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.
Matrix Biol ; 112: 90-115, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35963565

RESUMEN

Obesity triggers skeletal muscle physio-pathological alterations. However, the crosstalk between adipose tissue and myogenic cells remains poorly understood during obesity. We identified NID-1 among the adipose tissue secreted factors impairing myogenic potential of human myoblasts and murine muscle stem cells in vitro. Mice under High Fat Diet (HFD) displayed increased NID-1 expression in the skeletal muscle endomysium associated with intramuscular fat adipose tissue expansion and compromised muscle stem cell function. We show that NID-1 is highly secreted by skeletal muscle fibro-adipogenic/mesenchymal progenitors (FAPs) during obesity. We demonstrate that increased muscle NID-1 impairs muscle stem cells proliferation and primes the fibrogenic differentiation of FAPs, giving rise to an excessive deposition of extracellular matrix. Finally, we propose a model in which obesity leads to skeletal muscle extracellular matrix remodeling by FAPs, mediating the alteration of myogenic function by adipose tissue and highlighting the key role of NID-1 in the crosstalk between adipose tissue and skeletal muscle.


Asunto(s)
Adipogénesis , Desarrollo de Músculos , Animales , Diferenciación Celular , Matriz Extracelular , Humanos , Ratones , Músculo Esquelético/metabolismo , Obesidad/genética , Obesidad/metabolismo
2.
Nat Commun ; 13(1): 1039, 2022 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-35210422

RESUMEN

The contractile properties of adult myofibers are shaped by their Myosin heavy chain isoform content. Here, we identify by snATAC-seq a 42 kb super-enhancer at the locus regrouping the fast Myosin genes. By 4C-seq we show that active fast Myosin promoters interact with this super-enhancer by DNA looping, leading to the activation of a single promoter per nucleus. A rainbow mouse transgenic model of the locus including the super-enhancer recapitulates the endogenous spatio-temporal expression of adult fast Myosin genes. In situ deletion of the super-enhancer by CRISPR/Cas9 editing demonstrates its major role in the control of associated fast Myosin genes, and deletion of two fast Myosin genes at the locus reveals an active competition of the promoters for the shared super-enhancer. Last, by disrupting the organization of fast Myosin, we uncover positional heterogeneity within limb skeletal muscles that may underlie selective muscle susceptibility to damage in certain myopathies.


Asunto(s)
Fibras Musculares Esqueléticas , Miosinas , Animales , Ratones , Ratones Transgénicos , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Cadenas Pesadas de Miosina/genética , Cadenas Pesadas de Miosina/metabolismo , Miosinas/genética , Miosinas/metabolismo , Fenotipo
3.
PLoS Genet ; 16(11): e1009164, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33175861

RESUMEN

The chromosome translocations generating PAX3-FOXO1 and PAX7-FOXO1 chimeric proteins are the primary hallmarks of the paediatric fusion-positive alveolar subtype of Rhabdomyosarcoma (FP-RMS). Despite the ability of these transcription factors to remodel chromatin landscapes and promote the expression of tumour driver genes, they only inefficiently promote malignant transformation in vivo. The reason for this is unclear. To address this, we developed an in ovo model to follow the response of spinal cord progenitors to PAX-FOXO1s. Our data demonstrate that PAX-FOXO1s, but not wild-type PAX3 or PAX7, trigger the trans-differentiation of neural cells into FP-RMS-like cells with myogenic characteristics. In parallel, PAX-FOXO1s remodel the neural pseudo-stratified epithelium into a cohesive mesenchyme capable of tissue invasion. Surprisingly, expression of PAX-FOXO1s, similar to wild-type PAX3/7, reduce the levels of CDK-CYCLIN activity and increase the fraction of cells in G1. Introduction of CYCLIN D1 or MYCN overcomes this PAX-FOXO1-mediated cell cycle inhibition and promotes tumour growth. Together, our findings reveal a mechanism that can explain the apparent limited oncogenicity of PAX-FOXO1 fusion transcription factors. They are also consistent with certain clinical reports indicative of a neural origin of FP-RMS.


Asunto(s)
Transdiferenciación Celular/genética , Transformación Celular Neoplásica/genética , Proteínas de Fusión Oncogénica/metabolismo , Factores de Transcripción Paired Box/metabolismo , Rabdomiosarcoma Alveolar/genética , Animales , Biopsia , Embrión de Pollo , Niño , Ciclina D1/genética , Conjuntos de Datos como Asunto , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Proteína Proto-Oncogénica N-Myc/genética , Invasividad Neoplásica/genética , Células-Madre Neurales/patología , Tubo Neural/citología , Proteínas de Fusión Oncogénica/genética , Factor de Transcripción PAX3/genética , Factor de Transcripción PAX3/metabolismo , Factor de Transcripción PAX7/genética , Factor de Transcripción PAX7/metabolismo , Factores de Transcripción Paired Box/genética , Rabdomiosarcoma Alveolar/patología , Fase S/genética
4.
BMC Med Genet ; 21(1): 182, 2020 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-32943010

RESUMEN

BACKGROUND: Mandibulofacial dysostosis with microcephaly (MFDM) is a rare autosomal dominant genetic disease characterized by intellectual and growth retardations, as well as major microcephaly, induced by missense and splice site variants or microdeletions in the EFTUD2 gene. CASE PRESENTATION: Here, we investigate the case of a young girl with symptoms of MFDM and a normal karyotype. Whole-exome sequencing of the family was performed to identify genetic alterations responsible for this phenotype. We identified a de novo synonymous variant in the EFTUD2 gene. We demonstrated that this synonymous variant disrupts the donor splice-site in intron 9 resulting in the skipping of exon 9 and a frameshift that leads to a premature stop codon. CONCLUSIONS: We present the first case of MFDM caused by a synonymous variant disrupting the donor splice site, leading to exon skipping.


Asunto(s)
Disostosis Mandibulofacial/genética , Microcefalia/genética , Mutación , Factores de Elongación de Péptidos/genética , Empalme del ARN , Ribonucleoproteína Nuclear Pequeña U5/genética , Secuencia de Bases , Niño , Femenino , Humanos , Cariotipo , Fenotipo
5.
Cell Stem Cell ; 24(6): 958-973.e9, 2019 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-31006622

RESUMEN

Muscle satellite cells (MuSCs) are the quiescent muscle stem cells required for adult skeletal muscle repair. The impact of environmental stress such as pollution on MuSC behavior remains unexplored. We evaluated the impact of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) exposure, a ubiquitous and highly toxic pollutant, on MuSCs by combining in vivo mouse molecular genetic models with ex vivo studies. While all MuSCs express the transcription factor PAX7, we show that a subset also express PAX3 and exhibit resistance to environmental stress. Upon systemic TCDD treatment, PAX3-negative MuSCs display impaired survival, atypical activation, and sporadic differentiation through xenobiotic aryl hydrocarbon receptor signaling. We further show that PAX3-positive MuSCs become sensitized to environmental stress when PAX3 function is impaired and that PAX3-mediated induction of mTORC1 is required for protection. Our study, therefore, identifies a functional heterogeneity of MuSCs in response to environmental stress controlled by PAX3.


Asunto(s)
Células Madre Adultas/fisiología , Contaminación Ambiental/efectos adversos , Factor de Transcripción PAX3/metabolismo , Factor de Transcripción PAX7/metabolismo , Dibenzodioxinas Policloradas/efectos adversos , Células Satélite del Músculo Esquelético/fisiología , Animales , Diferenciación Celular , Supervivencia Celular , Células Cultivadas , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor de Transcripción PAX3/genética , Factor de Transcripción PAX7/genética , Receptores de Hidrocarburo de Aril/metabolismo , Transducción de Señal
6.
Elife ; 72018 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-29882512

RESUMEN

Muscle satellite cells are the primary source of stem cells for postnatal skeletal muscle growth and regeneration. Understanding genetic control of satellite cell formation, maintenance, and acquisition of their stem cell properties is on-going, and we have identified SOXF (SOX7, SOX17, SOX18) transcriptional factors as being induced during satellite cell specification. We demonstrate that SOXF factors regulate satellite cell quiescence, self-renewal and differentiation. Moreover, ablation of Sox17 in the muscle lineage impairs postnatal muscle growth and regeneration. We further determine that activities of SOX7, SOX17 and SOX18 overlap during muscle regeneration, with SOXF transcriptional activity requisite. Finally, we show that SOXF factors also control satellite cell expansion and renewal by directly inhibiting the output of ß-catenin activity, including inhibition of Ccnd1 and Axin2. Together, our findings identify a key regulatory function of SoxF genes in muscle stem cells via direct transcriptional control and interaction with canonical Wnt/ß-catenin signaling.


Asunto(s)
Autorrenovación de las Células , Proteínas HMGB/metabolismo , Factores de Transcripción SOXF/metabolismo , Células Satélite del Músculo Esquelético/metabolismo , beta Catenina/metabolismo , Animales , Proteína Axina/metabolismo , Línea Celular , Ciclina D1/metabolismo , Proteínas HMGB/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Factores de Transcripción SOXF/genética , Células Satélite del Músculo Esquelético/citología , Vía de Señalización Wnt
7.
Dev Biol ; 432(1): 24-33, 2017 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-28625870

RESUMEN

Transcription factors are key orchestrators of the emergence of neuronal diversity within the developing spinal cord. As such, the two paralogous proteins Pax3 and Pax7 regulate the specification of progenitor cells within the intermediate neural tube, by defining a neat segregation between those fated to form motor circuits and those involved in the integration of sensory inputs. To attain insights into the molecular means by which they control this process, we have performed detailed phenotypic analyses of the intermediate spinal interneurons (IN), namely the dI6, V0D, V0VCG and V1 populations in compound null mutants for Pax3 and Pax7. This has revealed that the levels of Pax3/7 proteins determine both the dorso-ventral extent and the number of cells produced in each subpopulation; with increasing levels leading to the dorsalisation of their fate. Furthermore, thanks to the examination of mutants in which Pax3 transcriptional activity is skewed either towards repression or activation, we demonstrate that this cell diversification process is mainly dictated by Pax3/7 ability to repress gene expression. Consistently, we show that Pax3 and Pax7 inhibit the expression of Dbx1 and of its repressor Prdm12, fate determinants of the V0 and V1 interneurons, respectively. Notably, we provide evidence for the activity of several cis-regulatory modules of Dbx1 to be sensitive to Pax3 and Pax7 transcriptional activity levels. Altogether, our study provides insights into how the redundancy within a TF family, together with discrete dynamics of expression profiles of each member, are exploited to generate cellular diversity. Furthermore, our data supports the model whereby cell fate choices in the neural tube do not rely on binary decisions but rather on inhibition of multiple alternative fates.


Asunto(s)
Proteínas de Homeodominio/fisiología , Interneuronas/fisiología , Proteínas del Tejido Nervioso/fisiología , Factor de Transcripción PAX3/fisiología , Factor de Transcripción PAX7/fisiología , Médula Espinal/citología , Animales , Diferenciación Celular/fisiología , Embrión de Pollo , Regulación del Desarrollo de la Expresión Génica , Interneuronas/citología , Ratones , Tubo Neural/fisiología , Médula Espinal/embriología , Células Madre/citología , Células Madre/fisiología
8.
Genesis ; 55(4)2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28196404

RESUMEN

p57Kip2 (p57) is a maternally expressed imprinted gene regulating growth arrest which belongs to the CIP/KIP family of cyclin-dependent kinase inhibitors. While initially identified as a cell cycle arrest protein through inhibition of cyclin and cyclin-dependent kinase complexes, p57 activity has also been linked to differentiation, apoptosis, and senescence. In addition, p57 has recently been shown to be involved in tumorigenesis and cell fate decisions in stem cells. Yet, p57 function in adult tissues remains poorly characterized due to the perinatal lethality of p57 knock-out mice. To analyze p57 tissue-specific activity, we generated a conditional mouse line (p57FL-ILZ/+ ) by flanking the coding exons 2-3 by LoxP sites. To track p57-expressing or mutant cells, the p57FL-ILZ allele also contains an IRES-linked ß-galactosidase reporter inserted in the 3' UTR of the gene. Here, we show that the ß-galactosidase reporter expression pattern recapitulates p57 tissue specificity during development and in postnatal mice. Furthermore, we crossed the p57FL-ILZ/+ mice with PGK-Cre mice to generate p57cKO-ILZ/+ animals with ubiquitous loss of p57. p57cKO-ILZ/+ mice display developmental phenotypes analogous to previously described p57 knock-outs. Thus, p57FL-ILZ/+ is a new genetic tool allowing expression and functional conditional analyses of p57.


Asunto(s)
Inhibidor p57 de las Quinasas Dependientes de la Ciclina/genética , Marcación de Gen/métodos , Mutación , Alelos , Animales , Células Cultivadas , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/metabolismo , Embrión de Mamíferos/metabolismo , Femenino , Genes Reporteros , Masculino , Ratones , Ratones Endogámicos C57BL , Especificidad de Órganos , Fenotipo , beta-Galactosidasa/genética , beta-Galactosidasa/metabolismo
9.
Front Cell Dev Biol ; 4: 58, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27446912

RESUMEN

Skeletal muscle growth and regeneration require a population of muscle stem cells, the satellite cells, located in close contact to the myofiber. These cells are specified during fetal and early postnatal development in mice from a Pax3/7 population of embryonic progenitor cells. As little is known about the genetic control of their formation and maintenance, we performed a genome-wide chronological expression profile identifying the dynamic transcriptomic changes involved in establishment of muscle stem cells through life, and acquisition of muscle stem cell properties. We have identified multiple genes and pathways associated with satellite cell formation, including set of genes specifically induced (EphA1, EphA2, EfnA1, EphB1, Zbtb4, Zbtb20) or inhibited (EphA3, EphA4, EphA7, EfnA2, EfnA3, EfnA4, EfnA5, EphB2, EphB3, EphB4, EfnBs, Zfp354c, Zcchc5, Hmga2) in adult stem cells. Ephrin receptors and ephrins ligands have been implicated in cell migration and guidance in many tissues including skeletal muscle. Here we show that Ephrin receptors and ephrins ligands are also involved in regulating the adult myogenic program. Strikingly, impairment of EPHB1 function in satellite cells leads to increased differentiation at the expense of self-renewal in isolated myofiber cultures. In addition, we identified new transcription factors, including several zinc finger proteins. ZFP354C and ZCCHC5 decreased self-renewal capacity when overexpressed, whereas ZBTB4 increased it, and ZBTB20 induced myogenic progression. The architectural and transcriptional regulator HMGA2 was involved in satellite cell activation. Together, our study shows that transcriptome profiling coupled with myofiber culture analysis, provides an efficient system to identify and validate candidate genes implicated in establishment/maintenance of muscle stem cells. Furthermore, tour de force transcriptomic profiling provides a wealth of data to inform for future stem cell-based muscle therapies.

10.
Dev Cell ; 33(1): 56-66, 2015 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-25800090

RESUMEN

Exposure to environmental teratogenic pollutant leads to severe birth defects. However, the biological events underlying these developmental abnormalities remain undefined. Here, we report a molecular link between an environmental stress response pathway and key developmental genes during craniofacial development. Strikingly, mutant mice with impaired Pax3/7 function display severe craniofacial defects. We show that these are associated with an upregulation of the signaling pathway mediated by the Aryl hydrocarbon receptor (AHR), the receptor to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), revealing a genetic interaction between Pax3 and AHR signaling. Activation of AHR signaling in Pax3-deficient embryos drives facial mesenchymal cells out of the cell cycle through the upregulation of p21 expression. Accordingly, inhibiting AHR activity rescues the cycling status of these cells and the facial closure of Pax3/7 mutants. Together, our findings demonstrate that the regulation of AHR signaling by Pax3/7 is required to protect against TCDD/AHR-mediated teratogenesis during craniofacial development.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Anomalías Congénitas/prevención & control , Anomalías Craneofaciales/prevención & control , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Contaminantes Ambientales/toxicidad , Factor de Transcripción PAX7/fisiología , Factores de Transcripción Paired Box/fisiología , Receptores de Hidrocarburo de Aril/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Western Blotting , Linaje de la Célula/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Anomalías Congénitas/etiología , Anomalías Craneofaciales/inducido químicamente , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Embrión de Mamíferos/citología , Embrión de Mamíferos/efectos de los fármacos , Embrión de Mamíferos/metabolismo , Femenino , Perfilación de la Expresión Génica , Ratones , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos , Factor de Transcripción PAX3 , Dibenzodioxinas Policloradas/análogos & derivados , Dibenzodioxinas Policloradas/toxicidad , Embarazo , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Hidrocarburo de Aril/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
11.
PLoS One ; 10(2): e0117370, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25658845

RESUMEN

Xenopus is an excellent tetrapod model for studying normal and pathological motoneuron ontogeny due to its developmental morpho-physiological advantages. In mammals, the urotensin II-related peptide (UTS2B) gene is primarily expressed in motoneurons of the brainstem and the spinal cord. Here, we show that this expression pattern was conserved in Xenopus and established during the early embryonic development, starting at the early tailbud stage. In late tadpole stage, uts2b mRNA was detected both in the hindbrain and in the spinal cord. Spinal uts2b+ cells were identified as axial motoneurons. In adult, however, the uts2b expression was only detected in the hindbrain. We assessed the ability of the uts2b promoter to drive the expression of a fluorescent reporter in motoneurons by recombineering a green fluorescent protein (GFP) into a bacterial artificial chromosome (BAC) clone containing the entire X. tropicalis uts2b locus. After injection of this construction in one-cell stage embryos, a transient GFP expression was observed in the spinal cord of about a quarter of the resulting animals from the early tailbud stage and up to juveniles. The GFP expression pattern was globally consistent with that of the endogenous uts2b in the spinal cord but no fluorescence was observed in the brainstem. A combination of histological and electrophysiological approaches was employed to further characterize the GFP+ cells in the larvae. More than 98% of the GFP+ cells expressed choline acetyltransferase, while their projections were co-localized with α-bungarotoxin labeling. When tail myotomes were injected with rhodamine dextran amine crystals, numerous double-stained GFP+ cells were observed. In addition, intracellular electrophysiological recordings of GFP+ neurons revealed locomotion-related rhythmic discharge patterns during fictive swimming. Taken together our results provide evidence that uts2b is an appropriate driver to express reporter genes in larval motoneurons of the Xenopus spinal cord.


Asunto(s)
Cromosomas Artificiales Bacterianos/metabolismo , Neuronas Motoras/metabolismo , Péptidos/metabolismo , Urotensinas/metabolismo , Xenopus/metabolismo , Animales , Animales Modificados Genéticamente/genética , Animales Modificados Genéticamente/metabolismo , Cromosomas Artificiales Bacterianos/genética , Fenómenos Electrofisiológicos , Embrión no Mamífero/metabolismo , Genes Reporteros , Hibridación in Situ , Microscopía Fluorescente , Péptidos/genética , Médula Espinal/metabolismo , Urotensinas/genética , Xenopus/crecimiento & desarrollo
12.
EMBO Mol Med ; 6(11): 1455-75, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25262827

RESUMEN

Mutations in amphiphysin-2/BIN1, dynamin 2, and myotubularin are associated with centronuclear myopathy (CNM), a muscle disorder characterized by myofibers with atypical central nuclear positioning and abnormal triads. Mis-splicing of amphiphysin-2/BIN1 is also associated with myotonic dystrophy that shares histopathological hallmarks with CNM. How amphiphysin-2 orchestrates nuclear positioning and triad organization and how CNM-associated mutations lead to muscle dysfunction remains elusive. We find that N-WASP interacts with amphiphysin-2 in myofibers and that this interaction and N-WASP distribution are disrupted by amphiphysin-2 CNM mutations. We establish that N-WASP functions downstream of amphiphysin-2 to drive peripheral nuclear positioning and triad organization during myofiber formation. Peripheral nuclear positioning requires microtubule/Map7/Kif5b-dependent distribution of nuclei along the myofiber and is driven by actin and nesprins. In adult myofibers, N-WASP and amphiphysin-2 are only involved in the maintenance of triad organization but not in the maintenance of peripheral nuclear positioning. Importantly, we confirmed that N-WASP distribution is disrupted in CNM and myotonic dystrophy patients. Our results support a role for N-WASP in amphiphysin-2-dependent nuclear positioning and triad organization and in CNM and myotonic dystrophy pathophysiology.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Músculo Esquelético/fisiopatología , Miopatías Estructurales Congénitas/fisiopatología , Proteínas Nucleares/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Proteína Neuronal del Síndrome de Wiskott-Aldrich/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Adulto , Humanos , Fibras Musculares Esqueléticas/metabolismo , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Proteínas Nucleares/genética , Proteínas Supresoras de Tumor/genética
13.
Development ; 141(14): 2780-90, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25005473

RESUMEN

A central question in development is to define how the equilibrium between cell proliferation and differentiation is temporally and spatially regulated during tissue formation. Here, we address how interactions between cyclin-dependent kinase inhibitors essential for myogenic growth arrest (p21(cip1) and p57(kip2)), the Notch pathway and myogenic regulatory factors (MRFs) orchestrate the proliferation, specification and differentiation of muscle progenitor cells. We first show that cell cycle exit and myogenic differentiation can be uncoupled. In addition, we establish that skeletal muscle progenitor cells require Notch signaling to maintain their cycling status. Using several mouse models combined with ex vivo studies, we demonstrate that Notch signaling is required to repress p21(cip1) and p57(kip2) expression in muscle progenitor cells. Finally, we identify a muscle-specific regulatory element of p57(kip2) directly activated by MRFs in myoblasts but repressed by the Notch targets Hes1/Hey1 in progenitor cells. We propose a molecular mechanism whereby information provided by Hes/Hey downstream of Notch as well as MRF activities are integrated at the level of the p57(kip2) enhancer to regulate the decision between progenitor cell maintenance and muscle differentiation.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteínas de Ciclo Celular/metabolismo , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/metabolismo , Proteínas de Homeodominio/metabolismo , Músculo Esquelético/crecimiento & desarrollo , Proteína MioD/metabolismo , Factor 5 Regulador Miogénico/metabolismo , Receptores Notch/metabolismo , Animales , Puntos de Control del Ciclo Celular , Diferenciación Celular , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Elementos de Facilitación Genéticos/genética , Extremidades/embriología , Regulación del Desarrollo de la Expresión Génica , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/metabolismo , Ratones , Ratones Transgénicos , Desarrollo de Músculos , Músculo Esquelético/citología , Músculo Esquelético/embriología , Músculo Esquelético/metabolismo , Proteína MioD/genética , Mioblastos/citología , Mioblastos/metabolismo , Especificidad de Órganos , Factor de Transcripción PAX7/metabolismo , Transducción de Señal , Células Madre/citología , Células Madre/metabolismo , Factor de Transcripción HES-1
14.
J Cell Sci ; 125(Pt 5): 1099-105, 2012 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22349700

RESUMEN

The position of the nucleus is regulated in different developmental stages and cellular events. During polarization, the nucleus moves away from the future leading edge and this movement is required for proper cell migration. Nuclear movement requires the LINC complex components nesprin-2G and SUN2, which form transmembrane actin-associated nuclear (TAN) lines at the nuclear envelope. Here we show that the nuclear envelope protein Samp1 (NET5) is involved in nuclear movement during fibroblast polarization and migration. Moreover, we demonstrate that Samp1 is a component of TAN lines that contain nesprin-2G and SUN2. Finally, Samp1 associates with SUN2 and lamin A/C, and the presence of Samp1 at the nuclear envelope requires lamin A/C. These results support a role for Samp1 in the association between the LINC complex and lamins during nuclear movement.


Asunto(s)
Núcleo Celular/fisiología , Proteínas de la Membrana/metabolismo , Membrana Nuclear/metabolismo , Proteínas Nucleares/metabolismo , Animales , Línea Celular , Movimiento Celular/fisiología , Núcleo Celular/metabolismo , Lamina Tipo A/genética , Lamina Tipo A/metabolismo , Proteínas de la Membrana/genética , Ratones , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/genética , Interferencia de ARN , ARN Interferente Pequeño , Proteínas de Schizosaccharomyces pombe/metabolismo , Proteínas de Unión a Telómeros/metabolismo
15.
Dev Cell ; 21(2): 273-87, 2011 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-21782525

RESUMEN

Coordinating the balance between progenitor self-renewal and myogenic differentiation is required for a regulated expansion of the developing muscles. Previous observation that neural crest cells (NCCs) migrate throughout the somite regions, where trunk skeletal muscles first emerge, suggests a potential role for these cells in influencing early muscle formation. However, specific signaling interactions between NCCs and skeletal muscle cells remain unknown. Here we show that mice with specific NCC and peripheral nervous system defects display impaired survival of skeletal muscle and show skeletal muscle progenitor cell (MPC) depletion due to precocious commitment to differentiation. We show that reduced NCC-derived Neuregulin1 (Nrg1) in the somite region perturbs ErbB3 signaling in uncommitted MPCs. Using a combination of explant culture experiments and genetic ablation in the mouse, we demonstrate that Nrg1 signals provided by the NCC lineage play a critical role in sustainable myogenesis, by restraining MPCs from precocious differentiation.


Asunto(s)
Diferenciación Celular/fisiología , Linaje de la Célula/fisiología , Desarrollo de Músculos/fisiología , Músculo Esquelético/citología , Cresta Neural/citología , Neurregulina-1/metabolismo , Receptor ErbB-3/metabolismo , Transducción de Señal/fisiología , Animales , Caspasa 3/metabolismo , Movimiento Celular/genética , Movimiento Celular/fisiología , Embrión de Mamíferos , Regulación del Desarrollo de la Expresión Génica/genética , Ratones , Ratones Transgénicos , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Músculo Esquelético/embriología , Proteína MioD/metabolismo , Neurregulina-1/genética , Proteínas de Neurofilamentos/metabolismo , Técnicas de Cultivo de Órganos , Factor de Transcripción PAX7/metabolismo , Receptor ErbB-3/genética , Factores de Transcripción SOXE/deficiencia , Factores de Transcripción SOXE/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteína Wnt1/genética
16.
Blood ; 116(13): 2345-55, 2010 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-20548097

RESUMEN

Polyploidization of megakaryocytes (MKs), the platelet precursors, occurs by endomitosis, a mitotic process that fails at late stages of cytokinesis. Expression and function of Aurora B kinase during endomitosis remain controversial. Here, we report that Aurora B is normally expressed during the human MK endomitotic process. Aurora B localized normally in the midzone or midbody during anaphase and telophase in low ploidy megakaryocytes and in up to 16N rare endomitotic MKs was observed. Aurora B was also functional during cytokinesis as attested by phosphorylation of both its activation site and MgcRacGAP, its main substrate. However, despite its activation, Aurora B did not prevent furrow regression. Inhibition of Aurora B by AZD1152-HQPA decreased cell cycle entry both in 2N to 4N and polyploid MKs and induced apoptosis mainly in 2N to 4N cells. In both MK classes, AZD1152-HQPA induced p53 activation and retinoblastoma hypophosphorylation. Resistance of polyploid MKs to apoptosis correlated to a high BclxL level. Aurora B inhibition did not impair MK polyploidization but profoundly modified the endomitotic process by inducing a mis-segregation of chromosomes and a mitotic failure in anaphase. This indicates that Aurora B is dispensable for MK polyploidization but is necessary to achieve a normal endomitotic process.


Asunto(s)
Megacariocitos/citología , Megacariocitos/enzimología , Mitosis/genética , Mitosis/fisiología , Poliploidía , Proteínas Serina-Treonina Quinasas/fisiología , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Aurora Quinasa B , Aurora Quinasas , Segregación Cromosómica/efectos de los fármacos , Segregación Cromosómica/fisiología , Fase G1/efectos de los fármacos , Fase G1/fisiología , Humanos , Técnicas In Vitro , Proteínas Inhibidoras de la Apoptosis , Megacariocitos/efectos de los fármacos , Proteínas Asociadas a Microtúbulos/metabolismo , Mitosis/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Fase S/efectos de los fármacos , Fase S/fisiología , Huso Acromático/enzimología , Survivin
17.
Blood ; 112(8): 3164-74, 2008 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-18684864

RESUMEN

Megakaryocyte (MK) is the naturally polyploid cell that gives rise to platelets. Polyploidization occurs by endomitosis, which was a process considered to be an incomplete mitosis aborted in anaphase. Here, we used time-lapse confocal video microscopy to visualize the endomitotic process of primary human megakaryocytes. Our results show that the switch from mitosis to endomitosis corresponds to a late failure of cytokinesis accompanied by a backward movement of the 2 daughter cells. No abnormality was observed in the central spindle of endomitotic MKs. A furrow formation was present, but the contractile ring was abnormal because accumulation of nonmuscle myosin IIA was lacking. In addition, a defect in cell elongation was observed in dipolar endomitotic MKs during telophase. RhoA and F-actin were partially concentrated at the site of furrowing. Inhibition of the Rho/Rock pathway caused the disappearance of F-actin at midzone and increased MK ploidy level. This inhibition was associated with a more pronounced defect in furrow formation as well as in spindle elongation. Our results suggest that the late failure of cytokinesis responsible for the endomitotic process is related to a partial defect in the Rho/Rock pathway activation.


Asunto(s)
Megacariocitos/citología , Mitosis , Quinasas Asociadas a rho/metabolismo , Actinas/metabolismo , Antígenos CD34/biosíntesis , Separación Celular , Citocinesis , Humanos , Lentivirus/metabolismo , Microscopía Confocal , Microscopía por Video , Ploidias , Transducción de Señal , Telofase
18.
J Leukoc Biol ; 82(5): 1278-88, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17684043

RESUMEN

Homophilic ligation of CD31, a member of the Ig superfamily of adhesion receptors, promotes macrophage clearance of apoptotic leukocytes by a mechanism hitherto not described. In studying CD31-dependent regulation of beta1-integrin binding of fibronectin-coated Latex beads, we discovered a role for the voltage-gated potassium channel ether-à-go-go-related gene (ERG) as a downstream effector of CD31 signaling. ERG was identified by tandem mass spectrometry as a 140-kDa protein, which was selectively modified with biotin following the targeted delivery of a biotin-transfer reagent to CD31 using Fab fragments of an anti-CD31 mAb. Similar results were obtained with macrophages but not K562 cells, expressing a truncated cytoplasmic tail of CD31, which failed to regulate bead binding. Colocalization of CD31 with ERG was confirmed by immunofluorescence for K562 cells and macrophages. We now demonstrate that the resting membrane potential of macrophages is depolarized on contact with apoptotic cells and that CD31 inhibits the ERG current, which would otherwise function to repolarize. Sustained depolarization favored the firm binding of phagocytic targets, a prerequisite for efficient engulfment. Our results identify ERG as a downstream effector of CD31 in the regulation of integrin-dependent binding of apoptotic cells by macrophages.


Asunto(s)
Apoptosis , Integrina beta1/metabolismo , Macrófagos/fisiología , Fagocitos/fisiología , Fagocitosis/fisiología , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/fisiología , Anticuerpos Monoclonales/farmacología , Reactivos de Enlaces Cruzados/farmacología , Electrofisiología , Canales de Potasio Éter-A-Go-Go/metabolismo , Fibronectinas/metabolismo , Técnica del Anticuerpo Fluorescente , Humanos , Células K562 , Activación de Macrófagos , Potenciales de la Membrana , Monocitos/citología , Monocitos/metabolismo
19.
Blood ; 110(1): 345-53, 2007 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-17374740

RESUMEN

Primary myelofibrosis (PMF) is the most serious myeloproliferative disorder, characterized by clonal myeloproliferation associated with cytokine-mediated bone marrow stromal reaction including fibrosis and osteosclerosis. Current drug therapy remains mainly palliative. Because the NF-kappaB pathway is implicated in the abnormal release of cytokines in PMF, the proteasome inhibitor bortezomib might be a potential therapy. To test its effect, we used the lethal murine model of myelofibrosis induced by thrombopoietin (TPO) overexpression. In this TPO(high) model, the development of the disease is related to a deregulated MPL signaling, as recently described in PMF patients. We first demonstrated that bortezomib was able to inhibit TPO-induced NF-kappaB activation in vitro in murine megakaryocytes. It also inhibited NF-kappaB activation in vivo in TPO(high) mice leading to decreased IL-1alpha plasma levels. After 4 weeks of treatment, bortezomib decreased TGF-beta1 levels in marrow fluids and impaired marrow and spleen fibrosis development. After 12 weeks of treatment, bortezomib also impaired osteosclerosis development through osteoprotegerin inhibition. Moreover, this drug reduced myeloproliferation induced by high TPO level. Finally, bortezomib dramatically improved TPO(high) mouse survival (89% vs 8% at week 52). We conclude that bortezomib appears as a promising therapy for future treatment of PMF patients.


Asunto(s)
Ácidos Borónicos/farmacología , Osteosclerosis/tratamiento farmacológico , Mielofibrosis Primaria/tratamiento farmacológico , Inhibidores de Proteasas/farmacología , Pirazinas/farmacología , Trombopoyetina/efectos adversos , Animales , Bortezomib , Modelos Animales de Enfermedad , Ratones , FN-kappa B/efectos de los fármacos , FN-kappa B/metabolismo , Osteosclerosis/inducido químicamente , Mielofibrosis Primaria/inducido químicamente , Tasa de Supervivencia , Trombopoyetina/sangre
20.
Blood ; 109(10): 4229-36, 2007 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-17244674

RESUMEN

Platelets are released by megakaryocytes (MKs) via cytoplasmic extensions called proplatelets, which require profound changes in the microtubule and actin organization. Here, we provide evidence that the Rho/ROCK pathway, a well-known regulator of actin cytoskeleton, acts as a negative regulator of proplatelet formation (PPF). Rho is expressed at a high level during the entire MK differentiation including human CD34(+) cells. Thrombopoietin stimulates its activity but at a higher extent in immature than in mature MKs. Overexpression of a dominant-negative or a spontaneously active RhoA leads to an increase or a decrease in PPF indicating that Rho activation inhibits PPF. This inhibitory effect is mediated through the main Rho effector, Rho kinase (ROCK), the inhibition of which also increases PPF. Furthermore, inhibition of Rho or ROCK in MKs leads to a decrease in myosin light chain 2 (MLC2) phosphorylation, which is required for myosin contractility. Interestingly, inhibition of the MLC kinase also decreases MLC2 phosphorylation while increasing PPF. Taken together, our results suggest that MLC2 phosphorylation is regulated by both ROCK and MLC kinase and plays an important role in platelet biogenesis by controlling PPF and fragmentation.


Asunto(s)
Plaquetas/citología , Péptidos y Proteínas de Señalización Intracelular/fisiología , Megacariocitos/citología , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas de Unión al GTP rho/fisiología , Miosinas Cardíacas/metabolismo , Diferenciación Celular/genética , Células Cultivadas , Activación Enzimática/efectos de los fármacos , Regulación de la Expresión Génica , Hematopoyesis/efectos de los fármacos , Humanos , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intracelular/genética , Cadenas Ligeras de Miosina/metabolismo , Quinasa de Cadena Ligera de Miosina/antagonistas & inhibidores , Quinasa de Cadena Ligera de Miosina/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Transducción de Señal , Trombopoyetina/farmacología , Proteínas de Unión al GTP rho/antagonistas & inhibidores , Proteínas de Unión al GTP rho/genética , Proteínas de Unión al GTP rho/metabolismo , Quinasas Asociadas a rho
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...