Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 9.353
Filtrer
1.
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.
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
5.
Int J Cardiol ; 410: 132232, 2024 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-38844090

RÉSUMÉ

Arterial hypertension (AH) is one of the most common pathologic conditions and uncontrolled AH is a leading risk factor for cardiovascular disease and mortality. AH chronically causes myocardial and arterial remodelling with hemodynamic changes affecting the heart and other organs, with potentially irreversible consequences leading to poor outcomes. Therefore, a proper and early treatment of AH is crucial after the diagnosis. Beyond medical treatment, physical exercise also plays a therapeutic role in reducing blood pressure, given its potential effects on sympathetic tone, renin-angiotensin-aldosterone system, and endothelial function. International scientific societies recommend physical exercise among lifestyle modifications to treat AH in the first stages of the disease. Moreover, some studies have also shown its usefulness in addition to drugs to reduce blood pressure further. Therefore, an accurate, personalized exercise prescription is recommended to optimize the prevention and treatment of hypertension. On the other hand, uncontrolled AH in athletes requires proper risk stratification and careful evaluation to practice competitive sports safely. Moreover, the differential diagnosis between hypertensive heart disease and athlete's heart is sometimes challenging and requires a careful and comprehensive interpretation in order not to misinterpret the clinical findings. The present review aims to discuss the relationship between hypertensive heart disease and physical exercise, from diagnostic tools to prevention and treatment strategies.


Sujet(s)
Exercice physique , Hypertension artérielle , Humains , Exercice physique/physiologie , Hypertension artérielle/diagnostic , Hypertension artérielle/physiopathologie , Hypertension artérielle/thérapie , Diagnostic différentiel , Cardiopathies/diagnostic , Cardiopathies/physiopathologie , Cardiopathies/étiologie , Traitement par les exercices physiques/méthodes
6.
J Am Heart Assoc ; 13(9): e034731, 2024 May 07.
Article de Anglais | MEDLINE | ID: mdl-38700011

RÉSUMÉ

BACKGROUND: Cardiac damage induced by ischemic stroke, such as arrhythmia, cardiac dysfunction, and even cardiac arrest, is referred to as cerebral-cardiac syndrome (CCS). Cardiac macrophages are reported to be closely associated with stroke-induced cardiac damage. However, the role of macrophage subsets in CCS is still unclear due to their heterogeneity. Sympathetic nerves play a significant role in regulating macrophages in cardiovascular disease. However, the role of macrophage subsets and sympathetic nerves in CCS is still unclear. METHODS AND RESULTS: In this study, a middle cerebral artery occlusion mouse model was used to simulate ischemic stroke. ECG and echocardiography were used to assess cardiac function. We used Cx3cr1GFPCcr2RFP mice and NLRP3-deficient mice in combination with Smart-seq2 RNA sequencing to confirm the role of macrophage subsets in CCS. We demonstrated that ischemic stroke-induced cardiac damage is characterized by severe cardiac dysfunction and robust infiltration of monocyte-derived macrophages into the heart. Subsequently, we identified that cardiac monocyte-derived macrophages displayed a proinflammatory profile. We also observed that cardiac dysfunction was rescued in ischemic stroke mice by blocking macrophage infiltration using a CCR2 antagonist and NLRP3-deficient mice. In addition, a cardiac sympathetic nerve retrograde tracer and a sympathectomy method were used to explore the relationship between sympathetic nerves and cardiac macrophages. We found that cardiac sympathetic nerves are significantly activated after ischemic stroke, which contributes to the infiltration of monocyte-derived macrophages and subsequent cardiac dysfunction. CONCLUSIONS: Our findings suggest a potential pathogenesis of CCS involving the cardiac sympathetic nerve-monocyte-derived macrophage axis.


Sujet(s)
Modèles animaux de maladie humaine , Accident vasculaire cérébral ischémique , Macrophages , Souris de lignée C57BL , Protéine-3 de la famille des NLR contenant un domaine pyrine , Animaux , Macrophages/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/génétique , Protéine-3 de la famille des NLR contenant un domaine pyrine/déficit , Accident vasculaire cérébral ischémique/physiopathologie , Accident vasculaire cérébral ischémique/métabolisme , Accident vasculaire cérébral ischémique/anatomopathologie , Récepteurs CCR2/génétique , Récepteurs CCR2/métabolisme , Mâle , Souris knockout , Souris , Infarctus du territoire de l'artère cérébrale moyenne/physiopathologie , Infarctus du territoire de l'artère cérébrale moyenne/anatomopathologie , Système nerveux sympathique/physiopathologie , Myocarde/anatomopathologie , Myocarde/métabolisme , Cardiopathies/étiologie , Cardiopathies/physiopathologie , Cardiopathies/anatomopathologie , Récepteur-1 de la chimiokine CX3C/génétique , Récepteur-1 de la chimiokine CX3C/métabolisme , Récepteur-1 de la chimiokine CX3C/déficit
7.
Heart Lung ; 67: 70-79, 2024.
Article de Anglais | MEDLINE | ID: mdl-38714139

RÉSUMÉ

BACKGROUND: Inflammation is implicated in cardiovascular disease (CVD) pathogenesis, but causal roles of specific circulating inflammatory cytokines remain unclear. Mendelian randomization (MR) studies are well-poised to provide etiological insights beyond constraints of conventional research. METHODS: We conducted a large-scale MR study to investigate potential causal relationships of 91 inflammatory proteins with CVD outcomes and cardiac remodeling using summary-level genetic data. Outcomes included coronary artery disease, myocardial infarction, stroke, atrial fibrillation, heart failure, abdominal aortic aneurysm, deep vein thrombosis of lower extremities, pulmonary embolism, cardiac structure and functional parameters. Inverse-variance weighted analysis was undertaken as the primary analysis, with several sensitivity analyses applied. RESULTS: Hepatocyte growth factor (HGF) demonstrated a causal relationship with increased susceptibility to both any stroke (OR 1.111; 95 % CI 1.044 - 1.183; P = 9.50e-04) and ischemic stroke (OR 1.121; 95 % CI 1.047 - 1.200; P = 1.04e-03). Programmed cell death 1 ligand 1 (PD-L1) was negatively associated with atrial fibrillation risk (OR 0.936, 95 % CI 0.901 - 0.973; P = 7.69e-04). CCL20, CDCP1, Flt3L and IL-10RA were identified as causal coronary artery disease risk factors, while LIF and ST1A1 had protective effects. IL-4 and LIF-R demonstrated causal links with right heart functional changes. CONCLUSIONS: Our MR study nominates specific circulating inflammatory cytokines as potential targets for CVD treatment and prevention. Further research into mechanisms and clinical translation are warranted.


Sujet(s)
Cytokines , Analyse de randomisation mendélienne , Humains , Cytokines/sang , Cytokines/métabolisme , Analyse de randomisation mendélienne/méthodes , Cardiopathies/sang , Cardiopathies/étiologie , Cardiopathies/physiopathologie , Inflammation/sang , Mâle , Femelle
8.
Am J Physiol Heart Circ Physiol ; 327(1): H108-H117, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38758123

RÉSUMÉ

Frailty reflects the heterogeneity in aging and may lead to the development of hypertension and heart disease, but the frailty-cardiovascular relationship and whether physical activity modifies this relationship in males and females are unclear. We tested whether higher frailty was positively associated with hypertension and heart disease in males and females and whether habitual movement mediated this relationship. The relationship between baseline frailty with follow-up hypertension and heart disease was investigated using the Canadian Longitudinal Study on Aging at 3-year follow-up data (males: n = 13,095; females: n = 13,601). Frailty at baseline was determined via a 73-item deficit-based index, activity at follow-up was determined via the Physical Activity Scale for the Elderly, and cardiovascular function was self-reported. Higher baseline frailty level was associated with a greater likelihood of hypertension and heart disease at follow-up, with covariate-adjusted odds ratios of 1.08-1.09 (all, P < 0.001) for a 0.01 increase in frailty index score. Among males and females, sitting time and strenuous physical activity were independently associated with hypertension, with these activity behaviors being partial mediators (except male-sitting time) for the frailty-hypertension relationship (explained 5-10% of relationship). The strength of this relationship was stronger among females. Only light-moderate activity partially mediated the relationship (∼6%) between frailty and heart disease in females, but no activity measure was a mediator for males. Higher frailty levels were associated with a greater incidence of hypertension and heart disease, and strategies that target increases in physical activity and reducing sitting may partially uncouple this relationship with hypertension, particularly among females.NEW & NOTEWORTHY Longitudinally, our study demonstrates that higher baseline frailty levels are associated with an increased risk of hypertension and heart disease in a large sample of Canadian males and females. Movement partially mediated this relationship, particularly among females.


Sujet(s)
Vieillissement , Exercice physique , Fragilité , Hypertension artérielle , Humains , Mâle , Femelle , Hypertension artérielle/physiopathologie , Hypertension artérielle/épidémiologie , Hypertension artérielle/diagnostic , Sujet âgé , Fragilité/physiopathologie , Fragilité/épidémiologie , Fragilité/diagnostic , Canada/épidémiologie , Études longitudinales , Adulte d'âge moyen , Sujet âgé de 80 ans ou plus , Facteurs sexuels , Personne âgée fragile , Pression sanguine , Facteurs âges , Facteurs de risque , Cardiopathies/épidémiologie , Cardiopathies/physiopathologie , Appréciation des risques
9.
Cardiovasc Toxicol ; 24(7): 687-699, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38816669

RÉSUMÉ

The abuse of methamphetamine is a significant threat to cardiovascular health and has detrimental effects on the myocardium. The present study aims to explore potential interventions that can mitigate myocardial pyroptosis in rats following methamphetamine withdrawal. A total of 104 male Wistar rats were randomly assigned to eight groups. The rats underwent a methamphetamine administration protocol, receiving intraperitoneal injections of 10 mg/kg during the 1st week, followed by a weekly dose escalation of 1 mg/kg from the second to the 6th week and two times per day. Concurrently, the rats engaged in 6 weeks of moderate-intensity treadmill aerobic training, lasting 60 min per day, 5 days a week. Simultaneously, the Nutrition bio-shield Superfood (NBS) supplement was administered at a dosage of 25 g/kg daily for 6 weeks. The study assessed the expression levels of Caspase-1, Interleukin-1beta (IL-1ß), and Interleukin-18 (IL-18) genes in myocardial tissue. Data analysis utilized a one-way analysis of variance (p ≤ 0.05). The findings revealed that methamphetamine usage significantly elevated the expression of Caspase-1, IL-1ß, and IL-18 genes (p ≤ 0.05). Conversely, methamphetamine withdrawal led to a notable reduction in the expression of these genes (p ≤ 0.05). Noteworthy reductions in Caspase-1, IL-1ß, and IL-18 expression were observed following aerobic training, supplementation, and the combined approach (p ≤ 0.05). The chronic use of methamphetamine was associated with cardiac tissue damage. This study highlights the potential of aerobic training and NBS Superfood supplementation in mitigating the harmful effects of methamphetamine-induced myocardial pyroptosis. The observed reductions in gene expression levels indicate promising interventions to address the cardiovascular consequences of methamphetamine abuse. The findings of this study suggest that a combination of aerobic exercise and NBS Superfood supplementation can provide a promising approach to mitigate the deleterious effects of methamphetamine on the heart. These findings can be useful for healthcare professionals and policymakers to design effective interventions to prevent and manage the adverse effects of methamphetamine abuse.


Sujet(s)
Cardiotoxicité , Compléments alimentaires , Modèles animaux de maladie humaine , Cardiopathies , Interleukine-18 , Métamfétamine , Conditionnement physique d'animal , Pyroptose , Rat Wistar , Animaux , Métamfétamine/toxicité , Métamfétamine/administration et posologie , Mâle , Conditionnement physique d'animal/physiologie , Conditionnement physique d'animal/méthodes , Pyroptose/effets des médicaments et des substances chimiques , Interleukine-18/métabolisme , Interleukine-18/génétique , Cardiopathies/induit chimiquement , Cardiopathies/prévention et contrôle , Cardiopathies/anatomopathologie , Cardiopathies/physiopathologie , Cardiopathies/métabolisme , Syndrome de sevrage/physiopathologie , Syndrome de sevrage/métabolisme , Syndrome de sevrage/prévention et contrôle , Caspase-1/métabolisme , Caspase-1/génétique , Stimulants du système nerveux central/toxicité , Stimulants du système nerveux central/administration et posologie , Interleukine-1 bêta/métabolisme , Interleukine-1 bêta/génétique , Myocarde/métabolisme , Myocarde/anatomopathologie , Rats , Troubles liés aux amphétamines/physiopathologie , Troubles liés aux amphétamines/métabolisme , Troubles liés aux amphétamines/thérapie , Facteurs temps
10.
Curr Cardiol Rep ; 26(6): 561-580, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38753291

RÉSUMÉ

PURPOSE OF REVIEW: Artificial intelligence (AI) is transforming electrocardiography (ECG) interpretation. AI diagnostics can reach beyond human capabilities, facilitate automated access to nuanced ECG interpretation, and expand the scope of cardiovascular screening in the population. AI can be applied to the standard 12-lead resting ECG and single-lead ECGs in external monitors, implantable devices, and direct-to-consumer smart devices. We summarize the current state of the literature on AI-ECG. RECENT FINDINGS: Rhythm classification was the first application of AI-ECG. Subsequently, AI-ECG models have been developed for screening structural heart disease including hypertrophic cardiomyopathy, cardiac amyloidosis, aortic stenosis, pulmonary hypertension, and left ventricular systolic dysfunction. Further, AI models can predict future events like development of systolic heart failure and atrial fibrillation. AI-ECG exhibits potential in acute cardiac events and non-cardiac applications, including acute pulmonary embolism, electrolyte abnormalities, monitoring drugs therapy, sleep apnea, and predicting all-cause mortality. Many AI models in the domain of cardiac monitors and smart watches have received Food and Drug Administration (FDA) clearance for rhythm classification, while others for identification of cardiac amyloidosis, pulmonary hypertension and left ventricular dysfunction have received breakthrough device designation. As AI-ECG models continue to be developed, in addition to regulatory oversight and monetization challenges, thoughtful clinical implementation to streamline workflows, avoiding information overload and overwhelming of healthcare systems with false positive results is necessary. Research to demonstrate and validate improvement in healthcare efficiency and improved patient outcomes would be required before widespread adoption of any AI-ECG model.


Sujet(s)
Intelligence artificielle , Électrocardiographie , Humains , Électrocardiographie/méthodes , Cardiopathies/diagnostic , Cardiopathies/physiopathologie
11.
Pacing Clin Electrophysiol ; 47(7): 953-965, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38751036

RÉSUMÉ

BACKGROUND: The disease related to the heart is serious and can lead to death. Precise heart disease prediction is imperative for the effective treatment of cardiac patients. This can be attained by machine learning (ML) techniques using healthcare data. Several models on the basis of ML predict and identify disease in the heart, but this model cannot manage a huge database because of the deficiency of the smart model. This paper provides an optimized SpinalNet with a MapReduce model to categorize heart disease. OBJECTIVE: The objective is to design a big data approach for heart disease classification using the proposed Jellyfish Search Flow Regime Optimization (JSFRO)-based SpinalNet. METHOD: The binary image conversion is applied on Electrocardiogram (ECG) images for converting the image to binary image. MapReduce model is adapted, in which the mappers execute feature extraction and the reducer performs heart disease classification. In the mapper phase, the features like statistical features, shape features and temporal features are extracted and in reducer, the SpinalNet with JSFRO is considered. Here, the training of SpinalNet is done with JSFRO, which is produced by the unification of Jellyfish Search Optimization (JSO) and Flow Regime Optimization (FRO). METHOD: The JSFRO-based SpinalNet offered effectual performance with the finest accuracy of 90.8%, sensitivity of 95.2% and specificity of 93.6%.


Sujet(s)
Mégadonnées , Électrocardiographie , Cardiopathies , Apprentissage machine , Humains , Cardiopathies/physiopathologie , Algorithmes
12.
Europace ; 26(5)2024 May 02.
Article de Anglais | MEDLINE | ID: mdl-38691672

RÉSUMÉ

AIMS: Blood stasis is crucial in developing left atrial (LA) thrombi. LA appendage peak flow velocity (LAAFV) is a quantitative parameter for estimating thromboembolic risk. However, its impact on LA thrombus resolution and clinical outcomes remains unclear. METHODS AND RESULTS: The LAT study was a multicentre observational study investigating patients with atrial fibrillation (AF) and silent LA thrombi detected by transoesophageal echocardiography (TEE). Among 17 436 TEE procedures for patients with AF, 297 patients (1.7%) had silent LA thrombi. Excluding patients without follow-up examinations, we enrolled 169 whose baseline LAAFV was available. Oral anticoagulation use increased from 85.7% at baseline to 97.0% at the final follow-up (P < 0.001). During 1 year, LA thrombus resolution was confirmed in 130 (76.9%) patients within 76 (34-138) days. Conversely, 26 had residual LA thrombi, 8 had thromboembolisms, and 5 required surgical removal. These patients with failed thrombus resolution had lower baseline LAAFV than those with successful resolution (18.0 [15.8-22.0] vs. 22.2 [17.0-35.0], P = 0.003). Despite limited predictive power (area under the curve, 0.659; P = 0.001), LAAFV ≤ 20.0 cm/s (best cut-off) significantly predicted failed LA thrombus resolution, even after adjusting for potential confounders (odds ratio, 2.72; 95% confidence interval, 1.22-6.09; P = 0.015). The incidence of adverse outcomes including ischaemic stroke/systemic embolism, major bleeding, or all-cause death was significantly higher in patients with reduced LAAFV than in those with preserved LAAFV (28.4% vs. 11.6%, log-rank P = 0.005). CONCLUSION: Failed LA thrombus resolution was not rare in patients with AF and silent LA thrombi. Reduced LAAFV was associated with failed LA thrombus resolution and adverse clinical outcomes.


Sujet(s)
Anticoagulants , Auricule de l'atrium , Fibrillation auriculaire , Échocardiographie transoesophagienne , Thrombose , Humains , Fibrillation auriculaire/physiopathologie , Fibrillation auriculaire/complications , Mâle , Femelle , Auricule de l'atrium/imagerie diagnostique , Auricule de l'atrium/physiopathologie , Sujet âgé , Thrombose/physiopathologie , Thrombose/imagerie diagnostique , Thrombose/complications , Adulte d'âge moyen , Vitesse du flux sanguin , Anticoagulants/usage thérapeutique , Facteurs de risque , Résultat thérapeutique , Maladies asymptomatiques , Facteurs temps , Cardiopathies/physiopathologie , Cardiopathies/complications , Cardiopathies/imagerie diagnostique , Thromboembolie/étiologie , Thromboembolie/physiopathologie , Sujet âgé de 80 ans ou plus , Fonction auriculaire gauche
13.
Ther Adv Cardiovasc Dis ; 18: 17539447241253134, 2024.
Article de Anglais | MEDLINE | ID: mdl-38819836

RÉSUMÉ

Cardiac fibrosis is a pivotal cardiovascular disease (CVD) process and represents a notable health concern worldwide. While the complex mechanisms underlying CVD have been widely investigated, recent research has highlighted microRNA-21's (miR-21) role in cardiac fibrosis pathogenesis. In this narrative review, we explore the molecular interactions, focusing on the role of miR-21 in contributing to cardiac fibrosis. Various signaling pathways, such as the RAAS, TGF-ß, IL-6, IL-1, ERK, PI3K-Akt, and PTEN pathways, besides dysregulation in fibroblast activity, matrix metalloproteinases (MMPs), and tissue inhibitors of MMPs cause cardiac fibrosis. Besides, miR-21 in growth factor secretion, apoptosis, and endothelial-to-mesenchymal transition play crucial roles. miR-21 capacity regulatory function presents promising insights for cardiac fibrosis. Moreover, this review discusses numerous approaches to control miR-21 expression, including antisense oligonucleotides, anti-miR-21 compounds, and Notch signaling modulation, all novel methods of cardiac fibrosis inhibition. In summary, this narrative review aims to assess the molecular mechanisms of cardiac fibrosis and its essential miR-21 function.


Unraveling cardiac fibrosis: insights into microRNA-21's key role and promising approaches for controlCardiac fibrosis poses a significant global health threat and plays a central role in cardiovascular diseases. This examination delves into recent research revealing the participation of microRNA-21 (MiR-21) in the progression of cardiac fibrosis, providing insight into its critical function in this process. The investigation explores diverse molecular interactions, underscoring MiR-21's contribution to the development of cardiac fibrosis. Various signaling pathways, including the Renin-Angiotensin-Aldosterone System, TGF-ß, IL-6, IL-1, ERK, PI3K-Akt, and PTEN pathways, coupled with disturbances in fibroblast activity, matrix metalloproteinases (MMPs), and tissue inhibitors of MMPs (TIMPs), contribute to cardiac fibrosis. MiR-21's influence on growth factor secretion, apoptosis, and endothelial-to-mesenchymal transition further emphasizes its crucial role. What adds promise to MiR-21 is its capacity for regulation, providing potential insights into controlling cardiac fibrosis. The review also investigates various methods to modulate MiR-21 expression, such as antisense oligonucleotides, anti-miR-21 compounds, and Notch signaling modulation ­ innovative approaches showing potential in inhibiting cardiac fibrosis. In summary, this narrative review aims to dissect the complex molecular mechanisms behind cardiac fibrosis, explicitly emphasizing the indispensable role of MiR-21. By comprehending these mechanisms, researchers can lay the groundwork for inventive interventions and therapeutic strategies to hinder cardiac fibrosis, ultimately contributing to advancing cardiovascular health.


Sujet(s)
Fibrose , microARN , Transduction du signal , microARN/métabolisme , microARN/génétique , Humains , Animaux , Myocarde/anatomopathologie , Myocarde/métabolisme , Cardiopathies/génétique , Cardiopathies/métabolisme , Cardiopathies/anatomopathologie , Cardiopathies/physiopathologie
14.
Cardiovasc Toxicol ; 24(6): 550-562, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38696070

RÉSUMÉ

Trastuzumab is widely used in HER2 breast cancer. However, it may cause left ventricular (LV) dysfunction. A decrease in LV global longitudinal strain (GLS) has been previously demonstrated to be a good predictor of subsequent cancer therapy related dysfunction (CTRCD). Left atrial morphological remodeling during Trastuzumab therapy has also been shown. The aim of this study is exploring the relationship between early changes in left atrial function and the development of Trastuzumab-induced cardiotoxicity. Consecutive patients with diagnosis of HER2+non-metastatic breast cancer treated with Trastuzumab were prospectively enrolled. A clinical, conventional, and advanced echocardiographic assessment was performed at baseline and every three months, until a one-year follow-up was reached. One-hundred-sixteen patients completed the 12 months follow-up, 10 (9%) cases of CTRCD were observed, all after the sixth month. GLS and LVEF significantly decreased in the CTRCD group at 6 months of follow-up, with an earlier (3 months) significant worsening in left atrial morpho-functional parameters. Systolic blood pressure, early peak atrial longitudinal strain (PALS), peak atrial contraction (PACS) and left atrial volume (LAVI) changes resulted independent predictors of CTRCD at multivariable logistic regression analysis. Moreover, early changes in PALS and PACS resulted good predictors of CTRCD development (AUC 0.85; p = 0.008, p < 0.001 and 0.77; p = 0.008, respectively). This prospective study emphasizes that the decline in PALS and PACS among trastuzumab-treated patients could possibly increase the accuracy in identifying future CTRCD in non-metastatic HER2 breast cancer cases, adding predictive value to conventional echocardiographic assessment.


Sujet(s)
Antinéoplasiques immunologiques , Fonction auriculaire gauche , Tumeurs du sein , Cardiotoxicité , Récepteur ErbB-2 , Trastuzumab , Fonction ventriculaire gauche , Humains , Trastuzumab/effets indésirables , Femelle , Tumeurs du sein/traitement médicamenteux , Adulte d'âge moyen , Récepteur ErbB-2/métabolisme , Études prospectives , Antinéoplasiques immunologiques/effets indésirables , Fonction ventriculaire gauche/effets des médicaments et des substances chimiques , Fonction auriculaire gauche/effets des médicaments et des substances chimiques , Adulte , Facteurs temps , Facteurs de risque , Résultat thérapeutique , Sujet âgé , Valeur prédictive des tests , Appréciation des risques , Remodelage auriculaire/effets des médicaments et des substances chimiques , Cardiopathies/induit chimiquement , Cardiopathies/physiopathologie , Cardiopathies/imagerie diagnostique , Dysfonction ventriculaire gauche/induit chimiquement , Dysfonction ventriculaire gauche/physiopathologie , Dysfonction ventriculaire gauche/imagerie diagnostique , Atrium du coeur/effets des médicaments et des substances chimiques , Atrium du coeur/physiopathologie , Atrium du coeur/imagerie diagnostique , Débit systolique/effets des médicaments et des substances chimiques
15.
Cardiovasc Toxicol ; 24(6): 563-575, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38700665

RÉSUMÉ

Anthracycline antibiotic is one of the most effective anti-tumor drugs used to manage certain types of breast cancers, lymphomas, and leukemias. However, anthracyclines induce a dose-dependent cardiotoxicity that may progress to heart failure. Thus, using a sensitive predictor of early cardiac dysfunction in patients treated with anthracyclines can help detect subclinical cardiac dysfunction early and help initiate interventions to protect these patients. Among parameters of myocardial measure, cardiac magnetic resonance (CMR)-measured native myocardial T1 mapping is considered a sensitive and accurate quantitative measure of early subclinical cardiac changes, particularly cardiac inflammation and fibrosis. However, to understand the quality and the validity of the current evidence supporting the use of these measures in patients treated with anthracyclines, we aimed to conduct a systematic review of clinical studies of this measure to detect early myocardial changes in cancer patients treated with anthracyclines. The primary outcome was the level of native T1 mapping. We performed fixed-effects meta-analyses and assessed certainty in effect estimates. Of the 1780 publications reviewed (till 2022), 23 were retrieved, and 9 articles met the inclusion criteria. Our study showed that exposure to anthracycline was associated with a significant elevation of native myocardial T1 mapping from baseline (95% CI 0.1121 to 0.5802; p = 0.0037) as well as compared to healthy control patients (95% CI 0.2925 to 0.7448; p < 0.0001). No significant publication bias was noted on the assessment of the funnel plot and Egger's test. According to the Q test, there was no significant heterogeneity in the included studies (I2 = 0.0000% versus healthy controls and I2 = 14.0666% versus baseline). Overall, our study suggests that native myocardial T1 mapping is useful for detecting anthracycline-induced cardiotoxicity in patients with cancer.


Sujet(s)
Anthracyclines , Antibiotiques antinéoplasiques , Cardiotoxicité , Cardiopathies , Tumeurs , Valeur prédictive des tests , Humains , Anthracyclines/effets indésirables , Tumeurs/traitement médicamenteux , Antibiotiques antinéoplasiques/effets indésirables , Femelle , Cardiopathies/induit chimiquement , Cardiopathies/imagerie diagnostique , Cardiopathies/physiopathologie , Cardiopathies/diagnostic , Mâle , Adulte d'âge moyen , Diagnostic précoce , Facteurs de risque , Adulte , Sujet âgé , Appréciation des risques , Imagerie par résonance magnétique , Fonction ventriculaire gauche/effets des médicaments et des substances chimiques , Jeune adulte
19.
Curr Probl Cardiol ; 49(7): 102587, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38653442

RÉSUMÉ

Inherited metabolic diseases (IMD) are caused by the functional defect of an enzyme, of genetic origin, that provokes a blockage in a specific metabolic pathway. Individually, IMD are considered rare diseases, with an incidence of less than 1/100,000 births. The symptoms are usually multisystemic, but frequently include cardiac manifestations. Of these, the most common are cardiomyopathies, especially hypertrophic cardiomyopathy. In addition, they can cause dilated or restrictive cardiomyopathy and non-compacted cardiomyopathy of the left ventricle. Characteristic signs also include rhythm alterations (atrio-ventricular conduction disturbances, Wolff-Parkinson-White syndrome or ventricular arrhythmias), valvular pathology and ischaemic coronary pathologies. The aim of this study is to present a narrative review of the IMD that may produce cardiac involvement. We describe both the specific cardiac manifestations of each disease and the systemic symptoms that guide diagnosis.


Sujet(s)
Cardiopathies , Humains , Cardiopathies/étiologie , Cardiopathies/diagnostic , Cardiopathies/physiopathologie , Cardiomyopathies/étiologie , Cardiomyopathies/diagnostic , Cardiomyopathies/physiopathologie , Erreurs innées du métabolisme/diagnostic , Erreurs innées du métabolisme/complications
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...