511
Views
1
CrossRef citations to date
0
Altmetric
Review

Does the gut microbiome mediate antipsychotic-induced metabolic side effects in schizophrenia?

ORCID Icon, ORCID Icon, ORCID Icon &
Pages 625-639 | Received 12 Oct 2021, Accepted 10 Feb 2022, Published online: 28 Feb 2022

References

  • American Psychiatric Association. Diagnostic and statistical manual of mental disorders (DSM-5®); Arlington, VA: American Psychiatric Pub, 2013.
  • Charlson FJ, Ferrari AJ, and Santomauro DF, et al. Global epidemiology and burden of schizophrenia: findings from the global burden of disease study 2016. Schizophr Bull 2018, 44 (6), 1195–1203.
  • Whiteford HA, Degenhardt L, and Rehm J, et al. Global burden of disease attributable to mental and substance use disorders: findings from the global burden of disease study 2010. Lancet 2013, 382 (9904), 1575–1586.
  • Lieberman JA, Rosenheck RA, and Davis SM, et al. Effectiveness of antipsychotic drugs in patients with chronic schizophrenia. N Engl J Med 2005, 353 (12), 1209–1223.
  • Siskind D, McCartney L, Goldschlager R, et al. First- and second–Generation antipsychotics in treatment–Refractory schizophrenia: systematic review and meta–Analysis. Br J Psychiatry 2016, 209 (5), 385–392.
  • Land R, Siskind D, McArdle P, et al. The impact of clozapine on hospital use: a systematic review and meta–Analysis. Acta Psychiatr Scand 2017, 135 (4), 296–309.
  • Vermeulen JM, van Rooijen G, van de Kerkhof MPJ, et al. Clozapine and long–Term mortality risk in patients with schizophrenia: a systematic review and meta–Analysis of studies lasting 1.1–12.5 years. Schizophr Bull 2019, 45 (2), 315–329.
  • Siskind D, Siskind V, Kisely S Clozapine response rates among people with treatment-resistant schizophrenia: data from a systematic review and meta–Analysis. Can J Psychiatry 2017, 62 (11), 772–777.
  • De Hert M, Dockx L, Bernagie C, et al. Prevalence and severity of antipsychotic related constipation in patients with schizophrenia: a retrospective descriptive study. BMC Gastroenterol 2011, 11 (1), 17.
  • De Hert MA, van Winkel R, Van Eyck D, et al. Prevalence of the metabolic syndrome in patients with schizophrenia treated with antipsychotic medication. Schizophr Res 2006, 83 (1), 87–93.
  • Marteene W, Winckel K, Hollingworth S, et al. Strategies to counter antipsychotic–Associated weight gain in patients with schizophrenia. Expert Opin Drug Saf 2019, 18 (12), 1149–1160.
  • McEvoy JP, Meyer JM, Goff DC, et al. Prevalence of the metabolic syndrome in patients with schizophrenia: baseline results from the Clinical Antipsychotic Trials of Intervention Effectiveness (CATIE) schizophrenia trial and comparison with national estimates from NHANES III. Schizophr Res 2005, 80 (1), 19–32.
  • Mitchell AJ, Vancampfort D, and Sweers K, et al. Prevalence of metabolic syndrome and metabolic abnormalities in schizophrenia and related disorders—A systematic review and meta–Analysis. Schizophr Bull 2013, 39 (2), 306–318.
  • Simpson CA, Diaz–Arteche C, Eliby D, et al. The gut microbiota in anxiety and depression - A systematic review. Clin Psychol Rev 2021, 83, 101943.
  • Lucidi L, Pettorruso M, Vellante F, et al. Gut microbiota and bipolar disorder: an overview on a novel biomarker for diagnosis and treatment. Int J Mol Sci 2021, 22 (7), 3723.
  • Bastiaanssen TFS, Cussotto S, Claesson MJ, et al. Unraveling the role of the microbiome in major depressive disorder. Harv Rev Psychiatry 2020, 28 (1), 26–39.
  • Schwarz E, Maukonen J, Hyytiäinen T, et al. Analysis of microbiota in first episode psychosis identifies preliminary associations with symptom severity and treatment response. Schizophr Res 2018, 192, 398–403.
  • Yuan X, Zhang P, Wang Y, et al. Changes in metabolism and microbiota after 24–Week risperidone treatment in drug naïve, normal weight patients with first episode schizophrenia. Schizophr Res 2018, 201, 299–306.
  • Zhang X, Pan L-Y, Zhang Z, et al. Analysis of gut mycobiota in first–Episode, drug-naïve Chinese patients with schizophrenia: a pilot study. Behav Brain Res 2020, 379, 112374.
  • Zhu F, Ju Y, Wang W, et al. Metagenome–Wide association of gut microbiome features for schizophrenia. Nat Commun 2020, 11 (1), 1612.
  • Bahr SM, Tyler BC, Wooldridge N, et al. Use of the second–Generation antipsychotic, risperidone, and secondary weight gain are associated with an altered gut microbiota in children. Transl Psychiatry 2015, 5 (10), e652–e652.
  • Flowers SA, Evans SJ, Ward KM, et al. Interaction between atypical antipsychotics and the gut microbiome in a bipolar disease cohort. Pharmacotherapy 2017, 37 (3), 261–267.
  • Zheng P, Zeng B, Liu M, et al. The gut microbiome from patients with schizophrenia modulates the glutamate–Glutamine-GABA cycle and schizophrenia–Relevant behaviors in mice. Sci Adv. 2019, 5 (2), eaau8317.
  • Davey KJ, Cotter PD, O’Sullivan O, et al. Antipsychotics and the gut microbiome: olanzapine–Induced metabolic dysfunction is attenuated by antibiotic administration in the rat. Transl Psychiatry 2013, 3 (10), e309–e309.
  • Morgan AP, Crowley JJ, Nonneman RJ, et al. The antipsychotic olanzapine interacts with the gut microbiome to cause weight gain in mouse. PLoS ONE 2014, 9 (12), e115225.
  • Divac N, Prostran M, Jakovcevski I, et al. Second–Generation antipsychotics and extrapyramidal adverse effects. Biomed Res Int 2014, 2014, 1–6.
  • Carlsson A Antipsychotic drugs, neurotransmitters, and schizophrenia. Ajp 1978, 135 (2), 164–173.
  • Dazzan P, Morgan KD, Orr K, et al. Different effects of typical and atypical antipsychotics on grey matter in first episode psychosis: the ÆSOP study. Neuropsychopharmacol 2005, 30 (4), 765–774.
  • Leucht S, Leucht C, Huhn M, et al. Sixty years of placebo–Controlled antipsychotic drug trials in acute schizophrenia: systematic review, bayesian meta–Analysis, and meta–Regression of efficacy predictors. Am J Psychiatry 2017, 174 (10), 927–942.
  • Correll CU, Solmi M, Veronese N, et al., Incidence and mortality from cardiovascular disease in patients with pooled and specific severe mental illness: a large–Scale meta–Analysis of 3,211,768 patients and 113,383,368 Controls. World Psychiatry 2017, 16 (2), 163–180.
  • Westman J, Eriksson SV, Gissler M, et al. Increased cardiovascular mortality in people with schizophrenia: a 24–Year national register study. Epidemiol Psychiatr Sci 2018, 27 (5), 519–527.
  • Lawrence D, Hancock KJ, Kisely S The gap in life expectancy from preventable physical illness in psychiatric patients in western australia: retrospective Analysis of population based registers. BMJ 2013, 346 (may21 1), f2539–f2539.
  • Bai Y-M, Chen J-Y, Chen -T-T, et al. Weight gain with clozapine: 8-Year cohort naturalistic study among hospitalized Chinese schizophrenia patients. Schizophr Res 2009, 108 (1–3), 122–126.
  • Henderson DC Clozapine: diabetes mellitus, weight gain, and lipid abnormalities. A Five-Year Natural;istic Study. J Clin Psychiatry. 2000, 157,975–981.
  • Kinon BJ, Kaiser CJ, Ahmed S, et al. Association between early and rapid weight gain and change in weight over one year of olanzapine therapy in patients with schizophrenia and related disorders. J Clin Psychopharmacol 2005, 25 (3), 255–258.
  • Rummel-Kluge C, Komossa K, and Schwarz S, et al. Head-to-Head comparisons of metabolic side effects of second generation antipsychotics in the treatment of schizophrenia: a systematic review and meta-Analysis. Schizophr Res 2010, 123 (2–3), 225–233.
  • Hirsch L, Yang J, Bresee L, et al. Second-Generation antipsychotics and metabolic side effects: a systematic review of population-Based studies. Drug Saf 2017, 40 (9), 771–781.
  • Mayfield K, Siskind D, Winckel K, et al. Glucagon-like peptide-1 agonists combating clozapine-Associated obesity and diabetes. J Psychopharmacol 2016, 30 (3), 227–236.
  • Salvi V, Mencacci C, Barone-Adesi F H1-Histamine receptor affinity predicts weight gain with antidepressants. Eur Neuropsychopharmacol 2016, 26 (10), 1673–1677.
  • Hong KS, Park T Searching susceptibility genes for antipsychotic-Induced weight gain: is the 5-HT2C receptor gene a promising candidate? Per Med 2007, 4 (3), 357–361.
  • Templeman LA, Reynolds GP, Arranz B, et al. Polymorphisms of the 5-HT2C receptor and leptin genes are associated with antipsychotic drug-Induced weight gain in caucasian subjects with a first-Episode psychosis. Pharmacogenet Genomics 2005, 15 (4), 195–200.
  • Baik J-H Dopamine signaling in food addiction: role of dopamine D2 receptors. BMB Rep 2013, 46 (11), 519–526.
  • Grajales D, Ferreira V, Valverde ÁM Second-Generation antipsychotics and dysregulation of glucose metabolism: beyond weight gain. Cells 2019, 8 (11), 1336.
  • Pillinger T, Beck K, Gobjila C, et al. Impaired glucose homeostasis in first-Episode schizophrenia: a systematic review and meta-Analysis. JAMA Psychiatry 2017, 74 (3), 261–269.
  • Firth J, Siddiqi N, Koyanagi A, et al. The lancet psychiatry commission: a blueprint for protecting physical health in people with mental illness. Lancet Psychiatry 2019, 6 (8), 675–712.
  • Duncan EJ, Woolson SL, Hamer RM, et al. Risk of lipid abnormality with haloperidol, olanzapine, quetiapine, and risperidone in a veterans affairs population. Int Clin Psychopharmacol 2009, 24 (4), 204–213.
  • Cryan JF, O’Riordan KJ, and Cowan CSM, et al. The microbiota-Gut-Brain axis. Physiol Rev 2019, 99 (4), 1877–2013.
  • Dabke K, Hendrick G, Devkota S The gut microbiome and metabolic syndrome. J Clin Investig 2019, 129 (10), 4050–4057.
  • Bliss ES, Whiteside E The gut-Brain axis, the human gut microbiota and their integration in the development of obesity. Front Physiol. 2018, 9, 900.
  • Lim YY, Lee YS, Ooi DSQ Engineering the gut microbiome for treatment of obesity: a review of current understanding and progress. Biotechnol J 2020, 15 (10), 2000013.
  • Lee CJ, Sears CL, Maruthur N Gut microbiome and its role in obesity and insulin resistance. Ann N Y Acad Sci 2020, 1461 (1), 37–52.
  • Cai M, Loy SL, Tan KH, et al. Association of elective and emergency cesarean delivery with early childhood overweight at 12 months of age. JAMA Network Open 2018, 1 (7), e185025.
  • Golofast B, Vales K The connection between microbiome and schizophrenia. Neurosci Biobehav Rev 2020, 108, 712–731.
  • Li H, Zhou Y, Liu J The impact of cesarean section on offspring overweight and obesity: a systematic review and meta-Analysis. Int J Obes 2013, 37 (7), 893–899.
  • Timmerman HM, Rutten NBMM, Boekhorst J, et al. Intestinal colonisation patterns in breastfed and formula-Fed infants during the first 12 weeks of life reveal sequential microbiota signatures. Sci Rep 2017, 7 (1), 8327.
  • Cong X, Judge M, Xu W, et al. Influence of feeding type on gut microbiome development in hospitalized preterm infants. Nurs Res 2017, 66 (2), 123–133.
  • David LA, Maurice CF, Carmody RN, et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature 2014, 505 (7484), 559–563.
  • Goodrich JK, Waters JL, Poole AC, et al. Human genetics shape the gut microbiome. Cell 2014, 159 (4), 789–799.
  • Ridaura VK, Faith JJ, Rey FE, et al. Gut microbiota from twins discordant for obesity modulate metabolism in mice. Science 2013, 341 (6150), 1241214.
  • Turnbaugh PJ, Ley RE, Mahowald MA, et al. An obesity-Associated gut microbiome with increased capacity for energy harvest. Nature 2006, 444 (7122), 1027–1031. https://doi.org/10.1038/nature05414.
  • The Human Microbiome Project Consortium. Structure, function and diversity of the healthy human microbiome. Nature 2012, 486 (7402), 207–214.
  • The human microbiome project consortium, A framework for human microbiome research. Nature 2012, 486 (7402), 215–221.
  • Sunjaya AP, Sunjaya AF, Kumala M Manipulating the gut microbiota to manage obesity. Adv Sci Lett 2018, 24 (8), 6198–6202.
  • Bäckhed F, Manchester JK, Semenkovich CF, et al. Mechanisms underlying the resistance to diet-Induced obesity in germ-Free mice. Pnas 2007, 104 (3), 979–984.
  • Backhed F, Ding H, Wang T, et al. The gut microbiota as an environmental factor that regulates fat storage. Proc Nat Acad Sci 2004, 101 (44), 15718–15723.
  • Kootte RS, Levin E, Salojärvi J, et al. Improvement of insulin sensitivity after lean donor feces in metabolic syndrome is driven by baseline intestinal microbiota composition. Cell Metab 2017, 26 (4), 611–619.e6.
  • Alang N, Kelly CR Weight gain after fecal microbiota transplantation. Open Forum Infect Dis 2015, 2 (1), ofv004.
  • Vrieze A, Van Nood E, Holleman F, et al. Transfer of intestinal microbiota from lean donors increases insulin sensitivity in individuals with metabolic syndrome. Gastroenterology 2012, 143 (4), 913–916.e7.
  • Yun Y, Kim H-N, Kim SE, et al. Comparative analysis of gut microbiota associated with body mass index in a large Korean cohort. BMC Microbiol 2017, 17 (1), 151.
  • Misiak B, Łoniewski I, Marlicz W, et al. The HPA axis dysregulation in severe mental illness: can we shift the blame to gut microbiota? Prog Neuro Psychopharmacol Biol Psychiatry 2020, 102, 109951.
  • Chen Y, Xu J, Chen Y Regulation of neurotransmitters by the gut microbiota and effects on cognition in neurological disorders. Nutrients 2021, 13 (6), 2099. https://doi.org/10.3390/nu13062099.
  • Shpakov AO, Derkach KV, Berstein LM Brain signaling systems in the type 2 diabetes and metabolic syndrome: promising target to treat and prevent these diseases. Future Sci OA 2015, 1 (3), FSO25. https://doi.org/10.4155/fso.15.23.
  • Chen X, Devaraj S Gut microbiome in obesity, metabolic syndrome, and diabetes. Curr Diab Rep 2018, 18 (12), 129.
  • Bonaz B, Bazin T, Pellissier S The vagus nerve at the interface of the microbiota-Gut-Brain axis. Front Neurosci. 2018, 12, 49.
  • Perry RJ, Peng L, Barry NA, et al. Acetate mediates a microbiome–Brain–β-Cell axis to promote metabolic syndrome. Nature 2016, 534 (7606), 213–217.
  • Frost G, Sleeth ML, Sahuri-Arisoylu M, et al. The short-Chain fatty acid acetate reduces appetite via a central homeostatic mechanism. Nat Commun 2014, 5 (1), 3611.
  • Hills R, Pontefract B, Mishcon H, et al. Gut microbiome: profound implications for diet and disease. Nutrients 2019, 11 (7), 1613.
  • Sharma S, Tripathi P Gut microbiome and type 2 diabetes: where we are and where to go? J Nutr Biochem 2019, 63, 101–108.
  • Zhao Y, Wang Z Gut microbiome and cardiovascular disease. Curr Opin Cardiol 2020, 35 (3), 207–218.
  • Zhao Q, Wang X, Hu Q, et al. Suppression of TLR4 by MiR-448 is involved in diabetic development via regulating macrophage polarization. J Pharm Pharmacol 2019, 71 (5), 806–815.
  • Vallianou NG, Stratigou T, Tsagarakis S Microbiome and diabetes: where are we now? Diabetes Res Clin Pract 2018, 146, 111–118.
  • Menon D, Coll R, O’Neill LAJ, et al. GSTO1-1 modulates metabolism in macrophages activated through the LPS and TLR4 pathway. J Cell Sci 2015, 128 (10), 1982–1990.
  • Zuany-Amorim C, Hastewell J, Walker C Toll-like receptors as potential therapeutic targets for multiple diseases. Nat Rev Drug Discov 2002, 1 (10), 797–807.
  • Marrie RA, Walld R, Bolton JM, et al. Increased incidence of psychiatric disorders in immune-Mediated INFLAMMATORY DISEAse. J Psychosom Res 2017, 101, 17–23.
  • Nowakowski J, Chrobak AA, Dudek D Psychiatric illnesses in inflammatory bowel diseases - Psychiatric comorbidity and biological underpinnings. Psychiatr Pol 2016, 50 (6), 1157–1166.
  • Yu LC-H Microbiota dysbiosis and barrier dysfunction in inflammatory bowel disease and colorectal cancers: exploring a common ground hypothesis. J Biomed Sci 2018, 25 (1), 79.
  • Ma W, Nguyen LH, Song M, et al. Dietary fiber intake, the gut microbiome, and chronic systemic inflammation in a cohort of adult men. Genome Med 2021, 13 (1), 102.
  • Chen Y, Meng P, Cheng S, et al. Assessing the effect of interaction between C-Reactive protein and gut microbiome on the risks of anxiety and depression. Mol Brain 2021, 14 (1), 133.
  • Orsolini L, Sarchione F, Vellante F, et al. Protein-C reactive as biomarker predictor of schizophrenia phases of illness? A systematic review. Curr Neuropharmacol 2018, 16 (5), 583–606.
  • Ullah I, Awan HA, Aamir A, et al. Role and perspectives of inflammation and C-Reactive protein (CRP) in psychosis: an economic and Widespread Tool for Assessing the Disease. Int J Mol Sci 2021, 22 (23), 13032. https://doi.org/10.3390/ijms222313032.
  • Lontchi-Yimagou E, Sobngwi E, Matsha TE, et al. Diabetes mellitus and inflammation. Curr Diab Rep 2013, 13 (3), 435–444. https://doi.org/10.1007/s11892-013-0375-y.
  • Pirola L, Ferraz JC Role of pro- and anti-Inflammatory phenomena in the physiopathology of type 2 diabetes and obesity. Wjbc 2017, 8 (2), 120.
  • Keane KN, Calton EK, Carlessi R, et al. The bioenergetics of inflammation: insights into obesity and type 2 diabetes. Eur J Clin Nutr 2017, 71 (7), 904–912.
  • Sen T, Cawthon CR, Ihde BT, et al. Diet-Driven microbiota dysbiosis is associated with vagal remodeling and obesity. Physiol Behav 2017, 173, 305–317.
  • Larsen N, Vogensen FK, van den Berg FWJ, et al. Gut microbiota in human adults with type 2 diabetes differs from non-Diabetic adults. PLoS ONE 2010, 5 (2), e9085.
  • Tirosh A, Calay ES, Tuncman G, et al. The short-Chain fatty acid propionate increases glucagon and FABP4 production, impairing insulin action in mice and humans. Sci Transl Med. 2019, 11 (489), eaav0120.
  • De Vadder F, Kovatcheva-Datchary P, Goncalves D, et al. Microbiota-Generated metabolites promote metabolic benefits via gut-Brain neural circuits. Cell 2014, 156 (1–2), 84–96.
  • Wang Z, Zhao Y Gut microbiota derived metabolites in cardiovascular health and disease. Protein Cell 2018, 9 (5), 416–431.
  • Koeth RA, Wang Z, Levison BS, et al. Intestinal microbiota metabolism of L-Carnitine, a nutrient in red meat, promotes atherosclerosis. Nat Med 2013, 19 (5), 576–585.
  • Wang Z, Klipfell E, Bennett BJ, et al. Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease. Nature 2011, 472 (7341), 57–63.
  • Gregory JC, Buffa JA, Org E, et al. Transmission of atherosclerosis susceptibility with gut microbial transplantation. J Biol Chem 2015, 290 (9), 5647–5660.
  • Zhu W, Gregory JC, Org E, et al. Gut microbial metabolite tmao enhances platelet hyperreactivity and thrombosis risk. Cell 2016, 165 (1), 111–124. https://doi.org/10.1016/j.cell.2016.02.011.
  • Randrianarisoa E, Lehn-Stefan A, Wang X, et al. Relationship of serum trimethylamine N-Oxide (TMAO) levels with early atherosclerosis in humans. Sci Rep 2016, 6 (1), 26745.
  • Senthong V, Wang Z, Li XS, et al. Intestinal Microbiota-Generated metabolite trimethylamine- N- Oxide and 5-Year mortality risk in stable coronary artery disease: the contributory role of intestinal microbiota in a COURAGE-Like patient cohort. journal of the American Heart Association 2016, 5 (6). 6
  • Cao X-S, Chen J, Zou J-Z, et al. Association of indoxyl sulfate with heart failure among patients on hemodialysis. clinical Journal of the American Society of Nephrology 2015, 10 (1), 111–119.
  • Fan P-C, Chang JC-H, Lin C-N, et al. Serum indoxyl sulfate predicts adverse cardiovascular events in patients with chronic kidney disease. J Formosan Med Assoc 2019, 118 (7), 1099–1106.
  • Kim HY, Yoo T-H, Hwang Y, et al. Indoxyl sulfate (IS)-mediated immune dysfunction provokes endothelial damage in patients with End-Stage Renal Disease (ESRD). Sci Rep 2017, 7 (1), 3057.
  • Yang K, Du C, Wang X, et al. Indoxyl Sulfate induces platelet hyperactivity and contributes to chronic kidney disease–Associated thrombosis in mice. Blood 2017, 129 (19), 2667–2679.
  • Wang C-H, Cheng M-L, Liu M-H, et al. Increased P-cresyl sulfate level is independently associated with poor outcomes in patients with heart failure. Heart and Vessels 2016, 31 (7), 1100–1108.
  • Lin C-J, Chuang C-K, Jayakumar T, et al. Serum p -cresyl sulfate predicts cardiovascular disease and mortality in elderly hemodialysis patients. archives of Medical Science 2013, 4, 662–668.
  • Gryp T, Vanholder R, Vaneechoutte M, et al. P-Cresyl sulfate. Toxins (Basel) 2017, 9 (2), 52.
  • Han H, Zhu J, Zhu Z, et al. p -Cresyl sulfate aggravates cardiac dysfunction associated with chronic kidney disease by enhancing apoptosis of cardiomyocytes. journal of the American Heart Association 2015, 4 (6). 6
  • Brown CN, Fultz EK, Ferdousian S, et al. Transgenic analyses of homer2 function within nucleus accumbens subregions in the regulation of methamphetamine reward and reinforcement in mice. Front Psychiatry 2020, 11. 11
  • Tuomisto H A diversity of beta diversities: straightening up a concept gone awry. Part 1. Defining beta diversity as a function of alpha and gamma diversity. Ecography 2010, 33 (1), 2–22.
  • Rinninella E, Raoul P, Cintoni M, et al. What is the healthy gut microbiota composition? A changing ecosystem across age, environment, diet, and diseases. Microorganisms 2019, 7 (1), 14.
  • Ma X, Asif H, Dai L, et al. Alteration of the gut microbiome in first-Episode drug-Naïve and chronic medicated schizophrenia correlate with regional brain volumes. J Psychiatr Res 2020, 123, 136–144.
  • Xu R, Wu B, Liang J, et al. Altered gut microbiota and mucosal immunity in patients with schizophrenia. Brain Behav Immun 2020, 85, 120–127.
  • Nguyen TT Differences in gut microbiome composition between persons with chronic schizophrenia and healthy comparison subjects. Schizophr Res 2019 204 , 23–29.
  • Shen Y, Xu J, Li Z, et al. Analysis of gut microbiota diversity and auxiliary diagnosis as a biomarker in patients with schizophrenia: a cross-Sectional study. Schizophr Res 2018, 197, 470–477.
  • Rizzatti G, Lopetuso LR, Gibiino G, et al. Proteobacteria: a common factor in human diseases. Biomed Res Int 2017, 2017, 1–7.
  • Shin N-R, Whon TW, Bae J-W Proteobacteria: microbial signature of dysbiosis in gut microbiota. Trends Biotechnol 2015, 33 (9), 496–503.
  • He Y, Kosciolek T, Tang J, et al. Gut microbiome and magnetic resonance spectroscopy study of subjects at ultra-High risk for psychosis may support the membrane hypothesis. Eur Psychiatry 2018, 53, 37–45.
  • Bahr SM, Weidemann BJ, Castro AN, et al. Risperidone-Induced weight gain is mediated through shifts in the gut microbiome and suppression of energy expenditure. EBioMedicine 2015, 2 (11), 1725–1734.
  • Davey KJ, O’Mahony SM, Schellekens H, et al. Gender-dependent consequences of chronic olanzapine in the rat: effects on body weight, inflammatory, metabolic and microbiota parameters. Psychopharmacology (Berl) 2012, 221 (1), 155–169.
  • Bobes J, Rejas J, Garcia-Garcia M, et al. Weight gain in patients with schizophrenia treated with risperidone, olanzapine, quetiapine or haloperidol: results of the EIRE study. Schizophr Res 2003, 62 (1–2), 77–88.
  • Gautam S, Jain S, Bhargava M Weight gain with olanzapine: drug, gender or age? Indian J Psychiatry 2006, 48 (1), 39–42.
  • Jones CA, Watson DJG, Fone KCF Animal models of schizophrenia. Br J Pharmacol 2011, 164 (4), 1162–1194.
  • Zhu F, Guo R, Wang W, et al. Transplantation of microbiota from drug-Free patients with schizophrenia causes schizophrenia-like abnormal behaviors and dysregulated kynurenine metabolism in mice. Mol Psychiatry 2020, 25 (11), 2905–2918.
  • Sarkar A, Lehto SM, Harty S, et al. Psychobiotics and the manipulation of bacteria–Gut–Brain signals. Trends Neurosci 2016, 39 (11), 763–781. https://doi.org/10.1016/j.tins.2016.09.002.
  • Dickerson FB, Stallings C, Origoni A, et al. Effect of probiotic supplementation on schizophrenia symptoms and association with gastrointestinal functioning: a randomized, placebo-Controlled trial. Prim Care Companion CNS Disord 2014, 16 (1).
  • Severance EG, Gressitt KL, Stallings CR, et al. Probiotic normalization of candida albicans in schizophrenia: a randomized, placebo-Controlled, longitudinal pilot study. Brain Behav Immun 2017, 62, 41–45.
  • Tomasik J, Yolken RH, Bahn S, et al. Immunomodulatory effects of probiotic supplementation in schizophrenia patients: a randomized, placebo-Controlled trial. BiomarkInsights 2015, 10, BMI.S22007.
  • Flowers SA, Baxter NT, Ward KM, et al. Effects of atypical antipsychotic treatment and resistant starch supplementation on gut microbiome composition in a cohort of patients with bipolar disorder or schizophrenia. Pharmacotherapy 2019, 39 (2), 161–170.
  • Okubo R, Koga M, Katsumata N, et al. Effect of bifidobacterium breve A-1 on anxiety and depressive symptoms in schizophrenia: a proof-of-Concept study. J Affect Disord 2019, 245, 377–385.
  • Ghaderi A, Banafshe HR, Mirhosseini N, et al. Clinical and metabolic response to vitamin d plus probiotic in schizophrenia patients. BMC Psychiatry 2019, 19 (1), 77.
  • Yang Y, Long Y, Kang D, et al. Effect of bifidobacterium on olanzapine-Induced body weight and appetite changes in patients with psychosis. Psychopharmacology (Berl) 2021. https://doi.org/10.1007/s00213-021-05866-z.
  • Kao A-C-C, Spitzer S, Anthony DC, et al. Prebiotic attenuation of olanzapine-Induced weight gain in rats: analysis of central and peripheral biomarkers and gut microbiota. Transl Psychiatry 2018, 8 (1), 1–12.
  • Vulevic J, Tzortzis G, Juric A, et al. Effect of a prebiotic galactooligosaccharide mixture (B-GOS®) on gastrointestinal symptoms in adults selected from a general population who suffer with bloating, abdominal pain, or flatulence. Neurogastroenterol Motility 2018, 30 (11), e13440.
  • Huang D, Gao J, Li C, et al. A potential probiotic bacterium for antipsychotic-Induced metabolic syndrome: mechanisms underpinning how akkermansia muciniphila subtype improves olanzapine-Induced glucose homeostasis in mice. Psychopharmacology (Berl) 2021. https://doi.org/10.1007/s00213-021-05878-9.
  • Depommier C, Everard A, Druart C, et al. Supplementation with akkermansia muciniphila in overweight and obese human volunteers: a proof-of-Concept exploratory study. Nat Med 2019, 25 (7), 1096–1103.
  • O’Mahony SM, Clarke G, Borre YE, et al., Tryptophan metabolism and the brain-Gut-Microbiome axis. Behav Brain Res 2015, 277, 32–48.
  • Siskind D, Gallagher E, Winckel K, et al. Does switching antipsychotics ameliorate weight gain in patients with severe mental illness? A systematic review and meta-Analysis. Schizophr Bull 2021, 47 (4), 948–958.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.