Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell Mol Life Sci ; 79(5): 259, 2022 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-35474498

RESUMEN

Delayed wound healing and chronic skin lesions represent a major health problem. Over the past years, growth factors mediated by platelet-rich plasma (PRP) and cell-based therapies were developed as effective and affordable treatment able to improve wound healing capacity. We have advanced existing concepts to develop a highly efficient high-throughput protocol with proven application for the isolation of PRP and pro-angiogenic cells (AngioPRP). This protocol outlines the effectiveness of AngioPRP in promoting the critical healing process including wound closure, re-epithelialization, granulation tissue growth, and blood vessel regeneration. We coupled this effect with normalization of mechanical properties of rescued mouse wounds, which is sustained by a correct arrangement of elastin and collagen fibers. Proteomic analysis of treated wounds demonstrated a fingerprint of AngioPRP based on the up-regulation of detoxification pathway of glutathione metabolism, correlated to a decrease in inflammatory response. Overall, these results have enabled us to provide a framework for how AngioPRP supports wound healing, opening avenues for further clinical advances.


Asunto(s)
Plaquetas , Plasma Rico en Plaquetas , Animales , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Ratones , Plasma Rico en Plaquetas/metabolismo , Proteómica , Cicatrización de Heridas/fisiología
2.
Hum Mol Genet ; 26(19): 3682-3698, 2017 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-28666318

RESUMEN

α-Dystroglycanopathies are a group of muscular dystrophies characterized by α-DG hypoglycosylation and reduced extracellular ligand-binding affinity. Among other genes involved in the α-DG glycosylation process, fukutin related protein (FKRP) gene mutations generate a wide range of pathologies from mild limb girdle muscular dystrophy 2I (LGMD2I), severe congenital muscular dystrophy 1C (MDC1C), to Walker-Warburg Syndrome and Muscle-Eye-Brain disease. FKRP gene encodes for a glycosyltransferase that in vivo transfers a ribitol phosphate group from a CDP -ribitol present in muscles to α-DG, while in vitro it can be secreted as monomer of 60kDa. Consistently, new evidences reported glycosyltransferases in the blood, freely circulating or wrapped within vesicles. Although the physiological function of blood stream glycosyltransferases remains unclear, they are likely released from blood borne or distant cells. Thus, we hypothesized that freely or wrapped FKRP might circulate as an extracellular glycosyltransferase, able to exert a "glycan remodelling" process, even at distal compartments. Interestingly, we firstly demonstrated a successful transduction of MDC1C blood-derived CD133+ cells and FKRP L276IKI mouse derived satellite cells by a lentiviral vector expressing the wild-type of human FKRP gene. Moreover, we showed that LV-FKRP cells were driven to release exosomes carrying FKRP. Similarly, we observed the presence of FKRP positive exosomes in the plasma of FKRP L276IKI mice intramuscularly injected with engineered satellite cells. The distribution of FKRP protein boosted by exosomes determined its restoration within muscle tissues, an overall recovery of α-DG glycosylation and improved muscle strength, suggesting a systemic supply of FKRP protein acting as glycosyltransferase.


Asunto(s)
Distrofia Muscular de Cinturas/genética , Distrofia Muscular de Cinturas/terapia , Proteínas/metabolismo , Animales , Modelos Animales de Enfermedad , Distroglicanos/metabolismo , Exosomas , Glicosilación , Glicosiltransferasas/metabolismo , Humanos , Ratones , Ratones Transgénicos , Músculo Esquelético/metabolismo , Distrofia Muscular de Cinturas/metabolismo , Mioblastos/metabolismo , Pentosiltransferasa , Proteínas/genética , Células Satélite del Músculo Esquelético/trasplante , Transferasas
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.
J Neurosci Res ; 96(9): 1576-1585, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30113722

RESUMEN

Spinocerebellar ataxias (SCAs) are a genetically heterogeneous group of cerebellar degenerative disorders, characterized by progressive gait unsteadiness, hand incoordination, and dysarthria. Ataxia type 1 (SCA1) is caused by the expansion of a CAG trinucleotide repeat in the SCA1 gene resulting in the atypical extension of a polyglutamine (polyQ) tract within the ataxin-1 protein. Our main objective was to investigate the mitochondrial oxidative metabolism in the cerebellum of transgenic SCA1 mice. SCA1 transgenic mice develop clinical features in the early life stages (around 5 weeks of age) presenting pathological cerebellar signs with concomitant progressive Purkinje neuron atrophy and relatively little cell loss; this evidence suggests that the SCA1 phenotype is not the result of cell death per se, but a possible effect of cellular dysfunction that occurs before neuronal demise. We studied the mitochondrial oxidative metabolism in cerebellar cells from both homozygous and heterozygous transgenic SCA1 mice, aged 2 and 6 months. Histochemical examination showed a cytochrome-c-oxidase (COX) deficiency in the Purkinje cells (PCs) of both heterozygous and homozygous mice, the oxidative defect being more prominent in older mice, in which the percentage of COX-deficient PC was up to 30%. Using a laser-microdissector, we evaluated the mitochondrial DNA (mtDNA) content on selectively isolated COX-competent and COX-deficient PC by quantitative Polymerase Chain Reaction and we found mtDNA depletion in those with oxidative dysfunction. In conclusion, the selective oxidative metabolism defect observed in neuronal PC expressing mutant ataxin occurs as early as 8 weeks of age thus representing an early step in the PC degeneration process in SCA1 disease.


Asunto(s)
Deficiencia de Citocromo-c Oxidasa/metabolismo , ADN Mitocondrial/genética , Células de Purkinje/metabolismo , Ataxias Espinocerebelosas/genética , Ataxias Espinocerebelosas/metabolismo , Animales , Ataxina-1/genética , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones Transgénicos , Células de Purkinje/ultraestructura
5.
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
6.
BMC Med Genet ; 17(1): 55, 2016 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-27515321

RESUMEN

BACKGROUND: The dystrophin gene is the one of the largest described in human beings and mutations associated to this gene are responsible for Duchenne or Becker muscular dystrophies. CASE PRESENTATION: Here we describe a nucleotide substitution in the acceptor splice site of intron 26 (c.3604-1G > C) carried by a 6-year-old boy who presented with a history of progressive proximal muscle weakness and elevated serum creatine kinase levels. RNA analysis showed that the first two nucleotides of the mutated intron 26 (AC) were not recognized by the splicing machinery and a new splicing site was created within exon 27, generating a premature stop codon and avoiding protein translation. CONCLUSIONS: The evaluation of the pathogenic effect of the mutation by mRNA analysis will be useful in the optics of an antisense oligonucleotides (AON)-based therapy.


Asunto(s)
Distrofina/genética , Mutación del Sistema de Lectura , Distrofia Muscular de Duchenne/genética , Sitios de Empalme de ARN , Sustitución de Aminoácidos , Niño , Humanos , Intrones , Masculino , Análisis de Secuencia de ARN
7.
J Muscle Res Cell Motil ; 37(3): 101-15, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27443559

RESUMEN

Myofibrillar myopathies (MFMs) are genetically heterogeneous dystrophies characterized by the disintegration of Z-disks and myofibrils and are associated with mutations in genes encoding Z-disk or Z-disk-related proteins. The c.626 C > T (p.P209L) mutation in the BAG3 gene has been described as causative of a subtype of MFM. We report a sporadic case of a 26-year-old Italian woman, affected by MFM with axonal neuropathy, cardiomyopathy, rigid spine, who carries the c.626 C > T mutation in the BAG3 gene. The patient and her non-consanguineous healthy parents and brother were studied with whole exome sequencing (WES) to further investigate the genetic basis of this complex phenotype. In the patient, we found that the BAG3 mutation is associated with variants in the NRAP and FHL1 genes that encode muscle-specific, LIM domain containing proteins. Quantitative real time PCR, immunohistochemistry and Western blot analysis of the patient's muscular biopsy showed the absence of NRAP expression and FHL1 accumulation in aggregates in the affected skeletal muscle tissue. Molecular dynamic analysis of the mutated FHL1 domain showed a modification in its surface charge, which could affect its capability to bind its target proteins. To our knowledge this is the first study reporting, in a BAG3 MFM, the simultaneous presence of genetic variants in the BAG3 and FHL1 genes (previously described as independently associated with MFMs) and linking the NRAP gene to MFM for the first time.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Reguladoras de la Apoptosis/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas con Dominio LIM/genética , Proteínas Musculares/genética , Miopatías Estructurales Congénitas/genética , Adulto , Exoma , Femenino , Humanos , Italia , Transfección
8.
EMBO Mol Med ; 16(4): 927-944, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38438561

RESUMEN

Cell therapy for muscular dystrophy has met with limited success, mainly due to the poor engraftment of donor cells, especially in fibrotic muscle at an advanced stage of the disease. We developed a cell-mediated exon skipping that exploits the multinucleated nature of myofibers to achieve cross-correction of resident, dystrophic nuclei by the U7 small nuclear RNA engineered to skip exon 51 of the dystrophin gene. We observed that co-culture of genetically corrected human DMD myogenic cells (but not of WT cells) with their dystrophic counterparts at a ratio of either 1:10 or 1:30 leads to dystrophin production at a level several folds higher than what predicted by simple dilution. This is due to diffusion of U7 snRNA to neighbouring dystrophic resident nuclei. When transplanted into NSG-mdx-Δ51mice carrying a mutation of exon 51, genetically corrected human myogenic cells produce dystrophin at much higher level than WT cells, well in the therapeutic range, and lead to force recovery even with an engraftment of only 3-5%. This level of dystrophin production is an important step towards clinical efficacy for cell therapy.


Asunto(s)
Distrofina , Distrofia Muscular de Duchenne , Animales , Humanos , Ratones , Modelos Animales de Enfermedad , Distrofina/genética , Exones , Vectores Genéticos , Ratones Endogámicos mdx , Músculos , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia
9.
Cell Rep ; 43(3): 113854, 2024 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-38412099

RESUMEN

The definition of cell metabolic profile is essential to ensure skeletal muscle fiber heterogeneity and to achieve a proper equilibrium between the self-renewal and commitment of satellite stem cells. Heme sustains several biological functions, including processes profoundly implicated with cell metabolism. The skeletal muscle is a significant heme-producing body compartment, but the consequences of impaired heme homeostasis on this tissue have been poorly investigated. Here, we generate a skeletal-muscle-specific feline leukemia virus subgroup C receptor 1a (FLVCR1a) knockout mouse model and show that, by sustaining heme synthesis, FLVCR1a contributes to determine the energy phenotype in skeletal muscle cells and to modulate satellite cell differentiation and muscle regeneration.


Asunto(s)
Proteínas de Transporte de Membrana , Células Satélite del Músculo Esquelético , Ratones , Animales , Proteínas de Transporte de Membrana/metabolismo , Hemo/metabolismo , Ratones Noqueados , Músculo Esquelético/metabolismo , Metabolismo Energético , Células Satélite del Músculo Esquelético/metabolismo , Diferenciación Celular/fisiología
10.
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
11.
Adv Exp Med Biol ; 777: 229-43, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23161086

RESUMEN

Stem cells are used in cell therapy for degenerative disorders. The main advantage of stem cells is that they can replenish their numbers for long periods through cell division and produce a progeny that can differentiate into multiple cell lineages with specific functions. CD133 is a member of a novel family of cell surface glycoproteins. The expression of human CD133 (AC133 antigen) was originally described in the hematopoietic CD34(+) stem cells, but now it becomes more and more evident that CD133 is a marker of stem and progenitor cell populations originating from various tissues and organs. The main objective of this chapter is to describe the potential sources of CD133(+) stem cells that harbor the ability to engraft, proliferate, and differentiate into functional cells. The characterization of such CD133(+) stem cells unlocks new opportunities in the treatment of degenerative diseases such as Duchenne muscular dystrophy.


Asunto(s)
Antígenos CD34 , Células Madre Hematopoyéticas , Antígenos CD34/biosíntesis , Biomarcadores/metabolismo , Linaje de la Célula , Células Madre Hematopoyéticas/citología , Humanos
12.
Autophagy ; 17(9): 2494-2510, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33030392

RESUMEN

Dominant de novo mutations in the co-chaperone BAG3 cause a severe form of myofibrillar myopathy, exhibiting progressive muscle weakness, muscle structural failure, and protein aggregation. To elucidate the mechanism of disease in, and identify therapies for, BAG3 myofibrillar myopathy, we generated two zebrafish models, one conditionally expressing BAG3P209L and one with a nonsense mutation in bag3. While transgenic BAG3P209L-expressing fish display protein aggregation, modeling the early phase of the disease, bag3-/- fish exhibit exercise dependent fiber disintegration, and reduced swimming activity, consistent with later stages of the disease. Detailed characterization of the bag3-/- fish, revealed an impairment in macroautophagic/autophagic activity, a defect we confirmed in BAG3 patient samples. Taken together, our data highlights that while BAG3P209L expression is sufficient to promote protein aggregation, it is the loss of BAG3 due to its sequestration within aggregates, which results in impaired autophagic activity, and subsequent muscle weakness. We therefore screened autophagy-promoting compounds for their effectiveness at removing protein aggregates, identifying nine including metformin. Further evaluation demonstrated metformin is not only able to bring about the removal of protein aggregates in zebrafish and human myoblasts but is also able to rescue the fiber disintegration and swimming deficit observed in the bag3-/- fish. Therefore, repurposing metformin provides a promising therapy for BAG3 myopathy.Abbreviations:ACTN: actinin, alpha; BAG3: BAG cochaperone 3; CRYAB: crystallin alpha B; DES: desmin; DMSO: dimethyl sulfoxide; DNAJB6: DnaJ heat shock protein family (Hsp40) member B6; dpf: days post fertilization; eGFP: enhanced green fluorescent protein; FDA: Food and Drug Administration; FHL1: four and a half LIM domains 1; FLNC: filamin C; hpf: hours post-fertilization; HSPB8: heat shock protein family B [small] member 8; LDB3/ZASP: LIM domain binding 3; MYOT: myotilin; TTN: titin; WT: wild-type.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Proteínas Reguladoras de la Apoptosis , Metformina , Miopatías Estructurales Congénitas , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Autofagia , Proteínas del Choque Térmico HSP40/genética , Proteínas del Choque Térmico HSP40/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular , Proteínas con Dominio LIM , Metformina/farmacología , Chaperonas Moleculares/metabolismo , Proteínas Musculares , Músculos/metabolismo , Mutación , Miopatías Estructurales Congénitas/genética , Proteínas del Tejido Nervioso/metabolismo , Pez Cebra/metabolismo , Proteínas de Pez Cebra
13.
Am J Pathol ; 175(3): 1200-7, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19644013

RESUMEN

Parathyroid tissue is able to spontaneously induce angiogenesis, proliferate, and secrete parathyroid hormone when autotransplanted in patients undergoing total parathyroidectomy. Angiogenesis is also involved in parathyroid tumorigenesis. Here we investigated the anatomical and molecular relationship between endothelial and parathyroid cells within human parathyroid glands. Immunohistochemistry for CD34 antigen identified two subpopulations in normal and tumoral parathyroid glands: one constituted by cells lining small vessels that displayed endothelial antigens (factor VIII, isolectin, laminin, CD146) and the other constituted of single cells scattered throughout the parenchyma that did not express endothelial markers. These parathyroid-derived CD34(+) cells were negative for the hematopoietic and mesenchymal markers CD45, Thy-1/CD90, CD105, and CD117/c-kit; however, a subset of CD34(+) cells co-expressed the parathyroid specific genes glial cell missing B, parathyroid hormone, and calcium sensing receptor. When cultured, these cells released significant amount of parathyroid hormone. Parathyroid-derived CD34(+) cells, but not CD34(-) cells, proliferated slowly and differentiated into mature endothelial cells. CD34(+) cells from parathyroid tumors differed from those derived from normal parathyroid glands as: 1) they were more abundant and mainly scattered throughout the parenchyma; 2) they rarely co-expressed CD146; and 3) a fraction co-expressed nestin. In conclusion, we identified cells expressing endothelial and parathyroid markers in human adult parathyroid glands. These parathyroid/endothelial cells were more abundant and less committed in parathyroid tumors compared with normal glands, showing features of endothelial progenitors, which suggests that they might be involved in parathyroid tumorigenesis.


Asunto(s)
Antígenos CD34 , Biomarcadores , Diferenciación Celular , Glándulas Paratiroides/citología , Glándulas Paratiroides/metabolismo , Neoplasias de las Paratiroides/patología , Transformación Celular Neoplásica , Células Cultivadas , Células Endoteliales/citología , Células Endoteliales/metabolismo , Hematopoyesis , Humanos , Proteínas de Filamentos Intermediarios/metabolismo , Neovascularización Patológica , Proteínas del Tejido Nervioso/metabolismo , Nestina , Glándulas Paratiroides/irrigación sanguínea , Hormona Paratiroidea/metabolismo , Neoplasias de las Paratiroides/irrigación sanguínea , Neoplasias de las Paratiroides/metabolismo , Receptores Sensibles al Calcio/metabolismo , Células Madre/citología , Células Madre/metabolismo
14.
Cell Rep ; 31(5): 107597, 2020 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-32375047

RESUMEN

Fibrosis and fat replacement in skeletal muscle are major complications that lead to a loss of mobility in chronic muscle disorders, such as muscular dystrophy. However, the in vivo properties of adipogenic stem and precursor cells remain unclear, mainly due to the high cell heterogeneity in skeletal muscles. Here, we use single-cell RNA sequencing to decomplexify interstitial cell populations in healthy and dystrophic skeletal muscles. We identify an interstitial CD142-positive cell population in mice and humans that is responsible for the inhibition of adipogenesis through GDF10 secretion. Furthermore, we show that the interstitial cell composition is completely altered in muscular dystrophy, with a near absence of CD142-positive cells. The identification of these adipo-regulatory cells in the skeletal muscle aids our understanding of the aberrant fat deposition in muscular dystrophy, paving the way for treatments that could counteract degeneration in patients with muscular dystrophy.


Asunto(s)
Adipogénesis/fisiología , Diferenciación Celular/fisiología , Células Intersticiales del Testículo/citología , Músculo Esquelético/patología , Distrofia Muscular de Duchenne/patología , Animales , Fibrosis/metabolismo , Fibrosis/patología , Humanos , Masculino , Ratones , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/metabolismo
15.
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
16.
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
17.
Front Neurol ; 10: 755, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31396142

RESUMEN

Background: Nutritional compounds can exert both anti-inflammatory and anti-oxidant effects. Since these events exacerbate the pathophysiology of muscular dystrophies, we investigated nutraceutical supplementation as an adjuvant therapy in dystrophic patients, to low costs and easy route of administration. Moreover, this treatment could represent an alternative therapeutic strategy for dystrophic patients who do not respond to corticosteroid treatment. Objective: A 24 weeks randomized double-blind placebo-controlled clinical study was aimed at evaluating the safety and efficacy of daily oral administration of flavonoids- and omega3-based natural supplement (FLAVOMEGA) in patients affected by muscular dystrophy with recognized muscle inflammation. Design: We screened 60 patients diagnosed for Duchenne (DMD), Facioscapulohumeral (FSHD), and Limb Girdle Muscular Dystrophy (LGMD). Using a computer-generated random allocation sequence, we stratified patients in a 2:1:1 ratio (DMD:FSHD:LGMD) to one of two treatment groups: continuous FLAVOMEGA, continuous placebo. Of 29 patients included, only 24 completed the study: 15 were given FLAVOMEGA, 14 placebo. Results: FLAVOMEGA was well tolerated with no reported adverse events. Significant treatment differences in the change from baseline in 6 min walk distance (6MWD; secondary efficacy endpoint) (P = 0.033) and in isokinetic knee extension (P = 0.039) (primary efficacy endpoint) were observed in LGMD and FSHD subjects. Serum CK levels (secondary efficacy endpoint) decreased in all FLAVOMEGA treated groups with significant difference in DMD subjects (P = 0.039). Conclusions: Although the small number of patients and the wide range of disease severity among patients reduced statistical significance, we obtained an optimal profile of safety and tolerability for the compound, showing valuable data of efficacy in primary and secondary endpoints. Trial registration number: NCT03317171 Retrospectively registered 25/10/2017.

18.
Sci Rep ; 8(1): 14659, 2018 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-30279586

RESUMEN

Duchenne muscular dystrophy (DMD) is one of the most common and severe forms of muscular dystrophy. Oxidative myofibre content, muscle vasculature architecture and exercise tolerance are impaired in DMD. Several studies have demonstrated that nutrient supplements ameliorate dystrophic features, thereby enhancing muscle performance. Here, we report that dietary supplementation with a specific branched-chain amino acid-enriched mixture (BCAAem) increased the abundance of oxidative muscle fibres associated with increased muscle endurance in dystrophic mdx mice. Amelioration of the fatigue index in BCAAem-treated mdx mice was caused by a cascade of events in the muscle tissue, which were promoted by endothelial nitric oxide synthase (eNOS) activation and vascular endothelial growth factor (VEGF) expression. VEGF induction led to recruitment of bone marrow (BM)-derived endothelial progenitors (EPs), which increased the capillary density of dystrophic skeletal muscle. Functionally, BCAAem mitigated the dystrophic phenotype of mdx mice without inducing dystrophin protein expression or replacing the dystrophin-associated glycoprotein (DAG) complex in the membrane, which is typically lost in DMD. BCAAem supplementation could be an effective adjuvant strategy in DMD treatment.


Asunto(s)
Aminoácidos/administración & dosificación , Suplementos Dietéticos , Distrofia Muscular de Duchenne/dietoterapia , Animales , Modelos Animales de Enfermedad , Células Progenitoras Endoteliales/efectos de los fármacos , Células Progenitoras Endoteliales/metabolismo , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Ratones Noqueados , Fibras Musculares Esqueléticas/efectos de los fármacos , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patología , Fuerza Muscular/efectos de los fármacos , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/patología , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo III/metabolismo , Estrés Oxidativo/efectos de los fármacos , Resistencia Física/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/metabolismo
19.
J Clin Invest ; 114(2): 182-95, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15254585

RESUMEN

Duchenne muscular dystrophy (DMD) is a common X-linked disease characterized by widespread muscle damage that invariably leads to paralysis and death. There is currently no therapy for this disease. Here we report that a subpopulation of circulating cells expressing AC133, a well-characterized marker of hematopoietic stem cells, also expresses early myogenic markers. Freshly isolated, circulating AC133(+) cells were induced to undergo myogenesis when cocultured with myogenic cells or exposed to Wnt-producing cells in vitro and when delivered in vivo through the arterial circulation or directly into the muscles of transgenic scid/mdx mice (which allow survival of human cells). Injected cells also localized under the basal lamina of host muscle fibers and expressed satellite cell markers such as M-cadherin and MYF5. Furthermore, functional tests of injected muscles revealed a substantial recovery of force after treatment. As these cells can be isolated from the blood, manipulated in vitro, and delivered through the circulation, they represent a possible tool for future cell therapy applications in DMD disease or other muscular dystrophies.


Asunto(s)
Distrofina/metabolismo , Glicoproteínas/metabolismo , Células Madre Hematopoyéticas/fisiología , Músculo Esquelético/fisiología , Músculo Esquelético/fisiopatología , Distrofia Muscular de Duchenne/metabolismo , Péptidos/metabolismo , Antígeno AC133 , Adolescente , Adulto , Animales , Antígenos CD , Biomarcadores , Diferenciación Celular/fisiología , Trasplante de Células , Células Cultivadas , Niño , Preescolar , Técnicas de Cocultivo , Distrofina/genética , Células Madre Hematopoyéticas/citología , Humanos , Ratones , Ratones Endogámicos mdx , Ratones SCID , Ratones Transgénicos , Músculo Esquelético/citología , Músculo Esquelético/patología , Distrofia Muscular de Duchenne/patología , Distrofia Muscular de Duchenne/fisiopatología , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Células Satélite del Músculo Esquelético/citología , Células Satélite del Músculo Esquelético/fisiología , Transducción de Señal/fisiología , Proteínas Wnt
20.
Cell Transplant ; 16(1): 41-55, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17436854

RESUMEN

In animal models of neurological disorders for cerebral ischemia, Parkinson's disease, and spinal cord lesions, transplantation of mesenchymal stem cells (MSCs) has been reported to improve functional outcome. Three mechanisms have been suggested for the effects of the MSCs: transdifferentiation of the grafted cells with replacement of degenerating neural cells, cell fusion, and neuroprotection of the dying cells. Here we demonstrate that a restricted number of cells with differentiated astroglial features can be obtained from human adult MSCs (hMSCs) both in vitro using different induction protocols and in vivo after transplantation into the developing mouse brain. We then examined the in vitro differentiation capacity of the hMSCs in coculture with slices of neonatal brain cortex. In this condition the hMSCs did not show any neuronal transdifferentiation but expressed neurotrophin low-affinity (NGFR(p75)) and high-affinity (trkC) receptors and released nerve growth factor (NGF) and neurotrophin-3 (NT-3). The same neurotrophin's expression was demonstrated 45 days after the intracerebral transplantation of hMSCs into nude mice with surviving astroglial cells. These data further confirm the limited capability of adult hMSC to differentiate into neurons whereas they differentiated in astroglial cells. Moreover, the secretion of neurotrophic factors combined with activation of the specific receptors of transplanted hMSCs demonstrated an alternative mechanism for neuroprotection of degenerating neurons. hMSCs are further defined in their transplantation potential for treating neurological disorders.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Factores de Crecimiento Nervioso/metabolismo , Enfermedades Neurodegenerativas/terapia , Adulto , Animales , Encéfalo/cirugía , Diferenciación Celular , Técnica del Anticuerpo Fluorescente , Humanos , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/ultraestructura , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neurotrofina 3/metabolismo , Técnicas de Cultivo de Órganos , Trasplante Heterólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA