Hostname: page-component-78c5997874-8bhkd Total loading time: 0 Render date: 2024-11-03T04:20:11.446Z Has data issue: false hasContentIssue false

Vitamin D and depression in older adults: lessons learned from observational and clinical studies

Published online by Cambridge University Press:  13 January 2022

Gilciane Ceolin
Affiliation:
Postgraduate Program in Nutrition, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil Translational Nutritional Neuroscience working Group, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil
Luciana da Conceição Antunes
Affiliation:
Department of Nutrition, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil Translational Nutritional Neuroscience working Group, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil
Morgana Moretti
Affiliation:
Postgraduate Program in Biochemistry, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil
Débora Kurrle Rieger
Affiliation:
Department of Nutrition, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil Translational Nutritional Neuroscience working Group, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil
Júlia Dubois Moreira*
Affiliation:
Department of Nutrition, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil Translational Nutritional Neuroscience working Group, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil
*
*Corresponding author: Prof. Júlia D. Moreira, email [email protected]
Rights & Permissions [Opens in a new window]

Abstract

Depression is a mental disorder triggered by the interaction of social, psychological and biological factors that have an important impact on an individual’s life. Despite being a well-studied disease with several established forms of treatment, its prevalence is increasing, especially among older adults. New forms of treatment and prevention are encouraged, and some researchers have been discussing the effects of vitamin D (VitD) on depression; however, the exact mechanism by which VitD exerts its effects is not yet conclusive. In this study, we aimed to discuss the possible mechanisms underlying the association between VitD and depression in older adults. Therefore, we conducted a systematic search of databases for indexed articles published until 30 April 2021. The primary focus was on both observational studies documenting the association between VitD and depression/depressive symptoms, and clinical trials documenting the effects of VitD supplementation on depression/depressive symptoms, especially in older adults. Based on pre-clinical, clinical and observational studies, it is suggested that the maintenance of adequate VitD concentrations is an important issue, especially in older adults, which are a risk population for both VitD deficiency and depression. Nevertheless, it is necessary to carry out more studies using longitudinal approaches in low- and middle-income countries to develop a strong source of evidence to formulate guidelines and interventions.

Type
Review Article
Copyright
© The Author(s), 2022. Published by Cambridge University Press on behalf of The Nutrition Society

Introduction

Depression is a mental disorder that causes clinically significant suffering and/or impairment in social, professional, economic and other important areas of an individual’s life, and is the main cause of suicide in more severe cases(14). The prevalence of depression in 2015 was estimated to be 4·4 % globally, with a higher prevalence among those between 55 and 77 years of age. Women appear to be more affected (7·5 %) than men (5·5 %)(5). Among those over 60 years old, depression occurs in 7·0 % of the general older population(6). According to the Global Burden of Disease, Injuries, and Risk Factors – GBD survey, depression is among the top three causes of disability(7). There has been a significant increase in the global burden of disease in years lived with disabilities (YLDs) in the past 20 years due to depressive disorders. In 1990, depression occupied the fourth position, moving to the third in 2007 with an increase of 33·4 %, and remained in the third position between 2007 and 2017; however, it has increased by 14·3 %(Reference James, Abate and Abate8). Moreover, as many people with depressive symptoms are undiagnosed, the prevalence of depressive disorders is probably higher than reported(4).

Mental disorders are among the main problems in public health, and mood disorders are diseases with higher costs to health systems worldwide(Reference Chisholm, Sweeny and Sheehan2,Reference Olesen, Gustavsson and Svensson3,Reference DiLuca and Olesen9,Reference Knapp and Wong10) . According to the Mental Health Atlas of the World Health Organization (WHO), low- and middle-income countries spend less than $1 per year per capita in the treatment and prevention of mental disorders, compared with an average of >$80 in high-income countries, owing to socioeconomic issues(11). Therefore, there is an urgent need to identify the modifiable risk factors associated with the aetiology of depression, helping with the treatment and prevention of this disorder, especially in low- and middle-income countries(Reference Whiteford, Ferrari and Degenhardt12,13) .

Depression is a complex disease triggered by the interaction between social, psychological and biological factors(4,Reference Li, D’Arcy and Meng14) . In older adults, depression can be triggered by a series of factors such as limitations in daily activities, cognition, mobility and social changes such as retirement, social isolation and relocation to long-term institutions(Reference Andrade, Wu and Lebrão15). Among the biological factors, genetic predisposition, neurotransmitter and neuroendocrine system imbalance, functional and structural brain anatomy, and cognition are the most studied mechanisms(Reference Kaltenboeck and Harmer16,Reference Otte, Gold and Penninx17) . Recently, nutritional factors have shown an important relationship with the evolution, prevention and treatment of mental disorders(Reference Lai, Hiles and Bisquera18). The association between VitD and depression has emerged in scientific scenarios, and this nutrient seems to be relevant in the prevention of depressive symptom development. However, the mechanism by which VitD exerts its effects remains unclear(Reference Camargo, Dalmagro and Rikel19,Reference Fedotova, Dudnichenko and Kruzliak20) .

Many clinical trials have been conducted to investigate the potential therapeutic effect of VitD on patients with depression, but the results remain inconclusive due to methodological issues(Reference Spedding21). VitD is a fat-soluble vitamin that is present in two forms: VitD2 (ergosterol) and VitD3 (cholecalciferol). It is obtained from diet, supplementation and sun exposure(Reference Bikle22,Reference Jäpelt and Jakobsen23) . VitD has a well-established role in mineral bone metabolism, but its effects are not restricted to bone health and are also important in maintaining many biological processes, such as the regulation of gene expression, cell proliferation and differentiation, and immune system regulation(Reference Johnson and Mohn24Reference Umar, Sastry and Chouchane26). In the central nervous system (CNS), the presence of nuclear (vitamin D receptor, VDR) and membrane (protein disulphide isomerase family A member 3, PDIA3) receptors for VitD and some enzymes (cytochrome P450 family enzymes CYP27a1, CYP27b1 and CYP24a1) responsible for converting its active form has raised the hypothesis that VitD may be involved in the pathophysiology of depression(Reference Berridge27Reference Landel, Stephan and Cui31).

Low serum VitD concentrations [25-hydroxycholecalciferol, 25(OH)D] have been considered a public health problem worldwide, especially in the elderly(Reference Palacios and Gonzalez32). For older adults, the prevalence of 25(OH)D deficiency (<50 nmol/l or <20 ng/ml) was 36 % in the United States(Reference Ganji, Zhang and Tangpricha33), 19 % in Canada(Reference Whiting, Langlois and Vatanparast34), 36 % in China(35 and 4–89 % in European countries(Reference Palacios and Gonzalez32). In low- and middle-income countries, the prevalence was approximately 41 % for older adults in Brazil(Reference Pereira-Santos, Santos and Carvalho36), 91 % in India(Reference Marwaha, Tandon and Garg37) and 46 % in Guatemala(Reference Sud, Montenegro-Bethancourt and Bermúdez38). However, different cut-off points have been suggested, and a single value to define VitD deficiency or insufficiency has been debated(Reference Bouillon39). Moreover, the establishment of desirable serum VitD concentrations is based on bone health to maintain mineral and skeletal homoeostasis(Reference Bouillon39,Reference Sempos, Heijboer and Bikle40) .

It is important to mention that VitD levels via skin synthesis and intestinal absorption are influenced by various factors such as skin pigmentation, latitude, season, age, obesity and inflammatory bowel diseases, among others(Reference Arabi, El Rassi and El-Hajj Fuleihan41Reference Amrein, Scherkl and Hoffmann43). Due to reduced sun exposure, decreased skin synthesis and dietary intake, and intestinal malabsorption, the elderly are among the top risk groups for VitD deficiency(Reference Arabi, El Rassi and El-Hajj Fuleihan41,Reference Cesari, Incalzi and Zamboni44) . They also present significant complications related to low VitD concentrations (<20 ng/ml), such as the risk of fractures due to fragility and bone loss, which contribute to age-related muscle weakness and sarcopenia(Reference Kesby, Eyles and Burne28,Reference Amrein, Scherkl and Hoffmann43,Reference Holick45,Reference Luo, Quan and Lin46) . In addition, VitD concentrations <20 ng/ml have been associated with an increased risk of all-cause mortality(Reference Dudenkov, Mara and Petterson47).

In this review, we aimed to update the role of VitD in depression, discussing the metabolism of VitD, its mechanism of action in the brain and the main evidence of pre-clinical, clinical and observational studies, especially those involving older adults, a population risk for both conditions, in an attempt to highlight the potential preventive and therapeutic effects of this nutrient. Also, we aimed to suggest future directions for new studies. To this end, we conducted a systematic search for articles published until 30 April 30 2021. The databases used were PubMed, Scopus, Embase, Science Direct and Web of Science (details are presented in the supplementary material).

Vitamin D: synthesis and metabolism

The synthesis of VitD (Fig. 1) by epidermal epithelial cells begins when the exposure to ultraviolet B radiation (UVB, 290–315 nm) promotes the non-enzymatic transformation of 7-dehydrocholesterol (7-DHC or pro-VitD) in pre-VitD3(Reference Tian and Holick48,Reference Bikle and Christakos49) . A photolytic break forms a secosteroid molecule, which then undergoes an isomerisation reaction induced by heat to transform it into VitD3 (or cholecalciferol), a process that takes about 8 h(Reference Tian and Holick48Reference Wacker and Holick50). Keratinocytes are the main cells of the epidermis that have the enzymatic machinery to metabolise VitD in its active form and express the vitamin D receptor (VDR)(Reference Bikle22,Reference Bouillon, Marcocci and Carmeliet51) . In contrast, the synthesis of the active form of VitD from either food or supplementation begins with incorporation into micelles and absorption through the enterocyte membrane by apical membrane transporters or by passive diffusion(Reference Reboul52). A fraction of VitD is incorporated into the chylomicrons, which are transported to the lymphatic system and then to the venous system by vitamin D binding protein (DBP)(Reference Wacker and Holick50). The other fraction is incorporated into adipose tissue and skeletal muscles(Reference Gil, Plaza-Diaz and Mesa53).

Fig. 1. Vitamin D synthesis, metabolism and target tissue actions. (1) The synthesis of VitD from sunlight initiates in the skin when 7-DHC is converted in pre-VitD3 and then VitD3 [25(OH)D3 or cholecalciferol] and is carried by DBP through blood circulation. (Reference Chisholm, Sweeny and Sheehan2) The VitD from dietary intake (VitD2/ergocalciferol and D3/cholecalciferol) is absorbed in the small intestine and packed into chylomicrons to reach the systemic circulation. Both VitD3 and VitD2 are also transported through blood circulation by DBP to the liver, where they are converted to 25-hydroxyvitamin D [calcidiol or 25(OH)D] by the action of 25-hydroxylases. (3) 25(OH)D coupled to DBP is transported to the target organs such as kidney, bones, adipose tissue, muscle and brain, and cells such as in the immune system containing the enzyme 1-α-hydroxylase, which convert 25(OH)D to 1,25-dihydroxyvitamin D [calcitriol or 1,25(OH)2D3], the active form of VitD. VitD act through both genomic and non-genomic pathways. In the genomic pathway, VitD active form enters the nucleus linked to the VDR where it binds to the RXR and then binds to the VDRE, resulting in modulation of target gene expression. In the non-genomic pathway, the VitD active form binds to the PDIA3 and starts signalling cascades, including the activation of phospholipase A2 activating protein (PLAA), phospholipase A2 (PLA2), phospholipase C (PLC) and opening Ca2+ channels that results in the activation of secondary messengers. This figure was made using BioRender (license: YN235V4QZA)

Both VitD2 and VitD3 are transported in the blood by DBP and must undergo activation through two consecutive enzymatic hydroxylation reactions in the liver and kidneys. In the liver, VitD2 and VitD3 are converted into 25-hydroxyvitamin D (calcidiol or 25(OH)D) by the action of 25-hydroxylases (cytochrome P450 enzymes group, CYP2R1 or CYP27A1)(Reference Holick54Reference Wacker and Holick56). The 25(OH)D coupled with DBP is transported to various tissues with cells containing the enzyme 1-α-hydroxylase (CYP27B1), as in the kidney, where it converts 25(OH)D to 1,25-dihydroxyvitamin D (calcitriol or 1,25(OH)2D3), the active form of VitD(Reference Holick54Reference Wacker and Holick56).

The conversion of 1,25(OH)2D3 in the kidney is regulated by several factors, including circulating concentrations of parathyroid hormone (PTH) in the parathyroid glands, serum phosphorus, calcium, fibroblast growth factor 23 (FGF-23) in the bone and its self-regulation. 1,25(OH)2D3 decreases its own synthesis by negative feedback; it decreases the secretion of parathyroid hormone and increases the expression of 24-hydroxylase(Reference Holick57). This self-regulation by the expression of 24-hydroxylase is found in most tissues and is essential for the catabolism of 25(OH)D and 1,25(OH)2D3(Reference Bikle, Patzek and Wang58).

The biological effects of 1,25(OH)2D3 are largely mediated by VDR, which is expressed in almost all human cells(Reference Bouillon, Carmeliet and Verlinden59,Reference Haussler, Jurutka and Mizwicki60) . The VDR belongs to a subfamily of nuclear receptors, which contains two sites for ligand binding called the genomic pocket (VDR-GP), which binds in a bowl-like configuration for gene transcription, and the alternative pocket (VDR-AP), which connects in a planar-like configuration for quick responses(Reference Haussler, Jurutka and Mizwicki60). When VDR-GP binds to 1,25(OH)2D3, it enters the cell nucleus and binds to the retinoid X receptor (RXR). This complex then binds to the vitamin D responsive element (VDRE) in the promoter regions of the target genes by recruiting co-activator or co-repressor complexes that regulate the transcription of genes either positively or negatively(Reference Gil, Plaza-Diaz and Mesa53,Reference Haussler, Jurutka and Mizwicki60) . The other suggested VitD receptor is PDIA3, also known as endoplasmic reticulum protein (ERp60, ERp57 and Grp58) or VitD membrane-associated rapid-response steroid-binding protein (1,25-MARRS)(Reference Chen, Olivares-Navarrete and Wang61). PDIA3 is present in caveolae (lipid rafts) and is linked to the rapid responses of 1,25(OH)2D3 by activating signalling cascades, where it physically interacts with downstream mediators(Reference Chen, Olivares-Navarrete and Wang61,Reference Boyan, Chen and Schwartz62) , including the activation of phospholipase A2 activating protein (PLAA), phospholipase A2 (PLA2), phospholipase C (PLC) and opening Ca2+ channels that result in the activation of secondary messengers(Reference Zmijewski and Carlberg63). PDIA3 is involved in the function of immune and musculoskeletal systems as well as mammary gland growth and development, and participates in the intestinal uptake of calcium and phosphate(Reference Zmijewski and Carlberg63). PDIA3 also mediates the effect of 1,25(OH)2D3 on the regulation of osteoblasts and chondrocytes(Reference Doroudi, Plaisance and Boyan64).

Vitamin D: mechanism of action in the brain

The first evidence of the role of VitD in brain function began with autoradiographic findings of the presence of VDR in the brain tissue of laboratory animals(Reference Stumpf, Sar and Clark65). VDR is found in neurons and glial cells in most regions of the brain, including the cortex (temporal, frontal, parietal and cingulate); deep grey matter (thalamus, basal ganglia, hypothalamus, hippocampus and amygdala); cerebellum, nuclei of the brain stem and substantia nigra (an area abundant in dopaminergic neurons); spinal cord; and ventricular system(Reference DeLuca, Kimball and Kolasinski66). In addition, an alternative mechanism was observed in post-mortem human brain tissue samples. It was demonstrated that 1,25(OH)2D3 can be activated locally through the expression of the enzyme 1α-hydroxylase, which is classically expressed in the kidney and is responsible for catalysing the conversion of 25(OH)D into 1,25(OH)2D3, showing that both forms (VitD and 25(OH)D) can pass through the blood–brain barrier(Reference Eyles, Smith and Kinobe67,Reference Pardridge, Sakiyama and Coty68) .

It has been proposed that within the neurovascular unit, the machinery for conversion of both VitD forms involves the cytochrome P450 family enzymes CYP27a1, CYP27b1 and CYP24a1 which are expressed in neurons, and CYP27a1 which is expressed in all neural cell types and is highly expressed in endothelial cells(Reference Landel, Stephan and Cui31). The active form of VitD triggers genomic actions associated with VDR or non-genomic actions related to PDIA3, which is expressed in small amounts in extra-cerebral tissues such as the liver and kidney. On the other hand, PDIA3 is highly expressed in the brain and appears to be the main brain receptor for VitD in neural tissue (Fig. 2).

Fig. 2. The role of vitamin D in depression. (1) In the brain, both active and inactive VitD is carried through blood circulation binding to DBP and can permeate the blood–brain barrier. All brain cells (endothelial cells (A), astrocytes (B), neurons (C), oligodendrocytes (D) and microglia (E)) have the machinery to transform VitD. VitD is turned into 25(OH)D by CYP27a1 in endothelial cells and neurons, and it is metabolized to 1,25(OH)2D3 by CYP27b1 in neurons or microglia. All brain cells can express VDR, but it is highly expressed by astrocytes. When it enters the cell, 1,25(OH)2D3 can bind to VDR, and then to the RXR in the nucleus. The complex VDR–RXR binds to the VDRE and initiates gene transcription or can be inactivated when in excess by CYP24a1. All brain cells can express PDIA3, but it is highly expressed in endothelial cells where 1,25(OH)2D3 can bind it, and PDIA3 physically interacts with downstream mediators to initiate rapid responses and induce signalling cascades. (2) VitD regulates the expression of many processes related to depression. It maintains Ca2+ homoeostasis, activates the expression of many antioxidant genes, regulates the formation of serotonin and dopamine, and reduces inflammation by reducing the expression of inflammatory cytokines. TPH2, tryptophan hydroxylase 2; SERT, serotonin reuptake transporter; GDNF, glial cell-derived neurotrophic factor; COMT, catechol-O-methyltransferase; NRF2, nuclear factor-erythroid-2-related factor 2; γ-GT, γ-glutamyl transpeptidase; GCLC, glutamate-cysteine ligase; GR, glutathione reductase; GPx, glutathione peroxidase; NF-κB, nuclear factor-kappa B. This figure was made using BioRender (license: FB235V4MBD)

VitD is known as a neurosteroid because of its important role in the CNS in processes related to cell differentiation, production and release of neurotrophic factors, synthesis of neurotransmitters, intracellular calcium homoeostasis, influence on the redox state, function and metabolism of neuronal cells and cognition (Fig. 2)(Reference Mayne and Burne29,Reference Eyles, Feron and Cui69) . The active form of VitD stimulates the synthesis of nerve growth factor (NGF) which acts on cholinergic neurons, and positively regulates the synthesis of neurotrophic factors derived from the glial cell line (GDNF), which acts on dopaminergic neurons, and neurotrophin 3 (NT-3), which is key to neuronal promotion, survival, differentiation and plasticity(Reference DeLuca, Kimball and Kolasinski66). Due to its involvement in several brain functions, observational studies in humans subjects have linked low serum VitD concentrations with some brain disorders such as schizophrenia, failure in synaptic plasticity related to learning and memory, cognitive decline and mood disorders(Reference Berridge27,Reference Mayne and Burne29,Reference Cui, Gooch and Petty70) .

Vitamin D and depressive symptoms: evidence from pre-clinical and clinical studies

Pre-clinical studies

Depression is a multifactorial disease, which makes it challenging to identify the precise biological mechanisms that link VitD to depression. However, some hypotheses have been proposed based on the experimental research data. Calcium homoeostasis, glutamatergic/GABAergic and monoaminergic system modulation, influence on circadian rhythm, anti-inflammatory properties and redox balance modulation are among the most investigated mechanisms.

The homoeostasis of intracellular and extracellular calcium (Ca2+) is an important factor responsible for driving the onset of depression, which links VitD with the development of depressive symptoms because of its interaction with excitatory synapses(Reference Berridge27). The imbalance in intracellular Ca2+ is caused by an elevation in glutamate and by activation of the phosphoinositide signalling pathway that generates inositol triphosphate (IP3) which releases Ca2+ from internal stores(Reference Berridge27,Reference Warsh, Andreopoulos and Li71,Reference Yuan, Kiselyov and Shin72) . The elevation of Ca2+ can affect both ionotropic (N-methyl-d-aspartate) and metabotropic (mGluR) receptors(Reference Kandel, Schwartz and Jessell73). This change in neural activity drives excitatory neurons and is responsible for the decline in the activity and the number of GABAergic inhibitory neurons, as well as modulation of the activity of other neurotransmitter systems, including the inhibition of the serotonergic system and the release of norepinephrine and dopamine(Reference Croarkin, Levinson and Daskalakis74). However, 1,25(OH)2D can act in this pathway by inducing the expression of proteins related to the maintenance of Ca2+ homoeostasis, such as calbindin, parvalbumin, Na+/Ca2+ exchanger (NCX1) and pump Ca2+-ATPase (PMCA). It also regulates Ca2+ concentrations by reducing the expression of the CaV1·2 calcium channel(Reference Berridge27,Reference Bivona, Agnello and Bellia75) .

Concerning other neurotransmitter systems, it has been proposed that depression could result from a deficiency of serotonin (5-HT) in the synaptic cleft(Reference Domínguez-López, Howell and Gobbi76Reference Ogawa, Fujii and Koga78). 5-HT is derived from the essential amino acid tryptophan. To produce 5-HT in the brain, tryptophan must first be transported across the blood–brain barrier and then metabolised by the enzyme tryptophan hydroxylase 2 (TPH2). VDR activation by 1,25(OH)2D3 can induce the expression of the TPH2 gene in serotonergic neurons(Reference Kaneko, Sabir and Dussik79,Reference Patrick and Ames80) . In addition, 1,25(OH)2D3 could act in the repression of the serotonin reuptake transporter (SERT or 5-HTT), and the mitochondrial enzyme responsible for 5-HT catabolism, monoamine oxidase-A, resulting in potentiated serotonergic transmission(Reference Sabir, Haussler and Mallick81).

In the dopaminergic system, VitD is involved in the maturation of dopaminergic neurons. VDR is present in the nucleus of positive neurons for tyrosine hydroxylase (TH), and can stimulate glial cell line-derived neurotrophic factor (GDNF) in dopaminergic neurons(Reference Cui, Pelekanos and Liu82). VDR also modulates metabolism through the genomic regulation of catechol-O-methyl transferase (COMT) expression, a key enzyme involved in dopamine turnover(Reference Cui, Pelekanos and Liu82,Reference Cui, Pertile and Liu83) . In addition, in a rat model of depression, VitD appears to produce therapeutic effects comparable to antidepressant drugs such as fluoxetine, improving anhedonia-like symptoms, probably by regulating the effect of dopamine-related actions on the nucleus accumbens(Reference Sedaghat, Yousefian and Vafaei84).

From a chronobiological perspective, a growing body of evidence suggests that VitD participates in the mechanisms orchestrating the circadian rhythm, suggesting that hypovitaminosis D might play a role in sleep disorders(Reference McCarty, Reddy and Keigley85). VitD has been associated with the regulation and maintenance of optimal sleep(Reference Mosavat, Smyth and Arabiat86). The mediating role of VitD in the circadian rhythm is supported by studies demonstrating the association between lower concentrations of VitD and sleep(Reference Jones, Redmond and Fulford87,Reference Muscogiuri, Barrea and Scannapieco88) . In addition, a circadian oscillation pattern can be equally observed in plasma 1,25(OH)2D3 concentration and DBP, which corroborates the association between VitD and the circadian system(Reference Jones, Redmond and Fulford87).

Because sunlight partially regulates the synthesis of VitD and is the main zeitgeber in the regulation of the circadian rhythm, it is conceivable that VitD might contribute to the transduction of signs regulating it(Reference Lucock, Jones and Martin89,Reference Romano, Muscogiuri and Di Benedetto90) . The suprachiasmatic nucleus (SCN) is a hypothalamic structure found directly above the optic chiasm, and its strategic anatomical position allows prompt central response to sunlight stimuli through the retina. SCN is the main oscillator, which accounts for the control of circadian rhythms by regulating several body functions during a 24-h cycle, sending peripheral signals through neurohumoral mechanisms(Reference Dibner, Schibler and Albrecht91). For this reason, the authors postulated that VitD is likely involved in the regulation of the sleep/wake rhythm(Reference Romano, Muscogiuri and Di Benedetto90).

Melatonin is a neurohormone involved in the regulation of mammalian circadian rhythms and sleep. It is released in response to darkness and is synthesised by the pineal gland(Reference Stehle, von Gall and Korf92). Its synthesis occurs from the metabolism of serotonin(Reference Zhao, Yu and Shen93), which, in turn, is also regulated by VitD. Along with VDR, 1,25(OH)2D triggers the central expression of TPH2, the gene responsible for encoding the enzyme catalysing the conversion of tryptophan into 5-hydroxytryptophan, which is then metabolised into serotonin and subsequently as melatonin(Reference Eyles, Smith and Kinobe67,Reference Kaneko, Sabir and Dussik79) . Therefore, it is thought that the combination of deficits in serum VitD levels and circadian rhythm impairments could induce a robust increase in depressive symptoms and/or act as an interplay variable in the pathophysiology of major depressive disorder.

Regarding anti-inflammatory pathways, it is also relevant to point out that both melatonin and VitD mediate the mitochondrial function in homoeostasis, such as down-regulating mechanistic target of rapamycin (mTOR), inducible nitric oxide synthase (iNOS) and nuclear factor kappa B (NF-κB) pathways, and up-regulating Sirtuin-1 (SIRT-1) and adenosine monophosphate-activated protein kinase (AMPK) pathways, which are critical mechanisms to avoid anomalous inflammatory responses related to oxidative stress and apoptosis(Reference Mocayar Marón, Ferder and Reiter94).

Pro-inflammatory cytokines, interleukins and other inflammatory markers, such as prostaglandins and acute-phase C-reactive protein, have been implicated to play role in the pathophysiology of depression(Reference Berk, Williams and Jacka95Reference Ticinesi, Meschi and Lauretani97). Inflammation leads to increased blood–brain barrier permeability, allowing easier entry of inflammatory molecules into the CNS(Reference Lee and Giuliani98). At a cellular level, it has been observed that tumour necrosis factor α (TNF-α) can induce glutamate release by activated microglia in vitro, leading to excitotoxic damage to neurons(Reference Takeuchi, Jin and Wang99). Some cytokines can directly increase enzymatic activity for converting tryptophan to kynurenine and decreasing the production of serotonin(Reference Capuron, Ravaud and Gualde100Reference Zhang, Terreni and De Simoni102). Considering that macrophages, dendritic cells and activated B and T lymphocytes express 1α-hydroxylase and VDR, VitD could act by modulating the immune response and regulating cytokine expression(Reference Ticinesi, Meschi and Lauretani97,Reference Calton, Keane and Newsholme103) . Moreover, it was demonstrated that the activity of NF-κB, a transcription factor involved in the synthesis of pro-inflammatory cytokines, was inhibited by 1,25(OH)2D3, which helps to maintain the balance of T-helper (Th) cells, inhibiting the production of Th1 and Th17 cytokines and increasing Th2 cytokine synthesis(Reference Bivona, Agnello and Bellia75).

Interestingly, Boontanrart et al. (2016) reported that activated microglia were associated with an increased expression of VitD receptor and Cyp27b1, which encodes the 1α-hydroxylase enzyme for converting 25(OH)D into its active form, thereby enhancing their responsiveness to 25(OH)D. Moreover, activated microglia exposed to 25(OH)D had reduced expression of pro-inflammatory cytokines, interleukin (IL)-6, IL-12 and TNF-α, and increased expression of IL-10. The decrease in pro-inflammatory cytokines was dependent on IL-10 induction of suppressor of cytokine signalling-3 (SOCS3). Therefore, 25(OH)D increases the expression of IL-10, creating a feedback loop via SOCS3 which reduces the pro-inflammatory immune response by activated microglia and probably protects the CNS from damage(Reference Boontanrart, Hall and Spanier104). In agreement with these findings, Lee et al. (2020) showed that VitD signalling in neurons elicits an anti-inflammatory state in microglia. Moreover, the partial deletion of VDR in neurons during early life exacerbates CNS autoimmunity in adult mice. Therefore, by changing the immune response of microglia, VitD may be an interesting mechanism for avoiding a prolonged inflammatory state in the CNS(Reference Lee, Selhorst and Lampe105).

In addition, VDR activation stimulates the expression of many antioxidant genes, such as the nuclear factor erythroid-2 (NRF2), γ-glutamyl transpeptidase (γ-GT), glutamate-cysteine ligase (GCLC), glutathione reductase (GR) and glutathione peroxidase (GPx)(Reference Berridge27). VitD negatively regulates the expression of iNOS in monocyte-derived cells, and increases the activity of γ-GT, an important enzyme in the glutathione pathway(Reference Garcion, Sindji and Leblondel106,Reference Garcion, Sindji and Montero-Menei107) . Reinforcing the modulation of oxidative stress as a mechanism associated with the antidepressant-like effect of VitD, repeated administration of this compound (2·5, 7·5 and 25 µg/kg for 7 d) prevented depressive-like behavior and brain oxidative stress induced by chronic administration of corticosterone (21 d) in male and female mice(Reference Camargo, Dalmagro and Platt108,Reference da Silva Souza, da Rosa and Neis109) . It has been demonstrated that reactive oxygen species (ROS) trigger a variety of molecular cascades that increase the permeability of the blood–brain barrier, allowing inflammatory cytokines to enter the CNS(Reference Neurauter, Schrocksnadel and Scholl-Burgi110). Moreover, it has been well established that inflammation and oxidative stress, which mutually amplify each other, play an important role in the pathophysiology of depression and can be a target for therapeutic strategies(Reference Lindqvist, Dhabhar and James111).

Clinical studies

Nineteen randomised clinical trials using VitD supplementation for depressive symptoms in adults were published up to 2020 (Table 1). Nine studies were double-blinded, and twelve included individuals aged >65 years. Most of the studies were conducted in high-income countries (13/19). Seven studies were conducted with community-dwelling, healthy volunteers or individuals with no specification, and three studies only with VitD-deficient individuals(Reference Zhu, Zhang and Wang112Reference Vieth, Kimball and Hu114). Six included only individuals with the diagnosis of depression, and two with individuals with VitD deficiency and diagnosed depression(Reference Vellekkatt, Menon and Rajappa115,Reference de Koning, Lips and Penninx116) . Considering only the studies that included individuals with a diagnosis of depression (with or without VitD deficiency), 4/8 presented improvement in depressive symptoms after VitD supplementation.

Table 1 Vitamin D supplementation and depression/depressive symptoms: clinical trials with older adults

NA, not assessed; RCT, randomised controlled trial; VitD, vitamin D; PHQ-8, patient health questionnaire depression scale; MINI, mini-international neuropsychiatric interview; HAMD-17, Hamilton depression rating scale-17; RSAS, revised social anhedonia scale; RPAS, revised physical anhedonia scale; HAMA-14, Hamilton anxiety rating scale-14; HDRS-17, Hamilton depression rating scale-17; BDI, Beck depression inventory; PANAS, positive and negative affect schedule; DASS-21, 21-item depression; CES-D, Center for Epidemiological Studies Depression; GDS-15, 15-item geriatric depression scale; MDI, major depression inventory; BDI-II, Beck depression inventory-II; FCPS, Fawcett–Clark pleasure capacity scale; GDS-LF30, long form 30-item GDS; HDRS-24, Hamilton depression rating scale-24; BDI-21, Beck depression inventory-21; HADS-14, hospital anxiety and depression scale; MADRS, Montgomery–sberg depression rating scale.

Seven (7/19) studies reported an improvement in depressive symptoms after VitD supplementation, eleven reported no improvement and one study lacked the power to assess due to sampling size(Reference Aucoin, Cooley and Anand123). Considering the studies that observed depressive symptom improvement, five of seven were conducted with individuals with depression, and one of these (1/7) reported individuals with concomitant depression and VitD deficiency. VitD doses ranged from 600 to 300 000 IU, and the majority (6/7) used VitD doses above the dietary reference intake (DRI) (> 4000 IU/d). VitD doses of 600–4000 IU were used on a daily basis; 20 000–50 000 IU were used weekly; and the effect of a single dose of 150 000–300 000 IU was evaluated.

Compared with the seven studies with positive results, the eleven studies that did not report improvements tended to use lower VitD doses (<4000 IU) and longer periods (from 6 months to 5 years of supplementation). Of the eleven negative studies, only four used higher doses: Sanders et al. (2011) used a single dose of 500 000 IU in the winter for 3–5 years; Dean et al. (2011) used 5000 IU/d for 6 weeks; Kjægaard et al. (2012) used 20 000 IU/week for 6 months; and Gugger et al. (2019) used 24 000 IU or 60 000 IU for 12 months(Reference Kjærgaard, Waterloo and Wang113,Reference Gugger, Marzel and Orav120,Reference Dean, Bellgrove and Hall128,Reference Sanders, Stuart and Williamson129) . The age range was higher in the studies that did not observe any improvement in depressive symptoms (individuals >70 years).

Two meta-analyses have shown controversial results in clinical trials with VitD supplementation. Spedding et al. (2014) showed that VitD supplementation (daily doses of ≥800 IU) could have an effect comparable to that of antidepressants in depressive symptoms(Reference Spedding21). Due to the methodological variability of the studies, the other meta-analysis conducted by Gowda et al. (2015) showed results that did not support this hypothesis(Reference Gowda, Mutowo and Smith131). In addition, a 5-year follow-up study found no potential effect of VitD on the incidence of depression(Reference Okereke, Reynolds and Mischoulon117). Comparing the findings of the published meta-analysis with the studies searched in the present review, we observed that studies that did not observe improvements in depressive symptoms were conducted with older people with no diagnosis of depression, with lower VitD doses and for longer periods of follow-up. On the contrary, studies with positive results were conducted with younger populations with a diagnosis of depression and higher VitD doses for short periods of follow-up.

Key points of pre-clinical and clinical studies

Pre-clinical studies have pointed to the potential and possible effect of vitD on depression. However, despite a considerable number of clinical studies, it has not yet been possible to prove whether VitD can prevent or be used as an adjuvant treatment in depression. The data remain controversial. In addition, it is not possible yet to define which doses/amount of vitamin D would be most appropriate for depression.

Vitamin D and depressive symptoms: evidence from observational studies

Table 2 summarises the information from forty-four observational studies that investigated the relationship between VitD and depression/depressive symptoms in both adults and older adults since 2006.

Table 2. Vitamin D supplementation and depression/depressive symptoms: observational studies with older adults

95 % CI, 95% confidence interval; OR, odds ratio; SE, standard error; ES, effect size; IRR, incidence rate ratio; B, unstandardised beta; RR, relative risk; 25(OH)D, 25-hydroxycholecalciferol; 25(OH)D2, 25-hydroxyvitamin D2; 25(OH)D2, 25-hydroxyvitamin D3; DSM-IV, diagnostic and statistical manual of mental disorders; CIDI, composite international diagnostic interview; IDS-SR, self-report version of the inventory of depressive symptoms; GDS-15, geriatric depression scale with 15 items; CES-D, Center for Epidemiologic Studies Depression scale; HAM-D-17, Hamilton depression rating scale-17 items; HAD, hospital anxiety and depression scale; PHQ-9, patient health questionnaire-9; BDI, Beck depression inventory; CIDI, composite international diagnostic interview; IDS-SR, inventory of depressive symptoms – self-report; SCL, symptom checklist; HDRS, Hamilton depression rating scale; HICDA, hospital international classification of disease adaptation; DASS21, depression anxiety stress scale.

From over 15 years of research published, we observed that most studies included a mixed population with adults and older adults (27/44), were composed of people from cohort studies (27/44) and high-income economies countries (38/44), and used screening scales of depressive symptoms (37/44). The majority of studies performed a cross-sectional (27/44), followed by both a cross-sectional and longitudinal (10/44), and, finally, a longitudinal analysis (7/44). Considering the studies that included only older adults (≥60 years, 17/44), most were composed of people from a cohort (14/17) and performed a cross-sectional (10/17), followed by both a cross-sectional and longitudinal (4/17) and, finally, a longitudinal analysis (3/17). Moreover, only three studies were performed in low- or middle-income countries. This is an important issue because, according to the Mental Health Action Plan 2013–2030, there is an imbalance between research in high- and low/middle-income countries that needs to be corrected to ensure that they have appropriate cultural and economic strategies to respond to mental health needs and priorities(13). One of their main goals is to strengthen information systems, evidence and research on mental health, and it suggests the development of more studies from low/middle-income countries.

It is difficult to compare the main differences between the studies because each study was different in terms of the method used to analyse data, the cut-off point for the classification of serum VitD concentrations and the screening for depressive symptoms or diagnosis for depression. However, an increasing number of studies have found an association between VitD and both depressive symptoms (32/44) and depression (7/44), specifically in those with cross-sectional analyses (24/44 and 7/44, respectively). Considering the studies in which researchers stratified the analysis by sex (7/44), the association was divergent because some authors(Reference de Oliveira, Hirani and Biddulph147,Reference Milaneschi, Shardell and Corsi169) found an association in both sexes, while other studies found an association for women(Reference Toffanello, Sergi and Veronese163,Reference de Koning, Elstgeest and Comijs175) or men(Reference Rhee, Lee and Ahn139,Reference Song, Kim and Rhee154,Reference Imai, Halldorsson and Eiriksdottir161) . In studies that included both adults and older adults, only five (5/27) reported no association(Reference Sahasrabudhe, Lee and Scott136,Reference Granlund, Ramnemark and Andersson138,Reference Husemoen, Ebstrup and Mortensen157,Reference Nanri, Mizoue and Matsushita171,Reference Pan, Lu and Franco172) .

Among the studies that exclusively analysed data of older adults, those that performed a cross-sectional analysis (10/17) found an association between VitD and either depression(Reference Imai, Halldorsson and Eiriksdottir161,Reference Hoogendijk, Lips and Dik173) or depressive symptoms(Reference Ceolin, Matsuo and Confortin135,Reference Yao, Fu and Zhang146,Reference Song, Kim and Rhee154Reference Rocha-Lima, Custódio and Moreira156,Reference Lapid, Cha and Takahashi165,Reference Stewart and Hirani170,Reference Wilkins, Sheline and Roe174) , but two studies that stratified the analysis by sex found an association only for men(Reference Song, Kim and Rhee154,Reference Imai, Halldorsson and Eiriksdottir161) . In studies that performed either longitudinal or cross-sectional and longitudinal analyses combined, the results are controversial. In the longitudinal analysis, one(Reference van den Berg, Marijnissen and van den Brink153) did not find any effect of VitD on the course of depression or remission, while another found a decrease in the score of depression with an increase in VitD(Reference va, n den Berg, Hegeman and van den Brink134), and another(Reference Milaneschi, Shardell and Corsi169) found an increase in depression score for a low level of VitD at 3 and 6 years follow-up in women and 3 years follow-up for men. In the cross-sectional and longitudinal combined analysis, some found a cross-sectional but not longitudinal association(Reference Almeida, Hankey and Yeap159,Reference Chan, Chan and Woo167) , another study(Reference Williams, Sink and Tooze160) did not find an association at baseline and 1 year follow-up, just one found an association at 4 years follow-up and another found a cross-sectional association only for women and not in the follow-up(Reference Toffanello, Sergi and Veronese163). Nevertheless, most of these studies found a higher risk for depression when considering VitD concentrations below 20 ng/ml or 50 nmol/l(Reference Ceolin, Matsuo and Confortin135,Reference Yao, Fu and Zhang146,Reference Song, Kim and Rhee154,Reference Almeida, Hankey and Yeap159,Reference Williams, Sink and Tooze160,Reference Toffanello, Sergi and Veronese163,Reference Milaneschi, Shardell and Corsi169,Reference Wilkins, Sheline and Roe174) . Other studies found higher risk when concentrations were below 10 ng/ml or 30 nmol/l(Reference Rocha-Lima, Custódio and Moreira156,Reference Imai, Halldorsson and Eiriksdottir161,Reference Lapid, Cha and Takahashi165,Reference Stewart and Hirani170) , and two studies found a lower risk for depression in concentrations >36·7 nmol/l(Reference Brouwer-Brolsma, Vaes and van der Zwaluw176) and 92 nmol/l(Reference Chan, Chan and Woo167). Moreover, a meta-analysis with a mixed population showed that an increase of 10 ng/ml in individuals with low serum concentrations of 25(OH)D had a protective effect against depression, with a decrease of 4 % in the risk of depression in cross-sectional studies, and a decrease of 8 % in the incidence of depression in cohort studies(Reference Ju, Lee and Jeong177). In studies involving only the elderly population, the same 10 ng/ml increase in serum 25(OH)D level was associated with a 12 % reduction in the risk of depression(Reference Li, Sun and Wang178).

Key points of observational studies

Despite the controversial results from observational studies, the majority have pointed to a higher risk of depression with low levels of VitD (20 ng/ml or 50 nmol/l). However, the variability in methodology between studies is important to note. At this moment, it is not possible to suggest a possible VitD cut-off point specific for depression. Few studies were carried out with only older adults, as well as in low- and middle-income countries. Few longitudinal studies were carried out to demonstrate causality of depression due to low levels.

Future perspective

Older adults are considered a risk group for both depression and vitamin D deficiency, which justifies further studies to focus on this population. The ageing process is associated with a reduced ability to sustain homoeostasis, which could make elderly people more susceptible to pathological alterations, including neuropsychiatric disorders(Reference Sibille179,Reference Pomatto and Davies180) . Also, women in menopausal transition are at risk of depression due to a lot of changes (i.e. hormone-related context, stressful events in life)(Reference Soares and Shea181). Moreover, older adults with depression present a higher risk of mortality(Reference Brandão, Fontenelle and da Silva182), especially in low- and middle-income countries, and have difficulties accessing treatment(4,Reference Lopes, Hellwig and e Silva183) . Another important factor is related to the adverse effects caused by antidepressant medications and the polypharmacy common in the elderly owing to the concomitance of several pathologies, which can facilitate the discontinuation of treatment(Reference Falci, Mambrini and Castro-Costa184Reference Read, Gee and Diggle186).

Facing the urgency to identify the modifiable risk factors associated with the aetiology of depression, helping with the treatment and prevention of this disorder, it is important to carry out more studies following a proper methodology since we have an important background related to pre-clinical studies. As highlighted by the WHO, these studies need to be developed especially in low- and middle-income countries, since these places have higher prevalence of depression(Reference Whiteford, Ferrari and Degenhardt12,13) . Further, observational studies have pointed to the preventive effect of adequate serum vitamin D concentrations on the development of depressive symptoms. More longitudinal studies have been suggested(Reference Li, Sun and Wang178,Reference Parker, Brotchie and Graham187) to better elucidate the preventive effects of VitD on depression/depressive symptoms.

Besides, the variability in the diagnosis of depression, differences in VitD cut-off reference values and methods for serum VitD analysis could influence those findings that are still controversial(Reference Spedding21,Reference Wong, Ima-Nirwana and Chin188,Reference Jorde and Kubiak189) . Recently, the use of the standardised measurement of VitD proposed by the VitD Standardization Program (VDSP) has been recommended to improve clinical and public health practice, and it is important for future studies to apply this in their methodology(Reference Sempos and Binkley190,Reference Giustina, Adler and Binkley191) . Considering the randomised control trial (RCT) that included the elderly population (>65 years), most of them did not present any improvements in depressive symptoms after VitD supplementation. This could be due to the lower VitD doses used in those studies, and because they were not performed in older individuals diagnosed with depression. This is an important aspect to be addressed in future RCTs.

Conclusion

Overall, this updated review suggests that the monitoring and maintenance of adequate VitD concentrations is crucial, especially in older adults, a population at risk for both VitD deficiency and depression. Several pre-clinical, clinical and observational studies have suggested that VitD could have a beneficial effect on depression/depressive symptoms due to its genomic and non-genomic actions in many pathways involved in the pathophysiology of depression.

Although studies presented controversial results, clinical studies have shown that older adults with depression/depressive symptoms could benefit from higher doses of VitD supplementation for short periods. However, more RCTs are needed to confirm which doses and for how long the treatment is needed to achieve the greatest benefit. From the observational studies, the results are still controversial, but the majority have reported an association between low serum concentrations of VitD and high risk for depression/depressive symptoms in older adults, pointing to a possible preventive effect of VitD. Additional studies with prospective designs, especially in low- and middle-income countries, will possibly help to better elucidate the impact of deficient VitD status for mental health in adulthood and, consequently, for the elderly.

Supplementary material

For supplementary material accompanying this paper visit https://doi.org/10.1017/S0954422422000026

Financial support

This work was supported by the Coordenação de Aperfeiçoamento de Pessoal de Nível Superior – CAPES/Brasil (Finance Code 001).

Conflict of interest

None.

Authorship

All authors contributed to conception of this study. Material was prepared and the first draft of the manuscript was written by G.C. and J.D.M., and all authors commented on previous versions of the manuscript. All authors read and approved the final manuscript.

References

American Psychiatric Association (2014) Manual diagnóstico e estatístico de transtornos mentais: DSM-5 [Diagnostic and Statistical Manual of MentalDisorders. DSM-5]. 5 th ed. Porto Alegre: Artmed.Google Scholar
Chisholm, D, Sweeny, K, Sheehan, P, et al. (2016) Scaling-up treatment of depression and anxiety: a global return on investment analysis. Lancet Psychiat 3, 415424.CrossRefGoogle ScholarPubMed
Olesen, J, Gustavsson, A, Svensson, M, et al. (2012) The economic cost of brain disorders in Europe. Eur J Neurol 19, 155162.CrossRefGoogle ScholarPubMed
World Health Organization (2020) Depression [Internet]. [cited 2020 Feb 13]. Available from: https://www.who.int/news-room/fact-sheets/detail/depression Google Scholar
World Health Organization (2017) Depression and Other Common Mental Disorders: Global Health Estimates [Internet]. Geneva: World Health Organization, p. 27. Available from: http://www.who.int/mental_health/management/depression/prevalence_global_health_estimates/en/ Google Scholar
World Health Organization (2017) Mental health of older adults [Internet]. [cited 2021 Apr 22]. Available from: https://www.who.int/news-room/fact-sheets/detail/mental-health-of-older-adults Google Scholar
Global Health Metrics (2019) Depressive Disorders — Level 3 Cause [Internet]. Institute for Health Metrics and Evaluation. [cited 2021 Jan 6]. Available from: http://www.healthdata.org/results/gbd_summaries/2019/depressive-disorders-level-3-cause Google Scholar
James, SL, Abate, D, Abate, KH, et al. (2018) Global, regional, and national incidence, prevalence, and years lived with disability for 354 diseases and injuries for 195 countries and territories, 1990–2017: a systematic analysis for the Global Burden of Disease Study 2017. Lancet; 392, 17891858.CrossRefGoogle Scholar
DiLuca, M & Olesen, J (2014) The cost of brain diseases: a burden or a challenge? Neuron 82, 12051208.CrossRefGoogle ScholarPubMed
Knapp, M & Wong, G (2020) Economics and mental health: the current scenario. World Psychiatry 19, 314.CrossRefGoogle ScholarPubMed
World Health Organization, editor. (2018) Mental Health Atlas 2017 [Internet]. Geneva, Switzerland: World Health Organization. Available from: https://apps.who.int/iris/bitstream/handle/10665/272735/9789241514019-eng.pdf Google Scholar
Whiteford, HA, Ferrari, AJ, Degenhardt, L, et al. (2015) The global burden of mental, neurological and substance use disorders: an analysis from the Global Burden of Disease Study 2010. PloS One 10, e0116820.CrossRefGoogle ScholarPubMed
World Health Organization (2013) WHO | Mental Health Action Plan 2013–2020 [Internet]. WHO. [cited 2020 Feb 13]. Available from: http://www.who.int/entity/mental_health/publications/action_plan/en/index.html Google Scholar
Li, M, D’Arcy, C & Meng, X (2016) Maltreatment in childhood substantially increases the risk of adult depression and anxiety in prospective cohort studies: systematic review, meta-analysis, and proportional attributable fractions. Psychol Med 46, 717730.CrossRefGoogle ScholarPubMed
Andrade, FCD, Wu, F, Lebrão, ML, et al. (2016) Life expectancy without depression increases among Brazilian older adults. Rev Saúde Pública 50, 12.CrossRefGoogle ScholarPubMed
Kaltenboeck, A & Harmer, C (2018) The neuroscience of depressive disorders: a brief review of the past and some considerations about the future. Brain Neurosci Adv SAGE Publications Ltd STM; 2, 2398212818799269.CrossRefGoogle ScholarPubMed
Otte, C, Gold, SM, Penninx, BW, et al. (2016) Major depressive disorder. Nat Rev Dis Primers 2, 120.CrossRefGoogle ScholarPubMed
Lai, JS, Hiles, S, Bisquera, A, et al. (2014) A systematic review and meta-analysis of dietary patterns and depression in community-dwelling adults. Am J Clin Nutr 99, 181197.CrossRefGoogle ScholarPubMed
Camargo, A, Dalmagro, AP, Rikel, L, et al. (2018) Cholecalciferol counteracts depressive-like behavior and oxidative stress induced by repeated corticosterone treatment in mice. Eur J Pharmacol 833, 451461.CrossRefGoogle ScholarPubMed
Fedotova, J, Dudnichenko, T, Kruzliak, P, et al. (2016) Different effects of vitamin D hormone treatment on depression-like behavior in the adult ovariectomized female rats. Biomed Pharmacother 84, 18651872.CrossRefGoogle ScholarPubMed
Spedding, S (2014) Vitamin D and depression: a systematic review and meta-analysis comparing studies with and without biological flaws. Nutrients 6, 15011518.CrossRefGoogle ScholarPubMed
Bikle, DD (2012) Vitamin D and the skin: physiology and pathophysiology. Rev Endocr Metab Disord 13, 319.CrossRefGoogle ScholarPubMed
Jäpelt, RB & Jakobsen, J (2013) Vitamin D in plants: a review of occurrence, analysis, and biosynthesis. Front Plant Sci Frontiers Media SA; [cited 2020 Apr 2]; 4. Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3651966/ CrossRefGoogle ScholarPubMed
Johnson, EJ & Mohn, ES (2015) Fat-soluble vitamins. Nutr Prim Care Provider 111, 3844.CrossRefGoogle ScholarPubMed
Norman, AW (2012) The history of the discovery of vitamin D and its daughter steroid hormone. ANM 61, 199206.Google ScholarPubMed
Umar, M, Sastry, KS & Chouchane, AI (2018) Role of vitamin D beyond the skeletal function: a review of the molecular and clinical studies. Int J Mol Sci [cited 2020 Sep 17]; 19. Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6032242/ CrossRefGoogle ScholarPubMed
Berridge, MJ (2017) Vitamin D and depression: cellular and regulatory mechanisms. Pharmacol Rev 69, 8092.CrossRefGoogle ScholarPubMed
Kesby, JP, Eyles, DW, Burne, THJ, et al. (2011) The effects of vitamin D on brain development and adult brain function. Mol Cell Endocrinol 347, 121127.CrossRefGoogle ScholarPubMed
Mayne, PE & Burne, THJ (2019) Vitamin D in synaptic plasticity, cognitive function, and neuropsychiatric illness. Trends Neurosci 42, 293306.CrossRefGoogle ScholarPubMed
Smaga, I, Niedzielska, E, Gawlik, M, et al. (2015) Oxidative stress as an etiological factor and a potential treatment target of psychiatric disorders. Part 2. Depression, anxiety, schizophrenia and autism. Pharmacol Rep. 67, 569580.CrossRefGoogle Scholar
Landel, V, Stephan, D, Cui, X, et al. (2018) Differential expression of vitamin D-associated enzymes and receptors in brain cell subtypes. J Steroid Biochem Mol Biol 177, 129134.CrossRefGoogle ScholarPubMed
Palacios, C & Gonzalez, L (2014) Is vitamin D deficiency a major global public health problem? J Steroid Biochem Mol Biol 144, Pt A, 138145.CrossRefGoogle ScholarPubMed
Ganji, V, Zhang, X & Tangpricha, V (2012) Serum 25-hydroxyvitamin D concentrations and prevalence estimates of hypovitaminosis D in the U.S. population based on assay-adjusted data. J Nutr 142, 498507.CrossRefGoogle ScholarPubMed
Whiting, SJ, Langlois, KA, Vatanparast, H, et al. (2011) The vitamin D status of Canadians relative to the 2011 Dietary Reference intakes: an examination in children and adults with and without supplement use. Am J Clin Nutr 94, 128135.CrossRefGoogle Scholar
Lu, H-K, Zhang, Z, Ke, Y-H, et al. (2012) High prevalence of vitamin D insufficiency in China: relationship with the levels of parathyroid hormone and markers of bone turnover. PloS One 7, e47264.CrossRefGoogle Scholar
Pereira-Santos, M, Santos, JYG dos, Carvalho, GQ, et al. (2019) Epidemiology of vitamin D insufficiency and deficiency in a population in a sunny country: geospatial meta-analysis in Brazil. Crit Rev Food Sci Nutr 59, 21022109.CrossRefGoogle Scholar
Marwaha, RK, Tandon, N, Garg, MK, et al. (2011) Vitamin D status in healthy Indians aged 50 years and above. J Assoc Phys India 59, 706709.Google ScholarPubMed
Sud, SR, Montenegro-Bethancourt, G, Bermúdez, OI, et al. (2010) Older Mayan residents of the western highlands of Guatemala lack sufficient levels of vitamin D. Nutr Res 30, 739746.CrossRefGoogle ScholarPubMed
Bouillon, R (2017) Comparative analysis of nutritional guidelines for vitamin D. Nat Rev Endocrinol Nature Publishing Group; 13, 466479.CrossRefGoogle ScholarPubMed
Sempos, CT, Heijboer, AC, Bikle, DD, et al. (2018) Vitamin D assays and the definition of hypovitaminosis D: results from the First International Conference on Controversies in Vitamin D. Br J Clin Pharmacol 84, 21942207.CrossRefGoogle ScholarPubMed
Arabi, A, El Rassi, R & El-Hajj Fuleihan, G (2010) Hypovitaminosis D in developing countries—prevalence, risk factors and outcomes. Nat Rev Endocrinol 6, 550561.CrossRefGoogle ScholarPubMed
Feizabad, E, Hossein-Nezhad, A, Maghbooli, Z, et al. (2017) Impact of air pollution on vitamin D deficiency and bone health in adolescents. Arch Osteoporos 12, 34.CrossRefGoogle ScholarPubMed
Amrein, K, Scherkl, M, Hoffmann, M, et al. (2020) Vitamin D deficiency 2.0: an update on the current status worldwide. Eur J Clin Nutr Nature Publishing Group 74, 14981513.CrossRefGoogle ScholarPubMed
Cesari, M, Incalzi, RA, Zamboni, V, et al. (2011) Vitamin D hormone: a multitude of actions potentially influencing the physical function decline in older persons. Geriatr Gerontol Int 11, 133142.CrossRefGoogle ScholarPubMed
Holick, MF (2006) High prevalence of vitamin D inadequacy and implications for health. Mayo Clin Proc 81, 353373.CrossRefGoogle ScholarPubMed
Luo, J, Quan, Z, Lin, S, et al. (2018) The association between blood concentration of 25- hydroxyvitamin D and sarcopenia: a meta-analysis. Asia Pac J Clin Nutr 27, 12581270.Google ScholarPubMed
Dudenkov, DV, Mara, KC, Petterson, TM, et al. (2018) Serum 25-Hydroxyvitamin D values and risk of all-cause and cause-specific mortality: a population-based cohort study. Mayo Clin Proc 93, 721730.CrossRefGoogle ScholarPubMed
Tian, XQ & Holick, MF (1995) Catalyzed thermal isomerization between previtamin D3 and vitamin D3 via beta-cyclodextrin complexation. J Biol Chem 270, 87068711.CrossRefGoogle ScholarPubMed
Bikle, D & Christakos, S (2020) New aspects of vitamin D metabolism and action — addressing the skin as source and target. Nat Rev Endocrinol Nature Publishing Group; 16, 234252.CrossRefGoogle ScholarPubMed
Wacker, M & Holick, MF (2013) Sunlight and vitamin D: a global perspective for health. Dermato-Endocrinol 5, 51108.CrossRefGoogle ScholarPubMed
Bouillon, R, Marcocci, C, Carmeliet, G, et al. (2019) Skeletal and extraskeletal actions of vitamin D: current evidence and outstanding questions. Endocr Rev Oxford Academic; 40, 11091151.CrossRefGoogle ScholarPubMed
Reboul, E (2015) Intestinal absorption of vitamin D: from the meal to the enterocyte. Food Funct The Royal Society of Chemistry; 6, 356362.CrossRefGoogle Scholar
Gil, Á, Plaza-Diaz, J & Mesa, MD (2018) Vitamin D: classic and novel actions. ANM Karger Publishers; 72, 8795.Google ScholarPubMed
Holick, MF (2004) Sunlight and vitamin D for bone health and prevention of autoimmune diseases, cancers, and cardiovascular disease. Am J Clin Nutr Oxford Academic; 80, 1678S1688S.CrossRefGoogle ScholarPubMed
Prabhu, AV, Luu, W, Li, D, et al. (2016) DHCR7: a vital enzyme switch between cholesterol and vitamin D production. Prog Lipid Res 64, 138151.CrossRefGoogle ScholarPubMed
Wacker, M & Holick, MF (2013) Vitamin D—effects on skeletal and extraskeletal health and the need for supplementation. Nutrients 5, 111148.CrossRefGoogle ScholarPubMed
Holick, MF (2007) Vitamin D deficiency. N Engl J Med 357, 266281.CrossRefGoogle ScholarPubMed
Bikle, DD, Patzek, S & Wang, Y (2018) Physiologic and pathophysiologic roles of extra renal CYP27b1: case report and review. Bone Rep 8, 255267.CrossRefGoogle ScholarPubMed
Bouillon, R, Carmeliet, G, Verlinden, L, et al. (2008) Vitamin D and human health: lessons from vitamin D receptor null mice. Endocr Rev 29, 726776.CrossRefGoogle ScholarPubMed
Haussler, MR, Jurutka, PW, Mizwicki, M, et al. (2011) Vitamin D receptor (VDR)-mediated actions of 1α,25(OH)2vitamin D3: genomic and non-genomic mechanisms. Best Pract Res Clin Endocrinol Metab 25, 543559.CrossRefGoogle ScholarPubMed
Chen, J, Olivares-Navarrete, R, Wang, Y, et al. (2010) Protein-disulfide Isomerase-associated 3 (Pdia3) mediates the membrane response to 1,25-dihydroxyvitamin D3 in osteoblasts. J Biol Chem American Society for Biochemistry and Molecular Biology; 285, 3704137050.CrossRefGoogle ScholarPubMed
Boyan, BD, Chen, J & Schwartz, Z (2012) Mechanism of Pdia3-dependent 1α,25-dihydroxy vitamin D3 signaling in musculoskeletal cells. Steroids 77, 892896.CrossRefGoogle ScholarPubMed
Zmijewski, MA & Carlberg, C (2020) Vitamin D receptor(s): in the nucleus but also at membranes? Exp Dermatol 29, 876884.CrossRefGoogle ScholarPubMed
Doroudi, M, Plaisance, MC, Boyan, BD, et al. (2015) Membrane actions of 1α,25(OH)2D3 are mediated by Ca2+/calmodulin-dependent protein kinase II in bone and cartilage cells. J Steroid Biochem Mol Biol 145, 6574.CrossRefGoogle Scholar
Stumpf, WE, Sar, M, Clark, SA, et al. (1982) Brain target sites for 1,25-dihydroxyvitamin D3. Science American Association for the Advancement of Science; 215, 14031405.CrossRefGoogle ScholarPubMed
DeLuca, GC, Kimball, SM, Kolasinski, J, et al. (2013) Review: I role of vitamin D in nervous system health and disease. Neuropathol Appl Neurobiol John Wiley & Sons, Ltd; 39, 458484.CrossRefGoogle ScholarPubMed
Eyles, DW, Smith, S, Kinobe, R, et al. (2005) Distribution of the vitamin D receptor and 1α-hydroxylase in human brain. J Chem Neuroanat 29, 2130.CrossRefGoogle ScholarPubMed
Pardridge, WM, Sakiyama, R & Coty, WA (1985) Restricted transport of vitamin D and A derivatives through the rat blood-brain barrier. J Neurochem 44, 11381141.CrossRefGoogle Scholar
Eyles, DW, Feron, F, Cui, X, et al. (2009) Developmental vitamin D deficiency causes abnormal brain development. Psychoneuroendocrinology 34, S247S257.CrossRefGoogle ScholarPubMed
Cui, X, Gooch, H, Petty, A, et al. (2017) Vitamin D and the brain: genomic and non-genomic actions. Mol Cell Endocrinol 453, 131143.CrossRefGoogle ScholarPubMed
Warsh, JJ, Andreopoulos, S & Li, PP (2004) Role of intracellular calcium signaling in the pathophysiology and pharmacotherapy of bipolar disorder: current status. Clin Neurosci Res 4, 201213.CrossRefGoogle Scholar
Yuan, JP, Kiselyov, K, Shin, DM, et al. (2003) Homer binds TRPC family channels and is required for gating of TRPC1 by IP3 receptors. Cell 114, 777789.CrossRefGoogle ScholarPubMed
Kandel, ER, Schwartz, JH, Jessell, TM, et al. (2014) Princípios de neurociências [Principles of Neuroscience], 5th ed. Porto Alegre: AMGH.Google Scholar
Croarkin, PE, Levinson, AJ & Daskalakis, ZJ (2011) Evidence for GABAergic inhibitory deficits in major depressive disorder. Neurosci Biobehav Rev 35, 818825.CrossRefGoogle ScholarPubMed
Bivona, G, Agnello, L, Bellia, C, et al. (2019) Non-Skeletal Activities of Vitamin D: From Physiology to Brain Pathology. Medicina (Kaunas) [cited 2020 Oct 18]; 55. Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6680897/ Google ScholarPubMed
Domínguez-López, S, Howell, R & Gobbi, G (2012) Characterization of serotonin neurotransmission in knockout mice: implications for major depression. Rev Neurosci De Gruyter; 23, 429443.CrossRefGoogle ScholarPubMed
Fakhoury, M (2016) Revisiting the serotonin hypothesis: implications for major depressive disorders. Mol Neurobiol 53, 27782786.CrossRefGoogle ScholarPubMed
Ogawa, S, Fujii, T, Koga, N, et al. (2014) Plasma L-tryptophan concentration in major depressive disorder: new data and meta-analysis. J Clin Psychiatry 75, e906e915.CrossRefGoogle ScholarPubMed
Kaneko, I, Sabir, MS, Dussik, CM, et al. (2015) 1,25-Dihydroxyvitamin D regulates expression of the tryptophan hydroxylase 2 and leptin genes: implication for behavioral influences of vitamin D. FASEB J 29, 40234035.CrossRefGoogle ScholarPubMed
Patrick, RP & Ames, BN (2015) Vitamin D and the omega-3 fatty acids control serotonin synthesis and action, part 2: relevance for ADHD, bipolar disorder, schizophrenia, and impulsive behavior. FASEB J Federation of American Societies for Experimental Biology; 29, 22072222.CrossRefGoogle ScholarPubMed
Sabir, MS, Haussler, MR, Mallick, S, et al. (2018) 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 [cited 2020 Oct 1]; 13. Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6042449/ CrossRefGoogle ScholarPubMed
Cui, X, Pelekanos, M, Liu, P-Y, et al. (2013) The vitamin D receptor in dopamine neurons; its presence in human substantia nigra and its ontogenesis in rat midbrain. Neuroscience 236, 7787.CrossRefGoogle ScholarPubMed
Cui, X, Pertile, R, Liu, P, et al. (2015) Vitamin D regulates tyrosine hydroxylase expression: N-cadherin a possible mediator. Neuroscience 304, 90100.CrossRefGoogle ScholarPubMed
Sedaghat, K, Yousefian, Z, Vafaei, AA, et al. (2019) Mesolimbic dopamine system and its modulation by vitamin D in a chronic mild stress model of depression in the rat. Behav Brain Res 356, 156169.CrossRefGoogle Scholar
McCarty, DE, Reddy, A, Keigley, Q, et al. (2012) Vitamin D, race, and excessive daytime sleepiness. J Clin Sleep Med 8, 693697.CrossRefGoogle ScholarPubMed
Mosavat, M, Smyth, A, Arabiat, D, et al. (2020) Vitamin D and sleep duration: is there a bidirectional relationship? Horm Mol Biol Clin Investig 41.Google Scholar
Jones, KS, Redmond, J, Fulford, AJ, et al. (2017) Diurnal rhythms of vitamin D binding protein and total and free vitamin D metabolites. J Steroid Biochem Mol Biol 172, 130135.CrossRefGoogle ScholarPubMed
Muscogiuri, G, Barrea, L, Scannapieco, M, et al. (2019) The lullaby of the sun: the role of vitamin D in sleep disturbance. Sleep Med 54, 262265.CrossRefGoogle ScholarPubMed
Lucock, M, Jones, P, Martin, C, et al. (2015) Vitamin D: beyond metabolism. J Evid Based Complement Altern Med SAGE Publications Inc STM; 20, 310322.CrossRefGoogle ScholarPubMed
Romano, F, Muscogiuri, G, Di Benedetto, E, et al. (2020) Vitamin D and sleep regulation: is there a role for vitamin D? Curr Pharm Des 26, 24922496.CrossRefGoogle Scholar
Dibner, C, Schibler, U & Albrecht, U (2010) The mammalian circadian timing system: organization and coordination of central and peripheral clocks. Annu Rev Physiol 72, 517549.CrossRefGoogle ScholarPubMed
Stehle, JH, von Gall, C & Korf, H-W (2003) Melatonin: a clock-output, a clock-input. J Neuroendocrinol 15, 383389.CrossRefGoogle ScholarPubMed
Zhao, D, Yu, Y, Shen, Y, et al. (2019) Melatonin synthesis and function: evolutionary history in animals and plants. Front Endocrinol [Internet]. Frontiers; [cited 2021 May 4]; 10. Available from: https://www.frontiersin.org/articles/10.3389/fendo.2019.00249/full CrossRefGoogle ScholarPubMed
Mocayar Marón, FJ, Ferder, L, Reiter, RJ, et al. (2020) Daily and seasonal mitochondrial protection: unraveling common possible mechanisms involving vitamin D and melatonin. J Steroid Biochem Mol Biol 199, 105595.CrossRefGoogle ScholarPubMed
Berk, M, Williams, LJ, Jacka, FN, et al. (2013) So depression is an inflammatory disease, but where does the inflammation come from? BMC Med 11, 200.CrossRefGoogle ScholarPubMed
Swardfager, W, Rosenblat, JD, Benlamri, M, et al. (2016) Mapping inflammation onto mood: inflammatory mediators of Anhedonia. Neurosci Biobehav Rev 64, 148166.CrossRefGoogle ScholarPubMed
Ticinesi, A, Meschi, T, Lauretani, F, et al. (2016) Nutrition and inflammation in older individuals: focus on vitamin D, n-3 polyunsaturated fatty acids and whey proteins. Nutrients [Internet]. [cited 2020 Apr 14]; 8. Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4848655/ CrossRefGoogle ScholarPubMed
Lee, C-H & Giuliani, F (2019) The role of inflammation in depression and fatigue. Front Immunol [Internet] Jul 19 [cited 2020 Oct 9]; 10. Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6658985/ CrossRefGoogle Scholar
Takeuchi, H, Jin, S, Wang, J, et al. (2006) Tumor necrosis factor-α induces neurotoxicity via glutamate release from hemichannels of activated microglia in an Autocrine Manner. J Biol Chem American Society for Biochemistry and Molecular Biology; 281, 2136221368.CrossRefGoogle Scholar
Capuron, L, Ravaud, A, Gualde, N, et al. (2001) Association between immune activation and early depressive symptoms in cancer patients treated with interleukin-2-based therapy. Psychoneuroendocrinology 26, 797808.CrossRefGoogle ScholarPubMed
Capuron, L, Neurauter, G, Musselman, DL, et al. (2003) Interferon-alpha–induced changes in tryptophan metabolism: relationship to depression and paroxetine treatment. Biol Psychiatry 54, 906914.CrossRefGoogle ScholarPubMed
Zhang, J, Terreni, L, De Simoni, MG, et al. (2001) Peripheral interleukin-6 administration increases extracellular concentrations of serotonin and the evoked release of serotonin in the rat striatum. Neurochem Int 38, 303308.CrossRefGoogle ScholarPubMed
Calton, EK, Keane, KN, Newsholme, P, et al. (2015) The impact of vitamin D levels on inflammatory status: a systematic review of immune cell studies. PLoS One [cited 2020 Apr 17]; 10. Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4631349/ CrossRefGoogle ScholarPubMed
Boontanrart, M, Hall, SD, Spanier, JA, et al. (2016) Vitamin D3 alters microglia immune activation by an IL-10 dependent SOCS3 mechanism. J Neuroimmunol 292, 126136.CrossRefGoogle ScholarPubMed
Lee, PW, Selhorst, A, Lampe, SG, et al. (2020) Neuron-specific vitamin D signaling attenuates microglia activation and CNS autoimmunity. Front Neurol 11, 19.CrossRefGoogle ScholarPubMed
Garcion, E, Sindji, L, Leblondel, G, et al. (1999) 1,25-dihydroxyvitamin D3 regulates the synthesis of gamma-glutamyl transpeptidase and glutathione levels in rat primary astrocytes. J Neurochem 73, 859866.CrossRefGoogle ScholarPubMed
Garcion, E, Sindji, L, Montero-Menei, C, et al. (1998) Expression of inducible nitric oxide synthase during rat brain inflammation: regulation by 1,25-dihydroxyvitamin D3. Glia 22, 282294.3.0.CO;2-7>CrossRefGoogle ScholarPubMed
Camargo, A, Dalmagro, AP, Platt, N, et al. (2020) Cholecalciferol abolishes depressive-like behavior and hippocampal glucocorticoid receptor impairment induced by chronic corticosterone administration in mice. Pharmacol Biochem Behav 196, 172971.CrossRefGoogle ScholarPubMed
da Silva Souza, SV, da Rosa, PB, Neis, VB, et al. (2020) Effects of cholecalciferol on behavior and production of reactive oxygen species in female mice subjected to corticosterone-induced model of depression. Naunyn-Schmiedeberg’s Arch Pharmacol 393, 111120.CrossRefGoogle ScholarPubMed
Neurauter, G, Schrocksnadel, K, Scholl-Burgi, S, et al. (2008) Chronic immune stimulation correlates with reduced phenylalanine turnover. Curr Drug Metab 9, 622627.CrossRefGoogle ScholarPubMed
Lindqvist, D, Dhabhar, FS, James, SJ, et al. (2017) Oxidative stress, inflammation and treatment response in major depression. Psychoneuroendocrinology 76, 197205.CrossRefGoogle ScholarPubMed
Zhu, C, Zhang, Y, Wang, T, et al. (2020) Vitamin D supplementation improves anxiety but not depression symptoms in patients with vitamin D deficiency. Brain Behav 10, e01760.CrossRefGoogle Scholar
Kjærgaard, M, Waterloo, K, Wang, CEA, et al. (2012) Effect of vitamin D supplement on depression scores in people with low levels of serum 25-hydroxyvitamin D: nested case—control study and randomised clinical trial. Br J Psychiatry Cambridge University Press; 201, 360368.CrossRefGoogle ScholarPubMed
Vieth, R, Kimball, S, Hu, A, et al. (2004) Randomized comparison of the effects of the vitamin D3 adequate intake versus 100 mcg (4000 IU) per day on biochemical responses and the wellbeing of patients. Nutr J 3, 8.CrossRefGoogle ScholarPubMed
Vellekkatt, F, Menon, V, Rajappa, M, et al. (2020) 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 129, 250256.CrossRefGoogle ScholarPubMed
de Koning, EJ, Lips, P, Penninx, BWJH, et al. (2019) Vitamin D supplementation for the prevention of depression and poor physical function in older persons: the D-Vitaal study, a randomized clinical trial. Am J Clin Nutr 110, 11191130.CrossRefGoogle ScholarPubMed
Okereke, OI, Reynolds, CF, Mischoulon, D, et al. (2020) Effect of long-term vitamin D3 supplementation vs placebo on risk of depression or clinically relevant depressive symptoms and on change in mood scores: a randomized clinical trial. JAMA 324, 471480.CrossRefGoogle ScholarPubMed
Alghamdi, S, Alsulami, N, Khoja, S, et al. (2020) Vitamin D supplementation ameliorates severity of major depressive disorder. J Mol Neurosci 70, 230235.CrossRefGoogle ScholarPubMed
Zajac, IT, Barnes, M, Cavuoto, P, et al. (2020) The effects of vitamin D-enriched mushrooms and vitamin D3 on cognitive performance and mood in healthy elderly adults: a randomised, double-blinded, placebo-controlled trial. Nutrients 12, 3847.CrossRefGoogle ScholarPubMed
Gugger, A, Marzel, A, Orav, EJ, et al. (2019) Effect of monthly high-dose vitamin D on mental health in older adults: secondary analysis of a RCT. J Am Geriatr Soc 67, 12111217.CrossRefGoogle ScholarPubMed
Alavi, NM, Khademalhoseini, S, Vakili, Z, et al. (2019) Effect of vitamin D supplementation on depression in elderly patients: a randomized clinical trial. Clin Nutr Elsevier; 38, 20652070.CrossRefGoogle ScholarPubMed
Hansen, JP, Pareek, M, Hvolby, A, et al. (2019) Vitamin D3 supplementation and treatment outcomes in patients with depression (D3-vit-dep). BMC Res Notes 12, 203.CrossRefGoogle ScholarPubMed
Aucoin, M, Cooley, K, Anand, L, et al. (2018) Adjunctive vitamin D in the treatment of non-remitted depression: lessons from a failed clinical trial. Complement Ther Med 36, 3845.CrossRefGoogle ScholarPubMed
Yalamanchili, V & Gallagher, JC (2018) Dose ranging effects of vitamin D3 on the geriatric depression score: a clinical trial. J Steroid Biochem Mol Biol 178, 6064.CrossRefGoogle ScholarPubMed
Mozaffari-Khosravi, H, Nabizade, L, Yassini-Ardakani, SM, et al. (2013) 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 33, 378385.CrossRefGoogle ScholarPubMed
Khoraminya, N, Tehrani-Doost, M, Jazayeri, S, et al. (2013) Efeitos terapêuticos da vitamina D como terapia adjuvantunctionaltina em pacientes com transtounctionalsivo maior [Therapeutic effects of vitamin Das adjunctive therapy to fluoxetine in patients with major depressive disorder]. Aust N Z J Psychiatry 47, 271275.CrossRefGoogle Scholar
Bertone-Johnson, ER, Powers, SI, Spangler, L, et al. (2012) Vitamin D supplementation and depression in the women’s health initiative calcium and vitamin D trial. Am J Epidemiol 176, 113.CrossRefGoogle ScholarPubMed
Dean, AJ, Bellgrove, MA, Hall, T, et al. (2011) Effects of vitamin D supplementation on cognitive and emotional functioning in young adults – a randomised controlled trial. PLOS One Public Library of Science; 6, e25966.CrossRefGoogle ScholarPubMed
Sanders, KM, Stuart, AL, Williamson, EJ, et al. (2011) Annual high-dose vitamin D3 and mental well-being: randomised controlled trial. Br J Psychiatry Cambridge University Press; 198, 357364.CrossRefGoogle ScholarPubMed
Jorde, R, Sneve, M, Figenschau, Y, et al. (2008) Effects of vitamin D supplementation on symptoms of depression in overweight and obese subjects: randomized double blind trial. J Intern Med 264, 599609.CrossRefGoogle ScholarPubMed
Gowda, U, Mutowo, MP, Smith, BJ, et al. (2015) Vitamin D supplementation to reduce depression in adults: meta-analysis of randomized controlled trials. Nutrition 31, 421429.CrossRefGoogle ScholarPubMed
Di Gessa, G, Biddulph, JP, Zaninotto, P, et al. (2021) Changes in vitamin D levels and depressive symptoms in later life in England. Sci Rep 11, 7724.CrossRefGoogle ScholarPubMed
Mulugeta, A, Lumsden, A & Hyppönen, E (2021) Relationship between Serum 25(OH)D and depression: causal evidence from a bi-directional Mendelian randomization study. Nutrients Multidisciplinary Digital Publishing Institute; 13, 109.CrossRefGoogle Scholar
va, n den Berg, KS, Hegeman, JM, van den Brink, RHS, et al. (2021) A prospective study into change of vitamin D levels, depression and frailty among depressed older persons. Int J Geriatr Psychiatry 36, 10291036.Google Scholar
Ceolin, G, Matsuo, LH, Confortin, SC, et al. (2020) Lower serum 25-hydroxycholecalciferol is associated with depressive symptoms in older adults in Southern Brazil. Nutr J 19, 123.CrossRefGoogle ScholarPubMed
Sahasrabudhe, N, Lee, JS, Scott, TM, et al. (2020) Serum vitamin D and depressive symptomatology among Boston-area Puerto Ricans. J Nutr 150, 32313240.CrossRefGoogle ScholarPubMed
Köhnke, C, Herrmann, M & Berger, K (2020) Associations of major depressive disorder and related clinical characteristics with 25-hydroxyvitamin D levels in middle-aged adults. Nutr Neurosci Taylor & Francis; 0, 110.Google Scholar
Granlund, LE, Ramnemark, AK, Andersson, C, et al. (2020) Vitamin D status was not associated with anxiety, depression, or health-related quality of life in Middle Eastern and African-born immigrants in Sweden. Nutr Res 75, 109118.CrossRefGoogle ScholarPubMed
Rhee, SJ, Lee, H & Ahn, YM (2020) Serum vitamin D concentrations are associauncwith depressive symptoms in mIthe Sixth Korea National Health and Nutrition Examination Survey 2014. Front Psychiatry. Frontiers; [cited 2021 Mar 25]; 11. Available from: https://www.frontiersin.org/articles/10.3389/fpsyt.2020.00756/full CrossRefGoogle Scholar
Bigman, G (2020) Vitamin D metabolites, D3 and D2, and their independent associations with depression symptoms among adults in the United States. Null Taylor & Francis; 19. doi: 10.1080/1028415X.2020.1794422.Google ScholarPubMed
Ronaldson, A, Arias de la Torre, J, Gaughran, F, et al. (2020) Prospective associations between vitamin D and depression in middle-aged adults: findings from the UK Biobank cohort. Psychol Med 21, 19.Google Scholar
Briggs, R, McCarroll, K, O’Halloran, A, et al. (2019) Vitamin D deficiency is assunced with an increased likelihood of incident depression in community-dwelling older adults. J Am Med Dir Assoc 20, 517523.CrossRefGoogle ScholarPubMed
Elstgeest, LEM, de Koning, EJ, Brouwer, IA, et al. (2018) Change in serum 25-hydroxyvitamin D and parallel change in depressive symptoms in Dutch older adults. Eur J Endocrinol 179, 239249.CrossRefGoogle ScholarPubMed
Sherchand, O, Sapkota, N, Chaudhari, RK, et al. (2018) Association between vitamin D deficiency and depression in Nepalese population. Psychiatry Res 267, 266271.CrossRefGoogle ScholarPubMed
Vidgren, M, Virtanen, JK, Tolmunen, T, et al. (2018) Serum concentrations of 25-hydroxyvitamin D and depression in a general middle-aged to elderly population in Finland. J Nutr Health Aging 22, 159164.CrossRefGoogle Scholar
Yao, Y, Fu, S, Zhang, H, et al. (2018) The prevalence of depressive symptoms in Chinese longevous persons and its correlation with vitamin D status. BMC Geriatr 18, 198.CrossRefGoogle ScholarPubMed
de Oliveira, C, Hirani, V & Biddulph, JP (2018) Associations between vitamin D levels and depressive symptoms in later life: euncce from the English Longitudinal Study of Ageing (ELSA). J Gerontol A Biol Sci Med Sci 73, 13771382.CrossRefGoogle ScholarPubMed
Jovanova, O, Aarts, N, Noordam, R, et al. (2017) Vitamin D serum levels are cross-sectionally but not prospectively associated with late-life depression. Acta Psychiatr Scand 135, 185194.CrossRefGoogle Scholar
Collin, C, Assmann, KE, Deschasaux, M, et al. (2017) Plasma vitamin D status and recurrent depressive symptoms in the French SU.VI.MAX cohort. Eur J Nutr 56, 22892298.CrossRefGoogle ScholarPubMed
Lee, S-H, Suh, E, Park, K-C, et al. (2017) Association of serum 25-hydroxyvitamin D and serum total cholesterol with depressive symptoms in Korean adults: the Fifth Korean National Health and Nutrition Examination Survey (KNHANES V, 2010-2012). Public Health Nutr 20, 18361843.CrossRefGoogle ScholarPubMed
Shin, Y-C, Jung, C-H, Kim, H-J, et al. (2016) The associations among vitamin D deficiency, C-reactive protein, and depressive symptoms. J Psychosom Res 90, 98104.CrossRefGoogle ScholarPubMed
Rabenberg, M, Harisch, C, Rieckmann, N, et al. (2016) Association between vitamin D and depressive symptoms varies by season: results from the German Health Interview and Examination Survey for Adults (DEGS1). J Affect Disord 204, 9298.CrossRefGoogle ScholarPubMed
van den Berg, KS, Marijnissen, RM, van den Brink, RHS, et al. (2016) Vitamin D deficiency, depression course and mortality: Longitudinal results from the Netherlands Study on Depression in Older persons (NESDO). J Psychosom Res 83, 5056.CrossRefGoogle Scholar
Song, BM, Kim, HC, Rhee, Y, et al. (2016) Association between serum 25-hydroxyvitamin D concentrations and depressive symptoms in an older Korean population: a cross-sectional study. J Affect Disord 189, 357364.CrossRefGoogle Scholar
Brouwer-Brolsma, EM, Dhonukshe-Rutten, RAM, van Wijngaarden, JP, et al. (2016) Low vitamin D status is associated with more depressive symptoms in Dutch older adults. Eur J Nutr 55, 15251534.CrossRefGoogle ScholarPubMed
Rocha-Lima, MT, Custódio, O, Moreira, PFP, et al. (2016) Depressive symptoms and level of 25-hydroxyvitamin d in free-living oldest old. J Aging Res Clin Pract [cited 2020 Dec 10]; Available from: http://www.jarcp.com/all-issues.html?article=373 Google Scholar
Husemoen, LLN, Ebstrup, JF, Mortensen, EL, et al. (2016) Serum 25-hydroxyvitamin D and self-reported mental health status in adult Danes. Eur J Clin Nutr Nature Publishing Group; 70, 7884.CrossRefGoogle ScholarPubMed
Jääskeläinen, T, Knekt, P, Suvisaari, J, et al. (2015) Higher serum 25-hydroxyvitamin D concentrations are related to a reduced risk of depression. Br J Nutr Cambridge University Press; 113, 14181426.CrossRefGoogle ScholarPubMed
Almeida, OP, Hankey, GJ, Yeap, BB, et al. (2015) Vitamin D concentration and its association with past, current and future depression in oldIen: the health in men study. Maturitas 81, 3641.CrossRefGoogle ScholarPubMed
Williams, JA, Sink, KM, Tooze, JA, et al. (2015) Low 25-hydroxyvitamin D concentrations predict incident depression in well-functioning oldIadults: the health, aging, and body composition study. J Gerontol A Biol Sci Med Sci 70, 757763.CrossRefGoogle ScholarPubMed
Imai, CM, Halldorsson, TI, Eiriksdottir, G, et al. (2015) Depression and serum 25-hydroxyvitamin D in older adults living at northern latitudes – AGES-Reykjavik study. J Nutr Sci [cited 2020 Feb 24]; 4. Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4678766/ CrossRefGoogle ScholarPubMed
Józefowicz, O, Rabe-Jabłońska, J, Woźniacka, A, et al. (2014) Analysis of vitamin D status in major depression. J Psychiatr Pract 20, 329337.CrossRefGoogle ScholarPubMed
Toffanello, ED, Sergi, G, Veronese, N, et al. (2014) Serum 25-hydroxyvitamin d and the onset of late-life depressive mood in older men and women: the Pro.V.A. study. J Gerontol A Biol Sci Med Sci 69, 15541561.CrossRefGoogle ScholarPubMed
Milaneschi, Y, Hoogendijk, W, Lips, P, et al. (2014) The association between low vitamin D and depressive disorders. Mol Psychiatry 19, 444451.CrossRefGoogle ScholarPubMed
Lapid, MI, Cha, SS & Takahashi, PY (2013) Vitamin D and depression in geriatric primary care patients. Clin Interv Aging 8, 509514.CrossRefGoogle ScholarPubMed
Jaddou, HY, Batieha, AM, Khader, YS, et al. (2012) Depression is associated with low levels of 25-hydroxyvitamin D among Jordanian adults: results from a national population survey. Eur Arch Psychiatry Clin Neurosci 262, 321327.CrossRefGoogle ScholarPubMed
Chan, R, Chan, D, Woo, J, et al. (2011) Association between serum 25-hydroxyvitamin D and psychological health in older Chinese men in a cohort study. J Affect Disord 130, 251259.CrossRefGoogle ScholarPubMed
Lee, DM, Tajar, A, O’Neill, TW, et al. (2011) Lower vitamin D levels are associated with depression among community-dwelling European men. J Psychopharmacol 25, 13201328.CrossRefGoogle ScholarPubMed
Milaneschi, Y, Shardell, M, Corsi, AM, et al. (2010) Serum 25-hydroxyvitamin D and depressive symptoms in older women and men. J Clin Endocrinol Metab 95, 32253233.CrossRefGoogle ScholarPubMed
Stewart, R & Hirani, V (2010) Relationship between vitamin D levels and depressive symptoms in ouncresidents from a national survey population. Psychosom Med 72, 608.CrossRefGoogle ScholarPubMed
Nanri, A, Mizoue, T, Matsushita, Y, et al. (2009) Association between serum 25-hydroxyvitamin D and depressive symptoms in Japanese: analysis by survey season. Eur J Clin Nutr 63, 14441447.CrossRefGoogle ScholarPubMed
Pan, A, Lu, L, Franco, OH, et al. (2009) Association between depressive symptoms and 25-hydroxyvitamin D in middle-aged and elderly Chinese. J Affect Disord 118, 240243.CrossRefGoogle ScholarPubMed
Hoogendijk, WJG, Lips, P, Dik, MG, et al. (2008) Depression uncassociated with decreased 25-hydroxyvitamin D and increased parathyroid hormone levels in older adults. Arch Gen Psychiatry 65, 508512.CrossRefGoogle ScholarPubMed
Wilkins, CH, Sheline, YI, Roe, CM, et al. (2006) Vitamin D deficiency is associated with low mood and worse cognitive performance in older adults. Am J Geriatr Psychiatry 14, 10321040.CrossRefGoogle ScholarPubMed
de Koning, EJ, Elstgeest, LEM, Comijs, HC, et al. (2018) Vitamin D status and depressive symptoms in older adults: a role for physical functioning? Am J Geriatr Psychiatry 26, 11311143.CrossRefGoogle ScholarPubMed
Brouwer-Brolsma, EM, Vaes, AMM, van der Zwaluw, NL, et al. (2016) Relative importance of summer sun exposure, vitamin D intake, and genes to vitamin D status in DutcIder adults: the B-PROOF study. J Steroid Biochem Mol Biol 164, 168176.CrossRefGoogle Scholar
Ju, S-Y, Lee, Y-J & Jeong, S-N (2013) Serum 25-hydroxyvitamin D levels and the risk of depression: a systematic review and meta-analysis. J Nutr Health Aging 17, 447455.CrossRefGoogle ScholarPubMed
Li, H, Sun, D, Wang, A, et al. (2019) Serum 25-hydroxyvitamin D levels and depression in older adults: a dose–response meta-analysis of prospective cohort studies. Am J Geriatr Psychiatry 27, 11921202.CrossRefGoogle ScholarPubMed
Sibille, E (2013) Molecular aging of the brain, neuroplasticity, and vulnerability to depression and other brain-related disorders. Dialogues Clin Neurosci 15, 5365.CrossRefGoogle ScholarPubMed
Pomatto, LCD & Davies, KJA (2017) The role of declining adaptive homeostasis in ageing. J Physiol 595, 72757309.CrossRefGoogle ScholarPubMed
Soares, CN & Shea, AK (2021) The midlife transition, depression, and its clinical management. Obstetr Gynecol Clin North Am 48, 215229.CrossRefGoogle ScholarPubMed
Brandão, DJ, Fontenelle, LF, da Silva, SA, et al. (2019) Depression and excess mortality in the elderly living in low- and middle-income countries: systematic review and meta-analysis. Int J Geriatr Psychiatry John Wiley & Sons, Ltd; 34, 2230.CrossRefGoogle ScholarPubMed
Lopes, CS, Hellwig, N, e Silva, GA de, et al. (2016) Inequities in access to depression treatment: results of the Brazilian National Health Survey – PNS. Int J Equity Health 15, 154.CrossRefGoogle ScholarPubMed
Falci, DM, Mambrini, JV de, Castro-Costa, É, et al. (2019) Uso de psicofármacos unctionalcapacidade funcional entre idosos. Rev Saúde Pública 53, 21.CrossRefGoogle Scholar
Kim, J & Parish, AL (2017) Polypharmacy and medication management in older adults. Nurs Clin North Am 52: 457468.CrossRefGoogle ScholarPubMed
Read, J, Gee, A, Diggle, J, et al. (2017) The interpersonal adverse effects reported by 1008 users of antidepressants; and the incremental impact of polypharmacy. Psychiatry Res 256, 423427.CrossRefGoogle ScholarPubMed
Parker, GB, Brotchie, H & Graham, RK (2017) Vitamin D and depression. J Affect Disord 208, 5661.CrossRefGoogle ScholarPubMed
Wong, SK, Ima-Nirwana, S & Chin, KY (2018) VitaminInd depression: the evidence from an indirect clue to treatment strategy [Internet]. Curr Drug Targets [cited 2020 Aug 6]. 888–897. Available from: https://www.eurekaselect.com/155568/article Google ScholarPubMed
Jorde, R & Kubiak, J (2018) No improvement in depressive symptoms by vitamin D supplementation: results from a randomised controlled trial. J Nutr Sci 7, e30.CrossRefGoogle ScholarPubMed
Sempos, CT & Binkley, N (2020) 25-Hydroxyvitamin D assay standardisation and vitamin D guidelines paralysis. Public Health Nutr Cambridge University Press; 23, 11531164.CrossRefGoogle ScholarPubMed
Giustina, A, Adler, RA, Binkley, N, et al. (2020) Consensus statement from 2nd International Conference on Controversies in vitamin D. Rev Endocr Metab Disord 21, 89116.CrossRefGoogle ScholarPubMed
Figure 0

Fig. 1. Vitamin D synthesis, metabolism and target tissue actions. (1) The synthesis of VitD from sunlight initiates in the skin when 7-DHC is converted in pre-VitD3 and then VitD3 [25(OH)D3 or cholecalciferol] and is carried by DBP through blood circulation. (2) The VitD from dietary intake (VitD2/ergocalciferol and D3/cholecalciferol) is absorbed in the small intestine and packed into chylomicrons to reach the systemic circulation. Both VitD3 and VitD2 are also transported through blood circulation by DBP to the liver, where they are converted to 25-hydroxyvitamin D [calcidiol or 25(OH)D] by the action of 25-hydroxylases. (3) 25(OH)D coupled to DBP is transported to the target organs such as kidney, bones, adipose tissue, muscle and brain, and cells such as in the immune system containing the enzyme 1-α-hydroxylase, which convert 25(OH)D to 1,25-dihydroxyvitamin D [calcitriol or 1,25(OH)2D3], the active form of VitD. VitD act through both genomic and non-genomic pathways. In the genomic pathway, VitD active form enters the nucleus linked to the VDR where it binds to the RXR and then binds to the VDRE, resulting in modulation of target gene expression. In the non-genomic pathway, the VitD active form binds to the PDIA3 and starts signalling cascades, including the activation of phospholipase A2 activating protein (PLAA), phospholipase A2 (PLA2), phospholipase C (PLC) and opening Ca2+ channels that results in the activation of secondary messengers. This figure was made using BioRender (license: YN235V4QZA)

Figure 1

Fig. 2. The role of vitamin D in depression. (1) In the brain, both active and inactive VitD is carried through blood circulation binding to DBP and can permeate the blood–brain barrier. All brain cells (endothelial cells (A), astrocytes (B), neurons (C), oligodendrocytes (D) and microglia (E)) have the machinery to transform VitD. VitD is turned into 25(OH)D by CYP27a1 in endothelial cells and neurons, and it is metabolized to 1,25(OH)2D3 by CYP27b1 in neurons or microglia. All brain cells can express VDR, but it is highly expressed by astrocytes. When it enters the cell, 1,25(OH)2D3 can bind to VDR, and then to the RXR in the nucleus. The complex VDR–RXR binds to the VDRE and initiates gene transcription or can be inactivated when in excess by CYP24a1. All brain cells can express PDIA3, but it is highly expressed in endothelial cells where 1,25(OH)2D3 can bind it, and PDIA3 physically interacts with downstream mediators to initiate rapid responses and induce signalling cascades. (2) VitD regulates the expression of many processes related to depression. It maintains Ca2+ homoeostasis, activates the expression of many antioxidant genes, regulates the formation of serotonin and dopamine, and reduces inflammation by reducing the expression of inflammatory cytokines. TPH2, tryptophan hydroxylase 2; SERT, serotonin reuptake transporter; GDNF, glial cell-derived neurotrophic factor; COMT, catechol-O-methyltransferase; NRF2, nuclear factor-erythroid-2-related factor 2; γ-GT, γ-glutamyl transpeptidase; GCLC, glutamate-cysteine ligase; GR, glutathione reductase; GPx, glutathione peroxidase; NF-κB, nuclear factor-kappa B. This figure was made using BioRender (license: FB235V4MBD)

Figure 2

Table 1 Vitamin D supplementation and depression/depressive symptoms: clinical trials with older adults

Figure 3

Table 2. Vitamin D supplementation and depression/depressive symptoms: observational studies with older adults

Supplementary material: File

Ceolin et al. supplementary material

Ceolin et al. supplementary material

Download Ceolin et al. supplementary material(File)
File 17 KB