Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Estelle Baron is active.

Publication


Featured researches published by Estelle Baron.


Human Molecular Genetics | 2015

Testing the burden of rare variation in arrhythmia-susceptibility genes provides new insights into molecular diagnosis for Brugada syndrome

Solena Le Scouarnec; Matilde Karakachoff; Jean-Baptiste Gourraud; Pierre Lindenbaum; Stéphanie Bonnaud; Vincent Portero; Laetitia Duboscq-Bidot; Xavier Daumy; Floriane Simonet; Raluca Teusan; Estelle Baron; Jade Violleau; Elodie Persyn; Lise Bellanger; Julien Barc; Stéphanie Chatel; Raphaël P. Martins; Philippe Mabo; Frederic Sacher; Michel Haïssaguerre; Florence Kyndt; Sébastien Schmitt; Stéphane Bézieau; Hervé Le Marec; Christian Dina; Jean-Jacques Schott; Vincent Probst; Richard Redon

The Brugada syndrome (BrS) is a rare heritable cardiac arrhythmia disorder associated with ventricular fibrillation and sudden cardiac death. Mutations in the SCN5A gene have been causally related to BrS in 20-30% of cases. Twenty other genes have been described as involved in BrS, but their overall contribution to disease prevalence is still unclear. This study aims to estimate the burden of rare coding variation in arrhythmia-susceptibility genes among a large group of patients with BrS. We have developed a custom kit to capture and sequence the coding regions of 45 previously reported arrhythmia-susceptibility genes and applied this kit to 167 index cases presenting with a Brugada pattern on the electrocardiogram as well as 167 individuals aged over 65-year old and showing no history of cardiac arrhythmia. By applying burden tests, a significant enrichment in rare coding variation (with a minor allele frequency below 0.1%) was observed only for SCN5A, with rare coding variants carried by 20.4% of cases with BrS versus 2.4% of control individuals (P = 1.4 × 10(-7)). No significant enrichment was observed for any other arrhythmia-susceptibility gene, including SCN10A and CACNA1C. These results indicate that, except for SCN5A, rare coding variation in previously reported arrhythmia-susceptibility genes do not contribute significantly to the occurrence of BrS in a population with European ancestry. Extreme caution should thus be taken when interpreting genetic variation in molecular diagnostic setting, since rare coding variants were observed in a similar extent among cases versus controls, for most previously reported BrS-susceptibility genes.


Journal of the American College of Cardiology | 2012

Multifocal ectopic Purkinje-related premature contractions: a new SCN5A-related cardiac channelopathy.

Gabriel Laurent; Samuel Saal; Mohamed Yassine Amarouch; Delphine M. Béziau; Roos F. Marsman; Laurence Faivre; Julien Barc; Christian Dina; Géraldine Bertaux; Olivier Barthez; Christel Thauvin-Robinet; Philippe Charron; Véronique Fressart; Alice Maltret; Elisabeth Villain; Estelle Baron; Jean Mérot; Rodolphe Turpault; Yves Coudière; Flavien Charpentier; Jean-Jacques Schott; Gildas Loussouarn; Arthur A.M. Wilde; Jean-Eric Wolf; Isabelle Baró; Florence Kyndt; Vincent Probst

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.


Journal of the American College of Cardiology | 2011

Screening for copy number variation in genes associated with the long QT syndrome: clinical relevance.

Julien Barc; François Briec; Sébastien Schmitt; Florence Kyndt; Martine Le Cunff; Estelle Baron; Claude Vieyres; Frederic Sacher; Richard Redon; Cédric Le Caignec; Hervé Le Marec; Vincent Probst; Jean-Jacques Schott

OBJECTIVES The aim of this study was to investigate, in a set of 93 mutation-negative long QT syndrome (LQTS) probands, the frequency of copy number variants (CNVs) in LQTS genes. BACKGROUND LQTS is an inherited cardiac arrhythmia characterized by a prolonged heart rate-corrected QT (QTc) interval associated with sudden cardiac death. Recent studies suggested the involvement of duplications or deletions in the occurrence of LQTS. However, their frequency remains unknown in LQTS patients. METHODS Point mutations in KCNQ1, KCNH2, and SCN5A genes were excluded by denaturing high-performance liquid chromatography or direct sequencing. We applied Multiplex Ligation-dependent Probe Amplification (MLPA) to detect CNVs in exons of these 3 genes. Abnormal exon copy numbers were confirmed by quantitative multiplex PCR of short fluorescent fragment (QMPSF). Array-based comparative genomic hybridization (array CGH) analysis was performed using Agilent Human Genome 244K Microarrays to further map the genomic rearrangements. RESULTS We identified 3 different deletions in 3 unrelated families: 1 in KCNQ1 and 2 involving KCNH2. We showed in the largest family that the deletion involving KCNH2 is fully penetrant and segregates with the long QT phenotype in 7 affected members. CONCLUSIONS Our study demonstrates that CNVs in KCNQ1 and KCNH2 explain around 3% of LQTS in patients with no point mutation in these genes. This percentage is likely higher than the frequency of point mutations in ANKB, KCNE1, KCNE2, KCNJ2, CACNA1C, CAV3, SCN4B, AKAP9, and SNTA1 together. Thus, we propose that CNV screening in KCNQ1 and KCNH2 may be performed routinely in LQTS patients.


Circulation-arrhythmia and Electrophysiology | 2012

A connexin40 mutation associated with a malignant variant of progressive familial heart block type I.

Naomasa Makita; Akiko Seki; Naokata Sumitomo; Halina S. Chkourko; Shigetomo Fukuhara; Hiroshi Watanabe; Wataru Shimizu; Connie R. Bezzina; Can Hasdemir; Hideo Mugishima; Takeru Makiyama; Alban Baruteau; Estelle Baron; Minoru Horie; Nobuhisa Hagiwara; Arthur A.M. Wilde; Vincent Probst; Hervé Le Marec; Dan M. Roden; Naoki Mochizuki; Jean-Jacques Schott; Mario Delmar

Background— Progressive familial heart block type I (PFHBI) is a hereditary arrhythmia characterized by progressive conduction disturbances in the His-Purkinje system. PFHBI has been linked to genes such as SCN5A that influence cardiac excitability but not to genes that influence cell-to-cell communication. Our goal was to explore whether nucleotide substitutions in genes coding for connexin proteins would associate with clinical cases of PFHBI and if so, to establish a genotype-cell phenotype correlation for that mutation. Methods and Results— We screened 156 probands with PFHBI. In addition to 12 sodium channel mutations, we found a germ line GJA5 (connexin40 [Cx40]) mutation (Q58L) in 1 family. Heterologous expression of Cx40-Q58L in connexin-deficient neuroblastoma cells resulted in marked reduction of junctional conductance (Cx40-wild type [WT], 22.2±1.7 nS, n=14; Cx40-Q58L, 0.56±0.34 nS, n=14; P<0.001) and diffuse localization of immunoreactive proteins in the vicinity of the plasma membrane without formation of gap junctions. Heteromeric cotransfection of Cx40-WT and Cx40-Q58L resulted in homogenous distribution of proteins in the plasma membrane rather than in membrane plaques in ≈50% of cells; well-defined gap junctions were observed in other cells. Junctional conductance values correlated with the distribution of gap junction plaques. Conclusions— Mutation Cx40-Q58L impairs gap junction formation at cell-cell interfaces. This is the first demonstration of a germ line mutation in a connexin gene that associates with inherited ventricular arrhythmias and emphasizes the importance of Cx40 in normal propagation in the specialized conduction system.


International Journal of Cardiology | 2016

Targeted resequencing identifies TRPM4 as a major gene predisposing to progressive familial heart block type I

Xavier Daumy; Mohamed Yassine Amarouch; Pierre Lindenbaum; Stéphanie Bonnaud; Eric Charpentier; Beatrice Bianchi; Sabine Naomi Nafzger; Estelle Baron; Swanny Fouchard; Aurélie Thollet; Florence Kyndt; Julien Barc; Solena Le Scouarnec; Naomasa Makita; Hervé Le Marec; Christian Dina; Jean-Baptiste Gourraud; Vincent Probst; Hugues Abriel; Richard Redon; Jean-Jacques Schott

BACKGROUND Progressive cardiac conduction disease (PCCD) is one of the most common cardiac conduction disturbances. It has been causally related to rare mutations in several genes including SCN5A, SCN1B, TRPM4, LMNA and GJA5. METHODS AND RESULTS In this study, by applying targeted next-generation sequencing (NGS) in 95 unrelated patients with PCCD, we have identified 13 rare variants in the TRPM4 gene, two of which are currently absent from public databases. This gene encodes a cardiac calcium-activated cationic channel which precise role and importance in cardiac conduction and disease is still debated. One novel variant, TRPM4-p.I376T, is carried by the proband of a large French 4-generation pedigree. Systematic familial screening showed that a total of 13 family members carry the mutation, including 10 out of the 11 tested affected individuals versus only 1 out of the 21 unaffected ones. Functional and biochemical analyses were performed using HEK293 cells, in whole-cell patch-clamp configuration and Western blotting. TRPM4-p.I376T results in an increased current density concomitant to an augmented TRPM4 channel expression at the cell surface. CONCLUSIONS This study is the first extensive NGS-based screening of TRPM4 coding variants in patients with PCCD. It reports the third largest pedigree diagnosed with isolated Progressive Familial Heart Block type I and confirms that this subtype of PCCD is caused by mutation-induced gain-of-expression and function of the TRPM4 ion channel.


Journal of the American Heart Association | 2016

Variants of Transient Receptor Potential Melastatin Member 4 in Childhood Atrioventricular Block

Ninda Ratna Maharani Syam; Stéphanie Chatel; Lijo Cherian Ozhathil; Valentin Sottas; Jean-Sébastien Rougier; Alban Baruteau; Estelle Baron; Mohamed Yassine Amarouch; Xavier Daumy; Vincent Probst; Jean-Jacques Schott; Hugues Abriel

Background Transient receptor potential melastatin member 4 (TRPM4) is a nonselective cation channel. TRPM4 mutations have been linked to cardiac conduction disease and Brugada syndrome. The mechanisms underlying TRPM4‐dependent conduction slowing are not fully understood. The aim of this study was to characterize TRPM4 genetic variants found in patients with congenital or childhood atrioventricular block. Methods and Results Ninety‐one patients with congenital or childhood atrioventricular block were screened for candidate genes. Five rare TRPM4 genetic variants were identified and investigated. The variants were expressed heterologously in HEK293 cells. Two of the variants, A432T and A432T/G582S, showed decreased expression of the protein at the cell membrane; inversely, the G582S variant showed increased expression. Further functional characterization of these variants using whole‐cell patch‐clamp configuration showed a loss of function and a gain of function, respectively. We hypothesized that the observed decrease in expression was caused by a folding and trafficking defect. This was supported by the observation that incubation of these variants at lower temperature partially rescued their expression and function. Previous studies have suggested that altered SUMOylation of TRPM4 may cause a gain of function; however, we did not find any evidence that supports SUMOylation as being directly involved for the gain‐of‐function variant. Conclusions This study underpins the role of TRPM4 in the cardiac conduction system. The loss‐of‐function variants A432T/G582S found in 2 unrelated patients with atrioventricular block are most likely caused by misfolding‐dependent altered trafficking. The ability to rescue this variant with lower temperature may provide a novel use of pharmacological chaperones in treatment strategies.


Heart | 2012

Identification of a strong genetic background for progressive cardiac conduction defect by epidemiological approach

Jean Baptiste Gourraud; Florence Kyndt; Swanny Fouchard; Eric Rendu; Philippe Jaafar; Claude Gully; Karim Gacem; Jean Marc Dupuis; Aurelie Longueville; Estelle Baron; Matilde Karakachoff; Jean Pierre Cebron; Stéphanie Chatel; Jean-Jacques Schott; Hervé Le Marec; Vincent Probst

Introduction Progressive cardiac conduction defect (PCCD) is a frequent disease attributed to degeneration and fibrosis of the His bundle. Over the past years, gene defects have been identified demonstrating that PCCD could be a genetic disease. The aim of this study was to show a familial aggregation for PCCD using a genetic epidemiological approach to improve in fine genetic knowledge of the transmission of the disease. Methods and results Using the French social security number, the authors have been able to determine the city of birth of the 6667 patients implanted with a pacemaker (PM) for PCCD between 1995 and 2005 in the western part of France. The authors then mapped the frequency of PM implantations for PCCD. A large heterogeneity of the frequency of the disease has been observed, with a frequency of 0.21% in the major city (Nantes) ranging up to 2.28% in specific parishes. Familial studies performed in the parishes with the highest frequency of the disease allowed the authors to identify five large families with PCCD. Clinical investigations demonstrated phenotype heterogeneity between families. Three patterns have been differentiated. Conclusions This study demonstrates a disparate geographical repartition of the frequency of PM implantation in the area of the authors at least in part related to a hereditary factor. The identification of five large families affected by PCCD using epidemiological approach underlines the existence of a major genetic background in PCCD.


Journal of the American Heart Association | 2016

Dysfunction of the Voltage‐Gated K+ Channel β2 Subunit in a Familial Case of Brugada Syndrome

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; Richard Redon

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 SCN5A, 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 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. Conclusions Altogether, our results indicate that Kvβ2 dysfunction can contribute to the Brugada electrocardiographic pattern.


Archives of Cardiovascular Diseases Supplements | 2015

0185 : Genetic screening identifies a high proportion of mutations in patients with idiopathic ventricular fibrillation and sudden cardiac death

Vincent Probst; Solena Le Scouarnec; Florence Kyndt; Jean-Jacques Schott; Jean-Baptiste Gourraud; Frederic Sacher; Philippe Mabo; Matilde Karakachoff; Stéphanie Bonnaud; Jade Violleau; Eloi Marijon; Florence Dumas; Alain Cariou; Estelle Baron; Pierre Lindenbaum; Xavier Jouven; Richard Redon

Introduction Several gene defects are associated with idiopathic ventricular fibrillation (IVF) and sudden cardiac death (SCD). The recent development of NGS-based mutation screening provides a unique opportunity to estimate extensively the spectrum and prevalence of rare variants in genes associated with cardiac diseases. Methods Cohort 1 was composed of 75 patients resuscitated from cardiac arrest due to IVF. All patients have undergone a complete clinical cardiac examination including 12 lead-ECG, cardiac echography, coronography and exercise test. Cohort 2 was composed of 99 victims of SCD related to ventricular fibrillation younger than 45 years old and without explanation for the SCD at the time of the reanimation. Genetic screening was based on the use of the HaloPlex(tm) Target Enrichment System (Agilent Technologies) prior to HiSeq sequencing (Illumina). The custom kit designed for this study covers 163 genes previously reported as involved in cardiac arrhythmias, conduction defect and cardiomyopathies. Results In cohort 1, the mean age was 36±10 years with a male predominance (52 males, 69%). In cohort 2, the mean age was 37±7 years with a male predominance (76 males, 79%). In cohort 1, we identified 50 putative mutations in 35 patients (47%). In cohort 2, we identified 30 putative mutations in 24 patients (24%). Table Cohort 1 (n=75) Cohort 2 (n=99) SCN5A 7 (9%) 0 HCM and DCM genes 22 (29%) 17 (17%) ARVD/C genes 4 (5%) 3 (3%) LQTS genes (excluding SCN5A) 4 (5%) 4 (4%) CPVT genes (excluding SCN5A) 7 (9%) 3 (3%) BRS genes 3 (4%) 1 (1%) Other 3 (4%) 2 (2%) Conclusion Our study identified mutations in almost 50 % of IVF patients after a complete cardiac evaluation. These results suggest that molecular analysis must be part of the work up in this kind of patients. In young patients affected by unexplained sudden death, the molecular analyses are less contributive probably because of a more important percentage of patients affected by ischemic cardiomyopathies.


Archive | 2012

SCN5A-related cardiac channelopathy

Gabriel Laurent; Samuel Saal; Mohamed Yassine Amarouch; Delphine M. Béziau; Laurence Faivre; Julien Barc; Christian Dina; Géraldine Bertaux; Olivier Barthez; Christel Thauvin-Robinet; Philippe Charron; Véronique Fressart; Alice Maltret; Elisabeth Villain; Estelle Baron; Rodolphe Turpault; Flavien Charpentier; Jean-Jacques Schott; Gildas Loussouarn; Arthur A.M. Wilde; Isabelle Baró; Florence Kyndt; Vincent Probst

Collaboration


Dive into the Estelle Baron's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hervé Le Marec

French Institute of Health and Medical Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge