Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.785
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 183(1): 46-61.e21, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32941802

RESUMEN

Metazoan organisms rely on conserved stress response pathways to alleviate adverse conditions and preserve cellular integrity. Stress responses are particularly important in stem cells that provide lifetime support for tissue formation and repair, but how these protective systems are integrated into developmental programs is poorly understood. Here we used myoblast differentiation to identify the E3 ligase CUL2FEM1B and its substrate FNIP1 as core components of the reductive stress response. Reductive stress, as caused by prolonged antioxidant signaling or mitochondrial inactivity, reverts the oxidation of invariant Cys residues in FNIP1 and allows CUL2FEM1B to recognize its target. The ensuing proteasomal degradation of FNIP1 restores mitochondrial activity to preserve redox homeostasis and stem cell integrity. The reductive stress response is therefore built around a ubiquitin-dependent rheostat that tunes mitochondrial activity to redox needs and implicates metabolic control in coordination of stress and developmental signaling.


Asunto(s)
Proteínas Portadoras/metabolismo , Estrés Oxidativo/fisiología , Especies Reactivas de Oxígeno/metabolismo , Antioxidantes/metabolismo , Proteínas Portadoras/genética , Diferenciación Celular , Células HEK293 , Homeostasis , Humanos , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Mitocondrias , Desarrollo de Músculos/fisiología , Mioblastos/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Transducción de Señal , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación
2.
Cell ; 183(7): 1913-1929.e26, 2020 12 23.
Artículo en Inglés | MEDLINE | ID: mdl-33333020

RESUMEN

Neurons in the cerebral cortex connect through descending pathways to hindbrain and spinal cord to activate muscle and generate movement. Although components of this pathway have been previously generated and studied in vitro, the assembly of this multi-synaptic circuit has not yet been achieved with human cells. Here, we derive organoids resembling the cerebral cortex or the hindbrain/spinal cord and assemble them with human skeletal muscle spheroids to generate 3D cortico-motor assembloids. Using rabies tracing, calcium imaging, and patch-clamp recordings, we show that corticofugal neurons project and connect with spinal spheroids, while spinal-derived motor neurons connect with muscle. Glutamate uncaging or optogenetic stimulation of cortical spheroids triggers robust contraction of 3D muscle, and assembloids are morphologically and functionally intact for up to 10 weeks post-fusion. Together, this system highlights the remarkable self-assembly capacity of 3D cultures to form functional circuits that could be used to understand development and disease.


Asunto(s)
Corteza Cerebral/fisiología , Corteza Motora/fisiología , Organoides/fisiología , Animales , Calcio/metabolismo , Diferenciación Celular , Células Cultivadas , Vértebras Cervicales , Regulación de la Expresión Génica , Glutamatos/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/citología , Ratones , Músculos/fisiología , Mioblastos/metabolismo , Red Nerviosa/fisiología , Optogenética , Organoides/ultraestructura , Rombencéfalo/fisiología , Esferoides Celulares/citología , Médula Espinal/citología
3.
Cell ; 162(2): 300-313, 2015 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-26144318

RESUMEN

The transition from proliferating precursor cells to post-mitotic differentiated cells is crucial for development, tissue homeostasis, and tumor suppression. To study cell-cycle exit during differentiation in vivo, we developed a conditional knockout and lineage-tracing system for Caenorhabditis elegans. Combined lineage-specific gene inactivation and genetic screening revealed extensive redundancies between previously identified cell-cycle inhibitors and the SWI/SNF chromatin-remodeling complex. Muscle precursor cells missing either SWI/SNF or G1/S inhibitor function could still arrest cell division, while simultaneous inactivation of these regulators caused continued proliferation and a C. elegans tumor phenotype. Further genetic analyses support that SWI/SNF acts in concert with hlh-1 MyoD, antagonizes Polycomb-mediated transcriptional repression, and suppresses cye-1 Cyclin E transcription to arrest cell division of muscle precursors. Thus, SWI/SNF and G1/S inhibitors provide alternative mechanisms to arrest cell-cycle progression during terminal differentiation, which offers insight into the frequent mutation of SWI/SNF genes in human cancers.


Asunto(s)
Caenorhabditis elegans/citología , Caenorhabditis elegans/crecimiento & desarrollo , Músculos/citología , Animales , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Ciclo Celular , Diferenciación Celular , Linaje de la Célula , Proliferación Celular , Proteínas Cromosómicas no Histona/metabolismo , Proteínas Musculares , Músculos/metabolismo , Mioblastos/citología , Mioblastos/metabolismo , Factores Reguladores Miogénicos/metabolismo , Proteínas Nucleares , Proteínas del Grupo Polycomb/metabolismo , Factores de Transcripción/metabolismo
4.
Cell ; 158(3): 477-8, 2014 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-25083863

RESUMEN

Regulation of gene expression in mammalian mitochondria by microRNAs is reported by Zhang et al. During muscle cell differentiation, localization of a miRNA is increased within mitochondria, where it interacts with Ago2 to selectively activate translation of mtDNA-encoded mRNAs. The findings represent a new mitochondrial regulatory pathway and a potentially powerful means to purposefully manipulate mtDNA expression.


Asunto(s)
Diferenciación Celular , MicroARNs/metabolismo , Mitocondrias/metabolismo , Mioblastos/metabolismo , Miocitos Cardíacos/metabolismo , Biosíntesis de Proteínas , Animales
5.
Cell ; 158(3): 607-19, 2014 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-25083871

RESUMEN

MicroRNAs are well known to mediate translational repression and mRNA degradation in the cytoplasm. Various microRNAs have also been detected in membrane-compartmentalized organelles, but the functional significance has remained elusive. Here, we report that miR-1, a microRNA specifically induced during myogenesis, efficiently enters the mitochondria where it unexpectedly stimulates, rather than represses, the translation of specific mitochondrial genome-encoded transcripts. We show that this positive effect requires specific miR:mRNA base-pairing and Ago2, but not its functional partner GW182, which is excluded from the mitochondria. We provide evidence for the direct action of Ago2 in mitochondrial translation by crosslinking immunoprecipitation coupled with deep sequencing (CLIP-seq), functional rescue with mitochondria-targeted Ago2, and selective inhibition of the microRNA machinery in the cytoplasm. These findings unveil a positive function of microRNA in mitochondrial translation and suggest a highly coordinated myogenic program via miR-1-mediated translational stimulation in the mitochondria and repression in the cytoplasm.


Asunto(s)
Diferenciación Celular , MicroARNs/metabolismo , Mitocondrias/metabolismo , Mioblastos/metabolismo , Miocitos Cardíacos/metabolismo , Biosíntesis de Proteínas , Animales , Proteínas Argonautas/metabolismo , Línea Celular , Ratones , Mioblastos/citología , Miocitos Cardíacos/citología
6.
Cell ; 157(7): 1657-70, 2014 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-24949975

RESUMEN

Voltage-gated Na and Ca2+ channels comprise distinct ion channel superfamilies, yet the carboxy tails of these channels exhibit high homology, hinting at a long-shared and purposeful module. For different Ca2+ channels, carboxyl-tail interactions with calmodulin do elaborate robust and similar forms of Ca2+ regulation. However, Na channels have only shown subtler Ca2+ modulation that differs among reports, challenging attempts at unified understanding. Here, by rapid Ca2+ photorelease onto Na channels, we reset this view of Na channel regulation. For cardiac-muscle channels (NaV1.5), reported effects from which most mechanistic proposals derive, we observe no Ca2+ modulation. Conversely, for skeletal-muscle channels (NaV1.4), we uncover fast Ca2+ regulation eerily similar to that of Ca2+ channels. Channelopathic myotonia mutations halve NaV1.4 Ca2+ regulation, and transplanting the NaV1.4 carboxy tail onto Ca2+ channels recapitulates Ca2+ regulation. Thus, we argue for the persistence and physiological relevance of an ancient Ca2+ regulatory module across Na and Ca2+ channels.


Asunto(s)
Canales de Calcio/metabolismo , Calcio/metabolismo , Calmodulina/química , Canales de Sodio Activados por Voltaje/química , Secuencia de Aminoácidos , Animales , Canales de Calcio/genética , Calmodulina/metabolismo , Cobayas , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Células Musculares/metabolismo , Mioblastos/metabolismo , Filogenia , Ratas , Alineación de Secuencia , Canales de Sodio Activados por Voltaje/genética , Canales de Sodio Activados por Voltaje/metabolismo
7.
Genes Dev ; 35(17-18): 1209-1228, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34413137

RESUMEN

The generation of myotubes from fibroblasts upon forced MyoD expression is a classic example of transcription factor-induced reprogramming. We recently discovered that additional modulation of signaling pathways with small molecules facilitates reprogramming to more primitive induced myogenic progenitor cells (iMPCs). Here, we dissected the transcriptional and epigenetic dynamics of mouse fibroblasts undergoing reprogramming to either myotubes or iMPCs using a MyoD-inducible transgenic model. Induction of MyoD in fibroblasts combined with small molecules generated Pax7+ iMPCs with high similarity to primary muscle stem cells. Analysis of intermediate stages of iMPC induction revealed that extinction of the fibroblast program preceded induction of the stem cell program. Moreover, key stem cell genes gained chromatin accessibility prior to their transcriptional activation, and these regions exhibited a marked loss of DNA methylation dependent on the Tet enzymes. In contrast, myotube generation was associated with few methylation changes, incomplete and unstable reprogramming, and an insensitivity to Tet depletion. Finally, we showed that MyoD's ability to bind to unique bHLH targets was crucial for generating iMPCs but dispensable for generating myotubes. Collectively, our analyses elucidate the role of MyoD in myogenic reprogramming and derive general principles by which transcription factors and signaling pathways cooperate to rewire cell identity.


Asunto(s)
Desarrollo de Músculos , Proteína MioD , Animales , Diferenciación Celular/genética , Ratones , Desarrollo de Músculos/genética , Fibras Musculares Esqueléticas , Músculo Esquelético , Proteína MioD/genética , Proteína MioD/metabolismo , Mioblastos/metabolismo , Células Madre/metabolismo
8.
Cell ; 152(3): 584-98, 2013 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-23374351

RESUMEN

Eukaryotic cells have a layer of heterochromatin at the nuclear periphery. To investigate mechanisms regulating chromatin distribution, we analyzed heterochromatin organization in different tissues and species, including mice with mutations in the lamin B receptor (Lbr) and lamin A (Lmna) genes that encode nuclear envelope (NE) proteins. We identified LBR- and lamin-A/C-dependent mechanisms tethering heterochromatin to the NE. The two tethers are sequentially used during cellular differentiation and development: first the LBR- and then the lamin-A/C-dependent tether. The absence of both LBR and lamin A/C leads to loss of peripheral heterochromatin and an inverted architecture with heterochromatin localizing to the nuclear interior. Myoblast transcriptome analyses indicated that selective disruption of the LBR- or lamin-A-dependent heterochromatin tethers have opposite effects on muscle gene expression, either increasing or decreasing, respectively. These results show how changes in NE composition contribute to regulating heterochromatin positioning, gene expression, and cellular differentiation during development.


Asunto(s)
Heterocromatina/metabolismo , Lamina Tipo A/metabolismo , Desarrollo de Músculos , Mioblastos/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Animales , Perfilación de la Expresión Génica , Ratones , Mioblastos/citología , Membrana Nuclear/metabolismo , Receptor de Lamina B
9.
Genome Res ; 34(5): 665-679, 2024 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-38777608

RESUMEN

Facioscapulohumeral muscular dystrophy (FSHD) is linked to abnormal derepression of the transcription activator DUX4. This effect is localized to a low percentage of cells, requiring single-cell analysis. However, single-cell/nucleus RNA-seq cannot fully capture the transcriptome of multinucleated large myotubes. To circumvent these issues, we use multiplexed error-robust fluorescent in situ hybridization (MERFISH) spatial transcriptomics that allows profiling of RNA transcripts at a subcellular resolution. We simultaneously examined spatial distributions of 140 genes, including 24 direct DUX4 targets, in in vitro differentiated myotubes and unfused mononuclear cells (MNCs) of control, isogenic D4Z4 contraction mutant and FSHD patient samples, as well as the individual nuclei within them. We find myocyte nuclei segregate into two clusters defined by the expression of DUX4 target genes, which is exclusively found in patient/mutant nuclei, whereas MNCs cluster based on developmental states. Patient/mutant myotubes are found in "FSHD-hi" and "FSHD-lo" states with the former signified by high DUX4 target expression and decreased muscle gene expression. Pseudotime analyses reveal a clear bifurcation of myoblast differentiation into control and FSHD-hi myotube branches, with variable numbers of DUX4 target-expressing nuclei found in multinucleated FSHD-hi myotubes. Gene coexpression modules related to extracellular matrix and stress gene ontologies are significantly altered in patient/mutant myotubes compared with the control. We also identify distinct subpathways within the DUX4 gene network that may differentially contribute to the disease transcriptomic phenotype. Taken together, our MERFISH-based study provides effective gene network profiling of multinucleated cells and identifies FSHD-induced transcriptomic alterations during myoblast differentiation.


Asunto(s)
Fibras Musculares Esqueléticas , Distrofia Muscular Facioescapulohumeral , Mioblastos , Análisis de la Célula Individual , Transcriptoma , Distrofia Muscular Facioescapulohumeral/genética , Distrofia Muscular Facioescapulohumeral/patología , Distrofia Muscular Facioescapulohumeral/metabolismo , Humanos , Mioblastos/metabolismo , Análisis de la Célula Individual/métodos , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Diferenciación Celular/genética , Hibridación Fluorescente in Situ , Perfilación de la Expresión Génica/métodos
10.
Cell ; 150(4): 710-24, 2012 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-22901804

RESUMEN

The muscleblind-like (Mbnl) family of RNA-binding proteins plays important roles in muscle and eye development and in myotonic dystrophy (DM), in which expanded CUG or CCUG repeats functionally deplete Mbnl proteins. We identified transcriptome-wide functional and biophysical targets of Mbnl proteins in brain, heart, muscle, and myoblasts by using RNA-seq and CLIP-seq approaches. This analysis identified several hundred splicing events whose regulation depended on Mbnl function in a pattern indicating functional interchangeability between Mbnl1 and Mbnl2. A nucleotide resolution RNA map associated repression or activation of exon splicing with Mbnl binding near either 3' splice site or near the downstream 5' splice site, respectively. Transcriptomic analysis of subcellular compartments uncovered a global role for Mbnls in regulating localization of mRNAs in both mouse and Drosophila cells, and Mbnl-dependent translation and protein secretion were observed for a subset of mRNAs with Mbnl-dependent localization. These findings hold several new implications for DM pathogenesis.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Distrofia Miotónica/metabolismo , Empalme del ARN , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo , Transcriptoma , Regiones no Traducidas 3' , Animales , Proteínas de Unión al ADN/genética , Proteínas de Drosophila , Drosophila melanogaster/metabolismo , Exones , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Mioblastos/metabolismo , Distrofia Miotónica/genética , Proteínas Nucleares , Especificidad de Órganos , Sitios de Empalme de ARN , Proteínas de Unión al ARN/genética
11.
Nature ; 591(7849): 281-287, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33568815

RESUMEN

Skeletal muscle regenerates through the activation of resident stem cells. Termed satellite cells, these normally quiescent cells are induced to proliferate by wound-derived signals1. Identifying the source and nature of these cues has been hampered by an inability to visualize the complex cell interactions that occur within the wound. Here we use muscle injury models in zebrafish to systematically capture the interactions between satellite cells and the innate immune system after injury, in real time, throughout the repair process. This analysis revealed that a specific subset of macrophages 'dwell' within the injury, establishing a transient but obligate niche for stem cell proliferation. Single-cell profiling identified proliferative signals that are secreted by dwelling macrophages, which include the cytokine nicotinamide phosphoribosyltransferase (Nampt, which is also known as visfatin or PBEF in humans). Nampt secretion from the macrophage niche is required for muscle regeneration, acting through the C-C motif chemokine receptor type 5 (Ccr5), which is expressed on muscle stem cells. This analysis shows that in addition to their ability to modulate the immune response, specific macrophage populations also provide a transient stem-cell-activating niche, directly supplying proliferation-inducing cues that govern the repair process that is mediated by muscle stem cells. This study demonstrates that macrophage-derived niche signals for muscle stem cells, such as NAMPT, can be applied as new therapeutic modalities for skeletal muscle injury and disease.


Asunto(s)
Macrófagos/metabolismo , Músculo Esquelético/citología , Músculo Esquelético/lesiones , Mioblastos/citología , Nicotinamida Fosforribosiltransferasa/metabolismo , Nicho de Células Madre , Pez Cebra/metabolismo , Animales , Proliferación Celular , Modelos Animales de Enfermedad , Humanos , Macrófagos/citología , Masculino , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Mioblastos/metabolismo , Nicotinamida Fosforribosiltransferasa/genética , Factor de Transcripción PAX7/metabolismo , RNA-Seq , Receptores CCR5/genética , Receptores CCR5/metabolismo , Regeneración/fisiología , Análisis de la Célula Individual , Pez Cebra/inmunología
12.
Mol Cell ; 74(3): 609-621.e6, 2019 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-30922843

RESUMEN

Adult tissue repair and regeneration require stem-progenitor cells that can self-renew and generate differentiated progeny. Skeletal muscle regenerative capacity relies on muscle satellite cells (MuSCs) and their interplay with different cell types within the niche. However, our understanding of skeletal muscle tissue cellular composition is limited. Here, using a combined approach of single-cell RNA sequencing and mass cytometry, we precisely mapped 10 different mononuclear cell types in adult mouse muscle. We also characterized gene signatures and determined key discriminating markers of each cell type. We identified two previously understudied cell populations in the interstitial compartment. One expresses the transcription factor scleraxis and generated tenocytes in vitro. The second expresses markers of smooth muscle and mesenchymal cells (SMMCs) and, while distinct from MuSCs, exhibited myogenic potential and promoted MuSC engraftment following transplantation. The blueprint presented here yields crucial insights into muscle-resident cell-type identities and can be exploited to study muscle diseases.


Asunto(s)
Diferenciación Celular/genética , Linaje de la Célula/genética , Fibras Musculares Esqueléticas/citología , Células Satélite del Músculo Esquelético/citología , Animales , Ratones , Desarrollo de Músculos/genética , Fibras Musculares Esqueléticas/metabolismo , Mioblastos/citología , Mioblastos/metabolismo , Células Satélite del Músculo Esquelético/metabolismo , Análisis de la Célula Individual , Células Madre/citología , Células Madre/metabolismo
13.
Proc Natl Acad Sci U S A ; 121(23): e2217971121, 2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38805272

RESUMEN

Myogenesis is a multistep process that requires a spatiotemporal regulation of cell events resulting finally in myoblast fusion into multinucleated myotubes. Most major insights into the mechanisms underlying fusion seem to be conserved from insects to mammals and include the formation of podosome-like protrusions (PLPs) that exert a driving force toward the founder cell. However, the machinery that governs this process remains poorly understood. In this study, we demonstrate that MTM1 is the main enzyme responsible for the production of phosphatidylinositol 5-phosphate, which in turn fuels PI5P 4-kinase α to produce a minor and functional pool of phosphatidylinositol 4,5-bisphosphate that concentrates in PLPs containing the scaffolding protein Tks5, Dynamin-2, and the fusogenic protein Myomaker. Collectively, our data reveal a functional crosstalk between a PI-phosphatase and a PI-kinase in the regulation of PLP formation.


Asunto(s)
Fusión Celular , Mioblastos , Fosfatos de Fosfatidilinositol , Podosomas , Animales , Fosfatos de Fosfatidilinositol/metabolismo , Ratones , Mioblastos/metabolismo , Mioblastos/citología , Podosomas/metabolismo , Proteínas Tirosina Fosfatasas no Receptoras/metabolismo , Proteínas Tirosina Fosfatasas no Receptoras/genética , Desarrollo de Músculos/fisiología
14.
Proc Natl Acad Sci U S A ; 121(17): e2312330121, 2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38625936

RESUMEN

The apolipoprotein B messenger RNA editing enzyme, catalytic polypeptide (APOBEC) family is composed of nucleic acid editors with roles ranging from antibody diversification to RNA editing. APOBEC2, a member of this family with an evolutionarily conserved nucleic acid-binding cytidine deaminase domain, has neither an established substrate nor function. Using a cellular model of muscle differentiation where APOBEC2 is inducibly expressed, we confirmed that APOBEC2 does not have the attributed molecular functions of the APOBEC family, such as RNA editing, DNA demethylation, and DNA mutation. Instead, we found that during muscle differentiation APOBEC2 occupied a specific motif within promoter regions; its removal from those regions resulted in transcriptional changes. Mechanistically, these changes reflect the direct interaction of APOBEC2 with histone deacetylase (HDAC) transcriptional corepressor complexes. We also found that APOBEC2 could bind DNA directly, in a sequence-specific fashion, suggesting that it functions as a recruiter of HDAC to specific genes whose promoters it occupies. These genes are normally suppressed during muscle cell differentiation, and their suppression may contribute to the safeguarding of muscle cell fate. Altogether, our results reveal a unique role for APOBEC2 within the APOBEC family.


Asunto(s)
Cromatina , Proteínas Musculares , Desaminasas APOBEC/genética , Desaminasas APOBEC-1/genética , Diferenciación Celular/genética , Cromatina/genética , Citidina Desaminasa/metabolismo , ADN , Fibras Musculares Esqueléticas/metabolismo , Proteínas Musculares/metabolismo , Mioblastos/metabolismo , ARN Mensajero/genética , Animales , Ratones
15.
Proc Natl Acad Sci U S A ; 121(19): e2321438121, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38687782

RESUMEN

Successful CRISPR/Cas9-based gene editing in skeletal muscle is dependent on efficient propagation of Cas9 to all myonuclei in the myofiber. However, nuclear-targeted gene therapy cargos are strongly restricted to their myonuclear domain of origin. By screening nuclear localization signals and nuclear export signals, we identify "Myospreader," a combination of short peptide sequences that promotes myonuclear propagation. Appending Myospreader to Cas9 enhances protein stability and myonuclear propagation in myoblasts and myofibers. AAV-delivered Myospreader dCas9 better inhibits transcription of toxic RNA in a myotonic dystrophy mouse model. Furthermore, Myospreader Cas9 achieves higher rates of gene editing in CRISPR reporter and Duchenne muscular dystrophy mouse models. Myospreader reveals design principles relevant to all nuclear-targeted gene therapies and highlights the importance of the spatial dimension in therapeutic development.


Asunto(s)
Sistemas CRISPR-Cas , Núcleo Celular , Edición Génica , Terapia Genética , Músculo Esquelético , Distrofia Muscular de Duchenne , Edición Génica/métodos , Animales , Ratones , Músculo Esquelético/metabolismo , Núcleo Celular/metabolismo , Terapia Genética/métodos , Distrofia Muscular de Duchenne/terapia , Distrofia Muscular de Duchenne/genética , Humanos , Señales de Localización Nuclear/genética , Proteína 9 Asociada a CRISPR/metabolismo , Proteína 9 Asociada a CRISPR/genética , Modelos Animales de Enfermedad , Mioblastos/metabolismo
16.
Genes Dev ; 33(11-12): 626-640, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-30975722

RESUMEN

Rhabdomyosarcoma (RMS) is an aggressive pediatric cancer composed of myoblast-like cells. Recently, we discovered a unique muscle progenitor marked by the expression of the Twist2 transcription factor. Genomic analyses of 258 RMS patient tumors uncovered prevalent copy number amplification events and increased expression of TWIST2 in fusion-negative RMS. Knockdown of TWIST2 in RMS cells results in up-regulation of MYOGENIN and a decrease in proliferation, implicating TWIST2 as an oncogene in RMS. Through an inducible Twist2 expression system, we identified Twist2 as a reversible inhibitor of myogenic differentiation with the remarkable ability to promote myotube dedifferentiation in vitro. Integrated analysis of genome-wide ChIP-seq and RNA-seq data revealed the first dynamic chromatin and transcriptional landscape of Twist2 binding during myogenic differentiation. During differentiation, Twist2 competes with MyoD at shared DNA motifs to direct global gene transcription and repression of the myogenic program. Additionally, Twist2 shapes the epigenetic landscape to drive chromatin opening at oncogenic loci and chromatin closing at myogenic loci. These epigenetic changes redirect MyoD binding from myogenic genes toward oncogenic, metabolic, and growth genes. Our study reveals the dynamic interplay between two opposing transcriptional regulators that control the fate of RMS and provides insight into the molecular etiology of this aggressive form of cancer.


Asunto(s)
Carcinogénesis , Desarrollo de Músculos , Proteína MioD/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Rabdomiosarcoma/genética , Rabdomiosarcoma/metabolismo , Proteína 1 Relacionada con Twist/genética , Proteína 1 Relacionada con Twist/metabolismo , Células Cultivadas , Ensamble y Desensamble de Cromatina , ADN/metabolismo , Transición Epitelial-Mesenquimal , Amplificación de Genes , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Secuencias Hélice-Asa-Hélice , Humanos , Proteína MioD/química , Mioblastos/metabolismo , Proteínas Nucleares/genética , Proteínas Represoras/química , Proteína 1 Relacionada con Twist/química
17.
J Cell Sci ; 137(6)2024 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-38345101

RESUMEN

Understanding how biophysical and biochemical microenvironmental cues together influence the regenerative activities of muscle stem cells and their progeny is crucial in strategizing remedies for pathological dysregulation of these cues in aging and disease. In this study, we investigated the cell-level influences of extracellular matrix (ECM) ligands and culture substrate stiffness on primary human myoblast contractility and proliferation within 16 h of plating and found that tethered fibronectin led to stronger stiffness-dependent responses compared to laminin and collagen. A proteome-wide analysis further uncovered cell metabolism, cytoskeletal and nuclear component regulation distinctions between cells cultured on soft and stiff substrates. Interestingly, we found that softer substrates increased the incidence of myoblasts with a wrinkled nucleus, and that the extent of wrinkling could predict Ki67 (also known as MKI67) expression. Nuclear wrinkling and Ki67 expression could be controlled by pharmacological manipulation of cellular contractility, offering a potential cellular mechanism. These results provide new insights into the regulation of human myoblast stiffness-dependent contractility response by ECM ligands and highlight a link between myoblast contractility and proliferation.


Asunto(s)
Matriz Extracelular , Membrana Nuclear , Humanos , Antígeno Ki-67/metabolismo , Matriz Extracelular/metabolismo , Mioblastos/metabolismo , Proliferación Celular
18.
Development ; 150(11)2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-37272529

RESUMEN

The mechanism of pattern formation during limb muscle development remains poorly understood. The canonical view holds that naïve limb muscle progenitor cells (MPCs) invade a pre-established pattern of muscle connective tissue, thereby forming individual muscles. Here, we show that early murine embryonic limb MPCs highly accumulate pSMAD1/5/9, demonstrating active signaling of bone morphogenetic proteins (BMP) in these cells. Overexpression of inhibitory human SMAD6 (huSMAD6) in limb MPCs abrogated BMP signaling, impaired their migration and proliferation, and accelerated myogenic lineage progression. Fewer primary myofibers developed, causing an aberrant proximodistal muscle pattern. Patterning was not disturbed when huSMAD6 was overexpressed in differentiated muscle, implying that the proximodistal muscle pattern depends on BMP-mediated expansion of MPCs before their differentiation. We show that limb MPCs differentially express Hox genes, and Hox-expressing MPCs displayed active BMP signaling. huSMAD6 overexpression caused loss of HOXA11 in early limb MPCs. In conclusion, our data show that BMP signaling controls expansion of embryonic limb MPCs as a prerequisite for establishing the proximodistal muscle pattern, a process that involves expression of Hox genes.


Asunto(s)
Proteínas Morfogenéticas Óseas , Músculo Esquelético , Animales , Humanos , Ratones , Proteínas Morfogenéticas Óseas/metabolismo , Diferenciación Celular/fisiología , Genes Homeobox , Músculo Esquelético/metabolismo , Mioblastos/metabolismo , Proteína smad6/metabolismo
19.
EMBO Rep ; 25(8): 3627-3650, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38982191

RESUMEN

Skeletal muscle regeneration involves a signaling network that regulates the proliferation, differentiation, and fusion of muscle precursor cells to injured myofibers. IRE1α, one of the arms of the unfolded protein response, regulates cellular proteostasis in response to ER stress. Here, we demonstrate that inducible deletion of IRE1α in satellite cells of mice impairs skeletal muscle regeneration through inhibiting myoblast fusion. Knockdown of IRE1α or its downstream target, X-box protein 1 (XBP1), also inhibits myoblast fusion during myogenesis. Transcriptome analysis revealed that knockdown of IRE1α or XBP1 dysregulates the gene expression of molecules involved in myoblast fusion. The IRE1α-XBP1 axis mediates the gene expression of multiple profusion molecules, including myomaker (Mymk). Spliced XBP1 (sXBP1) transcription factor binds to the promoter of Mymk gene during myogenesis. Overexpression of myomaker in IRE1α-knockdown cultures rescues fusion defects. Inducible deletion of IRE1α in satellite cells also inhibits myoblast fusion and myofiber hypertrophy in response to functional overload. Collectively, our study demonstrates that IRE1α promotes myoblast fusion through sXBP1-mediated up-regulation of the gene expression of multiple profusion molecules, including myomaker.


Asunto(s)
Fusión Celular , Endorribonucleasas , Desarrollo de Músculos , Músculo Esquelético , Mioblastos , Proteínas Serina-Treonina Quinasas , Transducción de Señal , Proteína 1 de Unión a la X-Box , Animales , Proteína 1 de Unión a la X-Box/metabolismo , Proteína 1 de Unión a la X-Box/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Ratones , Mioblastos/metabolismo , Mioblastos/citología , Músculo Esquelético/metabolismo , Músculo Esquelético/citología , Desarrollo de Músculos/genética , Endorribonucleasas/metabolismo , Endorribonucleasas/genética , Células Satélite del Músculo Esquelético/metabolismo , Regeneración/genética , Diferenciación Celular/genética , Regulación de la Expresión Génica , Proteínas de la Membrana , Proteínas Musculares
20.
Cell ; 147(2): 358-69, 2011 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-22000014

RESUMEN

Recently, a new regulatory circuitry has been identified in which RNAs can crosstalk with each other by competing for shared microRNAs. Such competing endogenous RNAs (ceRNAs) regulate the distribution of miRNA molecules on their targets and thereby impose an additional level of post-transcriptional regulation. Here we identify a muscle-specific long noncoding RNA, linc-MD1, which governs the time of muscle differentiation by acting as a ceRNA in mouse and human myoblasts. Downregulation or overexpression of linc-MD1 correlate with retardation or anticipation of the muscle differentiation program, respectively. We show that linc-MD1 "sponges" miR-133 and miR-133 [corrected] to regulate the expression of MAML1 and MEF2C, transcription factors that activate muscle-specific gene expression. Finally, we demonstrate that linc-MD1 exerts the same control over differentiation timing in human myoblasts, and that its levels are strongly reduced in Duchenne muscle cells. We conclude that the ceRNA network plays an important role in muscle differentiation.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Desarrollo de Músculos , Músculo Esquelético/citología , ARN no Traducido/metabolismo , Animales , Secuencia de Bases , Proteínas de Unión al ADN/genética , Humanos , Proteínas de Dominio MADS/genética , Factores de Transcripción MEF2 , Ratones , MicroARNs/metabolismo , Datos de Secuencia Molecular , Músculo Esquelético/embriología , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/embriología , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/patología , Mioblastos/metabolismo , Factores Reguladores Miogénicos/genética , Proteínas Nucleares/genética , Procesamiento Postranscripcional del ARN , ARN Largo no Codificante , Factores de Transcripción/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA