Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.592
Filter
1.
J Dev Orig Health Dis ; 15: e9, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38721989

ABSTRACT

Sodium overload during childhood impairs baroreflex sensitivity and increases arterial blood pressure and heart rate in adulthood; these effects persist even after high-salt diet (HSD) withdrawal. However, the literature lacks details on the effects of HSD during postnatal phases on cardiac ischemia/reperfusion responses in adulthood. The current study aimed to elucidate the impact of HSD during infancy adolescence on isolated heart function and cardiac ischemia/reperfusion responses in adulthood. Male 21-day-old Wistar rats were treated for 60 days with hypertonic saline solution (NaCl; 0.3M; experimental group) or tap water (control group). Subsequently, both groups were maintained on a normal sodium diet for 30 days. Subsequently, the rats were euthanized, and their hearts were isolated and perfused according to the Langendorff technique. After 30 min of the basal period, the hearts were subjected to 20 min of anoxia, followed by 20 min of reperfusion. The basal contractile function was unaffected by HSD. However, HSD elevated the left ventricular end-diastolic pressure during reperfusion (23.1 ± 5.2 mmHg vs. 11.6 ± 1.4 mmHg; p < 0.05) and increased ectopic incidence period during reperfusion (208.8 ± 32.9s vs. 75.0 ± 7.8s; p < 0.05). In conclusion, sodium overload compromises cardiac function after reperfusion events, diminishes ventricular relaxation, and increases the severity of arrhythmias, suggesting a possible arrhythmogenic effect of HSD in the postnatal phases.


Subject(s)
Arrhythmias, Cardiac , Myocardial Reperfusion Injury , Rats, Wistar , Animals , Rats , Arrhythmias, Cardiac/etiology , Arrhythmias, Cardiac/physiopathology , Male , Myocardial Reperfusion Injury/etiology , Myocardial Reperfusion Injury/physiopathology , Diastole/physiology , Sodium Chloride, Dietary/adverse effects , Heart Rate/physiology
2.
J Am Coll Cardiol ; 83(22): 2196-2213, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38811097

ABSTRACT

Despite impressive improvements in the care of patients with ST-segment elevation myocardial infarction, mortality remains high. Reperfusion is necessary for myocardial salvage, but the abrupt return of flow sets off a cascade of injurious processes that can lead to further necrosis. This has been termed myocardial ischemia-reperfusion injury and is the subject of this review. The pathologic and molecular bases for myocardial ischemia-reperfusion injury are increasingly understood and include injury from reactive oxygen species, inflammation, calcium overload, endothelial dysfunction, and impaired microvascular flow. A variety of pharmacologic strategies have been developed that have worked well in preclinical models and some have shown promise in the clinical setting. In addition, there are newer mechanical approaches including mechanical unloading of the heart prior to reperfusion that are in current clinical trials.


Subject(s)
Myocardial Reperfusion Injury , Humans , Myocardial Reperfusion Injury/physiopathology , Myocardial Reperfusion Injury/etiology , Myocardial Infarction/physiopathology , Myocardial Reperfusion/methods , ST Elevation Myocardial Infarction/physiopathology , ST Elevation Myocardial Infarction/therapy
3.
Methods Mol Biol ; 2803: 189-203, 2024.
Article in English | MEDLINE | ID: mdl-38676894

ABSTRACT

Acute myocardial infarction continues to account for a growing burden of heart failure worldwide. Despite existing therapies, new approaches for reducing the extent of damage and better managing heart failure progression are urgently needed. Preclinical large animal models are a critical step in the translation of scientific discoveries toward clinical trials and therapeutic application. In this chapter, we detail methods to induce swine models of myocardial infarction through catheter-mediated approaches involving either temporary (ischemia-reperfusion) or permanent (thrombus injection or embolic coil) occlusions. These techniques are relatively low in invasiveness, while infarct size with corresponding cardiac dysfunction can be controlled by adjusting the location of coronary occlusion. We also describe methods for cardiac angiography and echocardiography in pigs. This is the second edition of a previously published chapter with modifications.


Subject(s)
Disease Models, Animal , Myocardial Infarction , Myocardial Reperfusion Injury , Animals , Myocardial Infarction/therapy , Myocardial Infarction/pathology , Swine , Myocardial Reperfusion Injury/therapy , Myocardial Reperfusion Injury/etiology , Myocardial Reperfusion Injury/pathology , Echocardiography/methods , Coronary Angiography/methods , Embolism/etiology , Embolism/therapy , Embolism/pathology
4.
Article in English | MEDLINE | ID: mdl-38684422

ABSTRACT

PURPOSE: The study aims to assess the effects of dexmedetomidine (Dex) pretreatment on patients during cardiac valve replacement under cardiopulmonary bypass. METHODS: For patients in the Dex group (n = 52), 0.5 µg/kg Dex was given before anesthesia induction, followed by 0.5 µg/kg/h pumping injection before aortic occlusion. For patients in the control group (n = 52), 0.125 ml/kg normal saline was given instead of Dex. RESULTS: The patients in the Dex group had longer time to first dose of rescue propofol than the control group (P = 0.003). The Dex group required less total dosage of propofol than the control group (P = 0.0001). The levels of cardiac troponin I (cTnI), creatine kinase isoenzyme MB (CK-MB), malondialdehyde (MDA), and tumor necrosis factor-α (TNF-α) were lower in the Dex group than the control group at T4, 8 h after the operation (T5), and 24 h after the operation (T6) (P <0.01). The Dex group required less time for mechanical ventilation than the control group (P = 0.003). CONCLUSION: The study suggests that 0.50 µg/kg Dex pretreatment could reduce propofol use and the duration of mechanical ventilation, and confer myocardial protection without increased adverse events during cardiac valve replacement.


Subject(s)
Biomarkers , Cardiopulmonary Bypass , Dexmedetomidine , Heart Valve Prosthesis Implantation , Propofol , Respiration, Artificial , Troponin I , Dexmedetomidine/administration & dosage , Dexmedetomidine/adverse effects , Humans , Cardiopulmonary Bypass/adverse effects , Male , Heart Valve Prosthesis Implantation/adverse effects , Female , Time Factors , Middle Aged , Treatment Outcome , Propofol/adverse effects , Propofol/administration & dosage , Biomarkers/blood , Troponin I/blood , Creatine Kinase, MB Form/blood , Adrenergic alpha-2 Receptor Agonists/adverse effects , Adrenergic alpha-2 Receptor Agonists/administration & dosage , Tumor Necrosis Factor-alpha/blood , Malondialdehyde/blood , Aged , Adult , Anesthetics, Intravenous/adverse effects , Anesthetics, Intravenous/administration & dosage , Myocardial Reperfusion Injury/prevention & control , Myocardial Reperfusion Injury/etiology
5.
Int J Mol Med ; 53(5)2024 05.
Article in English | MEDLINE | ID: mdl-38577949

ABSTRACT

Several studies have shown that berberine (BBR) is effective in protecting against myocardial ischemia­reperfusion injury (MI/RI). However, the precise molecular mechanism remains elusive. The present study observed the mechanism and the safeguarding effect of BBR against hypoxia/reoxygenation (H/R) myocardial injury in H9c2 cells. BBR pretreatment significantly improved the decrease of cell viability, P62 protein, Rho Family GTPase 3 (RhoE) protein, ubiquinone subunit B8 protein, ubiquinol­cytochrome c reductase core protein U, the Bcl­2­associated X protein/B­cell lymphoma 2 ratio, glutathione (GSH) and the GSH/glutathione disulphide (GSSG) ratio induced by H/R, while reducing the increase in lactate dehydrogenase, microtubule­associated protein 1 light 3 protein, caspase­3 activity, reactive oxygen species, GSSG and malonaldehyde caused by H/R. Transmission electron microscopy and LysoTracker Red DND­99 staining results showed that BBR pretreatment inhibited H/R­induced excessive autophagy by mediating RhoE. BBR also inhibited mitochondrial permeability transition, maintained the stability of the mitochondrial membrane potential, reduced the apoptotic rate, and increased the level of caspase­3. However, the protective effects of BBR were attenuated by pAD/RhoE­small hairpin RNA, rapamycin (an autophagy activator) and compound C (an AMP­activated protein kinase inhibitor). These new findings suggested that BBR protects the myocardium from MI/RI by inhibiting excessive autophagy, maintaining mitochondrial function, improving the energy supply and redox homeostasis, and attenuating apoptosis through the RhoE/AMP­activated protein kinase pathway.


Subject(s)
AMP-Activated Protein Kinases , Autophagy , Berberine , Myocardial Reperfusion Injury , AMP-Activated Protein Kinases/metabolism , Apoptosis , Berberine/pharmacology , Caspase 3/metabolism , Glutathione Disulfide/metabolism , Ischemia/metabolism , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/prevention & control , Myocardial Reperfusion Injury/etiology , Myocardium/pathology , Myocytes, Cardiac/metabolism , Animals , Rats
6.
Eur J Pediatr ; 183(6): 2783-2789, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38568244

ABSTRACT

Myocardial injury in open-heart surgery is related to several factors including ischemia-reperfusion injury, generation of reactive oxygen species, increased production of inflammatory mediators, and enhancement of apoptosis of cardiomyocytes. The aim of this study was to study the effect of L-carnitine on myocardial injury in children undergoing open-heart surgery. This clinical trial was performed on 60 children with congenital heart disease (CHD) who underwent open-heart surgery. They were randomized into two groups: L-carnitine group who received L-carnitine 50 mg\kg\day once daily for 1 month before cardiac surgery and control group who received placebo for 1 month before cardiac surgery. Left ventricular cardiac function was assessed by conventional echocardiography to measure left ventricular ejection fraction (LVEF) and two-dimensional speckle tracking echocardiography (2D-STE) to determine left ventricular global longitudinal strain (2D-LV GLS). Blood samples were obtained pre-operatively at baseline before the administration of L-carnitine or placebo and 12 h post-operatively to measure the level of malondialdehyde (MDA), superoxide dismutase (SOD), fas, caspase-3, creatinine kinase-MB (CK-MB), and troponin I. L-carnitine group had significantly lower post-operative level of oxidative stress marker (MDA), apoptosis markers (fas and caspase-3), and myocardial injury markers (CK-MB and troponin I), but they had significantly higher SOD post-operative level compared to the control group. In addition, post-operative LVEF and 2D-LVGLS were significantly lower in the control group compared to L-carnitine group.   Conclusion: L-carnitine can reduce myocardial injury, improve post-operative left ventricular cardiac function, and may provide myocardium protection in children with CHD who underwent open-heart surgery.   Trial registration: The clinical trial was registered at www.pactr.org with registration number PACTR202010570607420 at 29/10/2020 before recruiting the patients. What is Known: • Myocardial injury in open-heart surgery is related to several factors including ischemia-reperfusion injury, generation of reactive oxygen species, increased production of inflammatory mediators, and enhancement of apoptosis of cardiomyocytes. • L-carnitine was reported to have myocardial protective effects in rheumatic valvular surgery and coronary artery bypass graft (CABG) in adults; however, there is no evidence on its effectiveness in children undergoing open-heart surgery. What is New: • L-carnitine significantly lowered the post-operative level of oxidative stress marker (MDA), apoptosis markers (fas and caspase-3), and myocardial injury markers (CK-MB and troponin I) in the treatment group. • L-carnitine can reduce myocardial injury, improve post-operative left ventricular cardiac function, and may provide myocardium protection in children with CHD who underwent open-heart surgery.


Subject(s)
Cardiac Surgical Procedures , Carnitine , Echocardiography , Heart Defects, Congenital , Oxidative Stress , Humans , Carnitine/therapeutic use , Male , Female , Heart Defects, Congenital/surgery , Child, Preschool , Oxidative Stress/drug effects , Cardiac Surgical Procedures/adverse effects , Infant , Apoptosis/drug effects , Myocardial Reperfusion Injury/prevention & control , Myocardial Reperfusion Injury/etiology , Child , Double-Blind Method , Biomarkers/blood , Ventricular Function, Left/drug effects , Postoperative Complications/prevention & control , Postoperative Complications/drug therapy , Treatment Outcome
7.
J Am Heart Assoc ; 13(3): e033102, 2024 Feb 06.
Article in English | MEDLINE | ID: mdl-38293938

ABSTRACT

BACKGROUND: Microvascular obstruction (MVO) and intramyocardial hemorrhage (IMH) are well-established imaging biomarkers of failed myocardial tissue reperfusion in patients with ST-segment elevation-myocardial infarction treated with percutaneous coronary intervention. MVO and IMH are associated with an increased risk of adverse outcome independent of infarct size, but whether the size of the culprit lesion vessel plays a role in the occurrence and severity of reperfusion injury is currently unknown. This study aimed to evaluate the association between culprit lesion vessel size and the occurrence and severity of reperfusion injury as determined by cardiac magnetic resonance imaging. METHODS AND RESULTS: Patients (n=516) with first-time ST-segment-elevation myocardial infarction underwent evaluation with cardiac magnetic resonance at 4 (3-5) days after infarction. MVO was assessed with late gadolinium enhancement imaging and IMH with T2* mapping. Vessel dimensions were determined using catheter-based reference. Median culprit lesion vessel size was 3.1 (2.7-3.6) mm. MVO and IMH were found in 299 (58%) and 182 (35%) patients. Culprit lesion vessel size was associated with body surface area, diabetes, total ischemic time, postinterventional thrombolysis in myocardial infarction flow, and infarct size. There was no association between vessel size and MVO or IMH in univariable and multivariable analysis (P>0.05). These findings were consistent across patient subgroups with left anterior descending artery and non-left anterior descending artery infarctions and those with thrombolysis in myocardial infarction 3 flow post-percutaneous coronary intervention. CONCLUSIONS: Comprehensive characterization of myocardial tissue reperfusion injury by cardiac magnetic resonance revealed no association between culprit lesion vessel size and the occurrence of MVO and IMH in patients treated with primary percutaneous coronary intervention for ST-segment-elevation myocardial infarction.


Subject(s)
Myocardial Infarction , Myocardial Reperfusion Injury , Percutaneous Coronary Intervention , ST Elevation Myocardial Infarction , Humans , Contrast Media , Gadolinium , Magnetic Resonance Imaging , ST Elevation Myocardial Infarction/diagnostic imaging , ST Elevation Myocardial Infarction/therapy , ST Elevation Myocardial Infarction/complications , Myocardial Infarction/therapy , Myocardial Reperfusion Injury/etiology , Myocardial Reperfusion Injury/complications , Myocardial Reperfusion/adverse effects , Hemorrhage/epidemiology , Percutaneous Coronary Intervention/adverse effects , Microcirculation
8.
Med ; 5(1): 10-31, 2024 Jan 12.
Article in English | MEDLINE | ID: mdl-38218174

ABSTRACT

Ischemic heart disease is the greatest health burden and most frequent cause of death worldwide. Myocardial ischemia/reperfusion is the pathophysiological substrate of ischemic heart disease. Improvements in prevention and treatment of ischemic heart disease have reduced mortality in developed countries over the last decades, but further progress is now stagnant, and morbidity and mortality from ischemic heart disease in developing countries are increasing. Significant problems remain to be resolved and require a better pathophysiological understanding. The present review attempts to briefly summarize the state of the art in myocardial ischemia/reperfusion research, with a view on both its coronary vascular and myocardial aspects, and to define the cutting edges where further mechanistic knowledge is needed to facilitate translation to clinical practice.


Subject(s)
Myocardial Ischemia , Myocardial Reperfusion Injury , Humans , Myocardial Reperfusion Injury/etiology , Myocardial Reperfusion Injury/prevention & control , Myocardial Ischemia/prevention & control , Myocardial Reperfusion/adverse effects , Myocardium
9.
J Nanobiotechnology ; 21(1): 421, 2023 Nov 14.
Article in English | MEDLINE | ID: mdl-37957640

ABSTRACT

BACKGROUND: Myocardial ischemia-reperfusion (I/R) injury is accompanied by an imbalance in the cardiac autonomic nervous system, characterized by over-activated sympathetic tone and reduced vagal nerve activity. In our preceding study, we pioneered the development of the magnetic vagus nerve stimulation (mVNS) system. This system showcased precise vagus nerve stimulation, demonstrating remarkable effectiveness and safety in treating myocardial infarction. However, it remains uncertain whether mVNS can mitigate myocardial I/R injury and its specific underlying mechanisms. In this study, we utilized a rat model of myocardial I/R injury to delve into the therapeutic potential of mVNS against this type of injury. RESULTS: Our findings revealed that mVNS treatment led to a reduction in myocardial infarct size, a decrease in ventricular fibrillation (VF) incidence and a curbing of inflammatory cytokine release. Mechanistically, mVNS demonstrated beneficial effects on myocardial I/R injury by inhibiting NLRP3-mediated pyroptosis through the M2AChR/OGDHL/ROS axis. CONCLUSIONS: Collectively, these outcomes highlight the promising potential of mVNS as a treatment strategy for myocardial I/R injury.


Subject(s)
Myocardial Infarction , Myocardial Reperfusion Injury , Vagus Nerve Stimulation , Animals , Rats , Magnetic Phenomena , Myocardial Infarction/therapy , Myocardial Reperfusion Injury/therapy , Myocardial Reperfusion Injury/etiology , NLR Family, Pyrin Domain-Containing 3 Protein , Pyroptosis , Reactive Oxygen Species
10.
EBioMedicine ; 95: 104744, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37556943

ABSTRACT

BACKGROUND: Macrophage infiltration and polarization are integral to the progression of heart failure and cardiac fibrosis after ischemia/reperfusion (IR). Interleukin 34 (IL-34) is an inflammatory regulator related to a series of autoimmune diseases. Whether IL-34 mediates inflammatory responses and contributes to cardiac remodeling and heart failure post-IR remains unclear. METHODS: IL-34 knock-out mice were used to determine the role of IL-34 on cardiac remodeling after IR surgery. Then, immunofluorescence, flow cytometry assays, and RNA-seq analysis were performed to explore the underlying mechanisms of IL-34-induced macrophage recruitment and polarization, and further heart failure after IR. FINDINGS: By re-analyzing single-cell RNA-seq and single-nucleus RNA-seq data of murine and human ischemic hearts, we showed that IL-34 expression was upregulated after IR. IL-34 knockout mitigated cardiac remodeling, cardiac dysfunction, and fibrosis after IR and vice versa. RNA-seq analysis revealed that IL-34 deletion correlated negatively with immune responses and chemotaxis after IR injury. Consistently, immunofluorescence and flow cytometry assays demonstrated that IL-34 deletion attenuated macrophage recruitment and CCR2+ macrophage polarization. Mechanistically, IL-34 deficiency repressed both the canonical and noncanonical NF-κB signaling pathway, leading to marked reduction of P-IKKß and P-IκBα kinase levels; downregulation of NF-κB p65, RelB, and p52 expression, which drove the decline in chemokine CCL2 expression. Finally, IL-34 and CCL2 levels were increased in the serum of acute coronary syndrome patients, with a positive correlation between circulating IL-34 and CCL2 levels in clinical patients. INTERPRETATION: In conclusion, IL-34 sustains NF-κB pathway activation to elicit increased CCL2 expression, which contributes to macrophage recruitment and polarization, and subsequently exacerbates cardiac remodeling and heart failure post-IR. Strategies targeting IL-34-centered immunomodulation may provide new therapeutic approaches to prevent and reverse cardiac remodeling and heart failure in clinical MI patients after percutaneous coronary intervention. FUNDING: This study was supported by the National Nature Science Foundation of China (81670352 and 81970327 to R T, 82000368 to Q F).


Subject(s)
Heart Failure , Interleukins , Myocardial Ischemia , Myocardial Reperfusion Injury , NF-kappa B , Animals , Humans , Mice , Heart Failure/etiology , Heart Failure/metabolism , I-kappa B Kinase/metabolism , Interleukins/genetics , Interleukins/metabolism , Macrophages/metabolism , Mice, Knockout , Myocardial Ischemia/metabolism , Myocardial Reperfusion Injury/etiology , Myocardial Reperfusion Injury/metabolism , Myocardium/metabolism , NF-kappa B/metabolism , Signal Transduction , Ventricular Remodeling
11.
Eur J Pharmacol ; 954: 175865, 2023 Sep 05.
Article in English | MEDLINE | ID: mdl-37406848

ABSTRACT

Excessive autophagy induced by reperfusion is one of the causes of severe myocardial injury. Tanshinone IIA (TSN) protects the myocardium against ischemia/reperfusion (I/R) injury. The mechanism by which the inhibition of excessive autophagy contributes to the myocardial protection by TSN is unclear. The protective effects and mechanisms of TSN were studied in H9c2 cells and rats after anoxia/reoxygenation (A/R)-or I/R-induced myocardial injury. The results showed that after the injury, cell viability decreased, lactate dehydrogenase and caspase 3 activity and apoptosis increased, and autophagy was excessively activated. Further, redox imbalance and energy stress, mitochondrial dysfunction, reduced myocardial function, increased infarct area, and severely damaged morphology were observed in rats. TSN increased 14-3-3η expression and regulated Akt/Beclin1 pathway, inhibited excessive autophagy, and significantly reversed the functional, enzymological and morphological indexes in vivo and in vitro. However, the protective effects of TSN were mimicked by 3-methyladenine (an autophagy inhibitor) and were attenuated by pAD/14-3-3η-shRNA, API-2 (an Akt inhibitor), and rapamycin (an autophagy activator). In conclusion, TSN could increase 14-3-3η expression and regulate Akt/Beclin1 pathway, inhibit excessive autophagy, maintain the mitochondrial function, improve energy supply and redox equilibrium, alleviate apoptosis, and ultimately protect myocardium against I/R injury.


Subject(s)
Myocardial Reperfusion Injury , Proto-Oncogene Proteins c-akt , Rats , Animals , Proto-Oncogene Proteins c-akt/metabolism , Beclin-1/metabolism , Myocytes, Cardiac , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/prevention & control , Myocardial Reperfusion Injury/etiology , Myocardium/metabolism , Apoptosis , Autophagy , Ischemia/metabolism
12.
Medicine (Baltimore) ; 102(9): e33025, 2023 Mar 03.
Article in English | MEDLINE | ID: mdl-36862913

ABSTRACT

BACKGROUND: Cardiac surgery using cardiopulmonary bypass has been shown to cause reversible postischemic cardiac dysfunction and is associated with reperfusion injury and myocardial cell death. Therefore, it is very important to have a series of measures in place to reduce oxygen consumption and provide myocardial protection. We performed a protocol for systematic review and meta-analysis to evaluate the effect of dexmedetomidine administration on myocardial ischemia/reperfusion injury in patients undergoing cardiac surgery with cardiopulmonary bypass. METHODS: This review protocol is registered in the PROSPERO International Prospective Register of systematic reviews, registration number CRD42023386749. A literature search is performed in January 2023 without restriction to regions, publication types or languages. The primary sources were the electronic databases of PubMed, Embase, Web of Science, the Cochrane Central Register of Controlled Trials, Chinese National Knowledge Infrastructure database, Chinese Biomedical Database, and Chinese Science and Technology Periodical database. Risk of bias will be assessed according to the Cochrane Risk of Bias Tool. The meta-analysis is performed using Reviewer Manager 5.4. RESULTS: The results of this meta-analysis will be submitted to a peer-reviewed journal for publication. CONCLUSION: This meta-analysis will evaluate the efficacy and safety of dexmedetomidine in patients undergoing cardiac surgery with cardiopulmonary bypass.


Subject(s)
Cardiac Surgical Procedures , Dexmedetomidine , Myocardial Reperfusion Injury , Humans , Dexmedetomidine/therapeutic use , Myocardial Reperfusion Injury/etiology , Myocardial Reperfusion Injury/prevention & control , Cardiopulmonary Bypass/adverse effects , Systematic Reviews as Topic , Meta-Analysis as Topic , Cardiac Surgical Procedures/adverse effects , Review Literature as Topic
13.
Metabolomics ; 19(2): 12, 2023 02 07.
Article in English | MEDLINE | ID: mdl-36750484

ABSTRACT

INTRODUCTION: Our untargeted metabolic data unveiled that Acyl-CoAs undergo dephosphorylation, however little is known about these novel metabolites and their physiology/pathology relevance. OBJECTIVES: To understand the relationship between acyl-CoAs dephosphorylation and energy status as implied in our previous work, we seek to investigate how ischemia (energy depletion) triggers metabolic changes, specifically acyl-CoAs dephosphorylation in this work. METHODS: Rat hearts were isolated and perfused in Langendorff mode for 15 min followed by 0, 5, 15, and 30 minutes of global ischemia. The heart tissues were harvested for metabolic analysis. RESULTS: As expected, ATP and phosphocreatine were significantly decreased during ischemia. Most short- and medium-chain acyl-CoAs progressively increased with ischemic time from 0 to 15 min, whereas a 30-minute ischemia did not lead to further change. Unlike other acyl-CoAs, propionyl-CoA accumulated progressively in the hearts that underwent ischemia from 0 to 30 min. Progressive dephosphorylation occurred to all assayed acyl-CoAs and free CoA regardless their level changes during the ischemia. CONCLUSION: The present work further confirms that dephosphorylation of acyl-CoAs is an energy-dependent process and how this dephosphorylation is mediated warrants further investigations. It is plausible that dephosphorylation of acyl-CoAs and limited anaplerosis are involved in ischemic injuries to heart. Further investigations are warranted to examine the mechanisms of acyl-CoA dephosphorylation and how the dephosphorylation is possibly involved in ischemic injuries.


Subject(s)
Acyl Coenzyme A , Heart , Metabolomics , Myocardial Ischemia , Animals , Rats , Acyl Coenzyme A/metabolism , Heart/physiopathology , Myocardial Ischemia/etiology , Myocardial Ischemia/metabolism , Myocardial Reperfusion Injury/etiology , Myocardial Reperfusion Injury/metabolism , Phosphorylation , Perfusion/adverse effects , Perfusion/methods
14.
Trends Cardiovasc Med ; 33(6): 357-366, 2023 08.
Article in English | MEDLINE | ID: mdl-35181472

ABSTRACT

Acute myocardial infarction is caused by a sudden coronary artery occlusion and leads to ischemia in the corresponding myocardial territory which generally results in myocardial necrosis. Without restoration of coronary perfusion, myocardial scar formation will cause adverse remodelling of the myocardium and heart failure. Successful introduction of percutaneous coronary intervention and surgical coronary artery bypass grafting made it possible to achieve early revascularisation/reperfusion, hence limiting the ischemic zone of myocardium. However, reperfusion by itself paradoxically triggers an exacerbated and accelerated injury in the myocardium, called ischemia-reperfusion (I/R) injury. This mechanism is partially driven by inflammation through multiple interacting pathways. In this review we summarize the current insights in mechanisms of I/R injury and the influence of altered inflammation. Multiple pharmacological and interventional therapeutic strategies (ischemic conditioning) have proven to be beneficial during I/R in preclinical models but were notoriously unsuccessful upon clinical translation. In this review we focus on common mechanisms of I/R injury, altered inflammation and potential therapeutic strategies. We hypothesize that a dual approach may be of value because I/R injury patients are predestined with multiple comorbidities and systemic low-grade inflammation, which requires targeted intervention before other strategies can be fully effective.


Subject(s)
Myocardial Infarction , Myocardial Ischemia , Myocardial Reperfusion Injury , Humans , Myocardial Reperfusion Injury/prevention & control , Myocardial Reperfusion Injury/etiology , Myocardium/metabolism , Heart , Inflammation/metabolism
15.
Theranostics ; 12(17): 7237-7249, 2022.
Article in English | MEDLINE | ID: mdl-36438504

ABSTRACT

Background: The only effective treatment for myocardial infarction (MI) is the timely restoration of coronary blood flow in the infarcted area, but further reperfusion exacerbates myocardial injury and leads to distal coronary no-reflow, which affects patient prognosis. Angiogenesis could be an important therapeutic strategy for re-establishing the blood supply to save the ischemic myocardium after MI. Basic fibroblast growth factor (bFGF) has been shown to promote angiogenesis. However, direct intravenous administration of bFGF is not a viable option given its poor half-life in vivo. Methods: Herein, we developed a peptide Lys-Lys-Pro-Leu-Gly-Leu-Ala-Gly-Phe-Phe (K2) to encapsulate bFGF to form bFGF@K2 micelle and proposed an enzyme-instructed self-assembly (EISA) strategy to deliver and slowly release bFGF in the ischemic myocardium. Results: The bFGF@K2 micelle exerted a stronger cardioprotective effect than free bFGF in a rat model of myocardial ischemia-reperfusion (MI/R). In vitro results revealed that the bFGF@K2 micelle could be cleaved by matrix metallopeptidase 9 (MMP-9) to yield bFGF@Nanofiber through amphipathic changes. In vivo experiments indicated that intravenous administration of bFGF@K2 micelle could lead to their restructuring into bFGF@Nanofiber and long term retention of bFGF in the ischemic myocardium of rat due to high expression of MMP-9 and assembly-induced retention (AIR) effect, respectively. Twenty-eight days after MI/R model establishment, bFGF@K2 micelle treatment significantly reduced fibrosis and improved cardiac function of the rats. Conclusion: We predict that our strategy could be applied in clinic for MI treatment in the future.


Subject(s)
Fibroblast Growth Factor 2 , Matrix Metalloproteinase 9 , Myocardial Infarction , Myocardial Reperfusion Injury , Nanofibers , Animals , Rats , Fibroblast Growth Factor 2/administration & dosage , Fibroblast Growth Factor 2/therapeutic use , Matrix Metalloproteinase 9/metabolism , Micelles , Myocardial Infarction/complications , Myocardial Infarction/drug therapy , Myocardial Infarction/metabolism , Myocardial Infarction/therapy , Myocardium/metabolism , Myocardium/pathology , Nanofibers/administration & dosage , Nanofibers/therapeutic use , Neovascularization, Pathologic , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/etiology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/therapy
16.
Sci Rep ; 12(1): 19349, 2022 11 11.
Article in English | MEDLINE | ID: mdl-36369283

ABSTRACT

To establish a simple myocardial ischemia‒reperfusion injury (MIRI) manifestation grading system based on clinical manifestations and coronary angiography during primary percutaneous coronary intervention (PPCI). All STEMI patients treated with PPCI from June 2018 to November 2019 were included. According to the MIRI manifestation grade, patients were divided into four grades (I-IV). Laboratory and clinical indicators of the patients and the occurrence of major adverse cardiac events (MACEs) within one year of follow-up were analyzed. A total of 300 patients were included. The higher the MIRI manifestation grade, the lower was the high-density lipoprotein cholesterol (HDL-C); the higher were the C-reactive protein (CRP), lipoprotein(a) [LP(a)], and peak levels of high-sensitivity troponin T (hs-cTnT), creatine kinase (CK-MB), and N-terminal pro-B-type natriuretic peptide (NT-proBNP); and the higher were the proportions of right coronary artery (RCA) and multivessel lesions (P < 0.05). The left ventricular end-diastolic dimension (LVEDD) and E/e' values of patients with higher grades were significantly increased, while the LVEF, left ventricular short-axis functional shortening (LVFS) and E/A values were significantly decreased (P < 0.05). The one-year cumulative incidence of major adverse cardiac events (MACEs) in patients with grade I-IV disease was 7.7% vs. 26.9% vs. 48.4% vs. 93.3%, respectively, P < 0.05. The higher the MIRI manifestation grade, the more obvious is the impact on diastolic and systolic function and the higher is the cumulative incidence of MACEs within one year, especially in patients with multivessel disease, low HDL-C, high CRP, high LP(a) levels, and the RCA as the infarction-related artery.


Subject(s)
Myocardial Reperfusion Injury , Percutaneous Coronary Intervention , ST Elevation Myocardial Infarction , Humans , ST Elevation Myocardial Infarction/surgery , ST Elevation Myocardial Infarction/etiology , Biomarkers , Percutaneous Coronary Intervention/adverse effects , Creatine Kinase, MB Form , Myocardial Reperfusion Injury/etiology , C-Reactive Protein/metabolism
17.
Int J Mol Sci ; 23(19)2022 Oct 03.
Article in English | MEDLINE | ID: mdl-36233031

ABSTRACT

CXCR4 antagonists have been claimed to reduce mortality after myocardial infarction in myocardial infarction (MI) animals, presumably due to suppressing inflammatory responses caused by myocardial ischemia-reperfusion injury, thus, subsequently facilitating tissue repair and cardiac function recovery. This study aims to determine whether a newly designed CXCR4 antagonist DBPR807 could exert better vascular-protective effects than other clinical counterparts (e.g., AMD3100) to alleviate cardiac damage further exacerbated by reperfusion. Consequently, we find that instead of traditional continuous treatment or multiple-dose treatment at different intervals of time, a single-dose treatment of DBPR807 before reperfusion in MI animals could attenuate inflammation via protecting oxidative stress damage and preserve vascular/capillary density and integrity via mobilizing endothelial progenitor cells, leading to a desirable fibrosis reduction and recovery of cardiac function, as evaluated with the LVEF (left ventricular ejection fraction) in infarcted hearts in rats and mini-pigs, respectively. Thus, it is highly suggested that CXCR4 antagonists should be given at a single high dose prior to reperfusion to provide the maximal cardiac functional improvement. Based on its favorable efficacy and safety profiles indicated in tested animals, DBPR807 has a great potential to serve as an adjunctive medicine for percutaneous coronary intervention (PCI) therapies in acute MI patients.


Subject(s)
Myocardial Infarction , Myocardial Reperfusion Injury , Percutaneous Coronary Intervention , Receptors, CXCR4 , Animals , Myocardial Infarction/therapy , Myocardial Reperfusion Injury/etiology , Rats , Receptors, CXCR4/antagonists & inhibitors , Stroke Volume , Swine , Swine, Miniature , Ventricular Function, Left
18.
STAR Protoc ; 3(4): 101751, 2022 12 16.
Article in English | MEDLINE | ID: mdl-36209428

ABSTRACT

Myocardial infarction (MI) is the primary cause of death worldwide, but there are no clinically relevant models to study MI. Here, we describe an ischemia/reperfusion (I/R) injury model typical of MI using mouse or human 3D in vitro cardiac spheroids (CSs). First, we demonstrated the culture and maintenance of CSs. Then, we detailed how to expose CSs to pathophysiological oxygen concentrations to induce I/R injury. The protocol can be used in combination with viability, contractility, and mRNA expression level measurements. For complete details on the use and execution of this protocol, please refer to Sharma et al. (2022).


Subject(s)
Myocardial Infarction , Myocardial Reperfusion Injury , Humans , Mice , Animals , Myocardial Reperfusion Injury/etiology , Disease Models, Animal , Heart , Myocardial Infarction/complications
19.
Int J Cardiol ; 368: 62-68, 2022 12 01.
Article in English | MEDLINE | ID: mdl-35987313

ABSTRACT

BACKGROUND: Nicorandil, an adenosine triphosphate-sensitive potassium channel agonist and nitric oxide donor, is a coronary vasodilator used to treat ischemia-induced chest pain, but it's potential cardioprotective benefits during open heart surgery have not been thoroughly investigated. The study objective was to assess the impact of nicorandil on postoperative ventricular dysfunction and end-organ injury in an established experimental model of open-heart surgery with cardiopulmonary bypass (CPB) and cardioplegic arrest. We hypothesized that nicorandil would attenuate myocardial ischemia-reperfusion (IR) injury, preserve ventricular function, and reduce end-organ injury. METHODS: Rabbits were cannulated for CPB, followed by 60 min of aortic cross-clamp (ACC) with cold cardioplegic arrest, and 120 min of recovery after ACC removal. Nicorandil (or normal saline vehicle) was given intravenously 5 min before ACC and continued throughout the recovery period. Left ventricular developed pressure (LVDP), systolic contractility (LV + dP/dt), and diastolic relaxation (LV -dP/dt) were continuously recorded, and blood and tissue samples were collected for measurement of oxidant stress (OS), inflammation, apoptosis, and organ injury. RESULTS: Nicorandil significantly attenuated IR-induced LV dysfunction compared to saline control (R-120: LV + dP/dt: 1596 ± 397 vs. 514 ± 269 mmHg/s, p = 0.010; LV -dP/dt: -1524 ± 432 vs. -432 ± 243 mmHg/s, p < 0.001; LVDP: 55 ± 11 vs. 22 ± 5 mmHg, p = 0.046). Furthermore, nicorandil inhibited IR-induced increases in OS, inflammation, apoptosis, and organ injury. CONCLUSIONS: Nicorandil exhibits myocardial protection by attenuation of IR-induced LV dysfunction associated with OS, inflammation, apoptosis, and organ injury. Nicorandil should be explored further as a potential therapeutic strategy for limiting global IR injury during open-heart surgery in humans.


Subject(s)
Myocardial Reperfusion Injury , Ventricular Dysfunction , Adenosine Triphosphate , Animals , Cardiopulmonary Bypass/adverse effects , Humans , Inflammation/drug therapy , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/etiology , Myocardial Reperfusion Injury/prevention & control , Nicorandil/pharmacology , Nicorandil/therapeutic use , Nitric Oxide Donors/therapeutic use , Oxidants , Potassium Channels , Rabbits , Saline Solution , Vasodilator Agents/pharmacology , Vasodilator Agents/therapeutic use
20.
Asian Cardiovasc Thorac Ann ; 30(8): 894-905, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35837687

ABSTRACT

BACKGROUND: It remains unclear whether the Rho-kinase (ROCK) inhibition in combination with mechanical circulatory support (MCS) had a synergic protective effect on myocardial ischemia (MI)/reperfusion injury in therapeutic strategies for acute myocardial infarction (AMI). We report the results of an approach using a rat model consisting of a miniaturized cardiopulmonary bypass (CPB) and AMI. METHODS: A total of 25 male Wistar rats were randomized into 5 groups: (1) Sham: a suture was passed under the left anterior descending artery (LAD) creating no MI. A vehicle solution (0.9% saline) was injected intraperitoneally. (2) Myocardial ischemia (MI) + vehicle (MI + V): LAD was ligated for 30 min and reperfused for 120 min, followed by administration of vehicle solution. (3) MI + fasudil (MI + F): the work sequence of group 2, but the selective ROCK inhibitor fasudil (10 mg/kg) was administered instead. (4) MI + V + CPB: CPB was initiated 15 min after the ligation of the LAD to the end of the reperfusion, in addition to the work sequence in group 2. (5) In the MI + F + CPB group, the work sequence of group 4, but with fasudil administration (10 mg/kg). RESULTS: Measurements of cardiac function through conductance catheter indicated that the drop of + dP/dt after reperfusion was moderately limited in MI + F + CPB (vs. MI + V, dP/dt p = 0.22). The preload recruitable stroke work was moderately improved in the MI + F + CPB (p = 0.23) compared with the corresponding control animals (MI + V). Phosphorylated protein kinase B expression in the MI + V + CPB and MI + F + CPB was higher than that in MI + V (p = 0.33). CONCLUSION: Therefore, fasudil administration with MCS resulted in a moderately better left ventricular performance.


Subject(s)
Coronary Artery Disease , Myocardial Infarction , Myocardial Ischemia , Myocardial Reperfusion Injury , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , Animals , Humans , Male , Myocardial Reperfusion Injury/etiology , Myocardial Reperfusion Injury/prevention & control , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , Saline Solution/therapeutic use , Treatment Outcome , rho-Associated Kinases/metabolism , rho-Associated Kinases/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...