by decreasing myosin light chain phosphorylation. Circ Res.

2013;112:1230-1233

78. Shimoda LA, Laurie SS. Vascular remodeling in pulmonary hypertension. J

Mol Med (Berl). 2013;91:297-309

149

79. Pak O, Aldashev A, Welsh D, Peacock A. The effects of hypoxia on the cells

of the pulmonary vasculature. Eur Respir J. 2007;30:364-372

80. Schultz K, Fanburg BL, Beasley D. Hypoxia and hypoxia-inducible factor-

1alpha promote growth factor-induced proliferation of human vascular

smooth muscle cells. Am J Physiol Heart Circ Physiol. 2006;290:H2528-

2534

81. Schultz K, Murthy V, Tatro JB, Beasley D. Prolyl hydroxylase 2 deficiency

limits proliferation of vascular smooth muscle cells by hypoxia-inducible

factor-1{alpha}-dependent mechanisms. Am J Physiol Lung Cell Mol

Physiol. 2009;296:L921-927

82. BelAiba RS, Bonello S, Zähringer C, Schmidt S, Hess J, Kietzmann T,

Görlach A. Hypoxia up-regulates hypoxia-inducible factor-1α transcription

by involving phosphatidylinositol 3-kinase and nuclear factor κb in

pulmonary artery smooth muscle cells. Mol Biol Cell. 2007;18:4691-4697

83. Diebold I, Djordjevic T, Hess J, Görlach A. Rac-1 promotes pulmonary

artery smooth muscle cell proliferation by upregulation of plasminogen

activator inhibitor-1: Role of nfκb-dependent hypoxia-inducible factor-1α

transcription. Thromb Haemost. 2008;100:1021-1028

84. Shimoda L. Hypoxic regulation of ion channels and transporters in

pulmonary vascular smooth muscle. In: Yuan JXJ, Ward JPT, eds.

Membrane receptors, channels and transporters in pulmonary circulation.

Humana Press; 2010:221-235.

150

85. Zhang R, Wu Y, Zhao M, Liu C, Zhou L, Shen S, Liao S, Yang K, Li Q, Wan

H. Role of hif-1alpha in the regulation ace and ace2 expression in hypoxic

human pulmonary artery smooth muscle cells. Am J Physiol Lung Cell Mol

Physiol. 2009;297:L631-640

86. Hartsfield CL, Alam J, Choi AM. Differential signaling pathways of ho-1 gene

expression in pulmonary and systemic vascular cells. Am J Physiol.

1999;277:L1133-1141

87. Yang Z, Oemar BS, Carrel T, Kipfer B, Julmy F, Luscher TF. Different

proliferative properties of smooth muscle cells of human arterial and venous

bypass vessels : Role of pdgf receptors, mitogen-activated protein kinase,

and cyclin-dependent kinase inhibitors. Circulation. 1998;97:181-187

88. Weiss S, Frischknecht K, Greutert H, Payeli S, Steffel J, Luscher TF, Carrel

TP, Tanner FC. Different migration of vascular smooth muscle cells from

human coronary artery bypass vessels. Role of rho/rock pathway. J Vasc

Res. 2007;44:149-156

89. Chanakira A, Dutta R, Charboneau R, Barke R, Santilli SM, Roy S. Hypoxia

differentially regulates arterial and venous smooth muscle cell proliferation

via pdgfr-beta and vegfr-2 expression. Am J Physiol Heart Circ Physiol.

2012;302:H1173-1184

90. Osada-Oka M, Ikeda T, Akiba S, Sato T. Hypoxia stimulates the autocrine

regulation of migration of vascular smooth muscle cells via hif-1alpha-

dependent expression of thrombospondin-1. J Cell Biochem.

2008;104:1918-1926

151

91. Lambert CM, Roy M, Robitaille GA, Richard DE, Bonnet S. Hif-1 inhibition

decreases systemic vascular remodelling diseases by promoting apoptosis

through a hexokinase 2-dependent mechanism. Cardiovasc Res.

2010;88:196-204

92. Erdozain OJ, Pegrum S, Winrow VR, Horrocks M, Stevens CR. Hypoxia in

abdominal aortic aneurysm supports a role for hif-1α and ets-1 as drivers of

matrix metalloproteinase upregulation in human aortic smooth muscle cells.

J Vasc Res. 2011;48:163-170

93. Imanishi M, Tomita S, Ishizawa K, Kihira Y, Ueno M, Izawa-Ishizawa Y,

Ikeda Y, Yamano N, Tsuchiya K, Tamaki T. Smooth muscle cell specific hif-

1alpha deficiency suppresses angiotensin ii-induced vascular remodeling in

mice. Cardiovasc Res. 2014;102:460-468

94. Eckhart AD, Yang N, Xin X, Faber JE. Characterization of the α1b-

gene promoter region and hypoxia regulatory elements

in vascular smooth muscle. Proceedings of the National Academy of

Sciences. 1997;94:9487-9492

95. Osada-Oka M, Ikeda T, Imaoka S, Akiba S, Sato T. Vegf-enhanced

proliferation under hypoxia by an autocrine mechanism in human vascular

smooth muscle cells. Journal of Atherosclerosis and Thrombosis.

2008;15:26-33

96. Guo J, Chen H, Ho J, Mancini J, Sontag T, Laporte SA, Richard DE, Lebrun

J-J. Tgfβ-induced grk2 expression attenuates angii-regulated vascular

152

smooth muscle cell proliferation and migration. Cell Signal. 2009;21:899-

905

97. Huang Y, Di Lorenzo A, Jiang W, Cantalupo A, Sessa WC, Giordano FJ.

Hypoxia-inducible factor-1alpha in vascular smooth muscle regulates blood

pressure homeostasis through a peroxisome proliferator-activated receptor-

gamma-angiotensin ii receptor type 1 axis. Hypertension. 2013;62:634-640

98. Fowkes FG, Rudan D, Rudan I, Aboyans V, Denenberg JO, McDermott

MM, Norman PE, Sampson UK, Williams LJ, Mensah GA, Criqui MH.

Comparison of global estimates of prevalence and risk factors for peripheral

artery disease in 2000 and 2010: A systematic review and analysis. Lancet.

2013;382:1329-1340

99. Hirsch AT, Hiatt WR. Pad awareness, risk, and treatment: New resources

for survival--the USA partners program. Vasc Med. 2001;6:9-12

100. Srivaratharajah K, Abramson BL. Women and peripheral arterial disease: A

review of sex differences in epidemiology, clinical manifestations, and

outcomes. Can J Cardiol. 2018;34:356-361

101. Price JF, Mowbray PI, Lee AJ, Rumley A, Lowe GD, Fowkes FG.

Relationship between smoking and cardiovascular risk factors in the

development of peripheral arterial disease and coronary artery disease:

Edinburgh artery study. Eur Heart J. 1999;20:344-353

102. Fowkes FG, Housley E, Riemersma RA, Macintyre CC, Cawood EH,

Prescott RJ, Ruckley CV. Smoking, lipids, glucose intolerance, and blood

pressure as risk factors for peripheral atherosclerosis compared with

153

ischemic heart disease in the edinburgh artery study. Am J Epidemiol.

1992;135:331-340

103. Diehm C, Allenberg JR, Pittrow D, Mahn M, Tepohl G, Haberl RL, Darius H,

Burghaus I, Trampisch HJ. Mortality and vascular morbidity in older adults

with asymptomatic versus symptomatic peripheral artery disease.

