Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.785
Filter
1.
Medicine (Baltimore) ; 103(19): e37965, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38728489

ABSTRACT

BACKGROUND: Heart failure is a common and severe condition, often complicated by diastolic dysfunction. Current standard therapies such as ACEIs and ARBs have limited efficacy in managing diastolic function. Sacubitril/Valsartan, an emerging therapy, warrants rigorous investigation to elucidate its impact on diastolic function in heart failure patients. METHODS: This systematic review and meta-analysis were conducted adhering to Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines and utilized the PICO schema. Searches were performed on 4 databases-PubMed, Embase, Web of Science, and Cochrane Library-without temporal restrictions. Inclusion and exclusion criteria were strictly defined, and quality assessments were conducted using the Cochrane Collaboration Risk of Bias tool. Both fixed-effects and random-effects models were used for statistical analysis, depending on inter-study heterogeneity assessed by I2 statistics and Chi-square tests. RESULTS: Out of 1129 identified publications, 8 studies met the criteria and were included in the meta-analysis. These studies consisted of both randomized controlled trials and cohort studies and featured diverse global populations. Significant reductions were found in the echocardiographic parameter E/e' ratio and LAVi upon treatment with Sacubitril/Valsartan compared to standard therapies, with mean differences of -1.38 and -4.62, respectively, both with P values < .01. CONCLUSIONS: This meta-analysis demonstrates that Sacubitril/Valsartan significantly improves diastolic function parameters in heart failure patients compared to standard treatments. These findings underscore the potential benefits of Sacubitril/Valsartan in the management of heart failure, particularly for patients with diastolic dysfunction.


Subject(s)
Aminobutyrates , Angiotensin Receptor Antagonists , Biphenyl Compounds , Drug Combinations , Heart Failure , Valsartan , Humans , Valsartan/therapeutic use , Aminobutyrates/therapeutic use , Biphenyl Compounds/therapeutic use , Heart Failure/drug therapy , Heart Failure/physiopathology , Angiotensin Receptor Antagonists/therapeutic use , Tetrazoles/therapeutic use , Diastole/drug effects
2.
Cardiovasc Res ; 120(6): 596-611, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38198753

ABSTRACT

AIMS: A mechanistic link between depression and risk of arrhythmias could be attributed to altered catecholamine metabolism in the heart. Monoamine oxidase-A (MAO-A), a key enzyme involved in catecholamine metabolism and longstanding antidepressant target, is highly expressed in the myocardium. The present study aimed to elucidate the functional significance and underlying mechanisms of cardiac MAO-A in arrhythmogenesis. METHODS AND RESULTS: Analysis of the TriNetX database revealed that depressed patients treated with MAO inhibitors had a lower risk of arrhythmias compared with those treated with selective serotonin reuptake inhibitors. This effect was phenocopied in mice with cardiomyocyte-specific MAO-A deficiency (cMAO-Adef), which showed a significant reduction in both incidence and duration of catecholamine stress-induced ventricular tachycardia compared with wild-type mice. Additionally, cMAO-Adef cardiomyocytes exhibited altered Ca2+ handling under catecholamine stimulation, with increased diastolic Ca2+ reuptake, reduced diastolic Ca2+ leak, and diminished systolic Ca2+ release. Mechanistically, cMAO-Adef hearts had reduced catecholamine levels under sympathetic stress, along with reduced levels of reactive oxygen species and protein carbonylation, leading to decreased oxidation of Type II PKA and CaMKII. These changes potentiated phospholamban (PLB) phosphorylation, thereby enhancing diastolic Ca2+ reuptake, while reducing ryanodine receptor 2 (RyR2) phosphorylation to decrease diastolic Ca2+ leak. Consequently, cMAO-Adef hearts exhibited lower diastolic Ca2+ levels and fewer arrhythmogenic Ca2+ waves during sympathetic overstimulation. CONCLUSION: Cardiac MAO-A inhibition exerts an anti-arrhythmic effect by enhancing diastolic Ca2+ handling under catecholamine stress.


Subject(s)
Calcium Signaling , Calcium-Binding Proteins , Calcium , Catecholamines , Disease Models, Animal , Monoamine Oxidase Inhibitors , Monoamine Oxidase , Myocytes, Cardiac , Ryanodine Receptor Calcium Release Channel , Animals , Monoamine Oxidase/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Catecholamines/metabolism , Monoamine Oxidase Inhibitors/pharmacology , Calcium Signaling/drug effects , Ryanodine Receptor Calcium Release Channel/metabolism , Humans , Calcium-Binding Proteins/metabolism , Calcium-Binding Proteins/genetics , Calcium/metabolism , Male , Mice, Knockout , Tachycardia, Ventricular/enzymology , Tachycardia, Ventricular/prevention & control , Tachycardia, Ventricular/metabolism , Tachycardia, Ventricular/physiopathology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Mice, Inbred C57BL , Phosphorylation , Reactive Oxygen Species/metabolism , Heart Rate/drug effects , Female , Diastole/drug effects , Cyclic AMP-Dependent Protein Kinases/metabolism , Action Potentials/drug effects , Cells, Cultured , Mice
3.
Clin Exp Pharmacol Physiol ; 49(8): 848-857, 2022 08.
Article in English | MEDLINE | ID: mdl-35596518

ABSTRACT

LCZ696, an angiotensin receptor-neprilysin inhibitor, has shown promising clinical efficacy in patients with heart failure (HF) with reduced ejection fraction. However, its potential effects on heart failure with preserved ejection fraction (HFpEF) are still not fully understood. We evaluated the effect of LCZ696 on HFpEF in transverse aortic constriction mice and compared it with the effect of the angiotensin receptor blocker, valsartan. We found that LCZ696 improved cardiac diastolic function by reducing ventricular hypertrophy and fibrosis in mice with overload-induced diastolic dysfunction. In addition, there was superior inhibition of LCZ696 than stand-alone valsartan. As a potential underlying mechanism, we demonstrated that LCZ696 behaves as a potent suppressor of calcium-mediated calcineurin-nuclear factor of activated T cells (NFAT) signalling transduction pathways. Hence, we demonstrated the protective effects of LCZ696 in overload-induced HFpEF and provided a pharmaceutical therapeutic strategy for related diseases.


Subject(s)
Aminobutyrates , Angiotensin Receptor Antagonists , Biphenyl Compounds , Cardiomegaly , Heart Failure , Neprilysin , Stroke Volume , Valsartan , Aminobutyrates/pharmacology , Aminobutyrates/therapeutic use , Angiotensin Receptor Antagonists/pharmacology , Animals , Biphenyl Compounds/pharmacology , Biphenyl Compounds/therapeutic use , Cardiomegaly/drug therapy , Diastole/drug effects , Disease Models, Animal , Drug Combinations , Heart Failure/drug therapy , Heart Failure/physiopathology , Mice , Neprilysin/antagonists & inhibitors , Receptors, Angiotensin/therapeutic use , Stroke Volume/drug effects , Stroke Volume/physiology , Tetrazoles/pharmacology , Tetrazoles/therapeutic use , Valsartan/pharmacology , Valsartan/therapeutic use , Ventricular Dysfunction/drug therapy , Ventricular Dysfunction/physiopathology
4.
J Cachexia Sarcopenia Muscle ; 13(3): 1565-1581, 2022 06.
Article in English | MEDLINE | ID: mdl-35301823

ABSTRACT

BACKGROUND: About half of heart failure (HF) patients, while having preserved left ventricular function, suffer from diastolic dysfunction (so-called HFpEF). No specific therapeutics are available for HFpEF in contrast to HF where reduced ejection fractions (HFrEF) can be treated pharmacologically. Myocardial titin filament stiffening, endothelial dysfunction, and skeletal muscle (SKM) myopathy are suspected to contribute to HFpEF genesis. We previously described small molecules interfering with MuRF1 target recognition thereby attenuating SKM myopathy and dysfunction in HFrEF animal models. The aim of the present study was to test the efficacy of one small molecule (MyoMed-205) in HFpEF and to describe molecular changes elicited by MyoMed-205. METHODS: Twenty-week-old female obese ZSF1 rats received the MuRF1 inhibitor MyoMed-205 for 12 weeks; a comparison was made to age-matched untreated ZSF1-lean (healthy) and obese rats as controls. LV (left ventricle) function was assessed by echocardiography and by invasive haemodynamic measurements until week 32. At week 32, SKM and endothelial functions were measured and tissues collected for molecular analyses. Proteome-wide analysis followed by WBs and RT-PCR was applied to identify specific genes and affected molecular pathways. MuRF1 knockout mice (MuRF1-KO) SKM tissues were included to validate MuRF1-specificity. RESULTS: By week 32, untreated obese rats had normal LV ejection fraction but augmented E/e' ratios and increased end diastolic pressure and myocardial fibrosis, all typical features of HFpEF. Furthermore, SKM myopathy (both atrophy and force loss) and endothelial dysfunction were detected. In contrast, MyoMed-205 treated rats had markedly improved diastolic function, less myocardial fibrosis, reduced SKM myopathy, and increased SKM function. SKM extracts from MyoMed-205 treated rats had reduced MuRF1 content and lowered total muscle protein ubiquitination. In addition, proteomic profiling identified eight proteins to respond specifically to MyoMed-205 treatment. Five out of these eight proteins are involved in mitochondrial metabolism, dynamics, or autophagy. Consistent with the mitochondria being a MyoMed-205 target, the synthesis of mitochondrial respiratory chain complexes I + II was increased in treated rats. MuRF1-KO SKM controls also had elevated mitochondrial complex I and II activities, also suggesting mitochondrial activity regulation by MuRF1. CONCLUSIONS: MyoMed-205 improved myocardial diastolic function and prevented SKM atrophy/function in the ZSF1 animal model of HFpEF. Mechanistically, SKM benefited from an attenuated ubiquitin proteasome system and augmented synthesis/activity of proteins of the mitochondrial respiratory chain while the myocardium seemed to benefit from reduced titin modifications and fibrosis.


Subject(s)
Heart Failure , Muscle Proteins , Muscle, Skeletal , Small Molecule Libraries , Tripartite Motif Proteins , Ubiquitin-Protein Ligases , Animals , Connectin/metabolism , Diastole/drug effects , Female , Fibrosis , Heart Failure/drug therapy , Heart Failure/metabolism , Heart Failure/pathology , Mice , Mice, Knockout , Muscle Proteins/antagonists & inhibitors , Muscle Proteins/metabolism , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscular Atrophy/drug therapy , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , Myocardium/pathology , Rats , Small Molecule Libraries/pharmacology , Stroke Volume/drug effects , Tripartite Motif Proteins/antagonists & inhibitors , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/metabolism
5.
Life Sci ; 291: 120270, 2022 Feb 15.
Article in English | MEDLINE | ID: mdl-34990651

ABSTRACT

Approximately 12-18% of hypertensive patients are diagnosed with resistant hypertension (RH). The risk of having worse cardiovascular outcomes is twice higher in those patients. The low effectiveness of conventional antihypertensive drugs in RH emphasizes the need to evaluate complementary drug therapies to achieve blood pressure (BP) control. Previous studies have demonstrated that phosphodiesterase 5 (PDE-5) inhibitors improve hemodynamics and reduce BP on essential hypertension. So, the authors aimed to summarize current clinical trials-based evidence published concerning the use of PDE-5 inhibitors on BP, cardiovascular function, and hemodynamics of patients with RH. We searched MEDLINE, EMBASE, LILACS, ClinicalTrials.gov, and WHO International Clinical Trials Registry databases on May 15th, 2020 using pre-defined search terms. Two independent reviewers assessed and extracted data from clinical trials that evaluated the effect of PDE-5 inhibitors on BP. We have included five articles in this systematic review. Four of them developed a single-day protocol, while one has developed a 14-day study. The main findings indicate that PDE-5 inhibitors ameliorate BP, vascular hemodynamics, and diastolic function parameters. Some data demonstrated improvement of endothelial function, but it was not a consensus. The side effects seemed to be limited and well-tolerated. In brief, our systematic review highlights the potential of PDE-5 inhibitors as a therapeutic alternative in addition to the multiple-drug regime for RH. Larger studies are still needed to determine whether the beneficial effects of PDE-5 inhibitors on RH would be maintained with chronic administration.


Subject(s)
Hypertension/drug therapy , Phosphodiesterase 5 Inhibitors/pharmacology , Antihypertensive Agents/pharmacology , Blood Pressure/drug effects , Cardiovascular Diseases , Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism , Diastole/drug effects , Humans , Hypertension/physiopathology , Phosphodiesterase 5 Inhibitors/metabolism
6.
Nutrients ; 13(12)2021 Nov 24.
Article in English | MEDLINE | ID: mdl-34959763

ABSTRACT

The role of magnesium in blood pressure has been studied among hypertensive patients; however, there is a dearth of studies exploring the role of magnesium in hypertensive crises. The primary objective of this study was to evaluate the relationship between serum magnesium and blood pressure in patients with hypertensive crises. This was a single-center, retrospective, chart review, cross-sectional study of patients with hypertensive crises. Patients were included if they were eighteen years of age or older, with an international classification disease ninth revision (ICD-9) code of 401.9 (hypertensive crises: emergency or urgency) and a documented magnesium level on their electronic medical record. The primary outcome of the study was the correlation between serum magnesium and blood pressure (systolic blood pressure and diastolic blood pressure) in patients with hypertensive crises. Two hundred and ninety-three patients were included in the study. The primary outcome result showed that serum magnesium was positively correlated with systolic blood pressure (r = 0.143, p = 0.014), but not diastolic blood pressure. Conclusion: This study found a significant positive association between magnesium and systolic blood pressure, but not diastolic blood pressure, among patients with hypertensive crises. This positive association of serum magnesium with systolic blood pressure was maintained after adjusting for covariates. This study's findings suggest a potential role of magnesium in blood pressure among patients with hypertensive crises.


Subject(s)
Blood Pressure/drug effects , Hypertension/blood , Magnesium/blood , Adult , Aged , Aged, 80 and over , Cross-Sectional Studies , Diastole/drug effects , Female , Humans , Male , Middle Aged , Retrospective Studies , Systole/drug effects , Young Adult
7.
Int Heart J ; 62(6): 1297-1304, 2021.
Article in English | MEDLINE | ID: mdl-34853223

ABSTRACT

Vascular endothelial growth factor receptor tyrosine kinase inhibitors (VEGFR-TKIs) frequently induce cardiovascular adverse events, though VEGFR-TKIs contribute to the improvement of the prognosis of patients with malignancies. It is widely accepted that VEGFR-TKIs impair left ventricular systolic functions; however, their effects on diastolic functions remain to be fully elucidated. The purpose of this study was to analyze the impact of VEGFR-TKIs on left ventricular diastolic functions. This study was designed as a retrospective single-center cohort study in Japan. We assessed 24 cases who received VEGFR-TKI monotherapy (sunitinib, sorafenib, pazopanib, axitinib) with left ventricular ejection fraction (LVEF) above 50% during the therapy at the Osaka University Hospital from January 2008 to June 2019. Left ventricular diastolic functions were evaluated by the change in echocardiographic parameters before and after the VEGFR-TKI treatment. Both septal e' and lateral e's decreased after treatment (septal e': before, 6.1 ± 1.8; after, 5.0 ± 1.9; n = 21, P < 0.01; lateral e': before, 8.7 ± 2.8; after, 6.9 ± 2.3; n = 21, P < 0.01). E/A declined after VEGFR-TKIs administration, though not statistically significantly. In 20 cases with at least one risk factor for heart failure with preserved ejection fraction (HFpEF), E/A significantly decreased (0.87 ± 0.34 versus 0.68 ± 0.14; P < 0.05) as well as the septal and lateral e's. These results suggest that treatment with VEGFR-TKIs impairs left ventricular diastolic functions in patients with preserved LVEF, especially in those with risk factors for HFpEF.


Subject(s)
Diastole/drug effects , Protein Kinase Inhibitors/adverse effects , Ventricular Dysfunction, Left/chemically induced , Aged , Cohort Studies , Echocardiography , Female , Humans , Male , Neoplasms/drug therapy , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Retrospective Studies , Stroke Volume
8.
Nutrients ; 13(10)2021 Oct 15.
Article in English | MEDLINE | ID: mdl-34684609

ABSTRACT

BACKGROUND: The association between high fructose consumption and elevated blood pressure continues to be controversial, especially in adolescence. The aim of this study was to assess the association between fructose consumption and elevated blood pressure in an European adolescent population. METHODS: A total of 1733 adolescents (mean ± SD age: 14.7 ± 1.2; percentage of girls: 52.8%) were analysed from the Healthy Lifestyle in Europe by Nutrition in Adolescence (HELENA) study in eight European countries. Blood pressure was measured using validated devices and methods for measuring systolic blood pressure (SBP) and diastolic blood pressure (DBP). Dietary data were recorded via repeated 24 h recalls (using specifically developed HELENA-DIAT software) and converted into pure fructose (monosaccharide form) and total fructose exposure (pure fructose + fructose from sucrose) intake using a specific fructose composition database. Food categories were separated at posteriori in natural vs. were non-natural foods. Elevated BP was defined according to the 90th percentile cut-off values and was compared according to tertiles of fructose intake using univariable and multivariable mixed logistic regression models taking into account confounding factors: centre, sex, age and z-score-BMI, MVPA (Moderate to Vigorous Physical Activity) duration, tobacco consumption, salt intake and energy intake. RESULTS: Pure fructose from non-natural foods was only associated with elevated DBP (DBP above the 10th percentile in the highest consuming girls (OR = 2.27 (1.17-4.40); p = 0.015) after adjustment for cofounding factors. CONCLUSIONS: Consuming high quantities of non-natural foods was associated with elevated DBP in adolescent girls, which was in part due to high fructose levels in these foods categories. The consumption of natural foods containing fructose, such as whole fruits, does not impact blood pressure and should continue to remain a healthy dietary habit.


Subject(s)
Blood Pressure/physiology , Diastole/physiology , Eating , Fructose/adverse effects , Adolescent , Blood Pressure/drug effects , Child , Diastole/drug effects , Female , Humans
9.
Oxid Med Cell Longev ; 2021: 1257283, 2021.
Article in English | MEDLINE | ID: mdl-34630844

ABSTRACT

Heart failure (HF) is a clinical syndrome caused by impairment of ventricular filling, ejection of blood, or both and is categorized as HF with reduced ejection fraction (HFrEF) or HF with preserved ejection fraction (HFpEF) based on left ventricular function. Cardiac fibrosis contributes to left ventricular dysfunction and leads to the development of HF. Ivabradine, an If current selective specific inhibitor, has been shown to improve the prognosis of patients with HF. However, the effects of ivabradine on cardiac function and fibrosis in HFpEF and HFrEF and the underlying mechanism remain unclear. In the present study, we utilized mouse models to mimic HFpEF and HFrEF and evaluated the therapeutic effects of ivabradine. By treating mice with different doses (10 mg/kg/d and 20 mg/kg/d) of ivabradine for 4 or 8 weeks, we found that a high dose of ivabradine improved cardiac diastolic function in HFpEF mice and ameliorated cardiac diastolic and systolic function and ventricular tachycardia incidence in HFrEF mice. Moreover, ivabradine significantly reduced the activation of cardiac fibroblasts and myocardial fibrosis in mice. Mechanistically, microRNA-133a, which was upregulated by ivabradine, targeted connective tissue growth factor and collagen 1 in cardiac fibroblasts and might contribute to the protective role of ivabradine. Together, our work utilized mouse models to study HFpEF and HFrEF, demonstrated the protective role of ivabradine in HFpEF and HFrEF, and elucidated the potential underlying mechanism, which provides an effective strategy for related diseases.


Subject(s)
Cardiotonic Agents/administration & dosage , Heart Failure/drug therapy , Heart Failure/metabolism , Ivabradine/administration & dosage , MicroRNAs/metabolism , Stroke Volume/drug effects , Up-Regulation/drug effects , Ventricular Dysfunction, Left/drug therapy , Ventricular Dysfunction, Left/metabolism , Animals , Animals, Newborn , Cells, Cultured , Diastole/drug effects , Disease Models, Animal , Fibroblasts/metabolism , Heart Ventricles/cytology , Male , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Signal Transduction/genetics , Systole/drug effects , Transfection , Treatment Outcome
10.
Exp Biol Med (Maywood) ; 246(23): 2511-2521, 2021 12.
Article in English | MEDLINE | ID: mdl-34342551

ABSTRACT

Chronic kidney disease (CKD) is a major contributor to the development of heart failure with preserved ejection fraction (HFpEF), whereas the underlying mechanism of cardiorenal HFpEF is still elusive. The aim of this study was to investigate the role of cardiac fibrosis in a rat model of cardiorenal HFpEF and explore whether treatment with Telmisartan, an inhibitor of renin-angiotensin-aldosterone system (RAAS), can ameliorate cardiac fibrosis and preserve diastolic function in cardiorenal HFpEF. Male rats were subjected to 5/6 subtotal nephrectomy (SNX) or sham operation (Sham), and rats were allowed four weeks to recover and form a stable condition of CKD. Telmisartan or vehicle was then administered p.o. (8 mg/kg/d) for 12 weeks. Blood pressure, brain natriuretic peptide (BNP), echocardiography, and cardiac magnetic resonance imaging were acquired to evaluate cardiac structural and functional alterations. Histopathological staining, real-time polymerase chain reaction (PCR) and western blot were performed to evaluate cardiac remodeling. SNX rats showed an HFpEF phenotype with increased BNP, decreased early to late diastolic transmitral flow velocity (E/A) ratio, increased left ventricular (LV) hypertrophy and preserved ejection fraction (EF). Pathology revealed increased cardiac fibrosis in cardiorenal HFpEF rats compared with the Sham group, while chronic treatment with Telmisartan significantly decreased cardiac fibrosis, accompanied by reduced markers of fibrosis (collagen I and collagen III) and profibrotic cytokines (α-smooth muscle actin, transforming growth factor-ß1, and connective tissue growth factor). In addition, myocardial inflammation was decreased after Telmisartan treatment, which was in a linear correlation with cardiac fibrosis. Telmisartan also reversed LV hypertrophy and E/A ratio, indicating that Telmisartan can improve LV remodeling and diastolic function in cardiorenal HFpEF. In conclusion, cardiac fibrosis is central to the pathology of cardiorenal HFpEF, and RAAS modulation with Telmisartan is capable of alleviating cardiac fibrosis and preserving diastolic dysfunction in this rat model.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Cardio-Renal Syndrome/drug therapy , Fibrosis/drug therapy , Heart Failure/drug therapy , Telmisartan/pharmacology , Animals , Antihypertensive Agents/pharmacology , Blood Flow Velocity/drug effects , Blood Pressure/drug effects , Cardio-Renal Syndrome/pathology , Diastole/drug effects , Disease Models, Animal , Echocardiography , Fibrosis/pathology , Heart Failure/pathology , Hypertrophy, Left Ventricular/drug therapy , Male , Natriuretic Peptide, Brain/analysis , Rats , Rats, Sprague-Dawley , Renal Insufficiency, Chronic/drug therapy , Renal Insufficiency, Chronic/pathology , Stroke Volume/drug effects , Ventricular Function, Left/drug effects , Ventricular Remodeling/drug effects
11.
Proc Natl Acad Sci U S A ; 118(28)2021 07 13.
Article in English | MEDLINE | ID: mdl-34260402

ABSTRACT

Sinoatrial node myocytes (SAMs) act as cardiac pacemaker cells by firing spontaneous action potentials (APs) that initiate each heartbeat. The funny current (If) is critical for the generation of these spontaneous APs; however, its precise role during the pacemaking cycle remains unresolved. Here, we used the AP-clamp technique to quantify If during the cardiac cycle in mouse SAMs. We found that If is persistently active throughout the sinoatrial AP, with surprisingly little voltage-dependent gating. As a consequence, it carries both inward and outward current around its reversal potential of -30 mV. Despite operating at only 2 to 5% of its maximal conductance, If carries a substantial fraction of both depolarizing and repolarizing net charge movement during the firing cycle. We also show that ß-adrenergic receptor stimulation increases the percentage of net depolarizing charge moved by If, consistent with a contribution of If to the fight-or-flight increase in heart rate. These properties were confirmed by heterologously expressed HCN4 channels and by mathematical models of If Modeling further suggested that the slow rates of activation and deactivation of the HCN4 isoform underlie the persistent activity of If during the sinoatrial AP. These results establish a new conceptual framework for the role of If in pacemaking, in which it operates at a very small fraction of maximal activation but nevertheless drives membrane potential oscillations in SAMs by providing substantial driving force in both inward and outward directions.


Subject(s)
Biological Clocks/physiology , Electrophysiological Phenomena , Myocytes, Cardiac/physiology , Sinoatrial Node/physiology , Action Potentials/drug effects , Action Potentials/physiology , Animals , Biological Clocks/drug effects , Computer Simulation , Diastole/drug effects , Diastole/physiology , Electrophysiological Phenomena/drug effects , HEK293 Cells , Humans , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Ivabradine/pharmacology , Membrane Transport Modulators/pharmacology , Mice, Inbred C57BL , Myocytes, Cardiac/drug effects , Sinoatrial Node/drug effects
12.
Eur J Pharmacol ; 904: 174170, 2021 Aug 05.
Article in English | MEDLINE | ID: mdl-33984298

ABSTRACT

Diastolic dysfunction is a major feature of hypertrophic cardiomyopathy (HCM). Data from patient tissue and animal models associate increased Ca2+ sensitivity of myofilaments with altered Na+ and Ca2+ ion homeostasis in cardiomyocytes with diastolic dysfunction. In this study, we tested the acute effects of ouabain on ventricular myocytes of an HCM mouse model. The effects of ouabain on contractility and Ca2+ transients were tested in intact adult mouse ventricular myocytes (AMVMs) of Mybpc3-targeted knock-in (KI) and wild-type (WT) mice. Concentration-response assessment of contractile function revealed low sensitivity of AMVMs to ouabain (10 µM) compared to literature data on human cardiomyocytes (100 nM). Three hundred µM ouabain increased contraction amplitude (WT ~1.8-fold; KI ~1.5-fold) and diastolic intracellular Ca2+ in both WT and KI (+12-18%), but further decreased diastolic sarcomere length in KI cardiomyocytes (-5%). Western Blot analysis of whole heart protein extracts revealed 50% lower amounts of Na+/K+ ATPase (NKA) in KI than in WT. Ouabain worsened the diastolic phenotype of KI cardiomyocytes at concentrations which did not impair WT diastolic function. Ouabain led to an elevation of intracellular Ca2+, which was poorly tolerated in KI showing already high cytosolic Ca2+ at baseline due to increased myofilament Ca2+ sensitivity. Lower amounts of NKA in KI could amplify the need to exchange excessive intracellular Na+ for Ca2+ and thereby explain the general tendency to higher diastolic Ca2+ in KI.


Subject(s)
Cardiomyopathy, Hypertrophic/genetics , Cardiomyopathy, Hypertrophic/metabolism , Diastole/drug effects , Enzyme Inhibitors/toxicity , Myocytes, Cardiac/drug effects , Ouabain/toxicity , Sarcomeres/drug effects , Animals , Calcium/metabolism , Carrier Proteins/genetics , Disease Models, Animal , Gene Knock-In Techniques , Mice , Myocardial Contraction/drug effects , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Sodium-Potassium-Exchanging ATPase/metabolism , Systole/drug effects
13.
Bioengineered ; 12(1): 1826-1837, 2021 12.
Article in English | MEDLINE | ID: mdl-33975512

ABSTRACT

Ivabradine (Iva), a heart rate reducing agent that specifically inhibits the pacemaker I(f) ionic current, has been demonstrated to be cardioprotective in many cardiovascular diseases. Autophagy is an evolutionarily conserved metabolic process that regulates cardiac homeostasis. This study is aimed to explore whether autophagy is functionally involved in the cardioprotective effect of Iva in a rat model of myocardial infarction (MI). We observed that Iva treatment (po, 10 mg/kg/day) showed significant recovery on the hemodynamics parameters in MI rats, including left ventricular systolic pressure, left ventricular end diastolic pressure, and maximal ascending/descending rate of left ventricular pressure. Also, Iva treatment dramatically decreased infarct size, inhibited myocardial apoptosis, and reduced the levels of pro-inflammatory cytokines tumor necrosis factor (TNF)-α, interleukin (IL)-1ß and IL-6 in MI rats. Moreover, Iva treatment enhanced autophagy and inhibited PI3K/AKT/mTOR/p70S6K pathway in MI rats. Simultaneously, we observed that autophagy enhancer rapamycin (ip, 10 mg/kg/day) showed similar cardioprotective effects with Iva. Furthermore, we observed that addition of autophagy inhibitor 3-methyladenine (ip, 10 mg/kg/day) counteracted the therapeutic effect of Iva, addressing that Iva attenuated post-MI cardiac injury by enhancing autophagy. In summary, these findings demonstrated that Iva attenuated MI in rats by enhancing autophagy, and PI3K/AKT/mTOR/p70S6K pathway might be involved in the process. Autophagy activation by Iva may be a potential therapeutic strategy for the treatment of MI.


Subject(s)
Autophagy , Cardiotonic Agents/therapeutic use , Ivabradine/therapeutic use , Myocardial Infarction/drug therapy , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Cardiotonic Agents/pharmacology , Cytokines/metabolism , Diastole/drug effects , Heart Ventricles/drug effects , Heart Ventricles/physiopathology , Hemodynamics/drug effects , Inflammation Mediators/metabolism , Ivabradine/pharmacology , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Rats, Sprague-Dawley , Signal Transduction , Systole/drug effects
14.
High Blood Press Cardiovasc Prev ; 28(4): 383-391, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33950510

ABSTRACT

INTRODUCTION: Hypertensive mediated heart disease is the consequence of anatomical and functional changes in cardiovascular system. The benefits on left ventricular (LV) diastolic impairment and remodeling of hypertension treatment are well established. AIM: To evaluate LV structure, systolic and diastolic function of treated hypertensive patients on a medium to long term follow-up. METHODS: Prospectively observational cohort study. Hypertensive patients over 18 years, ultrasound evaluation of LV structure and diastolic and systolic function, follow-up at least once a year. Diastolic function assessed following recommendations of the American Society of Echocardiography and the European Association of Cardiovascular Imaging. RESULTS: 285 patients, mean follow up of 1731 ± 952 days. Sample mean age 56.3 ± 12.5 years, 166 patients (58.3%) were males. Baseline blood pressure 147.8 ± 19/86.8 ± 11 mm Hg, 5 years blood pressure 134.4 ± 15.7/79 ± 9 mm Hg (p < 0.005 SBP and p < 0.01 DBP). Baseline fixed dose combinations 115 patients (40.4%), follow-up 53.1% (p < 0.05). LV remodeling was detected in 88 patients (30.9%) vs. 30.1% at 5 years (p = NS). The frequency of an E/e' ratio > 14 was reduced from 38 patients (13.3%) to 3.6% (p < 0.001), e' septal velocity < 7 cm/sec or e' lateral velocity < 10 cm/sec was reduced from 38.6% (110 patients) to 19.3% (p < 0.001). Baseline normal diastolic function was detected in 85.6% (244 patients) and 94% at the end of the follow-up (p < 0.02). CONCLUSIONS: In this observational cohort followed by a mean of 5 years, the main benefit of hypertension treatment was the prevention or regression of diastolic dysfunction.


Subject(s)
Antihypertensive Agents/therapeutic use , Heart Diseases/etiology , Hypertension/complications , Hypertension/drug therapy , Adult , Aged , Blood Pressure/drug effects , Diastole/drug effects , Female , Follow-Up Studies , Heart Diseases/physiopathology , Heart Diseases/prevention & control , Humans , Hypertension/physiopathology , Hypertrophy, Left Ventricular/etiology , Hypertrophy, Left Ventricular/physiopathology , Hypertrophy, Left Ventricular/prevention & control , Male , Middle Aged , Prospective Studies , Systole/drug effects , Ventricular Dysfunction, Left/etiology , Ventricular Dysfunction, Left/physiopathology , Ventricular Dysfunction, Left/prevention & control
15.
Am J Physiol Heart Circ Physiol ; 320(6): H2416-H2428, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33989083

ABSTRACT

Endothelial cells (ECs) secrete different paracrine signals that modulate the function of adjacent cells; two examples of these paracrine signals are nitric oxide (NO) and neuregulin-1 (NRG1), a cardioprotective growth factor. Currently, it is undetermined whether one paracrine factor can compensate for the loss of another. Herein, we hypothesized that NRG1 can compensate for endothelial NO synthase (eNOS) deficiency. We characterized eNOS null and wild-type (WT) mice by cardiac ultrasound and histology and we determined circulating NRG1 levels. In a separate experiment, eight groups of mice were divided into four groups of eNOS null mice and WT mice; half of the mice received angiotensin II (ANG II) to induce a more severe phenotype. Mice were randomized to daily injections with NRG1 or vehicle for 28 days. eNOS deficiency increased NRG1 plasma levels, indicating that ECs increase their NRG1 expression when NO production is deleted. eNOS deficiency also increased blood pressure, lowered heart rate, induced cardiac fibrosis, and affected diastolic function. In eNOS null mice, ANG II administration not only increased cardiac fibrosis but also induced cardiac hypertrophy and renal fibrosis. NRG1 administration prevented cardiac and renal hypertrophy and fibrosis caused by ANG II infusion and eNOS deficiency. Moreover, Nrg1 expression in the myocardium is shown to be regulated by miR-134. This study indicates that administration of endothelium-derived NRG1 can compensate for eNOS deficiency in the heart and kidneys.NEW & NOTEWORTHY ECs compensate for eNOS deficiency by increasing the secretion of NRG1. NRG1 administration prevents cardiac and renal hypertrophy and fibrosis caused by ANG II infusion and eNOS deficiency. NRG1 expression is regulated by miR-134.


Subject(s)
Endothelial Cells/metabolism , Heart Rate/genetics , Heart/drug effects , MicroRNAs/metabolism , Myocardium/pathology , Neuregulin-1/metabolism , Nitric Oxide Synthase Type III/genetics , Nitric Oxide/metabolism , Angiotensin II/pharmacology , Animals , Cardiomegaly/genetics , Cardiomegaly/pathology , Diastole/drug effects , Fibrosis/genetics , Fibrosis/pathology , Gene Expression Regulation , Heart Rate/drug effects , Kidney/pathology , Mice , Mice, Knockout , Neuregulin-1/pharmacology , Nitric Oxide Synthase Type III/metabolism , Random Allocation , Vasoconstrictor Agents/pharmacology
16.
J Cardiovasc Pharmacol ; 77(6): 805-813, 2021 06 01.
Article in English | MEDLINE | ID: mdl-34001722

ABSTRACT

ABSTRACT: Levosimendan, a calcium sensitizer, exerts inotropic action through improving left ventricular ejection fraction. We noticed that only few clinical studies are published in which the effects of levosimendan on cardiac function are studied by echocardiography. When screening the literature (PubMed, Embase, and CENTRAL, from inception to August 2020), we found 29 randomized controlled trials on levosimendan containing echocardiographic data. We included those studies, describing a total of 574 heart failure patients, in our meta-analysis and extracted 14 ultrasonic parameters, pooling the effect estimates using a random-effect model. Our analysis of the diastolic parameters of the left ventricle shows that levosimendan reduce the early/late transmitral diastolic peak flow velocity ratio [standardized mean difference (SMD) -0.45 to 95% confidence interval (CI) (-0.87 to -0.03), P = 0.037] and E/e' (e': mitral annulus peak early diastolic wave velocity using tissue-doppler imaging) [SMD -0.59, 95% CI (-0.8 to -0.39), P < 0.001]. As it regards the systolic parameters of the right ventricle, levosimendan increased tricuspid annular plane systolic excursion [SMD 0.62, 95% CI (0.28 to 0.95), P < 0.001] and tricuspid annular peak systolic velocity [SMD 0.75, 95% CI (0.35 to 1.16), P < 0.001], and reduced systolic pulmonary artery pressure [SMD -1.02, 95% CI (-1.32, -0.73), P < 0.001]. As it regards the diastolic parameters of the right ventricle, levosimendan was associated with the decrease of Aa (peak late diastolic tricuspid annular velocity using tissue-doppler imaging) [SMD -0.38, 95% CI (-0.76 to 0), P = 0.047] and increase of Ea (peak early diastolic tricuspid annular velocity using tissue-doppler imaging) [SMD 1.03, 95% CI (0.63 to 1.42), P < 0.001] and Ea/Aa [SMD 0.86, 95% CI (0.18 to 1.54), P = 0.013]. We show that levosimendan is associated with an amelioration in the diastolic and systolic functions of both ventricles in heart failure patients.


Subject(s)
Cardiotonic Agents/pharmacology , Heart Failure/drug therapy , Simendan/pharmacology , Diastole/drug effects , Echocardiography , Heart Failure/physiopathology , Humans , Randomized Controlled Trials as Topic , Stroke Volume/drug effects , Systole/drug effects , Ventricular Function, Left/drug effects , Ventricular Function, Right/drug effects
17.
J Diabetes Res ; 2021: 8838026, 2021.
Article in English | MEDLINE | ID: mdl-33855087

ABSTRACT

OBJECTIVES: A single-arm prospective study was conducted among Japanese patients with type 2 diabetes having preserved ejection fraction. The aim was to investigate (1) whether liraglutide therapy could improve B-type natriuretic peptide (BNP) levels and diastolic cardiac function assessed by the E-wave to E' ratio (E/E') using transthoracic echocardiography (TTE), and (2) whether E/E' contributed to BNP improvement independent of bodyweight reduction (UMIN000005565). METHODS: Patients with type 2 diabetes and left ventricular ejection fraction (LVEF) ≥ 40% without heart failure symptoms were enrolled, and daily injection with liraglutide (0.9 mg) was introduced. Cardiac functions were assessed by TTE before and after 26 weeks of liraglutide treatment. Diastolic cardiac function was defined as septal E/E' ≥ 13.0. RESULTS: Thirty-one patients were analyzed. BNP and E/E' improved, with BNP levels declining from 36.8 ± 30.5 pg/mL to 26.3 ± 25.9 pg/mL (p = 0.0014) and E/E' dropping from 12.7 ± 4.7 to 11.0 ± 3.3 (p = 0.0376). The LVEF showed no significant changes. E/E' improved only in patients with E/E' ≥ 13.0. Favorable changes in E/E' were canceled when adjusted for body mass index (BMI). Multivariate linear regression analysis revealed that the left ventricular diastolic diameter and ∆E/E'/∆BMI contributed to ∆BNP/baseline BNP (p = 0.0075, R 2 = 0.49264). CONCLUSIONS: Liraglutide had favorable effects on BNP and E/E' but not on LVEF. E/E' improvement was only seen in patients with diastolic cardiac function. Body weight reduction affected the change of E/E'. The BMI-adjusted E/E' significantly contributed to the relative change of BNP. GLP-1 analog treatment could be considered a therapeutic option against diabetic diastolic cardiac dysfunction regardless of body weight. This trial is registered with the University Hospital Medical Information Network in Japan, with clinical trial registration number: UMIN000005565.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Diastole/drug effects , Liraglutide/therapeutic use , Natriuretic Peptide, Brain/blood , Stroke Volume/physiology , Weight Loss/drug effects , Aged , Diabetes Mellitus, Type 2/physiopathology , Diastole/physiology , Female , Humans , Liraglutide/pharmacology , Male , Middle Aged
18.
Int J Mol Sci ; 22(6)2021 Mar 17.
Article in English | MEDLINE | ID: mdl-33802928

ABSTRACT

Blood platelets' adenosine receptors (AR) are considered to be a new target for the anti-platelet therapy. This idea is based on in vitro studies which show that signaling mediated by these receptors leads to a decreased platelet response to activating stimuli. In vivo evidence for the antithrombotic activity of AR agonists published to date were limited, however, to the usage of relatively high doses given in bolus. The present study was aimed at verifying if these substances used in lower doses in combination with inhibitors of P2Y12 could serve as components of dual anti-platelet therapy. We have found that a selective A2A agonist 2-hexynyl-5'-N-ethylcarboxamidoadenosine (HE-NECA) improved the anti-thrombotic properties of either cangrelor or prasugrel in the model of ferric chloride-induced experimental thrombosis in mice. Importantly, HE-NECA was effective not only when applied in bolus as other AR agonists in the up-to-date published studies, but also when given chronically. In vitro thrombus formation under flow conditions revealed that HE-NECA enhanced the ability of P2Y12 inhibitors to decrease fibrinogen content in thrombi, possibly resulting in their lower stability. Adenosine receptor agonists possess a certain hypotensive effect and an ability to increase the blood-brain barrier permeability. Therefore, the effects of anti-thrombotic doses of HE-NECA on blood pressure and the blood-brain barrier permeability in mice were tested. HE-NECA applied in bolus caused a significant hypotension in mice, but the effect was much lower when the substance was given in doses corresponding to that obtained by chronic administration. At the same time, no significant effect of HE-NECA was observed on the blood-brain barrier. We conclude that chronic administration of the A2A agonist can be considered a potential component of a dual antithrombotic therapy. However, due to the hypotensive effect of the substances, dosage and administration must be elaborated to minimize the side-effects. The total number of animals used in the experiments was 146.


Subject(s)
Adenosine Monophosphate/analogs & derivatives , Adenosine-5'-(N-ethylcarboxamide)/analogs & derivatives , Antithrombins/pharmacology , Fibrinogen/metabolism , Prasugrel Hydrochloride/pharmacology , Purinergic P1 Receptor Agonists/pharmacology , Thrombosis/metabolism , Adenosine Monophosphate/pharmacology , Adenosine-5'-(N-ethylcarboxamide)/pharmacology , Adult , Animals , Blood Pressure/drug effects , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/pathology , Chlorides , Diastole/drug effects , Female , Ferric Compounds , Humans , Laser-Doppler Flowmetry , Male , Mice, Inbred C57BL , Permeability/drug effects , Platelet Activation/drug effects , Platelet Aggregation/drug effects , Purinergic P2Y Receptor Antagonists/pharmacology , Systole/drug effects
19.
Am J Obstet Gynecol ; 225(5): 544.e1-544.e9, 2021 11.
Article in English | MEDLINE | ID: mdl-33887239

ABSTRACT

BACKGROUND: Nifedipine is a widely used drug in pregnancies complicated by maternal hypertensive disorders that can be associated with placental insufficiency and fetal hypoxemia. The evidence regarding fetal myocardial responses to nifedipine in hypoxemia is limited. OBJECTIVE: We hypothesized that nifedipine would not impair fetal sheep cardiac function under hypoxemic environment. In particular, we investigated the effects of nifedipine on fetal ventricular functional parameters and cardiac output. STUDY DESIGN: A total of 21 chronically instrumented fetal sheep at 122 to 134 gestational days (term, 145 days) were included in this study. Fetal cardiac function was evaluated by measuring global longitudinal strain, indices describing ventricular systolic and diastolic function, and cardiac outputs using two-dimensional speckle tracking and tissue and spectral pulsed-wave Doppler echocardiography. Fetal carotid artery blood pressure and blood gas values were invasively monitored. After baseline data collection, fetal hypoxemia was induced by maternal hyperoxygenation. After hypoxemia phase data collection, 9 fetuses received nifedipine infusion, and 12 fetuses received saline infusion. Data were collected 30 and 120 minutes after the infusion was started. After 120 minutes of data collection, maternal and fetal oxygenation were normalized, and normoxemia phase data were collected, while infusion was continued. RESULTS: Hypoxemia decreased fetal carotid artery mean arterial pressure from 40 (8) mm Hg to 35 (8) mm Hg (P<.007), and left ventricular global longitudinal strain showed less deformation than at baseline (P=.001). Under hypoxemia, nifedipine caused a reduction in right ventricular global longitudinal strain (P<.05), a decrease in right ventricular isovolumic relaxation velocity and its deceleration (P<.01) indicating diastolic dysfunction, and a drop in right ventricular cardiac output (P<.05). Nifedipine did not alter fetal left ventricular functional parameters or cardiac output. When normoxemia was restored, fetal right ventricular functional parameters and cardiac output returned to baseline level. CONCLUSION: In hypoxemic fetus, nifedipine impaired right ventricular function and reduced its cardiac output. The detrimental effects of nifedipine on fetal right ventricular function were abolished, when normoxemia was restored. Our findings suggest that in a hypoxemic environment nifedipine triggers detrimental effects on fetal right ventricular function.


Subject(s)
Calcium Channel Blockers/adverse effects , Cardiac Output/drug effects , Fetal Hypoxia/complications , Nifedipine/adverse effects , Ventricular Dysfunction, Right/chemically induced , Animals , Arterial Pressure/drug effects , Carotid Arteries/drug effects , Diastole/drug effects , Echocardiography, Doppler, Pulsed , Fetal Monitoring , Models, Animal , Sheep
20.
Int J Mol Sci ; 22(8)2021 Apr 15.
Article in English | MEDLINE | ID: mdl-33921053

ABSTRACT

Tetracycline antibiotics act by inhibiting bacterial protein translation. Given the bacterial ancestry of mitochondria, we tested the hypothesis that doxycycline-which belongs to the tetracycline class-reduces mitochondrial function, and results in cardiac contractile dysfunction in cultured H9C2 cardiomyoblasts, adult rat cardiomyocytes, in Drosophila and in mice. Ampicillin and carbenicillin were used as control antibiotics since these do not interfere with mitochondrial translation. In line with its specific inhibitory effect on mitochondrial translation, doxycycline caused a mitonuclear protein imbalance in doxycycline-treated H9C2 cells, reduced maximal mitochondrial respiration, particularly with complex I substrates, and mitochondria appeared fragmented. Flux measurements using stable isotope tracers showed a shift away from OXPHOS towards glycolysis after doxycycline exposure. Cardiac contractility measurements in adult cardiomyocytes and Drosophila melanogaster hearts showed an increased diastolic calcium concentration, and a higher arrhythmicity index. Systolic and diastolic dysfunction were observed after exposure to doxycycline. Mice treated with doxycycline showed mitochondrial complex I dysfunction, reduced OXPHOS capacity and impaired diastolic function. Doxycycline exacerbated diastolic dysfunction and reduced ejection fraction in a diabetes mouse model vulnerable for metabolic derangements. We therefore conclude that doxycycline impairs mitochondrial function and causes cardiac dysfunction.


Subject(s)
Anti-Bacterial Agents/pharmacology , Doxycycline/pharmacology , Mitochondria, Heart/metabolism , Myocardial Contraction/drug effects , Aging/metabolism , Animals , Calcium/metabolism , Calcium Signaling/drug effects , Cell Respiration/drug effects , Cytosol/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/physiopathology , Diastole/drug effects , Drosophila melanogaster/drug effects , Drosophila melanogaster/physiology , Electron Transport Complex I/metabolism , Electron Transport Complex II/metabolism , Glucose/metabolism , Glycolysis/drug effects , Male , Mice, Inbred C57BL , Mitochondria, Heart/drug effects , Mitochondrial Proteins/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Nuclear Proteins/metabolism , Oxidative Phosphorylation/drug effects , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...