Network


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

Hotspot


Dive into the research topics where Jean Mérot is active.

Publication


Featured researches published by Jean Mérot.


The EMBO Journal | 2001

Polarized trafficking and surface expression of the AQP4 water channel are coordinated by serial and regulated interactions with different clathrin-adaptor complexes

Ricardo Madrid; Sophie Le Maout; Marie‐Bénédicte Barrault; Katy Janvier; Serge Benichou; Jean Mérot

Aquaporin 4 (AQP4) is the predominant water channel in the brain. It is targeted to specific membrane domains of astrocytes and plays a crucial role in cerebral water balance in response to brain edema formation. AQP4 is also specifically expressed in the basolateral membranes of epithelial cells. However, the molecular mechanisms involved in its polarized targeting and membrane trafficking remain largely unknown. Here, we show that two independent C‐terminal signals determine AQP4 basolateral membrane targeting in epithelial MDCK cells. One signal involves a tyrosine‐based motif; the other is encoded by a di‐leucine‐like motif. We found that the tyrosine‐based basolateral sorting signal also determines AQP4 clathrin‐dependent endocytosis through direct interaction with the μ subunit of AP2 adaptor complex. Once endocytosed, a regulated switch in μ subunit interaction changes AP2 adaptor association to AP3. We found that the stress‐induced kinase casein kinase (CK)II phosphorylates the Ser276 immediately preceding the tyrosine motif, increasing AQP4–μ3A interaction and enhancing AQP4–lysosomal targeting and degradation. AQP4 phosphorylation by CKII may thus provide a mechanism that regulates AQP4 cell surface expression.


Circulation Research | 2005

Impaired KCNQ1-KCNE1 and Phosphatidylinositol-4,5-Bisphosphate Interaction Underlies the Long QT Syndrome

Kyu-Ho Park; Julien Piron; Shehrazade Dahimène; Jean Mérot; Isabelle Baró; Denis Escande; Gildas Loussouarn

Nearly a hundred different KCNQ1 mutations have been reported as leading to the cardiac long QT syndrome, characterized by prolonged QT interval, syncopes, and sudden death. We have previously shown that phosphatidylinositol-4,5-bisphosphate (PIP2) regulates the KCNQ1–KCNE1 complex. In the present study, we show that PIP2 affinity is reduced in three KCNQ1 mutant channels (R243H, R539W, and R555C) associated with the long QT syndrome. In giant excised patches, direct application of PIP2 on the cytoplasmic face of the three mutant channels counterbalances the loss of function. Reintroduction of a positive charge by application of methanethiosulfonate ethylammonium on the cytoplasmic face of R555C mutant channels also restores channel activity. The channel affinity for a soluble analog of PIP2 is decreased in the three mutant channels. By using a model that describes the KCNQ1–KCNE1 channel behavior and by fitting the relationship between the kinetics of deactivation and the current amplitude obtained in whole-cell experiments, we estimated the PIP2 binding and dissociation rates on wild-type and mutant channels. The dissociation rate of the three mutants was higher than for the wild-type channel, suggesting a decreased affinity for PIP2. PIP2 binding was magnesium-dependent, and the PIP2-dependent equilibrium constant in the absence of magnesium was higher with the wild-type than with the mutant channels. Altogether, our data suggest that a reduced PIP2 affinity of KCNQ1 mutants can lead to the long QT syndrome.


Nature Reviews Cardiology | 2015

Mitral valve disease—morphology and mechanisms

Robert A. Levine; Albert Hagège; Daniel P. Judge; Muralidhar Padala; Jacob P. Dal-Bianco; Elena Aikawa; Jonathan Beaudoin; Joyce Bischoff; Nabila Bouatia-Naji; Patrick Bruneval; Jonathan T. Butcher; Alain Carpentier; Miguel Chaput; Adrian H. Chester; Catherine Clusel; Francesca N. Delling; Harry C. Dietz; Christian Dina; Ronen Durst; Leticia Fernandez-Friera; Mark D. Handschumacher; Morten O. Jensen; Xavier Jeunemaitre; Hervé Le Marec; Thierry Le Tourneau; Roger R. Markwald; Jean Mérot; Emmanuel Messas; David P. Milan; Tui Neri

Mitral valve disease is a frequent cause of heart failure and death. Emerging evidence indicates that the mitral valve is not a passive structure, but—even in adult life—remains dynamic and accessible for treatment. This concept motivates efforts to reduce the clinical progression of mitral valve disease through early detection and modification of underlying mechanisms. Discoveries of genetic mutations causing mitral valve elongation and prolapse have revealed that growth factor signalling and cell migration pathways are regulated by structural molecules in ways that can be modified to limit progression from developmental defects to valve degeneration with clinical complications. Mitral valve enlargement can determine left ventricular outflow tract obstruction in hypertrophic cardiomyopathy, and might be stimulated by potentially modifiable biological valvular–ventricular interactions. Mitral valve plasticity also allows adaptive growth in response to ventricular remodelling. However, adverse cellular and mechanobiological processes create relative leaflet deficiency in the ischaemic setting, leading to mitral regurgitation with increased heart failure and mortality. Our approach, which bridges clinicians and basic scientists, enables the correlation of observed disease with cellular and molecular mechanisms, leading to the discovery of new opportunities for improving the natural history of mitral valve disease.


The Journal of Physiology | 2008

Kv7.1 (KCNQ1) properties and channelopathies

David Peroz; Nicolas Rodriguez; Frank S. Choveau; Isabelle Baró; Jean Mérot; Gildas Loussouarn

KCNQ1 is the pore‐forming subunit of a channel complex whose expression and function have been rather well characterized in the heart. Almost 300 mutations of KCNQ1 have been identified in patients and a vast majority of the described mutations are linked to the long QT syndrome. Only a few mutations are linked to other pathologies such as atrial fibrillation and the short QT syndrome. However, a considerable amount of work remains to be done to get a clear picture of the molecular mechanisms responsible for the pathogenesis related to each mutation. The present review gives three examples of recent studies towards this goal and illustrates the diversity of the molecular mechanisms involved.


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.


Biochimica et Biophysica Acta | 1989

Two types of K+ channels in the apical membrane of rabbit proximal tubule in primary culture

Jean Mérot; Michel Bidet; Sophie Le Maout; M. Tauc; P. Poujeol

The patch-clamp technique was used to investigate ionic channels in the apical membrane of rabbit proximal tubule cells in primary culture. Cell-attached recordings revealed the presence of a highly selective K+ channel with a conductance of 130 pS. The channel activity was increased with membrane depolarization. Experiments performed on excised patches showed that the channel activity depended on the free Ca2+ concentration on the cytoplasmic face of the membrane and that decreasing the cytoplasmic pH from 7.2 to 6.0 also decreased the channel activity. In symmetrical 140 mM KCl solutions the channel conductance was 200 pS. The channel was blocked by barium, tetraethylammonium and Leiurus quinquestriatus scorpion venom (from which charybdotoxin is extracted) when applied to the extracellular face of the channel. Barium and quinidine also blocked the channel when applied to the cytoplasmic face of the membrane. Another K+ channel with a conductance of 42 pS in symmetrical KCl solutions was also observed in excised patches. The channel was blocked by barium and apamin, but not by tetraethylammonium applied to the extracellular face of the membrane. Using the whole-cell recording configuration we determined a K+ conductance of 4.96 nS per cell that was blocked by 65% when 10 mM tetraethylammonium was applied to the bathing medium.


Circulation Research | 2008

Long QT Syndrome–Associated Mutations in KCNQ1 and KCNE1 Subunits Disrupt Normal Endosomal Recycling of IKs Channels

Guiscard Seebohm; Nathalie Strutz-Seebohm; Oana N. Ureche; Ulrike Henrion; Ravshan Baltaev; Andreas F. Mack; Ganna Korniychuk; Katja Steinke; Daniel Tapken; Arne Pfeufer; Stefan Kääb; Cecilia Bucci; Bernard Attali; Jean Mérot; Jeremy M. Tavaré; Uta C. Hoppe; Michael C. Sanguinetti; Florian Lang

Physical and emotional stress is accompanied by release of stress hormones such as the glucocorticoid cortisol. This hormone upregulates the serum- and glucocorticoid-inducible kinase (SGK)1, which in turn stimulates IKs, a slow delayed rectifier potassium current that mediates cardiac action potential repolarization. Mutations in IKs channel &agr; (KCNQ1, KvLQT1, Kv7.1) or &bgr; (KCNE1, IsK, minK) subunits cause long QT syndrome (LQTS), an inherited cardiac arrhythmia associated with increased risk of sudden death. Together with the GTPases RAB5 and RAB11, SGK1 facilitates membrane recycling of KCNQ1 channels. Here, we show altered SGK1-dependent regulation of LQTS-associated mutant IKs channels. Whereas some mutant KCNQ1 channels had reduced basal activity but were still activated by SGK1, currents mediated by KCNQ1(Y111C) or KCNQ1(L114P) were paradoxically reduced by SGK1. Heteromeric channels coassembled of wild-type KCNQ1 and the LQTS-associated KCNE1(D76N) mutant were similarly downregulated by SGK1 because of a disrupted RAB11-dependent recycling. Mutagenesis experiments indicate that stimulation of IKs channels by SGK1 depends on residues H73, N75, D76, and P77 in KCNE1. Identification of the IKs recycling pathway and its modulation by stress-stimulated SGK1 provides novel mechanistic insight into potentially fatal cardiac arrhythmias triggered by physical or psychological stress.


Circulation Research | 2006

14-3-3 Is a Regulator of the Cardiac Voltage-Gated Sodium Channel Nav1.5

Marie Allouis; Françoise Le Bouffant; Ronald Wilders; David Peroz; Jean-Jacques Schott; Jacques Noireaud; Hervé Le Marec; Jean Mérot; Denis Escande; Isabelle Baró

The voltage-sensitive Na+ channel Nav1.5 plays a crucial role in generating and propagating the cardiac action potential and its dysfunction promotes cardiac arrhythmias. The channel takes part into a large molecular complex containing regulatory proteins. Thus, factors that modulate its biosynthesis, localization, activity, and/or degradation are of great interest from both a physiological and pathological standpoint. Using a yeast 2-hybrid screen, we unveiled a novel partner, 14-3-3&eegr;, interacting with the Nav1.5 cytoplasmic I interdomain. The interaction was confirmed by coimmunoprecipitation of 14-3-3 and full-length Nav1.5 both in COS-7 cells expressing recombinant Nav1.5 and in mouse cardiac myocytes. Using immunocytochemistry, we also found that 14-3-3 and Nav1.5 colocalized at the intercalated discs. We tested the functional link between Nav1.5 and 14-3-3&eegr; using the whole-cell patch-clamp configuration. Coexpressing Nav1.5, the &bgr;1 subunit and 14-3-3&eegr; induced a negative shift in the inactivation curve of the Na+ current, a delayed recovery from inactivation, but no changes in the activation curve or in the current density. The negative shift was reversed, and the recovery from inactivation was normalized by overexpressing the Nav1.5 cytoplasmic I interdomain interacting with 14-3-3&eegr;. Reversal was also obtained with the dominant negative R56,60A 14-3-3&eegr; mutant, suggesting that dimerization of 14-3-3 is needed for current regulation. Computer simulations suggest that the absence of 14-3-3 could exert proarrhythmic effects on cardiac electrical restitution properties. Based on these findings, we propose that the 14-3-3 protein is a novel component of the cardiac Na+ channel acting as a cofactor for the regulation of the cardiac Na+ current.


Journal of Molecular and Cellular Cardiology | 2010

Delayed rectifier K+ currents and cardiac repolarization

Flavien Charpentier; Jean Mérot; Gildas Loussouarn; Isabelle Baró

The two components of the cardiac delayed rectifier current have been the subject of numerous studies since firstly described. This current controls the action potential duration and is highly regulated. After identification of the channel subunits underlying IKs, KCNQ1 associated with KCNE1, and IKr, HERG, their involvement in human cardiac channelopathies have provided various models allowing the description of the molecular mechanisms of the KCNQ1 and HERG channels trafficking, activity and regulation. More recently, studies have been focusing on the unveiling of different partners of the pore-forming proteins that contribute to their maturation, trafficking, activity and/or degradation, on one side, and on their respective expression in the heterogeneous cardiac tissue, on the other side. The aim of this review is to report and discuss the major works on IKs and IKr and the most recent ones that help to understand the precise function of these currents in the heart.


Circulation Research | 2006

The N-Terminal Juxtamembranous Domain of KCNQ1 is Critical for Channel Surface Expression. Implications in the Romano-Ward LQT1 Syndrome

Shehrazade Dahimène; Sébastien Alcoléa; Patrice Naud; Philippe Jourdon; Denis Escande; Robert Brasseur; Annick Thomas; Isabelle Baró; Jean Mérot

N-terminal mutations in the KCNQ1 channel are frequently linked to fatal arrhythmias in newborn children and adolescents but the cellular mechanisms involved in this dramatic issue remain, however, to be discovered. Here, we analyzed the trafficking of a series of N-terminal truncation mutants and identified a critical trafficking motif of KCNQ1. This determinant is located in the juxtamembranous region preceding the first transmembrane domain of the protein. Three mutations (Y111C, L114P and P117L) implicated in inherited Romano-Ward LQT1 syndrome, are embedded within this domain. Reexpression studies in both COS-7 cells and cardiomyocytes showed that the mutant proteins fail to exit the endoplasmic reticulum. KCNQ1 subunits harboring Y111C or L114P exert a dominant negative effect on the wild-type KCNQ1 subunit by preventing plasma membrane trafficking of heteromultimeric channels. The P117L mutation had a less pronounced effect on the trafficking of heteromultimeric channels but altered the kinetics of the current. Furthermore, we showed that the trafficking determinant in KCNQ1 is structurally and functionally conserved in other KCNQ channels and constitutes a critical trafficking determinant of the KCNQ channel family. Computed structural predictions correlated the potential structural changes introduced by the mutations with impaired protein trafficking. In conclusion, our studies unveiled a new role of the N-terminus of KCNQ channels in their trafficking and its implication in severe forms of LQT1 syndrome.

Collaboration


Dive into the Jean Mérot'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
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Russell A. Norris

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Albert Hagège

Paris Descartes University

View shared research outputs
Researchain Logo
Decentralizing Knowledge