Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 15.757
1.
Open Biol ; 14(6): 230427, 2024 Jun.
Article En | MEDLINE | ID: mdl-38862020

Hypertrophic cardiomyopathy (HCM) is a monogenic cardiac disorder commonly induced by sarcomere gene mutations. However, the mechanism for HCM is not well defined. Here, we generated transgenic MYH7 R453C and MYH6 R453C piglets and found both developed typical cardiac hypertrophy. Unexpectedly, we found serious fibrosis and cardiomyocyte loss in the ventricular of MYH7 R453C, not MYH6 R453C piglets, similar to HCM patients. Then, RNA-seq analysis and western blotting identified the activation of ERK1/2 and PI3K-Akt pathways in MYH7 R453C. Moreover, we observed an increased expression of fetal genes and an excess of reactive oxygen species (ROS) in MYH7 R453C piglet models, which was produced by Nox4 and subsequently induced inflammatory response. Additionally, the phosphorylation levels of Smad2/3, ERK1/2 and NF-kB p65 proteins were elevated in cardiomyocytes with the MYH7 R453C mutation. Furthermore, epigallocatechin gallate, a natural bioactive compound, could be used as a drug to reduce cell death by adjusting significant downregulation of the protein expression of Bax and upregulated Bcl-2 levels in the H9C2 models with MYH7 R453C mutation. In conclusion, our study illustrated that TGF-ß/Smad2/3, ERK1/2 and Nox4/ROS pathways have synergistic effects on cardiac remodelling and inflammation in MYH7 R453C mutation.


Myosin Heavy Chains , NADPH Oxidase 4 , NF-kappa B , Reactive Oxygen Species , Signal Transduction , Transforming Growth Factor beta , Animals , Myosin Heavy Chains/metabolism , Myosin Heavy Chains/genetics , Transforming Growth Factor beta/metabolism , NADPH Oxidase 4/metabolism , NADPH Oxidase 4/genetics , Reactive Oxygen Species/metabolism , NF-kappa B/metabolism , Swine , Myocytes, Cardiac/metabolism , Humans , Cardiac Myosins/metabolism , Cardiac Myosins/genetics , Disease Models, Animal , MAP Kinase Signaling System , Animals, Genetically Modified , Smad2 Protein/metabolism , Smad2 Protein/genetics , Mutation , Smad3 Protein/metabolism , Smad3 Protein/genetics , Ventricular Remodeling , Cardiomyopathy, Hypertrophic/metabolism , Cardiomyopathy, Hypertrophic/genetics , Cardiomyopathy, Hypertrophic/pathology , Rats
2.
Circ Res ; 134(12): 1752-1766, 2024 Jun 07.
Article En | MEDLINE | ID: mdl-38843295

Heart failure (HF) is characterized by a progressive decline in cardiac function and represents one of the largest health burdens worldwide. Clinically, 2 major types of HF are distinguished based on the left ventricular ejection fraction (EF): HF with reduced EF and HF with preserved EF. While both types share several risk factors and features of adverse cardiac remodeling, unique hallmarks beyond ejection fraction that distinguish these etiologies also exist. These differences may explain the fact that approved therapies for HF with reduced EF are largely ineffective in patients suffering from HF with preserved EF. Improving our understanding of the distinct cellular and molecular mechanisms is crucial for the development of better treatment strategies. This article reviews the knowledge of the immunologic mechanisms underlying HF with reduced and preserved EF and discusses how the different immune profiles elicited may identify attractive therapeutic targets for these conditions. We review the literature on the reported mechanisms of adverse cardiac remodeling in HF with reduced and preserved EF, as well as the immune mechanisms involved. We discuss how the knowledge gained from preclinical models of the complex syndrome of HF as well as from clinical data obtained from patients may translate to a better understanding of HF and result in specific treatments for these conditions in humans.


Heart Failure , Stroke Volume , Ventricular Remodeling , Humans , Heart Failure/physiopathology , Heart Failure/immunology , Animals , Myocarditis/physiopathology , Myocarditis/immunology , Ventricular Function, Left , Myocardium/pathology , Myocardium/metabolism , Myocardium/immunology
3.
J Pharmacol Sci ; 155(4): 121-130, 2024 Aug.
Article En | MEDLINE | ID: mdl-38880546

The atrophic myocardium resulting from mechanical unloading and nutritional deprivation is considered crucial as maladaptive remodeling directly associated with heart failure, as well as interstitial fibrosis. Conversely, myocardial hypertrophy resulting from hemodynamic loading is perceived as compensatory stress adaptation. We previously reported the abundant presence of highly redox-active polysulfide molecules, termed supersulfide, with two or more sulfur atoms catenated in normal hearts, and the supersulfide catabolism in pathologic hearts after myocardial infarction correlated with worsened prognosis of heart failure. However, the impact of supersulfide on myocardial remodeling remains unclear. Here, we investigated the involvement of supersulfide metabolism in cardiomyocyte remodeling, using a model of adenosine 5'-triphosphate (ATP) receptor-stimulated atrophy and endothelin-1 receptor-stimulated hypertrophy in neonatal rat cardiomyocytes. Results revealed contrasting changes in intracellular supersulfide and its catabolite, hydrogen sulfide (H2S), between cardiomyocyte atrophy and hypertrophy. Stimulation of cardiomyocytes with ATP decreased supersulfide activity, while H2S accumulation itself did not affect cardiomyocyte atrophy. This supersulfide catabolism was also involved in myofibroblast formation of neonatal rat cardiac fibroblasts. Thus, unraveling supersulfide metabolism during myocardial remodeling may lead to the development of novel therapeutic strategies to improve heart failure.


Hydrogen Sulfide , Myocytes, Cardiac , Sulfides , Ventricular Remodeling , Animals , Myocytes, Cardiac/metabolism , Sulfides/metabolism , Sulfides/pharmacology , Hydrogen Sulfide/metabolism , Cells, Cultured , Adenosine Triphosphate/metabolism , Rats , Atrophy , Cardiomegaly/metabolism , Cardiomegaly/pathology , Heart Failure/metabolism , Heart Failure/pathology , Animals, Newborn , Rats, Sprague-Dawley
4.
Nat Commun ; 15(1): 4757, 2024 Jun 04.
Article En | MEDLINE | ID: mdl-38834564

Semaglutide, a glucagon-like peptide-1 receptor agonist, is clinically used as a glucose-lowering and weight loss medication due to its effects on energy metabolism. In heart failure, energy production is impaired due to altered mitochondrial function and increased glycolysis. However, the impact of semaglutide on cardiomyocyte metabolism under pressure overload remains unclear. Here we demonstrate that semaglutide improves cardiac function and reduces hypertrophy and fibrosis in a mouse model of pressure overload-induced heart failure. Semaglutide preserves mitochondrial structure and function under chronic stress. Metabolomics reveals that semaglutide reduces mitochondrial damage, lipid accumulation, and ATP deficiency by promoting pyruvate entry into the tricarboxylic acid cycle and increasing fatty acid oxidation. Transcriptional analysis shows that semaglutide regulates myocardial energy metabolism through the Creb5/NR4a1 axis in the PI3K/AKT pathway, reducing NR4a1 expression and its translocation to mitochondria. NR4a1 knockdown ameliorates mitochondrial dysfunction and abnormal glucose and lipid metabolism in the heart. These findings suggest that semaglutide may be a therapeutic agent for improving cardiac remodeling by modulating energy metabolism.


Energy Metabolism , Glucagon-Like Peptides , Nuclear Receptor Subfamily 4, Group A, Member 1 , Animals , Male , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Energy Metabolism/drug effects , Mice , Glucagon-Like Peptides/pharmacology , Glucagon-Like Peptides/therapeutic use , Heart Failure/drug therapy , Heart Failure/metabolism , Mice, Inbred C57BL , Ventricular Remodeling/drug effects , Lipid Metabolism/drug effects , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Disease Models, Animal , Myocardium/metabolism , Myocardium/pathology , Signal Transduction/drug effects , Mitochondria/metabolism , Mitochondria/drug effects , Cardiomegaly/drug therapy , Cardiomegaly/metabolism
5.
Int J Mol Sci ; 25(11)2024 Jun 05.
Article En | MEDLINE | ID: mdl-38892396

Cardiac arrhythmias remain a significant concern with Ibrutinib (IBR), a first-generation Bruton's tyrosine kinase inhibitor (BTKi). Acalabrutinib (ABR), a next-generation BTKi, is associated with reduced atrial arrhythmia events. However, the role of ABR in ventricular arrhythmia (VA) has not been adequately evaluated. Our study aimed to investigate VA vulnerability and ventricular electrophysiology following chronic ABR therapy in male Sprague-Dawley rats utilizing epicardial optical mapping for ventricular voltage and Ca2+ dynamics and VA induction by electrical stimulation in ex-vivo perfused hearts. Ventricular tissues were snap-frozen for protein analysis for sarcoplasmic Ca2+ and metabolic regulatory proteins. The results show that both ABR and IBR treatments increased VA vulnerability, with ABR showing higher VA regularity index (RI). IBR, but not ABR, is associated with the abbreviation of action potential duration (APD) and APD alternans. Both IBR and ABR increased diastolic Ca2+ leak and Ca2+ alternans, reduced conduction velocity (CV), and increased CV dispersion. Decreased SERCA2a expression and AMPK phosphorylation were observed with both treatments. Our results suggest that ABR treatment also increases the risk of VA by inducing proarrhythmic changes in Ca2+ signaling and membrane electrophysiology, as seen with IBR. However, the different impacts of these two BTKi on ventricular electrophysiology may contribute to differences in VA vulnerability and distinct VA characteristics.


Agammaglobulinaemia Tyrosine Kinase , Arrhythmias, Cardiac , Benzamides , Piperidines , Rats, Sprague-Dawley , Animals , Benzamides/pharmacology , Benzamides/therapeutic use , Male , Rats , Agammaglobulinaemia Tyrosine Kinase/metabolism , Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/chemically induced , Piperidines/pharmacology , Piperidines/therapeutic use , Action Potentials/drug effects , Ventricular Remodeling/drug effects , Protein Kinase Inhibitors/pharmacology , Pyrazines/pharmacology , Calcium/metabolism , Adenine/analogs & derivatives , Adenine/pharmacology , Adenine/adverse effects , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Heart Ventricles/drug effects , Heart Ventricles/metabolism , Heart Ventricles/physiopathology , Pyrimidines/pharmacology , Calcium Signaling/drug effects , Pyrazoles/pharmacology
6.
Int J Mol Sci ; 25(11)2024 Jun 06.
Article En | MEDLINE | ID: mdl-38892449

Modified mRNAs (modRNAs) are an emerging delivery method for gene therapy. The success of modRNA-based COVID-19 vaccines has demonstrated that modRNA is a safe and effective therapeutic tool. Moreover, modRNA has the potential to treat various human diseases, including cardiac dysfunction. Acute myocardial infarction (MI) is a major cardiac disorder that currently lacks curative treatment options, and MI is commonly accompanied by fibrosis and impaired cardiac function. Our group previously demonstrated that the matricellular protein CCN5 inhibits cardiac fibrosis (CF) and mitigates cardiac dysfunction. However, it remains unclear whether early intervention of CF under stress conditions is beneficial or more detrimental due to potential adverse effects such as left ventricular (LV) rupture. We hypothesized that CCN5 would alleviate the adverse effects of myocardial infarction (MI) through its anti-fibrotic properties under stress conditions. To induce the rapid expression of CCN5, ModRNA-CCN5 was synthesized and administrated directly into the myocardium in a mouse MI model. To evaluate CCN5 activity, we established two independent experimental schemes: (1) preventive intervention and (2) therapeutic intervention. Functional analyses, including echocardiography and magnetic resonance imaging (MRI), along with molecular assays, demonstrated that modRNA-mediated CCN5 gene transfer significantly attenuated cardiac fibrosis and improved cardiac function in both preventive and therapeutic models, without causing left ventricular rupture or any adverse cardiac remodeling. In conclusion, early intervention in CF by ModRNA-CCN5 gene transfer is an efficient and safe therapeutic modality for treating MI-induced heart failure.


CCN Intercellular Signaling Proteins , Fibrosis , Genetic Therapy , Myocardial Infarction , RNA, Messenger , Animals , Mice , Genetic Therapy/methods , Myocardial Infarction/therapy , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , CCN Intercellular Signaling Proteins/genetics , CCN Intercellular Signaling Proteins/metabolism , Myocardium/metabolism , Myocardium/pathology , Disease Models, Animal , Male , Gene Transfer Techniques , Ventricular Remodeling/genetics , Mice, Inbred C57BL , Humans
7.
J Cell Mol Med ; 28(12): e18407, 2024 Jun.
Article En | MEDLINE | ID: mdl-38894630

Chronic intermittent hypoxia (CIH) is associated with an increased risk of cardiovascular diseases. Previously, we have shown that berberine (BBR) is a potential cardioprotective agent. However, its effect and mechanism on CIH-induced cardiomyopathy remain uncovered. This study was designed to determine the effects of BBR against CIH-induced cardiac damage and to explore the molecular mechanisms. Mice were exposed to 5 weeks of CIH with or without the treatment of BBR and adeno-associated virus 9 (AAV9) carrying SIRT6 or SIRT6-specific short hairpin RNA. The effect of BBR was evaluated by echocardiography, histological analysis and western blot analysis. CIH caused the inactivation of myocardial SIRT6 and AMPK-FOXO3a signalling. BBR dose-dependently ameliorated cardiac injury in CIH-induced mice, as evidenced by increased cardiac function and decreased fibrosis. Notably, SIRT6 overexpression mimicked these beneficial effects, whereas infection with recombinant AAV9 carrying SIRT6-specific short hairpin RNA abrogated them. Mechanistically, BBR reduced oxidative stress damage and preserved mitochondrial function via activating SIRT6-AMPK-FOXO3a signalling, enhancing mitochondrial biogenesis as well as PINK1-Parkin-mediated mitophagy. Taken together, these data demonstrate that SIRT6 activation protects against the pathogenesis of CIH-induced cardiac dysfunction. BBR attenuates CIH-induced myocardial injury by improving mitochondrial biogenesis and PINK1-Parkin-dependent mitophagy via the SIRT6-AMPK-FOXO3a signalling pathway.


Berberine , Forkhead Box Protein O3 , Hypoxia , Signal Transduction , Sirtuins , Berberine/pharmacology , Berberine/therapeutic use , Animals , Sirtuins/metabolism , Sirtuins/genetics , Signal Transduction/drug effects , Hypoxia/metabolism , Mice , Male , Forkhead Box Protein O3/metabolism , Forkhead Box Protein O3/genetics , Oxidative Stress/drug effects , Mice, Inbred C57BL , AMP-Activated Protein Kinases/metabolism , Mitochondria/metabolism , Mitochondria/drug effects , Mitophagy/drug effects , Ventricular Remodeling/drug effects , Disease Models, Animal
8.
Eur J Sport Sci ; 24(4): 474-486, 2024 Apr.
Article En | MEDLINE | ID: mdl-38895874

Even if more and more women are involved in strength-training (ST) programs in fitness centers, studies on strength gain, body composition, and cardiac remodeling were mainly conducted in men and whether they are similar in women remains to be explored. In this context, the aim of our study was to assess the effect of a supervised ST program on strength gains, body composition, and cardiac remodeling in previously untrained women and men. 17 healthy and previously untrained young women and 17 young men participated in a supervised 16-week ST program built according to the recommendation of the American College of Sports Medicine in terms of intensity, and strictly using similar volume and intensity in both groups. Strength performance, body composition, and cardiac remodeling were evaluated every 4 weeks. Cardiac adaptations were assessed using resting echocardiography, including regional 2D-Strain analysis of the left atrium and ventricle (LA and LV, respectively). Despite lower values at baseline, women exhibited similar or even higher strength gains compared to men. ST induced a decrease of body and abdominal fat mass and an increase of lean body mass in both groups. Similar cardiac remodeling was observed in women, and women, including an early and progressive LV and LA enlargement throughout the ST program, without any alteration of LV diastolic and systolic functions. These findings underlie that ST programs are highly suitable for women to enhance their strength performance and their cardiovascular health.


Body Composition , Resistance Training , Ventricular Remodeling , Humans , Resistance Training/methods , Male , Female , Body Composition/physiology , Ventricular Remodeling/physiology , Young Adult , Adult , Echocardiography , Muscle Strength/physiology , Physical Functional Performance , Adaptation, Physiological , Sex Factors
9.
Sci Robot ; 9(91): eadj9769, 2024 Jun 12.
Article En | MEDLINE | ID: mdl-38865476

Our understanding of cardiac remodeling processes due to left ventricular pressure overload derives largely from animal models of aortic banding. However, these studies fail to enable control over both disease progression and reversal, hindering their clinical relevance. Here, we describe a method for progressive and reversible aortic banding based on an implantable expandable actuator that can be finely tuned to modulate aortic banding and debanding in a rat model. Through catheterization, imaging, and histologic studies, we demonstrate that our platform can recapitulate the hemodynamic and structural changes associated with pressure overload in a controllable manner. We leveraged soft robotics to enable noninvasive aortic debanding, demonstrating that these changes can be partly reversed because of cessation of the biomechanical stimulus. By recapitulating longitudinal disease progression and reversibility, this animal model could elucidate fundamental mechanisms of cardiac remodeling and optimize timing of intervention for pressure overload.


Aorta , Disease Models, Animal , Animals , Rats , Robotic Surgical Procedures/instrumentation , Hemodynamics , Ventricular Remodeling/physiology , Male , Equipment Design , Rats, Sprague-Dawley , Robotics/instrumentation , Constriction , Biomechanical Phenomena
10.
FASEB J ; 38(11): e23709, 2024 Jun 15.
Article En | MEDLINE | ID: mdl-38809700

Brown adipose tissue (BAT) is correlated to cardiovascular health in rodents and humans, but the physiological role of BAT in the initial cardiac remodeling at the onset of stress is unknown. Activation of BAT via 48 h cold (16°C) in mice following transverse aortic constriction (TAC) reduced cardiac gene expression for LCFA uptake and oxidation in male mice and accelerated the onset of cardiac metabolic remodeling, with an early isoform shift of carnitine palmitoyltransferase 1 (CPT1) toward increased CPT1a, reduced entry of long chain fatty acid (LCFA) into oxidative metabolism (0.59 ± 0.02 vs. 0.72 ± 0.02 in RT TAC hearts, p < .05) and increased carbohydrate oxidation with altered glucose transporter content. BAT activation with TAC reduced early hypertrophic expression of ß-MHC by 61% versus RT-TAC and reduced pro-fibrotic TGF-ß1 and COL3α1 expression. While cardiac natriuretic peptide expression was yet to increase at only 3 days TAC, Nppa and Nppb expression were elevated in Cold TAC versus RT TAC hearts 2.7- and 2.4-fold, respectively. Eliminating BAT thermogenic activation with UCP1 KO mice eliminated differences between Cold TAC and RT TAC hearts, confirming effects of BAT activation rather than autonomous cardiac responses to cold. Female responses to BAT activation were blunted, with limited UCP1 changes with cold, partly due to already activated BAT in females at RT compared to thermoneutrality. These data reveal a previously unknown physiological mechanism of UCP1-dependent BAT activation in attenuating early cardiac hypertrophic and profibrotic signaling and accelerating remodeled metabolic activity in the heart at the onset of cardiac stress.


Adipose Tissue, Brown , Fibrosis , Uncoupling Protein 1 , Animals , Adipose Tissue, Brown/metabolism , Mice , Male , Uncoupling Protein 1/metabolism , Fibrosis/metabolism , Carnitine O-Palmitoyltransferase/metabolism , Carnitine O-Palmitoyltransferase/genetics , Mice, Inbred C57BL , Cardiomegaly/metabolism , Cardiomegaly/pathology , Myocardium/metabolism , Myocardium/pathology , Stress, Physiological , Ventricular Remodeling/physiology , Mice, Knockout , Cold Temperature
12.
J Am Heart Assoc ; 13(10): e028006, 2024 May 21.
Article En | MEDLINE | ID: mdl-38726894

BACKGROUND: S100a8/9 (S100 calcium binding protein a8/9) belongs to the S100 family and has gained a lot of interest as a critical regulator of inflammatory response. Our previous study found that S100a8/9 homolog promoted aortic valve sclerosis in mice with chronic kidney disease. However, the role of S100a8/9 in pressure overload-induced cardiac hypertrophy remains unclear. The present study was to explore the role of S100a8/9 in cardiac hypertrophy. METHODS AND RESULTS: Cardiomyocyte-specific S100a9 loss or gain of function was achieved using an adeno-associated virus system, and the model of cardiac hypertrophy was established by aortic banding-induced pressure overload. The results indicate that S100a8/9 expression was increased in response to pressure overload. S100a9 deficiency alleviated pressure overload-induced hypertrophic response, whereas S100a9 overexpression accelerated cardiac hypertrophy. S100a9-overexpressed mice showed increased FGF23 (fibroblast growth factor 23) expression in the hearts after exposure to pressure overload, which activated calcineurin/NFAT (nuclear factor of activated T cells) signaling in cardiac myocytes and thus promoted hypertrophic response. A specific antibody that blocks FGFR4 (FGF receptor 4) largely abolished the prohypertrophic response of S100a9 in mice. CONCLUSIONS: In conclusion, S100a8/9 promoted the development of cardiac hypertrophy in mice. Targeting S100a8/9 may be a promising therapeutic approach to treat cardiac hypertrophy.


Calgranulin A , Calgranulin B , Disease Models, Animal , Fibroblast Growth Factor-23 , Fibroblast Growth Factors , Myocytes, Cardiac , NFATC Transcription Factors , Up-Regulation , Animals , Calgranulin A/metabolism , Calgranulin A/genetics , Fibroblast Growth Factors/metabolism , Fibroblast Growth Factors/genetics , Calgranulin B/metabolism , Calgranulin B/genetics , NFATC Transcription Factors/metabolism , NFATC Transcription Factors/genetics , Fibroblast Growth Factor-23/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Signal Transduction , Cardiomegaly/metabolism , Cardiomegaly/pathology , Mice, Inbred C57BL , Male , Mice, Knockout , Calcineurin/metabolism , Mice , Hypertrophy, Left Ventricular/metabolism , Hypertrophy, Left Ventricular/physiopathology , Hypertrophy, Left Ventricular/genetics , Hypertrophy, Left Ventricular/pathology , Ventricular Remodeling
13.
J Am Heart Assoc ; 13(9): e032172, 2024 May 07.
Article En | MEDLINE | ID: mdl-38700022

BACKGROUND: The purpose of this study was to investigate a therapeutic approach targeting the inflammatory response and consequent remodeling from ischemic myocardial injury. METHODS AND RESULTS: Coronary thrombus aspirates were collected from patients at the time of ST-segment-elevation myocardial infarction and subjected to array-based proteome analysis. Clinically indistinguishable at myocardial infarction (MI), patients were stratified into vulnerable and resilient on the basis of 1-year left ventricular ejection fraction and death. Network analysis from coronary aspirates revealed prioritization of tumor necrosis factor-α signaling in patients with worse clinical outcomes. Infliximab, a tumor necrosis factor-α inhibitor, was infused intravenously at reperfusion in a porcine MI model to assess whether infliximab-mediated immune modulation impacts post-MI injury. At 3 days after MI (n=7), infliximab infusion increased proregenerative M2 macrophages in the myocardial border zone as quantified by immunofluorescence (24.1%±23.3% in infliximab versus 9.29%±8.7% in sham; P<0.01). Concomitantly, immunoassays of coronary sinus samples quantified lower troponin I levels (41.72±7.34 pg/mL versus 58.11±10.75 pg/mL; P<0.05) and secreted protein analysis revealed upregulation of injury-modifying interleukin-2, -4, -10, -12, and -18 cytokines in the infliximab-treated cohort. At 4 weeks (n=12), infliximab treatment resulted in significant protective influence, improving left ventricular ejection fraction (53.9%±5.4% versus 36.2%±5.3%; P<0.001) and reducing scar size (8.31%±10.9% versus 17.41%±12.5%; P<0.05). CONCLUSIONS: Profiling of coronary thrombus aspirates in patients with ST-segment-elevation MI revealed highest association for tumor necrosis factor-α in injury risk. Infliximab-mediated immune modulation offers an actionable pathway to alter MI-induced inflammatory response, preserving contractility and limiting adverse structural remodeling.


Disease Models, Animal , Infliximab , Ventricular Remodeling , Infliximab/therapeutic use , Infliximab/pharmacology , Animals , Humans , Male , Middle Aged , Ventricular Remodeling/drug effects , Female , ST Elevation Myocardial Infarction/drug therapy , ST Elevation Myocardial Infarction/immunology , Ventricular Function, Left/drug effects , Swine , Aged , Tumor Necrosis Factor-alpha/metabolism , Stroke Volume/drug effects , Coronary Thrombosis/prevention & control , Coronary Thrombosis/drug therapy , Myocardium/pathology , Myocardium/metabolism , Myocardium/immunology , Troponin I/blood , Troponin I/metabolism , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism
14.
Front Med ; 18(3): 484-498, 2024 Jun.
Article En | MEDLINE | ID: mdl-38743133

lncRNA ZNF593 antisense (ZNF593-AS) transcripts have been implicated in heart failure through the regulation of myocardial contractility. The decreased transcriptional activity of ZNF593-AS has also been detected in cardiac hypertrophy. However, the function of ZNF593-AS in cardiac hypertrophy remains unclear. Herein, we report that the expression of ZNF593-AS reduced in a mouse model of left ventricular hypertrophy and cardiomyocytes in response to treatment with the hypertrophic agonist phenylephrine (PE). In vivo, ZNF593-AS aggravated pressure overload-induced cardiac hypertrophy in knockout mice. By contrast, cardiomyocyte-specific transgenic mice (ZNF593-AS MHC-Tg) exhibited attenuated TAC-induced cardiac hypertrophy. In vitro, vector-based overexpression using murine or human ZNF593-AS alleviated PE-induced myocyte hypertrophy, whereas GapmeR-induced inhibition aggravated hypertrophic phenotypes. By using RNA-seq and gene set enrichment analyses, we identified a link between ZNF593-AS and oxidative phosphorylation and found that mitofusin 2 (Mfn2) is a direct target of ZNF593-AS. ZNF593-AS exerts an antihypertrophic effect by upregulating Mfn2 expression and improving mitochondrial function. Therefore, it represents a promising therapeutic target for combating pathological cardiac remodeling.


Cardiomegaly , GTP Phosphohydrolases , Myocytes, Cardiac , RNA, Long Noncoding , Up-Regulation , Animals , Mice , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Myocytes, Cardiac/metabolism , Cardiomegaly/genetics , Cardiomegaly/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Disease Models, Animal , Ventricular Remodeling/genetics , Humans , Mice, Knockout , Male , Mice, Transgenic , Mice, Inbred C57BL
15.
Eur J Obstet Gynecol Reprod Biol ; 298: 108-115, 2024 Jul.
Article En | MEDLINE | ID: mdl-38749071

OBJECTIVES: Hypertensive disorders of pregnancy (HDP) are among the leading causes of maternal morbidity and mortality. The primary objective of this study was to ascertain whether maternal cardiac remodeling is more prevalent in HDP than normotensive pregnancy and if significant change in aortic root size is involved. The secondary objective was to determine the types of cardiac remodeling often associated with HDP. METHODS: A systematic search was conducted across four electronic databases, including Medline, PubMed, Cochrane and EMBASE. The reference lists of selected articles were also searched to ensure no relevant studies were missed. The Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) guidelines were followed in this systematic review. RESULTS: Out of 5,278 articles identified by the search terms, 9 were eligible for inclusion in the meta-analysis. The investigation unveiled a greater prevalence of maternal cardiac remodeling in HDP than normotensive pregnancies. The commonest type of maternal cardiac remodeling in both HDP and normotensive pregnancies was eccentric left ventricular hypertrophy, followed by concentric left ventricular remodeling which was more specific to HDP. Notably, left atrial diameter was significantly increased in HDP than normotensive pregnancies, suggesting higher prevalence of diastolic dysfunction. Additionally, the aortic root dimension was significantly increased in HDP than normotensive pregnancies. CONCLUSION: This study underscores the importance of monitoring cardiac health in pregnancy, particularly in those with hypertensive disorders, in order to mitigate potential complications and improve maternal outcomes. Finally, the risk of aortic dissection that may occur as a long-term effect of aortic root enlargement in women with history of HDP ought to be investigated in future studies.


Hypertension, Pregnancy-Induced , Ventricular Remodeling , Humans , Female , Pregnancy , Ventricular Remodeling/physiology , Hypertension, Pregnancy-Induced/physiopathology , Hypertension, Pregnancy-Induced/epidemiology , Hypertrophy, Left Ventricular/physiopathology , Hypertrophy, Left Ventricular/epidemiology
16.
FASEB J ; 38(9): e23654, 2024 May 15.
Article En | MEDLINE | ID: mdl-38717442

Heart failure and cardiac remodeling are both characterized by mitochondrial dysfunction. Healthy mitochondria are required for adequate contractile activity and appropriate regulation of cell survival. In the mammalian heart, enhancement of the mitochondrial unfolded protein response (UPRmt) is cardioprotective under pressure overload conditions. We explored the UPRmt and the underlying regulatory mechanism in terms of hypertension-induced cardiac remodeling and the cardioprotective effect of metformin. Male spontaneously hypertensive rats and angiotensin II-treated neonatal rat cardiomyocytes were used to induce cardiac hypertrophy. The results showed that hypertension induced the formation of aberrant mitochondria, characterized by a reduced mtDNA/nDNA ratio and swelling, as well as lower levels of mitochondrial complexes I to V and inhibition of the expression of one protein subunit of each of complexes I to IV. Such changes eventually enlarged cardiomyocytes and increased cardiac fibrosis. Metformin treatment increased the mtDNA/nDNA ratio and regulated the UPRmt, as indicated by increased expression of activating transcription factor 5, Lon protease 1, and heat shock protein 60, and decreased expression of C/EBP homologous protein. Thus, metformin improved mitochondrial ultrastructure and function in spontaneously hypertensive rats. In vitro analyses revealed that metformin reduced the high levels of angiotensin II-induced mitochondrial reactive oxygen species in such animals and stimulated nuclear translocation of heat shock factor 1 (HSF1). Moreover, HSF1 small-interfering RNA reduced the metformin-mediated improvements in mitochondrial morphology and the UPRmt by suppressing hypertrophic signals and cardiomyocyte apoptosis. These results suggest that HSF1/UPRmt signaling contributes to the beneficial effects of metformin. Metformin-mediated targeting of mitochondrial protein homeostasis and modulation of HSF1 levels have potential therapeutic implications in terms of cardiac remodeling.


Heat Shock Transcription Factors , Metformin , Myocytes, Cardiac , Unfolded Protein Response , Animals , Male , Rats , Angiotensin II/pharmacology , Cardiomegaly/metabolism , Cardiomegaly/drug therapy , Cardiomegaly/pathology , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Heat Shock Transcription Factors/drug effects , Heat Shock Transcription Factors/metabolism , Hypertension/metabolism , Hypertension/drug therapy , Metformin/pharmacology , Mitochondria, Heart/metabolism , Mitochondria, Heart/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Rats, Inbred SHR , Rats, Inbred WKY , Transcription Factors/metabolism , Transcription Factors/genetics , Unfolded Protein Response/drug effects , Ventricular Remodeling/drug effects
17.
Exp Mol Pathol ; 137: 104895, 2024 Jun.
Article En | MEDLINE | ID: mdl-38703553

Lipidome perturbation occurring during meta-inflammation is associated to left ventricle (LV) remodeling though the activation of the NLRP3 inflammasome, a key regulator of chronic inflammation in obesity-related disorders. Little is known about phosphatidylcholine (PC) and phosphatidylethanolamine (PE) as DAMP-induced NLRP3 inflammasome. Our study is aimed to evaluate if a systemic reduction of PC/PE molar ratio can affect NLRP3 plasma levels in cardiovascular disease (CVD) patients with insulin resistance (IR) risk. Forty patients from IRCCS Policlinico San Donato were enrolled, and their blood samples were drawn before heart surgery. LV geometry measurements were evaluated by echocardiography and clinical data associated to IR risk were collected. PC and PE were quantified by ESI-MS/MS. Circulating NLRP3 was quantified by an ELISA assay. Our results have shown that CVD patients with IR risk presented systemic lipid impairment of PC and PE species and their ratio in plasma was inversely associated to NLRP3 levels. Interestingly, CVD patients with IR risk presented LV changes directly associated to increased levels of NLRP3 and a decrease in PC/PE ratio in plasma, highlighting the systemic effect of meta-inflammation in cardiac response. In summary, PC and PE can be considered bioactive mediators associated to both the NLRP3 and LV changes in CVD patients with IR risk.


Cardiovascular Diseases , Inflammasomes , Insulin Resistance , NLR Family, Pyrin Domain-Containing 3 Protein , Phosphatidylcholines , Phosphatidylethanolamines , Ventricular Remodeling , Humans , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Phosphatidylcholines/blood , Inflammasomes/metabolism , Male , Female , Middle Aged , Phosphatidylethanolamines/blood , Phosphatidylethanolamines/metabolism , Cardiovascular Diseases/blood , Cardiovascular Diseases/pathology , Aged
18.
Int J Cardiol ; 409: 132203, 2024 Aug 15.
Article En | MEDLINE | ID: mdl-38795973

BACKGROUND: Sacubitril/valsartan has been demonstrated to promote left ventricular (LV) reverse remodelling and improve outcomes in patients with heart failure (HF) with reduced ejection fraction (EF). Its molecular and tissue effects have not been fully elucidated yet, due to the paucity of preclinical studies, mostly based on ischaemic models. We aimed to evaluate the effects of sacubitril/valsartan on LV remodelling, myocardial fibrosis and mitochondrial biology in a murine model of non-ischaemic LV dysfunction. METHODS: Adult transgenic male mice with cardiac-specific hyperaldosteronism (AS mice) received subcutaneous isoproterenol injections to induce LV systolic dysfunction. After 7 days, mice were randomized to a 2-week treatment with saline (ISO-AS n = 15), valsartan (ISO + V n = 12) or sacubitril/valsartan (ISO + S/V n = 12). Echocardiography was performed at baseline, at day 7, and after each of the 2 weeks of treatment. After sacrifice at day 21, histological and immunochemical assays were performed. A control group of AS mice was also obtained (Ctrl-AS n = 8). RESULTS: Treatment with sacubitril/valsartan, but not with valsartan, induced a significant improvement in LVEF (p = 0.009 vs ISO-AS) and fractional shortening (p = 0.032 vs ISO-AS) after 2- week treatment. In both ISO + V and ISO + S/V groups, a trend toward reduction of the cardiac collagen 1/3 expression ratio was detected. ISO + V and ISO + S/V groups showed a significant recovery of mitochondrial morphology and inner membrane function meant for oxidative phosphorylation. CONCLUSION: In a murine model of non-ischaemic HF, sacubitril/valsartan proved to have beneficial effects on LV systolic function, and on cardiac energetics, by improving mitochondrial activity.


Aminobutyrates , Biphenyl Compounds , Disease Models, Animal , Drug Combinations , Fibrosis , Isoproterenol , Tetrazoles , Valsartan , Ventricular Dysfunction, Left , Ventricular Remodeling , Animals , Aminobutyrates/pharmacology , Biphenyl Compounds/pharmacology , Mice , Male , Ventricular Remodeling/drug effects , Tetrazoles/pharmacology , Fibrosis/chemically induced , Ventricular Dysfunction, Left/chemically induced , Ventricular Dysfunction, Left/physiopathology , Isoproterenol/toxicity , Mice, Transgenic , Mitochondria, Heart/drug effects , Mitochondria, Heart/metabolism , Angiotensin Receptor Antagonists/pharmacology , Random Allocation
19.
Int Immunopharmacol ; 134: 112143, 2024 Jun 15.
Article En | MEDLINE | ID: mdl-38692016

Chronic inflammation is a significant contributor to hypertensive heart failure. Carnosol (Car), primarily derived from the sage plant (Salvia carnosa), exhibits anti-inflammatory properties in a range of systems. Nevertheless, the influence of angiotensin II (Ang II) on cardiac remodeling remains uncharted. Car was shown to protect mice's hearts against Ang II-induced heart damage at dosages of 20 and 40 mg/kg/d. This protection was evident in a concentration-related decrease in the remodeling of the heart and dysfunction. Examination of the transcriptome revealed that the pivotal roles in mediating the protective effects of Car involved inhibiting Ang II-induced inflammation and the activation of the mitogen-activated protein kinase (MAPK) pathway. Furthermore, Car was found to inhibit p38 phosphorylation, therefore reducing the level of inflammation in cultured cardiomyocytes and mouse hearts. This effect was attributed to the direct binding to p38 and inhibition of p38 protein phosphorylation by Car both in vitro and in vivo. In addition, the effects of Car on inflammation were neutralized when p38 was blocked in cardiomyocytes.


Abietanes , Angiotensin II , Anti-Inflammatory Agents , Mice, Inbred C57BL , Myocytes, Cardiac , Ventricular Remodeling , p38 Mitogen-Activated Protein Kinases , Animals , Angiotensin II/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Male , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Mice , Abietanes/pharmacology , Abietanes/therapeutic use , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Ventricular Remodeling/drug effects , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/chemically induced , Phosphorylation/drug effects , Cells, Cultured
20.
Front Med ; 18(3): 499-515, 2024 Jun.
Article En | MEDLINE | ID: mdl-38806989

Cardiac fibrosis caused by ventricular remodeling and dysfunction such as post-myocardial infarction (MI) can lead to heart failure. RNA N6-methyladenosine (m6A) methylation has been shown to play a pivotal role in the occurrence and development of many illnesses. In investigating the biological function of the m6A reader YTHDF1 in cardiac fibrosis, adeno-associated virus 9 was used to knock down or overexpress the YTHDF1 gene in mouse hearts, and MI surgery in vivo and transforming growth factor-ß (TGF-ß)-activated cardiac fibroblasts in vitro were performed to establish fibrosis models. Our results demonstrated that silencing YTHDF1 in mouse hearts can significantly restore impaired cardiac function and attenuate myocardial fibrosis, whereas YTHDF1 overexpression could further enhance cardiac dysfunction and aggravate the occurrence of ventricular pathological remodeling and fibrotic development. Mechanistically, zinc finger BED-type containing 6 mediated the transcriptional function of the YTHDF1 gene promoter. YTHDF1 augmented AXL translation and activated the TGF-ß-Smad2/3 signaling pathway, thereby aggravating the occurrence and development of cardiac dysfunction and myocardial fibrosis. Consistently, our data indicated that YTHDF1 was involved in activation, proliferation, and migration to participate in cardiac fibrosis in vitro. Our results revealed that YTHDF1 could serve as a potential therapeutic target for myocardial fibrosis.


Axl Receptor Tyrosine Kinase , Fibrosis , Myocardial Infarction , Proto-Oncogene Proteins , RNA-Binding Proteins , Receptor Protein-Tyrosine Kinases , Animals , Mice , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Male , Mice, Inbred C57BL , Signal Transduction , Myocardium/pathology , Myocardium/metabolism , Transforming Growth Factor beta/metabolism , Ventricular Remodeling/genetics , Disease Models, Animal , Adenosine/analogs & derivatives , Adenosine/metabolism , Fibroblasts/metabolism
...