Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Cell Biol Int ; 46(4): 637-648, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-34989460

RESUMO

Melatonin has been proven to reduce myocardial ischemia-reperfusion (MI/R) injury. However, in most studies, melatonin was administered before MI/R, thus, the results lack clinical significance in patients with acute myocardial infarction. We hypothesize that melatonin posttreatment at different times has different curative effects. Administered of Melatonin (150 µM) at different times after the onset of reoxygenation (t = -15, 0, 5, 10, 15, and 30 min). Cellular apoptosis, oxidative stress, and mitochondrial function were assessed. Mitophagy-related protein levels, mitochondrial membrane potential (MMP), and mitochondrial permeability transition pore (mPTP) activity were also measured. A/R injury upregulated mitophagy, which was associated with increased cellular apoptosis, oxidative stress, and mitochondrial dysfunction. Melatonin posttreatment (t = -15, 0, 5, 10, 15, and 30 min) significantly inhibited excessive mitophagy after A/R injury, reduced cellular apoptosis and oxidative stress, restored mitochondrial function and MMP, and restrained mPTP opening. The therapeutic time window in which melatonin posttreatment protected H9c2 cells against A/R injury was large (from -15 to 30 min after the onset of reperfusion), but the earlier the melatonin administration was, the better its protective effect was. This mechanism is likely due to a reduction in mPTP activity and MMP collapse, which lead to the inhibition of mitophagy.


Assuntos
Melatonina , Traumatismo por Reperfusão Miocárdica , Apoptose , Humanos , Hipóxia/metabolismo , Melatonina/farmacologia , Melatonina/uso terapêutico , Mitofagia , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Traumatismo por Reperfusão Miocárdica/metabolismo , Miócitos Cardíacos/metabolismo
2.
Neuroimmunomodulation ; 29(2): 117-127, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34856557

RESUMO

INTRODUCTION: Isoflurane (ISO) may cause neuronal apoptosis and synaptic disorder during development, and damage long-term learning and memory function. This observation aimed to study the function of H19 in vitro and in vivo tests and the further mechanism was identified. METHODS: ISO cell models and rat models were established and reactive oxygen species (ROS) identified. The viability and apoptosis of HT22 cells were detected by the MTT and flow cytometer. Morris water maze test was conducted to analyze the neurotoxicity of ISO on spatial learning and memory ability. Quantitative PCR was the method to verify the expression of H19. The concentration of inflammatory indicators was identified by enzyme-linked immunosorbent assay. RESULTS: 1.5% and 2% ISO led to the neurotoxicity of HT22 cells and increased expression of H19. Silenced H19 meliorated these adverse impacts of ISO. Interference of H19 exerted neuroprotective roles by repressing modified neurological severity score, inhibiting escape latency, elevating distance and time of target area, and controlling ROS and inflammation. MiR-17-5p might be a promising competing endogenous RNA of H19. The expression of miR-17-5p was reduced in the ISO group and reversed by the absence of H19. CONCLUSION: Our results of in vitro and in vivo assay indicated that the absence of HT22 is a neuroprotective regulator of cognition and inflammation by accumulating miR-17-5p.


Assuntos
Isoflurano , MicroRNAs , RNA Longo não Codificante , Animais , Cognição , Isoflurano/toxicidade , MicroRNAs/genética , Doenças Neuroinflamatórias , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Ratos
3.
Br J Anaesth ; 127(4): 620-628, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34446224

RESUMO

BACKGROUND: Previous studies on the association between renal tissue desaturation and acute kidney injury (AKI) in infant cardiac surgery are limited by small sample sizes and inconsistent results. This prospective study aimed to determine the association between renal desaturation and AKI in infants undergoing surgical repair of an isolated ventricular septal defect (VSD). METHODS: Infants undergoing VSD repair involving cardiopulmonary bypass participated in this prospective cohort study. The exposure of interest was renal tissue desaturation, defined as at least 20% decrease in saturation from baseline for at least 60 consecutive seconds. Intraoperative care was not guided by renal oxygenation, as the anaesthesiologists were blinded to the monitor. The outcome was AKI arising within postoperative Days 1-3. The primary analysis was based on propensity score-matched infants with and without intraoperative renal desaturation. RESULTS: Intraoperative renal desaturation was detected in 38 of 242 infants using near-infrared spectroscopy. This group of infants was matched with 114 infants without intraoperative renal saturation after propensity score matching. Acute kidney injury occurred in 47% (18/38) and 27% (31/114) of infants with or without renal desaturation, respectively. Infants with renal desaturation had higher odds of developing AKI than infants without renal desaturation based on conditional logistic regression (odds ratio 2.79; 95% confidence interval: 1.21-6.44; P=0.016). The cumulative time of renal desaturation correlated moderately with the ratio of postoperative peak creatinine to preoperative baseline creatinine (r=0.51; P<0.001). CONCLUSIONS: Intraoperative renal desaturation is associated with increased odds of developing AKI after surgical repair of an isolated VSD involving cardiopulmonary bypass in infants. CLINICAL TRIAL REGISTRATION: NCT03941015.


Assuntos
Injúria Renal Aguda/etiologia , Procedimentos Cirúrgicos Cardíacos/métodos , Comunicação Interventricular/cirurgia , Complicações Pós-Operatórias/epidemiologia , Injúria Renal Aguda/epidemiologia , Ponte Cardiopulmonar/métodos , Estudos de Coortes , Creatinina/metabolismo , Feminino , Humanos , Lactente , Rim/metabolismo , Masculino , Oxigênio/metabolismo , Estudos Prospectivos , Espectroscopia de Luz Próxima ao Infravermelho
4.
Cardiol Young ; 31(3): 371-376, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33541455

RESUMO

OBJECTIVE: To analyse the changes of different ventilation on regional cerebral oxygen saturation and cerebral blood flow in infants during ventricular septal defect repair. METHODS: Ninety-two infants younger than 1 year were enrolled in the study. End-expiratory tidal pressure of carbon dioxide was maintained at 40-45 and 35-39 mmHg in relative low and high ventilation groups. Regional cerebral oxygen saturation and flow velocity of the middle cerebral artery were recorded after anaesthesia (T0), cut pericardium (T1), separation from cardiopulmonary bypass (T2), the end of modified ultrafiltration, (T3) and at the end of operation (T4). RESULTS: The relative low ventilation group exhibited a significantly high regional cerebral oxygen saturation at each time point except for T2 (T0:77 ± 4, T1:76 ± 5, T3:76 ± 8, T4:76 ± 8, respectively, p < 0.001). Flow velocity of the middle cerebral artery in the relative low ventilation group was higher compared to the relative high ventilation group at each time point except for T2 (T0:53 ± 14, T1:54 ± 15, T3:53 ± 17, T4:52 ± 16, respectively, p < 0.001). Between the two groups, T2 showed the lowest middle cerebral artery flow velocity (relative low ventilation: 39 ± 15, relative high ventilation: 39 ± 11, p < 0.001). CONCLUSION: The infants' regional cerebral oxygen saturation and middle cerebral artery flow velocity performed better in the range of 40-45 mmHg end-expiratory tidal pressure of carbon dioxide during CHD surgery. Modified ultrafiltration increased cerebral oxygen saturation. It was important to regulate ventilation in order to balance cerebral oxygen in infants.


Assuntos
Comunicação Interventricular , Ultrafiltração , Ponte Cardiopulmonar , Circulação Cerebrovascular , Humanos , Lactente , Oxigênio
5.
Paediatr Anaesth ; 30(9): 977-983, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32648613

RESUMO

BACKGROUND: Impaired cerebrovascular reactivity to carbon dioxide was proposed to contribute to neurological morbidity in children undergoing cardiac surgery. The objective of this study was to assess carbon dioxide reactivity and regional cerebral oxygen saturation in children younger than 3 years. METHODS: This study enrolled children younger than 3 years undergoing ventricular septal defect repair. The cohort was divided into three age groups: younger than 6 months, 6-12 months, and 12-36 months. Under steady-state anesthesia, carbon dioxide reactivity was calculated by measuring changes in middle cerebral artery blood flow velocity using transcranial Doppler sonography. Regional cerebral oxygen saturation changes were measured by near-infrared spectroscopy while endtidal carbon dioxide pressure was adjusted from 30 to 45 mm Hg. RESULTS: Carbon dioxide reactivity showed a statistically significant increasing relationship with age (younger than 6 months group: 4.42% ± 2.73%, 6-12 months group: 5.86% ± 1.91%, 12-36 months group: 7.58% ± 1.49%; P < .001). Regional cerebral oxygen saturation showed a statistically significant increasing relationship with age (younger than 6 months group: 65% ± 6%, 6-12 months group: 68% ± 5%, 12-36 months group: 70% ± 5%; P = .027). Regional cerebral oxygen saturation showed a statistically significant increasing relationship with endtidal carbon dioxide pressure in all children (P < .001). CONCLUSION: Abnormal carbon dioxide reactivity is prevalent in children younger than 3 years and the degree varies according to age.


Assuntos
Dióxido de Carbono , Comunicação Interventricular , Velocidade do Fluxo Sanguíneo , Circulação Cerebrovascular , Criança , Comunicação Interventricular/diagnóstico por imagem , Comunicação Interventricular/cirurgia , Humanos , Lactente , Recém-Nascido , Oxigênio , Ultrassonografia Doppler Transcraniana
6.
Exp Cell Res ; 367(2): 257-263, 2018 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-29630893

RESUMO

Penehyclidine hydrochloride (PHC) preconditioning can alleviate myocardial ischemia/reperfusion (I/R) injury and inhibits the upregulation of voltage-dependent anion channel 1 (VDAC1) during I/R. To validate that VDAC1 is a bona fide target of PHC for the protection against myocardial I/R injury, VDAC1 expression construct was delivered by lentiviruses into rat left ventricular myocardium before PHC preconditioning and myocardial I/R. Overexpression of VDAC1 exacerbated cardiac dysfunction and myocardial injury following I/R, and abolished the cardioprotective effect of PHC during I/R injury. Moreover, VDAC1 overexpression with myocardial I/R further increased cytochrome c release from mitochondria to cytoplasm, elevated the levels of cleaved caspase-3 and Bax, and decreased the level of Bcl-2 as compared with I/R alone, and PHC-mediated inhibition of mitochondria-dependent apoptosis during myocardial I/R was abolished by VDAC1 overexpression. In addition, VDAC1 was overexpressed in H9c2 cardiomyocytes undergoing anoxia/reoxygenation (A/R) with or without PHC pretreatment. The in vitro results showed that overexpression of VDAC1 further reduced mitochondrial membrane potential, increased mitochondrial membrane permeability and enhanced mitochondria-dependent apoptosis in H9c2 cells after A/R, and VDAC1 overexpression abrogated the protective effect of PHC on the mitochondrial function and integrity during A/R. In conclusion, exogenous overexpression of VDAC1 during myocardial I/R inhibits the cardioprotective effects of PHC. These effects may be associated with the suppression of VDAC1 expression.


Assuntos
Cardiotônicos/uso terapêutico , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Quinuclidinas/uso terapêutico , Canal de Ânion 1 Dependente de Voltagem/metabolismo , Animais , Apoptose , Permeabilidade da Membrana Celular , Masculino , Potencial da Membrana Mitocondrial , Mitocôndrias/metabolismo , Traumatismo por Reperfusão Miocárdica/metabolismo , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/fisiologia , Ratos , Ratos Wistar , Canal de Ânion 1 Dependente de Voltagem/genética
7.
Artigo em Inglês | MEDLINE | ID: mdl-38988172

RESUMO

BACKGROUND: Melatonin (MT) has been demonstrated to have cardioprotective effects. Nevertheless, the precise mechanism through which MT provides protection against the etiology of LPS-induced myocardial injury remains uncertain. In this investigation, our objective was to explore the impact of MT on LPS-induced myocardial injury in an in vitro setting. METHODS: H9C2 cells were categorized into four groups: a control group (H9C2 group), an MT group, an LPS group, and an MT + LPS group. The H9C2 group received treatment with sterile saline solution, the LPS group was exposed to 5 µg/mL LPS for 24 hours, the MT + LPS group underwent pretreatment with 150 µmol/L MT for 2 hours, followed by exposure to 5 µg/mL LPS for 24 hours, and the MT group received only 150 µmol/L MT for 2 hours. Cell viability and lactate dehydrogenase (LDH) release were assessed using the CCK-8 assay and LDH activity assay, respectively. The levels of reactive oxygen species (ROS) were quantified in each group of cells, and the percentage of propidium iodide (PI)-stained apoptotic cells was determined by flow cytometry. The mRNA levels of caspase11, GSDMD, and IL-18 in each group of cells were quantified. RESULTS: MT treatment significantly protected H9C2 cells from LPS-induced damage, as evidenced by decreased LDH release. LPS treatment markedly increased ROS levels in H9C2 cells, which were subsequently reduced by MT. LPS caused a substantial decrease in superoxide dismutase (SOD) activity and a significant increase in malondialdehyde (MDA) levels, while MT treatment significantly reversed these effects. Additionally, MT markedly enhanced the proportion of viable H9C2 cells compared to LPS-treated controls, as evidenced by the PI staining assay. LPS upregulated both mRNA levels and protein levels of IL-18 in H9C2 cells. However, MT treatment effectively mitigated this LPS-induced increase. Furthermore, MT significantly decreased LPS-induced protein levels of cleaved-caspase 11 and GSDMD-N in H9C2 cells. CONCLUSION: Overall, our findings suggest that MT inhibits the Caspase11-GSDMD signaling pathway via pyroptosis-related proteins (caspase-11 and GSDMD-N) and reduces the expression of inflammation-related cytokines (IL-18), thereby exerting a protective effect on H9C2 cells after LPS injury.

8.
Int Immunopharmacol ; 137: 112373, 2024 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-38852523

RESUMO

Although penehyclidine hydrochloride (PHC) has been identified to alleviate myocardial injury induced by ischemia/reperfusion (I/R), the regulatory molecules and related mechanisms are unknown. In this study, bioinformatics, molecular biology, and biochemistry methods were used to explore the molecular mechanisms and targets of PHC. In the myocardial ischemia-reperfusion injury (MIRI)-induced rat model, PHC pretreatment significantly improved cardiac function (p < 0.01). Multiple differentially expressed genes, including Z-DNA binding protein 1 (ZBP1), were identified through mRNA sequencing analysis of myocardial ischemic penumbra tissue in MIRI rats. The transduction of the ZBP1 adenovirus vector (Ad-Zbp1) in PHC-pretreated rats exhibited a reversible augmentation in myocardial infarct size (p < 0.01), pronounced pathological damage to the myocardial tissue, as well as a significant elevation of serum myocardial enzymes (p < 0.05). The interaction among ZBP1, fas-associating via death domain (FADD), and receptor-interacting serine/threonine-protein kinase 3 (RIPK3) leads to a remarkable up-regulation of cleaved-Caspase-1 (Cl-Casp-1), N-terminal gasdermin D (N-GSDMD), phospho-mixed lineage kinase domain-like Ser358 (p-MLKLS358), and other regulatory proteins, thereby triggering pyroptosis, apoptosis, and necroptosis (PANoptosis) in cardiomyocytes of MIRI rats. Moreover, the transduction of Ad-Zbp1 in the oxygen-glucose deprivation/re-oxygenation (OGD/R)-induced H9c2 cell model also dramatically augmented the number of cell deaths. However, the intervention of PHC considerably enhanced cell viability (p < 0.01), effectively mitigated the release of myocardial enzymes (p < 0.05), and markedly attenuated the expression levels of PANoptosis regulatory proteins through restraint of ZBP1 expression. Therefore, the therapeutic efficacy of PHC in improving MIRI might be attributed to targeting ZBP1-mediated PANoptosis.


Assuntos
Traumatismo por Reperfusão Miocárdica , Quinuclidinas , Ratos Sprague-Dawley , Animais , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Traumatismo por Reperfusão Miocárdica/patologia , Traumatismo por Reperfusão Miocárdica/metabolismo , Masculino , Ratos , Quinuclidinas/uso terapêutico , Quinuclidinas/farmacologia , Necroptose/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Linhagem Celular , Apoptose/efeitos dos fármacos , Miocárdio/patologia , Miocárdio/metabolismo , Modelos Animais de Doenças , Humanos , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/genética
9.
Eur J Pharmacol ; 963: 176292, 2024 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-38128867

RESUMO

Excessive or uncontrolled mitophagy may result in a drastic shortage of healthy mitochondrial for ATP supply after reperfusion, leading to irreversible myocardial damage. Melatonin, a hormone produced by the pineal gland, has been proven to ameliorate myocardial ischemia-reperfusion (I/R) injury via regulating mitophagy. However, its underlying mechanism has not been fully elucidated. The present study focused on the role of mitophagy in the cardioprotective effects of melatonin by using the myocardial I/R rat model. The rats were pretreated with or without the apelin inhibitor ML221, the sirtuin 3 (SIRT3) inhibitor 3-TYP and then subjected to I/R injury, with melatonin administrated 10 min before reperfusion. The effects of melatonin on myocardial infarct size, biomarkers of myocardial injury, oxidative stress, and mitochondrial function were detected, and the expression of apelin, SIRT3, and mitophagy-related proteins were also measured. Excessive mitophagy was activated after I/R injury and was correlated with oxidative stress and mitochondrial dysfunction. Melatonin pretreatment ameliorated myocardial injury by decreasing oxidative stress, restoring mitochondrial function, and inhibiting excessive mitophagy. However, ML221 or 3-TYP disrupted these beneficial effects of melatonin on I/R injury. Taken together, these results suggest that melatonin pretreatment ameliorates myocardial I/R injury through regulating the apelin/SIRT3 pathway to inhibit excessive mitophagy.


Assuntos
Melatonina , Traumatismo por Reperfusão Miocárdica , Traumatismo por Reperfusão , Sirtuína 3 , Ratos , Animais , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Traumatismo por Reperfusão Miocárdica/metabolismo , Sirtuína 3/metabolismo , Melatonina/farmacologia , Melatonina/uso terapêutico , Melatonina/metabolismo , Mitofagia , Apelina
10.
Comb Chem High Throughput Screen ; 26(6): 1204-1213, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-35838232

RESUMO

AIMS AND OBJECTIVE: The lack of effective treatments for myocardial ischemiareperfusion (MI-R) injury severely restricts the effectiveness of the treatment of ischemic heart disease. In the present research, we aimed to investigate the protective effect and molecular mechanism of penehyclidine hydrochloride (PHC) on MI-R cells. METHODS: Cell viability was quantified using CCK8. Cell apoptosis was analyzed using flow cytometry. Western blot and Elisa assays were used for the detection of target proteins. RESULTS: PHC pretreatment attenuated the inhibition of cell viability and decreased the percentage of apoptosis induced by simulated ischemia reperfusion (SIR). Platelet-derived growth factor B (PDGF-B) and its downstream AKT pathway were activated in PHC pretreated cells. After siRNAPDGF- B transfection, cell viability was inhibited and apoptosis was activated in PHC pretreated SIR cells, suggesting that PHC protected cells from SIR. PDGF-B knockdown also increased the levels of CK, LDH, IL-6 and TNF-α in PHC pretreated SIR cells. The effect of AKT inhibitor on H9C2 cells was consistent with that of PDGF-B knockdown. CONCLUSION: PHC pretreatment can protect cardiomyocytes from the decrease of cell activity and the increase of apoptosis caused by reperfusion through up-regulating PDGF-B to activate PI3K pathway. Our study indicates that PHC is a potential drug to protect cells from reperfusion injury and PDGF-B is a potential target for preventing MI-R injury.


Assuntos
Miócitos Cardíacos , Traumatismo por Reperfusão , Ratos , Animais , Miócitos Cardíacos/metabolismo , Ratos Wistar , Proteínas Proto-Oncogênicas c-sis/metabolismo , Proteínas Proto-Oncogênicas c-sis/farmacologia , Proteínas Proto-Oncogênicas c-sis/uso terapêutico , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Traumatismo por Reperfusão/tratamento farmacológico , Isquemia/metabolismo , Apoptose
11.
J Cardiovasc Transl Res ; 16(6): 1373-1382, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37584895

RESUMO

Penehyclidine hydrochloride (PHC) is an anticholinergic drug with cardioprotective effects. Ferroptosis is closely related to myocardial ischaemia-reperfusion injury (MIRI). In the present study, MIRI was induced in rats by left anterior descending coronary artery ligation. PHC pretreatment increased haemodynamic parameters and histopathological damage and reduced myocardial infarction size in the MIRI model. PHC pretreatment also inhibited ferroptosis, which was characterized by the decreased levels of Fe2+, 4-hydroxynonenal and ACSL4, and increased levels of GPX4, GSH-Px and GST. In response to 6 h of oxygen-glucose deprivation and 18 h of reoxygenation, PHC pretreatment had the same effects on these factors in H9c2 cells and reduced lipid ROS levels. Furthermore, ACSL4 overexpression reversed the protective effects of PHC on H9c2 cells. These results indicated that PHC inhibited MIRI through ACSL4-mediated ferroptosis. This study demonstrated that PHC could inhibit ferroptosis in MIRI and the relationship among PHC, ACSL4, ferroptosis and MIRI. This study demonstrated the inhibitory effect of PHC on ferroptosis and showed that PHC affects MIRI through ACSL4-mediated ferroptosis in vivo and in vitro.


Assuntos
Ferroptose , Traumatismo por Reperfusão Miocárdica , Ratos , Animais , Traumatismo por Reperfusão Miocárdica/patologia , Cardiotônicos , Quinuclidinas/farmacologia , Quinuclidinas/uso terapêutico
12.
Neurotox Res ; 40(6): 1895-1901, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36223054

RESUMO

This study attempted to explore whether miR-363-3p play a role in the isoflurane (ISO)-mediated protective effect of cardiomyocyte injury induced by hypoxia/reoxygenation (H/R). A myocardial cell injury model was established, and the different preconditioning ISO concentrations were screened and determined. The miR-363-3p level was detected by RT-qPCR. The effects of miR-363-3p on proliferation and apoptosis of H9c2 cells were detected by CCK-8 assay and flow cytometry. Myocardial injury indexes were determined by enzyme-linked immunosorbent assay (ELISA). The interaction of miR-363-3p with the 3'-UTR of the KLF2 gene was confirmed by luciferase reporter gene assay. ISO pretreatment can reduce the up-regulation of miR-363-3p after H/R injury. ISO pretreatment reduces the inhibition of cell viability and the promotion of cell apoptosis induced by H/R stimuli, while the overexpression of miR-363-3p counteracts the protective effect of ISO pretreatment. Meanwhile, ISO pretreatment also reduced the level of markers of H/R-induced myocardial injury. Moreover, luciferase reporter analysis showed that KLF2 was the downstream target gene of miR-363-3p. ISO pretreatment may alleviate H/R-induced cardiomyocyte injury by regulating miR-363-3p.


Assuntos
Isoflurano , MicroRNAs , Humanos , Miócitos Cardíacos , Isoflurano/farmacologia , MicroRNAs/genética , MicroRNAs/metabolismo , Hipóxia Celular , Hipóxia/metabolismo , Apoptose
13.
Biofactors ; 46(1): 83-93, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31618500

RESUMO

MicroRNAs have been reported to be implicated in myocardial ischemia/reperfusion (I/R) injury. The purpose of this study was to investigate the effect of miR-183-5p on I/R injury. Overexpression of miR-183-5p by agomiR transfection alleviated cardiac dysfunction and significantly reduced the infarct size in rats with myocardial I/R. MiR-183-5p also alleviated myocardial apoptosis with reduced apoptotic cells and lower levels of apoptosis associated proteins. in vitro experiments were conducted on rat H9c2 cells treated with anoxia/reoxygenation (A/R). Annexin V/propidium iodide (PI) staining and flow cytometry reported that the ratio of apoptotic cells decreased by miR-183-5p transfection before A/R treatment. Moreover, according to binding sequence prediction and Dual luciferase reporter assay, we explored that voltage-dependent anion channel 1 (VDAC1), which aggravates myocardial injury and apoptosis reported in our former research, was a target of miR-183-5p. In conclusion, miR-183-5p can efficiently attenuate I/R injury and miR-183-5p may exert its effect through repressing VDAC1 expression.


Assuntos
MicroRNAs/metabolismo , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Substâncias Protetoras/metabolismo , Canal de Ânion 1 Dependente de Voltagem/antagonistas & inibidores , Animais , Apoptose/genética , Modelos Animais de Doenças , Masculino , MicroRNAs/genética , Traumatismo por Reperfusão Miocárdica/genética , Traumatismo por Reperfusão Miocárdica/metabolismo , Ratos , Ratos Wistar , Canal de Ânion 1 Dependente de Voltagem/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA