Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Mucosal Immunol ; 17(2): 303-313, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38428738

RESUMO

The mammalian gastrointestinal tract hosts a diverse community of trillions of microorganisms, collectively termed the microbiota, which play a fundamental role in regulating tissue physiology and immunity. Recent studies have sought to dissect the cellular and molecular mechanisms mediating communication between the microbiota and host immune system. Epithelial cells line the intestine and form an initial barrier separating the microbiota from underlying immune cells, and disruption of epithelial function has been associated with various conditions ranging from infection to inflammatory bowel diseases and cancer. From several studies, it is now clear that epithelial cells integrate signals from commensal microbes. Importantly, these non-hematopoietic cells also direct regulatory mechanisms that instruct the recruitment and function of microbiota-sensitive immune cells. In this review, we discuss the central role that has emerged for epithelial cells in orchestrating intestinal immunity and highlight epithelial pathways through which the microbiota can calibrate tissue-intrinsic immune responses.


Assuntos
Doenças Inflamatórias Intestinais , Microbiota , Animais , Humanos , Intestinos , Doenças Inflamatórias Intestinais/metabolismo , Sistema Imunitário , Mucosa Intestinal , Mamíferos
2.
J Exp Med ; 220(6)2023 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-36976181

RESUMO

Intestinal epithelial cells (IECs) constitute a critical first line of defense against microbes. While IECs are known to respond to various microbial signals, the precise upstream cues regulating diverse IEC responses are not clear. Here, we discover a dual role for IEC-intrinsic interleukin-1 receptor (IL-1R) signaling in regulating intestinal homeostasis and inflammation. Absence of IL-1R in epithelial cells abrogates a homeostatic antimicrobial program including production of antimicrobial peptides (AMPs). Mice deficient for IEC-intrinsic IL-1R are unable to clear Citrobacter rodentium (C. rodentium) but are protected from DSS-induced colitis. Mechanistically, IL-1R signaling enhances IL-22R-induced signal transducer and activator of transcription 3 (STAT3) phosphorylation in IECs leading to elevated production of AMPs. IL-1R signaling in IECs also directly induces expression of chemokines as well as genes involved in the production of reactive oxygen species. Our findings establish a protective role for IEC-intrinsic IL-1R signaling in combating infections but a detrimental role during colitis induced by epithelial damage.


Assuntos
Colite , Receptores de Interleucina-1 , Camundongos , Animais , Receptores de Interleucina-1/genética , Receptores de Interleucina-1/metabolismo , Intestinos , Colite/metabolismo , Inflamação/metabolismo , Células Epiteliais/metabolismo , Homeostase , Mucosa Intestinal/metabolismo
3.
Cell Host Microbe ; 29(12): 1744-1756.e5, 2021 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-34678170

RESUMO

Interactions between the microbiota and mammalian host are essential for defense against infection, but the microbial-derived cues that mediate this relationship remain unclear. Here, we find that intestinal epithelial cell (IEC)-associated commensal bacteria, segmented filamentous bacteria (SFB), promote early protection against the pathogen Citrobacter rodentium, independent of CD4+ T cells. SFB induced histone modifications in IECs at sites enriched for retinoic acid receptor motifs, suggesting that SFB may enhance defense through retinoic acid (RA). Consistent with this, inhibiting RA signaling suppressed SFB-induced protection. Intestinal RA levels were elevated in SFB mice, despite the inhibition of mammalian RA production, indicating that SFB directly modulate RA. Interestingly, RA was produced by intestinal bacteria, and the loss of bacterial-intrinsic aldehyde dehydrogenase activity decreased the RA levels and increased infection. These data reveal RA as an unexpected microbiota-derived metabolite that primes innate defense and suggests that pre- and probiotic approaches to elevate RA could prevent or combat infections.


Assuntos
Bactérias/metabolismo , Enteropatias/metabolismo , Simbiose , Tretinoína/metabolismo , Animais , Bacillus cereus , Bifidobacterium bifidum , Linfócitos T CD4-Positivos , Citrobacter rodentium , Células Epiteliais , Código das Histonas , Interações entre Hospedeiro e Microrganismos , Enteropatias/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microbiota , Óxido Nítrico , Transdução de Sinais
4.
Front Immunol ; 10: 928, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31134059

RESUMO

Numerous bacterial pathogens infect the mammalian host by initially associating with epithelial cells that line the intestinal lumen. Recent work has revealed that commensal bacteria that reside in the intestine promote defense against pathogenic infection, however whether the microbiota direct host pathways that alter pathogen adherence is not well-understood. Here, by comparing germ-free mice, we identify that the microbiota decrease bacterial pathogen adherence and dampen epithelial expression of the cell surface glycoprotein C-type lectin 2e (Clec2e). Functional studies revealed that overexpression of this lectin promotes adherence of intestinal bacterial pathogens to mammalian cells. Interestingly, microbiota-sensitive downregulation of Clec2e corresponds with decreased histone acetylation of the Clec2e gene in intestinal epithelial cells. Histone deacetylation and transcriptional regulation of Clec2e depends on expression and recruitment of the histone deacetylase HDAC3. Thus, commensal bacteria epigenetically instruct epithelial cells to decrease expression of a C-type lectin that promotes pathogen adherence, revealing a novel mechanism for how the microbiota promote innate defense against infection.


Assuntos
Aderência Bacteriana/fisiologia , Epigênese Genética , Células Epiteliais/metabolismo , Intestinos/microbiologia , Lectinas Tipo C/genética , Microbiota/fisiologia , Acetilação , Animais , Regulação da Expressão Gênica , Células HEK293 , Histona Desacetilases/genética , Histona Desacetilases/metabolismo , Histonas/metabolismo , Humanos , Intestinos/citologia , Lectinas Tipo C/metabolismo , Camundongos Endogâmicos C57BL , Organismos Livres de Patógenos Específicos
5.
PLoS Pathog ; 13(5): e1006388, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28542482

RESUMO

Interferons (IFNs) target macrophages to regulate inflammation and resistance to microbial infections. The type II IFN (IFNγ) acts on a cell surface receptor (IFNGR) to promote gene expression that enhance macrophage inflammatory and anti-microbial activity. Type I IFNs can dampen macrophage responsiveness to IFNγ and are associated with increased susceptibility to numerous bacterial infections. The precise mechanisms responsible for these effects remain unclear. Type I IFNs silence macrophage ifngr1 transcription and thus reduce cell surface expression of IFNGR1. To test how these events might impact macrophage activation and host resistance during bacterial infection, we developed transgenic mice that express a functional FLAG-tagged IFNGR1 (fGR1) driven by a macrophage-specific promoter. Macrophages from fGR1 mice expressed physiologic levels of cell surface IFNGR1 at steady state and responded equivalently to WT C57Bl/6 macrophages when treated with IFNγ alone. However, fGR1 macrophages retained cell surface IFNGR1 and showed enhanced responsiveness to IFNγ in the presence of type I IFNs. When fGR1 mice were infected with the bacterium Listeria monocytogenes their resistance was significantly increased, despite normal type I and II IFN production. Enhanced resistance was dependent on IFNγ and associated with increased macrophage activation and antimicrobial function. These results argue that down regulation of myeloid cell IFNGR1 is an important mechanism by which type I IFNs suppress inflammatory and anti-bacterial functions of macrophages.


Assuntos
Listeria monocytogenes/fisiologia , Listeriose/imunologia , Macrófagos/imunologia , Receptores de Interferon/genética , Animais , Regulação para Baixo , Feminino , Humanos , Interferon Tipo I/imunologia , Listeriose/genética , Listeriose/microbiologia , Ativação de Macrófagos , Masculino , Camundongos , Camundongos Knockout , Receptores de Interferon/imunologia , Receptor de Interferon gama
6.
J Immunol ; 191(6): 3384-92, 2013 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-23935197

RESUMO

The ability of type I IFNs to increase susceptibility to certain bacterial infections correlates with downregulation of myeloid cell surface IFNGR, the receptor for the type II IFN (IFN-γ), and reduced myeloid cell responsiveness to IFN-γ. In this study, we show that the rapid reductions in mouse and human myeloid cell surface IFNGR1 expression that occur in response to type I IFN treatment reflect a rapid silencing of new ifngr1 transcription by repressive transcriptional regulators. Treatment of macrophages with IFN-ß reduced cellular abundance of ifngr1 transcripts as rapidly and effectively as actinomycin D treatment. IFN-ß treatment also significantly reduced the amounts of activated RNA polymerase II (pol II) and acetylated histones H3 and H4 at the ifngr1 promoter and the activity of an IFNGR1-luc reporter construct in macrophages. The suppression of IFNGR1-luc activity required an intact early growth response factor (Egr) binding site in the proximal ifngr1 promoter. Three Egr proteins and two Egr/NGFI-A binding (Nab) proteins were found to be expressed in bone macrophages, but only Egr3 and Nab1 were recruited to the ifngr1 promoter upon IFN-ß stimulation. Knockdown of Nab1 in a macrophage cell line prevented downregulation of IFNGR1 and prevented the loss of acetylated histones from the ifngr1 promoter. These data suggest that type I IFN stimulation induces a rapid recruitment of a repressive Egr3/Nab1 complex that silences transcription from the ifngr1 promoter. This mechanism of gene silencing may contribute to the anti-inflammatory effects of type I IFNs.


Assuntos
Proteína 3 de Resposta de Crescimento Precoce/metabolismo , Inativação Gênica/fisiologia , Interferon Tipo I/metabolismo , Receptores de Interferon/metabolismo , Proteínas Repressoras/metabolismo , Animais , Western Blotting , Imunoprecipitação da Cromatina , Regulação para Baixo , Proteína 3 de Resposta de Crescimento Precoce/imunologia , Citometria de Fluxo , Humanos , Interferon Tipo I/imunologia , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Células Mieloides/imunologia , Células Mieloides/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Receptores de Interferon/imunologia , Proteínas Repressoras/imunologia , Transcrição Gênica , Receptor de Interferon gama
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA