Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Pathol ; 257(3): 352-366, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35297529

RESUMEN

Muscular dystrophies are genetic diseases characterized by chronic inflammation and fibrosis. Macrophages are immune cells that sustain muscle regeneration upon acute injury but seem deleterious in the context of chronic muscle injury such as in muscular dystrophies. Here, we observed that the number of macrophages expressing the transcription factor Nfix increases in two distinct mouse models of muscular dystrophies. We showed that the deletion of Nfix in macrophages in dystrophic mice delays the establishment of fibrosis and muscle wasting, and increases grasp force. Macrophages lacking Nfix expressed more TNFα and less TGFß1, thus promoting apoptosis of fibro-adipogenic progenitors. Moreover, pharmacological treatment of dystrophic mice with a ROCK inhibitor accelerated fibrosis through the increase of Nfix expression by macrophages. Thus, we have identified Nfix as a macrophage profibrotic factor in muscular dystrophies, whose inhibition could be a therapeutic route to reduce severity of the dystrophic disease. © 2022 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Asunto(s)
Macrófagos , Distrofias Musculares , Factores de Transcripción NFI , Animales , Fibrosis , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Ratones Endogámicos mdx , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Distrofias Musculares/metabolismo , Distrofias Musculares/patología , Factores de Transcripción NFI/deficiencia , Factores de Transcripción NFI/genética , Factores de Transcripción NFI/metabolismo
2.
J Clin Invest ; 129(10): 4408-4418, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31498149

RESUMEN

Reactive astrocytes are associated with every form of neurological injury. Despite their ubiquity, the molecular mechanisms controlling their production and diverse functions remain poorly defined. Because many features of astrocyte development are recapitulated in reactive astrocytes, we investigated the role of nuclear factor I-A (NFIA), a key transcriptional regulator of astrocyte development whose contributions to reactive astrocytes remain undefined. Here, we show that NFIA is highly expressed in reactive astrocytes in human neurological injury and identify unique roles across distinct injury states and regions of the CNS. In the spinal cord, after white matter injury (WMI), NFIA-deficient astrocytes exhibit defects in blood-brain barrier remodeling, which are correlated with the suppression of timely remyelination. In the cortex, after ischemic stroke, NFIA is required for the production of reactive astrocytes from the subventricular zone (SVZ). Mechanistically, NFIA directly regulates the expression of thrombospondin 4 (Thbs4) in the SVZ, revealing a key transcriptional node regulating reactive astrogenesis. Together, these studies uncover critical roles for NFIA in reactive astrocytes and illustrate how region- and injury-specific factors dictate the spectrum of reactive astrocyte responses.


Asunto(s)
Astrocitos/metabolismo , Astrocitos/patología , Sistema Nervioso Central/lesiones , Sistema Nervioso Central/metabolismo , Factores de Transcripción NFI/metabolismo , Adulto , Animales , Barrera Hematoencefálica , Diferenciación Celular , Sistema Nervioso Central/patología , Humanos , Ratones , Ratones Noqueados , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/patología , Factores de Transcripción NFI/deficiencia , Factores de Transcripción NFI/genética , Oligodendroglía/metabolismo , Oligodendroglía/patología , Remielinización , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/patología , Trombospondinas/genética , Trombospondinas/metabolismo
3.
PLoS One ; 13(10): e0205298, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30289920

RESUMEN

The majority of apparently balanced translocation (ABT) carriers are phenotypically normal. However, several mechanisms were proposed to underlie phenotypes in affected ABT cases. In the current study, whole-genome mate-pair sequencing (WG-MPS) followed by Sanger sequencing was applied to further characterize de novo ABTs in three affected individuals. WG-MPS precisely mapped all ABT breakpoints and revealed three possible underlying molecular mechanisms. Firstly, in a t(X;1) carrier with hearing loss, a highly skewed X-inactivation pattern was observed and the der(X) breakpoint mapped ~87kb upstream an X-linked deafness gene namely POU3F4, thus suggesting an underlying long-range position effect mechanism. Secondly, cryptic complexity and a chromothripsis rearrangement was identified in a t(6;7;8;12) carrier with intellectual disability. Two translocations and a heterozygous deletion disrupted SOX5; a dominant nervous system development gene previously reported in similar patients. Finally, a direct gene disruption mechanism was proposed in a t(4;9) carrier with dysmorphic facial features and speech delay. In this case, the der(9) breakpoint directly disrupted NFIB, a gene involved in lung maturation and development of the pons with important functions in main speech processes. To conclude, in contrast to familial ABT cases with identical rearrangements and discordant phenotypes, where translocations are considered coincidental, translocations seem to be associated with phenotype presentation in affected de novo ABT cases. In addition, this study highlights the importance of investigating both coding and non-coding regions to decipher the underlying pathogenic mechanisms in these patients, and supports the potential introduction of low coverage WG-MPS in the clinical investigation of de novo ABTs.


Asunto(s)
Facies , Pérdida Auditiva/genética , Discapacidad Intelectual/genética , Trastornos del Desarrollo del Lenguaje/genética , Translocación Genética , Secuencia de Bases , Puntos de Rotura del Cromosoma , Femenino , Expresión Génica , Pérdida Auditiva/diagnóstico , Pérdida Auditiva/fisiopatología , Humanos , Discapacidad Intelectual/diagnóstico , Discapacidad Intelectual/fisiopatología , Cariotipo , Trastornos del Desarrollo del Lenguaje/diagnóstico , Trastornos del Desarrollo del Lenguaje/fisiopatología , Factores de Transcripción NFI/deficiencia , Factores de Transcripción NFI/genética , Factores del Dominio POU/deficiencia , Factores del Dominio POU/genética , Fenotipo , Factores de Transcripción SOXD/deficiencia , Factores de Transcripción SOXD/genética , Secuenciación Completa del Genoma
4.
Cell Tissue Res ; 374(3): 531-540, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30091046

RESUMEN

Enamel makes up the outermost layer of the crown and its hardness protects other dental tissues from various stimuli. Enamel cannot be regenerated once damaged because ameloblasts are lost during the tooth eruption. Since the ameloblast differentiation mechanism is still unknown, further research is essential for developing treatments for defective or damaged enamel. Previously, we have reported that osteoblast differentiation and bone formation were regulated through the runt-related transcription factor 2 (Runx2)-nuclear factor 1-C (Nfic)-osterix (Osx) pathway where Nfic directly controls Osx expression. This pathway regulates odontoblast differentiation and dentin formation as well. The aim of this study was to investigate if the same pathway is applicable for ameloblast differentiation. Structural enamel defects with disorganized ameloblasts and decreased proliferation activity of the cervical loop were observed in Nfic-/- mice incisors. Expression of the ameloblast differentiation markers was also downregulated significantly in Nfic-/- mice. Real-time PCR analyses suggested that Runx2, Nfic, and Osx regulate the expression of ameloblast differentiation markers, where Runx2 is upstream of Nfic, and Nfic controls Osx expression. Therefore, we suggest the Runx2-Nfic-Osx pathway as one of the key factors that regulate ameloblast differentiation.


Asunto(s)
Ameloblastos/citología , Ameloblastos/metabolismo , Diferenciación Celular , Esmalte Dental/metabolismo , Factores de Transcripción NFI/metabolismo , Transducción de Señal , Factor de Transcripción Sp7/metabolismo , Animales , Biomarcadores/metabolismo , Línea Celular , Linaje de la Célula , Proliferación Celular , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Esmalte Dental/ultraestructura , Ratones , Factores de Transcripción NFI/deficiencia , Diente/metabolismo , Diente/ultraestructura , Microtomografía por Rayos X
5.
Development ; 145(3)2018 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-29437824

RESUMEN

Our understanding of the transcriptional programme underpinning adult hippocampal neurogenesis is incomplete. In mice, under basal conditions, adult hippocampal neural stem cells (AH-NSCs) generate neurons and astrocytes, but not oligodendrocytes. The factors limiting oligodendrocyte production, however, remain unclear. Here, we reveal that the transcription factor NFIX plays a key role in this process. NFIX is expressed by AH-NSCs, and its expression is sharply upregulated in adult hippocampal neuroblasts. Conditional ablation of Nfix from AH-NSCs, coupled with lineage tracing, transcriptomic sequencing and behavioural studies collectively reveal that NFIX is cell-autonomously required for neuroblast maturation and survival. Moreover, a small number of AH-NSCs also develop into oligodendrocytes following Nfix deletion. Remarkably, when Nfix is deleted specifically from intermediate progenitor cells and neuroblasts using a Dcx-creERT2 driver, these cells also display elevated signatures of oligodendrocyte gene expression. Together, these results demonstrate the central role played by NFIX in neuroblasts within the adult hippocampal stem cell neurogenic niche in promoting the maturation and survival of these cells, while concomitantly repressing oligodendrocyte gene expression signatures.


Asunto(s)
Hipocampo/citología , Hipocampo/metabolismo , Factores de Transcripción NFI/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neurogénesis/fisiología , Animales , Astrocitos/citología , Astrocitos/metabolismo , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Supervivencia Celular , Proteína Doblecortina , Femenino , Regulación del Desarrollo de la Expresión Génica , Hipocampo/crecimiento & desarrollo , Masculino , Trastornos de la Memoria/genética , Trastornos de la Memoria/patología , Trastornos de la Memoria/fisiopatología , Ratones , Ratones Noqueados , Factores de Transcripción NFI/deficiencia , Factores de Transcripción NFI/genética , Neurogénesis/genética , Neuronas/citología , Neuronas/metabolismo , Oligodendroglía/citología , Oligodendroglía/metabolismo , Nicho de Células Madre/genética , Nicho de Células Madre/fisiología , Regulación hacia Arriba
6.
Development ; 145(2)2018 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-29247144

RESUMEN

During appendicular skeletal development, the bi-potential cartilage anlagen gives rise to transient cartilage, which is eventually replaced by bone, and to articular cartilage that caps the ends of individual skeletal elements. While the molecular mechanism that regulates transient cartilage differentiation is relatively well understood, the mechanism of articular cartilage differentiation has only begun to be unraveled. Furthermore, the molecules that coordinate the articular and transient cartilage differentiation processes are poorly understood. Here, we have characterized in chick the regulatory roles of two transcription factors, NFIA and GATA3, in articular cartilage differentiation, maintenance and the coordinated differentiation of articular and transient cartilage. Both NFIA and GATA3 block hypertrophic differentiation. Our results suggest that NFIA is not sufficient but necessary for articular cartilage differentiation. Ectopic activation of GATA3 promotes articular cartilage differentiation, whereas inhibition of GATA3 activity promotes transient cartilage differentiation at the expense of articular cartilage. We propose a novel transcriptional circuitry involved in embryonic articular cartilage differentiation, maintenance and its crosstalk with the transient cartilage differentiation program.


Asunto(s)
Proteínas Aviares/metabolismo , Cartílago Articular/embriología , Cartílago Articular/metabolismo , Factor de Transcripción GATA3/metabolismo , Factores de Transcripción NFI/metabolismo , Animales , Animales Modificados Genéticamente , Proteínas Aviares/deficiencia , Proteínas Aviares/genética , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Embrión de Pollo , Condrocitos/citología , Condrocitos/metabolismo , Femenino , Factor de Transcripción GATA3/genética , Técnicas de Silenciamiento del Gen , Masculino , Ratones , Ratones Noqueados , Modelos Biológicos , Factores de Transcripción NFI/deficiencia , Factores de Transcripción NFI/genética , Embarazo , ARN Interferente Pequeño/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
7.
Nat Cell Biol ; 19(9): 1081-1092, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28812581

RESUMEN

Brown fat dissipates energy as heat and protects against obesity. Here, we identified nuclear factor I-A (NFIA) as a transcriptional regulator of brown fat by a genome-wide open chromatin analysis of murine brown and white fat followed by motif analysis of brown-fat-specific open chromatin regions. NFIA and the master transcriptional regulator of adipogenesis, PPARγ, co-localize at the brown-fat-specific enhancers. Moreover, the binding of NFIA precedes and facilitates the binding of PPARγ, leading to increased chromatin accessibility and active transcription. Introduction of NFIA into myoblasts results in brown adipocyte differentiation. Conversely, the brown fat of NFIA-knockout mice displays impaired expression of the brown-fat-specific genes and reciprocal elevation of muscle genes. Finally, expression of NFIA and the brown-fat-specific genes is positively correlated in human brown fat. These results indicate that NFIA activates the cell-type-specific enhancers and facilitates the binding of PPARγ to control the brown fat gene program.


Asunto(s)
Adipocitos Marrones/metabolismo , Adipogénesis/genética , Tejido Adiposo Pardo/metabolismo , Factores de Transcripción NFI/metabolismo , PPAR gamma/metabolismo , Transcripción Genética , Células 3T3-L1 , Tejido Adiposo Pardo/citología , Animales , Sitios de Unión , Cromatina/genética , Cromatina/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Elementos de Facilitación Genéticos , Regulación de la Expresión Génica , Genotipo , Secuenciación de Nucleótidos de Alto Rendimiento , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Desarrollo de Músculos , Mioblastos/metabolismo , Factores de Transcripción NFI/deficiencia , Factores de Transcripción NFI/genética , PPAR gamma/genética , Fenotipo , Unión Proteica , Interferencia de ARN , Transducción de Señal , Factores de Tiempo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transfección
8.
Infect Immun ; 85(4)2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28167668

RESUMEN

Myeloid progenitor-derived suppressor cells (MDSCs) arise from myeloid progenitors and suppress both innate and adaptive immunity. MDSCs expand during the later phases of sepsis in mice, promote immunosuppression, and reduce survival. Here, we report that the myeloid differentiation-related transcription factor nuclear factor I-A (NFI-A) controls MDSC expansion during sepsis and impacts survival. Unlike MDSCs, myeloid cells with conditional deletion of the Nfia gene normally differentiated into effector cells during sepsis, cleared infecting bacteria, and did not express immunosuppressive mediators. In contrast, ectopic expression of NFI-A in myeloid progenitors from NFI-A myeloid cell-deficient mice impeded myeloid cell maturation and promoted immune repressor function. Importantly, surviving septic mice with conditionally deficient NFI-A myeloid cells were able to respond to challenge with bacterial endotoxin by mounting an acute inflammatory response. Together, these results support the concept of NFI-A as a master molecular transcriptome switch that controls myeloid cell differentiation and maturation and that malfunction of this switch during sepsis promotes MDSC expansion that adversely impacts sepsis outcome.


Asunto(s)
Células Mieloides/metabolismo , Factores de Transcripción NFI/deficiencia , Sepsis/genética , Sepsis/mortalidad , Animales , Biomarcadores , Citocinas/sangre , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Marcación de Gen , Vectores Genéticos/genética , Inmunidad , Inmunomodulación , Inmunofenotipificación , Recuento de Leucocitos , Leucocitos/inmunología , Leucocitos/metabolismo , Lipopolisacáridos/inmunología , Masculino , Ratones , Ratones Noqueados , Células Mieloides/inmunología , Células Supresoras de Origen Mieloide/inmunología , Células Supresoras de Origen Mieloide/metabolismo , Fenotipo , Sepsis/inmunología
9.
Biol Chem ; 397(11): 1173-1185, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27276529

RESUMEN

The protein inhibitor of activated STAT1 (PIAS1) plays important roles in regulating virus-induced chronic hepatitis, but the interaction between hepatitis B virus (HBV) and hPIAS1 is not clear. Our aim was to verify if HBV encoding proteins enhance the transcription of hPIAS1 and which cis-elements and transcription factors were involved in the mechanism. In order to do, so a series of molecular biological methods, along with functional and histological studies, were performed. We found that the HBV surface protein (HBs) enhanced hPIAS1 transcription through the activities of TAL1, E47, myogenin (MYOG), and NFI, dependent on the activation of p38MAPK and ERK signaling pathways in vitro, which might contribute to the ineffectiveness of treatment in CHB patients. Furthermore, liver samples from patients with high HBsAg levels and HBV DNA displayed increased hPIAS1 expression and high levels of TAL1, E47, MYOG, and NFI, compared to those patients with low HBsAg levels and HBV DNA, and healthy controls. These findings suggest that the HBs protein-induced hPIAS1 transcription requires TAL1, E47, MYOG, NFI, and MAPK signal pathways. It provides new potential targets for antiviral therapeutic strategies for controlling HBV-associated diseases.


Asunto(s)
Virus de la Hepatitis B/metabolismo , Sistema de Señalización de MAP Quinasas , Proteínas Nucleares/metabolismo , Proteínas Inhibidoras de STAT Activados/genética , Transcripción Genética , Proteínas del Envoltorio Viral/metabolismo , Adulto , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/deficiencia , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Células CHO , Cricetinae , Cricetulus , Femenino , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Células Hep G2 , Antígenos de Superficie de la Hepatitis B/metabolismo , Virus de la Hepatitis B/fisiología , Humanos , Masculino , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Miogenina/genética , Miogenina/metabolismo , Factores de Transcripción NFI/deficiencia , Factores de Transcripción NFI/genética , Factores de Transcripción NFI/metabolismo , Proteínas Nucleares/deficiencia , Proteínas Nucleares/genética , Fosforilación , Regiones Promotoras Genéticas/genética , Proteínas Proto-Oncogénicas/deficiencia , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteína 1 de la Leucemia Linfocítica T Aguda , Factor de Transcripción 3/deficiencia , Factor de Transcripción 3/genética , Factor de Transcripción 3/metabolismo
10.
Brain Res ; 1616: 71-87, 2015 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-25960350

RESUMEN

Nuclear factor one X (NFIX) has been shown to play a pivotal role during the development of many regions of the brain, including the neocortex, the hippocampus and the cerebellum. Mechanistically, NFIX has been shown to promote neural stem cell differentiation through the activation of astrocyte-specific genes and via the repression of genes central to progenitor cell self-renewal. Interestingly, mice lacking Nfix also exhibit other phenotypes with respect to development of the central nervous system, and whose underlying causes have yet to be determined. Here we examine one of the phenotypes displayed by Nfix(-/-) mice, namely hydrocephalus. Through the examination of embryonic and postnatal Nfix(-/-) mice we reveal that hydrocephalus is first seen at around postnatal day (P) 10 in mice lacking Nfix, and is fully penetrant by P20. Furthermore, we examined the subcommissural organ (SCO), the Sylvian aqueduct and the ependymal layer of the lateral ventricles, regions that when malformed and functionally perturbed have previously been implicated in the development of hydrocephalus. SOX3 is a factor known to regulate SCO development. Although we revealed that NFIX could repress Sox3-promoter-driven transcriptional activity in vitro, SOX3 expression within the SCO was normal within Nfix(-/-) mice, and Nfix mutant mice showed no abnormalities in the structure or function of the SCO. Moreover, these mutant mice exhibited no overt blockage of the Sylvian aqueduct. However, the ependymal layer of the lateral ventricles was frequently absent in Nfix(-/-) mice, suggesting that this phenotype may underlie the development of hydrocephalus within these knockout mice.


Asunto(s)
Epéndimo/patología , Regulación del Desarrollo de la Expresión Génica/genética , Hidrocefalia/patología , Ventrículos Laterales/patología , Factores de Transcripción NFI/deficiencia , Factores de Edad , Animales , Animales Recién Nacidos , Biología Computacional , Modelos Animales de Enfermedad , Embrión de Mamíferos , Epéndimo/embriología , Epéndimo/crecimiento & desarrollo , Hidrocefalia/genética , Ventrículos Laterales/embriología , Ventrículos Laterales/crecimiento & desarrollo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Factores de Transcripción NFI/genética , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo
11.
Liver Int ; 35(4): 1185-94, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25293436

RESUMEN

BACKGROUND & AIMS: Knockout studies of the murine Nuclear Factor I-C (NFI-C) transcription factor revealed abnormal skin wound healing and growth of its appendages, suggesting a role in controlling cell proliferation in adult regenerative processes. Liver regeneration following partial hepatectomy (PH) is a well-established regenerative model whereby changes elicited in hepatocytes lead to their rapid and phased proliferation. Although NFI-C is highly expressed in the liver, no hepatic function was yet established for this transcription factor. This study aimed to determine whether NFI-C may play a role in hepatocyte proliferation and liver regeneration. METHODS: Liver regeneration and cell proliferation pathways following two-thirds PH were investigated in NFI-C knockout (ko) and wild-type (wt) mice. RESULTS: We show that the absence of NFI-C impaired hepatocyte proliferation because of plasminogen activator I (PAI-1) overexpression and the subsequent suppression of urokinase plasminogen activator (uPA) activity and hepatocyte growth factor (HGF) signalling, a potent hepatocyte mitogen. This indicated that NFI-C first acts to promote hepatocyte proliferation at the onset of liver regeneration in wt mice. The subsequent transient down regulation of NFI-C, as can be explained by a self-regulatory feedback loop with transforming growth factor beta 1 (TGF-ß1), may limit the number of hepatocytes entering the first wave of cell division and/or prevent late initiations of mitosis. CONCLUSION: NFI-C acts as a regulator of the phased hepatocyte proliferation during liver regeneration.


Asunto(s)
Proliferación Celular , Regeneración Hepática , Hígado/metabolismo , Factores de Transcripción NFI/metabolismo , Animales , Retroalimentación Fisiológica , Regulación de la Expresión Génica , Genotipo , Hepatectomía/métodos , Factor de Crecimiento de Hepatocito/genética , Factor de Crecimiento de Hepatocito/metabolismo , Hígado/patología , Hígado/fisiopatología , Hígado/cirugía , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Factores de Transcripción NFI/deficiencia , Factores de Transcripción NFI/genética , Fenotipo , Inhibidor 1 de Activador Plasminogénico/genética , Inhibidor 1 de Activador Plasminogénico/metabolismo , Transducción de Señal , Factores de Tiempo , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo , Activador de Plasminógeno de Tipo Uroquinasa/genética , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo
12.
Blood ; 122(17): 2987-96, 2013 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-24041575

RESUMEN

Hematopoietic stem cells are both necessary and sufficient to sustain the complete blood system of vertebrates. Here we show that Nfix, a member of the nuclear factor I (Nfi) family of transcription factors, is highly expressed by hematopoietic stem and progenitor cells (HSPCs) of murine adult bone marrow. Although short hairpin RNA-mediated knockdown of Nfix expression in Lineage(-)Sca-1(+)c-Kit(+) HSPCs had no effect on in vitro cell growth or viability, Nfix-depleted HSPCs displayed a significant loss of colony-forming potential, as well as short- and long-term in vivo hematopoietic repopulating activity. Analysis of recipient mice at 4 to 20 days posttransplant revealed that Nfix-depleted HSPCs are established in the bone marrow, but fail to persist due to increased apoptotic cell death. Gene expression profiling of Nfix-depleted HSPCs reveals that loss of Nfix expression in HSPCs is concomitant with a decrease in the expression of multiple genes known to be important for HSPCs survival, such as Erg, Mecom, and Mpl. These data reveal that Nfix is a novel regulator of HSPCs survival posttransplantation and establish a role for Nfi genes in the regulation of this cellular compartment.


Asunto(s)
Células Madre Adultas/metabolismo , Células de la Médula Ósea/metabolismo , Hematopoyesis/genética , Células Madre Hematopoyéticas/metabolismo , Factores de Transcripción NFI/genética , Células Madre Adultas/citología , Animales , Antígenos Ly/genética , Antígenos Ly/metabolismo , Apoptosis , Células de la Médula Ósea/citología , Supervivencia Celular , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Factores de Transcripción NFI/deficiencia , Factores de Transcripción NFI/metabolismo , Proteínas Oncogénicas/genética , Proteínas Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-kit/genética , Proteínas Proto-Oncogénicas c-kit/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Receptores de Trombopoyetina/genética , Receptores de Trombopoyetina/metabolismo , Transducción de Señal , Factores de Transcripción , Regulador Transcripcional ERG
13.
Nature ; 495(7439): 98-102, 2013 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-23389444

RESUMEN

Adult stem cells reside in specialized niches where they receive environmental cues to maintain tissue homeostasis. In mammals, the stem cell niche within hair follicles is home to epithelial hair follicle stem cells and melanocyte stem cells, which sustain cyclical bouts of hair regeneration and pigmentation. To generate pigmented hairs, synchrony is achieved such that upon initiation of a new hair cycle, stem cells of each type activate lineage commitment. Dissecting the inter-stem-cell crosstalk governing this intricate coordination has been difficult, because mutations affecting one lineage often affect the other. Here we identify transcription factor NFIB as an unanticipated coordinator of stem cell behaviour. Hair follicle stem-cell-specific conditional targeting of Nfib in mice uncouples stem cell synchrony. Remarkably, this happens not by perturbing hair cycle and follicle architecture, but rather by promoting melanocyte stem cell proliferation and differentiation. The early production of melanin is restricted to melanocyte stem cells at the niche base. Melanocyte stem cells more distant from the dermal papilla are unscathed, thereby preventing hair greying typical of melanocyte stem cell differentiation mutants. Furthermore, we pinpoint KIT-ligand as a dermal papilla signal promoting melanocyte stem cell differentiation. Additionally, through chromatin-immunoprecipitation with high-throughput-sequencing and transcriptional profiling, we identify endothelin 2 (Edn2) as an NFIB target aberrantly activated in NFIB-deficient hair follicle stem cells. Ectopically induced Edn2 recapitulates NFIB-deficient phenotypes in wild-type mice. Conversely, endothelin receptor antagonists and/or KIT blocking antibodies prevent precocious melanocyte stem cell differentiation in the NFIB-deficient niche. Our findings reveal how melanocyte and hair follicle stem cell behaviours maintain reliance upon cooperative factors within the niche, and how this can be uncoupled in injury, stress and disease states.


Asunto(s)
Folículo Piloso/citología , Melanocitos/citología , Factores de Transcripción NFI/metabolismo , Nicho de Células Madre , Células Madre/citología , Células Madre/metabolismo , Animales , Apoptosis , Diferenciación Celular , Proliferación Celular , Inmunoprecipitación de Cromatina , Endotelina-2/genética , Endotelina-2/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Cabello/citología , Cabello/crecimiento & desarrollo , Color del Cabello , Folículo Piloso/metabolismo , Melanocitos/metabolismo , Ratones , Factores de Transcripción NFI/deficiencia , Factores de Transcripción NFI/genética , Análisis de Secuencia , Factor de Células Madre/metabolismo
14.
Int Immunol ; 23(6): 385-90, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21602176

RESUMEN

Nuclear factor I (NFI)-A is a member of the NFI family of transcription factors implicated in regulation of granulocyte differentiation. However, its role in the lymphoid lineage is not known. NFI-A deficiency results in perinatal lethality, thus precluding analysis of the role of NFI-A in lymphocyte development and function. Using recombination activation gene-2-deficient (RAG-2(-/-)) blastocysts and embryonic stem cells with homozygous NFI-A gene deletion, we show an essential role for NFI-A in T-cell activation. NFI-A(-/-)→RAG-2(-/-) chimeric mice had normal distributions of CD4(-)CD8(-) double negative, CD4(+)CD8(+) double positive, CD4(+)CD8(-) and CD4(-)CD8(+)-single positive cells in the thymus and CD4(+)CD8(-) and CD4(-)CD8(+) cells in spleen and lymph nodes. However, NFI-A(-/-)→RAG-2(-)(/)(-) mice had severely reduced thymus size and hypocellularity. The decrease in thymocytes and peripheral T cells in NFI-A(-/-)→RAG-2(-/-) chimeric mice is attributed to proliferative defects associated with decreased blast transformation, CD69 expression and DNA synthesis in response to T antigen receptor stimulation. Interestingly, NFI-A-null T cells showed increased levels of c-myc transcription that is inhibited in response to antigen receptor-mediated activation. These studies demonstrate for the first time a requirement for the NFI-A transcription factor in antigen receptor-induced T-cell activation events.


Asunto(s)
Proteínas de Unión al ADN/deficiencia , Activación de Linfocitos/genética , Factores de Transcripción NFI/metabolismo , Linfocitos T/inmunología , Animales , Ciclo Celular/inmunología , Proliferación Celular , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/inmunología , Prueba de Complementación Genética , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factores de Transcripción NFI/deficiencia , Factores de Transcripción NFI/genética , Linfocitos T/citología , Linfocitos T/metabolismo
15.
J Neurosci ; 30(27): 9127-39, 2010 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-20610746

RESUMEN

The balance between self-renewal and differentiation of neural progenitor cells is an absolute requirement for the correct formation of the nervous system. Much is known about both the pathways involved in progenitor cell self-renewal, such as Notch signaling, and the expression of genes that initiate progenitor differentiation. However, whether these fundamental processes are mechanistically linked, and specifically how repression of progenitor self-renewal pathways occurs, is poorly understood. Nuclear factor I A (Nfia), a gene known to regulate spinal cord and neocortical development, has recently been implicated as acting downstream of Notch to initiate the expression of astrocyte-specific genes within the cortex. Here we demonstrate that, in addition to activating the expression of astrocyte-specific genes, Nfia also downregulates the activity of the Notch signaling pathway via repression of the key Notch effector Hes1. These data provide a significant conceptual advance in our understanding of neural progenitor differentiation, revealing that a single transcription factor can control both the activation of differentiation genes and the repression of the self-renewal genes, thereby acting as a pivotal regulator of the balance between progenitor and differentiated cell states.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular/genética , Regulación del Desarrollo de la Expresión Génica/fisiología , Proteínas de Homeodominio/metabolismo , Factores de Transcripción NFI/fisiología , Células Madre/fisiología , Telencéfalo/citología , Factores de Edad , Análisis de Varianza , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Bromodesoxiuridina/metabolismo , Recuento de Células/métodos , Ventrículos Cerebrales/citología , Ventrículos Cerebrales/embriología , Inmunoprecipitación de Cromatina/métodos , Ensayo de Cambio de Movilidad Electroforética/métodos , Embrión de Mamíferos , Regulación del Desarrollo de la Expresión Génica/genética , Hipocampo/citología , Hipocampo/crecimiento & desarrollo , Proteínas de Homeodominio/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis por Micromatrices/métodos , Mutación/genética , Factores de Transcripción NFI/deficiencia , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Factor 6 de Transcripción de Unión a Octámeros/genética , Factor 6 de Transcripción de Unión a Octámeros/metabolismo , Regiones Promotoras Genéticas/fisiología , Receptores de Ácido Kaínico/genética , Receptores de Ácido Kaínico/metabolismo , Telencéfalo/embriología , Factor de Transcripción HES-1 , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
16.
Neural Dev ; 4: 43, 2009 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-19961580

RESUMEN

BACKGROUND: Agenesis of the corpus callosum is associated with many human developmental syndromes. Key mechanisms regulating callosal formation include the guidance of axons arising from pioneering neurons in the cingulate cortex and the development of cortical midline glial populations, but their molecular regulation remains poorly characterised. Recent data have shown that mice lacking the transcription factor Nfib exhibit callosal agenesis, yet neocortical callosal neurons express only low levels of Nfib. Therefore, we investigate here how Nfib functions to regulate non-cell-autonomous mechanisms of callosal formation. RESULTS: Our investigations confirmed a reduction in glial cells at the midline in Nfib-/- mice. To determine how this occurs, we examined radial progenitors at the cortical midline and found that they were specified correctly in Nfib mutant mice, but did not differentiate into mature glia. Cellular proliferation and apoptosis occurred normally at the midline of Nfib mutant mice, indicating that the decrease in midline glia observed was due to deficits in differentiation rather than proliferation or apoptosis. Next we investigated the development of callosal pioneering axons in Nfib-/- mice. Using retrograde tracer labelling, we found that Nfib is expressed in cingulate neurons and hence may regulate their development. In Nfib-/- mice, neuropilin 1-positive axons fail to cross the midline and expression of neuropilin 1 is diminished. Tract tracing and immunohistochemistry further revealed that, in late gestation, a minor population of neocortical axons does cross the midline in Nfib mutants on a C57Bl/6J background, forming a rudimentary corpus callosum. Finally, the development of other forebrain commissures in Nfib-deficient mice is also aberrant. CONCLUSION: The formation of the corpus callosum is severely delayed in the absence of Nfib, despite Nfib not being highly expressed in neocortical callosal neurons. Our results indicate that in addition to regulating the development of midline glial populations, Nfib also regulates the expression of neuropilin 1 within the cingulate cortex. Collectively, these data indicate that defects in midline glia and cingulate cortex neurons are associated with the callosal dysgenesis seen in Nfib-deficient mice, and provide insight into how the development of these cellular populations is controlled at a molecular level.


Asunto(s)
Cuerpo Calloso/embriología , Cuerpo Calloso/fisiopatología , Factores de Transcripción NFI/metabolismo , Neocórtex/embriología , Neocórtex/fisiopatología , Animales , Apoptosis/fisiología , Axones/fisiología , Diferenciación Celular/fisiología , Movimiento Celular/fisiología , Proliferación Celular , Giro del Cíngulo/embriología , Giro del Cíngulo/fisiopatología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Ratones , Ratones Noqueados , Factores de Transcripción NFI/deficiencia , Factores de Transcripción NFI/genética , Proteínas del Tejido Nervioso/metabolismo , Neuroglía/fisiología , Neuronas/fisiología , Neuropilina-1/metabolismo , Prosencéfalo/embriología , Prosencéfalo/fisiopatología , Células Madre/fisiología
17.
Mol Cell Biol ; 29(22): 6006-17, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19752192

RESUMEN

Transforming growth factor beta (TGF-beta) and platelet-derived growth factor A (PDGFAlpha) play a central role in tissue morphogenesis and repair, but their interplay remain poorly understood. The nuclear factor I C (NFI-C) transcription factor has been implicated in TGF-beta signaling, extracellular matrix deposition, and skin appendage pathologies, but a potential role in skin morphogenesis or healing had not been assessed. To evaluate this possibility, we performed a global gene expression analysis in NFI-C(-/-) and wild-type embryonic primary murine fibroblasts. This indicated that NFI-C acts mostly to repress gene expression in response to TGF-beta1. Misregulated genes were prominently overrepresented by regulators of connective tissue inflammation and repair. In vivo skin healing revealed a faster inflammatory stage and wound closure in NFI-C(-/-) mice. Expression of PDGFA and PDGF-receptor alpha were increased in wounds of NFI-C(-/-) mice, explaining the early recruitment of macrophages and fibroblasts. Differentiation of fibroblasts to contractile myofibroblasts was also elevated, providing a rationale for faster wound closure. Taken together with the role of TGF-beta in myofibroblast differentiation, our results imply a central role of NFI-C in the interplay of the two signaling pathways and in regulation of the progression of tissue regeneration.


Asunto(s)
Factores de Transcripción NFI/metabolismo , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal , Piel/patología , Factor de Crecimiento Transformador beta1/metabolismo , Cicatrización de Heridas , Animales , Diferenciación Celular , Movimiento Celular , Células Cultivadas , Embrión de Mamíferos/citología , Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Eliminación de Gen , Regulación de la Expresión Génica , Macrófagos/citología , Ratones , Ratones Noqueados , Factores de Transcripción NFI/deficiencia , Análisis de Secuencia por Matrices de Oligonucleótidos , Reproducibilidad de los Resultados , Piel/metabolismo
18.
J Biol Chem ; 284(25): 17293-17303, 2009 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-19386589

RESUMEN

Our previous studies have demonstrated that nuclear factor I-C (NFI-C) null mice developed short molar roots that contain aberrant odontoblasts and abnormal dentin formation. Based on these findings, we performed studies to elucidate the function of NFI-C in odontoblasts. Initial studies demonstrated that aberrant odontoblasts become dissociated and trapped in an osteodentin-like mineralized tissue. Abnormal odontoblasts exhibit strong bone sialoprotein expression but a decreased level of dentin sialophosphoprotein expression when compared with wild type odontoblasts. Loss of Nfic results in an increase in p-Smad2/3 expression in aberrant odontoblasts and pulp cells in the subodontoblastic layer in vivo and primary pulp cells from Nfic-deficient mice in vitro. Cell proliferation analysis of both cervical loop and ectomesenchymal cells of the Nfic-deficient mice revealed significantly decreased proliferative activity compared with wild type mice. In addition, Nfic-deficient primary pulp cells showed increased expression of p21 and p16 but decreased expression of cyclin D1 and cyclin B1, strongly suggesting cell growth arrest caused by a lack of Nfic activity. Analysis of the pulp and abnormal dentin in Nfic-deficient mice revealed an increase in apoptotic activity. Further, Nfic-deficient primary pulp cells exhibited an increase in caspase-8 and -3 activation, whereas the cleaved form of Bid was hardly detected. These results indicate that the loss of Nfic leads to the suppression of odontogenic cell proliferation and differentiation and induces apoptosis of aberrant odontoblasts during root formation, thereby contributing to the formation of short roots.


Asunto(s)
Factores de Transcripción NFI/fisiología , Odontoblastos/citología , Odontoblastos/metabolismo , Odontogénesis/fisiología , Raíz del Diente/crecimiento & desarrollo , Raíz del Diente/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Diferenciación Celular/fisiología , Proliferación Celular , Células Cultivadas , Cartilla de ADN/genética , Dentina/metabolismo , Regulación del Desarrollo de la Expresión Génica , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Factores de Transcripción NFI/antagonistas & inhibidores , Factores de Transcripción NFI/deficiencia , Factores de Transcripción NFI/genética , Odontogénesis/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Interferencia de ARN , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Raíz del Diente/citología
19.
J Neurosci ; 28(47): 12328-40, 2008 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-19020026

RESUMEN

The hippocampus plays an integral role in spatial navigation, learning and memory, and is a major site for adult neurogenesis. Critical to these functions is the proper organization of the hippocampus during development. Radial glia are known to regulate hippocampal formation, but their precise function in this process is yet to be defined. We find that in Nuclear Factor I b (Nfib)-deficient mice, a subpopulation of glia from the ammonic neuroepithelium of the hippocampus fail to develop. This results in severe morphological defects, including a failure of the hippocampal fissure, and subsequently the dentate gyrus, to form. As in wild-type mice, immature nestin-positive glia, which encompass all types of radial glia, populate the hippocampus in Nfib-deficient mice at embryonic day 15. However, these fail to mature into GLAST- and GFAP-positive glia, and the supragranular glial bundle is absent. In contrast, the fimbrial glial bundle forms, but alone is insufficient for proper hippocampal morphogenesis. Dentate granule neurons are present in the mutant hippocampus but their migration is aberrant, likely resulting from the lack of the complete radial glial scaffold usually provided by both glial bundles. These data demonstrate a role for Nfib in hippocampal fissure and dentate gyrus formation, and that distinct glial bundles are critical for correct hippocampal morphogenesis.


Asunto(s)
Hipocampo/citología , Hipocampo/embriología , Morfogénesis , Neuroglía/fisiología , Factores de Edad , Animales , Recuento de Células , Movimiento Celular/genética , Proliferación Celular , Células Cultivadas , Embrión de Mamíferos , Transportador 1 de Aminoácidos Excitadores/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica/genética , Proteína Ácida Fibrilar de la Glía , Ácido Glutámico/metabolismo , Histonas/metabolismo , Proteínas de Filamentos Intermediarios/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Morfogénesis/genética , Factores de Transcripción NFI/deficiencia , Proteínas del Tejido Nervioso/metabolismo , Nestina , Neuronas/metabolismo , Embarazo , Factores de Tiempo
20.
Genome Biol ; 8(5): R72, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17475010

RESUMEN

BACKGROUND: Nuclear factor I-A (NFI-A), a phylogenetically conserved transcription/replication protein, plays a crucial role in mouse brain development. Previous studies have shown that disruption of the Nfia gene in mice leads to perinatal lethality, corpus callosum agenesis, and hydrocephalus. RESULTS: To identify potential NFI-A target genes involved in the observed tissue malformations, we analyzed gene expression in brains from Nfia-/- and Nfia+/+ littermate mice at the mRNA level using oligonucleotide microarrays. In young postnatal animals (postnatal day 16), 356 genes were identified as being differentially regulated, whereas at the late embryonic stage (embryonic day 18) only five dysregulated genes were found. An in silico analysis identified phylogenetically conserved NFI binding sites in at least 70 of the differentially regulated genes. Moreover, assignment of gene function showed that marker genes for immature neural cells and neural precursors were expressed at elevated levels in young postnatal Nfia-/- mice. In contrast, marker genes for differentiated neural cells were downregulated at this stage. In particular, genes relevant for oligodendrocyte differentiation were affected. CONCLUSION: Our findings suggest that brain development, especially oligodendrocyte maturation, is delayed in Nfia-/- mice during the early postnatal period, which at least partly accounts for their phenotype. The identification of potential NFI-A target genes in our study should help to elucidate NFI-A dependent transcriptional pathways and contribute to enhanced understanding of this period of brain formation, especially with regard to the function of NFI-A.


Asunto(s)
Encéfalo/crecimiento & desarrollo , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Factores de Transcripción NFI/deficiencia , Animales , Sitios de Unión , Diferenciación Celular/genética , Ratones , Ratones Noqueados , Factores de Transcripción NFI/metabolismo , Factores de Transcripción NFI/fisiología , Neuronas/citología , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...