Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 6.558
Filtrar
1.
Gut Microbes ; 16(1): 2398126, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39254265

RESUMO

The intestine exhibits distinct characteristics along its length, with a substantial immune cell reservoir and diverse microbiota crucial for maintaining health. This study investigates how anatomical location and regional microbiota influence intestinal immune cell abundance. Using conventionally colonized and germ-free mice, segment-specific immune cell composition and microbial communities were assessed. Metagenomic sequencing analyzed microbiome variations, while flow cytometry and immunofluorescence examined immune cell composition. Microbiome composition varied significantly along the intestine, with diversity and abundance increasing from upper to lower segments. Immune cells showed distinct segment-specific patterning influenced by microbial colonization and localization. T cell subsets displayed varied dependence on microbiome presence and anatomical location. This study highlights locoregional differences in intestinal immune cell and microbiome composition, identifying immune subsets susceptible to microbiota presence. The findings provide context for understanding immune cell alterations in disease models.


Assuntos
Bactérias , Microbioma Gastrointestinal , Camundongos Endogâmicos C57BL , Animais , Camundongos , Bactérias/classificação , Bactérias/genética , Bactérias/isolamento & purificação , Bactérias/imunologia , Intestinos/microbiologia , Intestinos/imunologia , Intestinos/citologia , Metagenômica , Vida Livre de Germes , Feminino , Subpopulações de Linfócitos T/imunologia , Masculino , Mucosa Intestinal/microbiologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/citologia
2.
Physiol Plant ; 176(5): e14502, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39238133

RESUMO

Existing research has underscored the vital interplay between host organisms and their associated microbiomes, which affects health and function. In both plants and animals, host factors critically shape microbial communities and influence growth, health, and immunity. Post-harvest plants, such as those used in kimchi, a traditional Korean dish, offer a unique avenue for exploring host-microbe dynamics during fermentation. Despite the emphasis on lactic acid bacteria (LAB) in fermentation studies, the roles of host factors remain unclear. This study aimed to investigate the influence of these factors on plant transcriptomes during kimchi fermentation. We individually inoculated nine LAB strains into germ-free kimchi to generate LAB-mono-associated gnotobiotic kimchi and performed RNA-sequencing analysis for the host vegetables during fermentation. The transcriptomes of post-harvest vegetables in kimchi change over time, and microbes affect the transcriptome profiles of vegetables. Differentially expressed gene analyses revealed that microbes affected the temporal expression profiles of several genes in the plant transcriptomes in unique directions depending on the introduced LAB strains. Cluster analysis with other publicly available transcriptomes of post-harvest vegetables and fruits further revealed that the plant transcriptome is more profoundly influenced by the environment harboring the host than by host phylogeny. Our results bridge the gap in understanding the bidirectional relationship between host vegetables and microbes during food fermentation, illuminating the complex interplay between vegetable transcriptomes, fermentative microbes, and the fermentation process in food production. The different transcriptomic responses elicited by specific LAB strains suggest the possibility of microbial manipulation to achieve the desired fermentation outcomes.


Assuntos
Fermentação , Vida Livre de Germes , Verduras , Verduras/genética , Verduras/microbiologia , Transcriptoma/genética , Alimentos Fermentados/microbiologia , Regulação da Expressão Gênica de Plantas , Lactobacillales/genética , Lactobacillales/fisiologia , Lactobacillales/metabolismo
3.
BMC Microbiol ; 24(1): 364, 2024 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-39333864

RESUMO

BACKGROUND: Dysbiosis of the microbiome is a key hallmark of polycystic ovary syndrome (PCOS). However, the interaction between the host and microbiome and its relevance to the pathogenesis of PCOS remain unclear. METHODS: To evaluate the role of the commensal gut microbiome in PCOS, we gavaged germ-free mice with the fecal microbiota from patients with PCOS or healthy individuals and evaluated the reproductive endocrine features of the recipient mice. RESULTS: Mice transplanted with fecal microbiota from PCOS patients and those transplanted from healthy controls presented different bacterial profiles and reproductive endocrine features. The fecal microbiota of the mice in the PCOS group was enriched in Phocaeicola, Mediterraneibacter, Oscillospiraceae, Lawsonibacter and Rikenellaceae. Fecal microbiota transplantation (FMT) from PCOS patients induced increased disruption of ovarian functions, lipo-metabolic disturbance, insulin resistance and an obese-like phenotype in recipient mice. CONCLUSION: Our findings suggest that the microbiome may govern the set point of PCOS-bearing individuals and that gut ecosystem manipulation may be a useful marker and target for the management of PCOS.


Assuntos
Disbiose , Transplante de Microbiota Fecal , Microbioma Gastrointestinal , Síndrome do Ovário Policístico , Síndrome do Ovário Policístico/microbiologia , Síndrome do Ovário Policístico/terapia , Animais , Feminino , Camundongos , Humanos , Disbiose/microbiologia , Fezes/microbiologia , Doenças Metabólicas/microbiologia , Doenças Metabólicas/etiologia , Doenças Metabólicas/terapia , Bactérias/classificação , Bactérias/isolamento & purificação , Bactérias/genética , Resistência à Insulina , Ovário/microbiologia , Vida Livre de Germes , Modelos Animais de Doenças , Adulto
4.
Microbiome ; 12(1): 179, 2024 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-39307855

RESUMO

BACKGROUND: Dietary fibers can alter microbial metabolic output in support of healthy immune function; however, the impact of distinct fiber sources and immunomodulatory effects beyond short-chain fatty acid production are underexplored. In an effort to discern the effects of diverse fibers on host immunity, we employed five distinct rodent diets with varying fiber content and source in specific-pathogen-free, gnotobiotic (containing a 14-member synthetic human gut microbiota), and germ-free mice. RESULTS: Broad-scale metabolomics analysis of cecal contents revealed that fiber deprivation consistently reduced the concentrations of microbiota-produced B vitamins. This phenomenon was not always explained by reduced biosynthesis, rather, metatranscriptomic analyses pointed toward increased microbial usage of certain B vitamins under fiber-free conditions, ultimately resulting in a net reduction of host-available B vitamins. Broad immunophenotyping indicated that the local gut effector immune populations and activated T cells accumulate in a microbiota-dependent manner. Supplementation with the prebiotic inulin recovered the availability of microbially produced B vitamins and restored immune homeostasis. CONCLUSIONS: Our findings highlight the potential to use defined fiber polysaccharides to boost microbiota-derived B vitamin availability in an animal model and to regulate local innate and adaptive immune populations of the host. Video abstract.


Assuntos
Fibras na Dieta , Microbioma Gastrointestinal , Complexo Vitamínico B , Fibras na Dieta/metabolismo , Animais , Camundongos , Complexo Vitamínico B/metabolismo , Camundongos Endogâmicos C57BL , Vida Livre de Germes , Masculino , Ceco/microbiologia , Humanos , Bactérias/classificação , Bactérias/metabolismo , Metabolômica , Prebióticos
5.
Viruses ; 16(9)2024 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-39339908

RESUMO

For nearly twenty years, gnotobiotic (Gn) pigs have been used as a model of human norovirus (HuNoV) infection and disease. Unique in their ability to develop diarrhea and shed virus post oral challenge, Gn pigs have since been used to evaluate the infectivity of several genogroup II HuNoV strains. Nearly all major pandemic GII.4 variants have been tested in Gn pigs, with varying rates of infectivity. Some induce an asymptomatic state despite being shed in large quantities in stool, and others induce high incidence of both diarrhea and virus shedding. Non-GII.4 strains, including GII.12 and GII.6, have also been evaluated in Gn pigs. Again, rates of diarrhea and virus shedding tend to vary between studies. Several factors may influence these findings, including age, dosage, biological host factors, or bacterial presence. The impact of these factors is nuanced and requires further evaluation to elucidate the exact mechanisms behind increases or decreases in infection rates. Regardless, the value of Gn pig models in HuNoV research cannot be understated, and the model will surely continue to contribute to the field in years to come.


Assuntos
Infecções por Caliciviridae , Diarreia , Modelos Animais de Doenças , Vida Livre de Germes , Norovirus , Eliminação de Partículas Virais , Animais , Norovirus/genética , Norovirus/patogenicidade , Norovirus/fisiologia , Norovirus/classificação , Suínos , Infecções por Caliciviridae/virologia , Infecções por Caliciviridae/veterinária , Diarreia/virologia , Diarreia/veterinária , Humanos , Fezes/virologia , Genótipo
6.
Gut Microbes ; 16(1): 2392872, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39189608

RESUMO

We sought to better understand how intestinal microbiota confer protection against Clostridioides difficile (C. difficile) infection (CDI). We utilized gnotobiotic altered Schaedler flora (ASF) mice, which lack the abnormalities of germfree (GF) mice as well as the complexity and heterogeneity of antibiotic-treated mice. Like GF mice, ASF mice were highly prone to rapid lethal CDI, without antibiotics, while very low infectious doses resulted in chronic CDI. Administering such chronic CDI mice an undefined preparation of Clostridia lowered C. difficile levels by several logs. Importantly, such resolution of CDI was associated with colonization of Lachnospiraceae. Fractionation of the Clostridia population to enrich for Lachnospiraceae led to the appreciation that its CDI-impeding property strongly associated with a specific Lachnospiraceae strain, namely uncultured bacteria and archaea (UBA) 3401. UBA3401 was recalcitrant to being propagated as a pure culture but could be maintained in ASF mice, wherein it comprised up to about 50% of the intestinal microbiota, which was sufficient to generate a high-quality genomic sequence of this bacterium. Sequence analysis and ex vivo study of UBA3401 indicated that it had the ability to secrete substance(s) that directly impeded C. difficile growth. Moreover, in vivo administration of UBA3401/ASF feces provided strong protection to C. difficile challenge. Thus, UBA3401 may contribute to and/or provide a means to study microbiota-mediated CDI resistance.


Assuntos
Clostridiales , Clostridioides difficile , Infecções por Clostridium , Microbioma Gastrointestinal , Vida Livre de Germes , Animais , Camundongos , Clostridioides difficile/genética , Clostridioides difficile/crescimento & desenvolvimento , Clostridioides difficile/fisiologia , Clostridioides difficile/patogenicidade , Infecções por Clostridium/microbiologia , Infecções por Clostridium/prevenção & controle , Microbioma Gastrointestinal/efeitos dos fármacos , Clostridiales/genética , Clostridiales/crescimento & desenvolvimento , Camundongos Endogâmicos C57BL , Modelos Animais de Doenças , Fezes/microbiologia , Feminino , Antibacterianos/farmacologia
7.
Gut Microbes ; 16(1): 2387875, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39133869

RESUMO

The intestinal microbiome during infancy and childhood has distinct metabolic functions and microbial composition compared to adults. We recently published a gnotobiotic mouse model of the pre-weaning microbiome (PedsCom), which retains a pre-weaning configuration during the transition from a milk-based diet to solid foods, leads to a stunted immune system, and increases susceptibility to enteric infection. Here, we compared the phylogenetic and metabolic relationships of the PedsCom consortium to two adult-derived gnotobiotic communities, Altered Schaedler Flora and Oligo-Mouse Microbiota 12 (Oligo-MM12). We find that PedsCom contains several unique functions relative to these adult-derived mouse consortia, including differences in carbohydrate and lipid metabolism genes. Notably, amino acid degradation metabolic modules are more prevalent among PedsCom isolates, which is in line with the ready availability of these nutrients in milk. Indeed, metabolomic analysis revealed significantly lower levels of total free amino acids and lower levels of specific amino acids abundant in milk (e.g. glutamine and glutamic acid) in the intestinal contents of adult PedsCom colonized mice compared to Oligo-MM12 controls. Metabolomic analysis of pre-weaning intestinal contents also showed lower levels of amino acids that are replete in milk compared to germ-free controls. Thus, enhanced amino acid metabolism is a prominent feature of the pre-weaning microbiome that may facilitate design of early-life microbiome interventions.


Assuntos
Aminoácidos , Bactérias , Microbioma Gastrointestinal , Vida Livre de Germes , Leite , Desmame , Animais , Aminoácidos/metabolismo , Microbioma Gastrointestinal/fisiologia , Camundongos , Leite/microbiologia , Leite/metabolismo , Bactérias/classificação , Bactérias/metabolismo , Bactérias/genética , Bactérias/isolamento & purificação , Filogenia , Feminino , Camundongos Endogâmicos C57BL
8.
Gut Microbes ; 16(1): 2392874, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39163515

RESUMO

Alterations in intestinal permeability and the gut microbiome caused by alcohol abuse are associated with alcoholic liver disease and with worsening of inflammatory bowel diseases (IBD) symptoms. To resolve the direct effects of chronic ethanol consumption on the colon and its microbiome in the absence of acute or chronic alcohol-induced liver disease, we developed a mouse model of chronic binge drinking that uncovers how alcohol may enhance susceptibility to colitis via the microbiota. Employing daily ethanol gavage, we recapitulate key features of binge ethanol consumption. We found that binge ethanol drinking worsens intestinal infection, colonic injury and inflammation, and this effect persists beyond the drinking period. Using gnotobiotics, we showed that alcohol-driven susceptibility to colitis is microbiota-dependent and transferable to ethanol-naïve mice by microbiome transplantation. Allobaculum spp. expanded in binge drinking mice, and administration of Allobaculum fili was sufficient to enhance colitis in non-drinking mice. Our study provides a model to study binge drinking-microbiota interactions and their effects on host disease and reinforces the pathogenic function of Allobaculum spp. as colitogenic bacteria. Our findings illustrate how chronic binge drinking-induced alterations of the microbiome may affect susceptibility to IBD onset or flares.


Assuntos
Consumo Excessivo de Bebidas Alcoólicas , Colite , Colo , Microbioma Gastrointestinal , Camundongos Endogâmicos C57BL , Animais , Consumo Excessivo de Bebidas Alcoólicas/complicações , Microbioma Gastrointestinal/efeitos dos fármacos , Camundongos , Colite/microbiologia , Colite/induzido quimicamente , Colo/microbiologia , Colo/patologia , Modelos Animais de Doenças , Bactérias/classificação , Bactérias/isolamento & purificação , Bactérias/genética , Etanol/efeitos adversos , Suscetibilidade a Doenças , Masculino , Vida Livre de Germes , Inflamação/microbiologia , Doenças Inflamatórias Intestinais/microbiologia , Doenças Inflamatórias Intestinais/patologia
9.
Mol Metab ; 88: 102004, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39127167

RESUMO

BACKGROUND: Recent advances have significantly expanded our understanding of the gut microbiome's influence on host physiology and metabolism. However, the specific role of certain microorganisms in gestational health and fetal development remains underexplored. OBJECTIVE: This study investigates the impact of Bifidobacterium breve UCC2003 on fetal brain metabolism when colonized in the maternal gut during pregnancy. METHODS: Germ-free pregnant mice were colonized with or without B. breve UCC2003 during pregnancy. The metabolic profiles of fetal brains were analyzed, focusing on the presence of key metabolites and the expression of critical metabolic and cellular pathways. RESULTS: Maternal colonization with B. breve resulted in significant metabolic changes in the fetal brain. Specifically, ten metabolites, including citrate, 3-hydroxyisobutyrate, and carnitine, were reduced in the fetal brain. These alterations were accompanied by increased abundance of transporters involved in glucose and branched-chain amino acid uptake. Furthermore, supplementation with this bacterium was associated with elevated expression of critical metabolic pathways such as PI3K-AKT, AMPK, STAT5, and Wnt-ß-catenin signaling, including its receptor Frizzled-7. Additionally, there was stabilization of HIF-2 protein and modifications in genes and proteins related to cellular growth, axogenesis, and mitochondrial function. CONCLUSIONS: The presence of maternal B. breve during pregnancy plays a crucial role in modulating fetal brain metabolism and growth. These findings suggest that Bifidobacterium could modify fetal brain development, potentially offering new avenues for enhancing gestational health and fetal development through microbiota-targeted interventions.


Assuntos
Bifidobacterium breve , Encéfalo , Microbioma Gastrointestinal , Animais , Feminino , Camundongos , Bifidobacterium breve/metabolismo , Encéfalo/metabolismo , Gravidez , Microbioma Gastrointestinal/fisiologia , Feto/metabolismo , Vida Livre de Germes , Desenvolvimento Fetal , Camundongos Endogâmicos C57BL
10.
Brain Behav Immun ; 122: 301-312, 2024 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-39128572

RESUMO

Recent research has unveiled conflicting evidence regarding the link between aggression and the gut microbiome. Here, we compared behavior profiles of control, germ-free (GF), and antibiotic-treated mice, as well as re-colonized GF mice to understand the impact of the gut microbiome on aggression using the resident-intruder paradigm. Our findings revealed a link between gut microbiome depletion and higher aggression, accompanied by notable changes in urine metabolite profiles and brain gene expression. This study extends beyond classical murine models to humanized mice to reveal the clinical relevance of early-life antibiotic use on aggression. Fecal microbiome transplant from infants exposed to antibiotics in early life (and sampled one month later) into mice led to increased aggression compared to mice receiving transplants from unexposed infants. This study sheds light on the role of the gut microbiome in modulating aggression and highlights its potential avenues of action, offering insights for development of therapeutic strategies for aggression-related disorders.


Assuntos
Agressão , Encéfalo , Transplante de Microbiota Fecal , Microbioma Gastrointestinal , Agressão/fisiologia , Animais , Microbioma Gastrointestinal/fisiologia , Camundongos , Transplante de Microbiota Fecal/métodos , Masculino , Encéfalo/metabolismo , Antibacterianos/farmacologia , Comportamento Animal/fisiologia , Vida Livre de Germes , Camundongos Endogâmicos C57BL , Humanos
11.
Curr Microbiol ; 81(9): 267, 2024 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-39003673

RESUMO

In this study, we evaluated the impact of human gut microbiota on the immune pathways in the respiratory tract using a gnotobiotic (Gn) piglet model. We humanized piglets with rural and urban infant fecal microbiota (RIFM and UIFM, respectively) and then infected them with a H1N1 swine influenza virus. We analyzed the microbial diversity and structure of the intestinal and respiratory tracts of the piglets before and after the influenza virus infection and measured the viral load and immune responses. We found that the viral load in the upper respiratory tract of UIFM transplanted piglets was higher than their rural cohorts (RIFM), while virus-specific antibody responses were comparable. The relative cytokine gene expression in the tracheobronchial (respiratory tract) and mesenteric (gastrointestinal) lymph nodes, lungs, blood, and spleen of RIFM and UIFM piglets revealed a trend in reciprocal regulation of proinflammatory, innate, and adaptive immune-associated cytokines as well as the frequency of T-helper/memory cells, cytotoxic T cells, and myeloid immune cell subsets. We also observed different phylum-level shifts of the fecal microbiota in response to influenza virus infection between the two piglet groups, suggesting the potential impact of the gut microbiota on the immune responses to influenza virus infection and lung microbiota. In conclusion, Gn piglets humanized with diverse infant fecal microbiota had differential immune regulation, with UIFM favoring the activation of proinflammatory immune mediators following an influenza virus infection compared to their rural RIFM cohorts. Furthermore, Gn piglets can be a useful model in investigating the impact of diverse human microbiota of the gastrointestinal tract, probably also the respiratory tract, on respiratory health and testing specific probiotic- or prebiotic-based therapeutics.


Assuntos
Citocinas , Modelos Animais de Doenças , Fezes , Microbioma Gastrointestinal , Vida Livre de Germes , Imunidade nas Mucosas , Vírus da Influenza A Subtipo H1N1 , Animais , Suínos , Fezes/microbiologia , Fezes/virologia , Humanos , Vírus da Influenza A Subtipo H1N1/imunologia , Citocinas/metabolismo , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/virologia , Carga Viral , Lactente , Influenza Humana/imunologia , Influenza Humana/microbiologia , Influenza Humana/virologia
12.
Nature ; 632(8023): 174-181, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38987594

RESUMO

Changes in the gut microbiome have pivotal roles in the pathogenesis of acute graft-versus-host disease (aGVHD) after allogenic haematopoietic cell transplantation (allo-HCT)1-6. However, effective methods for safely resolving gut dysbiosis have not yet been established. An expansion of the pathogen Enterococcus faecalis in the intestine, associated with dysbiosis, has been shown to be a risk factor for aGVHD7-10. Here we analyse the intestinal microbiome of patients with allo-HCT, and find that E. faecalis escapes elimination and proliferates in the intestine by forming biofilms, rather than by acquiring drug-resistance genes. We isolated cytolysin-positive highly pathogenic E. faecalis from faecal samples and identified an anti-E. faecalis enzyme derived from E. faecalis-specific bacteriophages by analysing bacterial whole-genome sequencing data. The antibacterial enzyme had lytic activity against the biofilm of E. faecalis in vitro and in vivo. Furthermore, in aGVHD-induced gnotobiotic mice that were colonized with E. faecalis or with patient faecal samples characterized by the domination of Enterococcus, levels of intestinal cytolysin-positive E. faecalis were decreased and survival was significantly increased in the group that was treated with the E. faecalis-specific enzyme, compared with controls. Thus, administration of a phage-derived antibacterial enzyme that is specific to biofilm-forming pathogenic E. faecalis-which is difficult to eliminate with existing antibiotics-might provide an approach to protect against aGVHD.


Assuntos
Bacteriófagos , Enterococcus faecalis , Microbioma Gastrointestinal , Doença Enxerto-Hospedeiro , Adulto , Idoso , Animais , Feminino , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Adulto Jovem , Bacteriófagos/enzimologia , Bacteriófagos/genética , Biofilmes/efeitos dos fármacos , Biofilmes/crescimento & desenvolvimento , Disbiose/complicações , Disbiose/microbiologia , Enterococcus faecalis/efeitos dos fármacos , Enterococcus faecalis/genética , Enterococcus faecalis/crescimento & desenvolvimento , Enterococcus faecalis/metabolismo , Enterococcus faecalis/virologia , Fezes/microbiologia , Vida Livre de Germes , Doença Enxerto-Hospedeiro/complicações , Doença Enxerto-Hospedeiro/microbiologia , Doença Enxerto-Hospedeiro/prevenção & controle , Doença Enxerto-Hospedeiro/terapia , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Técnicas In Vitro , Intestinos/efeitos dos fármacos , Intestinos/microbiologia , Perforina/metabolismo , Fatores de Risco , Transplante Homólogo/efeitos adversos , Sequenciamento Completo do Genoma , Farmacorresistência Bacteriana/efeitos dos fármacos , Antibacterianos/farmacologia
13.
J Med Microbiol ; 73(7)2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-39028257

RESUMO

Clostridioides difficile is the most common cause of nosocomial antibiotic-associated diarrhoea and is responsible for a spectrum of diseases characterized by high levels of recurrence and morbidity. In some cases, complications can lead to death. Currently, several types of animal models have been developed to study various aspects of C. difficile infection (CDI), such as colonization, virulence, transmission and recurrence. These models have also been used to test the role of environmental conditions, such as diet, age and microbiome that modulate infection outcome, and to evaluate several therapeutic strategies. Different rodent models have been used successfully, such as the hamster model and the gnotobiotic and conventional mouse models. These models can be applied to study either the initial CDI infectious process or recurrences. The applications of existing rodent models and their advantages and disadvantages are discussed here.


Assuntos
Clostridioides difficile , Infecções por Clostridium , Modelos Animais de Doenças , Animais , Infecções por Clostridium/microbiologia , Clostridioides difficile/patogenicidade , Camundongos , Cricetinae , Humanos , Roedores/microbiologia , Vida Livre de Germes
14.
Gut Microbes ; 16(1): 2382324, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39069899

RESUMO

The human gut microbiota is a complex community comprising hundreds of species, with a few present in high abundance and the vast majority in low abundance. The biological functions and effects of these low-abundant species on their hosts are not yet fully understood. In this study, we assembled a bacterial consortium (SC-4) consisting of B. paravirosa, C. comes, M. indica, and A. butyriciproducens, which are low-abundant, short-chain fatty acid (SCFA)-producing bacteria isolated from healthy human gut, and tested its effect on host health using germ-free and human microbiota-associated colitis mouse models. The selection also favored these four bacteria being reduced in abundance in either Ulcerative Colitis (UC) or Crohn's disease (CD) metagenome samples. Our findings demonstrate that SC-4 can colonize germ-free (GF) mice, increasing mucin thickness by activating MUC-1 and MUC-2 genes, thereby protecting GF mice from Dextran Sodium Sulfate (DSS)-induced colitis. Moreover, SC-4 aided in the recovery of human microbiota-associated mice from DSS-induced colitis, and intriguingly, its administration enhanced the alpha diversity of the gut microbiome, shifting the community composition closer to control levels. The results showed enhanced phenotypes across all measures when the mice were supplemented with inulin as a dietary fiber source alongside SC-4 administration. We also showed a functional redundancy existing in the gut microbiome, resulting in the low abundant SCFA producers acting as a form of insurance, which in turn accelerates recovery from the dysbiotic state upon the administration of SC-4. SC-4 colonization also upregulated iNOS gene expression, further supporting its ability to produce an increasing number of goblet cells. Collectively, our results provide evidence that low-abundant SCFA-producing species in the gut may offer a novel therapeutic approach to IBD.


Assuntos
Bactérias , Colite , Sulfato de Dextrana , Disbiose , Ácidos Graxos Voláteis , Microbioma Gastrointestinal , Animais , Ácidos Graxos Voláteis/metabolismo , Humanos , Disbiose/microbiologia , Camundongos , Bactérias/classificação , Bactérias/isolamento & purificação , Bactérias/genética , Bactérias/metabolismo , Colite/microbiologia , Colite/induzido quimicamente , Modelos Animais de Doenças , Camundongos Endogâmicos C57BL , Consórcios Microbianos , Masculino , Feminino , Colite Ulcerativa/microbiologia , Colite Ulcerativa/metabolismo , Vida Livre de Germes
15.
Nature ; 632(8027): 1137-1144, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39085612

RESUMO

Exposure to environmental pollutants and human microbiome composition are important predisposition factors for tumour development1,2. Similar to drug molecules, pollutants are typically metabolized in the body, which can change their carcinogenic potential and affect tissue distribution through altered toxicokinetics3. Although recent studies demonstrated that human-associated microorganisms can chemically convert a wide range of xenobiotics and influence the profile and tissue exposure of resulting metabolites4,5, the effect of microbial biotransformation on chemical-induced tumour development remains unclear. Here we show that the depletion of the gut microbiota affects the toxicokinetics of nitrosamines, which markedly reduces the development and severity of nitrosamine-induced urinary bladder cancer in mice6,7. We causally linked this carcinogen biotransformation to specific gut bacterial isolates in vitro and in vivo using individualized bacterial culture collections and gnotobiotic mouse models, respectively. We tested gut communities from different human donors to demonstrate that microbial carcinogen metabolism varies between individuals and we showed that this metabolic activity applies to structurally related nitrosamine carcinogens. Altogether, these results indicate that gut microbiota carcinogen metabolism may be a contributing factor for chemical-induced carcinogenesis, which could open avenues to target the microbiome for improved predisposition risk assessment and prevention of cancer.


Assuntos
Carcinogênese , Carcinógenos , Microbioma Gastrointestinal , Nitrosaminas , Neoplasias da Bexiga Urinária , Animais , Feminino , Humanos , Masculino , Camundongos , Biotransformação , Carcinogênese/induzido quimicamente , Carcinogênese/metabolismo , Carcinogênese/patologia , Carcinógenos/química , Carcinógenos/metabolismo , Carcinógenos/farmacocinética , Carcinógenos/toxicidade , Microbioma Gastrointestinal/fisiologia , Vida Livre de Germes , Camundongos Endogâmicos C57BL , Nitrosaminas/química , Nitrosaminas/metabolismo , Nitrosaminas/farmacocinética , Nitrosaminas/toxicidade , Neoplasias da Bexiga Urinária/induzido quimicamente , Neoplasias da Bexiga Urinária/etiologia , Neoplasias da Bexiga Urinária/metabolismo , Neoplasias da Bexiga Urinária/patologia , Neoplasias da Bexiga Urinária/prevenção & controle , Suscetibilidade a Doenças
16.
Cell Host Microbe ; 32(6): 820-836, 2024 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-38870899

RESUMO

Microbial communities that colonize the human gastrointestinal (GI) tract defend against pathogens through a mechanism known as colonization resistance (CR). Advances in technologies such as next-generation sequencing, gnotobiotic mouse models, and bacterial cultivation have enhanced our understanding of the underlying mechanisms and the intricate microbial interactions involved in CR. Rather than being attributed to specific microbial clades, CR is now understood to arise from a dynamic interplay between microbes and the host and is shaped by metabolic, immune, and environmental factors. This evolving perspective underscores the significance of contextual factors, encompassing microbiome composition and host conditions, in determining CR. This review highlights recent research that has shifted its focus toward elucidating how these factors interact to either promote or impede enteric infections. It further discusses future research directions to unravel the complex relationship between host, microbiota, and environmental determinants in safeguarding against GI infections to promote human health.


Assuntos
Microbioma Gastrointestinal , Humanos , Animais , Camundongos , Interações entre Hospedeiro e Microrganismos , Trato Gastrointestinal/microbiologia , Bactérias/genética , Bactérias/classificação , Interações Hospedeiro-Patógeno , Vida Livre de Germes , Interações Microbianas
17.
Int J Mol Sci ; 25(11)2024 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-38892368

RESUMO

Intestinal epithelium renewal strictly depends on fine regulation between cell proliferation, differentiation, and apoptosis. While murine intestinal microbiota has been shown to modify some epithelial cell kinetics parameters, less is known about the role of the human intestinal microbiota. Here, we investigated the rate of intestinal cell proliferation in C3H/HeN germ-free mice associated with human flora (HFA, n = 8), and in germ-free (n = 15) and holoxenic mice (n = 16). One hour before sacrifice, all mice were intraperitoneally inoculated with 5-bromodeoxyuridine (BrdU), and the number of BrdU-positive cells/total cells (labelling index, LI), both in the jejunum and the colon, was evaluated by immunohistochemistry. Samples were also observed by scanning electron microscopy (SEM). Moreover, the microbiota composition in the large bowel of the HFA mice was compared to that of of human donor's fecal sample. No differences in LI were found in the small bowels of the HFA, holoxenic, and germ-free mice. Conversely, the LI in the large bowel of the HFA mice was significantly higher than that in the germ-free and holoxenic counterparts (p = 0.017 and p = 0.048, respectively). In the holoxenic and HFA mice, the SEM analysis disclosed different types of bacteria in close contact with the intestinal epithelium. Finally, the colonic microbiota composition of the HFA mice widely overlapped with that of the human donor in terms of dominant populations, although Bifidobacteria and Lactobacilli disappeared. Despite the small sample size analyzed in this study, these preliminary findings suggest that human intestinal microbiota may promote a high proliferation rate of colonic mucosa. In light of the well-known role of uncontrolled proliferation in colorectal carcinogenesis, these results may deserve further investigation in a larger population study.


Assuntos
Proliferação de Células , Colo , Microbioma Gastrointestinal , Mucosa Intestinal , Animais , Humanos , Mucosa Intestinal/microbiologia , Mucosa Intestinal/metabolismo , Camundongos , Colo/microbiologia , Colo/metabolismo , Masculino , Vida Livre de Germes , Feminino , Camundongos Endogâmicos C3H , Fezes/microbiologia
18.
FASEB J ; 38(11): e23648, 2024 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-38822661

RESUMO

Previous studies on germ-free (GF) animals have described altered anxiety-like and social behaviors together with dysregulations in brain serotonin (5-HT) metabolism. Alterations in circulating 5-HT levels and gut 5-HT metabolism have also been reported in GF mice. In this study, we conducted an integrative analysis of various behaviors as well as markers of 5-HT metabolism in the brain and along the GI tract of GF male mice compared with conventional (CV) ones. We found a strong decrease in locomotor activity, accompanied by some signs of increased anxiety-like behavior in GF mice compared with CV mice. Brain gene expression analysis showed no differences in HTR1A and TPH2 genes. In the gut, we found decreased TPH1 expression in the colon of GF mice, while it was increased in the cecum. HTR1A expression was dramatically decreased in the colon, while HTR4 expression was increased both in the cecum and colon of GF mice compared with CV mice. Finally, SLC6A4 expression was increased in the ileum and colon of GF mice compared with CV mice. Our results add to the evidence that the microbiota is involved in regulation of behavior, although heterogeneity among studies suggests a strong impact of genetic and environmental factors on this microbiota-mediated regulation. While no impact of GF status on brain 5-HT was observed, substantial differences in gut 5-HT metabolism were noted, with tissue-dependent results indicating a varying role of microbiota along the GI tract.


Assuntos
Comportamento Animal , Vida Livre de Germes , Serotonina , Animais , Serotonina/metabolismo , Camundongos , Masculino , Microbioma Gastrointestinal/fisiologia , Encéfalo/metabolismo , Triptofano Hidroxilase/metabolismo , Triptofano Hidroxilase/genética , Ansiedade/metabolismo , Ansiedade/microbiologia , Proteínas da Membrana Plasmática de Transporte de Serotonina/metabolismo , Proteínas da Membrana Plasmática de Transporte de Serotonina/genética , Camundongos Endogâmicos C57BL , Receptor 5-HT1A de Serotonina/metabolismo , Receptor 5-HT1A de Serotonina/genética , Colo/metabolismo , Colo/microbiologia
19.
Gut Microbes ; 16(1): 2363015, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38845453

RESUMO

Gut microbiota is responsible for essential functions in human health. Several communication axes between gut microbiota and other organs via neural, endocrine, and immune pathways have been described, and perturbation of gut microbiota composition has been implicated in the onset and progression of an emerging number of diseases. Here, we analyzed peripheral nerves, dorsal root ganglia (DRG), and skeletal muscles of neonatal and young adult mice with the following gut microbiota status: a) germ-free (GF), b) gnotobiotic, selectively colonized with 12 specific gut bacterial strains (Oligo-Mouse-Microbiota, OMM12), or c) natural complex gut microbiota (CGM). Stereological and morphometric analyses revealed that the absence of gut microbiota impairs the development of somatic median nerves, resulting in smaller diameter and hypermyelinated axons, as well as in smaller unmyelinated fibers. Accordingly, DRG and sciatic nerve transcriptomic analyses highlighted a panel of differentially expressed developmental and myelination genes. Interestingly, the type III isoform of Neuregulin1 (NRG1), known to be a neuronal signal essential for Schwann cell myelination, was overexpressed in young adult GF mice, with consequent overexpression of the transcription factor Early Growth Response 2 (Egr2), a fundamental gene expressed by Schwann cells at the onset of myelination. Finally, GF status resulted in histologically atrophic skeletal muscles, impaired formation of neuromuscular junctions, and deregulated expression of related genes. In conclusion, we demonstrate for the first time a gut microbiota regulatory impact on proper development of the somatic peripheral nervous system and its functional connection to skeletal muscles, thus suggesting the existence of a novel 'Gut Microbiota-Peripheral Nervous System-axis.'


Assuntos
Gânglios Espinais , Microbioma Gastrointestinal , Junção Neuromuscular , Animais , Junção Neuromuscular/microbiologia , Camundongos , Gânglios Espinais/metabolismo , Gânglios Espinais/microbiologia , Vida Livre de Germes , Nervos Periféricos/microbiologia , Nervos Periféricos/crescimento & desenvolvimento , Músculo Esquelético/microbiologia , Camundongos Endogâmicos C57BL , Neuregulina-1/metabolismo , Neuregulina-1/genética , Masculino , Bactérias/classificação , Bactérias/genética , Bactérias/isolamento & purificação , Bactérias/metabolismo , Células de Schwann/microbiologia , Células de Schwann/metabolismo
20.
Gut Microbes ; 16(1): 2361660, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38935764

RESUMO

The microbiota significantly impacts digestive epithelium functionality, especially in nutrient processing. Given the importance of iron for both the host and the microbiota, we hypothesized that host-microbiota interactions fluctuate with dietary iron levels. We compared germ-free (GF) and conventional mice (SPF) fed iron-containing (65 mg/Kg) or iron-depleted (<6 mg/Kg) diets. The efficacy of iron privation was validated by iron blood parameters. Ferritin and Dmt1, which represent cellular iron storage and transport respectively, were studied in tissues where they are abundant: the duodenum, liver and lung. When the mice were fed an iron-rich diet, the microbiota increased blood hemoglobin and hepcidin and the intestinal ferritin levels, suggesting that the microbiota helps iron storage. When iron was limiting, the microbiota inhibited the expression of the intestinal Dmt1 transporter, likely via the pathway triggered by Hif-2α. The microbiota assists the host in storing intestinal iron when it is abundant and competes with the host by inhibiting Dmt1 in conditions of iron scarcity. Comparison between duodenum, liver and lung indicates organ-specific responses to microbiota and iron availability. Iron depletion induced temporal changes in microbiota composition and activity, reduced α-diversity of microbiota, and led to Lactobacillaceae becoming particularly more abundant after 60 days of privation. By inoculating GF mice with a simplified bacterial mixture, we show that the iron-depleted host favors the gut fitness of Bifidobacterium longum.


Assuntos
Proteínas de Transporte de Cátions , Duodeno , Microbioma Gastrointestinal , Hepcidinas , Ferro da Dieta , Fígado , Animais , Camundongos , Microbioma Gastrointestinal/fisiologia , Ferro da Dieta/metabolismo , Ferro da Dieta/administração & dosagem , Proteínas de Transporte de Cátions/metabolismo , Proteínas de Transporte de Cátions/genética , Fígado/metabolismo , Fígado/microbiologia , Duodeno/metabolismo , Duodeno/microbiologia , Hepcidinas/metabolismo , Ferritinas/metabolismo , Vida Livre de Germes , Interações entre Hospedeiro e Microrganismos , Pulmão/microbiologia , Pulmão/metabolismo , Ferro/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Bactérias/classificação , Bactérias/metabolismo , Bactérias/genética , Bactérias/isolamento & purificação , Camundongos Endogâmicos C57BL , Hemoglobinas/metabolismo , Masculino
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...