Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Front Genet ; 13: 1056114, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36685855

RESUMO

In 2002 we published an article describing a population of vessel-associated progenitors that we termed mesoangioblasts (MABs). During the past decade evidence had accumulated that during muscle development and regeneration things may be more complex than a simple sequence of binary choices (e.g., dorsal vs. ventral somite). LacZ expressing fibroblasts could fuse with unlabelled myoblasts but not among themselves or with other cell types. Bone marrow derived, circulating progenitors were able to participate in muscle regeneration, though in very small percentage. Searching for the embryonic origin of these progenitors, we identified them as originating at least in part from the embryonic aorta and, at later stages, from the microvasculature of skeletal muscle. While continuing to investigate origin and fate of MABs, the fact that they could be expanded in vitro (also from human muscle) and cross the vessel wall, suggested a protocol for the cell therapy of muscular dystrophies. We tested this protocol in mice and dogs before proceeding to the first clinical trial on Duchenne Muscular Dystrophy patients that showed safety but minimal efficacy. In the last years, we have worked to overcome the problem of low engraftment and tried to understand their role as auxiliary myogenic progenitors during development and regeneration.

2.
Development ; 141(9): 1821-34, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24757004

RESUMO

The embryonic endothelium is a known source of hematopoietic stem cells. Moreover, vessel-associated progenitors/stem cells with multilineage mesodermal differentiation potential, such as the 'embryonic mesoangioblasts', originate in vitro from the endothelium. Using a genetic lineage tracing approach, we show that early extra-embryonic endothelium generates, in a narrow time-window and prior to the hemogenic endothelium in the major embryonic arteries, hematopoietic cells that migrate to the embryo proper, and are subsequently found within the mesenchyme. A subpopulation of these cells, distinct from embryonic macrophages, co-expresses mesenchymal and hematopoietic markers. In addition, hemogenic endothelium-derived cells contribute to skeletal and smooth muscle, and to other mesodermal cells in vivo, and display features of embryonic mesoangioblasts in vitro. Therefore, we provide new insights on the distinctive characteristics of the extra-embryonic and embryonic hemogenic endothelium, and we identify the putative in vivo counterpart of embryonic mesoangioblasts, suggesting their identity and developmental ontogeny.


Assuntos
Linhagem da Célula , Hemangioblastos/citologia , Mesoderma/citologia , Animais , Biomarcadores/metabolismo , Caderinas/metabolismo , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Regulação da Expressão Gênica , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Integrases/metabolismo , Macrófagos/citologia , Macrófagos/metabolismo , Mesoderma/embriologia , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Músculo Esquelético/citologia , Músculo Esquelético/embriologia , Músculo Liso/citologia , Músculo Liso/embriologia , Receptores de Complemento 3b/metabolismo , Recombinação Genética/genética
3.
Cell Stem Cell ; 11(4): 471-6, 2012 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-23040476

RESUMO

Reprogramming of somatic cells into neurons provides a new approach toward cell-based therapy of neurodegenerative diseases. A major challenge for the translation of neuronal reprogramming into therapy is whether the adult human brain contains cell populations amenable to direct somatic cell conversion. Here we show that cells from the adult human cerebral cortex expressing pericyte hallmarks can be reprogrammed into neuronal cells by retrovirus-mediated coexpression of the transcription factors Sox2 and Mash1. These induced neuronal cells acquire the ability of repetitive action potential firing and serve as synaptic targets for other neurons, indicating their capability of integrating into neural networks. Genetic fate-mapping in mice expressing an inducible Cre recombinase under the tissue-nonspecific alkaline phosphatase promoter corroborated the pericytic origin of the reprogrammed cells. Our results raise the possibility of functional conversion of endogenous cells in the adult human brain to induced neuronal fates.


Assuntos
Reprogramação Celular , Córtex Cerebral/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Neurais/citologia , Neurogênese , Neurônios/citologia , Pericitos/citologia , Potenciais de Ação , Adulto , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Diferenciação Celular , Células Cultivadas , Humanos , Camundongos , Rede Nervosa , Doenças Neurodegenerativas/terapia , Retroviridae , Fatores de Transcrição SOXB1/metabolismo , Transplante de Células-Tronco , Transmissão Sináptica
4.
Development ; 138(20): 4523-33, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21903674

RESUMO

Mice deficient in α-sarcoglycan (Sgca-null mice) develop progressive muscular dystrophy and serve as a model for human limb girdle muscular dystrophy type 2D. Sgca-null mice suffer a more severe myopathy than that of mdx mice, the model for Duchenne muscular dystrophy. This is the opposite of what is observed in humans and the reason for this is unknown. In an attempt to understand the cellular basis of this severe muscular dystrophy, we isolated clonal populations of myogenic progenitor cells (MPCs), the resident postnatal muscle progenitors of dystrophic and wild-type mice. MPCs from Sgca-null mice generated much smaller clones than MPCs from wild-type or mdx dystrophic mice. Impaired proliferation of Sgca-null myogenic precursors was confirmed by single fiber analysis and this difference correlated with Sgca expression during MPC proliferation. In the absence of dystrophin and associated proteins, which are only expressed after differentiation, SGCA complexes with and stabilizes FGFR1. Deficiency of Sgca leads to an absence of FGFR1 expression at the membrane and impaired MPC proliferation in response to bFGF. The low proliferation rate of Sgca-null MPCs was rescued by transduction with Sgca-expressing lentiviral vectors. When transplanted into dystrophic muscle, Sgca-null MPCs exhibited reduced engraftment. The reduced proliferative ability of Sgca-null MPCs explains, at least in part, the severity of this muscular dystrophy and also why wild-type donor progenitor cells engraft efficiently and consequently ameliorate disease.


Assuntos
Fatores de Crescimento de Fibroblastos/metabolismo , Desenvolvimento Muscular/fisiologia , Distrofia Muscular Animal/metabolismo , Distrofia Muscular Animal/patologia , Mioblastos/citologia , Mioblastos/metabolismo , Sarcoglicanopatias/metabolismo , Sarcoglicanopatias/patologia , Sarcoglicanas/metabolismo , Animais , Sequência de Bases , Linhagem Celular , Proliferação de Células , Primers do DNA/genética , Humanos , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Camundongos Knockout , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/terapia , Mioblastos/transplante , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Sarcoglicanopatias/genética , Sarcoglicanopatias/terapia
5.
J Clin Invest ; 120(1): 11-9, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20051632

RESUMO

Skeletal muscle damaged by injury or by degenerative diseases such as muscular dystrophy is able to regenerate new muscle fibers. Regeneration mainly depends upon satellite cells, myogenic progenitors localized between the basal lamina and the muscle fiber membrane. However, other cell types outside the basal lamina, such as pericytes, also have myogenic potency. Here, we discuss the main properties of satellite cells and other myogenic progenitors as well as recent efforts to obtain myogenic cells from pluripotent stem cells for patient-tailored cell therapy. Clinical trials utilizing these cells to treat muscular dystrophies, heart failure, and stress urinary incontinence are also briefly outlined.


Assuntos
Músculo Esquelético/citologia , Transplante de Células-Tronco , Células-Tronco/fisiologia , Animais , Ensaios Clínicos como Assunto , Insuficiência Cardíaca/terapia , Humanos , Mesoderma/citologia , Distrofias Musculares/etiologia , Distrofias Musculares/terapia , Fator Regulador Miogênico 5/genética , Fator de Transcrição PAX3 , Fator de Transcrição PAX7/genética , Fatores de Transcrição Box Pareados/genética , Regeneração , Incontinência Urinária por Estresse/terapia
6.
Cardiovasc Res ; 83(4): 707-16, 2009 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-19457891

RESUMO

AIMS: Our objective was to test whether progenitor cell proliferation and differentiation potential may vary depending upon the disease of the donor. METHODS AND RESULTS: Human cardiac mesoangioblasts were isolated from cardiac muscle biopsies of patients undergoing open heart surgery for correction of mitral regurgitation following an acute myocardial infarction (MR-MI) or correction of mitral and aortic regurgitation with ensuing left ventricular hypertrophy (MAR-LVH). The cells express surface markers and cardiac genes similar to mouse cardiac mesoangioblasts; they have limited self-renewing and clonogenic activity and are committed mainly to cardiogenesis. Although cardiac differentiation can be induced by 5-azacytidine or by co-culture with rat neonatal cardiomyocytes, human cells do not contract spontaneously like their mouse counterparts. When locally injected in the infarcted myocardium of immunodeficient mice, cardiac mesoangioblasts generate a chimeric heart that contains human myocytes and some capillaries; likewise, they colonize chick embryo hearts when transplanted in ovo. At variance with cells from patients with MR-MI, when isolation was performed on biopsies from MAR-LVH, cells could be isolated in much lower numbers, proliferated less extensively and failed to differentiate. CONCLUSION: Cardiac mesoangioblasts are present in the human heart but this endogenous progenitor population is progressively exhausted, possibly by continuous and inefficient regeneration attempts.


Assuntos
Células-Tronco Adultas/patologia , Cardiomiopatia Hipertrófica/patologia , Miocárdio/patologia , Células-Tronco Adultas/transplante , Animais , Insuficiência da Valva Aórtica/patologia , Diferenciação Celular , Proliferação de Células , Separação Celular , Células Cultivadas , Embrião de Galinha , Humanos , Hipertrofia Ventricular Esquerda/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos SCID , Insuficiência da Valva Mitral/patologia , Infarto do Miocárdio/patologia , Infarto do Miocárdio/terapia , Miócitos Cardíacos/patologia , Ratos , Transplante de Células-Tronco
7.
Nat Cell Biol ; 9(3): 255-67, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17293855

RESUMO

Cells derived from blood vessels of human skeletal muscle can regenerate skeletal muscle, similarly to embryonic mesoangioblasts. However, adult cells do not express endothelial markers, but instead express markers of pericytes, such as NG2 proteoglycan and alkaline phosphatase (ALP), and can be prospectively isolated from freshly dissociated ALP(+) cells. Unlike canonical myogenic precursors (satellite cells), pericyte-derived cells express myogenic markers only in differentiated myotubes, which they form spontaneously with high efficiency. When transplanted into severe combined immune deficient-X-linked, mouse muscular dystrophy (scid-mdx) mice, pericyte-derived cells colonize host muscle and generate numerous fibres expressing human dystrophin. Similar cells isolated from Duchenne patients, and engineered to express human mini-dystrophin, also give rise to many dystrophin-positive fibres in vivo. These data show that myogenic precursors, distinct from satellite cells, are associated with microvascular walls in the human skeletal muscle, may represent a correlate of embryonic 'mesoangioblasts' present after birth and may be a promising candidate for future cell-therapy protocols in patients.


Assuntos
Células-Tronco Adultas/citologia , Músculo Esquelético/citologia , Pericitos/citologia , Regeneração/fisiologia , Células Satélites de Músculo Esquelético/citologia , Adolescente , Adulto , Células-Tronco Adultas/metabolismo , Células-Tronco Adultas/transplante , Idoso , Animais , Antígenos CD/análise , Técnicas de Cultura de Células/métodos , Criança , Pré-Escolar , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos mdx , Camundongos Nus , Camundongos SCID , Pessoa de Meia-Idade , Proteínas Musculares/análise , Proteínas Musculares/genética , Músculo Esquelético/química , Músculo Esquelético/fisiologia , Distrofia Muscular de Duchenne/fisiopatologia , Distrofia Muscular de Duchenne/cirurgia , Pericitos/química , Pericitos/transplante , Células Satélites de Músculo Esquelético/metabolismo , Células Satélites de Músculo Esquelético/transplante , Transplante de Células-Tronco/métodos , Resultado do Tratamento
8.
Curr Protoc Stem Cell Biol ; Chapter 2: Unit 2B.1, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18785178

RESUMO

Mesoangioblasts are recently identified stem/progenitor cells, associated with small vessels of the mesoderm in mammals. Originally described in the mouse embryonic dorsal aorta, similar though not identical cells have been later identified and characterized from postnatal small vessels of skeletal muscle and heart (not described in this unit). They have in common the anatomical location, the expression of endothelial and/or pericyte markers, the ability to proliferate in culture, and the ability to undergo differentiation into various types of mesoderm cells upon proper culture conditions. Currently, the developmental origin of mesoangioblasts, their phenotypic heterogeneity, and the relationship with other mesoderm stem cells are not understood in detail and are the subject of active research. However, from a practical point of view, these cells have been successfully used in cell transplantation protocols that have yielded a significant rescue of structure and function in skeletal muscle of dystrophic mice and dogs. Since the corresponding human cells have been recently isolated and characterized, a clinical trial with these cells is planned in the near future. This unit provides detailed methods for isolation, culture, and characterization of mesoangioblasts.


Assuntos
Células-Tronco Adultas/citologia , Técnicas de Cultura de Células/métodos , Separação Celular/métodos , Células-Tronco Embrionárias/citologia , Animais , Aorta/citologia , Aorta/embriologia , Diferenciação Celular , Cães , Humanos , Mesoderma/irrigação sanguínea , Mesoderma/citologia , Camundongos , Músculo Esquelético/irrigação sanguínea , Músculo Esquelético/citologia , Mioblastos Esqueléticos/citologia , Pericitos/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA