Next Article in Journal
Effect of Docosahexaenoic Acid (DHA) Supplementation of Preterm Infants on Growth, Body Composition, and Blood Pressure at 7-Years Corrected Age: Follow-Up of a Randomized Controlled Trial
Previous Article in Journal
Whole Dietary Patterns, Cognitive Decline and Cognitive Disorders: A Systematic Review of Prospective and Intervention Studies
Previous Article in Special Issue
Nutrition Strategies Promoting Healthy Aging: From Improvement of Cardiovascular and Brain Health to Prevention of Age-Associated Diseases
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Role of Vitamin D Deficiency in the Pathogenesis of Cardiovascular and Cerebrovascular Diseases

by
Éva Pál
1,2,*,
Zoltán Ungvári
3,4,5,6,
Zoltán Benyó
1,2,† and
Szabolcs Várbíró
7,8,†
1
Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary
2
Eötvös Loránd Research Network, Semmelweis University (ELKH-SU), Cerebrovascular and Neurocognitive Disorders Research Group, 1094 Budapest, Hungary
3
Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
4
Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
5
International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Departments of Public Health and Translational Medicine, Semmelweis University, 1085 Budapest, Hungary
6
The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
7
Department of Obstetrics and Gynecology, Semmelweis University, 1082 Budapest, Hungary
8
Workgroup for Science Management, Doctoral School, Semmelweis University, 1085 Budapest, Hungary
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Nutrients 2023, 15(2), 334; https://doi.org/10.3390/nu15020334
Submission received: 5 December 2022 / Revised: 29 December 2022 / Accepted: 30 December 2022 / Published: 9 January 2023
(This article belongs to the Special Issue Dietary Interventions against Age-Related Vascular Pathologies)

Abstract

:
Deficiency in vitamin D (VitD), a lipid-soluble vitamin and steroid hormone, affects approximately 24% to 40% of the population of the Western world. In addition to its well-documented effects on the musculoskeletal system, VitD also contributes importantly to the promotion and preservation of cardiovascular health via modulating the immune and inflammatory functions and regulating cell proliferation and migration, endothelial function, renin expression, and extracellular matrix homeostasis. This brief overview focuses on the cardiovascular and cerebrovascular effects of VitD and the cellular, molecular, and functional changes that occur in the circulatory system in VitD deficiency (VDD). It explores the links among VDD and adverse vascular remodeling, endothelial dysfunction, vascular inflammation, and increased risk for cardiovascular and cerebrovascular diseases. Improved understanding of the complex role of VDD in the pathogenesis of atherosclerotic cardiovascular diseases, stroke, and vascular cognitive impairment is crucial for all cardiologists, dietitians, and geriatricians, as VDD presents an easy target for intervention.

1. Introduction

Vitamin D (VitD) is a lipid-soluble vitamin that functions as a steroid hormone [1]. VitD is best known for its role in calcium and bone homeostasis. Since the public has been educated about the importance of VitD, foods are now fortified with VitD and VitD containing dietary supplements are widely available, public health authorities thought that health problems resulting from VitD deficiency (VDD) had been resolved. Yet, it became apparent that, while rickets due to severe VitD is indeed very rare, moderate VDD is still quite prevalent and represents an important health challenge.
Estimates of the prevalence of VDD ranges from 24% (United States) to 40% (European Union) [2,3,4,5,6,7,8]. It is now evident that, in addition to the musculoskeletal system, most tissues and cell types express VitD receptors and VitD, in addition to its role in promoting and maintaining skeletal health, confers complex health benefits in multiple organ systems, including the immune system and the cardiovascular system [9,10,11]. Accordingly, efforts have been made to examine the causes, complex consequences, and prevention strategies of the “world pandemic” of VDD [2]. VDD has already been linked to increased risk for several diseases, including common cancers (colon, breast, and prostate cancers), diabetes mellitus, coronary artery disease, ischemic stroke, and autoimmune diseases [2].
Cardiovascular and cerebrovascular diseases belong to the leading causes of death and disability worldwide [12,13]. Importantly, the prevalence of cardiovascular diseases has been reported to increase with aging [14]. Since the average life expectancy is increasing globally, the burden to prevent and treat cardiovascular diseases will increase in the following decades. Several studies investigating the physiological and pathophysiological properties of the cardiovascular and cerebrovascular system have focused on the role of endothelial function in the maintenance of vascular health. Endothelial dysfunction, characterized by imbalanced vasodilation and vasoconstriction, elevated reactive oxygen species, and proinflammatory factors, decreases nitric oxide bioavailability and contributes significantly to the development and progression of cardiovascular and cerebrovascular diseases [15,16,17]. Although many mechanisms and risk factors of cardiovascular disorders have already been identified, the prevention and treatment strategies of vascular diseases are yet to be improved. To preserve cardiovascular health, even in advanced age, the pathogenesis and the risk factors of cardiovascular diseases have to be further investigated. In addition to traditional risk factors (e.g., hypertension, diabetes mellitus), VDD appears to increase the risk of cardiovascular and cerebrovascular disorders [1,13,18].
In this review, the effects of VitD on the functional and structural integrity of the circulatory system are considered, in terms of potential mechanisms involved in endothelial dysfunction and accelerated vascular aging phenotypes associated with VDD. The role of VDD in the pathogenesis of specific cardiovascular and cerebrovascular diseases (including stroke and vascular cognitive impairment (VCI)) is discussed [13].

2. Vitamin D Biosynthesis and Metabolism

Vitamin D can represent either vitamin D2 (ergocalciferol) or vitamin D3 (cholecalciferol), both of which are produced naturally by ultraviolet B radiation (290 to 315 nm wavelength) from ergosterol in yeast and mushrooms or from 7-dehydrocholesterol in the epidermis [2]. Humans acquire VitD mainly from its precursors upon exposure to sunlight and, to a lesser extent, from certain foods, such as oily fish [1]. Following the exposure of skin to sunlight, 7-dehydrocholesterol is converted first to pre-vitamin D3, which spontaneously isomerizes to vitamin D3 in a thermosensitive process [19]. Vitamin D2 or D3 from ingested food is incorporated into chylomicrons, followed by absorption into the lymphatic system and entering the venous blood. Inactive vitamin D (as well as its metabolites) circulates within the blood stream bound to carrier proteins, mainly to the VitD binding protein [20], and is subsequently metabolized in two steps to its active form, 1,25-dihydroxyvitamin D (1,25(OH)2D, calcitriol). First, the biomarker of VitD status, 25-hydroxyvitamin D (25(OH)D) is produced mostly by cytochrome P450 (CYP) 2R1 (but also by CYP27A1, CYP3A4, and CYP2J3) in the liver [2]. Next, 1,25(OH)2D is formed exclusively by CYP27B1 (25-hydroxyvitamin D-1α-hydroxylase), particularly in the kidney [19,21]. In addition to the kidney, many extrarenal tissues express CYP27B1; therefore, those are also capable of producing the active form of VitD [22]. The extrarenal production of 1,25(OH)2D is stimulated mainly by cytokines and appears to be important in the paracrine regulation of cell function [23]. Unlike extrarenal CYP27B1, renal 1α-hydroxylase is tightly regulated by the parathyroid hormone, fibroblast growth factor 23, and the plasma levels of 1,25(OH)2D, calcium, and phosphate ions [19,22]. In order to avoid the accumulation of 1,25(OH)2D or 25(OH)D, the target cells of VitD express CYP24A1 (24-hydroxylase), which converts 1,25(OH)2D to biologically inactive calcitroic acid [19], whereas, in the kidney, it is 24-hydroxylase catabolyses 25(OH)D, when a sufficient amount of 1,25(OH)2D has already been produced [21].

3. Mechanism of Action of Vitamin D

3.1. Genomic Actions

The biological actions of 1,25(OH)2D are mediated by the vitamin D receptor (VDR), which belongs to the nuclear receptor superfamily and acts as a ligand-activated transcription factor [20]. VDR was first discovered in chicken intestines [24], but was later found to be present in almost all cells and tissues [25]. VDR regulates the expression of numerous genes the promoters that contain specific DNA sequences known as vitamin D response elements (VDRE) [1,25]. The binding of 1,25(OH)2D to VDR induces conformational changes in the receptor that facilitate its interaction with the retinoid X receptor (RXR) and, subsequently, the formation of a VDR/RXR heterodimer, which provides adequate DNA binding affinity [20,26]. The ligand-bound VDR/RXR heterodimeric complex binds to the VDRE on the target genes and acts as a transcription factor that up- or downregulates their transcription [20,27]. The action of 1,25(OH)2D depends, however, on the involvement of tissue specific co-factors, for instance, the steroid-specific coactivators, and subsequently on the formation of transcriptional complexes [25]. The genomic actions of 1,25(OH)2D are schematically shown in Figure 1.

3.2. Non-Genomic Actions

Interestingly, VitD has been reported to have some rapid actions that are unlikely to involve the direct regulation of gene expression. These effects may, rather, be mediated by a membrane-associated VDR, which has been less well-characterized than the nuclear VDR [25]. The non-genomic actions of VitD include the activation of signaling molecules (e.g., phospholipase C (PLC), phospholipase A2 (PLA2), phosphatidylinositol-3-kinase (PI3K)), the rapid generation of second messengers, such as Ca2+, the activation of protein kinases, and the opening of the Ca2+ and Cl channels [28]. Surprisingly, however, these rapid, non-genomic actions appear to require the presence of the nuclear VDR, implying cooperation between the membrane-associated and nuclear VDRs [25]. In addition, the ligand-bound nuclear VDR has been reported to have non-classical, non-genomic actions. In that case, VitD regulates the target gene expression via protein–protein interactions, instead of binding to the VDRE on target genes [28].

4. Physiological Significance of Optimal Vitamin D Status

4.1. Determinants of Vitamin D Status

Vitamin D deficiency and insufficiency—defined usually as 25(OH)D levels below 20 ng/mL (50 nmol/L) and within the range of 21–29 ng/mL (52.5–72.5 nmol/L), respectively [10]—affect approximately one billion people worldwide [2]. Lifestyle and environmental factors limiting sunlight exposure of the skin are the main causes of VDD, but the decreased synthesis of 25(OH)D or 1,25(OH)2D, and heritable disorders such as hereditary VitD-resistant rickets could also reduce the bioavailability of VitD [2]. Several diseases, including cardiovascular disorders, have been associated with decreased VitD serum levels [2]. For instance, lower 25(OH)D serum levels (~27 ng/mL) have been reported in patients with hypertension, compared to normotensive individuals (~31 ng/mL) [29]. Similarly, Melamed et al. reported lower 25(OH)D levels among patients with peripheral arterial disease (21.5 ng/mL vs. 24.6 ng/mL in healthy subjects) [30]. Since there are few foods naturally containing VitD (such as cod liver oil, shiitake mushrooms, egg yolk), in general, sufficient VitD supply can be provided only by exposure to sunlight or by taking VitD supplements [2,11]. As a variety of factors could reduce the cutaneous production of VitD, such as ultraviolet protection, increased skin pigmentation, age, and seasonal and geographical variation [11], it is recommended to take VitD supplements: in general, 1000–2000 IU/day are needed to reach and maintain 25(OH)D levels greater than 30 ng/mL in the majority of the healthy population, in order to prevent VDD [10]. Unlike VDD, VitD intoxication (25(OH)D levels higher than 150 ng/mL (374 nmol/L)) is extremely rare, particularly because it cannot be caused by exposure to sunlight, since excess pre-vitamin D3 or vitamin D3 is destroyed by sunlight itself [2].

4.2. Physiological Role of Vitamin D

VitD appears to control the expression of more than 200 genes, as well as several signaling molecules and second messengers, including those not typically associated with mineral homeostasis (Table 1) [26,28]. The active form of VitD regulates, for instance, cellular proliferation, differentiation, apoptosis, angiogenesis, oxidative stress, membrane transport, matrix homeostasis, cell adhesion, immune functions, insulin secretion, and renin expression (Figure 1) [2,26,28,31]; thus, VitD plays an integral physiological role in nonskeletal tissues. Consequently, in addition to its well-characterized roles in calcium and phosphate homeostasis, as well as in bone metabolism, VitD exerts beneficial effects, for instance, on glucose homeostasis, the immune response, and the cardiovascular system [2]. More severe VDD impairs the mineral and bone homeostasis characterized by rickets and growth retardation in children, as well as osteomalacia, osteoporosis, and decreased muscle strength or sarcopenia in adults and the elderly [2]. Additionally, VDD is also associated with increased risk for cancer (e.g., colon, prostate, and breast cancer), diabetes mellitus, metabolic syndrome, infections, autoimmune diseases, depression, schizophrenia, and cardiovascular diseases [2,32].
Although VitD intoxication is rare, it can be caused by taking extensively high doses of VitD supplements [2]. The clinical manifestations of VitD toxicity are related primarily to hypercalcemia, and they include confusion, depression, psychosis, gastrointestinal disorders, renal failure, and cardiovascular symptoms, such as hypertension and bradyarrhythmia [39]. Surprisingly, it appears that both low and high 25(OH)D levels are associated with increased risk of total [40] and cardiovascular mortality [16,41], implying a U-shaped association between VitD concentrations and health. Although VitD appears to have a broad therapeutic window, the latter still has to be defined, especially for preventing cardiovascular and cerebrovascular diseases [31]. Nevertheless, optimal VitD supply is considered to be a prerequisite for health in all age groups [42].

5. Impacts of Vitamin D on the Cardiovascular System

5.1. Cellular Effects of Vitamin D

There is a growing body of evidence linking VDD to cardiovascular diseases [1]. For instance, VDD is associated with atherosclerosis, hypertension, cardiac hypertrophy, cerebrovascular diseases, coronary heart disease, and peripheral artery disease [2], as well as with several cardiovascular risk factors, such as dyslipidemia, insulin resistance, diabetes mellitus, and abdominal obesity [1,2,32]. VitD exerts a direct effect on the cardiovascular system, since VDRs have been found in cardiomyocytes [43], vascular smooth muscle cells (VSMCs) [44], endothelial cells [45], circulating monocytes, macrophages, dendritic cells, activated T lymphocytes [46], and platelets [47]. Furthermore, CYP27B1 (25-hydroxyvitamin D-1α-hydroxylase) is expressed in most of these cells, which enables the local synthesis of 1,25(OH)2D [1]. The cardiovascular protective effects of VitD include the modulation of immune, inflammatory, and endothelial functions [1]. Furthermore, VitD regulates cell proliferation and migration, renin expression, and extracellular matrix homeostasis, and it may attenuate the adverse effects of advanced glycation end products on endothelial cells [1,32]. In addition, VitD has an antithrombotic effect, since it downregulates the tissue factor, plasminogen activator inhibitor-1, and thrombospondin-1, whereas it upregulates thrombomodulin expression in monocytes and VSMCs [1,48]. Furthermore, VitD inhibits formation of foam cells and cholesterol uptake by macrophages; thus, it also exerts antiatherogenic effects [32]. Figure 2 summarizes the effects of VitD related to the cardiovascular system.

5.2. Impacts of Vitamin D on Blood Pressure and Cardiac Functions

VitD appears to have a beneficial effect on arterial blood pressure, and consequently, VDD is linked to hypertension [1]. However, the association between VitD levels and blood pressure values is not fully confirmed [49], especially in young healthy subjects [50,51]. Nevertheless, the impact of VitD on blood pressure has been attributed particularly to the negative regulation of the renin–angiotensin system (RAS) [52], since VitD appears to decrease the activity of the cyclic adenosine monophosphate response element in the renin gene promoter [38]. Consequently, VDR deficiency increases the expression of renin and, therefore, the production of angiotensin II, which can result in hypertension and cardiac hypertrophy [52]. Surprisingly, however, normotensive VDR knockout mice also developed cardiac hypertrophy [43], which could imply that VitD acts directly on cardiomyocytes [31,53]. Accordingly, VitD has been reported to stimulate cardiomyocyte relaxation, which could improve coronary perfusion during diastole, and it also regulates the gene expression profile of the extracellular matrix in the heart [32]. In conclusion, experimental animal models of VDD indicate that VitD prevents hypertension; however, the causal association between VitD levels and blood pressure values is still not fully confirmed.

5.3. Effects of Vitamin D on Angiogenesis and Vascular Remodeling

VitD has been reported to regulate the expression of several genes involved in cell proliferation and differentiation [35], as well as in extracellular matrix homeostasis (Table 1) [1] and, therefore, VitD likely modulates processes of angiogenesis and vascular remodeling. Accordingly, preclinical studies show that VitD attenuates pathological vascular remodeling both in intrarenal arteries in kidney fibrosis [54] and in basilar arteries after subarachnoid hemorrhage in rodent models [55]. Further, VDD has been reported to decrease the lumen and increase the wall thickness of coronary arterioles of female rats [56]. Altered VSMC migration and proliferation may be responsible for the vascular remodeling in VDD [16]. However, the literature is controversial regarding the effect of VitD treatment on VSMCs. Some studies using rat VSMCs report enhanced migration and proliferation [57,58], whereas others found VitD-induced inhibition of rat and human VSMC growth [44,59]. The effect of VitD treatment on VSMCs appears to depend on the applied dose. For instance, physiological doses of VitD inhibit VSMC proliferation [16] via blunting c-myc RNA induction [58], up-regulating the negative modulators of cell proliferation, including transforming growth factor β (TGF-β) [33], or decreasing cyclin-dependent kinase 2 (Cdk2) activity [60].
In addition, VitD participates in the modulation of endothelial cell proliferation and matrix homeostasis, due to the regulation of vascular endothelial growth factor (VEGF) and matrix metalloproteinases (MMPs) [1]. VEGF is known to stimulate endothelial cell proliferation and migration and mediate vascular growth and angiogenesis [61], whereas MMPs regulate angiogenesis and vascular remodeling by degrading extracellular matrix proteins [62]. Experimental animal models of VDD and clinical studies have reported that VDD decreased the expression of tissue inhibitors of MMP-1 and MMP-3, but upregulated the expression of MMP-2 and MMP-9 [1]. These alterations in extracellular matrix homeostasis may contribute to the development of vascular calcification [63]. Furthermore, VitD induces the upregulation of VEGF in endothelial progenitor cells [36], as well as in mature endothelial cells [64], and in VSMCs [27,57], implying the role of VitD in vasculogenesis, angiogenesis, and endothelial repair [61]. Surprisingly, however, VitD has also been reported to inhibit angiogenesis partly via reducing the protein expression of VEGF in various human tumor cells [65]. Thus, the effect of VitD on angiogenesis is ambiguous; however, the inhibitory impact of VitD on VEGF expression and angiogenesis has been reported almost exclusively in cancer studies [66]. In addition, VitD appears to stimulate the ability of multipotent mesenchymal stromal cells to promote vasculogenesis [67].
Furthermore, VitD regulates the elastin and collagen content of the vessel wall [37]; thus, it influences vascular resistance and arterial stiffness [68]. For instance, Andrukhova et al. reported increased collagen and decreased elastin content of the ascending aorta of 9-month-old VDR-deficient mice [37]. Correspondingly, Salum et al. found that VitD could preserve the structure of elastic fibers and the ratio of elastic fibers to collagen in the tunica media of the aorta in experimental diabetes [69]. Since the increase in the collagen-to-elastin ratio could increase arterial stiffness [68], VitD appears to participate in the maintenance of the normal elasticity of the vessel wall.
In summary, VitD is likely to be involved in the modulation of vascular remodeling, arterial stiffness, and angiogenesis. For instance, VitD has been reported to participate in the regulation of endothelial and smooth muscle cell proliferation, the extracellular matrix homeostasis, and the collagen–elastin content of arterial wall. Figure 3 represents various actions of VDD on the vascular wall, which may be responsible for vascular remodeling in VDD.

5.4. Impact of Vitamin D on Endothelial Function

5.4.1. Vitamin D and the Nitric Oxide System

Normal endothelium-dependent, NO-mediated vasodilation is essential for maintenance of cardiovascular and cerebrovascular health [15,70,71,72,73,74]. Low levels of VitD are associated with diminished flow-mediated vasodilation in humans, which could be attributed to the endothelial dysfunction characteristic for VDD [32], as summarized in Figure 3. There is strong evidence that endothelial VDR plays an important role in preserving endothelial function [75]. According to in vitro studies using rodent and human tissues, VitD has been reported to upregulate the expression of endothelial nitric oxide synthase (eNOS) [76], to increase the dimer to monomer ratio of the eNOS protein [77], and to modulate the phosphorylation of eNOS [55,78], leading to increased eNOS activity and, thus, to enhanced NO production [79]. The vasoprotective action of VitD may involve activation of synergistic signaling pathways, including those mediated by cyclic adenosine monophosphate-activated protein kinase (AMPK) [55], the phosphoinositide-3-kinase (PI3K)/Akt, p38 mitogen-activated protein kinase (MAPK), and the extracellular signal-regulated kinase (ERK)/MAPK pathways [78]. In preclinical models, VitD treatment increased eNOS protein expression, improving endothelium-dependent vasorelaxation [80,81]. Conversely, in VDR-deficient mice, endothelium-mediated vasorelaxation of the aorta is impaired, due to reduced eNOS expression [75]. Additionally, diminished NO production has been reported in mice with functionally inactive VDR [37] and endothelial dysfunction in resistance arteries of rats exposed to early-life VDD [82]. In conclusion, according to human studies and animal experimentations, VitD may play an important role in preserving endothelial function, therefore preventing the development of age-related cardiovascular diseases.

5.4.2. Oxidative Stress, Inflammation, and Vitamin D

VDD also promotes oxidative stress and inflammation, which may contribute to the genesis of both endothelial dysfunction and atherogenesis (Figure 3) [16,83,84]. For instance, VitD-deficient diet has been reported to increase the production of reactive oxygen species (ROS) in the aortic wall of rats [85] and down-regulate the antioxidant, ROS-scavenging enzyme, and cytosolic copper-zinc superoxide dismutase (CuZn-SOD) in the mesenteric arteries of mice [86]. VitD was also shown to decrease the expression of the ROS-generating NADPH oxidase enzymes in endothelial cells cultured from rodent and human tissues [77,84,87,88]. Thus, the anti-oxidative, vasoprotective action of physiological VitD levels is likely mediated synergistically by the upregulation of anti-oxidative pathways and down-regulation of pro-oxidative mechanisms [64,87]. By altering these pathways VDD may promote increased ROS production in the cardiovascular system [84]. The increase of oxidative stress, in turn, could lead to the inactivation of NO or oxidation of tetrahydrobiopterin, a critical cofactor for eNOS, which leads to eNOS uncoupling and, thus, to endothelial dysfunction [89].
VitD appears to suppress inflammation via several mechanisms, such as inhibition of the prostaglandin and cyclooxygenase pathways, upregulation of anti-inflammatory cytokines (interleukin (IL)-4 and IL-10), downregulation of pro-inflammatory cytokines (IL-1, IL-2, IL-6, IL-23, tumor necrosis factor-α (TNF-α), and interferon-γ), decreasing cytokine-induced expression of adhesion molecules, and downregulation of the RAS [32] [1]. In VDR-KO mice, the lack of VitD-mediated renin suppression leads to an increase in angiotensin II levels [52], which can promote vascular inflammation [83]. VitD has been reported to suppress the expression of inflammatory mediators, such as TNF-α, cyclooxygenase 2 (COX-2), and monocyte chemoattractant protein-1 (MCP-1), in the aorta of apolipoprotein E (ApoE)-deficient atherosclerotic mice [90]. Additionally, VitD downregulated the expression of COX-2 and the thromboxane prostanoid (TP) receptor in renal artery segments and aortic endothelial cells of ovariectomized rats, thus improving endothelial function [91]. Furthermore, VitD inhibits the activation of nuclear factor-κB (NF-κB) [92], and it decreases the expression of IL-6 in human endothelial cells [93]; thus, it prevents endothelial inflammation, improves flow-mediated vasodilation, and protects against atherosclerosis [1]. Correspondently, VDD may be associated with an increased production of pro-inflammatory mediators in mice [86], which is likely to promote vascular inflammation (Figure 3) [46]. In addition, animal models and clinical studies evidence that VitD suppresses the responses of T helper type 1 (TH1) and T helper type 17 (TH17) cells, whereas it supports regulatory T (Treg) and T helper type 2 (TH2) cells, which also contributes to the prevention of atherosclerosis [1,94,95].
The influence of VitD status on oxidative and inflammatory pathways suggests that VitD plays an important role in preventing oxidative stress and inflammation in the vascular wall. Thus, VDD is likely to increase the risk of vascular inflammation, contributing to the development of cardiovascular and cerebrovascular diseases.

5.5. Effects of Vitamin D on the Vascular Tone

As VitD influences endothelial function, especially NO bioavailability [1], it is not surprising that VitD contributes to the regulation of vascular tone. For instance, enhanced myogenic tone [82] and increased angiotensin II-induced vasoconstriction [86] of mesenteric arteries have been reported in the rodent models of VDD. Interestingly, in the coronary arteries of female rats, VDD appears to reduce both the pressure-induced myogenic tone and vasoconstriction evoked by a thromboxane A2 (TXA2) agonist [56]. In addition to the modulation of NO production, VitD appears to reduce the endothelium-dependent contraction of the aorta of spontaneously hypertensive rats, due to reducing calcium influx into the endothelial cells, thereby decreasing the production of endothelium-derived contracting factors [96,97]. Furthermore, VitD normalized the vascular reactivity of mesenteric arteries of spontaneously hypertensive rats via restoring the function of apamin- and ATP-sensitive K+ channels in VSMCs [98]. In conclusion, VDD might modulate vascular tone in various vascular beds, especially due to altering endothelial release of vasoactive mediators and reactivity to vasoconstrictor agents.

5.6. Impacts of Vitamin D on Vascular Permeability

VitD might play a role in preserving endothelial barrier function in various organs, including the brain, kidneys, and lungs [99,100,101,102,103]. In a combined rat model of chronic kidney disease and VDD, supplementation with paricalcitol (an active VitD analogue) resulted in the restoration of the impaired aortic endothelial permeability induced by chronic kidney disease [103]. Paricalcitol has also been reported to alter cell adhesion molecule expression and decrease vascular permeability in mice with endotoxemia [100]. In lungs, VitD might decrease alveolar–capillary permeability. In VDR-KO mice, increased permeability of alveolar wall has been reported, which was associated with altered expression of tight junction and adherens junction components [103]. According to a human study, high-dose treatment with oral cholecalciferol reduced the alterations in the pulmonary vascular permeability index in adults; however, the extravascular lung water index was not affected [101]. In the cerebrovascular system, VDD compromised the blood–brain barrier in a rat model of transient middle cerebral artery occlusion characterized by increased immunglobulin extravasation and decreased expression of tight junction proteins [102]. Thus, VDD may exacerbate the consequences of ischemic stroke partly due to increased blood–brain barrier permeability [102]. Furthermore, in a mouse model of cerebral cavernous malformation disease, cholecalciferol supplementation has been reported to reduce the lesion burden [104]. Additionally, cholecalciferol and its metabolites, 25(OH)D and 1,25(OH)2D, modulated endothelial stability via non-genomic actions; thus, they reduced vascular permeability in human endothelial cell monolayer and in mouse cerebral arteries [105].

6. Vitamin D Deficiency, Cardiovascular Diseases, Coronary Artery Disease, and Heart Failure

Cardiovascular diseases belong to the leading causes of morbidity and mortality worldwide [12,18]. The association between VitD levels and cardiovascular disorders has been widely investigated and extensively reviewed in the past few decades [1,12,18,34,106,107]. According to human studies, VDD has been linked to the increased risk of coronary artery disease (CAD), myocardial infarction, and heart failure [18,108]. However, whether VitD supplements exert a benefical effect on the development and progression of cardiovascular diseases is still under debate [12].
Since VDD is associated with several risk factors for cardiovascular diseases, it is likely that it increases the incidence of cardiovascular disorders indirectly. For instance, low VitD levels have been reported to increase the risk of hypertension, atherosclerosis, inflammation, and diabetes mellitus [106]. Of note, the causal associaton between some risk factors, for instance hypertension, and VDD are still not proven unambiguously [49]. VitD also might impact the pathogenesis of cardiovascular diseases, including CAD directly. In rodent models of VDD, morphological remodeling and altered vasoreactivity of intramural coronary arteries have been reported [109,110,111]. The aforementioned alterations in the coronary arterioles may impair the coronary circulation, therefore increasing the risk for cardiovascular disease. Endothelial dysfunction and subsequent atherosclerosis of coronary arteries are one of the leading causes for CAD [106]. As VitD improves endothelial function, due to several mechanisms, including preservation of endothelial NO and preventing vascular inflammation (see details above), it is presumable that VDD might contribute to the pathogenesis of CAD via facilitating endothelial dysfunction. Additionally, in VDD, the cholesterol uptake by macrophages is promoted, leading to increased macrophage-derived foam cell formation in the endothelium, which facilitates the progression of atherosclerosis [106]. In addition to the effects of VitD on coronary arteries, VitD has been reported to act directly on cardiomyocytes [22,40]. In VDR-KO mice, cardiac hypertrophy has been observed [29], whereas VitD treatment improved cardiomyocyte relaxation and, therefore, coronary perfusion during diastole in rodents [23]. Furthermore, VitD has been reported to play a role in extracellular matrix homeostasis in cardiomyocytes (see details above) [1]; thus, VitD influences heart failure development [12,112].
Taken together, VDD may favor the development of cardiovascular diseases, including CAD and heart failure, via influencing both coronary perfusion and cardiac function. Additionally, VDD has been linked to several risk factors of cardiovascular diseases; therefore, it also increases the incidence of CAD and heart failure indirectly. Table 2 summarizes the role of VDD in the pathogenesis of cardiovascular and cerebrovascular diseases.

7. Vitamin D Deficiency, Cerebrovascular Diseases, Stroke, and Vascular Cognitive Impairment

7.1. Role of VDD in the Pathogenesis of Cerebrovascular Disease and Stroke

Atherosclerotic cerebrovascular diseases and consequential ischemic strokes belong to the leading causes of death and disability worldwide [113,114,115]. Strokes due to atherosclerosis of a larger artery account for approximately one third of all stroke cases [116]. For instance, carotid artery atherosclerosis [117,118,119] may lead to ischemia, as a result of distal embolization or due to the hypoperfusion of brain tissue supplied by a severely narrowed or occluded vessel [116]. Strong epidemiological evidence link VDD to the increased risk of cerebrovascular diseases, including ischemic stroke [13,120,121,122,123,124]. VDD is particularly frequent in people who have suffered stroke, which is attributed to their limited mobility, advanced age, or malnutrition (i.e., conditions leading to decreased bioavailability of VitD) [13]. In humans, VDD has also been causally linked to poor post-stroke outcome [125,126], for instance, more severe cognitive impairment [13], and it is also associated with higher risk of death at one or two years following stroke and with greater risk of early recurrent stroke [13]. Although VDD appears to increase the risk for cerebrovascular diseases [13], large Mendelian randomization studies have failed to provide evidence for causal association between 25(OH)D levels and ischemic stroke [127,128,129]. Thus, furher studies are needed to establish the beneficial effect of VitD supplementation on reducing the incidence and severity of stroke [13,120]. Preclinical studies investigating the role of VitD in the outcome of stroke also yielded controversial results. For instance, in rodent models, VDD has been reported to increase the infarction volume, exacerbate the behavioral impairment, and compromise the blood–brain barrier after cerebrovascular occlusion [102,130]. Furthermore, VitD supplementation reduced the ischemia-induced brain damage in rodent brains [131,132]. Yet, in other rodent studies, VDD did not significantly affect the extent of brain injury following ischemic stroke [133]. Thus, further experimental studies using innovative models are warranted to characterize the impact of VDD on the cerebrovascular circulation. Epidemiological evidence also links VDD to chronic brain injury associated with cerebral small vessel disease [134], cerebral cavernous malformation disease [13], and increased arterial stiffness-related cerebrovascular damage [135].
Taken together, there is strong evidence, based on epidemiological and preclinical studies, that VDD is associated with cerebrovascular diseases, including ischemic stroke. Nevertheless, further studies are needed to establish the molecular and cellular mechanisms of the beneficial effect of VitD supplementation on reducing the risk and severity of ischemic stroke and to gain further insight into the cerebrovascular actions of VitD.

7.2. Role of VDD in the Pathogenesis of Vascular Cognitive Impairment

In addition to stroke, cerebrovascular diseases also manifest with cognitive impairment. The term vascular cognitive impairment (VCI) refers to diverse forms of cognitive disorders (ranging from mild cognitive impairment to vascular dementia), which associate with various cerebrovascular pathologies, ranging from cerebral small vessel disease [136,137,138,139,140,141,142,143,144,145,146,147,148,149,150,151] to large vessel atherosclerosis and stroke [152,153,154,155]. VCI is the second most common cause of age-related cognitive impairment and dementia after Alzheimer’s disease (AD). Additionally, the pathogenesis of AD itself also involves microvascular mechanisms; therefore, it is also considered a special form of VCI by many investigators [156,157,158,159,160,161,162,163,164,165]. In recent years, increasing evidence has been causally linked VDD to the genesis of VCI [166,167] and AD [168,169,170,171,172,173,174,175,176,177] in humans. Epidemiological studies suggest a more than doubled risk of mild cognitive impairment in older adults with VDD [178,179,180,181,182]. According to human studies, VitD levels also predict structural brain alterations, reduced hippocampal volume, and disrupted connectivity [183,184]. Further, VDR gene polymorphisms are associated with cognitive decline [185,186]. In conclusion, despite epidemiological studies implying a clear association between VDD and VCI, the underlying mechanisms are yet to be investigated.
Importantly, VCI and AD are quintessential diseases of aging. Their incidence exponentially increases with advanced aging, and the fundamental cellular and molecular mechanisms of aging contribute to their pathogenesis [187,188]. Importantly, there is initial evidence that VitD contributes to the regulation of epigenetic mechanisms of aging and that VitD supplementation decelerates epigenetic aging in subjects with VDD [189,190]. Other mechanisms of aging that may be affected by VDD include sirtuin pathways [191] and cellular senescence [192]. There is also evidence supporting an association between VDD and frailty in aging [193].

7.3. Effects of VitD and VDD on Cerebrovascular Homeostasis

VitD is known to influence several physiological processes relevant to cerebrovascular and brain pathophysiology [1]. Thus, VDD may affect cerebrovascular regulation and brain homeostasis and increase the risk and worsen the severity of ischemic stroke and VCI via multiple direct and indirect mechanisms.
The direct effects of VDD by which it may increase risk of stroke include enhanced platelet aggregation, upregulation of tissue factor expression, downregulation of antithrombin and thrombomodulin expression, impaired biosynthesis of neurotrophic factors and neurotransmitters, and compromised detoxification pathways of the brain [120]. Since VDD is associated with several risk factors for stroke (Table 2), it may also increase the incidence of cerebrovascular diseases indirectly [120]. For instance, VDD is linked to hypertension [1], which is ultimately one of the major modifiable risk factors for cerebral ischemia [115]. Diabetes mellitus and insulin resistance are also associated with VDD, which may be attributed to the impaired β-cell function and insulin sensitivity of the target cells in VDD [194,195]. VDD stimulates the secretion of the parathyroid hormone; thus, it results in secondary hyperparathyroidism [196]. Since elevated parathyroid hormone levels have been found in stroke patients, an association between parathyroid hormone (PTH) levels and cerebrovascular diseases is presumable [196]. Furthermore, VDD favors inflammation [1], which may play a central role in the pathogenesis and progression of stroke [115]. In addition, VDD has been associated with subclinical carotid atherosclerosis [197]. Since atherosclerosis, particularly that of the carotid arteries, may lead to cerebral ischemia [196], VitD is likely to prevent stroke events, partly by being protective against atherosclerosis [16].
In order to provide continuous oxygen and nutrient supply for neurons, the cerebral circulation is tightly controlled by myogenic, metabolic, endothelial, and neurovascular mechanisms [73,198,199,200,201,202,203,204,205,206,207,208,209]. VitD also affects many of these critical mechanisms involved in the regulation of cerebral blood flow. The autoregulation of cerebral blood flow [210,211,212,213] ensures that cerebral blood flow remains constant, despite fluctuations in arterial blood pressure within the range of 60–150 mmHg [198,214]. However, when blood pressure is not within the limits of autoregulation, there is a risk of brain injury [215,216], including that of cerebral microhemorrhages [217,218]. The available preclinical evidence suggest that VDD may interfere with autoregulation of cerebral blood flow [219,220]. This concept is also supported by the findings that VDD impairs both cerebrovascular morphology and function in rodents [110,219,220,221]. Among others, VDD induces hypertrophic remodeling, resulting from enhanced vascular smooth muscle cell proliferation in the cerebral arteries of male rats [221]. VDD may also cause an increase in vessel tone and a decrease in endothelial relaxation capacity in the cerebral arteries of rats [221]. These alterations may result from enhanced vasoconstrictor prostanoid production, due to increased COX-2 expression and from decreased eNOS expression, leading to diminished NO production in cerebral vessels (Figure 3) [221,222,223,224]. Functional impairment and pathological remodeling of the cerebral vessels in VDD compromises cerebrovascular adaptation to ischemic events [110], increasing the risk for ischemic stroke and/or worsening its outcome [225,226,227,228,229,230]. Interestingly, in preclinical models, females appear to be better protected from the cerebrovascular effects of VDD than males; therefore, sex differences should be considered in clinical studies investigating the cerebrovascular manifestations of VDD, as well [219,220].
The critical neurovascular mechanisms by which functional and phenotypic alterations of the cerebral microcirculation [231,232,233] may promote the pathogenesis of VCI in older adults include cerebral microvascular rarefaction [234], impairment of neurovascular coupling responses (also known as functional hyperemia, which is responsible for moment-to-moment adjustment of cerebral blood flow to increased oxygen and nutrient demand of activated neurons [235,236,237,238,239,240]), and disruption of the blood–brain barrier [214,241,242,243,244,245,246,247,248]. VDR is known to be expressed on each cell type within the neurovascular unit, including endothelial cells, smooth muscle cells, pericytes [249], astrocytes, and neurons [250]. Thus, it is expected that VDD may affect the diverse physiological processes mediated by these cells. VDD was reported to aggravate capillary rarefaction in the kidney [251], whereas VitD was able to increase capillary density in the heart [252]. Yet, the effects of VitD and VDD on capillary density in the brain remains to be elucidated. There is promising preclinical evidence that VitD can improve blood–brain barrier integrity in various pathophysiological conditions [253,254,255]. Neurovascular coupling responses are, at least in part, mediated by endothelium-derived NO [256,257]. It is important, in that regard, that VDD was shown to improve endothelial function in vessels of the systemic circulation (see above). Thus, translational studies are warranted to explore the link among VDD, impaired neurovascular coupling responses, and impaired cognitive performance in older adults. The tone of cerebral resistance arterioles is also influenced by arterial partial pressure of carbon dioxide (pCO2) and, to a lesser extent, by partial pressure of oxygen (pO2) [198]. Importantly, hypoxia and hypercapnia promote the release of vasoactive mediators, including NO, from cerebral vessels, and the subsequent vasodilation increases the blood flow and, thus, tissue oxygenation [198]. It can be expected that changes in endothelial function due to VDD or VitD therapy would also affect the aforementioned vasoregulatory mechanisms. This hypothesis needs to be tested experimentally. Since the central nervous system is highly vulnerable to changes in cerebral blood flow [200], the synergistic impairment of multiple cerebral vascular homeostatic mechanisms is likely to exacerbate neurological disorders and promote cognitive decline [115,215].

8. Conclusions

Several human observational and animal experimental studies imply that VDD favors the development, worsens the outcome of cerebrovascular disorders, and promotes the genesis of cognitive decline. Therefore, the understanding of the mechanisms underlying the beneficial effect of VitD supplementation on reducing the incidence and severity of stroke, as well as the prevention of VCI, warrants further studies. Several questions remain to be answered. For instance, how do the time of onset and duration of VDD influence its cerebrovascular consequences? Are the cerebrovascular consequences of VDD reversible? Do cerebrovascular and cognitive outcomes correlate in clinical studies investigating the effects of VitD [258,259,260]? What is the mechanism underlying the sex-dependence of the cerebrovascular manifestation of VDD? How does VitD modulate the cellular mechanisms of aging in cells of the neurovascular unit? How do the effects of VitD on pathogenic mechanisms underlying VCI and AD differ [261,262]? How do combination treatments [263] consisting of VitD and specific interventions targeting cellular mechanisms of aging affect cerebrovascular and brain pathologies?
The high incidence of cardiovascular and cerebrovascular diseases worldwide, especially in advanced age, highlights the importance of investigating the roles and mechanisms of modifiable cardiovascular risk factors, including VDD, in the pathogenesis of vascular disorders. Although strong evidence links VDD to cerebrovascular diseases, the understanding of the complete mechanism underlying the impact of VDD on the development and severity of ischemic stroke and VCI warrants further well-designed experimental, epidemiological, and clinical studies.

Author Contributions

Conceptualization, É.P., Z.U., S.V. and Z.B.; writing—original draft preparation, É.P., Z.U., Z.B. and S.V.; writing—review and editing, É.P., Z.U., Z.B. and S.V.; visualization, É.P.; supervision, Z.B. and S.V. All authors have read and agreed to the published version of the manuscript.

Funding

Authors’ original research summarized in the present review was supported by the Hungarian National Research, Development, and Innovation Office (OTKA K-125174, K-139230, and PD-143327), by the Ministry of Innovation and Technology of Hungary from the NRDI Fund (ÚNKP-22-4-II-SE-17, 2020-1.1.6-JÖVŐ-2021-00010, 2020-1.1.6-JÖVŐ-2021-00013; TKP2021-NKTA-47, RRF-2.3.1-21-2022-00003, and TKP2021-EGA-25), by the Semmelweis Science and Innovation Fund (STIA-KFI-2021 and STIA-OTKA-2021), the European University for Well-Being (EUniWell) program (grant agreement number: 101004093/EUniWell/EAC-A02-2019/EAC-A02-2019-1), and by the Dean of the Faculty of Medicine, Semmelweis University. ZU was supported by grants from the National Institute on Aging (R01AG055395, R01AG068295; R01AG070915), the National Institute of Neurological Disorders and Stroke (R01NS100782), and the National Cancer Institute (R01CA255840). The Central Library of Semmelweis University also kindly provided fund for open access publication fees.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors are grateful to Erzsébet Fejes for critically reading the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Norman, P.E.; Powell, J.T. Vitamin D and cardiovascular disease. Circ. Res. 2014, 114, 379–393. [Google Scholar] [CrossRef] [PubMed]
  2. Holick, M.F. Vitamin D deficiency. N. Engl. J. Med. 2007, 357, 266–281. [Google Scholar] [CrossRef] [PubMed]
  3. Lips, P. Vitamin D deficiency and secondary hyperparathyroidism in the elderly: Consequences for bone loss and fractures and therapeutic implications. Endocr. Rev. 2001, 22, 477–501. [Google Scholar] [CrossRef]
  4. Cui, A.; Xiao, P.; Ma, Y.; Fan, Z.; Zhou, F.; Zheng, J.; Zhang, L. Prevalence, trend, and predictor analyses of vitamin D deficiency in the US population, 2001–2018. Front. Nutr. 2022, 9, 965376. [Google Scholar] [CrossRef]
  5. da Silveira, E.A.; Moura, L.; Castro, M.C.R.; Kac, G.; Hadler, M.; Noll, P.; Noll, M.; Rezende, A.T.O.; Delpino, F.M.; Oliveira, C. Prevalence of Vitamin D and Calcium Deficiency and Insufficiency in Women of Childbearing Age and Associated Risk Factors: A Systematic Review and Meta-Analysis. Nutrients 2022, 14, 4351. [Google Scholar] [CrossRef] [PubMed]
  6. Ravinder, S.S.; Padmavathi, R.; Maheshkumar, K.; Mohankumar, M.; Maruthy, K.N.; Sankar, S.; Balakrishnan, K. Prevalence of vitamin D deficiency among South Indian pregnant women. J. Fam. Med. Prim. Care 2022, 11, 2884–2889. [Google Scholar]
  7. van der Meer, I.M.; Middelkoop, B.J.; Boeke, A.J.; Lips, P. Prevalence of vitamin D deficiency among Turkish, Moroccan, Indian and sub-Sahara African populations in Europe and their countries of origin: An overview. Osteoporos. Int. 2011, 22, 1009–1021. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Bakaloudi, D.R.; Chourdakis, M. A critical update on the role of mild and serious vitamin D deficiency prevalence and the COVID-19 epidemic in Europe. Nutrition 2022, 93, 111441. [Google Scholar] [CrossRef]
  9. Grober, U.; Reichrath, J.; Holick, M.F. Live longer with vitamin D? Nutrients 2015, 7, 1871–1880. [Google Scholar] [CrossRef] [Green Version]
  10. Holick, M.F.; Binkley, N.C.; Bischoff-Ferrari, H.A.; Gordon, C.M.; Hanley, D.A.; Heaney, R.P.; Murad, M.H.; Weaver, C.M. Evaluation, treatment, and prevention of vitamin D deficiency: An Endocrine Society clinical practice guideline. J. Clin. Endocrinol. Metab. 2011, 96, 1911–1930. [Google Scholar] [CrossRef] [Green Version]
  11. Hossein-nezhad, A.; Holick, M.F. Vitamin D for health: A global perspective. Mayo Clin. Proc. 2013, 88, 720–755. [Google Scholar] [CrossRef] [PubMed]
  12. Cosentino, N.; Campodonico, J.; Milazzo, V.; De Metrio, M.; Brambilla, M.; Camera, M.; Marenzi, G. Vitamin D and Cardiovascular Disease: Current Evidence and Future Perspectives. Nutrients 2021, 13, 3603. [Google Scholar] [CrossRef] [PubMed]
  13. Kim, H.A.; Perrelli, A.; Ragni, A.; Retta, F.; De Silva, T.M.; Sobey, C.G.; Retta, S.F. Vitamin D Deficiency and the Risk of Cerebrovascular Disease. Antioxidants 2020, 9, 327. [Google Scholar] [CrossRef] [PubMed]
  14. North, B.J.; Sinclair, D.A. The intersection between aging and cardiovascular disease. Circ. Res. 2012, 110, 1097–1108. [Google Scholar] [CrossRef] [PubMed]
  15. Lenz, I.J.; Plesnila, N.; Terpolilli, N.A. Role of endothelial nitric oxide synthase for early brain injury after subarachnoid hemorrhage in mice. J. Cereb. Blood Flow Metab. 2021, 41, 1669–1681. [Google Scholar] [CrossRef]
  16. Menezes, A.R.; Lamb, M.C.; Lavie, C.J.; DiNicolantonio, J.J. Vitamin D and atherosclerosis. Curr. Opin. Cardiol. 2014, 29, 571–577. [Google Scholar] [CrossRef]
  17. Sun, H.J.; Wu, Z.Y.; Nie, X.W.; Bian, J.S. Role of Endothelial Dysfunction in Cardiovascular Diseases: The Link Between Inflammation and Hydrogen Sulfide. Front. Pharmacol. 2019, 10, 1568. [Google Scholar] [CrossRef] [Green Version]
  18. de la Guía-Galipienso, F.; Martínez-Ferran, M.; Vallecillo, N.; Lavie, C.J.; Sanchis-Gomar, F.; Pareja-Galeano, H. Vitamin D and cardiovascular health. Clin. Nutr. 2021, 40, 2946–2957. [Google Scholar] [CrossRef] [PubMed]
  19. Bikle, D.D. Vitamin D metabolism, mechanism of action, and clinical applications. Chem. Biol. 2014, 21, 319–329. [Google Scholar] [CrossRef] [Green Version]
  20. Christakos, S.; Dhawan, P.; Verstuyf, A.; Verlinden, L.; Carmeliet, G. Vitamin D: Metabolism, Molecular Mechanism of Action, and Pleiotropic Effects. Physiol. Rev. 2016, 96, 365–408. [Google Scholar] [CrossRef] [Green Version]
  21. Prosser, D.E.; Jones, G. Enzymes involved in the activation and inactivation of vitamin D. Trends Biochem. Sci. 2004, 29, 664–673. [Google Scholar] [CrossRef] [PubMed]
  22. Richart, T.; Li, Y.; Staessen, J.A. Renal versus extrarenal activation of vitamin D in relation to atherosclerosis, arterial stiffening, and hypertension. Am. J. Hypertens. 2007, 20, 1007–1015. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Ferder, M.; Inserra, F.; Manucha, W.; Ferder, L. The world pandemic of vitamin D deficiency could possibly be explained by cellular inflammatory response activity induced by the renin-angiotensin system. Am. J. Physiol. Cell Physiol. 2013, 304, C1027–C1039. [Google Scholar] [CrossRef] [Green Version]
  24. Brumbaugh, P.F.; Haussler, M.R. Specific binding of 1alpha,25-dihydroxycholecalciferol to nuclear components of chick intestine. J. Biol. Chem. 1975, 250, 1588–1594. [Google Scholar] [CrossRef] [PubMed]
  25. Rosen, C.J.; Adams, J.S.; Bikle, D.D.; Black, D.M.; Demay, M.B.; Manson, J.E.; Murad, M.H.; Kovacs, C.S. The nonskeletal effects of vitamin D: An Endocrine Society scientific statement. Endocr. Rev. 2012, 33, 456–492. [Google Scholar] [PubMed] [Green Version]
  26. Pike, J.W.; Meyer, M.B.; Benkusky, N.A.; Lee, S.M.; St John, H.; Carlson, A.; Onal, M.; Shamsuzzaman, S. Genomic Determinants of Vitamin D-Regulated Gene Expression. Vitam. Horm. 2016, 100, 21–44. [Google Scholar] [PubMed] [Green Version]
  27. Jamali, N.; Sorenson, C.M.; Sheibani, N. Vitamin D and regulation of vascular cell function. Am. J. Physiol. Heart Circ. Physiol. 2018, 314, H753–H765. [Google Scholar] [CrossRef] [Green Version]
  28. Hii, C.S.; Ferrante, A. The Non-Genomic Actions of Vitamin D. Nutrients 2016, 8, 135. [Google Scholar] [CrossRef] [Green Version]
  29. Martins, D.; Wolf, M.; Pan, D.; Zadshir, A.; Tareen, N.; Thadhani, R.; Felsenfeld, A.; Levine, B.; Mehrotra, R.; Norris, K. Prevalence of cardiovascular risk factors and the serum levels of 25-hydroxyvitamin D in the United States: Data from the Third National Health and Nutrition Examination Survey. Arch. Intern. Med. 2007, 167, 1159–1165. [Google Scholar] [CrossRef] [Green Version]
  30. 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]
  31. Bouillon, R.; Carmeliet, G.; Verlinden, L.; van Etten, E.; Verstuyf, A.; Luderer, H.F.; Lieben, L.; Mathieu, C.; Demay, M. Vitamin D and human health: Lessons from vitamin D receptor null mice. Endocr. Rev. 2008, 29, 726–776. [Google Scholar] [PubMed] [Green Version]
  32. Mozos, I.; Marginean, O. Links between Vitamin D Deficiency and Cardiovascular Diseases. BioMed Res. Int. 2015, 2015, 109275. [Google Scholar] [CrossRef] [PubMed]
  33. Wu-Wong, J.R.; Nakane, M.; Ma, J.; Ruan, X.; Kroeger, P.E. Effects of Vitamin D analogs on gene expression profiling in human coronary artery smooth muscle cells. Atherosclerosis 2006, 186, 20–28. [Google Scholar] [CrossRef] [PubMed]
  34. Bouillon, R. Vitamin D and cardiovascular disorders. Osteoporos. Int. 2019, 30, 2167–2181. [Google Scholar] [CrossRef] [PubMed]
  35. Wu-Wong, J.R.; Nakane, M.; Ma, J.; Ruan, X.; Kroeger, P.E. VDR-mediated gene expression patterns in resting human coronary artery smooth muscle cells. J. Cell. Biochem. 2007, 100, 1395–1405. [Google Scholar] [CrossRef]
  36. Grundmann, M.; Haidar, M.; Placzko, S.; Niendorf, R.; Darashchonak, N.; Hubel, C.A.; von Versen-Höynck, F. Vitamin D improves the angiogenic properties of endothelial progenitor cells. Am. J. Physiol. Cell Physiol. 2012, 303, C954–C962. [Google Scholar] [CrossRef]
  37. Andrukhova, O.; Slavic, S.; Zeitz, U.; Riesen, S.C.; Heppelmann, M.S.; Ambrisko, T.D.; Markovic, M.; Kuebler, W.M.; Erben, R.G. Vitamin D is a regulator of endothelial nitric oxide synthase and arterial stiffness in mice. Mol. Endocrinol. 2014, 28, 53–64. [Google Scholar] [CrossRef] [Green Version]
  38. 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]
  39. Marcinowska-Suchowierska, E.; Kupisz-Urbańska, M.; Łukaszkiewicz, J.; Płudowski, P.; Jones, G. Vitamin D Toxicity-A Clinical Perspective. Front. Endocrinol. 2018, 9, 550. [Google Scholar] [CrossRef] [Green Version]
  40. Zittermann, A.; Iodice, S.; Pilz, S.; Grant, W.B.; Bagnardi, V.; Gandini, S. Vitamin D deficiency and mortality risk in the general population: A meta-analysis of prospective cohort studies. Am. J. Clin. Nutr. 2012, 95, 91–100. [Google Scholar] [CrossRef] [Green Version]
  41. Zittermann, A.; Pilz, S. Vitamin D and Cardiovascular Disease: An Update. Anticancer Res. 2019, 39, 4627–4635. [Google Scholar] [CrossRef] [PubMed]
  42. DeLuca, H.F. Overview of general physiologic features and functions of vitamin D. Am. J. Clin. Nutr. 2004, 80, 1689s–1696s. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Chen, S.; Law, C.S.; Grigsby, C.L.; Olsen, K.; Hong, T.T.; Zhang, Y.; Yeghiazarians, Y.; Gardner, D.G. Cardiomyocyte-specific deletion of the vitamin D receptor gene results in cardiac hypertrophy. Circulation 2011, 124, 1838–1847. [Google Scholar] [CrossRef] [Green Version]
  44. Somjen, D.; Weisman, Y.; Kohen, F.; Gayer, B.; Limor, R.; Sharon, O.; Jaccard, N.; Knoll, E.; Stern, N. 25-hydroxyvitamin D3-1alpha-hydroxylase is expressed in human vascular smooth muscle cells and is upregulated by parathyroid hormone and estrogenic compounds. Circulation 2005, 111, 1666–1671. [Google Scholar] [CrossRef] [Green Version]
  45. Merke, J.; Milde, P.; Lewicka, S.; Hugel, U.; Klaus, G.; Mangelsdorf, D.J.; Haussler, M.R.; Rauterberg, E.W.; Ritz, E. Identification and regulation of 1,25-dihydroxyvitamin D3 receptor activity and biosynthesis of 1,25-dihydroxyvitamin D3. Studies in cultured bovine aortic endothelial cells and human dermal capillaries. J. Clin. Investig. 1989, 83, 1903–1915. [Google Scholar] [CrossRef] [PubMed]
  46. Guillot, X.; Semerano, L.; Saidenberg-Kermanac’h, N.; Falgarone, G.; Boissier, M.C. Vitamin D and inflammation. Joint Bone Spine 2010, 77, 552–557. [Google Scholar] [CrossRef]
  47. Silvagno, F.; De Vivo, E.; Attanasio, A.; Gallo, V.; Mazzucco, G.; Pescarmona, G. Mitochondrial localization of vitamin D receptor in human platelets and differentiated megakaryocytes. PLoS ONE 2010, 5, e8670. [Google Scholar] [CrossRef] [Green Version]
  48. Aihara, K.; Azuma, H.; Akaike, M.; Ikeda, Y.; Yamashita, M.; Sudo, T.; Hayashi, H.; Yamada, Y.; Endoh, F.; Fujimura, M.; et al. Disruption of nuclear vitamin D receptor gene causes enhanced thrombogenicity in mice. J. Biol. Chem. 2004, 279, 35798–35802. [Google Scholar] [CrossRef] [Green Version]
  49. Mehta, V.; Agarwal, S. Does Vitamin D Deficiency Lead to Hypertension? Cureus 2017, 9, e1038. [Google Scholar] [CrossRef] [Green Version]
  50. Black, L.J.; Burrows, S.; Lucas, R.M.; Marshall, C.E.; Huang, R.C.; Chan She Ping-Delfos, W.; Beilin, L.J.; Holt, P.G.; Hart, P.H.; Oddy, W.H.; et al. Serum 25-hydroxyvitamin D concentrations and cardiometabolic risk factors in adolescents and young adults. Br. J. Nutr. 2016, 115, 1994–2002. [Google Scholar] [CrossRef] [Green Version]
  51. Muhairi, S.J.; Mehairi, A.E.; Khouri, A.A.; Naqbi, M.M.; Maskari, F.A.; Al Kaabi, J.; Al Dhaheri, A.S.; Nagelkerke, N.; Shah, S.M. Vitamin D deficiency among healthy adolescents in Al Ain, United Arab Emirates. BMC Public Health 2013, 13, 33. [Google Scholar] [CrossRef] [PubMed]
  52. Li, Y.C.; Kong, J.; Wei, M.; Chen, Z.F.; Liu, S.Q.; Cao, L.P. 1,25-Dihydroxyvitamin D(3) is a negative endocrine regulator of the renin-angiotensin system. J. Clin. Investig. 2002, 110, 229–238. [Google Scholar] [CrossRef] [PubMed]
  53. Pilz, S.; Tomaschitz, A.; Drechsler, C.; Dekker, J.M.; Marz, W. Vitamin D deficiency and myocardial diseases. Mol. Nutr. Food Res. 2010, 54, 1103–1113. [Google Scholar] [CrossRef] [PubMed]
  54. Arfian, N.; Kusuma, M.H.; Anggorowati, N.; Nugroho, D.B.; Jeffilano, A.; Suzuki, Y.; Ikeda, K.; Emoto, N. Vitamin D upregulates endothelin-1, ETBR, eNOS mRNA expression and attenuates vascular remodelling and ischemia in kidney fibrosis model in mice. Physiol. Res. 2018, 67, S137–S147. [Google Scholar] [CrossRef] [PubMed]
  55. Enkhjargal, B.; Malaguit, J.; Ho, W.M.; Jiang, W.; Wan, W.; Wang, G.; Tang, J.; Zhang, J.H. Vitamin D attenuates cerebral artery remodeling through VDR/AMPK/eNOS dimer phosphorylation pathway after subarachnoid hemorrhage in rats. J. Cereb. Blood Flow Metab. 2019, 39, 272–284. [Google Scholar] [CrossRef] [PubMed]
  56. Hadjadj, L.; Monori-Kiss, A.; Horvath, E.M.; Heinzlmann, A.; Magyar, A.; Sziva, R.E.; Miklos, Z.; Pal, E.; Gal, J.; Szabo, I.; et al. Geometric, elastic and contractile-relaxation changes in coronary arterioles induced by Vitamin D deficiency in normal and hyperandrogenic female rats. Microvasc. Res. 2019, 122, 78–84. [Google Scholar] [CrossRef]
  57. Cardus, A.; Parisi, E.; Gallego, C.; Aldea, M.; Fernandez, E.; Valdivielso, J.M. 1,25-Dihydroxyvitamin D3 stimulates vascular smooth muscle cell proliferation through a VEGF-mediated pathway. Kidney Int. 2006, 69, 1377–1384. [Google Scholar] [CrossRef] [Green Version]
  58. Mitsuhashi, T.; Morris, R.C., Jr.; Ives, H.E. 1,25-dihydroxyvitamin D3 modulates growth of vascular smooth muscle cells. J. Clin. Investig. 1991, 87, 1889–1895. [Google Scholar] [CrossRef] [PubMed]
  59. Carthy, E.P.; Yamashita, W.; Hsu, A.; Ooi, B.S. 1,25-Dihydroxyvitamin D3 and rat vascular smooth muscle cell growth. Hypertension 1989, 13, 954–959. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. 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] [Green Version]
  61. Ferrara, N. Vascular endothelial growth factor: Basic science and clinical progress. Endocr. Rev. 2004, 25, 581–611. [Google Scholar] [PubMed]
  62. Chow, A.K.; Cena, J.; Schulz, R. Acute actions and novel targets of matrix metalloproteinases in the heart and vasculature. Br. J. Pharmacol. 2007, 152, 189–205. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. 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. 2012, 27, 1800–1806. [Google Scholar] [CrossRef] [Green Version]
  64. Zhong, W.; Gu, B.; Gu, Y.; Groome, L.J.; Sun, J.; Wang, Y. Activation of vitamin D receptor promotes VEGF and CuZn-SOD expression in endothelial cells. J. Steroid Biochem. Mol. Biol. 2014, 140, 56–62. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Ben-Shoshan, M.; Amir, S.; Dang, D.T.; Dang, L.H.; Weisman, Y.; Mabjeesh, N.J. 1alpha,25-dihydroxyvitamin D3 (Calcitriol) inhibits hypoxia-inducible factor-1/vascular endothelial growth factor pathway in human cancer cells. Mol. Cancer Ther. 2007, 6, 1433–1439. [Google Scholar] [CrossRef] [Green Version]
  66. Krishna, S.M. Vitamin D as A Protector of Arterial Health: Potential Role in Peripheral Arterial Disease Formation. Int. J. Mol. Sci. 2019, 20, 4907. [Google Scholar] [CrossRef] [Green Version]
  67. Ye, B.; Weng, Y.; Lin, S.; Lin, J.; Huang, Z.; Huang, W.; Cai, X. 1,25(OH)2D3 Strengthens the Vasculogenesis of Multipotent Mesenchymal Stromal Cells from Rat Bone Marrow by Regulating the PI3K/AKT Pathway. Drug Des. Devel. Ther. 2020, 14, 1157–1167. [Google Scholar] [CrossRef] [Green Version]
  68. Shadwick, R.E. Mechanical design in arteries. J. Exp. Biol. 1999, 202, 3305–3313. [Google Scholar] [CrossRef]
  69. Salum, E.; Kampus, P.; Zilmer, M.; Eha, J.; Butlin, M.; Avolio, A.P.; Podramagi, T.; Arend, A.; Aunapuu, M.; Kals, J. Effect of vitamin D on aortic remodeling in streptozotocin-induced diabetes. Cardiovasc. Diabetol. 2012, 11, 58. [Google Scholar] [CrossRef] [Green Version]
  70. Austin, S.A.; Katusic, Z.S. Partial loss of endothelial nitric oxide leads to increased cerebrovascular beta amyloid. J. Cereb. Blood Flow Metab. 2020, 40, 392–403. [Google Scholar] [CrossRef]
  71. Liu, W.; Chen, Z.; Ortega, D.; Liu, X.; Huang, X.; Wang, L.; Chen, L.; Sun, J.; Hatsukami, T.S.; Yuan, C.; et al. Arterial elasticity, endothelial function and intracranial vascular health: A multimodal MRI study. J. Cereb. Blood Flow Metab. 2021, 41, 1390–1397. [Google Scholar] [CrossRef]
  72. Pradillo, J.M.; Hernandez-Jimenez, M.; Fernandez-Valle, M.E.; Medina, V.; Ortuno, J.E.; Allan, S.M.; Proctor, S.D.; Garcia-Segura, J.M.; Ledesma-Carbayo, M.J.; Santos, A.; et al. Influence of metabolic syndrome on post-stroke outcome, angiogenesis and vascular function in old rats determined by dynamic contrast enhanced MRI. J. Cereb. Blood Flow Metab. 2021, 41, 1692–1706. [Google Scholar] [CrossRef] [PubMed]
  73. Trigiani, L.J.; Bourourou, M.; Lacalle-Aurioles, M.; Lecrux, C.; Hynes, A.; Spring, S.; Fernandes, D.J.; Sled, J.G.; Lesage, F.; Schwaninger, M.; et al. A functional cerebral endothelium is necessary to protect against cognitive decline. J. Cereb. Blood Flow Metab. 2022, 42, 74–89. [Google Scholar] [CrossRef] [PubMed]
  74. Zhang, W.; Davis, C.M.; Zeppenfeld, D.M.; Golgotiu, K.; Wang, M.X.; Haveliwala, M.; Hong, D.; Li, Y.; Wang, R.K.; Iliff, J.J.; et al. Role of endothelium-pericyte signaling in capillary blood flow response to neuronal activity. J. Cereb. Blood Flow Metab. 2021, 41, 1873–1885. [Google Scholar] [CrossRef] [PubMed]
  75. Ni, W.; Watts, S.W.; Ng, M.; Chen, S.; Glenn, D.J.; Gardner, D.G. Elimination of vitamin D receptor in vascular endothelial cells alters vascular function. Hypertension 2014, 64, 1290–1298. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Martinez-Miguel, P.; Valdivielso, J.M.; Medrano-Andres, D.; Roman-Garcia, P.; Cano-Penalver, J.L.; Rodriguez-Puyol, M.; Rodriguez-Puyol, D.; Lopez-Ongil, S. The active form of vitamin D, calcitriol, induces a complex dual upregulation of endothelin and nitric oxide in cultured endothelial cells. Am. J. Physiol. Endocrinol. Metab. 2014, 307, E1085–E1096. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Hirata, M.; Serizawa, K.; Aizawa, K.; Yogo, K.; Tashiro, Y.; Takeda, S.; Moriguchi, Y.; Endo, K.; Fukagawa, M. 22-Oxacalcitriol prevents progression of endothelial dysfunction through antioxidative effects in rats with type 2 diabetes and early-stage nephropathy. Nephrol. Dial. Transplant. 2013, 28, 1166–1174. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Molinari, C.; Uberti, F.; Grossini, E.; Vacca, G.; Carda, S.; Invernizzi, M.; Cisari, C. 1alpha,25-dihydroxycholecalciferol induces nitric oxide production in cultured endothelial cells. Cell. Physiol. Biochem. 2011, 27, 661–668. [Google Scholar] [CrossRef]
  79. Dudzinski, D.M.; Michel, T. Life history of eNOS: Partners and pathways. Cardiovasc. Res. 2007, 75, 247–260. [Google Scholar] [CrossRef] [PubMed]
  80. Masszi, G.; Benko, R.; Csibi, N.; Horvath, E.M.; Tokes, A.M.; Novak, A.; Beres, N.J.; Tarszabo, R.; Buday, A.; Repas, C.; et al. Endothelial relaxation mechanisms and nitrative stress are partly restored by Vitamin D3 therapy in a rat model of polycystic ovary syndrome. Life Sci. 2013, 93, 133–138. [Google Scholar] [CrossRef] [Green Version]
  81. Masszi, G.; Novak, A.; Tarszabo, R.; Horvath, E.M.; Buday, A.; Ruisanchez, E.; Tokes, A.M.; Sara, L.; Benko, R.; Nadasy, G.L.; et al. Effects of vitamin D3 derivative--calcitriol on pharmacological reactivity of aortic rings in a rodent PCOS model. Pharmacol. Rep. 2013, 65, 476–483. [Google Scholar] [CrossRef] [PubMed]
  82. Tare, M.; Emmett, S.J.; Coleman, H.A.; Skordilis, C.; Eyles, D.W.; Morley, R.; Parkington, H.C. Vitamin D insufficiency is associated with impaired vascular endothelial and smooth muscle function and hypertension in young rats. J. Physiol. 2011, 589, 4777–4786. [Google Scholar] [CrossRef] [PubMed]
  83. Incalza, M.A.; D’Oria, R.; Natalicchio, A.; Perrini, S.; Laviola, L.; Giorgino, F. Oxidative stress and reactive oxygen species in endothelial dysfunction associated with cardiovascular and metabolic diseases. Vasc. Pharmacol. 2018, 100, 1–19. [Google Scholar]
  84. Kim, D.H.; Meza, C.A.; Clarke, H.; Kim, J.S.; Hickner, R.C. Vitamin D and Endothelial Function. Nutrients 2020, 12, 575. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Argacha, J.F.; Egrise, D.; Pochet, S.; Fontaine, D.; Lefort, A.; Libert, F.; Goldman, S.; van de Borne, P.; Berkenboom, G.; Moreno-Reyes, R. Vitamin D deficiency-induced hypertension is associated with vascular oxidative stress and altered heart gene expression. J. Cardiovasc. Pharmacol. 2011, 58, 65–71. [Google Scholar] [CrossRef] [PubMed]
  86. Pelham, C.J.; Drews, E.M.; Agrawal, D.K. Vitamin D controls resistance artery function through regulation of perivascular adipose tissue hypoxia and inflammation. J. Mol. Cell. Cardiol. 2016, 98, 1–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Dong, J.; Wong, S.L.; Lau, C.W.; Lee, H.K.; Ng, C.F.; Zhang, L.; Yao, X.; Chen, Z.Y.; Vanhoutte, P.M.; Huang, Y. Calcitriol protects renovascular function in hypertension by down-regulating angiotensin II type 1 receptors and reducing oxidative stress. Eur. Heart J. 2012, 33, 2980–2990. [Google Scholar] [CrossRef] [Green Version]
  88. Velimirovic, M.; Jevtic Dozudic, G.; Selakovic, V.; Stojkovic, T.; Puskas, N.; Zaletel, I.; Zivkovic, M.; Dragutinovic, V.; Nikolic, T.; Jelenkovic, A.; et al. Effects of Vitamin D3 on the NADPH Oxidase and Matrix Metalloproteinase 9 in an Animal Model of Global Cerebral Ischemia. Oxidative Med. Cell. Longev. 2018, 2018, 3273654. [Google Scholar] [CrossRef] [Green Version]
  89. Laursen, J.B.; Somers, M.; Kurz, S.; McCann, L.; Warnholtz, A.; Freeman, B.A.; Tarpey, M.; Fukai, T.; Harrison, D.G. Endothelial regulation of vasomotion in apoE-deficient mice: Implications for interactions between peroxynitrite and tetrahydrobiopterin. Circulation 2001, 103, 1282–1288. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Husain, K.; Suarez, E.; Isidro, A.; Ferder, L. Effects of paricalcitol and enalapril on atherosclerotic injury in mouse aortas. Am. J. Nephrol. 2010, 32, 296–304. [Google Scholar] [CrossRef]
  91. Dong, J.; Wong, S.L.; Lau, C.W.; Liu, J.; Wang, Y.X.; Dan He, Z.; Fai Ng, C.; Yu Chen, Z.; Yao, X.; Xu, A.; et al. Calcitriol restores renovascular function in estrogen-deficient rats through downregulation of cyclooxygenase-2 and the thromboxane-prostanoid receptor. Kidney Int. 2013, 84, 54–63. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Suzuki, Y.; Ichiyama, T.; Ohsaki, A.; Hasegawa, S.; Shiraishi, M.; Furukawa, S. Anti-inflammatory effect of 1alpha,25-dihydroxyvitamin D(3) in human coronary arterial endothelial cells: Implication for the treatment of Kawasaki disease. J. Steroid Biochem. Mol. Biol. 2009, 113, 134–138. [Google Scholar] [CrossRef]
  93. Jablonski, K.L.; Chonchol, M.; Pierce, G.L.; Walker, A.E.; Seals, D.R. 25-Hydroxyvitamin D deficiency is associated with inflammation-linked vascular endothelial dysfunction in middle-aged and older adults. Hypertension 2011, 57, 63–69. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Chun, R.F.; Liu, P.T.; Modlin, R.L.; Adams, J.S.; Hewison, M. Impact of vitamin D on immune function: Lessons learned from genome-wide analysis. Front. Physiol. 2014, 5, 151. [Google Scholar] [PubMed] [Green Version]
  95. Takeda, M.; Yamashita, T.; Sasaki, N.; Nakajima, K.; Kita, T.; Shinohara, M.; Ishida, T.; Hirata, K. Oral administration of an active form of vitamin D3 (calcitriol) decreases atherosclerosis in mice by inducing regulatory T cells and immature dendritic cells with tolerogenic functions. Arterioscler. Thromb. Vasc. Biol. 2010, 30, 2495–2503. [Google Scholar] [CrossRef] [Green Version]
  96. Wong, M.S.; Delansorne, R.; Man, R.Y.; Vanhoutte, P.M. Vitamin D derivatives acutely reduce endothelium-dependent contractions in the aorta of the spontaneously hypertensive rat. Am. J. Physiol. Heart Circ. Physiol. 2008, 295, H289–H296. [Google Scholar] [CrossRef] [PubMed]
  97. Wong, M.S.; Delansorne, R.; Man, R.Y.; Svenningsen, P.; Vanhoutte, P.M. Chronic treatment with vitamin D lowers arterial blood pressure and reduces endothelium-dependent contractions in the aorta of the spontaneously hypertensive rat. Am. J. Physiol. Heart Circ. Physiol. 2010, 299, H1226–H1234. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Borges, A.C.; Feres, T.; Vianna, L.M.; Paiva, T.B. Recovery of impaired K+ channels in mesenteric arteries from spontaneously hypertensive rats by prolonged treatment with cholecalciferol. Br. J. Pharmacol. 1999, 127, 772–778. [Google Scholar] [CrossRef] [Green Version]
  99. Chen, H.; Lu, R.; Zhang, Y.G.; Sun, J. Vitamin D Receptor Deletion Leads to the Destruction of Tight and Adherens Junctions in Lungs. Tissue Barriers 2018, 6, 1–13. [Google Scholar] [CrossRef] [Green Version]
  100. Lee, A.S.; Jung, Y.J.; Thanh, T.N.; Lee, S.; Kim, W.; Kang, K.P.; Park, S.K. Paricalcitol attenuates lipopolysaccharide-induced myocardial inflammation by regulating the NF-κB signaling pathway. Int. J. Mol. Med. 2016, 37, 1023–1029. [Google Scholar] [CrossRef] [Green Version]
  101. Parekh, D.; Dancer, R.C.A.; Scott, A.; D’Souza, V.K.; Howells, P.A.; Mahida, R.Y.; Tang, J.C.Y.; Cooper, M.S.; Fraser, W.D.; Tan, L.; et al. Vitamin D to Prevent Lung Injury following Esophagectomy-A Randomized, Placebo-Controlled Trial. Crit. Care Med. 2018, 46, e1128–e1135. [Google Scholar] [CrossRef] [PubMed]
  102. Sayeed, I.; Turan, N.; Stein, D.G.; Wali, B. Vitamin D deficiency increases blood-brain barrier dysfunction after ischemic stroke in male rats. Exp. Neurol. 2019, 312, 63–71. [Google Scholar] [CrossRef] [PubMed]
  103. Vila Cuenca, M.; Ferrantelli, E.; Meinster, E.; Pouw, S.M.; Kovačević, I.; de Menezes, R.X.; Niessen, H.W.; Beelen, R.H.J.; Hordijk, P.L.; Vervloet, M.G. Vitamin D Attenuates Endothelial Dysfunction in Uremic Rats and Maintains Human Endothelial Stability. J. Am. Heart Assoc. 2018, 7, e008776. [Google Scholar] [CrossRef]
  104. Gibson, C.C.; Zhu, W.; Davis, C.T.; Bowman-Kirigin, J.A.; Chan, A.C.; Ling, J.; Walker, A.E.; Goitre, L.; Delle Monache, S.; Retta, S.F.; et al. Strategy for identifying repurposed drugs for the treatment of cerebral cavernous malformation. Circulation 2015, 131, 289–299. [Google Scholar] [CrossRef]
  105. Gibson, C.C.; Davis, C.T.; Zhu, W.; Bowman-Kirigin, J.A.; Walker, A.E.; Tai, Z.; Thomas, K.R.; Donato, A.J.; Lesniewski, L.A.; Li, D.Y. Dietary Vitamin D and Its Metabolites Non-Genomically Stabilize the Endothelium. PLoS ONE 2015, 10, e0140370. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Aggarwal, R.; Akhthar, T.; Jain, S.K. Coronary artery disease and its association with Vitamin D deficiency. J. Midlife Health 2016, 7, 56–60. [Google Scholar] [CrossRef]
  107. Garcia, V.C.; Martini, L.A. Vitamin D and cardiovascular disease. Nutrients 2010, 2, 426–437. [Google Scholar] [CrossRef] [Green Version]
  108. Zittermann, A.; Koerfer, R. Vitamin D in the prevention and treatment of coronary heart disease. Curr. Opin. Clin. Nutr. Metab. Care 2008, 11, 752–757. [Google Scholar] [CrossRef] [PubMed]
  109. Fontányi, Z.; Sziva, R.E.; Pál, É.; Hadjadj, L.; Monori-Kiss, A.; Horváth, E.M.; Benkő, R.; Magyar, A.; Heinzlmann, A.; Benyó, Z.; et al. Vitamin D Deficiency Reduces Vascular Reactivity of Coronary Arterioles in Male Rats. Curr. Issues Mol. Biol. 2021, 43, 79–92. [Google Scholar] [CrossRef]
  110. Pál, É.; Hricisák, L.; Lékai, Á.; Nagy, D.; Fülöp, Á.; Erben, R.G.; Várbíró, S.; Sándor, P.; Benyó, Z. Ablation of Vitamin D Signaling Compromises Cerebrovascular Adaptation to Carotid Artery Occlusion in Mice. Cells 2020, 9, 1457. [Google Scholar] [CrossRef]
  111. Sziva, R.E.; Fontányi, Z.; Pál, É.; Hadjadj, L.; Monori-Kiss, A.; Horváth, E.M.; Benkő, R.; Magyar, A.; Heinzlmann, A.; Benyó, Z.; et al. Vitamin D Deficiency Induces Elevated Oxidative and Biomechanical Damage in Coronary Arterioles in Male Rats. Antioxidants 2020, 9, 997. [Google Scholar] [CrossRef] [PubMed]
  112. Latic, N.; Erben, R.G. Vitamin D and Cardiovascular Disease, with Emphasis on Hypertension, Atherosclerosis, and Heart Failure. Int. J. Mol. Sci. 2020, 21, 6483. [Google Scholar] [CrossRef] [PubMed]
  113. Engler-Chiurazzi, E.B.; Monaghan, K.L.; Wan, E.C.K.; Ren, X. Role of B cells and the aging brain in stroke recovery and treatment. Geroscience 2020, 42, 1199–1216. [Google Scholar] [CrossRef]
  114. Hankey, G.J. Stroke. Lancet 2017, 389, 641–654. [Google Scholar] [CrossRef] [PubMed]
  115. Sierra, C.; Coca, A.; Schiffrin, E.L. Vascular mechanisms in the pathogenesis of stroke. Curr. Hypertens. Rep. 2011, 13, 200–207. [Google Scholar] [CrossRef] [PubMed]
  116. Esenwa, C.; Gutierrez, J. Secondary stroke prevention: Challenges and solutions. Vasc. Health Risk Manag. 2015, 11, 437–450. [Google Scholar]
  117. Istvan, L.; Czako, C.; Benyo, F.; Elo, A.; Mihaly, Z.; Sotonyi, P.; Varga, A.; Nagy, Z.Z.; Kovacs, I. The effect of systemic factors on retinal blood flow in patients with carotid stenosis: An optical coherence tomography angiography study. Geroscience 2022, 44, 389–401. [Google Scholar] [CrossRef]
  118. Istvan, L.; Czako, C.; Elo, A.; Mihaly, Z.; Sotonyi, P.; Varga, A.; Ungvari, Z.; Csiszar, A.; Yabluchanskiy, A.; Conley, S.; et al. Imaging retinal microvascular manifestations of carotid artery disease in older adults: From diagnosis of ocular complications to understanding microvascular contributions to cognitive impairment. Geroscience 2021, 43, 1703–1723. [Google Scholar] [CrossRef]
  119. Cermakova, P.; Ding, J.; Meirelles, O.; Reis, J.; Religa, D.; Schreiner, P.J.; Jacobs, D.R.; Bryan, R.N.; Launer, L.J. Carotid Intima-Media Thickness and Markers of Brain Health in a Biracial Middle-Aged Cohort: CARDIA Brain MRI Sub-study. J. Gerontol. A Biol. Sci. Med. Sci. 2020, 75, 380–386. [Google Scholar] [CrossRef] [Green Version]
  120. Makariou, S.E.; Michel, P.; Tzoufi, M.S.; Challa, A.; Milionis, H.J. Vitamin D and stroke: Promise for prevention and better outcome. Curr. Vasc. Pharmacol. 2014, 12, 117–124. [Google Scholar] [CrossRef]
  121. Brondum-Jacobsen, P.; Nordestgaard, B.G.; Schnohr, P.; Benn, M. 25-hydroxyvitamin D and symptomatic ischemic stroke: An original study and meta-analysis. Ann. Neurol. 2013, 73, 38–47. [Google Scholar] [CrossRef] [PubMed]
  122. Kojima, G.; Bell, C.; Abbott, R.D.; Launer, L.; Chen, R.; Motonaga, H.; Ross, G.W.; Curb, J.D.; Masaki, K. Low dietary vitamin D predicts 34-year incident stroke: The Honolulu Heart Program. Stroke 2012, 43, 2163–2167. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Sun, Q.; Pan, A.; Hu, F.B.; Manson, J.E.; Rexrode, K.M. 25-Hydroxyvitamin D levels and the risk of stroke: A prospective study and meta-analysis. Stroke 2012, 43, 1470–1477. [Google Scholar] [CrossRef]
  124. Chowdhury, R.; Stevens, S.; Ward, H.; Chowdhury, S.; Sajjad, A.; Franco, O.H. Circulating vitamin D, calcium and risk of cerebrovascular disease: A systematic review and meta-analysis. Eur. J. Epidemiol. 2012, 27, 581–591. [Google Scholar] [CrossRef]
  125. Turetsky, A.; Goddeau, R.P., Jr.; Henninger, N. Low Serum Vitamin D Is Independently Associated with Larger Lesion Volumes after Ischemic Stroke. J. Stroke Cerebrovasc. Dis. 2015, 24, 1555–1563. [Google Scholar] [CrossRef] [PubMed]
  126. Zeng, Y.Y.; Yuan, C.X.; Wu, M.X.; Cheng, L.; Zhou, S.N.; Hu, P.L.; Fan, K.L.; Tang, W.J.; He, J.C. Low vitamin D levels and the long-term functional outcome of stroke up to 5 years. Brain Behav. 2021, 11, e2244. [Google Scholar] [CrossRef] [PubMed]
  127. Huang, T.; Afzal, S.; Yu, C.; Guo, Y.; Bian, Z.; Yang, L.; Millwood, I.Y.; Walters, R.G.; Chen, Y.; Chen, N.; et al. Vitamin D and cause-specific vascular disease and mortality: A Mendelian randomisation study involving 99,012 Chinese and 106,911 European adults. BMC Med. 2019, 17, 160. [Google Scholar] [CrossRef] [Green Version]
  128. Larsson, S.C.; Traylor, M.; Mishra, A.; Howson, J.M.M.; Michaelsson, K.; Markus, H.S. Serum 25-Hydroxyvitamin D Concentrations and Ischemic Stroke and Its Subtypes. Stroke 2018, 49, 2508–2511. [Google Scholar] [CrossRef]
  129. Leong, A.; Rehman, W.; Dastani, Z.; Greenwood, C.; Timpson, N.; Langsetmo, L.; Berger, C.; Fu, L.; Wong, B.Y.; Malik, S.; et al. The causal effect of vitamin D binding protein (DBP) levels on calcemic and cardiometabolic diseases: A Mendelian randomization study. PLoS Med. 2014, 11, e1001751. [Google Scholar] [CrossRef] [Green Version]
  130. Balden, R.; Selvamani, A.; Sohrabji, F. Vitamin D deficiency exacerbates experimental stroke injury and dysregulates ischemia-induced inflammation in adult rats. Endocrinology 2012, 153, 2420–2435. [Google Scholar] [CrossRef] [Green Version]
  131. Evans, M.A.; Kim, H.A.; Ling, Y.H.; Uong, S.; Vinh, A.; De Silva, T.M.; Arumugam, T.V.; Clarkson, A.N.; Zosky, G.R.; Drummond, G.R.; et al. Vitamin D3 Supplementation Reduces Subsequent Brain Injury and Inflammation Associated with Ischemic Stroke. Neuromolecular Med. 2018, 20, 147–159. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Wang, Y.; Chiang, Y.H.; Su, T.P.; Hayashi, T.; Morales, M.; Hoffer, B.J.; Lin, S.Z. Vitamin D(3) attenuates cortical infarction induced by middle cerebral arterial ligation in rats. Neuropharmacology 2000, 39, 873–880. [Google Scholar] [CrossRef] [PubMed]
  133. Evans, M.A.; Kim, H.A.; De Silva, T.M.; Arumugam, T.V.; Clarkson, A.N.; Drummond, G.R.; Zosky, G.R.; Broughton, B.R.; Sobey, C.G. Diet-induced vitamin D deficiency has no effect on acute post-stroke outcomes in young male mice. J. Cereb. Blood Flow Metab. 2018, 38, 1968–1978. [Google Scholar] [CrossRef] [PubMed]
  134. Chung, P.W.; Park, K.Y.; Kim, J.M.; Shin, D.W.; Park, M.S.; Chung, Y.J.; Ha, S.Y.; Ahn, S.W.; Shin, H.W.; Kim, Y.B.; et al. 25-hydroxyvitamin D status is associated with chronic cerebral small vessel disease. Stroke 2015, 46, 248–251. [Google Scholar] [CrossRef] [Green Version]
  135. Li, X.; Lyu, P.; Ren, Y.; An, J.; Dong, Y. Arterial stiffness and cognitive impairment. J. Neurol. Sci. 2017, 380, 1–10. [Google Scholar] [CrossRef]
  136. Bouvy, W.H.; van Veluw, S.J.; Kuijf, H.J.; Zwanenburg, J.J.; Kappelle, J.L.; Luijten, P.R.; Koek, H.L.; Geerlings, M.I.; Biessels, G.J.; Utrecht Vascular Cognitive Impairment Study, G. Microbleeds colocalize with enlarged juxtacortical perivascular spaces in amnestic mild cognitive impairment and early Alzheimer’s disease: A 7 Tesla MRI study. J. Cereb. Blood Flow Metab. 2020, 40, 739–746. [Google Scholar] [CrossRef] [Green Version]
  137. De Guio, F.; Duering, M.; Fazekas, F.; De Leeuw, F.E.; Greenberg, S.M.; Pantoni, L.; Aghetti, A.; Smith, E.E.; Wardlaw, J.; Jouvent, E. Brain atrophy in cerebral small vessel diseases: Extent, consequences, technical limitations and perspectives: The HARNESS initiative. J. Cereb. Blood Flow Metab. 2020, 40, 231–245. [Google Scholar] [CrossRef]
  138. Derraz, I.; Abdelrady, M.; Gaillard, N.; Ahmed, R.; Cagnazzo, F.; Dargazanli, C.; Lefevre, P.H.; Corti, L.; Riquelme, C.; Mourand, I.; et al. White Matter Hyperintensity Burden and Collateral Circulation in Large Vessel Occlusion Stroke. Stroke 2021, 52, 3848–3854. [Google Scholar] [CrossRef]
  139. Dewenter, A.; Gesierich, B.; Ter Telgte, A.; Wiegertjes, K.; Cai, M.; Jacob, M.A.; Marques, J.P.; Norris, D.G.; Franzmeier, N.; de Leeuw, F.E.; et al. Systematic validation of structural brain networks in cerebral small vessel disease. J. Cereb. Blood Flow Metab. 2022, 42, 1020–1032. [Google Scholar] [CrossRef]
  140. Guey, S.; Lesnik Oberstein, S.A.J.; Tournier-Lasserve, E.; Chabriat, H. Hereditary Cerebral Small Vessel Diseases and Stroke: A Guide for Diagnosis and Management. Stroke 2021, 52, 3025–3032. [Google Scholar] [CrossRef]
  141. Gurol, M.E.; Sacco, R.L.; McCullough, L.D. Multiple Faces of Cerebral Small Vessel Diseases. Stroke 2020, 51, 9–11. [Google Scholar] [CrossRef] [PubMed]
  142. Jansen, M.G.; Griffanti, L.; Mackay, C.E.; Anaturk, M.; Melazzini, L.; Lange, A.G.; Filippini, N.; Zsoldos, E.; Wiegertjes, K.; Leeuw, F.E.; et al. Association of cerebral small vessel disease burden with brain structure and cognitive and vascular risk trajectories in mid-to-late life. J. Cereb. Blood Flow Metab. 2022, 42, 600–612. [Google Scholar] [CrossRef] [PubMed]
  143. Jokinen, H.; Koikkalainen, J.; Laakso, H.M.; Melkas, S.; Nieminen, T.; Brander, A.; Korvenoja, A.; Rueckert, D.; Barkhof, F.; Scheltens, P.; et al. Global Burden of Small Vessel Disease-Related Brain Changes on MRI Predicts Cognitive and Functional Decline. Stroke 2020, 51, 170–178. [Google Scholar] [CrossRef] [PubMed]
  144. Kneihsl, M.; Hofer, E.; Enzinger, C.; Niederkorn, K.; Horner, S.; Pinter, D.; Fandler-Hofler, S.; Eppinger, S.; Haidegger, M.; Schmidt, R.; et al. Intracranial Pulsatility in Relation to Severity and Progression of Cerebral White Matter Hyperintensities. Stroke 2020, 51, 3302–3309. [Google Scholar] [CrossRef] [PubMed]
  145. Koton, S.; Schneider, A.L.C.; Windham, B.G.; Mosley, T.H.; Gottesman, R.F.; Coresh, J. Microvascular Brain Disease Progression and Risk of Stroke: The ARIC Study. Stroke 2020, 51, 3264–3270. [Google Scholar] [CrossRef] [PubMed]
  146. Lin, M.P.; Brott, T.G.; Liebeskind, D.S.; Meschia, J.F.; Sam, K.; Gottesman, R.F. Collateral Recruitment Is Impaired by Cerebral Small Vessel Disease. Stroke 2020, 51, 1404–1410. [Google Scholar] [CrossRef]
  147. Mayer, C.; Frey, B.M.; Schlemm, E.; Petersen, M.; Engelke, K.; Hanning, U.; Jagodzinski, A.; Borof, K.; Fiehler, J.; Gerloff, C.; et al. Linking cortical atrophy to white matter hyperintensities of presumed vascular origin. J. Cereb. Blood Flow Metab. 2021, 41, 1682–1691. [Google Scholar] [CrossRef] [PubMed]
  148. Pasi, M.; Cordonnier, C. Clinical Relevance of Cerebral Small Vessel Diseases. Stroke 2020, 51, 47–53. [Google Scholar] [CrossRef]
  149. Tsai, H.H.; Pasi, M.; Tsai, L.K.; Chen, Y.F.; Chen, Y.W.; Tang, S.C.; Gurol, M.E.; Yen, R.F.; Jeng, J.S. Superficial Cerebellar Microbleeds and Cerebral Amyloid Angiopathy: A Magnetic Resonance Imaging/Positron Emission Tomography Study. Stroke 2020, 51, 202–208. [Google Scholar] [CrossRef]
  150. Zwartbol, M.H.; van der Kolk, A.G.; Kuijf, H.J.; Witkamp, T.D.; Ghaznawi, R.; Hendrikse, J.; Geerlings, M.I.; on behalf of the UCC-SMART Study Group. Intracranial vessel wall lesions on 7T MRI and MRI features of cerebral small vessel disease: The SMART-MR study. J. Cereb. Blood Flow Metab. 2021, 41, 1219–1228. [Google Scholar] [CrossRef]
  151. Hilal, S.; Tan, C.S.; van Veluw, S.J.; Xu, X.; Vrooman, H.; Tan, B.Y.; Venketasubramanian, N.; Biessels, G.J.; Chen, C. Cortical cerebral microinfarcts predict cognitive decline in memory clinic patients. J. Cereb. Blood Flow Metab. 2020, 40, 44–53. [Google Scholar] [CrossRef] [PubMed]
  152. Girouard, H.; Munter, L.M. The many faces of vascular cognitive impairment. J. Neurochem. 2018, 144, 509–512. [Google Scholar] [CrossRef] [Green Version]
  153. Dichgans, M.; Leys, D. Vascular Cognitive Impairment. Circ. Res. 2017, 120, 573–591. [Google Scholar] [CrossRef] [PubMed]
  154. Gorelick, P.B.; Counts, S.E.; Nyenhuis, D. Vascular cognitive impairment and dementia. Biochim. Biophys. Acta 2016, 1862, 860–868. [Google Scholar] [CrossRef] [PubMed]
  155. Gorelick, P.B.; Bowler, J.V. Advances in vascular cognitive impairment. Stroke 2010, 41, e93–e98. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Benedictus, M.R.; Goos, J.D.; Binnewijzend, M.A.; Muller, M.; Barkhof, F.; Scheltens, P.; Prins, N.D.; van der Flier, W.M. Specific risk factors for microbleeds and white matter hyperintensities in Alzheimer’s disease. Neurobiol. Aging 2013, 34, 2488–2494. [Google Scholar] [CrossRef]
  157. Makedonov, I.; Black, S.E.; MacIntosh, B.J. Cerebral small vessel disease in aging and Alzheimer’s disease: A comparative study using MRI and SPECT. Eur. J. Neurol. 2013, 20, 243–250. [Google Scholar] [CrossRef] [PubMed]
  158. Heringa, S.M.; Reijmer, Y.D.; Leemans, A.; Koek, H.L.; Kappelle, L.J.; Biessels, G.J. Multiple microbleeds are related to cerebral network disruptions in patients with early Alzheimer’s disease. J. Alzheimers Dis. 2014, 38, 211–221. [Google Scholar] [CrossRef]
  159. Snyder, H.M.; Corriveau, R.A.; Craft, S.; Faber, J.E.; Greenberg, S.M.; Knopman, D.; Lamb, B.T.; Montine, T.J.; Nedergaard, M.; Schaffer, C.B.; et al. Vascular contributions to cognitive impairment and dementia including Alzheimer’s disease. Alzheimers Dement. 2015, 11, 710–717. [Google Scholar] [CrossRef] [Green Version]
  160. Halliday, M.R.; Rege, S.V.; Ma, Q.; Zhao, Z.; Miller, C.A.; Winkler, E.A.; Zlokovic, B.V. Accelerated pericyte degeneration and blood-brain barrier breakdown in apolipoprotein E4 carriers with Alzheimer’s disease. J. Cereb. Blood Flow Metab. 2016, 36, 216–227. [Google Scholar] [CrossRef]
  161. Nelson, A.R.; Sweeney, M.D.; Sagare, A.P.; Zlokovic, B.V. Neurovascular dysfunction and neurodegeneration in dementia and Alzheimer’s disease. Biochim. Biophys. Acta 2016, 1862, 887–900. [Google Scholar] [CrossRef] [PubMed]
  162. Montagne, A.; Zhao, Z.; Zlokovic, B.V. Alzheimer’s disease: A matter of blood-brain barrier dysfunction? J. Exp. Med. 2017, 214, 3151–3169. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Iadecola, C. Neurovascular regulation in the normal brain and in Alzheimer’s disease. Nat. Rev. Neurosci. 2004, 5, 347–360. [Google Scholar] [PubMed]
  164. Cortes-Canteli, M.; Iadecola, C. Alzheimer’s Disease and Vascular Aging: JACC Focus Seminar. J. Am. Coll. Cardiol. 2020, 75, 942–951. [Google Scholar] [CrossRef]
  165. Zhang, Y.; Bander, E.D.; Lee, Y.; Muoser, C.; Schaffer, C.B.; Nishimura, N. Microvessel occlusions alter amyloid-beta plaque morphology in a mouse model of Alzheimer’s disease. J. Cereb. Blood Flow Metab. 2020, 40, 2115–2131. [Google Scholar] [CrossRef]
  166. Moretti, R.; Caruso, P.; Dal Ben, M.; Conti, C.; Gazzin, S.; Tiribelli, C. Vitamin D, Homocysteine, and Folate in Subcortical Vascular Dementia and Alzheimer Dementia. Front. Aging Neurosci. 2017, 9, 169. [Google Scholar] [CrossRef] [Green Version]
  167. Prabhakar, P.; Chandra, S.R.; Supriya, M.; Issac, T.G.; Prasad, C.; Christopher, R. Vitamin D status and vascular dementia due to cerebral small vessel disease in the elderly Asian Indian population. J. Neurol. Sci. 2015, 359, 108–111. [Google Scholar] [CrossRef]
  168. Ouma, S.; Suenaga, M.; Bolukbasi Hatip, F.F.; Hatip-Al-Khatib, I.; Tsuboi, Y.; Matsunaga, Y. Serum vitamin D in patients with mild cognitive impairment and Alzheimer’s disease. Brain Behav. 2018, 8, e00936. [Google Scholar] [CrossRef] [Green Version]
  169. Jia, J.; Hu, J.; Huo, X.; Miao, R.; Zhang, Y.; Ma, F. Effects of vitamin D supplementation on cognitive function and blood Abeta-related biomarkers in older adults with Alzheimer’s disease: A randomised, double-blind, placebo-controlled trial. J. Neurol. Neurosurg. Psychiatry 2019, 90, 1347–1352. [Google Scholar] [PubMed]
  170. Bao, Z.; Wang, X.; Li, Y.; Feng, F. Vitamin D Alleviates Cognitive Dysfunction by Activating the VDR/ERK1/2 Signaling Pathway in an Alzheimer’s Disease Mouse Model. Neuroimmunomodulation 2020, 27, 178–185. [Google Scholar] [CrossRef]
  171. Du, Y.; Liang, F.; Zhang, L.; Liu, J.; Dou, H. Vitamin D Supplement for Prevention of Alzheimer’s Disease: A Systematic Review and Meta-Analysis. Am. J. Ther. 2020, 28, e638–e648. [Google Scholar] [CrossRef] [PubMed]
  172. Saad El-Din, S.; Rashed, L.; Medhat, E.; Emad Aboulhoda, B.; Desoky Badawy, A.; Mohammed ShamsEldeen, A.; Abdelgwad, M. Active form of vitamin D analogue mitigates neurodegenerative changes in Alzheimer’s disease in rats by targeting Keap1/Nrf2 and MAPK-38p/ERK signaling pathways. Steroids 2020, 156, 108586. [Google Scholar] [CrossRef] [PubMed]
  173. Zhang, H.; Wang, T.; Han, Z.; Wang, L.; Zhang, Y.; Wang, L.; Liu, G. Impact of Vitamin D Binding Protein Levels on Alzheimer’s Disease: A Mendelian Randomization Study. J. Alzheimers Dis. 2020, 74, 991–998. [Google Scholar] [CrossRef] [PubMed]
  174. Panza, F.; La Montagna, M.; Lampignano, L.; Zupo, R.; Bortone, I.; Castellana, F.; Sardone, R.; Borraccino, L.; Dibello, V.; Resta, E.; et al. Vitamin D in the development and progression of alzheimer’s disease: Implications for clinical management. Expert Rev. Neurother. 2021, 21, 287–301. [Google Scholar] [CrossRef]
  175. Dimitrakis, E.; Katsarou, M.S.; Lagiou, M.; Papastefanopoulou, V.; Spandidos, D.A.; Tsatsakis, A.; Papageorgiou, S.; Moutsatsou, P.; Antoniou, K.; Kroupis, C.; et al. Association of vitamin D receptor gene haplotypes with late-onset Alzheimer’s disease in a Southeastern European Caucasian population. Exp. Ther. Med. 2022, 24, 584. [Google Scholar] [CrossRef]
  176. Kang, J.; Park, M.; Lee, E.; Jung, J.; Kim, T. The Role of Vitamin D in Alzheimer’s Disease: A Transcriptional Regulator of Amyloidopathy and Gliopathy. Biomedicines 2022, 10, 1824. [Google Scholar] [CrossRef]
  177. Soares, J.Z.; Valeur, J.; Saltyte Benth, J.; Knapskog, A.B.; Selbaek, G.; Arefi, G.; Gilfillan, G.D.; Tollisen, A.; Bogdanovic, N.; Pettersen, R. Vitamin D in Alzheimer’s Disease: Low Levels in Cerebrospinal Fluid Despite Normal Amounts in Serum. J. Alzheimers Dis. 2022, 86, 1301–1314. [Google Scholar] [CrossRef]
  178. Etgen, T.; Sander, D.; Bickel, H.; Sander, K.; Forstl, H. Vitamin D deficiency, cognitive impairment and dementia: A systematic review and meta-analysis. Dement. Geriatr. Cogn. Disord. 2012, 33, 297–305. [Google Scholar] [CrossRef]
  179. Annweiler, C.; Fantino, B.; Schott, A.M.; Krolak-Salmon, P.; Allali, G.; Beauchet, O. Vitamin D insufficiency and mild cognitive impairment: Cross-sectional association. Eur. J. Neurol. 2012, 19, 1023–1029. [Google Scholar] [CrossRef]
  180. Arosio, B.; Rossi, P.D.; Ferri, E.; Cesari, M.; Vitale, G. Characterization of Vitamin D Status in Older Persons with Cognitive Impairment. Nutrients 2022, 14, 1142. [Google Scholar] [CrossRef]
  181. Balion, C.; Griffith, L.E.; Strifler, L.; Henderson, M.; Patterson, C.; Heckman, G.; Llewellyn, D.J.; Raina, P. Vitamin D, cognition, and dementia: A systematic review and meta-analysis. Neurology 2012, 79, 1397–1405. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  182. Llewellyn, D.J.; Langa, K.M.; Lang, I.A. Serum 25-hydroxyvitamin D concentration and cognitive impairment. J. Geriatr. Psychiatry Neurol. 2009, 22, 188–195. [Google Scholar] [CrossRef] [PubMed]
  183. Beydoun, M.A.; Shaked, D.; Hossain, S.; Beydoun, H.A.; Katzel, L.I.; Davatzikos, C.; Gullapalli, R.P.; Seliger, S.L.; Erus, G.; Evans, M.K.; et al. Vitamin D, Folate, and Cobalamin Serum Concentrations Are Related to Brain Volume and White Matter Integrity in Urban Adults. Front. Aging Neurosci. 2020, 12, 140. [Google Scholar] [CrossRef]
  184. Al-Amin, M.; Bradford, D.; Sullivan, R.K.P.; Kurniawan, N.D.; Moon, Y.; Han, S.H.; Zalesky, A.; Burne, T.H.J. Vitamin D deficiency is associated with reduced hippocampal volume and disrupted structural connectivity in patients with mild cognitive impairment. Hum. Brain Mapp. 2019, 40, 394–406. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Beydoun, M.A.; Ding, E.L.; Beydoun, H.A.; Tanaka, T.; Ferrucci, L.; Zonderman, A.B. Vitamin D receptor and megalin gene polymorphisms and their associations with longitudinal cognitive change in US adults. Am. J. Clin. Nutr. 2012, 95, 163–178. [Google Scholar] [CrossRef] [Green Version]
  186. Kuningas, M.; Mooijaart, S.P.; Jolles, J.; Slagboom, P.E.; Westendorp, R.G.; van Heemst, D. VDR gene variants associate with cognitive function and depressive symptoms in old age. Neurobiol. Aging 2009, 30, 466–473. [Google Scholar] [CrossRef]
  187. Ungvari, Z.; Tarantini, S.; Donato, A.J.; Galvan, V.; Csiszar, A. Mechanisms of Vascular Aging. Circ. Res. 2018, 123, 849–867. [Google Scholar] [CrossRef]
  188. Ungvari, Z.; Tarantini, S.; Sorond, F.; Merkely, B.; Csiszar, A. Mechanisms of Vascular Aging, A Geroscience Perspective: JACC Focus Seminar. J. Am. Coll. Cardiol. 2020, 75, 931–941. [Google Scholar] [CrossRef]
  189. Vetter, V.M.; Spira, D.; Banszerus, V.L.; Demuth, I. Epigenetic Clock and Leukocyte Telomere Length Are Associated with Vitamin D Status but not with Functional Assessments and Frailty in the Berlin Aging Study II. J. Gerontol. A Biol. Sci. Med. Sci. 2020, 75, 2056–2063. [Google Scholar] [CrossRef] [PubMed]
  190. Vetter, V.M.; Sommerer, Y.; Kalies, C.H.; Spira, D.; Bertram, L.; Demuth, I. Vitamin D supplementation is associated with slower epigenetic aging. Geroscience 2022, 44, 1847–1859. [Google Scholar] [CrossRef]
  191. Zhou, J.; Chen, H.; Wang, Q.; Chen, S.; Wang, R.; Wang, Z.; Yang, C.; Chen, A.; Zhao, J.; Zhou, Z.; et al. Sirt1 overexpression improves senescence-associated pulmonary fibrosis induced by vitamin D deficiency through downregulating IL-11 transcription. Aging Cell 2022, 21, e13680. [Google Scholar] [CrossRef] [PubMed]
  192. Yang, R.; Chen, J.; Zhang, J.; Qin, R.; Wang, R.; Qiu, Y.; Mao, Z.; Goltzman, D.; Miao, D. 1,25-Dihydroxyvitamin D protects against age-related osteoporosis by a novel VDR-Ezh2-p16 signal axis. Aging Cell 2020, 19, e13095. [Google Scholar] [CrossRef] [PubMed]
  193. Gomez-Cabrero, D.; Walter, S.; Abugessaisa, I.; Minambres-Herraiz, R.; Palomares, L.B.; Butcher, L.; Erusalimsky, J.D.; Garcia-Garcia, F.J.; Carnicero, J.; Hardman, T.C.; et al. A robust machine learning framework to identify signatures for frailty: A nested case-control study in four aging European cohorts. Geroscience 2021, 43, 1317–1329. [Google Scholar] [CrossRef]
  194. Sung, C.C.; Liao, M.T.; Lu, K.C.; Wu, C.C. Role of vitamin D in insulin resistance. J. Biomed. Biotechnol. 2012, 2012, 634195. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. Hadjadj, L.; Várbíró, S.; Horváth, E.M.; Monori-Kiss, A.; Pál, É.; Karvaly, G.B.; Heinzlmann, A.; Magyar, A.; Szabó, I.; Sziva, R.E.; et al. Insulin resistance in an animal model of polycystic ovary disease is aggravated by vitamin D deficiency: Vascular consequences. Diab. Vasc. Dis. Res. 2018, 15, 294–301. [Google Scholar] [CrossRef] [Green Version]
  196. Pilz, S.; Tomaschitz, A.; Drechsler, C.; Zittermann, A.; Dekker, J.M.; Marz, W. Vitamin D supplementation: A promising approach for the prevention and treatment of strokes. Curr. Drug Targets 2011, 12, 88–96. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  197. Carrelli, A.L.; Walker, M.D.; Lowe, H.; McMahon, D.J.; Rundek, T.; Sacco, R.L.; Silverberg, S.J. Vitamin D deficiency is associated with subclinical carotid atherosclerosis: The Northern Manhattan study. Stroke 2011, 42, 2240–2245. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  198. Benyo, Z.; Ruisanchez, E.; Leszl-Ishiguro, M.; Sandor, P.; Pacher, P. Endocannabinoids in cerebrovascular regulation. Am. J. Physiol. Heart Circ. Physiol. 2016, 310, H785–H801. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  199. Kim, K.J.; Diaz, J.R.; Presa, J.L.; Muller, P.R.; Brands, M.W.; Khan, M.B.; Hess, D.C.; Althammer, F.; Stern, J.E.; Filosa, J.A. Decreased parenchymal arteriolar tone uncouples vessel-to-neuronal communication in a mouse model of vascular cognitive impairment. Geroscience 2021, 43, 1405–1422. [Google Scholar] [CrossRef]
  200. Levit, A.; Hachinski, V.; Whitehead, S.N. Neurovascular unit dysregulation, white matter disease, and executive dysfunction: The shared triad of vascular cognitive impairment and Alzheimer disease. Geroscience 2020, 42, 445–465. [Google Scholar] [PubMed]
  201. Sabayan, B.; Westendorp, R.G.J. Neurovascular-glymphatic dysfunction and white matter lesions. Geroscience 2021, 43, 1635–1642. [Google Scholar] [CrossRef] [PubMed]
  202. Tarantini, S.; Balasubramanian, P.; Delfavero, J.; Csipo, T.; Yabluchanskiy, A.; Kiss, T.; Nyul-Toth, A.; Mukli, P.; Toth, P.; Ahire, C.; et al. Treatment with the BCL-2/BCL-xL inhibitor senolytic drug ABT263/Navitoclax improves functional hyperemia in aged mice. Geroscience 2021, 43, 2427–2440. [Google Scholar] [CrossRef]
  203. Tarantini, S.; Balasubramanian, P.; Yabluchanskiy, A.; Ashpole, N.M.; Logan, S.; Kiss, T.; Ungvari, A.; Nyul-Toth, A.; Schwartzman, M.L.; Benyo, Z.; et al. IGF1R signaling regulates astrocyte-mediated neurovascular coupling in mice: Implications for brain aging. Geroscience 2021, 43, 901–911. [Google Scholar] [CrossRef] [PubMed]
  204. Tarantini, S.; Nyul-Toth, A.; Yabluchanskiy, A.; Csipo, T.; Mukli, P.; Balasubramanian, P.; Ungvari, A.; Toth, P.; Benyo, Z.; Sonntag, W.E.; et al. Endothelial deficiency of insulin-like growth factor-1 receptor (IGF1R) impairs neurovascular coupling responses in mice, mimicking aspects of the brain aging phenotype. Geroscience 2021, 43, 2387–2394. [Google Scholar] [CrossRef] [PubMed]
  205. Gardner, A.W.; Montgomery, P.S.; Wang, M.; Shen, B.; Casanegra, A.I.; Silva-Palacios, F.; Ungvari, Z.; Yabluchanskiy, A.; Csiszar, A.; Waldstein, S.R. Cognitive decrement in older adults with symptomatic peripheral artery disease. Geroscience 2021, 43, 2455–2465. [Google Scholar] [CrossRef] [PubMed]
  206. Benyó, Z.; Wahl, M. Opiate receptor-mediated mechanisms in the regulation of cerebral blood flow. Cerebrovasc. Brain Metab. Rev. 1996, 8, 326–357. [Google Scholar]
  207. Görlach, C.; Benyó, Z.; Wahl, M. Endothelin-1-induced contraction in cerebral vessels mediated by phospholipase C/protein kinase C cascade. Kidney Int. Suppl. 1998, 67, S224–S225. [Google Scholar] [CrossRef] [Green Version]
  208. Iring, A.; Ruisanchez, É.; Leszl-Ishiguro, M.; Horváth, B.; Benkő, R.; Lacza, Z.; Járai, Z.; Sándor, P.; Di Marzo, V.; Pacher, P.; et al. Role of endocannabinoids and cannabinoid-1 receptors in cerebrocortical blood flow regulation. PLoS ONE 2013, 8, e53390. [Google Scholar] [CrossRef] [Green Version]
  209. Benyó, Z.; Lacza, Z.; Hortobágyi, T.; Görlach, C.; Wahl, M. Functional importance of neuronal nitric oxide synthase in the endothelium of rat basilar arteries. Brain Res. 2000, 877, 79–84. [Google Scholar] [CrossRef]
  210. Elting, J.W.J.; Tas, J.; Aries, M.J.; Czosnyka, M.; Maurits, N.M. Dynamic cerebral autoregulation estimates derived from near infrared spectroscopy and transcranial Doppler are similar after correction for transit time and blood flow and blood volume oscillations. J. Cereb. Blood Flow Metab. 2020, 40, 135–149. [Google Scholar] [CrossRef] [Green Version]
  211. Junejo, R.T.; Braz, I.D.; Lucas, S.J.; van Lieshout, J.J.; Phillips, A.A.; Lip, G.Y.; Fisher, J.P. Neurovascular coupling and cerebral autoregulation in atrial fibrillation. J. Cereb. Blood Flow Metab. 2020, 40, 1647–1657. [Google Scholar] [CrossRef]
  212. Nogueira, R.C.; Aries, M.; Minhas, J.S.; Petersen, N.H.; Xiong, L.; Kainerstorfer, J.M.; Castro, P. Review of studies on dynamic cerebral autoregulation in the acute phase of stroke and the relationship with clinical outcome. J. Cereb. Blood Flow Metab. 2022, 42, 430–453. [Google Scholar] [CrossRef] [PubMed]
  213. van Dalen, J.W.; Mutsaerts, H.J.; Petr, J.; Caan, M.W.; van Charante, E.P.M.; MacIntosh, B.J.; van Gool, W.A.; Nederveen, A.J.; Richard, E. Longitudinal relation between blood pressure, antihypertensive use and cerebral blood flow, using arterial spin labelling MRI. J. Cereb. Blood Flow Metab. 2021, 41, 1756–1766. [Google Scholar] [CrossRef] [PubMed]
  214. Wang, S.; Lv, W.; Zhang, H.; Liu, Y.; Li, L.; Jefferson, J.R.; Guo, Y.; Li, M.; Gao, W.; Fang, X.; et al. Aging exacerbates impairments of cerebral blood flow autoregulation and cognition in diabetic rats. Geroscience 2020, 42, 1387–1410. [Google Scholar] [CrossRef] [PubMed]
  215. Cipolla, M.J.; Liebeskind, D.S.; Chan, S.L. The importance of comorbidities in ischemic stroke: Impact of hypertension on the cerebral circulation. J. Cereb. Blood Flow Metab. 2018, 38, 2129–2149. [Google Scholar] [CrossRef] [PubMed]
  216. Szabó, C.; Csáki, C.; Benyó, Z.; Reivich, M.; Kovách, A.G. Role of the L-arginine-nitric oxide pathway in the changes in cerebrovascular reactivity following hemorrhagic hypotension and retransfusion. Circ. Shock 1992, 37, 307–316. [Google Scholar]
  217. Nyul-Toth, A.; Tarantini, S.; Kiss, T.; Toth, P.; Galvan, V.; Tarantini, A.; Yabluchanskiy, A.; Csiszar, A.; Ungvari, Z. Increases in hypertension-induced cerebral microhemorrhages exacerbate gait dysfunction in a mouse model of Alzheimer’s disease. Geroscience 2020, 42, 1685–1698. [Google Scholar] [CrossRef]
  218. Tarantini, S.; Yabluchanskiy, A.; Lindsey, M.L.; Csiszar, A.; Ungvari, Z. Effect of genetic depletion of MMP-9 on neurological manifestations of hypertension-induced intracerebral hemorrhages in aged mice. Geroscience 2021, 43, 2611–2619. [Google Scholar] [CrossRef]
  219. Hadjadj, L.; Pál, É.; Monori-Kiss, A.; Sziva, R.E.; Korsós-Novák, Á.; Mária Horváth, E.; Benkő, R.; Magyar, A.; Magyar, P.; Benyó, Z.; et al. Vitamin D deficiency and androgen excess result eutrophic remodeling and reduced myogenic adaptation in small cerebral arterioles in female rats. Gynecol. Endocrinol. 2019, 35, 529–534. [Google Scholar] [CrossRef]
  220. Pál, É.; Hadjadj, L.; Fontányi, Z.; Monori-Kiss, A.; Lippai, N.; Horváth, E.M.; Magyar, A.; Horváth, E.; Monos, E.; Nádasy, G.L.; et al. Gender, hyperandrogenism and vitamin D deficiency related functional and morphological alterations of rat cerebral arteries. PLoS ONE 2019, 14, e0216951. [Google Scholar] [CrossRef]
  221. Pál, É.; Hadjadj, L.; Fontányi, Z.; Monori-Kiss, A.; Mezei, Z.; Lippai, N.; Magyar, A.; Heinzlmann, A.; Karvaly, G.; Monos, E.; et al. Vitamin D deficiency causes inward hypertrophic remodeling and alters vascular reactivity of rat cerebral arterioles. PLoS ONE 2018, 13, e0192480. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  222. Lacza, Z.; Hermán, P.; Görlach, C.; Hortobágyi, T.; Sándor, P.; Wahl, M.; Benyó, Z. NO synthase blockade induces chaotic cerebral vasomotion via activation of thromboxane receptors. Stroke 2001, 32, 2609–2614. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  223. Benyó, Z.; Görlach, C.; Wahl, M. Involvement of thromboxane A2 in the mediation of the contractile effect induced by inhibition of nitric oxide synthesis in isolated rat middle cerebral arteries. J. Cereb. Blood Flow Metab. 1998, 18, 616–618. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  224. Horváth, B.; Lenzsér, G.; Benyó, B.; Németh, T.; Benko, R.; Iring, A.; Hermán, P.; Komjáti, K.; Lacza, Z.; Sándor, P.; et al. Hypersensitivity to thromboxane receptor mediated cerebral vasomotion and CBF oscillations during acute NO-deficiency in rats. PLoS ONE 2010, 5, e14477. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  225. Polycarpou, A.; Hricisak, L.; Iring, A.; Safar, D.; Ruisanchez, E.; Horvath, B.; Sandor, P.; Benyo, Z. Adaptation of the cerebrocortical circulation to carotid artery occlusion involves blood flow redistribution between cortical regions and is independent of eNOS. Am. J. Physiol. Heart Circ. Physiol. 2016, 311, H972–H980. [Google Scholar] [CrossRef] [PubMed]
  226. Shuaib, A.; Butcher, K.; Mohammad, A.A.; Saqqur, M.; Liebeskind, D.S. Collateral blood vessels in acute ischaemic stroke: A potential therapeutic target. Lancet Neurol. 2011, 10, 909–921. [Google Scholar] [CrossRef]
  227. Struys, T.; Govaerts, K.; Oosterlinck, W.; Casteels, C.; Bronckaers, A.; Koole, M.; Van Laere, K.; Herijgers, P.; Lambrichts, I.; Himmelreich, U.; et al. In vivo evidence for long-term vascular remodeling resulting from chronic cerebral hypoperfusion in mice. J. Cereb. Blood Flow Metab. 2017, 37, 726–739. [Google Scholar] [CrossRef] [Green Version]
  228. Brozici, M.; van der Zwan, A.; Hillen, B. Anatomy and functionality of leptomeningeal anastomoses: A review. Stroke 2003, 34, 2750–2762. [Google Scholar] [CrossRef] [Green Version]
  229. Liebeskind, D.S. Collateral circulation. Stroke 2003, 34, 2279–2284. [Google Scholar] [CrossRef] [Green Version]
  230. Zhang, H.; Prabhakar, P.; Sealock, R.; Faber, J.E. Wide genetic variation in the native pial collateral circulation is a major determinant of variation in severity of stroke. J. Cereb. Blood Flow Metab. 2010, 30, 923–934. [Google Scholar] [CrossRef]
  231. Kiss, T.; Nyul-Toth, A.; Balasubramanian, P.; Tarantini, S.; Ahire, C.; DelFavero, J.; Yabluchanskiy, A.; Csipo, T.; Farkas, E.; Wiley, G.; et al. Single-cell RNA sequencing identifies senescent cerebromicrovascular endothelial cells in the aged mouse brain. Geroscience 2020, 42, 429–444. [Google Scholar] [CrossRef] [PubMed]
  232. Kiss, T.; Nyul-Toth, A.; Balasubramanian, P.; Tarantini, S.; Ahire, C.; Yabluchanskiy, A.; Csipo, T.; Farkas, E.; Wren, J.D.; Garman, L.; et al. Nicotinamide mononucleotide (NMN) supplementation promotes neurovascular rejuvenation in aged mice: Transcriptional footprint of SIRT1 activation, mitochondrial protection, anti-inflammatory, and anti-apoptotic effects. Geroscience 2020, 42, 527–546. [Google Scholar] [CrossRef] [PubMed]
  233. Kiss, T.; Tarantini, S.; Csipo, T.; Balasubramanian, P.; Nyul-Toth, A.; Yabluchanskiy, A.; Wren, J.D.; Garman, L.; Huffman, D.M.; Csiszar, A.; et al. Circulating anti-geronic factors from heterochonic parabionts promote vascular rejuvenation in aged mice: Transcriptional footprint of mitochondrial protection, attenuation of oxidative stress, and rescue of endothelial function by young blood. Geroscience 2020, 42, 727–748. [Google Scholar] [CrossRef]
  234. Secomb, T.W.; Bullock, K.V.; Boas, D.A.; Sakadzic, S. The mass transfer coefficient for oxygen transport from blood to tissue in cerebral cortex. J. Cereb. Blood Flow Metab. 2020, 40, 1634–1646. [Google Scholar] [CrossRef]
  235. Dounavi, M.E.; Low, A.; McKiernan, E.F.; Mak, E.; Muniz-Terrera, G.; Ritchie, K.; Ritchie, C.W.; Su, L.; O’Brien, J.T. Evidence of cerebral hemodynamic dysregulation in middle-aged APOE epsilon4 carriers: The PREVENT-Dementia study. J. Cereb. Blood Flow Metab. 2021, 41, 2844–2855. [Google Scholar] [CrossRef]
  236. He, Y.; Wang, M.; Yu, X. High spatiotemporal vessel-specific hemodynamic mapping with multi-echo single-vessel fMRI. J. Cereb. Blood Flow Metab. 2020, 40, 2098–2114. [Google Scholar] [CrossRef]
  237. Huber, G.; Ogrodnik, M.; Wenzel, J.; Stolting, I.; Huber, L.; Will, O.; Peschke, E.; Matschl, U.; Hovener, J.B.; Schwaninger, M.; et al. Telmisartan prevents high-fat diet-induced neurovascular impairments and reduces anxiety-like behavior. J. Cereb. Blood Flow Metab. 2021, 41, 2356–2369. [Google Scholar] [CrossRef]
  238. Koush, Y.; de Graaf, R.A.; Kupers, R.; Dricot, L.; Ptito, M.; Behar, K.L.; Rothman, D.L.; Hyder, F. Metabolic underpinnings of activated and deactivated cortical areas in human brain. J. Cereb. Blood Flow Metab. 2021, 41, 986–1000. [Google Scholar] [CrossRef]
  239. Li, P.; Mu, J.; Ma, X.; Ding, D.; Ma, S.; Zhang, H.; Liu, J.; Zhang, M. Neurovascular coupling dysfunction in end-stage renal disease patients related to cognitive impairment. J. Cereb. Blood Flow Metab. 2021, 41, 2593–2606. [Google Scholar] [CrossRef]
  240. Sencan, I.; Esipova, T.; Kilic, K.; Li, B.; Desjardins, M.; Yaseen, M.A.; Wang, H.; Porter, J.E.; Kura, S.; Fu, B.; et al. Optical measurement of microvascular oxygenation and blood flow responses in awake mouse cortex during functional activation. J. Cereb. Blood Flow Metab. 2022, 42, 510–525. [Google Scholar] [CrossRef]
  241. Li, W.; Lo, E.H. Leaky memories: Impact of APOE4 on blood-brain barrier and dementia. J. Cereb. Blood Flow Metab. 2020, 40, 1912–1914. [Google Scholar] [CrossRef] [PubMed]
  242. Moon, W.J.; Lim, C.; Ha, I.H.; Kim, Y.; Moon, Y.; Kim, H.J.; Han, S.H. Hippocampal blood-brain barrier permeability is related to the APOE4 mutation status of elderly individuals without dementia. J. Cereb. Blood Flow Metab. 2021, 41, 1351–1361. [Google Scholar] [CrossRef] [PubMed]
  243. Kerkhofs, D.; Wong, S.M.; Zhang, E.; Uiterwijk, R.; Hoff, E.I.; Jansen, J.F.A.; Staals, J.; Backes, W.H.; van Oostenbrugge, R.J. Blood-brain barrier leakage at baseline and cognitive decline in cerebral small vessel disease: A 2-year follow-up study. Geroscience 2021, 43, 1643–1652. [Google Scholar] [CrossRef] [PubMed]
  244. Towner, R.A.; Gulej, R.; Zalles, M.; Saunders, D.; Smith, N.; Lerner, M.; Morton, K.A.; Richardson, A. Rapamycin restores brain vasculature, metabolism, and blood-brain barrier in an inflammaging model. Geroscience 2021, 43, 563–578. [Google Scholar] [CrossRef] [PubMed]
  245. Verheggen, I.C.M.; de Jong, J.J.A.; van Boxtel, M.P.J.; Gronenschild, E.; Palm, W.M.; Postma, A.A.; Jansen, J.F.A.; Verhey, F.R.J.; Backes, W.H. Increase in blood-brain barrier leakage in healthy, older adults. Geroscience 2020, 42, 1183–1193. [Google Scholar] [CrossRef] [PubMed]
  246. Verheggen, I.C.M.; de Jong, J.J.A.; van Boxtel, M.P.J.; Postma, A.A.; Jansen, J.F.A.; Verhey, F.R.J.; Backes, W.H. Imaging the role of blood-brain barrier disruption in normal cognitive ageing. Geroscience 2020, 42, 1751–1764. [Google Scholar] [CrossRef] [PubMed]
  247. Norling, A.M.; Gerstenecker, A.T.; Buford, T.W.; Khan, B.; Oparil, S.; Lazar, R.M. The role of exercise in the reversal of IGF-1 deficiencies in microvascular rarefaction and hypertension. Geroscience 2020, 42, 141–158. [Google Scholar]
  248. Jor’dan, A.J.; Manor, B.; Iloputaife, I.; Habtemariam, D.A.; Bean, J.F.; Sorond, F.A.; Lipsitz, L.A. Diminished Locomotor Control Is Associated With Reduced Neurovascular Coupling in Older Adults. J. Gerontol. A Biol. Sci. Med. Sci. 2020, 75, 1516–1522. [Google Scholar] [CrossRef]
  249. El-Atifi, M.; Dreyfus, M.; Berger, F.; Wion, D. Expression of CYP2R1 and VDR in human brain pericytes: The neurovascular vitamin D autocrine/paracrine model. Neuroreport 2015, 26, 245–248. [Google Scholar] [CrossRef]
  250. Gastfriend, B.D.; Foreman, K.L.; Katt, M.E.; Palecek, S.P.; Shusta, E.V. Integrative analysis of the human brain mural cell transcriptome. J. Cereb. Blood Flow Metab. 2021, 41, 3052–3068. [Google Scholar] [CrossRef]
  251. Afsar, B.; Afsar, R.E.; Dagel, T.; Kaya, E.; Erus, S.; Ortiz, A.; Covic, A.; Kanbay, M. Capillary rarefaction from the kidney point of view. Clin. Kidney J. 2018, 11, 295–301. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  252. Koleganova, N.; Piecha, G.; Ritz, E.; Gross, M.L. Calcitriol ameliorates capillary deficit and fibrosis of the heart in subtotally nephrectomized rats. Nephrol. Dial. Transplant. 2009, 24, 778–787. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  253. Won, S.; Sayeed, I.; Peterson, B.L.; Wali, B.; Kahn, J.S.; Stein, D.G. Vitamin D prevents hypoxia/reoxygenation-induced blood-brain barrier disruption via vitamin D receptor-mediated NF-kB signaling pathways. PLoS ONE 2015, 10, e0122821. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  254. Enkhjargal, B.; McBride, D.W.; Manaenko, A.; Reis, C.; Sakai, Y.; Tang, J.; Zhang, J.H. Intranasal administration of vitamin D attenuates blood-brain barrier disruption through endogenous upregulation of osteopontin and activation of CD44/P-gp glycosylation signaling after subarachnoid hemorrhage in rats. J. Cereb. Blood Flow Metab. 2017, 37, 2555–2566. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  255. Yang, J.; Wang, K.; Hu, T.; Wang, G.; Wang, W.; Zhang, J. Vitamin D3 Supplement Attenuates Blood-Brain Barrier Disruption and Cognitive Impairments in a Rat Model of Traumatic Brain Injury. Neuromolecular Med. 2021, 23, 491–499. [Google Scholar] [CrossRef] [PubMed]
  256. Chen, B.R.; Kozberg, M.G.; Bouchard, M.B.; Shaik, M.A.; Hillman, E.M. A critical role for the vascular endothelium in functional neurovascular coupling in the brain. J. Am. Heart Assoc. 2014, 3, e000787. [Google Scholar] [CrossRef]
  257. Tarantini, S.; Valcarcel-Ares, N.M.; Yabluchanskiy, A.; Fulop, G.A.; Hertelendy, P.; Gautam, T.; Farkas, E.; Perz, A.; Rabinovitch, P.S.; Sonntag, W.E.; et al. Treatment with the mitochondrial-targeted antioxidant peptide SS-31 rescues neurovascular coupling responses and cerebrovascular endothelial function and improves cognition in aged mice. Aging Cell 2018, 17, e12731. [Google Scholar] [CrossRef]
  258. Castle, M.; Fiedler, N.; Pop, L.C.; Schneider, S.J.; Schlussel, Y.; Sukumar, D.; Hao, L.; Shapses, S.A. Three Doses of Vitamin D and Cognitive Outcomes in Older Women: A Double-Blind Randomized Controlled Trial. J. Gerontol. A Biol. Sci. Med. Sci. 2020, 75, 835–842. [Google Scholar] [CrossRef]
  259. Griffin, T.P.; Wall, D.; Blake, L.; Griffin, D.G.; Robinson, S.M.; Bell, M.; Mulkerrin, E.C.; O’Shea, P.M. Vitamin D Status of Adults in the Community, in Outpatient Clinics, in Hospital, and in Nursing Homes in the West of Ireland. J. Gerontol. A Biol. Sci. Med. Sci. 2020, 75, 2418–2425. [Google Scholar] [CrossRef]
  260. Kositsawat, J.; Kuo, C.L.; Barry, L.C.; Melzer, D.; Bandinelli, S.; Ferrucci, L.; Wu, R.; Kuchel, G.A. Interaction Between Vitamin D and Interleukin 6 on Slow Gait Speed: 6-Year Follow-up Data of Older Adults From InCHIANTI. J. Gerontol. A Biol. Sci. Med. Sci. 2020, 75, 1161–1166. [Google Scholar] [CrossRef]
  261. Lai, R.H.; Hsu, C.C.; Yu, B.H.; Lo, Y.R.; Hsu, Y.Y.; Chen, M.H.; Juang, J.L. Vitamin D supplementation worsens Alzheimer’s progression: Animal model and human cohort studies. Aging Cell 2022, 21, e13670. [Google Scholar] [CrossRef] [PubMed]
  262. Lai, R.H.; Hsu, Y.Y.; Shie, F.S.; Huang, C.C.; Chen, M.H.; Juang, J.L. Non-genomic rewiring of vitamin D receptor to p53 as a key to Alzheimer’s disease. Aging Cell 2021, 20, e13509. [Google Scholar] [CrossRef] [PubMed]
  263. Rathmacher, J.A.; Pitchford, L.M.; Khoo, P.; Angus, H.; Lang, J.; Lowry, K.; Ruby, C.; Krajek, A.C.; Fuller, J.C.; Sharp, R.L. Long-term Effects of Calcium beta-Hydroxy-beta-Methylbutyrate and Vitamin D3 Supplementation on Muscular Function in Older Adults with and without Resistance Training: A Randomized, Double-blind, Controlled Study. J. Gerontol. A Biol. Sci. Med. Sci. 2020, 75, 2089–2097. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Genomic actions of vitamin D. 1,25-dihydroxyvitamin D3 (1,25(OH)2D3) binds to the vitamin D receptor (VDR) and promotes its heterodimerization with the retinoid X receptor (RXR). The ligand-bound VDR/RXR complex binds to the vitamin D response elements (VDRE) in the promoters of numerous genes and modulates their transcription. Therefore, 1,25(OH)2D3 regulates several physiological processes, such as cell proliferation, differentiation, and inflammation [13]. Figure reproduced from “Vitamin D Deficiency and the Risk of Cerebrovascular Disease.” by Kim et al., Antioxidants (Basel), 2020, 9, 327, doi:10.3390/antiox9040327. The original figure was published (and can be reproduced) under the terms of CC-BY 4.0 [13].
Figure 1. Genomic actions of vitamin D. 1,25-dihydroxyvitamin D3 (1,25(OH)2D3) binds to the vitamin D receptor (VDR) and promotes its heterodimerization with the retinoid X receptor (RXR). The ligand-bound VDR/RXR complex binds to the vitamin D response elements (VDRE) in the promoters of numerous genes and modulates their transcription. Therefore, 1,25(OH)2D3 regulates several physiological processes, such as cell proliferation, differentiation, and inflammation [13]. Figure reproduced from “Vitamin D Deficiency and the Risk of Cerebrovascular Disease.” by Kim et al., Antioxidants (Basel), 2020, 9, 327, doi:10.3390/antiox9040327. The original figure was published (and can be reproduced) under the terms of CC-BY 4.0 [13].
Nutrients 15 00334 g001
Figure 2. Overview of the cardiovascular system-related impacts of vitamin D (TSP: thrombospondin, PAI-1: plasminogen activator inhibitor-1, RAS: renin-angiotensin system, TH1: T helper type 1 cell, TH2: T-helper type 2 cell; “See Figure 4” refers to Figure 4 of the original article by Norman PE and Powell JT [1]). Figure reproduced from “Vitamin D and cardiovascular disease” by Norman PE and Powell JT, Circ Res 2014, 114, 379–393, doi:10.1161/circresaha.113.301241. [1] with permission of the publisher. Copyright 2014, American Heart Association, Inc.
Figure 2. Overview of the cardiovascular system-related impacts of vitamin D (TSP: thrombospondin, PAI-1: plasminogen activator inhibitor-1, RAS: renin-angiotensin system, TH1: T helper type 1 cell, TH2: T-helper type 2 cell; “See Figure 4” refers to Figure 4 of the original article by Norman PE and Powell JT [1]). Figure reproduced from “Vitamin D and cardiovascular disease” by Norman PE and Powell JT, Circ Res 2014, 114, 379–393, doi:10.1161/circresaha.113.301241. [1] with permission of the publisher. Copyright 2014, American Heart Association, Inc.
Nutrients 15 00334 g002
Figure 3. Representative actions of vitamin D deficiency in the (cerebro)vascular wall. Vitamin D deficiency impairs vascular functions via several pathways, including reductions in nitric oxide (NO) production, due to decreased endothelial nitric oxide synthase (eNOS) expression, phosphorylation, and dimerization. In addition, enhanced level of reactive oxygen species (ROS), resulting mainly from decreased expression of cytosolic copper-zinc superoxide dismutase (CuZn-SOD) and upregulation of the free radical generating nicotinamide adenine dinucleotide phosphate (NADPH) oxidase enzyme and its subunits in endothelial cells, contributes to endothelial dysfunction. Furthermore, vitamin D deficiency is associated with increased production of pro-inflammatory mediators, for instance, tumor necrosis factor-α (TNF-α) and cyclooxygenase 2 (COX-2), leading to vascular inflammation. Remodeling of extracellular matrix (ECM) and increased collagen–elastin ratio of vessel wall—particularly due to altered expression and activity of specific matrix metalloproteinases (MMPs)—as well as changes in vascular smooth muscle cell (VSMC) proliferation, may result in vascular remodeling and stiffening.
Figure 3. Representative actions of vitamin D deficiency in the (cerebro)vascular wall. Vitamin D deficiency impairs vascular functions via several pathways, including reductions in nitric oxide (NO) production, due to decreased endothelial nitric oxide synthase (eNOS) expression, phosphorylation, and dimerization. In addition, enhanced level of reactive oxygen species (ROS), resulting mainly from decreased expression of cytosolic copper-zinc superoxide dismutase (CuZn-SOD) and upregulation of the free radical generating nicotinamide adenine dinucleotide phosphate (NADPH) oxidase enzyme and its subunits in endothelial cells, contributes to endothelial dysfunction. Furthermore, vitamin D deficiency is associated with increased production of pro-inflammatory mediators, for instance, tumor necrosis factor-α (TNF-α) and cyclooxygenase 2 (COX-2), leading to vascular inflammation. Remodeling of extracellular matrix (ECM) and increased collagen–elastin ratio of vessel wall—particularly due to altered expression and activity of specific matrix metalloproteinases (MMPs)—as well as changes in vascular smooth muscle cell (VSMC) proliferation, may result in vascular remodeling and stiffening.
Nutrients 15 00334 g003
Table 1. Selected VitD-regulated genes linked to cardiovascular and cerebrovascular functions.
Table 1. Selected VitD-regulated genes linked to cardiovascular and cerebrovascular functions.
Gene NameCell TypeBiological Function
Transforming growth factor, beta 2 and 3 (TGFB2, TGFB3) [33]VSMCcell proliferation
Prostaglandin-endoperoxide synthase 1 (cyclooxygenase 1) (PTGS1) [33,34]VSMC, endothelial cellprostanoid synthesis
Vascular endothelial growth factor (VEGF) [35,36]VSMC, endothelial cellangiogenesis
Tissue inhibitor of metalloproteinase 1 and 2 (TIMP1, TIMP2) [34]cardiomyocyteECM homeostasis
Tissue inhibitor of metalloproteinase 3 (TIMP3) [35]VSMCECM homeostasis
Matrix metalloproteinase 2 and 9(MMP2, MMP9) [34]cardiomyocyteECM homeostasis
Nuclear factor kappa B (NFKB) [34]endothelial cellinflammation
Endothelial nitric oxide synthase (NOS3) [37]endothelial cellNO production
Interleukin 6 (IL6) [34]endothelial cellinflammation
Renin (REN) [38]human embryonic kidney cells, mesangial cellsblood pressure,
sodium retention
VSMC: vascular smooth muscle cell, ECM: extracellular matrix, NO: nitric oxide.
Table 2. Role of vitamin D deficiency (VDD) in the pathogenesis of cardiovascular and cerebrovascular diseases.
Table 2. Role of vitamin D deficiency (VDD) in the pathogenesis of cardiovascular and cerebrovascular diseases.
Risk Factor/MarkerMechanism
HypertensionRenin expression
Diabetes mellitusβ-cell function and insulin sensitivity
Cardiac hypertrophyECM remodeling
AtherosclerosisCholesterol uptake by macrophages
Vascular inflammationIncreased pro-inflammatory cytokines, decreased anti-inflammatory cytokines
Oxidative stressROS
Endothelial dysfunctioneNOS expression and activity,
reaction of NO with ROS
Vascular permeabilityTight junction proteins
Vascular remodelingVSMC proliferation, ECM remodeling, collagen–elastin content
ECM: extracellular matrix, ROS: reactive oxygen species, NO: nitric oxide, eNOS: endothelial nitric oxide synthase, VSMC: vascular smooth muscle cell.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Pál, É.; Ungvári, Z.; Benyó, Z.; Várbíró, S. Role of Vitamin D Deficiency in the Pathogenesis of Cardiovascular and Cerebrovascular Diseases. Nutrients 2023, 15, 334. https://doi.org/10.3390/nu15020334

AMA Style

Pál É, Ungvári Z, Benyó Z, Várbíró S. Role of Vitamin D Deficiency in the Pathogenesis of Cardiovascular and Cerebrovascular Diseases. Nutrients. 2023; 15(2):334. https://doi.org/10.3390/nu15020334

Chicago/Turabian Style

Pál, Éva, Zoltán Ungvári, Zoltán Benyó, and Szabolcs Várbíró. 2023. "Role of Vitamin D Deficiency in the Pathogenesis of Cardiovascular and Cerebrovascular Diseases" Nutrients 15, no. 2: 334. https://doi.org/10.3390/nu15020334

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