Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.753
Filtrar
1.
J Dev Orig Health Dis ; 15: e9, 2024 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-38721989

RESUMEN

Sodium overload during childhood impairs baroreflex sensitivity and increases arterial blood pressure and heart rate in adulthood; these effects persist even after high-salt diet (HSD) withdrawal. However, the literature lacks details on the effects of HSD during postnatal phases on cardiac ischemia/reperfusion responses in adulthood. The current study aimed to elucidate the impact of HSD during infancy adolescence on isolated heart function and cardiac ischemia/reperfusion responses in adulthood. Male 21-day-old Wistar rats were treated for 60 days with hypertonic saline solution (NaCl; 0.3M; experimental group) or tap water (control group). Subsequently, both groups were maintained on a normal sodium diet for 30 days. Subsequently, the rats were euthanized, and their hearts were isolated and perfused according to the Langendorff technique. After 30 min of the basal period, the hearts were subjected to 20 min of anoxia, followed by 20 min of reperfusion. The basal contractile function was unaffected by HSD. However, HSD elevated the left ventricular end-diastolic pressure during reperfusion (23.1 ± 5.2 mmHg vs. 11.6 ± 1.4 mmHg; p < 0.05) and increased ectopic incidence period during reperfusion (208.8 ± 32.9s vs. 75.0 ± 7.8s; p < 0.05). In conclusion, sodium overload compromises cardiac function after reperfusion events, diminishes ventricular relaxation, and increases the severity of arrhythmias, suggesting a possible arrhythmogenic effect of HSD in the postnatal phases.


Asunto(s)
Arritmias Cardíacas , Daño por Reperfusión Miocárdica , Ratas Wistar , Animales , Ratas , Arritmias Cardíacas/etiología , Arritmias Cardíacas/fisiopatología , Masculino , Daño por Reperfusión Miocárdica/etiología , Daño por Reperfusión Miocárdica/fisiopatología , Diástole/fisiología , Cloruro de Sodio Dietético/efectos adversos , Frecuencia Cardíaca/fisiología
2.
Iran Biomed J ; 28(2&3): 59-70, 2024 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-38770843

RESUMEN

Despite the unconditional success achieved in the treatment and prevention of AMI over the past 40 years, mortality in this disease remains high. Hence, it is necessary to develop novel drugs with mechanism of action different from those currently used in clinical practices. Studying the molecular mechanisms involved in the cardioprotective effect of adapting to cold could contribute to the development of drugs that increase cardiac tolerance to the impact of ischemia/reperfusion. An analysis of the published data shows that the long-term human stay in the Far North contributes to the occurrence of cardiovascular diseases. At the same time, chronic and continuous exposure to cold increases tolerance of the rat heart to ischemia/ reperfusion. It has been demonstrated that the cardioprotective effect of cold adaptation depends on the activation of ROS production, stimulation of the ß2-adrenergic receptor and protein kinase C, MPT pore closing, and KATP channel.


Asunto(s)
Adaptación Fisiológica , Frío , Humanos , Animales , Sistema Cardiovascular/fisiopatología , Sistema Cardiovascular/efectos de los fármacos , Daño por Reperfusión Miocárdica/fisiopatología , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión/fisiopatología , Daño por Reperfusión/metabolismo , Especies Reactivas de Oxígeno/metabolismo
3.
Eur J Pharmacol ; 974: 176633, 2024 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-38703975

RESUMEN

Cardiac arrest (CA) remains a leading cause of death, with suboptimal survival rates despite efforts involving cardiopulmonary resuscitation and advanced life-support technology. Post-resuscitation myocardial dysfunction (PRMD) is an important determinant of patient outcomes. Myocardial ischemia/reperfusion injury underlies this dysfunction. Previous reports have shown that ruthenium red (RR) has a protective effect against cardiac ischemia-reperfusion injury; however, its precise mechanism of action in PRMD remains unclear. This study investigated the effects of RR on PRMD and analyzed its underlying mechanisms. Ventricular fibrillation was induced in rats, which were then subjected to cardiopulmonary resuscitation to establish an experimental CA model. At the onset of return of spontaneous circulation, RR (2.5 mg/kg) was administered intraperitoneally. Our study showed that RR improved myocardial function and reduced the production of oxidative stress markers such as malondialdehyde (MDA), glutathione peroxidase (GSSG), and reactive oxygen species (ROS) production. RR also helped maintain mitochondrial structure and increased ATP and GTP levels. Additionally, RR effectively attenuated myocardial apoptosis. Furthermore, we observed downregulation of proteins closely related to mitophagy, including ubiquitin-specific protease 33 (USP33) and P62, whereas LC3B (microtubule-associated protein light chain 3B) was upregulated. The upregulation of mitophagy may play a critical role in reducing myocardial injury. These results demonstrate that RR may attenuate PRMD by promoting mitophagy through the inhibition of USP33. These effects are likely mediated through diverse mechanisms, including antioxidant activity, apoptosis suppression, and preservation of mitochondrial integrity and energy metabolism. Consequently, RR has emerged as a promising therapeutic approach for addressing post-resuscitation myocardial dysfunction.


Asunto(s)
Modelos Animales de Enfermedad , Paro Cardíaco , Mitofagia , Ratas Sprague-Dawley , Rojo de Rutenio , Animales , Mitofagia/efectos de los fármacos , Paro Cardíaco/complicaciones , Paro Cardíaco/tratamiento farmacológico , Paro Cardíaco/metabolismo , Paro Cardíaco/fisiopatología , Ratas , Masculino , Rojo de Rutenio/farmacología , Rojo de Rutenio/uso terapéutico , Estrés Oxidativo/efectos de los fármacos , Ubiquitina Tiolesterasa/metabolismo , Reanimación Cardiopulmonar , Regulación hacia Arriba/efectos de los fármacos , Miocardio/patología , Miocardio/metabolismo , Apoptosis/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Daño por Reperfusión Miocárdica/fisiopatología
4.
Bull Exp Biol Med ; 176(5): 539-542, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38717565

RESUMEN

Coronary occlusion (45 min) and reperfusion (120 min) in male Wistar rats in vivo, as well as total ischemia (45 min) of an isolated rat heart followed by reperfusion (30 min) were reproduced. The selective δ2-opioid receptor agonist deltorphin II (0.12 mg/kg and 152 nmol/liter) was administered intravenously 5 min before reperfusion in vivo or added to the perfusion solution at the beginning of reperfusion of the isolated heart. The peripheral opioid receptor antagonist naloxone methiodide and δ2-opioid receptor antagonist naltriben were used in doses of 5 and 0.3 mg/kg, respectively. It was found that the infarct-limiting effect of deltorphin II is associated with the activation of δ2-opioid receptors. We have demonstrated that deltorphin II can improve the recovery of the contractility of the isolated heart after total ischemia.


Asunto(s)
Daño por Reperfusión Miocárdica , Ratas Wistar , Receptores Opioides delta , Animales , Masculino , Receptores Opioides delta/agonistas , Receptores Opioides delta/metabolismo , Ratas , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/fisiopatología , Oligopéptidos/farmacología , Contracción Miocárdica/efectos de los fármacos , Corazón/efectos de los fármacos , Antagonistas de Narcóticos/farmacología , Infarto del Miocardio/metabolismo , Infarto del Miocardio/fisiopatología , Infarto del Miocardio/tratamiento farmacológico , Miocardio/metabolismo
5.
J Am Heart Assoc ; 13(9): e033744, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38686853

RESUMEN

BACKGROUND: The heart can metabolize the microbiota-derived short-chain fatty acid butyrate. Butyrate may have beneficial effects in heart failure, but the underlying mechanisms are unknown. We tested the hypothesis that butyrate elevates cardiac output by mechanisms involving direct stimulation of cardiac contractility and vasorelaxation in rats. METHODS AND RESULTS: We examined the effects of butyrate on (1) in vivo hemodynamics using parallel echocardiographic and invasive blood pressure measurements, (2) isolated perfused hearts in Langendorff systems under physiological conditions and after ischemia and reperfusion, and (3) isolated coronary arteries mounted in isometric wire myographs. We tested Na-butyrate added to injection solutions or physiological buffers and compared its effects with equimolar doses of NaCl. Butyrate at plasma concentrations of 0.56 mM increased cardiac output by 48.8±14.9%, stroke volume by 38.5±12.1%, and left ventricular ejection fraction by 39.6±6.2%, and lowered systemic vascular resistance by 33.5±6.4% without affecting blood pressure or heart rate in vivo. In the range between 0.1 and 5 mM, butyrate increased left ventricular systolic pressure by up to 23.7±3.4% in isolated perfused hearts and by 9.4±2.9% following ischemia and reperfusion, while reducing myocardial infarct size by 81.7±16.9%. Butyrate relaxed isolated coronary septal arteries concentration dependently with an EC50=0.57 mM (95% CI, 0.23-1.44). CONCLUSIONS: We conclude that butyrate elevates cardiac output through mechanisms involving increased cardiac contractility and vasorelaxation. This effect of butyrate was not associated with adverse myocardial injury in damaged hearts exposed to ischemia and reperfusion.


Asunto(s)
Butiratos , Cardiotónicos , Contracción Miocárdica , Vasodilatación , Vasodilatadores , Función Ventricular Izquierda , Animales , Masculino , Contracción Miocárdica/efectos de los fármacos , Función Ventricular Izquierda/efectos de los fármacos , Vasodilatación/efectos de los fármacos , Cardiotónicos/farmacología , Butiratos/farmacología , Vasodilatadores/farmacología , Preparación de Corazón Aislado , Ratas , Daño por Reperfusión Miocárdica/fisiopatología , Daño por Reperfusión Miocárdica/prevención & control , Daño por Reperfusión Miocárdica/metabolismo , Gasto Cardíaco/efectos de los fármacos , Volumen Sistólico/efectos de los fármacos , Ratas Wistar , Vasos Coronarios/efectos de los fármacos , Vasos Coronarios/fisiopatología , Relación Dosis-Respuesta a Droga , Modelos Animales de Enfermedad , Ratas Sprague-Dawley
6.
Cardiovasc Toxicol ; 24(5): 481-498, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38647950

RESUMEN

The hearts of subjects with diabetes are vulnerable to ischemia-reperfusion injury (IRI). In contrast, experimentally rodent hearts have been shown to be more resistant to IRI at the very early stages of diabetes induction than the heart of the non-diabetic control mice, and the mechanism is largely unclear. Ferroptosis has recently been shown to play an important role in myocardial IRI including that in diabetes, while the specific mechanisms are still unclear. Non-diabetic control (NC) and streptozotocin-induced diabetic (DM) mice were treated with the antioxidant N-acetylcysteine (NAC) in drinking water for 4 week starting at 1 week after diabetes induction. Mice were subjected to myocardial IRI induced by occluding the coronary artery for 30 min followed by 2 h of reperfusion, subsequently at 1, 2, and 5 week of diabetes induction. The post-ischemic myocardial infarct size in the DM mice was smaller than that in NC mice at 1 week of diabetes but greater than that in the NC mice at 2 and 5 week of diabetes, which were associated with a significant increase of ferroptosis at 2 and 5 week but a significant reduction of ferroptosis at 1 week of diabetes. NAC significantly attenuated post-ischemic ferroptosis as well as oxidative stress and reduced infarct size at 2 and 5 week of diabetes. Application of erastin, a ferroptosis inducer, reversed the cardioprotective effects of NAC. It is concluded that increased oxidative stress and ferroptosis are the major factors attributable to the increased vulnerability to myocardial IRI in diabetes and that attenuation of ferroptosis represents a major mechanism whereby NAC confers cardioprotection against myocardial IRI in diabetes.


Asunto(s)
Acetilcisteína , Antioxidantes , Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 1 , Ferroptosis , Ratones Endogámicos C57BL , Daño por Reperfusión Miocárdica , Animales , Daño por Reperfusión Miocárdica/prevención & control , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/fisiopatología , Acetilcisteína/farmacología , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/complicaciones , Masculino , Diabetes Mellitus Tipo 1/complicaciones , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Diabetes Mellitus Tipo 1/metabolismo , Antioxidantes/farmacología , Ferroptosis/efectos de los fármacos , Infarto del Miocardio/prevención & control , Infarto del Miocardio/patología , Infarto del Miocardio/metabolismo , Infarto del Miocardio/fisiopatología , Infarto del Miocardio/tratamiento farmacológico , Factores de Tiempo , Miocardio/patología , Miocardio/metabolismo , Ratones , Estrés Oxidativo/efectos de los fármacos
7.
Fundam Clin Pharmacol ; 38(3): 489-501, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38311344

RESUMEN

BACKGROUND: The high mortality rate of patients with acute myocardial infarction (AMI) remains the most pressing issue of modern cardiology. Over the past 10 years, there has been no significant reduction in mortality among patients with AMI. It is quite obvious that there is an urgent need to develop fundamentally new drugs for the treatment of AMI. Angiotensin 1-7 has some promise in this regard. OBJECTIVE: The objective of this article is analysis of published data on the cardioprotective properties of angiotensin 1-7. METHODS: PubMed, Scopus, Science Direct, and Google Scholar were used to search articles for this study. RESULTS: Angiotensin 1-7 increases cardiac tolerance to ischemia/reperfusion and mitigates adverse remodeling of the heart. Angiotensin 1-7 can prevent not only ischemic but also reperfusion cardiac injury. The activation of the Mas receptor plays a key role in these effects of angiotensin 1-7. Angiotensin 1-7 alleviates Ca2+ overload of cardiomyocytes and reactive oxygen species production in ischemia/reperfusion (I/R) of the myocardium. It is possible that both effects are involved in angiotensin 1-7-triggered cardiac tolerance to I/R. Furthermore, angiotensin 1-7 inhibits apoptosis of cardiomyocytes and stimulates autophagy of cells. There is also indirect evidence suggesting that angiotensin 1-7 inhibits ferroptosis in cardiomyocytes. Moreover, angiotensin 1-7 possesses anti-inflammatory properties, possibly achieved through NF-kB activity inhibition. Phosphoinositide 3-kinase, Akt, and NO synthase are involved in the infarct-reducing effect of angiotensin 1-7. However, the specific end-effector of the cardioprotective impact of angiotensin 1-7 remains unknown. CONCLUSION: The molecular nature of the end-effector of the infarct-limiting effect of angiotensin 1-7 has not been elucidated. Perhaps, this end-effector is the sarcolemmal KATP channel or the mitochondrial KATP channel.


Asunto(s)
Angiotensina I , Daño por Reperfusión Miocárdica , Fragmentos de Péptidos , Transducción de Señal , Angiotensina I/farmacología , Fragmentos de Péptidos/farmacología , Humanos , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Daño por Reperfusión Miocárdica/fisiopatología , Animales , Transducción de Señal/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Infarto del Miocardio/tratamiento farmacológico , Infarto del Miocardio/metabolismo , Remodelación Ventricular/efectos de los fármacos , Cardiotónicos/farmacología , Cardiotónicos/uso terapéutico , Apoptosis/efectos de los fármacos
8.
Can J Physiol Pharmacol ; 102(5): 331-341, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38118123

RESUMEN

Extracellular potassium concentration might modify electrophysiological properties in the border zone of ischemic myocardium. We evaluated the depolarization and repolarization characteristics across the ischemic-normal border under [K+] variation. Sixty-four-lead epicardial mapping was performed in 26 rats ([K+] 2.3-6.4 mM) in a model of acute ischemia/reperfusion. The animals with [K+] < 4.7 mM (low-normal potassium) had an ischemic zone with ST-segment elevation and activation delay, a border zone with ST-segment elevation and no activation delay, and a normal zone without electrophysiological abnormalities. The animals with [K+] >4.7 mM (normal-high potassium) had only the ischemic and normal zones and no transitional area. Activation-repolarization intervals and local conduction velocities were inversely associated with [K+] in linear regression analysis with adjustment for the zone of myocardium. The reperfusion extrasystolic burden (ESB) was greater in the low-normal as compared to normal-high potassium animals. Ventricular tachycardia/fibrillation incidence did not differ between the groups. In patch-clamp experiments, hypoxia shortened action potential duration at 5.4 mM but not at 1.3 mM of [K+]. IK(ATP) current was lower at 1.3 mM than at 5.4 mM of [K+]. We conclude that the border zone formation in low-normal [K+] was associated with attenuation of IK(ATP) response to hypoxia and increased reperfusion ESB.


Asunto(s)
Potenciales de Acción , Isquemia Miocárdica , Potasio , Animales , Potasio/sangre , Potasio/metabolismo , Masculino , Ratas , Isquemia Miocárdica/fisiopatología , Isquemia Miocárdica/sangre , Isquemia Miocárdica/metabolismo , Potenciales de Acción/fisiología , Daño por Reperfusión Miocárdica/sangre , Daño por Reperfusión Miocárdica/fisiopatología , Daño por Reperfusión Miocárdica/metabolismo , Ratas Wistar
9.
Pharm Biol ; 60(1): 553-561, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35244510

RESUMEN

CONTEXT: Acacetin is a natural source of flavonoids with anti-inflammatory and antioxidant effects. OBJECTIVE: This study determines acacetin's protective effect and mechanism on myocardial ischaemia/reperfusion (I/R) injury. MATERIALS AND METHODS: Sprague-Dawley rats were divided into sham and I/R injury and treatment with acacetin. Acacetin (10 mg/kg) was subcutaneously injected for 7 days. ECG and echocardiography were conducted to determine arrhythmia and heart function. The pathological characters of the heart were determined with triphenyl tetrazolium chloride staining, Haematoxylin & Eosin staining, and Masson staining. Expression of proteins in infarct tissues was examined with western blots. RESULTS: Administrated with acacetin in I/R rats significantly reduced the arrhythmia score from 4.90 to 2.50 and the reperfusion arrhythmia score from 3.79 to 1.82 in the vehicle or the acacetin group, respectively. LVEF was improved from 33.5% in the I/R group to 43.7% in the acacetin group, LVFS was increased from 16.4% to 24.5%, LVIDs was decreased from 6.5 to 5.3 mm. The inflammatory cell infiltration, myocardial fibrosis, and collagen 1 and 3 were reduced by acacetin. Acacetin promoted SOD and decreased MDA. In myocardial tissues, the expression level of TLR4 and IL-6 were restrained, and IL-10 was promoted. Apoptotic protein Bax was suppressed, and anti-apoptotic protein Bcl-2 was promoted in the acacetin group. Interestingly, the transcription factor Nrf-2/HO-1 pathway was also reversed by acacetin. DISCUSSION AND CONCLUSION: Our findings indicated that acacetin has a potential therapeutic effect in clinical application on treating I/R-induced heart injury.


Asunto(s)
Apoptosis/efectos de los fármacos , Flavonas/farmacología , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Estrés Oxidativo/efectos de los fármacos , Animales , Antiinflamatorios/farmacología , Antioxidantes/farmacología , Hemo Oxigenasa (Desciclizante)/metabolismo , Masculino , Daño por Reperfusión Miocárdica/fisiopatología , Factor 2 Relacionado con NF-E2/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ratas , Ratas Sprague-Dawley , Proteína X Asociada a bcl-2/metabolismo
10.
Pharm Biol ; 60(1): 384-393, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35188856

RESUMEN

CONTEXT: Therapeutic lymphangiogenesis is a new treatment for cardiovascular diseases. Our previous study showed M2b macrophages can alleviate myocardial ischaemia/reperfusion injury (MI/RI). However, the relation between M2b macrophages and lymphangiogenesis is not clear. OBJECTIVE: To investigate the effects of M2b macrophages on lymphangiogenesis after MI/RI. MATERIALS AND METHODS: Forty male Sprague-Dawley (SD) rats were randomized into Sham operation group (control, n = 8), MI/RI group (n = 16) and M2b macrophage transplantation group (n = 16). M2b macrophages (1 × 106) in 100 µL of normal saline or the same volume of vehicle was injected into the cardiac ischaemic zone. Two weeks later, echocardiography and lymphatic counts were performed, and the extent of myocardial fibrosis and the expression of vascular endothelial growth factor C (VEGFC) and VEGF receptor 3 (VEGFR3) were determined. In vitro, lymphatic endothelial cells (LECs) were cultured with M2b macrophages for 6-24 h, and the proliferation, migration and tube formation of the LECs were assessed. RESULTS: In vivo, M2b macrophage transplantation increased the level of lymphangiogenesis 2.11-fold, reduced 4.42% fibrosis, improved 18.65% left ventricular ejection fraction (LVEF) and upregulated the expressions of VEGFC and VEGFR3. In vitro, M2b macrophage increased the proliferation, migration, tube formation and VEGFC expression of LECs. M2b macrophage supernatant upregulated VEGFR3 expression of LECs. DISCUSSION AND CONCLUSIONS: Our study shows that M2b macrophages can promote lymphangiogenesis to reduce myocardial fibrosis and improve heart function, suggesting the possible use of M2b macrophage for myocardial protection therapy.


Asunto(s)
Linfangiogénesis/fisiología , Macrófagos/trasplante , Daño por Reperfusión Miocárdica/terapia , Animales , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Modelos Animales de Enfermedad , Ecocardiografía , Células Endoteliales/metabolismo , Fibrosis , Masculino , Daño por Reperfusión Miocárdica/fisiopatología , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Volumen Sistólico/fisiología , Función Ventricular Izquierda/fisiología
11.
Molecules ; 27(3)2022 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-35164296

RESUMEN

The transient vanilloid receptor potential type 1 (TRPV1) regulates neuronal and vascular functions mediated by nitric oxide (NO) and by the calcitonin gene-related peptide (CGRP). Here, we study the participation of TRPV1 in the regulation of myocardial injury caused by ischemia-reperfusion and in the control of NO, tetrahydrobiopterin (BH4), the cGMP pathway, CGRP, total antioxidant capacity (TAC), malondialdehyde (MDA) and phosphodiesterase-3 (PDE-3). Isolated hearts of Wistar rats perfused according to the Langendorff technique were used to study the effects of an agonist of TRPV1, capsaicin (CS), an antagonist, capsazepine (CZ), and their combination CZ+CS. The hearts were subjected to three conditions: (1) control, (2) ischemia and (3) ischemia-reperfusion. We determined cardiac mechanical activity and the levels of NO, cGMP, BH4, CGRP, TAC, MDA and PDE-3 in ventricular tissue after administration of CS, CZ and CZ+CS. Western blots were used to study the expressions of eNOS, iNOS and phosphorylated NOS (pNOS). Structural changes were determined by histological evaluation. CS prevented damage caused by ischemia-reperfusion by improving cardiac mechanical activity and elevating the levels of NO, cGMP, BH4, TAC and CGRP. TRPV1 and iNOS expression were increased under ischemic conditions, while eNOS and pNOS were not modified. We conclude that the activation of TRPV1 constitutes a therapeutic possibility to counteract the damage caused by ischemia and reperfusion by regulating the NO pathway through CGRP.


Asunto(s)
Corazón/fisiopatología , Daño por Reperfusión Miocárdica/fisiopatología , Óxido Nítrico/metabolismo , Estrés Oxidativo , Canales Catiónicos TRPV/metabolismo , Animales , Masculino , Daño por Reperfusión Miocárdica/metabolismo , Ratas , Ratas Wistar , Transducción de Señal
12.
Can J Physiol Pharmacol ; 100(3): 252-258, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-34990309

RESUMEN

This study was designed to assess the effect of soya phosphatidylcholine (SPC) against ischemia/reperfusion (I/R) injury and the possible underlying mechanism using experimental and computational studies. I/R injury was induced by global ischemia for 30 min followed by reperfusion for 120 min. The perfusion of the SPC was performed for 10 min before inducing global ischemia. In the mechanistic study, the involvement of specific cellular pathways was identified using various inhibitors such as ATP-dependent potassium channel (KATP) inhibitor (glibenclamide), protein kinase C (PKC) inhibitor (chelerythrine), non-selective nitric oxide synthase inhibitor (L-NAME), and endothelium remover (Triton X-100). The computational study of various ligands was performed on toll-like receptor 4 (TLR4) protein using AutoDock version 4.0. SPC (100 µM) significantly decreased the levels of cardiac damage markers and %infarction compared with the vehicle control (VC). Furthermore, cardiodynamics (indices of left ventricular contraction (dp/dtmax), indices of left ventricular relaxation (dp/dtmin), coronary flow, and antioxidant enzyme levels were significantly improved as compared with VC. This protective effect was attenuated by glibenclamide, chelerythrine, and Triton X-100, but it was not attenuated by L-NAME. The computational study showed a significant bonding affinity of SPC to the TLR4-MD2 complex. Thus, SPC reduced myocardial I/R injury in isolated perfused rat hearts, which might be governed by the KATP channel, PKC, endothelium response, and TLR4-MyD88 signaling pathway.


Asunto(s)
Daño por Reperfusión Miocárdica/tratamiento farmacológico , Daño por Reperfusión Miocárdica/etiología , Fosfatidilcolinas/uso terapéutico , Animales , Cardiotónicos , Simulación por Computador , Técnicas In Vitro , Masculino , Daño por Reperfusión Miocárdica/diagnóstico , Daño por Reperfusión Miocárdica/fisiopatología , Fosfatidilcolinas/administración & dosificación , Fosfatidilcolinas/farmacología , Ratas Wistar , Receptor Toll-Like 4
13.
Int J Med Sci ; 19(1): 65-73, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34975299

RESUMEN

Reperfusion injury following myocardial ischemia remained a challenge for optimal treatment of myocardial infarction. Ginsenosides Rb (G-Rb), the primary components of ginsenoside, have been reported to exert cardioprotective effects via numerous mechanisms. G-Rb1 mediate cardioprotective effects via various signaling pathways, including mitochondrial apoptotic pathway, PI3K/Akt/mTOR, HIF-1α and GRF91, RhoA, p38α MAPK, and eNOS. G-Rb2 activates the SIRT-1 pathway, while G-Rb3 promotes both JNK-mediated NF-κB and PERK/Nrf2/HMOX1. Generally, ginsenosides Rb1, 2, and 3 modulates oxidative stress, inflammation, and apoptosis, contributing to the improvement of structural, functional and biochemical parameters. In conclusion, G-Rb, particularly G-Rb1, have vast potential as a supplement in attenuating reperfusion injury. Translation into a clinical trial is warranted to confirm the beneficial effects of G-Rb.


Asunto(s)
Ginsenósidos/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Animales , Apoptosis , Cardiotónicos/efectos adversos , Cardiotónicos/uso terapéutico , Ginsenósidos/efectos adversos , Ginsenósidos/uso terapéutico , Inflamación/fisiopatología , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/fisiopatología , Estrés Oxidativo , Transducción de Señal
14.
Cardiovasc Drugs Ther ; 36(1): 15-29, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-33064235

RESUMEN

PURPOSE: Cardiac dysfunction can occur as a sequela of a state of prolonged pressure overload and postischemic injury. Flavonoids such as quercetin may be protective against cardiovascular disease. This study aimed to systematically assess the effects of quercetin on cardiac function in pressure overload and postischemia-reperfusion injury in rodents. METHODS: A systematic search of the literature up to May 2020 was conducted in PubMed, Ovid Medline, EBSCOhost, Scopus, and the Cochrane Library to identify relevant published studies on quercetin and cardiac function using standardized criteria. Meta-analyses were performed on animal studies of pressure overload and ischemia-reperfusion (I/R) injury. RESULTS: The effects of quercetin on cardiac function in both models were qualitatively reported in 14 studies. The effects of quercetin in four pressure-overload model studies involving 73 rodents and eight I/R-injury model studies involving 120 rodents were quantitatively assessed by meta-analysis. Quercetin improved the overall cardiac function in both pressure overload (n = 4 studies, n = 73 rodents; SMD = - 1.50; 95% CI: - 2.66 to - 0.33; P < 0.05; I2 = 74.05%) and I/R injury (n = 8 studies, n = 120 rodents; SMD = - 1.81; 95% CI: - 3.05 to - 0.56; P < 0.01; I2 = 84.93%) models. The improvement was associated with amelioration in cardiac structure in the pressure-overload model and both systolic and diastolic functioning in the I/R-injury model. CONCLUSION: The present meta-analysis suggested that quercetin has beneficial effects for improving cardiac left ventricular dysfunction in both pressure-overload and I/R-injury models.


Asunto(s)
Daño por Reperfusión Miocárdica/tratamiento farmacológico , Quercetina/farmacología , Disfunción Ventricular Izquierda/tratamiento farmacológico , Animales , Cardiotónicos/farmacología , Modelos Animales de Enfermedad , Ratones , Daño por Reperfusión Miocárdica/fisiopatología , Ratas , Disfunción Ventricular Izquierda/fisiopatología , Presión Ventricular/efectos de los fármacos
15.
Cardiovasc Drugs Ther ; 36(1): 1-13, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-32648168

RESUMEN

PURPOSE: Mitochondrial reactive oxygen species (ROS) production upon reperfusion of ischemic tissue initiates the ischemia/reperfusion (I/R) injury associated with heart attack. During ischemia, succinate accumulates and its oxidation upon reperfusion by succinate dehydrogenase (SDH) drives ROS production. Inhibition of succinate accumulation and/or oxidation by dimethyl malonate (DMM), a cell permeable prodrug of the SDH inhibitor malonate, can decrease I/R injury. However, DMM is hydrolysed slowly, requiring administration to the heart prior to ischemia, precluding its administration to patients at the point of reperfusion, for example at the same time as unblocking a coronary artery following a heart attack. To accelerate malonate delivery, here we developed more rapidly hydrolysable malonate esters. METHODS: We synthesised a series of malonate esters and assessed their uptake and hydrolysis by isolated mitochondria, C2C12 cells and in mice in vivo. In addition, we assessed protection against cardiac I/R injury by the esters using an in vivo mouse model of acute myocardial infarction. RESULTS: We found that the diacetoxymethyl malonate diester (MAM) most rapidly delivered large amounts of malonate to cells in vivo. Furthermore, MAM could inhibit mitochondrial ROS production from succinate oxidation and was protective against I/R injury in vivo when added at reperfusion. CONCLUSIONS: The rapidly hydrolysed malonate prodrug MAM can protect against cardiac I/R injury in a clinically relevant mouse model.


Asunto(s)
Cardiotónicos/farmacología , Malonatos/farmacología , Infarto del Miocardio/prevención & control , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Animales , Cardiotónicos/síntesis química , Cardiotónicos/química , Línea Celular , Modelos Animales de Enfermedad , Ésteres/química , Femenino , Humanos , Masculino , Malonatos/síntesis química , Malonatos/química , Ratones , Ratones Endogámicos C57BL , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Daño por Reperfusión Miocárdica/fisiopatología , Profármacos , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo , Ácido Succínico/metabolismo
16.
Microvasc Res ; 139: 104266, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34688627

RESUMEN

The no-reflow phenomenon induced by ischemia-reperfusion (I/R) injury seriously limits the therapeutic value of coronary recanalization and leads to a poor prognosis. Previous studies have shown that luteolin (LUT) is a vasoprotective factor. However, whether LUT can be used to prevent the no-reflow phenomenon remains unknown. Positron emission tomography perfusion imaging, performed to detect the effects of LUT on the no-reflow phenomenon in vivo, revealed that LUT treatment was able to reduce the no-reflow area in rat I/R models. In vitro, LUT was shown to reduce the hypoxia-reoxygenation injury-induced endothelial permeability and apoptosis. The levels of malondialdehyde, reactive oxygen species and NADPH were also measured and the results indicated that LUT could inhibit the oxidative stress. Western blot analysis revealed that LUT protected endothelial cells from I/R injury by regulating the Wnt/ß-catenin pathway. Overall, we concluded that the use of LUT to minimize I/R induced microvascular damage is a feasible strategy to prevent the no-reflow phenomenon.


Asunto(s)
Circulación Coronaria/efectos de los fármacos , Vasos Coronarios/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Luteolina/farmacología , Daño por Reperfusión Miocárdica/prevención & control , Fenómeno de no Reflujo/prevención & control , Vía de Señalización Wnt/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Permeabilidad Capilar/efectos de los fármacos , Células Cultivadas , Vasos Coronarios/diagnóstico por imagen , Vasos Coronarios/metabolismo , Vasos Coronarios/fisiopatología , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Células Endoteliales/patología , Humanos , Imagen de Perfusión Miocárdica , Daño por Reperfusión Miocárdica/diagnóstico por imagen , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/fisiopatología , Fenómeno de no Reflujo/diagnóstico por imagen , Fenómeno de no Reflujo/metabolismo , Fenómeno de no Reflujo/fisiopatología , Estrés Oxidativo/efectos de los fármacos , Tomografía de Emisión de Positrones , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo
17.
Biomed Pharmacother ; 145: 112432, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34798472

RESUMEN

Calenduloside E (CE) is a saponin isolated from Aralia elata (Miq) Seem, which has anti-cardiovascular disease effects. This study aims to evaluate the anti-myocardial ischemia-reperfusion injury (MIRI) mechanisms of CE and regulation of BAG3 on calcium overload. We adopted siRNA to interfere with BAG3 expression in H9c2 cardiomyocytes and used adenovirus to interfere with BAG3 expression (Ad-BAG3) in primary neonatal rat cardiomyocytes (PNRCMs) to clarify the role of BAG3 in mitigating MIRI by CE. The results showed that CE reduced calcium overload, and Ad-BAG3 had a significant regulatory effect on L-type Ca2+ channels (LTCC) but no effects on other calcium-related proteins. And BAG3 and LTCC were colocalized in myocardial tissue and BAG3 inhibited LTCC expression. Surprisingly, CE had no regulatory effect on LTCC mRNA, but CE promoted LTCC degradation through the autophagy-lysosomal pathway rather than the ubiquitination-protease pathway. Autophagy inhibitor played a negative regulation of cardiomyocyte contraction rhythm and field potential signals. Ad-BAG3 inhibited autophagy by regulating the expression of autophagy-related proteins and autophagy agonist treatment suppressed calcium overload. Therefore, CE promoted autophagy through BAG3, thereby regulating LTCC expression, inhibiting calcium overload, and ultimately reducing MIRI.


Asunto(s)
Calcio/metabolismo , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Miocitos Cardíacos/efectos de los fármacos , Ácido Oleanólico/análogos & derivados , Saponinas/farmacología , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Animales Recién Nacidos , Proteínas Reguladoras de la Apoptosis/genética , Autofagia/efectos de los fármacos , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo L/efectos de los fármacos , Canales de Calcio Tipo L/metabolismo , Línea Celular , Masculino , Daño por Reperfusión Miocárdica/fisiopatología , Ácido Oleanólico/farmacología , Ratas , Ratas Sprague-Dawley
18.
J Pharm Pharmacol ; 74(2): 282-291, 2022 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-34850055

RESUMEN

OBJECTIVES: Myocardial I/R injury is one of the most serious complications after reperfusion therapy in patients with myocardial infarction. Remifentanil has been found to protect the heart against I/R injury. However, its underlying mechanism remains uncertain in myocardial I/R injury. METHODS: The myocardial I/R injury rat model was established by 30 min of ischaemia followed by 24 h of reperfusion. The animal model was evaluated by the levels of TC, ALT and AST and H&E staining. The binding of miR-206-3p and TLR4 was predicted and verified using TargetScan software, luciferase reporter and RNA pull-down assays. The functional role and mechanism of remifentanil were identified by ultrasonic echocardiography, oxidative stress markers, H&E, Masson and TUNEL staining and western blot. KEY FINDINGS: The rat myocardial I/R injury model displayed a significantly high level of TC, ALT, AST, TLR4, p-IκBα and p-p65 and the presence of disorganized cells and inflammatory cell infiltration. The model also showed increased levels of LVEDD, LVESD, MDA, fibrosis and apoptosis and decreased levels of EF, FS, SOD and GSH, which were reversed with remifentanil treatment. Knockdown of miR-206-3p damaged cardiac function and aggravated oxidative stress. miR-206-3p could directly bind to TLR4. TLR4 overexpression destroyed cardiac function, exacerbated oxidative stress, increased levels of p-IκBα and p-p65 and aggravated pathology manifestation affected by remifentanil. CONCLUSIONS: Our results elucidated that remifentanil alleviated myocardial I/R injury by miR-206-3p/TLR4/NF-κB signalling axis.


Asunto(s)
Cardiotónicos/farmacología , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Estrés Oxidativo/efectos de los fármacos , Remifentanilo/farmacología , Animales , Línea Celular , Modelos Animales de Enfermedad , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Masculino , MicroARNs/genética , Daño por Reperfusión Miocárdica/fisiopatología , FN-kappa B/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor Toll-Like 4/metabolismo
19.
Pharmacol Res ; 175: 105986, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34800627

RESUMEN

During cardiac reperfusion after myocardial infarction, the heart is subjected to cascading cycles of ischaemia reperfusion injury (IRI). Patients presenting with this injury succumb to myocardial dysfunction resulting in myocardial cell death, which contributes to morbidity and mortality. New targeted therapies are required if the myocardium is to be protected from this injury and improve patient outcomes. Extensive research into the role of mitochondria during ischaemia and reperfusion has unveiled one of the most important sites contributing towards this injury; specifically, the opening of the mitochondrial permeability transition pore. The opening of this pore occurs during reperfusion and results in mitochondria swelling and dysfunction, promoting apoptotic cell death. Activation of mitochondrial ATP-sensitive potassium channels (mitoKATP) channels, uncoupling proteins, and inhibition of glycogen synthase kinase-3ß (GSK3ß) phosphorylation have been identified to delay mitochondrial permeability transition pore opening and reduce reactive oxygen species formation, thereby decreasing infarct size. Statins have recently been identified to provide a direct cardioprotective effect on these specific mitochondrial components, all of which reduce the severity of myocardial IRI, promoting the ability of statins to be a considerate preconditioning agent. This review will outline what has currently been shown in regard to statins cardioprotective effects on mitochondria during myocardial IRI.


Asunto(s)
Cardiotónicos/uso terapéutico , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Animales , Cardiotónicos/farmacología , Glucógeno Sintasa Quinasa 3 beta/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/efectos adversos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Mitocondrias Cardíacas/efectos de los fármacos , Mitocondrias Cardíacas/fisiología , Poro de Transición de la Permeabilidad Mitocondrial/metabolismo , Mitofagia/efectos de los fármacos , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/fisiopatología , Canales de Potasio/fisiología
20.
Pharm Biol ; 60(1): 38-45, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34860639

RESUMEN

CONTEXT: Ferroptosis was described as an important contributor to the myocardial ischaemia/reperfusion (MIR) injury, and britanin (Bri) was reported to exert antitumor and anti-inflammatory activities. OBJECTIVE: Our study explores the effect and mechanism of Bri on MIR damage. MATERIALS AND METHODS: The rat model of MIR was established by ligation of the left anterior descending coronary artery. Male Sprague-Dawley (SD) rats were divided into three groups: sham group (n = 6), MIR group (n = 6) and MIR + Bri group (n = 6; 50 mg/kg). Rats were intragastrically pre-treated with Bri or normal saline once daily for 3 days. To further verify the role and mechanism of Bri, H9C2 cells were subjected to hypoxia plus reoxygenation (H/R) to induce the in vitro model of MIR. RESULTS: Compared with MIR rats, Bri significantly decreased infarct area (22.50% vs. 38.67%), myocardial apoptosis (23.00% vs. 41.5%), creatine phosphokinase (0.57 U/mL vs. 0.76 U/mL), and lactate dehydrogenase levels (3.18 U/mL vs. 5.17 U/mL), concomitant with alleviation of ferroptosis. Mechanistically, Bri treatment induced the activation of the adenosine monophosphate activated protein kinase (AMPK)/glycogen synthase kinase 3ß (GSK3ß)/nuclear factor erythroid 2-related factor 2 (Nrf2) pathway in vivo. In addition, the AMPK/GSK3ß/Nrf2 pathway participated in the regulation of glutathione peroxidase 4 (GPX4) expression, and silencing of Nrf2 attenuated the effect of Bri on H/R-induced cell injury. DISCUSSION AND CONCLUSIONS: Bri protected against ferroptosis-mediated MIR damage by upregulating GPX4 through activation of the AMPK/GSK3ß/Nrf2 signalling, suggesting that Bri might become a novel therapeutic agent for MIR.


Asunto(s)
Ferroptosis/efectos de los fármacos , Lactonas/farmacología , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Fosfolípido Hidroperóxido Glutatión Peroxidasa/genética , Sesquiterpenos/farmacología , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Apoptosis/efectos de los fármacos , Línea Celular , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Masculino , Daño por Reperfusión Miocárdica/fisiopatología , Factor 2 Relacionado con NF-E2/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA