Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
1.
Am J Physiol Heart Circ Physiol ; 302(10): H2112-21, 2012 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-22447941

RESUMEN

We previously showed that treatment with tadalafil, a long-acting phosphodiesterase-5a (PDE5a) inhibitor, effectively prevented adverse left ventricular (LV) remodeling of the infarcted heart. We hypothesized that short-hairpin RNA (shRNA) therapy targeting PDE5a would simulate the effects of pharmacological intervention for treatment of postinfarction LV remodeling and dysfunction. Experimental model of myocardial infarction was developed in female mice by permanent ligation of left coronary artery. Immediately after that, an adenoviral vector encoding for shRNA sequence targeting PDE5a (Ad-shPDE5a) was injected intramyocardially, which specifically inhibited PDE5a in the heart. Four weeks later, Ad-shPDE5a treated mice showed significant mitigation of the left ventricle (LV) dilatation and dysfunction as indicated by smaller LV cavity and more preserved ejection fraction and fractional shortening. Infarction size and fibrosis were significantly reduced in Ad-shPDE5a-treated mice. Additionally, more salvaged cardiomyocytes, significantly reduced collagen contents, and higher blood vessel density were observed in Ad-shPDE5a-treated mice. The cytoprotective effects of Ad-shPDE5a were demonstrated in vitro in Ad-shPDE5a transfected cardiomyocytes cultured under oxygen glucose deprivation. Among downstream mediators of PDE5a signaling, cyclic GMP (cGMP) and cGMP-dependent protein kinase G (PKG) were activated with concomitant reduction in caspase-3 activity. However, no significant change in PKA and cAMP activities were observed in Ad-shPDE5a-treated hearts. Inhibition with shRNA improved cardiac remodeling and dysfunction by reducing infarction size and cardiac fibrosis and increased cGMP and PKG activity. These findings suggest that PDE5 inhibition with Ad-shPDE5a is a novel approach for treatment of myocardial infarction.


Asunto(s)
Adenoviridae/genética , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5/metabolismo , Infarto del Miocardio/complicaciones , ARN Interferente Pequeño/uso terapéutico , ARN Viral/uso terapéutico , Disfunción Ventricular Izquierda/prevención & control , Remodelación Ventricular , Animales , Células Cultivadas , Vasos Coronarios/fisiopatología , GMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5/efectos de los fármacos , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5/genética , Femenino , Ligadura/efectos adversos , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Infarto del Miocardio/etiología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , ARN Interferente Pequeño/farmacología , ARN Viral/farmacología , Transfección , Disfunción Ventricular Izquierda/fisiopatología , Remodelación Ventricular/efectos de los fármacos , Remodelación Ventricular/fisiología
2.
J Mol Med (Berl) ; 90(6): 719-30, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22237590

RESUMEN

We previously reported that mesenchymal stem cells (MSC) co-expressing Akt and angiopoietin-1 (Ang-1) preserved infarcted heart function via angiomyogenesis. The present study determined the mechanism of co-overexpression of Akt and Ang-1 in promoting endothelial commitment of MSC. The cells were transduced with vectors encoding for Akt ((Akt)MSC), Ang-1 ((Ang-1)MSC), and both Akt and Ang-1 ((AA)MSC) using Empty vector transduced MSC ((Emp)MSC) as control. Molecular studies indicated a coordinated interaction between Akt and Ang-1 in (AA)MSC and led to non-hypoxic stabilization of hypoxia inducible factor-1α (HIF-Iα) which accentuated under 4-h anoxia. We also observed HIF-Iα dependent induction of hemeoxygenase-1, endothelial specific markers and VEGF in (AA)MSC. Vascular commitment of (AA)MSC was confirmed by immunostaining, Western blotting and flow cytometry for endothelial specific early and late markers including Flt1, Flk1, Tie2, VCAM-1, and von Willebrand Factor-VIII (vWF-VIII) in HIF-Iα dependent fashion besides exhibiting higher emigrational activity and angiogenesis in vitro. (AA)MSC transplanted into rat model of myocardial infarction showed higher Flk1 and Flt1 positivity and also promoted intrinsic Flk1(+) and Flt1(+) cell mobilization into the infarcted heart. Given the ease of availability of MSC and simplicity of approach to co-overexpress Ang-1 and Akt to enhance their endothelial commitment, the strategy will be significant for cellular angiogenesis to treat ischemic heart.


Asunto(s)
Angiopoyetina 1/metabolismo , Células de la Médula Ósea/citología , Células Endoteliales/citología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Proteína Oncogénica v-akt/metabolismo , Proteínas Angiogénicas/genética , Proteínas Angiogénicas/metabolismo , Animales , Western Blotting , Linaje de la Célula , Femenino , Citometría de Flujo , Silenciador del Gen , Hipoxia , Células Madre Mesenquimatosas/citología , Estabilidad Proteica , Ratas
3.
PLoS One ; 6(8): e23667, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21886809

RESUMEN

UNLABELLED: The current protocols for generation of induced pluripotent stem (iPS) cells involve genome integrating viral vectors which may induce tumorgenesis. The aim of this study was to develop and optimize a non-viral method without genetic manipulation for reprogramming of skeletal myoblasts (SMs) using small molecules. METHODS AND RESULTS: SMs from young male Oct3/4-GFP(+) transgenic mouse were treated with DNA methyltransferase (DNMT) inhibitor, RG108. Two weeks later, GFP(+) colonies of SM derived iPS cells (SiPS) expressing GFP and with morphological similarity of mouse embryonic stem (ESCs) were formed and propagated in vitro. SiPS were positive for alkaline phosphatase activity, expressed SSEA1, displayed ES cell specific pluripotency markers and formed teratoma in nude mice. Optimization of culture conditions for embryoid body (EBs) formation yielded spontaneously contracting EBs having morphological, molecular, and ultra-structural similarities with cardiomyocytes and expressed early and late cardiac markers. miR profiling showed abrogation of let-7 family and upregulation of ESCs specific miR-290-295 cluster thus indicating that SiPS were similar to ESCs in miR profile. Four weeks after transplantation into the immunocompetent mice model of acute myocardial infarction (n = 12 per group), extensive myogenesis was observed in SiPS transplanted hearts as compared to DMEM controls (n = 6 per group). A significant reduction in fibrosis and improvement in global heart function in the hearts transplanted with SiPS derived cardiac progenitor cells were observed. CONCLUSIONS: Reprogramming of SMs by DNMT inhibitor is a simple, reproducible and efficient technique more likely to generate transgene integration-free iPS cells. Cardiac progenitors derived from iPS cells propagated extensively in the infarcted myocardium without tumorgenesis and improved cardiac function.


Asunto(s)
Reprogramación Celular/efectos de los fármacos , Indoles/farmacología , Mioblastos Esqueléticos/citología , Mioblastos Esqueléticos/efectos de los fármacos , Miocardio/citología , Propionatos/farmacología , Animales , Biomarcadores/metabolismo , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/antagonistas & inhibidores , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Células Madre Embrionarias/citología , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/metabolismo , Pruebas de Función Cardíaca/efectos de los fármacos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/ultraestructura , Masculino , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Mioblastos Esqueléticos/metabolismo , Contracción Miocárdica/efectos de los fármacos , Infarto del Miocardio/fisiopatología , Infarto del Miocardio/terapia , Miocardio/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Ftalimidas , Trasplante de Células Madre , Teratoma/patología , Triptófano/análogos & derivados , Remodelación Ventricular/efectos de los fármacos , Virus/metabolismo
4.
Rejuvenation Res ; 14(4): 393-403, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21595512

RESUMEN

Abstract improved growth characteristics of the aging bone marrow cells subsequent to neuropeptide Y (NPY)/neuropeptide Y Y5 receptor (NPY Y5R) ligand-receptor interaction. Bone marrow cells were isolated from neonatal (2-3 weeks), young (8-12 weeks), and old (24-28 months) rats on the basis of their preferential adherence to plastic surface. After culturing the cells at initial seeding density of 1×10(4) cells/cm(2), we found that the proliferation potential of bone marrow cells declined with age. Real-time polymerase chain reaction (PCR) and Western blotting showed that bone marrow cells in different age groups constitutively expressed NPY and NPY receptor subtypes (Y1R, Y2R, and Y5R). However, NPY and Y5R expression increased by more than 130-fold and decreased by 28-fold, respectively, in old bone marrow cells as compared to young bone marrow cells. NPY (10 nM) stimulated the proliferation of all bone marrow cells age groups, and their proliferation was blocked by Y5R antagonist. However, the pro-proliferative effect of NPY on old bone marrow cells was weaker than other cell groups due to lower Y5R expression. Y5R gene transfection of old bone marrow cells with subsequent NPY(3-36) (10 nM) treatment significantly increased proliferation of old bone marrow cells (>56%) as compared to green fluorescence protein-transfected control old bone marrow cells. Stimulation of old bone marrow cells by NPY treatment rejuvenated the growth characteristics of aging bone marrow cells as a result of Y5R overexpression.


Asunto(s)
Envejecimiento/metabolismo , Células de la Médula Ósea/citología , Neuropéptido Y/metabolismo , Receptores de Neuropéptido Y/metabolismo , Envejecimiento/efectos de los fármacos , Animales , Biomarcadores/metabolismo , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/enzimología , Proliferación Celular/efectos de los fármacos , Senescencia Celular/efectos de los fármacos , Células Clonales , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Ligandos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Neuropéptido Y/genética , Neuropéptido Y/farmacología , Fosforilación/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Ratas , Ratas Endogámicas F344 , Ratas Transgénicas , Receptores de Neuropéptido Y/genética , Células del Estroma/citología , Células del Estroma/efectos de los fármacos , Células del Estroma/enzimología
5.
Mol Pharm ; 8(5): 1446-57, 2011 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-21591779

RESUMEN

The conventional treatment modalities for ischemic heart disease only provide symptomatic relief to the patient without repairing and regenerating the damaged myocardium. Stem cell transplantation has emerged as a promising alternative therapeutic approach for cardiovascular diseases. Stem cells possess the potential of differentiation to adopt morphofunctional cardiac and vasculogenic phenotypes to repopulate the scar tissue and restore regional blood flow in the ischemic myocardium. These beneficial therapeutic effects make stem cell transplantation the method of choice for the treatment of ischemic heart disease. The efficacy of stem cell transplantation may be augmented by genetic manipulation of the cells prior to transplantation. Not only will insertion of therapeutic transgene(s) into the stem cells support the survival and differentiation of cells in the unfavorable microenvironment of the ischemic myocardium, but also the genetically manipulated stem cells will serve as a source of the transgene expression product in the heart for therapeutic benefits. We provide an overview of the extensively studied stem cell types for cardiac regeneration, the various methods in which these cells have been genetically manipulated and rationale of genetic modification of stem cells for use in regenerative cardiovascular therapeutics.


Asunto(s)
Técnicas de Transferencia de Gen , Terapia Genética , Infarto del Miocardio/terapia , Trasplante de Células Madre , Animales , Células Cultivadas , Terapia Combinada/efectos adversos , Técnicas de Transferencia de Gen/efectos adversos , Terapia Genética/efectos adversos , Corazón/fisiología , Humanos , Infarto del Miocardio/metabolismo , Infarto del Miocardio/fisiopatología , Miocardio/metabolismo , Comunicación Paracrina , Regeneración , Trasplante de Células Madre/efectos adversos , Transgenes
6.
Regen Med ; 6(2): 171-8, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21391851

RESUMEN

AIM: Genetic reprogramming of somatic cells with stemness genes to restore their pluripotent status is being studied extensively to generate pluripotent stem cells as an alternative to embryonic stem cells. This study was designed to examine the effectiveness of skeletal myoblast-derived induced pluripotent stem cells (SkiPS) from young male Oct4/GFP transgenic mice for regeneration of the infarcted heart. METHODS & RESULTS: A mouse model of permanent coronary artery ligation was developed in young female immunocompetent C57BL/6J or C57BL/6x129S4 SV/jae Oct4/GFP mice. SkiPS labeled with Q-dots (3 × 10(5) in 10 µl basal Dulbecco's modified Eagle's medium) were transplanted in and around the area of infarct immediately after coronary artery ligation (n = 16) under direct vision. Control mice (n = 12) were injected with the same number of skeletal myoblasts. Histological studies documented successful engraftment of SkiPS in all the surviving animals 4 weeks later. However, six of the 16 SkiPS-transplanted (37.5%) animal hearts showed intramural teratomas, whereas no tumor growth was observed in the control mice. Q-dot-labeled donor cells were also observed at the site of tumors. Histological studies revealed that teratomas were composed of cells from all of the three embryonic germ layers. Ultra-structure studies confirmed the histological findings and showed regions with well-organized myofibrillar structures in the tumors. CONCLUSION: Undifferentiated induced pluripotent stem cells should not be recommended for cardiac transplantation unless screened for specific teratogenic precursors or predifferentiated into cardiac lineage prior to transplantation.


Asunto(s)
Neoplasias Cardíacas/etiología , Células Madre Pluripotentes Inducidas/fisiología , Infarto del Miocardio/complicaciones , Infarto del Miocardio/patología , Teratoma/etiología , Animales , Transformación Celular Neoplásica/patología , Células Cultivadas , Reprogramación Celular/fisiología , Femenino , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Neoplasias Cardíacas/patología , Inmunocompetencia/fisiología , Células Madre Pluripotentes Inducidas/patología , Células Madre Pluripotentes Inducidas/trasplante , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mioblastos Esqueléticos/citología , Mioblastos Esqueléticos/fisiología , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Teratoma/patología
7.
Int J Stem Cells ; 4(1): 50-60, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24298334

RESUMEN

BACKGROUND AND OBJECTIVES: The incidence of human autologous transplanted skeletal myoblast (SkM) cell death in ischemic myocardium was higher in the first few days after cell therapy. We proposed that human SkM treated by human stromal cell-derived factor (SDF-1α) protein or tranfected by SDF-1α, precondition them against oxidative or anoxic injury. METHODS AND RESULTS: The purification of human SkM (80∼90%) culture was assessed for desmin and CXCR4 expression using immunostaining and flow cytometry respectively. Cells were transfected to overexpress SDF-1α or treated with rSDF-1α (10∼200 ng/ml, 1∼4 h) were either exposed to anoxia or treated with 100µM H2O2 for different time periods (1∼6 h anoxia) (1∼3 h H2O2). Optimized conditions for transfection of SDF-1α gene into human SkM were achieved, using FuGene(TM)6/phSDF-1α(3:2 v/w, 4 h transfection) with 125µ M ZnCl2 (p< 0.001), up to 7 days post-transfection as compared with transfected SkM without ZnCl2 and non-transfected control cells. Transfection efficiency was assessed by immunostaining, ELISA, western blots and PCR. LDH analysis showed significant decrease in release of LDH after exposure to 6 h anoxia or 100µ M H2O2 for 2 h as compared with the normal un-treated or un-transfected SkM (p< 0.001). In western blots assay, SDF-1α over-expressing human SkM or treated with rSDF-1α induced marked expression of total Akt (1.2-fold) and phospho-Akt (2.7-fold), Bcl2 (1.6-fold) and VEGF (5.8-fold) after exposure to 6 h anoxia as compared with human SkM controls. CONCLUSIONS: The preconditioning of donor transplanted human SkM with SDF-1α increased cell survival and promoted cytoprotective effect against oxidative or anoxic injury that may be an innovative approach for clinical application.

8.
Cardiovasc Res ; 88(2): 277-86, 2010 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-20833648

RESUMEN

AIMS: We previously reported that preconditioning of stem cells with insulin-like growth factor-1 (IGF-1) translocated connexin-43 (Cx-43) into mitochondria, causing cytoprotection. We posit that these preconditioning effects could be simulated by mitochondria-specific overexpression of Cx-43. METHODS AND RESULTS: During IGF-1-induced preconditioning of C57black/6 mouse bone marrow stem cell antigen-1(+) (Sca-1(+)) cells, Cx-43 was mainly translocated onto the mitochondrial inner membrane, which was abrogated by an extracellular signal-regulated kinases 1 and 2 (ERK1/2) blocker PD98059. To investigate the role of mitochondrial Cx-43, we successfully designed a vector coding for full-length mouse Cx-43 with a mitochondria-targeting sequence (mito-Cx-43) and cloned into a shuttle vector (pShuttle-IRES-hrGFP-1) for mitochondria-specific overexpression of Cx-43 (mito-Cx-43). Sca-1(+) cells with mito-Cx-43 reduced cytosolic accumulation of cytochrome c, lowered caspase-3 activity, and improved survival during index oxygen-glucose deprivation as determined by terminal deoxynucleotidyl transferase dUTP nick-end labelling and lactate dehydrogenase assays. Computational analysis revealed a B-cell lymphoma-2 (Bcl-2) homology domain-3 (BH3) motif in Cx-43 with a conserved pattern of amino acids consistent with the Bcl-2 family that regulated cytochrome c release. Moreover, computational secondary structure prediction indicated an extended α-helix in this region, a known condition for BH3-driven protein-protein interactions. CONCLUSION: Cx-43 translocation into mitochondria during preconditioning was ERK1/2-dependent. Expression of mito-Cx-43 simulated the cytoprotective effects of preconditioning in stem cells. Structural features of Cx-43 were shared with the Bcl-2 family as determined by computational analysis.


Asunto(s)
Apoptosis , Conexina 43/metabolismo , Mitocondrias/metabolismo , Células Madre/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Antígenos Ly/metabolismo , Apoptosis/efectos de los fármacos , Células Cultivadas , Conexina 43/química , Conexina 43/genética , Secuencia Conservada , Citocromos c/metabolismo , Citoprotección , Factor I del Crecimiento Similar a la Insulina/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Mitocondrias/efectos de los fármacos , Mitocondrias/patología , Membranas Mitocondriales/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Conformación Proteica , Inhibidores de Proteínas Quinasas/farmacología , Estructura Terciaria de Proteína , Transporte de Proteínas , Proteínas Proto-Oncogénicas c-bcl-2/química , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Células Madre/efectos de los fármacos , Células Madre/inmunología , Células Madre/patología , Relación Estructura-Actividad , Factores de Tiempo , Transfección , Regulación hacia Arriba
9.
Am J Physiol Heart Circ Physiol ; 299(5): H1395-404, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20833962

RESUMEN

We hypothesized that inhibition of the cGMP-specific enzyme phosphodiesterase 5A (PDE5A) promoted cGMP/protein kinase G (PKG) activity to condition stem cells for enhanced survival and proliferation. One-time tadalafil treatment (1 µM for 30 min) of mesenchymal stem cells ((Tada)MSCs) provided sustained protection of cells for 36 h. Higher cGMP activity with concomitantly increased PKG1 activity was observed in (Tada)MSCs, which peaked within 12 h after tadalafil treatment. Pretreatment with PKG1 blockers (1 µM KT-5823 or 20 nM K-252a) or transduction with adenoviral PKG1-short-hairpin RNA abolished tadalafil-induced cytoprotection of the cells. A higher proliferation rate was observed in (Tada)MSCs compared with nontreated MSCs ((Cont)MSCs). In a rat model of acute myocardial infarction, (Tada)MSCs transplanted 0 and 24 h after tadalafil treatment showed higher survival compared with (Cont)MSCs on day 2 and day 4 after engraftment. (Tada)MSCs transplanted 48 h after tadalafil treatment lost their protection on both day 2 and day 4 after engraftment, and their rate of survival was similar to (Cont)MSCs. Reduced terminal dUTP nick end-labeling positivity (P < 0.01 vs. (Cont)MSCs) and higher proliferation of (Tada)MSCs (P < 0.01 vs. (Cont)MSCs) was observed in the infarcted heart. Fluorescence immunostaining revealed neomyogenesis in both the infarct and peri-infarct areas. Blood vessel density was significantly increased in group 2 compared with group 1. Transthoracic echocardiographic heart function revealed significant preservation of the indexes of left ventricle contractility and attenuation of remodeling in (Tada)MSC-engrafted animal hearts (group 2) compared with (Cont)MSCs (group 1). PDE5A inhibition using long-acting tadalafil is an innovative approach to promote stem cell survival and proliferation in the infarcted heart.


Asunto(s)
Carbolinas/farmacología , Proliferación Celular/efectos de los fármacos , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5 , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Infarto del Miocardio/cirugía , Inhibidores de Fosfodiesterasa/farmacología , Animales , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , GMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Relación Dosis-Respuesta a Droga , Supervivencia de Injerto/efectos de los fármacos , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Modelos Animales , Neovascularización Fisiológica/fisiología , Ratas , Ratas Endogámicas F344 , Tadalafilo , Factores de Tiempo
10.
Am J Physiol Heart Circ Physiol ; 299(4): H1077-82, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20656888

RESUMEN

We have previously reported that preconditioning of bone marrow-derived mesenchymal stem cells (MSCs) with diazoxide (DZ) significantly improved cell survival via NF-κB signaling. Since micro-RNAs (miRNAs) are critical regulators of a wide variety of biological events, including apoptosis, proliferation, and differentiation, it is likely that DZ-induced survival is mediated by miRNAs. Here we show that miR-146a expressed during preconditioning with DZ is a key regulator of stem cell survival. Treatment of MSCs with DZ (200 µM) markedly increased miR-146a expression and promoted cell survival, as evaluated by lactate dehydrogenase release and transferase-mediated dUTP nick-end labeling staining. Interestingly, blocking NF-κB by IKK-γ NEMO binding domain inhibitor peptide did not induce miR-146a expression, indicating NF-κB regulates miR-146a expression. Moreover, blockade of miR-146a expression by antisense miR-146a inhibitor abolished DZ-induced cytoprotective effects, suggesting a critical role of miR-146a in MSC survival. Computational analysis found a consensus putative target site of miR-146a relevant to apoptosis in the 3' untranslated region of Fas mRNA. The role of Fas as a target gene was substantiated by abrogation of miR-146a, which markedly increased Fas protein expression. This was verified by luciferase reporter assay, which showed that forced expression of miR-146a downregulated Fas expression via targeting its 3'-UTR of this gene. Taken together, these data demonstrated that cytoprotection afforded by preconditioning of MSCs with DZ was regulated by miR-146a induction, which may be a novel therapeutic target in cardiac ischemic diseases.


Asunto(s)
Diazóxido/farmacología , Células Madre Mesenquimatosas/efectos de los fármacos , MicroARNs/metabolismo , FN-kappa B/metabolismo , Receptor fas/metabolismo , Animales , Apoptosis/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Masculino , Células Madre Mesenquimatosas/citología , Modelos Animales , Ratas , Ratas Endogámicas F344 , Transducción de Señal/fisiología , Trasplante de Células Madre
11.
J Cardiovasc Transl Res ; 3(2): 89-102, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20560023

RESUMEN

The harsh ischemic and cytokine-rich microenvironment in the infarcted myocardium, infiltrated by the inflammatory and immune cells, offers a significant challenge to the transplanted donor stem cells. Massive cell death occurs during transplantation as well as following engraftment which significantly lowers the effectiveness of the heart cell therapy. Various approaches have been adopted to overcome this problem nevertheless with multiple limitations with each of these current approaches. Cellular preconditioning and reprogramming by physical, chemical, genetic, and pharmacological manipulation of the cells has shown promise and "prime" the cells to the "state of readiness" to withstand the rigors of lethal ischemia in vitro as well as posttransplantation. This review summarizes the past and present novel approaches of ischemic preconditioning, pharmacological and genetic manipulation using preconditioning mimetics, recombinant growth factor protein treatment, and reprogramming of stem cells to overexpress survival signaling molecules, microRNAs, and trophic factors for intracrine, autocrine, and paracrine effects on cytoprotection.


Asunto(s)
Terapia Genética , Precondicionamiento Isquémico , Infarto del Miocardio/cirugía , Miocardio/patología , Trasplante de Células Madre , Células Madre/efectos de los fármacos , Animales , Apoptosis , Supervivencia Celular , Citoprotección , Humanos , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Péptidos y Proteínas de Señalización Intercelular/genética , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Regeneración , Células Madre/metabolismo , Células Madre/patología , Factores de Tiempo , Resultado del Tratamiento
12.
Heart Asia ; 2(1): 109-11, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-27325955

RESUMEN

BACKGROUND: There is mounting evidence to suggest that the heart has regenerative potential in the event of myocardial injury. Recent studies have shown that a resident population of cardiac progenitor cells (CPCs) in the heart contains both vasculogenic and myogenic lineages. CPCs are able to migrate to the site of injury in the heart for participation in the healing process. The resident CPCs in the heart may also be activated through outside pharmacological intervention to promote their participation in the intrinsic repair process. In the light of these characteristics, CPCs provide a logical source for the heart cell therapy. During the regenerative cardiac process, stem cell niches (a specialised environment surrounding stem cells) provide crucial support needed for their maintenance. DISCUSSION: Compromised niche function may lead to the selection of stem cells that no longer depend on self-renewal factors produced by its environment. The objective of stem cell transplantation associated with tissue-engineered approaches is to create a new modality in the treatment of heart failure. The use of efficient scaffolds will aid to re-establish a favourable microenvironment for stem cell survival, multiplication, differentiation and function. Cardiac tissue engineering using natural and/or synthetic materials in this regard provides a novel possibility in cardiovascular therapeutics.

13.
Antioxid Redox Signal ; 12(6): 693-702, 2010 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-19751147

RESUMEN

We proposed that pharmacological manipulation of mesenchymal stem cells (MSCs) with diazoxide enhanced their survival and regenerative potential via NFkappaB regulation. MSCs preconditioned ((PC)MSCs) with diazoxide and later subjected to oxidant stress with 100 micromol/L H(2)O(2) either immediately or after 24 h exhibited higher survival (p < 0.01 vs nonpreconditioned MSCs; (Non-PC)MSCs) with concomitantly increased phosphorylation of PI3K, Akt, GSK3beta (cytoplasmic), and NF-kappaB (p65) (nuclear). Akt kinase activity was determined as a function of GSK3beta activity. Pretreatment of (PC)MSCs with Wortmannin (Wt), NEMO-binding domain (NBD), or NF-kappaB (p50) siRNA abolished NF-kappaB (p65) activity. Preconditioning increased NF-kappaB-dependent elevation of secretable growth factors associated with their paracrine effects. Inhibition of PI3K activity with Wt reduced (PC)MSCs viability at both early and 24 h time-points. However, inhibition of NF-kappaB reduced viability of (PC)MSCs only at 24 h time-point. For in vivo studies, DMEM without cells (group-1) or containing 1 x 10(6) male (Non-PC)MSCs (group-2), (PC)MSCs (group-3), (PC)MSCs pretreated with Wortmannin (group-4) or NF-kappaB decoy (group-5) were transplanted in a female rat model of acute myocardial infarction. Group-3 showed highest cell survival and growth factor expression, increased angiomyogenesis, and functional improvement. We conclude that activation of NF-kappaB by preconditioning promoted (PC)MSCs survival and angiomyogenic potential in the infarcted heart.


Asunto(s)
Precondicionamiento Isquémico Miocárdico , Células Madre Mesenquimatosas/citología , Infarto del Miocardio/metabolismo , FN-kappa B/metabolismo , Neovascularización Fisiológica , Transducción de Señal , Androstadienos/farmacología , Animales , Supervivencia Celular/efectos de los fármacos , Diazóxido/farmacología , Modelos Animales de Enfermedad , Ecocardiografía , Femenino , Silenciador del Gen , Peróxido de Hidrógeno/farmacología , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , FN-kappa B/antagonistas & inhibidores , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Endogámicas F344 , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Wortmanina
14.
J Cell Mol Med ; 14(1-2): 323-36, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18681907

RESUMEN

The study aims to use cholesterol (Chol) + DOTAP liposome (CD liposome) based human vascular endothelial growth factor-165 (VEGF(165)) gene transfer into skeletal myoblasts (SkMs) for treatment of acute hind limb ischaemia in a rabbit model. The feasibility and efficacy of CD liposome mediated gene transfer with rabbit SkMs were characterized using plasmid carrying enhanced green fluorescent protein (pEGFP) and assessed by flow cytometry. After optimization, SkMs were transfected with CD lipoplexes carrying plasmid-VEGF(165) (CD-pVEGF(165)) and transplanted into rabbit ischaemic limb. Animals were randomized to receive intramuscular injection of Medium199 (M199; group 1), non-transfected SkM (group 2) or CD-pVEGF(165) transfected SkM (group 3). Flow cytometry revealed that up to 16% rabbit SkMs were successfully transfected with pEGFP. Based on the optimized transfection condition, transfected rabbit SkM expressed VEGF(165) up to day 18 with peak at day 2. SkMs were observed in all cell-transplanted groups, as visualized with 6-diamidino-2-phenylindole and bromodeoxyuridine. Angiographic blood vessel score revealed increased collateral vessel development in group 3 (39.7 +/- 2.0) compared with group 2 (21.6 +/- 1.1%, P < 0.001) and group 1 (16.9 +/- 1.1%, P < 0.001). Immunostaining for CD31 showed significantly increased capillary density in group 3 (14.88 +/- 0.9) compared with group 2 (8.5 +/- 0.49, P < 0.001) and group 1 (5.69 +/- 0.3, P < 0.001). Improved blood flow (ml/min./g) was achieved in animal group 3 (0.173 +/- 0.04) as compared with animal group 2 (0.122 +/- 0.016; P= 0.047) and group 1 (0.062 +/- 0.012; P < 0.001). In conclusion, CD liposome mediated VEGF(165) gene transfer with SkMs effectively induced neovascularization in the ischaemic hind limb and may serve as a safe and new therapeutic modality for the repair of acute ischaemic limb disease.


Asunto(s)
Extremidades , Isquemia/terapia , Liposomas/metabolismo , Mioblastos Esqueléticos/fisiología , Transfección , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/uso terapéutico , Animales , Trasplante de Células , Extremidades/irrigación sanguínea , Extremidades/patología , Extremidades/fisiopatología , Femenino , Humanos , Liposomas/ultraestructura , Mioblastos Esqueléticos/citología , Neovascularización Fisiológica , Tamaño de la Partícula , Conejos , Flujo Sanguíneo Regional , Transfección/métodos , Factor A de Crecimiento Endotelial Vascular/genética
15.
Antioxid Redox Signal ; 11(8): 1929-44, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19361254

RESUMEN

The conventional therapeutic modalities for myocardial infarction have limited success in preventing the progression of left ventricular remodeling and congestive heart failure. The heart cell therapy and therapeutic angiogenesis are two promising strategies for the treatment of ischemic heart disease. After extensive assessment of safety and effectiveness in vitro and in experimental animal studies, both of these approaches have accomplished the stage of clinical utility, albeit with limited success due to the inherent limitations and problems of each approach. Neomyogenesis without restoration of regional blood flow may be less meaningful. A combined stem-cell and gene-therapy approach of angiomyogenesis is expected to yield better results as compared with either of the approaches as a monotherapy. The combined therapy approach will help to restore the mechanical contractile function of the weakened myocardium and alleviate ischemic condition by restoration of regional blood flow. In providing an overview of both stem cell therapy and gene therapy, this article is an in-depth and critical appreciation of combined cell and gene therapy approach for myocardial repair.


Asunto(s)
Terapia Genética , Isquemia Miocárdica/patología , Isquemia Miocárdica/terapia , Neovascularización Patológica , Trasplante de Células Madre , Animales , Ratas
16.
Curr Opin Mol Ther ; 10(6): 611-21, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19051139

RESUMEN

Cell therapy is fast emerging as a potential therapeutic option in cardiovascular therapeutics. Because of their inherent myogenic differentiation potential, skeletal myoblasts (SkMs) have been extensively assessed in preclinical and clinical studies for their feasibility, safety and effectiveness for myocardial repair. Bioheart Inc is developing MyoCell, autologous SkMs delivered by MyoCath and MyoStar catheter delivery systems, for the treatment of cardiovascular diseases such as myocardial infarction and congestive heart failure. MyoCell is undergoing phase II/III clinical development and has so far demonstrated safety and efficacy, including improvements in cardiac function in phase I/II clinical trials.


Asunto(s)
Enfermedades Cardiovasculares/terapia , Mioblastos Esqueléticos/trasplante , Trasplante de Células/métodos , Ensayos Clínicos Fase II como Asunto , Ensayos Clínicos Fase III como Asunto , Insuficiencia Cardíaca/terapia , Humanos , Mioblastos Esqueléticos/metabolismo , Infarto del Miocardio/terapia , Resultado del Tratamiento
17.
Ann Thorac Surg ; 86(6): 1841-8, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19021990

RESUMEN

BACKGROUND: We sought to compare host immune cell kinetics, survival profile of donor skeletal myoblasts, and skeletal myoblast graft efficacy after autologous and allogeneic skeletal myoblast transplantation into a rat model of myocardial infarction. METHODS: One week after myocardial infarction, 128 animals were divided into four groups: group 1 (n = 24, receiving medium only), group 2 (n = 24, receiving medium and cyclosporine), group 3 (n = 40, autologous skeletal myoblast transplantation), and group 4 (n = 40, allogeneic skeletal myoblast transplantation with cyclosporine treatment). Rats were euthanized 10 minutes, 1 day, and 4, 7, and 28 days later. Host immune cell kinetics were assessed by immunohistochemical studies for macrophages, and CD4+ and CD8+ lymphocytes. Donor skeletal myoblast survival was confirmed by tracking prelabeled signals, and quantified by beta-gal assay. Heart function was evaluated by echocardiography. RESULTS: A transient immune cell infiltration was demonstrated in group 3, with macrophage infiltration on day 1 and day 4, CD8+ cell infiltration on day 4 and day 7, and CD4+ cell infiltration on day 4. In group 4, immunocyte infiltration was slightly more severe than that in group 3. Automyoblasts and allomyoblasts showed no significant difference of survival from day 1 to day 7 (p > 0.10); however, on day 28, automyoblasts showed better survival than allomyoblasts (p < 0.05). Transplantation of allomyoblasts increased systolic heart function and limited heart dilation after myocardial injury to a similar degree as automyoblasts (p > 0.10). CONCLUSIONS: The use of allomyoblasts is feasible and effective for cardiac repair with immunosuppressive treatment as compared with automyoblasts.


Asunto(s)
Trasplante de Células/métodos , Ciclosporinas/farmacología , Mioblastos Esqueléticos/trasplante , Infarto del Miocardio/cirugía , Animales , Modelos Animales de Enfermedad , Femenino , Rechazo de Injerto , Supervivencia de Injerto , Inmunohistoquímica , Inmunosupresores/farmacología , Masculino , Mioblastos Esqueléticos/inmunología , Infarto del Miocardio/patología , Probabilidad , Distribución Aleatoria , Ratas , Ratas Wistar , Regeneración/fisiología , Factores de Riesgo , Sensibilidad y Especificidad , Trasplante Autólogo , Trasplante Homólogo , Remodelación Ventricular/fisiología
18.
Circ Res ; 103(11): 1300-8, 2008 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-18948617

RESUMEN

We hypothesized that mesenchymal stem cells (MSCs) overexpressing insulin-like growth factor (IGF)-1 showed improved survival and engraftment in the infarcted heart and promoted stem cell recruitment through paracrine release of stromal cell-derived factor (SDF)-1alpha. Rat bone marrow-derived MSCs were used as nontransduced ((Norm)MSCs) or transduced with adenoviral-null vector ((Null)MSCs) or vector encoding for IGF-1 ((IGF-1)MSCs). (IGF-1)MSCs secreted higher IGF-1 until 12 days of observation (P<0.001 versus (Null)MSCs). Molecular studies revealed activation of phosphoinositide 3-kinase, Akt, and Bcl.xL and inhibition of glycogen synthase kinase 3beta besides release of SDF-1alpha in parallel with IGF-1 expression in (IGF-1)MSCs. For in vivo studies, 70 muL of DMEM without cells (group 1) or containing 1.5x10(6) (Null)MSCs (group 2) or (IGF-1)MSCs (group 3) were implanted intramyocardially in a female rat model of permanent coronary artery occlusion. One week later, immunoblot on rat heart tissue (n=4 per group) showed elevated myocardial IGF-1 and phospho-Akt in group 3 and higher survival of (IGF-1)MSCs (P<0.06 versus (Null)MSCs) (n=6 per group). SDF-1alpha was increased in group 3 animal hearts (20-fold versus group 2), with massive mobilization and homing of ckit(+), MDR1(+), CD31(+), and CD34(+) cells into the infarcted heart. Infarction size was significantly reduced in cell transplanted groups compared with the control. Confocal imaging after immunostaining for myosin heavy chain, actinin, connexin-43, and von Willebrand factor VIII showed extensive angiomyogenesis in the infarcted heart. Indices of left ventricular function, including ejection fraction and fractional shortening, were improved in group 3 as compared with group 1 (P<0.05). In conclusion, the strategy of IGF-1 transgene expression induced massive stem cell mobilization via SDF-1alpha signaling and culminated in extensive angiomyogenesis in the infarcted heart.


Asunto(s)
Células de la Médula Ósea/fisiología , Quimiocina CXCL12/fisiología , Corazón/fisiopatología , Movilización de Célula Madre Hematopoyética/métodos , Factor I del Crecimiento Similar a la Insulina/genética , Células Madre Mesenquimatosas/fisiología , Infarto del Miocardio/terapia , Cicatrización de Heridas/fisiología , Animales , Supervivencia Celular , Regulación de la Expresión Génica , Sustancias de Crecimiento/fisiología , Humanos , Factor I del Crecimiento Similar a la Insulina/deficiencia , Células Madre Mesenquimatosas/citología , Infarto del Miocardio/cirugía , Ratas , Ratas Transgénicas , Receptores CXCR4/fisiología , Transducción de Señal
19.
J Mol Cell Cardiol ; 45(4): 554-66, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18561945

RESUMEN

Stem cell transplantation has emerged as a potential modality in cardiovascular therapeutics due to their inherent characteristics of self-renewal, unlimited capacity for proliferation and ability to cross lineage restrictions and adopt different phenotypes. Constrained by extensive death in the unfriendly milieu of ischemic myocardium, the results of heart cell therapy in experimental animal models as well as clinical studies have been less than optimal. Several factors which play a role in early cell death after engraftment in the ischemic myocardium include: absence of survival factors in the transplanted heart, disruption of cell-cell interaction coupled with loss of survival signals from matrix attachments, insufficient vascular supply and elaboration of inflammatory cytokines resulting from ischemia and/or cell death. This article reviews various signaling pathways involved in triggering highly complex forms of cell death and provides critical appreciation of different novel anti-death strategies developed from the knowledge gained from using an ischemic preconditioning approach. The use of pharmacological preconditioning for up-regulation of pro-survival proteins and cardiogenic markers in the transplanted stem cells will be discussed.


Asunto(s)
Infarto del Miocardio/terapia , Trasplante de Células Madre , Acondicionamiento Pretrasplante/métodos , Animales , Antígenos de Diferenciación/metabolismo , Muerte Celular , Diferenciación Celular , Proliferación Celular , Supervivencia Celular , Modelos Animales de Enfermedad , Humanos , Infarto del Miocardio/metabolismo , Células Madre/metabolismo , Donantes de Tejidos
20.
Biomaterials ; 29(13): 2125-37, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18279953

RESUMEN

We aim to investigate the feasibility and efficacy of cholesterol (Chol)+DOTAP liposome (CD liposome) based human vascular endothelial growth factor-165 (hVEGF(165)) gene transfer into human skeletal myoblasts (hSkM) for cardiac repair. The feasibility and efficacy of CD liposome for gene transfer with hSkM was characterized using plasmid carrying enhanced green fluorescent protein (pEGFP). Based on the optimized transfection procedure, hSkM were transfected with CD lipoplexes carrying plasmid-hVEGF(165) (CD-phVEGF(165)). The genetically modified hSkM were transplanted into rat heart model of acute myocardial infarction. Flow cytometry revealed that about 7.99% hSkM could be transfected with pEGFP. Based on the optimized transfection condition, transfected hSkM expressed hVEGF(165) up to day-18 (1.7+/-0.1ng/ml) with peak at day-2 (13.1+/-0.52ng/ml) with >85% cell viability. Animal studies revealed that reduced apoptosis, improved angiogenesis with blood flow in group-3 animal's heart were achieved as compared to group-1 and 2. Ejection fraction was best recovered in group-3 animals. The study demonstrates that though gene transfection efficiency using CD liposome mediated hVEGF(165) gene transfer with hSkM was low; hVEGF(165) gene expression efficiency was sufficient to induce neovascularization, improve blood flow and injured heart function.


Asunto(s)
Colesterol , Ácidos Grasos Monoinsaturados , Liposomas , Mioblastos Esqueléticos/citología , Mioblastos Esqueléticos/metabolismo , Neovascularización Fisiológica , Compuestos de Amonio Cuaternario , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Apoptosis , Células Cultivadas , Colesterol/química , Factor de Crecimiento Epidérmico/genética , Factor de Crecimiento Epidérmico/metabolismo , Ácidos Grasos Monoinsaturados/química , Trasplante de Corazón , Liposomas/química , Microscopía Electrónica de Rastreo , Tamaño de la Partícula , Compuestos de Amonio Cuaternario/química , Ratas , Flujo Sanguíneo Regional , Transfección , Factor A de Crecimiento Endotelial Vascular/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...