134 results on '"Flavien Charpentier"'
Search Results
2. Intracardiac electrophysiology to characterize susceptibility to ventricular arrhythmias in murine models
- Author
-
Marine C. Ferrand, Gauthier Giordano, Nathalie Mougenot, Pierre-Léo Laporte, Nicolas Vignier, Arnaud Leclerc, Vincent Algalarrondo, Fabrice Extramiana, Flavien Charpentier, and Nathalie Neyroud
- Subjects
programmed electrical stimulation ,ventricular arrhythmias ,electrocardiography ,electrophysiology ,channelopathies ,cardiomyopathies ,Physiology ,QP1-981 - Abstract
Introduction: Sudden cardiac death (SCD) and ventricular fibrillation are rare but severe complications of many cardiovascular diseases and represent a major health issue worldwide. Although the primary causes are often acute or chronic coronary diseases, genetic conditions, such as inherited channelopathies or non-ischemic cardiomyopathies are leading causes of SCD among the young. However, relevant experimental models to study the underlying mechanisms of arrhythmias and develop new therapies are still needed. The number of genetically engineered mouse models with cardiac phenotype is growing, making electrophysiological studies in mice essential tools to study arrhythmogenicity and arrhythmia mechanisms and to test novel treatments. Recently, intracardiac catheterization via the jugular vein was described to induce and record ventricular arrhythmias in living anesthetized mice. Several strategies have been reported, developed in healthy wild-type animals and based on aggressive right ventricular stimulation.Methods: Here, we report a protocol based on programmed electrical stimulation (PES) performed in clinical practice in patients with cardiac rhythm disorders, adapted to two transgenic mice models of arrhythmia - Brugada syndrome and cardiolaminopathy.Results: We show that this progressive protocol, based on a limited number of right ventricular extrastimuli, enables to reveal different rhythmic phenotypes between control and diseased mice. In this study, we provide detailed information on PES in mice, including catheter positioning, stimulation protocols, intracardiac and surface ECG interpretation and we reveal a higher susceptibility of two mouse lines to experience triggered ventricular arrhythmias, when compared to control mice.Discussion: Overall, this technique allows to characterize arrhythmias and provides results in phenotyping 2 arrhythmogenic-disease murine models.
- Published
- 2024
- Full Text
- View/download PDF
3. Editorial: Experimental models and model organisms in cardiac electrophysiology: opportunities and challenges
- Author
-
Bianca J. J. M. Brundel, Chris Jopling, Igor R. Efimov, Flavien Charpentier, and Yoram Etzion
- Subjects
experimental models ,cardiac electrophysiology ,arrhythmia ,model organisms ,cardiac electrophysiologic technique ,Physiology ,QP1-981 - Published
- 2023
- Full Text
- View/download PDF
4. Predicting hERG repolarization power at 37°C from recordings at room temperature
- Author
-
Barbara B. R. Oliveira‐Mendes, Malak Alameh, Jérôme Montnach, Béatrice Ollivier, Solène Gibaud, Sylvain Feliciangeli, Florian Lesage, Flavien Charpentier, Gildas Loussouarn, Michel De Waard, and Isabelle Baró
- Subjects
Medicine (General) ,R5-920 - Published
- 2023
- Full Text
- View/download PDF
5. Long QT: Time to cut cholesterol?
- Author
-
Jin Li, Flavien Charpentier, and Ange Maguy
- Subjects
Diseases of the circulatory (Cardiovascular) system ,RC666-701 - Published
- 2023
- Full Text
- View/download PDF
6. SARS-CoV-2 E and 3a Proteins Are Inducers of Pannexin Currents
- Author
-
Barbara B. R. Oliveira-Mendes, Malak Alameh, Béatrice Ollivier, Jérôme Montnach, Nicolas Bidère, Frédérique Souazé, Nicolas Escriou, Flavien Charpentier, Isabelle Baró, Michel De Waard, and Gildas Loussouarn
- Subjects
COVID-19 ,SARS-CoV-2 ,viroporins ,E protein ,3a protein ,pannexin currents ,Cytology ,QH573-671 - Abstract
Controversial reports have suggested that SARS-CoV E and 3a proteins are plasma membrane viroporins. Here, we aimed at better characterizing the cellular responses induced by these proteins. First, we show that expression of SARS-CoV-2 E or 3a protein in CHO cells gives rise to cells with newly acquired round shapes that detach from the Petri dish. This suggests that cell death is induced upon expression of E or 3a protein. We confirmed this by using flow cytometry. In adhering cells expressing E or 3a protein, the whole-cell currents were not different from those of the control, suggesting that E and 3a proteins are not plasma membrane viroporins. In contrast, recording the currents on detached cells uncovered outwardly rectifying currents much larger than those observed in the control. We illustrate for the first time that carbenoxolone and probenecid block these outwardly rectifying currents; thus, these currents are most probably conducted by pannexin channels that are activated by cell morphology changes and also potentially by cell death. The truncation of C-terminal PDZ binding motifs reduces the proportion of dying cells but does not prevent these outwardly rectifying currents. This suggests distinct pathways for the induction of these cellular events by the two proteins. We conclude that SARS-CoV-2 E and 3a proteins are not viroporins expressed at the plasma membrane.
- Published
- 2023
- Full Text
- View/download PDF
7. Human MuStem Cell Grafting into Infarcted Rat Heart Attenuates Adverse Tissue Remodeling and Preserves Cardiac Function
- Author
-
Alice Rannou, Gilles Toumaniantz, Thibaut Larcher, Isabelle Leroux, Mireille Ledevin, Agnès Hivonnait, Candice Babarit, Romain Fleurisson, Laurence Dubreil, Séverine Ménoret, Ignacio Anegon, Flavien Charpentier, Karl Rouger, and Laetitia Guével
- Subjects
cell therapy ,myocardial infarction ,human adult stem cell ,MuStem cells ,regenerative medicine ,muscle-derived stem cells ,Genetics ,QH426-470 ,Cytology ,QH573-671 - Abstract
Myocardial infarction is one of the leading causes of mortality and morbidity worldwide. Whereas transplantation of several cell types into the infarcted heart has produced promising preclinical results, clinical studies using analogous human cells have shown limited structural and functional benefits. In dogs and humans, we have described a type of muscle-derived stem cells termed MuStem cells that efficiently promoted repair of injured skeletal muscle. Enhanced survival rate, long-term engraftment, and participation in muscle fiber formation were reported, leading to persistent tissue remodeling and clinical benefits. With the consideration of these features that are restricted or absent in cells tested so far for myocardial infarction, we wanted to investigate the capacity of human MuStem cells to repair infarcted hearts. Their local administration in immunodeficient rats 1 week after induced infarction resulted in reduced fibrosis and increased angiogenesis 3 weeks post-transplantation. Importantly, foci of human fibers were detected in the infarct site. Treated rats also showed attenuated left-ventricle dilation and preservation of contractile function. Interestingly, no spontaneous arrhythmias were observed. Our findings support the potential of MuStem cells, which have already been proposed as therapeutic candidates for dystrophic patients, to treat myocardial infarction and position them as an attractive tool for muscle-regenerative medicine.
- Published
- 2020
- Full Text
- View/download PDF
8. Generation of human induced pluripotent stem cell lines from four unrelated healthy control donors carrying European genetic background
- Author
-
Aurore Girardeau, Diane Atticus, Robin Canac, Bastien Cimarosti, Amandine Caillaud, Caroline Chariau, Floriane Simonet, Bertrand Cariou, Flavien Charpentier, Jean-Baptiste Gourraud, Vincent Probst, Nadjet Belbachir, Laurence Jesel, Patricia Lemarchand, Julien Barc, Richard Redon, Nathalie Gaborit, and Guillaume Lamirault
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Four human induced pluripotent stem cell (hiPSC) lines have been generated from healthy control European donors, and validated. This resource represents a useful tool for stem cell-based research, as references for developmental studies and disease modeling linked to any type of human tissue and organ, in an ethnical-, sex- and age-matched context. They providea reliable in-vitro model for single cell- and tissue-based investigations, and are also a valuable tool for genome editing-based studies.
- Published
- 2022
- Full Text
- View/download PDF
9. A consistent arrhythmogenic trait in Brugada syndrome cellular phenotype
- Author
-
Zeina R. Al Sayed, Mariam Jouni, Jean‐Baptiste Gourraud, Nadjet Belbachir, Julien Barc, Aurore Girardeau, Virginie Forest, Aude Derevier, Anne Gaignerie, Caroline Chariau, Bastien Cimarosti, Robin Canac, Pierre Olchesqui, Eric Charpentier, Jean‐Jacques Schott, Richard Redon, Isabelle Baró, Vincent Probst, Flavien Charpentier, Gildas Loussouarn, Kazem Zibara, Guillaume Lamirault, Patricia Lemarchand, and Nathalie Gaborit
- Subjects
Medicine (General) ,R5-920 - Published
- 2021
- Full Text
- View/download PDF
10. KV4.3 Expression Modulates NaV1.5 Sodium Current
- Author
-
Vincent Portero, Ronald Wilders, Simona Casini, Flavien Charpentier, Arie O. Verkerk, and Carol Ann Remme
- Subjects
transient outward current ,sodium current ,channels ,action potential ,myocyte ,arrhythmias ,Physiology ,QP1-981 - Abstract
In cardiomyocytes, the voltage-gated transient outward potassium current (Ito) is responsible for the phase-1 repolarization of the action potential (AP). Gain-of-function mutations in KCND3, the gene encoding the Ito carrying KV4.3 channel, have been associated with Brugada syndrome (BrS). While the role of Ito in the pro-arrhythmic mechanism of BrS has been debated, recent studies have suggested that an increased Ito may directly affect cardiac conduction. However, the effects of an increased Ito on AP upstroke velocity or sodium current at the cellular level remain unknown. We here investigated the consequences of KV4.3 overexpression on NaV1.5 current and consequent sodium channel availability. We found that overexpression of KV4.3 protein in HEK293 cells stably expressing NaV1.5 (HEK293-NaV1.5 cells) significantly reduced NaV1.5 current density without affecting its kinetic properties. In addition, KV4.3 overexpression decreased AP upstroke velocity in HEK293-NaV1.5 cells, as measured with the alternating voltage/current clamp technique. These effects of KV4.3 could not be explained by alterations in total NaV1.5 protein expression. Using computer simulations employing a multicellular in silico model, we furthermore demonstrate that the experimentally observed increase in KV4.3 current and concurrent decrease in NaV1.5 current may result in a loss of conduction, underlining the potential functional relevance of our findings. This study gives the first proof of concept that KV4.3 directly impacts on NaV1.5 current. Future studies employing appropriate disease models should explore the potential electrophysiological implications in (patho)physiological conditions, including BrS associated with KCND3 gain-of-function mutations.
- Published
- 2018
- Full Text
- View/download PDF
11. Dysfunction of the Voltage‐Gated K+ Channel β2 Subunit in a Familial Case of Brugada Syndrome
- Author
-
Vincent Portero, Solena Le Scouarnec, Zeineb Es‐Salah‐Lamoureux, Sophie Burel, Jean‐Baptiste Gourraud, Stéphanie Bonnaud, Pierre Lindenbaum, Floriane Simonet, Jade Violleau, Estelle Baron, Eléonore Moreau, Carol Scott, Stéphanie Chatel, Gildas Loussouarn, Thomas O'Hara, Philippe Mabo, Christian Dina, Hervé Le Marec, Jean‐Jacques Schott, Vincent Probst, Isabelle Baró, Céline Marionneau, Flavien Charpentier, and Richard Redon
- Subjects
Brugada syndrome ,cardiac arrhythmia ,clinical electrophysiology ,genetics ,KCNAB2/Kvβ2 ,potassium ion channels ,Diseases of the circulatory (Cardiovascular) system ,RC666-701 - Abstract
BackgroundThe Brugada syndrome is an inherited cardiac arrhythmia associated with high risk of sudden death. Although 20% of patients with Brugada syndrome carry mutations in SCN5A, the molecular mechanisms underlying this condition are still largely unknown. Methods and ResultsWe combined whole‐exome sequencing and linkage analysis to identify the genetic variant likely causing Brugada syndrome in a pedigree for which SCN5A mutations had been excluded. This approach identified 6 genetic variants cosegregating with the Brugada electrocardiographic pattern within the pedigree. In silico gene prioritization pointed to 1 variant residing in KCNAB2, which encodes the voltage‐gated K+ channel β2‐subunit (Kvβ2‐R12Q). Kvβ2 is widely expressed in the human heart and has been shown to interact with the fast transient outward K+ channel subunit Kv4.3, increasing its current density. By targeted sequencing of the KCNAB2 gene in 167 unrelated patients with Brugada syndrome, we found 2 additional rare missense variants (L13F and V114I). We then investigated the physiological effects of the 3 KCNAB2 variants by using cellular electrophysiology and biochemistry. Patch‐clamp experiments performed in COS‐7 cells expressing both Kv4.3 and Kvβ2 revealed a significant increase in the current density in presence of the R12Q and L13F Kvβ2 mutants. Although biotinylation assays showed no differences in the expression of Kv4.3, the total and submembrane expression of Kvβ2‐R12Q were significantly increased in comparison with wild‐type Kvβ2. ConclusionsAltogether, our results indicate that Kvβ2 dysfunction can contribute to the Brugada electrocardiographic pattern.
- Published
- 2016
- Full Text
- View/download PDF
12. Variable Na(v)1.5 protein expression from the wild-type allele correlates with the penetrance of cardiac conduction disease in the Scn5a(+/-) mouse model.
- Author
-
Anne-Laure Leoni, Bruno Gavillet, Jean-Sébastien Rougier, Céline Marionneau, Vincent Probst, Solena Le Scouarnec, Jean-Jacques Schott, Sophie Demolombe, Patrick Bruneval, Christopher L H Huang, William H Colledge, Andrew A Grace, Hervé Le Marec, Arthur A Wilde, Peter J Mohler, Denis Escande, Hugues Abriel, and Flavien Charpentier
- Subjects
Medicine ,Science - Abstract
Loss-of-function mutations in SCN5A, the gene encoding Na(v)1.5 Na+ channel, are associated with inherited cardiac conduction defects and Brugada syndrome, which both exhibit variable phenotypic penetrance of conduction defects. We investigated the mechanisms of this heterogeneity in a mouse model with heterozygous targeted disruption of Scn5a (Scn5a(+/-) mice) and compared our results to those obtained in patients with loss-of-function mutations in SCN5A.Based on ECG, 10-week-old Scn5a(+/-) mice were divided into 2 subgroups, one displaying severe ventricular conduction defects (QRS interval>18 ms) and one a mild phenotype (QRS< or = 18 ms; QRS in wild-type littermates: 10-18 ms). Phenotypic difference persisted with aging. At 10 weeks, the Na+ channel blocker ajmaline prolonged QRS interval similarly in both groups of Scn5a(+/-) mice. In contrast, in old mice (>53 weeks), ajmaline effect was larger in the severely affected subgroup. These data matched the clinical observations on patients with SCN5A loss-of-function mutations with either severe or mild conduction defects. Ventricular tachycardia developed in 5/10 old severely affected Scn5a(+/-) mice but not in mildly affected ones. Correspondingly, symptomatic SCN5A-mutated Brugada patients had more severe conduction defects than asymptomatic patients. Old severely affected Scn5a(+/-) mice but not mildly affected ones showed extensive cardiac fibrosis. Mildly affected Scn5a(+/-) mice had similar Na(v)1.5 mRNA but higher Na(v)1.5 protein expression, and moderately larger I(Na) current than severely affected Scn5a(+/-) mice. As a consequence, action potential upstroke velocity was more decreased in severely affected Scn5a(+/-) mice than in mildly affected ones.Scn5a(+/-) mice show similar phenotypic heterogeneity as SCN5A-mutated patients. In Scn5a(+/-) mice, phenotype severity correlates with wild-type Na(v)1.5 protein expression.
- Published
- 2010
- Full Text
- View/download PDF
13. Cardiac gene therapy with PDE2A limits remodeling and arrhythmias in mouse models of heart failure induced by catecholamines
- Author
-
Rima Kamel, Aurélia Bourcier, Jean Piero Margaria, Audrey Varin, Agnès Hivonnait, Françoise Mercier-Nomé, Delphine Mika, Alessandra Ghigo, Flavien Charpentier, Vincent Algalarrondo, Emilio Hirsch, Rodolphe Fischmeister, Grégoire Vandecasteele, and Jérôme Leroy
- Abstract
BACKGROUNDConstitutive cardiac PDE2 activation was shown to protect against contractile dysfunction and arrhythmia in heart failure (HF). However, it remains unknown whether an acute elevation of PDE2 is efficient to prevent maladaptive remodeling and arrhythmia. In this study we tested whether increasing acutely PDE2A activity in preclinical models of HF using cardiac PDE2 gene transfer could be of therapeutic value.METHODS AND RESULTSC57BL/6 male mice were injected with serotype 9 adeno-associated viruses (AAV9) encoding for PDE2A, or luciferase (LUC). Cardiac function assessed by echocardiography unveiled neither structural change nor dysfunction consecutive to PDE2A overexpression while AAV9 inoculation led to a ≈10-fold rise of PDE2A protein levels. Two weeks after AAV9 injections, mice were implanted with osmotic minipumps delivering NaCl or isoproterenol (Iso) (60 mg/kg/day) or Iso and phenylephrine (Iso+Phe, 30 mg/kg/day each) for 2 weeks. In LUC mice, chronic infusion with Iso increased left ventricular (LV) weight over body weight ratio, promoted fibrosis and decreased ejection fraction, but animals overexpressing PDE2A were protected towards these deleterious effects. Similarly, concomitant treatment with Iso+Phe promoted LV contractile dysfunction, fibrosis and apoptosis in LUC mice, while PDE2A overexpression limited these adverse outcomes. Furthermore, inotropic responses to Iso of ventricular cardiomyocytes isolated from Iso+Phe-LUC mice loaded with 1 µmol/L Fura-2AM and stimulated at 1 Hz to record calcium transients and sarcomere shortening were dampened. Chronic treatment with catecholamines favoured spontaneous calcium waves upon β-AR stimulation at the cellular level and promoted susceptibility to ventricular arrhythmiasin vivoevoked by catheter-mediated ventricular pacing after Iso and atropine injection. However, these adverse effects were blunted by the cardiac gene therapy with PDE2A.CONCLUSIONGene therapy with PDE2A limits cardiac adverse left ventricle remodeling and dysfunction induced by catecholamines as well as ventricular arrhythmias, providing evidence that acutely increasing PDE2A activity could prevent progression towards HF.
- Published
- 2023
- Full Text
- View/download PDF
14. Optical control of cardiac rhythm byin vivophotoactivation of an ERG channel peptide inhibitor
- Author
-
Jérôme Montnach, Hugo Millet, Antoine Persello, Hervé Meudal, Stephan De Waard, Pietro Mesrica, Barbara Ribeiro, Jérémie Richard, Agnès Hivonnait, Agnès Tessier, Benjamin Lauzier, Flavien Charpentier, Matteo E. Mangoni, Céline Landon, Chris Jopling, and Michel De Waard
- Abstract
RATIONALECardiac rhythm, conduction and synchronization of electrical activity require the coordinated action of different types of ion channels that differ according to transmural and regional specificities. Classical pharmacology affects these ion channels in a non-regionalized way which explains why treating arrhythmias, that often occur in specific foci, has often limited efficacy in addition to negative side-effects on non-targeted organs. Photopharmacology is an emergent technology that has the potential to counteract all the negative aspects of classical pharmacology by restricting drug activity in a spatio-temporal manner.OBJECTIVEWe tested the potential of photopharmacology in specifically regulating heart activity by using a caged derivative of a natural peptide inhibitor of the ERG channel, BeKm1. The peptide was uncaged and activity monitoredin vitroon a cell line expressing the hERG channel, on human cardiomyocytes derived from iPS cells, andex vivoandin vivoon zebrafish larvae and rat hearts.METHODS AND RESULTSCaged BeKm-1 is inactive and fully active upon uncaging. Uncaging of the peptide on human iPS-derived cardiomyocytes enlarges the action potential duration and triggers arrhythmias. Uncaging also triggers bradycardia and disturbs cardiac conduction within the atria in perfused rat hearts upon illumination. The potency of photopharmacology for cardiac electrical modulation was further validated in zebrafish larvae where illumination of the caged compound induces bradycardia and atrio-ventricular desynchrony. Finally, in anesthetized rats, illumination of the caged peptide in the right atria, containing the sino-atrial node, leads to bradycardia without arrhythmia.CONCLUSIONSThis report demonstrates that photopharmacology, using the caged peptide strategy, can be used for dynamically regulating cardiac electrical activityin vivoand that spatial illumination restriction can dissociate the bradycardic effect from the arrhythmic one. The technology is applicable to all kinds of cardiac ion channels and regions of interest to create arrhythmogenic models or investigate new clinical applications.
- Published
- 2023
- Full Text
- View/download PDF
15. Modelling sudden cardiac death risks factors in patients with coronavirus disease of 2019: the hydroxychloroquine and azithromycin case
- Author
-
Flavien Charpentier, Michel De Waard, Gildas Loussouarn, Jérôme Montnach, and Isabelle Baró
- Subjects
medicine.medical_specialty ,Long QT syndrome ,Dofetilide ,Azithromycin ,030204 cardiovascular system & hematology ,QT interval ,Asymptomatic ,Sudden cardiac death ,03 medical and health sciences ,0302 clinical medicine ,Basic Science ,Physiology (medical) ,Internal medicine ,Humans ,Medicine ,AcademicSubjects/MED00200 ,030212 general & internal medicine ,Subclinical infection ,SARS-CoV-2 ,business.industry ,COVID-19 ,Hydroxychloroquine ,medicine.disease ,COVID-19 Drug Treatment ,3. Good health ,Long QT Syndrome ,Death, Sudden, Cardiac ,Predictive model ,QT duration ,Cardiology ,medicine.symptom ,Cardiology and Cardiovascular Medicine ,business ,Arrhythmia ,medicine.drug - Abstract
Aims Coronavirus disease of 2019 (COVID-19) has rapidly become a worldwide pandemic. Many clinical trials have been initiated to fight the disease. Among those, hydroxychloroquine and azithromycin had initially been suggested to improve clinical outcomes. Despite any demonstrated beneficial effects, they are still in use in some countries but have been reported to prolong the QT interval and induce life-threatening arrhythmia. Since a significant proportion of the world population may be treated with such COVID-19 therapies, evaluation of the arrhythmogenic risk of any candidate drug is needed. Methods and results Using the O'Hara-Rudy computer model of human ventricular wedge, we evaluate the arrhythmogenic potential of clinical factors that can further alter repolarization in COVID-19 patients in addition to hydroxychloroquine (HCQ) and azithromycin (AZM) such as tachycardia, hypokalaemia, and subclinical to mild long QT syndrome. Hydroxychloroquine and AZM drugs have little impact on QT duration and do not induce any substrate prone to arrhythmia in COVID-19 patients with normal cardiac repolarization reserve. Nevertheless, in every tested condition in which this reserve is reduced, the model predicts larger electrocardiogram impairments, as with dofetilide. In subclinical conditions, the model suggests that mexiletine limits the deleterious effects of AZM and HCQ. Conclusion By studying the HCQ and AZM co-administration case, we show that the easy-to-use O'Hara-Rudy model can be applied to assess the QT-prolongation potential of off-label drugs, beyond HCQ and AZM, in different conditions representative of COVID-19 patients and to evaluate the potential impact of additional drug used to limit the arrhythmogenic risk.
- Published
- 2021
- Full Text
- View/download PDF
16. Molecular mechanisms of Brugada syndrome related to p.R211H variant in RRAD gene
- Author
-
Cyrielle Jajkiewicz, Tessier Agnès, Damien Minois, Derangeon Mickael, and Flavien Charpentier
- Subjects
Cardiology and Cardiovascular Medicine - Published
- 2023
- Full Text
- View/download PDF
17. PO-04-164 MOLECULAR MECHANISMS OF BRUGADA SYNDROME RELATED TO P.R211H MUTATION IN GENE
- Author
-
Cyrielle Jajkiewicz, Agnès Tessier, Damien Minois, Mickaël Derangeon, and Flavien Charpentier
- Subjects
Physiology (medical) ,Cardiology and Cardiovascular Medicine - Published
- 2023
- Full Text
- View/download PDF
18. Infanticide vs. inherited cardiac arrhythmias
- Author
-
Vicki Athanasopoulos, Todor Arsov, Matthew C. Cook, Sui Rong Wayne Chen, Peter J. Schwartz, Ruiwu Wang, Deborah DiSilvestre, Hariharan Raju, David A Wallace, Richard Redon, Marcin Adamski, Helene Halkjær Jensen, Ivy E. Dick, Antony Kaspi, Melanie Bahlo, Matthew A. Field, Jinhong Wei, Lia Crotti, Michael Toft Overgaard, Mette Nyegaard, Haloom Rafehi, Bárbara B Ribeiro de Oliveira-Mendes, Carola G. Vinuesa, Yafei Zhang, Flavien Charpentier, Isabelle Baró, Malene Brohus, Aalborg University [Denmark] (AAU), Australian National University (ANU), Columbia University Irving Medical Center (CUIMC), Aarhus University [Aarhus], Istituto Auxologico Italiano, Università degli Studi di Milano-Bicocca [Milano] (UNIMIB), unité de recherche de l'institut du thorax UMR1087 UMR6291 (ITX), Université de Nantes - UFR de Médecine et des Techniques Médicales (UFR MEDECINE), Université de Nantes (UN)-Université de Nantes (UN)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Macquarie University [Sydney], University of Maryland School of Medicine, University of Maryland System, University of Calgary, The Walter and Eliza Hall Institute of Medical Research (WEHI), University of Melbourne, Brohus, M, Arsov, T, Wallace, D, Jensen, H, Nyegaard, M, Crotti, L, Adamski, M, Zhang, Y, Field, M, Athanasopoulos, V, Baró, I, Ribeiro de Oliveira-Mendes, B, Redon, R, Charpentier, F, Raju, H, Disilvestre, D, Wei, J, Wang, R, Rafehi, H, Kaspi, A, Bahlo, M, Dick, I, Chen, S, Cook, M, Vinuesa, C, Overgaard, M, and Schwartz, P
- Subjects
Tachycardia ,MED/03 - GENETICA MEDICA ,Infanticide ,030204 cardiovascular system & hematology ,Ventricular tachycardia ,Ryanodine receptor 2 ,Sudden cardiac death ,ACTIVATION ,BSN ,Death, Sudden ,0302 clinical medicine ,VENTRICULAR-TACHYCARDIA ,AcademicSubjects/MED00200 ,CALMODULIN ,Child ,0303 health sciences ,High-Throughput Nucleotide Sequencing ,Smothering ,Sudden unexpected death ,3. Good health ,Child, Preschool ,Cardiology ,Female ,INACTIVATION ,medicine.symptom ,Cardiology and Cardiovascular Medicine ,medicine.medical_specialty ,Channelopathies and Cardiomyopathies ,Long QT syndrome ,Catecholaminergic polymorphic ventricular tachycardia ,03 medical and health sciences ,[SDV.MHEP.CSC]Life Sciences [q-bio]/Human health and pathology/Cardiology and cardiovascular system ,Clinical Research ,Physiology (medical) ,Internal medicine ,BASSOON ,medicine ,Humans ,030304 developmental biology ,MUTATIONS ,business.industry ,Calmodulinopathy ,Australia ,Infant ,Cardiac arrhythmia ,Arrhythmias, Cardiac ,Ryanodine Receptor Calcium Release Channel ,MED/11 - MALATTIE DELL'APPARATO CARDIOVASCOLARE ,medicine.disease ,Death, Sudden, Cardiac ,Tachycardia, Ventricular ,CALM2 ,business - Abstract
Aims In 2003, an Australian woman was convicted by a jury of smothering and killing her four children over a 10-year period. Each child died suddenly and unexpectedly during a sleep period, at ages ranging from 19 days to 18 months. In 2019 we were asked to investigate if a genetic cause could explain the children’s deaths as part of an inquiry into the mother’s convictions. Methods and results Whole genomes or exomes of the mother and her four children were sequenced. Functional analysis of a novel CALM2 variant was performed by measuring Ca2+-binding affinity, interaction with calcium channels and channel function. We found two children had a novel calmodulin variant (CALM2 G114R) that was inherited maternally. Three genes (CALM1-3) encode identical calmodulin proteins. A variant in the corresponding residue of CALM3 (G114W) was recently reported in a child who died suddenly at age 4 and a sibling who suffered a cardiac arrest at age 5. We show that CALM2 G114R impairs calmodulin's ability to bind calcium and regulate two pivotal calcium channels (CaV1.2 and RyR2) involved in cardiac excitation contraction coupling. The deleterious effects of G114R are similar to those produced by G114W and N98S, which are considered arrhythmogenic and cause sudden cardiac death in children. Conclusion A novel functional calmodulin variant (G114R) predicted to cause idiopathic ventricular fibrillation, catecholaminergic polymorphic ventricular tachycardia, or mild long QT syndrome was present in two children. A fatal arrhythmic event may have been triggered by their intercurrent infections. Thus, calmodulinopathy emerges as a reasonable explanation for a natural cause of their deaths., Graphical Abstract
- Published
- 2020
- Full Text
- View/download PDF
19. Human MuStem Cell Grafting into Infarcted Rat Heart Attenuates Adverse Tissue Remodeling and Preserves Cardiac Function
- Author
-
Thibaut Larcher, Laëtitia Guével, Laurence Dubreil, Agnès Hivonnait, Romain Fleurisson, Karl Rouger, Flavien Charpentier, Isabelle Leroux, Candice Babarit, Alice Rannou, Séverine Ménoret, Mireille Ledevin, Ignacio Anegon, and Gilles Toumaniantz
- Subjects
0301 basic medicine ,Cell type ,Pathology ,medicine.medical_specialty ,lcsh:QH426-470 ,muscle-derived stem cells ,regenerative medicine ,Infarction ,Article ,Cell therapy ,03 medical and health sciences ,0302 clinical medicine ,Fibrosis ,human adult stem cell ,Genetics ,medicine ,Myocardial infarction ,lcsh:QH573-671 ,Molecular Biology ,MuStem cells ,lcsh:Cytology ,business.industry ,Skeletal muscle ,medicine.disease ,3. Good health ,Transplantation ,lcsh:Genetics ,myocardial infarction ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Molecular Medicine ,cell therapy ,Stem cell ,business - Abstract
Myocardial infarction is one of the leading causes of mortality and morbidity worldwide. Whereas transplantation of several cell types into the infarcted heart has produced promising preclinical results, clinical studies using analogous human cells have shown limited structural and functional benefits. In dogs and humans, we have described a type of muscle-derived stem cells termed MuStem cells that efficiently promoted repair of injured skeletal muscle. Enhanced survival rate, long-term engraftment, and participation in muscle fiber formation were reported, leading to persistent tissue remodeling and clinical benefits. With the consideration of these features that are restricted or absent in cells tested so far for myocardial infarction, we wanted to investigate the capacity of human MuStem cells to repair infarcted hearts. Their local administration in immunodeficient rats 1 week after induced infarction resulted in reduced fibrosis and increased angiogenesis 3 weeks post-transplantation. Importantly, foci of human fibers were detected in the infarct site. Treated rats also showed attenuated left-ventricle dilation and preservation of contractile function. Interestingly, no spontaneous arrhythmias were observed. Our findings support the potential of MuStem cells, which have already been proposed as therapeutic candidates for dystrophic patients, to treat myocardial infarction and position them as an attractive tool for muscle-regenerative medicine., Graphical Abstract, Rannou and colleagues determine the repair potential of human muscle-derived stem cells faced with myocardial infarction. Following transplantation into rat infarcted hearts, they implant and form muscle fibers and cardiomyocytes. Also, they reduce fibrosis, enhance angiogenesis, and preserve cardiac function, positioning them as a promising therapeutic avenue for heart diseases.
- Published
- 2020
- Full Text
- View/download PDF
20. Beneficial effects of a cardiac gene therapy with phosphodiesterase pde2a in a mouse model of heart failure
- Author
-
Rima Kamel, Aurélia Bourcier, Jean Piero Margaria, Julie Larue, Audrey Varin, Alessandra Ghigo, Agnès Hivonnait, Françoise Mercier-Nomé, Delphine Mika, Vincent Algalarrondo, Flavien Charpentier, Emilio Hirsch, Grégoire Vandecasteele, Rodolphe Fischmeister, and Jérôme Leroy
- Subjects
Cardiology and Cardiovascular Medicine ,Molecular Biology - Published
- 2022
- Full Text
- View/download PDF
21. Molecular mechanisms of Brugada syndrome related to p.R211H mutation in RRAD gene
- Author
-
Cyrielle Jajkiewicz, Tessier Agnès, Derangeon Mickael, and Flavien Charpentier
- Subjects
Cardiology and Cardiovascular Medicine - Published
- 2022
- Full Text
- View/download PDF
22. Targeting the Microtubule EB1-CLASP2 Complex Modulates Na(V)1.5 at Intercalated Discs
- Author
-
Mischa Klerk, Calum A. MacRae, Marta Pérez-Hernández, Elisabeth M. Lodder, Paul W. Burridge, Christiaan C. Veerman, Isabella Mengarelli, Richard Redon, Vincent Portero, Gerard A Marchal, Carol Ann Remme, Mario Delmar, Franck Potet, Flavien Charpentier, Kaomei Guan, Svitlana Podliesna, Nuo Yu, Carlos G. Vanoye, Alfred Lewis George, David Y. Chiang, Niels Galjart, Simona Casini, Mariam Jouni, Arie O. Verkerk, Eli Rothenberg, Connie R. Bezzina, Cardiology, Graduate School, ACS - Heart failure & arrhythmias, Human Genetics, Medical Biology, APH - Methodology, and Cell biology
- Subjects
biology ,Physiology ,Chemistry ,cardiac ,Sodium channel ,Myocardium ,Fluorescence recovery after photobleaching ,Nav1.5 ,biology.organism_classification ,electrophysiology ,zebrafish ,Article ,Sodium Channels ,Cell biology ,Microtubule ,Cytoplasm ,GSK-3 ,biology.protein ,microscopy ,Cardiology and Cardiovascular Medicine ,Induced pluripotent stem cell ,Zebrafish ,myocyte, cardiac ,myocyte ,microtubule - Abstract
Rationale: Loss-of-function of the cardiac sodium channel Na V 1.5 causes conduction slowing and arrhythmias. Na V 1.5 is differentially distributed within subcellular domains of cardiomyocytes, with sodium current ( I Na ) being enriched at the intercalated discs (ID). Various pathophysiological conditions associated with lethal arrhythmias display ID-specific I Na reduction, but the mechanisms underlying microdomain-specific targeting of Na V 1.5 remain largely unknown. Objective: To investigate the role of the microtubule plus-end tracking proteins EB1 (end-binding protein 1) and CLASP2 (cytoplasmic linker associated protein 2) in mediating Na V 1.5 trafficking and subcellular distribution in cardiomyocytes. Methods and Results: EB1 overexpression in human-induced pluripotent stem cell-derived cardiomyocytes resulted in enhanced whole-cell I Na , increased action potential upstroke velocity ( V max ), and enhanced Na V 1.5 localization at the plasma membrane as detected by multicolor stochastic optical reconstruction microscopy. Fluorescence recovery after photobleaching experiments in HEK293A cells demonstrated that EB1 overexpression promoted Na V 1.5 forward trafficking. Knockout of MAPRE1 in human induced pluripotent stem cell-derived cardiomyocytes led to reduced whole-cell I Na , decreased V max , and action potential duration (APD) prolongation. Similarly, acute knockout of the MAPRE1 homolog in zebrafish ( mapre1b ) resulted in decreased ventricular conduction velocity and V max as well as increased APD. Stochastic optical reconstruction microscopy imaging and macropatch I Na measurements showed that subacute treatment (2–3 hours) with SB216763 (SB2), a GSK3β (glycogen synthase kinase 3β) inhibitor known to modulate CLASP2-EB1 interaction, reduced GSK3β localization and increased Na V 1.5 and I Na preferentially at the ID region of wild-type murine ventricular cardiomyocytes. By contrast, SB2 did not affect whole cell I Na or Na V 1.5 localization in cardiomyocytes from Clasp2 -deficient mice, uncovering the crucial role of CLASP2 in SB2-mediated modulation of Na V 1.5 at the ID. Conclusions: Our findings demonstrate the modulatory effect of the microtubule plus-end tracking protein EB1 on Na V 1.5 trafficking and function, and identify the EB1-CLASP2 complex as a target for preferential modulation of I Na within the ID region of cardiomyocytes.
- Published
- 2021
23. A consistent arrhythmogenic trait in Brugada syndrome cellular phenotype
- Author
-
Aude Derevier, Robin Canac, Isabelle Baró, Guillaume Lamirault, Vincent Probst, Jean-Jacques Schott, Anne Gaignerie, Aurore Girardeau, Richard Redon, Julien Barc, Flavien Charpentier, Bastien Cimarosti, Patricia Lemarchand, Nathalie Gaborit, Virginie Forest, Zeina R Al Sayed, Nadjet Belbachir, Mariam Jouni, Gildas Loussouarn, Kazem Zibara, Pierre Olchesqui, Eric Charpentier, Jean-Baptiste Gourraud, Caroline Chariau, unité de recherche de l'institut du thorax UMR1087 UMR6291 (ITX), Université de Nantes - UFR de Médecine et des Techniques Médicales (UFR MEDECINE), Université de Nantes (UN)-Université de Nantes (UN)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Structure fédérative de recherche François Bonamy (SFR François Bonamy), Université de Nantes (UN)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Institut de Recherche en Santé de l'Université de Nantes (IRS-UN), Lebanese University [Beirut] (LU), gaborit, nathalie, Unité de recherche de l'institut du thorax (ITX-lab), Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université de Nantes - UFR de Médecine et des Techniques Médicales (UFR MEDECINE), and Université de Nantes (UN)-Université de Nantes (UN)
- Subjects
Medicine (General) ,Heart Ventricles ,[SDV]Life Sciences [q-bio] ,Induced Pluripotent Stem Cells ,Medicine (miscellaneous) ,030204 cardiovascular system & hematology ,Letter to Editor ,NAV1.5 Voltage-Gated Sodium Channel ,03 medical and health sciences ,R5-920 ,0302 clinical medicine ,Text mining ,Humans ,Medicine ,Myocytes, Cardiac ,ComputingMilieux_MISCELLANEOUS ,Brugada Syndrome ,030304 developmental biology ,Brugada syndrome ,Genetics ,0303 health sciences ,business.industry ,Gene Expression Profiling ,Sodium ,Electric Conductivity ,Arrhythmias, Cardiac ,medicine.disease ,Cellular phenotype ,[SDV] Life Sciences [q-bio] ,Phenotype ,Gene Expression Regulation ,Case-Control Studies ,Mutation ,Trait ,Molecular Medicine ,business ,Biomarkers - Abstract
International audience
- Published
- 2021
- Full Text
- View/download PDF
24. Cardiac gene therapy with PDE2A limits ventricular remodeling, dysfunction and arrhythmias promoted in mice by chronic infusion with catecholamines
- Author
-
Rima Kamel, Aurélia Bourcier, Jean Piero Margaria, Audrey Varin, Alessandra Ghigo, Agnès Hivonnait, Françoise Nomé-Mercier, Delphine Mika, Vincent Algalarrondo, Emilio Hirsch, Flavien Charpentier, Grégoire Vandecasteele, Rodolphe Fischmeister, and Jérôme Leroy
- Subjects
Cardiology and Cardiovascular Medicine - Published
- 2022
- Full Text
- View/download PDF
25. Gap-134, a Connexin43 activator, prevents age-related development of ventricular fibrosis in Scn5a− mice
- Author
-
Hervé Le Marec, Jacques Lebreton, Agnès Carcouët, Audrey Donnart, Audrey Bihouée, Justine Patin, Isabelle Baró, Mickaël Derangeon, Agnès Hivonnait, Claire Castro, Arnaud Tessier, Flavien Charpentier, Marja Steenman, unité de recherche de l'institut du thorax UMR1087 UMR6291 (ITX), Université de Nantes - UFR de Médecine et des Techniques Médicales (UFR MEDECINE), Université de Nantes (UN)-Université de Nantes (UN)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Chimie Et Interdisciplinarité : Synthèse, Analyse, Modélisation (CEISAM), Université de Nantes - UFR des Sciences et des Techniques (UN UFR ST), Université de Nantes (UN)-Université de Nantes (UN)-Centre National de la Recherche Scientifique (CNRS)-Institut de Chimie du CNRS (INC), Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université de Nantes - UFR de Médecine et des Techniques Médicales (UFR MEDECINE), Université de Nantes (UN)-Université de Nantes (UN), Université Paris 1 Panthéon-Sorbonne - UFR Histoire (UP1 UFR09), Université Paris 1 Panthéon-Sorbonne (UP1), Université de Nantes - Faculté des Sciences et des Techniques, and Université de Nantes (UN)-Université de Nantes (UN)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Mice, 129 Strain ,Proline ,Cardiac fibrosis ,[SDV]Life Sciences [q-bio] ,NAV1.5 Voltage-Gated Sodium Channel ,03 medical and health sciences ,0302 clinical medicine ,[SDV.MHEP.CSC]Life Sciences [q-bio]/Human health and pathology/Cardiology and cardiovascular system ,Fibrosis ,Internal medicine ,Cardiac conduction ,medicine ,Animals ,[CHIM]Chemical Sciences ,Phosphorylation ,Cells, Cultured ,ComputingMilieux_MISCELLANEOUS ,Cell Proliferation ,Mice, Knockout ,Pharmacology ,Ventricular Remodeling ,Activator (genetics) ,business.industry ,Myocardium ,Fronce Danegaptide ZP 1609 ,Gap junction ,Fibroblasts ,Nantes ,medicine.disease ,Up-Regulation ,Disease Models, Animal ,030104 developmental biology ,Endocrinology ,Connexin 43 ,030220 oncology & carcinogenesis ,Benzamides ,Knockout mouse ,cardiovascular system ,F-44()(}() ,Pyrazoles ,Cardiomyopathies ,business ,Signal Transduction ,Transforming growth factor - Abstract
International audience; Na.LS Gap junction GW788388 Cardiac fibroblasts Down-regulation of Connexin43 (Cx43) has often been associated with the development of cardiac fibrosis. We showed previously that ScnSa heterozygous knockout mice (ScnS a+ 1-¡, which mimic familial progressive cardiac conduction defect, exhibit an age-dependent decrease of Cx43 expression and phosphorylation concomitantly with activation of TGF-¡3 pathway and fibrosis development in the myocardium between 45 and 60 weeks of age. The aim of this study was to investigate whether Gap-134 prevents Cx43 down-regulation with age and fibrosis development in ScnSa + t-mice. We observed in 60-week-old ScnSa+I-mouse heart a Cx43 expression and localization remodeling correlated with fibrosis. Chronic administration of a potent and selective gap junction modifier, Gap-134 (danegaptide), between 45 and 60 weeks, increased Cx43 expression and phosphorylation on serine 368 and prevented Cx43 delocalization. Furthermore, we found that Gap-134 prevented fibrosis despite the persistence of the conduction defects and the TGF-¡3 canonical pathway activation. ln conclusion, the present study demonstrates that the age-dependent decrease of Cx43 expression is involved in the ventricular fibrotic process occurring in ScnSa+I-mice. Finally, our study suggests that gap junction modifier, such as Gap-134, could be an effective anti-fibrotic agent in the context of age-dependent fibrosis in progressive cardiac conduction disease.
- Published
- 2020
- Full Text
- View/download PDF
26. KCNQ1 Antibodies for Immunotherapy of Long QT Syndrome Type 2
- Author
-
Jonas P. Wepfer, Jan P. Kucera, Jin Li, Virginie Forest, Flavien Charpentier, and Ange Maguy
- Subjects
endocrine system diseases ,Long QT syndrome ,medicine.medical_treatment ,610 Medicine & health ,CHO Cells ,030204 cardiovascular system & hematology ,Pharmacology ,medicine.disease_cause ,Proof of Concept Study ,Protein Structure, Secondary ,Membrane Potentials ,Autoimmunity ,Targeted therapy ,03 medical and health sciences ,Cricetulus ,0302 clinical medicine ,Cricetinae ,medicine ,Animals ,Humans ,Myocytes, Cardiac ,030212 general & internal medicine ,Patch clamp ,Induced pluripotent stem cell ,Cells, Cultured ,Autoantibodies ,business.industry ,urogenital system ,Chinese hamster ovary cell ,Immunotherapy ,medicine.disease ,Long QT Syndrome ,Electrophysiology ,HEK293 Cells ,KCNQ1 Potassium Channel ,570 Life sciences ,biology ,Rabbits ,Cardiology and Cardiovascular Medicine ,business - Abstract
Background Patients with long QT syndrome (LQTS) are predisposed to life-threatening arrhythmias. A delay in cardiac repolarization is characteristic of the disease. Pharmacotherapy, implantable cardioverter-defibrillators, and left cardiac sympathetic denervation are part of the current treatment options, but no targeted therapy for LQTS exists to date. Previous studies indicate that induced autoimmunity against the voltage-gated KCNQ1 K+ channels accelerates cardiac repolarization. Objectives However, a causative relationship between KCNQ1 antibodies and the observed electrophysiological effects has never been demonstrated, and thus presents the aim of this study. Methods The authors purified KCNQ1 antibodies and performed whole-cell patch clamp experiments as well as single-channel recordings on Chinese hamster ovary cells overexpressing IKs channels. The effect of purified KCNQ1 antibodies on human cardiomyocytes derived from induced pluripotent stem cells was then studied. Results The study demonstrated that KCNQ1 antibodies underlie the previously observed increase in repolarizing IKs current. The antibodies shift the voltage dependence of activation and slow the deactivation of IKs. At the single-channel level, KCNQ1 antibodies increase the open time and probability of the channel. In models of LQTS type 2 (LQTS2) using human induced pluripotent stem cell-derived cardiomyocytes, KCNQ1 antibodies reverse the prolonged cardiac repolarization and abolish arrhythmic activities. Conclusions Here, the authors provide the first direct evidence that KCNQ1 antibodies act as agonists on IKs channels. Moreover, KCNQ1 antibodies were able to restore alterations in cardiac repolarization and most importantly to suppress arrhythmias in LQTS2. KCNQ1 antibody therapy may thus present a novel promising therapeutic approach for LQTS2.
- Published
- 2020
- Full Text
- View/download PDF
27. Inhibition of G protein-gated K
- Author
-
Isabelle, Bidaud, Antony Chung You, Chong, Agnes, Carcouet, Stephan De, Waard, Flavien, Charpentier, Michel, Ronjat, Michel De, Waard, Dirk, Isbrandt, Kevin, Wickman, Anne, Vincent, Matteo E, Mangoni, and Pietro, Mesirca
- Subjects
Potassium Channels ,Calcium Channels, L-Type ,Arrhythmias ,Article ,NAV1.5 Voltage-Gated Sodium Channel ,Bee Venoms ,Disease Models, Animal ,Mice ,GTP-Binding Proteins ,Heart Conduction System ,Heart Rate ,Bradycardia ,Potassium Channel Blockers ,Animals ,Cardiomyopathies ,Sinoatrial Node - Abstract
Sinus node (SAN) dysfunction (SND) manifests as low heart rate (HR) and is often accompanied by atrial tachycardia or atrioventricular (AV) block. The only currently available therapy for chronic SND is the implantation of an electronic pacemaker. Because of the growing burden of SND in the population, new pharmacological therapies of chronic SND and heart block are desirable. We developed a collection of genetically modified mouse strains recapitulating human primary SND associated with different degrees of AV block. These mice were generated with genetic ablation of L-type Cav1.3 (Cav1.3−/−), T-type Cav3.1 (Cav3.1−/−), or both (Cav1.3−/−/Cav3.1−/−). We also studied mice haplo-insufficient for the Na+ channel Nav1.5 (Nav1.5+/) and mice in which the cAMP-dependent regulation of hyperpolarization-activated f-(HCN4) channels has been abolished (HCN4-CNBD). We analysed, by telemetric ECG recording, whether pharmacological inhibition of the G-protein-activated K+ current (IKACh) by the peptide tertiapin-Q could improve HR and AV conduction in these mouse strains. Tertiapin-Q significantly improved the HR of Cav1.3−/− (19%), Cav1.3−/−/Cav3.1−/− (23%) and HCN4-CNBD (14%) mice. Tertiapin-Q also improved cardiac conduction of Nav1.5+/− mice by 24%. Our data suggest that the development of pharmacological IKACh inhibitors for the management of SND and conduction disease is a viable approach.
- Published
- 2020
28. Cardiac Overexpression of PDE4B Blunts β-Adrenergic Response and Maladaptive Remodeling in Heart Failure
- Author
-
Delphine Mika, V. Algalarrondo, Valérie Domergue, Aurelia Bourcier, Matthieu Dessillons, Pauline Robert, Kaouter Bouadjel, Jérôme Leroy, Philippe Mateo, Marta Lindner, Florence Lefebvre, Jean-Baptiste Michel, A. Varin, Flavien Charpentier, Rodolphe Fischmeister, Jean Piero Margaria, Jane-Lise Samuel, Ibrahim Bedioune, Sarah Karam, Patrick Lechêne, Susana Gomez, Emilio Hirsch, Françoise Gaudin, Grégoire Vandecasteele, Charlène Coquard, Alessandra Ghigo, Signalisation et physiopathologie cardiovasculaire (CARPAT), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris-Saclay, Department of Molecular Biotechnologies and Health Sciences [Torino, Italy] (Hematology Division), Università degli studi di Torino (UNITO), Ingénierie et Plateformes au Service de l'Innovation Thérapeutique (IPSIT), Institut National de la Santé et de la Recherche Médicale (INSERM)-Institut de Chimie du CNRS (INC)-Université Paris-Saclay-Centre National de la Recherche Scientifique (CNRS), Marqueurs cardiovasculaires en situation de stress (MASCOT (UMR_S_942 / U942)), Institut National de la Santé et de la Recherche Médicale (INSERM)-Groupe Hospitalier Saint Louis - Lariboisière - Fernand Widal [Paris], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Centre National de la Recherche Scientifique (CNRS)-Université de Paris (UP)-Université Sorbonne Paris Nord, Laboratoire de Recherche Vasculaire Translationnelle (LVTS (UMR_S_1148 / U1148)), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Paris (UP)-Université Sorbonne Paris Nord, unité de recherche de l'institut du thorax UMR1087 UMR6291 (ITX), Université de Nantes - UFR de Médecine et des Techniques Médicales (UFR MEDECINE), Université de Nantes (UN)-Université de Nantes (UN)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, FISCHMEISTER, RODOLPHE, Università degli studi di Torino = University of Turin (UNITO), Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Centre National de la Recherche Scientifique (CNRS)-Université Paris Cité (UPCité)-Université Sorbonne Paris Nord, Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris Cité (UPCité)-Université Sorbonne Paris Nord, Unité de recherche de l'institut du thorax (ITX-lab), Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université de Nantes - UFR de Médecine et des Techniques Médicales (UFR MEDECINE), and Université de Nantes (UN)-Université de Nantes (UN)
- Subjects
Genetic enhancement ,Gene Expression ,heart failure ,030204 cardiovascular system & hematology ,phosphodiesterase 4 ,Negative regulator ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,PDE4B ,Transduction, Genetic ,Myocytes, Cardiac ,0303 health sciences ,Ventricular Remodeling ,cardiac remodeling ,cyclic AMP ,genetic therapy ,transgenic mice ,Phosphodiesterase ,Adrenergic beta-Agonists ,[SDV.MHEP.CSC] Life Sciences [q-bio]/Human health and pathology/Cardiology and cardiovascular system ,3. Good health ,Phenotype ,Heart Function Tests ,[SDV.BBM.GTP] Life Sciences [q-bio]/Biochemistry, Molecular Biology/Genomics [q-bio.GN] ,Disease Susceptibility ,Cardiology and Cardiovascular Medicine ,Genetically modified mouse ,Adenosine monophosphate ,medicine.medical_specialty ,Genetic Vectors ,Mice, Transgenic ,03 medical and health sciences ,[SDV.MHEP.CSC]Life Sciences [q-bio]/Human health and pathology/Cardiology and cardiovascular system ,[SDV.BBM.GTP]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Genomics [q-bio.GN] ,Physiology (medical) ,Internal medicine ,Receptors, Adrenergic, beta ,medicine ,Animals ,Humans ,030304 developmental biology ,business.industry ,Myocardium ,Isoproterenol ,β adrenergic ,medicine.disease ,Cyclic Nucleotide Phosphodiesterases, Type 4 ,Disease Models, Animal ,Endocrinology ,chemistry ,Heart failure ,business - Abstract
Background: The cyclic AMP (adenosine monophosphate; cAMP)-hydrolyzing protein PDE4B (phosphodiesterase 4B) is a key negative regulator of cardiac β-adrenergic receptor stimulation. PDE4B deficiency leads to abnormal Ca 2+ handling and PDE4B is decreased in pressure overload hypertrophy, suggesting that increasing PDE4B in the heart is beneficial in heart failure. Methods: We measured PDE4B expression in human cardiac tissues and developed 2 transgenic mouse lines with cardiomyocyte-specific overexpression of PDE4B and an adeno-associated virus serotype 9 encoding PDE4B. Myocardial structure and function were evaluated by echocardiography, ECG, and in Langendorff-perfused hearts. Also, cAMP and PKA (cAMP dependent protein kinase) activity were monitored by Förster resonance energy transfer, L-type Ca 2+ current by whole-cell patch-clamp, and cardiomyocyte shortening and Ca 2+ transients with an Ionoptix system. Heart failure was induced by 2 weeks infusion of isoproterenol or transverse aortic constriction. Cardiac remodeling was evaluated by serial echocardiography, morphometric analysis, and histology. Results: PDE4B protein was decreased in human failing hearts. The first PDE4B-transgenic mouse line (TG15) had a ≈15-fold increase in cardiac cAMP-PDE activity and a ≈30% decrease in cAMP content and fractional shortening associated with a mild cardiac hypertrophy that resorbed with age. Basal ex vivo myocardial function was unchanged, but β-adrenergic receptor stimulation of cardiac inotropy, cAMP, PKA, L-type Ca 2+ current, Ca 2+ transients, and cell contraction were blunted. Endurance capacity and life expectancy were normal. Moreover, these mice were protected from systolic dysfunction, hypertrophy, lung congestion, and fibrosis induced by chronic isoproterenol treatment. In the second PDE4B-transgenic mouse line (TG50), markedly higher PDE4B overexpression, resulting in a ≈50-fold increase in cardiac cAMP-PDE activity caused a ≈50% decrease in fractional shortening, hypertrophy, dilatation, and premature death. In contrast, mice injected with adeno-associated virus serotype 9 encoding PDE4B (10 12 viral particles/mouse) had a ≈50% increase in cardiac cAMP-PDE activity, which did not modify basal cardiac function but efficiently prevented systolic dysfunction, apoptosis, and fibrosis, while attenuating hypertrophy induced by chronic isoproterenol infusion. Similarly, adeno-associated virus serotype 9 encoding PDE4B slowed contractile deterioration, attenuated hypertrophy and lung congestion, and prevented apoptosis and fibrotic remodeling in transverse aortic constriction. Conclusions: Our results indicate that a moderate increase in PDE4B is cardioprotective and suggest that cardiac gene therapy with PDE4B might constitute a new promising approach to treat heart failure.
- Published
- 2020
- Full Text
- View/download PDF
29. Functional Impact of BeKm-1, a High-Affinity hERG Blocker, on Cardiomyocytes Derived from Human-Induced Pluripotent Stem Cells
- Author
-
Stephan De Waard, Jérôme Montnach, Barbara Ribeiro, Sébastien Nicolas, Virginie Forest, Flavien Charpentier, Matteo Elia Mangoni, Nathalie Gaborit, Michel Ronjat, Gildas Loussouarn, Patricia Lemarchand, Michel De Waard, Loussouarn, Gildas, Unité de recherche de l'institut du thorax (ITX-lab), Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université de Nantes - UFR de Médecine et des Techniques Médicales (UFR MEDECINE), Université de Nantes (UN)-Université de Nantes (UN), Inflammasome NLRP3 – NLRP3 Inflammasome, Centre International de Recherche en Infectiologie - UMR (CIRI), École normale supérieure - Lyon (ENS Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-École normale supérieure - Lyon (ENS Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), unité de recherche de l'institut du thorax UMR1087 UMR6291 (ITX), Université de Nantes - UFR de Médecine et des Techniques Médicales (UFR MEDECINE), Université de Nantes (UN)-Université de Nantes (UN)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Centre International de Recherche en Infectiologie (CIRI), École normale supérieure de Lyon (ENS de Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Université Jean Monnet - Saint-Étienne (UJM)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-École normale supérieure de Lyon (ENS de Lyon)-Université Claude Bernard Lyon 1 (UCBL), and Université de Lyon-Université de Lyon-Université Jean Monnet - Saint-Étienne (UJM)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
ERG1 Potassium Channel ,Patch-Clamp Techniques ,Pyridines ,[SDV.MHEP.PHY] Life Sciences [q-bio]/Human health and pathology/Tissues and Organs [q-bio.TO] ,Induced Pluripotent Stem Cells ,Action Potentials ,Scorpion Venoms ,[SDV.BBM.BP] Life Sciences [q-bio]/Biochemistry, Molecular Biology/Biophysics ,Models, Biological ,Article ,lcsh:Chemistry ,Piperidines ,[SDV.MHEP.CSC]Life Sciences [q-bio]/Human health and pathology/Cardiology and cardiovascular system ,Phenethylamines ,Potassium Channel Blockers ,[SDV.MHEP.PHY]Life Sciences [q-bio]/Human health and pathology/Tissues and Organs [q-bio.TO] ,Humans ,Myocytes, Cardiac ,hERG ,cardiovascular diseases ,lcsh:QH301-705.5 ,Sulfonamides ,Ion Transport ,Cell Differentiation ,[SDV.MHEP.CSC] Life Sciences [q-bio]/Human health and pathology/Cardiology and cardiovascular system ,[SDV.BBM.BP]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Biophysics ,Long QT Syndrome ,hiPS-cardiomyocytes ,HEK293 Cells ,lcsh:Biology (General) ,lcsh:QD1-999 ,Potassium ,cardiovascular system ,BeKm-1 ,LQTS ,Calcium ,Calcium Channels ,Anti-Arrhythmia Agents - Abstract
IKr current, a major component of cardiac repolarization, is mediated by human Ether-à, go-go-Related Gene (hERG, Kv11.1) potassium channels. The blockage of these channels by pharmacological compounds is associated to drug-induced long QT syndrome (LQTS), which is a life-threatening disorder characterized by ventricular arrhythmias and defects in cardiac repolarization that can be illustrated using cardiomyocytes derived from human-induced pluripotent stem cells (hiPS-CMs). This study was meant to assess the modification in hiPS-CMs excitability and contractile properties by BeKm-1, a natural scorpion venom peptide that selectively interacts with the extracellular face of hERG, by opposition to reference compounds that act onto the intracellular face. Using an automated patch-clamp system, we compared the affinity of BeKm-1 for hERG channels with some reference compounds. We fully assessed its effects on the electrophysiological, calcium handling, and beating properties of hiPS-CMs. By delaying cardiomyocyte repolarization, the peptide induces early afterdepolarizations and reduces spontaneous action potentials, calcium transients, and contraction frequencies, therefore recapitulating several of the critical phenotype features associated with arrhythmic risk in drug-induced LQTS. BeKm-1 exemplifies an interesting reference compound in the integrated hiPS-CMs cell model for all drugs that may block the hERG channel from the outer face. Being a peptide that is easily modifiable, it will serve as an ideal molecular platform for the design of new hERG modulators displaying additional functionalities.
- Published
- 2020
- Full Text
- View/download PDF
30. Human model of IRX5 mutations reveals key role for this transcription factor in ventricular conduction
- Author
-
Bastien Cimarosti, Guillaume Lamirault, Hanan Hamamy, Nathalie Gaborit, Céline Marionneau, Flavien Charpentier, Gildas Loussouarn, Laurent David, Kazem Zibara, Nicolas Jacob, Virginie Forest, Mariam Jouni, Caroline Chariau, Carine Bonnard, Hülya Kayserili, Bruno Reversade, Anne Gaignerie, Jean-Baptiste Gourraud, Robin Canac, Zeina R Al Sayed, Patricia Lemarchand, Aurore Girardeau, Karabey, Hülya Kayserili (ORCID 0000-0003-0376-499X & YÖK ID 7945), Reversade, Bruno, Al Sayed, Zeina R, Canac, Robin, Cimarosti, Bastien, Bonnard, Carine, Gourraud, Jean-Baptiste, Hamamy, Hanan, Girardeau, Aurore, Jouni, Mariam, Jacob, Nicolas, Gaignerie, Anne, Chariau, Caroline, David, Laurent, Forest, Virginie, Marionneau, Céline, Charpentier, Flavien, Loussouarn, Gildas, Lamirault, Guillaume, Zibara, Kazem, Lemarchand, Patricia, Gaborit, Nathalie, School of Medicine, unité de recherche de l'institut du thorax UMR1087 UMR6291 (ITX), Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université de Nantes - UFR de Médecine et des Techniques Médicales (UFR MEDECINE), Université de Nantes (UN)-Université de Nantes (UN), Agency for science, technology and research [Singapore] (A*STAR), Department of Genetic Medicine and Development [Geneva], Université de Genève (UNIGE), Koç University, Structure fédérative de recherche François Bonamy (SFR François Bonamy), Université de Nantes (UN)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Institut de Recherche en Santé de l'Université de Nantes (IRS-UN), Centre de Recherche en Transplantation et Immunologie (U1064 Inserm - CRTI), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Nantes - UFR de Médecine et des Techniques Médicales (UFR MEDECINE), Institut de transplantation urologie-néphrologie (ITUN), Université de Nantes (UN)-Centre hospitalier universitaire de Nantes (CHU Nantes), National University of Singapore (NUS), University of Amsterdam [Amsterdam] (UvA), Laboratory of Stem Cells [Lebanese, Beirut] (ER045-PRASE), Lebanese University [Beirut] (LU), This work was funded by grants from The National Research Agency [HEART iPS ANR-15-CE14-0019-01], and La Fédération Française de Cardiologie. Nathalie Gaborit was laureate of fellowships from Fondation Lefoulon-Delalande and International Incoming Fellowship FP7-PEOPLE-2012-IIF [PIIF-GA-2012-331436]. Zeina R. Al Sayed is supported by Eiffel scholarship program of Excellence (Campus France), by Doctoral School of Science and Technology-Lebanese University and The Fondation Genavie., ACS - Heart failure & arrhythmias, ARD - Amsterdam Reproduction and Development, Unité de recherche de l'institut du thorax (ITX-lab), and Université de Genève = University of Geneva (UNIGE)
- Subjects
conduction ,IRX5 mutations ,Physiology ,Transcription factor complex ,Connexin ,[SDV.BC]Life Sciences [q-bio]/Cellular Biology ,030204 cardiovascular system & hematology ,Biology ,arrhythmia ,Ventricular action potential ,03 medical and health sciences ,0302 clinical medicine ,Physiology (medical) ,transcription factors ,Cardiac conduction ,Transcription factor ,[SDV.BC] Life Sciences [q-bio]/Cellular Biology ,030304 developmental biology ,0303 health sciences ,GATA4 ,Sodium channel ,Depolarization ,Human-induced pluripotent stem cells ,Cell biology ,human induced pluripotent stem cells ,cardiovascular system ,IRX5 ,Cardiology and Cardiovascular Medicine ,Hamamy syndrome - Abstract
Aims: several inherited arrhythmic diseases have been linked to single gene mutations in cardiac ion channels and interacting proteins. However, the mechanisms underlying most arrhythmias, are thought to involve altered regulation of the expression of multiple effectors. In this study, we aimed to examine the role of a transcription factor (TF) belonging to the Iroquois homeobox family, IRX5, in cardiac electrical function. Methods and results: using human cardiac tissues, transcriptomic correlative analyses between IRX5 and genes involved in cardiac electrical activity showed that in human ventricular compartment, IRX5 expression strongly correlated to the expression of major actors of cardiac conduction, including the sodium channel, Nav1.5, and Connexin 40 (Cx40). We then generated human-induced pluripotent stem cells (hiPSCs) derived from two Hamamy syndrome-affected patients carrying distinct homozygous loss-of-function mutations in IRX5 gene. Cardiomyocytes derived from these hiPSCs showed impaired cardiac gene expression programme, including misregulation in the control of Nav1.5 and Cx40 expression. In accordance with the prolonged QRS interval observed in Hamamy syndrome patients, a slower ventricular action potential depolarization due to sodium current reduction was observed on electrophysiological analyses performed on patient-derived cardiomyocytes, confirming the functional role of IRX5 in electrical conduction. Finally, a cardiac TF complex was newly identified, composed by IRX5 and GATA4, in which IRX5 potentiated GATA4-induction of SCN5A expression. Conclusion: altogether, this work unveils a key role for IRX5 in the regulation of human ventricular depolarization and cardiac electrical conduction, providing therefore new insights into our understanding of cardiac diseases., National Research Agency; European Union (EU); Horizon 2020; Marie Curie Actions International Incoming Fellowship FP7-PEOPLE-2012-IIF; La Fédération Française de Cardiologie; Fondation LefoulonDelalande; Eiffel Scholarship Programme of Excellence (Campus France), Doctoral School of Science and Technology-Lebanese University and The Fondation Genavie
- Published
- 2020
- Full Text
- View/download PDF
31. Arrhythmias precede cardiomyopathy and remodeling of Ca2+ handling proteins in a novel model of long QT syndrome
- Author
-
Jean-Pierre Benitah, Nadjet Belbachir, Doron Shmerling, Jérôme Montnach, Isabelle Baró, Linwei Li, Agnès Carcouët, Gilles Toumaniantz, Claire Castro, Ana Maria Gomez, Gildas Loussouarn, Anne Julia Meinzinger, Flavien Charpentier, Franck F. Chizelle, Unité de recherche de l'institut du thorax (ITX-lab), Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université de Nantes - UFR de Médecine et des Techniques Médicales (UFR MEDECINE), Université de Nantes (UN)-Université de Nantes (UN), Signalisation et physiopathologie cardiovasculaire (UMRS1180), Institut National de la Santé et de la Recherche Médicale (INSERM), PolyGene AG, ANR-13-BSV1-0023,ARyRthmia,Arythmies dépendantes du calcium intracellulaire : mécanismes de la tachycardie ventriculaire catécholergique.(2013), ANR-12-BSV1-0013,PREVENTPCCD,Troubles progressifs de la conduction cardiaque liés à SCN5A: mécanismes physiopathologiques, biomarqueurs et prévention(2012), ANR-09-GENO-0003,CaRNaC,Le complexe du canal Na+ cardiaque Nav1.5 - Régulation et implication dans les arythmies(2009), European Project: 241526,EC:FP7:HEALTH,FP7-HEALTH-2009-single-stage,EUTRIGTREAT(2009), unité de recherche de l'institut du thorax UMR1087 UMR6291 (ITX), Université de Nantes - UFR de Médecine et des Techniques Médicales (UFR MEDECINE), and Université de Nantes (UN)-Université de Nantes (UN)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
0301 basic medicine ,medicine.medical_specialty ,[SDV]Life Sciences [q-bio] ,Long QT syndrome ,Cardiomyopathy ,Ranolazine ,030204 cardiovascular system & hematology ,Sudden death ,QT interval ,Afterdepolarization ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Ca2+/calmodulin-dependent protein kinase ,medicine ,cardiovascular diseases ,Molecular Biology ,ComputingMilieux_MISCELLANEOUS ,Chemistry ,medicine.disease ,3. Good health ,Phospholamban ,030104 developmental biology ,Endocrinology ,cardiovascular system ,Cardiology and Cardiovascular Medicine ,medicine.drug - Abstract
Aim Deletion of QKP1507-1509 amino-acids in SCN5A gene product, the voltage-gated Na+ channel Nav1.5, has been associated with a large phenotypic spectrum of type 3 long QT syndrome, conduction disorder, dilated cardiomyopathy and high incidence of sudden death. The aim of this study was to develop and characterize a novel model of type 3 long QT syndrome to study the consequences of the QKP1507–1509 deletion. Methods and results We generated a knock-in mouse presenting the delQKP1510–1512 mutation (Scn5a+/ΔQKP) equivalent to human deletion. Scn5a+/ΔQKP mice showed prolonged QT interval, conduction defects and ventricular arrhythmias at the age of 2 weeks, and, subsequently, structural defects and premature mortality. The mutation increased Na+ window current and generated a late Na+ current. Ventricular action potentials from Scn5a+/ΔQKP mice were prolonged. At the age of 4 weeks, Scn5a+/ΔQKP mice exhibited a remodeling leading to [Ca2+]i transients with higher amplitude and slower kinetics, combined with enhanced SR Ca2+ load. SERCA2 expression was not altered. However, total phospholamban expression was higher whereas the amount of Ca2+-calmodulin-dependent kinase II (CaMKII)-dependent T17-phosphorylated form was lower, in hearts from 4-week-old mice only. This was associated with a lower activity of CaMKII and lower calmodulin expression. In addition, Scn5a+/ΔQKP cardiomyocytes showed larger Ca2+ waves, correlated with the presence of afterdepolarizations during action potential recording. Ranolazine partially prevented action potential and QT interval prolongation in 4-week-old Scn5a+/ΔQKP mice and suppressed arrhythmias. Conclusion The Scn5a+/ΔQKP mouse model recapitulates the clinical phenotype of mutation carriers and provides new and unexpected insights into the pathological development of the disease in patients carrying the QKP1507–1509 deletion.
- Published
- 2018
- Full Text
- View/download PDF
32. An autoantibody profile detects Brugada syndrome and identifies abnormally expressed myocardial proteins
- Author
-
J Martjin Bos, Eric Schulze-Bahr, Sven Dittmann, Ardan M. Saguner, Robert M. Hamilton, Meena Fatah, Gonca Suna, Flavien Charpentier, Leonardo Bolognese, Kristopher S. Cunningham, Diptendu Chatterjee, Maurizio Pieroni, Danna A. Spears, Michael J. Ackerman, Firat Duru, Pasquale Notarstefano, Dipashree Chatterjee, University of Zurich, and Hamilton, Robert M
- Subjects
medicine.medical_specialty ,Heart Ventricles ,610 Medicine & health ,030204 cardiovascular system & hematology ,2705 Cardiology and Cardiovascular Medicine ,03 medical and health sciences ,Electrocardiography ,0302 clinical medicine ,Internal medicine ,Keratin ,Medicine ,Humans ,030304 developmental biology ,Brugada syndrome ,G alpha subunit ,Autoantibodies ,Brugada Syndrome ,chemistry.chemical_classification ,0303 health sciences ,biology ,business.industry ,fungi ,Autoantibody ,Arrhythmias, Cardiac ,medicine.disease ,Blot ,Endocrinology ,chemistry ,biology.protein ,10209 Clinic for Cardiology ,Biomarker (medicine) ,Immunohistochemistry ,Antibody ,Cardiology and Cardiovascular Medicine ,business - Abstract
Aims Brugada syndrome (BrS) is characterized by a unique electrocardiogram (ECG) pattern and life-threatening arrhythmias. However, the Type 1 Brugada ECG pattern is often transient, and a genetic cause is only identified in Methods and results For antibody (Ab) discovery, normal human ventricular myocardial proteins were solubilized and separated by isoelectric focusing (IEF) and molecular weight on two-dimensional (2D) gels and used to discover Abs by plating with sera from patients with BrS and control subjects. Target proteins were identified by mass spectrometry (MS). Brugada syndrome subjects were defined based on a consensus clinical scoring system. We assessed discovery and validation cohorts by 2D gels, western blots, and ELISA. We performed immunohistochemistry on myocardium from BrS subjects (vs. control). All (3/3) 2D gels exposed to sera from BrS patients demonstrated specific Abs to four proteins, confirmed by MS to be α-cardiac actin, α-skeletal actin, keratin, and connexin-43, vs. 0/8 control subjects. All (18/18) BrS subjects from our validation cohorts demonstrated the same Abs, confirmed by western blots, vs. 0/24 additional controls. ELISA optical densities for all Abs were elevated in all BrS subjects compared to controls. In myocardium obtained from BrS subjects, each protein, as well as SCN5A, demonstrated abnormal protein expression in aggregates. Conclusion A biomarker profile of autoantibodies against four cardiac proteins, namely α-cardiac actin, α-skeletal actin, keratin, and connexin-43, can be identified from sera of BrS patients and is highly sensitive and specific, irrespective of genetic cause for BrS. The four involved proteins, along with the SCN5A-encoded Nav1.5 alpha subunit are expressed abnormally in the myocardium of patients with BrS.
- Published
- 2019
33. RRAD mutation causes electrical and cytoskeletal defects in cardiomyocytes derived from a familial case of Brugada syndrome
- Author
-
Zeina R Al Sayed, Sabine Pattier, Hongchao Guo, Floriane Simonet, Solena Le Scouarnec, Emmanuelle Génin, Laurent David, Flavien Charpentier, Jean-Baptiste Gourraud, Caroline Chariau, Anne Gaignerie, Haodi Wu, Vincent Probst, Jean-François Deleuze, Stéphanie Bonnaud, Carol Scott, Florian Dilasser, Jean-Jacques Schott, Gervaise Loirand, Joseph C. Wu, Christian Dina, Isabelle Baró, Aurore Girardeau, Christophe Guilluy, Richard Redon, Vincent Sauzeau, Nathalie Gaborit, Sophie Burel, Matilde Karakachoff, Nadjet Belbachir, Laurence Jesel, Céline Marionneau, Vincent Portero, unité de recherche de l'institut du thorax UMR1087 UMR6291 (ITX), Université de Nantes - UFR de Médecine et des Techniques Médicales (UFR MEDECINE), Université de Nantes (UN)-Université de Nantes (UN)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Genetic and Cellular Engineering in Immunology and Regenerative Medicine (Team 2 - U1064 Inserm - CRTI), Centre de Recherche en Transplantation et Immunologie (U1064 Inserm - CRTI), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Nantes - UFR de Médecine et des Techniques Médicales (UFR MEDECINE), Université de Nantes (UN)-Université de Nantes (UN)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Nantes - UFR de Médecine et des Techniques Médicales (UFR MEDECINE), Université de Nantes (UN)-Université de Nantes (UN), Unité de recherche de l'institut du thorax (ITX-lab), Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université de Nantes - UFR de Médecine et des Techniques Médicales (UFR MEDECINE), and Le Bihan, Sylvie
- Subjects
RAD GTPase ,030204 cardiovascular system & hematology ,medicine.disease_cause ,Afterdepolarization ,03 medical and health sciences ,0302 clinical medicine ,Basic Science ,Medicine ,Missense mutation ,Brugada syndrome ,Induced pluripotent stem cell ,Endocardium ,030304 developmental biology ,0303 health sciences ,Mutation ,[SDV.MHEP] Life Sciences [q-bio]/Human health and pathology ,business.industry ,L-type calcium current ,Actin cytoskeleton ,medicine.disease ,Cell biology ,Electrophysiology ,Induced pluripotent stem cells ,Sodium current ,Cardiology and Cardiovascular Medicine ,business ,[SDV.MHEP]Life Sciences [q-bio]/Human health and pathology - Abstract
Aims The Brugada syndrome (BrS) is an inherited cardiac disorder predisposing to ventricular arrhythmias. Despite considerable efforts, its genetic basis and cellular mechanisms remain largely unknown. The objective of this study was to identify a new susceptibility gene for BrS through familial investigation. Methods and results Whole-exome sequencing performed in a three-generation pedigree with five affected members allowed the identification of one rare non-synonymous substitution (p.R211H) in RRAD, the gene encoding the RAD GTPase, carried by all affected members of the family. Three additional rare missense variants were found in 3/186 unrelated index cases. We detected higher levels of RRAD transcripts in subepicardium than in subendocardium in human heart, and in the right ventricle outflow tract compared to the other cardiac compartments in mice. The p.R211H variant was then subjected to electrophysiological and structural investigations in human cardiomyocytes derived from induced pluripotent stem cells (iPSC-CMs). Cardiomyocytes derived from induced pluripotent stem cells from two affected family members exhibited reduced action potential upstroke velocity, prolonged action potentials and increased incidence of early afterdepolarizations, with decreased Na+ peak current amplitude and increased Na+ persistent current amplitude, as well as abnormal distribution of actin and less focal adhesions, compared with intra-familial control iPSC-CMs Insertion of p.R211H-RRAD variant in control iPSCs by genome editing confirmed these results. In addition, iPSC-CMs from affected patients exhibited a decreased L-type Ca2+ current amplitude. Conclusion This study identified a potential new BrS-susceptibility gene, RRAD. Cardiomyocytes derived from induced pluripotent stem cells expressing RRAD variant recapitulated single-cell electrophysiological features of BrS, including altered Na+ current, as well as cytoskeleton disturbances.
- Published
- 2019
- Full Text
- View/download PDF
34. HIV-Tat induces a decrease in I Kr and I Ks via reduction in phosphatidylinositol-(4,5)-bisphosphate availability
- Author
-
Flavien Charpentier, Olfat A. Malak, Nadjet Belbachir, Mariam Jouni, C. Gauthier, Isabelle Baró, Kazem Zibara, Guillaume Lamirault, Zeina R Al Sayed, Bruno Beaumelle, Nathalie Gaborit, Zeineb Es-Salah-Lamoureux, Marine Gandon-Renard, Patricia Lemarchand, Gildas Loussouarn, unité de recherche de l'institut du thorax UMR1087 UMR6291 (ITX), Université de Nantes - UFR de Médecine et des Techniques Médicales (UFR MEDECINE), Université de Nantes (UN)-Université de Nantes (UN)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Centre d’Investigation Clinique de Nantes (CIC Nantes), Université de Nantes (UN)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre hospitalier universitaire de Nantes (CHU Nantes), Laboratory of Stem Cells [Lebanese, Beirut] (ER045-PRASE), Lebanese University [Beirut] (LU), Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier (UM)-Centre National de la Recherche Scientifique (CNRS), This work was funded by grants from the Fédération Française de Cardiologie, the Fondation Genavie, the Marie Curie European Actions (PIIF-GA-2012-331436 and PIOF-GA-2011-298280), the French Regional Council of Pays-de-la-Loire (Regional grant VACARME -www.vacarme-project.org) and the Agence Nationale de Recherche (ANR-15-CE14-0019-01). Dr Z. Es-Salah-Lamoureux was supported by grants from the Lefoulon Delalande Foundation, the Fondation pour la Recherche Médicale (SPF20111223614, FRM) and the Fondation Genavie. M. Jouni was awarded a scholarship from the Association of Scientific Orientation and Specialization (ASOS). Olfat Malak was laureate of the Line Pomaret-Delalande prize of the Fondation pour la Recherche Médicale (PLP20141031304, FRM). Zeina Reda Al Sayed was awarded by a Eiffel program from Campus France., Centre d’études d’Agents Pathogènes et Biotechologies pour la Santé (CPBS), Unité de recherche de l'institut du thorax (ITX-lab), Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université de Nantes - UFR de Médecine et des Techniques Médicales (UFR MEDECINE), Université de Nantes (UN)-Université de Nantes (UN), and Lemarchand, Patricia
- Subjects
0301 basic medicine ,Long QT syndrome ,Mutant ,hERG ,Biophysics ,[SDV.BC]Life Sciences [q-bio]/Cellular Biology ,010402 general chemistry ,01 natural sciences ,03 medical and health sciences ,Transactivation ,chemistry.chemical_compound ,[SDV.MHEP.CSC]Life Sciences [q-bio]/Human health and pathology/Cardiology and cardiovascular system ,medicine ,Induced pluripotent stem cell ,[SDV.BC] Life Sciences [q-bio]/Cellular Biology ,Molecular Biology ,ComputingMilieux_MISCELLANEOUS ,030304 developmental biology ,0303 health sciences ,biology ,Transfection ,medicine.disease ,Virology ,0104 chemical sciences ,3. Good health ,Cell biology ,030104 developmental biology ,Phosphatidylinositol 4,5-bisphosphate ,chemistry ,[SDV.MP.VIR]Life Sciences [q-bio]/Microbiology and Parasitology/Virology ,biology.protein ,Heterologous expression ,Cardiology and Cardiovascular Medicine - Abstract
International audience; Patients with HIV present with a higher prevalence of QT prolongation, of which molecularbases are still not clear. Among HIV proteins, Tat serves as a transactivator that stimulatesviral genes expression and is required for efficient HIV replication. Tat is actively secretedinto the blood by infected T-cells and affects organs such as the heart. Tat has been shown toalter cardiac repolarization in animal models but how this is mediated and whether this is alsothe case in human cells is unknown. In the present study, we show that Tat transfection inheterologous expression systems led to a decrease in hERG (underlying cardiac IKr) andhuman KCNE1-KCNQ1 (underlying cardiac IKs) currents and to an acceleration of theirdeactivation. This is consistent with a decrease in available phosphatidylinositol-(4,5)-bisphosphate (PIP2). A mutant Tat, unable to bind PIP2, did not reproduce the observedeffects. In addition, WT-Tat had no effect on a mutant KCNQ1 which is PIP2-insensitive,further confirming the hypothesis. Twenty four-hour incubation of human induced pluripotentstem cells-derived cardiomyocytes with Wild-type Tat reduced IKr and accelerated itsdeactivation. Concordantly, this Tat incubation led to a prolongation of the action potential(AP) duration. Events of AP alternans were also recorded in the presence of Tat, and wereexacerbated at a low pacing cycle length. Altogether, these data obtained on human K+channels both in heterologous expression systems and in human cardiomyocytes stronglysuggest that Tat sequesters PIP2, leading to a reduction of IKr and IKs, and provide a molecularmechanism for QT prolongation in HIV-infected patients.Key
- Published
- 2016
- Full Text
- View/download PDF
35. Arrhythmias precede cardiomyopathy and remodeling of Ca
- Author
-
Jérôme, Montnach, Franck F, Chizelle, Nadjet, Belbachir, Claire, Castro, Linwei, Li, Gildas, Loussouarn, Gilles, Toumaniantz, Agnès, Carcouët, Anne Julia, Meinzinger, Doron, Shmerling, Jean-Pierre, Benitah, Ana Maria, Gómez, Flavien, Charpentier, and Isabelle, Baró
- Subjects
Mice, Knockout ,Action Potentials ,Immunohistochemistry ,Propranolol ,Molecular Imaging ,NAV1.5 Voltage-Gated Sodium Channel ,Survival Rate ,Disease Models, Animal ,Electrocardiography ,Long QT Syndrome ,Mice ,Phenotype ,Echocardiography ,Heart Function Tests ,Disease Progression ,Animals ,Calcium ,Myocytes, Cardiac ,Cardiomyopathies ,Signal Transduction - Abstract
Deletion of QKP1507-1509 amino-acids in SCN5A gene product, the voltage-gated NaWe generated a knock-in mouse presenting the delQKP1510-1512 mutation (Scn5aThe Scn5a
- Published
- 2018
36. Mental stress test: a rapid, simple, and efficient test to unmask long QT syndrome
- Author
-
Vincent Probst, Julien Barc, François Huchet, Jean-Baptiste Gourraud, Hervé Le Marec, Béatrice Guyomarch, Richard Redon, Nathalie Gaborit, Pauline Etienne, Jean-Jacques Schott, F. Kyndt, Aurélie Thollet, Flavien Charpentier, unité de recherche de l'institut du thorax UMR1087 UMR6291 (ITX), Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université de Nantes - UFR de Médecine et des Techniques Médicales (UFR MEDECINE), and Université de Nantes (UN)-Université de Nantes (UN)
- Subjects
Adult ,Male ,medicine.medical_specialty ,congenital, hereditary, and neonatal diseases and abnormalities ,Adolescent ,Long QT syndrome ,[SDV]Life Sciences [q-bio] ,030204 cardiovascular system & hematology ,Qt duration ,QT interval ,03 medical and health sciences ,Electrocardiography ,Young Adult ,0302 clinical medicine ,Mutation Carrier ,Heart Rate ,Predictive Value of Tests ,Risk Factors ,Physiology (medical) ,Internal medicine ,Mental stress ,Heart rate ,medicine ,Humans ,KCNQ2 Potassium Channel ,In patient ,Genetic Predisposition to Disease ,030212 general & internal medicine ,Aged ,Aged, 80 and over ,Receiver operating characteristic ,business.industry ,Reproducibility of Results ,Mathematical Concepts ,Middle Aged ,medicine.disease ,3. Good health ,Long QT Syndrome ,Phenotype ,Case-Control Studies ,KCNQ1 Potassium Channel ,Mutation ,Ventricular Fibrillation ,Cardiology ,Female ,Cardiology and Cardiovascular Medicine ,business ,Stress, Psychological - Abstract
International audience; Aims: QT prolongation during mental stress test (MST) has been associated with familial idiopathic ventricular fibrillation. In long QT syndrome (LQTS), up to 30% of mutation carriers have normal QT duration. Our aim was to assess the QT response during MST, and its accuracy in the diagnosis of concealed LQTS. Methods and results: All patients who are carrier of a KCNQ1 or KCNH2 mutations without QT prolongation were enrolled. A control group was constituted of patients with negative exercise and epinephrine tests. Electrocardiogram were recorded at rest and at the maximum heart rate during MST and reviewed by two physicians. Among the 70 patients enrolled (median age 41±2.1 years, 46% male), 36 were mutation carrier for LQTS (20 KCNQ1 and 16 KCNH2), and 34 were controls. KCNQ1 and KCNH2 mutation carriers presented a longer QT interval at baseline [405(389; 416) and 421 (394; 434) ms, respectively] compared with the controls [361(338; 375)ms; P \textless 0.0001]. QT duration during MST varied by 9 (4; 18) ms in KCNQ1, 3 (-6; 16) ms in KCNH2, and by -22 (-29; -17) ms in controls (P \textless 0.0001). These QT variations were independent of heart rate (P \textless 0.3751). Receiver operating characteristic curve analysis identified a cut-off value of QT variation superior to -11 ms as best predictor of LQTS. It provided 97% sensitivity and 97% specificity of QT prolongation in the diagnosis of LQTS. Conclusion: We identified a paradoxical response of the QT interval during MST in LQTS. Easy to assess, MST may be efficient to unmask concealed LQTS in patients at risk of this pathology.
- Published
- 2018
- Full Text
- View/download PDF
37. C-terminal phosphorylation of NaV1.5 impairs FGF13-dependent regulation of channel inactivation
- Author
-
Gildas Loussouarn, Joan Heller Brown, Sophie Burel, Fabien C. Coyan, Jeanne M. Nerbonne, Flavien Charpentier, Lars S. Maier, Cheryl F. Lichti, Matthew R. Meyer, Maxime Lorenzini, Raymond R. Townsend, Céline Marionneau, unité de recherche de l'institut du thorax UMR1087 UMR6291 (ITX), Université de Nantes - UFR de Médecine et des Techniques Médicales (UFR MEDECINE), and Université de Nantes (UN)-Université de Nantes (UN)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
0301 basic medicine ,calmodulin ,Biochemistry & Molecular Biology ,Calmodulin ,[SDV]Life Sciences [q-bio] ,heart ,Nav1.5 ,Fibroblast growth factor ,Biochemistry ,Medical and Health Sciences ,Transgenic ,NAV1.5 Voltage-Gated Sodium Channel ,03 medical and health sciences ,Mice ,Ca2+/calmodulin-dependent protein kinase ,Animals ,Humans ,Phosphorylation ,Molecular Biology ,Ca2+/calmodulin-dependent protein kinase II (CaMKII) ,Heart Failure ,biology ,phosphorylation ,Sodium channel ,HEK 293 cells ,Phosphoproteomics ,phosphoproteomics ,Cell Biology ,channel inactivation ,Biological Sciences ,Molecular biology ,Fibroblast Growth Factors ,030104 developmental biology ,HEK293 Cells ,Amino Acid Substitution ,Mutation ,Chemical Sciences ,biology.protein ,FGF13 ,Ca2+/calmodulin-dependent protein kinase II ,calmodulin (CaM) ,Missense ,Calcium-Calmodulin-Dependent Protein Kinase Type 2 ,Ion Channel Gating ,sodium channel - Abstract
International audience; Voltage-gated Na(+) (NaV) channels are key regulators of myocardial excitability, and Ca(2+)/calmodulin-dependent protein kinase II (CaMKII)-dependent alterations in NaV1.5 channel inactivation are emerging as a critical determinant of arrhythmias in heart failure. However, the global native phosphorylation pattern of NaV1.5 subunits associated with these arrhythmogenic disorders and the associated channel regulatory defects remain unknown. Here, we undertook phosphoproteomic analyses to identify and quantify in situ the phosphorylation sites in the NaV1.5 proteins purified from adult WT and failing CaMKIIδc-overexpressing (CaMKIIδc-Tg) mouse ventricles. Of 19 native NaV1.5 phosphorylation sites identified, two C-terminal phosphoserines at positions 1938 and 1989 showed increased phosphorylation in the CaMKIIδc-Tg compared with the WT ventricles. We then tested the hypothesis that phosphorylation at these two sites impairs fibroblast growth factor 13 (FGF13)-dependent regulation of NaV1.5 channel inactivation. Whole-cell voltage-clamp analyses in HEK293 cells demonstrated that FGF13 increases NaV1.5 channel availability and decreases late Na(+) current, two effects that were abrogated with NaV1.5 mutants mimicking phosphorylation at both sites. Additional co-immunoprecipitation experiments revealed that FGF13 potentiates the binding of calmodulin to NaV1.5 and that phosphomimetic mutations at both sites decrease the interaction of FGF13 and, consequently, of calmodulin with NaV1.5. Together, we have identified two novel native phosphorylation sites in the C terminus of NaV1.5 that impair FGF13-dependent regulation of channel inactivation and may contribute to CaMKIIδc-dependent arrhythmogenic disorders in failing hearts.
- Published
- 2017
- Full Text
- View/download PDF
38. Transforming growth factor β receptor inhibition prevents ventricular fibrosis in a mouse model of progressive cardiac conduction disease
- Author
-
Cynthia Ore Cerpa, Christopher L.-H. Huang, Mickaël Derangeon, B. Jagu, Justine Patin, Jérôme Montnach, Isabelle Baró, Flavien Charpentier, Gilles Toumaniantz, Andrew A. Grace, William H. Colledge, Aurore Girardeau, unité de recherche de l'institut du thorax UMR1087 UMR6291 (ITX), Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université de Nantes - UFR de Médecine et des Techniques Médicales (UFR MEDECINE), Université de Nantes (UN)-Université de Nantes (UN), Université de Nantes - UFR de Médecine et des Techniques Médicales (UFR MEDECINE), and Université de Nantes (UN)-Université de Nantes (UN)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
Male ,0301 basic medicine ,Physiology ,[SDV]Life Sciences [q-bio] ,Connexin ,030204 cardiovascular system & hematology ,Nav1.5 ,Membrane Potentials ,NAV1.5 Voltage-Gated Sodium Channel ,0302 clinical medicine ,Heart Rate ,Transforming Growth Factor beta ,Fibrosis ,Receptor ,Mice, Knockout ,Voltage-Gated Sodium Channel Blockers ,Flecainide ,Ventricular Remodeling ,biology ,Age Factors ,Phenotype ,Benzamides ,cardiovascular system ,Female ,Cardiomyopathies ,Cardiology and Cardiovascular Medicine ,Anti-Arrhythmia Agents ,Signal Transduction ,Heterozygote ,medicine.medical_specialty ,Mice, 129 Strain ,Heart Ventricles ,Smad2/3 ,03 medical and health sciences ,Heart Conduction System ,Physiology (medical) ,Internal medicine ,Cardiac conduction ,medicine ,Animals ,Genetic Predisposition to Disease ,cardiovascular diseases ,business.industry ,CTGF ,Arrhythmias, Cardiac ,medicine.disease ,Disease Models, Animal ,Kinetics ,030104 developmental biology ,Endocrinology ,Connexin 43 ,biology.protein ,Pyrazoles ,Myocardial fibrosis ,GW788388 ,business ,Receptors, Transforming Growth Factor beta ,Transforming growth factor - Abstract
International audience; Aims: Loss-of-function mutations in SCN5A, the gene encoding NaV1.5 channel, have been associated with inherited progressive cardiac conduction disease (PCCD). We have proposed that Scn5a heterozygous knock-out (Scn5a+/-) mice, which are characterized by ventricular fibrotic remodelling with ageing, represent a model for PCCD. Our objectives were to identify the molecular pathway involved in fibrosis development and prevent its activation. Methods and results: Our study shows that myocardial interstitial fibrosis occurred in Scn5a+/- mice only after 45 weeks of age. Fibrosis was triggered by transforming growth factor β (TGF-β) pathway activation. Younger Scn5a+/- mice were characterized by a higher connexin 43 expression than wild-type (WT) mice. After the age of 45 weeks, connexin 43 expression decreased in both WT and Scn5a+/- mice, although the decrease was larger in Scn5a+/- mice. Chronic inhibition of cardiac sodium current with flecainide (50 mg/kg/day p.o) in WT mice from the age of 6 weeks to the age of 60 weeks did not lead to TGF-β pathway activation and fibrosis. Chronic inhibition of TGF-β receptors with GW788388 (5 mg/kg/day p.o.) in Scn5a+/- mice from the age of 45 weeks to the age of 60 weeks prevented the occurrence of fibrosis. However, current data could not detect reduction in QRS duration with GW788388. Conclusion: Myocardial fibrosis secondary to a loss of NaV1.5 is triggered by TGF-β signalling pathway. Those events are more likely secondary to the decreased NaV1.5 sarcolemmal expression rather than the decreased Na+ current per se. TGF-β receptor inhibition prevents age-dependent development of ventricular fibrosis in Scn5a+/- mouse.
- Published
- 2017
- Full Text
- View/download PDF
39. C-terminal phosphorylation of Na
- Author
-
Sophie, Burel, Fabien C, Coyan, Maxime, Lorenzini, Matthew R, Meyer, Cheryl F, Lichti, Joan H, Brown, Gildas, Loussouarn, Flavien, Charpentier, Jeanne M, Nerbonne, R Reid, Townsend, Lars S, Maier, and Céline, Marionneau
- Subjects
Heart Failure ,Mutation, Missense ,Mice, Transgenic ,Molecular Bases of Disease ,NAV1.5 Voltage-Gated Sodium Channel ,Fibroblast Growth Factors ,Mice ,HEK293 Cells ,Amino Acid Substitution ,Animals ,Humans ,Phosphorylation ,Calcium-Calmodulin-Dependent Protein Kinase Type 2 ,Ion Channel Gating - Abstract
Voltage-gated Na+ (NaV) channels are key regulators of myocardial excitability, and Ca2+/calmodulin-dependent protein kinase II (CaMKII)-dependent alterations in NaV1.5 channel inactivation are emerging as a critical determinant of arrhythmias in heart failure. However, the global native phosphorylation pattern of NaV1.5 subunits associated with these arrhythmogenic disorders and the associated channel regulatory defects remain unknown. Here, we undertook phosphoproteomic analyses to identify and quantify in situ the phosphorylation sites in the NaV1.5 proteins purified from adult WT and failing CaMKIIδc-overexpressing (CaMKIIδc-Tg) mouse ventricles. Of 19 native NaV1.5 phosphorylation sites identified, two C-terminal phosphoserines at positions 1938 and 1989 showed increased phosphorylation in the CaMKIIδc-Tg compared with the WT ventricles. We then tested the hypothesis that phosphorylation at these two sites impairs fibroblast growth factor 13 (FGF13)-dependent regulation of NaV1.5 channel inactivation. Whole-cell voltage-clamp analyses in HEK293 cells demonstrated that FGF13 increases NaV1.5 channel availability and decreases late Na+ current, two effects that were abrogated with NaV1.5 mutants mimicking phosphorylation at both sites. Additional co-immunoprecipitation experiments revealed that FGF13 potentiates the binding of calmodulin to NaV1.5 and that phosphomimetic mutations at both sites decrease the interaction of FGF13 and, consequently, of calmodulin with NaV1.5. Together, we have identified two novel native phosphorylation sites in the C terminus of NaV1.5 that impair FGF13-dependent regulation of channel inactivation and may contribute to CaMKIIδc-dependent arrhythmogenic disorders in failing hearts.
- Published
- 2017
40. The Sodium–Glucose Cotransporter 2 Inhibitor Dapagliflozin Prevents Cardiomyopathy in a Diabetic Lipodystrophic Mouse Model
- Author
-
Benoît Jagu, Jocelyne Magré, L. Dollet, Bertrand Cariou, Xavier Prieur, Xavier Marechal, David Montaigne, Michael Joubert, Cédric Le May, Audrey Ayer, Gilles Toumaniantz, Flavien Charpentier, Angela Tesse, Alain Manrique, Bart Staels, unité de recherche de l'institut du thorax UMR1087 UMR6291 (ITX), Université de Nantes - UFR de Médecine et des Techniques Médicales (UFR MEDECINE), Université de Nantes (UN)-Université de Nantes (UN)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université de Nantes - UFR de Médecine et des Techniques Médicales (UFR MEDECINE), and Université de Nantes (UN)-Université de Nantes (UN)
- Subjects
Blood Glucose ,Lipodystrophy ,Diabetic Cardiomyopathies ,[SDV]Life Sciences [q-bio] ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,Glucose uptake ,Left ,Cardiomyopathy ,030204 cardiovascular system & hematology ,Seipin ,chemistry.chemical_compound ,Mice ,Ventricular Dysfunction, Left ,0302 clinical medicine ,Glucosides ,Diabetic cardiomyopathy ,GTP-Binding Protein gamma Subunits ,Ventricular Dysfunction ,Medicine ,Ventricular Function ,Dapagliflozin ,ComputingMilieux_MISCELLANEOUS ,Mice, Knockout ,Hypertrophic cardiomyopathy ,Heart ,[SDV.MHEP.EM]Life Sciences [q-bio]/Human health and pathology/Endocrinology and metabolism ,Heterotrimeric GTP-Binding Proteins ,Magnetic Resonance Imaging ,3. Good health ,Echocardiography ,Type 2 ,medicine.drug ,medicine.medical_specialty ,Knockout ,030209 endocrinology & metabolism ,03 medical and health sciences ,Sodium-Glucose Transporter 2 ,[SDV.MHEP.CSC]Life Sciences [q-bio]/Human health and pathology/Cardiology and cardiovascular system ,Fluorodeoxyglucose F18 ,Internal medicine ,Diabetes Mellitus ,Internal Medicine ,[SDV.MHEP.PHY]Life Sciences [q-bio]/Human health and pathology/Tissues and Organs [q-bio.TO] ,Animals ,Hypoglycemic Agents ,Benzhydryl Compounds ,Sodium-Glucose Transporter 2 Inhibitors ,Pioglitazone ,Animal ,business.industry ,Insulin ,Myocardium ,[SDV.MHEP.HEG]Life Sciences [q-bio]/Human health and pathology/Hépatology and Gastroenterology ,Cardiomyopathy, Hypertrophic ,medicine.disease ,Disease Models, Animal ,Endocrinology ,chemistry ,Diabetes Mellitus, Type 2 ,Hypertrophic ,Hyperglycemia ,Positron-Emission Tomography ,Disease Models ,Thiazolidinediones ,Radiopharmaceuticals ,business - Abstract
International audience; Type 2 diabetes mellitus (T2DM) is a well-recognized independent risk factor for heart failure. T2DM is associated with altered cardiac energy metabolism, leading to ectopic lipid accumulation and glucose overload, the exact contribution of these two parameters remaining unclear. To provide new insight into the mechanism driving the development of diabetic cardiomyopathy, we studied a unique model of T2DM: lipodystrophic Bscl2(-/-) (seipin knockout [SKO]) mice. Echocardiography and cardiac magnetic resonance imaging revealed hypertrophic cardiomyopathy with left ventricular dysfunction in SKO mice, and these two abnormalities were strongly correlated with hyperglycemia. Surprisingly, neither intramyocardial lipid accumulation nor lipotoxic hallmarks were detected in SKO mice. [(18)F]Fludeoxyglucose positron emission tomography showed increased myocardial glucose uptake. Consistently, the O-GlcNAcylated protein levels were markedly increased in an SKO heart, suggesting a glucose overload. To test this hypothesis, we treated SKO mice with the hypoglycemic sodium-glucose cotransporter 2 (SGLT2) inhibitor dapagliflozin and the insulin sensitizer pioglitazone. Both treatments reduced the O-GlcNAcylated protein levels in SKO mice, and dapagliflozin successfully prevented the development of hypertrophic cardiomyopathy. Our data demonstrate that glucotoxicity by itself can trigger cardiac dysfunction and that a glucose-lowering agent can correct it. This result will contribute to better understanding of the potential cardiovascular benefits of SGLT2 inhibitors.
- Published
- 2017
- Full Text
- View/download PDF
41. Familial Catecholamine-Induced QT Prolongation in Unexplained Sudden Cardiac Death
- Author
-
Richard Redon, Jean-Baptiste Gourraud, Aurélie Thollet, Flavien Charpentier, François Huchet, Jean-Jacques Schott, Hervé Le Marec, Julien Barc, Florence Kyndt, Vincent Probst, unité de recherche de l'institut du thorax UMR1087 UMR6291 (ITX), Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université de Nantes - UFR de Médecine et des Techniques Médicales (UFR MEDECINE), and Université de Nantes (UN)-Université de Nantes (UN)
- Subjects
Male ,0301 basic medicine ,medicine.medical_specialty ,Adolescent ,Catecholamines blood ,[SDV]Life Sciences [q-bio] ,030204 cardiovascular system & hematology ,030105 genetics & heredity ,QT interval ,Sudden cardiac death ,Electrocardiography ,03 medical and health sciences ,Catecholamines ,0302 clinical medicine ,Internal medicine ,Mental stress ,medicine ,Humans ,medicine.diagnostic_test ,business.industry ,medicine.disease ,3. Good health ,Long QT Syndrome ,Death, Sudden, Cardiac ,Catecholamine ,Cardiology ,Female ,Cardiology and Cardiovascular Medicine ,business ,Stress, Psychological ,Cardiac deaths ,medicine.drug - Abstract
International audience; Despite major progress in molecular and phenotypic characterization of primary electrical disorders, many (aborted) sudden cardiac deaths (SCDs) occur in young victims without identifiable abnormalities (1). This study aimed to describe the use of mental stress test (MST) to identify catecholamine-induced QT prolongation (CIQTP) in SCD familial screening. MST was performed in the screening of 65 consec-utives families affected by unexplained SCD referred to the National Referral Centre for Inherited Cardiac Arrhythmias of Nantes. Conventional screening included echocardiography, exercise test, and epinephrine and ajmaline tests (1). MST was performed following a standardized protocol mostly based on mental arithmetic in stressful conditions (2). Two physicians blinded to patient clinical and genetic status reviewed all the electrocardiograms. Patients were considered affected for CIQTP when presenting prolonged QTc interval >480 ms or if >30 ms QT prolongation with the QTc interval >460 ms (1) during the different tests. We fortuitously unmasked a significant prolonga-tion of QTc duration in a young woman when mentioning the unexplained SCD of her sister at summer 2013. According to previous description of stress-induced repolarization abnormalities (2,3), we additionally performed MST during familial screening in 65 families with a familial history of SCD in young patients. Conventional screening identified 7 Brugada syndrome, 6 long QT syndrome (LQTS), and 3 familial cardiomyopathies. MST unmasked CIQTP in 4 families (mean age of SCD 18 years) (Figure 1A). In these families, 31 of the 57 tested family members were affected (QTc during MST 501 AE 36 ms vs. 430 AE 19 ms) with a flattening or double-hump T-wave aspect in 13 of 31 versus 1 of 26 in unaffected patients (p < 0.001) (Figure 1B). In 9 patients (including 2 with previous reported syncope) MST was the only positive test. Eight of these patients did not receive an epinephrine test because of contraindication (
- Published
- 2017
- Full Text
- View/download PDF
42. Courant sodique, dépolarisation et conduction cardiaque
- Author
-
Isabelle Baró, Mohamed Yassine Amarouch, and Flavien Charpentier
- Subjects
Chemistry ,Cardiology and Cardiovascular Medicine - Published
- 2013
- Full Text
- View/download PDF
43. Dysfunction of the Voltage‐Gated K + Channel β2 Subunit in a Familial Case of Brugada Syndrome
- Author
-
Gildas Loussouarn, Vincent Probst, Thomas O'Hara, Christian Dina, Carol Scott, Eléonore Moreau, Solena Le Scouarnec, Stéphanie Chatel, Stéphanie Bonnaud, Hervé Le Marec, Sophie Burel, Flavien Charpentier, Jade Violleau, Isabelle Baró, Estelle Baron, Pierre Lindenbaum, Vincent Portero, Jean-Jacques Schott, Floriane Simonet, Céline Marionneau, Philippe Mabo, Zeineb Es‐Salah‐Lamoureux, Jean-Baptiste Gourraud, Richard Redon, Jonchère, Laurent, Unité de recherche de l'institut du thorax (ITX-lab), Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université de Nantes - UFR de Médecine et des Techniques Médicales (UFR MEDECINE), Université de Nantes (UN)-Université de Nantes (UN), The Wellcome Trust Sanger Institute [Cambridge], CIC-IT Rennes, Hôpital Pontchaillou-Institut National de la Santé et de la Recherche Médicale (INSERM), Laboratoire Traitement du Signal et de l'Image (LTSI), Université de Rennes (UR)-Institut National de la Santé et de la Recherche Médicale (INSERM), Service de cardiologie et maladies vasculaires [Rennes] = Cardiac, Thoracic, and Vascular Surgery [Rennes], CHU Pontchaillou [Rennes], Leducq Foundation [CVD-05], Fondation pour la Recherche Medicale [DEQ20140329545], Inserm, French Regional Council of Pays-de-la-Loire, Fondation Lefoulon-Delalande, Fondation pour la Recherche Medicale, Fondation Genavie, Wellcome Trust [WT098051], European Commission [NavEx-256397], unité de recherche de l'institut du thorax UMR1087 UMR6291 (ITX), Université de Nantes - UFR de Médecine et des Techniques Médicales (UFR MEDECINE), Université de Nantes (UN)-Université de Nantes (UN)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Université de Rennes 1 (UR1), and Université de Rennes (UNIV-RENNES)-Université de Rennes (UNIV-RENNES)-Institut National de la Santé et de la Recherche Médicale (INSERM)
- Subjects
Male ,0301 basic medicine ,Arrhythmias ,030204 cardiovascular system & hematology ,whole exome sequencing ,NAV1.5 Voltage-Gated Sodium Channel ,Electrocardiography ,0302 clinical medicine ,Genotype ,Missense mutation ,Arrhythmia and Electrophysiology ,genetics ,Exome sequencing ,risk ,Original Research ,Brugada syndrome ,Genetics ,maps ,clinical electrophysiology ,potassium ion channels ,potassium channels ,[SDV.MHEP.CSC] Life Sciences [q-bio]/Human health and pathology/Cardiology and cardiovascular system ,Pedigree ,3. Good health ,Electrophysiology ,modulation ,Shal Potassium Channels ,Potassium Channels, Voltage-Gated ,Gain of Function Mutation ,Cardiology ,Female ,abnormalities ,Cardiology and Cardiovascular Medicine ,congenital, hereditary, and neonatal diseases and abnormalities ,medicine.medical_specialty ,gene prioritization ,j-wave syndromes ,[SDV.GEN.GH] Life Sciences [q-bio]/Genetics/Human genetics ,Polymorphism, Single Nucleotide ,Sudden death ,sudden cardiac death ,cardiac arrhythmia ,03 medical and health sciences ,[SDV.MHEP.CSC]Life Sciences [q-bio]/Human health and pathology/Cardiology and cardiovascular system ,Genetic linkage ,Internal medicine ,Exome Sequencing ,medicine ,Humans ,Genetic Predisposition to Disease ,cardiovascular diseases ,Gene ,disease ,business.industry ,KCNAB2/Kvβ2 ,mutations ,medicine.disease ,030104 developmental biology ,[SDV.GEN.GH]Life Sciences [q-bio]/Genetics/Human genetics ,Shaker Superfamily of Potassium Channels ,business - Abstract
Background The Brugada syndrome is an inherited cardiac arrhythmia associated with high risk of sudden death. Although 20% of patients with Brugada syndrome carry mutations in SCN 5A , the molecular mechanisms underlying this condition are still largely unknown. Methods and Results We combined whole‐exome sequencing and linkage analysis to identify the genetic variant likely causing Brugada syndrome in a pedigree for which SCN 5A mutations had been excluded. This approach identified 6 genetic variants cosegregating with the Brugada electrocardiographic pattern within the pedigree. In silico gene prioritization pointed to 1 variant residing in KCNAB 2 , which encodes the voltage‐gated K + channel β2‐subunit (Kvβ2‐R12Q). Kvβ2 is widely expressed in the human heart and has been shown to interact with the fast transient outward K + channel subunit Kv4.3, increasing its current density. By targeted sequencing of the KCNAB 2 gene in 167 unrelated patients with Brugada syndrome, we found 2 additional rare missense variants (L13F and V114I). We then investigated the physiological effects of the 3 KCNAB 2 variants by using cellular electrophysiology and biochemistry. Patch‐clamp experiments performed in COS ‐7 cells expressing both Kv4.3 and Kvβ2 revealed a significant increase in the current density in presence of the R12Q and L13F Kvβ2 mutants. Although biotinylation assays showed no differences in the expression of Kv4.3, the total and submembrane expression of Kvβ2‐R12Q were significantly increased in comparison with wild‐type Kvβ2. Conclusions Altogether, our results indicate that Kvβ2 dysfunction can contribute to the Brugada electrocardiographic pattern.
- Published
- 2016
- Full Text
- View/download PDF
44. 0062 : C-terminal phosphorylation of Nav1.5 channels impairs FGF13-dependent inactivation: potential impact in heart failure
- Author
-
Flavien Charpentier, Lars S. Maier, Cheryl Litchi, Céline Marionneau, Sophie Burel, R. Reid Townsend, Matthew R. Meyer, Maxime Lorenzini, Joan Heller Brown, Fabien C. Coyan, and Jeanne M. Nerbonne
- Subjects
medicine.medical_specialty ,biology ,Calmodulin ,business.industry ,Protein subunit ,HEK 293 cells ,Depolarization ,Nav1.5 ,Cell biology ,Endocrinology ,Internal medicine ,Ca2+/calmodulin-dependent protein kinase ,medicine ,biology.protein ,Phosphorylation ,Binding site ,business ,Cardiology and Cardiovascular Medicine - Abstract
Aims Voltage-gated Na+ (NaV) channels are key regulators of myocardial excitability, determining depolarization, duration, waveform and refractoriness of action potentials. In heart failure, action potentials are prolonged, and evidence suggests that this arrhythmogenic abnormality is associated with Ca2+/ Calmodulin-dependent protein Kinase II (CaMKII) and altered phosphorylation of the main ventricular NaV channel subunit, NaV1.5. The global native phosphorylation patterns of NaV1.5 channels in failing, compared with nonfailing, ventricles, and the associated channel regulatory defects, however, remain unknown. Methods and results Phosphoproteomic analyses were undertaken to identify and quantify in situ the phosphorylation sites on the NaV1.5 proteins purified from adult wild-type (WT) and failing CaMKIIdc-overexpressing (CaMKIIdc-Tg) mouse ventricles. Of the nineteen native NaV1.5 phosphorylation sites identified, two C-terminal phosphoserines at positions 1938 and 1989 showed increased phosphorylation in the CaMKIIdc-Tg, compared with the WT, ventricles. Because these two phosphoserines are located in close proximity to the binding site for Fibroblast Growth Factor 13 (FGF13), we tested the hypothesis that phosphorylation at these sites impairs the FGF13- mediated regulation of channel inactivation. Whole cell voltage-clamp analyses demonstrated that FGF13 increases NaV1.5 channel availability, accelerates recovery from inactivation and decreases the late Na+ current in HEK293 cells, three effects that were abrogated with NaV1.5 mutants mimicking phosphorylation at both sites. Further co-immunoprecipitation experiments revealed that phosphorylation at both sites decreases the interaction of FGF13 and of calmodulin with NaV1.5. Conclusions These analyses provide two novel native phosphorylation sites in the C-terminus of NaV1.5 which show increased phosphorylation in failing ventricles and impair FGF13-dependent inactivation. The author hereby declares no conflict of interest
- Published
- 2016
- Full Text
- View/download PDF
45. G protein-gated IKACh channels as therapeutic targets for treatment of sick sinus syndrome and heart block
- Author
-
Isabelle Bidaud, Pietro Mesirca, Kevin Wickman, Stéphane Evain, François Briec, Joerg Striessnig, Angelo G. Torrente, Laurine Marger, Anne Laure Leoni, Antony Chung You Chong, Flavien Charpentier, Matteo E. Mangoni, Khai Le Quang, Matthias Baudot, Joël Nargeot, Institut de Génomique Fonctionnelle (IGF), Université de Montpellier (UM)-Université Montpellier 1 (UM1)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Montpellier 2 - Sciences et Techniques (UM2)-Centre National de la Recherche Scientifique (CNRS), unité de recherche de l'institut du thorax UMR1087 UMR6291 (ITX), Université de Nantes - UFR de Médecine et des Techniques Médicales (UFR MEDECINE), Université de Nantes (UN)-Université de Nantes (UN)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Universität Innsbruck [Innsbruck], Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université de Nantes - UFR de Médecine et des Techniques Médicales (UFR MEDECINE), and Université de Nantes (UN)-Université de Nantes (UN)
- Subjects
0301 basic medicine ,Bradycardia ,medicine.medical_specialty ,Calcium Channels, L-Type ,Heart block ,Knockout ,[SDV]Life Sciences [q-bio] ,030204 cardiovascular system & hematology ,Sick sinus syndrome ,03 medical and health sciences ,Mice ,0302 clinical medicine ,GTP-Binding Proteins ,Internal medicine ,Heart rate ,medicine ,Animals ,humans ,Electronic pacemaker ,Mice, Knockout ,Sick Sinus Syndrome ,heart rate regulation ,Multidisciplinary ,Sinoatrial node ,business.industry ,GIRK4 ,Diastolic depolarization ,medicine.disease ,L-Type ,3. Good health ,SSS ,030104 developmental biology ,medicine.anatomical_structure ,Heart Block ,PNAS Plus ,Cardiology ,Calcium Channels ,medicine.symptom ,business ,Ion Channel Gating ,Cav1.3 - Abstract
International audience; Dysfunction of pacemaker activity in the sinoatrial node (SAN) underlies "sick sinus" syndrome (SSS), a common clinical condition characterized by abnormally low heart rate (bradycardia). If untreated, SSS carries potentially life-threatening symptoms, such as syncope and end-stage organ hypoperfusion. The only currently available therapy for SSS consists of electronic pacemaker implantation. Mice lacking L-type Cav1.3 Ca(2+) channels (Cav1.3(-/-)) recapitulate several symptoms of SSS in humans, including bradycardia and atrioventricular (AV) dysfunction (heart block). Here, we tested whether genetic ablation or pharmacological inhibition of the muscarinic-gated K(+) channel (IKACh) could rescue SSS and heart block in Cav1.3(-/-) mice. We found that genetic inactivation of IKACh abolished SSS symptoms in Cav1.3(-/-) mice without reducing the relative degree of heart rate regulation. Rescuing of SAN and AV dysfunction could be obtained also by pharmacological inhibition of IKACh either in Cav1.3(-/-) mice or following selective inhibition of Cav1.3-mediated L-type Ca(2+) (ICa,L) current in vivo. Ablation of IKACh prevented dysfunction of SAN pacemaker activity by allowing net inward current to flow during the diastolic depolarization phase under cholinergic activation. Our data suggest that patients affected by SSS and heart block may benefit from IKACh suppression achieved by gene therapy or selective pharmacological inhibition.
- Published
- 2016
- Full Text
- View/download PDF
46. Multifocal Ectopic Purkinje-Related Premature Contractions
- Author
-
Olivier Barthez, Philippe Charron, Samuel Saal, Jean-Eric Wolf, Rodolphe Turpault, Julien Barc, Gildas Loussouarn, Isabelle Baró, Florence Kyndt, Vincent Probst, Géraldine Bertaux, Mohamed Yassine Amarouch, Estelle Baron, Christel Thauvin-Robinet, Gabriel Laurent, Flavien Charpentier, Delphine M. Béziau, Laurence Faivre, Jean Mérot, Yves Coudière, Véronique Fressart, Christian Dina, Alice Maltret, Elisabeth Villain, Roos F. Marsman, Arthur A.M. Wilde, and Jean-Jacques Schott
- Subjects
0303 health sciences ,medicine.medical_specialty ,Purkinje fibers ,business.industry ,Sodium channel ,Cardiomyopathy ,Dilated cardiomyopathy ,030204 cardiovascular system & hematology ,medicine.disease ,Ventricular tachycardia ,Sudden death ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,Internal medicine ,cardiovascular system ,medicine ,Cardiology ,Repolarization ,cardiovascular diseases ,Cardiology and Cardiovascular Medicine ,business ,030304 developmental biology ,Cardiac channelopathy - Abstract
OBJECTIVES: The aim of this study was to describe a new familial cardiac phenotype and to elucidate the electrophysiological mechanism responsible for the disease. BACKGROUND: Mutations in several genes encoding ion channels, especially SCN5A, have emerged as the basis for a variety of inherited cardiac arrhythmias. METHODS: Three unrelated families comprising 21 individuals affected by multifocal ectopic Purkinje-related premature contractions (MEPPC) characterized by narrow junctional and rare sinus beats competing with numerous premature ventricular contractions with right and/or left bundle branch block patterns were identified. RESULTS: Dilated cardiomyopathy was identified in 6 patients, atrial arrhythmias were detected in 9 patients, and sudden death was reported in 5 individuals. Invasive electrophysiological studies demonstrated that premature ventricular complexes originated from the Purkinje tissue. Hydroquinidine treatment dramatically decreased the number of premature ventricular complexes. It normalized the contractile function in 2 patients. All the affected subjects carried the c.665G>A transition in the SCN5A gene. Patch-clamp studies of resulting p.Arg222Gln (R222Q) Nav1.5 revealed a net gain of function of the sodium channel, leading, in silico, to incomplete repolarization in Purkinje cells responsible for premature ventricular action potentials. In vitro and in silico studies recapitulated the normalization of the ventricular action potentials in the presence of quinidine. CONCLUSIONS: A new SCN5A-related cardiac syndrome, MEPPC, was identified. The SCN5A mutation leads to a gain of function of the sodium channel responsible for hyperexcitability of the fascicular-Purkinje system. The MEPPC syndrome is responsive to hydroquinidine.
- Published
- 2012
- Full Text
- View/download PDF
47. Early ion-channel remodeling and arrhythmias precede hypertrophy in a mouse model of complete atrioventricular block
- Author
-
Anne Bourge, Khai Le Quang, Olivier Bignolais, Aziza El Harchi, Julien Piron, Patrice Naud, Anne-Laure Leoni, Flavien Charpentier, François Briec, and Sophie Demolombe
- Subjects
Male ,Tachycardia ,medicine.medical_specialty ,Time Factors ,Heart Ventricles ,Action Potentials ,Down-Regulation ,Gene Expression ,Biology ,Real-Time Polymerase Chain Reaction ,Ventricular tachycardia ,Sudden death ,Ion Channels ,Muscle hypertrophy ,Electrocardiography ,Mice ,Ventricular hypertrophy ,Internal medicine ,medicine ,Animals ,Atrioventricular Block ,Molecular Biology ,medicine.diagnostic_test ,Gene Expression Profiling ,Myocardium ,Hemodynamics ,Arrhythmias, Cardiac ,Organ Size ,medicine.disease ,Up-Regulation ,Disease Models, Animal ,Protein Subunits ,Tachycardia, Ventricular ,cardiovascular system ,Cardiology ,Ventricular pressure ,Hypertrophy, Left Ventricular ,medicine.symptom ,Cardiology and Cardiovascular Medicine ,Atrioventricular block - Abstract
Complete atrioventricular block (CAVB) and related ventricular bradycardia are known to induce ventricular hypertrophy and arrhythmias. Different animal models of CAVB have been established with the most common being the dog model. Related studies were mainly focused on the consequences on the main repolarizing currents in these species, i.e. IKr and IKs, with a limited time point kinetics post-AVB. In order to explore at a genomic scale the electrical remodeling induced by AVB and its chronology, we have developed a novel model of CAVB in the mouse using a radiofrequency-mediated ablation procedure. We investigated transcriptional changes in ion channels and contractile proteins in the left ventricles as a function of time (12h, 1, 2 and 5 days after CAVB), using high-throughput real-time RT-PCR. ECG in conscious and anesthetized mice, left ventricular pressure recordings and patch-clamp were used for characterization of this new mouse model. As expected, CAVB was associated with a lengthening of the QT interval. Moreover, polymorphic ventricular tachycardia was recorded in 6/9 freely-moving mice during the first 24h post-ablation. Remarkably, myocardial hypertrophy was only evident 48 h post-ablation and was associated with increased heart weight and altered expression of contractile proteins. During the first 24 hours post-CAVB, genes encoding ion channel subunits were either up-regulated (such as Nav1.5, +74%) or down-regulated (Kv4.2, -43%; KChIP2, -47%; Navβ1, -31%; Cx43, -29%). Consistent with the transient alteration of Kv4.2 expression, I(to) was reduced at day 1, but restored at day 5. In conclusion, CAVB induces two waves of molecular remodeling: an early one (≤24 h) leading to arrhythmias, a later one related to hypertrophy. These results provide new molecular basis for ventricular tachycardia induced by AV block.
- Published
- 2011
- Full Text
- View/download PDF
48. Role of T-type calcium channel subunits in post-myocardial infarction remodelling probed with genetically engineered mice
- Author
-
Stanley Nattel, Philippe Comtois, Marc-Antoine Gillis, Danshi Li, Francine Duval, Khai Le Quang, Flavien Charpentier, Patrice Naud, Jean-Claude Tardif, Xiao-Yan Qi, Paul Levesque, Dobromir Dobrev, and Yanfen Shi
- Subjects
Male ,Cardiac function curve ,medicine.medical_specialty ,Mice, 129 Strain ,Time Factors ,Physiology ,Myocardial Infarction ,Stimulation ,Real-Time Polymerase Chain Reaction ,Ventricular Function, Left ,Calcium Channels, T-Type ,Electrocardiography ,Mice ,Heart Rate ,Physiology (medical) ,Internal medicine ,Heart rate ,medicine ,Animals ,RNA, Messenger ,Myocardial infarction ,Ventricular remodeling ,Ultrasonography ,Mice, Knockout ,Ejection fraction ,Ventricular Remodeling ,Voltage-dependent calcium channel ,Reverse Transcriptase Polymerase Chain Reaction ,Chemistry ,Myocardium ,Cardiac Pacing, Artificial ,Hemodynamics ,Stroke Volume ,medicine.disease ,Myocardial Contraction ,Mice, Inbred C57BL ,Disease Models, Animal ,Endocrinology ,Tachycardia, Ventricular ,Myocardial infarction complications ,Cardiology and Cardiovascular Medicine - Abstract
Aims Previous studies suggested that T-type Ca2+-current (ICaT)-blockers improve cardiac remodelling, but all available ICaT-blockers have non-specific actions on other currents and/or functions. To clarify the role of ICaT in cardiac remodelling, we studied mice with either of the principal cardiac ICaT-subunits (Cav3.1 or Cav3.2) knocked out. Methods and results Adult male Cav3.1- or Cav3.2-knockout (Cav3.1−/−, Cav3.2−/−) mice and respective wild-type (WT) littermate controls were subjected to left anterior descending coronary artery ligation to create myocardial infarction (MI). Echocardiography and programmed electrical stimulation were performed at baseline and 4 weeks post-MI. At baseline, Cav3.1−/− mice had slowed heart rates and longer PR intervals vs. WT, but no other electrophysiological and no haemodynamic differences. Cav3.2−/− showed no differences vs. WT. Contractile indices (left ventricular fractional shortening and ejection fraction) decreased more post-MI in Cav3.1−/− mice than in Cav3.1+/+ (e.g. by 34 and 29% for WT; 50 and 45% for Cav3.1−/−, respectively; P < 0.05 for each). Cav3.1−/− mice had increased ventricular tachycardia (VT) inducibility post-MI (9 of 11, 82%) vs. WT (3 of 10, 30%; P < 0.05). Cav3.2−/− mice were not different in cardiac function or VT inducibility vs. WT. Quantitative polymerase chain reaction showed that Cav3.1 is the major ICaT-subunit and that no compensatory Cav3.2 up-regulation occurs in Cav3.1−/− mice. Cav3.1−/− and Cav3.2−/− mice had no mRNA expression for the knocked-out gene, at baseline or post-MI. Conclusion Our findings suggest that, contrary to suggestions from previous studies with (imperfectly selective) pharmacological agents having T-type Ca2+-channel-blocking actions, elimination of Cav3.1 expression leads to impaired cardiac function and enhanced arrhythmia vulnerability post-MI, whereas Cav3.2 elimination has no effect.
- Published
- 2011
- Full Text
- View/download PDF
49. KCNE1-KCNQ1 osmoregulation by interaction of phosphatidylinositol-4,5-bisphosphate with Mg2+and polyamines
- Author
-
Nicolas Rodriguez, Isabelle Baró, Julien Piron, Flavien Charpentier, Frank S. Choveau, Jean Mérot, Gildas Loussouarn, and Mohammed Yassine Amarouch
- Subjects
0303 health sciences ,Osmotic concentration ,Physiology ,Biology ,Fusion protein ,Cell biology ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Membrane ,Phosphatidylinositol 4,5-bisphosphate ,chemistry ,Downregulation and upregulation ,Osmoregulation ,Extracellular ,lipids (amino acids, peptides, and proteins) ,030217 neurology & neurosurgery ,Intracellular ,030304 developmental biology - Abstract
KCNQ1 osmosensitivity is of physiological and pathophysiological relevance in epithelial and cardiac cells, but the mechanism involved remains elusive. In COS-7 cells expressing the KCNE1–KCNQ1 fusion protein, extracellular hypoosmolarity and hyperosmolarity modify the channel biophysical parameters. These changes are consistent with hypoosmolarity increasing the level of membrane phosphatidylinositol-4,5-bisphosphate (PIP2), which in turn upregulates KCNE1–KCNQ1 channels. We showed that increasing PIP2 levels with a water-soluble PIP2 analogue prevented channel upregulation in hypoosmotic condition, suggesting a variation of the channel–PIP2 interaction during channel osmoregulation. Furthermore, we showed that polyamines and Mg2+, already known to tonically inhibit KCNQ channels by screening PIP2 negative charges, are involved in the osmoregulatory process. Indeed, intracellular Mg2+ removal and polyamines chelation inhibited the channel osmoregulation. Thus, the dilution of those cations during cell swelling might decrease channel inhibition and explain the channel upregulation by hypoosmolarity. To support this idea, we quantified the role of Mg2+ in the osmodependent channel activity. Direct measurement of intracellular [Mg2+] variations during osmotic changes and characterization of the channel Mg2+ sensitivity showed that Mg2+ participates significantly to the osmoregulation. Using intracellular solutions that mimic the variation of Mg2+ and polyamines, we were able to recapitulate the current amplitude variations in response to extracellular osmolarity changes. Altogether, these results support the idea of a modulation of the channel–PIP2 interactions by Mg2+ and polyamines during cell volume changes. It is likely that this mechanism applies to other channels that are sensitive to both osmolarity and PIP2.
- Published
- 2010
- Full Text
- View/download PDF
50. P128Recovery of Scn5a-deficient mice cardiac conduction using AAVs
- Author
-
L Fosse, Michel Clergue, Alain Coulombe, Nathalie Mougenot, N Doisne, Flavien Charpentier, M Grauso-Culetto, Pascale Guicheney, and Nathalie Neyroud
- Subjects
medicine.medical_specialty ,Endocrinology ,Physiology ,business.industry ,Physiology (medical) ,Internal medicine ,Cardiac conduction ,medicine ,Deficient mouse ,Cardiology and Cardiovascular Medicine ,business - Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.