Circulation. 2009;120:2053-2061

104. Hirsch AT, Haskal ZJ, Hertzer NR, Bakal CW, Creager MA, Halperin JL,

Hiratzka LF, Murphy WR, Olin JW, Puschett JB, Rosenfield KA, Sacks D,

Stanley JC, Taylor LM, Jr., White CJ, White J, White RA, Antman EM, Smith

SC, Jr., Adams CD, Anderson JL, Faxon DP, Fuster V, Gibbons RJ, Hunt

SA, Jacobs AK, Nishimura R, Ornato JP, Page RL, Riegel B. Acc/aha 2005

practice guidelines for the management of patients with peripheral arterial

disease (lower extremity, renal, mesenteric, and abdominal aortic): A

collaborative report from the american association for vascular

surgery/society for vascular surgery, society for cardiovascular angiography

and interventions, society for vascular medicine and biology, society of

interventional radiology, and the acc/aha task force on practice guidelines

(writing committee to develop guidelines for the management of patients

with peripheral arterial disease): Endorsed by the american association of

cardiovascular and pulmonary rehabilitation; national heart, lung, and blood

institute; society for vascular nursing; transatlantic inter-society consensus;

and vascular disease foundation. Circulation. 2006;113:e463-654

154

105. Rooke TW, Hirsch AT, Misra S, Sidawy AN, Beckman JA, Findeiss LK,

Golzarian J, Gornik HL, Halperin JL, Jaff MR, Moneta GL, Olin JW, Stanley

JC, White CJ, White JV, Zierler RE. 2011 accf/aha focused update of the

guideline for the management of patients with peripheral artery disease

(updating the 2005 guideline). A Report of the American College of

Cardiology Foundation/American Heart Association Task Force on Practice

Guidelines. 2011;124:2020-2045

106. Rooke TW, Hirsch AT, Misra S, Sidawy AN, Beckman JA, Findeiss L,

Golzarian J, Gornik HL, Jaff MR, Moneta GL, Olin JW, Stanley JC, White

CJ, White JV, Zierler RE. Management of patients with peripheral artery

disease (compilation of 2005 and 2011 accf/aha guideline

recommendations): A report of the american college of cardiology

foundation/american heart association task force on practice guidelines. J

Am Coll Cardiol. 2013;61:1555-1570

107. DeWeese JA, Leather R, Porter J. Practice guidelines: Lower extremity

revascularization. J Vasc Surg. 1993;18:280-294

108. McGee SR, Boyko EJ. Physical examination and chronic lower-extremity

ischemia: A critical review. Arch Intern Med. 1998;158:1357-1364

109. Fontaine R, Kim M, Kieny R. [surgical treatment of peripheral circulation

disorders]. Helv Chir Acta. 1954;21:499-533

110. Pascarelli EF, Bertrand CA. Comparison of blood pressures in the arms and

legs. N Engl J Med. 1964;270:693-698

155

111. Wang JC, Criqui MH, Denenberg JO, McDermott MM, Golomb BA, Fronek

A. Exertional leg pain in patients with and without peripheral arterial

disease. Circulation. 2005;112:3501-3508

112. Collins R, Cranny G, Burch J, Aguiar-Ibanez R, Craig D, Wright K, Berry E,

Gough M, Kleijnen J, Westwood M. A systematic review of duplex

ultrasound, magnetic resonance angiography and computed tomography

angiography for the diagnosis and assessment of symptomatic, lower limb

peripheral arterial disease. Health Technol Assess. 2007;11:iii-iv, xi-xiii, 1-

184

113. Mark AC, Joseph L. Arterial diseases of the extremities. In: Kasper D, Fauci

A, Hauser S, Longo D, Jameson JL, Loscalzo J, eds. Harrison's principles

of internal medicine, 19e. New York, NY: McGraw-Hill Education; 2015.

114. Faber JE, Chilian WM, Deindl E, van Royen N, Simons M. A brief etymology

of the collateral circulation. Arterioscler Thromb Vasc Biol. 2014;34:1854-

1859

115. Simons M, Eichmann A. Molecular controls of arterial morphogenesis. Circ

Res. 2015;116:1712-1724

116. Pipp F, Boehm S, Cai WJ, Adili F, Ziegler B, Karanovic G, Ritter R, Balzer

J, Scheler C, Schaper W, Schmitz-Rixen T. Elevated fluid shear stress

enhances postocclusive collateral artery growth and gene expression in the

pig hind limb. Arterioscler Thromb Vasc Biol. 2004;24:1664-1668

117. Hoefer IE, den Adel B, Daemen MJ. Biomechanical factors as triggers of

vascular growth. Cardiovasc Res. 2013;99:276-283

156

118. Chilian WM, Mass HJ, Williams SE, Layne SM, Smith EE, Scheel KW.

Microvascular occlusions promote coronary collateral growth. Am J Physiol.

1990;258:H1103-1111

119. van den Wijngaard JP, Schulten H, van Horssen P, Ter Wee RD, Siebes M,

Post MJ, Spaan JA. Porcine coronary collateral formation in the absence of

a pressure gradient remote of the ischemic border zone. Am J Physiol Heart

Circ Physiol. 2011;300:H1930-1937

120. Dai X, Faber JE. Endothelial nitric oxide synthase deficiency causes

collateral vessel rarefaction and impairs activation of a cell cycle gene

network during arteriogenesis. Circ Res. 2010;106:1870-1881

121. Deindl E, Hoefer IE, Fernandez B, Barancik M, Heil M, Strniskova M,

Schaper W. Involvement of the fibroblast growth factor system in adaptive

and chemokine-induced arteriogenesis. Circ Res. 2003;92:561-568

122. Morrison AR, Yarovinsky TO, Young BD, Moraes F, Ross TD, Ceneri N,

Zhang J, Zhuang ZW, Sinusas AJ, Pardi R, Schwartz MA, Simons M,

Bender JR. Chemokine-coupled beta2 integrin-induced macrophage rac2-

myosin iia interaction regulates vegf-a mrna stability and arteriogenesis. J

Exp Med. 2014;211:1957-1968

123. Takeda Y, Costa S, Delamarre E, Roncal C, Leite de Oliveira R, Squadrito

ML, Finisguerra V, Deschoemaeker S, Bruyere F, Wenes M, Hamm A,

Serneels J, Magat J, Bhattacharyya T, Anisimov A, Jordan BF, Alitalo K,

Maxwell P, Gallez B, Zhuang ZW, Saito Y, Simons M, De Palma M,

157

Mazzone M. Macrophage skewing by phd2 haplodeficiency prevents

ischaemia by inducing arteriogenesis. Nature. 2011;479:122-126

124. Carmeliet P. Angiogenesis in health and disease. Nat Med. 2003;9:653-660

125. Geudens I, Gerhardt H. Coordinating cell behaviour during blood vessel

formation. Development. 2011;138:4569-4583

126. Gerhardt H, Golding M, Fruttiger M, Ruhrberg C, Lundkvist A, Abramsson

A, Jeltsch M, Mitchell C, Alitalo K, Shima D, Betsholtz C. Vegf guides

angiogenic sprouting utilizing endothelial tip cell filopodia. The Journal of

Cell Biology. 2003;161:1163-1177

127. Benjamin LE, Golijanin D, Itin A, Pode D, Keshet E. Selective ablation of

immature blood vessels in established human tumors follows vascular

endothelial growth factor withdrawal. J Clin Invest. 1999;103:159-165

128. Liu Y, Cox SR, Morita T, Kourembanas S. Hypoxia regulates vascular

endothelial growth factor gene expression in endothelial cells. Identification

of a 5' enhancer. Circ Res. 1995;77:638-643

129. Pugh CW, Ratcliffe PJ. Regulation of angiogenesis by hypoxia: Role of the

hif system. Nat Med. 2003;9:677-684

130. Forsythe JA, Jiang BH, Iyer NV, Agani F, Leung SW, Koos RD, Semenza

GL. Activation of vascular endothelial growth factor gene transcription by

hypoxia-inducible factor 1. Mol Cell Biol. 1996;16:4604-4613

131. Semenza GL. Vasculogenesis, angiogenesis, and arteriogenesis:

Mechanisms of blood vessel formation and remodeling. J Cell Biochem.

2007;102:840-847

158

132. Thurston G, Suri C, Smith K, McClain J, Sato TN, Yancopoulos GD,

McDonald DM. Leakage-resistant blood vessels in mice transgenically

overexpressing angiopoietin-1. Science. 1999;286:2511-2514

133. Murphy TP, Cutlip DE, Regensteiner JG, Mohler ER, 3rd, Cohen DJ,

Reynolds MR, Massaro JM, Lewis BA, Cerezo J, Oldenburg NC, Thum CC,

Jaff MR, Comerota AJ, Steffes MW, Abrahamsen IH, Goldberg S, Hirsch

AT. Supervised exercise, stent revascularization, or medical therapy for

claudication due to aortoiliac peripheral artery disease: The clever study. J

Am Coll Cardiol. 2015;65:999-1009

134. Murphy TP, Cutlip DE, Regensteiner JG, Mohler ER, Cohen DJ, Reynolds

MR, Massaro JM, Lewis BA, Cerezo J, Oldenburg NC, Thum CC, Goldberg

S, Jaff MR, Steffes MW, Comerota AJ, Ehrman J, Treat-Jacobson D, Walsh

ME, Collins T, Badenhop DT, Bronas U, Hirsch AT. Supervised exercise

versus primary stenting for claudication resulting from aortoiliac peripheral

artery disease: Six-month outcomes from the claudication: Exercise versus

endoluminal revascularization (clever) study. Circulation. 2012;125:130-

139

135. Norgren L, Hiatt WR, Dormandy JA, Nehler MR, Harris KA, Fowkes FG.

Inter-society consensus for the management of peripheral arterial disease

(tasc ii). J Vasc Surg. 2007;45 Suppl S:S5-67

136. Pan CW, Patel PM. Peripheral arterial disease. In: Ferri FF, ed. Ferri's

clinical advisor 2018. Elsevier; 2018:975-977.

159

137. Hiramoto JS, Teraa M, de Borst GJ, Conte MS. Interventions for lower

extremity peripheral artery disease. Nat Rev Cardiol. 2018;15:332-350

138. Castro C, Diez-Juan A, Cortes MJ, Andres V. Distinct regulation of mitogen-

activated protein kinases and p27kip1 in smooth muscle cells from different

vascular beds. A potential role in establishing regional phenotypic variance.

J Biol Chem. 2003;278:4482-4490

139. Limbourg A, Korff T, Napp LC, Schaper W, Drexler H, Limbourg FP.

Evaluation of postnatal arteriogenesis and angiogenesis in a mouse model

of hind-limb ischemia. Nat Protocols. 2009;4:1737-1748

140. Padgett ME, McCord TJ, McClung JM, Kontos CD. Methods for acute and

subacute murine hindlimb ischemia. Journal of Visualized Experiments :

JoVE. 2016:54166

141. Westvik TS, Fitzgerald TN, Muto A, Maloney SP, Pimiento JM, Fancher TT,

Magri D, Westvik HH, Nishibe T, Velazquez OC, Dardik A. Limb ischemia

after iliac ligation in aged mice stimulates angiogenesis without

arteriogenesis. J Vasc Surg. 2009;49:464-473

142. Bosch-Marce M, Okuyama H, Wesley JB, Sarkar K, Kimura H, Liu YV,

Zhang H, Strazza M, Rey S, Savino L, Zhou YF, McDonald KR, Na Y,

Vandiver S, Rabi A, Shaked Y, Kerbel R, LaVallee T, Semenza GL. Effects

of aging and hypoxia-inducible factor-1 activity on angiogenic cell

mobilization and recovery of perfusion after limb ischemia. Circ Res.

2007;101:1310-1318

160

143. Li M, Liu C, Bin J, Wang Y, Chen J, Xiu J, Pei J, Lai Y, Chen D, Fan C, Xie

J, Tao Y, Wu P. Mutant hypoxia inducible factor-1alpha improves

angiogenesis and tissue perfusion in ischemic rabbit skeletal muscle.

Microvasc Res. 2011;81:26-33

144. Patel TH, Kimura H, Weiss CR, Semenza GL, Hofmann LV. Constitutively

active hif-1alpha improves perfusion and arterial remodeling in an

endovascular model of limb ischemia. Cardiovasc Res. 2005;68:144-154

145. Milkiewicz M, Pugh CW, Egginton S. Inhibition of endogenous hif

inactivation induces angiogenesis in ischaemic skeletal muscles of mice. J

Physiol. 2004;560:21-26

146. Skuli N, Majmundar AJ, Krock BL, Mesquita RC, Mathew LK, Quinn ZL,

Runge A, Liu L, Kim MN, Liang J, Schenkel S, Yodh AG, Keith B, Simon

MC. Endothelial hif-2alpha regulates murine pathological angiogenesis and

revascularization processes. J Clin Invest. 2012;122:1427-1443

147. MacLauchlan SC, Calabro NE, Huang Y, Krishna M, Bancroft T, Sharma T,

Yu J, Sessa WC, Giordano F, Kyriakides TR. Hif-1alpha represses the

expression of the angiogenesis inhibitor thrombospondin-2. Matrix Biol.

2018;65:45-58

148. Iyer SR, Annex BH. Therapeutic angiogenesis for peripheral artery disease:

Lessons learned in translational science. JACC Basic to translational

science. 2017;2:503-512

149. Creager MA, Olin JW, Belch JJ, Moneta GL, Henry TD, Rajagopalan S,

Annex BH, Hiatt WR. Effect of hypoxia-inducible factor-1alpha gene therapy

161

on walking performance in patients with intermittent claudication.

Circulation. 2011;124:1765-1773

150. Rajagopalan S, Olin J, Deitcher S, Pieczek A, Laird J, Grossman PM,

Goldman CK, McEllin K, Kelly R, Chronos N. Use of a constitutively active

hypoxia-inducible factor-1alpha transgene as a therapeutic strategy in no-

option critical limb ischemia patients: Phase i dose-escalation experience.

Circulation. 2007;115:1234-1243

151. Carmeliet P. Mechanisms of angiogenesis and arteriogenesis. Nat Med.

2000;6:389-395

152. Fischer C, Schneider M, Carmeliet P. Principles and therapeutic

implications of angiogenesis, vasculogenesis and arteriogenesis. In:

Moncada S, Higgs A, eds. The vascular endothelium ii. Springer Berlin

Heidelberg; 2006:157-212.

153. Cooke JP, Losordo DW. Modulating the vascular response to limb ischemia.

Circ Res. 2015;116:1561-1578

154. Schwartz SM, Campbell GR, Campbell JH. Replication of smooth muscle

cells in vascular disease. Circ Res. 1986;58:427-444

155. Welsh DJ, Peacock AJ. Cellular responses to hypoxia in the pulmonary

circulation. High Alt Med Biol. 2013;14:111-116

156. Peng J, Zhang L, Drysdale L, Fong GH. The transcription factor epas-

1/hypoxia-inducible factor 2alpha plays an important role in vascular

remodeling. Proc Natl Acad Sci U S A. 2000;97:8386-8391

162

157. Tian H, Hammer RE, Matsumoto AM, Russell DW, McKnight SL. The

hypoxia-responsive transcription factor epas1 is essential for

catecholamine homeostasis and protection against heart failure during

embryonic development. Genes Dev. 1998;12:3320-3324

158. Compernolle V, Brusselmans K, Acker T, Hoet P, Tjwa M, Beck H,

Plaisance S, Dor Y, Keshet E, Lupu F, Nemery B, Dewerchin M, Van

Veldhoven P, Plate K, Moons L, Collen D, Carmeliet P. Loss of hif-2alpha

and inhibition of vegf impair fetal lung maturation, whereas treatment with

vegf prevents fatal respiratory distress in premature mice. Nat Med.

2002;8:702-710

159. Ramirez-Bergeron DL, Simon MC. Hypoxia-inducible factor and the

development of stem cells of the cardiovascular system. Stem Cells.

2001;19:279-286

160. Jain S, Maltepe E, Lu MM, Simon C, Bradfield CA. Expression of arnt, arnt2,

hif1α, hif2α and ah receptor mrnas in the developing mouse. Mech Dev.

1998;73:117-123

161. Pawlus MR, Hu CJ. Enhanceosomes as integrators of hypoxia inducible

factor (hif) and other transcription factors in the hypoxic transcriptional

response. Cell Signal. 2013;25:1895-1903

162. Ho TK, Rajkumar V, Ponticos M, Leoni P, Black DCM, Abraham DJ, Baker

DM. Increased endogenous angiogenic response and hypoxia-inducible

factor-1a; in human critical limb ischemia. J Vasc Surg. 2006;43:125-133

163

163. Lepore JJ, Cheng L, Min Lu M, Mericko PA, Morrisey EE, Parmacek MS.

High-efficiency somatic mutagenesis in smooth muscle cells and cardiac

myocytes in sm22alpha-cre transgenic mice. Genesis. 2005;41:179-184

164. Proweller A, Wright AC, Horng D, Cheng L, Lu MM, Lepore JJ, Pear WS,

Parmacek MS. Notch signaling in vascular smooth muscle cells is required

to pattern the cerebral vasculature. Proc Natl Acad Sci U S A.

2007;104:16275-16280

165. Lu Y, Zhang L, Liao X, Sangwung P, Prosdocimo DA, Zhou G, Votruba AR,

Brian L, Han YJ, Gao H, Wang Y, Shimizu K, Weinert-Stein K, Khrestian M,

Simon DI, Freedman NJ, Jain MK. Kruppel-like factor 15 is critical for

vascular . J Clin Invest. 2013;123:4232-4241

166. Tomita S, Sinal CJ, Yim SH, Gonzalez FJ. Conditional disruption of the aryl

hydrocarbon receptor nuclear translocator (arnt) gene leads to loss of target

gene induction by the aryl hydrocarbon receptor and hypoxia-inducible

factor 1alpha. Mol Endocrinol. 2000;14:1674-1681

167. Han Y, Yang K, Proweller A, Zhou G, Jain MK, Ramirez-Bergeron DL.

Inhibition of arnt severely compromises endothelial cell viability and function

in response to moderate hypoxia. Angiogenesis. 2012;15:409-420

168. Peng X, Wang J, Lassance-Soares RM, Najafi AH, Sood S, Aghili N,

Alderman LO, Panza JA, Faber JE, Wang S, Epstein SE, Burnett MS.

Gender differences affect blood flow recovery in a mouse model of hindlimb

ischemia. Am J Physiol Heart Circ Physiol. 2011;300:H2027-H2034

164

169. Bailey AM, O’Neill TJ, Morris CE, Peirce SM. Arteriolar remodeling following

ischemic injury extends from capillary to large arteriole in the

microcirculation. Microcirculation. 2008;15:389-404

170. Muller PY, Janovjak H, Miserez AR, Dobbie Z. Processing of gene

expression data generated by quantitative real-time rt-pcr. Biotechniques.

2002;32:1372-1374, 1376, 1378-1379

171. Wenger RH. Cellular adaptation to hypoxia: O2-sensing protein

hydroxylases, hypoxia-inducible transcription factors, and o2-regulated

gene expression. FASEB J. 2002;16:1151-1162

172. Salceda S, Beck I, Caro J. Absolute requirement of aryl hydrocarbon

receptor nuclear translocator protein for gene activation by hypoxia. Arch

Biochem Biophys. 1996;334:389-394

173. Gao S, Ho D, Vatner DE, Vatner SF. Echocardiography in mice. Curr Protoc

Mouse Biol. 2011;1:71–83

174. Schaad L, Hlushchuk R, Barré S, Gianni-Barrera R, Haberthür D, Banfi A,

Djonov V. Correlative imaging of the murine hind limb vasculature and

muscle tissue by microct and light microscopy. Sci Rep. 2017;7:41842

175. Chou SC, Azuma Y, Varia MA, Raleigh JA. Evidence that involucrin, a

marker for differentiation, is oxygen regulated in human squamous cell

carcinomas. Br J Cancer. 2004;90:728-735

176. Gross MW, Karbach U, Groebe K, Franko AJ, Mueller-Klieser W.

Calibration of misonidazole labeling by simultaneous measurement of

165

oxygen tension and labeling density in multicellular spheroids. Int J Cancer.

1995;61:567-573

177. Arpino JM, Nong Z, Li F, Yin H, Ghonaim N, Milkovich S, Balint B, O'Neil C,

Fraser GM, Goldman D, Ellis CG, Pickering JG. Four-dimensional

microvascular analysis reveals that regenerative angiogenesis in ischemic

muscle produces a flawed microcirculation. Circ Res. 2017;120:1453-1465

178. Dormandy JA, Rutherford RB. Management of peripheral arterial disease.

Tasc working group. Transatlantic inter-society consensus. J Vasc Surg.

2000;31:S1-s296

179. Browning E, Wang H, Hong N, Yu K, Buerk DG, DeBolt K, Gonder D,

Sorokina EM, Patel P, De Leon DD, Feinstein SI, Fisher AB, Chatterjee S.

Mechanotransduction drives post ischemic revascularization through k(atp)

channel closure and production of reactive oxygen species. Antioxid Redox

Signal. 2014;20:872-886

180. Chanakira A, Kir D, Barke RA, Santilli SM, Ramakrishnan S, Roy S. Hypoxia

differentially regulates arterial and venous smooth muscle cell migration.

PLoS One. 2015;10:e0138587

181. Deng B, Du J, Hu R, Wang AP, Wu WH, Hu CP, Li YJ, Li XH. Microrna-

103/107 is involved in hypoxia-induced proliferation of pulmonary arterial

smooth muscle cells by targeting hif-1beta. Life Sci. 2016;147:117-124

182. Kelly BD, Hackett SF, Hirota K, Oshima Y, Cai Z, Berg-Dixon S, Rowan A,

Yan Z, Campochiaro PA, Semenza GL. Cell type-specific regulation of

angiogenic growth factor gene expression and induction of angiogenesis in

166

nonischemic tissue by a constitutively active form of hypoxia-inducible

factor 1. Circ Res. 2003;93:1074-1081

183. Ramirez-Bergeron DL, Runge A, Adelman DM, Gohil M, Simon MC. Hif-

dependent hematopoietic factors regulate the development of the

embryonic vasculature. Dev Cell. 2006;11:81-92

184. Ramirez-Bergeron DL, Runge A, Dahl KD, Fehling HJ, Keller G, Simon MC.

Hypoxia affects mesoderm and enhances hemangioblast specification

during early development. Development. 2004;131:4623-4634

185. Maxwell PH, Dachs GU, Gleadle JM, Nicholls LG, Harris AL, Stratford IJ,

Hankinson O, Pugh CW, Ratcliffe PJ. Hypoxia-inducible factor-1 modulates

gene expression in solid tumors and influences both angiogenesis and

tumor growth. Proc Natl Acad Sci U S A. 1997;94:8104-8109

186. Semenza GL. A compendium of proteins that interact with hif-1α. Exp Cell

Res. 2017;356:128-135

187. Semenza GL. Vascular responses to hypoxia and ischemia. Arterioscler

Thromb Vasc Biol. 2010;30:648-652

188. Lijkwan MA, Hellingman AA, Bos EJ, van der Bogt KEA, Huang M,

Kooreman NG, de Vries MR, Peters HAB, Robbins RC, Hamming JF, Quax

PHA, Wu JC. Short hairpin rna gene silencing of prolyl hydroxylase-2 with

a minicircle vector improves neovascularization of hindlimb ischemia. Hum

Gene Ther. 2014;25:41-49

167

189. Moraes F, Paye J, Mac Gabhann F, Zhuang ZW, Zhang J, Lanahan AA,

Simons M. Endothelial cell-dependent regulation of arteriogenesis. Circ

Res. 2013;113:1076-1086

190. Shepherd AP, Öberg PÅ. Laser-doppler blood flowmetry. Boston: Kluwer

Academic Publishers; 1990.

191. Yu AY, Shimoda LA, Iyer NV, Huso DL, Sun X, McWilliams R, Beaty T,

Sham JSK, Wiener CM, Sylvester JT, Semenza GL. Impaired physiological

responses to chronic hypoxia in mice partially deficient for hypoxia-inducible

factor 1α. J Clin Invest. 1999;103:691-696

192. Brusselmans K, Compernolle V, Tjwa M, Wiesener MS, Maxwell PH, Collen

D, Carmeliet P. Heterozygous deficiency of hypoxia-inducible factor–2α

protects mice against pulmonary hypertension and right ventricular

dysfunction during prolonged hypoxia. J Clin Invest. 2003;111:1519-1527

193. Schermuly RT, Dony E, Ghofrani HA, Pullamsetti S, Savai R, Roth M,

Sydykov A, Lai YJ, Weissmann N, Seeger W, Grimminger F. Reversal of

experimental pulmonary hypertension by pdgf inhibition. J Clin Invest.

2005;115:2811-2821

194. Millette E, Rauch BH, Kenagy RD, Daum G, Clowes AW. Platelet-derived

growth factor–bb transactivates the fibroblast growth factor receptor to

induce proliferation in human smooth muscle cells. Trends Cardiovasc.

2006;16:25-28

195. Imanishi M, Chiba Y, Tomita N, Matsunaga S, Nakagawa T, Ueno M,

Yamamoto K, Tamaki T, Tomita S. Hypoxia-inducible factor-1α in smooth

168

muscle cells protects against aortic aneurysms—brief report. Arterioscler

Thromb Vasc Biol. 2016;36:2158-2162

196. Yoshida S, Nabzdyk CS, Pradhan L, LoGerfo FW. Thrombospondin-2 gene

silencing in human aortic smooth muscle cells improves cell attachment. J

Am Coll Surg. 2011;213:668-676

197. Grotendorst GR, Seppä HE, Kleinman HK, Martin GR. Attachment of

smooth muscle cells to collagen and their migration toward platelet-derived

growth factor. Proc Natl Acad Sci U S A. 1981;78:3669-3672

198. Barnes EA, Chen C-H, Sedan O, Cornfield DN. Loss of smooth muscle cell

hypoxia inducible factor-1α underlies increased vascular contractility in

pulmonary hypertension. FASEB J. 2017;31:650-662

199. Bateman RM, Sharpe MD, Ellis CG. Bench-to-bedside review:

Microvascular dysfunction in sepsis –hemodynamics, oxygen transport, and

nitric oxide. Critical Care. 2003;7:359-373

200. Johnson RJ, Rodriguez-Iturbe B, Kang DH, Feig DI, Herrera-Acosta J. A

unifying pathway for essential hypertension. Am J Hypertens. 2005;18:431-

440

201. Lyle MA, Davis JP, Brozovich FV. Regulation of pulmonary vascular smooth

muscle contractility in pulmonary arterial hypertension: Implications for

therapy. Front Physiol. 2017;8:614

202. Friedman SM, Nakashima M, Mar MA. Morphological assessment of

vasoconstriction and vascular hypertrophy in sustained hypertension in the

rat. Microvasc Res. 1971;3:416-425

169

203. Borton AH, Benson BL, Neilson LE, Saunders A, Alaiti MA, Huang AY, Jain

MK, Proweller A, Ramirez-Bergeron DL. Aryl hydrocarbon receptor nuclear

translocator in vascular smooth muscle cells is required for optimal

peripheral perfusion recovery. J Am Heart Assoc. 2018;7

204. Basu S, Srinivasan DK, Yang K, Raina H, Banerjee S, Zhang R, Fisher SA,

Proweller A. Notch transcriptional control of vascular smooth muscle

regulatory gene expression and function. J Biol Chem. 2013;288:11191-

11202

205. Kuang SQ, Kwartler CS, Byanova KL, Pham J, Gong L, Prakash SK, Huang

J, Kamm KE, Stull JT, Sweeney HL, Milewicz DM. Rare, nonsynonymous

variant in the smooth muscle-specific isoform of myosin heavy chain,

myh11, r247c, alters force generation in the aorta and phenotype of smooth

muscle cells. Circ Res. 2012;110:1411-1422

206. Zacour ME, Teoh H, Halayko AJ, Ward ME. Mechanisms of aortic smooth

muscle hyporeactivity after prolonged hypoxia in rats. J Appl Physiol.

2002;92:2625-2632

207. Labrousse-Arias D, Castillo-González R, Rogers NM, Torres-Capelli M,

Barreira B, Aragonés J, Cogolludo Á, Isenberg JS, Calzada MJ. Hif-2α-

mediated induction of pulmonary thrombospondin-1 contributes to hypoxia-

driven vascular remodelling and vasoconstriction. Cardiovasc Res.

2016;109:115-130

170

208. Filiano AJ, Bailey CD, Tucholski J, Gundemir S, Johnson GV.

Transglutaminase 2 protects against ischemic insult, interacts with hif1beta,

and attenuates hif1 signaling. FASEB J. 2008;22:2662-2675

209. McIntosh BE, Hogenesch JB, Bradfield CA. Mammalian per-arnt-sim

proteins in environmental adaptation. Annu Rev Physiol. 2010;72:625-645

210. Meeks MK, Ripley ML, Jin Z, Rembold CM. Heat shock protein 20-mediated

force suppression in forskolin-relaxed swine carotid artery. American

Journal of Physiology-Cell Physiology. 2005;288:C633-C639

211. Ye GJ, Aratyn-Schaus Y, Nesmith AP, Pasqualini FS, Alford PW, Parker

KK. The contractile strength of vascular smooth muscle myocytes is shape

dependent. Integr Biol (Camb). 2014;6:152-163

212. Alford PW, Nesmith AP, Seywerd JN, Grosberg A, Parker KK. Vascular

smooth muscle contractility depends on cell shape. Integr Biol (Camb).

