Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Apoptosis ; 27(3-4): 206-221, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35084609

RESUMEN

This study investigated how miR-136-5p partially affected cardiomyocyte pyroptosis in rats with coronary microembolization (CME). The cardiac function and structure of rats with CME were evaluated using echocardiography, hematoxylin and eosin staining, Masson staining, and troponin I level. Pyroptosis was induced by lipopolysaccharide (LPS) in isolated rat cardiomyocytes and evaluated by the expression of caspase-1, NOD-like receptor family pyrin domain-containing 3, interleukin-1ß, and gasdermin D-N. After cell transfection, the expression of Ataxin-1 like (ATXN1L), pyrin domain-containing 1 (PYDC1), and pyroptosis-related proteins was assessed. Dual-luciferase reporter and immunoprecipitation assays were used to verify the relationships among miR-136-5p, ATXN1L, and capicua (CIC). MiR-136-5p was under-expressed, whereas ATXN1L was overexpressed in rats with CME and in LPS-treated primary cardiomyocytes. MiR-136-5p targeted ATXN1L, and ATXN1L bound to CIC to suppress PYDC1 expression. MiR-136-5p overexpression suppressed pyroptosis by inhibiting the binding of ATXN1L with CIC and promoting PYDC1 expression, which was reversed by simultaneous elevation of ATXN1L. In conclusion, miR-136-5p suppressed pyroptosis by upregulating PYDC1 via ATXN1L/CIC axis, thereby attenuating cardiac damage caused by CME.


Asunto(s)
MicroARNs , Piroptosis , Animales , Apoptosis , Lipopolisacáridos , MicroARNs/metabolismo , Miocitos Cardíacos/metabolismo , Piroptosis/genética , Ratas
2.
J Cell Mol Med ; 24(19): 11500-11511, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32860492

RESUMEN

MiRNAs can be used as promising diagnostic biomarkers of heart failure, while lncRNAs act as competing endogenous RNAs of miRNAs. In this study, we collected peripheral blood monocytes from subjects with or without HF to explore the association between certain lncRNAs, miRNAs and HF. Heart failure patients with preserved or reduced ejection fraction were recruited for investigation. ROC analysis was carried out to evaluate the diagnostic values of certain miRNAs and lncRNAs in HF. Luciferase assays were used to study the regulatory relationship between above miRNAs and lncRNAs. LncRNA overexpression was used to explore the effect of certain miRNAs in H9C2 cells. Expression of miR-30c was significantly decreased in the plasma and peripheral blood monocytes of patients suffering from heart failure, especially in these with reduced ejection fraction. On the contrary, the expression of lncRNA-CASC7 was remarkably increased in the plasma and peripheral blood monocytes of patients suffering from heart failure. Both miR-30c and lncRNA-CASC7 expression showed a promising efficiency as diagnostic biomarkers of heart failure. Luciferase assays indicated that miR-30c played an inhibitory role in lncRNA-CASC7 and IL-11 mRNA expression. Moreover, the overexpression of lncRNA-CASC7 suppressed the expression of miR-30c while evidently increasing the expression of IL-11 mRNA and protein in H9C2 cells. This study clarified the relationship among miR-30c, lncRNA-CASC7 and IL-11 expression and the risk of heart failure and showed that lncRNA-CASC7 is potentially involved in the pathogenesis of HF via modulating the expression of miR-30c.


Asunto(s)
Regulación de la Expresión Génica , Insuficiencia Cardíaca/genética , MicroARNs/genética , ARN Largo no Codificante/metabolismo , Anciano , Animales , Secuencia de Bases , Biomarcadores/sangre , Línea Celular , Regulación hacia Abajo/genética , Femenino , Insuficiencia Cardíaca/sangre , Insuficiencia Cardíaca/diagnóstico , Insuficiencia Cardíaca/patología , Humanos , Interleucina-11/metabolismo , Masculino , MicroARNs/sangre , MicroARNs/metabolismo , Monocitos/metabolismo , ARN Largo no Codificante/genética , Curva ROC , Ratas , Regulación hacia Arriba/genética
3.
Am J Physiol Heart Circ Physiol ; 318(2): H332-H344, 2020 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-31858814

RESUMEN

Increased production of reactive oxygen species (ROS) significantly contributed to the pathogenesis of acute myocardial infarction (AMI). Recent studies suggest that hypoxia upregulated the long noncoding RNA taurine upregulated gene 1 (TUG1). In this study, we explored the functional significance and molecular mechanisms of TUG1/miR-132-3p axis in ischemia-challenged cardiomyocytes. In primary cardiomyocytes challenged with H2O2, expressions of miR-132-3p, TUG1, and other target proteins were measured by RT quantitative PCR or Western blot analysis; cell viability by 3-(4,5-dimethylthiazolyl-2)-2,5-diphenyltetrazolium bromide assay; apoptosis by annexin V and propidium iodide staining; the abundance of acetylated H3K9 or histone deacetylase 3 (HDAC3) within the promoter of target genes by chromatin immunoprecipitation; the direct interaction between miR-132-3p and HDAC3 or TUG1 by luciferase reporter assay. The biological significance of miR-132-3p, TUG1, and HDAC3 was assessed using miR-132-3p mimic, siRNA-targeting TUG1 and HDAC3 inhibitor RGF966, respectively, in H2O2-challenged cells in vitro or ischemia-reperfusion (I/R)-induced AMI in vivo. miR-132-3p was downregulated, whereas TUG1 upregulated in H2O2-challenged cardiomyocytes. Overexpressing miR-132-3p or knocking down TUG1 significantly improved viability, inhibited apoptosis, and reduced ROS production in H2O2-stressed cardiomyocytes in vitro and alleviated I/R-induced AMI in vivo. Mechanistically, TUG1 sponged miR-132-3p and upregulated HDAC3, which reduced the acetylation of H3K9 and epigenetically inhibited expressions of antioxidative genes, including Bcl-xL, Prdx2, and Hsp70. The TUG1/miR-132-3p/HDAC3 axis critically regulates ROS production and the pathogenic development of AMI. Targeting TUG1, upregulating miR-132-3p, or inhibiting HDAC3 may benefit AMI treatment.NEW & NOTEWORTHY Increased production of reactive oxygen species (ROS) significantly contributed to the pathogenesis of acute myocardial infarction (AMI). Recent studies suggest that hypoxia upregulated the long noncoding RNA taurine upregulated gene 1 (TUG1). However, the underlying mechanisms remain elusive. In the present study, we reported for the first time that H2O2 or ischemia-reperfusion-induced TUG1, by sponging microRNA 132-3p, activated histone deacetylase 3, which in turn targeted multiple protective genes, stimulated intracellular ROS accumulation, and aggravated the injury of AMI. Our findings might provide some insight to seek new targets for AMI treatment.


Asunto(s)
Histona Desacetilasas/genética , MicroARNs/genética , Isquemia Miocárdica/genética , ARN Largo no Codificante/genética , Acrilamidas/farmacología , Animales , Apoptosis , Epigénesis Genética , Técnicas de Silenciamiento del Gen , Humanos , Peróxido de Hidrógeno/metabolismo , Ratones , Ratones Endogámicos C57BL , MicroARNs/biosíntesis , Fenilendiaminas/farmacología , ARN Largo no Codificante/biosíntesis , ARN Interferente Pequeño/farmacología , Especies Reactivas de Oxígeno/metabolismo
4.
J Mol Cell Cardiol ; 133: 12-25, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31145943

RESUMEN

BACKGROUND: Long non-coding RNAs (lncRNAs) play a central role in regulating heart diseases. In the present study, we examined the effects of lncRNA taurine up-regulated gene 1 (TUG1) in ischemia/reperfusion (I/R)- or hydrogen peroxide-challenged cardiomyocytes, with specific focus on autophagy-induced cell apoptosis. METHODS: The expressions of miR-142-3p and TUG1 in H2O2-challenged cardiomyocytes and I/R-injured heart tissue were measured by RT-qPCR. Cell death was measured by trypan blue staining assay. Cell apoptosis was determined by Annexin V/PI staining and TUNEL assay. Autophagy was examined by quantifying cells or tissues containing LC3+ autophagic vacuoles by immunofluorescence, or by measuring the expressions of autophagy-related biomarkers by Western blot. The direct interaction between miR-142-3p and TUG1, high mobility group box 1 protein (HMGB1), or Ras-related C3 botulinum toxin substrate 1 (Rac1) was examined using luciferase reporter assay. The significance of miR-142-3p and TUG1 on cell apoptosis or autophagy was examined using both gain-of-function and loss-of-function approaches. The importance of HMGB1 or Rac1 was assessed using siRNA-mediated gene silencing. RESULTS: miR-142-3p was down-regulated, while TUG1 up-regulated in H2O2-challenged cardiomyocytes in vitro and I/R-injured heart tissues in vivo. Functionally, inhibition of TUG1 and overexpression of miR-142-3p inhibited cell apoptosis and autophagy in cardiomyocytes. The function of TUG1 were achieved by sponging miR-142-3p and releasing the suppression of the putative targets of miR-142-3p, HMGB1 and Rac1. Both HMGB1 and Rac1 essentially mediated cell apoptosis and autophagy induced by TUG1. CONCLUSIONS: TUG1, by targeting miR-142-3p and up-regulating HMGB1 and Rac1, plays a central role in stimulating autophagic cell apoptosis in ischemia/hypoxia-challenged cardiomyocytes. Down-regulating TUG1 or up-regulating miR-142-3p may ameliorate myocardial injury and protect against acute myocardial infarction.


Asunto(s)
Autofagia/genética , Proteína HMGB1/genética , MicroARNs/genética , Daño por Reperfusión Miocárdica/genética , ARN Largo no Codificante/genética , Proteína de Unión al GTP rac1/genética , Regiones no Traducidas 3' , Animales , Línea Celular Tumoral , Proliferación Celular/genética , Modelos Animales de Enfermedad , Técnicas de Silenciamiento del Gen , Silenciador del Gen , Humanos , Hipoxia/genética , Ratones , Daño por Reperfusión Miocárdica/patología , Miocitos Cardíacos/metabolismo
5.
J Cell Biochem ; 120(6): 10756-10766, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30652345

RESUMEN

Coronary microembolization (CME) occurs when atherosclerotic plaque debris is detached during the treatment of acute coronary syndrome with Percutaneous Coronary Intervention (PCI). The complications of distal microvascular embolism, including local myocardial inflammation, are the main causes of myocardial damage and are a strong predictor of poor long-term prognosis and major cardiac adverse events. microRNAs (miRNAs) are involved in the pathophysiological processes of cardiovascular inflammatory diseases. Dysregulation of microRNA (miR)-26a-5p, in particular, is associated with a variety of cardiovascular diseases. However, the role of miR-26a-5p in CME-induced myocardial injury is unclear. In this study, we developed an animal model of CME by injecting microembolic balls into the left ventricle of rats and found that miR-26a-5p expression decreased in myocardial tissue in response. Using a miR-26a-5p mimic, echocardiography, hematoxylin-eosin staining, and Western blot analysis we found that the diminished cardiac function and myocardial inflammation induced by CME is alleviated by miR-26a-5p overexpression. Furthermore, our results show that inhibitors of miR-26a-5p have the opposite effect. In addition, in vitro experiments using real-time PCR, Western blot analysis, and a dual luciferase reporter gene show that HMGA1 is a target gene of miR-26a-5p. Thus, overexpression of miR-26a-5p could be a novel therapy to improve CME-induced myocardial damage.


Asunto(s)
Vasos Coronarios/metabolismo , Embolia/genética , Proteína HMGA1a/genética , MicroARNs/genética , Miocardio/metabolismo , Animales , Antagomirs/genética , Antagomirs/metabolismo , Gasto Cardíaco , Vasos Coronarios/diagnóstico por imagen , Vasos Coronarios/patología , Modelos Animales de Enfermedad , Ecocardiografía , Embolia/diagnóstico por imagen , Embolia/etiología , Embolia/metabolismo , Femenino , Regulación de la Expresión Génica , Proteína HMGA1a/metabolismo , Masculino , MicroARNs/agonistas , MicroARNs/antagonistas & inhibidores , MicroARNs/metabolismo , Miocardio/patología , Oligorribonucleótidos/genética , Oligorribonucleótidos/metabolismo , Intervención Coronaria Percutánea/efectos adversos , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Volumen Sistólico , Troponina I/genética , Troponina I/metabolismo , Función Ventricular Izquierda
6.
Cytokine ; 111: 475-480, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29914794

RESUMEN

Oxidized low-density lipoprotein (ox-LDL) is a major risk factor for atherosclerosis and often causes injury to vascular endothelial cells. We found that pinocembrin, a natural antioxidant found in honey and certain herbs, protects human aortic endothelial cells (HAECs) from ox-LDL-induced injury. Pinocembrin suppresses the expression of pro-inflammatory vascular adhesion molecules (VCAM-1, ICAM-1 and E-selectin) and cytokines (TNF-α, IL-1ß, and IL-8), as well as ROS production induced by ox-LDL. Pinocembrin potently inhibits the attachment of monocytes to HAEC cells. Mechanistically, pinocembrin suppresses activation of the MAPK kinase p38 and NF-κB pathways in the context of ox-LDL. Our data indicate that pinocembrin is a promising versatile natural compound that can protect endothelial cells from injury.


Asunto(s)
Células Endoteliales/efectos de los fármacos , Flavanonas/farmacología , Lipoproteínas LDL/efectos adversos , Sustancias Protectoras/farmacología , Antioxidantes/farmacología , Aterosclerosis/tratamiento farmacológico , Aterosclerosis/metabolismo , Moléculas de Adhesión Celular/metabolismo , Células Cultivadas , Citocinas/metabolismo , Células Endoteliales/metabolismo , Humanos , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Monocitos/efectos de los fármacos , Monocitos/metabolismo , FN-kappa B/metabolismo , Especies Reactivas de Oxígeno , Transducción de Señal/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
7.
Mol Ther Nucleic Acids ; 35(1): 102106, 2024 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-38222298

RESUMEN

[This retracts the article DOI: 10.1016/j.omtn.2021.01.034.].

8.
Hypertens Res ; 46(6): 1493-1503, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36854725

RESUMEN

Resveratrol (RES) has been demonstrated to be protective in the cardiovascular system in animal studies, but the evidence is limited in humans. The purpose of the study was to evaluate the effect of RES supplementation on cardiac remodeling in patients with hypertension. Eighty Subjects were randomly divided into RES group (plus RES 400 mg/d in addition to conventional therapy, n = 43) and control group (conventional therapy, n = 37). The main outcomes of the study were changes within cardiac-remodeling parameters. Secondary outcomes were changes in anthropometric parameters, arterial stiffness parameters and mechanism indices. There was no statistically significant difference between the RES group and control group in terms of baseline characteristics. After 6 months, the RES group had smaller left atrial, lower E/e', higher left ventricular global longitudinal strain and lower biomarkers indicating cardiac fibrosis (expressed by decreases in procollagen type I C-peptide and galectin-3) compared to the control group. However, there was no significant difference in left ventricular structure between the two groups. Although the RES group showed a significant decrease in brachial-ankle pulse wave velocity compared to the pre-intervention value, the difference between the RES and the control groups was not obvious. What's more, compared with the control group, the serum levels of sirtuin3, superoxide dismutase and klotho were significantly increased in the RES group. In conclusion, RES supplementation can alleviate left atrial remodeling, improve left ventricular diastolic function and may alleviate cardiac fibrosis in hypertensive patients, and could be used as an adjunct to conventional therapies of hypertensive heart disease.


Asunto(s)
Hipertensión , Remodelación Ventricular , Humanos , Índice Tobillo Braquial , Suplementos Dietéticos , Fibrosis , Análisis de la Onda del Pulso , Resveratrol/farmacología , Función Ventricular Izquierda
10.
Mol Ther Nucleic Acids ; 23: 1258-1271, 2021 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-33717647

RESUMEN

Increasing evidence suggests that mitochondrial microRNAs (miRNAs) are implicated in the pathogenesis of cardiovascular diseases; however, their roles in ischemic heart disease remain unclear. Herein, we demonstrate that miR-146a is enriched in the mitochondrial fraction of cardiomyocytes, and its level significantly decreases after ischemic reperfusion (I/R) challenge. Cardiomyocyte-specific knockout of miR-146a aggravated myocardial infarction, apoptosis, and cardiac dysfunction induced by the I/R injury. Overexpression of miR-146a suppressed anoxia/reoxygenation-induced cardiomyocyte apoptosis by inhibiting the mitochondria-dependent apoptotic pathway and increasing the Bcl-2/Bax ratio. miR-146a overexpression also blocked mitochondrial permeability transition pore opening and attenuated the loss of mitochondrial membrane potential and cytochrome c leakage; meanwhile, miR-146a knockdown elicited the opposite effects. Additionally, miR-146a overexpression decreased cyclophilin D protein, not mRNA, expression. The luciferase reporter assay revealed that miR-146a binds to the coding sequence of the cyclophilin D gene. Restoration of cyclophilin D reversed the inhibitory action of miR-146a on cardiomyocyte apoptosis. Furthermore, cardiomyocyte-specific cyclophilin D deletion completely abolished the exacerbation of myocardial infarction and apoptosis observed in miR-146a cardiomyocyte-deficient mice. Collectively, these findings demonstrate that nuclear miR-146a translocates into the mitochondria and regulates mitochondrial function and cardiomyocyte apoptosis. Our study unveils a novel role for miR-146a in ischemic heart disease.

11.
Mol Ther Nucleic Acids ; 23: 1304-1322, 2021 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-33717651

RESUMEN

In this study, we aim to investigate the regulation of specific long non-coding RNAs (lncRNAs) on the progression of ischemia/reperfusion (I/R) injury. We identified and characterized the exosomes derived from mouse primary aortic endothelial cells. Subsequently, we found that these exosomes expressed typical exosomal markers and high levels of LINC00174, which significantly ameliorated I/R-induced myocardial damage and suppressed the apoptosis, vacuolation, and autophagy of myocardial cells. Mechanistic approaches revealed that LINC00174 directly interacted with SRSF1 to suppress the expression of p53, thus restraining the transcription of myocardin and repressing the activation of the Akt/AMPK pathway that was crucial for autophagy initiation in I/R-induced myocardial damage. Moreover, this molecular mechanism was verified by in vivo study. In summary, exosomal LINC00174 generated from vascular endothelial cells repressed p53-mediated autophagy and apoptosis to mitigate I/R-induced myocardial damage, suggesting that targeting LINC00174 may be a novel strategy to treat I/R-induced myocardial infarction.

12.
Drug Des Devel Ther ; 14: 4407-4422, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33122886

RESUMEN

OBJECTIVE: Coronary microembolization (CME) results in progressive contractile dysfunction associated with cardiomyocyte apoptosis. Alprostadil injection improves microcirculation, which is effective in treating various cardiovascular disorders. However, the therapeutic effects of alprostadil in CME-induced myocardia injury remain unknown. Therefore, we evaluated the effects of alprostadil injection on cardiac protection in a rat model of CME and explored the underlying mechanisms. METHODS: A rat model of CME was established by injecting polyethylene microspheres into the left ventricle. After injection of microspheres, rats in the alprostadil group received alprostadil via tail vein within 2 minutes. Cardiac function, histological alterations in myocardium, serum c-troponin I (cTnI) levels, myocardium adenosine triphosphate (ATP) concentrations, the activity of superoxide dismutase (SOD) and malondialdehyde (MDA) content in myocardium, and myocardial apoptosis-related proteins were detected 12 hours after CME modeling. RESULTS: Compared with the Sham group, ATP concentrations, SOD activity in the myocardium, and cardiac function were significantly decreased in a rat model of CME. In addition, serum cTnI levels, MDA content, expression levels of pro-apoptotic proteins, and the number of TUNEL-positive nuclei were remarkably higher in CME group than those in the Sham group. However, alprostadil treatment notably reduced serum cTnI levels and expression levels of pro-apoptotic proteins, while noticeably improved cardiac function, and accelerated SOD activity in the myocardium following CME. Additionally, it was unveiled that the protective effects of alprostadil injection inhibit CME-induced myocardial apoptosis in the myocardium potentially through regulation of the GSK-3ß/Nrf2/HO-1 signaling pathway. CONCLUSION: Alprostadil injection seems to significantly suppress oxidative stress, alleviate myocardial apoptosis in the myocardium, and improve cardiac systolic and diastolic functions following CME by regulating the GSK-3ß/Nrf2/HO-1 signaling pathway.


Asunto(s)
Alprostadil/farmacología , Apoptosis/efectos de los fármacos , Infarto del Miocardio/tratamiento farmacológico , Miocitos Cardíacos/efectos de los fármacos , Alprostadil/administración & dosificación , Animales , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Glucógeno Sintasa Quinasa 3 beta/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Hemo Oxigenasa (Desciclizante)/antagonistas & inhibidores , Hemo Oxigenasa (Desciclizante)/metabolismo , Masculino , Estructura Molecular , Infarto del Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Factor 2 Relacionado con NF-E2/antagonistas & inhibidores , Factor 2 Relacionado con NF-E2/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Relación Estructura-Actividad
13.
Cell Death Dis ; 10(2): 61, 2019 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-30683933

RESUMEN

Coronary microembolization (CME) is a common complication seen during primary percutaneous coronary intervention (pPCI). CME-induced myocardiac inflammation is the primary cause of myocardiac injury. Dysregulated miR-142-3p has been implicated in multiple cardiovascular diseases and is significantly downregulated in CME-induced myocardial injury. However, the role of miR-142-3p in CME-induced myocardial injury is unclear. This study herein built a porcine CME model by infusing microembolization spheres into the left anterior descending branch via a microcatheter, and detected the downregulation of miR-142-3p in the myocardial tissues of CME pigs. Echocardiography, hematoxylin basic fuchsin picric acid (HBFP) staining, and western blotting of NF-κB p65, TNF-α, IL-1ß, and IL-6 showed that the pharmacological overexpression of miR-142-3p using agomiR has improved cardiac function and attenuated CME-induced myocardiac inflammatory response, while its inhibition using antagomiR demonstrated inverse effects. Moreover, in vitro experiments demonstrated IRAK-1 as a direct target gene of miR-142-3p. Luciferase reporter assays, quantitative real-time polymerase chain reaction and western blotting demonstrated its effects in controlling the inflammation of cardiomyocytes. It is noteworthy that miR-142-3p was found to be decreased in the plasma of STEMI patients undergoing pPCI with no-reflow, indicating a potential clinical relevance of miR-142-3p. The receiver-operator characteristic curve indicated that plasma miR-142-3p might be an independent predictor of no-reflow during pPCI in patients with STEMI. Therefore, overexpression of miR-142-3p acts as a novel therapy for CME-induced myocardial injury.


Asunto(s)
Embolia/complicaciones , Quinasas Asociadas a Receptores de Interleucina-1/genética , MicroARNs/sangre , MicroARNs/metabolismo , Miocarditis/sangre , Miocarditis/etiología , Animales , Citocinas/metabolismo , Embolia/etiología , Femenino , Células HEK293 , Humanos , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , Lipopolisacáridos/farmacología , Masculino , MicroARNs/genética , Infarto del Miocardio/cirugía , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Intervención Coronaria Percutánea/efectos adversos , Ratas , Porcinos , Factor de Transcripción ReIA/metabolismo , Transfección
14.
Mol Ther Nucleic Acids ; 18: 110-122, 2019 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-31541797

RESUMEN

Myocardial infarction (MI) is a life-threatening cardiac event that results in extreme damage to the heart muscle. The Wnt signaling pathway has been implicated in the development of heart diseases. Hence, the current study aimed to investigate the role of microRNA (miRNA) in association with the Wnt signaling pathway to identify potential candidates for MI therapy. Differentially expressed miRNAs associated with MI occurrence were screened, and miR-494 was selected for subsequent experiments. Sprague-Dawley rats were included to establish a MI model via intraperitoneal injection of 0.1 mg/kg atropine sulfate and 40 mg/kg pentobarbital sodium. Then, the interaction between miR-494 and LRG1 was identified. The effect of miR-494 on expression of the Wnt signaling pathway-related genes, proliferation, migration, and invasion ability of fibroblasts and vascular endothelial cells (VECs) was subsequently evaluated through a series of gain- and loss-of-function experiments. The results revealed that miR-494 was poorly expressed and LRG1 was highly expressed in MI rats. miR-494 targets and downregulates LRG1, which resulted in the inactivation of the Wnt signaling pathway and promoted proliferation, migration, and invasion ability of fibroblasts and VECs. In conclusion, this study provided evidence suggesting that overexpressed miR-494 could potentially promote the proliferation, migration, and invasion of fibroblasts and VECs in MI through the inactivation of the Wnt signaling pathway by binding to LRG1.

15.
Biomed Pharmacother ; 106: 776-784, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29990871

RESUMEN

Coronary microembolization (CME) is a common complication during the treatment of acute coronary syndrome (ACS) and percutaneous coronary intervention (PCI). Nicorandil can be used to prevent myocardial injury after PCI to reduce the incidence of coronary no-reflow and slow flow, and play a role in myocardial protection, suggesting that its mechanism may be related to the inhibition of CME-induced inflammation of cardiomyocytes. However, the specific mechanism remains unclear. This study investigated the myocardial protective effects of nicorandil pretreatment on CME-induced myocardial injury and the specific mechanism of its inhibition of myocardial inflammation. An CME rat model exhibited CME-induced myocardial inflammation and the elevation of serum tumor necrosis factor-alpha (TNF-α) and interleukin (IL)-1ß based on echocardiography, myocardial enzyme detection, hematoxylin and eosin (HE) and hematoxylin-basic fuchsin-picric acid (HBFP) stainings, ELISA, quantitative real-time PCR, and western blotting. Nicorandil treatment seven days before CME induction effectively inhibited myocardial inflammation, ameliorated myocardial injury, and improved cardiac function, mainly by inhibiting Toll-like receptor 4 (TLR4)-mediated myeloid differentiation primary response protein 88 (MyD88)-dependent nuclear factor-kappa B (NF-κB) signaling. Rat neonatal cardiomyocyte experiments further confirmed that nicorandil ameliorated lipopolysaccharide (LPS)-induced myocardial inflammation and improved cardiomyocyte survival. The specific mechanisms mainly involved the inhibition of TLR4/MyD88/NF-κB signaling and the reduction of the inflammatory cytokines TNF-α and IL-1ß released from cardiomyocytes. In summary, nicorandil significantly protected cardiomyocytes from CME-induced myocardial injury mainly by inhibiting TLR4/MyD88/NF-κB signaling, thereby reducing the onset of CME-induced myocardial inflammation. This could be one of the important mechanisms for reducing postoperative myocardial injury via PCI-preoperative prophylactic treatment with nicorandil.


Asunto(s)
Antiinflamatorios/farmacología , Estenosis Coronaria/tratamiento farmacológico , Embolia/tratamiento farmacológico , Factor 88 de Diferenciación Mieloide/metabolismo , Infarto del Miocardio/prevención & control , Miocarditis/prevención & control , Miocitos Cardíacos/efectos de los fármacos , FN-kappa B/metabolismo , Nicorandil/farmacología , Receptor Toll-Like 4/metabolismo , Disfunción Ventricular Izquierda/prevención & control , Animales , Supervivencia Celular , Estenosis Coronaria/etiología , Estenosis Coronaria/metabolismo , Estenosis Coronaria/patología , Citoprotección , Modelos Animales de Enfermedad , Embolia/etiología , Embolia/metabolismo , Embolia/patología , Mediadores de Inflamación/sangre , Interleucina-1beta/sangre , Lipopolisacáridos/farmacología , Masculino , Microesferas , Infarto del Miocardio/etiología , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Miocarditis/etiología , Miocarditis/metabolismo , Miocarditis/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Ratas Sprague-Dawley , Transducción de Señal , Volumen Sistólico/efectos de los fármacos , Factor de Necrosis Tumoral alfa/sangre , Disfunción Ventricular Izquierda/etiología , Disfunción Ventricular Izquierda/metabolismo , Disfunción Ventricular Izquierda/fisiopatología , Función Ventricular Izquierda/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda