Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 2.539
Filtrer
1.
BMC Cardiovasc Disord ; 24(1): 339, 2024 Jul 04.
Article de Anglais | MEDLINE | ID: mdl-38965461

RÉSUMÉ

BACKGROUND: Zolpidem is a non-benzodiazepine hypnotic widely used to manage insomnia. Zolpidem-triggered atrial fibrillation (AF) in patients with cardiomyopathy has never been reported before. CASE PRESENTATION: A 40-year-old man with Duchenne muscular dystrophy-related cardiomyopathy attempted suicide and developed new-onset AF after zolpidem overdose. One year before admission, the patient visited our clinic due to chest discomfort and fatigue after daily walks for 1 month; both electrocardiography (ECG) and 24-hour Holter ECG results did not detect AF. After administration of cardiac medication (digoxin 0.125 mg/day, spironolactone 40 mg/day, furosemide 20 mg/day, bisoprolol 5 mg/day, sacubitril/valsartan 12/13 mg/day), he felt better. AF had never been observed before this admission via continuous monitoring during follow-up. Sixteen days before admission, the patient saw a sleep specialist and started zolpidem tartrate tablets (10 mg/day) due to insomnia for 6 months; ECG results revealed no significant change. The night before admission, the patient attempted suicide by overdosing on 40 mg of zolpidem after an argument, which resulted in severe lethargy. Upon admission, his ECG revealed new-onset AF, necessitating immediate cessation of zolpidem. Nine hours into admission, AF spontaneously terminated into normal sinus rhythm. Results from the ECG on the following days and the 24-hour Holter ECG at 1-month follow-up showed that AF was not detected. CONCLUSIONS: This study provides valuable clinical evidence indicating that zolpidem overdose may induce AF in patients with cardiomyopathy. It serves as a critical warning for clinicians when prescribing zolpidem, particularly for patients with existing heart conditions. Further large-scale studies are needed to validate this finding and to explore the mechanisms between zolpidem and AF.


Sujet(s)
Fibrillation auriculaire , Cardiomyopathies , Zolpidem , Humains , Zolpidem/effets indésirables , Mâle , Fibrillation auriculaire/traitement médicamenteux , Fibrillation auriculaire/diagnostic , Fibrillation auriculaire/induit chimiquement , Adulte , Cardiomyopathies/induit chimiquement , Cardiomyopathies/physiopathologie , Cardiomyopathies/diagnostic , Tentative de suicide , Mauvais usage des médicaments prescrits/diagnostic , Rythme cardiaque/effets des médicaments et des substances chimiques , Pyridines/effets indésirables
2.
J Am Heart Assoc ; 13(12): e033733, 2024 Jun 18.
Article de Anglais | MEDLINE | ID: mdl-38860414

RÉSUMÉ

BACKGROUND: Chronic sympathetic stimulation drives desensitization and downregulation of ß1 adrenergic receptor (ß1AR) in heart failure. We aim to explore the differential downregulation subcellular pools of ß1AR signaling in the heart. METHODS AND RESULTS: We applied chronic infusion of isoproterenol to induced cardiomyopathy in male C57BL/6J mice. We applied confocal and proximity ligation assay to examine ß1AR association with L-type calcium channel, ryanodine receptor 2, and SERCA2a ((Sarco)endoplasmic reticulum calcium ATPase 2a) and Förster resonance energy transfer-based biosensors to probe subcellular ß1AR-PKA (protein kinase A) signaling in ventricular myocytes. Chronic infusion of isoproterenol led to reduced ß1AR protein levels, receptor association with L-type calcium channel and ryanodine receptor 2 measured by proximity ligation (puncta/cell, 29.65 saline versus 14.17 isoproterenol, P<0.05), and receptor-induced PKA signaling at the plasma membrane (Förster resonance energy transfer, 28.9% saline versus 1.9% isoproterenol, P<0.05) and ryanodine receptor 2 complex (Förster resonance energy transfer, 30.2% saline versus 10.6% isoproterenol, P<0.05). However, the ß1AR association with SERCA2a was enhanced (puncta/cell, 51.4 saline versus 87.5 isoproterenol, P<0.05), and the receptor signal was minimally affected. The isoproterenol-infused hearts displayed decreased PDE4D (phosphodiesterase 4D) and PDE3A and increased PDE2A, PDE4A, and PDE4B protein levels. We observed a reduced role of PDE4 and enhanced roles of PDE2 and PDE3 on the ß1AR-PKA activity at the ryanodine receptor 2 complexes and myocyte shortening. Despite the enhanced ß1AR association with SERCA2a, the endogenous norepinephrine-induced signaling was reduced at the SERCA2a complexes. Inhibiting monoamine oxidase A rescued the norepinephrine-induced PKA signaling at the SERCA2a and myocyte shortening. CONCLUSIONS: This study reveals distinct mechanisms for the downregulation of subcellular ß1AR signaling in the heart under chronic adrenergic stimulation.


Sujet(s)
Canaux calciques de type L , Cyclic AMP-Dependent Protein Kinases , Régulation négative , Isoprénaline , Souris de lignée C57BL , Myocytes cardiaques , Récepteurs bêta-1 adrénergiques , Canal de libération du calcium du récepteur à la ryanodine , Sarcoplasmic Reticulum Calcium-Transporting ATPases , Transduction du signal , Animaux , Récepteurs bêta-1 adrénergiques/métabolisme , Mâle , Canal de libération du calcium du récepteur à la ryanodine/métabolisme , Isoprénaline/pharmacologie , Cyclic AMP-Dependent Protein Kinases/métabolisme , Myocytes cardiaques/métabolisme , Myocytes cardiaques/effets des médicaments et des substances chimiques , Sarcoplasmic Reticulum Calcium-Transporting ATPases/métabolisme , Canaux calciques de type L/métabolisme , Canaux calciques de type L/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Souris , Défaillance cardiaque/métabolisme , Défaillance cardiaque/induit chimiquement , Défaillance cardiaque/physiopathologie , Cardiomyopathies/métabolisme , Cardiomyopathies/induit chimiquement , Transfert d'énergie par résonance de fluorescence
3.
Biochim Biophys Acta Mol Basis Dis ; 1870(5): 167208, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38701956

RÉSUMÉ

OBJECTIVE: This study aims to investigate the cardiac protective effects and molecular mechanisms of electroacupuncture (EA) pre-treatment in lipopolysaccharide (LPS)-Induced Cardiomyopathy. METHODS AND RESULTS: Pre-treatment with EA was performed 30 min before intraperitoneal injection of LPS. Cardiac function changes in mice of the EA + LPS group were observed using electrocardiography, echocardiography, and enzyme linked immunosorbent assay (ELISA) and compared with the LPS group. The results demonstrated that EA pre-treatment significantly improved the survival rate of septic mice, alleviated the severity of endotoxemia, and exhibited notable cardiac protective effects. These effects were characterized by a reduction in ST-segment elevation on electrocardiography, an increase in ejection fraction (EF) and fraction shortening (FS) on echocardiography and a decrease in the expression of serum cardiac troponin I (cTn-I) levels. Serum exosomes obtained after EA pre-treatment were extracted and administered to septic mice, revealing significant cardiac protective effects of EA-derived exosomes. Furthermore, the antagonism of circulating exosomes in mice markedly suppressed the cardiac protective effects conferred by EA pre-treatment. Analysis of serum exosomes using quantitative reverse transcription-polymerase chain reaction (qRT-PCR) revealed a significant upregulation of miR-381 expression after EA pre-treatment. Inhibition or overexpression of miR-381 through serotype 9 adeno-associated virus (AAV9)-mediated gene delivery demonstrated that overexpression of miR-381 exerted a cardiac protective effect, while inhibition of miR-381 significantly attenuated the cardiac protective effects conferred by EA pre-treatment. CONCLUSIONS: Our research findings have revealed a novel endogenous cardiac protection mechanism, wherein circulating exosomes derived from EA pre-treatment mitigate LPS-induced cardiac dysfunction via miR-381.


Sujet(s)
Cardiomyopathies , Électroacupuncture , Exosomes , Lipopolysaccharides , microARN , Animaux , microARN/génétique , microARN/métabolisme , Exosomes/métabolisme , Exosomes/génétique , Électroacupuncture/méthodes , Souris , Cardiomyopathies/induit chimiquement , Cardiomyopathies/métabolisme , Cardiomyopathies/thérapie , Cardiomyopathies/anatomopathologie , Cardiomyopathies/génétique , Cardiomyopathies/prévention et contrôle , Lipopolysaccharides/toxicité , Mâle , Souris de lignée C57BL , Modèles animaux de maladie humaine
4.
Clin Cardiol ; 47(5): e24278, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38767024

RÉSUMÉ

Olanzapine, an atypical antipsychotic medication, has gained prominence in the treatment of schizophrenia and related psychotic disorders due to its effectiveness and perceived safety profile. However, emerging evidence suggests a potential link between olanzapine use and adverse cardiovascular effects, including cardiomyopathy. This narrative review explores the mechanisms, clinical implications, and management strategies associated with olanzapine-induced cardiomyopathy. A comprehensive review of the literature was conducted to investigate the relationship between olanzapine and cardiomyopathy. The search included epidemiological studies, clinical case reports, and mechanistic research focusing on the pathophysiology of olanzapine-induced cardiomyopathy. The review also examined treatment strategies for managing this potential complication. Olanzapine-induced cardiomyopathy is hypothesized to be associated with metabolic disturbances and receptor antagonism. The metabolic effects of olanzapine, such as weight gain, insulin resistance, and dyslipidemia, share similarities with obesity-related cardiomyopathy. Additionally, olanzapine's antagonism of certain receptors may contribute to cardiovascular stress. The review highlighted that patients with new-onset heart failure and significant weight gain while on olanzapine should be closely monitored for signs of cardiomyopathy. Early detection and prompt withdrawal of olanzapine, along with initiation of goal-directed medical therapy, are crucial for mitigating this potentially life-threatening condition. The relationship between olanzapine and cardiomyopathy is complex and not yet fully understood. However, the potential for significant cardiovascular risk necessitates vigilance among healthcare providers. Early identification and management of olanzapine-induced cardiomyopathy can improve patient outcomes. Further research is needed to elucidate the precise mechanisms behind this adverse effect and to develop optimized treatment strategies for patients requiring antipsychotic therapy.


Sujet(s)
Neuroleptiques , Cardiomyopathies , Obésité , Olanzapine , Humains , Olanzapine/effets indésirables , Neuroleptiques/effets indésirables , Cardiomyopathies/induit chimiquement , Cardiomyopathies/diagnostic , Obésité/complications , Schizophrénie/traitement médicamenteux , Diagnostic différentiel , Facteurs de risque
5.
Biomed Pharmacother ; 174: 116534, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38565062

RÉSUMÉ

The isoproterenol (ISO)-induced myocardial fibrosis is considered a reliable and repeatable experimental model characterized by a relatively low mortality rate. Although is well-known that ISO stimulates the ß1 adrenergic receptors at the myocardial level, a high degree of heterogeneity emerges around the doses and duration of the treatment generating unclear results. Therefore, we propose to gain insights into the progression of ISO-induced myocardial fibrosis, in order to critically analyze and optimize the experimental model. Male Wistar rats (12-14-week-old) were submitted to subcutaneous injection of ISO, in particular, two doses were selected: the commonly used dose of 5 mg/kg and a lower dose of 1 mg/kg, administered for 3 and 6 days. Biochemical and histological examinations were conducted either immediately after the last administration or after a recovering period of 7 or 14 days from the initial administration. Noteworthy, from our investigation emerged that even the lower dose of ISO was able to induce the maximal biochemical and histological alterations, suggesting that lower doses should be considered to control the progression of the damage more precisely and to identify a prodromic phase in which intervention with pharmacological or nutraceutical tools can be effectively attempted.


Sujet(s)
Fibrose , Isoprénaline , Myocarde , Rat Wistar , Animaux , Mâle , Myocarde/anatomopathologie , Myocarde/métabolisme , Rats , Cardiomyopathies/induit chimiquement , Cardiomyopathies/anatomopathologie , Cardiomyopathies/métabolisme , Cardiomyopathies/prévention et contrôle , Relation dose-effet des médicaments , Modèles animaux de maladie humaine
6.
Arch Toxicol ; 98(6): 1781-1794, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38573338

RÉSUMÉ

Doxorubicin (DOX) is one of the most frequently used chemotherapeutic drugs belonging to the class of anthracyclines. However, the cardiotoxic effects of anthracyclines limit their clinical use. Recent studies have suggested that ferroptosis is the main underlying pathogenetic mechanism of DOX-induced cardiomyopathy (DIC). BTB-and-CNC homology 1 (Bach1) acts as a key role in the regulation of ferroptosis. However, the mechanistic role of Bach1 in DIC remains unclear. Therefore, this study aimed to investigate the underlying mechanistic role of Bach1 in DOX-induced cardiotoxicity using the DIC mice in vivo (DOX at cumulative dose of 20 mg/kg) and the DOX-treated H9c2 cardiomyocytes in vitro (1 µM). Our results show a marked upregulation in the expression of Bach1 in the cardiac tissues of the DOX-treated mice and the DOX-treated cardiomyocytes. However, Bach1-/- mice exhibited reduced lipid peroxidation and less severe cardiomyopathy after DOX treatment. Bach1 knockdown protected against DOX-induced ferroptosis in both in vivo and in vitro models. Ferrostatin-1 (Fer-1), a potent inhibitor of ferroptosis, significantly alleviated DOX-induced cardiac damage. However, the cardioprotective effects of Bach1 knockdown were reversed by pre-treatment with Zinc Protoporphyrin (ZnPP), a selective inhibitor of heme oxygenase-1(HO-1). Taken together, these findings demonstrated that Bach1 promoted oxidative stress and ferroptosis through suppressing the expression of HO-1. Therefore, Bach1 may present as a promising new therapeutic target for the prevention and early intervention of DOX-induced cardiotoxicity.


Sujet(s)
Facteurs de transcription à motif basique et à glissière à leucines , Cardiomyopathies , Doxorubicine , Ferroptose , Heme oxygenase-1 , Souris de lignée C57BL , Souris knockout , Myocytes cardiaques , Stress oxydatif , Animaux , Ferroptose/effets des médicaments et des substances chimiques , Doxorubicine/toxicité , Stress oxydatif/effets des médicaments et des substances chimiques , Cardiomyopathies/induit chimiquement , Cardiomyopathies/métabolisme , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myocytes cardiaques/métabolisme , Myocytes cardiaques/anatomopathologie , Facteurs de transcription à motif basique et à glissière à leucines/métabolisme , Facteurs de transcription à motif basique et à glissière à leucines/génétique , Mâle , Souris , Heme oxygenase-1/métabolisme , Heme oxygenase-1/génétique , Lignée cellulaire , Rats , Cardiotoxicité , Antibiotiques antinéoplasiques/toxicité , Peroxydation lipidique/effets des médicaments et des substances chimiques , Protoporphyrines/pharmacologie , Transduction du signal/effets des médicaments et des substances chimiques , Cyclohexylamines , Protéines membranaires , Phénylènediamines
7.
J Nanobiotechnology ; 22(1): 195, 2024 Apr 20.
Article de Anglais | MEDLINE | ID: mdl-38643173

RÉSUMÉ

Doxorubicin (DOX) is a chemotherapeutic agent widely used for tumor treatment. Nonetheless its clinical application is heavily limited by its cardiotoxicity. There is accumulated evidence that transplantation of mesenchymal stem cell-derived exosomes (MSC-EXOs) can protect against Dox-induced cardiomyopathy (DIC). This study aimed to examine the cardioprotective effects of EXOs isolated from human induced pluripotent stem cell-derived MSCs (iPSC-MSCs) against DIC and explore the potential mechanisms. EXOs were isolated from the cultural supernatant of human BM-MSCs (BM-MSC-EXOs) and iPSC-MSCs (iPSC-MSC-EXOs) by ultracentrifugation. A mouse model of DIC was induced by intraperitoneal injection of Dox followed by tail vein injection of PBS, BM-MSC-EXOs, or iPSC-MSC-EXOs. Cardiac function, cardiomyocyte senescence and mitochondrial dynamics in each group were assessed. In vitro, neonatal mouse cardiomyocytes (NMCMs) were subjected to Dox and treated with BM-MSC-EXOs or iPSC-MSC-EXOs. The mitochondrial morphology and cellular senescence of NMCMs were examined by Mitotracker staining and senescence-associated-ß-galactosidase assay, respectively. Compared with BM-MSC-EXOs, mice treated with iPSC-MSC-EXOs displayed improved cardiac function and decreased cardiomyocyte mitochondrial fragmentation and senescence. In vitro, iPSC-MSC-EXOs were superior to BM-MSC-EXOs in attenuation of cardiomyocyte mitochondrial fragmentation and senescence caused by DOX. MicroRNA sequencing revealed a higher level of miR-9-5p in iPSC-MSC-EXOs than BM-MSC-EXOs. Mechanistically, iPSC-MSC-EXOs transported miR-9-5p into DOX-treated cardiomyocytes, thereby suppressing cardiomyocyte mitochondrial fragmentation and senescence via regulation of the VPO1/ERK signal pathway. These protective effects and cardioprotection against DIC were largely reversed by knockdown of miR-9-5p in iPSC-MSC-EXOs. Our results showed that miR-9-5p transferred by iPSC-MSC-EXOs protected against DIC by alleviating cardiomyocyte senescence via inhibition of the VPO1/ERK pathway. This study offers new insight into the application of iPSC-MSC-EXOs as a novel therapeutic strategy for DIC treatment.


Sujet(s)
Cardiomyopathies , Cellules souches pluripotentes induites , microARN , Humains , Souris , Animaux , Myocytes cardiaques/métabolisme , Cellules souches pluripotentes induites/métabolisme , microARN/génétique , microARN/métabolisme , Cardiomyopathies/induit chimiquement , Transduction du signal , Doxorubicine
8.
Sci Rep ; 14(1): 6971, 2024 03 23.
Article de Anglais | MEDLINE | ID: mdl-38521855

RÉSUMÉ

Doxorubicin has been used extensively as a potent anticancer agent, but its clinical use is limited by its cardiotoxicity. However, the underlying mechanisms remain to be fully elucidated. In this study, we tested whether NADPH oxidase 2 (Nox2) mediates cardiac sympathetic nerve terminal abnormalities and myocyte autophagy, resulting in cardiac atrophy and dysfunction in doxorubicin-induced heart failure. Nox2 knockout (KO) and wild-type (WT) mice were randomly assigned to receive a single injection of doxorubicin (15 mg/kg, i.p.) or saline. WT doxorubicin mice exhibited the decreases in survival rate, left ventricular (LV) wall thickness and LV fractional shortening and the increase in the lung wet-to-dry weight ratio 1 week after the injections. These alterations were attenuated in Nox2 KO doxorubicin mice. In WT doxorubicin mice, myocardial oxidative stress was increased, myocardial noradrenergic nerve fibers were reduced, myocardial expression of PGP9.5, GAP43, tyrosine hydroxylase and norepinephrine transporter was decreased, and these changes were prevented in Nox2 KO doxorubicin mice. Myocyte autophagy was increased and myocyte size was decreased in WT doxorubicin mice, but not in Nox2 KO doxorubicin mice. Nox2 mediates cardiac sympathetic nerve terminal abnormalities and myocyte autophagy-both of which contribute to cardiac atrophy and failure after doxorubicin treatment.


Sujet(s)
Cardiomyopathies , Myocytes cardiaques , NADPH Oxidase 2 , Animaux , Souris , Autophagie , Cardiomyopathies/induit chimiquement , Cardiomyopathies/métabolisme , Doxorubicine/pharmacologie , Souris de lignée C57BL , Souris knockout , Myocytes cardiaques/métabolisme , NADPH Oxidase 2/génétique , NADPH Oxidase 2/métabolisme , Stress oxydatif , Sympathectomie
9.
Int Immunopharmacol ; 131: 111897, 2024 Apr 20.
Article de Anglais | MEDLINE | ID: mdl-38513575

RÉSUMÉ

BACKGROUND: Esculin, a main active ingredient from Cortex fraxini, possesses biological activities such as anti-thrombosis, anti-inflammatory, and anti-oxidation effects. However, the effects of Esculin on septic cardiomyopathy remains unclear. This study aimed to explore the protective properties and mechanisms of Esculin in countering sepsis-induced cardiac trauma and dysfunction. METHODS AND RESULTS: In lipopolysaccharide (LPS)-induced mice model, Esculin could obviously improve heart injury and function. Esculin treatment also significantly reduced the production of inflammatory and apoptotic cells, the release of inflammatory cytokines, and the expression of oxidative stress-associated and apoptosis-associated markers in hearts compared to LPS injection alone. These results were consistent with those of in vitro experiments based on neonatal rat cardiomyocytes. Database analysis and molecular docking suggested that TLR4 was targeted by Esculin, as shown by stable hydrogen bonds formed between Esculin with VAL-308, ASN-307, CYS-280, CYS-304 and ASP-281 of TLR4. Esculin reversed LPS-induced upregulation of TLR4 and phosphorylation of NF-κB p65 in cardiomyocytes. The plasmid overexpressing TLR4 abolished the protective properties of Esculin in vitro. CONCLUSION: We concluded that Esculin could alleviate LPS-induced septic cardiomyopathy via binding to TLR4 to attenuate cardiomyocyte inflammation, oxidative stress and apoptosis.


Sujet(s)
Cardiomyopathies , Lipopolysaccharides , Souris , Rats , Animaux , Lipopolysaccharides/pharmacologie , Esculoside/pharmacologie , Récepteur de type Toll-4/métabolisme , Simulation de docking moléculaire , Anti-inflammatoires/pharmacologie , Cardiomyopathies/induit chimiquement , Cardiomyopathies/traitement médicamenteux , Facteur de transcription NF-kappa B/métabolisme
10.
Eur Rev Med Pharmacol Sci ; 28(5): 1641-1650, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38497849

RÉSUMÉ

OBJECTIVE: The pathogenesis of doxorubicin (DOX) induced cardiomyopathy (DCM) is still uncertain. We aimed to identify the critical genes and pathways involved in DCM based on bioinformatics analysis. MATERIALS AND METHODS: The GSE59672 and GSE23598 mice heart tissue microarray data were obtained from Gene Expression Omnibus (GEO) database. The "limma" package of R software was used to screen the differently expressed genes (DEGs). GO (Gene Ontology) and KEGG (Kyoto Encyclopedia of Genes and Genomes) analyses were performed on DEGs by using "clusterProfiler" package in R software. The PPI (Protein - Protein Interaction) network of DEGs constructed by STRING online database and thereby the top 15 hub genes selected by cytoHubba in Cytoscape software. The hub genes interaction was performed by GeneMANIA online database. The "Corrplot" R package was employed to assess hub genes correlation. RESULTS: Finally, a total of 492 and 501 DEGs were screened in GSE59672 and GSE23598 datasets, respectively. GO analyses revealed that DEGs were mainly involved in the regulation of extracellular matrix organization, metabolic process, regulation of collagen-containing extracellular matrix. KEGG pathway analyses indicated that DEGs were mainly involved in protein digestion and absorption, ECM-receptor interaction, phagosome, and p53 signaling pathway. Finally, the 8 hub genes were identified, including Col1a1, Col3a1, Col1a2, Col6a1, Ptprc, Tyrobp, Itgb2, and Ctss. CONCLUSIONS: The present study identified a series of key genes, including Col1a1, Col3a1, Col1a2, Col6a1, Ptprc, Tyrobp, Itgb2, and Ctss. In addition, important pathways were also discovered. The results of this study may provide a novel molecular mechanism and potential therapeutic targets for DCM.


Sujet(s)
Cardiomyopathies , Animaux , Souris , Cardiomyopathies/induit chimiquement , Cardiomyopathies/génétique , Antigènes CD18 , Biologie informatique , Bases de données factuelles , Doxorubicine/effets indésirables
11.
J Colloid Interface Sci ; 663: 1064-1073, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38458046

RÉSUMÉ

Doxorubicin (DOX) is widely used in clinic as a broad-spectrum chemotherapy drug, which can enhance the efficacy of chemodynamic therapy (CDT) by interfering tumor-related metabolize to increase H2O2 content. However, DOX can induce serious cardiomyopathy (DIC) due to its oxidative stress in cardiomyocytes. Eliminating oxidative stress would create a significant opportunity for the clinical application of DOX combined with CDT. To address this issue, we introduced sodium ascorbate (AscNa), the main reason is that AscNa can be catalyzed to produce H2O2 by the abundant Fe3+ in the tumor site, thereby enhancing CDT. While the content of Fe3+ in heart tissue is relatively low, so the oxidation of AscNa had tumor specificity. Meanwhile, due to its inherent reducing properties, AscNa could also eliminate the oxidative stress generated by DOX, preventing cardiotoxicity. Due to the differences between myocardial tissue and tumor microenvironment, a novel nanomedicine was designed. MoS2 was employed as a carrier and CDT catalyst, loaded with DOX and AscNa, coating with homologous tumor cell membrane to construct an acid-responsive nanomedicine MoS2-DOX/AscNa@M (MDA@M). In tumor cells, AscNa enhances the synergistic therapy of DOX and MoS2. In cardiomyocytes, AscNa could effectively reduce the cardiomyopathy induced by DOX. Overall, this study enhanced the clinical potential of chemotherapy synergistic CDT.


Sujet(s)
Cardiomyopathies , Tumeurs , Humains , Cardiotoxicité/traitement médicamenteux , Cardiotoxicité/étiologie , Cardiotoxicité/prévention et contrôle , Nanomédecine , Peroxyde d'hydrogène/métabolisme , Molybdène/métabolisme , Doxorubicine/pharmacologie , Myocytes cardiaques/métabolisme , Myocytes cardiaques/anatomopathologie , Cardiomyopathies/induit chimiquement , Cardiomyopathies/traitement médicamenteux , Cardiomyopathies/anatomopathologie , Acide ascorbique/pharmacologie , Lignée cellulaire tumorale , Tumeurs/métabolisme , Microenvironnement tumoral
12.
Sci Rep ; 14(1): 7227, 2024 03 27.
Article de Anglais | MEDLINE | ID: mdl-38538694

RÉSUMÉ

There is a scarcity of information on the population with diabetes mellitus type 2 and cardiomyopathy (PDMC) in COVID-19, especially on the association between anti-diabetic medications and COVID-19 outcomes. Study is designed as a retrospective cohort analysis covering 2020 and 2021. Data from National Diabetes Registry (CroDiab) were linked to hospital data, primary healthcare data, the SARS-CoV-2 vaccination database, and the SARS-CoV-2 test results database. Study outcomes were cumulative incidence of SARS-CoV-2 positivity, COVID-19 hospitalizations, and COVID-19 deaths. For outcome predictors, logistic regression models were developed. Of 231 796 patients with diabetes mellitus type 2 in the database, 14 485 patients had cardiomyopathy. The two2-year cumulative incidence of all three studies' COVID-19 outcomes was higher in PDMC than in the general diabetes population (positivity 15.3% vs. 14.6%, p = 0.01; hospitalization 7.8% vs. 4.4%, p < 0.001; death 2.6% vs. 1.2%, p < 0.001). Sodium-Glucose Transporter 2 (SGLT-2) inhibitors therapy was found to be protective of SARS-CoV-2 infections [OR 0.722 (95% CI 0.610-0.856)] and COVID-19 hospitalizations [OR 0.555 (95% CI 0.418-0.737)], sulfonylureas to be risk factors for hospitalization [OR 1.184 (95% CI 1.029-1.362)] and insulin to be a risk factor for hospitalization [OR 1.261 (95% CI 1.046-1.520)] and death [OR 1.431 (95% CI 1.080-1.897)]. PDMC are at greater risk of acquiring SARS-CoV-2 infection and having worse outcomes than the general diabetic population. SGLT-2 inhibitors therapy was a protective factor against SARS-CoV-2 infection and against COVID-19 hospitalization, sulfonylurea was the COVID-19 hospitalization risk factor, while insulin was a risk factor for all outcomes. Further research is needed in this diabetes sub-population.


Sujet(s)
COVID-19 , Cardiomyopathies , Diabète de type 2 , Inhibiteurs de la dipeptidyl-peptidase IV , Inhibiteurs du cotransporteur sodium-glucose de type 2 , Humains , Hypoglycémiants/usage thérapeutique , Études rétrospectives , Vaccins contre la COVID-19/usage thérapeutique , Inhibiteurs de la dipeptidyl-peptidase IV/usage thérapeutique , COVID-19/complications , SARS-CoV-2 , Diabète de type 2/complications , Diabète de type 2/traitement médicamenteux , Diabète de type 2/épidémiologie , Sulfonylurées/usage thérapeutique , Inhibiteurs du cotransporteur sodium-glucose de type 2/usage thérapeutique , Insuline/usage thérapeutique , Cardiomyopathies/induit chimiquement
13.
Rom J Intern Med ; 62(2): 210-215, 2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38377065

RÉSUMÉ

Hydroxychloroquine (HCQ) induced cardiotoxicity is a rare diagnosis and is often associated with chronic use of the medication. It has been shown that chronic HCQ use is associated with a drug-induced cardiomyopathy mainly driven by acquired lysosomal storage defects leading to hypertrophy and conduction abnormalities. As the only proven treatment is the discontinuation of the offending agent, prompt recognition is required to avoid further exposure to the drug and potential progression of disease. History, physical examination and advanced imaging modalities are useful diagnostic tools, but more invasive testing with an endomyocardial biopsy is required for definitive diagnosis. We present a descriptive case series of ten patients that were diagnosed with biopsy proven HCQ cardiotoxicity.


Sujet(s)
Antirhumatismaux , Cardiotoxicité , Hydroxychloroquine , Humains , Hydroxychloroquine/effets indésirables , Adulte d'âge moyen , Femelle , Mâle , Cardiotoxicité/étiologie , Antirhumatismaux/effets indésirables , Sujet âgé , Adulte , Biopsie , Cardiomyopathies/induit chimiquement
14.
Toxicology ; 503: 153752, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38369011

RÉSUMÉ

The study sought to assess the detrimental effects of isoproterenol (ISO) on major organs and investigate the potential reversibility of these adverse reactions in mice. Male mice were divided into normal control, 0.2 mg/kg.d and 3.0 mg/kg.d ISO groups, and were subcutaneously administered of the respective doses for 14 consecutive days. Subsequently, a recovery period experiment was conducted, replicating the aforementioned procedure, followed by an additional 2-week recovery period for the mice. Following 14 consecutive days of administration, mice treated with ISO exhibited notable cardiac damage manifested by abnormal ECG patterns, dysregulated energy metabolism, elevated cardiac hypertrophy, and increased heart pathological score. Additionally, the administration of ISO resulted in liver and kidney damage, as evidenced by increased pathological score, serum albumin level, and urea level. Lung damage was also observed, indicated by an increase in lung pathological score. Furthermore, the administration of ISO at a dosage of 3.0 mg/kg.d resulted in a decrease in liver mass index, serum iron content, and an increase in lung mass index. After a 2-week recovery period, mice treated with ISO showed abnormalities in ECG patterns and dysregulated myocardial energy metabolism, accompanied by a decrease in serum iron content. Histopathological examinations revealed continued pathological changes in the heart and lung, as well as significant hemosiderin deposition in the spleen. Furthermore, the group treated with ISO at a dosage of 3.0 mg/kg.d showed an increase in serum AST and TP levels. In summary, the study demonstrates that both 0.2 mg/kg.d and 3.0 mg/kg.d doses of ISO can induce damage to the heart, liver, lung, kidney, and spleen, with the higher dose causing more severe injuries. After a 2-week withdrawal period, the liver, kidney, and thymus injuries caused by 0.2 mg/kg ISO shows signs of recovery, while damage to the heart, lung, and spleen persists. The thymus injury mostly recovers, with minimal kidney pathology, but significant damage to the heart, liver, and lung remains even after the withdrawal period for the 3.0 mg/kg ISO dose.


Sujet(s)
Cardiomyopathies , Myocarde , Rats , Mâle , Souris , Animaux , Isoprénaline/toxicité , Isoprénaline/métabolisme , Rat Wistar , Myocarde/métabolisme , Cardiomyopathies/induit chimiquement , Métabolisme énergétique , Fer/métabolisme
16.
Circ Res ; 134(5): 505-525, 2024 03.
Article de Anglais | MEDLINE | ID: mdl-38422177

RÉSUMÉ

BACKGROUND: Chronic overconsumption of lipids followed by their excessive accumulation in the heart leads to cardiomyopathy. The cause of lipid-induced cardiomyopathy involves a pivotal role for the proton-pump vacuolar-type H+-ATPase (v-ATPase), which acidifies endosomes, and for lipid-transporter CD36, which is stored in acidified endosomes. During lipid overexposure, an increased influx of lipids into cardiomyocytes is sensed by v-ATPase, which then disassembles, causing endosomal de-acidification and expulsion of stored CD36 from the endosomes toward the sarcolemma. Once at the sarcolemma, CD36 not only increases lipid uptake but also interacts with inflammatory receptor TLR4 (Toll-like receptor 4), together resulting in lipid-induced insulin resistance, inflammation, fibrosis, and cardiac dysfunction. Strategies inducing v-ATPase reassembly, that is, to achieve CD36 reinternalization, may correct these maladaptive alterations. For this, we used NAD+ (nicotinamide adenine dinucleotide)-precursor nicotinamide mononucleotide (NMN), inducing v-ATPase reassembly by stimulating glycolytic enzymes to bind to v-ATPase. METHODS: Rats/mice on cardiomyopathy-inducing high-fat diets were supplemented with NMN and for comparison with a cocktail of lysine/leucine/arginine (mTORC1 [mechanistic target of rapamycin complex 1]-mediated v-ATPase reassembly). We used the following methods: RNA sequencing, mRNA/protein expression analysis, immunofluorescence microscopy, (co)immunoprecipitation/proximity ligation assay (v-ATPase assembly), myocellular uptake of [3H]chloroquine (endosomal pH), and [14C]palmitate, targeted lipidomics, and echocardiography. To confirm the involvement of v-ATPase in the beneficial effects of both supplementations, mTORC1/v-ATPase inhibitors (rapamycin/bafilomycin A1) were administered. Additionally, 2 heart-specific v-ATPase-knockout mouse models (subunits V1G1/V0d2) were subjected to these measurements. Mechanisms were confirmed in pharmacologically/genetically manipulated cardiomyocyte models of lipid overload. RESULTS: NMN successfully preserved endosomal acidification during myocardial lipid overload by maintaining v-ATPase activity and subsequently prevented CD36-mediated lipid accumulation, CD36-TLR4 interaction toward inflammation, fibrosis, cardiac dysfunction, and whole-body insulin resistance. Lipidomics revealed C18:1-enriched diacylglycerols as lipid class prominently increased by high-fat diet and subsequently reversed/preserved by lysine/leucine/arginine/NMN treatment. Studies with mTORC1/v-ATPase inhibitors and heart-specific v-ATPase-knockout mice further confirmed the pivotal roles of v-ATPase in these beneficial actions. CONCLUSION: NMN preserves heart function during lipid overload by preventing v-ATPase disassembly.


Sujet(s)
Cardiomyopathies , Insulinorésistance , Animaux , Souris , Rats , Adenosine triphosphatases , Arginine , Cardiomyopathies/induit chimiquement , Cardiomyopathies/prévention et contrôle , Antigènes CD36/génétique , Fibrose , Inflammation , Leucine , Lipides , Lysine , Complexe-1 cible mécanistique de la rapamycine , Myocytes cardiaques , Nicotinamide mononucléotide , Récepteur de type Toll-4/génétique
17.
Cell Signal ; 117: 111070, 2024 05.
Article de Anglais | MEDLINE | ID: mdl-38307305

RÉSUMÉ

Doxorubicin (Dox) is a potent antineoplastic agent, but its use is curtailed by severe cardiotoxicity, known as Dox-induced cardiomyopathy (DIC). The molecular mechanism underlying this cardiotoxicity remains unclear. Our current study investigates the role of Ubiquitin-Specific Protease 36 (USP36), a nucleolar deubiquitinating enzyme (DUB), in the progression of DIC and its mechanism. We found increased USP36 expression in neonatal rat cardiomyocytes and H9C2 cells exposed to Dox. Silencing USP36 significantly mitigated Dox-induced oxidative stress injury and apoptosis in vitro. Mechanistically, USP36 upregulation positively correlated with Poly (ADP-ribose) polymerase 1 (PARP1) expression, and its knockdown led to a reduction in PARP1 levels. Further investigation revealed that USP36 could bind to and mediate the deubiquitination of PARP1, thereby increasing its protein stability in cardiomyocytes upon Dox exposure. Moreover, overexpression of wild-type (WT) USP36 plasmid, but not its catalytically inactive mutant (C131A), stabilized PARP1 in HEK293T cells. We also established a DIC model in mice and observed significant upregulation of USP36 in the heart. Cardiac knockdown of USP36 in mice using a type 9 recombinant adeno-associated virus (rAAV9)-shUSP36 significantly preserved cardiac function after Dox treatment and protected against Dox-induced structural changes within the myocardium. In conclusion, these findings suggest that Dox promotes DIC progression by activating USP36-mediated PARP1 deubiquitination. This novel USP36/PARP1 axis may play a significant regulatory role in the pathogenesis of DIC.


Sujet(s)
Cardiomyopathies , Cardiotoxicité , Animaux , Humains , Souris , Rats , Apoptose , Cardiomyopathies/induit chimiquement , Cardiomyopathies/complications , Cardiotoxicité/métabolisme , Doxorubicine/effets indésirables , Doxorubicine/toxicité , Cellules HEK293 , Myocytes cardiaques/métabolisme , Stress oxydatif , Poly (ADP-Ribose) polymerase-1/métabolisme , Ubiquitin thiolesterase/métabolisme
19.
J Clin Oncol ; 42(11): 1265-1277, 2024 Apr 10.
Article de Anglais | MEDLINE | ID: mdl-38207238

RÉSUMÉ

PURPOSE: To leverage baseline global longitudinal strain (GLS) and N-terminal-pro-B-type natriuretic peptide (NT-proBNP) to identify childhood cancer survivors with a normal left ventricular ejection fraction (LVEF) at highest risk of future treatment-related cardiomyopathy. METHODS: St Jude Lifetime Cohort participants ≥5 years from diagnosis, at increased risk for cardiomyopathy per the International Guideline Harmonization Group (IGHG), with an LVEF ≥50% on baseline echocardiography (n = 1,483) underwent measurement of GLS (n = 1,483) and NT-proBNP (n = 1,052; 71%). Multivariable Cox regression models estimated hazard ratios (HRs) and 95% CIs for postbaseline cardiomyopathy (modified Common Terminology Criteria for Adverse Events ≥grade 2) incidence in association with echocardiogram-based GLS (≥-18) and/or NT-proBNP (>age-sex-specific 97.5th percentiles). Prediction performance was assessed using AUC in models with and without GLS and NT-proBNP and compared using DeLong's test for IGHG moderate- and high-risk individuals treated with anthracyclines. RESULTS: Among survivors (median age, 37.6; range, 10.2-70.4 years), 162 (11.1%) developed ≥grade 2 cardiomyopathy 5.1 (0.7-10.0) years from baseline assessment. The 5-year cumulative incidence of cardiomyopathy for survivors with and without abnormal GLS was, respectively, 7.3% (95% CI, 4.7 to 9.9) versus 4.4% (95% CI, 3.0 to 5.7) and abnormal NT-proBNP was 9.9% (95% CI, 5.8 to 14.1) versus 4.7% (95% CI, 3.2 to 6.2). Among survivors with a normal LVEF, abnormal baseline GLS and NT-proBNP identified anthracycline-exposed, IGHG-defined moderate-/high-risk survivors at a four-fold increased hazard of postbaseline cardiomyopathy (HR, 4.39 [95% CI, 2.46 to 7.83]; P < .001), increasing to a HR of 14.16 (95% CI, 6.45 to 31.08; P < .001) among survivors who received ≥250 mg/m2 of anthracyclines. Six years after baseline, AUCs for individual risk prediction were 0.70 for models with and 0.63 for models without GLS and NT-proBNP (P = .022). CONCLUSION: GLS and NT-proBNP should be considered for improved identification of survivors at high risk for future cardiomyopathy.


Sujet(s)
Survivants du cancer , Cardiomyopathies , Tumeurs , Mâle , Femelle , Humains , Enfant , Adulte , Peptide natriurétique cérébral , Débit systolique , Strain global longitudinal , Fonction ventriculaire gauche , Marqueurs biologiques , Tumeurs/traitement médicamenteux , Cardiomyopathies/induit chimiquement , Cardiomyopathies/diagnostic , Cardiomyopathies/épidémiologie , Fragments peptidiques , Cardiotoxicité/étiologie , Anthracyclines/effets indésirables , Antibiotiques antinéoplasiques/effets indésirables
20.
Apoptosis ; 29(5-6): 816-834, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38281279

RÉSUMÉ

Doxorubicin (DOX) is an anthracycline antibiotic used as an antitumor treatment. However, its clinical application is limited due to severe side effects such as cardiotoxicity. In recent years, numerous studies have demonstrated that cellular aging has become a therapeutic target for DOX-induced cardiomyopathy. However, the underlying mechanism and specific molecular targets of DOX-induced cardiomyocyte aging remain unclear. Poly (ADP-ribose) polymerase (PARP) is a family of protein post-translational modification enzymes in eukaryotic cells, including 18 members. PARP-1, the most well-studied member of this family, has become a potential molecular target for the prevention and treatment of various cardiovascular diseases, such as DOX cardiomyopathy and heart failure. PARP-1 and PARP-2 share 69% homology in the catalytic regions. However, they do not entirely overlap in function. The role of PARP-2 in cardiovascular diseases, especially in DOX-induced cardiomyocyte aging, is less studied. In this study, we found for the first time that down-regulation of PARP-2 can inhibit DOX-induced cellular aging in cardiomyocytes. On the contrary, overexpression of PARP-2 can aggravate DOX-induced cardiomyocyte aging and injury. Further research showed that PARP-2 inhibited the expression and activity of SIRT1, which in turn was involved in the development of DOX-induced cardiomyocyte aging and injury. Our findings provide a preliminary experimental basis for establishing PARP-2 as a new target for preventing and treating DOX cardiomyopathy and related drug development.


Sujet(s)
Vieillissement de la cellule , Doxorubicine , Myocytes cardiaques , Poly(ADP-ribose) polymerases , Sirtuine-1 , Doxorubicine/effets indésirables , Doxorubicine/pharmacologie , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myocytes cardiaques/anatomopathologie , Myocytes cardiaques/métabolisme , Sirtuine-1/métabolisme , Sirtuine-1/génétique , Animaux , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Poly(ADP-ribose) polymerases/métabolisme , Poly(ADP-ribose) polymerases/génétique , Rats , Cardiotoxicité/anatomopathologie , Cardiotoxicité/métabolisme , Cardiotoxicité/prévention et contrôle , Cardiotoxicité/étiologie , Apoptose/effets des médicaments et des substances chimiques , Rat Sprague-Dawley , Antibiotiques antinéoplasiques/toxicité , Antibiotiques antinéoplasiques/effets indésirables , Antibiotiques antinéoplasiques/pharmacologie , Cardiomyopathies/induit chimiquement , Cardiomyopathies/anatomopathologie , Cardiomyopathies/métabolisme , Cardiomyopathies/génétique , Humains
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...