Skip to main content Accessibility help
×
Hostname: page-component-76dd75c94c-x59qb Total loading time: 0 Render date: 2024-04-30T07:59:57.023Z Has data issue: false hasContentIssue false

Chapter 13 - Microbiome-Gut-Brain Interactions in Neurodevelopmental Disorders: Focus on Autism and Schizophrenia

Published online by Cambridge University Press:  02 September 2021

Golam Khandaker
Affiliation:
University of Cambridge
Neil Harrison
Affiliation:
Cardiff University Brain Research Imaging Centre (CUBRIC)
Edward Bullmore
Affiliation:
University of Cambridge
Robert Dantzer
Affiliation:
University of Texas, MD Anderson Cancer Center
Get access

Summary

Human brain development is a complex process. For instance, in the third week of gestation, many of the %7E 86 billion neurons observed in the adult brain are being produced (1). Subsequently, there is a constant refining and fine-tuning of these connections that continues after birth until late adolescence. This process of circuit remodelling is critical for normal brain function and behaviour (2) and factors that interfere with these processes can result in brain miswiring and associated behavioural manifestations. Genetic and environmental factors have long been recognized to modulate this complex process of brain development (2). However, there is a growing appreciation of the role that bacterial commensals of the gastrointestinal (GI) system (collectively referred to as gut microbiota which also includes viruses, fungi and protozoans) have in shaping host health and behaviour following birth (3,4). The gut microbiota plays a critical role in the maturation of the immune system especially in the early stages of life. The colonization and development of gut microbiota in early life has been associated with disease later in life (5). The gut microbiota comprises trillions of bacteria, which play a key role in morphological and neurological processes such as brain development, neurotransmission, neuroinflammation, neurogenesis and modulation of behaviour (6–9). Deleterious alterations to the gut microbiota through various factors such as diet, age, stress, mode of delivery and antibiotic administration can disrupt brain physiology and behaviour. Administration of antibiotics is one such example, where ablation of the gut microbiota composition can result in cognitive impairments (10), metabolic alterations (11) and disrupted immune function (12). Cumulative data from both preclinical and clinical studies suggest a key role for the gut microbiota in the onset of various psychiatric and neurological conditions including depression (13,14), anxiety (15), stroke (16), autism (17) and schizophrenia (18).

Type
Chapter
Information
Publisher: Cambridge University Press
Print publication year: 2021

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Azevedo, FA, Carvalho, LR, Grinberg, LT, et al. Equal numbers of neuronal and nonneuronal cells make the human brain an isometrically scaled-up primate brain. J. Comp. Neurol. 2009; 513:532541.CrossRefGoogle Scholar
Tognini, P. Gut microbiota: a potential regulator of neurodevelopment. Front Cell Neurosci. 2017;11:25.Google Scholar
Sekirov, I, Russell, SL, Antunes, LC, Finlay, BB. Gut microbiota in health and disease. Physiol Rev. 2010;90:859904.Google Scholar
Derrien, M, Alvarez, AS, de Vos, WM. The gut microbiota in the first decade of life. Trends Microbiol. 2019;27:9971010.Google Scholar
Zhuang, L, Chen, H, Zhang, S, Zhuang, J, Li, Q, Feng, Z. Intestinal microbiota in early life and its implications on childhood health. Genomics Proteomics Bioinformatics. 2019;17:1325.Google Scholar
Qin, J, Li, R, Raes, J, et al. A human gut microbial gene catalogue established by metagenomic sequencing. Nature. 2010;464:5965.Google Scholar
Dinan, TG, Cryan, JF. The microbiome-gut-brain axis in health and disease. Gastroenterol Clin North Am. 2017;46:7789.CrossRefGoogle ScholarPubMed
Sampson, TR, Debelius, JW, Thron, T, et al. Gut microbiota regulate motor deficits and neuroinflammation in a model of Parkinson’s disease. Cell. 2016;167:1469–80 e12.Google Scholar
Sherwin, E, Sandhu, KV, Dinan, TG, Cryan, JF. May the force be with you: the light and dark sides of the microbiota-gut-brain axis in neuropsychiatry. CNS Drugs. 2016;30:1019–41.Google Scholar
Frohlich, EE, Farzi, A, Mayerhofer, R, et al. Cognitive impairment by antibiotic-induced gut dysbiosis: analysis of gut microbiota-brain communication. Brain Behav Immun. 2016;56:140–55.Google Scholar
Yang, JH, Bhargava, P, McCloskey, D, et al. Antibiotic-induced changes to the host metabolic environment inhibit drug efficacy and alter immune function. Cell Host Microbe. 2017;22:757–65 e3.Google Scholar
Becattini, S, Taur, Y, Pamer, EG. Antibiotic-induced changes in the intestinal microbiota and disease. Trends Mol Med. 2016;22:458–78.Google Scholar
Kelly, JR, Borre, Y, O’Brien, C, et al. Transferring the blues: depression-associated gut microbiota induces neurobehavioural changes in the rat. J Psychiatr Res. 2016;82:109–18.CrossRefGoogle ScholarPubMed
Jiang, H, Ling, Z, Zhang, Y, et al. Altered fecal microbiota composition in patients with major depressive disorder. Brain Behav Immun. 2015;48:186–94.Google Scholar
Clapp, M, Aurora, N, Herrera, L, et al. Gut microbiota’s effect on mental health: The gut-brain axis. Clin Pract. 2017;7:987.CrossRefGoogle ScholarPubMed
1Ridler, C. Gut microbiota: gut bacteria affect post-ischaemic inflammation in stroke by modulating intestinal T cells. Nat Rev Gastroenterol Hepatol. 2016;13:250.Google Scholar
Mulle, JG, Sharp, WG, Cubells, JF. The gut microbiome: a new frontier in autism research. Curr Psychiatry Rep. 2013;15:337.Google Scholar
Dinan, TG, Borre, YE, Cryan, JF. Genomics of schizophrenia: time to consider the gut microbiome? Mol Psychiatry. 2014;19:1252–7.Google Scholar
Sandhu, KV, Sherwin, E, Schellekens, H, et al. Feeding the microbiota-gut-brain axis: diet, microbiome, and neuropsychiatry. Transl Res. 2017;179:223–44.Google Scholar
Arentsen, T, Qian, Y, Gkotzis, S, et al. The bacterial peptidoglycan-sensing molecule Pglyrp2 modulates brain development and behavior. Mol Psychiatry. 2017;22:257–66.CrossRefGoogle ScholarPubMed
Critchley, HD, Harrison, NA. Visceral influences on brain and behavior. Neuron. 2013;77(4):624–38.CrossRefGoogle ScholarPubMed
SAM, Carabotti M., Maselli, AM, Severia, C. The gut-brain axis: interactions between enteric microbiota, central and enteric nervous systems. Annals of Gastroenterology. 2015;28:203–9.Google Scholar
Lyte, M. Microbial endocrinology in the microbiome-gut-brain axis: how bacterial production and utilization of neurochemicals influence behavior. PLoS Pathog. 2013;9:e1003726.CrossRefGoogle ScholarPubMed
Foster, JA, Rinaman, L, Cryan, JF. Stress & the gut-brain axis: regulation by the microbiome. Neurobiol Stress. 2017;7:124–36.Google Scholar
Gautam, A, Kumar, R, Chakraborty, N, et al. Altered fecal microbiota composition in all male aggressor-exposed rodent model simulating features of post-traumatic stress disorder. J Neurosci Res. 2018;96:1311–23.CrossRefGoogle ScholarPubMed
Bravo, JA, Forsythe, P, Chew, MV, et al. 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;108:16050–5.Google Scholar
Allen, AP, Hutch, W, Borre, YE, et al. Bifidobacterium longum 1714 as a translational psychobiotic: modulation of stress, electrophysiology and neurocognition in healthy volunteers. Transl Psychiatry. 2016;6:e939.Google Scholar
Sudo, N, Chida, Y, Aiba, Y, et al. Postnatal microbial colonization programs the hypothalamic-pituitary-adrenal system for stress response in mice. J Physiol. 2004;558:263–75.Google Scholar
Poutahidis, T, Kearney, SM, Levkovich, T, et al. Microbial symbionts accelerate wound healing via the neuropeptide hormone oxytocin. PLoS One. 2013 Oct 30;8(10):e78898.Google Scholar
Buffington, SA, Di Prisco, GV, Auchtung, TA, et al. Microbial Reconstitution Reverses Maternal Diet-Induced Social and Synaptic Deficits in Offspring. Cell. 2016;165:1762–75.Google Scholar
Diaz Heijtz, R, Wang, S, Anuar, F, et al. Normal gut microbiota modulates brain development and behavior. Proc Natl Acad Sci USA. 2011;108:3047–52.Google Scholar
Clarke, G, Grenham, S, Scully, P, et al. The microbiome-gut-brain axis during early life regulates the hippocampal serotonergic system in a sex-dependent manner. Mol Psychiatry. 2013;18:666–73.Google Scholar
Saulnier, DM, Ringel, Y, Heyman, MB, et al. The intestinal microbiome, probiotics and prebiotics in neurogastroenterology. Gut Microbes. 2013;4:1727.Google Scholar
Burokas, A, Arboleya, S, Moloney, RD, et al. 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;82:472–87.Google Scholar
Marques, TM, Patterson, E, Wall, R, et al. Influence of GABA and GABA-producing Lactobacillus brevis DPC 6108 on the development of diabetes in a streptozotocin rat model. Benef Microbes. 2016;7:409–20.CrossRefGoogle Scholar
Sobko, T. Influence of the microflors on gastrointestinal nitric oxide generation. Unpublished Ph.D. thesis, Karolinska Institutet, Sweden, 2006.Google Scholar
Schicho, R, Krueger, D, Zeller, F, et al. Hydrogen sulfide is a novel prosecretory neuromodulator in the Guinea-pig and human colon. Gastroenterology. 2006;131:1542–52.Google Scholar
Latorre, R, Sternini, C, De Giorgio, R, Greenwood-Van Meerveld, B. Enteroendocrine cells: a review of their role in brain-gut communication. Neurogastroenterol Motil. 2016;28:620–30.CrossRefGoogle ScholarPubMed
Berthoud, HR. The vagus nerve, food intake and obesity. Regul Pept. 2008;149:1525.Google Scholar
Batterham, RL, Cowley, MA, Small, CJ, et al. Gut hormone PYY3-36 physiologically inhibits food intake. Nature. 2002;418:650–4.CrossRefGoogle Scholar
Koda, S, Date, Y, Murakami, N, et al. The role of the vagal nerve in peripheral PYY3-36-induced feeding reduction in rats. Endocrinology. 2005;146:2369–75.Google Scholar
Bercik, P, Denou, E, Collins, J, et al. The intestinal microbiota affect central levels of brain-derived neurotropic factor and behavior in mice. Gastroenterology. 2011;141:599609, e1-3.Google Scholar
Kunze, WA, Mao, YK, Wang, B, et al. Lactobacillus reuteri enhances excitability of colonic AH neurons by inhibiting calcium-dependent potassium channel opening. J Cell Mol Med. 2009;13:2261–70.Google Scholar
Macpherson, AJ, Slack, E, Geuking, MB, McCoy, KD. The mucosal firewalls against commensal intestinal microbes. Semin Immunopathol. 2009;31:145–9.CrossRefGoogle ScholarPubMed
McGuckin, MA, Linden, SK, Sutton, P, Florin, TH. Mucin dynamics and enteric pathogens. Nat Rev Microbiol. 2011;9:265–78.Google Scholar
Hooper, LV, Macpherson, AJ. Immune adaptations that maintain homeostasis with the intestinal microbiota. Nat Rev Immunol. 2010;10:159–69.CrossRefGoogle ScholarPubMed
Pamer, EG. Immune responses to commensal and environmental microbes. Nat Immunol. 2007;8:1173–8.Google Scholar
Cash, HL, Whitham, CV, Behrendt, CL, Hooper, LV. Symbiotic bacteria direct expression of an intestinal bactericidal lectin. Science. 2006;313:1126–30.Google Scholar
Bengmark, S. Gut microbiota, immune development and function. Pharmacol Res. 2013;69:87113.Google Scholar
Dantzer, R, O’Connor, JC, Freund, GG, Johnson, RW, Kelley, KW. From inflammation to sickness and depression: when the immune system subjugates the brain. Nat Rev Neurosci. 2008;9:4656.Google Scholar
Steer, T, Carpenter, H, Tuohy, K, Gibson, GR. Perspectives on the role of the human gut microbiota and its modulation by pro- and prebiotics. Nutr Res Rev. 2000;13:229–54.Google Scholar
Maes, M, Kubera, M, Leunis, J-C, Berk, M. Increased IgA and IgM responses against gut commensals in chronic depression: further evidence for increased bacterial translocation or leaky gut. J Affect Disord. 2012;141:5562.Google Scholar
Erny, D, Hrabe de Angelis, AL, Jaitin, D, et al. Host microbiota constantly control maturation and function of microglia in the CNS. Nat Neurosci. 2015;18:965–77.Google Scholar
Kau, AL, Ahern, PP, Griffin, NW, Goodman, AL, Gordon, JI. Human nutrition, the gut microbiome and the immune system. Nature. 2011;474:327–36.Google Scholar
Koh, A, De Vadder, F, Kovatcheva-Datchary, P, Backhed, F. From dietary fiber to host physiology: short-chain fatty acids as key bacterial metabolites. Cell. 2016;165:1332–45.Google Scholar
Stilling, RM, Dinan, TG, Cryan, JF. Microbial genes, brain & behaviour – epigenetic regulation of the gut-brain axis. Genes Brain Behav. 2014;13:6986.Google Scholar
Hullar, MA, Fu, BC. Diet, the gut microbiome, and epigenetics. Cancer J. 2014;20:170–5.Google Scholar
den Besten, G, van Eunen, K, Groen, AK, et al. The role of short-chain fatty acids in the interplay between diet, gut microbiota, and host energy metabolism. J Lipid Res. 2013;54:2325–40.Google Scholar
Stilling, RM, van de Wouw, M, Clarke, G, et al.. The neuropharmacology of butyrate: The bread and butter of the microbiota-gut-brain axis? Neurochem Int. 2016;99:110–32.Google Scholar
Nankova, BB AR, MacFabe, DF, La Gamma, EF. Enteric bacterial metabolites propionic and butyric acid modulate gene expression, including CREB-dependent catecholaminergic neurotransmission, in PC12 cells – possible relevance to autism spectrum disorders. PLOS One. 2014;9(8):e103740.Google Scholar
El-Ansary, AK, Ben Bacha, A, Kotb, M. Etiology of autistic features: the persisting neurotoxic effects of propionic acid. J Neuroinflammation. 2012;9:74.Google Scholar
Bourassa, MW, Alim, I, Bultman, SJ, Ratan, RR. Butyrate, neuroepigenetics and the gut microbiome: Can a high fiber diet improve brain health? Neurosci Lett. 2016;625:5663.Google Scholar
Luczynski, P, Whelan, SO, O’Sullivan, C, et al. Adult microbiota-deficient mice have distinct dendritic morphological changes: differential effects in the amygdala and hippocampus. Eur J Neurosci. 2016;44:2654–66.Google Scholar
Park, H, Poo, MM. Neurotrophin regulation of neural circuit development and function. Nat Rev Neurosci. 2013;14:723.Google Scholar
Tarsa, L, Goda, Y. Synaptophysin regulates activity-dependent synapse formation in cultured hippocampal neurons. Proc Natl Acad Sci USA. 2002;99:1012–6.Google Scholar
Kwon, SE, Chapman, ER. Synaptophysin regulates the kinetics of synaptic vesicle endocytosis in central neurons. Neuron. 2011;70:847–54.Google Scholar
El-Husseini AE, SE, Chetkovich, DM, Nicoll, RA, Bredt, DS. PSD-95 involvement in maturation of excitatory synapses. SCIENCE. 2000;290:1364–8.Google Scholar
Béïque J-C, Andrade R. PSD-95 regulates synaptic transmission and plasticity in rat cerebral cortex. The Journal of Physiology. 2003;546:859–67.Google Scholar
Neufeld, KM, Kang, N, Bienenstock, J, Foster, JA. Reduced anxiety-like behavior and central neurochemical change in germ-free mice. Neurogastroenterol Motil. 2011;23:255–64, e119.CrossRefGoogle ScholarPubMed
Hoban, AE, Stilling, RM, Ryan, FJ, et al. Regulation of prefrontal cortex myelination by the microbiota. Transl Psychiatry. 2016;6:e774.Google Scholar
Asan, E, Steinke, M, Lesch, KP. Serotonergic innervation of the amygdala: targets, receptors, and implications for stress and anxiety. Histochem Cell Biol. 2013;139:785813.Google Scholar
Kelly, JR, Minuto, C, Cryan, JF, Clarke, G, Dinan, TG. Cross talk: the microbiota and neurodevelopmental disorders. Front Neurosci. 2017;11:490.Google Scholar
Reigstad, CS, Salmonson, CE, Rainey, JF, 3rd, et al. Gut microbes promote colonic serotonin production through an effect of short-chain fatty acids on enterochromaffin cells. FASEB J. 2015;29:1395–403.Google Scholar
Desbonnet, L, Clarke, G, Traplin, A, et al. Gut microbiota depletion from early adolescence in mice: Implications for brain and behaviour. Brain Behav Immun. 2015;48:165–73.CrossRefGoogle ScholarPubMed
Mohle, L, Mattei, D, Heimesaat, MM, et al. Ly6C(hi) monocytes provide a link between antibiotic-induced changes in gut microbiota and adult hippocampal neurogenesis. Cell Rep. 2016;15:1945–56.CrossRefGoogle ScholarPubMed
Stiles, J, Jernigan, TL. The basics of brain development. Neuropsychol Rev. 2010;20:327–48.Google Scholar
O’Mahony, SM, Clarke, G, Borre, YE, Dinan, TG, Cryan, JF. Serotonin, tryptophan metabolism and the brain-gut-microbiome axis. Behav Brain Res. 2015;277:3248.Google Scholar
Jasarevic, E, Rodgers, AB, Bale, TL. A novel role for maternal stress and microbial transmission in early life programming and neurodevelopment. Neurobiol Stress. 2015;1:81–8.Google Scholar
Rogers, GB, Keating, DJ, Young, RL, et al. From gut dysbiosis to altered brain function and mental illness: mechanisms and pathways. Mol Psychiatry. 2016;21:738–48.Google Scholar
Kim, S, Kim, H, Yim, YS, et al. Maternal gut bacteria promote neurodevelopmental abnormalities in mouse offspring. Nature. 2017;549:528–32.Google Scholar
Sasaki, A, de Vega, W, Sivanathan, S, St-Cyr, S, McGowan, PO. Maternal high-fat diet alters anxiety behavior and glucocorticoid signaling in adolescent offspring. Neuroscience. 2014;272:92101.Google Scholar
Davis, EP, Sandman, CA. The timing of prenatal exposure to maternal cortisol and psychosocial stress is associated with human infant cognitive development. Child Dev. 2010;81:131–48.Google Scholar
Sandman, CA, Davis, EP, Buss, C, Glynn, LM. Exposure to prenatal psychobiological stress exerts programming influences on the mother and her fetus. Neuroendocrinology. 2012;95:721.Google Scholar
Golubeva, AV, Crampton, S, Desbonnet, L, et al. Prenatal stress-induced alterations in major physiological systems correlate with gut microbiota composition in adulthood. Psychoneuroendocrinology. 2015;60:5874.Google Scholar
Jansson, T, Powell, TL. Role of the placenta in fetal programming: underlying mechanisms and potential interventional approaches. Clin Sci (Lond). 2007;113:113.Google Scholar
Glover, V, Bergman, K, Sarkar, P, O’Connor, TG. Association between maternal and amniotic fluid cortisol is moderated by maternal anxiety. Psychoneuroendocrinology. 2009;34:430–5.Google Scholar
Côté, F FC, Bayard, E, Launay, JM, et al. Maternal serotonin is crucial for murine embryonic development. Proc Natl Acad Sci USA. 2007;104:329–34.Google Scholar
Bonnin, A, Levitt, P. Placental source for 5-HT that tunes fetal brain development. Neuropsychopharmacology. 2012;37:299300.Google Scholar
Braniste, V, Al-Asmakh, M, Kowal, C, et al. The gut microbiota influences blood-brain barrier permeability in mice. Sci Transl Med. 2014;6:263ra158.Google Scholar
Borre, YE, O’Keeffe, GW, Clarke, G, et al. Microbiota and neurodevelopmental windows: implications for brain disorders. Trends Mol Med. 2014;20:509–18.Google Scholar
O’Mahony, SM, Clarke, G, Dinan, TG, Cryan, JF. Early-life adversity and brain development: Is the microbiome a missing piece of the puzzle? Neuroscience. 2017;342:3754.Google Scholar
Timmerman, HM, Rutten, N, 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;7:8327.CrossRefGoogle ScholarPubMed
Rutayisire, E, Huang, K, Liu, Y, Tao, F. The mode of delivery affects the diversity and colonization pattern of the gut microbiota during the first year of infants’ life: a systematic review. BMC Gastroenterol. 2016;16:86.Google Scholar
Neu, J, Rushing, J. Cesarean versus vaginal delivery: long-term infant outcomes and the hygiene hypothesis. Clin Perinatol. 2011;38:321–31.Google Scholar
Musilova, S, Rada, V, Vlkova, E, Bunesova, V. Beneficial effects of human milk oligosaccharides on gut microbiota. Benef Microbes. 2014;5:273–83.Google Scholar
Bode, L. Human milk oligosaccharides: every baby needs a sugar mama. Glycobiology. 2012;22:1147–62.Google Scholar
Toscano, M, De Grandi, R, Grossi, E, Drago, L. Role of the human breast milk-associated microbiota on the newborns’ immune system: a mini review. Front Microbiol. 2017;8:2100.Google Scholar
Andreas, NJ, Kampmann, B, Mehring Le-Doare, K. Human breast milk: a review on its composition and bioactivity. Early Hum Dev. 2015;91:629–35.Google Scholar
Wall, R, Marques, TM, O’Sullivan, O, et al. Contrasting effects of Bifidobacterium breve NCIMB 702258 and Bifidobacterium breve DPC 6330 on the composition of murine brain fatty acids and gut microbiota. Am J Clin Nutr. 2012;95:1278–87.Google Scholar
Santos, F, Spinler, JK, Saulnier, DM, et al. Functional identification in Lactobacillus reuteri of a PocR-like transcription factor regulating glycerol utilization and vitamin B12 synthesis. Microb Cell Fact. 2011;10:55.Google Scholar
Innis, SM. Impact of maternal diet on human milk composition and neurological development of infants. Am J Clin Nutr. 2014;99:734S-41S.Google Scholar
Edlow, AG. Maternal obesity and neurodevelopmental and psychiatric disorders in offspring. Prenat Diagn. 2017;37:95110.Google Scholar
Kim, HJ, Cho, MH, Shim, WH, et al. Deficient autophagy in microglia impairs synaptic pruning and causes social behavioral defects. Mol Psychiatry. 2017;22:1576–84.CrossRefGoogle ScholarPubMed
Boksa, P. Abnormal synaptic pruning in schizophrenia: Urban myth or reality? J Psychiatry Neurosci. 2012;37:75–7.Google Scholar
Christensen, DL, Baio, J, Van Naarden Braun, K, et al. Prevalence and characteristics of autism spectrum disorder among children aged 8 years – autism and developmental disabilities monitoring network, 11 Sites, United States, 2012. Surveillance Summaries. 2016;65(3):123.Google Scholar
Lartseva, A, Dijkstra, T, Buitelaar, JK. Emotional language processing in autism spectrum disorders: a systematic review. Front Hum Neurosci. 2014;8:991.Google Scholar
Wang, S, Adolphs, R. Reduced specificity in emotion judgment in people with autism spectrum disorder. Neuropsychologia. 2017;99:286–95.Google Scholar
Li, Q, Zhou, JM. The microbiota-gut-brain axis and its potential therapeutic role in autism spectrum disorder. Neuroscience. 2016;324:131–9.Google Scholar
De Rubeis, S, He, X, Goldberg, AP, et al. Synaptic, transcriptional and chromatin genes disrupted in autism. Nature. 2014;515:209–15.Google Scholar
Gorrindo, P, Williams, KC, Lee, EB, et al. Gastrointestinal dysfunction in autism: parental report, clinical evaluation, and associated factors. Autism Res. 2012;5:101–8.Google Scholar
Adams, JB, Johansen, LJ, Powell, LD, Quig, D, Rubin, RA. Gastrointestinal flora and gastrointestinal status in children with autism–comparisons to typical children and correlation with autism severity. BMC Gastroenterol. 2011;11:22.Google Scholar
Buie, T, Fuchs, GJ, 3rd, Furuta, GT, et al. Recommendations for evaluation and treatment of common gastrointestinal problems in children with ASDs. Pediatrics. 2010;125(Suppl 1):S1929.Google Scholar
Mazurek, MO, Vasa, RA, Kalb, LG, et al. Anxiety, sensory over-responsivity, and gastrointestinal problems in children with autism spectrum disorders. J Abnorm Child Psychol. 2013;41:165–76.Google Scholar
Finegold, SM MD, Song, Y, Liu, C, et al. Gastrointestinal microflora studies in late-onset autism. Clin Infect Dis. 2002;35:S6S16.Google Scholar
Finegold, SM, Dowd, SE, Gontcharova, V, et al. Pyrosequencing study of fecal microflora of autistic and control children. Anaerobe. 2010;16:444–53.Google Scholar
De Angelis, M, Piccolo, M, Vannini, L, et al. Fecal microbiota and metabolome of children with autism and pervasive developmental disorder not otherwise specified. PLoS One. 2013;8:e76993.Google Scholar
Kang, DW, Park, JG, Ilhan, ZE, et al. Reduced incidence of Prevotella and other fermenters in intestinal microflora of autistic children. PLoS One. 2013;8:e68322.Google Scholar
Parracho, HM, Bingham, MO, Gibson, GR, McCartney, AL. Differences between the gut microflora of children with autistic spectrum disorders and that of healthy children. J Med Microbiol. 2005;54:987–91.Google Scholar
Pequegnat, B, Sagermann, M, Valliani, M, et al. A vaccine and diagnostic target for Clostridium bolteae, an autism-associated bacterium. Vaccine. 2013;31:2787–90.Google Scholar
Finegold, SM, Summanen, PH, Downes, J, Corbett, K, Komoriya, T. Detection of Clostridium perfringens toxin genes in the gut microbiota of autistic children. Anaerobe. 2017;45:133–7.Google Scholar
Shaw, W. Increased urinary excretion of a 3-(3-hydroxyphenyl)-3-hydroxypropionic acid (HPHPA), an abnormal phenylalanine metabolite of Clostridia spp. in the gastrointestinal tract, in urine samples from patients with autism and schizophrenia. Nutr Neurosci 2010;13:135–43.Google Scholar
Kang, DW, Adams, JB, Coleman, DM, et al. Long-term benefit of Microbiota Transfer Therapy on autism symptoms and gut microbiota. Sci Rep. 2019;9:5821.Google Scholar
Wang, L, Christophersen, CT, Sorich, MJ, et al. Elevated fecal short chain fatty acid and ammonia concentrations in children with autism spectrum disorder. Dig Dis Sci. 2012;57:2096–102.Google Scholar
de Magistris, L, Familiari, V, Pascotto, A, et al. Alterations of the intestinal barrier in patients with autism spectrum disorders and in their first-degree relatives. J Pediatr Gastroenterol Nutr. 2010;51:418–24.Google Scholar
Ashwood, P, Krakowiak, P, Hertz-Picciotto, I, et al.Elevated plasma cytokines in autism spectrum disorders provide evidence of immune dysfunction and are associated with impaired behavioral outcome. Brain Behav Immun. 2011;25:40–5.CrossRefGoogle ScholarPubMed
Son, JS, Zheng, LJ, Rowehl, LM, et al. Comparison of fecal microbiota in children with autism spectrum disorders and neurotypical siblings in the Simons Simplex Collection. PLoS One. 2015;10:e0137725.Google Scholar
Shultz, SR, Macfabe, DF, Martin, S, et al. Intracerebroventricular injections of the enteric bacterial metabolic product propionic acid impair cognition and sensorimotor ability in the Long-Evans rat: further development of a rodent model of autism. Behav Brain Res. 2009;200:3341.Google Scholar
Shultz, SR, MacFabe, DF, Ossenkopp, KP, et al. Intracerebroventricular injection of propionic acid, an enteric bacterial metabolic end-product, impairs social behavior in the rat: implications for an animal model of autism. Neuropharmacology. 2008;54:901–11.Google Scholar
Hsiao, EY, McBride, SW, Hsien, S, et al. Microbiota modulate behavioral and physiological abnormalities associated with neurodevelopmental disorders. Cell. 2013;155:1451–63.Google Scholar
Pobbe, RL, Pearson, BL, Defensor, EB, et al. Expression of social behaviors of C57BL/6J versus BTBR inbred mouse strains in the visible burrow system. Behav Brain Res. 2010;214:443–9.Google Scholar
Weissbrod, A, Shapiro, A, Vasserman, G, et al. Automated long-term tracking and social behavioural phenotyping of animal colonies within a semi-natural environment. Nat Commun. 2013;4:2018.Google Scholar
Pearson, BL, Pobbe, RL, Defensor, EB, et al. Motor and cognitive stereotypies in the BTBR T+tf/J mouse model of autism. Genes Brain Behav. 2011;10:228–35.Google Scholar
Karvat, G, Kimchi, T. Systematic autistic-like behavioral phenotyping of 4 mouse strains using a novel wheel-running assay. Behav Brain Res. 2012;233:405–14.Google Scholar
Stephenson, DT, O’Neill, SM, Narayan, S, et al. Histopathologic characterization of the BTBR mouse model of autistic-like behavior reveals selective changes in neurodevelopmental proteins and adult hippocampal neurogenesis. Mol Autism. 2011;2:7.Google Scholar
Heo, Y, Zhang, Y, Gao, D, Miller, VM, Lawrence, DA. Aberrant immune responses in a mouse with behavioral disorders. PLoS One. 2011;6:e20912.Google Scholar
Golubeva, AV, Joyce, SA, Moloney, G, et al. Microbiota-related changes in bile acid & tryptophan metabolism are associated with gastrointestinal dysfunction in a mouse model of autism. EBioMedicine. 2017;24:166–78.Google Scholar
Coretti, L, Cristiano, C, Florio, E, et al. Sex-related alterations of gut microbiota composition in the BTBR mouse model of autism spectrum disorder. Sci Rep. 2017;7:45356.Google Scholar
Perälä, J SJ, Saarni, SI, Kuoppasalmi, K, et al. Lifetime prevalence of psychotic and bipolar I disorders in a general population. Arch Gen Psychiatry. 2007;64:1928.Google Scholar
Hor, K, Taylor, M. Suicide and schizophrenia: a systematic review of rates and risk factors. J Psychopharmacol. 2010;24:8190.Google Scholar
JH, G. Understanding what causes schizophrenia: a developmental perspective. AM J Psychiatry. 2008;167:810.Google Scholar
Kahn, RS, Keefe, RS. Schizophrenia is a cognitive illness: time for a change in focus. JAMA Psychiatry. 2013;70:1107–12.Google Scholar
Hommer, RE, Swedo, SE. Schizophrenia and autism-related disorders. Schizophr Bull. 2015;41:313–4.Google Scholar
Corvin, A, Morris, DW. Genome-wide association studies: findings at the major histocompatibility complex locus in psychosis. Biol Psychiatry. 2014;75:276–83.Google Scholar
Miller, BJ, Buckley, P, Seabolt, W, Mellor, A, Kirkpatrick, B. Meta-analysis of cytokine alterations in schizophrenia: clinical status and antipsychotic effects. Biol Psychiatry. 2011;70:663–71.Google Scholar
Di Nicola, M, Cattaneo, A, Hepgul, N, et al. Serum and gene expression profile of cytokines in first-episode psychosis. Brain Behav Immun. 2013;31:90–5.Google Scholar
de Witte, L, Tomasik, J, Schwarz, E, et al. Cytokine alterations in first-episode schizophrenia patients before and after antipsychotic treatment. Schizophr Res. 2014;154:23–9.Google Scholar
Upthegrove, R, Manzanares-Teson, N, Barnes, NM. Cytokine function in medication-naive first episode psychosis: a systematic review and meta-analysis. Schizophr Res. 2014;155:101–8.Google Scholar
Cremaschi, L, Kardell, M, Johansson, V, et al. Prevalences of autoimmune diseases in schizophrenia, bipolar I and II disorder, and controls. Psychiatry Res. 2017;258:914.Google Scholar
Notter, T, Meyer, U. Microglia and schizophrenia: where next? Mol Psychiatry. 2017;22:788–9.Google Scholar
van Kesteren, CF, Gremmels, H, de Witte, LD, et al. Immune involvement in the pathogenesis of schizophrenia: a meta-analysis on postmortem brain studies. Transl Psychiatry. 2017;7:e1075.Google Scholar
Schwarcz, R, Hunter, CA. Toxoplasma gondii and schizophrenia: linkage through astrocyte-derived kynurenic acid? Schizophr Bull. 2007;33:652–3.Google Scholar
Prandovszky, E, Gaskell, E, Martin, H, et al. The neurotropic parasite Toxoplasma gondii increases dopamine metabolism. PLoS One. 2011;6:e23866.Google Scholar
Havlícek, J, Gasová, ZG, Smith, AP, et al. Decrease of psychomotor performance in subjects with latent ‘asymptomatic’ toxoplasmosis. Parasitology. 2001 May;122(Pt 5):515–20.Google Scholar
Molloy, MJ, Grainger, JR, Bouladoux, N, et al. Intraluminal containment of commensal outgrowth in the gut during infection-induced dysbiosis. Cell Host Microbe. 2013;14:318–28.Google Scholar
Arias, I, Sorlozano, A, Villegas, E, et al. Infectious agents associated with schizophrenia: a meta-analysis. Schizophr Res. 2012;136:128–36.Google Scholar
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;197:470–77.Google Scholar
Schwarz, E, Maukonen, J, Hyytiainen, T, Kieseppa, T, Oresic, M, Sabunciyan, S, et al. Analysis of microbiota in first episode psychosis identifies preliminary associations with symptom severity and treatment response. Schizophr Res. 2018;192:398403.Google Scholar
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:e309.Google Scholar
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;221:155–69.Google Scholar
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:e652.Google Scholar
Castro-Nallar, E, Bendall, ML, Perez-Losada, M, et al. Composition, taxonomy and functional diversity of the oropharynx microbiome in individuals with schizophrenia and controls. PeerJ. 2015;3:e1140.Google Scholar
Yolken, RH, Severance, EG, Sabunciyan, S, et al. Metagenomic sequencing indicates that the oropharyngeal phageome of individuals with schizophrenia differs from that of controls. Schizophr Bull. 2015;41:1153–61.Google Scholar
Chaves, C, Marque, CR, Trzesniak, C, et al. Glutamate-N-methyl-D-aspartate receptor modulation and minocycline for the treatment of patients with schizophrenia: an update. Braz J Med Biol Res. 2009;42(11):1002–14.Google Scholar
Solmi, M, Veronese, N, Thapa, N, et al. Systematic review and meta-analysis of the efficacy and safety of minocycline in schizophrenia. CNS Spectr. 2017;22:415–26.Google Scholar
Levkovitz, Y, Levi, U, Braw, Y, Cohen, H. 247 – Minocycline, a second-generation tetracycline, as a neuroprotective agent in an animal model of schizophrenia. Schizophrenia Research. 2008;98:135.Google Scholar
Zhang, L, Zhao, J. Profile of minocycline and its potential in the treatment of schizophrenia. Neuropsychiatr Dis Treat. 2014;10:1103–11.Google Scholar
Chaudhry, IB, Hallak, J, Husain, N, et al. Minocycline benefits negative symptoms in early schizophrenia: a randomised double-blind placebo-controlled clinical trial in patients on standard treatment. J Psychopharmacol. 2012;26:1185–93.Google Scholar
Liu, F, Guo, X, Wu, R, et al. Minocycline supplementation for treatment of negative symptoms in early-phase schizophrenia: a double blind, randomized, controlled trial. Schizophr Res. 2014;153:169–76.Google Scholar
Deakin, B, Suckling, J, Barnes, TRE, et al. The benefit of minocycline on negative symptoms of schizophrenia in patients with recent-onset psychosis (BeneMin): a randomised, double-blind, placebo-controlled trial. Lancet Psychiatry. 2018;5:885–94.Google Scholar
Dinan, TG, Stanton, C, Cryan, JF. Psychobiotics: a novel class of psychotropic. Biol Psychiatry. 2013;74:720–6.Google Scholar
Sarkar, A, Lehto, SM, Harty, S, et al. Psychobiotics and the manipulation of bacteria-gut-brain signals. Trends Neurosci. 2016;39:763–81.Google Scholar
Kang, DW, Adams, JB, Gregory, AC, et al. Microbiota transfer therapy alters gut ecosystem and improves gastrointestinal and autism symptoms: an open-label study. Microbiome. 2017;5:10.Google Scholar
Sandler, RH, Finegold, SM, Bolte, ER, et al. Short-term benefit from oral vancomycin treatment of regressive-onset autism. J Child Neurol. 2000;15(7):429–35.Google Scholar
Strati, F, Cavalieri, D, Albanese, D, et al. New evidences on the altered gut microbiota in autism spectrum disorders. Microbiome. 2017;5:24.Google Scholar
Gondalia, SV, Palombo, EA, Knowles, SR, et al. Molecular characterisation of gastrointestinal microbiota of children with autism (with and without gastrointestinal dysfunction) and their neurotypical siblings. Autism Res. 2012;5:419–27.Google Scholar
Cao, X, Lin, P, Jiang, P, Li, C. Characteristics of the gastrointestinal microbiome in children with autism spectrum disorder: a systematic review. Shanghai Arch Psychiatry. 2013;25:342–53.Google Scholar
Christian, LM, Galley, JD, Hade, EM, et al. Gut microbiome composition is associated with temperament during early childhood. Brain Behav Immun. 2015;45:118–27.Google Scholar
Tomova, A, Husarova, V, Lakatosova, S, et al. Gastrointestinal microbiota in children with autism in Slovakia. Physiol Behav. 2015;138:179–87.Google Scholar
Severance, EG, Yolken, RH, Eaton, WW. Autoimmune diseases, gastrointestinal disorders and the microbiome in schizophrenia: more than a gut feeling. Schizophr Res. 2016;176:2335.Google Scholar
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;62:41–5.Google Scholar

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×