Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 3.150
Filtrer
1.
FASEB J ; 38(13): e23780, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38948938

RÉSUMÉ

Aerobic training (AT), an effective form of cardiac rehabilitation, has been shown to be beneficial for cardiac repair and remodeling after myocardial infarction (MI). The p300/CBP-associated factor (PCAF) is one of the most important lysine acetyltransferases and is involved in various biological processes. However, the role of PCAF in AT and AT-mediated cardiac remodeling post-MI has not been determined. Here, we found that the PCAF protein level was significantly increased after MI, while AT blocked the increase in PCAF. AT markedly improved cardiac remodeling in mice after MI by reducing endoplasmic reticulum stress (ERS). In vivo, similar to AT, pharmacological inhibition of PCAF by Embelin improved cardiac recovery and attenuated ERS in MI mice. Furthermore, we observed that both IGF-1, a simulated exercise environment, and Embelin protected from H2O2-induced cardiomyocyte injury, while PCAF overexpression by viruses or the sirtuin inhibitor nicotinamide eliminated the protective effect of IGF-1 in H9C2 cells. Thus, our data indicate that maintaining low PCAF levels plays an essential role in AT-mediated cardiac protection, and PCAF inhibition represents a promising therapeutic target for attenuating cardiac remodeling after MI.


Sujet(s)
Infarctus du myocarde , Conditionnement physique d'animal , Remodelage ventriculaire , Facteurs de transcription CBP-p300 , Animaux , Facteurs de transcription CBP-p300/métabolisme , Facteurs de transcription CBP-p300/antagonistes et inhibiteurs , Infarctus du myocarde/métabolisme , Infarctus du myocarde/anatomopathologie , Souris , Remodelage ventriculaire/effets des médicaments et des substances chimiques , Remodelage ventriculaire/physiologie , Mâle , Souris de lignée C57BL , Myocytes cardiaques/métabolisme , Stress du réticulum endoplasmique/effets des médicaments et des substances chimiques
2.
Cells ; 13(13)2024 Jun 21.
Article de Anglais | MEDLINE | ID: mdl-38994928

RÉSUMÉ

Activation of the CXCL12/CXCR4/ACKR3 axis is known to aid myocardial repair through ischemia-triggered hypoxia-inducible factor-1α (HIF-1α). To enhance the upregulation of HIF-1α, we administered roxadustat, a novel prolyl hydroxylase inhibitor (PHI) clinically approved by the European Medicines Agency 2021 for the treatment of renal anemia, with the purpose of improving LV function and attenuating ischemic cardiomyopathy. METHODS: We evaluated roxadustat's impact on HIF-1 stimulation, cardiac remodeling, and function after MI. Therefore, we analyzed nuclear HIF-1 expression, the mRNA and protein expression of key HIF-1 target genes (RT-PCR, Western blot), inflammatory cell infiltration (immunohistochemistry), and apoptosis (TUNEL staining) 7 days after MI. Additionally, we performed echocardiography in male and female C57BL/6 mice 28 days post-MI. RESULTS: We found a substantial increase in nuclear HIF-1, associated with an upregulation of HIF-1α target genes like CXCL12/CXCR4/ACKR3 at the mRNA and protein levels. Roxadustat increased the proportion of myocardial reparative M2 CD206+ cells, suggesting beneficial alterations in immune cell migration and a trend towards reduced apoptosis. Echocardiography showed that roxadustat treatment significantly preserved ejection fraction and attenuated subsequent ventricular dilatation, thereby reducing adverse remodeling. CONCLUSIONS: Our findings suggest that roxadustat is a promising clinically approved treatment option to preserve myocardial function by attenuating adverse remodeling.


Sujet(s)
Glycine , Sous-unité alpha du facteur-1 induit par l'hypoxie , Isoquinoléines , Souris de lignée C57BL , Infarctus du myocarde , Remodelage ventriculaire , Animaux , Infarctus du myocarde/traitement médicamenteux , Infarctus du myocarde/anatomopathologie , Infarctus du myocarde/métabolisme , Souris , Remodelage ventriculaire/effets des médicaments et des substances chimiques , Glycine/analogues et dérivés , Glycine/pharmacologie , Glycine/usage thérapeutique , Mâle , Femelle , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Sous-unité alpha du facteur-1 induit par l'hypoxie/génétique , Isoquinoléines/pharmacologie , Isoquinoléines/usage thérapeutique , Apoptose/effets des médicaments et des substances chimiques , Chimiokine CXCL12/métabolisme , Chimiokine CXCL12/génétique , Myocarde/anatomopathologie , Myocarde/métabolisme
3.
Int J Med Sci ; 21(9): 1718-1729, 2024.
Article de Anglais | MEDLINE | ID: mdl-39006833

RÉSUMÉ

Isoproterenol (ISO) administration is a well-established model for inducing myocardial injury, replicating key features of human myocardial infarction (MI). The ensuing inflammatory response plays a pivotal role in the progression of adverse cardiac remodeling, characterized by myocardial dysfunction, fibrosis, and hypertrophy. The Mst1/Hippo signaling pathway, a critical regulator of cellular processes, has emerged as a potential therapeutic target in cardiovascular diseases. This study investigates the role of Mst1 in ISO-induced myocardial injury and explores its underlying mechanisms. Our findings demonstrate that Mst1 ablation in cardiomyocytes attenuates ISO-induced cardiac dysfunction, preserving cardiomyocyte viability and function. Mechanistically, Mst1 deletion inhibits cardiomyocyte apoptosis, oxidative stress, and calcium overload, key contributors to myocardial injury. Furthermore, Mst1 ablation mitigates endoplasmic reticulum (ER) stress and mitochondrial fission, both of which are implicated in ISO-mediated cardiac damage. Additionally, Mst1 plays a crucial role in modulating the inflammatory response following ISO treatment, as its deletion suppresses pro-inflammatory cytokine expression and neutrophil infiltration. To further investigate the molecular mechanisms underlying ISO-induced myocardial injury, we conducted a bioinformatics analysis using the GSE207581 dataset. GO and KEGG pathway enrichment analyses revealed significant enrichment of genes associated with DNA damage response, DNA repair, protein ubiquitination, chromatin organization, autophagy, cell cycle, mTOR signaling, FoxO signaling, ubiquitin-mediated proteolysis, and nucleocytoplasmic transport. These findings underscore the significance of Mst1 in ISO-induced myocardial injury and highlight its potential as a therapeutic target for mitigating adverse cardiac remodeling. Further investigation into the intricate mechanisms of Mst1 signaling may pave the way for novel therapeutic interventions for myocardial infarction and heart failure.


Sujet(s)
Voie de signalisation Hippo , Isoprénaline , Infarctus du myocarde , Myocytes cardiaques , Protein-Serine-Threonine Kinases , Transduction du signal , Animaux , Isoprénaline/effets indésirables , Myocytes cardiaques/métabolisme , Myocytes cardiaques/anatomopathologie , Myocytes cardiaques/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Protein-Serine-Threonine Kinases/métabolisme , Protein-Serine-Threonine Kinases/génétique , Souris , Humains , Infarctus du myocarde/anatomopathologie , Infarctus du myocarde/induit chimiquement , Infarctus du myocarde/métabolisme , Infarctus du myocarde/génétique , Remodelage ventriculaire/effets des médicaments et des substances chimiques , Stress oxydatif/effets des médicaments et des substances chimiques , Stress du réticulum endoplasmique/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques , Apoptose/génétique , Inflammation/induit chimiquement , Inflammation/métabolisme , Inflammation/génétique , Inflammation/anatomopathologie , Modèles animaux de maladie humaine , Protéines proto-oncogènes , Facteur de croissance des hépatocytes
4.
BMC Cardiovasc Disord ; 24(1): 356, 2024 Jul 12.
Article de Anglais | MEDLINE | ID: mdl-38997620

RÉSUMÉ

BACKGROUND AND AIMS: Sodium-glucose co-transporter 2 (SGLT2) inhibitors have beneficial effects in heart failure (HF), including reverse remodelling, but the mechanisms by which these benefits are conferred are unclear. Inflammation is implicated in the pathophysiology of heart failure (HF) and there are some pre-clinical data suggesting that SGLT2 inhibitors may reduce inflammation. There is however a lack of clinical data. The aim of our study was to investigate whether improvements in cardiac remodelling caused by dapagliflozin in individuals with type 2 diabetes (T2D) and left ventricular hypertrophy (LVH) were associated with its effects on inflammation. METHODS: We measured C-reactive protein (CRP), tumor necrosis factor alpha (TNF-α), interleukin-1ß (IL-1ß), interleukin 6 (IL-6), and interleukin 10 (IL-10) and neutrophil-to-lymphocyte ratio (NLR) in plasma samples of 60 patients with T2D and left ventricular hypertrophy (LVH) but without symptomatic HF from the DAPA-LVH trial in which participants were randomised dapagliflozin 10 mg daily or placebo for 12 months and underwent cardiac magnetic resonance imaging (CMR) at baseline and end of treatment. The primary analysis was to investigate the effect of dapagliflozin on inflammation and to assess the relationships between changes in inflammatory markers and LV mass and global longitudinal strain (GLS) and whether the effect of dapagliflozin on LV mass and GLS was modulated by baseline levels of inflammation. RESULTS: Following 12 months of treatment dapagliflozin significantly reduced CRP compared to placebo (mean difference of -1.96; 95% CI -3.68 to -0.24, p = 0.026). There were no significant statistical changes in other inflammatory markers. There were modest correlations between improvements in GLS and reduced inflammation (NLR (r = 0.311), IL-1ß (r = 0.246), TNF-α (r = 0.230)) at 12 months. CONCLUSIONS: Dapagliflozin caused a significant reduction in CRP compared to placebo. There were correlations between reductions in inflammatory markers including IL-1ß and improvements in global longitudinal strain (but not reduced LV mass). Reductions in systemic inflammation might play a contributory role in the cardiovascular benefits of dapagliflozin. TRIAL REGISTRATION: Clinicaltrials.gov NCT02956811 (06/11/2016).


Sujet(s)
Composés benzhydryliques , Marqueurs biologiques , Diabète de type 2 , Glucosides , Hypertrophie ventriculaire gauche , Médiateurs de l'inflammation , Inhibiteurs du cotransporteur sodium-glucose de type 2 , Fonction ventriculaire gauche , Remodelage ventriculaire , Humains , Glucosides/usage thérapeutique , Composés benzhydryliques/usage thérapeutique , Hypertrophie ventriculaire gauche/physiopathologie , Hypertrophie ventriculaire gauche/traitement médicamenteux , Hypertrophie ventriculaire gauche/imagerie diagnostique , Hypertrophie ventriculaire gauche/étiologie , Diabète de type 2/traitement médicamenteux , Diabète de type 2/sang , Diabète de type 2/complications , Diabète de type 2/diagnostic , Diabète de type 2/physiopathologie , Remodelage ventriculaire/effets des médicaments et des substances chimiques , Mâle , Femelle , Inhibiteurs du cotransporteur sodium-glucose de type 2/usage thérapeutique , Adulte d'âge moyen , Fonction ventriculaire gauche/effets des médicaments et des substances chimiques , Résultat thérapeutique , Médiateurs de l'inflammation/sang , Marqueurs biologiques/sang , Sujet âgé , Facteurs temps , Inflammation/traitement médicamenteux , Inflammation/sang , Inflammation/physiopathologie , Inflammation/diagnostic , Méthode en double aveugle , Anti-inflammatoires/usage thérapeutique , Cytokines/sang
5.
J Cell Mol Med ; 28(13): e18493, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38963241

RÉSUMÉ

Interleukin-5 (IL-5) has been reported to be involved in cardiovascular diseases, such as atherosclerosis and cardiac injury. This study aimed to investigate the effects of IL-5 on cardiac remodelling. Mice were infused with angiotensin II (Ang II), and the expression and source of cardiac IL-5 were analysed. The results showed that cardiac IL-5 expression was time- and dose-dependently decreased after Ang II infusion, and was mainly derived from cardiac macrophages. Additionally, IL-5-knockout (IL-5-/-) mice were used to observe the effects of IL-5 knockout on Ang II-induced cardiac remodelling. We found knockout of IL-5 significantly increased the expression of cardiac hypertrophy markers, elevated myocardial cell cross-sectional areas and worsened cardiac dysfunction in Ang II-infused mice. IL-5 deletion also promoted M2 macrophage differentiation and exacerbated cardiac fibrosis. Furthermore, the effects of IL-5 deletion on cardiac remodelling was detected after the STAT3 pathway was inhibited by S31-201. The effects of IL-5 on cardiac remodelling and M2 macrophage differentiation were reversed by S31-201. Finally, the effects of IL-5 on macrophage differentiation and macrophage-related cardiac hypertrophy and fibrosis were analysed in vitro. IL-5 knockout significantly increased the Ang II-induced mRNA expression of cardiac hypertrophy markers in myocardial cells that were co-cultured with macrophages, and this effect was reversed by S31-201. Similar trends in the mRNA levels of fibrosis markers were observed when cardiac fibroblasts and macrophages were co-cultured. In conclusions, IL-5 deficiency promote the differentiation of M2 macrophages by activating the STAT3 pathway, thereby exacerbating cardiac remodelling in Ang II-infused mice. IL-5 may be a potential target for the clinical prevention of cardiac remodelling.


Sujet(s)
Angiotensine-II , Cardiomégalie , Fibrose , Interleukine-5 , Macrophages , Souris knockout , Facteur de transcription STAT-3 , Transduction du signal , Remodelage ventriculaire , Animaux , Angiotensine-II/pharmacologie , Facteur de transcription STAT-3/métabolisme , Facteur de transcription STAT-3/génétique , Remodelage ventriculaire/effets des médicaments et des substances chimiques , Souris , Macrophages/métabolisme , Macrophages/effets des médicaments et des substances chimiques , Interleukine-5/métabolisme , Interleukine-5/génétique , Cardiomégalie/métabolisme , Cardiomégalie/anatomopathologie , Cardiomégalie/génétique , Cardiomégalie/induit chimiquement , Mâle , Souris de lignée C57BL , Différenciation cellulaire , Myocarde/métabolisme , Myocarde/anatomopathologie
6.
Phytother Res ; 38(7): 3763-3781, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38831669

RÉSUMÉ

Cardiac remodeling is a commonly observed pathophysiological phenomenon associated with the progression of heart failure in various cardiovascular disorders. Carnosol, a phenolic compound extracted from rosemary, possesses noteworthy pharmacological properties including anti-inflammatory, antioxidant, and anti-apoptotic activities. Considering the pivotal involvement of inflammation, oxidative stress, and apoptosis in cardiac remodeling, the present study aims to assess the effects of carnosol on cardiac remodeling and elucidate the underlying mechanisms. In an in vivo model, cardiac remodeling was induced by performing transverse aortic constriction (TAC) surgery on mice, while an in vitro model was established by treating neonatal rat cardiomyocytes (NRCMs) with Ang II. Our results revealed that carnosol treatment effectively ameliorated TAC-induced myocardial hypertrophy and fibrosis, thereby attenuating cardiac dysfunction in mice. Moreover, carnosol improved cardiac electrical remodeling and restored connexin 43 expression, thereby reducing the vulnerability to ventricular fibrillation (VF). Furthermore, carnosol significantly reduced Ang II-induced cardiomyocyte hypertrophy in NRCMs and alleviated the upregulation of hypertrophy and fibrosis markers. Both in vivo and in vitro models of cardiac remodeling exhibited the anti-inflammatory, anti-oxidative, and anti-apoptotic effects of carnosol. Mechanistically, these effects were mediated through the Sirt1/PI3K/AKT pathway, as the protective effects of carnosol were abrogated upon inhibition of Sirt1 or activation of the PI3K/AKT pathway. In summary, our study suggests that carnosol prevents cardiac structural and electrical remodeling by regulating the anti-inflammatory, anti-oxidative, and anti-apoptotic effects mediated by Sirt1/PI3K/AKT signaling pathways, thereby alleviating heart failure and VF.


Sujet(s)
Abiétanes , Défaillance cardiaque , Myocytes cardiaques , Remodelage ventriculaire , Animaux , Souris , Remodelage ventriculaire/effets des médicaments et des substances chimiques , Défaillance cardiaque/traitement médicamenteux , Abiétanes/pharmacologie , Myocytes cardiaques/effets des médicaments et des substances chimiques , Mâle , Rats , Souris de lignée C57BL , Modèles animaux de maladie humaine , Apoptose/effets des médicaments et des substances chimiques , Troubles du rythme cardiaque/traitement médicamenteux , Troubles du rythme cardiaque/prévention et contrôle , Protéines proto-oncogènes c-akt/métabolisme , Stress oxydatif/effets des médicaments et des substances chimiques , Phosphatidylinositol 3-kinases/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Antioxydants/pharmacologie , Fibrose , Sirtuine-1/métabolisme , Rat Sprague-Dawley , Angiotensine-II , Cardiomégalie/traitement médicamenteux
7.
Chem Biol Interact ; 398: 111104, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38906502

RÉSUMÉ

Interrupted ER homeostasis contributes to the etiology of obesity cardiomyopathy although it remains elusive how ER stress evokes cardiac anomalies in obesity. Our study evaluated the impact of ER stress inhibition on cardiac anomalies in obesity. Lean and ob/ob obese mice received chemical ER chaperone tauroursodeoxycholic acid (TUDCA, 50 mg/kg/d, p.o.) for 35 days prior to evaluation of glucose sensitivity, echocardiographic, myocardial geometric, cardiomyocyte mechanical and subcellular Ca2+ property, mitochondrial integrity, oxidative stress, apoptosis, and ferroptosis. Intracellular Ca2+ governing domains including sarco(endo)plasmic reticulum Ca2+-ATPase (SERCA) were monitored by45Ca2+uptake and immunoblotting. Our results noted that TUDCA alleviated myocardial remodeling (fibrosis, hypertrophy, enlarged LVESD), echocardiographic anomalies (compromised fractional shortening and ejection fraction), cardiomyocyte contractile dysfunction (amplitude and velocity of cell shortening, relengthening time) and intracellular Ca2+ anomalies (compromised subcellular Ca2+ release, clearance and SERCA function), mitochondrial damage (collapsed membrane potential, downregulated mitochondrial elements and ultrastructural alteration), ER stress (GRP78, eIF2α and ATF4), oxidative stress, apoptosis and ferroptosis [downregulated SLC7A11, GPx4 and upregulated transferrin receptor (TFRC)] without affecting global glucose sensitivity and serum Fe2+ in obese mice. Obesity-evoked change in HSP90, phospholamban and Na+-Ca2+ exchanger was spared by the chemical ER chaperone. Moreover, in vitro results noted that TUDCA, PERK inhibitor GSK2606414, TFRC neutralizing antibody and ferroptosis inhibitor LIP1 mitigated palmitic acid-elicited changes in lipid peroxidation and mechanical function. Our findings favored a role for ferroptosis in obesity cardiomyopathy downstream of ER stress.


Sujet(s)
Chaperonne BiP du réticulum endoplasmique , Stress du réticulum endoplasmique , Ferroptose , Obésité , Acide taurochénodésoxycholique , Acide taurochénodésoxycholique/pharmacologie , Animaux , Stress du réticulum endoplasmique/effets des médicaments et des substances chimiques , Souris , Ferroptose/effets des médicaments et des substances chimiques , Obésité/traitement médicamenteux , Obésité/métabolisme , Mâle , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myocytes cardiaques/métabolisme , Myocytes cardiaques/anatomopathologie , Calcium/métabolisme , Souris de lignée C57BL , Remodelage ventriculaire/effets des médicaments et des substances chimiques , Stress oxydatif/effets des médicaments et des substances chimiques , Contraction myocardique/effets des médicaments et des substances chimiques , Souris obèse
8.
J Cell Mol Med ; 28(12): e18407, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38894630

RÉSUMÉ

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.


Sujet(s)
Berbérine , Protéine O3 à motif en tête de fourche , Hypoxie , Transduction du signal , Sirtuines , Berbérine/pharmacologie , Berbérine/usage thérapeutique , Animaux , Sirtuines/métabolisme , Sirtuines/génétique , Transduction du signal/effets des médicaments et des substances chimiques , Hypoxie/métabolisme , Souris , Mâle , Protéine O3 à motif en tête de fourche/métabolisme , Protéine O3 à motif en tête de fourche/génétique , Stress oxydatif/effets des médicaments et des substances chimiques , Souris de lignée C57BL , AMP-Activated Protein Kinases/métabolisme , Mitochondries/métabolisme , Mitochondries/effets des médicaments et des substances chimiques , Mitophagie/effets des médicaments et des substances chimiques , Remodelage ventriculaire/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine
9.
Int J Mol Sci ; 25(11)2024 Jun 05.
Article de Anglais | MEDLINE | ID: mdl-38892396

RÉSUMÉ

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.


Sujet(s)
Agammaglobulinaemia tyrosine kinase , Troubles du rythme cardiaque , Benzamides , Pipéridines , Rat Sprague-Dawley , Animaux , Benzamides/pharmacologie , Benzamides/usage thérapeutique , Mâle , Rats , Agammaglobulinaemia tyrosine kinase/métabolisme , Agammaglobulinaemia tyrosine kinase/antagonistes et inhibiteurs , Troubles du rythme cardiaque/métabolisme , Troubles du rythme cardiaque/induit chimiquement , Pipéridines/pharmacologie , Pipéridines/usage thérapeutique , Potentiels d'action/effets des médicaments et des substances chimiques , Remodelage ventriculaire/effets des médicaments et des substances chimiques , Inhibiteurs de protéines kinases/pharmacologie , Pyrazines/pharmacologie , Calcium/métabolisme , Adénine/analogues et dérivés , Adénine/pharmacologie , Adénine/effets indésirables , Sarcoplasmic Reticulum Calcium-Transporting ATPases/métabolisme , Ventricules cardiaques/effets des médicaments et des substances chimiques , Ventricules cardiaques/métabolisme , Ventricules cardiaques/physiopathologie , Pyrimidines/pharmacologie , Signalisation calcique/effets des médicaments et des substances chimiques , Pyrazoles/pharmacologie
10.
Nat Commun ; 15(1): 4757, 2024 Jun 04.
Article de Anglais | MEDLINE | ID: mdl-38834564

RÉSUMÉ

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.


Sujet(s)
Métabolisme énergétique , Peptides glucagon-like , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires , Animaux , Mâle , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires/métabolisme , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires/génétique , Métabolisme énergétique/effets des médicaments et des substances chimiques , Souris , Peptides glucagon-like/pharmacologie , Peptides glucagon-like/usage thérapeutique , Défaillance cardiaque/traitement médicamenteux , Défaillance cardiaque/métabolisme , Souris de lignée C57BL , Remodelage ventriculaire/effets des médicaments et des substances chimiques , Métabolisme lipidique/effets des médicaments et des substances chimiques , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myocytes cardiaques/métabolisme , Protéine de liaison à l'élément de réponse à l'AMP cyclique/métabolisme , Modèles animaux de maladie humaine , Myocarde/métabolisme , Myocarde/anatomopathologie , Transduction du signal/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Mitochondries/effets des médicaments et des substances chimiques , Cardiomégalie/traitement médicamenteux , Cardiomégalie/métabolisme
11.
Kardiol Pol ; 82(6): 640-646, 2024.
Article de Anglais | MEDLINE | ID: mdl-38767161

RÉSUMÉ

BACKGROUND: Optimal glycemic control is necessary to prevent cardiovascular events in patients with type 2 diabetes. The positive impact of sodium-glucose cotransporter-2 inhibitors (SGLT2i) on cardiovascular events and mortality in these patients has been demonstrated by previous studies although the mechanism is unclear. AIMS: We aimed to compare the influence of SGLT2i on left ventricular remodeling and strain in diabetic patients with coronary artery disease (CAD) and without CAD during 6-month follow-up. METHODS: Between October 2021 and June 2022, 100 diabetic patients with preserved ejection fraction (HbA1c levels 6.5-10%) were started on SGLT2i (empagliflozin or dapagliflozin) and were prospectively followed up. Conventional and speckle-tracking echocardiography was performed by blinded sonographers, at baseline and then at 1 month and 6 months of treatment. After 6 months, the initial and biochemical blood tests were administered, and N-terminal pro-B-type natriuretic peptide levels of the patients were measured. RESULTS: Patients with CAD were older (P = 0.008), more frequently hypertensive (P = 0.035), and had dyslipidemia (P = 0.021). N-terminal pro-B-type natriuretic peptide levels did not change significantly after treatment in both groups. Left ventricular ejection fraction, global, 2-chamber, and 3-chamber strain values were improved significantly following SGLTi administration for the overall patient cohort, regardless of CAD status (P < 0.05 for all groups). CONCLUSIONS: Treatment with SGLT2i resulted in improvement in left ventricular strain parameters, which indicates that they might have a positive impact on outcomes for diabetic patients with preserved EF.


Sujet(s)
Maladie des artères coronaires , Diabète de type 2 , Peptide natriurétique cérébral , Fragments peptidiques , Inhibiteurs du cotransporteur sodium-glucose de type 2 , Humains , Mâle , Femelle , Adulte d'âge moyen , Diabète de type 2/complications , Diabète de type 2/traitement médicamenteux , Diabète de type 2/sang , Inhibiteurs du cotransporteur sodium-glucose de type 2/usage thérapeutique , Maladie des artères coronaires/traitement médicamenteux , Maladie des artères coronaires/sang , Maladie des artères coronaires/complications , Sujet âgé , Peptide natriurétique cérébral/sang , Fragments peptidiques/sang , Études de suivi , Débit systolique/effets des médicaments et des substances chimiques , Échocardiographie , Études prospectives , Remodelage ventriculaire/effets des médicaments et des substances chimiques , Composés benzhydryliques/usage thérapeutique , Composés benzhydryliques/pharmacologie , Glucosides
12.
J Am Heart Assoc ; 13(9): e032172, 2024 May 07.
Article de Anglais | MEDLINE | ID: mdl-38700022

RÉSUMÉ

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.


Sujet(s)
Modèles animaux de maladie humaine , Infliximab , Remodelage ventriculaire , Infliximab/usage thérapeutique , Infliximab/pharmacologie , Animaux , Humains , Mâle , Adulte d'âge moyen , Remodelage ventriculaire/effets des médicaments et des substances chimiques , Femelle , Infarctus du myocarde avec sus-décalage du segment ST/traitement médicamenteux , Infarctus du myocarde avec sus-décalage du segment ST/immunologie , Fonction ventriculaire gauche/effets des médicaments et des substances chimiques , Suidae , Sujet âgé , Facteur de nécrose tumorale alpha/métabolisme , Débit systolique/effets des médicaments et des substances chimiques , Thrombose coronarienne/prévention et contrôle , Thrombose coronarienne/traitement médicamenteux , Myocarde/anatomopathologie , Myocarde/métabolisme , Myocarde/immunologie , Troponine I/sang , Troponine I/métabolisme , Macrophages/effets des médicaments et des substances chimiques , Macrophages/immunologie , Macrophages/métabolisme
13.
Expert Opin Investig Drugs ; 33(6): 543-547, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38702878

RÉSUMÉ

INTRODUCTION: There are currently limited effective treatments available to improve lusitropy in patients suffering from heart failure with preserved ejection fraction. The role of PDE9A in diastolic dysfunction has been well-studied over recent years, with a special focus on its association with myocardial hypertrophy. Recent insights into PDE9A inhibition have brought to light the potential for reversal of cardiac remodeling, with multiple studies showing promising results in preclinical data. AREAS COVERED: This expert opinion provides an overview of the role of PDE9A in diastolic heart dysfunction along with the efficacy of PDE9A inhibitors in laboratory models of heart failure with preserved ejection fraction. EXPERT OPINION: The available data on PDE9A inhibition in preclinical studies suggest that there is potential for reversal of diastolic dysfunction and myocardial hypertrophy, however, conflicting data suggests that further studies are required before progressing to clinical trials.


Sujet(s)
3',5'-Cyclic-AMP Phosphodiesterases , Défaillance cardiaque , Inhibiteurs de la phosphodiestérase , Humains , Animaux , Défaillance cardiaque/traitement médicamenteux , Défaillance cardiaque/physiopathologie , Inhibiteurs de la phosphodiestérase/pharmacologie , 3',5'-Cyclic-AMP Phosphodiesterases/antagonistes et inhibiteurs , Cardiomégalie/traitement médicamenteux , Cardiomégalie/physiopathologie , Remodelage ventriculaire/effets des médicaments et des substances chimiques , Débit systolique/effets des médicaments et des substances chimiques
14.
FASEB J ; 38(9): e23654, 2024 May 15.
Article de Anglais | MEDLINE | ID: mdl-38717442

RÉSUMÉ

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.


Sujet(s)
Facteurs de transcription de choc thermique , Metformine , Myocytes cardiaques , Réponse aux protéines mal repliées , Animaux , Mâle , Rats , Angiotensine-II/pharmacologie , Cardiomégalie/métabolisme , Cardiomégalie/traitement médicamenteux , Cardiomégalie/anatomopathologie , Protéines de liaison à l'ADN/métabolisme , Protéines de liaison à l'ADN/génétique , Facteurs de transcription de choc thermique/effets des médicaments et des substances chimiques , Facteurs de transcription de choc thermique/métabolisme , Hypertension artérielle/métabolisme , Hypertension artérielle/traitement médicamenteux , Metformine/pharmacologie , Mitochondries du myocarde/métabolisme , Mitochondries du myocarde/effets des médicaments et des substances chimiques , Myocytes cardiaques/métabolisme , Myocytes cardiaques/effets des médicaments et des substances chimiques , Rats de lignée SHR , Rats de lignée WKY , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Réponse aux protéines mal repliées/effets des médicaments et des substances chimiques , Remodelage ventriculaire/effets des médicaments et des substances chimiques
15.
Int J Cardiol ; 409: 132203, 2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-38795973

RÉSUMÉ

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.


Sujet(s)
Amino-butyrates , Dérivés du biphényle , Modèles animaux de maladie humaine , Association médicamenteuse , Fibrose , Isoprénaline , Tétrazoles , Valsartan , Dysfonction ventriculaire gauche , Remodelage ventriculaire , Animaux , Amino-butyrates/pharmacologie , Dérivés du biphényle/pharmacologie , Souris , Mâle , Remodelage ventriculaire/effets des médicaments et des substances chimiques , Tétrazoles/pharmacologie , Fibrose/induit chimiquement , Dysfonction ventriculaire gauche/induit chimiquement , Dysfonction ventriculaire gauche/physiopathologie , Isoprénaline/toxicité , Souris transgéniques , Mitochondries du myocarde/effets des médicaments et des substances chimiques , Mitochondries du myocarde/métabolisme , Antagonistes des récepteurs aux angiotensines/pharmacologie , Répartition aléatoire
16.
Int Immunopharmacol ; 134: 112143, 2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38692016

RÉSUMÉ

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.


Sujet(s)
Abiétanes , Angiotensine-II , Anti-inflammatoires , Souris de lignée C57BL , Myocytes cardiaques , Remodelage ventriculaire , p38 Mitogen-Activated Protein Kinases , Animaux , Angiotensine-II/métabolisme , p38 Mitogen-Activated Protein Kinases/métabolisme , Mâle , Anti-inflammatoires/pharmacologie , Anti-inflammatoires/usage thérapeutique , Souris , Abiétanes/pharmacologie , Abiétanes/usage thérapeutique , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myocytes cardiaques/métabolisme , Myocytes cardiaques/anatomopathologie , Remodelage ventriculaire/effets des médicaments et des substances chimiques , Inflammation/traitement médicamenteux , Inflammation/métabolisme , Inflammation/induit chimiquement , Phosphorylation/effets des médicaments et des substances chimiques , Cellules cultivées
17.
Sci Rep ; 14(1): 12377, 2024 05 29.
Article de Anglais | MEDLINE | ID: mdl-38811632

RÉSUMÉ

Sacubitril/valsartan has been highly recognized as a treatment for Chronic heart failure (CHF). Its potential cardioprotective benefits and mechanisms, however, remain to be explored. Metabolomics can be used to identify the metabolic characteristics and related markers, as well as the influence of drugs, thereby opening up the new mechanism for sacubitril/valsartan therapy in CHF disease. In this study, the ligation of left anterior descending and exhaustive swimming were used to induce a rat model of CHF after myocardial infarction. The efficacy was appraised with echocardiography, serum NT-proBNP, and histopathologica. UPLC-Q/TOF-MS combined with multivariate statistical analysis approach were used to analyze the effect of sacubitril/valsartan on CHF rats. RT-qPCR and western blot were performed to investigate the tryptophan/kynurenine metabolism pathway. Accordingly, the basal cardiac function were increased, while the serum NT-proBNP and collagen volume fraction decreased in CHF rats with sacubitril/valsartan. Sacubitril/valsartan regulated the expression of kynurenine et.al 8 metabolomic biomarkers in CHF rats serum, and it contributed to the cardioprotective effects through tryptophan metabolism pathway. In addition, the mRNA and protein expression of the indoleamine 2,3-dioxygenase (IDO) in the myocardial tissue of CHF rats, were down-regulated by sacubitril/valsartan, which was the same with the IL-1ß, IFN-γ, TNF-α, COX-2, and IL-6 mRNA expression, and IL-1ß, IFN-γ, and TNF-α expression in serum. In conclusion, sacubitril/valsartan can ameliorate cardiac function and ventricular remodeling in CHF rats, at least in part through inhibition of tryptophan/kynurenine metabolism.


Sujet(s)
Amino-butyrates , Dérivés du biphényle , Association médicamenteuse , Défaillance cardiaque , Inflammation , Cynurénine , Tétrazoles , Tryptophane , Valsartan , Remodelage ventriculaire , Animaux , Amino-butyrates/pharmacologie , Valsartan/pharmacologie , Dérivés du biphényle/pharmacologie , Remodelage ventriculaire/effets des médicaments et des substances chimiques , Cynurénine/métabolisme , Défaillance cardiaque/traitement médicamenteux , Défaillance cardiaque/métabolisme , Rats , Tryptophane/métabolisme , Mâle , Tétrazoles/pharmacologie , Inflammation/traitement médicamenteux , Inflammation/métabolisme , Modèles animaux de maladie humaine , Peptide natriurétique cérébral/métabolisme , Peptide natriurétique cérébral/sang , Rat Sprague-Dawley
18.
Cardiovasc Toxicol ; 24(6): 527-538, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38720122

RÉSUMÉ

Adolescents commonly co-abuse many drugs including anabolic androgenic steroids either they are athletes or non-athletes. Stanozolol is the major anabolic used in recent years and was reported grouped with cannabis. The current study aimed at evaluating the biochemical and histopathological changes related to the hypertrophic effects of stanozolol and/or cannabis whether in condition of exercise practice or sedentary conditions. Adult male Wistar albino rats received either stanozolol (5 mg/kg, s.c), cannabis (10 mg/kg, i.p.), and a combination of both once daily for two months. Swimming exercise protocol was applied as a training model. Relative heart weight, oxidative stress biomarkers, cardiac tissue fibrotic markers were evaluated. Left ventricular morphometric analysis and collagen quantification was done. The combined treatment exhibited serious detrimental effects on the heart tissues. It increased heart tissue fibrotic markers (Masson's trichrome stain (p < 0.001), cardiac COL3 (p < 0.0001), and VEGF-A (p < 0.05)), lowered heart glutathione levels (p < 0.05) and dramatically elevated oxidative stress (increased malondialdehyde (p < 0.0001) and 8-OHDG (p < 0.0001)). Training was not ameliorating for the observed effects. Misuse of cannabis and stanozolol resulted in more hypertrophic consequences of the heart than either drug alone, which were at least largely assigned to oxidative stress, heart tissue fibrotic indicators, histological alterations, and morphometric changes.


Sujet(s)
Anabolisants , Cardiomégalie du sportif , Fibrose , Stress oxydatif , Rat Wistar , Stanozolol , Animaux , Stanozolol/toxicité , Mâle , Stress oxydatif/effets des médicaments et des substances chimiques , Anabolisants/toxicité , Cardiomégalie du sportif/effets des médicaments et des substances chimiques , Hypertrophie ventriculaire gauche/physiopathologie , Hypertrophie ventriculaire gauche/induit chimiquement , Hypertrophie ventriculaire gauche/métabolisme , Hypertrophie ventriculaire gauche/anatomopathologie , Hypertrophie ventriculaire gauche/prévention et contrôle , Remodelage ventriculaire/effets des médicaments et des substances chimiques , Myocarde/anatomopathologie , Myocarde/métabolisme , Dopage sportif , Marqueurs biologiques/métabolisme , Natation , Conditionnement physique d'animal/physiologie , Rats , Modèles animaux de maladie humaine
19.
Int J Mol Med ; 53(6)2024 06.
Article de Anglais | MEDLINE | ID: mdl-38666537

RÉSUMÉ

Fibroblast growth factor (FGF)21 is a peptide hormone that improves mitochondrial function and energy metabolism, and the deficiency of its co­receptor ß­klotho (KLB) causes decreased FGF21 sensitivity. The present study examined whether the cardiac delivery of plasmids containing the KLB gene via ultrasound­targeted microbubble destruction (UTMD) enhances the efficacy of FGF21 against heart failure post­acute myocardial infarction (AMI). For this purpose, the levels of FGF21 in patients and rats with heart dysfunction post­infarction were determined using ELISA. Sprague­Dawley rats received the 3X UTMD­mediated delivery of KLB@cationic microbubbles (KLB@CMBs) 1 week following the induction of AMI. Echocardiography, histopathology and biochemical analysis were performed at 4 weeks following the induction of AMI. The results revealed that patients with heart failure post­infarction had higher serum FGF21 levels than the healthy controls. However, the downstream signal, KLB, but not α­klotho, was reduced in the heart tissues of rats with AMI. As was expected, treatment with FGF21 did not substantially attenuate heart remodeling post­infarction. It was found that decreased receptors KLB in the heart may result in the insensitivity to FGF21 treatment. In vivo, the UTMD technology­mediated delivery of KLB@CMBs to the heart significantly enhanced the effects of FGF21 administration on cardiac remodeling and mitochondrial dysfunction in the rats following infarction. The delivery of KLB to the heart by UTMD and the administration of FGF21 attenuated mitochondrial impairment and oxidative stress by activating nuclear factor erythroid 2­related factor 2 signals. On the whole, the present study demonstrates that the cardiac delivery of KLB significantly optimizes the cardioprotective effects of FGF21 therapy on adverse heart remodeling. UTMD appears a promising interdisciplinary approach with which to improve heart failure post­myocardial infarction.


Sujet(s)
Facteurs de croissance fibroblastique , Protéines Klotho , Microbulles , Infarctus du myocarde , Rat Sprague-Dawley , Remodelage ventriculaire , Facteurs de croissance fibroblastique/administration et posologie , Facteurs de croissance fibroblastique/métabolisme , Facteurs de croissance fibroblastique/pharmacologie , Animaux , Infarctus du myocarde/métabolisme , Infarctus du myocarde/thérapie , Humains , Mâle , Rats , Remodelage ventriculaire/effets des médicaments et des substances chimiques , Femelle , Ondes ultrasonores , Myocarde/métabolisme , Myocarde/anatomopathologie , Défaillance cardiaque/métabolisme , Défaillance cardiaque/thérapie
20.
J Biomed Sci ; 31(1): 42, 2024 Apr 22.
Article de Anglais | MEDLINE | ID: mdl-38650023

RÉSUMÉ

BACKGROUND: Myocarditis substantially increases the risk of ventricular arrhythmia. Approximately 30% of all ventricular arrhythmia cases in patients with myocarditis originate from the right ventricular outflow tract (RVOT). However, the role of NLRP3 signaling in RVOT arrhythmogenesis remains unclear. METHODS: Rats with myosin peptide-induced myocarditis (experimental group) were treated with an NLRP3 inhibitor (MCC950; 10 mg/kg, daily for 14 days) or left untreated. Then, they were subjected to electrocardiography and echocardiography. Ventricular tissue samples were collected from each rat's RVOT, right ventricular apex (RVA), and left ventricle (LV) and examined through conventional microelectrode and histopathologic analyses. In addition, whole-cell patch-clamp recording, confocal fluorescence microscopy, and Western blotting were performed to evaluate ionic currents, intracellular Ca2+ transients, and Ca2+-modulated protein expression in individual myocytes isolated from the RVOTs. RESULTS: The LV ejection fraction was lower and premature ventricular contraction frequency was higher in the experimental group than in the control group (rats not exposed to myosin peptide). Myocarditis increased the infiltration of inflammatory cells into cardiac tissue and upregulated the expression of NLRP3; these observations were more prominent in the RVOT and RVA than in the LV. Furthermore, experimental rats treated with MCC950 (treatment group) improved their LV ejection fraction and reduced the frequency of premature ventricular contraction. Histopathological analysis revealed higher incidence of abnormal automaticity and pacing-induced ventricular tachycardia in the RVOTs of the experimental group than in those of the control and treatment groups. However, the incidences of these conditions in the RVA and LV were similar across the groups. The RVOT myocytes of the experimental group exhibited lower Ca2+ levels in the sarcoplasmic reticulum, smaller intracellular Ca2+ transients, lower L-type Ca2+ currents, larger late Na+ currents, larger Na+-Ca2+ exchanger currents, higher reactive oxygen species levels, and higher Ca2+/calmodulin-dependent protein kinase II levels than did those of the control and treatment groups. CONCLUSION: Myocarditis may increase the rate of RVOT arrhythmogenesis, possibly through electrical and structural remodeling. These changes may be mitigated by inhibiting NLRP3 signaling.


Sujet(s)
Troubles du rythme cardiaque , Myocardite , Protéine-3 de la famille des NLR contenant un domaine pyrine , Transduction du signal , Animaux , Rats , Troubles du rythme cardiaque/physiopathologie , Troubles du rythme cardiaque/étiologie , Troubles du rythme cardiaque/métabolisme , Furanes/pharmacologie , Indènes , Myocardite/métabolisme , Myocardite/physiopathologie , Protéine-3 de la famille des NLR contenant un domaine pyrine/antagonistes et inhibiteurs , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Rat Sprague-Dawley , Sulfonamides/pharmacologie , Remodelage ventriculaire/effets des médicaments et des substances chimiques , Remodelage ventriculaire/physiologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...