Modulation of Gut Microbial Diversity through Non-Pharmaceutical Approaches to Treat Schizophrenia
Abstract
:1. Introduction
2. Methodology
3. A Tridimensional Relationship of Gut Microbial Diversity, Neurotransmitters, and Schizophrenia
Gut Microorganisms | Role Associated with Neurotransmitter Regulation | References |
---|---|---|
Campylobacter jejuni | Affect glutamate synthesis and its metabolism | [15] |
Brevibacterium lactofermentum, Bacillus subtilis, Corynebacterium glutamicum, and Brevibacterium avium | Involved in conversion of L-glutamate into D-glutamate through glutamate racemase enzyme, thus disturbs glutamate metabolism | [15] |
Streptococcus vestibularis | Involved in synthesis and degradation of several types of neurotransmitters related to glutamate synthesis, GABA degradation, and isovaleric acid synthesis | [18] |
Erysipelotrichales, Bacteroidales, and Clostridiales | Affect glutamate activity through metabotropic glutamate receptors (mGluRs) | [19] |
Lactobacillus and Bifidobacterium species | Regulate GABA | [25] |
Bacillus and Serratia | Control dopamine | [26] |
Saccharomyces, Escherichia coli, and Bacillus | Regulate norepinephrine | [27] |
Candida, Escherichia, Streptococcus, and Enterococcus species | Regulate serotonin production | [27] |
4. Antipsychotics-Induced Side Effects and Gut Microbiota Dysbiosis in Schizophrenia
5. Gut Microbial Diversity Management Strategies to Treat Schizophrenia
5.1. Probiotics: Impact of Live Biotherapeutics on Disease Symptoms
5.2. Prebiotics: A Connection between Dietary Fiber, Gut Microbiota, and Schizophrenia
5.3. Polyphenols: A Potential Interplay between Polyphenols, Gut Microbiota, and Schizophrenia
6. Conclusions and Future Perspectives
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Ordieres, M.G.L. Schizophrenia: A Complex Mental Illness. In Psychiatry and Neuroscience Update; Springer: Cham, Switzerland, 2019; pp. 417–426. [Google Scholar]
- Kahn, R.S.; Sommer, I.E.; Murray, R.M.; Meyer-Lindenberg, A.; Weinberger, D.R.; Cannon, T.D.; O’Donovan, M.; Correll, C.U.; Kane, J.M.; Van Os, J. Schizophrenia. Nat. Rev. Dis. Primers 2015, 1, 1–23. [Google Scholar] [CrossRef] [PubMed]
- Correll, C.U.; Schooler, N.R. Negative symptoms in schizophrenia: A review and clinical guide for recognition, assessment, and treatment. Neuropsychiatr. Dis. Treat. 2020, 16, 519. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sommer, I.E.; Tiihonen, J.; van Mourik, A.; Tanskanen, A.; Taipale, H. The clinical course of schizophrenia in women and men—A nation-wide cohort study. NPJ Schizophr. 2020, 6, 1–7. [Google Scholar] [CrossRef] [PubMed]
- Buckley, P.F.; Miller, B.J.; Lehrer, D.S.; Castle, D.J. Psychiatric comorbidities and schizophrenia. Schizophr. Bull. 2009, 35, 383–402. [Google Scholar] [CrossRef] [Green Version]
- Olfson, M.; Gerhard, T.; Huang, C.; Crystal, S.; Stroup, T.S. Premature mortality among adults with schizophrenia in the united states. JAMA Psychiatry 2015, 72, 1172–1181. [Google Scholar] [CrossRef] [Green Version]
- Sawa, A.; Snyder, S.H. Schizophrenia: Diverse approaches to a complex disease. Science 2002, 296, 692–695. [Google Scholar] [CrossRef] [Green Version]
- Chow, J.; Lee, S.M.; Shen, Y.; Khosravi, A.; Mazmanian, S.K. Host–bacterial symbiosis in health and disease. Adv. Immunol. 2010, 107, 243–274. [Google Scholar]
- Rothschild, D.; Weissbrod, O.; Barkan, E.; Kurilshikov, A.; Korem, T.; Zeevi, D.; Costea, P.I.; Godneva, A.; Kalka, I.N.; Bar, N. Environment dominates over host genetics in shaping human gut microbiota. Nature 2018, 555, 210–215. [Google Scholar] [CrossRef]
- Yuan, X.; Kang, Y.; Zhuo, C.; Huang, X.-F.; Song, X. The gut microbiota promotes the pathogenesis of schizophrenia via multiple pathways. Biochem. Biophys. Res. Commun. 2019, 512, 373–380. [Google Scholar] [CrossRef]
- Schwarz, E.; Maukonen, J.; Hyytiäinen, T.; Kieseppä, T.; Orešič, M.; Sabunciyan, S.; Mantere, O.; Saarela, M.; Yolken, R.; Suvisaari, J. Analysis of microbiota in first episode psychosis identifies preliminary associations with symptom severity and treatment response. Schizophr. Res. 2018, 192, 398. [Google Scholar] [CrossRef] [Green Version]
- Yuan, X.; Wang, Y.; Li, X.; Jiang, J.; Kang, Y.; Pang, L.; Zhang, P.; Li, A.; Lv, L.; Andreassen, A.O.; et al. Gut microbial biomarkers for the treatment response in first-episode, drug-naive schizophrenia: A 24-week follow-up study. Transl. Psychiatry 2021, 11, 422. [Google Scholar] [CrossRef]
- Moher, D.; Liberati, A.; Tetzlaff, J.; Altman, D.G.; The PRISMA Group. Preferred reporting items for systematic reviews and meta-analyses: The PRISMA statement. PLoS Med. 2009, 6, e1000097. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Insel, T.R. Rethinking schizophrenia. Nature 2010, 468, 187–193. [Google Scholar] [CrossRef] [Green Version]
- Coyle, J.T. Glutamate and schizophrenia: Beyond the dopamine hypothesis. Cell. Mol. Neurobiol. 2006, 26, 363–382. [Google Scholar] [CrossRef]
- Munawar, N.; Ahsan, K.; Muhammad, K.; Ahmad, A.; Anwar, M.A.; Shah, I.; Al Ameri, A.K.; Al Mughairbi, F. Hidden role of gut microbiome dysbiosis in schizophrenia: Antipsychotics or psychobiotics as therapeutics? Int. J. Mol. Sci. 2021, 22, 7671. [Google Scholar] [CrossRef] [PubMed]
- Strandwitz, P. Neurotransmitter modulation by the gut microbiota. Brain Res. 2018, 1693, 128–133. [Google Scholar] [CrossRef]
- Chang, C.H.; Lin, C.H.; Lane, H.Y. D-glutamate and gut microbiota in alzheimer’s disease. Int. J. Mol. Sci. 2020, 21, 2676. [Google Scholar] [CrossRef] [Green Version]
- Zheng, P.; Zeng, B.; Liu, M.; Chen, J.; Pan, J.; Han, Y.; Liu, Y.; Cheng, K.; Zhou, C.; Wang, H. The gut microbiome from patients with schizophrenia modulates the glutamate-glutamine-GABA cycle and schizophrenia-relevant behaviors in mice. Sci. Adv. 2019, 5, eaau8317. [Google Scholar] [CrossRef] [Green Version]
- Zhu, F.; Guo, R.; Wang, W.; Ju, Y.; Wang, Q.; Ma, Q.; Sun, Q.; Fan, Y.; Xie, Y.; Yang, Z. Transplantation of microbiota from drug-free patients with schizophrenia causes schizophrenia-like abnormal behaviors and dysregulated kynurenine metabolism in mice. Mol. Psychiatry 2020, 25, 2905–2918. [Google Scholar] [CrossRef] [PubMed]
- Gubert, C.; Kong, G.; Uzungil, V.; Zeleznikow-Johnston, A.M.; Burrows, E.L.; Renoir, T.; Hannan, A.J. Microbiome profiling reveals gut dysbiosis in the metabotropic glutamate receptor 5 knockout mouse model of schizophrenia. Front. Cell Dev. Biol. 2020, 8, 1233. [Google Scholar] [CrossRef]
- Jameson, K.; Olson, C.; Kazmi, S.; Hsiao, E. Toward understanding microbiome-neuronal signaling. Mol. Cell 2020, 78, 577–583. [Google Scholar] [CrossRef]
- Chen, Y.; Xu, J.; Chen, Y. Regulation of neurotransmitters by the gut microbiota and effects on cognition in neurological disorders. Nutrients 2021, 13, 2099. [Google Scholar] [CrossRef]
- Stępnicki, P.; Kondej, M.; Kaczor, A.A. Current concepts and treatments of schizophrenia. Molecules 2018, 23, 2087. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- O’Mahony, S.M.; Clarke, G.; Borre, Y.; Dinan, T.; Cryan, J. Serotonin, tryptophan metabolism and the brain-gut-microbiome axis. Behav. Brain Res. 2015, 277, 32–48. [Google Scholar] [CrossRef]
- Desbonnet, L.; Garrett, L.; Clarke, G.; Bienenstock, J.; Dinan, T.G. The probiotic bifidobacteria infantis: An assessment of potential antidepressant properties in the rat. J. Psychiatr. Res. 2008, 43, 164–174. [Google Scholar] [CrossRef]
- Barrett, E.; Ross, R.; O’Toole, P.W.; Fitzgerald, G.F.; Stanton, C. Γ-aminobutyric acid production by culturable bacteria from the human intestine. J. Appl. Microbiol. 2012, 113, 411–417. [Google Scholar] [CrossRef]
- Lyte, M. Probiotics function mechanistically as delivery vehicles for neuroactive compounds: Microbial endocrinology in the design and use of probiotics. Bioessays 2011, 33, 574–581. [Google Scholar] [CrossRef]
- Golofast, B.; Vales, K. The connection between microbiome and schizophrenia. Neurosci. Biobehav. Rev. 2020, 108, 712–731. [Google Scholar] [CrossRef]
- Clarke, G.; Grenham, S.; Scully, P.; Fitzgerald, P.; Moloney, R.; Shanahan, F.; Dinan, T.; Cryan, J. The microbiome-gut-brain axis during early life regulates the hippocampal serotonergic system in a sex-dependent manner. Mol. Psychiatry 2013, 18, 666–673. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kelly, J.R.; Kennedy, P.J.; Cryan, J.F.; Dinan, T.G.; Clarke, G.; Hyland, N.P. Breaking down the barriers: The gut microbiome, intestinal permeability and stress-related psychiatric disorders. Front. Cell. Neurosci. 2015, 9, 392. [Google Scholar] [CrossRef] [Green Version]
- Desbonnet, L.; Clarke, G.; Traplin, A.; O’Sullivan, O.; Crispie, F.; Moloney, R.D.; Cotter, P.D.; Dinan, T.G.; Cryan, J.F. Gut microbiota depletion from early adolescence in mice: Implications for brain and behaviour. Brain Behav. Immun. 2015, 48, 165–173. [Google Scholar] [CrossRef]
- Rackers, H.S.; Thomas, S.; Williamson, K.; Posey, R.; Kimmel, M.C. Emerging literature in the microbiota-brain axis and perinatal mood and anxiety disorders. Psychoneuroendocrinology 2018, 95, 86–96. [Google Scholar] [CrossRef]
- Coyle, J.T. Nmda receptor and schizophrenia: A brief history. Schizophr. Bull. 2012, 38, 920–926. [Google Scholar] [CrossRef] [Green Version]
- Krystal, J.H.; Karper, L.P.; Seibyl, J.P.; Freeman, G.K.; Delaney, R.; Bremner, J.D.; Heninger, G.R.; Bowers, M.B.; Charney, D.S. Subanesthetic effects of the noncompetitive NMDA antagonist, ketamine, in humans: Psychotomimetic, perceptual, cognitive, and neuroendocrine responses. Arch. Gen. Psychiatry 1994, 51, 199–214. [Google Scholar] [CrossRef] [PubMed]
- Strube, W.; Marshall, L.; Quattrocchi, G.; Little, S.; Cimpianu, C.L.; Ulbrich, M.; Schneider-Axmann, T.; Falkai, P.; Hasan, A.; Bestmann, S. Glutamatergic contribution to probabilistic reasoning and jumping to conclusions in schizophrenia: A double-blind, randomized experimental trial. Biol. Psychiatry 2020, 88, 687–697. [Google Scholar] [CrossRef]
- Szczurowska, E.; Ahuja, N.; Jiruška, P.; Kelemen, E.; Stuchlík, A. Impairment of neural coordination in hippocampal neuronal ensembles after a psychotomimetic dose of dizocilpine. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2018, 81, 275–283. [Google Scholar] [CrossRef] [PubMed]
- Svoboda, J.; Stankova, A.; Entlerova, M.; Stuchlik, A. Acute administration of mk-801 in an animal model of psychosis in rats interferes with cognitively demanding forms of behavioral flexibility on a rotating arena. Front. Behav. Neurosci. 2015, 9, 75. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kubík, Š.; Buchtová, H.; Valeš, K.; Stuchlík, A. Mk-801 impairs cognitive coordination on a rotating arena (carousel) and contextual specificity of hippocampal immediate-early gene expression in a rat model of psychosis. Front. Behav. Neurosci. 2014, 8, 75. [Google Scholar] [CrossRef] [Green Version]
- Maqsood, R.; Stone, T.W. The gut-brain axis, BDNF, NMDA and CNS disorders. Neurochem. Res. 2016, 41, 2819–2835. [Google Scholar] [CrossRef]
- Nieto, R.; Kukuljan, M.; Silva, H. BDNF and schizophrenia: From neurodevelopment to neuronal plasticity, learning, and memory. Front. Psychiatry 2013, 4, 45. [Google Scholar] [CrossRef] [Green Version]
- Herken, J.; Bang, C.; Rühlemann, M.C.; Finke, C.; Klag, J.; Franke, A.; Prüss, H. Normal gut microbiome in nmda receptor encephalitis. Neurol.-Neuroimmunol. Neuroinflamm. 2019, 6, e632. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Breit, S.; Kupferberg, A.; Rogler, G.; Hasler, G. Vagus nerve as modulator of the brain–gut axis in psychiatric and inflammatory disorders. Front. Psychiatry 2018, 9, 44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fried, S.; Wemelle, E.; Cani, P.D.; Knauf, C. Interactions between the microbiota and enteric nervous system during gut-brain disorders. Neuropharmacology 2021, 197, 108721. [Google Scholar] [CrossRef]
- van der Esch, C.C.; Kloosterboer, S.M.; van der Ende, J.; Reichart, C.G.; Kouijzer, M.E.; de Kroon, M.M.; van Daalen, E.; Ester, W.A.; Rieken, R.; Dieleman, G.C. Risk factors and pattern of weight gain in youths using antipsychotic drugs. Eur. Child Adolesc. Psychiatry 2021, 30, 1263–1271. [Google Scholar] [CrossRef]
- Chen, J.; Huang, X.-F.; Shao, R.; Chen, C.; Deng, C. Molecular mechanisms of antipsychotic drug-induced diabetes. Front. Neurosci. 2017, 11, 643. [Google Scholar] [CrossRef] [PubMed]
- Chen, C.-H.; Shyue, S.-K.; Hsu, C.-P.; Lee, T.-S. Atypical antipsychotic drug olanzapine deregulates hepatic lipid metabolism and aortic inflammation and aggravates atherosclerosis. Cell. Physiol. Biochem. 2018, 50, 1216–1229. [Google Scholar] [CrossRef]
- Zhang, Y.; Liu, Y.; Su, Y.; You, Y.; Ma, Y.; Yang, G.; Song, Y.; Liu, X.; Wang, M.; Zhang, L. The metabolic side effects of 12 antipsychotic drugs used for the treatment of schizophrenia on glucose: A network meta-analysis. BMC Psychiatry 2017, 17, 373. [Google Scholar] [CrossRef] [Green Version]
- Kraeuter, A.-K.; Phillips, R.; Sarnyai, Z. The gut microbiome in psychosis from mice to men: A systematic review of preclinical and clinical studies. Front. Psychiatry 2020, 11, 799. [Google Scholar] [CrossRef]
- Gao, K.; Pi, Y.; Mu, C.L.; Peng, Y.; Huang, Z.; Zhu, W.Y. Antibiotics-induced modulation of large intestinal microbiota altered aromatic amino acid profile and expression of neurotransmitters in the hypothalamus of piglets. J. Neurochem. 2018, 146, 219–234. [Google Scholar] [CrossRef] [Green Version]
- Gao, K.; Pi, Y.; Mu, C.L.; Farzi, A.; Liu, Z.; Zhu, W.Y. Increasing carbohydrate availability in the hindgut promotes hypothalamic neurotransmitter synthesis: Aromatic amino acids linking the microbiota–brain axis. J. Neurochem. 2019, 149, 641–659. [Google Scholar] [CrossRef]
- Marques, C.; Meireles, M.; Faria, A.; Calhau, C. High-fat diet–induced dysbiosis as a cause of neuroinflammation. Biol. Psychiatry 2016, 80, e3–e4. [Google Scholar] [CrossRef] [PubMed]
- Davey, K.; Cotter, P.; O’sullivan, O.; Crispie, F.; Dinan, T.; Cryan, J.; O’Mahony, S. Antipsychotics and the gut microbiome: Olanzapine-induced metabolic dysfunction is attenuated by antibiotic administration in the rat. Transl. Psychiatry 2013, 3, e309. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Macedo, D.; Chaves Filho, A.J.M.; de Sousa, C.N.S.; Quevedo, J.; Barichello, T.; Júnior, H.V.N.; de Lucena, D.F. Antidepressants, antimicrobials or both? Gut microbiota dysbiosis in depression and possible implications of the antimicrobial effects of antidepressant drugs for antidepressant effectiveness. J. Affect. Disord. 2017, 208, 22–32. [Google Scholar] [CrossRef] [PubMed]
- Maier, L.; Pruteanu, M.; Kuhn, M.; Zeller, G.; Telzerow, A.; Anderson, E.E.; Brochado, A.R.; Fernandez, K.C.; Dose, H.; Mori, H. Extensive impact of non-antibiotic drugs on human gut bacteria. Nature 2018, 555, 623–628. [Google Scholar] [CrossRef]
- Manchia, M.; Fontana, A.; Panebianco, C.; Paribello, P.; Arzedi, C.; Cossu, E.; Garzilli, M.; Montis, M.A.; Mura, A.; Pisanu, C. Involvement of gut microbiota in schizophrenia and treatment resistance to antipsychotics. Biomedicines 2021, 9, 875. [Google Scholar] [CrossRef]
- Nguyen, T.T.; Kosciolek, T.; Maldonado, Y.; Daly, R.E.; Martin, A.S.; McDonald, D.; Knight, R.; Jeste, D.V. Differences in gut microbiome composition between persons with chronic schizophrenia and healthy comparison subjects. Schizophr. Res. 2019, 204, 23–29. [Google Scholar] [CrossRef]
- Zhu, F.; Ju, Y.; Wang, W.; Wang, Q.; Guo, R.; Ma, Q.; Sun, Q.; Fan, Y.; Xie, Y.; Yang, Z. Metagenome-wide association of gut microbiome features for schizophrenia. Nat. Commun. 2020, 11, 1612. [Google Scholar] [CrossRef] [Green Version]
- Ma, X.; Asif, H.; Dai, L.; He, Y.; Zheng, W.; Wang, D.; Ren, H.; Tang, J.; Li, C.; Jin, K. 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. [Google Scholar] [CrossRef]
- Gressier, F.; Porcelli, S.; Calati, R.; Serretti, A. Pharmacogenetics of clozapine response and induced weight gain: A comprehensive review and meta-analysis. Eur. Neuropsychopharmacol. 2016, 26, 163–185. [Google Scholar] [CrossRef]
- Penninx, B.W.; Lange, S.M. Metabolic syndrome in psychiatric patients: Overview, mechanisms, and implications. Dialogues Clin. Neurosci. 2018, 20, 63. [Google Scholar]
- Bahr, S.; Tyler, B.; Wooldridge, N.; Butcher, B.; Burns, T.; Teesch, L.; Oltman, C.; Azcarate-Peril, M.; Kirby, J.; Calarge, C. Use of the second-generation antipsychotic, risperidone, and secondary weight gain are associated with an altered gut microbiota in children. Transl. Psychiatry 2015, 5, e652. [Google Scholar] [CrossRef] [PubMed]
- Yuan, X.; Zhang, P.; Wang, Y.; Liu, Y.; Li, X.; Kumar, B.U.; Hei, G.; Lv, L.; Huang, X.F.; Fan, X.; et al. Changes in metabolism and microbiota after 24-week risperidone treatment in drug naive, normal weight patients with first episode schizophrenia. Schizophr. Res. 2018, 201, 299–306. [Google Scholar] [CrossRef]
- Pełka-Wysiecka, J.; Kaczmarczyk, M.; Bąba-Kubiś, A.; Liśkiewicz, P.; Wroński, M.; Skonieczna-Żydecka, K.; Marlicz, W.; Misiak, B.; Starzyńska, T.; Kucharska-Mazur, J. Analysis of gut microbiota and their metabolic potential in patients with schizophrenia treated with olanzapine: Results from a six-week observational prospective cohort study. J. Clin. Med. 2019, 8, 1605. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, J.C.W.; Gorbovskaya, I.; Hahn, M.K.; Muller, D.J. The gut microbiome in schizophrenia and the potential benefits of prebiotic and probiotic treatment. Nutrients 2021, 13, 1152. [Google Scholar] [CrossRef] [PubMed]
- Cussotto, S.; Walsh, J.; Golubeva, A.V.; Zhdanov, A.V.; Strain, C.R.; Fouhy, F.; Stanton, C.; Dinan, T.G.; Hyland, N.P.; Clarke, G. The gut microbiome influences the bioavailability of olanzapine in rats. EBioMedicine 2021, 66, 103307. [Google Scholar] [CrossRef]
- Flanagan, R.; Morgan, P. Is there a place for therapeutic drug monitoring of olanzapine? Clin. Pharm. 2011, 3, 348. [Google Scholar]
- Sen, M. Role of probiotics in health and disease–A review. Int. J. Adv. Life Sci. Res. 2019, 1–11. [Google Scholar]
- Tomasik, J.; Yolken, R.H.; Bahn, S.; Dickerson, F.B. Immunomodulatory effects of probiotic supplementation in schizophrenia patients: A randomized, placebo-controlled trial. Biomark. Insights 2015, 10, 47–54. [Google Scholar] [CrossRef]
- Severance, E.G.; Gressitt, K.L.; Stallings, C.R.; Katsafanas, E.; Schweinfurth, L.A.; Savage, C.L.; Adamos, M.B.; Sweeney, K.M.; Origoni, A.E.; Khushalani, S. Probiotic normalization of candida albicans in schizophrenia: A randomized, placebo-controlled, longitudinal pilot study. Brain Behav. Immun. 2017, 62, 41–45. [Google Scholar] [CrossRef] [Green Version]
- Dickerson, F.B.; Stallings, C.; Origoni, A.; Katsafanas, E.; Savage, C.L.; Schweinfurth, L.A.; Goga, J.; Khushalani, S.; Yolken, R.H. Effect of probiotic supplementation on schizophrenia symptoms and association with gastrointestinal functioning: A randomized, placebo-controlled trial. Prim. Care Companion CNS Disord. 2014, 16, 26294. [Google Scholar] [CrossRef] [Green Version]
- Okubo, R.; Koga, M.; Katsumata, N.; Odamaki, T.; Matsuyama, S.; Oka, M.; Narita, H.; Hashimoto, N.; Kusumi, I.; Xiao, J. 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. [Google Scholar] [CrossRef]
- Ghaderi, A.; Banafshe, H.R.; Mirhosseini, N.; Moradi, M.; Karimi, M.-A.; Mehrzad, F.; Bahmani, F.; Asemi, Z. Clinical and metabolic response to vitamin D plus probiotic in schizophrenia patients. BMC Psychiatry 2019, 19, 77. [Google Scholar] [CrossRef] [Green Version]
- Wu, D.; Lewis, E.D.; Pae, M.; Meydani, S.N. Nutritional modulation of immune function: Analysis of evidence, mechanisms, and clinical relevance. Front. Immunol. 2019, 9, 3160. [Google Scholar] [CrossRef] [PubMed]
- Xu, R.; Wu, B.; Liang, J.; He, F.; Gu, W.; Li, K.; Luo, Y.; Chen, J.; Gao, Y.; Wu, Z. Altered gut microbiota and mucosal immunity in patients with schizophrenia. Brain Behav. Immun. 2020, 85, 120–127. [Google Scholar] [CrossRef] [PubMed]
- Shen, Y.; Xu, J.; Li, Z.; Huang, Y.; Yuan, Y.; Wang, J.; Zhang, M.; Hu, S.; Liang, Y. 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. [Google Scholar] [CrossRef] [PubMed]
- Zhang, X.; Pan, L.-Y.; Zhang, Z.; Zhou, Y.-Y.; Jiang, H.-Y.; Ruan, B. Analysis of gut mycobiota in first-episode, drug-naïve chinese patients with schizophrenia: A pilot study. Behav. Brain Res. 2020, 379, 112374. [Google Scholar] [CrossRef]
- Ng, Q.X.; Soh, A.Y.S.; Venkatanarayanan, N.; Ho, C.Y.X.; Lim, D.Y.; Yeo, W.-S. A systematic review of the effect of probiotic supplementation on schizophrenia symptoms. Neuropsychobiology 2019, 78, 1–6. [Google Scholar] [CrossRef]
- Kang, D.; Zhang, F.; Yang, Y.; Liu, C.; Xiao, J.; Long, Y.; Huang, J.; Peng, X.; Wang, W.; Wang, X. Probiotic supplements reduce antipsychotic-induced metabolic disturbances in drug-naive first-episode schizophrenia. medRxiv 2021. [Google Scholar] [CrossRef]
- Sarkar, A.; Lehto, S.M.; Harty, S.; Dinan, T.G.; Cryan, J.F.; Burnet, P.W. Psychobiotics and the manipulation of bacteria–gut–brain signals. Trends Neurosci. 2016, 39, 763–781. [Google Scholar] [CrossRef] [Green Version]
- Suganya, K.; Koo, B.-S. Gut–brain axis: Role of gut microbiota on neurological disorders and how probiotics/prebiotics beneficially modulate microbial and immune pathways to improve brain functions. Int. J. Mol. Sci. 2020, 21, 7551. [Google Scholar] [CrossRef]
- Burokas, A.; Arboleya, S.; Moloney, R.D.; Peterson, V.L.; Murphy, K.; Clarke, G.; Stanton, C.; Dinan, T.G.; Cryan, J.F. Targeting the microbiota-gut-brain axis: Prebiotics have anxiolytic and antidepressant-like effects and reverse the impact of chronic stress in mice. Biol. Psychiatry 2017, 82, 472–487. [Google Scholar] [CrossRef] [PubMed]
- Barber, T.M.; Valsamakis, G.; Mastorakos, G.; Hanson, P.; Kyrou, I.; Randeva, H.S.; Weickert, M.O. Dietary influences on the microbiota–gut–brain axis. Int. J. Mol. Sci. 2021, 22, 3502. [Google Scholar] [CrossRef]
- Berding, K.; Carbia, C.; Cryan, J.F. Going with the grain: Fiber, cognition, and the microbiota-gut-brain-axis. Exp. Biol. Med. 2021, 246, 796–811. [Google Scholar] [CrossRef]
- Gibson, G.R.; Hutkins, R.; Sanders, M.E.; Prescott, S.L.; Reimer, R.A.; Salminen, S.J.; Scott, K.; Stanton, C.; Swanson, K.S.; Cani, P.D. Expert consensus document: The international scientific association for probiotics and prebiotics (isapp) consensus statement on the definition and scope of prebiotics. Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 491–502. [Google Scholar] [CrossRef] [Green Version]
- Food and Drug Administration. Food Labeling and Nutrition: Questions and Answers on Dietary Fiber. Available online: https://www.fda.gov/food/food-labeling-nutrition/questions-and-answers-dietary-fiber#synthetic_fibers (accessed on 17 December 2021).
- Mohanty, D.; Misra, S.; Mohapatra, S.; Sahu, P.S. Prebiotics and synbiotics: Recent concepts in nutrition. Food Biosci. 2018, 26, 152–160. [Google Scholar] [CrossRef]
- de Paulo Farias, D.; Neri-Numa, I.A.; de Araújo, F.F.; Pastore, G.M. A critical review of some fruit trees from the myrtaceae family as promising sources for food applications with functional claims. Food Chem. 2020, 306, 125630. [Google Scholar] [CrossRef]
- Quigley, E.M. Prebiotics and probiotics in digestive health. Clin. Gastroenterol. Hepatol. 2019, 17, 333–344. [Google Scholar] [CrossRef]
- Davani-Davari, D.; Negahdaripour, M.; Karimzadeh, I.; Seifan, M.; Mohkam, M.; Masoumi, S.J.; Berenjian, A.; Ghasemi, Y. Prebiotics: Definition, types, sources, mechanisms, and clinical applications. Foods 2019, 8, 92. [Google Scholar] [CrossRef] [Green Version]
- Black, S.; Kushner, I.; Samols, D. C-reactive protein. J. Biol. Chem. 2004, 279, 48487–48490. [Google Scholar] [CrossRef] [Green Version]
- Fernandes, B.; Steiner, J.; Bernstein, H.; Dodd, S.; Pasco, J.; Dean, O.; Nardin, P.; Goncalves, C.; Berk, M. C-reactive protein is increased in schizophrenia but is not altered by antipsychotics: Meta-analysis and implications. Mol. Psychiatry 2016, 21, 554–564. [Google Scholar] [CrossRef] [PubMed]
- McLoughlin, R.F.; Berthon, B.S.; Jensen, M.E.; Baines, K.J.; Wood, L.G. Short-chain fatty acids, prebiotics, synbiotics, and systemic inflammation: A systematic review and meta-analysis. Am. J. Clin. Nutr. 2017, 106, 930–945. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kao, A.; Harty, S.; Burnet, P. The influence of prebiotics on neurobiology and behavior. Int. Rev. Neurobiol. 2016, 131, 21–48. [Google Scholar] [PubMed]
- Kao, A.C.-C.; Burnet, P.W.; Lennox, B.R. Can prebiotics assist in the management of cognition and weight gain in schizophrenia? Psychoneuroendocrinology 2018, 95, 179–185. [Google Scholar] [CrossRef] [PubMed]
- Gronier, B.; Savignac, H.M.; Di Miceli, M.; Idriss, S.M.; Tzortzis, G.; Anthony, D.; Burnet, P.W. Increased cortical neuronal responses to NMDA and improved attentional set-shifting performance in rats following prebiotic (b-gos®) ingestion. Eur. Neuropsychopharmacol. 2018, 28, 211–224. [Google Scholar] [CrossRef] [PubMed]
- Swann, O.G.; Kilpatrick, M.; Breslin, M.; Oddy, W.H. Dietary fiber and its associations with depression and inflammation. Nutr. Rev. 2020, 78, 394–411. [Google Scholar] [CrossRef] [PubMed]
- Kim, C.-S.; Byeon, S.; Shin, D.-M. Sources of dietary fiber are differently associated with prevalence of depression. Nutrients 2020, 12, 2813. [Google Scholar] [CrossRef]
- Muth, A.-K.; Park, S.Q. The impact of dietary macronutrient intake on cognitive function and the brain. Clin. Nutr. 2021, 40, 3999–4010. [Google Scholar] [CrossRef]
- La Torre, D.; Verbeke, K.; Dalile, B. Dietary fibre and the gut–brain axis: Microbiota-dependent and independent mechanisms of action. Gut Microbiome 2021, 2, 1–39. [Google Scholar] [CrossRef]
- Guo, L.; Xiao, P.; Zhang, X.; Yang, Y.; Yang, M.; Wang, T.; Lu, H.; Tian, H.; Wang, H.; Liu, J. Inulin ameliorates schizophrenia via modulation of the gut microbiota and anti-inflammation in mice. Food Funct. 2021, 12, 1156–1175. [Google Scholar] [CrossRef]
- Bravo, J.A.; Forsythe, P.; Chew, M.V.; Escaravage, E.; Savignac, H.M.; Dinan, T.G.; Bienenstock, J.; Cryan, J.F. Ingestion of lactobacillus strain regulates emotional behavior and central gaba receptor expression in a mouse via the vagus nerve. Proc. Natl. Acad. Sci. USA 2011, 108, 16050–16055. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Messaoudi, M.; Lalonde, R.; Violle, N.; Javelot, H.; Desor, D.; Nejdi, A.; Bisson, J.-F.; Rougeot, C.; Pichelin, M.; Cazaubiel, M. 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, 105, 755–764. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Munawar, N. A note on prebiotics strategy to cope with current health challenges in human beings. Am. J. Biomed. Sci. Res. 2020, 8, 326–330. [Google Scholar] [CrossRef]
- Gómez-Pinilla, F. Brain foods: The effects of nutrients on brain function. Nat. Rev. Neurosci. 2008, 9, 568–578. [Google Scholar] [CrossRef] [Green Version]
- Ray, S.K.; Mukherjee, S. Evolving interplay between dietary polyphenols and gut microbiota—An emerging importance in healthcare. Front. Nutr. 2021, 8, 634944. [Google Scholar] [CrossRef]
- Cardona, F.; Andrés-Lacueva, C.; Tulipani, S.; Tinahones, F.J.; Queipo-Ortuño, M.I. Benefits of polyphenols on gut microbiota and implications in human health. J. Nutr. Biochem. 2013, 24, 1415–1422. [Google Scholar] [CrossRef] [Green Version]
- Corrêa, T.A.F.; Rogero, M.M.; Hassimotto, N.M.A.; Lajolo, F.M. The two-way polyphenols-microbiota interactions and their effects on obesity and related metabolic diseases. Front. Nutr. 2019, 6, 188. [Google Scholar] [CrossRef] [Green Version]
- Sun, C.; Zhao, C.; Guven, E.C.; Paoli, P.; Simal-Gandara, J.; Ramkumar, K.M.; Wang, S.; Buleu, F.; Pah, A.; Turi, V. Dietary polyphenols as antidiabetic agents: Advances and opportunities. Food Front. 2020, 1, 18–44. [Google Scholar] [CrossRef] [Green Version]
- Ofosu, F.K.; Daliri, E.B.-M.; Elahi, F.; Chelliah, R.; Lee, B.-H.; Oh, D.-H. New insights on the use of polyphenols as natural preservatives and their emerging safety concerns. Front. Sustain. Food Syst. 2020, 4, 223. [Google Scholar] [CrossRef]
- Silva, R.F.; Pogačnik, L. Polyphenols from food and natural products: Neuroprotection and safety. Antioxidants 2020, 9, 61. [Google Scholar] [CrossRef] [Green Version]
- Vauzour, D. Dietary polyphenols as modulators of brain functions: Biological actions and molecular mechanisms underpinning their beneficial effects. Oxidative Med. Cell. Longev. 2012, 2012, 914273. [Google Scholar] [CrossRef] [Green Version]
- Shabbir, U.; Tyagi, A.; Elahi, F.; Aloo, S.O.; Oh, D.-H. The potential role of polyphenols in oxidative stress and inflammation induced by gut microbiota in alzheimer’s disease. Antioxidants 2021, 10, 1370. [Google Scholar] [CrossRef] [PubMed]
- Figueira, I.; Garcia, G.; Pimpão, R.C.; Terrasso, A.; Costa, I.; Almeida, A.; Tavares, L.; Pais, T.; Pinto, P.; Ventura, M. Polyphenols journey through blood-brain barrier towards neuronal protection. Sci. Rep. 2017, 7, 11456. [Google Scholar] [CrossRef] [PubMed]
- Lv, M.; Yang, S.; Cai, L.; Qin, L.Q.; Li, B.Y.; Wan, Z. Effects of quercetin intervention on cognition function in app/ps1 mice was affected by vitamin d status. Mol. Nutr. Food Res. 2018, 62, 1800621. [Google Scholar] [CrossRef] [PubMed]
- Loftis, J.M.; Wilhelm, C.J.; Huckans, M. Effect of epigallocatechin gallate supplementation in schizophrenia and bipolar disorder: An 8-week, randomized, double-blind, placebo-controlled study. Ther. Adv. Psychopharmacol. 2013, 3, 21–27. [Google Scholar] [CrossRef] [Green Version]
- Nsor-Atindana, J.; Chen, M.; Goff, H.D.; Zhong, F.; Sharif, H.R.; Li, Y. Functionality and nutritional aspects of microcrystalline cellulose in food. Carbohydr. Polym. 2017, 172, 159–174. [Google Scholar] [CrossRef]
- Bishnoi, M.; Chopra, K.; Kulkarni, S.K. Protective effect of curcumin, the active principle of turmeric (curcuma longa) in haloperidol-induced orofacial dyskinesia and associated behavioural, biochemical and neurochemical changes in rat brain. Pharmacol. Biochem. Behav. 2008, 88, 511–522. [Google Scholar] [CrossRef]
- Naidu, P.; Kulkarni, S. Quercetin, a bioflavonoid, reverses haloperidol-induced catalepsy. Methods Find. Exp. Clin. Pharmacol. 2004, 26, 323–326. [Google Scholar] [CrossRef]
- Dietrich-Muszalska, A.; Kontek, B.; Olas, B.; Rabe-Jabłońska, J. Epicatechin inhibits human plasma lipid peroxidation caused by haloperidol in vitro. Neurochem. Res. 2012, 37, 557–562. [Google Scholar] [CrossRef]
- Dietrich-Muszalska, A.; Kopka, J.; Kontek, B. Polyphenols from berries of aronia melanocarpa reduce the plasma lipid peroxidation induced by ziprasidone. Schizophr. Res. Treat. 2014, 2014, 602390. [Google Scholar]
- Chen, L.; Zhang, C.; Han, Y.; Meng, X.; Zhang, Y.; Chu, H.; Ma, H. Gingko biloba extract (egb) inhibits oxidative stress in neuro 2a cells overexpressing appsw. BioMed Res. Int. 2019, 2019, 7034983. [Google Scholar] [CrossRef] [Green Version]
- Ramassamy, C.; Christen, Y.; Clostre, F.; Costentin, J. The ginkgo biloba extract, egb761, increases synaptosomal uptake of 5-hydroxytryptamine: In-vitro and ex-vivo studies. J. Pharm. Pharmacol. 1992, 44, 943–945. [Google Scholar] [CrossRef] [PubMed]
- Wan, M.L.Y.; Co, V.A.; El-Nezami, H. Dietary polyphenol impact on gut health and microbiota. Crit. Rev. Food Sci. Nutr. 2021, 61, 690–711. [Google Scholar] [CrossRef] [PubMed]
- Tzounis, X.; Rodriguez-Mateos, A.; Vulevic, J.; Gibson, G.R.; Kwik-Uribe, C.; Spencer, J.P. Prebiotic evaluation of cocoa-derived flavanols in healthy humans by using a randomized, controlled, double-blind, crossover intervention study. Am. J. Clin. Nutr. 2011, 93, 62–72. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ghoshal, U.C.; Shukla, R.; Ghoshal, U.; Gwee, K.-A.; Ng, S.C.; Quigley, E.M. The gut microbiota and irritable bowel syndrome: Friend or foe? Int. J. Inflamm. 2012, 2012, 151085. [Google Scholar] [CrossRef]
- Morgan, X.C.; Tickle, T.L.; Sokol, H.; Gevers, D.; Devaney, K.L.; Ward, D.V.; Reyes, J.A.; Shah, S.A.; LeLeiko, N.; Snapper, S.B. Dysfunction of the intestinal microbiome in inflammatory bowel disease and treatment. Genome Biol. 2012, 13, 1–18. [Google Scholar] [CrossRef]
- Wehkamp, J.; Harder, J.; Weichenthal, M.; Mueller, O.; Herrlinger, K.R.; Fellermann, K.; Schroeder, J.M.; Stange, E.F. Inducible and constitutive β-defensins are differentially expressed in crohn’s disease and ulcerative colitis. Inflamm. Bowel Dis. 2003, 9, 215–223. [Google Scholar] [CrossRef]
- Mayta-Apaza, A.C.; Pottgen, E.; De Bodt, J.; Papp, N.; Marasini, D.; Howard, L.; Abranko, L.; Van de Wiele, T.; Lee, S.-O.; Carbonero, F. Impact of tart cherries polyphenols on the human gut microbiota and phenolic metabolites in vitro and in vivo. J. Nutr. Biochem. 2018, 59, 160–172. [Google Scholar] [CrossRef]
- Queipo-Ortuño, M.I.; Boto-Ordóñez, M.; Murri, M.; Gomez-Zumaquero, J.M.; Clemente-Postigo, M.; Estruch, R.; Cardona Diaz, F.; Andres-Lacueva, C.; Tinahones, F.J. Influence of red wine polyphenols and ethanol on the gut microbiota ecology and biochemical biomarkers. Am. J. Clin. Nutr. 2012, 95, 1323–1334. [Google Scholar] [CrossRef]
- Kilua, A.; Nomata, R.; Nagata, R.; Fukuma, N.; Shimada, K.; Han, K.-H.; Fukushima, M. Purple sweet potato polyphenols differentially influence the microbial composition depending on the fermentability of dietary fiber in a mixed culture of swine fecal bacteria. Nutrients 2019, 11, 1495. [Google Scholar] [CrossRef] [Green Version]
- Mansoorian, B.; Combet, E.; Alkhaldy, A.; Garcia, A.L.; Edwards, C.A. Impact of fermentable fibres on the colonic microbiota metabolism of dietary polyphenols rutin and quercetin. Int. J. Environ. Res. Public Health 2019, 16, 292. [Google Scholar] [CrossRef] [Green Version]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Munawar, N.; Ahmad, A.; Anwar, M.A.; Muhammad, K. Modulation of Gut Microbial Diversity through Non-Pharmaceutical Approaches to Treat Schizophrenia. Int. J. Mol. Sci. 2022, 23, 2625. https://doi.org/10.3390/ijms23052625
Munawar N, Ahmad A, Anwar MA, Muhammad K. Modulation of Gut Microbial Diversity through Non-Pharmaceutical Approaches to Treat Schizophrenia. International Journal of Molecular Sciences. 2022; 23(5):2625. https://doi.org/10.3390/ijms23052625
Chicago/Turabian StyleMunawar, Nayla, Aftab Ahmad, Munir Ahmad Anwar, and Khalid Muhammad. 2022. "Modulation of Gut Microbial Diversity through Non-Pharmaceutical Approaches to Treat Schizophrenia" International Journal of Molecular Sciences 23, no. 5: 2625. https://doi.org/10.3390/ijms23052625