Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Nat Immunol ; 12(4): 320-6, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21336274

RESUMO

Lymphoid cells that express the nuclear hormone receptor RORγt are involved in containment of the large intestinal microbiota and defense against pathogens through the production of interleukin 17 (IL-17) and IL-22. They include adaptive IL-17-producing helper T cells (T(H)17 cells), as well as innate lymphoid cells (ILCs) such as lymphoid tissue-inducer (LTi) cells and IL-22-producing NKp46+ cells. Here we show that in contrast to T(H)17 cells, both types of RORγt+ ILCs constitutively produced most of the intestinal IL-22 and that the symbiotic microbiota repressed this function through epithelial expression of IL-25. This function was greater in the absence of adaptive immunity and was fully restored and required after epithelial damage, which demonstrates a central role for RORγt+ ILCs in intestinal homeostasis. Our data identify a finely tuned equilibrium among intestinal symbionts, adaptive immunity and RORγt+ ILCs.


Assuntos
Intestinos/imunologia , Tecido Linfoide/imunologia , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Transdução de Sinais/imunologia , Imunidade Adaptativa/imunologia , Animais , Antígenos Ly/genética , Antígenos Ly/metabolismo , Feminino , Citometria de Fluxo , Homeostase/imunologia , Humanos , Interleucina-17/genética , Interleucina-17/metabolismo , Interleucinas/genética , Interleucinas/metabolismo , Mucosa Intestinal/metabolismo , Intestinos/microbiologia , Tecido Linfoide/citologia , Tecido Linfoide/metabolismo , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Receptor 1 Desencadeador da Citotoxicidade Natural/genética , Receptor 1 Desencadeador da Citotoxicidade Natural/metabolismo , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Simbiose/imunologia , Fatores de Tempo , Interleucina 22
2.
J Allergy Clin Immunol ; 147(1): 335-348.e11, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32407834

RESUMO

BACKGROUND: The cross-talk between the host and its microbiota plays a key role in the promotion of health. The production of metabolites such as polyamines by intestinal-resident bacteria is part of this symbiosis shaping host immunity. The polyamines putrescine, spermine, and spermidine are abundant within the gastrointestinal tract and might substantially contribute to gut immunity. OBJECTIVE: We aimed to characterize the polyamine spermidine as a modulator of T-cell differentiation and function. METHODS: Naive T cells were isolated from wild-type mice or cord blood from healthy donors and submitted to polarizing cytokines, with and without spermidine treatment, to evaluate CD4+ T-cell differentiation in vitro. Moreover, mice were subjected to oral supplementation of spermidine, or its precursor l-arginine, to assess the frequency and total numbers of regulatory T (Treg) cells in vivo. RESULTS: Spermidine modulates CD4+ T-cell differentiation in vitro, preferentially committing naive T cells to a regulatory phenotype. After spermidine treatment, activated T cells lacking the autophagy gene Atg5 fail to upregulate Foxp3 to the same extent as wild-type cells. These results indicate that spermidine's polarizing effect requires an intact autophagic machinery. Furthermore, dietary supplementation with spermidine promotes homeostatic differentiation of Treg cells within the gut and reduces pathology in a model of T-cell transfer-induced colitis. CONCLUSION: Altogether, our results highlight the beneficial effects of spermidine, or l-arginine, on gut immunity by promoting Treg cell development.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Colite/imunologia , Imunidade nas Mucosas/efeitos dos fármacos , Espermidina/farmacologia , Linfócitos T Reguladores/imunologia , Animais , Diferenciação Celular/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout
3.
Circulation ; 139(15): 1798-1812, 2019 04 09.
Artigo em Inglês | MEDLINE | ID: mdl-30696265

RESUMO

BACKGROUND: Acute myocardial infarction (MI) elicits an inflammatory response that drives tissue repair and adverse cardiac remodeling. Inflammatory cell trafficking after MI is controlled by C-X-C motif chemokine ligand 12 (CXCL12) and its receptor, C-X-C motif chemokine receptor 4 (CXCR4). CXCR4 antagonists mobilize inflammatory cells and promote infarct repair, but the cellular mechanisms are unclear. METHODS: We investigated the therapeutic potential and mode of action of the peptidic macrocycle CXCR4 antagonist POL5551 in mice with reperfused MI. We applied cell depletion and adoptive transfer strategies using lymphocyte-deficient Rag1 knockout mice; DEREG mice, which express a diphtheria toxin receptor-enhanced green fluorescent protein fusion protein under the control of the promoter/enhancer region of the regulatory T (Treg) cell-restricted Foxp3 transcription factor; and dendritic cell-depleted CD11c-Cre iDTR mice. Translational potential was explored in a porcine model of reperfused MI using serial contrast-enhanced magnetic resonance imaging. RESULTS: Intraperitoneal POL5551 injections in wild-type mice (8 mg/kg at 2, 4, 6, and 8 days) enhanced angiogenesis in the infarct border zone, reduced scar size, and attenuated left ventricular remodeling and contractile dysfunction at 28 days. Treatment effects were absent in splenectomized wild-type mice, Rag1 knockout mice, and Treg cell-depleted DEREG mice. Conversely, treatment effects could be transferred into infarcted splenectomized wild-type mice by transplanting splenic Treg cells from POL5551-treated infarcted DEREG mice. Instructive cues provided by infarct-primed dendritic cells were required for POL5551 treatment effects. POL5551 injections mobilized Treg cells into the peripheral blood, followed by enhanced Treg cell accumulation in the infarcted region. Neutrophils, monocytes, and lymphocytes displayed similar mobilization kinetics, but their cardiac recruitment was not affected. POL5551, however, attenuated inflammatory gene expression in monocytes and macrophages in the infarcted region via Treg cells. Intravenous infusion of the clinical-stage POL5551 analogue POL6326 (3 mg/kg at 4, 6, 8, and 10 days) decreased infarct volume and improved left ventricular ejection fraction in pigs. CONCLUSIONS: These data confirm CXCR4 blockade as a promising treatment strategy after MI. We identify dendritic cell-primed splenic Treg cells as the central arbiters of these therapeutic effects and thereby delineate a pharmacological strategy to promote infarct repair by augmenting Treg cell function in vivo.


Assuntos
Anti-Inflamatórios/farmacologia , Infarto do Miocárdio/tratamento farmacológico , Miocárdio/metabolismo , Proteínas/farmacologia , Receptores CXCR4/antagonistas & inibidores , Linfócitos T Reguladores/efeitos dos fármacos , Animais , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Modelos Animais de Doenças , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Contração Miocárdica/efeitos dos fármacos , Infarto do Miocárdio/imunologia , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Miocárdio/imunologia , Miocárdio/patologia , Neovascularização Fisiológica/efeitos dos fármacos , Receptores CXCR4/metabolismo , Recuperação de Função Fisiológica , Transdução de Sinais , Sus scrofa , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Função Ventricular Esquerda/efeitos dos fármacos , Remodelação Ventricular/efeitos dos fármacos
4.
Eur J Immunol ; 49(5): 747-757, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30802297

RESUMO

Infection with Clostridium difficile is one of the major causes of health care acquired diarrhea and colitis. Signaling though MyD88 downstream of TLRs is critical for initiating the early protective host response in mouse models of C. difficile infection (CDI). In the intestine, MyD88 is expressed in various tissues and cell types, such as the intestinal epithelium and mononuclear phagocytes (MNP), including DC or macrophages. Using a genetic gain-of-function system, we demonstrate here that restricting functional MyD88 signaling to the intestinal epithelium, but also to MNPs is sufficient to protect mice during acute CDI by upregulation of the intestinal barrier function and recruitment of neutrophils. Nevertheless, we also show that mice depleted for CD11c-expressing MNPs in the intestine display no major defects in mounting an effective inflammatory response, indicating that the absence of these cells is irrelevant for inducing host protection during acute infection. Together, our results highlight the importance of epithelial-specific MyD88 signaling and demonstrate that although functional MyD88 signaling in DC and macrophages alone is sufficient to correct the phenotype of MyD88-deficiency, these cells do not seem to be essential for host protection in MyD88-sufficient animals during acute infection with C. difficile.


Assuntos
Clostridioides difficile/imunologia , Enterocolite Pseudomembranosa/imunologia , Enterocolite Pseudomembranosa/metabolismo , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Fator 88 de Diferenciação Mieloide/metabolismo , Transdução de Sinais , Animais , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Modelos Animais de Doenças , Enterocolite Pseudomembranosa/microbiologia , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Mucosa Intestinal/patologia , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos
5.
Semin Immunol ; 28(5): 514-524, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27825556

RESUMO

Recent advances in the field of immunometabolism support the concept that fundamental processes in T cell biology, such as TCR-mediated activation and T helper lineage differentiation, are closely linked to changes in the cellular metabolic programs. Although the major task of the intermediate metabolism is to provide the cell with a constant supply of energy and molecular precursors for the production of biomolecules, the dynamic regulation of metabolic pathways also plays an active role in shaping T cell responses. Key metabolic processes such as glycolysis, fatty acid and mitochondrial metabolism are now recognized as crucial players in T cell activation and differentiation, and their modulation can differentially affect the development of T helper cell lineages. In this review, we describe the diverse metabolic processes that T cells engage during their life cycle from naïve towards effector and memory T cells. We consider in particular how the cellular metabolism may actively support the function of T cells in their different states. Moreover, we discuss how molecular regulators such as mTOR or AMPK link environmental changes to adaptations in the cellular metabolism and elucidate the consequences on T cell differentiation and function.


Assuntos
Diferenciação Celular , Metabolismo Energético , Ativação Linfocitária/imunologia , Redes e Vias Metabólicas , Subpopulações de Linfócitos T/citologia , Subpopulações de Linfócitos T/fisiologia , Animais , Ácidos Graxos/metabolismo , Glicólise , Humanos , Memória Imunológica , Mitocôndrias/metabolismo , Fenótipo , Transdução de Sinais
6.
PLoS Pathog ; 13(5): e1006357, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28520792

RESUMO

MyD88-mediated signaling downstream of Toll-like receptors and the IL-1 receptor family is critically involved in the induction of protective host responses upon infections. Although it is known that MyD88-deficient mice are highly susceptible to a wide range of bacterial infections, the cell type-specific contribution of MyD88 in protecting the host against intestinal bacterial infection is only poorly understood. In order to investigate the importance of MyD88 in specific immune and nonimmune cell types during intestinal infection, we employed a novel murine knock-in model for MyD88 that enables the cell type-specific reactivation of functional MyD88 expression in otherwise MyD88-deficient mice. We report here that functional MyD88 signaling in CD11c+ cells was sufficient to activate intestinal dendritic cells (DC) and to induce the early group 3 innate lymphoid cell (ILC3) response as well as the development of colonic Th17/Th1 cells in response to infection with the intestinal pathogen C. rodentium. In contrast, restricting MyD88 signaling to several other cell types, including macrophages (MO), T cells or ILC3 did not induce efficient intestinal immune responses upon infection. However, we observed that the functional expression of MyD88 in intestinal epithelial cells (IEC) also partially protected the mice during intestinal infection, which was associated with enhanced epithelial barrier integrity and increased expression of the antimicrobial peptide RegIIIγ and the acute phase protein SAA1 by epithelial cells. Together, our data suggest that MyD88 signaling in DC and IEC is both essential and sufficient to induce a full spectrum of host responses upon intestinal infection with C. rodentium.


Assuntos
Citrobacter rodentium/imunologia , Infecções por Enterobacteriaceae/imunologia , Fator 88 de Diferenciação Mieloide/metabolismo , Transdução de Sinais , Animais , Colo/imunologia , Colo/microbiologia , Células Dendríticas/imunologia , Células Dendríticas/microbiologia , Infecções por Enterobacteriaceae/microbiologia , Infecções por Enterobacteriaceae/patologia , Células Epiteliais/imunologia , Células Epiteliais/microbiologia , Técnicas de Introdução de Genes , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Macrófagos/imunologia , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Fator 88 de Diferenciação Mieloide/genética , Receptores de Interleucina-1/metabolismo , Células Th1/imunologia , Células Th1/microbiologia , Células Th17/imunologia , Células Th17/microbiologia , Receptores Toll-Like/genética , Receptores Toll-Like/metabolismo
7.
PLoS Pathog ; 13(6): e1006454, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28628650

RESUMO

Human Cytomegalovirus (HCMV) is a widespread pathogen, infection with which can cause severe disease for immunocompromised individuals. The complex changes wrought on the host's immune system during both productive and latent HCMV infection are well known. Infected cells are masked and manipulated and uninfected immune cells are also affected; peripheral blood mononuclear cell (PBMC) proliferation is reduced and cytokine profiles altered. Levels increase of the anti-inflammatory cytokine IL-10, which may be important for the establishment of HCMV infections and is required for the development of high viral titres by murine cytomegalovirus. The mechanisms by which HCMV affects T cell IL-10 secretion are not understood. We show here that treatment of PBMC with purified pUL11 induces IL-10 producing T cells as a result of pUL11 binding to the CD45 phosphatase on T cells. IL-10 production induced by HCMV infection is also in part mediated by pUL11. Supernatants from pUL11 treated cells have anti-inflammatory effects on untreated PBMC. Considering the mechanism, CD45 can be a positive or negative regulator of TCR signalling, depending on its expression level, and we show that pUL11 also has concentration dependent activating or inhibitory effects on T cell proliferation and on the kinase function of the CD45 substrate Lck. pUL11 is therefore the first example of a viral protein that can target CD45 to induce T cells with anti-inflammatory properties. It is also the first HCMV protein shown to induce T cell IL-10 secretion. Understanding the mechanisms by which pUL11-induced changes in signal strength influence T cell development and function may provide the basis for the development of novel antiviral treatments and therapies against immune pathologies.


Assuntos
Linfócitos T CD4-Positivos/metabolismo , Infecções por Citomegalovirus/metabolismo , Citomegalovirus/metabolismo , Glicoproteínas/metabolismo , Interleucina-10/metabolismo , Antígenos Comuns de Leucócito/metabolismo , Proteínas Virais/metabolismo , Células Cultivadas , Citomegalovirus/genética , Infecções por Citomegalovirus/genética , Infecções por Citomegalovirus/virologia , Glicoproteínas/genética , Humanos , Interleucina-10/genética , Antígenos Comuns de Leucócito/genética , Leucócitos Mononucleares/metabolismo , Proteínas Virais/genética
8.
Trends Immunol ; 36(2): 81-91, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25592731

RESUMO

The specific regulation of cellular metabolic processes is of major importance for directing immune cell differentiation and function. We review recent evidence indicating that changes in basic cellular lipid metabolism have critical effects on T cell proliferation and cell fate decisions. While induction of de novo fatty acid (FA) synthesis is essential for activation-induced proliferation and differentiation of effector T cells, FA catabolism via ß-oxidation is important for the development of CD8(+) T cell memory as well as for the differentiation of CD4(+) regulatory T cells. We consider the influence of lipid metabolism and metabolic intermediates on the regulation of signaling and transcriptional pathways via post-translational modifications, and discuss how an improved understanding of FA metabolism may reveal strategies for manipulating immune responses towards therapeutic outcomes.


Assuntos
Ácidos Graxos/metabolismo , Imunomodulação , Linfócitos T/imunologia , Linfócitos T/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Humanos , Memória Imunológica , Lipídeos/biossíntese , Ativação Linfocitária , Oxirredução , Processamento de Proteína Pós-Traducional , Transdução de Sinais , Proteínas de Ligação a Elemento Regulador de Esterol/metabolismo , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Serina-Treonina Quinases TOR/metabolismo
9.
Eur J Inorg Chem ; 2018(23): 2689-2694, 2018 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-30034271

RESUMO

A multicomponent approach of the P n ligand complex [Cp*Fe(η5-P5)] (1: Cp* = η5-C5Me5) with the ditopic organic linkers 4,4'-bipyridine (2) or trans-1,2-di(pyridine-4-yl)ethene (3) in the presence of CuI salts of the anions [BF4]- and [PF6]- or the coordinating anion Br-, leads to the formation of four novel organometallic-organic hybrid polymers: the cationic 1D polymeric compounds [Cu4{Cp*Fe(µ3,η5:1:1-P5)}2(µ,η1:1-C10H8N2)4(CH3CN)4] n [BF4]4n (4) and [Cu4{Cp*Fe(µ3,η5:1:1-P5)}2(µ,η1:1-C10H8N2)4(CH3CN)4] n [PF6]4n (5) as well as the unique neutral threefold 2D → 2D interpenetrated networks [Cu2Cl2{Cp*Fe(µ3,η5:1:1-P5)}(µ,η1:1-C12H10N2)] n (6) and [Cu2Br2{Cp*Fe(µ3,η5:1:1-P5)}(µ,η1:1-C10H8N2)] n (7).

10.
Eur J Immunol ; 46(9): 2233-8, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27338930

RESUMO

Upon antigen-specific or allogeneic activation, T cells sharply increase their metabolic activity to cope with augmented needs for proliferation and effector functions. Therefore, enzymes involved in energy metabolism constitute attractive targets to modulate the activity of pathogenic effector T cells in the setting of graft-versus-host-disease (GVHD). Here, we show that T cells deficient for acetyl-CoA carboxylase 1 (TACC1) are dramatically less pathogenic than wild-type (WT) T cells in a lethal C57BL/6 into BALB/c model of acute GVHD and permitted sustained survival of recipient mice. In line with this clinical observation, higher frequencies of GVHD-suppressing Foxp3(+) regulatory T (Treg) cells were detected in the colon of TACC T-cell recipients. In vitro, T-cell stimulation with allogeneic DCs induced higher proportions of Treg cells but also led to diminished proliferation of TACC1 T cells compared to WT T cells. Furthermore, TACC1 T cells activated by allogeneic DCs showed impaired glycolysis and lipid synthesis. Thus, targeting de novo fatty acid synthesis via acetyl-CoA carboxylase inhibition may be a promising new strategy to prevent GVHD.


Assuntos
Acetil-CoA Carboxilase/antagonistas & inibidores , Ácidos Graxos/biossíntese , Doença Enxerto-Hospedeiro/etiologia , Doença Enxerto-Hospedeiro/prevenção & controle , Acetil-CoA Carboxilase/deficiência , Transferência Adotiva , Animais , Biomarcadores , Transplante de Medula Óssea , Diferenciação Celular , Modelos Animais de Doenças , Deleção de Genes , Doença Enxerto-Hospedeiro/mortalidade , Imunofenotipagem , Macrolídeos/farmacologia , Masculino , Camundongos , Fenótipo , Linfócitos T/citologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Transplante Homólogo
11.
Immunity ; 29(6): 958-70, 2008 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-19084435

RESUMO

Natural killer (NK) cells are innate lymphocytes with spontaneous antitumor activity, and they produce interferon-gamma (IFN-gamma) that primes immune responses. Whereas T helper cell subsets differentiate from naive T cells via specific transcription factors, evidence for NK cell diversification is limited. In this report, we characterized intestinal lymphocytes expressing the NK cell natural cytotoxicity receptor NKp46. Gut NKp46+ cells were distinguished from classical NK cells by limited IFN-gamma production and absence of perforin, whereas several subsets expressed the nuclear hormone receptor retinoic acid receptor-related orphan receptor t (RORgammat) and interleukin-22 (IL-22). Intestinal NKp46+IL-22+ cells were generated via a local process that was conditioned by commensal bacteria and required RORgammat. Mice lacking IL-22-producing NKp46+ cells showed heightened susceptibility to the pathogen Citrobacter rodentium, consistent with a role for intestinal NKp46+ cells in immune protection. RORgammat-driven diversification of intestinal NKp46+ cells thereby specifies an innate cellular defense mechanism that operates at mucosal surfaces.


Assuntos
Antígenos Ly/imunologia , Citrobacter rodentium/imunologia , Infecções por Enterobacteriaceae/imunologia , Interleucinas/imunologia , Intestinos/imunologia , Células Matadoras Naturais/imunologia , Receptor 1 Desencadeador da Citotoxicidade Natural/imunologia , Animais , Antígenos Ly/metabolismo , Infecções por Enterobacteriaceae/microbiologia , Imunidade Inata , Imunidade nas Mucosas/imunologia , Interleucinas/metabolismo , Mucosa Intestinal/metabolismo , Intestinos/microbiologia , Células Matadoras Naturais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor 1 Desencadeador da Citotoxicidade Natural/metabolismo , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares , Perforina/imunologia , Perforina/metabolismo , Receptores do Ácido Retinoico/imunologia , Receptores do Ácido Retinoico/metabolismo , Receptores dos Hormônios Tireóideos/imunologia , Receptores dos Hormônios Tireóideos/metabolismo , Transdução de Sinais/imunologia , Interleucina 22
12.
J Immunol ; 194(6): 2888-98, 2015 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-25694610

RESUMO

IL-17-producing Th17 cells mediate immune responses against a variety of fungal and bacterial infections. Signaling via NF-κB has been linked to the development and maintenance of Th17 cells. We analyzed the role of the unusual inhibitor of NF-κB, IκBNS, in the proliferation and effector cytokine production of murine Th17 cells. Our study demonstrates that nuclear IκBNS is crucial for murine Th17 cell generation. IκBNS is highly expressed in Th17 cells; in the absence of IκBNS, the frequencies of IL-17A-producing cells are drastically reduced. This was measured in vitro under Th17-polarizing conditions and confirmed in two colitis models. Mechanistically, murine IκBNS (-/-) Th17 cells were less proliferative and expressed markedly reduced levels of IL-2, IL-10, MIP-1α, and GM-CSF. Citrobacter rodentium was used as a Th17-inducing infection model, in which IκBNS (-/-) mice displayed an increased bacterial burden and diminished tissue damage. These results demonstrate the important function of Th17 cells in pathogen clearance, as well as in inflammation-associated pathology. We identified IκBNS to be crucial for the generation and function of murine Th17 cells upon inflammation and infection. Our findings may have implications for the therapy of autoimmune conditions, such as inflammatory bowel disease, and for the treatment of gut-tropic infections.


Assuntos
Diferenciação Celular/imunologia , Citrobacter rodentium/imunologia , Colite/imunologia , Infecções por Enterobacteriaceae/imunologia , Proteínas I-kappa B/imunologia , Células Th17/imunologia , Animais , Western Blotting , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Diferenciação Celular/genética , Proliferação de Células/genética , Citrobacter rodentium/fisiologia , Colite/genética , Colite/metabolismo , Citocinas/genética , Citocinas/imunologia , Citocinas/metabolismo , Infecções por Enterobacteriaceae/genética , Infecções por Enterobacteriaceae/microbiologia , Citometria de Fluxo , Expressão Gênica/imunologia , Interações Hospedeiro-Patógeno/imunologia , Proteínas I-kappa B/deficiência , Proteínas I-kappa B/genética , Camundongos da Linhagem 129 , Camundongos Knockout , NF-kappa B/imunologia , NF-kappa B/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Th17/metabolismo
13.
Nucleic Acids Res ; 43(3): 1537-48, 2015 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-25593324

RESUMO

Activated naive CD4(+) T cells are highly plastic cells that can differentiate into various T helper (Th) cell fates characterized by the expression of effector cytokines like IFN-γ (Th1), IL-4 (Th2) or IL-17A (Th17). Although previous studies have demonstrated that epigenetic mechanisms including DNA demethylation can stabilize effector cytokine expression, a comprehensive analysis of the changes in the DNA methylation pattern during differentiation of naive T cells into Th cell subsets is lacking. Hence, we here performed a genome-wide methylome analysis of ex vivo isolated naive CD4(+) T cells, Th1 and Th17 cells. We could demonstrate that naive CD4(+) T cells share more demethylated regions with Th17 cells when compared to Th1 cells, and that overall Th17 cells display the highest number of demethylated regions, findings which are in line with the previously reported plasticity of Th17 cells. We could identify seven regions located in Il17a, Zfp362, Ccr6, Acsbg1, Dpp4, Rora and Dclk1 showing pronounced demethylation selectively in ex vivo isolated Th17 cells when compared to other ex vivo isolated Th cell subsets and in vitro generated Th17 cells, suggesting that this unique epigenetic signature allows identifying and functionally characterizing in vivo generated Th17 cells.


Assuntos
Diferenciação Celular/genética , Epigênese Genética , Células Th17/citologia , Animais , Metilação de DNA , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Análise de Sequência com Séries de Oligonucleotídeos
14.
J Immunol ; 186(3): 1531-7, 2011 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-21178008

RESUMO

Retinoic acid-related orphan receptor (ROR)γt(+) TCRαß(+) cells expressing IL-17, termed Th17 cells, are most abundant in the intestinal lamina propria. Symbiotic microbiota are required for the generation of Th17 cells, but the requirement for microbiota-derived Ag is not documented. In this study, we show that normal numbers of Th17 cells develop in the intestine of mice that express a single TCR in the absence of cognate Ag, whereas the microbiota remains essential for their development. However, such mice, or mice monocolonized with the Th17-inducing segmented filamentous bacteria, fail to induce normal numbers of Foxp3(+) RORγt(+) T cells, the regulatory counterpart of IL-17(+)RORγt(+) T cells. These results demonstrate that a complex microbiota and cognate Ag are required to generate a properly regulated set of RORγt(+) T cells and Th17 cells.


Assuntos
Proliferação de Células , Interleucina-17/biossíntese , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/deficiência , Receptores de Antígenos de Linfócitos T/deficiência , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Auxiliares-Indutores/microbiologia , Sequência de Aminoácidos , Animais , Contagem de Linfócito CD4 , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Feminino , Fatores de Transcrição Forkhead/biossíntese , Fatores de Transcrição Forkhead/deficiência , Fatores de Transcrição Forkhead/genética , Vida Livre de Germes/genética , Vida Livre de Germes/imunologia , Infecções por Bactérias Gram-Positivas/imunologia , Infecções por Bactérias Gram-Positivas/patologia , Proteínas de Fluorescência Verde/biossíntese , Proteínas de Fluorescência Verde/genética , Interleucina-17/genética , Mucosa Intestinal/patologia , Ativação Linfocitária/genética , Ativação Linfocitária/imunologia , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Dados de Sequência Molecular , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/biossíntese , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/genética , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/fisiologia , Linfócitos T Auxiliares-Indutores/patologia
15.
Eur J Immunol ; 41(2): 473-84, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21268016

RESUMO

Rejection of solid organ allograft involves alloreactive T-cell expansion. The importance of NF-κB and NFAT in this process is underscored by the therapeutic efficacy of immunosuppressive agents, which target the two transcription factors. Since calpains, calcium-activated proteases, are involved in the activation of NF-κB and NFAT, we investigated the role of calpains in allograft rejection. In human transplant kidneys undergoing acute or chronic rejection, we show an increased expression of CAPN 1 gene encoding µ-calpain, associated with a marked expression of µ-calpain, mainly in infiltrating T cells. To address the role of calpain in rejection, we used a skin transplant model in transgenic mice expressing high levels of calpastatin, a calpain-specific inhibitor. We show that calpain inhibition extended skin allograft survival, from 11 to 20 days. This delay was associated with a limitation in allograft infiltration by T cells. In vitro, calpain inhibition by calpastatin transgene expression limited dramatically T-cell migration but, unexpectedly, increased slightly T-cell proliferation. Amplification of IL-2 signaling via the stabilization of IL-2R common γ-chain provided an explanation for the proliferation response. This is the first study establishing that calpain inhibition delays allograft rejection by slowing down T-cell migration rather than proliferation.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Calpaína/metabolismo , Rejeição de Enxerto/metabolismo , Acrilatos/uso terapêutico , Transferência Adotiva , Animais , Proteínas de Ligação ao Cálcio/genética , Calpaína/antagonistas & inibidores , Calpaína/genética , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Movimento Celular/genética , Movimento Celular/imunologia , Proliferação de Células/efeitos dos fármacos , Expressão Gênica/genética , Expressão Gênica/imunologia , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/patologia , Rejeição de Enxerto/prevenção & controle , Proteínas de Homeodomínio/genética , Humanos , Interleucina-2/metabolismo , Rim/imunologia , Rim/metabolismo , Transplante de Rim/imunologia , Ativação Linfocitária/genética , Ativação Linfocitária/imunologia , Teste de Cultura Mista de Linfócitos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , NF-kappa B/metabolismo , Fatores de Transcrição NFATC/metabolismo , Transplante de Pele/imunologia , Transplante de Pele/patologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Linfócitos T/patologia , Linfócitos T/transplante , Linfócitos T Citotóxicos/imunologia , Células Th17/imunologia , Células Th17/metabolismo , Transplante Homólogo
16.
Cell Microbiol ; 13(5): 653-9, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21338464

RESUMO

The mammalian intestine provides a unique niche for a large community of bacterial symbionts that complements the host in digestive and anabolic pathways, as well as in protection from pathogens. Only a few bacterial phyla have adapted to this predominantly anaerobic environment, but hundreds of different species create an ecosystem that affects many facets of the host's physiology. Recent data show how particular symbionts are involved in the maturation of the immune system, in the intestine and beyond, and how dysbiosis, or alteration of that community, can deregulate immunity and lead to immunopathology. The extensive and dynamic interactions between the symbionts and the immune system are key to homeostasis and health, and require all the blends of so-called regulatory and pro-inflammatory immune reactions. Unfortunately, pro-inflammatory immunity leading to the generation of Th17 cells has been mainly associated with its role in immunopathology. Here we discuss the view that the immune system in general, and type 17 immunity in particular, develop to maintain the equilibrium of the host with its symbionts.


Assuntos
Inflamação/imunologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Animais , Homeostase , Humanos , Simbiose , Células Th17/imunologia
17.
Nat Commun ; 13(1): 3998, 2022 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-35810180

RESUMO

Basic processes of the fatty acid metabolism have an important impact on the function of intestinal epithelial cells (IEC). However, while the role of cellular fatty acid oxidation is well appreciated, it is not clear how de novo fatty acid synthesis (FAS) influences the biology of IECs. We report here that interfering with de novo FAS by deletion of the enzyme Acetyl-CoA-Carboxylase (ACC)1 in IECs results in the loss of epithelial crypt structures and a specific decline in Lgr5+ intestinal epithelial stem cells (ISC). Mechanistically, ACC1-mediated de novo FAS supports the formation of intestinal organoids and the differentiation of complex crypt structures by sustaining the nuclear accumulation of PPARδ/ß-catenin in ISCs. The dependency of ISCs on cellular de novo FAS is tuned by the availability of environmental lipids, as an excess delivery of external fatty acids is sufficient to rescue the defect in crypt formation. Finally, inhibition of ACC1 reduces the formation of tumors in colitis-associated colon cancer, together highlighting the importance of cellular lipogenesis for sustaining ISC function and providing a potential perspective to colon cancer therapy.


Assuntos
Acetil-CoA Carboxilase , Lipogênese , Acetilcoenzima A/metabolismo , Acetil-CoA Carboxilase/metabolismo , Ácidos Graxos/metabolismo , Lipogênese/fisiologia , Células-Tronco/metabolismo
18.
J Exp Med ; 219(10)2022 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-35938981

RESUMO

Epigenetic modifications such as DNA methylation play an essential role in imprinting specific transcriptional patterns in cells. We performed genome-wide DNA methylation profiling of murine lymph node-derived ILCs, which led to the identification of differentially methylated regions (DMRs) and the definition of epigenetic marker regions in ILCs. Marker regions were located in genes with a described function for ILCs, such as Tbx21, Gata3, or Il23r, but also in genes that have not been related to ILC biology. Methylation levels of the marker regions and expression of the associated genes were strongly correlated, indicating their functional relevance. Comparison with T helper cell methylomes revealed clear lineage differences, despite partial similarities in the methylation of specific ILC marker regions. IL-33-mediated challenge affected methylation of ILC2 epigenetic marker regions in the liver, while remaining relatively stable in the lung. In our study, we identified a set of epigenetic markers that can serve as a tool to study phenotypic and functional properties of ILCs.


Assuntos
Imunidade Inata , Linfócitos , Animais , Biomarcadores , Metilação de DNA/genética , Epigênese Genética , Estudo de Associação Genômica Ampla , Camundongos
19.
J Immunol ; 182(9): 5789-99, 2009 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-19380827

RESUMO

Stromal cells in lymphoid tissues regulate lymphocyte recruitment and survival through the expression of specific chemokines and cytokines. During inflammation, the same signals recruit lymphocytes to the site of injury; however, the "lymphoid" stromal (LS) cells producing these signals remain poorly characterized. We find that mouse inflammatory lesions and tumors develop gp38(+) LS cells, in recapitulation of the development of LS cells early during the ontogeny of lymphoid organs and the intestine, and express a set of genes that promotes the development of lymphocyte-permissive tissues. These gp38(+) LS cells are induced by a robust pathway that requires myeloid cells but not known Toll- or NOD-like receptors, the inflammasome, or adaptive immunity. Parabiosis and inducible genetic cell fate mapping experiments indicate that local precursors, presumably resident fibroblasts rather that circulating precursors, massively proliferate and give rise to LS cells during inflammation. Our results show that LS cells are both programmed during ontogeny and reinduced during inflammation.


Assuntos
Movimento Celular/imunologia , Inflamação/imunologia , Inflamação/patologia , Tecido Linfoide/imunologia , Tecido Linfoide/patologia , Animais , Quimiocinas/biossíntese , Citocinas/biossíntese , Inflamação/embriologia , Peptídeos e Proteínas de Sinalização Intercelular/biossíntese , Mucosa Intestinal/embriologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/patologia , Leishmania major/imunologia , Leishmaniose Cutânea/imunologia , Leishmaniose Cutânea/patologia , Tecido Linfoide/embriologia , Glicoproteínas de Membrana/biossíntese , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Nus , Camundongos Transgênicos , Células Estromais/imunologia , Células Estromais/metabolismo , Células Estromais/patologia
20.
Proc Natl Acad Sci U S A ; 105(50): 19845-50, 2008 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-19057011

RESUMO

Invariant natural killer T (iNKT) cells constitute a subpopulation of T cells that recognize glycolipids presented by CD1d molecules. They are characterized by their prompt production of interleukin-4 (IL-4) and interferon-gamma (IFN-gamma), which enables them to modulate diverse immune responses. Recently, we enlarged this concept by identifying a distinct IL-17-producing iNKT cell subset, named iNKT17 cells. The mechanisms leading to the acquisition of this new iNKT cell activity are unknown. Herein we show that IL-17-producing iNKT cells are already present in the thymus, predominantly among a subset regarded so far as an immature stage of thymic iNKT cell development, the CD1d tetramer(pos)CD44(pos)NK1.1(neg)CD4(neg) cells. Using EGFP reporter mice, we demonstrate that the transcription factor ROR-gammat is critical for the thymic differentiation of this subset because only ROR-gammat(pos) iNKT cells are capable of massively secreting IL-17. Moreover, IL-17-producing CD1d tetramer(pos)CD44(pos)NK1.1(neg)CD4(neg) thymic iNKT cells have reached a mature differentiation stage because they fail to generate other cell subsets in fetal thymic organ culture. Conversely, thymic ROR-gammat(neg) iNKT cell precursors give rise to progeny, but acquire neither ROR-gammat expression nor the ability to secrete IL-17. In conclusion, our findings demonstrate an alternative thymic pathway leading to the development of iNKT17 cells that requires ROR-gammat expression.


Assuntos
Interleucina-17/metabolismo , Ativação Linfocitária , Células T Matadoras Naturais/imunologia , Receptores do Ácido Retinoico/fisiologia , Receptores dos Hormônios Tireóideos/fisiologia , Timo/imunologia , Animais , Diferenciação Celular/genética , Galactosilceramidas/imunologia , Proteínas de Fluorescência Verde/genética , Ativação Linfocitária/genética , Masculino , Camundongos , Camundongos Transgênicos , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares , Receptores do Ácido Retinoico/genética , Receptores dos Hormônios Tireóideos/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA