Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
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
2.
Stem Cells Transl Med ; 5(4): 451-63, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26956210

RESUMEN

UNLABELLED: Human placental mesenchymal stromal cells (pMSCs) have never been investigated in intrauterine growth restriction (IUGR). We characterized cells isolated from placental membranes and the basal disc of six IUGR and five physiological placentas. Cell viability and proliferation were assessed every 7 days during a 6-week culture. Expression of hematopoietic, stem, endothelial, and mesenchymal markers was evaluated by flow cytometry. We characterized the multipotency of pMSCs and the expression of genes involved in mitochondrial content and function. Cell viability was high in all samples, and proliferation rate was lower in IUGR compared with control cells. All samples presented a starting heterogeneous population, shifting during culture toward homogeneity for mesenchymal markers and occurring earlier in IUGR than in controls. In vitro multipotency of IUGR-derived pMSCs was restricted because their capacity for adipocyte differentiation was increased, whereas their ability to differentiate toward endothelial cell lineage was decreased. Mitochondrial content and function were higher in IUGR pMSCs than controls, possibly indicating a shift from anaerobic to aerobic metabolism, with the loss of the metabolic characteristics that are typical of undifferentiated multipotent cells. SIGNIFICANCE: This study demonstrates that the loss of endothelial differentiation potential and the increase of adipogenic ability are likely to play a significant role in the vicious cycle of abnormal placental development in intrauterine growth restriction (IUGR). This is the first observation of a potential role for placental mesenchymal stromal cells in intrauterine growth restriction, thus leading to new perspectives for the treatment of IUGR.


Asunto(s)
Retardo del Crecimiento Fetal/patología , Células Madre Mesenquimatosas/patología , Células Madre Mesenquimatosas/fisiología , Neovascularización Fisiológica , Placenta/patología , Estudios de Casos y Controles , Diferenciación Celular/genética , Movimiento Celular/genética , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Endotelio Vascular/fisiología , Femenino , Retardo del Crecimiento Fetal/genética , Humanos , Microvasos/fisiología , Neovascularización Fisiológica/genética , Placenta/irrigación sanguínea , Embarazo
3.
Curr Gene Ther ; 15(6): 563-71, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26415573

RESUMEN

Duchenne muscular dystrophy (DMD) is characterized by the loss of a functional dystrophin protein; the muscles of DMD patients progressively degenerate as a result of mechanical stress during contractions, and the condition eventually leads to premature death. By means antisense oligonucleotides (AONs), it is possible to modulate pre-mRNA splicing eliminating mutated exons and restoring dystrophin open reading frame. To overcome the hurdles in using AONs for therapeutic interventions, we exerted engineered human DMD stem cells with a lentivirus, which permanently and efficiently delivered the cloned AONs. Here we describe for the first time the exosome-mediated release of AONs from engineered human DMD CD133+ stem cells allowing the rescue of murine dystrophin expression. Finally, upon release, AONs could be internalized by host cells suggesting a potential role of exosomes acting as vesicular carriers for DMD gene therapy.


Asunto(s)
Distrofina/genética , Terapia Genética/métodos , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia , Células Madre/citología , Animales , Efecto Espectador/fisiología , Células Cultivadas , Distrofina/biosíntesis , Exones/genética , Humanos , Ratones , Ratones SCID , Músculo Esquelético/patología , Oligonucleótidos Antisentido/genética , Empalme del ARN/genética
4.
Biomed Res Int ; 2015: 680615, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25861640

RESUMEN

Duchenne muscular dystrophy (DMD), the most common form of muscular dystrophy, is characterized by muscular wasting caused by dystrophin deficiency that ultimately ends in force reduction and premature death. In addition to primary genetic defect, several mechanisms contribute to DMD pathogenesis. Recently, antioxidant supplementation was shown to be effective in the treatment of multiple diseases including muscular dystrophy. Different mechanisms were hypothesized such as reduced hydroxyl radicals, nuclear factor-κB deactivation, and NO protection from inactivation. Following these promising evidences, we investigated the effect of the administration of a mix of dietary natural polyphenols (ProAbe) on dystrophic mdx mice in terms of muscular architecture and functionality. We observed a reduction of muscle fibrosis deposition and myofiber necrosis together with an amelioration of vascularization. More importantly, the recovery of the morphological features of dystrophic muscle leads to an improvement of the endurance of treated dystrophic mice. Our data confirmed that ProAbe-based diet may represent a strategy to coadjuvate the treatment of DMD.


Asunto(s)
Músculo Esquelético/efectos de los fármacos , Distrofia Muscular Animal/tratamiento farmacológico , Distrofia Muscular de Duchenne/tratamiento farmacológico , Polifenoles/farmacología , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Fibrosis/tratamiento farmacológico , Fibrosis/metabolismo , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/metabolismo , Distrofia Muscular Animal/metabolismo , Distrofia Muscular de Duchenne/metabolismo , Miofibrillas/efectos de los fármacos , Miofibrillas/metabolismo , FN-kappa B/metabolismo , Óxido Nítrico/metabolismo
5.
Cell Transplant ; 24(2): 213-22, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-24268028

RESUMEN

We previously developed a collagen tube filled with autologous skin-derived stem cells (SDSCs) for bridging long rat sciatic nerve gaps. Here we present a case report describing a compassionate use of this graft for repairing the polyinjured motor and sensory nerves of the upper arms of a patient. Preclinical assessment was performed with collagen/SDSC implantation in rats after sectioning the sciatic nerve. For the patient, during the 3-year follow-up period, functional recovery of injured median and ulnar nerves was assessed by pinch gauge test and static two-point discrimination and touch test with monofilaments, along with electrophysiological and MRI examinations. Preclinical experiments in rats revealed rescue of sciatic nerve and no side effects of patient-derived SDSC transplantation (30 and 180 days of treatment). In the patient treatment, motor and sensory functions of the median nerve demonstrated ongoing recovery postimplantation during the follow-up period. The results indicate that the collagen/SDSC artificial nerve graft could be used for surgical repair of larger defects in major lesions of peripheral nerves, increasing patient quality of life by saving the upper arms from amputation.


Asunto(s)
Traumatismo Múltiple/terapia , Traumatismos de los Nervios Periféricos/terapia , Trasplante de Células Madre , Células Madre/citología , Animales , Encéfalo/diagnóstico por imagen , Colágeno/química , Femenino , Humanos , Inseminación Artificial Heteróloga , Masculino , Regeneración Nerviosa , Traumatismos de los Nervios Periféricos/diagnóstico por imagen , Traumatismos de los Nervios Periféricos/patología , Radiografía , Ratas , Ratas Desnudas , Recuperación de la Función , Nervio Ciático/patología , Piel/citología , Trasplante Autólogo , Adulto Joven
6.
PLoS One ; 9(12): e114787, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25493932

RESUMEN

Despite continuous improvements in therapeutic protocols, cancer-related mortality is still one of the main problems facing public health. The main cause of treatment failure is multi-drug resistance (MDR: simultaneous insensitivity to different anti-cancer agents), the underlying molecular and biological mechanisms of which include the activity of ATP binding cassette (ABC) proteins and drug compartmentalisation in cell organelles. We investigated the expression of the main ABC proteins and the role of cytoplasmic vacuoles in the MDR of six hepatocellular carcinoma (HCC) cell lines, and confirmed the accumulation of the yellow anti-cancer drug sunitinib in giant (four lines) and small cytoplasmic vacuoles of lysosomal origin (two lines). ABC expression analyses showed that the main ABC protein harboured by all of the cell lines was PGP, whose expression was not limited to the cell membrane but was also found on lysosomes. MTT assays showed that the cell lines with giant lysosomes were more resistant to sorafenib treatment than those with small lysosomes (p<0.01), and that verapamil incubation can revert this resistance, especially if it is administered after drug pre-incubation. The findings of this study demonstrate the involvement of PGP-positive lysosomes in drug sequestration and MDR in HCC cell lines. The possibility of modulating this mechanism using PGP inhibitors could lead to the development of new targeted strategies to enhance HCC treatment.


Asunto(s)
Carcinoma Hepatocelular/tratamiento farmacológico , Neoplasias Hepáticas/tratamiento farmacológico , Lisosomas/fisiología , Transportadoras de Casetes de Unión a ATP/metabolismo , Western Blotting , Carcinoma Hepatocelular/fisiopatología , Línea Celular Tumoral , Resistencia a Antineoplásicos/fisiología , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Indoles/uso terapéutico , Neoplasias Hepáticas/fisiopatología , Lisosomas/efectos de los fármacos , Pirroles/uso terapéutico , Sunitinib
7.
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
8.
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
9.
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
10.
Int J Nanomedicine ; 7: 3059-67, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22787400

RESUMEN

Recently published reports have described possible cellular therapy approaches to regenerate muscle tissues using arterial route delivery. However, the kinetic of distribution of these migratory stem cells within injected animal muscular dystrophy models is unknown. Using living X-ray computed microtomography, we established that intra-arterially injected stem cells traffic to multiple muscle tissues for several hours until their migration within dystrophic muscles. Injected stem cells express multiple traffic molecules, including VLA-4, LFA-1, CD44, and the chemokine receptor CXCR4, which are likely to direct these cells into dystrophic muscles. In fact, the majority of intra-arterially injected stem cells access the muscle tissues not immediately after the injection, but after several rounds of recirculation. We set up a new, living, 3D-imaging approach, which appears to be an important way to investigate the kinetic of distribution of systemically injected stem cells within dystrophic muscle tissues, thereby providing supportive data for future clinical applications.


Asunto(s)
Distrofias Musculares/cirugía , Trasplante de Células Madre/métodos , Células Madre/citología , Microtomografía por Rayos X/métodos , Antígeno AC133 , Animales , Antígenos CD/sangre , Antígenos CD/química , Células Cultivadas , Dextranos/química , Dextranos/farmacocinética , Modelos Animales de Enfermedad , Extremidades/diagnóstico por imagen , Glicoproteínas/sangre , Glicoproteínas/química , Humanos , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/metabolismo , Nanopartículas de Magnetita/química , Ratones , Ratones SCID , Péptidos/sangre , Péptidos/química , Reacción en Cadena en Tiempo Real de la Polimerasa , Investigación con Células Madre , Células Madre/química , Distribución Tisular
11.
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
12.
Tissue Eng Part B Rev ; 17(1): 1-11, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20846051

RESUMEN

Advances in stem cell research have provided important understanding of the cell biology and offered great promise for developing new strategies for tissue regeneration. Dynamic determination of stem cell migration and distribution in real time is essential for optimizing treatments in preclinical models and designing clinical protocols. Recent developments in the use of nanotechnologies have contributed to advance of the high-resolution in vivo imaging methods, including the positron emission tomography, the single-photon emission computed tomography, the magnetic resonance imaging, and microcomputed tomography. This review examines the use of nanotechnologies for stem cell tracking, the many contrast agents, and detectors that have been proposed and suggest future directions for mouse to human translation of these techniques, for both therapeutic and diagnostic purposes.


Asunto(s)
Rastreo Celular/métodos , Diagnóstico por Imagen/tendencias , Nanotecnología/métodos , Investigación Biomédica Traslacional/tendencias , Animales , Diagnóstico por Imagen/métodos , Humanos , Ratones , Modelos Biológicos , Especificidad de la Especie , Ingeniería de Tejidos/métodos , Ingeniería de Tejidos/tendencias
13.
Int J Mol Sci ; 11(3): 1070-81, 2010 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-20480000

RESUMEN

Advances in stem cell research have provided important understanding of the cell biology and offered great promise for developing new strategies for tissue regeneration. The beneficial effects of stem cell therapy depend also by the development of new approachs for the track of stem cells in living subjects over time after transplantation. Recent developments in the use of nanotechnologies have contributed to advance of the high-resolution in vivo imaging methods, including positron emission tomography (PET), single-photon emission tomography (SPECT), magnetic resonance (MR) imaging, and X-Ray computed microtomography (microCT). This review examines the use of nanotechnologies for stem cell tracking.


Asunto(s)
Rastreo Celular/métodos , Nanotecnología/métodos , Células Madre/citología , Animales , Humanos , Imagen por Resonancia Magnética , Radiografía , Células Madre/diagnóstico por imagen , Tomografía Computarizada de Emisión de Fotón Único
14.
Biomaterials ; 31(20): 5385-96, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20398929

RESUMEN

Ex vivo expansion of hematopoietic stem cells has been explored in the fields of stem cell biology, gene therapy and clinical transplantation. Recently, we demonstrated the existence of a circulating myogenic progenitor expressing the CD133 antigen. The relative inability of circulating CD133+ stem cells to reproduce themselves ex vivo imposes substantial limitations on their use for clinical applications in muscular dystrophies. Here we report that the use of cluster-assembled nanostructured titanium dioxide (ns-TiO(2)) substrates, in combination with cytokine enriched medium, enables high-level expansion of circulating CD133+ stem cells in vitro. Furthermore, we demonstrate that expanded circulating CD133+ stem cells retain their in vitro capacity to differentiate into myogenic cells. The exploitation of cluster-assembled ns-TiO(2) substrates for the expansion of CD133+ stem cells in vitro could therefore make the clinical application of these stem cells for the treatment of muscle diseases practical.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Células Musculares/citología , Nanoestructuras/química , Células Madre/citología , Titanio/química , Titanio/farmacología , Antígeno AC133 , Animales , Antígenos CD/metabolismo , Diferenciación Celular/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Forma de la Célula/efectos de los fármacos , Citocinas/farmacología , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Glicoproteínas/metabolismo , Hematopoyesis/efectos de los fármacos , Humanos , Inmunofenotipificación , Ratones , Microscopía de Fuerza Atómica , Células Musculares/efectos de los fármacos , Células Musculares/metabolismo , Desarrollo de Músculos/efectos de los fármacos , Péptidos/metabolismo , Células Madre/efectos de los fármacos , Células Madre/metabolismo
15.
J Cell Physiol ; 221(3): 526-34, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19688776

RESUMEN

Mutations in the dystrophin gene cause an X-linked genetic disorder: Duchenne muscular dystrophy (DMD). Stem cell therapy is an attractive method to treat DMD because a small number of cells are required to obtain a therapeutic effect. Here, we discussed about multiple types of myogenic stem cells and their possible use to treat DMD. The identification of a stem cell population providing efficient muscle regeneration is critical for the progression of cell therapy for DMD. We speculated that the most promising possibility for the treatment of DMD is a combination of different approaches, such as gene and stem cell therapy.


Asunto(s)
Terapia Genética/métodos , Distrofia Muscular de Duchenne/terapia , Trasplante de Células Madre , Antígeno AC133 , Animales , Antígenos CD/análisis , Glicoproteínas/análisis , Humanos , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/patología , Péptidos/análisis , Pericitos/citología , Células Satélite del Músculo Esquelético/citología , Células Satélite del Músculo Esquelético/trasplante , Células Madre/citología , Células Madre/metabolismo
16.
Mol Ther ; 17(10): 1771-8, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19623164

RESUMEN

In recent years, numerous reports have identified in mouse different sources of myogenic cells distinct from satellite cells that exhibited a variable myogenic potential in vivo. Myogenic stem cells have also been described in humans, although their regenerative potential has rarely been quantified. In this study, we have investigated the myogenic potential of human muscle-derived cells based on the expression of the stem cell marker CD133 as compared to bona fide satellite cells already used in clinical trials. The efficiency of these cells to participate in muscle regeneration and contribute to the renewal of the satellite cell pool, when injected intramuscularly, has been evaluated in the Rag2(-/-) gammaC(-/-) C5(-/-) mouse in which muscle degeneration is induced by cryoinjury. We demonstrate that human muscle-derived CD133+ cells showed a much greater regenerative capacity when compared to human myoblasts. The number of fibers expressing human proteins and the number of human cells in a satellite cell position are all dramatically increased when compared to those observed after injection of human myoblasts. In addition, CD133+/CD34+ cells exhibited a better dispersion in the host muscle when compared to human myoblasts. We propose that muscle-derived CD133+ cells could be an attractive candidate for cellular therapy.


Asunto(s)
Antígenos CD/inmunología , Glicoproteínas/inmunología , Desarrollo de Músculos/fisiología , Músculo Esquelético/citología , Mioblastos/citología , Péptidos/inmunología , Células Madre/citología , Antígeno AC133 , Animales , Diferenciación Celular/fisiología , Células Cultivadas , Humanos , Ratones , Ratones Mutantes , Desarrollo de Músculos/genética , Músculo Esquelético/inmunología , Mioblastos/fisiología , Células Madre/inmunología , Células Madre/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA