Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Pharmacol Res ; 127: 33-40, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28873339

RESUMEN

The adult myocardium, including human, harbours a population of resident multi-potent cardiac stem cells (CSCs), which when stimulated under the right conditions can give rise to new cardiomyocytes and vasculature. Elucidation of the cellular and molecular mechanisms that govern CSC biology and their role in myocardial regeneration will allow the design and development of optimal therapeutic interventions. It is now evident that different growth factors and cytokines govern CSC survival, proliferation, migration and differentiation, as well as playing a role in activating cardiac repair mechanisms such as improving angiogenesis, cardiomyocyte survival and limiting fibrosis. This review article will summarize the evidence for a role of VEGF, NRG-1, IGF-1, HGF, EGF, FGF and TGF-ß1 in modulating the repair and regeneration of cardiac tissue. It will also discuss the use of exosomes and exercise training as interventions to stimulate the endogenous repair and regenerative mechanisms in the damaged heart.


Asunto(s)
Citocinas/fisiología , Corazón/fisiología , Péptidos y Proteínas de Señalización Intercelular/fisiología , Regeneración/fisiología , Células Madre/fisiología , Animales , Ejercicio Físico/fisiología , Exosomas/fisiología , Humanos
2.
Stem Cell Res Ther ; 8(1): 158, 2017 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-28676130

RESUMEN

BACKGROUND: The development of cellular therapies to treat muscle wastage with disease or age is paramount. Resident muscle satellite cells are not currently regarded as a viable cell source due to their limited migration and growth capability ex vivo. This study investigated the potential of muscle-derived PW1+/Pax7- interstitial progenitor cells (PICs) as a source of tissue-specific stem/progenitor cells with stem cell properties and multipotency. METHODS: Sca-1+/PW1+ PICs were identified on tissue sections from hind limb muscle of 21-day-old mice, isolated by magnetic-activated cell sorting (MACS) technology and their phenotype and characteristics assessed over time in culture. Green fluorescent protein (GFP)-labelled PICs were used to determine multipotency in vivo in a tumour formation assay. RESULTS: Isolated PICs expressed markers of pluripotency (Oct3/4, Sox2, and Nanog), were clonogenic, and self-renewing with >60 population doublings, and a population doubling time of 15.8 ± 2.9 h. PICs demonstrated an ability to generate both striated and smooth muscle, whilst also displaying the potential to differentiate into cell types of the three germ layers both in vitro and in vivo. Moreover, PICs did not form tumours in vivo. CONCLUSION: These findings open new avenues for a variety of solid tissue engineering and regeneration approaches, utilising a single multipotent stem cell type isolated from an easily accessible source such as skeletal muscle.


Asunto(s)
Antígenos de Diferenciación/biosíntesis , Proliferación Celular , Regulación de la Expresión Génica , Células Madre Multipotentes/metabolismo , Células Satélite del Músculo Esquelético/metabolismo , Animales , Ratones , Células Madre Multipotentes/citología , Células Satélite del Músculo Esquelético/citología
3.
JACC Basic Transl Sci ; 2(6): 717-736, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30062184

RESUMEN

Skeletal muscle-derived PW1pos/Pax7neg interstitial cells (PICs) express and secrete a multitude of proregenerative growth factors and cytokines. Utilizing a porcine preclinical skeletal muscle injury model, delivery of allogeneic porcine PICs (pPICs) significantly improved and accelerated myofiber regeneration and neocapillarization, compared with saline vehicle control-treated muscles. Allogeneic pPICs did not contribute to new myofibers or capillaries and were eliminated by the host immune system. In conclusion, allogeneic pPIC transplantation stimulated the endogenous stem cell pool to bring about enhanced autologous skeletal muscle repair and regeneration. This allogeneic cell approach is considered a cost-effective, easy to apply, and readily available regenerative therapeutic strategy.

4.
Cell Death Differ ; 24(12): 2101-2116, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28800128

RESUMEN

Multipotent adult resident cardiac stem cells (CSCs) were first identified by the expression of c-kit, the stem cell factor receptor. However, in the adult myocardium c-kit alone cannot distinguish CSCs from other c-kit-expressing (c-kitpos) cells. The adult heart indeed contains a heterogeneous mixture of c-kitpos cells, mainly composed of mast and endothelial/progenitor cells. This heterogeneity of cardiac c-kitpos cells has generated confusion and controversy about the existence and role of CSCs in the adult heart. Here, to unravel CSC identity within the heterogeneous c-kit-expressing cardiac cell population, c-kitpos cardiac cells were separated through CD45-positive or -negative sorting followed by c-kitpos sorting. The blood/endothelial lineage-committed (Lineagepos) CD45posc-kitpos cardiac cells were compared to CD45neg(Lineageneg/Linneg) c-kitpos cardiac cells for stemness and myogenic properties in vitro and in vivo. The majority (~90%) of the resident c-kitpos cardiac cells are blood/endothelial lineage-committed CD45posCD31posc-kitpos cells. In contrast, the LinnegCD45negc-kitpos cardiac cell cohort, which represents ⩽10% of the total c-kitpos cells, contain all the cardiac cells with the properties of adult multipotent CSCs. These characteristics are absent from the c-kitneg and the blood/endothelial lineage-committed c-kitpos cardiac cells. Single Linnegc-kitpos cell-derived clones, which represent only 1-2% of total c-kitpos myocardial cells, when stimulated with TGF-ß/Wnt molecules, acquire full transcriptome and protein expression, sarcomere organisation, spontaneous contraction and electrophysiological properties of differentiated cardiomyocytes (CMs). Genetically tagged cloned progeny of one Linnegc-kitpos cell when injected into the infarcted myocardium, results in significant regeneration of new CMs, arterioles and capillaries, derived from the injected cells. The CSC's myogenic regenerative capacity is dependent on commitment to the CM lineage through activation of the SMAD2 pathway. Such regeneration was not apparent when blood/endothelial lineage-committed c-kitpos cardiac cells were injected. Thus, among the cardiac c-kitpos cell cohort only a very small fraction has the phenotype and the differentiation/regenerative potential characteristics of true multipotent CSCs.


Asunto(s)
Células Madre Adultas/enzimología , Células Madre Multipotentes/enzimología , Miocardio/enzimología , Proteínas Proto-Oncogénicas c-kit/biosíntesis , Células Madre Adultas/citología , Animales , Diferenciación Celular/fisiología , Células Cultivadas , Masculino , Ratones , Células Madre Multipotentes/citología , Miocardio/citología , Ratas , Ratas Wistar
5.
Stem Cells Transl Med ; 3(6): 702-12, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24744394

RESUMEN

Developing effective strategies for the regeneration of solid tissue requires an understanding of the biology underlying the tissue's endogenous repair mechanisms. PW1/Peg3(pos)/Pax7(neg) skeletal muscle-derived interstitial progenitor cells (PICs) were first identified recently in the interstitium of murine skeletal muscle and shown to contribute to muscle fiber regeneration in vivo. PICs, therefore, represent a novel candidate resident progenitor cell for muscle regeneration. To explore the potential of these cells for clinical translation, we must ascertain the presence of PICs in larger mammalian species and identify criteria to successfully isolate and expand this population. In this study, we report the isolation, characterization, and maintenance of multipotent PICs from juvenile porcine skeletal muscle. We show that porcine PICs can be reproducibly isolated from skeletal muscle, express stem/progenitor cell markers, and have a stable phenotype and karyotype through multiple passages. Furthermore, porcine PICs are clonogenic and multipotent, giving rise to skeletal myoblast/myotubes, smooth muscle, and endothelial cells. In addition, PICs can be induced to differentiate into cardiomyocyte-like cells. These results demonstrate, in an animal model with size and physiology extrapolatable to the human, that porcine skeletal muscle-derived PW1(pos)/Pax7(neg) PICs are a source of stem/progenitor cells. These findings open new avenues for a variety of solid tissue engineering and regeneration using a single multipotent stem cell type isolated from an easily accessible source, such as skeletal muscle.


Asunto(s)
Diferenciación Celular , Factores de Transcripción de Tipo Kruppel/metabolismo , Células Madre Multipotentes/metabolismo , Desarrollo de Músculos , Fibras Musculares Esqueléticas/metabolismo , Mioblastos Esqueléticos/metabolismo , Factor de Transcripción PAX7/metabolismo , Medicina Regenerativa/métodos , Adipogénesis , Animales , Antígenos CD34/metabolismo , Biomarcadores/metabolismo , Técnicas de Cultivo de Célula , Proliferación Celular , Separación Celular , Células Cultivadas , Células Endoteliales/metabolismo , Regulación del Desarrollo de la Expresión Génica , Cariotipo , Factores de Transcripción de Tipo Kruppel/genética , Antígenos Comunes de Leucocito/metabolismo , Miocitos Cardíacos/metabolismo , Miocitos del Músculo Liso/metabolismo , Factor de Transcripción PAX7/genética , Fenotipo , Porcinos , Factores de Tiempo
6.
Nat Protoc ; 9(7): 1662-81, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24945383

RESUMEN

This protocol describes the isolation of endogenous c-Kit (also known as CD117)-positive (c-Kit(+)), CD45-negative (CD45(-)) cardiac stem cells (eCSCs) from whole adult mouse and rat hearts. The heart is enzymatically digested via retrograde perfusion of the coronary circulation, resulting in rapid and extensive breakdown of the whole heart. Next, the tissue is mechanically dissociated further and cell fractions are separated by centrifugation. The c-Kit(+)CD45(-) eCSC population is isolated by magnetic-activated cell sorting technology and purity and cell numbers are assessed by flow cytometry. This process takes ∼4 h for mouse eCSCs or 4.5 h for rat eCSCs. We also describe how to characterize c-Kit(+)CD45(-) eCSCs. The c-Kit(+)CD45(-) eCSCs exhibit the defining characteristics of stem cells: they are self-renewing, clonogenic and multipotent. This protocol also describes how to differentiate eCSCs into three main cardiac lineages: functional, beating cardiomyocytes, smooth muscle, and endothelial cells. These processes take 17-20 d.


Asunto(s)
Miocardio/citología , Proteínas Proto-Oncogénicas c-kit/metabolismo , Células Madre/citología , Animales , Técnicas de Cultivo de Célula , Diferenciación Celular , Linaje de la Célula , Células Cultivadas , Ratones , Miocardio/metabolismo , Ratas , Células Madre/metabolismo
7.
J Am Heart Assoc ; 3(2): e000434, 2014 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-24670789

RESUMEN

BACKGROUND: Diabetes mellitus (DM) has multifactorial detrimental effects on myocardial tissue. Recently, carbonic anhydrases (CAs) have been shown to play a major role in diabetic microangiopathy but their role in the diabetic cardiomyopathy is still unknown. METHODS AND RESULTS: We obtained left ventricular samples from patients with DM type 2 (DM-T2) and nondiabetic (NDM) patients with postinfarct heart failure who were undergoing surgical coronary revascularization. Myocardial levels of CA-I and CA-II were 6- and 11-fold higher, respectively, in DM-T2 versus NDM patients. Elevated CA-I expression was mainly localized in the cardiac interstitium and endothelial cells. CA-I induced by high glucose levels hampers endothelial cell permeability and determines endothelial cell apoptosis in vitro. Accordingly, capillary density was significantly lower in the DM-T2 myocardial samples (mean±SE=2152±146 versus 4545±211/mm(2)). On the other hand, CA-II was mainly upregulated in cardiomyocytes. The latter was associated with sodium-hydrogen exchanger-1 hyperphosphorylation, exaggerated myocyte hypertrophy (cross-sectional area 565±34 versus 412±27 µm(2)), and apoptotic death (830±54 versus 470±34 per 10(6) myocytes) in DM-T2 versus NDM patients. CA-II is activated by high glucose levels and directly induces cardiomyocyte hypertrophy and death in vitro, which are prevented by sodium-hydrogen exchanger-1 inhibition. CA-II was shown to be a direct target for repression by microRNA-23b, which was downregulated in myocardial samples from DM-T2 patients. MicroRNA-23b is regulated by p38 mitogen-activated protein kinase, and it modulates high-glucose CA-II-dependent effects on cardiomyocyte survival in vitro. CONCLUSIONS: Myocardial CA activation is significantly elevated in human diabetic ischemic cardiomyopathy. These data may open new avenues for targeted treatment of diabetic heart failure.


Asunto(s)
Anhidrasa Carbónica II/metabolismo , Anhidrasa Carbónica I/metabolismo , Diabetes Mellitus Tipo 2/complicaciones , Cardiomiopatías Diabéticas/enzimología , Células Endoteliales/enzimología , Isquemia Miocárdica/enzimología , Miocitos Cardíacos/enzimología , Remodelación Ventricular , Anciano , Animales , Apoptosis , Glucemia/metabolismo , Anhidrasa Carbónica I/genética , Anhidrasa Carbónica II/genética , Cardiomegalia/enzimología , Cardiomegalia/patología , Proteínas de Transporte de Catión/metabolismo , Células Cultivadas , Cardiomiopatías Diabéticas/patología , Cardiomiopatías Diabéticas/fisiopatología , Células Endoteliales/patología , Activación Enzimática , Femenino , Humanos , Masculino , MicroARNs/metabolismo , Persona de Mediana Edad , Isquemia Miocárdica/patología , Isquemia Miocárdica/fisiopatología , Miocitos Cardíacos/patología , Fosforilación , Ratas , Ratas Wistar , Transducción de Señal , Intercambiador 1 de Sodio-Hidrógeno , Intercambiadores de Sodio-Hidrógeno/metabolismo , Regulación hacia Arriba , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
8.
PLoS One ; 8(1): e53933, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23349768

RESUMEN

BACKGROUND: Cell based therapies are required now to meet the critical care needs of paediatrics and healthy ageing in an increasingly long-lived human population. Repair of compromised tissue by supporting autologous regeneration is a life changing objective uniting the fields of medical science and engineering. Adipose stem cells (adSCs) are a compelling candidate for use in cell based medicine due to their plasticity and residence in numerous tissues. Adipose found in all animals contains a relatively high concentration of stem cells and is easily isolated by a minimally invasive clinical intervention; such as liposuction. METHODS: This study utilised primary rat adipose to validate a novel strategy for selecting adult stem cells. Experiments explored the use of large, very dense cell-specific antibody loaded isolation beads (diameter 5x-10x greater than target cells) which overcome the problem of endocytosis and have proved to be very effective in cell isolation from minimally processed primary tissue. The technique also benefited from pH mediated release, which enabled elution of captured cells using a simple pH shift. RESULTS: Large beads successfully captured and released adSCs from rat adipose, which were characterised using a combination of microscopy, flow cytometry and PCR. The resultant purified cell population retains minimal capture artefact facilitating autologous reperfusion or application in in vitro models. CONCLUSION: Although evidenced here for adSCs, this approach provides a technological advance at a platform level; whereby it can be applied to isolate any cell population for which there is a characterised surface antigen.


Asunto(s)
Tejido Adiposo/citología , Células Madre Adultas/citología , Separación Celular/métodos , Células Madre/citología , Tejido Adiposo/metabolismo , Adulto , Células Madre Adultas/metabolismo , Animales , Recuento de Células , Citometría de Flujo , Humanos , Microscopía Fluorescente , Ratas , Reproducibilidad de los Resultados , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre/metabolismo , Antígenos Thy-1/genética , Antígenos Thy-1/metabolismo
9.
Cell Regen ; 1(1): 6, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-25408869

RESUMEN

BACKGROUND: Human embryonic stem cells (hESCs) represent a tremendous resource for cell therapies and the study of human development; however to maintain their undifferentiated state in vitro they routinely require the use of mouse embryonic fibroblast (MEF) feeder-layers and exogenous protein media supplementation. RESULTS: These well established requirements can be overcome and in this study, it will be demonstrated that phenotypic stability of hESCs can be maintained using a novel, human plasma protein-based hydrogel as an extracellular culture matrix without the use of feeder cell co-culture. hESCs were resuspended in human platelet poor plasma (PPP), which was gelled by the addition of calcium containing DMEM-based hESC culture medium. Phenotypic and genomic expression of the pluripotency markers OCT4, NANOG and SOX2 were measured using immunohistochemistry and qRT-PCR respectively. Typical hESC morphology was demonstrated throughout in vitro culture and both viability and phenotypic stability were maintained throughout extended culture, up to 25 passages. CONCLUSIONS: PPP-derived hydrogel has demonstrated to be an efficacious alternative to MEF co-culture with its hydrophilicity allowing for this substrate to be delivered via minimally invasive procedures in a liquid phase with polymerization ensuing in situ. Together this provides a novel technique for the study of this unique group of stem cells in either 2D or 3D both in vitro and in vivo.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA