Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros

Base de datos
Tipo de estudio
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Clin Res Hepatol Gastroenterol ; : 102422, 2024 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-39025461

RESUMEN

BACKGROUND: Hepatic ischemia-reperfusion injury (HIRI) is a major cause of liver dysfunction after clinical liver surgery, which seriously affects the prognosis of patients. Remifentanil (RE) has been verified to attenuate HIRI. However, its therapeutic mechanism is still unclear. This study aimed to explore the protective mechanism of RE against HIRI. METHODS: A mouse HIRI model and an in vitro model of hypoxia/reoxygenation (H/R)-stimulated AML12 hepatocytes were established. Liver histopathological changes were evaluated by hematoxylin and eosin (HE) staining. Oxidative stress damage was assessed by malondialdehyde (MDA), superoxide dismutase (SOD), and reactive oxygen species (ROS) levels. Liver function was determined by serum alanine aminotransferase (ALT), aspartate aminotransferase (AST), lactate dehydrogenase (LDH). and adenosine triphosphate (ATP) levels. Cell counting kit-8 (CCK-8) assessed cell viability. Apoptosis was measured by terminal-deoxynucleoitidyl transferase mediated nick end labeling (TUNEL) and flow cytometry. The levels of inflammatory factors were detected by enzyme-linked immunosorbent assay (ELISA) kits. The differentially expressed genes were evaluated by mRNA microarray analysis. Western blotting and real-time quantitative polymerase chain reaction (RT-qPCR) were conducted to detect molecule expression. The binding of BTB and CNC homology 1 (BACH1) to peroxiredoxin 1 (PRDX1) was validated by chromatin immunoprecipitation (ChIP) and dual luciferase reporter assay. RESULTS: RE treatment improved liver function, and repressed oxidative stress damage and apoptosis in HIRI mice. Nine differentially expressed genes in the liver tissues of HIRI mice were selected by microarray analysis, among which BACH1 was down-regulated and PRDX1 was up-regulated after RE treatment. In addition, BACH1 directly bound to the promoter region of PRDX1 to inhibit its transcription and expression, which led to oxidative stress injury. BACH1 overexpression or PRDX1 silencing could counteract the beneficial effects of RE against HIRI. CONCLUSION: RE suppressed oxidative stress injury and inflammation via inactivation of the BACH1/PRDX1 axis, thereby ameliorating HIRI. Our findings enrich the understanding of the protective mechanisms of RE against HIRI, and provide novel evidence for its clinical application.

2.
ESC Heart Fail ; 11(2): 1205-1217, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38288506

RESUMEN

AIMS: Acute myocardial infarction (MI) is a significant contributor to death in individuals diagnosed with coronary heart disease on a worldwide level. The specific mechanism by which circRbms1 contributes to the damage caused by myocardial ischaemia-reperfusion (I/R) is not well understood. The primary aim of this study was to examine the role of circRbms1 and its associated mechanisms in the setting of I/R injury. METHODS AND RESULTS: An in vivo MI mice model and an in vitro MI cell model was established. The expression levels were detected using quantitative real-time PCR (qRT-PCR) and western blot. Cellular proliferation, apoptosis, pyroptosis, and autophagy were detected by immunostaining, immunohistochemistry, western blot, and transmission electron microscopy (TEM). Dual-luciferase reporter assay, RNA pull-down assay, and RIP assay were performed to validate the molecular interactions. CircRbms1 was up-regulated in A/R-induced HCMs and acted as a sponge for miR-142-3p, thereby targeting MST1. CircRbms1 could improve stability of MST1 by recruiting IGF2BP2 (all P < 0.05). CircRbms1 knockout reduced cell pyroptosis, improved autophagy and proliferation level in A/R-induced HCMs (all P < 0.05). CircRbms1 knockout alleviated cardiac dysfunction and cell pyroptosis and enhanced autophagy and proliferation in mice through the miR-142-3p/MST1 axis. CONCLUSIONS: CircRbms1 inhibited the miR-142-3p/MST1 axis and played a protective role in myocardial I/R injury. It may provide a new therapeutic target for I/R heart injury.


Asunto(s)
MicroARNs , Isquemia Miocárdica , Daño por Reperfusión Miocárdica , Animales , Ratones , Autofagia/genética , MicroARNs/genética , Daño por Reperfusión Miocárdica/genética , ARN Mensajero
3.
Am J Med Sci ; 367(1): 49-60, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37939881

RESUMEN

BACKGROUND: Myocardial ischemia-reperfusion (I/R), a harmful process in the treatment of cardiovascular diseases, can cause secondary damage to the cardiac tissues. Circular RNAs (circRNAs) are important regulators in a number of cardiac disorders. However, the role of circHDAC9 in myocardial I/R injury has not been clarified. METHODS: Human cardiac myocytes (HCMs) were treated with hypoxia/reoxygenation (H/R) and mice were subjected to I/R. Quantitative reverse transcriptase-polymerase chain reaction (RT-qPCR) was used to analyze the expression of circHDAC9, miR-671-5p, and SOX4, and western blot was used to detect SOX4 protein. The binding relationship among circHDAC9, miR-671-5p, and SOX4 was confirmed by RNA pull-down, luciferase, and RNA immunoprecipitation (RIP) assays. The effects of circHDAC9/miR-671-5p/SOX4 axis on the apoptosis, oxidative stress and inflammation were evaluated in both myocardial I/R injury models. RESULTS: The expression of circHDAC9 and SOX4 was noticeably elevated, whereas miR-671-5p expression was downregulated in both myocardial I/R injury models. circHDAC9 knockdown significantly reduced the apoptosis, activities of caspase-3 and caspase-9, ROS intensity, MDA activity, and concentrations of TNF-α, IL-1ß, and IL-6, but increased the viability and SOD activity in H/R-treated HCMs. Suppression of circHDAC9 dramatically reduced the levels of circHDAC9 and SOX4, while enhanced miR-671-5p expression in H/R-treated HCMs. CircHDAC9 functioned via sponging miR-671-5p to regulate SOX4 expression in vitro. Additionally, silencing of circHDAC9 improved the pathological abnormalities and cardiac dysfunction, and reduced the apoptosis, oxidative stress and inflammation in mice with myocardial I/R injury. CONCLUSIONS: Inhibition of circHDAC9 significantly improved myocardial I/R injury by regulating miR-671-5p/SOX4 signaling pathway.


Asunto(s)
MicroARNs , Daño por Reperfusión Miocárdica , ARN Circular , Animales , Humanos , Ratones , Apoptosis , Inflamación/patología , MicroARNs/genética , MicroARNs/metabolismo , Daño por Reperfusión Miocárdica/genética , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Miocitos Cardíacos/patología , Transducción de Señal , Factores de Transcripción SOXC/genética , Factores de Transcripción SOXC/metabolismo , Factores de Transcripción SOXC/farmacología , ARN Circular/metabolismo
4.
Exp Cell Res ; 422(2): 113417, 2023 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-36379276

RESUMEN

BACKGROUND: Diabetes cardiomyopathy (DCM) is one of the major risk factors for the heart failure of the diabetic patients. RIPK1 maybe participate in the regulation of the oxidative stress and inflammation during DCM. METHODS: H&E and Masson staining were utilized to assess the inflammation and fibrosis in myocardial tissues. CCK-8 and TUNEL staining were utilized to analyze cell viability and apoptosis, respectively. SOD activity and MDA content were detected utilizing the kits. The formation of autophagosomes was measured by immunofluorescence assay. RESULTS: RIPK1 and RPTOR (a component of mTORC1) expression and oxidative stress level were upregulated, but autophagy was decreased in the myocardial tissues of DCM rat characterized by the high body weight and blood glucose, abnormal cardiac function, myocardial inflammation and fibrosis. High glucose (HG) treatment resulted in cell viability and autophagy level decreasing, inflammatory cytokines expression increasing and oxidative stress increasing in cardiac fibroblasts (CFs). Meanwhile, RIPK1 and RPTOR expression also was increased in HG-treated cells. HG-induced CFs apoptosis, inflammation, oxidative stress and the inhibition of HG to cell viability and autophagy was partly reversed by the inhibitor of RIPK1 and mTORC1. CONCLUSION: Overall, RIPK1/mTORC1 signalling suppression improved HG-induced apoptosis, inflammation and oxidative stress through activation autophagy. These data provided a reliable evidence that RIPK1 may be a potential target for DCM therapeutic.


Asunto(s)
Diabetes Mellitus Experimental , Cardiomiopatías Diabéticas , Ratas , Animales , Cardiomiopatías Diabéticas/tratamiento farmacológico , Diabetes Mellitus Experimental/metabolismo , Estrés Oxidativo/fisiología , Apoptosis , Inflamación/genética , Inflamación/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Proteína Reguladora Asociada a mTOR , Fibrosis
5.
Bioengineered ; 13(5): 12726-12737, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35611768

RESUMEN

Evidence suggests circRBMS1 regulates mRNA to mediate cell apoptosis, inflammation, and oxidative stress in different diseases. MST1 is reported to be the target and activator of apoptosis-related molecules and signaling pathways. Hence, the present study aims to investigate the role of circ-RBMS1/miR-2355-3p/MST1 in the development of I/R injury. In vitro experiments showed increased circ-RBMS1 and decreased miR-2355-3p in H/R-induced HCMs. CircRBMS1 served as a sponge for miR-2355-3p and miR-2355-3p targeted MST1. Furthermore, knockout of circRBMS1 attenuated cell apoptosis, oxidized stress, and inflammation in H/R-induced HCMs. In vivo experiments indicated circRBMS1 knockdown attenuated cardiac function damage, cell apoptosis, oxidative stress injury and inflammatory response through miR-2355-3p/MST1 axis in mice. In summary, these results demonstrated circRBMS1 played a protective role in myocardial I/R injury though inhibition of miR-2355-3p/MST1 axis. It might provide a new therapeutic target for cardiac I/R injury.


Asunto(s)
MicroARNs , Daño por Reperfusión Miocárdica , Proteínas Serina-Treonina Quinasas , ARN Circular , Animales , Apoptosis/genética , Inflamación/metabolismo , Ratones , Ratones Noqueados , MicroARNs/genética , MicroARNs/metabolismo , Daño por Reperfusión Miocárdica/genética , Daño por Reperfusión Miocárdica/metabolismo , Miocitos Cardíacos/metabolismo , Proteínas Serina-Treonina Quinasas/genética , ARN Circular/genética
6.
Lab Invest ; 102(5): 505-514, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35066566

RESUMEN

LncRNAs and miRNAs are correlated with the pathogenesis of myocardial ischemia-reperfusion injury (MIRI). Whether lncRNA ROR or miR-185-5p plays a crucial role in MIRI is still unclear. In in-vitro, human cardiac myocytes (HCMs) were treated with hypoxia/reoxygenation (H/R). Wistar rats were used to set up an in-vitro I/R model by means of recanalization after ligation. Evaluation of the myocardial injury marker lactate dehydrogenase (LDH) in HCMs cells was performed. The expression of miR-185-5p and ROR, IL-1ß, and IL-18 were detected by qRT-PCR. ELISA was also performed to evaluate the secretion of IL-1ß and IL-18. Western blotting was carried out to determine CDK6, NLRP3, GSDMD-N, ASC, and cleaved-caspase1 protein expression. The relationship between miR-185-5p and CDK6 or ROR was confirmed by a dual-luciferase reporter assay. Our findings revealed that H/R treated HCMs showed a significantly decreased miR-185-5p expression and increased expression of CDK6 and ROR. ROR knockdown reduced H/R induced pyroptosis and inflammation, while knockdown of miR-185-5p accelerated the effect. Furthermore, miR-185-5p was negatively regulated and absorbed by ROR in HCMs. Overexpression of miR-185-5p reversed the H/R-induced cell pyroptosis and upregulation of LDH, IL-1ß, and IL-18. In HCMs, miR-185-5p was also negatively regulated and related to CDK6 expression. Moreover, overexpression of CDK6 significantly inhibited the effects of miR-185-5p mimics on the inflammatory response and pyroptosis of HCMs. Knockdown of ROR alleviated H/R-induced myocardial injury by elevating miR-185-5p and inhibiting CDK6 expression. Taken together, our results show that the ROR/miR-185-5p/CDK6 axis modulates cell pyroptosis induced by H/R and the inflammatory response of HCMs.


Asunto(s)
MicroARNs , Daño por Reperfusión Miocárdica , ARN Largo no Codificante , Animales , Hipoxia , Interleucina-18 , MicroARNs/genética , MicroARNs/metabolismo , Daño por Reperfusión Miocárdica/genética , ARN Largo no Codificante/genética , Ratas , Ratas Wistar
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA