Next Article in Journal
Antibacterial Activity and Anti-Quorum Sensing Mediated Phenotype in Response to Essential Oil from Melaleuca bracteata Leaves
Next Article in Special Issue
Therapeutic Effect of Iron Citrate in Blocking Calcium Deposition in High Pi-Calcified VSMC: Role of Autophagy and Apoptosis
Previous Article in Journal
Extracellular Vesicles—Connecting Kingdoms
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of Vascular Smooth Muscle Cells in Arterial Remodeling: Focus on Calcification-Related Processes

1
Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands
2
Department of Biomedical Engineering, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands
3
Department of Internal Medicine, Maastricht University Medical Centre (MUMC+), 6229 HX Maastricht, The Netherlands
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2019, 20(22), 5694; https://doi.org/10.3390/ijms20225694
Submission received: 8 October 2019 / Revised: 31 October 2019 / Accepted: 8 November 2019 / Published: 14 November 2019
(This article belongs to the Special Issue Mechanisms of Vascular Calcification)

Abstract

:
Arterial remodeling refers to the structural and functional changes of the vessel wall that occur in response to disease, injury, or aging. Vascular smooth muscle cells (VSMC) play a pivotal role in regulating the remodeling processes of the vessel wall. Phenotypic switching of VSMC involves oxidative stress-induced extracellular vesicle release, driving calcification processes. The VSMC phenotype is relevant to plaque initiation, development and stability, whereas, in the media, the VSMC phenotype is important in maintaining tissue elasticity, wall stress homeostasis and vessel stiffness. Clinically, assessment of arterial remodeling is a challenge; particularly distinguishing intimal and medial involvement, and their contributions to vessel wall remodeling. The limitations pertain to imaging resolution and sensitivity, so methodological development is focused on improving those. Moreover, the integration of data across the microscopic (i.e., cell-tissue) and macroscopic (i.e., vessel-system) scale for correct interpretation is innately challenging, because of the multiple biophysical and biochemical factors involved. In the present review, we describe the arterial remodeling processes that govern arterial stiffening, atherosclerosis and calcification, with a particular focus on VSMC phenotypic switching. Additionally, we review clinically applicable methodologies to assess arterial remodeling and the latest developments in these, seeking to unravel the ubiquitous corroborator of vascular pathology that calcification appears to be.

Graphical Abstract

1. Introduction

The vessel wall consists of multiple cells, including endothelial cells (ECs), vascular smooth muscle cells (VSMCs), fibroblasts and pericytes, and structural extracellular matrix (ECM) components, such as elastin and collagen [1]. ECs form the innermost layer, being the barrier between the blood-carrying lumen and vessel wall. In the vessel wall, ECs are surrounded by the tunica media, consisting almost entirely of circumferentially oriented VSMCs. Medial VSMCs, facilitating vessel dilation and constriction, are surrounded by interconnected elastic lamellae [2]. The outer layer of the vessel wall is the adventitia and contains ECM, maintaining structural integrity under peak mechanical load, and fibroblasts and progenitor (mesenchymal stem cell-like, MSC-like) cells. The interplay of all components of the vessel wall is needed to preserve vascular health [3,4,5].
The vessel wall is continuously exposed to biomechanical and biochemical stressors that elicit functional and adaptative responses. For instance, when blood flow is acutely increased, the increased wall shear stress (or friction) is sensed by ECs, which release relaxants. These relaxants (e.g., nitric oxide, NO) lower the active tone of VSMCs, leading to (flow-mediated) vessel dilation to counteract the initial increase in wall shear stress [6]. Long-term changes in wall shear stress provoke diameter adaptation [7]. Wall thickness changes are seen in response to increased wall stress (or wall tension), due to high blood pressure [8]. Such arterial remodeling responses shape early development of the macro-circulation, but are also affected by normal aging processes [9]. A multitude of biological processes contribute to pathological vascular remodeling, such as inflammation, oxidative stress, lipid accumulation, and degradation of the ECM [10,11]. Due to the heterogeneity of vascular remodeling, it is difficult to pin-point single biological processes responsible for vascular disease. Lessons from genetic disorders such as pseudoxanthoma elasticum (PXE), Marfan’s syndrome and Keutel syndrome, have addressed the complexity of arterial remodeling [12]. A better understanding of the complexity of arterial remodeling will help unravel its role as a cause or consequence of cardiovascular disease (CVD).
VSMCs in the medial layer of the vessel wall play a key role in arterial remodeling. VSMCs are the most abundant cell type in the arterial vessel wall and are pivotal in maintaining vessel structure and function [13]. VSMCs are considered heterogeneous and display a high degree of plasticity [5]. Under physiological conditions, VSMCs have a contractile phenotype which facilitates the contraction and dilation of the vasculature, which, in smaller resistance arteries, is key to the regulation of blood flow. Upon biological stress signals or vascular injury, VSMCs respond by losing contractility markers and differentiate towards a synthetic VSMC phenotype. Synthetic VSMCs subsequently express proteins involved in proliferation and migration [14]. Contractile VSMCs may adapt to stress by differentiating towards synthetic VSMCs which are able to newly synthesize ECM components such as collagen. To remodel the ECM, synthetic VSMCs produce metalloproteinases (MMPs), such as collagenases and elastases, that allow them to migrate to sites of injury [15,16,17]. VSMC-mediated remodeling of ECM within the vessel wall may result in increased arterial stiffness [13,18], contributing to systolic hypertension and altered hemodynamic conditions in end-organs such as the brain, kidneys and heart [19].
VSMCs are involved in many vascular diseases, such as atherosclerosis and aneurysm formation. In all these vascular pathologies, vascular calcification is involved [5,20,21]. Vascular calcification is defined as the deposition of calcium crystals within the vessel wall, initiated as microcalcification and propagating towards macrocalcifications [22], and, eventually, encroaching entire segments of the vasculature [23]. Vascular calcification is an active process with a key role for VSMCs, including apoptosis [24], osteochondrogenic transdifferentiation [25], extracellular vesicle release [2], calcium overload [26], and cellular senescence [27] (Figure 1). The aim of the present review is to provide an overview of the pathways underlying arterial remodeling, with a particular focus on the role of VSMCs and calcification-related processes.

2. Biology of Vascular Remodeling

2.1. Arterial Remodeling

Arterial remodeling reflects the adaptation of the vessel wall to biochemical and biomechanical stimuli [9,12]. In the vasculature, two types of remodeling can be distinguished: outward and inward remodeling, with respective hypertrophy (thickening) or hypotrophy (thinning) of the vessel wall. Large conduit vessels do not have the ability to constrict in response to stress, and therefore show hypertrophic remodeling. Atherosclerosis is characterized by an increase in vessel diameter, with the thickening of both the media and intima, and classically termed outward hypertrophic remodeling [28]. Aneurysm formation is characterized by an increase in vessel diameter, with a thinning of the vessel wall, and termed outward hypothropic remodeling [29] (Figure 2). Inward remodeling is less frequently seen and is observed in more muscular peripheral arteries, probably reflecting the sustained vasoconstriction of vessels [30].
Under low laminar flow, platelet derived growth factors (PDGF) and transforming growth factor-β (TGF-β) promote inward remodeling by increasing VSMC proliferation and collagen deposition [31]. Inward remodeling also contributes to atherosclerosis, but does not lead to an increased vessel size (Figure 2). Atherosclerosis development involves many cellular processes, like the recruitment of inflammatory cells by chemotaxis and infiltration [32].
VSMCs are connected to a fenestrated network of elastin and collagen fibers. The capacity of the vessel wall to elastically distend is important to accommodate the volume ejected with each heartbeat and to limit peripheral pressure pulsations. The active tone and spatial arrangement of VSMCs may influence the mechanical load on the ECM components and, therefore, modulate vessel diameter and stiffness [18]. Chronic exposure to high blood pressure increases tensile stress [9] to which VSMCs respond by proliferation, resulting in hyperplasia and thickening of the vascular wall. Concomitant activity of MMPs facilitates the structural breakdown of elastin ECM, and synthetic VSMCs produce collagen ECM, attempting to preserve stiffness homeostasis [33]. High blood pressure thus aggravates age-related stiffening of arteries [34,35,36]. Additional ECM disturbances, such as the presence of calcium crystals, have a further impact on the stiffening of the medial layer [37,38].
During aging, it is generally accepted that the number of cells in the vasculature decreases, although the causes of this finding remain to be established [34]. It has been hypothesized that VSMCs become senescent and that cell division rates decrease [39]. The recent literature ignores vessel wall cellularity and often refers to cellular processes, such as apoptosis, inflammation, calcification and epigenetic effects, all playing a part in vessel wall aging [20]. Additionally, with aging, collagen content in major arterial vasculature increases, whereas elastin content decreases and the number of VSMCs declines [40]. As a consequence, remaining VSMCs are embedded in a collagen-enriched ECM with fewer cellular focal adhesions [41,42,43]. Within arterial vessels, differences exist in the content of elastin to VSMC ratios [44]. Large arteries close to the heart contain more elastin and are therefore called “elastic” arteries. It is particularly elastic arteries that stiffen with age [45]. Large artery stiffening results in decreased arterial compliance, especially in those aged over 60 years [19,46,47]. Peripheral vessels contain more VSMCs relative to elastin and are termed “muscular” arteries. In muscular arteries, the relative elastin content increases with age, most likely caused by the decline in the number of VSMCs and decreased collagen content [44]. It should be noted that the absolute amount of ECM proteins in the vasculature decreases with age, but that fat and extracellular material, such as calcium crystals, increase [48]. Taken together, the number of VSMCs within the vasculature strongly correlates with vascular stiffening and the arterial remodeling processes [34].
Endothelial function plays an important role in arterial remodeling. Blood flow and normal wall shear stress stimulate ECs to produce NO (Figure 3). NO induces the relaxation of local VSMCs, which leads to dilation of the vessel wall. Endothelium-dependent vasodilation decreases with age, which appears to be associated (perhaps causally or bi-directionally) with a quiescent state of VSMCs and is clearly implicated to be associated with hypertension and CVD [49,50]. Under pathological conditions, ECs are known to produce cytokines and growth factors [51], which induces VSMCs phenotype switching from contractile to a more synthetic phenotype. An increased wall shear stress increases endothelial derived NO release, further decreasing VSMC proliferation and increasing VSMC apoptosis [52], resulting in outward remodeling and stiffened vasculature, due to the formation of the ECM matrix by remaining synthetic VSMCs [34,53].

2.1.1. Inflammation and Arterial Remodeling

Inflammatory processes play a significant role in arterial remodeling and are linked to the development of atherosclerosis. Chronic low-grade inflammation is a key driver of arterial aging. VSMC phenotypic switching and ECM biochemical changes activate inflammatory pathways and oxidative stress signaling [54]. Cytokines determine processes such as endothelial dysfunction, renin angiotensin activation and metalloproteinases release [55,56]. VSMCs are involved in local vessel wall inflammatory functions.

Proliferation

VSMCs have low turnover rates in healthy vessels, but increase proliferation upon vascular injury to initiate repair [57]. Increased expression of aging genes p16INK4a and p21 [58] have been linked to VSMC senescence and are present in atherosclerotic plaques, indicating lower proliferation and, thus, decreased repair capacity [59].

Cell Death

Cell death is linked to vascular disease, but absolute rates are difficult to determine. VSMC apoptosis in atherosclerosis has been linked to plaque vulnerability features [60], and chronic VSMC apoptosis has been shown to promote atherogenesis and plaque progression [61]. In vascular aging and medial remodeling, VSMCs cell death does not cause significant inflammation, due to the efficient IL-1β-mediated clearance of apoptotic bodies by VSMCs [62].

Platelets and Extracellular Vesicles (EVs)

Platelets have shown to affect VSMC inflammatory phenotypes and injury responses [63]. Additionally, platelet EVs induce an inflammatory response in VSMC in vitro [64]. Platelet factor 4 (PF4) has a central role in the stimulation of VSMC-mediated cytokine release, which is primarily affected by increased krüppel-like factor 4 (KLF-4) transcription. Experimental atherosclerosis models indicated that PF4 is proatherogenic and it has been found to penetrate deep into the vessel wall, underlining the importance of platelets aside from their effect on the endothelium [65].

2.2. The Role of VSMC Plasticity in Vascular Remodeling

Under physiological conditions, VSMCs display a contractile phenotype, in which markers such as alpha-smooth muscle actin (α-SMA), smooth muscle 22-alpha (SM22-α), smoothelin A/B (smtn), smooth muscle-myosin heavy chain (MYC11) and Calponin-1 (CNN-1) are highly expressed [14,66,67] (Table 1). VSMCs are heterogeneous with regard to their expression, are derived from different embryonic origins and have a strong genetic component causing VSMC diversity. Numerous in vivo studies have shown the existence of diverse VSMC populations within the same artery in rats [68], pigs [69] and humans [70]. We refer to VSMC plasticity here as the capability of VSMCs to switch phenotype. VSMC phenotypes are changed by environmental cues, such as soluble biochemical compounds, ECM proteins and biophysical conditions [33,71] (Figure 1). VSMCs exhibit a whole array of phenotypes, ranging from contractile–quiescent to migratory–proliferative–synthetic and osteogenic-, macrophage- or MSC-like [72,73,74] (Table 1). In the following subsections, we will briefly describe the most relevant factors that affect VSMC phenotype (Figure 1 and Table 2).

2.2.1. Biochemical Compounds

PDGF is an important signaling molecule in the initial phase of VSMC differentiation. During vascular development, PDGF causes mesenchymal cell recruitment and subsequent proliferation [75]. PDGF is known to induce VSMC differentiation towards a synthetic phenotype, as it downregulates the contractile marker αSMA in aortic VSMCs [76]. Additionally, PDGF increases proliferation and migration in pig and human coronary VSMCs [69,77,78]. Moreover, in vivo studies showed that inhibition of PDGF results in reduced VSMC proliferation and migration after arterial balloon injury [79,80].
TGF-β, as opposed to PDGF, induces a contractile and non-proliferative VSMC phenotype. An absence of TGF-β results in severe congenital cardiovascular disease, mainly shown in structural defects in experimental animals [81]. In vitro treatment of VSMCs with TGF-β induces expression of αSMA, MHC and CNN1 [69,82,83].
Activated coagulation proteins also have been shown to affect VSMC phenotype via protease activated receptors (PARs) [84]. Both tumor necrosis factor-alpha (TNF-α) and angiotensin II have been shown to induce the contractile, as well as the synthetic, phenotype of VSMCs. It has been shown that angiotensin II affects VSMC phenotypic plasticity via the induction of ROS and decreased scavenging activity of nitrate reductase (NAD(P)H) oxidases [85] and subsequently induces aneurysm formation via oxidative stress [21,86].

2.2.2. Extracellular Components

The ECM in which VSMCs are embedded also affects VSMC phenotype. VSMC phenotype is modulated via integrin receptors that are present on ECM proteins [87]. ECM consists of structural proteins, such as collagen, elastin and proteoglycans. For instance, the proteoglycan heparin promotes VSMC contractility. In vitro, heparin treatment of VSMCs induces a contractile phenotype and slows down its proliferation [69,88]. Collagen has pleiotropic effects on VSMC phenotype, depending on the type of collagen. Collagen type-I and fibronectin induce a synthetic VSMC phenotype [89,90] with proliferation [91,92]. On the contrary, collagen type-IV and laminin promote a contractile VSMC phenotype [89,93]. Intact elastin is associated with a contractile VSMC phenotype. The loss of elastin, by a genetic mutation, is associated with increased hypertrophy and hyperplasia of VSMCs [94,95]. Additionally, elastin remodeling, by the modulation of cathepsins and MMPs, promotes VSMC phenotypic switching that induces calcification [96,97].
Not only composition, but also the organization of structural fibers, determines VSMC phenotype. Culturing VSMCs in 3D compared to 2D increases contractile expression markers and induces TGF-β expression [98,99]. Culturing VSMCs on scaffold templates controls spatial organization and morphological response of VSMCs, indicating that VSMCs react to structural environmental changes to retain vessel function [100].

2.2.3. Biophysical Factors

Hemodynamics at the vessel level (flow and transmural pressure) also affect VSMCs by determining tensile loading and active tone (e.g., by NO). Changes in vessel wall shear stress modulate EC NO release, and influence VSMCs via cell–cell interaction with ECs [101]. Using co-cultures of ECs and VSMCs, shear stress induces a synthetic VSMC phenotype by decreasing α-SMA, MYH and smtn expression [69]. However, 2D co-culture systems do not mimic the complex vessel wall architecture in vivo. Changes in blood pressure directly transfer into changes in ECM tensile stress, which, as determined by (local) matrix elastic behavior and VSMC stiffness, results in VSMC deformation (i.e., mechanical strain). Alterations in VSMC strain are understood to modulate cellular phenotype [33]. Mechanical stretching forces applied directly to VSMCs enhance the expression of ECM proteins, such as collagen and fibronectin [102]. Biophysical factors in the physiological range promote VSMCs to maintain or adopt a contractile phenotype, whereas pathological biophysical stimuli promotes VSMCs to switch towards a synthetic phenotype.

2.2.4. Transcriptional Regulators

KLF4 is an important transcriptional regulator defining VSMC phenotype during development [103] and after vascular injury [104]. KLF4 expression is increased in lesions of ApoE-/- mice on a Western type diet [105]. Additionally, there is increased binding of KLF4 to αSMA and SM22α promotors upon vascular injury in mouse carotid arteries [104]. The loss of KLF4 has favorable effects, inhibiting plaque pathogenesis and reducing plaque vulnerability [106]. KLF4 is critical in the regulation of phenotypic switching of VSMCs, both in vitro and in vascular injury models. KLF4 expression results in profound activation of pluripotency genes such as Oct4 and Sox2, indicating that VSMCs can reactivate its pluripotency network in response to vascular stress or damage [5]. Moreover, KLF4 activates > 800 pro-inflammatory VSMC genes, of which many are atherosclerosis relevant [106]. Additionally, KLF4 knock-out in an atherosclerosis animal model revealed the switch of VSMCs toward a synthetic phenotype, while suppressing the macrophage-like form [106].

3. Clinical Features of Vascular Remodeling

3.1. Hypertension

Hypertension is widely accepted as a risk factor for the development of CVD. However, hypertension was first considered to be a consequence of aging and seen as insurmountable [107]. Later, several studies revealed that hypertension was associated with increased cardiovascular mortality [108,109]. A meta-analysis on blood pressure and cardiovascular disease showed that a rise of 20 mmHg in systolic blood pressure (SBP) and of 10 mmHg in diastolic blood pressure (DBP) was associated with a more than two-fold increase in vascular mortality [110]. More recently, the CALIBER study revealed that patients with hypertension (defined as > 140/90 mm Hg or those receiving blood pressure-lowering drugs) have a lifetime risk for overall CVD of 63.3% at 30 years of age (compared to 46.1% in patients with normal blood pressure) and develop CVD 5 years earlier [111]. Decreasing the SBP by 20 mmHg was associated with a 39% reduction in cardiovascular (CV) events in the total group and in a 69% reduction in patients between 60 and 69 years of age [112]. Currently, much of the focus on the primary and secondary prevention of CVD revolves around control of blood pressure [112].
The systolic aspect of hypertension is quite strongly determined by age- and disease-related decreases in arterial compliance [9,111,112,113]. Therefore, arterial remodeling processes affecting medial elastic properties are directly relevant in risk profiling and as treatment targets.

3.1.1. Cellular Components and Hypertension

High blood pressure is a multifactorial disorder in which genetic alterations, environmental factors and comorbidities interact [24,114]. ECs play an important role in the development of hypertension. In health, ECs are exposed to physiological shear stress, which is necessary to maintain proper functioning of the endothelium. The NO excreted by ECs regulates vascular tone, and thereby preserves ECM and VSMCs’ functioning [115]. In pathology, blood flow is more oscillatory, with higher peaks and blood stasis during diastole [116]. Turbulent flow and changes in shear stress, either high or low, affect EC function. Low shear stress areas are considered prone to developing atherosclerosis, inducing pro-inflammatory pathways and lead to dysregulation of the cytoskeleton and junctional proteins of ECs [115,117,118]. High shear stress induces morphological changes in ECs, as they align in the direction of the flow. Additionally, arterial outward remodeling occurs, leading to increased NO synthesis [119,120].
In hypertension, the amplitude and rate of elastic distension of arteries are increased, causing fatigue, damage and degradation of the vessel wall ECM [32]. Moreover, excessive VSMC strains induce phenotypic switching of these VSMCs to more synthetic types and, consequently, promotes arterial fibrosis, (further) compromising arterial compliance [121].

3.1.2. Calcification and Hypertension

Several pathologies are associated with VSMC phenotype switching [12]. Synthetic VSMCs produce EVs [2,122,123], that have been found in both the intimal and medial layers of the vessel wall [124]. VSMCs are known to release EVs upon phenotypic switching towards synthetic or osteogenic phenotype. EVs derived from VSMCs share similarities with EVs from osteoblasts, having calcium-binding capacities and osteoblast-like ECM production [2,80]. Recently, it was shown that not all EVs promote calcification [125]. This implies that EV content is variable and dependent on VSMC-mediated biogenesis. A specific subclass of EVs are exosomes, which express tetraspanins (CD9, CD63 and CD81) and differ in expression pattern, and, hence, mineralizing capacity [125]. Furthermore, calcifying conditions in vitro increase the multi-vesicular body, forming enzyme sphingomyelin phosphodiesterase 3 (SMPD3) and subsequent exosome genesis [80]. Consequently, the inhibition of SMPD3 ablates the generation of exosomes and their subsequent calcification. Additionally, sortilin has been suggested as a key player in the VSMC-mediated calcifying EVs genesis and release. Sortilin is a sorting receptor that directs target proteins to a designated location via the secretory or endocytic compartment [126]. Recent findings have identified that sortilin promotes vascular calcification via the trafficking and loading of tissue-nonspecific alkaline phosphatase into EVs [127]. Moreover, sortilin co-localized with calcification in human calcified vessels [127]. Differences between mineralizing and non-mineralizing EVs eventually determine calcification of the ECM in proximity of VSMCs. Hence, better insight into EV composition, such as lipid content and RNA and protein profile, might provide new insights into the mechanism by which VSMC derived EVs contribute to vascular calcification and result in novel targets for treatment.
In response to calcified ECM, neighboring ECs and VSMCs react and induce the production of osteogenic factors, such as bone morphogenetic protein 2 (BMP-2) and 4 (BMP-4) [128,129]. Besides this expression of bone-associated proteins, the number of VSMCs decreases with age, and an increase of collagen-to-elastin ratio further increases the stiffening of the vessel wall [130].
In hypertension, calcium handling is disturbed, which is associated with an increased activation of L-type calcium channels and sensitivity of (hypertensive) patients toward calcium channel blockers [131]. Increased intracellular calcium induces the activation of receptors coupled to phospholipase C, leading to the generation of second messengers that trigger cytokines, ROS and miRNAs, and also cellular derived EVs [132,133]. Additionally, increased intracellular calcium activates the contractile machinery of VSMCs, leading to hyper-contractility. Excessive intracellular calcium rapidly disintegrates both mitochondria and structural components of VSMCs, and results in calcium depositions within elastic fibers [130]. Consequently, elasticity of the vasculature decreases, further contributing to increased blood pressure [24,134] and VSMC stiffening [135,136]. Specific calcium antagonists, blocking L-type voltage-dependent calcium channels, reduce calcium internalization and normalize blood pressure [137,138,139]. Of note, these calcium channel blockers also prevent calcification of the vasculature [140].

3.1.3. Consequences of Vascular Calcification

Vascular calcification is associated with arterial stiffness [32,141]. In chronic kidney disease, medial calcification of large arteries is directly responsible for the increased stiffness (or reduced distensibility) [142,143]. The presence of vascular calcification (process) in the vessel wall contributes to ECM degradation and wall thickening [49,144]. Taken together, vascular calcification contributes directly and indirectly to arterial stiffening and, ultimately, hypertension [145,146]. Atherosclerosis has also been associated with arterial stiffness and hypertension [147,148]. Additionally, increased blood pressure and wall shear stress increase vulnerability and, consequently, atherosclerotic plaques become prone to rupture [30,149].

3.2. Atherosclerosis

Atherosclerosis is characterized by intimal plaque build-up and often referred to as a thickening of the vessel wall, ultimately narrowing the lumen of the vessel. Atherosclerosis is considered an inflammatory disease, starting with endothelial stress and subsequent monocyte infiltration [150,151]. Clinically, plaques may rupture, resulting in stroke or myocardial infarction [104]. Besides the classical view that systemic factors are causative for atherosclerosis, recent data suggest that local vascular processes also initiate atherogenesis [104]. Lineage tracing studies revealed that many of the cells present in atherosclerotic plaques are derived from VSMC precursors [30]. Additionally, it is now recognized that atherosclerotic plaque macrophages have lineage traces of VSMCs, indicating that cells in the intima originate from the vascular media, and not necessarily from the circulating mesenchymal cells that infiltrate the vessel wall at sites of injury [152].
Arterial remodeling is considered key to the development of atherosclerosis [148,153]. Arterial remodeling of the vessel wall can be initiated via many processes, including oxidative stress [2], proliferation [58], VSMC phenotype switching [154], cell infiltration, apoptosis and calcification [155,156,157].

3.3. Intimal and Medial Aspects of Vascular Calcification

Vascular calcification can be found at two anatomical locations, the medial and intimal layer. In the 1940s, post-mortem analysis of aortic specimens from CVD patients revealed that atherosclerosis is only present at vascular areas with either medial degradation or medial calcification [158,159,160,161]. Histological examination of aortas showed that medial calcification preceded atherosclerotic plaque build-up. Intimal calcification is seen as a hallmark of atherosclerosis and, clinically, is used as a marker for atherosclerotic burden [162]. For a long time, calcification has been considered as a passive degenerative detrimental process, not amendable for intervention. This view has recently changed, since statin treatment is associated with both increased plaque stability and increased plaque calcification [163,164,165]. However, the apparent protective role of calcification in stabilizing plaque is in contrast with the predictive value of calcification for CVD morbidity and mortality [160,164]. Moreover, medial calcification has been shown to change blood dynamics by inducing arterial remodeling processes and increasing arterial stiffness [166,167,168]. Medial calcification strongly correlates with the phenotypical switching of VSMCs and a rise in blood pressure. Macrocalcifications in patients who underwent a computed tomography (CT) scan were shown to have a three- to four-fold risk of developing fatal cardiovascular events [169]. Additionally, microcalcifications, which are precursors of macrocalcification, have been shown to induce plaque vulnerability [170].

Cellular Processes of Vascular Calcification

Specific stimuli, such as elevated calcium or phosphate levels, induce the switching of VSMCs to an osteogenic phenotypic where the cells acquire features of chondrocytes and osteoblasts [24]. Osteogenic VSMCs show an increased expression of osteogenic markers, such as alkaline phosphatase, BMP-2 and runt-related transcription factor 2 (Runx2), but show decreased calcification-inhibitor protein expression [171]. VSMCs exposed to elevated calcium levels display an intracellular calcium overload that may induce microcalcifications, eventually resulting in macrocalcification, which contributes to vascular stiffness and hypertension [172]. The process of calcification, culminating in either micro- or macrocalcification, compromises the structural integrity of the vessel wall and, hence, its functional properties.
Oxidative stress is an important player in the development of vascular calcification. Oxidative stress pathways in VSMCs have been linked to development aortic valve calcification [173]. Increased oxidative stress induces expression of Runx2, a key transcription factor associated with osteoblast differentiation [174]. VSMC-specific Runx2 knockout was shown to decrease vascular calcification and atherosclerosis development [175]. Additionally, increased H2O2 levels have been found in the close vicinity of calcification nucleation foci, together with an enhanced expression of oxidases [173]. High phosphate levels, present in chronic kidney disease, are associated with extent of vascular calcification [176]. In vitro, phosphate has been shown to induce mitochondrial-derived free radicals, by enhancing mitochondrial membrane potential [177]. Antioxidants inhibit phosphate-induced ROS production [177]. Increased oxidative stress is also found in diabetic patients [178], leading to an increased accumulation of advanced glycation end products (AGEs). AGEs have been shown to induce vascular calcification via increased oxidative stress of VSMC [179,180].

3.4. Fibrosis

Fibrosis of different organs contributes to up to 50% of worldwide mortality [181,182]. In the vasculature, myofibroblasts produce ECM proteins such as collagen and fibronectin [12]. Myofibroblasts are derived from MSC-like cells that reside in the vessel wall. The role of these MSC-like cells in the vessel wall is not fully understood, and is believed to play an important role in structural vessel repair. In vascular fibrosis, excessive collagen deposition leads to a reduced compliance of the vessel, which indicates increased arterial stiffness. Furthermore, high blood pressure induces structural changes including hypertrophy of the vessel wall [183,184] and cellular [185] and ECM remodeling [186].
Lineage tracing studies have shown that resident Gli1+ MSC-like cells are a major contributor of vascular fibrosis in arterial remodeling [187]. Acute vessel wall injury revealed that a significant number of Gli1+ cells were detected in the media and neointima, and that they express VSMC markers such as αSMA and CNN1. Additionally, some 50% of the newly formed VSMCs were derived from Gli+ progenitors, indicating the involvement of adventitial cells in arterial remodeling [188]. Likewise, angiotensin II induced hypertension was caused by an exuberant production of collagen in the adventitial layer [189]. VSMCs that migrate towards the adventitia start expressing stem cell markers Sca-1 and CD34 [186]. These “adventitial stem cells” change towards fibroblasts and deposit collagen and other ECM, contributing to adventitial fibrosis [190,191].
During atherosclerosis development, fate-tracing studies in mice have shown that MSC-like cells migrate into the media and neo-intima. Also, MSC-like Gli1+ cells were a major source of osteoblasts-like cells, significantly contributing to the process of vascular calcification in both vascular media and intima. The genetic ablation of Gli+ cells before injury dramatically reduced the severity of vascular calcification [192]. These data indicate that fibrotic processes are regulated by myofibroblasts and MSC-like cells and that these are important in the development of atherosclerosis and vascular calcification.

4. Assessing Vascular Remodeling and Disease

4.1. Pulse Wave Velocity (PWV), Distensibility Coefficient (DC) and Intima-Media Thickness (IMT)

Carotid-femoral PWV is considered the gold standard for assessing central arterial stiffness [193,194]. PWV, however, only measures vascular characteristics of the aorta and does not harbor information on any other vessels. Multi-slice CT, measuring calcification, reflects the atherosclerotic burden, and IMT measures atherosclerosis in the carotid arteries. Below, we describe in more detail measures of vascular remodeling and disease.

4.1.1. Pulse Wave Velocity

Measuring arterial stiffness in the clinic is challenging. The methods that are available for measuring arterial stiffness are non-invasive vascular imaging, such as ultrasound and magnetic resonance imaging (MRI), and high-fidelity recording of the pulse wave, performed by tonometry or ultrasound Doppler. Functional measures that can be obtained are pulse wave velocity (PWV) and the distensibility coefficient (DC). In the clinic, carotid–femoral PWV (cfPWV) adds to existing risk scores in cardiovascular risk management [195], while arterial stiffening has been shown to be independently associated with cardiovascular risk and mortality [16]. Brachial-ankle PWV is also used as an alternative to cfPWV, correlating reasonably well [16].
The interpretation of arterial stiffness findings in human studies is subject to (1) the stiffness measure/method used and (2) the mechanistic or constitutive focus of the study. Constitutive properties that are often considered to interpret arterial stiffness findings are those related to the ECM or VSMCs [196,197].

4.1.2. Distensibility Coefficient

Arterial stiffness can be determined by measuring local vessel diameter and distension by ultrasound or MRI [198]. The measured data can be used to calculate the local distensibility coefficient (DC) or compliance. PWV, as well as DC, are well known to be pressure dependent and require blood pressure adjustment [199]. Adjustment is valid when considering group analyses, but lacks applicability in the use of individual cases [32,200].
PWV is largely dependent on the elastic properties of the large arteries. Therefore, PWV should be interpreted with care, since it only reflects the stiffness of the aorta. In conduit arteries, the relative amount of VSMCs are limited and, thus, the contribution of ECM to arterial stiffening may be expected to be more pronounced. In peripheral, more muscular, arteries, age-related stiffening is not as straightforward [201,202]. PWV in the lower extremities can change significantly, which may explain why no evident relation between PWV and age has been reported [203]. Additionally, hypertrophy of VSMCs is commonly observed in hypertension [204], with the potential to (partially) determine PWV [205]. Thus, PWV is an important clinical marker for risk assessment, as it correlates with cardiovascular risk, mortality, and organ damage of the heart, kidney and brains [206,207].
Techniques for measuring PWV are developing. cfPWV is most commonly used for aortic stiffness, but is susceptible to local wave reflections and not suitable for obtaining information on carotid arteries [208]. Ultrasound ultrafast imaging techniques are being explored to directly measure the propagation of the pulse wave, with limited success for the common carotid artery [209]. Elastography is a promising technique, as it measures the propagation of shear waves in the tissue [210]. The propagation of the shear wave is directly proportional to the elastic modulus of the tissue [16]. PWV can also be measured using MRI, which also allows for segmentation. Its drawbacks are costs and long scanning times [33]. Currently, the available PWV toolbox can only be applied to large and medium-sized arteries, but techniques are developing for smaller vessels.
At present, the interpretation of stiffness measurements in terms of arterial wall constitutive properties remains a challenge [187]. Medial calcifications are known to co-localize with elastin fragmentation spots, but the mechanical contribution of the calcific deposits to vessel stiffness remains to be established; the elastin degradation alone could be sufficient to explain stiffening [211].

4.1.3. Intima-Media Thickness

IMT is used as a measurement for assessing the thickness of the tunica intima and media (estimated from lumen-intima and media-adventitia echo complexes). IMT is often measured using ultrasound and can be readily obtained in superficial arteries. One aspect that is not measured by IMT is adventitial thickness is a significant contributor of pathology development [212]. Wall thickness data are important in determining arterial wall material stiffness and wall stress. But, IMT can also be used to detect atherosclerosis, and is used to follow progression of the disease [213,214]. IMT is shown to be an independent predictor of overall cardiovascular events, including myocardial infarction and stroke [215]. Multiple studies have shown that increases in IMT can be significantly delayed using effective cardiovascular drugs, such as statins [216,217]. However, further observational studies revealed that this delay did not result in reduced cardiovascular events [218].
To study large populations, non-invasive techniques such as IMT are preferred over invasive measurement tools. As such, IMT provides a reliable tool for assessing vessel wall macro-structural changes. However, some limitations for IMTs exist. There is a lack of standardized protocols for the use of IMT, and attempts to establish them have not reached consensus yet [219]. IMTs of certain segments, such as the common carotid artery, are related to hypertension, blood pressure and vascular hypertrophy [220], whereas the internal carotid artery correlates with atherosclerotic plaques [221,222]. Taken together, additional risk assessments, such as plaque presence and stenosis severity, will be complementary to the use of IMT alone. A novel technique to determine extra-media thickness may have great potential in combination with IMT to discriminate between the medial and adventitial layer which aids in risk assessment [223,224]. Moreover, other imaging techniques, such as coronary artery calcium (CAC) scanning, have been demonstrated to be superior for predicting CVD events [155,225].

4.2. Calcification Imaging by Computed Tomography

Calcification imaging in the clinic is mostly based on CT, which is an angiography technique based on X-ray radiation and makes use of computer processing to create a detailed 3D image. Calcification scores, such as the coronary artery calcium (CAC) score, can be derived from CT scans and, in recent decades, this has been used as a measure of atherosclerotic burden. The Agatson score is the most frequently used method to calculate CAC. The Agatston score is defined as a weighed sum of the calcification area, with density given as Hounsfield Units [226]. CAC represents an accurate measure to assess atherosclerotic burden [227,228]. Additionally, CAC correlates with traditional risk factors for atherosclerosis, such as hypertension, hypercholesterolemia and diabetes [227,228]. CAC also associates with cardiovascular morbidities and mortalities, including cardiac death and stroke [229].
CAC is regarded as the best clinical predictor for the accumulation and progression of vascular calcification over time [230,231,232]. CAC can be used to identify high-risk individuals that need immediate medical attention or intervention [233]. Interventions to slow down or even regress calcification, using the supplementation of vitamin K, are currently being conducted [234]. High dietary intake of vitamin K is associated with a lower risk of coronary heart disease [235,236,237] and supplementation with vitamin K significantly delays the progression of calcification [238]. Conversely, statin treatment is known for its beneficial cardiovascular effects, however, it is also shown to increase CAC score [239]. This indicates that type of calcification, as well as the volume or density of calcification, determines whether it is plaque-stabilizing or plaque-destabilizing. Indeed, CAC volume is positively and independently associated with CVD, whereas CAC density was inversely correlated with CVD risk [240].
Measuring CAC as a clinical marker for CVD has some limitations. Firstly, CAC is limited to coronary arteries, which do not represent the calcification of other vessels of the arterial system. CAC does not represent total plaque size, as histopathological studies showed that the area of calcium deposition was much smaller than total plaque volume [241,242]. Additionally, CAC score poorly correlated with luminal narrowing and severity [217,243,244]. Secondly, distinction between medial and intimal calcification is not possible with current resolutions of CT. Intimal and medial calcification are different pathologies, with different risk factors for developing CVD [245]. Additionally, microcalcification (< 15 μm) cannot be assessed, as the resolution of CT is some 200 μm [246].
Thirdly, evidence suggests that atherosclerotic plaques that have denser calcification have smaller lipid cores, with increased stability of plaques, resulting in lower CVD risk [247]. Greater CAC volume, however, is associated with a higher risk of CVD [248]. This indicates that future research should integrate density and volume into CAC scoring. Finally, CAC involves exposure to radiation, which limits the use of calcium scores as a follow-up on atherosclerosis progression [249].

4.3. 18F-NaF Positron Emission Tomography

18F-NaF PET scanning gained recent attention as a novel imaging modality for vascular calcification [250]. In bone, 18F-NaF is incorporated into newly formed hydroxyapatite crystals by exchanging fluoride with the hydroxyl groups [251]. In the vasculature, 18F-NaF PET imaging has been reported to identify active (micro)calcification with high specificity [252]. Importantly, CT and echocardiography cannot identify active vascular calcification, which is considered the main driver of disease progression [253]. Assessing the molecular bioactivity of calcification in atherosclerotic plaques is of additive value to identify vulnerable plaques [254,255]. Additionally, ex vivo histological studies showed that the resolution of 18F-NaF has the potential to discriminate between micro-and macrocalcification [254]. Taken together, 18F-NaF uptake provides complementary information to CT, as it allows for the detection of ongoing active calcification of plaques, which is a major improvement to the prediction of future cardiovascular risk. Limitations of 18F-NaF PET imaging are the relatively high costs and exposure to radiation, but, also, 18F-NaF PET imaging lacks the resolution to discriminate between medial and intimal calcification.

5. Conclusions

Arterial remodeling is defined as the adaptation of the vessel wall away from a normal homeostatic state towards pathologic development, triggered by chronic exposure to stress signals. Different arterial remodeling processes have been described, such as inflammation, oxidative stress, lipid accumulation, and degradation of the ECM. Many of these are regulated by VSMCs and affect vessel wall morphology and properties. VSMCs exhibit cellular plasticity and, upon stress stimuli, they may differentiate towards synthetic, macrophage-like and osteogenic VSMCs. As a consequence, VSMCs initiate and support remodeling processes involving ECM synthesis and cell proliferation, migration and contraction, which aid in maintaining tissue functionality. Disruption of VSMC phenotypic plasticity has many pathological consequences, including the development of hypertension, atherosclerosis, aneurysm formation, intimal and medial calcification, and vascular fibrosis, ultimately leading to increased CVD morbidity and mortality. Early detection of vascular remodeling is key in the prevention and prognosis of CVD. Clinically, assessment of calcification-mediated remodeling remains difficult. Assessment tools include measurements of pulse wave velocity, distensibility coefficient and intima-media thickness. These techniques may capture changes in dynamic and geometric vessel wall properties, but cannot directly monitor the underlying arterial remodeling processes. The addition of techniques, such as 18F-NaF PET and CT imaging may provide further mechanistic insight into vascular calcification development and other VSMC-mediated processes.
Combining expertise in vascular biology, clinical vascular expertise and multi-scale/multi-modality imaging should advance the understanding and identification of critical determinants of vascular remodeling, ultimately resulting in tailor-made diagnoses and treatments.

Author Contributions

Conceptualization, A.J. and L.S.; writing—original draft preparation, A.J., K.R., A.K. and L.S.; writing—review and editing, A.J., K.R., A.K. and L.S.; visualization, A.J. and L.S.; supervision, L.S.; project administration, L.S.; funding acquisition, L.S.

Funding

This research was funded from the European Union’s Horizon 2020 research and innovation programmes under the Marie Sklodowska-Curie grant agreement No 675111, 722609 and 764474.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

α-SMAalpha-smooth muscle actin
AGEadvanced glycation end product
BMP-2bone morphogenetic protein 2
BMP-4bone morphogenetic protein 4
CACcoronary artery calcium
CD9cluster of differentiation 9
CD34cluster of differentiation 34
CD63cluster of differentiation 63
CD81cluster of differentiation 81
cfPWVcarotid-femoral pulse wave velocity
CNN-1calponin-1
CTcomputed tomography
CVcardiovascular
CVDcardiovascular disease
DBPdiastolic blood pressure
DCdistensibility coefficient
ECendothelial cell
ECMextracellular matrix
EVextracellular vesicle
Gli1glioma-associated oncogene
IL-1βinterleukin 1 beta
IMTintima-media thickness
KLF4kruppel-like factor 4
MMPmetalloproteinase
MRImagnetic resonance imaging
MSCmesenchymal stem cell
MYC11smooth muscle-myosin heavy chain
NAD(P)Hnitrate reductase
NOnitric oxide
Oct4octamer-binding transcription factor 4
p16INK4acyclin-dependent kinase inhibitor 2A
p21cyclin-dependent kinase inhibitor 1
PARprotease activated receptor
PF4platelet factor 4
PDGFplatelet derived growth factor
PXEpseudoxanthoma elasicum
PWVpulse wave velocity
ROSreactive oxygen species
Runx2runt-related transcription factor 2
SBPsystolic blood pressure
Sca-1Spinocerebellaire ataxie type 1
SM22-αsmooth muscle 22-alpha
SMPD3sphingomyelin phosphodiesterase 3
smtnSmoothelin
VSMCvascular smooth muscle cell

References

  1. Mazurek, R.; Dave, J.M.; Chandran, R.R.; Misra, A.; Sheikh, A.Q.; Greif, D.M. Vascular Cells in Blood Vessel Wall Development and Disease. Adv. Pharmacol. 2017, 78, 323–350. [Google Scholar] [PubMed]
  2. Schurgers, L.J.; Akbulut, A.C.; Kaczor, D.M.; Halder, M.; Koenen, R.R.; Kramann, R. Initiation and Propagation of Vascular Calcification Is Regulated by a Concert of Platelet- and Smooth Muscle Cell-Derived Extracellular Vesicles. Front. Cardiovasc. Med. 2018, 5, 36. [Google Scholar] [CrossRef] [PubMed]
  3. Roostalu, U.; Wong, J.K. Arterial smooth muscle dynamics in development and repair. Dev. Biol. 2018, 435, 109–121. [Google Scholar] [CrossRef] [PubMed]
  4. Hao, H.; Gabbiani, G.; Bochaton-Piallat, M.L. Arterial smooth muscle cell heterogeneity: Implications for atherosclerosis and restenosis development. Arterioscler. Thromb. Vasc. Biol. 2003, 23, 1510–1520. [Google Scholar] [CrossRef]
  5. Alexander, M.R.; Owens, G.K. Epigenetic control of smooth muscle cell differentiation and phenotypic switching in vascular development and disease. Annu. Rev. Physiol. 2012, 74, 13–40. [Google Scholar] [CrossRef]
  6. van Bussel, F.C.; van Bussel, B.C.; Hoeks, A.P.; Op’t Roodt, J.; Henry, R.M.; Ferreira, I.; Vanmolkot, F.H.; Schalkwijk, C.G.; Stehouwer, C.D.; Reesink, K.D. A control systems approach to quantify wall shear stress normalization by flow-mediated dilation in the brachial artery. PLoS ONE 2015, 10, e0115977. [Google Scholar] [CrossRef]
  7. van der Heijden, O.W.; Essers, Y.P.; Fazzi, G.; Peeters, L.L.; De Mey, J.G.; van Eys, G.J.J. Uterine artery remodeling and reproductive performance are impaired in endothelial nitric oxide synthase-deficient mice. Biol. Reprod. 2005, 72, 1161–1168. [Google Scholar] [CrossRef]
  8. Boutouyrie, P.; Laurent, S.; Girerd, X.; Benetos, A.; Lacolley, P.; Abergel, E.; Safar, M. Common carotid artery stiffness and patterns of left ventricular hypertrophy in hypertensive patients. Hypertension 1995, 25, 651–659. [Google Scholar] [CrossRef]
  9. Rourke, M.F.; Nichols, W.W. Aortic Diameter, Aortic Stiffness, and Wave Reflection Increase With Age and Isolated Systolic Hypertension. Hypertension 2005, 45, 652–658. [Google Scholar]
  10. Libby, P. Inflammation in atherosclerosis. Nature 2002, 420, 868–874. [Google Scholar] [CrossRef]
  11. Henning, R.J.; Bourgeois, M.; Harbison, R.D. Poly(ADP-ribose) Polymerase (PARP) and PARP Inhibitors: Mechanisms of Action and Role in Cardiovascular Disorders. Cardiovasc. Toxicol. 2018, 18, 493–506. [Google Scholar] [CrossRef] [PubMed]
  12. van Varik, B.J.; Rennenberg, R.J.M.W.; Reutelingsperger, C.P.; Kroon, A.A.; de Leeuw, P.W.; Schurgers, L.J. Mechanisms of arterial remodeling: Lessons from genetic diseases. Front. Genet. 2012, 3, 290. [Google Scholar] [CrossRef] [PubMed]
  13. Lacolley, P.; Regnault, V.; Segers, P.; Laurent, S. Vascular Smooth Muscle Cells and Arterial Stiffening: Relevance in Development, Aging, and Disease. Physiol. Rev. 2017, 97, 1555–1617. [Google Scholar] [CrossRef] [PubMed]
  14. Owens, G.K.; Kumar, M.S.; Wamhoff, B.R. Molecular Regulation of Vascular Smooth Muscle Cell Differentiation in Development and Disease. Physiol. Rev. 2004, 84, 767–801. [Google Scholar] [CrossRef] [PubMed]
  15. Willis, A.I.; Pierre-Paul, D.; Sumpio, B.E.; Gahtan, V. Vascular smooth muscle cell migration: Current research and clinical implications. Vasc. Endovasc. Surg. 2004, 38, 11–23. [Google Scholar] [CrossRef]
  16. Johnson, J.L. Matrix metalloproteinases: Influence on smooth muscle cells and atherosclerotic plaque stability. Expert Rev. Cardiovasc. Ther. 2014, 5, 265–282. [Google Scholar] [CrossRef]
  17. Peeters, S.A.; Engelen, L.; Buijs, J.; Chaturvedi, N.; Fuller, J.H.; Schalkwijk, C.G.; Stehouwer, C.D.; Group, E.P.C.S. Plasma levels of matrix metalloproteinase-2, -3, -10, and tissue inhibitor of metalloproteinase-1 are associated with vascular complications in patients with type 1 diabetes: The EURODIAB Prospective Complications Study. Cardiovasc. Diabetol. 2015, 14, 31. [Google Scholar] [CrossRef]
  18. Reesink, K.D.; Spronck, B. Constitutive interpretation of arterial stiffness in clinical studies: A methodological review. Am. J. Physiol. Heart Circ. Physiol. 2019, 316, H693–H709. [Google Scholar] [CrossRef]
  19. Smulyan, H.; Mookherjee, S.; Safar, M.E. The two faces of hypertension: Role of aortic stiffness. J. Am. Soc. Hypertens. 2016, 10, 175–183. [Google Scholar] [CrossRef]
  20. Lacolley, P.; Regnault, V.; Nicoletti, A.; Li, Z.; Michel, J.B. The vascular smooth muscle cell in arterial pathology: A cell that can take on multiple roles. Cardiovasc. Res. 2012, 95, 194–204. [Google Scholar] [CrossRef]
  21. Petsophonsakul, P.; Furmanik, M.; Forsythe, R.; Dweck, M.; Schurink, G.W.; Natour, E.; Reutelingsperger, C.; Jacobs, M.; Mees, B.; Schurgers, L. Role of Vascular Smooth Muscle Cell Phenotypic Switching and Calcification in Aortic Aneurysm Formation. Arterioscler. Thromb. Vasc. Biol. 2019, 27, 1151–1368. [Google Scholar] [CrossRef] [PubMed]
  22. Pawade, T.A.; Newby, D.E.; Dweck, M.R. Calcification in Aortic Stenosis: The Skeleton Key. J. Am. Coll. Cardiol. 2015, 66, 561–577. [Google Scholar] [CrossRef] [PubMed]
  23. Neven, E.; Haese, P.C. Vascular calcification in chronic renal failure: What have we learned from animal studies? Circ. Res. 2011, 108, 249–264. [Google Scholar] [CrossRef] [PubMed]
  24. Proudfoot, D. Molecular mechanisms of arterial calcification. Artery Res. 2009, 3, 128–131. [Google Scholar] [CrossRef]
  25. Tyson, K.L.; Reynolds, J.L.; McNair, R.; Zhang, Q.; Weissberg, P.L.; Shanahan, C.M. Osteo/chondrocytic transcription factors and their target genes exhibit distinct patterns of expression in human arterial calcification. Arterioscler. Thromb. Vasc. Biol. 2003, 23, 489–494. [Google Scholar] [CrossRef] [PubMed]
  26. Fleckenstein-Grün, G.; Frey, M.; Thimm, F.; Hofgärtner, W.; Fleckenstein, A. Calcium Overload—An Important Cellular Mechanism in Hypertension and Arteriosclerosis. Drugs 1992, 44, 23–30. [Google Scholar] [CrossRef]
  27. Shanahan, C.M. Mechanisms of vascular calcification in CKD-evidence for premature ageing? Nat. Rev. Nephrol. 2013, 9, 661–670. [Google Scholar] [CrossRef]
  28. O’Rourke, M.F.; Hashimoto, J. Mechanical Factors in Arterial Aging: A Clinical Perspective. J. Am. Coll. Cardiol. 2007, 50, 1–13. [Google Scholar] [CrossRef]
  29. Mulvany, M.J.; Baumbach, G.L.; Aalkjaer, C.; Heagerty, A.M.; Korsgaard, N.; Schiffrin, E.L.; Heistad, D.D. Vascular remodeling. Hypertension 1996, 28, 505–506. [Google Scholar]
  30. Mulvany, M.J. Small artery remodelling in hypertension: Causes, consequences and therapeutic implications. Med. Biol. Eng. Comp. 2008, 46, 461–467. [Google Scholar] [CrossRef]
  31. Ward, M.R.; Pasterkamp, G.; Yeung, A.C.; Borst, C. Arterial remodeling. Mechanisms and clinical implications. Circulation 2000, 102, 1186–1191. [Google Scholar] [CrossRef]
  32. Hansson, G.K.; Libby, P. The immune response in atherosclerosis: A double-edged sword. Nat. Rev. Immunol. 2006, 6, 508–519. [Google Scholar] [CrossRef]
  33. Humphrey, J.D.; Dufresne, E.R.; Schwartz, M.A. Mechanotransduction and extracellular matrix homeostasis. Nat. Rev. Mol. Cell Biol. 2014, 15, 802–812. [Google Scholar] [CrossRef] [Green Version]
  34. Greenwald, S.E. Ageing of the conduit arteries. J. Pathol. 2007, 211, 157–172. [Google Scholar] [CrossRef]
  35. Spronck, B.; Heusinkveld, M.H.G.; Donders, W.P.; de Lepper, A.G.W.; O’Roodt, J.; Kroon, A.A.; Delhaas, T.; Reesink, K.D. A constitutive modeling interpretation of the relationship among carotid artery stiffness, blood pressure, and age in hypertensive subjects. Am. J. Physiol. Heart Circ. Physiol. 2015, 308, H568–H582. [Google Scholar] [CrossRef] [Green Version]
  36. Humphrey, J.D.; Harrison, D.G.; Figueroa, C.A.; Lacolley, P.; Laurent, S. Central Artery Stiffness in Hypertension and Aging: A Problem with Cause and Consequence. Circ. Res. 2016, 118, 379–381. [Google Scholar] [CrossRef]
  37. Sekikawa, A.; Shin, C.; Curb, J.D.; Barinas-Mitchell, E.; Masaki, K.; El-Saed, A.; Seto, T.B.; Mackey, R.H.; Choo, J.; Fujiyoshi, A.; et al. Aortic stiffness and calcification in men in a population-based international study. Atherosclerosis 2012, 222, 473–477. [Google Scholar] [CrossRef] [Green Version]
  38. Kruger, T.; Oelenberg, S.; Kaesler, N.; Schurgers, L.J.; van de Sandt, A.M.; Boor, P.; Schlieper, G.; Brandenburg, V.M.; Fekete, B.C.; Veulemans, V.; et al. Warfarin Induces Cardiovascular Damage in Mice. Arterioscler. Thromb. Vasc. Biol. 2013, 33, 2618–2624. [Google Scholar] [CrossRef]
  39. Chi, C.; Li, D.J.; Jiang, Y.J.; Tong, J.; Fu, H.; Wu, Y.H.; Shen, F.M. Vascular smooth muscle cell senescence and age-related diseases: State of the art. Mol. Basis Dis. 2019, 1865, 1810–1821. [Google Scholar] [CrossRef]
  40. Sans, M.; Moragas, A. Mathematical morphologic analysis of the aortic medial structure. Biomechanical implications. Anal. Quant. Cytol. Histol. 1993, 15, 93–100. [Google Scholar]
  41. Cliff, W.J. The aortic tunica media in aging rats. Exp. Mol. Pathol. 1970, 13, 172–189. [Google Scholar] [CrossRef]
  42. Zieman, S.J.; Kass, D.A. Advanced Glycation Endproduct Crosslinking in the Cardiovascular System. Drugs 2004, 64, 459–470. [Google Scholar] [CrossRef]
  43. Wang, J.C.; Bennett, M. Aging and atherosclerosis: Mechanisms, functional consequences, and potential therapeutics for cellular senescence. Circ. Res. 2012, 111, 245–259. [Google Scholar] [CrossRef] [Green Version]
  44. Leondes, C.T. Biomechanical Systems: Techniques and Applications, Volume II: Cardiovascular Techniques; CRC Press: Boca Raton, FL, USA, 2000; pp. 6:1–6:61. [Google Scholar]
  45. Kelly, R.; Hayward, C.; Avolio, A.; O’Rourke, M. Noninvasive determination of age-related changes in the human arterial pulse. Circulation 1989, 80, 1652–1659. [Google Scholar] [CrossRef] [Green Version]
  46. Laogun, A.A.; Gosling, R.G. In vivo arterial compliance in man. Clin. Phys. Physiol. Meas. 1982, 3, 201–212. [Google Scholar] [CrossRef]
  47. Zieman, S.J.; Melenovsky, V.; Kass, D.A. Mechanisms, Pathophysiology, and Therapy of Arterial Stiffness. Arterioscler. Thromb. Vasc. Biol. 2005, 25, 932–943. [Google Scholar] [CrossRef] [Green Version]
  48. Cattell, M.A.; Anderson, J.C.; Hasleton, P.S. Age-related changes in amounts and concentrations of collagen and elastin in normotensive human thoracic aorta. Clin. Chim. Acta 1996, 245, 73–84. [Google Scholar] [CrossRef]
  49. Sun, Z. Aging, arterial stiffness, and hypertension. Hypertension 2015, 65, 252–256. [Google Scholar] [CrossRef] [Green Version]
  50. Brandes, R.P.; Fleming, I.; Busse, R. Endothelial aging. Cardiovasc. Res. 2005, 66, 286–294. [Google Scholar] [CrossRef] [Green Version]
  51. Urschel, K.; Cicha, I.; Daniel, W.G.; Garlichs, C.D. Shear stress patterns affect the secreted chemokine profile in endothelial cells. Clin. Hemorheol. Microcirc. 2012, 50, 143–152. [Google Scholar]
  52. Qiu, J.; Zheng, Y.; Hu, J.; Liao, D.; Gregersen, H.; Deng, X.; Fan, Y.; Wang, G. Biomechanical regulation of vascular smooth muscle cell functions: From in vitro to in vivo understanding. J. R. Soc. Interface 2014, 11, 20130852. [Google Scholar] [CrossRef] [Green Version]
  53. Yu, H.; Clarke, M.C.H.; Figg, N.; Littlewood, T.D.; Bennett, M.R. Smooth muscle cell apoptosis promotes vessel remodeling and repair via activation of cell migration, proliferation, and collagen synthesis. Arterioscler. Thromb. Vasc. Biol. 2011, 31, 2402–2409. [Google Scholar] [CrossRef] [Green Version]
  54. Mozos, I.; Luca, C.T. Crosstalk between Oxidative and Nitrosative Stress and Arterial Stiffness. Curr. Vasc. Pharmacol. 2017, 15, 446–456. [Google Scholar] [CrossRef]
  55. McMaster, W.G.; Kirabo, A.; Madhur, M.S.; Harrison, D.G. Inflammation, immunity, and hypertensive end-organ damage. Circ. Res. 2015, 116, 1022–1033. [Google Scholar] [CrossRef]
  56. Jain, S.; Khera, R.; Corrales-Medina, V.F.; Townsend, R.R.; Chirinos, J.A. Inflammation and arterial stiffness in humans. Atherosclerosis 2014, 237, 381–390. [Google Scholar] [CrossRef]
  57. Lutgens, E.; de Muinck, E.D.; Kitslaar, P.J.; Tordoir, J.H.; Wellens, H.J.; Daemen, M.J. Biphasic pattern of cell turnover characterizes the progression from fatty streaks to ruptured human atherosclerotic plaques. Cardiovasc. Res. 1999, 41, 473–479. [Google Scholar] [CrossRef] [Green Version]
  58. Matthews, C.; Gorenne, I.; Scott, S.; Figg, N.; Kirkpatrick, P.; Ritchie, A.; Goddard, M.; Bennett, M. Vascular smooth muscle cells undergo telomere-based senescence in human atherosclerosis: Effects of telomerase and oxidative stress. Circ. Res. 2006, 99, 156–164. [Google Scholar] [CrossRef] [Green Version]
  59. Moon, S.K.; Cha, B.Y.; Kim, C.H. In vitro cellular aging is associated with enhanced proliferative capacity, G1 cell cycle modulation, and matrix metalloproteinase-9 regulation in mouse aortic smooth muscle cells. Arch. Biochem. Biophys. 2003, 418, 39–48. [Google Scholar] [CrossRef]
  60. Clarke, M.C.; Figg, N.; Maguire, J.J.; Davenport, A.P.; Goddard, M.; Littlewood, T.D.; Bennett, M.R. Apoptosis of vascular smooth muscle cells induces features of plaque vulnerability in atherosclerosis. Nat. Med. 2006, 12, 1075–1080. [Google Scholar] [CrossRef]
  61. Clarke, M.C.H.; Littlewood, T.D.; Figg, N.; Maguire, J.J.; Davenport, A.P.; Goddard, M.; Bennett, M.R. Chronic apoptosis of vascular smooth muscle cells accelerates atherosclerosis and promotes calcification and medial degeneration. Circ. Res. 2008, 102, 1529–1538. [Google Scholar] [CrossRef] [Green Version]
  62. Bennett, M.R.; Gibson, D.F.; Schwartz, S.M.; Tait, J.F. Binding and phagocytosis of apoptotic vascular smooth muscle cells is mediated in part by exposure of phosphatidylserine. Circ. Res. 1995, 77, 1136–1142. [Google Scholar] [CrossRef] [PubMed]
  63. Shi, G.; Morrell, C.N. Platelets as initiators and mediators of inflammation at the vessel wall. Thromb. Res. 2011, 127, 387–390. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Vajen, T.; Benedikter, B.J.; Heinzmann, A.C.A.; Vasina, E.M.; Henskens, Y.; Parsons, M.; Maguire, P.B.; Stassen, F.R.; Heemskerk, J.W.M.; Schurgers, L.J.; et al. Platelet extracellular vesicles induce a pro-inflammatory smooth muscle cell phenotype. J. Extracell. Vesicles 2017, 6, 1322454. [Google Scholar] [CrossRef] [PubMed]
  65. Shi, G.; Field, D.J.; Long, X.; Mickelsen, D.; Ko, K.; Ture, S.; Korshunov, V.A.; Miano, J.M.; Morrell, C.N. Platelet factor 4 mediates vascular smooth muscle cell injury responses. Blood 2013, 121, 4417–4427. [Google Scholar] [CrossRef] [Green Version]
  66. Miano, J.M. Serum response factor: Toggling between disparate programs of gene expression. J. Mol. Cell. Cardiol. 2003, 35, 577–593. [Google Scholar] [CrossRef]
  67. Rensen, S.S.; Doevendans, P.A.; van Eys, G.J. Regulation and characteristics of vascular smooth muscle cell phenotypic diversity. Neth. Heart J. 2007, 15, 100–108. [Google Scholar] [CrossRef] [Green Version]
  68. Bochaton-Piallat, M.-L.; Ropraz, P.; Gabbiani, F.; Gabbiani, G. Phenotypic Heterogeneity of Rat Arterial Smooth Muscle Cell Clones. Arterioscler. Thromb. Vasc. Biol. 1996, 16, 815–820. [Google Scholar] [CrossRef]
  69. Hao, H.; Ropraz, P.; Verin, V.; Camenzind, E.; Geinoz, A.; Pepper, M.S.; Gabbiani, G.; Bochaton-Piallat, M.-L. Heterogeneity of Smooth Muscle Cell Populations Cultured From Pig Coronary Artery. Arterioscler. Thromb. Vasc. Biol. 2002, 22, 1093–1099. [Google Scholar] [CrossRef] [Green Version]
  70. Li, S.; Fan, Y.S.; Chow, L.H.; Van Den Diepstraten, C.; van Der Veer, E.; Sims, S.M.; Pickering, J.G. Innate diversity of adult human arterial smooth muscle cells: Cloning of distinct subtypes from the internal thoracic artery. Circ. Res. 2001, 89, 517–525. [Google Scholar] [CrossRef] [Green Version]
  71. Wanjare, M.; Kuo, F.; Gerecht, S. Derivation and maturation of synthetic and contractile vascular smooth muscle cells from human pluripotent stem cells. Cardiovasc. Res. 2013, 97, 321–330. [Google Scholar] [CrossRef] [Green Version]
  72. Gerthoffer, W.T. Mechanisms of vascular smooth muscle cell migration. Circ. Res. 2007, 100, 607–621. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Frismantiene, A.; Philippova, M.; Erne, P.; Resink, T.J. Smooth muscle cell-driven vascular diseases and molecular mechanisms of VSMC plasticity. Cell. Signal. 2018, 52, 48–64. [Google Scholar] [CrossRef] [PubMed]
  74. Allahverdian, S.; Chaabane, C.; Boukais, K.; Francis, G.A.; Bochaton-Piallat, M.L. Smooth muscle cell fate and plasticity in atherosclerosis. Cardiovasc. Res. 2018, 114, 540–550. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Schattemann, G.C.; Loushin, C.; Li, T.; Hart, C.E. PDGF-A is required for normal murine cardiovascular development. Dev. Biol. 1996, 176, 133–142. [Google Scholar] [CrossRef] [Green Version]
  76. Li, X.; van Putten, V.; Zarinetchi, F.; Nicks, E.M.; Thaler, S.; Heasley, E.L.; Nemenoff, A.R. Suppression of smooth-muscle α-actin expression by platelet-derived growth factor in vascular smooth-muscle cells involves Ras and cytosolic phospholipase A2. Biochem. J. 1997, 327, 709–716. [Google Scholar] [CrossRef] [Green Version]
  77. Li, J.Y.; Liu, C.P.; Shiao, W.C.; Jayakumar, T.; Li, Y.S.; Chang, N.C.; Huang, S.Y.; Hsieh, C.Y. Inhibitory effect of PDGF-BB and serum-stimulated responses in vascular smooth muscle cell proliferation by hinokitiol via up-regulation of p21 and p53. Arch. Med. Sci. 2018, 14, 579–587. [Google Scholar] [CrossRef] [Green Version]
  78. Ishigami, M.; Swertfeger, D.K.; Granholm, N.A.; Hui, D.Y. Apolipoprotein E inhibits platelet-derived growth factor-induced vascular smooth muscle cell migration and proliferation by suppressing signal transduction and preventing cell entry to G1 phase. J. Biol. Chem. 1998, 273, 20156–20161. [Google Scholar] [CrossRef] [Green Version]
  79. Kotani, M.; Fukuda, N.; Ando, H.; Hu, W.Y.; Kunimoto, S.; Saito, S.; Kanmatsuse, K. Chimeric DNA–RNA hammerhead ribozyme targeting PDGF A-chain mRNA specifically inhibits neointima formation in rat carotid artery after balloon injury. Cardiovasc. Res. 2003, 57, 265–276. [Google Scholar] [CrossRef] [Green Version]
  80. Deguchi, J.; Namba, T.; Hamada, H.; Nakaoka, T.; Abe, J.; Sato, O.; Miyata, T.; Makuuchi, M.; Kurokawa, K.; Takuwa, Y. Targeting endogenous platelet-derived growth factor B-chain by adenovirus-mediated gene transfer potently inhibits in vivo smooth muscle proliferation after arterial injury. Gene Ther. 1999, 6, 956–965. [Google Scholar] [CrossRef] [Green Version]
  81. Sanford, L.P.; Ormsby, I.; Gittenberger-de Groot, A.C.; Sariola, H.; Friedman, R.; Boivin, G.P.; Cardell, E.L.; Doetschman, T. TGFbeta2 knockout mice have multiple developmental defects that are non-overlapping with other TGFbeta knockout phenotypes. Development 1997, 124, 2659–2670. [Google Scholar]
  82. Hautmann, M.B.; Madsen, C.S.; Owens, G.K. A transforming growth factor beta (TGFbeta) control element drives TGFbeta-induced stimulation of smooth muscle alpha-actin gene expression in concert with two CArG elements. J. Biol. Chem. 1997, 272, 10948–10956. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Kapustin, A.N.; Chatrou, M.L.L.; Drozdov, I.; Zheng, Y.; Davidson, S.M.; Soong, D.; Furmanik, M.; Sanchis, P.; de Rosales, R.T.M.; Alvarez-Hernandez, D.; et al. Vascular smooth muscle cell calcification is mediated by regulated exosome secretion. Circ. Res. 2015, 116, 1312–1323. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Borissoff, J.I.; Spronk, H.M.H.; ten Cate, H. The hemostatic system as a modulator of atherosclerosis. N. Engl. J. Med. 2011, 364, 1746–1760. [Google Scholar] [CrossRef] [PubMed]
  85. Li, F.-F.; Shang, X.-K.; Du, X.-L.; Chen, S. Rapamycin Treatment Attenuates Angiotensin II -induced Abdominal Aortic Aneurysm Formation via VSMC Phenotypic Modulation and Down-regulation of ERK1/2 Activity. Curr. Med. Sci. 2018, 38, 93–100. [Google Scholar] [CrossRef] [PubMed]
  86. Jiang, F.; Yang, J.; Zhang, Y.; Dong, M.; Wang, S.; Zhang, Q.; Liu, F.F.; Zhang, K.; Zhang, C. Angiotensin-converting enzyme 2 and angiotensin 1–7: Novel therapeutic targets. Nat. Rev. Cardiol. 2014, 11, 413–426. [Google Scholar] [CrossRef]
  87. Moiseeva, E.P. Adhesion receptors of vascular smooth muscle cells and their functions. Cardiovasc. Res. 2001, 52, 372–386. [Google Scholar] [CrossRef]
  88. Zhang, J.; Wang, J.; Wei, Y.; Gao, C.; Chen, X.; Kong, W.; Kong, D.; Zhao, Q. ECM-mimetic heparin glycosamioglycan-functionalized surface favors constructing functional vascular smooth muscle tissue in vitro. Colloids Surf. B Biointerfaces 2016, 146, 280–288. [Google Scholar] [CrossRef]
  89. Sazonova, O.v.; Isenberg, B.C.; Herrmann, J.; Lee, K.L.; Purwada, A.; Valentine, A.D.; Buczek-Thomas, J.A.; Wong, J.Y.; Nugent, M.A. Extracellular matrix presentation modulates vascular smooth muscle cell mechanotransduction. Matrix Biol. 2015, 41, 36–43. [Google Scholar] [CrossRef]
  90. Finney, A.C.; Stokes, K.Y.; Pattillo, C.B.; Orr, A.W. Integrin signaling in atherosclerosis. Cell. Mol. Life Sci. 2017, 74, 2263–2282. [Google Scholar] [CrossRef]
  91. Ichii, T.; Koyama, H.; Tanaka, S.; Kim, S.; Shioi, A.; Okuno, Y.; Raines, E.W.; Iwao, H.; Otani, S.; Nishizawa, Y. Fibrillar collagen specifically regulates human vascular smooth muscle cell genes involved in cellular responses and the pericellular matrix environment. Circ. Res. 2001, 88, 460–467. [Google Scholar] [CrossRef] [Green Version]
  92. Chung, C.H.; Lin, K.T.; Chang, C.H.; Peng, H.C.; Huang, T.F. The integrin α2β1 agonist, aggretin, promotes proliferation and migration of VSMC through NF-kB translocation and PDGF production. Br. J. Pharmacol. 2009, 156, 846–856. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Thyberg, J.; Hultgårdh-Nilsson, A. Fibronectin and the basement membrane components laminin and collagen type IV influence the phenotypic properties of subcultured rat aortic smooth muscle cells differently. Cell Tissue Res. 1994, 276, 263–271. [Google Scholar] [CrossRef] [PubMed]
  94. O’Connor, W.N.; Davis, J.B.; Geissler, R.; Cottrill, C.M.; Noonan, J.A.; Todd, E.P. Supravalvular aortic stenosis. Clinical and pathologic observations in six patients. Arch. Pathol. Lab. Med. 1985, 109, 179–185. [Google Scholar]
  95. Bunton, T.E.; Biery, N.J.; Myers, L.; Gayraud, B.; Ramirez, F.; Dietz, H.C. Phenotypic alteration of vascular smooth muscle cells precedes elastolysis in a mouse model of Marfan syndrome. Circ. Res. 2001, 88, 37–43. [Google Scholar] [CrossRef] [Green Version]
  96. Bouvet, C.; Moreau, S.; Blanchette, J.; de Blois, D.; Moreau, P. Sequential activation of matrix metalloproteinase 9 and transforming growth factor beta in arterial elastocalcinosis. Arterioscler. Thromb. Vasc. Biol. 2008, 28, 856–862. [Google Scholar] [CrossRef]
  97. Pai, A.; Leaf, E.M.; El-Abbadi, M.; Giachelli, C.M. Elastin degradation and vascular smooth muscle cell phenotype change precede cell loss and arterial medial calcification in a uremic mouse model of chronic kidney disease. Am. J. Pathol. 2011, 178, 764–773. [Google Scholar] [CrossRef]
  98. Li, S.; Lao, J.; Benjamin, C.; Li, Y.S.; Zhao, Y.; Chu, J.; Chen, K.-D.; Tsou, T.C.; Peck, K.; Chien, S. Genomic analysis of smooth muscle cells in 3-dimensional collagen matrix1. FASEB J. 2002, 17, 97–99. [Google Scholar] [CrossRef]
  99. Tijore, A.; Behr, J.M.; Irvine, S.A.; Baisane, V.; Venkatraman, S. Bioprinted gelatin hydrogel platform promotes smooth muscle cell contractile phenotype maintenance. Biomed. Microdevics. 2018, 20, 32. [Google Scholar] [CrossRef]
  100. Sarkar, S.; Dadhania, M.; Rourke, P.; Desai, T.A.; Wong, J.Y. Vascular tissue engineering: Microtextured scaffold templates to control organization of vascular smooth muscle cells and extracellular matrix. Acta Biomater. 2005, 1, 93–100. [Google Scholar] [CrossRef]
  101. van Engeland, N.C.A.; Pollet, A.M.A.O.; den Toonder, J.M.J.; Bouten, C.V.C.; Stassen, O.M.J.A.; Sahlgren, C.M. A biomimetic microfluidic model to study signalling between endothelial and vascular smooth muscle cells under hemodynamic conditions. Lab Chip 2018, 18, 1607–1620. [Google Scholar] [CrossRef] [Green Version]
  102. O’Callaghan, C.J.; Williams, B. Mechanical Strain–Induced Extracellular Matrix Production by Human Vascular Smooth Muscle Cells. Hypertension 2000, 36, 319–324. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Cordes, K.R.; Sheehy, N.T.; White, M.P.; Berry, E.C.; Morton, S.U.; Muth, A.N.; Lee, T.H.; Miano, J.M.; Ivey, K.N.; Srivastava, D. miR-145 and miR-143 regulate smooth muscle cell fate and plasticity. Nature 2009, 460, 705. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Yoshida, T.; Kaestner, K.H.; Owens, G.K. Conditional Deletion of Krüppel-Like Factor 4 Delays Downregulation of Smooth Muscle Cell Differentiation Markers but Accelerates Neointimal Formation Following Vascular Injury. Circ. Res. 2008, 102, 1548–1557. [Google Scholar] [CrossRef] [PubMed]
  105. Cherepanova, O.A.; Pidkovka, N.A.; Sarmento, O.F.; Yoshida, T.; Gan, Q.; Adiguzel, E.; Bendeck, M.P.; Berliner, J.; Leitinger, N.; Owens, G.K. Oxidized Phospholipids Induce Type VIII Collagen Expression and Vascular Smooth Muscle Cell Migration. Circ. Res. 2009, 104, 609–618. [Google Scholar] [CrossRef] [Green Version]
  106. Shankman, L.S.; Gomez, D.; Cherepanova, O.A.; Salmon, M.; Alencar, G.F.; Haskins, R.M.; Swiatlowska, P.; Newman, A.A.C.; Greene, E.S.; Straub, A.C.; et al. KLF4-dependent phenotypic modulation of smooth muscle cells has a key role in atherosclerotic plaque pathogenesis. Nat. Med. 2015, 21, 628–637. [Google Scholar] [CrossRef] [Green Version]
  107. Kotchen, T.A. Historical trends and milestones in hypertension research: A model of the process of translational research. Hypertension 2011, 58, 522–538. [Google Scholar] [CrossRef] [Green Version]
  108. Franklin, S.S.; Wong, N.D. Hypertension and cardiovascular disease: Contributions of the framingham heart study. Glob. Heart 2013, 8, 49–57. [Google Scholar] [CrossRef] [Green Version]
  109. Dawber, T.R.; Kannel, W.B.; Revotskie, N.; Stokes, J.; Kagan, A.; Gordon, T. Some factors associated with the development of coronary heart disease: Six years’ follow-up experience in the Framingham study. Am. J. Public Health Nations Health 1959, 49, 1349–1356. [Google Scholar] [CrossRef]
  110. Lewington, S.; Clarke, R.; Qizilbash, N.; Peto, R.; Collins, R.; Collaboration, P.S. Age-specific relevance of usual blood pressure to vascular mortality: A meta-analysis of individual data for one million adults in 61 prospective studies. Lancet 2002, 360, 1903–1913. [Google Scholar]
  111. Rapsomaniki, E.; Timmis, A.; George, J.; Pujades-Rodriguez, M.; Shah, A.D.; Denaxas, S.; White, I.R.; Caulfield, M.J.; Deanfield, J.E.; Smeeth, L.; et al. Blood pressure and incidence of twelve cardiovascular diseases: Lifetime risks, healthy life-years lost, and age-specific associations in 1·25 million people. Lancet 2014, 383, 1899–1911. [Google Scholar] [CrossRef] [Green Version]
  112. Rahimi, K.; Emdin, C.A.; MacMahon, S. The epidemiology of blood pressure and its worldwide management. Circ. Res. 2015, 116, 925–936. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Najjar, S.S.; Scuteri, A.; Shetty, V.; Wright, J.G.; Muller, D.C.; Fleg, J.L.; Spurgeon, H.P.; Ferrucci, L.; Lakatta, E.G. Pulse wave velocity is an independent predictor of the longitudinal increase in systolic blood pressure and of incident hypertension in the Baltimore Longitudinal Study of Aging. J. Am. Coll. Cardiol. 2008, 51, 1377–1383. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Franklin, S.S.; Levy, D. Aging, blood pressure, and heart failure: What are the connections? Hypertension 2011, 58, 760–762. [Google Scholar] [CrossRef] [PubMed]
  115. Verwoert, G.C.; Franco, O.H.; Hoeks, A.P.G.; Reneman, R.S.; Hofman, A.; V Duijn, C.M.; Sijbrands, E.J.G.; Witteman, J.C.M.; Mattace-Raso, F.U.S. Arterial stiffness and hypertension in a large population of untreated individuals: The Rotterdam Study. J. Hypertens. 2014, 32, 1606–1612; discussion 1612. [Google Scholar] [CrossRef]
  116. Harvey, A.; Montezano, A.C.; Touyz, R.M. Vascular biology of ageing-Implications in hypertension. J. Mol. Cell. Cardiol. 2015, 83, 112–121. [Google Scholar] [CrossRef] [Green Version]
  117. Gimbrone, M.A.; García-Cardeña, G. Vascular endothelium, hemodynamics, and the pathobiology of atherosclerosis. Cardiovasc. Pathol. 2013, 22, 9–15. [Google Scholar] [CrossRef] [Green Version]
  118. Mitchell, G.F. Increased Aortic Stiffness: An Unfavorable Cardiorenal Connection. Hypertension 2004, 43, 151–153. [Google Scholar] [CrossRef] [Green Version]
  119. Dai, G.; Kaazempur-Mofrad, M.R.; Natarajan, S.; Zhang, Y.; Vaughn, S.; Blackman, B.R.; Kamm, R.D.; García-Cardeña, G.; Gimbrone, M.A. Distinct endothelial phenotypes evoked by arterial waveforms derived from atherosclerosis-susceptible and -resistant regions of human vasculature. Proc. Natl. Acad. Sci. USA 2004, 101, 14871–14876. [Google Scholar] [CrossRef] [Green Version]
  120. Passerini, A.G.; Polacek, D.C.; Shi, C.; Francesco, N.M.; Manduchi, E.; Grant, G.R.; Pritchard, W.F.; Powell, S.; Chang, G.Y.; Stoeckert, C.J.; et al. Coexisting proinflammatory and antioxidative endothelial transcription profiles in a disturbed flow region of the adult porcine aorta. Proc. Natl. Acad. Sci. USA 2004, 101, 2482–2487. [Google Scholar] [CrossRef] [Green Version]
  121. Blackman, B.R.; García-Cardeña, G.; Gimbrone, M.A. A New In Vitro Model to Evaluate Differential Responses of Endothelial Cells to Simulated Arterial Shear Stress Waveforms. J. Biomech. Eng. 2002, 124, 397–407. [Google Scholar] [CrossRef]
  122. Wang, Y.X.; Xiang, C.; Liu, B.; Zhu, Y.; Luan, Y.; Liu, S.T.; Qin, K.R. A multi-component parallel-plate flow chamber system for studying the effect of exercise-induced wall shear stress on endothelial cells. Biomed. Eng. Online 2016, 15, 154. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Williams, B. Mechanical influences on vascular smooth muscle cell function. J. Hypertens. 1998, 16, 1921–1929. [Google Scholar] [CrossRef] [PubMed]
  124. Kapustin, A.N.; Schoppet, M.; Schurgers, L.J.; Reynolds, J.L.; McNair, R.; Heiss, A.; Jahnen-Dechent, W.; Hackeng, T.M.; Schlieper, G.; Harrison, P.; et al. Prothrombin Loading of Vascular Smooth Muscle Cell–Derived Exosomes Regulates Coagulation and CalcificationHighlights. Arterioscler. Thromb. Vasc. Biol. 2017, 37, e22–e32. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Hutcheson, J.D.; Goettsch, C.; Pham, T.; Iwashita, M.; Aikawa, M.; Singh, S.A.; Aikawa, E. Enrichment of calcifying extracellular vesicles using density-based ultracentrifugation protocol. J. Extracell. Vesicles 2014, 3, 25129. [Google Scholar] [CrossRef]
  126. Tanimura, A.; McGregor, D.H.; Anderson, H.C. Matrix Vesicles in Atherosclerotic Calcification. Proc. Soc. Exp. Biol. Med. 2016, 172, 173–177. [Google Scholar] [CrossRef]
  127. Chen, N.X.; O’Neill, K.D.; Moe, S.M. Matrix vesicles induce calcification of recipient vascular smooth muscle cells through multiple signaling pathways. Kidney Int. 2018, 93, 343–354. [Google Scholar] [CrossRef] [Green Version]
  128. Carlo, A.S.; Nykjaer, A.; Willnow, T.E. Sorting receptor sortilin-a culprit in cardiovascular and neurological diseases. J. Mol. Med. (Berl.) 2014, 92, 905–911. [Google Scholar] [CrossRef]
  129. Goettsch, C.; Hutcheson, J.D.; Aikawa, M.; Iwata, H.; Pham, T.; Nykjaer, A.; Kjolby, M.; Rogers, M.; Michel, T.; Shibasaki, M.; et al. Sortilin mediates vascular calcification via its recruitment into extracellular vesicles. J. Clin. Invest. 2016, 126, 1323–1336. [Google Scholar] [CrossRef]
  130. Boström, K.I.; Rajamannan, N.M.; Towler, D.A. The regulation of valvular and vascular sclerosis by osteogenic morphogens. Circ. Res. 2011, 109, 564–577. [Google Scholar] [CrossRef]
  131. Shanahan, C.M.; Proudfoot, D.; Tyson, K.L.; Cary, N.R.; Edmonds, M.; Weissberg, P.L. Expression of mineralisation-regulating proteins in association with human vascular calcification. Z. Kardiol. 2000, 89, 63–68. [Google Scholar] [CrossRef]
  132. Touyz, R.M.; Alves-Lopes, R.; Rios, F.J.; Camargo, L.L.; Anagnostopoulou, A.; Arner, A.; Montezano, A.C. Vascular smooth muscle contraction in hypertension. Cardiovasc. Res. 2018, 114, 529–539. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Zhang, X.; Yan, S.M.; Zheng, H.L.; Hu, D.H.; Zhang, Y.T.; Guan, Q.H.; Ding, Q.L. A Mechanism Underlying Hypertensive Occurrence in the Metabolic Syndrome: Cooperative Effect of Oxidative Stress and Calcium Accumulation in Vascular Smooth Muscle Cells. Horm. Metab. Res. 2013, 46, 126–132. [Google Scholar] [PubMed]
  134. Fleckenstein, A.; Fleckenstein-Grün, G.; Frey, M.; Thimm, F. Experimental antiarteriosclerotic effects of calcium antagonists. J. Clin. Pharmacol. 1990, 30, 151–154. [Google Scholar] [CrossRef] [PubMed]
  135. Zhu, Y.; He, L.; Qu, J.; Zhou, Y. Regulation of Vascular Smooth Muscle Cell Stiffness and Adhesion by [Ca2+]i: An Atomic Force Microscopy-Based Study. Microsc. Microanal. 2018, 24, 708–712. [Google Scholar] [CrossRef]
  136. Godfraind, T. Discovery and Development of Calcium Channel Blockers. Front. Pharmacol. 2017, 8, 286. [Google Scholar] [CrossRef] [Green Version]
  137. Chen, N.X.; Kircelli, F.; O’Neill, K.D.; Chen, X.; Moe, S.M. Verapamil inhibits calcification and matrix vesicle activity of bovine vascular smooth muscle cells. Kidney Int. 2010, 77, 436–442. [Google Scholar] [CrossRef] [Green Version]
  138. Caballero-Gonzalez, F.J. Calcium Channel Blockers in the Management of Hypertension in the Elderly. Cardiovasc. Hematol. Agents Med. Chem. 2014, 12, 160–165. [Google Scholar] [CrossRef]
  139. Niederhoffer, N.; Marque, V.; Lartaud-Idjouadiene, I.; Duvivier, C.; Peslin, R.; Atkinson, J. Vasodilators, aortic elasticity, and ventricular end-systolic stress in nonanesthetized unrestrained rats. Hypertension 1997, 30, 1169–1174. [Google Scholar] [CrossRef]
  140. London, G.M.; Guérin, A.P.; Marchais, S.J.; Métivier, F.; Pannier, B.; Adda, H. Arterial media calcification in end-stage renal disease: Impact on all-cause and cardiovascular mortality. Nephrol. Dial. Transpl. 2003, 18, 1731–1740. [Google Scholar] [CrossRef]
  141. Odink, A.E.; Mattace-Raso, F.U.; van der Lugt, A.; Hofman, A.; Hunink, M.G.M.; Breteler, M.M.; Krestin, G.P.; Witteman, J.C. The association of arterial stiffness and arterial calcification: The Rotterdam study. J. Hum. Hypertens. 2008, 22, 205–207. [Google Scholar] [CrossRef] [Green Version]
  142. Guérin, A.P.; London, G.M.; Marchais, S.J.; Metivier, F. Arterial stiffening and vascular calcifications in end-stage renal disease. Nephrol. Dial. Transplant 2000, 15, 1014–1021. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Wagenseil, J.E.; Mecham, R.P. Elastin in large artery stiffness and hypertension. J. Cardiovasc. Transl. Res. 2012, 5, 264–273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Kaess, B.M.; Rong, J.; Larson, M.G.; Hamburg, N.M.; Vita, J.A.; Levy, D.; Benjamin, E.J.; Vasan, R.S.; Mitchell, G.F. Aortic stiffness, blood pressure progression, and incident hypertension. JAMA 2012, 308, 875–881. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Sehgel, N.L.; Zhu, Y.; Sun, Z.; Trzeciakowski, J.P.; Hong, Z.; Hunter, W.C.; Vatner, D.E.; Meininger, G.A.; Vatner, S.F. Increased vascular smooth muscle cell stiffness: A novel mechanism for aortic stiffness in hypertension. Am. J. Physiol. Heart Circ. Physiol. 2013, 305, H1281–H1287. [Google Scholar] [CrossRef]
  146. Csiszar, A.; Lehoux, S.; Ungvari, Z. Hemodynamic Forces, Vascular Oxidative Stress, and Regulation of BMP-2/4 Expression. Antioxid. Redox. Signal. 2009, 11, 1683–1697. [Google Scholar] [CrossRef] [Green Version]
  147. Richardson, P.D. Biomechanics of plaque rupture: Progress, problems, and new frontiers. Ann. Biomed. Eng. 2002, 30, 524–536. [Google Scholar] [CrossRef]
  148. Tang, D.; Yang, C.; Mondal, S.; Liu, F.; Canton, G.; Hatsukami, T.S.; Yuan, C. A negative correlation between human carotid atherosclerotic plaque progression and plaque wall stress: In vivo MRI-based 2D/3D FSI models. J. Biomech. 2008, 41, 727–736. [Google Scholar] [CrossRef] [Green Version]
  149. Hopkins, P.N. Molecular Biology of Atherosclerosis. Physiol. Rev. 2013, 93, 1317–1542. [Google Scholar] [CrossRef]
  150. Bennett, M.R.; Sinha, S.; Owens, G.K. Vascular Smooth Muscle Cells in Atherosclerosis. Circ. Res. 2016, 118, 692–702. [Google Scholar] [CrossRef]
  151. Ross, R. Atherosclerosis--an inflammatory disease. N. Engl. J. Med. 1999, 340, 115–126. [Google Scholar] [CrossRef]
  152. Chatrou, M.L.L.; Cleutjens, J.P.; van der Vusse, G.J.; Roijers, R.B.; Mutsaers, P.H.A.; Schurgers, L.J. Intra-Section Analysis of Human Coronary Arteries Reveals a Potential Role for Micro-Calcifications in Macrophage Recruitment in the Early Stage of Atherosclerosis. PLoS ONE 2015, 10, e0142335. [Google Scholar] [CrossRef]
  153. Feil, S.; Fehrenbacher, B.; Lukowski, R.; Essmann, F.; Schulze-Osthoff, K.; Schaller, M.; Feil, R. Transdifferentiation of vascular smooth muscle cells to macrophage-like cells during atherogenesis. Circ. Res. 2014, 115, 662–667. [Google Scholar] [CrossRef] [PubMed]
  154. Vaidya, D.; Szklo, M.; Cushman, M.; Holvoet, P.; Polak, J.; Bahrami, H.; Jenny, N.S.; Ouyang, P. Association of endothelial and oxidative stress with metabolic syndrome and subclinical atherosclerosis: Multi-ethnic study of atherosclerosis. Eur. J. Clin. Nutr. 2011, 65, 818–825. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Ross, R.; Glomset, J.A. Atherosclerosis and the arterial smooth muscle cell: Proliferation of smooth muscle is a key event in the genesis of the lesions of atherosclerosis. Science 1973, 180, 1332–1339. [Google Scholar] [CrossRef] [PubMed]
  156. Blumenthal, H. Calcification of the media of the human aorta and its relation to intimal arteriosclerosis, ageing and disease. Am. J. Pathol. 2007, 1–23. [Google Scholar]
  157. Sangiorgi, G.; Rumberger, J.A.; Severson, A.; Edwards, W.D.; Gregoire, J.; Fitzpatrick, L.A.; Schwartz, R.S. Arterial Calcification and Not Lumen Stenosis Is Highly Correlated With Atherosclerotic Plaque Burden in Humans: A Histologic Study of 723 Coronary Artery Segments Using Nondecalcifying Methodology. J. Am. Coll. Cardiol. 1998, 31, 126–133. [Google Scholar] [CrossRef] [Green Version]
  158. Pundziute, G.; Schuijf, J.D.; Jukema, J.W.; Decramer, I.; Sarno, G.; Vanhoenacker, P.K.; Reiber, J.H.C.; Schalij, M.J.; Wijns, W.; Bax, J.J. Head-to-head comparison of coronary plaque evaluation between multislice computed tomography and intravascular ultrasound radiofrequency data analysis. JACC. Cardiovasc. Interv. 2008, 1, 176–182. [Google Scholar] [CrossRef] [Green Version]
  159. Rodriguez-Granillo, G.A.; Carrascosa, P.; Bruining, N.; Waksman, R.; Garcia-Garcia, H.M. Defining the non-vulnerable and vulnerable patients with computed tomography coronary angiography: Evaluation of atherosclerotic plaque burden and composition. Eur. Heart J. Cardiovasc. Imaging 2016, 17, 481–491. [Google Scholar] [CrossRef] [Green Version]
  160. Nissen, S.E. Effect of intensive lipid lowering on progression of coronary atherosclerosis: Evidence for an early benefit from the Reversal of Atherosclerosis with Aggressive Lipid Lowering (REVERSAL) trial. Am. J. Cardiol. 2005, 96, 61F–68F. [Google Scholar] [CrossRef]
  161. Ridker, P.M.; Pradhan, A.; MacFadyen, J.G.; Libby, P.; Glynn, R.J. Cardiovascular benefits and diabetes risks of statin therapy in primary prevention: An analysis from the JUPITER trial. Lancet 2012, 380, 565–571. [Google Scholar] [CrossRef] [Green Version]
  162. Puri, R.; Libby, P.; Nissen, S.E.; Wolski, K.; Ballantyne, C.M.; Barter, P.J.; Chapman, M.J.; Erbel, R.; Raichlen, J.S.; Uno, K.; et al. Long-term effects of maximally intensive statin therapy on changes in coronary atheroma composition: Insights from SATURN. Eur. Heart J. Cardiovasc. Imaging 2014, 15, 380–388. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Räber, L.; Taniwaki, M.; Zaugg, S.; Kelbæk, H.; Roffi, M.; Holmvang, L.; Noble, S.; Pedrazzini, G.; Moschovitis, A.; Lüscher, T.F.; et al. Effect of high-intensity statin therapy on atherosclerosis in non-infarct-related coronary arteries (IBIS-4): A serial intravascular ultrasonography study. Eur. Heart J. 2015, 36, 490–500. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Rennenberg, R.J.M.W.; Kessels, A.G.H.; Schurgers, L.J.; van Engelshoven, J.M.A.; de Leeuw, P.W.; Kroon, A.A. Vascular calcifications as a marker of increased cardiovascular risk: A meta-analysis. Vasc. Health Risk Manag. 2009, 5, 185–197. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Essalihi, R.; Dao, H.H.; Yamaguchi, N.; Moreau, P. A new model of isolated systolic hypertension induced by chronic warfarin and vitamin K1 treatment. Am. J. Hypertens. 2003, 16, 103–110. [Google Scholar] [CrossRef] [Green Version]
  166. Essalihi, R.; Ouellette, V.; Hao Dao, H.; McKee, M.D.; Moreau, P. Phenotypic Modulation of Vascular Smooth Muscle Cells During Medial Arterial Calcification: A Role for Endothelin? J. Cardiovasc. Pharmacol. 2004, 44, S147–S150. [Google Scholar] [CrossRef]
  167. Lanzer, P.; Boehm, M.; Sorribas, V.; Thiriet, M.; Janzen, J.; Zeller, T.; St Hilaire, C.; Shanahan, C. Medial vascular calcification revisited: Review and perspectives. Eur. Heart J. 2014, 35, 1515–1525. [Google Scholar] [CrossRef]
  168. Ehara, S.; Kobayashi, Y.; Yoshiyama, M.; Shimada, K.; Shimada, Y.; Fukuda, D.; Nakamura, Y.; Yamashita, H.; Yamagishi, H.; Takeuchi, K.; et al. Spotty calcification typifies the culprit plaque in patients with acute myocardial infarction: An intravascular ultrasound study. Circulation 2004, 110, 3424–3429. [Google Scholar] [CrossRef] [Green Version]
  169. Vengrenyuk, Y.; Carlier, S.; Xanthos, S.; Cardoso, L.; Ganatos, P.; Virmani, R.; Einav, S.; Gilchrist, L.; Weinbaum, S. A hypothesis for vulnerable plaque rupture due to stress-induced debonding around cellular microcalcifications in thin fibrous caps. Proc. Natl. Acad. Sci. USA 2006, 103, 14678–14683. [Google Scholar] [CrossRef] [Green Version]
  170. Schurgers, L.J.; Joosen, I.A.; Laufer, E.M.; Chatrou, M.L.L.; Herfs, M.; Winkens, M.H.M.; Westenfeld, R.; Veulemans, V.; Krueger, T.; Shanahan, C.M.; et al. Vitamin K-antagonists accelerate atherosclerotic calcification and induce a vulnerable plaque phenotype. PLoS ONE 2012, 7, e43229. [Google Scholar] [CrossRef] [Green Version]
  171. Iyemere, V.P.; Proudfoot, D.; Weissberg, P.L.; Shanahan, C.M. Vascular smooth muscle cell phenotypic plasticity and the regulation of vascular calcification. J. Int. Med. 2006, 260, 192–210. [Google Scholar] [CrossRef]
  172. Shanahan, C.M.; Crouthamel, M.H.; Kapustin, A.; Giachelli, C.M. Arterial calcification in chronic kidney disease: Key roles for calcium and phosphate. Circ. Res. 2011, 109, 697–711. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Friedman, S.L.; Sheppard, D.; Duffield, J.S.; Violette, S. Therapy for fibrotic diseases: Nearing the starting line. Sci. Trans. Med. 2013, 5, sr1–sr167. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Hinz, B.; Phan, S.H.; Thannickal, V.J.; Prunotto, M.; Desmoulière, A.; Varga, J.; de Wever, O.; Mareel, M.; Gabbiani, G. Recent developments in myofibroblast biology: Paradigms for connective tissue remodeling. Am. J. Pathol. 2012, 180, 1340–1355. [Google Scholar] [CrossRef] [PubMed]
  175. Liberman, M.; Pesaro, A.E.P.; Carmo, L.S.; Serrano Jr, C.V. Vascular calcification: Pathophysiology and clinical implications. Einstein 2013, 11, 376–382. [Google Scholar] [CrossRef] [PubMed]
  176. Ducy, P.; Zhang, R.; Geoffroy, V.; Ridall, A.L.; Karsenty, G. Osf2/Cbfa1: A transcriptional activator of osteoblast differentiation. Cell 1997, 89, 747–754. [Google Scholar] [CrossRef] [Green Version]
  177. Sun, Y.; Byon, C.H.; Yuan, K.; Chen, J.; Mao, X.; Heath, J.M.; Javed, A.; Zhang, K.; Anderson, P.G.; Chen, Y. Smooth muscle cell-specific runx2 deficiency inhibits vascular calcification. Circ. Res. 2012, 111, 543–552. [Google Scholar] [CrossRef] [Green Version]
  178. Block, G.A.; Port, F.K. Re-evaluation of risks associated with hyperphosphatemia and hyperparathyroidism in dialysis patients: Recommendations for a change in management. Am. J. Kidney Dis. 2000, 35, 1226–1237. [Google Scholar] [CrossRef]
  179. Zhao, M.M.; Xu, M.J.; Cai, Y.; Zhao, G.; Guan, Y.; Kong, W.; Tang, C.; Wang, X. Mitochondrial reactive oxygen species promote p65 nuclear translocation mediating high-phosphate-induced vascular calcification in vitro and in vivo. Kidney Int. 2011, 79, 1071–1079. [Google Scholar] [CrossRef] [Green Version]
  180. Tiwari, B.K.; Pandey, K.B.; Abidi, A.B.; Rizvi, S.I. Markers of Oxidative Stress during Diabetes Mellitus. J. Biomark. 2013, 2013, 1–8. [Google Scholar] [CrossRef] [Green Version]
  181. Wei, Q.; Ren, X.; Jiang, Y.; Jin, H.; Liu, N.; Li, J. Advanced glycation end products accelerate rat vascular calcification through RAGE/oxidative stress. BMC Cardiovasc. Disord. 2013, 13, 13. [Google Scholar] [CrossRef] [Green Version]
  182. Tada, Y.; Yano, S.; Yamaguchi, T.; Okazaki, K.; Ogawa, N.; Morita, M.; Sugimoto, T. Advanced glycation end products-induced vascular calcification is mediated by oxidative stress: Functional roles of NAD(P)H-oxidase. Horm. Metab. Res. 2013, 45, 267–272. [Google Scholar] [CrossRef] [PubMed]
  183. Girerd, X.; Mourad, J.J.; Copie, X.; Moulin, C.; Acar, C.; Safar, M.; Laurent, S. Noninvasive detection of an increased vascular mass in untreated hypertensive patients. Am. J. Hypertens. 1994, 7, 1076–1084. [Google Scholar] [CrossRef] [PubMed]
  184. Fridez, P.; Zulliger, M.; Bobard, F.; Montorzi, G.; Miyazaki, H.; Hayashi, K.; Stergiopulos, N. Geometrical, functional, and histomorphometric adaptation of rat carotid artery in induced hypertension. J. Biomech. 2003, 36, 671–680. [Google Scholar] [CrossRef]
  185. Safar, M.; Chamiot-Clerc, P.; Dagher, G.; Renaud, J.F. Pulse pressure, endothelium function, and arterial stiffness in spontaneously hypertensive rats. Hypertension 2001, 38, 1416–1421. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  186. Arribas, S.M.; Hinek, A.; González, M.C. Elastic fibres and vascular structure in hypertension. Pharmacol. Ther. 2006, 111, 771–791. [Google Scholar] [CrossRef] [PubMed]
  187. Kramann, R.; Schneider, R.K.; DiRocco, D.P.; Machado, F.; Fleig, S.; Bondzie, P.A.; Henderson, J.M.; Ebert, B.L.; Humphreys, B.D. Perivascular Gli1+ progenitors are key contributors to injury-induced organ fibrosis. Cell Stem. Cell 2015, 16, 51–66. [Google Scholar] [CrossRef] [Green Version]
  188. Kramann, R.; Goettsch, C.; Wongboonsin, J.; Iwata, H.; Schneider, R.K.; Kuppe, C.; Kaesler, N.; Chang-Panesso, M.; Machado, F.G.; Gratwohl, S.; et al. Adventitial MSC-like Cells Are Progenitors of Vascular Smooth Muscle Cells and Drive Vascular Calcification in Chronic Kidney Disease. Cell Stem. Cell 2016, 19, 628–642. [Google Scholar] [CrossRef] [Green Version]
  189. Bersi, M.R.; Bellini, C.; Wu, J.; Montaniel, K.R.C.; Harrison, D.G.; Humphrey, J.D. Excessive Adventitial Remodeling Leads to Early Aortic Maladaptation in Angiotensin-Induced Hypertension. Hypertension 2016, 67, 890–896. [Google Scholar] [CrossRef] [Green Version]
  190. Majesky, M.W.; Horita, H.; Ostriker, A.; Lu, S.; Regan, J.N.; Bagchi, A.; Dong, X.R.; Poczobutt, J.; Nemenoff, R.A.; Weiser-Evans, M.C.M. Differentiated Smooth Muscle Cells Generate a Subpopulation of Resident Vascular Progenitor Cells in the Adventitia Regulated by Klf4Novelty and Significance. Circ. Res. 2017, 120, 296–311. [Google Scholar] [CrossRef]
  191. Wu, J.; Montaniel, K.R.C.; Saleh, M.A.; Xiao, L.; Chen, W.; Owens, G.K.; Humphrey, J.D.; Majesky, M.W.; Paik, D.T.; Hatzopoulos, A.K.; et al. Origin of Matrix-Producing Cells That Contribute to Aortic Fibrosis in Hypertension. Hypertension 2016, 67, 461–468. [Google Scholar] [CrossRef] [Green Version]
  192. Cecelja, M.; Chowienczyk, P. Role of arterial stiffness in cardiovascular disease. JRSM Cardiovasc. Dis. 2012, 1, 11. [Google Scholar] [CrossRef]
  193. Lee, H.Y.; Oh, B.H. Aging and arterial stiffness. Circ. J. 2010, 74, 2257–2262. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Mancia, G.; Fagard, R.; Narkiewicz, K.; Redon, J.; Zanchetti, A.; Bohm, M.; Christiaens, T.; Cifkova, R.; de Backer, G.; Dominiczak, A.; et al. The task force for the management ofarterial hypertension of the european society ofhypertension (esh) and of the european society of cardiology (esc). J. Hypertens. 2013, 31, 1281–1357. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. Nürnberger, J.; Keflioglu-Scheiber, A.; Opazo Saez, A.M.; Wenzel, R.R.; Philipp, T.; Schäfers, R.F. Augmentation index is associated with cardiovascular risk. J. Hypertens. 2002, 20, 2407–2414. [Google Scholar] [CrossRef] [PubMed]
  196. Vlachopoulos, C.; Ioakeimidis, N.; Aznaouridis, K.; Terentes-Printzios, D.; Rokkas, K.; Aggelis, A.; Panagiotakos, D.; Stefanadis, C. Prediction of Cardiovascular Events With Aortic Stiffness in Patients With Erectile Dysfunction. Hypertension 2014, 64, 672–678. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  197. Chow, B.; Rabkin, S.W. Brachial-ankle pulse wave velocity is the only index of arterial stiffness that correlates with a mitral valve indices of diastolic dysfunction, but no index correlates with left atrial size. Cardiol. Res. Pract. 2013, 2013, 986847. [Google Scholar] [CrossRef] [PubMed]
  198. Spronck, B.; Heusinkveld, M.H.G.; Vanmolkot, F.H.; Roodt, J.O.; Hermeling, E.; Delhaas, T.; Kroon, A.A.; Reesink, K.D. Pressure-dependence of arterial stiffness: Potential clinical implications. J. Hypertens. 2015, 33, 330–338. [Google Scholar] [CrossRef]
  199. Spronck, B.; Avolio, A.P.; Tan, I.; Butlin, M.; Reesink, K.D.; Delhaas, T. Arterial stiffness index beta and cardio-ankle vascular index inherently depend on blood pressure but can be readily corrected. J. Hypertens. 2017, 35, 98–104. [Google Scholar] [CrossRef]
  200. Spronck, B.; Delhaas, T.; de Lepper, A.G.; Giroux, J.; Goldwasser, F.; Boutouyrie, P.; Alivon, M.; Reesink, K.D. Patient-specific blood pressure correction technique for arterial stiffness: Evaluation in a cohort on anti-angiogenic medication. Hypertens. Res. 2017, 40, 752–757. [Google Scholar] [CrossRef]
  201. Dao, H.H.; Essalihi, R.; Bouvet, C.; Moreau, P. Evolution and modulation of age-related medial elastocalcinosis: Impact on large artery stiffness and isolated systolic hypertension. Cardiovasc. Res. 2005, 66, 307–317. [Google Scholar] [CrossRef] [Green Version]
  202. Laucyte-Cibulskiene, A.; Petraviciute, M.; Gudynaite, M.; Gumbys, L.; Valanciene, D.; Galiauskiene, K.; Ryliskyte, L.; Rimsevicius, L.; Miglinas, M.; Strupas, K. Mismatch between stiffness in elastic and muscular arteries as a predictor of vascular calcification in dialysis patients. Aging Clin. Exp. Res. 2018, 30, 375–382. [Google Scholar] [CrossRef] [PubMed]
  203. Sehgel, N.L.; Sun, Z.; Hong, Z.; Hunter, W.C.; Hill, M.A.; Vatner, D.E.; Vatner, S.F.; Meininger, G.A. Augmented Vascular Smooth Muscle Cell Stiffness and Adhesion When Hypertension Is Superimposed on Aging. Hypertension 2014, 65, 370–373. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  204. Imanishi, M.; Tomita, S.; Ishizawa, K.; Kihira, Y.; Ueno, M.; Izawa-Ishizawa, Y.; Ikeda, Y.; Yamano, N.; Tsuchiya, K.; Tamaki, T. Smooth muscle cell-specific Hif-1α deficiency suppresses angiotensin II-induced vascular remodelling in mice. Cardiovasc. Res. 2014, 102, 460–468. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  205. Gonzalez, W.; Fontaine, V.; Pueyo, M.E.; Laquay, N.; Messika-Zeitoun, D.; Philippe, M.; Arnal, J.F.; Jacob, M.P.; Michel, J.B. Molecular plasticity of vascular wall during N(G)-nitro-L-arginine methyl ester-induced hypertension: Modulation of proinflammatory signals. Hypertension 2000, 36, 103–109. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  206. Laurent, S.; Boutouyrie, P. Arterial stiffness and stroke in hypertension: Therapeutic implications for stroke prevention. CNS Drug. 2005, 19, 1–11. [Google Scholar] [CrossRef] [PubMed]
  207. Segers, P.; Swillens, A.; Taelman, L.; Vierendeels, J. Wave reflection leads to over- and underestimation of local wave speed by the PU- and QA-loop methods: Theoretical basis and solution to the problem. Physiol. Meas. 2014, 35, 847–861. [Google Scholar] [CrossRef]
  208. Hermeling, E.; Reesink, K.D.; Kornmann, L.M.; Reneman, R.S.; Hoeks, A.P. The dicrotic notch as alternative time-reference point to measure local pulse wave velocity in the carotid artery by means of ultrasonography. J. Hypertens. 2009, 27, 2028–2035. [Google Scholar] [CrossRef]
  209. Mirault, T.; Pernot, M.; Frank, M.; Couade, M.; Niarra, R.; Azizi, M.; Emmerich, J.; Jeunemaître, X.; Fink, M.; Tanter, M.; et al. Carotid stiffness change over the cardiac cycle by ultrafast ultrasound imaging in healthy volunteers and vascular Ehlers-Danlos syndrome. J. Hypertens. 2015, 33, 1890–1896. [Google Scholar] [CrossRef]
  210. de Korte, C.L.; Fekkes, S.; Nederveen, A.J.; Manniesing, R.; Hansen, H.R.H.G. Review: Mechanical Characterization of Carotid Arteries and Atherosclerotic Plaques. IEEE Trans. Ultrason. Ferroelectr. Freq. Control 2016, 63, 1613–1623. [Google Scholar] [CrossRef]
  211. Couade, M.; Pernot, M.; Prada, C.; Messas, E.; Emmerich, J.; Bruneval, P.; Criton, A.; Fink, M.; Tanter, M. Quantitative assessment of arterial wall biomechanical properties using shear wave imaging. Ultrasound Med. Biol. 2010, 36, 1662–1676. [Google Scholar] [CrossRef]
  212. Grotenhuis, H.B.; Westenberg, J.J.M.; Steendijk, P.; van der Geest, R.J.; Ottenkamp, J.; Bax, J.J.; Jukema, J.W.; de Roos, A. Validation and reproducibility of aortic pulse wave velocity as assessed with velocity-encoded MRI. J. Magn. Reson. Imaging 2009, 30, 521–526. [Google Scholar] [CrossRef] [PubMed]
  213. Polak, J.F.; Szklo, M.; Kronmal, R.A.; Burke, G.L.; Shea, S.; Zavodni, A.E.H.; O’Leary, D.H. The Value of Carotid Artery Plaque and Intima-Media Thickness for Incident Cardiovascular Disease: The Multi-Ethnic Study of Atherosclerosis. J. Am. Heart Assoc. 2013, 2. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  214. Iwakiri, T.; Yano, Y.; Sato, Y.; Hatakeyama, K.; Marutsuka, K.; Fujimoto, S.; Kitamura, K.; Kario, K.; Asada, Y. Usefulness of carotid intima-media thickness measurement as an indicator of generalized atherosclerosis: Findings from autopsy analysis. Atherosclerosis 2012, 225, 359–362. [Google Scholar] [CrossRef] [PubMed]
  215. Crouse, J.R.; Raichlen, J.S.; Riley, W.A.; Evans, G.W.; Palmer, M.K.; O’Leary, D.H.; Grobbee, D.E.; Bots, M.L.; METEOR Study Group. Effect of Rosuvastatin on Progression of Carotid Intima-Media Thickness in Low-Risk Individuals With Subclinical Atherosclerosis. JAMA 2007, 297, 1344–1353. [Google Scholar] [CrossRef] [Green Version]
  216. de Groot, E.; Jukema, J.W.; Montauban van Swijndregt, A.D.; Zwinderman, A.H.; Ackerstaff, R.G.A.; van der Steen, A.F.W.; Bom, N.; Lie, K.I.; Bruschke, A.V.G. B-Mode Ultrasound Assessment of Pravastatin Treatment Effect on Carotid and Femoral Artery Walls and Its Correlations With Coronary Arteriographic Findings: A Report of the Regression Growth Evaluation Statin Study (REGRESS). J. Am. Coll. Cardiol. 1998, 31, 1561–1567. [Google Scholar] [CrossRef] [Green Version]
  217. Lorenz, M.W.; Polak, J.F.; Kavousi, M.; Mathiesen, E.B.; Völzke, H.; Tuomainen, T.-P.; Sander, D.; Plichart, M.; Catapano, A.L.; Robertson, C.M.; et al. Carotid intima-media thickness progression to predict cardiovascular events in the general population (the PROG-IMT collaborative project): A meta-analysis of individual participant data. Lancet 2012, 379, 2053–2062. [Google Scholar] [CrossRef] [Green Version]
  218. Ravani, A.; Werba, J.P.; Frigerio, B.; Sansaro, D.; Amato, M.; Tremoli, E.; Baldassarre, D. Assessment and Relevance of Carotid Intima-Media Thickness (C-IMT) in Primary and Secondary Cardiovascular Prevention. Curr. Pharm. Des. 2015, 21, 1164–1171. [Google Scholar] [CrossRef] [Green Version]
  219. Zaid, M.; Fujiyoshi, A.; Kadota, A.; Abbott, R.D.; Miura, K. Coronary Artery Calcium and Carotid Artery Intima Media Thickness and Plaque: Clinical Use in Need of Clarification. J. Atheroscler. Thromb. 2017, 24, 227–239. [Google Scholar] [CrossRef] [Green Version]
  220. Iglesias del Sol, A.; Bots, M.L.; Grobbee, D.E.; Hofman, A.; Witteman, J.C.M. Carotid intima-media thickness at different sites: Relation to incident myocardial infarction. The Rotterdam Study. Eur. Heart J. 2002, 23, 934–940. [Google Scholar] [CrossRef] [Green Version]
  221. O’Leary, D.H.; Bots, M.L. Imaging of atherosclerosis: Carotid intima-media thickness. Eur. Heart J. 2010, 31, 1682–1689. [Google Scholar] [CrossRef] [Green Version]
  222. Skilton, M.R.; Sérusclat, A.; Sethu, A.H.A.U.; Brun, S.; Bernard, S.; Balkau, B.; Moulin, P.; Bonnet, F. Noninvasive measurement of carotid extra-media thickness: Associations with cardiovascular risk factors and intima-media thickness. JACC Cardiovasc. Imaging 2009, 2, 176–182. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  223. Malik, S.; Budoff, M.J.; Katz, R.; Blumenthal, R.S.; Bertoni, A.G.; Nasir, K.; Szklo, M.; Barr, R.G.; Wong, N.D. Impact of subclinical atherosclerosis on cardiovascular disease events in individuals with metabolic syndrome and diabetes: The multi-ethnic study of atherosclerosis. Diabetes Care 2011, 34, 2285–2290. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  224. Boulos, N.M.; Gardin, J.M.; Malik, S.; Postley, J.; Wong, N.D. Carotid Plaque Characterization, Stenosis, and Intima-Media Thickness According to Age and Gender in a Large Registry Cohort. Am. J. Cardiol. 2016, 117, 1185–1191. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  225. Agatston, A.S.; Janowitz, W.R.; Hildner, F.J.; Zusmer, N.R.; Viamonte, M.; Detrano, R. Quantification of coronary artery calcium using ultrafast computed tomography. J. Am. Coll. Cardiol. 1990, 15, 827–832. [Google Scholar] [CrossRef] [Green Version]
  226. Popovic, W.J.; Brown, J.E.; Adamson, J.W. The influence of thyroid hormones on in vitro erythropoiesis. Mediation by a receptor with beta adrenergic properties. J. Clin. Invest. 1977, 60, 907–913. [Google Scholar] [CrossRef]
  227. Greenland, P.; Bonow, R.O.; Brundage, B.H.; Budoff, M.J.; Eisenberg, M.J.; Grundy, S.M.; Lauer, M.S.; Post, W.S.; Raggi, P.; Redberg, R.F.; et al. ACCF/AHA 2007 clinical expert consensus document on coronary artery calcium scoring by computed tomography in global cardiovascular risk assessment and in evaluation of patients with chest pain: A report of the American College of Cardiology Foundation Clinical Expert Consensus Task Force (ACCF/AHA Writing Committee to Update the 2000 Expert Consensus Document on Electron Beam Computed Tomography). Circulation 2007, 115, 402–426. [Google Scholar]
  228. Fujiyoshi, A.; Miura, K.; Ohkubo, T.; Kadowaki, T.; Kadowaki, S.; Zaid, M.; Hisamatsu, T.; Sekikawa, A.; Budoff, M.J.; Liu, K.; et al. Cross-Sectional Comparison of Coronary Artery Calcium Scores Between Caucasian Men in the United States and Japanese Men in JapanThe Multi-Ethnic Study of Atherosclerosis and the Shiga Epidemiological Study of Subclinical Atherosclerosis. Am. J. Epidemiol. 2014, 180, 590–598. [Google Scholar] [CrossRef] [Green Version]
  229. Raggi, P.; Callister, T.Q.; Shaw, L.J. Progression of coronary artery calcium and risk of first myocardial infarction in patients receiving cholesterol-lowering therapy. Arterioscler. Thromb. Vasc. Biol. 2004, 24, 1272–1277. [Google Scholar] [CrossRef]
  230. Raggi, P. Cardiovascular disease: Coronary artery calcification predicts risk of CVD in patients with CKD. Nat. Rev. Nephrol. 2017, 13, 324–326. [Google Scholar] [CrossRef]
  231. Yamamoto, H.; Kitagawa, T.; Kihara, Y. Clinical Implications of the Coronary Artery Calcium Score in Japanese Patients. J. Atheroscler. Thromb. 2014, 21, 1101–1108. [Google Scholar] [CrossRef] [Green Version]
  232. Krueger, T.; Schlieper, G.; Schurgers, L.; Cornelis, T.; Cozzolino, M.; Jacobi, J.; Jadoul, M.; Ketteler, M.; Rump, L.C.; Stenvinkel, P.; et al. Vitamin K1 to slow vascular calcification in haemodialysis patients (VitaVasK trial): A rationale and study protocol. Nephrol. Dial. Trans. 2014, 29, 1633–1638. [Google Scholar] [CrossRef] [PubMed]
  233. Vossen, L.M.; Schurgers, L.J.; van Varik, B.J.; Kietselaer, B.L.J.H.; Vermeer, C.; Meeder, J.G.; Rahel, B.M.; van Cauteren, Y.J.M.; Hoffland, G.A.; Rennenberg, R.J.M.W.; et al. Menaquinone-7 Supplementation to Reduce Vascular Calcification in Patients with Coronary Artery Disease: Rationale and Study Protocol (VitaK-CAC Trial). Nutrients 2015, 7, 8905–8915. [Google Scholar] [CrossRef] [Green Version]
  234. Peeters, F.E.C.M.; van Mourik, M.J.W.; Meex, S.J.R.; Bucerius, J.; Schalla, S.M.; Gerretsen, S.C.; Mihl, C.; Dweck, M.R.; Schurgers, L.J.; Wildberger, J.E.; et al. Bicuspid Aortic Valve Stenosis and the Effect of Vitamin K2 on Calcification Using 18F-Sodium Fluoride Positron Emission Tomography/Magnetic Resonance: The BASIK2 Rationale and Trial Design. Nutrients 2018, 10, 386. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  235. Geleijnse, J.M.; Vermeer, C.; Grobbee, D.E.; Schurgers, L.J.; Knapen, M.H.J.; van der Meer, I.M.; Hofman, A.; Witteman, J.C.M. Dietary Intake of Menaquinone Is Associated with a Reduced Risk of Coronary Heart Disease: The Rotterdam Study. J. Nutr. 2004, 134, 3100–3105. [Google Scholar] [CrossRef] [PubMed]
  236. Brandenburg, V.M.; Reinartz, S.; Kaesler, N.; Krüger, T.; Dirrichs, T.; Kramann, R.; Peeters, F.; Floege, J.; Keszei, A.; Marx, N.; et al. Slower Progress of Aortic Valve Calcification With Vitamin K Supplementation: Results From a Prospective Interventional Proof-of-Concept Study. Circulation 2017, 135, 2081–2083. [Google Scholar] [CrossRef]
  237. Houslay, E.S.; Cowell, S.J.; Prescott, R.J.; Reid, J.; Burton, J.; Northridge, D.B.; Boon, N.A.; Newby, D.E.; Scottish Aortic Stenosis and Lipid Lowering Therapy, Impact on Regression trial Investigators. Progressive coronary calcification despite intensive lipid-lowering treatment: A randomised controlled trial. Heart 2006, 92, 1207–1212. [Google Scholar] [CrossRef] [Green Version]
  238. Chen, Z.; Qureshi, A.R.; Parini, P.; Hurt-Camejo, E.; Ripsweden, J.; Brismar, T.B.; Barany, P.; Jaminon, A.M.; Schurgers, L.J.; Heimbürger, O.; et al. Does statins promote vascular calcification in chronic kidney disease? Eur. J. Clin. Investig. 2017, 47, 137–148. [Google Scholar] [CrossRef]
  239. Terry, J.G.; Carr, J.J.; Kouba, E.O.; Davis, D.H.; Menon, L.; Bender, K.; Chandler, E.T.; Morgan, T.; Crouse, J.R. Effect of simvastatin (80 mg) on coronary and abdominal aortic arterial calcium (from the coronary artery calcification treatment with zocor [CATZ] study). Am. J. Cardiol. 2007, 99, 1714–1717. [Google Scholar] [CrossRef]
  240. Criqui, M.H.; Denenberg, J.O.; Ix, J.H.; McClelland, R.L.; Wassel, C.L.; Rifkin, D.E.; Carr, J.J.; Budoff, M.J.; Allison, M.A. Calcium density of coronary artery plaque and risk of incident cardiovascular events. JAMA 2014, 311, 271–278. [Google Scholar] [CrossRef]
  241. Rumberger, J.A.; Simons, D.B.; Fitzpatrick, L.A.; Sheedy, P.F.; Schwartz, R.S. Coronary Artery Calcium Area by Electron-Beam Computed Tomography and Coronary Atherosclerotic Plaque Area. Circulation 1995, 92, 2157–2162. [Google Scholar] [CrossRef]
  242. Cho, I.; Ó Hartaigh, B.; Gransar, H.; Valenti, V.; Lin, F.Y.; Achenbach, S.; Berman, D.S.; Budoff, M.J.; Callister, T.Q.; Al-Mallah, M.H.; et al. Prognostic implications of coronary artery calcium in the absence of coronary artery luminal narrowing. Atherosclerosis 2017, 262, 185–190. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  243. Amann, K. Media calcification and intima calcification are distinct entities in chronic kidney disease. Clin. J. Am. Soc. Nephrol. 2008, 3, 1599–1605. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  244. Vos, A.; Kockelkoren, R.; de Vis, J.B.; van der Schouw, Y.T.; van der Schaaf, I.C.; Velthuis, B.K.; Mali, W.P.T.M.; de Jong, P.A.; DUST study group. Risk factors for atherosclerotic and medial arterial calcification of the intracranial internal carotid artery. Atherosclerosis 2018, 276, 44–49. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  245. Alluri, K.; Joshi, P.H.; Henry, T.S.; Blumenthal, R.S.; Nasir, K.; Blaha, M.J. Scoring of coronary artery calcium scans: History, assumptions, current limitations, and future directions. Atherosclerosis 2015, 239, 109–117. [Google Scholar] [CrossRef] [PubMed]
  246. Tanaka, R.; Yoshioka, K.; Takagi, H.; Schuijf, J.D.; Arakita, K. Novel developments in non-invasive imaging of peripheral arterial disease with CT: Experience with state-of-the-art, ultra-high-resolution CT and subtraction imaging. Clin. Radiol. 2018, 74, 51–58. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  247. Goldberger, Z.D.; Valle, J.A.; Dandekar, V.K.; Chan, P.S.; Ko, D.T.; Nallamothu, B.K. Are changes in carotid intima-media thickness related to risk of nonfatal myocardial infarction? A critical review and meta-regression analysis. Am. Heart J. 2010, 160, 701–714. [Google Scholar] [CrossRef] [PubMed]
  248. Criqui, M.H.; Knox, J.B.; Denenberg, J.O.; Forbang, N.I.; McClelland, R.L.; Novotny, T.E.; Sandfort, V.; Waalen, J.; Blaha, M.J.; Allison, M.A. Coronary Artery Calcium Volume and Density: Potential Interactions and Overall Predictive Value: The Multi-Ethnic Study of Atherosclerosis. JACC Cardiovasc. Imaging 2017, 10, 845–854. [Google Scholar] [CrossRef]
  249. Kim, K.P.; Einstein, A.J.; Berrington de González, A. Coronary artery calcification screening: Estimated radiation dose and cancer risk. Arch. Int. Med. 2009, 169, 1188–1194. [Google Scholar] [CrossRef] [Green Version]
  250. Bucerius, J.; Dijkgraaf, I.; Mottaghy, F.M.; Schurgers, L.J. Target identification for the diagnosis and intervention of vulnerable atherosclerotic plaques beyond 18F-fluorodeoxyglucose positron emission tomography imaging: Promising tracers on the horizon. Eur. J. Nucl. Med. Mol Imaging 2019, 46, 251–265. [Google Scholar] [CrossRef] [Green Version]
  251. Blau, M.; Ganatra, R.; Bender, M.A. 18F-fluoride for bone imaging. Semin. Nucl. Med. 1972, 2, 31–37. [Google Scholar] [CrossRef]
  252. Dweck, M.R.; Chow, M.W.L.; Joshi, N.V.; Williams, M.C.; Jones, C.; Fletcher, A.M.; Richardson, H.; White, A.; McKillop, G.; van Beek, E.J.R.; et al. Coronary arterial 18F-sodium fluoride uptake: A novel marker of plaque biology. J. Am. Coll. Cardiol. 2012, 59, 1539–1548. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  253. Joshi, N.V.; Vesey, A.T.; Williams, M.C.; Shah, A.S.V.; Calvert, P.A.; Craighead, F.H.M.; Yeoh, S.E.; Wallace, W.; Salter, D.; Fletcher, A.M.; et al. 18F-fluoride positron emission tomography for identification of ruptured and high-risk coronary atherosclerotic plaques: A prospective clinical trial. Lancet 2014, 383, 705–713. [Google Scholar] [CrossRef] [Green Version]
  254. Irkle, A.; Vesey, A.T.; Lewis, D.Y.; Skepper, J.N.; Bird, J.L.E.; Dweck, M.R.; Joshi, F.R.; Gallagher, F.A.; Warburton, E.A.; Bennett, M.R.; et al. Identifying active vascular microcalcification by 18F-sodium fluoride positron emission tomography. Nat. Commun. 2015, 6, 7495. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  255. Hutcheson, J.D.; Maldonado, N.; Aikawa, E. Small entities with large impact: Microcalcifications and atherosclerotic plaque vulnerability. Curr. Opin. Lipidol. 2014, 25, 327–332. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. (a) Vascular smooth muscle cell (VMSC) phenotypic switching. Under physiological conditions, VSMCs display a contractile phenotype that regulates vessel structure and function. Under stress, arterial remodeling will occur, leading to VSMC phenotypic switching. Factors inducing the synthetic phenotype include platelet derived growth factors (PDGF), protease-activated receptors (PARs) and tumor necrosis factor-alpha (TNF-α). Synthetic VSMCs initiate vessel repair and can switch back to the contractile phenotype, driven by factors such as heparin or laminin. The osteogenic phenotype can be induced by prolonged exposure to BMP-2 or high phosphate. Osteogenic VSMCs shed extracellular vesicles that promote vascular calcification. Panel (b). Pathways affecting vascular calcification. Vascular calcification is an active process which can be initiated by several pathways, including: 1. biochemical factors, 2. physical factors, 3. vascular calcification (VC) inhibitors and 4. ECM factors. Biochemical factors, such as raised calcium and phosphate levels and PDGF, are stress molecules that induce VSMCs to switch towards a synthetic or osteogenic phenotype, including an increased release of extracellular vesicles. The ECM of the vessel wall directly influences VSMCs. Changes in collagen and elastin content cause VSMCs to change morphology and phenotype. VSMCs in turn, produce MMPs that induce structural changes in the vessel wall by rearranging collagen and elastin, promoting the migration and proliferation of VSMCs and other cell types. Physical factors such as shear stress and tensile stress affect ECs’ NO release, which influences the VSMC phenotype. Shear-induced stress induces a synthetic phenotype by decreasing a-SMA, MYH and smtn expression. Tensile stretch induces VSMCs to produce ECM proteins, such as collagen and fibronectin promoting vessel fibrosis. VC inhibitors, such as OPN, MGP and PP1, affect the VSMC phenotype by inducing changes in RNA expression patterns. Taken together, all stimuli play a part in an orchestrated VSMC response, ultimately promoting vascular calcification.
Figure 1. (a) Vascular smooth muscle cell (VMSC) phenotypic switching. Under physiological conditions, VSMCs display a contractile phenotype that regulates vessel structure and function. Under stress, arterial remodeling will occur, leading to VSMC phenotypic switching. Factors inducing the synthetic phenotype include platelet derived growth factors (PDGF), protease-activated receptors (PARs) and tumor necrosis factor-alpha (TNF-α). Synthetic VSMCs initiate vessel repair and can switch back to the contractile phenotype, driven by factors such as heparin or laminin. The osteogenic phenotype can be induced by prolonged exposure to BMP-2 or high phosphate. Osteogenic VSMCs shed extracellular vesicles that promote vascular calcification. Panel (b). Pathways affecting vascular calcification. Vascular calcification is an active process which can be initiated by several pathways, including: 1. biochemical factors, 2. physical factors, 3. vascular calcification (VC) inhibitors and 4. ECM factors. Biochemical factors, such as raised calcium and phosphate levels and PDGF, are stress molecules that induce VSMCs to switch towards a synthetic or osteogenic phenotype, including an increased release of extracellular vesicles. The ECM of the vessel wall directly influences VSMCs. Changes in collagen and elastin content cause VSMCs to change morphology and phenotype. VSMCs in turn, produce MMPs that induce structural changes in the vessel wall by rearranging collagen and elastin, promoting the migration and proliferation of VSMCs and other cell types. Physical factors such as shear stress and tensile stress affect ECs’ NO release, which influences the VSMC phenotype. Shear-induced stress induces a synthetic phenotype by decreasing a-SMA, MYH and smtn expression. Tensile stretch induces VSMCs to produce ECM proteins, such as collagen and fibronectin promoting vessel fibrosis. VC inhibitors, such as OPN, MGP and PP1, affect the VSMC phenotype by inducing changes in RNA expression patterns. Taken together, all stimuli play a part in an orchestrated VSMC response, ultimately promoting vascular calcification.
Ijms 20 05694 g001aIjms 20 05694 g001b
Figure 2. Vascular remodeling types. There are several types of arterial remodeling: hypotrophic, eutrophic and hypertrophic. Additionally, remodeling can be inward and outward. Hypotrophic remodeling results in a thinner vessel wall, which can be both inward and outward. In both cases, the wall-to-lumen ratio decreases. Hypertrophic remodeling results in the thickening of the vessel wall, that can also be inward and outward. The thickening of the vessel wall results in an increased wall-to-lumen ratio. In the eutrophic situation, wall-to-lumen ratios do not differ, but the size of the vessel can change. Atherosclerosis is characterized by an increased wall-to-lumen ratio, with both thickening of the media and intima and, therefore, is classified as inward or outward hypertrophic remodeling. Aneurysm formation is characterized by an increase in vessel diameter with a thinning of the vessel wall (outward hypotrophic remodeling). Inward remodeling is less frequent and is more associated with muscular peripheral arteries, reflecting sustained vasoconstriction of vessels.
Figure 2. Vascular remodeling types. There are several types of arterial remodeling: hypotrophic, eutrophic and hypertrophic. Additionally, remodeling can be inward and outward. Hypotrophic remodeling results in a thinner vessel wall, which can be both inward and outward. In both cases, the wall-to-lumen ratio decreases. Hypertrophic remodeling results in the thickening of the vessel wall, that can also be inward and outward. The thickening of the vessel wall results in an increased wall-to-lumen ratio. In the eutrophic situation, wall-to-lumen ratios do not differ, but the size of the vessel can change. Atherosclerosis is characterized by an increased wall-to-lumen ratio, with both thickening of the media and intima and, therefore, is classified as inward or outward hypertrophic remodeling. Aneurysm formation is characterized by an increase in vessel diameter with a thinning of the vessel wall (outward hypotrophic remodeling). Inward remodeling is less frequent and is more associated with muscular peripheral arteries, reflecting sustained vasoconstriction of vessels.
Ijms 20 05694 g002
Figure 3. Endothelial cell–smooth muscle cell communication. Endothelial cells (ECs) communicate with vascular smooth muscle cells (VSMCs) in two ways, called endothelial derived hyperpolarization (EDH). EDH has two major pathways: 1. Diffusion of nitric oxide (NO), which is produced by ECs and diffuses through the internal elastic lamina (IEL) to induce the relaxation of VSMCs. 2. Signaling through myoendothelial gap junctions (MEGJ) that connect ECs directly to VSMCs and cross the IEL. This direct communication between ECs and VSMC via MEGJ occurs via so-called endothelial derived hyperpolarizing factors (EDHF). 3. Major factors that stimulate EC NO secretion are flow or shear-stress alterations. Differences in wall shear rates are known to induce endothelial-derived changes in VSMC phenotype.
Figure 3. Endothelial cell–smooth muscle cell communication. Endothelial cells (ECs) communicate with vascular smooth muscle cells (VSMCs) in two ways, called endothelial derived hyperpolarization (EDH). EDH has two major pathways: 1. Diffusion of nitric oxide (NO), which is produced by ECs and diffuses through the internal elastic lamina (IEL) to induce the relaxation of VSMCs. 2. Signaling through myoendothelial gap junctions (MEGJ) that connect ECs directly to VSMCs and cross the IEL. This direct communication between ECs and VSMC via MEGJ occurs via so-called endothelial derived hyperpolarizing factors (EDHF). 3. Major factors that stimulate EC NO secretion are flow or shear-stress alterations. Differences in wall shear rates are known to induce endothelial-derived changes in VSMC phenotype.
Ijms 20 05694 g003
Table 1. Proteins that define VSMC phenotype.
Table 1. Proteins that define VSMC phenotype.
ProteinGene NameAbbreviationExpression
ContractileSynthetic
Alpha smooth muscle actinACTA-2⍺-SMA++
Smooth muscle myosin heavy chainMYH11SMMHC+
Smooth muscle 22 alphaTAGLNSM22-⍺+
SmoothelinSMTNSmtn+
CalponinCNN-1CNN-1+
Tumor necrosis factor alphaTNFATNF-⍺ +
S100 calcium binding protein A4S100A4S100A4+
Monocyte chemoattractant protein 1CCL2MCP-1+
+, positive effect; −, negative effect.
Table 2. Factors involved in VSMC phenotype switching.
Table 2. Factors involved in VSMC phenotype switching.
Factors Involved in VSMC Phenotype SwitchingPhenotype
Biochemical compoundsContractileSynthetic
PDGF+
TGF-β+
PARs+
TNF-⍺++
Angiotensin II++
Extracellular matrix components
Integrin: ⍺1β1, ⍺7β1, ⍺8β1+
Integrin: ⍺2β1, ⍺5β1, ⍺vβ3+
Collagen type I+
Collagen type IV+-
Elastin +-
Heparin+-
Fibronectin+
Laminin+
Physical factors
Tensile stress+
Shear stress+
Transcription
KLF4+
Oct4+
Abbreviations: PDGF, platelet derived growth factor; TGF-β, transforming growth factor beta; PARs Protease-activated receptors; TNF-⍺, tumor necrosis factor alpha; KLF4, krüppel-like factor 4; Oct4, octamer-binding transcription factor 4. +, positive effect; −, negative effect.

Share and Cite

MDPI and ACS Style

Jaminon, A.; Reesink, K.; Kroon, A.; Schurgers, L. The Role of Vascular Smooth Muscle Cells in Arterial Remodeling: Focus on Calcification-Related Processes. Int. J. Mol. Sci. 2019, 20, 5694. https://doi.org/10.3390/ijms20225694

AMA Style

Jaminon A, Reesink K, Kroon A, Schurgers L. The Role of Vascular Smooth Muscle Cells in Arterial Remodeling: Focus on Calcification-Related Processes. International Journal of Molecular Sciences. 2019; 20(22):5694. https://doi.org/10.3390/ijms20225694

Chicago/Turabian Style

Jaminon, Armand, Koen Reesink, Abraham Kroon, and Leon Schurgers. 2019. "The Role of Vascular Smooth Muscle Cells in Arterial Remodeling: Focus on Calcification-Related Processes" International Journal of Molecular Sciences 20, no. 22: 5694. https://doi.org/10.3390/ijms20225694

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop