Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Circulation ; 125(23): 2904-13, 2012 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-22589381

RESUMEN

BACKGROUND: Microsomal prostaglandin E(2) synthase-1 (mPGES-1), encoded by the Ptges gene, catalyzes prostaglandin E(2) biosynthesis and is expressed by leukocytes, cardiac myocytes, and cardiac fibroblasts. Ptges(-/-) mice develop more left ventricle (LV) dilation, worse LV contractile function, and higher LV end-diastolic pressure than Ptges(+/+) mice after myocardial infarction. In this study, we define the role of mPGES-1 in bone marrow-derived leukocytes in the recovery of LV function after coronary ligation. METHODS AND RESULTS: Cardiac structure and function in Ptges(+/+) mice with Ptges(+/+) bone marrow (BM(+/+)) and Ptges(+/+) mice with Ptges(-/-) BM (BM(-/-)) were assessed by morphometric analysis, echocardiography, and invasive hemodynamics before and 7 and 28 days after myocardial infarction. Prostaglandin levels and prostaglandin biosynthetic enzyme gene expression were measured by liquid chromatography-tandem mass spectrometry and real-time polymerase chain reaction, immunoblotting, immunohistochemistry, and immunofluorescence microscopy, respectively. After myocardial infarction, BM(-/-) mice had more LV dilation, worse LV systolic and diastolic function, higher LV end-diastolic pressure, more cardiomyocyte hypertrophy, and higher mortality but similar infarct size and pulmonary edema compared with BM(+/+) mice. BM(-/-) mice also had higher levels of COX-1 protein and more leukocytes in the infarct, but not the viable LV, than BM(+/+) mice. Levels of prostaglandin E(2) were higher in the infarct and viable myocardium of BM(-/-) mice than in BM(+/+) mice. CONCLUSIONS: Lack of mPGES-1 in bone marrow-derived leukocytes negatively regulates COX-1 expression, prostaglandin E(2) biosynthesis, and inflammation in the infarct and leads to impaired LV function, adverse LV remodeling, and decreased survival after acute myocardial infarction.


Asunto(s)
Oxidorreductasas Intramoleculares/metabolismo , Microsomas/enzimología , Células Mieloides/enzimología , Infarto del Miocardio/metabolismo , Disfunción Ventricular Izquierda/metabolismo , Animales , Presión Sanguínea/fisiología , Células de la Médula Ósea/enzimología , Ciclooxigenasa 1/metabolismo , Ciclooxigenasa 2/metabolismo , Diástole/fisiología , Modelos Animales de Enfermedad , Oxidorreductasas Intramoleculares/genética , Leucocitos/enzimología , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Infarto del Miocardio/genética , Infarto del Miocardio/mortalidad , Miocarditis/genética , Miocarditis/metabolismo , Miocarditis/mortalidad , Prostaglandina-E Sintasas , Sístole/fisiología , Disfunción Ventricular Izquierda/genética , Disfunción Ventricular Izquierda/mortalidad , Remodelación Ventricular/fisiología
2.
Blood ; 116(16): 2932-41, 2010 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-20606165

RESUMEN

Stem cells exhibit long-term self-renewal by asymmetric division and multipotent differentiation. During embryonic development, cell fate is determined with predictable orientation, differentiation, and partitioning to form the organism. This includes the formation of a hemangioblast from which 2 derivative cell clusters commit to either a hematopoietic or an endothelial lineage. Frequently, it is not clear whether tissue resident stem cells in the adult originate from the bone marrow. Here, we show that blast colony-forming cells exhibiting bilineage (hematopoietic and vascular) potential and long-term self-renewal originate from the uterus in the mouse. This is the first in vitro and in vivo evidence of an adult hemangioblast retained from development in the uterus. Our findings offer new understanding of uterine cell renewal and turnover and may provide insights and opportunities for the study of stem cell maintenance.


Asunto(s)
Hemangioblastos/citología , Hematopoyesis , Útero/citología , Animales , Linaje de la Célula , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Femenino , Ratones , Ratones Endogámicos C57BL
3.
Stem Cells ; 27(8): 1911-20, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19544423

RESUMEN

In recent years, the differentiation of bone marrow cells (BMCs) into myocytes has been extensively investigated, but the findings remain inconclusive. The purpose of this study was to determine the conditions necessary to induce myogenic differentiation in short-term cultures of adult BMCs, and to identify the BMC subpopulation responsible for this phenomenon. We report that high-density cultures of murine hematopoietic BMCs gave rise to spontaneous beating cell clusters in the presence of vascular endothelial and fibroblast growth factors. These clusters originated from c-kit(pos) cells. The formation of the clusters could be completely blocked by adding a c-kit/tyrosine kinase inhibitor, Gleevec (imatinib mesylate; Novartis International, Basel, Switzerland, http://www.novartis.com), to the culture. Cluster formation was also blunted in BMCs from c-kit-deficient (Kit(W)/Kit(W-v)) mice. Clustered cells expressed cardiomyocyte-specific transcription factor genes Gata-4 and Nkx2.5, sarcomeric proteins beta-MHC and MLC-2v, and ANF and connexin-43. Immunostaining revealed alpha-sarcomeric actinin expression in more than 90% of clustered cells. Under electron microscopy, the clustered cells exhibited a sarcomeric myofiber arrangement and z-bands. This study defines the microenvironment required to achieve a reproducible in vitro model of beating, myogenic cell clusters. This model could be used to examine the mechanisms responsible for the postnatal myogenic differentiation of BMCs. Our results identify c-kit(pos) bone marrow hematopoietic cells as the source of the myogenic clusters.


Asunto(s)
Células de la Médula Ósea/citología , Células Madre Hematopoyéticas/citología , Proteínas Proto-Oncogénicas c-kit/metabolismo , Adulto , Animales , Benzamidas , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Diferenciación Celular/fisiología , Linaje de la Célula , Movimiento Celular/fisiología , Células Cultivadas , Factor 2 de Crecimiento de Fibroblastos/farmacología , Expresión Génica , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Humanos , Mesilato de Imatinib , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Electrónica , Microscopía de Contraste de Fase , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Piperazinas/farmacología , Pirimidinas/farmacología , Factor A de Crecimiento Endotelial Vascular/farmacología
4.
Circulation ; 118(14 Suppl): S130-7, 2008 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-18824744

RESUMEN

BACKGROUND: The inability of skeletal myoblasts to transdifferentiate into cardiomyocytes suggests that their beneficial effects on cardiac function after a myocardial infarction are mediated by paracrine effects. We evaluated the roles of these factors in the preservation of matrix architecture (in the infarct and remote regions) by varying the timing (postmyocardial infarction) and delivery site of the implanted cells. METHODS AND RESULTS: Skeletal myoblasts (5x10(6)) or control media were injected into the infarct or noninfarcted myocardium at 5 or 30 days after coronary artery ligation in rats. Function was assessed by echocardiography before transplantation and 14 and 30 days thereafter and with a Millar catheter at 30 days after transplantation. Ventricular geometry, remote fibrillar collagen architecture, and changes in the matrix metalloproteinase-TIMP system were evaluated. Myoblast implantation in both sites and at both times preserved matrix architecture (length and width of collagen fibers) in the remote myocardium (in association with some decreases in remote myocardial matrix metalloprotease activity), improved global cardiac function, and attenuated the progressive increase in end diastolic volume (P<0.05 for all measures compared with medium controls). Cells delivered into the infarct region preserved scar thickness; cells delivered into the noninfarcted myocardium preserved wall thickness. CONCLUSIONS: Regardless of whether the cells were injected into the infarct or the noninfarcted myocardium early after an myocardial infarction or later, skeletal myoblasts improved cardiac function by preventing ventricular dilation and preserving matrix architecture in the remote region, likely mediated by paracrine effects.


Asunto(s)
Procedimientos Quirúrgicos Cardíacos , Mioblastos Esqueléticos/trasplante , Infarto del Miocardio/cirugía , Animales , Células Cultivadas , Colágeno/ultraestructura , Vasos Coronarios , Ecocardiografía , Femenino , Corazón/fisiopatología , Inyecciones , Ligadura , Metaloproteinasas de la Matriz/metabolismo , Infarto del Miocardio/diagnóstico por imagen , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocardio/enzimología , Miocardio/ultraestructura , Ratas , Ratas Endogámicas Lew , Volumen Sistólico , Factores de Tiempo , Función Ventricular , Remodelación Ventricular
5.
Circulation ; 117(13): 1701-10, 2008 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-18347209

RESUMEN

BACKGROUND: Pharmacological inhibition of cyclooxygenase-2 increases the risk of myocardial infarction (MI) and stroke. Microsomal prostaglandin (PG) E(2) synthase-1 (mPGES-1), encoded by the Ptges gene, functions downstream from cyclooxygenase-2 in the inducible PGE(2) biosynthetic pathway. We caused acute MI in Ptges(+/+) and Ptges(-/-) mice to define the role of mPGES-1 in cardiac ischemic injury. METHODS AND RESULTS: Twenty-eight days after MI, Ptges(-/-) mice develop more left ventricular (LV) dilation, have worse LV systolic and diastolic function, and have higher LV end-diastolic pressure than Ptges(+/+) mice but have similar pulmonary wet-to-dry weight ratios, cardiac mass, infarct size, and mortality. The length-to-width ratio of individual cardiomyocytes is significantly greater in Ptges(-/-) than Ptges(+/+) mice after MI, a finding consistent with eccentric cardiomyocyte hypertrophy in Ptges(-/-) mice. Expression of atrial natriuretic peptide, brain natriuretic peptide, and alpha- and beta-myosin heavy chain, markers of ventricular hypertrophy, is higher in the LV of Ptges(-/-) than Ptges(+/+) mice after MI. Ptges(+/+) mice express cyclooxygenase-2 and mPGES-1 protein in inflammatory cells adjacent to the infarct after MI but do not express these proteins in cardiomyocytes. Ptges(-/-) mice express cyclooxygenase-2 in inflammatory cells adjacent to the infarct and do not express mPGES-1 in any cells in the heart. Levels of PGE(2) but not PGD(2), thromboxane A(2), PGI(2), or PGF(2alpha) are higher in the infarct and LV remote from the infarct after MI in Ptges(+/+) than Ptges(-/-) mice. CONCLUSIONS: In Ptges(+/+) mice, mPGES-1 in inflammatory cells catalyzes PGE(2) biosynthesis in the LV after MI. Deletion of mPGES-1 leads to eccentric cardiac myocyte hypertrophy, LV dilation, and impaired LV contractile function after acute MI.


Asunto(s)
Eliminación de Gen , Oxidorreductasas Intramoleculares/deficiencia , Oxidorreductasas Intramoleculares/genética , Microsomas/enzimología , Infarto del Miocardio/enzimología , Infarto del Miocardio/fisiopatología , Remodelación Ventricular/genética , Animales , Oxidorreductasas Intramoleculares/fisiología , Masculino , Ratones , Ratones Endogámicos DBA , Ratones Noqueados , Infarto del Miocardio/genética , Prostaglandina-E Sintasas , Remodelación Ventricular/fisiología
6.
J Clin Invest ; 116(7): 1865-77, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16823487

RESUMEN

Clinical trials of bone marrow stem/progenitor cell therapy after myocardial infarction (MI) have shown promising results, but the mechanism of benefit is unclear. We examined the nature of endogenous myocardial repair that is dependent on the function of the c-kit receptor, which is expressed on bone marrow stem/progenitor cells and on recently identified cardiac stem cells. MI increased the number of c-kit+ cells in the heart. These cells were traced back to a bone marrow origin, using genetic tagging in bone marrow chimeric mice. The recruited c-kit+ cells established a proangiogenic milieu in the infarct border zone by increasing VEGF and by reversing the cardiac ratio of angiopoietin-1 to angiopoietin-2. These oscillations potentiated endothelial mitogenesis and were associated with the establishment of an extensive myofibroblast-rich repair tissue. Mutations in the c-kit receptor interfered with the mobilization of the cells to the heart, prevented angiogenesis, diminished myofibroblast-rich repair tissue formation, and led to precipitous cardiac failure and death. Replacement of the mutant bone marrow with wild-type cells rescued the cardiomyopathic phenotype. We conclude that, consistent with their documented role in tumorigenesis, bone marrow c-kit+ cells act as key regulators of the angiogenic switch in infarcted myocardium, thereby driving efficient cardiac repair.


Asunto(s)
Células de la Médula Ósea/fisiología , Citocinas/metabolismo , Células Madre Hematopoyéticas/fisiología , Miocardio , Neovascularización Fisiológica , Proteínas Proto-Oncogénicas c-kit/metabolismo , Animales , Células de la Médula Ósea/citología , Cardiomiopatías/metabolismo , Cardiomiopatías/patología , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Miocardio/citología , Miocardio/metabolismo , Miocardio/patología , Proteínas Proto-Oncogénicas c-kit/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Quimera por Trasplante , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
7.
FASEB J ; 22(3): 930-40, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17967925

RESUMEN

Cardiovascular disease is the number-one cause of mortality in the developed world. The aim of this study is to define the mechanisms by which bone marrow progenitor cells are mobilized in response to cardiac ischemic injury. We used a closed-chest model of murine cardiac infarction/reperfusion, which segregated the surgical thoracotomy from the induction of cardiac infarction, so that we could study isolated fluctuations in cytokines without the confounding impact of surgery. We show here that bone marrow activation of the c-kit tyrosine kinase receptor in response to released soluble KitL is necessary for bone marrow progenitor cell mobilization after ischemic cardiac injury. We also show that release of KitL and c-kit activation require the activity of matrix metalloproteinase-9 within the bone marrow compartment. Finally, we demonstrate that mice with c-kit dysfunction develop cardiac failure after myocardial infarction and that bone marrow transplantation rescues the failing cardiac phenotype. In light of the ongoing trials of progenitor cell therapy for heart disease, our study outlines the endogenous repair mechanisms that are invoked after cardiac injury. Amplification of this pathway may aid in restoration of cardiac function after myocardial infarction.


Asunto(s)
Médula Ósea/enzimología , Infarto del Miocardio/enzimología , Proteínas Proto-Oncogénicas c-kit/metabolismo , Células Madre/fisiología , Animales , Movimiento Celular , Células Cultivadas , Modelos Animales de Enfermedad , Células Endoteliales/citología , Activación Enzimática , Femenino , Humanos , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Infarto del Miocardio/sangre , Proteínas Proto-Oncogénicas c-kit/genética , ARN Mensajero/biosíntesis , Factor de Células Madre/sangre , Factor de Células Madre/metabolismo , Células Madre/citología , Regulación hacia Arriba
8.
Circulation ; 116(11 Suppl): I77-82, 2007 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-17846329

RESUMEN

BACKGROUND: We hypothesized that c-kit receptor function in the bone marrow is important for facilitating healing, leading to efficient cardiac repair after myocardial infarction (MI). METHODS AND RESULTS: We used Kit(W)/Kit(W-v) c-kit mutant mice and their wild-type littermates to assess the importance of c-kit function in cardiac remodeling after coronary ligation. We found that mutant mice developed 1.6-fold greater ventricular dilation (P=0.008) attributable to a 1.3-fold greater infarct expansion by day 14 after MI (P=0.01). The number of proliferating smooth muscle alpha-actin expressing cells was 1.8-fold lower in mutant mice at day 3 (P<0.01), resulting in a 1.6 to 1.8-fold reduction in total regional nonvascular smooth muscle alpha-actin expressing cells by both microscopy and flow cytometry (P<0.001 for both). This decrease was accompanied by a 1.4-fold reduction in the number of CD31 expressing blood vessels (P<0.05). Prior transplantation of wild-type bone marrow cells into mutant mice rescued the efficient establishment of vessel-rich repair tissue by inducing a 1.5-fold increase in nonvascular smooth muscle alpha-actin expressing cells and CD31 expressing blood vessels (P<0.05 for both). The increased recruitment of cells into the infarct region in the chimeric mice was associated with reduced infarct expansion (P<0.03) compared to wild-type levels. CONCLUSIONS: Bone marrow c-kit function critically impacts the myofibroblast repair response in infarcted hearts. Interventions that increase the infiltration of c-kit+ cells to the infarcted heart may potentiate this endogenous repair response, prevent infarct expansion, and improve the recovery of cardiac function after MI.


Asunto(s)
Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocardio/patología , Proteínas Proto-Oncogénicas c-kit/fisiología , Animales , Trasplante de Médula Ósea/métodos , Femenino , Ratones , Ratones Mutantes , Infarto del Miocardio/genética , Proteínas Proto-Oncogénicas c-kit/genética
9.
Eur J Heart Fail ; 10(1): 14-21, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18160341

RESUMEN

BACKGROUND: Diabetic cardiomyopathy is a common cause of heart failure in diabetic patients, but current treatments do not directly improve ventricular function. Cell transplantation can prevent cardiac dilatation after injury, and may also prevent congestive heart failure in diabetic cardiomyopathy. AIM: This study evaluated the functional effects of smooth muscle cells (SMCs) implanted into the myocardium of insulin- and non insulin-treated diabetic rats. METHODS: Four weeks after streptozotocin infusion, adult Wistar rats were implanted with BrdU-labelled SMCs or culture media (N=12/group). Six rats in each group were also treated with insulin. Echocardiograms were performed at 0, 4 and 8 weeks after streptozotocin injection, and histology and heart function were evaluated at 4 weeks after implantation. RESULTS: Blood glucose levels decreased after insulin treatment. Among cell-injected rats, histology indicated that those that did not receive insulin retained fewer surviving BrdU+ SMCs, and a smaller volume of myocardial tissue positive for alpha-smooth muscle actin. Cardiac function was preserved in the insulin-treated groups relative to those that did not receive insulin. Among insulin-treated rats, the cell-injected group functioned better than the media-injected group. CONCLUSIONS: Diabetic cardiomyopathy is partially treatable with insulin; however, a combination of SMC transplantation and insulin treatment produced the best functional result. Cell transplantation may prevent the progression of diabetic cardiomyopathy in patients whose glucose levels are controlled with insulin.


Asunto(s)
Cardiomiopatías/terapia , Diabetes Mellitus Experimental/complicaciones , Hipoglucemiantes/uso terapéutico , Insulina/uso terapéutico , Miocitos del Músculo Liso/trasplante , Animales , Aorta/citología , Glucemia/metabolismo , Presión Sanguínea/efectos de los fármacos , Presión Sanguínea/fisiología , Cardiomiopatías/etiología , Terapia Combinada/métodos , Diabetes Mellitus Experimental/sangre , Supervivencia de Injerto/efectos de los fármacos , Masculino , Distribución Aleatoria , Ratas , Ratas Wistar , Volumen Sistólico/efectos de los fármacos , Volumen Sistólico/fisiología , Resultado del Tratamiento , Función Ventricular Izquierda/efectos de los fármacos , Función Ventricular Izquierda/fisiología
10.
Cardiovasc Pathol ; 17(1): 32-9, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18160058

RESUMEN

INTRODUCTION: Murphy Roth Large (MRL) mice have a remarkable regenerative capacity. A recent report demonstrated rapid cardiac healing in these mice following cryogenically induced right ventricular injury, suggesting the potential for new regenerative therapies to restore cardiac function in mammals. We therefore evaluated the cardiac regenerative wound-healing response and functional recovery of MRL mice in response to a clinically relevant left ventricular coronary ligation. METHODS: Female MRL/MpJ+/+ and C57BL/6 mice underwent left coronary artery ligation. Cardiac function was evaluated by echocardiography at Days 0, 5, 15, and 60. At Day 96, invasive hemodynamics were assessed by pressure-volume loops using a Millar catheter. Hearts were perfusion fixed for histomorphometric analysis at Days 5, 15, and 96. Some hearts were fresh frozen at Days 5 and 15 for immunohistochemical analysis and digital quantitation of blood vessel density (CD31) and cellular proliferation (Ki67). RESULTS: MRL mice healed ear punch wounds (89% reduction in area) more extensively than C57BL/6 mice (28% reduction in area) but did not differ functionally from C57BL/6 animals before or after coronary ligation. In addition, blood vessel density and cell proliferation were similar between the two strains. CONCLUSIONS: Although MRL mice rapidly healed ear injury, they did not exhibit regeneration of the left ventricle or enhanced functional improvement in response to coronary ligation. The prospect of cardiac regeneration after myocardial infarction will require further studies designed to elucidate the possible mechanisms of functional restoration.


Asunto(s)
Infarto del Miocardio/fisiopatología , Miocardio/patología , Regeneración , Función Ventricular Izquierda , Cicatrización de Heridas , Animales , Proliferación Celular , Vasos Coronarios/cirugía , Modelos Animales de Enfermedad , Femenino , Ligadura , Ratones , Ratones Endogámicos C57BL , Infarto del Miocardio/patología , Miocitos Cardíacos/patología , Neovascularización Fisiológica , Recuperación de la Función , Factores de Tiempo
11.
Circ Res ; 99(6): 617-25, 2006 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-16931795

RESUMEN

Evidence suggests that bone marrow (BM) cells may give rise to a significant proportion of smooth muscle cells (SMCs) that contribute to intimal hyperplasia after vascular injury; however, the molecular pathways involved and the timeline of these events remain poorly characterized. We hypothesized that the stem cell factor (SCF)/c-Kit tyrosine kinase signaling pathway is critical to neointimal formation by BM-derived progenitors. Wire-induced femoral artery injury in mice reconstituted with wild-type BM cells expressing yellow fluorescent protein was performed, which revealed that 66+/-12% of the SMCs (alpha-smooth muscle actin-positive [alphaSMA(+)] cells) in the neointima were from BM. To characterize the role of the SCF/c-Kit pathway, we used c-Kit deficient W/W(v) and SCF-deficient Steel-Dickie mice. Strikingly, vascular injury in these mice resulted in almost a complete inhibition of neointimal formation, whereas wild-type BM reconstitution of c-Kit mutant mice led to neointimal formation in a similar fashion as wild-type animals, as did chronic administration of SCF in matrix metalloproteinase-9-deficient mice, a model of soluble SCF deficiency. Pharmacological antagonism of the SCF/c-Kit pathway with imatinib mesylate (Gleevec) or ACK2 (c-Kit antibody) also resulted in a marked reduction in intimal hyperplasia. Vascular injury resulted in the local upregulation of SCF expression. c-Kit(+) progenitor cells (PCs) homed to the injured vascular wall and differentiated into alphaSMA(+) cells. Vascular injury also caused an increase in circulating SCF levels which promoted CD34(+) PC mobilization, a response that was blunted in mutant and imatinib mesylate-treated mice. In vitro, SCF promoted adhesion of BM PCs to fibronectin. Additionally, anti-SCF antibodies inhibited adhesion of BM PCs to activated SMCs and diminished SMC differentiation. These data indicate that SCF/c-Kit signaling plays a pivotal role in the development of neointima by BM-derived PCs and that the inhibition of this pathway may serve as a novel therapeutic target to limit aberrant vascular remodeling.


Asunto(s)
Arteria Femoral/lesiones , Piperazinas/uso terapéutico , Pirimidinas/uso terapéutico , Transducción de Señal/efectos de los fármacos , Factor de Células Madre/fisiología , Túnica Íntima/crecimiento & desarrollo , Animales , Benzamidas , Células de la Médula Ósea/citología , Células de la Médula Ósea/fisiología , Adhesión Celular , Diferenciación Celular , Movimiento Celular/efectos de los fármacos , Fibronectinas/metabolismo , Regulación de la Expresión Génica/fisiología , Mesilato de Imatinib , Metaloproteinasa 9 de la Matriz/deficiencia , Ratones , Ratones Noqueados , Músculo Liso Vascular/citología , Factor de Células Madre/administración & dosificación , Factor de Células Madre/deficiencia , Factor de Células Madre/genética , Células Madre/citología , Células Madre/fisiología
12.
Coron Artery Dis ; 16(2): 85-91, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15735400

RESUMEN

In the last 15 years, heart cell implantation to regenerate infarcted myocardium has gone from the bench to clinical trial. Several phase I and II controlled randomized trials showed the feasibility, the side effects and the potential efficacy of cell implantation after myocardial infarction in humans. Preclinical experiments investigating the mechanisms of heart function improvement after cell implantation showed controversial results regarding implanted cell differentiation into cardiomyocytes and highlighted other effects including neovascularization and modifications of the extra cellular matrix remodelling. Ongoing clinical and experimental studies should pave the way for cell implantation to become a therapeutic option to prevent and treat post-myocardial infarction congestive heart failure in a near future.


Asunto(s)
Infarto del Miocardio/terapia , Miocardio/citología , Trasplante de Células Madre , Animales , Corazón/fisiología , Humanos , Infarto del Miocardio/fisiopatología , Regeneración/fisiología , Ingeniería de Tejidos
13.
Can J Cardiol ; 21(4): 355-9, 2005 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-15838563

RESUMEN

Heart failure is reaching epidemic proportions in Canada. The current medical and surgical therapies are inadequate to restore cardiac function. Increasingly, cell therapy has been investigated as a novel approach to regenerate the heart and to restore function. Animal studies have been promising and clinical trials are proceeding. Early results suggest that cell therapy may be efficacious in humans as well, but adverse events have been noted. Although the optimal cell type, mode of delivery and ideal patient population have not been identified, the bulk of the evidence suggests that both cardiologists and cardiac surgeons will use cellular therapy to treat heart failure in the future.


Asunto(s)
Trasplante de Células , Insuficiencia Cardíaca/terapia , Corazón/fisiología , Miocardio/citología , Regeneración , Animales , Trasplante de Médula Ósea , Humanos , Mioblastos Esqueléticos/trasplante , Neovascularización Fisiológica , Trasplante de Células Madre
14.
Ann Thorac Surg ; 75(5): 1414-21, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12735555

RESUMEN

BACKGROUND: Using radial artery grafts in patients with moderate to severe left ventricular dysfunction (LVD; ejection fraction < 35%) has been discouraged for the fear that postoperative vasopressor support may cause graft spasm and lead to ischemic complications. We, therefore, examined the safety of radial grafts in aortocoronary bypass (ACB) patients with LVD. METHODS: Data were collected from 5,455 patients who underwent isolated ACB between January 1995 and September 2001. One thousand eight hundred three patients received a radial artery graft (RadACB), and 3,652 patients did not (NoRadACB). Three hundred seven RadACB, and 819 NoRadACB operations were performed in LVD patients. A matched (age, sex, urgency of operation, diabetes, and renal insufficiency) cohort analysis was performed in LVD patients. Univariate and logistic regression analyses were performed in the entire population and the unmatched RadACB and NoRadACB patient subgroups to examine the effect of radial artery use on postoperative death or myocardial infarction rate. RESULTS: The matched cohort analysis revealed a similar rate of death or myocardial infarction (RadACB, 11 of 242 patients; NoRadACB, 16 of 242 patients; p = 0.32). Left ventricular dysfunction was associated with a higher rate of death or myocardial infarction in both unmatched groups (RadACB, odds ratio, 2.36; 95% confidence interval, 1.38 to 4.58; p = 0.004; NoRadACB, odds ratio, 1.62; 95% confidence interval, 1.18 to 2.24; p < 0.001) and in the entire population (odds ratio, 1.77; 95% confidence interval, 1.32 to 2.35; p = 0.003). An interaction term for patients with LVD and a radial artery graft, which was forced into the logistic regression model for the entire population, was not predictive of death or myocardial infarction (odds ratio, 1.52; 95% confidence interval, 0.75 to 3.10; p = 0.25). CONCLUSIONS: Left ventricular dysfunction carries similar risk for postoperative death or myocardial infarction in RadACB and NoRadACB patients. The presence of LVD in isolation is not a contraindication to the use of radial grafting.


Asunto(s)
Puente de Arteria Coronaria , Enfermedad Coronaria/cirugía , Arteria Radial/trasplante , Disfunción Ventricular Izquierda/complicaciones , Anciano , Estudios de Cohortes , Puente de Arteria Coronaria/efectos adversos , Puente de Arteria Coronaria/métodos , Puente de Arteria Coronaria/mortalidad , Enfermedad Coronaria/complicaciones , Femenino , Humanos , Modelos Logísticos , Masculino , Persona de Mediana Edad , Análisis Multivariante , Infarto del Miocardio/etiología , Estudios Retrospectivos , Factores de Riesgo
15.
Vasc Endovascular Surg ; 38(2): 149-55, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15064846

RESUMEN

The purpose of these authors' study was to analyze their center's experience with orthotopic heart transplantation (OHT) and abdominal aortic aneurysms (AAA) with particular attention to corticosteroid dosing, hemodynamic parameters, and aneurysm growth rate. A retrospective review of all patients (453) who underwent OHT at their university-affiliated medical center over an 18-year period (1981-1999) was undertaken. Nine (2%) patients who developed AAAs were identified and aneurysm growth was correlated with corticosteroid immunosuppression and hemodynamic parameters. The mean age of OHT patients was 44.5 +/-15 years and the majority were males (371 males, 82%). Median follow-up was 5.7 years. Ischemic cardiomyopathy (IC) was the most common indication for transplantation (45.5% of patients). All AAA patients were male (p=0.157), with a mean age of 58.4 +/-4.8 years (p=0.001), and had undergone OHT for IC (p=0.001). Mean arterial blood pressure and ejection fraction in the AAA patients had increased from pretransplant values of 107 mm Hg and 14.3 +/-5.7% to 142 mm Hg (p=0.017) and 54.1 +/-14.1% (p<0.001), respectively, before aneurysm repair. Mean aneurysm diameter at the time of repair was 6.0 +/-0.8 cm, and the average growth rate was 1.2 +/-0.4 cm/year in the 4 patients in whom it could be measured. Aneurysm repair was performed urgently in 2 patients and electively in 7 patients with 1 early postoperative death (11%). The extent of corticosteroid immunosuppression, corticosteroid pulses, and total corticosteroid dosing did not correlate with the rate of aneurysm growth. Improved hemodynamics and progressive posttransplant hypertension may contribute to aneurysm formation and growth in this group of patients.


Asunto(s)
Aneurisma de la Aorta Abdominal/etiología , Trasplante de Corazón , Corticoesteroides/toxicidad , Aneurisma de la Aorta Abdominal/cirugía , Distribución de Chi-Cuadrado , Femenino , Hemodinámica , Humanos , Hipertensión/complicaciones , Inmunosupresores/toxicidad , Masculino , Persona de Mediana Edad , Estudios Retrospectivos
16.
Surg Laparosc Endosc Percutan Tech ; 12(1): 52-7, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12008763

RESUMEN

We sought to determine the efficacy of using robotic assistance to facilitate endoscopic harvesting of internal thoracic arteries (ITAs). A total of 104 patients had ITAs harvested endoscopically with use of both the AESOP 3000 system (Computer Motion, Goleta, CA, U.S.A.) and Zeus robotic telesurgical system (Computer Motion). All ITAs were harvested with a harmonic scalpel (Ethicon Endosurgery, Cincinnati, OH, U.S.A.). With the left lung collapsed, ITAs were harvested with CO2 insufflation through three 5-mm ports in the left chest. All patients tolerated insufflation without hemodynamic compromise. Average ITA harvest time was 61.3 +/- 20.9 minutes. Intraoperative graft flows averaged 36.3 +/- 22.4 mL/min. There were three distal ITA injuries; all other vessels were patent after harvesting and demonstrated no angiographic evidence of injury. This article demonstrates a technique by which ITA can be safely harvested totally endoscopically with use of computer-enhanced robotic systems and a harmonic scalpel, allowing complete pedicle dissection through 5-mm ports with minimal ITA manipulation.


Asunto(s)
Robótica/métodos , Arterias Torácicas/cirugía , Cirugía Torácica Asistida por Video/métodos , Recolección de Tejidos y Órganos/métodos , Anciano , Femenino , Humanos , Masculino , Persona de Mediana Edad , Arterias Torácicas/trasplante , Cirugía Torácica Asistida por Video/instrumentación , Factores de Tiempo
19.
Ann Thorac Surg ; 89(6): 1918-24, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20494049

RESUMEN

BACKGROUND: Bicuspid aortic valve is associated with aortic aneurysm formation that may extend beyond the ascending aorta. METHODS: Between 1979 and 1997, 143 bicuspid aortic valve patients had aortic valve operations with replacement of an aneurysmal ascending aorta: 93 (65%) underwent full root replacement and 50 (35%) underwent separate valve and graft replacement. Distal aortic anastomosis was open in 42 patients (29%) and closed in 101 (71%). Late survival and complications were compared by surgical technique. RESULTS: Patients undergoing full root replacement tended to be younger (mean age 46 +/- 16 vs 59 +/- 13, p < 0.001) and presented with more aortic insufficiency (80% vs 35%, p < 0.001). Three (2.1%) hospital deaths occurred. Event-free survival was 82% (95% confidence interval, 75% to 88%) at 10 years and 41% (95% confidence interval, 11% to 71%) at 20 years. At a median follow-up of 11.5 years, the incidence of new aneurysms and late aortic complications were not significantly different among the procedure groups. Age at the time of operation was the only predictor of late survival (hazard ratio, 1.07; p = 0.007). CONCLUSIONS: Aortic root replacement with distal aortic reconstruction can be achieved with very low operative mortality and excellent long-term outcomes in patients with bicuspid aortic valve and dilated ascending aorta. The type of surgical procedure done in the aortic root and in the distal ascending aorta does not influence late survival, subsequent operation, or aortic complications. This is likely influenced by our patient-specific strategy when replacing the aortic root and distal ascending aorta.


Asunto(s)
Aneurisma de la Aorta/complicaciones , Aneurisma de la Aorta/cirugía , Válvula Aórtica/anomalías , Válvula Aórtica/cirugía , Procedimientos Quirúrgicos Cardíacos/efectos adversos , Procedimientos Quirúrgicos Cardíacos/métodos , Femenino , Humanos , Masculino , Persona de Mediana Edad , Complicaciones Posoperatorias/epidemiología , Estudios Retrospectivos , Resultado del Tratamiento
20.
J Thorac Cardiovasc Surg ; 139(5): 1286-94, 1294.e1-2, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-19931095

RESUMEN

OBJECTIVE: Cell therapy has received much attention for its potential to regenerate ischemic organs, but initial clinical trials in aged patients did not replicate the dramatic benefits recorded in preclinical studies with young animals. This study was designed to improve our understanding of age-related changes in the response to ischemic injury and the regenerative capacity of implanted cells in the context of cell therapy for older recipients. METHODS AND RESULTS: Restoration of regional perfusion after hind limb femoral artery ligation was impaired (P < .05) in old (vs young) rats, reflecting approximately 50% reductions in circulating endothelial progenitor cells and the release of vascular endothelial growth factor/basic fibroblast growth factor. Bone marrow stromal cells from young or old donors implanted into the ischemic hind limbs of young or old rats restored regional perfusion. Specifically, we documented significantly greater (P < .05) angiogenic potential in young (vs old) donor cells when recipient age was controlled and greater (P < .05) regenerative responses in young (vs old) recipients when donor cell age was controlled. Contributing to these differences were significantly greater survival in young (vs old) donor cells (in vitro and after implantation) and about 2-fold more production of vascular endothelial growth factor/basic fibroblast growth factor and mobilization of endogenous endothelial progenitor cells in young (vs old) rats in response to ischemia. CONCLUSIONS: The outcome of cell therapy in older recipients is determined by a combination of age effects on the donor cells and on the recipients' endogenous responses. Donor cell age and recipient age are equally important contributors to the outcome of cell therapy; thus, novel biointerventions will need to target both components of the process.


Asunto(s)
Envejecimiento , Trasplante de Médula Ósea , Células Endoteliales/trasplante , Isquemia/cirugía , Músculo Esquelético/irrigación sanguínea , Neovascularización Fisiológica , Trasplante de Células Madre , Factores de Edad , Animales , Movimiento Celular , Supervivencia Celular , Células Cultivadas , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Femenino , Arteria Femoral/cirugía , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Miembro Posterior , Isquemia/metabolismo , Isquemia/patología , Isquemia/fisiopatología , Flujometría por Láser-Doppler , Ligadura , Músculo Esquelético/patología , Ratas , Ratas Endogámicas Lew , Flujo Sanguíneo Regional , Factores de Tiempo , Factor A de Crecimiento Endotelial Vascular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA