Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
FASEB J ; 36(1): e22100, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34939244

RESUMO

This study aims to elucidate the relationships between gut microbiota, bile acid metabolism, and psychological comorbidity in Crohn's disease (CD). We profiled the fecal microbiota composition and quantified the bile acid pool of 39 CD patients and 14 healthy controls using 16S rRNA gene sequencing and liquid chromatography-tandem mass spectrometry, respectively. Significant reductions in the secondary bile acids, LCA and DCA, were found in both the feces and serum samples of CD patients, while the concentration of 7-DHCA was particularly higher in the serum of CD patients with psychological disorders. The fecal levels of HDCA and 12-DHCA of the CD patients were inversely correlated with their Self-Rated Depression Scale (SDS) scores, whereas the serum level of 7-DHCA was positively correlated with the SDS scores. In addition, the fecal levels of TDCA, TLCA, and TßMCA showed a positive correlation with the Self-Rated Anxiety Scale (SAS) scores. The fecal microbiota biodiversity was particularly declined in CD patients with psychological disorders. An enrichment of Ruminococcus gnavus in CD patients may cause psychological disorders by affecting the microbiota-gut-brain axis via its ability to degrade the gut barrier, regulate the tryptophan-kynurenine metabolism, and modulate bile acid metabolism. In addition, the overabundant Enterobacteriaceae and Lachnospiraceae in CD patients may contribute to psychological comorbidity via dysregulating their bile acids metabolism. Taken together, changes in the gut microbiota composition may cooperate with alterations in the bile acid metabolism that are involved in the development of psychological disorders in CD.


Assuntos
Ácidos e Sais Biliares/metabolismo , Clostridiales/metabolismo , Doença de Crohn , Disbiose , Enterobacteriaceae/metabolismo , Microbioma Gastrointestinal , Transtornos Mentais , Adulto , Doença de Crohn/metabolismo , Doença de Crohn/microbiologia , Doença de Crohn/psicologia , Disbiose/metabolismo , Disbiose/microbiologia , Disbiose/psicologia , Enterobacteriaceae/classificação , Feminino , Humanos , Masculino , Transtornos Mentais/metabolismo , Transtornos Mentais/microbiologia , Transtornos Mentais/psicologia
2.
J Nutr Biochem ; 99: 108865, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34582967

RESUMO

Pain-related functional gastrointestinal disorders (FGIDs) are characterized by visceral hypersensitivity (VHS) associated with alterations in the microbiota-gut-brain axis. Since human milk oligosaccharides (HMOs) modulate microbiota, gut and brain, we investigated whether HMOs impact VHS, and explored the role of gut microbiota. To induce VHS, C57BL/6JRj mice received hourly water avoidance stress (WAS) sessions for 10 d, or antibiotics (ATB) for 12 d. Challenged and unchallenged (Sham) animals were fed AIN93M diet (Cont) or AIN93M containing 1% of a 6-HMO mix (HMO6). VHS was assessed by monitoring the visceromotor response to colorectal distension. Fecal microbiome was analyzed by shotgun metagenomics. The effect of HMO6 sub-blends on VHS and nociceptive pathways was further tested using the WAS model. In mice fed Cont, WAS and ATB increased the visceromotor response to distension. HMO6 decreased WAS-mediated electromyographic rise at most distension volumes and overall Area Under Curve (AUC=6.12±0.50 in WAS/HMO6 vs. 9.46±0.50 in WAS/Cont; P<.0001). In contrast, VHS in ATB animals was not improved by HMO6. In WAS, HMO6 promoted most microbiota taxa and several functional pathways associated with low VHS and decreased those associated with high VHS. Among the sub-blends, 2'FL+DFL and LNT+6'SL reduced visceromotor response close to Sham/Cont values and modulated serotoninergic and CGRPα-related pathways. This research further substantiates the capacity of HMOs to modulate the microbiota-gut-brain communication and identifies mitigation of abdominal pain as a new HMO benefit. Ultimately, our findings suggest the value of specific HMO blends to alleviate pain associated FGIDs such as infantile colic or Irritable Bowel Syndrome.


Assuntos
Dor Abdominal/dietoterapia , Disbiose/dietoterapia , Microbioma Gastrointestinal , Leite Humano/metabolismo , Oligossacarídeos/metabolismo , Dor Abdominal/metabolismo , Dor Abdominal/microbiologia , Dor Abdominal/psicologia , Animais , Bactérias/classificação , Bactérias/genética , Bactérias/isolamento & purificação , Bactérias/metabolismo , Disbiose/metabolismo , Disbiose/microbiologia , Disbiose/psicologia , Fezes/microbiologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Oligossacarídeos/análise , Estresse Psicológico
3.
Gut Microbes ; 13(1): 2000275, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34856844

RESUMO

An array of chronic inflammatory diseases, including metabolic diseases such as obesity and diabetes, are thought to be promoted by disturbance of the intestinal microbiota. Such diseases disproportionately impact low-income communities, which are frequently afflicted by chronic stress and increased density housing. Hence, we hypothesized that overcrowded housing might promote stress, microbiota dysbiosis, inflammation, and, consequently, metabolic diseases. We tested this hypothesis in a tractable murine model of social overcrowding (SOC), in which mice were housed at twice normal density. SOC moderately impacted behavior in some widely used assays (Open Field, Elevated Plus Maze and Light/Dark tests) and resulted in a stark increase in corticosterone levels. Such indices of stress were associated with mild chronic gut inflammation, hyperglycemia, elevations in colonic cytokines, and alterations in gut microbiota composition. All of these consequences of SOC were eliminated by broad spectrum antibiotics, while some (inflammation and hyperglycemia) were transmitted by microbiota transplantation from SOC mice to germfree mice housed at normal density. Altogether, these results suggest a central role for intestinal microbiota in driving stress, inflammation, and chronic diseases that are promoted by overcrowded housing.


Assuntos
Aglomeração/psicologia , Microbioma Gastrointestinal , Estresse Psicológico/metabolismo , Estresse Psicológico/microbiologia , Animais , Antibacterianos/uso terapêutico , Glicemia/metabolismo , Corticosterona/metabolismo , Citocinas/metabolismo , Disbiose/metabolismo , Disbiose/microbiologia , Disbiose/psicologia , Disbiose/terapia , Transplante de Microbiota Fecal , Microbioma Gastrointestinal/efeitos dos fármacos , Trato Gastrointestinal/metabolismo , Trato Gastrointestinal/microbiologia , Hiperglicemia/microbiologia , Inflamação , Camundongos , Estresse Psicológico/psicologia , Estresse Psicológico/terapia
4.
Nutrients ; 13(11)2021 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-34836074

RESUMO

Children with autism spectrum disorder (ASD) report a higher frequency and severity of gastrointestinal disorders (GID) than typically developing (TD) children. GID-associated discomfort increases feelings of anxiety and frustration, contributing to the severity of ASD. Emerging evidence supports the biological intersection of neurodevelopment and microbiome, indicating the integral contribution of GM in the development and function of the nervous system, and mental health, and disease balance. Dysbiotic GM could be a contributing factor in the pathogenesis of GID in children with ASD. High-fat diets may modulate GM through accelerated growth of bile-tolerant bacteria, altered bacterial ratios, and reduced bacterial diversity, which may increase the risk of GID. Notably, saturated fatty acids are considered to have a pronounced effect on the increase of bile-tolerant bacteria and reduction in microbial diversity. Additionally, omega-3 exerts a favorable impact on GM and gut health due to its anti-inflammatory properties. Despite inconsistencies in the data elaborated in the review, the dietary fat composition, as part of an overall dietary intervention, plays a role in modulating GID, specifically in ASD, due to the altered microbiome profile. This review emphasizes the need to conduct future experimental studies investigating the effect of diets with varying fatty acid compositions on GID-specific microbiome profiles in children with ASD.


Assuntos
Transtorno do Espectro Autista/microbiologia , Gorduras na Dieta/farmacologia , Disbiose/psicologia , Gastroenteropatias/psicologia , Microbioma Gastrointestinal/efeitos dos fármacos , Eixo Encéfalo-Intestino/efeitos dos fármacos , Criança , Dieta/efeitos adversos , Dieta/psicologia , Humanos
5.
Nutrients ; 13(5)2021 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-34062976

RESUMO

Dysbiosis of the gut microbiota has been associated with different illnesses and emotional disorders such as stress. Traditional fermented foods that are rich in probiotics suggest modulation of dysbiosis, which protects against stress-induced disorders. The academic stress was evaluated in medical students using the SISCO Inventory of Academic Stress before and after ingestion of an aguamiel-based beverage fermented with Lactobacillus plantarum, Lactobacillus paracasei and Lactobacillus brevis (n = 27) and a control group (n = 18). In addition, microbial phyla in feces were quantified by qPCR. The results showed that the consumption of 100 mL of a beverage fermented with lactic acid bacteria (3 × 108 cfu/mL) for 8 weeks significantly reduced academic stress (p = 0.001), while the control group (placebo intervention) had no significant changes in the perception of academic stress (p = 0.607). Significant change (p = 0.001) was shown in the scores for environmental demands, and physical and psychological factors. Consumption of the fermented beverage significantly increased the phyla Firmicutes and Bacteroidetes but not Gammaproteobacteria. No significant changes were found in the control group, except for a slight increase in the phylum Firmicutes. The intake of this fermented beverage suggest a modulation of gut microbiota and possible reduction in stress-related symptoms in university students, without changing their lifestyle or diet.


Assuntos
Agave , Alimentos Fermentados/microbiologia , Probióticos/administração & dosagem , Estresse Psicológico/terapia , Estudantes de Medicina/psicologia , Adulto , Disbiose/microbiologia , Disbiose/psicologia , Fezes/microbiologia , Feminino , Fermentação , Microbioma Gastrointestinal , Humanos , Lactobacillales , Estudos Longitudinais , Masculino , México , Estudos Prospectivos , Método Simples-Cego , Estresse Psicológico/microbiologia , Adulto Jovem
6.
Neuropharmacology ; 192: 108598, 2021 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-33965398

RESUMO

Epidemiology and clinical research indicate that only a subset of people who are exposed to drugs of abuse will go on to develop a substance use disorder. Numerous factors impact individual susceptibility to developing a substance use disorder, including intrinsic biological factors, environmental factors, and interpersonal/social factors. Given the extensive morbidity and mortality that is wrought as a consequence of substance use disorders, a substantial body of research has focused on understanding the risk factors that mediate the shift from initial drug use to pathological drug use. Understanding these risk factors provides a clear path for the development of risk mitigation strategies to help reduce the burden of substance use disorders in the population. Here we will review the rapidly growing body of literature that examines the importance of interactions between the peripheral immune system, the gut microbiome, and the central nervous system (CNS) in mediating the transition to pathological drug use. While these systems had long been viewed as distinct, there is growing evidence that there is bidirectional communication between both the immune system and the gut microbiome that drive changes in neural and behavioral plasticity relevant to substance use disorders. Further, both of these systems are highly sensitive to environmental perturbations and are implicated in numerous neuropsychiatric conditions. While the field of study examining these interactions in substance use disorders is in its relative infancy, clarifying the relationship between gut-immune-brain signaling and substance use disorders has potential to improve our understanding of individual propensity to developing addiction and yield important insight into potential treatment options.


Assuntos
Eixo Encéfalo-Intestino/fisiologia , Encéfalo/imunologia , Microbioma Gastrointestinal/fisiologia , Neuroimunomodulação/fisiologia , Transtornos Relacionados ao Uso de Substâncias/imunologia , Comportamento Aditivo/imunologia , Comportamento Aditivo/metabolismo , Comportamento Aditivo/psicologia , Encéfalo/metabolismo , Disbiose/imunologia , Disbiose/psicologia , Humanos , Transtornos Relacionados ao Uso de Substâncias/metabolismo , Transtornos Relacionados ao Uso de Substâncias/psicologia
7.
Sci Rep ; 11(1): 9915, 2021 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-33972646

RESUMO

Psychological stress has been reported to relate to dysbiosis, imbalance of the intestinal microbiota composition, and contribute to the onset and exacerbation of depression, though, underlying mechanisms of psychological stress-related dysbiosis have been unknown. It has been previously established that α-defensins, which are effector peptides of innate enteric immunity produced by Paneth cells in the small intestine, play an important role in regulation of the intestinal microbiota. However, the relationship between disruption of intestinal ecosystem and α-defensin under psychological stress is yet to be determined. Here we show using chronic social defeat stress (CSDS), a mouse depression model that (1) the exposure to CSDS significantly reduces α-defensin secretion by Paneth cells and (2) induces dysbiosis and significant composition changes in the intestinal metabolites. Furthermore, (3) they are recovered by administration of α-defensin. These results indicate that α-defensin plays an important role in maintaining homeostasis of the intestinal ecosystem under psychological stress, providing novel insights into the onset mechanism of stress-induced depression, and may further contribute to discovery of treatment targets for depression.


Assuntos
Depressão/imunologia , Disbiose/imunologia , Estresse Psicológico/complicações , alfa-Defensinas/metabolismo , Administração Oral , Animais , Depressão/tratamento farmacológico , Depressão/microbiologia , Depressão/psicologia , Modelos Animais de Doenças , Disbiose/tratamento farmacológico , Disbiose/microbiologia , Disbiose/psicologia , Fezes/microbiologia , Microbioma Gastrointestinal/imunologia , Humanos , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Intestino Delgado/imunologia , Intestino Delgado/metabolismo , Intestino Delgado/microbiologia , Intestino Delgado/patologia , Masculino , Camundongos , Celulas de Paneth/imunologia , Celulas de Paneth/metabolismo , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Derrota Social , Estresse Psicológico/tratamento farmacológico , Estresse Psicológico/imunologia , Estresse Psicológico/psicologia , alfa-Defensinas/administração & dosagem , alfa-Defensinas/isolamento & purificação
8.
Am Heart J ; 239: 27-37, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33984318

RESUMO

BACKGROUND: Hypertension (HTN) is frequently linked with depression (DEP) in adults with cardiovascular disease (CVD), yet the underlying mechanism and successful management remain elusive. We approached this knowledge gap through the lens that humans are eukaryote-prokaryote "meta-organisms," such that cardiovascular disease dysregulation is a mosaic disorder involving dysbiosis of the gut. We hypothesized that patients diagnosed with hypertension plus depression harbor a unique gut microbial ecology with attending functional genomics engaged with their hosts' gut/brain axis physiology. METHODS: Stool microbiome DNA was analyzed by whole metagenome shotgun sequencing in 54 subjects parsed into cohorts diagnosed with HTN only (N = 18), DEP only (N = 7), DEP plus HTN (DEP-HTN) (N = 8), or reference subjects with neither HTN nor DEP (N = 21). A novel battery of machine-learning multivariate analyses of de-noised data yielded effect sizes and permutational covariance-based dissimilarities that significantly differentiated the cohorts (false discovery rate (FDR)-adjusted P ≤ .05); data clustering within 95% confidence interval). RESULTS: Metagenomic significant differences extricated the four cohorts. Data of the cohort exhibiting DEP-HTN were germane to the interplay of central control of blood pressure concomitant with the neuropathology of depressive disorders. DEP-HTN gut bacterial community ecology was defined by co-occurrence of Eubacterium siraeum, Alistipes obesi, Holdemania filiformis, and Lachnospiraceae bacterium 1.1.57FAA with Streptococcus salivariu. The corresponding microbial functional genomics of DEP-HTN engaged pathways degrading GABA and beneficial short chain fatty acids (SCFA), and are associated with enhanced sodium absorption and inflammasome induction. CONCLUSIONS: These data suggest a new putative endotype of hypertension, which we denote "depressive-hypertension" (DEP-HTN), for which we posit a model that is distinctive from either HTN alone or DEP alone. An "endotype" is a subtype of a heterogeneous pathophysiological mechanism. The DEP-HTN model incorporates a unique signature of microbial taxa and functional genomics with crosstalk that putatively intertwines host pathophysiology involving the gastrointestinal tract with disruptions in central control of blood pressure and mood. The DEP-HTN endotype model engages cardiology with gastroenterology and psychiatry, providing a proof-of-concept foundation to explore future treatments, diagnosis, and prevention of HTN-coupled mood disorders.


Assuntos
Afeto/fisiologia , Biota/genética , Depressão , Disbiose , Microbioma Gastrointestinal , Hipertensão , Adulto , Ciências Biocomportamentais , Depressão/diagnóstico , Depressão/metabolismo , Depressão/fisiopatologia , Disbiose/diagnóstico , Disbiose/fisiopatologia , Disbiose/psicologia , Fezes/microbiologia , Feminino , Microbioma Gastrointestinal/genética , Microbioma Gastrointestinal/fisiologia , Trato Gastrointestinal/microbiologia , Trato Gastrointestinal/fisiopatologia , Humanos , Hipertensão/diagnóstico , Hipertensão/metabolismo , Hipertensão/psicologia , Aprendizado de Máquina , Masculino , Redes e Vias Metabólicas , Metagenoma
9.
Nutrients ; 13(3)2021 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-33804493

RESUMO

Despite the beneficial actions of antibiotics against bacterial infections, the use of antibiotics is a crucial etiological factor influencing microbial dysbiosis-associated adverse outcomes in human health. Based on the assumption that gut microbial dysbiosis can provoke behavioral or psychological disorders, the present study evaluated anxiety-linked behavioral changes in a mouse model of streptomycin-induced dysbiosis. Measuring anxiety-like behavior using the light-dark box and elevated plus maze tests indicated that streptomycin treatment caused acute anxiety in mice. As an intervention for dysbiosis-associated distress, the probiotic strain Escherichia coli Nissle 1917 (EcN) was evaluated for its effects on streptomycin-induced behavioral changes in mice. EcN supplementation persistently ameliorated anxiety responses in mice with streptomycin-induced dysbiosis. As an outcome of anxiety, body weight changes were marginally affected by antibiotic treatment. However, mice supplemented with EcN displayed acute retardation of body weight gain, since EcN is known to reduce food intake and increase energy expenditure. Taken together, EcN treatment prominently counteracted streptomycin-induced anxiety in mice, with the metabolically beneficial retardation of body weight gain. The present model simulates psychological disorders in antibiotic users. As a promising intervention, EcN treatment can facilitate psychological relief under conditions of dysbiotic stress by blocking the pathologic gut-brain circuit.


Assuntos
Antibacterianos/farmacologia , Ansiedade/tratamento farmacológico , Disbiose/psicologia , Escherichia coli , Probióticos/farmacologia , Animais , Ansiedade/induzido quimicamente , Ansiedade/microbiologia , Suplementos Nutricionais , Disbiose/induzido quimicamente , Camundongos , Estreptomicina
10.
Medicine (Baltimore) ; 100(10): e18621, 2021 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-33725807

RESUMO

INTRODUCTION: Autism spectrum disorder (ASD) is a neurodevelopmental disorder with increasing incidence. The externalizing and internalizing problems among children with ASD often persistent and highly impair functioning of both the child and the family. Children with ASD often develop gut-related comorbidities and dysbiosis can have negative effects on not only the gastrointestinal (GI) tract, but also psychological symptoms. Dietary exclusions and probiotic supplements also have been investigated in the management of ASD symptoms. Especially, there is some anecdotal evidence that probiotics supplements are able to alleviate GI symptoms as well as improve behaviors in children with ASD. METHOD AND ANALYSIS: This review will report on overall studies that include randomized control trials, randomized cross-over studies and cluster-randomized trials designs that consider curative effect in children with ASD by probiotic supplements. We will search 6 databases: MEDLINE, Embase, Scopus, PubMed, The Cochrane Library, and Web of Science and we will perform a manual search the journal Autism and information of ongoing or unpublished studies. The Mixed Methods Appraisal Tool (MMAT) will be used to assess quality of articles and the Jadad scale will be used to assess for bias. Assessment of publication bias will be performed using funnel plots generated by Comprehensive Meta-Analysis (CMA) 3.0 software. Clarifying the evidence in this area will be important for future research directions when reformulating and promoting the therapeutic regime in the field. ETHICS AND DISSEMINATION: There are no human participants, data, or tissue being directly studied for the purposes of the review; therefore, ethics approval and consent to participate are not applicable. The results of this study will be presented at conferences and published in peer-reviewed journals. REGISTRATION AND STATUS: PROSPERO 2019 CRD42019132754.


Assuntos
Transtorno do Espectro Autista/dietoterapia , Disbiose/dietoterapia , Probióticos/administração & dosagem , Transtorno do Espectro Autista/epidemiologia , Transtorno do Espectro Autista/psicologia , Criança , Comorbidade , Disbiose/epidemiologia , Disbiose/psicologia , Humanos , Metanálise como Assunto , Revisões Sistemáticas como Assunto , Resultado do Tratamento
11.
Nutrients ; 13(2)2021 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-33669988

RESUMO

The gut microbiota (GM) represents a diverse and dynamic population of microorganisms and about 100 trillion symbiotic microbial cells that dwell in the gastrointestinal tract. Studies suggest that the GM can influence the health of the host, and several factors can modify the GM composition, such as diet, drug intake, lifestyle, and geographical locations. Gut dysbiosis can affect brain immune homeostasis through the microbiota-gut-brain axis and can play a key role in the pathogenesis of neurodegenerative diseases, including dementia and Alzheimer's disease (AD). The relationship between gut dysbiosis and AD is still elusive, but emerging evidence suggests that it can enhance the secretion of lipopolysaccharides and amyloids that may disturb intestinal permeability and the blood-brain barrier. In addition, it can promote the hallmarks of AD, such as oxidative stress, neuroinflammation, amyloid-beta formation, insulin resistance, and ultimately the causation of neural death. Poor dietary habits and aging, along with inflammatory responses due to dysbiosis, may contribute to the pathogenesis of AD. Thus, GM modulation through diet, probiotics, or fecal microbiota transplantation could represent potential therapeutics in AD. In this review, we discuss the role of GM dysbiosis in AD and potential therapeutic strategies to modulate GM in AD.


Assuntos
Doença de Alzheimer/microbiologia , Encéfalo/microbiologia , Disbiose/terapia , Microbioma Gastrointestinal/fisiologia , Mucosa Intestinal/metabolismo , Animais , Barreira Hematoencefálica/microbiologia , Dietoterapia , Disbiose/metabolismo , Disbiose/psicologia , Transplante de Microbiota Fecal , Humanos , Permeabilidade , Probióticos/uso terapêutico
12.
CNS Neurosci Ther ; 27(5): 505-514, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33464726

RESUMO

Various neurological disorders, such as stroke and Alzheimer's disease (AD), involve neuroinflammatory responses. The advent of the gut-brain axis enhances our understanding of neurological disease progression and secondary cell death. Gut microbiomes, especially those associated with inflammation, may reflect the dysbiosis of both the brain and the gut, opening the possibility to utilize inflammatory microbiomes as biomarkers and therapeutic targets. The gut-brain axis may serve as a contributing factor to disease pathology and offer innovative approaches in cell-based regenerative medicine for the treatment of neurological diseases. In reviewing the pathogenesis of stroke and AD, we also discuss the effects of gut microbiota on cognitive decline and brain pathology. Although the underlying mechanism of primary cell death from either disease is clearly distinct, both may be linked to gut-microbial dysfunction as a consequential aberration that is unique to each disease. Targeting peripheral cell death pathways that exacerbate disease symptoms, such as those arising from the gut, coupled with conventional central therapeutic approach, may improve stroke and AD outcomes.


Assuntos
Doença de Alzheimer/psicologia , Eixo Encéfalo-Intestino , Transtornos Cognitivos/etiologia , Disbiose/etiologia , Microbioma Gastrointestinal , Acidente Vascular Cerebral/complicações , Transtornos Cognitivos/microbiologia , Transtornos Cognitivos/psicologia , Disbiose/microbiologia , Disbiose/psicologia , Humanos , Acidente Vascular Cerebral/psicologia
13.
Nutrients ; 12(11)2020 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-33182607

RESUMO

Excessive expression of interleukin (IL)-1ß in the brain causes depression and cognitive dysfunction. Herein, we investigated the effect of Lactobacillus gasseri NK109, which suppressed IL-1ß expression in activated macrophages, on Escherichia coli K1-induced cognitive impairment and depression in mice. Germ-free and specific pathogen-free mice with neuropsychiatric disorders were prepared by oral gavage of K1. NK109 alleviated K1-induced cognition-impaired and depressive behaviors, decreased the expression of IL-1ß and populations of NF-κB+/Iba1+ and IL-1R+ cells, and increased the K1-suppressed population of BDNF+/NeuN+ cells in the hippocampus. However, its effects were partially attenuated by celiac vagotomy. NK109 treatment mitigated K1-induced colitis and gut dysbiosis. Tyndallized NK109, even if lysed, alleviated cognitive impairment and depression. In conclusion, NK109 alleviated neuropsychiatric disorders and colitis by modulating IL-1ß expression, gut microbiota, and vagus nerve-mediated gut-brain signaling.


Assuntos
Colite/terapia , Escherichia coli/fisiologia , Microbioma Gastrointestinal/fisiologia , Interleucina-1beta/metabolismo , Lactobacillus gasseri/fisiologia , Interações Microbianas/fisiologia , Animais , Comportamento Animal , Disfunção Cognitiva , Colite/psicologia , Depressão , Disbiose/psicologia , Disbiose/terapia , Escherichia coli/imunologia , Fezes/microbiologia , Microbioma Gastrointestinal/imunologia , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Lactobacillus gasseri/imunologia , Lipopolissacarídeos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Probióticos/administração & dosagem
14.
Nutrients ; 12(9)2020 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-32882999

RESUMO

Behavior and mood disorders have been linked to gut microbiota dysbiosis through the "microbiota-gut-brain axis". Microbiota-targeting interventions are promising therapeutic modalities to restore or even maintain normal microbiome composition and activity in these disorders. Here, we test the impact of a commercial synbiotic formulation on gut microbiota composition and metabolic activity. We employed an ex-vivo continuous fermentation model that simulates the proximal colon to assess the effect of this formulation on microbiota structure and functionality as compared to no treatment control and microcrystalline cellulose as a dietary fiber control. The test formulation did not alter the diversity of gut microbiota over 48 h of treatment. However, it induced the enrichment of Lactobacillus, Collinsella and Erysipelotrichaceae. The test formulation significantly increased the level of microbiota-generated butyrate within 12 h of treatment as compared to 24 h required by microcrystalline cellulose to boost its production. The test formulation did not lead to a significant change in amino acid profiles. These results provide evidence of potential benefits related to synbiotic effects and general gut health and support the potential of this food formulation as a therapeutic dietary intervention in mood and behavior disorders.


Assuntos
Colo/microbiologia , Ingredientes de Alimentos/microbiologia , Microbioma Gastrointestinal , Prebióticos/administração & dosagem , Simbióticos/administração & dosagem , Encéfalo/microbiologia , Butiratos/metabolismo , Fibras na Dieta/farmacologia , Disbiose/dietoterapia , Disbiose/psicologia , Fermentação , Alimentos Formulados/microbiologia , Humanos , Transtornos Mentais/dietoterapia , Transtornos Mentais/microbiologia , Transtornos do Humor/dietoterapia , Transtornos do Humor/microbiologia
15.
Curr Gastroenterol Rep ; 22(11): 53, 2020 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-32827270

RESUMO

PURPOSE OF REVIEW: To analyze current research involving elite athletes and the gut microbiome, with the goal to expand knowledge of potential sports performance strategies and interventions. RECENT FINDINGS: Most of the current research focuses on theoretical applications from microbiome research and known characteristics of performance, as few studies have actually been conducted in elite athletes. However, of the studies that have been conducted, the research is promising that dietary strategies may provide targeted interventions in the future. The majority of sports nutrition takes an external approach to diet: manipulating macronutrient intake based on type of training and performance goals. However, shifting to an internal focus, prioritizing the health of the gut microbiome, could be a new frontier for a competitive edge in athletics.


Assuntos
Desempenho Atlético , Microbioma Gastrointestinal/fisiologia , Trato Gastrointestinal/fisiologia , Esportes/fisiologia , Esportes/psicologia , Analgésicos/efeitos adversos , Animais , Bebidas/efeitos adversos , Osso e Ossos/fisiologia , Disbiose/psicologia , Microbioma Gastrointestinal/efeitos dos fármacos , Trato Gastrointestinal/efeitos dos fármacos , Trato Gastrointestinal/microbiologia , Humanos , Sistema Hipotálamo-Hipofisário/fisiologia , Estado de Hidratação do Organismo
17.
Artigo em Inglês | MEDLINE | ID: mdl-31689444

RESUMO

The pathoetiology and pathophysiology of borderline personality disorder (BPD) have been relatively under-explored. Consequently, no targetted pharmaceutical treatments or preventative interventions are available. The current article reviews the available data on the biological underpinnings of BPD, highlighting a role for early developmental processes, including prenatal stress and maternal dysbiosis, in BPD pathoetiology. Such factors are proposed to drive alterations in the infant's gut microbiome, in turn modulating amygdala development and the amygdala's two-way interactions with other brain regions. Alterations in opioidergic activity, including variations in the ratio of the mu-and kappa-opioid receptors seem a significant aspect of BPD pathophysiology, contributing to its comorbidities with depression, anxiety, impulsivity and addiction. Stress and dysphoria are commonly experienced in people classed with BPD. A growing body of data, across a host of medical conditions, indicate that stress and mood dysregulation may be intimately associated with gut dysbiosis and increased gut permeability, coupled to heightened levels of oxidative stress and immune-inflammatory activity. It urgently requires investigation as to the relevance of such gut changes in the course of BPD symptomatology. Accumulating data indicates that BPD symptom exacerbations may be linked to cyclical variations in estrogen, in turn decreasing serotonin and local melatonin synthesis, and thereby overlapping with the pathophysiology of migraine and endometriosis, which also have a heightened association with BPD. Future research directions and treatment implications are indicated.


Assuntos
Tonsila do Cerebelo/fisiopatologia , Transtorno da Personalidade Borderline/etiologia , Transtorno da Personalidade Borderline/fisiopatologia , Microbioma Gastrointestinal/fisiologia , Córtex Pré-Frontal/fisiopatologia , Receptores Opioides kappa/genética , Receptores Opioides mu/genética , Adulto , Tonsila do Cerebelo/metabolismo , Transtorno da Personalidade Borderline/mortalidade , Disbiose/complicações , Disbiose/psicologia , Feminino , Humanos , Recém-Nascido , Transtornos Mentais/complicações , Córtex Pré-Frontal/metabolismo , Gravidez , Efeitos Tardios da Exposição Pré-Natal , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/metabolismo , Estresse Psicológico/complicações , Estresse Psicológico/psicologia
18.
Med Hypotheses ; 134: 109410, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31627123

RESUMO

Cognitive functions, such as learning and memory, may be impaired during aging. Age-related cognitive impairment is associated with selective neuronal loss, oxidative changes that lead to microglia activation and neuroinflammation. In addition, it is associated to alteration reduction in trophic factors affecting neurogenesis and synaptic plasticity. In recent years, attention has been paid to the relationship between gut microbiota and brain. In aging, there is an alteration in microbiota, gut microbiota diversity is perturbed with an increase in pathogenic bacteria at the expense of beneficial ones. Dysbiosis may lead to chronic inflammation, and a decrease in bacteria metabolites such as short-chain fatty acids which have been related to an upregulation of neurotrophic factors. Supplementation with prebiotics and probiotics can modulate gut microbiota, returning it to a more physiological state; thus, they may be considered as a possible treatment for age-related cognitive impairment.


Assuntos
Disfunção Cognitiva/terapia , Disbiose/terapia , Prebióticos , Probióticos/uso terapêutico , Envelhecimento/psicologia , Fator Neurotrófico Derivado do Encéfalo/fisiologia , Morte Celular , Disfunção Cognitiva/microbiologia , Disfunção Cognitiva/fisiopatologia , Disfunção Cognitiva/prevenção & controle , Disbiose/psicologia , Microbioma Gastrointestinal/fisiologia , Humanos , Inflamação , Microglia/imunologia , Mitocôndrias/fisiologia , Neurogênese , Neurônios/patologia , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo
19.
Cells ; 8(10)2019 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-31546668

RESUMO

Alterations in Triggering Receptors Expressed on Myeloid cells (TREM-1/2) are bound to a variety of infectious, sterile inflammatory, and degenerative conditions, ranging from inflammatory bowel disease (IBD) to neurodegenerative disorders. TREMs are emerging as key players in pivotal mechanisms often concurring in IBD and neurodegeneration, namely microbiota dysbiosis, leaky gut, and inflammation. In conditions of dysbiosis, compounds released by intestinal bacteria activate TREMs on macrophages, leading to an exuberant pro-inflammatory reaction up to damage in the gut barrier. In turn, TREM-positive activated macrophages along with inflammatory mediators may reach the brain through the blood, glymphatic system, circumventricular organs, or the vagus nerve via the microbiota-gut-brain axis. This leads to a systemic inflammatory response which, in turn, impairs the blood-brain barrier, while promoting further TREM-dependent neuroinflammation and, ultimately, neural injury. Nonetheless, controversial results still exist on the role of TREM-2 compared with TREM-1, depending on disease specificity, stage, and degree of inflammation. Therefore, the present review aimed to provide an update on the role of TREMs in the pathophysiology of IBD and neurodegeneration. The evidence here discussed the highlights of the potential role of TREMs, especially TREM-1, in bridging inflammatory processes in intestinal and neurodegenerative disorders.


Assuntos
Doenças Inflamatórias Intestinais/genética , Doenças Neurodegenerativas/genética , Receptor Gatilho 1 Expresso em Células Mieloides/fisiologia , Animais , Barreira Hematoencefálica/fisiologia , Suscetibilidade a Doenças , Disbiose/complicações , Disbiose/genética , Disbiose/imunologia , Disbiose/psicologia , Microbioma Gastrointestinal/fisiologia , Humanos , Doenças Inflamatórias Intestinais/complicações , Doenças Inflamatórias Intestinais/microbiologia , Doenças Inflamatórias Intestinais/psicologia , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/fisiologia , Doenças Neurodegenerativas/imunologia , Doenças Neurodegenerativas/microbiologia , Receptores Imunológicos/genética , Receptores Imunológicos/fisiologia , Fatores de Risco , Receptor Gatilho 1 Expresso em Células Mieloides/genética
20.
Psychiatr Danub ; 31(Suppl 3): 381-385, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31488756

RESUMO

BACKGROUND: Psychiatric disorders may be correlated with a low-grade systemic inflammation but the origin of this inflammatory response remains unclear and both genetics and environmental factors seems to be concerned. Recent researches observed that gut microbiota seems to have an impact on the brain and immune processes. METHOD: We review recent literature to a better understanding of how microbiota interacts with brain, immunity and psychiatric disorders. We search on Pubmed, PsycINFO, PsycARTICLES and Sciencedirect articles with the keywords "gastrointestinal microbiota" and "mental disorders" or "psychological stress". RESULTS: We showed links between gut microbiota and brain-gut axis regulation, immune and endocrine system activity, neurophysiological changes, behavior variations and neuropsychiatric disorders. Communications between brain and gut are bidirectional via neural, endocrine and immune pathway. Microbiota dysbiosis and increase gut permeability with subsequent immune challenges seems to be the source of the chronic mild inflammation associated with neuropsychiatric disorders. Repeated immune or stress events early in life may lead to neurodevelopmental disorders or sickness behavior later in life. CONCLUSIONS: Psychological stress impact gut microbiota with subsequent immune activation leading to neurodevelopmental disorders or sickness behavior and altering neurophysiology and reactivity to stress or lifestyle.


Assuntos
Encéfalo/imunologia , Encéfalo/fisiopatologia , Microbioma Gastrointestinal/imunologia , Microbioma Gastrointestinal/fisiologia , Inflamação/imunologia , Inflamação/psicologia , Transtornos Mentais/imunologia , Transtornos Mentais/microbiologia , Disbiose/imunologia , Disbiose/microbiologia , Disbiose/psicologia , Sistema Endócrino/imunologia , Sistema Endócrino/metabolismo , Sistema Endócrino/microbiologia , Humanos , Inflamação/microbiologia , Neuropsiquiatria , Estresse Psicológico/imunologia , Estresse Psicológico/microbiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...