Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 153
Filtrar
1.
Cell ; 183(7): 1826-1847.e31, 2020 12 23.
Artículo en Inglés | MEDLINE | ID: mdl-33296702

RESUMEN

Inborn errors of human interferon gamma (IFN-γ) immunity underlie mycobacterial disease. We report a patient with mycobacterial disease due to inherited deficiency of the transcription factor T-bet. The patient has extremely low counts of circulating Mycobacterium-reactive natural killer (NK), invariant NKT (iNKT), mucosal-associated invariant T (MAIT), and Vδ2+ γδ T lymphocytes, and of Mycobacterium-non reactive classic TH1 lymphocytes, with the residual populations of these cells also producing abnormally small amounts of IFN-γ. Other lymphocyte subsets develop normally but produce low levels of IFN-γ, with the exception of CD8+ αß T and non-classic CD4+ αß TH1∗ lymphocytes, which produce IFN-γ normally in response to mycobacterial antigens. Human T-bet deficiency thus underlies mycobacterial disease by preventing the development of innate (NK) and innate-like adaptive lymphocytes (iNKT, MAIT, and Vδ2+ γδ T cells) and IFN-γ production by them, with mycobacterium-specific, IFN-γ-producing, purely adaptive CD8+ αß T, and CD4+ αß TH1∗ cells unable to compensate for this deficit.


Asunto(s)
Inmunidad Adaptativa , Inmunidad Innata , Interferón gamma/inmunología , Mycobacterium/inmunología , Proteínas de Dominio T Box/metabolismo , Secuencia de Aminoácidos , Secuencia de Bases , Linaje de la Célula , Preescolar , Cromatina/metabolismo , Islas de CpG/genética , Metilación de ADN/genética , Células Dendríticas/metabolismo , Epigénesis Genética , Femenino , Homocigoto , Humanos , Mutación INDEL/genética , Lactante , Interferón gamma/metabolismo , Células Asesinas Naturales/citología , Células Asesinas Naturales/metabolismo , Mutación con Pérdida de Función/genética , Masculino , Infecciones por Mycobacterium/genética , Infecciones por Mycobacterium/inmunología , Infecciones por Mycobacterium/microbiología , Linaje , Proteínas de Dominio T Box/química , Proteínas de Dominio T Box/deficiencia , Proteínas de Dominio T Box/genética , Linfocitos T Colaboradores-Inductores/inmunología , Transcriptoma/genética
2.
Nature ; 626(8001): 1042-1048, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38418917

RESUMEN

The loss of the tail is among the most notable anatomical changes to have occurred along the evolutionary lineage leading to humans and to the 'anthropomorphous apes'1-3, with a proposed role in contributing to human bipedalism4-6. Yet, the genetic mechanism that facilitated tail-loss evolution in hominoids remains unknown. Here we present evidence that an individual insertion of an Alu element in the genome of the hominoid ancestor may have contributed to tail-loss evolution. We demonstrate that this Alu element-inserted into an intron of the TBXT gene7-9-pairs with a neighbouring ancestral Alu element encoded in the reverse genomic orientation and leads to a hominoid-specific alternative splicing event. To study the effect of this splicing event, we generated multiple mouse models that express both full-length and exon-skipped isoforms of Tbxt, mimicking the expression pattern of its hominoid orthologue TBXT. Mice expressing both Tbxt isoforms exhibit a complete absence of the tail or a shortened tail depending on the relative abundance of Tbxt isoforms expressed at the embryonic tail bud. These results support the notion that the exon-skipped transcript is sufficient to induce a tail-loss phenotype. Moreover, mice expressing the exon-skipped Tbxt isoform develop neural tube defects, a condition that affects approximately 1 in 1,000 neonates in humans10. Thus, tail-loss evolution may have been associated with an adaptive cost of the potential for neural tube defects, which continue to affect human health today.


Asunto(s)
Empalme Alternativo , Evolución Molecular , Hominidae , Proteínas de Dominio T Box , Cola (estructura animal) , Animales , Humanos , Ratones , Empalme Alternativo/genética , Elementos Alu/genética , Modelos Animales de Enfermedad , Genoma/genética , Hominidae/anatomía & histología , Hominidae/genética , Intrones/genética , Defectos del Tubo Neural/genética , Defectos del Tubo Neural/metabolismo , Fenotipo , Isoformas de Proteínas/deficiencia , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas de Dominio T Box/deficiencia , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo , Cola (estructura animal)/anatomía & histología , Cola (estructura animal)/embriología , Exones/genética
3.
Nat Immunol ; 17(2): 179-86, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26595889

RESUMEN

Intestinal T cells and group 3 innate lymphoid cells (ILC3 cells) control the composition of the microbiota and gut immune responses. Within the gut, ILC3 subsets coexist that either express or lack the natural cytoxicity receptor (NCR) NKp46. We identified here the transcriptional signature associated with the transcription factor T-bet-dependent differentiation of NCR(-) ILC3 cells into NCR(+) ILC3 cells. Contrary to the prevailing view, we found by conditional deletion of the key ILC3 genes Stat3, Il22, Tbx21 and Mcl1 that NCR(+) ILC3 cells were redundant for the control of mouse colonic infection with Citrobacter rodentium in the presence of T cells. However, NCR(+) ILC3 cells were essential for cecal homeostasis. Our data show that interplay between intestinal ILC3 cells and adaptive lymphocytes results in robust complementary failsafe mechanisms that ensure gut homeostasis.


Asunto(s)
Inmunidad Innata , Interleucinas/biosíntesis , Linfocitos/inmunología , Linfocitos/metabolismo , Animales , Citrobacter rodentium/inmunología , Análisis por Conglomerados , Modelos Animales de Enfermedad , Infecciones por Enterobacteriaceae/genética , Infecciones por Enterobacteriaceae/inmunología , Infecciones por Enterobacteriaceae/metabolismo , Infecciones por Enterobacteriaceae/mortalidad , Infecciones por Enterobacteriaceae/patología , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Homeostasis , Subgrupos Linfocitarios/inmunología , Subgrupos Linfocitarios/metabolismo , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/deficiencia , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/genética , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Receptor 1 Gatillante de la Citotoxidad Natural/metabolismo , Transducción de Señal , Proteínas de Dominio T Box/deficiencia , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo , Transcriptoma , Interleucina-22
4.
Proc Natl Acad Sci U S A ; 118(6)2021 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-33526653

RESUMEN

Exacerbated immune responses and loss of self-tolerance lead to the development of autoimmunity and immunopathology. Novel therapies to target autoreactive T cells are still needed. Here, we report that Th2-polarized T cells lacking the transcription factor T-bet harbor strong immunomodulatory potential and suppress antigen-specific CD8+ T cells via IL-10. Tbx21-/- Th2 cells protected mice against virus-induced type 1 diabetes development and suppressed not only naive but also memory CD8+ T cell responses. IL-10-producing, but not IL-10-deficient Tbx21-/- Th2 cells down-regulated costimulatory molecules on dendritic cells and reduced their IL-12 production after lymphocytic choriomeningitis virus infection. Impaired dendritic cell activation hindered effector and cytotoxic CD8+ T cell development after infection. These findings indicate that Tbx21-/- Th2 cells strongly suppress proinflammatory responses of naive and memory T cells via IL-10. Thus, in vivo IL-10-secreting Th2 cells could harbor a therapeutic potential for the treatment of T cell-mediated inflammatory disorders.


Asunto(s)
Memoria Inmunológica , Interleucina-10/metabolismo , Proteínas de Dominio T Box/deficiencia , Proteínas de Dominio T Box/metabolismo , Células Th2/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/prevención & control , Regulación hacia Abajo , Epítopos/inmunología , Activación de Linfocitos/inmunología , Coriomeningitis Linfocítica/inmunología , Coriomeningitis Linfocítica/virología , Virus de la Coriomeningitis Linfocítica/inmunología , Ratones Endogámicos C57BL , Ratones Noqueados
5.
Circ Res ; 127(3): e94-e106, 2020 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-32290757

RESUMEN

RATIONALE: The heartbeat is organized by the cardiac conduction system (CCS), a specialized network of cardiomyocytes. Patterning of the CCS into atrial node versus ventricular conduction system (VCS) components with distinct physiology is essential for the normal heartbeat. Distinct node versus VCS physiology has been recognized for more than a century, but the molecular basis of this regional patterning is not well understood. OBJECTIVE: To study the genetic and genomic mechanisms underlying node versus VCS distinction and investigate rhythm consequences of failed VCS patterning. METHODS AND RESULTS: Using mouse genetics, we found that the balance between T-box transcriptional activator, Tbx5, and T-box transcriptional repressor, Tbx3, determined the molecular and functional output of VCS myocytes. Adult VCS-specific removal of Tbx5 or overexpression of Tbx3 re-patterned the fast VCS into slow, nodal-like cells based on molecular and functional criteria. In these cases, gene expression profiling showed diminished expression of genes required for VCS-specific fast conduction but maintenance of expression of genes required for nodal slow conduction physiology. Action potentials of Tbx5-deficient VCS myocytes adopted nodal-specific characteristics, including increased action potential duration and cellular automaticity. Removal of Tbx5 in vivo precipitated inappropriate depolarizations in the atrioventricular (His)-bundle associated with lethal ventricular arrhythmias. TBX5 bound and directly activated cis-regulatory elements at fast conduction channel genes required for fast physiological characteristics of the VCS action potential, defining the identity of the adult VCS. CONCLUSIONS: The CCS is patterned entirely as a slow, nodal ground state, with a T-box dependent, physiologically dominant, fast conduction network driven specifically in the VCS. Disruption of the fast VCS gene regulatory network allowed nodal physiology to emerge, providing a plausible molecular mechanism for some lethal ventricular arrhythmias.


Asunto(s)
Arritmias Cardíacas/metabolismo , Nodo Atrioventricular/metabolismo , Ventrículos Cardíacos/metabolismo , Proteínas de Dominio T Box/metabolismo , Transcripción Genética , Potenciales de Acción , Animales , Arritmias Cardíacas/genética , Arritmias Cardíacas/fisiopatología , Nodo Atrioventricular/fisiopatología , Tipificación del Cuerpo , Femenino , Regulación del Desarrollo de la Expresión Génica , Células HEK293 , Frecuencia Cardíaca , Ventrículos Cardíacos/fisiopatología , Humanos , Masculino , Ratones Noqueados , Proteínas de Dominio T Box/deficiencia , Proteínas de Dominio T Box/genética , Factores de Tiempo
6.
J Immunol ; 205(3): 708-719, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32591391

RESUMEN

Clearance of intracellular infections caused by Salmonella Typhimurium (STm) requires IFN-γ and the Th1-associated transcription factor T-bet. Nevertheless, whereas IFN-γ-/- mice succumb rapidly to STm infections, T-bet-/- mice do not. In this study, we assess the anatomy of immune responses and the relationship with bacterial localization in the spleens and livers of STm-infected IFN-γ-/- and T-bet-/- mice. In IFN-γ-/- mice, there is deficient granuloma formation and inducible NO synthase (iNOS) induction, increased dissemination of bacteria throughout the organs, and rapid death. The provision of a source of IFN-γ reverses this, coincident with subsequent granuloma formation and substantially extends survival when compared with mice deficient in all sources of IFN-γ. T-bet-/- mice induce significant levels of IFN-γ- after challenge. Moreover, T-bet-/- mice have augmented IL-17 and neutrophil numbers, and neutralizing IL-17 reduces the neutrophilia but does not affect numbers of bacteria detected. Surprisingly, T-bet-/- mice exhibit surprisingly wild-type-like immune cell organization postinfection, including extensive iNOS+ granuloma formation. In wild-type mice, most bacteria are within iNOS+ granulomas, but in T-bet-/- mice, most bacteria are outside these sites. Therefore, Th1 cells act to restrict bacteria within IFN-γ-dependent iNOS+ granulomas and prevent dissemination.


Asunto(s)
Granuloma/inmunología , Óxido Nítrico Sintasa de Tipo II/inmunología , Infecciones por Salmonella/inmunología , Salmonella typhimurium/inmunología , Proteínas de Dominio T Box/deficiencia , Células TH1/inmunología , Animales , Granuloma/genética , Interferón gamma/genética , Interferón gamma/inmunología , Interleucina-17/genética , Interleucina-17/inmunología , Ratones , Ratones Noqueados , Óxido Nítrico Sintasa de Tipo II/genética , Infecciones por Salmonella/genética , Salmonella typhimurium/genética , Proteínas de Dominio T Box/inmunología
7.
Nature ; 540(7633): 395-399, 2016 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-27974754

RESUMEN

Seahorses have a specialized morphology that includes a toothless tubular mouth, a body covered with bony plates, a male brood pouch, and the absence of caudal and pelvic fins. Here we report the sequencing and de novo assembly of the genome of the tiger tail seahorse, Hippocampus comes. Comparative genomic analysis identifies higher protein and nucleotide evolutionary rates in H. comes compared with other teleost fish genomes. We identified an astacin metalloprotease gene family that has undergone expansion and is highly expressed in the male brood pouch. We also find that the H. comes genome lacks enamel matrix protein-coding proline/glutamine-rich secretory calcium-binding phosphoprotein genes, which might have led to the loss of mineralized teeth. tbx4, a regulator of hindlimb development, is also not found in H. comes genome. Knockout of tbx4 in zebrafish showed a 'pelvic fin-loss' phenotype similar to that of seahorses.


Asunto(s)
Evolución Biológica , Proteínas de Peces/genética , Genoma/genética , Smegmamorpha/anatomía & histología , Smegmamorpha/genética , Aletas de Animales/anatomía & histología , Aletas de Animales/metabolismo , Animales , Secuencia Conservada/genética , Proteínas de Peces/deficiencia , Eliminación de Gen , Genómica , Miembro Posterior/anatomía & histología , Miembro Posterior/metabolismo , Masculino , Anotación de Secuencia Molecular , Familia de Multigenes/genética , Tasa de Mutación , Filogenia , Reproducción/fisiología , Proteínas de Dominio T Box/deficiencia , Proteínas de Dominio T Box/genética , Factores de Tiempo , Proteínas de Pez Cebra/deficiencia , Proteínas de Pez Cebra/genética
8.
Am J Med Genet A ; 185(3): 923-929, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33369127

RESUMEN

Long QT syndrome (LQTS) is a genetic disease resulting in a prolonged QT interval on a resting electrocardiogram, predisposing affected individuals to polymorphic ventricular tachycardia and sudden death. Although a number of genes have been implicated in this disease, nearly one in four individuals exhibiting the LQTS phenotype are genotype-negative. Whole-exome sequencing identified a missense T223M variant in TBX5 that cosegregates with prolonged QT interval in a family with otherwise genotype-negative LQTS and sudden death. The TBX5-T223M variant was absent among large ostensibly healthy populations (gnomAD) and predicted to be pathogenic by in silico modeling based on Panther, PolyPhen-2, Provean, SIFT, SNAP2, and PredictSNP prediction tools. The variant was located in a highly conserved region of TBX5 predicted to be part of the DNA-binding interface. A luciferase assay identified a 57.5% reduction in the ability of TBX5-T223M to drive expression at the atrial natriuretic factor promotor compared to wildtype TBX5 in vitro. We conclude that the variant is pathogenic in this family, and we put TBX5 forward as a disease susceptibility allele for genotype-negative LQTS. The identification of this familial variant may serve as a basis for the identification of previously unknown mechanisms of LQTS with broader implications for cardiac electrophysiology.


Asunto(s)
Muerte Súbita Cardíaca/etiología , Síndrome de QT Prolongado/genética , Mutación Missense , Mutación Puntual , Proteínas de Dominio T Box/genética , Adulto , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Factor Natriurético Atrial/genética , Niño , Preescolar , Electrocardiografía , Femenino , Humanos , Masculino , Persona de Mediana Edad , Modelos Moleculares , Linaje , Regiones Promotoras Genéticas , Conformación Proteica , Proteínas Recombinantes/metabolismo , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Proteínas de Dominio T Box/deficiencia , Secuenciación del Exoma
9.
Immunity ; 37(4): 674-84, 2012 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-23063332

RESUMEN

Mice lacking the transcription factor T-bet in the innate immune system develop microbiota-dependent colitis. Here, we show that interleukin-17A (IL-17A)-producing IL-7Rα(+) innate lymphoid cells (ILCs) were potent promoters of disease in Tbx21(-/-)Rag2(-/-) ulcerative colitis (TRUC) mice. TNF-α produced by CD103(-)CD11b(+) dendritic cells synergized with IL-23 to drive IL-17A production by ILCs, demonstrating a previously unrecognized layer of cellular crosstalk between dendritic cells and ILCs. We have identified Helicobacter typhlonius as a key disease trigger driving excess TNF-α production and promoting colitis in TRUC mice. Crucially, T-bet also suppressed the expression of IL-7R, a key molecule involved in controlling intestinal ILC homeostasis. The importance of IL-7R signaling in TRUC disease was highlighted by the dramatic reduction in intestinal ILCs and attenuated colitis following IL-7R blockade. Taken together, these data demonstrate the mechanism by which T-bet regulates the complex interplay between mucosal dendritic cells, ILCs, and the intestinal microbiota.


Asunto(s)
Colitis Ulcerosa/inmunología , Proteínas de Unión al ADN/inmunología , Inmunidad Innata , Linfocitos/inmunología , Receptores de Interleucina-7/inmunología , Proteínas de Dominio T Box/inmunología , Animales , Células Cultivadas , Enfermedad Crónica , Colitis Ulcerosa/microbiología , Colitis Ulcerosa/patología , Proteínas de Unión al ADN/deficiencia , Helicobacter/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Transducción de Señal , Proteínas de Dominio T Box/deficiencia
10.
Immunity ; 37(4): 660-73, 2012 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-23041064

RESUMEN

T-bet is a critical transcription factor for T helper 1 (Th1) cell differentiation. To study the regulation and functions of T-bet, we developed a T-bet-ZsGreen reporter mouse strain. We determined that interleukin-12 (IL-12) and interferon-γ (IFN-γ) were redundant in inducing T-bet in mice infected with Toxoplasma gondii and that T-bet did not contribute to its own expression when induced by IL-12 and IFN-γ. By contrast, T-bet and the transcription factor Stat4 were critical for IFN-γ production whereas IFN-γ signaling was dispensable for inducing IFN-γ. Loss of T-bet resulted in activation of an endogenous program driving Th2 cell differentiation in cells expressing T-bet-ZsGreen. Genome-wide analyses indicated that T-bet directly induced many Th1 cell-related genes but indirectly suppressed Th2 cell-related genes. Our study revealed redundancy and synergy among several Th1 cell-inducing pathways in regulating the expression of T-bet and IFN-γ, and a critical role of T-bet in suppressing an endogenous Th2 cell-associated program.


Asunto(s)
Transducción de Señal , Proteínas de Dominio T Box/inmunología , Células Th2/inmunología , Animales , Diferenciación Celular , Factor de Transcripción GATA3/inmunología , Interferón gamma/biosíntesis , Interferón gamma/inmunología , Interleucina-4/inmunología , Ratones , Ratones Noqueados , Factor de Transcripción STAT4/deficiencia , Factor de Transcripción STAT4/inmunología , Proteínas de Dominio T Box/deficiencia , Células TH1/inmunología , Células Th2/citología , Toxoplasma/inmunología , Toxoplasmosis/inmunología
11.
Nature ; 524(7564): 180-5, 2015 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-26245375

RESUMEN

The source of new hepatocytes in the uninjured liver has remained an open question. By lineage tracing using the Wnt-responsive gene Axin2 in mice, we identify a population of proliferating and self-renewing cells adjacent to the central vein in the liver lobule. These pericentral cells express the early liver progenitor marker Tbx3, are diploid, and thereby differ from mature hepatocytes, which are mostly polyploid. The descendants of pericentral cells differentiate into Tbx3-negative, polyploid hepatocytes, and can replace all hepatocytes along the liver lobule during homeostatic renewal. Adjacent central vein endothelial cells provide Wnt signals that maintain the pericentral cells, thereby constituting the niche. Thus, we identify a cell population in the liver that subserves homeostatic hepatocyte renewal, characterize its anatomical niche, and identify molecular signals that regulate its activity.


Asunto(s)
Proteína Axina/metabolismo , Diploidia , Hepatocitos/citología , Hepatocitos/metabolismo , Homeostasis , Hígado/citología , Animales , Biomarcadores/metabolismo , Linaje de la Célula , Proliferación Celular , Células Clonales/citología , Células Clonales/metabolismo , Células Endoteliales/metabolismo , Femenino , Hígado/irrigación sanguínea , Masculino , Ratones , Poliploidía , Regeneración , Coloración y Etiquetado , Nicho de Células Madre/fisiología , Células Madre/citología , Células Madre/metabolismo , Proteínas de Dominio T Box/deficiencia , Proteínas de Dominio T Box/metabolismo , Factores de Tiempo , Venas/citología , Venas/metabolismo , Vía de Señalización Wnt
12.
Allergol Int ; 70(4): 415-420, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34456137

RESUMEN

Monogenic diseases of the immune system, also known as inborn errors of immunity (IEIs), are caused by single-gene mutations and result in immune deficiency and dysregulation. More than 400 monogenic diseases have been described to date, and this number is rapidly expanding. The increasing availability of next-generation sequencing is now facilitating the diagnosis of IEIs. It is known that IEIs can predispose a person to not only infectious diseases but also cancer and immune disorders, such as inflammatory, autoimmune, and atopic diseases. IEIs with eosinophilia and atopic diseases can occur in several disorders. IEIs with eosinophilia have provided insights into human immunity and the pathogenesis of allergic diseases. Eosinophilia is not a rare finding in clinical practice, and it often poses problems in terms of etiologic research and differential diagnoses. Secondary eosinophilia is the most common form. The main underlying conditions are infectious diseases such as parasitic infections, allergic disorders, drug reactions, and of course IEIs. In clinical settings, the recognition of IEIs in the context of an allergic phenotype with eosinophilia is critical for prompt diagnosis and appropriate treatment aimed at modulating pathophysiological mechanisms and improving clinical symptoms.


Asunto(s)
Eosinofilia/inmunología , Enfermedades Genéticas Congénitas/inmunología , Enfermedades del Sistema Inmune/inmunología , Actinas/inmunología , Animales , Citocinas/inmunología , Humanos , Tolerancia Inmunológica , Receptores de Antígenos de Linfocitos T/inmunología , Proteínas de Dominio T Box/deficiencia , Proteínas de Dominio T Box/inmunología , Linfocitos T/inmunología
13.
Am J Respir Cell Mol Biol ; 61(4): 525-536, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-30965014

RESUMEN

Chronic obstructive pulmonary disease (COPD) is a progressive lung disease characterized by peripheral airways inflammation and emphysema. Emerging evidence indicates a contribution of both innate and adaptive immune cells to the development of COPD. Transcription factor T-bet modulates the function of immune cells and therefore might be involved in the pathogenesis of COPD. To elucidate the role for T-bet in elastase-induced emphysema, pathological phenotypes were compared between wild-type and T-bet-/- mice. T-bet-/- mice demonstrated enhanced emphysema development on histological analyses, with higher values of mean linear intercept and dynamic compliance relative to wild-type mice. The number of neutrophils in BAL fluids, lung IL-6 and IL-17 expression, and the proportion of CD4+ T cells positive for IL-17 or retinoic acid receptor-related orphan receptor-γt were higher in T-bet-/- mice than in wild-type mice. Although T-bet downregulates cytokine expression in bone marrow-derived macrophages and MH-S cells, a murine alveolar cell line, depending on the surrounding environment, IL-6 expression in alveolar macrophages isolated from elastase-treated mice was not dependent on T-bet. Coculture of bone marrow-derived macrophages and CD4+ T cells revealed that T-bet regulation of IL-17 expression was dependent on CD4+ T cells. Neutralizing antibodies against IL-6R or IL-17 ameliorated the development of emphysema in T-bet-/- mice. In conclusion, we demonstrate that T-bet ameliorates elastase-induced emphysema formation by modulating the host immune response in the lungs.


Asunto(s)
Enfisema Pulmonar/inmunología , Proteínas de Dominio T Box/fisiología , Inmunidad Adaptativa , Animales , Líquido del Lavado Bronquioalveolar/citología , Linfocitos T CD4-Positivos/química , Linfocitos T CD4-Positivos/inmunología , Quimiotaxis de Leucocito , Citocinas/metabolismo , Femenino , Inmunidad Innata , Pulmón/inmunología , Pulmón/metabolismo , Subgrupos Linfocitarios , Macrófagos/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Neutrófilos/fisiología , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/análisis , Elastasa Pancreática/toxicidad , Fenotipo , Enfisema Pulmonar/inducido químicamente , Enfisema Pulmonar/genética , Interferencia de ARN , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología , Proteínas de Dominio T Box/deficiencia , Proteínas de Dominio T Box/genética
14.
J Immunol ; 198(11): 4513-4523, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28461570

RESUMEN

Immunotherapeutic strategies for malignant glioma have to overcome the immunomodulatory activities of M2 monocytes that appear in the circulation and as tumor-associated macrophages (TAMs). M2 cell products contribute to the growth-promoting attributes of the tumor microenvironment (TME) and bias immunity toward type 2, away from the type 1 mechanisms with antitumor properties. To drive type 1 immunity in CNS tissues, we infected GL261 tumor-bearing mice with attenuated rabies virus (RABV). These neurotropic viruses spread to CNS tissues trans-axonally, where they induce a strong type 1 immune response that involves Th1, CD8, and B cell entry across the blood-brain barrier and virus clearance in the absence of overt sequelae. Intranasal infection with attenuated RABV prolonged the survival of mice bearing established GL261 brain tumors. Despite the failure of virus spread to the tumor, infection resulted in significantly enhanced tumor necrosis, extensive CD4 T cell accumulation, and high levels of the proinflammatory factors IFN-γ, TNF-α, and inducible NO synthase in the TME merely 4 d postinfection, before significant virus spread or the appearance of RABV-specific immune mechanisms in CNS tissues. Although the majority of infiltrating CD4 cells appeared functionally inactive, the proinflammatory changes in the TME later resulted in the loss of accumulating M2 and increased M1 TAMs. Mice deficient in the Th1 transcription factor T-bet did not gain any survival advantage from RABV infection, exhibiting only limited tumor necrosis and no change in TME cytokines or TAM phenotype and highlighting the importance of type 1 mechanisms in this process.


Asunto(s)
Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/patología , Encéfalo/virología , Virus de la Rabia/inmunología , Microambiente Tumoral/inmunología , Animales , Linfocitos B/inmunología , Barrera Hematoencefálica/inmunología , Barrera Hematoencefálica/patología , Barrera Hematoencefálica/virología , Encéfalo/inmunología , Neoplasias Encefálicas/virología , Linfocitos T CD4-Positivos , Citocinas/inmunología , Modelos Animales de Enfermedad , Femenino , Interferón gamma/biosíntesis , Interferón gamma/inmunología , Ratones , Necrosis/virología , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Óxido Nítrico Sintasa de Tipo II/metabolismo , Virus de la Rabia/genética , Virus de la Rabia/fisiología , Proteínas de Dominio T Box/deficiencia , Proteínas de Dominio T Box/metabolismo , Células Th2/inmunología , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/inmunología
15.
BMC Dev Biol ; 18(1): 5, 2018 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-29506474

RESUMEN

BACKGROUND: Tbx5 deficiency in zebrafish causes several abnormal phenotypes of the heart and pectoral fins. It has been reported that exogenous human growth hormone can enhance expression of downstream mediators in the growth hormone and insulin-like growth factor I (IGF-I) pathway and partially restore dysmorphogenesis in tbx5 morphants. This study aimed to further evaluate the effects of IGF-I on cell apoptosis and dysmorphogenesis in zebrafish embryos deficient for tbx5. RESULTS: Among the five studied groups of zebrafish embryos (wild-type embryos [WT], tbx5 morphants [MO], mismatched tbx5 morpholino-treated wild-type embryos [MIS], IGF-I-treated wild-type embryos [WTIGF1], and IGF-I-treated tbx5 morphants [MOIGF1]), the expression levels of the ifg1, igf1-ra, ifg-rb, erk1, and akt2 genes as well as the ERK and AKT proteins were significantly reduced in the MO group, but were partially restored in the MOIGF1 group. These expression levels remained normal in the WT, MIS, and WTIGF1 groups. Exogenous human IGF-I also reduced the incidence of phenotypic anomalies, decreased the expression levels of apoptotic genes and proteins, suppressed cell apoptosis, and improved survival of the MOIGF1 group. CONCLUSIONS: These results suggest that IGF-I has an anti-apoptotic protective effect in zebrafish embryos with tbx5 deficiency.


Asunto(s)
Apoptosis , Embrión no Mamífero/metabolismo , Factor I del Crecimiento Similar a la Insulina/farmacología , Morfogénesis , Proteínas de Dominio T Box/deficiencia , Pez Cebra/embriología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Embrión no Mamífero/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Humanos , Morfogénesis/efectos de los fármacos , Morfogénesis/genética , Morfolinos/farmacología , Miocardio/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Fenotipo , Fosforilación/efectos de los fármacos , Análisis de Supervivencia , Proteínas de Dominio T Box/metabolismo , Pez Cebra/genética
16.
BMC Genomics ; 19(1): 429, 2018 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-29866044

RESUMEN

BACKGROUND: Microdeletion of chromosome 22q11 is associated with significant developmental anomalies, including disruption of the cardiac outflow tract, thymic/parathyroid aplasia and cleft palate. Amongst the genes within this region, TBX1 is a major candidate for many of these developmental defects. Targeted deletion of Tbx1 in the mouse has provided significant insight into the function of this transcription factor during early development of the cardiac and pharyngeal systems. However, less is known about its role during palatogenesis. To assess the influence of Tbx1 function on gene expression profile within the developing palate we performed a microarray screen using total RNA isolated from the secondary palate of E13.5 mouse embryos wild type, heterozygous and mutant for Tbx1. RESULTS: Expression-level filtering and statistical analysis revealed a total of 577 genes differentially expressed across genotypes. Data were clustered into 3 groups based on comparison between genotypes. Group A was composed of differentially expressed genes in mutant compared to wild type (n = 89); Group B included differentially expressed genes in heterozygous compared to wild type (n = 400) and Group C included differentially expressed genes in mutant compared to heterozygous (n = 88). High-throughput quantitative real-time PCR (RT-PCR) confirmed a total of 27 genes significantly changed between wild type and mutant; and 27 genes between heterozygote and mutant. Amongst these, the majority were present in both groups A and C (26 genes). Associations existed with hypertrophic cardiomyopathy, cardiac muscle contraction, dilated cardiomyopathy, focal adhesion, tight junction and calcium signalling pathways. No significant differences in gene expression were found between wild type and heterozygous palatal shelves. CONCLUSIONS: Significant differences in gene expression profile within the secondary palate of wild type and mutant embryos is consistent with a primary role for Tbx1 during palatogenesis.


Asunto(s)
Eliminación de Gen , Perfilación de la Expresión Génica , Hueso Paladar/crecimiento & desarrollo , Proteínas de Dominio T Box/deficiencia , Proteínas de Dominio T Box/genética , Animales , Femenino , Genotipo , Ratones
17.
Basic Res Cardiol ; 113(3): 19, 2018 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-29564567

RESUMEN

Previous studies have suggested the involvement of CD4 + T lymphocytes in cardiac remodelling. T-bet can direct Th1 lineage commitment. This study aimed to investigate the functional significance of T-bet in cardiac remodelling induced by pressure overload using T-bet global knockout rats. Increased T-bet levels were observed in rodent and human hypertrophied hearts. T-bet deficiency resulted in a less severe hypertrophic phenotype in rats. CD4 + T-lymphocyte reconstitution in T-bet-/- rats resulted in aggravated cardiac remodelling. T-cell homing molecule expression and cytokine secretion were altered in T-bet-deficient rat hearts. Administration of exogenous interferon-γ (IFN-γ) offset T-bet deficiency-mediated cardioprotection. Cardiomyocytes cultured in T-bet-/- CD4 + T-cell-conditioned media showed a reduced hypertrophic response after hypertrophic stimuli, which was abolished by an IFN-γ-neutralizing antibody. Taken together, our findings show that T-bet deficiency attenuates pressure overload-induced cardiac remodelling in rats. Specifically, targeting T-bet in T cells may be of great importance for the treatment of pathological cardiac remodelling and heart failure.


Asunto(s)
Cardiomegalia/metabolismo , Cardiomiopatía Dilatada/metabolismo , Miocitos Cardíacos/metabolismo , Proteínas de Dominio T Box/deficiencia , Células TH1/metabolismo , Remodelación Ventricular , Traslado Adoptivo , Animales , Cardiomegalia/inmunología , Cardiomegalia/fisiopatología , Cardiomegalia/prevención & control , Cardiomiopatía Dilatada/inmunología , Cardiomiopatía Dilatada/fisiopatología , Cardiomiopatía Dilatada/prevención & control , Células Cultivadas , Quimiotaxis de Leucocito , Citocinas/inmunología , Citocinas/metabolismo , Técnicas de Silenciamiento del Gen , Genotipo , Humanos , Interferón gamma/farmacología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/inmunología , Comunicación Paracrina , Fenotipo , Ratas Sprague-Dawley , Ratas Transgénicas , Transducción de Señal , Proteínas de Dominio T Box/genética , Células TH1/efectos de los fármacos , Células TH1/inmunología , Células TH1/trasplante , Remodelación Ventricular/efectos de los fármacos , Remodelación Ventricular/genética
18.
J Immunol ; 196(12): 4893-904, 2016 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-27183623

RESUMEN

The discovery of Th17 cell plasticity, in which CD4(+) IL-17-producing Th17 cells give rise to IL-17/IFN-γ double-producing cells and Th1-like IFNγ(+) ex-Th17 lymphocytes, has raised questions regarding which of these cell types contribute to immunopathology during inflammatory diseases. In this study, we show using Helicobacter hepaticus-induced intestinal inflammation that IL-17A(Cre)- or Rag1(Cre)-mediated deletion of Tbx21 has no effect on the generation of IL-17/IFN-γ double-producing cells, but leads to a marked absence of Th1-like IFNγ(+) ex-Th17 cells. Despite the lack of Th1-like ex-Th17 cells, the degree of H. hepaticus-triggered intestinal inflammation in mice in which Tbx21 was excised in IL-17-producing or Rag1-expressing cells is indistinguishable from that observed in control mice. In stark contrast, using experimental autoimmune encephalomyelitis, we show that IL-17A(Cre)-mediated deletion of Tbx21 prevents the conversion of Th17 cells to IL-17A/IFN-γ double-producing cells as well as Th1-like IFN-γ(+) ex-Th17 cells. However, IL-17A(Cre)-mediated deletion of Tbx21 has only limited effects on disease course in this model and is not compensated by Ag-specific Th1 cells. IL-17A(Cre)-mediated deletion of Rorc reveals that RORγt is essential for the maintenance of the Th17 cell lineage, but not immunopathology during experimental autoimmune encephalomyelitis. These results show that neither the single Th17 subset, nor its progeny, is solely responsible for immunopathology or autoimmunity.


Asunto(s)
Enteritis/inmunología , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Proteínas de Dominio T Box/metabolismo , Células Th17/inmunología , Animales , Linfocitos T CD4-Positivos/inmunología , Diferenciación Celular , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/fisiopatología , Helicobacter hepaticus/inmunología , Interferón gamma/biosíntesis , Interleucina-17/inmunología , Intestinos/inmunología , Intestinos/microbiología , Intestinos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/deficiencia , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Proteínas de Dominio T Box/deficiencia , Proteínas de Dominio T Box/genética , Células Th17/patología
19.
Nature ; 485(7396): 104-8, 2012 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-22495305

RESUMEN

Adult stem cells sustain tissue maintenance and regeneration throughout the lifetime of an animal. These cells often reside in specific signalling niches that orchestrate the stem cell's balancing act between quiescence and cell-cycle re-entry based on the demand for tissue regeneration. How stem cells maintain their capacity to replenish themselves after tissue regeneration is poorly understood. Here we use RNA-interference-based loss-of-function screening as a powerful approach to uncover transcriptional regulators that govern the self-renewal capacity and regenerative potential of stem cells. Hair follicle stem cells provide an ideal model. These cells have been purified and characterized from their native niche in vivo and, in contrast to their rapidly dividing progeny, they can be maintained and passaged long-term in vitro. Focusing on the nuclear proteins and/or transcription factors that are enriched in stem cells compared with their progeny, we screened ∼2,000 short hairpin RNAs for their effect on long-term, but not short-term, stem cell self-renewal in vitro. To address the physiological relevance of our findings, we selected one candidate that was uncovered in the screen: TBX1. This transcription factor is expressed in many tissues but has not been studied in the context of stem cell biology. By conditionally ablating Tbx1 in vivo, we showed that during homeostasis, tissue regeneration occurs normally but is markedly delayed. We then devised an in vivo assay for stem cell replenishment and found that when challenged with repetitive rounds of regeneration, the Tbx1-deficient stem cell niche becomes progressively depleted. Addressing the mechanism of TBX1 action, we discovered that TBX1 acts as an intrinsic rheostat of BMP signalling: it is a gatekeeper that governs the transition between stem cell quiescence and proliferation in hair follicles. Our results validate the RNA interference screen and underscore its power in unearthing new molecules that govern stem cell self-renewal and tissue-regenerative potential.


Asunto(s)
Interferencia de ARN , Regeneración/fisiología , Células Madre/citología , Proteínas de Dominio T Box/metabolismo , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Proliferación Celular , Células Epidérmicas , Femenino , Folículo Piloso/citología , Masculino , Ratones , Regeneración/genética , Transducción de Señal , Células Madre/metabolismo , Proteínas de Dominio T Box/deficiencia , Proteínas de Dominio T Box/genética
20.
Mol Cell ; 40(4): 594-605, 2010 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-21095589

RESUMEN

The stable and heritable H3K27-methyl mark suppresses transcription of lineage-specific genes in progenitor cells. During developmental transitions, histone demethylases are required to dramatically alter epigenetic and gene expression states to create new cell-specific profiles. It is unclear why demethylase proteins that antagonize polycomb-mediated repression continue to be expressed in terminally differentiated cells where further changes in H3K27 methylation could be deleterious. In this study, we show that the H3K27 demethylases, Jmjd3 and UTX, mediate a functional interaction between the lineage-defining T-box transcription factor family and a Brg1-containing SWI/SNF remodeling complex. Importantly, Jmjd3 is required for the coprecipitation of Brg1 with the T-box factor, T-bet, and this interaction is necessary for Ifng remodeling in differentiated Th1 cells. Thus, Jmjd3 has a required role in general chromatin remodeling that is independent from its H3K27 demethylase potential. This function for H3K27 demethylase proteins may explain their presence in differentiated cells where the epigenetic profile is already established.


Asunto(s)
Ensamble y Desensamble de Cromatina/genética , Histona Demetilasas con Dominio de Jumonji/metabolismo , Proteínas Nucleares/metabolismo , Proteínas de Dominio T Box/metabolismo , Animales , Línea Celular , Cromatina/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , ADN Helicasas/metabolismo , Histona Demetilasas , Histonas/metabolismo , Interferón gamma/genética , Lisina/metabolismo , Ratones , Regiones Promotoras Genéticas/genética , Unión Proteica , Homología de Secuencia de Aminoácido , Proteínas de Dominio T Box/deficiencia , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA