Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
EMBO Mol Med ; 12(1): e11019, 2020 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-31793167

RESUMEN

Duchenne muscular dystrophy (DMD) is a debilitating fatal X-linked muscle disorder. Recent findings indicate that IGFs play a central role in skeletal muscle regeneration and development. Among IGFs, insulinlike growth factor 2 (IGF2) is a key regulator of cell growth, survival, migration and differentiation. The type 2 IGF receptor (IGF2R) modulates circulating and tissue levels of IGF2 by targeting it to lysosomes for degradation. We found that IGF2R and the store-operated Ca2+ channel CD20 share a common hydrophobic binding motif that stabilizes their association. Silencing CD20 decreased myoblast differentiation, whereas blockade of IGF2R increased proliferation and differentiation in myoblasts via the calmodulin/calcineurin/NFAT pathway. Remarkably, anti-IGF2R induced CD20 phosphorylation, leading to the activation of sarcoplasmic/endoplasmic reticulum Ca2+ -ATPase (SERCA) and removal of intracellular Ca2+ . Interestingly, we found that IGF2R expression was increased in dystrophic skeletal muscle of human DMD patients and mdx mice. Blockade of IGF2R by neutralizing antibodies stimulated muscle regeneration, induced force recovery and normalized capillary architecture in dystrophic mdx mice representing an encouraging starting point for the development of new biological therapies for DMD.


Asunto(s)
Músculo Esquelético/crecimiento & desarrollo , Distrofia Muscular de Duchenne/tratamiento farmacológico , Receptor IGF Tipo 2/antagonistas & inhibidores , Regeneración , Animales , Sitios de Unión , Niño , Humanos , Ratones , Ratones Endogámicos mdx , Mioblastos , Adulto Joven
2.
Mol Ther ; 24(11): 1949-1964, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27506452

RESUMEN

Duchenne muscular dystrophy is the most common genetic muscular dystrophy. It is caused by mutations in the dystrophin gene, leading to absence of muscular dystrophin and to progressive degeneration of skeletal muscle. We have demonstrated that the exon skipping method safely and efficiently brings to the expression of a functional dystrophin in dystrophic CD133+ cells injected scid/mdx mice. Golden Retriever muscular dystrophic (GRMD) dogs represent the best preclinical model of Duchenne muscular dystrophy, mimicking the human pathology in genotypic and phenotypic aspects. Here, we assess the capacity of intra-arterial delivered autologous engineered canine CD133+ cells of restoring dystrophin expression in Golden Retriever muscular dystrophy. This is the first demonstration of five-year follow up study, showing initial clinical amelioration followed by stabilization in mild and severe affected Golden Retriever muscular dystrophy dogs. The occurrence of T-cell response in three Golden Retriever muscular dystrophy dogs, consistent with a memory response boosted by the exon skipped-dystrophin protein, suggests an adaptive immune response against dystrophin.


Asunto(s)
Antígeno AC133/metabolismo , Inmunidad Adaptativa , Distrofia Muscular Animal/terapia , Trasplante de Células Madre/métodos , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Perros , Estudios de Seguimiento , Humanos , Distrofia Muscular Animal/inmunología , Células Madre/metabolismo , Trasplante Autólogo , Resultado del Tratamiento
3.
Development ; 143(4): 658-69, 2016 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-26884398

RESUMEN

Duchenne muscular dystrophy (DMD) is a progressive neuromuscular disorder characterized by muscle wasting and premature death. The defective gene is dystrophin, a structural protein, absence of which causes membrane fragility and myofiber necrosis. Several lines of evidence showed that in adult DMD patients dystrophin is involved in signaling pathways that regulate calcium homeostasis and differentiation programs. However, secondary aspects of the disease, such as inflammation and fibrosis development, might represent a bias in the analysis. Because fetal muscle is not influenced by gravity and does not suffer from mechanical load and/or inflammation, we investigated 12-week-old fetal DMD skeletal muscles, highlighting for the first time early alterations in signaling pathways mediated by the absence of dystrophin itself. We found that PLC/IP3/IP3R/Ryr1/Ca(2+) signaling is widely active in fetal DMD skeletal muscles and, through the calcium-dependent PKCα protein, exerts a fundamental regulatory role in delaying myogenesis and in myofiber commitment. These data provide new insights into the origin of DMD pathology during muscle development.


Asunto(s)
Señalización del Calcio , Feto/metabolismo , Inositol 1,4,5-Trifosfato/metabolismo , Desarrollo de Músculos , Músculo Esquelético/embriología , Distrofia Muscular de Duchenne/embriología , Distrofia Muscular de Duchenne/metabolismo , Animales , Biomarcadores/metabolismo , Biopsia , Calcio/metabolismo , Canales de Calcio/metabolismo , Feto/patología , Regulación del Desarrollo de la Expresión Génica , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Modelos Biológicos , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patología , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Distrofia Muscular Animal/metabolismo , Distrofia Muscular Animal/patología , Distrofia Muscular de Duchenne/patología , Factor de Transcripción PAX7/metabolismo , Proteína Quinasa C-alfa/metabolismo
4.
FEBS J ; 280(23): 6045-60, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24028392

RESUMEN

The protein dysferlin is abundantly expressed in skeletal and cardiac muscles, where its main function is membrane repair. Mutations in the dysferlin gene are involved in two autosomal recessive muscular dystrophies: Miyoshi myopathy and limb-girdle muscular dystrophy type 2B. Development of effective therapies remains a great challenge. Strategies to repair the dysferlin gene by skipping mutated exons, using antisense oligonucleotides (AONs), may be suitable only for a subset of mutations, while cell and gene therapy can be extended to all mutations. AON-treated blood-derived CD133+ stem cells isolated from patients with Miyoshi myopathy led to partial dysferlin reconstitution in vitro but failed to express dysferlin after intramuscular transplantation into scid/blAJ dysferlin null mice. We thus extended these experiments producing the full-length dysferlin mediated by a lentiviral vector in blood-derived CD133+ stem cells isolated from the same patients. Transplantation of engineered blood-derived CD133+ stem cells into scid/blAJ mice resulted in sufficient dysferlin expression to correct functional deficits in skeletal muscle membrane repair. Our data suggest for the first time that lentivirus-mediated delivery of full-length dysferlin in stem cells isolated from Miyoshi myopathy patients could represent an alternative therapeutic approach for treatment of dysferlinopathies.


Asunto(s)
Antígenos CD/metabolismo , Miopatías Distales/terapia , Glicoproteínas/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Musculares/metabolismo , Músculo Esquelético/patología , Atrofia Muscular/terapia , Oligonucleótidos Antisentido/farmacología , Péptidos/metabolismo , Trasplante de Células Madre , Células Madre/citología , Antígeno AC133 , Adulto , Animales , Western Blotting , Células Cultivadas , Miopatías Distales/genética , Miopatías Distales/patología , Disferlina , Técnica del Anticuerpo Fluorescente , Humanos , Técnicas para Inmunoenzimas , Hibridación Fluorescente in Situ , Inyecciones Intramusculares , Lentivirus/genética , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos A , Ratones SCID , Proteínas Musculares/genética , Músculo Esquelético/lesiones , Músculo Esquelético/metabolismo , Atrofia Muscular/genética , Atrofia Muscular/patología , Mutación/genética , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre/metabolismo
5.
FEBS J ; 280(17): 4251-62, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23206279

RESUMEN

Muscular dystrophies are heritable and heterogeneous neuromuscular disorders characterized by the primary wasting of skeletal muscle, usually caused by mutations in the proteins forming the link between the cytoskeleton and the basal lamina. As a result of mutations in the dystrophin gene, Duchenne muscular dystrophy patients suffer from progressive muscle atrophy and an exhaustion of muscular regenerative capacity. No efficient therapies are available. The evidence that adult stem cells were capable of participating in the regeneration of more than their resident organ led to the development of potential stem cell treatments for degenerative disorder. In the present review, we describe the different types of myogenic stem cells and their possible use for the progression of cell therapy in Duchenne muscular dystrophy.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos , Distrofia Muscular de Duchenne/terapia , Trasplante de Células Madre , Células Madre/citología , Animales , Humanos , Distrofia Muscular de Duchenne/metabolismo , Células Madre/metabolismo
6.
Int J Biochem Cell Biol ; 44(12): 2095-105, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22982241

RESUMEN

Among the scarce available data about the biological role of the membrane protein CD20, there is some evidence that this protein functions as a store-operated Ca(2+) channel and/or regulates transmembrane Ca(2+) trafficking. Recent findings indicate that store-operated Ca(2+) entry (SOCE) plays a central role in skeletal muscle function and development, but there remain a number of unresolved issues relating to SOCE modulation in this tissue. Here we describe CD20 expression in skeletal muscle, verifying its membrane localization in myoblasts and adult muscle fibers. Additionally, we show that inhibition of CD20 through antibody binding or gene silencing resulted in specific impairment of SOCE in C2C12 myoblasts. Our results provide novel insights into the CD20 expression pattern, and suggest that functional CD20 is required for SOCE to consistently occur in C2C12 myoblasts. These findings may contribute to future identification of mechanisms and molecules involved in the fine regulation of store-operated Ca(2+) entry in skeletal muscle.


Asunto(s)
Antígenos CD20/metabolismo , Señalización del Calcio/efectos de los fármacos , Expresión Génica , Fibras Musculares Esqueléticas/metabolismo , Secuencia de Aminoácidos , Animales , Anticuerpos/farmacología , Antígenos CD20/química , Antígenos CD20/genética , Antígenos CD20/inmunología , Línea Celular , Membrana Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones SCID , Datos de Secuencia Molecular , Músculo Esquelético/citología , Músculo Esquelético/metabolismo , Mioblastos/metabolismo , Interferencia de ARN
7.
PLoS One ; 7(8): e43464, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22912879

RESUMEN

BACKGROUND: MicroRNAs (miRNAs) have been recently involved in most of human diseases as targets for potential strategies to rescue the pathological phenotype. Since the skeletal muscle is a spread-wide highly differentiated and organized tissue, rescue of severely compromised muscle still remains distant from nowadays. For this reason, we aimed to identify a subset of miRNAs major involved in muscle remodelling and regeneration by analysing the miRNA-profile of single fibres isolated from dystrophic muscle, which was here considered as a model of chronic damage. METHODOLOGY/PRINCIPAL FINDINGS: The miRNA-signature associated to regenerating (newly formed) and remodelling (resting) fibres was investigated in animal models of muscular dystrophies and acute damage, in order to distinguish which miRNAs are primary related to muscle regeneration. In this study we identify fourteen miRNAs associated to dystrophic fibres responsible for muscle regeneration and remodelling, and confirm over-expression of the previously identified regeneration-associated myomiR-206. In particular, a functional binding site for myomiR-206 was identified and validated in the 3'untranslated region (3'UTR) of an X-linked member of a family of sequence independent chromatin-binding proteins (Hmgb3) that is preferentially expressed in hematopoietic stem cells. During regeneration of single muscle fibres, Hmgb3 messenger RNA (mRNA) and protein expression was gradually reduced, concurrent with the up-regulation of miR-206. CONCLUSION/SIGNIFICANCE: Our results elucidate a negative feedback circuit in which myomiR-206 represses Hmgb3 expression to modulate the regeneration of single muscle fibres after acute and chronic muscle damage. These findings suggest that myomiR-206 may be a potential therapeutic target in muscle diseases.


Asunto(s)
Proteína HMGB3/genética , MicroARNs/genética , Músculo Esquelético/metabolismo , Regeneración/genética , Regiones no Traducidas 3'/genética , Adolescente , Animales , Animales Recién Nacidos , Sitios de Unión/genética , Western Blotting , Niño , Preescolar , Perfilación de la Expresión Génica , Células HEK293 , Proteína HMGB3/metabolismo , Humanos , Lactante , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , MicroARNs/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patología , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Distrofias Musculares/genética , Distrofias Musculares/metabolismo , Distrofias Musculares/fisiopatología , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/metabolismo , Distrofia Muscular Animal/fisiopatología , Células 3T3 NIH , Análisis de Secuencia por Matrices de Oligonucleótidos
8.
Exp Cell Res ; 318(10): 1160-74, 2012 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-22465227

RESUMEN

Dysferlin mutations cause muscular dystrophy (dysferlinopathy) characterized by adult onset muscle weakness, high serum creatine kinase levels, attenuation of muscle regeneration and a prominent inflammatory infiltrate. In order to verify the role of lymphocytes and immune cells on this disease, we generated the Scid/A/J transgenic mice and compared these animals with the age-matched A/J mice. The absence of T and B lymphocytes in this animal model of dysferlinopathy resulted in an improvement of the muscle regeneration. Scid/A/J mice showed increased specific force in the myosin heavy chain 2A-expressing fibers of the diaphragm and abdominal muscles. Moreover, a partial reduction in complement deposition was observed together with a diminution in pro-inflammatory M1 macrophages. Consistent with this model, T and B lymphocytes seem to have a role in the muscle damaging immune response. The knowledge of the involvement of immune system in the development of dysferlinopathies could represent an important tool for their rescuing. By studying Scid/blAJ mice, we showed that it could be possible to modulate the pathological symptoms of these diseases by interfering with different components of the immune system.


Asunto(s)
Linfocitos B/patología , Proteínas de la Membrana/deficiencia , Músculo Esquelético/fisiopatología , Distrofia Muscular Animal/patología , Linfocitos T/patología , Animales , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Modelos Animales de Enfermedad , Disferlina , Distrofina/metabolismo , Células Endoteliales/patología , Femenino , Hibridación Genética , Técnicas In Vitro , Inflamación , Laminina/metabolismo , Macrófagos/patología , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones SCID , Contracción Muscular , Músculo Esquelético/irrigación sanguínea , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Distrofia Muscular Animal/metabolismo , Regeneración , Sarcoglicanos/metabolismo , Sarcolema/genética , Sarcolema/metabolismo , Sarcolema/patología
9.
BioDrugs ; 24(4): 237-47, 2010 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-20623990

RESUMEN

Muscular dystrophies are heritable, heterogeneous neuromuscular disorders and include Duchenne and Becker muscular dystrophies (DMD and BMD, respectively). DMD patients exhibit progressive muscle weakness and atrophy followed by exhaustion of muscular regenerative capacity, fibrosis, and eventually disruption of the muscle tissue architecture. In-frame mutations in the dystrophin gene lead to expression of a partially functional protein, resulting in the milder BMD. No effective therapies are available at present. Cell-based therapies have been attempted in an effort to promote muscle regeneration, with the hope that the host cells would repopulate the muscle and improve muscle function and pathology. Injection of adult myoblasts has led to the development of new muscle fibers, but several limitations have been identified, such as poor cell survival and limited migratory ability. As an alternative to myoblasts, stem cells were considered preferable for therapeutic applications because of their capacity for self-renewal and differentiation potential. In recent years, encouraging results have been obtained with adult stem cells to treat human diseases such as leukemia, Parkinson's disease, stroke, and muscular dystrophies. Embryonic stem cells (ESCs) can be derived from mammalian embryos in the blastocyst stage, and because they can differentiate into a wide range of specialized cells, they hold potential for use in treating almost all human diseases. Several ongoing studies focus on this possibility, evaluating differentiation of specific cell lines from human ESCs (hESCs) as well as the potential tumorigenicity of hESCs. The most important limitation with using hESCs is that it requires destruction of human blastocysts or embryos. Conversely, adult stem cells have been identified in various tissues, where they serve to maintain, generate, and replace terminally differentiated cells within their specific tissue as the need arises for cell turnover or from tissue injury. Moreover, these cells can participate in regeneration of more than just their specific tissue type. Here we describe multiple types of muscle- and fetal-derived myogenic stem cells, their characterization, and their possible use in treating muscular dystrophies such as DMD and BMD. We also emphasize that the most promising possibility for the management and therapy of DMD and BMD is a combination of different approaches, such as gene and stem cell therapy.


Asunto(s)
Células Madre Adultas/trasplante , Células Madre Embrionarias/trasplante , Músculo Esquelético/citología , Distrofias Musculares/terapia , Células Madre Pluripotentes/trasplante , Trasplante de Células Madre , Adulto , Células Madre Adultas/citología , Células Madre Adultas/fisiología , Animales , Diferenciación Celular , Células Madre Embrionarias/citología , Células Madre Embrionarias/fisiología , Femenino , Terapia Genética , Humanos , Masculino , Músculo Esquelético/fisiología , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/fisiología , Regeneración , Trasplante de Células Madre/tendencias
10.
PLoS One ; 3(5): e2218, 2008 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-18493616

RESUMEN

BACKGROUND: Various prognostic serum and cellular markers have been identified for many diseases, such as cardiovascular diseases and tumor pathologies. Here we assessed whether the levels of certain stem cells may predict the progression of Duchenne muscular dystrophy (DMD). METHODS AND FINDINGS: The levels of several subpopulations of circulating stem cells expressing the CD133 antigen were determined by flow cytometry in 70 DMD patients. The correlation between the levels and clinical status was assessed by statistical analysis. The median (+/-SD) age of the population was 10.66+/-3.81 (range 3 to 20 years). The levels of CD133+CXCR4+CD34- stem cells were significantly higher in DMD patients compared to healthy controls (mean+/-standard deviation: 17.38+/-1.38 vs. 11.0+/-1.70; P = 0.03) with a tendency towards decreased levels in older patients. Moreover, the levels of this subpopulation of cells correlated with the clinical condition. In a subgroup of 19 DMD patients after 24 months of follow-up, increased levels of CD133+CXCR4+CD34- cells was shown to be associated with a phenotype characterised by slower disease progression. The circulating CD133+CXCR4+CD34- cells in patients from different ages did not exhibit significant differences in their myogenic and endothelial in vitro differentiation capacity. CONCLUSIONS: Our results suggest that levels of CD133+CXCR4+CD34- could function as a new prognostic clinical marker for the progression of DMD.


Asunto(s)
Antígenos CD/sangre , Glicoproteínas/sangre , Distrofia Muscular de Duchenne/sangre , Péptidos/sangre , Antígeno AC133 , Adolescente , Adulto , Antígenos CD/clasificación , Estudios de Casos y Controles , Niño , Preescolar , Progresión de la Enfermedad , Glicoproteínas/clasificación , Humanos , Distrofia Muscular de Duchenne/inmunología , Análisis de Secuencia por Matrices de Oligonucleótidos , Péptidos/clasificación , Fenotipo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Índice de Severidad de la Enfermedad
11.
Blood ; 108(8): 2857-66, 2006 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-16809613

RESUMEN

Recently our group demonstrated the myogenic capacity of human CD133(+) cells isolated from peripheral blood when delivered in vivo through the arterial circulation into the muscle of dystrophic scid/mdx mice. CD133(+) stem cells express the adhesion molecules CD44, LFA-1, PSGL-1, alpha4-integrins, L-selectin, and chemokine receptor CCR7. Moreover these cells adhere in vitro to VCAM-1 spontaneously and after stimulation with CCL19. Importantly, after muscle exercise, we found that the expression of VCAM-1 is strongly up-regulated in dystrophic muscle vessels, whereas the number of rolling and firmly adhered CD133(+) stem cells significantly increased. Moreover, human dystrophin expression was significantly increased when muscle exercise was performed 24 hours before the intra-arterial injection of human CD133(+) cells. Finally, treatment of exercised dystrophic mice with anti-VCAM-1 antibodies led to a dramatic blockade of CD133(+) stem cell migration into the dystrophic muscle. Our results show for the first time that the expression of VCAM-1 on dystrophic muscle vessels induced by exercise controls muscle homing of human CD133(+) stem cells, opening new perspectives for a potential therapy of muscular dystrophy based on the intra-arterial delivery of CD133(+) stem cells.


Asunto(s)
Músculo Esquelético/irrigación sanguínea , Músculo Esquelético/metabolismo , Distrofia Muscular Animal/metabolismo , Distrofia Muscular Animal/terapia , Molécula 1 de Adhesión Celular Vascular/metabolismo , Antígeno AC133 , Animales , Antígenos CD/metabolismo , Adhesión Celular , Distrofina/metabolismo , Glicoproteínas/metabolismo , Humanos , Técnicas In Vitro , Inyecciones Intraarteriales , Ratones , Ratones Endogámicos mdx , Ratones SCID , Péptidos/metabolismo , Trasplante de Células Madre de Sangre Periférica , Receptores de Quimiocina/metabolismo , Trasplante Heterólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA