Next Article in Journal
The Aquaporin-3-Inhibiting Potential of Polyoxotungstates
Next Article in Special Issue
Vascular Calcification—New Insights into Its Mechanism
Previous Article in Journal
Historical Perspective: Models of Parkinson’s Disease
Previous Article in Special Issue
Novel Insight into Neuroimmune Regulatory Mechanisms and Biomarkers Linking Major Depression and Vascular Diseases: The Dilemma Continues
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of Vitamin D in Modulating Mesenchymal Stem Cells and Endothelial Progenitor Cells for Vascular Calcification

1
Division of Nephrology, Department of Medicine, Cardinal-Tien Hospital, New Taipei City 231, Taiwan
2
School of Medicine, Fu-Jen Catholic University, New Taipei City 234, Taiwan
3
Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
4
Division of Nephrology, Department of Medicine, Fu-Jen Catholic University Hospital, New Taipei City 243, Taiwan
5
Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
6
Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, Taipei 235, Taiwan
7
Division of Nephrology, Department of Internal Medicine, Tungs’ Taichung Metroharbor Hospital, Taichung City 43304, Taiwan
8
Department of Nephrology, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
9
College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
10
Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
11
Graduate Institute of Toxicology, National Taiwan University College of Medicine, Taipei 104, Taiwan
12
Nephrology division, Department of Internal Medicine, National Taiwan University Hospital, Taipei 100, Taiwan
13
Department of Internal Medicine, National Taiwan University Hospital BeiHu Branch, Taipei 108, Taiwan
14
Division of Nephrology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, and School of Medicine, Buddhist Tzu Chi University, Hualien, Taiwan
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2020, 21(7), 2466; https://doi.org/10.3390/ijms21072466
Submission received: 29 February 2020 / Revised: 26 March 2020 / Accepted: 30 March 2020 / Published: 2 April 2020
(This article belongs to the Special Issue Mechanisms of Vascular Calcification)

Abstract

:
Vascular calcification, which involves the deposition of calcifying particles within the arterial wall, is mediated by atherosclerosis, vascular smooth muscle cell osteoblastic changes, adventitial mesenchymal stem cell osteoblastic differentiation, and insufficiency of the calcification inhibitors. Recent observations implied a role for mesenchymal stem cells and endothelial progenitor cells in vascular calcification. Mesenchymal stem cells reside in the bone marrow and the adventitial layer of arteries. Endothelial progenitor cells that originate from the bone marrow are an important mechanism for repairing injured endothelial cells. Mesenchymal stem cells may differentiate osteogenically by inflammation or by specific stimuli, which can activate calcification. However, the bioactive substances secreted from mesenchymal stem cells have been shown to mitigate vascular calcification by suppressing inflammation, bone morphogenetic protein 2, and the Wingless-INT signal. Vitamin D deficiency may contribute to vascular calcification. Vitamin D supplement has been used to modulate the osteoblastic differentiation of mesenchymal stem cells and to lessen vascular injury by stimulating adhesion and migration of endothelial progenitor cells. This narrative review clarifies the role of mesenchymal stem cells and the possible role of vitamin D in the mechanisms of vascular calcification.

1. Introduction

Vascular calcification, which involves the deposition of calcifying particles within the endothelial layer or smooth muscle within the medial layer, is an important issue due to its associated complications, such as peripheral arterial occlusive disease and coronary artery disease [1,2,3]. Several conditions, including insulin resistance, hypertension, acute decompensated heart failure, chronic kidney disease (CKD), dyslipidemia, vitamin D deficiency, and metabolic syndrome, are associated with vascular calcification [4,5,6]. Vascular calcification is a predictor of overall mortality and poor arteriovenous graft maturation in patients with CKD [7,8]. As these risk factors can influence the endothelial layer and the smooth muscle cells simultaneously, measures to prevent them are vital.
Recently, mesenchymal stem cells (MSCs) and endothelial progenitor cells (EPCs) were considered important for the development of vascular calcification. MSCs are known as either marrow stromal cells, bone marrow fibroblasts, or skeletal stem cells. They could be classified as bone marrow derived MSCs or pericytes based on their origin [9]. Following their activation by inflammation or specific stimuli, they may differentiate osteogenically, which can activate calcification. Kramann et al. suggested that MSCs within the adventitial layer trigger vascular calcification by translocating into the endothelial and the medial layers [10]. Microvesicles derived from injured endothelial cells induce vascular calcification in part through the attraction of MSCs. Transcriptional modulation by specific agents, such as vitamin D, is a possible therapeutic approach to mitigating such vascular calcification [11].
EPCs originating from the bone marrow were shown to be an important mechanism in the repair of injured endothelial cells [12]. However, the EPC phenotype was altered under specific pathologic states, such as the accumulation of uremic toxins [13]. The EPCs with osteogenic character were related to the severity of the vascular calcification [14], and the pharmacologic dose of active vitamin D supplement might enhance the expression of calcifying EPCs in CKD patients [15]. However, nutritional vitamin D supplement may attenuate the severity of vascular calcification or aortic stiffness [16,17]. This significance deserves further clarify. This review explains a possible role of MSCs and EPCs in the mechanisms of vascular calcification and a possible role of vitamin D in that mechanism.

2. Mechanism of Vascular Calcification

2.1. Endothelial Injury Causing Vascular Calcification

Vascular calcification is characterized by the deposition of hydroxyapatite crystals within the arterial layer, which may originate from atherosclerosis or arteriosclerosis (Figure 1, blue arrow). [1]. Calcifying tissue within the vascular layer may originate from apoptosis within endothelial cells or osteoblastic changes in smooth muscle [18]. The intimal calcification is initiated by the focal retention of apo B–containing lipoproteins in the subendothelial extracellular matrix [19]. In the subendothelial layer, lipid-induced sequential migration of macrophages occurs. The macrophage phagocytizes the lipoprotein cholesterol complex. However, the excessive oxidized lipoprotein induces macrophage apoptosis [18]. The atheroma with apoptotic macrophages and oxidized lipoprotein serves as the necrotic core of the subendothelial layer and initiates the process of mineralization [20]. In addition to subendothelial lipid accumulation, the influences of stress on the endothelial layer, such as the activation of the renin–angiotensin–aldosterone system (RAAS), fluid overload, and insulin resistance, exacerbates the endothelial injury. Montezano et al. demonstrated the direct effect of angiotensin II on endothelial injuries; angiotensin II increased the release of reactive oxygen species by activating vascular nicotinamide adenine dinucleotide phosphate oxidase [21]. Instead of repairing in the endothelial layer, the replacement of the fibrotic tissue by fibroblasts reduced the endothelial compliance. Thus, endothelial injury due to calcification was aggravated by the increased shearing stress. Subendothelial lipid accumulation initiated endothelial injury, and the subsequent inflammation triggered by macrophages and the replacement by hydroxyapatite-associated crystals accelerated atherosclerosis and increased arterial stiffness.

2.2. The Role of Vascular Smooth Muscle Ccells (VSMCs) in Vascular Calcification

Vascular smooth muscle cells (VSMCs) within the medial layer of the arteries underwent rapid morphologic and functional changes after confronting environmental stimuli [22]. Specific stimuli on the smooth muscle layer activated osteoblastic-like differentiation, such as hyperphosphatemia. Hyperphosphatemia is a common complication in CKD patients because of the decrease in the renal clearance of phosphate, which is also related to cardiovascular mortality [23]. Giachelli et al. found that inorganic phosphate promoted the osteogenic differentiation of VSMC directly by induction of a sodium-dependent phosphate transporter (Pit-1) [24]. The core-binding factor α-1 (Cbfa-1) served as the transcription factor activated during the osteogenic differentiation by inducing the expression of tissue-nonspecific alkaline phosphatase [25,26,27].
During this osteoblastic transdifferentation process, translation of the Runx-activated canonical Wingless-INT (Wnt)-β-catenin signaling accelerated active calcium deposition and vascular calcification (Figure 1, yellow and blue arrow) [28]. Downstream bone morphogenetic protein 2 (BMP2) was activated by Wnt and propagated the osteogenic differentiation [29]. Hyperphosphatemia also stimulated the serum- and the glucocorticoid-inducible kinase (SGK1) with subsequent activation of the transcription factor NF-kB in VSMCs [30,31]. Therefore, hyperphosphatemia-mediated osteoblastic change within the smooth muscle layer may be a prominent mechanism of vascular calcification. However, the release of matrix vesicles from VSMCs was also associated with vascular calcification [32].
In hyperphosphatemia, proliferative VSMCs with low calcitonin and α-smooth muscle actin were found to serve as the transitional form between contractile and calcifying smooth muscle cells. Hyperphosphatemia also stimulated the VSMC apoptosis process [33]. The apoptotic body originating from VSMCs served as the nucleation site of mineral deposition [34]. Inorganic pyrophosphate originated from VSMC serves as the endogenous calcification inhibitors by the ectonucleotide pyrophosphatase/phosphodiesterase (ENPP1) mediated breakdown of nucleotide triphosphates or by the transmembrane protein ankylosis protein homolog (ANKH) mediated transportation. [35]. If the matrix vesicles (MV) contained sufficient calcification inhibitors, such as fetuin-A, the vascular calcification process would be mitigated [33,36]. Therefore, lessening the phosphate burden is important in preventing cardiovascular damage in CKD patients.
Beyond the hyperphosphatemia, protein-bound uremic toxin (e.g., indoxy sulfate) might induce phenotypic changes within the VSMCs by increasing the oxidative stress. Uremic toxins were able to alter the glucose metabolism within the VSMCs (and/or endothelial cells) and therefore increased the cellular release of the calcifying exosome into the artery and worsened the vascular calcification [37]. The uremic toxin also induced the osteoblastic differentiation of VSMCs and promoted further calcification [38]. This evidence indicated that, in CKD patients, the VSMC phenotype might be modulated, and osteoblastic differentiation might be initiated.
The role of vitamin D on VSMCs has been discussed in many studies. Valcheva et al. noticed that the VSMCs from vitamin D receptor-knock out mice had higher renin activity and premature senescence [39]. Based on the current evidence, in vitro studies demonstrated that vitamin D inhibited the mineralization of VSMCs treated with phosphate and tumor necrosis factor alpha (TNF-α) [40]. On the other hand, Chen et al. provided evidence that 1,25(OH)2D decreased VSMCs treated with endothelin mediated by cyclin-dependent kinase 2 (Cdk-2) activity [41]. Contrary evidence also demonstrated the vitamin D might stimulate vascular calcification by modulating the expression of parathyroid hormone-related peptide or the receptor activator of nuclear factor kappa-Β ligand/osteoprotegerin of VSMC [42,43]. The pharmacologic or the supraphysiologic concentrations of active or nutritional vitamin D might contribute to the vascular calcification in vivo studies [44,45]. Therefore, vitamin D has rather complex effects on calcification from the aspect of VSMC, and more advanced studies are needed to elucidate the role of vitamin D in vascular calcification.

2.3. The Role of Adventitial MSCs and Pericyte in Vascular Calcification

Adventitial MSCs (cluster of differentiation (CD)34+ CD31- CD146- CD45- [46]) are considered to contribute to vascular calcification. From the postmortem study of Yang et al., adventitial calcification occurred during the process of intracranial artery calcification [47], and the measurable adventitial vaso vasorum was predictive of the progressive atherosclerotic change in the intracranial arteries [48]. Researchers demonstrated that MSCs reside within the adventitial layer [46], and that MSC differentiation might be initiated after vascular injury. For instance, angiotensin II sensitized the MSCs with fibrogenic character by activating NF-κB [49].
Tang et al. provided evidence that the adventitial MSCs carrying the stem cell antigen 1 (Sca-1) surface protein were activated to repair arterial injuries [50]. At the same time, multiple inflammatory cells lie within the adventitial layer, and the pathologic status might dysregulate the repair process and induce vascular calcification (Figure 1, yellow arrow) [51]. Del Toro et al. reported that, in vivo, the adventitial MSC activated chemokine-mediated monocyte and neutrophil aggregation, thus exacerbating subendothelial injury [52]. Kramann et al. reported that vascular calcification could be reversed after the genetic ablation of glioma-associated oncogene homolog 1 (Gli1) for the migration of MSCs carrying human Gli1 from the adventitial layer to the smooth muscle and the endothelial layers in specific animal models (such as those fed with high-fat diets or nephrectomized rats) [10].
Sun et al. demonstrated that human adventitial progenitor cells carrying CD10, a common surface marker of acute lymphoblastic leukemia and lymphoid progenitors, have the potential for osteogenic differentiation through the Sonic hedgehog-signaling pathway [53]. In addition to MSCs, pericytes residing within the adventitia can migrate after intimal injury. Pericytes (with surface markers platelet-derived growth factor receptor (PDGF-R)β, α-smooth muscle actin (αSMA), and Neural/glial antigen 2 (NG-2) [54]) are located within the adventitial layer of the vasa vasorum. Vascular injury induces pericyte differentiation and migration during neointima formation and vascular calcification. After arterial injury, the pericyte itself contributed to the restenosis after arterial injury by modulating the PDGF signaling [54,55]. The role of MSCs within the adventitial layer is still not clear in humans. Based on the recent studies, the adventitial MSCs phenotype could be modified by endothelial injury and arteriosclerosis, and such modifications might worsen the vascular calcification. The strategy on modulating adventitial MSCs could be a new aspect in the future.

2.4. The Role of Matrix Vesicles/Exosomes and Calciprotein Particles Containing Insufficient Calcification Inhibitors

Plasma is always supersaturated with respect to the apatitic solid phase [56]. In research on osteoporosis and adynamic bone disease, the exchangeable calcium and phosphate pool was supersaturated, and sequential crystal formation occurred if there were no sufficient calcification inhibitors [57,58]. As mentioned previously, endothelial cells or VSMCs released exosomes or matrix vesicles when damaged. The chemokine homeostasis would be disrupted when recruiting erythrocytes, or platelets could release extracellular vesicles at the damaged endothelium [59]. In osteochondrogenic VSMCs, calcifying cells released matrix vesicles containing calcium, phosphate, lipoprotein, and calcification inhibitors [36]. The released exosome containing specific microRNA(miR), such as miR-135a(*), miR-762, miR-714, and miR-712 [60], or miR-32 [61], could be transported into nearby VSMC in a heparin sulphate proteoglycans (HSPG)-dependent manner [62], and such exosomes could stimulate osteogenic differentiation of VSMC.
The calcification inhibitors were assembled with apolipoprotein, crystalline, and amorphous hydroxyapatite calcium as calciprotein particles (CPPs) [63]. As the CPPs contained sufficient calcification inhibitors, such as fetuin-A, the CPPs were integrated into spherical rather than unstructured minerals. Such CPPs are called primary CPPs, and the primary CPPs were cleared through the scavenger receptor A, present on hepatic endothelial cells [63,64]. In subjects with insufficient calcification inhibitors, the CPPs turned into unstructured minerals with a diameter of 120–150 nm, which was larger than the primary CPPs (60–70 nm) [6]. These unstructured CPPs are called secondary CPPs, and such secondary CPPs were predictive for vascular calcification and cardiovascular mortality in uremic patients (Figure 1) [65]. Clinical evidence suggested that patients with CKD and a higher concentration of secondary CPPs had a higher incidence of vascular calcification [66]. Therefore, maintaining sufficient calcification inhibitors should be a therapeutic strategy for treating vascular calcification.
Among the calcification inhibitors, matrix Gla protein (MGP) phosphorylation and carboxylation provided the effectiveness for chelating calcium [67]. Vitamin K is essential for the post-translational conversion to γ-carboxyglutamate [68]. Under vitamin K sufficient status, phosphorylated MGP also avoided osteoblastic changes of VSMCs [69]. Mature MGP formed mineralized complex with fetuin-A, calcium, and phosphorus ion to lessen the mineral composition within vessels (Figure 1) [70]. In CKD patients, the secondary CPP was associated with insufficient MGP [71]. It is rational to supply vitamin K in subjects such as CKD patients with vitamin K deficiency [72]. Vitamin D deficiency, which is common in CKD patients, involves a functional vitamin K deficiency [73]. Cashman et al. provided evidence that the vitamin D status was correlated negatively with the uncarboxylated osteocalcin [74]. On the other hand, vitamin D might enhance the carboxylated MGP productions based on in vitro and in vivo evidence. In the osteoblast, vitamin D induced osteogenesis by enhancing ɣ-carboxylated-MGP-containing osteocalcin [75]. After treating the vitamins D and K with the osteoblast from the diabetic mice, the bone anabolism was enhanced [73]. In this manner, the extraosseous calcification might be lessened. Therefore, vitamin D supplements should be another strategy in treating vascular calcification based on the aspects of the CPPs.

3. The Role of EPCs, Hematopoietic Progenitor Cells, and MSCs in Vascular Calcification

From the traditional aspects, vascular calcification involves subendothelial hydroxyapatite formation, the osteogenic transformation of smooth muscle cells, and dysregulation/reductions in the activity of calcification inhibitors. In severe cases of ischemic limbs or peripheral occlusive arterial disease, the exhausted production of endothelial/hematopoietic stem cells and bone marrow MSCs contributes to the progression of vascular calcification.

3.1. EPCs and Arterial Calcification

As subendothelial atheroma occludes arteries, hypoxia-inducible factor-1-alpha (HIF-1-alpha) regulates the gene expression of vascular endothelial growth factor (VEGF). The activated VEGF was shown to modulate matrix metalloproteinase-9 (MMP-9) activity and increase the mobilization of EPCs [76]. In physiological hypoxia, angiogenesis was shown to repair a damaged endothelium by promoting the differentiation of EPCs [77]. The circulating EPCs migrated and invaded the subendothelial region to replace injured endothelial cells and regulated the differentiation of the surrounding stromal cells [78]. However, the circulating EPCs may be stimulated into endothelial regeneration or calcification. For example, in patients with end-stage renal disease, EPCs with surface markers of CD34+/CD133−/KDR+/CD45− were activated by active vitamin D, which lowered the expression of osteocalcin [79]. Furthermore, the concentration of circulating endothelial cells with markers of CD34+/CD133+/KDR+ can predict cardiovascular mortality in patients with atherosclerosis and those requiring hemodialysis [80]. However, EPCs bearing the markers CD34+/CD133+/VEGFR+ can enable vasculogenesis [81]. In patients with CKD, the accumulation of uremic toxin disrupted EPC migration into the endothelium. Wu et al. demonstrated that the protein-bound uremic toxin indoxyl sulfate down-regulated endothelial vacuolization by disrupting the effect of HIF-1-alpha [13]. Thus, indoxyl sulfate disrupted EPCs regeneration and endothelial repair.

3.2. Hematopoietic Progenitor Cells and Arterial Calcification

The hematopoietic progenitor cells originating in the bone marrow can differentiate into the myeloid and the lymphoid progenitor cells under oxidative stress. Dutta et al. first demonstrated in an animal model that a myocardial infarction stimulated hematopoietic progenitor cells production and worsened atherosclerosis [82]. Chronic stress decreased the expression of chemokine (C-X-C motif) ligand 12 CXCL12 within the bone marrow and facilitated the release of inflammatory monocytes and neutrophils [83]. The endothelial chondrocyte-like phenotype is common during vascular calcification, and monocytic cells can be programmed through stimulation of inflammatory cytokines, such as transforming growth factor-1β, to differentiate with chondrocyte characters, such as generate type II collagens [84].
Doehring et al. demonstrated that transplanted bone marrow CD34+/CD13+ myeloid progenitor cells transdifferentiated into chondrocyte-like cells in an atherosclerotic animal model [85]. Thus, bone marrow hematopoietic progenitor cells can be conditionally stimulated into monocytes or osteoclasts, which may regulate osteogenesis within the endothelial or the arterial smooth muscle cells. Recently, Cho et al. showed that bone marrow–derived hematopoietic progenitor cells (Sca-1+/PDGFRα−) have osteoclastogenic potency, which can lead to osteoclast-mediated bone resorption.
As inflammatory cytokines, such as interleukin-1 or interleukin-5, increased, Sca-1+/PDGFRα− decreased and was associated with more severe osteogenesis and vascular calcification within the vascular wall [86]. Recently, Frodermann et al. provided evidence that the exercise decreased the release of hematopoietic progenitor cells from the bone marrow by modulating the leptin release from the adipocyte. In this manner, the cardiovascular damage was relieved by lessening the inflammatory process [87]. This evidence gave us clues that the pathologic status induced the inflammatory differentiation of hematopoietic progenitor cells, and that such inflammation worsened the endothelial injury. Certain interventions lessening the differentiation might be a therapeutic strategy for treating endothelial injuries and sequential vascular calcification.

3.3. MSCs and Arterial Calcification

MSCs are multipotential stromal cells that can differentiate into osteoblasts, chondrocytes, or adipocytes. MSCs reside within adipose tissue, bone marrow, the umbilical cord, and the adventitial/medial layer of the vasculature. Cluster of differentiation (CD) markers indicate the origin of MSCs. For example, stromal stem cells from bone marrow have the surface markers SH2, SH3, CD29, CD44, CD71, CD90, CD106, CD120a, and CD124. The surface markers of MSCs determine whether they have the potential to differentiate into endothelial cells under specific stimuli. Miranville et al. demonstrated that adipose tissue-derived MSCs with CD34+/CD31− markers differentiated into endothelial cells and alleviated neointima formation [88]. However, MSCs residing within tissues other than the adventitial layer contributed to inflammation rather than differentiation into endothelial cells during osteogenic differentiation [89]. This was because MSCs that originated from adipose tissue or bone marrow required collagenase to cleave the hindrance posed by the stromal cells [90]. In summary, adipose MSCs have potential for osteogenic differentiation, and such characteristics might be related to the development of vascular calcification.

3.4. Extracelluar Vesicles and Calciprotein Particles Stimulated by MSCs

Extracellular vesicles are the double-layer phospholipid membrane vesicles released from cells. They encapsulate biological molecules such as nucleic acids, diverse cellular proteins, and metabolites [91,92]. As the extracellular vesicle might contain microRNA or specific proteins, it served as the intercellular communication [91]. MSCs had anti-inflammatory and or immunosuppressive properties [93], and the exosomes released from MSCs were identified as a possible therapeutic target for vascular calcification [94]. G Sahoo et al. showed that the exosome released from human stem cells induced endothelial viability in a paracrine manner [95]. Guo et al. reported that exosomes from bone marrow–derived MSCs bear the surface markers CD63 and CD81. Such exosomes hampered VSMC calcification by modulating the microRNA regulating the mitogen-activated protein kinase (MAPK) or the Wnt signaling pathways [96]. Wei et al. demonstrated that extracellular vesicles isolated from the MSCs and coated with heparin-based vehicles maintained patency after arterial graft in rats. This effect was modulated through the transfection of extracellular vesicles from atherogenic macrophages into anti-inflammatory and antiosteogenic macrophages [97]. From the evidence above, the undifferentiated MSCs had anti-inflammatory and/or immunosuppressive properties, and the extracellular vesicles released from MSCs might be a therapeutic strategy for vascular calcification by reducing inflammation.

4. Possible Therapeutic Roles of Vitamin D in MSCs and Vascular Calcification

Vitamin D is an essential hormone provided through exposure to sunlight or through intake from the diet. There are two major types, ergocalciferol and cholecalciferol. After being radiated by ultra-violet B (UVB) light at wavelengths of 290–315 nm, the ergosterol in plants or fungi is synthesized into ergocalciferol. Cholecalciferol originates from keratinocytes. After being radiated by UVB, 7-dehydrocholesterol is transformed into cholecalciferol [98]. The body’s synthesized cholecalciferol or ingested ergocalciferol/cholecalciferol is transported to the liver by a vitamin D transport protein and hydroxylased within the liver, where the vitamin D is transformed into 25-hydroxy vitamin D (25(OH)D, which is transported to the kidneys to be converted to 1,25(OH)2D by 1-alpha hydroxylase. The 1,25(OH)2D is then transported from the cytoplasm into the nucleus to interact with the vitamin D binding protein, which binds to the vitamin D receptor element so that vitamin D can influence the transcription of specific genes [99].
Vitamin D deficiency is common in CKD and diabetes mellitus for several reasons: (1) renal deterioration and proteinuria [100,101,102], (2) reduced 1-alpha hydroxylase activity within the kidney [103,104], (3) increased catabolism of 25(OH)D into inactive metabolite 24,25(OH)2D [103,105], and (4) pharmacological concentrations of vitamin D [106]. Current active vitamin D supplements have microgram concentrations [107]. In vitro studies demonstrated that supraphysiological concentrations of active vitamin D influenced the 25(OH)D production in the liver.
In CKD patients, 1,25(OH)2D was interfered with by fibroblast growth factor 23 (FGF23). FGF23 is the hormone secreted from osteocytes. In the CKD patients with decreased renal excretion of inorganic phosphates, FGF 23 served as the phosphaturic hormone to decrease the reabsorption of phosphate from the proximal tubule in the kidneys [108]. FGF23 directly suppressed the activity of 1-α hydroxylase and increased the activity of 25-hydroxyvitamin D3-24-hydroxylase [109,110]. The decrease of vitamin D and the increase of FGF23 interfered with the osteogenic differentiation of bone marrow MSCs in CKD patients [111,112,113]. Therefore, the correction of vitamin D deficiency is critical to the treatment of vascular calcification, and the synergy of vitamin D and MSCs should be considered in the treatment of vascular calcification.
Vitamin D deficiency is a risk factor and a predictor for cardiovascular disease [114]. In epidemiological studies, vitamin D deficiency was associated with a higher incidence of hypertension [115], coronary artery disease (CAD) [116], fatal stroke [117], and peripheral arterial disease [118]. Vitamin D deficiency itself was associated with impaired peripheral insulin sensitivity [119] and arterial stiffness [120]. The role of vitamin D in vascular disease involves immune modulation by moderating the release of anti-inflammatory cytokines by macrophages [121] or the reduction of RAAS hyperactivity [122,123]. Moreover, vitamin D can regulate carboxylation of the vitamin K-mediated MGP. Carboxylated MGP chelates excessive calcium and lessens extraosseous calcification. Vitamin D enhances osteocalcin and MGP production within osteoblasts. The downstream carboxylation of osteocalcin and MGP improves bone mineralization and mitigates extraskeletal calcification [6]. Beyond the aspects above, the adjunctive role of the vitamin D on MSCs or EPCs in treating vascular calcification is discussed as below.

4.1. The Influence of Vitamin D on EPCs in Vascular Calcification

Vitamin D receptor expression can predict cardiovascular disease. Ai et al. demonstrated that patients with CAD had fewer vitamin D receptors on EPCs than did control patients (Table 1) [124]. Vitamin D supplementation can accelerate EPC migration and differentiation through an angiogenesis-associated pathway. Grundmann et al. showed that endothelial colony-forming cells expressed mRNA of VEGF and pro-matrix metalloproteinase (pro-MMP) activity after treatment with physiological concentrations of 1,25(OH)2D in vitro (Table 1) [125]. Additionally, Schröder-Heurich et al. demonstrated that 1,25(OH)2D increased endothelial progenitor adhesion by alleviating the inflammatory signals of TNF-α in vitro (Table 1) [126].
Yu et al. found, in vitro, that physiological concentrations of 1,25(OH)2D altered the RNA expression profile of EPCs treated with high glucose [127]. Differentially expressed RNA influenced the activity of MMP and guanosine-5’-triphosphatase, which are related to EPC migration. These in vitro studies demonstrated that 1,25(OD)2D supplementation at physiological concentrations improved the adhesion of the EPCs in the injured endothelium and stimulated the migration of EPCs from the bone marrow.
Schröder-Heurich et al. also demonstrated that the adequate vitamin D supplement promoted the formation of VE-cadherin adhesion junctions on the EPCs. In this manner, the endothelial barrier integrity pretreated with TNF-α was repaired. Xu et al. also demonstrated that, in vitro, vitamin D alleviated EPC injuries, which were treated with Ang II by modulating the PPAR-γ/HO-1 pathway. The angiogenesis impaired by Ang II would be restored after vitamin D was supplied at cellular level (Table 1) [128]. At the same time, the study from Hammer et al. provided evidence that the calcitriol supplement improved EPCs viability in vitro (Table 1) [129]. These in vitro studies showed the possible therapeutic effect of the vitamin D on EPC migration and adhesion as well as the enhancement of the endothelial integrities under the circumstances involving vascular injury.

4.2. The Role of Vitamin D and MSCs/Pericytes in Vascular Calcification

Beyond the ability to differentiate osteoblasts, adipocytes, and chondroblasts, MSCs demonstrated anti-inflammatory and immune regulation functions [130,131]. An in vivo study initiated by Kramann et al. showed that the osteoblast-like character was initiated under specific circumstances, such as uremia [132]. The inflammatory cytokines released from the injured aorta, such as TGF-β1, mobilized MSC migration for neointimal formation [133]. However, Wang et al. provided in vitro evidence that the conditioned medium from MSCs retarded the VSMC osteoblastic change by blocking the bone morphogenetic protein (BMP) signaling and decreasing inflammatory cytokines in vitro [134,135]. Based on the in vitro evidence above, MSCs might provide the protective role in a paracrine manner to influence the calcification process, including anti-inflammatory effects, blocking the BMP2-Smad1/5/8 signal, downregulating the Wnt signal within VSMC, or attenuating the apoptosis of VSMC (Figure 2) [134,135,136,137].
Vitamin D deficiency was related to adventitial inflammation in clinical studies. Oma et al. noticed that the vitamin D concentration was inversely correlated with the monocyte infiltration within the adventitial layer in patients with CAD and inflammatory rheumatic disease [138]. Additionally, the vitamin D associated gene expression within aortic tissue might be influenced in patients with rheumatoid arthritis. Paraoxonases 2, which had antioxidative properties during atherosclerotic processes, was regulated by vitamin D. The expression was lessened during the inflammation [139]. Vitamin D was associated with lessening the inflammatory cytokine.
From the in vitro study initiated by Wang et al., the culture medium of MSCs decreased the calcium deposition in the VSMC because of the decreased expression of TNF-α, IL-1β, and IL-6 (Figure 2) [134]. Wasniks et al. also noticed that vitamin D decreased the TNF-α, and IL-6 secretions within osteocytes by suppressing M1 macrophages and influencing the osteogenic expression of MSCs [140]. Vitamin D had several roles in reducing the IL-1β-stimulated inflammatory profile in the adipocyte tissue, and such characteristics might be applied in lessening the calcification in MSCs in vitro [141].
A low vitamin D diet was observed to induce vascular calcification through the activation of BMP2 within the VSMC [142]. Fu et al. found that 1,25(OH)2D suppressed BMP2 activity in the bone marrow MSCs by binding the BMP2 promoting region [143]. Goltzman et al. also provided in vivo evidence that the vitamin D that originated from the osteocyte directly decreased the BMP2 release into serum and then mitigated the extraskeletal calcification [144].
Human marrow-derived MSCs (marrow stromal cells, hMSCs) give rise to osteoblasts, and their differentiation is stimulated by 1α,25(OH)2D, although hMSCs can also synthesize 1α,25(OH)2D. CKD reduces 1α,25(OH)2D production in kidneys and human MSCs [112]. Indeed, the vitamin D metabolism in hMSCs is regulated, as it is in the kidneys, and this promotes osteoblastogenesis in an autocrine/paracrine manner. CKD is associated with elevated circulating fibroblast growth factor 23 (FGF23). In vitro, rhFGF23 counters vitamin D-stimulated osteoblast differentiation of hMSCs by reducing the vitamin D receptor, CYP27B1/1α-hydroxylase, biosynthesis of 1α,25(OH)2D3, and signaling through BMP-7. Thus, the dysregulated vitamin D metabolism in hMSCs may contribute to impaired osteoblastogenesis and altered mineral metabolism in CKD subjects [113].
MSCs have the ability to reduce the VSMC calcification through down-regulating the Wnt signaling pathways. Guan et al. found that the culture medium from MSCs decreased the VSMC osteogenic differentiation by lowering Wnt 5a (Figure 2) [145]. Vitamin D regulated the expression of Wnt 5a in other systems, such as the respiratory tract [146]; therefore, it might provide a conjunctive role in decreasing vascular calcification.
From the evidence mentioned above, the adventitial MSCs carrying Gli-1 differentiated into osteoblast-like cells in the medial layer. However, the role of vitamin D on the MSCs in the adventitial layer is still under investigation. Recently, Hegner et al. noticed that the expression of the mammalian target of rapamycin (mTOR) influenced the calcification of MSCs in vitro [147]. They found that the activation of mammalian target of rapamycin complex 1 (mTORC1) was associated with the calcification of MSCs. When inhibiting mTORC1 by rapamycin, the mammalian target of rapamycin complex 2 (mTORC2) activity increased with a lessening of the calcification in MSCs. Vitamin D inhibited the mTORC1 activity through the inhibition of the tuberous sclerosis protein complex [148]. From this aspect, vitamin D might modulate the MSCs within the adventitia directly or influence the microenvironment.

4.3. The Role of Vitamin D in Adipose Tissue-Derived Stem Cells

The previous sections revealed that adipose tissue-derived MSCs have multipotency for differentiation into chondrocytes or smooth muscle cells. Adipose tissue-derived MSCs have vitamin D receptors within the nucleus, and the supplementation of the active form of vitamin D stimulated CYP24A1 activity and reduced 1,25(OH)2D expression within MSCs. However, the supplementation of the 25(OH)D increased intracellular active vitamin D production [149]. Thus, adipose tissue-derived MSCs can be modulated by vitamin D, especially nutritional vitamin D (e.g., cholecalciferol). From the study of Pesarini et al., vitamin D decreased the viability in time- and dose-dependent manners on the adipose tissue-derived MSCs and decreased the further adipose tissue formation [150].
Vitamin D induced the adipocyte stem cell osteogenic changes through activating bone morphogenetic protein 2 (BMP2) signaling [151]. At the same time, the supplementation of vitamin D modulated the chemokine-mediated inflammation induced by adipose tissue [152]. The vitamin D supplement might modulate the miR expression in the adipose tissue. Karkeni et al. also provided evidence that vitamin D lowered NF-κB signaling by alleviating the expression of miR 146a and miR-150 [153]. Thus, vitamin D decreased the adipocyte formation from stem cells by inducing apoptosis and modulating the inflammatory cytokine release within the adipocyte.
In addition to MSC migration, vitamin D may influence the differentiation of MSCs into adipocytes. MSCs within the bone marrow are the molecular switch between the osteoblastogenic and the adipocytic transformation. Several pathways, such as C/EBP-γ, C/EBP-α, and peroxisome proliferator-activated receptor-γ pathways, regulate MSC differentiation [154]. Vitamin D contributes to bone formation by activating the Wnt/β-catenin pathway. Lu et al. showed that active vitamin D induced bone formation by increasing the secretion of Wnt 10b by osteoclasts [155]. Therefore, vitamin D may play an adjunctive role in alleviating adipocyte transformation in MSCs and reducing the inflammation associated with vascular calcification.
The aforementioned evidence reveals that vitamin D may play a substantial role in modulating the therapeutic effect of MSCs in the treatment of vascular calcification.

5. Conclusions

Vascular calcification involves the deposition of calcifying particles within the endothelial and the medial layers after vascular damage. Recent reports on the MSCs lying within the adventitial layer demonstrated their role in developing vascular calcification. Therefore, the possible role of progenitor cells originating from bone marrow and soft tissue should be emphasized. Vitamin D deficiency is an important factor contributing to vascular calcification. Supplementation of vitamin D might modulate the calcification by modulating the MGP carboxylation. On the other hand, vitamin D might influence the phenotype of EPCs, hematopoietic progenitor cells, and MSCs. Vitamin D may be targeted along with MSCs in the treatment of vascular calcification.

Author Contributions

Study design: K.-C.L., C.-M.Z., R.-M.C., Y.-F.L.; Literature survey: Y.-C.H., W.-C.L., T.-H.Y., and C.-T.C.; Article drafting: Y.-C.H., C.-L.L., T.-H.Y., W.-C.L., C.-T.C., and K.-C.L.; All authors approved the final version of the manuscript.

Funding

The current study has been funded by Cardinal Tien Hospital (CTH108B-2A37).

Acknowledgments

This manuscript was edited by Wallace Academic Editing.

Conflicts of Interest

The authors have no relevant financial or non-financial competing interests to declare in relation to this manuscript.

Abbreviation

αSMAα-smooth muscle actin
Angangiotensin
BMPbone morphogenetic protein
BMP2bone morphogenetic protein2
C/EBPCCAAT/enhancer binding protein
CADcoronary artery disease
CBFA1core-binding factor α-1
CDcluster of differentiation
Cdk-2Cyclin-dependent kinase 2
CKDchronic kidney disease
CXCL1Chemokine (C-X-C motif) ligand 1
CXCL12Chemokine (C-X-C motif) ligand 12
CYP24A1Cytochrome P450 family 24 subfamily A member 1
EPCsendothelial progenitor cells
FGF23Fibroblast growth factor 23
Gli1The Human Glioma-Associated Oncogene Homolog 1
HIF-1-alphahypoxia-inducible factor-1-alpha
IL-1interleukin 1
IL-6Interleukin-6
KDRkinase insert domain receptor
LDLlow-density lipoprotein
M1 macrophageclassically activated macrophage
MAPKmitogen-activated protein kinase
MGPmatrix Gla protein
miRMicroRNA
MMPmatrix metalloproteinase
MSCsmesenchymal stem cells
mTORmammalian target of rapamycin
mTORC1mechanistic target of Rapamycin complex 1
mTORC2mechanistic target of Rapamycin complex 2
MVMatrix vesicle
NF-kBnuclear factor kappa-light-chain-enhancer of activated B
NG-2Neural/glial antigen 2
PDGFRPlatelet-derived growth factor receptors
PDGFRβPlatelet-derived growth factor receptor beta
PPAR-ɣperoxisome proliferator-activated receptor gamma
RAASrenin-angiotensin-aldosterone system
Sca-1stem cell antigen 1
SGK-1serum- and glucocorticoid-inducible kinase 1
Smad 1/5/8Mothers against decapentaplegic homolog 1/5/8
TNF-αtumor necrosis factor alpha
UVBultraviolet B
VEGFvascular endothelial growth factor
VSMCvascular smooth muscle cell
WntWingless-INT

References

  1. Nakahara, T.; Dweck, M.R.; Narula, N.; Pisapia, D.; Narula, J.; Strauss, H.W. Coronary Artery Calcification: From Mechanism to Molecular Imaging. JACC Cardiovasc. Imaging 2017, 10, 582–593. [Google Scholar] [CrossRef]
  2. Narula, N.; Dannenberg, A.J.; Olin, J.W.; Bhatt, D.L.; Johnson, K.W.; Nadkarni, G.; Min, J.; Torii, S.; Poojary, P.; Anand, S.S.; et al. Pathology of Peripheral Artery Disease in Patients with Critical Limb Ischemia. J. Am. Coll. Cardiol. 2018, 72, 2152–2163. [Google Scholar] [CrossRef]
  3. Mizuiri, S.; Nishizawa, Y.; Yamashita, K.; Mizuno, K.; Ishine, M.; Doi, S.; Masaki, T.; Shigemoto, K. Coronary artery calcification score and common iliac artery calcification score in non-dialysis CKD patients. Nephrology 2018, 23, 837–845. [Google Scholar] [CrossRef] [Green Version]
  4. Mathew, R.O.; Bangalore, S.; Lavelle, M.P.; Pellikka, P.A.; Sidhu, M.S.; Boden, W.E.; Asif, A. Diagnosis and management of atherosclerotic cardiovascular disease in chronic kidney disease: A review. Kidney Int. 2017, 91, 797–807. [Google Scholar] [CrossRef]
  5. Lin, J.S.; Evans, C.V.; Johnson, E.; Redmond, N.; Coppola, E.L.; Smith, N. Nontraditional Risk Factors in Cardiovascular Disease Risk Assessment: Updated Evidence Report and Systematic Review for the US Preventive Services Task Force. Jama 2018, 320, 281–297. [Google Scholar] [CrossRef] [Green Version]
  6. Hou, Y.-C.; Lu, C.-L.; Zheng, C.-M.; Chen, R.-M.; Lin, Y.-F.; Liu, W.-C.; Yen, T.-H.; Chen, R.; Lu, K.-C. Emerging Role of Vitamins D and K in Modulating Uremic Vascular Calcification: The Aspect of Passive Calcification. Nutrients 2019, 11, 152. [Google Scholar] [CrossRef] [Green Version]
  7. Gorriz, J.L.; Molina, P.; Cerveron, M.J.; Vila, R.; Bover, J.; Nieto, J.; Barril, G.; Martinez-Castelao, A.; Fernandez, E.; Escudero, V.; et al. Vascular calcification in patients with nondialysis CKD over 3 years. Clin. J. Am. Soc. Nephrol. Cjasn 2015, 10, 654–666. [Google Scholar] [CrossRef] [Green Version]
  8. Yap, Y.S.; Ting, K.T.; Chi, W.C.; Lin, C.H.; Liu, Y.C.; Chuang, W.L. Aortic Arch Calcification Predicts Patency Loss of Arteriovenous Fistula in End-Stage Renal Disease Patients. Sci. Rep. 2016, 6, 24943. [Google Scholar] [CrossRef]
  9. Crisan, M.; Yap, S.; Casteilla, L.; Chen, C.W.; Corselli, M.; Park, T.S.; Andriolo, G.; Sun, B.; Zheng, B.; Zhang, L.; et al. A perivascular origin for mesenchymal stem cells in multiple human organs. Cell Stem Cell 2008, 3, 301–313. [Google Scholar] [CrossRef] [Green Version]
  10. Kramann, R.; Goettsch, C.; Wongboonsin, J.; Iwata, H.; Schneider, R.K.; Kuppe, C.; Kaesler, N.; Chang-Panesso, M.; Machado, F.G.; Gratwohl, S.; et al. Adventitial MSC-like Cells Are Progenitors of Vascular Smooth Muscle Cells and Drive Vascular Calcification in Chronic Kidney Disease. Cell Stem Cell 2016, 19, 628–642. [Google Scholar] [CrossRef] [Green Version]
  11. Hou, Y.C.; Liu, W.C. Role of Vitamin D in Uremic Vascular Calcification. BioMed Res. 2017, 2017, 2803579. [Google Scholar] [CrossRef]
  12. Sun, R.; Huang, J.; Sun, B. Mobilization of endothelial progenitor cells in sepsis. Inflamm. Res. Off. J. Eur. Histamine Res. Soc. 2020, 69, 1–9. [Google Scholar] [CrossRef]
  13. Wu, C.C.; Hung, S.C.; Kuo, K.L.; Tarng, D.C. Impact of Indoxyl Sulfate on Progenitor Cell-Related Neovascularization of Peripheral Arterial Disease and Post-Angioplasty Thrombosis of Dialysis Vascular Access. Toxins 2017, 9, 25. [Google Scholar] [CrossRef]
  14. Yang, S.-W.; Hennessy, R.R.; Khosla, S.; Lennon, R.; Loeffler, D.; Sun, T.; Liu, Z.; Park, K.-H.; Wang, F.-L.; Lerman, L.O.; et al. Circulating osteogenic endothelial progenitor cell counts: New biomarker for the severity of coronary artery disease. Int. J. Cardiol. 2017, 227, 833–839. [Google Scholar] [CrossRef]
  15. Cianciolo, G.; Capelli, I.; Cappuccilli, M.; Scrivo, A.; Donadei, C.; Marchetti, A.; Rucci, P.; La Manna, G. Is chronic kidney disease-mineral and bone disorder associated with the presence of endothelial progenitor cells with a calcifying phenotype? Clin. Kidney J. 2017, 10, 389–396. [Google Scholar] [CrossRef] [Green Version]
  16. Dong, Y.; Stallmann-Jorgensen, I.S.; Pollock, N.K.; Harris, R.A.; Keeton, D.; Huang, Y.; Li, K.; Bassali, R.; Guo, D.H.; Thomas, J.; et al. A 16-week randomized clinical trial of 2000 international units daily vitamin D3 supplementation in black youth: 25-hydroxyvitamin D, adiposity, and arterial stiffness. J. Clin. Endocrinol. Metab. 2010, 95, 4584–4591. [Google Scholar] [CrossRef] [Green Version]
  17. Kumar, V.; Yadav, A.K.; Lal, A.; Kumar, V.; Singhal, M.; Billot, L.; Gupta, K.L.; Banerjee, D.; Jha, V. A Randomized Trial of Vitamin D Supplementation on Vascular Function in CKD. J. Am. Soc. Nephrol. Jasn 2017, 28, 3100–3108. [Google Scholar] [CrossRef] [Green Version]
  18. Mallat, Z.; Tedgui, A. Apoptosis in the vasculature: Mechanisms and functional importance. Br. J. Pharmacol. 2000, 130, 947–962. [Google Scholar] [CrossRef] [Green Version]
  19. Tabas, I.; Garcia-Cardena, G.; Owens, G.K. Recent insights into the cellular biology of atherosclerosis. J. Cell Biol. 2015, 209, 13–22. [Google Scholar] [CrossRef]
  20. Aikawa, E.; Nahrendorf, M.; Figueiredo, J.L.; Swirski, F.K.; Shtatland, T.; Kohler, R.H.; Jaffer, F.A.; Aikawa, M.; Weissleder, R. Osteogenesis associates with inflammation in early-stage atherosclerosis evaluated by molecular imaging in vivo. Circulation 2007, 116, 2841–2850. [Google Scholar] [CrossRef] [Green Version]
  21. Montezano, A.C.; Nguyen Dinh Cat, A.; Rios, F.J.; Touyz, R.M. Angiotensin II and vascular injury. Curr. Hypertens. Rep. 2014, 16, 431. [Google Scholar] [CrossRef]
  22. Wang, G.; Jacquet, L.; Karamariti, E.; Xu, Q. Origin and differentiation of vascular smooth muscle cells. J Physiol. 2015, 593, 3013–3030. [Google Scholar] [CrossRef] [Green Version]
  23. Lopes, A.A.; Tong, L.; Thumma, J.; Li, Y.; Fuller, D.S.; Morgenstern, H.; Bommer, J.; Kerr, P.G.; Tentori, F.; Akiba, T.; et al. Phosphate binder use and mortality among hemodialysis patients in the Dialysis Outcomes and Practice Patterns Study (DOPPS): Evaluation of possible confounding by nutritional status. Am. J. Kidney Dis. 2012, 60, 90–101. [Google Scholar] [CrossRef] [Green Version]
  24. Jono, S.; McKee, M.D.; Murry, C.E.; Shioi, A.; Nishizawa, Y.; Mori, K.; Morii, H.; Giachelli, C.M. Phosphate regulation of vascular smooth muscle cell calcification. Circ. Res. 2000, 87, E10–E17. [Google Scholar] [CrossRef]
  25. Li, X.; Yang, H.Y.; Giachelli, C.M. Role of the sodium-dependent phosphate cotransporter, Pit-1, in vascular smooth muscle cell calcification. Circ. Res. 2006, 98, 905–912. [Google Scholar] [CrossRef] [Green Version]
  26. Lang, F.; Ritz, E.; Alesutan, I.; Voelkl, J. Impact of aldosterone on osteoinductive signaling and vascular calcification. Nephron. Physiol. 2014, 128, 40–45. [Google Scholar] [CrossRef]
  27. Schlieper, G.; Schurgers, L.; Brandenburg, V.; Reutelingsperger, C.; Floege, J. Vascular calcification in chronic kidney disease: An update. Nephrol. Dial. Transplant. Off. Publ. Eur. Dial. Transpl. Assoc. Eur. Ren. Assoc. 2016, 31, 31–39. [Google Scholar] [CrossRef] [Green Version]
  28. Martinez-Moreno, J.M.; Munoz-Castaneda, J.R.; Herencia, C.; Oca, A.M.; Estepa, J.C.; Canalejo, R.; Rodriguez-Ortiz, M.E.; Perez-Martinez, P.; Aguilera-Tejero, E.; Canalejo, A.; et al. In vascular smooth muscle cells paricalcitol prevents phosphate-induced Wnt/beta-catenin activation. Am. J. Physiol. Ren. Physiol. 2012, 303, F1136–F1144. [Google Scholar] [CrossRef] [Green Version]
  29. Shao, J.-S.; Cheng, S.-L.; Pingsterhaus, J.M.; Charlton-Kachigian, N.; Loewy, A.P.; Towler, D.A. Msx2 promotes cardiovascular calcification by activating paracrine Wnt signals. J. Clin. Investig. 2005, 115, 1210–1220. [Google Scholar] [CrossRef] [Green Version]
  30. Voelkl, J.; Luong, T.T.; Tuffaha, R.; Musculus, K.; Auer, T.; Lian, X.; Daniel, C.; Zickler, D.; Boehme, B.; Sacherer, M.; et al. SGK1 induces vascular smooth muscle cell calcification through NF-kappaB signaling. J. Clin. Investig. 2018, 128, 3024–3040. [Google Scholar] [CrossRef]
  31. Voelkl, J.; Lang, F.; Eckardt, K.-U.; Amann, K.; Kuro-O, M.; Pasch, A.; Pieske, B.; Alesutan, I. Signaling pathways involved in vascular smooth muscle cell calcification during hyperphosphatemia. Cell. Mol. Life Sci. Cmls 2019, 76, 2077–2091. [Google Scholar] [CrossRef] [Green Version]
  32. Kapustin, A.N.; Chatrou, M.L.; Drozdov, I.; Zheng, Y.; Davidson, S.M.; Soong, D.; Furmanik, M.; Sanchis, P.; De Rosales, R.T.; Alvarez-Hernandez, D.; et al. Vascular smooth muscle cell calcification is mediated by regulated exosome secretion. Circ. Res. 2015, 116, 1312–1323. [Google Scholar] [CrossRef] [Green Version]
  33. Reynolds, J.L.; Joannides, A.J.; Skepper, J.N.; McNair, R.; Schurgers, L.J.; Proudfoot, D.; Jahnen-Dechent, W.; Weissberg, P.L.; Shanahan, C.M. 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. 2004, 15, 2857–2867. [Google Scholar] [CrossRef] [Green Version]
  34. Shroff, R.C.; McNair, R.; Skepper, J.N.; Figg, N.; Schurgers, L.J.; Deanfield, J.; Rees, L.; Shanahan, C.M. Chronic Mineral Dysregulation Promotes Vascular Smooth Muscle Cell Adaptation and Extracellular Matrix Calcification. J. Am. Soc. Nephrol. 2010, 21, 103–112. [Google Scholar] [CrossRef]
  35. Moochhala, S.H. Extracellular pyrophosphate in the kidney: How does it get there and what does it do? Nephron. Physiol. 2012, 120, p33–p38. [Google Scholar] [CrossRef]
  36. Nigwekar, S.U.; Thadhani, R.; Brandenburg, V.M. Calciphylaxis. New Engl. J. Med. 2018, 378, 1704–1714. [Google Scholar] [CrossRef]
  37. Opdebeeck, B.; Maudsley, S.; Azmi, A.; De Maré, A.; De Leger, W.; Meijers, B.; Verhulst, A.; Evenepoel, P.; D’Haese, P.C.; Neven, E. Indoxyl Sulfate and p-Cresyl Sulfate Promote Vascular Calcification and Associate with Glucose Intolerance. J. Am. Soc. Nephrol. 2019, 30, 751. [Google Scholar] [CrossRef]
  38. Adijiang, A.; Goto, S.; Uramoto, S.; Nishijima, F.; Niwa, T. Indoxyl sulphate promotes aortic calcification with expression of osteoblast-specific proteins in hypertensive rats. Nephrol. Dial. Transpl. 2008, 23, 1892–1901. [Google Scholar] [CrossRef] [Green Version]
  39. Valcheva, P.; Cardus, A.; Panizo, S.; Parisi, E.; Bozic, M.; Lopez Novoa, J.M.; Dusso, A.; Fernandez, E.; Valdivielso, J.M. Lack of vitamin D receptor causes stress-induced premature senescence in vascular smooth muscle cells through enhanced local angiotensin-II signals. Atherosclerosis 2014, 235, 247–255. [Google Scholar] [CrossRef]
  40. Aoshima, Y.; Mizobuchi, M.; Ogata, H.; Kumata, C.; Nakazawa, A.; Kondo, F.; Ono, N.; Koiwa, F.; Kinugasa, E.; Akizawa, T. Vitamin D receptor activators inhibit vascular smooth muscle cell mineralization induced by phosphate and TNF-alpha. Nephrol. Dial. Transplant. Off. Publ. Eur. Dial. Transpl. Assoc. Eur. Ren. Assoc. 2012, 27, 1800–1806. [Google Scholar] [CrossRef] [Green Version]
  41. Chen, S.; Law, C.S.; Gardner, D.G. Vitamin D-dependent suppression of endothelin-induced vascular smooth muscle cell proliferation through inhibition of CDK2 activity. J. Steroid Biochem. Mol. Biol. 2010, 118, 135–141. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Jono, S.; Nishizawa, Y.; Shioi, A.; Morii, H. 1,25-Dihydroxyvitamin D3 increases in vitro vascular calcification by modulating secretion of endogenous parathyroid hormone-related peptide. Circulation 1998, 98, 1302–1306. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Cardus, A.; Panizo, S.; Parisi, E.; Fernandez, E.; Valdivielso, J.M. Differential effects of vitamin D analogs on vascular calcification. J. Bone Miner. Res. Off. J. Am. Soc. Bone Miner. Res. 2007, 22, 860–866. [Google Scholar] [CrossRef] [PubMed]
  44. Bhat, O.M.; Yuan, X.; Camus, S.; Salloum, F.N.; Li, P.L. Abnormal Lysosomal Positioning and Small Extracellular Vesicle Secretion in Arterial Stiffening and Calcification of Mice Lacking Mucolipin 1 Gene. Int. J. Mol. Sci. 2020, 21, 1713. [Google Scholar] [CrossRef] [Green Version]
  45. Carmo, L.S.; Burdmann, E.A.; Fessel, M.R.; Almeida, Y.E.; Pescatore, L.A.; Farias-Silva, E.; Gamarra, L.F.; Lopes, G.H.; Aloia, T.P.A.; Liberman, M. Expansive Vascular Remodeling and Increased Vascular Calcification Response to Cholecalciferol in a Murine Model of Obesity and Insulin Resistance. Arterioscler. Thromb. Vasc. Biol. 2019, 39, 200–211. [Google Scholar] [CrossRef]
  46. Corselli, M.; Chen, C.W.; Sun, B.; Yap, S.; Rubin, J.P.; Peault, B. The tunica adventitia of human arteries and veins as a source of mesenchymal stem cells. Stem Cells Dev. 2012, 21, 1299–1308. [Google Scholar] [CrossRef] [Green Version]
  47. Yang, W.J.; Zheng, L.; Wu, X.H.; Huang, Z.Q.; Niu, C.B.; Zhao, H.L.; Leung, T.W.; Wong, L.K.; Chen, X.Y. Postmortem Study Exploring Distribution and Patterns of Intracranial Artery Calcification. Stroke 2018, 49, 2767–2769. [Google Scholar] [CrossRef]
  48. Zheng, L.; Yang, W.J.; Niu, C.B.; Zhao, H.L.; Wong, K.S.; Leung, T.W.H.; Chen, X.Y. Correlation of Adventitial Vasa Vasorum with Intracranial Atherosclerosis: A Postmortem Study. J. Stroke 2018, 20, 342–349. [Google Scholar] [CrossRef] [Green Version]
  49. Ijaz, T.; Sun, H.; Pinchuk, I.V.; Milewicz, D.M.; Tilton, R.G.; Brasier, A.R. Deletion of NF-κB/RelA in Angiotensin II-Sensitive Mesenchymal Cells Blocks Aortic Vascular Inflammation and Abdominal Aortic Aneurysm Formation. Arterioscler. Thromb. Vasc. Biol. 2017, 37, 1881–1890. [Google Scholar] [CrossRef] [Green Version]
  50. Tang, J.; Wang, H.; Huang, X.; Li, F.; Zhu, H.; Li, Y.; He, L.; Zhang, H.; Pu, W.; Liu, K.; et al. Arterial Sca1(+) Vascular Stem Cells Generate De Novo Smooth Muscle for Artery Repair and Regeneration. Cell Stem Cell 2020, 26, 81–96. [Google Scholar] [CrossRef]
  51. Tinajero, M.G.; Gotlieb, A.I. Recent Developments in Vascular Adventitial Pathobiology: The Dynamic Adventitia as a Complex Regulator of Vascular Disease. Am. J. Pathol. 2019. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Del Toro, R.; Chevre, R.; Rodriguez, C.; Ordonez, A.; Martinez-Gonzalez, J.; Andres, V.; Mendez-Ferrer, S. Nestin(+) cells direct inflammatory cell migration in atherosclerosis. Nat. Commun. 2016, 7, 12706. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Ding, L.; Vezzani, B.; Khan, N.; Su, J.; Xu, L.; Yan, G.; Liu, Y.; Li, R.; Gaur, A.; Diao, Z.; et al. CD10 expression identifies a subset of human perivascular progenitor cells with high proliferation and calcification potentials. Stem Cells 2019. [Google Scholar] [CrossRef]
  54. Tigges, U.; Komatsu, M.; Stallcup, W.B. Adventitial pericyte progenitor/mesenchymal stem cells participate in the restenotic response to arterial injury. J. Vasc. Res. 2013, 50, 134–144. [Google Scholar] [CrossRef] [Green Version]
  55. Folestad, E.; Kunath, A.; Wagsater, D. PDGF-C and PDGF-D signaling in vascular diseases and animal models. Mol. Asp. Med. 2018, 62, 1–11. [Google Scholar] [CrossRef] [PubMed]
  56. Sohnel, O.; Grases, F. Supersaturation of body fluids, plasma and urine, with respect to biological hydroxyapatite. Urol. Res. 2011, 39, 429–436. [Google Scholar] [CrossRef] [PubMed]
  57. Cannata-Andia, J.B.; Roman-Garcia, P.; Hruska, K. The connections between vascular calcification and bone health. Nephrol. Dial. Transplant. Off. Publ. Eur. Dial. Transpl. Assoc. Eur. Ren. Assoc. 2011, 26, 3429–3436. [Google Scholar] [CrossRef] [PubMed]
  58. Drouet, C. Apatite formation: Why it may not work as planned, and how to conclusively identify apatite compounds. Biomed Res. Int. 2013, 2013, 490946. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Jansen, F.; Li, Q.; Pfeifer, A.; Werner, N. Endothelial- and Immune Cell-Derived Extracellular Vesicles in the Regulation of Cardiovascular Health and Disease. JACC Basic Transl. Sci. 2017, 2, 790–807. [Google Scholar] [CrossRef]
