Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Nat Immunol ; 20(4): 471-481, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30778241

RESUMEN

Foxp3+ regulatory T cells (Treg cells) are crucial for the maintenance of immune homeostasis both in lymphoid tissues and in non-lymphoid tissues. Here we demonstrate that the ability of intestinal Treg cells to constrain microbiota-dependent interleukin (IL)-17-producing helper T cell (TH17 cell) and immunoglobulin A responses critically required expression of the transcription factor c-Maf. The terminal differentiation and function of several intestinal Treg cell populations, including RORγt+ Treg cells and follicular regulatory T cells, were c-Maf dependent. c-Maf controlled Treg cell-derived IL-10 production and prevented excessive signaling via the kinases PI(3)K (phosphatidylinositol-3-OH kinase) and Akt and the metabolic checkpoint kinase complex mTORC1 (mammalian target of rapamycin) and expression of inflammatory cytokines in intestinal Treg cells. c-Maf deficiency in Treg cells led to profound dysbiosis of the intestinal microbiota, which when transferred to germ-free mice was sufficient to induce exacerbated intestinal TH17 responses, even in a c-Maf-competent environment. Thus, c-Maf acts to preserve the identity and function of intestinal Treg cells, which is essential for the establishment of host-microbe symbiosis.


Asunto(s)
Inmunoglobulina A/biosíntesis , Intestinos/inmunología , Microbiota , Proteínas Proto-Oncogénicas c-maf/fisiología , Linfocitos T Reguladores/inmunología , Células Th17/inmunología , Animales , Células Cultivadas , Colitis/inmunología , Citocinas/metabolismo , Disbiosis , Regulación de la Expresión Génica , Homeostasis , Interleucina-10/biosíntesis , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas c-maf/genética , Proteínas Proto-Oncogénicas c-maf/metabolismo , Linfocitos T Reguladores/enzimología
3.
Nature ; 579(7800): 581-585, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32103173

RESUMEN

Adipose tissue is an energy store and a dynamic endocrine organ1,2. In particular, visceral adipose tissue (VAT) is critical for the regulation of systemic metabolism3,4. Impaired VAT function-for example, in obesity-is associated with insulin resistance and type 2 diabetes5,6. Regulatory T (Treg) cells that express the transcription factor FOXP3 are critical for limiting immune responses and suppressing tissue inflammation, including in the VAT7-9. Here we uncover pronounced sexual dimorphism in Treg cells in the VAT. Male VAT was enriched for Treg cells compared with female VAT, and Treg cells from male VAT were markedly different from their female counterparts in phenotype, transcriptional landscape and chromatin accessibility. Heightened inflammation in the male VAT facilitated the recruitment of Treg cells via the CCL2-CCR2 axis. Androgen regulated the differentiation of a unique IL-33-producing stromal cell population specific to the male VAT, which paralleled the local expansion of Treg cells. Sex hormones also regulated VAT inflammation, which shaped the transcriptional landscape of VAT-resident Treg cells in a BLIMP1 transcription factor-dependent manner. Overall, we find that sex-specific differences in Treg cells from VAT are determined by the tissue niche in a sex-hormone-dependent manner to limit adipose tissue inflammation.


Asunto(s)
Hormonas Esteroides Gonadales/metabolismo , Grasa Intraabdominal/inmunología , Caracteres Sexuales , Linfocitos T Reguladores/inmunología , Andrógenos/metabolismo , Animales , Quimiocina CCL2/inmunología , Cromatina/genética , Femenino , Regulación de la Expresión Génica , Inflamación/inmunología , Inflamación/metabolismo , Interleucina-33/inmunología , Grasa Intraabdominal/metabolismo , Masculino , Ratones , Factor 1 de Unión al Dominio 1 de Regulación Positiva/metabolismo , RNA-Seq , Receptores CCR2/metabolismo , Células del Estroma/citología , Células del Estroma/inmunología , Células del Estroma/metabolismo , Linfocitos T Reguladores/metabolismo , Transcripción Genética
4.
Eur Heart J ; 43(42): 4496-4511, 2022 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-35758064

RESUMEN

AIMS: Cardiotoxicity leading to heart failure (HF) is a growing problem in many cancer survivors. As specific treatment strategies are not available, RNA discovery pipelines were employed and a new and powerful circular RNA (circRNA)-based therapy was developed for the treatment of doxorubicin-induced HF. METHODS AND RESULTS: The circRNA sequencing was applied and the highly species-conserved circRNA insulin receptor (Circ-INSR) was identified, which participates in HF processes, including those provoked by cardiotoxic anti-cancer treatments. Chemotherapy-provoked cardiotoxicity leads to the down-regulation of Circ-INSR in rodents and patients, which mechanistically contributes to cardiomyocyte cell death, cardiac dysfunction, and mitochondrial damage. In contrast, Circ-INSR overexpression prevented doxorubicin-mediated cardiotoxicity in both rodent and human cardiomyocytes in vitro and in a mouse model of chronic doxorubicin cardiotoxicity. Breast cancer type 1 susceptibility protein (Brca1) was identified as a regulator of Circ-INSR expression. Detailed transcriptomic and proteomic analyses revealed that Circ-INSR regulates apoptotic and metabolic pathways in cardiomyocytes. Circ-INSR physically interacts with the single-stranded DNA-binding protein (SSBP1) mediating its cardioprotective effects under doxorubicin stress. Importantly, in vitro transcribed and circularized Circ-INSR mimics also protected against doxorubicin-induced cardiotoxicity. CONCLUSION: Circ-INSR is a highly conserved non-coding RNA which is down-regulated during cardiotoxicity and cardiac remodelling. Adeno-associated virus and circRNA mimics-based Circ-INSR overexpression prevent and reverse doxorubicin-mediated cardiomyocyte death and improve cardiac function. The results of this study highlight a novel and translationally important Circ-INSR-based therapeutic approach for doxorubicin-induced cardiac dysfunction.


Asunto(s)
Cardiotoxicidad , Cardiopatías , Ratones , Animales , Humanos , Cardiotoxicidad/etiología , Cardiotoxicidad/prevención & control , ARN Circular/genética , Receptor de Insulina/genética , Receptor de Insulina/metabolismo , Receptor de Insulina/farmacología , Proteómica , Apoptosis , Doxorrubicina/toxicidad , Miocitos Cardíacos/metabolismo , Cardiopatías/inducido químicamente , Cardiopatías/genética , Cardiopatías/prevención & control , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/farmacología , Proteínas Mitocondriales
5.
PLoS Biol ; 17(3): e2006716, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30856173

RESUMEN

The interdependence of selective cues during development of regulatory T cells (Treg cells) in the thymus and their suppressive function remains incompletely understood. Here, we analyzed this interdependence by taking advantage of highly dynamic changes in expression of microRNA 181 family members miR-181a-1 and miR-181b-1 (miR-181a/b-1) during late T-cell development with very high levels of expression during thymocyte selection, followed by massive down-regulation in the periphery. Loss of miR-181a/b-1 resulted in inefficient de novo generation of Treg cells in the thymus but simultaneously permitted homeostatic expansion in the periphery in the absence of competition. Modulation of T-cell receptor (TCR) signal strength in vivo indicated that miR-181a/b-1 controlled Treg-cell formation via establishing adequate signaling thresholds. Unexpectedly, miR-181a/b-1-deficient Treg cells displayed elevated suppressive capacity in vivo, in line with elevated levels of cytotoxic T-lymphocyte-associated 4 (CTLA-4) protein, but not mRNA, in thymic and peripheral Treg cells. Therefore, we propose that intrathymic miR-181a/b-1 controls development of Treg cells and imposes a developmental legacy on their peripheral function.


Asunto(s)
MicroARNs/metabolismo , Linfocitos T Reguladores/metabolismo , Animales , Citometría de Flujo , Ratones , Ratones Noqueados , MicroARNs/genética , Microscopía Confocal , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Timocitos/metabolismo
7.
Eur J Immunol ; 49(1): 121-132, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30281154

RESUMEN

The interdependence of posttranscriptional gene regulation via miRNA and transcriptional regulatory networks in lymphocyte development is poorly understood. Here, we identified miR-191 as direct upstream modulator of a transcriptional module comprising the transcription factors Foxp1, E2A, and Egr1. Deletion as well as ectopic expression of miR-191 resulted in developmental arrest in B lineage cells, indicating that fine tuning of the combined expression levels of Foxp1, E2A, and Egr1, which in turn control somatic recombination and cytokine-driven expansion, constitutes a prerequisite for efficient B-cell development. In conclusion, we propose that miR-191 acts as a rheostat in B-cell development by fine tuning a key transcriptional program.


Asunto(s)
Linfocitos B/fisiología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Factores de Transcripción Forkhead/metabolismo , MicroARNs/genética , Proteínas Represoras/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Diferenciación Celular , Células Cultivadas , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Factores de Transcripción Forkhead/genética , Redes Reguladoras de Genes , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Interferente Pequeño/genética , Recombinación Genética , Proteínas Represoras/genética , Transcripción Genética , Transgenes/genética
8.
J Immunol ; 195(10): 4832-40, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-26475928

RESUMEN

miRNAs regulate a large variety of developmental processes including development of the immune system. T cell development is tightly controlled through the interplay of transcriptional programs and cytokine-mediated signals. However, the role of individual miRNAs in this process remains largely elusive. In this study, we demonstrated that hematopoietic cell-specific loss of miR-17∼92, a cluster of six miRNAs implicated in B and T lineage leukemogenesis, resulted in profound defects in T cell development both at the level of prethymic T cell progenitors as well as intrathymically. We identified reduced surface expression of IL-7R and concomitant limited responsiveness to IL-7 signals as a common mechanism resulting in reduced cell survival of common lymphoid progenitors and thymocytes at the double-negative to double-positive transition. In conclusion, we identified miR-17∼92 as a critical modulator of multiple stages of T cell development.


Asunto(s)
Interleucina-7/inmunología , MicroARNs/inmunología , Transducción de Señal/fisiología , Linfocitos T/inmunología , Animales , Animales Modificados Genéticamente , Linfocitos B/citología , Linfocitos B/inmunología , Supervivencia Celular/genética , Supervivencia Celular/inmunología , Interleucina-7/genética , Ratones , MicroARNs/genética , Receptores de Interleucina-17/genética , Receptores de Interleucina-17/inmunología , Linfocitos T/citología
9.
Immunol Cell Biol ; 94(8): 741-6, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27089939

RESUMEN

Expression of microRNA miR-181a/b-1 is critical for intrathymic development of invariant natural killer T (iNKT) cells. However, the underlying mechanism has remained a matter of debate. On the one hand, growing evidence suggested that miR-181a/b-1 is instrumental in setting T-cell receptor (TCR) signaling threshold and thus permits agonist selection of iNKT cells through high-affinity TCR ligands. On the other hand, alterations in metabolic fitness mediated by miR-181a/b-1-dependent dysregulation of phosphatase and tensin homolog (Pten) have been proposed to cause the iNKT-cell defect in miR-181-a/b-1-deficient mice. To re-assess the hypothesis that modulation of TCR signal strength is the key mechanism by which miR-181a/b-1 controls the development of iNKT cells, we generated miR-181a/b-1-deficient mice expressing elevated levels of a Vα14Jα18 TCRα chain. In these mice, development of iNKT cells was fully restored. Furthermore, both subset distribution of iNKT cells as well as TCR Vß repertoire were independent of the presence of miR-181a/b-1 once a Vα14Jα18 TCRα chain was overexpressed. Finally, levels of Pten protein were similar in Vα14Jα18 transgenic mice irrespective of their miR-181a/b-1 status. Collectively, our data support a model in which miR-181 promotes development of iNKT cells primarily by generating a permissive state for agonist selection with alterations in metabolic fitness possibly constituting a secondary effect.


Asunto(s)
MicroARNs/metabolismo , Células T Asesinas Naturales/citología , Células T Asesinas Naturales/metabolismo , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Animales , Polaridad Celular , Subgrupos Linfocitarios/inmunología , Ratones Transgénicos , MicroARNs/genética , Fosfohidrolasa PTEN/metabolismo , Transgenes
10.
Leukemia ; 35(12): 3561-3567, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-33976371

RESUMEN

Humanized mouse models have become increasingly valuable tools to study human hematopoiesis and infectious diseases. However, human T-cell differentiation remains inefficient. We generated mice expressing human interleukin-7 (IL-7), a critical growth and survival factor for T cells, under the control of murine IL-7 regulatory elements. After transfer of human cord blood-derived hematopoietic stem and progenitor cells, transgenic mice on the NSGW41 background, termed NSGW41hIL7, showed elevated and prolonged human cellularity in the thymus while maintaining physiological ratios of thymocyte subsets. As a consequence, numbers of functional human T cells in the periphery were increased without evidence for pathological lymphoproliferation or aberrant expansion of effector or memory-like T cells. We conclude that the novel NSGW41hIL7 strain represents an optimized mouse model for humanization to better understand human T-cell differentiation in vivo and to generate a human immune system with a better approximation of human lymphocyte ratios.


Asunto(s)
Sangre Fetal/metabolismo , Células Madre Hematopoyéticas/metabolismo , Interleucina-7/metabolismo , Subgrupos de Linfocitos T/inmunología , Animales , Animales Modificados Genéticamente , Diferenciación Celular/fisiología , Sangre Fetal/citología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/inmunología , Humanos , Interleucina-7/genética , Ratones , Ratones Transgénicos , Especificidad de Órganos , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/metabolismo
11.
Nat Commun ; 11(1): 252, 2020 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-31937752

RESUMEN

Differentiation and homeostasis of Foxp3+ regulatory T (Treg) cells are strictly controlled by T-cell receptor (TCR) signals; however, molecular mechanisms that govern these processes are incompletely understood. Here we show that Bach2 is an important regulator of Treg cell differentiation and homeostasis downstream of TCR signaling. Bach2 prevents premature differentiation of fully suppressive effector Treg (eTreg) cells, limits IL-10 production and is required for the development of peripherally induced Treg (pTreg) cells in the gastrointestinal tract. Bach2 attenuates TCR signaling-induced IRF4-dependent Treg cell differentiation. Deletion of IRF4 promotes inducible Treg cell differentiation and rescues pTreg cell differentiation in the absence of Bach2. In turn, loss of Bach2 normalizes eTreg cell differentiation of IRF4-deficient Treg cells. Mechanistically, Bach2 counteracts the DNA-binding activity of IRF4 and limits chromatin accessibility, thereby attenuating IRF4-dependent transcription. Thus, Bach2 balances TCR signaling induced transcriptional activity of IRF4 to maintain homeostasis of thymically-derived and peripherally-derived Treg cells.


Asunto(s)
Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Animales , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/deficiencia , Diferenciación Celular/inmunología , Cromatina/metabolismo , Colitis/inmunología , Modelos Animales de Enfermedad , Epigénesis Genética/inmunología , Factores de Transcripción Forkhead/metabolismo , Tracto Gastrointestinal/inmunología , Regulación de la Expresión Génica/inmunología , Homeostasis/inmunología , Factores Reguladores del Interferón/deficiencia , Factores Reguladores del Interferón/metabolismo , Interleucina-10/biosíntesis , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Transducción de Señal/inmunología , Subgrupos de Linfocitos T/metabolismo , Linfocitos T Reguladores/metabolismo
12.
J Clin Invest ; 130(6): 3137-3150, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32125291

RESUMEN

The molecular mechanisms responsible for the high immunosuppressive capacity of CD4+ Tregs in tumors are not well known. High-dimensional single-cell profiling of T cells from chemotherapy-naive individuals with non-small-cell lung cancer identified the transcription factor IRF4 as specifically expressed by a subset of intratumoral CD4+ effector Tregs with superior suppressive activity. In contrast to the IRF4- counterparts, IRF4+ Tregs expressed a vast array of suppressive molecules, and their presence correlated with multiple exhausted subpopulations of T cells. Integration of transcriptomic and epigenomic data revealed that IRF4, either alone or in combination with its partner BATF, directly controlled a molecular program responsible for immunosuppression in tumors. Accordingly, deletion of Irf4 exclusively in Tregs resulted in delayed tumor growth in mice while the abundance of IRF4+ Tregs correlated with poor prognosis in patients with multiple human cancers. Thus, a common mechanism underlies immunosuppression in the tumor microenvironment irrespective of the tumor type.


Asunto(s)
Diferenciación Celular/inmunología , Factores Reguladores del Interferón/inmunología , Proteínas de Neoplasias/inmunología , Neoplasias/inmunología , Linfocitos T Reguladores/inmunología , Microambiente Tumoral/inmunología , Anciano , Anciano de 80 o más Años , Animales , Humanos , Masculino , Ratones , Persona de Mediana Edad , Neoplasias/patología , Linfocitos T Reguladores/patología
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda