Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
J Immunol ; 198(10): 3919-3926, 2017 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-28404635

RESUMEN

miR-23∼27∼24 was recently implicated in restricting Th2 immunity, as well as the differentiation and function of other effector T cell lineages. Interestingly, miR-24, unlike other family members, actually promotes Th1 and Th17 responses. In this article, we show that miR-24 drives the production of IFN-γ and IL-17 in T cells at least in part through targeting TCF1, a transcription factor known for its role in limiting Th1 and Th17 immunity. Surprisingly, whereas TCF1 was previously shown to promote Th2 responses through inducing GATA3, enforced TCF1 expression in miR-24-overexpressing T cells led to further downregulation of IL-4 and GATA3 expression, suggesting miR-24-mediated inhibition of Th2 immunity cannot be attributed to TCF1 repression by miR-24. Together, our data demonstrate a novel miR-24-TCF1 pathway in controlling effector cytokine production by T cells and further suggest miR-24 could function as a key upstream molecule regulating TCF1-mediated immune responses.


Asunto(s)
Factor Nuclear 1-alfa del Hepatocito/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Subgrupos de Linfocitos T/inmunología , Animales , Diferenciación Celular , Citocinas/biosíntesis , Citocinas/inmunología , Regulación hacia Abajo , Factor de Transcripción GATA3/biosíntesis , Factor Nuclear 1-alfa del Hepatocito/genética , Interferón gamma/biosíntesis , Interferón gamma/inmunología , Interleucina-17/biosíntesis , Interleucina-17/inmunología , Interleucina-4/genética , Interleucina-4/inmunología , Activación de Linfocitos , Ratones , Transducción de Señal , Subgrupos de Linfocitos T/metabolismo , Células TH1/inmunología , Células Th17/inmunología , Células Th2/inmunología
2.
PLoS Pathog ; 11(2): e1004635, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25658840

RESUMEN

IFNγ signaling drives dendritic cells (DCs) to promote type I T cell (Th1) immunity. Here, we show that activation of DCs by IFNγ is equally crucial for the differentiation of a population of T-bet+ regulatory T (Treg) cells specialized to inhibit Th1 immune responses. Conditional deletion of IFNγ receptor in DCs but not in Treg cells resulted in a severe defect in this specific Treg cell subset, leading to exacerbated immune pathology during parasitic infections. Mechanistically, IFNγ-unresponsive DCs failed to produce sufficient amount of IL-27, a cytokine required for optimal T-bet induction in Treg cells. Thus, IFNγ signalling endows DCs with the ability to efficiently control a specific type of T cell immunity through promoting a corresponding Treg cell population.


Asunto(s)
Diferenciación Celular/inmunología , Células Dendríticas/inmunología , Interferón gamma/inmunología , Linfocitos T Reguladores/inmunología , Toxoplasmosis/inmunología , Animales , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Inflamación/inmunología , Ratones , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/inmunología , Proteínas de Dominio T Box/inmunología , Linfocitos T Reguladores/citología , Células TH1/citología , Células TH1/inmunología
3.
Proc Natl Acad Sci U S A ; 111(23): 8559-64, 2014 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-24912151

RESUMEN

Chronic viral infections incapacitate adaptive immune responses by "exhausting" virus-specific T cells, inducing their deletion and reducing productive T-cell memory. Viral infection rapidly induces death receptor CD95 (Fas) expression by dendritic cells (DCs), making them susceptible to elimination by the immune response. Lymphocytic choriomeningitis virus (LCMV) clone 13, which normally establishes a chronic infection, is rapidly cleared in C57Black6/J mice with conditional deletion of Fas in DCs. The immune response to LCMV is characterized by an extended survival of virus-specific effector T cells. Moreover, transfer of Fas-negative DCs from noninfected mice to preinfected animals results in either complete clearance of the virus or a significant reduction of viral titers. Thus, DC-specific Fas expression plays a role in regulation of antiviral responses and suggests a strategy for stimulation of T cells in chronically infected animals and humans to achieve the clearance of persistent viruses.


Asunto(s)
Células Dendríticas/inmunología , Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica/inmunología , Receptor fas/inmunología , Animales , Línea Celular , Supervivencia Celular/inmunología , Chlorocebus aethiops , Enfermedad Crónica , Células Dendríticas/metabolismo , Células Dendríticas/virología , Eliminación de Gen , Interacciones Huésped-Patógeno/inmunología , Humanos , Activación de Linfocitos/inmunología , Coriomeningitis Linfocítica/genética , Coriomeningitis Linfocítica/virología , Virus de la Coriomeningitis Linfocítica/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T/virología , Células Vero , Receptor fas/genética , Receptor fas/metabolismo
4.
J Biol Chem ; 287(35): 29324-35, 2012 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-22773832

RESUMEN

Chronic nonhealing wounds, such as venous ulcers (VUs), are a widespread and serious medical problem with high morbidity and mortality. The molecular pathology of VUs remains poorly understood, impeding the development of effective treatment strategies. Using mRNA expression profiling of VUs biopsies and computational analysis, we identified a candidate set of microRNAs with lowered target gene expression. Among these candidates, miR-16, -20a, -21, -106a -130a, and -203 were confirmed to be aberrantly overexpressed in a cohort study of 10 VU patients by quantitative PCR and in situ hybridizations. These microRNAs were predicted to target multiple genes important for wound healing, including early growth response factor 3, vinculin, and leptin receptor (LepR). Overexpression of the top up-regulated miRNAs, miR-21 and miR-130a, in primary human keratinocytes down-regulated expression of the endogenous LepR and early growth response factor 3. The luciferase reporter assay verified LepR as a direct target for miR-21 and miR-130a. Both miR-21 and miR-130a delayed epithelialization in an acute human skin wound model. Furthermore, in vivo overexpression of miR-21 inhibited epithelialization and granulation tissue formation in a rat wound model. Our results identify a novel mechanism in which overexpression of specific set of microRNAs inhibits wound healing, resulting in new potential molecular markers and targets for therapeutic intervention.


Asunto(s)
Regulación de la Expresión Génica , MicroARNs/biosíntesis , Piel/lesiones , Piel/metabolismo , Cicatrización de Heridas/fisiología , Adulto , Anciano , Anciano de 80 o más Años , Animales , Pollos , Modelos Animales de Enfermedad , Perros , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Femenino , Humanos , Masculino , Ratones , MicroARNs/genética , Persona de Mediana Edad , Ratas , Ratas Long-Evans , Receptores de Leptina/genética , Receptores de Leptina/metabolismo , Piel/patología , Transcriptoma
5.
Nat Commun ; 9(1): 2757, 2018 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-30013024

RESUMEN

Reciprocal interactions between B and follicular T helper (Tfh) cells orchestrate the germinal center (GC) reaction, a hallmark of humoral immunity. Abnormal GC responses could lead to the production of pathogenic autoantibodies and the development of autoimmunity. Here we show that miR-146a controls GC responses by targeting multiple CD40 signaling pathway components in B cells; by contrast, loss of miR-146a in T cells does not alter humoral responses. However, specific deletion of both miR-146a and its paralog, miR-146b, in T cells increases Tfh cell numbers and enhanced GC reactions. Thus, our data reveal differential cell-intrinsic regulations of GC B and Tfh cells by miR-146a and miR-146b. Together, members of the miR-146 family serve as crucial molecular brakes to coordinately control GC reactions to generate protective humoral responses without eliciting unwanted autoimmunity.


Asunto(s)
Linfocitos B/inmunología , Centro Germinal/inmunología , MicroARNs/genética , Transducción de Señal/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Autoanticuerpos/biosíntesis , Autoinmunidad/genética , Linfocitos B/citología , Linfocitos B/efectos de los fármacos , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/inmunología , Antígenos CD40/genética , Antígenos CD40/inmunología , Diferenciación Celular , Regulación de la Expresión Génica , Centro Germinal/citología , Centro Germinal/efectos de los fármacos , Inmunidad Humoral/genética , Interleucina-4/farmacología , Ratones , Ratones Transgénicos , MicroARNs/inmunología , Cultivo Primario de Células , Isoformas de Proteínas/genética , Isoformas de Proteínas/inmunología , Linfocitos T Colaboradores-Inductores/citología , Linfocitos T Colaboradores-Inductores/efectos de los fármacos
6.
J Clin Invest ; 127(2): 530-542, 2017 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-28067667

RESUMEN

MicroRNAs (miRs) are tightly regulated in the immune system, and aberrant expression of miRs often results in hematopoietic malignancies and autoimmune diseases. Previously, it was suggested that elevated levels of miR-27 in T cells isolated from patients with multiple sclerosis facilitate disease progression by inhibiting Th2 immunity and promoting pathogenic Th1 responses. Here we have demonstrated that, although mice with T cell-specific overexpression of miR-27 harbor dysregulated Th1 responses and develop autoimmune pathology, these disease phenotypes are not driven by miR-27 in effector T cells in a cell-autonomous manner. Rather, dysregulation of Th1 responses and autoimmunity resulted from a perturbed Treg compartment. Excessive miR-27 expression in murine T cells severely impaired Treg differentiation. Moreover, Tregs with exaggerated miR-27-mediated gene regulation exhibited diminished homeostasis and suppressor function in vivo. Mechanistically, we determined that miR-27 represses several known as well as previously uncharacterized targets that play critical roles in controlling multiple aspects of Treg biology. Collectively, our data show that miR-27 functions as a key regulator in Treg development and function and suggest that proper regulation of miR-27 is pivotal to safeguarding Treg-mediated immunological tolerance.


Asunto(s)
Diferenciación Celular/inmunología , Regulación de la Expresión Génica/inmunología , Tolerancia Inmunológica , MicroARNs/inmunología , Linfocitos T Reguladores/inmunología , Animales , Diferenciación Celular/genética , Ratones , Ratones Transgénicos , MicroARNs/genética , Células TH1/inmunología , Células Th2/inmunología
7.
J Exp Med ; 213(2): 235-49, 2016 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-26834155

RESUMEN

Coordinated repression of gene expression by evolutionarily conserved microRNA (miRNA) clusters and paralogs ensures that miRNAs efficiently exert their biological impact. Combining both loss- and gain-of-function genetic approaches, we show that the miR-23∼27∼24 clusters regulate multiple aspects of T cell biology, particularly helper T (Th) 2 immunity. Low expression of this miRNA family confers proper effector T cell function at both physiological and pathological settings. Further studies in T cells with exaggerated regulation by individual members of the miR-23∼27∼24 clusters revealed that miR-24 and miR-27 collaboratively limit Th2 responses through targeting IL-4 and GATA3 in both direct and indirect manners. Intriguingly, although overexpression of the entire miR-23 cluster also negatively impacts other Th lineages, enforced expression of miR-24, in contrast to miR-23 and miR-27, actually promotes the differentiation of Th1, Th17, and induced regulatory T cells, implying that under certain conditions, miRNA families can fine tune the biological effects of their regulation by having individual members antagonize rather than cooperate with each other. Together, our results identify a miRNA family with important immunological roles and suggest that tight regulation of miR-23∼27∼24 clusters in T cells is required to maintain optimal effector function and to prevent aberrant immune responses.


Asunto(s)
MicroARNs/genética , MicroARNs/inmunología , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/inmunología , Animales , Asma/genética , Asma/inmunología , Asma/patología , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Modelos Animales de Enfermedad , Factor de Transcripción GATA3/biosíntesis , Factor de Transcripción GATA3/genética , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Interleucina-4/biosíntesis , Interleucina-4/genética , Activación de Linfocitos/genética , Ratones , Ratones Transgénicos , Familia de Multigenes , Fenotipo , Linfocitos T Colaboradores-Inductores/citología , Linfocitos T Colaboradores-Inductores/inmunología , Células Th2/citología , Células Th2/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA