Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 445
1.
Scand Cardiovasc J ; 58(1): 2347290, 2024 Dec.
Article En | MEDLINE | ID: mdl-38733316

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.


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.
Int J Mol Sci ; 23(5)2022 Mar 01.
Article En | MEDLINE | ID: mdl-35269870

Recent knowledge concerning the role of non-coding RNAs (ncRNAs) in myocardial ischemia/reperfusion (I/R) injury provides new insight into their possible roles as specific biomarkers for early diagnosis, prognosis, and treatment. MicroRNAs (miRNAs) have fewer than 200 nucleotides, while long ncRNAs (lncRNAs) have more than 200 nucleotides. The three types of ncRNAs (miRNAs, lncRNAs, and circRNAs) act as signaling molecules strongly involved in cardiovascular disorders (CVD). I/R injury of the heart is the main CVD correlated with acute myocardial infarction (AMI), cardiac surgery, and transplantation. The expression levels of many ncRNAs and miRNAs are highly modified in the plasma of MI patients, and thus they have the potential to diagnose and treat MI. Cardiomyocyte and endothelial cell death is the major trigger for myocardial ischemia-reperfusion syndrome (MIRS). The cardioprotective effect of inflammasome activation in MIRS and the therapeutics targeting the reparative response could prevent progressive post-infarction heart failure. Moreover, the pharmacological and genetic modulation of these ncRNAs has the therapeutic potential to improve clinical outcomes in AMI patients.


MicroRNAs , Myocardial Infarction , Myocardial Reperfusion Injury , RNA, Long Noncoding , Humans , MicroRNAs/metabolism , Myocardial Infarction/diagnosis , Myocardial Infarction/genetics , Myocardial Infarction/therapy , Myocardial Reperfusion Injury/diagnosis , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/prevention & control , Nucleotides , RNA, Long Noncoding/genetics , RNA, Untranslated/genetics , RNA, Untranslated/metabolism
3.
Can J Physiol Pharmacol ; 100(3): 252-258, 2022 Mar.
Article En | MEDLINE | ID: mdl-34990309

This study was designed to assess the effect of soya phosphatidylcholine (SPC) against ischemia/reperfusion (I/R) injury and the possible underlying mechanism using experimental and computational studies. I/R injury was induced by global ischemia for 30 min followed by reperfusion for 120 min. The perfusion of the SPC was performed for 10 min before inducing global ischemia. In the mechanistic study, the involvement of specific cellular pathways was identified using various inhibitors such as ATP-dependent potassium channel (KATP) inhibitor (glibenclamide), protein kinase C (PKC) inhibitor (chelerythrine), non-selective nitric oxide synthase inhibitor (L-NAME), and endothelium remover (Triton X-100). The computational study of various ligands was performed on toll-like receptor 4 (TLR4) protein using AutoDock version 4.0. SPC (100 µM) significantly decreased the levels of cardiac damage markers and %infarction compared with the vehicle control (VC). Furthermore, cardiodynamics (indices of left ventricular contraction (dp/dtmax), indices of left ventricular relaxation (dp/dtmin), coronary flow, and antioxidant enzyme levels were significantly improved as compared with VC. This protective effect was attenuated by glibenclamide, chelerythrine, and Triton X-100, but it was not attenuated by L-NAME. The computational study showed a significant bonding affinity of SPC to the TLR4-MD2 complex. Thus, SPC reduced myocardial I/R injury in isolated perfused rat hearts, which might be governed by the KATP channel, PKC, endothelium response, and TLR4-MyD88 signaling pathway.


Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/etiology , Phosphatidylcholines/therapeutic use , Animals , Cardiotonic Agents , Computer Simulation , In Vitro Techniques , Male , Myocardial Reperfusion Injury/diagnosis , Myocardial Reperfusion Injury/physiopathology , Phosphatidylcholines/administration & dosage , Phosphatidylcholines/pharmacology , Rats, Wistar , Toll-Like Receptor 4
4.
J Cell Mol Med ; 25(21): 10224-10235, 2021 11.
Article En | MEDLINE | ID: mdl-34601814

Cardiomyocyte apoptosis is the main reason of cardiac injury after myocardial ischaemia-reperfusion (I/R) injury (MIRI), but the role of p300/CBP-associated factor (PCAF) on myocardial apoptosis in MIRI is unknown. The aim of this study was to investigate the main mechanism of PCAF modulating cardiomyocyte apoptosis in MIRI. The MIRI model was constructed by ligation of the rat left anterior descending coronary vessel for 30 min and reperfusion for 24 h in vivo. H9c2 cells were harvested after induced by hypoxia for 6 h and then reoxygenation for 24 h (H/R) in vitro. The RNA interference PCAF expression adenovirus was transfected into rat myocardium and H9c2 cells. The area of myocardial infarction, cardiac function, myocardial injury marker levels, apoptosis, inflammation and oxidative stress were detected respectively. Both I/R and H/R remarkably upregulated the expression of PCAF, and downregulation of PCAF significantly attenuated myocardial apoptosis, inflammation and oxidative stress caused by I/R and H/R. In addition, downregulation of PCAF inhibited the activation of NF-κB signalling pathway in cardiomyocytes undergoing H/R. Pretreatment of lipopolysaccharide, a NF-κB pathway activator, could blunt these protective effects of PCAF downregulation on myocardial apoptosis in MIRI. These results highlight that downregulation of PCAF could reduce cardiomyocyte apoptosis by inhibiting the NF-κB pathway, thereby providing protection for MIRI. Therefore, PCAF might be a promising target for protecting against cardiac dysfunction induced by MIRI.


Apoptosis/genetics , Myocardial Reperfusion Injury/etiology , Myocardial Reperfusion Injury/metabolism , Myocytes, Cardiac/metabolism , NF-kappa B/metabolism , Signal Transduction , p300-CBP Transcription Factors/genetics , Animals , Biomarkers , Cell Line , Disease Models, Animal , Disease Susceptibility , Down-Regulation , Gene Expression Regulation , Heart Function Tests , Myocardial Reperfusion Injury/diagnosis , Oxidative Stress , Rats , p300-CBP Transcription Factors/metabolism
5.
J Cell Mol Med ; 25(21): 9939-9952, 2021 11.
Article En | MEDLINE | ID: mdl-34581005

Interleukin (IL)-7 is known to enhance the macrophages cytotoxic activity and that macrophages play a pivotal role in the development and progression of myocardial ischaemia/reperfusion (I/R) injury. However, the effects of IL-7 on macrophages infiltration and polarization in myocardial I/R injury are currently unclear. This study aimed to evaluate the effects of the IL-7 expression on myocardial I/R injury and their relationship with macrophages. The data showed that IL-7 expression in mouse heart tissue increases following I/R injury and that IL-7 knockout or anti-IL-7 antibody treatment significantly improve I/R injury, including reduction in myocardial infarction area, a serum troponin T level decreases and an improvement in cardiac function. On the other hand, recombinant IL-7 (rIL-7) supplementation induces opposite effects and the anti-IL-7 antibody significantly reduces the cardiomyocyte apoptosis and macrophage infiltration. rIL-7 cannot directly cause apoptosis, but it can induce cardiomyocyte apoptosis through macrophages, in addition to increase the macrophages migration in vitro. Anti-IL-7 antibody affects the cytokine production in T helper (Th) 1 and Th2 cells and also promotes the macrophages differentiation to M2 macrophages. However, anti-IL-7 antibody does not reduce the M1 macrophage number, and it only increases the ratio of M2/M1 macrophages in mice heart tissues after I/R injury. Taking together, these data reveal that IL-7 plays an intensifying role in myocardial I/R injury by promoting cardiomyocyte apoptosis through the regulation of macrophage infiltration and polarization.


Interleukin-7/metabolism , Macrophage Activation/immunology , Macrophages/immunology , Macrophages/metabolism , Myocardial Reperfusion Injury/etiology , Myocardial Reperfusion Injury/metabolism , Animals , Apoptosis/genetics , Biomarkers , Disease Models, Animal , Disease Susceptibility , Gene Expression , Heart Function Tests , Hemodynamics , Immunophenotyping , Interleukin-7/genetics , Macrophages/pathology , Mice , Mice, Knockout , Myocardial Reperfusion Injury/diagnosis , Myocytes, Cardiac/metabolism
7.
Int J Mol Sci ; 22(16)2021 Aug 10.
Article En | MEDLINE | ID: mdl-34445301

Angiotensin receptor neprilysin inhibitor (ARNI) treatment reduces functional mitral regurgitation (MR) to a greater extent than angiotensin receptor blocker (ARB) treatment alone, but the mechanism is unclear. We evaluated the mechanisms of how ARNI has an effect on functional MR. After inducing functional MR by left circumflex coronary artery occlusion, male Sprague Dawley rats (n = 31) were randomly assigned to receive the ARNI LCZ696, the ARB valsartan, or corn oil only (MR control). Excised mitral leaflets and left ventricle (LV) were analyzed, and valvular endothelial cells were evaluated focusing on molecular changes. LCZ696 significantly attenuated LV dilatation after 6 weeks when compared with the control group (LV end-diastolic volume, 461.3 ± 13.8 µL versus 525.1 ± 23.6 µL; p < 0.05), while valsartan did not (471.2 ± 8.9 µL; p > 0.05 to control). Histopathological analysis of mitral leaflets showed that LCZ696 strongly reduced fibrotic thickness compared to the control group (28.2 ± 2.7 µm vs. 48.8 ± 7.5 µm; p < 0.05). Transforming growth factor-ß and downstream phosphorylated extracellular-signal regulated kinase were also significantly lower in the LCZ696 group. Consequently, excessive endothelial-to-mesenchymal transition (EndoMT) was mitigated in the LCZ696 group compared to the control group and leaflet area was higher (11%) in the LCZ696 group than in the valsartan group. Finally, the MR extent was significantly lower in the LCZ696 group and functional improvement was observed. In conclusion, neprilysin inhibitor has positive effects on LV reverse remodeling and also attenuates fibrosis in MV leaflets and restores adaptive growth by directly modulating EndoMT.


Aminobutyrates/therapeutic use , Biphenyl Compounds/therapeutic use , Mitral Valve Insufficiency/drug therapy , Myocardial Infarction/drug therapy , Valsartan/therapeutic use , Aminobutyrates/pharmacology , Angiotensin Receptor Antagonists/pharmacology , Angiotensin Receptor Antagonists/therapeutic use , Animals , Biphenyl Compounds/pharmacology , Cells, Cultured , Drug Combinations , Endothelial Cells/drug effects , Endothelial Cells/physiology , Heart Failure/diagnosis , Heart Failure/drug therapy , Heart Failure/etiology , Heart Failure/physiopathology , Humans , Male , Mitral Valve/drug effects , Mitral Valve/pathology , Mitral Valve/physiology , Mitral Valve Insufficiency/diagnosis , Mitral Valve Insufficiency/etiology , Mitral Valve Insufficiency/physiopathology , Myocardial Infarction/complications , Myocardial Infarction/diagnosis , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/complications , Myocardial Reperfusion Injury/diagnosis , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/physiopathology , Neprilysin/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Valsartan/pharmacology , Ventricular Function, Left/drug effects , Ventricular Remodeling/drug effects
8.
Int J Med Sci ; 18(14): 3318-3325, 2021.
Article En | MEDLINE | ID: mdl-34400901

Purpose: Hydrogen (H2) is an antioxidant with anti-inflammatory and apoptosis functions.This study aimed to estimate the effects of H2 on acute myocardial infarction (AMI) in rats and its association with the inhibition of oxidative stress and cardiomyocyte pyroptosis. Methods: Sixty-four rats were randomly divided into three groups (Sham, AMI, and H2). The left anterior descending coronary artery (LAD) of rats in the AMI and H2 groups was ligated, while rats in the Sham group were threaded without ligation. In addition, 2% H2 was administered by inhalation for 24 h after ligation in the H2 group. Transthoracic echocardiography was performed after H2 inhalation, followed by collection of the serum and cardiac tissue of all rats. Results: H2 inhalation ameliorated the cardiac dysfunction, infarct size and inflammatory cell infiltration caused by AMI. Meanwhile, H2 inhalation reduced the concentration of serum Troponin I (TnI), brain natriuretic peptide (BNP), reactive oxygen species (ROS), cardiac malondialdehyde (MDA), and 8-OHdG. In addition, H2 inhalation inhibited cardiac inflammation and pyroptosis relative proteins expression. Conclusion: H2 effectively promoted heart functions in AMI rats by regulating oxidative stress and pyroptosis.


Antioxidants/administration & dosage , Hydrogen/administration & dosage , Myocardial Infarction/drug therapy , Myocardial Reperfusion Injury/prevention & control , Administration, Inhalation , Animals , Disease Models, Animal , Echocardiography , Humans , Inflammasomes/antagonists & inhibitors , Inflammasomes/metabolism , Male , Myocardial Infarction/complications , Myocardial Infarction/immunology , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/diagnosis , Myocardial Reperfusion Injury/immunology , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/immunology , Myocytes, Cardiac/pathology , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Oxidative Stress/drug effects , Oxidative Stress/immunology , Pyroptosis/drug effects , Pyroptosis/immunology , Rats , Reactive Oxygen Species/metabolism
9.
Curr Hypertens Rev ; 17(2): 121-130, 2021.
Article En | MEDLINE | ID: mdl-34225634

AIMS: Non-invasive indices to evaluate left ventricular changes during ischemic heart failure are needed to quantify the myocardial impairment and the effectiveness of therapeutic manoeuvres. The aims of this work were to calculate the Wall Thickening Fraction (WTF) and the Augmentation Index (AIx) and to assess the relationship between WTF and AIx using data obtained from an animal model with heart failure followed by a myocardial ischemia stage and a reperfusion stage. METHODS: Nine Corriedale sheep that had been monitored for 10 minutes during a basal stage underwent 5-minute myocardial ischemia, followed by 60-minute reperfusion. Seven of them were subjected to an induced heart failure through an overdose of halothane, two of which were treated with intra-aortic counterpulsation during the reperfusion stage. The remaining two animals were monitored during their ischemia-reperfusion stage. RESULTS: Data obtained in the 5 animals suffering from heart failure followed by myocardial ischemia showed that: a) heart failure induction determined decrease in cardiac output, cardiac index and systolic and diastolic aortic pressure (AoP) with respect to their basal values (p<0.05), b) myocardial ischemia decreased the WTF compared with basal and induced heart failure values (p<0.05), c) during the reperfusion stage accompanied by induced heart failure, WTF increased with respect to values observed during the ischemia induction stage (p<0.05); nevertheless, basal values were not recovered after reperfusion (p<0.05). During this 60-minute stage, systolic and diastolic AoP values were lower (p<0.05) than those at the basal stage. CONCLUSION: AIx and WTF values calculated from synchronically recorded values of aortic pressure and left ventricular wall thickness during the reperfusion stage in all animals (n = 9) showed a negative correlation (p<0.05). Analysed data provided evidence of a negative relationship between a left ventricular index of myocardial function and an arterial index obtained from AoP waves.


Heart Failure , Hypotension , Myocardial Ischemia , Myocardial Reperfusion Injury , Animals , Disease Models, Animal , Heart Failure/diagnosis , Ischemia , Myocardial Ischemia/diagnosis , Myocardial Reperfusion Injury/diagnosis , Reperfusion , Sheep , Ventricular Function, Left
10.
Int J Mol Sci ; 22(11)2021 May 27.
Article En | MEDLINE | ID: mdl-34072153

Preclinical studies have shown that postconditioning with hydrogen sulfide (H2S) exerts cardioprotective effects against myocardial ischemia-reperfusion injury (IRI). The aim of this study was to appraise the current evidence of the cardioprotective effects of H2S against IRI in order to explore the future implementation of H2S in clinical cardiac transplantation. The current literature on H2S postconditioning in the setting of global myocardial ischemia was systematically reviewed and analyzed, performing meta-analyses. A literature search of the electronic databases Medline, Embase and Cinahl identified 1835 studies that were subjected to our pre-defined inclusion criteria. Sixteen studies were considered eligible for inclusion. Postconditioning with H2S showed significant robust effects with regard to limiting infarct size (standardized mean difference (SMD) = -4.12, 95% CI [-5.53--2.71], p < 0.00001). Furthermore, H2S postconditioning consistently resulted in a significantly lower release of cardiac injury markers, lower levels of oxidative stress and improved cardiac function. Postconditioning with slow-releasing H2S donors offers a valuable opportunity for novel therapies within cardiac preservation for transplantation. Before clinical implication, studies evaluating the long-term effects of H2S treatment and effects of H2S treatment in large animal studies are warranted.


Heart , Hydrogen Sulfide/pharmacology , Organ Preservation Solutions , Organ Preservation , Animals , Biomarkers , Cryopreservation/methods , Heart Function Tests , Heart Transplantation/methods , Humans , Ischemic Postconditioning , Myocardial Reperfusion Injury/diagnosis , Myocardial Reperfusion Injury/etiology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/prevention & control , Organ Preservation/methods , Oxidative Stress/drug effects , Risk Factors , Tissue Donors
11.
Pharmacol Res ; 170: 105715, 2021 08.
Article En | MEDLINE | ID: mdl-34111564

The relevance of extracellular vesicles (EV) as mediators of cardiac damage or recovery upon Ischemia Reperfusion Injury (IRI) and Remote Ischemic PreConditioning (RIPC) is controversial. This study aimed to investigate whether serum-derived EV, recovered from patients with Acute Coronary Syndrome (ACS) and subjected to the RIPC or sham procedures, may be a suitable therapeutic approach to prevent IRI during Percutaneous-Coronary-Intervention (PCI). A double-blind, randomized, sham-controlled study (NCT02195726) has been extended, and EV were recovered from 30 patients who were randomly assigned (1:1) to undergo the RIPC- (EV-RIPC) or sham-procedures (EV-naive) before PCI. Patient-derived EV were analyzed by TEM, FACS and western blot. We found that troponin (TnT) was enriched in EV, compared to healthy subjects, regardless of diagnosis. EV-naive induced protection against IRI, both in-vitro and in the rat heart, unlike EV-RIPC. We noticed that EV-naive led to STAT-3 phosphorylation, while EV-RIPC to Erk-1/2 activation in the rat heart. Pre-treatment of the rat heart with specific STAT-3 and Erk-1/2 inhibitors led us to demonstrate that STAT-3 is crucial for EV-naive-mediated protection. In the same model, Erk-1/2 inhibition rescued STAT-3 activation and protection upon EV-RIPC treatment. 84 Human Cardiovascular Disease mRNAs were screened and DUSP6 mRNA was found enriched in patient-derived EV-naive. Indeed, DUSP6 silencing in EV-naive prevented STAT-3 phosphorylation and cardio-protection in the rat heart. This analysis of ACS-patients' EV proved: (i) EV-naive cardio-protective activity and mechanism of action; (ii) the lack of EV-RIPC-mediated cardio-protection; (iii) the properness of the in-vitro assay to predict EV effectiveness in-vivo.


Acute Coronary Syndrome/therapy , Arm/blood supply , Extracellular Vesicles/transplantation , Ischemic Preconditioning, Myocardial , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/metabolism , Percutaneous Coronary Intervention , Acute Coronary Syndrome/blood , Acute Coronary Syndrome/diagnosis , Aged , Animals , Case-Control Studies , Cell Line , Disease Models, Animal , Double-Blind Method , Dual Specificity Phosphatase 6/metabolism , Endothelial Cells/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Extracellular Vesicles/metabolism , Female , Humans , Ischemic Preconditioning, Myocardial/adverse effects , Male , Middle Aged , Myocardial Reperfusion Injury/diagnosis , Myocardial Reperfusion Injury/etiology , Myocardial Reperfusion Injury/metabolism , Myocytes, Cardiac/pathology , Percutaneous Coronary Intervention/adverse effects , Phosphorylation , Rats, Wistar , Regional Blood Flow , STAT3 Transcription Factor/metabolism , Time Factors , Treatment Outcome
12.
Heart Surg Forum ; 24(2): E409-E413, 2021 Apr 27.
Article En | MEDLINE | ID: mdl-33973512

BACKGROUND: To investigate the potential of intravenously administered porcine recombinant interferon-ß1a (IFN-ß1a) for myocardial protection during acute ischemia-reperfusion (IR) injury in an experimental animal model. METHODS: Twenty-two piglets (mean ± standard deviation, 26.7 ± 1.65 kg) were assigned to either the IFN group (n = 12) or the control group (n = 10). IR injury was induced by occluding the distal left descending coronary artery for 30 minutes, with a reperfusion period of 6 h. In the IFN group, the animals received 12.5 µg IFN-ß1a intravenously repeatedly; the control group received saline solution. The levels of interleukin-6 (IL-6) and cardiac troponin I (TnI) were measured, and the amount of myocardial damage was quantified by analyzing myocardial apoptosis and the mean fluorescence intensity (MFI) of methylene blue-stained cardiac tissue. RESULTS: In the IFN group, significantly more premature deaths occurred compared with the control group (25% versus 17%, P = .013). Between the groups, the mean heart rate was higher in the IFN group (102 ± 22 versus 80 ± 20 beats per minute, P = .02). IL-6 and TnI levels were comparable between the groups, with no significant difference, and there was no difference between the study groups in myocardial apoptosis in the infarcted myocardium. The percentage of MFI differed significantly between the IFN and control groups (90.75% ± 4.90% versus 96.02% ± 2.73%, P = .01). CONCLUSION: In this acute IR injury animal model, IFN-ß1a did not protect the myocardium from IR injury, but rather increased some of the unfavorable outcomes studied.


Interferon beta-1a/administration & dosage , Myocardial Infarction/complications , Myocardial Reperfusion Injury/drug therapy , Myocardium/pathology , Adjuvants, Immunologic/administration & dosage , Animals , Apoptosis , Disease Models, Animal , Injections, Intravenous , Myocardial Infarction/diagnosis , Myocardial Reperfusion Injury/diagnosis , Myocardial Reperfusion Injury/etiology , Swine
14.
Circulation ; 144(4): 303-317, 2021 07 27.
Article En | MEDLINE | ID: mdl-34015936

BACKGROUND: The benefits of exercise training in the cardiovascular system have been well accepted; however, the underlying mechanism remains to be explored. Here, we report the initial functional characterization of an exercise-induced cardiac physiological hypertrophy-associated novel long noncoding RNA (lncRNA). METHODS: Using lncRNA microarray profiling, we identified lncRNAs in contributing the modulation of exercise-induced cardiac growth that we termed cardiac physiological hypertrophy-associated regulator (CPhar). Mice with adeno-associated virus serotype 9 driving CPhar overexpression and knockdown were used in in vivo experiments. Swim training was used to induce physiological cardiac hypertrophy in mice, and ischemia reperfusion injury surgery was conducted to investigate the protective effects of CPhar in mice. To investigate the mechanisms of CPhar's function, we performed various analyses including quantitative reverse transcription polymerase chain reaction, Western blot, histology, cardiac function (by echocardiography), functional rescue experiments, mass spectrometry, in vitro RNA transcription, RNA pulldown, RNA immunoprecipitation, chromatin immunoprecipitation assay, luciferase reporter assay, and coimmunoprecipitation assays. RESULTS: We screened the lncRNAs in contributing the modulation of exercise-induced cardiac growth through lncRNA microarray profiling and found that CPhar was increased with exercise and was necessary for exercise-induced physiological cardiac growth. The gain and loss of function of CPhar regulated the expression of proliferation markers, hypertrophy, and apoptosis in cultured neonatal mouse cardiomyocytes. Overexpression of CPhar prevented myocardial ischemia reperfusion injury and cardiac dysfunction in vivo. We identified DDX17 (DEAD-Box Helicase 17) as a binding partner of CPhar in regulating CPhar downstream factor ATF7 (activating transcription factor 7) by sequestering C/EBPß (CCAAT/enhancer binding protein beta). CONCLUSIONS: Our study of this lncRNA CPhar provides new insights into the regulation of exercise-induced cardiac physiological growth, demonstrating the cardioprotective role of CPhar in the heart, and expanding our mechanistic understanding of lncRNA function, as well.


Biomarkers , Cardiomegaly/etiology , Endurance Training/adverse effects , Myocardial Reperfusion Injury/etiology , Myocytes, Cardiac/metabolism , RNA, Long Noncoding/genetics , Recovery of Function/genetics , Activating Transcription Factors/genetics , Activating Transcription Factors/metabolism , Animals , Apoptosis , CCAAT-Enhancer-Binding Protein-beta/genetics , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cardiomegaly/diagnosis , Disease Models, Animal , Echocardiography , Gene Expression Profiling , Mice , Myocardial Reperfusion Injury/diagnosis , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/physiopathology
15.
Medicine (Baltimore) ; 100(10): e24944, 2021 Mar 12.
Article En | MEDLINE | ID: mdl-33725857

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.


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
16.
Int J Mol Sci ; 22(3)2021 Jan 30.
Article En | MEDLINE | ID: mdl-33573240

Circular RNAs (circRNAs) are crucial in gene regulatory networks and disease development, yet circRNA expression in myocardial infarction (MI) is poorly understood. Here, we harvested myocardium samples from domestic pigs 3 days after closed-chest reperfused MI or sham surgery. Cardiac circRNAs were identified by RNA-sequencing of rRNA-depleted RNA from infarcted and healthy myocardium tissue samples. Bioinformatics analysis was performed using the CIRIfull and KNIFE algorithms, and circRNAs identified with both algorithms were subjected to differential expression (DE) analysis and validation by qPCR. Circ-RCAN2 and circ-C12orf29 expressions were significantly downregulated in infarcted tissue compared to healthy pig heart. Sanger sequencing was performed to identify the backsplice junctions of circular transcripts. Finally, we compared the expressions of circ-C12orf29 and circ-RCAN2 between porcine cardiac progenitor cells (pCPCs) that were incubated in a hypoxia chamber for different time periods versus normoxic pCPCs. Circ-C12orf29 did not show significant DE in vitro, whereas circ-RCAN2 exhibited significant ischemia-time-dependent upregulation in hypoxic pCPCs. Overall, our results revealed novel cardiac circRNAs with DE patterns in pCPCs, and in infarcted and healthy myocardium. Circ-RCAN2 exhibited differential regulation by myocardial infarction in vivo and by hypoxia in vitro. These results will improve our understanding of circRNA regulation during acute MI.


Gene Regulatory Networks , Myoblasts, Cardiac/pathology , Myocardial Infarction/complications , Myocardial Reperfusion Injury/genetics , RNA, Circular/metabolism , Animals , Cell Hypoxia/genetics , Computational Biology , Coronary Angiography , Disease Models, Animal , Down-Regulation , Female , Humans , Myoblasts, Cardiac/metabolism , Myocardial Infarction/diagnosis , Myocardial Infarction/genetics , Myocardial Reperfusion Injury/diagnosis , Myocardial Reperfusion Injury/pathology , Myocardium/pathology , RNA-Seq , Sus scrofa , Up-Regulation
17.
Angiology ; 72(6): 511-523, 2021 07.
Article En | MEDLINE | ID: mdl-33472383

We systematically searched the literature to assess the efficacy of trimetazidine in reducing periprocedural myocardial injury and improving postoperative left ventricular ejection fraction (LVEF) in patients with coronary artery disease (CAD) undergoing percutaneous coronary intervention (PCI). An electronic search was conducted based on the PubMed, Ovid, Scopus, Springer, CENTRAL, and Google Scholar databases; 14 randomized controlled trials (RCTs) were included. Our meta-analysis showed a significant reduction in cardiac troponin I (cTnI) levels with trimetazidine compared with controls (P < .00001) but not in serum creatine kinase-myocardial band levels (P = .49). There were significantly reduced odds of ischemic ST-T segment changes with trimetazidine (P = .0.03) but lack of significant difference in the incidence of anginal attacks between the 2 groups (P = .10). Results also suggest significantly higher LVEF with trimetazidine compared with controls (P < .00001). Meta-regression analysis indicated no influence of duration of trimetazidine therapy on cTnI levels. The administration of preprocedure trimetazidine may have a role in reducing periprocedural myocardial injury in patients with CAD undergoing PCI. Evidence also suggests that postoperative trimetazidine may improve LVEF in the short term. Lack of high-quality trials and the heterogeneity of studies limit the ability of our analysis to draw strong conclusions. Further well-designed RCTs are required to supplement current evidence.


Cardiovascular Agents/therapeutic use , Coronary Artery Disease/therapy , Myocardial Reperfusion Injury/prevention & control , Myocardium/pathology , Percutaneous Coronary Intervention , Stroke Volume/drug effects , Trimetazidine/therapeutic use , Ventricular Function, Left/drug effects , Biomarkers/blood , Cardiovascular Agents/adverse effects , Coronary Artery Disease/diagnostic imaging , Coronary Artery Disease/physiopathology , Female , Humans , Male , Middle Aged , Myocardial Reperfusion Injury/diagnosis , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocardium/metabolism , Percutaneous Coronary Intervention/adverse effects , Randomized Controlled Trials as Topic , Recovery of Function , Risk Factors , Treatment Outcome , Trimetazidine/adverse effects , Troponin I/blood
18.
Life Sci ; 260: 118400, 2020 Nov 01.
Article En | MEDLINE | ID: mdl-32918975

Clinical manifestations of COVID-19 affect many organs, including the heart. Cardiovascular disease is a dominant comorbidity and prognostic factors predicting risk for critical courses are highly needed. Moreover, immunomechanisms underlying COVID-induced myocardial damage are poorly understood. OBJECTIVE: To elucidate prognostic markers to identify patients at risk. RESULTS: Only patients with pericardial effusion (PE) developed a severe disease course, and those who died could be identified by a high CD8/Treg/monocyte ratio. Ten out of 19 COVID-19 patients presented with PE, 7 (78%) of these had elevated APACHE-II mortality risk-score, requiring mechanical ventilation. At admission, PE patients showed signs of systemic and cardiac inflammation in NMR and impaired cardiac function as detected by transthoracic echocardiography (TTE), whereas parameters of myocardial injury e.g. high sensitive troponin-t (hs-TnT) were not yet increased. During the course of disease, hs-TnT rose in 8 of the PE-patients above 16 ng/l, 7 had to undergo ventilatory therapy and 4 of them died. FACS at admission showed in PE patients elevated frequencies of CD3+CD8+ T cells among all CD3+ T-cells, and lower frequencies of Tregs and CD14+HLA-DR+-monocytes. A high CD8/Treg/monocyte ratio predicted a severe disease course in PE patients, and was associated with high serum levels of antiviral cytokines. By contrast, patients without PE and PE patients with a low CD8/Treg/monocyte ratio neither had to be intubated, nor died. CONCLUSIONS: PE predicts cardiac injury in COVID-19 patients. Therefore, TTE should be performed at admission. Immunological parameters for dysfunctional antiviral immunity, such as the CD8/Treg/monocyte ratio used here, supports risk assessment by predicting poor prognosis.


Betacoronavirus/isolation & purification , Biomarkers/analysis , Coronavirus Infections/mortality , Myocardial Reperfusion Injury/diagnosis , Myocardial Reperfusion Injury/mortality , Myocardium/pathology , Pneumonia, Viral/mortality , Risk Assessment/methods , Aged , COVID-19 , Coronavirus Infections/complications , Coronavirus Infections/transmission , Coronavirus Infections/virology , Female , Germany/epidemiology , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Myocardial Reperfusion Injury/epidemiology , Myocardial Reperfusion Injury/virology , Myocardium/metabolism , Pandemics , Pneumonia, Viral/complications , Pneumonia, Viral/transmission , Pneumonia, Viral/virology , Prognosis , Risk Factors , SARS-CoV-2 , Survival Rate
19.
Biochem Pharmacol ; 178: 114099, 2020 08.
Article En | MEDLINE | ID: mdl-32540483

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.


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.
J Cardiovasc Pharmacol Ther ; 25(6): 487-493, 2020 11.
Article En | MEDLINE | ID: mdl-32597205

Translation of the cardioprotective effect by pharmacological and mechanical conditioning therapies into improvement of clinical outcome for the patients has been disappointing. Confounding factors like comorbidity and comedications may explain some of the loss in translation. However, the substantial improvement of outcome in disease states involving ischemia-reperfusion injury, that is, planned cardiac surgery, elective percutaneous coronary intervention, and even primary percutaneous coronary intervention for ST-segment myocardial infarction (STEMI), is the most plausible explanation for the missed demonstration of a clinical benefit. Remote ischemic conditioning has demonstrated consistent cardioprotective effect in experimental and in clinical proof-of-concept studies. As an adjunctive cardioprotective treatment beyond reperfusion, remote ischemic conditioning should address target populations at risk of extensive tissue damage, including patients who experience complications, which may induce profound myocardial ischemia in relation to cardiac surgery or elective percutaneous coronary intervention. Moreover, patients with STEMI and predictable impaired clinical outcome due to delayed hospital admission, high Killip class, cardiogenic shock, and cardiac arrest remain target groups. For high-risk patients, daily remote ischemic conditioning or the corollary of blood flow-restricted exercise may be alternative cardioprotective options during postoperative and post-myocardial infarct rehabilitation.


Coronary Artery Bypass/adverse effects , Ischemic Postconditioning , Ischemic Preconditioning, Myocardial , Myocardial Reperfusion Injury/prevention & control , Percutaneous Coronary Intervention/adverse effects , Animals , Humans , Ischemic Postconditioning/adverse effects , Ischemic Preconditioning, Myocardial/adverse effects , Myocardial Reperfusion Injury/diagnosis , Myocardial Reperfusion Injury/etiology , Protective Factors , Risk Factors , Treatment Outcome
...