Next Article in Journal
Immunolocalization of Matrix Metalloproteinases 2 and 9 and Their Inhibitors in the Hearts of Rats Treated with Immunosuppressive Drugs—An Artificial Intelligence-Based Digital Analysis
Previous Article in Journal
Elevated NET, Calprotectin, and Neopterin Levels Discriminate between Disease Activity in COVID-19, as Evidenced by Need for Hospitalization among Patients in Northern Italy
Previous Article in Special Issue
Novel Multi-Antioxidant Approach for Ischemic Stroke Therapy Targeting the Role of Oxidative Stress
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Vitamin D and Cardiovascular Diseases: From Physiology to Pathophysiology and Outcomes

by
Matteo Nardin
1,2,
Monica Verdoia
3,4,
Simone Nardin
5,6,
Davide Cao
1,7,
Mauro Chiarito
1,8,
Elvin Kedhi
9,10,
Gennaro Galasso
11,
Gianluigi Condorelli
1,8 and
Giuseppe De Luca
12,13,*
1
Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20090 Milan, Italy
2
Internal Medicine, Department of Medicine, ASST Spedali Civili di Brescia, 25123 Brescia, Italy
3
Division of Cardiology, Ospedale degli Infermi, ASL Biella, 13875 Biella, Italy
4
Department of Translational Medicine, Eastern Piedmont University, 28100 Novara, Italy
5
U.O. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
6
Department of Internal Medicine and Medical Sciences, School of Medicine, University of Genova, 16126 Genova, Italy
7
Department of Cardiology, Humanitas Gavazzeni Hospital, 24125 Bergamo, Italy
8
Department of Cardiovascular Medicine, IRCCS-Humanitas Research Hospital, 20089 Rozzano, Italy
9
McGill University Health Center, Montreal, QC H3G 1A4, Canada
10
Department of Cardiology and Structural Heart Disease, University of Silesia, 40-032 Katowice, Poland
11
Department of Medicine, Surgery and Dentistry, University of Salerno, 84081 Baronissi, Italy
12
Division of Cardiology, AOU “Policlinico G. Martino”, Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy
13
Division of Cardiology, IRCCS Hospital Galeazzi-Sant’Ambrogio, 20157 Milan, Italy
*
Author to whom correspondence should be addressed.
Biomedicines 2024, 12(4), 768; https://doi.org/10.3390/biomedicines12040768
Submission received: 20 February 2024 / Revised: 18 March 2024 / Accepted: 26 March 2024 / Published: 30 March 2024
(This article belongs to the Special Issue Role of Natural Product in Cardiovascular Disease)

Abstract

:
Vitamin D is rightly recognized as an essential key factor in the regulation of calcium and phosphate homeostasis, affecting primary adequate bone mineralization. In the last decades, a more complex and wider role of vitamin D has been postulated and demonstrated. Cardiovascular diseases have been found to be strongly related to vitamin D levels, especially to its deficiency. Pre-clinical studies have suggested a direct role of vitamin D in the regulation of several pathophysiological pathways, such as endothelial dysfunction and platelet aggregation; moreover, observational data have confirmed the relationship with different conditions, including coronary artery disease, heart failure, and hypertension. Despite the significant evidence available so far, most clinical trials have failed to prove any positive impact of vitamin D supplements on cardiovascular outcomes. This discrepancy indicates the need for further information and knowledge about vitamin D metabolism and its effect on the cardiovascular system, in order to identify those patients who would benefit from vitamin D supplementation.

1. Introduction

Cardiovascular diseases continue to be the leading causes of mortality and morbidity worldwide. While significant progress has been made in the effective prevention of communicable diseases, particularly infections, and maternal–fetal mortality [1], this success has resulted in longer average life expectancies, leading to a more pronounced impact of cardiovascular conditions on public health [2].
Despite improvements in the treatment of the acute phase of cardiac diseases, such as myocardial infarction (MI) [3,4,5,6,7,8,9], primarily caused by atherothrombosis in the coronary vessels, the outcomes remain unsatisfactory in high-risk patient subgroups [10,11,12]. Therefore, there is a need for further research to explore new risk factors [13,14,15,16,17] and therapies [18] in coronary artery disease (CAD). A comprehensive understanding of the pathogenetic process is still lacking and, among the investigated factors, vitamin D has been hypothesized to play a role in the atherosclerosis process.
Since the early 20th Century, vitamin D has been shown to prevent and treat bone diseases including rickets and osteomalacia. Beyond its role in calcium and phosphate homeostasis, vitamin D has been significantly associated with cardiovascular disease [19,20]. The key rationale for this relationship is attributed to the transcription role of vitamin D, which has been proved to modulate the expression of approximately 3% of the human genome. Various observational studies have reported a significant association between vitamin D deficiency and various cardiovascular conditions, including but not limited to arterial stiffness, hypertension, atherosclerosis, and left ventricular hypertrophy [21,22]. Mechanisms regulated by vitamin D include endothelial dysfunction, cardiomyocyte function, and the calcification of valves and vessels [21,22,23].
In this review we aim to provide an overview of current evidence regarding the role of vitamin D in the cardiovascular system, covering molecular aspects and different clinical scenarios.

2. Methods and Search Strategy

After outlining the structure of our manuscript, we systematically searched the main medical electronic databases, such as PubMed, Medline, Scopus, Cochrane, and Google Scholar, utilizing specific keywords for each section.
For ‘vitamin D’, which was included in every search, we used the following set of terms: vitamin D, Vitamin D3, Vitamin D2, 25-hydroxyergocalciferol, 25-hydroxycholecalciferol, calcifediol, ercalcidiol, 1,25-dihydroxycholecalciferol, calcitriol, 1,25-dihydroxyergocalciferol, ercalcitriol, vitamin D binding protein, and vitamin D receptor. For ‘vitamin D metabolism’, we used terms such as metabolism, anabolism, catabolism, skin, liver, kidney, food, and hydroxylase. For ‘Endothelial dysfunction and inflammation’, we utilized terms such as endothelial dysfunction, inflammation, cytokines, reactive oxygen species, pro-inflammatory, interleukin, tumor necrosis factor, immunity, and immune cells. For ‘coronary artery disease’, we included terms such as coronary artery disease, atherosclerosis, lipid, lipoprotein, calcification, metabolic dysfunction, diabetes, acute coronary syndrome, myocardial infarction, and myocardial ischemia. For ‘platelet aggregation’, terms such as platelets, aggregation, aggregometry, and platelet reactivity were used. For ‘systemic arterial hypertension’, we utilized terms including hypertension, renin–angiotensin–aldosterone system, blood pressure, vascular resistance, reactive oxygen species, incidence, and prevalence. For ‘aortic stenosis’, we employed terms such as aortic valve stenosis, degenerative aortic stenosis, and valve calcification. For ‘heart failure’, terms such as heart failure, dyspnea, congestion, preserved ejection fraction, reduced ejection fraction, and left ventricle were used. For ‘genetics’, terms such as single nucleotide polymorphism, genetics, allele variant, and haplotype were utilized. For ‘therapeutic considerations’, terms such as trial, randomized clinical trial, supplement, outcome, mortality, and cardiovascular events were included. Priority was given to papers published since January 2004 and those found in indexed peer-reviewed journals. Each section was organized to present pre-clinical evidence followed by clinical findings. The therapeutic and outcome aspects were collectively addressed in a dedicated section.

3. Vitamin D Metabolism

The metabolism of vitamin D comprises intricate pathways (Figure 1) involving the skin, liver, kidney, and solar light to obtain the final active compound. It is important to note that the term vitamin D refers to a group of secosteroids derived from dietary intake or human cell production. In animal cells, the initial step involves 7-dehydrocholesterol, a precursor of cholesterol. Human skin cells also produce this compound, acting as pro-vitamin D. After exposure to solar ultraviolet B beams, the B ring of the steroid nucleus breaks, resulting in a secosteroid, which is called pre-vitamin D. This pre-vitamin D is then converted through spontaneous isomerization by a thermal reaction into cholecalciferol, also known as vitamin D3. The skin’s production of vitamin D3 is proportional to sun exposure and the quantity of melatonin. Vitamin D3 levels can also be increased through dietary intake from animal-based foods, mainly dairy products. Ingestion of plant-based food leads to the absorption of a similar compound named vitamin D2, derived from the plant cell steroid ergosterol. Both vitamin D3 and vitamin D2 are metabolized by hepatic 25-hydroxylases, encoded by the gene for cytochrome P450 2R1 (CYP2R1), into 25-hydroxycholecalciferol (25(OH)-D3), also known as calcifediol if from vitamin D3, or into 25-hydroxyergocalciferol (25(OH)-D2), also named ercalcidiol, if from vitamin D2. Routine blood measurements do not differentiate between 25(OH)-D3 and 25(OH)-D2, grouping them both as 25(OH)-D. General recommendations for vitamin D supplements assign the same conversion value to vitamin D3 and D2 [24], due to their similar [25], although not identical [26], ability to elevate serum 25(OH)-D levels. Most steps in the metabolism and actions of vitamins D2 and D3 are overlapping, but evidence suggests that cholecalciferol increases serum 25(OH)-D levels to a greater extent and maintains these higher levels for longer than ergocalciferol does [27], even though both forms are well absorbed in the gut [28,29]. Analogous evidence has been traditionally described in relation to rickets [30,31], but recent data from human transcriptome analyses has shown that gene expression related to immunity differs between vitamin D2 and D3 [32].
The final step involves hydroxylation on carbon 1, mediated by renal 1α-hydroxylase, encoded by gene CYP27B1, leading to the active vitamin D compounds 1,25-dihydroxycholecalciferol (1,25(OH)2-D3), also known as calcitriol if from vitamin D3, or 1,25-dihydroxyergocalciferol (1,25(OH)2-D2), also named ercalcitriol if from vitamin D2. Biological effects of compounds from vitamin D3 and vitamin D2 are widely accepted as equal, allowing the use of the comprehensive term 1,25(OH)2-D for both [33]. Both 25(OH)-D and 1,25(OH)2-D can by inactivated by 24-hydroylase, producing calcitroic acid (24,25(OH)2-D).
Due to the highly lipophilic properties of all vitamin D compounds, they bind to vitamin D binding protein (DBP) and, to a lesser extent, albumin in the bloodstream. Only a limited fraction of vitamin D is unbound to circulating proteins and is considered biologically active. The half-life of 25(OH)-D is significantly higher than that of the active compound 1,25(OH)2-D, at 10–20 d vs. 10–20 h. The target of 1,25(OH)2-D is the vitamin D receptor (VDR), located in the cytosol of in the nucleus of target cells. After crossing the cell membrane, calcitriol binds to VDR, causing a conformational change in the receptor and leading to heterodimerization with retinoic acid’s X-receptor. This complex then translocates into the nucleus, binding to vitamin D response elements on DNA and enhancing the calcium-binding protein synthesis. Non-genomic effects of vitamin D have been described, mostly involving the release of ionized calcium from intracellular stores.
Recent findings have shown that vitamin D can be activated in extra-renal tissues expressing the 1α-hydroxylase enzyme, suggesting a role of circulating 25(OH)-D as a substrate for local 1,25(OH)2-D synthesis. The extra-renal produced 1,25(OH)2-D may exert autocrine and paracrine effects. Therefore, the final biological effect of vitamin D is a sum of circulating calcitriol, derived by the kidney, and a locally produced fraction.
Classically, the metabolic pathways are finely regulated by calcium, phosphate, parathyroid hormone (PTH), and fibroblast growth factor 23 (FGF23). The peptide hormone PTH focuses on increasing blood calcium levels by acting on osteoclasts in bone, on calcium reabsorption in renal tubules, and on production of 1,25(OH)2-D in the kidney. Negative feedback is mediated by the increase in calcium and calcitriol levels [34,35].
FGF23 is a compound directly involved in inherited renal phosphate–wasting disease [36]. Released by osteoblasts and osteocytes, its synthesis is stimulated by 1,25(OH)2-D [37,38]. In the last decades, it has been identified as a significant regulator of vitamin D metabolism, inhibiting the kidney1α-hydroxylase [39]. Several pre-clinical studies have confirmed the link between bone and kidney mediated by FGF23 [38,40,41], forming a bone–kidney axis in the regulation of active vitamin D (Figure 2).
Emerging evidence in recent years has highlighted the endocrine role of vitamin D, not limited to calcium homeostasis but also affecting a wide range of cardiovascular diseases and components of metabolic syndromes. A key element is the presence of VDR in organs beyond those directly involved in calcium and phosphate regulation. Walters et al. identified VDR in rat hearts in 1986 [42], and VDR has also been found in endothelial cells, vascular smooth muscle cells, cardiac fibroblasts, and platelet surface [43,44,45,46], attributing a broad cardiovascular impact to vitamin D.

4. Endothelial Dysfunction and Inflammation

The definition of endothelial dysfunction encompasses a multitude of factors, signals, and compounds that share the common feature of impairing vascular endothelium homeostasis. Various pathological conditions, including atherosclerosis, hypertension, and platelet aggregation, are exacerbated by endothelial dysfunction.
Vitamin D can protect the endothelial function from different types of damage primarily mediated by reactive oxygen species (ROS) [47]. In the leptin-induced endothelial dysfunction model, calcitriol exhibits a protective role on human umbilical vein endothelial cells. This effect is mediated by downregulating vascular inflammatory mediators, such as monocyte chemoattractant protein 1 (MCP-1) and vascular cell adhesion molecule 1 (VCAM-1), along with pro-inflammatory factors, including nuclear factor-kB (NF-kB) [48,49]. A cross-sectional study in obese children observed an inverse association between 25(OH)-D and C-reactive protein (CRP), interleukin (IL)-6, malondialdehyde (MDA), and superoxide dismutase (SOD) [50]. In a randomized control trial with 114 type 2 diabetic patients, vitamin D supplementation led to improvements in HbA1c levels and a reduction in advanced oxidation protein products [51]. Conversely, vitamin D deficiency was found to be significantly associated with endothelial dysfunction [52]. Two meta-analyses reported that vitamin D supplementation significantly reduced CRP, MDA, and tumor necrosis factor-α (TNF-α) [53,54].
In addition to affecting inflammatory mediators, vitamin D could also influence immune cells and their polarization towards a pro-inflammatory phenotype [55]. Previous studies reported a reduction in lymphocytes and inflammatory cytokines secretion after vitamin D administration [56,57]. These findings were consistent among both diabetics and non-diabetics, suggesting the robustness of the association between vitamin D and immunity, irrespective of concomitant comorbidity [58]. Specifically, vitamin D reduced the expression of IL-1β, IL-6 and TNF-α, favoring IL-10 secretion [59,60], and promoted M2 macrophage differentiation while hampering the M1 type [61]. Vitamin D was reported to play a role in reducing neutrophil extracellular traps (NETs), that are deeply involved in the atherothrombotic process [62,63]. Calcitriol inhibits the generation of lymphocyte T helper 1 (TH1) and T helper 17 (TH17) [64,65], while promoting lymphocyte T helper 2 (TH2) and T regulatory (Treg) cells [66,67]. Therefore, a consensus has been reached regarding the essential role of vitamin D in regulating the immune system, marked by a general anti-inflammatory action [68].

5. Coronary Artery Disease

Atherosclerosis, closely linked with inflammation, affects both the initiation and progression of coronary artery disease (CAD). The protective role of vitamin D against CAD involves the modulation of the inflammatory response by reducing the expression of IL-1, IL-6, and TNF-α [19,69]. This subsequently decreases CRP and other pro-inflammatory cytokines, limiting the formation of macrophage-derived foam cells, a pivotal element in atherosclerosis progression [70]. Beyond its anti-inflammatory effects, calcitriol is hypothesized to impact lipid homeostasis, particularly by reducing low-density lipoprotein (LDL) uptake by macrophages through the limitation of scavenger receptor expression on the macrophage surface [71]. These findings were confirmed in subjects with various cardiovascular risk factors, including diabetes mellitus and obesity [72].
In clinical studies, serum vitamin D levels were found to be inversely associated with the extent of vascular calcifications [73] and with carotid intima–media thickness [74], which is considered an early marker of atherosclerosis. The concept of a bone–vascular axis has been explored in terms of rationale and evidence, but further clarification is needed to assess its clinical effect [75].
Considering the shared inflammatory signaling pathways between vitamin D and other cardiovascular risk factors, potential confounding interplays may exist. However, a large prospective study, the Health Professionals Follow-up Study, reported that the association between low vitamin D levels and an increased risk of acute myocardial infarction is substantially maintained, irrespective of major cardiovascular risk factors. This study, that included 18,225 men with up to a 10-year follow-up period, showed that patients with vitamin D levels ≥ 30 ng/mL had almost half the adjusted risk of myocardial infarction [76]. Subsequent meta-analyses confirmed a 1.5 times adjusted relative risk of major adverse cardiovascular events for lower (≤60 nmol/L) vs. higher (>60 nmol/L) vitamin D levels [77].
Among patients undergoing percutaneous coronary intervention (PCI), a consistent prevalence of vitamin D deficiency was detected, with a significant relationship with the prevalence and extent of CAD [78]. Similar results were found in patients with diabetes mellitus, although diabetes was not identified as a predictor of vitamin D deficiency [79].
In a subset of patients referred for a ST-segment elevation myocardial infarction (STEMI), vitamin D levels were inversely related to total cholesterol and LDL values, regardless of ongoing statin therapy at admission [80], as well as inflammatory biomarkers [81].
In the extensive Multi-Ethnic Study of Atherosclerosis (MESA), which involved 6436 patients with CAD and an observational period exceeding 8 y, individuals of the white race with lower serum 25(OH)-D concentration (<20 ng/mL) exhibited an elevated risk of experiencing cardiovascular events [82]. Also, among patients admitted for an acute coronary syndrome, several studies have confirmed a higher incidence of major adverse cardiovascular events [83,84,85], including impaired reperfusion in STEMI patients [86].
However, no significant link has been found between severe vitamin D deficiency (<10.2 ng/mL) and peri-procedural MI among patients undergoing PCI [87]. The main reason for this could be attributed to the different pathogenesis of acute and peri-procedural MI, with the latter strongly influenced by procedural features, including the instruments used and pharmacological treatments, such as glycoprotein IIB/IIIA inhibitors [88].
Possible explanations for these results may involve detrimental left ventricular remodeling; Padoan et al. suggested that adverse remodeling after a MI was observed in patients with lower vitamin D levels at baseline (12.6 vs. 18.7 ng/mL), with an increased risk of mortality at follow-up [89]. Definite answers, however, are still lacking, requiring further clarification of underlying mechanisms to design appropriate trials investigating effective therapeutic strategies.

6. Platelet Aggregation

Koyama et al. have suggested a pivotal antithrombotic role of vitamin D, reporting that calcitriol regulates thrombomodulin and tissue factor expression, thereby reducing platelet aggregation and thrombogenicity [90]. Furthermore, VDR has been identified in platelet mitochondria [46], indicating a direct role of vitamin D in regulating platelet function [91]. Non-genomic action in platelets is initiated by the binding of vitamin D and VDR, involving the regulation of intracellular calcium concentration, a crucial crossroad for platelet signaling leading to their aggregation [63].
Of particular interest is the impact of vitamin D levels during antiplatelet therapy, which is crucial in CAD patients requiring treatment. In patients on dual antiplatelet therapy (DAPT) with clopidogrel or ticagrelor, lower (≤9.1 ng/mL) vitamin D levels were related to a higher prevalence of high on-treatment residual platelet reactivity (HRPR), although these results did not achieve statistical significance at multivariate analysis [92]. However, in diabetic patients, the inverse relationship finding between HRPR prevalence and vitamin D levels reached statistical significance, especially for new P2Y12 inhibitors [93], suggesting a close connection between glycemic and vitamin D pathways in the cardiovascular field [94].
Further confirmations have been provided by Sultan et al., who found a significantly enhanced platelet reactivity among diabetic patients inversely correlated with vitamin D levels. Moreover, calcitriol administration to ex vivo platelets led to a significant reduction in platelet reactivity induced by collagen and ADP [95]. Involvement in the metabolism of homocysteine, a pro-thrombotic substance, has also been suggested for vitamin D [96]. These findings underscore the contribution of vitamin D to regulating platelet activity and thrombus formation. However, the exact molecular pathway induced by non-genomic VDR in platelets needs clarification, as well as the potential contribution of PTH [97].

7. Systemic Arterial Hypertension

As the foremost cardiovascular risk factor, systemic arterial hypertension deserves consideration regarding its potential link with vitamin D. The role of vitamin D in endothelial dysfunction, reactive oxygen species (ROS) production, and immune cell regulation could contribute to the development and maintenance of hypertension.
Specific research attention has been dedicated to the renin–angiotensin–aldosterone system (RAAS). In murine models, mice knockout for VDR exhibited higher blood pressure and developed cardiac hypertrophy due to enhanced renin expression, subsequently activating the signaling cascade [98]. The protective role of RAAS inhibitors was found to be more pronounced among human patients with lower values of vitamin D (12.7 ng/mL) [99].
However, initial animal models showed potential interference mediated by secondary hyperparathyroidism, leading to the introduction of a diet enriched with calcium and phosphate to normalize calcium homeostasis and renin concentration [100].
Conversely, aortic tissue in mice subjected to knockout for VDR and on an enriched diet exhibited an attenuated content of elastin and collagen. Additionally, endothelial nitric oxide synthase (eNOS) was reduced by half [100,101]. A complementary investigation in rats confirmed improved vascular tone with chronic calcitriol administration, resulting in upregulation of eNOS, reduction of ROS, and cyclooxygenase-1 (COX-1) expression [102].
Large-scale observational studies have linked vitamin D status to blood pressure values and the incidence of hypertension. In the National Health and Nutrition Examination Survey (NHANES) III, a significant inverse relationship between vitamin D and blood pressure values was reported, irrespective of age, sex, ethnicity, and physical activity [103]. Nevertheless, a noticeable attenuation of this correlation was found after accounting for body mass index and PTH levels, suggesting that PTH might mediate the observed impact of vitamin D on blood pressure [104], potentially as reported for HRPR prevalence in CAD patients [97].
Further investigations have suggested an attenuated age-related increment of blood pressure values, particularly systolic values, in people with normal vitamin D levels [105,106]. A meta-analysis of prospective observational studies reported a lower risk of hypertension of 12% for every 10 ng/mL increment in circulating 25(OH)-D levels [107]. However, other authors have not found the same trend [108], and suggested a more prominent role of PTH in the regulation of blood pressure rather than vitamin D [109], leading to an indeterminate answer to the question related to hypertension.

8. Aortic Stenosis

Degenerative aortic valve stenosis has garnered significant research attention in recent decades, particularly due to impressive therapeutic advancements using transcatheter approaches. However, its pathogenesis remains elusive, hindering a comprehensive understanding and the development of preventive strategies to alleviate the condition. Despite sharing several risk factors with CAD, a consistent percentage of patients do not exhibit concomitant significant CAD [110].
The role of vitamin D in degenerative aortic valve stenosis is controversial. It may either promote calcium deposition, leading to an increase in serum calcium, or mitigate the inflammatory environment that could advance valve disease progression. Despite pre-clinical evidence suggesting potential vessel calcification with exposure to high concentrations of vitamin D [111], murine models have shown that vitamin D supplementation does not exacerbate calcium deposition and aortic stenosis grade [112]. A more prominent role has been suggested for PTH. In an in vitro study, valvular endothelial cells exposed to high levels of PTH were found to undergo changes in transcription and protein secretion. This modification also impacts paracrine mediators that interact with valvular interstitial cells, stimulating them to switch their phenotype to an osteogenic profile [113].
Findings from a small-scale study proposed a potential linear relationship between low levels of vitamin D and aortic valve peak flow velocity, as well as stenosis progression [114]. A retrospective study demonstrated significantly lower survival and a higher incidence of aortic valve replacement among patients receiving calcium supplements, with or without vitamin D, compared to those not receiving calcium [115]. Conversely, another study found phosphate, but not calcium or vitamin D, to be significantly related to aortic valve stenosis incidence [116]. A large prospective observational Chinese study suggested a potential protective role of vitamin D in relation to degenerative aortic valve stenosis [117].
Further studies are needed to clarify the mechanisms related to the calcification process involving both valvular endothelial and valvular interstitial cells. The complex interplay of signals is not completely understood [118], making it challenging to definitively state the potential beneficial or detrimental impact of vitamin D.

9. Heart Failure

The term heart failure (HF) denotes a complex syndrome arising from structural and/or functional abnormalities of the heart, resulting in elevated intracardiac pressure and/or inadequate cardiac output at rest and/or during exercise. The pathophysiology of HF is characterized by several mechanisms, including hemodynamic impairment, neurohormonal activation, enhanced inflammation, and cardiac remodeling [119] (Figure 3).
Among the structural alterations observed in HF is left ventricular hypertrophy, typically occurring in response to volume or pressure overload, as seen in conditions such as hypertension or aortic valve stenosis. The thickening of the wall is often accompanied by an increase in chamber dimensions. Concurrently, with the enlargement of cardiomyocytes, interstitial fibrosis occurs, primarily reactive, while fibrosis developing after ischemia and cell loss is termed reparative. In both cases, there is an increase in extracellular matrix components in the myocardium, leading to stiffening of the walls and functional impairment [120].
There is consistent evidence supporting the anti-fibrotic and anti-hypertrophic action of vitamin D. Multipotent mesenchymal cells induced in a fibrotic phenotype, after exposure to 1,25(OH)2-D, exhibit decreased production of collagen, transforming growth factor-β1, and an increase in the production of metalloprotease-8 [121]. Moreover, in an animal model with mice subjected to knockout for the VDR gene, Chen et al. demonstrated a direct anti-hypertrophic activity in the heart from vitamin D-VDR activation which, in turn, involves the calcineurin/nuclear factor of activated T cell/modulatory calcineurin inhibitory protein 1 pathway [122].
Observational data revealed that patients with moderate or severe vitamin D deficiency (<20 ng/mL) displayed higher left ventricular wall thickness and diameter than non-deficient patients [123]. This association was also maintained in the context of severe aortic stenosis, demonstrating a linear relationship between vitamin D levels and wall thickness [124]. Similarly, left ventricular mass and myocardial performance index were impaired in a context of low vitamin D levels (<20 ng/mL) in a subset of hypertensive patients with preserved left ventricular ejection fraction [125].
Clinical studies have d a significant relationship between HF and vitamin D deficiency and its impact on outcomes. In a general healthcare population of more than 40,000 patients, vitamin D conferred an adjusted hazard ratio for new-onset HF of 1.31 and 2.01 for low (16–30 ng/mL) and very low (≤15 ng/mL) levels of vitamin D, respectively, compared with the normal value [126], and the general prevalence of HF progressively increased from normal (>30 ng/mL) to very low vitamin D levels. Functional capacity was also impaired, resulting in a consequent increase in hospitalization among patients with HF and vitamin D deficiency (≤10.9 ng/mL) [127].
Results from the NHANES cohort in the early 2000s, including 8351 patients, showed that 89% of patients with HF and CAD were deficient in vitamin D (<30 ng/mL) [128]. Additionally, Liu et al. demonstrated an adjusted hazard ratio of 1.09 (95% CI = 1.00–1.16) (p = 0.040) for all-cause mortality or re-hospitalization for HF at 18 months per 10 mmol/L decrease in vitamin D [129].

10. Genetics

The primary investigations related to genetic variants in vitamin D pathways have involved DBP, VDR, and hydroxylase enzymes [130]. In relation to DBP, two main single nucleotide polymorphisms (SNPs), rs7041 and rs4588, located in exon 11 of the coding gene, have been studied. In the ARIC study, carriers of the rs7041 substitution, genetically predisposed to low (17.2 ng/mL) 25(OH)-D levels, showed a mild increased risk of stroke at a median follow-up of 20 years [131]. However, no link with ischemic heart disease was found [132]. On the other hand, Powe et al. showed that the T allele of rs7041 was associated with lower levels of DBP, potentially increasing the free quotient of 25(OH)-D [133]; however, the impact on CAD remains undetermined [134]. The rs7041 was also investigated in relation to platelet aggregation: carriers with the G allele, which provides a higher concentration and affinity for vitamin D of DBP, were found to more frequently have HRPR in the case of lower serum vitamin D levels. Moreover, the results of the platelet aggregation ADP-test were significantly inversely related to vitamin D levels in G allele carriers [135].
The main SNPs of the VDR gene investigated affected both exons and intronic regions. The rs731236, also known as the associated restriction enzyme TaqI, is a synonymous T>C substitution on exon 9. A link with cardiovascular disease has been described in Pakistani [136] and Asian populations [137]. In French type 2 diabetics, TaqI, together with two SNPs of intronic regions, rs1544410 (G>A on intron 8) and rs7975232 (A>C on intron 8), was found significantly in linkage disequilibrium and significantly related to the prevalence of CAD [138]. The intronic SNPs rs1544410 (G>A) and rs7975232 (A>C), also named BsmI and ApaI, respectively, from the associated restriction enzyme, have been significantly related to low HDL cholesterol and low vitamin D. Homozygotes for the mutated A allele of BsmI resulted in an increased risk of CAD [139]. In other populations, BsmI but not TaqI significantly correlates with the incidence of CAD [140]. Other authors found no relationship between ApaI or TaqI and CAD, while a significant association was reported for another SNP, rs2228570, also known as FokI (A>T,C,G on exon 2) [141]. This leads to a shorter and more active VDR as homozygotes for the A allele of FokI SNPs had a significantly higher risk of CAD compared to other genotypes [142]. In a retrospective case–control study involving a Caucasian population, only SNP rs2228570 was significantly associated with the development of CV disease, while no influence was observed with the VDR polymorphisms BsmI, TaqI or ApaI [143]. Considering different ethnicities, in a Chinese study, two other SNPs in the non-coding region of the VDR gene, rs2189480 (C>A on the intronic region) and rs3847987 (C>A on the 3′- untranslated region), were found to be significantly related to cardiovascular disease [144]. Other researchers have investigated polymorphisms of VDR in relation to the development of restenosis after a PCI in a large cohort of patients. The main findings were consistent with a lower rate of coronary restenosis after PCI in patients presenting the AA haplotype in block 2 of the VDR gene compared to those with the GG haplotype. Considering that the block 2-AA haplotype has previously been shown to result in increased transcriptional activity of the VDR promoter, it appears that the higher the number of VDR, the lower risk of restenosis [145]. Genetic variants of VDR have been found to be significantly related to degenerative aortic stenosis in a pivotal study investigating the association of vitamin D with valvular disease [146].
Large studies adopting Mendelian randomization analyses investigated specific SNPs in relation to the risk of cardiovascular disease outcomes, including cardiovascular mortality, CAD, MI, and hypertension. Brøndum-Jacobsen et al. [147] and Afzal et al. [148] investigated four SNPs, two related to the 7-dehydrocholesterol reductase (DCHR7) gene (rs7944926 and rs11234027), and two in the CYP2R1 gene (rs10741657 and rs12794714). The main findings showed that genetically determined low 25(OH)-D levels were associated with increased all-cause mortality and cancer mortality, but not with increased cardiovascular mortality. Similarly, no evidence was found in relation to the risk of ischemic heart disease or MI. After selection through genome-wide association, four SNPs related to DCHR7, hydroxylase enzymes, and transport were tested in a Canadian population. The authors’ conclusion was that genetically lowered 25(OH)-D levels were not associated with an increased risk of CAD, suggesting that observational evidence of the relationship between circulating 25(OH)-D levels and CAD was possibly confounded or potentially due to reverse causation [149]. The most recent analyses included a large UK population and used non-linear Mendelian randomization after the identification of 35 genome-wide significant variants. The findings provided genetic evidence for an L-shaped association between 25(OH)-D and cardiovascular risk, which was largely restricted to individuals with low vitamin D status. The amelioration of vitamin D status among individuals with the lowest concentrations (<50 nmol/L) provided the strongest benefit [130].
Results on DNA are not conclusive, despite some evidence, but there are many confounds, and the final effect includes other elements. Studies involving non-coding RNA, in particular microRNA, are ongoing, but the evidence available so far is scant and limited in size in relation to the cardiovascular impact of vitamin D [150].

11. Therapeutic Considerations

There is a lack of agreement on optimal circulating vitamin D levels and thresholds to define its deficiency in the general population (Table 1). Moreover, the various types of supplements used, including vitamin D3, vitamin D2, and calcifediol, pose challenges in formulating conclusions.
In relation to lipid parameters, there is consistent heterogeneity in randomized trials, both in terms of baseline vitamin D levels and daily supplementation doses. A meta-analysis of 38 randomized clinical trials demonstrated a small but significant effect of vitamin D compared to a placebo in reducing total cholesterol, LDL, and triglycerides, and increasing high-density lipoprotein (HDL) [151]. Similar results for total cholesterol, LDL, and triglycerides were found in a more recent meta-analysis by Dibaba, including 41 randomized controlled trials with 3434 patients. However, HDL levels were not significantly modified by vitamin D supplements compared to a placebo [152]. Notably, there is evidence of a beneficial interaction between statins and vitamin D levels. The introduction of high-intensity statins in patients undergoing PCI increases vitamin D levels independently of ongoing supplementation and reduces platelet reactivity [153].
Despite robust evidence of the relationship between vitamin D and coronary artery disease (CAD), controversies persist regarding the impact of oral supplementation on cardiovascular outcomes [154]. The main evidence of benefits is based on the improvement of the inflammatory environment, including CRP and IL-6 [81]. Ergocalciferol administration among CAD patients did not result in beneficial changes in vascular function parameters after 3 mo [155]. In a small parallel-group, placebo-controlled, double-blind randomized trial in patients with a prior myocardial infarction (MI), a high dose of cholecalciferol showed no effect on cardiovascular outcomes, including blood pressure or cholesterol levels [156]. Conversely, Chinese CAD patients receiving calcitriol supplementation showed improvement in their SYNTAX score after 6 months [157].
Findings on hypertension are inconclusive. While some studies suggest that vitamin D supplementation, even at high doses, improved blood pressure values in the short term [158], a meta-analysis of 16 clinical trials did not show a significant impact on blood pressure from vitamin D supplements, except for patients with existing cardiometabolic impairment [159]. In young people with vitamin D deficiency, the impact of specific supplements is negligible [160].
The Randomised Evaluation of Calcium Or vitamin D (RECORD) trial, involving 5262 older people randomized to receive cholecalciferol (800 IU/day), calcium (1000 mg/day), cholecalciferol plus calcium, or a placebo, demonstrated a significant reduction in heart failure (HF) events with vitamin D supplementation compared to no supplementation, but no difference in myocardial infarction (MI) or stroke [161]. The VitamIN D treating patients with Chronic heArT failurE (VINDICATE) study, a randomized double-blind controlled trial in elderly patients with HF and vitamin D deficiency, showed a significant improvement in left ventricular structure and function with daily 100 μg (4000 IU) cholecalciferol [162]. However, when considering hard endpoints such as mortality, vitamin D supplementation among deficient patients with HF showed no difference compared to a placebo [163].
The following three large randomized controlled clinical trials investigated effects of vitamin supplementation on cardiovascular disease outcomes as a primary endpoint: the Vitamin D Assessment (ViDA) trial [164], the Vitamin D And Omega-3 Trial (VITAL) trial [165], and the Vitamin D3–Omega-3-Home Exercise-Healthy Ageing and Longevity (DO-HEALTH) trial [166].
The ViDA included 5108 persons from New Zealand, aged from 50 to 84 years, who were randomized to receive a bolus dose of 100,000 IU cholecalciferol or a placebo monthly. The median follow-up duration was 3.3 yr. The primary endpoint was a composite of cardiovascular disease incidence or death, and it occurred in 11.8% of the intervention group and 11.5% of the placebo group, yielding an adjusted hazard ratio of 1.02 (95% confidence interval (CI) = 0.87–1.20). Similar results were found in relation to baseline vitamin D status and for each of the secondary endpoints.
The VITAL trial recruited 25,871 patients from the USA, with an age of at least 50 yr for men and 55 for women. A factorial 2 × 2 design randomized participants to a daily dose of 2000 IU cholecalciferol and a daily dose of 1 g omega-3 fatty acid, both placebo-controlled. The median follow-up duration was 5.3 yr. The primary endpoints were invasive cancer of any type and major adverse cardiovascular events, and a composite of MI, stroke or death from cardiovascular causes. Results confirmed no significant reduction in either major adverse cardiovascular events or invasive cancer incidence.
The DO-HEALTH trial recruited 2157 adults, with an age of a least 70 years. A factorial 2 × 2 × 2 design was adopted to test cholecalciferol supplement, omega-3 supplement and a strength-exercise program. Supplementation of cholecalciferol was of 2000 IU daily. At a median follow-up of 2.99 yr, the two primary endpoints related to cardiovascular diseases, systolic and diastolic blood pressure, did not achieve statistical significance in relation to the cholecalciferol supplement compared to a placebo.
Altogether, the three large, randomized trials confirmed the absence of clinical benefit in terms of cardiovascular outcomes, despite differences in supplement dosage and follow-up duration. These data were also supported by meta-analyses, indicating the safety but null effect of vitamin D [167,168].
Table 1. The main international recommendation thresholds of circulating vitamin D and daily intake levels.
Table 1. The main international recommendation thresholds of circulating vitamin D and daily intake levels.
Target of Circulating 25(OH)-D (nmol/L)25(OH)-D Insufficiency (nmol/L)25(OH)-D Deficiency (nmol/L)Potentially Harmful Levels of Circulating 25(OH)-D (nmol/L)Recommended Daily Intake of Vitamin D Equivalent (IU/Day)Upper Tolerable Intake Levels of Vitamin D Equivalent (IU/Day)
Endocrine Society [169]
Males and females (9–18 years)7551–74≤50250600–10004000
Males and females (>18 years)7551–74≤502501500–200010,000
IOM [170]
Males and females (9–70 years)5030–49<301256004000
Males and females (>70 years)5030–49<301258004000
EFSA 2023 [24]
Males and females (>18 years)50<49 2506004000 (10,000) *
Mayo Clinic Recommendation [171]
Males and females (>18 years)62.525–62.4<25200800–200010,000 IU
Converter: 1 µg/L = 1 ng/mL = 2.5 nmol/L. 1 µg = 40 IU. * 10,000 IU is the threshold over which vitamin D toxicity was reported. EFSA suggests considering a 2.5 factor of safety, thus 4000 IU. 25(OH)-D = 25(OH) vitamin D; EFSA = European Food Safety Authority; IOM = Institute of Medicine; IU = international unit.

12. Conclusions and Future Perspectives

The numerous differences between the studies and trials conducted so far, in terms of patients’ risk profiles, baseline vitamin D status, supplementation doses, and primary endpoint selection, limit the ability to reach definitive findings and formulate recommendations. The results of pre-clinical studies and the observed significant independent relationships are strong and often concur in favor of a protective role of vitamin D in the cardiovascular field. The discrepancies within interventional clinical studies, both randomized and non-randomized, lead to the hypothesis that further clarification is required in relation to vitamin D metabolism, including genetics investigations. These investigations will be crucial in designing and conducting optimal randomized clinical trials for the appropriate population.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Roth, G.A.; Johnson, C.; Abajobir, A.; Abd-Allah, F.; Abera, S.F.; Abyu, G.; Ahmed, M.; Aksut, B.; Alam, T.; Alam, K.; et al. Global, Regional, and National Burden of Cardiovascular Diseases for 10 Causes, 1990 to 2015. J. Am. Coll. Cardiol. 2017, 70, 1–25. [Google Scholar] [CrossRef] [PubMed]
  2. Autier, P.; Boniol, M.; Pizot, C.; Mullie, P. Vitamin D status and ill health: A systematic review. lancet. Diabetes Endocrinol. 2014, 2, 76–89. [Google Scholar] [CrossRef]
  3. De Luca, G.; Navarese, E.P.; Cassetti, E.; Verdoia, M.; Suryapranata, H. Meta-analysis of randomized trials of glycoprotein IIb/IIIa inhibitors in high-risk acute coronary syndromes patients undergoing invasive strategy. Am. J. Cardiol. 2011, 107, 198–203. [Google Scholar] [CrossRef] [PubMed]
  4. De Luca, G.; Schaffer, A.; Wirianta, J.; Suryapranata, H. Comprehensive meta-analysis of radial vs femoral approach in primary angioplasty for STEMI. Int. J. Cardiol. 2013, 168, 2070–2081. [Google Scholar] [CrossRef] [PubMed]
  5. Verdoia, M.; Pergolini, P.; Rolla, R.; Nardin, M.; Schaffer, A.; Barbieri, L.; Marino, P.; Bellomo, G.; Suryapranata, H.; De Luca, G. Advanced age and high-residual platelet reactivity in patients receiving dual antiplatelet therapy with clopidogrel or ticagrelor. J. Thromb. Haemost. 2016, 14, 57–64. [Google Scholar] [CrossRef] [PubMed]
  6. Costa, F.; Montalto, C.; Branca, M.; Hong, S.-J.; Watanabe, H.; Franzone, A.; Vranckx, P.; Hahn, J.-Y.; Gwon, H.-C.; Feres, F.; et al. Dual antiplatelet therapy duration after percutaneous coronary intervention in high bleeding risk: A meta-analysis of randomized trials. Eur. Heart J. 2023, 44, 954–968. [Google Scholar] [CrossRef]
  7. Chichareon, P.; Modolo, R.; Collet, C.; Tenekecioglu, E.; Vink, M.A.; Oh, P.C.; Ahn, J.-M.; Musto, C.; Díaz de la Llera, L.S.; Cho, Y.-S.; et al. Efficacy and Safety of Stents in ST-Segment Elevation Myocardial Infarction. J. Am. Coll. Cardiol. 2019, 74, 2572–2584. [Google Scholar] [CrossRef] [PubMed]
  8. De Luca, G.; Smits, P.; Hofma, S.H.; Di Lorenzo, E.; Vlachojannis, G.J.; Van’t Hof, A.W.J.; van Boven, A.J.; Kedhi, E.; Stone, G.W.; Suryapranata, H.; et al. Everolimus eluting stent vs first generation drug-eluting stent in primary angioplasty: A pooled patient-level meta-analysis of randomized trials. Int. J. Cardiol. 2017, 244, 121–127. [Google Scholar] [CrossRef]
  9. Timmer, J.R.; van der Horst, I.C.C.; de Luca, G.; Ottervanger, J.P.; Hoorntje, J.C.A.; de Boer, M.-J.; Suryapranata, H.; Dambrink, J.-H.E.; Gosselink, M.; Zijlstra, F.; et al. Comparison of myocardial perfusion after successful primary percutaneous coronary intervention in patients with ST-elevation myocardial infarction with versus without diabetes mellitus. Am. J. Cardiol. 2005, 95, 1375–1377. [Google Scholar] [CrossRef]
  10. Bujak, K.; Vidal-Cales, P.; Gabani, R.; Rinaldi, R.; Gomez-Lara, J.; Ortega-Paz, L.; Jimenez-Diaz, V.; Jimenez-Kockar, M.; Jimenez-Quevedo, P.; Diletti, R.; et al. Relationship between stent length and very long-term target lesion failure following percutaneous coronary intervention for ST-elevation myocardial infarction in the drug-eluting stents era: Insights from the EXAMINATION-EXTEND study. Am. Heart J. 2023, 264, 72–82. [Google Scholar] [CrossRef]
  11. De Luca, G.; Dirksen, M.T.; Spaulding, C.; Kelbæk, H.; Schalij, M.; Thuesen, L.; van der Hoeven, B.; Vink, M.A.; Kaiser, C.; Musto, C.; et al. Time course, predictors and clinical implications of stent thrombosis following primary angioplasty. Insights from the DESERT cooperation. Thromb. Haemost. 2013, 110, 826–833. [Google Scholar] [CrossRef] [PubMed]
  12. De Luca, G.; van ’t Hof, A.W.J.; Ottervanger, J.P.; Hoorntje, J.C.A.; Gosselink, A.T.M.; Dambrink, J.-H.E.; de Boer, M.-J.; Suryapranata, H. Ageing, impaired myocardial perfusion, and mortality in patients with ST-segment elevation myocardial infarction treated by primary angioplasty. Eur. Heart J. 2005, 26, 662–666. [Google Scholar] [CrossRef] [PubMed]
  13. Barbato, E.; Piscione, F.; Bartunek, J.; Galasso, G.; Cirillo, P.; De Luca, G.; Iaccarino, G.; De Bruyne, B.; Chiariello, M.; Wijns, W. Role of beta2 adrenergic receptors in human atherosclerotic coronary arteries. Circulation 2005, 111, 288–294. [Google Scholar] [CrossRef] [PubMed]
  14. De Luca, G.; Verdoia, M.; Cassetti, E.; Schaffer, A.; Cavallino, C.; Bolzani, V.; Marino, P. Novara Atherosclerosis Study Group (NAS) High fibrinogen level is an independent predictor of presence and extent of coronary artery disease among Italian population. J. Thromb. Thrombolysis 2011, 31, 458–463. [Google Scholar] [CrossRef] [PubMed]
  15. Silverio, A.; Cancro, F.P.; Di Maio, M.; Bellino, M.; Esposito, L.; Centore, M.; Carrizzo, A.; Di Pietro, P.; Borrelli, A.; De Luca, G.; et al. Lipoprotein(a) levels and risk of adverse events after myocardial infarction in patients with and without diabetes. J. Thromb. Thrombolysis 2022, 54, 382–392. [Google Scholar] [CrossRef] [PubMed]
  16. Farina, F.M.; Serio, S.; Hall, I.F.; Zani, S.; Cassanmagnago, G.A.; Climent, M.; Civilini, E.; Condorelli, G.; Quintavalle, M.; Elia, L. The epigenetic enzyme DOT1L orchestrates vascular smooth muscle cell-monocyte crosstalk and protects against atherosclerosis via the NF-κB pathway. Eur. Heart J. 2022, 43, 4562–4576. [Google Scholar] [CrossRef] [PubMed]
  17. Barbieri, L.; Verdoia, M.; Schaffer, A.; Marino, P.; Suryapranata, H.; De Luca, G. Novara Atherosclerosis Study Group (NAS) Impact of sex on uric acid levels and its relationship with the extent of coronary artery disease: A single-centre study. Atherosclerosis 2015, 241, 241–248. [Google Scholar] [CrossRef] [PubMed]
  18. Schwartz, G.G.; Steg, P.G.; Szarek, M.; Bhatt, D.L.; Bittner, V.A.; Diaz, R.; Edelberg, J.M.; Goodman, S.G.; Hanotin, C.; Harrington, R.A.; et al. Alirocumab and Cardiovascular Outcomes after Acute Coronary Syndrome. N. Engl. J. Med. 2018, 379, 2097–2107. [Google Scholar] [CrossRef] [PubMed]
  19. Hammami, M.M.; Yusuf, A. Differential effects of vitamin D2 and D3 supplements on 25-hydroxyvitamin D level are dose, sex, and time dependent: A randomized controlled trial. BMC Endocr. Disord. 2017, 17, 12. [Google Scholar] [CrossRef]
  20. Chowdhury, R.; Kunutsor, S.; Vitezova, A.; Oliver-Williams, C.; Chowdhury, S.; Kiefte-de-Jong, J.C.; Khan, H.; Baena, C.P.; Prabhakaran, D.; Hoshen, M.B.; et al. Vitamin D and risk of cause specific death: Systematic review and meta-analysis of observational cohort and randomised intervention studies. BMJ 2014, 348, g1903. [Google Scholar] [CrossRef]
  21. Al Mheid, I.; Patel, R.; Murrow, J.; Morris, A.; Rahman, A.; Fike, L.; Kavtaradze, N.; Uphoff, I.; Hooper, C.; Tangpricha, V.; et al. Vitamin D status is associated with arterial stiffness and vascular dysfunction in healthy humans. J. Am. Coll. Cardiol. 2011, 58, 186–192. [Google Scholar] [CrossRef] [PubMed]
  22. Lee, J.H.; Gadi, R.; Spertus, J.A.; Tang, F.; O’Keefe, J.H. Prevalence of vitamin D deficiency in patients with acute myocardial infarction. Am. J. Cardiol. 2011, 107, 1636–1638. [Google Scholar] [CrossRef]
  23. Elamin, M.B.; Abu Elnour, N.O.; Elamin, K.B.; Fatourechi, M.M.; Alkatib, A.A.; Almandoz, J.P.; Liu, H.; Lane, M.A.; Mullan, R.J.; Hazem, A.; et al. Vitamin D and Cardiovascular Outcomes: A Systematic Review and Meta-Analysis. J. Clin. Endocrinol. Metab. 2011, 96, 1931–1942. [Google Scholar] [CrossRef]
  24. Turck, D.; Bohn, T.; Castenmiller, J.; de Henauw, S.; Hirsch-Ernst, K.; Knutsen, H.K.; Maciuk, A.; Mangelsdorf, I.; McArdle, H.J.; Pentieva, K.; et al. Scientific opinion on the tolerable upper intake level for vitamin D, including the derivation of a conversion factor for calcidiol monohydrate. EFSA J. 2023, 21, e08145. [Google Scholar] [CrossRef] [PubMed]
  25. Holick, M.F.; Biancuzzo, R.M.; Chen, T.C.; Klein, E.K.; Young, A.; Bibuld, D.; Reitz, R.; Salameh, W.; Ameri, A.; Tannenbaum, A.D. Vitamin D2 is as effective as vitamin D3 in maintaining circulating concentrations of 25-hydroxyvitamin D. J. Clin. Endocrinol. Metab. 2008, 93, 677–681. [Google Scholar] [CrossRef]
  26. Armas, L.A.G.; Hollis, B.W.; Heaney, R.P. Vitamin D2 is much less effective than vitamin D3 in humans. J. Clin. Endocrinol. Metab. 2004, 89, 5387–5391. [Google Scholar] [CrossRef]
  27. Alayed Albarri, E.M.; Sameer Alnuaimi, A.; Abdelghani, D. Effectiveness of vitamin D2 compared with vitamin D3 replacement therapy in a primary healthcare setting: A retrospective cohort study. Qatar Med. J. 2022, 2022, 29. [Google Scholar] [CrossRef]
  28. Tripkovic, L.; Lambert, H.; Hart, K.; Smith, C.P.; Bucca, G.; Penson, S.; Chope, G.; Hyppönen, E.; Berry, J.; Vieth, R.; et al. Comparison of vitamin D2 and vitamin D3 supplementation in raising serum 25-hydroxyvitamin D status: A systematic review and meta-analysis. Am. J. Clin. Nutr. 2012, 95, 1357–1364. [Google Scholar] [CrossRef] [PubMed]
  29. Lehmann, U.; Hirche, F.; Stangl, G.I.; Hinz, K.; Westphal, S.; Dierkes, J. Bioavailability of vitamin D2 and D3 in healthy volunteers, a randomized placebo-controlled trial. J. Clin. Endocrinol. Metab. 2013, 98, 4339–4345. [Google Scholar] [CrossRef]
  30. Jones, G. Extrarenal Vitamin D Activation and Interactions between Vitamin D2, Vitamin D3, and Vitamin D Analogs. Annu. Rev. Nutr. 2013, 33, 23–44. [Google Scholar] [CrossRef]
  31. Silva, M.C.; Furlanetto, T.W. Intestinal absorption of vitamin D: A systematic review. Nutr. Rev. 2018, 76, 60–76. [Google Scholar] [CrossRef]
  32. Durrant, L.R.; Bucca, G.; Hesketh, A.; Möller-Levet, C.; Tripkovic, L.; Wu, H.; Hart, K.H.; Mathers, J.C.; Elliott, R.M.; Lanham-New, S.A.; et al. Vitamins D2 and D3 Have Overlapping But Different Effects on the Human Immune System Revealed Through Analysis of the Blood Transcriptome. Front. Immunol. 2022, 13, 790444. [Google Scholar] [CrossRef] [PubMed]
  33. Mazzaferro, S.; Goldsmith, D.; Larsson, T.E.; Massy, Z.A.; Cozzolino, M. Vitamin D metabolites and/or analogs: Which D for which patient? Curr. Vasc. Pharmacol. 2014, 12, 339–349. [Google Scholar] [CrossRef] [PubMed]
  34. Martin, A.; David, V.; Quarles, L.D. Regulation and Function of the FGF23/Klotho Endocrine Pathways. Physiol. Rev. 2012, 92, 131–155. [Google Scholar] [CrossRef] [PubMed]
  35. Sato, T.; Courbebaisse, M.; Ide, N.; Fan, Y.; Hanai, J.-I.; Kaludjerovic, J.; Densmore, M.J.; Yuan, Q.; Toka, H.R.; Pollak, M.R.; et al. Parathyroid hormone controls paracellular Ca2+ transport in the thick ascending limb by regulating the tight-junction protein Claudin14. Proc. Natl. Acad. Sci. USA 2017, 114, E3344–E3353. [Google Scholar] [CrossRef] [PubMed]
  36. White, K.E.; Evans, W.E.; O’Riordan, J.L.H.; Speer, M.C.; Econs, M.J.; Lorenz-Depiereux, B.; Grabowski, M.; Meitinger, T.; Strom, T.M. Autosomal dominant hypophosphataemic rickets is associated with mutations in FGF23. Nat. Genet. 2000, 26, 345–348. [Google Scholar] [CrossRef]
  37. Masuyama, R.; Stockmans, I.; Torrekens, S.; Van Looveren, R.; Maes, C.; Carmeliet, P.; Bouillon, R.; Carmeliet, G. Vitamin D receptor in chondrocytes promotes osteoclastogenesis and regulates FGF23 production in osteoblasts. J. Clin. Investig. 2006, 116, 3150–3159. [Google Scholar] [CrossRef] [PubMed]
  38. Kolek, O.I.; Hines, E.R.; Jones, M.D.; LeSueur, L.K.; Lipko, M.A.; Kiela, P.R.; Collins, J.F.; Haussler, M.R.; Ghishan, F.K. 1alpha,25-Dihydroxyvitamin D3 upregulates FGF23 gene expression in bone: The final link in a renal-gastrointestinal-skeletal axis that controls phosphate transport. Am. J. Physiol. Gastrointest. Liver Physiol. 2005, 289, G1036–G1042. [Google Scholar] [CrossRef] [PubMed]
  39. Shimada, T.; Mizutani, S.; Muto, T.; Yoneya, T.; Hino, R.; Takeda, S.; Takeuchi, Y.; Fujita, T.; Fukumoto, S.; Yamashita, T. Cloning and characterization of FGF23 as a causative factor of tumor-induced osteomalacia. Proc. Natl. Acad. Sci. USA 2001, 98, 6500–6505. [Google Scholar] [CrossRef]
  40. Han, X.; Yang, J.; Li, L.; Huang, J.; King, G.; Quarles, L.D. Conditional Deletion of Fgfr1 in the Proximal and Distal Tubule Identifies Distinct Roles in Phosphate and Calcium Transport. PLoS ONE 2016, 11, e0147845. [Google Scholar] [CrossRef]
  41. Nguyen-Yamamoto, L.; Karaplis, A.C.; St–Arnaud, R.; Goltzman, D. Fibroblast Growth Factor 23 Regulation by Systemic and Local Osteoblast-Synthesized 1,25-Dihydroxyvitamin D. J. Am. Soc. Nephrol. 2017, 28, 586–597. [Google Scholar] [CrossRef] [PubMed]
  42. Walters, M.; Wicker, D.; Riggle, P. 1,25-Dihydroxyvitamin D3 receptors identified in the rat heart. J. Mol. Cell. Cardiol. 1986, 18, 67–72. [Google Scholar] [CrossRef] [PubMed]
  43. Schmidt, N.; Brandsch, C.; Kühne, H.; Thiele, A.; Hirche, F.; Stangl, G.I. Vitamin D Receptor Deficiency and Low Vitamin D Diet Stimulate Aortic Calcification and Osteogenic Key Factor Expression in Mice. PLoS ONE 2012, 7, e35316. [Google Scholar] [CrossRef] [PubMed]
  44. Merke, J.; Milde, P.; Lewicka, S.; Hügel, 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]
  45. Chen, S.; Glenn, D.J.; Ni, W.; Grigsby, C.L.; Olsen, K.; Nishimoto, M.; Law, C.S.; Gardner, D.G. Expression of the Vitamin D Receptor Is Increased in the Hypertrophic Heart. Hypertension 2008, 52, 1106–1112. [Google Scholar] [CrossRef] [PubMed]
  46. 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]
  47. Uberti, F.; Lattuada, D.; Morsanuto, V.; Nava, U.; Bolis, G.; Vacca, G.; Squarzanti, D.F.; Cisari, C.; Molinari, C. Vitamin D protects human endothelial cells from oxidative stress through the autophagic and survival pathways. J. Clin. Endocrinol. Metab. 2014, 99, 1367–1374. [Google Scholar] [CrossRef] [PubMed]
  48. Teixeira, T.M.; da Costa, D.C.; Resende, A.C.; Soulage, C.O.; Bezerra, F.F.; Daleprane, J.B. Activation of Nrf2-Antioxidant Signaling by 1,25-Dihydroxycholecalciferol Prevents Leptin-Induced Oxidative Stress and Inflammation in Human Endothelial Cells. J. Nutr. 2017, 147, 506–513. [Google Scholar] [CrossRef] [PubMed]
  49. Laera, N.; Malerba, P.; Vacanti, G.; Nardin, S.; Pagnesi, M.; Nardin, M. Impact of Immunity on Coronary Artery Disease: An Updated Pathogenic Interplay and Potential Therapeutic Strategies. Life 2023, 13, 2128. [Google Scholar] [CrossRef]
  50. Zhang, H.; Teng, J.; Li, Y.; Li, X.; He, Y.; He, X.; Sun, C. Vitamin D status and its association with adiposity and oxidative stress in schoolchildren. Nutrition 2014, 30, 1040–1044. [Google Scholar] [CrossRef]
  51. Cojic, M.; Kocic, R.; Klisic, A.; Kocic, G. The Effects of Vitamin D Supplementation on Metabolic and Oxidative Stress Markers in Patients with Type 2 Diabetes: A 6-Month Follow Up Randomized Controlled Study. Front. Endocrinol. 2021, 12, 610893. [Google Scholar] [CrossRef]
  52. Reynolds, J.A.; Haque, S.; Williamson, K.; Ray, D.W.; Alexander, M.Y.; Bruce, I.N. Vitamin D improves endothelial dysfunction and restores myeloid angiogenic cell function via reduced CXCL-10 expression in systemic lupus erythematosus. Sci. Rep. 2016, 6, 22341. [Google Scholar] [CrossRef]
  53. Mansournia, M.; Ostadmohammadi, V.; Doosti-Irani, A.; Ghayour-Mobarhan, M.; Ferns, G.; Akbari, H.; Ghaderi, A.; Talari, H.; Asemi, Z. The Effects of Vitamin D Supplementation on Biomarkers of Inflammation and Oxidative Stress in Diabetic Patients: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Horm. Metab. Res. 2018, 50, 429–440. [Google Scholar] [CrossRef] [PubMed]
  54. Moslemi, E.; Musazadeh, V.; Kavyani, Z.; Naghsh, N.; Shoura, S.M.S.; Dehghan, P. Efficacy of vitamin D supplementation as an adjunct therapy for improving inflammatory and oxidative stress biomarkers: An umbrella meta-analysis. Pharmacol. Res. 2022, 186, 106484. [Google Scholar] [CrossRef] [PubMed]
  55. Verdoia, M.; Nardin, M.; Rolla, R.; Negro, F.; Gioscia, R.; Saghir Afifeh, A.M.; Viglione, F.; Suryapranata, H.; Marcolongo, M.; De Luca, G. Cholecalciferol levels, inflammation and leukocytes parameters: Results from a large single-centre cohort of patients. Clin. Nutr. 2021, 40, 2228–2236. [Google Scholar] [CrossRef]
  56. Mahabadi-Ashtiyani, E.; Sheikh, V.; Borzouei, S.; Salehi, I.; Alahgholi-Hajibehzad, M. The increased T helper cells proliferation and inflammatory responses in patients with type 2 diabetes mellitus is suppressed by sitagliptin and vitamin D3 in vitro. Inflamm. Res. 2019, 68, 857–866. [Google Scholar] [CrossRef]
  57. Wang, W.; Zhang, J.; Wang, H.; Wang, X.; Liu, S. Vitamin D deficiency enhances insulin resistance by promoting inflammation in type 2 diabetes. Int. J. Clin. Exp. Pathol. 2019, 12, 1859–1867. [Google Scholar] [PubMed]
  58. Verdoia, M.; Nardin, M.; Rolla, R.; Negro, F.; Gioscia, R.; Afifeh, A.M.S.; Viglione, F.; Suryapranata, H.; Marcolongo, M.; De Luca, G.; et al. Association of lower vitamin D levels with inflammation and leucocytes parameters in patients with and without diabetes mellitus undergoing coronary angiography. Eur. J. Clin. Investig. 2021, 51, e13439. [Google Scholar] [CrossRef]
  59. Dionne, S.; Duchatelier, C.-F.; Seidman, E.G. The influence of vitamin D on M1 and M2 macrophages in patients with Crohn’s disease. Innate Immun. 2017, 23, 557–565. [Google Scholar] [CrossRef]
  60. Zhang, Y.; Leung, D.Y.M.; Richers, B.N.; Liu, Y.; Remigio, L.K.; Riches, D.W.; Goleva, E. Vitamin D Inhibits Monocyte/Macrophage Proinflammatory Cytokine Production by Targeting MAPK Phosphatase-1. J. Immunol. 2012, 188, 2127–2135. [Google Scholar] [CrossRef]
  61. Zhang, X.; Zhou, M.; Guo, Y.; Song, Z.; Liu, B. 1,25-Dihydroxyvitamin D 3 Promotes High Glucose-Induced M1 Macrophage Switching to M2 via the VDR-PPAR γ Signaling Pathway. Biomed Res. Int. 2015, 2015, 1–14. [Google Scholar] [CrossRef]
  62. Chen, C.; Liu, Z.; Huang, X. Anaerobic membrane bioreactors for sustainable and energy-efficient municipal wastewater treatment. In Current Developments in Biotechnology and Bioengineering; Elsevier, 2020; pp. 335–366.
  63. Nardin, M.; Verdoia, M.; Cao, D.; Nardin, S.; Kedhi, E.; Galasso, G.; van ’t Hof, A.W.J.; Condorelli, G.; De Luca, G. Platelets and the Atherosclerotic Process: An Overview of New Markers of Platelet Activation and Reactivity, and Their Implications in Primary and Secondary Prevention. J. Clin. Med. 2023, 12, 6074. [Google Scholar] [CrossRef]
  64. Staeva-Vieira, T.P.; Freedman, L.P. 1,25-Dihydroxyvitamin D3 Inhibits IFN-γ and IL-4 Levels During In Vitro Polarization of Primary Murine CD4+ T Cells. J. Immunol. 2002, 168, 1181–1189. [Google Scholar] [CrossRef] [PubMed]
  65. Chang, S.H.; Chung, Y.; Dong, C. Vitamin D Suppresses Th17 Cytokine Production by Inducing C/EBP Homologous Protein (CHOP) Expression. J. Biol. Chem. 2010, 285, 38751–38755. [Google Scholar] [CrossRef] [PubMed]
  66. Hau, C.S.; Shimizu, T.; Tada, Y.; Kamata, M.; Takeoka, S.; Shibata, S.; Mitsui, A.; Asano, Y.; Sugaya, M.; Kadono, T.; et al. The vitamin D3 analog, maxacalcitol, reduces psoriasiform skin inflammation by inducing regulatory T cells and downregulating IL-23 and IL-17 production. J. Dermatol. Sci. 2018, 92, 117–126. [Google Scholar] [CrossRef] [PubMed]
  67. Vanherwegen, A.-S.; Eelen, G.; Ferreira, G.B.; Ghesquière, B.; Cook, D.P.; Nikolic, T.; Roep, B.; Carmeliet, P.; Telang, S.; Mathieu, C.; et al. Vitamin D controls the capacity of human dendritic cells to induce functional regulatory T cells by regulation of glucose metabolism. J. Steroid Biochem. Mol. Biol. 2019, 187, 134–145. [Google Scholar] [CrossRef] [PubMed]
  68. Daryabor, G.; Gholijani, N.; Kahmini, F.R. A review of the critical role of vitamin D axis on the immune system. Exp. Mol. Pathol. 2023, 132–133, 104866. [Google Scholar] [CrossRef] [PubMed]
  69. Giulietti, A.; van Etten, E.; Overbergh, L.; Stoffels, K.; Bouillon, R.; Mathieu, C. Monocytes from type 2 diabetic patients have a pro-inflammatory profile. Diabetes Res. Clin. Pract. 2007, 77, 47–57. [Google Scholar] [CrossRef] [PubMed]
  70. Sharma, G.; She, Z.-G.; Valenta, D.T.; Stallcup, W.B.; Smith, J.W. Scavenger receptor-mediated targeting of macrophage foam cells in atherosclerotic plaque using oligonucleotide-functionalized nanoparticles. Nano Life 2010, 01, 207–214. [Google Scholar] [CrossRef]
  71. Suematsu, Y.; Nishizawa, Y.; Shioi, A.; Hino, M.; Tahara, H.; Inaba, M.; Morii, H.; Otani, S. Effect of 1,25-dihydroxyvitamin D3 on induction of scavenger receptor and differentiation of 12-O-tetradecanoylphorbol-13-acetate-treated THP-1 human monocyte like cells. J. Cell. Physiol. 1995, 165, 547–555. [Google Scholar] [CrossRef]
  72. Oh, J.; Weng, S.; Felton, S.K.; Bhandare, S.; Riek, A.; Butler, B.; Proctor, B.M.; Petty, M.; Chen, Z.; Schechtman, K.B.; et al. 1,25(OH)2 vitamin d inhibits foam cell formation and suppresses macrophage cholesterol uptake in patients with type 2 diabetes mellitus. Circulation 2009, 120, 687–698. [Google Scholar] [CrossRef] [PubMed]
  73. Barreto, D.V.; Barreto, F.C.; Liabeuf, S.; Temmar, M.; Boitte, F.; Choukroun, G.; Fournier, A.; Massy, Z.A. Vitamin D affects survival independently of vascular calcification in chronic kidney disease. Clin. J. Am. Soc. Nephrol. 2009, 4, 1128–1135. [Google Scholar] [CrossRef] [PubMed]
  74. Rodríguez, A.J.; Scott, D.; Srikanth, V.; Ebeling, P. Effect of vitamin D supplementation on measures of arterial stiffness: A systematic review and meta-analysis of randomized controlled trials. Clin. Endocrinol. 2016, 84, 645–657. [Google Scholar] [CrossRef] [PubMed]
  75. Nardin, M.; Verdoia, M.; Laera, N.; Cao, D.; De Luca, G. New Insights into Pathophysiology and New Risk Factors for ACS. J. Clin. Med. 2023, 12, 2883. [Google Scholar] [CrossRef] [PubMed]
  76. Giovannucci, E.; Liu, Y.; Hollis, B.W.; Rimm, E.B. 25-hydroxyvitamin D and risk of myocardial infarction in men: A prospective study. Arch. Intern. Med. 2008, 168, 1174–1180. [Google Scholar] [CrossRef]
  77. Wang, L.; Song, Y.; Manson, J.E.; Pilz, S.; März, W.; Michaëlsson, K.; Lundqvist, A.; Jassal, S.K.; Barrett-Connor, E.; Zhang, C.; et al. Circulating 25-hydroxy-vitamin D and risk of cardiovascular disease: A meta-analysis of prospective studies. Circ. Cardiovasc. Qual. Outcomes 2012, 5, 819–829. [Google Scholar] [CrossRef] [PubMed]
  78. Verdoia, M.; Schaffer, A.; Sartori, C.; Barbieri, L.; Cassetti, E.; Marino, P.; Galasso, G.; De Luca, G. Vitamin D deficiency is independently associated with the extent of coronary artery disease. Eur. J. Clin. Investig. 2014, 44, 634–642. [Google Scholar] [CrossRef]
  79. Nardin, M.; Verdoia, M.; Schaffer, A.; Barbieri, L.; Marino, P.; De Luca, G. Novara Atherosclerosis Study Group (NAS) Vitamin D status, diabetes mellitus and coronary artery disease in patients undergoing coronary angiography. Atherosclerosis 2016, 250, 114–121. [Google Scholar] [CrossRef] [PubMed]
  80. Verdoia, M.; Viglione, F.; Boggio, A.; Stefani, D.; Panarotto, N.; Malabaila, A.; Rolla, R.; Soldà, P.L.; Stecco, A.; Carriero, A.; et al. Relationship between vitamin D and cholesterol levels in STEMI patients undergoing primary percutaneous coronary intervention. Nutr. Metab. Cardiovasc. Dis. 2022, 32, 957–964. [Google Scholar] [CrossRef]
  81. Saghir Afifeh, A.M.; Verdoia, M.; Nardin, M.; Negro, F.; Viglione, F.; Rolla, R.; De Luca, G. Novara Atherosclerosis Study Group (NAS) Determinants of vitamin D activation in patients with acute coronary syndromes and its correlation with inflammatory markers. Nutr. Metab. Cardiovasc. Dis. 2021, 31, 36–43. [Google Scholar] [CrossRef]
  82. Robinson-Cohen, C.; Hoofnagle, A.N.; Ix, J.H.; Sachs, M.C.; Tracy, R.P.; Siscovick, D.S.; Kestenbaum, B.R.; de Boer, I.H. Racial differences in the association of serum 25-hydroxyvitamin D concentration with coronary heart disease events. JAMA 2013, 310, 179–188. [Google Scholar] [CrossRef] [PubMed]
  83. Correia, L.C.L.; Sodré, F.; Garcia, G.; Sabino, M.; Brito, M.; Kalil, F.; Barreto, B.; Lima, J.C.; Noya-Rabelo, M.M. Relation of severe deficiency of vitamin D to cardiovascular mortality during acute coronary syndromes. Am. J. Cardiol. 2013, 111, 324–327. [Google Scholar] [CrossRef]
  84. Verdoia, M.; Nardin, M.; Rolla, R.; Negro, F.; Gioscia, R.; Afifeh, A.M.S.; Viglione, F.; Suryapranata, H.; Marcolongo, M.; De Luca, G.; et al. Prognostic impact of Vitamin D deficiency in patients with coronary artery disease undergoing percutaneous coronary intervention. Eur. J. Intern. Med. 2021, 83, 62–67. [Google Scholar] [CrossRef] [PubMed]
  85. De Metrio, M.; Milazzo, V.; Rubino, M.; Cabiati, A.; Moltrasio, M.; Marana, I.; Campodonico, J.; Cosentino, N.; Veglia, F.; Bonomi, A.; et al. Vitamin D plasma levels and in-hospital and 1-year outcomes in acute coronary syndromes: A prospective study. Medicine 2015, 94, e857. [Google Scholar] [CrossRef] [PubMed]
  86. Verdoia, M.; Viglione, F.; Boggio, A.; Stefani, D.; Panarotto, N.; Malabaila, A.; Rolla, R.; Soldà, P.L.; De Luca, G. Novara Atherosclerosis Study Group (NAS) Vitamin D deficiency is associated with impaired reperfusion in STEMI patients undergoing primary percutaneous coronary intervention. Vascul. Pharmacol. 2021, 140, 106897. [Google Scholar] [CrossRef] [PubMed]
  87. Verdoia, M.; Ceccon, C.; Nardin, M.; Suryapranata, H.; De Luca, G. Novara Atherosclerosis Study Group (NAS) Vitamin D deficiency and periprocedural myocardial infarction in patients undergoing percutaneous coronary interventions. Cardiovasc. Revasc. Med. 2018, 19, 744–750. [Google Scholar] [CrossRef] [PubMed]
  88. De Luca, G.; Suryapranata, H.; Stone, G.W.; Antoniucci, D.; Tcheng, J.E.; Neumann, F.-J.; Bonizzoni, E.; Topol, E.J.; Chiariello, M. Relationship between patient’s risk profile and benefits in mortality from adjunctive abciximab to mechanical revascularization for ST-segment elevation myocardial infarction: A meta-regression analysis of randomized trials. J. Am. Coll. Cardiol. 2006, 47, 685–686. [Google Scholar] [CrossRef] [PubMed]
  89. Padoan, L.; Beltrami, A.P.; Stenner, E.; Beleù, A.; Ruscio, M.; Sinagra, G.; Aleksova, A. Left ventricular adverse remodeling after myocardial infarction and its association with vitamin D levels. Int. J. Cardiol. 2019, 277, 159–165. [Google Scholar] [CrossRef] [PubMed]
  90. Koyama, T.; Shibakura, M.; Ohsawa, M.; Kamiyama, R.; Hirosawa, S. Anticoagulant effects of 1alpha,25-dihydroxyvitamin D3 on human myelogenous leukemia cells and monocytes. Blood 1998, 92, 160–167. [Google Scholar] [CrossRef]
  91. López-Farré, A.J.; Mateos-Cáceres, P.J.; Sacristán, D.; Azcona, L.; Bernardo, E.; de Prada, T.P.; Alonso-Orgaz, S.; Fernández-Arquero, M.; Fernández-Ortiz, A.; Macaya, C. Relationship between vitamin D binding protein and aspirin resistance in coronary ischemic patients: A proteomic study. J. Proteome Res. 2007, 6, 2481–2487. [Google Scholar] [CrossRef]
  92. Verdoia, M.; Pergolini, P.; Rolla, R.; Sartori, C.; Nardin, M.; Schaffer, A.; Barbieri, L.; Daffara, V.; Marino, P.; Bellomo, G.; et al. Vitamin D levels and high-residual platelet reactivity in patients receiving dual antiplatelet therapy with clopidogrel or ticagrelor. Platelets 2016, 27, 576–582. [Google Scholar] [CrossRef] [PubMed]
  93. Verdoia, M.; Pergolini, P.; Nardin, M.; Rolla, R.; Negro, F.; Kedhi, E.; Suryapranata, H.; Marcolongo, M.; Carriero, A.; De Luca, G.; et al. Vitamin D levels and platelet reactivity in diabetic patients receiving dual antiplatelet therapy. Vascul. Pharmacol. 2019, 120, 106564. [Google Scholar] [CrossRef] [PubMed]
  94. Verdoia, M.; De Luca, G. Is there an actual link between vitamin D deficiency, cardiovascular disease, and glycemic control in patients with type 2 diabetes mellitus? Polish Arch. Intern. Med. 2023, 133, 16516. [Google Scholar] [CrossRef] [PubMed]
  95. Sultan, M.; Twito, O.; Tohami, T.; Ramati, E.; Neumark, E.; Rashid, G. Vitamin D diminishes the high platelet aggregation of type 2 diabetes mellitus patients. Platelets 2019, 30, 120–125. [Google Scholar] [CrossRef] [PubMed]
  96. Verdoia, M.; Nardin, M.; Gioscia, R.; Saghir Afifeh, A.M.; Viglione, F.; Negro, F.; Marcolongo, M.; De Luca, G. Novara Atherosclerosis Study Group (NAS) Association between vitamin D deficiency and serum Homocysteine levels and its relationship with coronary artery disease. J. Thromb. Thrombolysis 2021, 52, 523–531. [Google Scholar] [CrossRef] [PubMed]
  97. Verdoia, M.; Pergolini, P.; Rolla, R.; Nardin, M.; Barbieri, L.; Schaffer, A.; Bellomo, G.; Marino, P.; Suryapranata, H.; De Luca, G.; et al. Parathyroid Hormone Levels and High-Residual Platelet Reactivity in Patients Receiving Dual Antiplatelet Therapy with Acetylsalicylic Acid and Clopidogrel or Ticagrelor. Cardiovasc. Ther. 2016, 34, 209–215. [Google Scholar] [CrossRef] [PubMed]
  98. Li, Y.C.; Kong, J.; Wei, M.; Chen, Z.-F.; Liu, S.Q.; Cao, L.-P. 1,25-Dihydroxyvitamin D3 is a negative endocrine regulator of the renin-angiotensin system. J. Clin. Investig. 2002, 110, 229–238. [Google Scholar] [CrossRef] [PubMed]
  99. Verdoia, M.; Nardin, M.; Rolla, R.; Negro, F.; Gioscia, R.; Saghir Afifeh, A.M.; Viglione, F.; Suryapranata, H.; Marcolongo, M.; De Luca, G.; et al. Vitamin D levels condition the outcome benefits of renin-angiotensin system inhibitors (RASI) among patients undergoing percutaneous coronary intervention. Pharmacol. Res. 2020, 160, 105158. [Google Scholar] [CrossRef] [PubMed]
  100. 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]
  101. 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]
  102. Wong, M.S.K.; Delansorne, R.; Man, R.Y.K.; 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. Circ. Physiol. 2010, 299, H1226–H1234. [Google Scholar] [CrossRef] [PubMed]
  103. SCRAGG, R.; SOWERS, M.; BELL, C. Serum 25-hydroxyvitamin D, Ethnicity, and Blood Pressure in the Third National Health and Nutrition Examination Survey. Am. J. Hypertens. 2007, 20, 713–719. [Google Scholar] [CrossRef] [PubMed]
  104. He, J.L.; Scragg, R.K. Vitamin D, Parathyroid Hormone, and Blood Pressure in the National Health and Nutrition Examination Surveys. Am. J. Hypertens. 2011, 24, 911–917. [Google Scholar] [CrossRef] [PubMed]
  105. Wang, T.J.; Pencina, M.J.; Booth, S.L.; Jacques, P.F.; Ingelsson, E.; Lanier, K.; Benjamin, E.J.; D’Agostino, R.B.; Wolf, M.; Vasan, R.S. Vitamin D Deficiency and Risk of Cardiovascular Disease. Circulation 2008, 117, 503–511. [Google Scholar] [CrossRef] [PubMed]
  106. Judd, S.E.; Nanes, M.S.; Ziegler, T.R.; Wilson, P.W.; Tangpricha, V. Optimal vitamin D status attenuates the age-associated increase in systolic blood pressure in white Americans: Results from the third National Health and Nutrition Examination Survey. Am. J. Clin. Nutr. 2008, 87, 136–141. [Google Scholar] [CrossRef] [PubMed]
  107. Kunutsor, S.K.; Apekey, T.A.; Steur, M. Vitamin D and risk of future hypertension: Meta-analysis of 283,537 participants. Eur. J. Epidemiol. 2013, 28, 205–221. [Google Scholar] [CrossRef] [PubMed]
  108. Snijder, M.B.; Lips, P.; Seidell, J.C.; Visser, M.; Deeg, D.J.H.; Dekker, J.M.; Van Dam, R.M. Vitamin D status and parathyroid hormone levels in relation to blood pressure: A population-based study in older men and women. J. Intern. Med. 2007, 261, 558–565. [Google Scholar] [CrossRef] [PubMed]
  109. Reis, J.P.; von Mühlen, D.; Kritz-Silverstein, D.; Wingard, D.L.; Barrett-Connor, E. Vitamin D, Parathyroid Hormone Levels, and the Prevalence of Metabolic Syndrome in Community-Dwelling Older Adults. Diabetes Care 2007, 30, 1549–1555. [Google Scholar] [CrossRef] [PubMed]
  110. Stewart, B.F.; Siscovick, D.; Lind, B.K.; Gardin, J.M.; Gottdiener, J.S.; Smith, V.E.; Kitzman, D.W.; Otto, C.M. Clinical Factors Associated with Calcific Aortic Valve Disease. J. Am. Coll. Cardiol. 1997, 29, 630–634. [Google Scholar] [CrossRef]
  111. Zittermann, A.; Schleithoff, S.S.; Koerfer, R. Vitamin D and vascular calcification. Curr. Opin. Lipidol. 2007, 18, 41–46. [Google Scholar] [CrossRef]
  112. Colleville, B.; Perzo, N.; Avinée, G.; Dumesnil, A.; Ziegler, F.; Billoir, P.; Eltchaninoff, H.; Richard, V.; Durand, E. Impact of high-fat diet and vitamin D3 supplementation on aortic stenosis establishment in waved-2 epidermal growth factor receptor mutant mice. J. Integr. Med. 2019, 17, 107–114. [Google Scholar] [CrossRef] [PubMed]
  113. Vadana, M.; Cecoltan, S.; Ciortan, L.; Macarie, R.D.; Mihaila, A.C.; Tucureanu, M.M.; Gan, A.-M.; Simionescu, M.; Manduteanu, I.; Droc, I.; et al. Parathyroid Hormone Induces Human Valvular Endothelial Cells Dysfunction That Impacts the Osteogenic Phenotype of Valvular Interstitial Cells. Int. J. Mol. Sci. 2022, 23, 3776. [Google Scholar] [CrossRef]
  114. Tsuruda, T.; Funamoto, T.; Suzuki, C.; Yamamura, Y.; Nakai, M.; Chosa, E.; Kaikita, K. Increasing baseline aortic valve peak flow velocity is associated with progression of aortic valve stenosis in osteoporosis patients-a possible link to low vitamin D status. Arch. Osteoporos. 2023, 18, 129. [Google Scholar] [CrossRef]
  115. Kassis, N.; Hariri, E.H.; Karrthik, A.K.; Ahuja, K.R.; Layoun, H.; Saad, A.M.; Gad, M.M.; Kaur, M.; Bazarbashi, N.; Griffin, B.P.; et al. Supplemental calcium and vitamin D and long-term mortality in aortic stenosis. Heart 2022, 108, 964–972. [Google Scholar] [CrossRef] [PubMed]
  116. Xia, C.; Lei, W.; Hu, Y.; Yang, H.; Zeng, X.; Chen, M. Association of serum levels of calcium, phosphate, and vitamin D with risk of developing aortic stenosis: The UK Biobank cohort. Eur. J. Prev. Cardiol. 2022, 29, 1520–1528. [Google Scholar] [CrossRef]
  117. Huang, N.; Zhuang, Z.; Liu, Z.; Huang, T. Observational and Genetic Associations of Modifiable Risk Factors with Aortic Valve Stenosis: A Prospective Cohort Study of 0.5 Million Participants. Nutrients 2022, 14, 2273. [Google Scholar] [CrossRef] [PubMed]
  118. Butcher, J.T.; Nerem, R.M. Valvular endothelial cells regulate the phenotype of interstitial cells in co-culture: Effects of steady shear stress. Tissue Eng. 2006, 12, 905–915. [Google Scholar] [CrossRef]
  119. McDonagh, T.A.; Metra, M.; Adamo, M.; Gardner, R.S.; Baumbach, A.; Böhm, M.; Burri, H.; Butler, J.; Čelutkienė, J.; Chioncel, O.; et al. 2021 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure. Eur. Heart J. 2021, 42, 3599–3726. [Google Scholar] [CrossRef]
  120. Biernacka, A.; Frangogiannis, N.G. Aging and Cardiac Fibrosis. Aging Dis. 2011, 2, 158–173. [Google Scholar]
  121. Artaza, J.N.; Norris, K.C. Vitamin D reduces the expression of collagen and key profibrotic factors by inducing an antifibrotic phenotype in mesenchymal multipotent cells. J. Endocrinol. 2009, 200, 207–221. [Google Scholar] [CrossRef]
  122. 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]
  123. Ameri, P.; Canepa, M.; Milaneschi, Y.; Spallarossa, P.; Leoncini, G.; Giallauria, F.; Strait, J.B.; Lakatta, E.G.; Brunelli, C.; Murialdo, G.; et al. Relationship between vitamin D status and left ventricular geometry in a healthy population: Results from the Baltimore L ongitudinal Study of Aging. J. Intern. Med. 2013, 273, 253–262. [Google Scholar] [CrossRef]
  124. Verdoia, M.; Solli, M.; Ubertini, E.; Erbetta, R.; Gioscia, R.; Afifeh, A.M.S.; Viglione, F.; Rolla, R.; De Luca, G. Low vitamin D levels affect left ventricular wall thickness in severe aortic stenosis. J. Cardiovasc. Med. 2020, 21, 905–911. [Google Scholar] [CrossRef]
  125. Seker, T.; Gur, M.; Ucar, H.; Turkoglu, C.; Baykan, A.O.; Özaltun, B.; Harbalioglu, H.; Yuksel Kalkan, G.; Kaypakli, O.; Kuloglu, O.; et al. Lower serum 25-hydroxyvitamin D level is associated with impaired myocardial performance and left ventricle hypertrophy in newly diagnosed hypertensive patients. Anatol. J. Cardiol. 2015, 15, 744–750. [Google Scholar] [CrossRef] [PubMed]
  126. Anderson, J.L.; May, H.T.; Horne, B.D.; Bair, T.L.; Hall, N.L.; Carlquist, J.F.; Lappé, D.L.; Muhlestein, J.B. Intermountain Heart Collaborative (IHC) Study Group Relation of vitamin D deficiency to cardiovascular risk factors, disease status, and incident events in a general healthcare population. Am. J. Cardiol. 2010, 106, 963–968. [Google Scholar] [CrossRef] [PubMed]
  127. Nolte, K.; Herrmann-Lingen, C.; Platschek, L.; Holzendorf, V.; Pilz, S.; Tomaschitz, A.; Düngen, H.-D.; Angermann, C.E.; Hasenfuß, G.; Pieske, B.; et al. Vitamin D deficiency in patients with diastolic dysfunction or heart failure with preserved ejection fraction. ESC Hear. Fail. 2019, 6, 262–270. [Google Scholar] [CrossRef]
  128. Kim, D.H.; Sabour, S.; Sagar, U.N.; Adams, S.; Whellan, D.J. Prevalence of hypovitaminosis D in cardiovascular diseases (from the National Health and Nutrition Examination Survey 2001 to 2004). Am. J. Cardiol. 2008, 102, 1540–1544. [Google Scholar] [CrossRef] [PubMed]
  129. Liu, L.C.Y.; Voors, A.A.; van Veldhuisen, D.J.; van der Veer, E.; Belonje, A.M.; Szymanski, M.K.; Silljé, H.H.W.; van Gilst, W.H.; Jaarsma, T.; de Boer, R.A. Vitamin D status and outcomes in heart failure patients. Eur. J. Heart Fail. 2011, 13, 619–625. [Google Scholar] [CrossRef]
  130. Zhou, A.; Selvanayagam, J.B.; Hyppönen, E. Non-linear Mendelian randomization analyses support a role for vitamin D deficiency in cardiovascular disease risk. Eur. Heart J. 2022, 43, 1731–1739. [Google Scholar] [CrossRef]
  131. Schneider, A.L.C.; Lutsey, P.L.; Selvin, E.; Mosley, T.H.; Sharrett, A.R.; Carson, K.A.; Post, W.S.; Pankow, J.S.; Folsom, A.R.; Gottesman, R.F.; et al. Vitamin D, vitamin D binding protein gene polymorphisms, race and risk of incident stroke: The Atherosclerosis Risk in Communities (ARIC) study. Eur. J. Neurol. 2015, 22, 1220–1227. [Google Scholar] [CrossRef]
  132. Michos, E.D.; Misialek, J.R.; Selvin, E.; Folsom, A.R.; Pankow, J.S.; Post, W.S.; Lutsey, P.L. 25-hydroxyvitamin D levels, vitamin D binding protein gene polymorphisms and incident coronary heart disease among whites and blacks: The ARIC study. Atherosclerosis 2015, 241, 12–17. [Google Scholar] [CrossRef]
  133. Powe, C.E.; Evans, M.K.; Wenger, J.; Zonderman, A.B.; Berg, A.H.; Nalls, M.; Tamez, H.; Zhang, D.; Bhan, I.; Karumanchi, S.A.; et al. Vitamin D-binding protein and vitamin D status of black Americans and white Americans. N. Engl. J. Med. 2013, 369, 1991–2000. [Google Scholar] [CrossRef]
  134. Daffara, V.; Verdoia, M.; Rolla, R.; Nardin, M.; Marino, P.; Bellomo, G.; Carriero, A.; De Luca, G. Impact of polymorphism rs7041 and rs4588 of Vitamin D Binding Protein on the extent of coronary artery disease. Nutr. Metab. Cardiovasc. Dis. 2017, 27, 775–783. [Google Scholar] [CrossRef] [PubMed]
  135. Verdoia, M.; Daffara, V.; Pergolini, P.; Rolla, R.; Marino, P.; Bellomo, G.; Carriero, A.; De Luca, G. Vitamin D Binding Protein rs7041 polymorphism and high-residual platelet reactivity in patients receiving dual antiplatelet therapy with clopidogrel or ticagrelor. Vascul. Pharmacol. 2017, 93–95, 42–47. [Google Scholar] [CrossRef]
  136. Kulsoom, U.; Khan, A.; Saghir, T.; Nawab, S.N.; Tabassum, A.; Fatima, S.; Saleem, S.; Zehra, S. Vitamin D receptor gene polymorphism TaqI (rs731236) and its association with the susceptibility to coronary artery disease among Pakistani population. J. Gene Med. 2021, 23, e3386. [Google Scholar] [CrossRef]
  137. He, L.; Wang, M. Association of vitamin d receptor-a gene polymorphisms with coronary heart disease in Han Chinese. Int. J. Clin. Exp. Med. 2015, 8, 6224–6229. [Google Scholar] [PubMed]
  138. Ferrarezi, D.A.F.; Bellili-Muñoz, N.; Dubois-Laforgue, D.; Cheurfa, N.; Lamri, A.; Reis, A.F.; Le Feuvre, C.; Roussel, R.; Fumeron, F.; Timsit, J.; et al. Allelic variations of the vitamin D receptor (VDR) gene are associated with increased risk of coronary artery disease in type 2 diabetics: The DIABHYCAR prospective study. Diabetes Metab. 2013, 39, 263–270. [Google Scholar] [CrossRef]
  139. Sergeeva, E.G.; Ionova, Z.I. Association of BsmI and ApaI Polymorphisms of the Vitamin D Receptor Gene with Dyslipidemia in Patients with Coronary Artery Disease. J. Bioinforma. Diabetes 2020, 1, 12–19. [Google Scholar] [CrossRef]
  140. Akhlaghi, B.; Firouzabadi, N.; Foroughinia, F.; Nikparvar, M.; Dehghani, P. Impact of vitamin D receptor gene polymorphisms (TaqI and BsmI) on the incidence and severity of coronary artery disease: A report from southern Iran. BMC Cardiovasc. Disord. 2023, 23, 113. [Google Scholar] [CrossRef]
  141. Available online: https://www.ncbi.nlm.nih.gov/snp/rs2228570 (accessed on 25 March 2024).
  142. Fronczek, M.; Strzelczyk, J.K.; Osadnik, T.; Biernacki, K.; Ostrowska, Z. VDR Gene Polymorphisms in Healthy Individuals with Family History of Premature Coronary Artery Disease. Dis. Markers 2021, 2021, 8832478. [Google Scholar] [CrossRef]
  143. González Rojo, P.; Pérez Ramírez, C.; Gálvez Navas, J.M.; Pineda Lancheros, L.E.; Rojo Tolosa, S.; Ramírez Tortosa, M.d.C.; Jiménez Morales, A. Vitamin D-Related Single Nucleotide Polymorphisms as Risk Biomarker of Cardiovascular Disease. Int. J. Mol. Sci. 2022, 23, 8686. [Google Scholar] [CrossRef] [PubMed]
  144. Sun, H.; Long, S.R.; Li, X.; Ge, H.; Liu, X.; Wang, T.; Yu, F.; Wang, Y.; Xue, Y.; Li, W. Serum vitamin D deficiency and vitamin D receptor gene polymorphism are associated with increased risk of cardiovascular disease in a Chinese rural population. Nutr. Res. 2019, 61, 13–21. [Google Scholar] [CrossRef] [PubMed]
  145. Monraats, P.S.; Fang, Y.; Pons, D.; Pires, N.M.; Pols, H.A.; Zwinderman, A.H.; de Maat, M.P.; Doevendans, P.A.; DeWinter, R.J.; Tio, R.A.; et al. Vitamin D receptor: A new risk marker for clinical restenosis after percutaneous coronary intervention. Expert Opin. Ther. Targets 2010, 14, 243–251. [Google Scholar] [CrossRef] [PubMed]
  146. Ortlepp, J.R.; Hoffmann, R.; Ohme, F.; Lauscher, J.; Bleckmann, F.; Hanrath, P. The vitamin D receptor genotype predisposes to the development of calcific aortic valve stenosis. Heart 2001, 85, 635–638. [Google Scholar] [CrossRef]
  147. Brøndum-Jacobsen, P.; Benn, M.; Afzal, S.; Nordestgaard, B.G. No evidence that genetically reduced 25-hydroxyvitamin D is associated with increased risk of ischaemic heart disease or myocardial infarction: A Mendelian randomization study. Int. J. Epidemiol. 2015, 44, 651–661. [Google Scholar] [CrossRef] [PubMed]
  148. Afzal, S.; Brøndum-Jacobsen, P.; Bojesen, S.E.; Nordestgaard, B.G. Genetically low vitamin D concentrations and increased mortality: Mendelian randomisation analysis in three large cohorts. BMJ 2014, 349, g6330. [Google Scholar] [CrossRef]
  149. Manousaki, D.; Mokry, L.E.; Ross, S.; Goltzman, D.; Richards, J.B. Mendelian Randomization Studies Do Not Support a Role for Vitamin D in Coronary Artery Disease. Circ. Cardiovasc. Genet. 2016, 9, 349–356. [Google Scholar] [CrossRef]
  150. Rendina, D.; D Elia, L.; Abate, V.; Rebellato, A.; Buondonno, I.; Succoio, M.; Martinelli, F.; Muscariello, R.; De Filippo, G.; D Amelio, P.; et al. Vitamin D Status, Cardiovascular Risk Profile, and miRNA-21 Levels in Hypertensive Patients: Results of the HYPODD Study. Nutrients 2022, 14, 2683. [Google Scholar] [CrossRef]
  151. Mirhosseini, N.; Rainsbury, J.; Kimball, S.M. Vitamin D Supplementation, Serum 25(OH)D Concentrations and Cardiovascular Disease Risk Factors: A Systematic Review and Meta-Analysis. Front. Cardiovasc. Med. 2018, 5, 87. [Google Scholar] [CrossRef] [PubMed]
  152. Dibaba, D.T. Effect of vitamin D supplementation on serum lipid profiles: A systematic review and meta-analysis. Nutr. Rev. 2019, 77, 890–902. [Google Scholar] [CrossRef]
  153. Verdoia, M.; Pergolini, P.; Rolla, R.; Nardin, M.; Schaffer, A.; Barbieri, L.; Daffara, V.; Marino, P.; Bellomo, G.; Suryapranata, H.; et al. Impact of high-dose statins on vitamin D levels and platelet function in patients with coronary artery disease. Thromb. Res. 2017, 150, 90–95. [Google Scholar] [CrossRef] [PubMed]
  154. Verdoia, M.; Gioscia, R.; Nardin, M.; Rognoni, A.; De Luca, G. Low levels of vitamin D and coronary artery disease: Is it time for therapy? Kardiol. Pol. 2022, 80, 409–416. [Google Scholar] [CrossRef]
  155. Sokol, S.I.; Srinivas, V.; Crandall, J.P.; Kim, M.; Tellides, G.; Lebastchi, A.H.; Yu, Y.; Gupta, A.K.; Alderman, M.H. The effects of vitamin D repletion on endothelial function and inflammation in patients with coronary artery disease. Vasc. Med. 2012, 17, 394–404. [Google Scholar] [CrossRef]
  156. Witham, M.D.; Dove, F.J.; Khan, F.; Lang, C.C.; Belch, J.J.F.; Struthers, A.D. Effects of vitamin D supplementation on markers of vascular function after myocardial infarction—A randomised controlled trial. Int. J. Cardiol. 2013, 167, 745–749. [Google Scholar] [CrossRef]
  157. Wu, Z.; Wang, T.; Zhu, S.; Li, L. Effects of vitamin D supplementation as an adjuvant therapy in coronary artery disease patients. Scand. Cardiovasc. J. 2016, 50, 9–16. [Google Scholar] [CrossRef] [PubMed]
  158. Pfeifer, M.; Begerow, B.; Minne, H.W.; Nachtigall, D.; Hansen, C. Effects of a Short-Term Vitamin D3 and Calcium Supplementation on Blood Pressure and Parathyroid Hormone Levels in Elderly Women1. J. Clin. Endocrinol. Metab. 2001, 86, 1633–1637. [Google Scholar] [CrossRef] [PubMed]
  159. Kunutsor, S.K.; Burgess, S.; Munroe, P.B.; Khan, H. Vitamin D and high blood pressure: Causal association or epiphenomenon? Eur. J. Epidemiol. 2014, 29, 1–14. [Google Scholar] [CrossRef]
  160. Arora, P.; Song, Y.; Dusek, J.; Plotnikoff, G.; Sabatine, M.S.; Cheng, S.; Valcour, A.; Swales, H.; Taylor, B.; Carney, E.; et al. Vitamin D Therapy in Individuals with Prehypertension or Hypertension. Circulation 2015, 131, 254–262. [Google Scholar] [CrossRef]
  161. Ford, J.A.; MacLennan, G.S.; Avenell, A.; Bolland, M.; Grey, A.; Witham, M. RECORD Trial Group Cardiovascular disease and vitamin D supplementation: Trial analysis, systematic review, and meta-analysis. Am. J. Clin. Nutr. 2014, 100, 746–755. [Google Scholar] [CrossRef]
  162. Witte, K.K.; Byrom, R.; Gierula, J.; Paton, M.F.; Jamil, H.A.; Lowry, J.E.; Gillott, R.G.; Barnes, S.A.; Chumun, H.; Kearney, L.C.; et al. Effects of Vitamin D on Cardiac Function in Patients with Chronic HF: The VINDICATE Study. J. Am. Coll. Cardiol. 2016, 67, 2593–2603. [Google Scholar] [CrossRef]
  163. Zittermann, A.; Ernst, J.B.; Prokop, S.; Fuchs, U.; Dreier, J.; Kuhn, J.; Knabbe, C.; Birschmann, I.; Schulz, U.; Berthold, H.K.; et al. Effect of vitamin D on all-cause mortality in heart failure (EVITA): A 3-year randomized clinical trial with 4000 IU vitamin D daily. Eur. Heart J. 2017, 38, 2279–2286. [Google Scholar] [CrossRef]
  164. Scragg, R.; Stewart, A.W.; Waayer, D.; Lawes, C.M.M.; Toop, L.; Sluyter, J.; Murphy, J.; Khaw, K.-T.; Camargo, C.A. Effect of Monthly High-Dose Vitamin D Supplementation on Cardiovascular Disease in the Vitamin D Assessment Study. JAMA Cardiol. 2017, 2, 608. [Google Scholar] [CrossRef]
  165. Manson, J.E.; Cook, N.R.; Lee, I.-M.; Christen, W.; Bassuk, S.S.; Mora, S.; Gibson, H.; Gordon, D.; Copeland, T.; D’Agostino, D.; et al. Vitamin D Supplements and Prevention of Cancer and Cardiovascular Disease. N. Engl. J. Med. 2019, 380, 33–44. [Google Scholar] [CrossRef]
  166. Bischoff-Ferrari, H.A.; Vellas, B.; Rizzoli, R.; Kressig, R.W.; da Silva, J.A.P.; Blauth, M.; Felson, D.T.; McCloskey, E.V.; Watzl, B.; Hofbauer, L.C.; et al. Effect of Vitamin D Supplementation, Omega-3 Fatty Acid Supplementation, or a Strength-Training Exercise Program on Clinical Outcomes in Older Adults. JAMA 2020, 324, 1855. [Google Scholar] [CrossRef]
  167. Bolland, M.J.; Grey, A.; Gamble, G.D.; Reid, I.R. The effect of vitamin D supplementation on skeletal, vascular, or cancer outcomes: A trial sequential meta-analysis. lancet. Diabetes Endocrinol. 2014, 2, 307–320. [Google Scholar] [CrossRef]
  168. Barbarawi, M.; Kheiri, B.; Zayed, Y.; Barbarawi, O.; Dhillon, H.; Swaid, B.; Yelangi, A.; Sundus, S.; Bachuwa, G.; Alkotob, M.L.; et al. Vitamin D Supplementation and Cardiovascular Disease Risks in More Than 83 000 Individuals in 21 Randomized Clinical Trials: A Meta-analysis. JAMA Cardiol. 2019, 4, 765–776. [Google Scholar] [CrossRef]
  169. 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]
  170. Institute of Medicine (IOM). Dietary Reference Intakes for Calcium and Vitamin D; The National Academic Press: Washington, DC, USA, 2011.
  171. Kennel, K.A.; Drake, M.T.; Hurley, D.L. Vitamin D Deficiency in Adults: When to Test and How to Treat. Mayo Clin. Proc. 2010, 85, 752–758. [Google Scholar] [CrossRef]
Figure 1. Vitamin D synthesis pathway. The figure depicts the synthesis of vitamin D and its metabolites derived by ergosterol and cholesterol. CYP24A1 = cytochrome P450 family 24 subfamily A member 1, also named 25(OH)-D 24-hydroxylase; CYP27A1 = cytochrome P450 family 27 subfamily A member 1, also named sterol 27-hydroxylase; CYP27B1 = cytochrome P450 family 27 subfamily B member 1, also named 25(OH)-D 1α-hydroxylase; CYP2R1 = cytochrome P450 family 2 subfamily R member 1, also named vitamin D 25-hydroxylase; DBP = vitamin D binding protein; UVB = ultraviolet B; VDR = vitamin D receptor.
Figure 1. Vitamin D synthesis pathway. The figure depicts the synthesis of vitamin D and its metabolites derived by ergosterol and cholesterol. CYP24A1 = cytochrome P450 family 24 subfamily A member 1, also named 25(OH)-D 24-hydroxylase; CYP27A1 = cytochrome P450 family 27 subfamily A member 1, also named sterol 27-hydroxylase; CYP27B1 = cytochrome P450 family 27 subfamily B member 1, also named 25(OH)-D 1α-hydroxylase; CYP2R1 = cytochrome P450 family 2 subfamily R member 1, also named vitamin D 25-hydroxylase; DBP = vitamin D binding protein; UVB = ultraviolet B; VDR = vitamin D receptor.
Biomedicines 12 00768 g001
Figure 2. Hormonal regulation of vitamin D. The figure shows the hormonal regulation of the final step of vitamin D synthesis and the interplay at renal tubular cells (blue cells) of different signals. Green arrows indicate positive modulation; red arrows indicate inhibition. CYP24A1 = cytochrome P450 family 24 subfamily A member 1, also named 25(OH)-D 24-hydroxylase; CYP27B1 = cytochrome P450 family 27 subfamily B member 1, also named 25(OH)-D 1α-hydroxylase; FGF23 = fibroblast growth factor 23; FGFR1c = fibroblast growth factor receptor 1c; PTH = parathyroid hormone; PTHR1 = parathyroid hormone 1 receptor. Created with BioRender.com.
Figure 2. Hormonal regulation of vitamin D. The figure shows the hormonal regulation of the final step of vitamin D synthesis and the interplay at renal tubular cells (blue cells) of different signals. Green arrows indicate positive modulation; red arrows indicate inhibition. CYP24A1 = cytochrome P450 family 24 subfamily A member 1, also named 25(OH)-D 24-hydroxylase; CYP27B1 = cytochrome P450 family 27 subfamily B member 1, also named 25(OH)-D 1α-hydroxylase; FGF23 = fibroblast growth factor 23; FGFR1c = fibroblast growth factor receptor 1c; PTH = parathyroid hormone; PTHR1 = parathyroid hormone 1 receptor. Created with BioRender.com.
Biomedicines 12 00768 g002
Figure 3. Vitamin D targets in the cardiovascular field. The figure shows main targets and effect of vitamin D related to cardiovascular diseases. Green arrowheads indicate increase, red arrowheads indicate inhibition. ANP = atrial natriuretic peptide; Ca = calcium; CMs = cardiomyocytes; ECs = endothelial cells; IL-1 = interleukin-1; IL-6 = interleukin-6; LDL = low-density lipoprotein; MMP-2 = matrix metalloprotease-2; MMP-8 = matrix metalloprotease-8; NF-kB = nuclear factor-kB; NO = nitric oxide; P = phosphate; RAAS = renin angiotensin aldosterone system; TNF-α = tumor necrosis factor-α; VSMCs = vascular smooth muscle cells. Created with BioRender.com.
Figure 3. Vitamin D targets in the cardiovascular field. The figure shows main targets and effect of vitamin D related to cardiovascular diseases. Green arrowheads indicate increase, red arrowheads indicate inhibition. ANP = atrial natriuretic peptide; Ca = calcium; CMs = cardiomyocytes; ECs = endothelial cells; IL-1 = interleukin-1; IL-6 = interleukin-6; LDL = low-density lipoprotein; MMP-2 = matrix metalloprotease-2; MMP-8 = matrix metalloprotease-8; NF-kB = nuclear factor-kB; NO = nitric oxide; P = phosphate; RAAS = renin angiotensin aldosterone system; TNF-α = tumor necrosis factor-α; VSMCs = vascular smooth muscle cells. Created with BioRender.com.
Biomedicines 12 00768 g003
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

Nardin, M.; Verdoia, M.; Nardin, S.; Cao, D.; Chiarito, M.; Kedhi, E.; Galasso, G.; Condorelli, G.; De Luca, G. Vitamin D and Cardiovascular Diseases: From Physiology to Pathophysiology and Outcomes. Biomedicines 2024, 12, 768. https://doi.org/10.3390/biomedicines12040768

AMA Style

Nardin M, Verdoia M, Nardin S, Cao D, Chiarito M, Kedhi E, Galasso G, Condorelli G, De Luca G. Vitamin D and Cardiovascular Diseases: From Physiology to Pathophysiology and Outcomes. Biomedicines. 2024; 12(4):768. https://doi.org/10.3390/biomedicines12040768

Chicago/Turabian Style

Nardin, Matteo, Monica Verdoia, Simone Nardin, Davide Cao, Mauro Chiarito, Elvin Kedhi, Gennaro Galasso, Gianluigi Condorelli, and Giuseppe De Luca. 2024. "Vitamin D and Cardiovascular Diseases: From Physiology to Pathophysiology and Outcomes" Biomedicines 12, no. 4: 768. https://doi.org/10.3390/biomedicines12040768

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