Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Publication year range
1.
Hamostaseologie ; 43(4): 271-280, 2023 Aug.
Article in English | MEDLINE | ID: mdl-36882114

ABSTRACT

OBJECTIVE: Evidence has shown that microRNA (miR)-122-5p is a diagnostic biomarker of acute myocardial infarction. Here, we aimed to uncover the functions of miR-122-5p in the pathological process of myocardial ischemia-reperfusion injury (MI/RI). METHODS: An MI/RI model was established by left anterior descending coronary artery ligation in mice. The levels of miR-122-5p, suppressor of cytokine signaling-1 (SOCS1), phosphorylation of Janus kinase 2 (p-JAK2), and signal transducers and activators of transcription (p-STAT3) in the myocardial tissues of mice were measured. Downregulated miR-122-5p or upregulated SOCS1 recombinant adenovirus vectors were injected into mice before MI/RI modeling. The cardiac function, inflammatory response, myocardial infarction area, pathological damage, and cardiomyocyte apoptosis in the myocardial tissues of mice were evaluated. Cardiomyocytes were subjected to hypoxia/reoxygenation (H/R) injury and cardiomyocyte biological function was tested upon transfection of miR-122-5p inhibitor. The target relation between miR-122-5p and SOCS1 was evaluated. RESULTS: miR-122-5p expression and p-JAK2 and p-STAT3 expression were high, and SOCS1 expression was low in the myocardial tissues of MI/RI mice. Decreasing miR-122-5p or increasing SOCS1 expression inactivated the JAK2/STAT3 pathway to alleviate MI/RI by improving cardiac function and reducing inflammatory reaction, myocardial infarction area, pathological damage, and cardiomyocyte apoptosis in mice. Silencing of SOCS1 reversed depleted miR-122-5p-induced cardioprotection for MI/RI mice. In vitro experiments revealed that the downregulation of miR-122-5p induced proliferative, migratory, and invasive capabilities of H/R cardiomyocytes while inhibiting apoptosis. Mechanically, SOCS1 was a target gene of miR-122-5p. CONCLUSION: Our study summarizes that inhibition of miR-122-5p induces SOCS1 expression, thereby relieving MI/RI in mice.


Subject(s)
MicroRNAs , Myocardial Infarction , Myocardial Reperfusion Injury , Mice , Animals , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/prevention & control , MicroRNAs/genetics , MicroRNAs/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Myocardial Infarction/genetics , Myocardium/pathology
2.
J Biochem Mol Toxicol ; 36(10): e23164, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35848756

ABSTRACT

Numerous microRNAs (miRs) are abnormally expressed in response to hypoxia-induced myocardial damage. Herein, miR-34c-5p as a potential pharmaco-target was investigated in a mouse model of chronic intermittent hypoxia (CIH)-induced myocardial damage. A mouse model of myocardial damage was established using CIH with 7% or 21% O2 alternating 60 s for 12 h/day, 21% O2 for 12 h/day. AntagomiR-34c-5p (20 nM/0.1 ml; once a week for 12 weeks) was used as a miR-34c-5p inhibitor in a mouse model with tail-vein injection. In another experiment, mice were administrated with Sirt1 activator SRT1720 (50 mg/kg/day) by intraperitoneal injection. Gene Expression Omnibus database showed a significant upregulation of miR-34c-5p expression in the ischemic myocardium of male mice. In CIH-stimulated mice, miR-34c-5p expression was also significantly increased compared with normal mice. Treatment of antagomiR-34c-5p significantly restrained CIH-triggered myocardial apoptosis. After administration of antagomiR-34c-5p or Sirt1 activator SRT1720, cardiac hypertrophy and oxidative stress were attenuated in CIH-stimulated mice. We also found sirtuin 1 (Sirt1) as a direct target of miR-34c-5p, which was able to mediate Sirt1 protein expression in cardiomyocytes. AntagomiR-34c-5p injection markedly elevated Sirt1 protein expression in CIH-stimulated mice. AntagomiR-34c-5p or Sirt1 activator SRT1720 administration exhibited the antioxidative activity and cardioprotective roles in CIH-stimulated mice.


Subject(s)
MicroRNAs , Sirtuin 1 , Animals , Antagomirs , Apoptosis , Disease Models, Animal , Down-Regulation , Hypoxia/metabolism , Male , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Myocardium/metabolism , Sirtuin 1/genetics , Sirtuin 1/metabolism
3.
Dis Markers ; 2022: 9651092, 2022.
Article in English | MEDLINE | ID: mdl-35082934

ABSTRACT

OBJECTIVE: Heart failure and ventricular remodeling after acute myocardial infarction (AMI) are important factors affecting the prognosis of patients. Therefore, we expected to explore the therapeutic target of AMI by studying the effect of death-associated protein kinase 1 (DAPK1) on AMI rat model. MATERIALS AND METHODS: We used male Sprague-Dawley rats to make AMI model, and after 1, 3, 7, and 14 d, we detect the success rate of modeling and the expression change of DAPK1 through 2, 3, 5-triphenyl tetrazolium chloride staining, myocardial injury markers detection, echocardiographic detection, and histological experiment. In addition, we determined the effect of DAPK1 on AMI by subcutaneous injection of the DAPK1 inhibitor (TC-DAPK 6). The effect of DAPK1 on cardiomyocytes has also been verified in cell experiments on H9c2 cells. RESULTS: The expression of DAPK1 in AMI rats was significantly higher than that in sham group, and it increased with time. The expression of inflammatory factors (interleukin- (IL-) 1ß, IL-6, and tumor necrosis factor-α) in AMI rats treated by TC-DAPK 6 was reduced. In addition, TC-DAPK 6 also reduced the activity of malonaldehyde and increased the activities of superoxide dismutase, glutathione, and catalase. The expression of antioxidant molecules such as peroxiredoxin1/4 and glutathione peroxidase1/3 was also promoted by TC-DAPK 6. In H9c2 cells, TC-DAPK 6 also reduced its oxidative stress level. CONCLUSIONS: The increase of DAPK1 may be related to the pathogenesis of AMI. DAPK1 inhibitors protect cardiomyocytes from AMI-induced myocardial injury by reducing levels of inflammation and oxidative stress in myocardial tissue and cells.


Subject(s)
Death-Associated Protein Kinases/metabolism , Inflammation/metabolism , Myocardial Infarction/metabolism , Oxidative Stress/drug effects , Animals , Antioxidants/metabolism , Disease Models, Animal , Male , Rats , Rats, Sprague-Dawley
4.
J Cardiovasc Pharmacol ; 78(2): 247-252, 2021 06 18.
Article in English | MEDLINE | ID: mdl-34139744

ABSTRACT

ABSTRACT: CircRNA ACAP2 and miR-532 both promotes the apoptosis of cardiomyocytes, which contributes to myocardial infarction (MI). Therefore, ACAP2 and miR-532 may interact with each other to participate in MI. Plasma samples from both patients with MI (n = 65) and healthy controls (n = 65) were subjected to RNA extractions and real-time quantitative polymerase chain reaction to analyze the expression of ACAP2, mature miR-532, and premature miR-532. Correlations among them were analyzed by Pearson's correlation coefficient. Expression of both mature miR-532 and premature miR-532 in cardiomyocytes with ACAP2 overexpression was analyzed by real-time quantitative polymerase chain reaction to study the effects of ACAP2 overexpression on the maturation of miR-532. The role of ACAP2 and miR-532 in regulating the apoptosis of cardiomyocytes induced by hypoxia was analyzed by cell apoptosis assay. In this study, we found that ACAP2 and mature miR-532 were both upregulated in plasma from patients with MI. ACAP2 and mature miR-532 were inversely correlated, whereas ACAP2 and premature miR-532 were not significantly correlated. In cardiomyocytes, overexpression of ACAP2 increased the expression of mature miR-532, but not premature miR-532. Cell apoptosis analysis showed that ACAP2 and miR-532 overexpression promoted the apoptosis of cardiomyocytes induced by hypoxia treatment. In addition, miR-532 inhibitor reduced the effects of ACAP2 overexpression. ACAP2 is overexpressed in MI and may promote the maturation of miR-532 to induce the apoptosis of cardiomyocyte.


Subject(s)
Apoptosis/genetics , Membrane Proteins , MicroRNAs , Myocardial Infarction/metabolism , Myocytes, Cardiac/metabolism , Gene Expression Profiling/methods , Gene Expression Regulation , Humans , Hypoxia/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Circular , Up-Regulation
5.
Sci Total Environ ; 472: 1121-9, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24361747

ABSTRACT

Elemental profiles were determined for size-separated fugitive dust particles produced from Chinese desert and gobi soils. Seventeen surface soil samples from six Chinese deserts were collected, composited, resuspended, and sampled through TSP, PM10, and PM2.5 inlets onto Teflon® filters, which were analyzed for twenty-six elements. Two major dust sources could be distinguished based on differences in crustal and enriched elements-the northwestern (NW) region (Taklimakan Desert, Xinjiang Gobi, and Anxinan Gobi) and northern (N) region (Ulan Buh Desert, Central Inner Mongolia Desert, and Erenhot Gobi). The N sources showed lower concentrations of mineral elements (Fe, K, Na, Ti, Mn, Cr, and Rb in PM10, and Fe, K, Ti, Mn, Co, and V in PM2.5) and higher levels of contaminants (S, Zn, Mo, Cu, Cr, Pb, Cd, and As) than the NW ones, especially in PM2.5. Enrichment factors for Cu, Cr, Zn, Pb, As, Mo, and Cd calculated relative to the upper continental crust showed enrichments of one to two orders-of-magnitude, and they were much higher for N sources than NW ones, implying stronger anthropogenic impacts in north China. Aerosol elemental concentrations during dust events at Horqin, Beijing, and Xi'an matched the mass percentages of mineral elements from their presumptive sources better than the alternative ones, validating the differences between the NW and N sources. Additionally, Na/S, Mg/S, Fe/Al, K/Al, Si/Fe, and Na/Al ratios were suggested to differentiate the two dust source regions. The elemental ratios of Ca/Al, K/Al, Fe/Al, and Ti/Fe in the source regions matched those in aerosols collected downwind, and they can be considered as possible source indicators.


Subject(s)
Air Pollutants/analysis , Dust/analysis , Environmental Monitoring , Trace Elements/analysis , Aerosols/analysis , China , Desert Climate
6.
PLoS One ; 8(6): e67120, 2013.
Article in English | MEDLINE | ID: mdl-23826208

ABSTRACT

OBJECTIVE: To investigate whether lipoxin A4 (LXA4) increases expression of heme oxygenase-1(HO-1) in cardiomyocytes, whether LXA4-induced HO-1 protects cardiomyocytes against hypoxia/reoxygenation (H/R) injury, and what are the mechanisms involved in the LXA4-induced HO-1 induction. METHODS: Rat cardiomyocytes were exposed to H/R injury with or without preincubation with LXA4 or HO-1 inhibitor ZnPP-IX or various signal molecule inhibitors. Expressions of HO-1 protein and mRNA were analyzed by using Western blot and RT-PCR respectively. Activity of nuclear factor E2-related factor 2 (Nrf2) binding to the HO-1 E1 enhancer was assessed by chromatin immunoprecipitation. Nrf2 binding to the HO-1 antioxidant responsive element (ARE) were measured by using electrophoretic mobility shift assay. RESULTS: Pretreatment of the cells undergoing H/R lesion with LXA4 significantly reduced the lactate dehydrogenase and creatine kinase productions, increased the cell viability, and increased the expressions of HO-1 protein and mRNA and HO-1 promoter activity. HO-1 inhibition abolished the protective role of LXA4 on the cells undergoing H/R lesion. LXA4 increased p38 mitogen-activated protein kinase (p38 MAPK) activation, nuclear translocation of Nrf2, Nrf2 binding to the HO-1 ARE and E1 enhancer in cardiomyocytes with or without H/R exposure. CONCLUSION: The protection role of LXA4 against H/R injury of cardiomyocytes is related to upregulation of HO-1, via activation of p38 MAPK pathway and nuclear translocation of Nrf2 and Nrf2 binding to the HO-1 ARE and E1 enhancer, but not via activation of phosphatidyinositol-3-kinase or extracellular signal-regulated kinase pathway.


Subject(s)
Antioxidant Response Elements/genetics , Heme Oxygenase-1/biosynthesis , Hypoxia/enzymology , Lipoxins/pharmacology , Myocytes, Cardiac/pathology , NF-E2-Related Factor 2/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Cardiotonic Agents/pharmacology , Cell Shape/drug effects , Cell Survival/drug effects , Creatine Kinase/metabolism , Enzyme Induction/drug effects , Heme Oxygenase-1/genetics , Hypoxia/genetics , Hypoxia/pathology , L-Lactate Dehydrogenase/metabolism , Male , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/enzymology , Oxygen/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Transport/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , Transcription, Genetic/drug effects , Up-Regulation/drug effects
7.
Article in English | MEDLINE | ID: mdl-23602847

ABSTRACT

Studies have shown that lipoxin A4 (LXA4) and activation of LXA4 receptor provided protection against myocardial ischemia/reperfusion injury in animal models. However, the mechanisms by which LXA4 induced protective role on myocardial ischemia/reperfusion injury remains unclear. In the present studies, we investigated the protective effects of LXA4 on H9c2 cardiomyocytes exposed to hypoxia/reoxygenation (H/R) injury and involvement of heme oxygenase-1 (HO-1)- and K(+) channel-dependant pathways in the LXA4 action. H9c2 cardiomyocytes were pretreated with or without LXA4 or HO-1 specific interfering RNA (siRNA) or various blockers and openers of K(+) channels before exposing to H/R injury. The levels of lactate dehydrogenase (LDH) and creatine kinase (CK) in cellular supernatants and necrosis factor-α (TNF-α) in cellular lysates were measured by using ELISA. Expressions of HO-1 mRNA and protein were analyzed by using RT-PCR and Western blot respectively. Pretreatment of the cells undergoing H/R injury with LXA4 significantly reduced the LDH and CK levels induced by H/R injury, and increased the expressions and activity of HO-1. However, the protective effects of LXA4 were completely blocked by transfection of the cells with HO-1 siRNA, and were partially but significantly blocked by pretreatment of the cells with various blockers of K(+) channels. The LXA4-induced expressions of HO-1 in the cells were also inhibited by HO-1 siRNA and various blockers of K(+) channels. The inhibitory effects of LXA4 on enhanced TNF-α levels induced by H/R injury were abolished by transfection of the cells with HO-1 siRNA. In conclusion, the protective role of LXA4 on cardiomyocytes against H/R injury is related to upregulation of HO-1 via reduced production of TNF-α and activation of ATP-sensitive K(+) channels and calcium-sensitive K(+) channel.


Subject(s)
Lipoxins/metabolism , Myocytes, Cardiac/metabolism , Potassium Channels/metabolism , Animals , Cell Hypoxia , Creatine Kinase/genetics , Creatine Kinase/metabolism , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Lipoxins/pharmacology , Myocardial Reperfusion Injury/metabolism , Myocytes, Cardiac/pathology , RNA, Messenger/metabolism , Rats , Up-Regulation
8.
Zhong Xi Yi Jie He Xue Bao ; 2(2): 94-6, 2004 Mar.
Article in Chinese | MEDLINE | ID: mdl-15339465

ABSTRACT

OBJECTIVE: To evaluate the effect of Xiangdan Injection on mRNA expression of the endothelial vaso-active factors of patients with coronary heart disease and blood stasis. METHODS: Fifty-six patients were randomly divided into two groups:twenty-eight patients were treated according to the therapeutic guide for coronary heart disease as the control group and 28 were given the same treatment plus Xiangdan Injection as the treated group. The expressions of ET-1 and eNOS mRNA were examined with RT-PCR before experiment and ten days later. RESULTS: The positive rate of eNOS mRNA of the treated group increased, while the positive rate of ET-1 mRNA of the treated group decreased after ten day's treatment, with significant differences as compared with that before the experiment. Xiangdan Injection up-regulated the eNOS mRNA expression and suppressed the ET-1 mRNA expression. Changes of expression were not observed in the control group. CONCLUSION: Xiangdan Injection improves the endothelial function of patients with coronary heart disease and blood stasis by regulating the expressions of ET-1 and eNOS mRNA.


Subject(s)
Coronary Disease/drug therapy , Endothelin-1/genetics , Medicine, Chinese Traditional , Nitric Oxide Synthase/genetics , RNA, Messenger/analysis , Aged , Aged, 80 and over , Coronary Disease/metabolism , Coronary Thrombosis/drug therapy , Coronary Thrombosis/metabolism , Female , Humans , Injections , Male , Middle Aged , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type III
SELECTION OF CITATIONS
SEARCH DETAIL
...