Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Int J Mol Sci ; 24(15)2023 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-37569376

RESUMEN

Cardiac complications are frequently found following a stroke in humans whose pathophysiological mechanism remains poorly understood. We used machine learning to analyse a large set of data from a metabolipidomic study assaying 630 metabolites in a rat stroke model to investigate metabolic changes affecting the heart within 72 h after a stroke. Twelve rats undergoing a stroke and 28 rats undergoing the sham procedure were investigated. A plasmatic signature consistent with the literature with notable lipid metabolism remodelling was identified. The post-stroke heart showed a discriminant metabolic signature, in comparison to the sham controls, involving increased collagen turnover, increased arginase activity with decreased nitric oxide synthase activity as well as an altered amino acid metabolism (including serine, asparagine, lysine and glycine). In conclusion, these results demonstrate that brain injury induces a metabolic remodelling in the heart potentially involved in the pathophysiology of stroke heart syndrome.

2.
Basic Res Cardiol ; 115(2): 13, 2020 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-31925554

RESUMEN

The actual protective mechanisms underlying cardioprotection with remote ischemic conditioning (RIC) remain unclear. Recent data suggest that RIC induces kynurenine (KYN) and kynurenic acid synthesis, two metabolites derived from tryptophan (TRP), yet a causal relation between TRP pathway and RIC remains to be established. We sought to study the impact of RIC on the levels of TRP and its main metabolites within tissues, and to assess whether blocking kynurenine (KYN) synthesis from TRP would inhibit RIC-induced cardioprotection. In rats exposed to 40-min coronary occlusion and 2-h reperfusion, infarct size was significantly smaller in RIC-treated animals (35.7 ± 3.0% vs. 46.5 ± 2.2%, p = 0.01). This protection was lost in rats that received 1-methyl-tryptophan (1-MT) pretreatment, an inhibitor of KYN synthesis from TRP (infarct size = 46.2 ± 5.0%). Levels of TRP and nine compounds spanning its metabolism through the serotonin and KYN pathways were measured by reversed-phase liquid chromatography-tandem mass spectrometry in the liver, heart, and limb skeletal muscle, either exposed or not to RIC. In the liver, RIC induced a significant increase in xanthurenic acid, nicotinic acid, and TRP. Likewise, RIC increased NAD-dependent deacetylase sirtuin activity in the liver. Pretreatment with 1-MT suppressed the RIC-induced increases in NAD-dependent deacetylase sirtuin activity. Altogether, these findings indicate that RIC mechanism is dependent on TRP-KYN pathway activation.


Asunto(s)
Precondicionamiento Isquémico Miocárdico , Quinurenina/metabolismo , Infarto del Miocardio/prevención & control , Daño por Reperfusión Miocárdica/prevención & control , Miocardio/metabolismo , Triptófano/metabolismo , Animales , Modelos Animales de Enfermedad , Hígado/metabolismo , Masculino , Músculo Esquelético/metabolismo , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Miocardio/patología , Ratas Wistar
3.
Arterioscler Thromb Vasc Biol ; 36(8): 1598-606, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27255725

RESUMEN

OBJECTIVE: Myogenic tone (MT) of resistance arteries ensures autoregulation of blood flow in organs and relies on the intrinsic property of smooth muscle to contract in response to stretch. Nucleotides released by mechanical strain on cells are responsible for pleiotropic vascular effects, including vasoconstriction. Here, we evaluated the contribution of extracellular nucleotides to MT. APPROACH AND RESULTS: We measured MT and the associated pathway in mouse mesenteric resistance arteries using arteriography for small arteries and molecular biology. Of the P2 receptors in mouse mesenteric resistance arteries, mRNA expression of P2X1 and P2Y6 was dominant. P2Y6 fully sustained UDP/UTP-induced contraction (abrogated in P2ry6(-/-) arteries). Preventing nucleotide hydrolysis with the ectonucleotidase inhibitor ARL67156 enhanced pressure-induced MT by 20%, whereas P2Y6 receptor blockade blunted MT in mouse mesenteric resistance arteries and human subcutaneous arteries. Despite normal hemodynamic parameters, P2ry6(-/-) mice were protected against MT elevation in myocardial infarction-induced heart failure. Although both P2Y6 and P2Y2 receptors contributed to calcium mobilization, P2Y6 activation was mandatory for RhoA-GTP binding, myosin light chain, P42-P44, and c-Jun N-terminal kinase phosphorylation in arterial smooth muscle cells. In accordance with the opening of a nucleotide conduit in pressurized arteries, MT was altered by hemichannel pharmacological inhibitors and impaired in Cx43(+/-) and P2rx7(-/-) mesenteric resistance arteries. CONCLUSIONS: Signaling through P2 nucleotide receptors contributes to MT. This mechanism encompasses the release of nucleotides coupled to specific autocrine/paracrine activation of the uracil nucleotide P2Y6 receptor and may contribute to impaired tissue perfusion in cardiovascular diseases.


Asunto(s)
Arteriolas/metabolismo , Mesenterio/irrigación sanguínea , Receptores Purinérgicos P2/metabolismo , Vasoconstricción , Adenosina Trifosfatasas/metabolismo , Animales , Arteriolas/efectos de los fármacos , Arteriolas/fisiopatología , Presión Sanguínea , Señalización del Calcio , Células Cultivadas , Conexina 43/deficiencia , Conexina 43/genética , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Genotipo , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/fisiopatología , Hidrólisis , Mecanotransducción Celular , Ratones Noqueados , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Músculo Liso Vascular/metabolismo , Infarto del Miocardio/complicaciones , Miocitos del Músculo Liso/metabolismo , Cadenas Ligeras de Miosina/metabolismo , Fenotipo , Fosforilación , Agonistas del Receptor Purinérgico P2X/farmacología , Receptores Purinérgicos P2/deficiencia , Receptores Purinérgicos P2/efectos de los fármacos , Receptores Purinérgicos P2/genética , Receptores Purinérgicos P2X7/deficiencia , Receptores Purinérgicos P2X7/genética , Uridina Difosfato/farmacología , Vasoconstricción/efectos de los fármacos , Vasoconstrictores/farmacología , Proteínas de Unión al GTP rho/metabolismo , Proteína de Unión al GTP rhoA
4.
J Mol Cell Cardiol ; 65: 98-104, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24140799

RESUMEN

Remote ischemic preconditioning (RIPC) has emerged as a feasible and attractive therapeutic procedure for heart protection against ischemia/reperfusion (I/R) injury. However, its molecular mechanisms remain poorly understood. Hypoxia inducible factor-1α (HIF-1α) is a transcription factor that plays a key role in the cellular adaptation to hypoxia and ischemia. This study's aim was to test whether RIPC-induced cardioprotection requires HIF-1α upregulation to be effective. In the first study, wild-type mice and mice heterozygous for HIF1a (gene encoding the HIF-1α protein) were subjected to RIPC immediately before myocardial infarction (MI). RIPC resulted in a robust HIF-1α activation in the limb and acute cardioprotection in wild-type mice. RIPC-induced cardioprotection was preserved in heterozygous mice, despite the low HIF-1α expression in their limbs. In the second study, the role of HIF-1α in RIPC was evaluated using cadmium (Cd), a pharmacological HIF-1α inhibitor. Rats were subjected to MI (MI group) or to RIPC immediately prior to MI (R-MI group). Cd was injected 18 0min before RIPC (Cd-R-MI group). RIPC induced robust HIF-1α activation in rat limbs and significantly reduced infarct size (IS). Despite Cd's inhibition of HIF-1α activation, RIPC-induced cardioprotection was preserved in the Cd-R-MI group. RIPC applied immediately prior to MI increased HIF-1α expression and attenuated IS in rats and wild-type mice. However, RIPC-induced cardioprotection was preserved in partially HIF1a-deficient mice and in rats pretreated with Cd. When considered together, these results suggest that HIF-1α upregulation is unnecessary in acute RIPC.


Asunto(s)
Extremidades/irrigación sanguínea , Extremidades/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Precondicionamiento Isquémico , Animales , Cadmio/farmacología , Cardiotónicos/metabolismo , Activación Enzimática/efectos de los fármacos , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Subunidad alfa del Factor 1 Inducible por Hipoxia/deficiencia , Masculino , Ratones , Infarto del Miocardio/patología , Miocardio/enzimología , Miocardio/patología , Fosforilación/efectos de los fármacos , Ratas , Ratas Sprague-Dawley
5.
Cardiology ; 125(4): 242-9, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23816794

RESUMEN

PURPOSE: In acute myocardial infarction, left ventricular (LV) unloading reduces endothelin-1 (ET-1) release. We tested that endogenous ET-1 released during acute myocardial infarction might mediate ischemia/reperfusion (I/R) injury by stimulating increased intracellular calcium concentration, [Ca(2+)]i, and apoptosis. METHODS: Rabbits were subjected to 1 h of coronary artery occlusion followed by 3 h of reperfusion. Unloading was initiated 15 min prior to reperfusion and was maintained during reperfusion. The control group was subjected to reperfusion. Animals were treated with ET-1 receptor antagonist BQ123. In parallel, isolated rabbit cardiomyocytes subjected to simulated I/R with or without ET-1 or BQ123, intracellular Ca(2+) and cell death were assessed with flow cytometry. RESULTS: LV unloading prior to reperfusion reduced myocardial ET-1 release at 2 h of reperfusion. Infarct size was reduced in unloaded and BQ123 groups versus controls. LV unloading and BQ123 treatment reduced the percentage of apoptotic cells associated with increases in Bcl-2 protein levels in ischemic regions. BQ123 reduced both ET-1-induced [Ca(2+)]i increase and cell death for myocytes subjected to stimulated I/R. CONCLUSION: We propose that components of reperfusion injury involve ET-1 release which stimulates calcium overload and apoptosis. Intravenous ET-1 receptor blockade prior to reperfusion may be a protective adjunct to reperfusion therapy in acute myocardial infarction patients.


Asunto(s)
Endotelina-1/metabolismo , Infarto del Miocardio/fisiopatología , Daño por Reperfusión Miocárdica/fisiopatología , Animales , Apoptosis/fisiología , Calcio/metabolismo , Vasos Coronarios , Antagonistas de los Receptores de la Endotelina A , Hemodinámica/fisiología , Ligadura , Masculino , Daño por Reperfusión Miocárdica/metabolismo , Miocitos Cardíacos/metabolismo , Péptidos Cíclicos/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Conejos , Proteína X Asociada a bcl-2/metabolismo
6.
Pharmaceuticals (Basel) ; 16(10)2023 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-37895852

RESUMEN

BACKGROUND: Myocardial infarction is one of the leading causes of mortality worldwide; hence, there is an urgent need to discover novel cardioprotective strategies. Kynurenic acid (KYNA), a metabolite of the kynurenine pathway, has been previously reported to have cardioprotective effects. However, the mechanisms by which KYNA may be protective are still unclear. The current study addressed this issue by investigating KYNA's cardioprotective effect in the context of myocardial ischemia/reperfusion. METHODS: H9C2 cells and rats were exposed to hypoxia/reoxygenation or myocardial infarction, respectively, in the presence or absence of KYNA. In vitro, cell death was quantified using flow cytometry analysis of propidium iodide staining. In vivo, TTC-Evans Blue staining was performed to evaluate infarct size. Mitochondrial respiratory chain complex activities were measured using spectrophotometry. Protein expression was evaluated by Western blot, and mRNA levels by RT-qPCR. RESULTS: KYNA treatment significantly reduced H9C2-relative cell death as well as infarct size. KYNA did not exhibit any effect on the mitochondrial respiratory chain complex activity. SOD2 mRNA levels were increased by KYNA. A decrease in p62 protein levels together with a trend of increase in PARK2 may mark a stimulation of mitophagy. Additionally, ERK1/2, Akt, and FOXO3α phosphorylation levels were significantly reduced after the KYNA treatment. Altogether, KYNA significantly reduced myocardial ischemia/reperfusion injuries in both in vitro and in vivo models. CONCLUSION: Here we show that KYNA-mediated cardioprotection was associated with enhanced mitophagy and antioxidant defense. A deeper understanding of KYNA's cardioprotective mechanisms is necessary to identify promising novel therapeutic targets and their translation into the clinical arena.

7.
Am J Physiol Heart Circ Physiol ; 303(7): H871-7, 2012 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-22886414

RESUMEN

Remote ischemic conditioning (RCond) induced by short periods of ischemia and reperfusion of an organ or tissue before myocardial reperfusion is an attractive strategy of cardioprotection in the context of acute myocardial infarction. Nonetheless, its mechanism remains unknown. A humoral factor appears to be involved, although its identity is currently unknown. We hypothesized that the circulating microparticles (MPs) are the link between the remote tissue and the heart. MPs from rats and healthy humans undergoing RCond were characterized. In rats, RCond was induced by 10 min of limb ischemia. In humans, RCond was induced by three cycles of 5-min inflation and 5-min deflation of a blood-pressure cuff. In the second part of the study, rats underwent 40 min myocardial ischemia followed by 2 h reperfusion. Infarct size was measured and compared among three groups of rats: 1) myocardial infarction alone (MI) (n = 6); 2) MI + RCond started 20 min after coronary ligation (n = 6); and 3) MI + injection of RCond-derived rat MPs (MI + MPs) (n = 5). MPs from endothelial cells (CD54(+) and CD146(+) for rats and humans, respectively) and procoagulant MPs (Annexin V(+)) markedly increased after RCond, both in rats and humans. RCond reduced infarct size (24.4 ± 5.9% in MI + RCond vs. 54.6 ± 4.7% in MI alone; P < 0.01). Infarct size did not decrease in MI + MPs compared with MI alone (50.2 ± 6.4% vs. 54.6 ± 4.7%, not significantly different). RCond increased endothelium-derived and procoagulant MPs in both rats and humans. However, MP release did not appear to be a biological vector of RCond in our model.


Asunto(s)
Micropartículas Derivadas de Células/patología , Endotelio Vascular/patología , Miembro Posterior/irrigación sanguínea , Precondicionamiento Isquémico/métodos , Infarto del Miocardio/prevención & control , Daño por Reperfusión Miocárdica/prevención & control , Transducción de Señal , Extremidad Superior/irrigación sanguínea , Adulto , Animales , Biomarcadores/sangre , Coagulación Sanguínea , Micropartículas Derivadas de Células/metabolismo , Micropartículas Derivadas de Células/trasplante , Constricción , Modelos Animales de Enfermedad , Endotelio Vascular/metabolismo , Endotelio Vascular/trasplante , Humanos , Masculino , Infarto del Miocardio/sangre , Infarto del Miocardio/patología , Daño por Reperfusión Miocárdica/sangre , Daño por Reperfusión Miocárdica/patología , Miocardio/metabolismo , Miocardio/patología , Ratas , Ratas Wistar , Factores de Tiempo , Torniquetes
8.
Basic Res Cardiol ; 106(1): 147-62, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20981553

RESUMEN

Recent studies reported cardioprotective effects of erythropoietin (EPO) against ischemia-reperfusion (I/R) injury through activation of the reperfusion injury salvage kinase (RISK) pathway. As RISK has been reported to be impaired in diabetes and insulin resistance syndrome, we examined whether EPO-induced cardioprotection was maintained in rat models of type 1 diabetes and insulin resistance syndrome. Isolated hearts were obtained from three rat cohorts: healthy controls, streptozotocin (STZ)-induced diabetes, and high-fat diet (HFD)-induced insulin resistance syndrome. All hearts underwent 25 min ischemia and 30 min or 120 min reperfusion. They were assigned to receive either no intervention or a single dose of EPO at the onset of reperfusion. In hearts from healthy controls, EPO decreased infarct size (14.36 ± 0.60 and 36.22 ± 4.20% of left ventricle in EPO-treated and untreated hearts, respectively, p < 0.05) and increased phosphorylated forms of Akt, ERK1/2, and their downstream target GSK-3ß. In hearts from STZ-induced diabetic rats, EPO did not decrease infarct size (32.05 ± 2.38 and 31.88 ± 1.87% in EPO-treated and untreated diabetic rat hearts, respectively, NS) nor did it increase phosphorylation of Akt, ERK1/2, and GSK-3ß. In contrast, in hearts from HFD-induced insulin resistance rats, EPO decreased infarct size (18.66 ± 1.99 and 34.62 ± 3.41% in EPO-treated and untreated HFD rat hearts, respectively, p < 0.05) and increased phosphorylation of Akt, ERK1/2, and GSK-3ß. Administration of GSK-3ß inhibitor SB216763 was cardioprotective in healthy and diabetic hearts. STZ-induced diabetes abolished EPO-induced cardioprotection against I/R injury through a disruption of upstream signaling of GSK-3ß. In conclusion, direct inhibition of GSK-3ß may provide an alternative strategy to protect diabetic hearts against I/R injury.


Asunto(s)
Diabetes Mellitus Experimental/complicaciones , Eritropoyetina/uso terapéutico , Glucógeno Sintasa Quinasa 3/metabolismo , Resistencia a la Insulina , Daño por Reperfusión Miocárdica/prevención & control , Animales , Apoptosis/efectos de los fármacos , Cardiotónicos/administración & dosificación , Diabetes Mellitus Experimental/metabolismo , Grasas de la Dieta , Eritropoyetina/farmacología , Glucosa/toxicidad , Glucógeno Sintasa Quinasa 3 beta , Hemodinámica , Técnicas In Vitro , Sistema de Señalización de MAP Quinasas , Infarto del Miocardio/etiología , Infarto del Miocardio/patología , Infarto del Miocardio/prevención & control , Daño por Reperfusión Miocárdica/complicaciones , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Miocardio/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Wistar , Receptores de Eritropoyetina/metabolismo
9.
Basic Res Cardiol ; 106(6): 1329-39, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21833651

RESUMEN

Local ischemic postconditioning (IPost) and remote ischemic perconditioning (RIPer) are promising methods to decrease ischemia-reperfusion (I/R) injury. We tested whether the use of the two procedures in combination led to an improvement in cardioprotection through a higher activation of survival signaling pathways. Rats exposed to myocardial I/R were allocated to one of the following four groups: Control, no intervention at myocardial reperfusion; IPost, three cycles of 10-s coronary artery occlusion followed by 10-s reperfusion applied at the onset of myocardial reperfusion; RIPer, 10-min limb ischemia followed by 10-min reperfusion initiated 20 min after coronary artery occlusion; IPost+RIPer, IPost and RIPer in combination. Infarct size was significantly reduced in both IPost and RIPer (34.25 ± 3.36 and 24.69 ± 6.02%, respectively) groups compared to Control (54.93 ± 6.46%, both p < 0.05). IPost+RIPer (infarct size = 18.04 ± 4.86%) was significantly more cardioprotective than IPost alone (p < 0.05). RISK pathway (Akt, ERK1/2, and GSK-3ß) activation was enhanced in IPost, RIPer, and IPost+RIPer groups compared to Control. IPost+RIPer did not enhance RISK pathway activation as compared to IPost alone, but instead increased phospho-STAT-3 levels, highlighting the crucial role of the SAFE pathway. In IPost+RIPer, a SAFE inhibitor (AG490) abolished cardioprotection and blocked both Akt and GSK-3ß phosphorylations, whereas RISK inhibitors (wortmannin or U0126) abolished cardioprotection and blocked STAT-3 phosphorylation. In our experimental model, the combination of IPost and RIPer improved cardioprotection through the recruitment of the SAFE pathway. Our findings also indicate that cross talk exists between the RISK and SAFE pathways.


Asunto(s)
Poscondicionamiento Isquémico , Precondicionamiento Isquémico Miocárdico , Daño por Reperfusión Miocárdica/metabolismo , Transducción de Señal/fisiología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Western Blotting , Inhibidores Enzimáticos/farmacología , Miembro Posterior/irrigación sanguínea , Immunoblotting , Etiquetado Corte-Fin in Situ , Poscondicionamiento Isquémico/métodos , Precondicionamiento Isquémico Miocárdico/métodos , Masculino , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/fisiopatología , Ratas , Ratas Wistar , Transducción de Señal/efectos de los fármacos
10.
PLoS One ; 16(3): e0248554, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33765018

RESUMEN

Mitochondrial dynamics is a possible modulator of myocardial ischemia/reperfusion injuries (IRI). We previously reported that mice partially deficient in the fusion protein OPA1 exhibited higher IRI. Therefore, we investigated whether deficiency in the fission protein DRP1 encoded by Dnm1l gene would affect IRI in Dnm1l+/- mouse. After baseline characterization of the Dnm1l+/- mice heart, using echocardiography, electron microscopy, and oxygraphy, 3-month-old Dnm1l+/- and wild type (WT) mice were exposed to myocardial ischemia/reperfusion (I/R). The ischemic area-at-risk (AAR) and area of necrosis (AN) were delimited, and the infarct size was expressed by AN/AAR. Proteins involved in mitochondrial dynamics and autophagy were analyzed before and after I/R. Mitochondrial permeability transition pore (mPTP) opening sensitivity was assessed after I/R. Heart weight and left ventricular function were not significantly different in 3-, 6- and 12-month-old Dnm1l+/- mice than in WT. The cardiac DRP1 protein expression levels were 60% lower, whereas mitochondrial area and lipid degradation were significantly higher in Dnm1l+/- mice than in WT, though mitochondrial respiratory parameters and mPTP opening did not significantly differ. Following I/R, the infarct size was significantly smaller in Dnm1l+/- mice than in WT (34.6±3.1% vs. 44.5±3.3%, respectively; p<0.05) and the autophagic markers, LC3 II and P62 were significantly increased compared to baseline condition in Dnm1l+/- mice only. Altogether, data indicates that increasing fusion by means of Dnm1l deficiency was associated with protection against IRI, without alteration in cardiac or mitochondrial functions at basal conditions. This protection mechanism due to DRP1 haploinsufficiency increases the expression of autophagic markers.


Asunto(s)
Dinaminas/fisiología , Daño por Reperfusión Miocárdica/metabolismo , Animales , Dinaminas/genética , Haploinsuficiencia , Masculino , Ratones , Ratones Noqueados , Dinámicas Mitocondriales
11.
Am J Physiol Heart Circ Physiol ; 298(6): H1679-86, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20363897

RESUMEN

A noninvasive assessment of infarct size and transmural extension of myocardial infarction (TEMI) is fundamental in experimental models of ischemia-reperfusion. Conventional echocardiography parameters are limited in this purpose. This study was designed to examine whether speckle tracking imaging can be used in a rat model of ischemia-reperfusion to accurately detect the reduction of infarct size and TEMI induced by erythropoietin (EPO) as early as 24 h after reperfusion. Rats were randomly assigned to one of three groups: myocardial infarction (MI)-control group, 45 min ischemia followed by 24 h of reperfusion; MI-EPO group, similar surgery with a single bolus of EPO administered at the onset of reperfusion; and sham-operated group. Short-axis two-dimensional echocardiography was performed after reperfusion. Global radial (GS(r)) and circumferential (GS(cir)) strains were compared with infarct size and TEMI assessed after triphenyltetrazolium chloride staining. As a result, ejection fraction, shortening fraction, GS(r), and GS(cir) significantly correlated to infarct size, whereas only GS(r) and GS(cir) significantly correlated to TEMI. EPO significantly decreased infarct size (30.8 + or - 3.5 vs. 56.2 + or - 5.7% in MI-control, P < 0.001) and TEMI (0.37 + or - 0.05 vs. 0.77 + or - 0.05 in MI-control, P < 0.001). None of the conventional echocardiography parameters was significantly different between the MI-EPO and MI-control groups, whereas GS(r) was significantly higher in the MI-EPO group (29.1 + or - 4.7 vs. 16.4 + or - 3.3% in MI-control; P < 0.05). Furthermore, GS(cir) and GS(r) appeared to be the best parameters to identify a TEMI >0.75 24 h after reperfusion. In conclusion, these findings demonstrate the usefulness of speckle tracking imaging in the early evaluation of a cardioprotective strategy in a rat model of ischemia-reperfusion.


Asunto(s)
Ecocardiografía Doppler/métodos , Eritropoyetina/uso terapéutico , Infarto del Miocardio/diagnóstico por imagen , Infarto del Miocardio/tratamiento farmacológico , Daño por Reperfusión/complicaciones , Animales , Modelos Animales , Infarto del Miocardio/patología , Miocardio/patología , Necrosis , Ratas , Ratas Wistar , Terapia Recuperativa , Factores de Tiempo
12.
Shock ; 53(6): 730-736, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-31348147

RESUMEN

BACKGROUND: Acute myocardial infarction is a leading cause of death worldwide. Though highly beneficial, reperfusion of myocardium is associated with reperfusion injury. While indirect inhibition of Factor Xa has been shown to attenuate myocardial ischemia-reperfusion (I/R) injury, the underlying mechanism remains unclear. Our study sought to evaluate the effect of rivaroxaban (RIV), a direct inhibitor of Factor Xa, on myocardial I/R injury and determine its cellular targets. EXPERIMENTAL APPROACH: We used a rat model of 40-min coronary ligation followed by reperfusion. RIV (3 mg/kg) was given per os 1 h before reperfusion. Infarct size and myocardial proteic expression of survival pathways were assessed at 120 and 30 min of reperfusion, respectively. Plasmatic levels of P-selectin and von Willebrand factor were measured at 60 min of reperfusion. Cellular RIV effects were assessed using hypoxia-reoxygenation (H/R) models on human umbilical vein endothelial cells and on rat cardiomyoblasts (H9c2 cell line). KEY RESULTS: RIV decreased infarct size by 21% (42.9% vs. 54.2% in RIV-treated rats and controls respectively, P < 0.05) at blood concentrations similar to human therapeutic (387.7 ±â€Š152.3 ng/mL) levels. RIV had no effect on H/R-induced modulation of endothelial phenotype, nor did it alter myocardial activation of reperfusion injury salvage kinase and survivor activating factor enhancement pathways at 30 min after reperfusion. However, RIV exerted a cytoprotective effect on H9c2 cells submitted to H/R. CONCLUSIONS: RIV decreased myocardial I/R injury in rats at concentrations similar to human therapeutic ones. This protection was not associated with endothelial phenotype modulation but rather with potential direct cytoprotection on cardiomyocytes.


Asunto(s)
Cardiotónicos/uso terapéutico , Inhibidores del Factor Xa/uso terapéutico , Daño por Reperfusión Miocárdica/prevención & control , Rivaroxabán/uso terapéutico , Animales , Cardiotónicos/sangre , Cardiotónicos/farmacología , Factor Xa/metabolismo , Inhibidores del Factor Xa/sangre , Inhibidores del Factor Xa/farmacología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Masculino , Isquemia Miocárdica/complicaciones , Isquemia Miocárdica/terapia , Reperfusión Miocárdica , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Selectina-P/sangre , Ratas , Ratas Wistar , Reacción en Cadena en Tiempo Real de la Polimerasa , Rivaroxabán/sangre , Rivaroxabán/farmacología , Factor de von Willebrand/análisis
13.
Am J Physiol Heart Circ Physiol ; 297(6): H2035-43, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19617412

RESUMEN

Ischemic postconditioning (IPost) and erythropoietin (EPO) have been shown to attenuate myocardial reperfusion injury using similar signaling pathways. The aim of this study was to examine whether EPO is as effective as IPost in decreasing postischemic myocardial injury in both Langendorff-isolated-heart and in vivo ischemia-reperfusion rat models. Rat hearts were subjected to 25 min ischemia, followed by 30 min or 2 h of reperfusion in the isolated-heart study. Rats underwent 45 min ischemia, followed by 24 h of reperfusion in the in vivo study. In both studies, the control group (n=12; ischemia-reperfusion only) was compared with IPost (n=16; 3 cycles of 10 s reperfusion/10 s ischemia) and EPO (n=12; 1,000 IU/kg) at the onset of reperfusion. The following resulted. First, in the isolated hearts, IPost or EPO significantly improved postischemic recovery of left ventricular developed pressure. EPO induced better left ventricular developed pressure than IPost at 30 min of reperfusion (73.18+/-10.23 vs. 48.11+/-7.92 mmHg, P<0.05). After 2 h of reperfusion, the infarct size was significantly lower in EPO-treated hearts compared with IPost and control hearts (14.36+/-0.60%, 19.11+/-0.84%, and 36.21+/-4.20% of the left ventricle, respectively; P<0.05). GSK-3beta phosphorylation, at 30 min of reperfusion, was significantly higher with EPO compared with IPost hearts. Phosphatidylinositol 3-kinase and ERK1/2 inhibitors abolished both EPO- and IPost-mediated cardioprotection. Second, in vivo, IPost and EPO induced an infarct size reduction compared with control (40.5+/-3.6% and 28.9+/-3.1%, respectively, vs. 53.7+/-4.3% of the area at risk; P<0.05). Again, EPO decreased significantly more infarct size and transmurality than IPost (P<0.05). In conclusion, with the use of our protocols, EPO showed better protective effects than IPost against reperfusion injury through higher phosphorylation of GSK-3beta.


Asunto(s)
Cardiotónicos/uso terapéutico , Eritropoyetina/análogos & derivados , Infarto del Miocardio/prevención & control , Daño por Reperfusión Miocárdica/prevención & control , Miocardio/patología , Función Ventricular Izquierda/efectos de los fármacos , Animales , Circulación Coronaria/efectos de los fármacos , Darbepoetina alfa , Modelos Animales de Enfermedad , Eritropoyetina/farmacología , Femenino , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Frecuencia Cardíaca/efectos de los fármacos , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Infarto del Miocardio/enzimología , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Daño por Reperfusión Miocárdica/enzimología , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/fisiopatología , Miocardio/enzimología , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Wistar , Factores de Tiempo , Presión Ventricular/efectos de los fármacos
14.
Shock ; 51(6): 795-799, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-29846359

RESUMEN

Ischemia-reperfusion (I/R) injury is a leading cause of acute renal dysfunction. Remote ischemic conditioning (rIC) is known to protect organs exposed to I/R. We sought to investigate whether rIC would influence renal function recovery in a severe renal I/R injury rat model. Rats were randomly assigned to four experimental groups following median laparotomy and right nephrectomy: Sham (n = 6); 30-min left renal ischemia (RI) only (n = 20); RI + rIC (n = 20) (four 5-min cycles of limb ischemia interspersed with 5-min limb reperfusion during RI); and RI + erythropoietin pretreatment (EPO) (n = 20). Renal function was evaluated by assessing blood urea nitrogen (BUN) and serum creatinine (Cr) levels before surgery and after 1 day of reperfusion. All animals were monitored for 7 days for survival analysis. BUN and Cr baseline levels did not significantly differ between groups. At day 1, BUN and Cr were significantly higher than baseline values in all groups. BUN and Cr levels did not significantly differ at day 1 between RI and RI + rIC (P = 0.68). Conversely, EPO pretreatment injected 60 min before RI was associated with lower BUN and Cr levels compared with RI (P < 0.001 and P = 0.003, respectively) and RI + rIC (P < 0.001 and P = 0.001, respectively). In addition, 7-day survival rates were significantly higher in the Sham group (100%) compared with RI (50%; P = 0.039 vs. Sham) and RI + rIC (45%; P = 0.026 vs. Sham). Conversely, survival rate did not significantly differ between the Sham and RI + EPO groups (70%, P = 0.15). In conclusion, rIC affected neither acute renal dysfunction nor early mortality in a severe I/R renal injury rat model, contrary to EPO pretreatment.


Asunto(s)
Precondicionamiento Isquémico , Enfermedades Renales , Riñón , Daño por Reperfusión , Animales , Modelos Animales de Enfermedad , Riñón/metabolismo , Riñón/patología , Riñón/fisiopatología , Enfermedades Renales/metabolismo , Enfermedades Renales/patología , Enfermedades Renales/fisiopatología , Enfermedades Renales/prevención & control , Masculino , Ratas , Ratas Sprague-Dawley , Daño por Reperfusión/metabolismo , Daño por Reperfusión/parasitología , Daño por Reperfusión/patología , Daño por Reperfusión/prevención & control
15.
Catheter Cardiovasc Interv ; 72(4): 513-21, 2008 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-18814226

RESUMEN

OBJECTIVES: We tested the hypothesis that unloading the left ventricle with intra-aortic balloon counter-pulsation just prior to reperfusion provides infarct salvage compared with left ventricular (LV) unloading postreperfusion or reperfusion alone. BACKGROUND: Previous reports demonstrated infarct salvage with complete LV unloading with an LVAD prior to reperfusion; however, partial LV unloading using intra-aortic balloon pumps (IABPs) has not been evaluated. METHODS: Twenty-eight Yorkshire pigs were subjected to 1 hr of left anterior descending artery occlusion and 4 hr of reperfusion. An IABP was inserted and activated just prior to reperfusion (IABP-Pre), or 15 min after reperfusion (IABP-Post), or not at all (control). RESULTS: At baseline, the hemodynamic data were similar in the three groups. Myocardial infarct size expressed a percentage of zone at risk in control animals was 44.9% +/- 4.8%, IAPB-Pre group 20.9% +/- 5.1% (P < 0.05 compared to control), and IABP-Post group 33.2 +/- 6.1% (P = 0.16 vs. control group). There was a correlation between transcardiac endothelin-1 release at 15 min postreperfusion and infarct size (r = 0.59). CONCLUSION: LV unloading with an IABP prior to reperfusion reduces the extent of myocardial necrosis in hearts subjected to 1 hr of left anterior descending artery occlusion and 4 hr of reperfusion compared with either reperfusion alone or LV unloading after reperfusion. Inhibition of myocardial ET-1 release by LV unloading may be a significant mechanism of myocardial protection. These data suggest that in high-risk STEMI patients, IABP unloading prior to reperfusion might be more beneficial than IABP placement postreperfusion.


Asunto(s)
Angioplastia Coronaria con Balón , Endotelina-1/metabolismo , Contrapulsador Intraaórtico , Infarto del Miocardio/prevención & control , Daño por Reperfusión Miocárdica/prevención & control , Miocardio/metabolismo , Función Ventricular Izquierda , Angioplastia Coronaria con Balón/efectos adversos , Animales , Circulación Coronaria , Modelos Animales de Enfermedad , Corazón Auxiliar , Hemodinámica , Contrapulsador Intraaórtico/instrumentación , Infarto del Miocardio/metabolismo , Infarto del Miocardio/fisiopatología , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/fisiopatología , Miocardio/patología , Necrosis , Porcinos , Factores de Tiempo
16.
J Cardiovasc Pharmacol Ther ; 23(5): 446-455, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29658326

RESUMEN

BACKGROUND: Inflammation plays a crucial role in the pathophysiology of myocardial ischemia/reperfusion (I/R) injury. A clinical trial has recently reported a smaller infarct size in a cohort of patients with ST-segment elevation myocardial infarction (MI) treated with a short colchicine course. The mechanism underlying colchicine-induced cardioprotection in the early MI phase remains unclear. We hypothesized that a short pretreatment with colchicine could induce acute beneficial effects by protecting the heart against inflammation in myocardial I/R injury. METHODS AND RESULTS: Rats were subjected to 40-minute left anterior descending coronary occlusion, followed by 120-minute reperfusion. Colchicine (0.3 mg/kg) or a vehicle was administered per os 24 hours and immediately before surgery. Infarct size was significantly reduced in the colchicine group (35.6% ± 3.0% vs 46.6% ± 3.3%, P < .05). The beneficial effects of colchicine were associated with an increased systemic interleukin-10 (IL-10) level and decreased cardiac transforming growth factor-ß level. Interleukin-1ß was found to increase in a "time of reperfusion"-dependent manner. Colchicine inhibited messenger RNA expression of caspase-1 and pro-IL-18. Interleukin-1ß injected 10 minutes prior to myocardial ischemia induced greater infarct size (58.0% ± 2.0%, P < .05) as compared to the vehicle. Colchicine combined to IL-1ß injection significantly decreased infarct size (47.1% ± 2.2%, P < .05) as compared to IL-1ß alone, while colchicine alone exhibited a significantly more marked cardioprotective effect than the colchicine-IL-1ß association. CONCLUSION: The cardioprotection induced by a short colchicine pretreatment was associated with an anti-inflammatory effect in the early reperfusion phase in our rat MI model.


Asunto(s)
Antiinflamatorios/farmacología , Colchicina/farmacología , Infarto del Miocardio/prevención & control , Daño por Reperfusión Miocárdica/prevención & control , Miocitos Cardíacos/efectos de los fármacos , Animales , Caspasa 1/metabolismo , Modelos Animales de Enfermedad , Quimioterapia Combinada , Inflamasomas/efectos de los fármacos , Inflamasomas/metabolismo , Interleucina-10/sangre , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Interleucina-1beta/farmacología , Masculino , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Proteína con Dominio Pirina 3 de la Familia NLR/efectos de los fármacos , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Ratas Wistar , Factores de Tiempo , Factor de Crecimiento Transformador beta1/metabolismo
17.
Cell Calcium ; 39(1): 47-55, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16243395

RESUMEN

Diabetes mellitus causes multiple cardiovascular complications. Previous studies have shown that prolonged exposure (96 h) of human umbilical vein endothelial cells (HUVECs) to hyperglycemia causes a significant increase in apoptosis. We report here that this increase in apoptosis is associated with an increase in Ca(2+) current (whole cell patch-clamp recorded) resulting from Ca(2+) entry mediated by store-operated channels (SOCs). The number of apoptotic cells after prolonged high glucose (HG, 30 mmol/L) exposure was significantly reduced in the presence of the SOC inhibitor 2-APB or of La(3+). A marked increase (approximately 80%) in Ca(2+)-dependent calcineurin (CN-A) phosphatase activity also occurred after prolonged HG exposure. Prolonged HG exposure-induced increase in CN-A activity was prevented by 2-APB, and selective CN-A phosphatase inhibition by FK506 or calmodulin inhibition by calmidazolium decreased HG-induced apoptosis. Blocking hydrogen peroxide production using catalase or inhibiting the tyrosine kinase pp60(src) during prolonged exposure to HG, resulted in a marked decrease in apoptosis and was further associated with a significant reduction in CN-A phosphatase activity. The results demonstrate a significant role for Ca(2+) entry in HG-induced apoptosis in HUVECs, and suggest that this role is mediated via H(2)O(2) generation and the action of the Ca(2+)-activated protein phosphatase calcineurin.


Asunto(s)
Apoptosis , Calcineurina/metabolismo , Calcio/metabolismo , Endotelio Vascular/metabolismo , Glucosa/farmacología , Compuestos de Boro/farmacología , Canales de Calcio/efectos de los fármacos , Canales de Calcio/metabolismo , Calmodulina/antagonistas & inhibidores , Catalasa/farmacología , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Humanos , Peróxido de Hidrógeno/antagonistas & inhibidores , Peróxido de Hidrógeno/metabolismo , Hiperglucemia/metabolismo , Hiperglucemia/fisiopatología , Imidazoles/farmacología , Técnicas de Placa-Clamp , Monoéster Fosfórico Hidrolasas/metabolismo , Proteínas Proto-Oncogénicas pp60(c-src)/antagonistas & inhibidores , Tacrolimus/farmacología , Venas Umbilicales/citología
18.
Shock ; 45(2): 192-7, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26555744

RESUMEN

Remote ischemic preconditioning (RIPC) has emerged as an attractive strategy to protect the heart against ischemia-reperfusion (I/R) injury. The mechanisms by which remote ischemic conditioning (RIC) is protective are to date unknown, yet a well-accepted theory holds that the mitochondria play a central role. Mitochondria are dynamic organelles that undergo fusion and fission. Interventions that decrease mitochondrial fission or increase mitochondrial fusion have been associated with reduced I/R injury. However, whether RIPC influences mitochondrial dynamics or not has yet to be ascertained.We sought to determine the role played by mitochondrial dynamics in RIPC-induced cardioprotection. Male adult rats exposed in vivo to myocardial I/R were assigned to one of two groups, either undergoing 40 min of myocardial ischemia followed by 120 min of reperfusion (MI group) or four 5-min cycles of limb ischemia interspersed by 5 min of limb reperfusion, immediately prior to myocardial ischemia and 120 min of reperfusion (MI+RIPC group). After reperfusion, infarct size was assessed and myocardial tissue was analyzed by Western blot and electron microscopy. RIPC induced smaller infarct size (-28%), increased mitochondrial fusion protein OPA1, and preserved mitochondrial morphology. These findings suggest that mitochondrial dynamics play a role in the mechanisms of RIPC-induced cardioprotection.


Asunto(s)
Precondicionamiento Isquémico , Dinámicas Mitocondriales/efectos de los fármacos , Daño por Reperfusión Miocárdica/prevención & control , Animales , Modelos Animales de Enfermedad , Masculino , Microscopía Electrónica , Infarto del Miocardio/prevención & control , Miocardio/metabolismo , Miocardio/ultraestructura , Ratas , Ratas Wistar
19.
J Am Heart Assoc ; 5(9)2016 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-27664804

RESUMEN

BACKGROUND: Remote ischemic preconditioning (RIPC) is an attractive therapeutic procedure for protecting the heart against ischemia/reperfusion injury. Despite evidence of humoral mediators transported through the circulation playing a critical role, their actual identities so far remain unknown. We sought to identify plasmatic RIPC-induced metabolites that may play a role. METHODS AND RESULTS: Rat plasma samples from RIPC and control groups were analyzed using a targeted metabolomic approach aimed at measuring 188 metabolites. Principal component analysis and orthogonal partial least-squares discriminant analysis were used to identify the metabolites that discriminated between groups. Plasma samples from 50 patients subjected to RIPC were secondarily explored to confirm the results obtained in rats. Finally, a combination of the metabolites that were significantly increased in both rat and human plasma was injected prior to myocardial ischemia/reperfusion in rats. In the rat samples, 124 molecules were accurately quantified. Six metabolites (ornithine, glycine, kynurenine, spermine, carnosine, and serotonin) were the most significant variables for marked differentiation between the RIPC and control groups. In human plasma, analysis confirmed ornithine decrease and kynurenine and glycine increase following RIPC. Injection of the glycine and kynurenine alone or in combination replicated the protective effects of RIPC seen in rats. CONCLUSIONS: We have hereby reported significant variations in a cocktail of amino acids and biogenic amines after remote ischemic preconditioning in both rat and human plasma. CLINICAL TRIAL REGISTRATION: URL: http://www.clinicaltrials.gov. Unique identifier: NCT01390129.

20.
PLoS One ; 11(10): e0164066, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27723783

RESUMEN

BACKGROUND: Recent data suggests the involvement of mitochondrial dynamics in cardiac ischemia/reperfusion (I/R) injuries. Whilst excessive mitochondrial fission has been described as detrimental, the role of fusion proteins in this context remains uncertain. OBJECTIVES: To investigate whether Opa1 (protein involved in mitochondrial inner-membrane fusion) deficiency affects I/R injuries. METHODS AND RESULTS: We examined mice exhibiting Opa1delTTAG mutations (Opa1+/-), showing 70% Opa1 protein expression in the myocardium as compared to their wild-type (WT) littermates. Cardiac left-ventricular systolic function assessed by means of echocardiography was observed to be similar in 3-month-old WT and Opa1+/- mice. After subjection to I/R, infarct size was significantly greater in Opa1+/- than in WTs both in vivo (43.2±4.1% vs. 28.4±3.5%, respectively; p<0.01) and ex vivo (71.1±3.2% vs. 59.6±8.5%, respectively; p<0.05). No difference was observed in the expression of other main fission/fusion protein, oxidative phosphorylation, apoptotic markers, or mitochondrial permeability transition pore (mPTP) function. Analysis of calcium transients in isolated ventricular cardiomyocytes demonstrated a lower sarcoplasmic reticulum Ca2+ uptake, whereas cytosolic Ca2+ removal from the Na+/Ca2+ exchanger (NCX) was increased, whilst SERCA2a, phospholamban, and NCX protein expression levels were unaffected in Opa1+/- compared to WT mice. Simultaneous whole-cell patch-clamp recordings of mitochondrial Ca2+ movements and ventricular action potential (AP) showed impairment of dynamic mitochondrial Ca2+ uptake and a marked increase in the AP late repolarization phase in conjunction with greater occurrence of arrhythmia in Opa1+/- mice. CONCLUSION: Opa1 deficiency was associated with increased sensitivity to I/R, imbalance in dynamic mitochondrial Ca2+ uptake, and subsequent increase in NCX activity.


Asunto(s)
Calcio/metabolismo , GTP Fosfohidrolasas/metabolismo , Mitocondrias Cardíacas/metabolismo , Dinámicas Mitocondriales , Daño por Reperfusión Miocárdica/metabolismo , Miocitos Cardíacos/metabolismo , Animales , Modelos Animales de Enfermedad , GTP Fosfohidrolasas/genética , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Ratones , Ratones Mutantes , Mitocondrias Cardíacas/genética , Proteínas de Transporte de Membrana Mitocondrial/genética , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Poro de Transición de la Permeabilidad Mitocondrial , Daño por Reperfusión Miocárdica/genética , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/genética , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA