Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Ann Rheum Dis ; 82(9): 1142-1152, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37344156

RESUMEN

INTRODUCTION: Structural reorganisation of the synovium with expansion of fibroblast-like synoviocytes (FLS) and influx of immune cells is a hallmark of rheumatoid arthritis (RA). Activated FLS are increasingly recognised as a critical component driving synovial tissue remodelling by interacting with immune cells resulting in distinct synovial pathotypes of RA. METHODS: Automated high-content fluorescence microscopy of co-cultured cytokine-activated FLS and autologous peripheral CD4+ T cells from patients with RA was established to quantify cell-cell interactions. Phenotypic profiling of cytokine-treated FLS and co-cultured T cells was done by flow cytometry and RNA-Seq, which were integrated with publicly available transcriptomic data from patients with different histological synovial pathotypes. Computational prediction and knock-down experiments were performed in FLS to identify adhesion molecules for cell-cell interaction. RESULTS: Cytokine stimulation, especially with TNF-α, led to enhanced FLS-T cell interaction resulting in cell-cell contact-dependent activation, proliferation and differentiation of T cells. Signatures of cytokine-activated FLS were significantly enriched in RA synovial tissues defined as lymphoid-rich or leucocyte-rich pathotypes, with the most prominent effects for TNF-α. FLS cytokine signatures correlated with the number of infiltrating CD4+ T cells in synovial tissue of patients with RA. Ligand-receptor pair interaction analysis identified ICAM1 on FLS as an important mediator in TNF-mediated FLS-T cell interaction. Both, ICAM1 and its receptors were overexpressed in TNF-treated FLS and co-cultured T cells. Knock-down of ICAM1 in FLS resulted in reduced TNF-mediated FLS-T cell interaction. CONCLUSION: Our study highlights the role of cytokine-activated FLS in orchestrating inflammation-associated synovial pathotypes providing novel insights into disease mechanisms of RA.


Asunto(s)
Artritis Reumatoide , Sinoviocitos , Humanos , Citocinas , Factor de Necrosis Tumoral alfa/farmacología , Membrana Sinovial/patología , Sinoviocitos/patología , Fibroblastos/patología , Células Cultivadas
2.
Front Immunol ; 14: 1096096, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37033944

RESUMEN

Various autoimmune diseases are characterized by distinct cell subset distributions and activation profiles of peripheral blood mononuclear cells (PBMCs). PBMCs can therefore serve as an ideal biomarker material, which is easily accessible and allows for screening of multiple cell types. A detailed understanding of the immune landscape is critical for the diagnosis of patients with autoimmune diseases, as well as for a personalized treatment approach. In our study, we investigate the potential of multi-parameter spectral flow cytometry for the identification of patients suffering from autoimmune diseases and its power as an evaluation tool for in vitro drug screening approaches (advanced immunophenotyping). We designed a combination of two 22-color immunophenotyping panels for profiling cell subset distribution and cell activation. Downstream bioinformatics analyses included percentages of individual cell populations and median fluorescent intensity of defined markers which were then visualized as heatmaps and in dimensionality reduction approaches. In vitro testing of epigenetic immunomodulatory drugs revealed an altered activation status upon treatment, which supports the use of spectral flow cytometry as a high-throughput drug screening tool. Advanced immunophenotyping might support the exploration of novel therapeutic drugs and contribute to future personalized treatment approaches in autoimmune diseases and beyond.


Asunto(s)
Enfermedades Autoinmunes , Leucocitos Mononucleares , Humanos , Inmunofenotipificación , Medicina de Precisión , Evaluación Preclínica de Medicamentos , Enfermedades Autoinmunes/diagnóstico , Enfermedades Autoinmunes/tratamiento farmacológico
3.
Immunol Lett ; 247: 27-42, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35568324

RESUMEN

The two major subsets of peripheral T cells are classically divided into the CD4+ T helper cells and the cytotoxic CD8+ T cell lineage. However, the appearance of some effector CD4+ T cell populations displaying cytotoxic activity, in particular during viral infections, has been observed, thus breaking the functional dichotomy of CD4+ and CD8+ T lymphocytes. The strong association of the appearance of CD4+ cytotoxic T lymphocytes (CD4 CTLs) with viral infections suggests an important role of this subset in antiviral immunity by controlling viral replication and infection. Moreover, CD4 CTLs have been linked with anti-tumor activity and might also cause immunopathology in autoimmune diseases. This raises interest into the molecular mechanisms regulating CD4 CTL differentiation, which are poorly understood in comparison to differentiation pathways of other Th subsets. In this review, we provide a brief overview about key features of CD4 CTLs, including their role in viral infections and cancer immunity, and about the link between CD4 CTLs and immune-mediated diseases. Subsequently, we will discuss the current knowledge about transcriptional and epigenetic networks controlling CD4 CTL differentiation and highlight recent data suggesting a role for histone deacetylases in the generation of CD4 CTLs.


Asunto(s)
Linfocitos T CD4-Positivos , Linfocitos T Citotóxicos , Redes Reguladoras de Genes , Fenotipo , Linfocitos T Colaboradores-Inductores
4.
J Hepatol ; 75(5): 1164-1176, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34242699

RESUMEN

BACKGROUND & AIMS: 24-Norursodeoxycholic acid (NorUDCA) is a novel therapeutic bile acid used to treat immune-mediated cholestatic liver diseases, such as primary sclerosing cholangitis (PSC), where dysregulated T cells including CD8+ T cells contribute to hepatobiliary immunopathology. We hypothesized that NorUDCA may directly modulate CD8+ T cell function thus contributing to its therapeutic efficacy. METHODS: NorUDCA's immunomodulatory effects were first studied in Mdr2-/- mice, as a cholestatic model of PSC. To differentiate NorUDCA's immunomodulatory effects on CD8+ T cell function from its anticholestatic actions, we also used a non-cholestatic model of hepatic injury induced by an excessive CD8+ T cell immune response upon acute non-cytolytic lymphocytic choriomeningitis virus (LCMV) infection. Studies included molecular and biochemical approaches, flow cytometry and metabolic assays in murine CD8+ T cells in vitro. Mass spectrometry was used to identify potential CD8+ T cell targets modulated by NorUDCA. The signaling effects of NorUDCA observed in murine cells were validated in circulating T cells from patients with PSC. RESULTS: NorUDCA demonstrated immunomodulatory effects by reducing hepatic innate and adaptive immune cells, including CD8+ T cells in the Mdr2-/- model. In the non-cholestatic model of CD8+ T cell-driven immunopathology induced by acute LCMV infection, NorUDCA ameliorated hepatic injury and systemic inflammation. Mechanistically, NorUDCA demonstrated strong immunomodulatory efficacy in CD8+ T cells affecting lymphoblastogenesis, expansion, glycolysis and mTORC1 signaling. Mass spectrometry identified that NorUDCA regulates CD8+ T cells by targeting mTORC1. NorUDCA's impact on mTORC1 signaling was further confirmed in circulating PSC CD8+ T cells. CONCLUSIONS: NorUDCA has a direct modulatory impact on CD8+ T cells and attenuates excessive CD8+ T cell-driven hepatic immunopathology. These findings are relevant for treatment of immune-mediated liver diseases such as PSC. LAY SUMMARY: Elucidating the mechanisms by which 24-norursodeoxycholic acid (NorUDCA) works for the treatment of immune-mediated liver diseases, such as primary sclerosing cholangitis, is of considerable clinical interest. Herein, we uncovered an unrecognized property of NorUDCA in the immunometabolic regulation of CD8+ T cells, which has therapeutic relevance for immune-mediated liver diseases, including PSC.


Asunto(s)
Linfocitos T CD8-positivos/metabolismo , Inflamación/tratamiento farmacológico , Hígado/efectos de los fármacos , Ácido Ursodesoxicólico/análogos & derivados , Animales , Linfocitos T CD8-positivos/efectos de los fármacos , Modelos Animales de Enfermedad , Inflamación/fisiopatología , Hígado/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ácido Ursodesoxicólico/farmacología , Ácido Ursodesoxicólico/uso terapéutico
5.
J Autoimmun ; 119: 102610, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33621930

RESUMEN

CD4+ T cell trafficking is a fundamental property of adaptive immunity. In this study, we uncover a novel role for histone deacetylase 1 (HDAC1) in controlling effector CD4+ T cell migration, thereby providing mechanistic insight into why a T cell-specific deletion of HDAC1 protects against experimental autoimmune encephalomyelitis (EAE). HDAC1-deficient CD4+ T cells downregulated genes associated with leukocyte extravasation. In vitro, HDAC1-deficient CD4+ T cells displayed aberrant morphology and migration on surfaces coated with integrin LFA-1 ligand ICAM-1 and showed an impaired ability to arrest on and to migrate across a monolayer of primary mouse brain microvascular endothelial cells under physiological flow. Moreover, HDAC1 deficiency reduced homing of CD4+ T cells into the intestinal epithelium and lamina propria preventing weight-loss, crypt damage and intestinal inflammation in adoptive CD4+ T cell transfer colitis. This correlated with reduced expression levels of LFA-1 integrin chains CD11a and CD18 as well as of selectin ligands CD43, CD44 and CD162 on transferred circulating HDAC1-deficient CD4+ T cells. Our data reveal that HDAC1 controls T cell-mediated autoimmunity via the regulation of CD4+ T cell trafficking into the CNS and intestinal tissues.


Asunto(s)
Autoinmunidad , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Quimiotaxis de Leucocito/inmunología , Histona Desacetilasa 1/metabolismo , Inflamación/etiología , Inflamación/metabolismo , Animales , Biomarcadores , Adhesión Celular , Quimiotaxis de Leucocito/genética , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Encefalomielitis Autoinmune Experimental/diagnóstico , Encefalomielitis Autoinmune Experimental/etiología , Encefalomielitis Autoinmune Experimental/metabolismo , Células Endoteliales , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Histona Desacetilasa 1/genética , Inmunohistoquímica , Inflamación/diagnóstico , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Ratones , Ratones Noqueados
6.
Cell Rep ; 30(9): 3171-3182.e6, 2020 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-32130916

RESUMEN

Protein phosphatase 2A (PP2A) is an important regulator of signal transduction pathways and a tumor suppressor. Phosphorylation of the PP2A catalytic subunit (PP2AC) at tyrosine 307 has been claimed to inactivate PP2A and was examined in more than 180 studies using commercial antibodies, but this modification was never identified using mass spectrometry. Here we show that the most cited pTyr307 monoclonal antibodies, E155 and F-8, are not specific for phosphorylated Tyr307 but instead are hampered by PP2AC methylation at leucine 309 or phosphorylation at threonine 304. Other pTyr307 antibodies are sensitive to PP2AC methylation as well, and some cross-react with pTyr residues in general, including phosphorylated hemagglutinin tags. We identify pTyr307 using targeted mass spectrometry after transient overexpression of PP2AC and Src kinase. Yet under such conditions, none of the tested antibodies show exclusive pTyr307 specificity. Thus, data generated using these antibodies need to be revisited, and the mechanism of PP2A inactivation needs to be redefined.


Asunto(s)
Anticuerpos/metabolismo , Especificidad de Anticuerpos/inmunología , Leucina/metabolismo , Fosfotirosina/metabolismo , Proteína Fosfatasa 2/metabolismo , Procesamiento Proteico-Postraduccional , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/metabolismo , Especificidad de Anticuerpos/efectos de los fármacos , Reacciones Cruzadas/efectos de los fármacos , Factor de Crecimiento Epidérmico/farmacología , Células HEK293 , Humanos , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Metilación , Ratones , Ratones Endogámicos C57BL , Células 3T3 NIH , Péptidos/química , Péptidos/metabolismo , Fosforilación/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Vanadatos/farmacología , Familia-src Quinasas/metabolismo
7.
JCI Insight ; 5(4)2020 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-32102981

RESUMEN

Some effector CD4+ T cell subsets display cytotoxic activity, thus breaking the functional dichotomy of CD4+ helper and CD8+ cytotoxic T lymphocytes. However, molecular mechanisms regulating CD4+ cytotoxic T lymphocyte (CD4+ CTL) differentiation are poorly understood. Here we show that levels of histone deacetylases 1 and 2 (HDAC1-HDAC2) are key determinants of CD4+ CTL differentiation. Deletions of both Hdac1 and 1 Hdac2 alleles (HDAC1cKO-HDAC2HET) in CD4+ T cells induced a T helper cytotoxic program that was controlled by IFN-γ-JAK1/2-STAT1 signaling. In vitro, activated HDAC1cKO-HDAC2HET CD4+ T cells acquired cytolytic activity and displayed enrichment of gene signatures characteristic of effector CD8+ T cells and human CD4+ CTLs. In vivo, murine cytomegalovirus-infected HDAC1cKO-HDAC2HET mice displayed a stronger induction of CD4+ CTL features compared with infected WT mice. Finally, murine and human CD4+ T cells treated with short-chain fatty acids, which are commensal-produced metabolites acting as HDAC inhibitors, upregulated CTL genes. Our data demonstrate that HDAC1-HDAC2 restrain CD4+ CTL differentiation. Thus, HDAC1-HDAC2 might be targets for the therapeutic induction of CD4+ CTLs.


Asunto(s)
Linfocitos T CD4-Positivos/citología , Diferenciación Celular/fisiología , Histona Desacetilasa 1/fisiología , Histona Desacetilasa 2/fisiología , Linfocitos T Citotóxicos/fisiología , Animales , Linfocitos T CD4-Positivos/efectos de los fármacos , Ácidos Grasos/farmacología , Histona Desacetilasa 1/genética , Histona Desacetilasa 2/genética , Humanos , Ratones , Ratones Noqueados , Transducción de Señal/fisiología , Linfocitos T Citotóxicos/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología
8.
J Autoimmun ; 108: 102379, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31883829

RESUMEN

Rheumatoid Arthritis (RA) represents a chronic T cell-mediated inflammatory autoimmune disease. Studies have shown that epigenetic mechanisms contribute to the pathogenesis of RA. Histone deacetylases (HDACs) represent one important group of epigenetic regulators. However, the role of individual HDAC members for the pathogenesis of arthritis is still unknown. In this study we demonstrate that mice with a T cell-specific deletion of HDAC1 (HDAC1-cKO) are resistant to the development of Collagen-induced arthritis (CIA), whereas the antibody response to collagen type II was undisturbed, indicating an unaltered T cell-mediated B cell activation. The inflammatory cytokines IL-17 and IL-6 were significantly decreased in sera of HDAC1-cKO mice. IL-6 treated HDAC1-deficient CD4+ T cells showed an impaired upregulation of CCR6. Selective inhibition of class I HDACs with the HDAC inhibitor MS-275 under Th17-skewing conditions inhibited the upregulation of chemokine receptor 6 (CCR6) in mouse and human CD4+ T cells. Accordingly, analysis of human RNA-sequencing (RNA-seq) data and histological analysis of synovial tissue samples from human RA patients revealed the existence of CD4+CCR6+ cells with enhanced HDAC1 expression. Our data indicate a key role for HDAC1 for the pathogenesis of CIA and suggest that HDAC1 and other class I HDACs might be promising targets of selective HDAC inhibitors (HDACi) for the treatment of RA.


Asunto(s)
Artritis Reumatoide/etiología , Artritis Reumatoide/metabolismo , Susceptibilidad a Enfermedades , Histona Desacetilasa 1/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Animales , Artritis Reumatoide/patología , Biomarcadores , Colágeno/efectos adversos , Citocinas/metabolismo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Histona Desacetilasa 1/genética , Humanos , Mediadores de Inflamación/metabolismo , Ratones , Ratones Noqueados , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
9.
Cell Rep ; 29(13): 4447-4459.e6, 2019 12 24.
Artículo en Inglés | MEDLINE | ID: mdl-31875552

RESUMEN

Forkhead box protein P3+ (FOXP3+) regulatory T cells (Treg cells) play a key role in maintaining tolerance and immune homeostasis. Here, we report that a T cell-specific deletion of the transcription factor MAZR (also known as PATZ1) leads to an increased frequency of Treg cells, while enforced MAZR expression impairs Treg cell differentiation. Further, MAZR expression levels are progressively downregulated during thymic Treg cell development and during in-vitro-induced human Treg cell differentiation, suggesting that MAZR protein levels are critical for controlling Treg cell development. However, MAZR-deficient Treg cells show only minor transcriptional changes ex vivo, indicating that MAZR is not essential for establishing the transcriptional program of peripheral Treg cells. Finally, the loss of MAZR reduces the clinical score in dextran-sodium sulfate (DSS)-induced colitis, suggesting that MAZR activity in T cells controls the extent of intestinal inflammation. Together, these data indicate that MAZR is part of a Treg cell-intrinsic transcriptional network that modulates Treg cell development.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Represoras/metabolismo , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/metabolismo , Animales , Diferenciación Celular , Colitis/inmunología , Sulfato de Dextran , Humanos , Ratones Noqueados , Timo/citología , Transcripción Genética
10.
Front Immunol ; 10: 409, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30915074

RESUMEN

CD8 expression in T lymphocytes is tightly regulated by the activity of at least six Cd8 enhancers (E8I-E8VI), however their complex developmental stage-, subset-, and lineage-specific interplays are incompletely understood. Here we analyzed ATAC-seq data on the Immunological Genome Project database and identified a similar developmental regulation of chromatin accessibility of a subregion of E8I, designated E8I-core, and of E8VI. Loss of E8I-core led to a similar reduction in CD8 expression in naïve CD8+ T cells and in IELs as observed in E8I-/- mice, demonstrating that we identified the core enhancer region of E8I. While E8VI-/- mice displayed a mild reduction in CD8 expression levels on CD8SP thymocytes and peripheral CD8+ T cells, CD8 levels were further reduced upon combined deletion of E8I-core and E8VI. Moreover, activated E8I-core-/-E8VI-/- CD8+ T cells lost CD8 expression to a greater degree than E8I-core-/- and E8VI-/- CD8+ T cells, suggesting that the combined activity of both enhancers is required for establishment and maintenance of CD8 expression before and after TCR activation. Finally, we observed a severe reduction of CD4 CTLs among the TCRß+CD4+ IEL population in E8I-core-/- but not E8VI-/- mice. Such a reduction was not observed in Cd8a-/- mice, indicating that E8I-core controls the generation of CD4 CTLs independently of its role in Cd8a gene regulation. Further, the combined deletion of E8I-core and E8VI restored CD4 CTL subsets, suggesting an antagonistic function of E8VI in the generation of CD4 CTLs. Together, our study demonstrates a complex utilization and interplay of E8I-core and E8VI in regulating CD8 expression in cytotoxic lineage T cells and in IELs. Moreover, we revealed a novel E8I-mediated regulatory mechanism controlling the generation of intestinal CD4 CTLs.


Asunto(s)
Antígenos CD8/biosíntesis , Regulación de la Expresión Génica/inmunología , Mucosa Intestinal/inmunología , Linfocitos Intraepiteliales/inmunología , Linfocitos T Citotóxicos/inmunología , Animales , Linfocitos Intraepiteliales/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Linfocitos T Citotóxicos/metabolismo
11.
J Autoimmun ; 86: 51-61, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28964722

RESUMEN

Multiple sclerosis (MS) is a human neurodegenerative disease characterized by the invasion of autoreactive T cells from the periphery into the CNS. Application of pan-histone deacetylase inhibitors (HDACi) ameliorates experimental autoimmune encephalomyelitis (EAE), an animal model for MS, suggesting that HDACi might be a potential therapeutic strategy for MS. However, the function of individual HDAC members in the pathogenesis of EAE is not known. In this study we report that mice with a T cell-specific deletion of HDAC1 (using the Cd4-Cre deleter strain; HDAC1-cKO) were completely resistant to EAE despite the ability of HDAC1cKO CD4+ T cells to differentiate into Th17 cells. RNA sequencing revealed STAT1 as a prominent upstream regulator of differentially expressed genes in activated HDAC1-cKO CD4+ T cells and this was accompanied by a strong increase in phosphorylated STAT1 (pSTAT1). This suggests that HDAC1 controls STAT1 activity in activated CD4+ T cells. Increased pSTAT1 levels correlated with a reduced expression of the chemokine receptors Ccr4 and Ccr6, which are important for the migration of T cells into the CNS. Finally, EAE susceptibility was restored in WT:HDAC1-cKO mixed BM chimeric mice, indicating a cell-autonomous defect. Our data demonstrate a novel pathophysiological role for HDAC1 in EAE and provide evidence that selective inhibition of HDAC1 might be a promising strategy for the treatment of MS.


Asunto(s)
Encefalomielitis Autoinmune Experimental/metabolismo , Histona Desacetilasa 1/metabolismo , Esclerosis Múltiple/metabolismo , Factor de Transcripción STAT1/metabolismo , Células Th17/fisiología , Animales , Movimiento Celular , Células Cultivadas , Quimera , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inmunología , Histona Desacetilasa 1/genética , Humanos , Ratones , Ratones Noqueados , Esclerosis Múltiple/inmunología , Receptores CCR4/metabolismo , Receptores CCR6/metabolismo , Factor de Transcripción STAT1/genética
12.
Sci Rep ; 7(1): 15928, 2017 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-29162920

RESUMEN

Nuclear receptor corepressor 1 (NCOR1) is a transcriptional regulator bridging repressive chromatin modifying enzymes with transcription factors. NCOR1 regulates many biological processes, however its role in T cells is not known. Here we show that Cd4-Cre-mediated deletion of NCOR1 (NCOR1 cKOCd4) resulted in a reduction of peripheral T cell numbers due to a decrease in single-positive (SP) thymocytes. In contrast, double-positive (DP) thymocyte numbers were not affected in the absence of NCOR1. The reduction in SP cells was due to diminished survival of NCOR1-null postselection TCRßhiCD69+ and mature TCRßhiCD69- thymocytes. NCOR1-null thymocytes expressed elevated levels of the pro-apoptotic factor BIM and showed a higher fraction of cleaved caspase 3-positive cells upon TCR stimulation ex vivo. However, staphylococcal enterotoxin B (SEB)-mediated deletion of Vß8+ CD4SP thymocytes was normal, suggesting that negative selection is not altered in the absence of NCOR1. Finally, transgenic expression of the pro-survival protein BCL2 restored the population of CD69+ thymocytes in NCOR1 cKOCd4 mice to a similar percentage as observed in WT mice. Together, these data identify NCOR1 as a crucial regulator of the survival of SP thymocytes and revealed that NCOR1 is essential for the proper generation of the peripheral T cell pool.


Asunto(s)
Co-Represor 1 de Receptor Nuclear/metabolismo , Timocitos/citología , Timocitos/metabolismo , Animales , Supervivencia Celular , Eliminación de Gen , Subunidad alfa del Receptor de Interleucina-7/metabolismo , Recuento de Linfocitos , Ratones Noqueados , Co-Represor 1 de Receptor Nuclear/deficiencia , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...