Next Article in Journal
Assessment of Brainstem Functional Organization in Healthy Adults and Overactive Bladder Patients Using Ultra-High Field fMRI
Next Article in Special Issue
Oral Administration of Vitamin D3 Prevents Corneal Damage in a Knock-Out Mouse Model of Sjögren’s Syndrome
Previous Article in Journal
An Immunoinformatics Approach to Design Novel and Potent Multi-Epitope-Based Vaccine to Target Lumpy Skin Disease
Previous Article in Special Issue
A Mathematical Model for Determining the Body’s Fluctuating Need for and Synthesis of Active Vitamin D
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Genetic Variations of the Vitamin D Metabolic Pathway and COVID-19 Susceptibility and Severity: Current Understanding and Existing Evidence

by
Nipith Charoenngam
1,2,*,
Aunchalee Jaroenlapnopparat
1,
Sofia K. Mettler
1 and
Ashna Grover
1
1
Department of Medicine, Mount Auburn Hospital, Harvard Medical School, Cambridge, MA 02138, USA
2
Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 73170, Thailand
*
Author to whom correspondence should be addressed.
Biomedicines 2023, 11(2), 400; https://doi.org/10.3390/biomedicines11020400
Submission received: 4 January 2023 / Revised: 20 January 2023 / Accepted: 25 January 2023 / Published: 29 January 2023
(This article belongs to the Special Issue Recent Advances in Vitamin D)

Abstract

:
The immunomodulatory and metabolic effects of vitamin D receptor (VDR) activation have been considered beneficial in mitigating the susceptibility and severity of COVID-19 infection. Furthermore, vitamin D-binding protein (DBP) has pleiotropic effects on the immune system that may influence inflammation associated with COVID-19. Multiple observational studies have demonstrated an association between low levels of serum 25-hydroxyvitamin D and risk and the severity of COVID-19 infection. However, the impact of vitamin D supplementation as an adjunctive treatment for COVID-19 based on evidence from randomized clinical trials is unclear. Equally important is that certain variations of the genes involved in the vitamin D metabolic pathway have been shown to affect immune function and linked with various clinical outcomes, including cardio-metabolic disorders, autoimmune diseases, infections, and cancers. This indicates inter-individual difference in body response to vitamin D. There is also emerging evidence that common polymorphisms of these genes may influence the susceptibility and severity of COVID-19, although the confidence of these findings is limited by a small number of studies and participants. Further studies are needed to address the potential role of VDR activation and DBP in the pathophysiology of COVID-19 which take into account the genetic variations of vitamin D metabolic pathway.

1. Introduction

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a novel strain of coronavirus that causes coronavirus disease 2019 (COVID-19). It quickly spread worldwide resulting in a pandemic due to its high transmissibility and infectivity [1,2]. While most patients with COVID-19 develop mild respiratory symptoms, a significant proportion of patients develop severe illness resulting in multiple organ failure and death [3,4]. Factors influencing the risk of developing severe COVID-19 include advanced age, smoking, and the presence of underlying comorbidities such as immunocompromised state, cardio-metabolic disorders, chronic kidney disease, and chronic lung disease [5].
Multiple studies have suggested that vitamin D deficiency may be associated with increased susceptibility to COVID-19 infection and risks of severe illness [6,7,8,9,10,11]. This indicates the potential therapeutic and preventive role of vitamin D in COVID-19, which not only is responsible for calcium and phosphate homeostasis, but also has biological actions on various tissues that express the vitamin D receptor (VDR) [12,13,14]. Interestingly, there is mounting evidence that genetic variations of proteins involved in vitamin D metabolic pathway can affect tissue response to vitamin D and thus may influence the risks and severity of multiple chronic diseases, including COVID-19. In this article we dissect the complexity of the vitamin D metabolic pathway and outline the skeletal and non-skeletal actions of vitamin D. We review the relationship between vitamin D and COVID-19 infection. We comprehensively assess the recent evidence on genetic variations of vitamin D metabolic pathway in association with various clinical outcomes and COVID-19 and also introduce the concept of individual responsiveness to vitamin D. Finally, we discuss how certain genetic polymorphisms in vitamin D-related genes could impact immune function.

2. Sources, Synthesis, and Metabolism of Vitamin D

Humans obtain vitamin D mainly from diet, supplements, and sunlight exposure. Vitamin D2, or ergocalciferol, is derived from the ingestion of yeasts and mushrooms [13,14]. Vitamin D3, or cholecalciferol, is endogenously synthesized from 7-dehydrocholesterol in the skin that is exposed to the UVB radiation and exogenously derived from animal products (i.e., cod liver oil, oily fish). Both vitamin D2 and D3 also exist in the form of supplements and fortification in dietary products, such as milk, cooking oil, and orange juice [13,14].
Once vitamin D (D2 and D3) enters the circulation it is either distributed and stored in the muscle and adipose tissue or metabolized by the enzyme 25-hydroxylase (mainly encoded by the CYP2R1 gene) in the liver into 25-hydroxyvitamin D [25(OH)D]. 25(OH)D is the major circulating form of vitamin D clinically measured to reflect vitamin D status in the body. 25(OH)D is further metabolized in the kidney by the enzyme 1α-hydroxylase (encoded by the CYP27B1 gene) into 1,25-dihydroxyvitamin D [1,25(OH)2D], which is the biologically active form of vitamin D [13,14]. Both the circulating 25(OH)D and 1,25(OH)2D bind with the vitamin D binding protein (DBP, encoded by the GC gene, produced by the liver). For most cells, only the unbound 25(OH)D and 1,25(OH)2D enter the cells, but in some organs (i.e., kidney, parathyroid glands and placenta), DBP contributes to the transport of 25(OH)D into the cell via a megalin/cubilin complex [15].
1,25(OH)2D interacts with the intracellular nuclear VDR (encoded by the VDR gene) that forms a heterodimer complex with the retinoid X receptor (RXR) in the target cells. The activated VDR-RXR heterodimer complex, in turn, alters the expression of its target genes by interacting with the vitamin D-response elements (VDREs) in the target genes [16,17]. The 1,25(OH)2D-VDR-RXR-VDRE interaction also regulates 1,25(OH)2D level by inducing its destruction by upregulating the expression of the gene CYP24A1, encoding the enzyme 25-hydroxyvitamin D-24-hydroxylase that metabolizes 1,25(OH)2D and 25(OH)D into inactive carboxylic acids [18]. In addition, the enzyme 1α-hydroxylase, responsible for conversion of 25(OH)D into 1,25(OH)2D in the kidney is enhanced by the action of parathyroid hormone (PTH) and inhibited by the actions of 1,25(OH)2D and fibroblast growth factor-23 (FGF23) [13,14,19].
VDR is widely expressed in various types of tissues including the skin, skeletal muscles, adipose tissues, endocrine pancreas, innate and adaptive immune cells, blood vessels, brain, breast, and placenta [13,14]. Although the kidneys are the main site of production of circulating 1,25(OH)2D, CYP27B1 is expressed in many other types of cells, such as immune cells, parathyroid, breast, microglia, colon, and keratinocytes [6,20]. It is therefore speculated that 25(OH)D may be converted locally in these tissues to 1,25(OH)2D to exert its tissue-specific actions in a paracrine or autocrine manner. However, the fate of vitamin D metabolites in different target tissues needs more investigation. A schematic representation of the synthesis and metabolism of vitamin D is shown in Figure 1.

3. Skeletal and Non-Skeletal Effects of Vitamin D

Activation of VDR affects calcium and phosphate homeostasis mainly by promoting intestinal calcium and phosphate absorption and renal tubular calcium reabsorption. VDR activation increases bone resorption by inducing the receptor activator of nuclear factor kappa B (RANK)-mediated osteoclast differentiation and activation. In addition, 1,25(OH)2D directly inhibits PTH production and induces FGF23 production by osteocytes as a part of negative feedback loops to maintain serum calcium and phosphate concentrations within the physiologic ranges [13,14,19] (Figure 1).
Vitamin D is also known as an immunomodulatory agent that regulates the innate and adaptive immune systems [12,21,22,23]. Activated macrophages express the enzyme 1α-hydroxylase (CYP27B1) that metabolizes 25(OH)D into 1,25(OH)2D, a process which is not regulated by PTH, unlike in the kidneys. 1,25(OH)2D acts in a paracrine and autocrine manner to promote the production of the endogenous antimicrobial peptides (i.e., cathelicidins and defensins) by the macrophages [24,25,26]. 1,25(OH)2D affects the antigen presenting cells by inhibiting their differentiation, antigen presentation, and production of co-stimulatory molecules and inflammatory cytokines, and promoting the expression of inhibitory molecules. Moreover, activation of VDR has been demonstrated to modulate the activity and cytokine production of different types of T cells by facilitating differentiation of regulatory T cells (Treg) and promoting a shift from T helper 1 (TH1) and T helper 17 (TH17) to T helper 2 (TH2) immune profiles [22,27,28,29]. In addition, there is evidence showing that VDR is upregulated in response to mitogen activation of cytotoxic T lymphocytes and B cells, suggesting a coordinated regulation of the VDR signaling pathway and response to stimuli [30,31,32]. The effects of vitamin D on the immune system are summarized in Figure 2.
Besides its effects on calcium and phosphate homeostasis and immune function, vitamin D has been found to have biologic activities in multiple organ systems. To name a few, it plays a role in controlling glucose homeostasis by enhancing pancreatic β-cell function and improving insulin sensitivity [33,34]. It protects against vascular endothelial dysfunction by inducing nitric oxide production and maintaining endothelial stability [35,36]. It also has pro-differentiation and anti-proliferation effects on the keratinocytes and many types of cancer cells [37,38].
The broad range of theoretical health benefits of VDR activation has been supported by a multitude of studies that have showed associations between increased level of serum 25(OH)D concentration and decreased risks of infections, multiple chronic diseases, and all-cause mortality [39,40,41]. However, large-scale clinical trials have failed to demonstrate the benefit of supplementing vitamin D to the general population without known vitamin D deficiency in reducing risks of most chronic diseases, including diabetes, cardiovascular diseases, and cancer, except for a potential benefit in reducing the risks of autoimmune diseases [42,43,44,45]. While vitamin D supplementation is generally considered to raise the serum 25(OH)D level, increasing serum 25(OH)D does not necessarily reflect the increased level of circulating 1,25(OH)2D or increased VDR activation. This is based on the observation that circulating 1,25(OH)2D concentration can be normal or even high in individuals with vitamin D deficiency (defined by serum 25(OH)D < 20 ng/mL) due to the presence of secondary hyperparathyroidism that increases renal production of 1,25(OH)2D as a compensatory mechanism [19,46]. Therefore, the relationship between serum 25(OH)D, VDR activation, and health outcomes may be complex and requires further investigation.

4. Vitamin D and COVID-19 Infection

There are several proposed mechanisms by which vitamin D could reduce the risk and severity of COVID-19 infection. First, 1,25(OH)2D induces the macrophages and respiratory epithelial cells to produce antimicrobial peptides, including cathelicidin LL-37 that not only acts against invading bacteria and fungi by disrupting their cell membranes but also exhibits direct antiviral effects by altering viability of host target cells and destabilizing viral envelopes [47,48,49,50]. In addition, cathelicidins have been demonstrated to prevent lung injury secondary to oxygen toxicity [51]. This mechanism can be supported by the finding from a pilot study that showed increased serum cathelicidin in patients with sepsis who received a single enteral dose of 400,000 IUs of vitamin D3 compared to placebo [52]. Furthermore, a meta-analysis of 25 randomized controlled trials prior to the COVID era showed that vitamin D supplementation could reduce the risk of acute respiratory tract infection by approximately 12% compared to placebo [53].
The second mechanism involves the effects of VDR activation on the adaptive immune system. As discussed above, 1,25(OH)2D suppresses the activities of TH1 and TH17 and induces Treg differentiation. This action results in decreased production of proinflammatory cytokines (i.e., IL-6, IL-8, IL-12, IL-17, TNF-α), which is thought to mitigate the cytokine storm seen in severe COVID-19 infection [22,27,28,29]. These actions of vitamin D are supported by clinical studies demonstrating improvement in inflammatory burden in autoimmune diseases mediated by TH1 and TH17 (i.e., rheumatoid arthritis, multiple sclerosis, psoriasis, and inflammatory bowel disease) [12,54,55,56,57].
Third, vitamin D plays a role in controlling the renin-angiotensin-aldosterone system [58,59]. Activation of VDR has been shown to inhibit expression of the angiotensin-converting enzyme 2 (ACE2) protein in the renal tubular cells, a receptor to which SARS-CoV-2 virus can bind to, and thus may protect against acute kidney injury caused by direct cytopathic effect of the virus [59,60]. 1,25(OH)2D also inhibits renin and angiotensin-converting enzyme and induces the expression of ACE2 in the lungs. This reduces the angiotensin II accumulation that is thought to mitigate the development of acute respiratory distress syndrome and cardiovascular complications in COVID-19 [61]. Decreased renin expression may also alleviate bradykinin storm [62].
Other proposed explanations include but are not limited to vitamin D’s pleiotropic actions which help stabilize vascular endothelium, prevent vascular thrombosis, and inhibit fibrotic changes in the lungs [35,63,64,65]. It is worth noting that DBP also has independent biological functions, including macrophage activation, fatty acid transport, chemotaxis, and actin scavenging [15]. These could possibly play a role in pathophysiology of COVID-19. Elevated serum F-actin concentration has been observed in patients with acute respiratory distress syndrome and is believed to be a causative factor of pulmonary vascular angiopathy [66,67]. DBP is thought to mitigate this process by working in the actin scavenging system with gelsolin to clear extracellular actin released by damaged cells from the circulation [68].
These proposed explanations have been supported by multitudes of studies showing an association between low levels of serum 25(OH)D and risk of COVID-19 infection and mortality even after controlling for potential confounders such as age, body mass index, and medical comorbidities [6,7,8,9,10,11]. On the other hand, the benefits of raising serum 25(OH)D by giving vitamin D supplementation to COVID-19 patients have been disputed. Although the impact of vitamin D supplementation in reducing COVID-19 associated mortality and other secondary morbidity outcomes was observable in meta-analyses, most individual clinical trials have failed to clearly demonstrate the benefit [69]. Of note, there is mounting evidence that the administration of 25(OH)D (calcifediol) may be of particular benefit in reducing the severity and inflammatory burden of COVID-19 [70,71,72,73,74]. This is possibly due to the pharmacokinetic difference that 25(OH)D is more bioavailable and able to raise serum 25(OH)D concentration faster than orally administered native vitamin D [74,75,76,77].

5. Genetic Variations of Vitamin D Metabolic Pathway and Various Clinical Outcomes

Variation in the expression and activities of proteins involved in vitamin D metabolism can result in alterations in the kinetics and actions of vitamin D. Decreased enzymatic activity of the 7-dehydrocholesterol reductase, encoded by the DHCR7 gene, results in accumulation of the substrate 7-dehydrocholesterol, thereby increasing endogenous production of vitamin D3 [78]. There are four major hepatic 25-hydroxylases encoded by the genes CYP2R1, CYP27A1, CYP3A4, and CYP2J3, which determine the rate of conversion of vitamin D to 25(OH)D [79]. Homozygous inactivating mutations of CYP27B1 cause hereditary vitamin D-resistant rickets type 1 due to an inability to convert 25(OH)D into the active form 1,25(OH)2D. Homozygous inactivating mutations of VDR cause hereditary vitamin D-resistant rickets type 2 which manifests as rickets resistant to 1,25(OH)2D treatment and alopecia [80]. Loss-of-function mutations of CYP24A1 lead to inability to catabolize 25(OH)D and 1,25(OH)2D that results in phenotypes of hypercalcemia and hypercalciuria that predispose affected individuals to kidney stones and nephrocalcinosis [81]. While loss-of-function mutations of CYP27B1, CYP24A1, and VDR have been shown to significantly affect calcium homeostasis. Homozygous inactivating mutation of GC causing complete deficiency of DBP has been reported in a woman born to consanguineous parents who had debilitating ankylosing spondylitis, low bone mass, and a normal calcium level [82].
Furthermore, certain genetic polymorphisms of these vitamin D-related genes mentioned above (VDR, CYP2R1, CYP27B1, GC, CYP24A1, DHCR7) have been shown to be associated with risks of several diseases in populational and functional studies. These diseases include but are not limited to osteoporosis, asthma, cardio-metabolic diseases, neurodegenerative diseases, autoimmune diseases, nonalcoholic fatty liver disease, type 1 diabetes, viral infections, pancreatitis, and several types of cancers [83,84,85,86,87,88,89,90,91,92,93,94,95,96,97].
The large volume of data supports the link between genetics of vitamin D metabolic pathway and non-skeletal outcomes. While it is inevitable that some of these findings are subject to false positive results secondary to multiple testing and/or post-hoc testing, it may implicate that there is inter-individual difference in responsiveness to vitamin D in the general population. This concept of individual responsiveness to vitamin D can be supported by a clinical trial of 71 patients with prediabetes receiving 3,200 IUs/day of vitamin D3 for 5 months that revealed robust changes in broad gene expression in approximately 50% of the patients [98]. This finding concurs with the observation from another study revealing that approximately 60% of healthy adults with baseline serum 25(OH)D <30 ng/mL who received 10,000 IUs per day of vitamin D3 for 6 months had robust response in genome-wide expression in peripheral blood mononuclear cells, compared to the other 40% with mild to moderate response despite comparable increases in the serum concentrations of 25(OH)D to the range of 60–90 ng/mL [99]. In addition, the subjects with robust genomic response to vitamin D displayed different patterns of serum metabolomic profiles compared with those with mild to moderate response [99,100]. The specific genomic or epigenomic factors that can explain this inter-individual difference are, however, currently unknown.

6. Genetic Variations of Vitamin D Pathway and COVID-19 Infection

A number of observational studies have evaluated the association between vitamin D-related polymorphisms and risk of COVID-19 infection and severity [101,102,103,104,105,106,107,108,109,110,111,112,113,114,115,116]. As shown in Table 1, most studies are from European and Middle eastern countries and included a relatively small number of participants. Six retrospective cohort studies from Turkey, Iran, Portugal, United Arab Emirates, Serbia, and Cuba investigated the association between vitamin D-related polymorphisms and COVID-19 severity [102,103,104,105,106,107]. Four case-control studies from Italy, Iran, and Cyprus reported the association between VDR polymorphism and COVID-19 susceptibility [108,109,110,111], while one study from Iran reported the association between VDR polymorphism and COVID-19 mortality [112].
As shown in Table 2, VDR polymorphisms including FokI (rs2228570, exon 2, C > T), TaqI (rs731236, exon 9, A > G) and BsmI (rs1544410, intron 8, G > A) were shown to be associated with the risk of COVID-19 and severity in more than one observational study [102,103,108,109,110,111,112]. However, the data from these studies are largely conflicting and inconclusive. A polymorphism of the CYP2R1 gene (rs10741657, 5′UTR, G > A) was found to influence the severity of COVID-19 in two studies in the United Arab Emirates and Serbia [105,106]. An ecological study by Batur et al. [101] demonstrated that the prevalence of GT and TT genotypes of a polymorphism of the GC gene (rs7041, or BsuRI, exon 11, G > T) in each of the ten countries was associated with increased prevalence and decreased mortality rates among these countries. However, this finding was not confirmed in an observational study by Jafarpoor et al. [109]. Many other polymorphisms of the intronic and untranslated regions of the genes DHCR7, GC, and VDR were found to be associated with increased risk of critical COVID-19 disease in a study by Al-Anouti et al. [105]. However, in this study, a high throughput method was used for genotyping and the analysis was not adjusted for multiple testing; therefore, false positive results were very likely.
Taken together, there is slight evidence that polymorphisms of the VDR and GC genes may influence the risks of infectivity and severity of COVID-19, although the confidence of these findings is low due to the discrepancy in the results across the studies and the concern for false positivity in genomic research. Additional well-powered studies (such as genome-wide association studies) are needed to further elucidate the potential association between genetic variations in the vitamin D metabolic pathway and COVID-19.

7. Mendelian Randomization Studies of Vitamin D and COVID-19

Many Mendelian randomization studies utilized data from the UK biobank and the COVID-19 Host Genetics Initiative and generated models to predict serum 25(OH)D level or vitamin D deficiency (Table 1) [113,114,115,116]. These studies did not find any association between genetically predicted serum 25(OH)D level and COVID-19 risk. The models included variants of the genes CYP2R1, CYP24A1, DHCR7, and GC among several other SNPs [113,114,115,116]. While these studies point against the benefit of raising serum 25(OH)D level for COVID-19 risk and severity, the findings should be interpreted with caution as these models that included only genetic variants were not associated with unmeasured variables. It is known that environmental factors (i.e., vitamin D intake and UVB exposure) are also major determinants of vitamin D status in addition to genetics [117,118,119].

8. Functional Studies of Genetic Variations of the VDR and GC Genes

There are a few experimental studies that demonstrated functional significance of genetic variations of the VDR and GC genes. For instance, the FokI polymorphism results in VDR proteins with different structures: long f-VDR and short F-VDR. It has been well demonstrated in a transfection experimental study by van Etten et al. [120] that the shorter F-VDR resulted in higher NF-kB and NFAT-driven transcription and higher IL-12p40 promoter-driven transcription. Furthermore, homozygous short FF VDR genotype, compared with long ff VDR genotype, results in higher mRNA and protein expression of IL-12 in the macrophages and dendritic cells and stronger phytohemagglutinin-induced lymphocytic proliferation [120]. The CDX2 site, located in the promoter region of the VDR, is a binding site of the transcription factor caudal type homeobox-2 (CDX-2). CDX2 polymorphism affects the binding affinity of the CDX-2 transcription factor, VDR promoter methylation and thus VDR mRNA expression [121,122]. In an ex vivo study of peripheral blood mononuclear cell cultures of 51 patients with pulmonary tuberculosis and 60 healthy subjects, the CDX2 GG genotype and the combination baT haplotype of the 3′ untranslated region SNPs Apa I, Bsm I, and Taq I were associated with decreased TH1 response and increased IL-10 in response to culture filtrate antigen and 1,25(OH)2D [123].
Variations of the GC gene result in different isoforms of the DBP distinguished by electrophoresis. Among over 120 isoforms, there are three major isoforms that have been widely studied, namely Gc1f (rs7041 T and rs4588 C alleles), Gc1s (rs7041 G and rs4588 C alleles and Gc2 (rs7041 T and rs4588 A alleles). The Gc1s isoform has a higher affinity for 25(OH)D than the Gc1f isoform, with the Gc2 allele in between [15]. In addition, the Gc1f isoform carries the highest ability to be converted into the macrophage activating factor (MAF) by the sequential reactions of β-galactosidase and sialidase by T and B cells, respectively [124]. This explains the functional significance of these major GC polymorphisms (rs7041 and rs4588), which are shown to be associated with various clinical outcomes, including the risk of COVID-19.

9. Conclusions

The immunomodulatory and metabolic effects of vitamin D and VDR activation have been considered to have therapeutic and preventive potential in COVID-19 infection. Furthermore, DBP, encoded by the GC gene, is known to have independent immunological and actin-scavenging effects that may affect inflammation in COVID-19. Observational studies have shown that serum 25(OH)D concentration is inversely associated with the risk and severity of COVID-19. However, evidence from randomized clinical trials of vitamin D supplementation is weaker. This may suggest that raising serum 25(OH)D may not completely reflect increased activation of the intracellular VDR in the widespread tissues.
Another important aspect of the vitamin D-COVID-19 theory is that common polymorphisms in the VDR and GC genes have been shown to influence the susceptibility and severity of COVID-19. Although the strength of evidence is limited by a small number of studies and participants as well as possible false positivity, there is biological plausibility based on experimental studies that certain polymorphisms of the VDR and GC genes can affect immune function.
Therefore, it is possible that inter-individual differences in the function of VDR and GC based on their genetic variations may have confounded the results of the clinical trials that aimed to determine the impact vitamin D supplementation on COVID-19 and other clinical outcomes. Further investigations are warranted to address the potential role of VDR activation and DBP in pathophysiology of COVID-19 considering the genetic variations of the vitamin D metabolic pathway. In particular, VDR and GC polymorphisms should be taken into account as potential effect modifiers in studies aiming to determine causality of vitamin D and COVID-19-related outcomes.

Author Contributions

Conceptualization, N.C.; data curation, N.C. and A.J.; writing—original draft preparation, N.C.; writing—review and editing, N.C., S.K.M. and A.G.; visualization, N.C.; supervision, N.C. 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. Hu, B.; Guo, H.; Zhou, P.; Shi, Z.-L. Characteristics of SARS-CoV-2 and COVID-19. Nat. Rev. Microbiol. 2020, 19, 141–154. [Google Scholar] [CrossRef]
  2. The Lancet Infectious Diseases. COVID-19, a pandemic or not? Lancet Infect. Dis. 2020, 20, 383. [Google Scholar] [CrossRef]
  3. Kordzadeh-Kermani, E.; Khalili, H.; Karimzadeh, I. Pathogenesis, clinical manifestations and complications of coronavirus disease 2019 (COVID-19). Future Microbiol. 2020, 15, 1287–1305. [Google Scholar] [CrossRef]
  4. Wiersinga, W.J.; Rhodes, A.; Cheng, A.C.; Peacock, S.J.; Prescott, H.C. Pathophysiology, transmission, diagnosis, and treatment of coronavirus disease 2019 (COVID-19): A Review. JAMA 2020, 324, 782–793. [Google Scholar] [CrossRef]
  5. Wolff, D.; Nee, S.; Hickey, N.S.; Marschollek, M. Risk factors for COVID-19 severity and fatality: A structured literature review. Infection 2020, 49, 15–28. [Google Scholar] [CrossRef]
  6. Charoenngam, N.; Shirvani, A.; Holick, M.F. Vitamin D and its potential benefit for the COVID-19 pandemic. Endocr. Pract. 2021, 27, 484–493. [Google Scholar] [CrossRef]
  7. Kaya, M.O.; Pamukçu, E.; Yakar, B. The role of vitamin D deficiency on COVID-19: A systematic review and meta-analysis of observational studies. Epidemiol. Health 2021, 43, e2021074. [Google Scholar] [CrossRef]
  8. Ebrahimzadeh, A.; Mohseni, S.; Narimani, B.; Ebrahimzadeh, A.; Kazemi, S.; Keshavarz, F.; Yaghoubi, M.J.; Milajerdi, A. Association between vitamin D status and risk of COVID-19 in-hospital mortality: A systematic review and meta-analysis of observational studies. Crit. Rev. Food Sci. Nutr. 2021, 1–11. [Google Scholar] [CrossRef]
  9. Charoenngam, N.; Shirvani, A.; Reddy, N.; Vodopivec, D.M.; Apovian, C.M.; Holick, M.F. Association of vitamin D status with hospital morbidity and mortality in adult hospitalized patients with COVID-19. Endocr. Pract. 2021, 27, 271–278. [Google Scholar] [CrossRef]
  10. Chiodini, I.; Gatti, D.; Soranna, D.; Merlotti, D.; Mingiano, C.; Fassio, A.; Adami, G.; Falchetti, A.; Eller-Vainicher, C.; Rossini, M.; et al. Vitamin D status and SARS-CoV-2 infection and COVID-19 clinical outcomes. Front. Public Health 2021, 9, 736665. [Google Scholar] [CrossRef]
  11. Borsche, L.; Glauner, B.; von Mendel, J. COVID-19 mortality risk correlates inversely with vitamin D3 status, and a mortality rate close to zero could theoretically be achieved at 50 ng/mL 25(OH)D3: Results of a systematic review and meta-analysis. Nutrients 2021, 13, 3596. [Google Scholar] [CrossRef]
  12. Charoenngam, N.; Holick, M.F. Immunologic effects of vitamin D on human health and disease. Nutrients 2020, 12, 2097. [Google Scholar] [CrossRef]
  13. 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]
  14. Holick, M.F. Vitamin D deficiency. N. Engl. J. Med. 2007, 357, 266–281. [Google Scholar] [CrossRef]
  15. Bikle, D.D.; Schwartz, J. Vitamin D binding protein, total and free vitamin D levels in different physiological and pathophysiological conditions. Front. Endocrinol. 2019, 10, 317. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Pike, J.W.; Meyer, M.B. The vitamin D receptor: New paradigms for the regulation of gene expression by 1,25-dihydroxyvitamin D(3). Endocrinol. Metab. Clin. N. Am. 2010, 39, 255–269. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Hossein-Nezhad, A.; Spira, A.; Holick, M.F. Influence of vitamin D status and vitamin D3 supplementation on genome wide expression of white blood cells: A randomized double-blind clinical trial. PLoS ONE 2013, 8, e58725. [Google Scholar] [CrossRef] [Green Version]
  18. Jones, G.; Prosser, D.E.; Kaufmann, M. 25-hydroxyvitamin D-24-hydroxylase (CYP24A1): Its important role in the degradation of vitamin D. Arch. Biochem. Biophys. 2012, 523, 9–18. [Google Scholar] [CrossRef] [PubMed]
  19. Blau, J.E.; Collins, M.T. The PTH-vitamin D-FGF23 axis. Rev. Endocr. Metab. Disord. 2015, 16, 165–174. [Google Scholar] [CrossRef] [PubMed]
  20. Bikle, D.D.; Patzek, S.; Wang, Y. Physiologic and pathophysiologic roles of extra renal CYP27b1: Case report and review. Bone Rep. 2018, 8, 255–267. [Google Scholar] [CrossRef]
  21. Charoenngam, N. Vitamin D and rheumatic diseases: A review of clinical evidence. Int. J. Mol. Sci. 2021, 22, 10659. [Google Scholar] [CrossRef] [PubMed]
  22. Aranow, C. Vitamin D and the immune system. J. Investig. Med. 2011, 59, 881–886. [Google Scholar] [CrossRef] [Green Version]
  23. Bikle, D.D. Vitamin D regulation of immune function. Curr. Osteoporos. Rep. 2022, 20, 186–193. [Google Scholar] [CrossRef]
  24. Gombart, A.F. The vitamin D-antimicrobial peptide pathway and its role in protection against infection. Future Microbiol. 2009, 4, 1151–1165. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Hewison, M. Vitamin D and immune function: An overview. Proc. Nutr. Soc. 2012, 71, 50–61. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Adams, J.S.; Hewison, M. Unexpected actions of vitamin D: New perspectives on the regulation of innate and adaptive immunity. Nat. Clin. Pract. Endocrinol. Metab. 2008, 4, 80–90. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Lemire, J.M.; Archer, D.C.; Beck, L.; Spiegelberg, H.L. Immunosuppressive actions of 1,25-dihydroxyvitamin D3: Preferential inhibition of Th1 functions. J. Nutr. 1995, 125, 1704S–1708S. [Google Scholar] [CrossRef]
  28. Boonstra, A.; Barrat, F.J.; Crain, C.; Heath, V.L.; Savelkoul, H.F.J.; O’Garra, A. 1α,25-dihydroxyvitamin D3 has a direct effect on naive CD4+ T cells to enhance the development of Th2 cells. J. Immunol. 2001, 167, 4974. [Google Scholar] [CrossRef] [Green Version]
  29. Tang, J.; Zhou, R.; Luger, D.; Zhu, W.; Silver, P.B.; Grajewski, R.S.; Su, S.B.; Chan, C.C.; Adorini, L.; Caspi, R.R. Calcitriol suppresses antiretinal autoimmunity through inhibitory effects on the Th17 effector response. J. Immunol. 2009, 182, 4624–4632. [Google Scholar] [CrossRef] [Green Version]
  30. Chen, S.; Sims, G.P.; Chen, X.X.; Gu, Y.Y.; Chen, S.; Lipsky, P.E. Modulatory effects of 1,25-dihydroxyvitamin D3 on human B cell differentiation. J. Immunol. 2007, 179, 1634–1647. [Google Scholar] [CrossRef] [Green Version]
  31. Kongsbak, M.; Levring, T.B.; Geisler, C.; von Essen, M.R. The vitamin d receptor and T cell function. Front. Immunol. 2013, 4, 148. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Sarkar, S.; Hewison, M.; Studzinski, G.P.; Li, Y.C.; Kalia, V. Role of vitamin D in cytotoxic T lymphocyte immunity to pathogens and cancer. Crit. Rev. Clin. Lab. Sci. 2016, 53, 132–145. [Google Scholar] [CrossRef] [PubMed]
  33. Sung, C.-C.; Liao, M.-T.; Lu, K.-C.; Wu, C.-C. Role of vitamin D in insulin resistance. J. Biomed. Biotechnol. 2012, 2012, 634195. [Google Scholar] [CrossRef] [Green Version]
  34. Wolden-Kirk, H.; Overbergh, L.; Christesen, H.T.; Brusgaard, K.; Mathieu, C. Vitamin D and diabetes: Its importance for beta cell and immune function. Mol. Cell. Endocrinol. 2011, 347, 106–120. [Google Scholar] [CrossRef]
  35. Gibson, C.C.; Davis, C.T.; Zhu, W.; Bowman-Kirigin, J.A.; Walker, A.E.; Tai, Z.; Thomas, K.R.; Donato, A.J.; Lesniewski, L.A.; Li, D.Y. Dietary vitamin D and its metabolites non-genomically stabilize the endothelium. PLoS ONE 2015, 10, e0140370. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Andrukhova, O.; Slavic, S.; Zeitz, U.; Riesen, S.C.; Heppelmann, M.S.; Ambrisko, T.D.; Markovic, M.; Kuebler, W.M.; Erben, R.G. Vitamin D is a regulator of endothelial nitric oxide synthase and arterial stiffness in mice. Mol. Endocrinol. 2014, 28, 53–64. [Google Scholar] [CrossRef] [Green Version]
  37. Bikle, D.D. Vitamin D and the skin: Physiology and pathophysiology. Rev. Endocr, Metab, Disord. 2012, 13, 3–19. [Google Scholar] [CrossRef] [Green Version]
  38. Fleet, J.C.; DeSmet, M.; Johnson, R.; Li, Y. Vitamin D and cancer: A review of molecular mechanisms. Biochem. J. 2012, 441, 61–76. [Google Scholar] [CrossRef] [Green Version]
  39. Hossein-nezhad, A.; Holick, M.F. Vitamin D for health: A global perspective. Mayo Clin. Proc. 2013, 88, 720–755. [Google Scholar] [CrossRef] [Green Version]
  40. Wacker, M.; Holick, M.F. Sunlight and vitamin D: A global perspective for health. Dermato-Endocrinology 2013, 5, 51–108. [Google Scholar] [CrossRef] [Green Version]
  41. Garland, C.F.; Kim, J.J.; Mohr, S.B.; Gorham, E.D.; Grant, W.B.; Giovannucci, E.L.; Baggerly, L.; Hofflich, H.; Ramsdell, J.W.; Zeng, K.; et al. Meta-analysis of all-cause mortality according to serum 25-hydroxyvitamin D. Am. J. Public Health 2014, 104, e43–e50. [Google Scholar] [CrossRef]
  42. Charoenngam, N.; Shirvani, A.; Holick, M.F. The ongoing D-lemma of vitamin D supplementation for nonskeletal health and bone health. Curr. Opin. Endocrinol. Diabetes Obes. 2019, 26, 301–305. [Google Scholar] [CrossRef] [PubMed]
  43. Manson, J.E.; Cook, N.R.; Lee, I.M.; Christen, W.; Bassuk, S.S.; Mora, S.; Gibson, H.; Gordon, D.; Copeland, T.; D’Agostino, D.; et al. Vitamin D supplements and prevention of cancer and cardiovascular disease. N. Engl. J. Med. 2019, 380, 33–44. [Google Scholar] [CrossRef]
  44. Pittas, A.G.; Dawson-Hughes, B.; Sheehan, P.; Ware, J.H.; Knowler, W.C.; Aroda, V.R.; Brodsky, I.; Ceglia, L.; Chadha, C.; Chatterjee, R.; et al. Vitamin D supplementation and prevention of type 2 diabetes. N. Engl. J. Med. 2019, 381, 520–530. [Google Scholar] [CrossRef] [Green Version]
  45. Hahn, J.; Cook, N.R.; Alexander, E.K.; Friedman, S.; Walter, J.; Bubes, V.; Kotler, G.; Lee, I.M.; Manson, J.E.; Costenbader, K.H. Vitamin D and marine omega 3 fatty acid supplementation and incident autoimmune disease: VITAL randomized controlled trial. BMJ 2022, 376, e066452. [Google Scholar] [CrossRef] [PubMed]
  46. Kennel, K.A.; Drake, M.T.; Hurley, D.L. Vitamin D deficiency in adults: When to test and how to treat. Mayo Clin. Proc. 2010, 85, 752–758. [Google Scholar] [CrossRef] [Green Version]
  47. Hansdottir, S.; Monick, M.M.; Hinde, S.L.; Lovan, N.; Look, D.C.; Hunninghake, G.W. Respiratory epithelial cells convert inactive vitamin D to its active form: Potential effects on host defense. J. Immunol. 2008, 181, 7090. [Google Scholar] [CrossRef] [Green Version]
  48. Tripathi, S.; Tecle, T.; Verma, A.; Crouch, E.; White, M.; Hartshorn, K.L. The human cathelicidin LL-37 inhibits influenza A viruses through a mechanism distinct from that of surfactant protein D or defensins. J. Gen. Virol. 2013, 94, 40–49. [Google Scholar] [CrossRef] [PubMed]
  49. Sousa, F.H.; Casanova, V.; Findlay, F.; Stevens, C.; Svoboda, P.; Pohl, J.; Proudfoot, L.; Barlow, P.G. Cathelicidins display conserved direct antiviral activity towards rhinovirus. Peptides 2017, 95, 76–83. [Google Scholar] [CrossRef]
  50. Barlow, P.G.; Svoboda, P.; Mackellar, A.; Nash, A.A.; York, I.A.; Pohl, J.; Davidson, D.J.; Donis, R.O. Antiviral activity and increased host defense against influenza infection elicited by the human cathelicidin LL-37. PLoS ONE 2011, 6, e25333. [Google Scholar] [CrossRef]
  51. Jiang, J.-S.; Chou, H.-C.; Chen, C.-M. Cathelicidin attenuates hyperoxia-induced lung injury by inhibiting oxidative stress in newborn rats. Free Radic. Biol. Med. 2020, 150, 23–29. [Google Scholar] [CrossRef] [PubMed]
  52. Quraishi, S.A.; De Pascale, G.; Needleman, J.S.; Nakazawa, H.; Kaneki, M.; Bajwa, E.K.; Camargo, C.A., Jr.; Bhan, I. Effect of cholecalciferol supplementation on vitamin D status and cathelicidin levels in sepsis: A randomized, placebo-controlled trial. Crit. Care Med. 2015, 43, 1928–1937. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Martineau, A.R.; Jolliffe, D.A.; Hooper, R.L.; Greenberg, L.; Aloia, J.F.; Bergman, P.; Dubnov-Raz, G.; Esposito, S.; Ganmaa, D.; Ginde, A.A.; et al. Vitamin D supplementation to prevent acute respiratory tract infections: Systematic review and meta-analysis of individual participant data. BMJ 2017, 356, i6583. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Guan, Y.; Hao, Y.; Guan, Y.; Bu, H.; Wang, H. The effect of vitamin D supplementation on rheumatoid arthritis patients: A systematic review and meta-analysis. Front. Med. 2020, 7, 596007. [Google Scholar] [CrossRef]
  55. McLaughlin, L.; Clarke, L.; Khalilidehkordi, E.; Butzkueven, H.; Taylor, B.; Broadley, S.A. Vitamin D for the treatment of multiple sclerosis: A meta-analysis. J. Neurol. 2018, 265, 2893–2905. [Google Scholar] [CrossRef]
  56. Stanescu, A.M.A.; Simionescu, A.A.; Diaconu, C.C. Oral vitamin D therapy in patients with psoriasis. Nutrients 2021, 13, 163. [Google Scholar] [CrossRef]
  57. Li, J.; Chen, N.; Wang, D.; Zhang, J.; Gong, X. Efficacy of vitamin D in treatment of inflammatory bowel disease: A meta-analysis. Medicine 2018, 97, e12662. [Google Scholar] [CrossRef]
  58. Ajabshir, S.; Asif, A.; Nayer, A. The effects of vitamin D on the renin-angiotensin system. J. Nephropathol. 2014, 3, 41–43. [Google Scholar] [CrossRef]
  59. Ali, R.M.; Al-Shorbagy, M.Y.; Helmy, M.W.; El-Abhar, H.S. Role of Wnt4/β-catenin, Ang II/TGFβ, ACE2, NF-κB, and IL-18 in attenuating renal ischemia/reperfusion-induced injury in rats treated with Vit D and pioglitazone. Eur. J. Pharmacol. 2018, 831, 68–76. [Google Scholar] [CrossRef]
  60. Wu, J.; Deng, W.; Li, S.; Yang, X. Advances in research on ACE2 as a receptor for 2019-nCoV. Cell. Mol. Life Sci. 2021, 78, 531–544. [Google Scholar] [CrossRef]
  61. Hanff, T.C.; Harhay, M.O.; Brown, T.S.; Cohen, J.B.; Mohareb, A.M. Is there an association between COVID-19 mortality and the renin-angiotensin system? A call for epidemiologic investigations. Clin. Infect. Dis. 2020, 71, 870–874. [Google Scholar] [CrossRef] [Green Version]
  62. Garvin, M.R.; Alvarez, C.; Miller, J.I.; Prates, E.T.; Walker, A.M.; Amos, B.K.; Mast, A.E.; Justice, A.; Aronow, B.; Jacobson, D. A mechanistic model and therapeutic interventions for COVID-19 involving a RAS-mediated bradykinin storm. eLife 2020, 9, e59177. [Google Scholar] [CrossRef]
  63. Evans, R.M.; Lippman, S.M. Shining light on the COVID-19 pandemic: A vitamin D receptor checkpoint in defense of unregulated wound healing. Cell. Metab. 2020, 32, 704–709. [Google Scholar] [CrossRef]
  64. Vila Cuenca, M.; Ferrantelli, E.; Meinster, E.; Pouw, S.M.; Kovačević, I.; de Menezes, R.X.; Niessen, H.W.; Beelen, R.H.J.; Hordijk, P.L.; Vervloet, M.G. Vitamin D Attenuates Endothelial Dysfunction in Uremic Rats and Maintains Human Endothelial Stability. J. Am. Heart Assoc. 2018, 7, e008776. [Google Scholar] [CrossRef] [PubMed]
  65. Mohammad, S.; Mishra, A.; Ashraf, M.Z. Emerging role of vitamin D and its associated molecules in pathways related to pathogenesis of thrombosis. Biomolecules 2019, 9, 649. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Erukhimov, J.A.; Tang, Z.-L.; Johnson, B.A.; Donahoe, M.P.; Razzack, J.A.; Gibson, K.F.; Lee, W.M.; Wasserloos, K.J.; Watkins, S.A.; Pitt, B.R. Actin-containing sera from patients with adult respiratory distress syndrome are toxic to sheep pulmonary endothelial cells. Am. J. Respir. Crit. Care Med. 2000, 162, 288–294. [Google Scholar] [CrossRef]
  67. Weise-Cross, L.; Sands, M.A.; Sheak, J.R.; Broughton, B.R.S.; Snow, J.B.; Gonzalez Bosc, L.V.; Jernigan, N.L.; Walker, B.R.; Resta, T.C. Actin polymerization contributes to enhanced pulmonary vasoconstrictor reactivity after chronic hypoxia. Am. J. Physiol. Heart Circ. Physiol. 2018, 314, H1011–H1021. [Google Scholar] [CrossRef] [PubMed]
  68. Speeckaert, M.M.; Speeckaert, R.; Delanghe, J.R. Vitamin D binding protein in COVID-19. Clin. Med. J. 2020, 20, e136–e137. [Google Scholar] [CrossRef]
  69. Menger, J.; Lee, Z.-Y.; Notz, Q.; Wallqvist, J.; Hasan, M.S.; Elke, G.; Dworschak, M.; Meybohm, P.; Heyland, D.K.; Stoppe, C. Administration of vitamin D and its metabolites in critically ill adult patients: An updated systematic review with meta-analysis of randomized controlled trials. Crit. Care 2022, 26, 268. [Google Scholar] [CrossRef]
  70. Quesada-Gomez, J.M.; Lopez-Miranda, J.; Entrenas-Castillo, M.; Casado-Díaz, A.; Nogues Y Solans, X.; Mansur, J.L.; Bouillon, R. Vitamin D endocrine system and COVID-19: Treatment with calcifediol. Nutrients 2022, 14, 2716. [Google Scholar] [CrossRef] [PubMed]
  71. Entrenas Castillo, M.; Entrenas Costa, L.M.; Vaquero Barrios, J.M.; Alcalá Díaz, J.F.; López Miranda, J.; Bouillon, R.; Quesada Gomez, J.M. Effect of calcifediol treatment and best available therapy versus best available therapy on intensive care unit admission and mortality among patients hospitalized for COVID-19: A pilot randomized clinical study. J. Steroid Biochem. Mol. Biol. 2020, 203, 105751. [Google Scholar] [CrossRef]
  72. Nogues, X.; Ovejero, D.; Pineda-Moncusí, M.; Bouillon, R.; Arenas, D.; Pascual, J.; Ribes, A.; Guerri-Fernandez, R.; Villar-Garcia, J.; Rial, A.; et al. Calcifediol treatment and COVID-19-related outcomes. J. Clin. Endocrinol. Metab. 2021, 106, e4017–e4027. [Google Scholar] [CrossRef]
  73. Alcala-Diaz, J.F.; Limia-Perez, L.; Gomez-Huelgas, R.; Martin-Escalante, M.D.; Cortes-Rodriguez, B.; Zambrana-Garcia, J.L.; Entrenas-Castillo, M.; Perez-Caballero, A.I.; López-Carmona, M.D.; Garcia-Alegria, J.; et al. Calcifediol treatment and hospital mortality due to COVID-19: A cohort study. Nutrients 2021, 13, 1760. [Google Scholar] [CrossRef]
  74. Maghbooli, Z.; Sahraian, M.A.; Jamalimoghadamsiahkali, S.; Asadi, A.; Zarei, A.; Zendehdel, A.; Varzandi, T.; Mohammadnabi, S.; Alijani, N.; Karimi, M.; et al. Treatment with 25-hydroxyvitamin D(3) (Calcifediol) is associated with a reduction in the blood neutrophil-to-lymphocyte ratio marker of disease severity in hospitalized patients with COVID-19: A pilot multicenter, randomized, placebo-controlled, double-blinded clinical trial. Endocr. Pract. 2021, 27, 1242–1251. [Google Scholar] [CrossRef]
  75. Charoenngam, N.; Kalajian, T.A.; Shirvani, A.; Yoon, G.H.; Desai, S.; McCarthy, A.; Apovian, C.M.; Holick, M.F. A pilot-randomized, double-blind crossover trial to evaluate the pharmacokinetics of orally administered 25-hydroxyvitamin D3 and vitamin D3 in healthy adults with differing BMI and in adults with intestinal malabsorption. Am. J. Clin. Nutr. 2021, 114, 1189–1199. [Google Scholar] [CrossRef]
  76. Charoenngam, N.; Mueller, P.M.; Holick, M.F. Evaluation of 14-day concentration-time curves of vitamin D3 and 25-hydroxyvitamin D3 in healthy adults with varying body mass index. Anticancer Res. 2022, 42, 5095. [Google Scholar] [CrossRef] [PubMed]
  77. Jetter, A.; Egli, A.; Dawson-Hughes, B.; Staehelin, H.B.; Stoecklin, E.; Goessl, R.; Henschkowski, J.; Bischoff-Ferrari, H.A. Pharmacokinetics of oral vitamin D3 and calcifediol. Bone 2014, 59, 14–19. [Google Scholar] [CrossRef] [PubMed]
  78. Prabhu, A.V.; Luu, W.; Sharpe, L.J.; Brown, A.J. Cholesterol-mediated degradation of 7-dehydrocholesterol reductase switches the balance from cholesterol to vitamin D synthesis. J. Biol. Chem. 2016, 291, 8363–8373. [Google Scholar] [CrossRef] [Green Version]
  79. Zhu, J.G.; Ochalek, J.T.; Kaufmann, M.; Jones, G.; Deluca, H.F. CYP2R1 is a major, but not exclusive, contributor to 25-hydroxyvitamin D production in vivo. Proc. Natl. Acad. Sci. USA 2013, 110, 15650–15655. [Google Scholar] [CrossRef] [Green Version]
  80. Charoenngam, N.; Nasr, A.; Shirvani, A.; Holick, M.F. Hereditary metabolic bone diseases: A review of pathogenesis, diagnosis and management. Genes 2022, 13, 1880. [Google Scholar] [CrossRef] [PubMed]
  81. Nesterova, G.; Malicdan, M.C.; Yasuda, K.; Sakaki, T.; Vilboux, T.; Ciccone, C.; Horst, R.; Huang, Y.; Golas, G.; Introne, W.; et al. 1,25-(OH)2D-24 hydroxylase (CYP24A1) deficiency as a cause of nephrolithiasis. Clin. J. Am. Soc. Nephrol. 2013, 8, 649–657. [Google Scholar] [CrossRef] [Green Version]
  82. Henderson, C.M.; Fink, S.L.; Bassyouni, H.; Argiropoulos, B.; Brown, L.; Laha, T.J.; Jackson, K.J.; Lewkonia, R.; Ferreira, P.; Hoofnagle, A.N.; et al. Vitamin D—Binding protein deficiency and homozygous deletion of the GC gene. N. Engl. J. Med. 2019, 380, 1150–1157. [Google Scholar] [CrossRef] [PubMed]
  83. Jaroenlapnopparat, A.; Suppakitjanusant, P.; Ponvilawan, B.; Charoenngam, N. Vitamin D-related genetic variations and nonalcoholic fatty liver disease: A systematic review. Int. J. Mol. Sci. 2022, 23, 9122. [Google Scholar] [CrossRef]
  84. Liao, J.L.; Qin, Q.; Zhou, Y.S.; Ma, R.P.; Zhou, H.C.; Gu, M.R.; Feng, Y.P.; Wang, B.Y.; Yang, L. Vitamin D receptor Bsm I polymorphism and osteoporosis risk in postmenopausal women: A meta-analysis from 42 studies. Genes Nutr. 2020, 15, 20. [Google Scholar] [CrossRef] [PubMed]
  85. Zhou, Y.; Li, S. Meta-analysis of vitamin D receptor gene polymorphisms in childhood asthma. Front. Pediatr. 2022, 10, 843691. [Google Scholar] [CrossRef] [PubMed]
  86. Nunes, I.F.O.C.; Cavalcante, A.A.C.M.; Alencar, M.V.O.B.; Carvalho, M.D.F.; Sarmento, J.L.R.; Teixeira, N.S.C.C.A.; Paiva, A.A.; Carvalho, L.R.; Nascimento, L.F.M.; Cruz, M.S.P.; et al. Meta-analysis of the association between the rs228570 vitamin D receptor gene polymorphism and arterial hypertension risk. Adv. Nutr. 2020, 11, 1211–1220. [Google Scholar] [CrossRef]
  87. Liu, N.; Zhang, T.; Ma, L.; Wei, W.; Li, Z.; Jiang, X.; Sun, J.; Pei, H.; Li, H. Vitamin D receptor gene polymorphisms and risk of alzheimer disease and mild cognitive impairment: A systematic review and meta-analysis. Adv. Nutr. 2021, 12, 2255–2264. [Google Scholar] [CrossRef]
  88. Wen, J.; Li, J.; Liang, X.; Wang, A. Association of polymorphisms in vitamin D-metabolizing enzymes DHCR7 and CYP2R1 with cancer susceptibility: A systematic review and meta-analysis. Dis. Mark. 2021, 2021, 6615001. [Google Scholar] [CrossRef]
  89. Tabaei, S.; Motallebnezhad, M.; Tabaee, S.S. Vitamin D receptor (VDR) gene polymorphisms and risk of coronary artery disease (CAD): Systematic review and meta-analysis. Biochem. Genet. 2021, 59, 813–836. [Google Scholar] [CrossRef]
  90. Li, X.; Gan, X.; Gong, J.; Mou, T.; Zhou, H.; Li, M. Association between vitamin D receptor polymorphisms and acute pancreatitis: A protocol for systematic review and meta analysis. Medicine 2021, 100, e25508. [Google Scholar] [CrossRef]
  91. Zhang, L.; Yin, X.; Wang, J.; Xu, D.; Wang, Y.; Yang, J.; Tao, Y.; Zhang, S.; Feng, X.; Yan, C. Associations between VDR gene polymorphisms and osteoporosis risk and bone mineral density in postmenopausal women: A systematic review and meta-analysis. Sci. Rep. 2018, 8, 981. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Tizaoui, K.; Kaabachi, W.; Hamzaoui, A.; Hamzaoui, K. Association between vitamin D receptor polymorphisms and multiple sclerosis: Systematic review and meta-analysis of case-control studies. Cell. Mol. Immunol. 2015, 12, 243–252. [Google Scholar] [CrossRef] [Green Version]
  93. Xue, L.-N.; Xu, K.-Q.; Zhang, W.; Wang, Q.; Wu, J.; Wang, X.-Y. Associations between vitamin D receptor polymorphisms and susceptibility to ulcerative colitis and Crohn’s disease: A meta-analysis. Inflamm. Bowel Dis. 2013, 19, 54–60. [Google Scholar] [CrossRef] [PubMed]
  94. Laplana, M.; Royo, J.L.; Fibla, J. Vitamin D receptor polymorphisms and risk of enveloped virus infection: A meta-analysis. Gene 2018, 678, 384–394. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. 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] [PubMed]
  96. McNally, J.D.; Sampson, M.; Matheson, L.A.; Hutton, B.; Little, J. Vitamin D receptor (VDR) polymorphisms and severe RSV bronchiolitis: A systematic review and meta-analysis. Pediatr. Pulmonol. 2014, 49, 790–799. [Google Scholar] [CrossRef] [PubMed]
  97. Tarighi, S.; Najafi, M.; Hossein-Nezhad, A.; Ghaedi, H.; Meshkani, R.; Moradi, N.; Fadaei, R.; Kazerouni, F.; Shanaki, M. Association between two common polymorphisms of vitamin D binding protein and the risk of coronary artery disease: A case-control study. J. Med. Biochem. 2017, 36, 349–357. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Carlberg, C.; Seuter, S.; de Mello, V.D.F.; Schwab, U.; Voutilainen, S.; Pulkki, K.; Nurmi, T.; Virtanen, J.; Tuomainen, T.-P.; Uusitupa, M. Primary vitamin D target genes allow a categorization of possible benefits of vitamin D₃ supplementation. PLoS ONE 2013, 8, e71042. [Google Scholar] [CrossRef] [Green Version]
  99. Shirvani, A.; Kalajian, T.A.; Song, A.; Holick, M.F. Disassociation of vitamin D’s calcemic activity and non-calcemic genomic activity and individual responsiveness: A randomized controlled double-blind clinical trial. Sci. Rep. 2019, 9, 17685. [Google Scholar] [CrossRef] [Green Version]
  100. Shirvani, A.; Kalajian, T.A.; Song, A.; Allen, R.; Charoenngam, N.; Lewanczuk, R.; Holick, M.F. Variable genomic and metabolomic responses to varying doses of vitamin D supplementation. Anticancer Res. 2020, 40, 535–543. [Google Scholar] [CrossRef]
  101. Karcioglu Batur, L.; Hekim, N. The role of DBP gene polymorphisms in the prevalence of new coronavirus disease 2019 infection and mortality rate. J. Med. Virol. 2021, 93, 1409–1413. [Google Scholar] [CrossRef]
  102. Apaydin, T.; Polat, H.; Dincer Yazan, C.; Ilgin, C.; Elbasan, O.; Dashdamirova, S.; Bayram, F.; Tukenmez Tigen, E.; Unlu, O.; Tekin, A.F.; et al. Effects of vitamin D receptor gene polymorphisms on the prognosis of COVID-19. Clin. Endocrinol. 2022, 96, 819–830. [Google Scholar] [CrossRef] [PubMed]
  103. Abdollahzadeh, R.; Shushizadeh, M.H.; Barazandehrokh, M.; Choopani, S.; Azarnezhad, A.; Paknahad, S.; Pirhoushiaran, M.; Makani, S.Z.; Yeganeh, R.Z.; Al-Kateb, A.; et al. Association of vitamin D receptor gene polymorphisms and clinical/severe outcomes of COVID-19 patients. Infect. Genet. Evol. 2021, 96, 105098. [Google Scholar] [CrossRef]
  104. Freitas, A.T.; Calhau, C.; Antunes, G.; Araújo, B.; Bandeira, M.; Barreira, S.; Bazenga, F.; Braz, S.; Caldeira, D.; Santos, S.C.R.; et al. Vitamin D-related polymorphisms and vitamin D levels as risk biomarkers of COVID-19 disease severity. Sci. Rep. 2021, 11, 20837. [Google Scholar] [CrossRef] [PubMed]
  105. Al-Anouti, F.; Mousa, M.; Karras, S.N.; Grant, W.B.; Alhalwachi, Z.; Abdel-Wareth, L.; Uddin, M.; Alkaabi, N.; Tay, G.K.; Mahboub, B.; et al. Associations between genetic variants in the vitamin D metabolism pathway and severity of COVID-19 among UAE Residents. Nutrients 2021, 13, 3680. [Google Scholar] [CrossRef] [PubMed]
  106. Kotur, N.; Skakic, A.; Klaassen, K.; Gasic, V.; Zukic, B.; Skodric-Trifunovic, V.; Stjepanovic, M.; Zivkovic, Z.; Ostojic, O.; Stevanovic, G.; et al. Association of vitamin D, zinc and selenium related genetic variants with COVID-19 disease severity. Front. Nutr. 2021, 8, 689419. [Google Scholar] [CrossRef] [PubMed]
  107. Peralta, E.M.; Rosales, Y.Z.; Mesa, T.C.; González, E.N.S.; Pérez, Y.H.; de los Ángeles González Torres, M.; Balbuena, H.R.; Teruel, B.M. TaqI polymorphism of the VDR gene: Aspects related to the clinical behavior of COVID-19 in Cuban patients. Egypt. J. Med. Hum. Genet. 2021, 22, 83. [Google Scholar] [CrossRef]
  108. Balzanelli, M.G.; Distratis, P.; Lazzaro, R.; Pham, V.H.; Tran, T.C.; Dipalma, G.; Bianco, A.; Serlenga, E.M.; Aityan, S.K.; Pierangeli, V.; et al. Analysis of gene single nucleotide polymorphisms in COVID-19 disease highlighting the susceptibility and the severity towards the Infection. Diagnostics 2022, 12, 2824. [Google Scholar] [CrossRef]
  109. Jafarpoor, A.; Jazayeri, S.M.; Bokharaei-Salim, F.; Ataei-Pirkooh, A.; Ghaziasadi, A.; Soltani, S.; Sadeghi, A.; Marvi, S.S.; Poortahmasebi, V.; Khorrami, S.M.S.; et al. VDR gene polymorphisms are associated with the increased susceptibility to COVID-19 among iranian population: A case-control study. Int. J. Immunogenet. 2022, 49, 243–253. [Google Scholar] [CrossRef] [PubMed]
  110. Mamurova, B.; Akan, G.; Tuncel, G.; Mogol, E.; Evren, E.U.; Evren, H.; Suer, H.K.; Sanlidag, T.; Ergoren, M.C. A strong association between the VDR gene markers and SARS-CoV-2 variant. Res. Sq. 2022. preprint. [Google Scholar] [CrossRef]
  111. Zeidan, N.M.S.; Lateef, H.M.A.E.; Selim, D.M.; Razek, S.A.; Abd-Elrehim, G.A.B.; Nashat, M.; ElGyar, N.; Waked, N.M.; Soliman, A.A.; Elhewala, A.A.; et al. Vitamin D deficiency and vitamin D receptor FokI polymorphism as risk factors for COVID-19. Pediatr. Res. 2022. [Google Scholar] [CrossRef]
  112. Albu-Mohammed, W.H.M.; Anvari, E.; Fateh, A. Evaluating the role of BglI rs739837 and TaqI rs731236 polymorphisms in vitamin D receptor with SARS-CoV-2 variants mortality rate. Genes 2022, 13, 2346. [Google Scholar] [CrossRef]
  113. Butler-Laporte, G.; Nakanishi, T.; Mooser, V.; Morrison, D.R.; Abdullah, T.; Adeleye, O.; Mamlouk, N.; Kimchi, N.; Afrasiabi, Z.; Rezk, N.; et al. Vitamin D and COVID-19 susceptibility and severity in the COVID-19 Host Genetics Initiative: A Mendelian randomization study. PLoS Med. 2021, 18, e1003605. [Google Scholar] [CrossRef] [PubMed]
  114. Patchen, B.K.; Clark, A.G.; Gaddis, N.; Hancock, D.B.; Cassano, P.A. Genetically predicted serum vitamin D and COVID-19: A Mendelian randomisation study. BMJ Nutr. Prev. Health 2021, 4, 213–225. [Google Scholar] [CrossRef]
  115. Amin, H.A.; Drenos, F. No evidence that vitamin D is able to prevent or affect the severity of COVID-19 in individuals with European ancestry: A Mendelian randomisation study of open data. BMJ Nutr. Prev. Health 2021, 4, 42–48. [Google Scholar] [CrossRef] [PubMed]
  116. Cui, Z.; Tian, Y. Using genetic variants to evaluate the causal effect of serum vitamin D concentration on COVID-19 susceptibility, severity and hospitalization traits: A Mendelian randomization study. J. Trans. Med. 2021, 19, 300. [Google Scholar] [CrossRef] [PubMed]
  117. Bouillon, R. Genetic and environmental determinants of vitamin D status. Lancet 2010, 376, 148–149. [Google Scholar] [CrossRef]
  118. Lucas, R.M.; Ponsonby, A.-L.; Dear, K.; Valery, P.C.; Taylor, B.; van der Mei, I.; McMichael, A.J.; Pender, M.P.; Chapman, C.; Coulthard, A.; et al. Vitamin D status: Multifactorial contribution of environment, genes and other factors in healthy Australian adults across a latitude gradient. J. Steroid Biochem. Mol. Biol. 2013, 136, 300–308. [Google Scholar] [CrossRef]
  119. Mezzavilla, M.; Tomei, S.; Alkayal, F.; Melhem, M.; Ali, M.M.; Al-Arouj, M.; Bennakhi, A.; Alsmadi, O.; Elkum, N. Investigation of genetic variation and lifestyle determinants in vitamin D levels in Arab individuals. J. Trans. Med. 2018, 16, 20. [Google Scholar] [CrossRef] [Green Version]
  120. Van Etten, E.; Verlinden, L.; Giulietti, A.; Ramos-Lopez, E.; Branisteanu, D.D.; Ferreira, G.B.; Overbergh, L.; Verstuyf, A.; Bouillon, R.; Roep, B.O.; et al. The vitamin D receptor gene FokI polymorphism: Functional impact on the immune system. Eur. J. Immunol. 2007, 37, 395–405. [Google Scholar] [CrossRef]
  121. Meyer, V.; Bornman, L. Cdx-2 polymorphism in the vitamin D receptor gene (VDR) marks VDR expression in monocyte/macrophages through VDR promoter methylation. Immunogenetics 2018, 70, 523–532. [Google Scholar] [CrossRef] [PubMed]
  122. Meyer, V.; Saccone, D.S.; Tugizimana, F.; Asani, F.F.; Jeffery, T.J.; Bornman, L. Methylation of the vitamin D receptor (VDR) gene, together with genetic variation, race, and environment influence the signaling efficacy of the toll-like receptor 2/1-VDR pathway. Front. Immunol. 2017, 8, 1048. [Google Scholar] [CrossRef] [Green Version]
  123. Selvaraj, P.; Vidyarani, M.; Alagarasu, K.; Prabhu Anand, S.; Narayanan, P.R. Regulatory role of promoter and 3′ UTR variants of vitamin D receptor gene on cytokine response in pulmonary tuberculosis. J. Clin. Immunol. 2008, 28, 306–313. [Google Scholar] [CrossRef]
  124. Nagasawa, H.; Sasaki, H.; Uto, Y.; Kubo, S.; Hori, H. Association of the macrophage activating factor (MAF) precursor activity with polymorphism in vitamin D-binding protein. Anticancer Res. 2004, 24, 3361. [Google Scholar]
Figure 1. Schematic representation of vitamin D metabolic pathway and the effects of vitamin D on calcium and phosphate homeostasis. Abbreviations: Ca: Calcium; FGF-23; Fibroblast growth factor-23; P: Phosphate; PTH: Parathyroid hormone; UVB: Ultraviolet-B radiation.
Figure 1. Schematic representation of vitamin D metabolic pathway and the effects of vitamin D on calcium and phosphate homeostasis. Abbreviations: Ca: Calcium; FGF-23; Fibroblast growth factor-23; P: Phosphate; PTH: Parathyroid hormone; UVB: Ultraviolet-B radiation.
Biomedicines 11 00400 g001
Figure 2. Effects of vitamin D on different cell types of the innate and adaptive immune systems. Abbreviations: 1,25(OH)2D: 1,25-dihydroxyvitamin D; 25(OH)D: 25-hydroxyvitamin D; CCR10: C-C chemokine receptor type 10; CTLA-4: cytotoxic T-lymphocyte-associated protein 4; CYP27B1: Cytochrome P450 family 27 subfamily B member 1; FoxP3: Foxhead box P3; IFN-γ: Interferon-γ; IgG: Immunoglobulin G; IgM: Immunoglobulin M; IL-1β; Interleukin-1β; IL-2: Interleukin-2; IL-4: Interleukin-4; IL-5: Interleukin-5; IL-8: Interleukin-8; IL-9: Interleukin-9; IL-10: Interleukin-10; IL-13: Interleukin-13; IL-17: Interleukin-17; IL-21: Interleukin-21; ILT3: Immunoglobulin-like transcript-3; NK: Natural killer; PD-L1: Programmed death-ligand 1; TH1; T helper 1; TH2: T helper 2; TH9: T helper 9; TH17: T helper 17; Treg: Regulatory T cell; TNF-α; Tumer necrosis factor-α; TLR2: Toll-like receptor 2; TLR4: Toll-like receptor 4 [21].
Figure 2. Effects of vitamin D on different cell types of the innate and adaptive immune systems. Abbreviations: 1,25(OH)2D: 1,25-dihydroxyvitamin D; 25(OH)D: 25-hydroxyvitamin D; CCR10: C-C chemokine receptor type 10; CTLA-4: cytotoxic T-lymphocyte-associated protein 4; CYP27B1: Cytochrome P450 family 27 subfamily B member 1; FoxP3: Foxhead box P3; IFN-γ: Interferon-γ; IgG: Immunoglobulin G; IgM: Immunoglobulin M; IL-1β; Interleukin-1β; IL-2: Interleukin-2; IL-4: Interleukin-4; IL-5: Interleukin-5; IL-8: Interleukin-8; IL-9: Interleukin-9; IL-10: Interleukin-10; IL-13: Interleukin-13; IL-17: Interleukin-17; IL-21: Interleukin-21; ILT3: Immunoglobulin-like transcript-3; NK: Natural killer; PD-L1: Programmed death-ligand 1; TH1; T helper 1; TH2: T helper 2; TH9: T helper 9; TH17: T helper 17; Treg: Regulatory T cell; TNF-α; Tumer necrosis factor-α; TLR2: Toll-like receptor 2; TLR4: Toll-like receptor 4 [21].
Biomedicines 11 00400 g002
Table 1. Characteristics of studies investigating the association between vitamin D-related genetic variations and COVID-19 susceptibility and severity.
Table 1. Characteristics of studies investigating the association between vitamin D-related genetic variations and COVID-19 susceptibility and severity.
StudyCountryStudy DesignPopulation Characteristics/MethodsStudied Vitamin D-Related Gene(s)Outcomes
Batur et al., 2021 [101]Multiple countriesEcological studyAllele frequencies were obtained from data reported in five cohort and two systematic review and meta-analysis studies. Number of cases of COVID-19 per million population was obtained from the WHO COVID-19 Situation Report—164 in each of the ten countries (China, Japan, Nigeria, Kenya, Mexico, Italy, Turkey, Finland, Germany, Czech). GC
-
Prevalence per million of COVID-19 in each country
-
Mortality rates per million of COVID-19 in each country
Apaydin et al., 2021 [102]TurkeyRetrospective cohort297 with RT-PCR confirmed COVID-19 admitted to Marmara University Education and Research Hospital between April and October 2020VDR
-
COVID-19 disease severity
-
Intensive care unit admission
-
Mortality
Abdollahzadeh et al., 2021 [103]IranRetrospective cohort500 COVID-19 patients hospitalized at different hospitals in Iran between 5 May and 25 September, 2020.VDR
-
Signs and symptoms of COVID-19
Freitas et al., 2021 [104]PortugalRetrospective cohort491 patients with laboratory confirmed COVID-19 from Santa Maria hospital and São João hospitalCYP2R1, CYP24A1, DHCR7, GC and VDR
-
COVID-19 disease severity
Al-Anouti et al., 2021 [105]United Arab EmiratesRetrospective cohort646 patients with RT-PCT confirmed COVID-19 infection from the Sheikh Khalifa Medical City, quarantine area in Abu Dhabi and Rashid Hospital in Dubai between April 2020 and January 2021CYP2R1, GC and VDR
-
Critical illness requiring hospital admission organ support (i.e., hypoxia, respiratory failure, septic shock or multiorgan failure)
Kotur et al., 2021 [106]SerbiaRetrospective cohort120 adult and pediatric patients with COVID-19 treated at the Clinic of Pulmonology, Clinical Center of Serbia and Children’s Hospital for Lung Diseases and Tuberculosis, Medical Center “Dr Dragiša Mišovic,” Belgrade, Serbia, between April and June of 2020.DHCR7, CYP2R1, GC, VDR
-
COVID-19 disease severity
Peralta et al., 2021 [107]CubaRetrospective cohort104 patients with COVID-19 randomly recruited from Cuban citizens aged >1 year old VDR
-
COVID-19 disease severity
Balzanelli et al., 2022 [108]ItalyCase-control41 patients with COVID-19 and 43 healthy controls recruited from the 118 Pre-hospital and Emergency Department of SG Moscati Hospital of Taranto, Italy between September 2020 and October 2020.VDR
-
COVID-19 susceptibility
Jafarpoor et al., 2022 [109]IranCase-control188 hospitalized patients with COVID-19 and 218 patients with suspected COVID-19 with mild signs recruited from hospitals affiliated with the Iran University of Medical Sciences between March 2020 and June 2020VDR and GC
-
COVID-19 hospitalization/susceptibility
Mamurova et al., 2022 (preprint) [110]CyprusCase-control600 patients admitted to Near East University Hospital consisting of 100 with Alpha variant, 100 with Delta variant, 100 with Omicron variant and 300 with negative COVID-19 RT-PCR testVDR
-
COVID-19 susceptibility
Zeidan et al., 2022 [111]EgyptCase-control180 patients with COVID-19 and 200 age-, sex-, season-at-enrollment-matched controls recruited from Cairo, Ain-Shams, and Assuit University hospitals between October 2020 and March 2021VDR
-
COVID-19 susceptibility
Albu-Mohammed et al., 2022 [112]IranCase-control1734 patients recovered patients with COVID-19 and 1450 deceased patients with COVID-19 referred to the Ilam University
of Medical Sciences between November 2020 and February 2022
VDR
-
COVID-19 mortality
Butler-Laporte et al., 2020 [113]The United KingdomMendelian randomization studyGenetic variants associated with 25(OH)D levels in a GWAS of 443,734 participants of European ancestry including 401,460 from the UK Biobank were used as instrumental variable. GWASs of COVID-19 susceptibility, hospitalization, and severe disease from the COVID-19 Host Genetics Initiative were used as outcome GWASs. CYP2R1, CYP24A1, DHCR7 and GC among other genes in a model predicting serum 25(OH)D level
-
COVID-19 susceptibility
-
Hospitalization
-
COVID-19 disease severity
Patchen et al., 2021 [114]The United KingdomMendelian randomization studyData from genome-wide analyses in the population-based UK Biobank and SUNLIGHT Consortium were used as instrumental variable. Data from the COVID-19 Host Genetics Initiative were used as outcome GWASs. Participants included 17,965 COVID-19 cases including 11,085 laboratory or physician-confirmed cases, 7885 hospitalized cases and 4336 severe respiratory cases and 1,370,547 controls, primarily of European ancestry.CYP2R1, CYP24A1, DHCR7 and GC among other genes in a model predicting serum 25(OH)D level
-
COVID-19 susceptibility
-
Hospitalization
-
COVID-19 disease severity
-
Severe respiratory COVID-19
Amin et al., 2022 [115]The United KingdomMendelian randomization studyData from a GWAS in the population-based UK Biobank were used as instrumental variable. Data from the COVID-19 Host Genetics Initiative were used as outcome GWASs.CYP2R1, CYP24A1, DHCR7 among GC among other genes in a model predicting vitamin D deficiency
-
COVID-19 susceptibility
-
COVID-19 severity
Cui et al., 2022 [116]The United KingdomMendelian randomization studyData from genome-wide analyses in the population-based UK Biobank and SUNLIGHT Consortium were used as instrumental variable. Data from the COVID-19 Host Genetics Initiative were used as outcome GWASs.Genes in a model predicting serum 25(OH)D level
-
COVID-19 susceptibility
-
Hospitalization
-
COVID-19 disease severity
Abbreviations: 25(OH)D: 25-hydroxyvitamin D; COVID-19: Coronavirus disease 2019; GWAS: Genome-wide association study; RT-PCR: Reverse transcription polymerase chain reaction; UK: The United Kingdom.
Table 2. Association between vitamin D-related genetic variations and COVID-19 susceptibility and severity.
Table 2. Association between vitamin D-related genetic variations and COVID-19 susceptibility and severity.
GeneSNPLocationAlleleFinding(s)
DHCR7rs12785878Intron 2T > G
-
Kotur et al., 2021 [106]: TT genotype associated with mild-moderate COVID-19 vs. severe COVID-19 (p = 0.03).
rs4944979Intron 16G > T
-
Al-Anouti et al., 2021 [105]: GG genotype associated with decreased risk of critical COVID-19 (p = 0.02).
rs4944997Intron 18G > A
-
Al-Anouti et al., 2021 [105]: GG genotype associated with decreased risk of critical COVID-19 (p = 0.02).
rs4944998Intron 18G > C
-
Al-Anouti et al., 2021 [105]: GG genotype associated with decreased risk of critical COVID-19 (p = 0.02).
rs4944076Intron 20A > G
-
Al-Anouti et al., 2021 [105]: AA genotype associated with decreased risk of critical COVID-19 (p = 0.008).
rs10898210Intron 20A > G
-
Al-Anouti et al., 2021 [105]: AA genotype associated with decreased risk of critical COVID-19 (p = 0.009).
CYP2R1rs107416575′UTRG > A
-
Al-Anouti et al., 2021 [105]: AA genotype associated with decreased risk of critical COVID-19 (p = 0.004).
-
Kotur et al., 2021 [106]: GG genotype associated with increased risk of severe COVID-19.
GCrs4588Exon 11C > A
-
Batur et al., 2021 [101]: no association with prevalence and mortality rates
rs7041 (BsuRI)Exon 11G > T
-
Batur et al., 2021 [101]: GT genotype associated with increased prevalence and mortality rates compared with TT genotype (p < 0.05).
-
Jafarpoor et al., 2022 [109]: no association with likelihood of COVID-19.
rs113876500Upstream of Exon 1G > T
-
Al-Anouti et al., 2021 [101]: AA genotype associated with decreased risk of critical COVID-19 (p = 0.02).
rs59241277Intron 1A > G
-
Al-Anouti et al., 2021 [101]: AA genotype associated with decreased risk of critical COVID-19 (p = 0.005).
rs182901986Intron 1G > A
-
Al-Anouti et al., 2021 [101]: GG genotype associated with decreased risk of critical COVID-19 (p = 0.01).
rs113574864Intron 6C > T
-
Al-Anouti et al., 2021 [101]: CC genotype associated with decreased risk of critical COVID-19 (p = 0.005).
rs60349934Intron 6T > C
-
Al-Anouti et al., 2021 [101]: TT genotype associated with decreased risk of critical COVID-19 (p = 0.01).
rs2282679Intron 12T > G
-
Freitas et al., 2021 [104]: associated with severity (p = 0.005).
-
Kotur et al., 2021 [106]: no association with COVID-19 severity.
VDRrs11568820 (CDX2)PromotorG > A
-
Abdollahzadeh et al., 2021 [103]: C allele associated with decreased COVID-19 severity compared with c allele (p < 0.05).
rs4516035 (EcoRV)PromotorT > C
-
Abdollahzadeh et al., 2021 [103]: e allele associated symptomatic COVID-19 compared with E allele (p < 0.05).
rs2228570 (FokI)Exon 2C > T
-
Apaydin et al., 2021 [102]: Ff (CT) genotype associated with increased COVID-19 severity.
-
Abdollahzadeh et al., 2021 [103]: f allele associated with symptomatic COVID-19 compared with f allele (p < 0.05).
-
Kotur et al., 2021 [106]: no association with COVID-19 severity.
-
Balzanelli et al., 2022 [108]: ff (TT) genotype associated with decreased COVID-19 susceptibility (p < 0.05).
-
Jafarpoor et al., 2022 [109]: f (T) allele associated with increased likelihood of COVID-19 (p = 0.001)
-
Zeiden et al., 2022 [111]: FF (CC) genotype associated with increased likelihood of COVID-19 compared with ff (TT) genotype (p < 0.001).
-
Mamuruva et al., 2022 (preprint) [110]: f (T) allele associated with increased likelihood of COVID-19.
rs731236 (TaqI)Exon 9A > G
-
Apaydin et al., 2021 [102]: TT genotype associated with ICU admission compared with Tt genotype (p = 0.08).
-
Abdollahzadeh et al., 2021 [103]: no association with COVID-19 severity.
-
Peralta et al. 2021 [107]: no association with symptomaticity of COVID-19
-
Balzanelli et al., 2022 [108]: tt genotype associated with decreased COVID-19 susceptibility (p < 0.05).
-
Jafarpoor et al., 2022 [109]: no association with likelihood of COVID-19.
-
Mamuruva et al., 2022 (preprint) [110]: t allele associated with increased likelihood of COVID-19.
-
Albu-Mohammed et al., 2022 [112]: Tt and tt genotype associated with increased COVID-19 Alpha variant mortality; tt genotype associated with increased COVID-19 Delta variant mortality; no association with COVID-19 Omicron BA.5 variant mortality
rs757343 (Tru9I)Exon 9A > G
-
Abdollahzadeh et al., 2021 [103]: u allele associated with increased COVID-19 severity compared with U allele (p < 0.05).
rs11574018Intron 1T > C
-
Al-Anouti et al., 2021 [101]: TT genotype associated with decreased risk of critical COVID-19 (p = 0.04).
rs11574024Intron 1G > T
-
Al-Anouti et al., 2021 [101]: GG genotype associated with decreased risk of critical COVID-19 (p = 0.04).
rs1544410 (BsmI)Intron 8G > A
-
Apaydin et al., 2021 [102]: no association with COVID-19 severity.
-
Abdollahzadeh et al., 2021 [103]: b allele associated with increased COVID-19 severity compared with B allele (p < 0.05).
-
Balzanelli et al., 2022 [108]: BB genotype associated with slightly increased COVID-19 susceptibility (p < 0.05).
rs7975232 (ApaI)Intron 8C > A
-
Apaydin et al., 2021 [102]: aa genotype associated with COVID-19 mortality (p = 0.001)
-
Abdollahzadeh et al., 2021 [103]: no association with COVID-19 severity.
-
Jafarpoor et al., 2022 [109]: no association with likelihood of COVID-19.
rs739837 (BglI)3′UTRG > T
-
Abdollahzadeh et al., 2021 [103]: no association with COVID-19 severity.
-
Albu-Mohammed et al., 2022 [112]: Gg and gg genotypes associated with increased COVID-19 Omicron BA.5 variant mortality; no association with COVID-19 Alpha and Delta variant mortality.
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

Charoenngam, N.; Jaroenlapnopparat, A.; Mettler, S.K.; Grover, A. Genetic Variations of the Vitamin D Metabolic Pathway and COVID-19 Susceptibility and Severity: Current Understanding and Existing Evidence. Biomedicines 2023, 11, 400. https://doi.org/10.3390/biomedicines11020400

AMA Style

Charoenngam N, Jaroenlapnopparat A, Mettler SK, Grover A. Genetic Variations of the Vitamin D Metabolic Pathway and COVID-19 Susceptibility and Severity: Current Understanding and Existing Evidence. Biomedicines. 2023; 11(2):400. https://doi.org/10.3390/biomedicines11020400

Chicago/Turabian Style

Charoenngam, Nipith, Aunchalee Jaroenlapnopparat, Sofia K. Mettler, and Ashna Grover. 2023. "Genetic Variations of the Vitamin D Metabolic Pathway and COVID-19 Susceptibility and Severity: Current Understanding and Existing Evidence" Biomedicines 11, no. 2: 400. https://doi.org/10.3390/biomedicines11020400

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