Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 122
Filtrar
1.
PLoS One ; 19(7): e0306582, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38959253

RESUMO

Schizophrenia is a severe, complex and long-term psychiatric disorder with unclear etiology. Gut microbes influence the central nervous system via the gut-brain axis. Consequently, investigations of the relationship between gut microbes and schizophrenia are warranted. This study involved 29 patients with schizophrenia and 30 age-matched normal controls. After 16S rRNA gene sequencing and whole-genome shotgun metagenomic sequencing, we analyzed microbial diversity, composition, and function. According to 16S rRNA and metagenomic gene sequencing results, patients with schizophrenia had higher abundances of Clostridium and Megasphaera. Functional analysis showed that sphingolipid, phosphonates and phosphinates, as well as glutamine metabolism were associated with the occurrence and development of schizophrenia. Our data suggest that the gut microbiota exerts an effect on patients with schizophrenia, providing valuable insights into the potential regulation of in the context of this disorder.


Assuntos
Microbioma Gastrointestinal , RNA Ribossômico 16S , Esquizofrenia , Esquizofrenia/microbiologia , Humanos , Masculino , Feminino , Adulto , RNA Ribossômico 16S/genética , Pessoa de Meia-Idade , Estudos de Casos e Controles , Metagenômica/métodos , Metagenoma
2.
Brain Behav ; 14(6): e3579, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38841824

RESUMO

BACKGROUND: Gut dysbiosis has been established as a characteristic of schizophrenia (SCH). However, the signatures regarding SCH patients with prominent negative symptoms (SCH-N) in young adults have been poorly elucidated. METHODS: Stool samples were obtained from 30 young adults with SCH-N, 32 SCH patients with prominent positive symptoms (SCH-P) along with 36 healthy controls (HCs). Microbial diversity and composition were analyzed by 16S rRNA gene sequencing. Meanwhile, psychiatric symptoms were assessed by the positive and negative syndrome scale (PANSS). RESULTS: There is a significant difference in ß-diversity but not α-diversity indexes among the three groups. Moreover, we found a higher abundance of Fusobacteria and Proteobacteria phyla and a lower abundance of Firmicutes phyla in SCH-N when compared with HC. Besides, we identified a diagnostic potential panel comprising six genera (Coprococcus, Monoglobus, Prevotellaceae_NK3B31_group, Escherichia-Shigella, Dorea, and Butyricicoccus) that can distinguish SCH-N from HC (area under the curve = 0.939). However, the difference in microbial composition between the SCH-N and SCH-P is much less than that between SCH-N and the HC, and SCH-N and SCH-P cannot be effectively distinguished by gut microbiota. CONCLUSION: The composition of gut microbiota was changed in the patients with SCH-N, which may help in further understanding of pathogenesis in young adults with SCH-N.


Assuntos
Fezes , Microbioma Gastrointestinal , RNA Ribossômico 16S , Esquizofrenia , Humanos , Esquizofrenia/microbiologia , RNA Ribossômico 16S/genética , Masculino , Adulto Jovem , Feminino , Adulto , Fezes/microbiologia , Disbiose/microbiologia
3.
Neurosci Biobehav Rev ; 162: 105722, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38754717

RESUMO

FRILEUX, M., BOLTRI M. and al. Cognition and Gut microbiota in schizophrenia spectrum and mood disorders: a Systematic Review. NEUROSCI BIOBEHAV REV (1) 2024 Schizophrenia spectrum disorders and major mood disorders are associated with cognitive impairments. Recent studies suggest a link between gut microbiota composition and cognitive functioning. Here, we review the relationship between gut microbiota and cognition in these disorders. To do this, we conducted a systematic review, searching Cochrane Central Register of Controlled Trials, EBSCOhost, Embase, Pubmed, Scopus, and Web of Science. Studies were included if they investigated the relationship between gut microbiota composition and cognitive function through neuropsychological assessments in patients with bipolar, depressive, schizophrenia spectrum, and other psychotic disorders. Ten studies were identified. Findings underscore a link between gut dysbiosis and cognitive impairment. This relationship identified specific taxa (Haemophilus, Bacteroides, and Alistipes) as potential contributors to bolstered cognitive performance. Conversely, Candida albicans, Toxoplasma gondii, Streptococcus and Deinococcus were associated with diminished performance on cognitive assessments. Prebiotics and probiotics interventions were associated with cognitive enhancements, particularly executive functions. These results emphasize the role of gut microbiota in cognition, prompting further exploration of the underlying mechanisms paving the way toward precision psychiatry.


Assuntos
Microbioma Gastrointestinal , Transtornos do Humor , Esquizofrenia , Humanos , Microbioma Gastrointestinal/fisiologia , Esquizofrenia/microbiologia , Esquizofrenia/fisiopatologia , Transtornos do Humor/microbiologia , Transtornos do Humor/etiologia , Cognição/fisiologia , Disfunção Cognitiva/microbiologia , Disfunção Cognitiva/etiologia , Disfunção Cognitiva/fisiopatologia , Disbiose/microbiologia
4.
Schizophr Res ; 267: 444-450, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38643725

RESUMO

Cognitive impairment is a core symptom of schizophrenia. The gut microbiota (GM) and oxidative stress may play important roles in the pathophysiological mechanisms of cognitive impairment. This study aimed to explore the relationship between GM and oxidative stress in the cognitive function of schizophrenia. GM obtained by 16S RNA sequencing and serum superoxide dismutase (SOD) levels from schizophrenia patients (N = 68) and healthy controls (HCs, N = 72) were analyzed. All psychiatric symptoms were assessed using the Positive and Negative Syndrome Scale (PANSS). Cognitive function was assessed using the MATRICS Consensus Cognitive Battery (MCCB). Correlation analysis was used to explore the relationship between GM, SOD, and cognitive function. Machine learning models were used to identify potential biomarkers. Compared to HCs, the relative abundances of Collinsella, undefined Ruminococcus, Lactobacillus, Eubacterium, Mogibacterium, Desulfovibrio, Bulleidia, Succinivibrio, Corynebacterium, and Atopobium were higher in patients with schizophrenia, but Faecalibacterium, Anaerostipes, Turicibacter, and Ruminococcus were lower. In patients with schizophrenia, the positive factor, general factor, and total score of MCCB positively correlated with Lactobacillus, Collinsella, and Lactobacillus, respectively; SOD negatively correlated with Eubacterium, Collinsella, Lactobacillus, Corynebacterium, Bulleidia, Mogibacterium, and Succinivibrio, but positively correlated with Faecalibacterium, Ruminococcus, and MCCB verbal learning index scores; Faecalibacterium and Turicibacter were positively correlated with MCCB visual learning index scores and speed of processing index scores, respectively. Our findings revealed a correlation between SOD and GM and confirmed that cognitive dysfunction in patients with schizophrenia involves abnormal SOD levels and GM changes.


Assuntos
Disfunção Cognitiva , Microbioma Gastrointestinal , Estresse Oxidativo , Esquizofrenia , Humanos , Esquizofrenia/fisiopatologia , Esquizofrenia/microbiologia , Esquizofrenia/complicações , Microbioma Gastrointestinal/fisiologia , Masculino , Feminino , Estresse Oxidativo/fisiologia , Adulto , Projetos Piloto , China , Disfunção Cognitiva/etiologia , Disfunção Cognitiva/fisiopatologia , Disfunção Cognitiva/microbiologia , Superóxido Dismutase/sangue , Pessoa de Meia-Idade , Adulto Jovem , Aprendizado de Máquina
5.
Nutrients ; 15(20)2023 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-37892465

RESUMO

Schizophrenia, a severe mental illness affecting about 1% of the population, manifests during young adulthood, leading to abnormal mental function and behavior. Its multifactorial etiology involves genetic factors, experiences of adversity, infection, and gene-environment interactions. Emerging research indicates that maternal infection or stress during pregnancy may also increase schizophrenia risk in offspring. Recent research on the gut-brain axis highlights the gut microbiome's potential influence on central nervous system (CNS) function and mental health, including schizophrenia. The gut microbiota, located in the digestive system, has a significant role to play in human physiology, affecting immune system development, vitamin synthesis, and protection against pathogenic bacteria. Disruptions to the gut microbiota, caused by diet, medication use, environmental pollutants, and stress, may lead to imbalances with far-reaching effects on CNS function and mental health. Of interest are short-chain fatty acids (SCFAs), metabolic byproducts produced by gut microbes during fermentation. SCFAs can cross the blood-brain barrier, influencing CNS activity, including microglia and cytokine modulation. The dysregulation of neurotransmitters produced by gut microbes may contribute to CNS disorders, including schizophrenia. This review explores the potential relationship between SCFAs, the gut microbiome, and schizophrenia. Our aim is to deepen the understanding of the gut-brain axis in schizophrenia and to elucidate its implications for future research and therapeutic approaches.


Assuntos
Microbioma Gastrointestinal , Esquizofrenia , Feminino , Gravidez , Humanos , Adulto Jovem , Adulto , Microbioma Gastrointestinal/fisiologia , Eixo Encéfalo-Intestino , Esquizofrenia/microbiologia , Barreira Hematoencefálica , Dieta , Ácidos Graxos Voláteis
6.
Environ Sci Pollut Res Int ; 30(44): 100006-100017, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37624502

RESUMO

In recent years, attention has been focused on the benefit of greenspace on mental health, and it is suggested this link may vary with the type of greenspace. More and more studies have emphasized the influence of the gut microbiome on schizophrenia (SCZ). However, the effects of greenspaces on the gut microbiota in SCZ and the effect of different types of greenspaces on the gut microbiota remain unclear. We aim to examine if there were variations in the effects of various greenspace types on the gut microbiome in SCZ. Besides, we sink to explore important taxonomic compositions associated with different greenspace types. We recruited 243 objects with schizophrenia from Anhui Mental Health Center and collected fecal samples for 16Sr RNA gene sequencing. Three types of greenery coverage were calculated with different circular buffers (800, 1500, and 3000 m) corresponding to individual addresses. The association between greenspace and microbiome composition was analyzed with permutational analysis of variance (PERMANOVA). We conducted the linear regression to capture specific gut microbiome taxa associated with greenery coverage. Tree coverage was consistently associated with microbial composition in both 1500 m (R2 = 0.007, P = 0.030) and 3000 m (R2 = 0.007, P = 0.039). In contrast, there was no association with grass cover in any of the buffer zones. In the regression analysis, higher tree coverage was significantly correlated with the relative abundance of several taxa. Among them, tree coverage was positively associated with increased Bifidobacterium longum (ß = 1.069, P = 0.004), which was the dominant composition in the gut microbiota. The relationship between greenspace and gut microbiome in SCZ differed by the type of greenspace. Besides, "tree coverage" may present a dominant effect on the important taxonomic composition. Our findings might provide instructive evidence for the design of urban greenspace to optimize health and well-being in SCZ as well as the whole people.


Assuntos
Microbioma Gastrointestinal , Esquizofrenia , Humanos , Esquizofrenia/microbiologia , Parques Recreativos , Fezes/microbiologia
7.
Artigo em Inglês | MEDLINE | ID: mdl-37473955

RESUMO

BACKGROUND: Previous studies have reported a variety of gut microbiota alterations in patients with schizophrenia. However, none of these studies has investigated gut microbiota in patients with the deficit subtype of schizophrenia (D-SCZ) that can be characterized by primary and enduring negative symptoms. Therefore, in this study we aimed to profile gut microbiota of individuals with D-SCZ, compared to those with non-deficit schizophrenia (ND-SCZ) and healthy controls (HCs). METHODS: A total of 115 outpatients (44 individuals with D-SCZ and 71 individuals with ND-SCZ) during remission of positive and disorganization symptoms as well as 120 HCs were enrolled. Gut microbiota was analyzed using the 16 rRNA amplicon sequencing. Additionally, the levels of C-reactive protein (CRP), glucose and lipid metabolism markers were determined in the peripheral blood samples. RESULTS: Altogether 14 genera showed differential abundance in patients with D-SCZ compared to ND-SCZ and HCs, including Candidatus Soleaferrea, Eubacterium, Fusobacterium, Lachnospiraceae UCG-002, Lachnospiraceae UCG-004, Lachnospiraceae UCG-010, Libanicoccus, Limosilactobacillus, Mogibacterium, Peptococcus, Prevotella, Prevotellaceae NK3B31 group, Rikenellaceae RC9 gut group, and Slackia after adjustment for potential confounding factors. Observed alterations were significantly associated with cognitive performance in both groups of patients. Moreover, several significant correlations of differentially abundant genera with the levels of CRP, lipid profile parameters, glucose and insulin were found across all subgroups of participants. CONCLUSION: Findings from the present study indicate that individuals with D-SCZ show a distinct profile of gut microbiota alterations that is associated with cognitive performance, metabolic parameters and subclinical inflammation.


Assuntos
Microbioma Gastrointestinal , Esquizofrenia , Humanos , Microbioma Gastrointestinal/genética , Esquizofrenia/microbiologia , Estudos de Casos e Controles , Glucose , Clostridiales
8.
Psychiatry Res ; 326: 115279, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37331068

RESUMO

Although increasing evidence links microbial dysbiosis with the risk for psychiatric symptoms through the microbiome-gut-brain axis (MGBA), the specific mechanisms remain poorly characterized. In a diagnostically heterogeneous group of treated psychiatric cases and nonpsychiatric controls, we characterized the gut and oral microbiome, plasma cytokines, and hippocampal inflammatory processes via proton magnetic resonance spectroscopic imaging (1H-MRSI). Using a transdiagnostic approach, these data were examined in association with schizophrenia-related symptoms measured by the Positive and Negative Syndrome Scale (PANSS). Psychiatric cases had significantly greater heterogeneity of gut alpha diversity and an enrichment of pathogenic taxa, like Veillonella and Prevotella, in the oral microbiome, which was an accurate classifier of phenotype. Cases exhibited significantly greater positive, negative, and general PANSS scores that uniquely correlated with bacterial taxa. Strong, positive correlations of bacterial taxa were also found with cytokines and hippocampal gliosis, dysmyelination, and excitatory neurotransmission. This pilot study supports the hypothesis that the MGBA influences psychiatric symptomatology in a transdiagnostic manner. The relative importance of the oral microbiome in peripheral and hippocampal inflammatory pathways was highlighted, suggesting opportunities for probiotics and oral health to diagnose and treat psychiatric conditions.


Assuntos
Microbioma Gastrointestinal , Microbiota , Esquizofrenia , Humanos , Esquizofrenia/microbiologia , Projetos Piloto , Biomarcadores , Citocinas
9.
J Psychiatr Res ; 162: 140-149, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37156128

RESUMO

The human gut microbiome regulates brain function through the microbiome-gut-brain axis and is implicated in several neuropsychiatric disorders. However, the relationship between the gut microbiome and the pathogenesis of schizophrenia (SCZ) is poorly defined, and very few studies have examined the effect of antipsychotic treatment response. We aim to study the differences in the gut microbiota among drug-naïve (DN SCZ) and risperidone-treated SCZ patients (RISP SCZ), compared to healthy controls (HCs). We recruited a total of 60 participants, from the clinical services of a large neuropsychiatric hospital, which included DN SCZ, RISP SCZ and HCs (n = 20 each). Fecal samples were analyzed using 16s rRNA sequencing in this cross-sectional study. No significant differences were found in taxa richness (alpha diversity) but microbial composition differed between SCZ patients (both DN and RISP) and HCs (PERMANOVA, p = 0.02). Linear Discriminant Analysis Effect Size (LEfSe) and Random Forest model identified the top six genera, which significantly differed in abundance between the study groups. A specific genus-level microbial panel of Ruminococcus, UCG005, Clostridium_sensu_stricto_1 and Bifidobacterium could discriminate SCZ patients from HCs with an area under the curve (AUC) of 0.79, HCs vs DN SCZ (AUC: 0.68), HCs vs RISP SCZ (AUC: 0.93) and DN SCZ vs RISP SCZ (AUC: 0.87). Our study identified distinct microbial signatures that could aid in the differentiation of DN SCZ, RISP SCZ, and HCs. Our findings contribute to a better understanding of the role of the gut microbiome in SCZ pathophysiology and suggest potential targeted interventions.


Assuntos
Microbioma Gastrointestinal , Esquizofrenia , Humanos , Microbioma Gastrointestinal/genética , Esquizofrenia/microbiologia , Estudos Transversais , RNA Ribossômico 16S/genética , Biomarcadores , Fezes/microbiologia
10.
Sci Rep ; 11(1): 18178, 2021 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-34518605

RESUMO

As a severe public health problem, methamphetamine (METH) abuse places a heavy burden on families and society. A growing amount of evidence has indicated communication between gut microbiota and the CNS in drug addiction, with associations to neural, endocrine and immune pathways. Thus, we searched for alterations in the gut microbiota and their potential effects in METH users through 16S rRNA gene sequencing. A decreased Shannon index indicated lower bacterial diversity in the METH users than in the age-matched control group. The gut microbial community composition in the METH users was also altered, including reductions in Deltaproteobacteria and Bacteroidaceae abundances and increases in Sphingomonadales, Xanthomonadales, Romboutsia and Lachnospiraceae abundances. Moreover, the Fusobacteria abundance was correlated with the duration of METH use. Enterobacteriaceae, Ruminococcaceae, Bacteroides, and Faecalibacterium had statistically significant correlations with items related to the positive and negative symptoms of schizophrenia and to general psychopathology in the METH users, and all have previously been reported to be altered in individuals with psychotic syndromes, especially depression. Abstraction, one of the items of the cognitive assessment, was positively related to Blautia. These findings revealed alterations in the gut microbiota of METH users, and these alterations may play a role in psychotic syndrome and cognitive impairment. Although the mechanisms behind the links between these disorders and METH abuse are unknown, the relationships may indicate similarities in the pathogenesis of psychosis induced by METH abuse and other causes, providing a new paradigm for addiction and METH use disorder treatment.


Assuntos
Transtornos Relacionados ao Uso de Anfetaminas/microbiologia , Fezes/microbiologia , Microbioma Gastrointestinal , Metanfetamina/efeitos adversos , Adulto , Fatores Etários , Biodiversidade , Estudos de Casos e Controles , Cognição , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Filogenia , Análise de Componente Principal , Esquizofrenia/microbiologia , Psicologia do Esquizofrênico
11.
Int J Mol Sci ; 22(14)2021 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-34299291

RESUMO

Schizophrenia is a chronic, heterogeneous neurodevelopmental disorder that has complex symptoms and uncertain etiology. Mounting evidence indicates the involvement of genetics and epigenetic disturbances, alteration in gut microbiome, immune system abnormalities, and environmental influence in the disease, but a single root cause and mechanism involved has yet to be conclusively determined. Consequently, the identification of diagnostic markers and the development of psychotic drugs for the treatment of schizophrenia faces a high failure rate. This article surveys the etiology of schizophrenia with a particular focus on gut microbiota regulation and the microbial signaling system that correlates with the brain through the vagus nerve, enteric nervous system, immune system, and production of postbiotics. Gut microbially produced molecules may lay the groundwork for further investigations into the role of gut microbiota dysbiosis and the pathophysiology of schizophrenia. Current treatment of schizophrenia is limited to psychotherapy and antipsychotic drugs that have significant side effects. Therefore, alternative therapeutic options merit exploration. The use of psychobiotics alone or in combination with antipsychotics may promote the development of novel therapeutic strategies. In view of the individual gut microbiome structure and personalized response to antipsychotic drugs, a tailored and targeted manipulation of gut microbial diversity naturally by novel prebiotics (non-digestible fiber) may be a successful alternative therapeutic for the treatment of schizophrenia patients.


Assuntos
Antipsicóticos/uso terapêutico , Microbioma Gastrointestinal/efeitos dos fármacos , Probióticos/uso terapêutico , Esquizofrenia/microbiologia , Esquizofrenia/terapia , Encéfalo/microbiologia , Disbiose/imunologia , Disbiose/metabolismo , Disbiose/microbiologia , Microbioma Gastrointestinal/fisiologia , Humanos , Sistema Imunitário , Prebióticos/microbiologia
12.
Nutrients ; 13(6)2021 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-34205336

RESUMO

Emerging evidence indicates that gut microbiota is important in the regulation of brain activity and cognitive functions. Microbes mediate communication among the metabolic, peripheral immune, and central nervous systems via the microbiota-gut-brain axis. However, it is not well understood how the gut microbiome and neurons in the brain mutually interact or how these interactions affect normal brain functioning and cognition. We summarize the mechanisms whereby the gut microbiota regulate the production, transportation, and functioning of neurotransmitters. We also discuss how microbiome dysbiosis affects cognitive function, especially in neurodegenerative diseases such as Alzheimer's disease and Parkinson's disease.


Assuntos
Cognição/fisiologia , Microbioma Gastrointestinal/fisiologia , Doenças Neurodegenerativas/microbiologia , Neurotransmissores/fisiologia , Doença de Alzheimer/microbiologia , Animais , Ansiedade/microbiologia , Transtorno do Espectro Autista/microbiologia , Encéfalo/fisiopatologia , Depressão/microbiologia , Disbiose/fisiopatologia , Humanos , Doença de Parkinson/microbiologia , Esquizofrenia/microbiologia
13.
Sci Rep ; 11(1): 9743, 2021 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-33963227

RESUMO

The effect of the gut microbiome on the central nervous system and its possible role in mental disorders have received increasing attention. However, knowledge about the relationship between the gut microbiome and brain structure and function is still very limited. Here, we used 16S rRNA sequencing with structural magnetic resonance imaging (sMRI) and resting-state functional (rs-fMRI) to investigate differences in fecal microbiota between 38 patients with schizophrenia (SZ) and 38 demographically matched normal controls (NCs) and explored whether such differences were associated with brain structure and function. At the genus level, we found that the relative abundance of Ruminococcus and Roseburia was significantly lower, whereas the abundance of Veillonella was significantly higher in SZ patients than in NCs. Additionally, the analysis of MRI data revealed that several brain regions showed significantly lower gray matter volume (GMV) and regional homogeneity (ReHo) but significantly higher amplitude of low-frequency fluctuation in SZ patients than in NCs. Moreover, the alpha diversity of the gut microbiota showed a strong linear relationship with the values of both GMV and ReHo. In SZ patients, the ReHo indexes in the right STC (r = - 0.35, p = 0.031, FDR corrected p = 0.039), the left cuneus (r = - 0.33, p = 0.044, FDR corrected p = 0.053) and the right MTC (r = - 0.34, p = 0.03, FDR corrected p = 0.052) were negatively correlated with the abundance of the genus Roseburia. Our results suggest that the potential role of the gut microbiome in SZ is related to alterations in brain structure and function. This study provides insights into the underlying neuropathology of SZ.


Assuntos
Encéfalo , Microbioma Gastrointestinal , Imageamento por Ressonância Magnética , Esquizofrenia , Adulto , Bactérias/classificação , Bactérias/genética , Bactérias/crescimento & desenvolvimento , Encéfalo/diagnóstico por imagem , Encéfalo/fisiopatologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Esquizofrenia/diagnóstico por imagem , Esquizofrenia/microbiologia , Esquizofrenia/fisiopatologia
14.
Nutrients ; 13(4)2021 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-33807241

RESUMO

The gut microbiome (GMB) plays an important role in developmental processes and has been implicated in the etiology of psychiatric disorders. However, the relationship between GMB and schizophrenia remains unclear. In this article, we review the existing evidence surrounding the gut microbiome in schizophrenia and the potential for antipsychotics to cause adverse metabolic events by altering the gut microbiome. We also evaluate the current evidence for the clinical use of probiotic and prebiotic treatment in schizophrenia. The current data on microbiome alteration in schizophrenia remain conflicting. Longitudinal and larger studies will help elucidate the confounding effect on the microbiome. Current studies help lay the groundwork for further investigations into the role of the GMB in the development, presentation, progression and potential treatment of schizophrenia.


Assuntos
Antipsicóticos/efeitos adversos , Microbioma Gastrointestinal/efeitos dos fármacos , Prebióticos , Probióticos , Esquizofrenia/tratamento farmacológico , Esquizofrenia/microbiologia , Antipsicóticos/uso terapêutico , Humanos
15.
Behav Brain Res ; 396: 112886, 2021 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-32890599

RESUMO

Patients with psychosis usually respond to one antipsychotic drug and not to another; one third fail to respond to any. Some patients, who initially do well, stop responding. Some develop serious side effects even at low doses. While several of the reasons for this variability are known, many are not. The aim of this review is to explore the potential role of intestinal organisms in response/non-response to antipsychotics. Much of the literature in this field is relatively new and still, for the most part, theoretical. A growing number of animal experiments and clinical trials are starting to point, however, to substantial effects of antipsychotics on the composition of gut bacteria and, reciprocally, to the effects of microbiota on the pharmacokinetics of antipsychotic medication. Because so many factors influence the constituents of the human intestine, it is difficult, at present, to sort out how much one or more either enhance or dampen the benefits of antipsychotics or the character/severity of the adverse effects they induce. Dietary and other therapies are being devised to reverse dysbiosis. If successful, such therapies plus the modification of factors that, together, are known to determine the composition of microbiota could help to maximize the effectiveness of currently available antipsychotic therapy.


Assuntos
Antipsicóticos/farmacologia , Disbiose , Microbioma Gastrointestinal/efeitos dos fármacos , Esquizofrenia , Animais , Disbiose/tratamento farmacológico , Disbiose/metabolismo , Disbiose/microbiologia , Humanos , Esquizofrenia/tratamento farmacológico , Esquizofrenia/metabolismo , Esquizofrenia/microbiologia
16.
Schizophr Res ; 234: 51-57, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-32334937

RESUMO

It is being increasingly recognized that human mucosal surfaces are not sterile but are colonized with microorganisms collectively known as the microbiome. The microbiome can alter brain functioning in humans and animals by way of a series of interactions operative in the brain-immune-gut interactome. We characterized the oropharyngeal microbiome in 316 individuals, including 121 with schizophrenia, 62 with mania, 48 with major depressive disorder, and 85 controls without a psychiatric disorder. We found that the oropharyngeal microflora of individuals with schizophrenia and individuals with mania differed from controls in composition and abundance as measured by the weighted UniFrac distance (both p < .003 adjusted for covariates and multiple comparisons). This measure in individuals with major depressive disorder did not differ from that of controls. We also identified five bacterial taxa which differed among the diagnostic groups. Three of the taxa, Neisseria subflava, Weeksellaceae, and Prevotella, were decreased in individuals with schizophrenia or mania as compared to controls, while Streptococci was increased in these groups. One taxa, Schlegelella, was only found in individuals with mania. Neisseria subflava was also positively associated with cognitive functioning as measured by the Repeatable Battery for the Assessment of Neuropsychological Status. There were no taxa significantly altered in individuals with major depression. Individuals with schizophrenia and mania have altered compositions of the oropharyngeal microbiome. An understanding of the biology of the microbiome and its effect on the brain might lead to new insights into the pathogenesis, and ultimately, the prevention and treatment of these disorders.


Assuntos
Transtorno Depressivo Maior , Mania , Microbiota , Orofaringe/microbiologia , Esquizofrenia , Comamonadaceae , Transtorno Depressivo Maior/microbiologia , Flavobacteriaceae , Humanos , Mania/microbiologia , Neisseria , Prevotella , Esquizofrenia/microbiologia , Streptococcus
17.
Artigo em Inglês | MEDLINE | ID: mdl-33045322

RESUMO

Cognitive impairment has been consistently found to be a core feature of serious mental illnesses such as schizophrenia and major mood disorders (major depression and bipolar disorder). In recent years, a great effort has been made in elucidating the biological causes of cognitive deficits and the search for new biomarkers of cognition. Microbiome and gut-brain axis (MGB) hormones have been postulated to be potential biomarkers of cognition in serious mental illnesses. The main aim of this review was to synthesize current evidence on the association of microbiome and gut-brain hormones on cognitive processes in schizophrenia and major mood disorders and the association of MGB hormones with stress and the immune system. Our review underscores the role of the MGB axis on cognitive aspects of serious mental illnesses with the potential use of agents targeting the gut microbiota as cognitive enhancers. However, the current evidence for clinical trials focused on the MGB axis as cognitive enhancers in these clinical populations is scarce. Future clinical trials using probiotics, prebiotics, antibiotics, or faecal microbiota transplantation need to consider potential mechanistic pathways such as the HPA axis, the immune system, or gut-brain axis hormones involved in appetite control and energy homeostasis.


Assuntos
Eixo Encéfalo-Intestino/fisiologia , Cognição/fisiologia , Microbiota/fisiologia , Transtornos do Humor/psicologia , Esquizofrenia/microbiologia , Psicologia do Esquizofrênico , Humanos , Transtornos do Humor/microbiologia , Probióticos/uso terapêutico
18.
Psychiatry Res ; 291: 113260, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32763534

RESUMO

Exploring the gut microbiota characteristics of patients with acute and remission schizophrenia (SCZ) and evaluating the potential of the gut microbiome as a non-invasive biomarker for SCZ. A total of 87 fecal samples were collected, including a total of 58 samples from 29 SCZ patients over two different periods (remission and onset period) and 29 samples from the control group for 16S rRNA Miseq.The changes of intestinal microbiota in SCZ patients from remission to onset were analyzed, and a random forest model was constructed to recognize biomarkers. The optimal three genus-level diagnosis biomarkers were identified through an AUC validation on a random forest model, furthermore, an AUC of 0.76 (95% CI (0.63, 0.89)) was achieved between 29 aSCZ and 29 HCs. Compared with the control group, the first 11 OUT-level' biomarkers were identified in rSCZ group. As a status marker of the disease, the AUC of 0.7 (95% CI (0.56, 0.84)) was achieved between 29 rSCZ and 29 HCs. There were differences between SCZ patients and HCs, acute and remission patients as well, suggesting that the potential of the gut microbiota as a non-invasive diagnostic tool. Moreover, the features of the gut microbiome of SCZ provide clues for disease prognosis assessment and targeted intervention.


Assuntos
Biodiversidade , Microbioma Gastrointestinal/genética , RNA Ribossômico 16S/genética , Esquizofrenia/diagnóstico , Esquizofrenia/genética , Adulto , Estudos de Coortes , Fezes/microbiologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Esquizofrenia/microbiologia
19.
Nutrients ; 12(4)2020 04 08.
Artigo em Inglês | MEDLINE | ID: mdl-32276499

RESUMO

BACKGROUND AND SIGNIFICANCE: There is a need to develop new hypothesis-driven treatment for both both major depression (MD) and schizophrenia in which the risk of depression is 5 times higher than the general population. Major depression has been also associated with poor illness outcomes including pain, metabolic disturbances, and less adherence. Conventional antidepressants are partly effective, and 44% of the subjects remain unremitted under treatment. Improving MD treatment efficacy is thus needed to improve the SZ prognosis. Microbiota-orientated treatments are currently one of the most promising tracks. METHOD: This work is a systematic review synthetizing data of arguments to develop microbiota-orientated treatments (including fecal microbiota transplantation (FMT)) in major depression and schizophrenia. RESULTS: The effectiveness of probiotic administration in MD constitutes a strong evidence for developing microbiota-orientated treatments. Probiotics have yielded medium-to-large significant effects on depressive symptoms, but it is still unclear if the effect is maintained following probiotic discontinuation. Several factors may limit MD improvement when using probiotics, including the small number of bacterial strains administered in probiotic complementary agents, as well as the presence of a disturbed gut microbiota that probably limits the probiotics' impact. FMT is a safe technique enabling to improve microbiota in several gut disorders. The benefit/risk ratio of FMT has been discussed and has been recently improved by capsule administration. CONCLUSION: Cleaning up the gut microbiota by transplanting a totally new human gut microbiota in one shot, which is referred to as FMT, is likely to strongly improve the efficacy of microbiota-orientated treatments in MD and schizophrenia and maintain the effect over time. This hypothesis should be tested in future clinical trials.


Assuntos
Terapia Biológica/métodos , Transtorno Depressivo Maior/microbiologia , Transtorno Depressivo Maior/terapia , Esquizofrenia/microbiologia , Esquizofrenia/terapia , Adulto , Transplante de Microbiota Fecal , Feminino , Humanos , Masculino , Microbiota , Pessoa de Meia-Idade , Probióticos/uso terapêutico , Resultado do Tratamento , Adulto Jovem
20.
Nat Commun ; 11(1): 1612, 2020 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-32235826

RESUMO

Evidence is mounting that the gut-brain axis plays an important role in mental diseases fueling mechanistic investigations to provide a basis for future targeted interventions. However, shotgun metagenomic data from treatment-naïve patients are scarce hampering comprehensive analyses of the complex interaction between the gut microbiota and the brain. Here we explore the fecal microbiome based on 90 medication-free schizophrenia patients and 81 controls and identify a microbial species classifier distinguishing patients from controls with an area under the receiver operating characteristic curve (AUC) of 0.896, and replicate the microbiome-based disease classifier in 45 patients and 45 controls (AUC = 0.765). Functional potentials associated with schizophrenia include differences in short-chain fatty acids synthesis, tryptophan metabolism, and synthesis/degradation of neurotransmitters. Transplantation of a schizophrenia-enriched bacterium, Streptococcus vestibularis, appear to induces deficits in social behaviors, and alters neurotransmitter levels in peripheral tissues in recipient mice. Our findings provide new leads for further investigations in cohort studies and animal models.


Assuntos
Microbioma Gastrointestinal/fisiologia , Metagenoma , Esquizofrenia/metabolismo , Esquizofrenia/microbiologia , Animais , Bactérias/classificação , Bactérias/genética , Comportamento Animal , Modelos Animais de Doenças , Transplante de Microbiota Fecal , Fezes/microbiologia , Microbioma Gastrointestinal/genética , Humanos , Masculino , Metagenômica/métodos , Camundongos , Camundongos Endogâmicos C57BL , RNA Ribossômico 16S , Curva ROC , Fatores de Risco , Comportamento Social , Streptococcus
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA