Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
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
2.
PLoS One ; 15(5): e0233044, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32453801

RESUMEN

Mice deficient in the transcription factor Runx3 develop a multitude of immune system defects, including early onset colitis. This paper demonstrates that Runx3 is expressed in colonic mononuclear phagocytes (MNP), including resident macrophages (RM) and dendritic cell subsets (cDC2). Runx3 deletion in MNP causes early onset colitis due to their impaired maturation. Mechanistically, the resulting MNP subset imbalance leads to up-regulation of pro-inflammatory genes as occurs in IL10R-deficient RM. In addition, RM and cDC2 display a marked decrease in expression of anti-inflammatory/TGF ß-regulated genes and ß-catenin signaling associated genes, respectively. MNP transcriptome and ChIP-seq data analysis suggest that a significant fraction of genes affected by Runx3 loss are direct Runx3 targets. Collectively, Runx3 imposes intestinal immune tolerance by regulating maturation of colonic anti-inflammatory MNP, befitting the identification of RUNX3 as a genome-wide associated risk gene for various immune-related diseases in humans, including gastrointestinal tract diseases such as Crohn's disease and celiac.


Asunto(s)
Colitis/inmunología , Colon/inmunología , Subunidad alfa 3 del Factor de Unión al Sitio Principal/genética , Sistema Mononuclear Fagocítico/inmunología , Animales , Diferenciación Celular , Colitis/genética , Modelos Animales de Enfermedad , Humanos , Ratones , Receptores de Interleucina-10/genética , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Regulación hacia Arriba , beta Catenina/metabolismo
3.
J Exp Med ; 217(1)2020 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-31653691

RESUMEN

The zinc finger transcription factor, Bcl11b, is expressed in T cells and group 2 innate lymphoid cells (ILC2s) among hematopoietic cells. In early T-lineage cells, Bcl11b directly binds and represses the gene encoding the E protein antagonist, Id2, preventing pro-T cells from adopting innate-like fates. In contrast, ILC2s co-express both Bcl11b and Id2. To address this contradiction, we have directly compared Bcl11b action mechanisms in pro-T cells and ILC2s. We found that Bcl11b binding to regions across the genome shows distinct cell type-specific motif preferences. Bcl11b occupies functionally different sites in lineage-specific patterns and controls totally different sets of target genes in these cell types. In addition, Bcl11b bears cell type-specific post-translational modifications and organizes different cell type-specific protein complexes. However, both cell types use the same distal enhancer region to control timing of Bcl11b activation. Therefore, although pro-T cells and ILC2s both need Bcl11b for optimal development and function, Bcl11b works substantially differently in these two cell types.


Asunto(s)
Linaje de la Célula/inmunología , Inmunidad Innata/inmunología , Linfocitos/inmunología , Proteínas Represoras/inmunología , Linfocitos T/inmunología , Proteínas Supresoras de Tumor/inmunología , Animales , Línea Celular , Ratones , Ratones Endogámicos C57BL , Procesamiento Proteico-Postraduccional/inmunología
4.
Sci Adv ; 5(4): eaau8389, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-31032403

RESUMEN

Patients with neurofibromatosis type 1 (NF1) are predisposed to develop neurofibromas, but the underlying molecular mechanisms of neurofibromagenesis are not fully understood. We showed dual genetic deletion of Runx1 and Runx3 in Schwann cells (SCs) and SC precursors delayed neurofibromagenesis and prolonged mouse survival. We identified peripheral myelin protein 22 (Pmp22/Gas3) related to neurofibroma initiation. Knockdown of Pmp22 with short hairpin RNAs increased Runx1fl/fl;Runx3fl/fl;Nf1fl/fl;DhhCre tumor-derived sphere numbers and enabled significantly more neurofibroma-like microlesions on transplantation. Conversely, overexpression of Pmp22 in mouse neurofibroma SCs decreased cell proliferation. Mechanistically, RUNX1/3 regulated alternative promoter usage and induced levels of protein expression of Pmp22 to control SC growth. Last, pharmacological inhibition of RUNX/core-binding factor ß (CBFB) activity significantly reduced neurofibroma volume in vivo. Thus, we identified a signaling pathway involving RUNX1/3 suppression of Pmp22 in neurofibroma initiation and/or maintenance. Targeting disruption of RUNX/CBFB interaction might provide a novel therapy for patients with neurofibroma.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Subunidad alfa 3 del Factor de Unión al Sitio Principal/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteínas de la Mielina/metabolismo , Neurofibroma/metabolismo , Alelos , Animales , Secuencia de Bases , Proliferación Celular , Supervivencia Celular , Subunidad beta del Factor de Unión al Sitio Principal/metabolismo , Femenino , Eliminación de Gen , Humanos , Masculino , Ratones , Ratones Desnudos , Regiones Promotoras Genéticas , ARN Interferente Pequeño/metabolismo , Células de Schwann/metabolismo , Transducción de Señal , Transcriptoma
5.
Dev Cell ; 42(4): 388-399.e3, 2017 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-28829946

RESUMEN

Maintaining posture requires tight regulation of the position and orientation of numerous spinal components. Yet, surprisingly little is known about this regulatory mechanism, whose failure may result in spinal deformity as in adolescent idiopathic scoliosis. Here, we use genetic mouse models to demonstrate the involvement of proprioception in regulating spine alignment. Null mutants for Runx3 transcription factor, which lack TrkC neurons connecting between proprioceptive mechanoreceptors and spinal cord, developed peripubertal scoliosis not preceded by vertebral dysplasia or muscle asymmetry. Deletion of Runx3 in the peripheral nervous system or specifically in peripheral sensory neurons, or of enhancer elements driving Runx3 expression in proprioceptive neurons, induced a similar phenotype. Egr3 knockout mice, lacking muscle spindles, but not Golgi tendon organs, displayed a less severe phenotype, suggesting that both receptor types may be required for this regulatory mechanism. These findings uncover a central role for the proprioceptive system in maintaining spinal alignment.


Asunto(s)
Subunidad alfa 3 del Factor de Unión al Sitio Principal/genética , Proteína 3 de la Respuesta de Crecimiento Precoz/genética , Mecanorreceptores/metabolismo , Propiocepción , Escoliosis/genética , Animales , Elementos de Facilitación Genéticos , Mecanorreceptores/fisiología , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/crecimiento & desarrollo , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiología , Fenotipo , Médula Espinal/crecimiento & desarrollo , Médula Espinal/metabolismo , Médula Espinal/fisiología
6.
Adv Exp Med Biol ; 962: 369-393, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28299669

RESUMEN

In this chapter we summarize the pros and cons of the notion that Runx3 is a major tumor suppressor gene (TSG). Inactivation of TSGs in normal cells provides a viability/growth advantage that contributes cell-autonomously to cancer. More than a decade ago it was suggested that RUNX3 is involved in gastric cancer development, a postulate extended later to other epithelial cancers portraying RUNX3 as a major TSG. However, evidence that Runx3 is not expressed in normal gastric and other epithelia has challenged the RUNX3-TSG paradigm. In contrast, RUNX3 is overexpressed in a significant fraction of tumor cells in various human epithelial cancers and its overexpression in pancreatic cancer cells promotes their migration, anchorage-independent growth and metastatic potential. Moreover, 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 have unequivocally demonstrated that RUNX3 is not a bona fide cell-autonomous TSG. Importantly, accumulating data demonstrated that RUNX3 functions in control of immunity and inflammation, thereby indirectly influencing epithelial tumor development.


Asunto(s)
Subunidad alfa 3 del Factor de Unión al Sitio Principal/genética , Inmunidad/genética , Inflamación/genética , Neoplasias/genética , Neoplasias/patología , Animales , Humanos , Inflamación/patología
7.
Genes Dev ; 30(23): 2607-2622, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-28007784

RESUMEN

The Runx3 transcription factor is essential for development and diversification of the dorsal root ganglia (DRGs) TrkC sensory neurons. In Runx3-deficient mice, developing TrkC neurons fail to extend central and peripheral afferents, leading to cell death and disruption of the stretch reflex circuit, resulting in severe limb ataxia. Despite its central role, the mechanisms underlying the spatiotemporal expression specificities of Runx3 in TrkC neurons were largely unknown. Here we first defined the genomic transcription unit encompassing regulatory elements (REs) that mediate the tissue-specific expression of Runx3. Using transgenic mice expressing BAC reporters spanning the Runx3 locus, we discovered three REs-dubbed R1, R2, and R3-that cross-talk with promoter-2 (P2) to drive TrkC neuron-specific Runx3 transcription. Deletion of single or multiple elements either in the BAC transgenics or by CRISPR/Cas9-mediated endogenous ablation established the REs' ability to promote and/or repress Runx3 expression in developing sensory neurons. Our analysis reveals that an intricate combinatorial interplay among the three REs governs Runx3 expression in distinct subtypes of TrkC neurons while concomitantly extinguishing its expression in non-TrkC neurons. These findings provide insights into the mechanism regulating cell type-specific expression and subtype diversification of TrkC neurons in developing DRGs.


Asunto(s)
Subunidad alfa 3 del Factor de Unión al Sitio Principal/genética , Ganglios Espinales/embriología , Regulación del Desarrollo de la Expresión Génica/genética , Neuronas/metabolismo , Elementos Reguladores de la Transcripción/genética , Animales , Ataxia/genética , Sitios de Unión , Subunidad alfa 3 del Factor de Unión al Sitio Principal/metabolismo , Embrión de Mamíferos , Ganglios Espinales/citología , Eliminación de Gen , Locomoción/genética , Masculino , Ratones , Ratones Transgénicos , Neuronas/citología , Regiones Promotoras Genéticas/genética , Unión Proteica , Factores de Transcripción/metabolismo
8.
Nat Immunol ; 16(11): 1124-33, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26414766

RESUMEN

Subsets of innate lymphoid cells (ILCs) reside in the mucosa and regulate immune responses to external pathogens. While ILCs can be phenotypically classified into ILC1, ILC2 and ILC3 subsets, the transcriptional control of commitment to each ILC lineage is incompletely understood. Here we report that the transcription factor Runx3 was essential for the normal development of ILC1 and ILC3 cells but not of ILC2 cells. Runx3 controlled the survival of ILC1 cells but not of ILC3 cells. Runx3 was required for expression of the transcription factor RORγt and its downstream target, the transcription factor AHR, in ILC3 cells. The absence of Runx3 in ILCs exacerbated infection with Citrobacter rodentium. Therefore, our data establish Runx3 as a key transcription factor in the lineage-specific differentiation of ILC1 and ILC3 cells.


Asunto(s)
Subunidad alfa 3 del Factor de Unión al Sitio Principal/metabolismo , Inmunidad Innata , Subgrupos Linfocitarios/inmunología , Subgrupos Linfocitarios/metabolismo , Animales , Antígenos Ly/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular/inmunología , Linaje de la Célula/inmunología , Citrobacter rodentium/inmunología , Citrobacter rodentium/patogenicidad , Subunidad alfa 3 del Factor de Unión al Sitio Principal/deficiencia , Subunidad alfa 3 del Factor de Unión al Sitio Principal/genética , Subunidad beta del Factor de Unión al Sitio Principal/deficiencia , Subunidad beta del Factor de Unión al Sitio Principal/genética , Subunidad beta del Factor de Unión al Sitio Principal/metabolismo , Infecciones por Enterobacteriaceae/etiología , Infecciones por Enterobacteriaceae/inmunología , Subunidad alfa del Receptor de Interleucina-7/metabolismo , Mucosa Intestinal/citología , Mucosa Intestinal/inmunología , Subgrupos Linfocitarios/citología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor 1 Gatillante de la Citotoxidad Natural/metabolismo , 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 , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Receptores de Hidrocarburo de Aril/genética , Receptores de Hidrocarburo de Aril/metabolismo
9.
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
10.
Mol Cell Biol ; 34(6): 1158-69, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24421391

RESUMEN

Natural killer cells belong to the family of innate lymphoid cells comprising the frontline defense against infected and transformed cells. Development and activation of natural killer cells is highly dependent on interleukin-15 signaling. However, very little is known about the transcription program driving this process. The transcription factor Runx3 is highly expressed in natural killer cells, but its function in these cells is largely unknown. We show that loss of Runx3 impaired interleukin-15-dependent accumulation of mature natural killer cells in vivo and under culture conditions and pregnant Runx3(-/-) mice completely lack the unique population of interleukin-15-dependent uterine natural killer cells. Combined chromatin immunoprecipitation sequencing and differential gene expression analysis of wild-type versus Runx3-deficient in vivo activated splenic natural killer cells revealed that Runx3 cooperates with ETS and T-box transcription factors to drive the interleukin-15-mediated transcription program during activation of these cells. Runx3 functions as a nuclear regulator during interleukin-15-dependent activation of natural killer cells by regulating the expression of genes involved in proliferation, maturation, and migration. Similar studies with additional transcription factors will allow the construction of a more detailed transcriptional network that controls natural killer cell development and function.


Asunto(s)
Subunidad alfa 3 del Factor de Unión al Sitio Principal/genética , Interleucina-15/genética , Células Asesinas Naturales/metabolismo , Activación de Linfocitos/genética , Factores de Transcripción/genética , Animales , Antígenos de Protozoos/genética , Antígenos de Protozoos/metabolismo , Movimiento Celular/genética , Proliferación Celular , Subunidad alfa 3 del Factor de Unión al Sitio Principal/metabolismo , Femenino , Interleucina-15/metabolismo , Interleucina-2/genética , Interleucina-2/metabolismo , Ratones , Regiones Promotoras Genéticas/genética , Factores de Transcripción/metabolismo , Transcripción Genética/genética
11.
PLoS One ; 8(11): e80467, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24236182

RESUMEN

The transcription factor Runx3 is highly expressed in CD8(+) T and NK cytotoxic lymphocytes and is required for their effective activation and proliferation but molecular insights into the transcription program regulated by Runx3 in these cells are still missing. Using Runx3-ChIP-seq and transcriptome analysis of wild type vs. Runx3(-/-) primary cells we have now identified Runx3-regulated genes in the two cell types at both resting and IL-2-activated states. Runx3-bound genomic regions in both cell types were distantly located relative to gene transcription start sites and were enriched for RUNX and ETS motifs. Bound genomic regions significantly overlapped T-bet and p300-bound enhancer regions in Runx3-expressing Th1 helper cells. Compared to resting cells, IL-2-activated CD8(+) T and NK cells contain three times more Runx3-regulated genes that are common to both cell types. Functional annotation of shared CD8(+) T and NK Runx3-regulated genes revealed enrichment for immune-associated terms including lymphocyte activation, proliferation, cytotoxicity, migration and cytokine production, highlighting the role of Runx3 in CD8(+) T and NK activated cells.


Asunto(s)
Subunidad alfa 3 del Factor de Unión al Sitio Principal/genética , Regulación de la Expresión Génica , Linfocitos T Citotóxicos/metabolismo , Transcripción Genética , Animales , Elementos de Facilitación Genéticos , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Histonas/metabolismo , Interleucina-2/metabolismo , Interleucina-2/farmacología , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Ratones , Ratones Noqueados , Motivos de Nucleótidos , Posición Específica de Matrices de Puntuación , Unión Proteica , Fase de Descanso del Ciclo Celular/genética , Linfocitos T Citotóxicos/efectos de los fármacos , Linfocitos T Citotóxicos/inmunología , Factor de Transcripción AP-1/metabolismo , Sitio de Iniciación de la Transcripción
12.
EMBO J ; 31(18): 3718-29, 2012 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-22903063

RESUMEN

The formation of functional connectivity in the nervous system is governed by axon guidance that instructs nerve growth and branching during development, implying a similarity between neuronal subtypes in terms of nerve extension. We demonstrate the molecular mechanism of another layer of complexity in vertebrates by defining a transcriptional program underlying growth differences between positionally different neurons. The rate of axon extension of the early subset of embryonic dorsal root ganglion sensory neurons is encoded in neurons at different axial levels. This code is determined by a segmental pattern of axial levels of Runx family transcription factor Runx3. Runx3 in turn determines transcription levels of genes encoding cytoskeletal proteins involved in axon extension, including Rock1 and Rock2 which have ongoing activities determining axon growth in early sensory neurons and blocking Rock activity reverses axon extension deficits of Runx3(-/-) neurons. Thus, Runx3 acts to regulate positional differences in axon extension properties apparently without affecting nerve guidance and branching, a principle that could be relevant to other parts of the nervous system.


Asunto(s)
Axones/fisiología , Subunidad alfa 3 del Factor de Unión al Sitio Principal/metabolismo , Regulación de la Expresión Génica , Células Receptoras Sensoriales/fisiología , Animales , Axones/metabolismo , Proliferación Celular , Embrión de Pollo , Ganglios Espinales/embriología , Ratones , Ratones Transgénicos , Modelos Genéticos , Sistema Nervioso/embriología , Neuronas/metabolismo , ARN/metabolismo , Factores de Tiempo
13.
Immunol Cell Biol ; 90(8): 827-30, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22370763

RESUMEN

Runx transcription factor family proteins have essential roles during T-cell development by either activating or repressing target genes. For instance, lineage- and stage-specific expression of Cd4 and ThPOK is controlled by a transcriptional silencer embedded in each locus, whose activity requires bindings of Runx complexes. The evolutionarily conserved VWRPY penta-peptide sequences in Runx proteins have been shown to be responsible for repressive function as a platform to recruit Groucho/TLE transcriptional corepressors. However, it remains elusive whether requirement for the VWRPY motif differs among Runx target genes. By examining mice lacking VWRPY motifs in both Runx1 and Runx3 proteins, here, we show a full and partial derepression of Cd4 and ThPOK in CD8-linegae T cells, respectively. Thus, whereas Cd4 silencing completely depends on the VWRPY motif, both VWRPY-dependent and -independent mechanisms operate to repress ThPOK gene. These results indicate that Runx proteins utilize different modes to repress expression of different target genes.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/química , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Subunidad alfa 3 del Factor de Unión al Sitio Principal/química , Subunidad alfa 3 del Factor de Unión al Sitio Principal/metabolismo , Regulación hacia Abajo/genética , Linfocitos T/citología , Linfocitos T/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Antígenos CD4/genética , Antígenos CD4/metabolismo , Ratones , Datos de Secuencia Molecular , Relación Estructura-Actividad , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
14.
EMBO Mol Med ; 3(10): 593-604, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21786422

RESUMEN

The Runx3 transcription factor regulates cell fate decisions during embryonic development and in adults. It was previously reported that Runx3 is strongly expressed in embryonic and adult gastrointestinal tract (GIT) epithelium (Ep) and that its loss causes gastric cancer. More than 280 publications have based their research on these findings and concluded that Runx3 is indeed a tumour suppressor (TS). In stark contrast, using various measures, we found that Runx3 expression is undetectable in GIT Ep. Employing a variety of biochemical and genetic techniques, including analysis of Runx3-GFP and R26LacZ/Runx3(Cre) or R26tdTomato/Runx3(Cre) reporter strains, we readily detected Runx3 in GIT-embedded leukocytes, dorsal root ganglia, skeletal elements and hair follicles. However, none of these approaches revealed detectable Runx3 levels in GIT Ep. Moreover, our analysis of the original Runx3(LacZ/LacZ) mice used in the previously reported study failed to reproduce the GIT expression of Runx3. The lack of evidence for Runx3 expression in normal GIT Ep creates a serious challenge to the published data and undermines the notion that Runx3 is a TS involved in cancer pathogenesis.


Asunto(s)
Subunidad alfa 3 del Factor de Unión al Sitio Principal/deficiencia , Subunidad alfa 3 del Factor de Unión al Sitio Principal/metabolismo , Mucosa Gástrica/metabolismo , Proteínas Supresoras de Tumor/deficiencia , Proteínas Supresoras de Tumor/metabolismo , Animales , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Subunidad alfa 3 del Factor de Unión al Sitio Principal/genética , Células Epiteliales/citología , Células Epiteliales/metabolismo , Mucosa Gástrica/citología , Genes Reporteros , Proteínas Fluorescentes Verdes/metabolismo , Inmunohistoquímica , Hibridación in Situ , Ratones , Ratones Noqueados , Especificidad de Órganos , Proteínas/metabolismo , ARN no Traducido , Proteínas Recombinantes de Fusión/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas Supresoras de Tumor/genética , beta-Galactosidasa/metabolismo
15.
Blood Cells Mol Dis ; 45(2): 112-6, 2010 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-20554226

RESUMEN

Runx3 protein products that are translated from the distal (P1)- and proximal (P2)-promoter transcripts appear on Western blots as a 47-46kDa doublet corresponding to full-length proteins bearing the P1- and P2-N-termini respectively. An additional 44kDa protein band, the origin and nature of which was unclear, is also detected. Transfection of full-length Runx3 cDNA bearing the P2 N-terminus (P2-cDNA) into HEK293 cells resulted in expression of both 46 and 44kDa proteins. Sequence analysis of the P2-cDNA revealed an in-frame ATG 90bp downstream (+90ATG) of the proximal +1ATG. Insertion of an N-terminal HA-tag into P2-cDNA immediately downstream of the +1ATG produced HA-tagged 46kDa and untagged 44kDa proteins, consistent with the possibility that the latter was translated through initiation at the internal +90ATG site. Deleting or blocking the activity of the +1ATG, the natural cap-dependent translation initiation site in P2-cDNA, abrogated production of the 46kDa Runx3 protein while facilitating production of the 44kDa product. These findings supported the notion that Runx3 44kDa protein resulted from internal translation initiation at the +90ATG. Northern blot and RT-PCR analyses performed on RNA from P2-cDNA transfected cells showed a single transcript and product respectively, of the expected size, ruling out the possibility that the 44kDa protein was translated from transcripts originating at a cryptic promoter or produced by alternative splicing. Taken together, the data indicate that the 44kDa protein results from translation initiation at the internal ATG and that Runx3, like its family members Runx1 and Runx2, contains a mechanism for internal mRNA translation initiation.


Asunto(s)
Subunidad alfa 3 del Factor de Unión al Sitio Principal/genética , Iniciación de la Cadena Peptídica Traduccional/genética , Biosíntesis de Proteínas/genética , Isoformas de Proteínas/biosíntesis , Animales , Western Blotting , Línea Celular , Codón Iniciador , Perros , Ratones , Análisis de Secuencia de ADN
16.
Blood Cells Mol Dis ; 43(1): 1-5, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19233693

RESUMEN

Runx3 is one of the three mammalian Runt domain transcription factors comprising the deeply conserved RUNX gene family. While the three proteins recognize the same DNA-motif, the functional overlaps are minor; each Runx has a distinct subset of biological functions. This lack of functional redundancy is the consequence of a tightly regulated spatio/temporal expression of the genes by transcriptional and post-transcriptional control mechanisms. Over the years several groups created Runx3-deficient mouse models. Analysis of these mice revealed various phenotypic features that result from loss of cell autonomous function of Runx3. Here we summarize the phenotypic similarities and dissimilarities between two of the Runx3-deficient mouse strains, discuss the basis of the discrepancies and highlight the crux of the dispute.


Asunto(s)
Subunidad alfa 3 del Factor de Unión al Sitio Principal/genética , Ratones Noqueados/fisiología , Fenotipo , Animales , Anticuerpos/inmunología , Ataxia/etiología , Ataxia/genética , Subunidad alfa 3 del Factor de Unión al Sitio Principal/inmunología , Subunidad alfa 3 del Factor de Unión al Sitio Principal/metabolismo , Epitelio/metabolismo , Epitelio/patología , Extremidades/patología , Regulación de la Expresión Génica , Hiperplasia/etiología , Hiperplasia/genética , Enfermedades Inflamatorias del Intestino/etiología , Enfermedades Inflamatorias del Intestino/genética , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Leucocitos/inmunología , Ratones
17.
J Exp Med ; 206(1): 51-9, 2009 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-19139168

RESUMEN

Activation of naive CD8(+) T cells with antigen induces their differentiation into effector cytolytic T lymphocytes (CTLs). CTLs lyse infected or aberrant target cells by exocytosis of lytic granules containing the pore-forming protein perforin and a family of proteases termed granzymes. We show that effector CTL differentiation occurs in two sequential phases in vitro, characterized by early induction of T-bet and late induction of Eomesodermin (Eomes), T-box transcription factors that regulate the early and late phases of interferon (IFN) gamma expression, respectively. In addition, we demonstrate a critical role for the transcription factor Runx3 in CTL differentiation. Runx3 regulates Eomes expression as well as expression of three cardinal markers of the effector CTL program: IFN-gamma, perforin, and granzyme B. Our data point to the existence of an elaborate transcriptional network in which Runx3 initially induces and then cooperates with T-box transcription factors to regulate gene transcription in differentiating CTLs.


Asunto(s)
Subunidad alfa 3 del Factor de Unión al Sitio Principal/fisiología , Regulación de la Expresión Génica , Proteínas de Dominio T Box/fisiología , Linfocitos T Citotóxicos/metabolismo , Animales , Northern Blotting , Western Blotting , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Células Cultivadas , Subunidad alfa 3 del Factor de Unión al Sitio Principal/genética , Citotoxicidad Inmunológica/inmunología , Granzimas/genética , Granzimas/metabolismo , Interferón gamma/metabolismo , Interleucina-2/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Modelos Biológicos , Perforina/genética , Perforina/metabolismo , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo , Linfocitos T Citotóxicos/citología , Linfocitos T Citotóxicos/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo
18.
Proc Natl Acad Sci U S A ; 106(1): 238-43, 2009 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-19114653

RESUMEN

The transcription factor Runx1 is a key regulator of definitive hematopoiesis in the embryo and the adult. Lineage-specific expression of Runx1 involves transcription and post-transcription control through usage of alternative promoters and diverse 3'UTR isoforms, respectively. We identified and mapped microRNA (miR) binding sites on Runx1 3'UTR and show that miR-27a, miR-9, miR-18a, miR-30c, and miR-199a* bind and post-transcriptionally attenuate expression of Runx1. miR-27a impacts on both the shortest (0.15 kb) and longest (3.8 kb) 3'UTRs and, along with additional miRs, might contribute to translation attenuation of Runx1 mRNA in the myeloid cell line 416B. Whereas levels of Runx1 mRNA in 416B and the B cell line 70Z were similar, the protein levels were not. Large amounts of Runx1 protein were found in 70Z cells, whereas only minute amounts of Runx1 protein were made in 416B cells and overexpression of Runx1 in 416B induced terminal differentiation associated with megakaryocytic markers. Induction of megakaryocytic differentiation in K562 cells by 12-o-tetradecanoylphorbol-13-acetate markedly increased miR-27a expression, concomitantly with binding of Runx1 to miR-27a regulatory region. The data indicate that miR-27a plays a regulatory role in megakaryocytic differentiation by attenuating Runx1 expression, and that, during megakaryopoiesis, Runx1 and miR-27a are engaged in a feedback loop involving positive regulation of miR-27a expression by Runx1.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/fisiología , Regulación de la Expresión Génica/fisiología , Megacariocitos/citología , MicroARNs/fisiología , Trombopoyesis/genética , Diferenciación Celular , Línea Celular , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Retroalimentación Fisiológica , Humanos , MicroARNs/genética , Unión Proteica , ARN Mensajero/análisis
19.
BMC Evol Biol ; 8: 228, 2008 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-18681949

RESUMEN

BACKGROUND: Members of the Runx family of transcriptional regulators, which bind DNA as heterodimers with CBFbeta, are known to play critical roles in embryonic development in many triploblastic animals such as mammals and insects. They are known to regulate basic developmental processes such as cell fate determination and cellular potency in multiple stem-cell types, including the sensory nerve cell progenitors of ganglia in mammals. RESULTS: In this study, we detect and characterize the hitherto unexplored Runx/CBFbeta genes of cnidarians and sponges, two basal animal lineages that are well known for their extensive regenerative capacity. Comparative structural modeling indicates that the Runx-CBFbeta-DNA complex from most cnidarians and sponges is highly similar to that found in humans, with changes in the residues involved in Runx-CBFbeta dimerization in either of the proteins mirrored by compensatory changes in the binding partner. In situ hybridization studies reveal that Nematostella Runx and CBFbeta are expressed predominantly in small isolated foci at the base of the ectoderm of the tentacles in adult animals, possibly representing neurons or their progenitors. CONCLUSION: These results reveal that Runx and CBFbeta likely functioned together to regulate transcription in the common ancestor of all metazoans, and the structure of the Runx-CBFbeta-DNA complex has remained extremely conserved since the human-sponge divergence. The expression data suggest a hypothesis that these genes may have played a role in nerve cell differentiation or maintenance in the common ancestor of cnidarians and bilaterians.


Asunto(s)
Cnidarios/genética , Subunidades alfa del Factor de Unión al Sitio Principal/genética , Subunidad beta del Factor de Unión al Sitio Principal/genética , Poríferos/genética , Factores de Transcripción/genética , Animales , Cnidarios/clasificación , Mapeo Contig , Subunidades alfa del Factor de Unión al Sitio Principal/química , Subunidad beta del Factor de Unión al Sitio Principal/química , Evolución Molecular , Etiquetas de Secuencia Expresada , Modelos Moleculares , Filogenia , Poríferos/clasificación , Conformación Proteica , Factores de Transcripción/química
20.
BMC Dev Biol ; 7: 84, 2007 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-17626615

RESUMEN

BACKGROUND: Alternative promoters usage is an important paradigm in transcriptional control of mammalian gene expression. However, despite the growing interest in alternative promoters and their role in genome diversification, very little is known about how and on what occasions those promoters are differentially regulated. Runx1 transcription factor is a key regulator of early hematopoiesis and a frequent target of chromosomal translocations in acute leukemias. Mice deficient in Runx1 lack definitive hematopoiesis and die in mid-gestation. Expression of Runx1 is regulated by two functionally distinct promoters designated P1 and P2. Differential usage of these two promoters creates diversity in distribution and protein-coding potential of the mRNA transcripts. While the alternative usage of P1 and P2 likely plays an important role in Runx1 biology, very little is known about the function of the P1/P2 switch in mediating tissue and stage specific expression of Runx1 during development. RESULTS: We employed mice bearing a hypomorphic Runx1 allele, with a largely diminished P2 activity, to investigate the biological role of alternative P1/P2 usage. Mice homozygous for the hypomorphic allele developed to term, but died within a few days after birth. During embryogenesis the P1/P2 activity is spatially and temporally modulated. P2 activity is required in early hematopoiesis and when attenuated, development of liver hematopoietic progenitor cells (HPC) was impaired. Early thymus development and thymopoiesis were also abrogated as reflected by thymic hypocellularity and loss of corticomedullary demarcation. Differentiation of CD4/CD8 thymocytes was impaired and their apoptosis was enhanced due to altered expression of T-cell receptors. CONCLUSION: The data delineate the activity of P1 and P2 in embryogenesis and describe previously unknown functions of Runx1. The findings show unequivocally that the role of P1/P2 during development is non redundant and underscore the significance of alternative promoter usage in Runx1 biology.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Regulación del Desarrollo de la Expresión Génica , Hematopoyesis Extramedular/genética , Regiones Promotoras Genéticas , Timo/embriología , Alelos , Animales , Apoptosis , Diferenciación Celular , Ensayo de Unidades Formadoras de Colonias , Cartilla de ADN , Embrión de Mamíferos , Células Madre Embrionarias/citología , Citometría de Flujo , Genes Letales , Células Madre Hematopoyéticas/citología , Hibridación in Situ , Ratones , Ratones Noqueados , Fenotipo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Timo/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA