Вы находитесь на странице: 1из 13

Atherosclerosis 248 (2016) 97e109

Contents lists available at ScienceDirect

Atherosclerosis
journal homepage: www.elsevier.com/locate/atherosclerosis

Review article

Vascular endothelium e Gatekeeper of vessel health


Paul A. Cahill a, Eileen M. Redmond b, *
a
Vascular Biology and Therapeutics Laboratory, Dublin City University, Dublin, Ireland
b
Department of Surgery, University of Rochester Medical Center, Rochester, NY, USA

a r t i c l e i n f o a b s t r a c t

Article history: The vascular endothelium is an interface between the blood stream and the vessel wall. Changes in this
Received 21 August 2015 single cell layer of the artery wall are believed of primary importance in the pathogenesis of vascular
Received in revised form disease/atherosclerosis. The endothelium responds to humoral, neural and especially hemodynamic
3 March 2016
stimuli and regulates platelet function, inammatory responses, vascular smooth muscle cell growth and
Accepted 4 March 2016
Available online 9 March 2016
migration, in addition to modulating vascular tone by synthesizing and releasing vasoactive substances.
Compromised endothelial function contributes to the pathogenesis of cardiovascular disease; endothelial
dysfunction is associated with risk factors, correlates with disease progression, and predicts cardio-
Keywords:
Endothelium
vascular events. Therapies for atherosclerosis have been developed, therefore, that are directed towards
Endothelial dysfunction improving endothelial function.
Atherosclerosis 2016 Elsevier Ireland Ltd. All rights reserved.
Cardiovascular disease
Vessel remodeling
Clinical therapies
Review

1. Atherosclerosis foam cell formation following uptake of oxidized LDL via scavenger
receptors (SR-A type I and II and CD36), and stimulation of the
Atherosclerosis is a chronic inammatory disease in which the migration of medial SMC into the intima where they proliferate in
artery wall thickens as a result of the accumulation of cholesterol, response to growth factors such as platelet derived growth factor
macrophages and smooth muscle cells (SMC), ultimately restricting (PDGF) [13]. In the intima, SMC produce extracellular matrix mol-
blood ow through the artery. It is the main pathologic condition ecules including collagen and elastin. The most common clinical
underlying coronary artery and cerebrovascular disease leading to complication of atherosclerosis occurs upon plaque rupture that
heart attack and stroke, respectively. In the pathogenesis of allows blood components to come into contact with plaque lipids
atherosclerosis (reviewed in Ref. [1e4]), increases in plasma low and tissue factor, resulting in thrombus formation. While several
density lipoprotein (LDL) leads to a proportional increase in the cell types are clearly involved in the pathogenesis of atherosclerotic
entry of cholesterol laden LDL particles into the arterial wall across plaques, endothelial compromise/dysfunction is deemed of
a compromised/dysfunctional endothelial monolayer, where it particular importance as it is a necessary and initiating occurrence
accumulates. Once there, it can become oxidized, by free radical for atherogenesis to proceed. The endothelial lining can be viewed
production from adjacent endothelium, smooth muscle cells or as the rst line of defense between risk factors and vascular disease.
isolated macrophages [5e8]. Oxidized LDL has numerous effects on
a variety of cells, many of which are believed to cumulatively
2. Arterial endothelium: structure, function
exacerbate atherothrombosis ([9] for review). These include pro-
motion of monocyte adhesion and inltration to the intima by
The vascular endothelium lines the entire circulatory system. In
causing production of monocyte chemotactic protein-1 (MCP-1) by
coronary arteries, this single layer, together with some extracellular
endothelium and expression by endothelium of monocyte-binding
matrix, comprises the tunica intima. Originally thought of simply as
proteins including intercellular adhesion molecule-1 (ICAM-1),
a passive barrier, it is now viewed as an organ whose normal
functioning is crucial to maintaining vascular health, and
* Corresponding author. University of Rochester Medical Center, Department of
whose dysfunction is key in the initiation, progression and clinical
Surgery, Box SURG, 601 Elmwood Avenue, Rochester, NY, 14642-8410, USA. complications of vascular disease. Vascular endothelium acts as
E-mail address: Eileen_Redmond@urmc.rochester.edu (E.M. Redmond). a selectively permeable barrier between extravascular and

http://dx.doi.org/10.1016/j.atherosclerosis.2016.03.007
0021-9150/ 2016 Elsevier Ireland Ltd. All rights reserved.
98 P.A. Cahill, E.M. Redmond / Atherosclerosis 248 (2016) 97e109

intravascular compartments and provides a nonthrombogenic lin- exposed to low, disturbed or oscillating ow, while laminar ow
ing for the cardiovascular system. It is poised in an anatomic and high shear stress are seemingly atheroprotective [22]. In
location that allows interaction not only with circulating blood addition to inhibition of thrombosis and inhibition of endothelial
components and cells, but also with cells in the vessel wall. apoptosis, limitation of permeability is now a well-characterized
Endothelial cells are long, at cells orientated in the long axis of the atheroprotective mechanism of laminar ow and high shear
vessel. An average endothelial cell is 20e40 mM long, 10e15 mM stress [22]. Conversely, at regions of pathological low shear stress,
wide and only 0.1e0.5 mM thick. Electron microscopy of arterial increased endothelial permeability results in enhanced inltration
endothelium reveals it to be of the continuous type characterized of LDL and its local accumulation, which is a critical initial event in
by tight junctions at the lateral borders of each cell that restrict the the development of atherosclerosis [23,24]. Rozenberg et al.,
movement of macromolecules, and a complex micro-vesicular recently reported that Histamine, acting via its H1 receptor drives
system implicated in macromolecular transport. Despite its the formation of atherosclerotic lesions through an increased
apparent morphological simplicity and relative homogeneity, there vascular permeability for LDL [25]. Mullick et al., showed that
is evidence of regional and species variation manifested by differ- exposure to cigarette smoke injures the endothelium, resulting in
ences in permeability, responsiveness and biosynthesis. increased arterial permeability and increased LDL accumulation
[26]. A recent study provided evidence of a role for lipoxygenase
3. Barrier function and its metabolite hydroxyeicosatetraenoic acid (HETE) in high fat
diet-induced endothelial tight junction disruption [27], thus
The role of the endothelium as a semipermeable barrier is one of providing a possible mechanistic link between lifestyle and
its most basic functions. It regulates transport of macromolecules atherosclerosis. Therefore, a wide variety of injurious stimuli (e.g.,
between the vascular lumen and vascular smooth muscle. There are certain hemodynamic forces, inammatory mediators, bacterial
several different mechanisms by which macromolecules cross the endotoxin LPS, environmental toxins, high fat diet) can contribute
endothelial barrier; through the endothelial cells themselves, by to endothelial dysfunction by increasing endothelial permeability
diffusing laterally within the endothelial cell membrane and thus and subsequently arterial lipid accumulation in the subendothelial
around the cell, through endothelial cell-to cell junctions, through space, thereby initiating atherosclerotic plaque development. On
endothelial gaps, or via vesicular transport. As most biologic mol- the other hand, factors shown to maintain endothelial barrier
ecules are large, hydrophilic, anionic molecules, which are unable function include high density lipoprotein (HDL), and physical ex-
to diffuse into and through bilayer membranes, most are thought to ercise (reviewed in [28]). With respect to the latter, there is
move through intercellular junctions between cells or by vesicular considerable evidence of a direct relationship between exercise and
transport, or the formation of transient channels resulting from vascular health. The effect of exercise on maintaining endothelial
vesicle fusion [10]. Reorganization of the inter-cellular junctions, barrier function is likely a consequence of exercise increasing blood
involving actin and myosin or direct dissolution of junctional ow and shear stress, which in turn releases vasoprotective mol-
contacts, is believed to be the primary mechanism by which ecules such as nitric oxide (NO) and prostacyclin (PGI2). It is also
endothelial permeability to water, small and large solutes is appreciated that Sphingosine-1-phosphate (S1P), a bioactive
increased [11]. sphingolipid associated with HDL and found mainly in the blood
and lymph, robustly promotes endothelial barrier function [29,30].
4. Changes in permeability In particular, growing evidence indicates that HDL-associated S1P
mediates the benecial effects on endothelial integrity [31,32].
Various physiologic and pathophysiologic stimuli can induce, Other endogenous factors known to increase endothelial barrier
acutely and chronically, dramatic changes in endothelial perme- function and decrease permeability are angiopoietin-1 [33], and the
ability. For example thrombin, histamine and other acute inam- second messenger cyclic adenosine monophosphate (cAMP) and
matory mediators can act on endothelium to stimulate opening of agonists such as Serotonin and b-adrenergic agonists that increase
their intercellular junctions at the level of adherens and tight it [34,35] (Table 1).
junctional complexes [12]. The signaling involved in mediating
these responses include protein kinase C (PKC)einduced phos- 5. Endothelial glycocalyx
phorylation of linking proteins at the cell-cell and cell-matrix
junctions, leading to actin reorganization, cell rounding and Consisting of a negatively charged, organized mesh of mem-
increased paracellular transport. Myosin light chain kinase (MLCK) branous glycoproteins, proteoglycans and associated plasma pro-
may also be activated by inammatory mediators leading to actin- teins, the endothelial glycocalyx (100e750 nm thick) is recognized
myosin based retraction of endothelial cells [12]. Vascular endo- as contributing, together with the endothelium, to the protection of
thelial growth factors (VEGFs) are key regulators of vascular the vascular wall against disease (for review [36,37]). Improved
permeability via nitric oxide synthase (NOS) regulation [13,14], xing and imaging techniques have allowed in vivo visualization of
facilitated by multiple mechanisms including gap formation and the endothelial glycocalyx and provided evidence that there are
vesiculo-vacuolar organelle formation [15] (reviewed in Ref. [16]). signicant reductions in its extent during acute and chronic in-
The bacterial endotoxin lipopolysaccharide (LPS) causes endothe- ammatory challenge in man. Indeed, destruction of the glycocalyx
lial hyperpermeability by stimulating the small GTPase, RhoA, and has been directly and indirectly evidenced in several studies, e.g.
its effector Rho Kinase (ROCK) [17]. Endothelial permeability is also after ischemic challenge [38e40], during redox stress [39], and
inuenced by uid shear stress, the tangential frictional force after inammation [40,41]. Deterioration or destruction of the
exerted by owing blood [10,18]. Investigation of the relationship glycocalyx is associated with pathophysiological sequelae including
between albumin permeability and shear stress magnitude in vivo increased endothelial permeability, platelet aggregation and loss of
revealed that endothelium exposed to low wall shear stresses was vascular responsiveness [42]. There are interesting data supporting
more likely to have elevated macromolecule permeability [19]. Of an important role for the glycocalyx in the initiation and progres-
importance with respect to vascular disease, low density lipopro- sion of atherosclerosis. Van den Berg et al., reported thinning of the
tein (LDL) accumulation is greater at these more permeable areas glycocalyx in mice fed a cholesterol rich diet, and demonstrated less
exposed to low wall shear stress [20,21]. It is now recognized that glycocalyx in regions of the vessel at high atherogenic risk (i.e., at
atherosclerotic lesions preferentially develop at vessel segments branch points, areas of curvature) [43]. Moreover, the authors
P.A. Cahill, E.M. Redmond / Atherosclerosis 248 (2016) 97e109 99

Table 1
Factors affecting endothelial cell permeability.

Increase permeability Decrease permeability

'Low' shear stress 'High' shear stress


Vascular endothelial growth factor (VEGF) High density lipoprotein (HDL)/Sphingosine 1 phosphate(S1P)
Lipopolysaccharide (LPS)
High fat diet Physical exercise
Phospholipid oxidative products Increased cellular cAMP (by e.g., Serotonin or b-adrenergics)
Inammatory mediators (e.g., thrombin, Histamine, TGF(b) Cigarette smoke Angiopoietin-1

found an inverse relation between glycocalyx thickness and the endothelial cells [62]. Hypercholesterolemic mice decient for
intima-media ratio [43]. These ndings support the hypothesis that syndecan 4 have increased atherosclerotic plaque when compared
diminution of the glycocalyx, perhaps as a consequence of turbu- with control mice [62]. A ow-dependent GTP exchange factor
lent ow/low shear stress, contributes to the vulnerability of re- called TIAM1 reportedly links platelet endothelial cell adhesion
gions that are at high atherogenic risk [44]. Vasculoprotective molecule 1(PECAM-1) mechanotransduction to focal activation of
effects attributed to the glycocalyx include fortifying endothelial the small GTPase Rac1 which in turn triggers the Nuclear factor
barrier function [45], inhibiting coagulation and leukocyte adhe- kappa B (NF-kB) pathway and production of reactive oxygen spe-
sion [46] and mediating shear stress-induced NO release [47,48]. cies [63]. Another mechanotransducer candidate is the ion channel
The seemingly atheroprotective role of the gylcocalyx means that it protein Piezo1, a mediator of shear stresseinduced calcium inux
is attracting attention as a potential novel diagnostic or therapeutic [64]. Piezo1-mediated calcium inux is important for calpain ac-
target for atherosclerosis [49,42]. Approaches to increase the gly- tivity and subsequent rearrangement of focal adhesions for ow
cocalyx or to reverse its damage, as well as tactics to maintain it alignment. Endothelial cells isolated from Piezo1/ mice failed to
mechanically and chemically, are being tested as viable clinical align properly when exposed to atheroprotective ow conditions
options at this stage [37,42]. Indeed, clinical trial data support a [65]. A role for the G proteinecoupled Sphoingosine-1-phosphate
protective role for the compounds Diazoxide and Sulodexide on receptor (S1P1) in endothelial mechanotransduction has also
endothelial glycocalyx in patients after coronary bypass grafting been put forward as the expression of S1P1 was critical for ow-
and in those with type 2 diabetes, respectively [50,51]. mediated directional alignment in cultured endothelial cells
in vitro and in vivo [66].
6. Hemodynamic forces Hemodynamic forces are ubiquitous and fundamental physio-
logic stimuli for vascular cells, and are believed to critically inu-
Cells of the arteries are continuously exposed to pulsatile blood ence atherogenesis by regulating endothelial cell function, smooth
ow and are therefore subjected to the hemodynamic forces shear muscle behavior, and the interaction of endothelial cells with
stress and cyclic strain. Shear stress is the tangential frictional force smooth muscle cells [67] and with leucocytes and other blood
acting predominately at the endothelial cell surface, whereas cyclic constituents [22,68,69]. Of note, ow patterns and hemodynamic
strain/stretch acts perpendicular to the vessel wall and affects both forces are not uniform throughout the vasculature. While blood
endothelial and smooth muscle cells together with the underlying ow is essentially laminar throughout the arterial system, in
matrix [52]. Shear stress (t) (expressed as dynes/cm2) can be straight parts of the arterial tree blood ow is generally steady and
calculated using the formula, t 4mQ/pr3, where m is blood vis- unidirectional and wall shear stress is high (>15 dyn/cm2; physi-
cosity, Q is blood ow volume, p is the ratio of the circumference of ologic ow). At branch points and curvatures, blood ow is
a circle to its diameter, and r is the radius of the blood vessel. The disturbed or oscillatory with low net wall shear stress (0e4 dyn/
pumping heart causes pulsatile changes in blood pressure that cm2; pathologic low ow). Atherosclerotic lesion development
stretch the vessel wall circumferentially and create cyclic strain in generally correlates with the latter [70]. This type of observation
vascular cells. The degree of stretch is around 9%e12% in the aorta gave rise to the concept, now accepted, that disturbed ow patterns
and 1%e2% in the carotid arteries [53]. A variety of devices have are atherogenic whereas steady high ow patterns are atheropro-
been employed by research laboratories to expose cultured cells to tective (reviewed in Ref. [71]). Several groups have investigated the
dened mechanical forces. Those used to apply shear stress include effects of various ow patterns on endothelial cell biology in vivo
the cone plate viscometer [54], the parallel ow chamber [55] and and in vitro over the last 30 years, and the emerging consensus is
the perfused capillary culture system [56,57]. In the case of cyclic that physiologic ow favors the expression of endothelial genes and
stretch, researchers have grown cells on a variety of elastic sub- bioactive products that are protective against atherosclerosis,
strates [58] that can then be periodically stretched; e.g., the Flex- whereas pathologic ow stimulates genes and products that pro-
ercell system [59]. In order to respond to mechanical forces such as mote atherogenesis [22,68,69,70,72] (Fig. 1). Indeed, studies using
shear stress or cyclic strain and convert it into a biochemical DNA microarrays have revealed striking differences between the
response endothelial cells must express special receptors or numbers and kinds of endothelial genes that respond to steady
mechanotransducers. Biomechanical studies have suggested many ow versus disturbed ow [73]. Of interest, it is emerging that ow
different mechanotransducer candidates including ion channels, modulates epigenetic DNA methylation patterns via alterations in
tyrosine kinase receptors, G-protein coupled receptors, a caveolae- DNA methyltransferase activity, in particular DNMT1, that may
mediated process, cell adhesion molecules, the glycocalyx and contribute to the endothelial phenotypes observed in regions of
primary cilia (Reviewed in Ref. [53,60]). Certain biomechanically disturbed ow [74]. For example, disturbed ow patterns increase
induced events appear to involve transcriptional regulation and methylation of the proximal promoter of Kruppel like factor 4
shear stress-response elements have been identied in the pro- (KLF4), thus inhibiting atheroprotective KLF4 transcription in
moters of multiple endothelial cell-expressed genes (e.g., [61]). atherosclerosis-susceptible regions [75]. Findings such as these
Recent studies have uncovered other contenders as mechano- suggest that DNA methylation may play a key role in maintaining
transducers. Syndecan 4, a transmembrane heparan sulfate pro- endothelial cell homeostasis and in vascular disease development
teoglycan, is required for proper ow alignment in cultured and encourage continued investigation into epigenetic
100 P.A. Cahill, E.M. Redmond / Atherosclerosis 248 (2016) 97e109

Fig. 1. Effect of different ow patterns on (a) atherosclerotic plaque development and (b) endothelial phenotype and activity. In straight parts of the arterial tree blood ow is steady
and unidirectional and wall shear stress high. At branch points and curvatures, blood ow is disturbed or oscillating and net wall shear stress is low (areas in blue in schematic of
arterial bifurcation). Steady, high shear ow is atheroprotective, whereas disturbed ow, low shear is atherogenic. (For interpretation of the references to colour in this gure
legend, the reader is referred to the web version of this article.)

mechanisms relevant to vascular health and disease. derived growth factor (PDGF) [94], basic broblast growth factor
(bFGF) [95], transforming growth factor beta (TGFb) [96] and
7. Hemodynamic forces and endothelium Interleukin-1 and -6 [97].
Modulatory inuences of shear stress on endothelial cell adhe-
Hemodynamic forces impact multiple aspects of endothelial cell sion molecule (e.g., vascular cell adhesion molecule-1, VCAM-1)
biology and function (reviewed in Ref. [53]), all of which may in- and selectin (e.g., P-selectin) expression have also been reported,
uence atherosclerotic plaque initiation and development. Shear effects with important implications to not only platelet aggregation
stress affects endothelial cell morphology. Endothelial cells [98], but also to the adhesion of leukocytes, in particular mono-
exposed to steady rapid ow are elongated in shape and aligned cytes, to the endothelium [99,100]. Of note, following their adhe-
with their long axis in the direction of ow, in contrast to the sion, migration of monocytes across the endothelial barrier is
rounder shape and non-uniform orientation and greater perme- considered a signicant early event in the development of
ability of EC in segments exposed to disturbed ow [76]. Shear atherosclerotic lesions [1].
stress plays a role in controlling endothelial cell proliferation and
apoptosis; e.g., steady ow reduced EC proliferation [77], whereas 7.1. Reactive oxygen species
disturbed ow increased EC turnover and stimulated apoptosis
[78]. Cyclic strain also promotes endothelial migration and in vitro Increased production of reactive oxygen species (ROS) contrib-
tube formation (an index of angiogenesis) [79]. Growth and utes to mechanisms of vascular/endothelial dysfunction and
migration of endothelial cells becomes important in the repair of atherosclerosis [101,102]. ROS is a term that includes oxygen radi-
areas denuded of endothelium as a result of injury, be it toxic or cals such as superoxide (O 2 ) and hydroxyl radical (OH), as well as
mechanical and/or iatrogenic. non-radical derivatives of O2 including hydrogen peroxide (H2O2)
Endothelial cells respond to hemodynamic forces by altering and Ozone (O3). Oxidative stress is mainly caused by an imbalance
their production of vasoactive substances (Fig. 2). Increases in shear between the activity of endogenous pro-oxidative enzymes (such
stress generally causes vasodilation, mediated for the most part by as nicotinamide adenine dinucleotide phosphate-oxidase (NADPH)
increased endothelila nitric oxide synthase (eNOS) activity and NO oxidase and xanthine oxidase) and anti-oxidative enzymes (such as
production [57,80]. Indeed, shear stress is considered the premiere superoxide dismutase, glutathione peroxidase and heme oxygen-
physiologic stimulus for this potent vasorelaxant molecule. Other ase) in favor of the former. Modulation of ROS production from
endothelial-derived vasoactive substances altered by shear stress endothelium by shear stress and cyclic stretch has been reported
include PGI2 [81,57,82] and endothelin-1 (ET-1) [83,84]. The vaso- [60,103]. For example, Chatterjee et al., showed that in response to a
dilators NO and PGI2, and the vasoconstrictor ET-1 are not only decrease of shear, such as occurs during ischemia, membrane de-
important in regulating vascular tone at the level of vascular polarization via ATP-sensitive K channel closure initiated a
smooth muscle, but in addition, these agents inhibit (NO, PGI2), and signaling cascade leading to NADPH oxidase activation and ROS
stimulate (ET-1), SMC growth and migration [85e87]. Nitric oxide production [104]. In contrast, shear stress upregulated the
and PGI2 are also potent inhibitors of platelet aggregation [88,89]. expression of antioxidant enzymes such as peroxiredoxin 1 (PRX 1)
Besides stimulating NO and PGI2, shear stress contributes to [105]. The accepted idea, from evidence garnered over the last 20
maintaining the endothelium as non-thrombogenic by stimulating years, is that ow patterns crucially regulate the ROS/NO balance,
thrombomodulin [90], heparin sulfate proteoglycans [91] and tis- with steady ow (high shear) causing lower levels of ROS and
sue type plasminogen activator (tPA) [92]. greater NO bioavailability (considered anti-atherogenic), whereas
Hemodynamic forces also affect the production by the endo- disturbed ow (low shear) results in higher ROS and lower NO
thelium of a variety of growth factors and cytokines, many of which bioavailability (considered pro-atherogenic) (reviewed in Ref. [60]).
can affect the underlying smooth muscle cells, stimulating them to Steady ow favors the activation of key transcription factors such as
de-differentiate from a quiescent, contractile phenotype to the Nuclear factor (erythroid-derived 2)-like 2 (Nrf2) and Krppel-like
migratory and proliferative synthetic phenotype fundamental to Factor 2 (KLF2) to promote an anti-atherogenic environment by
the pathogenesis of vascular disease [93]. These include platelet- enhancing the expression of superoxide dismutase and heme
P.A. Cahill, E.M. Redmond / Atherosclerosis 248 (2016) 97e109 101

Fig. 2. Shear stress and cyclic strain are important stimuli for endothelial cells (EC). These hemodynamic forces are sensed by mechanoreceptors that include ion channels, G protein
coupled receptors, integrins, cytoskeletal components and receptor tyrosine kinases. Hemodynamic forces modulate endothelial release and expression of a myriad of substances
including vasoactive agents, growth factors, cytokines, microRNAs (miRNAs), thrombomodulin and reactive oxygen species (ROS) that may affect the aggregation and adhesion of
cells circulating in the blood (i.e., platelets and monocytes), the permeability, adhesivity and angiogenic potential of EC themselves, as well as the growth, migration, vasoreactivity
and extracellular matrix (ECM) synthesis of the underlying vascular smooth muscle (SMC).

oxygenase [106], whereas irregular ow leads to activation of key hemodynamic/mechanical forces that could be an underlying
transcription factors such as AP-1 and NF-kB leading to enhanced mechanism for the progression of atherosclerosis [112].
expression of pro-atherogenic agents such as monocyte chemo-
tactic protein1 (MCP-1) and intracellular adhesion molecule 1 9. Physical exercise
(ICAM-1) [60].
Of interest, physical exercise seems to be naturally anti-
8. Cyclic strain atherogenic, preserving vascular function with aging (reviewed in
Refs. [113,114]). Some data suggest that exercise inhibits plaque
As mentioned earlier, endothelial and smooth muscle cells development and induces the regression of coronary stenosis
in vivo are also exposed to cyclic strain, a circumferential stretching [115,116]. Furthermore, experimental studies have revealed that
synchronous with cardiac cycle-induced changes in blood pressure. exercise prevents the conversion of plaques into a vulnerable
Like shear stress, cyclic strain is a force of physiological and path- phenotype, thus averting the appearance of fatal lesions [117].
ological interest, sensed by mechanosensors, that modulates While the molecular mechanisms involved are not yet fully un-
endothelial cell intracellular signaling resulting in changes in pro- derstood, strong evidence supports the importance of increased
liferation, apoptosis [107], migration [79], angiogenic potential shear stress, as a result of exercise-induced increased heart rate and
[79,59] and matrix remodeling [108,109,103]. Cyclic strain increases blood ow, as the primary signal acting on the endothelium pro-
under certain pathological conditions such as hypertension and duced by exercise [118], resulting in down-regulated endothelial
congenital heart disease. Kou et al., reported that lower (5%) cyclic angiotensin II type 1 receptor expression, leading to decreases in
strain increases, but higher (20%) cyclic strain decreases, endothe- NADPH oxidase activity and superoxide anion production, which in
lial cell survival and angiogenesis via the NAD(P)H subunit p22phox turn decreases ROS generation, and preserves endothelial NO
pathway [110]. Physiologic cyclic strain regulated, in a p38-and bioavailability and its protective anti-atherogenic effects [119]. Of
receptor tyrosine kinase-dependent manner, the release of further note, atherosclerosis is a chronic inammatory disease
thrombomodulin a membrane glycoprotein expressed on the [120] and several studies support an inverse correlation between
lumenal surface of endothelial cells that promotes anti-coagulant physical exercise and serum markers of inammation such as C-
and anti-inammatory properties [111]. Liu et al., reported that reactive protein (CRP), tumor necrosis factor-a (TNFa) and
cyclic strain stimulates Heme oxygenase 1 (HO-1) in a manner Interleukin-6 [121e123,124]. In addition, recent meta analysis of 28
dependent on the production of ROS and mediated by Nrf2 [107]. individual studies showed that physical exercise has several non-
They also showed that cyclic strain inhibits cytokine-mediated acute effects expected to reduce atherogenesis; i.e., decreasing
endothelial cell apoptosis and that HO-1 underlies this cytopro- adhesion molecule VCAM-1 and ICAM-1 expression, reducing the
tective effect [107]. Thus, there is ample evidence that hemody- angiogenic factor VEGF, and increasing endothelial progenitor cells
namic forces, in particular shear stress and cyclic strain, are (EPC) [125].
critically important stimuli for the endothelium, affecting endo-
thelial cell function as well as modulating the endothelial release 10. Endothelium's vasoactive role (vasodilators)
and expression of growth factors, cytokines, vasodilators, vaso-
constrictors, reactive oxygen species and cell adhesion molecules - 10.1. EDRF/NO
all of which impact the underlying vascular smooth muscle and/or
the interaction of circulating inammatory and immune cells with The smooth muscle response to sympathetic/parasympathetic
the endothelium and thereby affect atherogenesis (Fig. 2). There nerve stimulation is a major contributor to maintenance of car-
is also evidence that atherosclerosis risk factors themselves (e.g. diovascular tone throughout the vascular tree. However, starting
hypertension, dyslipidemia, smoking) result in altered local with the discovery in 1980 by Furchgott and Zawadski of
102 P.A. Cahill, E.M. Redmond / Atherosclerosis 248 (2016) 97e109

endothelial-derived relaxing factor (EDRF) [126], identied chem- activation of NOS) and folic acid (which may mimic BH4). Of note
ically as endogenous nitric oxide (NO) [127], endothelial cells were hormones differentially regulate eNOS and NO bioavailability, with
subsequently recognized as playing a signicant role in the local estrogen, insulin and growth hormone increasing it, and proges-
regulation of vascular tone. This local regulation has implications terone and glucocorticoids decreasing it [136]. Antioxidants such as
for atherogenesis. Nitric oxide is synthesized by the heme- melatonin and Vitamin C have been found to increase blood ow
containing, calcium and calmodulin-dependent enzyme nitric ox- via an NO-dependent mechanism [137]. Evidence is emerging that
ide synthase in endothelial cells (eNOS) from L-arginine in a reac- statins, a class of lipid-lowering drugs, may have differential effects
tion that produces stoichiometric amounts of L-citrulline [128,129]. on eNOS and iNOS [138e140]. Of potential interest to chocolate
Activation of NOS and release of the free radical NO, which easily lovers, Taubert et al., reported that regular intake of polyphenol-
diffuses across biological membranes, results in stimulation of a rich dark chocolate increases NO bioavailability [141]. Treatment
soluble guanylyl cyclase leading to a profound increase in intra- of endothelial cells in culture with alcohol increases NO production
cellular cGMP within the adjacent smooth muscle cells, thus by enhancing NOS activity [142e144], and alcohol feeding in rats
resulting in their relaxation and consequent vasodilation [129]. NO increases aortic NO bioavailability [145]. These NO effects might
has a wide range of actions important in maintaining vascular explain some of the putative cardioprotection seen with moderate
homeostasis. In addition to causing vasodilation, it has anti- alcohol consumption [146e148]. However, while there is a lot of
proliferative, antioxidant and anti-inammatory properties that promise in capitalizing on the pre-clinical ndings with respect to
inhibit atherogenesis ([130] for recent review). As mentioned pre- NO biology, translation to new modalities for cardiovascular dis-
viously, shear stress resulting from blood ow upregulates eNOS in ease management has not been uniformly fruitful [134]. Results of
endothelium. Shear stress-responsive elements have been identi- dietary nitrate supplementation in clinical trials, for example, have
ed in the eNOS promotor sequence on cloning of the eNOS cDNA yielded variable outcomes with some benecial, and some with no
and identication of the promotor region [131]. eNOS activity can measurable effect [149,150].
also be stimulated by receptor mediated agonists-e.g., bradykinin, Moreover, it is important to note that certain conditions such as
acetylcholine and thrombin via palmitoylation, binding to hypercholesterolemia, hypertension and diabetes can lead to
calmodulin/calcium, displacement of caveolin and release from the increased vascular production of superoxide (O 2 ). Superoxide can
plasma membrane [132]. react with the increased NO also produced in such circumstances
The benecial effect of physical exercise on cardiovascular dis- forming the strong oxidant peroxynitrite (ONOO) that causes
ease is believed partly due to increased NO bioavailability [119,133]. protein nitration, lipid peroxidation, DNA damage and cell death
Conversely, common risk factors for cardiovascular disease such as [151]. Excessive peroxinitrite is believed an important contributor
hypercholesterolemia, hypertension, smoking and diabetes melli- to cardiovascular pathologies including atherosclerosis. Thus, NO
tus are associated with reduced NO in the arterial wall [134] (Fig. 3). must be considered a pleiotropic molecule, with both physiological
Indeed, the term endothelial dysfunction has become synonymous and pathological potential as it has the capacity for a dual role as a
with reduced biological activity of NO. Because of these ndings, protective agent, or as a target for generating harmful peroxinitrite,
numerous therapies have been investigated based on enhancing depending on the oxidative milieu [152,151]. As with most things in
NO release, thereby reversing endothelial dysfunction and pre- biology, it is the balance that is important and that determines
venting atherogenesis. NO donors (e.g., nitroglycerin) are physiological homeostasis or pathology development.
frequently used in cardiovascular disease patients. NO bioavail-
ability can be restored by moderate physical exercise, antioxidants 10.2. PGI2
and L-arginine, the natural precursor of NO [134]. Several small-
scale studies have demonstrated that intravenous L-arginine aug- Another important endothelial-derived vasodilator and athe-
ments endothelial function and improves exercise ability in pa- roprotective agent, whose release is stimulated by shear stress and
tients with cardiovascular disease by enhancing vasodilation and in response to acetylcholine, is prostacyclin (PGI2) [153,154]. PGI2 is
reducing monocyte adhesion [135]. Other agents under investiga- synthesized by cyclooxygenase-2 (COX-2) from arachidonic acid
tion include tetrahydrobiopterin (BH4, a co-factor required for [155] and increases the second messenger cyclic adenosine
monophosphate (cAMP) in smooth muscle cells as well as in
platelets. PGI2 mediates its actions through specic cell surface, 7-
membrane spanning, G protein-coupled receptors, known as PG12
receptor or IP [156,157]. In contrast to NO, PGI2 does not appear to
contribute to the maintenance of basal vascular tone of large
conduit arteries. However, in patients with a decreased NO
bioavailability as in atherosclerosis, COX-2-derived prostaglandins
can play a compensatory role [158]. Moreover, PGI2 exerts impor-
tant platelet inhibitory effects and can act synergistically with NO
in this regard [159]. Like NO, PGI2 can also inhibit leukocyte
adhesion and vascular smooth muscle proliferation [160] and
crosstalk between the two agents exists. PGI2 facilitates the release
of NO by endothelial cells [161], and conversely, the action of PGI2
in vascular smooth muscle is potentiated by NO and NO indirectly
via cGMP inhibition of phosphodiesterase 3, prolongs the half-life
of cAMP [162]. PGI2's effects counteract those of thromboxane
(TXA2) that is synthesized through COX-1 and causes platelet ag-
gregation, vasoconstriction and vascular proliferation, and the
balance of these two prostanoids is thus important in cardiovas-
Fig. 3. Common risk factors for cardiovascular disease (CVD) such as those listed here
are associated with 'endothelial dysfunction', a condition synonymous with reduced
cular hemostasis [157]. In accordance with the concept of PGI2
biological activity of nitric oxide (NO), and believed to be an initiating factor for being atheroprotective, an orally active PGI2 stable analog, Bera-
atherogenesis. prost, or PGI2 synthase (PGIS) gene transfer inhibited neointimal
P.A. Cahill, E.M. Redmond / Atherosclerosis 248 (2016) 97e109 103

formation in animal models of arterial injury [163,164]. Beraprost is Several ET receptor antagonists have been developed including
widely used in therapy for pulmonary artery hypertension and mixed antagonists Bosetan and the more potent Macitentan, and
chronic arterial obstruction [165]. The withdrawal of the Cox-2 the selective ETA antagonist Ambrisentan [185,186]. Restoration of
inhibitor Vioxx in 2004 due to increased cardiovascular events the impaired activity of the NO system is seen following selective
supports the critical role of PGI2 in inhibiting atherothrombosis in ETA receptor antagonism in a range of animal models of athero-
humans. In a clinical study, the IP variant (R212C), which was sclerosis, together with a reversal of atherosclerotic lesion devel-
defective in cAMP production, was closely linked to disease severity opment [187,188]. However mixed ETA/B receptor antagonism also
and adverse cardiovascular events in patients with cardiovascular seemed benecial in inhibiting lesion development in mice [189].
diseases [166]. In addition to the classical IP-cAMP pathway, evi- Whether or not both receptors should be blocked to provide most
dence suggests that PGI2 also mediates its effects on the vasculature clinical benet in the case of cardiovascular disease is a matter of
via the nuclear receptor peroxisome proliferator activated receptor ongoing debate. More recently, combined inhibitors of ECE and
d (PPAR)[167]. neural endopeptidase (NEP) such as SLV306 (daglutril) are being
widely tested in clinical trials [186].
10.3. EDHF
11.2. Angiotensin-II/AT1R
Any vasodilator response that persists after combined inhibition
of NO (e.g., by L-NMMA) and PGI2 (e.g., by aspirin) is attributed to The renineangiotensin system (RAS) is a circulating hormonal
substances that cause hyperpolarization of the underlying vascular system that regulates blood pressure, blood ow, uid volume and
smooth muscle via a mechanism involving increased potassium electrolyte balance. Recent ndings that the RAS is activated locally
(K) conductance resulting in a reduction in intracellular K and in the heart and vessel wall have expanded that view and it is now
the subsequent depolarization of the cell and relaxation. Molecules appreciated that it plays a critical role in the initiation and pro-
and mediators postulated to act as EDHF include K [168], cyto- gression of atherosclerosis [190]. Indeed, with the exception of
chrome P450 metabolites [169], lipoxygenase products [170], renin (which cleaves angiotensinogen to form angiotensin I), all
reactive oxygen species (H2O2) [171], C-type natriuretic peptide components of the RAS are produced in the vasculature. Endothe-
[172], and electrical coupling through myoendothelial gap junc- lial cells express angiotensin converting enzyme (ACE), the dipep-
tions [173]. tidyl carboxypeptidase that converts angiotensin-1 to the
physiologically active angiotensin-II (Ang-II) [191]. Both local and
11. Vasoconstrictors circulating Ang-II exert their activities through the binding to Ang-
II type 1 (AT1) or type 2 (AT2) receptors. AT1 receptors are widely
Opposing the action of the endothelial-derived vasodilators expressed on different cell types involved in atherogenesis and
(NO, PGI2, EDHFs) are a number of endothelial-derived vasocon- mediate most of the physiological and pathophysiological actions
strictors, chief among them endothelin-1 (ET-1), angiotensin-II of Ang-II. A role for the AT2 receptor in cardiovascular disease re-
(ANG-II) and vasoconstrictor prostaglandins. mains controversial (for review [192]). Ang-II is a proathero-
sclerotic mediator; it causes vasoconstriction and regulates not
11.1. ET-1 only adhesion molecule (VCAM-1, ICAM-1, P-selectin) expression
but also cytokine, chemokine, and growth factor secretion within
In 1988, Dr. Yanagisawa's group identied endothelin-1 (ET-1), a the arterial wall [193]. ACE inhibitors and angiotensin receptor
21 amino acid peptide generated by the endothelin converting blockers (ARBs) are established pharmacological tools in both pri-
enzyme (ECE)-mediated cleavage of a larger precursor big endo- mary and secondary prevention of atherosclerotic cardiovascular
thelin-1 (Big ET-1), as the rst endothelium-derived contracting disease [194]. The endothelium is the main target organ for many
factor [174]. ET-1 is one of the most potent endogenous vasocon- cardiovascular drugs including ACE inhibitors, and evidence shows
strictors (for review [175]. It is produced mainly by endothelial that their benecial effects are not only due to blood pressure
cells, but also by vascular smooth muscle cells, leukocytes and lowering but also due to a direct anti-inammatory activity [194].
macrophages [175]. ET-1 and NO are functionally closely interde-
pendent [176]; NO strongly inhibits the release of ET-1 from the 12. Antithrombotic/prothrombotic balance
endothelium and ET-1 strongly inhibits NO-mediated vasodilation
and vice versa [175,177]. There are 3 isoforms of endothelin: ET-1, The endothelium crucially maintains the lining of the blood
-2 and -3, with ET-1 most relevant in the vasculature and in vessel as nonthrombogenic [195]. An important pathophysiological
contributing to the maintenance of vascular tone [178]. At elevated consequence of endothelial dysfunction is a disturbed hemostatic
concentrations, associated with pathological conditions, ET-1 is balance of thrombotic and brinolytic states leading to altered
also pro-inammatory and promotes smooth muscle proliferation blood uidity and possible pathologic hemorrhage or clot forma-
[179,180]. Elevated levels of ET-1 have been implicated in the tion at sites of vascular injury. In regard to its physiological active
pathogenesis of hypertension, vasospasm and heart failure, in anti-thrombotic role, endothelium associated factors such as the
addition to atherosclerosis. Increased expression of ET-1 and ECE is potent inhibitors of platelet aggregation PGI2 and NO, as well as
seen in human arteries at different stages of atherosclerosis [181]. heparin-like molecules, thrombomodulin, Ecto-ADPase, tissue
The biological effects of ET-1 are mediated through activation of plasminogen activator and urokinase all may contribute [196]. The
two receptors, ETA and ETB [182,183]. ETA and ETB receptors endothelium is also capable of active pro-thrombotic behavior; it
expressed on vascular smooth muscle cells mediate the vasocon- synthesizes thromboxane A2 (TxA2) which promotes platelet ag-
strictor effects of ET-1. The ETB receptor is also found on endothelial gregation, it expresses adhesive co-factors for platelets such as von
cells, where its activation results in vasodilation mediated by PGI2 Willebrand factor, bronectin and thrombospondin, and procoa-
and NO release [184] (Fig. 4). ETB also plays a role in the clearance of gulant factors such as factor V [197]. Moreover, the endothelium
circulating ET-1 mediated by ligand-receptor complex internaliza- can be activated by various pathophysiologic stimuli to express
tion and intracellular degradation. Therefore, a reduction in ETB tissue factor, a trigger for the brin-generating coagulation cascade
number, or ETB receptor blockade, might reduce ET-1 clearance, [198,199]. Endothelial cells can also generate an inhibitor of the
increasing ET-1 concentrations without altering production. brinolytic pathway (plasminogen activator inibiyor-1 (PAI-1),
104 P.A. Cahill, E.M. Redmond / Atherosclerosis 248 (2016) 97e109

Fig. 4. Big endothelin-1 (inactive precursor peptide) is cleaved by endothelin converting enzyme (ECE) to form Endothelin-1 (ET-1). Endothelin receptors ETA and ETB are expressed
on vascular smooth muscle cells (SMC) and mediate vasoconstriction. ETB is also expressed on endothelial cells (EC) where its activation can result in vasodilation mediated by nitric
oxide (NO) and prostacyclin (PGI2) release.

which can reduce the rate of brin breakdown [200]. Thus, in [212]. Decreased EPC numbers have also been associated with
physiologic conditions, the normal endothelium actively supports metabolic syndrome, which confers a high risk for cardiovascular
the uid state of owing blood and prevents activation of circu- disease [213]. Given the critical role of the endothelium in vascular
lating cells, whereas injury to the endothelium triggers a series of health and the importance of maintaining a healthy endothelium
biochemical and molecular reactions favoring blood arrest and as opposed to a dysfunctional one, clinical benet may be derived
vessel wall repair. In the latter case (i.e., dysfunctional endothe- from harnessing EPC that can differentiate into mature endothelial
lium), platelets may aggregate and stick to the endothelium thereby cells to use as therapy for vascular disease. With this in mind,
activating it and encouraging leukocyte adhesion, as well as strategies that have been employed to enhance EPC number and
releasing platelet-derived growth factors that stimulate intimal functionality include statins, ACE inhibitors, angiotensin receptor
hyperplasia [201,202]. blockers and peroxisome proliferator-activated receptor gamma
(PPAR-g) agonists, although the mechanisms involved remain un-
13. Endothelial progenitor cells clear [213]. Other clinical approaches involve providing exogenous
EPC to the circulation to replenish numbers reduced in diseased
Evidence suggests that the injured endothelial monolayer may patients [214]. Finally, the discovery of resident stem/progenitor
be regenerated by circulating bone marrow-derived endothelial cells in the vessel wall that can differentiate into all types of
progenitor cells (EPC), which accelerate re-endothelialization and vascular cells [215], and evidence that endothelial cells may
protect against the initiation and progression of atherosclerosis transform into smooth muscle-like cells [216], has complicated the
[203e205,206]. EPC represent less than 1% of the cells in blood. vascular progenitor cell research eld even further.
They are generally puried from blood by density gradient centri-
fugation for mononuclear cells followed by attachment onto tissue 14. MicroRNAs (miRNAs)
culture plates, and characterized at early stages after attachment by
positive expression for both an endothelial marker such as VEGFR2 Discovered in 1993, microRNAs (miRNAs) are short (19e25
and for hematopoietic stem cell markers CD133 and CD34. After nucleotides long) sequences of non-coding RNA that regulate genes
expansion for several weeks they express endothelial markers at the post-transcriptional level. It is estimated that there may be
including vascular endothelial (VE)-Cadherin and Von Williebrand up to 5000 miRs that nely tune the expression of a multitude of
factor (vWF) [207]. It should be noted, however, that controversy protein-coding genes [217], including those involved in signaling
exists with respect to the identication and characterization of pathways controlling proliferation, differentiation and apoptosis. It
EPC which may actually encompass several different populations, is hardly surprising then, that a burgeoning role for miRNAs in
as well as with regard to their denitive role in the pathophysiology endothelial dysfunction and atherogenesis has been recognized.
of atherosclerosis vs in angiogenesis [208,209]. Insufcient blood Indeed, an expanding body of literature indicates that miRNAs
ow resulting in local tissue ischemia is recognized as a potent affect nearly every aspect of atherosclerotic plaque initiation,
stimulus for recruitment of EPC [206]; EPC express chemokine re- development and progression including inammation/chemo-
ceptor type 4 (CXCR-4) receptors that bind stromal-derived factor-1 kines, cholesterol homeostasis, cell adhesion, vascular cell prolif-
(SDF-1) that is released at sites of ischemia [210]. Higher circulating eration/apoptosis, and EPC function (recently reviewed in
levels of progenitor cells reect greater repair capacity and have Ref. [218,219]). For example, recent studies report that miR-24
been shown to reduce the progression of atherosclerosis [211]. Age targeting matrix metalloproteinase-14 (MMP-14) regulates mac-
is a signicant risk factor for the development of cardiovascular rophages [220], miR-126-5p promotes endothelial proliferation by
disease and interestingly, EPCs are subject to age-associated suppressing Notch1 inhibitor delta-like 1 homolog (Dlk1) [221],
changes that diminish their number in circulation and function miR-126 also inhibits VCAM-1 expression and leukocyte adhesion
P.A. Cahill, E.M. Redmond / Atherosclerosis 248 (2016) 97e109 105

to endothelial cells [222], miR-145 regulates SMC differentiation 16. Summary


and promotes plaque stability [223], miR-155 reduces inamma-
tory responses and enhances macrophage cholesterol efux [224], The endothelium is critical in maintaining physiological balance
and miR-302a modulates cholesterol homeostasis by affecting the in the vasculature and may be considered the gatekeeper of vessel
ATP-binding cassette (ABC) transporter ABCA1 [225]. MicroRNAs health. Endothelial cells synthesize and release many factors that
are actively secreted by endothelial cells under physiological and locally regulate permeability, vascular tone, smooth muscle cell
pathophysiological conditions and can be quantied in blood. growth and migration, inammatory responses and platelet func-
These circulating miRNA, which are very stable, are being viewed as tion. Upsetting this tightly regulated balance, such as occurs when
promising biomarkers for a number of cardiovascular diseases the endothelium becomes dysfunctional in response to a variety of
[226]. Moreover, ow-sensitive miRNAs known as mechano-miRs different risk factors, leads to the initiation and development of
have also been identied in endothelial cells [227]. These include atherosclerotic lesions. The endothelium is, thus, an important
miR-10a, miR-23b, miR-21, miR-663, miR-92a, miR-143/145, miR- target for therapies, both current and in the development phase, for
101 and miR-126; all of which regulate EC gene expression, cardiovascular disease.
affecting EC health or dysfuction and thus, atherogenesis. Given the
important role of gene regulating miRs in endothelial function Conicts of interest
many are currently being considered as potential therapeutic tar-
gets to treat atherosclerosis. Therapies might consist of either Authors Profs Paul A Cahill and Eileen M Redmond, submitting
introducing atheroprotective miRs, or depleting atherogenic the review titled Vascular EndotheliumeGatekeeper of Vessel
miRs using anti-sense miRNAs. Health declare no conicts of interest.

15. Endothelial-mesenchymal transition (EndoMT) References

Vascular endothelial cells can demonstrate plasticity in both [1] R. Ross, Atherosclerosis-an inammatory disease, N. Engl. J. Med. 340 (1999)
developmental and pathological conditions. Generation of mesen- 115e126.
[2] J. Mestas, K. Ley, Monocyte-endothelial cell interactions in the development
chymal cells from endothelium, known as endothelial- of atherosclerosis, Trends Cardiovasc. Med. 18 (2008) 228e232.
mesenchymal transition (EndoMT), is a crucial step in endothelial [3] P. Libby, P.M. Ridker, G.K. Hansson, Progress and challenges in translating the
cell differentiation to several lineages including broblasts, mural biology of atherosclerosis, Nature 473 (2011) 317e325.
[4] C. Blasi, The autoimmune origin of atherosclerosis, Atherosclerosis 201
cells and adipocytes. EndoMT has been implicated in vascular
(2008) 17e32.
remodeling associated with vein grafting, arteriosclerosis and [5] D.W. Morel, P.E. DiCorleto, G.M. Chisolm, Endothelial and smooth muscle
atherosclerosis [228]. TGF-b has been identied as key in driving cells alter low density lipoprotein in vitro by free radical oxidation, Arte-
EndMT progression [229]. Murine cell lineageetracing experiments riosclerosis 4 (1984) 357e364.
[6] U.P. Steinbrecher, S. Parthasarathy, D.S. Leake, et al., Modication of low
showed that endothelial-derived cells contribute to neointimal density lipoprotein by endothelial cells involves lipid peroxidation and
formation through EndoMT following vein grafting which is degradation of low density lipoprotein phospholipids, Proc. Natl. Acad. Sci. U.
dependent on early activation of the TGF-b Smad2/3-Slug signaling S. A. 81 (1984) 3883e3887.
[7] M.K. Cathcart, D.W. Morel, G.M. Chisolm 3rd, Monocytes and neutrophils
pathway [230]. Antagonism of TGF-b signaling decreased EndoMT oxidize low density lipoprotein making it cytotoxic, J. Leukoc. Biol. 38 (1985)
and resulted in less neointimal formation when compared to con- 341e350.
trols. Histological examination of human vein grafts validated the [8] S. Parthasarathy, D.J. Printz, D. Boyd, et al., Macrophage oxidation of low
density lipoprotein generates a modied form recognized by the scavenger
murine data. Furthermore, as caveolin-1 is important for TGF-b receptor, Arteriosclerosis 6 (1986) 505e510.
receptor internalization and TGF-b signaling, it also appears critical [9] S. Mitra, T. Goyal, J.L. Mehta, Oxidized LDL, LOX-1 and atherosclerosis, Car-
for regulation of EndoMT [231]. EndoMT may be a link between diovasc. Drugs Ther. Int. Soc. Cardiovasc. Pharmacother. 25 (2011) 419e429.
[10] O. Ogunrinade, G.T. Kameya, G.A. Truskey, Effect of uid shear stress on the
factors initiating atherosclerosis, such as inammation and
permeability of the arterial endothelium, Ann. Biomed. Eng. 30 (2002)
disturbed ow, and the tissue remodeling that leads to plaque 430e446.
formation [232]. It appears that oscillatory shear stress reduces [11] C.C. Michel, F.E. Curry, Microvascular permeability, Physiol. Rev. 79 (1999)
703e761.
broblast growth factor receptor 1 (FGFR1) expression and FGF
[12] H. Lum, A.B. Malik, Mechanisms of increased endothelial permeability, Can. J.
signaling, resulting in activation of endothelial TGF-b signaling and Physiol. Pharmacol. 74 (1996) 787e800.
induction of EndoMT. Chen et al., demonstrated that ApoE / [13] R.G. Tilton, K.C. Chang, W.S. LeJeune, et al., Role for nitric oxide in the
mice with disrupted protective FGF endothelial signaling as a hyperpermeability and hemodynamic changes induced by intravenous
VEGF, Investig. Ophthalmol. Vis. Sci. 40 (1999) 689e696.
result of cell-specic deletion of FGF receptor substrate 2a exhibi- [14] K. Ashina, Y. Tsubosaka, K. Kobayashi, et al., VEGF-induced blood ow in-
ted extensive development of EndMT and increased neointima crease causes vascular hyper-permeability in vivo, Biochem. Biophy. Res.
formation [232]. Moreover, in human patients the level of coronary Commun. 464 (2015) 590e595.
[15] B.W. Wong, M. Rahmani, Z. Luo, et al., Vascular endothelial growth factor
artery disease correlated strongly with loss of endothelial FGFR1 increases human cardiac microvascular endothelial cell permeability to low-
expression, activation of endothelial TGF-b signaling, and the density lipoproteins, J. Heart Lung Transpl. 28 (2009) 950e957.
extent of EndoMT [232]. EndoMT is thought to promote plaque [16] D.O. Bates, Vascular endothelial growth factors and vascular permeability,
Cardiovasc. Res. 87 (2010) 262e271.
growth by increasing deposition of bronectin and increasing [17] A.D. Joshi, C. Dimitropoulou, G. Thangjam, et al., Heat shock protein 90 in-
expression of cell adhesion molecules ICAM and VCAM, thereby hibitors prevent LPS-induced endothelial barrier dysfunction by disrupting
further promoting recruitment of circulating monocytes and leu- RhoA signaling, Am. J. Respir. Cell Mol. Biol. 50 (2014) 170e179.
[18] J.M. Tarbell, Shear stress and the endothelial transport barrier, Cardiovasc.
kocytes and inducing formation of new mesenchymal cells, leading Res. 87 (2010) 320e330.
to expansion of neointima. In a separate study, Moonen et al., re- [19] J.A. LaMack, H.A. Himburg, X.M. Li, et al., Interaction of wall shear stress
ported that while steady ow inhibited EndoMT, endothelial cells magnitude and gradient in the prediction of arterial macromolecular
permeability, Ann. Biomed. Eng. 33 (2005) 457e464.
exposed to disturbed ow underwent EndoMT and exhibited
[20] J.C. Rutledge, F.R. Curry, J.F. Lenz, et al., Low density lipoprotein transport
atherogenic differentiation. Gain- and loss-of-function studies across a microvascular endothelial barrier after permeability is increased,
established a pivotal role for ERK5 signaling in the inhibition of Circ. Res. 66 (1990) 486e495.
EndoMT [233]. Collectively, these data suggest that inhibition of [21] J.V. Soulis, D.K. Fytanidis, V.C. Papaioannou, et al., Wall shear stress on LDL
accumulation in human RCAs, Med. Eng. Phys. 32 (2010) 867e877.
EndoMT might prove to be a novel therapeutic strategy to treat [22] B.C. Berk, W. Min, C. Yan, et al., Atheroprotective mechanisms activated by
atherosclerosis. uid shear stress in endothelial cells, Drug News Perspect. 15 (2002)
106 P.A. Cahill, E.M. Redmond / Atherosclerosis 248 (2016) 97e109

133e139. endothelial glycocalyx integrity during coronary artery bypass grafting,


[23] K. Skalen, M. Gustafsson, E.K. Rydberg, et al., Subendothelial retention of Scand. Cardiovasc. J. 46 (2012) 339e344.
atherogenic lipoproteins in early atherosclerosis, Nature 417 (2002) [51] L.N. Broekhuizen, B.A. Lemkes, H.L. Mooij, et al., Effect of sulodexide on
750e754. endothelial glycocalyx and vascular permeability in patients with type 2
[24] L. Badimon, R.F. Storey, G. Vilahur, Update on lipids, inammation and diabetes mellitus, Diabetologia 53 (2010) 2646e2655.
atherothrombosis, Thromb. Haemost. 105 (Suppl 1) (2011) S34eS42. [52] P.F. Davies, Flow-mediated endothelial mechanotransduction, Physiol. Rev.
[25] I. Rozenberg, S.H. Sluka, L. Rohrer, et al., Histamine H1 receptor promotes 75 (1995) 519e560.
atherosclerotic lesion formation by increasing vascular permeability for low- [53] J. Ando, K. Yamamoto, Effects of shear stress and stretch on endothelial
density lipoproteins, Arterioscler. Thromb. Vasc. Biol. 30 (2010) 923e930. function, Antioxid. Redox Signal 15 (2011) 1389e1403.
[26] A.E. Mullick, J.M. McDonald, G. Melkonian, et al., Reactive carbonyls from [54] M. Ohno, G.H. Gibbons, V.J. Dzau, et al., Shear stress elevates endothelial
tobacco smoke increase arterial endothelial layer injury, Am. J. Physiol. Heart cGMP. Role of a potassium channel and G protein coupling, Circulation 88
Circ. Physiol. 283 (2002) H591eH597. (1993) 193e197.
[27] V. Kundumani-Sridharan, E. Dyukova, D.E. Hansen 3rd, et al., 12/15- [55] A.R. Koslow, R.R. Stromberg, L.I. Friedman, et al., A ow system for the study
Lipoxygenase mediates high-fat diet-induced endothelial tight junction of shear forces upon cultured endothelial cells, J. Biomech. Eng. 108 (1986)
disruption and monocyte transmigration: a new role for 15(S)-hydrox- 338e341.
yeicosatetraenoic acid in endothelial cell dysfunction, J. Biol. Chem. 288 [56] E.M. Redmond, P.A. Cahill, J.V. Sitzmann, Perfused transcapillary smooth
(2013) 15830e15842. muscle and endothelial cell co-cultureea novel in vitro model, In Vitro Cell
[28] J.J. Whyte, M.H. Laughlin, The effects of acute and chronic exercise on the Dev. Biol. Anim. 31 (1995) 601e609.
vasculature, Acta Physiol. Oxf. 199 (2010) 441e450. [57] E.M. Redmond, P.A. Cahill, J.V. Sitzmann, Flow-mediated regulation of G-
[29] J.G. Garcia, F. Liu, A.D. Verin, et al., Sphingosine 1-phosphate promotes protein expression in cocultured vascular smooth muscle and endothelial
endothelial cell barrier integrity by Edg-dependent cytoskeletal rearrange- cells, Arterioscler. Thromb. Vasc. Biol. 18 (1998) 75e83.
ment, J. Clin. Investig. 108 (2001) 689e701. [58] K. Kanda, T. Matsuda, Behavior of arterial wall cells cultured on periodically
[30] Y. Xiong, T. Hla, S1P control of endothelial integrity, Curr. Top. Microbiol. stretched substrates, Cell Transplant. 2 (1993) 475e484.
Immunol. 378 (2014) 85e105. [59] D. Morrow, J.P. Cullen, P.A. Cahill, et al., Cyclic strain regulates the Notch/CBF-
[31] K.M. Argraves, P.J. Gazzolo, E.M. Groh, et al., High density lipoprotein- 1 signaling pathway in endothelial cells: role in angiogenic activity, Arte-
associated sphingosine 1-phosphate promotes endothelial barrier function, rioscler. Thromb. Vasc. Biol. 27 (2007) 1289e1296.
J. Biol. Chem. 283 (2008) 25074e25081. [60] H.J. Hsieh, C.A. Liu, B. Huang, et al., Shear-induced endothelial mechano-
[32] B.A. Wilkerson, G.D. Grass, S.B. Wing, et al., Sphingosine 1-phosphate (S1P) transduction: the interplay between reactive oxygen species (ROS) and nitric
carrier-dependent regulation of endothelial barrier: high density lipoprotein oxide (NO) and the pathophysiological implications, J. Biomed. Sci. 21 (2014)
(HDL)-S1P prolongs endothelial barrier enhancement as compared with 3.
albumin-S1P via effects on levels, trafcking, and signaling of S1P1, J. Biol. [61] N. Resnick, T. Collins, W. Atkinson, et al., Platelet-derived growth factor B
Chem. 287 (2012) 44645e44653. chain promoter contains a cis-acting uid shear-stress-responsive element,
[33] L. Pizurki, Z. Zhou, K. Glynos, et al., Angiopoietin-1 inhibits endothelial Proc. Natl. Acad. Sci. U. S. A. 90 (1993) 4591e4595.
permeability, neutrophil adherence and IL-8 production, Br. J. Pharmacol. [62] N. Baeyens, M.J. Mulligan-Kehoe, F. Corti, et al., Syndecan 4 is required for
139 (2003) 329e336. endothelial alignment in ow and atheroprotective signaling, Proc. Natl.
[34] E.G. Langeler, V.W. van Hinsbergh, Norepinephrine and iloprost improve Acad. Sci. U. S. A. 111 (2014) 17308e17313.
barrier function of human endothelial cell monolayers: role of cAMP, Am. J. [63] Y. Liu, C. Collins, W.B. Kiosses, et al., A novel pathway spatiotemporally ac-
Physiol. 260 (1991) C1052eC1059. tivates Rac1 and redox signaling in response to uid shear stress, J. Cell Biol.
[35] J. Surapisitchat, J.A. Beavo, Regulation of endothelial barrier function by cy- 201 (2013) 863e873.
clic nucleotides: the role of phosphodiesterases, Handb. Exp. Pharmacol. [64] J. Li, B. Hou, D.J. Beech, Endothelial Piezo1: life depends on it, Channels
(2011) 193e210. Austin 9 (2015) 1e2.
[36] D. Chappell, M. Jacob, O. Paul, et al., The glycocalyx of the human umbilical [65] S.S. Ranade, Z. Qiu, S.H. Woo, et al., Piezo1, a mechanically activated ion
vein endothelial cell: an impressive structure ex vivo but not in culture, Circ. channel, is required for vascular development in mice, Proc. Natl. Acad. Sci.
Res. 104 (2009) 1313e1317. U. S. A. 111 (2014) 10347e10352.
[37] C.S. Alphonsus, R.N. Rodseth, The endothelial glycocalyx: a review of the [66] B. Jung, H. Obinata, S. Galvani, et al., Flow-regulated endothelial S1P
vascular barrier, Anaesthesia 69 (2014) 777e784. receptor-1 signaling sustains vascular development, Dev. Cell 23 (2012)
[38] M. Rehm, D. Bruegger, F. Christ, et al., Shedding of the endothelial glycocalyx 600e610.
in patients undergoing major vascular surgery with global and regional [67] J.P. Cullen, S.M. Nicholl, S. Sayeed, et al., Plasminogen activator inhibitor-1
ischemia, Circulation 116 (2007) 1896e1906. deciency enhances ow-induced smooth muscle cell migration, Thromb.
[39] A. Beresewicz, E. Czarnowska, M. Maczewski, Ischemic preconditioning and Res. 114 (2004) 57e65.
superoxide dismutase protect against endothelial dysfunction and endo- [68] M.A. Gimbrone Jr., J.N. Topper, T. Nagel, et al., Endothelial dysfunction, he-
thelium glycocalyx disruption in the postischemic guinea-pig hearts, Mol. modynamic forces, and atherogenesis, Ann. N. Y. Acad. Sci. 902 (2000)
Cell. Biochem. 186 (1998) 87e97. 230e239 discussion 239e240.
[40] A.W. Mulivor, H.H. Lipowsky, Inammation- and ischemia-induced shedding [69] S. Pradhan, B. Sumpio, Molecular and biological effects of hemodynamics on
of venular glycocalyx, Am. J. Physiol. Heart Circ. Physiol. 286 (2004) vascular cells, Front. Biosci. J. Virtual Libr. 9 (2004) 3276e3285.
H1672eH1680. [70] J.J. Chiu, S. Chien, Effects of disturbed ow on vascular endothelium: path-
[41] C.B. Henry, B.R. Duling, TNF-alpha increases entry of macromolecules into ophysiological basis and clinical perspectives, Physiol. Rev. 91 (2011)
luminal endothelial cell glycocalyx, Am. J. Physiol. Heart Circ. Physiol. 279 327e387.
(2000) H2815eH2823. [71] K.S. Heo, K. Fujiwara, J. Abe, Shear stress and atherosclerosis, Mol. Cells 37
[42] B.F. Becker, D. Chappell, D. Bruegger, et al., Therapeutic strategies targeting (2014) 435e440.
the endothelial glycocalyx: acute decits, but great potential, Cardiovasc. [72] P.H. Stone, A.U. Coskun, S. Kinlay, et al., Effect of endothelial shear stress on
Res. 87 (2010) 300e310. the progression of coronary artery disease, vascular remodeling, and in-stent
[43] B.M. van den Berg, J.A. Spaan, T.M. Rolf, et al., Atherogenic region and diet restenosis in humans: in vivo 6-month follow-up study, Circulation 108
diminish glycocalyx dimension and increase intima-to-media ratios at mu- (2003) 438e444.
rine carotid artery bifurcation, Am. J. Physiol. Heart Circ. Physiol. 290 (2006) [73] A.R. Brooks, P.I. Lelkes, G.M. Rubanyi, Gene expression proling of human
H915eH920. aortic endothelial cells exposed to disturbed ow and steady laminar ow,
[44] M. Gouverneur, B. Berg, M. Nieuwdorp, et al., Vasculoprotective properties of Physiol. Genom. 9 (2002) 27e41.
the endothelial glycocalyx: effects of uid shear stress, J. Intern. Med. 259 [74] J. Dunn, S. Thabet, H. Jo, Flow-dependent epigenetic DNA methylation in
(2006) 393e400. endothelial gene expression and atherosclerosis, Arterioscler. Thromb. Vasc.
[45] V.H. Huxley, D.A. Williams, Role of a glycocalyx on coronary arteriole Biol. 35 (2015) 1562e1569.
permeability to proteins: evidence from enzyme treatments, Am. J. Physiol. [75] Y.Z. Jiang, J.M. Jimenez, K. Ou, et al., Hemodynamic disturbed ow induces
Heart Circ. Physiol. 278 (2000) H1177eH1185. differential DNA methylation of endothelial Kruppel-Like Factor 4 promoter
[46] A.A. Constantinescu, H. Vink, J.A. Spaan, Endothelial cell glycocalyx modu- in vitro and in vivo, Circ. Res. 115 (2014) 32e43.
lates immobilization of leukocytes at the endothelial surface, Arterioscler. [76] B.L. Langille, S.L. Adamson, Relationship between blood ow direction and
Thromb. Vasc. Biol. 23 (2003) 1541e1547. endothelial cell orientation at arterial branch sites in rabbits and mice, Circ.
[47] S. Mochizuki, H. Vink, O. Hiramatsu, et al., Role of hyaluronic acid glycos- Res. 48 (1981) 481e488.
aminoglycans in shear-induced endothelium-derived nitric oxide release, [77] S. Akimoto, M. Mitsumata, T. Sasaguri, et al., Laminar shear stress inhibits
Am. J. Physiol. Heart Circ. Physiol. 285 (2003) H722eH726. vascular endothelial cell proliferation by inducing cyclin-dependent kinase
[48] R. Kelly, T. Ruane-O'Hora, M.I. Noble, et al., Differential inhibition by inhibitor p21(Sdi1/Cip1/Waf1), Circ. Res. 86 (2000) 185e190.
hyperglycaemia of shear stress- but not acetylcholine-mediated dilatation in [78] P.F. Davies, A. Remuzzi, E.J. Gordon, et al., Turbulent uid shear stress in-
the iliac artery of the anaesthetized pig, J. Physiol. 573 (2006) 133e145. duces vascular endothelial cell turnover in vitro, Proc. Natl. Acad. Sci. U. S. A.
[49] A.J. Drake-Holland, M.I. Noble, The important new drug target in cardio- 83 (1986) 2114e2117.
vascular medicineethe vascular glycocalyx, Cardiovasc. Hematol. Disord. [79] N. Von Offenberg Sweeney, P.M. Cummins, E.J. Cotter, et al., Cyclic strain-
Drug Targets 9 (2009) 118e123. mediated regulation of vascular endothelial cell migration and tube forma-
[50] A.A. Mennander, A. Shalaby, N. Oksala, et al., Diazoxide may protect tion, Biochem. Biophy. Res. Commun. 329 (2005) 573e582.
P.A. Cahill, E.M. Redmond / Atherosclerosis 248 (2016) 97e109 107

[80] G.M. Rubanyi, J.C. Romero, P.M. Vanhoutte, Flow-induced release of endothelium and their roles in the regulation of endothelial function,
endothelium-derived relaxing factor, Am. J. Physiol. 250 (1986) J. Cardiovasc. Pharmacol. 61 (2013) 461e470.
H1145eH1149. [110] B. Kou, J. Zhang, D.R. Singer, Effects of cyclic strain on endothelial cell
[81] A. Bhagyalakshmi, J.A. Frangos, Mechanism of shear-induced prostacyclin apoptosis and tubulogenesis are dependent on ROS production via NAD(P)H
production in endothelial cells, Biochem. Biophy. Res. Commun. 158 (1989) subunit p22phox, Microvasc. Res. 77 (2009) 125e133.
31e37. [111] F.A. Martin, A. McLoughlin, K.D. Rochfort, et al., Regulation of thrombomo-
[82] R.J. Hendrickson, C. Cappadona, E.N. Yankah, et al., Sustained pulsatile ow dulin expression and release in human aortic endothelial cells by cyclic
regulates endothelial nitric oxide synthase and cyclooxygenase expression in strain, PLoS One 9 (2014) e108254.
co-cultured vascular endothelial and smooth muscle cells, J. Mol. Cell Cardiol. [112] Y. Jiang, K. Kohara, K. Hiwada, Association between risk factors for athero-
31 (1999) 619e629. sclerosis and mechanical forces in carotid artery, Stroke J. Cereb. Circ. 31
[83] M.J. Kuchan, J.A. Frangos, Shear stress regulates endothelin-1 release via (2000) 2319e2324.
protein kinase C and cGMP in cultured endothelial cells, Am. J. Physiol. 264 [113] J. Szostak, P. Laurant, The forgotten face of regular physical exercise: a
(1993) H150eH156. 'natural' anti-atherogenic activity, Clin. Sci. 121 (2011) 91e106.
[84] A.M. Malek, A.L. Greene, S. Izumo, Regulation of endothelin 1 gene by uid [114] J.R. Santos-Parker, T.J. LaRocca, D.R. Seals, Aerobic exercise and other healthy
shear stress is transcriptionally mediated and independent of protein kinase lifestyle factors that inuence vascular aging, Adv. Physiol. Educ. 38 (2014)
C and cAMP, Proc. Natl. Acad. Sci. U. S. A. 90 (1993) 5999e6003. 296e307.
[85] U.C. Garg, A. Hassid, Nitric oxide-generating vasodilators and 8-bromo-cyclic [115] M. Sandrock, C. Schulze, D. Schmitz, et al., Physical activity throughout life
guanosine monophosphate inhibit mitogenesis and proliferation of cultured reduces the atherosclerotic wall process in the carotid artery, Br. J. Sports
rat vascular smooth muscle cells, J. Clin. Investig. 83 (1989) 1774e1777. Med. 42 (2008) 839e844.
[86] G.F. Alberts, K.A. Peiey, A. Johns, et al., Constitutive endothelin-1 over- [116] J. Niebauer, R. Hambrecht, T. Velich, et al., Attenuated progression of coro-
expression promotes smooth muscle cell proliferation via an external nary artery disease after 6 years of multifactorial risk intervention: role of
autocrine loop, J. Biol. Chem. 269 (1994) 10112e10118. physical exercise, Circulation 96 (1997) 2534e2541.
[87] M. Kohno, K. Yokokawa, K. Yasunari, et al., Effect of the endothelin family of [117] G. Schuler, R. Hambrecht, G. Schlierf, et al., Regular physical exercise and
peptides on human coronary artery smooth-muscle cell migration, low-fat diet. Effects on progression of coronary artery disease, Circulation 86
J. Cardiovasc. Pharmacol. 31 (Suppl. 1) (1998) S84eS89. (1992) 1e11.
[88] L.J. Ignarro, Endothelium-derived nitric oxide: actions and properties, FASEB [118] M.H. Laughlin, S.C. Newcomer, S.B. Bender, Importance of hemodynamic
J. Off. Publ. Fed. Am. Soc. Exp. Biol. 3 (1989) 31e36. forces as signals for exercise-induced changes in endothelial cell phenotype,
[89] S. Moncada, Prostacyclin and arterial wall biology, Arteriosclerosis 2 (1982) J. Appl. Physiol. 104 (2008) 588e600.
193e207. [119] B. Ramkhelawon, J. Vilar, D. Rivas, et al., Shear stress regulates angiotensin
[90] Y. Takada, F. Shinkai, S. Kondo, et al., Fluid shear stress increases the type 1 receptor expression in endothelial cells, Circ. Res. 105 (2009)
expression of thrombomodulin by cultured human endothelial cells, Bio- 869e875.
chem. Biophy. Res. Commun. 205 (1994) 1345e1352. [120] P. Libby, Inammation in atherosclerosis, Nature 420 (2002) 868e874.
[91] T. Arisaka, M. Mitsumata, M. Kawasumi, et al., Effects of shear stress on [121] T.S. Church, C.E. Barlow, C.P. Earnest, et al., Associations between cardiore-
glycosaminoglycan synthesis in vascular endothelial cells, Ann. N. Y. Acad. spiratory tness and C-reactive protein in men, Arterioscler. Thromb. Vasc.
Sci. 748 (1995) 543e554. Biol. 22 (2002) 1869e1876.
[92] S.L. Diamond, S.G. Eskin, L.V. McIntire, Fluid ow stimulates tissue plas- [122] N. Kondo, M. Nomura, Y. Nakaya, et al., Association of inammatory marker
minogen activator secretion by cultured human endothelial cells, Science and highly sensitive C-reactive protein with aerobic exercise capacity,
243 (1989) 1483e1485. maximum oxygen uptake and insulin resistance in healthy middle-aged
[93] G.K. Owens, M.S. Kumar, B.R. Wamhoff, Molecular regulation of vascular volunteers, Circ. J. 69 (2005) 452e457.
smooth muscle cell differentiation in development and disease, Physiol. Rev. [123] S. Mora, N. Cook, J.E. Buring, et al., Physical activity and reduced risk of
84 (2004) 767e801. cardiovascular events: potential mediating mechanisms, Circulation 116
[94] M. Mitsumata, R.S. Fishel, R.M. Nerem, et al., Fluid shear stress stimulates (2007) 2110e2118.
platelet-derived growth factor expression in endothelial cells, Am. J. Physiol. [124] G.O. Gjevestad, K.B. Holven, S.M. Ulven, Effects of exercise on gene expres-
265 (1993) H3eH8. sion of inammatory markers in human peripheral blood cells: a systematic
[95] A.M. Malek, G.H. Gibbons, V.J. Dzau, et al., Fluid shear stress differentially review, Curr. Cardiovasc. Risk Rep. 9 (2015) 34.
modulates expression of genes encoding basic broblast growth factor and [125] H. Palmefors, S. DuttaRoy, B. Rundqvist, et al., The effect of physical activity
platelet-derived growth factor B chain in vascular endothelium, J. Clin. or exercise on key biomarkers in atherosclerosisea systematic review,
Investig. 92 (1993) 2013e2021. Atherosclerosis 235 (2014) 150e161.
[96] M. Ohno, J.P. Cooke, V.J. Dzau, et al., Fluid shear stress induces endothelial [126] R.F. Furchgott, J.V. Zawadzki, The obligatory role of endothelial cells in the
transforming growth factor beta-1 transcription and production. Modulation relaxation of arterial smooth muscle by acetylcholine, Nature 288 (1980)
by potassium channel blockade, J. Clin. Investig. 95 (1995) 1363e1369. 373e376.
[97] A.V. Sterpetti, A. Cucina, A.R. Morena, et al., Shear stress increases the release [127] L.J. Ignarro, R.E. Byrns, G.M. Buga, et al., Endothelium-derived relaxing factor
of interleukin-1 and interleukin-6 by aortic endothelial cells, Surgery 114 from pulmonary artery and vein possesses pharmacologic and chemical
(1993) 911e914. properties identical to those of nitric oxide radical, Circ. Res. 61 (1987)
[98] M. Merten, T. Chow, J.D. Hellums, et al., A new role for P-selectin in shear- 866e879.
induced platelet aggregation, Circulation 102 (2000) 2045e2050. [128] R.F. Furchgott, P.M. Vanhoutte, Endothelium-derived relaxing and con-
[99] J. Ando, H. Tsuboi, R. Korenaga, et al., Shear stress inhibits adhesion of tracting factors, FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol. 3 (1989)
cultured mouse endothelial cells to lymphocytes by downregulating VCAM-1 2007e2018.
expression, Am. J. Physiol. 267 (1994) C679eC687. [129] S. Moncada, E.A. Higgs, Endogenous nitric oxide: physiology, pathology and
[100] R.E. Gerszten, Y.C. Lim, H.T. Ding, et al., Adhesion of monocytes to vascular clinical relevance, Eur. J. Clin. Investig. 21 (1991) 361e374.
cell adhesion molecule-1-transduced human endothelial cells: implications [130] E. Gkaliagkousi, A. Ferro, Nitric oxide signalling in the regulation of cardio-
for atherogenesis, Circ. Res. 82 (1998) 871e878. vascular and platelet function, Front. Biosci. J. Virtual Libr. 16 (2011)
[101] H. Kaneto, N. Katakami, M. Matsuhisa, et al., Role of reactive oxygen species 1873e1897.
in the progression of type 2 diabetes and atherosclerosis, Mediat. Inamm. [131] W.C. Sessa, J.K. Harrison, C.M. Barber, et al., Molecular cloning and expression
2010 (2010) 453892. of a cDNA encoding endothelial cell nitric oxide synthase, J. Biol. Chem. 267
[102] V.M. Victor, M. Rocha, E. Sola, et al., Oxidative stress, endothelial dysfunction (1992) 15274e15276.
and atherosclerosis, Curr. Pharm. Des. 15 (2009) 2988e3002. [132] R. Govers, T.J. Rabelink, Cellular regulation of endothelial nitric oxide syn-
[103] K.G. Birukov, Cyclic stretch, reactive oxygen species, and vascular remodel- thase, American journal of physiology, Ren. Physiol. 280 (2001) F193eF206.
ing, Antioxid. Redox Signal 11 (2009) 1651e1667. [133] G. Schuler, V. Adams, Y. Goto, Role of exercise in the prevention of cardio-
[104] S. Chatterjee, E.A. Browning, N. Hong, et al., Membrane depolarization is the vascular disease: results, mechanisms, and new perspectives, Eur. Heart J. 34
trigger for PI3K/Akt activation and leads to the generation of ROS, Am. J. (2013) 1790e1799.
Physiol. Heart Circ. Physiol. 302 (2012) H105eH114. [134] G. Yetik-Anacak, J.D. Catravas, Nitric oxide and the endothelium: history and
[105] A.L. Mowbray, D.H. Kang, S.G. Rhee, et al., Laminar shear stress up-regulates impact on cardiovascular disease, Vasc. Pharmacol. 45 (2006) 268e276.
peroxiredoxins (PRX) in endothelial cells: PRX 1 as a mechanosensitive [135] F. de Nigris, L.O. Lerman, S.W. Ignarro, et al., Benecial effects of antioxidants
antioxidant, J. Biol. Chem. 283 (2008) 1622e1627. and L-arginine on oxidation-sensitive gene expression and endothelial NO
[106] W. Takabe, E. Warabi, N. Noguchi, Anti-atherogenic effect of laminar shear synthase activity at sites of disturbed shear stress, Proc. Natl. Acad. Sci. U. S.
stress via Nrf2 activation, Antioxid. Redox Signal 15 (2011) 1415e1426. A. 100 (2003) 1420e1425.
[107] X.M. Liu, K.J. Peyton, W. Durante, Physiological cyclic strain promotes [136] S.P. Duckles, V.M. Miller, Hormonal modulation of endothelial NO produc-
endothelial cell survival via the induction of heme oxygenase-1, Am. J. tion, Pugers Arch. 459 (2010) 841e851.
Physiol. Heart Circ. Physiol. 304 (2013) H1634eH1643. [137] A.S. Thakor, E.A. Herrera, M. Seron-Ferre, et al., Melatonin and vitamin C
[108] P.M. Cummins, N. von Offenberg Sweeney, M.T. Killeen, et al., Cyclic strain- increase umbilical blood ow via nitric oxide-dependent mechanisms,
mediated matrix metalloproteinase regulation within the vascular endo- J. Pineal Res. 49 (2010) 399e406.
thelium: a force to be reckoned with, Am. J. Physiol. Heart Circ. Physiol. 292 [138] F. Jantzen, S. Konemann, B. Wolff, et al., Isoprenoid depletion by statins
(2007) H28eH42. antagonizes cytokine-induced down-regulation of endothelial nitric oxide
[109] H.B. Liu, J. Zhang, S.Y. Xin, et al., Mechanosensitive properties in the expression and increases NO synthase activity in human umbilical vein
108 P.A. Cahill, E.M. Redmond / Atherosclerosis 248 (2016) 97e109

endothelial cells, J. Physiol. Pharmacol. Off. J. Pol. Physiol. Soc. 58 (2007) [167] H. Lim, S.K. Dey, A novel pathway of prostacyclin signaling-hanging out with
503e514. nuclear receptors, Endocrinology 143 (2002) 3207e3210.
[139] S. Malik, A.K. Sharma, S. Bharti, et al., In vivo cardioprotection by pitavastatin [168] G. Edwards, K.A. Dora, M.J. Gardener, et al., K is an endothelium-derived
from ischemic-reperfusion injury through suppression of IKK/NF-kappaB hyperpolarizing factor in rat arteries, Nature 396 (1998) 269e272.
and upregulation of pAkt-e-NOS, J. Cardiovasc. Pharmacol. 58 (2011) [169] S. Taddei, D. Versari, A. Cipriano, et al., Identication of a cytochrome P450
199e206. 2C9-derived endothelium-derived hyperpolarizing factor in essential hy-
[140] R. Datar, W.H. Kaesemeyer, S. Chandra, et al., Acute activation of eNOS by pertensive patients, J. Am. Coll. Cardiol. 48 (2006) 508e515.
statins involves scavenger receptor-B1, G protein subunit Gi, phospholipase [170] F.M. Faraci, C.G. Sobey, S. Chrissobolis, et al., Arachidonate dilates basilar
C and calcium inux, Br. J. Pharmacol. 160 (2010) 1765e1772. artery by lipoxygenase-dependent mechanism and activation of K()
[141] D. Taubert, R. Roesen, C. Lehmann, et al., Effects of low habitual cocoa intake channels, Am. J. Physiol. Regul. Integr. Comp. Physiol. 281 (2001)
on blood pressure and bioactive nitric oxide: a randomized controlled trial, R246eR253.
JAMA 298 (2007) 49e60. [171] A. Ellis, C.R. Triggle, Endothelium-derived reactive oxygen species: their
[142] R.K. Davda, L.J. Chandler, F.T. Crews, et al., Ethanol enhances the endothelial relationship to endothelium-dependent hyperpolarization and vascular
nitric oxide synthase response to agonists, Hypertension 21 (1993) 939e943. tone, Can. J. Physiol. Pharmacol. 81 (2003) 1013e1028.
[143] R.J. Hendrickson, P.A. Cahill, J.V. Sitzmann, et al., Ethanol enhances basal and [172] C.M. Wei, S. Hu, V.M. Miller, et al., Vascular actions of C-type natriuretic
ow-stimulated nitric oxide synthase activity in vitro by activating an peptide in isolated porcine coronary arteries and coronary vascular smooth
inhibitory guanine nucleotide binding protein, J. Pharmacol. Exp. Ther. 289 muscle cells, Biochem. Biophys. Res. Commun. 205 (1994) 765e771.
(1999) 1293e1300. [173] T.M. Grifth, A.T. Chaytor, D.H. Edwards, The obligatory link: role of gap
[144] C.D. Venkov, P.R. Myers, M.A. Tanner, et al., Ethanol increases endothelial junctional communication in endothelium-dependent smooth muscle hy-
nitric oxide production through modulation of nitric oxide synthase perpolarization, Pharmacol. Res. Off. J. Italian Pharmacol. Soc. 49 (2004)
expression, Thromb. Haemost. 81 (1999) 638e642. 551e564.
[145] D.J. Kleinhenz, R.L. Sutliff, J.A. Polikandriotis, et al., Chronic ethanol ingestion [174] M. Yanagisawa, A. Inoue, T. Ishikawa, et al., Primary structure, synthesis, and
increases aortic endothelial nitric oxide synthase expression and nitric oxide biological activity of rat endothelin, an endothelium-derived vasoconstrictor
production in the rat, Alcohol. Clin. Exp. Res. 32 (2008) 148e154. peptide, Proc. Natl. Acad. Sci. U. S. A. 85 (1988) 6964e6967.
[146] K.J. Mukamal, K.M. Conigrave, M.A. Mittleman, et al., Roles of drinking [175] E. Thorin, D.J. Webb, Endothelium-derived endothelin-1, Pugers Arch. Eur.
pattern and type of alcohol consumed in coronary heart disease in men, J. Physiol. 459 (2010) 951e958.
N. Engl. J. Med. 348 (2003) 109e118. [176] T.F. Luscher, Z. Yang, M. Tschudi, et al., Interaction between endothelin-1 and
[147] P.E. Ronksley, S.E. Brien, B.J. Turner, et al., Association of alcohol consump- endothelium-derived relaxing factor in human arteries and veins, Circ. Res.
tion with selected cardiovascular disease outcomes: a systematic review and 66 (1990) 1088e1094.
meta-analysis, BMJ 342 (2011) d671. [177] P. Gilbert, J. Tremblay, E. Thorin, Endothelium-derived endothelin-1 reduces
[148] P.A. Cahill, E.M. Redmond, Alcohol and cardiovascular disease -modulation of cerebral artery sensitivity to nitric oxide by a protein kinase C-independent
vascular cell function, Nutrients 4 (2012) 297e318. pathway, Stroke J. Cereb. Circ. 32 (2001) 2351e2355.
[149] V. Kapil, R.S. Khambata, A. Robertson, et al., Dietary nitrate provides sus- [178] G. Callera, R. Tostes, C. Savoia, et al., Vasoactive peptides in cardiovascular
tained blood pressure lowering in hypertensive patients: a randomized, (patho)physiology, Expert Rev. Cardiovasc. Ther. 5 (2007) 531e552.
phase 2, double-blind, placebo-controlled study, Hypertension 65 (2015) [179] D.W. Anggrahini, N. Emoto, K. Nakayama, et al., Vascular endothelial cell-
320e327. derived endothelin-1 mediates vascular inammation and neointima for-
[150] E. Marsch, T.L. Theelen, B.J. Janssen, et al., The effect of prolonged dietary mation following blood ow cessation, Cardiovasc. Res. 82 (2009) 143e151.
nitrate supplementation on atherosclerosis development, Atherosclerosis [180] M.E. Ivey, N. Osman, P.J. Little, Endothelin-1 signalling in vascular smooth
245 (2015) 212e221. muscle: pathways controlling cellular functions associated with atheroscle-
[151] T. Fukai, M. Ushio-Fukai, Superoxide dismutases: role in redox signaling, rosis, Atherosclerosis 199 (2008) 237e247.
vascular function, and diseases, Antioxid. Redox Signal 15 (2011) [181] C. Ihling, T. Szombathy, B. Bohrmann, et al., Coexpression of endothelin-
1583e1606. converting enzyme-1 and endothelin-1 in different stages of human
[152] S. Yamagishi, T. Matsui, Nitric oxide, a janus-faced therapeutic target for atherosclerosis, Circulation 104 (2001) 864e869.
diabetic microangiopathyeFriend or foe? Pharmacol. Res. 64 (2011) [182] H. Arai, S. Hori, I. Aramori, et al., Cloning and expression of a cDNA encoding
187e194. an endothelin receptor, Nature 348 (1990) 730e732.
[153] K. Okahara, B. Sun, J. Kambayashi, Upregulation of prostacyclin synthesis- [183] T. Sakurai, M. Yanagisawa, Y. Takuwa, et al., Cloning of a cDNA encoding a
related gene expression by shear stress in vascular endothelial cells, Arte- non-isopeptide-selective subtype of the endothelin receptor, Nature 348
rioscler. Thromb. Vasc. Biol. 18 (1998) 1922e1926. (1990) 732e735.
[154] A.J. Flammer, T.F. Luscher, Human endothelial dysfunction: EDRFs, Pugers [184] G. de Nucci, R. Thomas, P. D'Orleans-Juste, et al., Pressor effects of circulating
Arch. Eur. J. Physiol. 459 (2010) 1005e1013. endothelin are limited by its removal in the pulmonary circulation and by
[155] S. Moncada, R. Gryglewski, S. Bunting, et al., An enzyme isolated from ar- the release of prostacyclin and endothelium-derived relaxing factor, Proc.
teries transforms prostaglandin endoperoxides to an unstable substance that Natl. Acad. Sci. U. S. A. 85 (1988) 9797e9800.
inhibits platelet aggregation, Nature 263 (1976) 663e665. [185] N. Dhaun, D.M. Pollock, J. Goddard, et al., Selective and mixed endothelin
[156] S. Narumiya, Y. Sugimoto, F. Ushikubi, Prostanoid receptors: structures, receptor antagonism in cardiovascular disease, Trends Pharmacol. Sci. 28
properties, and functions, Physiol. Rev. 79 (1999) 1193e1226. (2007) 573e579.
[157] E.M. Smyth, T. Grosser, M. Wang, et al., Prostanoids in health and disease, [186] J.J. Maguire, A.P. Davenport, Endothelin@25-new agonists, antagonists, in-
J. Lipid Res. 50 (Suppl. S423-428) (2009). hibitors and emerging research frontiers: IUPHAR Review 12, Br. J. Phar-
[158] T. Szeran, N. Erdei, T. Fulop, et al., Increased cyclooxygenase-2 expression macol. 171 (2014) 5555e5572.
and prostaglandin-mediated dilation in coronary arterioles of patients with [187] M.C. Kowala, P.M. Rose, P.D. Stein, et al., Selective blockade of the endothelin
diabetes mellitus, Circ. Res. 99 (2006) e12e17. subtype A receptor decreases early atherosclerosis in hamsters fed choles-
[159] M.W. Radomski, R.M. Palmer, S. Moncada, Comparative pharmacology of terol, Am. J. Pathol. 146 (1995) 819e826.
endothelium-derived relaxing factor, nitric oxide and prostacyclin in plate- [188] M. Barton, C.C. Haudenschild, L.V. d'Uscio, et al., Endothelin ETA receptor
lets, Br. J. Pharmacol. 92 (1987) 181e187. blockade restores NO-mediated endothelial function and inhibits athero-
[160] J. Kawabe, F. Ushikubi, N. Hasebe, Prostacyclin in vascular diseases. e Recent sclerosis in apolipoprotein E-decient mice, Proc. Natl. Acad. Sci. U. S. A. 95
insights and future perspectives, Circ. J. Off. J. Jpn. Circ. Soc. 74 (2010) (1998) 14367e14372.
836e843. [189] S. Babaei, P. Picard, A. Ravandi, et al., Blockade of endothelin receptors
[161] H. Shimokawa, N.A. Flavahan, R.R. Lorenz, et al., Prostacyclin releases markedly reduces atherosclerosis in LDL receptor decient mice: role of
endothelium-derived relaxing factor and potentiates its action in coronary endothelin in macrophage foam cell formation, Cardiovasc. Res. 48 (2000)
arteries of the pig, Br. J. Pharmacol. 95 (1988) 1197e1203. 158e167.
[162] E. Delpy, H. Coste, A.C. Gouville, Effects of cyclic GMP elevation on [190] D. Fukuda, M. Sata, Role of bone marrow renin-angiotensin system in the
isoprenaline-induced increase in cyclic AMP and relaxation in rat aortic pathogenesis of atherosclerosis, Pharmacol. Ther. 118 (2008) 268e276.
smooth muscle: role of phosphodiesterase 3, Br. J. Pharmacol. 119 (1996) [191] C.R. Tirapelli, D. Bonaventura, L.F. Tirapelli, et al., Mechanisms underlying the
471e478. vascular actions of endothelin 1, angiotensin II and bradykinin in the rat
[163] M. Isogaya, N. Yamada, H. Koike, et al., Inhibition of restenosis by beraprost carotid, Pharmacology 84 (2009) 111e126.
sodium (a prostaglandin I2 analogue) in the atherosclerotic rabbit artery [192] C.A. Lemarie, E.L. Schiffrin, The angiotensin II type 2 receptor in cardiovas-
after angioplasty, J. Cardiovasc. Pharmacol. 25 (1995) 947e952. cular disease, J. Ren. Angiotensin Aldosterone System JRAAS 11 (2010)
[164] Y. Numaguchi, K. Naruse, M. Harada, et al., Prostacyclin synthase gene 19e31.
transfer accelerates reendothelialization and inhibits neointimal formation [193] M. Graninger, R. Reiter, C. Drucker, et al., Angiotensin receptor blockade
in rat carotid arteries after balloon injury, Arterioscler. Thromb. Vasc. Biol. 19 decreases markers of vascular inammation, J. Cardiovasc. Pharmacol. 44
(1999) 727e733. (2004) 335e339.
[165] A. Sugawara, M. Kudo, A. Saito, et al., Novel effects of beraprost sodium on [194] F. Montecucco, A. Pende, F. Mach, The renin-angiotensin system modulates
vasculatures, Int. Angiol. 29 (2010) 28e32. inammatory processes in atherosclerosis: evidence from basic research and
[166] E. Arehart, J. Stitham, F.W. Asselbergs, et al., Acceleration of cardiovascular clinical studies, Mediat. Inamm. 2009 (2009) 752406.
disease by a dysfunctional prostacyclin receptor mutation: potential impli- [195] S.P. Jackson, Arterial thrombosiseinsidious, unpredictable and deadly, Nat.
cations for cyclooxygenase-2 inhibition, Circ. Res. 102 (2008) 986e993. Med. 17 (2011) 1423e1436.
P.A. Cahill, E.M. Redmond / Atherosclerosis 248 (2016) 97e109 109

[196] R.C. Jin, B. Voetsch, J. Loscalzo, Endogenous mechanisms of inhibition of [216] K.H. Lao, L. Zeng, Q. Xu, Endothelial and smooth muscle cell transformation
platelet function, Microcirculation 12 (2005) 247e258. in atherosclerosis, Curr. Opin. Lipidol. 26 (5) (2015) 449e456.
[197] K.A. Tanaka, N.S. Key, J.H. Levy, Blood coagulation: hemostasis and thrombin [217] E. Londin, P. Loher, A.G. Telonis, et al., Analysis of 13 cell types reveals evi-
regulation, Anesth. Analg. 108 (2009) 1433e1446. dence for the expression of numerous novel primate- and tissue-specic
[198] P.L. Giesen, B.S. Fyfe, J.T. Fallon, et al., Intimal tissue factor activity is released microRNAs, Proc. Natl. Acad. Sci. U. S. A. 112 (2015) E1106eE1115.
from the arterial wall after injury, Thromb. Haemost. 83 (2000) 622e628. [218] D. Santovito, V. Egea, C. Weber, Small but smart: MicroRNAs orchestrate
[199] N. Mackman, The many faces of tissue factor, J. Thromb. Haemost. JTH 7 atherosclerosis development and progression, Biochim. Biophys. Acta (2015).
(Suppl. 1) (2009) 136e139. S1388-1981(15)00235-8.
[200] T. Murata, Y. Nakashima, C. Yasunaga, et al., Extracellular and cell-associated [219] A. Schober, M. Nazari-Jahantigh, C. Weber, MicroRNA-mediated mechanisms
localizations of plasminogen activators and plasminogen activator inhibitor- of the cellular stress response in atherosclerosis, Nat. Rev. Cardiol. 12 (2015)
1 in cultured endothelium, Exp. Mol. Pathol. 55 (1991) 105e118. 361e374.
[201] A. Schafer, J. Bauersachs, Endothelial dysfunction, impaired endogenous [220] K. Di Gregoli, N. Jenkins, R. Salter, et al., MicroRNA-24 regulates macrophage
platelet inhibition and platelet activation in diabetes and atherosclerosis, behavior and retards atherosclerosis, Arterioscler. Thromb. Vasc. Biol. 34
Curr. Vasc. Pharmacol. 6 (2008) 52e60. (2014) 1990e2000.
[202] Z.M. Ruggeri, Platelets in atherothrombosis, Nat. Med. 8 (2002) 1227e1234. [221] A. Schober, M. Nazari-Jahantigh, Y. Wei, et al., MicroRNA-126-5p promotes
[203] S. Fujiyama, K. Amano, K. Uehira, et al., Bone marrow monocyte lineage cells endothelial proliferation and limits atherosclerosis by suppressing Dlk1, Nat.
adhere on injured endothelium in a monocyte chemoattractant protein-1- Med. 20 (2014) 368e376.
dependent manner and accelerate reendothelialization as endothelial pro- [222] T.A. Harris, M. Yamakuchi, M. Ferlito, et al., MicroRNA-126 regulates endo-
genitor cells, Circ. Res. 93 (2003) 980e989. thelial expression of vascular cell adhesion molecule 1, Proc. Natl. Acad. Sci.
[204] N. Werner, J. Priller, U. Laufs, et al., Bone marrow-derived progenitor cells U. S. A. 105 (2008) 1516e1521.
modulate vascular reendothelialization and neointimal formation: effect of [223] F. Lovren, Y. Pan, A. Quan, et al., MicroRNA-145 targeted therapy reduces
3-hydroxy-3-methylglutaryl coenzyme a reductase inhibition, Arterioscler. atherosclerosis, Circulation 126 (2012) S81eS90.
Thromb. Vasc. Biol. 22 (2002) 1567e1572. [224] F. Du, F. Yu, Y. Wang, et al., MicroRNA-155 deciency results in decreased
[205] T. Asahara, T. Murohara, A. Sullivan, et al., Isolation of putative progenitor macrophage inammation and attenuated atherogenesis in apolipoprotein
endothelial cells for angiogenesis, Science 275 (1997) 964e967. E-decient mice, Arterioscler. Thromb. Vasc. Biol. 34 (2014) 759e767.
[206] P.A. Williams, E.A. Silva, The role of synthetic extracellular matrices in [225] S. Meiler, Y. Baumer, E. Toulmin, et al., MicroRNA 302a is a novel modulator
endothelial progenitor cell homing for treatment of vascular disease, Ann. of cholesterol homeostasis and atherosclerosis, Arterioscler. Thromb. Vasc.
Biomed. Eng. 43 (10) (2015) 2301e2313. Biol. 35 (2015) 323e331.
[207] L. Hou, J.J. Kim, Y.J. Woo, et al., Stem cell-based therapies to promote [226] E.E. Creemers, A.J. Tijsen, Y.M. Pinto, Circulating microRNAs: novel bio-
angiogenesis in ischemic cardiovascular disease, Am. J. Physiol. Heart Circ. markers and extracellular communicators in cardiovascular disease? Circ.
Physiol. 310 (4) (2015 15) H455eH465. Res. 110 (2012) 483e495.
[208] R. Madonna, R. De Caterina, Circulating endothelial progenitor cells: do they [227] S. Kumar, C.W. Kim, R.D. Simmons, et al., Role of ow-sensitive microRNAs in
live up to their name? Vasc. Pharmacol. 67e69 (2015) 2e5. endothelial dysfunction and atherosclerosis: mechanosensitive athero-miRs,
[209] M.K. Hagensen, J. Shim, T. Thim, et al., Circulating endothelial progenitor Arterioscler. Thromb. Vasc. Biol. 34 (2014) 2206e2216.
cells do not contribute to plaque endothelium in murine atherosclerosis, [228] K.M. Welch-Reardon, N. Wu, C.C. Hughes, A role for partial endothelial-
Circulation 121 (2010) 898e905. mesenchymal transitions in angiogenesis? Arterioscler. Thromb. Vasc. Biol.
[210] D.J. Ceradini, A.R. Kulkarni, M.J. Callaghan, et al., Progenitor cell trafcking is 35 (2015) 303e308.
regulated by hypoxic gradients through HIF-1 induction of SDF-1, Nat. Med. [229] L.A. van Meeteren, P. ten Dijke, Regulation of endothelial cell plasticity by
10 (2004) 858e864. TGF-beta, Cell Tissue Res. 347 (2012) 177e186.
[211] P. Liu, B. Zhou, D. Gu, et al., Endothelial progenitor cell therapy in athero- [230] B.C. Cooley, J. Nevado, J. Mellad, et al., TGF-beta signaling mediates
sclerosis: a double-edged sword? Ageing Res. Rev. 8 (2009) 83e93. endothelial-to-mesenchymal transition (EndMT) during vein graft remod-
[212] K. Williamson, S.E. Stringer, M.Y. Alexander, Endothelial progenitor cells eling, Sci. Transl. Med. 6 (2014) 227e234.
enter the aging arena, Front. Physiol. 3 (2012) 30. [231] Z. Li, P.J. Wermuth, B.S. Benn, et al., Caveolin-1 deciency induces sponta-
[213] S. Devaraj, I. Jialal, Dysfunctional endothelial progenitor cells in metabolic neous endothelial-to-mesenchymal transition in murine pulmonary endo-
syndrome, Exp. Diabet. Res. 2012 (2012) 585018. thelial cells in vitro, Am. J. Pathol. 182 (2013) 325e331.
[214] G.P. Fadini, C. Agostini, A. Avogaro, Autologous stem cell therapy for pe- [232] P.Y. Chen, L. Qin, N. Baeyens, et al., Endothelial-to-mesenchymal transition
ripheral arterial disease meta-analysis and systematic review of the litera- drives atherosclerosis progression, J. Clin. Investig. 2015 (2015).
ture, Atherosclerosis 209 (2010) 10e17. [233] J.R. Moonen, E.S. Lee, M. Schmidt, et al., Endothelial-to-mesenchymal tran-
[215] Z. Tang, A. Wang, F. Yuan, et al., Differentiation of multipotent vascular stem sition contributes to bro-proliferative vascular disease and is modulated by
cells contributes to vascular diseases, Nat. Commun. 3 (2012) 875. uid shear stress, Cardiovasc. Res. 108 (2015) 377e386.

Вам также может понравиться