Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 18 de 18
Filtrer
1.
Neuromuscul Disord ; 33(7): 546-550, 2023 07.
Article de Anglais | MEDLINE | ID: mdl-37315422

RÉSUMÉ

Pathogenic variants in the skeletal muscle α-actin 1 gene (ACTA1) cause a spectrum of myopathies with clinical and myopathological diversity. Clinical presentations occur from the prenatal period to adulthood, commonly with proximal-predominant weakness and rarely preferential distal weakness. Myopathological findings are wide-ranging, with nemaline rods being most frequent. Associated cardiomyopathy is rare and conduction defects are not reported. We describe a family with congenital myopathy with prominent finger flexor weakness and cardiomyopathy with cardiac conduction defects. The proband, a 48-year-old Caucasian male, his 73-year-old mother, 41-year-old sister, and 19-year-old nephew presented with prominent finger flexor weakness on a background of neonatal hypotonia and delayed motor milestones. All had progressive cardiomyopathy with systolic dysfunction and/or left ventricular dilation. The proband and sister had intraventricular conduction delay and left anterior fascicular block, respectively. The mother had atrial fibrillation. Muscle biopsy in the proband and sister demonstrated congenital fiber-type disproportion and rare nemaline rods in the proband. A novel dominant variant in ACTA1 (c.81C>A, p.Asp27Glu) segregated within the family. This family expands the genotypic and phenotypic spectrum of ACTA1-related myopathy, highlighting preferential finger flexor involvement with cardiomyopathy and conduction disease. We emphasize early and ongoing cardiac surveillance in ACTA1-related myopathy.


Sujet(s)
Cardiomyopathies , Myopathies némaline , Myopathies congénitales structurales , Adulte , Sujet âgé , Femelle , Humains , Nouveau-né , Mâle , Adulte d'âge moyen , Jeune adulte , Actines/génétique , Trouble de la conduction cardiaque/complications , Trouble de la conduction cardiaque/anatomopathologie , Cardiomyopathies/anatomopathologie , Mères , Faiblesse musculaire/anatomopathologie , Muscles squelettiques/anatomopathologie , Mutation , Myopathies némaline/anatomopathologie , Myopathies congénitales structurales/anatomopathologie
2.
Genes (Basel) ; 12(8)2021 08 21.
Article de Anglais | MEDLINE | ID: mdl-34440456

RÉSUMÉ

Cardiac conduction disease (CCD), which causes altered electrical impulse propagation in the heart, is a life-threatening condition with high morbidity and mortality. It exhibits genetic and clinical heterogeneity with diverse pathomechanisms, but in most cases, it disrupts the synchronous activity of impulse-generating nodes and impulse-conduction underlying the normal heartbeat. In this study, we investigated a consanguineous Pakistani family comprised of four patients with CCD. We applied whole exome sequencing (WES) and co-segregation analysis, which identified a novel homozygous missense mutation (c.1531T>C;(p.Ser511Pro)) in the highly conserved kinase domain of the cardiac troponin I-interacting kinase (TNNI3K) encoding gene. The behaviors of mutant and native TNNI3K were compared by performing all-atom long-term molecular dynamics simulations, which revealed changes at the protein surface and in the hydrogen bond network. Furthermore, intra and intermolecular interaction analyses revealed that p.Ser511Pro causes structural variation in the ATP-binding pocket and the homodimer interface. These findings suggest p.Ser511Pro to be a pathogenic variant. Our study provides insights into how the variant perturbs the TNNI3K structure-function relationship, leading to a disease state. This is the first report of a recessive mutation in TNNI3K and the first mutation in this gene identified in the Pakistani population.


Sujet(s)
Trouble de la conduction cardiaque/génétique , Prédisposition génétique à une maladie , Protein-Serine-Threonine Kinases/génétique , Troponine I/génétique , Adolescent , Adulte , Trouble de la conduction cardiaque/épidémiologie , Trouble de la conduction cardiaque/anatomopathologie , Enfant , Consanguinité , Femelle , Homozygote , Humains , Mâle , Adulte d'âge moyen , Mutation faux-sens/génétique , Pakistan/épidémiologie , Pedigree , Motifs et domaines d'intéraction protéique/génétique , Protein-Serine-Threonine Kinases/ultrastructure , Facteurs de transcription/génétique , Troponine I/ultrastructure , , Jeune adulte
3.
Bioorg Med Chem Lett ; 46: 128162, 2021 08 15.
Article de Anglais | MEDLINE | ID: mdl-34062251

RÉSUMÉ

In the United States, approximately one million individuals are hospitalized every year for arrhythmias, making arrhythmias one of the top causes of healthcare expenditures. Mexiletine is currently used as an antiarrhythmic drug but has limitations. The purpose of this work was to use normal and Long QT syndrome Type 3 (LQTS3) patient-derived human induced pluripotent stem cell (iPSC)-derived cardiomyocytes to identify an analog of mexiletine with superior drug-like properties. Compared to racemic mexiletine, medicinal chemistry optimization of substituted racemic pyridyl phenyl mexiletine analogs resulted in a more potent sodium channel inhibitor with greater selectivity for the sodium over the potassium channel and for late over peak sodium current.


Sujet(s)
Trouble de la conduction cardiaque/anatomopathologie , Cellules souches pluripotentes induites/composition chimique , Syndrome du QT long/anatomopathologie , Méxilétine/pharmacologie , Myocytes cardiaques/anatomopathologie , Canal sodique voltage-dépendant NAV1.5/métabolisme , Pyridines/pharmacologie , Relation dose-effet des médicaments , Humains , Méxilétine/composition chimique , Structure moléculaire , Pyridines/composition chimique , Relation structure-activité
4.
J Med Chem ; 64(9): 5384-5403, 2021 05 13.
Article de Anglais | MEDLINE | ID: mdl-33942619

RÉSUMÉ

Ventricular cardiac arrhythmia (VA) arises in acquired or congenital heart disease. Long QT syndrome type-3 (LQT3) is a congenital form of VA caused by cardiac sodium channel (INaL) SCN5A mutations that prolongs cardiac action potential (AP) and enhances INaL current. Mexiletine inhibits INaL and shortens the QT interval in LQT3 patients. Above therapeutic doses, mexiletine prolongs the cardiac AP. We explored structure-activity relationships (SAR) for AP shortening and prolongation using dynamic medicinal chemistry and AP kinetics in human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). Using patient-derived LQT3 and healthy hiPSC-CMs, we resolved distinct SAR for AP shortening and prolongation effects in mexiletine analogues and synthesized new analogues with enhanced potency and selectivity for INaL. This resulted in compounds with decreased AP prolongation effects, increased metabolic stability, increased INaL selectivity, and decreased avidity for the potassium channel. This study highlights using hiPSC-CMs to guide medicinal chemistry and "drug development in a dish".


Sujet(s)
Antiarythmiques/composition chimique , Trouble de la conduction cardiaque/anatomopathologie , Syndrome du QT long/anatomopathologie , Méxilétine/analogues et dérivés , Potentiels d'action/effets des médicaments et des substances chimiques , Animaux , Antiarythmiques/pharmacologie , Comportement animal/effets des médicaments et des substances chimiques , Trouble de la conduction cardiaque/métabolisme , Cellules cultivées , Conception de médicament , Stabilité de médicament , Période , Humains , Cellules souches pluripotentes induites/cytologie , Cellules souches pluripotentes induites/métabolisme , Syndrome du QT long/métabolisme , Mâle , Méxilétine/pharmacologie , Souris , Souris de lignée BALB C , Myocytes cardiaques/cytologie , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myocytes cardiaques/physiologie , Canal sodique voltage-dépendant NAV1.5/génétique , Canal sodique voltage-dépendant NAV1.5/métabolisme , Rats , Rat Sprague-Dawley , Relation structure-activité
5.
Cell Physiol Biochem ; 55(S3): 87-107, 2021 Mar 06.
Article de Anglais | MEDLINE | ID: mdl-33667333

RÉSUMÉ

Potassium channels of the tandem of two-pore-domain (K2P) family were among the last potassium channels cloned. However, recent progress in understanding their physiological relevance and molecular pharmacology revealed their therapeutic potential and thus these channels evolved as major drug targets against a large variety of diseases. However, after the initial cloning of the fifteen family members there was a lack of potent and/or selective modulators. By now a large variety of K2P channel modulators (activators and blockers) have been described, especially for TASK-1, TASK-3, TREK-1, TREK2, TRAAK and TRESK channels. Recently obtained crystal structures of K2P channels, alanine scanning approaches to map drug binding sites, in silico experiments with molecular dynamics simulations (MDs) combined with electrophysiological studies to reveal the mechanism of channel inhibition/activation, yielded a good understanding of the molecular pharmacology of these channels. Besides summarizing drugs that were identified to modulate K2P channels, the main focus of this article is on describing the differential binding sites and mechanisms of channel modulation that are utilized by the different K2P channel blockers and activators.


Sujet(s)
Trouble de la conduction cardiaque/traitement médicamenteux , Modulateurs du transport transmembranaire/pharmacologie , Migraines/traitement médicamenteux , Canaux potassiques à pores à domaines en tandem/métabolisme , Potassium/métabolisme , Potentiels d'action/effets des médicaments et des substances chimiques , Potentiels d'action/physiologie , Sites de fixation , Trouble de la conduction cardiaque/génétique , Trouble de la conduction cardiaque/métabolisme , Trouble de la conduction cardiaque/anatomopathologie , Expression des gènes , Humains , Ouverture et fermeture des portes des canaux ioniques/effets des médicaments et des substances chimiques , Transport des ions , Ligands , Modulateurs du transport transmembranaire/composition chimique , Modulateurs du transport transmembranaire/classification , Migraines/génétique , Migraines/métabolisme , Migraines/anatomopathologie , Simulation de dynamique moléculaire , Spécificité d'organe , Canaux potassiques à pores à domaines en tandem/classification , Canaux potassiques à pores à domaines en tandem/génétique , Liaison aux protéines , Isoformes de protéines/classification , Isoformes de protéines/génétique , Isoformes de protéines/métabolisme , Structure secondaire des protéines
6.
Med Mol Morphol ; 54(3): 259-264, 2021 Sep.
Article de Anglais | MEDLINE | ID: mdl-33651170

RÉSUMÉ

A 16-year-old Japanese man was admitted to our hospital because of syncope during exercise. His father and his younger brother had permanent pacemaker implantation because of sick sinus syndrome. Several examinations revealed first-degree atrioventricular block, complete right bundle branch block, sick sinus syndrome, and ventricular tachycardia with normal cardiac function. As no abnormalities were evident on coronary angiography, right ventricular endomyocardial biopsy was performed. It showed myocardial disarrangement and lipofuscin accumulation in hypertrophic myocytes. Moreover, electron microscopy showed a few degenerative myocytes, Z-band streaming, disarrangement, increased small capillaries with Weibel-Palade bodies in endothelial cells, and endothelial proliferations. Genetic analysis of the proband, his father, and his younger brother revealed a missense mutation, D1275N, in SCN5A, a gene which encodes sodium ion channel protein, are related to cardiomyopathy and arrhythmia. The proband was diagnosed with a cardiac conduction defect (CCD) and underwent permanent pacemaker implantation. These pathological findings suggest various myocardial changes presented in CCD patients with a missense mutation, D1275N, in SCN5A.


Sujet(s)
Trouble de la conduction cardiaque/génétique , Mutation faux-sens , Myocarde/anatomopathologie , Canal sodique voltage-dépendant NAV1.5/génétique , Adolescent , Troubles du rythme cardiaque/génétique , Troubles du rythme cardiaque/métabolisme , Troubles du rythme cardiaque/anatomopathologie , Trouble de la conduction cardiaque/métabolisme , Trouble de la conduction cardiaque/anatomopathologie , Trouble de la conduction cardiaque/thérapie , Humains , Mâle , Pedigree
7.
Int J Mol Sci ; 22(4)2021 Feb 04.
Article de Anglais | MEDLINE | ID: mdl-33557237

RÉSUMÉ

Brugada syndrome and early repolarization syndrome are both classified as J-wave syndromes, with a similar mechanism of arrhythmogenesis and with the same basis for genesis of the characteristic electrocardiographic features. The Brugada syndrome is now considered a conduction disorder based on subtle structural abnormalities in the right ventricular outflow tract. Recent evidence suggests structural substrate in patients with the early repolarization syndrome as well. We propose a unifying mechanism based on these structural abnormalities explaining both arrhythmogenesis and the electrocardiographic changes. In addition, we speculate that, with increasing technical advances in imaging techniques and their spatial resolution, these syndromes will be reclassified as structural heart diseases or cardiomyopathies.


Sujet(s)
Troubles du rythme cardiaque/anatomopathologie , Syndrome de Brugada/anatomopathologie , Trouble de la conduction cardiaque/anatomopathologie , Fibrose/physiopathologie , Système de conduction du coeur/malformations , Animaux , Troubles du rythme cardiaque/étiologie , Syndrome de Brugada/étiologie , Trouble de la conduction cardiaque/étiologie , Humains
8.
J Pediatr Endocrinol Metab ; 33(6): 713-720, 2020 May 22.
Article de Anglais | MEDLINE | ID: mdl-32441671

RÉSUMÉ

Objectives Investigation of the association between epicardial adipose tissue thickness (EATT) and P-wave dispersion (Pd), QT dispersion (QTd), corrected QT dispersion (QTcd) and Tp-e interval in children with Type 1 Diabetes Mellitus (T1DM) was aimed. Methods Forty-one children with T1DM and 41 age- and gender-matched healthy children were included in the study. Demographical characteristics of all cases were examined. In echocardiography; in addition to conventional echocardiographic measurements, end-systolic EATT was measured from right ventricular free wall. In electrocardiogram; Pd, QTd, QTcd and Tp-e interval durations, as well as Tp-e/QT and Tp-e/QTc ratios were calculated. Correlation values between EATT and electrocardiographic parameters were also noted. Results Mean age of the patient group was determined to be 12.43 ± 3.04 years and that of the control group was determined to be 12.08 ± 2.56 years. There was no significant difference between the groups in regard to age, gender, body weight, height and body mass index. In the patient group; EATT, Pd, QTd, QTcd and Tp-e interval were determined to be significantly higher compared to the control group. In the patient group, no significant correlation was determined between EATT and Pd, QTd, QTcd and Tp-e. However, when both patient and control groups were evaluated together, a statistically significant positive correlation was determined between EATT and Pd, QTd, QTcd and Tp-e. Conclusions In children with T1DM, an increase in epicardial adipose tissue thickness and in risk of cardiac arrhythmias has been demonstrated. To reveal the possible unfavorable effects of EATT on cardiac conduction system in T1DM patients needs further studies.


Sujet(s)
Tissu adipeux/anatomopathologie , Adiposité/physiologie , Diabète de type 1/diagnostic , Système de conduction du coeur/métabolisme , Péricarde/anatomopathologie , Tissu adipeux/imagerie diagnostique , Tissu adipeux/métabolisme , Adolescent , Troubles du rythme cardiaque/diagnostic , Troubles du rythme cardiaque/étiologie , Trouble de la conduction cardiaque/diagnostic , Trouble de la conduction cardiaque/étiologie , Trouble de la conduction cardiaque/anatomopathologie , Études cas-témoins , Enfant , Diabète de type 1/complications , Diabète de type 1/anatomopathologie , Diabète de type 1/physiopathologie , Angiopathies diabétiques/diagnostic , Angiopathies diabétiques/étiologie , Échocardiographie , Femelle , Système de conduction du coeur/imagerie diagnostique , Système de conduction du coeur/anatomopathologie , Humains , Mâle , Taille d'organe/physiologie , Péricarde/imagerie diagnostique , Péricarde/métabolisme , Facteurs de risque
9.
Pediatr Dev Pathol ; 23(4): 312-316, 2020 Aug.
Article de Anglais | MEDLINE | ID: mdl-31821773

RÉSUMÉ

Cartilaginous metaplasia involving the atrioventricular (AV) node is an uncommon entity that may cause sudden cardiac death secondary to dysrhythmias. We report 2 autopsy cases of full-term male newborns: 1 stillborn and 1 live-born, with antemortem bradycardia who died in the peripartum period. An examination of the cardiac conduction system in both cases demonstrated extensive cartilaginous metaplasia of the central fibrous body and involvement of the AV node and bundle of His. The cases highlight the recognition of cardiac conduction system anomalies as a cause of sudden perinatal death. In cases of perinatal death with preceding arrhythmia, postmortem sections of the cardiac conduction system are recommended to examine for cardiac conduction system anomaly.


Sujet(s)
Noeud atrioventriculaire/anatomopathologie , Trouble de la conduction cardiaque/congénital , Trouble de la conduction cardiaque/anatomopathologie , Mort subite cardiaque/étiologie , Autopsie , Trouble de la conduction cardiaque/diagnostic , Humains , Nouveau-né , Mâle , Métaplasie , Mort périnatale , Mortinatalité
11.
Circulation ; 140(8): e483-e503, 2019 08 20.
Article de Anglais | MEDLINE | ID: mdl-30586773

RÉSUMÉ

BACKGROUND: It is unclear whether physiologic pacing by either cardiac biventricular pacing (BiVP) or His bundle pacing (HisBP) may prevent adverse structural and functional consequences known to occur among some patients who receive right ventricular pacing (RVP). AIM: Our analysis sought to review existing literature to determine if BiVP and/or HisBP might prevent adverse remodeling and be associated with structural, functional, and clinical advantages compared with RVP among patients without severe left ventricular dysfunction (>35%) who required permanent pacing because of heart block. METHODS: A literature search was conducted using MEDLINE (through PubMed) and Embase to identify randomized trials and observational studies comparing the effects of BiVP or HisBP versus RVP on measurements of left ventricular dimensions, left ventricular ejection fraction (LVEF), heart failure functional classification, quality of life, 6-minute walk, hospitalizations, and mortality. Data from studies that met the appropriate population, intervention, comparator, and outcomes of interest were abstracted for meta-analysis. Studies that reported pooled outcomes among patients with LVEF both above and below 35% could not be included in the meta-analysis because of strict relationships with industry procedures that preclude retrieval of industry-retained unpublished data on the subset of patients with preserved left ventricular function. RESULTS: Evidence from 8 studies, including a total of 679 patients meeting the prespecified criteria for inclusion, was identified. Results were compared for BiVP versus RVP, HisBP versus RVP, and BiVP+HisBP versus RVP. Among patients who received physiologic pacing with either BiVP or HisBP, the LV end-diastolic and end-systolic volumes were significantly lower (mean duration of follow-up: 1.64 years; -2.77 mL [95% CI -4.37 to -1.1 mL]; P=0.001; and -7.09 mL [95% CI -11.27 to -2.91; P=0.0009) and LVEF remained preserved or increased (mean duration of follow-up: 1.57 years; 5.328% [95% CI: 2.86%-7.8%; P<0.0001). Data on clinical impact such as functional status and quality of life were not definitive. Data on hospitalizations were unavailable. There was no effect on mortality. Several studies stratified results by LVEF and found that patients with LVEF >35% but ≤52% were more likely to receive benefit from physiologic pacing. Patients with chronic atrial fibrillation who underwent atrioventricular node ablation and pacemaker implant demonstrated clear improvement in LVEF with BiVP or HisBP versus RVP. CONCLUSION: Among patients with LVEF >35%, the LVEF remained preserved or increased with either BiVP or HisBP compared with RVP. However, patient-centered clinical outcome improvement appears to be limited primarily to patients who have chronic atrial fibrillation with rapid ventricular response rates and have undergone atrioventricular node ablation.


Sujet(s)
Bradycardie/anatomopathologie , Trouble de la conduction cardiaque/anatomopathologie , Entraînement électrosystolique/méthodes , Fonction ventriculaire gauche/physiologie , Fibrillation auriculaire/complications , Bradycardie/complications , Bradycardie/thérapie , Trouble de la conduction cardiaque/complications , Trouble de la conduction cardiaque/thérapie , Humains , Guides de bonnes pratiques cliniques comme sujet , Qualité de vie , Débit systolique
12.
Med Sci (Paris) ; 34(10): 820-823, 2018 Oct.
Article de Français | MEDLINE | ID: mdl-30451676

RÉSUMÉ

Macrophages regulate cardiac homeostasis under pathological and physiological conditions. Recent studies have elegantly substantiated the presence of specific subset of macrophages residing within the distal atrioventricular node in mice and humans. These macrophages directly couple with cardiomyocytes via connexin-43-containing gap junctions and increase atrioventricular conduction by accelerating cardiomyocyte repolarization. Conditional deletion of connexin-43 in macrophages or congenital lack of macrophages delay nodal conduction and foster progressive atrioventricular block. Exhaustive understanding of the role of tissue-resident macrophages in normal and aberrant cardiac conduction could initiate the development of therapeutic strategies focused on the modulation of macrophage functions in heart arrhythmia.


Sujet(s)
Troubles du rythme cardiaque/étiologie , Rythme cardiaque/physiologie , Macrophages/anatomopathologie , Macrophages/physiologie , Animaux , Troubles du rythme cardiaque/anatomopathologie , Troubles du rythme cardiaque/thérapie , Trouble de la conduction cardiaque/étiologie , Trouble de la conduction cardiaque/anatomopathologie , Humains , Souris , Myocytes cardiaques/physiologie , Myocytes cardiaques/ultrastructure
15.
Cell Physiol Biochem ; 44(4): 1559-1577, 2017.
Article de Anglais | MEDLINE | ID: mdl-29197877

RÉSUMÉ

BACKGROUND/AIMS: Truncating LMNA gene mutations occur in many inherited cardiomyopathy cases, but the molecular mechanisms involved in the disease they cause have not yet been systematically investigated. Here, we studied a novel frameshift LMNA variant (p.D243Gfs*4) identified in three members of an Italian family co-segregating with a severe form of cardiomyopathy with conduction defects. METHODS: HEK293 cells and HL-1 cardiomyocytes were transiently transfected with either Lamin A or D243Gfs*4 tagged with GFP (or mCherry). D243Gfs*4 expression, cellular localization and its effects on diverse cellular mechanisms were evaluated with western blotting, laser-scanning confocal microscopy and video-imaging analysis in single cells. RESULTS: When expressed in HEK293 cells, GFP- (or mCherry)-tagged LMNA D243Gfs*4 colocalized with calnexin within the ER. ER mislocalization of LMNA D243Gfs*4 did not significantly induce ER stress response, abnormal Ca2+ handling and apoptosis when compared with HEK293 cells expressing another truncated mutant of LMNA (R321X) which similarly accumulates within the ER. Of note, HEK293-LMNA D243Gfs*4 cells showed a significant reduction of connexin 43 (CX43) expression level, which was completely rescued by activation of the WNT/ß-catenin signaling pathway. When expressed in HL-1 cardiomyocytes, D243Gfs*4 significantly impaired the spontaneous Ca2+ oscillations recorded in these cells as result of propagation of the depolarizing waves through the gap junctions between non-transfected cells surrounding a cell harboring the mutation. Furthermore, mCh-D243Gfs*4 HL-1 cardiomyocytes showed reduced CX43-dependent Lucifer Yellow (LY) loading and propagation. Of note, activation of ß-catenin rescued both LY loading and LMNA D243Gfs*4 -HL-1 cells spontaneous activity propagation. CONCLUSION: Overall, the present results clearly indicate the involvement of the aberrant CX43 expression/activity as a pathogenic mechanism for the conduction defects associated to this LMNA truncating alteration.


Sujet(s)
Trouble de la conduction cardiaque/génétique , Cardiomyopathies/génétique , Lamine A/génétique , Apoptose , Séquence nucléotidique , Calcium/métabolisme , Calnexine/métabolisme , Trouble de la conduction cardiaque/complications , Trouble de la conduction cardiaque/anatomopathologie , Cardiomyopathies/complications , Cardiomyopathies/anatomopathologie , Lignée cellulaire , Connexine 43 , Réticulum endoplasmique/métabolisme , Femelle , Jonctions communicantes/métabolisme , Cellules HEK293 , Humains , Lamine A/métabolisme , Répétitions microsatellites/génétique , Microscopie confocale , Adulte d'âge moyen , Mutagenèse dirigée , Myocytes cardiaques/cytologie , Myocytes cardiaques/métabolisme , Pedigree , Polymorphisme de nucléotide simple , Imagerie accélérée , Voie de signalisation Wnt
16.
Curr Opin HIV AIDS ; 12(6): 528-533, 2017 Nov.
Article de Anglais | MEDLINE | ID: mdl-28984702

RÉSUMÉ

PURPOSE OF REVIEW: The increasing prevalence of cardiovascular disease comorbidity in persons infected with the HIV has become a global concern. The electrocardiogram (ECG) is increasingly being utilized to provide clinically relevant information regarding cardiac arrhythmias and cardio-autonomic dysfunction. The purpose of this review is to summarize the latest research comparing QT and R-to-R interval length as a function of HIV+ status or antiretroviral therapy (ART) regimen. RECENT FINDINGS: Prolongation of the corrected QTc interval may be acquired in HIV+ ART-naive individuals, exacerbated by various classes of ART drugs, and is generally predictive of lethal cardiac arrhythmias, with effects observed from childhood to adulthood. Recent literature also suggests the trend of lower heart rate variability in HIV is indicative of cardiorespiratory and inflammatory-immune dysfunction. SUMMARY: These emergent studies support the clinical relevance of the ECG across the age and HIV disease spectrum. Furthermore, the reported findings have implications for the management of cardiovascular and chronic inflammatory disease comorbidity in persons living with HIV.


Sujet(s)
Antirétroviraux/usage thérapeutique , Variation intra-population/effets des médicaments et des substances chimiques , Trouble de la conduction cardiaque/diagnostic , Trouble de la conduction cardiaque/anatomopathologie , Infections à VIH/anatomopathologie , Système de conduction du coeur/effets des médicaments et des substances chimiques , Rythme cardiaque/effets des médicaments et des substances chimiques , Humains
17.
Heart Rhythm ; 14(11): 1704-1712, 2017 11.
Article de Anglais | MEDLINE | ID: mdl-28756098

RÉSUMÉ

BACKGROUND: Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) are in vitro models with the clear advantages of their human origin and suitability for human disease investigations. However, limitations include their incomplete characterization and variability reported in different cell lines and laboratories. OBJECTIVE: The purpose of this study was to investigate in silico ionic mechanisms potentially explaining the phenotypic variability of hiPSC-CMs in long QT syndrome type 3 (LQT3) and their response to antiarrhythmic drugs. METHODS: Populations of in silico hiPSC-CM models were constructed and calibrated for control (n = 1,463 models) and LQT3 caused by INaL channelopathy (n = 1,401 models), using experimental recordings for late sodium current (INaL) and action potentials (APs). Antiarrhythmic drug therapy was evaluated by simulating mexiletine and ranolazine multichannel effects. RESULTS: As in experiments, LQT3 hiPSC-CMs yield prolonged action potential duration at 90% repolarization (APD90) (+34.3% than controls) and large electrophysiological variability. LQT3 hiPSC-CMs with symptomatic APs showed overexpression of ICaL, IK1, and INaL, underexpression of IKr, and increased sensitivity to both drugs compared to asymptomatic LQT3 models. Simulations showed that both mexiletine and ranolazine corrected APD prolongation in the LQT3 population but also highlighted differences in drug response. Mexiletine stops spontaneous APs in more LQT3 hiPSC-CMs models than ranolazine (784/1,401 vs 53/1,401) due to its stronger action on INa. CONCLUSION: In silico simulations demonstrate our ability to recapitulate variability in LQT3 and control hiPSC-CM phenotypes, and the ability of mexiletine and ranolazine to reduce APD prolongation, in agreement with experiments. The in silico models also identify potential ionic mechanisms of phenotypic variability in LQT3 hiPSC-CMs, explaining APD prolongation in symptomatic vs asymptomatic LQT3 hiPSC-CMs.


Sujet(s)
Trouble de la conduction cardiaque/traitement médicamenteux , Cellules souches pluripotentes induites/anatomopathologie , Syndrome du QT long/traitement médicamenteux , Méxilétine/pharmacologie , Myocytes cardiaques/anatomopathologie , Potentiels d'action , Antiarythmiques/pharmacologie , Variation intra-population , Trouble de la conduction cardiaque/anatomopathologie , Trouble de la conduction cardiaque/physiopathologie , Lignée cellulaire , Simulation numérique , Humains , Cellules souches pluripotentes induites/effets des médicaments et des substances chimiques , Syndrome du QT long/anatomopathologie , Syndrome du QT long/physiopathologie , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myocytes cardiaques/métabolisme , Techniques de patch-clamp
18.
Acta Biochim Biophys Sin (Shanghai) ; 49(3): 270-276, 2017 Mar 01.
Article de Anglais | MEDLINE | ID: mdl-28159958

RÉSUMÉ

Cardiac sodium channel plays a key role in the fast depolarization and maintenance of impulse conduction in cardiomyocytes. Mutations of SCN5A gene can lead to many types of arrhythmias. A 14-year-old boy with familial paternal history of sudden unexpected nocturnal death was admitted to hospital with recurrent syncope. A cardiac channelopathy was suspected and a pathogenic ion channel was searched for mutation identification. The proband manifested sinus node dysfunction, ventricular tachycardia, cardiac conduction disturbance involving atrioventricular node and His bundle. The proband and his mother received whole exome sequencing. A heterozygous in-frame deletion N1380del on exon 23 of SCN5A gene locating in a highly conserved pore residue in domain III (S5-S6) was revealed in the proband. The mutation was assessed in other family members by Sanger sequencing. The proband's living uncle and two sisters were asymptomatic mutation carriers with different degrees of cardiac conduction disturbance. Functional analysis was conducted using whole-cell patch clamping in HEK293T cells transfected with wild-type or mutant channels. The HEK293T cells transfected with plasmid pcDNA3.1-N1380del-SCN5A had no detectable sodium current. Overall, N1380del mutation of SCN5A gene leads to loss of function of sodium channel. N1380del is a pathogenetic mutation which can cause cardiac conduction defect and ventricular tachycardia.


Sujet(s)
Trouble de la conduction cardiaque/génétique , Mutation , Canal sodique voltage-dépendant NAV1.5/génétique , Tachycardie ventriculaire/génétique , Adolescent , Trouble de la conduction cardiaque/anatomopathologie , Trouble de la conduction cardiaque/thérapie , Exons , Humains , Mâle , Phénotype , Pronostic , Tachycardie ventriculaire/anatomopathologie , Tachycardie ventriculaire/thérapie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...