Next Article in Journal
Flavonoid-Rich Sambucus nigra Berry Extract Enhances Nrf2/HO-1 Signaling Pathway Activation and Exerts Antiulcerative Effects In Vivo
Previous Article in Journal
Mitochondrial Dysfunction in the Pathogenesis and Treatment of Oral Inflammatory Diseases
Previous Article in Special Issue
In Vivo Regulation of Signal Transduction Pathways by Vitamin D Stabilizes Homeostasis of Human Immune Cells and Counteracts Molecular Stress
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of Vitamin D and Its Molecular Bases in Insulin Resistance, Diabetes, Metabolic Syndrome, and Cardiovascular Disease: State of the Art

1
Department of Internal Medicine, National Relevance and High Specialization Hospital Trust ARNAS Civico Di Cristina Benfratelli, 90127 Palermo, Italy
2
Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties, [PROMISE], University of Palermo, 90127 Palermo, Italy
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(20), 15485; https://doi.org/10.3390/ijms242015485
Submission received: 4 September 2023 / Revised: 11 October 2023 / Accepted: 18 October 2023 / Published: 23 October 2023
(This article belongs to the Special Issue The Role of Vitamin D in Human Health and Diseases 3.0)

Abstract

:
In the last decade, an increasing awareness was directed to the role of Vitamin D in non-skeletal and preventive roles for chronic diseases. Vitamin D is an essential hormone in regulating calcium/phosphorous balance and in the pathogenesis of inflammation, insulin resistance, and obesity. The main forms of vitamin D, Cholecalciferol (Vitamin D3) and Ergocalciferol (Vitamin D2) are converted into the active form (1,25-dihydroxyvitamin D) thanks to two hydroxylations in the liver, kidney, pancreas, and immune cells. Some anti-inflammatory cytokines are produced at higher levels by vitamin D, while some pro-inflammatory cytokines are released at lower levels. Toll-Like Receptor (TLR) expression is increased, and a pro-inflammatory state is also linked to low levels of vitamin D. Regardless of how it affects inflammation, various pathways suggest that vitamin D directly improves insulin sensitivity and secretion. The level of vitamin D in the body may change the ratio of pro- to anti-inflammatory cytokines, which would impact insulin action, lipid metabolism, and the development and function of adipose tissue. Many studies have demonstrated an inverse relationship between vitamin D concentrations and pro-inflammatory markers, insulin resistance, glucose intolerance, metabolic syndrome, obesity, and cardiovascular disease. It is interesting to note that several long-term studies also revealed an inverse correlation between vitamin D levels and the occurrence of diabetes mellitus. Vitamin D supplementation in people has controversial effects. While some studies demonstrated improvements in insulin sensitivity, glucose, and lipid metabolism, others revealed no significant effect on glycemic homeostasis and inflammation. This review aims to provide insight into the molecular basis of the relationship between vitamin D, insulin resistance, metabolic syndrome, type 1 and 2 diabetes, gestational diabetes, and cardiovascular diseases.

1. Introduction

In recent years, attention to the role of vitamin D in different fields is growing. Vitamin D is a liposoluble prohormone with endocrine, autocrine, and paracrine functions and is fundamental to bone metabolism [1]. Vitamin D has a role in extra-skeletal functions; consequentially, there is a relationship between vitamin D deficiency and some pathologic conditions, including diabetes, metabolic syndrome, non-alcoholic liver disease, autoimmune diseases, hypertension, cardiovascular disease, and cancer [2,3,4,5,6,7,8,9] (Figure 1). Moreover, the recent pandemic of COVID-19 has underlined the possible therapeutic role of Vitamin D in some aspects of the infection and the association between severe vitamin D deficiency and COVID-19-related health outcomes [10,11,12]. Many studies have reported the existence of immuno-modulatory effects of vitamin D and that its deficiency may be associated with a sub-inflammatory state [13]. Diabetes and metabolic syndrome represent a major clinical and public health problem. The disease burden related to diabetes and metabolic syndrome is increasing significantly, particularly in older subjects [14,15]. According to the International Diabetes Federation, data released in 2021 showed that 537 million adults live with diabetes worldwide. The total number is predicted to rise to 643 million by 2030 and to 783 million by 2045 instead of the previous estimation of 693 million [16]. Many epidemiological and observational studies have found an association between vitamin D insufficiency and the incidence of type 1 and type 2 Diabetes [17,18,19,20,21]. In this sense, many studies reported the existence of different mechanisms able to explain the potential role of vitamin D in glucose metabolism, such as the preservation of the β-cell function and slow failure of residual β-cell function in patients with type 1 diabetes and latent autoimmune diabetes [22,23]. Furthermore, vitamin D determines direct stimulation of insulin secretion and improves peripheral insulin resistance by reducing systemic inflammation via the vitamin D receptor on pancreatic beta cells and in muscles and the liver [24,25,26]. This last mechanism also plays a key role in metabolic syndrome development [27]. The lack of vitamin D receptors in cardiovascular tissue increased ventricular mass dysregulation of metalloproteinases and fibroblasts, promoting the fibrotic process and ventricular dilatation [28].
Given this background, an extensive search of SCOPUS, PubMed, and CENTRAL was performed using the following string ((vitamin D) or (calcifediol)) or (ergocholecalciferol)) AND (systematic review [pt] or meta-analysis [pt]) and 2017:2023 [dp]). The search string retrieved 1575 manuscripts. The hand-searching of principal generalist, human nutrition, and basic research journals was carried out as well. Two authors (V.O. and A.T.) independently reviewed the titles, abstracts, and full texts of the retrieved articles to determine their potential inclusion. Any disagreements were resolved via discussion with a third author (S.C.). Manuscripts regarding the role of vitamin D in insulin resistance, type 1 and 2 diabetes, gestational diabetes, metabolic syndrome, and cardiovascular disease were extracted for this review. This review aims to explore the molecular basis of the role of vitamin D in insulin resistance, type 1 and 2 diabetes, gestational diabetes, metabolic syndrome, and cardiovascular disease.

2. Vitamin D Metabolism

Vitamin D is a liposoluble prohormone that humans can acquire via nutrition and synthesis in the skin during exposure to UV radiation [29]. Vitamin D3 (Cholecalciferol) is the main source of vitamin D, and vitamin D2 (Ergocalciferol) is the form through which vitamin D exists.
Most of the amount of Cholecalciferol comes from the endogenous production in the skin after sun exposure; a small amount of Cholecalciferol has an exogenous origin and derives from foods. Ergocalciferol is contained in dairy products and nutritional supplements, and it is a non-animal form of vitamin D [30].
Once in circulation, Cholecalciferol and Ergocalciferol are converted in the liver tissue by the action of vitamin D-25-hydroxylase (CYP2R1) to 25-hydroxyvitamin D or calcifediol [25 (OH) D]; subsequently, 25(OH)D undergoes a second conversion, by the enzyme 25-hydroxyvitamin D-1α-hydroxylase (CYP27B1), into active and bioavailable vitamin D (1,25-dihydroxyvitamin or calcitriol—CT) [1,25 (OH)2 D] [31,32,33]. This reaction takes place mainly in the kidney. At that point, 1,25 (OH)2 D performs its functions by binding to the vitamin D receptor (VDR), expressed in the cytoplasm of cells, forming a VDR-RXR hormone complex (vitamin D receptor—retinoid X receptor) via the stimulation of the heterodimerization of the VDR with the retinoid X receptor [15]. In the nucleus, it regulates the expression of many genes via their up or downregulation [32]. 1,25 (OH)2 D has about 1000-fold higher affinity than 25(OH) for the VDR. CYP27B1 is also expressed in other tissues, like activated macrophages, microglia, parathyroid glands, breast, colon, and keratinocytes; 1,25 (OH)2 D has autocrine and paracrine effects [34,35] (Figure 2). It is known that vitamin D is associated with bone health and can play an essential role in other systems, including the immune system. These extra skeletal actions are available because of the presence of VDR and hydroxylation enzymes in different tissues such as the pancreas, kidney, muscles, liver, and others. Vitamin D supplementation (VDS) has hormonal, anti-inflammatory, anti-apoptotic, anti-fibrotic activities, antioxidant, and immune-modulatory effects [36,37], and also plays a role in insulin resistance via the reduction in the expression of some pro-inflammatory cytokines like interleukin-1 (IL-1) and IL-6 [38].

3. Vitamin D and Insulin Resistance

3.1. Vitamin D, Insulin Resistance, and Molecular Mechanisms

Vitamin D is involved in several non-skeletal health diseases, including common metabolic disorders like Metabolic Syndrome (MetS), Type 2 Diabetes (T2DM), Impaired Fasting Glucose (IFG), Non-Alcoholic Fatty Liver Disease (NAFLD), and Polycystic ovarian syndrome (PCOS), which are all characterized by insulin resistance (IR) [39,40,41]. It has been demonstrated that there is an inverse association between vitamin D deficiency and the Homeostatic Model Assessment of Insulin Resistance (HOMA-IR), which is used as the measure of insulin resistance and defined as an increase in insulin secretion necessary for the maintenance of glycemic homeostasis [42]. Therefore, the supplementation of vitamin D reduces the risk of insulin resistance and circulating levels of insulin [42,43]; the inverse correlation between vitamin D and HOMA-IR becomes more robust with increasing Body Mass Index (BMI) [42].
Molecular mechanisms underlying the pathophysiological hypothesis of the possible association between hypovitaminosis D and insulin resistance are mainly associated with the expression of insulin receptors, and the production of inflammatory cytokines and polymorphism of VDR expressed in the β-cells of the pancreas. In particular, vitamin D acts upon gene transcription via genomic and non-genomic mechanisms. Based on the above, hypovitaminosis D and insulin resistance are genetically interrelated [40,42,43].
Concerning insulin receptor expression, it was found that vitamin D increases receptor expression in muscle, liver, and adipose tissue, improving insulin sensitivity [42]. In detail, it was shown that vitamin D works as an epigenetic factor, affecting the transcription level of many genes involved in insulin sensitivity, like Insulin Receptor Substrate (IRS), which is increased by 2.4-fold in high-fat mice models treated with vitamin D [42]. As a result, insulin sensitivity improves in the target tissues because IRS protein increases insulin sensitivity [42]. In addition, vitamin D improves the sensitivity of insulin receptors to insulin and glucose transport and promotes the conversion of proinsulin to insulin [43,44,45].
Vitamin D deficiency increases the expression of pro-inflammatory cytokines, which can be the cause of insulin resistance in patients with relatively higher BMI; it has been observed that obesity is associated with hypovitaminosis D because of three reasons: less exposure to sunlight, the low intake of vitamin D via nutrition, and the sequestration of vitamin D in the adipose tissue [42]. In addition to this mechanism, it was found that high secretion of the anti-diabetic hormone leptin, whose levels are deregulated by abdominal adiposity, is associated with insulin resistance. This means that high doses of vitamin D supplements can decrease leptin levels and reduce BMI in insulin-resistant patients [42]. This effect would be linked to a reduced caloric intake mediated by the binding of vitamin D to its receptors in the paraventricular nucleus of the hypothalamus.
As regards VDR, it is an endocrine member of the nuclear receptor superfamily for steroid hormones, and it works as a transcription factor that mediates the action of vitamin D via the control of the expression of hormone-sensitive genes, like Calmodulin-Dependent Kinase (CaMKs), which in turn stimulates VDR-mediated transcription by phosphorylation levels of VDR [40]. The function of β-cells may be affected by vitamin D via direct and indirect mechanisms. The direct mechanism consists of binding of vitamin D to VDR in β-cells, helping in the release of insulin secretion [42,46]; the indirect mechanism is related to the regulation by vitamin D of calcium flux via the pancreatic β-cell because insulin secretion is strongly dependent on calcium [46]. This could be the reason why tissue calcium levels (adipose tissue and skeletal muscle) affect IR [46]. It was recently discovered that the deletion of macrophage VDR promotes insulin resistance [40].
Recently, it was found that the enzyme-activating vitamin D, 1-α-hydroxylase, is present in β-cells [46]. While the non-genomic functions of vitamin D are carried out via the activation of numerous signaling molecules (phosphatidylinositol-3 kinase, phospholipase C (PLC), Ca2+-calmodulin kinase II (CaMPKII), protein kinase A (PKA), mitogen-activated protein kinases (MAPK)2s2+, src, and protein kinase C (PKC)) that in turn interact with vitamin D response elements (VDRE) on the promoter of vitamin D-sensitive genes [42,47]. In addition to those mentioned, there are other mechanisms by which vitamin D, via an alternative non-genomic pathway, influences intracellular signaling molecules or transcription factors that condition the expression of various genes.
This action could explain some of the modulatory effects of vitamin D on innate and adaptive immunity, cell antiviral responses, and cell survival. This pathway involves the protein–protein interaction between the VDR and a target protein. Some of these target proteins are represented by kinase I-κB (IKK)β, one of the upstream regulators of the NF-κB canonical pathway [33], signal transducers and transcription activators (Stat)1, Runt-related transcription factor (RunX) 1 [35], c-jun, β-catenin, and cAMP response element binding protein [48].

3.2. Studies and Research

A recent meta-analysis, which included 9232 participants, has studied genetic associations of four polymorphisms in the VDR with insulin-resistant diseases, particularly TaqI, BsmI, ApaI, and FokI variants. It was found that there is an association between insulin resistance-related diseases (mostly with PCOS and MetS than T2DM) and the VDR ApaI variant (mostly G allele than T allele) in Asians and populations who lived in middle-latitude districts. The BsmI (mostly A allele than G allele) and TaqI variants (T/C allele) were more prevalent in dark-pigmented Caucasians. At the same time, there was no association between the VDR FokI variant and insulin resistance-related diseases in populations with different skin pigments and in different latitudes [40].
Beneficial effects of high-dose vitamin D (≥2000 mg/day) and calcium (≥1000 mg/day) in both short-term and long-term (>12 weeks) combined vitamin D and calcium supplementation were found [46]. However, the results obtained so far are conflictive because some trials reported that the supplementation of vitamin D does not reduce insulin resistance [29]. Further studies, like long-term and large-scale randomized controlled trials, are needed.

4. Vitamin D and Type 2 Diabetes Mellitus (T2DM)

4.1. Vitamin D, T2DM and Molecular Mechanisms

It is well known that T2DM is a public health challenge worldwide, accounting for approximately 87–91% of all cases of diabetes. Type 2 Diabetes Mellitus is a chronic metabolic disorder characterized by inadequate insulin production and consequentially high blood glucose [49]. T2DM constitutes an essential risk factor for premature death and adverse complications, micro and macrovascular, such as blindness, stroke, heart attack, amputation, and kidney failure [50], and also determines and impairs quality of life [51]. According to OMS, 762 million people worldwide suffer from pre-diabetes [52], which is strongly connected with obesity [53].
A lack or insufficiency of Vitamin D is associated with macrovascular and microvascular complications of T2DM [54]. Obesity, prediabetes, and T2DM are often characterized by low circulating vitamin D levels [55].
VDR, implicated in the systemic effect of vitamin D, is also expressed in high insulin-sensitive tissues (pancreas, adipose tissue, and muscle) [56]. In the body, vitamin D is an epigenetic factor mediating the transcription level and enhancing insulin sensitivity [42].

4.2. Studies and Research

According to a recent meta-analysis, vitamin D supplementation improves glycemic homeostasis and insulin sensitivity [57]. It also seems to work as an anti-diabetic factor by regulating insulin sensitivity and production, controlling parathyroid hormone levels, and anti-inflammatory cytokine effects [58,59]. Vitamin D has been identified as a potential prevention and treatment strategy [60]. Low 25-hydroxyvitamin D (25(OH) D) levels are highly prevalent among T2DM patients [61]. The effects of vitamin D supplementation may explain the association between vitamin D and T2DM because it prevented the increase in plasma HbA1c levels and in IR [62,63,64].
The gold standard for evaluating glycemic control in T2DM is represented by glycated hemoglobin (HbA1c) in line with the UK Prospective Diabetes Study [65].
In line with recent studies, vitamin D supplementation is implicated in plasma HbA1c reduction, suggesting that vitamin D can contribute to reducing the development of diabetic complications [66]. Also, studies have found that vitamin D supplementation improved beta cell function [67] and insulin sensitivity [56,68,69,70], especially in those at high risk for diabetes.
In particular, vitamin D has a role in lipid metabolism in adipose tissue [71] and may decrease inflammation [72]. In pancreatic tissue, Vitamin D protects β-cells function, reducing local inflammation [73,74]. A key role is represented by the activation of the VDR expressed in the pancreatic beta-cell. Indeed, mice lacking VDR have impaired insulin secretion [75], and the addition of Vitamin D stimulates pancreatic cells, resulting in increased insulin secretion [76]. It is worth outlining that the human insulin receptor gene promoter contains a Vitamin D response element, suggesting that transcriptional activation of the gene may be favored by calcitriol administration [77,78]. A calcium-dependent mechanism mediates insulin secretion. Vitamin D may play a role [79] in regulating the opening and closure of calcium channels, mediating the calcium flux in beta cells, and interacting with receptors (VDR and 1,25 D3-MARRS). Therefore, vitamin D deficiency causing an alteration in calcium flux could interfere with normal insulin secretion [80,81]. In addition, vitamin D is involved in skeletal muscle metabolism, insulin sensitivity, and lipid composition [82]. Consequently, increasing circulating vitamin D concentration could affect tissue energy and metabolism, improving systemic insulin sensitivity. The skeletal muscle is crucial in insulin sensitivity, involving 70–90% of total glucose disposal during the post-prandial period [83,84,85]. Thus, vitamin D supplementation might improve skeletal muscle glucose handling and, as a consequence, insulin sensitivity [86]. Vitamin D also regulates the adipose tissue, and hypovitaminosis may play a role in obesity and fat mass due to the restoration of Vitamin D, a fat-soluble vitamin, in the adipose tissue [87]. According to Bajaj et al., hypovitaminosis also seems to increase microvascular complications such as diabetes retinopathy, diabetic neuropathy, diabetic nephropathy, and diabetic foot ulcers [88], and a meta-analysis demonstrated that increased circulating vitamin D levels protect the kidney from injury and ameliorate proteinuria in T2DM patients [89]. Concerning microvascular complications, vitamin D deficiency may be involved in diabetic neuropathy interfering with nociceptor functions by causing diabetic nerve damage [90], and diabetic retinopathy increasing the severity and playing a role in the pathogenesis via its effects on the immune system and angiogenesis [91]. Lastly, a lack of Vitamin D promotes macrovascular complications such as endothelial dysfunction and arterial stiffness [92,93], peripheral arterial disease, and carotid arterial plaque [94]. Vitamin D might have a direct effect on vascular stiffness. Vascular smooth muscles (VSMCs) and endothelial cells express 1α-hydrolase, which is involved in the conversion of 25(OH)D to calcitriol [95]. It has been shown that this enzyme is activated in primary cultures of human umbilical vein endothelial cells by inflammatory molecules such as TNF-α and lipopolysaccharide [96]. In addition, it was found that vitamin D has a direct effect on vascular tone by reducing calcium influx [97]. Moreover, an extrarenal activation of vitamin D was suggested as a possible contributor to hypertension and arterial stiffness [98].
The vitamin D level is inversely related to blood pressure [99]. Experimental and human studies showed that subjects with 1,25(OH)2D3 deficiency have increased activity of the RAAS, both in the body and in the kidney, developing hypertension [100]. An increased plasma renin concentration and low 1,25(OH)2D3 levels may elevate sympathetic activity and enhance intra-glomerular pressure, predisposing to AH, a decline in GFR, and subsequent cardiovascular damage [101]. The knocking out of either the VDR or the 1α-hydroxylase gene in mice upregulates RAAS activity and induces HT [102,103], while treatment of these animals with 1,25(OH)2D3 suppresses the RAAS activity [103]. In addition, the VDR is expressed in vascular tissues, including the myocardium, renin-producing juxtaglomerular cells, and vascular smooth muscle, which directly influences calcium influx, muscle relaxation, and diastolic function [104,105].
Hence, hypovitaminosis D (as deficiency or insufficiency) embraces several complications in diabetic patients; therefore, screening for vitamin D levels in T2DM patients may play a crucial role in defining the outcomes.

5. Vitamin D and Type 1 Diabetes Mellitus (T1DM)

5.1. Vitamin D, T1DM and Molecular Mechanisms

Type 1 Diabetes Mellitus (T1DM) is a chronic autoimmune disease related to an immune system alteration that destroys pancreatic ß cells with a consequent quantitative or qualitative dysfunction of insulin [106]. The prevalence of T1DM has steadily increased over the past few decades in most countries [107]. Patients with T1DM are genetically susceptible to developing autoimmune diseases, with an increased risk of developing the disease among first-degree relatives [108,109]. Currently, the research aims to identify genetic and environmental factors predisposing to the onset of the disease. Current knowledge suggests that an important role could be played by vitamin D, which in the first years of life modulates the still-growing immune system, which plays a crucial role in the development of self-tolerance [110,111,112]. Vitamin D signaling impairment, especially in the first years of life, increases the risk of autoimmunity [113,114,115]. Given the role that vitamin D plays in the immune system, it is believed that it may have a protective role in the development of T1DM [116].
The discovery of vitamin D receptors throughout the body has opened up new reflections on its possible implication in other diseases, including autoimmune diseases such as T1DM and multiple sclerosis [117]. Indeed, VDR is also expressed in immune cells, effectively regulating innate and adaptive immune responses [118].
The expression of 1α-hydroxylase CYP27B1 in specific immune system cells explains how these can regulate vitamin D levels [119]. Some studies have shown that the activity of macrophages/monocytes, antigen pre-transmitter cells, T cells, and B cells is regulated by vitamin D [120]. It plays a role in the modulation of the activity of dendritic cells [121]. In the presence of 1,25(OH)2D3, dendritic cells produce fewer inflammatory factors such as tumor necrosis factor-α and interleukin-12, producing a less anti-inflammatory tolerance state characterized by increased production of interleukin-10 [121].
1,25(OH)2D3 promotes macrophage differentiation, which is essential for the activation of involutional inflammation in animal models of T1DM, to the anti-inflammatory phenotype (M1→M2) via the VDR- PPARgamma signaling pathway [122]. These properties of vitamin D on the regulation of the inflammatory response are very interesting in T1DM because, in the pancreas of affected patients, there is an inflammatory infiltrate composed of T lymphocytes, B lymphocytes, and macrophages. In animal models of T1DM, such as non-obese diabetic mice, high doses of calcitriol and non-high calcium vitamin D analogs arrest involutional inflammation, as indicated by reduced effector T cell numbers and the induction of T-reg cells [123,124,125].
Fronczak et al. reported that increased maternal intake of vitamin D in food reduced the risk of autoimmunity against pancreatic beta cells in their offspring; there is no effect of 1alpha,25-dihydroxyvitamin D3 on residual beta cell function and insulin requirements in adults [126].

5.2. Studies and Research

According to evidence (systematic reviews and meta-analyses) [127,128] on the link between vitamin D levels and T1DM, adequate vitamin D status in the first years of life reduces the risk of diabetes [17,110,112,129], and vitamin D deficiency is more common in people with T1DM [130,131]. A cross-sectional study revealed that 70% of children with T1DM had a vitamin D deficiency [132], and rickets are associated with an increased risk of T1DM [133]. Also, the TEDDY study reported that a higher infant concentration of 25(OH)D is associated with lower islet autoimmunity [134]. In contrast, a birth cohort study in Finland suggested that sufficient vitamin D supplementation could assist in decreasing T1DM risk [133].
The risk of developing T1DM before age 15 is associated with a reduction in serum vitamin D levels, as demonstrated by a case–control study that was part of EURODIAB (OR 0.63) [135]. Human studies report the relationship between VDR polymorphisms and T1DM risk and β cell function. Although 25D is the major circulating form, pancreatic β cells can convert 25D to 1,25D [136]. This implies that a small role in beta cell survival in T1DM can be played by exogenous and circulating 1,25 D. Anyhow, rising 25D levels could be helpful as a substrate for the formation of 1,25D by beta cells while circulating 1,25 D could exert autocrine and paracrine effects. Considering beta cell injury at the clinical diagnosis, vitamin D is much less likely to be helpful after disease onset [137]. A meta-analysis conducted by Najjar et al. found no critical effect of a genetically determined reduction in 25(OH)D concentrations by selected polymorphisms on T1DM risk. A meta-analysis conducted by Gregoriou et al. showed that vitamin D supplementation in patients with T1DM resulted in a reduction in daily insulin requirements, as well as improving fasting C-peptide (FCP), stimulated C-peptide (SCP), and HbA1c [138].
Although not all studies agree [139], there are many observational studies that show a strong association between vitamin D deficiency and T1DM [140].

6. Vitamin D and Gestational Diabetes Mellitus (GDM)

6.1. Pathophysiology of Vitamin D Levels in Pregnancy

In pregnancy, numerous physiological alterations of the maternal metabolism are necessary for the normal development of the fetus. During pregnancy, a relationship between the maternal and fetal vitamin D status underlines the importance of an adequate vitamin D level in this period. Gestational vitamin D metabolism adaptations include a characteristic physiological growing of 1,25(OH)2D in maternal blood. It rises at the beginning of gestation and reaches its highest levels in the third trimester, where it presents two to three times the levels found in non-pregnant women. Several studies have shown a correlation between vitamin D levels and GDM [141]. GDM is defined as glucose intolerance, and IR was first diagnosed in pregnant women [142]. GDM affects up to 14% of pregnancies [143]. Inadequate glycemic control in women with GDM leads to short- and long-term maternal complications, including gestational hypertension, preeclampsia, macrosomia, congenital abnormalities, hypoglycemia in the newborn, and an increased risk of T2DM after pregnancy [144,145]. GDM is compared to a form of impaired glucose tolerance, similar to prediabetes in non-pregnant individuals, and represents a global public health problem related to serious health problems in the mother and newborn [146]. Women with a history of GDM have an increased risk of developing IR syndrome (IRS) and cardiovascular disease (CVD) later in their lives [147]. The rate of women who develop T2DM within 5–10 years ranges from 20 to 60% [148,149]. The risks of occurrence of MetS and CVD are three times higher in women with GDM. Indeed, children born to women with GDM have a higher risk of developing impaired glucose tolerance and obesity. The pathogenesis of GDM has not yet been cleared. Some studies [150,151] suggest that the onset and development of GDM are closely related to genetic factors (insulin resistance, family history of diabetes, and immune dysfunction) and environmental factors (dietary structure and pancreatic β cell damage).

6.2. Vitamin D, GDM, and Molecular Mechanisms

Vitamin D can support insulin secretion and normal glucose tolerance [152]. Vitamin D deficiency seems closely related to the onset of GDM. Among the factors that may play a role in the onset of GDM is chronic low-grade inflammation [153]. The increased degree of inflammation in early pregnancy is related to an increased risk of GDM and the development of hyperglycemia [154]. Moreover, in women with GDM, oxidative stress has been found [155,156,157], while antioxidant status is downregulated [158]. Oxidative stress plays an important role in both the pathogenesis and complications of GDM [159]. A significant inverse association exists between serum vitamin D concentrations and low-grade inflammation [160]. The low levels of vitamin D trigger inflammatory responses via the NF-kB pathway by regulating p-p65/RelB in pancreas tissue [161] upwards. Excessive Ca2+ and reactive oxygen species (ROS) in ß cells, both in vitamin D deficiency, result in cell death and promote diabetes [162].
Furthermore, some genes that protect against the onset of diabetes are inactivated by hypermethylation [163]. Vitamin D prevents hypermethylation by increasing the expression of DNA demethylases in more regions of genes that protect against diabetes [162]. In addition, a significant inverse association was also found between serum calcium concentrations, which is positively regulated by vitamin D, and obesity risk as another diabetes risk factor [164].

6.3. Studies and Research

Several longitudinal prospective cohort studies have reported the risk of GDM with serum vitamin D concentrations in early pregnancy [165]. A meta-analysis conducted by Chunfeng Wu et al. [166] showed that vitamin D supplementation has a beneficial effect on lipidic assessment: it increases HDL-Cholesterol (HDL-C) levels and is useful for reducing serum Total Cholesterol (TC) and LDL-Cholesterol (LDL-C) levels of patients with GDM. However, no single opinion exists between this meta-analysis and the previous ones [167,168,169].
Preceding meta-analyses [168] pointed out that vitamin D can improve LDL-C levels but does not affect triglycerides (TG), TC, and HDL-C. The short duration of the studies could explain this. Several studies [170,171,172] have shown that when GDM patients have abnormal lipid metabolism, their risk of pregnancy complications increases. Studies [173,174] proved that vitamin D deficiency is associated with a higher incidence of T2DM and vitamin D supplementation can dramatically increase insulin sensitivity in people with IR and vitamin D deficiency. IR and insufficient secretion underlie the pathogenesis of GDM [175]. According to a network meta-analysis conducted by Shixiao Jin et al. to evaluate the effects of vitamin D supplementation, it was best for reducing fast plasma glucose (FPG) and improving HOMA-IR compared to the effects of other nutritional strategies [176]. Vitamin D deficiency is a frequent phenomenon after pregnancy; one study showed that at 25–28 weeks of gestation, the concentration of 25(OH)D (the active form of the vitamin within the body) in GDM patients is significantly reduced [177]. Another systematic review and meta-analysis conducted by Wang M. et al. showed how vitamin D supplementation in a population of women with GDM can statistically significantly reduce serum FPG, insulin, and HOMA-IR, as well as complications related to childbirth (cesarean section, maternal hospitalization, and postpartum hemorrhage) and newborns (hyperbilirubinemia, giant children, hypoglycemia, polyhydramnios, fetal distress, and premature delivery). Vitamin D deficiency is considered a potential risk factor for abnormal glucose metabolism; Zhang et al. [178] conducted a study that showed that low vitamin D levels in the blood may increase the risk of GDM and that adequate vitamin D supplementation may improve GDM status. 25(OH)D can not only regulate insulin secretion but also stimulate insulin receptor expression to promote insulin sensitivity [179], achieving the effect of lowering blood sugar. In addition, vitamin D has antioxidant effects, which can reduce β islet cell damage and apoptosis β of islet cells via active oxidative groups [180]. Patients with GDM can increase their 25(OH)D concentration via vitamin D supplementation, thereby improving insulin resistance and decreasing blood sugar [181].

7. Vitamin D, Metabolic Syndrome (MetS), and Cardiovascular Disease (CVD)

7.1. MetS and CVD: Burden of the Problem

MetS is related to abdominal obesity, IR, hypertension, and dyslipidemia [182]. The diagnosis of MetS includes waist circumference (WC), FPG, TG levels, HDL-C levels, total cholesterol levels, and blood pressure (BP) [183]. The MetS increases the risk of developing T2DM associated with long-term microvascular and macrovascular damage [184] and CVDs. CVDs are one of the significant causes of disability and death worldwide [185]. Atherosclerosis is the primary etiology of CVDs, and it is considered a chronic inflammatory condition [186]. Several studies have also documented that a decrease in antioxidant levels and an increase in inflammatory and oxidative stress biomarkers may be involved in the pathophysiology of T2DM complications [187] and the onset of CVDs [188]. The inflammatory process can be triggered by metabolic disorders such as atherogenic dyslipidemia (higher TG and apolipoprotein B, small low-density lipoprotein cholesterol LDL-C particles, and low HDL-C concentrations), T2DM, and increased inflammatory cytokines [189]. Consequently, the inflammatory cascade may initiate plaque formation, endothelial damage, and, ultimately, plaque rupture [186]. The pathophysiology of endothelial dysfunction includes the overproduction of reactive oxidative species, inflammatory cytokines, pro-atherogenic lipoproteins, and an imbalance between vasodilating and vasoconstricting molecules. The impairment of vasodilation may be due to reduced bioavailability of nitric oxide (NO), produced by the endothelial cells and involved in multiple physiological processes, including vasodilation, inflammation, and platelet aggregation [190]. On the other hand, dyslipidemia is associated with insulin resistance and elevated risk of CVD events [191,192]. There are numerous risk factors for MetS e CVDs; among these, the dietary factor is among the most important [193], such as high-calorie and high-fat diets [194].

7.2. Vitamin D, MetS, CVD, and Molecular Mechanisms

Vitamin D deficiency patients are a risk factor for MetS [195]. Vitamin D deficiency can affect insulin secretion and sensitivity and play an essential role in the onset of MetS [27]. Furthermore, a study found that vitamin D supplementation had a positive effect on lipid profile, IR, hyperglycemia, obesity, and hypertension and then on the treatment of MetS-related disorders [196].
Vitamin D can reduce Oxidative Stress (OS) using upregulating cellular Glutathione (GSH) and antioxidant systems such as glutathione peroxidase and superoxide dismutase [197]. Also, vitamin D can inhibit Reactive Oxygen Species (ROS) secretion [198]. VDRs are expressed in different tissues, notably endothelial cells, vascular smooth muscle cells, and cardiomyocytes, and regulate the expression of the target gene [199]. Vitamin D3, furthermore, is a direct transcriptional regulator of endothelial Nitric Oxide (NO) synthase. In this pathophysiological situation, OS plays a crucial role in cellular injury, in which the production of reactive ROS suppresses the antioxidant defense system of the cells, which consequently causes cellular death [200]. Under physiologic conditions, the antioxidant defense systems maintain the oxidant-antioxidant balance by adjusting the altering levels of oxidants [201]. The antioxidant defense systems include enzymes such as glutathione peroxidase, catalase, superoxide dismutase, and other compounds (albumin and GSH).
Furthermore, different nutrients such as vitamins and minerals can also affect the antioxidant balance [202,203,204]. Accordingly, vitamin D has been proposed to have antioxidant properties. The association between VDS and MetS is controversial. The benefits of VDS in the treatments of MetS and its disorders connected include improved arterial stiffness, mitochondrial oxidation, and phospholipid metabolism; increased lipoprotein lipase activity, peripheral insulin sensitivity, and β-cell function; and decreased inflammatory cytokines and parathyroid hormone levels and renin–angiotensin–aldosterone system activity [205,206,207,208].
Various underlying mechanisms have been suggested for the association of serum vitamin D with MetS and its components. Firstly, there is an inverse significant relation between blood vitamin D concentration and abdominal obesity.
Vitamin D is a fat-soluble vitamin and tends to be stored in adipose tissues; so, its bioavailability and circulating levels are lower in those with abdominal obesity, so the synthesis in the liver of obese individuals is lower compared to ordinary people [87]. Considering the central role of vitamin D in the expression of insulin receptors and increasing insulin responsiveness for glucose transporters (GLUTs), serum vitamin D deficiency is involved in the incidence of insulin resistance and type 2 diabetes [77]. Also, vitamin D is known as an antihypertensive agent because of its direct effect on vascular cells, suppression of the renin–angiotensin–aldosterone system, calcium metabolism, and prevention of secondary hyperparathyroidism [209], in particular, parathyroid hormone (PTH), which is also involved in the process of lipogenesis; Vitamin D deficiency can favor greater adiposity by promoting an increase in parathyroid hormone levels and the inflow of calcium in adipocytes, thus increasing lipogenesis and inhibiting the lipolysis in adipocytes [210].
Another hypothesis suggests that excess body fat retains the metabolites of this vitamin, and cholecalciferol produced in the skin or acquired via the diet is partially “sequestered” by body fat before being transported to the liver and undergoing the first hydroxylation [211].
Obese persons may have low levels of 25(OH)D in their blood due to adipose tissue impeding vitamin D absorption and usage or because they spend less time outside, leading to insufficient vitamin D production in the skin [212]. Other proposed mechanisms include high expressions of the vitamin D receptor in adipose tissue and the possibility of vitamin D playing a role in the pathogenesis of the metabolic syndrome [213]. The benefits of Vitamin D supplements in the treatments of MetS and its disorders connected include improved arterial stiffness, mitochondrial oxidation, and phospholipid metabolism; increased lipoprotein lipase activity, peripheral insulin sensitivity, and β-cell function; and decreased inflammatory cytokines and parathyroid hormone levels and renin–angiotensin–aldosterone system activity [205,206,207]. In conclusion, there is no clear and demonstrated relationship between low serum 25(OH)D levels and MetS because of the lack of long-term studies.

7.3. Studies and Research

Zhu and Heil reported that serum 25D level was linked to the risk factors for MetS [214]. In a meta-analysis study, Jafari et al. [215] reported that vitamin D supplementation improved the lipid profile of patients with T2DM. In another meta-analysis, vitamin D intake significantly decreased insulin resistance in people with T2DM [216]. Several RCTs have studied the impact of vitamin D supplements on lipid profiles, glucose homeostasis, and C-reactive protein (CRP) in persons with CVDs [217]. Some studies reported no significant relationship between VDS and MetS in adults [218,219,220]. Therefore, the association between VDS and MetS still needs evidence to demonstrate whether VDS helps treat MetS. In a meta-analysis, Ostadmohammadi, Milajerdi et al. demonstrated the beneficial effects of vitamin D supplementation on reductions in fasting glucose, insulin concentrations, and HOMA-IR. In addition, the pooled analysis revealed a significant increase in serum HDL-C concentrations after vitamin D therapy and a significant reduction in CRP levels. However, supplementation did not affect TG, TC, and LDL-C levels [221]. Kai-Jie, Zhong-Tao et al. conducted a meta-analysis to study the effect of vitamin D on MetS in adults using relevant biomarkers such as anthropometric parameters, BP, blood lipid profile, blood sugar, OS, and vitamin D toxicity. Vitamin D did not affect waist circumference, body mass index, body fat percentage, and BP. VDS significantly reduced FPG but did not affect HDL-C, LDL-C, TC, and TG blood levels. For OS parameters, VDS significantly lowered malondialdehyde and hypersensitive CRP [222]. De Paula et al. [223], in a systematic review, meta-analysis, and randomized clinical trials (RCTs), investigated the effects of micronutrients on BP in patients with T2DM. In this systematic review, a reduction in BP, especially systolic BP, was demonstrated. Observational and experimental data favor the concept that vitamin D is associated with the pathogenesis of arterial hypertension [224,225]. A possible mechanism for this link involves the inhibition of the renin–angiotensin–aldosterone system by vitamin D. Additionally, in the presence of hypovitaminosis D, an alternative mechanism could be related to the secondary hyperparathyroidism and relative hypocalcemia that are commonly seen in these patients [226]. In a meta-analysis, Hajhashemy Z, Shahdadian F et al. illustrated that the highest level of blood vitamin D, compared with the lowest level, was significantly linked to lower odds of MetS in cross-sectional studies on the adult population. In addition, based on dose–response analysis, each 25 nmol/L (or 10 ng/mL) increment in 25(OH)D was associated with a 15% decreased chance of MetS.

8. Discussion and Conclusions

Data reported in our review support the notion that Vitamin D levels are associated with T1DM and T2DM, GDM, MetS, and CVDs. There is some experimental and epidemiological evidence for the administration of Vitamin D in these different diseases. In Table 1, we have summarized the results of meta-analyses and systematic reviews on the effectiveness of vitamin D administration and their dosages in the various conditions that we have dealt with in this review. However, data from randomized clinical trials, which are highly heterogeneous, yielded contrasting data. Although the possibility of preventing the onset of the disease, vitamin D administration should be started very early in life or even during pregnancy in T1DM and GDM; moreover, different data showed that vitamin D administration improves glucose metabolism and the risk of T2DM and metabolic syndrome, randomized clinical studies showed contradictory results for vitamin D supplementation in the management of altered metabolic states. In this sense, further studies are necessary to determine the fundamental role of vitamin D deficiency and if it can be considered a causal factor in altered metabolism.

Author Contributions

Conceptualization, C.A. and S.C.; methodology, C.A. and S.C.; software, C.A. and S.C.; validation, C.A. and S.C.; formal analysis, V.O. and A.T.; investigation, V.O. and A.T.; resources, L.M., S.A., V.O. and A.T.; data curation, L.M., S.A., V.O. and A.T.; writing—original draft preparation, C.A.; writing—review and editing, C.A, L.M., S.A. and S.C.; visualization, L.M.; supervision, S.C.; project administration, C.A. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Dattola, A.; Silvestri, M.; Bennardo, L.; Passante, M.; Scali, E.; Patruno, C.; Nisticò, S.P. Role of Vitamins in Skin Health: A Systematic Review. Curr. Nutr. Rep. 2020, 9, 226–235. [Google Scholar] [CrossRef] [PubMed]
  2. Umar, M.; Sastry, K.S.; Chouchane, A.I. Role of Vitamin D Beyond the Skeletal Function: A Review of the Molecular and Clinical Studies. Int. J. Mol. Sci. 2018, 19, 1618. [Google Scholar] [CrossRef] [PubMed]
  3. Maddaloni, E.; Cavallari, I.; Napoli, N.; Conte, C. Vitamin D and Diabetes Mellitus. Front. Horm. Res. 2018, 50, 161–176. [Google Scholar] [CrossRef]
  4. Mitri, J.; Muraru, M.D.; Pittas, A.G. Vitamin D and Type 2 Diabetes: A Systematic Review. Eur. J. Clin. Nutr. 2011, 65, 1005–1015. [Google Scholar] [CrossRef] [PubMed]
  5. Targher, G.; Bertolini, L.; Scala, L.; Cigolini, M.; Zenari, L.; Falezza, G.; Arcaro, G. Associations between Serum 25-Hydroxyvitamin D3 Concentrations and Liver Histology in Patients with Non-Alcoholic Fatty Liver Disease. Nutr. Metab. Cardiovasc. Dis. 2007, 17, 517–524. [Google Scholar] [CrossRef]
  6. Chen, X.; Zhou, M.; Yan, H.; Chen, J.; Wang, Y.; Mo, X. Association of Serum Total 25-Hydroxy-Vitamin D Concentration and Risk of All-Cause, Cardiovascular and Malignancies-Specific Mortality in Patients with Hyperlipidemia in the United States. Front. Nutr. 2022, 9, 971720. [Google Scholar] [CrossRef]
  7. Wimalawansa, S.J. Non-Musculoskeletal Benefits of Vitamin D. J. Steroid Biochem. Mol. Biol. 2018, 175, 60–81. [Google Scholar] [CrossRef]
  8. Teleni, L.; Baker, J.; Koczwara, B.; Kimlin, M.G.; Walpole, E.; Tsai, K.; Isenring, E.A. Clinical Outcomes of Vitamin D Deficiency and Supplementation in Cancer Patients. Nutr. Rev. 2013, 71, 611–621. [Google Scholar] [CrossRef]
  9. Khademi, Z.; Hamedi-Shahraki, S.; Amirkhizi, F. Vitamin D Insufficiency Is Associated with Inflammation and Deregulation of Adipokines in Patients with Metabolic Syndrome. BMC Endocr. Disord. 2022, 22, 223. [Google Scholar] [CrossRef]
  10. Argano, C.; Mallaci Bocchio, R.; Natoli, G.; Scibetta, S.; Lo Monaco, M.; Corrao, S. Protective Effect of Vitamin D Supplementation on COVID-19-Related Intensive Care Hospitalization and Mortality: Definitive Evidence from Meta-Analysis and Trial Sequential Analysis. Pharmaceuticals 2023, 16, 130. [Google Scholar] [CrossRef]
  11. Corrao, S.; Mallaci Bocchio, R.; Lo Monaco, M.; Natoli, G.; Cavezzi, A.; Troiani, E.; Argano, C. Does Evidence Exist to Blunt Inflammatory Response by Nutraceutical Supplementation during COVID-19 Pandemic? An Overview of Systematic Reviews of Vitamin D, Vitamin C, Melatonin, and Zinc. Nutrients 2021, 13, 1261. [Google Scholar] [CrossRef] [PubMed]
  12. Argano, C.; Mallaci Bocchio, R.; Lo Monaco, M.; Scibetta, S.; Natoli, G.; Cavezzi, A.; Troiani, E.; Corrao, S. An Overview of Systematic Reviews of the Role of Vitamin D on Inflammation in Patients with Diabetes and the Potentiality of Its Application on Diabetic Patients with COVID-19. Int. J. Mol. Sci. 2022, 23, 2873. [Google Scholar] [CrossRef] [PubMed]
  13. Barragan, M.; Good, M.; Kolls, J.K. Regulation of Dendritic Cell Function by Vitamin D. Nutrients 2015, 7, 8127–8151. [Google Scholar] [CrossRef]
  14. Saklayen, M.G. The Global Epidemic of the Metabolic Syndrome. Curr. Hypertens. Rep. 2018, 20, 12. [Google Scholar] [CrossRef] [PubMed]
  15. Argano, C.; Natoli, G.; Mularo, S.; Nobili, A.; Monaco, M.L.; Mannucci, P.M.; Perticone, F.; Pietrangelo, A.; Corrao, S. Impact of Diabetes Mellitus and Its Comorbidities on Elderly Patients Hospitalized in Internal Medicine Wards: Data from the RePoSi Registry. Healthcare 2022, 10, 86. [Google Scholar] [CrossRef]
  16. International Diabetes Federation. Diabetes Is “A Pandemic of Unprecedented Magnitude” Now Affecting One in 10 Adults Worldwide. Diabetes Res. Clin. Pract. 2021, 181, 109133. [Google Scholar] [CrossRef]
  17. Dong, J.-Y.; Zhang, W.; Chen, J.J.; Zhang, Z.-L.; Han, S.-F.; Qin, L.-Q. Vitamin D Intake and Risk of Type 1 Diabetes: A Meta-Analysis of Observational Studies. Nutrients 2013, 5, 3551–3562. [Google Scholar] [CrossRef]
  18. Littorin, B.; Blom, P.; Schölin, A.; Arnqvist, H.J.; Blohmé, G.; Bolinder, J.; Ekbom-Schnell, A.; Eriksson, J.W.; Gudbjörnsdottir, S.; Nyström, L.; et al. Lower Levels of Plasma 25-Hydroxyvitamin D among Young Adults at Diagnosis of Autoimmune Type 1 Diabetes Compared with Control Subjects: Results from the Nationwide Diabetes Incidence Study in Sweden (DISS). Diabetologia 2006, 49, 2847–2852. [Google Scholar] [CrossRef]
  19. Song, Y.; Wang, L.; Pittas, A.G.; Del Gobbo, L.C.; Zhang, C.; Manson, J.E.; Hu, F.B. Blood 25-Hydroxy Vitamin D Levels and Incident Type 2 Diabetes: A Meta-Analysis of Prospective Studies. Diabetes Care 2013, 36, 1422–1428. [Google Scholar] [CrossRef]
  20. Afzal, S.; Bojesen, S.E.; Nordestgaard, B.G. Low 25-Hydroxyvitamin D and Risk of Type 2 Diabetes: A Prospective Cohort Study and Meta-analysis. Clin. Chem. 2013, 59, 381–391. [Google Scholar] [CrossRef]
  21. Lee, C.J.; Iyer, G.; Liu, Y.; Kalyani, R.R.; Bamba, N.; Ligon, C.B.; Varma, S.; Mathioudakis, N. The Effect of Vitamin D Supplementation on Glucose Metabolism in Type 2 Diabetes Mellitus: A Systematic Review and Meta-Analysis of Intervention Studies. J. Diabetes Complicat. 2017, 31, 1115–1126. [Google Scholar] [CrossRef] [PubMed]
  22. Sharma, S.; Biswal, N.; Bethou, A.; Rajappa, M.; Kumar, S.; Vinayagam, V. Does Vitamin D Supplementation Improve Glycaemic Control In Children With Type 1 Diabetes Mellitus?—A Randomized Controlled Trial. J. Clin. Diagn. Res. 2017, 11, SC15–SC17. [Google Scholar] [CrossRef] [PubMed]
  23. Ataie-Jafari, A.; Loke, S.-C.; Rahmat, A.B.; Larijani, B.; Abbasi, F.; Leow, M.K.S.; Yassin, Z. A Randomized Placebo-Controlled Trial of Alphacalcidol on the Preservation of Beta Cell Function in Children with Recent Onset Type 1 Diabetes. Clin. Nutr. 2013, 32, 911–917. [Google Scholar] [CrossRef]
  24. Contreras-Bolívar, V.; García-Fontana, B.; García-Fontana, C.; Muñoz-Torres, M. Mechanisms Involved in the Relationship between Vitamin D and Insulin Resistance: Impact on Clinical Practice. Nutrients 2021, 13, 3491. [Google Scholar] [CrossRef]
  25. Gysemans, C.A.; Cardozo, A.K.; Callewaert, H.; Giulietti, A.; Hulshagen, L.; Bouillon, R.; Eizirik, D.L.; Mathieu, C. 1,25-Dihydroxyvitamin D3 Modulates Expression of Chemokines and Cytokines in Pancreatic Islets: Implications for Prevention of Diabetes in Nonobese Diabetic Mice. Endocrinology 2005, 146, 1956–1964. [Google Scholar] [CrossRef]
  26. Park, S.; Kim, D.S.; Kang, S. Vitamin D Deficiency Impairs Glucose-Stimulated Insulin Secretion and Increases Insulin Resistance by Reducing PPAR-γ Expression in Nonobese Type 2 Diabetic Rats. J. Nutr. Biochem. 2016, 27, 257–265. [Google Scholar] [CrossRef]
  27. Melguizo-Rodríguez, L.; Costela-Ruiz, V.J.; García-Recio, E.; De Luna-Bertos, E.; Ruiz, C.; Illescas-Montes, R. Role of Vitamin D in the Metabolic Syndrome. Nutrients 2021, 13, 830. [Google Scholar] [CrossRef] [PubMed]
  28. Mancuso, P.; Rahman, A.; Hershey, S.D.; Dandu, L.; Nibbelink, K.A.; Simpson, R.U. 1,25-Dihydroxyvitamin-D3 Treatment Reduces Cardiac Hypertrophy and Left Ventricular Diameter in Spontaneously Hypertensive Heart Failure-Prone (Cp/+) Rats Independent of Changes in Serum Leptin. J. Cardiovasc. Pharmacol. 2008, 51, 559–564. [Google Scholar] [CrossRef] [PubMed]
  29. Pieńkowska, A.; Janicka, J.; Duda, M.; Dzwonnik, K.; Lip, K.; Mędza, A.; Szlagatys-Sidorkiewicz, A.; Brzeziński, M. Controversial Impact of Vitamin D Supplementation on Reducing Insulin Resistance and Prevention of Type 2 Diabetes in Patients with Prediabetes: A Systematic Review. Nutrients 2023, 15, 983. [Google Scholar] [CrossRef] [PubMed]
  30. Ross, A.C.; Taylor, C.L.; Yaktine, A.L.; Del Valle, H.B. (Eds.) Institute of Medicine (US) Committee to Review Dietary Reference Intakes for Vitamin D and Calcium. In Dietary Reference Intakes for Calcium and Vitamin D; The National Academies Collection: Reports Funded by National Institutes of Health; National Academies Press (US): Washington, DC, USA, 2011. [Google Scholar]
  31. Haussler, M.R.; Haussler, C.A.; Jurutka, P.W.; Thompson, P.D.; Hsieh, J.C.; Remus, L.S.; Selznick, S.H.; Whitfield, G.K. The Vitamin D Hormone and Its Nuclear Receptor: Molecular Actions and Disease States. J. Endocrinol. 1997, 154, S57–S73. [Google Scholar]
  32. Adams, J.S.; Rafison, B.; Witzel, S.; Reyes, R.E.; Shieh, A.; Chun, R.; Zavala, K.; Hewison, M.; Liu, P.T. Regulation of the Extrarenal CYP27B1-Hydroxylase. J. Steroid Biochem. Mol. Biol. 2014, 144 Pt A, 22–27. [Google Scholar] [CrossRef]
  33. Klopot, A.; Hance, K.W.; Peleg, S.; Barsony, J.; Fleet, J.C. Nucleo-Cytoplasmic Cycling of the Vitamin D Receptor in the Enterocyte-Like Cell Line, Caco-2. J. Cell. Biochem. 2007, 100, 617–628. [Google Scholar] [CrossRef] [PubMed]
  34. Charoenngam, N.; Shirvani, A.; Holick, M.F. Vitamin D for Skeletal and Non-Skeletal Health: What We Should Know. J. Clin. Orthop. Trauma 2019, 10, 1082–1093. [Google Scholar] [CrossRef] [PubMed]
  35. Battault, S.; Whiting, S.J.; Peltier, S.L.; Sadrin, S.; Gerber, G.; Maixent, J.M. Vitamin D Metabolism, Functions and Needs: From Science to Health Claims. Eur. J. Nutr. 2013, 52, 429–441. [Google Scholar] [CrossRef] [PubMed]
  36. Chagas, C.E.A.; Borges, M.C.; Martini, L.A.; Rogero, M.M. Focus on Vitamin D, Inflammation and Type 2 Diabetes. Nutrients 2012, 4, 52–67. [Google Scholar] [CrossRef] [PubMed]
  37. Hewison, M. An Update on Vitamin D and Human Immunity. Clin. Endocrinol. 2012, 76, 315–325. [Google Scholar] [CrossRef]
  38. Selvaraj, P.; Harishankar, M.; Afsal, K. Vitamin D: Immuno-Modulation and Tuberculosis Treatment. Can. J. Physiol. Pharmacol. 2015, 93, 377–384. [Google Scholar] [CrossRef]
  39. Wang, S.; Cai, B.; Han, X.; Gao, Y.; Zhang, X.; Wang, R.; Zhang, Y.; Chen, Q. Vitamin D Supplementation for Non-alcoholic Fatty Liver Disease in Type 2 Diabetes Mellitus: A Protocol for a Systematic Review and Meta-Analysis. Medicine 2020, 99, e20148. [Google Scholar] [CrossRef]
  40. Han, F.; Lv, Y.; Gong, L.; Liu, H.; Wan, Z.; Liu, L. VDR Gene Variation and Insulin Resistance Related Diseases. Lipids Health Dis. 2017, 16, 157. [Google Scholar] [CrossRef]
  41. Sindhughosa, D.A.; Wibawa, I.D.N.; Mariadi, I.K.; Somayana, G. Additional Treatment of Vitamin D for Improvement of Insulin Resistance in Non-Alcoholic Fatty Liver Disease Patients: A Systematic Review and Meta-Analysis. Sci. Rep. 2022, 12, 7716. [Google Scholar] [CrossRef]
  42. Rafiq, S.; Jeppesen, P.B. Vitamin D Deficiency Is Inversely Associated with Homeostatic Model Assessment of Insulin Resistance. Nutrients 2021, 13, 4358. [Google Scholar] [CrossRef] [PubMed]
  43. Mo, M.; Shao, B.; Xin, X.; Luo, W.; Si, S.; Jiang, W.; Wang, S.; Shen, Y.; Wu, J.; Yu, Y. The Association of Gene Variants in the Vitamin D Metabolic Pathway and Its Interaction with Vitamin D on Gestational Diabetes Mellitus: A Prospective Cohort Study. Nutrients 2021, 13, 4220. [Google Scholar] [CrossRef] [PubMed]
  44. Wang, M.; Chen, Z.; Hu, Y.; Wang, Y.; Wu, Y.; Lian, F.; Li, H.; Yang, J.; Xu, X. The Effects of Vitamin D Supplementation on Glycemic Control and Maternal-Neonatal Outcomes in Women with Established Gestational Diabetes Mellitus: A Systematic Review and Meta-Analysis. Clin. Nutr. 2021, 40, 3148–3157. [Google Scholar] [CrossRef] [PubMed]
  45. Huang, S.; Fu, J.; Zhao, R.; Wang, B.; Zhang, M.; Li, L.; Shi, C. The Effect of Combined Supplementation with Vitamin D and Omega-3 Fatty Acids on Blood Glucose and Blood Lipid Levels in Patients with Gestational Diabetes. Ann. Palliat. Med. 2021, 10, 5652–5658. [Google Scholar] [CrossRef]
  46. Asbaghi, O.; Khosroshahi, M.Z.; Kashkooli, S.; Abbasnezhad, A. Effect of Calcium-Vitamin D Co-Supplementation on Insulin, Insulin Sensitivity, and Glycemia: A Systematic Review and Meta-Analysis of Randomized Clinical Trials. Horm. Metab. Res. 2019, 51, 288–295. [Google Scholar] [CrossRef]
  47. Szymczak-Pajor, I.; Śliwińska, A. Analysis of Association between Vitamin D Deficiency and Insulin Resistance. Nutrients 2019, 11, 794. [Google Scholar] [CrossRef]
  48. Hii, C.S.; Ferrante, A. The Non-Genomic Actions of Vitamin D. Nutrients 2016, 8, 135. [Google Scholar] [CrossRef]
  49. Muoio, D.M.; Newgard, C.B. Mechanisms of Disease:Molecular and Metabolic Mechanisms of Insulin Resistance and Beta-Cell Failure in Type 2 Diabetes. Nat. Rev. Mol. Cell Biol. 2008, 9, 193–205. [Google Scholar] [CrossRef]
  50. Lotfy, M.; Adeghate, J.; Kalasz, H.; Singh, J.; Adeghate, E. Chronic Complications of Diabetes Mellitus: A Mini Review. Curr. Diabetes Rev. 2017, 13, 3–10. [Google Scholar] [CrossRef]
  51. Zheng, Y.; Ley, S.H.; Hu, F.B. Global Aetiology and Epidemiology of Type 2 Diabetes Mellitus and Its Complications. Nat. Rev. Endocrinol. 2018, 14, 88–98. [Google Scholar] [CrossRef]
  52. Rooney, M.R.; Fang, M.; Ogurtsova, K.; Ozkan, B.; Echouffo-Tcheugui, J.B.; Boyko, E.J.; Magliano, D.J.; Selvin, E. Global Prevalence of Prediabetes. Diabetes Care 2023, 46, 1388–1394. [Google Scholar] [CrossRef] [PubMed]
  53. Blaak, E.E.; Antoine, J.-M.; Benton, D.; Björck, I.; Bozzetto, L.; Brouns, F.; Diamant, M.; Dye, L.; Hulshof, T.; Holst, J.J.; et al. Impact of Postprandial Glycaemia on Health and Prevention of Disease. Obes. Rev. 2012, 13, 923–984. [Google Scholar] [CrossRef] [PubMed]
  54. Zakharova, I.; Klimov, L.; Kuryaninova, V.; Nikitina, I.; Malyavskaya, S.; Dolbnya, S.; Kasyanova, A.; Atanesyan, R.; Stoyan, M.; Todieva, A.; et al. Vitamin D Insufficiency in Overweight and Obese Children and Adolescents. Front. Endocrinol. 2019, 10, 103. [Google Scholar] [CrossRef]
  55. Kabadi, S.M.; Lee, B.K.; Liu, L. Joint Effects of Obesity and Vitamin D Insufficiency on Insulin Resistance and Type 2 Diabetes. Diabetes Care 2012, 35, 2048–2054. [Google Scholar] [CrossRef] [PubMed]
  56. Gagnon, C.; Lu, Z.X.; Magliano, D.J.; Dunstan, D.W.; Shaw, J.E.; Zimmet, P.Z.; Sikaris, K.; Grantham, N.; Ebeling, P.R.; Daly, R.M. Serum 25-Hydroxyvitamin D, Calcium Intake, and Risk of Type 2 Diabetes after 5 Years: Results from a National, Population-Based Prospective Study (the Australian Diabetes, Obesity and Lifestyle Study). Diabetes Care 2011, 34, 1133–1138. [Google Scholar] [CrossRef]
  57. Mirhosseini, N.; Vatanparast, H.; Mazidi, M.; Kimball, S.M. Vitamin D Supplementation, Glycemic Control, and Insulin Resistance in Prediabetics: A Meta-Analysis. J. Endocr. Soc. 2018, 2, 687–709. [Google Scholar] [CrossRef]
  58. Borissova, A.M.; Tankova, T.; Kirilov, G.; Dakovska, L.; Kovacheva, R. The Effect of Vitamin D3 on Insulin Secretion and Peripheral Insulin Sensitivity in Type 2 Diabetic Patients. Int. J. Clin. Pract. 2003, 57, 258–261. [Google Scholar] [CrossRef]
  59. Fadda, G.Z.; Akmal, M.; Lipson, L.G.; Massry, S.G. Direct Effect of Parathyroid Hormone on Insulin Secretion from Pancreatic Islets. Am. J. Physiol. 1990, 258, E975–E984. [Google Scholar] [CrossRef]
  60. Sherman, M.H.; Yu, R.T.; Engle, D.D.; Ding, N.; Atkins, A.R.; Tiriac, H.; Collisson, E.A.; Connor, F.; Van Dyke, T.; Kozlov, S.; et al. Vitamin D Receptor-Mediated Stromal Reprogramming Suppresses Pancreatitis and Enhances Pancreatic Cancer Therapy. Cell 2014, 159, 80–93. [Google Scholar] [CrossRef]
  61. 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]
  62. Cigolini, M.; Iagulli, M.P.; Miconi, V.; Galiotto, M.; Lombardi, S.; Targher, G. Serum 25-Hydroxyvitamin D3 Concentrations and Prevalence of Cardiovascular Disease among Type 2 Diabetic Patients. Diabetes Care 2006, 29, 722–724. [Google Scholar] [CrossRef] [PubMed]
  63. Scragg, R.; Sowers, M.; Bell, C. Serum 25-Hydroxyvitamin D, Diabetes, and Ethnicity in the Third National Health and Nutrition Examination Survey. Diabetes Care 2004, 27, 2813–2818. [Google Scholar] [CrossRef] [PubMed]
  64. Di Cesar, D.J.; Ploutz-Snyder, R.; Weinstock, R.S.; Moses, A.M. Vitamin D Deficiency Is More Common in Type 2 than in Type 1 Diabetes. Diabetes Care 2006, 29, 174. [Google Scholar] [CrossRef]
  65. UK Prospective Diabetes Study (UKPDS) Group. Intensive Blood-Glucose Control with Sulphonylureas or Insulin Compared with Conventional Treatment and Risk of Complications in Patients with Type 2 Diabetes (UKPDS 33). Lancet 1998, 352, 837–853. [Google Scholar] [CrossRef]
  66. Hu, Z.; Chen, J.; Sun, X.; Wang, L.; Wang, A. Efficacy of Vitamin D Supplementation on Glycemic Control in Type 2 Diabetes Patients: A Meta-Analysis of Interventional Studies. Medicine 2019, 98, e14970. [Google Scholar] [CrossRef] [PubMed]
  67. Mitri, J.; Dawson-Hughes, B.; Hu, F.B.; Pittas, A.G. Effects of Vitamin D and Calcium Supplementation on Pancreatic β Cell Function, Insulin Sensitivity, and Glycemia in Adults at High Risk of Diabetes: The Calcium and Vitamin D for Diabetes Mellitus (CaDDM) Randomized Controlled Trial. Am. J. Clin. Nutr. 2011, 94, 486–494. [Google Scholar] [CrossRef] [PubMed]
  68. Nazarian, S.; St Peter, J.V.; Boston, R.C.; Jones, S.A.; Mariash, C.N. Vitamin D3 Supplementation Improves Insulin Sensitivity in Subjects with Impaired Fasting Glucose. Transl. Res. 2011, 158, 276–281. [Google Scholar] [CrossRef] [PubMed]
  69. Pramono, A.; Jocken, J.W.E.; Blaak, E.E. Vitamin D Deficiency in the Aetiology of Obesity-Related Insulin Resistance. Diabetes Metab. Res. Rev. 2019, 35, e3146. [Google Scholar] [CrossRef]
  70. Alvarez, J.A.; Ashraf, A. Role of Vitamin D in Insulin Secretion and Insulin Sensitivity for Glucose Homeostasis. Int. J. Endocrinol. 2010, 2010, 351385. [Google Scholar] [CrossRef]
  71. Larrick, B.M.; Kim, K.-H.; Donkin, S.S.; Teegarden, D. 1,25-Dihydroxyvitamin D Regulates Lipid Metabolism and Glucose Utilization in Differentiated 3T3-L1 Adipocytes. Nutr. Res. 2018, 58, 72–83. [Google Scholar] [CrossRef]
  72. Karkeni, E.; Bonnet, L.; Marcotorchino, J.; Tourniaire, F.; Astier, J.; Ye, J.; Landrier, J.-F. Vitamin D Limits Inflammation-Linked MicroRNA Expression in Adipocytes In Vitro and In Vivo: A New Mechanism for the Regulation of Inflammation by Vitamin D. Epigenetics 2018, 13, 156–162. [Google Scholar] [CrossRef]
  73. Altieri, B.; Grant, W.B.; Della Casa, S.; Orio, F.; Pontecorvi, A.; Colao, A.; Sarno, G.; Muscogiuri, G. Vitamin D and Pancreas: The Role of Sunshine Vitamin in the Pathogenesis of Diabetes Mellitus and Pancreatic Cancer. Crit. Rev. Food Sci. Nutr. 2017, 57, 3472–3488. [Google Scholar] [CrossRef] [PubMed]
  74. Cade, C.; Norman, A.W. Vitamin D3 Improves Impaired Glucose Tolerance and Insulin Secretion in the Vitamin D-Deficient Rat In Vivo. Endocrinology 1986, 119, 84–90. [Google Scholar] [CrossRef] [PubMed]
  75. Zeitz, U.; Weber, K.; Soegiarto, D.W.; Wolf, E.; Balling, R.; Erben, R.G. Impaired Insulin Secretory Capacity in Mice Lacking a Functional Vitamin D Receptor. FASEB J. 2003, 17, 509–511. [Google Scholar] [CrossRef] [PubMed]
  76. Bouillon, R.; Carmeliet, G.; Verlinden, L.; van Etten, E.; Verstuyf, A.; Luderer, H.F.; Lieben, L.; Mathieu, C.; Demay, M. Vitamin D and Human Health: Lessons from Vitamin D Receptor Null Mice. Endocr. Rev. 2008, 29, 726–776. [Google Scholar] [CrossRef] [PubMed]
  77. Maestro, B.; Campión, J.; Dávila, N.; Calle, C. Stimulation by 1,25-Dihydroxyvitamin D3 of Insulin Receptor Expression and Insulin Responsiveness for Glucose Transport in U-937 Human Promonocytic Cells. Endocr. J. 2000, 47, 383–391. [Google Scholar] [CrossRef] [PubMed]
  78. Maestro, B.; Molero, S.; Bajo, S.; Dávila, N.; Calle, C. Transcriptional Activation of the Human Insulin Receptor Gene by 1,25-Dihydroxyvitamin D3. Cell Biochem. Funct. 2002, 20, 227–232. [Google Scholar] [CrossRef]
  79. Pittas, A.G.; Harris, S.S.; Stark, P.C.; Dawson-Hughes, B. The Effects of Calcium and Vitamin D Supplementation on Blood Glucose and Markers of Inflammation in Nondiabetic Adults. Diabetes Care 2007, 30, 980–986. [Google Scholar] [CrossRef]
  80. Bland, R.; Markovic, D.; Hills, C.E.; Hughes, S.V.; Chan, S.L.F.; Squires, P.E.; Hewison, M. Expression of 25-Hydroxyvitamin D3-1alpha-Hydroxylase in Pancreatic Islets. J. Steroid Biochem. Mol. Biol. 2004, 89–90, 121–125. [Google Scholar] [CrossRef]
  81. Reusch, J.E.; Begum, N.; Sussman, K.E.; Draznin, B. Regulation of GLUT-4 Phosphorylation by Intracellular Calcium in Adipocytes. Endocrinology 1991, 129, 3269–3273. [Google Scholar] [CrossRef]
  82. Ryan, Z.C.; Craig, T.A.; Folmes, C.D.; Wang, X.; Lanza, I.R.; Schaible, N.S.; Salisbury, J.L.; Nair, K.S.; Terzic, A.; Sieck, G.C.; et al. 1α,25-Dihydroxyvitamin D3 Regulates Mitochondrial Oxygen Consumption and Dynamics in Human Skeletal Muscle Cells. J. Biol. Chem. 2016, 291, 1514–1528. [Google Scholar] [CrossRef] [PubMed]
  83. Baron, A.D.; Brechtel, G.; Wallace, P.; Edelman, S.V. Rates and Tissue Sites of Non-Insulin- and Insulin-Mediated Glucose Uptake in Humans. Am. J. Physiol. 1988, 255, E769–E774. [Google Scholar] [CrossRef] [PubMed]
  84. DeFronzo, R.A.; Jacot, E.; Jequier, E.; Maeder, E.; Wahren, J.; Felber, J.P. The Effect of Insulin on the Disposal of Intravenous Glucose. Results from Indirect Calorimetry and Hepatic and Femoral Venous Catheterization. Diabetes 1981, 30, 1000–1007. [Google Scholar] [CrossRef]
  85. Jefferson, G.E.; Schnell, D.M.; Thomas, D.T.; Bollinger, L.M. Calcitriol Concomitantly Enhances Insulin Sensitivity and Alters Myocellular Lipid Partitioning in High Fat-Treated Skeletal Muscle Cells. J. Physiol. Biochem. 2017, 73, 613–621. [Google Scholar] [CrossRef] [PubMed]
  86. Krul-Poel, Y.H.M.; Ter Wee, M.M.; Lips, P.; Simsek, S. Management of endocrine disease: The Effect of Vitamin D Supplementation on Glycaemic Control in Patients with Type 2 Diabetes Mellitus: A Systematic Review and Meta-Analysis. Eur. J. Endocrinol. 2017, 176, R1–R14. [Google Scholar] [CrossRef] [PubMed]
  87. Earthman, C.P.; Beckman, L.M.; Masodkar, K.; Sibley, S.D. The Link between Obesity and Low Circulating 25-Hydroxyvitamin D Concentrations: Considerations and Implications. Int. J. Obes. 2012, 36, 387–396. [Google Scholar] [CrossRef]
  88. Bajaj, S.; Singh, R.P.; Dwivedi, N.C.; Singh, K.; Gupta, A.; Mathur, M. Vitamin D Levels and Microvascular Complications in Type 2 Diabetes. Indian J. Endocrinol. Metab. 2014, 18, 537–541. [Google Scholar] [CrossRef]
  89. Zhao, J.; Dong, J.; Wang, H.; Shang, H.; Zhang, D.; Liao, L. Efficacy and Safety of Vitamin D3 in Patients with Diabetic Nephropathy: A Meta-Analysis of Randomized Controlled Trials. Chin. Med. J. 2014, 127, 2837–2843. [Google Scholar]
  90. Shehab, D.; Al-Jarallah, K.; Mojiminiyi, O.A.; Al Mohamedy, H.; Abdella, N.A. Does Vitamin D Deficiency Play a Role in Peripheral Neuropathy in Type 2 Diabetes? Diabet. Med. 2012, 29, 43–49. [Google Scholar] [CrossRef]
  91. Assy, M.H.; Draz, N.A.; Fathy, S.E.; Hamed, M.G. Impact of Vitamin D Level in Diabetic People with Peripheral Neuropathy. Egypt. J. Neurol. Psychiatry Neurosurg. 2021, 57, 117. [Google Scholar] [CrossRef]
  92. Al-Shoumer, K.A.; Al-Essa, T.M. Is There a Relationship between Vitamin D with Insulin Resistance and Diabetes Mellitus? World J. Diabetes 2015, 6, 1057–1064. [Google Scholar] [CrossRef]
  93. Mohd Saffian, S.; Jamil, N.A.; Mohd Tahir, N.A.; Hatah, E. Vitamin D Insufficiency Is High in Malaysia: A Systematic Review and Meta-Analysis of Studies on Vitamin D Status in Malaysia. Front. Nutr. 2022, 9, 1050745. [Google Scholar] [CrossRef]
  94. Ding, Y.-H.; Wei, T.-M.; Qian, L.-Y.; Ma, Y.; Lao, D.-B.; Yao, B.; Pang, J. Association between Serum 25-Hydroxyvitamin D and Carotid Atherosclerotic Plaque in Chinese Type 2 Diabetic Patients. Medicine 2017, 96, e6445. [Google Scholar] [CrossRef]
  95. De Boer, I.H.; Kestenbaum, B.; Shoben, A.B.; Michos, E.D.; Sarnak, M.J.; Siscovick, D.S. 25-hydroxyvitamin D levels inversely associate with risk for developing coronary artery calcification. J. Am. Soc. Nephrol. 2009, 20, 1805–1812. [Google Scholar] [CrossRef]
  96. Zehnder, D.; Bland, R.; Chana, R.S.; Wheeler, D.C.; Howie, A.J.; Williams, M.C.; Stewart, P.M.; Hewison, M. Synthesis of 1,25-dihydroxyvitamin D3 by human endothelial cells is regulated by inflammatory cytokines: A novel autocrine determinant of vascular cell adhesion. J. Am. Soc. Nephrol. 2002, 13, 621–629. [Google Scholar] [CrossRef]
  97. Wong, M.S.; Delansorne, R.; Man, R.Y.; Vanhoutte, P.M. Vitamin D derivatives acutely reduce endothelium-dependent contractions in the aorta of the spontaneously hypertensive rat. Am. J. Physiol. Heart Circ. Physiol. 2008, 295, H289–H296. [Google Scholar] [CrossRef]
  98. Richart, T.; Li, Y.; Staessen, J.A. Renal versus extrarenal activation of vitamin D in relation to atherosclerosis, arterial stiffening, and hypertension. Am. J. Hypertens. 2007, 20, 1007–1015. [Google Scholar] [CrossRef]
  99. Karadeniz, Y.; Özpamuk-Karadeniz, F.; Ahbab, S.; Ataoğlu, E.; Can, G. Vitamin D Deficiency Is a Potential Risk for Blood Pressure Elevation and the Development of Hypertension. Medicina 2021, 57, 1297. [Google Scholar] [CrossRef]
  100. McMullan, C.J.; Borgi, L.; Curhan, G.C.; Fisher, N.; Forman, J.P. The effect of vitamin D on renin-angiotensin system activation and blood pressure: A randomized control trial. J. Hypertens. 2017, 35, 822–829. [Google Scholar] [CrossRef]
  101. Tomaschitz, A.; Pilz, S.; Ritz, E.; Grammer, T.; Drechsler, C.; Boehm, B.O.; März, W. Independent association between 1,25-dihydroxyvitamin D, 25-hydroxyvitamin D and the renin-angiotensin system: The Ludwigshafen Risk and Cardiovascular Health (LURIC) study. Clin. Chim. Acta 2010, 411, 1354–1360. [Google Scholar] [CrossRef]
  102. 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]
  103. Zhou, C.; Lu, F.; Cao, K.; Xu, D.; Goltzman, D.; Miao, D. Calcium-independent and 1,25(OH)2D3-dependent regulation of the renin-angiotensin system in 1alpha-hydroxylase knockout mice. Kidney Int. 2008, 74, 170–179. [Google Scholar] [CrossRef]
  104. 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]
  105. Jorde, R.; Figenschau, Y.; Emaus, N.; Hutchinson, M.; Grimnes, G. Serum 25-hydroxyvitamin D levels are strongly related to systolic blood pressure but do not predict future hypertension. Hypertension 2010, 55, 792–798. [Google Scholar] [CrossRef]
  106. Paschou, S.A.; Papadopoulou-Marketou, N.; Chrousos, G.P.; Kanaka-Gantenbein, C. On Type 1 Diabetes Mellitus Pathogenesis. Endocr. Connect. 2018, 7, R38–R46. [Google Scholar] [CrossRef]
  107. Soltesz, G.; Patterson, C.C.; Dahlquist, G. EURODIAB Study Group Worldwide Childhood Type 1 Diabetes Incidence—What Can We Learn from Epidemiology? Pediatr. Diabetes 2007, 8 (Suppl. S6), 6–14. [Google Scholar] [CrossRef]
  108. Cernea, S.; Dobreanu, M.; Raz, I. Prevention of Type 1 Diabetes: Today and Tomorrow. Diabetes Metab. Res. Rev. 2010, 26, 602–605. [Google Scholar] [CrossRef]
  109. Huber, A.; Menconi, F.; Corathers, S.; Jacobson, E.M.; Tomer, Y. Joint Genetic Susceptibility to Type 1 Diabetes and Autoimmune Thyroiditis: From Epidemiology to Mechanisms. Endocr. Rev. 2008, 29, 697–725. [Google Scholar] [CrossRef]
  110. Mathieu, C.; Badenhoop, K. Vitamin D and Type 1 Diabetes Mellitus: State of the Art. Trends Endocrinol. Metab. 2005, 16, 261–266. [Google Scholar] [CrossRef]
  111. Takiishi, T.; Gysemans, C.; Bouillon, R.; Mathieu, C. Vitamin D and Diabetes. Endocrinol. Metab. Clin. N. Am. 2010, 39, 419–446. [Google Scholar] [CrossRef]
  112. Storm, T.L.; Sørensen, O.H.; Lund, B.; Lund, B.; Christiansen, J.S.; Andersen, A.R.; Lumholtz, I.B.; Parving, H.H. Vitamin D Metabolism in Insulin-Dependent Diabetes Mellitus. Metab. Bone Dis. Relat. Res. 1983, 5, 107–110. [Google Scholar] [CrossRef]
  113. Luong, K.V.Q.; Nguyen, L.T.H.; Nguyen, D.N.P. The Role of Vitamin D in Protecting Type 1 Diabetes Mellitus. Diabetes Metab. Res. Rev. 2005, 21, 338–346. [Google Scholar] [CrossRef]
  114. Danescu, L.G.; Levy, S.; Levy, J. Vitamin D and Diabetes Mellitus. Endocrine 2009, 35, 11–17. [Google Scholar] [CrossRef]
  115. Yang, C.-Y.; Leung, P.S.C.; Adamopoulos, I.E.; Gershwin, M.E. The Implication of Vitamin D and Autoimmunity: A Comprehensive Review. Clin. Rev. Allergy Immunol. 2013, 45, 217–226. [Google Scholar] [CrossRef]
  116. Norris, J.M.; Johnson, R.K.; Stene, L.C. Type 1 Diabetes-Early Life Origins and Changing Epidemiology. Lancet Diabetes Endocrinol. 2020, 8, 226–238. [Google Scholar] [CrossRef]
  117. Jiang, X.; Kiel, D.P.; Kraft, P. The Genetics of Vitamin D. Bone 2019, 126, 59–77. [Google Scholar] [CrossRef]
  118. Holick, M.F. Vitamin D Deficiency. N. Engl. J. Med. 2007, 357, 266–281. [Google Scholar] [CrossRef]
  119. Bikle, D.D. Vitamin D Metabolism, Mechanism of Action, and Clinical Applications. Chem. Biol. 2014, 21, 319–329. [Google Scholar] [CrossRef]
  120. Dankers, W.; Colin, E.M.; van Hamburg, J.P.; Lubberts, E. Vitamin D in Autoimmunity: Molecular Mechanisms and Therapeutic Potential. Front. Immunol. 2016, 7, 697. [Google Scholar] [CrossRef]
  121. Penna, G.; Adorini, L. 1 Alpha,25-Dihydroxyvitamin D3 Inhibits Differentiation, Maturation, Activation, and Survival of Dendritic Cells Leading to Impaired Alloreactive T Cell Activation. J. Immunol. 2000, 164, 2405–2411. [Google Scholar] [CrossRef]
  122. Zhang, X.; Zhou, M.; Guo, Y.; Song, Z.; Liu, B. 1,25-Dihydroxyvitamin D₃ Promotes High Glucose-Induced M1 Macrophage Switching to M2 via the VDR-PPARγ Signaling Pathway. Biomed. Res. Int. 2015, 2015, 157834. [Google Scholar] [CrossRef]
  123. Casteels, K.M.; Mathieu, C.; Waer, M.; Valckx, D.; Overbergh, L.; Laureys, J.M.; Bouillon, R. Prevention of Type I Diabetes in Nonobese Diabetic Mice by Late Intervention with Nonhypercalcemic Analogs of 1,25-Dihydroxyvitamin D3 in Combination with a Short Induction Course of Cyclosporin A. Endocrinology 1998, 139, 95–102. [Google Scholar] [CrossRef] [PubMed]
  124. Mathieu, C.; Waer, M.; Laureys, J.; Rutgeerts, O.; Bouillon, R. Prevention of Autoimmune Diabetes in NOD Mice by 1,25 Dihydroxyvitamin D3. Diabetologia 1994, 37, 552–558. [Google Scholar] [CrossRef]
  125. Zella, J.B.; McCary, L.C.; DeLuca, H.F. Oral Administration of 1,25-Dihydroxyvitamin D3 Completely Protects NOD Mice from Insulin-Dependent Diabetes Mellitus. Arch. Biochem. Biophys. 2003, 417, 77–80. [Google Scholar] [CrossRef]
  126. Fronczak, C.M.; Barón, A.E.; Chase, H.P.; Ross, C.; Brady, H.L.; Hoffman, M.; Eisenbarth, G.S.; Rewers, M.; Norris, J.M. In Utero Dietary Exposures and Risk of Islet Autoimmunity in Children. Diabetes Care 2003, 26, 3237–3242. [Google Scholar] [CrossRef] [PubMed]
  127. Corrao, S.; Colomba, D.; Arnone, S.; Argano, C.; Di Chiara, T.; Scaglione, R.; Licata, G. Improving Efficacy of PubMed Clinical Queries for Retrieving Scientifically Strong Studies on Treatment. J. Am. Med. Inform. Assoc. 2006, 13, 485–487. [Google Scholar] [CrossRef]
  128. Corrao, S.; Colomba, D.; Argano, C.; Calvo, L.; Scaglione, R.; Licata, G. Optimized Search Strategy for Detecting Scientifically Strong Studies on Treatment through PubMed. Intern. Emerg. Med. 2012, 7, 283–287. [Google Scholar] [CrossRef]
  129. Hou, Y.; Song, A.; Jin, Y.; Xia, Q.; Song, G.; Xing, X. A Dose–Response Meta-Analysis between Serum Concentration of 25-Hydroxy Vitamin D and Risk of Type 1 Diabetes Mellitus. Eur. J. Clin. Nutr. 2021, 75, 1010–1023. [Google Scholar] [CrossRef]
  130. Bener, A.; Alsaied, A.; Al-Ali, M.; Al-Kubaisi, A.; Basha, B.; Abraham, A.; Guiter, G.; Mian, M. High Prevalence of Vitamin D Deficiency in Type 1 Diabetes Mellitus and Healthy Children. Acta Diabetol. 2009, 46, 183–189. [Google Scholar] [CrossRef]
  131. Omar, D.F.; Kamal, M.M.; El-Hefnawy, M.H.; El-Mesallamy, H.O. Serum Vitamin D and Its Upregulated Protein, Thioredoxin Interacting Protein, Are Associated With Beta-Cell Dysfunction in Adult Patients With Type 1 and Type 2 Diabetes. Can. J. Diabetes 2018, 42, 588–594. [Google Scholar] [CrossRef]
  132. ALkharashi, N.A. Estimation of Vitamin D Deficiency Prevalence among Saudi Children in Armed Forces Hospital and Riyadh Care Hospital in Riyadh, Kingdom of Saudi Arabia and Its Relation to Type 1 Diabetes Mellitus. Saudi Med. J. 2019, 40, 1290–1293. [Google Scholar] [CrossRef] [PubMed]
  133. Hyppönen, E.; Läärä, E.; Reunanen, A.; Järvelin, M.R.; Virtanen, S.M. Intake of Vitamin D and Risk of Type 1 Diabetes: A Birth-Cohort Study. Lancet 2001, 358, 1500–1503. [Google Scholar] [CrossRef] [PubMed]
  134. Norris, J.M.; Lee, H.-S.; Frederiksen, B.; Erlund, I.; Uusitalo, U.; Yang, J.; Lernmark, Å.; Simell, O.; Toppari, J.; Rewers, M.; et al. Plasma 25-Hydroxyvitamin D Concentration and Risk of Islet Autoimmunity. Diabetes 2018, 67, 146–154. [Google Scholar] [CrossRef] [PubMed]
  135. The EURODIAB Substudy 2 Study Group. Vitamin D Supplement in Early Childhood and Risk for Type I (Insulin-Dependent) Diabetes Mellitus. Diabetologia 1999, 42, 51–54. [Google Scholar] [CrossRef]
  136. Wei, Z.; Yoshihara, E.; He, N.; Hah, N.; Fan, W.; Pinto, A.F.M.; Huddy, T.; Wang, Y.; Ross, B.; Estepa, G.; et al. Vitamin D Switches BAF Complexes to Protect β Cells. Cell 2018, 173, 1135–1149.e15. [Google Scholar] [CrossRef] [PubMed]
  137. Yu, J.; Sharma, P.; Girgis, C.M.; Gunton, J.E. Vitamin D and Beta Cells in Type 1 Diabetes: A Systematic Review. Int. J. Mol. Sci. 2022, 23, 14434. [Google Scholar] [CrossRef]
  138. Gregoriou, E.; Mamais, I.; Tzanetakou, I.; Lavranos, G.; Chrysostomou, S. The Effects of Vitamin D Supplementation in Newly Diagnosed Type 1 Diabetes Patients: Systematic Review of Randomized Controlled Trials. Rev. Diabet. Stud. 2017, 14, 260–268. [Google Scholar] [CrossRef]
  139. Nascimento, B.F.; Moreira, C.F.F.; da Fonseca, E.R.; Fedeszen, P.M.K.; de Paula, T.P.; de Sena, A.S.S.; de Almeida, N.F.A.; Bandeira Filho, O.C.S.; Curval, D.R.; Padilha, P.C. Effects of vitamin D supplementation on glycemic control of children and adolescents with type 1 diabetes mellitus: A systematic review. J. Pediatr. Endocrinol. Metab. 2022, 35, 973–988. [Google Scholar] [CrossRef]
  140. Najjar, L.; Sutherland, J.; Zhou, A.; Hyppönen, E. Vitamin D and Type 1 Diabetes Risk: A Systematic Review and Meta-Analysis of Genetic Evidence. Nutrients 2021, 13, 4260. [Google Scholar] [CrossRef]
  141. von Websky, K.; Hasan, A.A.; Reichetzeder, C.; Tsuprykov, O.; Hocher, B. Impact of Vitamin D on Pregnancy-Related Disorders and on Offspring Outcome. J. Steroid Biochem. Mol. Biol. 2018, 180, 51–64. [Google Scholar] [CrossRef]
  142. American Diabetes Association 2. Classification and Diagnosis of Diabetes. Diabetes Care 2017, 40, S11–S24. [Google Scholar] [CrossRef] [PubMed]
  143. Jenum, A.K.; Mørkrid, K.; Sletner, L.; Vangen, S.; Torper, J.L.; Nakstad, B.; Voldner, N.; Rognerud-Jensen, O.H.; Berntsen, S.; Mosdøl, A.; et al. Impact of Ethnicity on Gestational Diabetes Identified with the WHO and the Modified International Association of Diabetes and Pregnancy Study Groups Criteria: A Population-Based Cohort Study. Eur. J. Endocrinol. 2012, 166, 317–324. [Google Scholar] [CrossRef] [PubMed]
  144. Noctor, E.; Crowe, C.; Carmody, L.A.; Kirwan, B.; O’Dea, A.; Glynn, L.G.; McGuire, B.E.; O’Shea, P.M.; Dunne, F.P. ATLANTIC-DIP: Prevalence of Metabolic Syndrome and Insulin Resistance in Women with Previous Gestational Diabetes Mellitus by International Association of Diabetes in Pregnancy Study Groups Criteria. Acta Diabetol. 2015, 52, 153–160. [Google Scholar] [CrossRef] [PubMed]
  145. Milajerdi, A.; Tehrani, H.; Haghighatdoost, F.; Larijani, B.; Surkan, P.J.; Azadbakht, L. Associations between Higher Egg Consumption during Pregnancy with Lowered Risks of High Blood Pressure and Gestational Diabetes Mellitus. Int. J. Vitam. Nutr. Res. 2018, 88, 166–175. [Google Scholar] [CrossRef] [PubMed]
  146. Groof, Z.; Garashi, G.; Husain, H.; Owayed, S.; AlBader, S.; Mouhsen, H.; Mohammad, A.; Ziyab, A.H. Prevalence, Risk Factors, and Fetomaternal Outcomes of Gestational Diabetes Mellitus in Kuwait: A Cross-Sectional Study. J. Diabetes Res. 2019, 2019, e9136250. [Google Scholar] [CrossRef]
  147. Verma, A.; Boney, C.M.; Tucker, R.; Vohr, B.R. Insulin Resistance Syndrome in Women with Prior History of Gestational Diabetes Mellitus. J. Clin. Endocrinol. Metab. 2002, 87, 3227–3235. [Google Scholar] [CrossRef]
  148. Andersson-Hall, U.; Gustavsson, C.; Pedersen, A.; Malmodin, D.; Joelsson, L.; Holmäng, A. Higher Concentrations of BCAAs and 3-HIB Are Associated with Insulin Resistance in the Transition from Gestational Diabetes to Type 2 Diabetes. J. Diabetes Res. 2018, 2018, 4207067. [Google Scholar] [CrossRef]
  149. Nutrition Therapy in Gestational Diabetes Mellitus: Time to Move Forward | Diabetes Care | American Diabetes Association. Available online: https://diabetesjournals.org/care/article/41/7/1343/36441/Nutrition-Therapy-in-Gestational-Diabetes-Mellitus (accessed on 23 August 2023).
  150. Ismail, N.A.; Mohamed Ismail, N.A.; Bador, K.M. Vitamin D in Gestational Diabetes Mellitus and Its Association with Hyperglycaemia, Insulin Sensitivity and Other Factors. J. Obstet. Gynaecol. 2021, 41, 899–903. [Google Scholar] [CrossRef]
  151. Magnusdottir, K.S.; Tryggvadottir, E.A.; Magnusdottir, O.K.; Hrolfsdottir, L.; Halldorsson, T.I.; Birgisdottir, B.E.; Hreidarsdottir, I.T.; Hardardottir, H.; Gunnarsdottir, I. Vitamin D Status and Association with Gestational Diabetes Mellitus in a Pregnant Cohort in Iceland. Food Nutr. Res. 2021, 65, 5574. [Google Scholar] [CrossRef]
  152. Agüero-Domenech, N.; Jover, S.; Sarrión, A.; Baranda, J.; Quesada-Rico, J.A.; Pereira-Expósito, A.; Gil-Guillén, V.; Cortés-Castell, E.; García-Teruel, M.J. Vitamin D Deficiency and Gestational Diabetes Mellitus in Relation to Body Mass Index. Nutrients 2021, 14, 102. [Google Scholar] [CrossRef]
  153. Khambule, L.; George, J.A. The Role of Inflammation in the Development of GDM and the Use of Markers of Inflammation in GDM Screening. Adv. Exp. Med. Biol. 2019, 1134, 217–242. [Google Scholar] [CrossRef] [PubMed]
  154. Wolf, M.; Sauk, J.; Shah, A.; Vossen Smirnakis, K.; Jimenez-Kimble, R.; Ecker, J.L.; Thadhani, R. Inflammation and Glucose Intolerance: A Prospective Study of Gestational Diabetes Mellitus. Diabetes Care 2004, 27, 21–27. [Google Scholar] [CrossRef] [PubMed]
  155. López-Tinoco, C.; Roca, M.; García-Valero, A.; Murri, M.; Tinahones, F.J.; Segundo, C.; Bartha, J.L.; Aguilar-Diosdado, M. Oxidative Stress and Antioxidant Status in Patients with Late-Onset Gestational Diabetes Mellitus. Acta Diabetol. 2013, 50, 201–208. [Google Scholar] [CrossRef] [PubMed]
  156. Shang, M.; Zhao, J.; Yang, L.; Lin, L. Oxidative Stress and Antioxidant Status in Women with Gestational Diabetes Mellitus Diagnosed by IADPSG Criteria. Diabetes Res. Clin. Pract. 2015, 109, 404–410. [Google Scholar] [CrossRef]
  157. Peuchant, E.; Brun, J.-L.; Rigalleau, V.; Dubourg, L.; Thomas, M.-J.; Daniel, J.-Y.; Leng, J.-J.; Gin, H. Oxidative and Antioxidative Status in Pregnant Women with Either Gestational or Type 1 Diabetes. Clin. Biochem. 2004, 37, 293–298. [Google Scholar] [CrossRef]
  158. Grissa, O.; Atègbo, J.-M.; Yessoufou, A.; Tabka, Z.; Miled, A.; Jerbi, M.; Dramane, K.L.; Moutairou, K.; Prost, J.; Hichami, A.; et al. Antioxidant Status and Circulating Lipids Are Altered in Human Gestational Diabetes and Macrosomia. Transl. Res. 2007, 150, 164–171. [Google Scholar] [CrossRef]
  159. Maged, A.M.; Torky, H.; Fouad, M.A.; GadAllah, S.H.; Waked, N.M.; Gayed, A.S.; Salem, A.K. Role of Antioxidants in Gestational Diabetes Mellitus and Relation to Fetal Outcome: A Randomized Controlled Trial. J. Matern. Fetal Neonatal Med. 2016, 29, 4049–4054. [Google Scholar] [CrossRef]
  160. Haidari, F.; Zakerkish, M.; Karandish, M.; Saki, A.; Pooraziz, S. Association between Serum Vitamin D Level and Glycemic and Inflammatory Markers in Non-Obese Patients with Type 2 Diabetes. Iran. J. Med. Sci. 2016, 41, 367–373. [Google Scholar]
  161. 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]
  162. Berridge, M.J. Vitamin D Deficiency and Diabetes. Biochem. J. 2017, 474, 1321–1332. [Google Scholar] [CrossRef]
  163. Johannesson, B.; Sui, L.; Freytes, D.O.; Creusot, R.J.; Egli, D. Toward Beta Cell Replacement for Diabetes. EMBO J. 2015, 34, 841–855. [Google Scholar] [CrossRef] [PubMed]
  164. Taheri, E.; Saedisomeolia, A.; Djalali, M.; Qorbani, M.; Madani Civi, M. The Relationship between Serum 25-Hydroxy Vitamin D Concentration and Obesity in Type 2 Diabetic Patients and Healthy Subjects. J. Diabetes Metab. Disord. 2012, 11, 16. [Google Scholar] [CrossRef] [PubMed]
  165. Milajerdi, A.; Abbasi, F.; Mousavi, S.M.; Esmaillzadeh, A. Maternal Vitamin D Status and Risk of Gestational Diabetes Mellitus: A Systematic Review and Meta-Analysis of Prospective Cohort Studies. Clin. Nutr. 2021, 40, 2576–2586. [Google Scholar] [CrossRef] [PubMed]
  166. Wu, C.; Song, Y.; Wang, X. Vitamin D Supplementation for the Outcomes of Patients with Gestational Diabetes Mellitus and Neonates: A Meta-Analysis and Systematic Review. Int. J. Clin. Pract. 2023, 2023, 1907222. [Google Scholar] [CrossRef] [PubMed]
  167. Akbari, M.; Moosazaheh, M.; Lankarani, K.B.; Tabrizi, R.; Samimi, M.; Karamali, M.; Jamilian, M.; Kolahdooz, F.; Asemi, Z. The Effects of Vitamin D Supplementation on Glucose Metabolism and Lipid Profiles in Patients with Gestational Diabetes: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Horm. Metab. Res. 2017, 49, 647–653. [Google Scholar] [CrossRef]
  168. Wang, H.; Xia, N.; Yang, Y.; Peng, D.-Q. Influence of Vitamin D Supplementation on Plasma Lipid Profiles: A Meta-Analysis of Randomized Controlled Trials. Lipids Health Dis. 2012, 11, 42. [Google Scholar] [CrossRef]
  169. Jahanjoo, F.; Farshbaf-Khalili, A.; Shakouri, S.K.; Dolatkhah, N. Maternal and Neonatal Metabolic Outcomes of Vitamin D Supplementation in Gestational Diabetes Mellitus: A Systematic Review and Meta-Analysis. Ann. Nutr. Metab. 2018, 73, 145–159. [Google Scholar] [CrossRef]
  170. Kron-Rodrigues, M.R.; Rudge, M.V.C.; Lima, S.A.M. Supplementation of Vitamin D in the Postdelivery Period of Women with Previous Gestational Diabetes Mellitus: Systematic Review and Meta-Analysis of Randomized Trials. Rev. Bras. Ginecol. Obstet. 2021, 43, 699–709. [Google Scholar] [CrossRef]
  171. Saha, S.; Saha, S. Changes in Anthropometric and Blood 25-Hydroxyvitamin D Measurements in Antenatal Vitamin Supplemented Gestational Diabetes Mellitus Patients: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. J. Turk. Ger. Gynecol. Assoc. 2021, 22, 217–234. [Google Scholar] [CrossRef]
  172. Wang, M.M.; Chen, Z.J.; Wang, Y.; Xu, X.R.; Li, H.J.; Yang, J. Effects of Vitamin D Supplementation on Serum Lipid Profiles and Neonatal Outcomes in Gestational Diabetes Mellitus:a Meta-analysis. Zhongguo Yi Xue Ke Xue Yuan Xue Bao 2021, 43, 82–91. [Google Scholar] [CrossRef]
  173. Yang, C.; Jing, W.; Ge, S.; Sun, W. Vitamin D Status and Vitamin D Deficiency Risk Factors among Pregnancy of Shanghai in China. BMC Pregnancy Childbirth 2021, 21, 431. [Google Scholar] [CrossRef] [PubMed]
  174. Chatzakis, C.; Sotiriadis, A.; Tsakmaki, E.; Papagianni, M.; Paltoglou, G.; Dinas, K.; Mastorakos, G. The Effect of Dietary Supplements on Oxidative Stress in Pregnant Women with Gestational Diabetes Mellitus: A Network Meta-Analysis. Nutrients 2021, 13, 2284. [Google Scholar] [CrossRef] [PubMed]
  175. Wang, L.; Zhang, C.; Song, Y.; Zhang, Z. Serum Vitamin D Deficiency and Risk of Gestational Diabetes Mellitus: A Meta-Analysis. Arch. Med. Sci. 2020, 16, 742–751. [Google Scholar] [CrossRef] [PubMed]
  176. Jin, S.; Sha, L.; Dong, J.; Yi, J.; Liu, Y.; Guo, Z.; Hu, B. Effects of Nutritional Strategies on Glucose Homeostasis in Gestational Diabetes Mellitus: A Systematic Review and Network Meta-Analysis. J. Diabetes Res. 2020, 2020, 6062478. [Google Scholar] [CrossRef]
  177. Soheilykhah, S.; Mojibian, M.; Rashidi, M.; Rahimi-Saghand, S.; Jafari, F. Maternal Vitamin D Status in Gestational Diabetes Mellitus. Nutr. Clin. Pract. 2010, 25, 524–527. [Google Scholar] [CrossRef]
  178. Zhang, Y.; Gong, Y.; Xue, H.; Xiong, J.; Cheng, G. Vitamin D and Gestational Diabetes Mellitus: A Systematic Review Based on Data Free of Hawthorne Effect. BJOG Int. J. Obstet. Gynaecol. 2018, 125, 784–793. Available online: https://obgyn.onlinelibrary.wiley.com/doi/abs/10.1111/1471-0528.15060 (accessed on 23 August 2023). [CrossRef]
  179. Vaidya, A.; Williams, J.S. Vitamin D and Insulin Sensitivity: Can Gene Association and Pharmacogenetic Studies of the Vitamin D Receptor Provide Clarity? Metabolism 2012, 61, 759–761. [Google Scholar] [CrossRef]
  180. Lu, M.; Xu, Y.; Lv, L.; Zhang, M. Association between Vitamin D Status and the Risk of Gestational Diabetes Mellitus: A Meta-Analysis. Arch. Gynecol. Obstet. 2016, 293, 959–966. [Google Scholar] [CrossRef]
  181. Chen, Y.; Zhu, B.; Wu, X.; Li, S.; Tao, F. Association between Maternal Vitamin D Deficiency and Small for Gestational Age: Evidence from a Meta-Analysis of Prospective Cohort Studies. BMJ Open 2017, 7, e016404. [Google Scholar] [CrossRef]
  182. Licata, G.; Argano, C.; Di Chiara, T.; Parrinello, G.; Scaglione, R. Obesity: A main factor of metabolic syndrome? Panminerva Med. 2006, 48, 77–85. [Google Scholar]
  183. Fahed, G.; Aoun, L.; Bou Zerdan, M.; Allam, S.; Bou Zerdan, M.; Bouferraa, Y.; Assi, H.I. Metabolic Syndrome: Updates on Pathophysiology and Management in 2021. Int. J. Mol. Sci. 2022, 23, 786. [Google Scholar] [CrossRef]
  184. Classification and Diagnosis of Diabetes: Standards of Medical Care in Diabetes—2022 | Diabetes Care | American Diabetes Association. Available online: https://diabetesjournals.org/care/article/45/Supplement_1/S17/138925/2-Classification-and-Diagnosis-of-Diabetes (accessed on 23 August 2023).
  185. Wang, H.; Naghavi, M.; Allen, C.; Barber, R.M.; Bhutta, Z.A.; Carter, A.; Casey, D.C.; Charlson, F.J.; Chen, A.Z.; Coates, M.M.; et al. Global, Regional, and National Life Expectancy, All-Cause Mortality, and Cause-Specific Mortality for 249 Causes of Death, 1980–2015: A Systematic Analysis for the Global Burden of Disease Study 2015. Lancet 2016, 388, 1459–1544. [Google Scholar] [CrossRef]
  186. Zhu, Y.; Xian, X.; Wang, Z.; Bi, Y.; Chen, Q.; Han, X.; Tang, D.; Chen, R. Research Progress on the Relationship between Atherosclerosis and Inflammation. Biomolecules 2018, 8, 80. [Google Scholar] [CrossRef] [PubMed]
  187. Hojs, R.; Ekart, R.; Bevc, S.; Hojs, N. Markers of Inflammation and Oxidative Stress in the Development and Progression of Renal Disease in Diabetic Patients. Nephron 2016, 133, 159–162. [Google Scholar] [CrossRef]
  188. Flaim, C.; Kob, M.; Di Pierro, A.M.; Herrmann, M.; Lucchin, L. Effects of a Whey Protein Supplementation on Oxidative Stress, Body Composition and Glucose Metabolism among Overweight People Affected by Diabetes Mellitus or Impaired Fasting Glucose: A Pilot Study. J. Nutr. Biochem. 2017, 50, 95–102. [Google Scholar] [CrossRef]
  189. Mannarino, E.; Pirro, M. Molecular Biology of Atherosclerosis. Clin. Cases Miner. Bone Metab. 2008, 5, 57–62. [Google Scholar]
  190. Hussin, A.M.; Ashor, A.W.; Schoenmakers, I.; Hill, T.; Mathers, J.C.; Siervo, M. Effects of Vitamin D Supplementation on Endothelial Function: A Systematic Review and Meta-Analysis of Randomised Clinical Trials. Eur. J. Nutr. 2017, 56, 1095–1104. [Google Scholar] [CrossRef] [PubMed]
  191. Jacobson, T.A.; Maki, K.C.; Orringer, C.E.; Jones, P.H.; Kris-Etherton, P.; Sikand, G.; La Forge, R.; Daniels, S.R.; Wilson, D.P.; Morris, P.B.; et al. National Lipid Association Recommendations for Patient-Centered Management of Dyslipidemia: Part 2. J. Clin. Lipidol. 2015, 9, S1–S122.e1. [Google Scholar] [CrossRef]
  192. Cholesterol Treatment Trialists’ (CTT) Collaboration; Fulcher, J.; O’Connell, R.; Voysey, M.; Emberson, J.; Blackwell, L.; Mihaylova, B.; Simes, J.; Collins, R.; Kirby, A.; et al. Efficacy and Safety of LDL-Lowering Therapy among Men and Women: Meta-Analysis of Individual Data from 174,000 Participants in 27 Randomised Trials. Lancet 2015, 385, 1397–1405. [Google Scholar] [CrossRef]
  193. Bovolini, A.; Garcia, J.; Andrade, M.A.; Duarte, J.A. Metabolic Syndrome Pathophysiology and Predisposing Factors. Int. J. Sports Med. 2021, 42, 199–214. [Google Scholar] [CrossRef]
  194. Castro-Barquero, S.; Ruiz-León, A.M.; Sierra-Pérez, M.; Estruch, R.; Casas, R. Dietary Strategies for Metabolic Syndrome: A Comprehensive Review. Nutrients 2020, 12, 2983. [Google Scholar] [CrossRef] [PubMed]
  195. Pan, G.-T.; Guo, J.-F.; Mei, S.-L.; Zhang, M.-X.; Hu, Z.-Y.; Zhong, C.-K.; Zeng, C.-Y.; Liu, X.-H.; Ma, Q.-H.; Li, B.-Y.; et al. Vitamin D Deficiency in Relation to the Risk of Metabolic Syndrome in Middle-Aged and Elderly Patients with Type 2 Diabetes Mellitus. J. Nutr. Sci. Vitaminol. 2016, 62, 213–219. [Google Scholar] [CrossRef]
  196. Faraji, S.; Alizadeh, M. Mechanistic Effects of Vitamin D Supplementation on Metabolic Syndrome Components in Patients with or without Vitamin D Deficiency. J. Obes. Metab. Syndr. 2020, 29, 270–280. [Google Scholar] [CrossRef] [PubMed]
  197. Sepidarkish, M.; Farsi, F.; Akbari-Fakhrabadi, M.; Namazi, N.; Almasi-Hashiani, A.; Maleki Hagiagha, A.; 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] [PubMed]
  198. Jain, S.K.; Micinski, D. Vitamin D Upregulates Glutamate Cysteine Ligase and Glutathione Reductase, and GSH Formation, and Decreases ROS and MCP-1 and IL-8 Secretion in High-Glucose Exposed U937 Monocytes. Biochem. Biophys. Res. Commun. 2013, 437, 7–11. [Google Scholar] [CrossRef]
  199. Brandenburg, V.M.; Vervloet, M.G.; Marx, N. The Role of Vitamin D in Cardiovascular Disease: From Present Evidence to Future Perspectives. Atherosclerosis 2012, 225, 253–263. [Google Scholar] [CrossRef]
  200. Siti, H.N.; Kamisah, Y.; Kamsiah, J. The Role of Oxidative Stress, Antioxidants and Vascular Inflammation in Cardiovascular Disease (a Review). Vascul. Pharmacol. 2015, 71, 40–56. [Google Scholar] [CrossRef]
  201. Sies, H. Total Antioxidant Capacity: Appraisal of a Concept. J. Nutr. 2007, 137, 1493–1495. [Google Scholar] [CrossRef]
  202. Namazi, N.; Larijani, B.; Azadbakht, L. Alpha-Lipoic Acid Supplement in Obesity Treatment: A Systematic Review and Meta-Analysis of Clinical Trials. Clin. Nutr. 2018, 37, 419–428. [Google Scholar] [CrossRef]
  203. Heshmati, J.; Farsi, F.; Shokri, F.; Rezaeinejad, M.; Almasi-Hashiani, A.; Vesali, S.; Sepidarkish, M. A Systematic Review and Meta-Analysis of the Probiotics and Synbiotics Effects on Oxidative Stress. J. Funct. Foods 2018, 46, 66–84. [Google Scholar] [CrossRef]
  204. Slominski, A.T.; Kim, T.-K.; Hobrath, J.V.; Janjetovic, Z.; Oak, A.S.W.; Postlethwaite, A.; Lin, Z.; Li, W.; Takeda, Y.; Jetten, A.M.; et al. Characterization of a New Pathway That Activates Lumisterol in Vivo to Biologically Active Hydroxylumisterols. Sci. Rep. 2017, 7, 11434. [Google Scholar] [CrossRef] [PubMed]
  205. Mahmood, S.F.; Idiculla, J.; Joshi, R.; Joshi, S.; Kulkarni, S. Vitamin D Supplementation in Adults with Vitamin D Deficiency and Its Effect on Metabolic Syndrome—A Randomized Controlled Study. Int. J. Vitam. Nutr. Res. 2016, 86, 121–126. [Google Scholar] [CrossRef] [PubMed]
  206. Teixeira, J.S.; Bull Ferreira Campos, A.; Cordeiro, A.; Pereira, S.E.; Saboya, C.J.; Ramalho, A. Vitamin D Nutritional Status and Its Relationship with Metabolic Changes in Adolescents and Adults with Severe Obesity. Nutr. Hosp. 2018, 35, 847–853. [Google Scholar] [CrossRef] [PubMed]
  207. Ferreira, P.P.; Cangussu, L.; Bueloni-Dias, F.N.; Orsatti, C.L.; Schmitt, E.B.; Nahas-Neto, J.; Nahas, E.A.P. Vitamin D Supplementation Improves the Metabolic Syndrome Risk Profile in Postmenopausal Women. Climacteric 2020, 23, 24–31. [Google Scholar] [CrossRef]
  208. Xu, Y.; Zhou, Y.; Liu, J.; Wang, C.; Qu, Z.; Wei, Z.; Zhou, D. Genetically Increased Circulating 25(OH)D Level Reduces the Risk of Type 2 Diabetes in Subjects with Deficiency of Vitamin D. Medicine 2020, 99, e23672. [Google Scholar] [CrossRef]
  209. Pilz, S.; Tomaschitz, A.; Ritz, E.; Pieber, T.R. Vitamin D status and arterial hypertension: A systematic review. Nat. Rev. Cardiol. 2009, 6, 621–630. [Google Scholar] [CrossRef]
  210. Shi, H.; Norman, A.W.; Okamura, W.H.; Sen, A.; Zemel, M.B. 1a,25-Dihydroxyvitamin D3 modulates human adipocyte metabolism via nongenomic action. FASEB J. 2001, 15, 2751–2753. [Google Scholar] [CrossRef]
  211. Clarke, N.M.P.; Page, J.E. Vitamin D deficiency. Curr. Opin. Pediatr. 2012, 24, 46–49. [Google Scholar] [CrossRef]
  212. Vimaleswaran, K.S.; Cavadino, A.; Berry, D.J.; Jorde, R.; Dieffenbach, A.K.; Lu, C.; Alves, A.C.; Heerspink, H.J.L.; Tikkanen, E.; Eriksson, J.; et al. Association of vitamin D status with arterial blood pressure and hypertension risk: A mendelian randomisation study. Lancet Diabetes Endocrinol. 2014, 2, 719–729. [Google Scholar] [CrossRef]
  213. Rosen, C.J.; Adams, J.S.; Bikle, D.D.; Black, D.M.; Demay, M.B.; Manson, J.E.; Murad, M.H.; Kovacs, C.S. The nonskeletal effects of vitamin D: An endocrine society scientific statement. Endocr. Rev. 2012, 33, 456–492. [Google Scholar] [CrossRef]
  214. Zhu, W.; Heil, D.P. Associations of Vitamin D Status with Markers of Metabolic Health: A Community-Based Study in Shanghai, China. Diabetes Metab. Syndr. 2018, 12, 727–732. [Google Scholar] [CrossRef] [PubMed]
  215. Jafari, T.; Fallah, A.A.; Barani, A. Effects of Vitamin D on Serum Lipid Profile in Patients with Type 2 Diabetes: A Meta-Analysis of Randomized Controlled Trials. Clin. Nutr. 2016, 35, 1259–1268. [Google Scholar] [CrossRef] [PubMed]
  216. Li, X.; Liu, Y.; Zheng, Y.; Wang, P.; Zhang, Y. The Effect of Vitamin D Supplementation on Glycemic Control in Type 2 Diabetes Patients: A Systematic Review and Meta-Analysis. Nutrients 2018, 10, 375. [Google Scholar] [CrossRef] [PubMed]
  217. 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]
  218. Ganji, V.; Sukik, A.; Alaayesh, H.; Rasoulinejad, H.; Shraim, M. Serum Vitamin D Concentrations Are Inversely Related to Prevalence of Metabolic Syndrome in Qatari Women. Biofactors 2020, 46, 180–186. [Google Scholar] [CrossRef] [PubMed]
  219. Wang, N.; Wang, C.; Chen, X.; Wan, H.; Chen, Y.; Chen, C.; Han, B.; Lu, Y. Vitamin D, Prediabetes and Type 2 Diabetes: Bidirectional Mendelian Randomization Analysis. Eur. J. Nutr. 2020, 59, 1379–1388. [Google Scholar] [CrossRef]
  220. Zheng, J.-S.; Luan, J.; Sofianopoulou, E.; Sharp, S.J.; Day, F.R.; Imamura, F.; Gundersen, T.E.; Lotta, L.A.; Sluijs, I.; Stewart, I.D.; et al. The Association between Circulating 25-Hydroxyvitamin D Metabolites and Type 2 Diabetes in European Populations: A Meta-Analysis and Mendelian Randomisation Analysis. PLoS Med. 2020, 17, e1003394. [Google Scholar] [CrossRef]
  221. Ostadmohammadi, V.; Milajerdi, A.; Ghayour-Mobarhan, M.; Ferns, G.; Taghizadeh, M.; Badehnoosh, B.; Mirzaei, H.; Asemi, Z. The Effects of Vitamin D Supplementation on Glycemic Control, Lipid Profiles and C-Reactive Protein Among Patients with Cardiovascular Disease: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Curr. Pharm. Des. 2019, 25, 201–210. [Google Scholar] [CrossRef]
  222. Qi, K.-J.; Zhao, Z.-T.; Zhang, W.; Yang, F. The Impacts of Vitamin D Supplementation in Adults with Metabolic Syndrome: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Front. Pharmacol. 2022, 13, 1033026. [Google Scholar] [CrossRef]
  223. De Paula, T.P.; Kramer, C.K.; Viana, L.V.; Azevedo, M.J. Effects of Individual Micronutrients on Blood Pressure in Patients with Type 2 Diabetes: A Systematic Review and Meta-Analysis of Randomized Clinical Trials. Sci. Rep. 2017, 7, 40751. [Google Scholar] [CrossRef]
  224. McGreevy, C.; Williams, D. New Insights about Vitamin D and Cardiovascular Disease: A Narrative Review. Ann. Intern. Med. 2011, 155, 820–826. [Google Scholar] [CrossRef] [PubMed]
  225. Grossman, E. Ambulatory Blood Pressure Monitoring in the Diagnosis and Management of Hypertension. Diabetes Care 2013, 36 (Suppl. S2), S307–S311. [Google Scholar] [CrossRef] [PubMed]
  226. Hajhashemy, Z.; Shahdadian, F.; Moslemi, E.; Mirenayat, F.S.; Saneei, P. Serum Vitamin D Levels in Relation to Metabolic Syndrome: A Systematic Review and Dose-Response Meta-Analysis of Epidemiologic Studies. Obes. Rev. 2021, 22, e13223. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The pleiotropic effect of vitamin D.
Figure 1. The pleiotropic effect of vitamin D.
Ijms 24 15485 g001
Figure 2. Vitamin D metabolism.
Figure 2. Vitamin D metabolism.
Ijms 24 15485 g002
Table 1. Synthesis of meta-analyses and systematic reviews on the effectiveness of various dosages of vitamin D administration in pathologic conditions analyzed.
Table 1. Synthesis of meta-analyses and systematic reviews on the effectiveness of various dosages of vitamin D administration in pathologic conditions analyzed.
Author/YearDesignDuration Participants
(I/C)
Dose of
Vitamin D
Results
VITAMIN D AND INSULIN RESISTANCE (IR)
Asbaghi et al.,
2019 [46]
MT (12 RCTs)From 6 to 312 weeks8946 healthy subjects
or patients with overweight/obesity, IFG, pre-diabetes, GDM, T2DM, PCOS, HIV infection
(4395/4551)
From 200 IU/day Vitamin D3 to 50,000 IU/week Vitamin D3 (with supplementation dose of calcium that ranged from 500 mg/day to 1000 mg/day)Reduce effects on FBG, circulating levels of insulin, and HOMA-IR
Sindhughosa et al., 2022 [41]MT (7 RCTs)From 10 to 52 weeks735 patients with NAFLD (423/312)From 1000 IU/day Vitamin D3 to 50,000 IU/week Vitamin D3 Improvement in IR (marked by a decrease in HOMA-IR), decrease in ALT levels
Pienkowska et al.,
2023 [29]
SR (8 RCTs)From 12 to 260 weeksFrom 66 to 2423
patients with prediabetes
From 1000 IU/day Vitamin D3 to 88,000 IU/week Vitamin D3 Only one trial showed improvements in FBG and HOMA-IR
VITAMIN D AND TYPE 2 DIABETES MELLITUS (T2DM)
Pittas et al.,
2007 [61]
MT (13 case–control studies; 15 cross-sectional studies; 12 RCTs)N/APatients with T2DM
or prediabetes
2000 IU/day Vitamin D3 or
Vitamin D3 700 IU/day with supplementation dose of 500 mg/day calcium citrate
Vitamin D and calcium insufficiency may negatively influence glycemia, whereas combined supplementation with both nutrients may be beneficial in optimizing glucose metabolism
Krul-Poel et al.,
2017 [86]
MT (23 RCTs)From 4 to 52 weeks1797 patients with T2DM: for the effect
on HbA1c 1475 patients (755/720), for the effect on FBG 1180 patients (608/572)
From 1000 IU/day Vitamin D3 to 45,000 IU/week Vitamina D3 or 11,200 IU/day Vitamin D3 for 2 weeks followed by 5600 IU/day for 10 weeks or from 100,000 to 300,000 IU Vitamin D3 single doseSignificant effect on FBG in a subgroup of studies (n = 4); no significant effect in change in HbA1c
Mirhosseini et al., 2018 [57]MT (28 RCTs)From 8 to 260 weeks3848 healthy subjects or patients with prediabetes and/or overweight or obesity, NAFLD, arterial hypertension, cervical intraepithelial neoplasia, premenopausal and postmenopausal womenFrom 420 IU/day to 88,880 IU/week Vitamin D3Significant reduction in HbA1c, FBG, and HOMA-IR
Hu et al.,
2019 [66]
MT (19 RCTs)From 4 to 24 weeks1374 patients
with T2DM
(747/627)
Up to 50,000 UI/weekly Vitamin D3 or 300,000 UI single injection Vitamin D3 Significant
reduction in HbA1c,
IR (marked by a decrease in HOMA-IR)
and insulin levels in the short-term vitamin D supplementation group
VITAMIN D AND TYPE 1 DIABETES MELLITUS (T1DM)
Gregoriou et al.,
2017 [138]
MT (7 RCTs)From 4 to 52 weeks287 patients
with T1DM
Calcitriol 0.25 μg per day or on alternate days plus insulin
Alphacalcidole 0.5 μg daily plus insulin
Cholecalciferol 2000 IU per day plus insulin for 18 mo
Cholecalciferol 70 IU/kg body weight/day plus insulin
Vitamin D supplementation in the form of alphacalcidole and chole- calciferol appears to be beneficial in daily insulin dose (DID), fasting C- peptide (FCP), stimulated C-peptide (SCP), and HbA1c.
Najjar et al.,
2021 [140]
MT (10 studies:
3 cohort;
5 case–control;
2 matched case–control)
N/A39,884 patients
with T1DM
(16,370/23,514)
N/ANo large effect of a genetically determined reduction in 25(OH)D concentrations by selected polymorphisms on T1D risk
Hou et al.,
2021 [129]
MT (16 studies:
12 case–control studies;
1 cross-sectional case–control study;
2 nested case–control study;
1 case–cohort study)
N/A10,605 patients
with T1DM
(3913/6692)
N/AResults demonstrated a significant inverse association between the 25(OH)D concentration in circulation and the risk of T1DM
Nascimento et al., 2022 [139]SR (10 studies)From 6 to 52 weeksChildren and adolescents (0–19 years) with T1DMCholecalciferol, with dosages ranging from 1000 to 160,000 IU. Just one study used vitamin D in the form of alfacalcidol at a dosage of 0.25 to 0.5 μg/dayThis study did not provide evidence to support the effect of vitamin D supplementation on glycemic control to aid in the treatment of T1DM
Yu et al.,
2022 [137]
SR (13 studies:
9 RCTs; 2 open-label case–control;
1 open label;
1 cohort)
From 4 to 12 weeks527 patients
with T1DM
The following therapeutic regimens were used:
1.25 D 0.25 μg 2nd daily; 25 D 2000 IU daily; 25 D to achieve serum 25 D > 125 nmol/L;
Alfacalcidol 0.25 μg bd 25 D; 60,000 IU monthly; Ergocalciferol (D2) 2 m of 50,000 IU/w; 25 D 2000 IU/d; 25D. 3000 IU/d; Calciferol 2000 IU/d + etanercept + GAD-alum
The maintenance of optimal circulating 25 D levels may reduce the risk of T1D and that may have potential for benefits in delaying the development of absolute or near-absolute C-peptide deficiency
VITAMIN D AND GESTATIONAL DIABETES MELLITUS (GDM)
Akbari et al.,
2017 [167]
MT (6 RCTs)From 6 to 12 weeks371 pregnant women with GDM (187/184)50,000 IU vitamin D3 2 times during the study or 50,000 IU vitamin D3 once every 2 weeks for 2 months, for a total of 200,000 IU vitamin D3 or 50,000 U vitamin D3 pearl twice during the study + 1000 mg calcium per day or 1000 IU vitamin D3 and 1000 mg evening primrose oil (EPO) or one intramuscular injection of 300,000 IU of vitamin D3 or a total of 700,000 IU vitamin D3 during pregnancyThis meta-analysis has demonstrated that vitamin D supplementation may lead to an improvement in HOMA-IR, QUICKI, and LDL-cholesterol levels but did not affect FPG, insulin, HbA1c, triglycerides, total-, and HDL-cholesterol levels; however, vitamin D supplementation increased HOMA-B.
Jahanjoo et al.,
2018 [169]
MT (5 RCTs)From 6 to 16 weeks310 women with GDM50,000 IU vitamin D3 2 times during the study or 200,000 IU vitamin D3 for each of the first 2 days, and then 50,000 IU per week thereafter, up to 700,000 IU in total. Those at week 28 of gestation or later were asked to take 100,000 IU weekly or 50,000 IU of vitamin D3 once every 2 weeksThis study showed that supplementation of GDM women with vitamin D may lead to an improvement in FPG, TC, LDL, HDL, and hs-CRP serum levels, as well as in newborns’ hyperbilirubinemia
Rodrigues et al.,
2019 [170]
MT (6 RCT studies)From 6 to 24 weeks and a study until delivery456 pregnant women with GDM diagnosed
in the second or third trimester of pregnancy
50,000 IU of vitamin D3 every 2 weeks
or 1000 UI daily
Improves adverse maternal and neonatal outcomes related to GDM
Milajerdi et al.,
2021 [145]
MT (29 studies: 18 cohort;
9 nested case–control; 1 prospective cross-sectional; 1 retrospective cohort)
N/A42,668 patients
with GDM or not
Blood vitamin D levelsThe lowest risk of GDM was found among those with serum vitamin D levels of 40 and 90 nmol/L
Wang et al.,
2021 [44]
MT (19 RCTs; of these, 13 concerned GDM)From 6 to 12 weeks1198 patients
with GDM
From 50,000 IU of vitamin D3 2 times/day to 1200
IU daily
The results showed that vitamin D supplementation during pregnancy could significantly reduce maternal cesarean section rate, maternal hospitalization rate, and postpartum hemorrhage in women with GDM
Chatzakis et al.,
2021 [174]
MT (15 studies:
9 cohort;
6 nestedcase–control)
N/A42,636 pregnant
women
(1848/40,788)
Blood vitamin D levelsThe result showed that lower levels of serum 25(OH)D were associated with a higher chance of GDM
Wu et al.,
2023 [166]
MT (20 RCT studies)From 2 to 16 weeks1682 pregnant women with GDM diagnosed
(837/845)
From 50,000 IU of vitamin D3 2 times/day to 1200
IU daily
Reduce serum LDL-C, TG, and TC levels and increase the serum HDL-C level. Reduce maternal and neonatal hyperbilirubinemia and hospitalization risk.
VITAMIN D, METABOLIC SYNDROME (MetS), AND CARDIOVASCULAR DISEASE (CVD)
De Paula TP et al.,
2017 [223]
MT (7 RCTs)From 3 to 52 weeks542 patients with
T2DM (472/70)
A single dose of vitamin D2 (100,000 IU) or vitamin D3 (100,000 IU or 200,000 IU)Reduction in BP, especially in systolic BP
Ostadmohammadi et al.,
2019 [221]
MT (8 RCTs) From 8 to 24 weeks630 adults with
CVD (305/325)
50,000 IU/week Vitamin D3 or
50,000 IU every two weeks or 300,000
IU single dose
Improving glycemic control, HDL-C, and CRP levels; it did not affect TG, TC, and LDL-C levels
Hajhashemy Z. et al.,
2021 [226]
Dose–response MT (43 epidemiological studies: 38 cross-sectional; 1 nested case
control; 4 cohort studies)
N/A309.206 adults
with or without
MetS
Blood Vitamin D levels in adultsInverse
association between serum vitamin D concentrations and risk of MetS
Qi K.J. et al.,
2022 [222]
MT (13 RCTs)From 8 to 24 weeks1.076 adults with
MetS (530/546)
From 1000 IU/day Vitamin D3 to 50,000 IU/weekDecreased BP, FPG, HOMA-IR, and CRP levels; it did not affect
HDL-C, LDL-C, TC, and TG levels
I/C: intervention/control; IR: insulin resistance; MT: meta-analysis; RCTs: randomized controlled trials; IFG: impaired fasting glucose; GDM: gestational diabetes mellitus; T2DM: type 2 diabetes mellitus; PCOS: polycystic ovary syndrome; FBG: fasting blood glucose; HOMA-IR: homeostatic model assessment of insulin resistance; NAFLD: non-alcoholic fatty liver disease; HbA1c: glycated hemoglobin; SR: systematic review; IR: insulin resistance; T1DM: type 1 diabetes mellitus; N/A: not applicable; MetS: metabolic syndrome; CVD: cardiovascular disease; BP: blood pressure; TG: triglycerides; TC: total cholesterol; LDL-C: LDL-cholesterol; HDL-C: HDL-cholesterol; CRP: C-reactive protein.
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

Argano, C.; Mirarchi, L.; Amodeo, S.; Orlando, V.; Torres, A.; Corrao, S. The Role of Vitamin D and Its Molecular Bases in Insulin Resistance, Diabetes, Metabolic Syndrome, and Cardiovascular Disease: State of the Art. Int. J. Mol. Sci. 2023, 24, 15485. https://doi.org/10.3390/ijms242015485

AMA Style

Argano C, Mirarchi L, Amodeo S, Orlando V, Torres A, Corrao S. The Role of Vitamin D and Its Molecular Bases in Insulin Resistance, Diabetes, Metabolic Syndrome, and Cardiovascular Disease: State of the Art. International Journal of Molecular Sciences. 2023; 24(20):15485. https://doi.org/10.3390/ijms242015485

Chicago/Turabian Style

Argano, Christiano, Luigi Mirarchi, Simona Amodeo, Valentina Orlando, Alessandra Torres, and Salvatore Corrao. 2023. "The Role of Vitamin D and Its Molecular Bases in Insulin Resistance, Diabetes, Metabolic Syndrome, and Cardiovascular Disease: State of the Art" International Journal of Molecular Sciences 24, no. 20: 15485. https://doi.org/10.3390/ijms242015485

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