2011;3:1063-1070

213. Riento K, Ridley AJ. Rocks: Multifunctional kinases in cell behaviour. Nat

Rev Mol Cell Biol. 2003;4:446-456

214. Loirand G, Guerin P, Pacaud P. Rho kinases in cardiovascular physiology

and pathophysiology. Circ Res. 2006;98:322-334

215. Tao J, Barnett JV, Watanabe M, Ramirez-Bergeron D. Hypoxia supports

epicardial cell differentiation in vascular smooth muscle cells through the

activation of the tgfbeta pathway. J Cardiovasc Dev Dis. 2018;5

171

216. Tang DD, Gunst SJ. Depletion of focal adhesion kinase by antisense

depresses contractile activation of smooth muscle. Am J Physiol Cell

Physiol. 2001;280:C874-883

217. Opazo Saez A, Zhang W, Wu Y, Turner CE, Tang DD, Gunst SJ. Tension

development during contractile stimulation of smooth muscle requires

recruitment of paxillin and vinculin to the membrane. Am J Physiol Cell

Physiol. 2004;286:C433-447

218. Romer LH, Birukov KG, Garcia JG. Focal adhesions: Paradigm for a

signaling nexus. Circ Res. 2006;98:606-616

219. Lee SH, Lee YJ, Han HJ. Role of hypoxia-induced fibronectin-integrin beta1

expression in embryonic stem cell proliferation and migration: Involvement

of pi3k/akt and fak. J Cell Physiol. 2011;226:484-493

220. Cowden Dahl KD, Robertson SE, Weaver VM, Simon MC. Hypoxia-

inducible factor regulates alphavbeta3 integrin cell surface expression. Mol

Biol Cell. 2005;16:1901-1912

221. Corley KM, Taylor CJ, Lilly B. Hypoxia-inducible factor 1alpha modulates

adhesion, migration, and fak phosphorylation in vascular smooth muscle

cells. J Cell Biochem. 2005;96:971-985

222. Steucke KE, Tracy PV, Hald ES, Hall JL, Alford PW. Vascular smooth

muscle cell functional contractility depends on extracellular mechanical

properties. J Biomech. 2015;48:3044-3051

223. Lehoux S, Castier Y, Tedgui A. Molecular mechanisms of the vascular

responses to haemodynamic forces. J Intern Med. 2006;259:381-392

172

224. Chen Q, Jin M, Yang F, Zhu J, Xiao Q, Zhang L. Matrix metalloproteinases:

Inflammatory regulators of cell behaviors in vascular formation and

remodeling. Mediators Inflamm. 2013;2013:928315

225. Stegemann JP, Hong H, Nerem RM. Mechanical, biochemical, and

extracellular matrix effects on vascular smooth muscle cell phenotype. J

Appl Physiol. 2005;98:2321-2327

226. Ahmad A, Ahmad S, Malcolm KC, Miller SM, Hendry-Hofer T, Schaack JB,

White CW. Differential regulation of pulmonary vascular cell growth by

hypoxia-inducible transcription factor–1α and hypoxia-inducible

transcription factor–2α. Am J Respir Cell Mol Biol. 2013;49:78-85

227. Israël A. The ikk complex, a central regulator of nf-κb activation. Cold Spring

Harb Perspect Biol. 2010;2:a000158

228. Vogel TP, Milner JD, Cooper MA. The ying and yang of stat3 in human

disease. J Clin Immunol. 2015;35:615-623

229. Adams JM, Cory S. The bcl-2 protein family: Arbiters of cell survival.

Science. 1998;281:1322-1326

230. Fouillade C, Monet-Lepretre M, Baron-Menguy C, Joutel A. Notch signalling

in smooth muscle cells during development and disease. Cardiovasc Res.

2012;95:138-146

231. Hoffman EC, Reyes H, Chu FF, Sander F, Conley LH, Brooks BA,

Hankinson O. Cloning of a factor required for activity of the ah (dioxin)

receptor. Science. 1991;252:954-958

173

232. Reyes H, Reisz-Porszasz S, Hankinson O. Identification of the ah receptor

nuclear translocator protein (arnt) as a component of the DNA binding form

of the ah receptor. Science. 1992;256:1193-1195

233. Xiao L, Zhang Z, Luo X. Roles of xenobiotic receptors in vascular

pathophysiology. Circ J. 2014;78:1520-1530

234. Seidel SD, Winters GM, Rogers WJ, Ziccardi MH, Li V, Keser B, Denison

MS. Activation of the ah receptor signaling pathway by prostaglandins. J

Biochem Mol Toxicol. 2001;15:187-196

235. Sinal CJ, Bend JR. Aryl hydrocarbon receptor-dependent induction of

cyp1a1 by bilirubin in mouse hepatoma hepa 1c1c7 cells. Mol Pharmacol.

1997;52:590-599

236. McMillan BJ, Bradfield CA. The aryl hydrocarbon receptor is activated by

modified low-density lipoprotein. Proc Natl Acad Sci U S A. 2007;104:1412-

1417

237. Gasiewicz TA, Singh KP, Bennett JA. The ah receptor in stem cell cycling,

regulation, and quiescence. Ann N Y Acad Sci. 2014;1310:44-50

238. Singh KP, Garrett RW, Casado FL, Gasiewicz TA. Aryl hydrocarbon

receptor-null allele mice have hematopoietic stem/progenitor cells with

abnormal characteristics and functions. Stem Cells Dev. 2011;20:769-784

239. Singh KP, Bennett JA, Casado FL, Walrath JL, Welle SL, Gasiewicz TA.

Loss of aryl hydrocarbon receptor promotes gene changes associated with

premature hematopoietic stem cell exhaustion and development of a

myeloproliferative disorder in aging mice. Stem Cells Dev. 2014;23:95-106

174

240. Stockinger B, Di Meglio P, Gialitakis M, Duarte JH. The aryl hydrocarbon

receptor: Multitasking in the immune system. Annu Rev Immunol.

2014;32:403-432

241. Zhang N. The role of endogenous aryl hydrocarbon receptor signaling in

cardiovascular physiology. J Cardiovasc Dis Res. 2011;2:91-95

242. Adelibieke Y, Yisireyili M, Ng HY, Saito S, Nishijima F, Niwa T. Indoxyl

sulfate induces il-6 expression in vascular endothelial and smooth muscle

cells through oat3-mediated uptake and activation of ahr/nf-kappab

pathway. Nephron Exp Nephrol. 2014;128:1-8

243. Ng HY, Bolati W, Lee CT, Chien YS, Yisireyili M, Saito S, Pei SN, Nishijima

F, Niwa T. Indoxyl sulfate downregulates mas receptor via aryl hydrocarbon

receptor/nuclear factor-kappa b, and induces cell proliferation and tissue

factor expression in vascular smooth muscle cells. Nephron. 2016;133:205-

212

244. Yisireyili M, Saito S, Abudureyimu S, Adelibieke Y, Ng HY, Nishijima F,

Takeshita K, Murohara T, Niwa T. Indoxyl sulfate-induced activation of

(pro)renin receptor promotes cell proliferation and tissue factor expression

in vascular smooth muscle cells. PLoS One. 2014;9:e109268

245. Moore DH, Ruska H. The fine structure of capillaries and small arteries. J

Biophys Biochem Cytol. 1957;3:457-462

246. Straub AC, Zeigler AC, Isakson BE. The myoendothelial junction:

Connections that deliver the message. Physiology. 2014;29:242-249

175

247. Isakson BE, Duling BR. Heterocellular contact at the myoendothelial

junction influences gap junction organization. Circ Res. 2005;97:44-51

248. Straub AC, Billaud M, Johnstone SR, Best AK, Yemen S, Dwyer ST, Looft-

Wilson R, Lysiak JJ, Gaston B, Palmer L, Isakson BE. Compartmentalized

connexin 43 s-nitrosylation/denitrosylation regulates heterocellular

communication in the vessel wall. Arterioscler Thromb Vasc Biol.

