Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 33
Filtrer
1.
Pharmacol Res ; 207: 107305, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39002868

RÉSUMÉ

Cardiomyopathy (CM) represents a heterogeneous group of diseases primarily affecting cardiac structure and function, with genetic and epigenetic dysregulation playing a pivotal role in its pathogenesis. Emerging evidence from the burgeoning field of epitranscriptomics has brought to light the significant impact of various RNA modifications, notably N6-methyladenosine (m6A), 5-methylcytosine (m5C), N7-methylguanosine (m7G), N1-methyladenosine (m1A), 2'-O-methylation (Nm), and 6,2'-O-dimethyladenosine (m6Am), on cardiomyocyte function and the broader processes of cardiac and vascular remodelling. These modifications have been shown to influence key pathological mechanisms including mitochondrial dysfunction, oxidative stress, cardiomyocyte apoptosis, inflammation, immune response, and myocardial fibrosis. Importantly, aberrations in the RNA methylation machinery have been observed in human CM cases and animal models, highlighting the critical role of RNA methylating enzymes and their potential as therapeutic targets or biomarkers for CM. This review underscores the necessity for a deeper understanding of RNA methylation processes in the context of CM, to illuminate novel therapeutic avenues and diagnostic tools, thereby addressing a significant gap in the current management strategies for this complex disease.


Sujet(s)
Cardiomyopathies , Épigenèse génétique , ARN , Humains , Animaux , Cardiomyopathies/génétique , Cardiomyopathies/métabolisme , Cardiomyopathies/thérapie , ARN/génétique , ARN/métabolisme , Méthylation ,
2.
Autophagy ; 20(8): 1712-1722, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38522082

RÉSUMÉ

MCOLN1/TRPML1 is a nonselective cationic channel specifically localized to the late endosome and lysosome. With its property of mediating the release of several divalent cations such as Ca2+, Zn2+ and Fe2+ from the lysosome to the cytosol, MCOLN1 plays a pivotal role in regulating a variety of cellular events including endocytosis, exocytosis, lysosomal biogenesis, lysosome reformation, and especially in Macroautophagy/autophagy. Autophagy is a highly conserved catabolic process that maintains cytoplasmic integrity by removing superfluous proteins and damaged organelles. Acting as the terminal compartments, lysosomes are crucial for the completion of the autophagy process. This review delves into the emerging role of MCOLN1 in controlling the autophagic process by regulating lysosomal ionic homeostasis, thereby governing the fundamental functions of lysosomes. Furthermore, this review summarizes the physiological relevance as well as molecular mechanisms through which MCOLN1 orchestrates autophagy, consequently influencing mitochondria turnover, cell apoptosis and migration. In addition, we have illustrated the implications of MCOLN1-regulated autophagy in the pathological process of cancer and myocardial ischemia-reperfusion (I/R) injury. In summary, given the involvement of MCOLN1-mediated autophagy in the pathogenesis of cancer and myocardial I/R injury, targeting MCOLN1 May provide clues for developing new therapeutic strategies for the treatment of these diseases. Exploring the regulation of MCOLN1-mediated autophagy in diverse diseases contexts will surely broaden our understanding of this pathway and offer its potential as a promising drug target.Abbreviation: CCCP:carbonyl cyanide3-chlorophenylhydrazone; CQ:chloroquine; HCQ: hydroxychloroquine;I/R: ischemia-reperfusion; MAP1LC3/LC3:microtubule associated protein 1 light chain 3; MCOLN1/TRPML1:mucolipin TRP cation channel 1; MLIV: mucolipidosis type IV; MTORC1:MTOR complex 1; ROS: reactive oxygenspecies; SQSTM1/p62: sequestosome 1.


Sujet(s)
Autophagie , Lysosomes , Canaux cationiques TRP , Humains , Autophagie/physiologie , Lysosomes/métabolisme , Animaux , Canaux cationiques TRP/métabolisme , Tumeurs/métabolisme , Tumeurs/anatomopathologie , Tumeurs/traitement médicamenteux , Lésion de reperfusion myocardique/métabolisme , Lésion de reperfusion myocardique/anatomopathologie
3.
Environ Toxicol ; 39(3): 1682-1699, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38041472

RÉSUMÉ

This study aims to explore the roles of phenylacetyl glutamine (PAGln) on myocardial infarction (MI) pathogenesis. Here, using targeted metabolomics analysis, it was found that the plasma metabolite PAGln was upregulated in coronary artery disease (CAD) patients and MI mice and could be an independent risk factor for CAD. In vivo and in vitro functional experiments revealed that PAGln pretreatment enhanced MI-induced myocardial injury and cardiac fibrosis, as evident by the increased infarct size, cardiomyocyte death, and the upregulated expression of cardiac fibrosis markers (Col1a1 and α-SMA). Combined with RNA-sequencing analysis and G protein-coupled receptor (GPCR) inhibitor, we found that the GPCR signaling activation is essential for PAGln-mediated effects on cardiomyocyte death. Furthermore, drug affinity responsive target stability and cellular thermal shift assay demonstrated that PAGln could interact with ß1-adrenergic receptor (AR). Moreover, ß1-AR blocker treatment indeed extended the cardiac remodeling after PAGln-enhanced MI. These results suggest that PAGln might be a potential therapeutic target for extending the cardiac remodeling window in MI patients that signals via ß1-AR.


Sujet(s)
Infarctus du myocarde , Myocytes cardiaques , Humains , Souris , Animaux , Myocytes cardiaques/métabolisme , Glutamine/métabolisme , Glutamine/usage thérapeutique , Remodelage ventriculaire , Infarctus du myocarde/traitement médicamenteux , Fibrose , Récepteurs adrénergiques/métabolisme , Récepteurs adrénergiques/usage thérapeutique , Myocarde/métabolisme
4.
Article de Chinois | WPRIM (Pacifique Occidental) | ID: wpr-1021686

RÉSUMÉ

BACKGROUND:N6-methyladenosine(m6A)is a hot research topic in the mechanism of pathological cardiac remodeling and plays an important role in the development of cardiovascular diseases. OBJECTIVE:To summarize the possible mechanism by which m6A modification in non-coding RNAs regulates the main processes of pathological cardiac remodeling,such as pathological cardiac hypertrophy,cardiomyocyte death,myocardial fibrosis and vascular remodeling. METHODS:"m6A,non-coding RNA,pathological cardiac hypertrophy,cardiomyocyte apoptosis,cardiomyocyte pyroptosis,cardiomyocyte ferroptosis,myocardial fibrosis,vascular remodeling"were used as search terms in Chinese and English.Relevant literature from CNKI,PubMed and Web of Science databases published from January 1974 to April 2023 was retrieved,and finally 86 eligible articles were reviewed. RESULTS AND CONCLUSION:m6A modification is a highly dynamic and reversible modification.Pathological cardiac remodeling mainly involves pathological cardiac hypertrophy,cardiomyocyte apoptosis,cardiomyocyte pyroptosis,cardiomyocyte ferroptosis,myocardial fibrosis and vascular remodeling.m6A-related enzymes can regulate pathological cardiac remodeling processes through various non-coding RNAs and different signaling pathways,which can be used as a new potential intervention for cardiovascular diseases.In pathological cardiac remodeling,research on the regulatory relationship between m6A modification and non-coding RNAs is still in its infancy.With the development of epigenetics,m6A modification in non-coding RNAs is expected to have a new development in the regulation of pathological cardiac remodeling.

5.
Int J Mol Sci ; 23(24)2022 Dec 09.
Article de Anglais | MEDLINE | ID: mdl-36555270

RÉSUMÉ

Acute myocardial infarction (AMI) is one of the major leading causes of death in humans globally. Recently, increased levels of recruited macrophages and AGE-albumin were observed in the hearts of humans and animals with acute myocardial infarction. Thus, the purposes of this study were to investigate whether the elevated levels of AGE-albumin from activated macrophage cells are implicated in ischemia-induced cardiomyocyte death and to develop therapeutic strategies for AMI based on its underlying molecular mechanisms with respect to AGEs. The present study demonstrated that activated macrophages and AGE-albumin were observed in heart tissues obtained from humans and rats with AMI incidences. In the cellular model of AMI, it was found that increased expression of AGE-albumin was shown to be co-localized with macrophages, and the presence of AGE-albumin led to increased expression of RAGE through the mitogen-activated protein kinase pathway. After revealing cardiomyocyte apoptosis induced by toxicity of the AGE-RAGE system, sRAGE-secreting MSCs were generated using the CRISPR/Cas9 platform to investigate the therapeutic effects of sRAGE-MSCs in an AMI rat model. Gene-edited sRAGE-MSCs showed greater therapeutic effects against AMI pathogenesis in rat models compared to mock MSCs, and promising results of the functional improvement of stem cells could result in significant improvements in the clinical management of cardiovascular diseases.


Sujet(s)
Infarctus du myocarde , Rodentia , Humains , Rats , Animaux , Infarctus du myocarde/métabolisme , Transduction du signal , Reperfusion , Albumines
6.
Kaohsiung J Med Sci ; 38(11): 1093-1102, 2022 Nov.
Article de Anglais | MEDLINE | ID: mdl-36254861

RÉSUMÉ

Myocardial ischemia/reperfusion (I/R) injury is associated with the poor outcome and higher mortality after myocardial infarction. Recent studies have revealed that miR-199a-5p participates in the process of myocardial I/R injury, but the precise roles and molecular mechanisms of miR-199a-5p in myocardial I/R injury remain not well-studied. Ferroptosis has been proposed to promote cardiomyocyte death, closely associated with myocardial I/R injury. Herein, the present study aimed to explore the function and mechanisms by which miR-199a-5p regulates whether miR-199a-5p contributes to ferroptosis-induced cardiomyocyte death responding to oxygen-glucose deprivation/reoxygenation (OGD/R) injury, an in vitro model of myocardial I/R injury focusing on Akt/eNOS signaling pathway. The results found that ferroptosis-induced cardiomyocyte death occurs and is accompanied by an increase in miR-199a-5p level in OGD/R-treated H9c2 cells. MiR-199a-5p inhibitor ameliorated ferroptosis-induced cardiomyocyte death as evidenced by the increased cell viability, the reduced reactive oxygen species (ROS) generation, lactate dehydrogenase (LDH) activity, malondialdehyde (MDA) and Fe2+ contents, and the up-regulated glutathione (GSH)/glutathione disulphide (GSSG) ratio as well as glutathione peroxidase 4 (Gpx4) protein expression in H9c2 cells-exposed to OGD/R, while miR-199a-5p mimic had the opposite effects. In addition, OGD/R led to the inhibition of Akt/eNOS signaling pathway, which was also blocked by miR-199a-5p inhibitor and aggravated by miR-199a-5p mimic. Furthermore, LY294002, an inhibitor of Akt/eNOS signaling pathway, abrogated miR-199a-5p inhibitor-induced the reduction of ferroptosis-induced cardiomyocyte death. In summary, our findings demonstrated that miR-199a-5p plays a central role in stimulating ferroptosis-induced cardiomyocyte death during ischemic/hypoxic injury via inhibiting Akt/eNOS signaling pathway.


Sujet(s)
Ferroptose , microARN , Lésion de reperfusion myocardique , Humains , Apoptose , Ferroptose/génétique , Glucose/métabolisme , microARN/métabolisme , Lésion de reperfusion myocardique/génétique , Myocytes cardiaques/métabolisme , Oxygène/métabolisme , Protéines proto-oncogènes c-akt/génétique , Protéines proto-oncogènes c-akt/métabolisme , Transduction du signal , Nitric oxide synthase type III
7.
Antioxidants (Basel) ; 11(8)2022 Jul 27.
Article de Anglais | MEDLINE | ID: mdl-36009181

RÉSUMÉ

RATIONALE: Intermittent hypoxia (IH) is one of the main features of sleep-disordered breathing (SDB). Recent findings indicate that hypoxia inducible factor-1 (HIF-1) promotes cardiomyocytes apoptosis during chronic IH, but the mechanisms involved remain to be elucidated. Here, we hypothesize that IH-induced ER stress is associated with mitochondria-associated ER membrane (MAM) alteration and mitochondrial dysfunction, through HIF-1 activation. METHODS: Right atrial appendage biopsies from patients with and without SDB were used to determine HIF-1α, Grp78 and CHOP expressions. Wild-type and HIF-1α+/- mice were exposed to normoxia (N) or IH (21-5% O2, 60 cycles/h, 8 h/day) for 21 days. Expressions of HIF-1α, Grp78 and CHOP, and apoptosis, were measured by Western blot and immunochemistry. In isolated cardiomyocytes, we examined structural integrity of MAM by proximity ligation assay and their function by measuring ER-to-mitochondria Ca2+ transfer by confocal microscopy. Finally, we measured mitochondrial respiration using oxygraphy and calcium retention capacity (CRC) by spectrofluorometry. MAM structure was also investigated in H9C2 cells incubated with 1 mM CoCl2, a potent HIF-1α inducer. RESULTS: In human atrial biopsies and mice, IH induced HIF-1 activation, ER stress and apoptosis. IH disrupted MAM, altered Ca2+ homeostasis, mitochondrial respiration and CRC. Importantly, IH had no effect in HIF-1α+/- mice. Similar to what observed under IH, HIF-1α overexpression was associated with MAM alteration in H9C2. CONCLUSION: IH-induced ER stress, MAM alterations and mitochondrial dysfunction were mediated by HIF-1; all these intermediate mechanisms ultimately inducing cardiomyocyte apoptosis. This suggests that HIF-1 modulation might limit the deleterious cardiac effects of SDB.

8.
Mol Med Rep ; 26(2)2022 Aug.
Article de Anglais | MEDLINE | ID: mdl-35703348

RÉSUMÉ

Sepsis­induced cardiac dysfunction is one of the most common types of organ dysfunction in sepsis; its pathogenesis is highly complex and not yet fully understood. Cardiomyocytes serve a key role in the pathophysiology of cardiac function; due to the limited ability of cardiomyocytes to regenerate, their loss contributes to decreased cardiac function. The activation of inflammatory signalling pathways affects cardiomyocyte function and modes of cardiomyocyte death in sepsis. Prevention of cardiomyocyte death is an important therapeutic strategy for sepsis­induced cardiac dysfunction. Thus, understanding the signalling pathways that activate cardiomyocyte death and cross­regulation between death modes are key to finding therapeutic targets. The present review focused on advances in understanding of sepsis­induced cardiomyocyte death pathways, including apoptosis, necroptosis, mitochondria­mediated necrosis, pyroptosis, ferroptosis and autophagy. The present review summarizes the effect of inflammatory activation on cardiomyocyte death mechanisms, the diversity of regulatory mechanisms and cross­regulation between death modes and the effect on cardiac function in sepsis to provide a theoretical basis for treatment of sepsis­induced cardiac dysfunction.


Sujet(s)
Cardiopathies , Sepsie , Apoptose , Autophagie , Cardiopathies/métabolisme , Humains , Myocytes cardiaques/métabolisme , Sepsie/métabolisme
9.
Front Cardiovasc Med ; 9: 847012, 2022.
Article de Anglais | MEDLINE | ID: mdl-35497981

RÉSUMÉ

Chemotherapy and targeted therapies have significantly improved the prognosis of oncology patients. However, these antineoplastic treatments may also induce adverse cardiovascular effects, which may lead to acute or delayed onset of cardiac dysfunction. These common cardiovascular complications, commonly referred to as cardiotoxicity, not only may require the modification, suspension, or withdrawal of life-saving antineoplastic therapies, with the risk of reducing their efficacy, but can also strongly impact the quality of life and overall survival, regardless of the oncological prognosis. The onset of cardiotoxicity may depend on the class, dose, route, and duration of administration of anticancer drugs, as well as on individual risk factors. Importantly, the cardiotoxic side effects may be reversible, if cardiac function is restored upon discontinuation of the therapy, or irreversible, characterized by injury and loss of cardiac muscle cells. Subclinical myocardial dysfunction induced by anticancer therapies may also subsequently evolve in symptomatic congestive heart failure. Hence, there is an urgent need for cardioprotective therapies to reduce the clinical and subclinical cardiotoxicity onset and progression and to limit the acute or chronic manifestation of cardiac damages. In this review, we summarize the knowledge regarding the cellular and molecular mechanisms contributing to the onset of cardiotoxicity associated with common classes of chemotherapy and targeted therapy drugs. Furthermore, we describe and discuss current and potential strategies to cope with the cardiotoxic side effects as well as cardioprotective preventive approaches that may be useful to flank anticancer therapies.

10.
Cell Mol Life Sci ; 79(6): 300, 2022 May 19.
Article de Anglais | MEDLINE | ID: mdl-35588335

RÉSUMÉ

Although acute melatonin treatment effectively reduces cardiac ischemia/reperfusion (I/R) injury in lean rats by modulating melatonin receptor 2 (MT2), there is no information regarding the temporal effects of melatonin administration during cardiac I/R injury in prediabetic obese rats. Prediabetic obese rats induced by chronic consumption of a high-fat diet (HFD) were used. The rats underwent a cardiac I/R surgical procedure (30-min of ischemia, followed by 120-min of reperfusion) and were randomly assigned to receive either vehicle or melatonin treatment. In the melatonin group, rats were divided into 3 different subgroups: (1) pretreatment, (2) treatment during ischemic period, (3) treatment at the reperfusion onset. In the pretreatment subgroup either a nonspecific MT blocker (Luzindole) or specific MT2 blocker (4-PPDOT) was also given to the rats prior to melatonin treatment. Pretreatment with melatonin (10 mg/kg) effectively reduced cardiac I/R injury by reducing infarct size, arrhythmia, and LV dysfunction. Reduction in impaired mitochondrial function, mitochondrial dynamic balance, oxidative stress, defective autophagy, and apoptosis were observed in rats pretreated with melatonin. Unfortunately, the cardioprotective benefits were not observed when 10-mg/kg of melatonin was acutely administered to the rats after cardiac ischemia. Thus, we increased the dose of melatonin to 20 mg/kg, and it was administered to the rats during ischemia or at the onset of reperfusion. The results showed that 20-mg/kg of melatonin effectively reduced cardiac I/R injury to a similar extent to the 10-mg/kg pretreatment regimen. The MT2 blocker inhibited the protective effects of melatonin. Acute melatonin treatment during cardiac I/R injury exerted protective effects in prediabetic obese rats. However, a higher dose of melatonin is required when given after the onset of cardiac ischemia. These effects of melatonin were mainly mediated through activation of MT2.


Sujet(s)
Mélatonine , Lésion de reperfusion myocardique , État prédiabétique , Animaux , Mélatonine/pharmacologie , Mélatonine/usage thérapeutique , Lésion de reperfusion myocardique/complications , Lésion de reperfusion myocardique/traitement médicamenteux , Obésité/complications , Obésité/traitement médicamenteux , État prédiabétique/complications , État prédiabétique/traitement médicamenteux , Rats , Rat Wistar
11.
Autophagy ; 18(12): 3053-3055, 2022 12.
Article de Anglais | MEDLINE | ID: mdl-35491864

RÉSUMÉ

Accumulating evidence suggests that macroautophagy/autophagy dysfunction plays a critical role in myocardial ischemia-reperfusion (I/R) injury. However, the underlying mechanisms responsible for malfunctional autophagy in cardiomyocytes subjected to I/R are poorly understood. As a result, there are no effective therapeutic options that target autophagy to prevent myocardial I/R injury. We recently revealed that MCOLN1/TRPML1, a lysosomal cationic channel, directly contributes to the inhibition of autophagic flux in cardiomyocytes post I/R. We found that MCOLN1 is activated secondary to reactive oxygen species (ROS) elevation following I/R, which in turn induces the release of lysosomal zinc into the cytosol. This ultimately blocks autophagic flux in cardiomyocytes by disrupting the fusion between autophagosomes containing engulfed mitochondria and lysosomes. Furthermore, we discovered that the MCOLN1-mediated inhibition of autophagy induced by I/R impairs mitochondrial function, which results in further detrimental ROS release that directly contributes to cardiomyocyte death. More importantly, restoration of blocked autophagic flux in cardiomyocytes subjected to I/R achieved by blocking MCOLN1 channels significantly rescues cardiomyocyte death in vitro and greatly improves cardiac function of mice subjected to I/R in vivo. Therefore, targeting MCOLN1 represents a novel therapeutic strategy to protect against myocardial I/R injury.Abbreviations: I/R: ischemia-reperfusion; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MCOLN1/TRPML1: mucolipin TRP cation channel 1; ROS: reactive oxygen species; SQSTM1/p62: sequestosome 1.


Sujet(s)
Lésion de reperfusion myocardique , Canaux cationiques TRP , Souris , Animaux , Myocytes cardiaques/métabolisme , Autophagie , Espèces réactives de l'oxygène/métabolisme , Lésion de reperfusion myocardique/prévention et contrôle , Autophagosomes/métabolisme , Canaux cationiques TRP/métabolisme
12.
Basic Res Cardiol ; 117(1): 20, 2022 04 07.
Article de Anglais | MEDLINE | ID: mdl-35389129

RÉSUMÉ

Accumulating evidence suggests that autophagy dysfunction plays a critical role in myocardial ischemia/reperfusion (I/R) injury. However, the underling mechanism of malfunctional autophagy in the cardiomyocytes subjected to I/R has not been well defined. As a result, there is no effective therapeutic option by targeting autophagy to prevent myocardial I/R injury. Here, we used both an in vitro and an in vivo I/R model to monitor autophagic flux in the cardiomyocytes, by exposing neonatal rat ventricular myocytes to hypoxia/reoxygenation and by subjecting mice to I/R, respectively. We observed that the autophagic flux in the cardiomyocytes subjected to I/R was blocked in both in vitro and in vivo models. Down-regulating a lysosomal cationic channel, TRPML1, markedly restored the blocked myocardial autophagic flux induced by I/R, demonstrating that TRPML1 directly contributes to the blocked autophagic flux in the cardiomyocytes subjected to I/R. Mechanistically, TRPML1 is activated secondary to ROS elevation following ischemia/reperfusion, which in turn induces the release of lysosomal zinc into the cytosol and ultimately blocks the autophagic flux in cardiomyocytes, presumably by disrupting the fusion between autophagosomes and lysosomes. As a result, the inhibited myocardial autophagic flux induced by TRPML1 disrupted mitochondria turnover and resulted in mass accumulation of damaged mitochondria and further ROS release, which directly led to cardiomyocyte death. More importantly, pharmacological and genetic inhibition of TRPML1 channels greatly reduced infarct size and rescued heart function in mice subjected to I/R in vivo by restoring impaired myocardial autophagy. In summary, our study demonstrates that secondary to ROS elevation, activation of TRPML1 results in autophagy inhibition in the cardiomyocytes subjected to I/R, which directly leads to cardiomyocyte death by disrupting mitochondria turnover. Therefore, targeting TRPML1 represents a novel therapeutic strategy to protect against myocardial I/R injury.


Sujet(s)
Lésion de reperfusion myocardique , Animaux , Apoptose , Autophagie , Souris , Lésion de reperfusion myocardique/traitement médicamenteux , Myocarde , Myocytes cardiaques , Rats , Espèces réactives de l'oxygène
13.
J Cardiovasc Transl Res ; 15(3): 444-455, 2022 06.
Article de Anglais | MEDLINE | ID: mdl-35182317

RÉSUMÉ

Myocardial infarction (MI) causes most of the mortality worldwide. Coronary obstruction-caused myocardial ischemic injury leads to permanent loss of the myocardium. Subsequent compensatory myocardial remodeling and heart failure would result in arrhythmia and even sudden death. The molecular mechanisms of these pathological processes remain to be thoroughly revealed. Circular RNAs (circRNAs) are special types of non-coding RNAs which can durably regulate gene expression and modulate cell fate. They had been reported to mediate ischemic myocardial injury and myocardial remodeling. circRNAs can be loaded into extracellular vesicles and released into extracellular space. More recently, it was uncovered that the extracellular circRNAs can regulate intercellular communications, similar to "first messengers." The cross-talk mediated by extracellular circRNAs had been demonstrated to play important roles in pathological processes. Here, we would like to review the modulation of extracellular circRNAs in ischemic myocardial injury and myocardial remodeling. We believe the extracellular circRNAs can bring new strategies of MI treatment.


Sujet(s)
Vésicules extracellulaires , microARN , Infarctus du myocarde , Vésicules extracellulaires/métabolisme , Humains , microARN/génétique , Infarctus du myocarde/métabolisme , Myocarde/anatomopathologie , ARN circulaire/génétique
14.
Cell Signal ; 92: 110281, 2022 04.
Article de Anglais | MEDLINE | ID: mdl-35151832

RÉSUMÉ

Myocardial infarction (MI), ischemia-reperfusion injury or chemotherapy can trigger excessive loss of terminally differentiated cardiomyocytes, leading to the development of heart failure. Whereas apoptosis has been considered to be the major form of cell death in various myocardial damage, the means by which to reduce cardiomyocyte loss are limited, and the mechanism that underlies cardiomyocyte apoptosis need to be further investigated. PH domain leucine-rich repeat protein phosphatase1 (PHLPP1) belongs to a novel family of Ser/Thr protein phosphatases that functions as a tumor suppressor. Here, we identified PHLPP1 as an important pro-apoptosis factor of cardiomyocytes in response to pathogenic stresses. The conditional PHLPP1 deficiency in cardiomyocytes alleviated myocardial ischemic injury, improved cardiac function and inhibited myocardial fibrosis, in turn preventing adverse cardiac remodeling and heart failure after MI. The conditional PHLPP1 deficiency in cardiomyocytes also attenuated doxorubicin (Dox)-induced myocardial injury, suppressed the inflammation and fibrosis in cardiac tissues, and protected from cardiac dysfunction. Mechanically, PHLPP1 bound the anti-apoptosis protein myeloid cell leukemia sequence 1 (Mcl-1) in cardiomyocytes. Thr163 phosphorylation of Mcl-1 was reported to slow Mcl-1 protein turnover. We further found that PHLPP1 deficiency enhanced Thr163 phosphorylation of Mcl-1, inhibited Mcl-1 degradation and maintained Mcl-1 protein expression level in myocardium and cardiomyocytes upon MI or Dox treatment. PHLPP1 could directly dephosphorylate Thr163 of Mcl-1. Thus, PHLPP1 promotes cardiomyocyte death and cardiac dysfunction through binding and enhancing Mcl-1 degradation under ischemic or toxic injury conditions, which sheds new light on the development of potential therapies to control cardiomyocyte loss.


Sujet(s)
Infarctus du myocarde , Myocytes cardiaques , Animaux , Apoptose , Humains , Souris , Souris de lignée C57BL , Protéine Mcl-1 , Infarctus du myocarde/métabolisme , Myocarde/métabolisme , Myocytes cardiaques/métabolisme , Protéines nucléaires/génétique , Protéines nucléaires/métabolisme , Phosphoprotein Phosphatases/métabolisme
15.
Cardiovasc Res ; 118(4): 1115-1125, 2022 03 16.
Article de Anglais | MEDLINE | ID: mdl-33878183

RÉSUMÉ

AIMS: Acute myocardial infarction causes lethal cardiomyocyte injury during ischaemia and reperfusion (I/R). Histones have been described as important Danger Associated Molecular Proteins (DAMPs) in sepsis. The objective of this study was to establish whether extracellular histone release contributes to myocardial infarction. METHODS AND RESULTS: Isolated, perfused rat hearts were subject to I/R. Nucleosomes and histone-H4 release was detected early during reperfusion. Sodium-ß-O-Methyl cellobioside sulfate (mCBS), a newly developed histone-neutralizing compound, significantly reduced infarct size whilst also reducing the detectable levels of histones. Histones were directly toxic to primary adult rat cardiomyocytes in vitro. This was prevented by mCBS or HIPe, a recently described, histone-H4 neutralizing peptide, but not by an inhibitor of TLR4, a receptor previously reported to be involved in DAMP-mediated cytotoxicity. Furthermore, TLR4-reporter HEK293 cells revealed that cytotoxicity of histone H4 was independent of TLR4 and NF-κB. In an in vivo rat model of I/R, HIPe significantly reduced infarct size. CONCLUSION: Histones released from the myocardium are cytotoxic to cardiomyocytes, via a TLR4-independent mechanism. The targeting of extracellular histones provides a novel opportunity to limit cardiomyocyte death during I/R injury of the myocardium.


Sujet(s)
Infarctus du myocarde , Lésion de reperfusion myocardique , Animaux , Cellules HEK293 , Histone/métabolisme , Humains , Infarctus du myocarde/métabolisme , Infarctus du myocarde/prévention et contrôle , Lésion de reperfusion myocardique/métabolisme , Lésion de reperfusion myocardique/prévention et contrôle , Myocarde/métabolisme , Myocytes cardiaques/métabolisme , Rats , Récepteur de type Toll-4/métabolisme
16.
Front Mol Biosci ; 8: 668129, 2021.
Article de Anglais | MEDLINE | ID: mdl-34141722

RÉSUMÉ

Cardiovascular disease is the leading cause of death worldwide. In spite of the mature managements of myocardial infarction (MI), post-MI reperfusion (I/R) injury results in high morbidity and mortality. Cardiomyocyte Ca2+ overload is a major factor of I/R injury, initiating a cascade of events contributing to cardiomyocyte death and myocardial dysfunction. Ca2+/calmodulin-dependent protein kinase II (CaMKII) plays a critical role in cardiomyocyte death response to I/R injury, whose activation is a key feature of myocardial I/R in causing intracellular mitochondrial swelling, endoplasmic reticulum (ER) Ca2+ leakage, abnormal myofilament contraction, and other adverse reactions. CaMKII is a multifunctional serine/threonine protein kinase, and CaMKIIδ, the dominant subtype in heart, has been widely studied in the activation, location, and related pathways of cardiomyocytes death, which has been considered as a potential targets for pharmacological inhibition. In this review, we summarize a brief overview of CaMKII with various posttranslational modifications and its properties in myocardial I/R injury. We focus on the molecular mechanism of CaMKII involved in regulation of cell death induced by myocardial I/R including necroptosis and pyroptosis of cardiomyocyte. Finally, we highlight that targeting CaMKII modifications and cell death involved pathways may provide new insights to understand the conversion of cardiomyocyte fate in the setting of myocardial I/R injury.

17.
J Adv Res ; 29: 33-44, 2021 03.
Article de Anglais | MEDLINE | ID: mdl-33842003

RÉSUMÉ

Introduction: Previous studies reported the beneficial effects of pretreatment with melatonin on the heart during cardiac ischemia/reperfusion (I/R) injury. However, the effects of melatonin given after cardiac ischemia, as well as its comparative temporal effects are unknown. These include pretreatment, during ischemia, and at the onset of reperfusion. Also, the association between melatonin receptors and cardiac arrhythmias, mitochondrial function and dynamics, autophagy, and mitophagy during cardiac I/R have not been investigated. Objectives: We tested two major hypotheses in this study. Firstly, the temporal effect of melatonin administration exerts different cardioprotective efficacy during cardiac I/R. Secondly, melatonin provides cardioprotective effects via MT2 activation, leading to improvement in cardiac mitochondrial function and dynamics, reduced excessive mitophagy and autophagy, and decreased cardiac arrhythmias, resulting in improved LV function. Methods: Male rats were subjected to cardiac I/R, and divided into 4 intervention groups: vehicle, pretreatment with melatonin, melatonin given during ischemia, and melatonin given at the onset of reperfusion. In addition, either a non-specific melatonin receptor (MT) blocker or specific MT2 blocker was given to rats. Results: Treatment with melatonin at all time points alleviated cardiac I/R injury to a similar extent, quantified by reduction in infarct size, arrhythmia score, LV dysfunction, cardiac mitochondrial dysfunction, imbalance of mitochondrial dynamics, excessive mitophagy, and a decreased Bax/Bcl2 ratio. In H9C2 cells, melatonin increased %cell viability by reducing mitochondrial dynamic imbalance and a decrease in Bax protein expression. The cardioprotective effects of melatonin were dependent on MT2 activation. Conclusion: Melatonin given before or after ischemia exerted equal levels of cardioprotection on the heart with I/R injury, and its beneficial effects on cardiac arrhythmias, cardiac mitochondrial function and dynamics were dependent upon the activation of MT2.


Sujet(s)
Cardiotoniques/pharmacologie , Mélatonine/pharmacologie , Lésion de reperfusion myocardique/traitement médicamenteux , Récepteur de la mélatonine de type MT2/métabolisme , Animaux , Apoptose/effets des médicaments et des substances chimiques , Troubles du rythme cardiaque/traitement médicamenteux , Autophagie/effets des médicaments et des substances chimiques , Cardiotoniques/métabolisme , Lignée cellulaire , Survie cellulaire/effets des médicaments et des substances chimiques , Mâle , Mitochondries du myocarde/effets des médicaments et des substances chimiques , Dynamique mitochondriale/effets des médicaments et des substances chimiques , Mitophagie/effets des médicaments et des substances chimiques , Infarctus du myocarde/traitement médicamenteux , Lésion de reperfusion myocardique/anatomopathologie , Myocytes cardiaques/effets des médicaments et des substances chimiques , Rats , Rat Wistar , Récepteurs à la mélatonine/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Fonction ventriculaire gauche/effets des médicaments et des substances chimiques
18.
Front Cardiovasc Med ; 8: 820416, 2021.
Article de Anglais | MEDLINE | ID: mdl-35127874

RÉSUMÉ

Heart failure is a syndrome in which the heart cannot pump enough blood to meet the body's needs, resulting from impaired ventricular filling or ejection of blood. Heart failure is still a global public health problem and remains a substantial unmet medical need. Therefore, it is crucial to identify new therapeutic targets for heart failure. Ca2+/calmodulin-dependent kinase II (CaMKII) is a serine/threonine protein kinase that modulates various cardiac diseases. CaMKII-δ9 is the most abundant CaMKII-δ splice variant in the human heart and acts as a central mediator of DNA damage and cell death in cardiomyocytes. Here, we proved that CaMKII-δ9 mediated cardiomyocyte death promotes cardiomyopathy and heart failure. However, CaMKII-δ9 did not directly regulate cardiac hypertrophy. Furthermore, we also showed that CaMKII-δ9 induced cell death in adult cardiomyocytes through impairing the UBE2T/DNA repair signaling. Finally, we demonstrated no gender difference in the expression of CaMKII-δ9 in the hearts, together with its related cardiac pathology. These findings deepen our understanding of the role of CaMKII-δ9 in cardiac pathology and provide new insights into the mechanisms and therapy of heart failure.

19.
Front Physiol ; 11: 551318, 2020.
Article de Anglais | MEDLINE | ID: mdl-33192549

RÉSUMÉ

Myocardial ischemia-reperfusion (MI/R) injury is characterized by iron deposition and reactive oxygen species production, which can induce ferroptosis. Ferroptosis has also been proposed to promote cardiomyocyte death. The current study sought to define the mechanism governing cardiomyocyte death in MI/R injury. An animal model of MI/R was established by ligation and perfusion of the left anterior descending coronary artery, and a cellular model of IR was constructed in cardiomyocytes. ChIP assay was then conducted to determine the interaction among USP22, SIRT1, p53, and SLC7A11. Loss- and gain-of-function assays were also conducted to determine the in vivo and in vitro roles of USP22, SIRT1, and SLC7A11. The infarct size and pathological changes of myocardial tissue were observed using TCC and hematoxylin-eosin staining, and the levels of cardiac function- and myocardial injury-related factors of rats were determined. Cardiomyocyte viability and apoptosis were evaluated in vitro, followed by detection of ferroptosis-related indicators (glutathione (GSH), reactive oxygen species, lipid peroxidation, and iron accumulation). USP22, SIRT1, and SLC7A11 expressions were found to be down-regulated, whereas p53 was highly expressed during MI/R injury. USP22, SIRT1, or SLC7A11 overexpression reduced the infarct size and ameliorated pathological conditions, cardiac function, as evidenced by reduced maximum pressure, ejection fraction, maximum pressure rate, and myocardial injury characterized by lower creatine phosphokinase and lactate dehydrogenase levels in vivo. Moreover, USP22, SIRT1, or SLC7A11 elevation contributed to enhanced cardiomyocyte viability and attenuated ferroptosis-induced cell death in vitro, accompanied by increased GSH levels, as well as decreased reactive oxygen species production, lipid peroxidation, and iron accumulation. Together, these results demonstrate that USP22 overexpression could inhibit ferroptosis-induced cardiomyocyte death to protect against MI/R injury via the SIRT1/p53/SLC7A11 association.

20.
BMC Cardiovasc Disord ; 20(1): 310, 2020 06 29.
Article de Anglais | MEDLINE | ID: mdl-32600304

RÉSUMÉ

BACKGROUND: Correct detection of human cardiomyocyte death is essential for definitive diagnosis and appropriate management of cardiovascular diseases. Although current strategies have proven utility in clinical cardiology, they have some limitations. Our aim was to develop a new approach to monitor myocardial death using methylation patterns of circulating cell-free DNA (cf-DNA). METHODS: We first examined the methylation status of FAM101A in heart tissue and blood of individual donors using quantitative methylation-sensitive PCR (qMS-PCR). The concentrations and kinetics of cardiac cf-DNA in plasma from five congenital heart disease (CHD) children before and after they underwent cardiac surgery at serial time points were then investigated. RESULTS: We identified demethylated FAM101A specifically present in heart tissue. Importantly, our time course experiments demonstrated that the plasma cardiac cf-DNA level increased quickly during the early post-cardiac surgery phase, peaking at 4-6 h, decreased progressively (24 h) and returned to baseline (72 h). Moreover, cardiac cf-DNA concentrations pre- and post-operation were closely correlated with plasma troponin levels. CONCLUSIONS: We proposed a novel strategy for the correct detection of cardiomyocyte death, based on analysis of plasma cf-DNA carrying the cardiac-specific methylation signature. Our pilot study may lead to new tests for human cardiac pathologies.


Sujet(s)
Acides nucléiques acellulaires/génétique , Méthylation de l'ADN , Cardiopathies congénitales/génétique , Cardiopathies congénitales/anatomopathologie , Myocytes cardiaques/anatomopathologie , Procédures de chirurgie cardiaque , Mort cellulaire , Enfant d'âge préscolaire , Épigénome , Femelle , Cardiopathies congénitales/sang , Cardiopathies congénitales/chirurgie , Humains , Nourrisson , Nouveau-né , Mâle , Protéines des microfilaments/génétique , Projets pilotes , Facteurs temps , Résultat thérapeutique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE