100 results on '"Gildas, Loussouarn"'
Search Results
2. A need for exhaustive and standardized characterization of ion channels activity. The case of KV11.1
- Author
-
Malak Alameh, Barbara Ribeiro Oliveira-Mendes, Florence Kyndt, Jordan Rivron, Isabelle Denjoy, Florian Lesage, Jean-Jacques Schott, Michel De Waard, and Gildas Loussouarn
- Subjects
KCNH2 gene ,variants ,electrophysiology ,long QT syndrome ,phenotyping ,Physiology ,QP1-981 - Abstract
hERG, the pore-forming subunit of the rapid component of the delayed rectifier K+ current, plays a key role in ventricular repolarization. Mutations in the KCNH2 gene encoding hERG are associated with several cardiac rhythmic disorders, mainly the Long QT syndrome (LQTS) characterized by prolonged ventricular repolarization, leading to ventricular tachyarrhythmias, sometimes progressing to ventricular fibrillation and sudden death. Over the past few years, the emergence of next-generation sequencing has revealed an increasing number of genetic variants including KCNH2 variants. However, the potential pathogenicity of the majority of the variants remains unknown, thus classifying them as variants of uncertain significance or VUS. With diseases such as LQTS being associated with sudden death, identifying patients at risk by determining the variant pathogenicity, is crucial. The purpose of this review is to describe, on the basis of an exhaustive examination of the 1322 missense variants, the nature of the functional assays undertaken so far and their limitations. A detailed analysis of 38 hERG missense variants identified in Long QT French patients and studied in electrophysiology also underlies the incomplete characterization of the biophysical properties for each variant. These analyses lead to two conclusions: first, the function of many hERG variants has never been looked at and, second, the functional studies done so far are excessively heterogeneous regarding the stimulation protocols, cellular models, experimental temperatures, homozygous and/or the heterozygous condition under study, a context that may lead to conflicting conclusions. The state of the literature emphasizes how necessary and important it is to perform an exhaustive functional characterization of hERG variants and to standardize this effort for meaningful comparison among variants. The review ends with suggestions to create a unique homogeneous protocol that could be shared and adopted among scientists and that would facilitate cardiologists and geneticists in patient counseling and management.
- Published
- 2023
- Full Text
- View/download PDF
3. A functional network of highly pure enteric neurons in a dish
- Author
-
Martial Caillaud, Morgane E. Le Dréan, Adrien De-Guilhem-de-Lataillade, Catherine Le Berre-Scoul, Jérôme Montnach, Steven Nedellec, Gildas Loussouarn, Vincent Paillé, Michel Neunlist, and Hélène Boudin
- Subjects
enteric neuron ,enteric synapse ,spontaneous activity ,lipid-mediated transfection ,multi-electrode array ,patch-clamp ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
The enteric nervous system (ENS) is the intrinsic nervous system that innervates the entire digestive tract and regulates major digestive functions. Recent evidence has shown that functions of the ENS critically rely on enteric neuronal connectivity; however, experimental models to decipher the underlying mechanisms are limited. Compared to the central nervous system, for which pure neuronal cultures have been developed for decades and are recognized as a reference in the field of neuroscience, an equivalent model for enteric neurons is lacking. In this study, we developed a novel model of highly pure rat embryonic enteric neurons with dense and functional synaptic networks. The methodology is simple and relatively fast. We characterized enteric neurons using immunohistochemical, morphological, and electrophysiological approaches. In particular, we demonstrated the applicability of this culture model to multi-electrode array technology as a new approach for monitoring enteric neuronal network activity. This in vitro model of highly pure enteric neurons represents a valuable new tool for better understanding the mechanisms involved in the establishment and maintenance of enteric neuron synaptic connectivity and functional networks.
- Published
- 2023
- Full Text
- View/download PDF
4. 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
5. Computer modeling of whole-cell voltage-clamp analyses to delineate guidelines for good practice of manual and automated patch-clamp
- Author
-
Jérôme Montnach, Maxime Lorenzini, Adrien Lesage, Isabelle Simon, Sébastien Nicolas, Eléonore Moreau, Céline Marionneau, Isabelle Baró, Michel De Waard, and Gildas Loussouarn
- Subjects
Medicine ,Science - Abstract
Abstract The patch-clamp technique and more recently the high throughput patch-clamp technique have contributed to major advances in the characterization of ion channels. However, the whole-cell voltage-clamp technique presents certain limits that need to be considered for robust data generation. One major caveat is that increasing current amplitude profoundly impacts the accuracy of the biophysical analyses of macroscopic ion currents under study. Using mathematical kinetic models of a cardiac voltage-gated sodium channel and a cardiac voltage-gated potassium channel, we demonstrated how large current amplitude and series resistance artefacts induce an undetected alteration in the actual membrane potential and affect the characterization of voltage-dependent activation and inactivation processes. We also computed how dose–response curves are hindered by high current amplitudes. This is of high interest since stable cell lines frequently demonstrating high current amplitudes are used for safety pharmacology using the high throughput patch-clamp technique. It is therefore critical to set experimental limits for current amplitude recordings to prevent inaccuracy in the characterization of channel properties or drug activity, such limits being different from one channel type to another. Based on the predictions generated by the kinetic models, we draw simple guidelines for good practice of whole-cell voltage-clamp recordings.
- Published
- 2021
- Full Text
- View/download PDF
6. Up-regulation of voltage-gated sodium channels by peptides mimicking S4-S5 linkers reveals a variation of the ligand-receptor mechanism
- Author
-
Olfat A. Malak, Fayal Abderemane-Ali, Yue Wei, Fabien C. Coyan, Gilyane Pontus, David Shaya, Céline Marionneau, and Gildas Loussouarn
- Subjects
Medicine ,Science - Abstract
Abstract Prokaryotic NaV channels are tetramers and eukaryotic NaV channels consist of a single subunit containing four domains. Each monomer/domain contains six transmembrane segments (S1-S6), S1-S4 being the voltage-sensor domain and S5-S6 the pore domain. A crystal structure of NaVMs, a prokaryotic NaV channel, suggests that the S4-S5 linker (S4-S5L) interacts with the C-terminus of S6 (S6T) to stabilize the gate in the open state. However, in several voltage-gated potassium channels, using specific S4-S5L-mimicking peptides, we previously demonstrated that S4-S5L/S6T interaction stabilizes the gate in the closed state. Here, we used the same strategy on another prokaryotic NaV channel, NaVSp1, to test whether equivalent peptides stabilize the channel in the open or closed state. A NaVSp1-specific S4-S5L peptide, containing the residues supposed to interact with S6T according to the NaVMs structure, induced both an increase in NaVSp1 current density and a negative shift in the activation curve, consistent with S4-S5L stabilizing the open state. Using this approach on a human NaV channel, hNaV1.4, and testing 12 hNaV1.4 S4-S5L peptides, we identified four activating S4-S5L peptides. These results suggest that, in eukaryotic NaV channels, the S4-S5L of DI, DII and DIII domains allosterically modulate the activation gate and stabilize its open state.
