Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 214
Filter
1.
Cardiovasc Toxicol ; 24(6): 550-562, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38696070

ABSTRACT

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


Subject(s)
Antineoplastic Agents, Immunological , Atrial Function, Left , Breast Neoplasms , Cardiotoxicity , Receptor, ErbB-2 , Trastuzumab , Ventricular Function, Left , Humans , Trastuzumab/adverse effects , Female , Breast Neoplasms/drug therapy , Middle Aged , Receptor, ErbB-2/metabolism , Prospective Studies , Antineoplastic Agents, Immunological/adverse effects , Ventricular Function, Left/drug effects , Atrial Function, Left/drug effects , Adult , Time Factors , Risk Factors , Treatment Outcome , Aged , Predictive Value of Tests , Risk Assessment , Atrial Remodeling/drug effects , Heart Diseases/chemically induced , Heart Diseases/physiopathology , Heart Diseases/diagnostic imaging , Ventricular Dysfunction, Left/chemically induced , Ventricular Dysfunction, Left/physiopathology , Ventricular Dysfunction, Left/diagnostic imaging , Heart Atria/drug effects , Heart Atria/physiopathology , Heart Atria/diagnostic imaging , Stroke Volume/drug effects
3.
J Cardiovasc Pharmacol ; 84(1): 118-123, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38547516

ABSTRACT

ABSTRACT: This study seeks to identify the anticoagulant efficacy of rivaroxaban treatment on thrombi detected using echocardiography of the left atrial appendage in 275 patients with persistent atrial fibrillation. During follow-up after 9-24 weeks of rivaroxaban treatment, patients were divided into "effective group" (n = 143) and "ineffective group" (n = 132) according to the thrombolytic effect of the drug. Left atrial diameter (LAD), left atrial ejection fraction (LAEF), left ventricular ejection fraction (LVEF), mean diameter of left atrial appendage (LAAD mean ), angle between left atrial appendage and left atrium (LAA-A), velocity of blood flow in left atrial appendage (LAA-v), and thrombus size were compared before and after drug administration. Following treatment, LAEF, LVEF, and LAA-v values were greater and LAD and LAAD mean values were lower in the effective ( P < 0.05). Logistic regression analysis showed significant correlations of LAD, LAEF, LVEF, LAA-A, and LAA-v with anticoagulant efficacy ( P < 0.05). The efficacy of rivaroxaban in treatment of left atrial auricular thrombosis in patients with persistent AF was correlated with LAD, LAEF, LVEF, LAA-A, and LAA-v. Multivariate logistic regression analysis further revealed LAEF [odds ratio (OR) 1.7, 95% confidence interval (CI), 0.45-16.9, P = 0.008], 3D-EF (OR 6.4, 95% CI, 1.06-16.9, P = 0.039) and left ventricular global longitudinal strain (OR 18.0, 95% CI, 1.38-35.68, P = 0.028) as factors related to left atrial appendage thrombus. Echocardiography with global longitudinal strain assessment could be effectively utilized to evaluate the functional parameters of LAA and thus aid in predicting the safety of rivaroxaban as an anticoagulation agent.


Subject(s)
Atrial Appendage , Atrial Fibrillation , Echocardiography, Three-Dimensional , Factor Xa Inhibitors , Rivaroxaban , Humans , Atrial Fibrillation/drug therapy , Atrial Fibrillation/physiopathology , Atrial Fibrillation/diagnostic imaging , Atrial Fibrillation/diagnosis , Female , Male , Rivaroxaban/therapeutic use , Rivaroxaban/administration & dosage , Aged , Middle Aged , Treatment Outcome , Atrial Appendage/diagnostic imaging , Atrial Appendage/physiopathology , Atrial Appendage/drug effects , Factor Xa Inhibitors/adverse effects , Factor Xa Inhibitors/therapeutic use , Thrombosis/diagnostic imaging , Thrombosis/drug therapy , Thrombosis/physiopathology , Predictive Value of Tests , Atrial Function, Left/drug effects , Thrombolytic Therapy , Ventricular Function, Left/drug effects , Time Factors
4.
High Blood Press Cardiovasc Prev ; 31(2): 157-166, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38530572

ABSTRACT

INTRODUCTION: Cardiac organ damage like left ventricular (LV) hypertrophy and left atrial (LA) enlargement is more prevalent in women than men with hypertension, but the mechanisms underlying this gender difference remain unclear. METHODS: We tested the association of drug nonadherence with the presence of LV hypertrophy and LA enlargement by echocardiography in 186 women and 337 men with uncontrolled hypertension defined as daytime systolic blood pressure (BP) ≥ 135mmHg despite the prescription of at least two antihypertensive drugs. Drug adherence was assessed by measurements of serum drug concentrations interpreted by an experienced pharmacologist. Aldosterone-renin-ratio (ARR) was measured on actual medication. RESULTS: Women had a higher prevalence of LV hypertrophy (46% vs. 33%) and LA enlargement (79% vs 65%, both p < 0.05) than men, while drug nonadherence (8% vs. 9%, p > 0.514) did not differ. Women were older and had lower serum renin concentration and higher ARR than men, while 24-h systolic BP (141 ± 9 mmHg vs. 142 ± 9 mmHg), and the prevalences of obesity (43% vs. 50%) did not differ (all p > 0.10). In multivariable analyses, female gender was independently associated with a two-fold increased risk of LV hypertrophy (OR 2.01[95% CI 1.30-3.10], p = 0.002) and LA enlargement (OR 1.90 [95% CI 1.17-3.10], p = 0.010), while no association with drug nonadherence was found. Higher ARR was independently associated with LV hypertrophy in men only (OR 2.12 [95% CI 1.12-4.00] p = 0.02). CONCLUSIONS: Among patients with uncontrolled hypertension, the higher prevalence of LV hypertrophy and LA enlargement in women was not explained by differences in drug nonadherence. REGISTRATION: URL:  https://www. CLINICALTRIALS: gov ; Unique identifier: NCT03209154.


Subject(s)
Antihypertensive Agents , Hypertension , Hypertrophy, Left Ventricular , Medication Adherence , Renin , Aged , Female , Humans , Male , Middle Aged , Aldosterone/blood , Antihypertensive Agents/therapeutic use , Arterial Pressure/drug effects , Atrial Function, Left/drug effects , Atrial Remodeling/drug effects , Biomarkers/blood , Cross-Sectional Studies , Health Status Disparities , Hypertension/drug therapy , Hypertension/physiopathology , Hypertension/epidemiology , Hypertrophy, Left Ventricular/epidemiology , Hypertrophy, Left Ventricular/physiopathology , Hypertrophy, Left Ventricular/diagnostic imaging , Prevalence , Renin/blood , Risk Assessment , Risk Factors , Sex Factors , Treatment Outcome , Ventricular Function, Left/drug effects , Ventricular Remodeling/drug effects
6.
Cardiovasc Diabetol ; 20(1): 25, 2021 01 25.
Article in English | MEDLINE | ID: mdl-33494751

ABSTRACT

BACKGROUND: Although the benefits of sodium-glucose cotransporter 2 inhibitors (SGLT2i) on cardiovascular events have been reported in patients with type 2 diabetes mellitus (T2DM) with or without heart failure (HF), the impact of SGLT2i on cardiac remodelling remains to be established. METHODS: We searched the PubMed, Embase, Cochrane Library and Web of Science databases up to November 16th, 2020, for randomized controlled trials reporting the effects of SGLT2i on parameters of cardiac structure, cardiac function, plasma N-terminal pro-brain natriuretic peptide (NT-proBNP) level or the Kansas City Cardiomyopathy Questionnaire (KCCQ) score in T2DM patients with or without chronic HF. The effect size was expressed as the mean difference (MD) or standardized mean difference (SMD) and its 95% confidence interval (CI). Subgroup analyses were performed based on the stage A-B or stage C HF population and HF types. RESULTS: Compared to placebo or other antidiabetic drugs, SGLT2i showed no significant effects on left ventricular mass index, left ventricular end diastolic volume index, left ventricular end systolic volume index, or left atrial volume index. SGLT2i improved left ventricular ejection fraction only in the subgroup of HF patients with reduced ejection fraction (MD 3.16%, 95% CI 0.11 to 6.22, p = 0.04; I2 = 0%), and did not affect the global longitudinal strain in the overall analysis including stage A-B HF patients. SGLT2i showed benefits in the E/e' ratio (MD - 0.45, 95% CI - 0.88 to - 0.03, p = 0.04; I2 = 0%), plasma NT-proBNP level (SMD - 0.09, 95% CI - 0.16 to - 0.03, p = 0.004; I2 = 0%), and the KCCQ score (SMD 3.12, 95% CI 0.76 to 5.47, p  = 0.01; I2 = 0%) in the overall population. CONCLUSION: The use of SGLT2i was associated with significant improvements in cardiac diastolic function, plasma NT-proBNP level, and the KCCQ score in T2DM patients with or without chronic HF, but did not significantly affect cardiac structural parameters indexed by body surface area. The LVEF level was improved only in HF patients with reduced ejection fraction.


Subject(s)
Atrial Function, Left/drug effects , Atrial Remodeling/drug effects , Diabetes Mellitus, Type 2/drug therapy , Heart Failure/drug therapy , Sodium-Glucose Transporter 2 Inhibitors/therapeutic use , Ventricular Function, Left/drug effects , Ventricular Remodeling/drug effects , Aged , Biomarkers/blood , Blood Glucose/drug effects , Blood Glucose/metabolism , Chronic Disease , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/diagnosis , Female , Heart Failure/diagnostic imaging , Heart Failure/physiopathology , Humans , Male , Middle Aged , Natriuretic Peptide, Brain/blood , Peptide Fragments/blood , Recovery of Function , Sodium-Glucose Transporter 2 Inhibitors/adverse effects , Stroke Volume/drug effects , Time Factors , Treatment Outcome
7.
Cardiovasc Diabetol ; 20(1): 7, 2021 01 07.
Article in English | MEDLINE | ID: mdl-33413413

ABSTRACT

BACKGROUND: Sodium-glucose linked transporter type 2 (SGLT-2) inhibition has been shown to reduce cardiovascular mortality in heart failure independently of glycemic control and prevents the onset of atrial arrhythmias, a common co-morbidity in heart failure with preserved ejection fraction (HFpEF). The mechanism behind these effects is not fully understood, and it remains unclear if they could be further enhanced by additional SGLT-1 inhibition. We investigated the effects of chronic treatment with the dual SGLT-1&2 inhibitor sotagliflozin on left atrial (LA) remodeling and cellular arrhythmogenesis (i.e. atrial cardiomyopathy) in a metabolic syndrome-related rat model of HFpEF. METHODS: 17 week-old ZSF-1 obese rats, a metabolic syndrome-related model of HFpEF, and wild type rats (Wistar Kyoto), were fed 30 mg/kg/d sotagliflozin for 6 weeks. At 23 weeks, LA were imaged in-vivo by echocardiography. In-vitro, Ca2+ transients (CaT; electrically stimulated, caffeine-induced) and spontaneous Ca2+ release were recorded by ratiometric microscopy using Ca2+-sensitive fluorescent dyes (Fura-2) during various experimental protocols. Mitochondrial structure (dye: Mitotracker), Ca2+ buffer capacity (dye: Rhod-2), mitochondrial depolarization (dye: TMRE) and production of reactive oxygen species (dye: H2DCF) were visualized by confocal microscopy. Statistical analysis was performed with 2-way analysis of variance followed by post-hoc Bonferroni and student's t-test, as applicable. RESULTS: Sotagliflozin ameliorated LA enlargement in HFpEF in-vivo. In-vitro, LA cardiomyocytes in HFpEF showed an increased incidence and amplitude of arrhythmic spontaneous Ca2+ release events (SCaEs). Sotagliflozin significantly reduced the magnitude of SCaEs, while their frequency was unaffected. Sotagliflozin lowered diastolic [Ca2+] of CaT at baseline and in response to glucose influx, possibly related to a ~ 50% increase of sodium sodium-calcium exchanger (NCX) forward-mode activity. Sotagliflozin prevented mitochondrial swelling and enhanced mitochondrial Ca2+ buffer capacity in HFpEF. Sotagliflozin improved mitochondrial fission and reactive oxygen species (ROS) production during glucose starvation and averted Ca2+ accumulation upon glycolytic inhibition. CONCLUSION: The SGLT-1&2 inhibitor sotagliflozin ameliorated LA remodeling in metabolic HFpEF. It also improved distinct features of Ca2+-mediated cellular arrhythmogenesis in-vitro (i.e. magnitude of SCaEs, mitochondrial Ca2+ buffer capacity, diastolic Ca2+ accumulation, NCX activity). The safety and efficacy of combined SGLT-1&2 inhibition for the treatment and/or prevention of atrial cardiomyopathy associated arrhythmias should be further evaluated in clinical trials.


Subject(s)
Arrhythmias, Cardiac/prevention & control , Atrial Function, Left/drug effects , Atrial Remodeling/drug effects , Glycosides/pharmacology , Heart Atria/drug effects , Heart Failure/drug therapy , Sodium-Glucose Transporter 1/antagonists & inhibitors , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Sodium-Glucose Transporter 2/metabolism , Animals , Arrhythmias, Cardiac/etiology , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/physiopathology , Calcium Signaling/drug effects , Disease Models, Animal , Heart Atria/metabolism , Heart Atria/physiopathology , Heart Failure/etiology , Heart Failure/metabolism , Heart Failure/physiopathology , Metabolic Syndrome/complications , Mitochondria, Heart/drug effects , Mitochondria, Heart/metabolism , Mitochondria, Heart/pathology , Mitochondrial Dynamics/drug effects , Mitochondrial Swelling/drug effects , Rats, Inbred WKY , Rats, Zucker , Reactive Oxygen Species/metabolism , Sodium-Calcium Exchanger/metabolism , Sodium-Glucose Transporter 1/metabolism
8.
Int J Cardiovasc Imaging ; 37(1): 155-162, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32803485

ABSTRACT

Survivors of childhood cancer are at risk of cancer therapy-related cardiac dysfunction (CTRCD) characterized by systolic impairment, with little known about diastolic function. Left atrial strain (LAS) is a surrogate measure of left ventricular filling. We hypothesized that survivors (1) have lower LAS versus controls, and (2) survivors exposed to higher anthracycline dosage have even lower LAS. Cross-sectional study of 45 survivors exposed to anthracyclines ≥ 1 year after chemotherapy and 45 healthy controls. Echo variables included mitral spectral and tissue Doppler, left ventricular ejection fraction (LV EF), LV dimension, LA volume, LV global longitudinal strain (GLS), and LAS. Peak strain (Ɛ) and strain rate (SR) at three phases were obtained: atrial contraction (ac), reservoir (res), and conduit (con). Two sub-analyses of cancer survivors were performed: (1) those with anthracycline dosage ≥ 250 mg/m2, and (2) those with Ɛres in the lowest quartile. On the whole, survivors had lower Ɛres and Ɛcon values. The majority of survivors had relatively normal LAS, while a subset had very low LAS values and were more likely to be older. Survivors exposed to ≥ 250 mg/m2 anthracycline also had lower Ɛres than those < 250 mg/m2. There were no differences in mitral spectral/tissue Doppler, LV dimension, left atrial volume, or GLS. A subset of childhood cancer survivors have lower LAS than their healthy counterparts, while most are essentially normal. Those exposed to higher anthracycline dosage have even lower Ɛres. Longitudinal study of LAS may prove useful in monitoring for CTRCD.


Subject(s)
Anthracyclines/adverse effects , Antibiotics, Antineoplastic/adverse effects , Atrial Function, Left/drug effects , Cancer Survivors , Echocardiography, Doppler , Heart Atria/drug effects , Heart Diseases/chemically induced , Neoplasms/drug therapy , Adolescent , Case-Control Studies , Child , Cross-Sectional Studies , Female , Heart Atria/diagnostic imaging , Heart Atria/physiopathology , Heart Diseases/diagnostic imaging , Heart Diseases/physiopathology , Humans , Male , Predictive Value of Tests , Time Factors , Treatment Outcome
9.
Circulation ; 142(25): 2443-2455, 2020 12 22.
Article in English | MEDLINE | ID: mdl-33092403

ABSTRACT

BACKGROUND: Ibrutinib is a Bruton tyrosine kinase inhibitor with remarkable efficacy against B-cell cancers. Ibrutinib also increases the risk of atrial fibrillation (AF), which remains poorly understood. METHODS: We performed electrophysiology studies on mice treated with ibrutinib to assess inducibility of AF. Chemoproteomic analysis of cardiac lysates identified candidate ibrutinib targets, which were further evaluated in genetic mouse models and additional pharmacological experiments. The pharmacovigilance database, VigiBase, was queried to determine whether drug inhibition of an identified candidate kinase was associated with increased reporting of AF. RESULTS: We demonstrate that treatment of mice with ibrutinib for 4 weeks results in inducible AF, left atrial enlargement, myocardial fibrosis, and inflammation. This effect was reproduced in mice lacking Bruton tyrosine kinase, but not in mice treated with 4 weeks of acalabrutinib, a more specific Bruton tyrosine kinase inhibitor, demonstrating that AF is an off-target side effect. Chemoproteomic profiling identified a short list of candidate kinases that was narrowed by additional experimentation leaving CSK (C-terminal Src kinase) as the strongest candidate for ibrutinib-induced AF. Cardiac-specific Csk knockout in mice led to increased AF, left atrial enlargement, fibrosis, and inflammation, phenocopying ibrutinib treatment. Disproportionality analyses in VigiBase confirmed increased reporting of AF associated with kinase inhibitors blocking Csk versus non-Csk inhibitors, with a reporting odds ratio of 8.0 (95% CI, 7.3-8.7; P<0.0001). CONCLUSIONS: These data identify Csk inhibition as the mechanism through which ibrutinib leads to AF. Registration: URL: https://ww.clinicaltrials.gov; Unique identifier: NCT03530215.


Subject(s)
Adenine/analogs & derivatives , Antineoplastic Agents/toxicity , Atrial Fibrillation/chemically induced , Atrial Function, Left/drug effects , CSK Tyrosine-Protein Kinase/antagonists & inhibitors , Heart Atria/drug effects , Heart Rate/drug effects , Piperidines/toxicity , Protein Kinase Inhibitors/toxicity , Action Potentials/drug effects , Adenine/toxicity , Agammaglobulinaemia Tyrosine Kinase/deficiency , Agammaglobulinaemia Tyrosine Kinase/genetics , Animals , Atrial Fibrillation/enzymology , Atrial Fibrillation/physiopathology , CSK Tyrosine-Protein Kinase/genetics , CSK Tyrosine-Protein Kinase/metabolism , Databases, Genetic , Heart Atria/enzymology , Heart Atria/physiopathology , Humans , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout , Risk Assessment , Risk Factors
10.
Eur J Pharmacol ; 881: 173120, 2020 Aug 15.
Article in English | MEDLINE | ID: mdl-32325147

ABSTRACT

Atrial structural and electrical remodelling play important roles in atrial fibrillation (AF). Sacubitril/valsartan attenuates cardiac remodelling in heart failure. However, the effect of sacubitril/valsartan on AF is unclear. The aim of this study was to evaluate the effect of sacubitril/valsartan on atrial electrical and structural remodelling in AF and investigate the underlying mechanism of action. Thirty-three rabbits were randomized into sham, RAP, and sac/val groups. HL-1 cells were subjected to control treatment or rapid pacing with or without LBQ657 and valsartan. Echocardiography, atrial electrophysiology, and histological examination were performed. The concentration of Ca2+ and expression levels of calcineurin, NFAT, p-NFAT, Cav1.2, collagen Ⅰ and Ⅲ, ANP, BNP, CNP, NT-proBNP, and ST2 in HL-1 cells, and IcaL in left atrial cells, were determined. We observed that compared to that in the sham group, the atrium and right ventricle were enlarged, myocardial fibrosis was markedly higher, AF inducibility was significantly elevated, and atrial effective refractory periods were shortened in the RAP group. These effects were significantly reversed by sacubitril/valsartan. Compared to that in the sham group, collagen Ⅰ and Ⅲ, NT-proBNP, ST2, calcineurin, and NFAT were significantly up-regulated, while p-NFAT and Cav1.2 were down-regulated in the RAP group, and sacubitril/valsartan inhibited these changes. Ca2+ concentration increased and ICaL density decreased in in vivo and in vitro AF models, reversed by sacubitril/valsartan. Sacubitril/valsartan attenuates atrial electrical remodelling and ameliorates structure remodelling in AF. This study paves the way for the possibility of clinical use of sacubitril/valsartan in AF patients.


Subject(s)
Aminobutyrates/pharmacology , Anti-Arrhythmia Agents/pharmacology , Atrial Fibrillation/drug therapy , Atrial Function, Left/drug effects , Atrial Remodeling/drug effects , Heart Atria/drug effects , Heart Rate/drug effects , Tetrazoles/pharmacology , Action Potentials/drug effects , Animals , Atrial Fibrillation/metabolism , Atrial Fibrillation/physiopathology , Biphenyl Compounds , Calcium Signaling/drug effects , Disease Models, Animal , Drug Combinations , Fibrosis , Heart Atria/metabolism , Heart Atria/physiopathology , Male , Rabbits , Valsartan
11.
J Vet Cardiol ; 28: 37-47, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32244178

ABSTRACT

OBJECTIVES: - To evaluate the effect of a single dose of pimobendan on left atrial (LA) function in healthy cats. ANIMALS: - Eleven client owned healthy cats. MATERIAL AND METHODS: - Standardized and repeated echocardiographic examinations were performed on healthy and conscious cats before and after a single dose of orally administered pimobendan (1.25 mg/cat). Left atrial systolic functional parameters were assessed. RESULTS: - Some of the tested parameters of LA function showed significant improvement after pimobendan administration, whereas no significant effect on left ventricular function was observed. In particular, LA minimal diameters obtained from M-mode images in short (p=0.018) and long (p=0.009) axis reduced after pimobendan administration, whereas LA fractional shortening from short (p=0.027) and long (p=0.042) axis and LA appendage emptying velocity (p<0.001) significantly increased. A mild increase in heart rate (p=0.001), and a transient increase on the peak systolic wave pulmonary vein velocity (p=0.008) were also recorded as a possible effect. CONCLUSIONS: - A single dose of pimobendan appears to impact LA function in healthy cats. However, because of the small number of cats included, and the absence of a placebo group, these results cannot be definitively separated from the effect of time. Additional studies are needed to understand if similar effects are observed in cats with cardiomyopathy and LA dilatation.


Subject(s)
Atrial Function, Left/drug effects , Pyridazines/pharmacology , Animals , Blood Flow Velocity/drug effects , Cats , Echocardiography/veterinary , Female , Heart Atria/diagnostic imaging , Heart Rate/drug effects , Male , Pilot Projects , Pulmonary Veins , Pyridazines/administration & dosage , Ventricular Function, Left/drug effects
12.
J Vet Med Sci ; 82(5): 546-552, 2020 May 15.
Article in English | MEDLINE | ID: mdl-32188801

ABSTRACT

This study aimed to assess the effects of atenolol on left ventricular (LV) and left atrial (LA) function in healthy cats and investigate the relationship between atenolol administration and LA enlargement (LAE) in cats with hypertrophic cardiomyopathy (HCM). In study 1, nine experimental cats were used to assess the effects of atenolol in healthy subjects. Cats were administered one of three medication protocols for 7 days: atenolol 6.25 mg/cat twice daily, 12.5 mg/cat twice daily, or placebo (biofermin) 1 tab/cat twice daily. In study 2, cats with HCM were retrospectively recruited and divided into four groups according to atenolol administration [(control group (Cont) or atenolol administration group (Ate)] and the presence or absence of LAE as follows: Cont LAE (-) group (n=42), Cont LAE (+) group (n=20), Ate LAE (-) group (n=17), and Ate LAE (+) group (n=12). LV and LA functions were compared in both studies. LV and LA functions were decreased by atenolol administration in study 1. In study 2, the peak myocardial velocity during early diastole (E') was significantly decreased in the Cont LAE (+), Ate LAE (-), and Ate LAE (+) groups compared to that in the Cont LAE (-) group, but there were no significant differences between LAE (+) groups. Multivariate logistic analysis revealed that atenolol administration was not associated with LAE. Diastolic dysfunction may be associated with LAE; however, atenolol administration did not affect LAE in cats with HCM.


Subject(s)
Adrenergic beta-1 Receptor Antagonists/pharmacology , Atenolol/pharmacology , Atrial Function, Left/drug effects , Cardiomyopathy, Hypertrophic/veterinary , Ventricular Function, Left/drug effects , Animals , Atrial Remodeling/drug effects , Cardiomyopathy, Hypertrophic/drug therapy , Cat Diseases/drug therapy , Cats , Echocardiography/veterinary , Female , Male , Retrospective Studies
13.
J Cardiovasc Transl Res ; 13(5): 731-740, 2020 10.
Article in English | MEDLINE | ID: mdl-32048199

ABSTRACT

Both diabetes mellitus (DM) and atrial fibrillation (AF) are usually associated with enhanced inflammatory response. The effect of the "NACHT, LRR and PYD domain containing protein 3" (NLRP3)-inflammasome/caspase-1/galectin-3 pathway and the potential benefits of NLRP3-inflammasome inhibitor glibenclamide (GLB) on atrial remodeling in the DM state are still unknown. Here, we demonstrated that higher AF inducibility and conduction inhomogeneity, slower epicardial conduction velocity, and increased amount of fibrosis in diabetic rabbits as against normal ones were markedly reduced by GLB. Atrial caspase-1 activity as well as serum IL-1ß and IL-18 levels were elevated in diabetic animals but suppressed by GLB. Moreover, GLB decreased the DM-induced protein expression enhancement of NLRP3, Gal-3, TGF-ß1, and CaV1.2 according to western blot analysis. Summarily, our findings indicate that the NLRP3-inflammasome/caspase-1/Gal-3 signaling pathway is related to the pathogenesis of AF in the diabetic state. NLRP3-inflammasome inhibitor GLB prevents AF inducibility and moderates atrial structural remodeling in DM.


Subject(s)
Atrial Fibrillation/etiology , Atrial Function, Left , Atrial Remodeling , Caspase 1/metabolism , Diabetes Mellitus, Experimental/complications , Galectin 3/metabolism , Heart Atria/metabolism , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Alloxan , Animals , Anti-Inflammatory Agents/pharmacology , Atrial Fibrillation/metabolism , Atrial Fibrillation/physiopathology , Atrial Fibrillation/prevention & control , Atrial Function, Left/drug effects , Atrial Remodeling/drug effects , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/physiopathology , Fibrosis , Glyburide/pharmacology , Heart Atria/drug effects , Heart Atria/physiopathology , Heart Rate , Inflammasomes/antagonists & inhibitors , Isolated Heart Preparation , Male , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Rabbits , Signal Transduction
14.
Open Vet J ; 9(4): 375-383, 2020 01.
Article in English | MEDLINE | ID: mdl-32042661

ABSTRACT

Background: Left atrial (LA) function is an important determinant of the left ventricular (LV) filling, playing a key role in maintaining optimal cardiac performance. Pimobendan is a phosphodiesterase III inhibitor with positive inotropic and vasodilator effects. The present study aims to investigate the effects of pimobendan on LA function in dogs with stage B2 myxomatous mitral valve disease (MMVD). Aim: The aim of this investigation was to study the effects of pimobendan on LA function in dogs with preclinical MMVD. Methods: Twenty-seven dogs with stage B2 MMVD were retrospectively included. LA function was assessed before and 1-6 months following pimobendan initiation. For each dog, two-dimensional (2D) echocardiography was performed to assess LA diameter and volume for each phase of the LA cycle and to assess complete, passive, and active LA function. Pulsed-wave tissue Doppler imaging (TDI) of the left ventricular longitudinal myocardial velocity associated with atrial contraction (A'), both at the level of the interventricular septum and the LV free wall, was also used as an indicator of LA function. Results: There were no significant differences in any of the left atrial variables pre- and posttreatment. Conclusion: Echocardiographic estimates of LA function by 2D diameters and volumes and TDI A' in dogs with MMVD do not change after treatment with pimobendan.


Subject(s)
Atrial Function, Left/drug effects , Dog Diseases/drug therapy , Heart Valve Diseases/veterinary , Pyridazines/pharmacology , Vasodilator Agents/pharmacology , Animals , Cardiotonic Agents/pharmacology , Dogs , Female , Heart Valve Diseases/drug therapy , Male , Phosphodiesterase Inhibitors/pharmacology
15.
BMC Cardiovasc Disord ; 20(1): 50, 2020 02 03.
Article in English | MEDLINE | ID: mdl-32013885

ABSTRACT

BACKGROUND: Previous studies demonstrated impaired lipid metabolism and augmented aerobic glycolysis in AF. The authors aimed to investigate whether the use of metformin, an AMPK activator, could reverse this metabolic remodeling in chronic AF and to explore the underlying mechanisms. METHODS: We conducted chronic AF animal models with 18 beagle dogs and divided them into SR (pacemaker implanted without pacing), AF (pacemaker implanted with sustained pacing at a frequency of 400 beats/min for 6 weeks), and metformin+AF group (daily oral administration of metformin was initiated 1 week before surgery and continued throughout the study period). After electrophysiological measurements, the left atrial appendage tissue samples were taken from the beating heart for further analysis. Protein expression, histological analysis, and biochemical measurements were conducted. RESULTS: The AF groups showed decreased expression of FAT/CD36, CPT-1, VLCAD, increased concentration of free fatty acid and triglyceride, and increased lipid deposition. The activation of AMPK/PGC-1α/PPARα pathway was decreased. The key factors of the Warburg effect, including HIF-1α, GLUT-1, PDK1, HK, and LDH, increased in AF group compared to SR group. The expression of PDH decreased significantly, accompanied by increased atrial lactate production. The extent of fibrosis increased significantly in the left atrial appendage of AF group. dERP, ∑WOV, and AF inducibility increased while ERP decreased in AF group compared to SR group. The use of metformin attenuated all these changes effectively. CONCLUSIONS: Metformin improves lipid metabolism and reverses the Warburg effect in chronic AF via AMPK activation. It attenuates atrial electrical and structural remodeling.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Atrial Appendage/drug effects , Atrial Fibrillation/drug therapy , Energy Metabolism/drug effects , Enzyme Activators/pharmacology , Lipid Metabolism/drug effects , Metformin/pharmacology , Animals , Atrial Appendage/enzymology , Atrial Appendage/physiopathology , Atrial Fibrillation/enzymology , Atrial Fibrillation/physiopathology , Atrial Function, Left/drug effects , Atrial Remodeling/drug effects , Chronic Disease , Disease Models, Animal , Dogs , Enzyme Activation , Heart Rate/drug effects , Male
16.
JACC Cardiovasc Imaging ; 13(5): 1107-1115, 2020 05.
Article in English | MEDLINE | ID: mdl-31954642

ABSTRACT

OBJECTIVES: The aim of this study was to assess the effect of congestion and decongestive therapy on left atrial (LA) mechanics and to determine the relationship between LA improvement after decongestive therapy and clinical outcome in immediate or chronic heart failure with reduced ejection fraction (HFrEF). BACKGROUND: LA mechanics are affected by volume/pressure overload in decompensated HFrEF. METHODS: A total of 31 patients with HFrEF and immediate heart failure (age 64 ± 15 years, 74% male, left ventricular ejection fraction 20 ± 12%) underwent serial echocardiography during decongestive therapy with simultaneous hemodynamic monitoring. LA function was assessed by strain (rate) imaging. Patients were re-evaluated 6 weeks after discharge and prospectively followed up for the composite endpoint of heart failure readmission and all-cause mortality. RESULTS: LA reservoir function was markedly reduced at baseline and improved with decongestion (peak atrial longitudinal strain from 6.4 ± 2.2% to 8.8 ± 3.0% and strain rate from 0.29 ± 0.11 s-1 to 0.38 ± 0.13 s-1), independent of changes in left ventricular global longitudinal strain, LA end-diastolic volume, and mitral regurgitation severity (p < 0.001). Both measures continued to rise at 6 weeks (up to 13.4 ± 6.1% and 0.50 ± 0.19 s-1, respectively; p < 0.001). LA pump strain rate only increased 6 weeks after discharge (-0.25 ± 0.12 s-1 to -0.55 ± 0.29 s-1; p < 0.010). Changes in LA mechanics correlated with changes in wedge pressure (r = -0.61; p < 0.001). Lower peak atrial longitudinal strain values after decongestion were associated with increased risk for the composite endpoint of heart failure and mortality (p < 0.019). CONCLUSIONS: LA reservoir and booster function, while severely impaired during immediate decompensation, significantly improve during and after decongestive therapy. Poor LA reservoir function after decongestion is associated with worse outcome.


Subject(s)
Atrial Function, Left , Echocardiography , Heart Failure/diagnostic imaging , Hemodynamic Monitoring , Hemodynamics , Aged , Atrial Function, Left/drug effects , Cardiovascular Agents/therapeutic use , Disease Progression , Female , Heart Failure/drug therapy , Heart Failure/mortality , Heart Failure/physiopathology , Hemodynamics/drug effects , Humans , Male , Middle Aged , Patient Readmission , Predictive Value of Tests , Prospective Studies , Recovery of Function , Time Factors , Treatment Outcome
18.
J Ethnopharmacol ; 248: 112317, 2020 Feb 10.
Article in English | MEDLINE | ID: mdl-31629862

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Arnebiae Radix, a common herbal medicine in China, is often utilized to treat blood-heat syndrome and has been reported to exert an effect on the heart. AIM OF THE STUDY: The combination of acetylcholine (Ach) and CaCl2 has been widely used to induce atrial fibrillation (AF) in animals. However, whether Arnebiae Radix displays any preventive action on Ach-CaCl2 induced AF in rats remains uncertain. In our study, we attempted to investigate the protective effects of Arnebiae Radix on Ach-CaCl2 induced AF compared to amiodarone, which was employed as the positive control. MATERIALS AND METHODS: To establish the AF model, SD rats were treated with a mixture of 0.1 mL/100 g Ach-CaCl2 (60 µg/mL Ach and 10 mg/mL CaCl2) by tail vein injection for 7 days. Rats were also given a gavage of Arnebiae Radix (0.18 g/mL) one week before or concurrently with the establishment of the AF model. At the end of the experimental period, the induction, duration and timing of AF were monitored using electrocardiogram recordings. Left atrial tissues were stained to observe the level of fibrosis. Electrophysiological measurements were used to examine atrial size and function. RESULTS: In Ach-CaCl2-induced AF rats, Arnebiae Radix decreased AF induction, duration and susceptibility to AF. In addition, Arnebiae Radix significantly reduced atrial fibrosis and inhibited atrial enlargement induced by Ach-CaCl2. Moreover, there was an apparent improvement in cardiac function in the Arnebiae Radix-treated group. CONCLUSIONS: Our findings indicate that Arnebiae Radix treatment can attenuate Ach-CaCl2-induced atrial injury and serve as an effective therapeutic strategy for the treatment of AF in the future.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Atrial Fibrillation/prevention & control , Atrial Function, Left/drug effects , Atrial Remodeling/drug effects , Boraginaceae , Heart Rate/drug effects , Plant Extracts/pharmacology , Acetylcholine , Animals , Anti-Arrhythmia Agents/isolation & purification , Atrial Fibrillation/chemically induced , Atrial Fibrillation/physiopathology , Boraginaceae/chemistry , Calcium Chloride , Disease Models, Animal , Fibrosis , Male , Plant Extracts/isolation & purification , Rats, Sprague-Dawley , Ventricular Function, Left/drug effects , Ventricular Remodeling/drug effects
19.
Circ Heart Fail ; 12(11): e005835, 2019 11.
Article in English | MEDLINE | ID: mdl-31684756

ABSTRACT

BACKGROUND: Hypertrophic cardiomyopathy (HCM) is a genetic cardiovascular disorder, primarily involving mutations in sarcomeric proteins. HCM patients present with hypertrophy, diastolic dysfunction, and fibrosis, but there is no specific treatment. The sphingosine-1-phosphate receptor modulator, FTY720/fingolimod, is approved for treatment of multiple sclerosis. We hypothesize that modulation of the sphingosine-1-phosphate receptor by FTY720 would be of therapeutic benefit in sarcomere-linked HCM. METHODS: We treated mice with an HCM-linked mutation in tropomyosin (Tm-E180G) and nontransgenic littermates with FTY720 or vehicle for 6 weeks. Compared with vehicle-treated, FTY720-treated Tm-E180G mice had a significant reduction in left atrial size (1.99±0.19 [n=7] versus 2.70±0.44 [n=6] mm; P<0.001) and improvement in diastolic function (E/A ratio: 2.69±0.38 [n=7] versus 5.34±1.19 [n=6]; P=0.004) as assessed by echocardiography. RESULTS: Pressure-volume relations revealed significant improvements in the end-diastolic pressure volume relationship, relaxation kinetics, preload recruitable stroke work, and ejection fraction. Detergent-extracted fiber bundles revealed a significant decrease in myofilament Ca2+-responsiveness (pCa50=6.15±0.11 [n=13] versus 6.24±0.06 [n=14]; P=0.041). We attributed these improvements to a downregulation of S-glutathionylation of cardiac myosin binding protein-C in FTY720-treated Tm-E180G mice and reduction in oxidative stress by downregulation of NADPH oxidases with no changes in fibrosis. CONCLUSIONS: This is the first demonstration that modulation of S1PR results in decreased myofilament-Ca2+-responsiveness and improved diastolic function in HCM. We associated these changes with decreased oxidative modification of myofilament proteins via downregulation of NOX2. Our data support the hypothesis that modification of sphingolipid signaling may be a novel therapeutic approach in HCM.


Subject(s)
Atrial Function, Left/drug effects , Atrial Remodeling/drug effects , Cardiomyopathy, Hypertrophic/drug therapy , Fingolimod Hydrochloride/pharmacology , Myocytes, Cardiac/drug effects , Sphingosine 1 Phosphate Receptor Modulators/pharmacology , Sphingosine-1-Phosphate Receptors/drug effects , Animals , Calcium Signaling/drug effects , Cardiomyopathy, Hypertrophic/genetics , Cardiomyopathy, Hypertrophic/metabolism , Cardiomyopathy, Hypertrophic/physiopathology , Diastole , Disease Models, Animal , Female , Fibrosis , Genetic Predisposition to Disease , Male , Mice, Mutant Strains , Mutation , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Myofibrils/drug effects , Myofibrils/metabolism , Myofibrils/pathology , Oxidative Stress/drug effects , Phenotype , Sphingosine-1-Phosphate Receptors/metabolism , Tropomyosin/genetics
20.
Cardiovasc Diabetol ; 18(1): 165, 2019 11 28.
Article in English | MEDLINE | ID: mdl-31779619

ABSTRACT

BACKGROUND: Diabetes mellitus is an important risk factor for atrial fibrillation (AF) development. Sodium-glucose co-transporter-2 (SGLT-2) inhibitors are used for the treatment of type 2 diabetes mellitus (T2DM). Their cardioprotective effects have been reported but whether they prevent AF in T2DM patients are less well-explored. We tested the hypothesis that the SGLT-2 inhibitor, empagliflozin, can prevent atrial remodeling in a diabetic rat model. METHODS: High-fat diet and low-dose streptozotocin (STZ) treatment were used to induce T2DM. A total of 96 rats were randomized into the following four groups: (i) control (ii) T2DM, (iii) low-dose empagliflozin (10 mg/kg/day)/T2DM; and (iv) high-dose empagliflozin (30 mg/kg/day)/T2DM by the intragastric route for 8 weeks. RESULTS: Compared with the control group, left atrial diameter, interstitial fibrosis and the incidence of AF inducibility were significantly increased in the DM group. Moreover, atrial mitochondrial respiratory function, mitochondrial membrane potential, and mitochondrial biogenesis were impaired. Empagliflozin treatment significantly prevented the development of these abnormalities in DM rats, likely via the peroxisome proliferator-activated receptor-c coactivator 1α (PGC-1α)/nuclear respiratory factor-1 (NRF-1)/mitochondrial transcription factor A (Tfam) signaling pathway. CONCLUSIONS: Empagliflozin can ameliorate atrial structural and electrical remodeling as well as improve mitochondrial function and mitochondrial biogenesis in T2DM, hence may be potentially used in the prevention of T2DM-related atrial fibrillation.


Subject(s)
Atrial Fibrillation/prevention & control , Atrial Function, Left/drug effects , Atrial Remodeling/drug effects , Benzhydryl Compounds/pharmacology , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 2/drug therapy , Glucosides/pharmacology , Heart Rate/drug effects , Mitochondria, Heart/drug effects , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Animals , Atrial Fibrillation/etiology , Atrial Fibrillation/metabolism , Atrial Fibrillation/physiopathology , Diabetes Mellitus, Experimental/etiology , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/physiopathology , Diet, High-Fat , Disease Models, Animal , Male , Membrane Potential, Mitochondrial/drug effects , Mitochondria, Heart/metabolism , Mitochondrial Proteins/metabolism , Organelle Biogenesis , Oxidative Stress/drug effects , Rats, Sprague-Dawley , Signal Transduction , Streptozocin
SELECTION OF CITATIONS
SEARCH DETAIL
...