Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Eur J Pharmacol ; 956: 175938, 2023 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-37536623

RESUMEN

Impaired endothelium-dependent vasodilation in atherosclerosis is a high-risk factor for myocardial infarction and ischemic stroke, and inflammation, necroptosis and apoptosis contribute to endothelial dysfunction in atherosclerosis. Although DL-3-n-butylphthalide (NBP) has been widely used in treating ischemic stroke, its effect on endothelium-dependent vasodilation remains unknown. This study aims to explore whether NBP is able to improve endothelium-dependent vasodilation in atherosclerosis and the underlying mechanisms. Male ApoE-/- mice were fed with a high-fat diet (HFD) for 9-16 weeks to establish a model of atherosclerosis. NBP were given to the mice after eating HFD for 6 weeks and atorvastatin served as a positive control. The endothelium-dependent vasodilation, the blood flow velocity, the atherosclerotic lesion area, the serum levels of lipids, inflammatory cytokines and necroptosis-relevant proteins (RIPK1, RIPK3 and MLKL), and the endothelial necroptosis and apoptosis within the aorta were measured. Human umbilical vein endothelial cells (HUVECs) were incubated with oxidized low-density lipoprotein (ox-LDL) for 48 h to mimic endothelial injury in atherosclerosis, lactate dehydrogenase release, the ratio of necroptosis and apoptosis and the expression of necroptosis-relevant proteins were examined. Similar to atorvastatin, NBP improves endothelium-dependent vasodilation, decreases aortic flow velocity and reduces atherosclerotic lesion area in HFD-fed ApoE-/- mice, concomitant with a reduction in serum lipids, inflammatory cytokines and necroptosis-relevant proteins, and endothelial necroptosis and apoptosis. Consistently, NBP inhibited necroptosis and apoptosis in ox-LDL-treated HUVECs. Based on these observations, we conclude that NBP exerts beneficial effects on improving the endothelium-dependent vasodilation in atherosclerosis via suppressing inflammation, endothelial necroptosis and apoptosis.


Asunto(s)
Aterosclerosis , Accidente Cerebrovascular Isquémico , Masculino , Humanos , Ratones , Animales , Dieta Alta en Grasa/efectos adversos , Vasodilatación , Atorvastatina/farmacología , Necroptosis , Aterosclerosis/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Inflamación/metabolismo , Endotelio/metabolismo , Citocinas/metabolismo , Accidente Cerebrovascular Isquémico/metabolismo , Apoptosis , Apolipoproteínas E/genética , Ratones Noqueados
2.
Eur J Pharmacol ; 950: 175774, 2023 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-37146710

RESUMEN

The dysregulation of nuclear factor erythroid 2-related factor 2 (Nrf2) and/or solute carrier family 7 member 11 (SLC7A11) is believed to contribute to ferroptosis in the hearts suffered ischemia/reperfusion (I/R), but the mechanisms behind the dysregulation of them are not fully elucidated. Mucosa associated lymphoid tissue lymphoma translocation gene 1 (MALT1) can function as a paracaspase to cleave specified substrates and it is predicted to interact with Nrf2. This study aims to explore whether targeting MALT1 can reduce I/R-induced ferroptosis via enhancing the Nrf2/SLC7A11 pathway. The SD rat hearts were subjected to 1h-ischemia plus 3h-reperfusion to establish the I/R injury model, which showed myocardial injuries (increase in infarct size and creatine kinase release) and up-regulation of MALT1 while downregulation of Nrf2 and SLC7A11 concomitant with the increased ferroptosis, reflecting by an increase in glutathione peroxidase 4 (GPX4) level while decreases in the levels of acyl-CoA synthetase long chain family member 4 (ACSL4), total iron, Fe2+ and lipid peroxidation (LPO); these phenomena were reversed in the presence of MI-2, a specific inhibitor of MALT1. Consistently, similar results were achieved in the cultured cardiomyocytes subjected to 8h-hypoxia plus 12h-reoxygenation. Furthermore, micafungin, an antifungal drug, could also exert beneficial effect on mitigating myocardial I/R injury via inhibition of MALT1. Based on these observations, we conclud that inhibition of MALT1 can reduce I/R-induced myocardial ferroptosis through enhancing the Nrf2/SLC7A11 pathway; and MALT1 may be used as a potential target to seek novel or existing drugs (such as micafungin) for treating myocardial infarction.


Asunto(s)
Ferroptosis , Daño por Reperfusión Miocárdica , Daño por Reperfusión , Animales , Ratas , Isquemia , Micafungina , Proteína 1 de la Translocación del Linfoma del Tejido Linfático Asociado a Mucosas , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Factor 2 Relacionado con NF-E2 , Ratas Sprague-Dawley , Reperfusión
3.
Chem Biol Interact ; 368: 110205, 2022 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-36195186

RESUMEN

Ferroptosis is an iron-dependent cell death and contributes to doxorubicin-induced cardiotoxicity, but the mechanisms behind intracellular iron overload in cardiomyocyte after administration of doxorubicin remain largely unknown. Ferritinophagy is a selective type of autophagy and could be a novel source for intracellular free iron. Spermatogenesis-associated protein 2 (SPATA2), a member of the TNF signaling pathway, can recruit cylindromatosis (CYLD, a deubiquitinating enzyme) to regulate cell death. This study aims to explore whether ferritinophagy is the source for intracellular iron overload in cardiomyocyte upon doxorubicin treatment and whether the SPATA2/CYLD pathway is involved in regulation of nuclear receptor coactivator 4 (NCOA4) level, the selective cargo receptor for ferritinophagy. The C57BL/6J mice were subjected to a single injection of doxorubicin, which showed the compromised cardiac functions, accompanied by the upregulation of SPATA2 and CYLD and the enhanced interaction between them, the increases in ferritinophagy (reflecting by increases in NCOA4 and ratio of LC3Ⅱ/LC3Ⅰ while decreases in NCOA4 ubiquitination and ferritin) and ferroptosis (reflecting by intracellular iron overload and increase of acyl-CoA synthetase long chain family member 4). Consistently, similar results were achieved in the cultured cardiomyocytes after incubation with doxorubicin. Knocked down of SPATA2 notably reduced doxorubicin-induced cardiomyocyte injury concomitant with the attenuated ferritinophagy and the decreased ferroptosis. Based on these observations, we conclude that a novel pathway of SPATA2/CYLD has been identified, which contributes to doxorubicin-induced cardiomyocyte ferroptosis via enhancing ferritinophagy through a mechanism involving the deubiquitination of NCOA4.


Asunto(s)
Ferroptosis , Sobrecarga de Hierro , Ratones , Masculino , Animales , Miocitos Cardíacos/metabolismo , Ratones Endogámicos C57BL , Autofagia , Hierro/metabolismo , Factores de Transcripción , Doxorrubicina/toxicidad , Enzima Desubiquitinante CYLD
4.
Eur J Pharmacol ; 898: 173999, 2021 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-33675785

RESUMEN

Necroptosis, ferroptosis and cyclophilin D (Cyp D)-dependent necrosis contribute to myocardial ischemia/reperfusion (I/R) injury, and ponatinib, deferoxamine and cyclosporine are reported to inhibit necroptosis, ferroptosis and Cyp D-dependent necrosis, respectively. This study aims to explore whether the any two combination between ponatinib, deferoxamine and cyclosporine exerts a better cardioprotective effect on I/R injury than single medicine does. The H9c2 cells were subjected to 10 h of hypoxia (H) plus 4 h of reoxygenation (R) to establish H/R injury model. The effects of any two combination between ponatinib, deferoxamine and cyclosporine on H/R injury were examined. On this basis, a I/R injury model in rat hearts was established to focus on the effect of ponatinib, deferoxamine and their combination on myocardial I/R injury and the underlying mechanisms. In H/R-treated H9c2 cells, all three medicines can attenuate H/R injury (decrease in LDH release and necrosis percent). However, only the combination of ponatinib with deferoxamine exerted synergistic effect on reducing H/R injury, showing simultaneous suppression of necroptosis and ferroptosis. Expectedly, administration of ponatinib or deferoxamine either before or after ischemia could suppress necroptosis or ferroptosis in the I/R-treated rat hearts as they did in vitro, concomitant with a decrease in myocardial infarct size and creatine kinase release, and the combination therapy is more efficient than single medication. Based on these observations, we conclude that the combination of ponatinib with deferoxamine reduces myocardial I/R injury via simultaneous inhibition of necroptosis and ferroptosis.


Asunto(s)
Deferoxamina/farmacología , Ferroptosis/efectos de los fármacos , Imidazoles/farmacología , Infarto del Miocardio/prevención & control , Daño por Reperfusión Miocárdica/prevención & control , Miocitos Cardíacos/efectos de los fármacos , Necroptosis/efectos de los fármacos , Piridazinas/farmacología , Animales , Línea Celular , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Quimioterapia Combinada , Peroxidación de Lípido/efectos de los fármacos , 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 , Ratas Sprague-Dawley , Transducción de Señal
5.
Free Radic Biol Med ; 162: 339-352, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33157209

RESUMEN

Iron overload triggers the ferroptosis in the heart following ischemia/reperfusion (I/R) and transferrin receptor 1 (TfR1) charges the cellular iron uptake. Bioinformatics analysis shows that the three molecules of ubiquitin-specific protease 7 (USP7), p53 and TfR1 form a unique pathway of USP7/p53/TfR1. This study aims to explore whether USP7/p53/TfR1 pathway promotes ferroptosis in rat hearts suffered I/R and the underlying mechanisms. The SD rat hearts were subjected to 1 h-ischemia plus 3 h-reperfusion, showing myocardial injury (increase in creatine kinase release, infarct size, myocardial fiber loss and disarray) and up-regulation of USP7, p53 and TfR1 concomitant with an increase of ferroptosis (reflecting by accumulation of iron and lipid peroxidation while decrease of glutathione peroxidase activity). Inhibition of USP7 activated p53 via suppressing deubiquitination, which led to down-regulation of TfR1, accompanied by the decreased ferroptosis and myocardial I/R injury. Next, H9c2 cells underwent hypoxia/reoxygenation (H/R) in vitro to mimic the myocardial I/R model in vivo. Consistent with the results in vivo, inhibition or knockdown of USP7 reduced the H/R injury (decrease of LDH release and necrosis) and enhanced the ubiquitination of p53 along with the decreased levels of p53 and TfR1 as well as the attenuated ferroptosis (manifesting as the decreased iron content and lipid peroxidation while the increased GPX activity). Knockdown of TfR1 inhibited H/R-induced ferroptosis without p53 deubiquitination. Based on these observations, we conclude that a novel pathway of USP7/p53/TfR1 has been identified in the I/R-treated rat hearts, where up-regulation of USP7promotes ferrptosis via activation of the p53/TfR1 pathway.


Asunto(s)
Ferroptosis , Corazón , Peptidasa Específica de Ubiquitina 7/genética , Animales , Isquemia , Ratas , Ratas Sprague-Dawley , Receptores de Transferrina , Reperfusión , Proteína p53 Supresora de Tumor/genética
6.
Naunyn Schmiedebergs Arch Pharmacol ; 394(2): 401-410, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32621060

RESUMEN

Ferroptosis is an iron-dependent regulated necrosis. This study aims to evaluate the contribution of ferroptosis to ischemia or reperfusion injury, and lay a basis for precise therapy of myocardial infarction. The Sprague-Dawley (SD) rat hearts were subjected to ischemia for different duration or the hearts were treated with 1 h-ischemia plus different duration of reperfusion. The myocardial injury was assessed by biochemical assays and hematoxylin & eosin (HE) staining. The ferroptosis was evaluated with the levels of acyl-CoA synthetase long-chain family member 4 (ACSL4), glutathione peroxidase 4 (GPX4), iron, and malondialdehyde. Iron chelator (deferoxamine) was applied to verify the contribution of ferroptosis to ischemia and reperfusion injury. The results showed that ischemic injury (infarction and CK release) was getting worse with the extension of ischemia, but no significant changes in ferroptosis indexes (ACSL4, GPX4, iron, and malondialdehyde) in cardiac tissues were observed. Differently, the levels of ACSL4, iron, and malondialdehyde were gradually elevated with the extension of reperfusion concomitant with a decrease of GPX4 level. In the ischemia-treated rat hearts, no significant changes in myocardial injury were observed in the presence of deferoxamine, while in the ischemia/reperfusion-treated rat hearts, myocardial injury was markedly attenuated in the presence of deferoxamine concomitant with a reduction of ferroptosis. Based on these observations, we conclude that ferroptosis occurs mainly in the phase of myocardial reperfusion but not ischemia. Thus, intervention of ferroptosis exerts beneficial effects on reperfusion injury but not ischemic injury, laying a basis for precise therapy for patients with myocardial infarction.


Asunto(s)
Ferroptosis , Isquemia , Daño por Reperfusión Miocárdica , Animales , Biomarcadores/sangre , Biomarcadores/metabolismo , Coenzima A Ligasas/metabolismo , Creatina Quinasa/sangre , Deferoxamina/farmacología , Hierro/metabolismo , Isquemia/metabolismo , Masculino , Malondialdehído/metabolismo , Daño por Reperfusión Miocárdica/sangre , Daño por Reperfusión Miocárdica/metabolismo , Miocardio/metabolismo , Fosfolípido Hidroperóxido Glutatión Peroxidasa/metabolismo , Ratas Sprague-Dawley , Sideróforos/farmacología
7.
Eur J Pharmacol ; 875: 173053, 2020 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-32135123

RESUMEN

RIPK1/RIPK3/MLKL (Receptor-interacting protein kinase 1/Receptor-interacting protein kinase 3/Mixed lineage kinase domain-like protein) pathway-mediated necroptosis contributes to myocardial ischemia/reperfusion (I/R) injury, and Arctiin can prevent myocardial fibrosis and hypertrophy. This study aims to explore the effect of Arctiin on myocardial I/R injury and the underlying mechanisms. SD rat hearts or cardiomyocytes were subjected to I/R or hypoxia/reoxygenation (H/R) to establish the I/R or H/R injury model. The methods of biochemistry, PI/DAPI (propidium iodide/4',6-Diamidino-2-Phenylindole) and H&E (Hematoxylin & eosin) staining were used to evaluate the I/R or H/R injury. The effects of Arctiin on necroptosis in I/R-treated hearts or H/R-treated cardiomyocytes were assessed. The results showed that Arctiin reduced myocardial I/R injury (decreases in myocardial infarction and creatine kinase release), concomitant with a decrease in levels of necroptosis-associated proteins (RIPK1/p-RIPK1, RIPK3/p-RIPK3 and MLKL/p-MLKL) in I/R-treated rat hearts. Consistently, the necrosis and LDH release in H/R-treated cardiomyocytes were attenuated in the presence of Arctiin, accompanied by suppression of necroptosis-relevant proteins. Furthermore, H/R-induced reactive oxygen species (ROS) generation and mitochondrial dysfunctions (increase in mitochondrial membrane potential and decrease in ATP production) were impaired by Arctiin. Using the program of the Molecular Operating Environment (MOE), we predict that RIPK1 and MLKL (but not RIPK3) might be the potential targets of Arctiin. Based on these observations, we conclude that Arctiin can protect the rat heart from I/R injury, and its beneficial effect is related to reduction of necroptosis via scavenging reactive oxygen species and restoring mitochondrial functions or targeting RIPK1 and/or MLKL.


Asunto(s)
Cardiotónicos/farmacología , Medicamentos Herbarios Chinos/farmacología , Furanos/farmacología , Glucósidos/farmacología , Daño por Reperfusión Miocárdica/prevención & control , Necroptosis/efectos de los fármacos , Animales , Cardiotónicos/uso terapéutico , Línea Celular , Modelos Animales de Enfermedad , Medicamentos Herbarios Chinos/uso terapéutico , Furanos/uso terapéutico , Glucósidos/uso terapéutico , Humanos , Masculino , Mitocondrias/efectos de los fármacos , Mitocondrias/patología , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Ratas , Especies Reactivas de Oxígeno/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores , Transducción de Señal/efectos de los fármacos
8.
Can J Physiol Pharmacol ; 98(5): 259-266, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-31825666

RESUMEN

Mitochondrial dysfunctions are responsible for myocardial injury upon ischemia/reperfusion (I/R), and mitochondrial E3 ubiquitin ligase 1 (Mul1) plays an important role in maintaining mitochondrial functions. This study aims to explore the function of Mul1 in myocardial I/R injury and the underlying mechanisms. The Sprague-Dawley rat hearts were subjected to 1 h of ischemia plus 3 h of reperfusion, which showed the I/R injury (increase in infarct size and creatine kinase release) and the elevated total and mitochondrial protein levels of Mul1 and p53 accompanied by the enhanced interactions between Mul1 and p53 as well as p53 and small a ubiquitin-like modifier (SUMO1). Consistently, hypoxia/reoxygenation (H/R) treated cardiac (H9c2) cells displayed cellular injury (apoptosis and necrosis), upregulation of total and mitochondrial protein levels of Mul1 and p53, and enhanced interactions between p53 and SUMO1 concomitant with mitochondrial dysfunctions (an increase in mitochondrial membrane potential and reactive oxygen species production with a decrease in ATP production); these phenomena were attenuated by knockdown of Mul1 expression. Based on these observations, we conclude that a novel role of Mul1 has been identified in the myocardial mitochondria, where Mul1 stabilizes and activates p53 through its function of SUMOylation following I/R, leading to p53-mediated mitochondrial dysfunction and cell death.


Asunto(s)
Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Regulación hacia Arriba , Adenosina Trifosfato/metabolismo , Animales , Línea Celular , Técnicas de Silenciamiento del Gen , Masculino , Potencial de la Membrana Mitocondrial , Proteínas Mitocondriales/deficiencia , Proteínas Mitocondriales/genética , Daño por Reperfusión Miocárdica/genética , Daño por Reperfusión Miocárdica/patología , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/deficiencia , Ubiquitina-Proteína Ligasas/genética
9.
Cardiovasc Drugs Ther ; 33(1): 13-23, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30637549

RESUMEN

PURPOSE: Necroptosis is an important form of cell death following myocardial ischemia/reperfusion (I/R) and phosphoglycerate mutase 5 (PGAM5) functions as the convergent point for multiple necrosis pathways. This study aims to investigate whether inhibition of PGAM5 could reduce I/R-induced myocardial necroptosis and the underlying mechanisms. METHODS: The SD rat hearts (or H9c2 cells) were subjected to 1-h ischemia (or 10-h hypoxia) plus 3-h reperfusion (or 4-h reoxygenation) to establish the I/R (or H/R) injury model. The myocardial injury was assessed by the methods of biochemistry, H&E (hematoxylin and eosin), and PI/DAPI (propidium iodide/4',6-diamidino-2-phenylindole) staining, respectively. Drug interventions or gene knockdown was used to verify the role of PGAM5 in I/R (or H/R)-induced myocardial necroptosis and possible mechanisms. RESULTS: The I/R-treated heart showed the injuries (increase in infarct size and creatine kinase release), upregulation of PGAM5, dynamin-related protein 1 (Drp1), p-Drp1-S616, and necroptosis-relevant proteins (RIPK1/RIPK3, receptor-interacting protein kinase 1/3; MLKL, mixed lineage kinase domain-like); these phenomena were attenuated by inhibition of PGAM5 or RIPK1. In H9c2 cells, H/R treatment elevated the levels of PGAM5, RIPK1, RIPK3, MLKL, Drp1, and p-Drp1-S616 and induced mitochondrial dysfunctions (elevation in mitochondrial membrane potential and ROS level) and cellular necrosis (increase in LDH release and the ratio of PI+/DAPI+ cells); these effects were blocked by inhibition or knockdown of PGAM5. CONCLUSIONS: Inhibition of PGAM5 can reduce necroptosis in I/R-treated rat hearts through suppression of Drp1; there is a positive feedback between RIPK1 and PGAM5, and PGAM5 might serve as a novel therapeutic target for prevention of myocardial I/R injury.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasa 1/metabolismo , Inhibidores Enzimáticos/farmacología , Glicolatos/farmacología , Proteínas Mitocondriales/antagonistas & inhibidores , Infarto del Miocardio/prevención & control , Daño por Reperfusión Miocárdica/prevención & control , Miocitos Cardíacos/efectos de los fármacos , Fosfoglicerato Mutasa/antagonistas & inhibidores , Fosfoproteínas Fosfatasas/antagonistas & inhibidores , Animales , Muerte Celular/efectos de los fármacos , Línea Celular , Modelos Animales de Enfermedad , Regulación hacia Abajo , Masculino , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Infarto del Miocardio/enzimología , Infarto del Miocardio/patología , Daño por Reperfusión Miocárdica/enzimología , Daño por Reperfusión Miocárdica/patología , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/patología , Fosfoglicerato Mutasa/genética , Fosfoglicerato Mutasa/metabolismo , Fosfoproteínas Fosfatasas/genética , Fosfoproteínas Fosfatasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Ratas Sprague-Dawley , Proteína Serina-Treonina Quinasas de Interacción con Receptores , Transducción de Señal/efectos de los fármacos
10.
Eur J Pharmacol ; 822: 1-12, 2018 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-29337194

RESUMEN

Fasudil is a potent Rho-kinase (ROCK) inhibitor and can relax smooth muscle or cardiac muscle contraction through decreasing the phosphorylation level of myosin regulatory light chain (p-MLC20 or p-MLC2v), while p-MLC2v can function as a transcription factor to promote the NADPH oxidase 2 (NOX2) expression in rat hearts subjected to ischemia/reperfusion (I/R). This study aims to explore whether fasudil can protect the rat hearts against I/R oxidative injury through suppressing NOX2 expression via reduction of p-MLC2v level. The SD rat hearts were subjected to 1h-ischemia plus 3h-reperfusion, which showed myocardial injuries (myocardial fiber loss and disarray, increase of creatine kinase release and myocardial infarction/apoptosis), increase in ROCK activity and nuclear p-MLC2v level concomitant with up-regulation of NOX2 and H2O2 production; these phenomena were attenuated by fasudil in a dose-dependent manner. Next, we verified the cardioprotective effect of fasudil and the underlying mechanisms in hypoxia-reoxygenation (H/R) -treated H9c2 cells. Consistent with the results in vivo, the H/R-treated H9c2 cells showed cellular injury (increase in apoptotic ratio), elevation in ROCK activity and nuclear p-MLC2v level, accompanied by up-regulation of NOX2 and H2O2 production; these effects were blocked in the presence of fasudil in a dose-dependent way. Based on these observations, we conclude that beneficial effect of fasudil against myocardial I/R or H/R oxidative injury is related to the suppression of NOX2 expression through decrease of the p-MLC2v level. Our findings also highlight that intervention of MLC2v phosphorylation by drugs may provide a novel strategy to protect heart from I/R oxidative injury.


Asunto(s)
1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/análogos & derivados , Corazón/efectos de los fármacos , Cadenas Ligeras de Miosina/metabolismo , NADPH Oxidasa 2/metabolismo , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/metabolismo , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/farmacología , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/uso terapéutico , Animales , Apoptosis/efectos de los fármacos , Miosinas Cardíacas/metabolismo , Línea Celular , Masculino , Miocardio/patología , Fosforilación/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Daño por Reperfusión/patología , Quinasas Asociadas a rho/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...