AccScience Publishing / AN / Volume 1 / Issue 3 / DOI: 10.36922/an.v1i3.272
Cite this article
101
Download
1338
Views
Journal Browser
Volume | Year
Issue
Search
News and Announcements
View All
REVIEW

Gut-brain axis in depression: Crosstalk between neuroinflammation and gut microbiota

Yang Cai1 Shenyang Zhang2 Sibo Zhao1 Honghong Yao1,3,4*
Show Less
1 Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing, China
2 Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
3 Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
4 Institute of Life Sciences, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
Advanced Neurology 2022, 1(3), 272 https://doi.org/10.36922/an.v1i3.272
Submitted: 22 November 2022 | Accepted: 30 December 2022 | Published: 16 January 2023
© 2023 by the Author(s). This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution 4.0 International License ( https://creativecommons.org/licenses/by/4.0/ )
Abstract

Depression is the most common mental disorder and the leading cause of disability and suicide worldwide. Recently, during the COVID-19 pandemic, there has been a dramatic increase in the number of patients with depression on a global scale. Therefore, new insights into the underlying pathophysiology of depression are urgently required to develop more effective therapeutic strategies. An interesting fact is the coexistence of increased intestinal permeability and disrupted blood-brain barrier observed in patients with depression and animal models. A growing number of studies have revealed that a bidirectional interaction exists between the brain and the gastrointestinal tract, commonly termed the gut-brain axis. Emerging evidence has suggested that the regulation of neuroinflammation and gut homeostasis through the gut-brain axis is influenced by stress and depression, in which gut microbiota and microbe-derived short-chain fatty acids play a crucial role. Here, we describe the molecular and cellular mechanisms involved in the crosstalk between neuroinflammation and gut microbiota in depression. We further highlight fecal microbiota transplantation and dietary supplementation in depression and discuss their potential as therapeutic targets for depression.

Keywords
Depression
Stress
Gut-brain axis
Microbiota
Short-chain fatty acids
Neuroinflammation
Nutrients
Funding
Science and Technology Innovation 2030-Major Project of the Ministry of Science and Technology of China
National Natural Science Foundation of Distinguished Young Scholars
National Natural Science Foundation of China
Fundamental Research Funds for the Central Universities
References
[1]

Global Health Data Exchange, 2019, Institute of Health Metrics and Evaluation. Available from: https://www.vizhub. healthdata.org/gbd-results [Last accesed on 2022 Oct 14]. 

[2]

World Health Organization, 2022, Mental Health and COVID-19: Early Evidence of the Pandemic’s Impact. Geneva: World Health Organization. Available from: https://www.who.int/publications/i/item/WHO-2019-nCoV-Sci_Brief- Mental_health-2022.1 [Last accessed on 2022 Oct 15].

[3]

World Health Organization, 2022, Depression: Fact Sheets. Available from: https://www.who.int/news-room/fact-sheets/detail/depression [Last accessed on 2022 Oct 14].

[4]

DeRubeis RJ, Siegle GJ, Hollon SD, 2008, Cognitive therapy versus medication for depression: Treatment outcomes and neural mechanisms. Nat Rev Neurosci, 9: 788–796. https://doi.org/10.1038/nrn2345

[5]

Heneka MT, Carson MJ, El Khoury J, et al., 2015, Neuroinflammation in Alzheimer’s disease. Lancet Neurol, 14: 388–405. https://doi.org/10.1016/S1474-4422(15)70016-5 

[6]

De Virgilio A, Greco A, Fabbrini G, et al., 2016, Parkinson’s disease: Autoimmunity and neuroinflammation. Autoimmun Rev, 15: 1005–1111. https://doi.org/10.1016/j.autrev.2016.07.022

[7]

Troubat R, Barone P, Leman S, et al., 2021, Neuroinflammation and depression: A review. Eur J Neurosci, 53: 151–171. https://doi.org/10.1111/ejn.14720

[8]

Raison CL, Capuron L, Miller AH, 2006, Cytokines sing the blues: inflammation and the pathogenesis of depression. Trends Immunol, 27: 24–31. https://doi.org/10.1016/j.it.2005.11.006 

[9]

Musselman DL, Lawson DH, Gumnick JF, et al., 2001, Paroxetine for the prevention of depression induced by high-dose interferon alfa. N Engl J Med, 344: 961–966. https://doi.org/10.1056/NEJM200103293441303 

[10]

Haapakoski R, Mathieu J, Ebmeier KP, et al., 2015, Cumulative meta-analysis of interleukins 6 and 1beta, tumour necrosis factor alpha and C-reactive protein in patients with major depressive disorder. Brain Behav Immun, 49: 206–215. https://doi.org/10.1016/j.bbi.2015.06.001

[11]

Kappelmann N, Lewis G, Dantzer R, et al., 2018, Antidepressant activity of anti-cytokine treatment: A systematic review and meta-analysis of clinical trials of chronic inflammatory conditions. Mol Psychiatry, 23: 335–343. https://doi.org/10.1038/mp.2016.167

[12]

Simpson CA, Diaz-Arteche C, Eliby D, et al., 2021, The gut microbiota in anxiety and depression- A systematic review. Clin Psychol Rev, 83: 101943. https://doi.org/10.1016/j.cpr.2020.101943

[13]

Foster JA, Neufeld KA, 2013, Gut-brain axis: How the microbiome influences anxiety and depression. Trends Neurosci, 36: 305–312. https://doi.org/10.1016/j.tins.2013.01.005 

[14]

Cryan JF, O’Riordan KJ, Cowan CS, et al., 2019, The microbiota-gut-brain axis. Physiol Rev, 99: 1877–2013. https://doi.org/10.1152/physrev.00018.2018 

[15]

Valles-Colomer M, Falony G, Darzi Y, et al., 2019, The neuroactive potential of the human gut microbiota in quality of life and depression. Nat Microbiol, 4: 623–632. https://doi.org/10.1038/s41564-018-0337-x

[16]

Jiang H, Ling Z, Zhang Y, et al., 2015, Altered fecal microbiota composition in patients with major depressive disorder. Brain Behav Immun, 48: 186–194. https://doi.org/10.1016/j.bbi.2015.03.016

[17]

Yang Z, Li J, Gui X, et al., 2020, Updated review of research on the gut microbiota and their relation to depression in animals and human beings. Mol Psychiatry, 25: 2759–2772. https://doi.org/10.1038/s41380-020-0729-1

[18]

Messaoudi M, Lalonde R, Violle N, et al., 2011, Assessment of psychotropic-like properties of a probiotic formulation (Lactobacillus helveticus R0052 and Bifidobacterium longum R0175) in rats and human subjects. Br J Nutr, 105: 755–764. https://doi.org/10.1017/S0007114510004319

[19]

Camilleri M, 2019, Leaky gut: Mechanisms, measurement and clinical implications in humans. Gut, 68: 1516–1526. https://doi.org/10.1136/gutjnl-2019-318427

[20]

Ohlsson L, Gustafsson A, Lavant E, et al., 2019, Leaky gut biomarkers in depression and suicidal behavior. Acta Psychiatr Scand, 139: 185–193. https://doi.org/10.1111/acps.12978

[21]

Vanuytsel T, van Wanrooy S, Vanheel H, et al., 2014, Psychological stress and corticotropin-releasing hormone increase intestinal permeability in humans by a mast cell-dependent mechanism. Gut, 63: 1293–1299. https://doi.org/10.1136/gutjnl-2013-305690 

[22]

Clark A, Mach, 2016, Exercise-induced stress behavior, gut-microbiota-brain axis and diet: A systematic review for athletes. J Int Soc Sports Nutr, 13: 43. https://doi.org/10.1186/s12970-016-0155-6

[23]

Maes M, Kubera M, Leunis JC, 2008, The gut-brain barrier in major depression: Intestinal mucosal dysfunction with an increased translocation of LPS from gram negative enterobacteria (leaky gut) plays a role in the inflammatory pathophysiology of depression. Neuro Endocrinol Lett, 29: 117–124.

[24]

Ait-Belgnaoui A, Durand H, Cartier C, et al., 2012, Prevention of gut leakiness by a probiotic treatment leads to attenuated HPA response to an acute psychological stress in rats. Psychoneuroendocrinology, 37: 1885–1895. https://doi.org/10.1016/j.psyneuen.2012.03.024

[25]

Keri S, Szabo C, Kelemen O, 2014, Expression of toll-like receptors in peripheral blood mononuclear cells and response to cognitive-behavioral therapy in major depressive disorder. Brain Behav Immun, 40: 235–243. https://doi.org/10.1016/j.bbi.2014.03.020

[26]

Eckburg PB, Bik EM, Bernstein CN, et al., 2005, Diversity of the human intestinal microbial flora. Science, 308: 1635–1638. https://doi.org/10.1126/science.1110591

[27]

Zheng P, Yang J, Li Y, et al., 2020, Gut microbial signatures can discriminate unipolar from bipolar depression. Adv Sci (Weinh), 7: 1902862. https://doi.org/10.1002/advs.201902862

[28]

Zheng P, Zeng B, Zhou C, et al., 2016, Gut microbiome remodeling induces depressive-like behaviors through a pathway mediated by the host’s metabolism. Mol Psychiatry, 21: 786–796. https://doi.org/10.1038/mp.2016.44 

[29]

Yang J, Zheng P, Li Y, et al., 2020, Landscapes of bacterial and metabolic signatures and their interaction in major depressive disorders. Sci Adv, 6: eaba8555. https://doi.org/10.1126/sciadv.aba8555

[30]

Lin CH, Chen CC, Chiang HL, et al., 2019, Altered gut microbiota and inflammatory cytokine responses in patients with Parkinson’s disease. J Neuroinflammation, 16: 129. https://doi.org/10.1186/s12974-019-1528-y

[31]

Bajaj JS, Hylemon PB, Ridlon JM, et al., 2012, Colonic mucosal microbiome differs from stool microbiome in cirrhosis and hepatic encephalopathy and is linked to cognition and inflammation. Am J Physiol Gastrointest Liver Physiol, 303: G675–G685. https://doi.org/10.1152/ajpgi.00152.2012

[32]

Pu Y, Zhang Q, Tang Z, et al., 2022, Fecal microbiota transplantation from patients with rheumatoid arthritis causes depression-like behaviors in mice through abnormal T cells activation. Transl Psychiatry, 12: 223. https://doi.org/10.1038/s41398-022-01993-z

[33]

Kelly JR, Borre Y, O’Brien C, et al., 2016, Transferring the blues: Depression-associated gut microbiota induces neurobehavioural changes in the rat. J Psychiatr Res, 82: 109–118.https://doi.org/10.1016/j.jpsychires.2016.07.019

[34]

Zhang Y, Huang R, Cheng M, et al., 2019, Gut microbiota from NLRP3-deficient mice ameliorates depressive-like behaviors by regulating astrocyte dysfunction via circHIPK2. Microbiome, 7: 116. https://doi.org/10.1186/s40168-019-0733-3

[35]

McGuinness AJ, Davis JA, Dawson SL, et al., 2022, A systematic review of gut microbiota composition in observational studies of major depressive disorder, bipolar disorder and schizophrenia. Mol Psychiatry, 27: 1920–1935. http://doi.org/10.1038/s41380-022-01456-3

[36]

Fung TC, Olson CA, Hsiao EY, 2017, Interactions between the microbiota, immune and nervous systems in health and disease. Nat Neurosci, 20: 145–155. https://doi.org/10.1038/nn.4476 

[37]

Rutsch A, Kantsjo JB, Ronchi F, 2020, The gut-brain axis: How microbiota and host inflammasome influence brain physiology and pathology. Front Immunol, 11: 604179. https://doi.org/10.3389/fimmu.2020.604179

[38]

Carlessi AS, Borba LA, Zugno AI, et al., 2021, Gut microbiota-brain axis in depression: The role of neuroinflammation. Eur J Neurosci, 53: 222–235. https://doi.org/10.1111/ejn.14631

[39]

Parker A, Fonseca S, Carding SR, 2020, Gut microbes and metabolites as modulators of blood-brain barrier integrity and brain health. Gut Microbes, 11: 135–157. https://doi.org/10.1080/19490976.2019.1638722

[40]

Zhao J, Bi W, Xiao S, et al., 2019, Neuroinflammation induced by lipopolysaccharide causes cognitive impairment in mice. Sci Rep, 9: 5790. https://doi.org/10.1038/s41598-019-42286-8

[41]

Kohler CA, Freitas TH, Maes M, et al., 2017, Peripheral cytokine and chemokine alterations in depression: A meta-analysis of 82 studies. Acta Psychiatr Scand, 135: 373–387. https://doi.org/10.1111/acps.12698

[42]

Alvarez-Mon MA, Gomez-Lahoz AM, Orozco A, et al., 2021, Expansion of CD4 T lymphocytes expressing interleukin 17 and tumor necrosis factor in patients with major depressive disorder. J Pers Med, 11: 220. https://doi.org/10.3390/jpm11030220

[43]

Zunszain PA, Anacker C, Cattaneo A, et al., 2011, Glucocorticoids, cytokines and brain abnormalities in depression. Prog Neuropsychopharmacol Biol Psychiatry, 35: 722–729. https://doi.org/10.1016/j.pnpbp.2010.04.011

[44]

Miller AH, Raison CL, 2016, The role of inflammation in depression: from evolutionary imperative to modern treatment target. Nat Rev Immunol, 16: 22–34. https://doi.org/10.1038/nri.2015.5

[45]

Wu H, Denna TH, Storkersen JN, et al., 2019, Beyond a neurotransmitter: The role of serotonin in inflammation and immunity. Pharmacol Res, 140: 100–114. https://doi.org/10.1016/j.phrs.2018.06.015

[46]

Correia AS, Cardoso A, Vale N, 2021, Highlighting immune system and stress in major depressive disorder, Parkinson’s, and Alzheimer’s Diseases, with a connection with serotonin. Int J Mol Sci, 22: 8525. https://doi.org/10.3390/ijms22168525 

[47]

Niklasson F, Agren H, 1984, Brain energy metabolism and blood-brain barrier permeability in depressive patients: Analyses of creatine, creatinine, urate, and albumin in CSF and blood. Biol Psychiatry, 19: 1183–1206.

[48]

Beurel E, Lowell JA, Jope RS, 2018, Distinct characteristics of hippocampal pathogenic T(H)17 cells in a mouse model of depression. Brain Behav Immun, 73: 180–191. https://doi.org/10.1016/j.bbi.2018.04.012

[49]

Beurel E, Harrington LE, Jope RS, 2013, Inflammatory T helper 17 cells promote depression-like behavior in mice. Biol Psychiatry, 73: 622–630. https://doi.org/10.1016/j.biopsych.2012.09.021

[50]

Lee J, Venna VR, Durgan DJ, et al., 2020, Young versus aged microbiota transplants to germ-free mice: Increased short-chain fatty acids and improved cognitive performance. Gut Microbes, 12: 1–14. https://doi.org/10.1080/19490976.2020.1814107 

[51]

O’Riordan KJ, Collins MK, Moloney GM, et al., 2022, Short chain fatty acids: Microbial metabolites for gut-brain axis signalling. Mol Cell Endocrinol, 546: 111572. https://doi.org/10.1016/j.mce.2022.111572

[52]

Tang CF, Wang CY, Wang JH, et al., Short-chain fatty acids ameliorate depressive-like behaviors of high fructose-fed mice by rescuing hippocampal neurogenesis decline and blood-brain barrier damage. Nutrients, 14: 1882. https://doi.org/10.3390/nu14091882

[53]

Rothhammer V, Mascanfroni ID, Bunse L, et al., 2016, Type I interferons and microbial metabolites of tryptophan modulate astrocyte activity and central nervous system inflammation via the aryl hydrocarbon receptor. Nat Med, 22: 586–957. https://doi.org/10.1038/nm.4106 

[54]

Scott SA, Fu J, Chang PV, 2020, Microbial tryptophan metabolites regulate gut barrier function via the aryl hydrocarbon receptor. Proc Natl Acad Sci U S A, 117: 19376–19387. https://doi.org/10.1073/pnas.2000047117

[55]

Barden N, 2004, Implication of the hypothalamic-pituitary-adrenal axis in the physiopathology of depression. J Psychiatry Neurosci, 29: 185–193.

[56]

Wang X, Li Y, Wu L, et al., 2021, Dysregulation of the gut-brain-skin axis and key overlapping inflammatory and immune mechanisms of psoriasis and depression. Biomed Pharmacother, 137: 111065. https://doi.org/10.1016/j.biopha.2020.111065

[57]

Sudo N, Chida Y, Aiba Y, et al., 2004, Postnatal microbial colonization programs the hypothalamic-pituitary-adrenal system for stress response in mice. J Physiol, 558: 263–275. https://doi.org/10.1113/jphysiol.2004.063388

[58]

Luckey TD, 1965, Effects of microbes on germfree animals. Adv Appl Microbiol, 7: 169–223. https://doi.org/10.1016/s0065-2164(08)70387-3

[59]

Hernandez-Chirlaque C, Aranda CH, Ocon B, et al., 2016, Germ-free and antibiotic-treated mice are highly susceptible to epithelial injury in DSS colitis. J Crohns Colitis, 10: 1324–1335. https://doi.org/10.1093/ecco-jcc/jjw096

[60]

Yang Y, Eguchi A, Wan X, et al., 2023, A role of gut-microbiota-brain axis via subdiaphragmatic vagus nerve in depression-like phenotypes in Chrna7 knock-out mice. Prog Neuropsychopharmacol Biol Psychiatry, 120: 110652. https://doi.org/10.1016/j.pnpbp.2022.110652

[61]

Rojas OL, Probstel Ak, Porfilio EA, et al., 2019, Recirculating intestinal IgA-producing cells regulate neuroinflammation via IL-10. Cell, 176: 610–624.e18. https://doi.org/10.1016/j.cell.2018.11.035

[62]

Fitzpatrick Z, Frazer G, Ferro A, et al., 2020, Gut-educated IgA plasma cells defend the meningeal venous sinuses. Nature, 587: 472–476. https://doi.org/10.1038/s41586-020-2886-4 

[63]

Sanmarco LM, Wheeler MA, Gutierrez-Vazquez C, et al., 2021, Gut-licensed IFNgamma(+) NK cells drive LAMP1(+) TRAIL(+) anti-inflammatory astrocytes. Nature, 590: 473–479. https://doi.org/10.1038/s41586-020-03116-4

[64]

Verkhratsky A, Illes P, Tang Y, et al., 2021, The anti-inflammatory astrocyte revealed: The role of the microbiome in shaping brain defences. Signal Transduct Target Ther, 6: 150. https://doi.org/10.1038/s41392-021-00577-5

[65]

Koh A, De Vadder F, Kovatcheva-Datchary P, et al., 2016, From dietary fiber to host physiology: Short-chain fatty acids as key bacterial metabolites. Cell, 165: 1332–1345. https://doi.org/10.1016/j.cell.2016.05.041

[66]

Cai Y, Folkerts J, Folkerts G, et al., 2020, Microbiota-dependent and -independent effects of dietary fibre on human health. Br J Pharmacol, 177: 1363–1381. https://doi.org/10.1111/bph.14871 

[67]

Z, Yi CX, Katiraei S, et al., 2018, Butyrate reduces appetite and activates brown adipose tissue via the gut-brain neural circuit. Gut, 67: 1269–1279. https://doi.org/10.1136/gutjnl-2017-314050

[68]

Arnoldussen IAC, Wiesmann M, Pelgrim CE, et al., 2017, Butyrate restores HFD-induced adaptations in brain function and metabolism in mid-adult obese mice. Int J Obes (Lond), 41: 935–944. https://doi.org/10.1038/ijo.2017.52

[69]

Liu J, Sun J, Wang F, et al., 2015, Neuroprotective effects of clostridium butyricum against vascular dementia in mice via metabolic butyrate. Biomed Res Int, 2015: 412946. https://doi.org/10.1155/2015/412946

[70]

Byrne CS, Chambers ES, Alhabeeb H, et al., 2016, Increased colonic propionate reduces anticipatory reward responses in the human striatum to high-energy foods. Am J Clin Nutr, 104: 5–14. https://doi.org/10.3945/ajcn.115.126706

[71]

van de Wouw M, Boehme M, Lyte JM, et al., 2018, Short-chain fatty acids: Microbial metabolites that alleviate stress-induced brain-gut axis alterations. J Physiol, 596: 4923–4944. https://doi.org/10.1113/JP276431

[72]

Falomir-Lockhart LJ, Cavazzutti GF, Gimenez E, et al., Fatty acid signaling mechanisms in neural cells: Fatty acid receptors. Front Cell Neurosci, 13: 162. https://doi.org/10.3389/fncel.2019.00162

[73]

Erny D, de Angelis AL, Jaitin D, et al., 2015, Host microbiota constantly control maturation and function of microglia in the CNS. Nat Neurosci, 18: 965–977. https://doi.org/10.1038/nn.4030

[74]

Maslowski KM, Vieira AT, Ng A, et al., 2009, Regulation of inflammatory responses by gut microbiota and chemoattractant receptor GPR43. Nature, 461: 1282–1286. https://doi.org/10.1038/nature08530

[75]

Oldendorf WH, 1973, Carrier-mediated blood-brain barrier transport of short-chain monocarboxylic organic acids. Am J Physiol, 224: 1450–1453. https://doi.org/10.1152/ajplegacy.1973.224.6.1450

[76]

Agirman G, Hsiao EY, 2021, SnapShot: The microbiota-gut-brain axis. Cell, 184: 2524–2524.e1. https://doi.org/10.1016/j.cell.2021.03.022

[77]

Xiao S, Jiang S, Qian D, et al., 2020, Modulation of microbially derived short-chain fatty acids on intestinal homeostasis, metabolism, and neuropsychiatric disorder. Appl Microbiol Biotechnol, 104: 589–601. https://doi.org/10.1007/s00253-019-10312-4

[78]

Hoyles L, Snelling T, Umlai UK, et al., 2018, Microbiome-host systems interactions: Protective effects of propionate upon the blood-brain barrier. Microbiome, 6: 55. https://doi.org/10.1186/s40168-018-0439-y

[79]

Braniste V, Al-Asmakh M, Kowal C, et al., 2014, The gut microbiota influences blood-brain barrier permeability in mice. Sci Transl Med, 6: 263ra158. https://doi.org/10.1126/scitranslmed.3009759

[80]

Morris G, Berk M, Carvalho A, et al., 2017, The role of the microbial metabolites including tryptophan catabolites and short chain fatty acids in the pathophysiology of immune-inflammatory and neuroimmune disease. Mol Neurobiol, 54: 4432–4451. https://doi.org/10.1007/s12035-016-0004-2

[81]

Liu RT, Rowan-Nash AD, Sheehan AE, et al., 2020, Reductions in anti-inflammatory gut bacteria are associated with depression in a sample of young adults. Brain Behav Immun, 88: 308–324. https://doi.org/10.1016/j.bbi.2020.03.026

[82]

Stilling RM, van de Wouw M, Clarke G, et al., 2016, The neuropharmacology of butyrate: The bread and butter of the microbiota-gut-brain axis? Neurochem Int, 99: 110–132. https://doi.org/10.1016/j.neuint.2016.06.011

[83]

Yamawaki Y, Yoshioka N, Nozaki K, et al., 2018, Sodium butyrate abolishes lipopolysaccharide-induced depression-like behaviors and hippocampal microglial activation in mice. Brain Res, 1680: 13–38. https://doi.org/10.1016/j.brainres.2017.12.004

[84]

Rao J, Qiao Y, Xie R, et al., 2021, Fecal microbiota transplantation ameliorates stress-induced depression-like behaviors associated with the inhibition of glial and NLRP3 inflammasome in rat brain. J Psychiatr Res, 137: 147–157. https://doi.org/10.1016/j.jpsychires.2021.02.057

[85]

Bercik P, Denou E, Collins J, et al., 2011, The intestinal microbiota affect central levels of brain-derived neurotropic factor and behavior in mice. Gastroenterology, 141: 599–609, 609 e1–3. https://doi.org/10.1053/j.gastro.2011.04.052

[86]

Cai T, Shi X, Yuan LZ, et al., 2019, Fecal microbiota transplantation in an elderly patient with mental depression. Int Psychogeriatr, 31: 1525–1526. http://doi.org/10.1017/S1041610219000115

[87]

Meyyappan AC, Forth E, Wallace CJK, et al., 2020, Effect of fecal microbiota transplant on symptoms of psychiatric disorders: A systematic review. BMC Psychiatry, 20: 299. https://doi.org/10.1186/s12888-020-02654-5

[88]

Fond GB, Lagier JC, Honore S, et al., 2020, Microbiota-orientated treatments for major depression and schizophrenia. Nutrients, 12: 1024. https://doi.org/10.3390/nu12041024 

[89]

Green JE, Berk M, Loughman A, et al., 2021, FMT for psychiatric disorders: Following the brown brick road into the future. Bipolar Disord, 23: 651–655. https://doi.org/10.1111/bdi.13124 

[90]

Gupta S, Mullish BH, Allegretti JR, 2021, Fecal microbiota transplantation: The evolving risk landscape. Am J Gastroenterol, 116: 647–656. http://doi.org/10.14309/ajg.0000000000001075 

[91]

Grigoryan Z, Shen MJ, Twardus SW, et al., 2020, Fecal microbiota transplantation: Uses, questions, and ethics. Med Microecol, 6: 100027. https://doi.org/10.1016/j.medmic.2020.100027

[92]

Wang S, Xu M, Wang W, et al., 2016, Systematic review: Adverse events of fecal microbiota transplantation. PLoS One, 11: e0161174. https://doi.org/10.1371/journal.pone.0161174 

[93]

DeFilipp Z, Bloom PP, Soto MT, et al., 2019, Drug-resistant E. coli bacteremia transmitted by fecal microbiota transplant. N Engl J Med, 381: 2043–2050. https://doi.org/10.1056/NEJMoa1910437

[94]

Zellmer C, Sater MR, Huntley MH, et al., 2021, Shiga toxin-producing Escherichia coli transmission via fecal microbiota transplant. Clin Infect Dis, 72: e876–e880. https://doi.org/10.1093/cid/ciaa1486 

[95]

Vrieze A, Van Nood E, Holleman F, et al., 2012, Transfer of intestinal microbiota from lean donors increases insulin sensitivity in individuals with metabolic syndrome. Gastroenterology, 143: 913–916.e7. https://doi.org/10.1053/j.gastro.2012.06.031 

[96]

Ridaura VK, Faith JJ, Rey FE, et al., 2013, Gut microbiota from twins discordant for obesity modulate metabolism in mice. Science, 341: 1241214. https://doi.org/10.1126/science.1241214

[97]

Vijay-Kumar M, Aitken JD, Carvalho FA, et al., 2010, Metabolic syndrome and altered gut microbiota in mice lacking Toll-like receptor 5. Science, 328: 228–231. https://doi.org/10.1126/science.1179721 

[98]

Kassam Z, Dubois N, Ramakrishna B, et al., 2019, Donor screening for fecal microbiota transplantation. N Engl J Med, 381: 2070–2072. https://doi.org/10.1056/NEJMc1913670

[99]

Bear TLK, Dalziel JE, Coad J, et al., 2020, The role of the gut microbiota in dietary interventions for depression and anxiety. Adv Nutr, 11: 890–907. https://doi.org/10.1093/advances/nmaa016

[100]

Li X, Chen M, Yao Z, et al., 2022, Dietary inflammatory potential and the incidence of depression and anxiety: A meta-analysis. J Health Popul Nutr, 41: 24. https://doi.org/10.1186/s41043-022-00303-z 

[101]

Chen GQ, Peng CL, Lian Y, et al., 2021, Association between dietary inflammatory index and mental health: A systematic review and dose-response meta-analysis. Front Nutr, 8: 662357. https://doi.org/10.3389/fnut.2021.662357

[102]

Ruusunen A, Lehto SM, Mursu J, et al., 2014, Dietary patterns are associated with the prevalence of elevated depressive symptoms and the risk of getting a hospital discharge diagnosis of depression in middle-aged or older Finnish men. J Affect Disord, 159: 1–6. https://doi.org/10.1016/j.jad.2014.01.020

[103]

Parletta N, Zarnowiecki D, Cho J, et al., 2019, A mediterranean-style dietary intervention supplemented with fish oil improves diet quality and mental health in people with depression: A randomized controlled trial (HELFIMED). Nutr Neurosci, 22: 474–487. https://doi.org/10.1080/1028415X.2017.1411320

[104]

Akbaraly TN, Brunner EJ, Ferrie JE, et al., 2009, Dietary pattern and depressive symptoms in middle age. Br J Psychiatry, 195: 408–413. https://doi.org/10.1192/bjp.bp.108.058925

[105]

Nanri A, Kimura Y, Matsushita Y, et al., 2020, Dietary patterns and depressive symptoms among Japanese men and women. Eur J Clin Nutr, 64: 832–839. https://doi.org/10.1038/ejcn.2010.86 

[106]

Gougeon L, Payette H, Morais JA, et al., 2016, Intakes of folate, Vitamin B6 and B12 and risk of depression in community-dwelling older adults: The quebec longitudinal study on nutrition and aging. Eur J Clin Nutr, 70: 380–385. https://doi.org/10.1038/ejcn.2015.202

[107]

Anglin RE, Samaan Z, Walter SD, et al., 2013, Vitamin D deficiency and depression in adults: Systematic review and meta-analysis. Br J Psychiatry, 202: 100–107. https://doi.org/10.1192/bjp.bp.111.106666

[108]

Jacka FN, Maes M, Pasco JA, et al., 2012, Nutrient intakes and the common mental disorders in women. J Affect Disord, 141: 79–85. https://doi.org/10.1016/j.jad.2012.02.018 

[109]

Miller AL, 2008, The methylation, neurotransmitter, and antioxidant connections between folate and depression. Altern Med Rev, 13: 216–226.

[110]

Wang J, Um P, Dickerman BA, et al., 2018, Zinc, magnesium, selenium and depression: A review of the evidence, potential mechanisms and implications. Nutrients, 10: 584. https://doi.org/10.3390/nu10050584 

[111]

Grosso G, Pajak A, Marventano S, et al., 2014, Role of omega-3 fatty acids in the treatment of depressive disorders: A comprehensive meta-analysis of randomized clinical trials. PLoS One, 9: e96905. https://doi.org/10.1371/journal.pone.0096905

[112]

Appleton KM, Rogers PJ, Ness AR, 2010, Updated systematic review and meta-analysis of the effects of n-3 long-chain polyunsaturated fatty acids on depressed mood. Am J Clin Nutr, 91: 757–770. https://doi.org/10.3945/ajcn.2009.28313

[113]

Pusceddu MM, El Aidy S, Crispie F, et al., 2015, N-3 Polyunsaturated fatty acids (PUFAs) reverse the impact of early-life stress on the gut microbiota. PLoS One, 10: e0139721. https://doi.org/10.1371/journal.pone.0139721

[114]

Robertson RC, Oriach CS, Murphy K, et al., 2017, Omega-3 polyunsaturated fatty acids critically regulate behaviour and gut microbiota development in adolescence and adulthood. Brain Behav Immun, 59: 21–37. https://doi.org/10.1016/j.bbi.2016.07.145

[115]

Grosso G, Galvano F, Marventano S, et al., 2014, Omega-3 fatty acids and depression: scientific evidence and biological mechanisms. Oxid Med Cell Longev, 2014: 313570. https://doi.org/10.1155/2014/313570

[116]

Savignac HM, Corona G, Mills H, et al., 2013, Prebiotic feeding elevates central brain derived neurotrophic factor, N-methyl-D-aspartate receptor subunits and D-serine. Neurochem Int, 63: 756–764. https://doi.org/10.1016/j.neuint.2013.10.006 

[117]

Schmidt K, Cowen PJ, Harmer CJ, et al., 2015, Prebiotic intake reduces the waking cortisol response and alters emotional bias in healthy volunteers. Psychopharmacology (Berl), 232: 1793–1801. https://doi.org/10.1007/s00213-014-3810-0

[118]

Kunz-Ebrecht SR, Kirschbaum C, Marmot M, et al., 2004, Differences in cortisol awakening response on work days and weekends in women and men from the Whitehall II cohort. Psychoneuroendocrinology, 29: 516–528. https://doi.org/10.1016/s0306-4530(03)00072-6

[119]

Pruessner JC, Wolf OT, Hellhammer DH, et al., 1997, Free cortisol levels after awakening: A reliable biological marker for the assessment of adrenocortical activity. Life Sci, 61: 2539–2549. https://doi.org/10.1016/s0024-3205(97)01008-4

[120]

Burokas A, Arboleya S, Moloney RD, et al., 2017, Targeting the microbiota-gut-brain axis: Prebiotics have anxiolytic and antidepressant-like effects and reverse the impact of  chronic stress in mice. Biol Psychiatry, 82: 472–487. https://doi.org/10.1016/j.biopsych.2016.12.031

[121]

Neufeld KA, O’Mahony SM, Hoban AE, et al., 2019, Neurobehavioural effects of Lactobacillus rhamnosus GG alone and in combination with prebiotics polydextrose and galactooligosaccharide in male rats exposed to early-life stress. Nutr Neurosci, 22: 425–434. https://doi.org/10.1080/1028415X.2017.1397875 

[122]

Haghighat N, Rajabi S, Mohammadshahi M, 2021, Effect of synbiotic and probiotic supplementation on serum brain-derived neurotrophic factor level, depression and anxiety symptoms in hemodialysis patients: A randomized, double-blinded, clinical trial. Nutr Neurosci, 24: 490–499. https://doi.org/10.1080/1028415X.2019.1646975

[123]

Marx W, Lane M, Hockey M, et al., 2021, Diet and depression: Exploring the biological mechanisms of action. Mol Psychiatry, 26: 134–150. https://doi.org/10.1038/s41380-020-00925-x

[124]

Okereke OI, Vyas CM, Mischoulon D, et al., 2021, Effect of long-term supplementation with marine omega-3 fatty acids vs placebo on risk of depression or clinically relevant depressive symptoms and on change in mood scores: A randomized clinical trial. JAMA, 326: 2385–2394. https://doi.org/10.1001/jama.2021.21187

[125]

Liu RT, Walsh RFL, Sheehan AE, 2019, Prebiotics and probiotics for depression and anxiety: A systematic review and meta-analysis of controlled clinical trials. Neurosci Biobehav Rev, 102: 13–23. https://doi.org/10.1016/j.neubiorev.2019.03.023

Conflict of interest
The authors declare that they have no conflicts of interest.
Share
Back to top
Advanced Neurology, Electronic ISSN: 2810-9619 Published by AccScience Publishing