Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.136
Filter
Add more filters

Publication year range
1.
FASEB J ; 38(9): e23654, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38717442

ABSTRACT

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.


Subject(s)
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
2.
J Cell Mol Med ; 28(12): e18407, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38894630

ABSTRACT

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.


Subject(s)
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
3.
J Biomed Sci ; 31(1): 42, 2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38650023

ABSTRACT

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.


Subject(s)
Arrhythmias, Cardiac , Myocarditis , NLR Family, Pyrin Domain-Containing 3 Protein , Signal Transduction , Animals , Rats , Arrhythmias, Cardiac/physiopathology , Arrhythmias, Cardiac/etiology , Arrhythmias, Cardiac/metabolism , Furans/pharmacology , Indenes , Myocarditis/metabolism , Myocarditis/physiopathology , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Rats, Sprague-Dawley , Sulfonamides/pharmacology , Ventricular Remodeling/drug effects , Ventricular Remodeling/physiology
4.
Circ Res ; 130(4): 436-454, 2022 02 18.
Article in English | MEDLINE | ID: mdl-35175847

ABSTRACT

Heart failure affects over 2.6 million women and 3.4 million men in the United States with known sex differences in epidemiology, management, response to treatment, and outcomes across a wide spectrum of cardiomyopathies that include peripartum cardiomyopathy, hypertrophic cardiomyopathy, stress cardiomyopathy, cardiac amyloidosis, and sarcoidosis. Some of these sex-specific considerations are driven by the cellular effects of sex hormones on the renin-angiotensin-aldosterone system, endothelial response to injury, vascular aging, and left ventricular remodeling. Other sex differences are perpetuated by implicit bias leading to undertreatment and underrepresentation in clinical trials. The goal of this narrative review is to comprehensively examine the existing literature over the last decade regarding sex differences in various heart failure syndromes from pathophysiological insights to clinical practice.


Subject(s)
Cardiomyopathies/physiopathology , Cardiomyopathies/therapy , Heart Failure/physiopathology , Heart Failure/therapy , Sex Characteristics , Angiotensin Receptor Antagonists/pharmacology , Angiotensin Receptor Antagonists/therapeutic use , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Cardiac Resynchronization Therapy/methods , Cardiomyopathies/blood , Cardiomyopathies/diagnosis , Female , Gonadal Steroid Hormones/blood , Heart Failure/blood , Heart Failure/diagnosis , Humans , Stroke Volume/drug effects , Stroke Volume/physiology , Ventricular Remodeling/drug effects , Ventricular Remodeling/physiology
5.
Diabetes Obes Metab ; 26(6): 2401-2411, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38528818

ABSTRACT

AIMS: To conduct an updated systematic review and meta-analysis to evaluate the efficacy of glucagon-like peptide-1 receptor agonists (GLP-1RAs) with regard to cardiac function and structure in people with or without type 2 diabetes mellitus (T2DM). MATERIALS AND METHODS: We conducted a systematic search using the PubMed, Embase and ClinicalTrials.gov online databases. The primary outcome of interest was changes in mitral inflow E-velocity to tissue Doppler e' velocity (E/e') ratio. Secondary outcomes included other indicators of cardiac reverse remodelling and functional capacity comprising changes in left ventricular mass (LVM), left ventricular global longitudinal strain, left ventricular end-diastolic volume, left ventricular end-systolic volume, left ventricular ejection fraction (LVEF), early to atrial mitral inflow velocity ratio, left atrial volume (LAV), N-terminal pro-B-type natriuretic peptide (NT-proBNP) levels and 6-min walk test (6MWT) results. RESULTS: A total of 15 trials involving 898 patients were included in this analysis. GLP-1RAs significantly improved E/e' ratio (mean difference [MD] = -0.73; 95% confidence interval [CI] -1.34, -0.13), LVM (MD = -3.86 g; 95% CI -7.60, -0.12), LAV (MD = -8.20 mL; 95% CI -12.37, -4.04), NT-proBNP level (standardized MD = -0.27; 95% CI -0.47, -0.06), and 6MWT result (MD = +22.31 m; 95% CI 1.64, 42.99). However, GLP-1RAs had no effect on LVEF (MD = +0.31%; 95% CI -1.02, 1.64). CONCLUSIONS: In this systematic review and meta-analysis, GLP-1RAs were found to have a positive impact on left ventricle diastolic function, hypertrophy, and exercise capacity, but had no effect on systolic function.


Subject(s)
Diabetes Mellitus, Type 2 , Glucagon-Like Peptide-1 Receptor , Humans , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/physiopathology , Glucagon-Like Peptide-1 Receptor/agonists , Hypoglycemic Agents/therapeutic use , Hypoglycemic Agents/pharmacology , Ventricular Function, Left/drug effects , Ventricular Remodeling/drug effects , Stroke Volume/drug effects , Female , Male , Middle Aged , Heart/drug effects , Natriuretic Peptide, Brain/blood , Glucagon-Like Peptide-1 Receptor Agonists
6.
J Cardiovasc Pharmacol ; 83(6): 612-620, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38547510

ABSTRACT

ABSTRACT: Pulmonary arterial hypertension (PAH) is characterized by increased pulmonary vascular resistance (PVR), imposing overload on the right ventricle (RV) and imbalance of the redox state. Our study investigated the influence of treatment with sulforaphane (SFN), found in cruciferous vegetables, on RV remodeling and redox homeostasis in monocrotaline (MCT)-induced PAH. Male Wistar rats were separated into 4 groups: control (CTR); CTR + SFN; MCT; and MCT + SFN. PAH induction was implemented by a single dose of MCT (60 mg/kg intraperitoneally). Treatment with SFN (2.5 mg/kg/day intraperitoneally) started on the seventh day after the MCT injection and persisted for 2 weeks. After 21 days of PAH induction, echocardiographic, hemodynamic, and oxidative stress evaluation was performed. The MCT group showed an increase in RV hypertrophy, RV systolic area, RV systolic, mean pulmonary artery pressure, and PVR and exhibited a decrease in the RV outflow tract acceleration time/ejection time ratio, RV fractional shortening, and tricuspid annular plane systolic excursion compared to CTR ( P < 0.05). SFN-treated PAH attenuated detrimental changes in tricuspid annular plane systolic excursion, mean pulmonary artery pressure, and PVR parameters. Catalase levels and the glutathione/Glutathione disulfide (GSSG) ratio were diminished in the MCT group compared to CTR ( P < 0.05). SFN increased catalase levels and normalized the glutathione/GSSG ratio to control levels ( P < 0.05). Data express the benefit of SFN treatment on the cardiac function of rats with PAH associated with the cellular redox state.


Subject(s)
Disease Models, Animal , Isothiocyanates , Monocrotaline , Oxidation-Reduction , Oxidative Stress , Rats, Wistar , Sulfoxides , Ventricular Function, Right , Animals , Sulfoxides/pharmacology , Isothiocyanates/pharmacology , Male , Ventricular Function, Right/drug effects , Oxidative Stress/drug effects , Antioxidants/pharmacology , Hypertrophy, Right Ventricular/physiopathology , Hypertrophy, Right Ventricular/metabolism , Hypertrophy, Right Ventricular/drug therapy , Homeostasis/drug effects , Ventricular Remodeling/drug effects , Myocardial Contraction/drug effects , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/physiopathology , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/chemically induced , Pulmonary Artery/drug effects , Pulmonary Artery/physiopathology , Pulmonary Artery/metabolism , Rats , Arterial Pressure/drug effects , Pulmonary Arterial Hypertension/drug therapy , Pulmonary Arterial Hypertension/physiopathology , Pulmonary Arterial Hypertension/metabolism , Ventricular Dysfunction, Right/physiopathology , Ventricular Dysfunction, Right/drug therapy , Ventricular Dysfunction, Right/metabolism
7.
J Cardiovasc Pharmacol ; 83(6): 588-601, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38547517

ABSTRACT

ABSTRACT: Chronic kidney disease (CKD) is a significant global health threat that imposes a substantial burden on both individuals and societies. CKD frequently correlates with cardiovascular events, particularly left ventricular hypertrophy (LVH), which contributes to the high mortality rate associated with CKD. Fibroblast growth factor 23 (FGF23), a hormone primarily involved in regulating calcium and phosphorus metabolism, has been identified as a major risk factor for LVH in CKD patients. Elevated serum FGF23 levels are known to induce LVH and myocardial fibrosis by activating the fibroblast growth factor receptor 4 (FGFR4) signal pathway. Therefore, targeting FGFR4 and its downstream signaling pathways holds potential as a treatment strategy for cardiac dysfunction in CKD. In our current study, we have discovered that Hypericin, a key component derived from Hypericum perforatum , has the ability to alleviate CKD-related LVH by targeting the FGFR4/phospholipase C gamma 1 (PLCγ1) signaling pathway. Through in vitro experiments using rat cardiac myocyte H9c2 cells, we observed that Hypericin effectively inhibits FGF23-induced hypertrophy and fibrosis by suppressing the FGFR4/PLCγ1/calcineurin/nuclear factor of activated T-cell (NFAT3) signaling pathway. In addition, our in vivo studies using mice on a high-phosphate diet and rat models of 5/6 nephrectomy demonstrated that Hypericin has therapeutic effects against CKD-induced LVH by modulating the FGFR4/PLCγ1/calcineurin/NFAT3 signaling pathway. In conclusion, our research highlights the potential of Hypericin as a candidate for the treatment of CKD-induced cardiomyopathy. By suppressing the FGFR4/PLCγ1 signaling pathway, Hypericin shows promise in attenuating LVH and myocardial fibrosis associated with CKD.


Subject(s)
Anthracenes , Disease Models, Animal , Fibroblast Growth Factor-23 , Fibroblast Growth Factors , Fibrosis , Hypertrophy, Left Ventricular , Mice, Inbred C57BL , Myocytes, Cardiac , Perylene , Receptor, Fibroblast Growth Factor, Type 4 , Renal Insufficiency, Chronic , Signal Transduction , Animals , Perylene/analogs & derivatives , Perylene/pharmacology , Signal Transduction/drug effects , Fibroblast Growth Factors/metabolism , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/drug therapy , Receptor, Fibroblast Growth Factor, Type 4/metabolism , Hypertrophy, Left Ventricular/physiopathology , Hypertrophy, Left Ventricular/metabolism , Hypertrophy, Left Ventricular/prevention & control , Hypertrophy, Left Ventricular/pathology , Hypertrophy, Left Ventricular/drug therapy , Rats , Male , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Cell Line , Anthracenes/pharmacology , Ventricular Function, Left/drug effects , Ventricular Remodeling/drug effects , Phospholipase C gamma/metabolism , NFATC Transcription Factors/metabolism , Mice
8.
Sleep Breath ; 28(2): 823-833, 2024 May.
Article in English | MEDLINE | ID: mdl-38147288

ABSTRACT

PURPOSE: Circadian disruption has been a common issue due to modern lifestyles. Ventricular remodeling (VR) is a pivotal progressive pathologic change after acute myocardial infarction (AMI) and circadian disruption may have a negative influence on VR according to the latest research. Whether or not Guanxin V (GXV) has a positive effect on VR after AMI with circadian disruption drew our interest. METHODS: Rats were randomly divided into a sham group, an AMI group, an AMI with circadian disruption group, and an AMI with circadian disruption treated with the GXV group according to a random number table. RNA sequencing (RNA-Seq) was utilized to confirm the different expressed genes regulated by circadian disruption. Cardiac function, inflammation factors, pathological evaluation, and mitochondrial dynamics after the intervention were conducted to reveal the mechanism by which GXV regulated VR after AMI with circadian disruption. RESULTS: RNA-Seq demonstrated that NF-κB was up-regulated by circadian disruption in rats with AMI. Functional and pathological evaluation indicated that compared with the AMI group, circadian disruption was associcataed with deteriorated cardiac function, expanded infarcted size, and exacerbated fibrosis and cardiomyocyte apoptosis. Further investigation demonstrated that mitochondrial dynamics imbalance was induced by circadian disruption. GXV intervention reversed the inflammatory status including down-regulation of NF-κB. Reserved cardiac function, limited infarct size, and ameliorated fibrosis and apoptosis were also observed in the GXV treated group. GXV maintained mitochondrial fission/fusion imbalance through suppressed expression of mitochondrial fission-associated proteins. CONCLUSION: The study findings suggest that identified mitochondrial dysfunctions may underlie the link between circadian disruption and VR. GXV may exert cardioprotection after AMI with circadian disruption through regulating mitochondrial dynamics.


Subject(s)
Mitochondrial Dynamics , Myocardial Infarction , Ventricular Remodeling , Animals , Myocardial Infarction/pathology , Myocardial Infarction/drug therapy , Myocardial Infarction/metabolism , Myocardial Infarction/genetics , Myocardial Infarction/physiopathology , Ventricular Remodeling/drug effects , Ventricular Remodeling/physiology , Rats , Mitochondrial Dynamics/drug effects , Male , Rats, Sprague-Dawley , Drugs, Chinese Herbal/pharmacology , Chronobiology Disorders/drug therapy , Chronobiology Disorders/physiopathology , Chronobiology Disorders/genetics , Disease Models, Animal
9.
Int J Mol Sci ; 25(8)2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38673951

ABSTRACT

Succinate dehydrogenase inhibition with malonate during initial reperfusion reduces myocardial infarct size in both isolated mouse hearts subjected to global ischemia and in in situ pig hearts subjected to transient coronary ligature. However, the long-term effects of acute malonate treatment are unknown. Here, we investigated whether the protective effects of succinate dehydrogenase inhibition extend to a reduction in scar size and adverse left ventricular remodeling 28 days after myocardial infarction. Initially, ten wild-type mice were subjected to 45 min of left anterior descending coronary artery (LAD) occlusion, followed by 24 h of reperfusion, and were infused during the first 15 min of reperfusion with saline with or without disodium malonate (10 mg/kg/min, 120 µL/kg/min). Malonate-treated mice depicted a significant reduction in infarct size (15.47 ± 3.40% of area at risk vs. 29.34 ± 4.44% in control animals, p < 0.05), assessed using triphenyltetrazolium chloride. Additional animals were then subjected to a 45 min LAD ligature, followed by 28 days of reperfusion. Treatment with a single dose of malonate during the first 15 min of reperfusion induced a significant reduction in scar area, measured using Picrosirius Red staining (11.94 ± 1.70% of left ventricular area (n = 5) vs. 23.25 ± 2.67% (n = 9), p < 0.05), an effect associated with improved ejection fraction 28 days after infarction, as determined using echocardiography, and an attenuated enhancement in expression of the pro-inflammatory and fibrotic markers NF-κB and Smad2/3 in remote myocardium. In conclusion, a reversible inhibition of succinate dehydrogenase with a single dose of malonate at the onset of reperfusion has long-term protective effects in mice subjected to transient coronary occlusion.


Subject(s)
Malonates , Myocardial Infarction , Myocardial Reperfusion Injury , Succinate Dehydrogenase , Ventricular Remodeling , Animals , Malonates/pharmacology , Myocardial Infarction/drug therapy , Myocardial Infarction/pathology , Mice , Succinate Dehydrogenase/metabolism , Succinate Dehydrogenase/antagonists & inhibitors , Male , Ventricular Remodeling/drug effects , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/pathology , Cicatrix/pathology , Cicatrix/drug therapy , Mice, Inbred C57BL
10.
Int J Mol Sci ; 25(11)2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38892396

ABSTRACT

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.


Subject(s)
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
11.
Circ Res ; 128(10): 1435-1450, 2021 05 14.
Article in English | MEDLINE | ID: mdl-33983832

ABSTRACT

Despite multiple attempts to develop a unifying hypothesis that explains the pathophysiology of heart failure with a reduced ejection fraction (HFrEF), no single conceptual model has withstood the test of time. In the present review, we discuss how the results of recent successful phase III clinical development programs in HFrEF are built upon existing conceptual models for drug development. We will also discuss where recent successes in clinical trials do not fit existing models to identify areas where further refinement of current paradigms may be needed. To provide the necessary structure for this review, we will begin with a brief overview of the pathophysiology of HFrEF, followed by an overview of the current conceptual models for HFrEF, and end with an analysis of the scientific rationale and clinical development programs for 4 new therapeutic classes of drugs that have improved clinical outcomes in HFrEF. The 4 new therapeutic classes discussed are ARNIs, SGLT2 (sodium-glucose cotransporter 2) inhibitors, soluble guanylate cyclase stimulators, and myosin activators. With the exception of SGLT2 inhibitors, each of these therapeutic advances was informed by the insights provided by existing conceptual models of heart failure. Although the quest to determine the mechanism of action of SGLT2 inhibitors is ongoing, this therapeutic class of drugs may represent the most important advance in cardiovascular therapeutics of recent decades and may lead to rethinking or expanding our current conceptual models for HFrEF.


Subject(s)
Heart Failure/drug therapy , Heart Failure/physiopathology , Stroke Volume , Aminobutyrates/therapeutic use , Angiotensin Receptor Antagonists/therapeutic use , Biphenyl Compounds/therapeutic use , Clinical Trials, Phase III as Topic , Drug Combinations , Drug Development , Enzyme Activators/therapeutic use , Heterocyclic Compounds, 2-Ring/therapeutic use , Humans , Models, Biological , Natriuretic Peptides/metabolism , Neprilysin/antagonists & inhibitors , Pyrimidines/therapeutic use , Sodium-Glucose Transporter 2 Inhibitors/therapeutic use , Soluble Guanylyl Cyclase/metabolism , Urea/analogs & derivatives , Urea/therapeutic use , Valsartan/therapeutic use , Ventricular Remodeling/drug effects , Ventricular Remodeling/physiology
13.
Proc Natl Acad Sci U S A ; 117(49): 31353-31364, 2020 12 08.
Article in English | MEDLINE | ID: mdl-33229578

ABSTRACT

Progressive remodeling of the heart, resulting in cardiomyocyte (CM) loss and increased inflammation, fibrosis, and a progressive decrease in cardiac function, are hallmarks of myocardial infarction (MI)-induced heart failure. We show that MCB-613, a potent small molecule stimulator of steroid receptor coactivators (SRCs) attenuates pathological remodeling post-MI. MCB-613 decreases infarct size, apoptosis, hypertrophy, and fibrosis while maintaining significant cardiac function. MCB-613, when given within hours post MI, induces lasting protection from adverse remodeling concomitant with: 1) inhibition of macrophage inflammatory signaling and interleukin 1 (IL-1) signaling, which attenuates the acute inflammatory response, 2) attenuation of fibroblast differentiation, and 3) promotion of Tsc22d3-expressing macrophages-all of which may limit inflammatory damage. SRC stimulation with MCB-613 (and derivatives) is a potential therapeutic approach for inhibiting cardiac dysfunction after MI.


Subject(s)
Cyclohexanones/pharmacology , Myocardial Infarction/physiopathology , Pyridines/pharmacology , Receptors, Steroid/metabolism , Ventricular Remodeling/drug effects , Animals , Cell Differentiation/drug effects , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Fibrosis , Heart Function Tests , Inflammation/pathology , Macrophages/drug effects , Macrophages/pathology , Mice , Myocardial Infarction/genetics , Myocardial Infarction/pathology , RAW 264.7 Cells , RNA/genetics , RNA/metabolism , Transcription, Genetic/drug effects
14.
Pharm Biol ; 61(1): 963-972, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37357417

ABSTRACT

CONTEXT: Punicalagin has myocardial protection; the mechanism of punicalagin on ventricular remodeling (VR) after acute myocardial infarction (AMI) remains unclear. OBJECTIVE: These studies explore the role and mechanism of punicalagin in preventing and treating VR after AMI. MATERIALS AND METHODS: Molecular docking was used to predict the targets of punicalagin. After 2 weeks of AMI model, the SD rats were randomly divided into model, and punicalagin (200, 400 mg/kg, gavage) groups for 4 weeks. Thoracotomy with perforation but no ligature was performed on rats in control group. The protein expression of nucleotide-binding oligomerization domain-like receptor family pyrin domain-containing 3 (NLRP3), apoptosis speck-like protein (ASC), caspase-1, gasdermin D (GSDMD), and GSDMD-N, the mRNA expression of NLRP3, caspase-1, GSDMD, interleukin-1ß (IL-1ß) and IL-18 were evaluated. RESULTS: Punicalagin had binding activities with NLRP3 (Vina score, -5.8), caspase-1 (Vina score, -6.7), and GSDMD (Vina score, -6.7). Punicalagin could improve cardiac function, alleviate cardiac pathological changes, minimize the excessive accumulation of collagen in the left ventricular myocardium (p < 0.01), and inhibit cardiomyocyte apoptosis (p < 0.01). Furthermore, punicalagin could inhibit the overexpression of NLRP3, caspase-1, and GSDMD via immunohistochemistry (p < 0.01). Punicalagin inhibited the protein levels of NLRP3, caspase-1, ASC, GSDMD, and GSDMD-N (p < 0.05, p < 0.01). Punicalagin reduced the mRNA expression of NLRP3, caspase-1, GSDMD, IL-1ß and IL-18 (p < 0.05, p < 0.01). CONCLUSIONS: Punicalagin may provide a useful treatment for the future myocardial protection.


Subject(s)
Hydrolyzable Tannins , Myocardial Infarction , Signal Transduction , Ventricular Remodeling , Hydrolyzable Tannins/administration & dosage , Animals , Rats , Ventricular Remodeling/drug effects , Myocardial Infarction/drug therapy , Myocardial Infarction/pathology , Signal Transduction/drug effects , Male , Rats, Sprague-Dawley , Molecular Docking Simulation , Fibrosis/drug therapy , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Apoptosis/drug effects , Caspase 1/metabolism
15.
J Cell Mol Med ; 26(2): 312-325, 2022 01.
Article in English | MEDLINE | ID: mdl-34845819

ABSTRACT

Persistent cardiac Ca2+ /calmodulin-dependent Kinase II (CaMKII) activation was considered to promote heart failure (HF) development, some studies believed that CaMKII was a target for therapy of HF. However, CaMKII was an important mediator for the ischaemia-induced coronary angiogenesis, and new evidence confirmed that angiogenesis inhibited cardiac remodelling and improved heart function, and some conditions which impaired angiogenesis aggravated ventricular remodelling. This study aimed to investigate the roles and the underlying mechanisms of CaMKII inhibitor in cardiac remodelling. First, we induced cardiac remodelling rat model by ISO, pre-treated by CaMKII inhibitor KN-93, evaluated heart function by echocardiography measurements, and performed HE staining, Masson staining, Tunel staining, Western blot and RT-PCR to test cardiac remodelling and myocardial microvessel density; we also observed ultrastructure of cardiac tissue with transmission electron microscope. Second, we cultured HUVECs, pre-treated by ISO and KN-93, detected cell proliferation, migration, tubule formation and apoptosis, and carried out Western blot to determine the expression of NOX2, NOX4, VEGF, VEGFR2, p-VEGFR2 and STAT3; mtROS level was also measured. In vivo, we found KN-93 severely reduced myocardial microvessel density, caused apoptosis of vascular endothelial cells, enhanced cardiac hypertrophy, myocardial apoptosis, collagen deposition, aggravated the deterioration of myocardial ultrastructure and heart function. In vitro, KN-93 inhibited HUVECs proliferation, migration and tubule formation, and promoted apoptosis of HUVECs. The expression of NOX2, NOX4, p-VEGFR2 and STAT3 were down-regulated by KN-93; mtROS level was severely reduced by KN-93. We concluded that KN-93 impaired angiogenesis and aggravated cardiac remodelling and heart failure via inhibiting NOX2/mtROS/p-VEGFR2 and STAT3 pathways.


Subject(s)
Benzylamines , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Heart Failure , Sulfonamides , Ventricular Remodeling , Animals , Benzylamines/adverse effects , Benzylamines/pharmacology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Endothelial Cells/metabolism , Myocytes, Cardiac/metabolism , NADPH Oxidase 2 , Neovascularization, Physiologic/drug effects , Rats , STAT3 Transcription Factor/metabolism , Sulfonamides/adverse effects , Sulfonamides/pharmacology , Vascular Endothelial Growth Factor Receptor-2/metabolism , Ventricular Remodeling/drug effects
16.
Circulation ; 144(11): 859-869, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34420373

ABSTRACT

BACKGROUND: Inflammation is a key factor of myocardial damage in reperfused ST-segment-elevation myocardial infarction. We hypothesized that colchicine, a potent anti-inflammatory agent, may reduce infarct size (IS) and left ventricular (LV) remodeling at the acute phase of ST-segment-elevation myocardial infarction. METHODS: In this double-blind multicenter trial, we randomly assigned patients admitted for a first episode of ST-segment-elevation myocardial infarction referred for primary percutaneous coronary intervention to receive oral colchicine (2-mg loading dose followed by 0.5 mg twice a day) or matching placebo from admission to day 5. The primary efficacy outcome was IS determined by cardiac magnetic resonance imaging at 5 days. The relative LV end-diastolic volume change at 3 months and IS at 3 months assessed by cardiac magnetic resonance imaging were among the secondary outcomes. RESULTS: We enrolled 192 patients, 101 in the colchicine group and 91 in the control group. At 5 days, the gadolinium enhancement-defined IS did not differ between the colchicine and placebo groups with a mean of 26 interquartile range (IQR) [16-44] versus 28.4 IQR [14-40] g of LV mass, respectively (P=0.87). At 3 months follow-up, there was no significant difference in LV remodeling between the colchicine and placebo groups with a +2.4% (IQR, -8.3% to 11.1%) versus -1.1% (IQR, -8.0% to 9.9%) change in LV end-diastolic volume (P=0.49). Infarct size at 3 months was also not significantly different between the colchicine and placebo groups (17 IQR [10-28] versus 18 IQR [10-27] g of LV mass, respectively; P=0.92). The incidence of gastrointestinal adverse events during the treatment period was greater with colchicine than with placebo (34% versus 11%, respectively; P=0.0002). CONCLUSIONS: In this randomized, placebo-controlled trial, oral administration of high-dose colchicine at the time of reperfusion and for 5 days did not reduce IS assessed by cardiac magnetic resonance imaging. Registration: URL: https://www.clinicaltrials.gov; Unique identifier: NCT03156816.


Subject(s)
Colchicine/therapeutic use , Myocardial Infarction/drug therapy , ST Elevation Myocardial Infarction/drug therapy , Ventricular Remodeling/drug effects , Acute Disease , Adult , Aged , Contrast Media/pharmacology , Female , Heart/drug effects , Hospitalization , Humans , Male , Middle Aged , Myocardium/pathology , Referral and Consultation
17.
Circulation ; 144(3): 199-209, 2021 07 20.
Article in English | MEDLINE | ID: mdl-33983794

ABSTRACT

BACKGROUND: Patients with left ventricular (LV) systolic dysfunction after myocardial infarction are at a high risk of developing heart failure. The addition of neprilysin inhibition to renin angiotensin system inhibition may result in greater attenuation of adverse LV remodeling as a result of increased levels of substrates for neprilysin with vasodilatory, antihypertrophic, antifibrotic, and sympatholytic effects. METHODS: We performed a prospective, multicenter, randomized, double-blind, active-comparator trial comparing sacubitril/valsartan 97/103 mg twice daily with valsartan 160 mg twice daily in patients ≥3 months after myocardial infarction with a LV ejection fraction ≤40% who were taking a renin angiotensin system inhibitor (equivalent dose of ramipril ≥2.5 mg twice daily) and a ß-blocker unless contraindicated or intolerant. Patients in New York Heart Association class ≥II or with signs and symptoms of heart failure were excluded. The primary outcome was change from baseline to 52 weeks in LV end-systolic volume index measured using cardiac magnetic resonance imaging. Secondary outcomes included other magnetic resonance imaging measurements of LV remodeling, change in NT-proBNP (N-terminal pro-B-type natriuretic peptide) and high-sensitivity cardiac troponin I, and a patient global assessment of change questionnaire. RESULTS: From July 2018 to June 2019, we randomized 93 patients with the following characteristics: mean age, 60.7±10.4 years; median time from myocardial infarction, 3.6 years (interquartile range, 1.2-7.2); mean LV ejection fraction, 36.8%±7.1%; and median NT-proBNP, 230 pg/mL (interquartile range, 124-404). Sacubitril/valsartan, compared with valsartan, did not significantly reduce LV end-systolic volume index; adjusted between-group difference, -1.9 mL/m2 (95% CI, -4.9 to 1.0); P=0.19. There were no significant between-group differences in NT-proBNP, high-sensitivity cardiac troponin I, LV end-diastolic volume index, left atrial volume index, LV ejection fraction, LV mass index, or patient global assessment of change. CONCLUSIONS: In patients with asymptomatic LV systolic dysfunction late after myocardial infarction, treatment with sacubitril/valsartan did not have a significant reverse remodeling effect compared with valsartan. Registration: URL: https://www.clinicaltrials.gov; Unique identifier: NCT03552575.


Subject(s)
Myocardial Infarction/complications , Neprilysin/antagonists & inhibitors , Ventricular Dysfunction, Left/diagnosis , Ventricular Dysfunction, Left/etiology , Ventricular Remodeling/drug effects , Aged , Aminobutyrates/administration & dosage , Asymptomatic Diseases , Biomarkers , Biphenyl Compounds/administration & dosage , Disease Susceptibility , Drug Combinations , Female , Follow-Up Studies , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Myocardial Infarction/drug therapy , Randomized Controlled Trials as Topic , Stroke Volume/drug effects , Treatment Outcome , Valsartan/administration & dosage , Ventricular Dysfunction, Left/drug therapy
18.
J Transl Med ; 20(1): 305, 2022 07 06.
Article in English | MEDLINE | ID: mdl-35794561

ABSTRACT

BACKGROUND: Increasing evidence indicates that myocardial oxidative injury plays a crucial role in the pathophysiology of cardiac hypertrophy (CH) and heart failure (HF). The active component of rhubarb, rhein exerts significant actions on oxidative stress and inflammation. Nonetheless, its role in cardiac remodeling remains unclear. METHODS: CH was induced by angiotensin II (Ang II, 1.4 mg/kg/d for 4 weeks) in male C57BL/6 J mice. Then, rhein (50 and 100 mg/kg) was injected intraperitoneally for 28 days. CH, fibrosis, oxidative stress, and cardiac function in the mice were examined. In vitro, neonatal rat cardiomyocytes (CMs) and cardiac fibroblasts (CFs) pre-treated with rhein (5 and 25 µM) were challenged with Ang II. We performed RNA sequencing to determine the mechanistic role of rhein in the heart. RESULTS: Rhein significantly suppressed Ang II-induced CH, fibrosis, and reactive oxygen species production and improved cardiac systolic dysfunction in vivo. In vitro, rhein significantly attenuated Ang II-induced CM hypertrophy and CF collagen expression. In addition, rhein obviously alleviated the increased production of superoxide induced by Ang II. Mechanistically, rhein inhibited FGF23 expression significantly. Furthermore, FGF23 overexpression abolished the protective effects of rhein on CMs, CFs, and cardiac remodeling. Rhein reduced FGF23 expression, mostly through the activation of AMPK (AMP-activated protein kinase). AMPK activity inhibition suppressed Ang II-induced CM hypertrophy and CF phenotypic transformation. CONCLUSION: Rhein inhibited Ang II-induced CH, fibrosis, and oxidative stress during cardiac remodeling through the AMPK-FGF23 axis. These findings suggested that rhein could serve as a potential therapy in cardiac remodeling and HF.


Subject(s)
AMP-Activated Protein Kinases , Angiotensin II , Anthraquinones , Fibroblast Growth Factor-23 , Heart Failure , Ventricular Remodeling , AMP-Activated Protein Kinases/metabolism , Angiotensin II/metabolism , Animals , Anthraquinones/pharmacology , Fibroblast Growth Factor-23/metabolism , Fibrosis , Heart Failure/drug therapy , Heart Failure/metabolism , Hypertrophy , Male , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Rats , Ventricular Remodeling/drug effects
19.
Microvasc Res ; 139: 104268, 2022 01.
Article in English | MEDLINE | ID: mdl-34728225

ABSTRACT

Neutrophils oscillate in number and phenotype after being released from bone marrow. Myocardial infarction (MI) outcome is associated with the time-of-day of ischemia onset. However, the underlying contributive factors of neutrophils to cardiac remodeling post MI remain unknown. We examined neutrophil infiltration into the heart and cardiac function and remodeling in C57BL/6J MI model created by permanent coronary ligation at different zeitgeber times (ZT). We found that cell surface markers (CD62L, CXCR2, CXCR4) of neutrophils in peripheral blood lost diurnal oscillation 24 h post MI. Meanwhile, circadian gene Bmal1, Nr1d1, and Clock mRNA expression displayed disrupted diurnal patterns. Flow cytometry showed augmented aged neutrophil (CD11b+Ly6G+CD62Llow) infiltration into the heart along with increased circulating aged neutrophils in MI groups with more infiltration at ZT5 (p < 0.05), but no difference for aged neutrophil infiltration at different ZT points in late stage. Infiltrated neutrophils had significantly higher CXCL2 and CXCR2 but lower CXCR4 gene expression (p < 0.05). Mice that underwent ligation at ZT5 had high mortality rate and large infarct size. Echocardiography showed that those mice had significantly larger end diastolic and systolic volume and lower ejection fraction (p < 0.05). Immunohistology revealed that those mice displayed more fibrosis, cardiomyocyte hypertrophy, and less angiogenesis compared to ZT13 or ZT21 group (p < 0.05). However, treatment with anti-CXCL2 antibody significantly reduced LV dilatation, fibrosis, hypertrophy and improved cardiac function. These results indicate greater aged neutrophil infiltration into the heart contributes to cardiac hypertrophy, fibrosis, and dysfunction which suggests that blocking neutrophil aging may be a therapeutic alternative following acute myocardial infarction.


Subject(s)
Circadian Rhythm , Myocardial Infarction/immunology , Myocardium/immunology , Neutrophil Infiltration , Neutrophils/immunology , Ventricular Function, Left , Ventricular Remodeling , Animals , Antibodies/pharmacology , Chemokine CXCL2/antagonists & inhibitors , Chemokine CXCL2/metabolism , Disease Models, Animal , Fibrosis , Male , Mice, Inbred C57BL , Myocardial Infarction/drug therapy , Myocardial Infarction/metabolism , Myocardial Infarction/physiopathology , Myocardium/metabolism , Myocardium/pathology , Neutrophil Infiltration/drug effects , Neutrophils/drug effects , Neutrophils/metabolism , Receptors, CXCR4/metabolism , Receptors, Interleukin-8B/metabolism , Time Factors , Ventricular Remodeling/drug effects
20.
Circ Res ; 126(8): 1007-1023, 2020 04 10.
Article in English | MEDLINE | ID: mdl-32098592

ABSTRACT

RATIONALE: Excessive Ang II (angiotensin II) levels lead to a profibrotic and hypertrophic milieu that produces deleterious remodeling and dysfunction in hypertension-associated heart failure. Agents that disrupt Ang II-induced cardiac dysfunction may have clinical utility in the treatment of hypertension-associated heart failure. OBJECTIVE: We have examined the potential effect of celastrol-a bioactive compound derived from the Celastraceae family-on Ang II-induced cardiac dysfunction. METHODS AND RESULTS: In rat primary cardiomyocytes and H9C2 (rat cardiomyocyte-like H9C2) cells, celastrol attenuates Ang II-induced cellular hypertrophy and fibrotic responses. Proteome microarrays, surface plasmon resonance, competitive binding assays, and molecular simulation were used to identify the molecular target of celastrol. Our data showed that celastrol directly binds to and inhibits STAT (signal transducer and activator of transcription)-3 phosphorylation and nuclear translocation. Functional tests demonstrated that the protection of celastrol is afforded through targeting STAT3. Overexpression of STAT3 dampens the effect of celastrol by partially rescuing STAT3 activity. Finally, we investigated the in vivo effect of celastrol treatment in mice challenged with Ang II and in the transverse aortic constriction model. We show that celastrol administration protected heart function in Ang II-challenged and transverse aortic constriction-challenged mice by inhibiting cardiac fibrosis and hypertrophy. CONCLUSIONS: Our studies show that celastrol inhibits Ang II-induced cardiac dysfunction by inhibiting STAT3 activity.


Subject(s)
Angiotensin II/toxicity , Drug Delivery Systems/methods , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/metabolism , Triterpenes/administration & dosage , Ventricular Remodeling/drug effects , Animals , Cell Line , HEK293 Cells , Humans , Mice , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Pentacyclic Triterpenes , Protein Structure, Tertiary , Random Allocation , Rats , STAT3 Transcription Factor/chemistry , Tripterygium , Triterpenes/chemistry , Ventricular Remodeling/physiology
SELECTION OF CITATIONS
SEARCH DETAIL