Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 2019 2022-07-11 23:18:58 |
2 format -9 word(s) 2010 2022-07-12 04:38:15 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Kouba, B.R.;  Camargo, A.;  Gil-Mohapel, J.;  Rodrigues, A.L.S. Vitamin D for Treatment of Depression and Anxiety. Encyclopedia. Available online: https://encyclopedia.pub/entry/25024 (accessed on 02 July 2024).
Kouba BR,  Camargo A,  Gil-Mohapel J,  Rodrigues ALS. Vitamin D for Treatment of Depression and Anxiety. Encyclopedia. Available at: https://encyclopedia.pub/entry/25024. Accessed July 02, 2024.
Kouba, Bruna R., Anderson Camargo, Joana Gil-Mohapel, Ana Lúcia S. Rodrigues. "Vitamin D for Treatment of Depression and Anxiety" Encyclopedia, https://encyclopedia.pub/entry/25024 (accessed July 02, 2024).
Kouba, B.R.,  Camargo, A.,  Gil-Mohapel, J., & Rodrigues, A.L.S. (2022, July 11). Vitamin D for Treatment of Depression and Anxiety. In Encyclopedia. https://encyclopedia.pub/entry/25024
Kouba, Bruna R., et al. "Vitamin D for Treatment of Depression and Anxiety." Encyclopedia. Web. 11 July, 2022.
Vitamin D for Treatment of Depression and Anxiety
Edit

Major depressive disorder and anxiety disorders are common and disabling conditions that affect millions of people worldwide. Despite being different disorders, symptoms of depression and anxiety frequently overlap in individuals, making them difficult to diagnose and treat adequately. Therefore, compounds capable of exerting beneficial effects against both disorders are of special interest. Noteworthily, vitamin D deficiency has been associated with an increased risk of developing depression and anxiety, and individuals with these psychiatric conditions have low serum levels of this vitamin. 

anxiety depression neuromodulator vitamin D

1. Introduction

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

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

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

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

In recent years, several preclinical studies have been conducted to investigate the possible antidepressant and anxiolytic effects of vitamin D.
Of note, chronic administration of cholecalciferol [5 mg/kg for 14 days; subcutaneous (s.c.) administration] elicited an antidepressant-like effect in the forced swim test in ovariectomized Wistar rats [35]. In addition, cholecalciferol supplementation (5 mg/kg for 14 days; s.c.) was capable of attenuating anxiety-like behaviors in the elevated plus-maze and the light–dark box tests in ovariectomized Wistar rats [10][36]. Based on these findings, further studies have been conducted to better understand the mechanisms underlying the anxiolytic and antidepressant effects of vitamin D in animal models [37][38].
Camargo et al. (2018) reported that cholecalciferol [2.5 µg/kg, orally by mouth (p.o.)], administered once a day in the last 7 days of chronic corticosterone administration (20 mg/kg, p.o., for 21 days), exerted an antidepressant-like effect in male mice subjected to the splash test and tail suspension test. Additionally, cholecalciferol treatment attenuated the increase in protein carbonyl and nitrite levels induced by corticosterone in the brain, suggesting that vitamin D3 has an antidepressant-like effect by, in part, modulating oxidative stress [37]. The administration of cholecalciferol (100 IU/kg, p.o.) for 7 days also abolished the depressive-like behavior in the tail suspension test induced by chronic corticosterone administration in female mice [39]. A significant decrease in ROS production in the hippocampus was observed after treatment with cholecalciferol, both in control and corticosterone-exposed mice, reinforcing the notion that the antidepressant-like effect of this vitamin may involve the modulation of oxidative stress [39]. More recently, a study by Neis et al. (2022) showed that repeated administration of cholecalciferol for 7 days (2.5 μg/kg, p.o.) abolished chronic unpredictable stress-induced depressive-like behavior in the tail suspension test, as well as a reduction in serotonin levels in the prefrontal cortex of female mice. Moreover, reinforcing the involvement of the serotonergic system in the antidepressant-like effect of cholecalciferol, the administration of the serotonin synthesis inhibitor p-chlorophenylalanine methyl ester was effective in abolishing the reduction in immobility time in the tail suspension test elicited by cholecalciferol [40].
The repeated administration of a low dose of cholecalciferol (2.5 μg/kg, p.o.) also caused an antidepressant-like effect and was effective in reducing the immunocontent of proteins that form the NLRP3 inflammasome, such as ASC [apoptosis-associated speck-like protein containing a caspase recruitment domain (CARD)], caspase-1, and thioredoxin-interacting protein (TXNIP) in the hippocampus of male mice [41]. Calcitriol treatment (100 ng/kg, p.o.) for 10 weeks in ovariectomized female Sprague–Dawley rats was also effective in producing neuroprotective effects by regulating the adenosine monophosphate (AMP)-activated protein kinase (AMPK)/NF-kB signaling pathway. Moreover, calcitriol treatment reduced the pro-inflammatory cytokines IL-1β, IL-6, and TNF-α, as well as iNOS and cyclooxygenase-2 (COX-2) levels in the hippocampus [42]. This suggests that the modulation of the NLRP3 inflammasome-driven pathway may underlie, at least in part, the antidepressant-like effect of this vitamin.
More recently, Bakhtiari-Dovvombaygi et al. (2021) also reported that the anti-inflammatory and antioxidant effects displayed by pretreatment with vitamin D3 (10,000 IU/kg for 28 days) in male rats underlie the ability of this vitamin to abrogate anxiety- and depressive-like behaviors induced by chronic unpredictable mild stress (CUMS) in the elevated plus-maze and forced swimming test. Indeed, these protective effects of vitamin D were accompanied by a decrease in cortical malondialdehyde and IL-6 levels, as well as an increase in total thiol levels and enhanced SOD and catalase activity [38]. Interestingly, another study observed that following 4 weeks of CUMS, the occurrence of depressive-like behaviors was associated with an increase in 1,25(OH)2D and VDR expression in the hippocampus of rats, suggesting a compensatory mechanism, by which vitamin D may protect against the development of depressive-like behaviors [43].
In addition to attenuating depressive- and anxiety-like behaviors through its anti-inflammatory and antioxidant properties, the modulation of neurotrophic factors has also been shown to contribute to the protective properties of vitamin D [44]. Xu et al. used male C57BL/6 mice to show that calcitriol (25 μg/kg/day for 4 weeks; i.c.v.) is effective in acting as an antidepressant in a post-stroke depression model by up-regulating VDR and BDNF expression [45]. In an ovariectomized Wistar rat model of depression induced by CUMS, vitamin D3 (5 mg/kg for 4 weeks; s.c.) treatment was able to reverse depression-like behaviors in the sucrose preference test and the forced swimming test by increasing BDNF and NT-3/NT-4 levels in the hippocampus [44]. Although these studies showed that vitamin D supplementation results in an increase in pro-neurogenic neurotrophins, such as BDNF, Groves et al. observed that vitamin D deficiency in BALB/c mice was associated with depressive-like behaviors without compromising hippocampal neurogenesis [46]. Therefore, further studies are needed to elucidate whether neurogenesis is critical for the anxiolytic and antidepressant effects of this vitamin.

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

Several studies have reported that vitamin D supplementation improves symptoms of depression and anxiety associated with various medical conditions, including type II diabetes, Crohn’s disease, ulcerative colitis, and obesity [47][48][49][50][51].
However, the potential therapeutic effects of vitamin D in individuals primarily diagnosed with depression or anxiety remain controversial. For example, vitamin D supplementation (1600 IU for 6 months) was shown to significantly improve anxiety symptoms, but not depressive symptoms, in patients with vitamin D deficiency [52]. Likewise, supplementation with 2800 IU of vitamin D in patients with depression did not promote a significant reduction in Hamilton D-17 scores [53]. On the other hand, supplementation with 50,000 IU of vitamin D for 2 weeks was able to improve depression severity, as assessed with the Beck Depression Inventory-II (BDI-II), although no changes in serotonin levels were detected [54]. However, in another study, cholecalciferol treatment (50,000 IU for 3 months) significantly increased serum serotonin levels, while decreasing BDI scores in women with moderate, severe, and extreme depression. Interestingly, among men, an improvement in the severity of depressive symptoms with vitamin D supplementation was only observed in those diagnosed with severe depression [55]. Beneficial effects of vitamin D (50,000 IU for 8 weeks) supplementation have also been observed in older adults (over 60 years of age) with depression [56]. However, lower doses of vitamin D (400 IU daily for 2 years) were not able to improve depressive symptoms [57]. Finally, a single dose of vitamin D (300,000 IU) was reported as an effective and safe intervention in MDD with concurrent vitamin D deficiency [58][59]. In patients with depression, the daily administration of 1500 IU vitamin D3 plus 20 mg fluoxetine for 8 weeks was superior to fluoxetine alone [60]. Another study reported that vitamin D supplementation (50,000 IU once/week for 3 months) in combination with standard of care improved the severity of anxiety in individuals diagnosed with Generalized Anxiety Disorder by increasing serotonin concentrations and decreasing the levels of the inflammatory biomarker neopterin [61]. Overall, although there is compelling clinical evidence pointing to the benefits of vitamin D for the management of depression and anxiety, it is important to note that divergent results have also been obtained [62]. Multiple factors may contribute to these discrepant results, including differences in the doses of vitamin D used, treatment time, serum 25-hydroxyvitamin D levels at baseline, nutritional condition at the onset of the treatment, age and sex of the individuals, as well as the presence of comorbidities that may influence the efficacy of vitamin D supplementation. Additionally, the heterogeneity observed in clinical studies may also be associated with genetic polymorphisms that may affect vitamin D efficacy [63][64][65].

References

  1. Craske, M.G.; Stein, M.B.; Eley, T.C.; Milad, M.R.; Holmes, A.; Rapee, R.M.; Wittchen, H.U. Anxiety Disorders. Nat. Rev. Dis. Primers 2017, 3, 17024.
  2. Otte, C.; Gold, S.M.; Penninx, B.W.; Pariante, C.M.; Etkin, A.; Fava, M.; Mohr, D.C.; Schatzberg, A.F. Major Depressive Disorder. Nat. Rev. Dis. Primers 2016, 2, 16065.
  3. Lamers, F.; Van Oppen, P.; Comijs, H.C.; Smit, J.H.; Spinhoven, P.; van Balkom, A.J.L.M.; Nolen, W.A.; Zitman, F.G.; Beekman, A.T.F.; Penninx, B.W.J.H. Comorbidity Patterns of Anxiety and Depressive Disorders in a Large Cohort Study: The Netherlands Study of Depression and Anxiety (NESDA). J. Clin. Psychiatry 2011, 72, 341–348.
  4. Malhi, G.S.; Mann, J.J. Depression. Lancet 2018, 392, 2299–2312.
  5. Schiele, M.A.; Domschke, K. Epigenetics at the Crossroads between Genes, Environment and Resilience in Anxiety Disorders: Epigenetics in Anxiety Disorders. Genes Brain Behav. 2018, 17, e12423.
  6. De Haan, P.; Klein, H.C.; ’t Hart, B.A. Autoimmune Aspects of Neurodegenerative and Psychiatric Diseases: A Template for Innovative Therapy. Front. Psychiatry 2017, 8, 46.
  7. Carlessi, A.S.; Borba, L.A.; Zugno, A.I.; Quevedo, J.; Réus, G.Z. Gut Microbiota-Brain Axis in Depression: The Role of Neuroinflammation. Eur. J. Neurosci. 2021, 53, 222–235.
  8. Kim, Y.K.; Jeon, S.W. Neuroinflammation and the Immune-Kynurenine Pathway in Anxiety Disorders. Curr. Neuropharmacol. 2018, 16, 574–582.
  9. Stevens, B.R.; Goel, R.; Seungbum, K.; Richards, E.M.; Holbert, R.C.; Pepine, C.J.; Raizada, M.K. Increased Human Intestinal Barrier Permeability Plasma Biomarkers Zonulin and FABP2 Correlated with Plasma LPS and Altered Gut Microbiome in Anxiety or Depression. Gut 2018, 67, 1555–1557.
  10. Fedotova, J.; Zarembo, D.; Dragasek, J.; Caprnda, M.; Kruzliak, P.; Dudnichenko, T. Modulating Effects of Cholecalciferol Treatment on Estrogen Deficiency-Induced Anxiety-Like Behavior of Adult Female Rats. Folia Med. 2017, 59, 139–158.
  11. Khairy, E.Y.; Attia, M.M. Protective Effects of Vitamin D on Neurophysiologic Alterations in Brain Aging: Role of Brain-Derived Neurotrophic Factor (BDNF). Nutr. Neurosci. 2021, 24, 650–659.
  12. Lefebvre d’Hellencourt, C.; Montero-Menei, C.N.; Bernard, R.; Couez, D. Vitamin D3 Inhibits Proinflammatory Cytokines and Nitric Oxide Production by the EOC13 Microglial Cell Line. J. Neurosci. Res. 2003, 71, 575–582.
  13. Morello, M.; Landel, V.; Lacassagne, E.; Baranger, K.; Annweiler, C.; Féron, F.; Millet, P. Vitamin D Improves Neurogenesis and Cognition in a Mouse Model of Alzheimer’s Disease. Mol. Neurobiol. 2018, 55, 6463–6479.
  14. Yong, H.Y.F.; Rawji, K.S.; Ghorbani, S.; Xue, M.; Yong, V.W. The Benefits of Neuroinflammation for the Repair of the Injured Central Nervous System. Cell. Mol. Immunol. 2019, 16, 540–546.
  15. Fullerton, J.N.; Gilroy, D.W. Resolution of Inflammation: A New Therapeutic Frontier. Nat. Rev. Drug Discov. 2016, 15, 551–567.
  16. Barton, G.M. A Calculated Response: Control of Inflammation by the Innate Immune System. J. Clin. Investig. 2008, 118, 413–420.
  17. Herman, F.J.; Pasinetti, G.M. Principles of Inflammasome Priming and Inhibition: Implications for Psychiatric Disorders. Brain. Behav. Immun. 2018, 73, 66–84.
  18. Orihuela, R.; McPherson, C.A.; Harry, G.J. Microglial M1/M2 Polarization and Metabolic States: Microglia Bioenergetics with Acute Polarization. Br. J. Pharmacol. 2016, 173, 649–665.
  19. Rudzki, L.; Maes, M. The Microbiota-Gut-Immune-Glia (MGIG) Axis in Major Depression. Mol. Neurobiol. 2020, 57, 4269–4295.
  20. Kelley, N.; Jeltema, D.; Duan, Y.; He, Y. The NLRP3 Inflammasome: An Overview of Mechanisms of Activation and Regulation. Int. J. Mol. Sci. 2019, 20, 3328.
  21. Song, N.; Li, T. Regulation of NLRP3 Inflammasome by Phosphorylation. Front. Immunol. 2018, 9, 2305.
  22. Liu, X.; Zhang, X.; Ding, Y.; Zhou, W.; Tao, L.; Lu, P.; Wang, Y.; Hu, R. Nuclear Factor E2-Related Factor-2 Negatively Regulates NLRP3 Inflammasome Activity by Inhibiting Reactive Oxygen Species-Induced NLRP3 Priming. Antioxid. Redox Signal. 2017, 26, 28–43.
  23. Xu, X.; Zhang, L.; Ye, X.; Hao, Q.; Zhang, T.; Cui, G.; Yu, M. Nrf2/ARE Pathway Inhibits ROS-Induced NLRP3 Inflammasome Activation in BV2 Cells after Cerebral Ischemia Reperfusion. Inflamm. Res. 2018, 67, 57–65.
  24. Sutterwala, F.S.; Haasken, S.; Cassel, S.L. Mechanism of NLRP3 Inflammasome Activation. Ann. N. Y. Acad. Sci. 2014, 1319, 82–95.
  25. Liddelow, S.A.; Guttenplan, K.A.; Clarke, L.E.; Bennett, F.C.; Bohlen, C.J.; Schirmer, L.; Bennett, M.L.; Münch, A.E.; Chung, W.S.; Peterson, T.C.; et al. Neurotoxic Reactive Astrocytes Are Induced by Activated Microglia. Nature 2017, 541, 481–487.
  26. Tejeda, G.; Díaz-Guerra, M. Integral Characterization of Defective BDNF/TrkB Signalling in Neurological and Psychiatric Disorders Leads the Way to New Therapies. Int. J. Mol. Sci. 2017, 18, 268.
  27. Farina, C.; Aloisi, F.; Meinl, E. Astrocytes Are Active Players in Cerebral Innate Immunity. Trends Immunol. 2007, 28, 138–145.
  28. Troubat, R.; Barone, P.; Leman, S.; Desmidt, T.; Cressant, A.; Atanasova, B.; Brizard, B.; El Hage, W.; Surget, A.; Belzung, C.; et al. Neuroinflammation and Depression: A Review. Eur. J. Neurosci. 2021, 53, 151–171.
  29. Bansal, Y.; Singh, R.; Parhar, I.; Kuhad, A.; Soga, T. Quinolinic Acid and Nuclear Factor Erythroid 2-Related Factor 2 in Depression: Role in Neuroprogression. Front. Pharmacol. 2019, 10, 452.
  30. Braidy, N.; Grant, R.; Brew, B.J.; Adams, S.; Jayasena, T.; Guillemin, G.J. Effects of Kynurenine Pathway Metabolites on Intracellular NAD + Synthesis and Cell Death in Human Primary Astrocytes and Neurons. Int. J. Tryptophan Res. 2009, 2, 61–69.
  31. Pierozan, P.; Biasibetti, H.; Schmitz, F.; Ávila, H.; Parisi, M.M.; Barbe-Tuana, F.; Wyse, A.T.S.; Pessoa-Pureur, R. Quinolinic Acid Neurotoxicity: Differential Roles of Astrocytes and Microglia via FGF-2-Mediated Signaling in Redox-Linked Cytoskeletal Changes. Biochim. Biophys. Acta BBA-Mol. Cell Res. 2016, 1863, 3001–3014.
  32. Agus, A.; Planchais, J.; Sokol, H. Gut Microbiota Regulation of Tryptophan Metabolism in Health and Disease. Cell Host Microbe 2018, 23, 716–724.
  33. Chen, L.M.; Bao, C.H.; Wu, Y.; Liang, S.H.; Wang, D.; Wu, L.Y.; Huang, Y.; Liu, H.R.; Wu, H.G. Tryptophan-Kynurenine Metabolism: A Link between the Gut and Brain for Depression in Inflammatory Bowel Disease. J. Neuroinflammation 2021, 18, 135.
  34. Siopi, E.; Chevalier, G.; Katsimpardi, L.; Saha, S.; Bigot, M.; Moigneu, C.; Eberl, G.; Lledo, P.M. Changes in Gut Microbiota by Chronic Stress Impair the Efficacy of Fluoxetine. Cell Rep. 2020, 30, 3682–3690.e6.
  35. Fedotova, J.; Dudnichenko, T.; Kruzliak, P.; Puchavskaya, Z. Different Effects of Vitamin D Hormone Treatment on Depression-like Behavior in the Adult Ovariectomized Female Rats. Biomed. Pharmacother. 2016, 84, 1865–1872.
  36. Fedotova, J.O. Vitamin D3 Treatment Differentially Affects Anxiety-like Behavior in the Old Ovariectomized Female Rats and Old Ovariectomized Female Rats Treated with Low Dose of 17β-Estradiol. BMC Med. Genet. 2019, 20, 49.
  37. Camargo, A.; Dalmagro, A.P.; Rikel, L.; da Silva, E.B.; Simão da Silva, K.A.B.; Zeni, A.L.B. Cholecalciferol Counteracts Depressive-like Behavior and Oxidative Stress Induced by Repeated Corticosterone Treatment in Mice. Eur. J. Pharmacol. 2018, 833, 451–461.
  38. Bakhtiari-Dovvombaygi, H.; Izadi, S.; Zare Moghaddam, M.; Hashemzehi, M.; Hosseini, M.; Azhdari-Zarmehri, H.; Dinpanah, H.; Beheshti, F. Beneficial Effects of Vitamin D on Anxiety and Depression-like Behaviors Induced by Unpredictable Chronic Mild Stress by Suppression of Brain Oxidative Stress and Neuroinflammation in Rats. Naunyn. Schmiedebergs Arch. Pharmacol. 2021, 394, 655–667.
  39. Da Silva Souza, S.V.; da Rosa, P.B.; Neis, V.B.; Moreira, J.D.; Rodrigues, A.L.S.; Moretti, M. Effects of Cholecalciferol on Behavior and Production of Reactive Oxygen Species in Female Mice Subjected to Corticosterone-Induced Model of Depression. Naunyn. Schmiedebergs Arch. Pharmacol. 2020, 393, 111–120.
  40. Neis, V.B.; Werle, I.; Moretti, M.; Rosa, P.B.; Camargo, A.; Dalsenter, Y.D.O.; Platt, N.; Rosado, A.F.; Engel, W.D.; de Almeida, G.R.L.; et al. Involvement of Serotonergic Neurotransmission in the Antidepressant-like Effect Elicited by Cholecalciferol in the Chronic Unpredictable Stress Model in Mice. Metab. Brain Dis. 2022, 37, 1597–1608.
  41. Camargo, A.; Dalmagro, A.P.; Platt, N.; Rosado, A.F.; Neis, V.B.; Zeni, A.L.B.; Kaster, M.P.; Rodrigues, A.L.S. Cholecalciferol Abolishes Depressive-like Behavior and Hippocampal Glucocorticoid Receptor Impairment Induced by Chronic Corticosterone Administration in Mice. Pharmacol. Biochem. Behav. 2020, 196, 172971.
  42. Zhang, W.; Guo, Y.; Wang, K.; Chen, L.; Jiang, P. Neuroprotective Effects of Vitamin D and 17ß-Estradiol against Ovariectomy-Induced Neuroinflammation and Depressive-like State: Role of the AMPK/NF-ΚB Pathway. Int. Immunopharmacol. 2020, 86, 106734.
  43. Jiang, P.; Zhang, W.Y.; Li, H.D.; Cai, H.L.; Liu, Y.P.; Chen, L.Y. Stress and Vitamin D: Altered Vitamin D Metabolism in Both the Hippocampus and Myocardium of Chronic Unpredictable Mild Stress Exposed Rats. Psychoneuroendocrinology 2013, 38, 2091–2098.
  44. Koshkina, A.; Dudnichenko, T.; Baranenko, D.; Fedotova, J.; Drago, F. Effects of Vitamin D3 in Long-Term Ovariectomized Rats Subjected to Chronic Unpredictable Mild Stress: BDNF, NT-3, and NT-4 Implications. Nutrients 2019, 11, 1726.
  45. Xu, Y.; Liang, L. Vitamin D3/Vitamin D Receptor Signaling Mitigates Symptoms of Post-Stroke Depression in Mice by Upregulating Hippocampal BDNF Expression. Neurosci. Res. 2021, 170, 306–313.
  46. Groves, N.J.; Bradford, D.; Sullivan, R.K.P.; Conn, K.A.; Aljelaify, R.F.; McGrath, J.J.; Burne, T.H.J. Behavioural Effects of Adult Vitamin D Deficiency in BALB/c Mice Are Not Associated with Proliferation or Survival of Neurons in the Adult Hippocampus. PLoS ONE 2016, 11, e0152328.
  47. Jorde, R.; Sneve, M.; Figenschau, Y.; Svartberg, J.; Waterloo, K. Effects of Vitamin D Supplementation on Symptoms of Depression in Overweight and Obese Subjects: Randomized Double-Blind Trial. J. Intern. Med. 2008, 264, 599–609.
  48. Narula, N.; Cooray, M.; Anglin, R.; Muqtadir, Z.; Narula, A.; Marshall, J.K. Impact of High-Dose Vitamin D3 Supplementation in Patients with Crohn’s Disease in Remission: A Pilot Randomized Double-Blind Controlled Study. Dig. Dis. Sci. 2017, 62, 448–455.
  49. Penckofer, S.; Byrn, M.; Adams, W.; Emanuele, M.A.; Mumby, P.; Kouba, J.; Wallis, D.E. Vitamin D Supplementation Improves Mood in Women with Type 2 Diabetes. J. Diabetes Res. 2017, 2017, 8232863.
  50. Sharifi, A.; Vahedi, H.; Nedjat, S.; Mohamadkhani, A.; Hosseinzadeh Attar, M.J. Vitamin D Decreases Beck Depression Inventory Score in Patients with Mild to Moderate Ulcerative Colitis: A Double-Blind Randomized Placebo-Controlled Trial. J. Diet. Suppl. 2019, 16, 541–549.
  51. Yosaee, S.; Soltani, S.; Esteghamati, A.; Motevalian, S.A.; Tehrani-Doost, M.; Clark, C.C.T.; Jazayeri, S. Effects of Zinc, Vitamin D, and Their Co-Supplementation on Mood, Serum Cortisol, and Brain-Derived Neurotrophic Factor in Patients with Obesity and Mild to Moderate Depressive Symptoms: A Phase II, 12-Wk, 2 × 2 Factorial Design, Double-Blind, Randomized, Placebo-Controlled Trial. Nutrition 2020, 71, 110601.
  52. Zhu, C.; Zhang, Y.; Wang, T.; Lin, Y.; Yu, J.; Xia, Q.; Zhu, P.; Zhu, D. Vitamin D Supplementation Improves Anxiety but Not Depression Symptoms in Patients with Vitamin D Deficiency. Brain Behav. 2020, 10, e01760.
  53. Hansen, J.P.; Pareek, M.; Hvolby, A.; Schmedes, A.; Toft, T.; Dahl, E.; Nielsen, C.T. Vitamin D3 Supplementation and Treatment Outcomes in Patients with Depression (D3-Vit-Dep). BMC Res. Notes 2019, 12, 203.
  54. Kaviani, M.; Nikooyeh, B.; Zand, H.; Yaghmaei, P.; Neyestani, T.R. Effects of Vitamin D Supplementation on Depression and Some Involved Neurotransmitters. J. Affect. Disord. 2020, 269, 28–35.
  55. Alghamdi, S.; Alsulami, N.; Khoja, S.; Alsufiani, H.; Tayeb, H.O.; Tarazi, F.I. Vitamin D Supplementation Ameliorates Severity of Major Depressive Disorder. J. Mol. Neurosci. 2020, 70, 230–235.
  56. Alavi, N.M.; Khademalhoseini, S.; Vakili, Z.; Assarian, F. Effect of Vitamin D Supplementation on Depression in Elderly Patients: A Randomized Clinical Trial. Clin. Nutr. 2019, 38, 2065–2070.
  57. Bertone-Johnson, E.R.; Powers, S.I.; Spangler, L.; Larson, J.; Michael, Y.L.; Millen, A.E.; Bueche, M.N.; Salmoirago-Blotcher, E.; Wassertheil-Smoller, S.; Brunner, R.L.; et al. Vitamin D Supplementation and Depression in the Women’s Health Initiative Calcium and Vitamin D Trial. Am. J. Epidemiol. 2012, 176, 1–13.
  58. Mozaffari-Khosravi, H.; Nabizade, L.; Yassini-Ardakani, S.M.; Hadinedoushan, H.; Barzegar, K. The Effect of 2 Different Single Injections of High Dose of Vitamin D on Improving the Depression in Depressed Patients With Vitamin D Deficiency: A Randomized Clinical Trial. J. Clin. Psychopharmacol. 2013, 33, 378–385.
  59. Vellekkatt, F.; Menon, V.; Rajappa, M.; Sahoo, J. Effect of Adjunctive Single Dose Parenteral Vitamin D Supplementation in Major Depressive Disorder with Concurrent Vitamin D Deficiency: A Double-Blind Randomized Placebo-Controlled Trial. J. Psychiatr. Res. 2020, 129, 250–256.
  60. Khoraminya, N.; Tehrani-Doost, M.; Jazayeri, S.; Hosseini, A.; Djazayery, A. Therapeutic Effects of Vitamin D as Adjunctive Therapy to Fluoxetine in Patients with Major Depressive Disorder. Aust. N. Z. J. Psychiatry 2013, 47, 271–275.
  61. Eid, A.; Khoja, S.; AlGhamdi, S.; Alsufiani, H.; Alzeben, F.; Alhejaili, N.; Tayeb, H.O.; Tarazi, F.I. Vitamin D Supplementation Ameliorates Severity of Generalized Anxiety Disorder (GAD). Metab. Brain Dis. 2019, 34, 1781–1786.
  62. Casseb, G.A.S.; Kaster, M.P.; Rodrigues, A.L.S. Potential Role of Vitamin D for the Management of Depression and Anxiety. CNS Drugs 2019, 33, 619–637.
  63. Valdivielso, J.M.; Fernandez, E. Vitamin D receptor polymorphisms and diseases. Clin. Chim. Acta 2006, 371, 1–12.
  64. Lye, M.S.; Tor, Y.S.; Tey, Y.Y.; Shahabudin, A.; Loh, S.P.; Ibrahim, N.; Stanslas, J.; Rosli, R.; Ling, K.H. BsmI-ApaI-TaqI TAC (BAt) Haplotype of Vitamin D Receptor Gene Is Associated with Increased Risk of Major Depressive Disorder. J. Mol. Neurosci. 2020, 71, 981–990.
  65. Usategui-Martín, R.; De Luis-Román, D.A.; Fernández-Gómez, J.M.; Ruiz-Mambrilla, M.; Pérez-Castrillón, J.L. Vitamin D Receptor (VDR) Gene Polymorphisms Modify the Response to Vitamin D Supplementation: A Systematic Review and Meta-Analysis. Nutrients 2022, 14, 360.
More
Information
Subjects: Neurosciences
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , ,
View Times: 678
Revisions: 2 times (View History)
Update Date: 12 Jul 2022
1000/1000
Video Production Service