Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 193
Filtrar
1.
J Cardiovasc Transl Res ; 7(5): 475-82, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24831573

RESUMEN

Stem cell therapy is a promising strategy in promoting cardiac repair in the setting of ischemic heart disease. Clinical and preclinical studies have shown that cell therapy improves cardiac function. Whether autologous or allogeneic cells should be used, and the need for immunosuppression in non-autologous settings, is a matter of debate. Cyclosporin A (CsA) is frequently used in preclinical trials to reduce cell rejection after non-autologous cell therapy. The direct effect of CsA on the function and survival of stem cells is unclear. Furthermore, the appropriate daily dosage of CsA in animal models has not been established. In this review, we discuss the pros and cons of the use of CsA on an array of stem cells both in vitro and in vivo. Furthermore, we present a small collection of data put forth by our group supporting the efficacy and safety of a specific daily CsA dosage in a pig model.


Asunto(s)
Ciclosporina/administración & dosificación , Supervivencia de Injerto/efectos de los fármacos , Cardiopatías/cirugía , Inmunosupresores/administración & dosificación , Regeneración/efectos de los fármacos , Trasplante de Células Madre , Células Madre/efectos de los fármacos , Animales , Supervivencia Celular/efectos de los fármacos , Ciclosporina/efectos adversos , Modelos Animales de Enfermedad , Esquema de Medicación , Cardiopatías/inmunología , Cardiopatías/patología , Cardiopatías/fisiopatología , Humanos , Inmunosupresores/efectos adversos , Trasplante de Células Madre/efectos adversos , Células Madre/inmunología , Porcinos , Factores de Tiempo , Resultado del Tratamiento
2.
Nutr Metab Cardiovasc Dis ; 24(4): 428-33, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24370448

RESUMEN

BACKGROUND AND AIMS: Omega-3 fatty acids suppress Thromboxane A(2) (TxA(2)) generation via mechanisms independent to that of aspirin therapy. We sought to evaluate whether baseline omega-3 fatty acid levels influence arachidonic acid proven platelet-cyclooxygenase-1 (COX-1) independent TxA(2) generation (TxA(2) generation despite adequate aspirin use). METHODS AND RESULTS: Subjects with acute myocardial infarction, stable CVD or at high risk for CVD, on adequate aspirin therapy were included in this study. Adequate aspirin action was defined as complete inhibition of platelet-COX-1 activity as assessed by <10% change in light transmission aggregometry to ≥1 mmol/L arachidonic acid. TxA(2) production was measured via liquid chromatography-tandem mass spectrometry for the stable TxA(2) metabolite 11-dehydro-thromboxane B2 (UTxB2) in urine. The relationship between baseline fatty acids, demographics and UTxB(2) were evaluated. Baseline omega-3 fatty acid levels were not associated with UTxB(2) concentration. However, smoking was associated with UTxB(2) in this study. CONCLUSION: Baseline omega-3 fatty acid levels do not influence TxA(2) generation in patients with or at high risk for CVD receiving adequate aspirin therapy. The association of smoking and TxA(2) generation, in the absence of platelet COX-1 activity, among aspirin treated patients warrants further study.


Asunto(s)
Aspirina/uso terapéutico , Plaquetas/efectos de los fármacos , Enfermedades Cardiovasculares/tratamiento farmacológico , Ciclooxigenasa 1/sangre , Inhibidores de la Ciclooxigenasa/uso terapéutico , Ácidos Grasos Omega-3/sangre , Tromboxano A2/sangre , Anciano , Biomarcadores/sangre , Biomarcadores/orina , Plaquetas/enzimología , Enfermedades Cardiovasculares/sangre , Enfermedades Cardiovasculares/enzimología , Cromatografía Liquida , Femenino , Humanos , Masculino , Persona de Mediana Edad , Agregación Plaquetaria/efectos de los fármacos , Pruebas de Función Plaquetaria , Fumar/efectos adversos , Fumar/sangre , Fumar/orina , Espectrometría de Masas en Tándem , Tromboxano B2/análogos & derivados , Tromboxano B2/orina
3.
Minerva Cardioangiol ; 61(6): 605-16, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24253454

RESUMEN

Early results from stem cell trials to treat myocardial infarction have shown promise. Several types of stem cells have moved through phase I trials to demonstrate safety and some at the same time have shown significant potential for myocardial regeneration and functional recovery. The means by which stem cells contribute to improving myocardial function, however, remains unknown. Challenges in labeling stem cells for tracking and fate determination after cell transplantation have precluded establishing whether transplanted stem cell engraftment, expansion after engraftment, endogenous stem cell activation or a combination of these mechanisms contribute to improved function. Cardiac magnetic resonance imaging (cMRI), due to its inherent capabilities, has emerged as the imaging modality of choice to provide important insights into remodeling of myocardium after stem cell transplantation and its consequences on cardiac function. Of cMRI capabilities, excellent spatial resolution is instrumental in assessment of global and regional function and feasibility of scar quantification sets it apart from other imaging modalities and facilitates critical analysis. These capabilities permit the identification of dysfunctional myocardium and scar and changes in these regions over time. The effect of stem cell therapeutics on dysfunctional myocardium and scar can then be highlighted in longitudinal assessment in clinical trials. This has been demonstrated in the inaugural Phase I SCIPIO trial where patients received autologous C-kitPos cardiac stem cell (CSC) transplantation. Although the global function improved significantly with CSC transplantation, regional/segmental analysis provided crucial insights into the effects of CSCs on the most dysfunctional myocardial segments. Magnetic resonance imaging is also a contending and complementing modality in molecular imaging essential for mechanistic studies.


Asunto(s)
Imagen por Resonancia Magnética/métodos , Infarto del Miocardio/terapia , Trasplante de Células Madre/métodos , Animales , Ensayos Clínicos Fase I como Asunto , Humanos , Infarto del Miocardio/fisiopatología , Miocardio/metabolismo , Miocardio/patología , Recuperación de la Función , Regeneración , Células Madre/citología , Remodelación Ventricular
4.
Scand J Immunol ; 74(1): 31-41, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21338382

RESUMEN

Polyvalent Ig preparations, derived from the pooled plasma of thousands of healthy donors, contain a complex mix of both 'acquired' and natural antibodies directed against pathogens as well as foreign and self/auto antigens (Ag). Depending on their formulation, donor pool size, etc., liquid Ig preparations contain monomeric and dimeric IgG. The dimeric IgG fraction is thought to represent mainly idiotype-antiidiotype Ab pairs. Treatment of all IgG fractions at pH 4 effectively monomerizes the IgG dimers resulting in separated idiotype-antiidiotype Ab pairs and thus in a comparable F(ab')(2) binding site availability of the different IgG fractions. Previously, we identified an increased anti-self-reactivity within the monomerized dimer fraction. This study addressed if, among the different IgG fractions, an analogous preferential reactivity was evident in the response against different pathogen-derived protein and carbohydrate antigens. Therefore, we assessed the activity of total unseparated IgG, the monomeric and dimeric IgG fractions against antigenic structures of bacterial and viral antigens/virulence factors. All fractions showed similar reactivity to protein antigens except for exotoxin A of Pseudomonas aeruginosa, where the dimeric fraction, especially when monomerized, showed a marked increase in reactivity. This suggests that the production of antiidiotypic IgG antibodies contributes to controlling the immune response to certain categories of pathogens. In contrast, the monomeric IgG fractions showed increased reactivity towards pathogen-associated polysaccharides, classically regarded as T-independent antigens. Taken together, the differential reactivity of the IgG fractions seems to indicate a preferential segregation of antibody reactivities according to the nature of the antigen.


Asunto(s)
Anticuerpos Antibacterianos/inmunología , Anticuerpos Antivirales/inmunología , Reacciones Antígeno-Anticuerpo , Antígenos Bacterianos/inmunología , Antígenos Virales/inmunología , Inmunoglobulina G/inmunología , Anticuerpos Antibacterianos/química , Anticuerpos Antibacterianos/aislamiento & purificación , Anticuerpos Antivirales/química , Anticuerpos Antivirales/aislamiento & purificación , Toxinas Bacterianas/inmunología , Línea Celular , Humanos , Inmunoglobulina G/química , Inmunoglobulina G/aislamiento & purificación , Pruebas de Neutralización , Polisacáridos Bacterianos/inmunología , Multimerización de Proteína , Toxoides/inmunología
5.
Neth Heart J ; 17(5): 199-207, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19484156

RESUMEN

Myocardial infarction results in loss of cardiomyocytes, scar formation, ventricular remodelling, and eventually heart failure. In recent years, cell therapy has emerged as a potential new strategy for patients with ischaemic heart disease. This includes embryonic and bone marrow derived stem cells. Recent clinical studies showed ostensibly conflicting results of intracoronary infusion of autologous bone marrow derived stem cells in patients with acute or chronic myocardial infarction. Anyway, these results have stimulated additional clinical and pre-clinical studies to further enhance the beneficial effects of stem cell therapy. Recently, the existence of cardiac stem cells that reside in the heart itself was demonstrated. Their discovery has sparked intense hope for myocardial regeneration with cells that are obtained from the heart itself and are thereby inherently programmed to reconstitute cardiac tissue. These cells can be detected by several surface markers (e.g. c-kit, Sca-1, MDR1, Isl-1). Both in vitro and in vivo differentiation into cardiomyocytes, endothelial cells and vascular smooth muscle cells has been demonstrated, and animal studies showed promising results on improvement of left ventricular function. This review will discuss current views regarding the feasibility of cardiac repair, and focus on the potential role of the resident cardiac stem and progenitor cells. (Neth Heart J 2009;17:199-207.).

6.
Minerva Cardioangiol ; 53(6): 549-64, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16333238

RESUMEN

Myocardial infarction and other pathologic conditions of the heart result in loss of cardiomyocytes, scar formation, ventricular remodeling, and eventually heart failure. Since pharmacologic and interventional strategies fail to regenerate dead myocardium, heart failure continues to be a major health problem worldwide. Recent studies in animal models of myocardial infarction and heart failure have demonstrated that various subsets of adult primitive cells can regenerate functional cardiomyocytes and cardiac vasculature with improvement in cardiac structure and function. Small clinical trials of cell therapy in patients with myocardial infarction and ischemic cardiomyopathy have recapitulated these beneficial effects in humans with infarct size reduction and improvement in ejection fraction, myocardial perfusion, and wall motion. Several phenotypically distinct cell populations have been utilized for cardiac regeneration, and the relative merits of one cell over another remain to be determined. The recent discovery of adult cardiac stem cells has sparked intense hope for myocardial regeneration with cells that are from the heart itself and are thereby inherently programmed to reconstitute cardiac tissue. The purpose of this review is to summarize the evidence regarding the feasibility of cardiac repair in humans via adult stem/progenitor cells, and to discuss the potential utility of cardiac stem cells for therapeutic myocardial regeneration.


Asunto(s)
Infarto del Miocardio/terapia , Miocitos Cardíacos/trasplante , Regeneración , Trasplante de Células Madre , Adulto , Animales , Ensayos Clínicos como Asunto , Estudios de Factibilidad , Humanos , Isquemia Miocárdica/terapia , Miocardio/citología
7.
Am J Physiol Heart Circ Physiol ; 281(6): H2500-10, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11709417

RESUMEN

Recent investigations have established a role for the beta II-isoform of protein kinase C (PKC beta II) in the induction of cardiac hypertrophy and failure. Although receptors for activated C kinase (RACKs) have been shown to direct PKC signal transduction, the mechanism through which RACK1, a selective PKC beta II RACK, participates in PKC beta II-mediated cardiac hypertrophy and failure remains undefined. We have previously reported that PKC epsilon activation modulates the expression of RACKs, and that altered epsilon-isoform of PKC (PKC epsilon)-RACK interactions may facilitate the genesis of cardiac phenotypes in mice. Here, we present evidence that high levels of PKC epsilon activity are commensurate with impaired left ventricular function (dP/dt = 6,074 +/- 248 mmHg/s in control vs. 3,784 +/- 269 mmHg/s in transgenic) and significant myocardial hypertrophy. More importantly, we demonstrate that high levels of PKC epsilon activation induce a significant colocalization of PKC beta II with RACK1 (154 +/- 7% of control) and a marked redistribution of PKC beta II to the particulate fraction (17 +/- 2% of total PKC beta II in control mice vs. 49 +/- 5% of total PKC beta II in hypertrophied mice), without compensatory changes of the other eight PKC isoforms present in the mouse heart. This enhanced PKC beta II activation is coupled with increased RACK1 expression and PKC beta II-RACK1 interactions, demonstrating PKC epsilon-induced PKC beta II signaling via a RACK1-dependent mechanism. Taken together with our previous findings regarding enhanced RACK1 expression and PKC epsilon-RACK1 interactions in the setting of cardiac hypertrophy and failure, these results suggest that RACK1 serves as a nexus for at least two isoforms of PKC, the epsilon-isoform and the beta II-isoform, thus coordinating PKC-mediated hypertrophic signaling.


Asunto(s)
Insuficiencia Cardíaca/metabolismo , Isoenzimas/genética , Isoenzimas/metabolismo , Miocardio/enzimología , Péptidos/metabolismo , Proteína Quinasa C/genética , Proteína Quinasa C/metabolismo , Actinas/genética , Animales , Cardiomegalia/metabolismo , Expresión Génica/fisiología , Humanos , Células Jurkat , Ratones , Ratones Transgénicos , Mutagénesis/fisiología , Contracción Miocárdica/fisiología , Cadenas Pesadas de Miosina/genética , Mapeo de Interacción de Proteínas , Proteína Quinasa C beta , Proteína Quinasa C-epsilon , Receptores de Cinasa C Activada , Transducción de Señal/fisiología
9.
J Mol Cell Cardiol ; 33(11): 1897-918, 2001 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11708836

RESUMEN

Over the past decade, an enormous number of studies (>100) have focused on the role of nitric oxide (NO) in myocardial ischemia. It is important to distinguish the function of NO in unstressed (non-preconditioned) myocardium from its function in preconditioned myocardium (i.e. myocardium that has shifted to a defensive phenotype in response to stress). Of the 92 studies that have examined the role of NO in modulating the severity of ischemia/reperfusion injury in non-preconditioned myocardium, the vast majority [67 (73%)] have concluded that NO (either endogenous or exogenous) has a protective effect and only 11 (12%) found a detrimental effect. The proportion of studies supporting a cytoprotective role of NO is similar in vivo[35 (71%) out of 49] and in vitro[32 (74%) out of 43]. With regard to the delayed acquisition of tolerance to ischemia [late preconditioning (PC)], overwhelming evidence indicates a critical role of NO in this phenomenon. Specifically, enhanced biosynthesis of NO by eNOS is essential to trigger the late phase of ischemia-induced and exercise-induced PC, and enhanced NO production by iNOS is obligatorily required to mediate the anti-stunning and anti-infarct actions of late PC elicited by five different stimuli (ischemia, adenosine A1 agonists, opioid delta1 agonists, endotoxin derivatives and exercise). Thus, NO plays a dual role in the pathophysiology of the late phase of PC, acting initially as the trigger and subsequently as the mediator of this adaptive response ("NO hypothesis of late PC"). The diversity of the PC stimuli that converge on iNOS implies that the upregulation of this enzyme is a central mechanism whereby the myocardium protects itself from ischemia. The NO hypothesis of late PC has thus revealed a cytoprotective function of iNOS in the heart, a novel paradigm which has recently been extended to other tissues, including kidney and intestine. Other corollaries of this hypothesis are that the heart responds to stress in a biphasic manner, utilizing eNOS as an immediate but short-term response and iNOS as a delayed but long-term defense, and that the fundamental difference between non-preconditioned and late preconditioned myocardium is the tissue level of iNOS-derived NO, which is tonically higher in the latter compared with the former. Hence, late PC can be viewed as a state of enhanced NO synthesis. The NO hypothesis of late PC has important therapeutic implications. In experimental animals, administration of NO donors in lieu of ischemia can faithfully reproduce the molecular and functional aspects of ischemia-induced late PC, indicating that NO is not only necessary but also sufficient to induce late PC. The recent demonstration that nitroglycerin also induces late PC in patients provides proof-of-principle for the concept that nitrates could be used as a PC-mimetic therapy for the prophylaxis of ischemic injury in the clinical arena. This novel application of nitrates could be as important as, or perhaps even more important than, their current use as antianginal and preload-reducing agents. In addition, gene transfer of either eNOS or iNOS has been shown to replicate the infarct-sparing actions of ischemic PC, suggesting that NOS gene therapy could be an effective strategy for alleviating ischemia/reperfusion injury. Ten years of research have demonstrated that NO plays a fundamental biological role in protecting the heart against ischemia/reperfusion injury. The time has come to translate this enormous body of experimental evidence into clinically useful therapies by harnessing the cytoprotective properties of NO.


Asunto(s)
Precondicionamiento Isquémico Miocárdico , Isquemia Miocárdica , Óxido Nítrico Sintasa/fisiología , Óxido Nítrico/fisiología , Animales , Humanos , Modelos Biológicos , Óxido Nítrico Sintasa de Tipo II , Fenotipo , Factores de Tiempo
10.
Basic Res Cardiol ; 96(5): 487-96, 2001 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-11605996

RESUMEN

The goal of this study was to determine whether the protective effects of the A3AR agonist N6-(3-iodobenzyl)adenosine-5'-N-methylcarboxamide (IB-MECA) against myocardial stunning are mediated by the A1AR. Six groups of conscious rabbits underwent a sequence of six 4-minute coronary occlusion (O)/4-minute reperfusion (R) cycles for three consecutive days (days 1, 2, and 3). In vehicle-treated rabbits (group I), the recovery of systolic wall thickening (WTh) in the ischemic/reperfused region was markedly depressed on day 1, indicating the presence of severe myocardial stunning. On days 2 and 3, however, the recovery of systolic WTh was markedly accelerated, indicating the presence of late ischemic preconditioning (PC). When rabbits were pretreated with the A1AR agonist 2-chloro-N6-cyclopentyladenosine (CCPA, 100 microg/kg i.v.) or with IB-MECA (100 microg/kg i.v.) 10 min prior to the first sequence of O/R cycles on day 1 (group III and V, respectively), the recovery of systolic WTh was markedly accelerated compared to vehicle-treated animals (reflected as an approximately 48% decrease in the total deficit of systolic WTh). The magnitude of the protection afforded by adenosine receptor agonists was equivalent to that provided by late ischemic PC. Pre-treating rabbits with the A1AR antagonist N-0861 completely blocked both the hemodynamic and the cardioprotective effects of CCPA (group IV). However, the same dose of N-0861 did not block the cardioprotective actions of IB-MECA (group VI). Importantly, N-0861 did not influence the degree of myocardial stunning in the absence of PC (group II) and it did not block the development of late ischemic PC. Taken together, these results provide conclusive evidence that the cardioprotective effects of IB-MECA are not mediated via the A1AR, supporting the concept that activation of A3ARs prior to an ischemic challenge provides protection against ischemia/reperfusion injury.


Asunto(s)
Adenosina/análogos & derivados , Adenosina/farmacología , Cardiotónicos/farmacología , Aturdimiento Miocárdico/tratamiento farmacológico , Aturdimiento Miocárdico/metabolismo , Receptores Purinérgicos P1/metabolismo , Animales , Estado de Conciencia , Frecuencia Cardíaca , Masculino , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Daño por Reperfusión Miocárdica/metabolismo , Miocardio/metabolismo , Conejos
11.
Proc Natl Acad Sci U S A ; 98(20): 11114-9, 2001 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-11553773

RESUMEN

Conflicting roles for protein kinase C (PKC) isozymes in cardiac disease have been reported. Here, deltaPKC-selective activator and inhibitor peptides were designed rationally, based on molecular modeling and structural homology analyses. Together with previously identified activator and inhibitor peptides of epsilonPKC, deltaPKC peptides were used to identify cardiac functions of these isozymes. In isolated cardiomyocytes, perfused hearts, and transgenic mice, deltaPKC and epsilonPKC had opposing actions on protection from ischemia-induced damage. Specifically, activation of epsilonPKC caused cardioprotection whereas activation of deltaPKC increased damage induced by ischemia in vitro and in vivo. In contrast, deltaPKC and epsilonPKC caused identical nonpathological cardiac hypertrophy; activation of either isozyme caused nonpathological hypertrophy of the heart. These results demonstrate that two related PKC isozymes have both parallel and opposing effects in the heart, indicating the danger in the use of therapeutics with nonselective isozyme inhibitors and activators. Moreover, reduction in cardiac damage caused by ischemia by perfusion of selective regulator peptides of PKC through the coronary arteries constitutes a major step toward developing a therapeutic agent for acute cardiac ischemia.


Asunto(s)
Cardiomegalia/fisiopatología , Corazón/efectos de los fármacos , Isoenzimas/metabolismo , Contracción Miocárdica/fisiología , Isquemia Miocárdica/fisiopatología , Fragmentos de Péptidos/farmacología , Péptidos/farmacología , Proteína Quinasa C/metabolismo , Secuencia de Aminoácidos , Animales , Células Cultivadas , Activación Enzimática , Corazón/fisiología , Corazón/fisiopatología , Hemodinámica/efectos de los fármacos , Hemodinámica/fisiología , Técnicas In Vitro , Isoenzimas/química , Masculino , Ratones , Ratones Transgénicos , Modelos Moleculares , Datos de Secuencia Molecular , Contracción Miocárdica/efectos de los fármacos , Fragmentos de Péptidos/síntesis química , Fragmentos de Péptidos/química , Péptidos/síntesis química , Péptidos/química , Conformación Proteica , Proteína Quinasa C/química , Proteína Quinasa C-delta , Proteína Quinasa C-epsilon , Ratas , Ratas Wistar , Alineación de Secuencia , Homología de Secuencia de Aminoácido
13.
Proc Natl Acad Sci U S A ; 98(16): 9050-5, 2001 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-11481471

RESUMEN

The goal of this study was to determine the role of the Janus tyrosine kinase (JAK)-signal transducers and activators of transcription (STAT) pathway in the late phase of ischemic preconditioning (PC). A total of 230 mice were used. At 5 min after ischemic PC (induced with six cycles of 4-min coronary occlusion/4-min reperfusion), immunoprecipitation with anti-phosphotyrosine (anti-pTyr) antibodies followed by immunoblotting with anti-JAK antibodies revealed increased tyrosine phosphorylation of JAK1 (+257 +/- 53%) and JAK2 (+238 +/- 35%), indicating rapid activation of these two kinases. Similar results were obtained by immunoblotting with anti-pTyr-JAK1 and anti-pTyr-JAK2 antibodies. Western analysis with anti-pTyr-STAT antibodies demonstrated a marked increase in nuclear pTyr-STAT1 (+301 +/- 61%) and pTyr-STAT3 (+253 +/- 60%) 30 min after ischemic PC, which was associated with redistribution of STAT1 and STAT3 from the cytosolic to the nuclear fraction and with an increase in STAT1 and STAT3 gamma-IFN activation site DNA-binding activity (+606 +/- 64%), indicating activation of STAT1 and STAT3. No nuclear translocation or tyrosine phosphorylation of STAT2, STAT4, STAT5A, STAT5B, or STAT6 was observed. Pretreatment with the JAK inhibitor AG-490 20 min before the six occlusion/reperfusion cycles blocked the enhanced tyrosine phosphorylation of JAK1 and JAK2 and the increased tyrosine phosphorylation, nuclear translocation, and enhanced DNA-binding activity of STAT1 and STAT3. The same dose of AG-490 abrogated the protection against myocardial infarction and the concomitant up-regulation of inducible NO synthase (iNOS) protein and activity observed 24 h after ischemic PC. Taken together, these results demonstrate that ischemic PC induces isoform-selective activation of JAK1, JAK2, STAT1, and STAT3, and that ablation of this response impedes the up-regulation of iNOS and the concurrent acquisition of ischemic tolerance. This study demonstrates that the JAK-STAT pathway plays an essential role in the development of late PC. The results reveal a signaling mechanism that underlies the transcriptional up-regulation of the cardiac iNOS gene and the adaptation of the heart to ischemic stress.


Asunto(s)
Precondicionamiento Isquémico , Proteínas Tirosina Quinasas/metabolismo , Transactivadores/metabolismo , Animales , Secuencia de Bases , Sondas de ADN , Masculino , Ratones , Ratones Endogámicos ICR , Infarto del Miocardio/enzimología , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Óxido Nítrico Sintasa/metabolismo , Tirfostinos/farmacología
14.
Circulation ; 104(6): 694-9, 2001 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-11489777

RESUMEN

BACKGROUND: Recent studies suggest that the late phase of ischemic preconditioning (PC) can be mimicked by pretreatment with NO donors. The ability of clinically relevant NO donors to induce PC against infarction, however, has not been evaluated. Furthermore, it is unknown whether tolerance to the hemodynamic actions of nitrates also extends to their PC effects. METHODS AND RESULTS: Conscious rabbits underwent a 30-minute coronary occlusion and 3 days of reperfusion. A 60-minute intravenous (IV) infusion of nitroglycerin (NTG) ending 1 hour before occlusion reduced infarct size, indicating an early PC effect. When the time interval between NTG infusion and occlusion was extended to 24 or 72 hours, the infarct-sparing action of NTG became even more pronounced, indicating a robust late PC effect. Transdermal NTG patches elicited a late PC effect that was (1) equivalent to that induced by IV NTG, demonstrating the efficacy of transdermal NTG as an alternative form of NTG delivery for inducing late PC, and (2) similar in nitrate-tolerant and -nontolerant rabbits, demonstrating that tolerance does not extend to the PC effects of NTG. CONCLUSIONS: In conscious rabbits, administration of NTG via either the IV or the transdermal route elicits a robust protective effect against infarction that lasts for 72 hours. The magnitude of NTG-induced cardioprotection is equivalent to that observed during the late phase of ischemic PC and is not affected by the development of tolerance. These findings reveal a new action of nitrates and support novel applications of these drugs for protecting the ischemic myocardium in patients.


Asunto(s)
Precondicionamiento Isquémico Miocárdico , Infarto del Miocardio/prevención & control , Nitroglicerina/farmacología , Vasodilatadores/farmacología , Administración Cutánea , Análisis de Varianza , Animales , Presión Sanguínea/efectos de los fármacos , Estado de Conciencia , Tolerancia a Medicamentos , Frecuencia Cardíaca/efectos de los fármacos , Infusiones Intravenosas , Infarto del Miocardio/patología , Conejos , Factores de Tiempo
15.
Am J Physiol Heart Circ Physiol ; 281(2): H959-68, 2001 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11454603

RESUMEN

Recent studies have demonstrated that the adenosine A1 receptor agonist 2-chloro-N6-cyclopentyladenosine (CCPA) and the adenosine A3 receptor agonist N6-(3-iodobenzyl)adenosine-5'-N-methyluronamide (IB-MECA) produce a delayed phase of protection against infarction similar to the late phase of ischemic preconditioning (PC). However, the mechanism for adenosine A1 or A3 receptor-induced late PC remains unknown. The goal of this study was to determine whether the delayed cardioprotective effects of adenosine A1 or A3 receptors are mediated by cyclooxygenase-2 (COX-2), which is an obligatory mediator of ischemic PC. We found that COX-2 protein expression (Western blotting) did not increase 24 h after the administration of either CCPA (100 microg/kg iv) or IB-MECA (300 microg/kg iv) compared with controls. To probe the role of constitutive COX-2 expression, conscious rabbits were subjected to 30-min coronary occlusion followed by 72-h reperfusion. Twenty-four hours before the occlusion, the rabbits were pretreated with CCPA (100 microg/kg iv) or IB-MECA (300 microg/kg iv). Both CCPA and IB-MECA resulted in a marked (approximately 47%) reduction in infarct size vs. controls [36.2 +/- 4.0% of the risk region (n = 9), 31.2 +/- 4.7% (n = 9), and 59.5 +/- 3.8% (n = 9), respectively; P < 0.05], similar to that induced by the late phase of ischemic PC [31.8 +/- 3.2% (n = 9)]. The selective COX-2 inhibitor N-(2-[cyclohexyloxy]4-nitrophenyl)methanesulfonamide (NS-398, 5 mg/kg), which abolished the protective effect of ischemic late PC, failed to block the protection of either CCPA or IB-MECA, indicating that COX-2 does not mediate the delayed protection of either CCPA or IB-MECA [CCPA + NS-398, 29.1 +/- 3.4% (n = 7); IB-MECA + NS-398, 34.9 +/- 2.9% (n = 8)]. NS-398 in itself did not affect infarct size [54.9 +/- 3.7% (n = 9)]. Taken together, these results demonstrate that, in contrast to ischemia-induced late PC, the mechanisms of adenosine A1 or A3 receptor-induced late PC is independent of COX-2.


Asunto(s)
Precondicionamiento Isquémico Miocárdico , Isoenzimas/fisiología , Prostaglandina-Endoperóxido Sintasas/fisiología , Receptores Purinérgicos P1/fisiología , Animales , Ciclooxigenasa 2 , Frecuencia Cardíaca , Masculino , Conejos , Receptor de Adenosina A3
16.
Circulation ; 103(24): 2935-41, 2001 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-11413083

RESUMEN

BACKGROUND: Experimental studies suggest that the cardioprotective effects of the late phase of ischemic preconditioning (PC) can be mimicked pharmacologically. However, to date, no drug has been tested with respect to its ability to elicit a late PC effect in humans. As a consequence, clinical exploitation of the powerful anti-stunning and anti-infarct actions of late PC has been elusive thus far. METHODS AND RESULTS: A total of 66 patients were randomized to receive a 4-hour intravenous infusion of nitroglycerin (NTG) or normal saline; on the following day, they underwent percutaneous transluminal coronary angioplasty (three 2-minute balloon inflations 5 minutes apart). Measurements of ST-segment shifts (intracoronary and surface ECGs), regional wall motion (quantitative 2D echocardiography), and chest pain score indicated that the infusion of NTG 24 hours before angioplasty rendered the myocardium relatively resistant to ischemia and that the degree of this cardioprotective effect was comparable to that afforded by the ischemia associated with the first balloon inflation in control subjects (early phase of ischemic PC). Collateral flow (estimated from a pressure-derived index) did not differ between control and NTG-pretreated patients, indicating that the enhanced tolerance to ischemia in NTG-pretreated patients cannot be accounted for by baseline differences in collateral function. CONCLUSIONS: NTG protects human myocardium against ischemia 24 hours after its administration. To the best of our knowledge, this is the first report that a late PC effect can be recruited pharmacologically in humans. The results suggest that prophylactic administration of nitrates could be a novel approach to the protection of the ischemic myocardium in patients.


Asunto(s)
Angioplastia Coronaria con Balón , Enfermedad Coronaria/terapia , Precondicionamiento Isquémico Miocárdico/métodos , Nitroglicerina/administración & dosificación , Velocidad del Flujo Sanguíneo/efectos de los fármacos , Presión Sanguínea/efectos de los fármacos , Dolor en el Pecho/tratamiento farmacológico , Circulación Colateral/efectos de los fármacos , Circulación Coronaria/efectos de los fármacos , Enfermedad Coronaria/diagnóstico , Ecocardiografía , Electrocardiografía/efectos de los fármacos , Pruebas de Función Cardíaca/efectos de los fármacos , Frecuencia Cardíaca/efectos de los fármacos , Humanos , Infusiones Intravenosas , Infarto del Miocardio/prevención & control , Aturdimiento Miocárdico/prevención & control , Método Simple Ciego , Resultado del Tratamiento , Vasodilatadores/administración & dosificación , Función Ventricular Izquierda/efectos de los fármacos
17.
Circ Res ; 88(12): 1306-13, 2001 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-11420308

RESUMEN

An essential role for protein kinase C epsilon (PKCepsilon) has been shown in multiple forms of cardioprotection; however, there is a distinct paucity of information concerning the signaling architecture that is responsible for the manifestation of a protective phenotype. We and others have recently shown that signal transduction may proceed via the formation of signaling complexes (Circ Res. 2001;88:59-62). In order to understand if the assembly of multiprotein complexes is the manner by which signaling is conducted in cardioprotection, we designed a series of experiments to characterize the associations of Src tyrosine kinase with PKCepsilon in a conscious rabbit model of nitric oxide (NO)-induced late preconditioning. Our data demonstrate that PKCepsilon and Src can form functional signaling modules in vitro: PKCepsilon interacts with Src; the association with PKCepsilon activates Src; and adult cardiac cells receiving recombinant adenoviruses encoding PKCepsilon exhibit increased Src activity. Furthermore, our results show that NO-induced late preconditioning involved PKCepsilon-Src module formation and enhanced the enzymatic activity of PKCepsilon-associated Src. Inhibition of PKC blocked cardioprotection, module formation, and PKCepsilon-associated Src activity, providing direct evidence for a functional role of the PKCepsilon-Src module in the orchestration of NO-induced cardioprotection in conscious rabbits.


Asunto(s)
Precondicionamiento Isquémico Miocárdico , Isoenzimas/metabolismo , Óxido Nítrico/metabolismo , Proteína Quinasa C/metabolismo , Transducción de Señal/fisiología , Familia-src Quinasas/metabolismo , Animales , Activación Enzimática/fisiología , Inhibidores Enzimáticos/farmacología , Glutatión Transferasa/genética , Isoenzimas/genética , Sustancias Macromoleculares , Ratones , Complejos Multiproteicos , Miocardio/citología , Miocardio/metabolismo , Óxido Nítrico/farmacología , Donantes de Óxido Nítrico/farmacología , Compuestos Nitrosos/farmacología , Unión Proteica/fisiología , Proteína Quinasa C/genética , Proteína Quinasa C-epsilon , Conejos , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transducción de Señal/efectos de los fármacos , Transfección , Familia-src Quinasas/genética
18.
Circulation ; 103(14): 1893-8, 2001 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-11294809

RESUMEN

BACKGROUND: Extracellular superoxide dismutase (Ec-SOD) may protect the heart against myocardial infarction (MI) because of its extended half-life and capacity to bind heparan sulfate proteoglycans on cellular surfaces. Accordingly, we used direct gene transfer to increase systemic levels of Ec-SOD and determined whether this gene therapy could protect against MI. METHODS AND RESULTS: The cDNA for human Ec-SOD was incorporated into a replication-deficient adenovirus (Ad5/CMV/Ec-SOD). Injection of this virus produced a high level of Ec-SOD in the liver, which was redistributed to the heart and other organs by injection of heparin. Untreated rabbits (group I) underwent a 30-minute coronary occlusion and 3 days of reperfusion. For comparison, preconditioned rabbits (group II) underwent a sequence of six 4-minute-occlusion/4-minute-reperfusion cycles 24 hours before the 30-minute occlusion. Control-treated rabbits (group III) were injected intravenously with Ad5/CMV/nls-LacZ, and gene-therapy rabbits (group IV) were injected with Ad5/CMV/Ec-SOD 3 days before the 30-minute occlusion. Both groups treated with Ad5 received intravenous heparin 2 hours before the 30-minute occlusion. Infarct size (percent risk area) was similar in groups I (57+/-6%) and III (58+/-5%). Ec-SOD gene therapy markedly reduced infarct size to 25+/-4% (P<0.01, group IV versus group III), a protection comparable to that of the late phase of ischemic preconditioning (29+/-3%, P<0.01 group II versus group I). CONCLUSIONS: Direct gene transfer of the cDNA encoding membrane-bound Ec-SOD affords powerful cardioprotection, providing proof of principle for the effectiveness of antioxidant gene therapy against MI.


Asunto(s)
Terapia Genética , Infarto del Miocardio/terapia , Superóxido Dismutasa/metabolismo , Animales , Arritmias Cardíacas/etiología , Arritmias Cardíacas/fisiopatología , Arritmias Cardíacas/prevención & control , Presión Sanguínea/fisiología , Células COS , Estado de Conciencia , Depuradores de Radicales Libres/metabolismo , Depuradores de Radicales Libres/uso terapéutico , Expresión Génica , Frecuencia Cardíaca/fisiología , Ventrículos Cardíacos/patología , Ventrículos Cardíacos/fisiopatología , Humanos , Masculino , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Daño por Reperfusión Miocárdica/complicaciones , Tamaño de los Órganos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Conejos , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/uso terapéutico , Superóxido Dismutasa/genética , Superóxido Dismutasa/uso terapéutico
19.
J Mol Cell Cardiol ; 33(4): 825-30, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11273734

RESUMEN

We used mice with genetic disruption of the A3 adenosine receptor (AR) gene (A3AR(-/-)mice) to assess the in vivo role of the A3AR in modulating myocardial ischemia/reperfusion injury and preconditioning (PC). Surprisingly, infarct size induced by 30 min of coronary artery occlusion and 24 h of reperfusion was 35% smaller in A3AR(-/-)compared to wild-type mice (A3AR(+/+)). The reduction in infarct size was not the result of differences in heart rate, body temperature or increased cardiac expression of A1ARs. However, neutrophil infiltration within infarcted regions was less in A3AR(-/-)mice. Furthermore, ischemic PC induced by either a single episode (one 5 min occlusion) or multiple episodes (six 4 min occlusions) of ischemia produced equivalent reductions in infarct size in A3AR(-/-)and A3AR(+/+)mice. These results indicate that, in the mouse, (i) A3ARs play an injurious role during acute myocardial ischemia/reperfusion injury, possibly by exacerbating the inflammatory response, and (ii) A3ARs are not necessary for the development of the early phase of ischemic PC.


Asunto(s)
Precondicionamiento Isquémico Miocárdico , Infarto del Miocardio/prevención & control , Isquemia Miocárdica/prevención & control , Daño por Reperfusión Miocárdica/prevención & control , Receptores Purinérgicos P1/fisiología , Animales , Temperatura Corporal , Marcación de Gen , Frecuencia Cardíaca , Inmunidad Innata , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Isquemia Miocárdica/patología , Isquemia Miocárdica/fisiopatología , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/fisiopatología , Neutrófilos/fisiología , Ensayo de Unión Radioligante , Receptor de Adenosina A3 , Receptores Purinérgicos P1/genética , Factores de Tiempo
20.
Circ Res ; 88(5): 520-8, 2001 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-11249876

RESUMEN

We investigated whether activation of A(1) or A(3) adenosine receptors (ARs) induces late preconditioning (PC) against infarction in conscious rabbits using the selective AR agonists 2-chloro-N(6)-cyclopentyladenosine (CCPA) and N(6)-3-iodobenzyladenosine-5'-N-methylcarboxamide (IB-MECA). In vitro radioligand binding and cAMP assays demonstrated CCPA to be approximately 200- to 400-fold selective for the rabbit A(1)AR and IB-MECA to be approximately 20-fold selective for the rabbit A(3)AR. We observed that (1) pretreatment of rabbits 24 hours earlier with CCPA (100 microgram/kg IV bolus) or IB-MECA (100 or 300 microgram/kg) resulted in an approximately 35% to 40% reduction in the size of the infarct induced by 30 minutes of coronary artery occlusion and 72 hours of reperfusion compared with vehicle-treated rabbits, whereas pretreatment with the selective A(2A)AR agonist CGS 21680 (100 microgram/kg) had no effect; (2) the delayed cardioprotective effect of CCPA, but not that of IB-MECA, was completely blocked by coadministration of the highly selective A(1)AR antagonist N-0861; (3) inhibition of nitric oxide synthase (NOS) with N(omega)-nitro-L-arginine during the 30-minute occlusion abrogated the infarct-sparing action of CCPA but not that of IB-MECA; and (4) inhibition of ATP-sensitive potassium (K(ATP)) channels with sodium 5-hydroxydecanoate during the 30-minute occlusion blocked the cardioprotective effects of both CCPA and IB-MECA. Taken together, these results indicate that activation of either A(1)ARs or A(3)ARs (but not A(2A)ARs) elicits delayed protection against infarction in conscious rabbits and that both A(1)AR- and A(3)AR-induced cardioprotection involves opening of K(ATP) channels. However, A(1)AR-induced late PC uses an NOS-dependent pathway whereas A(3)AR-induced late PC is mediated by an NOS-independent pathway.


Asunto(s)
Adenosina/análogos & derivados , Precondicionamiento Isquémico Miocárdico , Infarto del Miocardio/prevención & control , Receptores Purinérgicos P1/metabolismo , Adenina/análogos & derivados , Adenina/farmacología , Adenosina/metabolismo , Adenosina/farmacología , Animales , Unión Competitiva/efectos de los fármacos , Línea Celular , Estado de Conciencia , Ácidos Decanoicos/farmacología , Humanos , Hidroxiácidos/farmacología , Radioisótopos de Yodo , Membranas/metabolismo , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Nitroarginina/farmacología , Norbornanos/farmacología , Fenetilaminas/farmacología , Conejos , Ensayo de Unión Radioligante , Receptor de Adenosina A3 , Receptores Purinérgicos P1/efectos de los fármacos , Receptores Purinérgicos P1/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...