Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
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.
Cancer Med ; 12(3): 2945-2957, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36056757

RESUMO

BACKGROUND AND AIMS: Colorectal cancer (CRC) incidence is increasing in young patients without a clear etiology. Emerging data have implicated the fecal microbiome in CRC carcinogenesis. However, its impact on young onset CRC is poorly defined. METHODS: We performed a meta-analysis of fecal metagenomics sequencing data from n = 692 patients with CRC and n = 602 healthy controls from eleven studies to evaluate features of the fecal metagenome associated with CRC. We hypothesized that known carcinogenic virulence factors (colibactin, fadA) and species abundance may be differentially enriched in young CRC patients relative to older CRC patients and controls. RESULTS: Summary odds ratios (OR) for CRC were increased with the presence of colibactin (OR 1.92 95% CI 1.08-3.38), fadA (OR 4.57 95% CI 1.63-12.85), and F. nucleatum (OR 6.93 95% CI 3.01-15.96) in meta-analysis models adjusted for age, gender, and body mass index. The OR for CRC for the presence of E.coli was 2.02 (0.92-4.45). An increase in the prevalence of Fusobacterium nucleatum (OR = 1.40 [1.18; 1.65]) and Escherichia coli (OR = 1.14 [1.02; 1.28]) per 10-year increase in age was observed in models including samples from both CRC and healthy controls. Species relative abundance was differentially enriched in young CRC patients for five species-Intestinimonas butyriciproducens, Holdemania filiformis, Firimicutues bacterium CAG 83, Bilophilia wadsworthia, and Alistipes putredinis. CONCLUSION: In this study, we observed strong associations with CRC status for colibactin, fadA, and Fusobacterium nucleatum with CRC relative to controls. In addition, we identified several microbial species differentially enriched in young colorectal cancer patients. Studies targeting the young CRC patients are warranted to elucidate underlying preclinical mechanisms.


Assuntos
Neoplasias Colorretais , Microbiota , Humanos , Neoplasias Colorretais/genética , Metagenoma , Metagenômica , Fusobacterium nucleatum , Carcinogênese/genética
4.
Immunity ; 55(12): 2222-2224, 2022 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-36516815

RESUMO

Cellular dynamics that influence mucosal healing are not well understood. In this issue of Immunity, Frede, Czarnewski, Monasterio et al. find that B cells accumulate in the colon following intestinal injury. These B cells impair epithelial repair by hindering local stromal-epithelial interactions.


Assuntos
Intenção , Mucosa Intestinal , Colo , Células Epiteliais
5.
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
6.
Immunity ; 52(2): 275-294.e9, 2020 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-32075728

RESUMO

Type 3 innate lymphoid cells (ILC3s) are critical for lung defense against bacterial pneumonia in the neonatal period, but the signals that guide pulmonary ILC3 development remain unclear. Here, we demonstrated that pulmonary ILC3s descended from ILC precursors that populated a niche defined by fibroblasts in the developing lung. Alveolar fibroblasts produced insulin-like growth factor 1 (IGF1), which instructed expansion and maturation of pulmonary ILC precursors. Conditional ablation of IGF1 in alveolar fibroblasts or deletion of the IGF-1 receptor from ILC precursors interrupted ILC3 biogenesis and rendered newborn mice susceptible to pneumonia. Premature infants with bronchopulmonary dysplasia, characterized by interrupted postnatal alveolar development and increased morbidity to respiratory infections, had reduced IGF1 concentrations and pulmonary ILC3 numbers. These findings indicate that the newborn period is a critical window in pulmonary immunity development, and disrupted lung development in prematurely born infants may have enduring effects on host resistance to respiratory infections.


Assuntos
Imunidade Inata , Fator de Crescimento Insulin-Like I/metabolismo , Pulmão/imunologia , Linfócitos/citologia , Células Epiteliais Alveolares/metabolismo , Animais , Animais Recém-Nascidos , Displasia Broncopulmonar/imunologia , Diferenciação Celular , Proliferação de Células , Suscetibilidade a Doenças/imunologia , Humanos , Recém-Nascido , Recém-Nascido Prematuro , Fator de Crescimento Insulin-Like I/deficiência , Interleucinas/metabolismo , Pulmão/citologia , Pulmão/crescimento & desenvolvimento , Linfócitos/metabolismo , Camundongos , Pneumonia/imunologia , Proteína com Dedos de Zinco da Leucemia Promielocítica/metabolismo , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/metabolismo , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Transdução de Sinais , Interleucina 22
7.
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
8.
JCI Insight ; 3(18)2018 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-30232290

RESUMO

Altered response to the intestinal microbiota strongly associates with inflammatory bowel disease (IBD); however, how commensal microbial cues are integrated by the host during the pathogenesis of IBD is not understood. Epigenetics represents a potential mechanism that could enable intestinal microbes to modulate transcriptional output during the development of IBD. Here, we reveal a histone methylation signature of intestinal epithelial cells isolated from the terminal ilea of newly diagnosed pediatric IBD patients. Genes characterized by significant alterations in histone H3-lysine 4 trimethylation (H3K4me3) showed differential enrichment in pathways involving immunoregulation, cell survival and signaling, and metabolism. Interestingly, a large subset of these genes was epigenetically regulated by microbiota in mice and several microbiota-sensitive epigenetic targets demonstrated altered expression in IBD patients. Remarkably though, a substantial proportion of these genes exhibited H3K4me3 levels that correlated with the severity of intestinal inflammation in IBD, despite lacking significant differential expression. Collectively, these data uncover a previously unrecognized epigenetic profile of IBD that can be primed by commensal microbes and indicate sensitive targets in the epithelium that may underlie how microbiota predispose to subsequent intestinal inflammation and disease.


Assuntos
Doença de Crohn/metabolismo , Epigênese Genética , Microbioma Gastrointestinal/fisiologia , Inflamação , Adolescente , Animais , Criança , Células Epiteliais/metabolismo , Feminino , Histonas/metabolismo , Humanos , Íleo , Masculino , Metilação , Camundongos , Camundongos Endogâmicos C57BL
9.
Gastroenterology ; 155(2): 501-513, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29689264

RESUMO

BACKGROUND & AIMS: Intestinal microbiota modulate metabolism and associate closely with epithelial cells in the intestine. In intestinal epithelial cells (IECs), histone deacetylase 3 (HDAC3) integrates microbiota-derived signals to control intestinal homeostasis. We investigated whether HDAC3 in IECs regulates metabolism and the development of obesity in mice. METHODS: Adult C57BL/6 (control) mice and mice with constitutive or inducible IEC-specific disruption of Hdac3 (HDAC3ΔIEC mice) were placed on a standard chow or high-fat diet (HFD, 60% kcal from fat). We measured body composition, weight, glucose tolerance, and energy expenditure. IECs were isolated from small intestine and gene expression, and lipid levels were analyzed. HDAC3 levels were determined in 43 pediatric patient ileal biopsy samples and compared with body weight. RESULTS: Control mice fed an HFD gained weight, became obese, and had reduced glucose tolerance with increased serum insulin, whereas HFD-fed HDAC3ΔIEC mice did not develop obesity. Serum levels of triglycerides were reduced in HDAC3ΔIEC mice, and these mice had less liver fat and smaller adipocytes, compared with HFD-fed control mice. HDAC3ΔIEC mice had similar food intake and activity as control mice, but higher energy expenditure because of increased catabolism. IECs from HDAC3ΔIEC mice had altered expression levels of genes that regulate metabolism in response to the microbiota (such as Chka, Mttp, Apoa1, and Pck1) and accumulated triglycerides compared with IECs from control mice. The microbiota-derived short-chain fatty acid butyrate was decreased in obese mice. Butyrate significantly reduced the activity of HDAC3 and increased Pck1 expression in only control IECs. Administration of butyrate to control mice with diet-induced obesity, but not HDAC3ΔIEC mice, led to significant weight loss. Disruption of HDAC3 in IECs of mice after they became obese led to weight loss and improved metabolic profile. Levels of HDAC3 in intestinal biopsy samples correlated with patient weight. CONCLUSIONS: We found that epithelial HDAC3 promotes development of diet-induced obesity in studies of mice and that butyrate reduces activity of HDAC3 in IECs to prevent diet-induced obesity. This pathway might be manipulated to prevent or reduce obesity-associated disease.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Células Epiteliais/metabolismo , Microbioma Gastrointestinal/fisiologia , Histona Desacetilases/metabolismo , Obesidade/patologia , Animais , Biópsia , Peso Corporal/fisiologia , Criança , Modelos Animais de Doenças , Metabolismo Energético , Feminino , Histona Desacetilases/genética , Humanos , Íleo/citologia , Íleo/microbiologia , Íleo/patologia , Resistência à Insulina , Mucosa Intestinal/citologia , Mucosa Intestinal/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Obesidade/etiologia , Obesidade/fisiopatologia
10.
J Exp Med ; 212(10): 1513-28, 2015 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-26371187

RESUMO

Innate lymphoid cells (ILCs) are critical for maintaining epithelial barrier integrity at mucosal surfaces; however, the tissue-specific factors that regulate ILC responses remain poorly characterized. Using mice with intestinal epithelial cell (IEC)-specific deletions in either inhibitor of κB kinase (IKK)α or IKKß, two critical regulators of NFκB activation, we demonstrate that IEC-intrinsic IKKα expression selectively regulates group 3 ILC (ILC3)-dependent antibacterial immunity in the intestine. Although IKKß(ΔIEC) mice efficiently controlled Citrobacter rodentium infection, IKKα(ΔIEC) mice exhibited severe intestinal inflammation, increased bacterial dissemination to peripheral organs, and increased host mortality. Consistent with weakened innate immunity to C. rodentium, IKKα(ΔIEC) mice displayed impaired IL-22 production by RORγt(+) ILC3s, and therapeutic delivery of rIL-22 or transfer of sort-purified IL-22-competent ILCs from control mice could protect IKKα(ΔIEC) mice from C. rodentium-induced morbidity. Defective ILC3 responses in IKKα(ΔIEC) mice were associated with overproduction of thymic stromal lymphopoietin (TSLP) by IECs, which negatively regulated IL-22 production by ILC3s and impaired innate immunity to C. rodentium. IEC-intrinsic IKKα expression was similarly critical for regulation of intestinal inflammation after chemically induced intestinal damage and colitis. Collectively, these data identify a previously unrecognized role for epithelial cell-intrinsic IKKα expression and TSLP in regulating ILC3 responses required to maintain intestinal barrier immunity.


Assuntos
Quinase I-kappa B/metabolismo , Imunidade Inata/imunologia , Linfócitos/imunologia , Animais , Citrobacter rodentium/patogenicidade , Colite/imunologia , Colite/patologia , Colo/imunologia , Colo/metabolismo , Colo/microbiologia , Citocinas/metabolismo , Infecções por Enterobacteriaceae/imunologia , Infecções por Enterobacteriaceae/mortalidade , Células Epiteliais/metabolismo , Feminino , Quinase I-kappa B/genética , Quinase I-kappa B/imunologia , Interleucinas/genética , Interleucinas/metabolismo , Interleucinas/farmacologia , Linfócitos/microbiologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacologia , Linfopoietina do Estroma do Timo , Interleucina 22
11.
Toxicol Pathol ; 43(1): 101-6, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25330924

RESUMO

The mammalian gastrointestinal tract is home to trillions of commensal microorganisms that collectively make up the intestinal microbiota. These microbes are important environmental factors that regulate homeostasis, and alterations in the composition of the microbiota have been associated with several diseases, including inflammatory bowel disease, diabetes, and cancer. New research is beginning to uncover epigenomic pathways that may regulate this relationship with the microbiota. Epigenomic modifications alter the structure of the chromatin and therefore regulate the transcriptional program of a cell. These modifications are maintained by the dynamic activity of various modifying and demodifying enzymes, the activities of which can be influenced by metabolites and other environmental cues. Histone deacetylases (HDACs) are a class of epigenomic-modifying enzymes that are regulated by both endogenous and exogenous factors, and recent studies have suggested that host HDAC expression is important for regulating communication between the intestinal microbiota and mammalian host cells.


Assuntos
Epigenômica/métodos , Microbiota/genética , Animais , Humanos
12.
Nature ; 504(7478): 153-7, 2013 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-24185009

RESUMO

The development and severity of inflammatory bowel diseases and other chronic inflammatory conditions can be influenced by host genetic and environmental factors, including signals derived from commensal bacteria. However, the mechanisms that integrate these diverse cues remain undefined. Here we demonstrate that mice with an intestinal epithelial cell (IEC)-specific deletion of the epigenome-modifying enzyme histone deacetylase 3 (HDAC3(ΔIEC) mice) exhibited extensive dysregulation of IEC-intrinsic gene expression, including decreased basal expression of genes associated with antimicrobial defence. Critically, conventionally housed HDAC3(ΔIEC) mice demonstrated loss of Paneth cells, impaired IEC function and alterations in the composition of intestinal commensal bacteria. In addition, HDAC3(ΔIEC) mice showed significantly increased susceptibility to intestinal damage and inflammation, indicating that epithelial expression of HDAC3 has a central role in maintaining intestinal homeostasis. Re-derivation of HDAC3(ΔIEC) mice into germ-free conditions revealed that dysregulated IEC gene expression, Paneth cell homeostasis and intestinal barrier function were largely restored in the absence of commensal bacteria. Although the specific mechanisms through which IEC-intrinsic HDAC3 expression regulates these complex phenotypes remain to be determined, these data indicate that HDAC3 is a critical factor that integrates commensal-bacteria-derived signals to calibrate epithelial cell responses required to establish normal host-commensal relationships and maintain intestinal homeostasis.


Assuntos
Regulação da Expressão Gênica , Histona Desacetilases/metabolismo , Homeostase , Mucosa Intestinal/enzimologia , Intestinos/microbiologia , Simbiose , Adulto , Animais , Bactérias/genética , Colite Ulcerativa/enzimologia , Colite Ulcerativa/genética , Colite Ulcerativa/microbiologia , Doença de Crohn/enzimologia , Doença de Crohn/genética , Doença de Crohn/microbiologia , Feminino , Deleção de Genes , Perfilação da Expressão Gênica , Histona Desacetilases/genética , Humanos , Mucosa Intestinal/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Celulas de Paneth/citologia , Celulas de Paneth/metabolismo , RNA Ribossômico 16S/genética , Transdução de Sinais
13.
J Immunol ; 190(5): 2292-300, 2013 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23355735

RESUMO

Resistin-like molecule (RELM)α belongs to a family of secreted mammalian proteins that have putative immunomodulatory functions. Recent studies have identified a pathogenic role for RELMα in chemically induced colitis through effects on innate cell populations. However, whether RELMα regulates intestinal adaptive immunity to enteric pathogens is unknown. In this study, we employed Citrobacter rodentium as a physiologic model of pathogenic Escherichia coli-induced diarrheal disease, colitis, and Th17 cell responses. In response to Citrobacter, RELMα expression was induced in intestinal epithelial cells, infiltrating macrophages, and eosinophils of the infected colons. Citrobacter-infected RELMα(-/-) mice exhibited reduced infection-induced intestinal inflammation, characterized by decreased leukocyte recruitment to the colons and reduced immune cell activation compared with wild-type (WT) mice. Interestingly, Citrobacter colonization and clearance were unaffected in RELMα(-/-) mice, suggesting that the immune stimulatory effects of RELMα following Citrobacter infection were pathologic rather than host-protective. Furthermore, infected RELMα(-/-) mice exhibited decreased CD4(+) T cell expression of the proinflammatory cytokine IL-17A. To directly test whether RELMα promoted Citrobacter-induced intestinal inflammation via IL-17A, infected WT and IL-17A(-/-) mice were treated with rRELMα. RELMα treatment of Citrobacter-infected WT mice exacerbated intestinal inflammation and IL-17A expression whereas IL-17A(-/-) mice were protected from RELMα-induced intestinal inflammation. Finally, infected RELMα(-/-) mice exhibited reduced levels of serum IL-23p19 compared with WT mice, and RELMα(-/-) peritoneal macrophages showed deficient IL-23p19 induction. Taken together, these data identify a proinflammatory role for RELMα in bacterial-induced colitis and suggest that the IL-23/Th17 axis is a critical mediator of RELMα-induced inflammation.


Assuntos
Citrobacter rodentium/imunologia , Inflamação/imunologia , Peptídeos e Proteínas de Sinalização Intercelular/imunologia , Interleucina-17/imunologia , Intestinos/efeitos dos fármacos , Macrófagos Peritoneais/efeitos dos fármacos , Células Th17/efeitos dos fármacos , Imunidade Adaptativa/efeitos dos fármacos , Animais , Citrobacter rodentium/patogenicidade , Sulfato de Dextrana , Eosinófilos/efeitos dos fármacos , Eosinófilos/imunologia , Eosinófilos/patologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/imunologia , Células Epiteliais/patologia , Feminino , Expressão Gênica/efeitos dos fármacos , Inflamação/induzido quimicamente , Inflamação/microbiologia , Inflamação/patologia , Peptídeos e Proteínas de Sinalização Intercelular/deficiência , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Interleucina-17/deficiência , Interleucina-17/genética , Subunidade p19 da Interleucina-23/sangue , Subunidade p19 da Interleucina-23/imunologia , Intestinos/imunologia , Intestinos/microbiologia , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/farmacologia , Células Th17/imunologia , Células Th17/patologia
14.
Science ; 336(6086): 1321-5, 2012 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-22674331

RESUMO

The mammalian intestinal tract is colonized by trillions of beneficial commensal bacteria that are anatomically restricted to specific niches. However, the mechanisms that regulate anatomical containment remain unclear. Here, we show that interleukin-22 (IL-22)-producing innate lymphoid cells (ILCs) are present in intestinal tissues of healthy mammals. Depletion of ILCs resulted in peripheral dissemination of commensal bacteria and systemic inflammation, which was prevented by administration of IL-22. Disseminating bacteria were identified as Alcaligenes species originating from host lymphoid tissues. Alcaligenes was sufficient to promote systemic inflammation after ILC depletion in mice, and Alcaligenes-specific systemic immune responses were associated with Crohn's disease and progressive hepatitis C virus infection in patients. Collectively, these data indicate that ILCs regulate selective containment of lymphoid-resident bacteria to prevent systemic inflammation associated with chronic diseases.


Assuntos
Alcaligenes/fisiologia , Interleucinas/imunologia , Intestinos/imunologia , Linfócitos/imunologia , Tecido Linfoide/imunologia , Tecido Linfoide/microbiologia , Adulto , Alcaligenes/imunologia , Alcaligenes/isolamento & purificação , Animais , Translocação Bacteriana , Doença de Crohn/imunologia , Doença de Crohn/microbiologia , Hepatite C Crônica/imunologia , Hepatite C Crônica/microbiologia , Humanos , Imunidade Inata , Inflamação , Interleucinas/administração & dosagem , Interleucinas/biossíntese , Intestinos/microbiologia , Complexo Antígeno L1 Leucocitário/metabolismo , Fígado/microbiologia , Linfonodos/imunologia , Macaca mulatta , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Baço/microbiologia , Adulto Jovem , Interleucina 22
15.
Immunity ; 37(1): 158-70, 2012 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-22705104

RESUMO

Signals from commensal bacteria can influence immune cell development and susceptibility to infectious or inflammatory diseases. However, the mechanisms by which commensal bacteria regulate protective immunity after exposure to systemic pathogens remain poorly understood. Here, we demonstrate that antibiotic-treated (ABX) mice exhibit impaired innate and adaptive antiviral immune responses and substantially delayed viral clearance after exposure to systemic LCMV or mucosal influenza virus. Furthermore, ABX mice exhibited severe bronchiole epithelial degeneration and increased host mortality after influenza virus infection. Genome-wide transcriptional profiling of macrophages isolated from ABX mice revealed decreased expression of genes associated with antiviral immunity. Moreover, macrophages from ABX mice exhibited defective responses to type I and type II IFNs and impaired capacity to limit viral replication. Collectively, these data indicate that commensal-derived signals provide tonic immune stimulation that establishes the activation threshold of the innate immune system required for optimal antiviral immunity.


Assuntos
Bactérias/imunologia , Imunidade Inata , Vírus/imunologia , Imunidade Adaptativa , Animais , Antibacterianos/farmacologia , Infecções por Arenaviridae/genética , Infecções por Arenaviridae/imunologia , Bactérias/efeitos dos fármacos , Suscetibilidade a Doenças/imunologia , Interferons/imunologia , Vírus da Coriomeningite Linfocítica/imunologia , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/genética , Infecções por Orthomyxoviridae/imunologia
16.
Genes Dev ; 25(23): 2480-8, 2011 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-22156208

RESUMO

Macrophages, a key cellular component of inflammation, become functionally polarized in a signal- and context-specific manner. Th2 cytokines such as interleukin 4 (IL-4) polarize macrophages to a state of alternative activation that limits inflammation and promotes wound healing. Alternative activation is mediated by a transcriptional program that is influenced by epigenomic modifications, including histone acetylation. Here we report that macrophages lacking histone deacetylase 3 (HDAC3) display a polarization phenotype similar to IL-4-induced alternative activation and, furthermore, are hyperresponsive to IL-4 stimulation. Throughout the macrophage genome, HDAC3 deacetylates histone tails at regulatory regions, leading to repression of many IL-4-regulated genes characteristic of alternative activation. Following exposure to Schistosoma mansoni eggs, a model of Th2 cytokine-mediated disease that is limited by alternative activation, pulmonary inflammation was ameliorated in mice lacking HDAC3 in macrophages. Thus, HDAC3 functions in alternative activation as a brake whose release could be of benefit in the treatment of multiple inflammatory diseases.


Assuntos
Epigênese Genética , Histona Desacetilases/genética , Ativação de Macrófagos/genética , Macrófagos/metabolismo , Animais , Histona Desacetilases/metabolismo , Interleucina-4/genética , Interleucina-4/metabolismo , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos , Pneumonia/enzimologia , Pneumonia/imunologia , Pneumonia/parasitologia , Schistosoma mansoni , Células Th2/imunologia , Células Th2/metabolismo
17.
Nat Immunol ; 12(11): 1045-54, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21946417

RESUMO

Innate lymphoid cells (ILCs), a heterogeneous cell population, are critical in orchestrating immunity and inflammation in the intestine, but whether ILCs influence immune responses or tissue homeostasis at other mucosal sites remains poorly characterized. Here we identify a population of lung-resident ILCs in mice and humans that expressed the alloantigen Thy-1 (CD90), interleukin 2 (IL-2) receptor a-chain (CD25), IL-7 receptor a-chain (CD127) and the IL-33 receptor subunit T1-ST2. Notably, mouse ILCs accumulated in the lung after infection with influenza virus, and depletion of ILCs resulted in loss of airway epithelial integrity, diminished lung function and impaired airway remodeling. These defects were restored by administration of the lung ILC product amphiregulin. Collectively, our results demonstrate a critical role for lung ILCs in restoring airway epithelial integrity and tissue homeostasis after infection with influenza virus.


Assuntos
Homeostase , Imunidade Inata , Influenza Humana/imunologia , Pulmão/metabolismo , Infecções por Orthomyxoviridae/imunologia , Orthomyxoviridae/imunologia , Mucosa Respiratória/metabolismo , Remodelação das Vias Aéreas/efeitos dos fármacos , Remodelação das Vias Aéreas/imunologia , Anfirregulina , Animais , Antígenos CD/biossíntese , Células Cultivadas , Família de Proteínas EGF , Glicoproteínas/farmacologia , Homeostase/imunologia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Interleucina-33 , Interleucinas/metabolismo , Pulmão/imunologia , Pulmão/patologia , Pulmão/virologia , Camundongos , Camundongos Endogâmicos C57BL , Mucosa Respiratória/imunologia , Mucosa Respiratória/patologia , Mucosa Respiratória/virologia , Cicatrização
18.
J Neurosci ; 31(2): 764-74, 2011 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-21228185

RESUMO

Gene expression is dynamically regulated by chromatin modifications on histone tails, such as acetylation. In general, histone acetylation promotes transcription, whereas histone deacetylation negatively regulates transcription. The interplay between histone acetyltranserases and histone deacetylases (HDACs) is pivotal for the regulation of gene expression required for long-term memory processes. Currently, very little is known about the role of individual HDACs in learning and memory. We examined the role of HDAC3 in long-term memory using a combined genetic and pharmacologic approach. We used HDAC3-FLOX genetically modified mice in combination with adeno-associated virus-expressing Cre recombinase to generate focal homozygous deletions of Hdac3 in area CA1 of the dorsal hippocampus. To complement this approach, we also used a selective inhibitor of HDAC3, RGFP136 [N-(6-(2-amino-4-fluorophenylamino)-6-oxohexyl)-4-methylbenzamide]. Immunohistochemistry showed that focal deletion or intrahippocampal delivery of RGFP136 resulted in increased histone acetylation. Both the focal deletion of HDAC3 as well as HDAC3 inhibition via RGFP136 significantly enhanced long-term memory in a persistent manner. Next we examined expression of genes implicated in long-term memory from dorsal hippocampal punches using quantitative reverse transcription-PCR. Expression of nuclear receptor subfamily 4 group A, member 2 (Nr4a2) and c-fos was significantly increased in the hippocampus of HDAC3-FLOX mice compared with wild-type controls. Memory enhancements observed in HDAC3-FLOX mice were abolished by intrahippocampal delivery of Nr4a2 small interfering RNA, suggesting a mechanism by which HDAC3 negatively regulates memory formation. Together, these findings demonstrate a critical role for HDAC3 in the molecular mechanisms underlying long-term memory formation.


Assuntos
Benzamidas/farmacologia , Histona Desacetilases/fisiologia , Memória de Longo Prazo/fisiologia , Acetilação , Animais , Hipocampo/enzimologia , Inibidores de Histona Desacetilases/farmacologia , Histona Desacetilases/biossíntese , Histona Desacetilases/genética , Histonas/metabolismo , Memória de Longo Prazo/efeitos dos fármacos , Camundongos , Camundongos Mutantes , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares/biossíntese , Deleção de Sequência , Percepção Espacial/efeitos dos fármacos , Percepção Espacial/fisiologia
19.
Cancer Res ; 66(18): 9316-22, 2006 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-16982777

RESUMO

Cells are frequently challenged by DNA double-strand breaks (DSB) that threaten their normal function and survival. In mammalian cells, the repair of DSBs is predominantly mediated by the DNA-dependent protein kinase (DNA-PK) complex. We unexpectedly found that the corepressor silencing mediator for retinoid and thyroid hormone receptor (SMRT) associates with the DNA-PK repair complex. The SMRT/histone deacetylase 3 complex is required for the transcriptional repressive property of the Ku70 subunit of the repair complex. Moreover, SMRT, but not the related Nuclear Receptor Corepressor, is required for cellular recovery from DNA DSBs induced by ionizing radiation or DNA damage-inducing drugs. Thus, the corepressor SMRT plays a novel and critical role in the cellular response to DSBs.


Assuntos
Reparo do DNA/fisiologia , Proteínas de Ligação a DNA/fisiologia , Proteínas Repressoras/fisiologia , Antígenos Nucleares/metabolismo , Dano ao DNA , Proteína Quinase Ativada por DNA/metabolismo , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Células HeLa , Histona Desacetilases/metabolismo , Humanos , Autoantígeno Ku , Proteínas Nucleares/antagonistas & inibidores , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Correpressor 1 de Receptor Nuclear , Correpressor 2 de Receptor Nuclear , RNA Interferente Pequeno/genética , Proteínas Repressoras/antagonistas & inibidores , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Especificidade por Substrato , Ativação Transcricional , Transfecção
20.
EMBO J ; 25(17): 3966-74, 2006 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-16917504

RESUMO

Unliganded thyroid hormone receptor (TR) actively represses transcription via the nuclear receptor corepressor (N-CoR)/histone deacetylase 3 (HDAC3) complex. Although transcriptional activation by liganded receptors involves chromatin remodeling, the role of ATP-dependent remodeling in receptor-mediated repression is unknown. Here we report that SNF2H, the mammalian ISWI chromatin remodeling ATPase, is critical for repression of a genomically integrated, TR-regulated reporter gene. N-CoR and HDAC3 are both required for recruitment of SNF2H to the repressed gene. SNF2H does not interact directly with the N-CoR/HDAC3 complex, but binds to unacetylated histone H4 tails, suggesting that deacetylase activity of the corepressor complex is critical to SNF2H function. Indeed, HDAC3 as well as SNF2H are required for nucleosomal organization on the TR target gene. Consistent with these findings, reduction of SNF2H induces expression of an endogenous TR-regulated gene, dio1, in liver cells. Thus, although not apparent from studies of transiently transfected reporter genes, gene repression by TR involves the targeting of chromatin remodeling factors to repressed genes by the HDAC activity of nuclear receptor corepressors.


Assuntos
Adenosina Trifosfatases/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Proteínas de Ligação a DNA/metabolismo , Histona Desacetilases/metabolismo , Proteínas Nucleares/metabolismo , Receptores dos Hormônios Tireóideos/fisiologia , Proteínas Repressoras/metabolismo , Fatores de Transcrição/metabolismo , Adenosina Trifosfatases/genética , Animais , Montagem e Desmontagem da Cromatina , Proteínas Cromossômicas não Histona/genética , Proteínas de Ligação a DNA/genética , Genes Reporter , Histona Desacetilases/genética , Histonas/metabolismo , Humanos , Luciferases/genética , Camundongos , Proteínas Nucleares/genética , Correpressor 1 de Receptor Nuclear , Regiões Promotoras Genéticas , Interferência de RNA , Proteínas Repressoras/genética , Fatores de Transcrição/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA