Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Development ; 147(12)2020 06 26.
Artículo en Inglés | MEDLINE | ID: mdl-32541004

RESUMEN

Satellite cells (SC) are muscle stem cells that can regenerate adult muscles upon injury. Most SC originate from PAX7+ myogenic precursors set aside during development. Although myogenesis has been studied in mouse and chicken embryos, little is known about human muscle development. Here, we report the generation of human induced pluripotent stem cell (iPSC) reporter lines in which fluorescent proteins have been introduced into the PAX7 and MYOG loci. We use single cell RNA sequencing to analyze the developmental trajectory of the iPSC-derived PAX7+ myogenic precursors. We show that the PAX7+ cells generated in culture can produce myofibers and self-renew in vitro and in vivo Together, we demonstrate that cells exhibiting characteristics of human fetal satellite cells can be produced in vitro from iPSC, opening interesting avenues for muscular dystrophy cell therapy. This work provides significant insights into the development of the human myogenic lineage.


Asunto(s)
Diferenciación Celular , Factor de Transcripción PAX7/metabolismo , Células Satélite del Músculo Esquelético/metabolismo , Sistemas CRISPR-Cas/genética , Linaje de la Célula , Autorrenovación de las Células , Células Cultivadas , Genes Reporteros , Sitios Genéticos , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Fibras Musculares Esqueléticas/citología , Fibras Musculares Esqueléticas/metabolismo , Miogenina/genética , Factor de Transcripción PAX7/genética , ARN Guía de Kinetoplastida/metabolismo , Células Satélite del Músculo Esquelético/citología
2.
FASEB J ; 35(11): e21955, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34613626

RESUMEN

Kabuki syndrome (KS) is a rare genetic disorder caused primarily by mutations in the histone modifier genes KMT2D and KDM6A. The genes have broad temporal and spatial expression in many organs, resulting in complex phenotypes observed in KS patients. Hypotonia is one of the clinical presentations associated with KS, yet detailed examination of skeletal muscle samples from KS patients has not been reported. We studied the consequences of loss of KMT2D function in both mouse and human muscles. In mice, heterozygous loss of Kmt2d resulted in reduced neuromuscular junction (NMJ) perimeter, decreased muscle cell differentiation in vitro and impaired myofiber regeneration in vivo. Muscle samples from KS patients of different ages showed presence of increased fibrotic tissue interspersed between myofiber fascicles, which was not seen in mouse muscles. Importantly, when Kmt2d-deficient muscle stem cells were transplanted in vivo in a physiologic non-Kabuki environment, their differentiation potential is restored to levels undistinguishable from control cells. Thus, the epigenetic changes due to loss of function of KMT2D appear reversible through a change in milieu, opening a potential therapeutic avenue.


Asunto(s)
Anomalías Múltiples/metabolismo , Diferenciación Celular/genética , Proteínas de Unión al ADN/metabolismo , Cara/anomalías , Enfermedades Hematológicas/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Células Musculares/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Proteínas de Neoplasias/metabolismo , Transducción de Señal/genética , Enfermedades Vestibulares/metabolismo , Anomalías Múltiples/genética , Adolescente , Animales , Niño , Preescolar , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Femenino , Enfermedades Hematológicas/genética , N-Metiltransferasa de Histona-Lisina/genética , Humanos , Lactante , Masculino , Ratones , Ratones Transgénicos , Células Musculares/patología , Mutación , Proteína de la Leucemia Mieloide-Linfoide/genética , Proteínas de Neoplasias/genética , Unión Neuromuscular/genética , Unión Neuromuscular/metabolismo , Enfermedades Vestibulares/genética
3.
Nature ; 495(7441): 379-83, 2013 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-23485971

RESUMEN

Maintenance of body temperature is essential for the survival of homeotherms. Brown adipose tissue (BAT) is a specialized fat tissue that is dedicated to thermoregulation. Owing to its remarkable capacity to dissipate stored energy and its demonstrated presence in adult humans, BAT holds great promise for the treatment of obesity and metabolic syndrome. Rodent data suggest the existence of two types of brown fat cells: constitutive BAT (cBAT), which is of embryonic origin and anatomically located in the interscapular region of mice; and recruitable BAT (rBAT), which resides within white adipose tissue (WAT) and skeletal muscle, and has alternatively been called beige, brite or inducible BAT. Bone morphogenetic proteins (BMPs) regulate the formation and thermogenic activity of BAT. Here we use mouse models to provide evidence for a systemically active regulatory mechanism that controls whole-body BAT activity for thermoregulation and energy homeostasis. Genetic ablation of the type 1A BMP receptor (Bmpr1a) in brown adipogenic progenitor cells leads to a severe paucity of cBAT. This in turn increases sympathetic input to WAT, thereby promoting the formation of rBAT within white fat depots. This previously unknown compensatory mechanism, aimed at restoring total brown-fat-mediated thermogenic capacity in the body, is sufficient to maintain normal temperature homeostasis and resistance to diet-induced obesity. These data suggest an important physiological cross-talk between constitutive and recruitable brown fat cells. This sophisticated regulatory mechanism of body temperature may participate in the control of energy balance and metabolic disease.


Asunto(s)
Tejido Adiposo Pardo/citología , Tejido Adiposo Blanco/citología , Proteínas Morfogenéticas Óseas/metabolismo , Transducción de Señal , Tejido Adiposo Pardo/inervación , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Animales , Temperatura Corporal , Regulación de la Temperatura Corporal , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/genética , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/metabolismo , Proteínas Morfogenéticas Óseas/genética , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Metabolismo Energético , Ratones , Células Madre/citología
4.
Hum Mol Genet ; 23(21): 5781-92, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-24925318

RESUMEN

Dystroglycan is a transmembrane glycoprotein whose interactions with the extracellular matrix (ECM) are necessary for normal muscle and brain development, and disruptions of its function lead to dystroglycanopathies, a group of congenital muscular dystrophies showing extreme genetic and clinical heterogeneity. Specific glycans bound to the extracellular portion of dystroglycan, α-dystroglycan, mediate ECM interactions and most known dystroglycanopathy genes encode glycosyltransferases involved in glycan synthesis. POMK, which was found mutated in two dystroglycanopathy cases, is instead involved in a glycan phosphorylation reaction critical for ECM binding, but little is known about the clinical presentation of POMK mutations or of the function of this protein in the muscle. Here, we describe two families carrying different truncating alleles, both removing the kinase domain in POMK, with different clinical manifestations ranging from Walker-Warburg syndrome, the most severe form of dystroglycanopathy, to limb-girdle muscular dystrophy with cognitive defects. We explored POMK expression in fetal and adult human muscle and identified widespread expression primarily during fetal development in myocytes and interstitial cells suggesting a role for this protein during early muscle differentiation. Analysis of loss of function in the zebrafish embryo and larva showed that pomk function is necessary for normal muscle development, leading to locomotor dysfuction in the embryo and signs of muscular dystrophy in the larva. In summary, we defined diverse clinical presentations following POMK mutations and showed that this gene is necessary for early muscle development.


Asunto(s)
Estudios de Asociación Genética , Desarrollo de Músculos/genética , Mutación , Enfermedades Neuromusculares/diagnóstico , Enfermedades Neuromusculares/genética , Fenotipo , Proteínas Quinasas/genética , Adolescente , Adulto , Secuencia de Aminoácidos , Animales , Encéfalo/metabolismo , Encéfalo/patología , Niño , Preescolar , Consanguinidad , Análisis Mutacional de ADN , Distroglicanos/metabolismo , Exoma , Femenino , Expresión Génica , Técnicas de Silenciamiento del Gen , Silenciador del Gen , Estudio de Asociación del Genoma Completo , Glicosilación , Humanos , Imagen por Resonancia Magnética , Masculino , Datos de Secuencia Molecular , Linaje , Proteínas Quinasas/química , Alineación de Secuencia , Adulto Joven , Pez Cebra
5.
6.
FASEB J ; 28(7): 2955-69, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24687993

RESUMEN

Previously, we identified family with sequence similarity 65, member B (Fam65b), as a protein transiently up-regulated during differentiation and fusion of human myogenic cells. Silencing of Fam65b expression results in severe reduction of myogenin expression and consequent lack of myoblast fusion. The molecular function of Fam65b and whether misregulation of its expression could be causative of muscle diseases are unknown. Protein pulldowns were used to identify Fam65b-interacting proteins in differentiating human muscle cells and regenerating muscle tissue. In vitro, human muscle cells were treated with histone-deacetylase (HDAC) inhibitors, and expression of Fam65b and interacting proteins was studied. Nontreated cells were used as controls. In vivo, expression of Fam65b was down-regulated in developing zebrafish to determine the effects on muscle development. Fam65b binds to HDAC6 and dysferlin, the protein mutated in limb girdle muscular dystrophy 2B. The tricomplex Fam65b-HDAC6-dysferlin is transient, and Fam65b expression is necessary for the complex to form. Treatment of myogenic cells with pan-HDAC or HDAC6-specific inhibitors alters Fam65b expression, while dysferlin expression does not change. Inhibition of Fam65b expression in developing zebrafish results in abnormal muscle, with low birefringence, tears at the myosepta, and increased embryo lethality. Fam65b is an essential component of the HDAC6-dysferlin complex. Down-regulation of Fam65b in developing muscle causes changes consistent with muscle disease.-Balasubramanian, A., Kawahara, G., Gupta, V. A., Rozkalne, A., Beauvais, A., Kunkel, L. M., Gussoni, E. Fam65b is important for formation of the HDAC6-dysferlin protein complex during myogenic cell differentiation.


Asunto(s)
Diferenciación Celular/genética , Histona Desacetilasas/metabolismo , Proteínas de la Membrana/metabolismo , Células Musculares/metabolismo , Desarrollo de Músculos/genética , Proteínas Musculares/metabolismo , Proteínas/genética , Proteínas/metabolismo , Secuencia de Aminoácidos , Animales , Moléculas de Adhesión Celular , Células Cultivadas , Regulación hacia Abajo/genética , Disferlina , Histona Desacetilasa 6 , Histona Desacetilasas/genética , Humanos , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Proteínas Musculares/genética , Músculo Esquelético/metabolismo , Enfermedades Musculares/genética , Enfermedades Musculares/metabolismo , Unión Proteica/genética , Alineación de Secuencia , Tubulina (Proteína)/genética , Tubulina (Proteína)/metabolismo , Pez Cebra
7.
Hum Mol Genet ; 21(16): 3668-80, 2012 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-22634225

RESUMEN

Muscle side population (SP) cells are rare myogenic progenitors distinct from satellite cells, the known tissue-specific stem cells of skeletal muscle. Studies in mice demonstrated that muscle SP cells give rise to satellite cells in vivo. Given that muscle SP cells are heterogeneous, it has been difficult to prospectively enrich for myogenic progenitors within the SP fraction, particularly from human tissue. Further, conditions that favor the expansion of human muscle SP cells while retaining their myogenic potential have yet to be reported. In this study, human fetal muscle SP and main population (MP) cells were purified based on the expression of melanoma cell adhesion molecule (MCAM), a marker we previously reported to enrich for cells with myogenic potential. To define the relationship between MCAM expression and the degree of myogenic commitment, single cells were analyzed for the expression of myogenic-specific markers. Myogenic factors strongly associated with MCAM expression in single cells, particularly Myf5. Different MCAM+ populations, including SP cells, were expanded and assayed for fusion potential in vitro and engraftment potential in vivo. All MCAM+ subpopulations fused robustly into myotubes in vitro, whereas the MCAM- subpopulations did not. Further, MCAM+ SP cells exhibited the highest fusion potential in vitro and were the only fraction to engraft in vivo, although at low levels, following propagation. Thus, MCAM can be used to prospectively enrich for myogenic muscle SP cells in human fetal muscle. Moreover, we provide evidence that human MCAM+ SP cells have intrinsic myogenic activity that is retained after propagation.


Asunto(s)
Músculo Esquelético/citología , Células de Población Lateral/metabolismo , Animales , Biomarcadores/metabolismo , Antígeno CD146/metabolismo , Diferenciación Celular , Células Cultivadas , Humanos , Ratones , Ratones Endogámicos NOD , Fibras Musculares Esqueléticas/citología , Músculo Esquelético/embriología , Músculo Esquelético/metabolismo , Factor 5 Regulador Miogénico/metabolismo , Factor de Transcripción PAX7/metabolismo , Trasplante Heterólogo
8.
Nat Biotechnol ; 2024 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-39261590

RESUMEN

Experimental cell therapies for skeletal muscle conditions have shown little success, primarily because they use committed myogenic progenitors rather than true muscle stem cells, known as satellite cells. Here we present a method to generate in vitro-derived satellite cells (idSCs) from skeletal muscle tissue. When transplanted in small numbers into mouse muscle, mouse idSCs fuse into myofibers, repopulate the satellite cell niche, self-renew, support multiple rounds of muscle regeneration and improve force production on par with freshly isolated satellite cells in damaged skeletal muscle. We compared the epigenomic and transcriptional signatures between idSCs, myoblasts and satellite cells and used these signatures to identify core signaling pathways and genes that confer idSC functionality. Finally, from human muscle biopsies, we successfully generated satellite cell-like cells in vitro. After further development, idSCs may provide a scalable source of cells for the treatment of genetic muscle disorders, trauma-induced muscle damage and age-related muscle weakness.

9.
Am J Pathol ; 181(3): 961-8, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22841819

RESUMEN

X-linked myotubular myopathy is a severe congenital myopathy caused by deficiency of the lipid phosphatase, myotubularin. Recent studies of human tissue and animal models have discovered structural and physiological abnormalities in myotubularin-deficient muscle, but the impact of myotubularin deficiency on myogenic stem cells within muscles is unclear. In the present study, we evaluated the viability, proliferative capacity, and in vivo engraftment of myogenic cells obtained from severely symptomatic (Mtm1δ4) myotubularin-deficient mice. Mtm1δ4 muscle contains fewer myogenic cells than wild-type (WT) littermates, and the number of myogenic cells decreases with age. The behavior of Mtm1δ4 myoblasts is also abnormal, because they engraft poorly into C57BL/6/Rag1null/mdx5cv mice and display decreased proliferation and increased apoptosis compared with WT myoblasts. Evaluation of Mtm1δ4 animals at 21 and 42 days of life detected fewer satellite cells in Mtm1δ4 muscle compared with WT littermates, and the decrease in satellite cells correlated with progression of disease. In addition, analysis of WT and Mtm1δ4 regeneration after injury detected similar abnormalities of satellite cell function, with fewer satellite cells, fewer dividing cells, and increased apoptotic cells in Mtm1δ4 muscle. These studies demonstrate specific abnormalities in myogenic cell number and behavior that may relate to the progression of disease in myotubularin deficiency, and may also be used to develop in vitro assays by which novel treatment strategies can be assessed.


Asunto(s)
Apoptosis , Mioblastos/patología , Mioblastos/trasplante , Proteínas Tirosina Fosfatasas no Receptoras/deficiencia , Animales , Recuento de Células , Proliferación Celular , Supervivencia Celular , Progresión de la Enfermedad , Humanos , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/lesiones , Músculo Esquelético/patología , Mioblastos/metabolismo , Factor de Transcripción PAX7/metabolismo , Proteínas Tirosina Fosfatasas no Receptoras/metabolismo , Células Satélite del Músculo Esquelético/metabolismo , Células Satélite del Músculo Esquelético/patología
10.
Skelet Muscle ; 13(1): 18, 2023 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-37936227

RESUMEN

The tropomyosin genes (TPM1-4) contribute to the functional diversity of skeletal muscle fibers. Since its discovery in 1988, the TPM3 gene has been recognized as an indispensable regulator of muscle contraction in slow muscle fibers. Recent advances suggest that TPM3 isoforms hold more extensive functions during skeletal muscle development and in postnatal muscle. Additionally, mutations in the TPM3 gene have been associated with the features of congenital myopathies. The use of different in vitro and in vivo model systems has leveraged the discovery of several disease mechanisms associated with TPM3-related myopathy. Yet, the precise mechanisms by which TPM3 mutations lead to muscle dysfunction remain unclear. This review consolidates over three decades of research about the role of TPM3 in skeletal muscle. Overall, the progress made has led to a better understanding of the phenotypic spectrum in patients affected by mutations in this gene. The comprehensive body of work generated over these decades has also laid robust groundwork for capturing the multiple functions this protein plays in muscle fibers.


Asunto(s)
Enfermedades Musculares , Tropomiosina , Humanos , Tropomiosina/genética , Tropomiosina/metabolismo , Músculo Esquelético/metabolismo , Enfermedades Musculares/genética , Fibras Musculares Esqueléticas/metabolismo , Mutación
11.
Adv Biol (Weinh) ; 7(12): e2300157, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37434585

RESUMEN

Tetraspanins organize protein complexes at the cell membrane and are responsible for assembling diverse binding partners in changing cellular states. Tetraspanin CD82 is a useful cell surface marker for prospective isolation of human myogenic progenitors and its expression is decreased in Duchenne muscular dystrophy (DMD) cell lines. The function of CD82 in skeletal muscle remains elusive, partly because the binding partners of this tetraspanin in muscle cells have not been identified. CD82-associated proteins are sought to be identified in human myotubes via mass spectrometry proteomics, which identifies dysferlin and myoferlin as CD82-binding partners. In human dysferlinopathy (Limb girdle muscular dystrophy R2, LGMDR2) myogenic cell lines, expression of CD82 protein is near absent in two of four patient samples. In the cell lines where CD82 protein levels are unaffected, increased expression of the ≈72 kDa mini-dysferlin product is identified using an antibody recognizing the dysferlin C-terminus. These data demonstrate that CD82 binds dysferlin/myoferlin in differentiating muscle cells and its expression can be affected by loss of dysferlin in human myogenic cells.


Asunto(s)
Proteínas Musculares , Distrofias Musculares , Humanos , Disferlina/genética , Proteína Kangai-1 , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Distrofias Musculares/metabolismo , Tetraspaninas
12.
Proc Natl Acad Sci U S A ; 106(23): 9274-9, 2009 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-19470472

RESUMEN

Skeletal muscle is formed via fusion of myoblasts, a well-studied process in Drosophila. In vertebrates however, this process is less well understood, and whether there is evolutionary conservation with the proteins studied in flies is under investigation. Sticks and stones (Sns), a cell surface protein found on Drosophila myoblasts, has structural homology to nephrin. Nephrin is a protein expressed in kidney that is part of the filtration barrier formed by podocytes. No previous study has established any role for nephrin in skeletal muscle. We show, using two models, zebrafish and mice, that the absence of nephrin results in poorly developed muscles and incompletely fused myotubes, respectively. Although nephrin-knockout (nephrin(KO)) myoblasts exhibit prolonged activation of MAPK/ERK pathway during myogenic differentiation, expression of myogenin does not seem to be altered. Nevertheless, MAPK pathway blockade does not rescue myoblast fusion. Co-cultures of unaffected human fetal myoblasts with nephrin(KO) myoblasts or myotubes restore the formation of mature myotubes; however, the contribution of nephrin(KO) myoblasts is minimal. These studies suggest that nephrin plays a role in secondary fusion of myoblasts into nascent myotubes, thus establishing a possible functional conservation with Drosophila Sns.


Asunto(s)
Proteínas de la Membrana/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Mioblastos/metabolismo , Animales , Fusión Celular , Técnicas de Cocultivo , Humanos , Ratones , Músculo Esquelético/citología , Pez Cebra
13.
J Cell Biol ; 175(1): 99-110, 2006 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-17015616

RESUMEN

Skeletal muscle side population (SP) cells are thought to be "stem"-like cells. Despite reports confirming the ability of muscle SP cells to give rise to differentiated progeny in vitro and in vivo, the molecular mechanisms defining their phenotype remain unclear. In this study, gene expression analyses of human fetal skeletal muscle demonstrate that bone morphogenetic protein 4 (BMP4) is highly expressed in SP cells but not in main population (MP) mononuclear muscle-derived cells. Functional studies revealed that BMP4 specifically induces proliferation of BMP receptor 1a-positive MP cells but has no effect on SP cells, which are BMPR1a-negative. In contrast, the BMP4 antagonist Gremlin, specifically up-regulated in MP cells, counteracts the stimulatory effects of BMP4 and inhibits proliferation of BMPR1a-positive muscle cells. In vivo, BMP4-positive cells can be found in the proximity of BMPR1a-positive cells in the interstitial spaces between myofibers. Gremlin is expressed by mature myofibers and interstitial cells, which are separate from BMP4-expressing cells. Together, these studies propose that BMP4 and Gremlin, which are highly expressed by human fetal skeletal muscle SP and MP cells, respectively, are regulators of myogenic progenitor proliferation.


Asunto(s)
Proteínas Morfogenéticas Óseas/fisiología , Péptidos y Proteínas de Señalización Intercelular/fisiología , Fibras Musculares Esqueléticas/citología , Mioblastos Esqueléticos/citología , Proteína Morfogenética Ósea 4 , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/metabolismo , Proteínas Morfogenéticas Óseas/antagonistas & inhibidores , Proliferación Celular , Células Cultivadas , Feto , Humanos , Fibras Musculares Esqueléticas/metabolismo , Mioblastos Esqueléticos/metabolismo
14.
Muscle Nerve ; 43(1): 88-93, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21171099

RESUMEN

Erythropoietin promotes myoblast proliferation and inhibits fibrosis and thus it could impede the pathogenesis of muscle degenerative diseases. However, its stimulation of erythropoiesis limits its use as a therapeutic agent. An erythropoietin analog, carbamylated erythropoietin (C-EPO), retains these protective actions, yet it does not interact with the erythropoietin receptor. To determine whether treatment with C-EPO alleviates the signs of muscular dystrophy in an animal model of Duchenne muscular dystrophy, we treated mdx mice with intraperitoneal injections of 50 µg/kg and 100 µg/kg C-EPO for 4 and 12 weeks, and we monitored weight, serum creatine kinase levels, and changes in muscle histology. Moderate histological improvement was observed at 4 weeks, which did not translate into a significantly decreased level of serum creatine kinase. At the doses tested, C-EPO is not an effective therapeutic for the treatment of a mouse model of Duchenne muscular dystrophy.


Asunto(s)
Creatina Quinasa/antagonistas & inhibidores , Eritropoyetina/análogos & derivados , Músculo Esquelético/efectos de los fármacos , Distrofia Muscular Animal/tratamiento farmacológico , Animales , Proliferación Celular/efectos de los fármacos , Creatina Quinasa/metabolismo , Modelos Animales de Enfermedad , Eritropoyetina/administración & dosificación , Eritropoyetina/uso terapéutico , Femenino , Inyecciones Intraperitoneales , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiopatología , Distrofia Muscular Animal/metabolismo , Distrofia Muscular Animal/fisiopatología , Mioblastos/efectos de los fármacos , Mioblastos/metabolismo , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/uso terapéutico , Insuficiencia del Tratamiento
15.
Muscle Nerve ; 41(6): 746-50, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20513101

RESUMEN

We report a boy who received two allogeneic stem cell transplantations from umbilical cord donors to treat chronic granulomatous disease (CGD). The CGD was cured after the second transplantation, but 2.5 years later he was diagnosed with Duchenne muscular dystrophy (DMD). Examinations of his DNA, muscle tissue, and myoblast cultures derived from muscle tissue were performed to determine whether any donor dystrophin was being expressed. The boy was found to have a large-scale deletion on the X chromosome that spanned the loci for CYBB and DMD. The absence of dystrophin led to muscle histology characteristic of DMD. Analysis of myofibers demonstrated no definite donor cell engraftment. This case suggests that umbilical cord-derived hematopoietic stem cell transplantation will not be efficacious in the therapy of DMD without additional interventions that induce engraftment of donor cells in skeletal muscle.


Asunto(s)
Distrofina/deficiencia , Distrofina/genética , Enfermedad Granulomatosa Crónica/genética , Enfermedad Granulomatosa Crónica/cirugía , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/cirugía , Alemtuzumab , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales Humanizados , Anticuerpos Antineoplásicos/uso terapéutico , Niño , Mapeo Cromosómico , Cromosomas Humanos X , Ciclofosfamida/uso terapéutico , Estudios de Seguimiento , Eliminación de Gen , Regulación de la Expresión Génica , Humanos , Masculino , Reoperación , Trasplante Homólogo , Resultado del Tratamiento , Vidarabina/análogos & derivados , Vidarabina/uso terapéutico
16.
Skelet Muscle ; 10(1): 34, 2020 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-33243288

RESUMEN

BACKGROUND: Tetraspanins are a family of proteins known to assemble protein complexes at the cell membrane. They are thought to play diverse cellular functions in tissues by modifying protein-binding partners, thus bringing complexity and diversity in their regulatory networks. Previously, we identified the tetraspanin KAI/CD82 as a prospective marker for human muscle stem cells. CD82 expression appeared decreased in human Duchenne muscular dystrophy (DMD) muscle, suggesting a functional link to muscular dystrophy, yet whether this decrease is a consequence of dystrophic pathology or a compensatory mechanism in an attempt to rescue muscle from degeneration is currently unknown. METHODS: We studied the consequences of loss of CD82 expression in normal and dystrophic skeletal muscle and examined the dysregulation of downstream functions in mice aged up to 1 year. RESULTS: Expression of CD82 is important to sustain satellite cell activation, as in its absence there is decreased cell proliferation and less efficient repair of injured muscle. Loss of CD82 in dystrophic muscle leads to a worsened phenotype compared to control dystrophic mice, with decreased pulmonary function, myofiber size, and muscle strength. Mechanistically, decreased myofiber size in CD82-/- dystrophic mice is not due to altered PTEN/AKT signaling, although increased phosphorylation of mTOR at Ser2448 was observed. CONCLUSION: Basal CD82 expression is important to dystrophic muscle, as its loss leads to significantly weakened myofibers and impaired muscle function, accompanied by decreased satellite cell activity that is unable to protect and repair myofiber damage.


Asunto(s)
Proteína Kangai-1/metabolismo , Distrofia Muscular de Duchenne/metabolismo , Células Satélite del Músculo Esquelético/metabolismo , Animales , Proliferación Celular , Células Cultivadas , Femenino , Proteína Kangai-1/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Fuerza Muscular , Distrofia Muscular de Duchenne/genética , Fosfohidrolasa PTEN/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Células Satélite del Músculo Esquelético/fisiología , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo
17.
Methods Mol Biol ; 1889: 1-15, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30367405

RESUMEN

Primary myoblasts derived from human tissue are a valuable tool in research of muscle disease and pathophysiology. However, skeletal muscle biopsies, especially from diseased muscle, contain a plethora of non-myogenic cells, necessitating purification of the myogenic cell population. This protocol describes techniques for dissociation of cells from human skeletal muscle biopsies and enrichment for a highly myogenic population by fluorescence-activated cell sorting (FACS). We also describe methods for assessing myogenicity and population expansion for subsequent in vitro study.


Asunto(s)
Separación Celular , Citometría de Flujo , Músculo Esquelético/citología , Mioblastos Esqueléticos/citología , Mioblastos Esqueléticos/metabolismo , Biomarcadores , Diferenciación Celular , Separación Celular/métodos , Células Cultivadas , Citometría de Flujo/métodos , Técnica del Anticuerpo Fluorescente , Humanos , Inmunofenotipificación
18.
Dev Biol ; 311(2): 449-63, 2007 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-17919536

RESUMEN

Satellite cells are recognized as the main source for myoblasts in postnatal muscle. The possible participation of other cell types in myofiber maintenance remains a subject of debate. Here, we investigated the potential of vascular preparations from mouse retina to undergo myogenesis when cultured alone or with differentiated primary myogenic cultures. The choice of retina, an organ richly supplied with capillary network and anatomically separated from skeletal muscles, ensures that the vasculature preparation is devoid of satellite cells. We demonstrate that retina-derived cells spontaneously fuse with preexisting myotubes and contribute additional myonuclei, some of which initiate expression of muscle-specific genes after fusion. Myogenic differentiation of retinal cells prior to their fusion with preexisting myotubes was not detected. Although originating from vasculature preparations, nuclei undergoing myogenic reprogramming were contributed by cells that were neither endothelial nor blood borne. Our results suggest smooth muscle/pericytes as the possible source, and that myogenic reprogramming depends on the muscle specific transcription factor MyoD. Our studies provide insights into a novel avenue for myofiber maintenance, relying on nuclei of non-myogenic origin that undergo fusion and subsequent myogenic conversion within host myofibers. This process may support ongoing myofiber maintenance throughout life.


Asunto(s)
Fusión Celular , Desarrollo de Músculos/fisiología , Fibras Musculares Esqueléticas/metabolismo , Retina/citología , Animales , Células Cultivadas , Pollos , Técnicas de Cocultivo , Células Endoteliales/citología , Células Endoteliales/metabolismo , Regulación de la Expresión Génica , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Hibridación Fluorescente in Situ , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microcirculación/citología , Fibras Musculares Esqueléticas/citología , Músculo Liso/citología , Proteína MioD/genética , Proteína MioD/metabolismo , Factor 5 Regulador Miogénico/genética , Factor 5 Regulador Miogénico/metabolismo , Células Satélite del Músculo Esquelético/citología , Células Satélite del Músculo Esquelético/metabolismo
19.
J Clin Invest ; 110(6): 807-14, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12235112

RESUMEN

Duchenne muscular dystrophy (DMD) is a severe progressive muscle-wasting disorder caused by mutations in the dystrophin gene. Studies have shown that bone marrow cells transplanted into lethally irradiated mdx mice, the mouse model of DMD, can become part of skeletal muscle myofibers. Whether human marrow cells also have this ability is unknown. Here we report the analysis of muscle biopsies from a DMD patient (DMD-BMT1) who received bone marrow transplantation at age 1 year for X-linked severe combined immune deficiency and who was diagnosed with DMD at age 12 years. Analysis of muscle biopsies from DMD-BMT1 revealed the presence of donor nuclei within a small number of muscle myofibers (0.5-0.9%). The majority of the myofibers produce a truncated, in-frame isoform of dystrophin lacking exons 44 and 45 (not wild-type). The presence of bone marrow-derived donor nuclei in the muscle of this patient documents the ability of exogenous human bone marrow cells to fuse into skeletal muscle and persist up to 13 years after transplantation.


Asunto(s)
Células de la Médula Ósea/fisiología , Trasplante de Médula Ósea , Músculo Esquelético/patología , Distrofia Muscular de Duchenne/patología , Adolescente , Animales , Biopsia , Células de la Médula Ósea/ultraestructura , Núcleo Celular/ultraestructura , Niño , Distrofina/genética , Distrofina/metabolismo , Exones/genética , Femenino , Humanos , Inmunohistoquímica , Hibridación Fluorescente in Situ , Lactante , Masculino , Músculo Esquelético/citología , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/metabolismo , Inmunodeficiencia Combinada Grave/terapia , Factores de Tiempo
20.
Bio Protoc ; 7(21)2017 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-29152538

RESUMEN

Primary myoblast culture is a valuable tool in research of muscle disease, pathophysiology, and pharmacology. This protocol describes techniques for dissociation of cells from human skeletal muscle biopsies and enrichment for a highly myogenic population by fluorescence-activated cell sorting (FACS). We also describe methods for assessing myogenicity and population expansion for subsequent in vitro study.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA