info@biomedres.us   +1 (502) 904-2126   One Westbrook Corporate Center, Suite 300, Westchester, IL 60154, USA   Site Map
ISSN: 2574 -1241

Impact Factor : 0.548

  Submit Manuscript

Review ArticleOpen Access

Risk Factors, Biomarkers, Molecular and Cellular Mechanisms of Vascular Calcification Genesis and its Methods of Detection Volume 50- Issue 4

Leta Melaku*

  • Assistant Professor of Medical Physiology, Department of Biomedical Sciences, College of Health Sciences, Arsi University, Ethiopia

Received: May 01, 2023;   Published: May 25, 2023

*Corresponding author: Leta Melaku, Department of Biomedical Sciences, College of Health Sciences, Arsi University, Asella, Oromia, Ethiopia

DOI: 10.26717/BJSTR.2023.50.007982

Abstract PDF

ABSTRACT

Vascular calcification has traditionally been a passive process. However, in the last years, it has been proven to be an actively regulated biological process that is associated with crystallization of hydroxyapatite in the extracellular matrix and in cells of the media (VCm) or intima (VCi) of the arterial wall. Corresponding to bone mineralization, different pro and anti-calcifying mechanisms play an active role in mineral deposition in vascular cells. Evidence from clinical observations, animal models, and molecular studies suggest factors that regulate bone cell differentiation and mineralization, including fetuin A, BMP-2, matrix gla protein, osteopontin, osteoprotegerin, and inorganic pyrophosphate are used as biomarkers of vascular calcification. Under normal conditions, there is a balance between all calcification promoters and inhibitors, and it is possible that each pathological condition such as age, diabetes, dyslipidemia and hypertension, disrupts the balance with its own approach. The initiating factors and clinical consequences depend on the underlying disease state and the location of the calcification. Thereby, the pathogenesis of vascular calcification is a complex mechanism and not completely clear. On the other hand, diagnosis of the calcified arterial injury depends up on functional characteristics and imaging methods. In this article, the current knowledge of molecular and cellular mechanism, risk factors, biomarkers, and methods of detection of vascular calcification were reviewed.

Keywords: Vascular Calcification; Calcification Promoters and Inhibitors; Risk Factors; Clinical Assessment

Abbreviations: CT: Computed Tomography; VSMCs: Vascular Smooth Muscle Cells; IEM: Internal Elastic Membrane; CAD: Coronary Artery Disease; OPN: Osteopontin; OPG: Osteoprotegerin; TRAIL: TNF-Related Apoptosis-Inducing Ligand; OC: Osteocalcin; TNF: Tumour Necrosis Factor; ALP: Alkaline Phosphatase; Cbfa1: Core-Binding Factor Alpha 1; BMP: Bone Morphogenic Proteins; MBD: Mineral and Bone Disorders; VDN: Vitamin D Plus Oral Nicotine; AOPPs: Advanced Oxidation Protein Products; AV: Aortic Valve; MMP- 9: Matrix Metalloproteinase 9; TGF-β: Transforming Growth Factor-β; STC: Stanniocalcin; VC: Vascular Calcification; Cbfa-1: Core Binding Factor Alpha-1; RUNX2: Runt-Related Transcription Factor 2; MSX-2: Msh Homeobox 2; MGP: Matrix Gla Protein; MVSC: Multipotent Vascular Stem Cells; CASR: Calcium-Sensing Receptor; DXA: Dual-Energy X-Ray Absorptiometry; EBCT: Electron Beam Computer Tomography; MSCT: Multi-Slice Computer Tomography; MRI: Magnet Resonance Imaging; IVUS: Intravascular Ultrasound; OCT: Optical Coherence Tomography; AS: Arterial Stiffness

Introduction

Calcium is the most common element in bone, and 99% of the total body calcium is in bone in the form of a calcium phosphate crystalline structure called hydroxyapatite [Ca10[PO4]6[OH]2] [1]. Calcium is also found outside of bone in a variety of tissues, broadly termed extra-skeletal calcification. In these sites, the calcium can be in multiple forms- including hydroxyapatite, magnesium whitlockite, and amorphous calcium phosphate [2]. In vertebrates, all extracellular body fluids are supersaturated with respect to calcium and phosphate, resulting in a tendency for spontaneous calcium phosphate precipitation, which is often expressed as the calcium 3 inorganic phosphate product [3]. Vascular mineralization is a process in which mineral is pathologically deposited in blood vessels, mainly in large elastic and muscular arteries such as the aorta, coronaries, and carotid and peripheral arteries [4-7]. Arterial calcification can occur in both intimal (VCi) and medial layers (VCm). The two forms of vascular calcification are distinct in their morphology and pathology [8]. Intimal calcification is exclusively associated with atherosclerosis and morphologically appears as punctate and disorganized mineral deposition in the arterial wall intima. Intimal calcification forms an important part of atherosclerotic plaques, which constitute VSMCs, macrophages, lipid, connective tissue, and necrotic debris [9]. Intimal calcification is ubiquitously associated with atheroma, can be used as a surrogate marker of atherosclerosis [10,11], and is predictive of future cardiovascular events [12,13]. Calcification had been thought to occur late in the disease course. Non-contrast computed tomography (CT) is the most sensitive method to quantify vascular calcification, although it measures total vessel calcium content and does not distinguish between intimal and medial mineralization.

However, the wider consensus is that calcification in coronary arteries in the general population (non-diabetic and non-chronic kidney disease) predominantly affects the intima [14,15]. In contrast, the medial layer may also be affected, leading to thickening of the medial layer of larger elastic arteries resulting in arteriosclerosis of smaller elastic arteries classically described as Mönckeberg’s calcification, or medial calcinosis [16,17]. VCm confirmed by histology were observed in large elastic type arteries (ascending aorta), medium sized visceral and kidney arteries, and small transitional arteries (coronary, temporal, uterine, ovarian, parathyroid, mammary gland, and other) with diameter of at least 0.5 mm [18,19]. The four stages of lesion progression distinguish the extent and severity of Mönckeberg’s calcification (Table 1) [20-22]. In stage 1, calcifications appear on haematoxylin–eosin (H&E) staining as irregular blue or violet deposits embedded within the media [23]. On a high-resolution light microscopy using H&E, Elastica-van-Gieson, von Kossa, or Alizarinstaining deposits consisting of fine granulations, which increase in size and become confluent, are revealed. Both intra- and extracellular deposits are present. Intracellular deposits are in vascular smooth muscle cells (VSMCs); extracellular deposits are largely associated with damaged and fractured elastic fibres embedded within the extracellular matrix. In muscular and transitional arteries, with H&Estaining granular calcifications develop alongside the internal elastic membrane (IEM) and nearby VSMC.

Table 1. Stages of medial calcifications and histological aspects.

biomedres-openaccess-journal-bjstr

In experience, the involvement of the IEM is common. These bands of calcium-rich deposits may thicken, becoming solid plates extending deep into the inner layer of the media. With further progression of the disease calcifications may distort the junctions of the innermost and outermost layers of the media, spanning up to three quadrants of the cross section (stage 2) or it may involve the entire circumference (stage 3). In stages 2 and 3, large conglomerates of calcifications may form solid plates or sheaths, progressively distorting the architecture of the media; intrusions upon the intima are then common [24].

In the absence of atherosclerotic lesions, the intima shows a subendothelial hyperplasia. In stage 4 of VC m foci of bone formation within the arterial media may be found, calcifications may undergo osseous metaplasia giving rise to true bony trabeculae. These structures delineate true medullary spaces harbouring haematopoietic cells interspersed with adipocytes [25]. In the arterial wall, calcification deposits associated with VCm may be perceived as foreign bodies and induce granuloma formation within the media; these structures often contain multinucleated giant cells. Other inflammatory components such as foam cells, lymphocytes, and mast cells may also be present. In advanced stages, large calcifications may induce secondary changes in the intima such as subendothelial hyperplasia characterized by an increase in cellularity (e.g., myofibroblasts, fibroblasts, fibrocytes) and ulcerations characterized by infiltrations of the intima or even protrusions into the lumen.

It should be noted that VCm lesions do not spontaneously regress, and the clinical complications may vary according to the site and the amount of calcification. This disease of small vessels is also more common in patients with diabetes, renal failure and advanced aging [26]. The clinical manifestations of vascular calcification depend on the location within the arterial wall and the tissue perfused [27]. Intimal, atherosclerotic calcification can lead to myocardial infarction from stenosis and acute thrombus, or ischemia in both coronary and peripheral arteries. Medial or circumferential calcification can lead to reduced compliance due to arterial stiffening, resulting in an impaired vasodilation during ischemia that, in theory could lead to arrhythmias and sudden death. With medial calcification (arteriosclerosis) of the aorta, there will be increased pulse wave velocity, elevated pulse pressure, and systolic hypertension. Lastly, calcification of the arterioles of the skin and other organs can lead to calciphylaxis and ischemic gut [28]. Therefore, coronary calcium quantified by noncontrast CT can be taken as a measure of intimal calcification in the general population [14,15]. Numerous studies have investigated the association of cardiovascular risk with mVC in the general population as well as in diabetic, hypertensive and ESRD patients [29-31]. These studies have established in the general population, the amount of vascular calcification, as measured and quantified by multislice computed tomography, is an important predictor of all-cause mortality, vascular complications, and myocardial infarction [32].

On the epidemiological scenario, vascular calcification increases with age, atherosclerosis, renal failure, diabetes mellitus, hypercholesterolemia, osteoporosis, obesity, smoking, menopause, and lack of physical exercise [33,34]. Calcification of the aorta may affect 65% of people in a general population with a mean age of 60 years and correlates with coronary calcification identified by multidetector computed tomography, with a positive predictive value to increase cardiovascular morbidity and mortality in asymptomatic patients at intermediate risk [35]. Additionally, calcification of the abdominal aorta is associated with increased cardiovascular mortality, even when adjusted for age [35]. Conversely, calcification of the coronary arteries is associated with a greater risk of myocardial infarction and with increased incidence of adverse events during percutaneous and surgical myocardial revascularization [36]. Aortic valve sclerosis has a 40% prevalence in octogenarian patients [34], and initiates the process of calcific aortic valve stenosis, in which mineralization of the cusp has a pathophysiological mechanism like vascular calcification [37]. Calcific aortic valve stenosis is a predictor of cardiovascular risk in the elderly [38]. In opposition to control valves, calcified aortic valves express more alkaline phosphatase and matrix metalloproteinase 2 [39]. Despite the calcified degeneration of the aortic valve being associated with atherosclerosis and its risk factors, studies that used statins to treat patients with aortic valve stenosis did not demonstrate decreased aortic valve stenosis progression [40].

Calcification Promoters and Inhibitors

Calcification Inhibitors

Under normal conditions blood vessel cells express mineralizationinhibiting molecules [41]. The loss of their expression, as happens in CKD, causes what is known as “loss of natural inhibition”, giving rise to spontaneous calcification and increased mortality [41]. A list with these calcification inhibiting molecules has been drawn up after mutation analysis on mice, including among others:

Fibroblast Growth Factor-23 (FGF-23): FGF-23 is an approximately 30 kDA protein released by bone that requires the presence of the cofactor Klotho for its classical effects [42]. FGF-23 promotes phosphate excretion by reducing its proximal reabsorption by reducing the expression of NPT2a and NPT2c mRNA, sodium/ phosphate transporters [43]. FGF-23 also decreases conversion of calcidiol into its active form by reducing 1α-hydroxylase activity [44]. Thereby, gastrointestinal absorption of calcium and phosphate is reduced. In parathyroid glands, FGF-23 decreases PTH secretion and parathyroid cell proliferation [45]. FGF-23 null mice develop hypercalcitriolemia and VC [42]. Although the mechanistic link remains to be explained, FGF-23 may serve as a novel risk marker for the cardiovascular mortality in CKD [44]. In patients with coronary artery disease (CAD), the same independent link between FGF-23 and mortality has been demonstrated [46]. In contrast to FGF-23, Klotho excess has never been shown to be noxious [47]. Interestingly, Klotho levels are up regulated by vitamin D receptor agonists (calcitriol or paricalcitol) in CKD mice submitted to a high phosphate diet. These mice show half less calcification than those who did not receive therapy. Phosphaturia is increased whereas phosphatemia and FGF-23 levels are lowered [48]. In contrast, vascular Klotho deficiency favors the development of arterial calcification and mediates resistance to beneficial vascular effects of FGF-23 [47].

Fetuin-A (FET-A): Fet-A is a serum 59-kDa glycoprotein that inhibits ectopic vascular calcification [41] and produced by the liver that possess a systemic action [49,50]. It is a powerful inhibitor of hydroxyapatite formation, reducing the formation of crystals in in vitro solutions containing calcium and phosphorus without affecting those that are already formed [41,51]. Mice that are deficient in this protein develop extensive calcifications in soft tissue such as the myocardium, kidneys, tongue, and skin [52]. Fet-A is thought to inhibit calcification by binding early calcium phosphate crystals and by inhibiting crystal growth and mineral deposition [42]. This could be facilitated by the formation of large calciprotein particles [51,53]. Indeed, the accumulation of naked calcium phosphate crystals is responsible for extraosseous calcification and causes inflammation. These crystals are usually digested by the cells of the reticuloendothelial system such as macrophages. In contact with the crystals, macrophages secrete proinflammatory cytokines and undergo more apoptosis [42]. The formation of fetuin-A calciprotein particles (CPP) facilitates the clearance of these crystals and thereby reduces their negative impact. Fet-A likely plays a very important role in the stabilization of these complexes and reduces the inflammatory response [42]. Fet-A binds and sequesters insoluble mineral nuclei, forming soluble colloidal CPP, thereby inhibiting crystal growth and aggregation [42]. Macrophages secrete less cytokines and undergo less apoptosis phenomenon compared to reactions caused by naked crystals. This property of Fet-A to decrease inflammation may be influenced by the phosphorylation degree of the glycoprotein [54]. In these studies, lower serum Fet-A concentrations have been associated with increases in calcification scores, arterial stiffness, mortality and incidence of cardiovascular events [55-58].

Osteopontin (OPN): Osteopontin (OPN) is a phosphoprotein that is usually found in mineralized tissue such as bones and teeth [41,59]. It inhibits mineralization by blocking hydroxyapatite formation and activating osteoclast function [60]. Although it is not found in normal arteries, its expression is detected in atherosclerotic plaques and calcified calcified vessels. OPN knock-out mice do not develop VC but, when these mice are bred with MGP knock-out mice, the VCs are more important than in simple MGP knock-out mice [61]. OPN must be phosphorylated to act as a calcification inhibitor [42,62]. OPN inhibits mineralization of VSMC by binding to the mineralized crystal surface [63]. On the contrary to the fully phosphorylated OPN, cleaved OPN could act as a proinflammatory cytokine and a proangiogenic factor facilitating vascular mineralization [60,64]. The possibility that OPN could serve as a calcification serum marker is controversial [42]. Berezin et al showed that OPN was a good predictor of coronary calcification in type two diabetes mellitus patients [65]. Tousilis et al found a positive association between OPN and arterial stiffness in coronary artery disease [66]. Indeed, the discrepancy between the different studies may perhaps be explained by the differences in patient populations. It is thought OPN plays a key role in inflammatory process [42]. Its relationship with diseases related to inflammation such as atherosclerosis, obesity and autoimmune diseases has already been shown [67-69]. It has also been suggested that hyperglycemia could up-regulate OPN and thereby lead to VSMCs proliferation [70].

Osteoprotegerin [O]: Osteoprotegerin [OPG] is a member of the tumour necrosis factor receptor family that has been identified as a regulator of bone resorption [71]. OPG is produced by many tissues, including the cardiovascular system, lungs, kidney and immune system [72]. OPG is a regulatory factor produced by bone marrow derived stromal cells [42]. OPG plays a pivotal role in the regulation of the bone turnover, inhibiting osteoclast differentiation and acting like a decoy receptor for the receptor activator of NF-КB ligand (RANKL system) [73]. It interferes with the interaction between RANK (expressed by osteoclast-like cells) and RANKL (expressed by osteoblast-like cells). OPG is also thought to inhibit alkaline phosphatase activity [74]. OPG levels are significantly higher in CKD patients, in relation with the severity of renal failure. Although OPG is known to impede osteoclast differentiation in bone, OPG is usually considered as a protective factor against VC as it blocks the bone remodeling process in the vascular tissue [42]. OPG is also a neutralizer of the pro-apoptotic actions of TRAIL (TNF-related apoptosis-inducing ligand), which strongly activates vascular cells apoptosis [75]. Apoptotic bodies can also lead to mineralization. In support of that, it has been observed that OPG deficient mice do develop both severe aortic calcifications and osteoporosis [76,77]. Interestingly, OPG seems to be a marker of VC onset rather than a severity or progression predictor [42,78]. Osteocalcin (OC): OC, a vitamin-K dependent matrix protein that inhibits calcium salt precipitation in vitro [79], shows a strong affinity for hydroxyapatite [42]. OC has been found in calcified atherosclerotic plaques and calcified aortic valves [80]. It was generally thought that OC inhibits crystal growth [81] and limits bone formation [82]. Nonetheless, its utility as serum marker is still discussed in conflicting studies. Aoki, et al. [83] did not show any relationship between OC and VC in type 2 diabetes mellitus patients whereas Kim, et al. [84] found an inverse correlation between OC and Agatston calcification score in Asian women, even after adjusting for age [42]. To define if OC can be used as a diagnostic or a screening tool, the role of OC in the pathogenesis of VC clearly remains to be clarified.

Pyrophosphate (PPi): PPi is a small molecule made of two phosphate ions [42]. It acts as a calcification inhibitor by inhibiting hydroxyapatite crystal formation [85]. Once again, knock-out mice (in fact, knock-out mice for a precursor) develop VCs [86]. Absence of PPi would promote VSMC differentiation but the mechanism is not fully understood [87,88]. O’Neill, et al. demonstrated the negative association between PPi and VC in CKD [89]. Although the short halflife of PPi limits the possibility for improving VC by bolus injections, daily peritoneal dialysis achieved with a solution which contains PPi in CKD mouse model do succeed in inhibiting calcification [90]. O’Neill et al demonstrated that daily intraperitoneal injections in rats could also reduce both incidence and amount of calcification [91]. PPi has been shown to inhibit mineralization on rat aortic VSMCs cultures too [92]. Furthermore, biphosphonates, non-hydrolysable analogs of PPi, have also proved their ability to inhibit aortic calcifications in experimental renal failure rats. Calcification was stopped in cultures of rat aortas as well as in vivo model [42]. It supports the idea that biphosphonates have direct effects on VC, independent of bone [93], maybe via a down regulation of Notch1-RBP-Jk signaling pathway and MsX2 gene induction [94]. ATP, which is a polyphosphate associated with nucleoside, might also act as calcium phosphate deposition inhibitor, not only as the source of PPi but also as a direct inhibitor [95]. Even if PPi seems to be a promising marker, its determination has been performed in a single center only and the transferability to other centers should be validated.

Matrix Gla Protein (MGP): MGP is a vitamin K, 14-kDa γ-carboxylated protein expressed by chondrocytes, VSMCs, endothelial cells and fibroblasts [42]. Its role as a calcification inhibitor has been illustrated by MGP knock-out mice who develop extensive arterial calcifications [96,97]. In 2002, Moe et al demonstrated a correlation between vascular MGP expression and the calcification of epigastric arteries in dialysis patients [98,99]. MGP-deficiency in humans leads to Keutel syndrome, a rare genetic disease hallmarked by abnormal soft tissue calcification [96]. MGP binds calcium crystals, inhibits crystal growth, and plays a role in the normal phenotype of VSMCs in preventing the osteoblastic differentiation [100,101]. MGP also binds and inactivates a pro-mineralization factor, bone morphogenetic protein-2 (BMP-2) [102]. Among other effects, BMP- 2 promotes osteogenic conversion of VSMCs via MSX2 transcription factor [42]. MGP could also protect mineral nucleation sites on elastin and thereby prevent spontaneous calcification of the elastic laminae [42]. In support of that, the irregular calcification of the thoracic and abdominal aorta segments in MGP −/− mice correlates with the local variations of the elastin content [96]. Parallel to this study, other authors hypothesized a mineralization process by size exclusion, in which MGP proves to be essential to prevent mineralization within fibrils [42].

Calcification Activators

There are studies that speculate that, as well as hyperphosphataemia and hypercalcaemia, there are substances present in the blood serum of patients with CKD capable of stimulating calcification [103]. Bovine VSMC in the presence of uraemic serum increases the expression of calcification-related proteins. Many uraemic factors have been identified that can induce osteogenic genes, transforming osteoblasts and secreting some bone matrix proteins in the walls of blood vessels and soft tissue. Some of these factors are tumour necrosis factor (TNF) [104], inflammatory cytokines [105], fibronectin [106], type-I collagen [106] and 25-hydroxycholesterol [107]. These uraemic serum substances stimulate the expression of molecules essential to vesicular calcification.

Alkaline Phosphatase: Alkaline phosphatase (ALP) is one of the osteoblastic phenotype markers and is considered essential in the vascular calcification process [41] t has been detected in vascular and heart valve calcifications. ALP expressed on the surface of cells can act on phosphate liberators, releasing inorganic phosphate [108] Inflammatory cytokines and vitamin D induce its up-regulation and mineralization [109].

Core-Binding Factor Alpha 1: Core-binding factor alpha 1 (Cbfa1) is the main regulator of bone cell differentiation [41]. fa1- deficient mice have problems with cartilage formation and bone mineralisation [110]. acts as a transcription factor that accelerates the expression of important osteoblast lineage genes such as osteocalcin, osteopontin, ALP or type-I collagen [111]. s expression is up regulated by phosphate43 and uraemic toxins [103].

Bone Morphogenetic Protein – 2 (BMP-2): Bone morphogenic proteins (BMP) are a group of, at least, 30 proteins that receive their name from their osteoinductive properties [41]. Bone morphogenetic proteins (BMPs) belong to a subdivision of TGF-β like growth factors family. BMPs regulate growth, differentiation, and development in the embryo as well as during tissue remodeling processes in the adult organism. BMP-2 is an important molecule iVC [he regulation of bone formation as well as in VC [41,42]. In bone, it promotes osteoblast differentiation and mineralization [112]. Inhibition of BMP-2 inhibits osteoblast differentiation and bone formation in vivo and in vitro [113] and protects against atherosclerosis and VC [114]. They act by binding to a heterodimeric system of transmembrane receptors (BMP- 1 and BMP-2 receptor) that trimerises upon binding. The binding of a BMP to its specific type II receptor results in the type 1 receptor being activated. This causes phosphorylation and nuclear translocation of the Smad transcription factors thus modifying the transcription rate of target genes [115]. They then induce ectopic bone formation [116]. Sclerostin: Sclerostin is an osteocyte-specific glycoprotein and is considered as a potent inhibitor of bone formation [117,118].

It inhibits specific co-receptors needed for β-catenin-dependant signaling activation [119]. This pathway is involved in osteoblastmediated bone formation [120]. It is thought that sclerostin plays a role in bone mechanosensibilisation [42]. When bone undergoes a substantial strain, sclerostin production would be decreased and bone could thus increase its formation in response to mechanical stress [121]. As β-catenin belongs to Wnt cascade signaling and as Wnt pathway is thought to be implicated in development of VC, it is interesting to investigate a potential association between sclerostin levels and VCs [42]. In non-CKD patients, some studies have demonstrated a positive association between sclerostin levels and VC [122,123] whereas in other ones, there was not a significant correlation between the two parameters [124,125].

Rankl: RANKL (also known as OPGL) is a protein consisting of 316 amino acids with a molecular weight of 38kD. Its expression is also modulated by several cytokines, glucocorticoids and PTH [126]. RANKL is produced by osteoblast lineage cells and activated T cells. It promotes osteoclast formation, fusion, differentiation, activation and survival, leading to increased bone resorption and bone loss [127]. RANKL stimulates its specific receptor RANK, which is expressed in fewer cells such as progenitor cells and mature osteoclasts, activated T cells and dendritic cells [128-130]. The activation of RANK by RANKL triggers the NF-ΚB intracellular signalling cascade. The final stage of RANK activation is the NK-κB translocation into the nucleus, which can take place by the classical or alternative pathway [41]. Both pathways are regulated by their kinases which are, respectively, IKK‚ and IKKα. The NK-κB translocation to the nucleus modulates the expression of different genes, e.g., BMP4 [131]. The biological effects of OPG are the opposite of RANKL-mediated effects, since OPG acts as a soluble inhibitor that prevents RANKL interaction and the subsequent stimulation of its RANK receptor [132]. Many trials have shown that VC as well as arterial stiffness and cardiovascular events are inversely related to serum RANKL [133-135] and positively related to serum OPG [136-138].

Risk Factors for Vascular Calcification

The risk factors for VC are divided into traditional: involving advanced age, hypertension, diabetes, smoking, dyslipidemia, and others; and the non-traditional ones: including inflammation, oxidative stress, and mineral and bone disorders (MBD) of CKD, among other factors [139].

Age

Age is the strongest predictor of coronary artery disease [140] but multiple other clinical risk factors have been implicated in the pathogenesis of arterial calcification. Coronary artery calcification is also more prevalent and more severe among CKD patients than in the general population, and studies in CKD patients offer insight into the pathogenesis. In patients not yet on dialysis, over 50% have coronary artery calcification [141] whereas 70–90% of prevalent dialysis patients have significant coronary artery calcification [142,143]. Histologic studies comparing dialysis patients to non-CKD patients who died of a coronary event showed that dialysis patients had more calcification in the atheromatous plaques, but not more plaque. Dialysis patients also had a thicker medial layer [144]. Studies evaluating distal segments of the coronary arteries found medial calcification adjacent to the internal elastic lamina in dialysis patients [14] and in patients with advanced CKD [15]. Moe et al had found isolated medial calcification in the absence of intimal calcification in the inferior epigastric artery of patients undergoing a renal transplant [98]. Thus, calcification can occur both in intimal and medial arterial layers and in different vascular beds. In a study of 4544 patients, the presence of calcification in the thoracic aorta, carotids and iliac arteries were associated with all-cause mortality with hazard ratios of 2.1, 1.6, and 1.67, respectively, whereas coronary artery calcification was associated with a hazard ratio of 3.4 for cardiovascular mortality [145]. At the present time, it appears that there may be different initiating factors in different vascular beds and in the intima and media, but a common downstream process of de-differentiation to an osteoblast like phenotype.

Hypertension

Hypertension is associated with vascular remodelling and arteriosclerosis. In clinical studies, hypertension is not a commonly cited risk factor for calcification, perhaps because most subjects with calcification have hypertension as a clinical manifestation of the arteriosclerosis. The renin-angiotensin system is known to be a major pathogenic factor in VSMC apoptosis, growth, and differentiation, and therefore it likely plays a role in calcification [146]. Armstrong et al fed rabbits an atherogenic diet with high dose vitamin D to induce calcification along the internal elastic lamina and the media layer. There was upregulation of BMP-2 and down regulation of alphasmooth muscle actin suggesting a dedifferentiation from a vascular smooth muscle cell phenotype to an osteoblast like phenotype. Furthermore, calcified arteries had upregulation of angiotensin 1 receptor and treatment with an angiotensin receptor blocker prevented the calcification [147]. In contrast, in 5/6th nephrectomized rats (a model of CKD), treatment with enalapril improved myocardial hypertrophy and progression of renal disease but had no effect on vascular calcification [148]. In a rat model of arterial calcification induced by intramuscular administration of vitamin D plus oral nicotine (VDN), increased calcium content of arteries was associated with increased levels of angiotensin II and adlosterone in the tissue; treatment with captopril or spironolactone reduced the calcification [149]. Thus, the renin-angiotensin and aldosterone pathway appear to play a role in arterial calcification. Whether this is due to the reduction of underlying remodelling (arteriosclerosis), or a direct inhibition of the osteogenic transformation will require additional studies.

Diabetes

Patients with diabetes had increased calcification compared to non-diabetic patients and there was increased expression in the medial layer of bone matrix proteins in the arteries such as osteopontin, type I collagen and alkaline phosphatase [150]. In vitro, there is a study that found VSMC incubated with high glucose led to an increase in the expression of the osteoblast transcription factor RUNX2, BMP-2 and osteocalcin and enhanced calcification in bovine VSMC. The protein kinase C signaling pathway was involved in this high glucose-induced expression of RUNX2 and bone matrix proteins [150]. Another group found that when fed high fat diet, the Ldlr−/− diabetic mouse develops hyperglycemia, dyslipidemia and aortic calcification with concomitant upregulation of aortic BMP2 and Msx2 gene expression [151]. Increased glucose increased the BMP-2/Msx2- Wnt pathway, leading to an osteogenic phenotype in a subset of the myofibroblasts; inhibition of the BMP-2 pathway reduced arterial calcification [152]. Interestingly, the location of BMP2 and BMP4 differed in diabetic aortas in that BMP-4 was found in the endothelium and BMP2 throughout the vascular wall [152]. These results suggest that the increased vascular calcification in diabetes is at least partially due to the direct effects of hyperglycemia on transforming the VSMC to osteoblast like phenotype via multiple mechanisms.

Dyslipidemia

Although clinically the role of lipids in vascular calcification is unclear, during osteogenic differentiation, calcifying vascular cells (CVCs, a clone of VSMC that readily calcify) accumulate not only minerals but also lipids such as triglycerides [153]. In vitro, HDL inhibits the osteogenic differentiation pathway [153]. In CVCs, stearate, compared to other fatty acids, promoted mineralization whereas inhibition of acetyl-CoA carboxylase or acyl-CoA synthetase reduced mineralization [154]. In these same CVC, n-3 unsaturated fatty acids play a protective role through a p38-MAPK (mitogenactivated protein kinase) and PPARγ (peroxisome proliferator activated receptor gamma) dependent mechanism [155]. Finally, oxidized lipids such as oxysterols and oxidized phospholipids illicit procalcific effects in vascular cells as detailed below [156]. Thus, dyslipidemia, rather than elevated LDL cholesterol appears to be a major causative factor in vascular calcification.

Inflammation

Inflammation is a known non-traditional risk factor for atherosclerosis and vascular disease in the normal population and in CKD and is associated with increased mortality [157]. Both CRP [158] and inflammatory cytokines [159] are associated with increased coronary artery calcification in patients with CKD. Interestingly, osteogenesis is associated with local inflammation and macrophage infiltration in atherosclerosis in ApoE−/− mice as revealed by molecular imaging in vivo [160]. Tumor necrosis factor alpha can induce mineralization of calcifying vascular cells in vitro [104] and co-culture of these cells with monocyte/ macrophages (the source of most cytokines) can accelerate mineralization [161]. In human VSMC, the phosphatidylinositol 3-kinase (PI3K)/Akt pathway may inhibit inflammation induced calcification, perhaps by mediating alkaline phosphatase which is a ‘marker’ of osteoblast phenotype but also a potent inhibitor of a naturally occurring inhibitor of calcification, pyrophosphate [162]. Cytokine stimulation of alkaline phosphatase from VSMCs probably plays an important role also in calcification associated with diabetes, since the TNF-α inhibitor infliximab was shown to reduce the osteogenic phenotype of VSMC and the extent of medial calcification in LDLR−/− diabetic mice, without reducing obesity, hypercholesterolemia, and hyperglycemia [151].

Oxidative Stress

CKD is a state of increased oxidative stress due to impaired antioxidative mechanisms [163]. Elevations in asymmetric dimethylarginine, a naturally occurring inhibitor of NO synthase, are associated with increased intima-medial thickness in the carotid arteries, concentric left ventricular hypertrophy, and mortality in dialysis patients [164]. In a rat model of CKD, the antioxidant Tempol inhibited vascular calcification by reducing oxidative stress and inhibiting osteogenic transdifferentiation of vascular smooth muscle cells [165]. In the general population, there is growing evidence indicates that there is a correlation between oxidative stress and the development of vascular calcification [166-168]. Macrophages, endothelial cells, and smooth muscle cells produce reactive oxygen species such as hydrogen peroxide and superoxide anion in response to several stimuli. The free radical nitric oxide (NO) is generated from the endothelium from L -arginine by the enzyme NO synthase and leads to production of hydroxyl or peroxyl radicals. When VSMC are treated with β-glycerophosphate or uremic serum for 24 h, the production of H2O2 and early expression of NADPH oxidase sub-unit p22(phox) are increased. The elevated oxidative stress was associated with increased expression of RUNX2 and alkaline phosphatase and calcification of VSMC [166]. An important contributor to oxidative stress in atherosclerotic lesions is the formation of hydrogen peroxide from various sources in vascular cells [37]. A recent study by Byon et al [168] demonstrated that H2O2 induces a switch of VSMC from contractile to osteogenic phenotype associated with an increased expression of RUNX2 and calcification in VSMC. Furthermore, inhibition of H2O2-activated AKT signaling pathways blocked increased expression of RUNX2 and VSMC calcification [168]. A similar study has also demonstrated that advanced oxidation protein products [AOPPs] induce vascular calcification by promoting osteoblast differentiation of human vascular smooth muscle cells via the ERK signaling pathway [167]. In a rabbit model of atherogenesis fed high dose vitamin D, there was increased oxidative stress and aortic valve (AV) calcification/stenosis. The latter could be abrogated by the antioxidant lipoic acid [37]. Lipid oxidation products have direct effects on both bone forming and bone-resorbing cells. Oxidized LDL directly inhibits differentiation of osteoblasts [156] while directly inducing differentiation of osteoclasts [169]. Oxidized lipids also regulate osteoclastogenic cytokines produced by osteoblasts [170]. Thus, oxidative stress may be causative in vascular calcification, and may also explain the relationship between increased coronary artery calcification and osteoporosis found in both CKD and the general population [171].

Advanced Glycation End-Products (AGEs)

Proteins can be modified indirectly by reactive carbonyl compounds formed by auto-oxidation of carbohydrates and lipids, leading to eventual formation of AGEs. AGEs have been found in arterial and cardiac tissue as well as atherosclerotic lesions in dialysis patients [172]. Circulating AGEs such as pentosidine are elevated in patients on dialysis [173]. AGE-modified elastin and calcification has been found to co-localize in the aortic media of dialysis patients and binding of mineral to elastin is thought to be an important factor in the pathogenesis of medial calcification [174]. In cultured VSMC, AGEs can accelerate calcification of microvascular pericytes [175]. AGEs induced the expression of RUNX2 mRNA and alkaline phosphatase activity and calcification [176]. The receptor for AGE (RAGE) is expressed in a variety of cells including VSMC [177] and these AGE mediated changes in VSMCs were partially attenuated by a neutralizing antibody to RAGE [178]. A study by Suga, et al. [179] demonstrated that activation of RAGE inhibited VSMC phenotypic gene expression and induces osteogenic differentiation of VSMC. This RAGE mediated effect was via Notch/Msx2 induction in VSMC. The results suggest that AGEs that accumulate in diabetes could elicit the osteoblastic differentiation of VSMCs, thereby contributing to vascular calcification via the RAGE pathway.

Abnormal Mineral Metabolism

Hyperphosphatemia: Abnormal mineral metabolism has been recognized as a nontraditional risk factor in the development of vascular calcification in CKD patients and is associated with increased mortality in both pre-dialysis and dialysis patients [27,180]. Hyperphosphatemia is associated with the prevalence and progression of vascular calcification in dialysis patients [181]. Several studies have demonstrated that the use of noncalcium-based as compared with calcium-based phosphate binders attenuated vascular calcification and mortality in dialysis patients [182,183]. In the general population, phosphorus levels in the upper quartile of the normal range are also associated with increased cardiovascular and all-cause mortality [184]. In vitro, phosphate increased the calcification of VSMC in dose-dependent manner [185]. High phosphate induced the loss of VSMC markers, such as smooth muscle (SM) α-actin and SM22α and increased the expression of the osteochondrogenic markers Runx2, osterix, osteopontin, and alkaline phosphatase [62,186]. A recent study has demonstrated that bovine VSMC incubated with calcification media (10 mM β-glycerolphosphate as a phosphate donor) generated cellular matrix vesicles that have high annexin II and VI content and the ability to mineralize extracellular matrix compared to that from bovine VSMC incubated without phosphate [187]. The matrix vesicles serve as nucleation sites for calcification, like the vesicles that bud from osteoblasts and hypertrophic chondrocytes in normal bone formation. Phosphate transport to cells is primarily mediated by sodium-dependent phosphate (NaPi) cotransporters [188] and treatment with phosphonoformic acid (PFA, a competitive inhibitor of NaPi transport) inhibits phosphate uptake and VSMC osteochondrogenic differentiation [185].

The type III NaPi co-transporters, PiT-1, is highly expressed in VSMC [189] and the knockdown PiT-1 with siRNA suppressed phosphate induced calcification and blocked induction of the osteogenic markers Runx2/Cbfa1 and osteopontin [189]. However, our group has shown that bovine VSMCs incubated with pooled uremic sera from dialysis patients had increased calcification, above that induced by phosphorus but only when phosphorus is available [190]. The addition of PFA (inhibitor of NaPi transport) or levamisole (inhibitor of alkaline phosphatase) only partially inhibited uremic serum-induced osteopontin upregulation. The cyclic adenosine monophosphate (cAMP)/protein kinase A signaling pathway was involved in uremic serum-induced upregulation of RUNX2 and alkaline phosphatase [191]. High phosphate may also regulate matrix mineralization through elastin degradation. A soluble elastin-derived peptide can induce mineralization of human VSMCs in the presence of high phosphorus concentration [192]. Treatment of rat VSMC with elastin peptide induced the expression of elastin–laminin receptors along with increased expression of osteoblastic transcription factor RUNX2 and alkaline phosphatase [193]. TGF-β which is known to upregulate RUNX2 [194], had synergistic effect on VSMC phenotypic change. In a rat aortic ring model, treatment with high phosphate and warfarin increased matrix metalloproteinase 9 (MMP-9) activity followed by transforming growth factor-β (TGF-β) signaling and aortic calcification [195]. One recent study demonstrated that MMP- 2 and MMP-9 expression and activity are increased with progressive CKD, and blockade of MMP activity can inhibit arterial calcification [196]. This matrix degradation or alteration may be an initial step in calcification.

Hypercalcemia: There is an association with elevated serum calcium and the development of vascular calcification in the CKD population [180]. In addition, the use of calcium containing phosphate binders which induce positive calcium balance is associated with increased arterial calcification in the majority of studies [197]. In vitro, calcium alone can increase human VSMCs calcification [198]. Furthermore, calcium and phosphorus had synergistic roles in inducing mineralization of VSMC [199]. In an aorta ring culture model, elevated calcium was more potent than phosphorus to induce VSMC calcification for a given concentration of calcium and phosphorus, called the Ca × P product [200,201]. Calcium also stimulates VSMC matrix vesicle release [199]. Study by Chen et al have demonstrated that calcified VSMC derived cellular MV are enriched with annexin II and VI but with little fetuin-A [187]. Furthermore, blockade of annexin calcium channel activity with K201or the L-type calcium channel blocker verapamil significantly inhibit MV activity and the calcification of VSMC [202]. Shanahan and colleagues also demonstrated that blockade of intracellular calcium increase can inhibit MV calcification [1]. Abnormal mineral metabolism contributes to the development of vascular calcification by multiple mechanisms.

Fibroblast Growth Factor 23: The hormone fibroblast growth factor 23 (FGF-23) is predominately expressed in osteocytes and is involved in mineral homeostasis by inducing hyperphosphaturia, inhibiting calcitriol synthesis and inhibiting PTH secretion [203]. In the kidney it exerts its biological functions by binding to the FGF receptor in the presence of the cofactor Klotho [204]. Several studies have demonstrated that FGF-23 is associated with coronary artery and aortic vascular calcification in CKD and dialysis patients [205,206]. Targeted deletion of FGF-23 or Klotho in mice resulted in hyperphosphatemia and vascular calcification [204,207]. In moderately uremic mice fed high-phosphate diets, elevated serum FGF-23 and osteopontin levels, but not serum phosphorus levels, were associated with extensive arterial-medial calcifications [208]. A recent study by Takei et al has demonstrated that the expression of stanniocalcin (STC) 2, a calcium/phosphate-regulating hormone, is increased and colocalized in calcified lesions of FGF-23 or Klotho null mice [209]. Although the mechanism by which FGF-23 affects vascular calcification is not clear at present, these data suggest that another mechanism by which phosphate affects vascular calcification may be through phosphorus-mediated elevation of FGF-23 levels.

Molecular and Cellular Mechanism of Arterial Calcification

Occurrence of vascular calcification (VC) has been discovered in the “Iceman” who lived 5000 years ago [210] and scientists had already paid attention to this phenomenon and to its relationship with renal disease in the 19th century [211]. Traditionally, two major forms of ectopic [pathologic] calcifications were distinguished; dystrophic refers to VC occurring in damaged tissues while metastatic was associated with systemic disorders of calcium and phosphate metabolism; these descriptions reflect the differences between vascular ossifications [active process] and petrifications [passive process] described by Virchow [212]. Initially VC was considered to be a passive process, the result of Ca2+ and phosphate ions exceeding solubility in tissue fluid, thereby inducing the precipitation and deposition of hydroxyapatite crystals [213]; however, VC is now considered as an active process that is complex, actively regulated via a variety of molecular signalling pathways and associated with crystallization of hydroxyapatite in the extracellular matrix and in cells of the media (VCm) or intima (VCi) of the arterial wall by involving the differentiation of macrophages and vascular smooth muscle cells (VSMCs) into osteoclast-like cell [214-216]. While considerable progress elucidating the signalling pathways regulating VC formation has been achieved, the exact molecular basis of VC remains elusive [217,218]. Within coming new research data, the already large number of molecular mechanisms suggested to contribute to VC formation continues to grow. The mechanism of arterial calcification is complex, but multiple investigators agree that the first step appears to be de-differentiation or transformation of vascular smooth muscle cells (VSMC) to an osteoblast/chondrocytic phenotype (Figure 1).

Figure 1.

biomedres-openaccess-journal-bjstr

VSMC originate from a similar mesenchymal stem cell as osteoblasts, the latter occurring with up-regulation of the transcription factor core binding factor alpha-1 (Cbfa-1) now called Runt-related transcription factor 2 (RUNX2) or msh homeobox 2 (MSX-2). These cells then do what a normal osteoblast does: secrete matrix proteins [98]. The signals that induce this transformation are multiple and once the matrix is laid down, these cells then mineralize the matrix through the secretion of matrix vesicles [187], or through apoptosis [219]. Phosphorus and calcium increase the mineralizing potential of these matrix vesicles [187,202]. It appears that while deposition of hydroxyapatite represents the resulting commonality of VC, different initiating and propagating molecular mechanisms, as well as diverse crystalline compositions of calcium apatite crystals may be present in various forms of VC [220-222]. The underlying pathophysiological mechanisms resulting in VC can be broadly described as: genetic predisposition certainly plays an important role in the genesis of this phenomenon [213]. According to Rutsch et al, 40 – 50% of cases of coronary calcification can be attributed to genetics [223]. Genes ENPP1 and NT5E are respectively implicated in infancy and idiopathic VC. The first one encodes a protein which transforms ATP to adenosine and pyrophosphate (PPi, inhibitor of calcification) whereas the second one converts AMP into adenosine and inorganic phosphate (Pi, accelerator of mineralization) [4,214].

The VC phenotype caused by mutations in these genes underlines the role of PPi and Pi in pathogenesis. Mutations in ABCC6, a gene encoding a nucleoside-sensitive transporter, have also been linked to hereditary calcification [42]. Alternative action of ABCC6 may include deficient hepatic production of inhibitory factor of matrix Gla protein (MGP), an important inhibitor of calcification [224]. Another major mechanism of development of VCs is looks like process through which bone formation occurs (Figure 1) [215,225]. First, vascular smooth muscle cells (VSMCs) undergo osteogenic differentiation into phenotypically distinct osteoblast-like cells [103,225]. In the case of renal failure, phosphate plays a key role in this mechanism [226]. In vitro, high extracellular phosphate concentrations induce a rise in intracellular phosphate concentration which is actively mediated by Pit-1, a sodium dependent phosphate co-transporter [185,189]. This increasing phosphate concentration in the VSMC induces a phenotypic switch of VSMCs into osteoblast-like cells [185,186,225]. The protein Cfba1/Runx2 [core-binding factor subunit 1α/runt-related transcription factor 2] is a specific and indispensable transcriptional regulator for this osteoblastic differentiation. Its expression is also enhanced with high extracellular phosphate [100,185,186]. These “new” cells will express alkaline phosphatase (ALP), secrete, under the control of Cfba-1, bone-associated proteins (such as osteopontin [227], collagen type 1, osteoprotegerin, bone morphogenic protein-2 and osteocalcin [228]) and release mineralization-competent MVs in the extracellular matrix [199,226]. VSMCs release MVs under normal physiological conditions and these MVs are protected from mineralization by the presence of calcification inhibitors [42]. Under pathological conditions, a combination of factors makes the MVs “mineralization competent” [229].

Moreover, an increase of intracellular phosphate level mediated by Na/Pi transporter is thought to induce VSMC apoptosis through an unclear process that possibly involves a disruption in mitochondrial metabolism [230]. Some studies suggest that apoptosis leads to calcification [231,232]. The MVs, in which proapoptotic factor BAX (BCL2-associated X protein) have been identified [233], may be remnants of apoptotic cells. As MVs have the capacity to concentrate and crystallize calcium, apoptosis could be a key regulator of VC [230]. More recently, a different point of view has emerged according to which phenotypically distinct osteoblast-like cells might originate from stem cells rather than VSMCs [42]. A new mechanism called “Circulating cell theory”, suggesting an active role for circulating cells arising from sources such as bone marrow, has been postulated to contribute towards VC. It is well known that the extracellular fluid is a metastable soup about calcium and phosphate concentrations and that active inhibitors of calcification must be present, both circulating and locally, to prevent the spontaneous formation of apatite: a situation that certainly applies to the CKD population [23]. The active inhibition process involves vascular smooth muscle cells and several proteins, including some that are vitamin K-dependent. Under the influence of chemo-attractants [released by damaged endothelium for instance], these bone marker-positive cells may home to diseased arteries. Under pathologic conditions such as an imbalance between promoters and inhibitors of VC, this population may further undergo osteogenic differentiation in the lesions, which could promote vessel mineralization [42,234]. Another recent study has also claimed that multipotent vascular stem cells (MVSC) present in blood vessel wall might differentiate into osteoblast-like cells [235].

Nevertheless, this point of view is still very controversial. Although the role of phosphate is well established in osteoblastic differentiation process, many other factors can influence this conversion and accurate causal mechanisms remained not completely understood. Under normal conditions, VSMCs produce endogenous inhibitors of calcification such as matrix Gla protein (MGP), osteopontin, osteoprotegerin and pyrophosphate [6]. A long-term exposure of VSMCs to a variety of stresses can overwhelm the action of these inhibitors and induce differentiation [229]. Among these chronic stresses, ionic disorders (especially hyperphosphatemia and hypercalcemia) are incriminated but inflammation, hormonal perturbation, metabolic disorders, and oxidative stress can also lead to VC. Oxidative stress in VSMCs, generated by hyperlipidemia and oxidized lipoproteins or uremic milieu [166], causes the expression of runx2[168], osterix and govers Wnt signaling [236], leading to osteogenic differentiation. Inflammatory cytokines, such as TNF-α, can also induce calcification via Msx2/Wnt/β-catenin pathway [237]. In support of that, calcium deposits colocalize with inflammatory cells in vitro [161] and in vivo [160]. Moreover, it has been suggested that mineral crystals may themselves be pro-inflammatory, creating a vicious cycle of inflammation and calcification [220,238]. The receptor for advanced glycation end products (RAGE) endogenously expressed in endothelial cells and its ligands (in which S100 family proteins are found), are also known to be involved in atherosclerotic formation and VC [42]. It has been suggested that galectin-3 and RAGE modulate vascular osteogenesis in part via Wnt/β-catenin signaling [239]. Several trials have shown a raise in serum levels of S100/calgranulins in vascular disease [240,241]. Thereby, S100 proteins could be a potential biomarker and therapeutic target to develop [242].

Involved in the control of both parathyroid hormone (PTH) and calcitonin secretion, the calcium-sensing receptor (CaSR) is a G protein–coupled cell surface receptor that can sense extracellular calcium ions. Evidence have been provided to demonstrate that a decrease in the CaSR protein expression in the vasculature is directly involved in the development of VC [243,244]. It is of particular interest to note that calcimimetics, which are allosteric drug compounds that selectively target the CaSR, decrease VC at least in part through local control of the CaSR expression in VSMC [245,246]. However, so far, the mechanism whereby the CaSR exert its protective effect remains largely unknown. Hormones have pleiotropic effects on calcific vasculopathy. For example, the adipose-derived factor, leptin, promotes VC in vitro [247] and in vivo [248]. Adiponectin-deficient mice have increased vascular calcification [249]. The influence of PTH is part of bone turnover process. A disruption between promoters and inhibitors can also generate VC. Moreover, similar to bone formation, there might a balance between VC and its resorption. Indeed, monocytes and macrophages contained in the calcified wall can differentiate into an osteoclast-like phenotype and counteracts the action of VSMCs that have undergone osteoblast differentiation [250]. Hyperphosphatemia would disadvantage osteoclast phenotype by down-regulating RANK ligand-induced signalling [251] but this is not clear whether osteoclast-like cells can really counteract VC or solely witness vascular remodelling process. All these modifications will favour for an optimal microenvironment for hydroxyapatite formation and calcification. Similar osteogenic differentiation is also observed, in vivo, in animal and human uremic models [98,103,186].

Clinical Assessment Methods of Vascular Calcification

There are several methods to assess the amount of arterial calcification: conventional radiography, dual-energy X-ray absorptiometry (DXA), multi-slice computer tomography (MSCT), electron beam computer tomography [EBCT], magnet resonance imaging [MRI], ultrasound, intravascular ultrasound (IVUS) and optical coherence tomography (OCT) [252]. Unfortunately, except for intravascular ultrasound, none of these techniques can distinguish intimal from medial calcification [140,182]. In clinical practice, there are also other established methods for measuring arterial stiffness (AS) with functional measurements and diverse imaging methods. However, the direct prediction of mVC is not easy for all these methods.

Functional (Hemodynamic) Measurements

A clinically easily applicable method to assess mVC is the measurement of the ankle-/brachial index (ABI) with a high ABI (> 1.3) serves as marker of VC [253]. Conversely, the estimation of local stiffness is an established only by direct measurement of parameters strongly linked to stiffness. The pulse wave velocity (PWV) is a robust and reliable parameter that is considered as gold standard for AS assessment. The determination of PWV, is a one of the simplest ways to estimate the level of AS, includes the measurements of the distance covered by the wave and the time required to cover the distance (PWV = distance/time delay). Basically, the regional PWV of each vessel in the body can be measured. However, aorta and its major branches represent the main sites of interest and has clinical relevance, because they contribute to the larger part of the arterial buffering function [254] and responsible for most of the pathophysiological sequels of increased stiffness. Carotid-femoral PWV is very suitable and easily applicable in the clinic, as it assesses the aortoiliac pathway. There are also established methods for the measurement of the PWV in the upper [brachial PWV] and lower limbs (femoro-tibial PWV) [255]. The carotid-femoral PWV has predictive value for cardiovascular events in several epidemiologic studies in different populations [256-258] while PWV measurements outside the aortic track demonstrated no correlation with cardiovascular events [259].

Another method used to assess local stiffness is the measurement of arterial distensibility using ultrasound or echotracking devices for the detection of diameter changes of the vessel during systole and diastole [260]. The method’s limitations are its dependence on high spatial resolution and the high degree of technical expertise required. Furthermore, only superficial arteries, such as the common carotid or femoral artery, are examined because its depth penetration is limited. The determination of pulse pressure (PP) and the augmentation index (Aix) is another method to assess AS. In case of stiff arteries, the reflected wave arrives earlier in the central arteries and augments the systolic pressure. As a result, the PP and AIx ((first systolic peak-second systolic peak)/ (pulse pressure increase)) [252]. The limitation of PP and AIx determination is their dependency on other conditions such as heart rate, ventricular contractility, duration and pattern of ventricular ejection, reflectance point and measured vessel segment. It has been elegantly shown by Scuteri, et al. [261] that PWV and therefore AS increase with age independently of blood pressure development. Furthermore, patients with ESRD on regular haemodialysis usually have higher PWV in comparison with healthy controls [262], and the PWV increase over time is higher than in the healthy population. Therefore, PWV seems to be a marker for timedependent ageing processes in the vascular system and less sufficient as specific indicator for AS [255,263].

Imaging Methods

Conventional radiography is a semi-qualitative method with several established scores. The abdominal aortic calcification score was proposed by Kauppila, et al. [264] to assess the extent of calcification of the abdominal aorta in front of the lumbar vertebrae on a lateral X-ray of the lumbar spine. However, conventional radiography of peripheral vessels may be a useful marker to measure Vamp [265]. Conventional radiography can indirectly indicate the presence of VCm by a ‘tram track’ calcification pattern in comparison with a ‘patchy’ pattern typically when atherosclerotic plaques are calcified [266,267]. Linear ‘tram-tracks’ are a typical appearance of VCm on conventional radiography [268], whereas a ‘patchy’ pattern typically suggesting atherosclerosis [266,267]. However, the sensitivity and specificity of conventional radiographs for detecting VCm or discrimination between VCm and intimal calcification remain uncertain [269,270]. Dual-energy X-ray absorptiometry is also a well-established method usually used for the measurement of bone mineral density but can also be used for simultaneous semiquantitative assessment of VC [271]. Both MSCT and EBCT are also very sensitive and precise imaging techniques for the detection and quantification of calcification [272]. MRI is, in general, a superior method for imaging soft tissue while it is not suitable for reliable assessment of VC, due to very short calcium echo time [273]. During endovascular interventions, IVUS [274] and OCT [275] can be also used to detect and quantify the amount of VC. Although OCT provides higher resolution than IVUS, its penetration depth is not sufficient to evaluate the entire medial layer. In the clinical setting, multidetector computed tomography is often used and generates a quantitative calcium score [276,277], which is a potent predictor for cardiovascular events [278]. Most studies identified intimal calcification as predictor of a vulnerable plaque phenotype, the punctated “spotty calcification” [277,279].

Circulating Biomarkers

A comprehensive approach including gathering of exact patient history and performing hemodynamic measurements and imaging studies is needed to determine the presence of arteriosclerosis, to quantify its amount and to provide discrimination from atherosclerosis. Currently, the latter issue is not still difficult as hemodynamic measurements nor imaging studies can certainly exactly distinguish between intimal and medial calcification. Therefore, new specific probes imaging microcalcification can provide a platform to study the earliest events associated with VC at the molecular and cellular level. The use of circulating biomarkers such as MGP for detecting or screening VC is an attractive possibility. Vitamin K-dependent proteins have been associated with the earliest calcification areas in the plaque [280]. It was the uncarboxylated form of MGP that strongly correlated with both medial and intimal calcification [280,281]. By measuring circulating MGP isoforms it was shown that most of the healthy population have sub-optimal levels of vascular vitamin K [282,283]. Preliminary data confirmed MGP are associated with aspects of cardiovascular disease as patients with high VC scores display high levels of inactive MGP, especially dialysis patients [284-286].

Conclusion

Vascular calcification is recognized as an active cellular process that occurs in response to metabolic insults that is intimately entwined with aging, abnormal mineral metabolism, and other related chronic diseases (i.e., DM, CKD). Within vascular microenvironment itself, a dense and interconnected network of calcification inhibitors and promoters were highlighted. Under normal conditions, there is a balance between all these parameters. Currently, vascular calcification is regulated by a complex pathophysiological mechanism, primarily triggered when there is an imbalance between inhibitors and promoters, in favour of osteogenic proteins and transcription factors synthesis, in detriment of bone reabsorption mediators. According to active theory, VSMCs undergo differentiation into osteoblast-like cells, in great part because of an increased intracellular phosphate concentration that is likely mediated by the co-transporter Pit-1 in response to extracellular hyperphosphatemia. Other risk factors such as advanced age, smoking, inflammation, oxidative stress, mineral and bone disorders (MBD) are also known to be associated to VSMCs conversion. As evidenced by different clinical observations, animal models, and molecular studies, the exact molecular and cellular mechanism of vascular calcification is still far from being fully elucidated. Thereby, the challenge remains to understand which mechanisms are active and/or predominate under various disease states, and to develop effective therapeutic strategies that may prevent and potentially reverse vascular calcification. Correspondingly, qualities that would be appreciated for selecting a good vascular calcification biomarker depend on its capacities to achieve clinical goals, particularly its ability to select high risk patients for further investigation, to make a reliable calcification assessment, to provide a prognostic, to help in treatment choice or to follow up the treatment efficiency. However, functional characteristics and imaging methods are commonly used for the diagnosis of the calcified arterial injury.

Declarations

Ethics Approval and Consent to Participate

Not applicable.

Consent For Publication

Not applicable.

Availability of Data and Material

The datasets used and analyzed during the current study are available from the corresponding author on reasonable request.

Competing Interests

The author declares that there is no conflict of interests regarding the publication of this paper.

Funding

Nil support in financial or other manner

Authors’ Contributions

LM had participated in the design of the study, data analyses, and manuscript preparation; and the authors could have read and approved the final manuscript.

Acknowledgements

The Author is grateful to College of Health Sciences Research and Community Office of Arsi University as well as researchers who their documents were used in the preparation of the review.

References

  1. Kapustin A, Davies J, Reynolds J, McNair R, Jones G, et al. (2011) Calcium regulates key components of vascular smooth muscle cell-derived matrix vesicles to enhance mineralization. Circ Res 109(1): e1-12.
  2. Chen N, Moe S (2012) Vascular Calcification: Pathophysiology and Risk Factors. Curr Hypertens Rep 14(3): 228-237.
  3. Heiss A, Eckert T, Aretz A, Richtering W, van Dorp W, et al. (2008) Hierarchical role of fetuin-A and acidic serum proteins in the formation and stabilization of calcium phosphate particles. J Biol Chem 283: 14815-14825.
  4. Karwowski W, Naumnik B, Szczepanski M, Mysliwiec M (2012) The mechanism of vascular calcification - A systematic review. Med Sci Monit 18: RA1-11.
  5. Kaneto H, Katakami N, Matsuhisa M, Matsuoka T (2010) Role of reactive oxygen species in the progression of type 2 diabetes and atherosclerosis. Mediat Inflamm 2010: 453892.
  6. Shanahan C, Crouthamel M, Kapustin A, Giachelli C (2011) Arterial calcification in chronic kidney disease: key roles for calcium and phosphate. Circ Res 109: 697-711.
  7. Schurgers L (2013) Vitamin K: key vitamin in controlling vascular calcification in chronic kidney disease. Kidney International 83: 782-784.
  8. Proudfoot D, Shanahan C (2001) Biology of calcification in vascular cells: Intima versus media. Herz 26: 245-251.
  9. Ross R (1993) The pathogenesis of atherosclerosis: a perspective for the 1990s. Nature 362: 801-809.
  10. Agatston A, Janowitz W, Hildner F, Zusmer N, Viamonte M, et al. (1990) Quantification of coronary artery calcium using ultrafast computed tomography. J Am Coll Cardiol 15: 827-832.
  11. O Rourke R, Brundage B, Froelicher V, Greenland P, Grundy S, et al. (2000) American College of Cardiology/American Heart Association Expert Consensus Document on electron-beam computed tomography for the diagnosis and prognosis of coronary artery disease. J Am Coll Cardiol 36: 126-140.
  12. Simon A, Levenson J (1993) Early detection of subclinical atherosclerosis in asymptomatic subjects at high risk for cardiovascular disease. Clin Exp Hypertens 15: 1069-1076.
  13. Wexler L, Brundage B, Crouse J, Detrano R, Fuster V, et al. (1996) Coronary artery calcification: pathophysiology, epidemiology, imaging methods, and clinical implications. A statement for health professionals from the American Heart Association Writing Group. Circulation 94: 1175-1192.
  14. Gross M, Meyer H, Ziebart H, Rieger P, Wenzel U, et al. (2007) Calcification of coronary intima and media: immunohistochemistry, backscatter imaging, and x-ray analysis in renal and nonrenal patients. Clin J Am Soc Nephrol 2(1): 121-134.
  15. Nakamura S, Ishibashi Ueda H, Niizuma S, Yoshihara F, Horio T, et al. (2009) Coronary calcification in patients with chronic kidney disease and coronaryartery disease. Clin J Am Soc Nephrol 4(12): 1892-1900.
  16. Moe S, Chen N (2004) Pathophysiology of vascular calcification in chronic kidney disease. Circ Res 95(6): 560-567.
  17. Proudfoot D, Shanahan C, Weissberg P (1998) Vascular calcification: new insights into an old problem [editorial; comment]. Journal of Pathology 185(1): 1-3.
  18. Saxena A, Waddell I, Friesen R, Michalski R (2005) Monckeberg medial calcific sclerosis mimicking malignant calcification pattern at mammography. J Clin Pathol 58: 447-448.
  19. Castillo B, Torczynski E, Edward D (1999) Monckeberg’s sclerosis in temporal artery biopsy specimens. Br J Ophthalmol 83: 1091-1092.
  20. Stary H, Chandler A, Glagov S, Guyton J, Insull W, et al. (1994) A definition of initial, fatty streak, and intermediate lesions of atherosclerosis. Arterioscler Thromb 14: 840-856.
  21. Stary H, Chandler A, Dinsmore R, Fuster V, Glagov S, et al. (1995) A definition of advanced types of atherosclerotic lesions and a histological classification of atherosclerosis: a report from the Committee on Vascular Lesions of the Council on Arteriosclerosis, American Heart Association. Circulation 92: 1355-1374.
  22. Stary H (2000) Natural history of calcium deposits in atherosclerosis progression and regression. Z Kardiol 89(Suppl 2): 28-35.
  23. Lanzer P, Boehm M, Sorribas V, Thiriet M, Janzen J, et al. (2014) Medial vascular calcification revisited: Review and perspectives. European Heart Journal 35: 1515-1525.
  24. Janzen J, Vuong P (2001) Arterial calcifications: morphological aspects and their pathological implications. Z Kardiol 90(Suppl. 3): 6-11.
  25. Janzen J, Bultmann B, Leitritz M, Rothenberger Janzen K, Vuong P (2003) Histopathological aspects of arterial calcifications. Perfusion 16: 136-140.
  26. Lehto S, Niskanen L, Suhonen M, Ronnemaa T, Laakso M (1996) Medial artery calcification. A neglected harbinger of cardiovascular complications in non-insulin-dependent diabetes mellitus. Arterioscler Thromb Vasc Biol 16(8): 978-983.
  27. Moe S, Chen N (2008) Mechanisms of vascular calcification in chronic kidney disease. J Am Soc Nephrol 19(2): 213-216.
  28. Moe S, Chen N (2003) Calciphylaxis and vascular calcification: a continuum of extra-skeletal osteogenesis. Pediatr Nephrol 18: 969-975.
  29. Budoff M, Shaw L, Liu S, Weinstein S, Mosler T, et al. (2007) Long-term prognosis associated with coronary calcification: observations from a registry of 25,253 patients. J Am Coll Cardiol 49: 1860-1870.
  30. Rennenberg R, Kessels A, Schurgers L, van Engelshoven J, de Leeuw P, et al. (2009) Vascular calcifications as a marker of increased cardiovascular risk: a meta-analysis. Vasc Health Risk Manag 5: 185-197.
  31. Gorriz J, Molina P, Cerveron M, Vila R, Bover J, et al. (2015) Vascular calcification in patients with nondialysis CKD over 3 years. Clin J Am Soc Nephrol 10: 654-666.
  32. Drueke T, Massy Z (2010) Atherosclerosis in CKD: differences from the general population. Nat Rev Nephrol 6: 723-735.
  33. Wolisi G, Moe S (2005) The role of vitamin D in vascular calcification in chronic kidney disease. Semin Dial 18(4): 307-314.
  34. Aronow W, Schwartz K, Koenigsberg M (1987) Correlation of serum lipids, calcium, and phosphorus, diabetes mellitus and history of systemic hypertension with presence or absence of calcified or thickened aortic cusps or root in elderly patients. Am J Cardiol 59(9): 998-999.
  35. Wilson P, Kauppila L, O’Donnell C, Kiel D, Hannan M, et al. (2001) Abdominal aortic calcific deposits are an important predictor of vascular morbidity and mortality. Circulation 103(11): 1529-1534.
  36. Wayhs R, Zelinger A, Raggi P (2002) High coronary artery calcium scores pose an extremely elevated risk for hard events. J Am Coll Cardiol 39(2): 225-230.
  37. Liberman M, Bassi E, Martinatti M, Lario F, Wosniak J, et al. (2008) Oxidant generation predominates around calcifying foci and enhances progression of aortic valve calcification. Arterioscler Thromb Vasc Biol 28(3): 463-470.
  38. Otto C, Lind B, Kitzman D, Gersh B, Siscovick D (1999) Association of aorticvalve sclerosis with cardiovascular mortality and morbidity in the elderly. N Engl J Med 341(3): 142-147.
  39. Jian B, Jones P, Li Q, Mohler E, Schoen F, et al. (2001) Matrix metalloproteinase-2 is associated with tenascin-C in calcific aortic stenosis. Am J Pathol 159(1): 321-327.
  40. Cowell S, Newby D, Prescott R, Bloomfield P, Reid J, et al. (2005) A randomized trial of intensive lipid-lowering therapy in calcific aortic stenosis. N Engl J Med 352(23): 2389-2397.
  41. Valdivielso J (2011) Vascular calcification: types and mechanisms. Nefrologia 31(2): 142-147.
  42. Evrard S, Delanaye P, Kamel S, Cristol J, Cavalier E (2015) Vascular calcification: from pathophysiology to biomarkers. Clinica Chimica Acta 438: 401-414.
  43. Prie D, Torres P, Friedlander G (2009) A new axis of phosphate balance control: fibroblast growth factor 23-Klotho. Nephrol Ther 5: 513-519.
  44. Hu P, Xuan Q, Hu B, Lu L, Wang J, et al. (2012) Fibroblast growth factor-23 helps explain the biphasic cardiovascular effects of vitamin D in chronic kidney disease. Int J Biol Sci 8: 663-671.
  45. Toussaint N, Pedagogos E, Tan S (2012) Phosphate in early chronic kidney disease: associations with clinical outcomes and a target to reduce cardiovascular risk. Nephrology 17: 433-444.
  46. Parker B, Schurgers L, Brandenburg V (2010) The associations of fibroblast growth factor 23 and uncarboxylated matrix Gla protein with mortality in coronary artery disease: The Heart and Soul Study. Ann Intern Med 152: 640-648.
  47. Massy Z, Drueke T (2013) Vascular calcification. Curr Opin Nephrol Hypertens 22: 405-412.
  48. Lau W, Leaf E, Hu M (2012) Vitamin D receptor agonists increase klotho and osteopontin while decreasing aortic calcification in mice with chronic kidney disease fed a high phosphate diet. Kidney Int 82: 1261-1270.
  49. Ketteler M, Bongartz P, Westenfeld R (2003) Association of low fetuin-A (AHSG) concentrations in serum with cardiovascular mortality in patients on dialysis: a cross-sectional study. Lancet 361: 827-833.
  50. Oikawa O, Higuchi T, Yamazaki T, Yamamoto C, Fukuda N, et al. (2007) Evaluation of serum fetuin-A relationships with biochemical parameters in patients on hemodialysis. Clin Exp Nephrol 11: 304-308.
  51. Heiss A, DuChesne A, Denecke B, Joachim Grötzinger, Kazuhiko Yamamoto, et al. (2003) Structural basis of calcification inhibition by alpha 2-HS glycoprotein/fetuin-A Formation of colloidal calciprotein particles. J Biol Chem 278(15): 13333-13341.
  52. Schafer C, Heiss A, Schwarz A, Jurgen Floege, Werner Muller Esterl, et al. (2003) The serum protein alpha 2-Heremans-Schmid glycoprotein/fetuin-A is a systemically acting inhibitor of ectopic calcification. J Clin Invest 112(3): 357-366.
  53. Price P, Lim J (2003) The inhibition of calcium phosphate precipitation by fetuin is accompanied by the formation of a fetuin-mineral complex. J Biol Chem 278(24): 22144-22152.
  54. Smith E, Hanssen E, McMahon L, Holt S (2013) Fetuin-A-containing calciprotein particles reduce mineral stress in the macrophage. PLoS One.
  55. Lee C, Chua S, Hsu C, Terry Ting Yu Chiu, Yueh Ting Lee, et al. (2013) Biomarkers associated with vascular and valvular calcification in chronic hemodialysis patients. Dis Markers 34(4): 229-235.
  56. Chen H, Chiu Y, Hsu S, Pai M, Yang J, et al. (2013) Low serum fetuin A levels and incident stroke in patients with maintenance haemodialysis. Eur J Clin Invest 43(4): 387-396.
  57. Abdel Wahab A, Fathy O, Al Harizy R (2013) Negative correlation between fetuin-A and indices of vascular disease in systemic lupus erythematosus patients with and without lupus nephritis. Arab J Nephrol Transplant 6(1): 11-20.
  58. Marechal C, Schlieper G, Nguyen P, Thilo Krüger, Emmanuel Coche (2011) Serum fetuin-A levels are associated with vascular calcifications and predict cardiovascular events in renal transplant recipients. Clin J Am Soc Nephrol 6(5): 974-985.
  59. Giachelli C, Steitz S (2000) Osteopontin: A versatile regulator of inflammation and biomineralization. Matrix Biol 19(7): 615-622.
  60. Scatena M, Liaw L, Giachelli C (2007) Osteopontin: amultifunctionalmolecule regulating chronic inflammation and vascular disease. Arterioscler Thromb Vasc Biol 27(11): 2302-2309.
  61. Speer M, McKee M, Guldberg R, Hsueh Ying Yang, Elyse Tung (2002) Inactivation of the osteopontin gene enhances vascular calcification of matrix Gla protein-deficient mice: evidence for osteopontin as an inducible inhibitor of vascular calcification in vivo. J Exp Med 196(8): 1047-1055.
  62. Jono S, Peinado C, Giachelli C (2000) Phosphorylation of osteopontin is required for inhibition of vascular smooth muscle cell calcification. J Biol Chem 275(26): 20197-20203.
  63. Wada T, McKee M, Steitz S, Giachelli C (1999) Calcification of vascular smooth muscle cell cultures: inhibition by osteopontin. Circ Res 84(2): 166-178.
  64. Qin X, Corriere M, Matrisian L, Guzman R (2006) Matrix metalloproteinase inhibition attenuates aortic calcification. Arterioscler Thromb Vasc Biol 26(7): 1510-1516.
  65. Berezin A, Kremzer A (2013) Circulating osteopontin as a marker of early coronary vascular calcification in type two diabetesmellitus patients with known asymptomatic coronary artery disease. Atherosclerosis 229(2): 475-481.
  66. Tousoulis D, Siasos G, Maniatis K, Evangelos Oikonomou, Stamatios Kioufis, et al. (2013) Serum osteoprotegerin and osteopontin levels are associated with arterial stiffness and the presence and severity of coronary artery disease. Int J Cardiol 167(5): 1924-1928.
  67. Albu A, Fodor D, Bondor C, Craciun A (2013) Bone metabolism regulators and arterial stiffness in postmenopausal women. Maturitas 76(2): 146-150.
  68. Kiefer F, Zeyda M, Gollinger K, Lukas Kenner, Thomas M Stulnig, et al. (2010) Neutralization of osteopontin inhibits obesity-induced inflammation and insulin resistance. Diabetes 59(4): 935-946.
  69. Zheng Y, Wang Z, Deng L, Michael P Gantier, Jun Ping Liu, et al. (2012) Osteopontin promotes inflammation in patients with acute coronary syndrome through its activity on IL-17 producing cells. Eur J Immunol 42(10): 2803-2814.
  70. Sun J, Xu Y, Dai Z, Sun Y (2009) Intermittent high glucose enhances proliferation of vascular smooth muscle cells by upregulating osteopontin. Mol Cell Endocrinol 313(1-2): 64-69.
  71. Simonet W, Lacey D, Dunstan C, W Pattison, P Campbell (1997) Osteoprotegerin: A novel secreted protein involved in the regulation of bone density. Cell 89(2): 309-319.
  72. Collin Osdoby P (2004) Regulation of vascular calcification by osteoclast regulatory factors RANKL and osteoprotegerin. Circ Res 95(11): 1046-1057.
  73. Lacey D, Timms E, Tan H, T Burgess, R Elliott, et al. (1998) Osteoprotegerin ligand is a cytokine that regulates osteoclast differentiation and activation. Cell 93(2): 165-176.
  74. Bennett B, Scatena M, Kirk E, Cecilia M Giachelli, Michael E Rosenfeld, et al. (2006) Osteoprotegerin inactivation accelerates advanced atherosclerotic lesion progression and calcification in older ApoE-/- mice. Arterioscler Thromb Vasc Biol 26(9): 2117-2124.
  75. Collin Osdoby P, Rothe L, Bekker S, Anderson F, Huang Y, et al. (2002) Basic fibroblast growth factor stimulates osteoclast recruitment, development, and bone pit resorption in association with angiogenesis in vivo on the chick chorioallantoic membrane and activates isolated avian osteoclast resorption in vitro. J Bone Miner Res 17(10): 1859-1871.
  76. Bucay N, Sarosi I, Dunstan C, David L Lacey, William J Boyle, et al. (1998) Osteoprotegerin-deficient mice develop early onset osteoporosis and arterial calcification. Genes Dev 12(9): 1260-1268.
  77. Morony S, Sage A, Corbin T, Lu J, Tintut Y, et al. (2012) Enhanced mineralization potential of vascular cells from SM22alpha-Rankl (tg) mice. Calcif Tissue Int 91(6): 379-386.
  78. Morena M, Jaussent I, Halkovich A, Anne Marie Dupuy, Anne Sophie Bargnoux, et al. (2012) Bone biomarkers help grading severity of coronary calcifications in non-dialysis chronic kidney disease patients. PLoS One.
  79. Van de Loo P, Soute B, van Haarlem L, Vermeer C (1987) The effect of Gla-containing proteins on the precipitation of insoluble salts. Biochem Biophys Res Commun 142(1): 113-119.
  80. Levy R, Gundberg C, Scheinman R (1983) The identification of the vitamin K-dependent bone protein osteocalcin as one of the gamma-carboxyglutamic acid containing proteins present in calcified atherosclerotic plaque and mineralized heart valves. Atherosclerosis 46(1): 49-56.
  81. Hunter G, Hauschka P, Poole A, Rosenberg L, Goldberg H, et al. (1996) Nucleation and inhibition of hydroxyapatite formation by mineralized tissue proteins. Biochem J 317(1): 59-64.
  82. Ducy P, Desbois C, Boyce B, C Dunstan, E Smith, et al. (1996) Increased bone formation in osteocalcin-deficient mice. Nature 382(6590): 448-452.
  83. Aoki A, Murata M, Asano T, Aki Ikoma, Masami Sasaki, et al. (2013) Association of serum osteoprotegerin with vascular calcification in patients with type 2 diabetes. Cardiovasc Diabetol 12: 11.
  84. Kim K, Kim K, Park K, Yumie Rhee, Yong Ho Lee, et al. (2012) Aortic calcification and bone metabolism: the relationship between aortic calcification, BMD, vertebral fracture, 25-hydroxyvitamin D, and osteocalcin. Calcif Tissue Int 91(6): 370-378.
  85. Harmey D, Hessle L, Narisawa S, Johnson K, Terkeltaub R, et al. (2004) Concerted regulation of inorganic pyrophosphate and osteopontin by akp2, enpp1, and ank: an integrated model of the pathogenesis of mineralization disorders. Am J Pathol 164(4): 1199-1209.
  86. Rutsch F, Ruf N, Vaingankar S, Bernd Hinrichs, Wendy Smith, et al. (2003) Mutations in ENPP1 are associated with “idiopathic” infantile arterial calcification. Nat Genet 34(4): 379-381.
  87. Johnson K, Polewski M, van Etten D, Terkeltaub R (2005) Chondrogenesis mediated by PPi depletion promotes spontaneous aortic calcification in NPP1-/- mice. Arterioscler Thromb Vasc Biol 25(4): 686-691.
  88. Towler D (2005) Inorganic pyrophosphate: a paracrine regulator of vascular calcification and smooth muscle phenotype. Arterioscler Thromb Vasc Biol 25(4): 651-654.
  89. O Neill W, Sigrist M, McIntyre C (2010) Plasma pyrophosphate and vascular calcification in chronic kidney disease. Nephrol Dial Transplant 25(1): 187-191.
  90. Riser B, Barreto F, Rezg R, Paul W Valaitis, Chyung S Cook, et al. (2011) Daily peritoneal administration of sodium pyrophosphate in a dialysis solution prevents the development of vascular calcification in a mouse model of uraemia. Nephrol Dial Transplant 26(10): 3349-3357.
  91. O Neill W, Lomashvili K, Malluche H, Faugere M, Riser B, et al. (2011) Treatment with pyrophosphate inhibits uremic vascular calcification. Kidney Int 79(5): 512-517.
  92. Villa Bellosta R, Sorribas V (2011) Calcium phosphate deposition with normal phosphate concentration. Role of pyrophosphate. Circ J 75(11): 2705-2710.
  93. Lomashvili K, Monier Faugere M, Wang X, Malluche H, O Neill W (2009) Effect of bisphosphonates on vascular calcification and bone metabolism in experimental renal failure. Kidney Int 75(6): 617-625.
  94. Zhou S, Fang X, Xin H, Guan S (2013) Effects of alendronate on the Notch1RBPJkappa signaling pathway in the osteogenic differentiation and mineralization of vascular smooth muscle cells. Mol Med Rep 8(1): 89-94.
  95. Villa Bellosta R, Sorribas V (2013) Prevention of vascular calcification by polyphosphates and nucleotides- role of ATP. Circ J 77(8): 2145-2151.
  96. Khavandgar Z, Roman H, Li J (2013) Elastin haploinsufficiency impedes the progression of arterial calcification in MGP-deficient mice. J Bone Miner Res.
  97. Luo G, Ducy P, McKee, GJ Pinero, E Loyer, et al. (1997) M Spontaneous calcification of arteries and cartilage in mice lacking matrix GLA protein. Nature 386(6620): 78-81.
  98. Moe S, O Neill K, Duan D, Ahmed S, Chen N, et al. (2002) Medial artery calcification in ESRD patients is associated with deposition of bone matrix proteins. Kidney Int 61(2): 638-647.
  99. Moe S, Reslerova M, Ketteler M, Kalisha O neill, Danxia Duan, et al. (2005) Role of calcification inhibitors in the pathogenesis of vascular calcification in chronic kidney disease (CKD). Kidney Int 67(6): 2295-22304.
  100. Mizobuchi M, Towler D, Slatopolsky E (2009) Vascular calcification: the killer of patients with chronic kidney disease. J Am Soc Nephrol 20(7): 1453-1464.
  101. Roy M, Nishimoto S (2002) Matrix Gla protein binding to hydroxyapatite is dependent on the ionic environment: calcium enhances binding affinity, but phosphate and magnesium decrease affinity. Bone 31(2): 296-302.
  102. Sweatt A, Sane D, Hutson S, Wallin R (2003) Matrix Gla protein (MGP) and bone morphogenetic protein-2 in aortic calcified lesions of aging rats. J Thromb Haemost 1(1): 178-185.
  103. Moe S, Duan D, Doehle B, O Neill K, Chen N, et al. (2003) Uremia induces the osteoblast differentiation factor Cbfa1 in human blood vessels. Kidney Int 63(3): 1003-1011.
  104. Tintut Y, Patel J, Parhami F, Demer L (2000) Tumor necrosis factor-alpha promotes in vitro calcification of vascular cells via the cAMP pathway. Circulation 102(21): 2636-2642.
  105. Stenvinkel P, Ketteler M, Johnson R (2005) IL-10, IL-6, and TNF-alpha: central factors in the altered cytokine network of uremia-the good, the bad, and the ugly. Kidney Int 67(4): 1216-1233.
  106. Watson K, Parhami F, Shin V, Demer L (1998) Fibronectin and collagen I matrixes promote calcification of vascular cells in vitro, whereas collagen IV matrix is inhibitory. Arterioscler Thromb Vasc Biol 18(12): 1964-1971.
  107. Watson K, Bostrom K, Ravindranath R (1994) TGF-beta 1 and 25- hydroxycholesterol stimulate osteoblast-like vascular cells to calcify. J Clin Invest 93(5): 2106-2113.
  108. Shioi A, Katagi M, Okuno Y (2002) Induction of bone-type alkaline phosphatase in human vascular smooth muscle cells: roles of tumor necrosis factor-alpha and oncostatin M derived from macrophages. Circ Res 91(1): 9-16.
  109. Jono S, Nishizawa Y, Shioi A, Morii H (1998) 1, 25-dihydroxyvitamin D3 increases in vitro vascular calcification by modulating secretion of endogenous parathyroid hormone-related peptide. Circulation 98(13): 1302-1306.
  110. Ducy P, Zhang R, Geoffroy V (1997) Osf2/Cbfa1: a transcriptional activator of osteoblast differentiation. Cell 89(5): 747-754.
  111. Tyson K, Reynolds J, McNair R (2003) Osteo/chondrocytic transcription factors and their target genes exhibit distinct patterns of expression in human arterial calcification. Arterioscler Thromb Vasc Biol 23(3): 489-494.
  112. Li X, Yang H, Giachelli C (2008) BMP-2 promotes phosphate uptake, phenotypic modulation, and calcification of human vascular smooth muscle cells. Atherosclerosis 199(2): 271-277.
  113. Abe E, Yamlamoto M, Taguchi Y (2000) Essential requirement of BMPs-2/4 for both osteoblast and osteoclast formation in murine bone marrow cultures from adult mice: antagonism by noggin. J Bone Miner Res 15(4): 663-673.
  114. Yao Y, Bennett B, Wang X (2010) Inhibition of bone morphogenetic proteins protects against atherosclerosis and vascular calcification. Circ Res 107(4): 485-494.
  115. Chen D, Zhao M, Mundy G (2004) Bone morphogenetic proteins. Growth Factors 22(4): 233-241.
  116. Wang E, Rosen V, D Alessandro J (1990) Recombinant human bone morphogenetic protein induces bone formation. Proc Natl Acad Sci USA 87(6): 2220-2224.
  117. Balemans W, Ebeling M, Patel N (2001) Increased bone density in sclerosteosis is due to the deficiency of a novel secreted protein (SOST). HumMol Genet 10(5): 537-543.
  118. Brunkow M, Gardner J, Van Ness J (2001) Bone dysplasia sclerosteosis results from loss of the SOST gene product, A novel cystine knot-containing protein. Am J Hum Genet 68(3): 577-589.
  119. Joiner D, Ke J, Zhong Z, Xu H, Williams B, et al. (2013) LRP5 and LRP6 in development and disease. Trends Endocrinol Metab 24(1): 31-39.
  120. Krishnan V, Bryant H, Macdougald O (2006) Regulation of bone mass by Wnt signaling. J Clin Invest 116(5): 1202-1209.
  121. Robling A, Niziolek P, Baldridge L (2008) Mechanical stimulation of bone in vivo reduces osteocyte expression of Sost/sclerostin. J Biol Chem 283(9): 5866-5875.
  122. Koos R, Brandenburg V, Mahnken A (2013) Sclerostin as a potential novel biomarker for aortic valve calcification: an in-vivo and ex-vivo J Heart Valve Dis 22(3): 317-325.
  123. Hampson G, Edwards S, Conroy S, Blake G, Fogelman I, et al. (2013) The relationship between inhibitors of the Wnt signalling pathway (Dickkopf-1(DKK1) and sclerostin), bone mineral density, vascular calcification, and arterial stiffness in post-menopausal women. Bone 56(1): 42-47.
  124. Szulc P, Bertholon C, Borel O, Marchand F, Chapurlat R, et al. (2013) Lower fracture risk in older men with higher sclerostin concentration: A prospective analysis from the MINOS study. J Bone Miner Res 28(4): 855-864.
  125. Morales Santana S, Garcia Fontana B, Garcia Martin A (2013) Atherosclerotic disease in type 2 diabetes is associated with an increase in sclerostin levels. Diabetes Care 36(6): 1667-1674.
  126. Kong Y, Boyle W, Penninger J (2000) Osteoprotegerin ligand: a regulator of immune responses and bone physiology. Immunol Today 21(10): 495-502.
  127. Kong Y, Feige U, Sarosi I (1999) Activated T cells regulate bone loss and joint destruction in adjuvant arthritis through osteoprotegerin ligand. Nature 402(6759): 304-309.
  128. Anderson D, Maraskovsky E, Billingsley W (1997) A homologue of the TNF receptor and its ligand enhance T-cell growth and dendritic-cell function. Nature 390(6656): 175-179.
  129. Myers D, Collier F, Minkin C (1999) Expression of functional RANK on mature rat and human osteoclasts. FEBS Lett 463(3): 295-300.
  130. Green E, Flavell R (1999) TRANCE-RANK, a new signal pathway involved in lymphocyte development and T cell activation. J Exp Med 189(7): 1017-1020.
  131. Kanegae Y, Tavares A, Izpisua Belmonte J, Verma I (1998) Role of Rel/NF-kappa B transcription factors during the outgrowth of the vertebrate limb. Nature 392(6676): 611-614.
  132. Yasuda H, Shima N, Nakagawa N (1998) Osteoclast differentiation factor is a ligand for osteoprotegerin/osteoclastogenesis-inhibitory factor and is identical to TRANCE/RANKL. Proc Natl Acad Sci USA 95(7): 3597-3602.
  133. Morena M, Terrier N, Jaussent I (2006) Plasma osteoprotegerin is associated with mortality in hemodialysis patients. J Am Soc Nephrol 17(1): 262-270.
  134. Hwang J, Wei J, Abbara S, Grinspoon S, Lo J, et al. (2012) Receptor activator of nuclear factorkappaB ligand (RANKL) and its relationship to coronary atherosclerosis in HIV patients. J Acquir Immune Defic Syndr 61(3): 359-363.
  135. Ozkok A, Caliskan Y, Sakaci T (2012) Osteoprotegerin/RANKL axis and progression of coronary artery calcification in hemodialysis patients. Clin J Am Soc Nephrol 7(6): 965-973.
  136. Meneghini M, Regalia A, Alfieri C (2013) Calcium and osteoprotegerin levels predict the progression of the abdominal aortic calcifications after kidney transplantation. Transplantation 96(1): 42-48.
  137. Gordin D, Soro Paavonen A, Thomas M (2013) Osteoprotegerin is an independent predictor of vascular events in Finnish adults with type 1 diabetes. Diabetes Care 36(7): 1827-1833.
  138. Winther S, Christensen J, Flyvbjerg A (2013) Osteoprotegerin and mortality in hemodialysis patients with cardiovascular disease. Clin Nephrol 80(3): 161-167.
  139. Lewis R (2012) Mineral and bone disorders in chronic kidney disease: new insights into mechanism and management. Ann Clin Biochem 49(5): 432-440.
  140. Raggi P, Gongora M, Gopal A, Callister T, Budoff M, et al. (2008) Coronary artery calcium to predict all-cause mortality in elderly men and women. J Am Coll Cardiol 52(1): 17-23.
  141. Mehrotra R, Budoff M, Hokanson J, Ipp E, Takasu J, et al. (2005) Progression of coronary artery calcification in diabetics with and without chronic kidney disease. Kidney Int 68(3): 1258-1266.
  142. Block G, Spiegel D, Ehrlich J, Mehta R, Lindbergh J, et al. (2005) Effects of sevelamer and calcium on coronary artery calcification in patients new to hemodialysis. Kidney Int 68(4): 1815-1824.
  143. Moe S, O Neill K, Reslerova M, Fineberg N, Persohn S, et al. (2004) Natural history of vascular calcification in dialysis and transplant patients. Nephrol Dial Transplant 19(9): 2387-2393.
  144. Schwarz U, Buzello M, Ritz E, Stein G, Raabe G, et al. (2000) Morphology of coronary atherosclerotic lesions in patients with end-stage renal failure. Nephrology, Dialysis, Transplantation 15(2): 218-223.
  145. Allison M, Hsi S, Wassel C, Morgan C, Ix J, et al. (2012) Calcified atherosclerosis in different vascular beds and the risk of mortality. Arterioscler Thromb Vasc Biol 32(1): 140-146.
  146. Savoia C, Burger D, Nishigaki N, Montezano A, Touyz R, et al. (2011) Angiotensin II and the vascular phenotype in hypertension. Expert Rev Mol Med 13: e11.
  147. Armstrong Z, Boughner D, Drangova M, Rogers K (2011) Angiotensin II type 1 receptor blocker inhibits arterial calcification in a pre-clinical model. Cardiovasc Res 90(1): 165-170.
  148. Tokumoto M, Mizobuchi M, Finch J, Nakamura H, Martin D, et al. (2009) Blockage of the renin-angiotensin system attenuates mortality but not vascular calcification in uremic rats: sevelamer carbonate prevents vascular calcification. Am J Nephrol 29(6): 582-591.
  149. Wu S, Yu Y, Cai Y, Jia L, Wang X, et al. (2012) Endogenous aldosterone is involved in vascular calcification in rat. Exp Biol Med (Maywood) 237(1): 31-37.
  150. Chen N, Duan D, O Neill K, Moe S (2006) High glucose increases the expression of Cbfa1 and BMP-2 and enhances the calcification of vascular smooth muscle cells. Nephrol Dial Transplant 21(12): 3435-3442.
  151. Al-Aly Z, Shao J, Lai C, Huang E, Cai J, et al. (2007) Aortic Msx2- Wnt calcification cascade is regulated by TNF-alpha-dependent signals in diabetic Ldlr−/− mice. Arterioscler Thromb Vasc Biol 27(12): 2589-2596.
  152. Bostrom K, Jumabay M, Matveyenko A, Nicholas S, Yao Y, et al. (2011) Activation of vascular bone morphogenetic protein signaling in diabetes mellitus. Circ Res 108(4): 446-457.
  153. Parhami F, Basseri B, Hwang J, Tintut Y, Demer L, et al. (2002) High-density lipoprotein regulates calcification of vascular cells. Circ Res 91(7): 570-576.
  154. Ting T, Miyazaki-Anzai S, Masuda M, Levi M, Demer L, et al. (2011) Increased lipogenesis and stearate accelerate vascular calcification in calcifying vascular cells. J Biol Chem 286(27): 23938-23949.
  155. Abedin M, Lim J, Tang T, Park D, Demer L, et al. (2006) N-3 fatty acids inhibit vascular calcification via the p38-mitogen-activated protein kinase and peroxisome proliferator-activated receptor-gamma pathways. Circ Res 98(6): 727-729.
  156. Parhami F, Morrow A, Balucan J, Leitinger N, Watson A, et al. (1997) Lipid oxidation products have opposite effects on calcifying vascular cell and bone cell differentiation. A possible explanation for the paradox of arterial calcification in osteoporotic patients. Arteriosclerosis, Thrombosis & Vascular Biology 17(4): 680-687.
  157. Ridker P (2003) Clinical application of C-reactive protein for cardiovascular disease detection and prevention. Circulation 107(3): 363-369.
  158. Oh J, Wunsch R, Turzer M, Bahner M, Raggi P, et al. (2002) Advanced coronary and carotid arteriopathy in young adults with childhood-onset chronic renal failure. Circulation 106(1): 100-105.
  159. Stompor T, Krasniak A, Sulowicz W, Dembinska-Kiec A, Janda K, et al. (2005) Changes in common carotid artery intima-media thickness over 1 year in patients on peritoneal dialysis. Nephrol Dial Transplant 20(2): 404-412.
  160. Aikawa E, Nahrendorf M, Figueiredo J, Swirski F, Shtatland T, et al. (2007) Osteogenesis associates with inflammation in early-stage atherosclerosis evaluated by molecular imaging in vivo 116(24): 2841-2850.
  161. Tintut Y, Patel J, Territo M, Saini T, Parhami F, et al. (2002) Monocyte/macrophage regulation of vascular calcification in vitro. Circulation 105(5): 650-655.
  162. Okazaki H, Shioi A, Hirowatari K, Koyama H, Fukumoto S, et al. Phosphatidylinositol 3-kinase/Akt pathway regulates inflammatory mediators-induced calcification of human vascular smooth muscle cells. Osaka City Med J 55(2): 71-80.
  163. Amore A, Coppo R (2002) Immunological basis of inflammation in dialysis. Nephrol Dial Transplant 17(Suppl 8): 16-24.
  164. Zoccali C, Mallamaci F, Tripepi G (2004) Novel cardiovascular risk factors in end-stage renal disease. J Am Soc Nephrol 15(Suppl 1): S77-S80.
  165. Yamada S, Taniguchi M, Tokumoto M, Toyonaga J, Fujisaki K, et al. (2011) The antioxidant tempol ameliorates arterial medial calcification in uremic rats: Important role of oxidative stress in the pathogenesis of vascular calcification in chronic kidney disease. J Bone Miner Res 27(2): 474-485.
  166. Sutra T, Morena M, Bargnoux A, Caporiccio B, Canaud B, et al. (2008) Superoxide production: A procalcifying cell signalling event in osteoblastic differentiation of vascular smooth muscle cells exposed to calcification media. Free Radic Res 42(9): 789-797.
  167. You H, Yang H, Zhu Q, Li M, Xue J, et al. (2009) Advanced oxidation protein products induce vascular calcification by promoting osteoblastic trans-differentiation of smooth muscle cells via oxidative stress and ERK pathway. Ren Fail 31(4): 313-319.
  168. Byon C, Javed A, Dai Q, Kappes J, Clemens T, et al. (2008) Oxidative stress induces vascular calcification through modulation of the osteogenic transcription factor Runx2 by AKT signaling. J Biol Chem 283(22): 15319-15327.
  169. Tintut Y, Parhami F, Tsingotjidou A, Tetradis S, Territo, et al. (2002) M 8-Isoprostaglandin E2 enhances receptor-activated NFkappa B ligand (RANKL)-dependent osteoclastic potential of marrow hematopoietic precursors via the cAMP pathway. J Biol Chem 277(16): 14221-14226.
  170. Tseng W, Lu J, Bishop G, Watson A, Sage A, et al. (2010) Regulation of interleukin-6 expression in osteoblasts by oxidized phospholipids. J Lipid Res 51(5): 1010-1016.
  171. Demer L (2002) Vascular calcification and osteoporosis: inflammatory responses to oxidized lipids. Int J Epidemiol 31(4): 737-741.
  172. Niwa T, Katsuzaki T, Miyazaki S, Miyazaki T, Ishizaki Y, et al. (1997) Immunohistochemical detection of imidazolone, a novel advanced glycation product, in kidneys and aortas of diabetic patients. J Clin Invest 99(6): 1272-1280.
  173. Miyata T, Sprague S (1996) Advanced glycation of beta 2-microglobulin in the pathogenesis of bone lesions in dialysis-associated amyloidosis. Nephrology, Dialysis, Transplantation 11(Suppl 3): 86-90.
  174. Sakata N, Noma A, Yamamoto Y, Okamoto K, Meng J, et al. (2003) Modification of elastin by pentosidine is associated with the calcification of aortic media in patients with end-stage renal disease. Nephrol Dial Transplant 18(8): 1601-1619.
  175. Yamagishi S, Fujimori H, Yonekura H, Tanaka N, Yamamoto H, et al. (1999) Advanced glycation endproducts accelerate calcification in microvascular pericytes. Biochem Biophys Res Commun 258(2): 353-357.
  176. Tanikawa T, Okada Y, Tanikawa R, Tanaka Y (2009) Advanced glycation end products induce calcification of vascular smooth muscle cells through RAGE/p38 MAPK. J Vasc Res 46(6): 572-580.
  177. Naka Y, Bucciarelli L, Wendt T, Lee L, Rong L, et al. (2004) RAGE axis: Animal models and novel insights into the vascular complications of diabetes. Arterioscler Thromb Vasc Biol 24(8): 1342-1349.
  178. Ren X, Shao H, Wei Q, Sun Z, Liu N, et al. (2009) Advanced glycation end-products enhance calcification in vascular smooth muscle cells. J Int Med Res 37(3): 847-854.
  179. Suga T, Iso T, Shimizu T, Tanaka T, Yamagishi S, et al. (2011) Activation of receptor for advanced glycation end products induces osteogenic differentiation of vascular smooth muscle cells. J Atheroscler Thromb 18(8): 670-683.
  180. Block G, Hulbert-Shearon T, Levin N, Port F (1998) Association of serum phosphorus and calcium × phosphate product with mortality risk in chronic hemodialysis patients: A national study. American Journal of Kidney Diseases 31(4): 607-617.
  181. Goodman W, Goldin J, Kuizon B, Yoon C, Gales B, et al. (2000) Coronary-artery calcification in young adults with end-stage renal disease who are undergoing dialysis. N Engl J Med 342: 1478-1483.
  182. Block G, Raggi P, Bellasi A, Kooienga L, Spiegel D, et al. (2007) Mortality effect of coronary calcification and phosphate binder choice in incident hemodialysis patients. Kidney Int 71: 238-239.
  183. Chertow G, Burke S, Raggi P (2002) Sevelamer attenuates the progression of coronary and aortic calcification in hemodialysis patients. Kidney Int 62(1): 245-252.
  184. Tonelli M, Sacks F, Pfeffer M, Gao Z, Curhan G, et al. (2005) Relation between serum phosphate level and cardiovascular event rate in people with coronary disease. Circulation 112(17): 2627-2633.
  185. Jono S, McKee M, Murry C, Shioi A, Nishizawa Y, et al. (2000) Phosphate regulation of vascular smooth muscle cell calcification. Circ Res 87: E10-17.
  186. Steitz S, Speer M, Curinga G, Yang H, Haynes P, et al. (2001) Smooth muscle cell phenotypic transition associated with calcification: upregulation of Cbfa1 and downregulation of smooth muscle lineage markers. Circ Res 89(12): 1147-1154.
  187. Chen N, O Neill K, Chen X, Moe S (2008) Annexin-mediated matrix vesicle calcification in vascular smooth muscle cells. J Bone Miner Res 23(11): 1798-1805.
  188. Werner A, Dehmelt L, Nalbant P (1998) Na+- dependent phosphate cotransporters: The NaPi protein families. Journal of Experimental Biology 201(Pt 23): 3135-3142.
  189. Li X, Yang H, Giachelli C (2006) Role of the sodium-dependent phosphate cotransporter, Pit-1, in vascular smooth muscle cell calcification. Circ Res 98: 905-912.
  190. Chen N, O Neill K, Duan D, Moe S (2002) Phosphorus and uremic serum up-regulate osteopontin expression in vascular smooth muscle cells. Kidney Int 62(5): 1724-1731.
  191. Chen N, Duan D, O Neill K, Wolisi G, Koczman J, et al. (2006) The mechanisms of uremic serum-induced expression of bone matrix proteins in bovine vascular smooth muscle cells. Kidney Int 70(6): 1046-1053.
  192. Hosaka N, Mizobuchi M, Ogata H, Kumata C, Kondo F, et al. (2009) Elastin degradation accelerates phosphate-induced mineralization of vascular smooth muscle cells. Calcif Tissue Int 85(6): 523-529.
  193. Simionescu A, Philips K, Vyavahare N (2005) Elastin-derived peptides and TGF-beta1 induce osteogenic responses in smooth muscle cells. Biochem Biophys Res Commun 334(2): 524-232.
  194. Lee K, Kim H, Li Q, Chi X, Ueta C, et al. (2000) Runx2 is a common target of transforming growth factor beta1 and bone morphogenetic protein 2, and cooperation between Runx2 and Smad5 induces osteoblast-specific gene expression in the pluripotent mesenchymal precursor cell line C2C12. Mol Cell Biol 20(23): 8783-8792.
  195. Bouvet C, Moreau S, Blanchette J, de Blois D, Moreau P, et al. (2008) Sequential activation of matrix metalloproteinase 9 and transforming growth factor beta in arterial elastocalcinosis. Arterioscler Thromb Vasc Biol 28(5): 856-862.
  196. Chen N, O Neill K, Chen K, Kraiwporn X, Gattone V, et al. (2011) Activation of Arterial Matrix Metalloproteinases Leads to Vascular Calcification in Chronic Kidney Disease. American Journal of Nephrology 34(3): 211-219.
  197. Moe S, Chertow G (2006) The Case against Calcium-Based Phosphate Binders. Clin J Am Soc Nephrol 1(4): 697-703.
  198. Yang H, Curinga G, Giachelli C (2004) Elevated extracellular calcium levels induce smooth muscle cell matrix mineralization in vitro. Kidney Int 66(6): 2293-2299.
  199. Reynolds J, Joannides A, Skepper J, McNair R, Schurgers L, et al. (2004) Human vascular smooth muscle cells undergo vesicle-mediated calcification in response to changes in extracellular calcium and phosphate concentrations: A potential mechanism for accelerated vascular calcification in ESRD. J Am Soc Nephrol 15(11): 2857-2867.
  200. Lomashvili K, Cobbs S, Hennigar R, Hardcastle K, O Neill W, et al. (2004) Phosphate-induced vascular calcification: role of pyrophosphate and osteopontin. J Am Soc Nephrol 15(6): 1392-1401.
  201. Shroff R, McNair R, Skepper J, Figg N, Schurgers L, et al. (2010) Chronic mineral dysregulation promotes vascular smooth muscle cell adaptation and extracellular matrix calcification. J Am Soc Nephrol 21(1): 103-112.
  202. Chen N, Kircelli F, ONeill K, Chen X, Moe S (2010) Verapamil inhibits calcification and matrix vesicle activity of bovine vascular smooth muscle cells. Kidney Int 77(5): 436-442.
  203. Wolf M (2009) Fibroblast growth factor 23 and the future of phosphorus management. Curr Opin Nephrol Hypertens 18(6): 463-468.
  204. Kuro oM, Matsumura Y, Aizawa H, Kawaguchi H, Suga T, et al. (1997) Mutation of the mouse klotho gene leads to a syndrome resembling ageing. Nature 390(6655): 45-51.
  205. Desjardins L, Liabeuf S, Renard C, Lenglet A, Lemke H, et al. (2012) FGF 23 is independently associated with vascular calcification but not bone mineral density in patients at various CKD stages. Osteoporos Int 23(7): 2017-2025.
  206. Balci M, Kirkpantur A, Gulbay M, Gurbuz O (2010) Plasma fibroblast growth factor-23 levels are independently associated with carotid artery atherosclerosis in maintenance hemodialysis patients. Hemodial Int 14(4): 425-432.
  207. Stubbs J, Liu S, Quarles L (2007) Role of fibroblast growth factor 23 in phosphate homeostasis and pathogenesis of disordered mineral metabolism in chronic kidney disease. Semin Dial 20(4): 302-308.
  208. El Abbadi M, Pai A, Leaf E, Yang H, Bartley B, et al. (2009) Phosphate feeding induces arterial medial calcification in uremic mice: role of serum phosphorus, fibroblast growth factor-23, and osteopontin. Kidney Int 75(12): 1297-1307.
  209. Takei Y, Yamamoto H, Sato T, Otani A, Kozai M, et al. (2012) Stanniocalcin 2 is associated with ectopic calcification in alpha-klotho mutant mice and inhibits hyperphosphatemia-induced calcification in aortic vascular smooth muscle cells. Bone 50(4): 998-1005.
  210. Murphy Jr W, Nedden Dz D, Gostner P, Knapp R, Recheis W, et al. (2003) The iceman: discovery and imaging. Radiology 226(3): 614-629.
  211. Virchow R (1855) Cell Theory and Neoplasia. Arch Pathol Anat 8: 103-113.
  212. Virchow R (1966) Die Cellularpathologie in ihrer Begrundung auf physiologische und pathologische Gewebslehre. In: Verlag von August Hirschwald, Berlin: 1858. Hildesheim: Georg Olms Verlagsbuchhandlung pp. 327-329.
  213. Rocha Singh K, Zeller T, Jaff M (2014) Peripheral Arterial Calcification: Prevalence, Mechanism, Detection, and Clinical Implications. Catheterization and Cardiovascular Interventions 83(6): E212-220.
  214. Sage A, Tintut Y, Demer L (2010) Regulatory mechanisms in vascular calcification. Nat Rev Cardiol 7(9): 528-536.
  215. Shao J, Cheng S, Sadhu J, Towler D (2010) Inflammation and the osteogenic regulation of vascular calcification: A review and perspective. Hypertension 55(3): 579-592.
  216. Nitta K (2011) Vascular calcification in patients with chronic kidney disease. Ther Apher Dial 15(6): 513-521.
  217. Bostrom K, Watson K, Horn S, Wortham C, Herman I, et al. (1993) Bone morphogenetic protein expression in human atherosclerotic lesions. J Clin Invest 91(4): 1800-1809.
  218. Sallam T, Cheng H, Demer L, Tintut Y (2013) Regulatory circuits controlling vascular calcification. Cell Mol Life Sci 70(17): 3187-3197.
  219. Shroff R, McNair R, Figg N, Skepper J, Schurgers L, et al. (2008) Dialysis accelerates medial vascular calcification in part by triggering smooth muscle cell apoptosis. Circulation 118(17): 1748-1757.
  220. Shanahan C (2007) Inflammation ushers in calcification: A cycle of damage and protection? Circulation 116(24): 2782-2785.
  221. Reid D, Shanahan C, Duer M, Arroyo L, Schoppet M, et al. (2012) Lipids in biocalcification: contrasts and similarities between intimal and medial vascular calcification and bone by NMR. J Lipid Res 53(8): 1569-1575.
  222. Wu M, Rementer C, Giachelli C (2013) Vascular calcification: an update on mechanisms and challenges in treatment. Calcif Tissue Int 93(4): 365-373.
  223. Rutsch F, Nitschke Y, Terkeltaub R (2011) Genetics in arterial calcification: pieces of a puzzle and cogs in a wheel. Circ Res 109(5): 578-592.
  224. Li Q, Jiang Q, Schurgers L, Uitto J (2007) Pseudoxanthoma elasticum: reduced gammaglutamyl carboxylation of matrix gla protein in a mouse model (Abcc6-/-). Biochem Biophys Res Commun 364(2): 208-213.
  225. Giachelli C (2004) Vascular calcification mechanisms. J Am Soc Nephrol 15(12): 2959-2964.
  226. Giachelli C (2003) Vascular calcification: in vitro evidence for the role of inorganic phosphate. J Am Soc Nephrol 14(9 suppl 4): S300-304.
  227. Giachelli C, Bae N, Almeida M, Denhardt D, Alpers C, et al. (1993) Osteopontin is elevated during neointima formation in rat arteries and is a novel component of human atherosclerotic plaques. J Clin Invest 92(4): 1686-1696.
  228. Levy R, Schoen F, Levy J, Nelson A, Howard S, et al. (1983) Biologic determinants of dystrophic calcification and osteocalcin deposition in glutaraldehyde-preserved porcine aortic valve leaflets implanted subcutaneously in rats. Am J Pathol 113(2): 143-155.
  229. Kapustin A, Shanahan C (2012) Calcium regulation of vascular smooth muscle cell derived matrix vesicles. Trends Cardiovasc Med 22(5): 133-137.
  230. Kendrick J, Chonchol M (2011) The role of phosphorus in the development and progression of vascular calcification. Am J Kidney Dis 58(5): 826-834.
  231. Giachelli C, Speer M, Li X, Rajachar R, Yang H (2005) Regulation of vascular calcification: roles of phosphate and osteopontin. Circ Res 96: 717-722.
  232. Kockx M, De Meyer G, Muhring J, Jacob W, Bult H, et al. (1998) Apoptosis and related proteins in different stages of human atherosclerotic plaques. Circulation 97: 2307-2315.
  233. Proudfoot D, Skepper J, Hegyi L, Bennett M, Shanahan C, et al. (2000) Apoptosis regulates human vascular calcification in vitro: evidence for initiation of vascular calcification by apoptotic bodies. Circ Res 87: 1055-1062.
  234. Pal S, Golledge J (2011) Osteoprogenitors in vascular calcification: a circulating cell theory. J Atheroscler Thromb 18: 551-559.
  235. Tang Z, Wang A, Yuan F (2012) Differentiation of multipotent vascular stem cells contributes to vascular diseases. Nat Commun 3: 875.
  236. Shao J, Cheng S, Pingsterhaus J, Charlton Kachigian N, Loewy A, et al. (2005) Msx2 promotes cardiovascular calcification by activating paracrine Wnt signals. J Clin Invest 115: 1210-1220.
  237. Lee H, Woo K, Ryoo H, Baek J (2010) Tumor necrosis factor-alpha increases alkaline phosphatase expression in vascular smooth muscle cells via MSX2 induction. Biochem Biophys Res Commun 391: 1087-1092.
  238. Smith E, Ford M, Tomlinson L, Rajkumar C, McMahon L, et al. (2012) Phosphorylated fetuin-A-containing calciprotein particles are associated with aortic stiffness and a procalcific milieu in patients with pre-dialysis CKD. Nephrol Dial Transplant 27: 1957-1966.
  239. Menini S, Iacobini C, Ricci C (2013) The galectin 3/RAGE dyad modulates vascular osteogenesis in atherosclerosis. Cardiovasc Res 100: 472-480.
  240. Hofmann Bowman M, Schmidt A (2011) S100/calgranulins EN-RAGEing the blood vessels: implications for inflammatory responses and atherosclerosis. Am J Cardiovasc Dis 1: 92-100.
  241. Averill M, Kerkhoff C, Bornfeldt K (2012) S100A8 and S100A9 in cardiovascular biology and disease. Arterioscler Thromb Vasc Biol 32: 223-229.
  242. Hofmann Bowman M, McNally E (2012) Genetic pathways of vascular calcification. Trends Cardiovasc Med 22: 93-98.
  243. Caudrillier A, Mentaverri R, Brazier M, Kamel S, Massy Z (2010) Calcium-sensing receptor as a potential modulator of vascular calcification in chronic kidney disease. J Nephrol 23: 17-22.
  244. Alam M, Kirton J, Wilkinson F (2009) Calcification is associated with loss of functional calcium-sensing receptor in vascular smooth muscle cells. Cardiovasc Res 81: 260-268.
  245. Ivanovski O, Nikolov I, Joki N (2009) The calcimimetic R-568 retards uremiaenhanced vascular calcification and atherosclerosis in apolipoprotein E deficient (apoE-/-) mice. Atherosclerosis 205: 55-62.
  246. Henaut L, Boudot C, Massy Z (2014) Calcimimetics increase CaSR expression and reduce mineralization in vascular smooth muscle cells: mechanisms of action. Cardiovasc Res 101: 256-265.
  247. Parhami F, Tintut Y, Ballard A, Fogelman A, Demer L (2001) Leptin enhances the calcification of vascular cells: artery wall as a target of leptin. Circ Res 88: 954-960.
  248. Zeadin M, Butcher M, Werstuck G, Khan M, Yee C, et al. (2009) Effect of leptin on vascular calcification in apolipoprotein E-deficient mice. Arterioscler Thromb Vasc Biol 29: 2069-2075.
  249. Hill J, Olson E, Griendling K, Kitsis R, Stull J (2012) Muscle: fundamental biology and mechanisms of disease. Elsevier Science & Technology Books.
  250. Doherty T, Uzui H, Fitzpatrick L (2002) Rationale for the role of osteoclast-like cells in arterial calcification. FASEB J 16: 577-582.
  251. Mozar A, Haren N, Chasseraud M, Loïc Louvet, Cécile Mazière, et al. (2008) High extracellular inorganic phosphate concentration inhibits RANK-RANKL signaling in osteoclast-like cells. J Cell Physiol 215: 47-54.
  252. Tolle M, Reshetnik A, Schuchardt M, Hohne M, van der Giet M, et al. (2015) Arteriosclerosis and vascular calcification: causes, clinical assessment, and therapy. Eur J Clin Invest 45(9): 976-985.
  253. Lilly S, Qasim A, Mulvey C, Churchill T, Reilly M, et al. (2013) non-compressible arterial disease and the risk of coronary calcification in type-2 diabetes. Atherosclerosis 230: 17-22.
  254. Latham R, Westerhof N, Sipkema P, Rubal B, Reuderink P, et al. (1985) Regional wave travel and reflections along the human aorta: a study with six simultaneous micromanometric pressures. Circulation 72: 1257-1269.
  255. Laurent S, Cockcroft J, Van Bortel L, Boutouyrie P, Giannattasio C, et al. (2006) Expert consensus document on arterial stiffness: methodological issues and clinical applications. Eur Heart J 27: 2588-2605.
  256. Roos C, Auger D, Djaberi R, de Koning E, Rabelink T, et al. (2013) Relationship between left ventricular diastolic function and arterial stiffness in asymptomatic patients with diabetes mellitus. Int J Cardiovasc Imaging 29: 609-616.
  257. Terai M, Ohishi M, Ito N, Takagi T, Tatara Y, et al. (2008) Comparison of arterial functional evaluations as a predictor of cardiovascular events in hypertensive patients: the Non-Invasive Atherosclerotic Evaluation in Hypertension (NOAH) study. Hypertens Res 31: 1135-1145.
  258. Wang K, Cheng H, Sung S, Chuang S, Li C, et al. (2010) Wave reflection and arterial stiffness in the prediction of 15- year all-cause and cardiovascular mortalities: a community-based study. Hypertension 55: 799-805.
  259. Pannier B, Guerin A, Marchais S, Safar M, London G, et al. (2005) Stiffness of capacitive and conduit arteries: prognostic significance for end-stage renal disease patients. Hypertension 45: 592-596.
  260. Tardy Y, Meister J, Perret F, Brunner H, Arditi M, et al. (1991) Non-invasive estimate of the mechanical properties of peripheral arteries from ultrasonic and photoplethysmographic measurements. Clin Phys Physiol Meas 12: 39-54.
  261. Scuteri A, Morrell C, Orru M, Strait J, Tarasov K, et al. (2014) Longitudinal perspective on the conundrum of central arterial stiffness, blood pressure, and aging. Hypertension 64: 1219-1227.
  262. Blacher J, Guerin A, Pannier B, Marchais S, Safar M, et al. (1999) Impact of aortic stiffness on survival in end-stage renal disease. Circulation 99: 2434-2439.
  263. LeBoeuf A, Mac-Way F, Utescu M, De Serres S, Douville P, et al. (2011) Impact of dialysate calcium concentration on the progression of aortic stiffness in patients on haemodialysis. Nephrol Dial Transplant 26: 3695-3701.
  264. Kauppila L, Polak J, Cupples L, Hannan M, Kiel D, et al. (1997) New indices to classify location, severity, and progression of calcific lesions in the abdominal aorta: A 25-year follow-up study. Atherosclerosis 132: 245-250.
  265. Duhn V, D Orsi E, Johnson S, D Orsi C, Adams A, et al. (2011) Breast arterial calcification: A marker of medial vascular calcification in chronic kidney disease. Clin J Am Soc Nephrol 6: 377-382.
  266. Otsuka F, Sakakura K, Yahagi K, Joner M, Virmani R, et al. (2014) Has our understanding of calcification in human coronary atherosclerosis progressed? Arterioscler Thromb Vasc Biol 34: 724-736.
  267. Abdelmalek J, Stark P, Walther C, Ix J, Rifkin D, et al. (2012) Associations between coronary calcification on chest radiographs and mortality in hemodialysis patients. Am J Kidney Dis 60: 990-997.
  268. London G, Guerin A, Marchais S, Metivier F, Pannier B, et al. (2003) Arterial media calcification in end-stage renal disease: impact on all-cause and cardiovascular mortality. Nephrol Dial Transplant 18(9): 1731-1740.
  269. Lindbom A (1950) Arteriosclerosis and arterial thrombosis in the lower limb; a roentgenological study. Acta Radiol Suppl 80: 1-80.
  270. Schlieper G, Aretz A, Verberckmoes S, Kruger T, Behets G, et al. (2010) Ultrastructural analysis of vascular calcifications in uremia. J Am Soc Nephrol 21: 689-696.
  271. Schousboe J, Wilson K, Kiel D (2006) Detection of abdominal aortic calcification with lateral spine imaging using DXA. J Clin Densitom 9: 302-308.
  272. Knez A, Becker C, Becker A, Leber A, White C, et al. (2002) Determination of coronary calcium with multi-slice spiral computed tomography: a comparative study with electron-beam CT. Int J Cardiovasc Imaging 18: 295-303.
  273. De Rotte A, Koning W, Truijman M, den Hartog A, Bovens S, et al. (2014) Seven-tesla magnetic resonance imaging of atherosclerotic plaque in the significantly stenosed carotid artery: A feasibility study. Invest Radiol 49: 749-757.
  274. Dangas G, Maehara A, Evrard S, Sartori S, Li J, et al. (2014) Coronary artery calcification is inversely related to body morphology in patients with significant coronary artery disease: A three-dimensional intravascular ultrasound study. Eur Heart J Cardiovasc Imaging 15: 201-209.
  275. Mehanna E, Bezerra H, Prabhu D, Brandt E, Chamie D, et al. (2013) Volumetric characterization of human coronary calcification by frequency-domain optical coherence tomography. Circ J 77: 2334-2340.
  276. Nasir K, Rivera J, Yoon Y, Chang S, Choi S, et al. (2010) Variation in atherosclerotic plaque composition according to increasing coronary artery calcium scores on computed tomography angiography. Int J Cardiovasc Imaging 26: 923-932.
  277. Motoyama S, Kondo T, Sarai M, Sugiura A, Harigaya H, et al. (2007) Multislice computed tomographic characteristics of coronary lesions in acute coronary syndromes. J Am Coll Cardiol 50(4): 319-326.
  278. Keelan P, Bielak L, Ashai K, Jamjoum L, Denktas A, et al. (2001) Long-term prognostic value of coronary calcification detected by electron-beam computed tomography in patients undergoing coronary angiography. Circulation 104(4): 412-417.
  279. Ehara S, Kobayashi Y, Yoshiyama M, Shimada K, Shimada Y, et al. (2004) Spotty calcification typifies the culprit plaque in patients with acute myocardial infarction: an intravascular ultrasound study. Circulation 110(22): 3424-3429.
  280. Roijers R, Debernardi N, Cleutjens J, Schurgers L, Mutsaers P, et al. (2011) Microcalcifications in early intimal lesions of atherosclerotic human coronary arteries. Am J Pathol 178: 2879-2887.
  281. Schurgers L, Teunissen K, Knapen M, Martijn Kwaijtaal, Rob van Diest, et al. (2005) Novel conformation-specific antibodies against matrix gamma-carboxyglutamic acid (Gla) protein: undercarboxylated matrix Gla protein as marker for vascular calcification. Arterioscler Thromb Vasc Biol 25: 1629-1633.
  282. Cranenburg E, Koos R, Schurgers L, Magdeleyns E, Schoonbrood T, et al. (2010) Characterisation and potential diagnostic value of circulating matrix Gla protein (MGP) species. Thromb Haemost 104(4): 811-822.
  283. Schurgers L, Cranenburg E, Vermeer C (2008) Matrix Gla-protein: the calcification inhibitor in need of vitamin K. Thromb Haemost 100(4): 593-603.
  284. Ueland T, Gullestad L, Dahl C, Aukrust P, Aakhus S, et al. (2010) Undercarboxylated matrix Gla protein is associated with indices of heart failure and mortality in symptomatic aortic stenosis. J Intern Med 268(4): 483-492.
  285. Schurgers L, Barreto D, Barreto F, Sophie Liabeuf, Cédric Renard, et al. (2010) The circulating inactive form of matrix gla protein is a surrogate marker for vascular calcification in chronic kidney disease: A preliminary report. Clin J Am Soc Nephrol 5(4): 568-575.
  286. Schlieper G, Westenfeld R, Kruger T, Ellen C Cranenburg, Elke J Magdeleyns, et al. (2011) Circulating nonphosphorylated carboxylated matrix gla protein predicts survival in ESRD. J Am Soc Nephrol 22(2): 387-395.