Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
ESC Heart Fail ; 11(1): 601-605, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38030138

ABSTRACT

AIM: Acyl ghrelin increases cardiac output (CO) in heart failure with reduced ejection fraction (HFrEF). This could impair the right ventricular-pulmonary arterial coupling (RVPAC), both through an increased venous return and right ventricular afterload. We aim to investigate if acyl ghrelin increases CO with or without worsening the right-sided haemodynamics in HFrEF assessed by RVPAC. METHODS AND RESULTS: The Karolinska Acyl ghrelin Trial was a randomized double-blind placebo-controlled trial of acyl ghrelin versus placebo (120-min intravenous infusion) in HFrEF. RVPAC was assessed echocardiographically at baseline and 120 min. ANOVA was used for difference in change between acyl ghrelin versus placebo, adjusted for baseline values. Of the 30 randomized patients, 22 had available RVPAC (acyl ghrelin n = 12, placebo n = 10). Despite a 15% increase in CO in the acyl ghrelin group (from 4.0 (3.5-4.6) to 4.6 (3.9-6.1) L/min, P = 0.003), RVPAC remained unchanged; 5.9 (5.3-7.6) to 6.3 (4.8-7.5) mm·(m/s)-1 , P = 0.372, while RVPAC was reduced in the placebo group, 5.2 (4.3-6.4) to 4.8 (4.2-5.8) mm·(m/s)-1 , P = 0.035. Comparing change between groups, CO increased in the acyl ghrelin group versus placebo (P = 0.036) while RVPAC and the right ventricular pressure gradient remained unchanged. CONCLUSION: Treatment with acyl ghrelin increases CO while preserving or even improving RVPAC in HFrEF, possibly due to increased contractility, reduced PVR and/or reduced left sided filling pressures. These potential effects strengthen the role of acyl ghrelin therapy in HFrEF with right ventricular failure.


Subject(s)
Heart Failure , Hypertension, Pulmonary , Humans , Stroke Volume , Heart Failure/drug therapy , Ghrelin/pharmacology , Ghrelin/therapeutic use , Cardiac Output
3.
Front Cardiovasc Med ; 10: 1228789, 2023.
Article in English | MEDLINE | ID: mdl-37608817

ABSTRACT

Cardiovascular diseases (CVD) are still the leading cause of death worldwide. The improved survival of patients with comorbidities such as type 2 diabetes, hypertension, obesity together with the extension of life expectancy contributes to raise the prevalence of CVD in the increasingly aged society. Therefore, a translational research platform that enables precise evaluation of cardiovascular function in healthy and disease condition and assess the efficacy of novel pharmacological treatments, could implement basic science and contribute to reduce CVD burden. Heart failure is a deadly syndrome characterized by the inability of the heart to meet the oxygen demands of the body (unless there is a compensatory increased of filling pressure) and can manifest either with reduced ejection fraction (HFrEF) or preserved ejection fraction (HFpEF). The development and progression of HFrEF is mostly attributable to impaired contractile performance (systole), while in HFpEF the main problem resides in decreased ability of left ventricle to relax and allow the blood filling (diastole). Murine preclinical models have been broadly used in research to understand pathophysiologic mechanisms of heart failure and test the efficacy of novel therapies. Several methods have been employed to characterise cardiac systolic and diastolic function including Pressure Volume (PV) loop hemodynamic analysis, echocardiography and Magnetic Resonance Imaging (MRI). The choice of one methodology or another depends on many aspects including budget available, skills of the operator and design of the study. The aim of this review is to discuss the importance of several methodologies that are commonly used to characterise the cardiovascular phenotype of preclinical models of heart failure highlighting advantages and limitation of each procedure. Although it requires highly skilled operators for execution, PV loop analysis represents the "gold standard" methodology that enables the assessment of left ventricular performance also independently of vascular loading conditions and heart rate, which conferee a really high physiologic importance to this procedure.

4.
Eur Heart J ; 44(22): 2009-2025, 2023 Jun 09.
Article in English | MEDLINE | ID: mdl-36916707

ABSTRACT

BACKGROUND AND AIMS: Ghrelin is an endogenous appetite-stimulating peptide hormone with potential cardiovascular benefits. Effects of acylated (activated) ghrelin were assessed in patients with heart failure and reduced ejection fraction (HFrEF) and in ex vivo mouse cardiomyocytes. METHODS AND RESULTS: In a randomized placebo-controlled double-blind trial, 31 patients with chronic HFrEF were randomized to synthetic human acyl ghrelin (0.1 µg/kg/min) or placebo intravenously over 120 min. The primary outcome was change in cardiac output (CO). Isolated mouse cardiomyocytes were treated with acyl ghrelin and fractional shortening and calcium transients were assessed. Acyl ghrelin but not placebo increased cardiac output (acyl ghrelin: 4.08 ± 1.15 to 5.23 ± 1.98 L/min; placebo: 4.26 ± 1.23 to 4.11 ± 1.99 L/min, P < 0.001). Acyl ghrelin caused a significant increase in stroke volume and nominal increases in left ventricular ejection fraction and segmental longitudinal strain and tricuspid annular plane systolic excursion. There were no effects on blood pressure, arrhythmias, or ischaemia. Heart rate decreased nominally (acyl ghrelin: 71 ± 11 to 67 ± 11 b.p.m.; placebo 69 ± 8 to 68 ± 10 b.p.m.). In cardiomyocytes, acyl ghrelin increased fractional shortening, did not affect cellular Ca2+ transients, and reduced troponin I phosphorylation. The increase in fractional shortening and reduction in troponin I phosphorylation was blocked by the acyl ghrelin antagonist D-Lys 3. CONCLUSION: In patients with HFrEF, acyl ghrelin increased cardiac output without causing hypotension, tachycardia, arrhythmia, or ischaemia. In isolated cardiomyocytes, acyl ghrelin increased contractility independently of preload and afterload and without Ca2+ mobilization, which may explain the lack of clinical side effects. Ghrelin treatment should be explored in additional randomized trials. CLINICAL TRIAL REGISTRATION: ClinicalTrials.gov Identifier: NCT05277415.


Subject(s)
Heart Failure , Ventricular Dysfunction, Left , Humans , Mice , Animals , Myocytes, Cardiac/metabolism , Calcium/metabolism , Ghrelin/pharmacology , Ghrelin/therapeutic use , Stroke Volume , Ventricular Function, Left , Troponin I/metabolism
5.
Acta Physiol (Oxf) ; 236(3): e13869, 2022 11.
Article in English | MEDLINE | ID: mdl-36002394

ABSTRACT

AIMS: Cardiac contractile dysfunction is prevalent in rheumatoid arthritis (RA), with an increased risk for heart failure. A hallmark of RA has increased levels of peptidyl arginine deaminases (PAD) that convert arginine to citrulline leading to ubiquitous citrullination, including in the heart. We aimed to investigate whether PAD-dependent citrullination in the heart was linked to contractile function in a mouse model of RA during the acute inflammatory phase. METHODS: We used hearts from the collagen-induced arthritis (CIA) mice, with overt arthritis, and control mice to analyze cardiomyocyte Ca2+ handling and fractional shortening, the force-Ca2+ relationship in isolated myofibrils, the levels of PAD, protein post-translational modifications, and Ca2+ handling protein. Then, we used an in vitro model to investigate the role of TNF-α in the PAD-mediated citrullination of proteins in cardiomyocytes. RESULTS: Cardiomyocytes from CIA mice displayed larger Ca2+ transients than controls, whereas cell shortening was similar in the two groups. Myofibrils from CIA hearts required higher [Ca2+ ] to reach 50% of maximum shortening, ie Ca2+ sensitivity was lower. This was associated with increased PAD2 expression and α-actin citrullination. TNF-α increased PAD-mediated citrullination which was blocked by pre-treatment with the PAD inhibitor 2-chloroacetamide. CONCLUSION: Using a mouse RA model we found evidence of impaired cardiac contractile function linked to reduced Ca2+ sensitivity, increased expression of PAD2, and citrullination of α-actin, which was triggered by TNF-α. This provides molecular and physiological evidence for acquired cardiomyopathy and a potential mechanism for RA-associated heart failure.


Subject(s)
Arthritis, Experimental , Arthritis, Rheumatoid , Heart Failure , Animals , Mice , Citrullination , Citrulline/metabolism , Protein-Arginine Deiminases/genetics , Protein-Arginine Deiminases/metabolism , Tumor Necrosis Factor-alpha , Disease Models, Animal , Actins , Hydrolases/metabolism , Arthritis, Rheumatoid/metabolism , Arthritis, Experimental/metabolism , Arginine/pharmacology
6.
FASEB J ; 35(12): e22010, 2021 12.
Article in English | MEDLINE | ID: mdl-34724256

ABSTRACT

The hypoxia-inducible nuclear-encoded mitochondrial protein NADH dehydrogenase (ubiquinone) 1 alpha subcomplex, 4-like 2 (NDUFA4L2) has been demonstrated to decrease oxidative phosphorylation and production of reactive oxygen species in neonatal cardiomyocytes, brain tissue and hypoxic domains of cancer cells. Prolonged local hypoxia can negatively affect skeletal muscle size and tissue oxidative capacity. Although skeletal muscle is a mitochondrial rich, oxygen sensitive tissue, the role of NDUFA4L2 in skeletal muscle has not previously been investigated. Here we ectopically expressed NDUFA4L2 in mouse skeletal muscles using adenovirus-mediated expression and in vivo electroporation. Moreover, femoral artery ligation (FAL) was used as a model of peripheral vascular disease to induce hind limb ischemia and muscle damage. Ectopic NDUFA4L2 expression resulted in reduced mitochondrial respiration and reactive oxygen species followed by lowered AMP, ADP, ATP, and NAD+ levels without affecting the overall protein content of the mitochondrial electron transport chain. Furthermore, ectopically expressed NDUFA4L2 caused a ~20% reduction in muscle mass that resulted in weaker muscles. The loss of muscle mass was associated with increased gene expression of atrogenes MurF1 and Mul1, and apoptotic genes caspase 3 and Bax. Finally, we showed that NDUFA4L2 was induced by FAL and that the Ndufa4l2 mRNA expression correlated with the reduced capacity of the muscle to generate force after the ischemic insult. These results show, for the first time, that mitochondrial NDUFA4L2 is a novel regulator of skeletal muscle mass and force. Specifically, induced NDUFA4L2 reduces mitochondrial activity leading to lower levels of important intramuscular metabolites, including adenine nucleotides and NAD+ , which are hallmarks of mitochondrial dysfunction and hence shows that dysfunctional mitochondrial activity may drive muscle wasting.


Subject(s)
Electron Transport Complex I/metabolism , Hypoxia/physiopathology , Mitochondria/metabolism , Muscle, Skeletal/pathology , Muscular Atrophy/pathology , Animals , Cell Proliferation , Electron Transport Complex I/genetics , Female , Mice , Mice, Inbred C57BL , Muscle, Skeletal/metabolism , Muscular Atrophy/metabolism , Reactive Oxygen Species
7.
Front Cell Dev Biol ; 9: 640723, 2021.
Article in English | MEDLINE | ID: mdl-33644077

ABSTRACT

Extracellular vesicles (EVs), which are cell released double layered membrane particles, have been found in every circulating body fluid, and provide a tool for conveying diverse information between cells, influencing both physiological and pathological conditions. Viruses can hijack the EVs secretory pathway to exit infected cells and use EVs endocytic routes to enter uninfected cells, suggesting that EVs and viruses can share common cell entry and biogenesis mechanisms. SARS-CoV-2 is responsible of the coronavirus disease 2019 (Covid-19), which may be accompanied by severe multi-organ manifestations. EVs may contribute to virus spreading via transfer of virus docking receptors such as CD9 and ACE2. Covid-19 is known to affect the renin angiotensin system (RAS), and could promote secretion of harmful EVs. In this scenario EVs might be linked to cardiovascular manifestations of the Covid-19 disease through unbalance in RAS. In contrast EVs derived from mesenchymal stem cells or cardiosphere derived cells, may promote cardiovascular function due to their beneficial effect on angiogenesis, fibrosis, contractility and immuno-modulation. In this article we assessed the potential impact of EVs in cardiovascular manifestations of Covid-19 and highlight potential strategies to control the extracellular signaling for future therapies.

9.
Cardiovasc Res ; 116(3): 619-632, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31382275

ABSTRACT

AIMS: Polycystic ovary syndrome (PCOS) is a common endocrinopathy that is suggested to increase the risk for cardiovascular disease. How PCOS may lead to adverse cardiac outcomes is unclear and here we hypothesized that prenatal exposure to dihydrotestosterone (DHT) and/or maternal obesity in mice induce adverse metabolic and cardiac programming in female offspring that resemble the reproductive features of the syndrome. METHODS AND RESULTS: The maternal obese PCOS phenotype was induced in mice by chronic high-fat-high-sucrose consumption together with prenatal DHT exposure. The prenatally androgenized (PNA) female offspring displayed cardiac hypertrophy during adulthood, an outcome that was not accompanied by aberrant metabolic profile. The expression of key genes involved in cardiac hypertrophy was up-regulated in the PNA offspring, with limited or no impact of maternal obesity. Furthermore, the activity of NADPH oxidase, a major source of reactive oxygen species in the cardiovascular system, was down-regulated in the PNA offspring heart. We next explored for early transcriptional changes in the heart of newly born PNA offspring, which could account for the long-lasting changes observed in adulthood. Neonatal PNA hearts displayed an up-regulation of transcription factors involved in cardiac hypertrophic remodelling and of the calcium-handling gene, Slc8a2. Finally, to determine the specific role of androgens in cardiovascular function, female mice were continuously exposed to DHT from pre-puberty to adulthood, with or without the antiandrogen flutamide. Continuous exposure to DHT led to adverse left ventricular remodelling, and increased vasocontractile responses, while treatment with flutamide partly alleviated these effects. CONCLUSION: Taken together, our results indicate that intrauterine androgen exposure programmes long-lasting heart remodelling in female mouse offspring that is linked to left ventricular hypertrophy and highlight the potential risk of developing cardiac dysfunction in daughters of mothers with PCOS.


Subject(s)
Dihydrotestosterone , Hypertrophy, Left Ventricular/etiology , Polycystic Ovary Syndrome/chemically induced , Ventricular Dysfunction, Left/etiology , Ventricular Function, Left , Ventricular Remodeling , Animals , Diet, High-Fat , Dietary Sucrose , Disease Models, Animal , Energy Metabolism , Female , Gene Expression Regulation , Hypertrophy, Left Ventricular/genetics , Hypertrophy, Left Ventricular/metabolism , Hypertrophy, Left Ventricular/physiopathology , Maternal Exposure , Mesenteric Arteries/metabolism , Mesenteric Arteries/physiopathology , Mice, Inbred C57BL , Myocardium/metabolism , Myocardium/pathology , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Obesity/complications , Pregnancy , Sex Factors , Sexual Development , Sodium-Calcium Exchanger/genetics , Sodium-Calcium Exchanger/metabolism , Ventricular Dysfunction, Left/genetics , Ventricular Dysfunction, Left/metabolism , Ventricular Dysfunction, Left/physiopathology
10.
Heart ; 104(24): 2026-2034, 2018 12.
Article in English | MEDLINE | ID: mdl-29804097

ABSTRACT

OBJECTIVES: Patients with rheumatoid arthritis (RA) display an increased risk of heart failure independent of traditional cardiovascular risk factors. To elucidate myocardial disease in RA, we have investigated molecular and cellular remodelling of the heart in an established mouse model of RA. METHODS: The collagen antibody-induced arthritis (CAIA) RA mouse model is characterised by joint inflammation and increased inflammatory markers in the serum. We used CAIA mice in the postinflammatory phase that resembles medically controlled RA or RA in remission. Hearts were collected for cardiomyocyte isolation, biochemistry and histology analysis. RESULTS: Hearts from mice subjected to CAIA displayed hypertrophy (heart/body weight, mean±SD: 5.9±0.8vs 5.1±0.7 mg/g, p<0.05), fibrosis and reduced left ventricular fractional shortening compared with control. Cardiomyocytes from CAIA mice showed reduced cytosolic [Ca2+]i transient amplitudes (F/F0, mean±SD: 3.0±1.2vs 3.6±1.5, p<0.05) that was linked to reductions in sarcoplasmic reticulum (SR) Ca2+ store (F/F0, mean±SD: 3.5±1.3vs 4.4±1.3, p<0.01) measured with Ca2+ imaging. This was associated to lower fractional shortening in the cardiomyocytes from the CAIA mice (%FS, mean±SD: 3.4±2.2 vs 4.6%±2.3%, p<0.05). Ca2+ handling proteins displayed oxidation-dependent posttranslational modifications that together with an increase in superoxide dismutase expression indicate a cell environment with oxidative stress. CONCLUSIONS: This study shows that inflammation during active RA has long-term consequences on molecular remodelling and contractile function of the heart, which further supports that rheumatology patients should be followed for development of heart failure.


Subject(s)
Arthritis, Experimental/complications , Arthritis, Rheumatoid/complications , Cardiomyopathies/etiology , Myocardial Contraction , Myocytes, Cardiac/metabolism , Oxidative Stress , Ventricular Function, Left , Animals , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Arthritis, Experimental/physiopathology , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Arthritis, Rheumatoid/physiopathology , Calcium Channels, L-Type/metabolism , Calcium Signaling , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Cardiomyopathies/physiopathology , Female , Fibrosis , Male , Mice, Inbred BALB C , Myocytes, Cardiac/pathology , Protein Processing, Post-Translational , Sarcoplasmic Reticulum/metabolism , Ventricular Remodeling
11.
Hypertension ; 71(3): 507-517, 2018 03.
Article in English | MEDLINE | ID: mdl-29335250

ABSTRACT

MitoAKAPs (mitochondrial A kinase anchoring proteins), encoded by the Akap1 gene, regulate multiple cellular processes governing mitochondrial homeostasis and cell viability. Although mitochondrial alterations have been associated to endothelial dysfunction, the role of mitoAKAPs in the vasculature is currently unknown. To test this, postischemic neovascularization, vascular function, and arterial blood pressure were analyzed in Akap1 knockout mice (Akap1-/- ) and their wild-type (wt) littermates. Primary cultures of aortic endothelial cells (ECs) were also obtained from Akap1-/- and wt mice, and ECs migration, proliferation, survival, and capillary-like network formation were analyzed under different experimental conditions. After femoral artery ligation, Akap1-/- mice displayed impaired blood flow and functional recovery, reduced skeletal muscle capillary density, and Akt phosphorylation compared with wt mice. In Akap1-/- ECs, a significant enhancement of hypoxia-induced mitophagy, mitochondrial dysfunction, reactive oxygen species production, and apoptosis were observed. Consistently, capillary-like network formation, migration, proliferation, and AKT phosphorylation were reduced in Akap1-/- ECs. Alterations in Akap1-/- ECs behavior were also confirmed in Akap1-/- mice, which exhibited a selective reduction in acetylcholine-induced vasorelaxation in mesenteric arteries and a mild but significant increase in arterial blood pressure levels compared with wt. Finally, overexpression of a constitutively active Akt mutant restored vascular reactivity and ECs function in Akap1-/- conditions. These results demonstrate the important role of mitoAKAPs in the modulation of multiple ECs functions in vivo and in vitro, suggesting that mitochondria-dependent regulation of ECs might represent a novel therapeutic approach in cardiovascular diseases characterized by endothelial dysfunction.


Subject(s)
A Kinase Anchor Proteins/metabolism , Endothelial Cells/pathology , Mitochondria/pathology , Neovascularization, Pathologic/pathology , Reactive Oxygen Species/metabolism , Analysis of Variance , Animals , Cell Movement/physiology , Cells, Cultured , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Nitric Oxide/metabolism , Phosphorylation , Random Allocation , Reference Values , Risk Factors , Statistics, Nonparametric , Vascular Diseases/pathology , Vascular Diseases/physiopathology
13.
PLoS One ; 11(5): e0154076, 2016.
Article in English | MEDLINE | ID: mdl-27136357

ABSTRACT

A-kinase anchoring proteins (AKAPs) transmit signals cues from seven-transmembrane receptors to specific sub-cellular locations. Mitochondrial AKAPs encoded by the Akap1 gene have been shown to modulate mitochondrial function and reactive oxygen species (ROS) production in the heart. Under conditions of hypoxia, mitochondrial AKAP121 undergoes proteolytic degradation mediated, at least in part, by the E3 ubiquitin ligase Seven In-Absentia Homolog 2 (Siah2). In the present study we hypothesized that Akap1 might be crucial to preserve mitochondrial function and structure, and cardiac responses to myocardial ischemia. To test this, eight-week-old Akap1 knockout mice (Akap1-/-), Siah2 knockout mice (Siah2-/-) or their wild-type (wt) littermates underwent myocardial infarction (MI) by permanent left coronary artery ligation. Age and gender matched mice of either genotype underwent a left thoracotomy without coronary ligation and were used as controls (sham). Twenty-four hours after coronary ligation, Akap1-/- mice displayed larger infarct size compared to Siah2-/- or wt mice. One week after MI, cardiac function and survival were also significantly reduced in Akap1-/- mice, while cardiac fibrosis was significantly increased. Akap1 deletion was associated with remarkable mitochondrial structural abnormalities at electron microscopy, increased ROS production and reduced mitochondrial function after MI. These alterations were associated with enhanced cardiac mitophagy and apoptosis. Autophagy inhibition by 3-methyladenine significantly reduced apoptosis and ameliorated cardiac dysfunction following MI in Akap1-/- mice. These results demonstrate that Akap1 deficiency promotes cardiac mitochondrial aberrations and mitophagy, enhancing infarct size, pathological cardiac remodeling and mortality under ischemic conditions. Thus, mitochondrial AKAPs might represent important players in the development of post-ischemic cardiac remodeling and novel therapeutic targets.


Subject(s)
A Kinase Anchor Proteins/metabolism , Mitochondria/metabolism , A Kinase Anchor Proteins/genetics , Adenine/analogs & derivatives , Adenine/pharmacology , Animals , Apoptosis/drug effects , Blotting, Western , Disease Models, Animal , Echocardiography , In Situ Nick-End Labeling , Mice , Mice, Knockout , Microscopy, Electron , Mitochondria/genetics , Mitochondria/pathology , Mitochondria/ultrastructure , Mitophagy/drug effects , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , Reactive Oxygen Species/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
14.
Basic Res Cardiol ; 111(3): 34, 2016 May.
Article in English | MEDLINE | ID: mdl-27071401

ABSTRACT

The inorganic anion nitrate (NO3 (-)), which is naturally enriched in certain vegetables (e.g., spinach and beetroot), has emerged as a dietary component that can regulate diverse bodily functions, including blood pressure, mitochondrial efficiency, and skeletal muscle force. It is not known if dietary nitrate improves cardiac contractility. To test this, mice were supplemented for 1-2 weeks with sodium nitrate in the drinking water at a dose similar to a green diet. The hearts from nitrate-treated mice showed increased left ventricular pressure and peak rate of pressure development as measured with the Langendorff heart technique. Cardiomyocytes from hearts of nitrate-treated and control animals were incubated with the fluorescent indicator Fluo-3 to measure cytoplasmic free [Ca(2+)] and fractional shortening. Cardiomyocytes from nitrate-treated mice displayed increased fractional shortening, which was linked to larger Ca(2+) transients. Moreover, nitrate hearts displayed increased protein expression of the L-type Ca(2+) channel/dihydropyridine receptor and peak L-type Ca(2+) channel currents. The nitrate-treated hearts displayed increased concentration of cAMP but unchanged levels of cGMP compared with controls. These findings provide the first evidence that dietary nitrate can affect the expression of important Ca(2+) handling proteins in the heart, resulting in increased cardiomyocyte Ca(2+) signaling and improved left ventricular contractile function. Our observation shows that dietary nitrate impacts cardiac function and adds understanding to inorganic nitrate as a physiological modulator.


Subject(s)
Calcium Signaling/physiology , Heart/drug effects , Heart/physiology , Myocardial Contraction/drug effects , Nitrates/pharmacology , Animals , Blotting, Western , Diet , Isolated Heart Preparation , Male , Mice , Mice, Inbred C57BL , Myocardial Contraction/physiology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , Patch-Clamp Techniques
15.
Sci Transl Med ; 7(319): 319ra207, 2015 Dec 23.
Article in English | MEDLINE | ID: mdl-26702095

ABSTRACT

Uncoordinated contraction from electromechanical delay worsens heart failure pathophysiology and prognosis, but restoring coordination with biventricular pacing, known as cardiac resynchronization therapy (CRT), improves both. However, not every patient qualifies for CRT. We show that heart failure with synchronous contraction is improved by inducing dyssynchrony for 6 hours daily by right ventricular pacing using an intracardiac pacing device, in a process we call pacemaker-induced transient asynchrony (PITA). In dogs with heart failure induced by 6 weeks of atrial tachypacing, PITA (starting on week 3) suppressed progressive cardiac dilation as well as chamber and myocyte dysfunction. PITA enhanced ß-adrenergic responsiveness in vivo and normalized it in myocytes. Myofilament calcium response declined in dogs with synchronous heart failure, which was accompanied by sarcomere disarray and generation of myofibers with severely reduced function, and these changes were absent in PITA-treated hearts. The benefits of PITA were not replicated when the same number of right ventricular paced beats was randomly distributed throughout the day, indicating that continuity of dyssynchrony exposure is necessary to trigger the beneficial biological response upon resynchronization. These results suggest that PITA could bring the benefits of CRT to the many heart failure patients with synchronous contraction who are not CRT candidates.


Subject(s)
Disease Progression , Heart Failure/pathology , Heart Failure/therapy , Pacemaker, Artificial , Animals , Calcium/metabolism , Dogs , Heart Failure/physiopathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Myofibrils/metabolism , Proteomics , Receptors, Adrenergic, beta/metabolism , Sarcomeres/metabolism
16.
Cardiovasc Res ; 107(4): 431-41, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26101262

ABSTRACT

AIMS: Coronary artery disease is the leading cause of death in western countries, and its association with lower extremity peripheral artery disease (LE-PAD) represents an independent predictor of worse outcome. However, the molecular mechanisms underlying these effects are currently unknown. METHODS AND RESULTS: To investigate these processes, we used in vitro approaches and several mouse models: (i) unilateral limb ischaemia by left common femoral artery ligation [peripheral ischaemia (PI), n = 38]; (ii) myocardial infarction by permanent ligation of the left descending coronary artery (MI, n = 40); (iii) MI after 5 weeks of limb ischaemia (PI + MI, n = 44); (iv) sham operation (SHAM, n = 20). Compared with MI, PI + MI hearts were characterized by a significant increase in cardiomyocyte apoptosis, larger infarct areas, and decreased cardiac function. By using a proteomic approach, we identified a ≅ 8 kDa circulating peptide, Dermcidin (DCD), secreted by ischaemic skeletal muscles, enhancing cardiomyocytes apoptosis under hypoxic conditions and infarct size after permanent coronary artery ligation. siRNA interference experiments to reduce DCD circulating levels significantly reduced infarct size and ameliorated cardiac function after MI. CONCLUSION: Our data demonstrate that chronic limb ischaemia activates detrimental pathways in the ischaemic heart through humoral mechanisms of remote organ crosstalk. Thus, DCD may represent a novel important myokine modulating cardiomyocyte survival and function.


Subject(s)
Coronary Vessels/surgery , Dermcidins/metabolism , Muscle, Skeletal/blood supply , Myocardial Infarction/surgery , Myocytes, Cardiac/metabolism , Animals , Disease Models, Animal , Ligation/methods , Mice , Muscle, Skeletal/surgery , Myocardial Infarction/metabolism , Myocardial Ischemia/metabolism
17.
Circulation ; 131(24): 2120-30, 2015 Jun 16.
Article in English | MEDLINE | ID: mdl-25995315

ABSTRACT

BACKGROUND: Whether biomechanical force on the heart can induce exosome secretion to modulate cardiovascular function is not known. We investigated the secretion and activity of exosomes containing a key receptor in cardiovascular function, the angiotensin II type I receptor (AT1R). METHODS AND RESULTS: Exosomes containing AT1Rs were isolated from the media overlying AT1R-overexpressing cells exposed to osmotic stretch and from sera of mice undergoing cardiac pressure overload. The presence of AT1Rs in exosomes was confirmed by both electron microscopy and radioligand receptor binding assays and shown to require ß-arrestin2, a multifunctional adaptor protein essential for receptor trafficking. We show that functional AT1Rs are transferred via exosomes in an in vitro model of cellular stretch. Using mice with global and cardiomyocyte conditional deletion of ß-arrestin2, we show that under conditions of in vivo pressure overload the cellular source of the exocytosis of exosomes containing AT1R is the cardiomyocyte. Exogenously administered AT1R-enriched exosomes target cardiomyocytes, skeletal myocytes, and mesenteric resistance vessels and are sufficient to confer blood pressure responsiveness to angiotensin II infusion in AT1R knockout mice. CONCLUSIONS: AT1R-enriched exosomes are released from the heart under conditions of in vivo cellular stress to likely modulate vascular responses to neurohormonal stimulation. In the context of the whole organism, the concept of G protein-coupled receptor trafficking should consider circulating exosomes as part of the reservoir of functional AT1Rs.


Subject(s)
Exosomes/chemistry , Myocytes, Cardiac/chemistry , Receptor, Angiotensin, Type 1/blood , Stress, Mechanical , Animals , Arrestins/deficiency , Arrestins/genetics , Arrestins/physiology , Blood Pressure , Constriction , Exosomes/physiology , Female , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron , Muscle Cells/metabolism , Myocytes, Cardiac/ultrastructure , Osmotic Pressure , Protein Transport , RNA Interference , RNA, Small Interfering/pharmacology , Radioligand Assay , Receptor, Angiotensin, Type 1/deficiency , Receptor, Angiotensin, Type 1/genetics , Vascular Resistance , beta-Arrestins
18.
PLoS One ; 9(9): e106024, 2014.
Article in English | MEDLINE | ID: mdl-25181347

ABSTRACT

Re-induction of fetal genes and/or re-expression of postnatal genes represent hallmarks of pathological cardiac remodeling, and are considered important in the progression of the normal heart towards heart failure (HF). Whether epigenetic modifications are involved in these processes is currently under investigation. Here we hypothesized that histone chromatin modifications may underlie changes in the gene expression program during pressure overload-induced HF. We evaluated chromatin marks at the promoter regions of the sarcoplasmic reticulum Ca2+ATPase (SERCA-2A) and ß-myosin-heavy chain (ß-MHC) genes (Atp2a2 and Myh7, respectively) in murine hearts after one or eight weeks of pressure overload induced by transverse aortic constriction (TAC). As expected, all TAC hearts displayed a significant reduction in SERCA-2A and a significant induction of ß-MHC mRNA levels. Interestingly, opposite histone H3 modifications were identified in the promoter regions of these genes after TAC, including H3 dimethylation (me2) at lysine (K) 4 (H3K4me2) and K9 (H3K9me2), H3 trimethylation (me3) at K27 (H3K27me3) and dimethylation (me2) at K36 (H3K36me2). Consistently, a significant reduction of lysine-specific demethylase KDM2A could be found after eight weeks of TAC at the Atp2a2 promoter. Moreover, opposite changes in the recruitment of DNA methylation machinery components (DNA methyltransferases DNMT1 and DNMT3b, and methyl CpG binding protein 2 MeCp2) were found at the Atp2a2 or Myh7 promoters after TAC. Taken together, these results suggest that epigenetic modifications may underlie gene expression reprogramming in the adult murine heart under conditions of pressure overload, and might be involved in the progression of the normal heart towards HF.


Subject(s)
Epigenesis, Genetic , Heart Failure/genetics , Myosin Heavy Chains/genetics , Pressure , Promoter Regions, Genetic , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Animals , Chromatin/metabolism , Gene Expression Profiling , Histones/metabolism , Jumonji Domain-Containing Histone Demethylases/genetics , Lysine/metabolism , Male , Mice, Inbred C57BL , Myosin Heavy Chains/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics
19.
Circ Res ; 115(2): 273-83, 2014 Jul 07.
Article in English | MEDLINE | ID: mdl-24807785

ABSTRACT

RATIONALE: The sympathetic nervous system plays a fundamental role in the regulation of myocardial function. During chronic pressure overload, overactivation of the sympathetic nervous system induces the release of catecholamines, which activate ß-adrenergic receptors in cardiomyocytes and lead to increased heart rate and cardiac contractility. However, chronic stimulation of ß-adrenergic receptors leads to impaired cardiac function, and ß-blockers are widely used as therapeutic agents for the treatment of cardiac disease. MicroRNA-133 (miR-133) is highly expressed in the myocardium and is involved in controlling cardiac function through regulation of messenger RNA translation/stability. OBJECTIVE: To determine whether miR-133 affects ß-adrenergic receptor signaling during progression to heart failure. METHODS AND RESULTS: Based on bioinformatic analysis, ß1-adrenergic receptor (ß1AR) and other components of the ß1AR signal transduction cascade, including adenylate cyclase VI and the catalytic subunit of the cAMP-dependent protein kinase A, were predicted as direct targets of miR-133 and subsequently validated by experimental studies. Consistently, cAMP accumulation and activation of downstream targets were repressed by miR-133 overexpression in both neonatal and adult cardiomyocytes following selective ß1AR stimulation. Furthermore, gain-of-function and loss-of-function studies of miR-133 revealed its role in counteracting the deleterious apoptotic effects caused by chronic ß1AR stimulation. This was confirmed in vivo using a novel cardiac-specific TetON-miR-133 inducible transgenic mouse model. When subjected to transaortic constriction, TetON-miR-133 inducible transgenic mice maintained cardiac performance and showed attenuated apoptosis and reduced fibrosis compared with control mice. CONCLUSIONS: miR-133 controls multiple components of the ß1AR transduction cascade and is cardioprotective during heart failure.


Subject(s)
Cyclic AMP/physiology , MicroRNAs/physiology , Myocytes, Cardiac/physiology , Receptors, Adrenergic, beta-1/physiology , Second Messenger Systems/physiology , 3' Untranslated Regions/physiology , Adenylyl Cyclases/physiology , Animals , Apoptosis , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/physiology , Disease Progression , Gene Expression Regulation/drug effects , Genes, Reporter , Guanine Nucleotide Exchange Factors/physiology , Male , Metoprolol/pharmacology , Metoprolol/therapeutic use , Mice , Mice, Inbred C57BL , Mice, Transgenic , MicroRNAs/genetics , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/drug effects , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/genetics
20.
Circ Res ; 114(5): 833-44, 2014 Feb 28.
Article in English | MEDLINE | ID: mdl-24334028

ABSTRACT

RATIONALE: MicroRNAs (miRs) are small, noncoding RNAs that function to post-transcriptionally regulate gene expression. First transcribed as long primary miR transcripts (pri-miRs), they are enzymatically processed in the nucleus by Drosha into hairpin intermediate miRs (pre-miRs) and further processed in the cytoplasm by Dicer into mature miRs where they regulate cellular processes after activation by a variety of signals such as those stimulated by ß-adrenergic receptors (ßARs). Initially discovered to desensitize ßAR signaling, ß-arrestins are now appreciated to transduce multiple effector pathways independent of G-protein-mediated second messenger accumulation, a concept known as biased signaling. We previously showed that the ß-arrestin-biased ßAR agonist, carvedilol, activates cellular pathways in the heart. OBJECTIVE: Here, we tested whether carvedilol could activate ß-arrestin-mediated miR maturation, thereby providing a novel potential mechanism for its cardioprotective effects. METHODS AND RESULTS: In human cells and mouse hearts, carvedilol upregulates a subset of mature and pre-miRs, but not their pri-miRs, in ß1AR-, G-protein-coupled receptor kinase 5/6-, and ß-arrestin1-dependent manner. Mechanistically, ß-arrestin1 regulates miR processing by forming a nuclear complex with hnRNPA1 and Drosha on pri-miRs. CONCLUSIONS: Our findings indicate a novel function for ß1AR-mediated ß-arrestin1 signaling activated by carvedilol in miR biogenesis, which may be linked, in part, to its mechanism for cell survival.


Subject(s)
Arrestins/metabolism , MicroRNAs/genetics , Receptors, Adrenergic, beta-1/metabolism , Signal Transduction/physiology , Adrenergic beta-1 Receptor Agonists/pharmacology , Animals , Arrestins/genetics , Carbazoles/pharmacology , Carvedilol , G-Protein-Coupled Receptor Kinase 5/metabolism , G-Protein-Coupled Receptor Kinases/metabolism , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/metabolism , Propanolamines/pharmacology , RNA Processing, Post-Transcriptional/physiology , Receptors, Adrenergic, beta-1/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , beta-Arrestins
SELECTION OF CITATIONS
SEARCH DETAIL
...