Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Sci Immunol ; 7(68): eabi4919, 2022 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-35179948

RESUMEN

The response of naive CD8+ T cells to their cognate antigen involves rapid and broad changes to gene expression that are coupled with extensive chromatin remodeling, but the mechanisms governing these changes are not fully understood. Here, we investigated how these changes depend on the basic leucine zipper ATF-like transcription factor Batf, which is essential for the early phases of the process. Through genome scale profiling, we characterized the role of Batf in chromatin organization at several levels, including the accessibility of key regulatory regions, the expression of their nearby genes, and the interactions that these regions form with each other and with key transcription factors. We identified a core network of transcription factors that cooperated with Batf, including Irf4, Runx3, and T-bet, as indicated by their colocalization with Batf and their binding in regions whose accessibility, interactions, and expression of nearby genes depend on Batf. We demonstrated the synergistic activity of this network by overexpressing the different combinations of these genes in fibroblasts. Batf and Irf4, but not Batf alone, were sufficient to increase accessibility and transcription of key loci, normally associated with T cell function. Addition of Runx3 and T-bet further contributed to fine-tuning of these changes and was essential for establishing chromatin loops characteristic of T cells. These data provide a resource for studying the epigenomic and transcriptomic landscape of effector differentiation of cytotoxic T cells and for investigating the interdependency between transcription factors and its effects on the epigenome and transcriptome of primary cells.


Asunto(s)
Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/inmunología , Linfocitos T CD8-positivos/inmunología , Diferenciación Celular/inmunología , Subunidad alfa 3 del Factor de Unión al Sitio Principal/inmunología , Factores Reguladores del Interferón/inmunología , Proteínas de Dominio T Box/inmunología , Animales , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/deficiencia , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Subunidad alfa 3 del Factor de Unión al Sitio Principal/genética , Epigénesis Genética/genética , Femenino , Factores Reguladores del Interferón/genética , Ratones , Ratones Noqueados , Ratones Transgénicos , Proteínas de Dominio T Box/genética
2.
Front Immunol ; 12: 535039, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33815354

RESUMEN

The BTB zinc finger transcription factor MAZR (also known as PATZ1) controls, partially in synergy with the transcription factor Runx3, the development of CD8 lineage T cells. Here we explored the role of MAZR as well as combined activities of MAZR/Runx3 during cytotoxic T lymphocyte (CTL) and memory CD8+ T cell differentiation. In contrast to the essential role of Runx3 for CTL effector function, the deletion of MAZR had a mild effect on the generation of CTLs in vitro. However, a transcriptome analysis demonstrated that the combined deletion of MAZR and Runx3 resulted in much more widespread downregulation of CTL signature genes compared to single Runx3 deletion, indicating that MAZR partially compensates for loss of Runx3 in CTLs. Moreover, in line with the findings made in vitro, the analysis of CTL responses to LCMV infection revealed that MAZR and Runx3 cooperatively regulate the expression of CD8α, Granzyme B and perforin in vivo. Interestingly, while memory T cell differentiation is severely impaired in Runx3-deficient mice, the deletion of MAZR leads to an enlargement of the long-lived memory subset and also partially restored the differentiation defect caused by loss of Runx3. This indicates distinct functions of MAZR and Runx3 in the generation of memory T cell subsets, which is in contrast to their cooperative roles in CTLs. Together, our study demonstrates complex interplay between MAZR and Runx3 during CTL and memory T cell differentiation, and provides further insight into the molecular mechanisms underlying the establishment of CTL and memory T cell pools.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Subunidad alfa 3 del Factor de Unión al Sitio Principal/inmunología , Memoria Inmunológica/inmunología , Proteínas de Neoplasias/inmunología , Proteínas Represoras/inmunología , Linfocitos T Citotóxicos/inmunología , Animales , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/virología , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Subunidad alfa 3 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 3 del Factor de Unión al Sitio Principal/metabolismo , Regulación de la Expresión Génica/inmunología , Interacciones Huésped-Patógeno/inmunología , Activación de Linfocitos/inmunología , Coriomeningitis Linfocítica/inmunología , Coriomeningitis Linfocítica/metabolismo , Coriomeningitis Linfocítica/virología , Virus de la Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica/fisiología , Ratones Noqueados , Ratones Transgénicos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Linfocitos T Citotóxicos/metabolismo , Linfocitos T Citotóxicos/virología
3.
Proc Natl Acad Sci U S A ; 118(4)2021 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-33479171

RESUMEN

Runt domain-related (Runx) transcription factors are essential for early T cell development in mice from uncommitted to committed stages. Single and double Runx knockouts via Cas9 show that target genes responding to Runx activity are not solely controlled by the dominant factor, Runx1. Instead, Runx1 and Runx3 are coexpressed in single cells; bind to highly overlapping genomic sites; and have redundant, collaborative functions regulating genes pivotal for T cell development. Despite stable combined expression levels across pro-T cell development, Runx1 and Runx3 preferentially activate and repress genes that change expression dynamically during lineage commitment, mostly activating T-lineage genes and repressing multipotent progenitor genes. Furthermore, most Runx target genes are sensitive to Runx perturbation only at one stage and often respond to Runx more for expression transitions than for maintenance. Contributing to this highly stage-dependent gene regulation function, Runx1 and Runx3 extensively shift their binding sites during commitment. Functionally distinct Runx occupancy sites associated with stage-specific activation or repression are also distinguished by different patterns of partner factor cobinding. Finally, Runx occupancies change coordinately at numerous clustered sites around positively or negatively regulated targets during commitment. This multisite binding behavior may contribute to a developmental "ratchet" mechanism making commitment irreversible.


Asunto(s)
Linaje de la Célula/inmunología , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 3 del Factor de Unión al Sitio Principal/genética , Células Precursoras de Linfocitos T/inmunología , Linfocitos T/inmunología , Transcriptoma , Animales , Diferenciación Celular , Linaje de la Célula/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/inmunología , Subunidad alfa 3 del Factor de Unión al Sitio Principal/inmunología , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Subunidad alfa del Receptor de Interleucina-2/genética , Subunidad alfa del Receptor de Interleucina-2/inmunología , Masculino , Ratones , Células Precursoras de Linfocitos T/citología , Cultivo Primario de Células , Proteínas Represoras/genética , Proteínas Represoras/inmunología , Linfocitos T/clasificación , Linfocitos T/citología , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/inmunología
4.
Aging (Albany NY) ; 12(24): 26095-26120, 2020 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-33401247

RESUMEN

The heterogeneity and complexity of tumor-immune microenvironments lead to diverse immunotherapy effects among colon cancer patients. It is crucial to identify immune microenvironment-related biomarkers and construct prognostic risk models. In this study, the immune and stromal scores of 415 cases from TCGA were calculated using the ESTIMATE algorithm. AXIN2, CCL22, CLEC10A, CRIP2, RUNX3, and TRPM5 were screened and established a prognostic immune-related gene (IRG) signature using by univariate, LASSO, and multivariate Cox regression models. The predicted performance of IRG signature was external validated by GSE39582 (n=519). Stratified survival analysis showed IRG signature was an effective predictor of survival in patients with different clinical characteristics. The protein expression level of six genes was validated by immunohistochemistry analysis. Difference analysis indicated the mutation rate, immune cell of resting NK cells and regulatory T cells infiltration and four immune checkpoints of PD-1, PD-L1, LAG3 and VSIR expression levels in the high-risk group were significantly higher than those in the low-risk group. A nomogram incorporating the gene signatures and clinical factors was demonstrated had a good accuracy (1-, 3-, and 5-year AUC= 0.799, 0.791, 0.738). Our study identified a novel IRG signature, which may provide some references for the clinical precision immunotherapy of patients.


Asunto(s)
Adenocarcinoma/genética , Neoplasias del Colon/genética , Transcriptoma , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/inmunología , Adenocarcinoma/inmunología , Adenocarcinoma/mortalidad , Adenocarcinoma/patología , Anciano , Antígenos CD/genética , Antígenos CD/inmunología , Proteína Axina/genética , Proteína Axina/inmunología , Antígenos B7/genética , Antígenos B7/inmunología , Antígeno B7-H1/genética , Antígeno B7-H1/inmunología , Quimiocina CCL22/genética , Quimiocina CCL22/inmunología , Neoplasias del Colon/inmunología , Neoplasias del Colon/mortalidad , Neoplasias del Colon/patología , Subunidad alfa 3 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 3 del Factor de Unión al Sitio Principal/inmunología , Bases de Datos Genéticas , Femenino , Humanos , Células Asesinas Naturales , Proteínas con Dominio LIM/genética , Proteínas con Dominio LIM/inmunología , Lectinas Tipo C/genética , Lectinas Tipo C/inmunología , Linfocitos Infiltrantes de Tumor , Masculino , Persona de Mediana Edad , Análisis Multivariante , Nomogramas , Pronóstico , Receptor de Muerte Celular Programada 1/genética , Receptor de Muerte Celular Programada 1/inmunología , Modelos de Riesgos Proporcionales , Tasa de Supervivencia , Linfocitos T Reguladores , Canales Catiónicos TRPM/genética , Canales Catiónicos TRPM/inmunología , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología , Proteína del Gen 3 de Activación de Linfocitos
5.
Digestion ; 101(2): 156-164, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-30763934

RESUMEN

BACKGROUND/AIMS: Runt-related transcription factor (RUNX) 3 is a tumor suppressor whose expression is reduced in non-neoplastic rectal mucosa of patients with ulcerative colitis (UC) with coexisting colitis-associated cancer (CAC). We aimed to evaluate RUNX3 utility as a predictive marker for CAC using immunohistochemistry (IHC) for non-neoplastic UC mucosa. METHODS: We retrospectively compared the RUNX3 expression detected by IHC between non-neoplastic rectal biopsy specimens from 20 cases with invasive cancer (CAC group) and 20 cases selected from 138 patients without CAC (non-CAC group) that were treated during the same period (2006-2017) and were matched for sex, duration, extension, and age. We validated the results using tissue microarrays (TMA) of 44 operated cases with CAC. The RUNX3 expression level was determined by calculating the percentage of RUNX3-positive-cells. RESULTS: The RUNX3 expression was lower in the CAC than that in the non-CAC group (35.6 vs. 70.7%, p = 0.03). For a cutoff value of 58%, the sensitivity and specificity for predicting CAC were 75.0 and 70.0% respectively. The immunostaining results for the TMA showed the same trend; 74% of cases with CAC were negative for the RUNX3 expression. CONCLUSION: RUNX3 immunostaining of non-neoplastic mucosa is useful for identifying UC patients at a high risk of developing CAC.


Asunto(s)
Colitis Ulcerosa/genética , Neoplasias Colorrectales/genética , Subunidad alfa 3 del Factor de Unión al Sitio Principal/metabolismo , Anciano , Biomarcadores de Tumor/inmunología , Biomarcadores de Tumor/metabolismo , Biopsia , Colitis Ulcerosa/inmunología , Neoplasias Colorrectales/inmunología , Subunidad alfa 3 del Factor de Unión al Sitio Principal/inmunología , Femenino , Predisposición Genética a la Enfermedad , Humanos , Inmunohistoquímica , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Masculino , Persona de Mediana Edad , Recto/inmunología , Recto/metabolismo , Valores de Referencia , Estudios Retrospectivos , Sensibilidad y Especificidad , Análisis de Matrices Tisulares
6.
Med Sci Monit ; 25: 7795-7807, 2019 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-31622282

RESUMEN

BACKGROUND The methylation status of RUNX3 promoter region, its impact on RUNX3 gene expression, and Th1/Th2 imbalance are unknown in bronchiolitis. This study aimed to explore the predictors of bronchiolitis developing into asthma. MATERIAL AND METHODS The methylation status of RUNX3 promoter was assessed using Illumina HiSeq platform method. The relative RUNX3 mRNA levels in PBMCs were measured by qRT-PCR. Serum IL-4 and IFN-γ concentrations were measured by ELISA. RESULTS A series of sites with significantly higher levels of methylation as compared to their corresponding controls were identified, including 24 sites in group Ba vs. group Cn, 13 sites in group Ba vs. group Ca, 7 sites in group Ba vs. group Bn, 16 sites in group Bn vs. group Cn, 11 sites in group Ca vs. group Cn, and 23 sites in group B vs. group C; P<0.05. The relative mRNA levels in group Ba were significantly lower than those in groups Cn, Ca, Bn; P<0.05. The serum IL-4 concentrations in group Ba were significantly higher than those in group Cn; P<0.05. The serum IFN-γ concentrations in group Ba were significantly lower than those in groups Cn, Ca, Bn; P<0.05. Correlation analysis showed that differentially methylated RUNX3 promoter region sites were significantly negatively correlated with levels of relative RUNX3 mRNA and IFN-γ, and were significantly positively correlated with IL-4 levels. CONCLUSIONS The methylation status of RUNX3 promoter region plays a role in Th1/Th2 imbalance by silencing RUNX3 gene expression, which can serve as predictive marker for the development of bronchiolitis into asthma.


Asunto(s)
Bronquiolitis/genética , Bronquiolitis/inmunología , Subunidad alfa 3 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 3 del Factor de Unión al Sitio Principal/inmunología , Metilación de ADN , Células TH1/inmunología , Células Th2/inmunología , Asma/genética , Asma/inmunología , Biomarcadores/análisis , Estudios de Casos y Controles , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Pronóstico , Regiones Promotoras Genéticas , ARN Mensajero/genética , ARN Mensajero/inmunología , Células TH1/metabolismo , Células Th2/metabolismo , Transcriptoma
7.
Nat Commun ; 10(1): 3306, 2019 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-31341159

RESUMEN

Memory CD8+ T cells have the ability to provide lifelong immunity against pathogens. Although memory features generally arise after challenge with a foreign antigen, naïve CD8 single positive (SP) thymocytes may acquire phenotypic and functional characteristics of memory cells in response to cytokines such as interleukin-4. This process is associated with the induction of the T-box transcription factor Eomesodermin (EOMES). However, the underlying molecular mechanisms remain ill-defined. Using epigenomic profiling, we show that these innate memory CD8SP cells acquire only a portion of the active enhancer repertoire of conventional memory cells. This reprograming is secondary to EOMES recruitment, mostly to RUNX3-bound enhancers. Furthermore, EOMES is found within chromatin-associated complexes containing BRG1 and promotes the recruitment of this chromatin remodelling factor. Also, the in vivo acquisition of EOMES-dependent program is BRG1-dependent. In conclusion, our results support a strong epigenetic basis for the EOMES-driven establishment of CD8+ T cell innate memory program.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Subunidad alfa 3 del Factor de Unión al Sitio Principal/fisiología , ADN Helicasas/fisiología , Epigénesis Genética , Memoria Inmunológica , Proteínas Nucleares/fisiología , Proteínas de Dominio T Box/metabolismo , Factores de Transcripción/fisiología , Animales , Subunidad alfa 3 del Factor de Unión al Sitio Principal/inmunología , Subunidad alfa 3 del Factor de Unión al Sitio Principal/metabolismo , ADN Helicasas/inmunología , ADN Helicasas/metabolismo , Femenino , Perfilación de la Expresión Génica , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Proteínas Nucleares/inmunología , Proteínas Nucleares/metabolismo , Proteínas de Dominio T Box/genética , Factores de Transcripción/inmunología , Factores de Transcripción/metabolismo
8.
Front Immunol ; 9: 2101, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30258450

RESUMEN

Innate lymphoid cells (ILCs) are the most recently identified family of the innate immune system and are hypothesized to modulate immune functions prior to the generation of adaptive immune responses. Subsets of ILCs reside in the mucosa and regulate immune responses to external pathogens; however, their role and the mechanism by which they protect against intracellular bacterial infection is not completely understood. In this report, using S. typhimurium and L. monocytogenes, we found that the levels of group 1 ILCs and NCR+ ILC3s were increased upon infection and that these increases were associated with Runt-related transcription factor 3 (Runx3) expression. Runx3 fl/fl PLZF-cre mice were much more sensitive to infection with the intracellular bacterial pathogens S. typhimurium and L. monocytogenes partially due to abnormal Group 1 ILC and NCR+ILC3 function. We also found that Runx3 directly binds to the Il12Rß2 promoter and intron 8 to accelerate the expression of Il12Rß2 and modulates IFNγ secretion triggered by the IL12/ STAT4 axis. Therefore, we demonstrate that Runx3 influences group 1 ILC- and NCR+ILC3-mediated immune protection against intracellular bacterial infections of both the gut and liver.


Asunto(s)
Subunidad alfa 3 del Factor de Unión al Sitio Principal/inmunología , Inmunidad Innata , Interleucina-12/inmunología , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Linfocitos/inmunología , Infecciones por Salmonella/inmunología , Salmonella typhimurium/inmunología , Transducción de Señal/inmunología , Animales , Subunidad alfa 3 del Factor de Unión al Sitio Principal/genética , Interleucina-12/genética , Listeriosis/genética , Listeriosis/patología , Linfocitos/patología , Ratones , Ratones Transgénicos , Infecciones por Salmonella/genética , Infecciones por Salmonella/patología , Transducción de Señal/genética
9.
Clin Chim Acta ; 487: 75-79, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30218658

RESUMEN

Runx3, a member of the Runt-related transcription factor family, has attracted extensive attention due to its important role in the development of immune systems, especially in the differentiation of T cells. Accumulated evidence indicated that altered expression of Runx3 regulates a variety of target genes in different tissues/cells. Studies in animal models suggested that Runx3 may regulate the development of T cell lineage including those of innate lymphoid cells, Treg cells and dendritic cells, which may contribute to the development of hypersensitivity and asthma. Specifically, Runx3 modulates Th1/Th2 balance and hence, the production of interleukins, which induce inflammatory responses. Understanding the roles and mechanisms of Runx3 in the regulation of immune function provides a basis for the design of novel preventive and treatment models for bronchial asthma. This article reviews published data from cell lines, animal models, and patients, concerning the relationship between Runx3 expression alteration and asthma. Epigenetic regulation of Runx3 by DNA hypermethylation and microRNA, and the implication of these pathways in asthma are also discussed.


Asunto(s)
Asma/genética , Subunidad alfa 3 del Factor de Unión al Sitio Principal/genética , Animales , Asma/inmunología , Asma/metabolismo , Subunidad alfa 3 del Factor de Unión al Sitio Principal/inmunología , Subunidad alfa 3 del Factor de Unión al Sitio Principal/metabolismo , Humanos , Inflamación/inmunología , Inflamación/metabolismo
10.
Crit Rev Immunol ; 38(1): 63-78, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29717663

RESUMEN

The transcription factor RUNX3 is a prominent regulator of multiple hematopoietic cell lineages. Gene loss of function studies demonstrated the unique and essential roles of this master regulator in differentiated lymphoid and myeloid cells. As a complementary approach, RUNX3 was upregulated in various leukocyte subsets to probe the instructive role of this 'multilineage'-specific transcription factor. In this report, we overview the immunomodulatory functions of RUNX3 within the hematopoietic compartment to gain insight into the consequences of Runx3 deletion or overexpression in committed immune cells. Genetic studies revealed the essential role of RUNX3 in Langerhans cell development. Moreover, this transcription factor is necessary for the differentiation and maintenance of the cytotoxic CD8+ T cells. In addition, T helper, natural killer, and B cells are also influenced by RUNX3. Importantly, the ectopic expression of Runx3 enhances the immunogenicity of cytotoxic T cells and pluripotent stem-cell-derived dendritic cells, suggesting that this protein can be applied in cell-based immunotherapies.


Asunto(s)
Linfocitos B/inmunología , Subunidad alfa 3 del Factor de Unión al Sitio Principal/inmunología , Linfocitos T/inmunología , Animales , Linfocitos B/citología , Células Dendríticas/citología , Células Dendríticas/inmunología , Humanos , Linfocitos T/citología
11.
J Immunol ; 200(4): 1513-1526, 2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29305435

RESUMEN

Agonists to the TNF/TNFR costimulatory receptors CD134 (OX40) and CD137 (4-1BB) elicit antitumor immunity. Dual costimulation with anti-CD134 plus anti-CD137 is particularly potent because it programs cytotoxic potential in CD8+ and CD4+ T cells. Cytotoxicity in dual-costimulated CD4 T cells depends on the T-box transcription factor eomesodermin (Eomes), which we report is induced via a mechanism that does not rely on IL-2, in contrast to CD8+ CTL, but rather depends on the CD8 T cell lineage commitment transcription factor Runx3, which supports Eomes expression in mature CD8+ CTLs. Further, Eomes and Runx3 were indispensable for dual-costimulated CD4 T cells to mediate antitumor activity in an aggressive melanoma model. Runx3 is also known to be expressed in standard CD4 Th1 cells where it fosters IFN-γ expression; however, the CD4 T cell lineage commitment factor ThPOK represses transcription of Eomes and other CD8 lineage genes, such as Cd8a Hence, CD4 T cells can differentiate into Eomes+ cytotoxic CD4+CD8+ double-positive T cells by terminating ThPOK expression. In contrast, dual-costimulated CD4 T cells express Eomes, despite the continued expression of ThPOK and the absence of CD8α, indicating that Eomes is selectively released from ThPOK repression. Finally, although Eomes was induced by CD137 agonist, but not CD134 agonist, administered individually, CD137 agonist failed to induce CD134-/- CD4 T cells to express Eomes or Runx3, indicating that both costimulatory pathways are required for cytotoxic Th1 programming, even when only CD137 is intentionally engaged with a therapeutic agonist.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Melanoma Experimental/inmunología , Proteínas de Dominio T Box/biosíntesis , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/agonistas , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología , Animales , Diferenciación Celular/inmunología , Subunidad alfa 3 del Factor de Unión al Sitio Principal/inmunología , Inmunoterapia , Activación de Linfocitos/inmunología , Melanoma Experimental/metabolismo , Ratones , Ratones Transgénicos , Receptores OX40/agonistas , Receptores OX40/inmunología , Factores de Transcripción/inmunología , Factores de Transcripción/metabolismo
12.
Nat Immunol ; 18(11): 1218-1227, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28945245

RESUMEN

T cell antigen receptor (TCR) signaling in the thymus initiates positive selection, but the CD8+-lineage fate is thought to be induced by cytokines after TCR signaling has ceased, although this remains controversial and unproven. We have identified four cytokines (IL-6, IFN-γ, TSLP and TGF-ß) that did not signal via the common γ-chain (γc) receptor but that, like IL-7 and IL-15, induced expression of the lineage-specifying transcription factor Runx3d and signaled the generation of CD8+ T cells. Elimination of in vivo signaling by all six of these 'lineage-specifying cytokines' during positive selection eliminated Runx3d expression and completely abolished the generation of CD8+ single-positive thymocytes. Thus, this study proves that signaling during positive selection by lineage-specifying cytokines is responsible for all CD8+-lineage-fate 'decisions' in the thymus.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linaje de la Célula/inmunología , Citocinas/inmunología , Timo/inmunología , Animales , Linfocitos T CD8-positivos/metabolismo , Subunidad alfa 3 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 3 del Factor de Unión al Sitio Principal/inmunología , Subunidad alfa 3 del Factor de Unión al Sitio Principal/metabolismo , Citocinas/metabolismo , Citometría de Flujo , Expresión Génica/inmunología , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/inmunología , Timocitos/inmunología , Timocitos/metabolismo , Timo/citología , Timo/metabolismo
13.
J Immunol ; 198(1): 239-248, 2017 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-27852743

RESUMEN

Application of dendritic cells (DCs) to prime responses to tumor Ags provides a promising approach to immunotherapy. However, only a limited number of DCs can be manufactured from adult precursors. In contrast, pluripotent embryonic stem (ES) cells represent an inexhaustible source for DC production, although it remains a major challenge to steer directional differentiation because ES cell-derived cells are typically immature with impaired functional capacity. Consistent with this notion, we found that mouse ES cell-derived DCs (ES-DCs) represented less mature cells compared with bone marrow-derived DCs. This finding prompted us to compare the gene expression profile of the ES cell- and adult progenitor-derived, GM-CSF-instructed, nonconventional DC subsets. We quantified the mRNA level of 17 DC-specific transcription factors and observed that 3 transcriptional regulators (Irf4, Spi-B, and Runx3) showed lower expression in ES-DCs than in bone marrow-derived DCs. In light of this altered gene expression, we probed the effects of these transcription factors in developing mouse ES-DCs with an isogenic expression screen. Our analysis revealed that forced expression of Irf4 repressed ES-DC development, whereas, in contrast, Runx3 improved the ES-DC maturation capacity. Moreover, LPS-treated and Runx3-activated ES-DCs exhibited enhanced T cell activation and migratory potential. In summary, we found that ex vivo-generated ES-DCs had a compromised maturation ability and immunogenicity. However, ectopic expression of Runx3 enhances cytokine-driven ES-DC development and acts as an instructive tool for the generation of mature DCs with enhanced immunogenicity from pluripotent stem cells.


Asunto(s)
Diferenciación Celular/fisiología , Subunidad alfa 3 del Factor de Unión al Sitio Principal/biosíntesis , Células Dendríticas/citología , Expresión Génica Ectópica/fisiología , Células Madre Embrionarias/citología , Animales , Western Blotting , Separación Celular , Células Cultivadas , Subunidad alfa 3 del Factor de Unión al Sitio Principal/inmunología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Células Madre Embrionarias/inmunología , Células Madre Embrionarias/metabolismo , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Activación de Linfocitos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/inmunología , Células Madre Pluripotentes/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Transcriptoma
14.
J Immunol ; 195(6): 2879-87, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26254341

RESUMEN

Th-inducing Pox virus and zinc finger/Krüppel-like factor (ThPOK) is a key commitment factor for CD4(+) lineage T cells and is essential for the maintenance of CD4 lineage integrity; thus, the expression of ThPOK has to be tightly controlled. In this article, we demonstrate that Myc-associated zinc finger-related factor (MAZR) and Runt-related transcription factor 1 (Runx1) together repressed ThPOK in preselection double-positive thymocytes, whereas MAZR acted in synergy with Runx3 in the repression of ThPOK in CD8(+) T cells. Furthermore, MAZR-Runx1 and MAZR-Runx3 double-mutant mice showed enhanced derepression of Cd4 in double-negative thymocytes and in CD8(+) T cells in comparison with Runx1 or Runx3 single-deficient mice, respectively, indicating that MAZR modulates Cd4 silencing. Thus, our data demonstrate developmental stage-specific synergistic activities between MAZR and Runx/core-binding factor ß (CBFß) complexes. Finally, retroviral Cre-mediated conditional deletion of MAZR in peripheral CD8(+) T cells led to the derepression of ThPOK, thus showing that MAZR is also part of the molecular machinery that maintains a repressed state of ThPOK in CD8(+) T cells.


Asunto(s)
Linfocitos T CD8-positivos/citología , Subunidad alfa 2 del Factor de Unión al Sitio Principal/inmunología , Subunidad alfa 3 del Factor de Unión al Sitio Principal/inmunología , Proteínas de Neoplasias/inmunología , Proteínas Represoras/inmunología , Factores de Transcripción/inmunología , Animales , Antígenos CD4/biosíntesis , Antígenos CD4/genética , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Diferenciación Celular/inmunología , Línea Celular , Linaje de la Célula/inmunología , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 3 del Factor de Unión al Sitio Principal/genética , Factores de Unión al Sitio Principal/inmunología , Técnicas de Inactivación de Genes , Células HEK293 , Humanos , Ratones , Ratones Transgénicos , Proteínas de Neoplasias/genética , Proteínas Represoras/genética , Timocitos/citología , Timocitos/inmunología
15.
Biochim Biophys Acta ; 1855(2): 131-43, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25641675

RESUMEN

Inactivation of tumor suppressor genes (TSG) in normal cells provides a viability/growth advantage that contributes cell-autonomously to cancer. More than a decade ago claims arose that the RUNX3 member of the RUNX transcription factor family is a major TSG inactivated in gastric cancer, a postulate extended later to other cancers. However, evidence that Runx3 is not expressed in normal gastric and other epithelia has challenged the RUNX3-TSG paradigm. Here we critically re-appraise this paradigm in light of recent high-throughput, quantitative genome-wide studies on thousands of human samples of various tumors and new investigations of the role of Runx3 in mouse cancer models. Collectively, these studies unequivocally demonstrate that RUNX3 is not a bona fide cell-autonomous TSG. Accordingly, RUNX3 is not recognized as a TSG and is not included among the 2000 cancer genes listed in the "Cancer Gene Census" or "Network for Cancer Genes" repositories. In contrast, RUNX3 does play important functions in immunity and inflammation and may thereby indirectly influence epithelial tumor development.


Asunto(s)
Subunidad alfa 3 del Factor de Unión al Sitio Principal/genética , Inmunidad Innata/genética , Inflamación/genética , Neoplasias/genética , Subunidad alfa 3 del Factor de Unión al Sitio Principal/inmunología , Genes Supresores de Tumor , Humanos , Inflamación/inmunología , Inflamación/patología , Neoplasias/inmunología , Neoplasias/patología , Neoplasias Glandulares y Epiteliales/genética , Neoplasias Glandulares y Epiteliales/inmunología , Neoplasias Glandulares y Epiteliales/patología
16.
J Immunol ; 193(8): 4032-42, 2014 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-25217159

RESUMEN

Differentiation of CD4(+) helper and CD8(+) cytotoxic αß T cells from CD4(+)CD8(+) thymocytes involves upregulation of lineage-specifying transcription factors and transcriptional silencing of CD8 or CD4 coreceptors, respectively, in MHC class II or I (MHCII or I)-restricted thymocytes. In this study, we demonstrate that inactivation of the Dicer RNA endonuclease in murine thymocytes impairs initiation of Cd4 and Cd8 silencing, leading to development of positively selected MHCI- and MHCII-restricted mature CD4(+)CD8(+) thymocytes. Expression of the antiapoptotic BCL2 protein or inactivation of the p53 proapoptotic protein rescues these thymocytes from apoptosis, increasing their frequency and permitting accumulation of CD4(+)CD8(+) αß T cells in the periphery. Dicer-deficient MHCI-restricted αß T cells fail to normally silence Cd4 and display impaired induction of the CD8 lineage-specifying transcription factor Runx3, whereas Dicer-deficient MHCII-restricted αß T cells show impaired Cd8 silencing and impaired induction of the CD4 lineage-specifying transcription factor Thpok. Finally, we show that the Drosha RNA endonuclease, which functions upstream of Dicer in microRNA biogenesis, also regulates Cd4 and Cd8 silencing. Our data demonstrate a previously dismissed function for the microRNA biogenesis machinery in regulating expression of lineage-specifying transcription factors and silencing of Cd4 and Cd8 during αß T cell differentiation.


Asunto(s)
ARN Helicasas DEAD-box/genética , MicroARNs/genética , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Ribonucleasa III/genética , Linfocitos T Citotóxicos/citología , Linfocitos T Colaboradores-Inductores/citología , Animales , Apoptosis/inmunología , Antígenos CD4/genética , Antígenos CD4/inmunología , Antígenos CD8/genética , Antígenos CD8/inmunología , Diferenciación Celular/inmunología , Linaje de la Célula/inmunología , Subunidad alfa 3 del Factor de Unión al Sitio Principal/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Antígenos de Histocompatibilidad Clase II/inmunología , Ratones , Ratones Noqueados , MicroARNs/biosíntesis , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Linfocitos T Citotóxicos/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Factores de Transcripción/inmunología , Proteína p53 Supresora de Tumor/metabolismo , Regulación hacia Arriba
17.
Nat Immunol ; 15(10): 947-56, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25129370

RESUMEN

The transcription factor ThPOK promotes CD4(+) T cell differentiation in the thymus. Here, using a mouse strain that allows post-thymic gene deletion, we show that ThPOK maintains CD4(+) T lineage integrity and couples effector differentiation to environmental cues after antigenic stimulation. ThPOK preserved the integrity and amplitude of effector responses and was required for proper differentiation of types 1 and 2 helper T cells in vivo by restraining the expression and function of Runx3, a nuclear factor crucial for cytotoxic T cell differentiation. The transcription factor LRF acts redundantly with ThPOK to prevent the transdifferentiation of mature CD4(+) T cells into CD8(+) T cells. As such, the ThPOK-LRF transcriptional module was essential for CD4(+) T cell integrity and responses.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Proteínas de Unión al ADN/inmunología , Timo/inmunología , Factores de Transcripción/inmunología , Animales , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular/inmunología , Linaje de la Célula/genética , Linaje de la Célula/inmunología , Subunidad alfa 3 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 3 del Factor de Unión al Sitio Principal/inmunología , Subunidad alfa 3 del Factor de Unión al Sitio Principal/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Citometría de Flujo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Análisis de Secuencia por Matrices de Oligonucleótidos , Células TH1/inmunología , Células TH1/metabolismo , Células Th2/inmunología , Células Th2/metabolismo , Timo/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcriptoma/inmunología
18.
Immunity ; 41(2): 169-71, 2014 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-25148017

RESUMEN

Very little is known about the transcription factor network that regulates the development of intestinal intraepithelial lymphocytes (IELs). In this issue of Immunity, Klose et al. (2014b) and Reis et al. (2014) demonstrate an essential role for T-bet in regulating the CD8αα IEL differentiation program.


Asunto(s)
Subunidad alfa 3 del Factor de Unión al Sitio Principal/inmunología , Proteínas de Dominio T Box/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Animales
19.
Immunity ; 41(2): 244-56, 2014 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-25148025

RESUMEN

The intestinal epithelium harbors large populations of activated and memory lymphocytes, yet these cells do not cause tissue damage in the steady state. We investigated how intestinal T cell effector differentiation is regulated upon migration to the intestinal epithelium. Using gene loss- and gain-of-function strategies, as well as reporter approaches, we showed that cooperation between the transcription factors T-bet and Runx3 resulted in suppression of conventional CD4(+) T helper functions and induction of an intraepithelial lymphocyte (IEL) program that included expression of IEL markers such as CD8αα homodimers. Interferon-γ sensing and T-bet expression by CD4(+) T cells were both required for this program, which was distinct from conventional T helper differentiation but shared by other IEL populations, including TCRαß(+)CD8αα(+) IELs. We conclude that the gut environment provides cues for IEL maturation through the interplay between T-bet and Runx3, allowing tissue-specific adaptation of mature T lymphocytes.


Asunto(s)
Subunidad alfa 3 del Factor de Unión al Sitio Principal/inmunología , Proteínas de Dominio T Box/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Antígenos CD8/biosíntesis , Diferenciación Celular/inmunología , Células Cultivadas , Colitis/genética , Colitis/inmunología , Proteínas de Unión al ADN/inmunología , Interferón gamma/inmunología , Interleucinas/inmunología , Mucosa Intestinal/citología , Mucosa Intestinal/inmunología , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Antígenos de Linfocitos T alfa-beta/biosíntesis , Receptores de Citocinas/genética , Receptores de Interferón/genética , Receptores de Interleucina , Transducción de Señal/inmunología , Proteínas de Dominio T Box/biosíntesis , Proteínas de Dominio T Box/genética , Factores de Transcripción/inmunología , Tretinoina , Regulación hacia Arriba , Receptor de Interferón gamma
20.
PLoS One ; 8(10): e77490, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24204843

RESUMEN

Classical dendritic cells (cDC) are specialized antigen-presenting cells mediating immunity and tolerance. cDC cell-lineage decisions are largely controlled by transcriptional factor regulatory cascades. Using an in vivo cell-specific targeting of Runx3 at various stages of DC lineage development we show that Runx3 is required for cell-identity, homeostasis and function of splenic Esam(hi) DC. Ablation of Runx3 in DC progenitors led to a substantial decrease in splenic CD4(+)/CD11b(+) DC. Combined chromatin immunoprecipitation sequencing and gene expression analysis of purified DC-subsets revealed that Runx3 is a key gene expression regulator that facilitates specification and homeostasis of CD11b(+)Esam(hi) DC. Mechanistically, loss of Runx3 alters Esam(hi) DC gene expression to a signature characteristic of WT Esam(low) DC. This transcriptional reprogramming caused a cellular change that diminished phagocytosis and hampered Runx3(-/-) Esam(hi) DC capacity to prime CD4(+) T cells, attesting to the significant role of Runx3 in specifying Esam(hi) DC identity and function.


Asunto(s)
Antígeno CD11b/genética , Moléculas de Adhesión Celular/genética , Reprogramación Celular/genética , Subunidad alfa 3 del Factor de Unión al Sitio Principal/genética , Células Dendríticas/metabolismo , Bazo/metabolismo , Transcripción Genética , Animales , Antígeno CD11b/inmunología , Antígenos CD4/genética , Antígenos CD4/inmunología , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Moléculas de Adhesión Celular/inmunología , Comunicación Celular , Diferenciación Celular , Linaje de la Célula/inmunología , Reprogramación Celular/inmunología , Subunidad alfa 3 del Factor de Unión al Sitio Principal/deficiencia , Subunidad alfa 3 del Factor de Unión al Sitio Principal/inmunología , Células Dendríticas/citología , Células Dendríticas/inmunología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Inmunoprecipitación , Ratones , Ratones Transgénicos , Fagocitosis , Análisis de Secuencia de ADN , Transducción de Señal , Bazo/citología , Bazo/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...