Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Extracell Vesicles ; 10(3): e12045, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33456725

RESUMEN

Extracellular vesicles (EVs) from cardiac stromal cells, developed as therapeutic candidates, improve dystrophic muscle function when administered parenterally, but oral delivery remains untested. We find that casein, the dominant protein in breast milk, enhances the uptake and bioactivity of ingested heart-derived EVs, altering gene expression in blood cells and enhancing muscle function in mdx mice with muscular dystrophy. Thus, EVs, administered orally, are absorbed and exert disease-modifying bioactivity in vivo. Formulating EVs with casein enhances uptake and markedly expands the range of potential therapeutic applications.


Asunto(s)
Caseínas/metabolismo , Vesículas Extracelulares/metabolismo , Distrofias Musculares/terapia , Animales , Ingestión de Alimentos , Femenino , Ratones , Ratones Endogámicos mdx , Leche Humana/metabolismo , Enfermedades Musculares/terapia , Mioblastos Cardíacos/metabolismo , Células del Estroma/metabolismo
3.
JCI Insight ; 4(7)2019 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-30944252

RESUMEN

Dystrophin deficiency leads to progressive muscle degeneration in Duchenne muscular dystrophy (DMD) patients. No known cure exists, and standard care relies on the use of antiinflammatory steroids, which are associated with side effects that complicate long-term use. Here, we report that a single intravenous dose of clinical-stage cardiac stromal cells, called cardiosphere-derived cells (CDCs), improves the dystrophic phenotype in mdx mice. CDCs augment cardiac and skeletal muscle function, partially reverse established heart damage, and boost the regenerative capacity of skeletal muscle. We further demonstrate that CDCs work by secreting exosomes, which normalize gene expression at the transcriptome level, and alter cell signaling and biological processes in mdx hearts and skeletal muscle. The work reported here motivated the ongoing HOPE-2 clinical trial of systemic CDC delivery to DMD patients, and identifies exosomes as next-generation cell-free therapeutic candidates for DMD.


Asunto(s)
Células Madre Adultas/trasplante , Distrofina/metabolismo , Exosomas/metabolismo , Distrofia Muscular de Duchenne/terapia , Miocardio/citología , Células Madre Adultas/metabolismo , Animales , Modelos Animales de Enfermedad , Distrofina/genética , Femenino , Regulación de la Expresión Génica , Humanos , Inyecciones Intravenosas , Ratones , Ratones Endogámicos mdx , Distrofia Muscular de Duchenne/genética , Mutación
4.
Stem Cell Reports ; 10(3): 942-955, 2018 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-29478899

RESUMEN

Genetic deficiency of dystrophin leads to disability and premature death in Duchenne muscular dystrophy (DMD), affecting the heart as well as skeletal muscle. Here, we report that clinical-stage cardiac progenitor cells, known as cardiosphere-derived cells (CDCs), improve cardiac and skeletal myopathy in the mdx mouse model of DMD. Injection of CDCs into the hearts of mdx mice augments cardiac function, ambulatory capacity, and survival. Exosomes secreted by human CDCs reproduce the benefits of CDCs in mdx mice and in human induced pluripotent stem cell-derived Duchenne cardiomyocytes. Surprisingly, CDCs and their exosomes also transiently restored partial expression of full-length dystrophin in mdx mice. The findings further motivate the testing of CDCs in Duchenne patients, while identifying exosomes as next-generation therapeutic candidates.


Asunto(s)
Exosomas/fisiología , Distrofia Muscular de Duchenne/terapia , Animales , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Modelos Animales de Enfermedad , Distrofina/metabolismo , Exosomas/metabolismo , Femenino , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiología , Distrofia Muscular Animal/metabolismo , Distrofia Muscular Animal/terapia , Distrofia Muscular de Duchenne/metabolismo , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/fisiología
5.
J Am Coll Cardiol ; 66(6): 599-611, 2015 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-26248985

RESUMEN

BACKGROUND: Cardiosphere-derived cells mediate therapeutic regeneration in patients after myocardial infarction and are undergoing further clinical testing for cardiomyopathy. The beneficial effects of cardiosphere-derived cells are mediated by the secretion of exosomes and possibly other extracellular membrane vesicles (EMVs). OBJECTIVES: This study sought to investigate the effect of cardiosphere-derived EMVs (CSp-EMVs) on fibroblasts in vitro and tested whether priming with CSp-EMVs could confer salutary properties on fibroblasts in vivo. METHODS: CSp-EMVs were isolated from serum-free media conditioned for 3 days by cardiospheres. Dermal fibroblasts were primed with CSp-EMVs for 24 h followed by exosomal micro-ribonucleic acid profiling. In vivo, we injected CSp-EMV-primed or -unprimed dermal fibroblasts (or CSp-EMVs) in a chronic rat model of myocardial infarction and defined the functional and structural consequences. RESULTS: CSp-EMVs amplified their own biological signals: exposure of "inert" fibroblasts to CSp-EMVs rendered the fibroblasts therapeutic. Intramyocardially injected CSp-EMV-primed (but not unprimed) fibroblasts increased global pump function and vessel density while reducing scar mass. CSp-EMV priming caused fibroblasts to secrete much higher levels of stromal-cell-derived factor 1 and vascular endothelial growth factor and dramatically changed the micro-ribonucleic acid profile of fibroblast-secreted EMVs in vitro. The priming was followed by significant angiogenic and cardioprotective effects. CONCLUSIONS: CSp-EMVs alter fibroblast phenotype and secretome in a salutary positive-feedback loop. The phenotypic conversion of inert cells to therapeutically active cells reveals a novel mechanism for amplification of exosome bioactivity.


Asunto(s)
Albúminas/fisiología , Apoptosis/fisiología , Membrana Celular/fisiología , Fibroblastos/fisiología , Infarto del Miocardio/patología , Neovascularización Fisiológica/fisiología , Albúminas/uso terapéutico , Animales , Membrana Celular/trasplante , Células Cultivadas , Vesículas Cubiertas por Clatrina/fisiología , Vesículas Cubiertas por Clatrina/trasplante , Femenino , Fibroblastos/trasplante , Fibrosis/patología , Fibrosis/terapia , Humanos , Masculino , Infarto del Miocardio/terapia , Poliésteres/uso terapéutico , Ratas , Ratas Endogámicas BN , Ratas Endogámicas WKY , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA