Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 3.126
Filtrer
1.
Int J Mol Sci ; 25(16)2024 Aug 12.
Article de Anglais | MEDLINE | ID: mdl-39201467

RÉSUMÉ

The clinical significance of B-type natriuretic peptide (BNP) and N-terminal pro-B-type natriuretic peptide (NT-proBNP) in pediatric patients remains an area of evolving understanding, particularly regarding their utility in the presence or absence of pre-existing heart conditions. While clear cutoff values and established roles in heart failure are understood in adult patients, pediatric norms vary with age, complicating interpretation. Notably, the emergence of multi-system inflammatory syndrome in children (MIS-C) has highlighted the importance of these markers not only in the detection of acute heart failure but also as a marker of disease severity and even as a differential diagnosis tool. This review summarizes current knowledge on the utility of BNP and NT-proBNP in pediatric patients. Their unique physiology, including circulation and compensation mechanisms, likely influence BNP and NT-proBNP release, potentially even in non-heart failure states. Factors such as dynamic volemic changes accompanying inflammatory diseases in children may contribute. Thus, understanding the nuanced roles of BNP and NT-proBNP in pediatric populations is crucial for the accurate diagnosis, management, and differentiation of cardiac and non-cardiac conditions.


Sujet(s)
Marqueurs biologiques , Défaillance cardiaque , Peptide natriurétique cérébral , Fragments peptidiques , Humains , Peptide natriurétique cérébral/sang , Peptide natriurétique cérébral/métabolisme , Enfant , Fragments peptidiques/sang , Marqueurs biologiques/sang , Défaillance cardiaque/sang , Défaillance cardiaque/métabolisme , Défaillance cardiaque/diagnostic , Cardiopathies/sang , Cardiopathies/métabolisme , Cardiopathies/diagnostic , Syndrome de réponse inflammatoire généralisée/sang , Syndrome de réponse inflammatoire généralisée/diagnostic , Pertinence clinique
2.
Vascul Pharmacol ; 155: 107324, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38985581

RÉSUMÉ

Doxorubicin (DOX) is a highly effective chemotherapeutic agent whose clinical use is hindered by the onset of cardiotoxic effects, resulting in reduced ejection fraction within the first year from treatment initiation. Recently it has been demonstrated that DOX accumulates within mitochondria, leading to disruption of metabolic processes and energetic imbalance. We previously described that phosphoinositide 3-kinase γ (PI3Kγ) contributes to DOX-induced cardiotoxicity, causing autophagy inhibition and accumulation of damaged mitochondria. Here we intend to describe the maladaptive metabolic rewiring occurring in DOX-treated hearts and the contribution of PI3Kγ signalling to this process. Metabolomic analysis of DOX-treated WT hearts revealed an accumulation of TCA cycle metabolites due to a cycle slowdown, with reduced levels of pyruvate, unchanged abundance of lactate and increased Acetyl-CoA production. Moreover, the activity of glycolytic enzymes was upregulated, and fatty acid oxidation downregulated, after DOX, indicative of increased glucose oxidation. In agreement, oxygen consumption was increased in after pyruvate supplementation, with the formation of cytotoxic ROS rather than energy production. These metabolic changes were fully prevented in KD hearts. Interestingly, they failed to increase glucose oxidation in response to DOX even with autophagy inhibition, indicating that PI3Kγ likely controls the fuel preference after DOX through an autophagy-independent mechanism. In vitro experiments showed that inhibition of PI3Kγ inhibits pyruvate dehydrogenase (PDH), the key enzyme of Randle cycle regulating the switch from fatty acids to glucose usage, while decreasing DOX-induced mobilization of GLUT-4-carrying vesicles to the plasma membrane and limiting the ensuing glucose uptake. These results demonstrate that PI3Kγ promotes a maladaptive metabolic rewiring in DOX-treated hearts, through a two-pronged mechanism controlling PDH activation and GLUT-4-mediated glucose uptake.


Sujet(s)
Cardiotoxicité , Doxorubicine , Métabolisme énergétique , Acides gras , Glucose , Oxydoréduction , Animaux , Doxorubicine/toxicité , Glucose/métabolisme , Acides gras/métabolisme , Métabolisme énergétique/effets des médicaments et des substances chimiques , Phosphatidylinositol 3-kinases de classe Ib/métabolisme , Glycolyse/effets des médicaments et des substances chimiques , Autophagie/effets des médicaments et des substances chimiques , Mâle , Transduction du signal/effets des médicaments et des substances chimiques , Myocytes cardiaques/métabolisme , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myocytes cardiaques/anatomopathologie , Cycle citrique/effets des médicaments et des substances chimiques , Souris de lignée C57BL , Cardiopathies/induit chimiquement , Cardiopathies/métabolisme , Cardiopathies/anatomopathologie , Cardiopathies/prévention et contrôle , Cardiopathies/physiopathologie , Mitochondries du myocarde/métabolisme , Mitochondries du myocarde/effets des médicaments et des substances chimiques , Mitochondries du myocarde/anatomopathologie , Mitochondries du myocarde/enzymologie , Souris knockout , Modèles animaux de maladie humaine , Espèces réactives de l'oxygène/métabolisme , Transporteur de glucose de type 4/métabolisme , Antibiotiques antinéoplasiques/toxicité , Antibiotiques antinéoplasiques/effets indésirables
4.
Stem Cell Res Ther ; 15(1): 219, 2024 Jul 19.
Article de Anglais | MEDLINE | ID: mdl-39026298

RÉSUMÉ

Epicardial epithelial-to-mesenchymal transition (EMT) plays a pivotal role in both heart development and injury response and involves dynamic cellular changes that are essential for cardiogenesis and myocardial repair. Specifically, epicardial EMT is a crucial process in which epicardial cells lose polarity, migrate into the myocardium, and differentiate into various cardiac cell types during development and repair. Importantly, following EMT, the epicardium becomes a source of paracrine factors that support cardiac growth at the last stages of cardiogenesis and contribute to cardiac remodeling after injury. As such, EMT seems to represent a fundamental step in cardiac repair. Nevertheless, endogenous EMT alone is insufficient to stimulate adequate repair. Redirecting and amplifying epicardial EMT pathways offers promising avenues for the development of innovative therapeutic strategies and treatment approaches for heart disease. In this review, we present a synthesis of recent literature highlighting the significance of epicardial EMT reactivation in adult heart disease patients.


Sujet(s)
Transition épithélio-mésenchymateuse , Péricarde , Humains , Péricarde/métabolisme , Péricarde/cytologie , Animaux , Cardiopathies/métabolisme , Cardiopathies/anatomopathologie , Cardiopathies/thérapie , Myocarde/métabolisme , Myocarde/anatomopathologie , Différenciation cellulaire
5.
J Clin Invest ; 134(13)2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38949027

RÉSUMÉ

Biological sex is an important modifier of physiology and influences pathobiology in many diseases. While heart disease is the number one cause of death worldwide in both men and women, sex differences exist at the organ and cellular scales, affecting clinical presentation, diagnosis, and treatment. In this Review, we highlight baseline sex differences in cardiac structure, function, and cellular signaling and discuss the contribution of sex hormones and chromosomes to these characteristics. The heart is a remarkably plastic organ and rapidly responds to physiological and pathological cues by modifying form and function. The nature and extent of cardiac remodeling in response to these stimuli are often dependent on biological sex. We discuss organ- and molecular-level sex differences in adaptive physiological remodeling and pathological cardiac remodeling from pressure and volume overload, ischemia, and genetic heart disease. Finally, we offer a perspective on key future directions for research into cardiac sex differences.


Sujet(s)
Caractères sexuels , Remodelage ventriculaire , Humains , Femelle , Mâle , Animaux , Cardiopathies/anatomopathologie , Cardiopathies/métabolisme , Cardiopathies/physiopathologie , Cardiopathies/génétique , Hormones sexuelles stéroïdiennes/métabolisme , Coeur/physiopathologie , Coeur/physiologie , Myocarde/anatomopathologie , Myocarde/métabolisme
6.
Int J Mol Sci ; 25(14)2024 Jul 12.
Article de Anglais | MEDLINE | ID: mdl-39062885

RÉSUMÉ

BACKGROUND: Mitochondria play a crucial role in adapting to fluctuating energy demands, particularly in various heart diseases. This study investigates mitochondrial morphology near intercalated discs in left ventricular (LV) heart tissues, comparing samples from patients with sinus rhythm (SR), atrial fibrillation (AF), dilated cardiomyopathy (DCM), and ischemic cardiomyopathy (ICM). METHODS: Transmission electron microscopy was used to analyze mitochondria within 0-3.5 µm and 3.5-7 µm of intercalated discs in 9 SR, 10 AF, 9 DCM, and 8 ICM patient samples. Parameters included mean size in µm2 and elongation, count, percental mitochondrial area in the measuring frame, and a conglomeration score. RESULTS: AF patients exhibited higher counts of small mitochondria in the LV myocardium, resembling SR. DCM and ICM groups had fewer, larger, and often hydropic mitochondria. Accumulation rates and percental mitochondrial area were similar across groups. Significant positive correlations existed between other defects/size and hydropic mitochondria and between count/area and conglomeration score, while negative correlations between count and size/other defects and between hydropic mitochondria and count could be seen as well. CONCLUSION: Mitochondrial parameters in the LV myocardium of AF patients were similar to those of SR patients, while DCM and ICM displayed distinct changes, including a decrease in number, an increase in size, and compromised mitochondrial morphology. Further research is needed to fully elucidate the pathophysiological role of mitochondrial morphology in different heart diseases, providing deeper insights into potential therapeutic targets and interventions.


Sujet(s)
Mitochondries du myocarde , Humains , Mâle , Femelle , Projets pilotes , Adulte d'âge moyen , Sujet âgé , Mitochondries du myocarde/métabolisme , Mitochondries du myocarde/ultrastructure , Cardiomyopathie dilatée/anatomopathologie , Cardiomyopathie dilatée/métabolisme , Cardiopathies/métabolisme , Cardiopathies/anatomopathologie , Microscopie électronique à transmission , Adulte , Ventricules cardiaques/anatomopathologie , Ventricules cardiaques/métabolisme , Ventricules cardiaques/ultrastructure , Fibrillation auriculaire/métabolisme , Fibrillation auriculaire/anatomopathologie , Fibrillation auriculaire/physiopathologie , Myocarde/métabolisme , Myocarde/anatomopathologie , Myocarde/ultrastructure
7.
Biomolecules ; 14(7)2024 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-39062467

RÉSUMÉ

Hyperuricemia (HUA) may lead to myocardial cell damage, thereby promoting the occurrence and adverse outcomes of heart diseases. In this review, we discuss the latest clinical research progress, and explore the impact of HUA on myocardial damage-related diseases such as myocardial infarction, arrhythmias, and heart failure. We also combined recent findings from basic research to analyze potential mechanisms linking HUA with myocardial injury. In different pathological models (such as direct action of high uric acid on myocardial cells or combined with myocardial ischemia-reperfusion model), HUA may cause damage by activating the NOD-like receptor protein 3 inflammasome-induced inflammatory response, interfering with cardiac cell energy metabolism, affecting antioxidant defense systems, and stimulating reactive oxygen species production to enhance the oxidative stress response, ultimately resulting in decreased cardiac function. Additionally, we discuss the impact of lowering uric acid intervention therapy and potential safety issues that may arise. However, as the mechanism underlying HUA-induced myocardial injury is poorly defined, further research is warranted to aid in the development novel therapeutic strategies for HUA-related cardiovascular diseases.


Sujet(s)
Cardiopathies , Hyperuricémie , Stress oxydatif , Acide urique , Humains , Hyperuricémie/métabolisme , Hyperuricémie/traitement médicamenteux , Hyperuricémie/complications , Animaux , Cardiopathies/métabolisme , Acide urique/métabolisme , Espèces réactives de l'oxygène/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Inflammasomes/métabolisme
8.
Biomolecules ; 14(7)2024 Jul 19.
Article de Anglais | MEDLINE | ID: mdl-39062578

RÉSUMÉ

Calcium regulation is essential in virtually any cell due to its critical role as a second messenger in multiple signaling pathways [...].


Sujet(s)
Calcium , Découverte de médicament , Cardiopathies , Humains , Calcium/métabolisme , Découverte de médicament/méthodes , Cardiopathies/métabolisme , Cardiopathies/traitement médicamenteux , Cinétique , Animaux , Signalisation calcique
10.
Circ Res ; 135(5): 596-613, 2024 Aug 16.
Article de Anglais | MEDLINE | ID: mdl-39056179

RÉSUMÉ

BACKGROUND: Macrophages are key players in obesity-associated cardiovascular diseases, which are marked by inflammatory and immune alterations. However, the pathophysiological mechanisms underlying macrophage's role in obesity-induced cardiac inflammation are incompletely understood. Our study aimed to identify the key macrophage population involved in obesity-induced cardiac dysfunction and investigate the molecular mechanism that contributes to the inflammatory response. METHODS: In this study, we used single-cell RNA-sequencing analysis of Cd45+CD11b+F4/80+ cardiac macrophages to explore the heterogeneity of cardiac macrophages. The CCR2+ (C-C chemokine receptor 2) macrophages were specifically removed by a dual recombinase approach, and the macrophage CCR2 was deleted to investigate their functions. We also performed cleavage under target and tagmentation analysis, chromatin immunoprecipitation-polymerase chain reaction, luciferase assay, and macrophage-specific lentivirus transfection to define the impact of lysozyme C in macrophages on obesity-induced inflammation. RESULTS: We find that the Ccr2 cluster undergoes a functional transition from homeostatic maintenance to proinflammation. Our data highlight specific changes in macrophage behavior during cardiac dysfunction under metabolic challenge. Consistently, inducible ablation of CCR2+CX3CR1+ macrophages or selective deletion of macrophage CCR2 prevents obesity-induced cardiac dysfunction. At the mechanistic level, we demonstrate that the obesity-induced functional shift of CCR2-expressing macrophages is mediated by the CCR2/activating transcription factor 3/lysozyme 1/NF-κB (nuclear factor kappa B) signaling. Finally, we uncover a noncanonical role for lysozyme 1 as a transcription activator, binding to the RelA promoter, driving NF-κB signaling, and strongly promoting inflammation and cardiac dysfunction in obesity. CONCLUSIONS: Our findings suggest that lysozyme 1 may represent a potential target for the diagnosis of obesity-induced inflammation and the treatment of obesity-induced heart disease.


Sujet(s)
Macrophages , Lysozyme , Obésité , Récepteurs CCR2 , Animaux , Obésité/complications , Obésité/métabolisme , Macrophages/métabolisme , Récepteurs CCR2/métabolisme , Récepteurs CCR2/génétique , Souris , Lysozyme/métabolisme , Lysozyme/génétique , Souris de lignée C57BL , Mâle , Souris knockout , Transduction du signal , Inflammation/métabolisme , Inflammation/génétique , Cardiopathies/étiologie , Cardiopathies/métabolisme , Cardiopathies/génétique
11.
J Biochem Mol Toxicol ; 38(8): e23783, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39056209

RÉSUMÉ

Cyclophosphamide (CYP) is widely used to treat various types of cancer. In addition to the therapeutic properties of this drug, unfortunately, its side effects are still not fully understood. This study investigated the protective effect of curcumin (CURC) and berberine (BER) on CYP-induced cardiac damage. Thirty-six male rats were equally divided into the control, dimethyl sulfoxide (DMSO), CYP, CYP + CURC, CYP + BER and CYP + BER + CURC groups. Troponin-I, Creatine kinase-myocardial band (CK-MB), total cholesterol, triglyceride levels in serum samples, and reactive oxygen species (ROS), poly(ADP-ribose) polymerase-1 (PARP-1), and transient receptor potential melastatin 2 (TRPM2) channel levels in heart tissue were measured using an enzyme-linked immunoassay (ELISA) kit. In addition, histopathological examination and immunohistochemical investigation of the TRPM2 channel, fibroblast specific protein-1 (FSP1), transforming growth factor-beta- 1 (TGF-ß1) and α-smooth muscle actin (α-SMA) expressions were determined in heart tissue. The CYP group's troponin-I, total cholesterol, triglyceride, CK-MB, ROS, PARP-1 and TRPM2 channel levels were higher than in the other groups in the ELISA measurements (p < 0.05). In contrast, these parameters in the group treated with CURC and BER together with CYP were lower than in the CYP group (p < 0.05). Additionally, CUR and BER reduced CYP-induced pathological damage, TRPM2, FSP1, TGF-ß1 and α-SMA expressions. The data showed that CYP administration can cause cardiac damage by increasing the TRPM2 channel, TGF-ß1, FSP1 and α-SMA expression levels. Therefore, we concluded that CURC and BER administration following CYP application may be used as therapeutic agents to prevent CYP-induced cardiac damage.


Sujet(s)
Berbérine , Curcumine , Cyclophosphamide , Fibrose , Myocarde , Canaux cationiques TRPM , Animaux , Canaux cationiques TRPM/métabolisme , Cyclophosphamide/toxicité , Cyclophosphamide/effets indésirables , Mâle , Rats , Curcumine/pharmacologie , Berbérine/pharmacologie , Myocarde/métabolisme , Myocarde/anatomopathologie , Marqueurs biologiques/métabolisme , Marqueurs biologiques/sang , Lipides/sang , Rat Wistar , Cardiopathies/induit chimiquement , Cardiopathies/métabolisme , Cardiopathies/anatomopathologie , Cardiopathies/prévention et contrôle , Cardiopathies/traitement médicamenteux
12.
Front Biosci (Landmark Ed) ; 29(6): 234, 2024 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-38940046

RÉSUMÉ

Drosophila melanogaster has been used as a model system to identify and characterize genetic contributions to development, homeostasis, and to investigate the molecular determinants of numerous human diseases. While there exist many differences at the genetic, structural, and molecular level, many signalling components and cellular machineries are conserved between Drosophila and humans. For this reason, Drosophila can and has been used extensively to model, and study human pathologies. The extensive genetic resources available make this model system a powerful one. Over the years, the sophisticated and rapidly expanding Drosophila genetic toolkit has provided valuable novel insights into the contribution of genetic components to human diseases. The activity of Notch signalling is crucial during development and conserved across the Metazoa and has been associated with many human diseases. Here we highlight examples of mechanisms involving Notch signalling that have been elucidated from modelling human diseases in Drosophila melanogaster that include neurodegenerative diseases, congenital diseases, several cancers, and cardiac disorders.


Sujet(s)
Drosophila melanogaster , Récepteurs Notch , Transduction du signal , Animaux , Drosophila melanogaster/génétique , Drosophila melanogaster/métabolisme , Récepteurs Notch/métabolisme , Récepteurs Notch/génétique , Transduction du signal/génétique , Humains , Modèles animaux de maladie humaine , Tumeurs/génétique , Tumeurs/métabolisme , Maladies neurodégénératives/génétique , Maladies neurodégénératives/métabolisme , Cardiopathies/génétique , Cardiopathies/métabolisme
13.
Cells ; 13(11)2024 May 28.
Article de Anglais | MEDLINE | ID: mdl-38891063

RÉSUMÉ

During mammalian heart development, the clustered genes encoding peptide hormones, Natriuretic Peptide A (NPPA; ANP) and B (NPPB; BNP), are transcriptionally co-regulated and co-expressed predominately in the atrial and ventricular trabecular cardiomyocytes. After birth, expression of NPPA and a natural antisense transcript NPPA-AS1 becomes restricted to the atrial cardiomyocytes. Both NPPA and NPPB are induced by cardiac stress and serve as markers for cardiovascular dysfunction or injury. NPPB gene products are extensively used as diagnostic and prognostic biomarkers for various cardiovascular disorders. Membrane-localized guanylyl cyclase receptors on many cell types throughout the body mediate the signaling of the natriuretic peptide ligands through the generation of intracellular cGMP, which interacts with and modulates the activity of cGMP-activated kinase and other enzymes and ion channels. The natriuretic peptide system plays a fundamental role in cardio-renal homeostasis, and its potent diuretic and vasodilatory effects provide compensatory mechanisms in cardiac pathophysiological conditions and heart failure. In addition, both peptides, but also CNP, have important intracardiac actions during heart development and homeostasis independent of the systemic functions. Exploration of the intracardiac functions may provide new leads for the therapeutic utility of natriuretic peptide-mediated signaling in heart diseases and rhythm disorders. Here, we review recent insights into the regulation of expression and intracardiac functions of NPPA and NPPB during heart development, homeostasis, and disease.


Sujet(s)
Coeur , Homéostasie , Peptides natriurétiques , Humains , Animaux , Peptides natriurétiques/métabolisme , Cardiopathies/métabolisme , Cardiopathies/génétique , Cardiopathies/anatomopathologie
14.
Int J Mol Sci ; 25(11)2024 May 25.
Article de Anglais | MEDLINE | ID: mdl-38891948

RÉSUMÉ

Cardiovascular diseases (CVDs), particularly heart failure, are major contributors to early mortality globally. Heart failure poses a significant public health problem, with persistently poor long-term outcomes and an overall unsatisfactory prognosis for patients. Conventionally, treatments for heart failure have focused on lowering blood pressure; however, the development of more potent therapies targeting hemodynamic parameters presents challenges, including tolerability and safety risks, which could potentially restrict their clinical effectiveness. Adenosine has emerged as a key mediator in CVDs, acting as a retaliatory metabolite produced during cellular stress via ATP metabolism, and works as a signaling molecule regulating various physiological processes. Adenosine functions by interacting with different adenosine receptor (AR) subtypes expressed in cardiac cells, including A1AR, A2AAR, A2BAR, and A3AR. In addition to A1AR, A3AR has a multifaceted role in the cardiovascular system, since its activation contributes to reducing the damage to the heart in various pathological states, particularly ischemic heart disease, heart failure, and hypertension, although its role is not as well documented compared to other AR subtypes. Research on A3AR signaling has focused on identifying the intricate molecular mechanisms involved in CVDs through various pathways, including Gi or Gq protein-dependent signaling, ATP-sensitive potassium channels, MAPKs, and G protein-independent signaling. Several A3AR-specific agonists, such as piclidenoson and namodenoson, exert cardioprotective impacts during ischemia in the diverse animal models of heart disease. Thus, modulating A3ARs serves as a potential therapeutic approach, fueling considerable interest in developing compounds that target A3ARs as potential treatments for heart diseases.


Sujet(s)
Cardiopathies , Récepteur A3 à l'adénosine , Transduction du signal , Humains , Animaux , Transduction du signal/effets des médicaments et des substances chimiques , Récepteur A3 à l'adénosine/métabolisme , Cardiopathies/métabolisme , Cardiopathies/traitement médicamenteux , Agonistes du récepteur A3 à l'adénosine/usage thérapeutique , Agonistes du récepteur A3 à l'adénosine/pharmacologie , Adénosine/métabolisme
16.
Cardiovasc Toxicol ; 24(8): 818-835, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38896162

RÉSUMÉ

Doxorubicin (DOX) is an effective anticancer agent, yet its clinical utility is hampered by dose-dependent cardiotoxicity. This study explores the cardioprotective potential of Marein (Mar) against DOX-induced cardiac injury and elucidates underlying molecular mechanisms. Neonatal rat cardiomyocytes (NRCMs) and murine models were employed to assess the impact of Mar on DOX-induced cardiotoxicity (DIC). In vitro, cell viability, oxidative stress were evaluated. In vivo, a chronic injection method was employed to induce a DIC mouse model, followed by eight weeks of Mar treatment. Cardiac function, histopathology, and markers of cardiotoxicity were assessed. In vitro, Mar treatment demonstrated significant cardioprotective effects in vivo, as evidenced by improved cardiac function and reduced indicators of cardiac damage. Mechanistically, Mar reduced inflammation, oxidative stress, and apoptosis in cardiomyocytes, potentially via activation of the Focal Adhesion Kinase (FAK)/AKT pathway. Mar also exhibited an anti-ferroptosis effect. Interestingly, Mar did not compromise DOX's efficacy in cancer cells, suggesting a dual benefit in onco-cardiology. Molecular docking studies suggested a potential interaction between Mar and FAK. This study demonstrates Mar's potential as a mitigator of DOX-induced cardiotoxicity, offering a translational perspective on its clinical application. By activating the FAK/AKT pathway, Mar exerts protective effects against DOX-induced cardiomyocyte damage, highlighting its promise in onco-cardiology. Further research is warranted to validate these findings and advance Mar as a potential adjunctive therapy in cancer treatment.


Sujet(s)
Apoptose , Cardiotoxicité , Modèles animaux de maladie humaine , Doxorubicine , Focal adhesion kinase 1 , Cardiopathies , Myocytes cardiaques , Stress oxydatif , Protéines proto-oncogènes c-akt , Transduction du signal , Animaux , Doxorubicine/toxicité , Protéines proto-oncogènes c-akt/métabolisme , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myocytes cardiaques/anatomopathologie , Myocytes cardiaques/enzymologie , Myocytes cardiaques/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Focal adhesion kinase 1/métabolisme , Stress oxydatif/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques , Humains , Cardiopathies/induit chimiquement , Cardiopathies/métabolisme , Cardiopathies/prévention et contrôle , Cardiopathies/enzymologie , Cardiopathies/anatomopathologie , Mâle , Anthraquinones/pharmacologie , Souris de lignée C57BL , Rat Sprague-Dawley , Rats , Lignée cellulaire tumorale , Cytoprotection , Cellules cultivées , Antibiotiques antinéoplasiques/toxicité , Souris
17.
Adv Exp Med Biol ; 1441: 341-364, 2024.
Article de Anglais | MEDLINE | ID: mdl-38884720

RÉSUMÉ

Epigenetics is the study of heritable changes to the genome and gene expression patterns that are not caused by direct changes to the DNA sequence. Examples of these changes include posttranslational modifications to DNA-bound histone proteins, DNA methylation, and remodeling of nuclear architecture. Collectively, epigenetic changes provide a layer of regulation that affects transcriptional activity of genes while leaving DNA sequences unaltered. Sequence variants or mutations affecting enzymes responsible for modifying or sensing epigenetic marks have been identified in patients with congenital heart disease (CHD), and small-molecule inhibitors of epigenetic complexes have shown promise as therapies for adult heart diseases. Additionally, transgenic mice harboring mutations or deletions of genes encoding epigenetic enzymes recapitulate aspects of human cardiac disease. Taken together, these findings suggest that the evolving field of epigenetics will inform our understanding of congenital and adult cardiac disease and offer new therapeutic opportunities.


Sujet(s)
Méthylation de l'ADN , Épigenèse génétique , Humains , Animaux , Méthylation de l'ADN/génétique , Cardiopathies congénitales/génétique , Histone/métabolisme , Histone/génétique , Maturation post-traductionnelle des protéines , Souris , Cardiopathies/génétique , Cardiopathies/métabolisme , Mutation
18.
Free Radic Biol Med ; 221: 13-22, 2024 Aug 20.
Article de Anglais | MEDLINE | ID: mdl-38697490

RÉSUMÉ

Ischemic heart diseases and cardiomyopathies are characterized by hypoxia, energy starvation and mitochondrial dysfunction. HIF-1 acts as a cellular oxygen sensor, tuning the balance of metabolic and oxidative stress pathways to provide ATP and sustain cell survival. Acting on mitochondria, HIF-1 regulates different processes such as energy substrate utilization, oxidative phosphorylation and mitochondrial dynamics. In turn, mitochondrial homeostasis modifications impact HIF-1 activity. This underlies that HIF-1 and mitochondria are tightly interconnected to maintain cell homeostasis. Despite many evidences linking HIF-1 and mitochondria, the mechanistic insights are far from being understood, particularly in the context of cardiac diseases. Here, we explore the current understanding of how HIF-1, reactive oxygen species and cell metabolism are interconnected, with a specific focus on mitochondrial function and dynamics. We also discuss the divergent roles of HIF in acute and chronic cardiac diseases in order to highlight that HIF-1, mitochondria and oxidative stress interaction deserves to be deeply investigated. While the strategies aiming at stabilizing HIF-1 have provided beneficial effects in acute ischemic injury, some deleterious effects were observed during prolonged HIF-1 activation. Thus, deciphering the link between HIF-1 and mitochondria will help to optimize HIF-1 modulation and provide new therapeutic perspectives for the treatment of cardiovascular pathologies.


Sujet(s)
Cardiopathies , Stress oxydatif , Espèces réactives de l'oxygène , Humains , Cardiopathies/métabolisme , Cardiopathies/anatomopathologie , Cardiopathies/génétique , Animaux , Espèces réactives de l'oxygène/métabolisme , Facteur-1 induit par l'hypoxie/métabolisme , Facteur-1 induit par l'hypoxie/génétique , Mitochondries du myocarde/métabolisme , Mitochondries du myocarde/anatomopathologie , Mitochondries/métabolisme , Mitochondries/anatomopathologie , Mitochondries/génétique , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Sous-unité alpha du facteur-1 induit par l'hypoxie/génétique , Phosphorylation oxydative , Ischémie myocardique/métabolisme , Ischémie myocardique/anatomopathologie , Ischémie myocardique/génétique
19.
J Am Heart Assoc ; 13(9): e032067, 2024 May 07.
Article de Anglais | MEDLINE | ID: mdl-38700010

RÉSUMÉ

BACKGROUND: Doxorubicin and other anthracyclines are crucial cancer treatment drugs. However, they are associated with significant cardiotoxicity, severely affecting patient care and limiting dosage and usage. Previous studies have shown that low carbon monoxide (CO) concentrations protect against doxorubicin toxicity. However, traditional methods of CO delivery pose complex challenges for daily administration, such as dosing and toxicity. To address these challenges, we developed a novel oral liquid drug product containing CO (HBI-002) that can be easily self-administered by patients with cancer undergoing doxorubicin treatment, resulting in CO being delivered through the upper gastrointestinal tract. METHODS AND RESULTS: HBI-002 was tested in a murine model of doxorubicin cardiotoxicity in the presence and absence of lung or breast cancer. The mice received HBI-002 twice daily before doxorubicin administration and experienced increased carboxyhemoglobin levels from a baseline of ≈1% to 7%. Heart tissue from mice treated with HBI-002 had a 6.3-fold increase in CO concentrations and higher expression of the cytoprotective enzyme heme oxygenase-1 compared with placebo control. In both acute and chronic doxorubicin toxicity scenarios, HBI-002 protected the heart from cardiotoxic effects, including limiting tissue damage and cardiac dysfunction and improving survival. In addition, HBI-002 did not compromise the efficacy of doxorubicin in reducing tumor volume, but rather enhanced the sensitivity of breast 4T1 cancer cells to doxorubicin while simultaneously protecting cardiac function. CONCLUSIONS: These findings strongly support using HBI-002 as a cardioprotective agent that maintains the therapeutic benefits of doxorubicin cancer treatment while mitigating cardiac damage.


Sujet(s)
Antibiotiques antinéoplasiques , Monoxyde de carbone , Cardiotoxicité , Doxorubicine , Protéines membranaires , Animaux , Doxorubicine/toxicité , Monoxyde de carbone/métabolisme , Antibiotiques antinéoplasiques/toxicité , Femelle , Administration par voie orale , Souris , Heme oxygenase-1/métabolisme , Cardiopathies/induit chimiquement , Cardiopathies/prévention et contrôle , Cardiopathies/métabolisme , Cardiopathies/anatomopathologie , Modèles animaux de maladie humaine , Souris de lignée C57BL , Carboxyhémoglobine/métabolisme , Fonction ventriculaire gauche/effets des médicaments et des substances chimiques , Humains
20.
Physiology (Bethesda) ; 39(5): 0, 2024 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-38713090

RÉSUMÉ

Oxidative phosphorylation is regulated by mitochondrial calcium (Ca2+) in health and disease. In physiological states, Ca2+ enters via the mitochondrial Ca2+ uniporter and rapidly enhances NADH and ATP production. However, maintaining Ca2+ homeostasis is critical: insufficient Ca2+ impairs stress adaptation, and Ca2+ overload can trigger cell death. In this review, we delve into recent insights further defining the relationship between mitochondrial Ca2+ dynamics and oxidative phosphorylation. Our focus is on how such regulation affects cardiac function in health and disease, including heart failure, ischemia-reperfusion, arrhythmias, catecholaminergic polymorphic ventricular tachycardia, mitochondrial cardiomyopathies, Barth syndrome, and Friedreich's ataxia. Several themes emerge from recent data. First, mitochondrial Ca2+ regulation is critical for fuel substrate selection, metabolite import, and matching of ATP supply to demand. Second, mitochondrial Ca2+ regulates both the production and response to reactive oxygen species (ROS), and the balance between its pro- and antioxidant effects is key to how it contributes to physiological and pathological states. Third, Ca2+ exerts localized effects on the electron transport chain (ETC), not through traditional allosteric mechanisms but rather indirectly. These effects hinge on specific transporters, such as the uniporter or the Na+/Ca2+ exchanger, and may not be noticeable acutely, contributing differently to phenotypes depending on whether Ca2+ transporters are acutely or chronically modified. Perturbations in these novel relationships during disease states may either serve as compensatory mechanisms or exacerbate impairments in oxidative phosphorylation. Consequently, targeting mitochondrial Ca2+ holds promise as a therapeutic strategy for a variety of cardiac diseases characterized by contractile failure or arrhythmias.


Sujet(s)
Calcium , Mitochondries du myocarde , Humains , Animaux , Calcium/métabolisme , Mitochondries du myocarde/métabolisme , Phosphorylation oxydative , Espèces réactives de l'oxygène/métabolisme , Myocarde/métabolisme , Cardiopathies/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE