Next Article in Journal
Relaxin Inhibits the Cardiac Myofibroblast NLRP3 Inflammasome as Part of Its Anti-Fibrotic Actions via the Angiotensin Type 2 and ATP (P2X7) Receptors
Next Article in Special Issue
Anxiety-like Behavior and GABAAR/BDZ Binding Site Response to Progesterone Withdrawal in a Stress-Vulnerable Strain, the Wistar Kyoto Rats
Previous Article in Journal
Systematic Review: Targeted Molecular Imaging of Angiogenesis and Its Mediators in Rheumatoid Arthritis
Previous Article in Special Issue
Peripheral Serotonin Deficiency Affects Anxiety-like Behavior and the Molecular Response to an Acute Challenge in Rats
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Molecular Basis Underlying the Therapeutic Potential of Vitamin D for the Treatment of Depression and Anxiety

by
Bruna R. Kouba
1,
Anderson Camargo
1,
Joana Gil-Mohapel
2,3,* and
Ana Lúcia S. Rodrigues
1,*
1
Center of Biological Sciences, Department of Biochemistry, Federal University of Santa Catarina, Florianópolis 88040-900, SC, Brazil
2
Island Medical Program, Faculty of Medicine, University of British Columbia, Victoria, BC V8P 5C2, Canada
3
Division of Medical Sciences, University of Victoria, Victoria, BC V8P 5C2, Canada
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(13), 7077; https://doi.org/10.3390/ijms23137077
Submission received: 31 May 2022 / Revised: 22 June 2022 / Accepted: 24 June 2022 / Published: 25 June 2022

Abstract

:
Major depressive disorder and anxiety disorders are common and disabling conditions that affect millions of people worldwide. Despite being different disorders, symptoms of depression and anxiety frequently overlap in individuals, making them difficult to diagnose and treat adequately. Therefore, compounds capable of exerting beneficial effects against both disorders are of special interest. Noteworthily, vitamin D deficiency has been associated with an increased risk of developing depression and anxiety, and individuals with these psychiatric conditions have low serum levels of this vitamin. Indeed, in the last few years, vitamin D has gained attention for its many functions that go beyond its effects on calcium–phosphorus metabolism. Particularly, antioxidant, anti-inflammatory, pro-neurogenic, and neuromodulatory properties seem to contribute to its antidepressant and anxiolytic effects. Therefore, in this review, we highlight the main mechanisms that may underlie the potential antidepressant and anxiolytic effects of vitamin D. In addition, we discuss preclinical and clinical studies that support the therapeutic potential of this vitamin for the management of these disorders.

1. Introduction

Major depressive disorder (MDD) and anxiety disorders, which in this review will be referred to as depression and anxiety, are devastating and highly prevalent clinical entities that constitute one of the leading causes of disability worldwide [1,2]. These disorders often occur concomitantly and their symptoms frequently overlap in individuals, and patients with these two comorbidities often present with a higher severity and duration of symptoms [3]. Therefore, the diagnosis and treatment of these disorders remain a challenge in the clinical setting [1,4]. Despite being different disorders, the etiology of depression and anxiety involves similar factors, such as genetic predispositions, environmental aspects, and several biological mechanisms [1,2,5]. Among the main biological mechanisms implicated in the pathophysiology of these disorders, compelling evidence has pointed to neuroinflammation as a key factor in the onset and progression of these disorders [6]. Notably, other biological mechanisms that have been implicated in depression and anxiety, such as gut dysbiosis, impaired neurogenesis, and monoaminergic dysfunction, may be triggered by a neuroinflammatory process, opening new perspectives for studying molecular targets and neuroprotective agents against these mood disturbances [4,7,8,9]. In this regard, in recent years, vitamin D has gained prominence due to its antioxidant, anti-inflammatory, pro-neurogenic, and neuromodulatory properties that appear to be fundamental to its antidepressant and anxiolytic effects [10,11,12,13]. Given this background, in this review, we highlight the main mechanisms that may underlie the potential antidepressant and anxiolytic effects of vitamin D. In addition, we discuss preclinical and clinical studies that support the therapeutic potential of this vitamin for the management of these mood disorders.

2. Literature Data Searching

This review presents a mechanistic overview of the clinical and preclinical research regarding the effects of vitamin D on depression and anxiety. The search included original manuscripts and contemporary reviews published in English, assessed by specific search terms in the title or abstract of the manuscripts available through PubMed. The search terms used were “vitamin D”, “calcitriol”, “cholecalciferol”, “calcidiol”, “anxiety”, “depression”, “major depressive disorder”, “neuroinflammation”, “inflammation” “neurogenesis”, and “monoaminergic system”. We performed a specific screening of the clinical and preclinical studies that investigated the role of vitamin D in anxiety and depression. To review the molecular basis underlying the therapeutic potential of vitamin D for the treatment of depression and anxiety, we selected preclinical and clinical studies published over a 28-year period (1994 to 2022).

3. Neuroinflammation as a Key Pathophysiological Mechanism Related to Mood Disorders

Neuroinflammation is a complex process that comprises a defense mechanism in the central nervous system (CNS), protecting and restoring the structure and function of the brain against infection and injury, through the modulation of neurogenesis, axonal regeneration, and remyelination of neural cells [14]. However, chronic and exacerbated inflammatory responses can produce harmful effects in the brain. These inflammatory processes may involve inflammation-related signaling molecules, microbiota, as well as immune and brain cells [7,15].
Microglia, nervous-system-specific immune cells, are of special interest to neuroinflammatory responses. Under normal conditions, microglia assume a phenotype defined by a ramified morphology and highly motile processes for constant monitoring of the brain parenchyma (M2 phenotype). After an insult, promoted by pathogen-associated molecular patterns and/or damage-associated molecular patterns that interact with pattern recognition receptors (PRRs), such as Toll-like receptors (TLRs), microglia retract their processes and adopt an ameboid form (M1 phenotype) [16,17,18,19]. In addition to morphological changes, the binding of these molecular patterns to these receptors induces the priming of NLRP3 [nucleotide-binding oligomerization domain (NOD)-, leucine-rich repeats (LRR)- and pyrin domain-containing protein 3] and pro-interleukin (IL)-1β expression via nuclear factor kappa B (NF-kB) and myeloid differentiation primary response 88 (Myd88) pathways [17,20,21]. Noteworthily, the expression of NLRP3 may be inhibited under certain conditions, particularly upon activation of nuclear factor erythroid 2-related factor 2 (Nrf2), the main regulator of the antioxidant response [22,23]. Subsequently, different stimuli capable of promoting mitochondrial dysfunction, calcium and potassium ion flux, reactive oxygen species (ROS) production, and lysosomal damage activate the NLRP3 inflammasome, promoting the autoproteolytic activation of pro-caspase-1 [20]. Caspase-1 can subsequently cleave pro-interleukin-1β and pro-interleukin-18 into their active forms, interleukin-1β (IL-1β) and interleukin-18 (IL-18), respectively [17,24]. In addition, activation of NLRP3 can lead to gasdermin D-mediated formation of membrane pores and subsequently pyroptosis [17].
Mediators released by activated microglia induce astrocyte polarization [25]. This polarization contributes to the impairment of signaling pathways that play a crucial role in neuronal survival and synaptic plasticity, such as the brain-derived neurotrophic factor (BDNF)/tropomyosin-related kinase B (TrkB) signaling pathway [26]. Chronic activation of astrocytes also results in increased levels of chemokines, such as chemokine ligand 2, which interact with peripheral immune cell receptors to induce infiltration of macrophages and monocytes from the circulation into the CNS and resulting in increased blood–brain barrier permeability [27,28].
Interestingly, neuroinflammatory-process-derived pro-inflammatory cytokines, such as interferon-gamma (IFN-γ) and tumor necrosis factor-alpha (TNF-α), cause an increase in the activity of the enzyme indoleamine 2,3-dioxygenase in astrocytes, microglia, and inflammatory cells. The activation of this enzyme increases the formation of quinolinic acid, an N-methyl-D-aspartate (NMDA) receptor agonist, through the kynurenine pathway, contributing to glutamatergic system disturbance and compromising the synthesis of serotonin by depleting tryptophan [8,29,30,31]. Within this scenario, it is worth noting that more than 90% of the body’s serotonin is produced in the gut, particularly by enterochromaffin cells, and alterations in the intestinal microbiota triggered by inflammatory processes have been shown to directly compromise the synthesis of this monoamine [32,33]. Indeed, it is important to note that changes in gut microbiota have been reported to impair the efficacy of antidepressants, such as fluoxetine [34].
In summary, neuroinflammation results from intrinsic communication among peripheral immune cells, gut microbiota, and immune cells present in the brain, and these interactions culminate in glutamatergic dysregulation, as well as reduced synaptic plasticity and monoamine synthesis. Together, these factors contribute to the atrophy and neuronal loss seen in patients with chronic depression and anxiety disorders [35]. In this regard, compounds with anti-inflammatory, pro-neurogenic, and neuromodulatory properties, such as vitamin D, are of special interest for the treatment of these mood disorders.

4. Metabolism and Biological Functions of Vitamin D

Vitamin D is a steroid hormone that can be obtained from the diet in two forms: (i) vitamin D2 (ergocalciferol) found in yeast, mushrooms, as well as plants, and (ii) vitamin D3 (cholecalciferol) found in foods of animal origin, such as cod liver oil and oily fish [36]. However, the main source of vitamin D is cholecalciferol synthesized endogenously through solar ultraviolet B (UVB) radiation at wavelengths of 290–315 nm [36,37]. Through this process, UVB photons penetrate into the dermis and are absorbed by 7-dehydrocholesterol, which is then converted into previtamin D3 [37]. Previtamin D3 subsequently isomerizes into vitamin D3, which is then transported in the circulation by vitamin D-binding protein (DBP) [37]. In the liver, vitamin D3 is hydroxylated at C-25 by 25-hydroxylase, mainly by cytochrome P450 Family 2 Subfamily R Member 1 (CYP2R1), to produce 25-hydroxyvitamin D3 (25(OH)D3) [38,39]. Following this, 25(OH)D3 can be stored or can undergo a second hydroxylation in the kidneys by 25-hydroxyvitamin D-1α-hydroxylase (Cytochrome P450 Family 27 Subfamily B Member 1; CYP27B1), forming its biologically active form, 1α,25-dihydroxyvitamin D (1,25(OH)2D3), also known as calcitriol [40]. It is estimated that humans can synthesize quantities equivalent to the oral intake of 20,000 IU of this vitamin [38].
Vitamin D is widely known to play critical roles in calcium–phosphorus metabolism, acting in the maintenance of bone homeostasis [38]. In addition, vitamin D also exerts pleiotropic functions that have gained increased attention in the last few years [41]. These biological actions of calcitriol are mediated by vitamin D receptors (VDRs) [42]. The interaction between calcitriol and VDR allows other transcription factors, such as the retinoid X receptor (RXR), to interact with this complex, forming a heterodimer [42]. This heterodimer can then bind to vitamin D-responsive elements (VDREs), and by recruiting complexes of either co-activators or co-repressors, it can modulate gene expression [42,43]. It is estimated that 200 to 2000 genes are modulated directly or indirectly by vitamin D [42]. Due to the numerous biological actions mediated by vitamin D, the measurement of its serum concentration becomes indispensable. In this regard, the Endocrine Society’s Clinical Practice Guideline defines vitamin D deficiency as values <20 ng/mL; insufficiency between 21–29 ng/mL; and sufficiency between 30–100 ng/mL [36,40]. However, reference values for vitamin D remain a matter of debate [44].

4.1. Antioxidant and Anti-Inflammatory Properties of Vitamin D

Neurons and glial cells express VDR and vitamin D-metabolizing enzymes in various regions, such as the prefrontal cortex and hippocampus, suggesting a possible role for vitamin D in depression and anxiety disorders [45]. In line with this, several studies have suggested an inverse association between serum vitamin D levels and symptoms related to depression and anxiety [46,47,48]. In addition to glial cells, VDRs and CYP27B1 are also expressed in several other immune cells, such as CD4+ and CD8+ T cells, B cells, macrophages, neutrophils, and dendritic cells [49]. Notably, it has been shown that calcitriol is capable of modulating innate and adaptive immune responses [49]. Furthermore, the positive modulation of antioxidant enzymes by calcitriol is known to contribute to redox homeostasis and, consequently, to the attenuation of the neuroinflammatory process [50,51]. For example, calcitriol has been shown to inhibit the synthesis of interleukin interleukin-6 (IL-6), TNF-α, and nitric oxide (NO) by activated microglia [12]. Likewise, it has been shown to decrease the expression of IL-1β, TNF-α, and inducible nitric oxide synthase (iNOS), the enzyme responsible for the synthesis of NO under inflammatory conditions, in the prefrontal cortex of rats [52].
In addition to IL-6 and TNF-α, vitamin D3 has also been shown to reduce the expression of interleukin-12 (IL-12) and increase the expression of the anti-inflammatory cytokine interleukin-10 (IL-10) [53]. Furthermore, vitamin D was shown to up-regulate the expression of other anti-inflammatory cytokines, including interleukin-14 (IL-4), and transforming growth factor-beta (TGF-β) [54]. Calcitriol is also capable of up-regulating TLR10 while down-regulating TLR2 and TLR4, reinforcing its role in attenuating microglial activation [55,56].
In this context, it has been reported that vitamin D mitigates the activation of the NLRP3 inflammasome [57,58]. Although a study by Camargo et al. (2020) did not find changes in NLRP3 expression in mice treated for 7 days with cholecalciferol, this treatment reduced the immunocontent of other inflammasome-related proteins, including ASC (apoptosis-associated speck-like protein containing a caspase recruitment domain), TXNIP (thioredoxin interacting protein), and caspase-1 in the hippocampus of mice. However, Xin et al. (2019) showed that calcitriol attenuated NLRP3 expression by inhibiting the NF-kB pathway in human bronchial epithelial cells. Likewise, treatment with a VDR agonist (paricalcitol) was able to attenuate the expression of NLRP3, gasdermin D, caspase-1, and IL-1β, proteins related to the NF-kB-mediated pyroptosis process in cisplatin-induced acute kidney injury models [59]. One of the possible mechanisms by which vitamin D may reduce the activation of NF-kB is by up-regulating the NF-kB inhibitor IkBα (nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor, alpha) [60]. In addition, it has been reported that VDR can bind NLRP3, inhibiting the activation of this complex [61,62].
Calcitriol is also able of suppressing NLRP1 inflammasome activation by up-regulating the nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway [50]. Indeed, the ability of calcitriol to activate Nrf2 signaling seems to be directly associated with its neuroprotective properties [63]. Once activated, Nrf2 promotes the transcription of several antioxidant enzymes [64]. In line with this, studies have shown that vitamin D is able to induce the expression of heme oxygenase-1 (HO-1), catalase, superoxide dismutase (SOD), and glutamate-cysteine ligase (the rate-limiting enzyme in the synthesis of glutathione, an endogenous antioxidant), thereby inhibiting ROS accumulation in the brain [51,65,66,67].

4.2. Pro-Neurogenic and Neuromodulatory Properties of Vitamin D

Vitamin D deficiency during pregnancy has been demonstrated to directly affect brain development in the offspring [68,69,70]. Vitamin D-deficient offspring exhibit a thinner cortex, reduced expression of genes and proteins related to cytoskeleton maintenance, synaptic plasticity, neurotransmission, cell proliferation, and growth, as well as decreased levels of nerve growth factor (NGF) [70,71,72,73]. Although no alterations in BDNF and glial cell line-derived neurotrophic factor (GDNF) levels were found in vitamin D-deficient offspring, different lines of evidence have shown that this vitamin is effective in inducing the synthesis of these neurotrophins [74,75]. In addition, vitamin D also participates in the synthesis of the neurotrophin NT-3, a key regulator of survival, growth, and differentiation of neurons [76,77].
Indeed, neurotrophins bind to receptor tyrosine kinases (Trk), which, in turn, contribute to promoting the survival and growth of neurons as well as synaptogenesis [78]. In this regard, a study by Atif et al. showed that a combined treatment with progesterone and vitamin D was effective in activating the BDNF/TrkB signaling pathway [79]. Although this study did not explore the mechanisms underlying this, it is known that activated TrkB can phosphorylate and activate the mammalian target of rapamycin (mTOR), resulting in the translation of synaptic proteins, including postsynaptic density protein-95 kDa (PSD-95), glutamate α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor subunit 1 (GluA1), and synapsin [80,81]. Noteworthily, synapsins can increase neurotransmitter release in the synaptic cleft [82]. Interestingly, vitamin D supplementation has been shown to promote an increase in serum serotonin in individuals suffering from depression [83] as well as serum dopamine levels in children with attention-deficit/hyperactivity disorder (ADHD) [84]. Within this scenario, it has been shown that vitamin D can directly increase the biosynthesis of monoamines by augmenting the expression of the enzymes tyrosine hydroxylase and tryptophan hydroxylase, which mediate the synthesis of dopamine/noradrenaline and serotonin, respectively, particularly in the prefrontal cortex [85]. In addition, VDR overexpression has been shown to increase dopaminergic neuron differentiation, and consequently, dopamine production [86]. Moreover, Morello et al. (2018) showed that calcitriol can cause neuronal proliferation in primary cultures of murine neural progenitor cells and improve neurogenesis in transgenic Alzheimer’s disease (AD)-like mice [5XFAD model, which expresses human amyloid precursor protein (APP) and presenilin 1 (PSEN1) transgenes with a total of five AD-linked mutations: the Swedish (K670N/M671L), Florida (I716V), and London (V717I) mutations in APP, and the M146L and L286V mutations in PSEN1). Finally, calcitriol was also able to inhibit serotonin reuptake transport and the expression of the monoamine oxidase-A (MAO-A) gene, suggesting that this vitamin and antidepressants may share common mechanisms of action [87].

4.3. Vitamin D: Modulation of Gut Microbiota

The immunological and neuromodulatory role of vitamin D occurs, in part, through its interaction with the gut microbiota [88]. Indeed, serum vitamin D concentration may contribute to changes in the composition of gut microbiota. Furthermore, vitamin D deficiency, VDR, or CYP27B1 depletion have been shown to result in an increase in the Bacteroidetes and Proteobacteria phyla, triggering epithelial barrier dysfunction and intestinal inflammation [89,90]. Interestingly, VDR has been shown to negatively regulate bacteria-induced intestinal NF-kB activation, attenuating inflammatory responses. Therefore, VDR is an important contributor to intestinal homeostasis and bacterial infection protection [91]. The main neuromodulatory, anti-inflammatory, and antioxidant properties of vitamin D are illustrated in Figure 1.

5. Preclinical Studies: Effects of Vitamin D in Models of Depression and Anxiety

In recent years, several preclinical studies have been conducted to investigate the possible antidepressant and anxiolytic effects of vitamin D (Table 1).
Of note, chronic administration of cholecalciferol [5 mg/kg for 14 days; subcutaneous (s.c.) administration] elicited an antidepressant-like effect in the forced swim test in ovariectomized Wistar rats [92]. In addition, cholecalciferol supplementation (5 mg/kg for 14 days; s.c.) was capable of attenuating anxiety-like behaviors in the elevated plus-maze and the light–dark box tests in ovariectomized Wistar rats [10,93]. Based on these findings, further studies have been conducted to better understand the mechanisms underlying the anxiolytic and antidepressant effects of vitamin D in animal models [94,98].
Camargo et al. (2018) reported that cholecalciferol [2.5 µg/kg, orally by mouth (p.o.)], administered once a day in the last 7 days of chronic corticosterone administration (20 mg/kg, p.o., for 21 days), exerted an antidepressant-like effect in male mice subjected to the splash test and tail suspension test. Additionally, in this study, cholecalciferol treatment attenuated the increase in protein carbonyl and nitrite levels induced by corticosterone in the brain, suggesting that vitamin D3 has an antidepressant-like effect by, in part, modulating oxidative stress [94]. The administration of cholecalciferol (100 IU/kg, p.o.) for 7 days also abolished the depressive-like behavior in the tail suspension test induced by chronic corticosterone administration in female mice [95]. In this study, a significant decrease in ROS production in the hippocampus was observed after treatment with cholecalciferol, both in control and corticosterone-exposed mice, reinforcing the notion that the antidepressant-like effect of this vitamin may involve the modulation of oxidative stress [95]. More recently, a study by Neis et al. (2022) showed that repeated administration of cholecalciferol for 7 days (2.5 μg/kg, p.o.) abolished chronic unpredictable stress-induced depressive-like behavior in the tail suspension test, as well as a reduction in serotonin levels in the prefrontal cortex of female mice. Moreover, reinforcing the involvement of the serotonergic system in the antidepressant-like effect of cholecalciferol, in this study, the administration of the serotonin synthesis inhibitor p-chlorophenylalanine methyl ester was effective in abolishing the reduction in immobility time in the tail suspension test elicited by cholecalciferol [96].
The repeated administration of a low dose of cholecalciferol (2.5 μg/kg, p.o.) also caused an antidepressant-like effect and was effective in reducing the immunocontent of proteins that form the NLRP3 inflammasome, such as ASC [apoptosis-associated speck-like protein containing a caspase recruitment domain (CARD)], caspase-1, and thioredoxin-interacting protein (TXNIP) in the hippocampus of male mice [57]. Calcitriol treatment (100 ng/kg, p.o.) for 10 weeks in ovariectomized female Sprague–Dawley rats was also effective in producing neuroprotective effects by regulating the adenosine monophosphate (AMP)-activated protein kinase (AMPK)/NF-kB signaling pathway. Moreover, calcitriol treatment reduced the pro-inflammatory cytokines IL-1β, IL-6, and TNF-α, as well as iNOS and cyclooxygenase-2 (COX-2) levels in the hippocampus [97]. This suggests that the modulation of the NLRP3 inflammasome-driven pathway may underlie, at least in part, the antidepressant-like effect of this vitamin.
More recently, Bakhtiari-Dovvombaygi et al. (2021) also reported that the anti-inflammatory and antioxidant effects displayed by pretreatment with vitamin D3 (10,000 IU/kg for 28 days) in male rats underlie the ability of this vitamin to abrogate anxiety- and depressive-like behaviors induced by chronic unpredictable mild stress (CUMS) in the elevated plus-maze and forced swimming test. Indeed, these protective effects of vitamin D were accompanied by a decrease in cortical malondialdehyde and IL-6 levels, as well as an increase in total thiol levels and enhanced SOD and catalase activity [98]. Interestingly, another study observed that following 4 weeks of CUMS, the occurrence of depressive-like behaviors was associated with an increase in 1,25(OH)2D and VDR expression in the hippocampus of rats, suggesting a compensatory mechanism, by which vitamin D may protect against the development of depressive-like behaviors [99].
In addition to attenuating depressive- and anxiety-like behaviors through its anti-inflammatory and antioxidant properties, the modulation of neurotrophic factors has also been shown to contribute to the protective properties of vitamin D [76]. Xu et al. used male C57BL/6 mice to show that calcitriol (25 μg/kg/day for 4 weeks; i.c.v.) is effective in acting as an antidepressant in a post-stroke depression model by up-regulating VDR and BDNF expression [75]. In an ovariectomized Wistar rat model of depression induced by CUMS, vitamin D3 (5 mg/kg for 4 weeks; s.c.) treatment was able to reverse depression-like behaviors in the sucrose preference test and the forced swimming test by increasing BDNF and NT-3/NT-4 levels in the hippocampus [76]. Although these studies showed that vitamin D supplementation results in an increase in pro-neurogenic neurotrophins, such as BDNF, Groves et al. observed that vitamin D deficiency in BALB/c mice was associated with depressive-like behaviors without compromising hippocampal neurogenesis [100]. Therefore, further studies are needed to elucidate whether neurogenesis is critical for the anxiolytic and antidepressant effects of this vitamin.

6. Clinical Studies: Effects of Vitamin D in Depression and Anxiety

Several studies have reported that vitamin D supplementation improves symptoms of depression and anxiety associated with various medical conditions, including type II diabetes, Crohn’s disease, ulcerative colitis, and obesity [101,102,103,104,105].
However, the potential therapeutic effects of vitamin D in individuals primarily diagnosed with depression or anxiety remain controversial. For example, vitamin D supplementation (1600 IU for 6 months) was shown to significantly improve anxiety symptoms, but not depressive symptoms, in patients with vitamin D deficiency [106]. Likewise, supplementation with 2800 IU of vitamin D in patients with depression did not promote a significant reduction in Hamilton D-17 scores [107]. On the other hand, supplementation with 50,000 IU of vitamin D for 2 weeks was able to improve depression severity, as assessed with the Beck Depression Inventory-II (BDI-II), although no changes in serotonin levels were detected [108]. However, in another study, cholecalciferol treatment (50,000 IU for 3 months) significantly increased serum serotonin levels, while decreasing BDI scores in women with moderate, severe, and extreme depression. Interestingly, among men, an improvement in the severity of depressive symptoms with vitamin D supplementation was only observed in those diagnosed with severe depression [83]. Beneficial effects of vitamin D (50,000 IU for 8 weeks) supplementation have also been observed in older adults (over 60 years of age) with depression [109]. However, lower doses of vitamin D (400 IU daily for 2 years) were not able to improve depressive symptoms [110]. Finally, a single dose of vitamin D (300,000 IU) was reported as an effective and safe intervention in MDD with concurrent vitamin D deficiency [111,112]. In patients with depression, the daily administration of 1500 IU vitamin D3 plus 20 mg fluoxetine for 8 weeks was superior to fluoxetine alone [113]. Another study reported that vitamin D supplementation (50,000 IU once/week for 3 months) in combination with standard of care improved the severity of anxiety in individuals diagnosed with Generalized Anxiety Disorder by increasing serotonin concentrations and decreasing the levels of the inflammatory biomarker neopterin [114]. Overall, although there is compelling clinical evidence pointing to the benefits of vitamin D for the management of depression and anxiety, it is important to note that divergent results have also been obtained [46]. Multiple factors may contribute to these discrepant results, including differences in the doses of vitamin D used, treatment time, serum 25-hydroxyvitamin D levels at baseline, nutritional condition at the onset of the treatment, age and sex of the individuals, as well as the presence of comorbidities that may influence the efficacy of vitamin D supplementation. Additionally, the heterogeneity observed in clinical studies may also be associated with genetic polymorphisms that may affect vitamin D efficacy [115,116,117].

7. Conclusions and Future Directions

In this review, we discussed the main evidence underlining the therapeutic potential of vitamin D in the management of depression and anxiety disorders. Of particular relevance, compelling evidence suggests that vitamin D possesses antioxidant, anti-inflammatory, pro-neurogenic, and neuromodulatory properties, and, thus, may act in a similar manner to classic antidepressants. In support, several preclinical studies have shown the beneficial effects of vitamin D supplementation in animal models of these mood disorders. Given this, some studies have investigated the possible synergistic or additive beneficial effects of vitamin D and current pharmacotherapy for the management of anxiety and depression. However, the clinical studies that assess the efficacy of vitamin D supplementation in the treatment of depression and anxiety are still scarce and their results are, at times, controversial. The discrepant results from clinical trials call for the need to conduct future studies, so as to establish a protocol for vitamin D supplementation that is effective in preventing or attenuating depressive and anxiety symptoms. Therefore, appropriately randomized clinical trials assessing the potential of vitamin D as co-adjuvant for the treatment of these mood disorders are warranted, so as to ascertain the true therapeutic value of this vitamin in the context of depression and anxiety.

Author Contributions

B.R.K.: conceptualization and writing of original manuscript draft. A.C.: writing of original manuscript draft. J.G.-M.: review and editing of the manuscript and funding acquisition for manuscript publication. A.L.S.R.: conceptualization, supervision, review and editing of the manuscript, and funding acquisition. All authors have read and agreed to the published version of the manuscript.

Funding

The authors acknowledge funding from Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq; #421143/2018-5 and #312215/2021-5) and Coordenação de Aperfeiçoamento de Pessoal de Nível Superior (CAPES). J.G.M. acknowledges funding from the University of Victoria (UVic, Victoria, BC, Canada)—São Paulo Research Foundation (FAPESP, São Paulo, SP, Brazil) SPRINT partnership (UVic-FAPESP SPRINT 1/2018 Grant). A.L.S.R. is CNPq Research Fellow.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Craske, M.G.; Stein, M.B.; Eley, T.C.; Milad, M.R.; Holmes, A.; Rapee, R.M.; Wittchen, H.U. Anxiety Disorders. Nat. Rev. Dis. Primers 2017, 3, 17024. [Google Scholar] [CrossRef] [PubMed]
  2. Otte, C.; Gold, S.M.; Penninx, B.W.; Pariante, C.M.; Etkin, A.; Fava, M.; Mohr, D.C.; Schatzberg, A.F. Major Depressive Disorder. Nat. Rev. Dis. Primers 2016, 2, 16065. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Lamers, F.; Van Oppen, P.; Comijs, H.C.; Smit, J.H.; Spinhoven, P.; van Balkom, A.J.L.M.; Nolen, W.A.; Zitman, F.G.; Beekman, A.T.F.; Penninx, B.W.J.H. Comorbidity Patterns of Anxiety and Depressive Disorders in a Large Cohort Study: The Netherlands Study of Depression and Anxiety (NESDA). J. Clin. Psychiatry 2011, 72, 341–348. [Google Scholar] [CrossRef]
  4. Malhi, G.S.; Mann, J.J. Depression. Lancet 2018, 392, 2299–2312. [Google Scholar] [CrossRef]
  5. Schiele, M.A.; Domschke, K. Epigenetics at the Crossroads between Genes, Environment and Resilience in Anxiety Disorders: Epigenetics in Anxiety Disorders. Genes Brain Behav. 2018, 17, e12423. [Google Scholar] [CrossRef] [Green Version]
  6. De Haan, P.; Klein, H.C.; ’t Hart, B.A. Autoimmune Aspects of Neurodegenerative and Psychiatric Diseases: A Template for Innovative Therapy. Front. Psychiatry 2017, 8, 46. [Google Scholar] [CrossRef] [Green Version]
  7. Carlessi, A.S.; Borba, L.A.; Zugno, A.I.; Quevedo, J.; Réus, G.Z. Gut Microbiota-Brain Axis in Depression: The Role of Neuroinflammation. Eur. J. Neurosci. 2021, 53, 222–235. [Google Scholar] [CrossRef]
  8. Kim, Y.K.; Jeon, S.W. Neuroinflammation and the Immune-Kynurenine Pathway in Anxiety Disorders. Curr. Neuropharmacol. 2018, 16, 574–582. [Google Scholar] [CrossRef]
  9. Stevens, B.R.; Goel, R.; Seungbum, K.; Richards, E.M.; Holbert, R.C.; Pepine, C.J.; Raizada, M.K. Increased Human Intestinal Barrier Permeability Plasma Biomarkers Zonulin and FABP2 Correlated with Plasma LPS and Altered Gut Microbiome in Anxiety or Depression. Gut 2018, 67, 1555–1557. [Google Scholar] [CrossRef]
  10. Fedotova, J.; Zarembo, D.; Dragasek, J.; Caprnda, M.; Kruzliak, P.; Dudnichenko, T. Modulating Effects of Cholecalciferol Treatment on Estrogen Deficiency-Induced Anxiety-Like Behavior of Adult Female Rats. Folia Med. 2017, 59, 139–158. [Google Scholar] [CrossRef]
  11. Khairy, E.Y.; Attia, M.M. Protective Effects of Vitamin D on Neurophysiologic Alterations in Brain Aging: Role of Brain-Derived Neurotrophic Factor (BDNF). Nutr. Neurosci. 2021, 24, 650–659. [Google Scholar] [CrossRef] [PubMed]
  12. Lefebvre d’Hellencourt, C.; Montero-Menei, C.N.; Bernard, R.; Couez, D. Vitamin D3 Inhibits Proinflammatory Cytokines and Nitric Oxide Production by the EOC13 Microglial Cell Line. J. Neurosci. Res. 2003, 71, 575–582. [Google Scholar] [CrossRef] [PubMed]
  13. Morello, M.; Landel, V.; Lacassagne, E.; Baranger, K.; Annweiler, C.; Féron, F.; Millet, P. Vitamin D Improves Neurogenesis and Cognition in a Mouse Model of Alzheimer’s Disease. Mol. Neurobiol. 2018, 55, 6463–6479. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Yong, H.Y.F.; Rawji, K.S.; Ghorbani, S.; Xue, M.; Yong, V.W. The Benefits of Neuroinflammation for the Repair of the Injured Central Nervous System. Cell. Mol. Immunol. 2019, 16, 540–546. [Google Scholar] [CrossRef] [PubMed]
  15. Fullerton, J.N.; Gilroy, D.W. Resolution of Inflammation: A New Therapeutic Frontier. Nat. Rev. Drug Discov. 2016, 15, 551–567. [Google Scholar] [CrossRef]
  16. Barton, G.M. A Calculated Response: Control of Inflammation by the Innate Immune System. J. Clin. Investig. 2008, 118, 413–420. [Google Scholar] [CrossRef]
  17. Herman, F.J.; Pasinetti, G.M. Principles of Inflammasome Priming and Inhibition: Implications for Psychiatric Disorders. Brain. Behav. Immun. 2018, 73, 66–84. [Google Scholar] [CrossRef]
  18. Orihuela, R.; McPherson, C.A.; Harry, G.J. Microglial M1/M2 Polarization and Metabolic States: Microglia Bioenergetics with Acute Polarization. Br. J. Pharmacol. 2016, 173, 649–665. [Google Scholar] [CrossRef]
  19. Rudzki, L.; Maes, M. The Microbiota-Gut-Immune-Glia (MGIG) Axis in Major Depression. Mol. Neurobiol. 2020, 57, 4269–4295. [Google Scholar] [CrossRef]
  20. Kelley, N.; Jeltema, D.; Duan, Y.; He, Y. The NLRP3 Inflammasome: An Overview of Mechanisms of Activation and Regulation. Int. J. Mol. Sci. 2019, 20, 3328. [Google Scholar] [CrossRef] [Green Version]
  21. Song, N.; Li, T. Regulation of NLRP3 Inflammasome by Phosphorylation. Front. Immunol. 2018, 9, 2305. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Liu, X.; Zhang, X.; Ding, Y.; Zhou, W.; Tao, L.; Lu, P.; Wang, Y.; Hu, R. Nuclear Factor E2-Related Factor-2 Negatively Regulates NLRP3 Inflammasome Activity by Inhibiting Reactive Oxygen Species-Induced NLRP3 Priming. Antioxid. Redox Signal. 2017, 26, 28–43. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Xu, X.; Zhang, L.; Ye, X.; Hao, Q.; Zhang, T.; Cui, G.; Yu, M. Nrf2/ARE Pathway Inhibits ROS-Induced NLRP3 Inflammasome Activation in BV2 Cells after Cerebral Ischemia Reperfusion. Inflamm. Res. 2018, 67, 57–65. [Google Scholar] [CrossRef] [PubMed]
  24. Sutterwala, F.S.; Haasken, S.; Cassel, S.L. Mechanism of NLRP3 Inflammasome Activation. Ann. N. Y. Acad. Sci. 2014, 1319, 82–95. [Google Scholar] [CrossRef]
  25. Liddelow, S.A.; Guttenplan, K.A.; Clarke, L.E.; Bennett, F.C.; Bohlen, C.J.; Schirmer, L.; Bennett, M.L.; Münch, A.E.; Chung, W.S.; Peterson, T.C.; et al. Neurotoxic Reactive Astrocytes Are Induced by Activated Microglia. Nature 2017, 541, 481–487. [Google Scholar] [CrossRef]
  26. Tejeda, G.; Díaz-Guerra, M. Integral Characterization of Defective BDNF/TrkB Signalling in Neurological and Psychiatric Disorders Leads the Way to New Therapies. Int. J. Mol. Sci. 2017, 18, 268. [Google Scholar] [CrossRef] [Green Version]
  27. Farina, C.; Aloisi, F.; Meinl, E. Astrocytes Are Active Players in Cerebral Innate Immunity. Trends Immunol. 2007, 28, 138–145. [Google Scholar] [CrossRef]
  28. Troubat, R.; Barone, P.; Leman, S.; Desmidt, T.; Cressant, A.; Atanasova, B.; Brizard, B.; El Hage, W.; Surget, A.; Belzung, C.; et al. Neuroinflammation and Depression: A Review. Eur. J. Neurosci. 2021, 53, 151–171. [Google Scholar] [CrossRef]
  29. Bansal, Y.; Singh, R.; Parhar, I.; Kuhad, A.; Soga, T. Quinolinic Acid and Nuclear Factor Erythroid 2-Related Factor 2 in Depression: Role in Neuroprogression. Front. Pharmacol. 2019, 10, 452. [Google Scholar] [CrossRef]
  30. Braidy, N.; Grant, R.; Brew, B.J.; Adams, S.; Jayasena, T.; Guillemin, G.J. Effects of Kynurenine Pathway Metabolites on Intracellular NAD + Synthesis and Cell Death in Human Primary Astrocytes and Neurons. Int. J. Tryptophan Res. 2009, 2, 61–69. [Google Scholar] [CrossRef]
  31. Pierozan, P.; Biasibetti, H.; Schmitz, F.; Ávila, H.; Parisi, M.M.; Barbe-Tuana, F.; Wyse, A.T.S.; Pessoa-Pureur, R. Quinolinic Acid Neurotoxicity: Differential Roles of Astrocytes and Microglia via FGF-2-Mediated Signaling in Redox-Linked Cytoskeletal Changes. Biochim. Biophys. Acta BBA-Mol. Cell Res. 2016, 1863, 3001–3014. [Google Scholar] [CrossRef]
  32. Agus, A.; Planchais, J.; Sokol, H. Gut Microbiota Regulation of Tryptophan Metabolism in Health and Disease. Cell Host Microbe 2018, 23, 716–724. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Chen, L.M.; Bao, C.H.; Wu, Y.; Liang, S.H.; Wang, D.; Wu, L.Y.; Huang, Y.; Liu, H.R.; Wu, H.G. Tryptophan-Kynurenine Metabolism: A Link between the Gut and Brain for Depression in Inflammatory Bowel Disease. J. Neuroinflammation 2021, 18, 135. [Google Scholar] [CrossRef] [PubMed]
  34. Siopi, E.; Chevalier, G.; Katsimpardi, L.; Saha, S.; Bigot, M.; Moigneu, C.; Eberl, G.; Lledo, P.M. Changes in Gut Microbiota by Chronic Stress Impair the Efficacy of Fluoxetine. Cell Rep. 2020, 30, 3682–3690.e6. [Google Scholar] [CrossRef] [PubMed]
  35. Duman, R.S. Neuronal Damage and Protection in the Pathophysiology and Treatment of Psychiatric Illness: Stress and Depression. Dialogues Clin. Neurosci. 2009, 11, 239–255. [Google Scholar] [CrossRef] [PubMed]
  36. 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]
  37. Holick, M.F. Vitamin D: A D-Lightful Solution for Health. J. Investig. Med. 2011, 59, 872–880. [Google Scholar] [CrossRef]
  38. Holick, M.F. Vitamin D Deficiency. N. Engl. J. Med. 2007, 357, 266–281. [Google Scholar] [CrossRef]
  39. Zhu, J.; DeLuca, H.F. Vitamin D 25-Hydroxylase—Four Decades of Searching, Are We There Yet? Arch. Biochem. Biophys. 2012, 523, 30–36. [Google Scholar] [CrossRef]
  40. 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]
  41. Christakos, S.; Dhawan, P.; Verstuyf, A.; Verlinden, L.; Carmeliet, G. Vitamin D: Metabolism, Molecular Mechanism of Action, and Pleiotropic Effects. Physiol. Rev. 2016, 96, 365–408. [Google Scholar] [CrossRef] [PubMed]
  42. Wacker, M.; Holick, M. Vitamin D—Effects on Skeletal and Extraskeletal Health and the Need for Supplementation. Nutrients 2013, 5, 111–148. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Haussler, M.R.; Whitfield, G.K.; Kaneko, I.; Haussler, C.A.; Hsieh, D.; Hsieh, J.-C.; Jurutka, P.W. Molecular Mechanisms of Vitamin D Action. Calcif. Tissue Int. 2013, 92, 77–98. [Google Scholar] [CrossRef] [PubMed]
  44. Alshahrani, F.; Aljohani, N. Vitamin D: Deficiency, Sufficiency and Toxicity. Nutrients 2013, 5, 3605–3616. [Google Scholar] [CrossRef] [Green Version]
  45. Eyles, D.W.; Smith, S.; Kinobe, R.; Hewison, M.; McGrath, J.J. Distribution of the Vitamin D Receptor and 1α-Hydroxylase in Human Brain. J. Chem. Neuroanat. 2005, 29, 21–30. [Google Scholar] [CrossRef]
  46. Casseb, G.A.S.; Kaster, M.P.; Rodrigues, A.L.S. Potential Role of Vitamin D for the Management of Depression and Anxiety. CNS Drugs 2019, 33, 619–637. [Google Scholar] [CrossRef]
  47. Hoogendijk, W.J.G.; Lips, P.; Dik, M.G.; Deeg, D.J.H.; Beekman, A.T.F.; Penninx, B.W.J.H. Depression Is Associated With Decreased 25-Hydroxyvitamin D and Increased Parathyroid Hormone Levels in Older Adults. Arch. Gen. Psychiatry 2008, 65, 508. [Google Scholar] [CrossRef]
  48. Pu, D.; Luo, J.; Wang, Y.; Ju, B.; Lv, X.; Fan, P.; He, L. Prevalence of Depression and Anxiety in Rheumatoid Arthritis Patients and Their Associations with Serum Vitamin D Level. Clin. Rheumatol. 2018, 37, 179–184. [Google Scholar] [CrossRef]
  49. Baeke, F.; Takiishi, T.; Korf, H.; Gysemans, C.; Mathieu, C. Vitamin D: Modulator of the Immune System. Curr. Opin. Pharmacol. 2010, 10, 482–496. [Google Scholar] [CrossRef]
  50. Nakajo, T.; Katayoshi, T.; Kitajima, N.; Tsuji-Naito, K. 1,25-Dihydroxyvitamin D3 Attenuates IL-1β Secretion by Suppressing NLRP1 Inflammasome Activation by Upregulating the NRF2-HO-1 Pathway in Epidermal Keratinocytes. Redox Biol. 2021, 48, 102203. [Google Scholar] [CrossRef]
  51. Yamini, P.; Ray, R.S.; Chopra, K. Vitamin D3 Attenuates Cognitive Deficits and Neuroinflammatory Responses in ICV-STZ Induced Sporadic Alzheimer’s Disease. Inflammopharmacology 2018, 26, 39–55. [Google Scholar] [CrossRef]
  52. Cui, C.; Xu, P.; Li, G.; Qiao, Y.; Han, W.; Geng, C.; Liao, D.; Yang, M.; Chen, D.; Jiang, P. Vitamin D Receptor Activation Regulates Microglia Polarization and Oxidative Stress in Spontaneously Hypertensive Rats and Angiotensin II-Exposed Microglial Cells: Role of Renin-Angiotensin System. Redox Biol. 2019, 26, 101295. [Google Scholar] [CrossRef] [PubMed]
  53. Boontanrart, M.; Hall, S.D.; Spanier, J.A.; Hayes, C.E.; Olson, J.K. Vitamin D3 Alters Microglia Immune Activation by an IL-10 Dependent SOCS3 Mechanism. J. Neuroimmunol. 2016, 292, 126–136. [Google Scholar] [CrossRef] [PubMed]
  54. Calvello, R.; Cianciulli, A.; Nicolardi, G.; De Nuccio, F.; Giannotti, L.; Salvatore, R.; Porro, C.; Trotta, T.; Panaro, M.A.; Lofrumento, D.D. Vitamin D Treatment Attenuates Neuroinflammation and Dopaminergic Neurodegeneration in an Animal Model of Parkinson’s Disease, Shifting M1 to M2 Microglia Responses. J. Neuroimmune Pharmacol. 2017, 12, 327–339. [Google Scholar] [CrossRef] [PubMed]
  55. Verma, R.; Jung, J.H.; Kim, J.Y. 1,25-Dihydroxyvitamin D3 up-Regulates TLR10 While down-Regulating TLR2, 4, and 5 in Human Monocyte THP-1. J. Steroid Biochem. Mol. Biol. 2014, 141, 1–6. [Google Scholar] [CrossRef]
  56. Verma, R.; Kim, J.Y. 1,25-Dihydroxyvitamin D3 Facilitates M2 Polarization and Upregulates TLR10 Expression on Human Microglial Cells. Neuroimmunomodulation 2016, 23, 75–80. [Google Scholar] [CrossRef]
  57. Camargo, A.; Dalmagro, A.P.; Platt, N.; Rosado, A.F.; Neis, V.B.; Zeni, A.L.B.; Kaster, M.P.; Rodrigues, A.L.S. Cholecalciferol Abolishes Depressive-like Behavior and Hippocampal Glucocorticoid Receptor Impairment Induced by Chronic Corticosterone Administration in Mice. Pharmacol. Biochem. Behav. 2020, 196, 172971. [Google Scholar] [CrossRef]
  58. Xin, L.; Che, B.; Zhai, B.; Luo, Q.; Zhang, C.; Wang, J.; Wang, S.; Fan, G.; Liu, Z.; Feng, J.; et al. 1,25-Dihydroxy Vitamin D3 Attenuates the Oxidative Stress-Mediated Inflammation Induced by PM2.5via the P38/NF-ΚB/NLRP3 Pathway. Inflammation 2019, 42, 702–713. [Google Scholar] [CrossRef]
  59. Jiang, S.; Zhang, H.; Li, X.; Yi, B.; Huang, L.; Hu, Z.; Li, A.; Du, J.; Li, Y.; Zhang, W. Vitamin D/VDR Attenuate Cisplatin-Induced AKI by down-Regulating NLRP3/Caspase-1/GSDMD Pyroptosis Pathway. J. Steroid Biochem. Mol. Biol. 2021, 206, 105789. [Google Scholar] [CrossRef]
  60. Cohen-Lahav, M.; Shany, S.; Tobvin, D.; Chaimovitz, C.; Douvdevani, A. Vitamin D Decreases NFκB Activity by Increasing IκBα Levels. Nephrol. Dial. Transplant. 2006, 21, 889–897. [Google Scholar] [CrossRef] [Green Version]
  61. Huang, H.; Hong, J.Y.; Wu, Y.J.; Wang, E.Y.; Liu, Z.Q.; Cheng, B.H.; Mei, L.; Liu, Z.G.; Yang, P.C.; Zheng, P.Y. Vitamin D Receptor Interacts with NLRP3 to Restrict the Allergic Response. Clin. Exp. Immunol. 2018, 194, 17–26. [Google Scholar] [CrossRef] [Green Version]
  62. Rao, Z.; Chen, X.; Wu, J.; Xiao, M.; Zhang, J.; Wang, B.; Fang, L.; Zhang, H.; Wang, X.; Yang, S.; et al. Vitamin D Receptor Inhibits NLRP3 Activation by Impeding Its BRCC3-Mediated Deubiquitination. Front. Immunol. 2019, 10, 2783. [Google Scholar] [CrossRef]
  63. Cui, C.; Wang, C.; Jin, F.; Yang, M.; Kong, L.; Han, W.; Jiang, P. Calcitriol Confers Neuroprotective Effects in Traumatic Brain Injury by Activating Nrf2 Signaling through an Autophagy-Mediated Mechanism. Mol. Med. 2021, 27, 118. [Google Scholar] [CrossRef] [PubMed]
  64. Fão, L.; Mota, S.I.; Rego, A.C. Shaping the Nrf2-ARE-Related Pathways in Alzheimer’s and Parkinson’s Diseases. Ageing Res. Rev. 2019, 54, 100942. [Google Scholar] [CrossRef] [PubMed]
  65. Ali, A.; Shah, S.A.; Zaman, N.; Uddin, M.N.; Khan, W.; Ali, A.; Riaz, M.; Kamil, A. Vitamin D Exerts Neuroprotection via SIRT1/Nrf-2/NF-KB Signaling Pathways against D-Galactose-Induced Memory Impairment in Adult Mice. Neurochem. Int. 2021, 142, 104893. [Google Scholar] [CrossRef] [PubMed]
  66. Mehrabadi, S.; Sadr, S.S. Administration of Vitamin D3 and E Supplements Reduces Neuronal Loss and Oxidative Stress in a Model of Rats with Alzheimer’s Disease. Neurol. Res. 2020, 42, 862–868. [Google Scholar] [CrossRef] [PubMed]
  67. Shinpo, K.; Kikuchi, S.; Sasaki, H.; Moriwaka, F.; Tashiro, K. Effect of 1,25-Dihydroxyvitamin D3 on Cultured Mesencephalic Dopaminergic Neurons to the Combined Toxicity Caused by L-Buthionine Sulfoximine and 1-Methyl-4-Phenylpyridine. J. Neurosci. Res. 2000, 62, 374–382. [Google Scholar] [CrossRef]
  68. Cui, X.; McGrath, J.J.; Burne, T.H.J.; Mackay-Sim, A.; Eyles, D.W. Maternal Vitamin D Depletion Alters Neurogenesis in the Developing Rat Brain. Int. J. Dev. Neurosci. 2007, 25, 227–232. [Google Scholar] [CrossRef]
  69. Zhu, Y.; Zhou, R.; Yang, R.; Zhang, Z.; Bai, Y.; Chang, F.; Li, L.; Sokabe, M.; Goltzman, D.; Miao, D.; et al. Abnormal Neurogenesis in the Dentate Gyrus of Adult Mice Lacking 1,25-Dihydroxy Vitamin D3 (1,25-(OH)2D3). Hippocampus 2012, 22, 421–433. [Google Scholar] [CrossRef]
  70. Eyles, D.; Almeras, L.; Benech, P.; Patatian, A.; Mackay-Sim, A.; McGrath, J.; Féron, F. Developmental Vitamin D Deficiency Alters the Expression of Genes Encoding Mitochondrial, Cytoskeletal and Synaptic Proteins in the Adult Rat Brain. J. Steroid Biochem. Mol. Biol. 2007, 103, 538–545. [Google Scholar] [CrossRef]
  71. Almeras, L.; Eyles, D.; Benech, P.; Laffite, D.; Villard, C.; Patatian, A.; Boucraut, J.; Mackay-Sim, A.; McGrath, J.; Féron, F. Developmental Vitamin D Deficiency Alters Brain Protein Expression in the Adult Rat: Implications for Neuropsychiatric Disorders. PROTEOMICS 2007, 7, 769–780. [Google Scholar] [CrossRef] [PubMed]
  72. Eyles, D.; Brown, J.; Mackay-Sim, A.; McGrath, J.; Feron, F. Vitamin D3 and Brain Development. Neuroscience 2003, 118, 641–653. [Google Scholar] [CrossRef]
  73. Féron, F.; Burne, T.H.J.; Brown, J.; Smith, E.; McGrath, J.J.; Mackay-Sim, A.; Eyles, D.W. Developmental Vitamin D3 Deficiency Alters the Adult Rat Brain. Brain Res. Bull. 2005, 65, 141–148. [Google Scholar] [CrossRef] [PubMed]
  74. Naveilhan, P.; Neveu, I.; Wion, D.; Brachet, P. 1,25-Dihydroxyvitamin D3, an Inducer of Glial Cell Line-Derived Neurotrophic Factor. NeuroReport 1996, 7, 2171–2175. [Google Scholar] [CrossRef] [PubMed]
  75. Xu, Y.; Liang, L. Vitamin D3/Vitamin D Receptor Signaling Mitigates Symptoms of Post-Stroke Depression in Mice by Upregulating Hippocampal BDNF Expression. Neurosci. Res. 2021, 170, 306–313. [Google Scholar] [CrossRef]
  76. Koshkina, A.; Dudnichenko, T.; Baranenko, D.; Fedotova, J.; Drago, F. Effects of Vitamin D3 in Long-Term Ovariectomized Rats Subjected to Chronic Unpredictable Mild Stress: BDNF, NT-3, and NT-4 Implications. Nutrients 2019, 11, 1726. [Google Scholar] [CrossRef] [Green Version]
  77. Neveu, I.; Naveilhan, P.; Baudet, C.; Brachet, P.; Metsis, M. 1,25-Dihydroxyvitamin D3 Regulates NT-3, NT-4 but Not BDNF MRNA in Astrocytes. NeuroReport 1994, 6, 124–126. [Google Scholar] [CrossRef]
  78. Scott-Solomon, E.; Kuruvilla, R. Mechanisms of Neurotrophin Trafficking via Trk Receptors. Mol. Cell. Neurosci. 2018, 91, 25–33. [Google Scholar] [CrossRef]
  79. Atif, F.; Yousuf, S.; Sayeed, I.; Ishrat, T.; Hua, F.; Stein, D.G. Combination Treatment with Progesterone and Vitamin D Hormone Is More Effective than Monotherapy in Ischemic Stroke: The Role of BDNF/TrkB/Erk1/2 Signaling in Neuroprotection. Neuropharmacology 2013, 67, 78–87. [Google Scholar] [CrossRef] [Green Version]
  80. Abdallah, C.G.; Sanacora, G.; Duman, R.S.; Krystal, J.H. Ketamine and Rapid-Acting Antidepressants: A Window into a New Neurobiology for Mood Disorder Therapeutics. Annu. Rev. Med. 2015, 66, 509–523. [Google Scholar] [CrossRef] [Green Version]
  81. Li, N.; Lee, B.; Liu, R.J.; Banasr, M.; Dwyer, J.M.; Iwata, M.; Li, X.Y.; Aghajanian, G.; Duman, R.S. MTOR-Dependent Synapse Formation Underlies the Rapid Antidepressant Effects of NMDA Antagonists. Science 2010, 329, 959–964. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Jovanovic, J.N.; Czernik, A.J.; Fienberg, A.A.; Greengard, P.; Sihra, T.S. Synapsins as Mediators of BDNF-Enhanced Neurotransmitter Release. Nat. Neurosci. 2000, 3, 323–329. [Google Scholar] [CrossRef] [PubMed]
  83. Alghamdi, S.; Alsulami, N.; Khoja, S.; Alsufiani, H.; Tayeb, H.O.; Tarazi, F.I. Vitamin D Supplementation Ameliorates Severity of Major Depressive Disorder. J. Mol. Neurosci. 2020, 70, 230–235. [Google Scholar] [CrossRef] [PubMed]
  84. Seyedi, M.; Gholami, F.; Samadi, M.; Djalali, M.; Effatpanah, M.; Yekaninejad, M.S.; Hashemi, R.; Abdolahi, M.; Chamari, M.; Honarvar, N.M. The Effect of Vitamin D3 Supplementation on Serum BDNF, Dopamine, and Serotonin in Children with Attention-Deficit/Hyperactivity Disorder. CNS Neurol. Disord.-Drug Targets 2019, 18, 496–501. [Google Scholar] [CrossRef]
  85. Jiang, P.; Zhang, L.H.; Cai, H.L.; Li, H.D.; Liu, Y.P.; Tang, M.M.; Dang, R.L.; Zhu, W.Y.; Xue, Y.; He, X. Neurochemical Effects of Chronic Administration of Calcitriol in Rats. Nutrients 2014, 6, 6048–6059. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Pertile, R.A.N.; Cui, X.; Eyles, D.W. Vitamin D Signaling and the Differentiation of Developing Dopamine Systems. Neuroscience 2016, 333, 193–203. [Google Scholar] [CrossRef]
  87. Sabir, M.S.; Haussler, M.R.; Mallick, S.; Kaneko, I.; Lucas, D.A.; Haussler, C.A.; Whitfield, G.K.; Jurutka, P.W. Optimal Vitamin D Spurs Serotonin: 1,25-Dihydroxyvitamin D Represses Serotonin Reuptake Transport (SERT) and Degradation (MAO-A) Gene Expression in Cultured Rat Serotonergic Neuronal Cell Lines. Genes Nutr. 2018, 13, 19. [Google Scholar] [CrossRef]
  88. Luthold, R.V.; Fernandes, G.R.; Franco-de-Moraes, A.C.; Folchetti, L.G.D.; Ferreira, S.R.G. Gut Microbiota Interactions with the Immunomodulatory Role of Vitamin D in Normal Individuals. Metabolism 2017, 69, 76–86. [Google Scholar] [CrossRef]
  89. Assa, A.; Vong, L.; Pinnell, L.J.; Avitzur, N.; Johnson-Henry, K.C.; Sherman, P.M. Vitamin D Deficiency Promotes Epithelial Barrier Dysfunction and Intestinal Inflammation. J. Infect. Dis. 2014, 210, 1296–1305. [Google Scholar] [CrossRef] [Green Version]
  90. Ooi, J.H.; Li, Y.; Rogers, C.J.; Cantorna, M.T. Vitamin D Regulates the Gut Microbiome and Protects Mice from Dextran Sodium Sulfate–Induced Colitis. J. Nutr. 2013, 143, 1679–1686. [Google Scholar] [CrossRef]
  91. Wu, S.; Liao, A.P.; Xia, Y.; Chun Li, Y.; Li, J.D.; Sartor, R.B.; Sun, J. Vitamin D Receptor Negatively Regulates Bacterial-Stimulated NF-ΚB Activity in Intestine. Am. J. Pathol. 2010, 177, 686–697. [Google Scholar] [CrossRef] [PubMed]
  92. Fedotova, J.; Dudnichenko, T.; Kruzliak, P.; Puchavskaya, Z. Different Effects of Vitamin D Hormone Treatment on Depression-like Behavior in the Adult Ovariectomized Female Rats. Biomed. Pharmacother. 2016, 84, 1865–1872. [Google Scholar] [CrossRef] [PubMed]
  93. Fedotova, J.O. Vitamin D3 Treatment Differentially Affects Anxiety-like Behavior in the Old Ovariectomized Female Rats and Old Ovariectomized Female Rats Treated with Low Dose of 17β-Estradiol. BMC Med. Genet. 2019, 20, 49. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Camargo, A.; Dalmagro, A.P.; Rikel, L.; da Silva, E.B.; Simão da Silva, K.A.B.; Zeni, A.L.B. Cholecalciferol Counteracts Depressive-like Behavior and Oxidative Stress Induced by Repeated Corticosterone Treatment in Mice. Eur. J. Pharmacol. 2018, 833, 451–461. [Google Scholar] [CrossRef]
  95. Da Silva Souza, S.V.; da Rosa, P.B.; Neis, V.B.; Moreira, J.D.; Rodrigues, A.L.S.; Moretti, M. Effects of Cholecalciferol on Behavior and Production of Reactive Oxygen Species in Female Mice Subjected to Corticosterone-Induced Model of Depression. Naunyn. Schmiedebergs Arch. Pharmacol. 2020, 393, 111–120. [Google Scholar] [CrossRef]
  96. Neis, V.B.; Werle, I.; Moretti, M.; Rosa, P.B.; Camargo, A.; Dalsenter, Y.D.O.; Platt, N.; Rosado, A.F.; Engel, W.D.; de Almeida, G.R.L.; et al. Involvement of Serotonergic Neurotransmission in the Antidepressant-like Effect Elicited by Cholecalciferol in the Chronic Unpredictable Stress Model in Mice. Metab. Brain Dis. 2022, 37, 1597–1608. [Google Scholar] [CrossRef]
  97. Zhang, W.; Guo, Y.; Wang, K.; Chen, L.; Jiang, P. Neuroprotective Effects of Vitamin D and 17ß-Estradiol against Ovariectomy-Induced Neuroinflammation and Depressive-like State: Role of the AMPK/NF-ΚB Pathway. Int. Immunopharmacol. 2020, 86, 106734. [Google Scholar] [CrossRef]
  98. Bakhtiari-Dovvombaygi, H.; Izadi, S.; Zare Moghaddam, M.; Hashemzehi, M.; Hosseini, M.; Azhdari-Zarmehri, H.; Dinpanah, H.; Beheshti, F. Beneficial Effects of Vitamin D on Anxiety and Depression-like Behaviors Induced by Unpredictable Chronic Mild Stress by Suppression of Brain Oxidative Stress and Neuroinflammation in Rats. Naunyn. Schmiedebergs Arch. Pharmacol. 2021, 394, 655–667. [Google Scholar] [CrossRef]
  99. Jiang, P.; Zhang, W.Y.; Li, H.D.; Cai, H.L.; Liu, Y.P.; Chen, L.Y. Stress and Vitamin D: Altered Vitamin D Metabolism in Both the Hippocampus and Myocardium of Chronic Unpredictable Mild Stress Exposed Rats. Psychoneuroendocrinology 2013, 38, 2091–2098. [Google Scholar] [CrossRef]
  100. Groves, N.J.; Bradford, D.; Sullivan, R.K.P.; Conn, K.A.; Aljelaify, R.F.; McGrath, J.J.; Burne, T.H.J. Behavioural Effects of Adult Vitamin D Deficiency in BALB/c Mice Are Not Associated with Proliferation or Survival of Neurons in the Adult Hippocampus. PLoS ONE 2016, 11, e0152328. [Google Scholar] [CrossRef] [Green Version]
  101. Jorde, R.; Sneve, M.; Figenschau, Y.; Svartberg, J.; Waterloo, K. Effects of Vitamin D Supplementation on Symptoms of Depression in Overweight and Obese Subjects: Randomized Double-Blind Trial. J. Intern. Med. 2008, 264, 599–609. [Google Scholar] [CrossRef] [PubMed]
  102. Narula, N.; Cooray, M.; Anglin, R.; Muqtadir, Z.; Narula, A.; Marshall, J.K. Impact of High-Dose Vitamin D3 Supplementation in Patients with Crohn’s Disease in Remission: A Pilot Randomized Double-Blind Controlled Study. Dig. Dis. Sci. 2017, 62, 448–455. [Google Scholar] [CrossRef] [PubMed]
  103. Penckofer, S.; Byrn, M.; Adams, W.; Emanuele, M.A.; Mumby, P.; Kouba, J.; Wallis, D.E. Vitamin D Supplementation Improves Mood in Women with Type 2 Diabetes. J. Diabetes Res. 2017, 2017, 8232863. [Google Scholar] [CrossRef]
  104. Sharifi, A.; Vahedi, H.; Nedjat, S.; Mohamadkhani, A.; Hosseinzadeh Attar, M.J. Vitamin D Decreases Beck Depression Inventory Score in Patients with Mild to Moderate Ulcerative Colitis: A Double-Blind Randomized Placebo-Controlled Trial. J. Diet. Suppl. 2019, 16, 541–549. [Google Scholar] [CrossRef] [PubMed]
  105. Yosaee, S.; Soltani, S.; Esteghamati, A.; Motevalian, S.A.; Tehrani-Doost, M.; Clark, C.C.T.; Jazayeri, S. Effects of Zinc, Vitamin D, and Their Co-Supplementation on Mood, Serum Cortisol, and Brain-Derived Neurotrophic Factor in Patients with Obesity and Mild to Moderate Depressive Symptoms: A Phase II, 12-Wk, 2 × 2 Factorial Design, Double-Blind, Randomized, Placebo-Controlled Trial. Nutrition 2020, 71, 110601. [Google Scholar] [CrossRef]
  106. Zhu, C.; Zhang, Y.; Wang, T.; Lin, Y.; Yu, J.; Xia, Q.; Zhu, P.; Zhu, D. Vitamin D Supplementation Improves Anxiety but Not Depression Symptoms in Patients with Vitamin D Deficiency. Brain Behav. 2020, 10, e01760. [Google Scholar] [CrossRef] [PubMed]
  107. Hansen, J.P.; Pareek, M.; Hvolby, A.; Schmedes, A.; Toft, T.; Dahl, E.; Nielsen, C.T. Vitamin D3 Supplementation and Treatment Outcomes in Patients with Depression (D3-Vit-Dep). BMC Res. Notes 2019, 12, 203. [Google Scholar] [CrossRef] [Green Version]
  108. Kaviani, M.; Nikooyeh, B.; Zand, H.; Yaghmaei, P.; Neyestani, T.R. Effects of Vitamin D Supplementation on Depression and Some Involved Neurotransmitters. J. Affect. Disord. 2020, 269, 28–35. [Google Scholar] [CrossRef]
  109. Alavi, N.M.; Khademalhoseini, S.; Vakili, Z.; Assarian, F. Effect of Vitamin D Supplementation on Depression in Elderly Patients: A Randomized Clinical Trial. Clin. Nutr. 2019, 38, 2065–2070. [Google Scholar] [CrossRef]
  110. Bertone-Johnson, E.R.; Powers, S.I.; Spangler, L.; Larson, J.; Michael, Y.L.; Millen, A.E.; Bueche, M.N.; Salmoirago-Blotcher, E.; Wassertheil-Smoller, S.; Brunner, R.L.; et al. Vitamin D Supplementation and Depression in the Women’s Health Initiative Calcium and Vitamin D Trial. Am. J. Epidemiol. 2012, 176, 1–13. [Google Scholar] [CrossRef]
  111. Mozaffari-Khosravi, H.; Nabizade, L.; Yassini-Ardakani, S.M.; Hadinedoushan, H.; Barzegar, K. The Effect of 2 Different Single Injections of High Dose of Vitamin D on Improving the Depression in Depressed Patients With Vitamin D Deficiency: A Randomized Clinical Trial. J. Clin. Psychopharmacol. 2013, 33, 378–385. [Google Scholar] [CrossRef] [PubMed]
  112. Vellekkatt, F.; Menon, V.; Rajappa, M.; Sahoo, J. Effect of Adjunctive Single Dose Parenteral Vitamin D Supplementation in Major Depressive Disorder with Concurrent Vitamin D Deficiency: A Double-Blind Randomized Placebo-Controlled Trial. J. Psychiatr. Res. 2020, 129, 250–256. [Google Scholar] [CrossRef] [PubMed]
  113. Khoraminya, N.; Tehrani-Doost, M.; Jazayeri, S.; Hosseini, A.; Djazayery, A. Therapeutic Effects of Vitamin D as Adjunctive Therapy to Fluoxetine in Patients with Major Depressive Disorder. Aust. N. Z. J. Psychiatry 2013, 47, 271–275. [Google Scholar] [CrossRef] [PubMed]
  114. Eid, A.; Khoja, S.; AlGhamdi, S.; Alsufiani, H.; Alzeben, F.; Alhejaili, N.; Tayeb, H.O.; Tarazi, F.I. Vitamin D Supplementation Ameliorates Severity of Generalized Anxiety Disorder (GAD). Metab. Brain Dis. 2019, 34, 1781–1786. [Google Scholar] [CrossRef] [PubMed]
  115. Valdivielso, J.M.; Fernandez, E. Vitamin D receptor polymorphisms and diseases. Clin. Chim. Acta 2006, 371, 1–12. [Google Scholar] [CrossRef]
  116. Lye, M.S.; Tor, Y.S.; Tey, Y.Y.; Shahabudin, A.; Loh, S.P.; Ibrahim, N.; Stanslas, J.; Rosli, R.; Ling, K.H. BsmI-ApaI-TaqI TAC (BAt) Haplotype of Vitamin D Receptor Gene Is Associated with Increased Risk of Major Depressive Disorder. J. Mol. Neurosci. 2020, 71, 981–990. [Google Scholar] [CrossRef]
  117. Usategui-Martín, R.; De Luis-Román, D.A.; Fernández-Gómez, J.M.; Ruiz-Mambrilla, M.; Pérez-Castrillón, J.L. Vitamin D Receptor (VDR) Gene Polymorphisms Modify the Response to Vitamin D Supplementation: A Systematic Review and Meta-Analysis. Nutrients 2022, 14, 360. [Google Scholar] [CrossRef]
Figure 1. Antioxidant, anti-inflammatory, and neuromodulatory properties of vitamin D. Neurons and glial cells are able to express VDR in regions such as the prefrontal cortex and hippocampus. In these regions, the positive modulation of antioxidant enzymes, such as HO-1, CAT, and SOD, by calcitriol contributes to redox homeostasis and, consequently, to the attenuation of the neuroinflammatory process. Calcitriol is also able to increase the expression of IkBα, thus, inhibiting the nuclear translocation of NF-kB. As a consequence, less synthesis, oligomerization, and activation of the NLRP3 inflammasome occurs, resulting in attenuation of pro-inflammatory cytokines (such as IL-1β and TNF-α) and an increase in anti-inflammatory cytokines (such as IL-10 and IL-4) (A). In addition, vitamin D has also been shown to modulate the intestinal microbiota (C). Finally, vitamin D actively regulates the synthesis of monoamines and BDNF, thus, favoring the process of synaptic plasticity (B). Abbreviations: ↓: decreased; ↑: increased; BDNF: brain-derived neurotrophic factor; CAT: catalase; HO-1: heme oxygenase-1; IL-1β: interleukin-1β; IL-4: interleukin-4; IL-10: interleukin-10; IkBα: nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor, alpha; MAO: monoamine oxidase; NF-kB: nuclear factor kappa B; NLRP3: NOD-like receptor family pyrin domain-containing 3; ROS: reactive oxygen species; SERT: serotonin transporters; SOD: superoxide dismutase; TNF-α: tumor necrosis factor-alpha; VDR: vitamin D receptor.
Figure 1. Antioxidant, anti-inflammatory, and neuromodulatory properties of vitamin D. Neurons and glial cells are able to express VDR in regions such as the prefrontal cortex and hippocampus. In these regions, the positive modulation of antioxidant enzymes, such as HO-1, CAT, and SOD, by calcitriol contributes to redox homeostasis and, consequently, to the attenuation of the neuroinflammatory process. Calcitriol is also able to increase the expression of IkBα, thus, inhibiting the nuclear translocation of NF-kB. As a consequence, less synthesis, oligomerization, and activation of the NLRP3 inflammasome occurs, resulting in attenuation of pro-inflammatory cytokines (such as IL-1β and TNF-α) and an increase in anti-inflammatory cytokines (such as IL-10 and IL-4) (A). In addition, vitamin D has also been shown to modulate the intestinal microbiota (C). Finally, vitamin D actively regulates the synthesis of monoamines and BDNF, thus, favoring the process of synaptic plasticity (B). Abbreviations: ↓: decreased; ↑: increased; BDNF: brain-derived neurotrophic factor; CAT: catalase; HO-1: heme oxygenase-1; IL-1β: interleukin-1β; IL-4: interleukin-4; IL-10: interleukin-10; IkBα: nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor, alpha; MAO: monoamine oxidase; NF-kB: nuclear factor kappa B; NLRP3: NOD-like receptor family pyrin domain-containing 3; ROS: reactive oxygen species; SERT: serotonin transporters; SOD: superoxide dismutase; TNF-α: tumor necrosis factor-alpha; VDR: vitamin D receptor.
Ijms 23 07077 g001
Table 1. Preclinical studies on effects of vitamin D in models of depression and anxiety.
Table 1. Preclinical studies on effects of vitamin D in models of depression and anxiety.
StudyAnimal Model TreatmentBehavioral AlterationsBiochemical Alterations
Fedotova et al., 2016 [92]Ovariectomized Wistar ratsCholecalciferol (5 mg/kg for 14 days; s.c.)Antidepressant-like effect in the FST Not evaluated
Fedotova et al., 2017 [10]Ovariectomized Wistar ratsCholecalciferol (5 mg/kg for 14 days; s.c.)Anxiolytic-like effect in EPM and LDTNot evaluated
Fedotova 2019 [93]Ovariectomized Wistar ratsCholecalciferol (5 mg/kg for 14 days; s.c.)Anxiolytic-like effect in EPM and LDTNot evaluated
Camargo et al., 2018 [94]Corticosterone (21 days) in male Swiss miceCholecalciferol (2.5 µg/kg, for 7 days; p.o.)Antidepressant-like effect in the splash test and TST↓ Protein carbonyl and nitrite levels
da Silva Souza et al., 2020 [95]Corticosterone (21days) in female Swiss miceCholecalciferol (100 IU/kg, p.o.) for 7 daysAntidepressant-like effect in the TST↓ ROS
Camargo et al., 2020 [57]Corticosterone (21 days) in male Swiss miceCholecalciferol (2.5 μg/kg, p.o.) for 7 daysAntidepressant-like effect in the TST and splash test↓ ASC, caspase-1, and TXNIP
Neis et al., 2022 [96]CUMS in female Swiss miceCholecalciferol (2.5 μg/kg, p.o.) for 7 daysAntidepressant-like effect in the TST↑ serotonin levels in the prefrontal cortex
Zhang et al., 2020 [97]Ovariectomized female Sprague-Dawley ratsCalcitriol (100 ng/kg, p.o.) for 10 weeksAntidepressant-like effect in the FST and novelty-suppressed feeding test↓ IL-1β, IL-6, and TNF-α, iNOS and COX-2
Bakhtiari-Dovvombaygi et al., 2021 [98]CUMS in male Wistar ratsVitamin D3 (10,000 IU/kg) for 28 daysAnxiolytic and antidepressant-like effect in EPM and FST↓ Malondialdehyde and IL-6
↑ Total thiols, SOD, and CAT
Jiang et al., 2013 [99]CUMS (4 weeks) in male Sprague-Dawley ratsWithout treatmentDepressive-like behavior in sucrose preference test↑ 1,25(OH)2D and VDR
Koshkina et al., 2019 [76]CUMS in ovariectomized Wistar ratVitamin D3 (5 mg/kg for 4 weeks; s.c.)Antidepressant-like effect in FST and sucrose preference test↑BDNF and NT-3/NT-4
Xu and Liang 2021 [75]post-stroke depression model (male C57BL/6 mice)Calcitriol (25 μg/kg/day for 4 weeks; i.c.v.)Antidepressant-like effect in FST and sucrose preference test↑ VDR and BDNF
↓ decreased; ↑ increased; ASC: apoptosis-associated speck-like protein containing a caspase recruitment domain (CARD); BDNF: brain-derived neurotrophic factor; CAT: catalase; COX-2: cyclooxygenase-2; CUMS: chronic unpredictable mild stress; EPM: elevated plus maze; FST: forced swimming test; i.c.v.: intracerebroventricular; IL-1β: interleukin-1β; IL-6: interleukin-6; iNOS: inducible nitric oxide synthase; LTD: light–dark box test; p.o.: by mouth (orally); ROS, reactive oxygen species; s.c.: subcutaneous; SOD: superoxide dismutase; TNF-α: tumor necrosis factor-alpha; TST: tail suspension test; TXNIP: thioredoxin-interacting protein; VDR: vitamin D receptor.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kouba, B.R.; Camargo, A.; Gil-Mohapel, J.; Rodrigues, A.L.S. Molecular Basis Underlying the Therapeutic Potential of Vitamin D for the Treatment of Depression and Anxiety. Int. J. Mol. Sci. 2022, 23, 7077. https://doi.org/10.3390/ijms23137077

AMA Style

Kouba BR, Camargo A, Gil-Mohapel J, Rodrigues ALS. Molecular Basis Underlying the Therapeutic Potential of Vitamin D for the Treatment of Depression and Anxiety. International Journal of Molecular Sciences. 2022; 23(13):7077. https://doi.org/10.3390/ijms23137077

Chicago/Turabian Style

Kouba, Bruna R., Anderson Camargo, Joana Gil-Mohapel, and Ana Lúcia S. Rodrigues. 2022. "Molecular Basis Underlying the Therapeutic Potential of Vitamin D for the Treatment of Depression and Anxiety" International Journal of Molecular Sciences 23, no. 13: 7077. https://doi.org/10.3390/ijms23137077

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