Next Article in Journal
Protein-Balanced Dietary Habits Benefit Cognitive Function in Japanese Older Adults
Next Article in Special Issue
Effect of Vitamin D Supplementation on Depression in Adults: A Systematic Review of Randomized Controlled Trials (RCTs)
Previous Article in Journal
Genomic or Non-Genomic? A Question about the Pleiotropic Roles of Vitamin D in Inflammatory-Based Diseases
Previous Article in Special Issue
Protection of Vitamin C on Oxidative Damage Caused by Long-Term Excess Iodine Exposure in Wistar Rats
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Effects of Vitamin D on Cardiovascular Risk and Oxidative Stress

1
National Institute of Cardiology, Brazilian Ministry of Health, Rio de Janeiro 21040-900, Brazil
2
Nutrindo Ideais Performance and Nutrition Research Center, Rio de Janeiro 22640-100, Brazil
3
Extreme Sports Nutrition Institute—INEE, Belo Horizonte 31270-901, Brazil
*
Author to whom correspondence should be addressed.
G.R. and B.S.-S. contributed equally to the article and are co-first authors.
Nutrients 2023, 15(3), 769; https://doi.org/10.3390/nu15030769
Submission received: 20 December 2022 / Revised: 30 January 2023 / Accepted: 31 January 2023 / Published: 2 February 2023
(This article belongs to the Special Issue Vitamins and Human Health)

Abstract

:
Introduction: Vitamin D has been primarily studied as an important factor influencing bone and calcium metabolism. Metabolites of vitamin D are essential for whole-body calcium homeostasis, maintaining serum calcium levels within a narrow range by regulating this process in the bones and gut. Nevertheless, its deficiency is also related to increased risk of type 2 diabetes mellitus (T2DM), metabolic syndrome (MS), and cardiovascular disease (CVD)—with increased visceral adipose tissue and body mass index (BMI), as well as the frequently associated hypercholesterolemia. It has been reported that vitamin D levels are inversely related to cardiovascular (CV) risk in men and women. However, the effects of vitamin D on distinct outcomes in women and the dose of supplementation needed to improve clinical endpoints have not been established. 25-Hydroxyvitamin D [25(OH)D] reduces systemic inflammatory mediators in CVD and favors the release of anti-inflammatory cytokines from the immune system. In addition, 25(OH)D can be primarily converted into calcitriol (1,25-dihydroxycholecalciferol [1,25(OH)2D]) in the kidneys through the action of the 1-α-hydroxylase enzyme. Calcitriol, through the downregulation mechanism of renin expression, renin–angiotensin–aldosterone system (RAAS) activity, and its interaction with the vitamin D receptor, can bring CV benefits. The calcitriol form also lowers parathyroid hormone (PTH) levels by indirectly causing a reduction in aldosterone and mineralocorticoid synthesis. Elevated plasma aldosterone is related to endothelial dysfunction and CVD in hypovitaminosis D status. Conclusion: Vitamin D supplementation may benefit certain risk groups, as it improves metabolic variables, reducing oxidative stress and CV outcomes. More studies are needed to define interventions with vitamin D in men and women.

1. Introduction

Recent studies have shown that vitamin D deficiency is a global health problem [1,2,3]. The minimum laboratory value of 25-hydroxyvitamin D [25(OH)D], 20 ng/mL, is not reached by 65% of the world’s elderly population, and deficiency is present even in younger individuals [4]. Vitamin D is one of the main factors influencing bone and calcium metabolism. Its deficiency is also related to a higher risk of type 1 diabetes, type 2 diabetes mellitus (T2DM), metabolic syndrome (MS), coronavirus disease 2019 (COVID-19), and cardiovascular disease (CVD), including arterial hypertension (AH) [1,5,6,7,8,9].
This review article assesses the potential relevance of vitamin D concentrations and their relationships with oxidative stress and cardiovascular (CV) risk. Some evidence suggests that vitamin D levels are inversely related to blood pressure and the risk of AH, based on studies in Western populations [9,10,11]. Animal studies provide strong support for the function of 1,25-dihydroxycholecalciferol [1,25(OH)2D] through the downregulation of renin expression and renin–angiotensin–aldosterone system (RAAS) activity through its interaction with the vitamin D receptor [12]. Vitamin D’s function as a potent negative endocrine regulator of renin gene expression provides insights into the development of AH and how vitamin D deficiency could favor its development or progression [13]. The 25(OH)D is converted to 1,25(OH)2D in the kidneys through the action of the 1-α-hydroxylase enzyme. However, macrophages, endothelial cells, and smooth muscle cells are also capable of performing this conversion. This increase in the local production of active vitamin D acts as an autocrine and paracrine factor, which is critical for specific cell functions [14].
A growing body of evidence from animal and human studies illustrates that vitamin D decreases systemic inflammatory mediators in CVD and favors the release of anti-inflammatory cytokines from the immune system [15,16]. In diabetics, for example, 1,25(OH)2D has been shown to suppress the uptake of cholesterol into the endothelium by macrophages and reverse the increased cholesterol deposition through the downregulation of endoplasmic reticulum activity. Furthermore, this suppression of the endoplasmic reticulum activity shifts M2 macrophages to the M1 phenotype, suggesting that vitamin D’s regulation of endoplasmic reticulum activity may be a potential therapy for patients with atherosclerosis. Macrophages recruited to the subendothelial space respond to environmental signals that dictate their differentiation into different phenotypes. In atherosclerotic plaques, M1 macrophages express membrane receptors that facilitate the egress of immune cells from the atherosclerotic plaque. In contrast, M2 macrophages, induced by interleukins (e.g., IL-4, IL-10), are known to attenuate excessive inflammation, facilitating collagen production and fibrosis [17,18].
In preclinical studies, for example, the absence of vitamin D receptor signaling increased blood pressure by increasing renin release and accelerating atherogenesis, possibly through the local activation of macrophages and the RAAS [18].

2. Material and Methods

The presented scientific literature was reviewed and the studies were downloaded from the PubMed (https://pubmed.ncbi.nlm.nih.gov/ (accessed on 1 December 2022)), ScienceDirect (https://sciencedirect.com/ (accessed on 1 December 2022)), and SciELO (https://scielo.com.br/ (accessed on 1 December 2022)) databases. Combinations of several search terms—such as “vitamin D”, “cholecalciferol”, “calcitriol”, “1,25 dihydroxycholecalciferol”, “25 hydroxycholecalciferol”, “CVD”, “cardiovascular”, “antioxidant”, “anti-inflammatory”, “oxidative stress”, “redox”, “hypertension”, “hypercholesterolemic”, and “antihypertensive”—were applied. After the search, the studies were classified according to the health-specific parameters of the text.

3. Results

3.1. Vitamin D and Its Physiology

Vitamin D is a generic name used to designate a group of fat-soluble compounds of endogenous or exogenous origin that have the physiological characteristics of a hormone [16]. The endogenous form is produced in the epidermis, by the action of ultraviolet B rays, from the activation of 7-dehydrocholesterol (provitamin D3) with the formation of vitamin D3, which undergoes hydroxylation primarily in the liver and in the proximal convoluted tubules of the kidneys, resulting in 1,25(OH)2D or calcitriol, which is the most active form of vitamin D [16].
In humans, exposure to the Sun is responsible for about 90–95% of the supply of vitamin D. The exogenous sources—i.e., food or ergocalciferol—are unimportant, as significant amounts of vitamin D are found only in oily fish and mushrooms cultivated in the Sun. The effectiveness of endogenous production of vitamin D depends on environmental factors related to latitude, season, and the time of day when Sun exposure occurs. Genetic factors, body composition, physical activity, topical use of sunscreens, and skin pigmentation are also important [19,20].
According to consensus, obesity is strongly related to serum levels of 25(OH)D, with a negative correlation between vitamin D levels and body mass index (BMI) [21] that could be explained both by the shorter time of exposure to the Sun—linked to the more sedentary habits of the obese population—and by the reduction in vitamin D’s bioavailability, related to the increase in the storage of vitamin D in the adipose tissue [22]. It has also been proposed that hypovitaminosis D could promote a physiological elevation of parathyroid hormone (PTH) levels by increasing the intracellular calcium concentration in adipocytes which, in turn, would stimulate lipogenesis and weight gain [23].
To clarify this issue, Vimaleswaran and colleagues recently performed a bidirectional Mendelian randomization study that analyzed 21 cohorts with a total of 42,024 patients, demonstrating that genetically determined 25(OH)D levels were not significantly correlated with BMI [24]. The authors concluded that obesity should be seen as an important causal risk factor that accounts for approximately one-third of cases of vitamin D deficiency. Thus, the obesity “epidemic” may be contributing to the high prevalence of insufficiency (25(OH)D < 30 ng/mL) and deficiency (25(OH)D < 20 ng/mL) of vitamin D according to criteria established by the Endocrine Society, currently observed in almost half of the world’s population [25]. The recommendation for vitamin D supplementation includes the following criteria: 400–1000 IU/day, 600–1000 IU/day, and 1500–2000 IU/day for infants up to 1 year of age, children up to 18 years of age, and adults, respectively. The Endocrine Society also states that obese adults require 2–3 times more vitamin D than eutrophic adults [26].
From a physiological point of view, the active form of vitamin D increases intestinal calcium absorption and extracellular calcium concentration, indirectly suppressing PTH secretion, in addition to having a direct inhibitory effect on gene transcription in the parathyroid glands. In vitamin D deficiency, there is an elevation of PTH and, consequently, an increase in renal tubular reabsorption of calcium and its mobilization from the bone through osteoclastic stimulation [27].
The main functions of vitamin D are related to calcium/phosphorus homeostasis and the promotion of bone mineralization. However, considering that the vitamin D receptor coding gene transcription can regulate about 3% of the human genome, and that vitamin D receptors are present in several tissues (e.g., parathyroid cells, renal tubules, pancreatic β cells, immune system cells, neural cells, cardiac musculature, the muscular layer of vessels and the endothelium), many studies have suggested that vitamin D deficiency may contribute to the pathogenesis of conditions such as autoimmune diseases, cancer, COVID-19, glucose and lipid alterations, AH, MS, endothelial dysfunction, and CVD [2,7,8,9,28,29,30,31,32,33]
Evidence demonstrates that a low concentration of 25(OH)D is considered to be a risk factor for chronic diseases and is also related to all-cause mortality [34]. Thus, it is expected to increase morbidity and mortality for diseases associated with vitamin D deficiency in the coming years [35]. Evidence on the non-skeletal health benefits of vitamin D has gained momentum in recent years, although guidelines still only support its benefits for skeletal outcomes [34].
Vitamin D deficiency has been associated with several conditions and diseases, including immunological pathologies, cancer, and COVID-19. In fact, recent research has supported the hypothesis that vitamin D deficiency is related to the incidence, severity, and mortality attributed to COVID-19 [7,8]. It has been demonstrated that, in men, the administration of 25(OH)D, without neglecting the hypercalcemic risk, can be considered a suitable indication for treating patients with COVID-19 [7]. Furthermore, studies have concluded that individuals with a higher risk of severe COVID-19 correspond to those with a higher risk of severe vitamin D deficiency, including the elderly and obese [8]. However, although a role for vitamin D has been proposed in counteracting COVID-19 through potential modulation of immune dysfunction and cytokine storm, to date, there is still a lack of evidence in the literature in terms of differences in serum levels in large cohorts, according to disease state [9].
In addition, one of the big questions about vitamin D supplementation is whether there are a variety of recommended vitamin D thresholds that are used to study their health effects. An insufficient level of vitamin D—considered to be <50 nmol/L—is most often used to describe hypovitaminosis D in recent studies. Thus, it seems to be difficult to interpret some studies that consider normal values of vitamin D to be between 25 and 49 nmol/L [36].
In chronic diseases such as T2DM, vitamin D supplementation has shown a 10% risk reduction in overweight individuals with pre-T2DM for progression to T2DM. However, this effect was smaller than that produced by the use of oral hypoglycemic agents or lifestyle modifications [37,38]. Additionally, with regard to cancer, there are strong preclinical data linking vitamin D to cell-cycle control. Furthermore, many observational studies have associated low vitamin D status with an increased risk of cancer or poor prognosis. There appears to be a benefit of vitamin D supplementation on cancer mortality, especially when the follow-up is longer than 4 years [37,38,39].
The relationship between vitamin D and stroke has been investigated in some studies, and it has been proposed that vitamin D could be a prognostic biomarker for functional outcomes in stroke patients. However, the evidence is still inadequate to support the effectiveness of vitamin D supplementation in stroke and post-stroke recovery [40].
Vitamin D appears to have an important immunomodulatory effect due to its influence on all cells of the immune system through the production of cytokines. Patients with vitamin D deficiency, after receiving vitamin D supplementation, have a reduced risk of upper respiratory infections. Another documented benefit was in patients with asthma or respiratory disease, where vitamin D supplementation reduced the risk of acute upper respiratory infections and improved expiratory lung function. With regard to autoimmune diseases, there may be a worsening in cases of vitamin D deficiency. This is due to the adaptive immune system being negatively regulated by calcitriol and, therefore, vitamin D deficiency may predispose individuals to autoimmune diseases [41].
Regarding bone metabolism, vitamin D status is related to inadequate bone health—especially in high-risk groups, such as cancer patients. A very low prevalence of patients with adequate bone health and normal vitamin D status was observed in a cohort of women with breast cancer. Thus, bone health and vitamin D status must be appropriately assessed and managed to reduce the risk of fragility fractures in patients with cancer and other chronic diseases [39].

3.2. Vitamin D, Oxidative Stress, and Inflammation

Oxidative stress has confirmed its importance in cellular damage through the action of reactive oxygen species (ROS). However, there is a balance between oxidant and antioxidant activity under normal conditions. The antioxidant defense system includes glutathione peroxidase, superoxide dismutase, and various vitamins, including vitamin D, minerals, and other compounds. Vitamin D, in particular, is known to have the capacity to reduce oxidative stress through the positive regulation of cellular glutathione and superoxide dismutase. Several meta-analyses and clinical studies with different groups have demonstrated the important role of vitamin D in the management of oxidative stress (Table 1).
Sepidarkish and colleagues presented a meta-analysis of 13 clinical trials, finding that vitamin D supplementation increased levels of serum total antioxidant capacity (TAC) (SMD: 0.54 mmol/L, 95%CI: 0.29 to 0.79; I2 = 65.4%, p = 0.001) and total serum glutathione (GSH) (SMD: 0.33; 95% CI: 0.11 to 0.54, p = 0.003; I2 = 61.2%, p = 0.001). Malondialdehyde (MDA) decreased significantly (SMD: −0.40 mmol/L, 95% CI: −0.60 to −0.21, p < 0.001), while nitric oxide (NO) was not significantly different (0.17, 95% CI: −0.10 to 0.45, I2 = 71.1%, p = 0.21). [46].
A comprehensive meta-analysis listed 23 studies in different groups of individuals with different clinical health conditions, genders, and ages, finding improvements in MDA levels (ES = −0.37; 95% CI: −0.48, −0.25, p < 0.001) and in the inflammatory markers high-sensitivity C-reactive protein (hs-CRP) (ES = −0.42; 95% CI: −0.55, −0.29, p < 0.001) and tumor necrosis factor (TNF)-α (ES = −0.27; 95% CI: −0.42, −0.12; p < 0.001) at the time of vitamin D supplementation [48].
In specific groups, such as women with polycystic ovary syndrome (PCOS), it was found that vitamin D supplementation led to an increase in TAC, leading to reductions in the MDA levels (−1.64, 95% CI −2.26 to −1.02, p < 0.001) and in the oxidative stress and inflammatory marker hs-CRP (−1.03; 95% CI, −1.58, −0.49; p < 0.001); however, there were no changes in NO or GSH [44].
In a systematic review including 33 randomized clinical trials of diabetic patients to analyze the relationship of vitamin D supplementation with some markers, there were decreases in hs-CRP (WMD 0.27; 95% CI, −0.35, −0.20; p < 0.001) and MDA (WMD − 043, 95% CI −0.62, −0.25, p < 0.001), as with the group of women with PCOS. Along the way, there were still significant increases in the release markers of NO (WMD 4.33, 95% CI 0.96, 7.70), GSH (WMD 82.59, 95% CI 44.37, 120.81, p < 0.001), and TAC (WMD 57.34, 95% CI 33.48, 81.20, p < 0.001) [45].
A randomized controlled study corroborating these findings, also including T2DM patients, performed some interventions including the supplementation of at least 14,000 IU per week (deficient group: initial supplementation was 50,000 IU per week of vitamin D for three months, and then supplementation was reduced to 14,000 IU per week for another three months), finding a decrease in the concentration of ROS at a follow-up period of 6 months and a decrease in the inflammatory marker CRP, although this was not statistically significant [47].
In another study analyzing younger age groups, the authors found similar results. In a Chinese study involving more than 1400 children aged 7 to 11 years, comparing deficiency and sufficiency groups, there was an inverse association of 25(OH)D levels with hs-CRP (1.21 ± 0.13 vs.1.04 ± 0.14), interleukin-6 (IL-6) (3.83 ± 0.33 vs. 3.58 ± 0.34), MDA (2.87 ± 0.21 vs. 2.83 ± 0.27), and superoxide dismutase (SOD) (95.38 ± 12.22 vs. 127.62 ± 15.98 U/mL, p < 0.001) [43]. Corroborating these findings, an observational study carried out with 66 obese children found a correlation between vitamin D deficiency and increased levels of oxidative stress and inflammation markers. After partial correlation analysis, it was found that there was an inverse relationship of vitamin D levels with 3-nitrotyrosine (r = −0.424, p = 0.001), soluble vascular cell adhesion molecule-1 (sVCAM-1) (r = −0.272, p = 0.032), and the inflammation marker IL-6 (18.0 [12.2–25.7] vs. 13.7 [10.7–19.3] pg/mL, p < 0.036) [42].
As seen in the studies cited above, chronic low-grade inflammation that is typically characterized by increased levels of C-reactive protein (CRP), interleukin-6 (IL-6), and tumor necrosis factor-α (TNF-α) is associated with increased risk of type 2 diabetes mellitus (T2DM) and cardiovascular diseases (CVD). A series of in vitro and in vivo studies have demonstrated that vitamin D supplementation decreases the levels of inflammatory cytokines and increases the concentrations of anti-inflammatory markers [17,42,43,44,45,46,47,48]. Vitamin D and its metabolite can inhibit nuclear transcription factor κB activity by increasing IκB expression and reducing IκB-α phosphorylation. Therefore, vitamin D has the potential to inhibit the production of pro-inflammatory factors (42,44). Thus, vitamin D can decrease the generation of free radicals and inflammatory cytokines, mainly by deactivating NF-κB-dependent pathways, showing its favorable effects on oxidative stress reduction [46].
In summary, vitamin D should be considered as an adjuvant therapy for treating inflammation and increased oxidative stress diseases. More research is still required to determine the dosages and duration of treatment.

3.3. Vitamin D and Cardiovascular Risk

Several studies have demonstrated an essential link between vitamin D and CV risk (Table 2). Observational and prospective studies show that the prevalence of hypovitaminosis D is significantly higher in adults with T2DM when matched by sex and age to non-diabetics. These T2DM patients with hypovitaminosis D have significant thickening of the common carotid intima compared to those presenting normal serum 25(OH)D levels [49]. However, when 108 healthy postmenopausal women (mean age: 55 years) received a daily supplement containing minerals and vitamin D, no statistical significance was seen for intima–media thickness, compliance coefficient, or distensibility coefficient when compared to a placebo [50]. Oh and colleagues, in an in vitro study, found that 1,25(OH)2D suppressed macrophage foam cell formation in a cultured positive medium by reducing acetylated or oxidized LDL uptake. The study was designed using macrophages from 62 diabetic, hypertensive, and vitamin-D-deficient (<80 nmol/L) women (mean 55 years), and almost 50% less cholesteryl ester formation was seen when vitamin D was present in the medium [17].
The mechanisms by which the reduction in vitamin D concentrations could trigger T2DM are not fully understood. There is evidence that vitamin D directly stimulates insulin secretion by pancreatic β cells, and some other indirect actions have been proposed [62]. Additionally, considering the relevance of 25(OH)D in the regulation of the immune system, as suggested by the higher prevalence of hypovitaminosis D in patients with autoimmune diseases (e.g., Crohn’s disease, T2DM, rheumatoid arthritis, lupus, and multiple sclerosis) and some types of cancer (e.g., colon, prostate, breast, and pancreas), the possibility was also raised that exacerbation of inflammatory responses, which accompanies hypovitaminosis D states, may contribute to the worsening of insulin resistance [5,49,50,62,63].
Although observational studies indicate a positive association between vitamin D deficiency, reduction in HDL cholesterol/apolipoprotein A levels, and elevation of total cholesterol/triglycerides [64], recent clinical research has not observed significant benefits in plasma lipid concentrations with vitamin D supplementation [65]. Therefore, the consequences of hypovitaminosis D on the lipoprotein profile have yet to be established. Myrup and colleagues accompanied 74 postmenopausal healthy Danish women (>70 years) for 18 months. The protocol of supplementation with 0.5 μg of 1,25(OH)2D/day for 1 year presented no statistical significance for any of the parameters analyzed (i.e., total cholesterol (TC); high-density lipoprotein cholesterol (HDL-c); blood pressure at 0, 3, 6, 9, 12, and 18 months) [51]. On the other hand, a study of 463 postmenopausal women (45–75 years; mean age 58 years) showed that women with low 25(OH)D levels (<30 ng/mL) had higher levels of TC (p = 0.031), triglycerides (OR 1.55, 95%CI = 1.13–2.35), and HDL-c (OR 1.60, 95%CI = 1.19–2.40) [6].
Vitamin D supplementation has been found to improve several markers of cardiovascular health, including enhanced lipid profiles [57,59,66]. Thus, 25(OH)D deficiency is related to a significant worsening in TC, LDL, and triglyceride levels, as observed in a study reporting alterations of +9.4%, +13.5%, and +26.4%, respectively [57]. Vitamin D sufficiency has been shown to reduce TC, LDL, and triglycerides, with mean differences of −0.17, −0.19, and −0.12, respectively [59].
The link between AH and 25(OH)D deficiency is supported by prospective studies and some meta-analyses of observational studies [1,16]. Data from the National Health and Nutrition Examination Surveys (NHANES) from 2001 to 2006 showed an inverse association between serum 25(OH)D and systolic blood pressure after adjusting for age, gender, ethnicity, and BMI. More than 5000 adolescents aged 12 to 19 years were included in this study. Systolic blood pressure was significantly higher in the low-serum-25(OH)D group (serum 25(OH)D3 < 48.1 nmol/L) than in the medium (serum 25(OH)D 48.1 to <66.2 nmol/L) and high groups (serum 25(OH)D > 66.2 nmol/L) (109.8 ± 0.5 vs. 108.2 ± 0.4 and 108.4 ± 0.4 mmHg, respectively; p = 0.01) [56]. Similarly, in a study of over 1400 adolescents aged 13 to 15 years, it was found that 25(OH)D deficiency (serum 25(OH)D3 < 50 nmol/L) was associated with elevated levels of diastolic blood pressure. In this study, diastolic blood pressure was higher in the 25(OH)D-deficient group compared with the 25(OH)D3-sufficient group (p = 0.04) [61]. Other studies observed similar results, where increasing serum 25(OH)D levels resulted in significant decreases in systolic and diastolic blood pressure [58,67]. It was observed that for every 10 mmol/L increase in serum 25(OH)D there was an associated decrease in diastolic blood pressure (p = 0.02), independent of the BMI [58].
Studies demonstrate an inverse correlation between serum 25(OH)D levels and blood pressure—most evident when analyzing systolic blood pressure (SBP) in women over 50 years of age [68]—higher blood pressure levels in individuals with a history of less exposure to ultraviolet rays [69,70], and lower risk of AH in middle-aged women who ingest higher amounts of calcium/vitamin D through the consumption of milk and dairy products [71]. In 2001, Pfeifer and colleagues developed a study aiming to compare the effects of calcium (1.200 mg/day) versus calcium plus vitamin D supplementation (1.200 mg/day and 800 IU/day, respectively) on the SBP levels of postmenopausal women (n = 148; mean age = 74 years). Supplementation of vitamin D with calcium decreased SBP by 9.3% (p = 0.02) and heart rate by 5.4% (p = 0.02) when compared to the supplementation with calcium alone. Furthermore, 60 subjects (81%) in the calcium plus vitamin D group showed a decrease of 5 mmHg or more in SBP, compared with 35 (47%) subjects in the calcium group (p = 0.04) [52].
A large meta-analysis of prospective studies included more than 280,000 participants [13], including 55,816 patients with AH. The analysis included men and women with 25(OH)D levels measured at baseline. The participants with the highest levels of 25(OH)D were found to have a 30% lower risk of AH. Another study evaluated the relationship between 25(OH)D, PTH levels, and AH in the elderly [72]. In that study, vitamin D supplementation caused a reduction in PTH levels, which are a potentially modifiable determinant of blood pressure. Similar observations were described in a study that investigated 25(OH)D levels in patients with metabolic syndrome—low circulating 25(OH)D was associated with high PTH levels [73]. In that study, with more than 1000 adult participants, it was suggested that hyperparathyroidism might contribute to the worsening of metabolic syndrome and AH.
Over seven years, a prospective study found no significant differences in the adjusted mean changes in systolic or diastolic blood pressure by quartile of 25(OH)D. Incident AH was slightly and statistically significantly lower in the third quartile of 25(OH)D serum levels (48–<65 nmol/L) compared with the lowest quartile in 4863 healthy postmenopausal women (mean age = 66 years) [54].
In a prospective case–control study with 1484 middle-aged women, the authors observed a relative AH risk of 1.47 (95% CI: 1.10–1.97) for women with vitamin D deficiency compared to those with adequate serum levels [10]. The pathophysiological mechanisms suggested for this association include the elevation of serum PTH levels—directly related to the reduction in 25(OH)D—and the deleterious effects of hypovitaminosis D on activation of the RAAS and endothelial function [74].
The effects of PTH on blood pressure and CVD, as suggested by clinical observations [75] and by the induction of AH with the infusion of PTH, are probably mediated by the action of aldosterone. Indeed, a positive correlation between PTH and aldosterone levels was described in 2009 by Brunaud and colleagues, where several clinical and experimental studies showed that PTH could directly or indirectly stimulate (via the RAAS) the synthesis of mineralocorticoids [54,74,75]. It is essential to highlight that, in a large prospective study published in 2014, after the adjustment for potential confounders, the elevation of PTH—rather than the reduction in 25(OH)D concentrations—was associated with the risk of AH [27].
In the last decade, experimental studies with animals have shown that calcitriol is a potent suppressor of gene transcription that determines renin synthesis. Human studies have demonstrated that high calcitriol levels are associated with the suppression of plasma renin activity [76]. In fact, mice lacking vitamin D receptors have excessive plasma renin activity associated with AH [12]. These findings are of practical importance, since vitamin D replacement and the development of vitamin D analogs with negligible effects on calcium metabolism could suppress the RAAS, benefiting some patients with AH. However, the drug intervention studies currently available are insufficient to corroborate this benefit.
It is possible that elevations in plasma aldosterone also play a role in the endothelial dysfunction that has already been described in hypovitaminosis D states [53,55,60,77,78,79,80]. This hypothesis was supported by a large population analysis that demonstrated a positive correlation between aldosterone/renin levels and changes in endothelial function. Despite this, these results could be limited in middle-aged individuals due to the methods of assessing flow-mediated dilation of the brachialis. In this study, the vitamin D deficiency group (25(OH)D level of <20 ng/mL) had significantly higher prevalence of severe coronary artery disease (CAD) (53% vs. 38% p = 0.03), diffuse CAD (56% vs. 34% p = 0.03), and impaired endothelial-dependent vasodilation (FMD) (values < 4.5%; 50.6% vs. 7%; p < 0.002), as well as significantly lower flow-mediated dilation of the brachial artery (4.57% vs. 10.68%: p < 0.001) [80].
Recent in vitro studies also demonstrate that the benefits of vitamin D on endothelial function may be related to increased expression of the enzyme nitric oxide synthetase [81,82] and endothelial growth factor [83,84], inhibition of adhesion molecules, macrophage migration, and muscle cell proliferation [84,85], reduction in oxidative stress, and production of pro-inflammatory cytokines [32,79].
The effect of vitamin D replacement on endothelial function has been poorly investigated. Although some studies have not observed significant impacts of vitamin D supplementation on endothelial function, adhesion molecule production, or hs-CRP [32,85,86], the normalization of the production of ROS and the overexpression of the angiotensin II type 1 receptor in rats have already been documented [86]. Some controlled clinical studies have shown that vitamin D replacement can improve the parameters of vascular function [60]. Tarcin and colleagues (2009) presented significantly lower FMD in 25(OH)D-deficient subjects (25(OH)D < 25 nmol/l) than in controls (7.0 ± 3.2% vs. 11.2 ± 5.2%; p = 0.001), improved levels after replacement therapy (10.4 ± 3.3%; p = 0.002), and significantly lower post-treatment values of thiobarbituric-acid-reactive substances (TBARS) (4.7 ± 1.7 to 2.9 ± 0.7 ng/mg MDA; p = 0.0001) [60]. Therefore, even when analyzing this early sign of atherosclerosis and vascular injury, current evidence does not allow for estimating the real benefits of vitamin D replacement in the prevention or treatment of CVD, and further studies are needed.

4. Discussion

Oxidative stress is a transient or permanent disturbance in the state of oxidative equilibrium that generates regulatory changes within the cell, depending on the specific target and concentrations of ROS. Under regulatory conditions, ROS concentrations fluctuate in a controlled manner and are modulated by enzymatic and non-enzymatic antioxidant systems. Thus, there is a balance between oxidant and antioxidant activity. ROS levels increase if this homeostatic state fails, generating inflammation and dysfunctional physiological effects [46,48].
Oxidative imbalance has been associated with the progression of atherosclerosis and CV outcomes. Its increase impacts the development of several atherosclerotic risk factors, including vitamin D deficiency. Specific oxidative pathways involving pro-oxidant and antioxidant enzymes, such as nicotinamide adenine dinucleotide phosphate oxidase (NADPH), myeloperoxidase, SOD, and glutathione peroxidase, seem to play an essential role in the production of ROS. Vitamin D, in particular, is known to have the ability to reduce oxidative stress through the upregulation of cellular glutathione and SOD [44,46].
Observational and prospective studies demonstrate a correlation between hypovitaminosis D, increased levels of oxidative stress markers, and significant thickening of the common carotid intima—a common indicator of CV dysfunction. On the other hand, there is a lower risk of AH in individuals with normal serum levels of 25(OH)D who receive greater amounts of calcium/vitamin D through the consumption of milk and derivatives, for example. The pathophysiological mechanisms suggested for this association with the relative risk of developing AH include hypovitaminosis D’s deleterious effects on the RAAS’s activation and endothelial function [42,49,83].
The correlation between vitamin D deficiency and increased levels of oxidative stress markers is known. Therefore, vitamin D should be considered as an adjuvant therapy in the treatment of inflammatory diseases and/or cardiometabolic conditions. However, the effect of vitamin D replacement has still been scarcely investigated, and current evidence does not allow for estimating the actual benefits, dosages, and duration of treatment—especially for the prevention and/or treatment of CVD. More studies are also needed to investigate new strategies to modulate oxidative stress and to determine which lifestyle modifications should be implemented. When it comes to a nutritional approach, the Mediterranean diet and the Dietary Approach to Stop Hypertension are especially recommended by the American Heart Association as preventive or treatment approaches for CVD. A common feature of these dietary patterns is that they are rich in antioxidants. Therefore, adherence to healthy dietary patterns should always be investigated in patients with cardiometabolic disease [87,88,89].

5. Conclusions

Vitamin D supplementation could favor some risk groups, especially patients with chronic diseases, by reducing oxidative stress and endothelial damage. In fact, an increase in CVDs is observed with aging, and other conditions such as obesity, T2DM, AH, COVID-19, dyslipidemia, and smoking impair NO production, increasing RAAS activation and oxidative stress, which may contribute to endothelial dysfunction in these patients. Further studies analyzing the effects of vitamin D on endothelial function are of great interest, since the correction of hypovitaminosis D could minimize the damage to the endothelial function and, consequently, reduce the CV risk.

Author Contributions

Conceptualization, G.R. and B.S.-S.; methodology, B.S.-S.; data curation, G.R.; writing—original draft preparation, T.B., R.P. (Rayssa Petronio), D.A. and R.P. (Raphaela Paes); writing—review and editing, G.R. and B.S.-S. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Essential Nutrition Ind de Alimentos.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

List of Abbreviations

[25(OH)D]25-Hydroxyvitamin D
[1,25(OH)2D]1,25-Dihydroxyvitamin D
AHArterial hypertension
BMIBody mass index
CADCoronary artery disease
COVID-19Coronavirus disease 2019
CRPC-reactive protein
CVDCardiovascular disease
CVCardiovascular
DMDiabetes mellitus
FMDEndothelial-dependent vasodilation
GSHGlutathione
HAHypertension
HDL-cHigh-density lipoprotein cholesterol
hs-CRPHigh-sensitivity C-reactive protein
IL-4Interleukin-4
IL-6Interleukin-6
IL-10Interleukin-10
IRInsulin resistance
MDAMalondialdehyde
MSMetabolic syndrome
NADPHNicotinamide adenine dinucleotide phosphate oxidase
NHANESNational Health and Nutrition Examination Surveys
NONitric oxide
PTHParathyroid hormone
PCOSPolycystic ovary syndrome
RASRenin–angiotensin system
RAASRenin–angiotensin–aldosterone system
ROSReactive oxygen species
SBPSystolic blood pressure
SODSuperoxide dismutase
sVCAM-1Soluble vascular cell adhesion molecule-1
TACAntioxidant capacity levels
TBARSThiobarbituric-acid-reactive substances
TCTotal cholesterol
T2DMType 2 diabetes mellitus
TNFTumor necrosis factor

References

  1. Holick, M.F. Vitamin D: Important for prevention of osteoporosis, cardiovascular heart disease, type 1 diabetes, autoimmune diseases, and some cancers. South. Med. J. 2005, 98, 1024–1027. [Google Scholar] [CrossRef]
  2. Holick, M.F. Vitamin D deficiency. N. Engl. J. Med. 2007, 357, 266–281. [Google Scholar] [CrossRef]
  3. Holick, M.F.; Chen, T.C. Vitamin D deficiency: A worldwide problem with health consequences. Am. J. Clin. Nutr. 2008, 87, 1080s–1086s. [Google Scholar] [CrossRef]
  4. Ferrone, F.; Pepe, J.; Danese, V.; Fassino, V.; Cecchetti, V.; De Lucia, F.; Biamonte, F.; Colangelo, L.; Ferrazza, G.; Panzini, E. The relative influence of serum ionized calcium and 25-hydroxyvitamin D in regulating PTH secretion in healthy subjects. Bone 2019, 125, 6. [Google Scholar] [CrossRef]
  5. Pittas, A.G.; Lau, J.; Hu, F.B.; Dawson-Hughes, B. The role of vitamin D and calcium in type 2 diabetes. A systematic review and meta-analysis. J. Clin. Endocrinol. Metab. 2007, 92, 2017–2029. [Google Scholar] [CrossRef]
  6. Schmitt, E.B.; Nahas-Neto, J.; Bueloni-Dias, F.; Poloni, P.; Orsatti, C.L.; Nahas, E.A.P. Vitamin D deficiency is associated with metabolic syndrome in postmenopausal women. Maturitas 2018, 107, 97–102. [Google Scholar] [CrossRef]
  7. Gallelli, L.; Mannino, G.C.; Luciani, F.; de Sire, A.; Mancuso, E.; Gangemi, P.; Cosco, L.; Monea, G.; Averta, C.; Minchella, P.; et al. Vitamin D Serum Levels in Subjects Tested for SARS-CoV-2: What Are the Differences among Acute, Healed, and Negative COVID-19 Patients? A Multicenter Real-Practice Study. Nutrients 2021, 13, 3932. [Google Scholar] [CrossRef]
  8. Jin, J.-M.; Bai, P.; Bai, P.; He, W.; Wu, F.; Liu, X.; Han, D.; Liu, S.; Yang, J. Gender differences in patients with COVID-19: Focus on severity and mortality. Front. Public Health 2020, 8, 152. [Google Scholar] [CrossRef]
  9. Quesada-Gomez, J.M.; Entrenas-Castillo, M.; Bouillon, R. Vitamin D receptor stimulation to reduce acute respiratory distress syndrome (ARDS) in patients with coronavirus SARS-CoV-2 infections: Revised. J. Steroid Biochem. Mol. Biol. 2020, 202, 105719. [Google Scholar] [CrossRef]
  10. Forman, J.P.; Curhan, G.C.; Taylor, E.N. Plasma 25-hydroxyvitamin D levels and risk of incident hypertension among young women. Hypertension 2008, 52, 828–832. [Google Scholar] [CrossRef]
  11. Martins, D.; Wolf, M.; Pan, D.; Zadshir, A.; Tareen, N.; Thadhani, R.; Felsenfeld, A.; Levine, B.; Mehrotra, R.; Norris, K. Prevalence of cardiovascular risk factors and the serum levels of 25-hydroxyvitamin D in the United States: Data from the Third National Health and Nutrition Examination Survey. Arch. Intern. Med. 2007, 167, 1159–1165. [Google Scholar] [CrossRef]
  12. Li, Y.C.; Kong, J.; Wei, M.; Chen, Z.; Liu, S.; Cao, L. 1,25-dihydroxyvitamin D (3) is a negative endocrine regulator of the renin-angiotensin system. J. Clin. Invest. 2002, 110, 229–238. [Google Scholar] [CrossRef]
  13. 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]
  14. Burgaz, A.; Orsini, N.; Larsson, S.C.; Wolk, A. Blood 25-hydroxyvitamin D concentration and hypertension: A meta-analysis. J. Hypertens. 2011, 29, 636–645. [Google Scholar] [CrossRef] [PubMed]
  15. Weng, S.; Sprague, J.E.; Oh, J.; Riek, A.E.; Chin, K.; Garcia, M.; Bernal-Mizrachi, C. Vitamin D deficiency induces high blood pressure and accelerates atherosclerosis in mice. PLoS ONE 2013, 8, e54625. [Google Scholar] [CrossRef]
  16. Dusso, A.S.; Brown, A.J.; Slatopolsky, E. Vitamin D. Am. J. Physiol. Ren. Physiol. 2005, 289, F8–F28. [Google Scholar] [CrossRef]
  17. 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]
  18. Szeto, F.L.; Reardon, C.A.; Yoon, D.; Wang, Y.; Wong, K.E.; Chen, Y.; Kong, J.; Liu, S.Q.; Thadhani, R.; Getz, G.S. Vitamin d receptor signaling inhibits atherosclerosis in mice. Mol. Endocrinol. 2012, 26, 1091–1101. [Google Scholar] [CrossRef]
  19. DeLuca, H.F. Overwiew of general physiologic features and functions of vitamin D. Am. J. Clin. Nutr. 2004, 80 (Suppl. 6), 1689S–1696S. [Google Scholar] [CrossRef]
  20. Valtueña, J.; Gonzalez-Gross, M.; Huybrechts, I.; Breidenassel, C.; Ferrari, M.; Mouratidou, T.; Gottrand, F.; Dallongeville, J.; Azzini, E.; Sioen, I.; et al. Factors Associated with vitamin D deficiency in European adolescents: The HELENA Study. J. Nutr. Sci. Vitaminol. 2013, 59, 161–171. [Google Scholar] [PubMed]
  21. Earthman, C.P.; Beckman, L.M.; Masodkar, K.; Sibley, S.D. The link between obesity and 25-hydroxyvitamin D concentrations: Considerations and implications. Int. J. Obes. 2012, 36, 387–396. [Google Scholar] [CrossRef]
  22. Wortsman, J.; Matsuoka, L.Y.; Chen, T.C.; Lu, Z.; Holick, M.F. Decreased bioavailability of vitamin D in Obesity. Am. J. Clin. Nutr. 2000, 72, 690–693. [Google Scholar] [CrossRef]
  23. Snijder, M.B.; van Dam, R.M.; Visser, M.; Deeg, D.J.H.; Dekker, J.M.; Bouter, L.M.; Seidell, J.C.; Lips, P. Adiposity in relation to vitamin D status and parathyroid hormone levels: A population-based study in older men and women. J. Clin. Endocrinol. Metab. 2005, 90, 4119–4123. [Google Scholar] [CrossRef]
  24. Vimaleswaran, K.S.; Berry, D.J.; Lu, C.; Tikkanen, E.; Pilz, S.; Hiraki, L.T.; Cooper, J.D.; Dastani, Z.; Li, R.; Houston, D.K.; et al. Causal relationship between obesity and vitamin D status: Bi-directional Mendelian randomization analysis of multiple cohorts. PLoS Med. 2013, 10, e1001383. [Google Scholar] [CrossRef]
  25. 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]
  26. Kimball, M.; Holick, M. Official recommendations for vitamin D through the life stages in developed countries. Eur. J. Clin. Nutr. 2020, 74, 1514–1518. [Google Scholar] [CrossRef]
  27. Van Ballegooijen, A.J.; Kestenbaum, B.; Sachs, M.C.; de Boer, I.H.; Siscovick, D.S.; Hoofnagle, A.N.; Ix, J.H.; Visser, M.; Brouwer, I.A. Association of 25-hydroxyvitamin D and parathyroid hormone with incident hypertension: MESA (Multi-Ethnic Study of Atherosclerosis). J. Am. Coll. Cardiol. 2014, 63, 1214–1222. [Google Scholar] [CrossRef] [PubMed]
  28. Bouillon, R.; Marcocci, C.; Carmeliet, G.; Bikle, D.; White, J.H.; Dawson-Hughes, B.; Lips, P.; Munns, C.F.; Lazaretti-Castro, M.; Giustina, A.; et al. Skeletal and extraskeletal actions of vitamin D: Current evidence and outstanding questions. Endocr. Rev. 2019, 40, 51. [Google Scholar]
  29. Borba, V.Z.C.; Mañas, N.C.; Kulak, C.A.M. Deficiência de vitamina D—Por que, quando e como tratar. In Endocrinologia Clínica; Vilar, L., Ed.; Guanabara Koogan Ltda.: Rio de Janeiro, Brazil, 2013; pp. 1050–1064. [Google Scholar]
  30. Mechica, J.B. Raquitismo e osteomalacia. Arq. Bras. Endocrinol. Metabol. 1999, 43, 457–466. [Google Scholar] [CrossRef]
  31. Al-Daghri, N.M.; Alkharfy, K.M.; Al-Othman, A.; El-Kholie, E.; Moharram, O.; Alokail, M.S.; Al-Saleh, Y.; Sabico, S.; Kumar, S.; Chrousos, G.P. Vitamin D Supplementation an adjuvant therapy for patients with T2DM: An 18–month prospective interventional study. Cardiovasc. Diabetol. 2012, 11, 85. [Google Scholar] [CrossRef]
  32. Bokyung, M. Effects of Vitamin D on Blood Pressure and Endothelial Function. Korean J. Physiol. Pharmacol. 2013, 17, 385–392. [Google Scholar]
  33. Mc Gill, A.T.; Stewart, J.M.; Lithander, F.E.; Strik, C.M.; Poppitt, S.D. Relationships of low serum vitamin D3 with anthropometry in overweight and obesity. Nutr. J. 2008, 7, 4. [Google Scholar] [CrossRef] [PubMed]
  34. Yousef, S.; Colman, I.; Papadimitropoulos, M.; Manuel, D.; Hossain, A.; Faris, M.; Wells, G.A. Vitamin D and Chronic Diseases among First-Generation Immigrants: A Large-Scale Study Using Canadian Health Measures Survey (CHMS) Data. Nutrients 2022, 14, 1760. [Google Scholar] [CrossRef] [PubMed]
  35. Jablonski, N.G.; Chaplin, G. Human skin pigmentation, migration and disease susceptibility. Philos. Trans. R. Soc. B Biol. Sci. 2012, 367, 785–792. [Google Scholar] [CrossRef] [PubMed]
  36. Mithal, A.; Wahl, D.A.; Bonjour, J.-P.; Burckhardt, P.; Dawson-Hughes, B.; Eisman, J.A.; El-Hajj Fuleihan, G.; Josse, R.G.; Lips, P.; Morales-Torres, J. Global vitamin D status and determinants of hypovitaminosis D. In Osteoporosis International; Springer: London, UK, 2009; pp. 1807–1820. [Google Scholar]
  37. Diabetes Prevention Program Research Group. Long-term effects of lifestyle intervention or metformin on diabetes development and microvascular complications over 15-year follow-up: The Diabetes Prevention Program Outcomes Study. Lancet Diabetes Endocrinol. 2015, 3, 866–875. [Google Scholar] [CrossRef]
  38. Pittas, A.G.; Jorde, R.; Kawahara, T.; Dawson-Hughes, B. Vitamin D supplementation for prevention of type 2 diabetes mellitus: To D or not to D? J. Clin. Endocrinol. Metab. 2020, 105, 3721–3733. [Google Scholar] [CrossRef]
  39. de Sire, A.; Gallelli, L.; Marotta, N.; Lippi, L.; Fusco, N.; Calafiore, D.; Cione, E.; Muraca, L.; Maconi, A.; De Sarro, G.; et al. Vitamin D Deficiency in Women with Breast Cancer: A Correlation with Osteoporosis? A Machine Learning Approach with Multiple Factor Analysis. Nutrients 2022, 14, 1586. [Google Scholar] [CrossRef]
  40. Siniscalchi, A.; Lochner, P.; Anticoli, S.; Chirchiglia, D.; De Sarro, G.; Gallelli, L. What is the Current Role for Vitamin D and the Risk of Stroke? Curr. Neurovasc. Res. 2019, 16, 178–183. [Google Scholar] [CrossRef]
  41. Bouillon, R.; Manousaki, D.; Rosen, C.; Trajanoska, K.; Rivadeneira, F.; Richards, J.B. The health effects of vitamin D supplementation: Evidence from human studies. Nat. Rev. Endocrinol. 2022, 18, 96–110. [Google Scholar] [CrossRef]
  42. Codoner-Franch, P.; Tavarez-Alonso, S.; Simo-Jorda, R.; Laporta-Martin, P.; Carratala-Calvo, A.; Alonso-Iglesias, E. Vitamin D status is linked to biomarkers of oxidative stress, Inflammation, and endothelial activation in obese children. J. Pediatr. 2012, 161, 848–854. [Google Scholar] [CrossRef]
  43. 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] [PubMed]
  44. Akbari, M.; Ostadmohammadi, V.; Lankarani, K.B.; Tabrizi, R.; Kolahdooz, F.; Heydari, S.T.; Kavari, S.H.; Mirhosseini, N.; Mafi, A.; Dastorani, M.; et al. The Effects of Vitamin D Supplementation on Biomarkers of Inflammation and Oxidative Stress Among Women with Polycystic Ovary Syndrome: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Horm. Metab. Res. 2018, 50, 271–279. [Google Scholar] [CrossRef] [PubMed]
  45. Mansournia, M.; Ostadmohammadi, V.; Doosti-Irani, A.; Ghayour-Mobarhan, M.; Ferns, G.; Akbari, H.; Ghaderi, A.; Reza, 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]
  46. Sepidarkish, M.; Farsi, F.; Akbari-Fakhrabadi, M.; Namazi, N.; Almasi-Hashiani, A.; Hagiagha, A.M.; Heshmati, J. The effect of vitamin D supplementation on oxidative stress parameters: A systematic review and meta-analysis of clinical trials. Pharmacol. Res. 2019, 139, 141–152. [Google Scholar] [CrossRef]
  47. 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] [PubMed]
  48. Moslemi, E.; Musazadeh, V.; Kavyani, Z.; Naghsh, N.; Shoura, 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]
  49. Targher, G.; Bertolini, L.; Padovani, R.; Zenari, L.; Scala, L.; Cigolini, M.; Arcaro, G. Serum 25- Hydroxyvitamin D3 concentrations and carotid artery intima-thickness among type 2 diabetic patients. Clin. Endocrinol. 2006, 65, 593–597. [Google Scholar] [CrossRef]
  50. Braam, L.; Hoeks, A.P.G.; Brouns, F.; Hamulyak, K.; Gerichhausen, M.J.W.; Vermeer, C. Beneficial effects of vitamins D and K on the elastic properties of the vessel wall in postmenopausal women: A follow-up study. Thromb. Haemost. 2004, 91, 373–380. [Google Scholar] [CrossRef]
  51. Myrup, B.; Jensen, G.F.; McNair, P. Cardiovascular Risk Factors During Estrogen-Norethindrone and Cholecalciferol Treatment. Arch. Intern. Med. 1997, 152, 2265–2268. [Google Scholar] [CrossRef]
  52. 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 Women. J. Clin. Endocrinol. Metab. 2001, 86, 1633–1637. [Google Scholar] [CrossRef]
  53. Bolland, M.J.; Bacon, C.J.; Horne, A.M.; Mason, B.H.; Ames, R.W.; Wang, T.K.M.; Grey, A.B.; Gamble, G.D.; Reid, I.R. Vitamin D insufficiency and health outcomes over 5 y in older women. Am. J. Clin. Nutr. 2010, 91, 82–89. [Google Scholar] [CrossRef] [PubMed]
  54. Margolis, K.L.; Martin, L.W.; Ray, R.M.; Kerby, T.J.; Allison, M.A.; Curb, J.D.; Kotchen, T.A.; Liu, S.; Wassertheil-Smoller, S.; Manson, J.E. A Prospective Study of Serum 25-Hydroxyvitamin D Levels, Blood Pressure, and Incident Hypertension in Postmenopausal Women. Am. J. Epidemiol. 2011, 175, 22–32. [Google Scholar] [CrossRef] [PubMed]
  55. Syal, S.K.; Kapoor, A.; Bhatia, E.; Sinha, A.; Kumar, S.; Tewari, S.; Garg, N.; Goel, P. Vitamin D deficiency, coronary artery disease, and endothelial dysfunction: Observations from a coronary angiographic study in Indian patients. J. Invasive Cardiol. 2012, 24, 385–389. [Google Scholar]
  56. Ganji, V.; Zhang, X.; Shaikh, N.; Tangpricha, V. Serum 25-hydroxyvitamin D concentrations are associated with prevalence of metabolic syndrome and various cardiometabolic risk factors in US children and adolescents based on assay-adjusted serum 25-hydroxyvitamin D data from NHANES 2001–2006. Am. J. Clin. Nutr. 2011, 94, 225–233. [Google Scholar] [CrossRef] [PubMed]
  57. Lupton, J.R.; Faridi, K.F.; Martin, S.S.; Sharma, S.; Kulkarni, K.; Jones, S.R.; Michos, E.D. Deficient serum 25-hydroxyvitamin D is associated with an atherogenic lipid profile: The very large database of lipids (VLDL-3) study. J. Clin. Lipidol. 2016, 10, 72–81. [Google Scholar] [CrossRef]
  58. Petersen, R.A.; Dalskov, S.-M.; Sorensen, L.B.; Hjorth, M.F.; Andersen, R.; Tetens, I.; Krarup, H.; Ritz, C.; Astrup, A.; Michaelsen, K.F. Vitamin D status is associated with cardiometabolic markers in 8–11-year-old children, independently of body fat and physical activity. Br. J. Nutr. 2015, 114, 1647–1655. [Google Scholar] [CrossRef]
  59. Skaaby, T.; Husemoen, L.L.N.; Pisinger, C.; Jorgensen, T.; Thuesen, B.H.; Fenger, M.; Linneberg, A. Vitamin D status and changes in cardiovascular risk factors: A prospective study of a general population. Cardiology 2012, 123, 62–70. [Google Scholar] [CrossRef]
  60. Tarcin, O.; Yavuz, D.G.; Ozben, B.; Telli, A.; Ogunc, A.V.; Yuksel, M.; Toprak, A.; Yazici, D.; Sancak, S.; Deyneli, O.; et al. Effect of vitamin D deficiency and replacement on endothelial function in asymptomatic subjects. J. Clin. Endocrinol. Metab. 2009, 94, 4023–4030. [Google Scholar] [CrossRef]
  61. Tomaino, K.; Romero, K.; Robinson, C.; Baumann, L.; Hansel, N.; Pollard, S.; Gilman, R.; Lima, E.; Checkley, W.; PURA Study Investigators. Association between serum 25-hydroxy Vitamin D levels and blood pressure among adolescents in two resource-limited settings in Peru. Am. J. Hypertens. 2015, 28, 1017–1023. [Google Scholar] [CrossRef]
  62. Chiu, K.C.; Chu, A.; Go, V.; Liang, W.; Saad, M.F. Hypovitaminosis D is associated with insulin resistance and beta cell dysfunction. Am. J. Clin. Nutr. 2004, 79, 820–825. [Google Scholar] [CrossRef]
  63. Shoelson, S.E.; Herrero, L.; Naaz, A. Obesity, inflammation, and insulin resistance. Gastroenterology 2007, 132, 2169–2180. [Google Scholar] [CrossRef]
  64. Jaimungal, S.; Wehmeier, K.; Mooradian, A.D.; Haas, M.J. The emerging evidence for vitamin D-mediated regulation of apolipoprotein A-1 synthesis. Nutr. Res. 2011, 31, 805–812. [Google Scholar] [CrossRef]
  65. Ponda, M.P.; Huang, X.; Odeh, M.A.; Breslow, J.L.; Kaufman, H.W. Vitamin D may not improve lipid levels: A serial clinical laboratory data study. Circulation 2012, 126, 270–277. [Google Scholar] [CrossRef]
  66. Warren, T.; Mcallister, R.; Morgan, A.; Rai, T.; Mcgilligan, V.; Ennis, M.; Page, C.; Kelly, C.; Peace, A.; Corfe, B.; et al. The Interdependency and Co-Regulation of the Vitamin D and Cholesterol Metabolism. Cells 2021, 6, 2007. [Google Scholar] [CrossRef]
  67. Pacifico, L.; Anania, C.; Osborn, J.F.; Ferraro, F.; Bonci, E.; Olivero, E.; Chiesa, C. Low 25(OH)D3 levels are associated with total adiposity, metabolic syndrome, and hypertension in Caucasian children and adolescents. Eur. J. Endocrinol. 2011, 165, 603–611. [Google Scholar] [CrossRef]
  68. 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]
  69. Rostand, S.G. Ultraviolet light may contribute to geographic and racial blood pressure differences. Hypertension 1997, 30, 150–156. [Google Scholar] [CrossRef]
  70. Krause, R.; Buhring, M.; Hopfenmuller, W.; Holick, M.F.; Sharma, A.M. Ultraviolet B and blood pressure. Lancet 1998, 352, 709–710. [Google Scholar] [CrossRef]
  71. Wang, L.; Manson, J.E.; Buring, J.E.; Lee, I.-M.; Sesso, H.D. Dietary intake of dairy products, calcium, and vitamin D and the risk of hypertension in middle-aged and older women. Hypertension 2008, 51, 1073–1079. [Google Scholar] [CrossRef]
  72. Snijder, M.B.; Lips, P.T.; Seidell, J.C.; Visser, M.; Deeg, D.J.; 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. Int. Med. 2007, 261, 558–565. [Google Scholar] [CrossRef]
  73. 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]
  74. Rubin, M.R.; Maurer, M.S.; McMahon, D.J.; Bilezikian, J.P.; Silverberg, S.J. Arterial stiffness in mild primary hyperparathyroidism. J. Clin. Endocrinol. Metab. 2005, 90, 3326–3330. [Google Scholar] [CrossRef]
  75. Brunaud, L.; Germain, A.; Zarnegar, R.; Rancier, M.; Alrasheedi, S.; Caillard, C.; Ayav, A.; Weryha, G.; Mirallie, E.; Bresler, L. Serum aldosterone is correlated positively to parathyroid hormone (PTH) levels in patients with primary hyperparathyroidism. Surgery 2009, 146, 1035–1041. [Google Scholar] [CrossRef] [PubMed]
  76. Tomaschitz, A.; Ritz, E.; Pieske, B.; Rus-Machan, J.; Kienreich, K.; Verheyen, N.; Gaksch, M.; Grubler, M.; Fahrleitner-Pammer, A.; Mrak, P.; et al. Aldosterone and parathyroid hormone interactions as mediators of metabolic and cardiovascular disease. Metabolism 2014, 63, 20–31. [Google Scholar] [CrossRef]
  77. Tomaschitz, A.; Ritz, E.; Pieske, B.; Fahrleitner-Pammer, A.; Kienreich, K.; Horina, J.H.; Drechsler, C.; Marz, W.; Ofner, M.; Pieber, T.R.; et al. Aldosterone and parathyroid hormone: A precarious couple for cardiovascular disease. Cardiovasc. Res. 2012, 94, 10–19. [Google Scholar] [CrossRef] [PubMed]
  78. Resnick, L.M.; Muller, F.D.; Laragh, J.H. Calcium-regulating hormones in essential hypertension. Relation to plasma renin activity and sodium metabolism. Ann. Intern. Med. 1986, 105, 649–654. [Google Scholar] [CrossRef]
  79. Oz, F.; Cizgici, A.Y.; Oflaz, H.; Elitok, A.; Karaayvaz, E.B.; Mercanoglu, F.; Bugra, Z.; Omer, B.; Adalet, K.; Oncul, A. Impact of vitamin D insufficiency on the epicardial coronary flow velocity and endothelial function. Coron Artery Dis 2013, 24, 392–397. [Google Scholar] [CrossRef]
  80. Brewer, L.C.; Michos, E.D.; Reis, J.P. Vitamin D in atherosclerosis, vascular disease, and endothelial function. Curr. Drug Targets 2011, 12, 54–60. [Google Scholar] [CrossRef]
  81. 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]
  82. Molinari, C.; Uberti, F.; Grossini, E.; Vacca, G.; Carda, S.; Invernizzi, M.; Cisari, C. 1ɑ,25-Dihydroxycholecalciferol induces nitric oxide production in cultured endothelial cells. Cell. Physiol. Biochem. 2011, 27, 661–668. [Google Scholar] [CrossRef]
  83. Cardus, A.; Panizo Garcia, S.; Encinas Martin, M.; Dolcet Roca, X.; Gallego, C.; Aldea, M.; Fernandez, E.; Valdivielso Revilla, J.M. 1,25-dihydroxyvitamin D(3) regulates VEGF production through a vitamin D response element in the VEGF promoter. Atherosclerosis 2009, 204, 85–89. [Google Scholar] [CrossRef]
  84. Zhong, W.; Gu, B.; Gu, Y.; Groome, L.J.; Sun, J.; Wang, Y. Activation of vitamin D receptor promoters VEGF and CuZn-SOD expression in endothelial cells. J. Steroid. Biochem. Mol. Biol. 2013, 204, 85–89. [Google Scholar]
  85. Sypniewska, G.; Pollak, J.; Strozecki, P.; Camil, F.; Kretowicz, M.; Janikowski, G.; Mankowska-Cyl, A.; Pater, A.; Manitius, J. 25-hydroxyvitamin d, biomarkers of endothelial dysfunction and subclinical organ damage in adults with hypertension. Am. J. Hypertens. 2014, 27, 114–121. [Google Scholar] [CrossRef] [PubMed]
  86. Dong, J.H.; Wong, S.L.; Lau, C.W.; Lee, H.K.; Ng, C.F.; Zhang, L.; Yao, X.; Chen, Z.Y.; Vanhoutte, P.M.; Huang, Y. Calcitriol protects renovascular function in hypertension by down-regulating angiotensin II type I receptors and reducing oxidative stress. Eur. Heart J. 2012, 33, 2980–2990. [Google Scholar] [CrossRef] [PubMed]
  87. Harris, R.A.; Pedersen-White, J.; Guo, D.; Stallmann-Jorgensen, I.S.; Keeton, D.; Huang, Y.; Shah, Y.; Zhu, H.; Dong, Y. Vitamin D3 supplementation for 16 weeks improves flow-mediated dilatation in overweight African American adults. Am. J. Hypertens. 2011, 24, 557–562. [Google Scholar] [CrossRef] [PubMed]
  88. Stricker, H.; Bianda, F.T.; Guidicelli-Nicolosi, S.; Limoni, C.; Colucci, G. Effect of a single, oral, high-dose vitamin D supplementation on endothelial function in patients with peripheral arterial disease: A randomized controlled pilot study. Eur. J. Vasc. Endovasc. Surg. 2012, 44, 307–312. [Google Scholar] [CrossRef] [PubMed]
  89. Carey, R.M.; Whelton, P.K.; 2017 ACC/AHA Hypertension Guideline Writing Committee. Prevention, detection, evaluation, and management of high blood pressure in adults: Synopsis of the 2017 American College of Cardiology/American Heart Association Hypertension Guideline. Ann. Int. Med. 2018, 168, 351–358. [Google Scholar] [CrossRef]
Table 1. Vitamin D, oxidative stress and inflammation studies.
Table 1. Vitamin D, oxidative stress and inflammation studies.
Author(s), Year, Reference NumberCharacteristic of Study/ParticipantsVitamin D Protocol Oxidative Stress Parameters AnalyzedResults
Codoñer-Franch et al. (2012) [42]Cross-sectional study (66 obese Caucasian children aged 7–14 year)-3-nitrotyrosine; sVCAM-1; IL-6Inverse relationship between serum 25(OH)D levels with 3-nitrotyrosine (r = −0.424, p = 0.001) and sVCAM-1 (r = −0.272, p = 0.032). Comparing deficiency and sufficiency groups, IL-6 decreases significantly (18.0 [12.2–25.7] vs. 13.7 [10.7–19.3] pg/mL, p < 0.036).
Zhang et al. (2014) [43]Cross-sectional study (1488 chinese children aged 7–11 year)-CRP; IL-6; MDA; SODInverse association of serum 25(OH)D levels with CRP (1.21 ± 0.13 vs.1.04 ± 0.14), IL-6 (3.83 ± 0.33 vs. 3.58 ± 0.34), MDA (2.87 ± 0.21 vs. 2.83 ± 0.27), and SOD (95.38 ± 12.22 vs. 127.62 ± 15.98 U/mL, p < 0.001) comparing deficiency and sufficiency groups.
Akbari et al. (2018) [44]Systematic review and meta-analysis of randomized (7 studies involving polycystic ovary syndrome women)controlled trialsDifferent vitamin D supplementation protocols (400–7142 IU/d)CRP; MDA; NO; GSHVitamin D supplementation led to an increase in TAC, leading to a reduction in CRP (−1.03; 95%CI, −1.58, −0.49; p < 0.001) and MDA (−1.64, 95%CI −2.26 to −1.02, p < 0.001), however, there was no change in NO or GSH.
Mansournia et al. (2018) [45]Meta-analysis (33 studies involving adults)Different vitamin D supplementation protocols (10–11,200 IU/d)CRP; MDA; NO; GSH; TACSupplementation significantly reduced CRP (WMD 0.27; 95%CI, −0.35, −0.20; p < 0.001) and MDA (WMD −0.43, 95%CI −0.62, −0.25, p < 0.001); and increased NO (WMD 4.33, 95%CI 0.96, 7.70), GSH (WMD 82.59, 95%CI 44.37, 120.81, p < 0.001) and TAC (WMD 57.34, 95%CI 33.48, 81.20, p < 0.001)
Sepidarkish et al. (2018) [46]Meta-analysis (17 studies involving adults)Different vitamin D supplementation protocols (300–20,000 IU/m)TAC; GSH; MDA; NOSupplementation in 13 clinical trials showed increased levels of serum TAC (SMD: 0.54 mmol/L, 95%CI: 0.29 to 0.79; I2 = 65.4%, p = 0.001) and GSH (SMD: 0.33; 95%CI: 0.11 to 0.54, p = 0.003; I2 = 61.2%, p = 0.001). MDA decreased significantly (SMD: −0.40 mmol/L, 95%CI: −0.60 to −0.21, p < 0.001) and NO was not significant different (0.17, 95%CI: −0.10 to 0.45, I2 = 71.1%, p = 0.21).
Cojic et al. (2021) [47]Randomized Controlled Study (114 T2DM analyzed patients)14,000 IU/wk vitamin D supplementation for 6 m. Vitamin D deficient group [25(OH)D ≤ 50 nmol/L]: initial correction of vitamin D levels 50,000 IU/wk for 3 m. HbA1c; AOPP; HOMA-IR; MDA; CRP; TG/TBARS at 0, 3 and 6 mImproved HbA1c levels over the 3-month and 6-month period. Significant decrease inAOPP levels over the 3-month period. HOMA-IR, MDA and TG/TBARS was not statistically significant. The inflammatory marker—CRP had a decrease but not statistically significant.
Moslemi et al. (2022) [48]Meta-analysis (23 studies involving adults)Different vitamin D supplementation protocols (100–30,708 IU/d)CRP; TNF-α; MDA; IL-6; TAC; GSHSupplementation significantly reduced CRP (ES = −0.42; 95%CI: −0.55, −0.29, p < 0.001), TNF-α (ES = −0.27; 95%CI: −0.42, −0.12; p < 0.001), MDA concentrations (ES = −0.37; 95%CI: −0.48, −0.25, p < 0.001). No significant difference at IL-6 levels (ES = −0.35, 95%CI: −0.80, 0.10; p = 0.125), TAC (ES = 0.68; 95%CI: −0.31, 1.66, p = 0.179), GSH activity (ES = 0.08; 95%CI: −0.44, 0.60, p = 0.757)
Abreviations: 25-hydroxyvitamin D (25(OH)D); soluble Vascular Cell Adhesion Molecule-1 (sVCAM-1); Interleukin-6 (IL-6); Serum C-reactive protein (CRP); Malondialdehyde (MDA); Superoxide dismutase (SOD); Nitric oxide (NO); Glutathione (GSH); Total antioxidant capacity (TAC); Glycated haemoglobin (HbA1c); Advanced oxidation protein products (AOPP); Insulin resitance index (HOMA-IR); Tryglicerides/Thiobarbituric acid-reactive substances (TG/TBARS); Tumor necrosis factor-α (TNF-α); Type 2 Diabetes Mellitus (T2DM); Month (m); Week/s (wk); Year/s (yr); Day/s (d). Red color: Inflammatory markers.
Table 2. Vitamin D and CVD risk studies.
Table 2. Vitamin D and CVD risk studies.
Author(s), Year, Reference NumberCharacteristic of Study/ParticipantsVitamin D Protocol CVD Parameters AnalyzedResults
Myrup; Jensen; McNair (1992) [51]Double-blind, randomized study/74 healthy Danish women (>70 year)1 year of dailly 0.5 μg 1,25(OH)2D supplementationTC; HDL; BP at 0,3,6,9,12,18 m. No statistical significance for all parameters.
Pfeifer et al. (2001) [52]Double-blind, randomized, placebo-controlled study/148 (25(OH)D serum < 50 nmol/L) women (>70 year; mean 74 year) 8 wk of dailly supplementation of calcium 1.200 mg plus 800 IU of vitamin 1.25(OH)2D or just calcium 1.200 mg. SBP; DBP; HR at 0 and 8 wk.Compared with calcium, supplementation with vitamin D3 plus calcium decrease SBP in 9.3% (p = 0.02) and decrease HR in 5.4% (p = 0.02). Sixty subjects (81%) in the vitamin D3 plus calcium group compared with 35 (47%) subjects in the calcium group showed a decrease in SBP of 5 mmHg or more (p = 0.04)
Braam et al. (2004) [50]Randomized placebo-controlled study/108 healthy women (>50 year and <60 year; mean 55 year)3 year placebo or a daily supplement containingminerals and vitamin D. CC; DC; IMT; E at 0 and 3 yr.No statistical significance for all parameters.
Targher et al. (2004) [49]Case-control study/ 390 diabetes patients (130 with vitamin D hypovitaminosis: 25(OH)D ≤ 37.5 nmol/L) and 390 matched-control individuals. (mean 58 year)Compared winter serum [25(OH)D] concentrations in 390consecutive type 2 diabetic patients and 390 nondiabetic controlswho were comparable for age and sex.Prevalence of hypovitaminosis; IMT; HbA1c; Fibrinogen; CRPIn diabetic patients, prevalence of hypovitaminosis D was higher (34.0 vs. 16.4%, p < 0.001)and significantly higher IMT (1.10 ± 0.15 vs. 0.87 ± 0.14 p < 0.001), HbA1c (7.5 ± 1.3 vs. 7.2 ± 1.4 p < 0.05), fibrinogen (4.69 ± 0.8 vs. 4.21 ± 0.9 p < 0.001)and CRP
(4.98 ± 6·5 vs. 4.30 ± 6.1 p < 0.001) concentrations was seen in these condition.
Bolland et al. (2009) [53]Randomized placebo-controlled study/1471 (25(OH)D serum >25 nmol/L) healthy women (>55 year; mean 74 year) supplementing calcium-Incidence of stroke; MI; CA; HF; adverse BP; TC at 0 and 5 yr.No statistical significance for all parameters comparing deficient (<50 nmol/L serum 25OHD) and non-deficient (>50 nmol/L serum 25OHD) at 0 and 5 yr.
Oh et al. (2009) [17]In vitro study/Macrophage of 62 diabetics, hypertensive and vitamin D deficient (<80 nmol/L) patients (mean 55 year)Same patient’s macrophages were cultured in vitamin D deficient or in 1,25(OH)2D3 medium and exposed to modified LDL.Foam cell formation.1,25(OH)2D suppressed foam cell formation by reducing acetylated or oxidized LDL uptake - almost 50%less cholesteryl ester formation.
Margolis et al. (2012) [54]Longitudional study/4863 healthy women (50–79 yr; mean 66 year)-SBP; DBPOver 7 yr, there were no significant differences in the adjusted mean change in systolic or diastolic blood pressure by quartile of 25(OH)D. Incident hypertension was slightly and statistic significantly lower in the third quartil of 25(OH)D (48– < 65 nmol/L) compared with the lowest quartile.
Syal et al. (2012) [55]Observational study/100 Indian individuals - 81% male (37–75 yr, mean 57 year)-Prevalence of severe CAD; diffuse CAD; FMD; brachial artery FMD Vitamin D deficiency group (25(OH)D level of <20 ng/mL) had significantly higher prevalence of severe CAD (53% vs. 38% p = 0.03), diffuse CAD (56% vs. 34% p = 0.03), impaired FMD (values < 4.5%; 50.6% vs. 7%; p < 0.002) and significantly lower brachial artery flow-mediated dilation (4.57% vs. 10.68%: p < 0.001).
Schmitt et al. (2018) [6]Observational, cross-sectional cohort study/463 women (45–75 yr; mean 58 year)-TC; Triglycerides; HDL. Women with low 25(OH)D levels (<30 ng/mL) had higher TC (p = 0.031); triglycerides (OR 1.55, 95%CI = 1.13–2.35) and low HDL levels (OR 1.60, 95%CI = 1.19–2.40).
Children, Adolescents and Young Adults Studies
Ganji et al. (2011) [56]Observational, cross-sectional cohort study/5867 children and adolescents (12–19 year). Serum 25(OH)D: low (<48.1 nmol/mL), medium (48.1 to <66.2 nmol/L), and high (>66.2 nmol/L).-SBPSBP significantly higher in the low serum 25(OH)D group than in the medium group and high groups (109.8 ± 0.5 vs. 108.2 ± 0.4 and 108.4 ± 0.4 mmHg, p = 0.01)
Lupton et al. (2016) [57]Observational, cross-sectional cohort study/20,360 adults. Serum 25(OH)D: deficient (<20 ng/mL), intermediate (≥20–30 ng/mL), and optimal (≥30 ng/mL).-TC; Triglycerides; LDL. 25(OH)D deficiency is related to a significant worsening in TC, LDL, and triglycerides levels observed in a study reporting alteration of +9.4%, +13.5%, and +26.4%, respectively
Petersen et al. (2015) [58]Observational, cross-sectional cohort study/782 children (8–11 year). -DBPFor every 10 mmol/L increase in serum 25(OH)D there is an association with a decrease in DBP (p = 0.02), independent of the BMI.
Skaaby et al. (2012) [59]Prospective Study/ 4330 adults. Serum 25(OH)D: sufficiency (> 48.0nmol/l).-TC; Triglycerides; LDL. Vitamin D sufficiency has been shown to reduce TC, LDL, and triglycerides with mean differences of −0.17, −0.19, and −0.12, respectively
Tarcin et al. (2009) [60]Case-control study/23 asymptomatic vitamin D-deficient (25(OH)D < 25 nmol/l) and 23 matched-control individuals (mean 25(OH)D level of 75 nmol/l). (mean 23 year)Vitamin D-deficient group: 300,000 IU/mintramuscular for 3 monthsFMD; TBARSSignificantly lower FMD in 25(OH)D-deficient subjects than controls (7.0 ± 3.2% vs. 11.2 ± 5.2%; p = 0.001), improved after replacement therapy (10.4 ± 3.3%; p = 0.002). Significantly lower post treatment values of TBARS (4.7 ± 1.7 to 2.9 ± 0.7 ng/mgMDA; p = 0.0001).
Tomaino et al. (2015) [61]Observational, cross-sectional cohort study/1441 adolescents (13–15 year). Serum 25(OH)D: deficiency (serum 25(OH)D3 < 50 nmol/L).-DBP DBP higher in the 25(OH)D-deficient group compared with the 25(OH)D3-sufficient group (p = 0.04)
Abreviations: 1,25-dihydroxyvitamin D(1,25(OH)2D); Total cholesterol (TC); High density lipoprotein (HDL); Blood pressure (BP); 25-hydroxyvitamin D (25(OH)D); Sistolic blood pressure (SBP); Diastolic blood pressure (DBP); Heart rate (HR); Flow-mediated dilation (FMD); Thiobarbituric acid-reactive substances (TBARS); Compliance coefficient (CC); Distensibility coefficient (DC); Intima-media thickness (IMT); Glycated haemoglobin (HbA1c); Serum C-reactive protein (CRP); Young’s Modulus (E); Myocardial infarction (MI); Cancer (CA); Heart failure (HF); Coronary artery disease (CAD); Low density lipoprotein (LDL); Month (m); Week/s (wk); Year/s (yr); Day/s (d). For convertion of 25(OH)D from ng/ml to nmol/L, multiply by 2.496.
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

Renke, G.; Starling-Soares, B.; Baesso, T.; Petronio, R.; Aguiar, D.; Paes, R. Effects of Vitamin D on Cardiovascular Risk and Oxidative Stress. Nutrients 2023, 15, 769. https://doi.org/10.3390/nu15030769

AMA Style

Renke G, Starling-Soares B, Baesso T, Petronio R, Aguiar D, Paes R. Effects of Vitamin D on Cardiovascular Risk and Oxidative Stress. Nutrients. 2023; 15(3):769. https://doi.org/10.3390/nu15030769

Chicago/Turabian Style

Renke, Guilherme, Bernardo Starling-Soares, Thomaz Baesso, Rayssa Petronio, Danilo Aguiar, and Raphaela Paes. 2023. "Effects of Vitamin D on Cardiovascular Risk and Oxidative Stress" Nutrients 15, no. 3: 769. https://doi.org/10.3390/nu15030769

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