Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.641
Filter
3.
Nat Commun ; 15(1): 6058, 2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39025877

ABSTRACT

Heart failure causes considerable morbidity and mortality worldwide. Clinically applied drugs for the treatment of heart failure are still severely limited by poor delivery efficiency to the heart and off-target consumption. Inspired by the high heart delivery efficiency of inhaled drugs, we present an inhalable cardiac-targeting peptide (CTP)-modified calcium phosphate (CaP) nanoparticle for the delivery of TP-10, a selective inhibitor of PDE10A. The CTP modification significantly promotes cardiomyocyte and fibroblast targeting during the pathological state of heart failure in male mice. TP-10 is subsequently released from TP-10@CaP-CTP and effectively attenuates cardiac remodelling and improved cardiac function. In view of these results, a low dosage (2.5 mg/kg/2 days) of inhaled medication exerted good therapeutic effects without causing severe lung injury after long-term treatment. In addition, the mechanism underlying the amelioration of heart failure is investigated, and the results reveal that the therapeutic effects of this system on cardiomyocytes and cardiac fibroblasts are mainly mediated through the cAMP/AMPK and cGMP/PKG signalling pathways. By demonstrating the targeting capacity of CTP and verifying the biosafety of inhalable CaP nanoparticles in the lung, this work provides a perspective for exploring myocardium-targeted therapy and presents a promising clinical strategy for the long-term management of heart failure.


Subject(s)
Heart Failure , Myocytes, Cardiac , Nanomedicine , Nanoparticles , Animals , Male , Heart Failure/drug therapy , Heart Failure/prevention & control , Mice , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Administration, Inhalation , Nanoparticles/chemistry , Nanomedicine/methods , Peptides/pharmacology , Peptides/administration & dosage , Myocardium/metabolism , Myocardium/pathology , Fibroblasts/drug effects , Fibroblasts/metabolism , Mice, Inbred C57BL , Signal Transduction/drug effects , Cyclic GMP/metabolism , Lung/drug effects , Lung/pathology , Lung/metabolism , Disease Models, Animal , Calcium Phosphates
4.
Cardiovasc Diabetol ; 23(1): 269, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39044275

ABSTRACT

Heart failure with preserved ejection fraction (HFpEF) is a mortal clinical syndrome without effective therapies. Empagliflozin (EMPA) improves cardiovascular outcomes in HFpEF patients, but the underlying mechanism remains elusive. Here, mice were fed a high-fat diet (HFD) supplemented with L-NAME for 12 weeks and subsequently intraperitoneally injected with EMPA for another 4 weeks. A 4D-DIA proteomic assay was performed to detect protein changes in the failing hearts. We identified 310 differentially expressed proteins (DEPs) (ctrl vs. HFpEF group) and 173 DEPs (HFpEF vs. EMPA group). The regulation of immune system processes was enriched in all groups and the interferon response genes (STAT1, Ifit1, Ifi35 and Ifi47) were upregulated in HFpEF mice but downregulated after EMPA administration. In addition, EMPA treatment suppressed the increase in the levels of aging markers (p16 and p21) in HFpEF hearts. Further bioinformatics analysis verified STAT1 as the hub transcription factor during pathological changes in HFpEF mice. We next treated H9C2 cells with IFN-γ, a primary agonist of STAT1 phosphorylation, to investigate whether EMPA plays a beneficial role by blocking STAT1 activation. Our results showed that IFN-γ treatment caused cardiomyocyte senescence and STAT1 activation, which were inhibited by EMPA administration. Notably, STAT1 inhibition significantly reduced cellular senescence possibly by regulating STING expression. Our findings revealed that EMPA mitigates cardiac inflammation and aging in HFpEF mice by inhibiting STAT1 activation. The STAT1-STING axis may act as a pivotal mechanism in the pathogenesis of HFpEF, especially under inflammatory and aging conditions.


Subject(s)
Benzhydryl Compounds , Cellular Senescence , Disease Models, Animal , Glucosides , Heart Failure , Membrane Proteins , Mice, Inbred C57BL , Myocytes, Cardiac , STAT1 Transcription Factor , Signal Transduction , Sodium-Glucose Transporter 2 Inhibitors , Stroke Volume , Ventricular Function, Left , Animals , STAT1 Transcription Factor/metabolism , Heart Failure/metabolism , Heart Failure/physiopathology , Heart Failure/prevention & control , Heart Failure/drug therapy , Heart Failure/pathology , Benzhydryl Compounds/pharmacology , Glucosides/pharmacology , Cellular Senescence/drug effects , Signal Transduction/drug effects , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Male , Stroke Volume/drug effects , Ventricular Function, Left/drug effects , Membrane Proteins/metabolism , Membrane Proteins/genetics , Cell Line , Interferon-gamma/metabolism , Phosphorylation , Mice
5.
Basic Res Cardiol ; 119(4): 691-697, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38864895

ABSTRACT

The mitochondrial metabolite succinate is a key driver of ischemia/reperfusion injury (IRI). Targeting succinate metabolism by inhibiting succinate dehydrogenase (SDH) upon reperfusion using malonate is an effective therapeutic strategy to achieve cardioprotection in the short term (< 24 h reperfusion) in mouse and pig in vivo myocardial infarction (MI) models. We aimed to assess whether inhibiting IRI with malonate given upon reperfusion could prevent post-MI heart failure (HF) assessed after 28 days. Male C57BL/6 J mice were subjected to 30 min left anterior coronary artery (LAD) occlusion, before reperfusion for 28 days. Malonate or without-malonate control was infused as a single dose upon reperfusion. Cardiac function was assessed by echocardiography and fibrosis by Masson's trichrome staining. Reperfusion without malonate significantly reduced ejection fraction (~ 47%), fractional shortening (~ 23%) and elevated collagen deposition 28 days post-MI. Malonate, administered as a single infusion (16 mg/kg/min for 10 min) upon reperfusion, gave a significant cardioprotective effect, with ejection fraction (~ 60%) and fractional shortening (~ 30%) preserved and less collagen deposition. Using an acidified malonate formulation, to enhance its uptake into cardiomyocytes via the monocarboxylate transporter 1, both 1.6 and 16 mg/kg/min 10 min infusion led to robust long-term cardioprotection with preserved ejection fraction (> 60%) and fractional shortening (~ 30%), as well as significantly less collagen deposition than control hearts. Malonate administration upon reperfusion prevents post-MI HF. Acidification of malonate enables lower doses of malonate to also achieve long-term cardioprotection post-MI. Therefore, the administration of acidified malonate upon reperfusion is a promising therapeutic strategy to prevent IRI and post-MI HF.


Subject(s)
Disease Models, Animal , Heart Failure , Malonates , Mice, Inbred C57BL , Myocardial Infarction , Myocardial Reperfusion Injury , Animals , Malonates/pharmacology , Male , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/prevention & control , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/prevention & control , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Heart Failure/prevention & control , Heart Failure/metabolism , Heart Failure/drug therapy , Heart Failure/physiopathology , Heart Failure/etiology , Mice , Myocardium/metabolism , Myocardium/pathology , Ventricular Function, Left/drug effects , Fibrosis , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Time Factors
6.
Circ Res ; 135(2): 280-297, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38847080

ABSTRACT

BACKGROUND: Heart failure (HF) is one of the leading causes of mortality worldwide. Extracellular vesicles, including small extracellular vesicles or exosomes, and their molecular cargo are known to modulate cell-to-cell communication during multiple cardiac diseases. However, the role of systemic extracellular vesicle biogenesis inhibition in HF models is not well documented and remains unclear. METHODS: We investigated the role of circulating exosomes during cardiac dysfunction and remodeling in a mouse transverse aortic constriction (TAC) model of HF. Importantly, we investigate the efficacy of tipifarnib, a recently identified exosome biogenesis inhibitor that targets the critical proteins (Rab27a [Ras associated binding protein 27a], nSMase2 [neutral sphingomyelinase 2], and Alix [ALG-2-interacting protein X]) involved in exosome biogenesis for this mouse model of HF. In this study, 10-week-old male mice underwent TAC surgery were randomly assigned to groups with and without tipifarnib treatment (10 mg/kg 3 times/wk) and monitored for 8 weeks, and a comprehensive assessment was conducted through performed echocardiographic, histological, and biochemical studies. RESULTS: TAC significantly elevated circulating plasma exosomes and markedly increased cardiac left ventricular dysfunction, cardiac hypertrophy, and fibrosis. Furthermore, injection of plasma exosomes from TAC mice induced left ventricular dysfunction and cardiomyocyte hypertrophy in uninjured mice without TAC. On the contrary, treatment of tipifarnib in TAC mice reduced circulating exosomes to baseline and remarkably improved left ventricular functions, hypertrophy, and fibrosis. Tipifarnib treatment also drastically altered the miRNA profile of circulating post-TAC exosomes, including miR 331-5p, which was highly downregulated both in TAC circulating exosomes and in TAC cardiac tissue. Mechanistically, miR 331-5p is crucial for inhibiting the fibroblast-to-myofibroblast transition by targeting HOXC8, a critical regulator of fibrosis. Tipifarnib treatment in TAC mice upregulated the expression of miR 331-5p that acts as a potent repressor for one of the fibrotic mechanisms mediated by HOXC8. CONCLUSIONS: Our study underscores the pathological role of exosomes in HF and fibrosis in response to pressure overload. Tipifarnib-mediated inhibition of exosome biogenesis and cargo sorting may serve as a viable strategy to prevent progressive cardiac remodeling in HF.


Subject(s)
Extracellular Vesicles , Heart Failure , Quinolones , Animals , Male , Mice , Cardiotonic Agents/pharmacology , Cardiotonic Agents/therapeutic use , Disease Models, Animal , Extracellular Vesicles/drug effects , Heart Failure/pathology , Heart Failure/prevention & control , Quinolones/pharmacology , Quinolones/therapeutic use , Random Allocation , Up-Regulation/drug effects , MicroRNAs , Myofibroblasts/drug effects , Myofibroblasts/metabolism
7.
JACC Heart Fail ; 12(7): 1157-1165, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38878010

ABSTRACT

As a result of the widespread use of reperfusion therapies and secondary prevention over the last 30 years, there has been a dramatic reduction in the risk of mortality and development of heart failure (HF) following acute myocardial infarction (MI). Despite this, the development of chronic HF remains a common occurrence in the days, months, and years following MI. Neurohormonal inhibition remains the mainstay of pharmacologic prevention of HF following MI, with recent trials showing an additive benefit of a neprilysin inhibitor or a sodium glucose co-transporter 2 inhibitor in reducing the risk of development of HF but no significant effect on mortality. Novel imaging tools may help refine risk stratification in high-risk patients and allow greater targeting of preventative therapies in patients most likely to benefit. Research is ongoing into novel therapies aiming to minimize the degree of myocardial damage and prevention of progressive adverse remodeling following MI.


Subject(s)
Heart Failure , Myocardial Infarction , Humans , Heart Failure/prevention & control , Myocardial Infarction/complications , Myocardial Infarction/prevention & control , Secondary Prevention/methods , Sodium-Glucose Transporter 2 Inhibitors/therapeutic use
9.
Toxicol Appl Pharmacol ; 487: 116957, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38735590

ABSTRACT

Heart failure is associated with histone deacetylase (HDAC) regulation of gene expression, the inhibition of which is thought to be beneficial for heart failure therapy. Here, we explored the cardioprotective effects and underlying mechanism of a novel selenium-containing HDAC inhibitor, Se-SAHA, on isoproterenol (ISO)-induced heart failure. We found that pretreatment with Se-SAHA attenuated ISO-induced cardiac hypertrophy and fibrosis in neonatal rat ventricular myocytes (NRVMs). Se-SAHA significantly attenuated the generation of ISO-induced reactive oxygen species (ROS) and restored the expression levels of superoxide dismutase 2 (SOD2) and heme oxygenase-1 (HO-1) in vitro. Furthermore, Se-SAHA pretreatment prevented the accumulation of autophagosomes. Se-SAHA reversed the high expression of HDAC1 and HDAC6 induced by ISO incubation. However, after the addition of the HDAC agonist, the effect of Se-SAHA on blocking autophagy was inhibited. Using ISO-induced mouse models, cardiac ventricular contractile dysfunction, hypertrophy, and fibrosis was reduced treated by Se-SAHA. In addition, Se-SAHA inhibited HDAC1 and HDAC6 overexpression in ISO-treated mice. Se-SAHA treatment significantly increased the activity of SOD2 and improved the ability to eliminate free radicals. Se-SAHA hindered the excessive levels of the microtubule-associated protein 1 light chain 3 (LC3)-II and Beclin-1 in heart failure mice. Collectively, our results indicate that Se-SAHA exerts cardio-protection against ISO-induced heart failure via antioxidative stress and autophagy inhibition.


Subject(s)
Autophagy , Heart Failure , Histone Deacetylase Inhibitors , Isoproterenol , Mice, Inbred C57BL , Myocytes, Cardiac , Oxidative Stress , Rats, Sprague-Dawley , Animals , Isoproterenol/toxicity , Heart Failure/chemically induced , Heart Failure/prevention & control , Heart Failure/pathology , Heart Failure/drug therapy , Autophagy/drug effects , Histone Deacetylase Inhibitors/pharmacology , Oxidative Stress/drug effects , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Myocytes, Cardiac/metabolism , Male , Rats , Mice , Superoxide Dismutase/metabolism , Reactive Oxygen Species/metabolism , Antioxidants/pharmacology , Fibrosis , Cells, Cultured , Cardiomegaly/chemically induced , Cardiomegaly/prevention & control , Cardiomegaly/pathology
10.
Nat Med ; 30(8): 2337-2342, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38749475

ABSTRACT

Comparative evidence for the effects of bariatric metabolic surgery (BMS) and glucagon-like peptide-1 receptor agonists (GLP-1RA) on cardiovascular outcomes is limited. Here, in an observational, retrospective cohort study, we compared the incidence of congestive heart failure (CHF) in adults living with obesity and diabetes without history of CHF (primary CHF) treated with BMS versus GLP-1RA. The population cohort comprised members of Clalit Health Services with no prior history of ischemic heart disease, ischemic stroke or CHF. During the time period of 2008-2021, patients who underwent their first BMS were matched 1:1 with patients who initiated treatment with GLP-1RA, based on clinical characteristics. The study included 2,205 matched pairs of patients (64.5% female), followed for a median of 6.6 years and up to 12 years. Primary incidence of CHF occurred in 26 (1.2%) patients treated with BMS and in 90 patients treated with GLP-1RA (4.1%) (adjusted hazard ratio 0.43, 95% confidence interval 0.27-0.68). Further adjustment for weight reduction did not significantly diminish this association (hazard ratio adjusted for weight reduction 0.48, 95% confidence interval 0.28-0.82), indicating that the differential effect was not mediated through the relative advantage of BMS in maximal weight reduction. In this study, BMS was associated with a stronger reduction in primary incidence of CHF compared with treatment with GLP-1RA. With the increasing use of highly potent next-generation GLP-1RAs, further comparative long-term studies are warranted.


Subject(s)
Bariatric Surgery , Glucagon-Like Peptide-1 Receptor , Heart Failure , Humans , Heart Failure/epidemiology , Heart Failure/prevention & control , Female , Male , Glucagon-Like Peptide-1 Receptor/agonists , Middle Aged , Retrospective Studies , Adult , Obesity/surgery , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/complications , Treatment Outcome , Incidence , Aged , Glucagon-Like Peptide-1 Receptor Agonists
11.
Biochem Pharmacol ; 225: 116305, 2024 07.
Article in English | MEDLINE | ID: mdl-38768763

ABSTRACT

Heart failure (HF) prevalence is rising due to reduced early mortality and demographic change. Relaxin (RLN) mediates protective effects in the cardiovascular system through Relaxin-receptor 1 (RXFP1). Cardiac overexpression of RXFP1 with additional RLN supplementation attenuated HF in the pressure-overload transverse aortic constriction (TAC) model. Here, we hypothesized that robust transgenic RXFP1 overexpression in cardiomyocytes (CM) protects from TAC-induced HF even in the absence of RLN. Hence, transgenic mice with a CM-specific overexpression of human RXFP1 (hRXFP1tg) were generated. Receptor functionality was demonstrated by in vivo hemodynamics, where the administration of RLN induced positive inotropy strictly in hRXFP1tg. An increase in phospholamban-phosphorylation at serine 16 was identified as a molecular correlate. hRXFP1tg were protected from TAC without additional RLN administration, presenting not only less decline in systolic left ventricular (LV) function but also abrogated LV dilation and pulmonary congestion compared to WT mice. Molecularly, transgenic hearts exhibited not only a significantly attenuated fetal and fibrotic gene activation but also demonstrated less fibrotic tissue and CM hypertrophy in histological sections. These protective effects were evident in both sexes. Similar cardioprotective effects of hRXFP1tg were detectable in a RLN-knockout model, suggesting an alternative mechanism of receptor activation through intrinsic activity, alternative endogenous ligands or crosstalk with other receptors. In summary, CM-specific RXFP1 overexpression provides protection against TAC even in the absence of endogenous RLN. This suggests RXFP1 overexpression as a potential therapeutic approach for HF, offering baseline protection with optional RLN supplementation for specific activation.


Subject(s)
Myocytes, Cardiac , Receptors, G-Protein-Coupled , Receptors, Peptide , Relaxin , Animals , Humans , Male , Mice , Heart Failure/metabolism , Heart Failure/prevention & control , Heart Failure/genetics , Mice, Inbred C57BL , Mice, Transgenic , Myocytes, Cardiac/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Peptide/genetics , Receptors, Peptide/metabolism , Relaxin/genetics , Relaxin/metabolism
13.
Herz ; 49(3): 233-246, 2024 Jun.
Article in German | MEDLINE | ID: mdl-38709278

ABSTRACT

Cardiac device therapy provides not only treatment options for bradyarrhythmia but also advanced treatment for heart failure and preventive measures against sudden cardiac death. In heart failure treatment it enables synergistic reverse remodelling and reduces pharmacological side effects. Cardiac resynchronization therapy (CRT) has revolutionized the treatment of reduced left ventricular ejection fraction (LVEF) and left bundle branch block by decreasing the mortality and morbidity with improvement of the quality of life and resilience. Conduction system pacing (CSP) as an alternative method of physiological stimulation can improve heart function and reduce the risk of pacemaker-induced cardiomyopathy. Leadless pacers and subcutaneous/extravascular defibrillators offer less invasive options with lower complication rates. The prevention of infections through preoperative and postoperative strategies enhances the safety of these therapies.


Subject(s)
Defibrillators, Implantable , Heart Failure , Humans , Cardiac Resynchronization Therapy/methods , Death, Sudden, Cardiac/prevention & control , Evidence-Based Medicine , Heart Failure/therapy , Heart Failure/prevention & control , Pacemaker, Artificial , Treatment Outcome
14.
J Mol Cell Cardiol ; 191: 12-22, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38643934

ABSTRACT

Doxorubicin (DOX) is a widely used chemotherapeutic agent that can cause serious cardiotoxic side effects, leading to heart failure (HF). Impaired mitochondrial function is thought to be key factor driving progression into HF. We have previously shown in a rat model of DOX-HF that heart failure with reduced ejection fraction correlates with mitochondrial loss and dysfunction. Adenosine monophosphate-dependent kinase (AMPK) is a cellular energy sensor, regulating mitochondrial biogenesis and energy metabolism, including fatty acid oxidation. We hypothesised that AMPK activation could restore mitochondrial function and therefore be a novel cardioprotective strategy for the prevention of DOX-HF. Consequently, we set out to assess whether 5-aminoimidazole-4-carboxamide 1-ß-D-ribofuranoside (AICAR), an activator of AMPK, could prevent cardiac functional decline in this chronic intravenous rat model of DOX-HF. In line with our hypothesis, AICAR improved cardiac systolic function. AICAR furthermore improved cardiac mitochondrial fatty acid oxidation, independent of mitochondrial number, and in the absence of observable AMPK-activation. In addition, we found that AICAR prevented loss of myocardial mass. RNAseq analysis showed that this may be driven by normalisation of pathways associated with ribosome function and protein synthesis, which are impaired in DOX-treated rat hearts. AICAR furthermore prevented dyslipidemia and excessive body-weight loss in DOX-treated rats, which may contribute to preservation of myocardial mass. Though it is unclear whether AICAR exerted its cardioprotective effect through cardiac or extra-cardiac AMPK-activation or via an AMPK-independent effect, these results show promise for the use of AICAR as a cardioprotective agent in DOX-HF to both preserve cardiac function and mass.


Subject(s)
AMP-Activated Protein Kinases , Aminoimidazole Carboxamide , Cardiotonic Agents , Doxorubicin , Heart Failure , Ribonucleotides , Animals , Doxorubicin/adverse effects , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Heart Failure/chemically induced , Heart Failure/prevention & control , Heart Failure/metabolism , Heart Failure/pathology , Heart Failure/drug therapy , Ribonucleotides/pharmacology , Male , Cardiotonic Agents/pharmacology , Rats , AMP-Activated Protein Kinases/metabolism , Mitochondria, Heart/metabolism , Mitochondria, Heart/drug effects , Myocardium/metabolism , Myocardium/pathology , Fatty Acids/metabolism , Disease Models, Animal
15.
Br J Pharmacol ; 181(16): 2964-2990, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38679957

ABSTRACT

BACKGROUND AND PURPOSE: Cancer therapy-related cardiovascular adverse events (CAEs) in presence of comorbidities, are in the spotlight of the cardio-oncology guidelines. Carfilzomib (Cfz), indicated for relapsed/refractory multiple myeloma (MM), presents with serious CAEs. MM is often accompanied with co-existing comorbidities. However, Cfz use in MM patients with cardiometabolic syndrome (CMS) or in heart failure with reduced ejection fraction (HFrEF), is questionable. EXPERIMENTAL APPROACH: ApoE-/- and C57BL6/J male mice received 14 weeks Western Diet (WD) (CMS models). C57BL6/J male mice underwent permanent LAD ligation for 14 days (early-stage HFrEF model). CMS- and HFrEF-burdened mice received Cfz for two consecutive or six alternate days. Daily metformin and atorvastatin administrations were performed additionally to Cfz, as prophylactic interventions. Mice underwent echocardiography, while proteasome activity, biochemical and molecular analyses were conducted. KEY RESULTS: CMS did not exacerbate Cfz left ventricular (LV) dysfunction, whereas Cfz led to metabolic complications in both CMS models. Cfz induced autophagy and Ca2+ homeostasis dysregulation, whereas metformin and atorvastatin prevented Cfz-mediated LV dysfunction and molecular deficits in the CMS-burdened myocardium. Early-stage HFrEF led to depressed LV function and increased protein phosphatase 2A (PP2A) activity. Cfz further increased myocardial PP2A activity, inflammation and Ca2+-cycling dysregulation. Metformin co-administration exerted an anti-inflammatory potential on the myocardium without improving LV function. CONCLUSION AND IMPLICATIONS: CMS and HFrEF seem to exacerbate Cfz-induced CAEs, by presenting metabolism-related hidden toxicity and PP2A-related cardiac inflammation, respectively. Metformin retains its prophylactic potential in the presence of CMS, while mitigating inflammation and Ca2+ signalling dysregulation in the HFrEF myocardium.


Subject(s)
Cardiotoxicity , Heart Failure , Mice, Inbred C57BL , Oligopeptides , Animals , Male , Cardiotoxicity/prevention & control , Oligopeptides/pharmacology , Oligopeptides/administration & dosage , Heart Failure/chemically induced , Heart Failure/metabolism , Heart Failure/drug therapy , Heart Failure/prevention & control , Mice , Metabolic Syndrome/chemically induced , Metabolic Syndrome/drug therapy , Metabolic Syndrome/metabolism
17.
N Engl J Med ; 390(16): 1455-1466, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38587237

ABSTRACT

BACKGROUND: Empagliflozin improves cardiovascular outcomes in patients with heart failure, patients with type 2 diabetes who are at high cardiovascular risk, and patients with chronic kidney disease. The safety and efficacy of empagliflozin in patients who have had acute myocardial infarction are unknown. METHODS: In this event-driven, double-blind, randomized, placebo-controlled trial, we assigned, in a 1:1 ratio, patients who had been hospitalized for acute myocardial infarction and were at risk for heart failure to receive empagliflozin at a dose of 10 mg daily or placebo in addition to standard care within 14 days after admission. The primary end point was a composite of hospitalization for heart failure or death from any cause as assessed in a time-to-first-event analysis. RESULTS: A total of 3260 patients were assigned to receive empagliflozin and 3262 to receive placebo. During a median follow-up of 17.9 months, a first hospitalization for heart failure or death from any cause occurred in 267 patients (8.2%) in the empagliflozin group and in 298 patients (9.1%) in the placebo group, with incidence rates of 5.9 and 6.6 events, respectively, per 100 patient-years (hazard ratio, 0.90; 95% confidence interval [CI], 0.76 to 1.06; P = 0.21). With respect to the individual components of the primary end point, a first hospitalization for heart failure occurred in 118 patients (3.6%) in the empagliflozin group and in 153 patients (4.7%) in the placebo group (hazard ratio, 0.77; 95% CI, 0.60 to 0.98), and death from any cause occurred in 169 (5.2%) and 178 (5.5%), respectively (hazard ratio, 0.96; 95% CI, 0.78 to 1.19). Adverse events were consistent with the known safety profile of empagliflozin and were similar in the two trial groups. CONCLUSIONS: Among patients at increased risk for heart failure after acute myocardial infarction, treatment with empagliflozin did not lead to a significantly lower risk of a first hospitalization for heart failure or death from any cause than placebo. (Funded by Boehringer Ingelheim and Eli Lilly; EMPACT-MI ClinicalTrials.gov number, NCT04509674.).


Subject(s)
Heart Failure , Myocardial Infarction , Sodium-Glucose Transporter 2 Inhibitors , Aged , Female , Humans , Male , Middle Aged , Benzhydryl Compounds/therapeutic use , Benzhydryl Compounds/adverse effects , Double-Blind Method , Follow-Up Studies , Glucosides/therapeutic use , Glucosides/adverse effects , Heart Failure/etiology , Heart Failure/mortality , Heart Failure/prevention & control , Hospitalization , Kaplan-Meier Estimate , Myocardial Infarction/complications , Myocardial Infarction/drug therapy , Myocardial Infarction/mortality , Sodium-Glucose Transporter 2 Inhibitors/therapeutic use , Sodium-Glucose Transporter 2 Inhibitors/adverse effects , Treatment Outcome , Heart Disease Risk Factors
18.
Inflammation ; 47(2): 789-806, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38446361

ABSTRACT

Altered cardiac innate immunity is highly associated with the progression of cardiac disease states and heart failure. S100A8/A9 is an important component of damage-associated molecular patterns (DAMPs) that is critically involved in the pathogenesis of heart failure, thus considered a promising target for pharmacological intervention. In the current study, initially, we validated the role of S100A8/A9 in contributing to cardiac injury and heart failure via the overactivation of the ß-adrenergic pathway and tested the potential use of paquinimod as a pharmacological intervention of S100A8/A9 activation in preventing cardiac dysfunction, collagen deposition, inflammation, and immune cell infiltration in ß-adrenergic overactivation-mediated heart failure. This finding was further confirmed by the cardiomyocyte-specific silencing of S100A9 via the use of the adeno-associated virus (AAV) 9-mediated short hairpin RNA (shRNA) gene silencing system. Most importantly, in the assessment of the underlying cellular mechanism by which activated S100A8/A9 cause aggravated progression of cardiac fibrosis and heart failure, we discovered that the activated S100A8/A9 can promote fibroblast-macrophage interaction, independent of inflammation, which is likely a key mechanism leading to the enhanced collagen production. Our results revealed that targeting S100A9 provides dual beneficial effects, which is not only a strategy to counteract cardiac inflammation but also preclude cardiac fibroblast-macrophage interactions. The findings of this study also indicate that targeting S100A9 could be a promising strategy for addressing cardiac fibrosis, potentially leading to future drug development.


Subject(s)
Calgranulin B , Myocytes, Cardiac , Animals , Mice , Adrenergic beta-Agonists/pharmacology , Calgranulin A/metabolism , Calgranulin B/metabolism , Calgranulin B/genetics , Fibroblasts/metabolism , Fibroblasts/drug effects , Fibrosis , Heart Failure/metabolism , Heart Failure/prevention & control , Inflammation/metabolism , Macrophages/metabolism , Macrophages/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology
19.
Heart Lung ; 65: 101-108, 2024.
Article in English | MEDLINE | ID: mdl-38457967

ABSTRACT

BACKGROUND: Growing evidence has shown that antioxidant diets protect against heart failure (HF). However, the association between the composite dietary antioxidant index (CDAI), an important measure of overall antioxidants in the diet, and HF has received little attention. OBJECTIVE: The purpose of this study was to examine the relationship between the CDAI and HF. METHODS: A secondary cross-sectional analysis of the 2003 to 2019 National Health and Nutrition Examination Survey (NHANES) was performed. Weighted multivariable logistic regression was used to test the association between the CDAI and HF in four different models, with subgroup analysis and an interaction test subsequently performed. RESULTS: A total of 37,390 participants were included. The HF groups had lower CDAI levels than those in the non-HF group (0.29 ± 0.04 vs. -0.74 ± 0.16, p < 0.0001). After adjusting for demographic characteristics, lifestyle factors, and disease history, a negative association was found between the CDAI and HF (OR: 0.97, 95 % CI: 0.94, 1.00). There was an inverse trend whereby increasing the CDAI was associated with decreasing the odds of HF (p for trend < 0.001). The subgroup analysis and interaction test showed no significant dependence on demographic characteristics, lifestyle factors, and disease history with regard to this association (all p for interaction > 0.05). CONCLUSION: The CDAI was inversely associated with HF in US adults, with higher CDAI levels possibly being associated with a lower incidence of HF, suggesting that dietary antioxidants may help prevent HF.


Subject(s)
Antioxidants , Heart Failure , Adult , Humans , Nutrition Surveys , Cross-Sectional Studies , Heart Failure/epidemiology , Heart Failure/prevention & control , Diet
20.
Cardiovasc Diabetol ; 23(1): 112, 2024 Mar 30.
Article in English | MEDLINE | ID: mdl-38555463

ABSTRACT

BACKGROUND: Glucagon-like peptide-1 receptor agonists (GLP-1RAs) are effective antidiabetic drugs with potential cardiovascular benefits. Despite their well-established role in reducing the risk of major adverse cardiovascular events (MACE), their impact on heart failure (HF) remains unclear. Therefore, our study examined the cardioprotective effects of tirzepatide (TZT), a novel glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide 1 (GLP-1) receptor agonist. METHODS: A three-steps approach was designed: (i) Meta-analysis investigation with the primary objective of assessing major adverse cardiovascular events (MACE) occurrence from major randomized clinical trials.; (ii) TZT effects on a human cardiac AC16 cell line exposed to normal (5 mM) and high (33 mM) glucose concentrations for 7 days. The gene expression and protein levels of primary markers related to cardiac fibrosis, hypertrophy, and calcium modulation were evaluated. (iii) In silico data from bioinformatic analyses for generating an interaction map that delineates the potential mechanism of action of TZT. RESULTS: Meta-analysis showed a reduced risk for MACE events by TZT therapy (HR was 0.59 (95% CI 0.40-0.79, Heterogeneity: r2 = 0.01, I2 = 23.45%, H2 = 1.31). In the human AC16 cardiac cell line treatment with 100 nM TZT contrasted high glucose (HG) levels increase in the expression of markers associated with fibrosis, hypertrophy, and cell death (p < 0.05 for all investigated markers). Bioinformatics analysis confirmed the interaction between the analyzed markers and the associated pathways found in AC16 cells by which TZT affects apoptosis, fibrosis, and contractility, thus reducing the risk of heart failure. CONCLUSION: Our findings indicate that TZT has beneficial effects on cardiac cells by positively modulating cardiomyocyte death, fibrosis, and hypertrophy in the presence of high glucose concentrations. This suggests that TZT may reduce the risk of diabetes-related cardiac damage, highlighting its potential as a therapeutic option for heart failure management clinical trials. Our study strongly supports the rationale behind the clinical trials currently underway, the results of which will be further investigated to gain insights into the cardiovascular safety and efficacy of TZT.


Subject(s)
Diabetes Mellitus, Type 2 , Diabetes Mellitus , Gastric Inhibitory Polypeptide , Glucagon-Like Peptide-2 Receptor , Heart Failure , Humans , Heart Failure/prevention & control , Diabetes Mellitus/diagnosis , Diabetes Mellitus/drug therapy , Hypertrophy , Hypoglycemic Agents/pharmacology , Myocytes, Cardiac , Fibrosis , Glucose , Glucagon-Like Peptide-1 Receptor
SELECTION OF CITATIONS
SEARCH DETAIL