Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
1.
Cell Death Dis ; 12(7): 677, 2021 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-34226515

RESUMEN

Muscular dystrophies are debilitating neuromuscular disorders for which no cure exists. As this disorder affects both cardiac and skeletal muscle, patients would benefit from a cellular therapy that can simultaneously regenerate both tissues. The current protocol to derive bipotent mesodermal progenitors which can differentiate into cardiac and skeletal muscle relies on the spontaneous formation of embryoid bodies, thereby hampering further clinical translation. Additionally, as skeletal muscle is the largest organ in the human body, a high myogenic potential is necessary for successful regeneration. Here, we have optimized a protocol to generate chemically defined human induced pluripotent stem cell-derived mesodermal progenitors (cdMiPs). We demonstrate that these cells contribute to myotube formation and differentiate into cardiomyocytes, both in vitro and in vivo. Furthermore, the addition of valproic acid, a clinically approved small molecule, increases the potential of the cdMiPs to contribute to myotube formation that can be prevented by NOTCH signaling inhibitors. Moreover, valproic acid pre-treated cdMiPs injected in dystrophic muscles increase physical strength and ameliorate the functional performances of transplanted mice. Taken together, these results constitute a novel approach to generate mesodermal progenitors with enhanced myogenic potential using clinically approved reagents.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Mesodermo/efectos de los fármacos , Desarrollo de Músculos/efectos de los fármacos , Fibras Musculares Esqueléticas/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Receptores Notch/metabolismo , Ácido Valproico/farmacología , Animales , Linaje de la Célula , Células Cultivadas , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Femenino , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/trasplante , Masculino , Mesodermo/citología , Mesodermo/metabolismo , Mesodermo/trasplante , Ratones , Ratones Noqueados , Contracción Muscular , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/trasplante , Fuerza Muscular , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiopatología , Distrofias Musculares/genética , Distrofias Musculares/metabolismo , Distrofias Musculares/fisiopatología , Distrofias Musculares/cirugía , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/trasplante , Fenotipo , Ratas , Transducción de Señal
2.
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
3.
Cells ; 8(5)2019 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-31075875

RESUMEN

The skeletal muscle is considered to be an ideal target for stem cell therapy as it has an inherent regenerative capacity. Upon injury, the satellite cells, muscle stem cells that reside under the basal lamina of the myofibres, start to differentiate in order to reconstitute the myofibres while maintaining the initial stem cell pool. In recent years, it has become more and more evident that epigenetic mechanisms such as histon modifications, DNA methylations and microRNA modulations play a pivatol role in this differentiation process. By understanding the mechanisms behind myogenesis, researchers are able to use this knowledge to enhance the differentiation and engraftment potential of different muscle stem cells. Besides manipulation on an epigenetic level, recent advances in the field of genome-engineering allow site-specific modifications in the genome of these stem cells. Combining epigenetic control of the stem cell fate with the ability to site-specifically correct mutations or add genes for further cell control, can increase the use of stem cells as treatment of muscular dystrophies drastically. In this review, we will discuss the advances that have been made in genome-engineering and the epigenetic regulation of muscle stem cells and how this knowledge can help to get stem cell therapy to its full potential.


Asunto(s)
Epigénesis Genética , Desarrollo de Músculos/genética , Células Madre/metabolismo , Diferenciación Celular/genética , Humanos , Modelos Biológicos , Mutación/genética
4.
Theranostics ; 8(10): 2799-2813, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29774076

RESUMEN

Rationale: Pluripotent stem cells (PSCs) are being investigated as a cell source for regenerative medicine since they provide an infinitive pool of cells that are able to differentiate towards every cell type of the body. One possible therapeutic application involves the use of these cells to treat myocardial infarction (MI), a condition where billions of cardiomyocytes (CMs) are lost. Although several protocols have been developed to differentiate PSCs towards CMs, none of these provide a completely pure population, thereby still posing a risk for neoplastic teratoma formation. Therefore, we developed a strategy to (i) monitor cell behavior noninvasively via site-specific integration of firefly luciferase (Fluc) and the human positron emission tomography (PET) imaging reporter genes, sodium iodide symporter (hNIS) and somatostatin receptor type 2 (hSSTr2), and (ii) perform hSSTr2-mediated suicide gene therapy via the clinically used radiopharmacon 177Lu-DOTATATE. Methods: Human embryonic stem cells (ESCs) were gene-edited via zinc finger nucleases to express Fluc and either hNIS or hSSTr2 in the safe harbor locus, adeno-associated virus integration site 1. Firstly, these cells were exposed to 4.8 MBq 177Lu-DOTATATE in vitro and cell survival was monitored via bioluminescence imaging (BLI). Afterwards, hNIS+ and hSSTr2+ ESCs were transplanted subcutaneously and teratomas were allowed to form. At day 59, baseline 124I and 68Ga-DOTATATE PET and BLI scans were performed. The day after, animals received either saline or 55 MBq 177Lu-DOTATATE. Weekly BLI scans were performed, accompanied by 124I and 68Ga-DOTATATE PET scans at days 87 and 88, respectively. Finally, hSSTr2+ ESCs were differentiated towards CMs and transplanted intramyocardially in the border zone of an infarct that was induced by left anterior descending coronary artery ligation. After transplantation, the animals were monitored via BLI and PET, while global cardiac function was evaluated using cardiac magnetic resonance imaging. Results: Teratoma growth of both hNIS+ and hSSTr2+ ESCs could be followed noninvasively over time by both PET and BLI. After 177Lu-DOTATATE administration, successful cell killing of the hSSTr2+ ESCs was achieved both in vitro and in vivo, indicated by reductions in total tracer lesion uptake, BLI signal and teratoma volume. As undifferentiated hSSTr2+ ESCs are not therapeutically relevant, they were differentiated towards CMs and injected in immune-deficient mice with a MI. Long-term cell survival could be monitored without uncontrolled cell proliferation. However, no improvement in the left ventricular ejection fraction was observed. Conclusion: We developed isogenic hSSTr2-expressing ESCs that allow noninvasive cell monitoring in the context of PSC-derived regenerative therapy. Furthermore, we are the first to use the hSSTr2 not only as an imaging reporter gene, but also as a suicide mechanism for radionuclide therapy in the setting of PSC-derived cell treatment.


Asunto(s)
Células Madre Embrionarias Humanas/trasplante , Infarto del Miocardio/terapia , Receptores de Somatostatina/metabolismo , Trasplante de Células Madre/métodos , Teratoma/diagnóstico por imagen , Animales , Línea Celular , Femenino , Genes Reporteros , Células Madre Embrionarias Humanas/citología , Células Madre Embrionarias Humanas/metabolismo , Humanos , Luciferasas/genética , Luciferasas/metabolismo , Ratones , Ratones Desnudos , Infarto del Miocardio/diagnóstico por imagen , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Octreótido/análogos & derivados , Compuestos Organometálicos , Tomografía de Emisión de Positrones , Radiofármacos , Receptores de Somatostatina/genética , Trasplante de Células Madre/efectos adversos , Teratoma/etiología
5.
J Nucl Med ; 58(10): 1659-1665, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28596158

RESUMEN

Molecular imaging is indispensable for determining the fate and persistence of engrafted stem cells. Standard strategies for transgene induction involve the use of viral vectors prone to silencing and insertional mutagenesis or the use of nonhuman genes. Methods: We used zinc finger nucleases to induce stable expression of human imaging reporter genes into the safe-harbor locus adeno-associated virus integration site 1 in human embryonic stem cells. Plasmids were generated carrying reporter genes for fluorescence, bioluminescence imaging, and human PET reporter genes. Results: In vitro assays confirmed their functionality, and embryonic stem cells retained differentiation capacity. Teratoma formation assays were performed, and tumors were imaged over time with PET and bioluminescence imaging. Conclusion: This study demonstrates the application of genome editing for targeted integration of human imaging reporter genes in human embryonic stem cells for long-term molecular imaging.


Asunto(s)
Células Madre Embrionarias/metabolismo , Endorribonucleasas/metabolismo , Edición Génica , Genes Reporteros/genética , Genoma Humano/genética , Tomografía de Emisión de Positrones , Dedos de Zinc , Animales , Diferenciación Celular , Línea Celular , Endorribonucleasas/química , Femenino , Expresión Génica , Humanos , Hígado/citología , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA