Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Circulation ; 106(12 Suppl 1): I176-82, 2002 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-12354729

RESUMEN

BACKGROUND: The optimal cardiac graft for the repair of congenital heart defects will be composed of autologous cells and will grow with the child. The biodegradable material should permit rapid cellular growth and delayed degradation with minimal inflammation. We compared a new material, epsilon-caprolactone-co-L-lactide sponge reinforced with knitted poly-L-lactide fabric (PCLA), to gelatin (GEL) and polyglycolic acid (PGA), which are previously evaluated materials. METHODS: Syngenic rat aortic smooth muscle cells (SMCs, 2x10(6)) were seeded onto GEL, PGA, and PCLA patches and cultured (n=11 per group). The DNA content in each patch was measured at 1, 2, and 3 weeks after seeding. Histological examination was performed 2 weeks after seeding. Cell-seeded patches were employed to replace a surgically created defect in the right ventricular outflow tract (RVOT) of rats (n=5 per group). Histology was studied at 8 weeks following implantation. RESULTS: In vitro studies showed that the DNA content increased significantly (P<0.05) in all patches between 1 and 3 weeks after seeding. Histology and staining SMCs for anti-alpha-smooth muscle actin (alphaSMA) revealed better growth of cells in the interstices of the grafts with GEL and PCLA than the PGA graft. In vivo studies demonstrated that seeded SMCs survived at least 8 weeks after the patch implantation in all groups. PCLA scaffolds were replaced by more cells with larger alphaSMA-positive areas and by more extracellular matrix with larger elastin-positive areas than with GEL and PGA. The patch did not thin and expanded significantly. The GEL and PGA patches thinned and expanded. All grafts had complete endothelialization on the endocardial surface. CONCLUSIONS: SMC-seeded biodegradable materials can be employed to repair the RVOT. The novel PCLA patches permitted better cellular penetration in vitro and did not thin or dilate in vivo and did not produce an inflammatory response. The cell-seeded PCLA patch may permit the construction of an autologous patch to repair congenital heart defects.


Asunto(s)
Materiales Biocompatibles/farmacología , Cardiopatías/cirugía , Músculo Liso Vascular/trasplante , Poliésteres/farmacología , Ingeniería de Tejidos/métodos , Implantes Absorbibles , Animales , División Celular , Supervivencia Celular , Células Cultivadas , ADN/análisis , Endotelio Vascular/crecimiento & desarrollo , Matriz Extracelular/metabolismo , Esponja de Gelatina Absorbible/farmacología , Cardiopatías Congénitas/cirugía , Cardiopatías/patología , Trasplante de Corazón , Ventrículos Cardíacos/cirugía , Masculino , Músculo Liso Vascular/irrigación sanguínea , Músculo Liso Vascular/citología , Miocardio/citología , Miocardio/metabolismo , Neovascularización Fisiológica , Ácido Poliglicólico/farmacología , Ratas , Ratas Endogámicas Lew
2.
Circulation ; 108(6): 736-40, 2003 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-12874180

RESUMEN

BACKGROUND: Adipocyte-derived hormones may represent a mechanism linking insulin resistance to cardiovascular disease. In the present study, we evaluated the direct effects of resistin, a novel adipocyte-derived hormone, on endothelial activation. METHODS AND RESULTS: Endothelial cells (ECs) were incubated with human recombinant resistin (10 to 100 ng/ML, 24 hours), and endothelin-1 (ET-1) release, ET-1 mRNA expression, and nitric oxide (NO) production were assessed. Transient transfection assays were used to evaluate the effects of resistin on transcription of human ET-1 gene promoter. Furthermore, the effects of resistin on AP-1-mutated ET-1 promoter were evaluated. The effects of resistin on expression of vascular cell adhesion molecule (VCAM-1) and monocyte chemoattractant chemokine (MCP-1) were studied in addition to CD40 receptor, CD40 ligand-induced MCP-1 expression, and tumor necrosis factor receptor-associated factor-3 (TRAF3), an inhibitor of CD40 signaling. Incubation of ECs with resistin resulted in an increase in ET-1 release and ET-1 mRNA expression, with no change in NO production. Whereas treatment with resistin resulted in an increase in ET-1 promoter activity, the AP-1-mutated promoter was inactive after resistin stimulation. Additionally, resistin-treated cells showed increased expression of VCAM-1 and MCP-1, with concomitant reductions in TRAF-3 expression. Resistin did not alter CD40 receptor expression; however, increased CD40 ligand induced MCP-1 production. CONCLUSIONS: The novel adipokine resistin exerts direct effects to promote EC activation by promoting ET-1 release, in part by inducing ET-1 promoter activity via the AP-1 site. Furthermore, resistin upregulates adhesion molecules and chemokines and downregulates TRAF-3, an inhibitor of CD40 ligand signaling. In this fashion, resistin may be mechanistically linked to cardiovascular disease in the metabolic syndrome.


Asunto(s)
Adipocitos/metabolismo , Endotelina-1/genética , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/fisiología , Hormonas Ectópicas/farmacología , Péptidos y Proteínas de Señalización Intercelular , Ligando de CD40/farmacología , Comunicación Celular/fisiología , Células Cultivadas , Quimiocina CCL2/metabolismo , Endotelina-1/metabolismo , Endotelio Vascular/citología , Hormonas Ectópicas/genética , Humanos , Óxido Nítrico/metabolismo , Regiones Promotoras Genéticas/efectos de los fármacos , Regiones Promotoras Genéticas/fisiología , Proteínas/metabolismo , ARN Mensajero/metabolismo , Proteínas Recombinantes/farmacología , Resistina , Factor 3 Asociado a Receptor de TNF , Transfección , Molécula 1 de Adhesión Celular Vascular/metabolismo
3.
Circulation ; 109(7): 833-6, 2004 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-14967730

RESUMEN

BACKGROUND: Because complement-mediated vascular injury participates in atherosclerosis and C-reactive protein (CRP) can activate the complement cascade, we sought to determine whether CRP affects the expression of the protective complement-inhibitory factors on the cell surface of endothelial cells (ECs). METHODS AND RESULTS: Human coronary artery or human saphenous vein ECs were incubated with CRP (0 to 100 microg/mL, 0 to 72 hours), and the expression of the complement-inhibitory proteins decay-accelerating factor (DAF), membrane cofactor protein (CD46), and CD59 were measured by flow cytometry. Incubation with CRP resulted in a significant increase in the expression of all 3 proteins. CRP-induced upregulation of DAF required increased steady-state mRNA and de novo protein synthesis. The increased expression of complement-inhibitory proteins was functionally effective, resulting in significant reduction of complement-mediated lysis of antibody-coated human saphenous vein ECs. CONCLUSIONS: These observations provide evidence for a possible protective role for CRP in atherogenesis.


Asunto(s)
Antígenos CD/biosíntesis , Arteriosclerosis/prevención & control , Proteína C-Reactiva/fisiología , Antígenos CD55/biosíntesis , Antígenos CD59/biosíntesis , Endotelio Vascular/efectos de los fármacos , Glicoproteínas de Membrana/biosíntesis , Citotoxicidad Celular Dependiente de Anticuerpos/efectos de los fármacos , Antígenos CD/genética , Arteriosclerosis/metabolismo , Proteína C-Reactiva/farmacología , Antígenos CD55/genética , Antígenos CD59/genética , Células Cultivadas/efectos de los fármacos , Células Cultivadas/metabolismo , Vasos Coronarios/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Endotelio Vascular/citología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Proteína Cofactora de Membrana , Glicoproteínas de Membrana/genética , ARN Mensajero/biosíntesis , Vena Safena/citología
4.
Circulation ; 107(13): 1783-90, 2003 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-12665485

RESUMEN

BACKGROUND: Accumulating evidence suggests that C-reactive protein (CRP), in addition to predicting vascular disease, may actively facilitate lesion formation by inciting endothelial cell activation. Given the central importance of angiotensin type 1 receptor (AT1-R) in the pathogenesis of atherosclerosis, we examined the effects of CRP on AT1-R expression and kinetics in vascular smooth muscle (VSM) cells. In addition, the effects of CRP on VSM migration, proliferation, and reactive oxygen species (ROS) production were evaluated in the presence and absence of the angiotensin receptor blocker, losartan. Lastly, the effects of CRP (and losartan) on neointimal formation were examined in vivo in a rat carotid angioplasty model. METHODS AND RESULTS: The effects of human recombinant CRP (0 to 100 microg/mL) on AT1-R transcript, mRNA stability, and protein expression were studied in cultured human VSM cells. AT1-R binding was assessed with 125I-labeled angiotensin II (Ang II). VSM migration was assessed with wound cell migration assays, whereas VSM proliferation was determined with [3H]-incorporation and cell number. The effects of CRP (and losartan) on Ang II-induced ROS production were evaluated by 2',7'-dichlorofluorescein fluorescence. Lastly, the effects of CRP (and losartan) on neointimal formation, VSM cell migration, proliferation, and matrix formation were studied in vivo in a rat carotid artery balloon injury model. CRP markedly upregulated AT1-R mRNA and protein expression and increased AT1-R number on VSM cells. CRP promoted VSM migration and proliferation in vitro and increased ROS production. Furthermore, CRP potentiated the effects of Ang II on these processes. In the rat carotid artery angioplasty model, exposure to CRP resulted in an increase in cell migration and proliferation, collagen and elastin content, and AT1-R expression, as well as an increase in neointimal formation; these effects were attenuated by losartan. CONCLUSIONS: CRP, at concentrations known to predict cardiovascular events, upregulates AT1-R-mediated atherosclerotic events in vascular smooth muscle in vitro and in vivo. These data lend credence to the notion that CRP functions as a proatherosclerotic factor as well as a powerful risk marker.


Asunto(s)
Proteína C-Reactiva/farmacología , Músculo Liso Vascular/metabolismo , Receptores de Angiotensina/biosíntesis , Regulación hacia Arriba , Angioplastia de Balón/efectos adversos , Animales , Estenosis Carotídea/etiología , Estenosis Carotídea/patología , División Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Regulación de la Expresión Génica , Humanos , Cinética , Masculino , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/fisiología , ARN Mensajero/biosíntesis , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/análisis , Receptor de Angiotensina Tipo 1 , Receptores de Angiotensina/genética , Receptores de Angiotensina/metabolismo
5.
Circulation ; 108 Suppl 1: II219-25, 2003 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-12970236

RESUMEN

BACKGROUND: Recurrent ventricular dilatation can occur after surgical repair of a left ventricular (LV) aneurysm. Use of an autologous bioengineered muscle graft to replace resected scar tissue may prevent recurrent dilatation and improve cardiac function. METHODS: Vascular smooth muscle cells (SMCs, 5 x 10(6) cells) from rat aortas were seeded onto synthetic PCLA (sponge polymer of epsilon-caprolactone-co-L-lactide reinforced with knitted poly-L-lactide fabric) patches and cultured for 2 weeks to allow tissue formation. Syngenic rats underwent proximal left coronary artery ligation to create a transmural myocardial scar. Four weeks after coronary ligation, cell-seeded patches (n=15) or unseeded patches (n=12) were used for a modified endoventricular circular patch plasty (EVCPP) repair of the infarct area. Ligated controls (n=14) and nonligated normal rats (n=10) had sham surgeries without EVCPP. Cardiac function was assessed by echocardiography and isolated Langendorff heart perfusion. Graft histology and morphology was also assessed. RESULTS: After 8 weeks in vivo, seeded patches were thicker (P<0.05) and smaller in area (P<0.003) than unseeded patches. Only seeded patches had prominent elastic tissue formation (P<0.001) in association with SMCs. LV systolic function by echocardiography was improved in the seeded group compared with both unseeded (P<0.002) and control groups (P<0.0001). LV volumes in both patch repair groups were comparable but were significantly smaller (P<0.05) than controls. LV distensibility tended toward improvement in the seeded group as compared with unseeded hearts, but the difference did not achieve statistical significance (P=0.06). CONCLUSIONS: Surgical repair with muscle-cell seeded grafts reduced abnormal chamber distensibility and improved LV function after myocardial infarction as compared with unseeded grafts. Bioengineered muscle grafts may be superior to synthetic materials for the surgical repair of LV scar.


Asunto(s)
Aneurisma Cardíaco/cirugía , Músculo Liso Vascular/trasplante , Animales , Células Cultivadas , Aneurisma Cardíaco/patología , Aneurisma Cardíaco/fisiopatología , Masculino , Infarto del Miocardio/mortalidad , Infarto del Miocardio/patología , Ratas , Ratas Endogámicas Lew , Ingeniería de Tejidos , Función Ventricular Izquierda , Remodelación Ventricular
6.
Circulation ; 106(12 Suppl 1): I137-42, 2002 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-12354723

RESUMEN

BACKGROUND: Surgical repair of congenital and acquired cardiac defects may be enhanced by the use of autologous bioengineered muscle grafts. These tissue-engineered constructs are not optimal in their formation and function. We hypothesized that a mechanical stretch regimen applied to human heart cells that were seeded on a three-dimensional gelatin scaffold (Gelfoam) would improve tissue formation and enhance graft strength. METHODS AND RESULTS: Heart cells from children undergoing repair of Tetralogy of Fallot were isolated and cultured. Heart cells were seeded on gelatin-matrix scaffolds (Gelfoam) and subjected to cyclical mechanical stress (n=7) using the Bio-Stretch Apparatus (80 cycles/minute for 14 days). Control scaffolds (n=7) were maintained under identical conditions but without cyclical stretch. Cell counting, histology, and computerized image analysis determined cell proliferation and their spatial distribution within the tissue-engineered grafts. Collagen matrix formation and organization was determined with polarized light and laser confocal microscopy. Uniaxial tensile testing assessed tissue-engineered graft function. Human heart cells proliferated within the gelatin scaffold. Remarkably, grafts that were subjected to cyclical stretch demonstrated increased cell proliferation and a marked improvement of cell distribution. Collagen matrix formation and organization was enhanced by mechanical stretch. Both maximal tensile strength and resistance to stretch were improved by cyclical mechanical stretch. CONCLUSION: The cyclical mechanical stretch regimen enhanced the formation of a three-dimensional tissue-engineered cardiac graft by improving the proliferation and distribution of seeded human heart cells and by stimulating organized matrix formation resulting in an order of magnitude increase in the mechanical strength of the graft.


Asunto(s)
Miocardio/citología , Ingeniería de Tejidos/métodos , Implantes Absorbibles , División Celular , Células Cultivadas , Niño , Colágeno/análisis , Colágeno/ultraestructura , Matriz Extracelular/química , Matriz Extracelular/ultraestructura , Esponja de Gelatina Absorbible , Cardiopatías Congénitas/cirugía , Trasplante de Corazón , Humanos , Cinética , Miocardio/química , Miocardio/ultraestructura , Estrés Mecánico , Resistencia a la Tracción
7.
Circulation ; 105(16): 1890-6, 2002 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-11997273

RESUMEN

BACKGROUND: C-reactive protein (CRP) has been suggested to actively participate in the development of atherosclerosis. In the present study, we examined the role of the potent endothelium-derived vasoactive factor endothelin-1 (ET-1) and the inflammatory cytokine interleukin-6 (IL-6) as mediators of CRP-induced proatherogenic processes. METHODS AND RESULTS: Saphenous vein endothelial cells (HSVECs) were incubated with human recombinant CRP (25 microg/mL, 24 hours) and the expression of vascular cell adhesion molecule (VCAM-1), intracellular adhesion molecule (ICAM-1), and monocyte chemoattractant chemokine-1 was determined. The effects of CRP on LDL uptake were assessed in macrophages using immunofluorescent labeling of CD32 and CD14. In each study, the effect of endothelin antagonism (bosentan) and IL-6 inhibition (monoclonal anti-IL-6 antibodies) was examined. The effects of CRP on the secretion of ET-1 and IL-6 from HSVECs were also evaluated. Incubation of HSVECs with recombinant human CRP resulted in a marked increase in ICAM-1 and VCAM-1 expression (P<0.001). Likewise, CRP caused a significant increase in monocyte chemoattractant chemokine-1 production, a key mediator of leukocyte transmigration (P<0.001). CRP caused a marked and sustained increase in native LDL uptake by macrophages (P<0.05). These proatherosclerotic effects of CRP were mediated, in part, via increased secretion of ET-1 and IL-6 (P<0.01) and were attenuated by both bosentan and IL-6 antagonism (P<0.01). CONCLUSIONS: CRP actively promotes a proatherosclerotic and proinflammatory phenotype. These effects are mediated, in part, via the production of ET-1 and IL-6 and are attenuated by mixed ET(A/B) receptor antagonism and IL-6 inhibition. Bosentan may be useful in decreasing CRP-mediated vascular disease.


Asunto(s)
Proteína C-Reactiva/antagonistas & inhibidores , Endotelina-1/fisiología , Endotelio Vascular/metabolismo , Interleucina-6/fisiología , Anticuerpos/farmacología , Arteriosclerosis/etiología , Biomarcadores/análisis , Bosentán , Proteína C-Reactiva/farmacología , Moléculas de Adhesión Celular/metabolismo , Células Cultivadas , Quimiocina CCL2/biosíntesis , Endotelio Vascular/efectos de los fármacos , Humanos , Inflamación/inmunología , Lipoproteínas LDL/metabolismo , Macrófagos/metabolismo , Modelos Cardiovasculares , Sulfonamidas/farmacología
8.
Circulation ; 106(8): 913-9, 2002 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-12186793

RESUMEN

BACKGROUND: Given the central importance of nitric oxide (NO) in the development and clinical course of cardiovascular diseases, we sought to determine whether the powerful predictive value of C-reactive protein (CRP) might be explained through an effect on NO production. METHODS AND RESULTS: Endothelial cells (ECs) were incubated with recombinant CRP (0 to 100 microg/mL, 24 hours), and NO and cyclic guanosine monophosphate (cGMP) production was assessed. The effects of CRP on endothelial NO synthase (eNOS) protein, mRNA expression, and mRNA stability were also examined. In a separate study, the effects of CRP (25 microg/mL) on EC cell survival, apoptosis, and in vitro angiogenesis were evaluated. Incubation of ECs with CRP resulted in a significant inhibition of basal and stimulated NO release, with concomitant reductions in cGMP production. CRP caused a marked downregulation of eNOS mRNA and protein expression. Actinomycin D studies suggested that eNOS downregulation was related to decreased mRNA stability. In conjunction with a decrease in NO production, CRP inhibited both basal and vascular endothelial growth factor-stimulated angiogenesis as assessed by EC migration and capillary-like tube formation. CRP did not induce EC survival but did, however, promote apoptosis in a NO-dependent fashion. CONCLUSIONS: CRP, at concentrations known to predict adverse vascular events, directly quenches the production of the NO, in part, through posttranscriptional effect on eNOS mRNA stability. Diminished NO bioactivity, in turn, inhibits angiogenesis, an important compensatory mechanism in chronic ischemia. Through decreasing NO synthesis, CRP may facilitate the development of diverse cardiovascular diseases. Risk reduction strategies designed to lower plasma CRP may be effective by improving NO bioavailability.


Asunto(s)
Proteína C-Reactiva/farmacología , Endotelio Vascular/metabolismo , Neovascularización Fisiológica , Óxido Nítrico/biosíntesis , Apoptosis , Proteína C-Reactiva/fisiología , Enfermedades Cardiovasculares/etiología , Células Cultivadas , GMP Cíclico/biosíntesis , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Humanos , Neovascularización Fisiológica/efectos de los fármacos , Óxido Nítrico Sintasa/genética , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico Sintasa de Tipo III , Estabilidad del ARN , ARN Mensajero/biosíntesis
9.
Circulation ; 109(17): 2058-67, 2004 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-15078802

RESUMEN

BACKGROUND: Myocardial ischemia provides a potent stimulus to angiogenesis, and the mobilization and differentiation of endothelial progenitor cells (EPCs) has been shown to be important in this process. An elevated level of C-reactive protein (CRP) has emerged as one of the most powerful predictors of cardiovascular disease. However, the impact of CRP on EPC biology is unknown. METHODS AND RESULTS: EPCs were isolated from the peripheral venous blood of healthy male volunteers. Cells were cultured in endothelial cell basal medium-2 in the absence and presence of CRP (5 to 20 microg/mL), rosiglitazone (1 micromol/L), and/or vascular endothelial growth factor. EPC differentiation, survival, and function were assayed. CRP at concentrations > or =15 microg/mL significantly reduced EPC cell number, inhibited the expression of the endothelial cell-specific markers Tie-2, EC-lectin, and VE-cadherin, significantly increased EPC apoptosis, and impaired EPC-induced angiogenesis. EPC-induced angiogenesis was dependent on the presence of nitric oxide, and CRP treatment caused a decrease in endothelial nitric oxide synthase mRNA expression by EPCs. However, all of these detrimental CRP-mediated effects on EPCs were attenuated by pretreatment with rosiglitazone, a peroxisome proliferator-activated receptor-gamma (PPARgamma) agonist. CONCLUSIONS: Human recombinant CRP, at concentrations known to predict adverse vascular outcomes, directly inhibits EPC differentiation, survival, and function, key components of angiogenesis and the response to chronic ischemia. This occurs in part via an effect of CRP to reduce EPC eNOS expression. The PPARgamma agonist rosiglitazone inhibits the negative effects of CRP on EPC biology. The ability of CRP to inhibit EPC differentiation and survival may represent an important mechanism that further links inflammation to cardiovascular disease.


Asunto(s)
Proteína C-Reactiva/farmacología , Células Endoteliales/efectos de los fármacos , Apoptosis/efectos de los fármacos , Proteína C-Reactiva/antagonistas & inhibidores , Enfermedades Cardiovasculares/etiología , Diferenciación Celular/efectos de los fármacos , Células Cultivadas/efectos de los fármacos , Células Endoteliales/citología , Células Endoteliales/fisiología , Inducción Enzimática/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Inflamación , Masculino , Neovascularización Fisiológica/efectos de los fármacos , Óxido Nítrico/biosíntesis , Óxido Nítrico/fisiología , Óxido Nítrico Sintasa/biosíntesis , Óxido Nítrico Sintasa/genética , Óxido Nítrico Sintasa de Tipo III , Receptores Citoplasmáticos y Nucleares/agonistas , Receptores Citoplasmáticos y Nucleares/biosíntesis , Receptores Citoplasmáticos y Nucleares/genética , Proteínas Recombinantes/farmacología , Rosiglitazona , Tiazolidinedionas/farmacología , Factores de Transcripción/agonistas , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética , Factor A de Crecimiento Endotelial Vascular/farmacología
10.
Circulation ; 110(16): 2401-9, 2004 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-15262835

RESUMEN

BACKGROUND: Despite the mounting clinical burden of heart failure, the biomolecules that control myocardial tissue remodeling are poorly understood. TIMP-3 is an endogenous inhibitor of matrix metalloproteinases (MMPs) that has been found to be deficient in failing human myocardium. We hypothesized that TIMP-3 expression prevents maladaptive tissue remodeling in the heart, and accordingly, its deficiency in mice would alone be sufficient to trigger progressive cardiac remodeling and dysfunction similar to human heart failure. METHODS AND RESULTS: Mice with a targeted timp-3 deficiency were evaluated with aging and compared with age-matched wild-type littermates. Loss of timp-3 function triggered spontaneous LV dilatation, cardiomyocyte hypertrophy, and contractile dysfunction at 21 months of age consistent with human dilated cardiomyopathy. Its absence also resulted in interstitial matrix disruption with elevated MMP-9 activity, and activation of the proinflammatory tumor necrosis factor-alpha cytokine system, molecular hallmarks of human myocardial remodeling. CONCLUSIONS: TIMP-3 deficiency disrupts matrix homeostasis and the balance of inflammatory mediators, eliciting the transition to cardiac dilation and dysfunction. Therapeutic restoration of myocardial TIMP-3 may provide a novel approach to limit cardiac remodeling and the progression to failure in patients with dilated cardiomyopathy.


Asunto(s)
Cardiomiopatía Dilatada/enzimología , Inhibidor Tisular de Metaloproteinasa-3/deficiencia , Proteínas ADAM , Proteína ADAM17 , Animales , Cardiomiopatía Dilatada/diagnóstico por imagen , Cardiomiopatía Dilatada/genética , Cardiomiopatía Dilatada/patología , Colágeno/análisis , Progresión de la Enfermedad , Matriz Extracelular/química , Homeostasis , Hipertrofia , Macrófagos/patología , Metaloproteinasa 9 de la Matriz/análisis , Metaloendopeptidasas/análisis , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Cardiovasculares , Contracción Miocárdica , Miocitos Cardíacos/patología , Nitritos/análisis , Receptores Tipo II del Factor de Necrosis Tumoral/análisis , Inhibidor Tisular de Metaloproteinasa-3/genética , Inhibidor Tisular de Metaloproteinasa-3/fisiología , Factor de Necrosis Tumoral alfa/análisis , Ultrasonografía
11.
J Thorac Cardiovasc Surg ; 129(4): 904-11, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15821662

RESUMEN

BACKGROUND: Cell transplantation improves heart function after myocardial infarction. This study investigated the survival of implanted cells in normal and infarcted myocardium. METHODS: Male rat aortic smooth muscle cells were cultured. For the in vitro study, male smooth muscle cells mixed with female smooth muscle cells or male smooth muscle cells injected into a piece of female rat myocardium were used to evaluate the accuracy of quantitative real-time polymerase chain reaction to measure Y chromosomes. For the in vivo study, 2 million live or dead male smooth muscle cells were injected into normal or infarcted female myocardium. At 1 hour and 1 and 4 weeks after transplantation, hearts, lungs, and kidneys were harvested for measurement of Y chromosomes. RESULTS: In vitro, the accuracy of polymerase chain reaction measurement was excellent in cultured cells (r2 = 0.996) and the myocardium (r2 = 0.786). In vivo, 1 hour after 2 x 10(6) cell implantation, live cell numbers decreased to 1.0 +/- 0.2 x 10 6 and 1.1 +/- 0.3 x 10(6) , and dead cell numbers decreased to 0.9 +/- 0.2 x 10(6) and 0.8 +/- 0.2 x 10(6) in the normal and infarcted myocardium, respectively (P < .01 for all groups). Lungs and kidneys contained 8.5% and 1.5% of the implanted cells, but no cells were detected at 1 week. At 1 week, no dead smooth muscle cells were detected in the normal or infarcted myocardium. The numbers of live cells at 1 and 4 weeks were 0.48 +/- 0.06 x 10(6) and 0.27 +/- 0.07 x 10(6) in normal myocardium and 0.29 +/- 0.08 x 10(6) and 0.18 +/- 0.05 x 10(6) in infarcted myocardium. CONCLUSIONS: One hour after implantation, only 50% of smooth muscle cells remained in the implanted area. Some implanted cells deposited in other tissue. Implanted cell survival progressively decreased during the 4-week study.


Asunto(s)
Músculo Liso Vascular/citología , Miocitos del Músculo Liso/trasplante , Animales , Antimetabolitos , Encéfalo/patología , Bromodesoxiuridina , Muerte Celular , Supervivencia Celular , Células Cultivadas , ADN/genética , Femenino , Supervivencia de Injerto , Riñón/patología , Pulmón/patología , Masculino , Infarto del Miocardio/cirugía , Miocardio/patología , Reacción en Cadena de la Polimerasa , Ratas , Ratas Endogámicas Lew , Factores de Tiempo , Cromosoma Y/genética
12.
J Thorac Cardiovasc Surg ; 126(6): 1886-91, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14688701

RESUMEN

OBJECTIVES: Elevated levels of C-reactive protein are one of the strongest prognostic factors in atherosclerosis. In addition to predicting vascular disease, C-reactive protein may directly facilitate the development of a proinflammatory and proatherosclerotic phenotype. Recent studies have demonstrated marked up-regulation of various adhesion molecules and inflammatory responses in endothelial cells subjected to C-reactive protein. The nuclear factor-kappaB signal transduction is known to play a key role in the expression of these proatherogenic entities. This study examines the direct effects of C-reactive protein on nuclear factor-kappaB activation and related mechanisms in saphenous vein endothelial cells. METHODS: The activation of nuclear factor-kappaB was determined by confocal microscopy assessing the nuclear localization of nuclear factor-kappaB in endothelial cells incubated with C-reactive protein (50 microg/mL) for 30 minutes and 3 hours. Cells not incubated with C-reactive protein were used as negative controls, and cells incubated with tumor necrosis factor-alpha (10 ng/mL) for 15 minutes were used as positive controls in all studies. The degradation of IkappaB-alpha and IkappaB-beta was assessed by Western blotting of the cell lysates obtained from cells incubated with human recombinant C-reactive protein (50 microg/mL) for 15 minutes, 30 minutes, and 1 hour. RESULTS: Nuclear factor-kappaB nuclear translocation in endothelial cells increased significantly after 30 minutes of incubation with C-reactive protein (P <.01). Nuclear localization of nuclear factor-kappaB returned to baseline levels after 3 hours of incubation with C-reactive protein. Incubation with C-reactive protein resulted in degradation of IkappaB-alpha that was maximal at 30 minutes (P <.05). C-reactive protein showed no significant effect on IkappaB-beta degradation. CONCLUSIONS: These data demonstrate, for the first time, that C-reactive protein activates the nuclear factor-kappaB signal transduction pathway in endothelial cells. Degradation of IkappaB-alpha, but not IkappaB-beta, seems to be the major pathway leading to nuclear factor-kappaB nuclear translocation and activation induced by C-reactive protein. These data support the concept that C-reactive protein, at concentrations known to predict diverse vascular insults, directly facilitates a proinflammatory and proatherosclerotic phenotype through activation of nuclear factor-kappaB. These data have important implications for saphenous vein atherosclerosis in patients with elevated C-reactive protein levels.


Asunto(s)
Arteriosclerosis/metabolismo , Proteína C-Reactiva/farmacología , Endotelio Vascular/metabolismo , FN-kappa B/metabolismo , Vena Safena/metabolismo , Transducción de Señal , Arteriosclerosis/fisiopatología , Western Blotting , Proteína C-Reactiva/fisiología , Células Cultivadas , Histocitoquímica , Humanos , Proteínas I-kappa B/metabolismo , Microscopía Confocal , Proteínas Recombinantes/farmacología , Transducción de Señal/efectos de los fármacos
13.
J Thorac Cardiovasc Surg ; 126(5): 1537-48, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14666030

RESUMEN

OBJECTIVE: Cell transplantation to prevent congestive heart failure in patients with inherited dilated cardiomyopathy might require the use of noncardiac donor cells unaffected by the genetic defect and cryopreservation to permit cell storage until the time of transplantation. However, the effects of cryopreservation on peripheral muscle cells harvested from a cardiomyopathic recipient and their subsequent ability to restore cardiac structure and function after transplantation are unknown. METHODS: Skeletal myoblasts and vascular smooth muscle cells from cardiomyopathic hamsters (delta-sarcoglycan-deficient BIO 53.58 hamster) and age-matched normal donor hamsters were isolated, expanded in culture, and cryopreserved. After reanimation in culture, cell morphology and growth rate were assessed and compared with values seen in noncryopreserved cells. A total of 4 x 10(6) previously cryopreserved skeletal myoblasts (n = 10) and vascular smooth muscle cells (n = 10) harvested from cardiomyopathic donors were then transplanted into the left ventricles of 17-week-old BIO 53.58 hamsters. Hearts injected with culture medium alone (n = 11) served as controls. Heart function was assessed 5 weeks after transplantation on a Langendorff apparatus, and left ventricular geometry was quantified by means of computerized planimetry. Staining with 5-bromo-2'-deoxyuridine identified the injected cells. RESULTS: Vascular smooth muscle cells from cardiomyopathic donors had an abnormal morphology and diminished growth rates in culture compared with vascular smooth muscle cells from normal donors. These markers of injury were exacerbated by cryopreservation. In contrast, vascular smooth muscle cells from normal donors and skeletal myoblasts from either cardiomyopathic or normal donors appeared normal in culture and were unaffected by cryopreservation. Both cryopreserved vascular smooth muscle cells and skeletal myoblasts from cardiomyopathic donors formed a viable muscle-resembling tissue that prevented wall thinning, limited left ventricular dilatation, and preserved global systolic function in hamsters with a genetic dilated cardiomyopathy. However, attenuation of cardiac remodeling and preservation of global heart function was greater after skeletal myoblast transplantation compared with vascular smooth muscle cell transplantation in parallel to the in vitro morphologic and growth characteristics of these cells. CONCLUSIONS: Cryostorage of healthy donor cells does not prevent the benefits of cell transplantation on limiting remodeling and preserving cardiac function in the failing heart. The health of donor cells in vitro predicts their subsequent benefits on cardiac structure and function after transplantation. Cryopreservation of donor cells might facilitate a clinically applicable and effective approach for ventricular restoration with cell-transplantation therapy for patients with inherited dilated cardiomyopathy.


Asunto(s)
Cardiomiopatía Dilatada/cirugía , Criopreservación/métodos , Músculo Liso Vascular/patología , Músculo Liso Vascular/trasplante , Función Ventricular Izquierda/fisiología , Remodelación Ventricular/fisiología , Análisis de Varianza , Animales , Volumen Cardíaco , División Celular , Trasplante de Células/métodos , Cricetinae , Modelos Animales de Enfermedad , Pruebas de Función Cardíaca , Inmunohistoquímica , Masculino , Músculo Esquelético/citología , Músculo Esquelético/patología , Músculo Esquelético/trasplante , Músculo Liso Vascular/citología , Fotomicrografía , Probabilidad , Valores de Referencia , Sensibilidad y Especificidad , Trasplante Autólogo
14.
J Thorac Cardiovasc Surg ; 127(4): 1180-7, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15052220

RESUMEN

OBJECTIVE: We evaluated the effect of transplanted cell type, time, and region of the heart on transgene expression to determine the potential of combined gene and cell delivery for myocardial repair. METHODS: Lewis rats underwent myocardial cryoinjury 3 weeks before transplantation with heart cells (a mixed culture of cardiomyocytes, smooth muscle cells, endothelial cells and fibroblasts, n = 13), vascular endothelial growth factor-transfected heart cells (n = 13), skeletal myoblasts (n = 13), vascular endothelial growth factor-transfected skeletal myoblasts (n = 13), or medium (control, n = 12). Vascular endothelial growth factor expression in the scar, border zone, and normal myocardium was evaluated at 3 days and at 1, 2, and 4 weeks by means of quantitative polymerase chain reaction. Transplanted cells and vascular endothelial growth factor protein were identified immunohistologically on myocardial sections. RESULTS: Vascular endothelial growth factor levels were very low in control scars but increased transiently after medium injection. Transplantation with heart cells and skeletal myoblasts significantly increased vascular endothelial growth factor expression in the scar and border zone. Transplantation of vascular endothelial growth factor-transfected heart cells and vascular endothelial growth factor-transfected skeletal myoblasts further augmented vascular endothelial growth factor expression, resulting in 4- to 5-fold greater expression of vascular endothelial growth factor in the scar at 1 week. Peak vascular endothelial growth factor expression was greater and earlier in vascular endothelial growth factor-transfected heart cells than in vascular endothelial growth factor-transfected skeletal myoblasts. Vascular endothelial growth factor was primarily expressed by the transplanted cells. Some of the transplanted heart cells and vascular endothelial growth factor-transfected heart cells were identified in the endothelial layer of blood vessels in the scar. CONCLUSIONS: Transplantation of heart cells and skeletal myoblasts induces vascular endothelial growth factor expression in myocardial scars and is greatly augmented by prior transfection with a vascular endothelial growth factor transgene. Vascular endothelial growth factor expression is limited to the scar and border zone for 4 weeks. Both heart cells and skeletal myoblasts may be excellent delivery vehicles for cell-based myocardial gene therapy.


Asunto(s)
Trasplante de Células , Regulación de la Expresión Génica/genética , Miocitos Cardíacos/trasplante , Transgenes/genética , Factor A de Crecimiento Endotelial Vascular/genética , Animales , Modelos Animales de Enfermedad , Femenino , Inmunohistoquímica , Masculino , Modelos Cardiovasculares , Mioblastos Esqueléticos/metabolismo , Mioblastos Esqueléticos/trasplante , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Infarto del Miocardio/terapia , Miocitos Cardíacos/metabolismo , Neovascularización Fisiológica/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Endogámicas Lew , Transfección , Factor A de Crecimiento Endotelial Vascular/biosíntesis
15.
J Thorac Cardiovasc Surg ; 123(1): 89-95, 2002 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11782760

RESUMEN

OBJECTIVE: Hypertrophic obstructive cardiomyopathy has been proposed to be the result of gene mutations of contractile proteins. However, we have previously shown significant elevation of insulin-like growth factor-I (IGF-I) and transforming growth factor-beta1 (TGF-beta1) at the messenger RNA, protein, and receptor levels in patients with hypertrophic obstructive cardiomyopathy when compared with myocardium from patients without this disorder. We hypothesized that this growth factor overexpression is a regional phenomenon. To test this hypothesis, we compared levels of IGF-I and TGF-beta1 in hypertrophic and nonhypertrophic myocardium within the same group of patients with hypertrophic obstructive cardiomyopathy. METHODS: Two biopsy specimens were obtained from each patient undergoing septal myectomy for severely symptomatic hypertrophic obstructive cardiomyopathy, from hypertrophied septum and from nonhypertrophied myocardium (8 patients in total). Clinical data were prospectively recorded. Messenger RNA levels for growth factor were quantified by means of multiplex reverse transcriptase-polymerase chain reaction, expressed as a densitometric ratio of growth factor/glyceraldehyde-3-phosphate dehydrogenase. Protein levels were quantified by means of chemiluminescent slot blot analysis. Growth factor proteins were used to generate a standard curve. RESULTS: IGF-I messenger RNA and protein levels in hypertrophic myocardium were 2.6 and 2.9 times greater, respectively, than in nonhypertrophic myocardium of the same patients (both P <.01). TGF-beta 1 messenger RNA and protein levels in the hypertrophic myocardium were 2.5 and 2.8 times greater, respectively, than the levels in the nonhypertrophied myocardium (both P <.01). There was a significant correlation between the IGF-I protein ratio (hypertrophic/nonhypertrophic myocardium) and the inducible left ventricular outflow tract gradients measured at cardiac catheterization (r = 0.77, P =.025). CONCLUSIONS: Myocardial overexpression of IGF-I and TGF-beta1 is a regional phenomenon in patients with hypertrophic obstructive cardiomyopathy and is likely involved in the pathogenesis of the disorder.


Asunto(s)
Cardiomiopatía Hipertrófica/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Miocardio/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Cardiomiopatía Hipertrófica/cirugía , Femenino , Humanos , Factor I del Crecimiento Similar a la Insulina/genética , Masculino , Persona de Mediana Edad , ARN Mensajero/metabolismo , Receptor IGF Tipo 1/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta1
16.
J Thorac Cardiovasc Surg ; 124(6): 1157-64, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12447182

RESUMEN

OBJECTIVES: Nonbiodegradable synthetic materials have been widely used to repair cardiac defects. Material-related failures, however, such as lack of growth, thrombosis, and infection, do occur. Because a biodegradable scaffold can be replaced by the patient's own cells and will be treated as a foreign body for a limited period, we compared four biodegradable materials (gelatin, polyglycolic acid (PGA), and copolymer made of epsilon-caprolactone and l-lactic acid reinforced with a poly-l-lactide knitted [KN-PCLA] or woven fabric [WV-PCLA]) with a nonbiodegradable polytetrafluoroethylene (PTFE) material. An animal heart model was tested that simulates the in vivo clinical condition to which a synthetic material would be used. METHODS: The five patches were used to repair transmural defects surgically created in the right ventricular outflow tracts of adult rat hearts (n = 5, each patch group). The PTFE patch group served as a control group. At 8 weeks after implantation, the biomaterials were excised. Patch size, patch thickness, infiltrated cell number, extracellular matrix composition, and patch degradation were evaluated. RESULTS: The PTFE patch itself did not change in size except for increasing in thickness because of fibroblast and collagen coverage of both its surfaces. Host cells did not migrate into the PTFE biomaterial. In contrast, cells migrated into the biodegrading gelatin, PGA, and KN-PCLA and WV-PCLA scaffolds. Cellular ingrowth per unit patch area was highest in the KN-PCLA patch. The KN-PCLA patch increased modestly in size and thinness. The WV-PCNA patch did not change in size or thickness. Fibroblasts and collagen were the dominant cellular infiltrate and extracellular matrix formed in the biodegrading scaffolds. The in vivo rates of biomaterial degradation, thinning, and expansion were material specific. All the subendocardial patch surfaces were covered with endothelial cells. No thrombi were seen. CONCLUSIONS: The unique, spongy matrix structure of the PCLA patch favored cell colonization relative to the other patches. The strong, durable outer poly-l-lactide fabric layers in these patches offered physical, biocompatible, and bioresorbable advantages relative to the other biodegradable materials studied. Host cells migrated into all the biomaterials. The cells secreted matrix and formed tissue, which was endothelialized on the endocardial surface. The biomaterial degradation rates and the tissue formation rates were material related. The PCLA grafts hold promise to become a suitable patch for surgical repair.


Asunto(s)
Materiales Biocompatibles , Cardiopatías Congénitas/cirugía , Implantes Absorbibles , Animales , Biodegradación Ambiental , Caproatos , Endocardio/ultraestructura , Gelatina , Lactonas , Masculino , Poliésteres , Ácido Poliglicólico , Politetrafluoroetileno , Ratas , Ratas Endogámicas Lew , Obstrucción del Flujo Ventricular Externo/cirugía
17.
J Thorac Cardiovasc Surg ; 123(6): 1114-9, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12063457

RESUMEN

OBJECTIVES: Evidence has accrued to suggest that diabetic patients face an increased risk of ischemic events and low output syndrome and might mount an inordinate response to ischemia and reperfusion. Because hyperglycemia is a potent stimulus for endothelin-1 production, we hypothesized that increased production, action, or both of endothelin-1 in diabetes might represent an important mediator of endothelial dysfunction in patients with that disease. To this aim, we compared the effects of cardioplegic arrest and reperfusion on coronary sinus effluent endothelin-1 levels and atrial arteriolar vascular responses in diabetic and case-matched nondiabetic patients undergoing coronary artery bypass grafting. METHODS: In study 1 coronary sinus effluent endothelin-1 levels were assessed at baseline and at 1 and 10 minutes after reperfusion in 13 diabetic and 12 nondiabetic patients matched for age, ejection fraction, Parsonnet score, and crossclamp time. In study 2 vascular responses of atrial arterioles subjected to perioperative ischemia-reperfusion were evaluated with videomicroscopy. Atrial microvessels (from appendages) were obtained before and after removal of the aortic crossclamp, and vascular responses to exogenously administered endothelin-1 (10(-10) mol/L) and substance P (10(-8) mol/L) were studied in the presence or absence of BQ-123, an endothelin A receptor antagonist. RESULTS: Diabetic patients elaborated more endothelin-1 at 1 and 10 minutes after reperfusion (P =.01). Endothelin-1-mediated vasoconstriction was similar in diabetic and nondiabetic atrial microvessels before cardioplegic arrest and cardiopulmonary bypass. After cardiopulmonary bypass and reperfusion, endothelin-1-mediated vasoconstriction was enhanced in both groups; however, this response was greater in microvessels from diabetic patients (P =.02). BQ-123, the endothelin A antagonist, attenuated the effects of bypass and reperfusion on endothelin-1-mediated vasoconstriction in both groups (P =.01). Substance P-mediated vasodilatation was similar in diabetic and nondiabetic atrial microvessels before bypass. After bypass and reperfusion, substance P-mediated vasodilatation was diminished in both groups; however, this response was more pronounced in the diabetic group (P =.003). BQ-123 coincubation restored substance P-mediated vasodilatation in both groups. CONCLUSIONS: We determined the following: (1) the coronary effluent release of endothelin-1 is higher in diabetic than in nondiabetic patients after cardiopulmonary bypass and reperfusion; (2) diabetic coronary microvessels respond to bypass and reperfusion with greater endothelin-1-mediated vasoconstriction and diminished nitric oxide-mediated vasodilatation; and (3) these effects are attenuated by endothelin antagonism. Endothelin-1 might be an important mediator of ischemia-reperfusion injury in patients with diabetes. Furthermore, use of endothelin receptor antagonists might be a novel strategy for improving the resistance of the diabetic heart to cardioplegic arrest and reperfusion.


Asunto(s)
Puente de Arteria Coronaria , Vasos Coronarios/fisiopatología , Angiopatías Diabéticas/fisiopatología , Endotelina-1/sangre , Endotelio Vascular/fisiopatología , Anciano , Antagonistas de los Receptores de Endotelina , Paro Cardíaco Inducido , Humanos , Persona de Mediana Edad , Péptidos Cíclicos/farmacología , Periodo Posoperatorio
18.
J Thorac Cardiovasc Surg ; 123(6): 1132-40, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12063460

RESUMEN

OBJECTIVE: The study evaluated the utility of transplanting bone marrow stromal cells in a porcine myocardial infarction model. METHODS: A myocardial infarction was created by occluding the distal left anterior descending artery in pigs with coils and Gelfoam sponge. Sternal bone marrow was aspirated, and stromal cells were cultured and induced to differentiate to a myogenic phenotype with 5-azacytidine. Four weeks after coronary artery occlusion, sestamibi technetium single-photon emission computed tomographic scans were performed, and then either a graft of 100 x 10(6) bone marrow stromal cells (n = 5, 30% labeled with bromodeoxyuridine) or culture medium (n = 6) was injected into the infarct region. Four weeks later the tomographic scans were repeated and cardiac function was assessed with pressure and volume measurements. Morphologic and histologic characteristics of the heart were also studied. RESULTS: Histologic examination found bromodeoxyuridine-labeled cells within the infarct region in islands that had sarcomeres and Z-bands and stained positively for cardiac specific troponin I. The bone marrow stromal cell transplant sites had a greater (P <.05) capillary density than did the control sites. The tomographic scans showed that the hearts with the cell transplants had increases in stroke volume, regional perfusion, and wall motion (P <.05 for all groups) relative to the control hearts. The pressure-volume analysis showed improvement (P <.05) in end-systolic elastance and preload recruitable stroke work in the transplantation group relative to the control group. The left ventricular chamber size was smaller (P <.05) and the scar thickness was greater (P <.05) in the hearts with transplanted cells than in the control hearts (P =.06). CONCLUSION: 5-Azacytidine-treated bone marrow stromal cells transplanted into the myocardial infarct region formed islands of cardiac-like tissue, induced angiogenesis, prevented thinning and dilatation of the infarct region, and improved regional and global contractile function.


Asunto(s)
Trasplante de Médula Ósea , Infarto del Miocardio/cirugía , Animales , Modelos Animales de Enfermedad , Ventrículos Cardíacos/patología , Inmunohistoquímica , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Neovascularización Fisiológica , Radiofármacos , Volumen Sistólico , Células del Estroma/trasplante , Porcinos , Tecnecio Tc 99m Sestamibi , Tomografía Computarizada de Emisión de Fotón Único , Trasplante Autólogo , Función Ventricular
19.
J Thorac Cardiovasc Surg ; 123(6): 1074-83, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12063453

RESUMEN

OBJECTIVES: Contemporary cardioprotective strategies to prevent perioperative ischemia-reperfusion injury have focused on the l-arginine nitric oxide pathway. Tetrahydrobiopterin is an absolute cofactor required for the enzyme nitric oxide synthase and is thus a critical determinant of nitric oxide production. We hypothesized that ischemia-reperfusion results in diminished levels of tetrahydrobiopterin, which might represent a key cellular defect underlying endothelial and myocyte dysfunction after ischemia-reperfusion. To this aim, we examined the effects of tetrahydrobiopterin supplementation in (1) an in vivo experimental model of global ischemia-reperfusion and (2) an in vitro human ventricular heart cell model of simulated ischemia-reperfusion. Measures of endothelial function, oxidant production, cell survival, and cardiac function were used to assess outcome. METHODS: In study 1 Wistar rats were divided into one of 2 groups (n = 10 per group). One group received tetrahydrobiopterin (25 mg x kg(-1) x d(-1) for 7 days), and the other group served as the control group. Hearts were subjected to 30 minutes of ischemia followed by 30 minutes of reperfusion, and left ventricular developed pressure, left ventricular systolic pressure, and left ventricular end-diastolic pressure were determined by using the modified Langendorff technique. In study 2 we quantitated myocardial malondialdehyde, a marker of lipid peroxidation, in ventricular tissues from both groups of animals using butanol phase extraction and spectrophotometric analysis. In study 3 coronary vascular responses were determined in vascular segments of the left coronary artery in both groups of animals after ischemia-reperfusion. Endothelium-dependent and endothelium-independent vasodilatation to acetylcholine and sodium nitroprusside, respectively, were compared between groups. In study 4, using a human ventricular heart cell model of simulated ischemia-reperfusion, we studied the effects of tetrahydrobiopterin (20 micromol/L) on cellular injury (as assessed by means of trypan blue uptake). RESULTS: After ischemia-reperfusion, myocardial dysfunction was evidenced by a decrease in left ventricular developed pressure and an increase in left ventricular end-diastolic pressure (P =.01 compared with baseline). Hearts from tetrahydrobiopterin-treated rats exhibited protection against ischemia-reperfusion injury (left ventricular developed pressure: 74 +/- 4 vs control 42 +/- 8 mm Hg, P =.01; left ventricular end-diastolic pressure: 12 +/- 3 vs 34 +/- 7 mm Hg, P =.01). Furthermore, tetrahydrobiopterin treatment attenuated the rise in malondialdehyde levels after ischemia-reperfusion (P =.01). After reperfusion, coronary endothelial function to acetylcholine was attenuated (P =.003 vs sham-treated mice), whereas responses to sodium nitroprusside remained unchanged. Tetrahydrobiopterin-treated rats exhibited an improvement in acetylcholine-mediated vasorelaxation (P =.01 vs ischemia-reperfusion group). Cellular injury, as assessed by means of trypan blue uptake, was higher in human ventricular heart cells subjected to simulated ischemia-reperfusion; this effect was prevented with tetrahydrobiopterin treatment (P =.001). CONCLUSIONS: Supplemental tetrahydrobiopterin provides a novel cardioprotective effect on left ventricular function, endothelial-vascular reactivity, oxidative damage, and cardiomyocyte injury after ischemia-reperfusion injury and might represent an important cellular target for future operative myocardial protection strategies.


Asunto(s)
Antioxidantes/farmacología , Biopterinas/análogos & derivados , Biopterinas/farmacología , Corazón/efectos de los fármacos , Daño por Reperfusión Miocárdica/prevención & control , Función Ventricular Izquierda/efectos de los fármacos , Adolescente , Animales , Niño , Preescolar , Endotelio Vascular/efectos de los fármacos , Humanos , Técnicas In Vitro , Peroxidación de Lípido , Masculino , Daño por Reperfusión Miocárdica/fisiopatología , Ratas , Ratas Wistar
20.
J Thorac Cardiovasc Surg ; 123(6): 1120-4, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12063458

RESUMEN

OBJECTIVES: We have previously demonstrated an importance of endothelin-1 in diabetic patients undergoing bypass surgery. Recent evidence suggests that cardiomyocytes might also produce endothelin-1, which might directly impair myocyte contractility by increasing intracellular calcium levels. Because hyperglycemia is a potent stimulus of endothelin-1 production, we hypothesized that increased production, action, or both of endothelin-1 might be a mediator of direct cardiomyocyte injury in diabetes. Therefore we studied the effects of endothelin receptor blockers (BQ-123 and bosentan) on hyperglycemia-induced endothelin-1 production and cellular injury after ischemia-reperfusion. METHODS: Using a human ventricular heart cell model of simulated ischemia-reperfusion, we studied the effects of normoglycemia (5 mmol/L, 48 hours) and hyperglycemia (25 mmol/L, 48 hours) on cellular injury and endothelin-1 production. Furthermore, the effects of selective endothelin-A and mixed endothelin-A/B receptor antagonism (with BQ-123 and bosentan, respectively) were evaluated. RESULTS: Cellular injury, as assessed by means of trypan blue uptake, was higher in human ventricular heart cells subjected to hyperglycemia and simulated ischemia-reperfusion injury (P =.01); this effect was prevented with both BQ-123 and bosentan (P =.01). In addition, heart cells from the hyperglycemic group elaborated more endothelin-1 after ischemia-reperfusion (P =.02). CONCLUSIONS: Endothelin-1 production and cellular injury were greater in human ventricular heart cells subjected to hyperglycemic conditions and simulated ischemia-reperfusion. These effects are mediated by endothelin-A receptors because both BQ-123 and bosentan exerted similar degrees of protection. Endothelin receptor blockade is a novel strategy to improve the resistance of the diabetic heart to cardioplegic arrest and reperfusion.


Asunto(s)
Antihipertensivos/farmacología , Angiopatías Diabéticas/fisiopatología , Antagonistas de los Receptores de Endotelina , Endotelina-1/sangre , Daño por Reperfusión Miocárdica/fisiopatología , Péptidos Cíclicos/farmacología , Sulfonamidas/farmacología , Bosentán , Células Cultivadas , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA