Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 607
Filter
1.
Scand Cardiovasc J ; 58(1): 2347290, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38733316

ABSTRACT

Objectives. The aim of this study was to investigate the expression of long non-coding RNA (lncRNA) brain and reproductive organ-expressed protein (BRE) antisense RNA 1 (BRE-AS1) in patients with acute myocardial infarction (AMI) and its effect on ischemia/reperfusion (I/R)-induced oxidative stress and apoptosis of cardiomyocytes. Methods. Serum BRE-AS1 levels in patients with AMI was detected using quantitative real-time polymerase chain reaction (qRT-PCR). The diagnostic and prognostic values of BRE-AS1 were evaluated. H9c2 cells were treated with hypoxia/reoxygenation to establish an in vitro myocardial infarction cell model. The levels of inflammatory cytokines such as tumor necrosis factor-α (TNF-α), interleukin-1ß (IL-1ß), and IL-6 were detected by enzyme-linked immunosorbent assay (ELISA). Levels of lactate dehydrogenase (LDH), malondialdehyde (MDA), superoxide dismutase (SOD), and glutathione peroxidase (GSH-Px) were determined by commercial kits. Cell counting kit-8 (CCK-8) and flow cytometry were used to evaluate the cell viability and cell apoptosis. Results. The expression of BRE-AS1 in serum of patients with AMI is upregulated, which shows the clinical diagnostic value for AMI. In the I/R injury cell model, the knockout of BRE-AS1 can significantly alleviate the increase in TNF-α, IL-1ß, and IL-6 levels, inhibit the production of LDH and MDA, increase the activities of SOD and GSH-Px, promote the cell viability and suppress cell apoptosis. Conclusions. Abnormally elevated BRE-AS1 has a high diagnostic value for AMI as well as a prognostic value for major adverse cardiovascular events (MACEs). The elevation of BRE-AS1 promoted oxidative stress injury and cell apoptosis in vitro.


Subject(s)
Apoptosis , Inflammation Mediators , Myocardial Infarction , Myocytes, Cardiac , Oxidative Stress , RNA, Long Noncoding , RNA, Long Noncoding/blood , RNA, Long Noncoding/metabolism , RNA, Long Noncoding/genetics , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Humans , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/blood , Myocardial Infarction/genetics , Myocardial Infarction/diagnosis , Male , Middle Aged , Female , Inflammation Mediators/metabolism , Inflammation Mediators/blood , Cell Line , Animals , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/blood , Myocardial Reperfusion Injury/diagnosis , Myocardial Reperfusion Injury/genetics , Rats , Cytokines/metabolism , Cytokines/blood , Signal Transduction , Case-Control Studies , Aged , Up-Regulation
2.
Can J Physiol Pharmacol ; 102(5): 331-341, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38118123

ABSTRACT

Extracellular potassium concentration might modify electrophysiological properties in the border zone of ischemic myocardium. We evaluated the depolarization and repolarization characteristics across the ischemic-normal border under [K+] variation. Sixty-four-lead epicardial mapping was performed in 26 rats ([K+] 2.3-6.4 mM) in a model of acute ischemia/reperfusion. The animals with [K+] < 4.7 mM (low-normal potassium) had an ischemic zone with ST-segment elevation and activation delay, a border zone with ST-segment elevation and no activation delay, and a normal zone without electrophysiological abnormalities. The animals with [K+] >4.7 mM (normal-high potassium) had only the ischemic and normal zones and no transitional area. Activation-repolarization intervals and local conduction velocities were inversely associated with [K+] in linear regression analysis with adjustment for the zone of myocardium. The reperfusion extrasystolic burden (ESB) was greater in the low-normal as compared to normal-high potassium animals. Ventricular tachycardia/fibrillation incidence did not differ between the groups. In patch-clamp experiments, hypoxia shortened action potential duration at 5.4 mM but not at 1.3 mM of [K+]. IK(ATP) current was lower at 1.3 mM than at 5.4 mM of [K+]. We conclude that the border zone formation in low-normal [K+] was associated with attenuation of IK(ATP) response to hypoxia and increased reperfusion ESB.


Subject(s)
Action Potentials , Myocardial Ischemia , Potassium , Animals , Potassium/blood , Potassium/metabolism , Male , Rats , Myocardial Ischemia/physiopathology , Myocardial Ischemia/blood , Myocardial Ischemia/metabolism , Action Potentials/physiology , Myocardial Reperfusion Injury/blood , Myocardial Reperfusion Injury/physiopathology , Myocardial Reperfusion Injury/metabolism , Rats, Wistar
3.
Cells ; 10(12)2021 11 27.
Article in English | MEDLINE | ID: mdl-34943838

ABSTRACT

The majority of cardiovascular deaths are associated with acute coronary syndrome, especially ST-elevation myocardial infarction. Therapeutic reperfusion alone can contribute up to 40 percent of total infarct size following coronary artery occlusion, which is called ischemia-reperfusion injury (IRI). Its size depends on many factors, including the main risk factors of cardiovascular mortality, such as age, sex, systolic blood pressure, smoking, and total cholesterol level as well as obesity, diabetes, and physical effort. Extracellular vesicles (EVs) are membrane-coated particles released by every type of cell, which can carry content that affects the functioning of other tissues. Their role is essential in the communication between healthy and dysfunctional cells. In this article, data on the variability of the content of EVs in patients with the most prevalent cardiovascular risk factors is presented, and their influence on IRI is discussed.


Subject(s)
Extracellular Vesicles/metabolism , Myocardial Reperfusion Injury/blood , Myocardial Reperfusion Injury/epidemiology , Age Factors , Animals , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/physiopathology , Risk Factors , Sex Characteristics
4.
Sci Rep ; 11(1): 16999, 2021 08 20.
Article in English | MEDLINE | ID: mdl-34417540

ABSTRACT

The effect of uridine on the myocardial ischemic and reperfusion injury was investigated. A possible mechanism of its cardioprotective action was established. Two rat models were used: (1) acute myocardial ischemia induced by occlusion of the left coronary artery for 60 min; and (2) myocardial ischemia/reperfusion with 30-min ischemia and 120-min reperfusion. In both models, treatment with uridine (30 mg/kg) prevented a decrease in cell energy supply and in the activity of the antioxidant system, as well as an increase in the level of lipid hydroperoxides and diene conjugates. This led to a reduction of the necrosis zone in the myocardium and disturbances in the heart rhythm. The blocker of the mitochondrial ATP-dependent potassium (mitoKATP) channel 5-hydroxydecanoate limited the positive effects of uridine. The data indicate that the cardioprotective action of uridine may be related to the activation of the mitoKATP channel. Intravenously injected uridine was more rapidly eliminated from the blood in hypoxia than in normoxia, and the level of the mitoKATP channel activator UDP in the myocardium after uridine administration increased. The results suggest that the use of uridine can be a potentially effective approach to the management of cardiovascular diseases.


Subject(s)
Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardium/pathology , Potassium Channels/metabolism , Uridine/pharmacology , Acute Disease , Adenosine Triphosphate/metabolism , Animals , Antioxidants/metabolism , Arrhythmias, Cardiac/blood , Arrhythmias, Cardiac/drug therapy , Arrhythmias, Cardiac/etiology , Disease Models, Animal , Lipid Peroxidation/drug effects , Male , Myocardial Reperfusion Injury/blood , Myocardium/metabolism , Rats, Wistar , Tachycardia/blood , Tachycardia/complications , Uridine/blood , Uridine/therapeutic use , Uridine Diphosphate/metabolism , Uridine Triphosphate/metabolism , Ventricular Fibrillation/complications , Ventricular Fibrillation/drug therapy
5.
Microvasc Res ; 138: 104235, 2021 11.
Article in English | MEDLINE | ID: mdl-34453991

ABSTRACT

Electroacupuncture (EA) intervention has a remarkable cardioprotection against myocardial ischemia reperfusion injury (MIRI). Recently, it has been suggested that the gut microbiota plays an important role in regulating the progression and prognosis of MIRI. The purpose of this study was to illustrate the relationship between gut microbiota and cardioprotection of EA on MIRI. We conducted a MIRI model by ligating the left anterior descending coronary artery for 30 min followed by reperfusion in male Sprague Dawley rats, which then received 7 days of EA intervention. Echocardiography was employed to evaluate left ventricular function. Fecal samples were collected for microbial analysis by 16S rDNA high-throughput sequencing. Blood samples and myocardium were collected for inflammatory cytokine detection by enzyme linked immunosorbent assay (ELISA) and Western blot. Hematoxylin & eosin (HE) staining and immunofluorescence of ileum tissue were performed for intestinal damage evaluation. After 7 days of EA intervention, the left ventricular function was improved with significantly increased ejection fraction and fractional shortening. Furthermore, we found that EA intervention reversed the changed gut microbiota induced by MIRI, including Clostridiales, RF39, S24-7, Desulfovibrio, and Allobaculum, improved the impaired gut barrier, reduced the production and circulation of lipopolysaccharide (LPS), inhibited the level of interleukin 6 (IL-6) and interleukin 12 (IL-12) in periphery and decreased the expression of Toll like receptor 4 (TLR4) and IL-6 in myocardium. EA intervention could improve the impaired gut mucosal barrier and reduce the production and circulation of LPS after MIRI through regulating gut microbiota, thus inhibiting the circulation and myocardium inflammation and finally exerted the cardioprotective effect.


Subject(s)
Bacteria/metabolism , Electroacupuncture , Gastrointestinal Microbiome , Inflammation Mediators/metabolism , Intestinal Mucosa/microbiology , Lipopolysaccharides/blood , Myocardial Reperfusion Injury/prevention & control , Myocardium/metabolism , Acute-Phase Proteins , Animals , Bacteria/growth & development , Carrier Proteins/blood , Disease Models, Animal , Dysbiosis , Male , Membrane Glycoproteins/blood , Myocardial Reperfusion Injury/blood , Myocardial Reperfusion Injury/microbiology , Myocardial Reperfusion Injury/pathology , Myocardium/pathology , Rats, Sprague-Dawley , Ventricular Function, Left
6.
Cardiovasc Toxicol ; 21(11): 889-900, 2021 11.
Article in English | MEDLINE | ID: mdl-34324134

ABSTRACT

Complete vascular occlusion to distant tissue prior to an ischemic cardiac event can provide significant cardioprotection via remote ischemic preconditioning (RIPC). Despite understanding its mechanistic basis, its translation to clinical practice has been unsuccessful, likely secondary to the inherent impossibility of predicting (and therefore preconditioning) an ischemic event, as well as the discomfort that is associated with traditional, fully occlusive RIPC stimuli. Our laboratory has previously shown that non-occlusive banding (NOB) via wrapping of a leather band (similar to a traditional Jewish ritual) can elicit an RIPC response in healthy human subjects. This study sought to further the pain-mediated aspect of this observation in a mouse model of NOB with healthy mice that were exposed to treatment with and without lidocaine to inhibit pain sensation prior to ischemia/reperfusion injury. We demonstrated that NOB downregulates key inflammatory markers resulting in a preconditioning response that is partially mediated via pain sensation.


Subject(s)
Anesthetics, Local/pharmacology , Forelimb/blood supply , Ischemic Preconditioning/methods , Lidocaine/pharmacology , Myocardial Infarction/prevention & control , Myocardial Reperfusion Injury/prevention & control , Pain Threshold/drug effects , Radial Artery/physiology , Animals , Cytokines/blood , Cytokines/genetics , Disease Models, Animal , Echocardiography , Ligation , Male , Mice, Inbred C57BL , Myocardial Infarction/blood , Myocardial Infarction/diagnostic imaging , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/blood , Myocardial Reperfusion Injury/diagnostic imaging , Myocardial Reperfusion Injury/physiopathology , Myocardium/metabolism , Myocardium/pathology , Radial Artery/diagnostic imaging , Regional Blood Flow , Time Factors
7.
Basic Res Cardiol ; 116(1): 36, 2021 05 26.
Article in English | MEDLINE | ID: mdl-34037861

ABSTRACT

We studied the translational cardioprotective potential of P2Y12 inhibitors against acute myocardial ischemia/reperfusion injury (IRI) in an animal model of acute myocardial infarction and in patients with ST-elevation myocardial infarction (STEMI) undergoing primary percutaneous coronary intervention (PPCI). P2Y12 inhibitors may have pleiotropic effects to induce cardioprotection against acute myocardial IRI beyond their inhibitory effects on platelet aggregation. We compared the cardioprotective effects of clopidogrel, prasugrel, and ticagrelor on infarct size in an in vivo rat model of acute myocardial IRI, and investigated the effects of the P2Y12 inhibitors on enzymatic infarct size (48-h area-under-the-curve (AUC) troponin T release) and clinical outcomes in a retrospective study of STEMI patients from the CONDI-2/ERIC-PPCI trial using propensity score analyses. Loading with ticagrelor in rats reduced infarct size after acute myocardial IRI compared to controls (37 ± 11% vs 52 ± 8%, p < 0.01), whereas clopidogrel and prasugrel did not (50 ± 11%, p > 0.99 and 49 ± 9%, p > 0.99, respectively). Correspondingly, troponin release was reduced in STEMI patients treated with ticagrelor compared to clopidogrel (adjusted 48-h AUC ratio: 0.67, 95% CI 0.47-0.94). Compared to clopidogrel, the composite endpoint of cardiac death or hospitalization for heart failure within 12 months was reduced in STEMI patients loaded with ticagrelor (HR 0.63; 95% CI 0.42-0.94) but not prasugrel (HR 0.84, 95% CI 0.43-1.63), prior to PPCI. Major adverse cardiovascular events did not differ between clopidogrel, ticagrelor, or prasugrel. The cardioprotective effects of ticagrelor in reducing infarct size may contribute to the clinical benefit observed in STEMI patients undergoing PPCI.


Subject(s)
Blood Platelets/drug effects , Myocardial Reperfusion Injury/prevention & control , Percutaneous Coronary Intervention , Platelet Aggregation Inhibitors/therapeutic use , Platelet Aggregation/drug effects , Purinergic P2Y Receptor Antagonists/therapeutic use , Receptors, Purinergic P2Y12/drug effects , ST Elevation Myocardial Infarction/therapy , Translational Research, Biomedical , Aged , Animals , Blood Platelets/metabolism , Clopidogrel/therapeutic use , Disease Models, Animal , Female , Humans , Male , Middle Aged , Myocardial Reperfusion Injury/blood , Myocardial Reperfusion Injury/etiology , Myocardial Reperfusion Injury/physiopathology , Percutaneous Coronary Intervention/adverse effects , Platelet Aggregation Inhibitors/adverse effects , Prasugrel Hydrochloride/therapeutic use , Purinergic P2Y Receptor Antagonists/adverse effects , Randomized Controlled Trials as Topic , Rats , Receptors, Purinergic P2Y12/metabolism , Retrospective Studies , ST Elevation Myocardial Infarction/physiopathology , Ticagrelor/therapeutic use , Treatment Outcome
8.
Mol Neurobiol ; 58(9): 4268-4279, 2021 Sep.
Article in English | MEDLINE | ID: mdl-33982231

ABSTRACT

Increased histone deacetylase 3 (HDAC3) has been demonstrated to contribute to the pathogenesis of myocardial ischemia-reperfusion injury (MI/RI). Therefore, the goal of this study was to investigate how HDAC3 regulated MI/RI by mediating microRNA (miR)-494-3p/dromodomain-containing protein 4 (BRD4) axis. The MI/RI model was established by ligating the right anterior descending coronary artery. Cardiomyocytes from newborn mice were treated with hypoxia/reoxygenation (H/R). Gain-of-function and loss-of-function approaches were implemented to figure out the roles of miR-494-3p and HDAC3 in MI/RI. miR-494-3p, HDAC3, and BRD4 in myocardial tissues of mice with MI/RI and H/R-treated cardiomyocytes were detected. The relationships between miR-494-3p and HDAC3 and BRD4 were identified. Reduced miR-494-3p and upregulated HDAC3 and BRD4 exhibited in myocardial tissues of mice with MI/RI and H/R-treated cardiomyocytes. Inhibited HDAC3 or elevated miR-494-3p repressed the inflammation and apoptosis, improved cardiac function, and ameliorated myocardial injury in myocardial tissues of mice with MI/RI. Suppression of HDAC3 or elevation of miR-494-3p depressed inflammation and apoptosis and promoted cell viability of primary cardiomyocytes. miR-494-3p targeted BRD4. The study concludes that suppressed HDAC3 plays a protective role in MI/RI by upregulation of miR-494-3p and inhibition of BRD4, which could be helpful for MI/RI therapy.


Subject(s)
Histone Deacetylases/metabolism , Inflammation/metabolism , MicroRNAs/metabolism , Myocardial Reperfusion Injury/metabolism , Myocytes, Cardiac/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Animals , Apoptosis/physiology , Disease Models, Animal , Female , Inflammation/blood , Interleukin-1/blood , Interleukin-6/blood , Mice , Myocardial Reperfusion Injury/blood , Myocardium/metabolism , Tumor Necrosis Factor-alpha/blood
9.
Medicine (Baltimore) ; 100(10): e24944, 2021 Mar 12.
Article in English | MEDLINE | ID: mdl-33725857

ABSTRACT

INTRODUCTION: The morbidity and mortality of acute myocardial infarction patients still remains high after percutaneous coronary intervention (PCI). Myocardial ischemia-reperfusion (MIR) injury is one of the important reasons. Although the phenomenon of MIR injury can paradoxically reduce the beneficial effects of myocardial reperfusion, there currently remains no effective therapeutic agent for preventing MIR. Previous studies have shown that Yiqi Liangxue Shengji prescription (YLS) is effective in improving clinical symptoms and ameliorating the major adverse cardiovascular events of coronary heart disease patients undergoing PCI. This study aims to evaluate the effectiveness and safety of YLS in patients with acute myocardial infarction (AMI) after PCI. METHODS: This study is a randomized, double-blinded, placebo-controlled, single-central clinical trial. A total of 140 participants are randomly allocated to 2 groups: the intervention group and the placebo group. Based on routine medications, the intervention group will be treated with YLS and the placebo group will be treated with YLS placebo. All participants will receive a 8-week treatment and then be followed up for another 12 months. The primary outcome measures are N terminal pro B type natriuretic peptide (NT-proBNP) and left ventricular ejection fraction. Secondary outcomes are plasma levels of microRNA-145, plasma cardiac enzyme, and Troponin I levels in blood samples, changes in ST-segment in ECG, Seattle Angina Questionnaire, the efficacy of angina symptoms, and occurrence of major adverse cardiac events. All the data will be recorded in case report forms and analyzed by SPSS V.17.0. DISCUSSION: The trial will investigate whether the postoperative administration of YLS in patients with AMI after PCI will improve cardiac function. And it explores microRNAs (miRNA)-145 as detection of blood-based biomarkers for AMI by evaluating the relation between miRNAs in plasma and cardiac function. TRIAL REGISTRATION: Chinese Clinical Trials Registry identifier ChiCTR2000038816. Registered on October 10, 2020.


Subject(s)
Coronary Disease/complications , Drugs, Chinese Herbal/therapeutic use , Myocardial Infarction/drug therapy , Myocardial Reperfusion Injury/prevention & control , Adolescent , Adult , Aged , Aged, 80 and over , Coronary Disease/surgery , Drugs, Chinese Herbal/pharmacology , Electrocardiography , Female , Humans , Male , MicroRNAs/blood , Middle Aged , Myocardial Infarction/blood , Myocardial Infarction/diagnosis , Myocardial Infarction/etiology , Myocardial Reperfusion Injury/blood , Myocardial Reperfusion Injury/diagnosis , Myocardial Reperfusion Injury/etiology , Natriuretic Peptide, Brain/blood , Peptide Fragments/blood , Percutaneous Coronary Intervention , Randomized Controlled Trials as Topic , Treatment Outcome , Troponin I/blood , Ventricular Function, Left/drug effects , Young Adult
10.
J Am Heart Assoc ; 10(7): e019216, 2021 04 06.
Article in English | MEDLINE | ID: mdl-33728934

ABSTRACT

Background Perioperative blood glucose level has shown an association with postoperative outcomes. We compared the incidences of myocardial injury after noncardiac surgery (MINS) and 30-day mortality, according to preoperative blood glucose and hemoglobin A1c (HbA1c) levels. Methods and Results The patients were divided according to blood glucose level within 1 day before surgery. The hyperglycemia group was defined with fasting glucose >140 mg/dL or random glucose >180 mg/dL. In addition, we compared the outcomes according to HbA1c >6.5% among patients with available HbA1c within 3 months before surgery. The primary outcome was MINS, and 30-day mortality was also compared. A total of 12 304 patients were enrolled and divided into 2 groups: 8324 (67.7%) in the normal group and 3980 (32.3%) in the hyperglycemia group. After adjustment with inverse probability of weighting, the hyperglycemia group exhibited significantly higher incidences of MINS and 30-day mortality (18.7% versus 27.6%; odds ratio, 1.29; 95% CI, 1.18-1.42; P<0.001; and 2.0% versus 5.1%; hazard ratio, 2.00; 95% CI, 1.61-2.49; P<0.001, respectively). In contrast to blood glucose, HbA1c was not associated with MINS or 30-day mortality. Conclusions Preoperative hyperglycemia was associated with MINS and 30-day mortality, whereas HbA1c was not. Immediate glucose control may be more crucial than long-term glucose control in patients undergoing noncardiac surgery. Registration URL: https://www.cris.nih.go.kr; Unique identifier: KCT0004244.


Subject(s)
Blood Glucose/metabolism , Glycated Hemoglobin/metabolism , Myocardial Reperfusion Injury/blood , Postoperative Complications/blood , Surgical Procedures, Operative/adverse effects , Female , Follow-Up Studies , Humans , Incidence , Male , Middle Aged , Myocardial Reperfusion Injury/epidemiology , Myocardial Reperfusion Injury/etiology , Postoperative Complications/epidemiology , Postoperative Complications/etiology , Postoperative Period , Republic of Korea/epidemiology , Retrospective Studies , Survival Rate/trends
11.
J Cell Physiol ; 236(9): 6657-6665, 2021 09.
Article in English | MEDLINE | ID: mdl-33554327

ABSTRACT

Oxygen is often administered to patients and occasionally to healthy individuals as well; however, the cellular toxicity of oxygen, especially following prolonged exposure, is widely known. To evaluate the potential effect of oxygen exposure on circulating stem/progenitor cells and cardiac ischemia/reperfusion (I/R) injury, we exposed healthy adult mice to 100% oxygen for 20 or 60 min. We then examined the c-kit-positive stem/progenitor cells and colony-forming cells and measured the cytokine/chemokine levels in peripheral blood. We also induced cardiac I/R injury in mice at 3 h after 60 min of oxygen exposure and examined the recruitment of inflammatory cells and the fibrotic area in the heart. The proportion of c-kit-positive stem/progenitor cells significantly increased in peripheral blood at 3 and 24 h after oxygen exposure for either 20 or 60 min (p < .01 vs. control). However, the abundance of colony-forming cells in peripheral blood conversely decreased at 3 and 24 h after oxygen exposure for only 60 min (p < .05 vs. control). Oxygen exposure for either 20 or 60 min resulted in significantly decreased plasma vascular endothelial growth factor levels at 3 h, whereas oxygen exposure for only 60 min reduced plasma insulin-like growth factor 1 levels at 24 h (p < .05 vs. control). Protein array indicated the increase in the levels of some cytokines/chemokines, such as CXCL6 (GCP-2) at 24 h after 60 min of oxygen exposure. Moreover, oxygen exposure for 60 min enhanced the recruitment of Ly6g- and CD11c-positive inflammatory cells at 3 days (p < .05 vs. control) and increased the fibrotic area at 14 days in the heart after I/R injury (p < .05 vs. control). Prolonged oxygen exposure induced the mobilization and functional impairment of stem/progenitor cells and likely enhanced inflammatory responses to exacerbate cardiac I/R injury in healthy mice.


Subject(s)
Myocardial Reperfusion Injury/pathology , Oxygen/adverse effects , Stem Cells/pathology , Animals , Chemokine CXCL12/blood , Colony-Forming Units Assay , Inflammation Mediators/blood , Male , Mice, Inbred C57BL , Myocardial Reperfusion Injury/blood , Myocardium/metabolism , Myocardium/pathology , Proto-Oncogene Proteins c-kit/metabolism , Reactive Oxygen Species/metabolism , Vascular Endothelial Growth Factor A/blood
13.
Microcirculation ; 28(1): e12657, 2021 01.
Article in English | MEDLINE | ID: mdl-32892409

ABSTRACT

OBJECTIVE: This study examined the correlation between serum miR-98-5p levels and indices of microvascular reperfusion in patients undergoing primary percutaneous coronary intervention (pPCI) after ST-segment elevation myocardial infarction (STEMI). Additionally, we evaluated the mechanisms by which miR-98-5p promoted ischemia/reperfusion (I/R)-induced injury in both cultured cell lines and an animal model. METHODS: Circulating miR-98-5p levels were measured and compared from 171 STEMI patients undergoing pPCI, who were divided into two groups: no-reflow and reflow. The levels of miR-98-5p, nerve growth factor (NGF), and transient receptor potential vanilloid 1 (TRPV1) were analyzed in cultured human coronary endothelial cells (HCECs) exposed to hypoxia/reoxygenation (H/R). The effects of antagomir-98-5p on myocardial I/R-induced microvascular dysfunction in vivo were evaluated. Target gene expression and activity were assessed. RESULTS: Higher miR-98-5p levels were associated with compromised indices of microvascular reperfusion. In vitro experiments on HCECs showed that exposure to H/R significantly increased miR-98-5p levels. We identified NGF as a novel target of miR-98-5p. Further, antagomir-98-5p relieved microvascular dysfunction and enhanced the expression of NGF and TRPV1 in the rat myocardial I/R model. CONCLUSIONS: MiR-98-5p promotes microvascular dysfunction by targeting the NGF-TRPV1 axis. Serum miR-98-5p serves as a potential biomarker for microvascular reperfusion.


Subject(s)
Coronary Vessels/metabolism , MicroRNAs/blood , Microvessels/metabolism , Myocardial Reperfusion Injury/blood , Nerve Growth Factor/blood , Aged , Biomarkers/blood , Cells, Cultured , Coronary Vessels/pathology , Endothelial Cells/metabolism , Endothelial Cells/physiology , Female , Follow-Up Studies , Gene Expression Regulation , Humans , Male , Microvessels/pathology , Middle Aged , Myocardial Reperfusion Injury/pathology
14.
Cell Biochem Funct ; 39(2): 287-295, 2021 Mar.
Article in English | MEDLINE | ID: mdl-32767595

ABSTRACT

Ischaemia/reperfusion (I/R) injury can lead to severe arrhythmia and aggravate myocardial damage. Exosomes are small-membrane vesicles that play a protective role in myocardial I/R injury. This study aimed to explore the protective effects of ischaemic preconditioning (IPC)-induced serum exosomes (IPC-Exo) on myocardial I/R injury in rats and its underlying mechanism. Serum exosomes were extracted from IPC rats and quantified using a bicinchoninic acid assay kit. IPC-Exo (50 µg) was injected into the infarcted myocardium immediately after ligation. Rats were randomly divided into Sham, I/R, IPC-Exo + I/R, I/R + LY294002, and I/R + IPC-Exo + LY294002 groups. Haemodynamic parameters were measured by physiological recording. Transthoracic echocardiography was used to detect cardiac function. The serum levels of creatine kinase isomer-MB, lactate dehydrogenase, aspartate transaminase, tumour necrosis factor-alpha, interleukin (IL)-1ß, and IL-10 were detected by enzyme-linked immunosorbent assay. Triphenyl tetrazolium chloride staining was used to measure the myocardial infarct size. Apoptosis in myocardial tissues was detected by TUNEL staining. Western blotting was used to detect the levels of PI3K/AKT and apoptosis-related proteins. Our results showed that treatment with IPC-Exo ameliorated cardiac function and reduced inflammatory factor production, cardiomyocyte apoptosis, and myocardial infarct size. Moreover, IPC-Exo treatment promoted the protein expression of Bcl-2, p-PI3K, and p-AKT but inhibited that of caspase-3 and Bax. However, treatment with LY294002 significantly reversed that IPC-Exo-induced increase in p-PI3K and p-AKT levels, improvement of haemodynamics, and decrease of inflammatory factor production and apoptosis in the I/R + IPC-Exo group. Taken together, our results suggest that IPC-Exo may alleviate I/R injury via activating the PI3K/AKT signalling pathway.


Subject(s)
Exosomes/metabolism , Ischemic Preconditioning , Myocardial Reperfusion Injury/pathology , Signal Transduction , Animals , Apoptosis/drug effects , Caspase 3/metabolism , Chromones/pharmacology , Creatine Kinase, MB Form/metabolism , Hemodynamics/drug effects , Male , Morpholines/pharmacology , Myocardial Reperfusion Injury/blood , Myocardial Reperfusion Injury/metabolism , Myocardium/cytology , Myocardium/metabolism , Myocardium/pathology , Phosphatidylinositol 3-Kinases/chemistry , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Tetraspanin 30/metabolism , Tumor Necrosis Factor-alpha/metabolism , Ventricular Function, Left/physiology
15.
Naunyn Schmiedebergs Arch Pharmacol ; 394(2): 401-410, 2021 02.
Article in English | MEDLINE | ID: mdl-32621060

ABSTRACT

Ferroptosis is an iron-dependent regulated necrosis. This study aims to evaluate the contribution of ferroptosis to ischemia or reperfusion injury, and lay a basis for precise therapy of myocardial infarction. The Sprague-Dawley (SD) rat hearts were subjected to ischemia for different duration or the hearts were treated with 1 h-ischemia plus different duration of reperfusion. The myocardial injury was assessed by biochemical assays and hematoxylin & eosin (HE) staining. The ferroptosis was evaluated with the levels of acyl-CoA synthetase long-chain family member 4 (ACSL4), glutathione peroxidase 4 (GPX4), iron, and malondialdehyde. Iron chelator (deferoxamine) was applied to verify the contribution of ferroptosis to ischemia and reperfusion injury. The results showed that ischemic injury (infarction and CK release) was getting worse with the extension of ischemia, but no significant changes in ferroptosis indexes (ACSL4, GPX4, iron, and malondialdehyde) in cardiac tissues were observed. Differently, the levels of ACSL4, iron, and malondialdehyde were gradually elevated with the extension of reperfusion concomitant with a decrease of GPX4 level. In the ischemia-treated rat hearts, no significant changes in myocardial injury were observed in the presence of deferoxamine, while in the ischemia/reperfusion-treated rat hearts, myocardial injury was markedly attenuated in the presence of deferoxamine concomitant with a reduction of ferroptosis. Based on these observations, we conclude that ferroptosis occurs mainly in the phase of myocardial reperfusion but not ischemia. Thus, intervention of ferroptosis exerts beneficial effects on reperfusion injury but not ischemic injury, laying a basis for precise therapy for patients with myocardial infarction.


Subject(s)
Ferroptosis , Ischemia , Myocardial Reperfusion Injury , Animals , Biomarkers/blood , Biomarkers/metabolism , Coenzyme A Ligases/metabolism , Creatine Kinase/blood , Deferoxamine/pharmacology , Iron/metabolism , Ischemia/metabolism , Male , Malondialdehyde/metabolism , Myocardial Reperfusion Injury/blood , Myocardial Reperfusion Injury/metabolism , Myocardium/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Rats, Sprague-Dawley , Siderophores/pharmacology
16.
PLoS One ; 15(12): e0244792, 2020.
Article in English | MEDLINE | ID: mdl-33382840

ABSTRACT

Blood coagulation is central to myocardial ischemia and reperfusion (IR) injury. Studies on the light elicited circadian rhythm protein Period 2 (PER2) using whole body Per2-/- mice found deficient platelet function and reduced clotting which would be expected to protect from myocardial IR-injury. In contrast, intense light induction of PER2 protected from myocardial IR-injury while Per2 deficiency was detrimental. Based on these conflicting data, we sought to evaluate the role of platelet specific PER2 in coagulation and myocardial ischemia and reperfusion injury. We demonstrated that platelets from mice with tissue-specific deletion of Per2 in the megakaryocyte lineage (Per2loxP/loxP-PF4-CRE) significantly clot faster than platelets from control mice. We further found increases in infarct sizes or plasma troponin levels in Per2loxP/loxP-PF4-CRE mice when compared to controls. As intense light increases PER2 protein in human tissues, we also performed translational studies and tested the effects of intense light therapy on coagulation in healthy human subjects. Our human studies revealed that intense light therapy repressed procoagulant pathways in human plasma samples and significantly reduced the clot rate. Based on these results we conclude that intense light elicited PER2 has an inhibitory function on platelet aggregation in mice. Further, we suggest intense light as a novel therapy to prevent or treat clotting in a clinical setting.


Subject(s)
Blood Coagulation/physiology , Blood Platelets/metabolism , Myocardial Ischemia/metabolism , Myocardial Reperfusion Injury/metabolism , Period Circadian Proteins/metabolism , Phototherapy , Animals , Humans , Light , Male , Mice , Myocardial Ischemia/blood , Myocardial Reperfusion Injury/blood , Period Circadian Proteins/genetics , Platelet Aggregation/physiology , Proteomics
17.
Int Immunopharmacol ; 89(Pt B): 107086, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33068868

ABSTRACT

AIMS: The purpose of this study was to investigate the protective effects of puerarin and elucidate the underlying mechanisms of puerarin in myocardial ischemia/reperfusion (MI/R) injury. MAIN METHODS: C57BL/6 mice were exposed to puerarin (100 mg/kg) with or without the SIRT1 inhibitor nicotinamide (500 mg/kg) and then subjected to MI/R operation. Myocardial infarct size, serum creatine kinase-MB (CK-MB) activity, apoptotic cell death, and cardiac structure and function were examined to evaluate MI/R injury. RT-PCR and western blotting were used to determine the inflammatory response and inflammasome activation, as well as activation of SIRT1/NF-κB pathway. RESULTS: Puerarin significantly reduced myocardial infarct size, serum CK-MB activity, and apoptotic cell death, and improved cardiac structural damage and dysfunction. Moreover, puerarin notably decreased the mRNA and protein levels of TNF-α, IL-6, and IL-1ß, indicating that puerarin attenuated MI/R-induced inflammation. Furthermore, puerarin markedly decreased the protein levels of Ac-NF-κB, NLRP3, cleaved caspase-1, cleaved IL-1ß, and cleaved IL-18 and increased the protein level of SIRT1. More importantly, the SIRT1 inhibitor nicotinamide prevented these puerarin-induced cardioprotective effects and regulation of the SIRT1/NF-κB pathway, as well as the NLRP3 inflammasome activation. CONCLUSION: Puerarin protected against MI/R injury by inhibiting inflammatory responses probably via the SIRT1/NF-κB pathway, and inhibition of the NLRP3 inflammasome was also involved in puerarin-induced cardioprotective effects. These results suggest that puerarin may be a novel candidate for the treatment of ischemic heart disease.


Subject(s)
Cardiotonic Agents/pharmacology , Inflammation/metabolism , Isoflavones/pharmacology , Myocardial Reperfusion Injury/prevention & control , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Signal Transduction/drug effects , Animals , Cardiotonic Agents/administration & dosage , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacology , Heart/drug effects , Inflammasomes/antagonists & inhibitors , Inflammasomes/drug effects , Inflammation/etiology , Male , Mice, Inbred C57BL , Myocardial Reperfusion Injury/blood , Myocardial Reperfusion Injury/complications , Myocardial Reperfusion Injury/pathology , Myocardium/metabolism , Myocardium/pathology , NF-kappa B p50 Subunit/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Niacinamide/administration & dosage , Niacinamide/pharmacology , Sirtuin 1/metabolism
18.
Expert Opin Pharmacother ; 21(15): 1851-1865, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32659185

ABSTRACT

INTRODUCTION: Achieving reperfusion immediately after acute myocardial infarction improves outcomes; despite this, patients remain at a high risk for mortality and morbidity at least for the first year after the event. Ischemia-reperfusion injury (IRI) has a complex pathophysiology and plays an important role in myocardial tissue injury, repair, and remodeling. AREAS COVERED: In this review, the authors discuss the various mechanisms and their pharmacological agents currently available for reducing myocardial ischemia-reperfusion injury (IRI). They review important original investigations and trials in various clinical databases for treatments targeting IRI. EXPERT OPINION: Encouraging results observed in many preclinical studies failed to show similar success in attenuating myocardial IRI in large-scale clinical trials. Identification of critical risk factors for IRI and targeting them individually rather than one size fits all approach should be the major focus of future research. Various newer therapies like tocilizumab, anakinra, colchicine, revacept, and therapies targeting the reperfusion injury salvage kinase pathway, survivor activating factor enhancement, mitochondrial pathways, and angiopoietin-like peptide 4 hold promise for the future.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Cardiotonic Agents/therapeutic use , Myocardial Infarction/drug therapy , Myocardial Reperfusion Injury/drug therapy , Platelet Aggregation Inhibitors/therapeutic use , Animals , Anti-Inflammatory Agents/administration & dosage , Cardiotonic Agents/administration & dosage , Humans , Molecular Targeted Therapy , Myocardial Infarction/blood , Myocardial Infarction/complications , Myocardial Infarction/immunology , Myocardial Reperfusion Injury/blood , Myocardial Reperfusion Injury/etiology , Myocardial Reperfusion Injury/immunology , Platelet Aggregation Inhibitors/administration & dosage
19.
Biochem Pharmacol ; 178: 114099, 2020 08.
Article in English | MEDLINE | ID: mdl-32540483

ABSTRACT

There is some recent evidence that cardiac ischemia/reperfusion (I/R) injury induces intestinal damage within days, which contributes to adverse cardiovascular outcomes after myocardial infarction. However, it is not clear whether remote gut injury has any detectable early signs, and whether different interventions aiming to reduce cardiac damage are also effective at protecting the intestine. Previously, we found that chronic treatment with rofecoxib, a selective inhibitor of cyclooxygenase-2 (COX-2), limited myocardial infarct size to a comparable extent as cardiac ischemic preconditioning (IPC) in rats subjected to 30-min coronary artery occlusion and 120-min reperfusion. In the present study, we aimed to analyse the early intestinal alterations caused by cardiac I/R injury, with or without the above-mentioned infart size-limiting interventions. We found that cardiac I/R injury induced histological changes in the small intestine within 2 h, which were accompanied by elevated tissue level of COX-2 and showed positive correlation with the activity of matrix metalloproteinase-2 (MMP-2), but not of MMP-9 in the plasma. All these changes were prevented by rofecoxib treatment. By contrast, cardiac IPC failed to reduce intestinal injury and plasma MMP-2 activity, although it prevented the transient reduction in jejunal blood flow in response to cardiac I/R. Our results demonstrate for the first time that rapid development of intestinal damage follows cardiac I/R, and that two similarly effective infarct size-limiting interventions, rofecoxib treatment and cardiac IPC, have different impacts on cardiac I/R-induced gut injury. Furthermore, intestinal damage correlates with plasma MMP-2 activity, which may be a biomarker for its early diagnosis.


Subject(s)
Cardiotonic Agents/pharmacology , Cyclooxygenase 2 Inhibitors/pharmacology , Cyclooxygenase 2/genetics , Intestine, Small/drug effects , Lactones/pharmacology , Matrix Metalloproteinase 2/genetics , Myocardial Reperfusion Injury/prevention & control , Sulfones/pharmacology , Animals , Biomarkers/blood , Coronary Occlusion/surgery , Coronary Vessels/surgery , Cyclooxygenase 2/blood , Disease Models, Animal , Drug Administration Schedule , Gene Expression , Intestine, Small/pathology , Ischemic Preconditioning/methods , Male , Matrix Metalloproteinase 2/blood , Matrix Metalloproteinase 9/blood , Matrix Metalloproteinase 9/genetics , Myocardial Reperfusion Injury/blood , Myocardial Reperfusion Injury/diagnosis , Myocardial Reperfusion Injury/genetics , Myocardium/enzymology , Myocardium/pathology , Rats , Rats, Wistar
20.
Am J Physiol Heart Circ Physiol ; 319(1): H32-H41, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32412785

ABSTRACT

Disruption of the normal gut microbiome (dysbiosis) is implicated in the progression and severity of myriad disorders, including hypercholesterolemia and cardiovascular disease. Probiotics attenuate and reverse gut dysbiosis to improve cardiovascular risk factors like hypertension and hypercholesterolemia. Lactobacillus reuteri is a well-studied lactic acid-producing probiotic with known cholesterol-lowering properties and anti-inflammatory effects. In the present study, we hypothesized that L. reuteri delivered to hypercholesterolemic low-density lipoprotein receptor knockout (LDLr KO) mice will reduce cholesterol levels and minimize cardiac injury from an ischemic insult. L. reuteri [1 × 109 or 50 × 106 colony-forming units (CFU)/day] was administered by oral gavage to wild-type mice and LDLr KO for up to 6 wk followed by an ischemia-reperfusion (I/R) protocol. After 4 wk of gavage, total serum cholesterol in wild-type mice receiving saline was 113.5 ± 5.6 mg/dL compared with 113.3 ± 6.8 and 101.9 ± 7.5 mg/dL in mice receiving 1 × 109 or 50 × 106 CFU/day, respectively. Over the same time frame, administration of L. reuteri at 1 × 109 or 50 × 106 CFU/day did not lower total serum cholesterol (283.0 ± 11.1, 263.3 ± 5.0, and 253.1 ± 7.0 mg/dL; saline, 1 × 109 or 50 × 106 CFU/day, respectively) in LDLr KO mice. Despite no impact on total serum cholesterol, L. reuteri administration significantly attenuated cardiac injury following I/R, as evidenced by smaller infarct sizes compared with controls in both wild-type and LDLr KO groups. In conclusion, daily L. reuteri significantly protected against cardiac injury without lowering cholesterol levels, suggesting anti-inflammatory properties of L. reuteri uncoupled from improvements in serum cholesterol.NEW & NOTEWORTHY We demonstrated that daily delivery of Lactobacillus reuteri to wild-type and hypercholesterolemic lipoprotein receptor knockout mice attenuated cardiac injury following ischemia-reperfusion without lowering total serum cholesterol in the short term. In addition, we validated protection against cardiac injury using histology and immunohistochemistry techniques. L. reuteri offers promise as a probiotic to mitigate ischemic cardiac injury.


Subject(s)
Cholesterol/blood , Myocardial Reperfusion Injury/microbiology , Probiotics/therapeutic use , Animals , Gastrointestinal Microbiome , Limosilactobacillus reuteri/pathogenicity , Male , Mice , Mice, Inbred C57BL , Myocardial Reperfusion Injury/blood , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/prevention & control , Probiotics/administration & dosage , Receptors, LDL/deficiency , Receptors, LDL/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...