Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.985
Filter
1.
Mol Med Rep ; 30(2)2024 Aug.
Article in English | MEDLINE | ID: mdl-38904206

ABSTRACT

Myocardial ischemia/reperfusion injury (MIRI) is a significant challenge in the management of myocardial ischemic disease. Extensive evidence suggests that the macrophage­mediated inflammatory response may play a vital role in MIRI. Mesenchymal stem cells and, in particular, exosomes derived from these cells, may be key mediators of myocardial injury and repair. However, whether exosomes protect the heart by regulating the polarization of macrophages and the exact mechanisms involved are poorly understood. The present study aimed to determine whether exosomes secreted by bone marrow mesenchymal stem cells (BMSC­Exo) harboring miR­25­3p can alter the phenotype of macrophages by affecting the JAK2/STAT3 signaling pathway, which reduces the inflammatory response and protects against MIRI. An in vivo MIRI model was established in rats by ligating the anterior descending region of the left coronary artery for 30 min followed by reperfusion for 120 min, and BMSC­Exo carrying miR­25­3p (BMSC­Exo­25­3p) were administered through tail vein injection. A hypoxia­reoxygenation model of H9C2 cells was established, and the cells were cocultured with BMSC­Exo­25­3p in vitro. The results of the present study demonstrated that BMSC­Exo or BMSC­Exo­25­3p could be taken up by cardiomyocytes in vivo and H9C2 cells in vitro. BMSC­Exo­25­3p demonstrated powerful cardioprotective effects by decreasing the cardiac infarct size, reducing the incidence of malignant arrhythmias and attenuating myocardial enzyme activity, as indicated by lactate dehydrogenase and creatine kinase levels. It induced M1­like macrophage polarization after myocardial ischemia/reperfusion (I/R), as evidenced by the increase in iNOS expression through immunofluorescence staining and upregulation of proinflammatory cytokines through RT­qPCR, such as interleukin­1ß (IL­1ß) and interleukin­6 (IL­6). As hypothesized, BMSC­Exo­25­3p inhibited M1­like macrophage polarization and proinflammatory cytokine expression while promoting M2­like macrophage polarization. Mechanistically, the JAK2/STAT3 signaling pathway was activated after I/R in vivo and in LPS­stimulated macrophages in vitro, and BMSC­Exo­25­3p pretreatment inhibited this activation. The results of the present study indicate that the attenuation of MIRI by BMSC­Exo­25­3p may be related to JAK2/STAT3 signaling pathway inactivation and subsequent inhibition of M1­like macrophage polarization.


Subject(s)
Exosomes , Macrophages , Mesenchymal Stem Cells , MicroRNAs , Myocardial Reperfusion Injury , STAT3 Transcription Factor , Animals , MicroRNAs/genetics , MicroRNAs/metabolism , Exosomes/metabolism , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/prevention & control , Rats , Macrophages/metabolism , Male , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , STAT3 Transcription Factor/metabolism , Janus Kinase 2/metabolism , Signal Transduction , Rats, Sprague-Dawley , Disease Models, Animal , Myocytes, Cardiac/metabolism , Cell Line
2.
Cardiovasc Diabetol ; 23(1): 202, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38867293

ABSTRACT

The specific pathophysiological pathways through which diabetes exacerbates myocardial ischemia/reperfusion (I/R) injury remain unclear; however, dysregulation of immune and inflammatory cells, potentially driven by abnormalities in their number and function due to diabetes, may play a significant role. In the present investigation, we simulated myocardial I/R injury by inducing ischemia through ligation of the left anterior descending coronary artery in mice for 40 min, followed by reperfusion for 24 h. Previous studies have indicated that protein kinase Cß (PKCß) is upregulated under hyperglycemic conditions and is implicated in the development of various diabetic complications. The Y4 RNA fragment is identified as the predominant small RNA component present in the extracellular vesicles of cardio sphere-derived cells (CDCs), exhibiting notable anti-inflammatory properties in the contexts of myocardial infarction and cardiac hypertrophy. Our investigation revealed that the administration of Y4 RNA into the ventricular cavity of db/db mice following myocardial I/R injury markedly enhanced cardiac function. Furthermore, Y4 RNA was observed to facilitate M2 macrophage polarization and interleukin-10 secretion through the suppression of PKCß activation. The mechanism by which Y4 RNA affects PKCß by regulating macrophage activation within the inflammatory environment involves the inhibition of ERK1/2 phosphorylation In our study, the role of PKCß in regulating macrophage polarization during myocardial I/R injury was investigated through the use of PKCß knockout mice. Our findings indicate that PKCß plays a crucial role in modulating the inflammatory response associated with macrophage activation in db/db mice experiencing myocardial I/R, with a notable exacerbation of this response observed upon significant upregulation of PKCß expression. In vitro studies further elucidated the protective mechanism by which Y4 RNA modulates the PKCß/ERK1/2 signaling pathway to induce M2 macrophage activation. Overall, our findings suggest that Y4 RNA plays an anti-inflammatory role in diabetic I/R injury, suggesting a novel therapeutic approach for managing myocardial I/R injury in diabetic individuals.


Subject(s)
Disease Models, Animal , Macrophages , Mice, Inbred C57BL , Myocardial Reperfusion Injury , Protein Kinase C beta , Signal Transduction , Animals , Protein Kinase C beta/metabolism , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/enzymology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/prevention & control , Myocardial Reperfusion Injury/genetics , Macrophages/metabolism , Macrophages/enzymology , Male , Interleukin-10/metabolism , Interleukin-10/genetics , Mice , Diabetic Cardiomyopathies/enzymology , Diabetic Cardiomyopathies/pathology , Diabetic Cardiomyopathies/metabolism , Diabetic Cardiomyopathies/etiology , Diabetic Cardiomyopathies/genetics , Diabetic Cardiomyopathies/physiopathology , Cells, Cultured , Phenotype , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , Myocytes, Cardiac/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Macrophage Activation , Mitogen-Activated Protein Kinase 1/metabolism , Ventricular Function, Left , Phosphorylation
4.
Theranostics ; 14(8): 3267-3281, 2024.
Article in English | MEDLINE | ID: mdl-38855181

ABSTRACT

Background: Myocardial infarction (MI) as a consequence of atherosclerosis-associated acute thrombosis is a leading cause of death and disability globally. Antiplatelet and anticoagulant drugs are standard therapies in preventing and treating MI. However, all clinically used drugs are associated with bleeding complications, which ultimately limits their use in patients with a high risk of bleeding. We have developed a new recombinant drug, targ-HSA-TAP, that combines targeting and specific inhibition of activated platelets as well as anticoagulation. This drug is designed and tested for a prolonged circulating half-life, enabling unique thromboprophylaxis without bleeding complications. Methods: Targ-HSA-TAP combines a single-chain antibody (scFv) that targets activated glycoprotein IIb/IIIa on activated platelets, human serum albumin (HSA) for prolonged circulation, and tick anticoagulant peptide (TAP) for coagulation FX inhibition. A non-binding scFv is employed as a non-targeting control (non-targ-HSA-TAP). Its efficacy was investigated in vivo using murine models of acute thrombosis and cardiac ischemia-reperfusion (I/R) injury. Results: Our experiments confirmed the targeting specificity of targ-HSA-TAP to activated platelets and demonstrated effective prevention of platelet aggregation and thrombus formation, as well as FXa inhibition in vitro. Thromboprophylactic administration of targ-HSA-TAP subcutaneously in mice prevented occlusion of the carotid artery after ferric chloride injury as compared to non-targ-HSA-TAP and PBS-control treated mice. By comparing the therapeutic outcomes between targ-TAP and targ-HSA-TAP, we demonstrate the significant improvements brought by the HSA fusion in extending the drug's half-life and enhancing its therapeutic window for up to 16 h post-administration. Importantly, tail bleeding time was not prolonged with targ-HSA-TAP in contrast to the clinically used anticoagulant enoxaparin. Furthermore, in a murine model of cardiac I/R injury, mice administered targ-HSA-TAP 10 h before injury demonstrated preserved cardiac function, with significantly higher ejection fraction and fractional shortening, as compared to the non-targ-HSA-TAP and PBS control groups. Advanced strain analysis revealed reduced myocardial deformation and histology confirmed a reduced infarct size in targ-HSA-TAP treated mice compared to control groups. Conclusion: The inclusion of HSA represents a significant advancement in the design of targeted therapeutic agents for thromboprophylaxis. Our activated platelet-targeted targ-HSA-TAP is a highly effective antithrombotic drug with both anticoagulant and antiplatelet effects while retaining normal hemostasis. The long half-life of targ-HSA-TAP provides the unique opportunity to use this antithrombotic drug for more effective, long-lasting and safer anti-thrombotic prophylaxis. In cases where MI occurs, this prophylactic strategy reduces thrombus burden and effectively reduces cardiac I/R injury.


Subject(s)
Blood Platelets , Hemorrhage , Serum Albumin, Human , Thrombosis , Animals , Mice , Thrombosis/prevention & control , Thrombosis/drug therapy , Humans , Hemorrhage/prevention & control , Blood Platelets/drug effects , Blood Platelets/metabolism , Disease Models, Animal , Male , Anticoagulants/pharmacology , Anticoagulants/therapeutic use , Single-Chain Antibodies/pharmacology , Single-Chain Antibodies/therapeutic use , Myocardial Reperfusion Injury/prevention & control , Myocardial Reperfusion Injury/drug therapy , Myocardial Infarction/drug therapy , Mice, Inbred C57BL , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/therapeutic use
5.
PLoS One ; 19(6): e0304761, 2024.
Article in English | MEDLINE | ID: mdl-38843265

ABSTRACT

Type 2 diabetes predisposes patients to heart disease, which is the primary cause of death across the globe. Type 2 diabetes often accompanies obesity and is defined by insulin resistance and abnormal glucose handling. Insulin resistance impairs glucose uptake and results in hyperglycemia, which damages tissues such as kidneys, liver, and heart. 2-oxoglutarate (2-OG)- and iron-dependent oxygenases (2-OGDOs), a family of enzymes regulating various aspects of cellular physiology, have been studied for their role in obesity and diet-induced insulin resistance. However, nothing is known of the 2-OGDO family member 2-oxoglutarate and iron-dependent prolyl hydroxylase domain containing protein 1 (OGFOD1) in this setting. OGFOD1 deletion leads to protection in cardiac ischemia-reperfusion injury and cardiac hypertrophy, which are two cardiac events that can lead to heart failure. Considering the remarkable correlation between heart disease and diabetes, the cardioprotection observed in OGFOD1-knockout mice led us to challenge these knockouts with high-fat diet. Wildtype mice fed a high-fat diet developed diet-induced obesity, insulin resistance, and glucose intolerance, but OGFOD1 knockout mice fed this same diet were resistant to diet-induced obesity and insulin resistance. These results support OGFOD1 down-regulation as a strategy for preventing obesity and insulin handling defects.


Subject(s)
Diet, High-Fat , Insulin Resistance , Mice, Knockout , Obesity , Animals , Obesity/metabolism , Obesity/genetics , Mice , Diet, High-Fat/adverse effects , Male , Prolyl Hydroxylases/metabolism , Prolyl Hydroxylases/genetics , Glucose Intolerance/metabolism , Glucose Intolerance/genetics , Mice, Inbred C57BL , Gene Deletion , Cardiomegaly/metabolism , Cardiomegaly/prevention & control , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/prevention & control , Myocardial Reperfusion Injury/genetics
6.
J Physiol Investig ; 67(3): 129-138, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38902960

ABSTRACT

Ischemia-reperfusion (IR) injury remains a pivotal contributor to myocardial damage following acute coronary events and revascularization procedures. Phosphoinositide 3-kinase (PI3K), a key mediator of cell survival signaling, plays a central role in regulating inflammatory responses and cell death mechanisms. Trans-chalcone (Tch), a natural compound known for its anti-inflammatory activities, has shown promise in various disease models. The aim of the current study was to investigate the potential protective effects of Tch against myocardial injury induced by ischemia and reperfusion challenges by targeting the PI3K-inflammasome interaction. Experimental models utilizing male rats subjected to an in vivo model of IR injury and myocardial infarction were employed. Administration of Tch (100 µg/kg, intraperitoneally) significantly reduced myocardial injury, as indicated by limited infarct size and decreased levels of the myocardial enzyme troponin. Mechanistically, Tch upregulated PI3K expression, thereby inhibiting the activity of the NOD-like receptor protein 3 inflammasome followed by the activation of pro-inflammatory cytokines interleukin-1ß (IL-1ß) and IL-18. Moreover, it mitigated oxidative stress and suppressed vascular-intercellular adhesion molecules, contributing to its cardioprotective effects. The PI3K/Akt pathway inhibitor LY294002 considerably attenuated the beneficial effects of Tch. These findings highlight the therapeutic potential of Tch in ameliorating myocardial injury associated with IR insults through its modulation of the PI3K/Akt-inflammasome axis. The multifaceted mechanisms underlying its protective effects signify Tch as a promising candidate for further exploration in developing targeted therapies aimed at mitigating ischemic heart injury and improving clinical outcomes in cardiovascular diseases characterized by IR injury.


Subject(s)
Myocardial Reperfusion Injury , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Rats, Sprague-Dawley , Animals , Male , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/prevention & control , Myocardial Reperfusion Injury/pathology , Proto-Oncogene Proteins c-akt/metabolism , Rats , Phosphatidylinositol 3-Kinases/metabolism , Inflammasomes/metabolism , Inflammasomes/drug effects , Chalcone/pharmacology , Chalcone/analogs & derivatives , Chalcone/therapeutic use , Signal Transduction/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
7.
Sci Rep ; 14(1): 14350, 2024 06 21.
Article in English | MEDLINE | ID: mdl-38906975

ABSTRACT

Cardiac ischemic preconditioning (Pre) reduces cardiac ischemia-reperfusion injury (IRI) by stimulating opioid receptors. Chronic use of opioids can alter the signaling pathways. We investigated the effects of chronic methadone use on IRI and Pre. The experiments were performed on isolated hearts of male Wistar rats in four groups: IRI, Methadone + IRI (M-IRI), Pre + IRI (Pre-IRI), Methadone + Pre + IRI (M-Pre-IRI). The infarct size (IS) in the Pre-IRI group was smaller than the IRI group (26.8% vs. 47.8%, P < 0.05). In the M-IRI and M-Pre-IRI groups, the infarct size was similar to the IRI group. Akt (Ak strain transforming) phosphorylation in the Pre-IRI, M-IRI, and M-Pre-IRI groups was significantly higher than in the IRI group (0.56 ± 0.15, 0.63 ± 0.20, and 0.93 ± 0.18 vs 0.28 ± 0.17 respectively). STAT3 (signal transducer and activator of transcription 3) phosphorylation in the Pre-IRI and M-Pre-IRI groups (1.38 ± 0.14 and 1.46 ± 0.33) was significantly higher than the IRI and M-IRI groups (0.99 ± 0.1 and 0.98 ± 0.2). Thus, chronic use of methadone not only has no protective effect against IRI but also destroys the protective effects of ischemic preconditioning. This may be due to the hyperactivation of Akt and changes in signaling pathways.


Subject(s)
Ischemic Preconditioning, Myocardial , Methadone , Myocardial Reperfusion Injury , Proto-Oncogene Proteins c-akt , Rats, Wistar , STAT3 Transcription Factor , Animals , Methadone/pharmacology , STAT3 Transcription Factor/metabolism , Male , Proto-Oncogene Proteins c-akt/metabolism , Phosphorylation/drug effects , Rats , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/prevention & control , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/pathology , Ischemic Preconditioning, Myocardial/methods , Signal Transduction/drug effects , Reperfusion Injury/metabolism , Reperfusion Injury/prevention & control , Reperfusion Injury/drug therapy , Reperfusion Injury/pathology
8.
Sci Rep ; 14(1): 12949, 2024 06 05.
Article in English | MEDLINE | ID: mdl-38839839

