Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
mBio ; 14(4): e0150423, 2023 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-37526424

RESUMO

The inflammatory bowel diseases (IBD) occur in genetically susceptible individuals who mount inappropriate immune responses to their microbiota leading to chronic intestinal inflammation. Whereas IBD clinical presentation is well described, how interactions between microbiota and host genotype impact early subclinical stages of the disease remains unclear. The transcription factor hepatocyte nuclear factor 4 alpha (HNF4A) has been associated with human IBD, and deletion of Hnf4a in intestinal epithelial cells (IECs) in mice (Hnf4aΔIEC) leads to spontaneous colonic inflammation by 6-12 mo of age. Here, we tested if pathology in Hnf4aΔIEC mice begins earlier in life and if microbiota contribute to that process. Longitudinal analysis revealed that Hnf4aΔIEC mice reared in specific pathogen-free (SPF) conditions develop episodic elevated fecal lipocalin 2 (Lcn2) and loose stools beginning by 4-5 wk of age. Lifetime cumulative Lcn2 levels correlated with histopathological features of colitis at 12 mo. Antibiotic and gnotobiotic tests showed that these phenotypes in Hnf4aΔIEC mice were dependent on microbiota. Fecal 16S rRNA gene sequencing in SPF Hnf4aΔIEC and control mice disclosed that genotype significantly contributed to differences in microbiota composition by 12 mo, and longitudinal analysis of the Hnf4aΔIEC mice with the highest lifetime cumulative Lcn2 revealed that microbial community differences emerged early in life when elevated fecal Lcn2 was first detected. These microbiota differences included enrichment of a novel phylogroup of Akkermansia muciniphila in Hnf4aΔIEC mice. We conclude that HNF4A functions in IEC to shape composition of the gut microbiota and protect against episodic inflammation induced by microbiota throughout the lifespan. IMPORTANCE The inflammatory bowel diseases (IBD), characterized by chronic inflammation of the intestine, affect millions of people around the world. Although significant advances have been made in the clinical management of IBD, the early subclinical stages of IBD are not well defined and are difficult to study in humans. This work explores the subclinical stages of disease in mice lacking the IBD-associated transcription factor HNF4A in the intestinal epithelium. Whereas these mice do not develop overt disease until late in adulthood, we find that they display episodic intestinal inflammation, loose stools, and microbiota changes beginning in very early life stages. Using germ-free and antibiotic-treatment experiments, we reveal that intestinal inflammation in these mice was dependent on the presence of microbiota. These results suggest that interactions between host genotype and microbiota can drive early subclinical pathologies that precede the overt onset of IBD and describe a mouse model to explore those important processes.


Assuntos
Colite , Fator 4 Nuclear de Hepatócito , Doenças Inflamatórias Intestinais , Microbiota , Animais , Humanos , Camundongos , Antibacterianos , Colite/induzido quimicamente , Fator 4 Nuclear de Hepatócito/genética , Inflamação/patologia , Doenças Inflamatórias Intestinais/genética , Intestinos , RNA Ribossômico 16S/genética
2.
Cell Mol Gastroenterol Hepatol ; 14(2): 465-493, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35533983

RESUMO

BACKGROUND & AIMS: The intestine constantly interprets and adapts to complex combinations of dietary and microbial stimuli. However, the transcriptional strategies by which the intestinal epithelium integrates these coincident sources of information remain unresolved. We recently found that microbiota colonization suppresses epithelial activity of hepatocyte nuclear factor 4 nuclear receptor transcription factors, but their integrative regulation was unknown. METHODS: We compared adult mice reared germ-free or conventionalized with a microbiota either fed normally or after a single high-fat meal. Preparations of unsorted jejunal intestinal epithelial cells were queried using lipidomics and genome-wide assays for RNA sequencing and ChIP sequencing for the activating histone mark H3K27ac and hepatocyte nuclear factor 4 alpha. RESULTS: Analysis of lipid classes, genes, and regulatory regions identified distinct nutritional and microbial responses but also simultaneous influence of both stimuli. H3K27ac sites preferentially increased by high-fat meal in the presence of microbes neighbor lipid anabolism and proliferation genes, were previously identified intestinal stem cell regulatory regions, and were not hepatocyte nuclear factor 4 alpha targets. In contrast, H3K27ac sites preferentially increased by high-fat meal in the absence of microbes neighbor targets of the energy homeostasis regulator peroxisome proliferator activated receptor alpha, neighbored fatty acid oxidation genes, were previously identified enterocyte regulatory regions, and were hepatocyte factor 4 alpha bound. CONCLUSIONS: Hepatocyte factor 4 alpha supports a differentiated enterocyte and fatty acid oxidation program in germ-free mice, and that suppression of hepatocyte factor 4 alpha by the combination of microbes and high-fat meal may result in preferential activation of intestinal epithelial cell proliferation programs. This identifies potential transcriptional mechanisms for intestinal adaptation to multiple signals and how microbiota may modulate intestinal lipid absorption, epithelial cell renewal, and systemic energy balance.


Assuntos
Duodeno , Microbioma Gastrointestinal , Mucosa Intestinal , Animais , Duodeno/metabolismo , Duodeno/microbiologia , Ácidos Graxos/metabolismo , Fator 4 Nuclear de Hepatócito/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Lipídeos , Camundongos
3.
Sci Adv ; 7(30)2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34301599

RESUMO

Bile salt synthesis, secretion into the intestinal lumen, and resorption in the ileum occur in all vertebrate classes. In mammals, bile salt composition is determined by host and microbial enzymes, affecting signaling through the bile salt-binding transcription factor farnesoid X receptor (Fxr). However, these processes in other vertebrate classes remain poorly understood. We show that key components of hepatic bile salt synthesis and ileal transport pathways are conserved and under control of Fxr in zebrafish. Zebrafish bile salts consist primarily of a C27 bile alcohol and a C24 bile acid that undergo multiple microbial modifications including bile acid deconjugation that augments Fxr activity. Using single-cell RNA sequencing, we provide a cellular atlas of the zebrafish intestinal epithelium and uncover roles for Fxr in transcriptional and differentiation programs in ileal and other cell types. These results establish zebrafish as a nonmammalian vertebrate model for studying bile salt metabolism and Fxr signaling.


Assuntos
Ácidos e Sais Biliares , Peixe-Zebra , Animais , Ácidos e Sais Biliares/metabolismo , Intestinos , Fígado/metabolismo , Mamíferos/metabolismo , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Peixe-Zebra/metabolismo
4.
Nat Rev Gastroenterol Hepatol ; 18(1): 7-23, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33024279

RESUMO

The intestinal epithelium serves the unique and critical function of harvesting dietary nutrients, while simultaneously acting as a cellular barrier separating tissues from the luminal environment and gut microbial ecosystem. Two salient features of the intestinal epithelium enable it to perform these complex functions. First, cells within the intestinal epithelium achieve a wide range of specialized identities, including different cell types and distinct anterior-posterior patterning along the intestine. Second, intestinal epithelial cells are sensitive and responsive to the dynamic milieu of dietary nutrients, xenobiotics and microorganisms encountered in the intestinal luminal environment. These diverse identities and responsiveness of intestinal epithelial cells are achieved in part through the differential transcription of genes encoded in their shared genome. Here, we review insights from mice and other vertebrate models into the transcriptional regulatory mechanisms underlying intestinal epithelial identity and microbial responsiveness, including DNA methylation, chromatin accessibility, histone modifications and transcription factors. These studies are revealing that most transcription factors involved in intestinal epithelial identity also respond to changes in the microbiota, raising both opportunities and challenges to discern the underlying integrative transcriptional regulatory networks.


Assuntos
Diferenciação Celular/genética , Microambiente Celular/fisiologia , Microbioma Gastrointestinal , Mucosa Intestinal/fisiologia , Animais , Diferenciação Celular/fisiologia , Microambiente Celular/genética , Microbioma Gastrointestinal/genética , Microbioma Gastrointestinal/fisiologia , Regulação da Expressão Gênica , Humanos , Mucosa Intestinal/citologia , Mucosa Intestinal/microbiologia , Intestinos/microbiologia , Intestinos/fisiologia , Camundongos , Modelos Animais , Nutrigenômica , Fenômenos Fisiológicos da Nutrição/genética , Fenômenos Fisiológicos da Nutrição/fisiologia , Transcrição Gênica/genética , Transcrição Gênica/fisiologia , Peixe-Zebra
5.
Proc Natl Acad Sci U S A ; 116(25): 12428-12436, 2019 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-31160464

RESUMO

The nervous system regulates host immunity in complex ways. Vertebrate olfactory sensory neurons (OSNs) are located in direct contact with pathogens; however, OSNs' ability to detect danger and initiate immune responses is unclear. We report that nasal delivery of rhabdoviruses induces apoptosis in crypt OSNs via the interaction of the OSN TrkA receptor with the viral glycoprotein in teleost fish. This signal results in electrical activation of neurons and very rapid proinflammatory responses in the olfactory organ (OO), but dampened inflammation in the olfactory bulb (OB). CD8α+ cells infiltrate the OO within minutes of nasal viral delivery, and TrkA blocking, but not caspase-3 blocking, abrogates this response. Infiltrating CD8α+ cells were TCRαß T cells with a nonconventional phenotype that originated from the microvasculature surrounding the OB and not the periphery. Nasal delivery of viral glycoprotein (G protein) recapitulated the immune responses observed with the whole virus, and antibody blocking of viral G protein abrogated these responses. Ablation of crypt neurons in zebrafish resulted in increased susceptibility to rhabdoviruses. These results indicate a function for OSNs as a first layer of pathogen detection in vertebrates and as orchestrators of nasal-CNS antiviral immune responses.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Vírus da Necrose Hematopoética Infecciosa/imunologia , Neurônios Receptores Olfatórios/fisiologia , Receptor trkA/metabolismo , Animais , Apoptose , Caspase 3/metabolismo , Mucosa Nasal/imunologia , Mucosa Nasal/virologia , Neurônios Receptores Olfatórios/citologia , Neurônios Receptores Olfatórios/virologia , Oncorhynchus mykiss
6.
Front Immunol ; 8: 559, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28555138

RESUMO

Commensal microorganisms inhabit every mucosal surface of teleost fish. At these surfaces, microorganisms directly and indirectly shape the teleost immune system. This review provides a comprehensive overview of how the microbiota and microbiota-derived products influence both the mucosal and systemic immune system of fish. The cross talk between the microbiota and the teleost immune system shifts significantly under stress or disease scenarios rendering commensals into opportunists or pathogens. Lessons learnt from germ-free fish models as well as from oral administration of live probiotics to fish highlight the vast impact that microbiota have on immune development, antibody production, mucosal homeostasis, and resistance to stress. Future studies should dissect the specific mechanisms by which different members of the fish microbiota and the metabolites they produce interact with pathogens, with other commensals, and with the teleost immune system.

7.
Sci Rep ; 7: 41753, 2017 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-28150752

RESUMO

Commensal bacteria co-exist on the mucosal surfaces of all vertebrates. The host's mucosal immune system must tolerate commensals while fighting pathogens. One of the mechanisms used by the mucosal immune system to maintain homeostasis is the secretion of immunoglobulins (Igs) across epithelial barriers, which is achieved via the polymeric immunoglobulin receptor (pIgR). Rainbow trout pIgR is known to transport IgT and IgM across epithelia. However, other biological functions for trout pIgR or trout secretory component (tSC) remain unknown. This study investigates the interaction of tSC with commensal bacteria, pathogenic bacteria and a fungal pathogen. Our results show that the majority of trout skin and gut bacteria are coated in vivo by tSC. In vitro, tSC present in mucus coats trout commensal isolates such as Microbacterium sp., Staphylococcus warneri, Flectobacillus major, Arthrobacter stackebrantii, and Flavobacterium sp. and the pathogens Vibrio anguillarum and Edwardsiella ictaluri with coating levels ranging from 8% to 70%. Moreover, we found that the majority of tSC is in free form in trout mucus and free tSC is able to directly bind bacteria. We propose that binding of free SC to commensal bacteria is a key and conserved mechanism for maintenance of microbial communities in vertebrate mucosal surfaces.


Assuntos
Interações Hospedeiro-Patógeno , Microbiota , Mucosa/metabolismo , Mucosa/microbiologia , Oncorhynchus mykiss/metabolismo , Oncorhynchus mykiss/microbiologia , Animais , Aderência Bacteriana , Microbioma Gastrointestinal , Imunoglobulinas/metabolismo , Muco/metabolismo , Muco/microbiologia , Pele/metabolismo , Pele/microbiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...