Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Eur Heart J ; 36(12): 743-50, 2015 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-24835485

RESUMEN

AIM: There is now compelling evidence that cells committed to a cardiac lineage are most effective for improving the function of infarcted hearts. This has been confirmed by our pre-clinical studies entailing transplantation of human embryonic stem cell (hESC)-derived cardiac progenitors in rat and non-human primate models of myocardial infarction. These data have paved the way for a translational programme aimed at a phase I clinical trial. METHODS AND RESULTS: The main steps of this programme have included (i) the expansion of a clone of pluripotent hESC to generate a master cell bank under good manufacturing practice conditions (GMP); (ii) a growth factor-induced cardiac specification; (iii) the purification of committed cells by immunomagnetic sorting to yield a stage-specific embryonic antigen (SSEA)-1-positive cell population strongly expressing the early cardiac transcription factor Isl-1; (iv) the incorporation of these cells into a fibrin scaffold; (v) a safety assessment focused on the loss of teratoma-forming cells by in vitro (transcriptomics) and in vivo (cell injections in immunodeficient mice) measurements; (vi) an extensive cytogenetic and viral testing; and (vii) the characterization of the final cell product and its release criteria. The data collected throughout this process have led to approval by the French regulatory authorities for a first-in-man clinical trial of transplantation of these SSEA-1(+) progenitors in patients with severely impaired cardiac function. CONCLUSION: Although several facets of this manufacturing process still need to be improved, these data may yet provide a useful platform for the production of hESC-derived cardiac progenitor cells under safe and cost-effective GMP conditions.


Asunto(s)
Células Madre Embrionarias Humanas/trasplante , Separación Inmunomagnética/métodos , Bancos de Tejidos/organización & administración , Animales , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Ensayos Clínicos Fase I como Asunto , Análisis Citogenético , Estudios de Evaluación como Asunto , Humanos , Ratones SCID , Miocitos Cardíacos/citología , Miocitos Cardíacos/trasplante , Conservación de Tejido/métodos , Andamios del Tejido
2.
Circulation ; 122(11 Suppl): S118-23, 2010 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-20837902

RESUMEN

BACKGROUND: The safety and efficacy of myocardial regeneration using embryonic stem cells are limited by the risk of teratoma and the high rate of cell death. METHODS AND RESULTS: To address these issues, we developed a composite construct made of a sheet of adipose tissue-derived stroma cells and embryonic stem cell-derived cardiac progenitors. Ten Rhesus monkeys underwent a transient coronary artery occlusion followed, 2 weeks later, by the open-chest delivery of the composite cell sheet over the infarcted area or a sham operation. The sheet was made of adipose tissue-derived stroma cells grown from a biopsy of autologous adipose tissue and cultured onto temperature-responsive dishes. Allogeneic Rhesus embryonic stem cells were committed to a cardiac lineage and immunomagnetically sorted to yield SSEA-1(+) cardiac progenitors, which were then deposited onto the cell sheet. Cyclosporine was given for 2 months until the animals were euthanized. Preimplantation studies showed that the SSEA-1(+) progenitors expressed cardiac markers and had lost pluripotency. After 2 months, there was no teratoma in any of the 5 cell-treated monkeys. Analysis of >1500 histological sections showed that the SSEA-1(+) cardiac progenitors had differentiated into cardiomyocytes, as evidenced by immunofluorescence and real-time polymerase chain reaction. There were also a robust engraftment of autologous adipose tissue-derived stroma cells and increased angiogenesis compared with the sham animals. CONCLUSIONS: These data collected in a clinically relevant nonhuman primate model show that developmentally restricted SSEA-1(+) cardiac progenitors appear to be safe and highlight the benefit of the epicardial delivery of a construct harboring cells with a cardiomyogenic differentiation potential and cells providing them the necessary trophic support.


Asunto(s)
Tejido Adiposo/citología , Células Madre Embrionarias/trasplante , Infarto del Miocardio/terapia , Miocardio/patología , Regeneración , Trasplante de Células Madre/métodos , Tejido Adiposo/trasplante , Animales , Diferenciación Celular , Modelos Animales de Enfermedad , Humanos , Antígeno Lewis X , Macaca mulatta , Ratones , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Neovascularización Fisiológica , Células del Estroma , Trasplante Autólogo , Trasplante Homólogo
3.
Circulation ; 110(12): 1626-31, 2004 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-15364802

RESUMEN

BACKGROUND: The benefits of skeletal myoblast (SM) transplantation on infarcted myocardium have been investigated extensively; however, little is known about its effects in nonischemic cardiomyopathy models. To address this issue, we tested SM transplantation in CHF147 Syrian hamsters, a strain characterized by a delta-sarcoglycan deficiency that phenotypically features the human setting of primary dilated cardiomyopathy. METHODS AND RESULTS: Cell culture techniques were used to prepare approximately 5x10(6) muscle cells from autologous tibialis anterior muscle, of which 50% were SMs (desmin staining). The cells were injected in 6 sites across the left ventricular wall (n=14). Control animals (n=12) received equivalent volumes of culture medium. Left ventricular systolic function was assessed in a blinded fashion from 2D echocardiographic left ventricular fractional area change, before transplantation, and 4 weeks later. Explanted hearts were processed for the detection of myotubes and quantification of fibrosis. Baseline functional data did not differ between the 2 groups. Four weeks after transplantation, 6 of the 10 surviving grafted hamsters were improved compared with 0 of the 8 survivors of the control group. This translated into a 6% decrease in fractional area change in controls compared with a 24% increase in cell-transplanted hamsters (P=0.001). Engrafted myotubes were consistently detected in all SM transplanted hearts by immunohistochemistry, whereas fibrosis was not worsened by cell injections. CONCLUSIONS: These data suggest that the functional benefits of SM transplantation might extend to nonischemic cardiomyopathy.


Asunto(s)
Cardiomiopatía Dilatada/terapia , Músculo Esquelético/citología , Mioblastos/trasplante , Animales , Cardiomiopatía Dilatada/genética , Cardiomiopatía Dilatada/patología , Cardiomiopatía Dilatada/fisiopatología , Células Cultivadas/trasplante , Cricetinae , Femenino , Fibrosis , Ventrículos Cardíacos/fisiopatología , Inyecciones , Masculino , Mesocricetus , Miocardio/patología , Distribución Aleatoria , Sarcoglicanos/deficiencia , Método Simple Ciego , Trasplante Heterotópico , Función Ventricular Izquierda
4.
J Thorac Cardiovasc Surg ; 130(1): 173-9, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15999059

RESUMEN

OBJECTIVE: Early cell death remains a major limitation of skeletal myoblast transplantation. Because the poor vascularization of the target scars contributes to cell loss, we assessed the effects of combining skeletal myoblast transplantation with administration of hypoxia-inducible factor 1alpha, a master gene that controls the expression of a wide array of angiogenic factors. METHODS: A myocardial infarction was created in 56 rats by means of coronary artery ligation. Eight days later, rats were randomly allocated to receive in-scar injections of culture medium (control animals, n = 11), skeletal myoblasts (5 x 10(6) , n = 13), adenovirus-encoded hypoxia-inducible factor 1alpha (1.0 x 10(10) pfu/mL, n = 7), or skeletal myoblasts (5 x 10(6)) in combination with an empty vector (n = 3) or active hypoxia-inducible factor 1alpha (1.0 x 10(10) pfu/mL, n = 13). A fifth group (n = 9) underwent a staged approach in which hypoxia-inducible factor 1alpha (1.0 x 10(10) pfu/mL) was injected at the time of infarction, followed 8 days later by skeletal myoblasts (5 x 10(6)). Left ventricular function was assessed echocardiographically before transplantation and 1 month thereafter. Explanted hearts were then processed for the immunohistochemical detection of myotubes, quantification of angiogenesis, myoblast engraftment, and cell survival. RESULTS: Baseline ejection fractions were not significantly different among groups (35%-40%). One month later, ejection fraction had decreased from baseline in control hearts and in those injected with hypoxia-inducible factor 1alpha. In contrast, it did not deteriorate after injections of skeletal myoblasts alone or combined with either the empty vector or active hypoxia-inducible factor 1alpha administered sequentially. The most striking change occurred in the skeletal myoblast plus hypoxia-inducible factor 1alpha combined group in which ejection fraction increased dramatically (by 27%) above baseline levels and was thus markedly higher than in all other groups ( P = .0001 and P = .001 vs control animals and animals receiving hypoxia-inducible factor 1alpha, respectively). Compared with skeletal myoblasts alone, the coadministration of hypoxia-inducible factor 1alpha resulted in a significantly greater degree of angiogenesis, cell engraftment, and cell survival. CONCLUSION: Induction of angiogenesis is an effective means of potentiating the functional benefits of myoblast transplantation, and hypoxia-inducible factor 1alpha can successfully achieve this goal.


Asunto(s)
Mioblastos Esqueléticos/trasplante , Neovascularización Fisiológica/efectos de los fármacos , Factores de Transcripción/farmacología , Adenoviridae , Animales , Muerte Celular/fisiología , Femenino , Subunidad alfa del Factor 1 Inducible por Hipoxia , Masculino , Mioblastos Esqueléticos/fisiología , Infarto del Miocardio/cirugía , Distribución Aleatoria , Ratas , Ratas Wistar , Factores de Transcripción/sangre , Factores de Transcripción/uso terapéutico , Función Ventricular Izquierda
5.
Ann Thorac Surg ; 95(6): 2036-41, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23623545

RESUMEN

BACKGROUND: Surgical treatment of retroperitoneal tumors with cavoatrial involvement can be challenging. Completeness of resection of the cava tumor extension is crucial for the patient's survival. We report a monocentric experience with the use of cardiopulmonary bypass and deep hypothermic low flow for the surgical resection of caval and atrial involvement of retroperitoneal tumors. METHODS: Between 2006 and 2011, 9 patients were admitted in our cardiovascular surgery department for retroperitoneal tumors with cavoatrial extension. Every case was performed with cardiopulmonary bypass under deep hypothermia (18°C) with a continuous low-flow perfusion (1 to 1.5 L/min). Cardiopulmonary bypass output was tuned to obtain a nearly bloodless field. Reconstruction of the atriohepatic confluent was carried out with a pericardium patch without inferior vena cava reconstruction. RESULTS: There was no perioperative death. Mean duration of deep hypothermic low flow was 52.2 ± 18.2 minutes. The lowest mean esophageal temperature obtained during procedure was 18.2° ± 1.4°C. No neurologic event was noted postoperatively. Three patients had early complications: one reintervention for bleeding, one reintervention for mediastinitis, and one transient moderate renal failure. After a year, all patients were alive with patent atriohepatic reconstruction. CONCLUSIONS: Cardiopulmonary bypass with deep hypothermic low flow facilitates tumor resection and reconstruction of the atriohepatic confluent. It provides satisfactory postoperative results. It should be considered as an option in the management of these retroperitoneal tumors with cavoatrial involvement.


Asunto(s)
Procedimientos Quirúrgicos Cardíacos/métodos , Atrios Cardíacos/cirugía , Hipotermia Inducida/métodos , Neoplasias Primarias Secundarias/cirugía , Neoplasias Retroperitoneales/cirugía , Vena Cava Inferior/cirugía , Adulto , Anciano , Estudios de Cohortes , Terapia Combinada/métodos , Femenino , Estudios de Seguimiento , Atrios Cardíacos/patología , Hepatectomía/métodos , Humanos , Neoplasias Renales/patología , Neoplasias Renales/cirugía , Laparotomía/métodos , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/cirugía , Masculino , Persona de Mediana Edad , Neoplasias Primarias Secundarias/patología , Nefrectomía/métodos , Neoplasias Retroperitoneales/patología , Estudios Retrospectivos , Medición de Riesgo , Esternotomía/métodos , Factores de Tiempo , Resultado del Tratamiento , Vena Cava Inferior/patología , Adulto Joven
6.
Cardiovasc Res ; 99(4): 640-7, 2013 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-23771945

RESUMEN

AIMS: Few studies have assessed the effects of cell therapy in non-ischaemic cardiomyopathies which, however, contribute to a large number of cardiac failures. Assuming that such conditions are best suited for a global delivery of cells, we assessed the effects of epicardially delivered adipose tissue-derived stroma cell (ADSC) sheets in a mouse model of dilated cardiomyopathy based on cardiac-specific and tamoxifen-inducible invalidation of serum response factor. METHODS AND RESULTS: Three weeks after tamoxifen administration, the function of the left ventricle (LV) was assessed by echocardiography. Twenty-nine mice were then allocated to control (n = 9, non-transgenic), sham (n = 10, transgenic non-treated), and treated (n = 10, transgenic) groups. In the treated group, 3 × 10(6) allogeneic ADSCs were cultured for 2 days onto temperature-responsive polymers and the generated sheets were then transplanted over the surface of the heart. In 10 additional mice, the sheet was made of green fluorescent protein (GFP)-labelled ADSCs to track cell fate. Function, engraftment, and fibrosis were blindly assessed after 3 weeks. In the non-treated group, fractional shortening declined compared with baseline, whereas the sheet application resulted in its stabilization. This correlated with a lesser degree of LV remodelling, as LV end-diastolic and end-systolic diameters did not differ from baseline values. Many GFP(+) cells were identified in the epicardial graft and in the myocardium. Treated animals also displayed a reduced expression of the stress-induced atrial natriuretic factor and beta-myosin heavy chain genes. These protective effects were also accompanied by a reduction of myocardial fibrosis. CONCLUSION: These results strongly suggest the functional relevance of epicardially delivered cell-seeded biomaterials to non-ischaemic heart failure.


Asunto(s)
Tejido Adiposo/citología , Cardiomiopatía Dilatada/terapia , Células del Estroma/trasplante , Animales , Cardiomiopatía Dilatada/patología , Fibrosis , Antígeno Ki-67/análisis , Ratones , Ratones Endogámicos C57BL , Miocardio/patología , Pericardio , Células del Estroma/citología , Células del Estroma/fisiología , Remodelación Ventricular
7.
Interact Cardiovasc Thorac Surg ; 13(2): 211-3, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21565839

RESUMEN

A 75-year-old male, known to have achalasia, was admitted to the intensive care unit with massive upper gastrointestinal bleeding and sepsis. He had a history of purulent pericarditis 18 months earlier. He also presented with atrial fibrillation associated with a cerebral transient ischemic accident two months earlier. A contrast computed tomography scan showed an atrio-esophageal fistula with active extravasation of contrast. He was operated on via a median sternotomy, and the defects in the atrial wall, inferior vena cava and diaphragm were closed using pericardial patches. An esophagectomy was to be performed 24 hours later, but the patient died from septic shock and multiple organ failure before his second procedure.


Asunto(s)
Procedimientos Quirúrgicos Cardíacos/métodos , Acalasia del Esófago/complicaciones , Fístula Esofágica/etiología , Esofagectomía/métodos , Atrios Cardíacos , Cardiopatías/etiología , Anciano , Diagnóstico Diferencial , Acalasia del Esófago/cirugía , Fístula Esofágica/diagnóstico , Fístula Esofágica/cirugía , Resultado Fatal , Fístula/diagnóstico , Fístula/etiología , Fístula/cirugía , Estudios de Seguimiento , Cardiopatías/diagnóstico , Cardiopatías/cirugía , Humanos , Masculino , Tomografía Computarizada por Rayos X
8.
J Clin Invest ; 120(4): 1125-39, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20335662

RESUMEN

Cell therapy holds promise for tissue regeneration, including in individuals with advanced heart failure. However, treatment of heart disease with bone marrow cells and skeletal muscle progenitors has had only marginal positive benefits in clinical trials, perhaps because adult stem cells have limited plasticity. The identification, among human pluripotent stem cells, of early cardiovascular cell progenitors required for the development of the first cardiac lineage would shed light on human cardiogenesis and might pave the way for cell therapy for cardiac degenerative diseases. Here, we report the isolation of an early population of cardiovascular progenitors, characterized by expression of OCT4, stage-specific embryonic antigen 1 (SSEA-1), and mesoderm posterior 1 (MESP1), derived from human pluripotent stem cells treated with the cardiogenic morphogen BMP2. This progenitor population was multipotential and able to generate cardiomyocytes as well as smooth muscle and endothelial cells. When transplanted into the infarcted myocardium of immunosuppressed nonhuman primates, an SSEA-1+ progenitor population derived from Rhesus embryonic stem cells differentiated into ventricular myocytes and reconstituted 20% of the scar tissue. Notably, primates transplanted with an unpurified population of cardiac-committed cells, which included SSEA-1- cells, developed teratomas in the scar tissue, whereas those transplanted with purified SSEA-1+ cells did not. We therefore believe that the SSEA-1+ progenitors that we have described here have the potential to be used in cardiac regenerative medicine.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Células Madre Multipotentes/trasplante , Infarto del Miocardio/terapia , Miocitos Cardíacos/citología , Trasplante de Células Madre , Animales , Proteína Morfogenética Ósea 2/farmacología , Diferenciación Celular , Células Cultivadas , Células Madre Embrionarias/citología , Humanos , Antígeno Lewis X/análisis , Macaca mulatta , MicroARNs/análisis , Células Madre Multipotentes/citología , Factor 3 de Transcripción de Unión a Octámeros/análisis , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/análisis
9.
J Thorac Cardiovasc Surg ; 135(3): 673-8, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18329492

RESUMEN

OBJECTIVE: Cardiac stem cell transplantation as a potential means of regenerating infarcted myocardium is currently receiving a great deal of interest. However, data on these endogenous cardiac precursors are primarily derived from animal studies, and their clinical relevance still remains elusive. METHODS: We prospectively screened 32 endomyocardial biopsies harvested from heart transplant recipients (off rejection episodes) and 18 right appendage biopsies collected during coronary artery bypass surgery, and processed the tissue specimens for the immunohistochemical detection of markers of stemness (c-kit, MDR-1, Isl-1), hematopoietic origin (CD45), mast cells (tryptase), endothelial cells (CD105), and cardiac lineage (Nkx2.5). Confocal microscopy was used for colocalization experiments. Three right appendage biopsies were also cultured for 2 to 3 weeks, at the completion of which c-kit-positive cells were sorted by flow cytometry. RESULTS: In endomyocardial biopsies, a median number of 2.7 (1.8-4) c-kit-positive cells/mm(2) were found, and this number was even significantly smaller in right appendage biopsies (1 [0.5-1.8] c-kit-positive cell/mm(2), P = .01). All of these c-kit-positive cells co-stained for CD45 and were more specifically identified as mast cells by their positive staining for the specific tryptase marker. However, none of the c-kit-positive cells expressed the markers of stemness MDR-1 and Isl-1 or colocalized with CD105. Flow cytometry confirmed the small number of c-kit-positive cells in cultured right atrial appendages. CONCLUSION: These data raise a cautionary note on the therapeutic exploitation of cardiac stem cells in patients with ischemic cardiomyopathy, who may be the elective candidates for regenerative therapy.


Asunto(s)
Apéndice Atrial/patología , Biopsia con Aguja/métodos , Miocitos Cardíacos/patología , Células Madre/fisiología , Adulto , Separación Celular/métodos , Células Cultivadas , Puente de Arteria Coronaria , Endocardio/patología , Femenino , Técnica del Anticuerpo Fluorescente , Trasplante de Corazón/patología , Humanos , Masculino , Microscopía Confocal , Persona de Mediana Edad , Miocardio/patología , Miocitos Cardíacos/trasplante , Probabilidad , Sensibilidad y Especificidad , Estadísticas no Paramétricas , Trasplante de Células Madre , Células Madre/ultraestructura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA