Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 84
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 15(12): 1097-8, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25396343

RESUMEN

Humans deficient in the adaptor MyD88 or the kinase IRAK4 suffer from primary immunodeficiency. Blood cells from these patients show defective induction of specific subsets of genes after exposure to microbial stimuli in vitro.


Asunto(s)
Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/inmunología , Quinasas Asociadas a Receptores de Interleucina-1/genética , Mutación , Factor 88 de Diferenciación Mieloide/genética , Femenino , Humanos , Quinasas Asociadas a Receptores de Interleucina-1/inmunología , Masculino , Enfermedades de Inmunodeficiencia Primaria
2.
Nat Immunol ; 19(6): 518-520, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29777225
3.
Proc Natl Acad Sci U S A ; 118(9)2021 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-33619107

RESUMEN

Reactivation of human cytomegalovirus (HCMV) from latency is a major health consideration for recipients of stem-cell and solid organ transplantations. With over 200,000 transplants taking place globally per annum, virus reactivation can occur in more than 50% of cases leading to loss of grafts as well as serious morbidity and even mortality. Here, we present the most extensive screening to date of epigenetic inhibitors on HCMV latently infected cells and find that histone deacetylase inhibitors (HDACis) and bromodomain inhibitors are broadly effective at inducing virus immediate early gene expression. However, while HDACis, such as myeloid-selective CHR-4487, lead to production of infectious virions, inhibitors of bromodomain (BRD) and extraterminal proteins (I-BETs), including GSK726, restrict full reactivation. Mechanistically, we show that BET proteins (BRDs) are pivotally connected to regulation of HCMV latency and reactivation. Through BRD4 interaction, the transcriptional activator complex P-TEFb (CDK9/CycT1) is sequestered by repressive complexes during HCMV latency. Consequently, I-BETs allow release of P-TEFb and subsequent recruitment to promoters via the superelongation complex (SEC), inducing transcription of HCMV lytic genes encoding immunogenic antigens from otherwise latently infected cells. Surprisingly, this occurs without inducing many viral immunoevasins and, importantly, while also restricting viral DNA replication and full HCMV reactivation. Therefore, this pattern of HCMV transcriptional dysregulation allows effective cytotoxic immune targeting and killing of latently infected cells, thus reducing the latent virus genome load. This approach could be safely used to pre-emptively purge the virus latent reservoir prior to transplantation, thereby reducing HCMV reactivation-related morbidity and mortality.


Asunto(s)
Proteínas de Ciclo Celular/genética , Citomegalovirus/inmunología , ADN Viral/genética , Epigénesis Genética , Histona Desacetilasas/genética , Factor B de Elongación Transcripcional Positiva/genética , Factores de Transcripción/genética , Azepinas/farmacología , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Benzodiazepinas/farmacología , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/inmunología , Ciclina T/genética , Ciclina T/inmunología , Quinasa 9 Dependiente de la Ciclina/genética , Quinasa 9 Dependiente de la Ciclina/inmunología , Citomegalovirus/efectos de los fármacos , Citomegalovirus/genética , Infecciones por Citomegalovirus/genética , Infecciones por Citomegalovirus/inmunología , Infecciones por Citomegalovirus/patología , Replicación del ADN/efectos de los fármacos , ADN Viral/antagonistas & inhibidores , ADN Viral/inmunología , Genes Inmediatos-Precoces , Genes Reporteros , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/inmunología , Interacciones Huésped-Patógeno , Humanos , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Modelos Biológicos , Factor B de Elongación Transcripcional Positiva/inmunología , Cultivo Primario de Células , Regiones Promotoras Genéticas , Linfocitos T Citotóxicos/efectos de los fármacos , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/virología , Células THP-1 , Talidomida/análogos & derivados , Talidomida/farmacología , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/inmunología , Transcripción Genética , Activación Viral/efectos de los fármacos , Latencia del Virus/efectos de los fármacos
4.
Curr Issues Mol Biol ; 45(5): 4228-4245, 2023 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-37232738

RESUMEN

SP140 is an epigenetic reader protein expressed predominantly in immune cells. GWAS studies have shown an association between SP140 single nucleotide polymorphisms (SNPs) and diverse autoimmune and inflammatory diseases, suggesting a possible pathogenic role for SP140 in immune-mediated diseases. We previously demonstrated that treatment of human macrophages with the novel selective inhibitor of the SP140 protein (GSK761) reduced the expression of endotoxin-induced cytokines, implicating a role of SP140 in the function of inflammatory macrophages. In this study, we investigated the effects of GSK761 on in vitro human dendritic cell (DC) differentiation and maturation, assessing the expression of cytokines and co-stimulatory molecules and their capacity to stimulate T-cell activation and induce phenotypic changes. In DCs, lipopolysaccharide (LPS) stimulation induced an increase in SP140 expression and its recruitment to transcription start sites (TSS) of pro-inflammatory cytokine genes. Moreover, LPS-induced cytokines such as TNF, IL-6, and IL-1ß were reduced in GSK761- or SP140 siRNA- treated DCs. Although GSK761 did not significantly affect the expression of surface markers that define the differentiation of CD14+ monocytes into immature DCs (iDCs), subsequent maturation of iDCs to mature DCs was significantly inhibited. GSK761 strongly reduced expression of the maturation marker CD83, the co-stimulatory molecules CD80 and CD86, and the lipid-antigen presentation molecule CD1b. Finally, when the ability of DCs to stimulate recall T-cell responses by vaccine-specific T cells was assessed, T cells stimulated by GSK761-treated DCs showed reduced TBX21 and RORA expression and increased FOXP3 expression, indicating a preferential generation of regulatory T cells. Overall, this study suggests that SP140 inhibition enhances the tolerogenic properties of DCs, supporting the rationale of targeting SP140 in autoimmune and inflammatory diseases where DC-mediated inflammatory responses contribute to disease pathogenesis.

5.
Clin Exp Immunol ; 211(2): 93-95, 2023 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-36807653

RESUMEN

The immune system plays a critical role in protecting the host against infection but is subject to numerous levels of control that are necessary to prevent pathological, tissue-damaging responses. Inappropriate inflammatory immune responses to self-antigens, innocuous commensal microorganisms, or environmental antigens can lead to chronic, debilitating, and degenerative diseases. Regulatory T cells have an essential, non-redundant, and dominant function in preventing pathological immune responses, as shown by the development of systemic fatal autoimmunity in humans and animals with a genetic deficiency in regulatory T cells. In addition to controlling immune responses, there is a growing understanding that regulatory T cells also contribute directly to tissue homeostasis by promoting tissue regeneration and repair. For these reasons, the prospect of enhancing regulatory T-cell numbers and/or function in patients represents an appealing therapeutic opportunity with potential applications in many diseases, including some where the pathological role of the immune system has only recently been recognized. Approaches to enhance regulatory T cells are now starting to be explored in clinical studies in humans. This review series brings together papers highlighting the Treg-enhancing approaches that are most advanced clinically and examples of therapeutic opportunities based on our growing understanding of regulatory T-cell functions.


Asunto(s)
Autoinmunidad , Linfocitos T Reguladores , Animales , Humanos , Autoantígenos
6.
Trends Immunol ; 41(1): 29-45, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31813765

RESUMEN

Memory T cells possess functional differences from naïve T cells that powerfully contribute to the efficiency of secondary immune responses. These abilities are imprinted during the primary response, linked to the acquisition of novel patterns of gene expression. Underlying this are alterations at the chromatin level (epigenetic modifications) that regulate constitutive and inducible gene transcription. T cell epigenetic memory can persist long-term, contributing to long-lasting immunity after infection or vaccination. However, acquired epigenetic states can also hinder effective tumor immunity or contribute to autoimmunity. The growing understanding of epigenetic gene regulation as it relates to both the stability and malleability of T cell memory may offer the potential to selectively modify T cell memory in disease by targeting epigenetic mechanisms.


Asunto(s)
Epigénesis Genética , Memoria Inmunológica , Linfocitos T , Cromatina/inmunología , Epigénesis Genética/inmunología , Regulación de la Expresión Génica , Humanos , Memoria Inmunológica/genética , Linfocitos T/inmunología
7.
BMC Biol ; 20(1): 182, 2022 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-35986286

RESUMEN

BACKGROUND: SP140 is a bromodomain-containing protein expressed predominantly in immune cells. Genetic polymorphisms and epigenetic modifications in the SP140 locus have been linked to Crohn's disease (CD), suggesting a role in inflammation. RESULTS: We report the development of the first small molecule SP140 inhibitor (GSK761) and utilize this to elucidate SP140 function in macrophages. We show that SP140 is highly expressed in CD mucosal macrophages and in in vitro-generated inflammatory macrophages. SP140 inhibition through GSK761 reduced monocyte-to-inflammatory macrophage differentiation and lipopolysaccharide (LPS)-induced inflammatory activation, while inducing the generation of CD206+ regulatory macrophages that were shown to associate with a therapeutic response to anti-TNF in CD patients. SP140 preferentially occupies transcriptional start sites in inflammatory macrophages, with enrichment at gene loci encoding pro-inflammatory cytokines/chemokines and inflammatory pathways. GSK761 specifically reduces SP140 chromatin binding and thereby expression of SP140-regulated genes. GSK761 inhibits the expression of cytokines, including TNF, by CD14+ macrophages isolated from CD intestinal mucosa. CONCLUSIONS: This study identifies SP140 as a druggable epigenetic therapeutic target for CD.


Asunto(s)
Enfermedad de Crohn , Inhibidores del Factor de Necrosis Tumoral , Antígenos Nucleares/genética , Antígenos Nucleares/metabolismo , Enfermedad de Crohn/genética , Enfermedad de Crohn/metabolismo , Citocinas/genética , Citocinas/metabolismo , Epigénesis Genética , Humanos , Macrófagos , Factores de Transcripción/genética
8.
Br J Clin Pharmacol ; 88(12): 5238-5256, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35655123

RESUMEN

AIMS: To improve the tolerability and therapeutic application of histone deacetylase inhibitors (HDACi), by application of an esterase-sensitive motif (ESM), to target pharmacological activity directly to mononuclear myeloid cells expressing the processing enzyme carboxylesterase-1 (CES1). METHODS: This first-in-human study comprised single and multiple ascending dose cohorts to determine safety and tolerability. Pharmacodynamic parameters included acetylation, cytokine inhibition and intracellular concentrations of processed acid metabolite in isolated monocytes. Mechanistic work was conducted in vitro and in a CES1/Es1elo mouse strain. RESULTS: ESM-HDAC391 showed transient systemic exposure (plasma half-life of 21-30 min) but selective retention of processed acid for at least 12 hours, resulting in robust targeted mechanistic engagement (increased acetylation in monocytes plus inhibition of ex vivo stimulated cytokine production). ESM-HDAC391 was well tolerated and clinical toxicities common to non-targeted HDACi were not observed. ESM-HDAC391 treatment was accompanied by the novel finding of a dose-dependent monocyte depletion that was transient and reversible and which plateaued at 0.06 × 109 monocytes/L after repeat dosing with 20 or 40 mg. Characterisation of monocyte depletion in transgenic mice (CES1/Es1elo ) suggested that colony stimulating factor 1 receptor loss on circulating cells contributed to ESM-HDAC-mediated depletion. Further mechanistic investigations using human monocytes in vitro demonstrated HDACi-mediated change in myeloid fate through modulation of colony stimulating factor 1 receptor and downstream effects on cell differentiation. CONCLUSION: These findings demonstrate selective targeting of monocytes in humans using the ESM approach and identify monocytopaenia as a novel outcome of ESM-HDACi treatment, with implications for potential benefit of these molecules in myeloid-driven diseases.


Asunto(s)
Esterasas , Inhibidores de Histona Desacetilasas , Humanos , Animales , Ratones , Inhibidores de Histona Desacetilasas/farmacología , Factor Estimulante de Colonias de Macrófagos , Citocinas
9.
J Immunol ; 205(9): 2479-2488, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-32978279

RESUMEN

The human TNF/LT locus genes TNF, LTA, and LTB are expressed in a cell type-specific manner. In this study, we show that a highly conserved NFAT binding site within the distal noncoding element hHS-8 coordinately controls TNF and LTA gene expression in human T cells. Upon activation of primary human CD4+ T cells, hHS-8 and the TNF and LTA promoters display increased H3K27 acetylation and nuclease sensitivity and coordinate induction of TNF, LTA, and hHS-8 enhancer RNA transcription occurs. Functional analyses using CRISPR/dead(d)Cas9 targeting of the hHS-8-NFAT site in the human T cell line CEM demonstrate significant reduction of TNF and LTA mRNA synthesis and of RNA polymerase II recruitment to their promoters. These studies elucidate how a distal element regulates the inducible cell type-specific gene expression program of the human TNF/LT locus and provide an approach for modulation of TNF and LTA transcription in human disease using CRISPR/dCas9.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Expresión Génica/genética , Linfotoxina-alfa/genética , Factor de Necrosis Tumoral alfa/genética , Acetilación , Sitios de Unión/genética , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Secuencia Conservada/genética , Elementos de Facilitación Genéticos/genética , Histonas/genética , Humanos , Leucocitos Mononucleares/metabolismo , Regiones Promotoras Genéticas/genética , Unión Proteica/genética , ARN Polimerasa II/genética , ARN Mensajero/genética , Células THP-1/metabolismo , Transcripción Genética/genética
10.
Ann Rheum Dis ; 80(10): 1268-1277, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34380700

RESUMEN

Increasingly earlier identification of individuals at high risk of rheumatoid arthritis (RA) (eg, with autoantibodies and mild symptoms) improves the feasibility of preventing or curing disease. The use of antigen-specific immunotherapies to reinstate immunological self-tolerance represent a highly attractive strategy due to their potential to induce disease resolution, in contrast to existing approaches that require long-term treatment of underlying symptoms.Preclinical animal models have been used to understand disease mechanisms and to evaluate novel immunotherapeutic approaches. However, models are required to understand critical processes supporting disease development such as the breach of self-tolerance that triggers autoimmunity and the progression from asymptomatic autoimmunity to joint pain and bone loss. These models would also be useful in evaluating the response to treatment in the pre-RA period.This review proposes that focusing on immune processes contributing to initial disease induction rather than end-stage pathological consequences is essential to allow development and evaluation of novel immunotherapies for early intervention. We will describe and critique existing models in arthritis and the broader field of autoimmunity that may fulfil these criteria. We will also identify key gaps in our ability to study these processes in animal models, to highlight where further research should be targeted.


Asunto(s)
Artritis Experimental/inmunología , Artritis Reumatoide/inmunología , Autoanticuerpos/inmunología , Autoinmunidad/inmunología , Inmunoterapia , Autotolerancia/inmunología , Animales , Anticuerpos Antiproteína Citrulinada/inmunología , Artritis Experimental/prevención & control , Artritis Experimental/terapia , Artritis Reumatoide/prevención & control , Artritis Reumatoide/terapia , Enfermedades Asintomáticas , Desensibilización Inmunológica , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Tolerancia Inmunológica/inmunología , Ratones , Ratas , Factor Reumatoide/inmunología
11.
Mol Cell ; 51(3): 310-25, 2013 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-23932714

RESUMEN

Recent studies suggest a hierarchical model in which lineage-determining factors act in a collaborative manner to select and prime cell-specific enhancers, thereby enabling signal-dependent transcription factors to bind and function in a cell-type-specific manner. Consistent with this model, TLR4 signaling primarily regulates macrophage gene expression through a pre-existing enhancer landscape. However, TLR4 signaling also induces priming of ∼3,000 enhancer-like regions de novo, enabling visualization of intermediates in enhancer selection and activation. Unexpectedly, we find that enhancer transcription precedes local mono- and dimethylation of histone H3 lysine 4 (H3K4me1/2). H3K4 methylation at de novo enhancers is primarily dependent on the histone methyltransferases Mll1, Mll2/4, and Mll3 and is significantly reduced by inhibition of RNA polymerase II elongation. Collectively, these findings suggest an essential role of enhancer transcription in H3K4me1/2 deposition at de novo enhancers that is independent of potential functions of the resulting eRNA transcripts.


Asunto(s)
Elementos de Facilitación Genéticos , Activación de Macrófagos/genética , Receptor Toll-Like 4/metabolismo , Animales , Secuencia de Bases , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Células Cultivadas , Metilación de ADN , Expresión Génica , Regulación de la Expresión Génica , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/genética , Histonas/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , FN-kappa B/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , ARN Polimerasa II/antagonistas & inhibidores , Análisis de Secuencia de ADN , Transducción de Señal , Transactivadores/metabolismo , Factor de Transcripción ReIA/metabolismo , Transcripción Genética
12.
Immunity ; 30(2): 171-3, 2009 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-19239898

RESUMEN

CD40L expressed on T cells plays an important role in stimulating the function of dendritic cells (DCs). In this issue of Immunity, Johnson et al. (2009) demonstrate a role for DC-expressed CD40L in priming CD8(+) T cell responses.


Asunto(s)
Ligando de CD40/inmunología , Ligando de CD40/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Linfocitos T/inmunología , Animales
13.
PLoS Biol ; 13(12): e1002316, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26646171

RESUMEN

Trypanosoma brucei, the causative agent of African sleeping sickness, is transmitted to its mammalian host by the tsetse. In the fly, the parasite's surface is covered with invariant procyclin, while in the mammal it resides extracellularly in its bloodstream form (BF) and is densely covered with highly immunogenic Variant Surface Glycoprotein (VSG). In the BF, the parasite varies this highly immunogenic surface VSG using a repertoire of ~2500 distinct VSG genes. Recent reports in mammalian systems point to a role for histone acetyl-lysine recognizing bromodomain proteins in the maintenance of stem cell fate, leading us to hypothesize that bromodomain proteins may maintain the BF cell fate in trypanosomes. Using small-molecule inhibitors and genetic mutants for individual bromodomain proteins, we performed RNA-seq experiments that revealed changes in the transcriptome similar to those seen in cells differentiating from the BF to the insect stage. This was recapitulated at the protein level by the appearance of insect-stage proteins on the cell surface. Furthermore, bromodomain inhibition disrupts two major BF-specific immune evasion mechanisms that trypanosomes harness to evade mammalian host antibody responses. First, monoallelic expression of the antigenically varied VSG is disrupted. Second, rapid internalization of antibodies bound to VSG on the surface of the trypanosome is blocked. Thus, our studies reveal a role for trypanosome bromodomain proteins in maintaining bloodstream stage identity and immune evasion. Importantly, bromodomain inhibition leads to a decrease in virulence in a mouse model of infection, establishing these proteins as potential therapeutic drug targets for trypanosomiasis. Our 1.25Å resolution crystal structure of a trypanosome bromodomain in complex with I-BET151 reveals a novel binding mode of the inhibitor, which serves as a promising starting point for rational drug design.


Asunto(s)
Modelos Moleculares , Proteínas Protozoarias/metabolismo , Factores de Transcripción/metabolismo , Trypanosoma brucei brucei/fisiología , Sustitución de Aminoácidos , Animales , Sitios de Unión , Línea Celular , Técnicas de Silenciamiento del Gen , Técnicas de Inactivación de Genes , Evasión Inmune , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Conformación Proteica , Isoformas de Proteínas/antagonistas & inhibidores , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Protozoarias/antagonistas & inhibidores , Proteínas Protozoarias/química , Proteínas Protozoarias/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Análisis de Supervivencia , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/química , Factores de Transcripción/genética , Tripanocidas/farmacología , Tripanocidas/uso terapéutico , Trypanosoma brucei brucei/efectos de los fármacos , Trypanosoma brucei brucei/inmunología , Trypanosoma brucei brucei/patogenicidad , Tripanosomiasis Africana/tratamiento farmacológico , Tripanosomiasis Africana/parasitología , Tripanosomiasis Africana/fisiopatología , Virulencia
14.
Blood ; 123(5): 697-705, 2014 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-24335499

RESUMEN

The bromodomain and extraterminal (BET) protein BRD2-4 inhibitors hold therapeutic promise in preclinical models of hematologic malignancies. However, translation of these data to molecules suitable for clinical development has yet to be accomplished. Herein we expand the mechanistic understanding of BET inhibitors in multiple myeloma by using the chemical probe molecule I-BET151. I-BET151 induces apoptosis and exerts strong antiproliferative effect in vitro and in vivo. This is associated with contrasting effects on oncogenic MYC and HEXIM1, an inhibitor of the transcriptional activator P-TEFb. I-BET151 causes transcriptional repression of MYC and MYC-dependent programs by abrogating recruitment to the chromatin of the P-TEFb component CDK9 in a BRD2-4-dependent manner. In contrast, transcriptional upregulation of HEXIM1 is BRD2-4 independent. Finally, preclinical studies show that I-BET762 has a favorable pharmacologic profile as an oral agent and that it inhibits myeloma cell proliferation, resulting in survival advantage in a systemic myeloma xenograft model. These data provide a strong rationale for extending the clinical testing of the novel antimyeloma agent I-BET762 and reveal insights into biologic pathways required for myeloma cell proliferation.


Asunto(s)
Antineoplásicos/uso terapéutico , Benzodiazepinas/uso terapéutico , Compuestos Heterocíclicos de 4 o más Anillos/uso terapéutico , Mieloma Múltiple/tratamiento farmacológico , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Benzodiazepinas/farmacología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Humanos , Ratones , Mieloma Múltiple/genética , Mieloma Múltiple/patología , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas de Unión al ARN/genética , Factores de Transcripción , Activación Transcripcional/efectos de los fármacos , Células Tumorales Cultivadas
15.
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
16.
Biomedicines ; 12(6)2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38927552

RESUMEN

T cell activation is critical for an effective immune response against pathogens. However, dysregulation contributes to the pathogenesis of autoimmune diseases, including Juvenile Idiopathic Arthritis (JIA). The molecular mechanisms underlying T cell activation are still incompletely understood. T cell activation promotes the acetylation of histone 3 at Lysine 27 (H3K27ac) at enhancer and promoter regions of proinflammatory cytokines, thereby increasing the expression of these genes which is essential for T cell function. Co-activators E1A binding protein P300 (P300) and CREB binding protein (CBP), collectively known as P300/CBP, are essential to facilitate H3K27 acetylation. Presently, the role of P300/CBP in human CD4+ T cells activation remains incompletely understood. To assess the function of P300/CBP in T cell activation and autoimmune disease, we utilized iCBP112, a selective inhibitor of P300/CBP, in T cells obtained from healthy controls and JIA patients. Treatment with iCBP112 suppressed T cell activation and cytokine signaling pathways, leading to reduced expression of many proinflammatory cytokines, including IL-2, IFN-γ, IL-4, and IL-17A. Moreover, P300/CBP inhibition in T cells derived from the inflamed synovium of JIA patients resulted in decreased expression of similar pathways and preferentially suppressed the expression of disease-associated genes. This study underscores the regulatory role of P300/CBP in regulating gene expression during T cell activation while offering potential insights into the pathogenesis of autoimmune diseases. Our findings indicate that P300/CBP inhibition could potentially be leveraged for the treatment of autoimmune diseases such as JIA in the future.

17.
Immunol Cell Biol ; 90(5): 492-7, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22391814

RESUMEN

Production of type I interferon (IFN-α/ß) is a common cellular response to virus infection. IFN-α/ß has a dual role in combating infection, triggering innate antiviral mechanisms and stimulating the generation of an adaptive immune response. This review focuses on the effects of IFN-α/ß on one particular immune cell type, the T cell, and the impact of IFN-α/ß-mediated signalling in T cells on the immune response. The critical role of T-cell responsiveness to IFN-α/ß for the generation of productive T-cell responses after infections with certain viruses in vivo is discussed in the context of in vitro experiments investigating the mechanisms by which IFN-α/ß modifies T-cell function. These studies reveal complex effects of IFN-α/ß on T cells, with the consequences of exposure to IFN-α/ß depending on the context of other signals received by the T cell.


Asunto(s)
Interferón Tipo I/inmunología , Linfocitos T/inmunología , Virosis/inmunología , Inmunidad Adaptativa , Animales , Microambiente Celular , Humanos , Inmunidad Innata , Receptor Cross-Talk/inmunología , Transducción de Señal/inmunología
18.
Hepatology ; 53(4): 1154-63, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21480323

RESUMEN

UNLABELLED: Therapy of chronic hepatitis C with pegylated interferon α (pegIFN-α) and ribavirin achieves sustained virological responses in approximately half of the patients. Nonresponse to treatment is associated with constitutively increased expression of IFN-stimulated genes in the liver already before therapy. This activation of the endogenous IFN system could prevent cells from responding to therapeutically injected (peg)IFN-α, because prolonged stimulation of cells with IFN-α induces desensitization of the IFN signal transduction pathway. Whether all types of IFNs induce refractoriness in the liver is presently unknown. We therefore treated mice with multiple injections and different combinations of IFN-α, IFN-ß, IFN-γ, and IFN-λ. Pretreatment of mice with IFN-α, IFN-ß, and IFN-λ induced a strong expression of the negative regulator ubiquitin-specific peptidase 18 in the liver and gut. As a result, IFN-α signaling was significantly reduced when mice where reinjected 16 hours after the first injection. Surprisingly, both IFN-ß and IFN-λ could activate the Janus kinase-signal transducer and activator of transcription (STAT) pathway and the expression of IFN-stimulated genes despite high levels of ubiquitin-specific peptidase 18. IFN-λ treatment of human liver biopsies ex vivo resulted in strong and maintained phosphorylation of STAT1, whereas IFN-α-induced STAT1 activation was transient. CONCLUSION: Contrary to the action of IFN-α, IFN-ß, and IFN-λ signaling in the liver does not become refractory during repeated stimulation of the IFN signal transduction pathway. The sustained efficacy of IFN-ß and IFN-λ could be an important advantage for the treatment patients who are nonresponders to pegIFN-α, through a preactivated endogenous IFN system.


Asunto(s)
Citocinas/fisiología , Interferón-alfa/farmacología , Interferón beta/farmacología , Interferón gamma/farmacología , Hígado/efectos de los fármacos , Transducción de Señal/fisiología , Animales , Línea Celular Tumoral , Endopeptidasas/metabolismo , Hepatitis C Crónica/fisiopatología , Humanos , Inductores de Interferón/farmacología , Intestino Delgado/efectos de los fármacos , Quinasas Janus/metabolismo , Hígado/fisiología , Masculino , Ratones , Factor de Transcripción STAT1/metabolismo , Transducción de Señal/efectos de los fármacos , Ubiquitina Tiolesterasa
19.
Nat Genet ; 54(6): 817-826, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35618845

RESUMEN

During activation, T cells undergo extensive gene expression changes that shape the properties of cells to exert their effector function. Understanding the regulation of this process could help explain how genetic variants predispose to immune diseases. Here, we mapped genetic effects on gene expression (expression quantitative trait loci (eQTLs)) using single-cell transcriptomics. We profiled 655,349 CD4+ T cells, capturing transcriptional states of unstimulated cells and three time points of cell activation in 119 healthy individuals. This identified 38 cell clusters, including transient clusters that were only present at individual time points of activation. We found 6,407 genes whose expression was correlated with genetic variation, of which 2,265 (35%) were dynamically regulated during activation. Furthermore, 127 genes were regulated by variants associated with immune-mediated diseases, with significant enrichment for dynamic effects. Our results emphasize the importance of studying context-specific gene expression regulation and provide insights into the mechanisms underlying genetic susceptibility to immune-mediated diseases.


Asunto(s)
Enfermedades del Sistema Inmune , Sitios de Carácter Cuantitativo , Linfocitos T CD4-Positivos , Regulación de la Expresión Génica/genética , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Humanos , Enfermedades del Sistema Inmune/genética , Polimorfismo de Nucleótido Simple , Sitios de Carácter Cuantitativo/genética , Transcriptoma
20.
Nat Commun ; 13(1): 2885, 2022 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-35610203

RESUMEN

Myeloid cells are central to homeostasis and immunity. Characterising in vitro myelopoiesis protocols is imperative for their use in research, immunotherapies, and understanding human myelopoiesis. Here, we generate a >470K cells molecular map of human induced pluripotent stem cells (iPSC) differentiation into macrophages. Integration with in vivo single-cell atlases shows in vitro differentiation recapitulates features of yolk sac hematopoiesis, before definitive hematopoietic stem cells (HSC) emerge. The diversity of myeloid cells generated, including mast cells and monocytes, suggests that HSC-independent hematopoiesis can produce multiple myeloid lineages. We uncover poorly described myeloid progenitors and conservation between in vivo and in vitro regulatory programs. Additionally, we develop a protocol to produce iPSC-derived dendritic cells (DC) resembling cDC2. Using CRISPR/Cas9 knock-outs, we validate the effects of key transcription factors in macrophage and DC ontogeny. This roadmap of myeloid differentiation is an important resource for investigating human fetal hematopoiesis and new therapeutic opportunities.


Asunto(s)
Células Madre Pluripotentes Inducidas , Mielopoyesis , Diferenciación Celular/genética , Linaje de la Célula/genética , Genómica , Hematopoyesis/genética , Humanos , Mielopoyesis/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA