Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Nat Immunol ; 16(12): 1228-34, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26523867

RESUMEN

The molecular mechanisms that link the sympathetic stress response and inflammation remain obscure. Here we found that the transcription factor Nr4a1 regulated the production of norepinephrine (NE) in macrophages and thereby limited experimental autoimmune encephalomyelitis (EAE), a mouse model of multiple sclerosis. Lack of Nr4a1 in myeloid cells led to enhanced NE production, accelerated infiltration of leukocytes into the central nervous system (CNS) and disease exacerbation in vivo. In contrast, myeloid-specific deletion of tyrosine hydroxylase (TH), the rate-limiting enzyme in catecholamine biosynthesis, protected mice against EAE. Furthermore, we found that Nr4a1 repressed autocrine NE production in macrophages by recruiting the corepressor CoREST to the Th promoter. Our data reveal a new role for macrophages in neuroinflammation and identify Nr4a1 as a key regulator of catecholamine production by macrophages.


Asunto(s)
Sistema Nervioso Central/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Inflamación/inmunología , Macrófagos/inmunología , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/inmunología , Sistema Nervioso Simpático/inmunología , Animales , Línea Celular , Células Cultivadas , Sistema Nervioso Central/metabolismo , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/metabolismo , Expresión Génica/inmunología , Humanos , Inflamación/genética , Inflamación/metabolismo , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Confocal , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/patología , Células Mieloides/inmunología , Células Mieloides/metabolismo , Norepinefrina/inmunología , Norepinefrina/metabolismo , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Conejos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sistema Nervioso Simpático/metabolismo , Tirosina 3-Monooxigenasa/genética , Tirosina 3-Monooxigenasa/inmunología , Tirosina 3-Monooxigenasa/metabolismo
3.
Immunity ; 34(4): 479-91, 2011 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-21458306

RESUMEN

The transcription factor FOXP3 is essential for the suppressive function of regulatory T cells that are required for maintaining self-tolerance. We have solved the crystal structure of the FOXP3 forkhead domain as a ternary complex with the DNA-binding domain of the transcription factor NFAT1 and a DNA oligonucleotide from the interleukin-2 promoter. A striking feature of this structure is that FOXP3 forms a domain-swapped dimer that bridges two molecules of DNA. Structure-guided or autoimmune disease (IPEX)-associated mutations in the domain-swap interface diminished dimer formation by the FOXP3 forkhead domain without compromising FOXP3 DNA binding. These mutations also eliminated T cell-suppressive activity conferred by FOXP3, both in vitro and in a murine model of autoimmune diabetes in vivo. We conclude that FOXP3-mediated suppressor function requires dimerization through the forkhead domain and that mutations in the dimer interface can lead to the systemic autoimmunity observed in IPEX patients.


Asunto(s)
Factores de Transcripción Forkhead/química , Factores de Transcripción Forkhead/inmunología , Linfocitos T Reguladores/química , Linfocitos T Reguladores/inmunología , Secuencia de Aminoácidos , Animales , Humanos , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Mutación , Factores de Transcripción NFATC/química , Factores de Transcripción NFATC/inmunología , Multimerización de Proteína , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína , Alineación de Secuencia
4.
Nature ; 497(7447): 122-6, 2013 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-23563267

RESUMEN

TET (ten-eleven-translocation) proteins are Fe(ii)- and α-ketoglutarate-dependent dioxygenases that modify the methylation status of DNA by successively oxidizing 5-methylcytosine to 5-hydroxymethylcytosine, 5-formylcytosine and 5-carboxycytosine, potential intermediates in the active erasure of DNA-methylation marks. Here we show that IDAX (also known as CXXC4), a reported inhibitor of Wnt signalling that has been implicated in malignant renal cell carcinoma and colonic villous adenoma, regulates TET2 protein expression. IDAX was originally encoded within an ancestral TET2 gene that underwent a chromosomal gene inversion during evolution, thus separating the TET2 CXXC domain from the catalytic domain. The IDAX CXXC domain binds DNA sequences containing unmethylated CpG dinucleotides, localizes to promoters and CpG islands in genomic DNA and interacts directly with the catalytic domain of TET2. Unexpectedly, IDAX expression results in caspase activation and TET2 protein downregulation, in a manner that depends on DNA binding through the IDAX CXXC domain, suggesting that IDAX recruits TET2 to DNA before degradation. IDAX depletion prevents TET2 downregulation in differentiating mouse embryonic stem cells, and short hairpin RNA against IDAX increases TET2 protein expression in the human monocytic cell line U937. Notably, we find that the expression and activity of TET3 is also regulated through its CXXC domain. Taken together, these results establish the separate and linked CXXC domains of TET2 and TET3, respectively, as previously unknown regulators of caspase activation and TET enzymatic activity.


Asunto(s)
5-Metilcitosina/metabolismo , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Factores de Transcripción/química , Factores de Transcripción/metabolismo , Animales , Secuencia de Bases , Caspasas/metabolismo , Dominio Catalítico , Islas de CpG/genética , Metilación de ADN/genética , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Dioxigenasas/química , Dioxigenasas/genética , Dioxigenasas/metabolismo , Regulación hacia Abajo , Células Madre Embrionarias/metabolismo , Activación Enzimática , Células HEK293 , Humanos , Ratones , Oxidación-Reducción , Regiones Promotoras Genéticas/genética , Dominios y Motivos de Interacción de Proteínas , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Proto-Oncogénicas/química , Proteínas Proto-Oncogénicas/genética , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Células U937
6.
J Immunol ; 195(7): 3180-9, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26324768

RESUMEN

Regulatory T cell (Treg) activity is modulated by a cooperative complex between the transcription factor NFAT and FOXP3, a lineage specification factor for Tregs. FOXP3/NFAT interaction is required to repress expression of IL-2, upregulate expression of the Treg markers CTLA4 and CD25, and confer suppressor function to Tregs. However, FOXP3 is expressed transiently in conventional CD4(+) T cells upon TCR stimulation and may lead to T cell hyporesponsiveness. We found that a short synthetic peptide able to inhibit FOXP3/NFAT interaction impaired suppressor activity of conventional Tregs in vitro. Specific inhibition of FOXP3/NFAT interaction with this inhibitory peptide revealed that FOXP3 downregulates NFAT-driven promoter activity of CD40L and IL-17. Inhibition of FOXP3/NFAT interaction upregulated CD40L expression on effector T cells and enhanced T cell proliferation and IL-2, IFN-γ, IL-6, or IL-17 production in response to TCR stimulation. The inhibitory peptide impaired effector T cell conversion into induced Tregs in the presence of TGF-ß. Moreover, in vivo peptide administration showed antitumor efficacy in mice bearing Hepa129 or TC1 tumor cells when combined with sorafenib or with an antitumor vaccine, respectively. Our results suggest that inhibition of NFAT/FOXP3 interaction might improve antitumor immunotherapies.


Asunto(s)
Ligando de CD40/biosíntesis , Factores de Transcripción Forkhead/metabolismo , Interleucina-17/biosíntesis , Factores de Transcripción NFATC/metabolismo , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Animales , Antineoplásicos/farmacología , Ligando de CD40/genética , Antígeno CTLA-4/biosíntesis , Proliferación Celular/genética , Femenino , Factores de Transcripción Forkhead/antagonistas & inhibidores , Humanos , Inmunoterapia , Interferón gamma/biosíntesis , Interleucina-17/genética , Interleucina-2/biosíntesis , Subunidad alfa del Receptor de Interleucina-2/biosíntesis , Interleucina-6/biosíntesis , Células Jurkat , Activación de Linfocitos/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Transgénicos , Factores de Transcripción NFATC/antagonistas & inhibidores , Neoplasias/terapia , Niacinamida/análogos & derivados , Niacinamida/farmacología , Ovalbúmina/inmunología , Fragmentos de Péptidos/farmacología , Compuestos de Fenilurea/farmacología , Regiones Promotoras Genéticas/genética , Sorafenib , Factor de Crecimiento Transformador beta/metabolismo
7.
Nature ; 468(7325): 839-43, 2010 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-21057493

RESUMEN

TET2 is a close relative of TET1, an enzyme that converts 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC) in DNA. The gene encoding TET2 resides at chromosome 4q24, in a region showing recurrent microdeletions and copy-neutral loss of heterozygosity (CN-LOH) in patients with diverse myeloid malignancies. Somatic TET2 mutations are frequently observed in myelodysplastic syndromes (MDS), myeloproliferative neoplasms (MPN), MDS/MPN overlap syndromes including chronic myelomonocytic leukaemia (CMML), acute myeloid leukaemias (AML) and secondary AML (sAML). We show here that TET2 mutations associated with myeloid malignancies compromise catalytic activity. Bone marrow samples from patients with TET2 mutations displayed uniformly low levels of 5hmC in genomic DNA compared to bone marrow samples from healthy controls. Moreover, small hairpin RNA (shRNA)-mediated depletion of Tet2 in mouse haematopoietic precursors skewed their differentiation towards monocyte/macrophage lineages in culture. There was no significant difference in DNA methylation between bone marrow samples from patients with high 5hmC versus healthy controls, but samples from patients with low 5hmC showed hypomethylation relative to controls at the majority of differentially methylated CpG sites. Our results demonstrate that Tet2 is important for normal myelopoiesis, and suggest that disruption of TET2 enzymatic activity favours myeloid tumorigenesis. Measurement of 5hmC levels in myeloid malignancies may prove valuable as a diagnostic and prognostic tool, to tailor therapies and assess responses to anticancer drugs.


Asunto(s)
5-Metilcitosina/metabolismo , Proteínas de Unión al ADN/metabolismo , Hidroxilación , Leucemia Mieloide Aguda/metabolismo , Proteínas Mutantes/metabolismo , Síndromes Mielodisplásicos/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Animales , Biocatálisis , Diferenciación Celular , Línea Celular , Islas de CpG/genética , Metilación de ADN , ADN de Neoplasias/química , ADN de Neoplasias/metabolismo , Proteínas de Unión al ADN/genética , Dioxigenasas , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Ratones , Ratones Endogámicos C57BL , Proteínas Mutantes/genética , Mutación , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/patología , Proteínas Proto-Oncogénicas/genética
8.
Proc Natl Acad Sci U S A ; 110(46): 18608-13, 2013 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-24163352

RESUMEN

Acquisition of effector properties is a key step in the generation of cytotoxic T lymphocytes (CTLs). Here we show that inflammatory signals regulate Dicer expression in CTLs, and that deletion or depletion of Dicer in mouse or human activated CD8(+) T cells causes up-regulation of perforin, granzymes, and effector cytokines. Genome-wide analysis of microRNA (miR, miRNA) changes induced by exposure of differentiating CTLs to IL-2 and inflammatory signals identifies miR-139 and miR-150 as components of an miRNA network that controls perforin, eomesodermin, and IL-2Rα expression in differentiating CTLs and whose activity is modulated by IL-2, inflammation, and antigenic stimulation. Overall, our data show that strong IL-2R and inflammatory signals act through Dicer and miRNAs to control the cytolytic program and other aspects of effector CTL differentiation.


Asunto(s)
Diferenciación Celular/fisiología , Regulación de la Expresión Génica/inmunología , MicroARNs/metabolismo , Ribonucleasa III/metabolismo , Transducción de Señal/inmunología , Linfocitos T Citotóxicos/fisiología , Virosis/inmunología , Traslado Adoptivo , Animales , Western Blotting , Biología Computacional , Cartilla de ADN/genética , Granzimas/metabolismo , Humanos , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Ratones , Perforina/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Análisis de Secuencia de ADN
9.
Proc Natl Acad Sci U S A ; 109(36): 14532-7, 2012 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-22912406

RESUMEN

Bromodomain-containing proteins bind acetylated lysine residues on histone tails and are involved in the recruitment of additional factors that mediate histone modifications and enable transcription. A compound, I-BET-762, that inhibits binding of an acetylated histone peptide to proteins of the bromodomain and extra-terminal domain (BET) family, was previously shown to suppress the production of proinflammatory proteins by macrophages and block acute inflammation in mice. Here, we investigated the effect of short-term treatment with I-BET-762 on T-cell function. Treatment of naïve CD4(+) T cells with I-BET-762 during the first 2 d of differentiation had long-lasting effects on subsequent gene expression and cytokine production. Gene expression analysis revealed up-regulated expression of several antiinflammatory gene products, including IL-10, Lag3, and Egr2, and down-regulated expression of several proinflammatory cytokines including GM-CSF and IL-17. The short 2-d treatment with I-BET-762 inhibited the ability of antigen-specific T cells, differentiated under Th1 but not Th17 conditions in vitro, to induce pathogenesis in an adoptive transfer model of experimental autoimmune encephalomyelitis. The suppressive effects of I-BET-762 on T-cell mediated inflammation in vivo were accompanied by decreased recruitment of macrophages, consistent with decreased GM-CSF production by CNS-infiltrating T cells. These effects were mimicked by an inhibitor of c-myc function, implicating reduced expression of c-myc and GM-CSF as one avenue by which I-BET-762 suppresses the inflammatory functions of T cells. Our study demonstrates that inhibiting the functions of BET-family proteins during early T-cell differentiation causes long-lasting suppression of the proinflammatory functions of Th1 cells.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Citocinas/metabolismo , Encefalomielitis Autoinmune Experimental/inmunología , Regulación de la Expresión Génica/inmunología , Proteínas Nucleares/inmunología , alfa-Amilasas Salivales/antagonistas & inhibidores , Factores de Transcripción/inmunología , Transcripción Genética/inmunología , Traslado Adoptivo , Animales , Benzodiazepinas/farmacología , Linfocitos T CD4-Positivos/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/inmunología , Citometría de Flujo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Histonas/metabolismo , Ratones , Ratones Endogámicos C57BL , Análisis por Micromatrices , Proteínas Nucleares/metabolismo , Fosforilación , Factor B de Elongación Transcripcional Positiva/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tiazoles/farmacología , Factores de Transcripción/metabolismo
10.
Proc Natl Acad Sci U S A ; 108(35): 14566-71, 2011 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-21873190

RESUMEN

The Ten-Eleven-Translocation 2 (TET2) gene encodes a member of TET family enzymes that alters the epigenetic status of DNA by oxidizing 5-methylcytosine to 5-hydroxymethylcytosine (5hmC). Somatic loss-of-function mutations of TET2 are frequently observed in patients with diverse myeloid malignancies, including myelodysplastic syndromes, myeloproliferative neoplasms, and chronic myelomonocytic leukemia. By analyzing mice with targeted disruption of the Tet2 catalytic domain, we show here that Tet2 is a critical regulator of self-renewal and differentiation of hematopoietic stem cells (HSCs). Tet2 deficiency led to decreased genomic levels of 5hmC and augmented the size of the hematopoietic stem/progenitor cell pool in a cell-autonomous manner. In competitive transplantation assays, Tet2-deficient HSCs were capable of multilineage reconstitution and possessed a competitive advantage over wild-type HSCs, resulting in enhanced hematopoiesis into both lymphoid and myeloid lineages. In vitro, Tet2 deficiency delayed HSC differentiation and skewed development toward the monocyte/macrophage lineage. Our data indicate that Tet2 has a critical role in regulating the expansion and function of HSCs, presumably by controlling 5hmC levels at genes important for the self-renewal, proliferation, and differentiation of HSCs.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Células Madre Hematopoyéticas/citología , Homeostasis , Proteínas Proto-Oncogénicas/fisiología , Animales , Diferenciación Celular , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Dioxigenasas , Hematopoyesis , Ratones , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas/deficiencia , Proteínas Proto-Oncogénicas/genética
11.
Proc Natl Acad Sci U S A ; 108(28): 11381-6, 2011 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-21709260

RESUMEN

Nuclear factor of activated T cells (NFAT) proteins are Ca(2+)-regulated transcription factors that control gene expression in many cell types. NFAT proteins are heavily phosphorylated and reside in the cytoplasm of resting cells; when cells are stimulated by a rise in intracellular Ca(2+), NFAT proteins are dephosphorylated by the Ca(2+)/calmodulin-dependent phosphatase calcineurin and translocate to the nucleus to activate target gene expression. Here we show that phosphorylated NFAT1 is present in a large cytoplasmic RNA-protein scaffold complex that contains a long intergenic noncoding RNA (lincRNA), NRON [noncoding (RNA) repressor of NFAT]; a scaffold protein, IQ motif containing GTPase activating protein (IQGAP); and three NFAT kinases, casein kinase 1, glycogen synthase kinase 3, and dual specificity tyrosine phosphorylation regulated kinase. Combined knockdown of NRON and IQGAP1 increased NFAT dephosphorylation and nuclear import exclusively after stimulation, without affecting the rate of NFAT rephosphorylation and nuclear export; and both NRON-depleted T cells and T cells from IQGAP1-deficient mice showed increased production of NFAT-dependent cytokines. Our results provide evidence that a complex of lincRNA and protein forms a scaffold for a latent transcription factor and its regulatory kinases, and support an emerging consensus that lincRNAs that bind transcriptional regulators have a similar scaffold function.


Asunto(s)
Factores de Transcripción NFATC/química , Factores de Transcripción NFATC/metabolismo , ARN/química , ARN/metabolismo , Transporte Activo de Núcleo Celular , Animales , Secuencia de Bases , Linfocitos T CD8-positivos/metabolismo , Cartilla de ADN/genética , Células HeLa , Humanos , Células Jurkat , Sustancias Macromoleculares/química , Sustancias Macromoleculares/metabolismo , Ratones , Ratones Noqueados , Modelos Biológicos , Fosforilación , ARN Largo no Codificante , ARN Interferente Pequeño/genética , ARN no Traducido/química , ARN no Traducido/genética , ARN no Traducido/metabolismo , Proteínas Activadoras de ras GTPasa/química , Proteínas Activadoras de ras GTPasa/deficiencia , Proteínas Activadoras de ras GTPasa/genética , Proteínas Activadoras de ras GTPasa/metabolismo
12.
J Exp Med ; 204(8): 1875-89, 2007 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-17664287

RESUMEN

Although inhibitory Fc gamma receptors have been demonstrated to promote mucosal tolerance, the role of activating Fc gamma receptors in modulating T helper type (Th)2-dependent inflammatory responses characteristic of asthma and allergies remains unclear. Here, we demonstrate that signaling via activating Fc gamma receptors in conjunction with Toll-like receptor 4 stimulation modulated cytokine production from bone marrow-derived dendritic cells (DCs) and augmented their ability to promote Th2 responses. Ligation of the low affinity receptor Fc gamma RIII was specifically required for the enhanced Th2 responses, as Fc gamma RIII(-/-) DCs failed to augment Th2-mediated airway inflammation in vivo or induce Th2 differentiation in vitro. Further, Fc gamma RIII(-/-) mice had impaired Th2 cytokine production and exhibited reduced airway inflammation, whereas no defect was found in Fc gamma RI(-/-) mice. The augmentation of Th2 immunity was regulated by interleukin 10 production from the DCs but was distinct and independent of the well-established role of Fc gamma RIII in augmenting antigen presentation. Thus, our studies reveal a novel and specific role for Fc gamma RIII signaling in the regulation of Th cell responses and suggest that in addition to immunoglobulin (Ig)E, antigen-specific IgG also contributes to the pathogenesis of Th2-mediated diseases such as asthma and allergies.


Asunto(s)
Regulación de la Expresión Génica , Receptores de IgG/metabolismo , Células Th2/metabolismo , Animales , Presentación de Antígeno , Asma/metabolismo , Células Dendríticas/metabolismo , Femenino , Hipersensibilidad/metabolismo , Inflamación , Interleucina-10/metabolismo , Ratones , Ratones Endogámicos BALB C , Transducción de Señal , Receptor Toll-Like 4/metabolismo
13.
Nat Rev Drug Discov ; 21(9): 655-675, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35637318

RESUMEN

Cell-based therapeutics are an emerging modality with the potential to treat many currently intractable diseases through uniquely powerful modes of action. Despite notable recent clinical and commercial successes, cell-based therapies continue to face numerous challenges that limit their widespread translation and commercialization, including identification of the appropriate cell source, generation of a sufficiently viable, potent and safe product that meets patient- and disease-specific needs, and the development of scalable manufacturing processes. These hurdles are being addressed through the use of cutting-edge basic research driven by next-generation engineering approaches, including genome and epigenome editing, synthetic biology and the use of biomaterials.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos , Edición Génica , Humanos
14.
Am J Respir Cell Mol Biol ; 43(3): 342-8, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19855087

RESUMEN

Our previous studies revealed that, in a murine model of asthma, mice that received Fas-deficient T cells developed a prolonged phase of airway inflammation, mucus production, and airway hyperreactivity that failed to resolve even 6 weeks after the last challenge. To investigate how Fas-Fas ligand (FasL) interaction occurs between T cells and other cells in vivo, Gld mice with abnormalities of the FasL signaling pathway were used. The reconstituted mice were made by transferring T cells from B6 or Gld mice to Rag(-/-) or FasL-deficient Rag(-/-) mice. We found that Rag(-/-) mice that received B6 T cells resolved the airway inflammation, whereas FasL-deficient Rag(-/-) mice that received Gld T cells developed a prolonged airway inflammation at Day 28, with decreased IFN-gamma production. Both FasL-deficient Rag(-/-) mice that received B6 T cells and Rag(-/-) mice that received Gld T cells also had completely resolved their airway inflammation by Day 28 after challenge. Interestingly, FasL-deficient Rag(-/-) mice that received Gld T cells eventually resolved airway inflammation at Day 42, with a similar level of IFN-gamma production to that of control group. These results demonstrate that FasL expression on either T cells only or non-T cells only was sufficient for the eventual resolution of airway inflammation, and the prolonged airway inflammation in FasL-deficient Rag(-/-) mice that received Gld T cells was correlated with decreased IFN-gamma production by Gld T cells.


Asunto(s)
Asma/prevención & control , Modelos Animales de Enfermedad , Proteína Ligando Fas/fisiología , Sistema Respiratorio/metabolismo , Linfocitos T/metabolismo , Traslado Adoptivo , Animales , Asma/inmunología , Asma/metabolismo , Células Cultivadas , Citocinas/metabolismo , Citometría de Flujo , Proteínas de Homeodominio/fisiología , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Noqueados , Linfocitos T/trasplante
15.
J Immunol ; 181(2): 1019-24, 2008 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-18606653

RESUMEN

The T cell costimulatory molecule ICOS regulates Th2 effector function in allergic airway disease. Recently, several studies with ICOS(-/-) mice have also demonstrated a role for ICOS in Th2 differentiation. To determine the effects of ICOS on the early immune response, we investigated augmenting ICOS costimulation in a Th2-mediated immune response to Schistosoma mansoni Ags. We found that augmenting ICOS costimulation with B7RP-1-Fc increased the accumulation of T and B cells in the draining lymph nodes postimmunization. Interestingly, the increased numbers were due in part to increased migration of undivided Ag-specific TCR transgenic T cells and surprisingly B cells, as well as non-TCR transgenic T cells. B7RP-1-Fc also increased the levels of the chemokines CCL21 and CXCL13 in the draining lymph node, suggesting ICOS costimulation contributes to migration by direct or indirect effects on dendritic cells, stromal cells and high endothelial venules. Further, the effects of B7RP-1-Fc were not dependent on immunization. Our data support a model in which ICOS costimulation augments the pool of lymphocytes in the draining lymph nodes, leading to an increase in the frequency of potentially reactive T and B cells.


Asunto(s)
Antígenos de Diferenciación de Linfocitos T/inmunología , Antígeno B7-1/inmunología , Citocinas/metabolismo , Ganglios Linfáticos/inmunología , Linfocitos/inmunología , Schistosoma mansoni/inmunología , Células Th2/inmunología , Animales , Antígenos de Diferenciación de Linfocitos T/metabolismo , Linfocitos B/inmunología , Antígeno B7-1/metabolismo , Movimiento Celular , Citocinas/inmunología , Femenino , Ligando Coestimulador de Linfocitos T Inducibles , Proteína Coestimuladora de Linfocitos T Inducibles , Ganglios Linfáticos/citología , Ganglios Linfáticos/metabolismo , Linfocitos/citología , Linfocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Linfocitos T/inmunología
16.
Cell Immunol ; 259(2): 177-84, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19646680

RESUMEN

Previous work has shown ICOS can function independently of CD28, but whether either molecule can compensate for the other in vivo is not known. Since ICOS is a potent inducer of Th2 cytokines and linked to allergy and elevated serum IgE in humans, we hypothesized that augmenting ICOS costimulation in murine allergic airway disease may overcome CD28 deficiency. While ICOS was expressed on T cells from CD28(-/-) mice, Th2-mediated airway inflammation was not induced in CD28(-/-) mice by increased ICOS costimulation. Further, we determined if augmenting CD28 costimulation could compensate for ICOS deficiency. ICOS(-/-) mice had a defect in airway eosinophilia that was not overcome by augmenting CD28 costimulation. CD28 costimulation also did not fully compensate for ICOS for antibody responses, germinal center formation or the development of follicular B helper T cells. CD28 and ICOS play complementary non-overlapping roles in the development of Th2 immunity in vivo.


Asunto(s)
Antígenos de Diferenciación de Linfocitos T/inmunología , Antígenos CD28/inmunología , Enfermedades Pulmonares/inmunología , Células Th2/inmunología , Animales , Líquido del Lavado Bronquioalveolar/inmunología , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Histocitoquímica , Inmunidad Celular/inmunología , Inmunoglobulina E/sangre , Proteína Coestimuladora de Linfocitos T Inducibles , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Organismos Libres de Patógenos Específicos
17.
Nat Commun ; 5: 4540, 2014 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-25105474

RESUMEN

Jarid2 is a reported component of three lysine methyltransferase complexes, polycomb repressive complex 2 (PRC2) that methylates histone 3 lysine 27 (H3K27), and GLP-G9a and SETDB1 complexes that methylate H3K9. Here we show that Jarid2 is upregulated upon TCR stimulation and during positive selection in the thymus. Mice lacking Jarid2 in T cells display an increase in the frequency of IL-4-producing promyelocytic leukemia zinc finger (PLZF)(hi) immature invariant natural killer T (iNKT) cells and innate-like CD8(+) cells; Itk-deficient mice, which have a similar increase of innate-like CD8(+) cells, show blunted upregulation of Jarid2 during positive selection. Jarid2 binds to the Zbtb16 locus, which encodes PLZF, and thymocytes lacking Jarid2 show increased PLZF and decreased H3K9me3 levels. Jarid2-deficient iNKT cells perturb Th17 differentiation, leading to reduced Th17-driven autoimmune pathology. Our results establish Jarid2 as a novel player in iNKT cell maturation that regulates PLZF expression by modulating H3K9 methylation.


Asunto(s)
Células Asesinas Naturales/citología , Complejo Represivo Polycomb 2/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Animales , Antígenos CD/metabolismo , Antígenos de Diferenciación de Linfocitos T/metabolismo , Células de la Médula Ósea/citología , Linfocitos T CD4-Positivos/citología , Linfocitos T CD8-positivos/citología , Diferenciación Celular , Separación Celular , Femenino , Citometría de Flujo , Histonas/química , Interleucina-4/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Lectinas Tipo C/metabolismo , Lisina/química , Masculino , Metilación , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , MicroARNs/metabolismo , Regiones Promotoras Genéticas , Proteína de la Leucemia Promielocítica con Dedos de Zinc , Transducción de Señal , Timo/metabolismo , Regulación hacia Arriba , Dedos de Zinc
18.
Nat Commun ; 4: 2990, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24356538

RESUMEN

Atopic asthma is an inflammatory pulmonary disease associated with Th2 adaptive immune responses triggered by innocuous antigens. While dendritic cells (DCs) are known to shape the adaptive immune response, the mechanisms by which DCs promote Th2 differentiation remain elusive. Herein we demonstrate that Th2-promoting stimuli induce DC expression of IRF4. Mice with conditional deletion of Irf4 in DCs show a dramatic defect in Th2-type lung inflammation, yet retain the ability to elicit pulmonary Th1 antiviral responses. Using loss- and gain-of-function analysis, we demonstrate that Th2 differentiation is dependent on IRF4 expression in DCs. Finally, IRF4 directly targets and activates the Il-10 and Il-33 genes in DCs. Reconstitution with exogenous IL-10 and IL-33 recovers the ability of Irf4-deficient DCs to promote Th2 differentiation. These findings reveal a regulatory module in DCs by which IRF4 modulates IL-10 and IL-33 cytokine production to specifically promote Th2 differentiation and inflammation.


Asunto(s)
Asma/metabolismo , Diferenciación Celular , Células Dendríticas/citología , Hipersensibilidad Inmediata/metabolismo , Factores Reguladores del Interferón/metabolismo , Pulmón/metabolismo , Células Th2/citología , Animales , Células de la Médula Ósea/citología , Femenino , Regulación de la Expresión Génica , Inflamación , Interleucina-10/metabolismo , Interleucinas , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Ácaros , Análisis de Secuencia por Matrices de Oligonucleótidos , Transducción de Señal
19.
PLoS One ; 4(11): e7525, 2009 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-19888475

RESUMEN

BACKGROUND: Inducible Costimulator (ICOS) is an important regulator of Th2 lymphocyte function and a potential immunotherapeutic target for allergy and asthma. A SNP in the ICOS 5' promoter in humans is associated with increased atopy and serum IgE in a founder population and increased ICOS surface expression and Th2 cytokine production from peripheral blood mononuclear cells. However, it is unknown if increased ICOS expression contributes to disease progression or is a result of disease pathology. METHODOLOGY/PRINCIPAL FINDINGS: We developed a mouse model in which ICOS surface expression levels are genetically predetermined to test our hypothesis that genetic regulation of ICOS expression controls the severity of Th2 responses in vivo. Using ICOS+/+ and ICOS+/- mice in a Th2 model of airway inflammation, we found that T cells from the ICOS+/- mice had reduced ICOS expression and decreased Th2-mediated inflammation in vivo. Although the activation status of the T cells did not differ, T cells isolated from the lungs and draining lymph nodes of ICOS+/- mice at the peak of inflammation produced less Th2 cytokines upon stimulation ex vivo. Using 4get mice, which express GFP upon IL-4 transcription, we determined that the decreased Th2 cytokines in ICOS+/- is due to reduced percentage of Th2 cells and not a defect in their ability to produce IL-4. CONCLUSION: These data suggest that in both mice and humans, the level of ICOS surface expression regulates the magnitude of the in vivo Th2 response, perhaps by influencing Th2 differentiation.


Asunto(s)
Antígenos de Diferenciación de Linfocitos T/genética , Antígenos de Diferenciación de Linfocitos T/fisiología , Regulación de la Expresión Génica , Células Th2/citología , Transcripción Genética , Animales , Células Presentadoras de Antígenos , Membrana Celular/metabolismo , Citocinas/metabolismo , Progresión de la Enfermedad , Femenino , Proteína Coestimuladora de Linfocitos T Inducibles , Inflamación , Interleucina-4/metabolismo , Leucocitos Mononucleares/citología , Ganglios Linfáticos/metabolismo , Recuento de Linfocitos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
20.
Science ; 324(5929): 930-5, 2009 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-19372391

RESUMEN

DNA cytosine methylation is crucial for retrotransposon silencing and mammalian development. In a computational search for enzymes that could modify 5-methylcytosine (5mC), we identified TET proteins as mammalian homologs of the trypanosome proteins JBP1 and JBP2, which have been proposed to oxidize the 5-methyl group of thymine. We show here that TET1, a fusion partner of the MLL gene in acute myeloid leukemia, is a 2-oxoglutarate (2OG)- and Fe(II)-dependent enzyme that catalyzes conversion of 5mC to 5-hydroxymethylcytosine (hmC) in cultured cells and in vitro. hmC is present in the genome of mouse embryonic stem cells, and hmC levels decrease upon RNA interference-mediated depletion of TET1. Thus, TET proteins have potential roles in epigenetic regulation through modification of 5mC to hmC.


Asunto(s)
5-Metilcitosina/metabolismo , Citosina/análogos & derivados , Proteínas de Unión al ADN/metabolismo , ADN/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , Citosina/análisis , Citosina/metabolismo , ADN/química , Metilación de ADN , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Fosfatos de Dinucleósidos/metabolismo , Células Madre Embrionarias/química , Células Madre Embrionarias/metabolismo , Humanos , Hidroxilación , Espectrometría de Masas , Ratones , Oxigenasas de Función Mixta , Datos de Secuencia Molecular , Proteínas Proto-Oncogénicas/química , Proteínas Proto-Oncogénicas/genética , Interferencia de ARN , Alineación de Secuencia , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA