Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
PLoS One ; 13(10): e0204595, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30303984

RESUMEN

The in vivo microenvironment of tissues provides myriad unique signals to cells. Thus, following isolation, many cell types change in culture, often preserving some but not all of their in vivo characteristics in culture. At least some of the in vivo microenvironment may be mimicked by providing specific cues to cultured cells. Here, we show that after isolation and during maintenance in culture, adherent rat islets reduce expression of key ß-cell transcription factors necessary for ß-cell function and that soluble pancreatic decellularized matrix (DCM) can enhance ß-cell gene expression. Following chromatographic fractionation of pancreatic DCM, we performed proteomics to identify soluble factors that can maintain ß-cell stability and function. We identified Apolipoprotein E (ApoE) as an extracellular protein that significantly increased the expression of key ß-cell genes. The ApoE effect on beta cells was mediated at least in part through the JAK/STAT signaling pathway. Together, these results reveal a role for ApoE as an extracellular factor that can maintain the mature ß-cell gene expression profile.


Asunto(s)
Apolipoproteínas E/metabolismo , Espacio Extracelular/metabolismo , Regulación de la Expresión Génica/fisiología , Células Secretoras de Insulina/metabolismo , Animales , Células Cultivadas , Proteoglicanos de Heparán Sulfato/metabolismo , Islotes Pancreáticos/metabolismo , Quinasas Janus/metabolismo , Proteoma , Proteómica , Ratas Sprague-Dawley , Receptores de LDL/metabolismo , Factores de Transcripción STAT/metabolismo , Técnicas de Cultivo de Tejidos
2.
J Biomed Opt ; 14(4): 044029, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19725740

RESUMEN

Cardiac architecture is inherently three-dimensional, yet most characterizations rely on two-dimensional histological slices or dissociated cells, which remove the native geometry of the heart. We previously developed a method for labeling intact heart sections without dissociation and imaging large volumes while preserving their three-dimensional structure. We further refine this method to permit quantitative analysis of imaged sections. After data acquisition, these sections are assembled using image-processing tools, and qualitative and quantitative information is extracted. By examining the reconstructed cardiac blocks, one can observe end-to-end adjacent cardiac myocytes (cardiac strands) changing cross-sectional geometries, merging and separating from other strands. Quantitatively, representative cross-sectional areas typically used for determining hypertrophy omit the three-dimensional component; we show that taking orientation into account can significantly alter the analysis. Using fast-Fourier transform analysis, we analyze the gross organization of cardiac strands in three dimensions. By characterizing cardiac structure in three dimensions, we are able to determine that the alpha crystallin mutation leads to hypertrophy with cross-sectional area increases, but not necessarily via changes in fiber orientation distribution.


Asunto(s)
Algoritmos , Anatomía Transversal/métodos , Corazón/anatomía & histología , Interpretación de Imagen Asistida por Computador/métodos , Imagenología Tridimensional/métodos , Microscopía de Fluorescencia por Excitación Multifotónica/métodos , Microtomía/métodos , Animales , Aumento de la Imagen/métodos , Ratones , Ratones Transgénicos , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
3.
Circulation ; 117(15): 1936-44, 2008 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-18378613

RESUMEN

BACKGROUND: ST2 is a member of the interleukin-1 receptor family with a soluble form that is markedly upregulated on application of biomechanical strain to cardiac myocytes. Circulating ST2 levels are elevated in the setting of acute myocardial infarction, but the predictive value of ST2 independent of traditional clinical factors and of an established biomarker of biomechanical strain, N-terminal prohormone B-type natriuretic peptide (NT-proBNP), has not been established. METHODS AND RESULTS: We measured ST2 at baseline in 1239 patients with ST-elevation myocardial infarction from the CLopidogrel as Adjunctive ReperfusIon TherapY-Thrombolysis in Myocardial Infarction 28 (CLARITY-TIMI 28) trial. Per trial protocol, patients were to undergo coronary angiography after 2 to 8 days and were followed up for 30 days for clinical events. In contrast to NT-proBNP, ST2 levels were independent of clinical factors potentially related to chronic increased left ventricular wall stress, including age, hypertension, prior myocardial infarction, and prior heart failure; levels also were only modestly correlated with NT-proBNP (r=0.14). After adjustment for baseline characteristics and NT-proBNP levels, an ST2 level above the median was associated with a significantly greater risk of cardiovascular death or heart failure (third quartile: adjusted odds ratio, 1.42; 95% confidence interval, 0.68 to 3.57; fourth quartile: adjusted odds ratio, 3.57; 95% confidence interval, 1.87 to 6.81; P<0.0001 for trend). When both ST2 and NT-proBNP were added to a model containing traditional clinical predictors, the c statistic significantly improved from 0.82 (95% confidence interval, 0.77 to 0.87) to 0.86 (95% confidence interval, 0.81 to 0.90) (P=0.017). CONCLUSIONS: In ST-elevation myocardial infarction, high baseline ST2 levels are a significant predictor of cardiovascular death and heart failure independently of baseline characteristics and NT-proBNP, and the combination of ST2 and NT-proBNP significantly improves risk stratification. These data highlight the prognostic value of multiple, complementary biomarkers of biomechanical strain in ST-elevation myocardial infarction.


Asunto(s)
Electrocardiografía , Infarto del Miocardio/sangre , Péptido Natriurético Encefálico/sangre , Fragmentos de Péptidos/sangre , Receptores de Superficie Celular/sangre , Anciano , Biomarcadores , Enfermedades Cardiovasculares/sangre , Enfermedades Cardiovasculares/diagnóstico por imagen , Enfermedades Cardiovasculares/mortalidad , Comorbilidad , Angiografía Coronaria , Femenino , Estudios de Seguimiento , Insuficiencia Cardíaca/sangre , Insuficiencia Cardíaca/epidemiología , Insuficiencia Cardíaca/etiología , Humanos , Proteína 1 Similar al Receptor de Interleucina-1 , Masculino , Persona de Mediana Edad , Infarto del Miocardio/fisiopatología , Miocitos Cardíacos/fisiología , Ensayos Clínicos Controlados Aleatorios como Asunto/estadística & datos numéricos , Estrés Mecánico , Análisis de Supervivencia
4.
J Histotechnol ; 31(3): 129-134, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20622939

RESUMEN

One of the challenges in labeling tissues for fluorescence microscopy is minimizing sample processing while maintaining or improving the information generated by the fluorescent label. Generally, tissues are extracted, fixed, and embedded in mounting media (such as paraffin), sectioned, and then postprocessed by removing the paraffin, blocking, labeling, and washing. Despite all of these steps, the consistency of labeling quality can vary as a result of several factors, including heterogeneity in labeling efficiency from slide to slide, the necessity of postprocessing to obtain information on sequential sections of tissue, interference from the mounting media, and loss of native three-dimensional structural information, especially in thicker sections. A method for embedding and processing tissues that have been labeled by intravital staining is described in this study. Intravital staining is the process in which live-cell dyes and other labels are injected into the bloodstream before fixation of the tissues. Tissues processed this way can be imaged upon sectioning without further staining and retain their native, three-dimensional information, thereby improving the information retained by the labels and speeding up sample processing.

5.
Circ Res ; 101(12): 1328-38, 2007 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-17916779

RESUMEN

Biomechanical overload induces cardiac hypertrophy and heart failure, and reactive oxygen species (ROS) play a role in both processes. Thioredoxin-Interacting Protein (Txnip) is encoded by a mechanically-regulated gene that controls cell growth and apoptosis in part through interaction with the endogenous dithiol antioxidant thioredoxin. Here we show that Txnip is a critical regulator of the cardiac response to pressure overload. We generated inducible cardiomyocyte-specific and systemic Txnip-null mice (Txnip-KO) using Flp/frt and Cre/loxP technologies. Compared with littermate controls, Txnip-KO hearts had attenuated cardiac hypertrophy and preserved left ventricular (LV) contractile reserve through 4 weeks of pressure overload; however, the beneficial effects were not sustained and Txnip deletion ultimately led to maladaptive LV remodeling at 8 weeks of pressure overload. Interestingly, these effects of Txnip deletion on cardiac performance were not accompanied by global changes in thioredoxin activity or ROS; instead, Txnip-KO hearts had a robust increase in myocardial glucose uptake. Thus, deletion of Txnip plays an unanticipated role in myocardial energy homeostasis rather than redox regulation. These results support the emerging concept that the function of Txnip is not as a simple thioredoxin inhibitor but as a metabolic control protein.


Asunto(s)
Presión Sanguínea/genética , Cardiomegalia/genética , Cardiomegalia/fisiopatología , Proteínas Portadoras/genética , Eliminación de Gen , Marcación de Gen , Tiorredoxinas/genética , Animales , Cardiomegalia/metabolismo , Proteínas Portadoras/fisiología , Femenino , Marcación de Gen/métodos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Tiorredoxinas/fisiología , Remodelación Ventricular/fisiología
6.
Circulation ; 116(15): 1683-92, 2007 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-17875967

RESUMEN

BACKGROUND: Local delivery of chemotactic factors represents a novel approach to tissue regeneration. However, successful chemokine protein delivery is challenged by barriers including the rapid diffusion of chemokines and cleavage of chemokines by proteases that are activated in injured tissues. Stromal cell-derived factor-1 (SDF-1) is a well-characterized chemokine for attracting stem cells and thus a strong candidate for promoting regeneration. However, SDF-1 is cleaved by exopeptidases and matrix metalloproteinase-2, generating a neurotoxin implicated in some forms of dementia. METHODS AND RESULTS: We designed a new chemokine called S-SDF-1(S4V) that is resistant to matrix metalloproteinase-2 and exopeptidase cleavage but retains chemotactic bioactivity, reducing the neurotoxic potential of native SDF-1. To deliver S-SDF-1(S4V), we expressed and purified fusion proteins to tether the chemokine to self-assembling peptides, which form nanofibers and allow local delivery. Intramyocardial delivery of S-SDF-1(S4V) after myocardial infarction recruited CXCR4+/c-Kit+ stem cells (46+/-7 to 119+/-18 cells per section) and increased capillary density (from 169+/-42 to 283+/-27 per 1 mm2). Furthermore, in a randomized, blinded study of 176 rats with myocardial infarction, nanofiber delivery of the protease-resistant S-SDF-1(S4V) improved cardiac function (ejection fraction increased from 34.0+/-2.5% to 50.7+/-3.1%), whereas native SDF-1 had no beneficial effects. CONCLUSIONS: The combined advances of a new, protease-resistant SDF-1 and nanofiber-mediated delivery promoted recruitment of stem cells and improved cardiac function after myocardial infarction. These data demonstrate that driving chemotaxis of stem cells by local chemokine delivery is a promising new strategy for tissue regeneration.


Asunto(s)
Quimiocinas CXC/genética , Metaloproteinasa 2 de la Matriz/metabolismo , Infarto del Miocardio/fisiopatología , Neovascularización Fisiológica , Células Madre/fisiología , Animales , Presión Sanguínea , Arterias Carótidas/fisiología , Quimiocina CXCL12 , Quimiocinas CXC/aislamiento & purificación , Quimiocinas CXC/farmacología , Dipeptidil Peptidasa 4/metabolismo , Resistencia a Medicamentos , Movilización de Célula Madre Hematopoyética , Masculino , Mutación , Péptido Hidrolasas/metabolismo , Plásmidos , Ratas , Ratas Sprague-Dawley , Regeneración
7.
Nat Med ; 13(8): 970-4, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17660827

RESUMEN

An emerging concept is that the mammalian myocardium has the potential to regenerate, but that regeneration might be too inefficient to repair the extensive myocardial injury that is typical of human disease. However, the degree to which stem cells or precursor cells contribute to the renewal of adult mammalian cardiomyocytes remains controversial. Here we report evidence that stem cells or precursor cells contribute to the replacement of adult mammalian cardiomyocytes after injury but do not contribute significantly to cardiomyocyte renewal during normal aging. We generated double-transgenic mice to track the fate of adult cardiomyocytes in a 'pulse-chase' fashion: after a 4-OH-tamoxifen pulse, green fluorescent protein (GFP) expression was induced only in cardiomyocytes, with 82.7% of cardiomyocytes expressing GFP. During normal aging up to one year, the percentage of GFP+ cardiomyocytes remained unchanged, indicating that stem or precursor cells did not refresh uninjured cardiomyocytes at a significant rate during this period of time. By contrast, after myocardial infarction or pressure overload, the percentage of GFP+ cardiomyocytes decreased from 82.8% in heart tissue from sham-treated mice to 67.5% in areas bordering a myocardial infarction, 76.6% in areas away from a myocardial infarction, and 75.7% in hearts subjected to pressure overload, indicating that stem cells or precursor cells had refreshed the cardiomyocytes.


Asunto(s)
Envejecimiento/fisiología , Linaje de la Célula , Lesiones Cardíacas/genética , Lesiones Cardíacas/patología , Miocitos Cardíacos/citología , Miocitos Cardíacos/patología , Células Madre/citología , Células Madre/metabolismo , Animales , Proliferación Celular , Perfilación de la Expresión Génica , Ratones , Ratones Transgénicos , Miocitos Cardíacos/metabolismo
8.
Circulation ; 114(7): 637-44, 2006 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-16894033

RESUMEN

BACKGROUND: Local delivery methods can target therapies to specific tissues and potentially avoid toxicity to other organs. Platelet-derived growth factor can protect the myocardium, but it also plays an important role in promoting pulmonary hypertension. It is not known whether local myocardial delivery of platelet-derived growth factor during myocardial infarction (MI) can lead to sustained cardiac benefit without causing pulmonary hypertension. METHODS AND RESULTS: We performed a randomized and blinded experiment of 127 rats that survived experimental MI or sham surgery. We delivered platelet-derived growth factor (PDGF)-BB with self-assembling peptide nanofibers (NFs) to provide controlled release within the myocardium. There were 6 groups with n > or = 20 in each group: sham, sham+NF, sham+NF/PDGF, MI, MI+NF, and MI+NF/PDGF. Serial echocardiography from 1 day to 3 months showed significant improvement of ventricular fractional shortening, end-systolic dimension, and end-diastolic dimension with local PDGF delivery (P < 0.05 for MI+NF/PDGF versus MI or MI+NF). Catheterization at 4 months revealed improved ventricular function in the controlled delivery group (left ventricular end-diastolic pressure, cardiac index, +dP/dt, -dP/dt, and time constant of exponential decay all P < 0.05 for MI+NF/P versus MI or MI+NF). Infarcted myocardial volume was reduced by NF/PDGF therapy (34.0 +/- 13.3% in MI, 28.9 +/- 12.9% in MI+NF, and 12.0 +/- 5.8% in MI+NF/PDGF; P < 0.001). There was no evidence of pulmonary toxicity from the therapy, with no differences in right ventricular end-systolic pressure, right ventricular dP/dt, bromodeoxyuridine staining, or pulmonary artery medial wall thickness. CONCLUSIONS: Intramyocardial delivery of PDGF by self-assembling peptide NFs leads to long-term improvement in cardiac performance after experimental infarction without apparent pulmonary toxicity. Local myocardial protection may allow prevention of heart failure without systemic toxicity.


Asunto(s)
Hipertensión Pulmonar/inducido químicamente , Hipertensión Pulmonar/prevención & control , Infarto del Miocardio/fisiopatología , Factor de Crecimiento Derivado de Plaquetas/administración & dosificación , Factor de Crecimiento Derivado de Plaquetas/farmacología , Función Ventricular/efectos de los fármacos , Animales , Sistemas de Liberación de Medicamentos , Ecocardiografía , Hemodinámica/efectos de los fármacos , Hemodinámica/fisiología , Hipertensión Pulmonar/fisiopatología , Inyecciones/métodos , Pulmón/efectos de los fármacos , Pulmón/fisiopatología , Masculino , Infarto del Miocardio/tratamiento farmacológico , Nanotecnología , Factor de Crecimiento Derivado de Plaquetas/efectos adversos , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Flujo Sanguíneo Regional/efectos de los fármacos , Función Ventricular/fisiología
9.
Am J Physiol Heart Circ Physiol ; 290(1): H232-9, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16126817

RESUMEN

Matrix metalloproteinases (MMPs) are postulated to be necessary for neovascularization during wound healing. MMP-9 deletion alters remodeling postmyocardial infarction (post-MI), but whether and to what degree MMP-9 affects neovascularization post-MI is unknown. Neovascularization was evaluated in wild-type (WT; n = 63) and MMP-9 null (n = 55) mice at 7-days post-MI. Despite similar infarct sizes, MMP-9 deletion improved left ventricular function as evaluated by hemodynamic analysis. Blood vessel quantity and quality were evaluated by three independent studies. First, vessel density was increased in the infarct of MMP-9 null mice compared with WT, as quantified by Griffonia (Bandeiraea) simplicifolia lectin I (GSL-I) immunohistochemistry. Second, preexisting vessels, stained in vivo with FITC-labeled GSL-I pre-MI, were present in the viable but not MI region. Third, a technetium-99m-labeled peptide (NC100692), which selectively binds to activated alpha(v)beta3-integrin in angiogenic vessels, was injected into post-MI mice. Relative NC100692 activity in myocardial segments with diminished perfusion (0-40% nonischemic) was higher in MMP-9 null than in WT mice (383 +/- 162% vs. 250 +/- 118%, respectively; P = 0.002). The unique finding of this study was that MMP-9 deletion stimulated, rather than impaired, neovascularization in remodeling myocardium. Thus targeted strategies to inhibit MMP-9 early post-MI will likely not impair the angiogenic response.


Asunto(s)
Eliminación de Gen , Metaloproteinasa 9 de la Matriz/genética , Infarto del Miocardio/fisiopatología , Neovascularización Fisiológica/fisiología , Animales , Vasos Coronarios/patología , Ligadura , Macrófagos/patología , Metaloproteinasa 9 de la Matriz/deficiencia , Ratones , Ratones Noqueados , Infarto del Miocardio/patología , Remodelación Ventricular/fisiología
10.
J Clin Invest ; 116(1): 237-48, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16357943

RESUMEN

Endothelial cells can protect cardiomyocytes from injury, but the mechanism of this protection is incompletely described. Here we demonstrate that protection of cardiomyocytes by endothelial cells occurs through PDGF-BB signaling. PDGF-BB induced cardiomyocyte Akt phosphorylation in a time- and dose-dependent manner and prevented apoptosis via PI3K/Akt signaling. Using injectable self-assembling peptide nanofibers, which bound PDGF-BB in vitro, sustained delivery of PDGF-BB to the myocardium at the injected sites for 14 days was achieved. A blinded and randomized study in 96 rats showed that injecting nanofibers with PDGF-BB, but not nanofibers or PDGF-BB alone, decreased cardiomyocyte death and preserved systolic function after myocardial infarction. A separate blinded and randomized study in 52 rats showed that PDGF-BB delivered with nanofibers decreased infarct size after ischemia/reperfusion. PDGF-BB with nanofibers induced PDGFR-beta and Akt phosphorylation in cardiomyocytes in vivo. These data demonstrate that endothelial cells protect cardiomyocytes via PDGF-BB signaling and that this in vitro finding can be translated into an effective in vivo method of protecting myocardium after infarction. Furthermore, this study shows that injectable nanofibers allow precise and sustained delivery of proteins to the myocardium with potential therapeutic benefits.


Asunto(s)
Endotelio Vascular/fisiología , Corazón/fisiología , Factor de Crecimiento Derivado de Plaquetas/farmacología , Animales , Apoptosis/efectos de los fármacos , Becaplermina , Sistemas de Liberación de Medicamentos , Endotelio Vascular/efectos de los fármacos , Corazón/efectos de los fármacos , Sistema de Conducción Cardíaco , Células Musculares/citología , Células Musculares/efectos de los fármacos , Células Musculares/fisiología , Miocardio/citología , Fibras Nerviosas/efectos de los fármacos , Fibras Nerviosas/fisiología , Factor de Crecimiento Derivado de Plaquetas/administración & dosificación , Factor de Crecimiento Derivado de Plaquetas/uso terapéutico , Proteínas Proto-Oncogénicas c-sis , Ratas
11.
Proc Natl Acad Sci U S A ; 102(30): 10622-7, 2005 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-16020536

RESUMEN

Growth factor signaling can affect tissue remodeling through autocrine/paracrine mechanisms. Recent evidence indicates that EGF receptor transactivation by heparin-binding EGF (HB-EGF) contributes to hypertrophic signaling in cardiomyocytes. Here, we show that HB-EGF operates in a spatially restricted circuit in the extracellular space within the myocardium, revealing the critical nature of the local microenvironment in intercellular signaling. This highly localized microenvironment of HB-EGF signaling was demonstrated with 3D morphology, consistent with predictions from a computational model of EGF signaling. HB-EGF secretion by a given cardiomyocyte in mouse left ventricles led to cellular hypertrophy and reduced expression of connexin43 in the overexpressing cell and in immediately adjacent cells but not in cells farther away. Thus, HB-EGF acts as an autocrine and local paracrine cardiac growth factor that leads to loss of gap junction proteins within a spatially confined microenvironment. These findings demonstrate how cells can coordinate remodeling with their immediate neighboring cells with highly localized extracellular EGF signaling.


Asunto(s)
Conexina 43/metabolismo , Factor de Crecimiento Epidérmico/metabolismo , Hipertrofia Ventricular Izquierda/metabolismo , Miocardio/citología , Transducción de Señal , Remodelación Ventricular/fisiología , Análisis de Varianza , Animales , Northern Blotting , Western Blotting , Cartilla de ADN , Ensayo de Inmunoadsorción Enzimática , Receptores ErbB/metabolismo , Técnicas de Transferencia de Gen , Heparina/metabolismo , Inmunohistoquímica , Miocardio/metabolismo , Ratas , Ratas Sprague-Dawley
12.
Am J Physiol Heart Circ Physiol ; 288(4): H1802-9, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15563531

RESUMEN

This study compared the effects of rosuvastatin on left ventricular infarct size in mice after permanent coronary occlusion vs. 60 min of ischemia followed by 24 h of reperfusion. Statins can inhibit neutrophil adhesion, increase nitric oxide synthase (NOS) expression, and mobilize progenitor stem cells after ischemic injury. Mice received blinded and randomized administration of rosuvastatin (20 mg.kg(-1).day(-1)) or saline from 2 days before surgery until death. After 60 min of ischemia with reperfusion, infarct size was reduced by 18% (P = 0.03) in mice randomized to receive rosuvastatin (n = 18) vs. saline (n = 22) but was similar after permanent occlusion in rosuvastatin (n = 17) and saline (n = 20) groups (P = not significant). Myocardial infarct size after permanent left anterior descending coronary artery occlusion (n = 6) tended to be greater in NOS3-deficient mice than in the wild-type saline group (33 +/- 4 vs. 23 +/- 2%, P = 0.08). Infarct size in NOS3-deficient mice was not modified by treatment with rosuvastatin (34 +/- 5%, n = 6, P = not significant vs. NOS3-deficient saline group). After 60 min of ischemia-reperfusion, neutrophil infiltration was similar in rosuvastatin and saline groups as was the percentage of CD34(+), Sca-1(+), and c-Kit(+) cells. Left ventricular NOS3 mRNA and protein levels were unchanged by rosuvastatin. Rosuvastatin reduces infarct size after 60 min of ischemia-reperfusion but not after permanent coronary occlusion, suggesting a potential anti-inflammatory effect. Although we were unable to demonstrate that the myocardial protection was due to an effect on neutrophil infiltration, stem cell mobilization, or induction of NOS3, these data suggest that rosuvastatin may be particularly beneficial in myocardial protection after ischemia-reperfusion injury.


Asunto(s)
Fluorobencenos/farmacología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Infarto del Miocardio/tratamiento farmacológico , Infarto del Miocardio/patología , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Daño por Reperfusión Miocárdica/patología , Pirimidinas/farmacología , Sulfonamidas/farmacología , Animales , Enfermedad Coronaria/tratamiento farmacológico , Enfermedad Coronaria/patología , Ventrículos Cardíacos/patología , Movilización de Célula Madre Hematopoyética , Ligadura , Masculino , Ratones , Ratones Endogámicos C57BL , Miocardio/patología , Óxido Nítrico Sintasa/genética , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico Sintasa de Tipo II , Óxido Nítrico Sintasa de Tipo III , ARN Mensajero/análisis , Rosuvastatina Cálcica , Células Madre/efectos de los fármacos
13.
Circulation ; 109(21): 2581-6, 2004 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-15123525

RESUMEN

BACKGROUND: Although cellular redox balance plays an important role in mechanically induced cardiac hypertrophy, the mechanisms of regulation are incompletely defined. Because thioredoxin is a major intracellular antioxidant and can also regulate redox-dependent transcription, we explored the role of thioredoxin activity in mechanically overloaded cardiomyocytes in vitro and in vivo. METHODS AND RESULTS: Overexpression of thioredoxin induced protein synthesis in cardiomyocytes (127+/-5% of controls, P<0.01). Overexpression of thioredoxin-interacting protein (Txnip), an endogenous thioredoxin inhibitor, reduced protein synthesis in response to mechanical strain (89+/-5% reduction, P<0.01), phenylephrine (80+/-3% reduction, P<0.01), or angiotensin II (80+/-4% reduction, P<0.01). In vivo, myocardial thioredoxin activity increased 3.5-fold compared with sham controls after transverse aortic constriction (P<0.01). Aortic constriction did not change thioredoxin expression but reduced Txnip expression by 40% (P<0.05). Gene transfer studies showed that cells that overexpress Txnip develop less hypertrophy after aortic constriction than control cells in the same animals (28.1+/-5.2% reduction versus noninfected cells, P<0.01). CONCLUSIONS: Thus, even though thioredoxin is an antioxidant, activation of thioredoxin participates in the development of pressure-overload cardiac hypertrophy, demonstrating the dual function of thioredoxin as both an antioxidant and a signaling protein. These results also support the emerging concept that the thioredoxin inhibitor Txnip is a critical regulator of biomechanical signaling.


Asunto(s)
Cardiomegalia/metabolismo , Proteínas Portadoras/fisiología , Corazón/efectos de los fármacos , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Tiorredoxinas/metabolismo , Angiotensina II/farmacología , Animales , Enfermedades de la Aorta/complicaciones , Cardiomegalia/etiología , Cardiomegalia/genética , Proteínas Portadoras/genética , Proteínas de Ciclo Celular , Tamaño de la Célula , Células Cultivadas/efectos de los fármacos , Células Cultivadas/metabolismo , Células Cultivadas/patología , Constricción Patológica/complicaciones , Modelos Animales de Enfermedad , Vectores Genéticos/genética , Vectores Genéticos/farmacología , Ligadura , Masculino , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Oxidación-Reducción , Fenilefrina/farmacología , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno , Transducción de Señal , Método Simple Ciego , Estrés Mecánico , Tiorredoxinas/genética , Activación Transcripcional/efectos de los fármacos , Activación Transcripcional/fisiología
14.
Circ Res ; 93(3): 238-45, 2003 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-12855673

RESUMEN

Matrix metalloproteinase-mediated degradation of type I collagen may play a role in cardiac remodeling after strain or injury. To explore this hypothesis, we used mice homozygous (r/r) for a targeted mutation in Col1a1; these mice synthesize collagen I that resists collagenase cleavage at Gly975-Leu976. A total of 64 r/r and 84 littermate wild-type mice (WT) underwent experimental pressure overload by transverse aortic constriction (TAC) or myocardial infarction (MI). Echocardiographic, hemodynamic, and histological parameters were evaluated up to 12 weeks after TAC or 21 days after MI. At 4 weeks after TAC, collagen levels, wall thickness, and echocardiographic parameters were similar in the 2 groups. At 12 weeks after TAC, r/r mice had smaller LV dimensions (ESD: 2.7+/-0.2 mm WT versus 1.7+/-0.2 mm r/r, P<0.013; EDD: 3.8+/-0.2 mm WT versus 3.1+/-0.1 mm r/r, P<0.013); better fractional shortening (30+/-2% WT versus 46+/-4% r/r; P<0.013); and lower LV/body weight ratios (7.3+/-0.6 WT and 5.1+/-0.5 r/r; P<0.013). Surprisingly, these differences were not accompanied by differences in collagen accumulation, myocyte cross-sectional areas, wall thickness, or microvessel densities. Furthermore, no differences in LV remodeling assessed by echocardiography, fibrosis, or hemodynamic parameters were found between r/r and WT mice after MI. Thus, a mutation that encodes a collagenase cleavage-resistant collagen I does not affect early LV remodeling after TAC or MI, suggesting that collagen cleavage at this site is not the mechanism by which metalloproteinases mediate LV remodeling. Collagen cleavage could, however, have a role in preservation of cardiac function in late remodeling by mechanisms independent of collagen accumulation. We were not able to detect collagen cleavage fragments, and could not, therefore, rule out the possibility of collagen cleavage at additional sites.


Asunto(s)
Colágeno/genética , Mutación , Miocardio/patología , Remodelación Ventricular/genética , Animales , Aorta/patología , Cardiomegalia/genética , Cardiomegalia/patología , Colágeno/metabolismo , Constricción Patológica/patología , Cruzamientos Genéticos , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Ecocardiografía , Endopeptidasas/metabolismo , Fibrosis/complicaciones , Fibrosis/patología , Hemodinámica , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Infarto del Miocardio/complicaciones , Infarto del Miocardio/patología , Miocardio/metabolismo , Tasa de Supervivencia
15.
Circulation ; 106(23): 2961-6, 2002 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-12460879

RESUMEN

BACKGROUND: We identified an interleukin-1 receptor family member, ST2, as a gene markedly induced by mechanical strain in cardiac myocytes and hypothesized that ST2 participates in the acute myocardial response to stress and injury. METHODS AND RESULTS: ST2 mRNA was induced in cardiac myocytes by mechanical strain (4.7+/-0.9-fold) and interleukin-1beta (2.0+/-0.2-fold). Promoter analysis revealed that the proximal and not the distal promoter of ST2 is responsible for transcriptional activation in cardiac myocytes by strain and interleukin-1beta. In mice subjected to coronary artery ligation, serum ST2 was transiently increased compared with unoperated controls (20.8+/-4.4 versus 0.8+/-0.8 ng/mL, P<0.05). Soluble ST2 levels were increased in the serum of human patients (N=69) 1 day after myocardial infarction and correlated positively with creatine kinase (r=0.41, P<0.001) and negatively with ejection fraction (P=0.02). CONCLUSIONS: These data identify ST2 release in response to myocardial infarction and suggest a role for this innate immune receptor in myocardial injury.


Asunto(s)
Proteínas de la Membrana/metabolismo , Infarto del Miocardio/metabolismo , Miocardio/metabolismo , Receptores de Interleucina-1/metabolismo , Angiotensina II/farmacología , Antagonistas de Receptores de Angiotensina , Animales , Animales Recién Nacidos , Biomarcadores/sangre , Células Cultivadas , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Humanos , Imidazoles/farmacología , Interleucina-1/farmacología , Proteína 1 Similar al Receptor de Interleucina-1 , Interleucina-4/farmacología , Lipopolisacáridos/farmacología , Proteínas de la Membrana/sangre , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Infarto del Miocardio/sangre , Miocardio/citología , Ensayos de Protección de Nucleasas , Ésteres del Forbol/farmacología , Regiones Promotoras Genéticas/efectos de los fármacos , Regiones Promotoras Genéticas/fisiología , Piridinas/farmacología , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor de Angiotensina Tipo 1 , Receptores de Superficie Celular , Receptores de Interleucina , Receptores de Interleucina-1/sangre , Receptores de Interleucina-1/genética , Estrés Mecánico , Volumen Sistólico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...