ABSTRACT

Growth/differentiation factor-15 (GDF15) is considered an unfavourable prognostic biomarker for cardiovascular disease in clinical data, while experimental studies suggest it has cardioprotective potential. This study focuses on the direct cardiac effects of GDF15 during ischemia-reperfusion injury in Wistar male rats, employing concentrations relevant to patients at high cardiovascular risk. Initially, we examined circulating levels and heart tissue expression of GDF15 in rats subjected to ischemia-reperfusion and sham operations in vivo. We then evaluated the cardiac effects of GDF15 both in vivo and ex vivo, administering recombinant GDF15 either before 30 min of ischemia (preconditioning) or at the onset of reperfusion (postconditioning). We compared infarct size and cardiac contractile recovery between control and rGDF15-treated rats. Contrary to our expectations, ischemia-reperfusion did not increase GDF15 plasma levels compared to sham-operated rats. However, cardiac protein and mRNA expression increased in the infarcted zone of the ischemic heart after 24 h of reperfusion. Notably, preconditioning with rGDF15 had a cardioprotective effect, reducing infarct size both in vivo (65 ± 5% in control vs. 42 ± 6% in rGDF15 groups) and ex vivo (60 ± 4% in control vs. 45 ± 4% in rGDF15 groups), while enhancing cardiac contractile recovery ex vivo. However, postconditioning with rGDF15 did not alter infarct size or the recovery of contractile parameters in vivo or ex vivo. These novel findings reveal that the short-term exogenous administration of rGDF15 before ischemia, at physiologically relevant levels, protects the heart against ischemia-reperfusion injury in both in vivo and ex vivo settings. The ex vivo results indicate that rGDF15 operates independently of the inflammatory, endocrine and nervous systems, suggesting direct and potent cardioprotective properties against ischemia-reperfusion injury.


Subject(s)
Growth Differentiation Factor 15 , Myocardial Infarction , Rats, Wistar , Growth Differentiation Factor 15/metabolism , Growth Differentiation Factor 15/genetics , Animals , Male , Myocardial Infarction/metabolism , Rats , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/prevention & control , Myocardium/metabolism , Myocardium/pathology , Cardiotonic Agents/pharmacology , Cardiotonic Agents/administration & dosage , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology , Ischemic Preconditioning, Myocardial/methods
9.
J Biol Chem ; 300(6): 107372, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38754781

ABSTRACT

OMT-28 is a metabolically robust small molecule developed to mimic the structure and function of omega-3 epoxyeicosanoids. However, it remained unknown to what extent OMT-28 also shares the cardioprotective and anti-inflammatory properties of its natural counterparts. To address this question, we analyzed the ability of OMT-28 to ameliorate hypoxia/reoxygenation (HR)-injury and lipopolysaccharide (LPS)-induced endotoxemia in cultured cardiomyocytes. Moreover, we investigated the potential of OMT-28 to limit functional damage and inflammasome activation in isolated perfused mouse hearts subjected to ischemia/reperfusion (IR) injury. In the HR model, OMT-28 (1 µM) treatment largely preserved cell viability (about 75 versus 40% with the vehicle) and mitochondrial function as indicated by the maintenance of NAD+/NADH-, ADP/ATP-, and respiratory control ratios. Moreover, OMT-28 blocked the HR-induced production of mitochondrial reactive oxygen species. Pharmacological inhibition experiments suggested that Gαi, PI3K, PPARα, and Sirt1 are essential components of the OMT-28-mediated pro-survival pathway. Counteracting inflammatory injury of cardiomyocytes, OMT-28 (1 µM) reduced LPS-induced increases in TNFα protein (by about 85% versus vehicle) and NF-κB DNA binding (by about 70% versus vehicle). In the ex vivo model, OMT-28 improved post-IR myocardial function recovery to reach about 40% of the baseline value compared to less than 20% with the vehicle. Furthermore, OMT-28 (1 µM) limited IR-induced NLRP3 inflammasome activation similarly to a direct NLRP3 inhibitor (MCC950). Overall, this study demonstrates that OMT-28 possesses potent cardio-protective and anti-inflammatory properties supporting the hypothesis that extending the bioavailability of omega-3 epoxyeicosanoids may improve their prospects as therapeutic agents.


Subject(s)
Cardiotonic Agents , Myocytes, Cardiac , Animals , Mice , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Cardiotonic Agents/pharmacology , Cardiotonic Agents/chemistry , Inflammasomes/metabolism , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/prevention & control , Lipopolysaccharides/pharmacology , Male , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Mice, Inbred C57BL , Sirtuin 1/metabolism , Anti-Inflammatory Agents/pharmacology , Reactive Oxygen Species/metabolism , Fatty Acids, Omega-3/pharmacology , Fatty Acids, Omega-3/chemistry , Endotoxemia/drug therapy , Endotoxemia/metabolism
10.
Cardiovasc Toxicol ; 24(7): 646-655, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38801481

ABSTRACT

This research is concentrated on investigating the role and mechanism of miR-652-3p in the protective effects of isoflurane (ISO) against myocardial ischemia-reperfusion (I/R) injury. H9c2 cells underwent pretreatment with varying concentrations of ISO, and subsequently, a hypoxia/reoxygenation (H/R) model was constructed. The levels of miR-652-3p, ISL LIM homeobox 1 (ISL1), and inflammatory cytokines interleukin (IL)-6 and tumor necrosis factor-alpha (TNF-α) were evaluated through reverse transcription polymerase chain reaction (RT-qPCR). Enzyme-linked immunosorbent assay was employed to investigate concentrations of myocardial injury markers, such as creatine kinase-MB (CK-MB) and cardiac troponin I (cTnI). Cell counting kit-8 was used to evaluate cell viability, while flow cytometry was utilized to measure apoptosis. Additionally, a dual luciferase reporter assay was conducted to validate the targeting relationship between ISL1 and miR-652-3p. Herein, we confirmed that the level of miR-652-3p was gradually increased with prolonged hypoxia; nevertheless, this increase was suppressed by ISO pretreatment (P < 0.05). Additionally, ISO pretreatment prevented the decrease in cell viability, increase in apoptosis, and overproduction of IL-6, TNF-α, CK-MB, and cTnI induced by H/R (P < 0.05). However, the inhibitory effects of ISO were counteracted by the increased levels of miR-652-3p (P < 0.05). ISL1 is a potential target of miR-652-3p. H/R induction suppressed ISL1 levels compared to the control, but ISO treatment increased its expression (P < 0.05). Overexpression of ISL1 inhibited the elimination of the protective effect of ISO on myocardial damage induced by the elevation of miR-652-3p (P < 0.05). The findings of this research confirm that miR-652-3p attenuated the protective effect of ISO on cardiomyocytes in myocardial ischemia by targeting ISL1.


Subject(s)
Apoptosis , Cell Hypoxia , Interleukin-6 , Isoflurane , LIM-Homeodomain Proteins , MicroRNAs , Myocardial Reperfusion Injury , Myocytes, Cardiac , Transcription Factors , MicroRNAs/metabolism , MicroRNAs/genetics , Isoflurane/pharmacology , LIM-Homeodomain Proteins/metabolism , LIM-Homeodomain Proteins/genetics , Animals , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/enzymology , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/prevention & control , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/genetics , Cell Line , Apoptosis/drug effects , Rats , Transcription Factors/metabolism , Transcription Factors/genetics , Interleukin-6/metabolism , Interleukin-6/genetics , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/metabolism , Gene Expression Regulation/drug effects , Inflammation Mediators/metabolism , Creatine Kinase, MB Form/metabolism , Creatine Kinase, MB Form/blood , Troponin I/metabolism , Cytoprotection
11.
Cardiovasc Toxicol ; 24(7): 637-645, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38720121

ABSTRACT

To investigate the role of microRNA-195-3p (miR-195-3p) in hypoxia/reoxygenation (H/R)-induced cardiomyocyte injury. AC16 human cardiomyocyte cells were cultured and pretreated with different concentrations of isoflurane (ISO) (1%, 2%, and 3%), followed by 6 h each of hypoxia and reoxygenation to construct H/R cell models. The optimum ISO concentration was assessed based on the cell viability. miR-195-3p expression was regulated by in vitro cell transfection. Cell viability was determined by MTT assay, and apoptosis was evaluated by flow cytometry. The levels of myocardial injury and inflammation were determined by enzyme-linked immunosorbent assay. Compared with the control group, the cell viability of the H/R group had significantly decreased and that of ISO pretreatment had increased in a dose-dependent manner. Therefore, we selected a 2% ISO concentration for pretreatment. MiR-195-3p expression had significantly increased in the H/R group and decreased after 2% ISO pretreatment. Additionally, the number of apoptotic cells and the levels of lactate dehydrogenase, creatine kinase-myoglobin binding, cardiac troponin I, interleukin (IL)-1ß, IL-6, and tumor necrosis factor-α had increased significantly. ISO preconditioning inhibited H/R-induced AC16 cell damage, whereas miR-195-3p overexpression reversed the protective effects of ISO on cardiomyocytes. The expression of phosphatase and tensin homolog deleted on chromosome 10 (PTEN) was reduced in the H/R-induced AC16 cells, and PTEN is a downstream target gene of miR-195-3p. Preconditioning with 2% ISO plays a protective role in H/R-induced AC16 cell damage by inhibiting miR-195-3p expression.


Subject(s)
Apoptosis , Cell Hypoxia , Isoflurane , MicroRNAs , Myocardial Reperfusion Injury , Myocytes, Cardiac , Signal Transduction , MicroRNAs/metabolism , MicroRNAs/genetics , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Isoflurane/pharmacology , Isoflurane/toxicity , Humans , Apoptosis/drug effects , Cell Line , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/prevention & control , Myocardial Reperfusion Injury/genetics , Inflammation Mediators/metabolism , Cell Survival/drug effects , Down-Regulation , Dose-Response Relationship, Drug , Cytokines/metabolism
12.
Am J Physiol Heart Circ Physiol ; 327(1): H70-H79, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38700468

ABSTRACT

Activation of the vagus nerve mediates cardioprotection and attenuates myocardial ischemia/reperfusion (I/R) injury. In response to vagal activation, acetylcholine (ACh) is released from the intracardiac nervous system (ICNS) and activates intracellular cardioprotective signaling cascades. Recently, however, a nonneuronal cholinergic cardiac system (NNCCS) in cardiomyocytes has been described as an additional source of ACh. To investigate whether the NNCCS mediates cardioprotection in the absence of vagal and ICNS activation, we used a reductionist approach of isolated adult rat ventricular cardiomyocytes without neuronal cells, using hypoxic preconditioning (HPC) as a protective stimulus. Adult rat ventricular cardiomyocytes were isolated, the absence of neuronal cells was confirmed, and HPC was induced by 10/20 min hypoxia/reoxygenation (H/R) before subjection to 30/5 min H/R to simulate I/R injury. Cardiomyocyte viability was assessed by trypan blue staining at baseline and after HPC+H/R or H/R. Intra- and extracellular ACh was quantified using liquid chromatography-coupled mass spectrometry at baseline, after HPC, after hypoxia, and after reoxygenation, respectively. In a subset of experiments, muscarinic and nicotinic ACh receptor (m- and nAChR) antagonists were added during HPC or during H/R. Cardiomyocyte viability at baseline (69 ± 4%) was reduced by H/R (10 ± 3%). With HPC, cardiomyocyte viability was preserved after H/R (25 ± 6%). Intra- and extracellular ACh increased during hypoxia; HPC further increased both intra- and extracellular ACh (from 0.9 ± 0.7 to 1.5 ± 1.0 nmol/mg; from 0.7 ± 0.6 to 1.1 ± 0.7 nmol/mg, respectively). The addition of mAChR and nAChR antagonists during HPC had no impact on HPC's protection; however, protection was abrogated when antagonists were added during H/R (cardiomyocyte viability after H/R: 23 ± 5%; 13 ± 4%). In conclusion, activation of the NNCCS is involved in cardiomyocyte protection; HPC increases intra- and extracellular ACh during H/R, and m- and nAChRs are causally involved in HPC's cardiomyocyte protection during H/R. The interplay between upstream ICNS activation and NNCCS activation in myocardial cholinergic metabolism and cardioprotection needs to be investigated in future studies.NEW & NOTEWORTHY The intracardiac nervous system is considered to be involved in ischemic conditioning's cardioprotection through the release of acetylcholine (ACh). However, we demonstrate that hypoxic preconditioning (HPC) protects from hypoxia/reoxygenation injury and increases intra- and extracellular ACh during hypoxia in isolated adult ventricular rat cardiomyocytes. HPC's protection involves cardiomyocyte muscarinic and nicotinic ACh receptor activation. Thus, besides the intracardiac nervous system, a nonneuronal cholinergic cardiac system may also be causally involved in cardiomyocyte protection by ischemic conditioning.


Subject(s)
Acetylcholine , Myocardial Reperfusion Injury , Myocytes, Cardiac , Animals , Myocytes, Cardiac/metabolism , Acetylcholine/pharmacology , Acetylcholine/metabolism , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/prevention & control , Myocardial Reperfusion Injury/physiopathology , Myocardial Reperfusion Injury/pathology , Male , Cell Hypoxia , Rats , Non-Neuronal Cholinergic System , Ischemic Preconditioning, Myocardial , Rats, Sprague-Dawley , Cell Survival , Receptors, Muscarinic/metabolism , Cells, Cultured , Muscarinic Antagonists/pharmacology
13.
Vascul Pharmacol ; 155: 107378, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38729253

ABSTRACT

OBJECTIVES: Flavonoids are polyphenolic compounds found in a wide range of foods, including fruits, vegetables, tea plants, and other natural products. They have been mainly classified as flavanols, flavonols, flavones, isoflavones, flavanones, and flavanonols. In this comprehensive review, we will discuss preclinical pieces of evidence on the potential of flavonoids for the prevention/treatment of myocardial ischemia-reperfusion (IR) injury. KEY FINDINGS: In-vitro and in-vivo studies have shown that flavonoids play an important role in preventing ischemic heart disease (IHD). They possess strong anti-oxidant, anti-inflammatory, anti-bacterial, anti-thrombotic, anti-apoptotic, and anti-carcinogenic activities. In addition, at a molecular level, flavonoids also modulate various pathways like MAPK, NFκB etc. to confer beneficial effects. SUMMARY: The current review of flavonoids in myocardial ischemia-reperfusion injury furnishes updated information that could drive future research. The in-vitro and in-vivo experiments have demonstrated various favourable pharmacological properties of flavonoids. This review provides valuable information to conduct clinical studies, validating the safety aspects of flavonoids in the clinical domain.


Subject(s)
Flavonoids , Myocardial Reperfusion Injury , Humans , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/prevention & control , Myocardial Reperfusion Injury/physiopathology , Flavonoids/pharmacology , Animals , Signal Transduction/drug effects , Antioxidants/pharmacology , Anti-Inflammatory Agents/pharmacology , Myocardium/metabolism , Myocardium/pathology , Apoptosis/drug effects , Cardiovascular Agents/pharmacology , Cardiovascular Agents/therapeutic use
15.
Biomed Pharmacother ; 175: 116689, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38703508

ABSTRACT

Ischemic heart disease invariably leads to devastating damage to human health. Nicotinamide ribose (NR), as one of the precursors of NAD+ synthesis, has been discovered to exert a protective role in various neurological and cardiovascular disorders. Our findings demonstrated that pretreatment with 200 mg/kg NR for 3 h significantly reduced myocardial infarct area, decreased levels of CK-MB and LDH in serum, and improved cardiac function in the rats during myocardial ischemia-reperfusion (I/R) injury. Meanwhile, 0.5 mM NR also effectively increased the viability and decreased the LDH release of H9c2 cells during OGD/R. We had provided evidence that NR pretreatment could decrease mitochondrial reactive oxygen species (mtROS) production and MDA content, and enhance SOD activity, thereby mitigating mitochondrial damage and inhibiting apoptosis during myocardial I/R injury. Further investigations revealed that NR increased NAD+ content and upregulated SIRT3 protein expression in myocardium. Through using of SIRT3 small interfering RNA and the SIRT3 deacetylase activity inhibitor 3-TYP, we had confirmed that the cardioprotective effect of NR on cardiomyocytes was largely dependent on the inhibition of mitochondrial oxidative stress via SIRT3-SOD2 axis. Overall, our study suggested that exogenous supplementation with NR mitigated mitochondrial damage and inhibited apoptosis during myocardial I/R injury by reducing mitochondrial oxidative stress via SIRT3-SOD2-mtROS pathway.


Subject(s)
Apoptosis , Myocardial Reperfusion Injury , Niacinamide , Oxidative Stress , Pyridinium Compounds , Rats, Sprague-Dawley , Signal Transduction , Sirtuin 3 , Superoxide Dismutase , Animals , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/prevention & control , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Sirtuin 3/metabolism , Signal Transduction/drug effects , Male , Niacinamide/pharmacology , Niacinamide/analogs & derivatives , Superoxide Dismutase/metabolism , Rats , Apoptosis/drug effects , Oxidative Stress/drug effects , Pyridinium Compounds/pharmacology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Reactive Oxygen Species/metabolism , Cell Line , Cardiotonic Agents/pharmacology , Sirtuins
16.
Eur J Pharmacol ; 976: 176698, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-38821168

ABSTRACT

Myocardial infarction (MI) is a life-threatening cardiovascular disease that, on average, results in 8.5 million deaths worldwide each year. Timely revascularization of occluded vessels is a critical method of myocardial salvage. However, reperfusion paradoxically leads to the worsening of myocardial damage known as myocardial ischaemia/reperfusion injury (MI/RI). Therefore, reducing the size of myocardial infarction after reperfusion is critical and remains an important therapeutic goal. The susceptibility of the myocardium to MI/RI may be increased by diabetes. Currently, some traditional antidiabetic agents such as metformin reduce MI/RI by decreasing inflammation, inhibiting oxidative stress, and improving vascular endothelial function. This appears to be a new direction for the treatment of MI/RI. Recent cardiovascular outcome trials have shown that several oral antidiabetic agents, including glucagon-like peptide-1 receptor agonists (GLP-1RAs), dipeptidyl peptidase-4 inhibitors (DPP-4is), and sodium-glucose-linked transporter-2 inhibitors (SGLT-2is), not only have good antidiabetic effects but also have a protective effect on myocardial protection. This article aims to discuss the mechanisms and effects of oral antidiabetic agents, including GLP-1RAs, DPP-4is, and SGLT-2is, on MI/RI to facilitate their clinical application.


Subject(s)
Dipeptidyl-Peptidase IV Inhibitors , Glucagon-Like Peptide-1 Receptor , Hypoglycemic Agents , Myocardial Reperfusion Injury , Sodium-Glucose Transporter 2 Inhibitors , Humans , Glucagon-Like Peptide-1 Receptor/agonists , Animals , Myocardial Reperfusion Injury/prevention & control , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/metabolism , Sodium-Glucose Transporter 2 Inhibitors/therapeutic use , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Hypoglycemic Agents/therapeutic use , Hypoglycemic Agents/pharmacology , Administration, Oral , Glucagon-Like Peptide-1 Receptor Agonists
17.
Biomed Pharmacother ; 176: 116764, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38805965

ABSTRACT

Ischemic heart disease (IHD) is a condition where the heart muscle does not receive enough blood flow, leading to cardiac dysfunction. Restoring blood flow to the coronary artery is an effective clinical therapy for myocardial ischemia. This strategy helps lower the size of the myocardial infarction and improves the prognosis of patients. Nevertheless, if the disrupted blood flow to the heart muscle is restored within a specific timeframe, it leads to more severe harm to the previously deprived heart tissue. This condition is referred to as myocardial ischemia/reperfusion injury (MIRI). Until now, there is a dearth of efficacious strategies to prevent and manage MIRI. Hormones are specialized substances that are produced directly into the circulation by endocrine organs or tissues in humans and animals, and they have particular effects on the body. Hormonal medications utilize human or animal hormones as their active components, encompassing sex hormones, adrenaline medications, thyroid hormone medications, and others. While several studies have examined the preventive properties of different endocrine hormones, such as estrogen and hormone analogs, on myocardial injury caused by ischemia-reperfusion, there are other hormone analogs whose mechanisms of action remain unexplained and whose safety cannot be assured. The current study is on hormones and hormone medications, elucidating the mechanism of hormone pharmaceuticals and emphasizing the cardioprotective effects of different endocrine hormones. It aims to provide guidance for the therapeutic use of drugs and offer direction for the examination of MIRI in clinical therapy.


Subject(s)
Myocardial Reperfusion Injury , Myocardial Reperfusion Injury/prevention & control , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/metabolism , Humans , Animals , Hormones/metabolism , Hormones/therapeutic use , Cardiotonic Agents/pharmacology , Cardiotonic Agents/therapeutic use
18.
BMC Cardiovasc Disord ; 24(1): 280, 2024 May 29.
Article in English | MEDLINE | ID: mdl-38811893

ABSTRACT

BACKGROUND: Myocardial ischemia-reperfusion injury (I/RI) is a major cause of perioperative cardiac-related adverse events and death. Studies have shown that sevoflurane postconditioning (SpostC), which attenuates I/R injury and exerts cardioprotective effects, regulates mitochondrial dynamic balance via HIF-1α, but the exact mechanism is unknown. This study investigates whether the PI3K/AKT pathway in SpostC regulates mitochondrial dynamic balance by mediating HIF-1α, thereby exerting myocardial protective effects. METHODS: The H9C2 cardiomyocytes were cultured to establish the hypoxia-reoxygenation (H/R) model and randomly divided into 4 groups: Control group, H/R group, sevoflurane postconditioning (H/R + SpostC) group and PI3K/AKT blocker (H/R + SpostC + LY) group. Cell survival rate was determined by CCK-8; Apoptosis rate was determined by flow cytometry; mitochondrial membrane potential was evaluated by Mito Tracker™ Red; mRNA expression levels of AKT, HIF-1α, Opa1and Drp1 were detected by quantitative real-time polymerase chain reaction (qRT-PCR); Western Blot assay was used to detect the protein expression levels of AKT, phosphorylated AKT (p-AKT), HIF-1α, Opa1 and Drp1. RESULTS: Compared with the H/R group, the survival rate of cardiomyocytes in the H/R + SpostC group increased, the apoptosis rate decreased and the mitochondrial membrane potential increased. qRT-PCR showed that the mRNA expression of HIF-1α and Opa1 were higher in the H/R + SpostC group compared with the H/R group, whereas the transcription level of Drp1 was lower in the H/R + SpostC group. In the H/R + SpostC + LY group, the mRNA expression of HIF-1α was lower than the H/R + SpostC group. There was no difference in the expression of Opa1 mRNA between the H/R group and the H/R + SpostC + LY group. WB assay results showed that compared with the H/R group, the protein expression levels of HIF-1α, Opa1, P-AKT were increased and Drp1 protein expression levels were decreased in the H/R + SpostC group. HIF-1α, P-AKT protein expression levels were decreased in the H/R + SpostC + LY group compared to the H/R + SpostC group. CONCLUSION: SpostC mediates HIF-1α-regulated mitochondrial fission and fusion-related protein expression to maintain mitochondrial dynamic balance by activating the PI3K/AKT pathway and increasing AKT phosphorylation, thereby attenuating myocardial I/R injury.


Subject(s)
Apoptosis , Hypoxia-Inducible Factor 1, alpha Subunit , Membrane Potential, Mitochondrial , Mitochondria, Heart , Mitochondrial Dynamics , Myocardial Reperfusion Injury , Myocytes, Cardiac , Phosphatidylinositol 3-Kinase , Proto-Oncogene Proteins c-akt , Sevoflurane , Signal Transduction , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Proto-Oncogene Proteins c-akt/metabolism , Animals , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/enzymology , Sevoflurane/pharmacology , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/prevention & control , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/enzymology , Mitochondrial Dynamics/drug effects , Cell Line , Rats , Apoptosis/drug effects , Phosphatidylinositol 3-Kinase/metabolism , Mitochondria, Heart/drug effects , Mitochondria, Heart/metabolism , Mitochondria, Heart/pathology , Mitochondria, Heart/enzymology , Membrane Potential, Mitochondrial/drug effects , Cell Hypoxia , Dynamins/metabolism , Dynamins/genetics , GTP Phosphohydrolases/metabolism , GTP Phosphohydrolases/genetics , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Cytoprotection , Ischemic Postconditioning , Phosphorylation
20.
Int J Mol Sci ; 25(9)2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38732142

ABSTRACT

The high mortality rate among patients with acute myocardial infarction (AMI) is one of the main problems of modern cardiology. It is quite obvious that there is an urgent need to create more effective drugs for the treatment of AMI than those currently used in the clinic. Such drugs could be enzyme-resistant peptide analogs of glucagon-like peptide-1 (GLP-1). GLP-1 receptor (GLP1R) agonists can prevent ischemia/reperfusion (I/R) cardiac injury. In addition, chronic administration of GLP1R agonists can alleviate the development of adverse cardiac remodeling in myocardial infarction, hypertension, and diabetes mellitus. GLP1R agonists can protect the heart against oxidative stress and reduce proinflammatory cytokine (IL-1ß, TNF-α, IL-6, and MCP-1) expression in the myocardium. GLP1R stimulation inhibits apoptosis, necroptosis, pyroptosis, and ferroptosis of cardiomyocytes. The activation of the GLP1R augments autophagy and mitophagy in the myocardium. GLP1R agonists downregulate reactive species generation through the activation of Epac and the GLP1R/PI3K/Akt/survivin pathway. The GLP1R, kinases (PKCε, PKA, Akt, AMPK, PI3K, ERK1/2, mTOR, GSK-3ß, PKG, MEK1/2, and MKK3), enzymes (HO-1 and eNOS), transcription factors (STAT3, CREB, Nrf2, and FoxO3), KATP channel opening, and MPT pore closing are involved in the cardioprotective effect of GLP1R agonists.


Subject(s)
Cardiotonic Agents , Glucagon-Like Peptide-1 Receptor , Signal Transduction , Humans , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/metabolism , Cardiotonic Agents/pharmacology , Cardiotonic Agents/therapeutic use , Signal Transduction/drug effects , Animals , Peptides/pharmacology , Peptides/therapeutic use , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/prevention & control , Myocardial Reperfusion Injury/pathology , Myocardial Infarction/metabolism , Myocardial Infarction/drug therapy , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Glucagon-Like Peptide-1 Receptor Agonists
SELECTION OF CITATIONS
SEARCH DETAIL
...