Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Nature ; 586(7828): 281-286, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32968276

RESUMO

'Dysbiosis' of the maternal gut microbiome, in response to challenges such as infection1, altered diet2 and stress3 during pregnancy, has been increasingly associated with abnormalities in brain function and behaviour of the offspring4. However, it is unclear whether the maternal gut microbiome influences neurodevelopment during critical prenatal periods and in the absence of environmental challenges. Here we investigate how depletion and selective reconstitution of the maternal gut microbiome influences fetal neurodevelopment in mice. Embryos from antibiotic-treated and germ-free dams exhibited reduced brain expression of genes related to axonogenesis, deficient thalamocortical axons and impaired outgrowth of thalamic axons in response to cell-extrinsic factors. Gnotobiotic colonization of microbiome-depleted dams with a limited consortium of bacteria prevented abnormalities in fetal brain gene expression and thalamocortical axonogenesis. Metabolomic profiling revealed that the maternal microbiome regulates numerous small molecules in the maternal serum and the brains of fetal offspring. Select microbiota-dependent metabolites promoted axon outgrowth from fetal thalamic explants. Moreover, maternal supplementation with these metabolites abrogated deficiencies in fetal thalamocortical axons. Manipulation of the maternal microbiome and microbial metabolites during pregnancy yielded adult offspring with altered tactile sensitivity in two aversive somatosensory behavioural tasks, but no overt differences in many other sensorimotor behaviours. Together, our findings show that the maternal gut microbiome promotes fetal thalamocortical axonogenesis, probably through signalling by microbially modulated metabolites to neurons in the developing brain.


Assuntos
Encéfalo/embriologia , Encéfalo/metabolismo , Disbiose/microbiologia , Feto/embriologia , Feto/metabolismo , Microbioma Gastrointestinal/fisiologia , Mães , Animais , Axônios/metabolismo , Encéfalo/citologia , Córtex Cerebral/citologia , Córtex Cerebral/embriologia , Córtex Cerebral/metabolismo , Simulação por Computador , Disbiose/sangue , Disbiose/patologia , Feminino , Feto/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Gravidez , Complicações na Gravidez/sangue , Complicações na Gravidez/microbiologia , Complicações na Gravidez/patologia , Análise de Componente Principal , Tálamo/citologia , Tálamo/embriologia , Tálamo/metabolismo
2.
BMC Gastroenterol ; 24(1): 158, 2024 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-38720308

RESUMO

BACKGROUND: Primary sclerosing cholangitis (PSC) is a complex disease with pathogenic mechanisms that remain to be elucidated. Previous observational studies with small sample sizes have reported associations between PSC, dyslipidemia, and gut microbiota dysbiosis. However, the causality of these associations is uncertain, and there has been no systematic analysis to date. METHODS: The datasets comprise data on PSC, 179 lipid species, and 412 gut microbiota species. PSC data (n = 14,890) were sourced from the International PSC Study Group, while the dataset pertaining to plasma lipidomics originated from a study involving 7174 Finnish individuals. Data on gut microbiota species were derived from the Dutch Microbiome Project study, which conducted a genome-wide association study involving 7738 participants. Furthermore, we employed a two-step Mendelian randomization (MR) analysis to quantify the proportion of the effect of gut microbiota-mediated lipidomics on PSC. RESULTS: Following a rigorous screening process, our MR analysis revealed a causal relationship between higher levels of gene-predicted Phosphatidylcholine (O-16:1_18:1) (PC O-16:1_18:1) and an increased risk of developing PSC (inverse variance-weighted method, odds ratio (OR) 1.30, 95% confidence interval (CI) 1.03-1.63). There is insufficient evidence to suggest that gene-predicted PSC impacts the levels of PC O-16:1_18:1 (OR 1.01, 95% CI 0.98-1.05). When incorporating gut microbiota data into the analysis, we found that Eubacterium rectale-mediated genetic prediction explains 17.59% of the variance in PC O-16:1_18:1 levels. CONCLUSION: Our study revealed a causal association between PC O-16:1_18:1 levels and PSC, with a minor portion of the effect mediated by Eubacterium rectale. This study aims to further explore the pathogenesis of PSC and identify promising therapeutic targets. For patients with PSC who lack effective treatment options, the results are encouraging.


Assuntos
Colangite Esclerosante , Microbioma Gastrointestinal , Lipidômica , Análise da Randomização Mendeliana , Humanos , Colangite Esclerosante/sangue , Colangite Esclerosante/microbiologia , Colangite Esclerosante/genética , Microbioma Gastrointestinal/genética , Masculino , Estudo de Associação Genômica Ampla , Feminino , Fosfatidilcolinas/sangue , Disbiose/sangue , Pessoa de Meia-Idade , Adulto
4.
Hepatology ; 71(2): 522-538, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31228214

RESUMO

Chronic alcohol consumption causes increased intestinal permeability and changes in the intestinal microbiota composition, which contribute to the development and progression of alcohol-related liver disease. In this setting, little is known about commensal fungi in the gut. We studied the intestinal mycobiota in a cohort of patients with alcoholic hepatitis, patients with alcohol use disorder, and nonalcoholic controls using fungal-specific internal transcribed spacer amplicon sequencing of fecal samples. We further measured serum anti-Saccharomyces cerevisiae antibodies (ASCA) as a systemic immune response to fungal products or fungi. Candida was the most abundant genus in the fecal mycobiota of the two alcohol groups, whereas genus Penicillium dominated the mycobiome of nonalcoholic controls. We observed a lower diversity in the alcohol groups compared with controls. Antibiotic or steroid treatment was not associated with a lower diversity. Patients with alcoholic hepatitis had significantly higher ASCA levels compared to patients with alcohol use disorder and to nonalcoholic controls. Within the alcoholic hepatitis cohort, patients with levels of at least 34 IU/mL had a significantly lower 90-day survival (59%) compared with those with ASCA levels less than 34 IU/mL (80%) with an adjusted hazard ratio of 3.13 (95% CI, 1.11-8.82; P = 0.031). Conclusion: Patients with alcohol-associated liver disease have a lower fungal diversity with an overgrowth of Candida compared with controls. Higher serum ASCA was associated with increased mortality in patients with alcoholic hepatitis. Intestinal fungi may serve as a therapeutic target to improve survival, and ASCA may be useful to predict the outcome in patients with alcoholic hepatitis.


Assuntos
Disbiose/etiologia , Disbiose/imunologia , Hepatite Alcoólica/complicações , Hepatite Alcoólica/imunologia , Intestinos/microbiologia , Micobioma , Adulto , Idoso , Anticorpos Antifúngicos/sangue , Candida/imunologia , Estudos de Coortes , Disbiose/sangue , Feminino , Hepatite Alcoólica/sangue , Humanos , Fenômenos do Sistema Imunitário , Masculino , Pessoa de Meia-Idade , Saccharomyces cerevisiae/imunologia
5.
J Psychiatry Neurosci ; 46(3): E358-E368, 2021 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-34008933

RESUMO

Background: The microbiota interacts with the brain through the gut-brain axis, and a distinct dysbiosis may lead to major depressive episodes. Bacteria can pass through the gut barrier and be found in the blood. Using a multiomic approach, we investigated whether a distinct blood microbiome and metabolome was associated with major depressive episodes, and how it was modulated by treatment. Methods: In this case-control multiomic study, we analyzed the blood microbiome composition, inferred bacterial functions and metabolomic profile of 56 patients experiencing a current major depressive episode and 56 matched healthy controls, before and after treatment, using 16S rDNA sequencing and liquid chromatography coupled to tandem mass spectrometry. Results: The baseline blood microbiome in patients with a major depressive episode was distinct from that of healthy controls (patients with a major depressive episode had a higher proportion of Janthinobacterium and lower levels of Neisseria) and changed after antidepressant treatment. Predicted microbiome functions confirmed by metabolomic profiling showed that patients who were experiencing a major depressive episode had alterations in the cyanoamino acid pathway at baseline. High baseline levels of Firmicutes and low proportions of Bosea and Tetrasphaera were associated with response to antidepressant treatment. Based on inferred baseline metagenomic profiles, bacterial pathways that were significantly associated with treatment response were related to xenobiotics, amino acids, and lipid and carbohydrate metabolism, including tryptophan and drug metabolism. Metabolomic analyses showed that plasma tryptophan levels are independently associated with response to antidepressant treatment. Limitations: Our study has some limitations, including a lack of information on blood microbiome origin and the lack of a validation cohort to confirm our results. Conclusion: Patients with depression have a distinct blood microbiome and metabolomic signature that changes after treatment. Dysbiosis could be a new therapeutic target and prognostic tool for the treatment of patients who are experiencing a major depressive episode.


Assuntos
Antidepressivos/uso terapêutico , Sangue/microbiologia , Eixo Encéfalo-Intestino/efeitos dos fármacos , Transtorno Depressivo Maior/tratamento farmacológico , Transtorno Depressivo Maior/microbiologia , Disbiose/microbiologia , Metaboloma/efeitos dos fármacos , Microbiota/efeitos dos fármacos , Adulto , Antidepressivos/farmacologia , Bactérias/classificação , Bactérias/efeitos dos fármacos , Sangue/efeitos dos fármacos , Metabolismo dos Carboidratos/efeitos dos fármacos , Estudos de Casos e Controles , Transtorno Depressivo Maior/sangue , Transtorno Depressivo Maior/complicações , Disbiose/sangue , Disbiose/complicações , Disbiose/metabolismo , Feminino , Microbioma Gastrointestinal/efeitos dos fármacos , Humanos , Metabolismo dos Lipídeos/efeitos dos fármacos , Masculino
6.
Can J Physiol Pharmacol ; 99(1): 9-17, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-32706987

RESUMO

Periodontal disease is one of the most common conditions resulting from poor oral hygiene and is characterized by a destructive process in the periodontium that essentially includes gingiva, alveolar mucosa, cementum, periodontal ligament, and alveolar bone. Notably, the destructive event in the alveolar bone has been linked to homocysteine (Hcy) metabolism; however, it has not been fully investigated. Therefore; the implication of Hcy towards initiation, progression, and maintenance of the periodontal disease remains incompletely understood. Higher levels of Hcy (also known as hyperhomocysteinemia (HHcy)) exerts deleterious effects on gum health and teeth in distinct ways. Firstly, increased production of proinflammatory cytokines such as TNF-α, IL-1ß, IL-6, and IL-8 leads to an inflammatory cascade of events that affect methionine (Met) and Hcy metabolism (i.e., 1-carbon metabolism) leading to HHcy. Secondly, metabolic dysregulation during chronic medical conditions increases systemic inflammation leading to a decrease in vitamins, more specifically B6, B12, and folic acid, that play important roles as cofactors in Hcy metabolism. Also, given the folate level in the HHcy state that is important during dysbiosis, these two conditions appear to be intimately related, and in this context, HHcy-induced dysbiosis may be one of the potential causes of periodontal disease. This paper sums up the link between periodontitis and HHcy, with a special emphasis on the "oral-gut microbiome axis" and the potential probiotic intervention towards warding off some of the serious periodontal disease conditions.


Assuntos
Disbiose/complicações , Microbioma Gastrointestinal/fisiologia , Homocisteína/metabolismo , Hiper-Homocisteinemia/imunologia , Periodontite/imunologia , Disbiose/sangue , Disbiose/imunologia , Disbiose/microbiologia , Ácido Fólico/sangue , Ácido Fólico/metabolismo , Homocisteína/sangue , Homocisteína/imunologia , Humanos , Hiper-Homocisteinemia/sangue , Hiper-Homocisteinemia/metabolismo , Metionina/metabolismo , Periodontite/sangue , Periodontite/metabolismo , Probióticos
7.
Int J Mol Sci ; 22(9)2021 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-34068595

RESUMO

Systemic inflammation, from gut translocation of organismal molecules, might worsen uremic complications in acute kidney injury (AKI). The monitoring of gut permeability integrity and/or organismal molecules in AKI might be clinically beneficial. Due to the less prominence of Candida albicans in human intestine compared with mouse gut, C. albicans were orally administered in bilateral nephrectomy (BiN) mice. Gut dysbiosis, using microbiome analysis, and gut permeability defect (gut leakage), which was determined by fluorescein isothiocyanate-dextran and intestinal tight-junction immunofluorescent staining, in mice with BiN-Candida was more severe than BiN without Candida. Additionally, profound gut leakage in BiN-Candida also resulted in gut translocation of lipopolysaccharide (LPS) and (1→3)-ß-D-glucan (BG), the organismal components from gut contents, that induced more severe systemic inflammation than BiN without Candida. The co-presentation of LPS and BG in mouse serum enhanced inflammatory responses. As such, LPS with Whole Glucan Particle (WGP, a representative BG) induced more severe macrophage responses than LPS alone as determined by supernatant cytokines and gene expression of downstream signals (NFκB, Malt-1 and Syk). Meanwhile, WGP alone did not induced the responses. In parallel, WGP (with or without LPS), but not LPS alone, accelerated macrophage ATP production (extracellular flux analysis) through the upregulation of genes in mitochondria and glycolysis pathway (using RNA sequencing analysis), without the induction of cell activities. These data indicated a WGP pre-conditioning effect on cell energy augmentation. In conclusion, Candida in BiN mice accelerated gut translocation of BG that augmented cell energy status and enhanced pro-inflammatory macrophage responses. Hence, gut fungi and BG were associated with the enhanced systemic inflammation in acute uremia.


Assuntos
Injúria Renal Aguda/metabolismo , Candida albicans/metabolismo , Inflamação/sangue , Proteoglicanas/sangue , Injúria Renal Aguda/genética , Injúria Renal Aguda/microbiologia , Animais , Candida/metabolismo , Candida albicans/patogenicidade , Disbiose/sangue , Metabolismo Energético , Humanos , Inflamação/microbiologia , Inflamação/patologia , Inflamação/cirurgia , Lipopolissacarídeos/sangue , Macrófagos/metabolismo , Macrófagos/microbiologia , Macrófagos/patologia , Camundongos , Microbiota/genética , Nefrectomia/efeitos adversos
8.
Physiol Genomics ; 52(8): 314-321, 2020 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-32628083

RESUMO

Lipocalin 2 (Lcn2) is a multifunctional innate immune protein that limits microbial overgrowth. Our previous study demonstrated that the gut microbiota directly induces intestinal Lcn2 production, and Lcn2-deficient (Lcn2-/-) mice exhibit gut dysbiosis. Coincidentally, gut dysbiosis is associated with metabolic syndrome pathogenesis, and elevated Lcn2 levels has been considered a potential clinical biomarker of metabolic syndrome. Yet whether Lcn2 mitigates or exacerbates metabolic syndrome remains inconclusive. Our objective was to determine whether Lcn2 deficiency-induced compositional changes in gut microbiota contribute to gain in adiposity in aged mice. Utilizing Lcn2-/- mice and their wild-type (WT) littermates, we measured metabolic markers, including fasting blood glucose, serum lipids, fat pad weight, and insulin resistance at ages 3, 6, and 9 mo old. Relative to WT mice, aged Lcn2-/- mice exhibited a gain in adiposity associated with numerous features of metabolic syndrome, including insulin resistance and dyslipidemia. Surprisingly, supplementation with a high-fat diet did not further aggravate metabolic syndrome that spontaneously occurs in Lcn2-/- mice by 6 mo of age. Interestingly, chow-fed Lcn2-/- mice displayed marked differences in the bacterial abundance and metabolomic profile of the gut microbiota compared with WT mice. Overall, our results demonstrate that Lcn2 is essential to maintain metabolic and gut microbiotal homeostasis, where deficiency induces spontaneous delayed onset of metabolic syndrome.


Assuntos
Envelhecimento/metabolismo , Disbiose/complicações , Disbiose/metabolismo , Dislipidemias/complicações , Microbioma Gastrointestinal/genética , Lipocalina-2/deficiência , Síndrome Metabólica/complicações , Síndrome Metabólica/metabolismo , Tecido Adiposo , Adiposidade/genética , Animais , Glicemia/análise , Dieta Hiperlipídica , Modelos Animais de Doenças , Disbiose/sangue , Disbiose/microbiologia , Dislipidemias/sangue , Dislipidemias/microbiologia , Feminino , Microbioma Gastrointestinal/imunologia , Homeostase/genética , Lipocalina-2/genética , Masculino , Síndrome Metabólica/sangue , Síndrome Metabólica/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Obesidade/metabolismo
9.
Respir Res ; 21(1): 271, 2020 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-33076910

RESUMO

BACKGROUND: The role of the microbiota in the pathogenesis of chronic obstructive pulmonary disease (COPD) following exposure to ambient particulate matter (PM) is largely unknown. METHODS: Fifty-four male Sprague-Dawley rats were exposed to clean air, biomass fuel (BMF), or motor vehicle exhaust (MVE) for 4, 12, and 24 weeks. We performed pulmonary inflammation evaluation, morphometric measurements, and lung function analysis in rat lung at three different times points during exposure. Lung and gut microbial composition was assessed by 16S rRNA pyrosequencing. Serum lipopolysaccharide levels were measured and short-chain fatty acids in colon contents were quantified. RESULTS: After a 24-week PM exposure, rats exhibited pulmonary inflammation and pathological changes characteristic of COPD. The control and PM exposure (BMF and MVE) groups showed similar microbial diversity and composition in rat lung. However, the gut microbiota after 24 weeks PM exposure was characterized by decreased microbial richness and diversity, distinct overall microbial composition, lower levels of short-chain fatty acids, and higher serum lipopolysaccharide. CONCLUSION: Chronic exposure to ambient particulate matter induces gut microbial dysbiosis and metabolite shifts in a rat model of chronic obstructive pulmonary disease.


Assuntos
Disbiose/induzido quimicamente , Microbioma Gastrointestinal/efeitos dos fármacos , Exposição por Inalação/efeitos adversos , Pulmão/efeitos dos fármacos , Material Particulado/toxicidade , Doença Pulmonar Obstrutiva Crônica/induzido quimicamente , Animais , Líquido da Lavagem Broncoalveolar , Modelos Animais de Doenças , Disbiose/sangue , Disbiose/fisiopatologia , Microbioma Gastrointestinal/fisiologia , Pulmão/fisiopatologia , Masculino , Material Particulado/administração & dosagem , Doença Pulmonar Obstrutiva Crônica/sangue , Doença Pulmonar Obstrutiva Crônica/fisiopatologia , Ratos , Ratos Sprague-Dawley
10.
Clin Sci (Lond) ; 134(23): 3159-3174, 2020 12 11.
Artigo em Inglês | MEDLINE | ID: mdl-33215637

RESUMO

Gut microbiota dysbiosis has been studied under the pathological conditions of osteoarthritis (OA). However, the effect of antibiotic-induced gut flora dysbiosis on OA remains incompletely understood at present. Herein, we used a mouse (8 weeks) OA model of destabilization of the medial meniscus (DMM) and gut microbiome dysbiosis induced by antibiotic treatment with ampicillin and neomycin for 8 weeks. The results show that antibiotic-induced intestinal microbiota dysbiosis reduced the serum level of lipopolysaccharide (LPS) and the inflammatory response, such as suppression of the levels of tumour necrosis factor-α (TNF-α) and interleukin-6 (IL-6), which can lead to decreased matrix metalloprotease-13 (MMP-13) expression and improvement of OA after joint injury. In addition, trabecular thickness (Tb.Th) and osteophyte scores were increased significantly in antibiotic-induced male mice compared with female mice. We further used network correlation analysis to verify the effect of gut microbiota dysbiosis on OA. Therefore, the present study contributes to our understanding of the gut-joint axis in OA and reveals the relationship between the inflammatory response, sex and gut microbiota, which may provide new strategies to prevent the symptoms and long-term sequelae of OA. Conclusion: Our data showed that gut microbiome dysbiosis alleviates the progression of OA.


Assuntos
Progressão da Doença , Disbiose/microbiologia , Microbioma Gastrointestinal , Osteoartrite/microbiologia , Osteoartrite/patologia , Animais , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Biomarcadores/sangue , Osso e Ossos/efeitos dos fármacos , Osso e Ossos/patologia , Calcificação Fisiológica/efeitos dos fármacos , Cartilagem/efeitos dos fármacos , Cartilagem/patologia , Disbiose/sangue , Disbiose/complicações , Disbiose/tratamento farmacológico , Feminino , Microbioma Gastrointestinal/efeitos dos fármacos , Inflamação/patologia , Masculino , Metaloproteinase 13 da Matriz/metabolismo , Meniscos Tibiais/efeitos dos fármacos , Meniscos Tibiais/patologia , Camundongos Endogâmicos C57BL , Esclerose/complicações , Esclerose/patologia , Caracteres Sexuais
11.
Can J Physiol Pharmacol ; 98(11): 803-809, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32150694

RESUMO

The objective was to explore if and how the microbiota changed in polycystic ovary syndrome (PCOS) women compared with healthy women. Eight obese PCOS (PO group), 10 nonobese PCOS (PN group), and nine healthy normal weight women (control) (C group) were enrolled. Insulin (INS), testosterone (T), follicle-stimulating hormone (FSH), luteinizing hormone (LH), estrogen (E2), and dehydroepiandrosterone (DHEA) were detected with radioimmunoassay. Antimullerian hormone (AMH), fasting glucose, and hemoglobin A1c (HbA1c) were determined by a chemiluminescence immunoassay, glucose oxidase method, and HPLC, respectively. Gut microbiota composition was evaluated by PCR. Alpha diversity was assessed using Chao1 and the Shannon index. PCOS women showed significantly higher T, LH, and LH/FSH and lower FSH levels than the C group (p < 0.05). The AMH level was significantly higher in the PO than in the PN group (p < 0.05). The PO group presented a significantly higher fasting INS level and HMOA-IR scores than the other groups, lower observed SVs and alpha diversity than the C group, higher beta diversity than the PN group (p < 0.05), and decreased abundances of genera (mainly butyrate producers). Regression analysis showed that decreased abundances of several genera were correlated with higher circulating T and impaired glucose metabolism. PCOS is associated with changes in the gut microbiota composition. Obesity has a driving role in the development of dysbiotic gut microbiota in PCOS.


Assuntos
Disbiose/metabolismo , Obesidade/complicações , Síndrome do Ovário Policístico/complicações , Adulto , Glicemia/metabolismo , Índice de Massa Corporal , Estudos de Casos e Controles , Disbiose/sangue , Disbiose/diagnóstico , Disbiose/microbiologia , Jejum/sangue , Feminino , Microbioma Gastrointestinal/fisiologia , Voluntários Saudáveis , Humanos , Insulina/sangue , Insulina/metabolismo , Obesidade/sangue , Obesidade/metabolismo , Obesidade/microbiologia , Síndrome do Ovário Policístico/sangue , Síndrome do Ovário Policístico/metabolismo , Síndrome do Ovário Policístico/microbiologia , Testosterona/sangue , Testosterona/metabolismo , Adulto Jovem
12.
Med Sci Monit ; 26: e926789, 2020 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-33223514

RESUMO

BACKGROUND The mechanism of how intermittent fasting (IF) improves metabolism is not fully understood. Our study aimed to explore the effect of IF on lipid metabolism in obese mice, specifically on the intestinal flora. MATERIAL AND METHODS Diet-induced obese (DIO) mice were subjected to ad libitum (AL) feeding or IF (alternate-day fasting) for 30 days. We examined the lipid metabolism, fat distribution, gene expression of lipid metabolism, and intestinal flora in the mice. RESULTS Despite having access to the same high-fat diet as the AL-fed groups, IF mice displayed pronounced weight loss, and their lipid metabolism significantly improved, mainly reflected in lower serum lipid levels and ameliorated liver steatosis. IF also reduced metabolic endotoxemia in DIO mice. The 16S ribosomal deoxyribonucleic acid gene amplicon sequencing suggested that IF did not change the community richness but had a tendency to increase community diversity in the intestinal flora. In addition, IF significantly reduced the ratio of Firmicutes to Bacteroidetes and increased the relative abundance of Allobaculum in the intestinal flora. CONCLUSIONS IF can improve fat metabolism, reduce fat accumulation, promote white fat conversion to beige, and improve gut microbiota.


Assuntos
Jejum , Microbioma Gastrointestinal , Metabolismo dos Lipídeos , Tecido Adiposo Bege/patologia , Tecido Adiposo Branco/patologia , Animais , Peso Corporal , Dieta Hiperlipídica , Análise Discriminante , Disbiose/sangue , Disbiose/microbiologia , Ingestão de Energia , Comportamento Alimentar , Lipopolissacarídeos/sangue , Masculino , Camundongos Endogâmicos C57BL , Camundongos Obesos
13.
J Allergy Clin Immunol ; 144(1): 157-170.e8, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30768991

RESUMO

BACKGROUND: Diet-induced obesity and food allergies increase in tandem, but a potential cause-and-effect relationship between these diseases of affluence remains to be tested. OBJECTIVE: We sought to test the role of high dietary fat intake, diet-induced obesity, and associated changes in gut microbial community structure on food allergy pathogenesis. METHODS: Mice were fed a high-fat diet (HFD) for 12 weeks before food allergen sensitization on an atopic dermatitis-like skin lesion, followed by intragastric allergen challenge to induce experimental food allergy. Germ-free animals were colonized with a signature HFD or lean microbiota for 8 weeks before induction of food allergy. Food-induced allergic responses were quantified by using a clinical allergy score, serum IgE levels, serum mouse mast cell protease 1 concentrations, and type 2 cytokine responses. Accumulation of intestinal mast cells was examined by using flow cytometry and chloroacetate esterase tissue staining. Changes in the gut microbial community structure were assessed by using high-throughput 16S ribosomal DNA gene sequencing. RESULTS: HFD-induced obesity potentiates food-induced allergic responses associated with dysregulated intestinal effector mast cell responses, increased intestinal permeability, and gut dysbiosis. An HFD-associated microbiome was transmissible to germ-free mice, with the gut microbial community structure of recipients segregating according to the microbiota input source. Independent of an obese state, an HFD-associated gut microbiome was sufficient to confer enhanced susceptibility to food allergy. CONCLUSION: These findings identify HFD-induced microbial alterations as risk factors for experimental food allergy and uncouple a pathogenic role of an HFD-associated microbiome from obesity. Postdieting microbiome alterations caused by overindulgence of dietary fat might increase susceptibility to food allergy.


Assuntos
Dieta Hiperlipídica , Hipersensibilidade Alimentar/microbiologia , Microbioma Gastrointestinal , Animais , DNA Bacteriano/análise , Disbiose/sangue , Disbiose/microbiologia , Feminino , Hipersensibilidade Alimentar/sangue , Imunoglobulina E/sangue , Masculino , Camundongos Endogâmicos C57BL , Obesidade/sangue , Obesidade/microbiologia
14.
Gastroenterology ; 155(2): 383-390.e8, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29729257

RESUMO

BACKGROUND & AIMS: Colorectal cancer (CRC) development has been associated with increased proportions of Bacteroides fragilis and certain Streptococcus, Fusobacterium, and Peptostreptococcus species in the intestinal microbiota. We investigated associations between bacteremia from specific intestinal microbes and occurrence of CRC. METHODS: We performed a retrospective study after collecting data on 13,096 adult patients (exposed group) in Hong Kong hospitalized with bacteremia (identified by blood culture test) without a previous diagnosis of cancer from January 1, 2006 through December 31, 2015. We collected data on intestinal microbes previously associated with CRC (genera Bacteroides, Clostridium, Filifactor, Fusobacterium, Gemella, Granulicatella, Parvimonas, Peptostreptococcus, Prevotella, Solobacterium, and Streptococcus). Clinical information, including patient demographics, comorbid medical conditions, date of bacteremia, and bacterial species identified, were collected. The incidence of biopsy-proved CRC was compared between the exposed and unexposed (patients without bacteremia matched for age, sex, and comorbidities) groups. RESULTS: The risk of CRC was increased in patients with bacteremia from B fragilis (hazard ratio [HR] = 3.85, 95% CI = 2.62-5.64, P = 5.5 × 10-12) or Streptococcus gallolyticus (HR = 5.73, 95% CI = 2.18-15.1, P = 4.1 × 10-4) compared with the unexposed group. In addition, the risk of CRC was increased in patients with bacteremia from Fusobacterium nucleatum (HR = 6.89, 95% CI = 1.70-27.9, P = .007), Peptostreptococcus species (HR = 3.06, 95% CI = 1.47-6.35, P = .003), Clostridium septicum (HR = 17.1, 95% CI = 1.82-160, P = .013), Clostridium perfringens (HR = 2.29, 95% CI = 1.16-4.52, P = .017), or Gemella morbillorum (HR = 15.2, 95% CI = 1.54-150, P = .020). We observed no increased risk in patients with bacteremia caused by microbes not previously associated with colorectal neoplasms. CONCLUSIONS: In a retrospective analysis of patients hospitalized for bacteremia, we associated later diagnosis of CRC with B fragilis and S gallolyticus and other intestinal microbes. These bacteria might have entered the bloodstream from intestinal dysbiosis and perturbed barrier function. These findings support a model in which specific members of the intestinal microbiota promote colorectal carcinogenesis. Clinicians should evaluate patients with bacteremia from these species for neoplastic lesions in the colorectum.


Assuntos
Bacteriemia/microbiologia , Colo/microbiologia , Neoplasias Colorretais/sangue , Disbiose/sangue , Microbioma Gastrointestinal , Adulto , Idoso , Idoso de 80 Anos ou mais , Bacteroides fragilis/isolamento & purificação , Bacteroides fragilis/patogenicidade , Biópsia , Carcinogênese , Colo/patologia , Neoplasias Colorretais/diagnóstico , Neoplasias Colorretais/epidemiologia , Neoplasias Colorretais/microbiologia , Disbiose/diagnóstico , Disbiose/epidemiologia , Disbiose/microbiologia , Feminino , Hong Kong/epidemiologia , Humanos , Incidência , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos , Streptococcus gallolyticus/isolamento & purificação , Streptococcus gallolyticus/patogenicidade
15.
Ann Rheum Dis ; 78(5): 590-593, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30760471

RESUMO

OBJECTIVES: Rheumatoid arthritis (RA) has been associated with a relative expansion of faecal Prevotellaceae. To determine the microbiome composition and prevalence of Prevotella spp. in a group of individuals at increased risk for RA, but prior to the development of the disease. METHODS: In an ongoing cohort study of first-degree relatives (FDRs) of patients with RA, we identified 'FDR controls', asymptomatic and without autoantibodies, and individuals in pre-clinical RA stages, who had either developed anticitrullinated peptide antibodies or rheumatoid factor positivity and/or symptoms and signs associated with possible RA. Stool sampling and culture-independent microbiota analyses were performed followed by descriptive statistics and statistical analyses of community structures. RESULTS: A total of 133 participants were included, of which 50 were categorised as 'FDR controls' and 83 in 'pre-clinical RA stages'. The microbiota of individuals in 'pre-clinical RA stages' was significantly altered compared with FDR controls. We found a significant enrichment of the bacterial family Prevotellaceae, particularly Prevotella spp., in the 'pre-clinical RA' group (p=0.04). CONCLUSIONS: Prevotella spp. enrichment in individuals in pre-clinical stages of RA, before the onset of RA, suggests a role of intestinal dysbiosis in the development of RA.


Assuntos
Artrite Reumatoide/microbiologia , Fezes/microbiologia , Microbioma Gastrointestinal/imunologia , Prevotella/imunologia , Idoso , Artrite Reumatoide/genética , Artrite Reumatoide/imunologia , Autoanticorpos/sangue , Autoanticorpos/imunologia , Estudos de Casos e Controles , Disbiose/sangue , Disbiose/imunologia , Disbiose/microbiologia , Feminino , Predisposição Genética para Doença , Humanos , Masculino , Pessoa de Meia-Idade , Linhagem , Fator Reumatoide/sangue , Fatores de Risco
16.
Metabolomics ; 15(11): 140, 2019 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-31605240

RESUMO

INTRODUCTION: Low gut microbiome richness is associated with dyslipidemia and insulin resistance, and ceramides and other sphingolipids are implicated in the development of diabetes. OBJECTIVES: Determine whether circulating sphingolipids, particularly ceramides, are associated with alterations in the gut microbiome among obese patients with increased diabetes risk. METHODS: This was a cross-sectional and longitudinal retrospective analysis of a dietary/weight loss intervention. Fasted serum was collected from 49 participants (41 women) and analyzed by HPLC-MS/MS to quantify 45 sphingolipids. Shotgun metagenomic sequencing of stool was performed to profile the gut microbiome. RESULTS: Confirming the link to deteriorated glucose homeostasis, serum ceramides were positively correlated with fasting glucose, but inversely correlated with fasting and OGTT-derived measures of insulin sensitivity and ß-cell function. Significant associations with gut dysbiosis were demonstrated, with SM and ceramides being inversely correlated with gene richness. Ceramides with fatty acid chain lengths of 20-24 carbons were the most associated with low richness. Diet-induced weight loss, which improved gene richness, decreased most sphingolipids. Thirty-one MGS, mostly corresponding to unidentified bacteria species, were inversely correlated with ceramides, including a number of Bifidobacterium and Methanobrevibacter smithii. Higher ceramide levels were also associated with increased metagenomic modules for lipopolysaccharide synthesis and flagellan synthesis, two pathogen-associated molecular patterns, and decreased enrichment of genes involved in methanogenesis and bile acid metabolism. CONCLUSION: This study identifies an association between gut microbiota richness, ceramides, and diabetes risk in overweight/obese humans, and suggests that the gut microbiota may contribute to dysregulation of lipid metabolism in metabolic disorders.


Assuntos
Ceramidas/sangue , Disbiose/sangue , Glucose/metabolismo , Metabolômica , Obesidade/sangue , Adulto , Ceramidas/metabolismo , Cromatografia Líquida de Alta Pressão , Estudos Transversais , Disbiose/metabolismo , Feminino , Microbioma Gastrointestinal , Humanos , Masculino , Pessoa de Meia-Idade , Obesidade/metabolismo , Estudos Retrospectivos , Esfingolipídeos/sangue , Esfingolipídeos/metabolismo , Espectrometria de Massas em Tandem
17.
Diabetes Obes Metab ; 21(3): 479-490, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30328245

RESUMO

Obesity is a global pandemic with immense health consequences for individuals and societies. Multiple factors, including environmental influences and genetic predispositions, are known to affect the development of obesity. Despite an increasing understanding of the factors driving the obesity epidemic, therapeutic interventions to prevent or reverse obesity are limited in their impact. Manipulation of the human gut microbiome provides a new potential therapeutic approach in the fight against obesity. Specific gut bacteria and their metabolites are known to affect host metabolism and feeding behaviour, and dysbiosis of this biosystem may lead to metabolic syndrome. Potential therapies to alter the gut microbiota to treat obesity include dietary changes, supplementation of the diet with probiotic organisms and prebiotic compounds that influence bacterial growth, and the use of faecal microbiota transplant, in which gut microbiota from healthy individuals are introduced into the gut. In this review, we examine the growing scientific evidence supporting the mechanisms by which the human gut microbiota may influence carbohydrate metabolism and obesity, and the various possible therapies that may utilize the gut microbiota to help correct metabolic dysfunction.


Assuntos
Transplante de Microbiota Fecal , Microbioma Gastrointestinal/fisiologia , Obesidade/microbiologia , Obesidade/terapia , Glicemia/metabolismo , Disbiose/sangue , Disbiose/complicações , Disbiose/microbiologia , Disbiose/terapia , Comportamento Alimentar/fisiologia , Humanos , Síndrome Metabólica/sangue , Síndrome Metabólica/complicações , Síndrome Metabólica/microbiologia , Síndrome Metabólica/terapia , Obesidade/sangue , Obesidade/complicações , Prebióticos/administração & dosagem , Probióticos/uso terapêutico
18.
J Proteome Res ; 17(3): 1120-1128, 2018 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-29364680

RESUMO

Equine grass sickness (EGS) is a frequently fatal disease of horses, responsible for the death of 1 to 2% of the U.K. horse population annually. The etiology of this disease is currently uncharacterized, although there is evidence it is associated with Clostridium botulinum neurotoxin in the gut. Prevention is currently not possible, and ileal biopsy diagnosis is invasive. The aim of this study was to characterize the fecal microbiota and biofluid metabolic profiles of EGS horses, to further understand the mechanisms underlying this disease, and to identify metabolic biomarkers to aid in diagnosis. Urine, plasma, and feces were collected from horses with EGS, matched controls, and hospital controls. Sequencing the16S rRNA gene of the fecal bacterial population of the study horses found a severe dysbiosis in EGS horses, with an increase in Bacteroidetes and a decrease in Firmicutes bacteria. Metabolic profiling by 1H nuclear magnetic resonance spectroscopy found EGS to be associated with the lower urinary excretion of hippurate and 4-cresyl sulfate and higher excretion of O-acetyl carnitine and trimethylamine-N-oxide. The predictive ability of the complete urinary metabolic signature and using the four discriminatory urinary metabolites to classify horses by disease status was assessed using a second (test) set of horses. The urinary metabolome and a combination of the four candidate biomarkers showed promise in aiding the identification of horses with EGS. Characterization of the metabolic shifts associated with EGS offers the potential of a noninvasive test to aid premortem diagnosis.


Assuntos
Acetilcarnitina/urina , Cresóis/urina , Disbiose/diagnóstico , Hipuratos/urina , Doenças dos Cavalos/diagnóstico , Metilaminas/urina , Ésteres do Ácido Sulfúrico/urina , Acetilcarnitina/sangue , Animais , Bacteroidetes/classificação , Bacteroidetes/isolamento & purificação , Biomarcadores/sangue , Biomarcadores/urina , Clostridium botulinum/metabolismo , Clostridium botulinum/patogenicidade , Cresóis/sangue , Disbiose/sangue , Disbiose/microbiologia , Disbiose/urina , Fezes/microbiologia , Firmicutes/classificação , Firmicutes/isolamento & purificação , Microbioma Gastrointestinal , Hipuratos/sangue , Doenças dos Cavalos/sangue , Doenças dos Cavalos/microbiologia , Doenças dos Cavalos/urina , Cavalos , Espectroscopia de Ressonância Magnética , Metilaminas/sangue , RNA Ribossômico 16S/genética , Ésteres do Ácido Sulfúrico/sangue
19.
Liver Transpl ; 24(6): 752-761, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29500907

RESUMO

Liver transplantation (LT) improves daily function and ameliorates gut microbial composition. However, the effect of LT on microbial functionality, which can be related to overall patient benefit, is unclear and could affect the post-LT course. The aims were to determine the effect of LT on gut microbial functionality focusing on endotoxemia, bile acid (BA), ammonia metabolism, and lipidomics. We enrolled outpatient patients with cirrhosis on the LT list and followed them until 6 months after LT. Microbiota composition (Shannon diversity and individual taxa) and function analysis (serum endotoxin, urinary metabolomics and serum lipidomics, and stool BA profile) and cognitive tests were performed at both visits. We enrolled 40 patients (age, 56 ± 7 years; mean Model for End-Stage Liver Disease score, 22.6). They received LT 6 ± 3 months after enrollment and were re-evaluated 7 ± 3 months after LT with a stable course. A significant improvement in cognition with increase in microbial diversity, increase in autochthonous and decrease in potentially pathogenic taxa, and reduced endotoxemia were seen after LT compared with baseline. Stool BAs increased significantly after LT, and there was evidence of greater bacterial action (higher secondary, oxo and iso-BAs) after LT although the levels of conjugated BAs remained similar. There was a reduced serum ammonia and corresponding rise in urinary phenylacetylglutamine after LT. There was an increase in urinary trimethylamine-N-oxide, which was correlated with specific changes in serum lipids related to cell membrane products. The ultimate post-LT lipidomic profile appeared beneficial compared with the profile before LT. In conclusion, LT improves gut microbiota diversity and dysbiosis, which is accompanied by favorable changes in gut microbial functionality corresponding to BAs, ammonia, endotoxemia, lipidomic, and metabolomic profiles. Liver Transplantation 24 752-761 2018 AASLD.


Assuntos
Disbiose/microbiologia , Doença Hepática Terminal/cirurgia , Microbioma Gastrointestinal/fisiologia , Cirrose Hepática/cirurgia , Transplante de Fígado , Ácidos e Sais Biliares/sangue , Cognição/fisiologia , Disbiose/sangue , Disbiose/fisiopatologia , Doença Hepática Terminal/sangue , Doença Hepática Terminal/microbiologia , Endotoxemia/diagnóstico , Endotoxemia/microbiologia , Endotoxemia/fisiopatologia , Fezes/microbiologia , Feminino , Humanos , Metabolismo dos Lipídeos/fisiologia , Fígado/metabolismo , Fígado/cirurgia , Cirrose Hepática/sangue , Cirrose Hepática/microbiologia , Testes de Função Hepática , Masculino , Metaboloma/fisiologia , Pessoa de Meia-Idade , Índice de Gravidade de Doença , Resultado do Tratamento
20.
Metab Eng ; 49: 128-142, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30075203

RESUMO

Malnutrition is a severe non-communicable disease, which is prevalent in children from low-income countries. Recently, a number of metagenomics studies have illustrated associations between the altered gut microbiota and child malnutrition. However, these studies did not examine metabolic functions and interactions between individual species in the gut microbiota during health and malnutrition. Here, we applied genome-scale metabolic modeling to model the gut microbial species, which were selected from healthy and malnourished children from three countries. Our analysis showed reduced metabolite production capabilities in children from two low-income countries compared with a high-income country. Additionally, the models were also used to predict the community-level metabolic potentials of gut microbes and the patterns of pairwise interactions among species. Hereby we found that due to bacterial interactions there may be reduced production of certain amino acids in malnourished children compared with healthy children from the same communities. To gain insight into alterations in the metabolism of malnourished (stunted) children, we also performed targeted plasma metabolic profiling in the first 2 years of life of 25 healthy and 25 stunted children. Plasma metabolic profiling further revealed that stunted children had reduced plasma levels of essential amino acids compared to healthy controls. Our analyses provide a framework for future efforts towards further characterization of gut microbial metabolic capabilities and their contribution to malnutrition.


Assuntos
Aminoácidos/sangue , Transtornos da Nutrição Infantil , Disbiose , Microbioma Gastrointestinal , Genoma Bacteriano , Criança , Transtornos da Nutrição Infantil/sangue , Transtornos da Nutrição Infantil/genética , Transtornos da Nutrição Infantil/microbiologia , Pré-Escolar , Disbiose/sangue , Disbiose/genética , Disbiose/microbiologia , Feminino , Humanos , Masculino
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA