Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Front Cardiovasc Med ; 9: 854361, 2022.
Article in English | MEDLINE | ID: mdl-35360022

ABSTRACT

Background: Activation of the renin-angiotensin-aldosterone system (RAAS) plays a critical role in the development of hypertension. Published evidence on a putative "memory effect" of AngII on the vascular components is however scarce. Aim: To evaluate the long-term effects of transient exposure to AngII on the mouse heart and the arterial tissue. Methods: Blood pressure, cardiovascular tissue damage and remodeling, and systemic oxidative stress were evaluated in C57/B6/J mice at the end of a 2-week AngII infusion (AngII); 2 and 3 weeks after the interruption of a 2-week AngII treatment (AngII+2W and AngII +3W; so-called "memory" conditions) and control littermate (CTRL). RNAseq profiling of aortic tissues was used to identify potential key regulated genes accounting for legacy effects on the vascular phenotype. RNAseq results were validated by RT-qPCR and immunohistochemistry in a reproduction cohort of mice. Key findings were reproduced in a homotypic cell culture model. Results: The 2 weeks AngII infusion induced cardiac hypertrophy and aortic damage that persisted beyond AngII interruption and despite blood pressure normalization, with a sustained vascular expression of ICAM1, infiltration by CD45+ cells, and cell proliferation associated with systemic oxidative stress. RNAseq profiling in aortic tissue identified robust Acta2 downregulation at transcript and protein levels (α-smooth muscle actin) that was maintained beyond interruption of AngII treatment. Among regulators of Acta2 expression, the transcription factor Myocardin (Myocd), exhibited a similar expression pattern. The sustained downregulation of Acta2 and Myocd was associated with an increase in H3K27me3 in nuclei of aortic sections from mice in the "memory" conditions. A sustained downregulation of ACTA2 and MYOCD was reproduced in the cultured human aortic vascular smooth muscle cells upon transient exposure to Ang II. Conclusion: A transient exposure to Ang II produces prolonged vascular remodeling with robust ACTA2 downregulation, associated with epigenetic imprinting supporting a "memory" effect despite stimulus withdrawal.

2.
Cardiovasc Drugs Ther ; 36(5): 793-803, 2022 10.
Article in English | MEDLINE | ID: mdl-34138361

ABSTRACT

CONTEXT: Duchenne muscular dystrophy (DMD) is associated with a progressive alteration in cardiac function. OBJECTIVE: The aim of this study was to detect early cardiac dysfunction using the high sensitive two-dimensional speckle-tracking echocardiography (2D strain) in mdx mouse model and to investigate the potential preventive effects of the S107 ryanodine receptor (RyR2) stabilizer on early onset of DMD-related cardiomyopathy. METHODS AND RESULTS: Conventional echocardiography and global and segmental left ventricle (LV) 2D strains were assessed in male mdx mice and control C57/BL10 mice from 2 to 12 months of age. Up to 12 months of age, mdx mice showed preserved myocardial function as assessed by conventional echocardiography. However, global longitudinal, radial, and circumferential LV 2D strains significantly declined in mdx mice compared to controls from the 9 months of age. Segmental 2D strain analysis found a predominant alteration in posterior, inferior, and lateral LV segments, with a more marked impairment with aging. Then, mdx mice were treated with S107 in the drinking water at a dose of 250 mg/L using two different protocols: earlier therapy from 2 to 6 months of age and later therapy from 6 to 9 months of age. The treatment with S107 was efficient only when administered earlier in very young animals (from 2 to 6 months of age) and prevented the segmental alterations seen in non-treated mdx mice. CONCLUSIONS: This is the first animal study to evaluate the therapeutic effect of a drug targeting early onset of DMD-related cardiomyopathy, using 2D strain echocardiography. Speckle-tracking analyses revealed early alterations of LV posterior segments that could be prevented by 4 months of RyR2 stabilization.


Subject(s)
Cardiomyopathies , Drinking Water , Muscular Dystrophy, Duchenne , Animals , Cardiomyopathies/diagnostic imaging , Cardiomyopathies/drug therapy , Cardiomyopathies/etiology , Male , Mice , Mice, Inbred mdx , Muscular Dystrophy, Duchenne/complications , Muscular Dystrophy, Duchenne/diagnosis , Muscular Dystrophy, Duchenne/drug therapy , Ryanodine Receptor Calcium Release Channel
3.
Otol Neurotol ; 42(6): e779-e787, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33871251

ABSTRACT

INTRODUCTION: There remains no standard imaging method that allows computer-assisted surgery of the cochlea in real time. However, recent evidence suggests that high-frequency ultrasound (HFUS) could permit real-time visualization of cochlear architecture. Registration with an imaging modality that suffers neither attenuation nor conical deformation could reveal useful anatomical landmarks to surgeons. Our study aimed to address the feasibility of an automated three-dimensional (3D) HFUS/microCT registration, and to evaluate the identification of cochlear structures using 2D/3D HFUS and microCT. METHODS: MicroCT, and 2D/3D 40 MHz US in B-mode were performed on ex vivo guinea pig cochlea. An automatic rigid registration algorithm was applied to segmented 3D images. This automatic registration was then compared to a reference method using manual annotated landmarks placed by two senior otologists. Inter- and intrarater reliabilities were evaluated using intraclass correlation coefficient (ICC) and the mean registration error was calculated. RESULTS: 3D HFUS/microCT automatic registration was successful. Excellent levels of concordance were achieved with regards intra-rater reliability for both raters with micro-CT and US images (ICC ranging from 0.98 to 1, p < 0.001) and with regards inter-rater reliability (ICC ranging from 0.99 to 1, p < 0.001). The mean HFUS/microCT automated RE for both observers was 0.17 ±â€Š0.03 mm [0.10-0.25]. Identification of the basilar membrane, modiolus, scala tympani, and scala vestibuli was possible with 2D/3D HFUS and micro-CT. CONCLUSIONS: HFUS/microCT image registration is feasible. 2D/3D HFUS and microCT allow the visualization of cochlear structures. Many potential clinical applications are conceivable.


Subject(s)
Cochlea , Surgery, Computer-Assisted , Algorithms , Animals , Cochlea/diagnostic imaging , Cochlea/surgery , Feasibility Studies , Guinea Pigs , Imaging, Three-Dimensional , Reproducibility of Results , X-Ray Microtomography
4.
Cells ; 9(12)2020 12 02.
Article in English | MEDLINE | ID: mdl-33276630

ABSTRACT

The third isotype of beta-adrenoreceptors (ß3-AR) has recently come (back) into focus after the observation of its expression in white and beige human adipocytes and its implication in metabolic regulation. This coincides with the recent development and marketing of agonists at the human receptor with superior specificity. Twenty years ago, however, we and others described the expression of ß3-AR in human myocardium and its regulation of contractility and cardiac remodeling. Subsequent work from many laboratories has since expanded the characterization of ß3-AR involvement in many aspects of cardiovascular physio(patho)logy, justifying the present effort to update current paradigms under the light of the most recent evidence.


Subject(s)
Adipocytes, Beige/metabolism , Gene Expression Regulation , Myocardium/metabolism , Receptors, Adrenergic, beta-3/chemistry , Receptors, Adrenergic, beta-3/metabolism , Animals , Antioxidants/metabolism , Cardiovascular Diseases/metabolism , Cardiovascular System/metabolism , Catecholamines/metabolism , Gene Expression Profiling , Heart Failure/metabolism , Humans , Protein Isoforms , Signal Transduction
5.
Sci Transl Med ; 12(564)2020 10 07.
Article in English | MEDLINE | ID: mdl-33028705

ABSTRACT

Pathological remodeling of the myocardium has long been known to involve oxidant signaling, but strategies using systemic antioxidants have generally failed to prevent it. We sought to identify key regulators of oxidant-mediated cardiac hypertrophy amenable to targeted pharmacological therapy. Specific isoforms of the aquaporin water channels have been implicated in oxidant sensing, but their role in heart muscle is unknown. RNA sequencing from human cardiac myocytes revealed that the archetypal AQP1 is a major isoform. AQP1 expression correlates with the severity of hypertrophic remodeling in patients with aortic stenosis. The AQP1 channel was detected at the plasma membrane of human and mouse cardiac myocytes from hypertrophic hearts, where it colocalized with NADPH oxidase-2 and caveolin-3. We show that hydrogen peroxide (H2O2), produced extracellularly, is necessary for the hypertrophic response of isolated cardiac myocytes and that AQP1 facilitates the transmembrane transport of H2O2 through its water pore, resulting in activation of oxidant-sensitive kinases in cardiac myocytes. Structural analysis of the amino acid residues lining the water pore of AQP1 supports its permeation by H2O2 Deletion of Aqp1 or selective blockade of the AQP1 intrasubunit pore inhibited H2O2 transport in mouse and human cells and rescued the myocyte hypertrophy in human induced pluripotent stem cell-derived engineered heart muscle. Treatment of mice with a clinically approved AQP1 inhibitor, Bacopaside, attenuated cardiac hypertrophy. We conclude that cardiac hypertrophy is mediated by the transmembrane transport of H2O2 by the water channel AQP1 and that inhibitors of AQP1 represent new possibilities for treating hypertrophic cardiomyopathies.


Subject(s)
Aquaporin 1 , Induced Pluripotent Stem Cells , Animals , Humans , Hydrogen Peroxide/metabolism , Induced Pluripotent Stem Cells/metabolism , Mice , Myocardium/metabolism , Myocytes, Cardiac/metabolism
6.
Theranostics ; 10(18): 8130-8142, 2020.
Article in English | MEDLINE | ID: mdl-32724462

ABSTRACT

Anthracyclines are key chemotherapeutic agents used in various adult and pediatric cancers, however, their clinical use is limited due to possible congestive heart failure (HF) caused by acute and irreversible cardiotoxicity. Currently, there is no method to predict the future development of the HF in these patients. In order to identify early biomarkers to predict anthracycline cardiotoxicity in long-term survivors of childhood cancer, this longitudinal study aimed to analyze early and late in-vivo regional myocardial anthracycline-induced cardiotoxicity, related to in-vitro cardiac myocytes dysfunction, in a juvenile rat model. Methods: Young male Wistar rats (4 weeks-old) were treated with different cumulative doses of doxorubicin (7.5, 10 or 12.5 mg/kg) or NaCl (0.9%) once a week for 6 weeks by intravenous injection. Cardiac function was evaluated in-vivo by conventional (left ventricular ejection fraction, LVEF) and regional two-dimensional (2D) speckle tracking echocardiography over the 4 months after the last injection. The animals were assigned to preserved (pEF) or reduced EF (rEF) groups at the end of the protocol and were compared to controls. Results: We observed a preferential contractile dysfunction of the base of the heart, further altered in the posterior segment, even in pEF group. The first regional alterations appeared 1 month after chemotherapy. Functional investigation of cardiomyocytes isolated from the LV base 1 month after doxorubicin treatment showed that early in-vivo contractile alterations were associated with both decreased myofilament Ca2+ sensitivity and length-dependent activation. Changes in post-translational modifications (phosphorylation; S-glutathionylation) and protein degradation of the cardiac myosin binding protein-C may contribute to these alterations. Conclusion: Our data suggest that screening of the contractile defaults of the base of the heart by regional 2D strain echocardiography is useful to detect subclinical myocardial dysfunction prior to the development of delayed anthracycline-induced cardiomyopathy in pediatric onco-cardiology.


Subject(s)
Antibiotics, Antineoplastic/adverse effects , Cardiotoxicity/diagnosis , Doxorubicin/adverse effects , Myocardial Contraction/drug effects , Neoplasms/drug therapy , Animals , Antibiotics, Antineoplastic/administration & dosage , Cancer Survivors , Cardiotoxicity/etiology , Disease Models, Animal , Doxorubicin/administration & dosage , Echocardiography , Heart Ventricles/diagnostic imaging , Heart Ventricles/drug effects , Humans , Injections, Intravenous , Longitudinal Studies , Male , Myocardial Contraction/physiology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , Rats , Rats, Wistar , Stroke Volume/drug effects , Ventricular Function, Left/drug effects
7.
Br J Pharmacol ; 177(19): 4448-4463, 2020 10.
Article in English | MEDLINE | ID: mdl-32608017

ABSTRACT

BACKGROUND AND PURPOSE: The artemisinin derivative, artemether, has antimalarial activity with potential neurotoxic and cardiotoxic effects. Artemether in nanocapsules (NC-ATM) is more efficient than free artemether for reducing parasitaemia and increasing survival of Plasmodium berghei-infected mice. NCs also prevent prolongation of the QT interval of the ECG. Here, we assessed cellular cardiotoxicity of artemether and how this toxicity was prevented by nanoencapsulation. EXPERIMENTAL APPROACH: Mice were treated with NC-ATM orally (120 mg·kg-1 twice daily) for 4 days. Other mice received free artemether, blank NCs, and vehicle for comparison. We measured single-cell contraction, intracellular Ca2+ transient using fluorescent Indo-1AM Ca2+ dye, and electrical activity using the patch-clamp technique in freshly isolated left ventricular myocytes. The acute effect of free artemether was also tested on cardiomyocytes of untreated animals. KEY RESULTS: Artemether prolonged action potentials (AP) upon acute exposure (at 0.1, 1, and 10 µM) of cardiomyocytes from untreated mice or after in vivo treatment. This prolongation was unrelated to blockade of K+ currents, increased Ca2+ currents or promotion of a sustained Na+ current. AP lengthening was abolished by the NCX inhibitor SEA-0400. Artemether promoted irregular Ca2+ transients during pacing and spontaneous Ca2+ events during resting periods. NC-ATM prevented all effects. Blank NCs had no effects compared with vehicle. CONCLUSION AND IMPLICATIONS: Artemether induced NCX-dependent AP lengthening (explaining QTc prolongation) and disrupted Ca2+ handling, both effects increasing pro-arrhythmogenic risks. NCs prevented these adverse effects, providing a safe alternative to the use of artemether alone, especially to treat malaria.


Subject(s)
Calcium , Myocytes, Cardiac , Action Potentials , Animals , Arrhythmias, Cardiac , Artemether , Calcium/metabolism , Mice , Myocytes, Cardiac/metabolism , Patch-Clamp Techniques , Sodium-Calcium Exchanger
8.
Biochim Biophys Acta Mol Basis Dis ; 1865(1): 230-242, 2019 01.
Article in English | MEDLINE | ID: mdl-30463691

ABSTRACT

Heart failure with preserved ejection fraction (HFpEF) is a common clinical syndrome associated with high morbidity and mortality. Therapeutic options are limited due to a lack of knowledge of the pathology and its evolution. We investigated the cellular phenotype and Ca2+ handling in hearts recapitulating HFpEF criteria. HFpEF was induced in a portion of male Wistar rats four weeks after abdominal aortic banding. These animals had nearly normal ejection fraction and presented elevated blood pressure, lung congestion, concentric hypertrophy, increased LV mass, wall stiffness, impaired active relaxation and passive filling of the left ventricle, enlarged left atrium, and cardiomyocyte hypertrophy. Left ventricular cell contraction was stronger and the Ca2+ transient larger. Ca2+ cycling was modified with a RyR2 mediated Ca2+ leak from the sarcoplasmic reticulum and impaired Ca2+ extrusion through the Sodium/Calcium exchanger (NCX), which promoted an increase in diastolic Ca2+. The Sarcoplasmic/endoplasmic reticulum Ca2+ ATPase (SERCA2a) and NCX protein levels were unchanged. The phospholamban (PLN) to SERCA2a ratio was augmented in favor of an inhibitory effect on the SERCA2a activity. Conversely, PLN phosphorylation at the calmodulin-dependent kinase II (CaMKII)-specific site (PLN-Thr17), which promotes SERCA2A activity, was increased as well, suggesting an adaptive compensation of Ca2+ cycling. Altogether our findings show that cardiac remodeling in hearts with a HFpEF status differs from that known for heart failure with reduced ejection fraction. These data also underscore the interdependence between systolic and diastolic "adaptations" of Ca2+ cycling with complex compensative interactions between Ca2+ handling partner and regulatory proteins.


Subject(s)
Calcium/metabolism , Heart Failure/metabolism , Myocytes, Cardiac/metabolism , Stroke Volume , Animals , Calcium-Binding Proteins/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Disease Models, Animal , Heart Ventricles/metabolism , Homeodomain Proteins/metabolism , Hypertension/metabolism , Male , Rats , Rats, Wistar , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcoplasmic Reticulum/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases , Sodium-Calcium Exchanger/metabolism , Ventricular Dysfunction, Left/metabolism
9.
Nat Rev Cardiol ; 15(5): 292-316, 2018 05.
Article in English | MEDLINE | ID: mdl-29388567

ABSTRACT

Nitric oxide (NO) signalling has pleiotropic roles in biology and a crucial function in cardiovascular homeostasis. Tremendous knowledge has been accumulated on the mechanisms of the nitric oxide synthase (NOS)-NO pathway, but how this highly reactive, free radical gas signals to specific targets for precise regulation of cardiovascular function remains the focus of much intense research. In this Review, we summarize the updated paradigms on NOS regulation, NO interaction with reactive oxidant species in specific subcellular compartments, and downstream effects of NO in target cardiovascular tissues, while emphasizing the latest developments of molecular tools and biomarkers to modulate and monitor NO production and bioavailability.


Subject(s)
Cardiovascular Diseases/metabolism , Cardiovascular System/metabolism , Nitric Oxide Synthase/metabolism , Nitric Oxide/metabolism , Signal Transduction , Adenylyl Cyclases/metabolism , Adrenergic beta-3 Receptor Agonists/therapeutic use , Animals , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/physiopathology , Cardiovascular System/drug effects , Cardiovascular System/physiopathology , Enzyme Activation , Enzyme Activators/therapeutic use , Humans , Nitric Oxide Donors/therapeutic use , Phosphodiesterase 5 Inhibitors/therapeutic use , Reactive Oxygen Species/metabolism , Receptors, Adrenergic, beta-3/drug effects , Receptors, Adrenergic, beta-3/metabolism , Signal Transduction/drug effects
10.
Cardiovasc Res ; 114(5): 643-644, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29438492
11.
Sci Rep ; 7: 46620, 2017 04 20.
Article in English | MEDLINE | ID: mdl-28425489

ABSTRACT

Restless Legs Syndrome (RLS) is a genetically complex neurological disorder in which overlapping genetic risk factors may contribute to the diversity and heterogeneity of the symptoms. The main goal of the study was to investigate, through analysis of heart rate variability (HRV), whether in RLS patients the MEIS1 polymorphism at risk influences the sympathovagal regulation in different sleep stages. Sixty-four RLS patients with periodic leg movement index above 15 per hour, and 38 controls underwent one night of video-polysomnographic recording. HRV in the frequency- and time- domains was analyzed during nighttime sleep. All RLS patients were genotyped, and homozygotes for rs2300478 in the MEIS1 locus were used for further analysis. Comparison of the sympathovagal pattern of RLS patients to control subjects did not show significant differences after adjustments for confounding factors in frequency-domain analyses, but showed an increased variability during N2 and N3 stages in time-domain analyses in RLS patients. Sorting of RLS patients according to MEIS1 polymorphism reconfirmed the association between MEIS1 and PLMS, and showed a significant increased sympathovagal balance during N3 stage in those homozygotes for the risk allele. RLS patients should be considered differently depending on MEIS1 genotype, some being potentially at risk for cardiovascular disorders.


Subject(s)
Myeloid Ecotropic Viral Integration Site 1 Protein/genetics , Polymorphism, Single Nucleotide , Restless Legs Syndrome/genetics , Restless Legs Syndrome/physiopathology , Sympathetic Nervous System/physiopathology , Vagus Nerve/physiopathology , Adult , Aged , Female , Genotype , Heart Rate/physiology , Humans , Leg/physiopathology , Male , Middle Aged , Movement , Polysomnography , Sleep Stages/physiology
12.
Sci Rep ; 7: 44998, 2017 03 28.
Article in English | MEDLINE | ID: mdl-28349937

ABSTRACT

Chagas disease is a neglected parasitic disease caused by the protozoan Trypanosoma cruzi. New antitrypanosomal options are desirable to prevent complications, including a high rate of cardiomyopathy. Recently, a natural substance, lychnopholide, has shown therapeutic potential, especially when encapsulated in biodegradable polymeric nanocapsules. However, little is known regarding possible adverse effects of lychnopholide. Here we show that repeated-dose intravenous administration of free lychnopholide (2.0 mg/kg/day) for 20 days caused cardiopathy and mortality in healthy C57BL/6 mice. Echocardiography revealed concentric left ventricular hypertrophy with preserved ejection fraction, diastolic dysfunction and chamber dilatation at end-stage. Single cardiomyocytes presented altered contractility and Ca2+ handling, with spontaneous Ca2+ waves in diastole. Acute in vitro lychnopholide application on cardiomyocytes from healthy mice also induced Ca2+ handling alterations with abnormal RyR2-mediated diastolic Ca2+ release. Strikingly, the encapsulation of lychnopholide prevented the cardiac alterations induced in vivo by the free form repeated doses. Nanocapsules alone had no adverse cardiac effects. Altogether, our data establish lychnopholide presented in nanocapsule form more firmly as a promising new drug candidate to cure Chagas disease with minimal cardiotoxicity. Our study also highlights the potential of nanotechnology not only to improve the efficacy of a drug but also to protect against its adverse effects.


Subject(s)
Biocompatible Materials , Cardiotonic Agents/pharmacology , Lactones/adverse effects , Nanocapsules , Polymers , Sesquiterpenes/adverse effects , Trypanocidal Agents/adverse effects , Trypanosoma cruzi/drug effects , Animals , Biocompatible Materials/chemistry , Calcium/metabolism , Calcium Signaling/drug effects , Cardiotonic Agents/chemistry , Cardiotoxicity , Chagas Disease/diagnosis , Chagas Disease/drug therapy , Chagas Disease/mortality , Chagas Disease/parasitology , Echocardiography , Male , Mice , Molecular Imaging , Mortality , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Nanocapsules/chemistry , Polymers/chemistry
13.
Free Radic Biol Med ; 102: 229-239, 2017 01.
Article in English | MEDLINE | ID: mdl-27932075

ABSTRACT

Acute myocardial infarction leads to an increase in oxidative stress and lipid peroxidation. 4(RS)-4-F4t-Neuroprostane (4-F4t-NeuroP) is a mediator produced by non-enzymatic free radical peroxidation of the cardioprotective polyunsaturated fatty acid, docosahexaenoic acid (DHA). In this study, we investigated whether intra-cardiac delivery of 4-F4t-NeuroP (0.03mg/kg) prior to occlusion (ischemia) prevents and protects rat myocardium from reperfusion damages. Using a rat model of ischemic-reperfusion (I/R), we showed that intra-cardiac infusion of 4-F4t-NeuroP significantly decreased infarct size following reperfusion (-27%) and also reduced ventricular arrhythmia score considerably during reperfusion (-41%). Most notably, 4-F4t-NeuroP decreased ventricular tachycardia and post-reperfusion lengthening of QT interval. The evaluation of the mitochondrial homeostasis indicates a limitation of mitochondrial swelling in response to Ca2+ by decreasing the mitochondrial permeability transition pore opening and increasing mitochondria membrane potential. On the other hand, mitochondrial respiration measured by oxygraphy, and mitochondrial ROS production measured with MitoSox red® were unchanged. We found decreased cytochrome c release and caspase 3 activity, indicating that 4-F4t-NeuroP prevented reperfusion damages and reduced apoptosis. In conclusion, 4-F4t-NeuroP derived from DHA was able to protect I/R cardiac injuries by regulating the mitochondrial homeostasis.


Subject(s)
Docosahexaenoic Acids/administration & dosage , Mitochondria, Heart/drug effects , Myocardial Infarction/metabolism , Myocardial Reperfusion Injury/drug therapy , Neuroprostanes/administration & dosage , Animals , Docosahexaenoic Acids/metabolism , Heart/drug effects , Heart/physiopathology , Humans , Lipid Peroxidation/genetics , Mitochondria, Heart/metabolism , Mitochondria, Heart/pathology , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardium/metabolism , Myocardium/pathology , Oxidative Stress/genetics , Protective Agents/administration & dosage , Rats , Reactive Oxygen Species/metabolism , Tachycardia, Ventricular/drug therapy , Tachycardia, Ventricular/metabolism , Tachycardia, Ventricular/pathology
15.
Elife ; 52016 Feb 08.
Article in English | MEDLINE | ID: mdl-26857994

ABSTRACT

Although cardio-vascular incidents and sudden cardiac death (SCD) are among the leading causes of premature death in the general population, the origins remain unidentified in many cases. Genome-wide association studies have identified Meis1 as a risk factor for SCD. We report that Meis1 inactivation in the mouse neural crest leads to an altered sympatho-vagal regulation of cardiac rhythmicity in adults characterized by a chronotropic incompetence and cardiac conduction defects, thus increasing the susceptibility to SCD. We demonstrated that Meis1 is a major regulator of sympathetic target-field innervation and that Meis1 deficient sympathetic neurons die by apoptosis from early embryonic stages to perinatal stages. In addition, we showed that Meis1 regulates the transcription of key molecules necessary for the endosomal machinery. Accordingly, the traffic of Rab5(+) endosomes is severely altered in Meis1-inactivated sympathetic neurons. These results suggest that Meis1 interacts with various trophic factors signaling pathways during postmitotic neurons differentiation.


Subject(s)
Autonomic Nervous System Diseases/genetics , Death, Sudden, Cardiac , Genetic Predisposition to Disease , Neoplasm Proteins/deficiency , Animals , Apoptosis , Autonomic Nervous System Diseases/pathology , Endosomes/metabolism , Gene Silencing , Homeodomain Proteins , Mice , Myeloid Ecotropic Viral Integration Site 1 Protein
16.
Heart Rhythm ; 13(1): 165-74, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26190315

ABSTRACT

BACKGROUND: Short QT syndrome is associated with an increased risk of cardiac arrhythmias and unexpected sudden death. Until now, only mutations in genes encoding the cardiac potassium and calcium channels have been implicated in early T-wave repolarization. OBJECTIVE: The purpose of this study was to confirm a relationship between a short QT syndrome and carnitine deficiency. METHODS: We report 3 patients affected by primary systemic carnitine deficiency and an associated short QT syndrome. Ventricular fibrillation during early adulthood was the initial symptom in 1 case. To confirm the relationship between carnitine, short QT syndrome, and arrhythmias, we used a mouse model of carnitine deficiency induced by long-term subcutaneous perfusion of MET88. RESULTS: MET88-treated mice developed cardiac hypertrophy associated with a remodeling of the mitochondrial network. The continuous monitoring of electrocardiograms confirmed a shortening of the QT interval, which was negatively correlated with the plasma carnitine concentration. As in humans, such alterations coincided with the genesis of ventricular premature beats and ventricular tachycardia and fibrillation. CONCLUSION: Altogether, these results suggest that long-chain fatty acid metabolism influence the morphology and the electrical function of the heart.


Subject(s)
Arrhythmias, Cardiac , Cardiomyopathies , Carnitine/deficiency , Hyperammonemia , Muscular Diseases , Adult , Animals , Arrhythmias, Cardiac/diagnosis , Arrhythmias, Cardiac/etiology , Arrhythmias, Cardiac/physiopathology , Arrhythmias, Cardiac/prevention & control , Cardiomyopathies/complications , Cardiomyopathies/diagnosis , Cardiomyopathies/drug therapy , Cardiomyopathies/genetics , Cardiomyopathies/physiopathology , Carnitine/administration & dosage , Carnitine/blood , Carnitine/genetics , Child, Preschool , Echocardiography, Doppler/methods , Electrocardiography/methods , Electrophysiologic Techniques, Cardiac , Female , Humans , Hyperammonemia/complications , Hyperammonemia/diagnosis , Hyperammonemia/drug therapy , Hyperammonemia/genetics , Hyperammonemia/physiopathology , Infant , Male , Mice , Mice, Inbred C57BL , Mitochondria, Heart/metabolism , Mitochondria, Heart/pathology , Muscular Diseases/complications , Muscular Diseases/diagnosis , Muscular Diseases/drug therapy , Muscular Diseases/genetics , Muscular Diseases/physiopathology , Treatment Outcome
17.
Sci Rep ; 5: 17969, 2015 Dec 10.
Article in English | MEDLINE | ID: mdl-26655634

ABSTRACT

Ranolazine is a recently developed drug used for the treatment of patients with chronic stable angina. It is a selective inhibitor of the persistent cardiac Na(+) current (INa), and is known to reduce the Na(+)-dependent Ca(2+) overload that occurs in cardiomyocytes during ischemia. Vascular effects of ranolazine, such as vasorelaxation,have been reported and may involve multiple pathways. As voltage-gated Na(+) channels (Nav) present in arteries play a role in contraction, we hypothesized that ranolazine could target these channels. We studied the effects of ranolazine in vitro on cultured aortic smooth muscle cells (SMC) and ex vivo on rat aortas in conditions known to specifically activate or promote INa. We observed that in the presence of the Nav channel agonist veratridine, ranolazine inhibited INa and intracellular Ca(2+) calcium increase in SMC, and arterial vasoconstriction. In arterial SMC, ranolazine inhibited the activity of tetrodotoxin-sensitive voltage-gated Nav channels and thus antagonized contraction promoted by low KCl depolarization. Furthermore, the vasorelaxant effects of ranolazine, also observed in human arteries and independent of the endothelium, involved antagonization of the α1-adrenergic receptor. Combined α1-adrenergic antagonization and inhibition of SMCs Nav channels could be involved in the vascular effects of ranolazine.


Subject(s)
Adrenergic alpha-1 Receptor Antagonists/pharmacology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Ranolazine/pharmacology , Receptors, Adrenergic, alpha-1/metabolism , Sodium Channel Blockers/pharmacology , Voltage-Gated Sodium Channels/metabolism , Action Potentials/drug effects , Animals , Aorta , Humans , Muscle Contraction/drug effects , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Rats , Uterine Artery/drug effects , Uterine Artery/metabolism , Vasodilation/drug effects
18.
Free Radic Biol Med ; 86: 269-78, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25911196

ABSTRACT

Neuroprostanes are lipid mediators produced by nonenzymatic free radical peroxidation of docosahexaenoic acid (DHA). DHA is associated with a lower atherosclerosis risk, suggesting a beneficial role in cardiovascular diseases. The aim of this study was to investigate the influence of DHA peroxidation on its potentially antiarrhythmic properties (AAP) in isolated ventricular cardiomyocytes and in vivo in post-myocardial infarcted mice. Calcium imaging and biochemical experiments indicate that cardiac arrhythmias induced by isoproterenol are associated with Ca(2+) leak from the sarcoplasmic reticulum after oxidation and phosphorylation of the type 2 ryanodine receptor (RyR2) leading to dissociation of the FKBP12.6/RyR2 complex. Both oxidized DHA and 4(RS)-4-F4t-NeuroP prevented cellular arrhythmias and posttranslational modifications of the RyR2 leading to a stabilized FKBP12.6/RyR2 complex. DHA per se did not have AAP. The AAP of 4(RS)-4-F4t-NeuroP was also observed in vivo. In this study, we challenged the paradigm that spontaneously formed oxygenated metabolites of lipids are undesirable as they are unconditionally toxic. This study reveals that the lipid mediator 4(RS)-4-F4t-neuroprostane derived from nonenzymatic peroxidation of docosahexaenoic acid can counteract such deleterious effects through cardiac antiarrhythmic properties. Our findings demonstrate 4(RS)-4-F4t-NeuroP as a mediator of the cardioprotective AAP of DHA. This discovery opens new perspectives for products of nonenzymatic oxidized ω3 polyunsaturated fatty acids as potent mediators in diseases that involve ryanodine complex destabilization such as ischemic events.


Subject(s)
Docosahexaenoic Acids/pharmacology , Neuroprostanes/physiology , Animals , Arrhythmias, Cardiac/metabolism , Calcium Signaling , Cells, Cultured , Docosahexaenoic Acids/physiology , Heart Ventricles/pathology , Lipid Peroxidation , Male , Mice, Inbred C57BL , Myocardial Infarction/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Oxidative Stress , Protective Factors
20.
Antioxid Redox Signal ; 18(9): 1009-20, 2013 Mar 20.
Article in English | MEDLINE | ID: mdl-22978600

ABSTRACT

AIMS: Heart failure (HF) is characterized by regionalized contractile alterations resulting in loss of the transmural contractile gradient across the left ventricular free wall. We tested whether a regional alteration in mitochondrial oxidative metabolism during HF could affect myofilament function through protein kinase A (PKA) signaling. RESULTS: Twelve weeks after permanent left coronary artery ligation that induced myocardial infarction (MI), subendocardial (Endo) cardiomyocytes had decreased activity of complex I and IV of the mitochondrial electron transport chain and produced twice more superoxide anions than sham Endo and subepicardial cells. This effect was associated with a reduced antioxidant activity of superoxide dismutase and Catalase only in MI Endo cells. The myofilament contractile properties (Ca(2+) sensitivity and maximal tension), evaluated in skinned cardiomyocytes, were also reduced only in MI Endo myocytes. Conversely, in MI rats treated with the antioxidant N-acetylcysteine (NAC) for 4 weeks, the generation of superoxide anions in Endo cardiomyocytes was normalized and the contractile properties of skinned cardiomyocytes restored. This effect was accompanied by improved in vivo contractility. The beneficial effects of NAC were mediated, at least, in part, through reduction of the PKA activity, which was higher in MI myofilaments, particularly, the PKA-mediated hyperphosphorylation of cardiac Troponin I. INNOVATION: The Transmural gradient in the mitochondrial content/activity is lost during HF and mediates reactive oxygen species-dependent contractile dysfunction. CONCLUSIONS: Regionalized alterations in redox signaling affect the contractile machinery of sub-Endo myocytes through a PKA-dependent pathway that contributes to the loss of the transmural contractile gradient and impairs global contractility.


Subject(s)
Heart Failure/physiopathology , Myocardial Contraction , Myocardial Ischemia/physiopathology , Reactive Oxygen Species/metabolism , Acetylcysteine/therapeutic use , Animals , Antioxidants/therapeutic use , Calcium/pharmacology , Catalase/metabolism , Cyclic AMP/physiology , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/pharmacology , Cyclic AMP-Dependent Protein Kinases/metabolism , Heart Failure/etiology , Heart Failure/metabolism , Lipid Peroxidation , Male , Mitochondria, Heart/metabolism , Myocardial Contraction/drug effects , Myocardial Ischemia/complications , Myocardial Ischemia/metabolism , Myocytes, Cardiac/metabolism , Myofibrils/drug effects , Myofibrils/physiology , Oxidative Stress/drug effects , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Random Allocation , Rats , Rats, Wistar , Recombinant Proteins/pharmacology , Superoxide Dismutase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...