Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
8.
PLoS One ; 12(12): e0189324, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29228025

RESUMEN

Primordial growth failure has been linked to defects in the biology of cell division and replication. The complex processes involved in microtubule spindle formation, organization and function have emerged as a dominant patho-mechanism in these conditions. The majority of reported disease genes encode for centrosome and centriole proteins, leaving kinetochore proteins by which the spindle apparatus interacts with the chromosomes largely unaccounted for. We report a novel disease gene encoding the constitutive inner kinetochore member CENPT, which is involved in kinetochore targeting and assembly, resulting in severe growth failure in two siblings of a consanguineous family. We herein present studies on the molecular and cellular mechanisms that explain how genetic mutations in this gene lead to primordial growth failure. In both, affected human cell lines and a zebrafish knock-down model of Cenpt, we observed aberrations in cell division with abnormal accumulation of micronuclei and of nuclei with increased DNA content arising from incomplete and/or irregular chromosomal segregation. Our studies underscore the critical importance of kinetochore function for overall body growth and provide new insight into the cellular mechanisms implicated in the spectrum of these severe growth disorders.


Asunto(s)
Proteínas Cromosómicas no Histona/genética , Trastornos del Crecimiento/genética , Animales , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Modelos Animales , Pez Cebra/genética
9.
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.

10.
Cell Stem Cell ; 20(1): 1-3, 2017 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-28061348

RESUMEN

Exosome-dependent intercellular communication is an emerging signaling mechanism involved in tissue repair and regeneration. Now in this issue of Cell Stem Cell, Fry et al. (2016) show that muscle stem cells communicate with fibrogenic cells by exosomal trafficking of microRNA-206 to regulate extracellular matrix deposition and muscle tissue remodeling.


Asunto(s)
Comunicación Celular , Exosomas , Humanos , Mioblastos , Regeneración , Células Madre
11.
Cell Stem Cell ; 19(6): 800-807, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27641304

RESUMEN

Cell-surface markers for prospective isolation of stem cells from human skeletal muscle have been difficult to identify. Such markers would be powerful tools for studying satellite cell function during homeostasis and in pathogenesis of diseases such as muscular dystrophies. In this study, we show that the tetraspanin KAI/CD82 is an excellent marker for prospectively isolating stem cells from human fetal and adult skeletal muscle. Human CD82+ muscle cells robustly engraft into a mouse model of muscular dystrophy. shRNA knockdown of CD82 in myogenic cells reduces myoblast proliferation, suggesting it is functionally involved in muscle homeostasis. CD82 physically interacts with alpha7beta1 integrin (α7ß1-ITG) and with α-sarcoglycan, a member of the Dystrophin-Associated Glycoprotein Complex (DAPC), both of which have been linked to muscular dystrophies. Consistently, CD82 expression is decreased in Duchenne muscular dystrophy patients. Together, these findings suggest that CD82 function may be important for muscle stem cell function in muscular disorders.


Asunto(s)
Separación Celular/métodos , Proteína Kangai-1/metabolismo , Distrofias Musculares/metabolismo , Distrofias Musculares/patología , Células Satélite del Músculo Esquelético/metabolismo , Adulto , Animales , Antígenos CD/metabolismo , Biomarcadores/metabolismo , Antígeno CD146/metabolismo , Células HEK293 , Humanos , Inmunoprecipitación , Cadenas alfa de Integrinas/metabolismo , Ratones SCID , Distrofia Muscular Animal/patología , Sarcoglicanos/metabolismo
12.
Nat Biotechnol ; 33(9): 962-9, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26237517

RESUMEN

During embryonic development, skeletal muscles arise from somites, which derive from the presomitic mesoderm (PSM). Using PSM development as a guide, we establish conditions for the differentiation of monolayer cultures of mouse embryonic stem (ES) cells into PSM-like cells without the introduction of transgenes or cell sorting. We show that primary and secondary skeletal myogenesis can be recapitulated in vitro from the PSM-like cells, providing an efficient, serum-free protocol for the generation of striated, contractile fibers from mouse and human pluripotent cells. The mouse ES cells also differentiate into Pax7(+) cells with satellite cell characteristics, including the ability to form dystrophin(+) fibers when grafted into muscles of dystrophin-deficient mdx mice, a model of Duchenne muscular dystrophy (DMD). Fibers derived from ES cells of mdx mice exhibit an abnormal branched phenotype resembling that described in vivo, thus providing an attractive model to study the origin of the pathological defects associated with DMD.


Asunto(s)
Diferenciación Celular , Modelos Animales de Enfermedad , Fibras Musculares Esqueléticas/patología , Distrofia Muscular de Duchenne/patología , Células Madre Pluripotentes/patología , Animales , Células Cultivadas , Ratones , Ratones Transgénicos
13.
J Vis Exp ; (95): 52307, 2015 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-25651101

RESUMEN

The generation of patient-specific cell lines represents an invaluable tool for diagnostic or translational research, and these cells can be collected from skin or muscle biopsy tissue available during the patient's diagnostic workup. In this protocol, we describe a technique for live cell isolation from small amounts of muscle or skin tissue for primary cell culture. Additionally, we provide a technique for the immortalization of myogenic cell lines and fibroblast cell lines from primary cells. Once cell lines are immortalized, substantial expansion of patient-derived cells can be achieved. Immortalized cells are amenable to many downstream applications, including drug screening and in vitro correction of the genetic mutation. Altogether, these protocols provide a reliable tool to generate and preserve patient-derived cells for downstream applications.


Asunto(s)
Técnicas Citológicas/métodos , Fibroblastos/citología , Técnicas de Diagnóstico Molecular/métodos , Músculos/citología , Piel/citología , Biopsia/métodos , Línea Celular , Separación Celular/métodos , Humanos
14.
J Vis Exp ; (89)2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-25078247

RESUMEN

Skeletal muscle is a unique tissue because of its structure and function, which requires specific protocols for tissue collection to obtain optimal results from functional, cellular, molecular, and pathological evaluations. Due to the subtlety of some pathological abnormalities seen in congenital muscle disorders and the potential for fixation to interfere with the recognition of these features, pathological evaluation of frozen muscle is preferable to fixed muscle when evaluating skeletal muscle for congenital muscle disease. Additionally, the potential to produce severe freezing artifacts in muscle requires specific precautions when freezing skeletal muscle for histological examination that are not commonly used when freezing other tissues. This manuscript describes a protocol for rapid freezing of skeletal muscle using isopentane (2-methylbutane) cooled with liquid nitrogen to preserve optimal skeletal muscle morphology. This procedure is also effective for freezing tissue intended for genetic or protein expression studies. Furthermore, we have integrated our freezing protocol into a broader procedure that also describes preferred methods for the short term triage of tissue for (1) single fiber functional studies and (2) myoblast cell culture, with a focus on the minimum effort necessary to collect tissue and transport it to specialized research or reference labs to complete these studies. Overall, this manuscript provides an outline of how fresh tissue can be effectively distributed for a variety of phenotypic studies and thereby provides standard operating procedures (SOPs) for pathological studies related to congenital muscle disease.


Asunto(s)
Músculo Esquelético/anatomía & histología , Músculo Esquelético/fisiología , Enfermedades Musculares/congénito , Enfermedades Musculares/patología , Fijación del Tejido/métodos , Animales , Técnicas de Cultivo de Célula/métodos , Modelos Animales de Enfermedad , Congelación , Ratones , Músculo Esquelético/patología , Fenotipo
15.
Nat Commun ; 5: 4063, 2014 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-24898859

RESUMEN

Developing human muscle contains inter-myofibre progenitors expressing Bmp-receptor 1a (Bmpr1a) and Myf5 that respond to stimulation with Bmp4. Here we ablate Bmpr1a in Myf5- and MyoD-expressing cells in vivo. Mutant mice reveal increased intramuscular fat and reduced myofibre size in selected muscles, or following muscle injury. Myo-endothelial progenitors are the most affected cell type: clonal studies demonstrate that ablation of Bmpr1a in myo-endothelial cells results in decreased myogenic activity, while adipogenic differentiation is significantly increased. Downstream phospho-Smad 1, 5, 8 signalling is also severely decreased in mutant myo-endothelial cells. Lineage tracing of endothelial cells using VE-cadherin(Cre) driver failed to reveal a significant contribution of these cells to developing or injured skeletal muscle. Thus, myo-endothelial progenitors with functioning Bmpr1a signalling demonstrate myogenic potential, but their main function in vivo is to inhibit intramuscular adipogenesis, both through a cell-autonomous and a cell-cell interaction mechanism.


Asunto(s)
Adipogénesis/genética , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/genética , Músculo Esquelético/metabolismo , Células Madre/metabolismo , Animales , Antígenos CD , Cadherinas , Células Endoteliales/metabolismo , Ratones , Ratones Noqueados , Proteína MioD/metabolismo , Factor 5 Regulador Miogénico/metabolismo , Transducción de Señal , Proteína Smad1/metabolismo , Proteína Smad5/metabolismo , Proteína Smad8/metabolismo
16.
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
17.
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
18.
Hum Gene Ther ; 25(1): 73-81, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24152287

RESUMEN

Abstract Stem cell transplantation is being tested as a potential therapy for a number of diseases. Stem cells isolated directly from tissue specimens or generated via reprogramming of differentiated cells require rigorous testing for both safety and efficacy in preclinical models. The availability of mice with immune-deficient background that carry additional mutations in specific genes facilitates testing the efficacy of cell transplantation in disease models. The muscular dystrophies are a heterogeneous group of disorders, of which Duchenne muscular dystrophy is the most severe and common type. Cell-based therapy for muscular dystrophy has been under investigation for several decades, with a wide selection of cell types being studied, including tissue-specific stem cells and reprogrammed stem cells. Several immune-deficient mouse models of muscular dystrophy have been generated, in which human cells obtained from various sources are injected to assess their preclinical potential. After transplantation, the presence of engrafted human cells is detected via immunofluorescence staining, using antibodies that recognize human, but not mouse, proteins. Here we show that one antibody specific to human spectrin, which is commonly used to evaluate the efficacy of transplanted human cells in mouse muscle, detects myofibers in muscles of NOD/Rag1(null)mdx(5cv), NOD/LtSz-scid IL2Rγ(null) mice, or mdx nude mice, irrespective of whether they were injected with human cells. These "reactive" clusters are regenerating myofibers, which are normally present in dystrophic tissue and the spectrin antibody is likely recognizing utrophin, which contains spectrin-like repeats. Therefore, caution should be used in interpreting data based on detection of single human-specific proteins, and evaluation of human stem cell engraftment should be performed using multiple human-specific labeling strategies.


Asunto(s)
Fibras Musculares Esqueléticas/metabolismo , Regeneración , Espectrina/metabolismo , Donantes de Tejidos , Animales , Antígeno CD146/metabolismo , Modelos Animales de Enfermedad , Reacciones Falso Positivas , Femenino , Humanos , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos mdx , Fibras Musculares Esqueléticas/citología , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/terapia , Mioblastos Esqueléticos/citología , Mioblastos Esqueléticos/metabolismo , Mioblastos Esqueléticos/trasplante
19.
FEBS J ; 280(23): 6097-113, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24102982

RESUMEN

Mammalian muscle cell differentiation is a complex process of multiple steps for which many of the factors involved have not yet been defined. In a screen to identify the regulators of myogenic cell fusion, we found that the gene for G-protein coupled receptor 56 (GPR56) was transiently up-regulated during the early fusion of human myoblasts. Human mutations in the gene for GPR56 cause the disease bilateral frontoparietal polymicrogyria; however, the consequences of receptor dysfunction on muscle development have not been explored. Using knockout mice, we defined the role of GPR56 in skeletal muscle. GPR56(-/-) myoblasts have decreased fusion and smaller myotube sizes in culture. In addition, a loss of GPR56 expression in muscle cells results in decreases or delays in the expression of myogenic differentiation 1, myogenin and nuclear factor of activated T-cell (NFAT)c2. Our data suggest that these abnormalities result from decreased GPR56-mediated serum response element and NFAT signalling. Despite these changes, no overt differences in phenotype were identified in the muscle of GPR56 knockout mice, which presented only a mild but statistically significant elevation of serum creatine kinase compared to wild-type. In agreement with these findings, clinical data from 13 bilateral frontoparietal polymicrogyria patients revealed mild serum creatine kinase increase in only two patients. In summary, targeted disruption of GPR56 in mice results in myoblast abnormalities. The absence of a severe muscle phenotype in GPR56 knockout mice and human patients suggests that other factors may compensate for the lack of this G-protein coupled receptor during muscle development and that the motor delay observed in these patients is likely not a result of primary muscle abnormalities.


Asunto(s)
Fusión Celular , Malformaciones del Desarrollo Cortical/patología , Desarrollo de Músculos/fisiología , Mioblastos/citología , Factores de Transcripción NFATC/metabolismo , Receptores Acoplados a Proteínas G/fisiología , Elemento de Respuesta al Suero/genética , Animales , Western Blotting , Comunicación Celular , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Humanos , Técnicas para Inmunoenzimas , Luciferasas/metabolismo , Masculino , Malformaciones del Desarrollo Cortical/genética , Malformaciones del Desarrollo Cortical/metabolismo , Ratones , Ratones Noqueados , Proteína MioD/genética , Proteína MioD/metabolismo , Mioblastos/metabolismo , Miogenina/genética , Miogenina/metabolismo , Factores de Transcripción NFATC/antagonistas & inhibidores , Factores de Transcripción NFATC/genética , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA