Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
J Cell Physiol ; 238(1): 242-256, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36538623

RESUMEN

Myocardial hypertrophy is associated with a significant increase in intracellular Ca2+ , which can be induced by long-chain fatty acid. Palmitic acid methyl ester (PAME), a fatty acid ester released from adipose tissue, superior cervical ganglion, and retina, has been found to have anti-inflammation, antifibrosis, and peripheral vasodilation effects. However, the effects of PAME on cardiomyocytes are still unclear. The aim of this study was to determine whether PAME could disrupt the intracellular Ca2+ balance, leading to cardiomyocyte hypertrophy. Neonatal rat cardiomyocytes were treated with various concentrations (10-100 µM) of PAME for 1-4 days. Cytosolic Ca2+ and mitochondrial Ca2+ concentrations were examined using Fura-2 AM and Rhod-2, respectively. After treatment with PAME for 4 days, mitochondrial Ca2+ , an indicator of the state of mitochondrial permeability transition pore (MPTP), and cell death were monitored by flow cytometric analysis. ATP levels were detected using the ATP assay kit. Cardiomyocyte hypertrophy was analyzed by measuring the cardiac hypertrophy biomarker and cell area using quantitative real time-polymerase chain reaction, Western Blot analysis and immunofluorescence analysis. Our results show that PAME concentration- and time-dependently increased cytosolic and mitochondria Ca2+ through the mitochondrial calcium uniporter. Moreover, treatment with PAME for 4 days caused MPTP opening, thereby reducing ATP production and enhancing reactive oxygen species (ROS) generation, and finally led to cardiomyocyte hypertrophy. These effects caused by PAME treatment were attenuated by the G-protein coupled receptor 40 (GPR40) inhibitor. In conclusion, PAME impaired mitochondrial function, which in turn led to cardiomyocyte hypertrophy through increasing the mitochondrial Ca2+ levels mediated by activating the GPR40 signaling pathway.


Asunto(s)
Calcio , Mitocondrias , Palmitatos , Receptores Acoplados a Proteínas G , Animales , Ratas , Adenosina Trifosfato/metabolismo , Calcio/metabolismo , Cardiomegalia/inducido químicamente , Cardiomegalia/metabolismo , Mitocondrias/metabolismo , Miocitos Cardíacos/metabolismo , Palmitatos/farmacología , Receptores Acoplados a Proteínas G/metabolismo , Células Cultivadas
2.
J Mol Cell Cardiol ; 118: 122-132, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29577873

RESUMEN

Intermittent hypoxia (IH) has been shown to exert cardioprotective effects against ischemia/reperfusion (I/R) injury through the preservation of ion homeostasis. I/R dramatically elevated cytosolic Zn2+ and caused cardiomyocyte death. However, the role of IH exposure in the relationship between Zn2+ regulation and cardioprotection is still unclear. The aim of the present study was to study whether IH exposure could help in intracellular Zn2+ regulation, hence contributing to cardioprotection against I/R injury. Adult rat cardiomyocytes were exposed to IH (5% O2, 5% CO2 and balanced N2) for 30 min followed by 30 min of normoxia (21% O2, 5% CO2 and balanced N2). Changes in intracellular Zn2+ concentration were determined using a Zn2+-specific fluorescent dye, FluoZin-3 or RhodZin-3. Fluorescence was monitored under an inverted fluorescent or confocal microscope. The results demonstrated that I/R or 2,2'-dithiodipyridine (DTDP), a reactive disulphide compound, induced Zn2+ release from metallothioneins (MTs), subsequently causing cytosolic Zn2+ overload, which in turn increased intracellular Zn2+ entry into the mitochondria via a Ca2+ uniporter, hence inducing mitochondrial membrane potential loss, and eventually led to cell death. However, the cytosolic Zn2+ overload and cell death caused by I/R or DTDP was significantly reduced by treatment of cardiomyocytes with IH. The findings from this study suggest that IH might exert its cardioprotective effect through reducing the I/R-induced cytosolic Zn2+ overload and cell death in cardiomyocytes.


Asunto(s)
Hipoxia/patología , Espacio Intracelular/metabolismo , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Especies Reactivas de Oxígeno/metabolismo , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Zinc/metabolismo , 2,2'-Dipiridil/análogos & derivados , 2,2'-Dipiridil/farmacología , Animales , Cardiotónicos/metabolismo , Muerte Celular/efectos de los fármacos , Disulfuros/farmacología , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Metalotioneína/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Modelos Biológicos , Miocitos Cardíacos/efectos de los fármacos , Ratas Sprague-Dawley
3.
Cell Physiol Biochem ; 46(3): 1252-1262, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29672298

RESUMEN

BACKGROUND/AIMS: Intermittent hypoxia (IH) has been shown to exert preconditioning-like cardioprotective effects. It also has been reported that IH preserves intracellular pH (pHi) during ischemia and protects cardiomyocytes against ischemic reperfusion injury. However, the exact mechanism is still unclear. METHODS: In this study, we used proton indicator BCECF-AM to analyze the rate of pHi recovery from acidosis in the IH model of rat neonatal cardiomyocytes. Neonatal cardiomyocytes were first treated with repetitive hypoxia-normoxia cycles for 1-4 days. Cells were then acid loaded with NH4Cl, and the rate of pHi recovery from acidosis was measured. RESULTS: We found that the pHi recovery rate from acidosis was much slower in the IH group than in the room air (RA) group. When we treated cardiomyocytes with Na+-H+ exchange (NHE) inhibitors (Amiloride and HOE642) or Na+-free Tyrode solution during the recovery, there was no difference between RA and IH groups. We also found intracellular Na+ concentration ([Na+]i) significantly increased after IH exposure for 4 days. However, the phenomenon could be abolished by pretreatment with ROS inhibitors (SOD and phenanathroline), intracellular calcium chelator or Na+-Ca2+ exchange (NCX) inhibitor. Furthermore, the pHi recovery rate from acidosis became faster in the IH group than in the RA group when inhibition of NCX activity. CONCLUSIONS: These results suggest that IH would induce the elevation of ROS production. ROS then activates Ca2+-efflux mode of NCX and results in intracellular Na+ accumulation. The rise of [Na+]i further inhibits the activity of NHE-mediated acid extrusion and retards the rate of pHi recovery from acidosis during IH.


Asunto(s)
Hipoxia de la Célula , Intercambiador de Sodio-Calcio/metabolismo , Sodio/metabolismo , Amilorida/farmacología , Animales , Células Cultivadas , Femenino , Guanidinas/farmacología , Concentración de Iones de Hidrógeno , Masculino , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Proteína Quinasa C/metabolismo , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Intercambiador de Sodio-Calcio/antagonistas & inhibidores , Sulfonas/farmacología , Superóxido Dismutasa/metabolismo
4.
Chin J Physiol ; 58(4): 254-62, 2015 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-26211649

RESUMEN

Intermittent hypoxia (IH) occurs frequently in patients with obstructive sleep apnoea and can cause ventricular dysfunction. However, whether myocardial inflammation and sodium-hydrogen exchanger-1 (NHE-1) expression play an important role in IH-induced ventricular dysfunction remains unclear. This study aimed to investigate whether short-term exercise provides a protective effect on IH-induced left ventricular (LV) function impairment. Male Sprague-Dawley rats were randomly assigned to 4 groups: control (CON), IH, exercise (EXE) or IH interspersed with EXE (IHEXE). IH rats were exposed to repetitive hypoxia/reoxygenation cycles (2%-6% O2 for 2-5 s per 75 s, followed by 21% O2 for 6 h/day) during the light phase for 12 consecutive days. EXE rats were habituated to treadmill running for 5 days, permitted 2 days of rest, and followed by 5 exercise bouts (30 m/min for 60 min on a 2% grade) on consecutive days during the dark phase. IHEXE rats were exposed to IH during the light phase interspersed with exercise programs during the dark phase on the same day. Cardiac function was quantified by echocardiographic evaluation. Myocardial levels of tumour necrosis factor-alpha (TNF-α), interleukin-6 (IL-6) and NHE-1 were determined. IH rats showed LV dysfunction characterized by lower LV fractional shortening (LVFS%) and LV ejection fraction (LVEF%). LV dysfunction was associated with higher myocardial levels of TNF-α, IL-6 and NHE-1 mRNA and protein. These changes were not observed in IHEXE rats (P > 0.05 for all). EXE rats showed lower levels of NHE-1 protein than CON rats (P < 0.05). However, the levels of LVFS%, LVEF%, TNF-α and IL-6 protein and NHE-1 mRNA did not differ between EXE and CON rats (P > 0.05 for all). These data indicated that exercise may provide a protective effect on IH-induced LV dysfunction by attenuating IH-induced myocardial NHE-1 hyperactivity.


Asunto(s)
Hipoxia/complicaciones , Condicionamiento Físico Animal , Intercambiadores de Sodio-Hidrógeno/fisiología , Disfunción Ventricular Izquierda/prevención & control , Animales , Interleucina-6/análisis , Masculino , Ratas , Ratas Sprague-Dawley , Factor de Necrosis Tumoral alfa/análisis
5.
Cell Physiol Biochem ; 33(2): 513-27, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24557000

RESUMEN

BACKGROUND/AIMS: Intermittent hypoxia (IH) may exert pre-conditioning-like cardioprotective effects and alter Ca(2+) regulation; however, the exact mechanism of these effects remains unclear. Thus, we examined Ca(2+)-handling mechanisms induced by IH in rat neonatal cardiomyocytes. METHODS: Cardiomyocytes were exposed to repetitive hypoxia-re-oxygenation cycles for 1-4 days. Mitochondrial reactive oxygen species (ROS) generation was determined by flow cytometry, and intracellular Ca(2+) concentrations were measured using a live-cell fluorescence imaging system. Protein kinase C (PKC) isoforms and Ca(2+)-handling proteins were analysed using immunofluorescence and western blotting. RESULTS: After IH exposure for 4 days, the rate of Ca(2+) extrusion from the cytosol to the extracellular milieu during 40-mM KCl-induced Ca(2+) mobilization increased significantly, whereas ROS levels increased mildly. IH activated PKC isoforms, which translocated to the membrane from the cytosol, and Na(+)/Ca(2+) exchanger-1, leading to enhanced Ca(2+) efflux capacity. Simultaneously, IH increased sarcoplasmic reticulum (SR) Ca(2+)-ATPase and ryanodine receptor 2 (RyR-2) activities and RyR-2 expression, resulting in improved Ca(2+) uptake and release capacity of SR in cardiomyocytes. CONCLUSIONS: IH-induced mild elevations in ROS generation can enhance Ca(2+) efflux from the cytosol to the extracellular milieu and Ca(2+)-mediated SR regulation in cardiomyocytes, resulting in enhanced Ca(2+)-handling ability.


Asunto(s)
Señalización del Calcio , Calcio/metabolismo , Miocitos Cardíacos/metabolismo , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Animales , Hipoxia de la Célula , Células Cultivadas , Miocitos Cardíacos/patología , Ratas , Ratas Sprague-Dawley
6.
J Biomed Sci ; 21: 46, 2014 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-24885237

RESUMEN

BACKGROUND: Intermittent hypoxia (IH) plays a critical role in sleep breathing disorder-associated hippocampus impairments, including neurocognitive deficits, irreversible memory and learning impairments. IH-induced neuronal injury in the hippocampus may result from reduced precursor cell proliferation and the relative numbers of postmitotic differentiated neurons. However, the mechanisms underlying IH-induced reactive oxygen species (ROS) generation effects on cell proliferation and neuronal differentiation remain largely unknown. RESULTS: ROS generation significantly increased after 1-4 days of IH without increased pheochromocytoma-12 (PC12) cell death, which resulted in increased protein phosphatase 2A (PP2A) mRNA and protein levels. After 3-4 days of IH, extracellular signal-regulated kinases 1/2 (ERK1/2) protein phosphorylation decreased, which could be reversed by superoxide dismutase (SOD), 1,10-phenanthroline (Phe), the PP2A phosphorylation inhibitors, okadaic acid (OKA) and cantharidin, and the ERK phosphorylation activator nicotine (p < 0.05). In particular, the significantly reduced cell proliferation and increased proportions of cells in the G0/G1 phase after 1-4 days of IH (p < 0.05), which resulted in decreased numbers of PC12 cells, could be reversed by treatment with SOD, Phe, PP2A inhibitors and an ERK activator. In addition, the numbers of nerve growth factor (NGF)-induced PC12 cells with neurite outgrowths after 3-4 days of IH were less than those after 4 days of RA, which was also reversed by SOD, Phe, PP2A inhibitors and an ERK activator. CONCLUSIONS: Our results suggest that IH-induced ROS generation increases PP2A activation and subsequently downregulates ERK1/2 activation, which results in inhibition of PC12 cell proliferation through G0/G1 phase arrest and NGF-induced neuronal differentiation.


Asunto(s)
Diferenciación Celular/genética , Activación Enzimática/genética , Proteína Fosfatasa 2/biosíntesis , Trastornos del Sueño-Vigilia/enzimología , Animales , Proliferación Celular/efectos de los fármacos , Hipocampo/enzimología , Hipoxia/complicaciones , Hipoxia/enzimología , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Factor de Crecimiento Nervioso/farmacología , Células PC12 , Fosforilación , Proteína Fosfatasa 2/antagonistas & inhibidores , Proteína Fosfatasa 2/genética , Ratas , Especies Reactivas de Oxígeno/metabolismo , Trastornos del Sueño-Vigilia/complicaciones , Trastornos del Sueño-Vigilia/patología , Superóxido Dismutasa/metabolismo
7.
Eur J Pharmacol ; 957: 176031, 2023 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-37660967

RESUMEN

Myocardial ischemia-reperfusion (I/R) injury triggers several cell death types, including apoptosis, autophagy, and ferroptosis. Licochalcone A (LCA), a natural flavonoid compound isolated from the root of Glycyrrhiza glabra, has been demonstrated to exert potential pharmacological benefits, such as antioxidant, antitumor, and anti-inflammatory activities. The present study aimed to investigate the involvement of ferroptosis in the pathogenesis of I/R and determine whether LCA can inhibit ferroptosis to prevent the myocardial I/R injury in rats. The effects of LCA on myocardial I/R injury were detected by examining the left ventricular-developed pressure and triphenyltetrazolium chloride staining. We conducted Western blotting analyses, ELISA assay, and quantitative real-time PCR to determine the levels of ferroptosis-related molecules. To demonstrate the cardioprotective effect of LCA in vitro, H9c2 and primary neonatal rat cardiomyocytes were co-treated with ferroptosis inducers (erastin, RSL3, or Fe-SP) and LCA for 16 and 24 h. Our ex vivo study showed that LCA increased the cardiac contractility, and reduced the infarct volume and ferroptosis-related biomarkers in rat hearts after I/R. Moreover, LCA reduced the levels of ferroptosis inducers-induced reactive oxygen species generation, lipid peroxidation, and ferroptosis-related biomarkers in cultured H9c2 cells and cardiomyocytes. LCA also reduced the Fe-SP-increased nuclear factor erythroid 2-related factor 2 and heme oxygenase-1 protein levels in cultured cardiomyocytes. In the present study, we showed that the LCA-induced cardioprotective effects in attenuating the myocardial I/R injury were correlated with ferroptosis regulation, and provided a possible new therapeutic strategy for prevention or therapy of the myocardial I/R injury.


Asunto(s)
Chalconas , Ferroptosis , Animales , Ratas , Chalconas/farmacología , Chalconas/uso terapéutico , Fenómenos Fisiológicos Cardiovasculares , Isquemia
8.
J Clin Med ; 12(13)2023 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-37445227

RESUMEN

A high malondialdehyde-oxidized low-density lipoprotein (MDA-oxLDL) level is associated with atherosclerotic cardiovascular diseases and major adverse cardiovascular events. A higher cardio-ankle vascular index (CAVI) is independently associated with an increased risk of cardiovascular events, cardiovascular mortality, myocardial infarction, and stroke in patients with cardiovascular risk. Thus, this study aimed to evaluate the relationship between serum MDA-oxLDL levels and CAVI in patients with triple-vessel coronary artery disease who underwent coronary artery bypass graft (CABG) surgery. Fasting blood samples and baseline characteristics were obtained from 88 patients who had undergone CABG. A commercialized enzyme-linked immunosorbent assay was used to measure MDA-oxLDL levels. An automatic pulse wave analyzer was used to measure CAVI values, and each side of CAVI values of ≥9 was designated as arterial stiffness. In total, 47 participants were assigned to the arterial stiffness group. More patients had diabetes mellitus, were older, and had higher serum MDA-oxLDL levels in the arterial stiffness group than in the control group. A multivariate logistic regression analysis disclosed that MDA-oxLDL and diabetes mellitus were independent predictors of arterial stiffness. Moreover, according to the Spearman's correlation analysis, the serum MDA-oxLDL level was positively associated with both left and right CAVI. Serum MDA-oxLDL levels were positively associated with arterial stiffness in patients who had undergone CABG.

9.
Int J Cardiol ; 375: 74-86, 2023 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-36513286

RESUMEN

BACKGROUND: Ischemia/reperfusion (I/R) is associated with severe cellular damage and death. Ferroptosis, a new form of regulated cell death caused by the accumulation of iron-mediated lipid peroxidation, has been found in several diseases including I/R injury, which was reported to be suppressed by flavonoids. Baicalein (BAI) and luteolin (Lut) are flavonoids and were shown to reduce the myocardial I/R injury. BAI was found to suppress ferroptosis in cancer cells via reducing reactive oxygen species (ROS) generation. However, the anti-ferroptosis effect of Lut on ferroptosis has not been reported. This study aimed to investigate whether ferroptosis reduction contributes to the BAI- and Lut-protected cardiomyocytes. METHODS: This research used erastin, RSL3, and Fe-SP to induce ferroptosis. Cell viability was examined using MTT assay. Annexin V-FITC, CM-H2DCFDA, and Phen Green SK diacetate (PGSK) fluorescent intensity were detected to analyze apoptotsis, ROS levels, and Fe2+ concentrations, respectively. qPCR and Western blot analysis were conducted to detect the levels of mRNA and protein, respectively. RESULTS: Our data show that BAI and Lut protected cardiomyocytes against ferroptosis caused by ferroptosis inducers and I/R. Moreover, both BAI and Lut decreased ROS and malondialdehyde (MDA) generation and the protein levels of ferroptosis markers, and restored Glutathione peroxidase 4 (GPX4) protein levels in cardiomyocytes reduced by ferroptosis inducers. BAI and Lut reduced the I/R-induced myocardium infarction and decreased the levels of Acsl4 and Ptgs2 mRNA. CONCLUSIONS: BAI and Lut could protect the cardiomyocytes against the I/R-induced ferroptosis via suppressing accumulation of ROS and MDA.


Asunto(s)
Daño por Reperfusión Miocárdica , Miocitos Cardíacos , Ratas , Animales , Miocitos Cardíacos/metabolismo , Luteolina/farmacología , Luteolina/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Daño por Reperfusión Miocárdica/prevención & control , Daño por Reperfusión Miocárdica/metabolismo , ARN Mensajero/metabolismo , Reperfusión , Isquemia/metabolismo
10.
J Biomed Sci ; 19: 29, 2012 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-22405498

RESUMEN

BACKGROUND: Episodic cessation of airflow during sleep in patients with sleep apnea syndrome results in intermittent hypoxia (IH). Our aim was to investigate the effects of IH on cerebellar granule cells and to identify the mechanism of IH-induced cell death. METHODS: Cerebellar granule cells were freshly prepared from neonatal Sprague-Dawley rats. IH was created by culturing the cerebellar granule cells in the incubators with oscillating O2 concentration at 20% and 5% every 30 min for 1-4 days. The results of this study are based on image analysis using a confocal microscope and associated software. Cellular oxidative stress increased with increase in IH. In addition, the occurrence of cell death (apoptosis and necrosis) increased as the duration of IH increased, but decreased in the presence of an iron chelator (phenanthroline) or poly (ADP-ribose) polymerase (PARP) inhibitors [3-aminobenzamide (3-AB) and DPQ]. The fluorescence of caspase-3 remained the same regardless of the duration of IH, and Western blots did not detect activation of caspase-3. However, IH increased the ratio of apoptosis-inducing factor (AIF) translocation to the nucleus, while PARP inhibitors (3-AB) reduced this ratio. RESULTS: According to our findings, IH increased oxidative stress and subsequently leading to cell death. This effect was at least partially mediated by PARP activation, resulting in ATP depletion, calpain activation leading to AIF translocation to the nucleus. CONCLUSIONS: We suggest that IH induces cell death in rat primary cerebellar granule cells by stimulating oxidative stress PARP-mediated calpain and AIF activation.


Asunto(s)
Hipoxia de la Célula/efectos de los fármacos , Cerebelo/efectos de los fármacos , Cerebelo/metabolismo , Estrés Oxidativo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Activación Transcripcional/efectos de los fármacos , Animales , Factor Inductor de la Apoptosis/metabolismo , Benzamidas/farmacología , Calpaína/metabolismo , Caspasa 3/metabolismo , Muerte Celular/efectos de los fármacos , Cerebelo/citología , Expresión Génica/efectos de los fármacos , Oxígeno/administración & dosificación , Fenantrolinas/farmacología , Poli(ADP-Ribosa) Polimerasa-1 , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Ratas , Ratas Sprague-Dawley
11.
Tzu Chi Med J ; 34(3): 310-317, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35912047

RESUMEN

Objectives: Cardiovascular diseases are one of the primary causes of death. Cardiomyocyte loss is a significant feature of cardiac injury. Ferroptosis is iron-dependent cell death, which occurs due to excess iron and reactive oxygen species (ROS) accumulation causing lipid peroxidation, and subsequent cell death. Ferroptosis has been confirmed to mediate ischemia/reperfusion-induced cardiomyopathy and chemotherapy-induced cardiotoxicity. Berberine (BBR) has been proven to protect the heart from cardiomyopathies, including cardiac hypertrophy, heart failure, myocardial infarction, and arrhythmias. It protects cardiomyocytes from apoptosis and autophagy. However, the relation between BBR and ferroptosis is still unknown. This study aimed to confirm if BBR reduces cardiac cell loss via inhibiting ferroptosis. Materials and Methods: We used erastin and Ras-selective lethal small molecule 3 (RSL3) to establish a ferroptosis model in an H9c2 cardiomyoblast cell line and rat neonatal cardiomyocytes to prove that BBR has a protective effect on cardiac cells via inhibiting ferroptosis. Results: In H9c2 cardiomyoblasts, the results showed that BBR reduced erastin and RSL3-induced cell viability loss. Moreover, BBR decreased ROS accumulation and lipid peroxidation in cells induced with ferroptosis. Furthermore, quantitative polymerase chain reaction results showed that Ptgs2 mRNA was reduced in BBR-treated cells. In rat neonatal cardiomyocytes, BBR reduced RSL3-induced loss of cell viability. Conclusion: These results indicated that BBR inhibited ferroptosis via reducing ROS generation and reducing lipid peroxidation in erastin and RSL3-treated cardiac cells.

12.
Oxid Med Cell Longev ; 2022: 9523491, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35082973

RESUMEN

Ferroptosis is an iron-dependent form of cell death caused by the inactivation of glutathione peroxidase 4 (GPX4) and accumulation of lipid peroxides. Ferroptosis has been found to participate in the ischemia-reperfusion (I/R) injury, leading to heart dysfunction and myocardial cell death. Xanthohumol (XN), a prenylated flavonoid isolated from Humulus lupulus, has multiple pharmacological activities, such as anti-inflammatory and antioxidant. This study is aimed at investigating whether XN could attenuate the I/R-induced ferroptosis in cardiomyocytes and the underlying mechanisms. Cardiomyocytes were treated with Fe-SP and RSL3, and the rat hearts were treated with I/R. The results from the present study show that XN was able to protect cardiomyocytes against Fe-SP- and RSL3-induced ferroptotic cell death by decreasing the production of lipid peroxidation and ROS, chelating iron, reducing the NRF2 protein level, and modulating the protein levels of GPX4. Moreover, XN significantly decreased the mRNA levels of ferroptosis markers, Ptgs2 and Acsl4, and the protein levels of ACSL4 and NRF2 and modulated the protein levels of GPX4 in I/R-treated hearts. The findings from the present study suggest that XN might have the therapeutic potential for the I/R-induced ferroptosis injury.


Asunto(s)
Ferroptosis/efectos de los fármacos , Flavonoides/uso terapéutico , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Miocardio/patología , Propiofenonas/uso terapéutico , Animales , Flavonoides/farmacología , Propiofenonas/farmacología , Ratas , Ratas Sprague-Dawley
13.
Sleep Breath ; 15(4): 845-54, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21136300

RESUMEN

RATIONALE: Chronic intermittent hypoxia (CIH) is thought to induce several cardiovascular effects in patients with obstructive sleep apnoea (OSA). However, the effects of CIH on patients with long-standing hypertension are unknown. PURPOSE: This prospective study aimed to investigate the influence of combined OSA and hypertension on cardiomyocyte death. METHODS: Wistar-Kyoto rats (WKY) and spontaneously hypertensive rats (SHR) were exposed to repetitive hypoxia-reoxygenation cycles (30 s of 5% O(2); 45 s of 21% O(2)) or room air for 6 h/day during the light phase (10 a.m.-4 p.m.) for 10, 20, or 30 days, and the levels of necrosis and apoptosis induced in their left ventricular cardiomyocyte were examined. RESULTS: CIH increased the accumulation of reactive oxygen species, which induced cardiomyocyte necrosis in WKY and SHR (both p < 0.05). Cardiomyocyte oxidative stress levels by CIH were higher in SHR than in WKY (p < 0.05); therefore, cardiomyocyte necrosis was amplified (p < 0.05). Notably, if a superoxide-scavenging agent is injected beforehand, cardiomyocyte necrosis can be effectively inhibited (p < 0.05). When WKY and SHR are exposed to CIH, increases in mitochondria-released cytochrome c and activation of caspase-3 are found in the cytosolic fraction only in WKY. CONCLUSIONS: CIH causes cardiomyocyte loss in SHR mainly through cardiomyocyte necrosis. In WKY however, CIH simultaneously induces apoptosis and necrosis of cardiomyocytes.


Asunto(s)
Muerte Celular/fisiología , Hipertensión/patología , Miocitos Cardíacos/patología , Disfunción Ventricular Izquierda/patología , Animales , Peroxidación de Lípido/fisiología , Masculino , Microscopía Fluorescente , Necrosis , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Especies Reactivas de Oxígeno/metabolismo , Espectrometría de Fluorescencia , Superóxido Dismutasa/metabolismo
14.
Eur J Appl Physiol ; 111(8): 1939-50, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21249391

RESUMEN

We investigated whether exercise provides beneficial effects to attenuate intermittent hypoxia (IH)-induced myocardial apoptosis. Male Sprague-Dawley rats were randomly assigned to four groups: control (CON), IH, exercise (EXE) or IH interspersed with EXE (IHEXE). IH rats were exposed to repetitive hypoxia-reoxygenation cycles (30 s of 5% O(2); 45 s of 21% O(2), 6 h day(-1)) during the light phase (1000-1600 h) for 12 consecutive days. EXE rats were habituated to treadmill running for 5 days, permitted 2 days of rest, followed by 5 exercise bouts (30 m min(-1) for 60 min on a 2% grade) on consecutive days during the dark phase (2000-2200 h). IHEXE rats were exposed to IH during the light phase interspersed with exercise programs during the dark phase on the same day. Apoptosis levels, cytochrome c (Cyt-c), cleaved caspase-3, oxidative stress and antioxidant capacity were determined in the left ventricular (LV) myocardium. IH rats showed higher myocardial levels of the apoptotic index, mitochondria-released Cyt-c, cleaved caspase-3 and oxidative stress and lower catalase activity levels than CON rats (p < 0.05, for all). These changes were not observed in EXE rats (p > 0.05, for all) except that catalase activity increased (p < 0.05). IHEXE rats showed lower myocardial levels of apoptotic index, mitochondria-released Cyt-c, cleaved caspase-3 and oxidative stress and higher catalase activity levels (p < 0.05, for all) than IH rats. We conclude that short-term exercise provides potent cardioprotective effects by attenuating IH-induced myocardial apoptosis.


Asunto(s)
Apoptosis/fisiología , Infarto del Miocardio/etiología , Infarto del Miocardio/prevención & control , Condicionamiento Físico Animal/fisiología , Disfunción Ventricular Izquierda/prevención & control , Animales , Hipoxia de la Célula/fisiología , Peroxidación de Lípido/fisiología , Masculino , Infarto del Miocardio/metabolismo , Isquemia Miocárdica/etiología , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/patología , Isquemia Miocárdica/fisiopatología , Periodicidad , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ratas , Ratas Sprague-Dawley , Factores de Tiempo , Disfunción Ventricular Izquierda/etiología , Disfunción Ventricular Izquierda/metabolismo
15.
Tzu Chi Med J ; 33(4): 323-331, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34760626

RESUMEN

Catecholaminergic polymorphic ventricular tachycardia (CPVT), a rare autosomal dominant or recessive disease, usually results in syncope or sudden cardiac death. Most CPVT patients do not show abnormal cardiac structure and electrocardiogram features and symptoms, usually onset during adrenergically mediated physiological conditions. CPVT tends to occur at a younger age and is not easy to be diagnosed and managed. The main cause of CPVT is associated with mishandling Ca2+ in cardiomyocytes. Intracellular Ca2+ is strictly controlled by a protein located in the sarcoplasm reticulum (SR), such as ryanodine receptor, histidine-rich Ca2+-binding protein, triadin, and junctin. Mutation in these proteins results in misfolding or malfunction of these proteins, thereby affecting their Ca2+-binding affinity, and subsequently disturbs Ca2+ homeostasis during excitation-contraction coupling (E-C coupling). Furthermore, transient disturbance of Ca2+ homeostasis increases membrane potential and causes Ca2+ store overload-induced Ca2+ release, which in turn leads to delayed after depolarization and arrhythmia. Previous studies have focused on the interaction between ryanodine receptors and protein kinase or phosphatase in the cytosol. However, recent studies showed the regulation signaling for ryanodine receptor not only from the cytosol but also within the SR. The changing of Ca2+ concentration is critical for protein interaction inside the SR which changes protein conformation to regulate the open probability of ryanodine receptors. Thus, it influences the threshold of Ca2+ released from the SR, making it easier to release Ca2+ during E-C coupling. In this review, we briefly discuss how Ca2+ handling protein variations affect the Ca2+ handling in CPVT.

16.
Eur J Pharmacol ; 905: 174183, 2021 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-34015318

RESUMEN

This study aimed to investigate whether methyl palmitate (MP) exerts cardioprotective effect against the ischemia/reperfusion (I/R) injury and its mechanisms underlying. The cultured adult cardiomyocytes were treated with vehicle or lactic acid ischemic buffer (pH 6.8) during hypoxia/reoxygenation. In addition, the cardioprotective effect of MP was evaluated using the ex vivo heart model of I/R injury. Here, we found that MP significantly reduced the I/R-induced cardiomyocyte death. Treatment with GW1100 (a GPR40-antagonist) or wortmannin (a phosphatidylinositol 3-kinase, PI3K, specific inhibitor) significantly attenuated the level of phospho-AKT (p-AKT) and abolished the MP-induced cardioprotection against the I/R-induced injury. Using the ex vivo I/R model, we also demonstrated that pretreatment with MP significantly reduced the size of myocardial infarction and the levels of cleaved-caspase 3 and MDA, and increased the protein levels of GPR40 and p-AKT induced by I/R. The cardioprotective effect of MP was evaluated also using the in vivo heart model of I/R injury. We demonstrated that post-ischemic treatment with MP significantly attenuated the size of myocardial infarction and the serum level of CK-MB induced by in vivo I/R model. Taken together, our data suggest that MP could provide significant cardioprotection against the I/R injury, and the underlying mechanisms by which MP prevented the cardiomyocyte death might be mediated through the GPR40-activated PI3K/AKT signaling pathways. These findings suggest the potential applications of MP in the treatment of I/R-induced heart injury.


Asunto(s)
Cardiotónicos/farmacología , Daño por Reperfusión Miocárdica/prevención & control , Palmitatos/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Cardiotónicos/uso terapéutico , Forma MB de la Creatina-Quinasa/sangre , Masculino , Modelos Biológicos , Infarto del Miocardio/tratamiento farmacológico , Infarto del Miocardio/patología , Miocitos Cardíacos/efectos de los fármacos , Palmitatos/uso terapéutico , Cultivo Primario de Células , Ratas Sprague-Dawley
17.
Stem Cells Int ; 2021: 9938649, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34650609

RESUMEN

Adipogenic differentiation from stem cells has become a research target due to the increasing interest in obesity. It has been indicated that adipocytes can secrete palmitic acid methyl ester (PAME), which is able to regulate stem cell proliferation. However, the effects of PAME on adipogenic differentiation in stem cell remain unclear. Here, we present that the adipogenic differentiation medium supplemented with PAME induced the differentiation of rat adipose tissue-derived mesenchymal stem cells (rAD-MSCs) into adipocyte. rAD-MSCs were treated with PAME for 12 days and then subjected to various analyses. The results from the present study show that PAME significantly increased the levels of adipogenic differentiation markers, PPARγ and Gpd1, and enhanced adipogenic differentiation in rAD-MSCs. Furthermore, the level of GPR40/120 protein increased during induction of adipocyte differentiation in rAD-MSCs. Cotreatment with PAME and a GPR40/120 antagonist together inhibited the PAME-enhanced adipogenic differentiation. Moreover, PAME significantly increased phosphorylation of extracellular signal-regulated kinases (ERK), but not AKT and mTOR. Cotreatment with PAME and a GPR40/120 antagonist together inhibited the PAME-enhanced ERK phosphorylation and adipogenic differentiation. PAME also increased the intracellular Ca2+ levels. Cotreatment with PAME and a Ca2+ chelator or a phospholipase C (PLC) inhibitor prevented the PAME-enhanced ERK phosphorylation and adipogenic differentiation. Our data suggest that PAME activated the GPR40/120/PLC-mediated pathway, which in turn increased the intracellular Ca2+ levels, thereby activating the ERK, and eventually enhanced adipogenic differentiation in rAD-MSCs. The findings from the present study might help get insight into the physiological roles and molecular mechanism of PAME in regulating stem cell differentiation.

18.
Antioxidants (Basel) ; 10(11)2021 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-34829537

RESUMEN

To identify the core structure of 2-aminoethoxydiphenyl borate (2-APB) responsible for the anti-oxidative and protective effect on the ischemia/reperfusion (I/R)-induced heart injury, various 2-APB analogues were analyzed, and several antioxidant assays were performed. Cell viability was determined using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Myocardial infarct size was quantified using triphenyl tetrazolium chloride (TTC) staining. The levels of tumor necrosis factor-alpha (TNF-α) and cleaved-caspase-3 protein were evaluated as an indicator for the anti-inflammatory and anti-apoptotic effect, respectively. Our data show that 2-APB, diphenylborinic anhydride (DPBA) and 3-(diphenylphosphino)-1-propylamine (DP3A) all exerted the anti-oxidative activity, but only 2-APB and DPBA can scavenge H2O2. 2-APB and DPBA can potently inhibit hydrogen peroxide (H2O2)- and hypoxanthine/xanthine oxidase (HX/XOD)-induced increases in intracellular H2O2 and H9c2 cell death. 2-APB and DPBA were able to decrease the I/R-induced adult rat cardiomyocytes death, myocardial infarct size, and the levels of malondialdehyde (MDA) and creatine kinase-MB (CK-MB). Our results suggest that the two benzene rings with a boron atom comprise the core structure of 2-APB responsible for the anti-oxidative effect mediated through the reaction with H2O2 and generation of phenolic compounds, which in turn reduced the I/R-induced oxidative stress and injury in the rat heart.

19.
Biomolecules ; 11(11)2021 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-34827665

RESUMEN

Myocardial ischemia/reperfusion (I/R) injury has been associated with ferroptosis, which is characterized by an iron-dependent accumulation of lipid peroxide to lethal levels. Gossypol acetic acid (GAA), a natural product taken from the seeds of cotton plants, prevents oxidative stress. However, the effects of GAA on myocardial I/R-induced ferroptosis remain unclear. This study investigated the ability of GAA to attenuate I/R-induced ferroptosis in cardiomyocytes along with the underlying mechanisms in a well-established rat model of myocardial I/R and isolated neonatal rat cardiomyocytes. H9c2 cells and cardiomyocytes were treated with the ferroptosis inducers erastin, RSL3, and Fe-SP. GAA could protect H9c2 cells against ferroptotic cell death caused by these ferroptosis inducers by decreasing the production of malondialdehyde and reactive oxygen species, chelating iron content, and downregulating mRNA levels of Ptgs2. GAA could prevent oxygen-glucose deprivation/reperfusion-induced cell death and lipid peroxidation in the cardiomyocytes. Moreover, GAA significantly attenuated myocardial infarct size, reduced lipid peroxidation, decreased the mRNA levels of the ferroptosis markers Ptgs2 and Acsl4, decreased the protein levels of ACSL4 and NRF2, and increased the protein levels of GPX4 in I/R-induced ex vivo rat hearts. Thus, GAA may play a cytoprotectant role in ferroptosis-induced cardiomyocyte death and myocardial I/R-induced ferroptotic cell death.


Asunto(s)
Daño por Reperfusión Miocárdica , Animales , Ferroptosis , Gosipol/análogos & derivados , Masculino , Ratas
20.
Stem Cells Int ; 2019: 7606238, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31885624

RESUMEN

Bone marrow-derived mesenchymal cells (BM-MSCs) are able to differentiate into adipocytes, which can secrete adipokines to affect BM-MSC proliferation and differentiation. Recent evidences indicated that adipocytes can secrete fatty acid metabolites, such as palmitic acid methyl ester (PAME), which is able to cause vasorelaxation and exerts anti-inflammatory effects. However, effects of PAME on BM-MSC proliferation remain unclear. The aim of this study was to investigate the effect of PAME on human BM-MSC (hBM-MSC) proliferation and its underlying molecular mechanisms. hBM-MSCs were treated with PAME for 48 h and then subjected to various analyses. The results from the present study show that PAME significantly reduced the levels of G2/M phase regulatory proteins, cyclin-dependent kinase 1 (Cdk1), and cyclin B1 and inhibited proliferation in hBM-MSCs. Moreover, the level of Mdm2 protein decreased, while the levels of p21 and p53 protein increased in the PAME-treated hBM-MSCs. However, PAME treatment did not significantly affect apoptosis/necrosis, ROS generation, and the level of Cdc25C protein. PAME also induced intracellular acidosis and increased intracellular Ca2+ levels. Cotreatment with PAME and Na+/H+ exchanger inhibitors together further reduced the intracellular pH but did not affect the PAME-induced decreases of cell proliferation and increases of the cell population at the G2/M phase. Cotreatment with PAME and a calcium chelator together inhibited the PAME-increased intracellular Ca2+ levels but did not affect the PAME-induced cell proliferation inhibition and G2/M cell cycle arrest. Moreover, the half-life of p53 protein was prolonged in the PAME-treated hBM-MSCs. Taken together, these results suggest that PAME induced p53 stabilization, which in turn increased the levels of p53/p21 proteins and decreased the levels of Cdk1/cyclin B1 proteins, thereby preventing the activation of Cdk1, and eventually caused cell cycle arrest at the G2/M phase. The findings from the present study might help get insight into the physiological roles of PAME in regulating hBM-MSC proliferation.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA