Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
J Mol Cell Cardiol ; 48(6): 1215-24, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20064522

RESUMEN

The therapeutic effects of mesenchymal stem cells (MSCs) transplantation are increasingly thought to be mediated by MSC secretion. We have previously demonstrated that human ESC-derived MSCs (hESC-MSCs) produce cardioprotective microparticles in pig model of myocardial ischemia/reperfusion (MI/R) injury. As the safety and availability of clinical grade human ESCs remain a concern, MSCs from fetal tissue sources were evaluated as alternatives. Here we derived five MSC cultures from limb, kidney and liver tissues of three first trimester aborted fetuses and like our previously described hESC-derived MSCs; they were highly expandable and had similar telomerase activities. Each line has the potential to generate at least 10(16-19) cells or 10(7-10) doses of cardioprotective secretion for a pig model of MI/R injury. Unlike previously described fetal MSCs, they did not express pluripotency-associated markers such as Oct4, Nanog or Tra1-60. They displayed a typical MSC surface antigen profile and differentiated into adipocytes, osteocytes and chondrocytes in vitro. Global gene expression analysis by microarray and qRT-PCR revealed a typical MSC gene expression profile that was highly correlated among the five fetal MSC cultures and with that of hESC-MSCs (r(2)>0.90). Like hESC-MSCs, they produced secretion that was cardioprotective in a mouse model of MI/R injury. HPLC analysis of the secretion revealed the presence of a population of microparticles with a hydrodynamic radius of 50-65 nm. This purified population of microparticles was cardioprotective at approximately 1/10 dosage of the crude secretion.


Asunto(s)
Técnicas de Cultivo de Célula , Células Madre Embrionarias/citología , Células Madre Mesenquimatosas/citología , Animales , Antígenos/química , Diferenciación Celular , Cromatografía Líquida de Alta Presión/métodos , Perfilación de la Expresión Génica , Humanos , Células Madre Mesenquimatosas/metabolismo , Ratones , Microesferas , Análisis de Secuencia por Matrices de Oligonucleótidos , Tamaño de la Partícula , Daño por Reperfusión/patología , Porcinos , Distribución Tisular
2.
Arterioscler Thromb Vasc Biol ; 24(4): 691-6, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-14764422

RESUMEN

OBJECTIVE: Endothelial differentiation is a fundamental process in angiogenesis and vasculogenesis with implications in development, normal physiology, and pathology. To better understand this process, an in vitro cellular system that recapitulates endothelial differentiation and is amenable to experimental manipulations is required. METHODS AND RESULTS: Embryonic cell lines that differentiate exclusively into endothelial cells were derived from early mouse embryos using empirical but reproducible culture techniques without viral or chemical transformation. The cells were not pluripotent and expressed reduced levels of Oct 4 and Rex-1. They were non-tumorigenic with a population doubling time of approximately 15 hours. When plated on matrigel, they readily differentiated to form patent tubular structures with diameters of 30 to 150 microm. The differentiated cells endocytosed acetylated low-density lipoprotein (LDL) and began to express endothelial-specific markers such as CD34, CD31, Flk-1, TIE2, P-selectin, Sca-1, and thy-1. They also expressed genes essential for differentiation and maintenance of endothelial lineages, eg, Flk-1, vascular endothelial growth factor (VEGF), and angiopoietin-1. When transplanted into animal models, these cells incorporated into host vasculature. CONCLUSIONS: These cell lines can undergo in vitro and in vivo endothelial differentiation that recapitulated known endothelial differentiation pathways. Therefore, they are ideal for establishing an in vitro cellular system to study endothelial differentiation.


Asunto(s)
Embrión de Mamíferos/citología , Células Endoteliales/citología , Endotelio Vascular/citología , Factores de Transcripción , Animales , Antígenos de Diferenciación/análisis , Biomarcadores/análisis , Blastocisto/citología , Técnicas de Cultivo de Célula/métodos , Diferenciación Celular , Línea Celular , Linaje de la Célula , Células Cultivadas/citología , Células Clonales/citología , Células Clonales/metabolismo , Proteínas de Unión al ADN/análisis , Endocitosis , Células Endoteliales/metabolismo , Genes RAG-1 , Lipoproteínas LDL/metabolismo , Hígado/irrigación sanguínea , Hígado/lesiones , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Trasplante de Neoplasias , Neovascularización Patológica/patología , Neovascularización Fisiológica , Factor 3 de Transcripción de Unión a Octámeros , Teratoma/irrigación sanguínea , beta-Galactosidasa/análisis
3.
Biofactors ; 23(2): 69-73, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16179748

RESUMEN

Homocysteine, cytokines (IL-18, IL-6, IL-8) are involved in vascular inflammation and coronary artery disease. Homocysteine influences endothelial IL-6 and IL-8 cytokine expression and release, however, an association between homocysteine and IL-18 has not been previously investigated in endothelial/smooth muscle cells and or in coronary artery disease. We report in 9 coronary artery bypass surgery (CABG) patients a positive correlation r = 0.86 between homocysteine and IL-18 plasma levels (p < 0.05). Plasma IL-18 levels are significantly higher in those patients with elevated homocysteine compared to those with normal levels (p < 0.02; 153 +/- 19 pg/ml versus 116 +/- 14 pg/ml respectively). Our in vitro cell culture studies suggest that the source of IL-18 in CABG patients with elevated homocysteine is not from vascular smooth muscle or endothelial cells.


Asunto(s)
Puente de Arteria Coronaria , Enfermedad de la Arteria Coronaria/sangre , Homocisteína/sangre , Interleucina-18/sangre , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Masculino , Persona de Mediana Edad
4.
FEBS Lett ; 546(2-3): 237-40, 2003 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-12832047

RESUMEN

Dyslipidemia, inflammation and gender are major risk factors in cardiovascular disease. Here we show that hepatic expression of Peroxisome proliferator-activated receptor alpha (PPARalpha), a nuclear receptor that regulates lipid metabolism and inflammation, is regulated in a gender-specific manner during lipopolysaccharide (LPS)-induced systemic inflammation. Immediately following LPS-induced systemic inflammation, hepatic PPARalpha mRNA level decreased dramatically in mice. It was restored to baseline within 24 h in females but remained below baseline for >72 h in male mice. In gonadectomized mice of both sexes, PPARalpha mRNA level was restored to baseline within 48 h after the initial decrease.


Asunto(s)
Hipolipemiantes/farmacología , Inflamación/fisiopatología , Hígado/metabolismo , Receptores Citoplasmáticos y Nucleares/genética , Factores Sexuales , Factores de Transcripción/genética , Animales , Secuencia de Bases , Cartilla de ADN , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Lipopolisacáridos/farmacología , Masculino , Ratones , ARN Mensajero/genética , ARN Mensajero/metabolismo
5.
Cardiovasc Pathol ; 13(2): 91-7, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15033158

RESUMEN

INTRODUCTION: The laboratory mouse is a powerful tool in cardiovascular research. In this report, we describe a method for a reproducible mouse myocardial infarction model that would allow subsequent comparative and quantitative studies on molecular and pathophysiological variables. METHODS: (A) The distribution of the major coronary arteries including the septal artery in the left ventricle of the C57BL/6J mice (n=20) was mapped by perfusion of latex dye or fluorescent beads through the aorta. (B) The territory of myocardial infarction after the ligation of the most proximal aspect of the left anterior descending (LAD) coronary artery was quantified. (C) The consistency in the histological changes parallel to the infarction at different time points was analyzed. RESULTS: (A) The coronary artery tree of the mouse is different from human and, particularly, in regard to the blood supply of the septum. (B) Contrary to previous belief, the septal coronary artery in the mouse is variable in origin. (C) A constant ligation of the LAD immediately below the left auricular level ensures a statistically significant reproducible infarct size. (D) The ischemic changes can be monitored at a histological level in a way similar to what is described in the human. CONCLUSION: We illustrate a method for maximal reproducibility of experimental acute myocardial infarction in the mouse model, due to a consistent loss of perfusion in the lower half of the left ventricle. This will allow the study of molecular and physiological variables in a controlled and quantifiable experimental model environment.


Asunto(s)
Vasos Coronarios/anatomía & histología , Modelos Animales de Enfermedad , Infarto del Miocardio/patología , Animales , Vasos Coronarios/lesiones , Ligadura/métodos , Ratones , Ratones Endogámicos C57BL , Reproducibilidad de los Resultados
6.
Redox Rep ; 7(4): 215-8, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12396666

RESUMEN

Several recent studies have shown that purified subsets of bone marrow (BM) cells can differentiate into endothelial, cardiac, and other cell types. During coronary artery bypass graft (CABG) surgery, sternal BM is routinely discarded. To determine if this BM can be used to induce angiogenesis and augment perfusion of the cardiac tissues after CABG, a simplified and more practical approach of using whole BM extract was tried to determine whether it would be adequate for the induction of BM-derived angiogenesis in experimental acute limb ischemia. BM was prepared from FVB/N-TgN(TIE2 lacZ)182 Sato (Tie2-lacZ) or B6.129S7-Gtrosa 26 (Rosa 26) mice that express beta-galactosidase (beta-gal) in endothelial cells and most adult tissues, respectively. Acute limb ischemia was induced in either C57BL6/J or FVB/N mice by double ligation of the left femoral artery just distal to the profunda femoral artery branch. Occlusion of the ligated artery was verified by angiography. The study group (n = 31) received an intramuscular injection of 50 micro l containing 1 x 10(6) BM cells, 5 mm proximal to the site of ligation. Experimental controls (n = 21) had an intramuscular injection of 50 micro l of saline. Angiogenesis in the mice was assessed by histological analysis. BM-derived beta-gal(+) cells were observed to aggregate in the vicinity of the ligated artery and not in the injected musculature BM-derived endothelial cells were incorporated within capillaries and small size blood vessels near the site of ligation. Generation of BM-derived blood vessels in experimental acute limb ischemia does not require purification of specific subset of cells. The elimination of cell purification will enhance the ease of using BM transplantation in generating blood vessels.


Asunto(s)
Trasplante de Médula Ósea , Isquemia/fisiopatología , Neovascularización Fisiológica , Enfermedad Aguda , Animales , Células de la Médula Ósea/fisiología , Diferenciación Celular , Ratones , Ratones Endogámicos C57BL
7.
In Vitro Cell Dev Biol Anim ; 40(5-6): 143-9, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15479118

RESUMEN

We have previously isolated mouse embryonic cell lines with endothelial potential using a simple empirical approach. In an attempt to isolate similar cell lines from adult mouse bone marrow (BM), BM cells were cultured on mitotically inactive mouse embryonic fibroblast (MEF) feeder cells. Several cell lines with putative endothelial potential were generated. They expressed endothelial-specific genes and formed vascular-like structures when plated on matrigel. When transplanted into appropriate mouse models, they incorporated into the endothelium of the vasculature. Similar cell lines were also obtained using human or porcine BM. None of these lines induced tumor formation when transplanted into immunodeficient Rag1-/- mice. However, all the lines were aneuploid with genetic markers from BM samples and the MEF feeder, suggesting that they resulted from a non-species-specific fusion of a BM cell and mitotically inactive MEF. Together, these lines demonstrated for the first time that BM cells can also undergo fusion with commonly used mitotically inactive feeder cells. Although these fusion cell lines were culture artifacts, their derivation would be useful in understanding fusion of BM cells with other cell types, and their endothelial potential will also be useful in characterizing endothelial differentiation.


Asunto(s)
Células de la Médula Ósea/citología , Células Endoteliales/citología , Endotelio Vascular/citología , Fibroblastos/citología , Células Híbridas/citología , Aneuploidia , Animales , Biomarcadores/análisis , Fusión Celular , Línea Celular , Línea Celular Transformada/citología , Línea Celular Transformada/metabolismo , Colágeno , Combinación de Medicamentos , Embrión de Mamíferos/citología , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Humanos , Laminina , Hígado/irrigación sanguínea , Ratones , Ratones Endogámicos C57BL , Mitomicina/farmacología , Infarto del Miocardio , Neovascularización Fisiológica/fisiología , Reacción en Cadena de la Polimerasa/métodos , Proteoglicanos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Porcinos
9.
Stem Cell Res ; 4(3): 214-22, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20138817

RESUMEN

Human ESC-derived mesenchymal stem cell (MSC)-conditioned medium (CM) was previously shown to mediate cardioprotection during myocardial ischemia/reperfusion injury through large complexes of 50-100 nm. Here we show that these MSCs secreted 50- to 100-nm particles. These particles could be visualized by electron microscopy and were shown to be phospholipid vesicles consisting of cholesterol, sphingomyelin, and phosphatidylcholine. They contained coimmunoprecipitating exosome-associated proteins, e.g., CD81, CD9, and Alix. These particles were purified as a homogeneous population of particles with a hydrodynamic radius of 55-65 nm by size-exclusion fractionation on a HPLC. Together these observations indicated that these particles are exosomes. These purified exosomes reduced infarct size in a mouse model of myocardial ischemia/reperfusion injury. Therefore, MSC mediated its cardioprotective paracrine effect by secreting exosomes. This novel role of exosomes highlights a new perspective into intercellular mediation of tissue injury and repair, and engenders novel approaches to the development of biologics for tissue repair.


Asunto(s)
Exosomas/metabolismo , Células Madre Mesenquimatosas/metabolismo , Isquemia Miocárdica/terapia , Daño por Reperfusión/terapia , Animales , Antígenos CD/metabolismo , Proteínas de Unión al Calcio/metabolismo , Cardiotónicos/uso terapéutico , Cromatografía Líquida de Alta Presión , Modelos Animales de Enfermedad , Exosomas/fisiología , Humanos , Glicoproteínas de Membrana/metabolismo , Células Madre Mesenquimatosas/citología , Ratones , Microscopía Electrónica , Tetraspanina 28 , Tetraspanina 29
11.
Mol Cell Proteomics ; 6(10): 1680-9, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17565974

RESUMEN

Transplantation of mesenchymal stem cells (MSCs) has been used to treat a wide range of diseases, and the mechanism of action is postulated to be mediated by either differentiation into functional reparative cells that replace injured tissues or secretion of paracrine factors that promote tissue repair. To complement earlier studies that identified some of the paracrine factors, we profiled the paracrine proteome to better assess the relevance of MSC paracrine factors to the wide spectrum of MSC-mediated therapeutic effects. To evaluate the therapeutic potential of the MSC paracrine proteome, a chemically defined serum-free culture medium was conditioned by MSCs derived from human embryonic stem cells using a clinically compliant protocol. The conditioned medium was analyzed by multidimensional protein identification technology and cytokine antibody array analysis and revealed the presence of 201 unique gene products. 86-88% of these gene products had detectable transcript levels by microarray or quantitative RT-PCR assays. Computational analysis predicted that these gene products will significantly drive three major groups of biological processes: metabolism, defense response, and tissue differentiation including vascularization, hematopoiesis, and skeletal development. It also predicted that the 201 gene products activate important signaling pathways in cardiovascular biology, bone development, and hematopoiesis such as Jak-STAT, MAPK, Toll-like receptor, transforming growth factor-beta, and mTOR (mammalian target of rapamycin) signaling pathways. This study identified a large number of MSC secretory products that have the potential to act as paracrine modulators of tissue repair and replacement in diseases of the cardiovascular, hematopoietic, and skeletal tissues. Moreover our results suggest that human embryonic stem cell-derived MSC-conditioned medium has the potency to treat a variety of diseases in humans without cell transplantation.


Asunto(s)
Células Madre Embrionarias/citología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Proteoma/metabolismo , Cromatografía Liquida , Medios de Cultivo Condicionados , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Genoma Humano , Humanos , Espectrometría de Masas , Redes y Vías Metabólicas , Análisis por Matrices de Proteínas , Proteoma/química , Proteoma/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reproducibilidad de los Resultados
12.
Stem Cells ; 25(2): 425-36, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17053208

RESUMEN

Adult tissue-derived mesenchymal stem cells (MSCs) have demonstrated therapeutic efficacy in treating diseases or repairing damaged tissues through mechanisms thought to be mediated by either cell replacement or secretion of paracrine factors. Characterized, self-renewing human ESCs could potentially be an invariable source of consistently uniform MSCs for therapeutic applications. Here we describe a clinically relevant and reproducible manner of generating identical batches of hESC-derived MSC (hESC-MSC) cultures that circumvents exposure to virus, mouse cells, or serum. Trypsinization and propagation of HuES9 or H1 hESCs in feeder- and serum-free selection media generated three polyclonal, karyotypically stable, and phenotypically MSC-like cultures that do not express pluripotency-associated markers but displayed MSC-like surface antigens and gene expression profile. They differentiate into adipocytes, osteocytes, and chondrocytes in vitro. Gene expression and fluorescence-activated cell sorter analysis identified CD105 and CD24 as highly expressed antigens on hESC-MSCs and hESCs, respectively. CD105+, CD24- monoclonal isolates have a typical MSC gene expression profiles and were identical to each other with a highly correlated gene expression profile (r(2) > .90). We have developed a protocol to reproducibly generate clinically compliant and identical hESC-MSC cultures.


Asunto(s)
Antígenos CD/inmunología , Antígeno CD24/inmunología , Diferenciación Celular , Células Madre Embrionarias/citología , Células Madre Mesenquimatosas/citología , Receptores de Superficie Celular/inmunología , Adipogénesis , Animales , Antígenos CD/genética , Antígenos CD/metabolismo , Antígeno CD24/genética , Antígeno CD24/metabolismo , Separación Celular , Células Cultivadas , Condrogénesis , Células Madre Embrionarias/metabolismo , Células Madre Embrionarias/trasplante , Endoglina , Citometría de Flujo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Células Madre Mesenquimatosas/metabolismo , Ratones , Osteogénesis , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Trasplante de Células Madre
13.
Stem Cell Res ; 1(2): 129-37, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19383393

RESUMEN

Although paracrine effects of mesenchymal stem cells (MSCs) have been suggested previously, cardioprotection by human MSC secretions has never been demonstrated. Human MSC-conditioned medium (CM) was collected by following a clinically compliant protocol. In a porcine model of ischemia and reperfusion injury, intravenous and intracoronary MSC-CM treatment significantly reduced myocardial nuclear oxidative stress as determined by immunostaining for 8-hydroxy-2'-deoxyguanosine. In addition, expression levels of phospho-SMAD2 and active caspase 3 were diminished following CM treatment, suggesting that TGF-beta signaling and apoptosis were reduced. This was associated with a 60% reduction in infarct size and marked improvement of systolic and diastolic cardiac performance as assessed with echocardiography and pressure volume loops. Fractionation studies revealed that only the fraction of the CM containing products >1000 kDa (100-220 nm) provided cardioprotection in a mouse model of ischemia and reperfusion injury. This indicates that the responsible paracrine factor of human MSCs is likely a large complex rather than a single small molecule. These data identify human MSC-CM as a promising therapeutic option to reduce myocardial infarct size in patients with acute MI and suggest that the use of stem cell secretions could extend the applicability of stem cells for therapeutic purposes.


Asunto(s)
Medios de Cultivo Condicionados/farmacología , Células Madre Mesenquimatosas/metabolismo , Infarto del Miocardio/terapia , Animales , Caspasa 3/análisis , Modelos Animales de Enfermedad , Daño por Reperfusión Miocárdica/terapia , Estrés Oxidativo/efectos de los fármacos , Comunicación Paracrina , Proteína Smad2/análisis , Porcinos , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA