Abstract

The coronavirus disease 2019 (COVID-19) pandemic was declared a public health emergency of international concern by the World Health Organization. COVID-19 has high transmissibility and could result in acute lung injury in a fraction of patients. By counterbalancing the activity of the renin-angiotensin system, angiotensin-converting enzyme 2, which is the fusion receptor of the virus, plays a protective role against the development of complications of this viral infection. Vitamin D can induce the expression of angiotensin-converting enzyme 2 and regulate the immune system through different mechanisms. Epidemiologic studies of the relationship between vitamin D and various respiratory infections were reviewed and, here, the postulated mechanisms and clinical data supporting the protective role of vitamin D against COVID-19–mediated complications are discussed.

INTRODUCTION

Coronaviruses (CoVs) are positive, single-stranded RNA viruses capable of mutation and recombination and can infect mammals and birds, which can result in a spectrum of diseases.1–3 They are among the enveloped viruses with a small, membrane-integrated protein envelope that contributes to various aspects of the virus’s life cycle, including its assembly, budding, and pathogenesis,4 and makes them vulnerable to antimicrobial peptides.5 In humans, CoVs cause respiratory tract infections (RTIs) that are usually mild and are responsible for almost 15% of the epidemic common colds in winters. Coronaviruses also are able to contaminate a wide diversity of animals and cross species barriers, which results in outbreaks of severe (and sometimes lethal) RTIs such as severe acute respiratory syndrome (SARS) and the Middle East respiratory syndrome.1 In December 2019, Wuhan, China, became the origin of another outbreak of severe pneumonia (officially named coronavirus disease 2019 [COVID-19] by the World Health Organization) caused by a novel coronavirus called SARS-CoV-2.6–8 After an incubation period of 2–14 days (median, approximately 5 days),9 SARS-CoV-2 results in an RTI with manifestations ranging from asymptomatic or mild to a very severe form of the disease. The overall mortality rate among those hospitalized in China is 6.4%10 and the estimated mortality rate outside China is 15.2%11; the estimated basic reproduction number is between 2.24 and 3.58.2,12 As of this writing, most reported patients (mainly from China) are middle-aged adults and the disease seems to be more fatal in men and elderly patients.13–17 However, our knowledge regarding this disease is still in its infancy and rapidly evolving.

BACKGROUND

COVID-19 pathology

Applying a low-input, metagenomic, next-generation sequencing approach on RNA extracted from bronchoalveolar lavage of 2 patients with typical clinical features of the disease, Chen et al18 sequenced the entire genome of the SARS-CoV-2 virus encompassing 29 881 nucleotides.18 Advanced analyses demonstrated a 94.6% similarity between SARS-CoV-2 and SARS-CoV in the domains used for coronavirus species classification, suggesting the 2 viruses emerged from 1 species.19 More than a decade ago, it was demonstrated that SARS-CoV takes advantage of angiotensin-converting enzyme 2 (ACE2) as an essential receptor for binding to the host cells and infecting them.20 Importantly, Zhou et al,19 through a well-designed set of studies, demonstrated that SARS-CoV-2 also could use ACE2 as a receptor for cellular attachment in the ACE2-expressing cells of humans, civet, pig, and Chinese horseshoe bats (but not mouse), implying ACE2 is the fusion receptor of this virus.19

Apart from lung, ACE2 receptor is expressed in many other tissues, such as heart, endothelium, kidney, and intestine,21 which coincides with the tissue tropism of the SARS-CoV-2 virus. A review of preliminary studies showed that besides the main clinical findings (including fever, cough, and fatigue), acute respiratory distress, acute cardiac injury, acute renal failure, and diarrhea are among the most prevalent manifestations of COVID-19 infection.22,23 Angiotensin-converting enzyme 2 is the second angiotensin-converting enzyme and its main function is the regulation of the renin-angiotensin system (RAS).20 The RAS is a complex system consisting of a various enzymes, peptides, and receptors, including angiotensin-I-converting enzyme (ACE) and ACE2, and has an important role in several biological functions, such as regulation of the body blood pressure and water balance.24 The effects of RAS on lung injury could be mediated by its influence on the tone and permeability of lung vessels, vitality of epithelial cells, and the activation of fibroblasts.

Angiotensin-converting enzyme 2, in contrast to ACE, angiotensin (Ang) II, and Ang II type 1 receptor (AT1R), alleviated lung injury in experimental and in vivo models.25 Angiotensin-converting enzyme converts Ang I into Ang II, and binding of Ang II to AT1R contributes to a variety of consequences, such as vasoconstriction, proliferation, inflammation, and apoptosis. However, Ang-(1–7), a heptapeptide produced through hydrolysis of Ang II by ACE2, upon interaction with its receptor, neutralizes different functions of Ang II, conferring a counter-regulatory function for ACE2 against ACE.24,26 In 2002, in a candidate gene study, Marshall et al27 detected an association between the presence of a deletion polymorphism in the human ACE gene (DCP1), which is related to the higher activity of the ACE, with the development of acute respiratory distress syndrome and its associated mortality.27 After this study, Imai et al,28 in 2005, showed in an animal study that ACE2 can protect mice from development of sepsis-induced severe acute lung injury (ALI). In contrast, other constituents of the RAS (ie, ACE, Ang II, and AT1R) worsened the disease course and damaged pulmonary function. In this study, ACE-deficient mice demonstrated improvement in the disease manifestations, while a significant deterioration was detected in the lung performance of the Ace2 knockout mice. Also, the catalytic activity of ACE2 was suggested to protect against the development of ALI, whereas upregulation of Ang II by ACE was shown to play an important role in the pathogenesis of acute lung failure. On the other hand, the development of ALI was demonstrated to downregulate the ACE2 protein. Presumably, it was the first study that implied a direct protective role for ACE2 against the development of (sepsis-induced) lung injury.28 Later, these results were supported by other experimental studies that revealed a role for this enzyme in the protection against acute lung failure. In other words, ACE2 could be considered a key factor in both cell fusion of the virus and its pathogenicity, including lung, cardiovascular, and renal involvement.20

Vitamin D metabolism and deficiency

Although for years vitamin D was considered as a food constituent playing an important role in bone metabolism, to date, it is known as a steroid hormone with important regulatory roles in different physiological systems and pathways of the human body, including the immune system.29,30 Compelling evidence from around the world has illustrated an association between vitamin D deficiency and different infectious diseases, predominantly viral infections accompanied by a poor response to the standard treatments.30–34 Different clinical trials have demonstrated an association between vitamin D deficiency and increased risk of pulmonary infection, which is supported by various laboratory experimental studies highlighting different mechanisms, including the negative regulatory role of vitamin D on the renin-angiotensin signaling pathway. Vitamin D is produced in the skin from 7-dehydrocholesterol by ultraviolet radiation. Bound to the vitamin D–binding protein, vitamin D is transported to the liver, where it is hydroxylated to the major circulating form of vitamin D (25(OH)D) by at least 1 cytochrome P450 (CYP) hydroxylase. After transportation of 25(OH)D to the kidney, mediated by another CYP hydroxylase, the hormonally active form of vitamin D (1,25(OH)2D3) is synthesized.35,36 Although achieving the sufficient level of serum vitamin D is not far reaching, vitamin D deficiency is a global health issue. It is generally estimated that > 3 billion individuals around the world have vitamin D insufficiency, of whom half are suffering from actual vitamin D deficiency.37

ESTABLISHED CLINICAL EVIDENCE

Accumulating epidemiologic studies have demonstrated that low levels of plasma vitamin D could increase the incidence or severity of respiratory viral infections in humans, suggesting an important potential role for this vitamin in the prevention or treatment of viral RTIs.38 In a meta-analysis performed to study the association between low serum 25(OH)D concentration and RTIs in 11 randomized controlled trials (RCTs), including 5660 patients in total, Bergman et al39 reported a significant protective effect for vitamin D against RTI (odds ratio [OR], 0.64; 95%CI, 0.49–0.84). This protective effect was stronger when daily doses of vitamin D (vs bolus doses) were taken.39 In another meta-analysis, Pham et al40 discovered an inverse association between the circulating 25(OH)D level and risk (OR, 1.83; 95%CI, 1.42–2.37) as well as the severity (OR, 2.46; 95%CI, 1.65–3.66) of acute respiratory tract infections (ARTI). In this study, the highest risk of ARTI was associated with 25(OH)D concentrations < 15 ng/mL40 (to convert to nanomoles per liter, multiply by 2.496).

Vitamin D levels and dosing

Reviewing the current evidence suggests that the protective effect of vitamin D supplementation on reducing the incidence or severity of RTI only appears, or is most prominent in, patients deficient in vitamin D.41 Although, practically speaking, a serum 25(OH)D level < 20 ng/mL is considered deficient and < 30 ng/mL as insufficiency,42 in a secondary analysis of a US survey including 18 883 participants, 24% of individuals with 25(OH)D levels < 10 ng/mL reported having a recent RTI. In this study, cumulatively, recent RTI was reported by 44% of participants with vitamin D insufficiency, whereas only 17% of people with sufficient serum concentrations of vitamin D reported experiencing a recent RTI.43 Likewise, a case-control study of Bangladeshi children detected blood 25(OH)D concentrations of < 11.7 ng/mL to be linked with acute lower respiratory infection (ALRI) development. In this study, with each 4 ng/mL increase in the level of plasma vitamin D, the odds of ALRI development decreased by half.44 Also, in a case-control study conducted with 150 Indian children (aged 2–60 months), serum 25(OH)D concentrations > 9.01 ng/mL had a significantly protective effect against the development of severe ALRI (OR, 0.09; 95%CI, 0.03–0.24).45 In another Indian case-control study, in which the association between ALRI and serum vitamin D concentration was investigated, the mean ± SD serum 25(OH)D concentration of patients with ALRI (13.91 ± 2.99 ng/mL) was significantly less than that of the control subjects (19.38 ± 7.83 ng/mL).46 Also, in a Japanese case study of hospitalized children with ALRI, a significant correlation between serum vitamin D concentrations < 15 ng/mL and the need for ventilator management was detected.47 In addition, a hospital-based case-control study of Turkish newborns reported a significant difference between those with ALRI and healthy participants in terms of their own mean serum 25(OH)D level as well as their mothers’. In this study, neonates with blood 25(OH)D levels < 10 ng/mL (OR, 4.25; 95%CI, 1.05–17.07) compared with those with blood 25(OH)D concentrations < 20 ng/mL (OR, 1.83; 95%CI, 0.31–10.52) were more susceptible to ALRI development.48 The results of this study were confirmed by other studies of Turkish and Chinese infants.49,50 Moreover, in another neonatal study, the mean cord blood 25(OH)D level of neonates hospitalized because of an ALRI was significantly lower than that of the control subjects, who were newborns not hospitalized because of an ALRI (14.82 ng/mL vs 22.43 ng/mL).51

To date, the majority of the studies seeking an association between blood concentrations of vitamin D and ALI have been directed toward children. In 2 distinct meta-analyses, both published in 2017, by pooling the results of different studies of children, vitamin D deficiency was significantly associated with a higher risk for ALRI development.52,53 Furthermore, an association was detected between vitamin D deficiency and the severity of lower RTI in 1 of these studies.53 Finally, a meta-analysis on the results of 25 RCTs, including 10 933 individuals of all ages (range, 0 to 95 years), confirmed the association between vitamin D deficiency and ARTI. In this analysis, supplementation with vitamin D decreased the risk of ARTI among all study participants (OR, 0.88; 95%CI, 0.81–0.96). Through a subgroup analysis, Martineau et al54 confined the protective effect of vitamin D only for the daily or weekly intake of the supplement, in contrast to the bolus doses. Furthermore, within the group that took daily or weekly vitamin D supplementation, the best protective results were detected among individuals with 25(OH)D levels < 10.02 ng/mL. Martineau et al54 concluded that vitamin D was safe and protective against ARTI, with more prominent results in patients with severe vitamin D deficiency who received daily or weekly doses of the supplement. The results of this study were in line with the outcomes of a prior meta-analysis of RCTs published in 2013 by Bergman et al,39 who concluded that daily vitamin D supplementation was more protective compared with bolus doses (OR, 0.51 vs 0.86, respectively).39

Critically ill settings

Mortality rates have been reported to be higher in vitamin D–deficient patients in the medical intensive care unit (ICU) compared with patients with 25(OH)D insufficiency or those with sufficient 25(OH)D levels (mean ± SD 25(OH)D level of survivors vs nonsurvivors, 27.9 ± 9.7 vs 9.7 ± 4.7 ng/mL, respectively).55 Interestingly, another study conducted in a critical illness setting showed that although taking vitamin D during ICU hospitalization was not beneficial, patients who took vitamin D supplements before ICU admission experienced more ICU-free and ventilator-free days, indicating the importance of vitamin D supplementation in patients who may eventually need to be admitted to ICU.56 In an RCT conducted in 2016 in the United States, 30 patients in the ICU who were receiving mechanical ventilatory support were assigned to 1 of 3 groups: (1) placebo (n = 10), (2) 50 000 IU/d vitamin D3 for 5 sequential days (ie, total dose, 250 000 IU; n = 9), and (3) 100 000 IU/d vitamin D3 for 5 sequential days (ie, total dose, 500 000 IU; n = 11).57 In this study, administration of high doses of vitamin D3 safely increased the plasma concentration of 25(OH)D to the sufficient range (mean ± SD plasma 25(OH)D levels, 45.7 ± 19.6 ng/mL and 55.2 ± 14.4 ng/mL after the 250 000 IU and 500 000 IU vitamin D3 supplementations, respectively). Both regimens of vitamin D resulted in shortening hospitalization time. Although not significantly different, a decreasing trend was also noticed in length of ICU stay (mean ± SD, 23 ± 14 days for the placebo group vs 17 ± 14 and 15 ± 10 days in the 250 000 IU and 500 000 IU vitamin D3–supplementation groups, respectively) and in the number of days receiving ventilatory support (mean ± SD, 20 ± 15 in placebo vs 12 ± 10 and 14 ± 10 in the 250 000 IU and 500 000 IU vitamin D3–supplementation groups, respectively).57

Vitamin D supplementation and risk of toxicity

Ideally, the human body acquires a major part of its daily vitamin D need from certain foods, including dairy products, oily fish, and egg yolk, in addition to UVB-based synthesis in the skin.58,59 However, levels are not sufficient in a considerable part of the population worldwide and vitamin D deficiency is a global health issue for both sexes in all age groups.37,39 Furthermore, it should be noted that several groups of individuals, such as obese people, dark-skinned individuals living at high latitudes, seniors, patients with certain diseases such as renal disorders, as well as those who do not have enough vitamin D sources in their daily diet are at higher risk for vitamin D deficiency.55,60

To attain to an optimal 25(OH)D plasma level (30–50 ng/mL), international guidelines recommend daily supplementation of 400 IU–2000 IU for the general population (based on age, sex, body weight, skin color, time outdoors, and geographic latitude) throughout the year.58,59,61,62 Adults deficient in vitamin D are recommended to take weekly doses of 50 000 IU for up to 3 months or daily doses of 6000 IU followed by maintenance doses of 1500–2000 IU/d after achieving optimal blood concentration.58 The recommended doses increase for pregnant and breastfeeding women, obese adults (body mass index > 30 kg/m2), seniors, night workers, and dark-skinned people, as well as patients with a disability; and decrease for neonates and children.62,63 However, each country may have developed a clinical practice guideline with the recommendations best suited for its own residents.

Supplementation with both D2 (ergocalciferol) and D3 (cholecalciferol) is possible and the 2 types of the vitamin have been shown to be effective in maintaining the optimal level of vitamin D concentration.63,64 However, a study in 2004 of 20 healthy volunteers demonstrated a lower potency of less than one-third in addition to a shorter half-life for ergocalciferol relative to cholecalciferol.65 This result was confirmed by an RCT performed 15 years later.66 Therefore, it seems that D2 is less effective and has a shorter duration of action in humans compared with vitamin D3.65,66

A concern that may arise in this context is the possibility of vitamin D toxicity. Acute vitamin D toxicity, defined by blood 25(OH)D concentration > 150 ng/mL, is extremely rare and the acute toxic dose for vitamin D has not yet been well defined.67–69 On the basis of current evidence, the chronic toxicity appears after daily intake of > 40 000–60 000 IU for ≥ 1 months consecutively.67–71 Therefore, the possibility of vitamin D toxicity is slight. However, a report by the US Institute of Medicine in 2011, based on several association studies, suggested a possible association between serum 25(OH)D concentrations > 50 ng/mL and detrimental outcomes, such as increasing the risk of falls, certain cancers, and even death.69,72 In an observational cohort study with 5 years’ follow-up of approximately 250 000 Danish participants, the association between serum concentration of 25(OH)D and death due to cardiovascular disease, acute myocardial infarction, or stroke was examined. The lowest mortality risk was reported in association with vitamin D concentration of 22 ng/mL, whereas lower and higher extremes of vitamin D concentration were associated with increased mortality risk in a reverse J-shape (ie, the highest risk at the lower concentration levels). Compared with the optimal concentration (22 ng/mL), the vitamin D plasma level of 39.3 ng/mL was associated with a mortality hazard ratio of 1.3.73 However, in another observational study, analyzing data of 26 916 individuals from 8 European cohorts, increasing serum 25(OH)D concentrations up to 39.3 ng/mL was not associated with changes in the mortality rate.74 Nevertheless, 2 RCTs conducted with seniors ≥ 70 years demonstrated that high single doses of vitamin D (monthly or annually) could be related to a higher risk of falls and fractures in this age group.75,76 By and large, according to present knowledge, vitamin D toxicity could occur mainly as a result of taking extremely high doses of vitamin D and with the very high concentrations of blood 25(OH)D (ie, > 150 ng/mL).69

POTENTIAL MECHANISMS OF VITAMIN D IN MITIGATING COVID-19 PROGRESSION

RAS regulation

Various clinical and epidemiologic studies have demonstrated a strong link between vitamin D and the regulation of the RAS complex network.55,77–79 In lipopolysaccharide-induced ALI in rats, the active form of vitamin D can inhibit the biosynthesis of renin, ACE, and Ang II, and induce expression of ACE2. As a result, vitamin D, by inducing the activity of the ACE2/Ang-(1–7) axis and reducing renin and the activity of ACE/Ang II pathway, could negatively regulate the RAS.24 Blocking of the (pro)renin receptor in a randomized rodent model demonstrated protection against lipopolysaccharide-induced ALI by reduction of the inflammatory response in the pulmonary cells.80 A reduction in interstitial edema and hemorrhage, as well as a decline in the leukocyte count and levels of tumor necrosis factor-α and various interleukins, including interleukin-6, were also observed in this study.80 In addition, vitamin D could suppress the expression of renin, and this effect seems to be independent of the Ang II feedback regulation.77 In wild-type mice, vitamin D deficiency results in renin synthesis, and supplementation with 1,25(OH)2D3 suppresses the expression of renin.77 Furthermore, in an in vitro study using a cell line with high expression level of renin, Li et al77 found that 1,25(OH)2D3 directly and extensively suppressed the transcription of the renin gene by a vitamin D receptor (VDR)-mediated mechanism.77 Particularly, it has been observed that poor vitamin D status could result in increased susceptibility to infection by enveloped viruses.81 The nontrivial role played by the different elements of the RAS in the development of COVID-19 complications and the regulatory association between vitamin D and those elements underscore the importance of this postulated mechanism. However, in addition to the particular association between vitamin D and the CoV-specific receptor, ACE2, there are other interesting connections between this vitamin and the respiratory system and viral infections. Although the exact underlying mechanisms by which the poor vitamin D status could contribute to the development and worsening of the viral diseases remain to be fully discovered, there are a few hypotheses.82

Vitamin D receptor

Most of the immune system cells, including dendritic cells and T and B lymphocytes, express a high level of VDR, which functions as a transcription factor; upon binding to vitamin D, VDR modulates the responses of the cells to the viruses.61,81 Vitamin D receptor is also expressed in pulmonary tissue. In 2 distinct in vivo murine studies that compared the severity of lipopolysaccharide-induced ALI between VDR-knockout and wild-type mice, more severe ALI83,84 and higher mortality rate83 were reported in the VDR-knockout mice. Other important observations in the lung cells of VDR-knockout mice included elevated alveolar permeability and pulmonary vascular leakiness, increased neutrophil infiltration, apoptosis, pulmonary inflammation, and expression of pro-inflammatory cytokines and chemokines, in addition to the inordinate expression of Ang II.83,84 Moreover, FokI T allele was significantly associated with an increased viral infection susceptibility to the enveloped viruses.81

FokI is a major polymorphism in VDR that is associated with functional modifications in this receptor, such as inducing alterations in the interaction between VDR and its transcription factor that result in the changes in its transcriptional activity.81,85

Upregulation of the CYP27B1 gene and downstream antimicrobial peptides

In 2008, Hansdottir et al86 showed that primary lung epithelial cells express relatively high basal levels of CYP27B1 and low levels of CYP24A1. CYP27B1 encodes the 1α-hydroxylase enzyme that is responsible for the final step in the synthesis of the active, hormonal form of vitamin D (1,25(OH)2D3) from its circulating form (25(OH)D), whereas the CYP24A1 gene translates to 24-hydroxylase, which catalyzes the degradation of the active form of vitamin D. This locally generated, active vitamin D is believed to play an important role in the modulation of the immune system.86 Many immune cells, owing to the expression of CYP27B1, are also able to convert the inactive form of vitamin D to its active form. This activation is linked to the production and expression regulation of antimicrobial peptides such as α- and β-defensins and cathelicidins.30,87 This is one of the important roles played by vitamin D in the regulation of the immune system and confers critical antimicrobial functions and immunomodulatory activities in innate and adaptive immunity.88–93

Defensins are expressed by human airway epithelia and are present in airway secretions, playing a role in respiratory mucosal defence.94,95 Locally activated vitamin D can directly induce the expression of cathelicidin peptides.96 Intriguingly, Hansdottir et al,86 through a set of experiments on the live respiratory syncytial virus, detected that viral RNA is able to increase the expression of 1α-hydroxylase and, subsequently, conversion of vitamin D in primary epithelial cells, and they suggested the active form of vitamin D and viral RNA act synergistically to increase the expression of the antimicrobial peptides, particularly cathelicidin.86

Cathelicidins are antimicrobial peptides that are part of the innate immune system of many vertebrates, including humans, with a vast spectrum of direct and indirect antimicrobial activity against different pathogens, including enveloped viruses.97,98 The vitamin D-cathelicidin axis is considered an important player in the regulation of the human immune system and modulation of innate and adaptive immunity.99 LL-37 is the only identified member of the cathelicidin family in humans that is expressed by respiratory epithelial cells; it regulates and boosts the microbial combating capacity of the host cells against a wide range of respiratory pathogens.88,100,101 Vitamin D can induce the expression of the LL-37 gene (CAMP).102–104

Regulation of the inflammatory response

Regulating the activity of the important nuclear factor-κB (NF-κB) pathway, via various mechanisms, is among other suggested immunomodulatory functions of vitamin D.105–109 In an investigation of the influence of vitamin D on respiratory syncytial virus–infected airway epithelial cells in an in vitro model, researchers demonstrated that a locally produced active form of vitamin D could induce IκBα in the airway epithelium. IκBα is an inhibitor of NF-κB that can reduce the expression of the NF-κB–driven genes in response to the virus infection. IFN-b is one of these genes that translates to proteins with prominent antiviral activity. However, intriguingly, despite the reduction in the expression of IFN-b, this process does not increase the virus replication rate, implying it plays a role in controlling the inflammatory response of the body’s cells to the virus infection, which results in diminished severity of the disease and a lowering of the morbidity and mortality risks of the infection.38 Also, in a review article110 summarizing the results of in vitro investigations performed to explore the immunomodulatory effects of vitamin D in the virus-infected human respiratory epithelial cells, even though this vitamin and its metabolites were not shown to confer an inhibitory effect against virus replication in the cells, all the reviewed studies highlighted the influence of vitamin D on the expression and secretion of chemokines and pro-inflammatory cytokines. Although administration of vitamin D was associated with an increased secretion of the chemokines of CXCL8 and CXCL10 in rhinovirus-infected epithelial cells, vitamin D supplementation decreased the production of a wide range of different pro-inflammatory cytokines and chemokines in respiratory syncytial virus– and influenza A–infected cells.110 Postulated mechanisms through which vitamin D could play a protective role against viral RTIs are summarized in Figure 1.

Figure 1

Postulated mechanisms through which vitamin D could play a protective role against viral RTI. (Upper box) Basal condition. Angiotensin (Ang) I, the product of the renin-mediated conversion of angiotensinogen, is hydrolyzed to Ang II by the peptidase angiotensin-I-converting enzyme (ACE). Ang II, upon binding to Ang II type 1 receptor (AT1R), confers its deleterious effects such as RTI through different mechanisms, including the development of interstitial edema and hemorrhage, and increasing the expression of pro-inflammatory cytokines and chemokines. Angiotensin-converting enzyme 2 (ACE2) hydrolyzes Ang II to Ang-(1–7), which upon interaction with its receptor (Mas), neutralizes different functions of Ang II and alleviates lung injury. IκBα is an inhibitor of NF-κB and reduces the expression of the NF-κB–driven genes (such as interferon β1 [IFN-b]) in response to the virus infection. IFN-b encodes a protein that, in response to a pathogen, induces transcription of inflammatory cytokines and chemokines. (Lower box) After treatment with vitamin D. Full arrowheads indicate the proposed effects of vitamin D. (A) The active form of vitamin D can suppress the expression of renin, ACE, and Ang II, and induce the expression of ACE2. (B) Suppression of the renin-angiotensin system could reduce the interstitial edema and hemorrhage and decrease the count of leukocytes and levels of pro-inflammatory cytokines (eg, TNF-α, IL-1β, IL-6, and IL-12) and pro-inflammatory transcription factors (eg, NF-κB). (C) The locally produced active form of vitamin D could induce IκBα in the airway epithelium which through inhibition of NF-κB reduces the expression of the NF-κB–driven genes in response to the virus infection. (D) Locally activated vitamin D can regulate the expression of certain host defense peptides (HDPs) such as cathelicidin, α- and β-defensins, and hepcidins. HDPs play a critical role in innate and adaptive immunity and through different mechanisms, such as reduction of the viral replication and infectivity,110 regulates and boosts the microbial combating capacity of the host against a wide range of the respiratory pathogens. *The CYP27B1 gene, which encodes 1α-hydroxylase, is highly expressed in many immune cells and primary lung epithelial cells. 1α-Hydroxylase catalyzes the final step in the synthesis of the active form of vitamin D3. Abbreviations: CYP27B1, cytochrome P450 family 27 subfamily b member 1; IL, interleukin; NF-κB, nuclear factor-κB; RTI, respiratory tract infection; TNF, tumor necrosis factor.

CONCLUSION

ACE2 is considered as receptor for SARS-CoV-2 through which the virus attaches to the cell. However, ACE2 also plays a protective role in the development of complicated manifestations of this viral infection.20 The active form of vitamin D can induce the expression of ACE224 and regulate the immune system through different distinct mechanisms. There is evidence from laboratory experiments that vitamin D can reduce the replication of other respiratory viruses and this is supported by clinical data demonstrating a preventive role for vitamin D against viral RTIs, particularly in those suffering from very low plasma concentrations of vitamin D. Taking this information together and regarding the important role of ACE2 in the prevention of COVID-19 complications on one hand and its regulatory association with vitamin D on the other hand, it seems rational to consider a potential role for vitamin D against SARS-Cov-2, although to date, we do not know for sure whether such an effect exists. Presumably, the most protective effect of supplementation appears in patients deficient in vitamin D (ie, those with serum 25(OH)D levels < 10 ng/mL) and possibly prior to the emergence of complications. Although vitamin D is safe (toxicity being quite rare), easily available, and affordable, its deficiency is a worldwide health problem.37 Therefore, supplementation with daily (preferred) or weekly doses of vitamin D, aimed at increasing the concentration of serum 25(OH)D to the optimal level of 30–50 ng/mL69,111–114 could be considered a global strategy, being more important in countries with a high prevalence of vitamin D deficiency and particularly in those patients with COVID-19 who are a high risk of ICU admission.

Acknowledgments

The author wishes to thank Prof. Joseph Rafter for kindly reviewing this manuscript.

Author contributions. F.H. is solely responsible for all aspects of this report and is accountable for all aspects of the work.

Funding. None.

Declaration of interest. None.

References

1

McIntosh
K
,
Perlman
S.
Coronaviruses, including severe acute respiratory syndrome (SARS) and Middle East respiratory syndrome (MERS). In:
Douglas
Mandell
, ed.
Mandell, Douglas, and Bennett’s Principles and Practice of Infectious Diseases
. Vol
2
, 8th ed.
Philadelphia, PA
: Elsevier Saunders;
2014
:
1928
1936.e2
. doi: .

2

Lai
CC
,
Shih
TP
,
Ko
WC
, et al. 
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and coronavirus disease-2019 (COVID-19): the epidemic and the challenges
.
Int J Antimicrob Agents
.
2020
;
55
:
105924
.

3

Malik
YA.
Properties of coronavirus and SARS-CoV-2
.
Malays J Pathol.
2020
;
42
:
3
11
.

4

Schoeman
D
,
Fielding
BC.
Coronavirus envelope protein: current knowledge
.
Virol J.
2019
;
16
:
69

5

Peters
BM
,
Shirtliff
ME
,
Jabra-Rizk
MA.
Antimicrobial peptides: primeval molecules or future drugs?
PLoS Pathog.
2010
;
6
:
E1001067
.

6

Wang
C
,
Horby
PW
,
Hayden
FG
, et al. 
A novel coronavirus outbreak of global health concern
.
Lancet
.
2020
;
395
:
470
473
.

7

Robson
B.
Computers and viral diseases. Preliminary bioinformatics studies on the design of a synthetic vaccine and a preventative peptidomimetic antagonist against the SARS-CoV-2 (2019-nCoV, COVID-19) coronavirus
.
Comput Biol Med
.
2020
;
119
:
103670
.

8

Zhu
N
,
Zhang
D
,
Wang
W
, et al. 
A novel coronavirus from patients with pneumonia in China, 2019
.
N Engl J Med.
2020
;
382
:
727
733
.

9

Lauer
SA
,
Grantz
KH
,
Bi
Q
, et al. 
The incubation period of coronavirus disease 2019 (CoVID-19) from publicly reported confirmed cases: estimation and application
.
Ann Intern Med
.
2020
;
172
:
577
582
.

10

Wang
K
,
Zuo
P
,
Liu
Y
, et al. 
Clinical and laboratory predictors of in-hospital mortality in patients with COVID-19: a cohort study in Wuhan, China
.
Clin Infect Dis
. 2020; doi: 10.1093/cid/ciaa538.

11

Baud
D
,
Qi
X
,
Nielsen-Saines
K
, et al. 
Real estimates of mortality following COVID-19 infection
.
Lancet Infect Dis
.
2020
;
20
:
773
.

12

Zhao
S
,
Lin
Q
,
Ran
J
, et al. 
Preliminary estimation of the basic reproduction number of novel coronavirus (2019-nCoV) in China, from 2019 to 2020: a data-driven analysis in the early phase of the outbreak
.
Int J Infect Dis
.
2020
;
92
:
214
217
.

13

Huang
C
,
Wang
Y
,
Li
X
, et al. 
Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China
Lancet
.
2020
;
395
:
497
506
.

14

Chen
N
,
Zhou
M
,
Dong
X
, et al. 
Epidemiological and clinical characteristics of 99 cases of 2019 novel coronavirus pneumonia in Wuhan, China: a descriptive study
.
Lancet
.
2020
;
395
:
507
513
.

15

Chan
JFW
,
Yuan
S
,
Kok
KH
, et al. 
A familial cluster of pneumonia associated with the 2019 novel coronavirus indicating person-to-person transmission: a study of a family cluster
.
Lancet
.
2020
;
395
:
514
523
.

16

Du Y, Tu L, Zhu P, et al. Clinical Features of 85 Fatal Cases of COVID-19 from Wuhan. A Retrospective Observational Study. Am J Respir Crit Care Med. 2020;

201
:
1372
1379
. et al. 

17

Borges do Nascimento
IJ
,
Cacic
N
,
Abdulazeem
HM
, et al. 
Novel coronavirus infection (COVID-19) in humans: a scoping review and meta-analysis
.
J Clin Med.
2020
;
9
:
941
.

18

Chen
L
,
Liu
W
,
Zhang
Q
, et al. 
RNA based mNGS approach identifies a novel human coronavirus from two individual pneumonia cases in 2019 Wuhan outbreak
.
Emerg Microbes Infect
.
2020
;
9
:
313
319
.

19

Zhou
P
,
Yang
X
,
Lou Wang
XG
, et al. 
A pneumonia outbreak associated with a new coronavirus of probable bat origin
.
Nature
.
2020
;
579
:
270
273
.

20

Kuba
K
,
Imai
Y
,
Rao
S
, et al. 
Lessons from SARS: control of acute lung failure by the SARS receptor ACE2
.
J Mol Med.
2006
;
84
:
814
820
.

21

Zhang
H
,
Penninger
JM
,
Li
Y
, et al. 
Angiotensin-converting enzyme 2 (ACE2) as a SARS-CoV-2 receptor: molecular mechanisms and potential therapeutic target
.
Intensive Care Med.
2020
;
46
:
586
590
.

22

Millán-Oñate
J
,
Rodriguez-Morales
AJ
,
Camacho-Moreno
G
, et al. 
A new emerging zoonotic virus of concern: the 2019 novel Coronavirus (COVID-19)
.
Infect.
2020
;
24
:
187
.

23

Han
Q
,
Lin
Q
,
Jin
S
, et al. 
Coronavirus 2019-nCoV: a brief perspective from the front line
.
J Infect
.
2020
;
80
:
373
377
.

24

Xu
J
,
Yang
J
,
Chen
J
, et al. 
Vitamin D alleviates lipopolysaccharide-induced acute lung injury via regulation of the renin-angiotensin system
.
Mol Med Rep
.
2017
;
16
:
7432
7438
.

25

Fanelli
V
,
Ranieri
VM.
Mechanisms and clinical consequences of acute lung injury
.
Ann Am Thorac Soc.
2015
;
12
:
S3
S8
.

26

Ferrario
CM
,
Varagic
J.
The ANG-(1-7)/ACE2/mas axis in the regulation of nephron function
.
Am J Physiol - Ren Physiol
.
2010
;
298
:
F1297
F1305
.

27

Marshall
RP
,
Webb
S
,
Bellingan
GJ
, et al. 
Angiotensin converting enzyme insertion/deletion polymorphism is associated with susceptibility and outcome in acute respiratory distress syndrome
.
Am J Respir Crit Care Med.
2002
;
166
:
646
650
.

28

Imai
Y
,
Kuba
K
,
Rao
S
, et al. 
Angiotensin-converting enzyme 2 protects from severe acute lung failure
.
Nature
.
2005
;
436
:
112
116
.

29

Holick
M.
Vitamin D and bone health
.
J Nutr
.
1996
;
126
:
1159S
1164S
.

30

Azmi
H
,
Hassou
N
,
Ennaji
MM.
Vitamin D immunomodulatory role in chronic and acute viral diseases. In: Ennaji MM, ed.
Emerging and Reemerging Viral Pathogens
.
London, United Kingdom
: Elsevier;
2020
:
489
506
.

31

Walker
VP
,
Modlin
RL.
The vitamin D connection to pediatric infections and immune function
.
Pediatr Res.
2009
;
65
:
106R
1113
.

32

Cannell
JJ
,
Vieth
R
,
Umhau
JC
, et al. 
Epidemic influenza and vitamin D
.
Epidemiol Infect.
2006
;
134
:
1129
1140
.

33

Schwalfenberg
GK.
A review of the critical role of vitamin D in the functioning of the immune system and the clinical implications of vitamin D deficiency
.
Mol Nutr Food Res.
2011
;
55
:
96
108
.

34

Hansdottir
S
,
Monick
MM.
Vitamin D effects on lung immunity and respiratory diseases
.
Vitam Horm.
2011
;
86
:
217
237
.

35

Bikle
DD.
Vitamin D metabolism, mechanism of action, and clinical applications
.
Chem Biol
.
2014
;
21
:
319
329
.

36

Christakos
S
,
Ajibade
D
,
Dhawan
P
, et al. 
Vitamin D: metabolism
.
Rheum Dis Clin
.
2012
;
38
:
1
11
.

37

Wimalawansa
SJ.
Causes, benefits and consequences of vitamin D deficiency
.
J Community Med Health Res
.
2019
;
2
:
1
–4.

38

Hansdottir
S
,
Monick
MM
,
Lovan
N
, et al. 
Vitamin D decreases respiratory syncytial virus induction of NF-κB–linked chemokines and cytokines in airway epithelium while maintaining the antiviral state
.
J Immunol.
2010
;
184
:
965
974
.

39

Bergman
P
,
Lindh
ÅU
,
Björkhem-Bergman
L
, et al. 
Vitamin D and respiratory tract infections: a systematic review and meta-analysis of randomized controlled trials
.
PLoS One
.
2013
;
8
:
E65835
.

40

Pham
H
,
Rahman
A
,
Majidi
A
, et al. 
Acute respiratory tract infection and 25-hydroxyvitamin D concentration: a systematic review and meta-analysis
.
Int J Environ Res Public Health
.
2019
;
16
:
3020
.

41

Beard
JA
,
Bearden
A
,
Striker
R.
Vitamin D and the anti-viral state
.
J Clin Virol
.
2011
;
50
:
194
200
.

42

Holick
MF.
Medical progress: vitamin D deficiency
.
N Engl J Med.
2007
;
357
:
266
281
.

43

Ginde
AA
,
Mansbach
JM
,
Camargo
CA.
Association between serum 25-hydroxyvitamin D level and upper respiratory tract infection in the Third National Health and Nutrition Examination Survey
.
Arch Intern Med.
2009
;
169
:
384
390
.

44

Roth
DE
,
Shah
R
,
Black
RE
, et al. 
Vitamin D status and acute lower respiratory infection in early childhood in Sylhet, Bangladesh
.
Wiley Online Libr
.
2010
;
99
:
389
393
.

45

Wayse
V
,
Yousafzai
A
,
Mogale
K
, et al. 
Association of subclinical vitamin D deficiency with severe acute lower respiratory infection in Indian children under 5 y
.
Eur J Clin Nutr.
2004
;
58
:
563
567
.

46

Prasad
R
,
Sharma
A
,
Das
BK
, et al. 
Serum retinol, vitamin D and zinc levels in under five children with acute lower respiratory tract infections
.
Indian J Pediatr.
2019
;
86
:
196
197
.

47

Inamo
Y
,
Hasegawa
M
,
Saito
K
, et al. 
Serum vitamin D concentrations and associated severity of acute lower respiratory tract infections in Japanese hospitalized children
.
Pediatr Int
.
2011
;
53
:
199
201
.

48

Karatekin
G
,
Kaya
A
,
Salihoğlu
Ö
, et al. 
Association of subclinical vitamin D deficiency in newborns with acute lower respiratory infection and their mothers
.
Eur J Clin Nutr.
2009
;
63
:
473
477
.

49

Dinlen
N
,
Zenciroglu
A
,
Beken
S
, et al. 
Association of vitamin D deficiency with acute lower respiratory tract infections in newborns
.
J Matern Neonatal Med
.
2016
;
29
:
928
932
.

50

Jia
KP
,
Zhao
LF
,
Feng
N
, et al. 
Lower level of vitamin D3 is associated with susceptibility to acute lower respiratory tract infection (ALRTI) and severity: a hospital based study in Chinese infants
.
Int J Clin Exp Med
.
2017
;
10
:
7997
8003
.

51

Binks
MJ
,
Smith-Vaughan
HC
,
Marsh
R
, et al. 
Cord blood vitamin D and the risk of acute lower respiratory infection in indigenous infants in the Northern Territory
.
Med J Aust
.
2016
;
204
:
238.e1
238.e7
.

52

Allam
MF
,
Farid
MM
,
Allam
MF
, et al. 
Relation between vitamin D deficiency and acute lower respiratory tract infection: a meta-analysis
.
Egypt J Paediatr.
2017
;
34
:
285
295
.

53

Jat
KR.
Vitamin D deficiency and lower respiratory tract infections in children: a systematic review and meta-analysis of observational studies
.
Trop Doct.
2017
;
47
:
77
84
.

54

Martineau
AR
,
Jolliffe
DA
,
Greenberg
L
, et al. 
Vitamin D supplementation to prevent acute respiratory infections: individual participant data meta-analysis
.
Health Technol Assess.
2019
;
23
:
1
44
.

55

Venkatram
S
,
Chilimuri
S
,
Adrish
M
, et al. 
Vitamin D deficiency is associated with mortality in the medical intensive care unit
.
Crit Care.
2011
;
15
:
R292
.

56

Leclair
TR
,
Zakai
N
,
Bunn
JY
, et al. 
Vitamin D supplementation in mechanically ventilated patients in the medical intensive care unit
.
J Parenter Enter Nutr
.
2019
;
43
:
1037
1043
.

57

Han
JE
,
Jones
JL
,
Tangpricha
V
, et al. 
High dose vitamin D administration in ventilated intensive care unit patients: a pilot double blind randomized controlled trial
.
J Clin Transl Endocrinol
.
2016
;
4
:
59
65
.

58

Holick
MF
,
Binkley
NC
,
Bischoff-Ferrari
HA
, et al. 
Evaluation, treatment, and prevention of vitamin D deficiency: an Endocrine Society clinical practice guideline
.
J Clin Endocrinol Metab
.
2011
;
96
:
1911
1930
.

59

Rusinska
A
,
Płudowski
P
,
Walczak
M
, et al. 
Vitamin D supplementation guidelines for general population and groups at risk of vitamin D deficiency in Poland-Recommendations of the Polish society of pediatric endocrinology and diabetes and the expert panel with participation of national specialist c
.
Front Endocrinol (Lausanne)
.
2018
;
9
:
246
.

60

Grant
WB
,
Al Anouti
F
,
Moukayed
M.
Targeted 25-hydroxyvitamin D concentration measurements and vitamin D3 supplementation can have important patient and public health benefits
.
Eur J Clin Nutr.
2020
;
74
:
366
376
.

61

Pludowski
P
,
Holick
MF
,
Grant
WB
, et al. 
Vitamin D supplementation guidelines
.
J Steroid Biochem Mol Biol
.
2018
;
175
:
125
135
.

62

Haq
A
,
Wimalawansa
SJ
,
Pludowski
P
, et al. 
Clinical practice guidelines for vitamin D in the United Arab Emirates
.
J Steroid Biochem Mol Biol
.
2018
;
175
:
4
11
.

63

Kennel
KA
,
Drake
MT
,
Hurley
DL.
Vitamin D deficiency in adults: when to test and how to treat
.
Mayo Clin Proc
.
2010
;
85
:
752
758
.

64

Baroni
L
,
Goggi
S
,
Battaglino
R
, et al. 
Vegan nutrition for mothers and children: practical tools for healthcare providers
.
Nutrients
.
2018
;
11
:
5
.

65

Armas
LAG
,
Hollis
BW
,
Heaney
RP.
Vitamin D2 is much less effective than vitamin D3 in humans
.
J Clin Endocrinol Metab
.
2004
;
89
:
5387
5391
.

66

Martineau
AR
,
Thummel
KE
,
Wang
Z
, et al. 
Differential effects of oral boluses of vitamin D2 vs vitamin D3 on vitamin D metabolism: a randomized controlled trial
.
J Clin Endocrinol Metab
.
2019
;
104
:
5831
5839
.

67

Roop
JK.
Hypervitaminosis-an emerging pathological condition
.
Int J Heal Sci Res
.
2018
;
8
:
280
.

68

Rosenbloom
M.
At what dose is vitamin D toxic? Available at: https://www.medscape.com/answers/819426-102369/at-what-dose-is-vitamin-d-toxic. Accessed March 3,
2020
.

69

Marcinowska-Suchowierska
E
,
Kupisz-Urbanska
M
,
Lukaszkiewicz
J
, et al. 
Vitamin D toxicity–a clinical perspective
.
Front Endocrinol (Lausanne).
2018
;
9
:
550
.

70

Zeratsky
K.
Vitamin D toxicity: what if you get too much? Mayo Clinic. Available at: https://www.mayoclinic.org/healthy-lifestyle/nutrition-and-healthy-eating/expert-answers/vitamin-d-toxicity/faq-20058108. Accessed March 3,
2020
.

71

Vieth
R.
Vitamin D toxicity, policy, and science
.
J Bone Miner Res.
2007
;
22
:
V64
V68
.

72

Del Valle
H
,
Yaktine
A
,
Taylor
C
, eds.
Dietary Reference Intakes for Calcium and Vitamin D
. Washington, DC:
National Academies Press
;
2011
. doi:

73

Durup
D
,
Jørgensen
HL
,
Christensen
J
, et al. 
A reverse J-shaped association between serum 25-hydroxyvitamin D and cardiovascular disease mortality: the CopD Study
.
J Clin Endocrinol Metab
.
2015
;
100
:
2339
2346
.

74

Gaksch
M
,
Jorde
R
,
Grimnes
G
, et al. 
Vitamin D and mortality: individual participant data meta-analysis of standardized 25-hydroxyvitamin D in 26916 individuals from a European consortium
.
PLoS One.
2017
;
12
:
E0170791
.

75

Sanders
KM
,
Stuart
AL
,
Williamson
EJ
, et al. 
Annual high-dose oral vitamin D and falls and fractures in older women: a randomized controlled trial
.
JAMA
.
2010
;
303
:
1815
1822
.

76

Bischoff-Ferrari
HA
,
Dawson-Hughes
B
,
Orav
EJ
, et al. 
Monthly high-dose vitamin D treatment for the prevention of functional decline: a randomized clinical trial
.
JAMA Intern Med.
2016
;
176
:
175
183
.

77

Li
YC
,
Qiao
G
,
Uskokovic
M
, et al. 
Vitamin D: a negative endocrine regulator of the renin-angiotensin system and blood pressure
.
J Steroid Biochem Mol Biol
.
2004
;
89-90
:
387
392
.

78

Li
YC.
Vitamin D regulation of the renin-angiotensin system
.
J Cell Biochem.
2003
;
88
:
327
331
.

79

Li
Y.
Vitamin D and the renin-angiotensin system.: Feldman D, ed.
Vitamin D
.
London, United Kingdom
: Academic Press;
2018
:
825
847
.

80

Ishii
K
,
Takeuchi
H
,
Fukunaga
K
, et al. 
Attenuation of lipopolysaccharide-induced acute lung injury after (pro)renin receptor blockade
.
Exp Lung Res
.
2015
;
41
:
199
207
.

81

Laplana
M
,
Royo
JL
,
Fibla
J.
Vitamin D receptor polymorphisms and risk of enveloped virus infection: a meta-analysis
.
Gene
.
2018
;
678
:
384
394
.

82

Teymoori-Rad
M
,
Shokri
F
,
Salimi
V
, et al. 
The interplay between vitamin D and viral infections
.
Rev Med Virol.
2019
;
29
:
E2032
.

83

Kong
J
,
Zhu
X
,
Shi
Y
, et al. 
VDR attenuates acute lung injury by blocking Ang-2-Tie-2 pathway and renin-angiotensin system
.
Mol Endocrinol
.
2013
;
27
:
2116
2125
.

84

Shi
YY
,
Liu
TJ
,
Fu
JH
, et al. 
Vitamin D/VDR signaling attenuates lipopolysaccharide-induced acute lung injury by maintaining the integrity of the pulmonary epithelial barrier
.
Mol Med Rep
.
2016
;
13
:
1186
1194
.

85

Jurutka
PW
,
Remus
LS
,
Whitfield
GK
, et al. 
The polymorphic N terminus in human vitamin D receptor isoforms influences transcriptional activity by modulating interaction with transcription factor IIB
.
Mol Endocrinol
.
2000
;
14
:
401
420
.

86

Hansdottir
S
,
Monick
MM
,
Hinde
SL
, et al. 
Respiratory epithelial cells convert inactive vitamin D to its active form: potential effects on host defense
.
J Immunol.
2008
;
181
:
7090
7099
.

87

Youssef
DA
,
Miller
CWT
,
El-Abbassi
AM
, et al. 
Antimicrobial implications of vitamin D
.
Dermatoendocrinol
.
2011
;
3
:
220
229
.

88

Doss
M
,
White
MR
,
Tecle
T
, et al. 
Human defensins and LL-37 in mucosal immunity
.
J Leukoc Biol
.
2010
;
87
:
79
92
.

89

Barlow
PGP
,
Li
Y
,
Wilkinson
T
, et al. 
The human cationic host defense peptide LL‐37 mediates contrasting effects on apoptotic pathways in different primary cells of the innate immune system
.
J Leukoc Biol
.
2006
;
80
:
509
520
.

90

Albanna
E
,
Ali
Y
,
Elkashnia
R.
Vitamin D and LL-37 in children with pneumonia
.
Egypt J Pediatr Allergy Immunol
.
2010
;
8
:
81
86
.

91

Brice
DC
,
Diamond
G.
Antiviral activities of human host defense peptides
.
Curr Med Chem.
2020
;
27
:
1420
1443
.

92

Silva
ON
,
Porto
WF
,
Ribeiro
SM
, et al. 
Host-defense peptides and their potential use as biomarkers in human diseases
.
Drug Discov Today
.
2018
;
23
:
1666
1971
.

93

Shin
D-M
,
Jo
E-K.
Antimicrobial peptides in innate immunity against mycobacteria
.
Immune Netw.
2011
;
11
:
245
252
.

94

McCray
PB
,
Bentley
L.
Human airway epithelia express a β-defensin
.
Am J Respir Cell Mol Biol.
1997
;
16
:
343
349
.

95

Singh
PK
,
Jia
HP
,
Wiles
K
, et al. 
Production of β-defensins by human airway epithelia
.
Proc Natl Acad Sci USA
.
1998
;
95
:
14961
14966
.

96

White
JH.
Vitamin D as an inducer of cathelicidin antimicrobial peptide expression: past, present and future
.
J Steroid Biochem Mol Biol
.
2010
;
121
:
234
238
.

97

Agier
J
,
Efenberger
M
,
Brzezińska-Błaszczyk
E.
Cathelicidin impact on inflammatory cells
.
Cent Eur J Immunol.
2015
;
2
:
225
235
.

98

Zanetti
M
,
Gennaro
R
,
Romeo
D.
Cathelicidins: a novel protein family with a common proregion and a variable C-terminal antimicrobial domain
.
FEBS Lett
.
1995
;
374
:
1
5
.

99

Chung
C
,
Silwal
P
,
Kim
I
, et al. 
Vitamin D-cathelicidin axis: at the crossroads between protective immunity and pathological inflammation during infection
.
Immune Netw.
2020
;
20
:e12.

100

Bucki
R
,
Leszczyńska
K
,
Namiot
A
, et al. 
Cathelicidin LL-37: a multitask antimicrobial peptide
.
Arch Immunol Ther Exp.
2010
;
58
:
15
25
.

101

Agerberth
B
,
Gunne
H
,
Odeberg
J
, et al. 
FALL-39, a putative human peptide antibiotic, is cysteine-free and expressed in bone marrow and testis
.
Proc Natl Acad Sci USA
.
1995
;
92
:
195
199
.

102

Gombart
AF
,
O’Kelly
J
,
Saito
T
, et al. 
Regulation of the CAMP gene by 1,25(OH)2D3 in various tissues
.
J Steroid Biochem Mol Biol
.
2007
;
103
:
552
557
.

103

Weber
G
,
Heilborn
JD
,
Jimenez
CIC
, et al. 
Vitamin D induces the antimicrobial protein hCAP18 in human skin
.
J Invest Dermatol
.
2005
;
124
:
1080
1082
.

104

Wang
T-T
,
Nestel
FP
,
Bourdeau
V
, et al. 
Cutting edge: 1,25-dihydroxyvitamin D 3 is a direct inducer of antimicrobial peptide gene expression
.
J Immunol.
2004
;
173
:
2909
2912
.

105

Szeto
FL
,
Sun
J
,
Kong
J
, et al. 
Involvement of the vitamin D receptor in the regulation of NF-κB activity in fibroblasts
.
J Steroid Biochem Mol Biol
.
2007
;
103
:
563
566
.

106

Deb
DK
,
Chen
Y
,
Zhang
Z
, et al. 
1,25-Dihydroxyvitamin D3 suppresses high glucose-induced angiotensinogen expression in kidney cells by blocking the NF-κB pathway
.
Am J Physiol - Ren Physiol
.
2009
;
296
:F1212–F1218.

107

Riis
JL
,
Johansen
C
,
Gesser
B
, et al.  1α.
25(OH)2D3 regulates NF-κB DNA binding activity in cultured normal human keratinocytes through an increase in IκBα expression
.
Arch Dermatol Res.
2004
;
296
:
195
202
.

108

Sun
J
,
Kong
J
,
Duan
Y
, et al. 
Increased NF-κB activity in fibroblasts lacking the vitamin D receptor
.
Am J Physiol - Endocrinol Metab
.
2006
;
291
:E315–E322.

109

Cohen-Lahav
M
,
Shany
S
,
Tobvin
D
, et al. 
Vitamin D decreases NFκB activity by increasing IκBα levels
.
Nephrol Dial Transplant
.
2006
;
21
:
889
897
.

110

Greiller
CL
,
Martineau
AR.
Modulation of the immune response to respiratory viruses by vitamin D
.
Nutrients
.
2015
;
7
:
4240
4270
.

111

Marshall
B
,
Bennett
N
,
Smith
A
, et al. 
Can vitamin D prevent acute respiratory infections?
J Fam Pract.
2019
;
68
:
230
231
.

112

Scragg
R.
The Vitamin D Assessment (ViDA) study – design and main findings
.
J Steroid Biochem Mol Biol
.
2020
;
198
:
105562
.

113

Scragg
R
,
Waayer
D
,
Stewart
AW
, et al. 
The Vitamin D Assessment (ViDA) Study: design of a randomized controlled trial of vitamin D supplementation for the prevention of cardiovascular disease, acute respiratory infection, falls and non-vertebral fractures
.
J Steroid Biochem Mol Biol
.
2016
;
164
:
318
325
.

114

Sabetta
JR
,
Depetrillo
P
,
Cipriani
RJ
, et al. 
Serum 25-hydroxyvitamin D and the incidence of acute viral respiratory tract infections in healthy adults
.
PLoS One
.
2010
;
5
:
E11088
.

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://academic.oup.com/journals/pages/open_access/funder_policies/chorus/standard_publication_model)