Skip to main content

Advertisement

Log in

Sex Differences in the Gut-Brain Axis: Implications for Mental Health

  • SEX AND GENDER ISSUES IN BEHAVIORAL HEALTH (CN EPPERSON AND L HANTSOO, SECTION EDITORS)
  • Published:
Current Psychiatry Reports Aims and scope Submit manuscript

Abstract

Purpose of Review

The purpose of this article is to highlight how sex differences in the gut-brain axis may contribute to the discrepancies in incidence of neurodevelopmental, psychiatric, and neurodegenerative disorders between females and males. We focus on autism spectrum disorder, psychotic disorders, stress and anxiety disorders, depression, Alzheimer’s disease, and Parkinson’s disease and additionally discuss the comorbidity between inflammatory bowel disorder and mental health disorders.

Recent Findings

Human and animal studies show that sex may modify the relationship between the gut or immune system and brain and behavior. Sex also appears to modify the effect of microbial treatments such as probiotics and antibiotics on brain and behavior.

Summary

There is emerging evidence that assessing the role of sex in the gut-brain axis may help elucidate the etiology of and identify effective treatments for neurodevelopmental, psychiatric, and neurodegenerative disorders.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1

Similar content being viewed by others

References

Papers of particular interest, published recently, have been highlighted as: • Of importance •• Of major importance

  1. Afifi M. Gender differences in mental health. Singapore Med J Stamford Publishing PTE LTD. 2007;48:385.

    CAS  Google Scholar 

  2. May T, Adesina I, McGillivray J, Rinehart NJ. Sex differences in neurodevelopmental disorders. Curr Opin Neurol LWW. 2019;32:622–6.

    Article  Google Scholar 

  3. Tierney MC, Curtis AF, Chertkow H, Rylett RJ. Integrating sex and gender into neurodegeneration research: a six-component strategy. Alzheimer’s Dement Transl Res Clin Interv Elsevier. 2017;3:660–7.

    Article  Google Scholar 

  4. •• Jaggar M, Rea K, Spichak S, Dinan TG, Cryan JF. You’ve got male: sex and the microbiota-gut-brain axis across the lifespan. Front Neuroendocrinol. 2020; This comprehensive review is an excellent complement to the present study, as it reviews the role of sex in the gut-brain across the lifespan.

  5. Jašarević E, Morrison KE, Bale TL. Sex differences in the gut microbiome - brain axis across the lifespan. Philos Trans R Soc B Biol Sci. 2016;371:12–7.

    Article  Google Scholar 

  6. Elsesser K. The Myth Of Biological Sex. Forbes [Internet]. 2020 Jun 15; Available from: https://www.forbes.com/sites/kimelsesser/2020/06/15/the-myth-of-biological-sex/

  7. Carabotti M, Scirocco A, Maselli MA, Severi C. The gut-brain axis: interactions between enteric microbiota, central and enteric nervous systems. Ann Gastroenterol Q Publ Hell Soc Gastroenterol. The Hellenic Society of Gastroenterology; 2015;28:203.

  8. Gill SR, Pop M, DeBoy RT, Eckburg PB, Turnbaugh PJ, Samuel BS, et al. Metagenomic analysis of the human distal gut microbiome. Science (80- ). American Association for the Advancement of Science; 2006;312:1355–9.

  9. Wang H-X, Wang Y-P. Gut microbiota-brain axis. Chin Med J (Engl). Wolters Kluwer Health; 2016;129:2373.

  10. Kavvadia M, Santis GLD, Cascapera S, Lorenzo AD. Psychobiotics as integrative therapy for neuropsychiatric disorders with special emphasis on the microbiota-gut-brain axis. Biomed Prev. 2017;2.

  11. • Liang D, Leung RK-K, Guan W, Au WW. Involvement of gut microbiome in human health and disease: brief overview, knowledge gaps and research opportunities. Gut Pathog BioMed Central. 2018;10:1–9 This article reviews the current knowledge base about the microbiome and proposes future directions for novel investigations.

    Article  Google Scholar 

  12. • Dinan TG, Cryan JF. Brain-gut-microbiota axis and mental health. Psychosom Med LWW. 2017;79:920–6 This article reviews the pre-clinical evidence supporting a linkage between the gut and mental health, and questions if these links will still be present in patients.

    Article  Google Scholar 

  13. Borre YE, O’Keeffe GW, Clarke G, Stanton C, Dinan TG, Cryan JF. Microbiota and neurodevelopmental windows: implications for brain disorders. Trends Mol Med [Internet]. Elsevier Ltd; 2014;20:509–518. Available from: https://doi.org/10.1016/j.molmed.2014.05.002

  14. • Clapp M, Aurora N, Herrera L, Bhatia M, Wilen E, Wakefield S. Gut microbiota’s effect on mental health: the gut-brain axis. Clin Pract. 2017; This review article discusses the connection between dysbiosis and mental health disorders, as well as the potential of probiotics as a treatment option.

  15. Dinan TG, Stanton C, Cryan JF. Psychobiotics: a novel class of psychotropic. Biol Psychiatry Elsevier. 2013;74:720–6.

    Article  CAS  Google Scholar 

  16. Kerry RG, Patra JK, Gouda S, Park Y, Shin H-S, Das G. Benefaction of probiotics for human health: a review. J food drug anal Elsevier. 2018;26:927–39.

    Article  Google Scholar 

  17. American Psychiatric Association. Diagnostic and statistical manual of mental disorders (DSM-5®). American Psychiatric Pub; 2013.

  18. Shaw KA, Maenner MJ, Baio J, Washington A, Christensen DL, Wiggins LD, et al. Early identification of autism spectrum disorder among children aged 4 years - Early autism and developmental disabilities monitoring network, six sites, United States, 2016. MMWR Surveill Summ 2020;

  19. Maenner MJ. Prevalence of autism spectrum disorder among children aged 8 years—autism and developmental disabilities monitoring network, 11 sites, United States, 2016. MMWR Surveill Summ. 2020;69.

  20. Schieve LA, Gonzalez V, Boulet SL, Visser SN, Rice CE, Braun KVN, et al. Concurrent medical conditions and health care use and needs among children with learning and behavioral developmental disabilities, National Health Interview Survey, 2006–2010. Res Dev Disabil Elsevier; 2012;33:467–476.

  21. Marler S, Ferguson BJ, Lee EB, Peters B, Williams KC, McDonnell E, et al. Association of rigid-compulsive behavior with functional constipation in autism spectrum disorder. J Autism Dev Disord. 2017.

  22. Ho LKH, Tong VJW, Syn N, Nagarajan N, Tham EH, Tay SK, et al. Gut microbiota changes in children with autism spectrum disorder: a systematic review. Gut Pathog Springer. 2020;12:6.

    Article  CAS  Google Scholar 

  23. •• Wang M, Zhou J, He F, Cai C, Wang H, Wang Y, et al. Alteration of gut microbiota-associated epitopes in children with autism spectrum disorders. Brain Behav Immun. 2019; This study of children with autism found that sex was associated with gut microbiome-associated epitopes, providing evidence that sex may influence the gut-immune connection.

  24. •• Coretti L, Cristiano C, Florio E, Scala G, Lama A, Keller S, et al. Sex-related alterations of gut microbiota composition in the BTBR mouse model of autism spectrum disorder. Sci Rep Nature Publishing Group. 2017;7:45356 This original study showed that in a mouse model of autism, sex was differential associated with relative abundance of gut microbes and with immune expression.

    CAS  Google Scholar 

  25. Foley KA, MacFabe DF, Vaz A, Ossenkopp K-P, Kavaliers M. Sexually dimorphic effects of prenatal exposure to propionic acid and lipopolysaccharide on social behavior in neonatal, adolescent, and adult rats: implications for autism spectrum disorders. Int J Dev Neurosci Elsevier. 2014;39:68–78.

    Article  CAS  Google Scholar 

  26. MacFabe DF, Rodríguez-Capote K, Hoffman JE, Franklin AE, Mohammad-Asef Y, Taylor AR, et al. A novel rodent model of autism: intraventricular infusions of propionic acid increase locomotor activity and induce neuroinflammation and oxidative stress in discrete regions of adult rat brain. Am J Biochem Biotechnol. 2008;4:146–66.

    Article  CAS  Google Scholar 

  27. MacFabe DF, Cain DP, Rodriguez-Capote K, Franklin AE, Hoffman JE, Boon F, et al. Neurobiological effects of intraventricular propionic acid in rats: possible role of short chain fatty acids on the pathogenesis and characteristics of autism spectrum disorders. Behav Brain Res Elsevier. 2007;176:149–69.

    Article  CAS  Google Scholar 

  28. Garbett KA, Hsiao EY, Kalman S, Patterson PH, Mirnics K. Effects of maternal immune activation on gene expression patterns in the fetal brain. Transl Psychiatry [Internet]. 2012;2:e98. Available from: http://www.ncbi.nlm.nih.gov/pubmed/22832908.

  29. Nouel D, Burt M, Zhang Y, Harvey L, Boksa P. Prenatal exposure to bacterial endotoxin reduces the number of GAD67-and reelin-immunoreactive neurons in the hippocampus of rat offspring. Eur Neuropsychopharmacol Elsevier; 2012;22:300–307.

  30. Depino AM. Peripheral and central inflammation in autism spectrum disorders. Mol Cell Neurosci Elsevier. 2013;53:69–76.

    Article  CAS  Google Scholar 

  31. Malkova N V., Yu CZ, Hsiao EY, Moore MJ, Patterson PH. Maternal immune activation yields offspring displaying mouse versions of the three core symptoms of autism. Brain Behav Immun [Internet]. Elsevier Inc.; 2012;26:607–16. Available from: https://doi.org/10.1016/j.bbi.2012.01.011.

  32. McGrath J, Saha S, Chant D, Welham J. Schizophrenia: A concise overview of incidence, prevalence, and mortality. Epidemiol. Rev. 2008.

  33. Jongsma HE, Turner C, Kirkbride JB, Jones PB. International incidence of psychotic disorders, 2002–17: a systematic review and meta-analysis. Lancet Public Heal Elsevier. 2019;4:e229–44.

    Article  Google Scholar 

  34. Castro-Nallar E, Bendall ML, Pérez-Losada M, Sabuncyan S, Severance EG, Dickerson FB, et al. Composition, taxonomy and functional diversity of the oropharynx microbiome in individuals with schizophrenia and controls. PeerJ. 2015.

  35. Severance EG, Prandovszky E, Castiglione J, Yolken RH. Gastroenterology issues in schizophrenia: why the gut matters. Curr Psychiatry Rep. 2015.

  36. Schwarz E, Maukonen J, Hyytiäinen T, Kieseppä T, Orešič M, Sabunciyan S, et al. Analysis of microbiota in first episode psychosis identifies preliminary associations with symptom severity and treatment response. Schizophr Res. 2018.

  37. Čiháková D, Eaton WW, Talor MV, Harkus UH, Demyanovich HK, Rodriguez K, et al. Gliadin-related antibodies in schizophrenia. Schizophr Res. 2018;195:585.

    Article  Google Scholar 

  38. Jackson J, Eaton W, Cascella N, Fasano A, Santora D, Sullivan K, et al. Gluten sensitivity and relationship to psychiatric symptoms in people with schizophrenia. Schizophr Res Elsevier. 2014;159:539–42.

    Article  Google Scholar 

  39. Kelly DL, Demyanovich HK, Rodriguez KM, Čiháková D, Talor M V, McMahon RP, et al. Randomized controlled trial of a gluten-free diet in patients with schizophrenia positive for antigliadin antibodies (AGA IgG): a pilot feasibility study. J psychiatry Neurosci JPN. Can Med Assoc; 2019;44:269.

  40. Severance EG, Gressitt KL, Stallings CR, Katsafanas E, Schweinfurth LA, Savage CL, et al. Candida albicans exposures, sex specificity and cognitive deficits in schizophrenia and bipolar disorder. npj Schizophr. 2016.

  41. •• Severance EG, Gressitt KL, Stallings CR, Katsafanas E, Schweinfurth LA, Savage CLG, et al. Probiotic normalization of Candida albicans in schizophrenia: a randomized, placebo-controlled, longitudinal pilot study. Brain Behav Immun [Internet]. Elsevier Inc.; 2017;62:41–5. Available from: https://doi.org/10.1016/j.bbi.2016.11.019. This clinical study found that male, but not female, patients with schizophrenia treated with probiotics had reduced antibodies to Candida albicans and improvements in symptoms of schizophrenia.

  42. Kessler RC, Petukhova M, Sampson NA, Zaslavsky AM, Wittchen HU. Twelve-month and lifetime prevalence and lifetime morbid risk of anxiety and mood disorders in the United States. Int J Methods Psychiatr Res. 2012.

  43. Jiang H yin, Zhang X, Yu Z he, Zhang Z, Deng M, Zhao J hua, et al. Altered gut microbiota profile in patients with generalized anxiety disorder. J Psychiatr Res. 2018.

  44. Tran N, Zhebrak M, Yacoub C, Pelletier J, Hawley D. The gut-brain relationship: investigating the effect of multispecies probiotics on anxiety in a randomized placebo-controlled trial of healthy young adults. J Affect Disord. 2019.

  45. Christian LM, Galley JD, Hade EM, Schoppe-Sullivan S, Kamp Dush C, Bailey MT. Gut microbiome composition is associated with temperament during early childhood. Brain Behav Immun. 2015.

  46. •• Audet MC. Stress-induced disturbances along the gut microbiota-immune-brain axis and implications for mental health: does sex matter? Front Neuroendocrinol. 2019. This article reviews how the sexually dimorphic alterations to the microbiome and sex hormones may explain the female bias in mood and stress disorders.

  47. Sovijit WN, Sovijit WE, Pu S, Usuda K, Inoue R, Watanabe G, et al. Ovarian progesterone suppresses depression and anxiety-like behaviors by increasing the Lactobacillus population of gut microbiota in ovariectomized mice. Neurosci res. Elsevier; 2019.

  48. Braniste V, Leveque M, Buisson-Brenac C, Bueno L, Fioramonti J, Houdeau E. Oestradiol decreases colonic permeability through oestrogen receptor β-mediated up-regulation of occludin and junctional adhesion molecule-A in epithelial cells. J Physiol Wiley Online Library. 2009;587:3317–28.

    CAS  Google Scholar 

  49. • Cowan CSM, Richardson R. Early-life stress leads to sex-dependent changes in pubertal timing in rats that are reversed by a probiotic formulation. Dev Psychobiol Wiley Online Library. 2019;61:679–87 This original study demonstrated that in rats, early life maternal separation stress with differentially related to pubertal timing in male versus female animals, but probiotic treatment restored timing in both sexes.

    Article  CAS  Google Scholar 

  50. •• Davis DJ, Hecht PM, Jasarevic E, Beversdorf DQ, Will MJ, Fritsche K, et al. Sex-specific effects of docosahexaenoic acid (DHA) on the microbiome and behavior of socially-isolated mice. Brain Behav Immun. 2017; This study found that treatment of male, but not female, rats with an omega-3 fatty acid following social isolation led to reductions in symptoms of anxiety and depression, suggesting behavioral treatments affecting the gut may have differential affects by sex.

  51. Ferrari AJ, Somerville AJ, Baxter AJ, Norman R, Patten SB, Vos T, et al. Global variation in the prevalence and incidence of major depressive disorder: a systematic review of the epidemiological literature. Psychol Med Cambridge University Press. 2013;43:471.

    CAS  Google Scholar 

  52. Substance Abuse and Mental Health Services Administration. Results from the 2017 National Survey on Drug Use and Health. Vol I Summ Natl Find [Internet] 2018; Available from: https://www.samhsa.gov/data/sites/default/files/cbhsq-reports/NSDUHDetailedTabs2017/NSDUHDetailedTabs2017.htm#tab8-56A

  53. Maes M, Kubera M, Leunis JC. 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. 2008.

  54. Berg RD. Bacterial translocation from the gastrointestinal tract. Mech Pathog Enteric Dis 2. Springer; 1999. p. 11–30.

  55. Jiang H, Ling Z, Zhang Y, Mao H, Ma Z, Yin Y, et al. Altered fecal microbiota composition in patients with major depressive disorder. Brain Behav Immun. 2015;

  56. Naseribafrouei A, Hestad K, Avershina E, Sekelja M, Linløkken A, Wilson R, et al. Correlation between the human fecal microbiota and depression. Neurogastroenterol Motil. 2014.

  57. Messaoudi M, Lalonde R, Violle N, Javelot H, Desor D, Nejdi A, et al. Assessment of psychotropic-like properties of a probiotic formulation (Lactobacillus helveticus R0052 and Bifidobacterium longum R0175) in rats and human subjects. Br J Nutr. 2011.

  58. Foster J A, McVey Neufeld KA. Gut-brain axis: how the microbiome influences anxiety and depression. Trends Neurosci [Internet]. Elsevier Ltd; 2013;36:305–12. Available from: https://doi.org/10.1016/j.tins.2013.01.005.

  59. Emge JR, Huynh K, Miller EN, Kaur M, Reardon C, Barrett KE, et al. Modulation of the microbiota-gut-brain axis by probiotics in a murine model of inflammatory bowel disease. Am J Physiol Gastrointest Liver Physiol. 2016.

  60. Luo J, Wang T, Liang S, Hu X, Li W, Jin F. Experimental gastritis leads to anxiety- and depression-like behaviors in female but not male rats. Behav Brain Funct. 2013.

  61. Liu L, Li Q, Sapolsky R, Liao M, Mehta K, Bhargava A, et al. Transient gastric irritation in the neonatal rats leads to changes in hypothalamic CRF expression, depression- and anxiety-like behavior as adults. PLoS One. 2011.

  62. •• Chen JJ, Zheng P, Liu YY, Zhong XG, Wang HY, Guo YJ, et al. Sex differences in gut microbiota in patients with major depressive disorder. Neuropsychiatr Dis Treat. 2018. This study identified specific sex differences in the microbiota of male and female MDD patients, specifically noting the differential levels of phyla Actinobacteria and Bacteroidetes.

  63. Dickerson F, Severance E, Yolken R. The microbiome, immunity, and schizophrenia and bipolar disorder. Brain Behav Immun. 2017.

  64. Nguyen TT, Kosciolek T, Eyler LT, Knight R, Jeste DV. Overview and systematic review of studies of microbiome in schizophrenia and bipolar disorder. J. Psychiatr. Res. 2018.

  65. Coello K, Hansen TH, Sørensen N, Munkholm K, Kessing LV, Pedersen O, et al. Gut microbiota composition in patients with newly diagnosed bipolar disorder and their unaffected first-degree relatives. Brain Behav Immun. 2019.

  66. Flowers SA, Evans SJ, Ward KM, McInnis MG, Ellingrod VL. Interaction between atypical antipsychotics and the gut microbiome in a bipolar disease cohort. Pharmacotherapy. 2017;

  67. Karling P, Maripuu M, Wikgren M, Adolfsson R, Norrback KF. Association between gastrointestinal symptoms and affectivity in patients with bipolar disorder. World J Gastroenterol. 2016.

  68. Alzheimer’s association. 2019 Alzheimer’s disease facts and figures includes a special report on Alzheimer’s detection in the primary care setting: connecting patients and physicians. Alzheimers Dement Vol 15, Issue 3. 2019.

  69. Chêne G, Beiser A, Au R, Preis SR, Wolf PA, Dufouil C, et al. Gender and incidence of dementia in the Framingham Heart Study from mid-adult life. Alzheimer’s Dement. 2015;

  70. Vogt NM, Kerby RL, Dill-McFarland KA, Harding SJ, Merluzzi AP, Johnson SC, et al. Gut microbiome alterations in Alzheimer’s disease. Sci Rep [Internet]. 2017 [cited 2018 Dec 31];7:13537. Available from: http://www.nature.com/articles/s41598-017-13601-y

  71. Kowalski K, Mulak A. Brain-gut-microbiota axis in Alzheimer’s disease. J Neurogastroenterol Motil. 2019.

  72. Mielke MM, Vemuri P, Rocca WA. Clinical epidemiology of Alzheimer’s disease: assessing sex and gender differences. Epidemiol: Clin; 2014.

    Google Scholar 

  73. Pinares-Garcia P, Stratikopoulos M, Zagato A, Loke H, Lee J. Sex: A significant risk factor for neurodevelopmental and neurodegenerative disorders. Brain Sci. 2018.

  74. • Buckley RF, Mormino EC, Rabin JS, Hohman TJ, Landau S, Hanseeuw BJ, et al. Sex differences in the association of global amyloid and regional tau deposition measured by positron emission tomography in clinically normal older adults. JAMA Neurol. 2019; This neuroimaging study identified that clinically typical women exhibited greater AD pathology than men, demonstrating biological support for sex differences in AD development.

  75. •• Cox LM, Abou-El-Hassan H, Maghzi AH, Vincentini J, Weiner HL. The sex-specific interaction of the microbiome in neurodegenerative diseases. Brain Res. 2019. This comprehensive review of human and animal studies evaluates how sex-specific interactions within the microbiota influences the development and presentation of neurodegenerative diseases.

  76. Merlo S, Spampinato SF, Sortino MA. Estrogen and Alzheimer’s disease: still an attractive topic despite disappointment from early clinical results. Eur J Pharmacol. Elsevier; 2017;817:51–8.

  77. Wang C, Zhang F, Jiang S, Siedlak SL, Shen L, Perry G, et al. Estrogen receptor-α is localized to neurofibrillary tangles in Alzheimer’s disease. Sci Rep Nature Publishing Group. 2016;6:1–10.

    Google Scholar 

  78. Baker JM, Al-Nakkash L, Herbst-Kralovetz MM. Estrogen–gut microbiome axis: physiological and clinical implications. Maturitas Elsevier. 2017;103:45–53.

    Article  CAS  Google Scholar 

  79. Flores R, Shi J, Fuhrman B, Xu X, Veenstra TD, Gail MH, et al. Fecal microbial determinants of fecal and systemic estrogens and estrogen metabolites: a cross-sectional study. J Transl Med Springer. 2012;10:253.

    Article  CAS  Google Scholar 

  80. Minter MR, Zhang C, Leone V, Ringus DL, Zhang X, Oyler-Castrillo P, et al. Antibiotic-induced perturbations in gut microbial diversity influences neuro-inflammation and amyloidosis in a murine model of Alzheimer’s disease. Sci Rep. 2016.

  81. •• Minter MR, Hinterleitner R, Meisel M, Zhang C, Leone V, Zhang X, et al. Antibiotic-induced perturbations in microbial diversity during post-natal development alters amyloid pathology in an aged APPSWE/PS1ΔE9 murine model of Alzheimer’s disease. Sci Rep. 2017; This original study showed that early post-natal microbial changes associated with broad-spectrum antibiotic treatment led to reductions in neuropathology among male mice.

  82. Schneider RB, Iourinets J, Richard IH. Parkinson’s disease psychosis: presentation, diagnosis and management. Manag: Neurodegener. Dis; 2017.

    Google Scholar 

  83. Hirsch L, Jette N, Frolkis A, Steeves T, Pringsheim T. The incidence of Parkinson’s disease: a systematic review and meta-analysis. Neuroepidemiology Karger Publishers. 2016;46:292–300.

    Article  Google Scholar 

  84. Tysnes OB, Storstein A. Epidemiology of Parkinson’s disease. J. Neural Transm. 2017.

  85. Scheperjans F, Aho V, Pereira PAB, Koskinen K, Paulin L, Pekkonen E, et al. Gut microbiota are related to Parkinson’s disease and clinical phenotype. Mov Disord. 2015;30:350–8.

    Article  Google Scholar 

  86. Bullich C, Keshavarzian A, Garssen J, Kraneveld A, Perez-Pardo P. Gut vibes in Parkinson’s disease: the microbiota-gut-brain axis. Mov Disord Clin Pract. 2019.

  87. Del Tredici K, Braak H. A not entirely benign procedure: progression of Parkinson’s disease. Acta Neuropathol. 2008.

  88. Lionnet A, Leclair-Visonneau L, Neunlist M, Murayama S, Takao M, Adler CH, et al. Does Parkinson’s disease start in the gut? Acta Neuropathol. 2018.

  89. Hill-Burns EM, Debelius JW, Morton JT, Wissemann WT, Lewis MR, Wallen ZD, et al. Parkinson’s disease and Parkinson’s disease medications have distinct signatures of the gut microbiome. Mov Disord. 2017;32:739–49.

    Article  CAS  Google Scholar 

  90. Petrov VA, Saltykova IV, Zhukova IA, Alifirova VM, Zhukova NG, Dorofeeva YB, et al. Analysis of gut microbiota in patients with Parkinson’s disease. Bull Exp Biol Med. 2017.

  91. Gerhardt S. Mohajeri MH. Nutrients: Changes of colonic bacterial composition in Parkinson’s disease and other neurodegenerative diseases; 2018.

    Google Scholar 

  92. • Tenkorang MA, Snyder B, Cunningham RL. Sex-related differences in oxidative stress and neurodegeneration. Steroids. 2018; This article delineates how sex differences in the generation of oxidative stress may underlie the sex differences in neurodegenerative disorders.

  93. Mariani E, Lombardini L, Facchin F, Pizzetti F, Frabetti F, Tarozzi A, et al. Sex-specific transcriptome differences in substantia nigra tissue: a meta-analysis of Parkinson’s disease data. Genes (Basel). 2018.

  94. • Siani F, Greco R, Levandis G, Ghezzi C, Daviddi F, Demartini C, et al. Influence of estrogen modulation on glia activation in a murine model of Parkinson’s disease. Front Neurosci. 2017; This original study, utilizing a murine model of PD, investigated the protective role of estrogen against nigrostriatal degeneration, demonstrating that estrogen treatments could even reverse neural degeneration.

  95. van Kessel SP, Frye AK, El-Gendy AO, Castejon M, Keshavarzian A, van Dijk G, et al. Gut bacterial tyrosine decarboxylases restrict levels of levodopa in the treatment of Parkinson’s disease. Nat Commun. 2019.

  96. Baumgart DC, Carding SR. Inflammatory bowel disease: cause and immunobiology. Lancet. 2007.

  97. Baumgart DC, Sandborn WJ. Inflammatory bowel disease: clinical aspects and established and evolving therapies. Lancet. 2007.

  98. Molodecky NA, Soon S, Rabi DM, Ghali WA, Ferris M, Chernoff G, et al. Increasing incidence and prevalence of the inflammatory bowel diseases with time, based on systematic review. Gastroenterology Elsevier. 2012;142:46–54.

    Article  Google Scholar 

  99. Shivashankar R, Tremaine WJ, Harmsen WS, Loftus Jr E V. Incidence and prevalence of Crohn’s disease and ulcerative colitis in Olmsted County, Minnesota from 1970 through 2010. Clin Gastroenterol Hepatol. Elsevier; 2017;15:857–863.

  100. Shah SC, Khalili H, Gower-Rousseau C, Olen O, Benchimol EI, Lynge E, et al. Sex-based differences in incidence of inflammatory bowel diseases—pooled analysis of population-based studies from Western countries. Gastroenterology. 2018.

  101. Neuendorf R, Harding A, Stello N, Hanes D, Wahbeh H. Depression and anxiety in patients with inflammatory bowel disease: a systematic review. J Psychosom Res. 2016.

  102. Fuller-Thomson E, Sulman J. Depression and inflammatory bowel disease: findings from two nationally representative Canadian surveys. Inflamm Bowel Dis. 2006.

  103. Panara AJ, Yarur AJ, Rieders B, Proksell S, Deshpande AR, Abreu MT, et al. The incidence and risk factors for developing depression after being diagnosed with inflammatory bowel disease: a cohort study. Aliment Pharmacol Ther. 2014.

  104. Mikocka-Walus A, Knowles SR, Keefer L, Graff L. Controversies revisited: a systematic review of the comorbidity of depression and anxiety with inflammatory bowel diseases. Inflamm Bowel Dis. Oxford University Press Oxford, UK; 2016;22:752–62.

  105. Brooks AJ, Rowse G, Ryder A, Peach EJ, Corfe BM, Lobo AJ. Systematic review: psychological morbidity in young people with inflammatory bowel disease–risk factors and impacts. Aliment Pharmacol Ther Wiley Online Library. 2016;44:3–15.

    Article  CAS  Google Scholar 

  106. Bernstein CN. Psychological stress and depression: risk factors for IBD? Dig Dis Karger Publishers. 2016;34:58–63.

    Article  Google Scholar 

  107. Mikocka-Walus A, Pittet V, Rossel JB, von Känel R. Symptoms of depression and anxiety are independently associated with clinical recurrence of inflammatory bowel disease. Clin Gastroenterol Hepatol. 2016.

  108. Blondel-Kucharski F, Chircop C, Marquis P, Cortot A, Baron F, Gendre JP, et al. Health-related quality of life in Crohn’s disease: a prospective longitudinal study in 231 patients. Am J Gastroenterol. 2001.

  109. Hauser G, Tkalcić M, Stimac D, Milić S, Sincić BM. Gender related differences in quality of life and affective status in patients with inflammatory bowel disease. Coll Antropol. 2011;

  110. Hoivik ML, Moum B, Solberg IC, Cvancarova M, Hoie O, Vatn MH, et al. Health-related quality of life in patients with ulcerative colitis after a 10-year disease course: results from the IBSEN study. Inflamm Bowel Dis. 2012.

Download references

Acknowledgments

Alexa Curhan Budavari and Katrina M. Rodriguez are supported by the National Institute of Mental Health’s Psychiatric Epidemiology Training Program (5T32MH014592-39; PI: Volk, Heather).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Calliope Holingue.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

This article is part of the Topical Collection on Sex and Gender Issues in Behavioral Health

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Holingue, C., Budavari, A.C., Rodriguez, K.M. et al. Sex Differences in the Gut-Brain Axis: Implications for Mental Health. Curr Psychiatry Rep 22, 83 (2020). https://doi.org/10.1007/s11920-020-01202-y

Download citation

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1007/s11920-020-01202-y

Keywords

Navigation