2011;31:399-407

249. Rhodin JA. The ultrastructure of mammalian arterioles and precapillary

sphincters. J Ultrastruct Res. 1967;18:181-223

250. Sandow SL, Hill CE. Incidence of myoendothelial gap junctions in the

proximal and distal mesenteric arteries of the rat is suggestive of a role in

endothelium-derived hyperpolarizing factor-mediated responses. Circ Res.

2000;86:341-346

251. Heberlein KR, Straub AC, Best AK, Greyson MA, Looft-Wilson RC, Sharma

PR, Meher A, Leitinger N, Isakson BE. Plasminogen activator inhibitor-1

regulates myoendothelial junction formation. Circ Res. 2010;106:1092-

1102

252. Baeten JT, Lilly B. Notch signaling in vascular smooth muscle cells. Adv

Pharmacol. 2017;78:351-382

253. Yang K, Proweller A. Vascular smooth muscle notch signals regulate

endothelial cell sensitivity to angiogenic stimulation. J Biol Chem.

2011;286:13741-13753

176

254. Yang K, Banerjee S, Proweller A. Regulation of pre-natal circle of willis

assembly by vascular smooth muscle notch signaling. Dev Biol.

2013;381:107-120

255. Sahlgren C, Gustafsson MV, Jin S, Poellinger L, Lendahl U. Notch signaling

mediates hypoxia-induced tumor cell migration and invasion. Proc Natl

Acad Sci U S A. 2008;105:6392-6397

256. Diez H, Fischer A, Winkler A, Hu CJ, Hatzopoulos AK, Breier G, Gessler M.

Hypoxia-mediated activation of dll4-notch-hey2 signaling in endothelial

progenitor cells and adoption of arterial cell fate. Exp Cell Res. 2007;313:1-

9

257. Hu YY, Fu LA, Li SZ, Chen Y, Li JC, Han J, Liang L, Li L, Ji CC, Zheng MH,

Han H. Hif-1alpha and hif-2alpha differentially regulate notch signaling

through competitive interaction with the intracellular domain of notch

receptors in glioma stem cells. Cancer Lett. 2014;349:67-76

258. Zheng X, Narayanan S, Zheng X, Luecke-Johansson S, Gradin K, Catrina

SB, Poellinger L, Pereira TS. A notch-independent mechanism contributes

to the induction of hes1 gene expression in response to hypoxia in p19 cells.

Exp Cell Res. 2017;358:129-139

259. Zheng X, Linke S, Dias JM, Zheng X, Gradin K, Wallis TP, Hamilton BR,

Gustafsson M, Ruas JL, Wilkins S, Bilton RL, Brismar K, Whitelaw ML,

Pereira T, Gorman JJ, Ericson J, Peet DJ, Lendahl U, Poellinger L.

Interaction with factor inhibiting hif-1 defines an additional mode of cross-

177

coupling between the notch and hypoxia signaling pathways. Proc Natl

Acad Sci U S A. 2008;105:3368-3373

260. Armulik A, Genové G, Mäe M, Nisancioglu MH, Wallgard E, Niaudet C, He

L, Norlin J, Lindblom P, Strittmatter K, Johansson BR, Betsholtz C.

Pericytes regulate the blood–brain barrier. Nature. 2010;468:557

261. Moura RP, Almeida A, Sarmento B. The role of non-endothelial cells on the

penetration of nanoparticles through the blood brain barrier. Prog Neurobiol.

2017;159:39-49

262. Volz KS, Jacobs AH, Chen HI, Poduri A, McKay AS, Riordan DP, Kofler N,

Kitajewski J, Weissman I, Red-Horse K. Pericytes are progenitors for

coronary artery smooth muscle. Elife. 2015;4

263. Ding R, Darland DC, Parmacek MS, D'Amore PA. Endothelial-

mesenchymal interactions in vitro reveal molecular mechanisms of smooth

muscle/pericyte differentiation. Stem Cells Dev. 2004;13:509-520

264. Shen Z, Li C, Frieler RA, Gerasimova AS, Lee SJ, Wu J, Wang MM, Lumeng

CN, Brosius FC, Duan SZ, Mortensen RM. Smooth muscle protein 22

alpha-cre is expressed in myeloid cells in mice. Biochem Biophys Res

Commun. 2012;422:639-642

265. Voelkel NF, Tuder RM. Hypoxia-induced pulmonary vascular remodeling: A

model for what human disease? J Clin Invest. 2000;106:733-738

266. Liu W, Zhang Y, Lu L, Wang L, Chen M, Hu T. Expression and correlation

of hypoxia-inducible factor-1α (hif-1α) with pulmonary artery remodeling

and right ventricular hypertrophy in experimental pulmonary embolism.

178

Medical Science Monitor : International Medical Journal of Experimental

and Clinical Research. 2017;23:2083-2088

267. Ball MK, Waypa GB, Mungai PT, Nielsen JM, Czech L, Dudley VJ, Beussink

L, Dettman RW, Berkelhamer SK, Steinhorn RH, Shah SJ, Schumacker PT.

Regulation of hypoxia-induced pulmonary hypertension by vascular smooth

muscle hypoxia-inducible factor-1α. Am J Respir Crit Care Med.

2014;189:314-324

268. Reho JJ, Zheng X, Fisher SA. Smooth muscle contractile diversity in the

control of regional circulations. American Journal of Physiology - Heart and

Circulatory Physiology. 2014;306:H163-H172

269. Sausbier M, Schubert R, Voigt V, Hirneiss C, Pfeifer A, Korth M, Kleppisch

T, Ruth P, Hofmann F. Mechanisms of no/cgmp-dependent vasorelaxation.

Circ Res. 2000;87:825-830

270. Starke RM, Chalouhi N, Ding D, Raper DM, McKisic MS, Owens GK, Hasan

DM, Medel R, Dumont AS. Vascular smooth muscle cells in cerebral

aneurysm pathogenesis. Transl Stroke Res. 2014;5:338-346

271. Milewicz DM, Trybus KM, Guo DC, Sweeney HL, Regalado E, Kamm K,

Stull JT. Altered smooth muscle cell force generation as a driver of thoracic

aortic aneurysms and dissections. Arterioscler Thromb Vasc Biol.

2017;37:26-34

272. Branchetti E, Poggio P, Sainger R, Shang E, Grau JB, Jackson BM, Lai EK,

Parmacek MS, Gorman RC, Gorman JH, Bavaria JE, Ferrari G. Oxidative

179

stress modulates vascular smooth muscle cell phenotype via ctgf in thoracic

aortic aneurysm. Cardiovasc Res. 2013;100:316-324

273. Crosas-Molist E, Meirelles T, López-Luque J, Serra-Peinado C, Selva J,

Caja L, Gorbenko del Blanco D, Uriarte JJ, Bertran E, Mendizábal Y,

Hernández V, García-Calero C, Busnadiego O, Condom E, Toral D,

Castellà M, Forteza A, Navajas D, Sarri E, Rodríguez-Pascual F, Dietz HC,

Fabregat I, Egea G. Vascular smooth muscle cell phenotypic changes in

patients with marfan syndrome. Arterioscler Thromb Vasc Biol.

2015;35:960-972

274. Inamoto S, Kwartler CS, Lafont AL, Liang YY, Fadulu VT, Duraisamy S,

Willing M, Estrera A, Safi H, Hannibal MC, Carey J, Wiktorowicz J, Tan FK,

Feng XH, Pannu H, Milewicz DM. Tgfbr2 mutations alter smooth muscle cell

phenotype and predispose to thoracic aortic aneurysms and dissections.

Cardiovasc Res. 2010;88:520-529

275. Ailawadi G, Moehle CW, Pei H, Walton SP, Yang Z, Kron IL, Lau CL, Owens

GK. Smooth muscle phenotypic modulation is an early event in aortic

aneurysms. J Thorac Cardiovasc Surg. 2009;138:1392-1399

276. Zhang J, Wang L, Fu W, Wang C, Guo D, Jiang J, Wang Y. Smooth muscle

cell phenotypic diversity between dissected and unaffected thoracic aortic

media. J Cardiovasc Surg (Torino). 2013;54:511-521

277. An Z, Liu Y, Song ZG, Tang H, Yuan Y, Xu ZY. Mechanisms of aortic

dissection smooth muscle cell phenotype switch. J Thorac Cardiovasc

Surg. 2017;154:1511-1521.e1516

180

278. Tsai SH, Huang PH, Hsu YJ, Peng YJ, Lee CH, Wang JC, Chen JW, Lin

SJ. Inhibition of hypoxia inducible factor-1alpha attenuates abdominal aortic

aneurysm progression through the down-regulation of matrix

metalloproteinases. Sci Rep. 2016;6:28612

279. Shankman LS, Gomez D, Cherepanova OA, Salmon M, Alencar GF,

Haskins RM, Swiatlowska P, Newman AA, Greene ES, Straub AC, Isakson

B, Randolph GJ, Owens GK. Klf4-dependent phenotypic modulation of

smooth muscle cells has a key role in atherosclerotic plaque pathogenesis.

Nat Med. 2015;21:628-637

280. Gordon D, Reidy MA, Benditt EP, Schwartz SM. Cell proliferation in human

coronary arteries. Proc Natl Acad Sci U S A. 1990;87:4600-4604

281. Aikawa M, Sakomura Y, Ueda M, Kimura K, Manabe I, Ishiwata S,

Komiyama N, Yamaguchi H, Yazaki Y, Nagai R. Redifferentiation of smooth

muscle cells after coronary angioplasty determined via myosin heavy chain

expression. Circulation. 1997;96:82-90

282. Kocher O, Gabbiani G. Cytoskeletal features of normal and atheromatous

human arterial smooth muscle cells. Hum Pathol. 1986;17:875-880

283. Mosse PR, Campbell GR, Campbell JH. Smooth muscle phenotypic

expression in human carotid arteries. Ii. Atherosclerosis-free diffuse intimal

thickenings compared with the media. Arteriosclerosis. 1986;6:664-669

284. van der Loop FT, Gabbiani G, Kohnen G, Ramaekers FC, van Eys GJ.

Differentiation of smooth muscle cells in human blood vessels as defined

181

by smoothelin, a novel marker for the contractile phenotype. Arterioscler

Thromb Vasc Biol. 1997;17:665-671

285. Hill BJ, Wamhoff BR, Sturek M. Functional nucleotide receptor expression

and sarcoplasmic reticulum morphology in dedifferentiated porcine

coronary smooth muscle cells. J Vasc Res. 2001;38:432-443

286. Kocher O, Gabbiani F, Gabbiani G, Reidy MA, Cokay MS, Peters H, Hüttner

I. Phenotypic features of smooth muscle cells during the evolution of

experimental carotid artery intimal thickening. Biochemical and morphologic

studies. Laboratory investigation; a journal of technical methods and

pathology. 1991;65:459-470

287. Dixon JL, Stoops JD, Parker JL, Laughlin MH, Weisman GA, Sturek M.

Dyslipidemia and vascular dysfunction in diabetic pigs fed an atherogenic

diet. Arterioscler Thromb Vasc Biol. 1999;19:2981-2992

288. Falk E, Nakano M, Bentzon JF, Finn AV, Virmani R. Update on acute

coronary syndromes: The pathologists' view. Eur Heart J. 2013;34:719-728

289. Newby AC, Zaltsman AB. Fibrous cap formation or destruction--the critical

importance of vascular smooth muscle cell proliferation, migration and

matrix formation. Cardiovasc Res. 1999;41:345-360

290. Bitto A, De Caridi G, Polito F, Calò M, Irrera N, Altavilla D, Spinelli F,

Squadrito F. Evidence for markers of hypoxia and apoptosis in explanted

human carotid atherosclerotic plaques. J Vasc Surg. 2010;52:1015-1021

291. Sluimer JC, Gasc J-M, van Wanroij JL, Kisters N, Groeneweg M, Sollewijn

Gelpke MD, Cleutjens JP, van den Akker LH, Corvol P, Wouters BG,

182

Daemen MJ, Bijnens A-PJ. Hypoxia, hypoxia-inducible transcription factor,

and macrophages in human atherosclerotic plaques are correlated with

intraplaque angiogenesis. J Am Coll Cardiol. 2008;51:1258-1265

292. Castellano J, Aledo R, Sendra J, Costales P, Juan-Babot O, Badimon L,

Llorente-Cortés V. Hypoxia stimulates low-density lipoprotein receptor–

related protein-1 expression through hypoxia-inducible factor-1α in human

vascular smooth muscle cells. Arterioscler Thromb Vasc Biol.

2011;31:1411-1420

293. Liu D, Lei L, Desir M, Huang Y, Cleman J, Jiang W, Fernandez-Hernando

C, Di Lorenzo A, Sessa WC, Giordano FJ. Smooth muscle hypoxia-

inducible factor 1alpha links intravascular pressure and atherosclerosis--

brief report. Arterioscler Thromb Vasc Biol. 2016;36:442-445

294. Ben-Shoshan J, Afek A, Maysel-Auslender S, Barzelay A, Rubinstein A,

Keren G, George J. Hif-1α overexpression and experimental murine

atherosclerosis. Arterioscler Thromb Vasc Biol. 2009;29:665-670

295. Ghaderian SM, Lindsey NJ, Graham AM, Homer-Vanniasinkam S,

Akbarzadeh Najar R. Pathogenic mechanisms in varicose vein disease:

The role of hypoxia and inflammation. Pathology. 2010;42:446-453

296. Lee JD, Yang WK, Lee TH. Increased expression of hypoxia-inducible

factor-1alpha and bcl-2 in varicocele and varicose veins. Ann Vasc Surg.

2012;26:1100-1105

183

297. Lim CS, Gohel MS, Shepherd AC, Paleolog E, Davies AH. Venous hypoxia:

A poorly studied etiological factor of varicose veins. J Vasc Res.

2011;48:185-194

298. Lim CS, Kiriakidis S, Paleolog EM, Davies AH. Increased activation of the

hypoxia-inducible factor pathway in varicose veins. J Vasc Surg.

2012;55:1427-1439

299. Lim CS, Kiriakidis S, Sandison A, Paleolog EM, Davies AH. Hypoxia-

inducible factor pathway and diseases of the vascular wall. J Vasc Surg.

2013;58:219-230

300. Misra S, Shergill U, Yang B, Janardhanan R, Misra KD. Increased

expression of hif-1alpha, vegf-a and its receptors, mmp-2, timp-1, and

adamts-1 at the venous stenosis of arteriovenous fistula in a mouse model

with renal insufficiency. J Vasc Interv Radiol. 2010;21:1255-1261

184