  60. Gui, T.; Zhou, G.; Sun, Y.; Shimokado, A.; Itoh, S.; Oikawa, K.; Muragaki, Y. MicroRNAs that target Ca(2+) transporters are involved in vascular smooth muscle cell calcification. Lab. Investig. A J. Tech. Methods Pathol. 2012, 92, 1250–1259. [Google Scholar] [CrossRef] [Green Version]
  61. Liu, J.; Xiao, X.; Shen, Y.; Chen, L.; Xu, C.; Zhao, H.; Wu, Y.; Zhang, Q.; Zhong, J.; Tang, Z.; et al. MicroRNA-32 promotes calcification in vascular smooth muscle cells: Implications as a novel marker for coronary artery calcification. PLoS ONE 2017, 12, e0174138. [Google Scholar] [CrossRef]
  62. Zhang, C.; Zhang, K.; Huang, F.; Feng, W.; Chen, J.; Zhang, H.; Wang, J.; Luo, P.; Huang, H. Exosomes, the message transporters in vascular calcification. J. Cell. Mol. Med. 2018, 22, 4024–4033. [Google Scholar] [CrossRef]
  63. Herrmann, M.; Schafer, C.; Heiss, A.; Graber, S.; Kinkeldey, A.; Buscher, A.; Schmitt, M.M.; Bornemann, J.; Nimmerjahn, F.; Herrmann, M.; et al. Clearance of fetuin-A--containing calciprotein particles is mediated by scavenger receptor-A. Circ. Res. 2012, 111, 575–584. [Google Scholar] [CrossRef] [PubMed]
  64. Jahnen-Dechent, W.; Heiss, A.; Schafer, C.; Ketteler, M. Fetuin-A regulation of calcified matrix metabolism. Circ. Res. 2011, 108, 1494–1509. [Google Scholar] [CrossRef] [PubMed]
  65. Pasch, A.; Block, G.A.; Bachtler, M.; Smith, E.R.; Jahnen-Dechent, W.; Arampatzis, S.; Chertow, G.M.; Parfrey, P.; Ma, X.; Floege, J. Blood Calcification Propensity, Cardiovascular Events, and Survival in Patients Receiving Hemodialysis in the EVOLVE Trial. Clin. J. Am. Soc. Nephrol. Cjasn 2017, 12, 315–322. [Google Scholar] [CrossRef] [Green Version]
  66. Chen, W.; Anokhina, V.; Dieudonne, G.; Abramowitz, M.K.; Kashyap, R.; Yan, C.; Wu, T.T.; de Mesy Bentley, K.L.; Miller, B.L.; Bushinsky, D.A. Patients with advanced chronic kidney disease and vascular calcification have a large hydrodynamic radius of secondary calciprotein particles. Nephrol. Dial. Transplant. Off. Publ. Eur. Dial. Transpl. Assoc. Eur. Ren. Assoc. 2019, 34, 992–1000. [Google Scholar] [CrossRef] [PubMed]
  67. Mazzaferro, S.; Tartaglione, L.; Rotondi, S.; Bover, J.; Goldsmith, D.; Pasquali, M. News on biomarkers in CKD-MBD. Semin. Nephrol. 2014, 34, 598–611. [Google Scholar] [CrossRef]
  68. Shearer, M.J.; Okano, T. Key Pathways and Regulators of Vitamin K Function and Intermediary Metabolism. Annu. Rev. Nutr. 2018, 38, 127–151. [Google Scholar] [CrossRef]
  69. Wuyts, J.; Dhondt, A. The role of vitamin K in vascular calcification of patients with chronic kidney disease. Acta Clin. Belg. 2016, 71, 462–467. [Google Scholar] [CrossRef]
  70. Wen, L.; Chen, J.; Duan, L.; Li, S. Vitamin Kdependent proteins involved in bone and cardiovascular health (Review). Mol. Med. Rep. 2018, 18, 3–15. [Google Scholar] [CrossRef] [Green Version]
  71. Viegas, C.S.B.; Santos, L.; Macedo, A.L.; Matos, A.A.; Silva, A.P.; Neves, P.L.; Staes, A.; Gevaert, K.; Morais, R.; Vermeer, C.; et al. Chronic Kidney Disease Circulating Calciprotein Particles and Extracellular Vesicles Promote Vascular Calcification: A Role for GRP (Gla-Rich Protein). Arterioscler. Thromb. Vasc. Biol. 2018, 38, 575–587. [Google Scholar] [CrossRef] [Green Version]
  72. Silaghi, C.N.; Ilyes, T.; Filip, V.P.; Farcas, M.; van Ballegooijen, A.J.; Craciun, A.M. Vitamin K Dependent Proteins in Kidney Disease. Int. J. Mol. Sci. 2019, 20, 1571. [Google Scholar] [CrossRef] [Green Version]
  73. Poon, C.C.; Li, R.W.; Seto, S.W.; Kong, S.K.; Ho, H.P.; Hoi, M.P.; Lee, S.M.; Ngai, S.M.; Chan, S.W.; Leung, G.P.; et al. In vitro vitamin K(2) and 1alpha,25-dihydroxyvitamin D(3) combination enhances osteoblasts anabolism of diabetic mice. Eur. J. Pharmacol. 2015, 767, 30–40. [Google Scholar] [CrossRef]
  74. O’Connor, E.; Molgaard, C.; Michaelsen, K.F.; Jakobsen, J.; Lamberg-Allardt, C.J.; Cashman, K.D. Serum percentage undercarboxylated osteocalcin, a sensitive measure of vitamin K status, and its relationship to bone health indices in Danish girls. Br. J. Nutr. 2007, 97, 661–666. [Google Scholar] [CrossRef] [Green Version]
  75. Miyake, N.; Hoshi, K.; Sano, Y.; Kikuchi, K.; Tadano, K.; Koshihara, Y. 1,25-Dihydroxyvitamin D3 promotes vitamin K2 metabolism in human osteoblasts. Osteoporos. Int. A J. Establ. Result Coop. Between Eur. Found. Osteoporos. Natl. Osteoporos. Found. USA 2001, 12, 680–687. [Google Scholar] [CrossRef]
  76. Briasoulis, A.; Tousoulis, D.; Antoniades, C.; Papageorgiou, N.; Stefanadis, C. The role of endothelial progenitor cells in vascular repair after arterial injury and atherosclerotic plaque development. Cardiovasc. Ther. 2011, 29, 125–139. [Google Scholar] [CrossRef]
  77. Li, Y.; Sun, R.; Zou, J.; Ying, Y.; Luo, Z. Dual Roles of the AMP-Activated Protein Kinase Pathway in Angiogenesis. Cells 2019, 8, 752. [Google Scholar] [CrossRef] [Green Version]
  78. Schipani, E.; Wu, C.; Rankin, E.B.; Giaccia, A.J. Regulation of Bone Marrow Angiogenesis by Osteoblasts during Bone Development and Homeostasis. Front. Endocrinol. 2013, 4, 85. [Google Scholar] [CrossRef] [Green Version]
  79. Cianciolo, G.; La Manna, G.; Della Bella, E.; Cappuccilli, M.L.; Angelini, M.L.; Dormi, A.; Capelli, I.; Laterza, C.; Costa, R.; Alviano, F.; et al. Effect of vitamin D receptor activator therapy on vitamin D receptor and osteocalcin expression in circulating endothelial progenitor cells of hemodialysis patients. Blood Purif. 2013, 35, 187–195. [Google Scholar] [CrossRef]
  80. Lu, C.-L.; Leu, J.-G.; Liu, W.-C.; Zheng, C.-M.; Lin, Y.-F.; Shyu, J.-F.; Wu, C.-C.; Lu, K.-C. Endothelial Progenitor Cells Predict Long-Term Mortality in Hemodialysis Patients. Int. J. Med. Sci. 2016, 13, 240–247. [Google Scholar] [CrossRef] [Green Version]
  81. Bahlmann, F.H.; Speer, T.; Fliser, D. Endothelial progenitor cells in chronic kidney disease. Nephrol. Dial. Transplant. 2009, 25, 341–346. [Google Scholar] [CrossRef] [Green Version]
  82. Dutta, P.; Courties, G.; Wei, Y.; Leuschner, F.; Gorbatov, R.; Robbins, C.S.; Iwamoto, Y.; Thompson, B.; Carlson, A.L.; Heidt, T.; et al. Myocardial infarction accelerates atherosclerosis. Nature 2012, 487, 325–329. [Google Scholar] [CrossRef] [Green Version]
  83. Heidt, T.; Sager, H.B.; Courties, G.; Dutta, P.; Iwamoto, Y.; Zaltsman, A.; von Zur Muhlen, C.; Bode, C.; Fricchione, G.L.; Denninger, J.; et al. Chronic variable stress activates hematopoietic stem cells. Nat. Med. 2014, 20, 754–758. [Google Scholar] [CrossRef] [Green Version]
  84. Pufe, T.; Petersen, W.; Fandrich, F.; Varoga, D.; Wruck, C.J.; Mentlein, R.; Helfenstein, A.; Hoseas, D.; Dressel, S.; Tillmann, B.; et al. Programmable cells of monocytic origin (PCMO): A source of peripheral blood stem cells that generate collagen type II-producing chondrocytes. J. Orthop. Res. Off. Publ. Orthop. Res. Soc. 2008, 26, 304–313. [Google Scholar] [CrossRef]
  85. Doehring, L.C.; Heeger, C.; Aherrahrou, Z.; Kaczmarek, P.M.; Erdmann, J.; Schunkert, H.; Ehlers, E.M. Myeloid CD34+CD13+ precursor cells transdifferentiate into chondrocyte-like cells in atherosclerotic intimal calcification. Am. J. Pathol. 2010, 177, 473–480. [Google Scholar] [CrossRef]
  86. Cho, H.J.; Lee, J.W.; Cho, H.J.; Lee, C.S.; Kim, H.S. Identification of Adult Mesodermal Progenitor Cells and Hierarchy in Atherosclerotic Vascular Calcification. Stem Cells 2018, 36, 1075–1096. [Google Scholar] [CrossRef] [Green Version]
  87. Frodermann, V.; Rohde, D.; Courties, G.; Severe, N.; Schloss, M.J.; Amatullah, H.; McAlpine, C.S.; Cremer, S.; Hoyer, F.F.; Ji, F.; et al. Exercise reduces inflammatory cell production and cardiovascular inflammation via instruction of hematopoietic progenitor cells. Nat. Med. 2019, 25, 1761–1771. [Google Scholar] [CrossRef]
  88. Miranville, A.; Heeschen, C.; Sengenes, C.; Curat, C.A.; Busse, R.; Bouloumie, A. Improvement of postnatal neovascularization by human adipose tissue-derived stem cells. Circulation 2004, 110, 349–355. [Google Scholar] [CrossRef] [Green Version]
  89. Hu, Y.; Davison, F.; Ludewig, B.; Erdel, M.; Mayr, M.; Url, M.; Dietrich, H.; Xu, Q. Smooth muscle cells in transplant atherosclerotic lesions are originated from recipients, but not bone marrow progenitor cells. Circulation 2002, 106, 1834–1839. [Google Scholar] [CrossRef] [Green Version]
  90. Planat-Benard, V.; Silvestre, J.S.; Cousin, B.; Andre, M.; Nibbelink, M.; Tamarat, R.; Clergue, M.; Manneville, C.; Saillan-Barreau, C.; Duriez, M.; et al. Plasticity of human adipose lineage cells toward endothelial cells: Physiological and therapeutic perspectives. Circulation 2004, 109, 656–663. [Google Scholar] [CrossRef]
  91. Rani, S.; Ryan, A.E.; Griffin, M.D.; Ritter, T. Mesenchymal Stem Cell-derived Extracellular Vesicles: Toward Cell-free Therapeutic Applications. Mol. Ther. J. Am. Soc. Gene Ther. 2015, 23, 812–823. [Google Scholar] [CrossRef] [Green Version]
  92. Pan, B.T.; Johnstone, R.M. Fate of the transferrin receptor during maturation of sheep reticulocytes in vitro: Selective externalization of the receptor. Cell 1983, 33, 967–978. [Google Scholar] [CrossRef]
  93. Griffin, M.D.; Ryan, A.E.; Alagesan, S.; Lohan, P.; Treacy, O.; Ritter, T. Anti-donor immune responses elicited by allogeneic mesenchymal stem cells: What have we learned so far? Immunol. Cell Biol. 2013, 91, 40–51. [Google Scholar] [CrossRef]
  94. Wang, Y.; Ma, W.Q.; Zhu, Y.; Han, X.Q.; Liu, N. Exosomes Derived From Mesenchymal Stromal Cells Pretreated With Advanced Glycation End Product-Bovine Serum Albumin Inhibit Calcification of Vascular Smooth Muscle Cells. Front. Endocrinol. 2018, 9, 524. [Google Scholar] [CrossRef] [Green Version]
  95. Sahoo, S.; Klychko, E.; Thorne, T.; Misener, S.; Schultz, K.M.; Millay, M.; Ito, A.; Liu, T.; Kamide, C.; Agrawal, H.; et al. Exosomes from human CD34(+) stem cells mediate their proangiogenic paracrine activity. Circ. Res. 2011, 109, 724–728. [Google Scholar] [CrossRef] [Green Version]
  96. Guo, Y.; Bao, S.; Guo, W.; Diao, Z.; Wang, L.; Han, X.; Guo, W.; Liu, W. Bone marrow mesenchymal stem cell-derived exosomes alleviate high phosphorus-induced vascular smooth muscle cells calcification by modifying microRNA profiles. Funct. Integr. Genom. 2019, 19, 633–643. [Google Scholar] [CrossRef]
  97. Wei, Y.; Wu, Y.; Zhao, R.; Zhang, K.; Midgley, A.C.; Kong, D.; Li, Z.; Zhao, Q. MSC-derived sEVs enhance patency and inhibit calcification of synthetic vascular grafts by immunomodulation in a rat model of hyperlipidemia. Biomaterials 2019, 204, 13–24. [Google Scholar] [CrossRef]
  98. Mousavi, S.E.; Amini, H.; Heydarpour, P.; Amini Chermahini, F.; Godderis, L. Air pollution, environmental chemicals, and smoking may trigger vitamin D deficiency: Evidence and potential mechanisms. Environ. Int. 2019, 122, 67–90. [Google Scholar] [CrossRef]
  99. Holick, M.F. Vitamin D: A D-Lightful health perspective. Nutr. Rev. 2008, 66, S182–S194. [Google Scholar] [CrossRef]
  100. Gonzalez, E.A.; Sachdeva, A.; Oliver, D.A.; Martin, K.J. Vitamin D insufficiency and deficiency in chronic kidney disease. A single center observational study. Am. J. Nephrol. 2004, 24, 503–510. [Google Scholar] [CrossRef]
  101. LaClair, R.E.; Hellman, R.N.; Karp, S.L.; Kraus, M.; Ofner, S.; Li, Q.; Graves, K.L.; Moe, S.M. Prevalence of calcidiol deficiency in CKD: A cross-sectional study across latitudes in the United States. Am. J. Kidney Dis. 2005, 45, 1026–1033. [Google Scholar] [CrossRef]
  102. Banerjee, S.; Basu, S.; Sengupta, J. Vitamin D in nephrotic syndrome remission: A case-control study. Pediatric Nephrol. 2013, 28, 1983–1989. [Google Scholar] [CrossRef]
  103. Dusso, A.S.; Tokumoto, M. Defective renal maintenance of the vitamin D endocrine system impairs vitamin D renoprotection: A downward spiral in kidney disease. Kidney Int. 2011, 79, 715–729. [Google Scholar] [CrossRef] [Green Version]
  104. Yamaguchi, S.; Maruyama, T.; Wakino, S.; Tokuyama, H.; Hashiguchi, A.; Tada, S.; Homma, K.; Monkawa, T.; Thomas, J.; Miyashita, K.; et al. A case of severe osteomalacia caused by Tubulointerstitial nephritis with Fanconi syndrome in asymptomotic primary biliary cirrhosis. Bmc Nephrol. 2015, 16, 187. [Google Scholar] [CrossRef] [Green Version]
  105. Wang, L.; Gao, Z.; Wang, L.; Gao, Y. Upregulation of nuclear factor-kappaB activity mediates CYP24 expression and reactive oxygen species production in indoxyl sulfate-induced chronic kidney disease. Nephrology 2016, 21, 774–781. [Google Scholar] [CrossRef]
  106. Michaud, J.; Naud, J.; Ouimet, D.; Demers, C.; Petit, J.L.; Leblond, F.A.; Bonnardeaux, A.; Gascon-Barre, M.; Pichette, V. Reduced hepatic synthesis of calcidiol in uremia. J. Am. Soc. Nephrol. 2010, 21, 1488–1497. [Google Scholar] [CrossRef] [Green Version]
  107. Harinarayan, C.V. Vitamin D and diabetes mellitus. Hormones 2014, 13, 163–181. [Google Scholar] [CrossRef]
  108. Lu, X.; Hu, M.C. Klotho/FGF23 Axis in Chronic Kidney Disease and Cardiovascular Disease. Kidney Dis. 2017, 3, 15–23. [Google Scholar] [CrossRef] [Green Version]
  109. Beckman, M.J.; Tadikonda, P.; Werner, E.; Prahl, J.; Yamada, S.; DeLuca, H.F. Human 25-hydroxyvitamin D3-24-hydroxylase, a multicatalytic enzyme. Biochemistry 1996, 35, 8465–8472. [Google Scholar] [CrossRef]
  110. Shimada, T.; Yamazaki, Y.; Takahashi, M.; Hasegawa, H.; Urakawa, I.; Oshima, T.; Ono, K.; Kakitani, M.; Tomizuka, K.; Fujita, T.; et al. Vitamin D receptor-independent FGF23 actions in regulating phosphate and vitamin D metabolism. Am. J. Physiol. Ren. Physiol. 2005, 289, F1088–F1095. [Google Scholar] [CrossRef]
  111. Lou, Y.-R.; Toh, T.C.; Tee, Y.H.; Yu, H. 25-Hydroxyvitamin D3 induces osteogenic differentiation of human mesenchymal stem cells. Sci. Rep. 2017, 7, 42816. [Google Scholar] [CrossRef] [Green Version]
  112. Zhou, S.; Glowacki, J. Chronic kidney disease and vitamin D metabolism in human bone marrow-derived MSCs. Ann. New York Acad. Sci. 2017, 1402, 43–55. [Google Scholar] [CrossRef]
  113. Meng, F.; Bertucci, C.; Gao, Y.; Li, J.; Luu, S.; LeBoff, M.S.; Glowacki, J.; Zhou, S. Fibroblast growth factor 23 counters vitamin D metabolism and action in human mesenchymal stem cells. J. Steroid Biochem. Mol. Biol. 2020, 199, 105587. [Google Scholar] [CrossRef]
  114. Judd, S.E.; Tangpricha, V. Vitamin D deficiency and risk for cardiovascular disease. Am. J. Med. Sci. 2009, 338, 40–44. [Google Scholar] [CrossRef] [Green Version]
  115. Forman, J.P.; Giovannucci, E.; Holmes, M.D.; Bischoff-Ferrari, H.A.; Tworoger, S.S.; Willett, W.C.; Curhan, G.C. Plasma 25-hydroxyvitamin D levels and risk of incident hypertension. Hypertension 2007, 49, 1063–1069. [Google Scholar] [CrossRef] [Green Version]
  116. Kim, D.H.; Sabour, S.; Sagar, U.N.; Adams, S.; Whellan, D.J. Prevalence of hypovitaminosis D in cardiovascular diseases (from the National Health and Nutrition Examination Survey 2001 to 2004). Am. J. Cardiol. 2008, 102, 1540–1544. [Google Scholar] [CrossRef]
  117. Pilz, S.; Dobnig, H.; Fischer, J.E.; Wellnitz, B.; Seelhorst, U.; Boehm, B.O.; Marz, W. Low vitamin d levels predict stroke in patients referred to coronary angiography. Stroke 2008, 39, 2611–2613. [Google Scholar] [CrossRef] [Green Version]
  118. Melamed, M.L.; Muntner, P.; Michos, E.D.; Uribarri, J.; Weber, C.; Sharma, J.; Raggi, P. Serum 25-hydroxyvitamin D levels and the prevalence of peripheral arterial disease: Results from NHANES 2001 to 2004. Arterioscler. Thromb. Vasc. Biol. 2008, 28, 1179–1185. [Google Scholar] [CrossRef] [Green Version]
  119. Yaribeygi, H.; Maleki, M.; Sathyapalan, T.; Iranpanah, H.; Orafai, H.M.; Jamialahmadi, T.; Sahebkar, A. The molecular mechanisms by which vitamin D improve glucose homeostasis: A mechanistic review. Life Sci. 2020, 244, 117305. [Google Scholar] [CrossRef]
  120. Sypniewska, G.; Pollak, J.; Strozecki, P.; Camil, F.; Kretowicz, M.; Janikowski, G.; Mankowska-Cyl, A.; Pater, A.; Manitius, J. 25-hydroxyvitamin D, biomarkers of endothelial dysfunction and subclinical organ damage in adults with hypertension. Am. J. Hypertens. 2014, 27, 114–121. [Google Scholar] [CrossRef] [Green Version]
  121. Tedgui, A.; Mallat, Z. Cytokines in atherosclerosis: Pathogenic and regulatory pathways. Physiol. Rev. 2006, 86, 515–581. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Yuan, W.; Pan, W.; Kong, J.; Zheng, W.; Szeto, F.L.; Wong, K.E.; Cohen, R.; Klopot, A.; Zhang, Z.; Li, Y.C. 1,25-dihydroxyvitamin D3 suppresses renin gene transcription by blocking the activity of the cyclic AMP response element in the renin gene promoter. J. Biol. Chem. 2007, 282, 29821–29830. [Google Scholar] [CrossRef] [Green Version]
  123. Tiryaki, O.; Usalan, C.; Sayiner, Z.A. Vitamin D receptor activation with calcitriol for reducing urinary angiotensinogen in patients with type 2 diabetic chronic kidney disease. Ren. Fail. 2016, 38, 222–227. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Ai, S.; He, Z.; Ding, R.; Wu, F.; Huang, Z.; Wang, J.; Huang, S.; Dai, X.; Zhang, J.; Chen, J.; et al. Reduced Vitamin D Receptor on Circulating Endothelial Progenitor Cells: A New Risk Factor of Coronary Artery Diseases. J. Atheroscler. Thromb. 2018, 25, 410–421. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Grundmann, M.; Haidar, M.; Placzko, S.; Niendorf, R.; Darashchonak, N.; Hubel, C.A.; von Versen-Hoynck, F. Vitamin D improves the angiogenic properties of endothelial progenitor cells. Am. J. Physiol. Cell Physiol. 2012, 303, C954–C962. [Google Scholar] [CrossRef] [PubMed]
  126. Schröder-Heurich, B.; Hardenberg, S.v.; Brodowski, L.; Kipke, B.; Meyer, N.; Borns, K.; Kaisenberg, C.S.V.; Brinkmann, H.; Claus, P.; Versen-Höynck, F.V. Vitamin D improves endothelial barrier integrity and counteracts inflammatory effects on endothelial progenitor cells. Faseb J. 2019, 33, 9142–9153. [Google Scholar] [CrossRef]
  127. Yu, P.; Song, H.; Gao, J.; Li, B.; Liu, Y.; Wang, Y. Vitamin D (1,25-(OH)2D3) regulates the gene expression through competing endogenous RNAs networks in high glucose-treated endothelial progenitor cells. J. Steroid Biochem. Mol. Biol. 2019, 193, 105425. [Google Scholar] [CrossRef]
  128. Xu, W.; Hu, X.; Qi, X.; Zhu, R.; Li, C.; Zhu, Y.; Yin, S.; Cheng, L.; Zhu, R. Vitamin D Ameliorates Angiotensin II-Induced Human Endothelial Progenitor Cell Injury via the PPAR-gamma/HO-1 Pathway. J. Vasc. Res. 2019, 56, 17–27. [Google Scholar] [CrossRef]
  129. Hammer, Y.; Soudry, A.; Levi, A.; Talmor-Barkan, Y.; Leshem-Lev, D.; Singer, J.; Kornowski, R.; Lev, E.I. Effect of vitamin D on endothelial progenitor cells function. PloS ONE 2017, 12, e0178057. [Google Scholar] [CrossRef] [Green Version]
  130. Dominici, M.; Le Blanc, K.; Mueller, I.; Slaper-Cortenbach, I.; Marini, F.; Krause, D.; Deans, R.; Keating, A.; Prockop, D.; Horwitz, E. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 2006, 8, 315–317. [Google Scholar] [CrossRef]
  131. Shafiee, A.; Patel, J.; Lee, J.S.; Hutmacher, D.W. Mesenchymal stem/stromal cells enhance engraftment, vasculogenic and pro-angiogenic activities of endothelial colony forming cells in immunocompetent hosts. Sci. Rep. 2017, 7, 13558. [Google Scholar] [CrossRef]
  132. Kramann, R.; Couson, S.K.; Neuss, S.; Kunter, U.; Bovi, M.; Bornemann, J.; Knuchel, R.; Jahnen-Dechent, W.; Floege, J.; Schneider, R.K. Exposure to uremic serum induces a procalcific phenotype in human mesenchymal stem cells. Arterioscler. Thromb. Vasc. Biol. 2011, 31, e45–e54. [Google Scholar] [CrossRef] [Green Version]
  133. Wang, W.; Li, C.; Pang, L.; Shi, C.; Guo, F.; Chen, A.; Cao, X.; Wan, M. Mesenchymal stem cells recruited by active TGFβ contribute to osteogenic vascular calcification. Stem Cells Dev. 2014, 23, 1392–1404. [Google Scholar] [CrossRef] [Green Version]
  134. Wang, S.; Tong, M.; Hu, S.; Chen, X. The Bioactive Substance Secreted by MSC Retards Mouse Aortic Vascular Smooth Muscle Cells Calcification. BioMed Res. Int. 2018, 2018, 6053567. [Google Scholar] [CrossRef] [Green Version]
  135. Wang, S.; Hu, S.; Wang, J.; Liu, Y.; Zhao, R.; Tong, M.; Cui, H.; Wu, N.; Chen, X. Conditioned medium from bone marrow-derived mesenchymal stem cells inhibits vascular calcification through blockade of the BMP2-Smad1/5/8 signaling pathway. Stem Cell Res. Ther. 2018, 9, 160. [Google Scholar] [CrossRef] [Green Version]
  136. Xie, C.; Ouyang, L.; Chen, J.; Zhang, H.; Luo, P.; Wang, J.; Huang, H. The Emerging Role of Mesenchymal Stem Cells in Vascular Calcification. Stem Cells Int. 2019, 2019, 2875189. [Google Scholar] [CrossRef]
  137. Lee, K.-M.; Kang, H.-A.; Park, M.; Lee, H.-Y.; Choi, H.-R.; Yun, C.-H.; Oh, J.-W.; Kang, H.-S. Interleukin-24 attenuates β-glycerophosphate-induced calcification of vascular smooth muscle cells by inhibiting apoptosis, the expression of calcification and osteoblastic markers, and the Wnt/β-catenin pathway. Biochem. Biophys. Res. Commun. 2012, 428, 50–55. [Google Scholar] [CrossRef]
  138. Oma, I.; Andersen, J.K.; Lyberg, T.; Molberg, O.; Whist, J.E.; Fagerland, M.W.; Almdahl, S.M.; Hollan, I. Plasma vitamin D levels and inflammation in the aortic wall of patients with coronary artery disease with and without inflammatory rheumatic disease. Scand. J. Rheumatol. 2017, 46, 198–205. [Google Scholar] [CrossRef]
  139. Oma, I.; Olstad, O.K.; Andersen, J.K.; Lyberg, T.; Molberg, Ø.; Fostad, I.; Wang Fagerland, M.; Almdahl, S.M.; Rynning, S.E.; Yndestad, A.; et al. Differential expression of vitamin D associated genes in the aorta of coronary artery disease patients with and without rheumatoid arthritis. PLoS ONE 2018, 13, e0202346. [Google Scholar] [CrossRef]
  140. Wasnik, S.; Rundle, C.H.; Baylink, D.J.; Yazdi, M.S.; Carreon, E.E.; Xu, Y.; Qin, X.; Lau, K.W.; Tang, X. 1,25-Dihydroxyvitamin D suppresses M1 macrophages and promotes M2 differentiation at bone injury sites. Jci Insight 2018, 3, e98773. [Google Scholar] [CrossRef] [Green Version]
  141. Zhu, J.; Bing, C.; Wilding, J.P.H. Vitamin D receptor ligands attenuate the inflammatory profile of IL-1beta-stimulated human white preadipocytes via modulating the NF-kappaB and unfolded protein response pathways. Biochem. Biophys. Res. Commun. 2018, 503, 1049–1056. [Google Scholar] [CrossRef]
  142. Schmidt, N.; Brandsch, C.; Kühne, H.; Thiele, A.; Hirche, F.; Stangl, G.I. Vitamin D receptor deficiency and low vitamin D diet stimulate aortic calcification and osteogenic key factor expression in mice. PLoS ONE 2012, 7, e35316. [Google Scholar] [CrossRef]
  143. Fu, B.; Wang, H.; Wang, J.; Barouhas, I.; Liu, W.; Shuboy, A.; Bushinsky, D.A.; Zhou, D.; Favus, M.J. Epigenetic regulation of BMP2 by 1,25-dihydroxyvitamin D3 through DNA methylation and histone modification. PLoS ONE 2013, 8, e61423. [Google Scholar] [CrossRef]
  144. Nguyen-Yamamoto, L.; Tanaka, K.-I.; St-Arnaud, R.; Goltzman, D. Vitamin D-regulated osteocytic sclerostin and BMP2 modulate uremic extraskeletal calcification. Jci Insight 2019, 4, e126467. [Google Scholar] [CrossRef]
  145. Zhu, M.; Fang, X.; Zhou, S.; Li, W.; Guan, S. Indirect coculture of vascular smooth muscle cells with bone marrow mesenchymal stem cells inhibits vascular calcification and downregulates the Wnt signaling pathways. Mol. Med. Rep. 2016, 13, 5141–5148. [Google Scholar] [CrossRef]
  146. Huang, Y.; Wang, L.; Jia, X.X.; Lin, X.X.; Zhang, W.X. Vitamin D alleviates airway remodeling in asthma by down-regulating the activity of Wnt/beta-catenin signaling pathway. Int. Immunopharmacol. 2019, 68, 88–94. [Google Scholar] [CrossRef]
  147. Schaub, T.; Gurgen, D.; Maus, D.; Lange, C.; Tarabykin, V.; Dragun, D.; Hegner, B. mTORC1 and mTORC2 Differentially Regulate Cell Fate Programs to Coordinate Osteoblastic Differentiation in Mesenchymal Stromal Cells. Sci. Rep. 2019, 9, 20071. [Google Scholar] [CrossRef]
  148. Lisse, T.S.; Hewison, M. Vitamin D: A new player in the world of mTOR signaling. Cell Cycle 2011, 10, 1888–1889. [Google Scholar] [CrossRef] [Green Version]
  149. Valle, Y.L.; Almalki, S.G.; Agrawal, D.K. Vitamin D machinery and metabolism in porcine adipose-derived mesenchymal stem cells. Stem Cell Res. Ther. 2016, 7, 118. [Google Scholar] [CrossRef] [Green Version]
  150. Pesarini, J.R.; Oliveira, R.J.; Pessatto, L.R.; Antoniolli-Silva, A.; Felicidade, I.; Nardi, N.B.; Camassola, M.; Mantovani, M.S.; Ribeiro, L.R. Vitamin D: Correlation with biochemical and body composition changes in a southern Brazilian population and induction of cytotoxicity in mesenchymal stem cells derived from human adipose tissue. Biomed. Pharmacother. Biomed. Pharmacother. 2017, 91, 861–871. [Google Scholar] [CrossRef] [Green Version]
  151. Song, I.; Kim, B.S.; Kim, C.S.; Im, G.I. Effects of BMP-2 and vitamin D3 on the osteogenic differentiation of adipose stem cells. Biochem. Biophys. Res. Commun. 2011, 408, 126–131. [Google Scholar] [CrossRef] [PubMed]
  152. Karkeni, E.; Marcotorchino, J.; Tourniaire, F.; Astier, J.; Peiretti, F.; Darmon, P.; Landrier, J.-F. Vitamin D Limits Chemokine Expression in Adipocytes and Macrophage Migration In Vitro and in Male Mice. Endocrinology 2015, 156, 1782–1793. [Google Scholar] [CrossRef]
  153. Karkeni, E.; Bonnet, L.; Marcotorchino, J.; Tourniaire, F.; Astier, J.; Ye, J.; Landrier, J.F. Vitamin D limits inflammation-linked microRNA expression in adipocytes in vitro and in vivo: A new mechanism for the regulation of inflammation by vitamin D. Epigenetics 2018, 13, 156–162. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Muruganandan, S.; Roman, A.A.; Sinal, C.J. Adipocyte differentiation of bone marrow-derived mesenchymal stem cells: Cross talk with the osteoblastogenic program. Cell. Mol. Life Sci. CMLS 2009, 66, 236–253. [Google Scholar] [CrossRef] [PubMed]
  155. Lu, C.-L.; Shyu, J.-F.; Wu, C.-C.; Hung, C.-F.; Liao, M.-T.; Liu, W.-C.; Zheng, C.-M.; Hou, Y.-C.; Lin, Y.-F.; Lu, K.-C. Association of Anabolic Effect of Calcitriol with Osteoclast-Derived Wnt 10b Secretion. Nutrients 2018, 10, 1164. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. The mechanism of the vascular calcification based on endothelial injury, vascular smooth muscle cell (VSMC) calcification, mesenchymal stem cells (MSCs)/pericytes in the adventitial layer, and the deficiency of calcification inhibitors. Cells from all layers of the vessel wall transformed into osteoblast-like cells. The atherosclerosis within the endothelium induced endothelial calcification by releasing matrix vesicles (MV)/extracellular vesicles (EV) with insufficient matrix Gla protein (MGP)/fetuin A. On the other hand, atherosclerosis also stimulated VSMCs to release MV/EV with insufficient fetuin A after being injured by uremic toxin or renin–angiotensin–aldosterone system (RAAS) activation. The adventitial MSCs/pericyte migrated to the medial layer and transdifferentiated into osteoblast-like cells, which contributed to calcification of the medial layer. CPP: calciprotein particle.
Figure 1. The mechanism of the vascular calcification based on endothelial injury, vascular smooth muscle cell (VSMC) calcification, mesenchymal stem cells (MSCs)/pericytes in the adventitial layer, and the deficiency of calcification inhibitors. Cells from all layers of the vessel wall transformed into osteoblast-like cells. The atherosclerosis within the endothelium induced endothelial calcification by releasing matrix vesicles (MV)/extracellular vesicles (EV) with insufficient matrix Gla protein (MGP)/fetuin A. On the other hand, atherosclerosis also stimulated VSMCs to release MV/EV with insufficient fetuin A after being injured by uremic toxin or renin–angiotensin–aldosterone system (RAAS) activation. The adventitial MSCs/pericyte migrated to the medial layer and transdifferentiated into osteoblast-like cells, which contributed to calcification of the medial layer. CPP: calciprotein particle.
Ijms 21 02466 g001
Figure 2. The adjunctive role of vitamin D in treating vascular calcification based on MSCs and endothelial progenitor cells (EPCs). In physiological hypoxia, angiogenesis can repair a damaged endothelium by promoting the differentiation of EPCs. The circulating EPCs migrated and invaded the subendothelial region to replace the injured endothelial cells; they also regulated the differentiation of the surrounding stromal cells and therefore reduced calcification. The MSCs mitigated calcification by lowering the proinflammatory cytokines or reducing the VSMC osteogenic expression. Vitamin D served as the adjunctive role in mitigating calcification by influencing EPCs and MSCs in several manners. For EPCs, vitamin D enhanced the EPCs mobilization during angiogenesis by increasing the vascular endothelial growth factor (VEGF) release. Vitamin D also enhanced the EPC adhesion and migration. Under the inflammatory status, such as high TNF-α scenario, the further vascular calcifications could be lessened by the usage of vitamin-D. For MSCs, vitamin D influenced the secretion of proinflammatory cytokines, such as TNF-α, interleukin 1(IL-1), and interleukin 6 (IL-6), which might induce osteogenic MSCs. Vitamin D decreased the VSMC osteogenic differentiation by decreasing the BMP2-Smad1/5/8 (mothers against decapentaplegic homolog 1/5/8) signal or Wnt5a expression.
Figure 2. The adjunctive role of vitamin D in treating vascular calcification based on MSCs and endothelial progenitor cells (EPCs). In physiological hypoxia, angiogenesis can repair a damaged endothelium by promoting the differentiation of EPCs. The circulating EPCs migrated and invaded the subendothelial region to replace the injured endothelial cells; they also regulated the differentiation of the surrounding stromal cells and therefore reduced calcification. The MSCs mitigated calcification by lowering the proinflammatory cytokines or reducing the VSMC osteogenic expression. Vitamin D served as the adjunctive role in mitigating calcification by influencing EPCs and MSCs in several manners. For EPCs, vitamin D enhanced the EPCs mobilization during angiogenesis by increasing the vascular endothelial growth factor (VEGF) release. Vitamin D also enhanced the EPC adhesion and migration. Under the inflammatory status, such as high TNF-α scenario, the further vascular calcifications could be lessened by the usage of vitamin-D. For MSCs, vitamin D influenced the secretion of proinflammatory cytokines, such as TNF-α, interleukin 1(IL-1), and interleukin 6 (IL-6), which might induce osteogenic MSCs. Vitamin D decreased the VSMC osteogenic differentiation by decreasing the BMP2-Smad1/5/8 (mothers against decapentaplegic homolog 1/5/8) signal or Wnt5a expression.
Ijms 21 02466 g002
Table 1. The influence of vitamin D on EPCs in the development of vascular calcification.
Table 1. The influence of vitamin D on EPCs in the development of vascular calcification.
Performance of EPCsCharacteristics Surface Marker
Vitamin D receptors on EPCsDecrease in coronary artery disease (CAD) [124]CD45dim, CD34+, and KDR+
EPCs migration and differentiationAccelerated [125]CD34+, CD31+, CD45−, and CD133−
Endothelial colony-forming cells expressed mRNA of VEGF and pro–matrix metalloproteinase (pro-MMP) activityIncreased [125]CD34+, CD31+, CD45−, and CD133−
Endothelial progenitor adhesionIncreased [126]CD31+, CD45+, and CD133+
Migration of the EPCs from the bone marrowIncreased [127]1,1′-Dioctadecyl-3,3,3′,3′-tetramethylindocarbocyanine-labeled acetylated low density lipoprotein and fluorescein isothiocyanate -Ulex europaeus agglutinin-1
Formation of VE-cadherin adhesion junctions on the EPCsIncreased [126]CD31+, CD45+, and CD133+
EPC injury by Ang II through modulating the PPAR-γ/HO-1 pathwayDecreased [128]VEGF-2+ and CD13+
EPC viabilityImproved [129]CD34+ and KDR+

Share and Cite

MDPI and ACS Style

Hou, Y.-C.; Lu, C.-L.; Zheng, C.-M.; Liu, W.-C.; Yen, T.-H.; Chen, R.-M.; Lin, Y.-F.; Chao, C.-T.; Lu, K.-C. The Role of Vitamin D in Modulating Mesenchymal Stem Cells and Endothelial Progenitor Cells for Vascular Calcification. Int. J. Mol. Sci. 2020, 21, 2466. https://doi.org/10.3390/ijms21072466

AMA Style

Hou Y-C, Lu C-L, Zheng C-M, Liu W-C, Yen T-H, Chen R-M, Lin Y-F, Chao C-T, Lu K-C. The Role of Vitamin D in Modulating Mesenchymal Stem Cells and Endothelial Progenitor Cells for Vascular Calcification. International Journal of Molecular Sciences. 2020; 21(7):2466. https://doi.org/10.3390/ijms21072466

Chicago/Turabian Style

Hou, Yi-Chou, Chien-Lin Lu, Cai-Mei Zheng, Wen-Chih Liu, Tzung-Hai Yen, Ruei-Ming Chen, Yuh-Feng Lin, Chia-Ter Chao, and Kuo-Cheng Lu. 2020. "The Role of Vitamin D in Modulating Mesenchymal Stem Cells and Endothelial Progenitor Cells for Vascular Calcification" International Journal of Molecular Sciences 21, no. 7: 2466. https://doi.org/10.3390/ijms21072466

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

Article Metrics

Back to TopTop