Acessibilidade / Reportar erro

Gut microbiome in neuropsychiatric disorders

O microbioma intestinal nas doenças neuropsiquiátricas

ABSTRACT

Background:

Neuropsychiatric disorders are a significant cause of death and disability worldwide. The mechanisms underlying these disorders include a constellation of structural, infectious, immunological, metabolic, and genetic etiologies. Advances in next-generation sequencing techniques have demonstrated that the composition of the enteric microbiome is dynamic and plays a pivotal role in host homeostasis and several diseases. The enteric microbiome acts as a key mediator in neuronal signaling via metabolic, neuroimmune, and neuroendocrine pathways.

Objective:

In this review, we aim to present and discuss the most current knowledge regarding the putative influence of the gut microbiome in neuropsychiatric disorders.

Methods:

We examined some of the preclinical and clinical evidence and therapeutic strategies associated with the manipulation of the gut microbiome.

Results:

targeted taxa were described and grouped from major studies to each disease.

Conclusions:

Understanding the complexity of these ecological interactions and their association with susceptibility and progression of acute and chronic disorders could lead to novel diagnostic biomarkers based on molecular targets. Moreover, research on the microbiome can also improve some emerging treatment choices, such as fecal transplantation, personalized probiotics, and dietary interventions, which could be used to reduce the impact of specific neuropsychiatric disorders. We expect that this knowledge will help physicians caring for patients with neuropsychiatric disorders.

Keywords:
Gastrointestinal Microbiome; Metagenomics; Nervous System Diseases; Transplantation; Precision Medicine

RESUMO

Antecedentes:

Os transtornos neuropsiquiátricos são uma importante causa de morte e invalidez no mundo. Os mecanismos subjacentes a esses transtornos incluem uma constelação de etiologias estruturais, infecciosas, imunológicas, metabólicas e genéticas. Avanços nas técnicas de sequenciamento do DNA têm demonstrado que a composição do microbioma entérico é dinâmica e desempenha um papel fundamental não apenas na homeostase do hospedeiro, mas também em várias doenças. O microbioma entérico atua como mediador na sinalização das vias metabólica, neuroimune e neuroendócrina.

Objetivo:

Apresentar os estudos mais recentes sobre a possível influência do microbioma intestinal nas diversas doenças neuropsiquiátricas e discutir tanto os resultados quanto a eficácia dos tratamentos que envolvem a manipulação do microbioma intestinal.

Métodos:

foram examinadas algumas das evidências pré-clínicas e clínicas e estratégias terapêuticas associadas à manipulação do microbioma intestinal.

Resultados:

os táxons-alvo foram descritos e agrupados a partir dos principais estudos para cada doença.

Conclusões:

Entender a fundo a complexidade das interações ecológicas no intestino e sua associação com a suscetibilidade a certas doenças agudas e crônicas pode levar ao desenvolvimento de novos biomarcadores diagnósticos com base em alvos moleculares. Além disso, o estudo do microbioma intestinal pode auxiliar na otimização de tratamentos não farmacológicos emergentes, tais como o transplante de microbiota fecal, o uso de probióticos e intervenções nutricionais personalizadas. Dessa forma, terapias alternativas poderiam ser usadas para reduzir o impacto dos transtornos neuropsiquiátricos na saúde pública. Esperamos que esse conhecimento seja útil para médicos que cuidam de pacientes com diversos transtornos neuropsiquiátricos.

Palavras-chave:
Microbioma Gastrointestinal; Metagenômica; Doenças do Sistema Nervoso; Transplante; Medicina de Precisão

INTRODUCTION

Over the past decade, microbiomics have emerged as a new field led by advances in culture-independent methods and next-generation sequencing technologies. These methods have provided a broader understanding of how the interaction between microbes and humans can profoundly influence host homeostasis and different disease states11. Holmes E, Li JV, Athanasiou T, Ashrafian H, Nicholson JK. Understanding the role of gut microbiome - host metabolic signal disruption in health and disease. Trends Microbiol. 2011 Jul;19(7):349-59. https://doi.org/10.1016/j.tim.2011.05.006
https://doi.org/https://doi.org/10.1016/...
. The literature has reflected the growing number of studies on the putative influence of the microbiome on human health and disease (Figure 1). In humans, bacteria are the most prevalent domain, and it has been estimated that the ratio of microbes to human cells in an adult is nearly 1:122. Sender R, Fuchs S, Milo R. Revised estimates for the number of human and bacteria cells in the body. PLoS Biol. 2016 Aug;14(8):e1002533. https://doi.org/10.1371/journal.pbio.1002533
https://doi.org/https://doi.org/10.1371/...
. The human microbiome is defined as the compendium of microbial communities (including bacteria, archaea, viruses, protozoa, and fungi) living in a given body niche33. Turnbaugh PJ, Ley RE, Hamady M, Fraser-liggett CM, Knight R, Gordon JI. The Human Microbiome Project. Nature. 2007 Oct;449(7164):804-10. https://doi.org/10.1038/nature06244
https://doi.org/https://doi.org/10.1038/...
,44. Inda ME, Broset E, Lu TK, de la Fuente-Nunez C. Emerging frontiers in microbiome engineering. Trends Immunol. 2019 Oct;40(10):952-73. https://doi.org/10.1016/j.it.2019.08.007
https://doi.org/https://doi.org/10.1016/...
.

Figure 1.
Citations in PubMed related to microbiome research over the last decade.

It has been demonstrated that the gut microbiome can be shaped by prenatal conditions, lifestyle, aging, host genetics, antibiotic use, and even geography. Moreover, it has been shown that microbial aggregates on the large intestine can modulate a wide range of host physiological processes related to immune system maturation, food metabolism, energy production, and brain development55. Rhee SH, Pothoulakis C, Mayer EA. Principles and clinical implications of the brain-gut-enteric microbiota axis. Nat Rev Gastroenterol Hepatol. 2009 May;6(5):306-14. https://doi.org/10.1038/nrgastro.2009.35
https://doi.org/https://doi.org/10.1038/...
,66. Feng Q, Chen WD, Wang YD. Gut microbiota: An integral moderator in health and disease. Front Microbiol. 2018 Feb;9:151. https://doi.org/10.3389/fmicb.2018.00151
https://doi.org/https://doi.org/10.3389/...
.

The crosstalk between the gut microbiome and the brain is known as the gut microbiota-brain axis (MBA), which involves immunological, neuroendocrine, metabolic, and stress-response pathways77. Fung TC, Olson CA, Hsiao EY. Interactions between the microbiota, immune and nervous systems in health and disease. Nat Neurosci. 2017 Feb;20(2):145-55. https://doi.org/10.1038/nn.4476
https://doi.org/https://doi.org/10.1038/...
. This axis appears to be a cardinal mediator in a repertory of complex traits that range from metabolic to neuropsychiatric conditions88. Griffiths JA, Mazmanian SK. Emerging evidence linking the gut microbiome to neurologic disorders. Genome Med. 2018 Dec;10(1):98. https://doi.org/10.1186/s13073-018-0609-3
https://doi.org/https://doi.org/10.1186/...
. Despite the vast contribution of animal models to elucidate biological mechanisms underlying host-microbiota interactions, there are difficulties in translating the findings in animal experiments to human research. Also, differences in methodologic standards, metadata curation, and reference databases management (https://portal.hmpdacc.org/; https://www.sanger.ac.uk/resources/downloads/bacteria/metahit/) can be potential pitfalls for study comparison, interpretation, and reproducibility99. Allaband C, McDonald D, Vázquez-Baeza Y, Minich JJ, Tripathi A, Brenner DA, et al. Microbiome 101: studying, analyzing, and interpreting gut microbiome data for clinicians. Clin Gastroenterol Hepatol. 2019 Jan;17(2):218-30. https://doi.org/10.1016/j.cgh.2018.09.017
https://doi.org/https://doi.org/10.1016/...
.

Since cohorts in microbiome studies tend to be small and heterogeneous, it is difficult to know which observations are generalizable to larger patient populations. Metabolomic, proteomic, and big data analyses of microbiome function will be critical to determine if the conclusions of these studies can apply to the clinical setting. Further experimental studies using in vitro or in vivo models are needed to understand the direct impact and causal relationships between host and microbes in order to control both known and potential hidden confounders. In this way, it still not currently possible to determine whether changes in the microbiota could be merely an epiphenomenon rather than the primary cause of the problem. Thus, few clinicians currently recognize that the gut microbiome is relevant to human neurophysiology because the nature of the relationship between microbiota and disease is still unclear.

In this context, we aim to present and discuss the current literature on the putative biological significance of the gut microbiome in neuropsychiatric disorders. We hope to show why this field is emerging as a possible source of therapeutic targets in these disorders and how it can be incorporated into personalized medicine strategies44. Inda ME, Broset E, Lu TK, de la Fuente-Nunez C. Emerging frontiers in microbiome engineering. Trends Immunol. 2019 Oct;40(10):952-73. https://doi.org/10.1016/j.it.2019.08.007
https://doi.org/https://doi.org/10.1016/...
. We will describe only the major studies related to each disease; however, additional references are presented in Tables 1, 2, 3, 4, 5.

Table 1.
Summary of the main studies about gut microbiome in multiple sclerosis.
Table 2.
Summary of the main studies about gut microbiome in stroke.
Table 3.
Summary of main studies about gut microbiome and dementias.
Table 4.
Summary of the main studies about gut microbiome in epilepsy.
Table 5.
Summary of the main studies about gut microbiome in Parkinson’s disease.

GUT MICROBIOME STRUCTURE AND FUNCTION: INTESTINAL AND SYSTEMIC IMPACT

The gastrointestinal (GI) tract is considered the organ that harbors the highest amount of commensal microorganisms, reaching 101313. Cario E, Gerken G, Podolsky DK. Toll-like receptor 2 enhances ZO-1-associated intestinal epithelial barrier integrity via protein kinase C. Gastroenterology. 2004 Jul;127(1):224-38. https://doi.org/10.1053/j.gastro.2004.04.015
https://doi.org/https://doi.org/10.1053/...
bacteria/mL2. Bacteroidetes, Firmicutes, Actinobacteria, Verrumicrobia, Proteobacteria, and Fusobacteria have been identified as the core enterotypes at the phylum level, which consist of more than 1000 species33. Turnbaugh PJ, Ley RE, Hamady M, Fraser-liggett CM, Knight R, Gordon JI. The Human Microbiome Project. Nature. 2007 Oct;449(7164):804-10. https://doi.org/10.1038/nature06244
https://doi.org/https://doi.org/10.1038/...
. These consortia tend to remain steady throughout adult life, but preclinical evidence in both animals and humans have revealed that pre- and post-natal colonization of the gut has specific signatures and interindividual variations (~20%), depending on critical events such as maternal conditions, perinatal infections, mode of delivery, breastfeeding, diet, antibiotic exposure, and host genetics55. Rhee SH, Pothoulakis C, Mayer EA. Principles and clinical implications of the brain-gut-enteric microbiota axis. Nat Rev Gastroenterol Hepatol. 2009 May;6(5):306-14. https://doi.org/10.1038/nrgastro.2009.35
https://doi.org/https://doi.org/10.1038/...
,66. Feng Q, Chen WD, Wang YD. Gut microbiota: An integral moderator in health and disease. Front Microbiol. 2018 Feb;9:151. https://doi.org/10.3389/fmicb.2018.00151
https://doi.org/https://doi.org/10.3389/...
.

Different approaches comparing germ-free mice and conventionally colonized controls demonstrated that the abnormal composition of the gut microbiota led to functional and structural changes in the gastrointestinal tract. A variety of morphological defects on mucosa-associated lymphoid tissue and intestinal microvasculature, including reducing the number of Paneth cells, Peyer’s patches, and villi size, were already described in mice models1010. Stappenbeck TS, Hooper L V., Gordon JI. Developmental regulation of intestinal angiogenesis by indigenous microbes via Paneth cells. Proc Natl Acad Sci U S A. 2002 Nov;99(24):15451-5. https://doi.org/10.1073/pnas.202604299
https://doi.org/https://doi.org/10.1073/...
. Similarly, the loss of microbial diversity can negatively affect the expression of genes related to nutrient absorption, mucosal cellularity, and barrier fortification1111. Grenham S, Clarke G, Cryan JF, Dinan TG. Brain-gut-microbe communication in health and disease. Front Physiol. 2011 Oct;2(94):1-15. https://doi.org/10.3389/fphys.2011.00094
https://doi.org/https://doi.org/10.3389/...
. In vitro and in vivo studies have examined the importance of Toll-like receptors (TLRs) present on the surface of intestinal epithelial cells and immune T cells1212. Rakoff-Nahoum S, Paglino J, Eslami-Varzaneh F, Edberg S, Medzhitov R. Recognition of commensal microflora by toll-like receptors is required for intestinal homeostasis. Cell. 2004 Jul;118(2):229-41. https://doi.org/10.1016/j.cell.2004.07.002
https://doi.org/https://doi.org/10.1016/...
. The signaling transmission mediated by TLRs can also modulate mechanisms encompassing the host’s tolerance to commensal bacteria and the inhibition of pro-inflammatory cytokines such as the tumor necrosis factor (TNF), interleukin-6 (IL-6), and interleukin-1B (IL-1B)1313. Cario E, Gerken G, Podolsky DK. Toll-like receptor 2 enhances ZO-1-associated intestinal epithelial barrier integrity via protein kinase C. Gastroenterology. 2004 Jul;127(1):224-38. https://doi.org/10.1053/j.gastro.2004.04.015
https://doi.org/https://doi.org/10.1053/...
. These findings have been classically associated with several GI disorders, including inflammatory bowel disease (IBD), Crohn’s disease (CD), and ulcerative colitis (UC), as well as irritable bowel syndrome (IBS), functional dyspepsia (FD), and colorectal cancer (CRC)1414. Wang Y, Kasper LH. The role of microbiome in central nervous system disorders. Brain Behav Immun. 2014 May;38:1-12. https://doi.org/10.1016/j.bbi.2013.12.015
https://doi.org/https://doi.org/10.1016/...
,1515. Powell N, Walker MM, Talley NJ. The mucosal immune system: master regulator of bidirectional gut-brain communications. Nat Rev Gastroenterol Hepatol. 2017 Mar;14(3):143-59. https://doi.org/10.1038/nrgastro.2016.191
https://doi.org/https://doi.org/10.1038/...
. Besides, observational studies have indicated that the loss of beneficial microorganisms affects ecological interactions among local populations and drive systemic diseases. Some of the metabolic processes that are impaired in patients with obesity, diabetes mellitus, and non-alcoholic fatty liver disease are associated with the synthesis of vitamins, xenobiotic transformation, and bacteria-derived molecules production, including secondary bile acids and short-chain fatty acids (SCFA) production1616. Chen X, D’Souza R, Hong ST. The role of gut microbiota in the gut-brain axis: Current challenges and perspectives. Protein Cell. 2013 Jun;4(6):403-14. https://doi.org/10.1007/s13238-013-3017-x
https://doi.org/https://doi.org/10.1007/...
,1717. Martin CR, Osadchiy V, Kalani A, Mayer EA. The brain-gut-microbiome axis. Cell Mol Gastroenterol Hepatol. 2018 Apr;6(2):133-48. https://doi.org/10.1016/j.jcmgh.2018.04.003
https://doi.org/https://doi.org/10.1016/...
.

INTESTINAL MICROBIOTA AFFECTS BRAIN FUNCTION AND HAS IMMUNOMODULATORY PROPERTIES

The human gut contains its own neural system, consisting of more than one hundred million neuron cells (108)1818. Calvani R, Picca A, Rita M, Landi F, Bernabei R, Marzetti E. Of microbes and minds: a narrative review on the second brain aging. Front Med (Lausanne). 2018 Mar;5:53. https://doi.org/10.3389/fmed.2018.00053
https://doi.org/https://doi.org/10.3389/...
. Crosstalk between the central nervous system (CNS) and the gut microbiome is known as the gut microbiota-brain axis, and it is orchestrated at different anatomical levels. It occurs through an intricate network of afferent and efferent circuits alongside the vagus nerve, enteric nervous system, and hypothalamic-pituitary-adrenal axis. Thus, the gut connectome integrates neuroendocrine, enteroendocrine, neuroimmune, and metabolic signaling pathways responsible for regulating functions associated with digestion, tract motility, and brain development. Furthermore, gut microbes can control host responses to vascular injury and autoimmunity by modifications in both the blood-brain barrier (BBB) and brain lymphatic system77. Fung TC, Olson CA, Hsiao EY. Interactions between the microbiota, immune and nervous systems in health and disease. Nat Neurosci. 2017 Feb;20(2):145-55. https://doi.org/10.1038/nn.4476
https://doi.org/https://doi.org/10.1038/...
,1515. Powell N, Walker MM, Talley NJ. The mucosal immune system: master regulator of bidirectional gut-brain communications. Nat Rev Gastroenterol Hepatol. 2017 Mar;14(3):143-59. https://doi.org/10.1038/nrgastro.2016.191
https://doi.org/https://doi.org/10.1038/...
.

Additionally, evidence from intervention studies in germ-free mice, where a diverse microbiota is absent, has also shown the great potential that microorganisms have in regulating microglia differentiation and maturation. Other features commonly displayed in gnotobiotic models are the abnormal expression of proteins related to synaptic plasticity, such as the brain-derived neurotrophic factor and the impairment in global cognition responses. For instance, in a study carried out by Möhle et al., the authors pointed out that adult hippocampal neurogenesis and global cognition responses can be restored after oral supplementation with probiotics via expansion of Ly6C+ monocytes1919. Möhle L, Mattei D, Heimesaat MM, Bereswill S, Fischer A, Alutis M, et al. Ly6Chi monocytes provide a link between antibiotic-induced changes in gut microbiota and adult hippocampal neurogenesis. Cell Rep. 2016 May;15(9):1945-56. https://doi.org/10.1016/j.celrep.2016.04.074
https://doi.org/https://doi.org/10.1016/...
,2020. Erny D, De Angelis ALH, Jaitin D, Wieghofer P, Staszewski O, David E, et al. Host microbiota constantly control maturation and function of microglia in the CNS. Nat Neurosci. 2015 Jul;18(7):965-77. https://doi.org/10.1038/nn.4030
https://doi.org/https://doi.org/10.1038/...
. Notably, it has been reported that microbial metabolites such as SCFAs, secondary bile acids, precursors of the lipid biosynthesis (propionate), and specific amino acids (tryptophan) are critical in modulating the release of host cytokines and hormones such as peptide YY, vasoactive intestinal peptide, glucagon-like peptide-1 (GLP-1), and melatonin, as well as neurotransmitters such as serotonin (5-HT), catecholamines, and gamma-aminobutyric acid (GABA). All of these affect brain functions related to feeding, stress response, emotional behaviors, aging, and cognition1717. Martin CR, Osadchiy V, Kalani A, Mayer EA. The brain-gut-microbiome axis. Cell Mol Gastroenterol Hepatol. 2018 Apr;6(2):133-48. https://doi.org/10.1016/j.jcmgh.2018.04.003
https://doi.org/https://doi.org/10.1016/...
,2121. Dinan TG, Cryan JF. The microbiome-gut-brain axis in health and disease. Gastroenterol Clin North Am. 2017 Mar;46(1):77-89. https://doi.org/10.1016/j.gtc.2016.09.007
https://doi.org/https://doi.org/10.1016/...
. Furthermore, intestinal dysbiosis appears to be linked to the development of brain autoimmunity driven by specific T cell subtypes, pro-inflammatory cytokines, endothelial barrier impairment, and neurodegeneration77. Fung TC, Olson CA, Hsiao EY. Interactions between the microbiota, immune and nervous systems in health and disease. Nat Neurosci. 2017 Feb;20(2):145-55. https://doi.org/10.1038/nn.4476
https://doi.org/https://doi.org/10.1038/...
,2222. Colpitts SL, Kasper LH. Influence of the gut microbiome on autoimmunity in the central nervous system. J Immunol. 2017 Jan;198(2):596-604. https://doi.org/10.4049/jimmunol.1601438
https://doi.org/https://doi.org/10.4049/...
. The next section will explore some of the neuropsychiatric disorders thought to be influenced by microbiome disturbances.

MULTIPLE SCLEROSIS

Previous studies have linked T helper 17 cells (Th17) to MS pathogenesis through its effect in exacerbating experimental autoimmune encephalomyelitis (EAE). Regulatory T cells (Treg) have an essential role in suppressing inflammation in the CNS in EAE models2222. Colpitts SL, Kasper LH. Influence of the gut microbiome on autoimmunity in the central nervous system. J Immunol. 2017 Jan;198(2):596-604. https://doi.org/10.4049/jimmunol.1601438
https://doi.org/https://doi.org/10.4049/...
,2323. Hughes LE, Smith PA, Bonell S, Natt RS, Wilson C, Rashid T, et al. Cross-reactivity between related sequences found in Acinetobacter sp., Pseudomonas aeruginosa, myelin basic protein and myelin oligodendrocyte glycoprotein in multiple sclerosis. J Neuroimmunol. 2003 Nov;144(1-2):105-15. https://doi.org/10.1016/s0165-5728(03)00274-1
https://doi.org/https://doi.org/10.1016/...
. Cekanaviciute et al. have identified increased EAE disease scores and deficient IL-10+ Treg induction in mice colonized with microbiota from patients with MS2424. Cekanaviciute E, Yoo BB, Runia TF, Debelius JW, Singh S, Nelson CA, et al. Gut bacteria from multiple sclerosis patients modulate human T cells and exacerbate symptoms in mouse models. Proc Natl Acad Sci U S A. 2017 Oct;114(40):10713-8. https://doi.org/10.1073/pnas.1711235114
https://doi.org/https://doi.org/10.1073/...
. Furthermore, some butyrate-producing bacteria, mainly belonging to the Firmicutes phylum, have also been implicated in the pathogenesis of MS. Butyrate is known to inhibit pro-inflammatory pathways and prevent systemic exposure to intestinal antigens2525. Canani RB, Costanzo M Di, Leone L, Pedata M, Meli R, Calignano A. Potential beneficial effects of butyrate in intestinal and extraintestinal diseases. World J Gastroenterol. 2011 Mar;17(12):1519-28. https://doi.org/10.3748/wjg.v17.i12.1519
https://doi.org/https://doi.org/10.3748/...
. Bacteria, such as Faecalibacterium from the Firmicutes phylum, were found to be reduced in MS patients2626. Cantarel BL, Waubant E, Chehoud C, Kuczynski J, Desantis TZ, Warrington J, et al. Gut microbiota in multiple sclerosis: Possible influence of immunomodulators. J Investig Med. 2015 Jun;63(5):729-34. https://doi.org/10.1097/JIM.0000000000000192
https://doi.org/https://doi.org/10.1097/...
.

In a study of adult patients, Jangi et al. found a higher abundance of Methanobrevibacter and Akkermansia with a lower prevalence of Prevotella, Butyricimonas, Colinsella, and Slackia in patients with MS compared to healthy controls. Combining microbiome results with the immunogenetics characteristics of patients with MS, they found a positive correlation between Methanobrevibacter and Akkermansia and a negative correlation of Butyricimonas with MAPK14, MAPK1, LTBR, STAT5B, CASP1, and HLA-DRB1 - genes associated with potentiation of the immune response in MS2727. Jangi S, Gandhi R, Cox LM, Li N, Von Glehn F, Yan R, et al. Alterations of the human gut microbiome in multiple sclerosis. Nat Commun. 2016 Jun;7:1-11. https://doi.org/10.1038/ncomms12015
https://doi.org/https://doi.org/10.1038/...
.

Tremlett et al. studied the microbiome in pediatric MS. A phylum-level analysis found a negative association between Bacteroidetes and CD4+ T cells and Tregs and a positive association between Actinobacteria and CD4+ T cells and Tr1 (IL-10), which represent some of the most common bacterial phyla of the human microbiota. The evenness of the gut microbiome also had a strong and negative association with Th17 and T helper 2 (Th2) response in the control group2828. Tremlett H, Fadrosh DW, Faruqi AA, Hart J, Roalstad S, Graves J, et al. Gut microbiota composition and relapse risk in pediatric MS: A pilot study. J Neurol Sci. 2016 Apr;363:153-7. https://doi.org/10.1016/j.jns.2016.02.042
https://doi.org/https://doi.org/10.1016/...
.

More recently, the interaction between microbiome and diet in MS has attracted attention. A study found an association between a lower number of relapses and a lower disability status scale after one year of a high vegetable/low protein diet (HV/LP diet) when compared with a classical Western diet characterized by regular consumption of red meat, processed meat, refined grains, sweetened food, salt, and saturated and omega-6 fatty acids. The HV/LP group had higher levels of bacteria from the Lachnospiraceae genus, and they were positively related to Treg cells2929. Saresella M, Mendozzi L, Rossi V, Mazzali F, Piancone F, LaRosa F, et al. Immunological and clinical effect of diet modulation of the gut microbiome in multiple sclerosis patients: A pilot study. Front Immunol. 2017 Oct;8:1-11. https://doi.org/10.3389/fimmu.2017.01391
https://doi.org/https://doi.org/10.3389/...
. Table 1 summarizes the most relevant microbiome findings in patients with MS.

AUTOIMMUNE ENCEPHALITIS

Immuno-mediated encephalitis is an emergent group of syndromes characterized by the development of acute or subacute progressive encephalopathy (less than three months onset) that occurs due to an abnormal antibody response against cell-surface, intracellular synaptic, or intraneuronal (nuclear or cytoplasmic) antigens3030. Graus F, Titulaer MJ, Balu R, Benseler S, Bien CG, Cellucci T, et al. A clinical approach to diagnosis of autoimmune encephalitis. Lancet Neurol. 2016 Apr;15(4):391-404. https://doi.org/10.1016/S1474-4422(15)00401-9
https://doi.org/https://doi.org/10.1016/...
,3131. Lancaster E, Dalmau J. Neuronal autoantigens-pathogenesis, associated disorders and antibody testing. Nat Rev Neurol. 2012 Jun;8(7):380-90. https://doi.org/10.1038/nrneurol.2012.99
https://doi.org/https://doi.org/10.1038/...
.

Most of what is known about the impact of intestinal dysbiosis in patients with autoimmune encephalitis (AE) comes from studies conducted in rodents. It has been suggested that changes in the intestinal microbiota could increase the susceptibility to AE through different mechanisms. Thus, increases in abundance and richness of specific pathobionts can provoke pro-inflammatory T cell cross-reactivity due to molecular similarities with neural proteins2222. Colpitts SL, Kasper LH. Influence of the gut microbiome on autoimmunity in the central nervous system. J Immunol. 2017 Jan;198(2):596-604. https://doi.org/10.4049/jimmunol.1601438
https://doi.org/https://doi.org/10.4049/...
. Microbiome products of diet fermentation such as the SCFAs have a major impact on gene expression of transcription factors via epigenetic mechanisms. Indeed, in a case-control study conducted by Gong et al., fecal samples were examined in 30 patients at different phases of anti- N-methyl-D-aspartate receptor (NMDAR) encephalitis. Patients in the acute phase had low Firmicutes to Bacteroidetes (F/B) ratios than the control group3232. Gong X, Liu X, Li C, Chen C, Lin J, Li A, et al. Alterations in the human gut microbiome in anti-N-methyl-D-aspartate receptor encephalitis. Ann Clin Transl Neurol. 2019 Sep;6(9):1771-81. https://doi.org/10.1002/acn3.50874
https://doi.org/https://doi.org/10.1002/...
. These results support the theory that an imbalance in commensal microbes negatively impacts the production of SCFAs. Also, main SCFAs such as acetate (C2), propionate (C3), and butyrate (C4) can alter a variety of cellular mechanisms involving the activity of G-protein coupled receptors (GPRs), the inhibition of histones deacetylases (HDACs), and nuclear factor-jB (NF-jB), and the biosynthesis of retinoic acid, which are all essential in maintaining Treg differentiation and hence reducing neuroinflammation3333. Haase S, Haghikia A, Wilck N, Müller DN, Linker RA. Impacts of microbiome metabolites on immune regulation and autoimmunity. Immunology. 2018 Jun;154(2):230-8. https://doi.org/10.1111/imm.12933
https://doi.org/https://doi.org/10.1111/...
,3434. Silva YP, Bernardi A, Frozza RL. The role of short-chain fatty acids from gut microbiota in gut-brain communication. Front Endocrinol (Lausanne). 2020 Jan;11:1-14. https://doi.org/10.3389/fendo.2020.00025
https://doi.org/https://doi.org/10.3389/...
.

Enrichment of the genus Fusobacterium was also reported in the anti-NMDAR AE. Fusobacteria species comprise gram-negative anaerobic bacilli, which are considered normal microbiota in oral cavity, GI, and urogenital tract. However, recent studies report that F. nucleatum can present pathogenic properties that have been implicated in oral and extraoral diseases, including neurological disorders3232. Gong X, Liu X, Li C, Chen C, Lin J, Li A, et al. Alterations in the human gut microbiome in anti-N-methyl-D-aspartate receptor encephalitis. Ann Clin Transl Neurol. 2019 Sep;6(9):1771-81. https://doi.org/10.1002/acn3.50874
https://doi.org/https://doi.org/10.1002/...
. It is believed that adhesion and invasion via hematogenous translocation are the primary strategies used by F. nucleatum to activate inflammatory and oncogenic genes, thus contributing to disease development and progression3535. Brennan CA, Garrett WS. Fusobacterium nucleatum - symbiont, opportunist and oncobacterium. Nat Rev Microbiol. 2019 Mar;17(3):156-66. https://doi.org/10.1038/s41579-018-0129-6
https://doi.org/https://doi.org/10.1038/...
.

MICROBIOME AND STROKE

Studies have shown that up to 50% of patients with stroke suffer from GI complications, which has a strong association with patient recovery, deterioration of neurological functions, and mortality3636. Wen SW, Wong CHY. An unexplored brain-gut microbiota axis in stroke. Gut Microbes. 2017 Nov;8(6):601-6. https://doi.org/10.1080/19490976.2017.1344809
https://doi.org/https://doi.org/10.1080/...
. These features regarding stroke make it an interesting condition to look for associations between brain and gut microbiota. Indeed, several studies have shown a link between stroke outcomes and microbiota regulation of the immune system and metabolism3737. Org E, Blum Y, Kasela S, Mehrabian M, Kuusisto J, Kangas AJ, et al. Relationships between gut microbiota, plasma metabolites, and metabolic syndrome traits in the METSIM cohort. Genome Biol. 2017 Apr;18(1):70. https://doi.org/10.1186/s13059-017-1194-2
https://doi.org/https://doi.org/10.1186/...
,3838. Haak BW, Westendorp WF, van Engelen TSR, Brands X, Brouwer MC, Vermeij JD, et al. Disruptions of anaerobic gut bacteria are associated with stroke and post-stroke infection: a prospective case-control study. Transl Stroke Res. 2020 Oct;1-12. https://doi.org/10.1007/s12975-020-00863-4
https://doi.org/https://doi.org/10.1007/...
,3939. Li N, Weng X, Sun C, Wu X, Lu M, Si Y, et al. Change of intestinal microbiota in cerebral ischemic stroke patients. BMC Microbiol. 2019 Aug;19(1):1-8. https://doi.org/10.1186/s12866-019-1552-1
https://doi.org/https://doi.org/10.1186/...
(Table 2).

The bidirectional communication between the brain and gut after stroke involves the vagus nerve, release of damage-associated molecular patterns (DAMPs), cytokines from the injury site of the brain and gut, and migration of inflammatory or immune cells from the gut to the injury site4040. Arya AK, Hu B. Brain-gut axis after stroke. Brain Circ. 2018 Oct-Dec;4(4):165-73. https://doi.org/10.4103/bc.bc_32_18
https://doi.org/https://doi.org/10.4103/...
. This communication occurs by complex signaling pathways from the vagus nerve to the enteric nervous system, the neuronal-glial-endothelial interactions, and DAMPs and cytokines-induced activation of gut inflammatory and immune cells4040. Arya AK, Hu B. Brain-gut axis after stroke. Brain Circ. 2018 Oct-Dec;4(4):165-73. https://doi.org/10.4103/bc.bc_32_18
https://doi.org/https://doi.org/10.4103/...
.

Several studies show significant changes in the microbial diversity in fecal samples of patients after an ischemic stroke, leading to gut alterations including dysbiosis, dysmotility, hemorrhage, and sepsis4040. Arya AK, Hu B. Brain-gut axis after stroke. Brain Circ. 2018 Oct-Dec;4(4):165-73. https://doi.org/10.4103/bc.bc_32_18
https://doi.org/https://doi.org/10.4103/...
,4141. Karlsson FH, Fåk F, Nookaew I, Tremaroli V, Fagerberg B, Petranovic D, et al. Symptomatic atherosclerosis is associated with an altered gut metagenome. Nat Commun. 2012 Dec;3(1):1245. https://doi.org/10.1038/ncomms2266
https://doi.org/https://doi.org/10.1038/...
. Furthermore, Singh et al. showed a reduction in microbiota diversity after stroke events that might be associated with stress response, impaired motility, and tissue necrosis4242. Singh V, Roth S, Llovera G, Sadler R, Garzetti D, Stecher B, et al. Microbiota dysbiosis controls the neuroinflammatory response after stroke. J Neurosci. 2016 Jul;36(28):7428-40. https://doi.org/10.1523/JNEUROSCI.1114-16.2016
https://doi.org/https://doi.org/10.1523/...
. These changes may cause gut permeability modifications and the increase of circulating lipopolysaccharides (LPS) molecules, which may influence systemic inflammation and immune response after stroke.

Circulating IL-17 released by γδ T cells and IL-10 released by regulatory T cells (Tregs) were associated with increased ischemic brain injury and neuroprotective properties, respectively, after ischemic brain injury4343. Benakis C, Brea D, Caballero S, Faraco G, Moore J, Murphy M, et al. Commensal microbiota affects ischemic stroke outcome by regulating intestinal γδ T cells. Nat Med. 2016 May;22(5):516-23. https://doi.org/10.1038/nm.4068
https://doi.org/https://doi.org/10.1038/...
. There is some evidence that these inflammatory molecules are regulated by gut microbiota, promoting a strong interrelation between brain and gut and influencing several neurological diseases4040. Arya AK, Hu B. Brain-gut axis after stroke. Brain Circ. 2018 Oct-Dec;4(4):165-73. https://doi.org/10.4103/bc.bc_32_18
https://doi.org/https://doi.org/10.4103/...
,4343. Benakis C, Brea D, Caballero S, Faraco G, Moore J, Murphy M, et al. Commensal microbiota affects ischemic stroke outcome by regulating intestinal γδ T cells. Nat Med. 2016 May;22(5):516-23. https://doi.org/10.1038/nm.4068
https://doi.org/https://doi.org/10.1038/...
. After a stroke, dysbiosis leads to an imbalance of T-cell subpopulations (Th1, Th2, Th9, Th17, Treg, and follicular T helper cells) that trigger several types of autoimmune and inflammatory disease4040. Arya AK, Hu B. Brain-gut axis after stroke. Brain Circ. 2018 Oct-Dec;4(4):165-73. https://doi.org/10.4103/bc.bc_32_18
https://doi.org/https://doi.org/10.4103/...
. For example, Th1 (production of IL-2 and interferon-gamma) and Th2 (IL-4, IL-5, and IL-13) induce inflammation; Th9 (IL-9 and TGF-β) and Treg (IL-10 and IL-35) have a neuroprotective function; Th17 activates matrix metalloproteinases and causes blood-brain barrier breakdown by secreting IL-17A, IL17-F, and IL-224040. Arya AK, Hu B. Brain-gut axis after stroke. Brain Circ. 2018 Oct-Dec;4(4):165-73. https://doi.org/10.4103/bc.bc_32_18
https://doi.org/https://doi.org/10.4103/...
. Benakis et al. demonstrated that antibiotic-treatment-induced dysbiosis could influence stroke outcome in vivo models by regulating T cells in the small intestine4343. Benakis C, Brea D, Caballero S, Faraco G, Moore J, Murphy M, et al. Commensal microbiota affects ischemic stroke outcome by regulating intestinal γδ T cells. Nat Med. 2016 May;22(5):516-23. https://doi.org/10.1038/nm.4068
https://doi.org/https://doi.org/10.1038/...
. Some studies also observed improved stroke outcomes by fecal microbiota transplantation to control post-stroke dysbiosis4343. Benakis C, Brea D, Caballero S, Faraco G, Moore J, Murphy M, et al. Commensal microbiota affects ischemic stroke outcome by regulating intestinal γδ T cells. Nat Med. 2016 May;22(5):516-23. https://doi.org/10.1038/nm.4068
https://doi.org/https://doi.org/10.1038/...
. Most interestingly, treatments involving reduction of Clostridiaceae and S24-7 spp. showed to be relevant to neuroprotection after stroke in mice4343. Benakis C, Brea D, Caballero S, Faraco G, Moore J, Murphy M, et al. Commensal microbiota affects ischemic stroke outcome by regulating intestinal γδ T cells. Nat Med. 2016 May;22(5):516-23. https://doi.org/10.1038/nm.4068
https://doi.org/https://doi.org/10.1038/...
.

Zeng et al. recently raised the possibility that the microbiome might be a novel risk factor for stroke4444. Zeng X, Gao X, Peng Y, Wu Q, Zhu J, Tan C, et al. Higher risk of stroke is correlated with increased opportunistic pathogen load and reduced levels of butyrate-producing bacteria in the gut. Front Cell Infect Microbiol. 2019 Feb;9:4. https://doi.org/10.3389/fcimb.2019.00004
https://doi.org/https://doi.org/10.3389/...
. With a risk stratification approach and comparing higher- versus lower-risk patients, they found an increased risk of stroke associated with enrichment of opportunistic pathogens (e.g., Proteobacteria, Bacilli, Enterobacteriaceae), low abundance of butyrate-producing bacteria (e.g., Lachnospiraceae, Ruminococcaceae), and reduced concentrations of fecal butyrate4444. Zeng X, Gao X, Peng Y, Wu Q, Zhu J, Tan C, et al. Higher risk of stroke is correlated with increased opportunistic pathogen load and reduced levels of butyrate-producing bacteria in the gut. Front Cell Infect Microbiol. 2019 Feb;9:4. https://doi.org/10.3389/fcimb.2019.00004
https://doi.org/https://doi.org/10.3389/...
,4545. Yin J, Liao SX, He Y, Wang S, Xia GH, Liu FT, et al. Dysbiosis of gut microbiota with reduced trimethylamine-n-oxide level in patients with large-artery atherosclerotic stroke or transient ischemic attack. J Am Heart Assoc. 2015 Nov;4(11):e002699. https://doi.org/10.1161/JAHA.115.002699
https://doi.org/https://doi.org/10.1161/...
.

Xia et al. developed a stroke dysbiosis index (SDI) based on the patient’s gut taxonomic differences compared to healthy individuals4646. Xia GH, You C, Gao XX, Zeng XL, Zhu JJ, Xu KY, et al. Stroke dysbiosis index (SDI) in gut microbiome are associated with brain injury and prognosis of stroke. Front Neurol. 2019 Apr;10:397. https://doi.org/10.3389/fneur.2019.00397
https://doi.org/https://doi.org/10.3389/...
. They observed that high SDI values predicted severe brain injury in patients with stroke. Furthermore, to investigate a putative causal effect of intestinal dysbiosis, the authors performed experiments using a middle cerebral artery occlusion model in animals colonized with microbiota from affected individuals. They observed that mice transplanted with intestinal microbiota from high-SDI patients also developed an exacerbated inflammatory response, hence, worsening the acute brain injury associated with stroke4646. Xia GH, You C, Gao XX, Zeng XL, Zhu JJ, Xu KY, et al. Stroke dysbiosis index (SDI) in gut microbiome are associated with brain injury and prognosis of stroke. Front Neurol. 2019 Apr;10:397. https://doi.org/10.3389/fneur.2019.00397
https://doi.org/https://doi.org/10.3389/...
.

Although the results are encouraging, as described above, additional studies with larger samples and different ethnic backgrounds are needed to validate these findings. However, if further confirmed, a careful approach, including microbiome screening as a possible preventive target for stroke management is needed.

GUT MICROBIOTA: THE RELATIONSHIP WITH DEMENTIA

Studies in germ-free animals exposed to microbial infections, human post-mortem brain samples, and microbiome analysis of living humans have revealed that disorder of the gut microbiota may underlie the development or exacerbation of Alzheimer’s disease (AD) pathology4747. Kowalski K, Mulak A. Brain-gut-microbiota axis in Alzheimer’s disease. J Neurogastroenterol Motil. 2019 Jan;25(1):48-60. https://doi.org/10.5056/jnm18087
https://doi.org/https://doi.org/10.5056/...
. Also, available data suggest that the gut microbiota in AD is characterized by a substantial reduction in beneficial microbial diversity and presence of pathogenic species such as Proteobacteria phylum, especially the Enterobacteriaceae family4848. Welcome MO. Gut microbiota disorder, gut epithelial and blood-brain barrier dysfunctions in etiopathogenesis of dementia: molecular mechanisms and signaling pathways. Neuromolecular Med. 2019 Sep;21(3):205-26. https://doi.org/10.1007/s12017-019-08547-5.
https://doi.org/https://doi.org/10.1007/...
. These shifts in microbial diversity may activate immune cells and stimulate overproduction of toxic metabolites or pro-inflammatory cytokines, which contribute to the destruction of the GI mucosa. It is well known that chronic inflammation and immune dysregulation precede cognitive decline by years4949. Fox M, Knorr DA, Haptonstall KM. Alzheimer’s disease and symbiotic microbiota: an evolutionary medicine perspective. Ann N Y Acad Sci. 2019 Aug;1449(1):3-24. https://doi.org/10.1111/nyas.14129
https://doi.org/https://doi.org/10.1111/...
. Increased inflammation makes gut microorganisms move from the GI tract through cells overlying the Peyer’s patches into blood and other tissues (a process known as atopobiosis)4747. Kowalski K, Mulak A. Brain-gut-microbiota axis in Alzheimer’s disease. J Neurogastroenterol Motil. 2019 Jan;25(1):48-60. https://doi.org/10.5056/jnm18087
https://doi.org/https://doi.org/10.5056/...
. Likewise, systemic inflammation can increase BBB permeability, exposing the brain to cytokines that can lead to neuroinflammation and neuronal cell death, promoting neurodegenerative diseases4848. Welcome MO. Gut microbiota disorder, gut epithelial and blood-brain barrier dysfunctions in etiopathogenesis of dementia: molecular mechanisms and signaling pathways. Neuromolecular Med. 2019 Sep;21(3):205-26. https://doi.org/10.1007/s12017-019-08547-5.
https://doi.org/https://doi.org/10.1007/...
,5050. Sochocka M, Donskow-Łysoniewska K, Diniz BS, Kurpas D, Brzozowska E, Leszek J. The gut microbiome alterations and inflammation-driven pathogenesis of Alzheimer’s disease-a critical review. Mol Neurobiol. 2019 Mar;56(3):1841-51. https://doi.org/10.1007/s12035-018-1188-4
https://doi.org/https://doi.org/10.1007/...
. Initially, the brain can resist, but the regenerative capacity, together with the microglia’s ability to clear toxic metabolites, decreases with time4949. Fox M, Knorr DA, Haptonstall KM. Alzheimer’s disease and symbiotic microbiota: an evolutionary medicine perspective. Ann N Y Acad Sci. 2019 Aug;1449(1):3-24. https://doi.org/10.1111/nyas.14129
https://doi.org/https://doi.org/10.1111/...
.

The outer membrane component of Gram-negative bacteria is LPS, capable of triggering systemic inflammation by increasing pro-inflammatory cytokines4848. Welcome MO. Gut microbiota disorder, gut epithelial and blood-brain barrier dysfunctions in etiopathogenesis of dementia: molecular mechanisms and signaling pathways. Neuromolecular Med. 2019 Sep;21(3):205-26. https://doi.org/10.1007/s12017-019-08547-5.
https://doi.org/https://doi.org/10.1007/...
,5151. Pistollato F, Cano SS, Elio I, Vergara MM, Giampieri F, Battino M. Role of gut microbiota and nutrients in amyloid formation and pathogenesis of Alzheimer disease. Nutr Rev. 2016 Oct;74(10):624-34. https://doi.org/10.1093/nutrit/nuw023
https://doi.org/https://doi.org/10.1093/...
. Lipopolysaccharides may also modify gut homeostasis and promote gut inflammation and permeability. The abundance of Gram-negative intestinal bacteria, such as the Enterobacteriaceae family in individuals with AD, results in increased translocation of LPS from the gut into the circulation, which in turn may contribute to AD pathology5252. Vogt NM, Kerby RL, Dill-McFarland KA, Harding SJ, Merluzzi AP, Johnson SC, et al. Gut microbiome alterations in Alzheimer’s disease. Sci Rep. 2017 Oct;7(1):13537. https://doi.org/10.1038/s41598-017-13601-y
https://doi.org/https://doi.org/10.1038/...
. Indeed, a study involving post-mortem brain tissue from patients with AD showed that LPS fragments co-localized with amyloid plaques in the hippocampus and neocortex5353. Liu P, Wu L, Peng G, Han Y, Tang R, Ge J, et al. Altered microbiomes distinguish Alzheimer’s disease from amnestic mild cognitive impairment and health in a Chinese cohort. Brain Behav Immun. 2019 Aug;80:633-43. https://doi.org/10.1016/j.bbi.2019.05.008
https://doi.org/https://doi.org/10.1016/...
.

Surprisingly, bacteria can produce their own amyloid, which maintains cellular junctions, promotes the formation of biofilms, and confers resistance against physical or immune destruction4747. Kowalski K, Mulak A. Brain-gut-microbiota axis in Alzheimer’s disease. J Neurogastroenterol Motil. 2019 Jan;25(1):48-60. https://doi.org/10.5056/jnm18087
https://doi.org/https://doi.org/10.5056/...
. Microbial and cerebral amyloids are structurally similar and can be recognized by the same TLR2/TLR1 receptor system5050. Sochocka M, Donskow-Łysoniewska K, Diniz BS, Kurpas D, Brzozowska E, Leszek J. The gut microbiome alterations and inflammation-driven pathogenesis of Alzheimer’s disease-a critical review. Mol Neurobiol. 2019 Mar;56(3):1841-51. https://doi.org/10.1007/s12035-018-1188-4
https://doi.org/https://doi.org/10.1007/...
. They might activate signaling pathways known to play a role in neurodegeneration and AD pathogenesis5050. Sochocka M, Donskow-Łysoniewska K, Diniz BS, Kurpas D, Brzozowska E, Leszek J. The gut microbiome alterations and inflammation-driven pathogenesis of Alzheimer’s disease-a critical review. Mol Neurobiol. 2019 Mar;56(3):1841-51. https://doi.org/10.1007/s12035-018-1188-4
https://doi.org/https://doi.org/10.1007/...
,5151. Pistollato F, Cano SS, Elio I, Vergara MM, Giampieri F, Battino M. Role of gut microbiota and nutrients in amyloid formation and pathogenesis of Alzheimer disease. Nutr Rev. 2016 Oct;74(10):624-34. https://doi.org/10.1093/nutrit/nuw023
https://doi.org/https://doi.org/10.1093/...
. The hypothesized mechanism is that bacteria-derived amyloids leak from the GI tract and accumulate in the brain, resulting in an increase of reactive oxygen species and activation of nuclear factor-κB, which upregulates the pro-inflammatory microRNA-34a5151. Pistollato F, Cano SS, Elio I, Vergara MM, Giampieri F, Battino M. Role of gut microbiota and nutrients in amyloid formation and pathogenesis of Alzheimer disease. Nutr Rev. 2016 Oct;74(10):624-34. https://doi.org/10.1093/nutrit/nuw023
https://doi.org/https://doi.org/10.1093/...
. Subsequently, microRNA-34a downregulates the expression of TREM2 (triggering receptor expressed on myeloid cells 2), leading to impairment of phagocytosis and contributing to the peptide accumulation β-amyloid1-425151. Pistollato F, Cano SS, Elio I, Vergara MM, Giampieri F, Battino M. Role of gut microbiota and nutrients in amyloid formation and pathogenesis of Alzheimer disease. Nutr Rev. 2016 Oct;74(10):624-34. https://doi.org/10.1093/nutrit/nuw023
https://doi.org/https://doi.org/10.1093/...
. Both amyloids and LPS are potent activators of the receptor for advanced glycation end-products (RAGE) and Toll-like receptors (TLR), and their co-activation amplifies pro-inflammatory signaling leading to sustained chronic inflammation in AD5050. Sochocka M, Donskow-Łysoniewska K, Diniz BS, Kurpas D, Brzozowska E, Leszek J. The gut microbiome alterations and inflammation-driven pathogenesis of Alzheimer’s disease-a critical review. Mol Neurobiol. 2019 Mar;56(3):1841-51. https://doi.org/10.1007/s12035-018-1188-4
https://doi.org/https://doi.org/10.1007/...
,5151. Pistollato F, Cano SS, Elio I, Vergara MM, Giampieri F, Battino M. Role of gut microbiota and nutrients in amyloid formation and pathogenesis of Alzheimer disease. Nutr Rev. 2016 Oct;74(10):624-34. https://doi.org/10.1093/nutrit/nuw023
https://doi.org/https://doi.org/10.1093/...
.

Commensal microbiota produces an assortment of neuroactive molecules, such as serotonin, kynurenine, GABA, catecholamines, histamine, and acetylcholine, among others5050. Sochocka M, Donskow-Łysoniewska K, Diniz BS, Kurpas D, Brzozowska E, Leszek J. The gut microbiome alterations and inflammation-driven pathogenesis of Alzheimer’s disease-a critical review. Mol Neurobiol. 2019 Mar;56(3):1841-51. https://doi.org/10.1007/s12035-018-1188-4
https://doi.org/https://doi.org/10.1007/...
. The consequence of a dysbiotic bowel in the metabolism of tryptophan and kynurenic pathways is documented in AD5050. Sochocka M, Donskow-Łysoniewska K, Diniz BS, Kurpas D, Brzozowska E, Leszek J. The gut microbiome alterations and inflammation-driven pathogenesis of Alzheimer’s disease-a critical review. Mol Neurobiol. 2019 Mar;56(3):1841-51. https://doi.org/10.1007/s12035-018-1188-4
https://doi.org/https://doi.org/10.1007/...
. Gut microbes may regulate the serotonergic system directly by producing serotonin or degrading the serotonin precursor, tryptophan4949. Fox M, Knorr DA, Haptonstall KM. Alzheimer’s disease and symbiotic microbiota: an evolutionary medicine perspective. Ann N Y Acad Sci. 2019 Aug;1449(1):3-24. https://doi.org/10.1111/nyas.14129
https://doi.org/https://doi.org/10.1111/...
. Escherichia coli, an Enterobacteriaceae member, plays an essential role in regulating production and availability of serotonin, acting as a transmitter both in the CNS and in the enteric nervous system5151. Pistollato F, Cano SS, Elio I, Vergara MM, Giampieri F, Battino M. Role of gut microbiota and nutrients in amyloid formation and pathogenesis of Alzheimer disease. Nutr Rev. 2016 Oct;74(10):624-34. https://doi.org/10.1093/nutrit/nuw023
https://doi.org/https://doi.org/10.1093/...
. Nevertheless, gut-derived serotonin only exerts indirect effects on brain functions. Despite that, the gut is the only source of tryptophan, derived either from the diet or microbial production. Tryptophan crosses the BBB to become available for serotonin synthesis in the brain4949. Fox M, Knorr DA, Haptonstall KM. Alzheimer’s disease and symbiotic microbiota: an evolutionary medicine perspective. Ann N Y Acad Sci. 2019 Aug;1449(1):3-24. https://doi.org/10.1111/nyas.14129
https://doi.org/https://doi.org/10.1111/...
. Gulaj et al. found reduced plasma concentration of tryptophan and kynurenic acid in 34 patients with AD, suggesting that dysregulation of the kynurenine route is present in AD5454. Gulaj E, Pawlak K, Bien B, Pawlak D. Kynurenine and its metabolites in Alzheimer’s disease patients. Adv Med Sci. 2010;55(2):204-11. https://doi.org/10.2478/v10039-010-0023-6
https://doi.org/https://doi.org/10.2478/...
.

Lactobacillus and Bifidobacterial genera can metabolize glutamate to produce GABA. Changes in gut microbiota might compromise the endogenous production of this inhibitory neurotransmitter5151. Pistollato F, Cano SS, Elio I, Vergara MM, Giampieri F, Battino M. Role of gut microbiota and nutrients in amyloid formation and pathogenesis of Alzheimer disease. Nutr Rev. 2016 Oct;74(10):624-34. https://doi.org/10.1093/nutrit/nuw023
https://doi.org/https://doi.org/10.1093/...
. Alterations in GABA signaling are linked to cognitive impairment and AD neuropathy4949. Fox M, Knorr DA, Haptonstall KM. Alzheimer’s disease and symbiotic microbiota: an evolutionary medicine perspective. Ann N Y Acad Sci. 2019 Aug;1449(1):3-24. https://doi.org/10.1111/nyas.14129
https://doi.org/https://doi.org/10.1111/...
. Likewise, Lactobacillus, Lactococcus, Streptococcus, and Enterococcus may produce histamine, which acts as a neurotransmitter essential for modulating neuroinflammation through TNF-α expression in the brain5555. Alkasir R, Li J, Li X, Jin M, Zhu B. Human gut microbiota: the links with dementia development. Protein Cell. 2017 Feb;8(2):90-102. https://doi.org/10.1007/s13238-016-0338-6
https://doi.org/https://doi.org/10.1007/...
. Furthermore, an N-methyl-D-aspartate (NMDA)-targeting neurotoxin that was observed to be raised in AD brains may be produced by gut cyanobacteria4949. Fox M, Knorr DA, Haptonstall KM. Alzheimer’s disease and symbiotic microbiota: an evolutionary medicine perspective. Ann N Y Acad Sci. 2019 Aug;1449(1):3-24. https://doi.org/10.1111/nyas.14129
https://doi.org/https://doi.org/10.1111/...
.

The role of diet in these mechanisms is still poorly understood but probably extremely important4747. Kowalski K, Mulak A. Brain-gut-microbiota axis in Alzheimer’s disease. J Neurogastroenterol Motil. 2019 Jan;25(1):48-60. https://doi.org/10.5056/jnm18087
https://doi.org/https://doi.org/10.5056/...
. Dietary patterns similar to the Mediterranean diet and Dietary Approaches to Stop Hypertension have been associated with a reduced risk of AD5656. Nagpal R, Neth BJ, Wang S, Craft S, Yadav H. Modified Mediterranean-ketogenic diet modulates gut microbiome and short-chain fatty acids in association with Alzheimer’s disease markers in subjects with mild cognitive impairment. EBioMedicine. 2019 Sep;47:529-42. https://doi.org/10.1016/j.ebiom.2019.08.032
https://doi.org/https://doi.org/10.1016/...
. In contrast, a Western-style diet represents a risk factor. The variety and composition of a diet and long-term dietary habits may influence the gut microbiota composition and shape the microbial community5151. Pistollato F, Cano SS, Elio I, Vergara MM, Giampieri F, Battino M. Role of gut microbiota and nutrients in amyloid formation and pathogenesis of Alzheimer disease. Nutr Rev. 2016 Oct;74(10):624-34. https://doi.org/10.1093/nutrit/nuw023
https://doi.org/https://doi.org/10.1093/...
. A newly proposed insight is that the transplantation of fecal microflora from healthy people to patients with AD can help restore the intestinal microbiota and reduce the negative impact of the dysbiotic microbiome on the gut and brain functions4747. Kowalski K, Mulak A. Brain-gut-microbiota axis in Alzheimer’s disease. J Neurogastroenterol Motil. 2019 Jan;25(1):48-60. https://doi.org/10.5056/jnm18087
https://doi.org/https://doi.org/10.5056/...
. The influence of gut microbiota on brain function is being investigated continuously. Table 3 presents additional studies on the role of gut microbiota and dementia.

EPILEPSY, KETOGENIC DIET, AND THE INTESTINAL MICROBIOME

About one-third of patients with epilepsy have seizures refractory to anti-seizure drugs (ASD)5757. Chen Z, Brodie MJ, Liew D, Kwan P. Treatment outcomes in patients with newly diagnosed epilepsy treated with established and new antiepileptic drugs a 30-year longitudinal cohort study. JAMA Neurol. 2018 Mar;75(3):279-86. https://doi.org/10.1001/jamaneurol.2017.3949
https://doi.org/https://doi.org/10.1001/...
. Non-pharmacological approaches, especially the ketogenic diet (KD), are alternatives in cases of pharmacoresistant epilepsy. The KD has been used for about one hundred years, demonstrating efficacy in reducing seizure frequency, mostly in children with difficult-to-control epilepsy5858. Devinsky O, Vezzani A, O’Brien TJ, Jette N, Scheffer IE, De Curtis M, et al. Epilepsy. Nat Rev Dis Primers. 2018 May;4:18024. https://doi.org/10.1038/nrdp.2018.24
https://doi.org/https://doi.org/10.1038/...
.

The KD appears to be a powerful contributor in modulating downstream effects on an individual’s gut metagenomic composition and metabolomic profile5959. Cabrera-Mulero A, Tinahones A, Bandera B, Moreno-Indias I, Macías-González M, Tinahones FJ. Keto microbiota: A powerful contributor to host disease recovery. Rev Endocr Metab Disord. 2019 Dec;20(4):415-25. https://doi.org/10.1007/s11154-019-09518-8
https://doi.org/https://doi.org/10.1007/...
. In a dietary intervention study conducted by Ang et al., two cohorts composed of obese patients and mice were followed up during short- and long-term periods. In humans, a baseline diet and a KD with only 5% of carbohydrates content were used. Mice were submitted to three different diet types: low-fat diet (LFD), high-fat diet (HFD), and KD. Results from a 16S amplicon-based metagenomic approach of stool samples and metabolomic profile in humans demonstrated a significant reduction in the relative abundance of Actinobacteria phylum and a marked decrease in different bifidobacterial species, suggesting that the KD is sufficient to shift the gut microbiota composition. Similar findings were seen in a mice model, where ketone bodies like βHB had a direct effect in suppressing microbial proliferation of bifidobacteria. Moreover, fecal microbial transplantation from patients into germ-free mice demonstrated that the colonization of KD-associated microorganisms drove the reduction of selected members of Bifidobacterium adolescentis, therefore modulating the induction of Th17 cells. These findings reveal a separate pathway whereby carbohydrate restriction, rather than high-fat intake, is the main contributor to gut microbiome structure and immune response6060. Ang QY, Alexander M, Newman JC, Tian Y, Cai J, Upadhyay V, et al. Ketogenic diets alter the gut microbiome resulting in decreased intestinal Th17 cells. Cell. 2020 Jun;181(6):1263-1275.e16. https://doi.org/10.1016/j.cell.2020.04.027
https://doi.org/https://doi.org/10.1016/...
.

Olson et al. demonstrated that the taxonomic composition of the gut microbiome in mice is altered after treatment with KD. They also raised the possibility that some KD-associated species such as Akkermansia muciniphila and Parabacteroides merdae and particular molecules predicted seizure protection with high accuracy and were necessary to reduce brain electrical activity6161. Olson CA, Vuong HE, Yano JM, Liang QY, Nusbaum DJ, Hsiao EY. The gut microbiota mediates the anti-seizure effects of the ketogenic diet. Cell. 2018 Jun;173(7):1728-1741.e13. https://doi.org/10.1016/j.cell.2018.04.027
https://doi.org/https://doi.org/10.1016/...
. Taken together, these findings are likely to be correlated with a decrease in the metabolism of ketogenic gamma-glutamylated (GG) amino acids and, therefore, to low concentrations of these amino acids in the colon, blood, and CNS. Likewise, this phenomenon can boost GABA bioavailability on the hippocampus, increasing seizure threshold in mice and contributing to the anti-seizure effect. Finally, based on these bacterial species and molecules, researchers could identify microbiome-based treatments such as microbiome transplant, live biotherapeutic products, and targeted pharmacological approaches that protect against seizures in mice6161. Olson CA, Vuong HE, Yano JM, Liang QY, Nusbaum DJ, Hsiao EY. The gut microbiota mediates the anti-seizure effects of the ketogenic diet. Cell. 2018 Jun;173(7):1728-1741.e13. https://doi.org/10.1016/j.cell.2018.04.027
https://doi.org/https://doi.org/10.1016/...
.

To further examine the potential of KD in shaping the intestinal microbiome in patients with epilepsy, Lindefeldt et al. analyzed the taxonomic and functional profile in children with difficult-to-control epilepsy using whole metagenomic sequencing6262. Lindefeldt M, Eng A, Darban H, Bjerkner A, Zetterström CK, Allander T, et al. The ketogenic diet influences taxonomic and functional composition of the gut microbiota in children with severe epilepsy. NPJ Biofilms Microbiomes. 2019 Jan;5(1):1-13. https://doi.org/10.1038/s41522-018-0073-2
https://doi.org/https://doi.org/10.1038/...
. The study consistently showed that i) there were differences in the composition of the patient’s gut regarding healthy controls before starting the intervention; ii) those differences were reflected in the reduction of relative abundances of butyrate-producing organisms such as Eubacterium rectale and Dialister during and after treatment with KD where variations were more noticeable; iii) KD decreased Bifidobacterium and Dialister had an impact not only on the production of non-digestible carbohydrates (e.g., lactate) but also on the final conversion to SCFAs (mainly acetate), which are crucial in brain physiology; finally, iv) shifts on gut microbiome associated with KD promoted the growth of Escherichia coli, which could trigger gut inflammation in epileptic patients6262. Lindefeldt M, Eng A, Darban H, Bjerkner A, Zetterström CK, Allander T, et al. The ketogenic diet influences taxonomic and functional composition of the gut microbiota in children with severe epilepsy. NPJ Biofilms Microbiomes. 2019 Jan;5(1):1-13. https://doi.org/10.1038/s41522-018-0073-2
https://doi.org/https://doi.org/10.1038/...
. These results are in line with a study carried out by Zhang et al., who explored the potential of microbial biomarkers in patients with refractory epilepsy who followed a KD for six months6363. Zhang Y, Zhou S, Zhou Y, Yu L, Zhang L, Wang Y. Altered gut microbiome composition in children with refractory epilepsy after ketogenic diet. Epilepsy Res. 2018 Sep;145:163-8. https://doi.org/10.1016/j.eplepsyres.2018.06.015
https://doi.org/https://doi.org/10.1016/...
. Overall, they observed low diversity and richness ratios in individuals undergoing a KD as well as an increase in the relative abundance of Bacteroidetes and reduction of Firmicutes and Actinobacteria phyla. They also reported a high abundance of Clostridia class organisms in non-responders. This class has been associated with tryptophan catabolites, which in turn are responsible for hormone secretion, neurotransmission, gut motility and permeability, and anti-oxidative effects6363. Zhang Y, Zhou S, Zhou Y, Yu L, Zhang L, Wang Y. Altered gut microbiome composition in children with refractory epilepsy after ketogenic diet. Epilepsy Res. 2018 Sep;145:163-8. https://doi.org/10.1016/j.eplepsyres.2018.06.015
https://doi.org/https://doi.org/10.1016/...
. Another work involving epilepsy and KD had a similar outcome and reported significant gut dysbiosis in the refractory group. Nonetheless, the enterotype Bacteroidetes was accumulated in both the healthy and epileptic groups after at least one week of high-fat diet therapy. This phylum has been related to seizure modulation by secretion of inflammatory cytokines, including IL6 and IL17. Also, Cronobacter was the predominant genus identified only in affected children, which decreased over the treatment period6464. Xie G, Qian Z, Qiu C-Z, Dai W-K, Wang H-P, Li Y-H, et al. Ketogenic diet poses a significant effect on imbalanced gut microbiota in infants with refractory epilepsy. World J Gastroenterol. 2017 Sep;23(33):6164-71. https://doi.org/10.3748/wjg.v23.i33.6164
https://doi.org/https://doi.org/10.3748/...
.

It has been observed that patients with epilepsy are presumably prone to gut dysbiosis and, hence, to chronic inflammation of the intestinal epithelium6565. Dahlin M, Prast-Nielsen S. The gut microbiome and epilepsy. EBioMedicine. 2019 Jun;44:741-6. https://doi.org/10.1016/j.ebiom.2019.05.024
https://doi.org/https://doi.org/10.1016/...
,6666. Lee K, Kim N, Shim JO, Kim G-H. Gut bacterial dysbiosis in children with intractable epilepsy. J Clin Med. 2021 Jan;10(1):5. https://doi.org/10.3390/jcm10010005
https://doi.org/https://doi.org/10.3390/...
,6767. Şafak B, Altunan B, Topçu B, Eren Topkaya A. The gut microbiome in epilepsy. Microb Pathog. 2020 Feb;139:103853. https://doi.org/10.1016/j.micpath.2019.103853
https://doi.org/https://doi.org/10.1016/...
. In a cohort of 91 individuals, Peng et al. revealed that patients with more than four seizures per year had a predominance of Ruminococcus and rare bacteria genera compared to a drug-sensitive group. These findings lead to various hypotheses: i) the use of several antiepileptic drugs (AED) induces intestinal dysbiosis; ii) the prevalence of rare microorganisms modulate metabolic pathways involving ABC transporters, therefore conferring chemoresistance to the treatment; iii) Bifidobacterial and Lactobacilli genera stimulate the production of GABA and are prevalent in the drug-sensitive group6868. Peng A, Qiu X, Lai W, Li W, Zhang L, Zhu X, et al. Altered composition of the gut microbiome in patients with drug-resistant epilepsy. Epilepsy Res. 2018 Nov;147:102-7. https://doi.org/10.1016/j.eplepsyres.2018.09.013
https://doi.org/https://doi.org/10.1016/...
.

So far, all of the studies involving different epilepsy phenotypes either in humans or mice report changes in the intestinal microbiome at baseline or after a KD6969. Gong X, Liu X, Chen C, Lin J, Li A, Guo K, et al. Alteration of gut microbiota in patients with epilepsy and the potential index as a biomarker. Front Microbiol. 2020 Sep;11:517797. https://doi.org/10.3389/fmicb.2020.517797
https://doi.org/https://doi.org/10.3389/...
. Still, there is little overlap across these studies in the exact microbial signatures that have been identified (Table 4). Thus, additional high-powered and well-controlled studies are needed to explore the issue better and propose new treatment options7070. Gómez-Eguílaz M, Ramón-Trapero JL, Pérez-Martínez L, Blanco JR. The beneficial effect of probiotics as a supplementary treatment in drug-resistant epilepsy: A pilot study. Benef Microbes. 2018 Dec;9(6):875-81. https://doi.org/10.3920/BM2018.0018
https://doi.org/https://doi.org/10.3920/...
,7171. Braakman HMH, van Ingen J. Can epilepsy be treated by antibiotics? J Neurol. 2018 Aug;265(8):1934-6. https://doi.org/10.1007/s00415-018-8943-3
https://doi.org/https://doi.org/10.1007/...
.

PARKINSON’S DISEASE AND MICROBIOME

It is well known that patients with Parkinson’s disease (PD) present severe non-motor symptoms at the prodromal phase of the disease, which are determinants of the quality of life in these individuals. Sensorial, neuropsychiatric, sleep dysfunction, and GI symptoms (constipation) are the most common phenotypes describe in this category7272. Kalia LV, Lang AE. Parkinson’s disease. Lancet. 2015 Aug;386(9996):896-912. https://doi.org/10.1016/S0140-6736(14)61393-3
https://doi.org/https://doi.org/10.1016/...
. Several studies using breath testing demonstrated that a considerable proportion of patients with PD have intestinal bacterial overgrowth and absorption issues, leading to intestinal constipation. On the other hand, 16S ribosomal RNA analysis from colonic biopsies and stool samples of adults with PD showed decreased SCFAs-producing bacteria such as Blautia, Coprococcus, and Roseburia7373. Keshavarzian A, Green SJ, Engen PA, Voigt RM, Naqib A, Forsyth CB, et al. Colonic bacterial composition in Parkinson’s disease. Mov Disord. 2015 Sep;30(10):1351-60. https://doi.org/10.1002/mds.26307
https://doi.org/https://doi.org/10.1002/...
,7474. Aho VTE, Houser MC, Pereira PAB, Chang J, Rudi K, Paulin L, et al. Relationships of gut microbiota, short-chain fatty acids, inflammation, and the gut barrier in Parkinson’s disease. Mol Neurodegener. 2021 Feb;16(1):6. https://doi.org/10.1186/s13024-021-00427-6.
https://doi.org/https://doi.org/10.1186/...
. Furthermore, depletion or increase in Prevotella and Lactobacillus genera was reported in several case-control studies performed in these patients7575. Aho VTE, Pereira PAB, Voutilainen S, Paulin L, Pekkonen E, Auvinen P, et al. Gut microbiota in Parkinson’s disease: temporal stability and relations to disease progression. EBioMedicine. 2019 Jun;44:691-707. https://doi.org/10.1016/j.ebiom.2019.05.064
https://doi.org/https://doi.org/10.1016/...
,7676. Li C, Cui L, Yang Y, Miao J, Zhao X, Zhang J, et al. Gut microbiota differs between parkinson’s disease patients and healthy controls in northeast China. Front Mol Neurosci. 2019 Jul;12:171. https://doi.org/10.3389/fnmol.2019.00171
https://doi.org/https://doi.org/10.3389/...
. This class of beneficial microorganisms is involved in BBB integrity, permeability, and neuronal inflammatory signaling7373. Keshavarzian A, Green SJ, Engen PA, Voigt RM, Naqib A, Forsyth CB, et al. Colonic bacterial composition in Parkinson’s disease. Mov Disord. 2015 Sep;30(10):1351-60. https://doi.org/10.1002/mds.26307
https://doi.org/https://doi.org/10.1002/...
. Moreover, the increase in Enterobacteriaceae members was directly proportional to the severity of symptoms like stability, gait, and rigidity7777. Scheperjans F, Aho V, Pereira PAB, Koskinen K, Paulin L, Pekkonen E, et al. Gut microbiota are related to Parkinson’s disease and clinical phenotype. Mov Disord. 2015 Mar;30(3):350-8. https://doi.org/10.1002/mds.26069
https://doi.org/https://doi.org/10.1002/...
. Conversely, in a study conducted in individuals with PD, the oral microbiome was analyzed. Male patients exhibited an increase in the abundance of Prevotella, which is considered an opportunistic pathobiont on the mouth, suggesting a strong role of these genera in periodontal disease7878. Pereira PAB, Aho VTE, Paulin L, Pekkonen E, Auvinen P, Scheperjans F. Oral and nasal microbiota in Parkinson’s disease. Parkinsonism Relat Disord. 2017 May;38:61-7. https://doi.org/10.1016/j.parkreldis.2017.02.026
https://doi.org/https://doi.org/10.1016/...
. Taken together, these findings support that modifications in bacteria density, taxonomic levels (dysbiosis), and mucin production may, in turn, boost the neuroglia system, triggering damage to the intestinal and brain barriers, leading to alpha-synuclein protein misfolding and finally aggregation. Likewise, chronic systemic exposure to LPS leads to the selective death of dopaminergic neurons in the substantia nigra7373. Keshavarzian A, Green SJ, Engen PA, Voigt RM, Naqib A, Forsyth CB, et al. Colonic bacterial composition in Parkinson’s disease. Mov Disord. 2015 Sep;30(10):1351-60. https://doi.org/10.1002/mds.26307
https://doi.org/https://doi.org/10.1002/...
,7979. Houser MC, Tansey MG. The gut-brain axis: Is intestinal inflammation a silent driver of Parkinson’s disease pathogenesis? NPJ Parkinsons Dis [Internet]. 2017 Jan;3(3):1-9. https://doi.org/10.1038/s41531-016-0002-0
https://doi.org/https://doi.org/10.1038/...
,8080. Liu B, Fang F, Pedersen NL, Tillander A, Ludvigsson JF, Ekbom A, et al. Vagotomy and Parkinson disease. Neurology. 2017 May;88(21):1996-2002. https://doi.org/10.1212/WNL.0000000000003961
https://doi.org/https://doi.org/10.1212/...
.

The vagus nerve (VN) has also been implicated in the pathogenesis of PD. Some researchers suggest that the VN can modulate neuroimmune and inflammatory signals either via top-down or through the microbiota-gut axis7979. Houser MC, Tansey MG. The gut-brain axis: Is intestinal inflammation a silent driver of Parkinson’s disease pathogenesis? NPJ Parkinsons Dis [Internet]. 2017 Jan;3(3):1-9. https://doi.org/10.1038/s41531-016-0002-0
https://doi.org/https://doi.org/10.1038/...
. Thus, it has been proposed that the VN could transport alpha-synuclein to the CNS and vice versa. After examining 9,430 vagotomies in Swedish patients, Liu et al. demonstrated that truncal, but not selective vagotomy, had a protective effect against PD development8080. Liu B, Fang F, Pedersen NL, Tillander A, Ludvigsson JF, Ekbom A, et al. Vagotomy and Parkinson disease. Neurology. 2017 May;88(21):1996-2002. https://doi.org/10.1212/WNL.0000000000003961
https://doi.org/https://doi.org/10.1212/...
. In contrast, recent studies in mouse models mimicking motor and non-motor symptoms of early and late stages of the disease point out that changes in the immune response to gut bacteria could affect motor symptoms in PD8181. Sun MF, Shen YQ. Dysbiosis of gut microbiota and microbial metabolites in Parkinson’s disease. Ageing Res Rev. 2018 Aug;45:53-61. https://doi.org/10.1016/j.arr.2018.04.004
https://doi.org/https://doi.org/10.1016/...
. In an experimental study performed by Sangjune et al., the authors demonstrated that pathologic species of a-synuclein could spread from the gut muscles to the brain through connections of the vagus nerve. These mice also showed neuropsychiatric symptoms, including anxiety, depression, olfactory dysfunction, and spatial learning and memory abnormalities. They also assessed another group of animals that were submitted to an injection of a-synuclein and vagotomy. The authors observed that no transmission of pathologic α-synuclein occurred in these animals, which were also free of the cardinal symptoms of PD8282. Kim S, Kwon SH, Kam TI, Panicker N, Karuppagounder SS, Lee S, et al. Transneuronal propagation of pathologic α-synuclein from the gut to the brain models Parkinson’s disease. Neuron. 2019 Aug;103(4):627-641.e7. https://doi.org/10.1016/j.neuron.2019.05.035
https://doi.org/https://doi.org/10.1016/...
.

Several studies have been published exploring the gut microbiome in PD (Table 5). However, the need for well-designed clinical studies exploring the role of the gut microbiome in PD in the clinical setting is still lacking.

Gut microbiota alterations in neuropsychiatric diseases

Autism Spectrum Disorder

Clinical observations indicate that patients with Autism Spectrum Disorder (ASD) have GI disturbances that include diarrhea, constipation, and abdominal pain8383. Lefter R, Ciobica A, Timofte D, Stanciu C, Trifan A. A descriptive review on the prevalence of gastrointestinal disturbances and their multiple associations in autism spectrum disorder. Medicina (Kaunas). 2019 Dec;56(1):11. https://doi.org/10.3390/medicina56010011.
https://doi.org/https://doi.org/10.3390/...
. Likewise, a growing number of studies have shown that patients with ASD have an altered gut microbiota composition compared to neurotypical individuals8484. 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 Mar;11:22. https://doi.org/10.1186/1471-230X-11-22.
https://doi.org/https://doi.org/10.1186/...
. Moreover, GI disturbances strongly correlate with the severity of ASD symptoms, and GI disturbances are markedly associated with GI dysbiosis8383. Lefter R, Ciobica A, Timofte D, Stanciu C, Trifan A. A descriptive review on the prevalence of gastrointestinal disturbances and their multiple associations in autism spectrum disorder. Medicina (Kaunas). 2019 Dec;56(1):11. https://doi.org/10.3390/medicina56010011.
https://doi.org/https://doi.org/10.3390/...
,8484. 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 Mar;11:22. https://doi.org/10.1186/1471-230X-11-22.
https://doi.org/https://doi.org/10.1186/...
. Therefore, it has been suggested that gut microbiota alterations may contribute to the pathogenesis of ASD(69). Although several studies have shown that adult rodents prenatally exposed to VPA, a model for ASD, exhibit gut dysbiosis8585. de Theije CGM, Wopereis H, Ramadan M, van Eijndthoven T, Lambert J, Knol J, et al. Altered gut microbiota and activity in a murine model of autism spectrum disorders. Brain Behav Immun. 2014 Mar;37:197-206. https://doi.org/10.1016/j.bbi.2013.12.005
https://doi.org/https://doi.org/10.1016/...
,8686. Liu F, Horton-Sparks K, Hull V, Li RW, Martínez-Cerdeño V. The valproic acid rat model of autism presents with gut bacterial dysbiosis similar to that in human autism. Mol Autism. 2018 Dec;9:61. https://doi.org/10.1186/s13229-018-0251-3
https://doi.org/https://doi.org/10.1186/...
, there is a lack of evidence of a causal link between abnormal microbiota and ASD-like behaviors.

Schizophrenia

Patients with schizophrenia commonly have GI disturbances, such as constipation and GI hypomotility and inflammation8787. Severance EG, Prandovszky E, Castiglione J, Yolken RH. Gastroenterology Issues in schizophrenia: why the gut matters. Curr Psychiatry Rep. 2015 May;17(5):27. https://doi.org/10.1007/s11920-015-0574-0
https://doi.org/https://doi.org/10.1007/...
,8888. Severance EG, Yolken RH, Eaton WW. Autoimmune diseases, gastrointestinal disorders and the microbiome in schizophrenia: more than a gut feeling. Schizophr Res. 2016 Sep;176(1):23-35. https://doi.org/10.1016/j.schres.2014.06.027
https://doi.org/https://doi.org/10.1016/...
. A growing body of evidence indicates that altered gut microbiota may account for the GI disturbances and the severity of symptoms in schizophrenia patients suggesting a key role of the gut microbiota in promoting the pathogenesis of schizophrenia. Interestingly, this suggestion has been recently proved by Zheng and colleagues. In this study, the authors found that the gut microbiota from patients with schizophrenia induced behavioral alterations and modulated brain excitability when transferred to mice8989. Zheng P, Zeng B, Liu M, Chen J, Pan J, Han Y, et al. Correction for the Research Article: The gut microbiome from patients with schizophrenia modulates the glutamate-glutamine-GABA cycle and schizophrenia-relevant behaviors in mice. Sci Adv. 2019 Jun 21;5(6):eaay2759. https://doi.org/10.1126/sciadv.aay2759
https://doi.org/https://doi.org/10.1126/...
. Other studies have also suggested that GI disturbances and gut microbiota alterations in schizophrenia may be related to the use of antipsychotic medication9090. Flowers SA, Baxter NT, Ward KM, Kraal AZ, McInnis MG, Schmidt TM, 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 Feb;39(2):161-70. https://doi.org/10.1002/phar.2214
https://doi.org/https://doi.org/10.1002/...
. However, gut microbiota seems to be altered in patients even before medication9191. Schwarz E, Maukonen J, Hyytiäinen T, Kieseppä T, Orešič M, Sabunciyan S, et al. Analysis of microbiota in first episode psychosis identifies preliminary associations with symptom severity and treatment response. Schizophr Res. 2018 Feb;192:398-403. https://doi.org/10.1016/j.schres.2017.04.017
https://doi.org/https://doi.org/10.1016/...
. Hence, to better understand the gut microbiota alterations in schizophrenia, experimental validation of clinical findings seems necessary. Furthermore, animal models of schizophrenia reinforce the clinical data showing that gut dysbiosis may be implicated in schizophrenia, mainly pointing to alterations in the Firmicutes phylum bacteria.

ANXIETY AND DEPRESSION DISORDERS

Preclinical studies have shown that the intestinal microbiome seems to play a crucial role in the pathophysiology of both neuropsychiatric disorders. Also, fecal microbiota from patients diagnosed with anxiety and depression induce behavioral and physiological features of these disorders when transplanted to microbiota-deficient animals, including anhedonia, anxiety-like behaviors, and altered tryptophan metabolism9292. Kelly JR, Borre Y, O’ Brien C, Patterson E, El Aidy S, Deane J, et al. Transferring the blues: Depression-associated gut microbiota induces neurobehavioural changes in the rat. J Psychiatr Res. 2016 Nov;82:109-18. https://doi.org/10.1016/j.jpsychires.2016.07.019
https://doi.org/https://doi.org/10.1016/...
,9393. Zheng P, Zeng B, Zhou C, Liu M, Fang Z, Xu X, et al. Gut microbiome remodeling induces depressive-like behaviors through a pathway mediated by the host’s metabolism. Mol Psychiatry. 2016 Jun;21(6):786-96. https://doi.org/10.1038/mp.2016.44
https://doi.org/https://doi.org/10.1038/...
,9494. Cryan JF, O’Riordan KJ, Cowan CSM, Sandhu KV, Bastiaanssen TFS, Boehme M, et al. The microbiota-gut-brain axis. Physiol Rev. 2019 Oct;99(4):1877-2013. https://doi.org/10.1152/physrev.00018.2018
https://doi.org/https://doi.org/10.1152/...
.

Park and colleagues showed that anxiety- and depression-like behaviors induced by olfactory bulbectomy (OBx) are related to colonic motility alterations and gut microbiota composition changes. In addition, the expression of hypothalamic corticotropin-hormone (CRH) was elevated in OBx mice, suggesting that GI disturbances and gut dysbiosis may be due to the recruitment of the hypothalamic-pituitary-adrenal axis9595. Park AJ, Collins J, Blennerhassett PA, Ghia JE, Verdu EF, Bercik P, et al. Altered colonic function and microbiota profile in a mouse model of chronic depression. Neurogastroenterol Motil. 2013 Sep;25(9):733-e575. https://doi.org/10.1111/nmo.12153
https://doi.org/https://doi.org/10.1111/...
,9696. de Weerth C. Do bacteria shape our development? Crosstalk between intestinal microbiota and HPA axis. Neurosci Biobehav Rev. 2017 Dec;83:458-71. https://doi.org/10.1016/j.neubiorev.2017.09.016
https://doi.org/https://doi.org/10.1016/...
. Murakami et al. analyzed the gut microbiota composition of fecal samples from Wistar rats submitted to maternal separation when neonates. This early-life stressor leads to anxiety- and depression-like behaviors in adulthood. The authors found that maternal separation induced a specific reduction in Bifidobacterium, Bacteroidetes, and Prevotella genera9797. Murakami T, Kamada K, Mizushima K, Higashimura Y, Katada K, Uchiyama K, et al. Changes in intestinal motility and gut microbiota composition in a rat stress model. Digestion. 2017;95(1):55-60. https://doi.org/10.1159/000452364
https://doi.org/https://doi.org/10.1159/...
. Li et al. analyzed the gut microbiota of fecal samples from mice submitted to the chronic unpredictable mild stress (CUMS) model. They found that the gut microbiota of CUMS-treated mice exhibits drastic alterations in microbiota composition, including an increased α-diversity and changes in the abundance of specific microbial phyla, such as Verrucomicrobia and Proteobacteria. At the genus level, animals exposed to CUMS exhibit an increased abundance in Helicobacter, Turicibacter, Parasutterella, Alistipes, Odoribacter, and Akkermansia, but a decrease in Barnesiella, Bifidobacterium, Lactobacillus, and Olsenella9898. Li N, Wang Q, Wang Y, Sun A, Lin Y, Jin Y, et al. Fecal microbiota transplantation from chronic unpredictable mild stress mice donors affects anxiety-like and depression-like behavior in recipient mice via the gut microbiota-inflammation-brain axis. Stress. 2019 Sep;22(5):592-602. https://doi.org/10.1080/10253890.2019.1617267
https://doi.org/https://doi.org/10.1080/...
.

FUTURE DIRECTIONS

Reports using cutting-edge technologies such as metagenomics and metabolomics are changing some of the established paradigms regarding the physiopathological mechanisms behind neuropsychiatric diseases. Using the potential of microbial profiles as biomarkers of neurological and mental health disorders may maximize the efficacy of existing therapies. Current challenges remain in establishing causation rather than association and translating basic science studies into clinical practice with the potential of targeting the microbiome for therapeutic purposes.

References

  • Support: This work was supported by a grant from Fundação de Amparo à Pesquisa do Estado de São Paulo (FAPESP, grant number 2013/07559-3), SP, Brazil and Coordenação de Aperfeiçoamento de Pessoal de Nível Superior (CAPES, grant number: 001), Brazil. DMM-G, ABG, AMC, DCR, and AD are supported by fellowships from FAPESP (grants #2018/00142-3, 2019/00213-0, 2019/25948-3, 2019/0048-0 and 2015/25607-0, respectively). I.L.-C. is supported by Conselho Nacional de Pesquisa (CNPq), Brazil (grant #311923/2019-4).

Publication Dates

  • Publication in this collection
    25 Mar 2022
  • Date of issue
    Feb 2022

History

  • Received
    12 Feb 2021
  • Reviewed
    13 Apr 2021
  • Accepted
    10 May 2021
Academia Brasileira de Neurologia - ABNEURO R. Vergueiro, 1353 sl.1404 - Ed. Top Towers Offices Torre Norte, 04101-000 São Paulo SP Brazil, Tel.: +55 11 5084-9463 | +55 11 5083-3876 - São Paulo - SP - Brazil
E-mail: revista.arquivos@abneuro.org