- Published
- 2020
- Full Text
- View/download PDF
7. A standardised hERG phenotyping pipeline to evaluate KCNH2 genetic variant pathogenicity
- Author
-
Barbara Oliveira‐Mendes, Sylvain Feliciangeli, Mélissa Ménard, Frank Chatelain, Malak Alameh, Jérôme Montnach, Sébastien Nicolas, Béatrice Ollivier, Julien Barc, Isabelle Baró, Jean‐Jacques Schott, Vincent Probst, Florence Kyndt, Isabelle Denjoy, Florian Lesage, Gildas Loussouarn, and Michel De Waard
- Subjects
arrhythmias ,diagnostic testing ,genetic variant ,hERG ion channel ,pathogenicity ,QT syndrome ,Medicine (General) ,R5-920 - Abstract
Abstract Background and aims Mutations in KCNH2 cause long or short QT syndromes (LQTS or SQTS) predisposing to life‐threatening arrhythmias. Over 1000 hERG variants have been described by clinicians, but most remain to be characterised. The objective is to standardise and accelerate the phenotyping process to contribute to clinician diagnosis and patient counselling. In silico evaluation was also included to characterise the structural impact of the variants. Methods We selected 11 variants from known LQTS patients and two variants for which diagnosis was problematic. Using the Gibson assembly strategy, we efficiently introduced mutations in hERG cDNA despite GC‐rich sequences. A pH‐sensitive fluorescent tag was fused to hERG for efficient evaluation of channel trafficking. An optimised 35‐s patch‐clamp protocol was developed to evaluate hERG channel activity in transfected cells. R software was used to speed up analyses. Results In the present work, we observed a good correlation between cell surface expression, assessed by the pH‐sensitive tag, and current densities. Also, we showed that the new biophysical protocol allows a significant gain of time in recording ion channel properties and provides extensive information on WT and variant channel biophysical parameters, that can all be recapitulated in a single parameter defined herein as the repolarisation power. The impacts of the variants on channel structure were also reported where structural information was available. These three readouts (trafficking, repolarisation power and structural impact) define three pathogenicity indexes that may help clinical diagnosis. Conclusions Fast‐track characterisation of KCNH2 genetic variants shows its relevance to discriminate mutants that affect hERG channel activity from variants with undetectable effects. It also helped the diagnosis of two new variants. This information is meant to fill a patient database, as a basis for personalised medicine. The next steps will be to further accelerate the process using an automated patch‐clamp system.
- Published
- 2021
- Full Text
- View/download PDF
8. Editorial: Molecular Mechanisms of Voltage-Gating in Ion Channels
- Author
-
Gildas Loussouarn and Mounir Tarek
- Subjects
voltage-gated ion channel ,patch - clamp technique ,toxins ,cryo-EM ,VSD gate coupling ,Therapeutics. Pharmacology ,RM1-950 - Published
- 2021
- Full Text
- View/download PDF
9. Nav1.2 and BK channel interaction shapes the action potential in the axon initial segment
- Author
-
Luiza Filipis, Laila Ananda Blömer, Jérôme Montnach, Gildas Loussouarn, Michel De Waard, and Marco Canepari
- Subjects
Physiology - Published
- 2023
- Full Text
- View/download PDF
10. 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
11. hERG S4-S5 linker acts as a voltage-dependent ligand that binds to the activation gate and locks it in a closed state
- Author
-
Olfat A. Malak, Zeineb Es-Salah-Lamoureux, and Gildas Loussouarn
- Subjects
Medicine ,Science - Abstract
Abstract Delayed-rectifier potassium channels (hERG and KCNQ1) play a major role in cardiac repolarization. These channels are formed by a tetrameric pore (S5–S6) surrounded by four voltage sensor domains (S1-S4). Coupling between voltage sensor domains and the pore activation gate is critical for channel voltage-dependence. However, molecular mechanisms remain elusive. Herein, we demonstrate that covalently binding, through a disulfide bridge, a peptide mimicking the S4-S5 linker (S4-S5L) to the channel S6 C-terminus (S6T) completely inhibits hERG. This shows that channel S4-S5L is sufficient to stabilize the pore activation gate in its closed state. Conversely, covalently binding a peptide mimicking S6T to the channel S4-S5L prevents its inhibiting effect and renders the channel almost completely voltage-independent. This shows that the channel S4-S5L is necessary to stabilize the activation gate in its closed state. Altogether, our results provide chemical evidence that S4-S5L acts as a voltage-controlled ligand that binds S6T to lock the channel in a closed state, elucidating the coupling between voltage sensors and the gate in delayed rectifier potassium channels and potentially other voltage-gated channels.
- Published
- 2017
- Full Text
- View/download PDF
12. CAVIN1-Mediated Endocytosis: A Novel Mechanism Underlying The Interindividual Variability In Drug-Induced Long QT
- Author
-
Zeina R. Al Sayed, Céline Pereira, Esthel Pénard, Adeline Mallet, Charlène Jouve, Nihar Masurkar, Gildas Loussouarn, David-Alexandre Trégouët, and Jean-Sébastien Hulot
- Abstract
BackgroundDrug-induced QT prolongation (diLQT) is a feared side-effect as exposing susceptible individuals to fatal arrhythmias. The occurrence of diLQT is primarily attributed to unintended drug interactions with cardiac ion channels, notably the hERG channels that generate the repolarizing current (IKr) and thereby regulate the late repolarization phase. There is an important inter-individual susceptibility to develop diLQT which is of unknown origin but can be reproduced in patient-specific iPSC-derived cardiomyocytes (iPS-CMs).ObjectiveWe aimed to investigate the dynamics of hERG channels in response to sotalol and to identify regulators of the susceptibility to developing diLQT.MethodsWe measured electrophysiological activity and cellular distribution of hERG channels after hERG blocker treatment in iPS-CMs derived from patients with highest or lowest sensitivity (HS or LS) to sotalol administrationin vivo(i.e., based on the measure of the maximal change in QT interval 3 hours after administration). Specific small-interfering RNAs (siRNA) and CAVIN1-T2A-GFP adenovirus were used to manipulateCAVIN1expression.ResultsWhile HS and LS iPS-CMs showed similar electrophysiological characteristics at the baseline, the late repolarization phase was prolonged, and IKrsignificantly decreased after exposure of HS iPS-CMs to low sotalol concentrations. IKrreduction was caused by a rapid translocation of hERG channel from the plasma membrane to the cytoskeleton upon sotalol application. This phenomenon was suppressed by blocking active endocytosis using dynasore.CAVIN1, essential for caveolae biogenesis, was two-times more expressed in HS iPS-CMs and its knockdown using siRNA decreased their sensitivity to sotalol.CAVIN1overexpression in LS iPS-CMs using adenovirus showed reciprocal effects. Mechanistically, we found that treatment with sotalol promoted trafficking of the hERG channel from the plasma membrane to the cytoskeleton through caveolae and in a manner dependent on CAVIN1 expression.CAVIN1silencing reduced the number of caveolae at the membrane and abrogated the internalization of hERG channel in sotalol-treated HS iPS-CMs. CAVIN1 also controlled cardiomyocyte responses to other hERG blockers such as E4031, vandetanib, and clarithromycin.ConclusionsOur study identifies unbridled turnover of the potassium channel hERG as a mechanism supporting the inter-individual susceptibility underlying diLQT development and demonstrates how this phenomenon is finely tuned by CAVIN1.CLINICAL PERSPECTIVEWhat is new?-The inter-individual susceptibility underlying diLQT development involves unbridled turnover of cardiac ion channels from the plasma membrane.-This phenomenon is finely tuned by CAVIN1, a protein that is essential for essential for caveolae biogenesis.-Treatment with hERG blocker promoted trafficking of the hERG channel from the plasma membrane to the cytoskeleton through caveolae and in a manner dependent on CAVIN1 expression.What are the clinical implications?-While congenital long QT is primarily from a genetic origin, the development of drug-induced long QT involves differences in the trafficking machinery of cardiac ion channels.-The prediction of CAVIN1 expression levels could help preventing drug-induced cardiotoxicity.
- Published
- 2023
- Full Text
- View/download PDF
13. The Integrative Approach to Study of the Structure and Functions of Cardiac Voltage-Dependent Ion Channels
- Author
-
M. G. Karlova, G. S. Glukhov, H. Zhang, Y. G. Kacher, Gildas Loussouarn, E. V. Zaklyazminskaya, and Olga Sokolova
- Subjects
0303 health sciences ,Materials science ,General Chemistry ,Gating ,Condensed Matter Physics ,Ion ,03 medical and health sciences ,Electrophysiology ,Molecular dynamics ,0302 clinical medicine ,Structural biology ,Structural proteomics ,Biophysics ,General Materials Science ,Patch clamp ,030217 neurology & neurosurgery ,Ion channel ,030304 developmental biology - Abstract
Membrane proteins, including ion channels, became the focus of structural proteomics midway through the 20th century. Methods for studying ion channels are diverse and include structural (X-ray crystallography, cryoelectron microscopy, currently X-ray free electron lasers) and functional (e.g., patch clamp) approaches. This review highlights the evolution of approaches to study of the structure of cardiac ion channels, provides an overview of new techniques of structural biology concerning ion channels, including the use of lipo- and nanodiscs, and discusses the contribution of electrophysiological studies and molecular dynamics to obtain a complete picture of the structure and functioning of cardiac ion channels. Electrophysiological studies have become a powerful tool for deciphering the mechanisms of ion conductivity and selectivity, gating and regulation, as well as testing molecules of pharmacological interest. Obtaining the atomic structure of ion channels became possible by the active development of X-ray crystallography and cryoelectron micro-scopy, and, recently, with the use of XFEL.
- Published
- 2021
- Full Text
- View/download PDF
14. 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
15. 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
16. A standardised hERG phenotyping pipeline to evaluate KCNH2 genetic variant pathogenicity
- Author
-
Julien Barc, Béatrice Ollivier, Sylvain Feliciangeli, Malak Alameh, Vincent Probst, Isabelle Denjoy, Barbara Oliveira-Mendes, Jean-Jacques Schott, Frank Chatelain, S. Nicolas, Florian Lesage, Jérôme Montnach, Isabelle Baró, Michel De Waard, F. Kyndt, Mélissa Ménard, Gildas Loussouarn, 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)-Nantes Université - UFR de Médecine et des Techniques Médicales (Nantes Univ - UFR MEDECINE), Nantes Université - pôle Santé, Nantes Université (Nantes Univ)-Nantes Université (Nantes Univ)-Nantes Université - pôle Santé, Nantes Université (Nantes Univ)-Nantes Université (Nantes Univ), Laboratory of Excellence in Ion Channel Science and Therapeutics [Valbonne] (LabEx ICST), Institut de pharmacologie moléculaire et cellulaire (IPMC), Université Nice Sophia Antipolis (1965 - 2019) (UNS), COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-Centre National de la Recherche Scientifique (CNRS)-Université Côte d'Azur (UCA)-Université Nice Sophia Antipolis (1965 - 2019) (UNS), COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-Centre National de la Recherche Scientifique (CNRS)-Université Côte d'Azur (UCA), Centre hospitalier universitaire de Nantes (CHU Nantes), AP-HP - Hôpital Bichat - Claude Bernard [Paris], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP), Garnier, Sophie, and Loussouarn, Gildas
- Subjects
genetic variant ,ERG1 Potassium Channel ,congenital, hereditary, and neonatal diseases and abnormalities ,Medicine (General) ,Gibson assembly ,In silico ,[SDV]Life Sciences [q-bio] ,hERG ,Medicine (miscellaneous) ,Action Potentials ,Computational biology ,diagnostic testing ,translational medicine ,R5-920 ,[SDV.MHEP.CSC]Life Sciences [q-bio]/Human health and pathology/Cardiology and cardiovascular system ,Transcriptional Regulator ERG ,Humans ,pathogenicity ,Research Articles ,biology ,Virulence ,Genetic variants ,Single parameter ,Arrhythmias, Cardiac ,Pathogenicity ,Patient counselling ,[SDV.MHEP.CSC] Life Sciences [q-bio]/Human health and pathology/Cardiology and cardiovascular system ,[SDV] Life Sciences [q-bio] ,Long QT Syndrome ,Clinical diagnosis ,hERG ion channel ,biology.protein ,Molecular Medicine ,QT syndrome ,arrhythmias ,Research Article - Abstract
Background and aims Mutations in KCNH2 cause long or short QT syndromes (LQTS or SQTS) predisposing to life‐threatening arrhythmias. Over 1000 hERG variants have been described by clinicians, but most remain to be characterised. The objective is to standardise and accelerate the phenotyping process to contribute to clinician diagnosis and patient counselling. In silico evaluation was also included to characterise the structural impact of the variants. Methods We selected 11 variants from known LQTS patients and two variants for which diagnosis was problematic. Using the Gibson assembly strategy, we efficiently introduced mutations in hERG cDNA despite GC‐rich sequences. A pH‐sensitive fluorescent tag was fused to hERG for efficient evaluation of channel trafficking. An optimised 35‐s patch‐clamp protocol was developed to evaluate hERG channel activity in transfected cells. R software was used to speed up analyses. Results In the present work, we observed a good correlation between cell surface expression, assessed by the pH‐sensitive tag, and current densities. Also, we showed that the new biophysical protocol allows a significant gain of time in recording ion channel properties and provides extensive information on WT and variant channel biophysical parameters, that can all be recapitulated in a single parameter defined herein as the repolarisation power. The impacts of the variants on channel structure were also reported where structural information was available. These three readouts (trafficking, repolarisation power and structural impact) define three pathogenicity indexes that may help clinical diagnosis. Conclusions Fast‐track characterisation of KCNH2 genetic variants shows its relevance to discriminate mutants that affect hERG channel activity from variants with undetectable effects. It also helped the diagnosis of two new variants. This information is meant to fill a patient database, as a basis for personalised medicine. The next steps will be to further accelerate the process using an automated patch‐clamp system., We developed a method for rapid analysis of the properties of new hERG channel variants that is helpful for clinicians to diagnose an arrhythmia and help counseling the patient. This method is rapid enough to provide a complete set of channel properties and will set the stage for a more extensive characterization in the future.
- Published
- 2021
- Full Text
- View/download PDF
17. Editorial: Molecular Mechanisms of Voltage-Gating in Ion Channels
- Author
-
Mounir Tarek, 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), 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
[SDV]Life Sciences [q-bio] ,hERG ,patch - clamp technique ,Gating ,RM1-950 ,patch-clamp technique ,03 medical and health sciences ,0302 clinical medicine ,Pharmacology (medical) ,Patch clamp ,Ion channel ,030304 developmental biology ,Pharmacology ,Membrane potential ,0303 health sciences ,biology ,Voltage-gated ion channel ,Chemistry ,toxins ,Depolarization ,Hyperpolarization (biology) ,biology.protein ,Biophysics ,cryo-EM ,VSD gate coupling ,Therapeutics. Pharmacology ,030217 neurology & neurosurgery ,voltage-gated ion channel - Abstract
International audience; Editorial on the Research Topic Molecular Mechanisms of Voltage-Gating in Ion Channels Voltage-gated ion channels are transmembrane proteins conducting ions according to the electrochemical gradient, when opened by voltage. Hence, in these channels, at least one of the channel gates regulating the ion flux is controlled by the transmembrane potential. They are frequently ion specific and therefore selectively permeable to sodium (Na V channels), potassium (K V channels), calcium (Ca V channels) or chloride (CLC channels) ions. Depending on the channels, opening of the activation gate is triggered by membrane depolarization (e.g. K V , Na V and Ca V channels) or hyperpolarization (HCN channels for instance). In addition, in many voltage-gated channels, a so-called inactivation gate is also present. Compared to the activation gate, the latter is, when voltage-dependent, oppositely coupled to the potential: In K V , Na V and Ca V channels, upon membrane depolarization, the inactivation gate closes whereas the activation gate opens. Various voltage-dependent channels have been identified, depending on the excitable cell types in which they are expressed and their physiological role. They are characterized by their conductance, ion selectivity, pharmacology and voltage-sensitivity. These properties are mainly dictated by the amino-acid sequence and structure of the pore forming subunit(s), the presence of accessory subunit(s), the membrane composition and the intra-and extracellular ions concentrations. Many mutations have been identified in these channels, impacting their functions and provoking diseases named channelopathies. In 2012, we hosted a Research Topic on the Molecular Mechanisms of Voltage Dependency (Loussouarn and Tarek, 2012), bringing together scientists to collaborate and showcase the latest developments in the field. Since this Frontiers Research Topic, the development of new approaches, such as the use of cryo-electron microscopy (cryo-EM) at the atomic scale and the original approach of split channels, to name a few, has led to a more precise understanding of the mechanisms of voltage-gating, their targeting by toxins, and also their physio-pathological implications. Given the wealth of recent electrophysiological, biochemical, optical, and structural data regarding ion channel voltage-dependence, we felt there was clearly a need for putting together a new Research Topic that would include up to date Reviews and Original Research describing molecular details of the functioning of these complex voltage-gated channels. The review of Brewer et al. underlines how ion channel structures and models reveal critical differences in the atomic details of KCNQ1, hERG, and Na V 1.5 structures associated with their distinct voltage-gating and implication in Long QT syndrome, and also their pharmacological profiles. Such structural data may help defining the pathogenicity of the hundreds of variants in absence of functional data. It also mentions an important point: Molecular Dynamics represents a useful tool in refining cryo-EM structures, which are often of lower resolution in the periphery of protein core structures.
- Published
- 2021
- Full Text
- View/download PDF
18. Voltage-dependent activation in EAG channels follows a ligand-receptor rather than a mechanical-lever mechanism
- Author
-
Olfat A. Malak, Gildas Loussouarn, K.S. Kudryashova, Olga Sokolova, Anastasia V. Grizel, and G. S. Gluhov
- Subjects
Models, Molecular ,0301 basic medicine ,ERG1 Potassium Channel ,hERG ,Allosteric regulation ,Gating ,Biochemistry ,03 medical and health sciences ,Protein Domains ,PAS domain ,Chlorocebus aethiops ,Animals ,Humans ,Receptor ,Molecular Biology ,Ion channel ,030102 biochemistry & molecular biology ,biology ,Chemistry ,Cell Biology ,Ligand (biochemistry) ,Ether-A-Go-Go Potassium Channels ,Electrophysiology ,030104 developmental biology ,COS Cells ,biology.protein ,Biophysics ,Peptides ,Ion Channel Gating ,Molecular Biophysics - Abstract
Ether-a-go-go family (EAG) channels play a major role in many physiological processes in humans, including cardiac repolarization and cell proliferation. Cryo-EM structures of two of them, K(V)10.1 and human ether-a-go-go-related gene (hERG or K(V)11.1), have revealed an original nondomain-swapped structure, suggesting that the mechanism of voltage-dependent gating of these two channels is quite different from the classical mechanical-lever model. Molecular aspects of hERG voltage-gating have been extensively studied, indicating that the S4-S5 linker (S4-S5(L)) acts as a ligand binding to the S6 gate (S6 C-terminal part, S6(T)) and stabilizes it in a closed state. Moreover, the N-terminal extremity of the channel, called N-Cap, has been suggested to interact with S4-S5(L) to modulate channel voltage-dependent gating, as N-Cap deletion drastically accelerates hERG channel deactivation. In this study, using COS-7 cells, site-directed mutagenesis, electrophysiological measurements, and immunofluorescence confocal microscopy, we addressed whether these two major mechanisms of voltage-dependent gating are conserved in K(V)10.2 channels. Using cysteine bridges and S4-S5(L)–mimicking peptides, we show that the ligand/receptor model is conserved in K(V)10.2, suggesting that this model is a hallmark of EAG channels. Truncation of the N-Cap domain, Per-Arnt-Sim (PAS) domain, or both in K(V)10.2 abolished the current and altered channel trafficking to the membrane, unlike for the hERG channel in which N-Cap and PAS domain truncations mainly affected channel deactivation. Our results suggest that EAG channels function via a conserved ligand/receptor model of voltage gating, but that the N-Cap and PAS domains have different roles in these channels.
- Published
- 2019
- Full Text
- View/download PDF
19. Disruption of a Conservative Motif in the C-Terminal Loop of the KCNQ1 Channel Causes LQT Syndrome
- Author
-
Maria Karlova, Denis V. Abramochkin, Ksenia B. Pustovit, Tatiana Nesterova, Valery Novoseletsky, Gildas Loussouarn, Elena Zaklyazminskaya, and Olga S. Sokolova
- Subjects
Male ,Heterozygote ,POTASSIUM CHANNEL KCNQ1 ,GENETICS ,CASE REPORT ,KCNQ1 PROTEIN, HUMAN ,METABOLISM ,HETEROZYGOTE ,Catalysis ,Inorganic Chemistry ,biophysics ,Humans ,Point Mutation ,PATCH-CLAMP ,Physical and Theoretical Chemistry ,MUTATION ,Molecular Biology ,Spectroscopy ,Aged ,CHILD, PRESCHOOL ,KCNQ1 ,MALE ,Organic Chemistry ,HUMAN ,HUMANS ,General Medicine ,IKS ,INHERITED CHANNELOPATHY ,POINT MUTATION ,Computer Science Applications ,KV7.1 ,Long QT Syndrome ,PRESCHOOL CHILD ,LONG QT SYNDROME ,Child, Preschool ,Kv7.1 ,IKs ,patch-clamp ,inherited channelopathy ,LQTS ,KCNQ1 Potassium Channel ,Mutation ,KCNQ1 POTASSIUM CHANNEL ,AGED - Abstract
We identified a single nucleotide variation (SNV) (c.1264A > G) in the KCNQ1 gene in a 5-year-old boy who presented with a prolonged QT interval. His elder brother and mother, but not sister and father, also had this mutation. This missense mutation leads to a p.Lys422Glu (K422E) substitution in the Kv7.1 protein that has never been mentioned before. We inserted this substitution in an expression plasmid containing Kv7.1 cDNA and studied the electrophysiological characteristics of the mutated channel expressed in CHO-K1, using the whole-cell configuration of the patch-clamp technique. Expression of the mutant Kv7.1 channel in both homo- and heterozygous conditions in the presence of auxiliary subunit KCNE1 results in a significant decrease in tail current densities compared to the expression of wild-type (WT) Kv7.1 and KCNE1. This study also indicates that K422E point mutation causes a dominant negative effect. The mutation was not associated with a trafficking defect; the mutant channel protein was confirmed to localize at the cell membrane. This mutation disrupts the poly-Lys strip in the proximal part of the highly conserved cytoplasmic A–B linker of Kv7.1 that was not shown before to be crucial for channel functioning. © 2022 by the authors. Russian Science Foundation, RSF: 22-14-00088; International Association for the Evaluation of Educational Achievement, IEA: 2773 This study was funded by Russian Science Foundation (22-14-00088 to O.S.S.). Authors thank Lisa Trifonova for proof-reading the manuscript and Lisha Mai for help with . G.L. would like to acknowledge support from the CNRS International Emerging Action (IEA) PRC RUSSIE 2019 (PRC no. 2773). O.S.S. and V.N. acknowledge the support from the Interdisciplinary Scientific and Educational School of Moscow Lomonosov University «Molecular Technologies of the Living Systems and Synthetic Biology». was created with the help of Biorender.com.
- Published
- 2022
- Full Text
- View/download PDF
20. A long QT mutation substitutes cholesterol for phosphatidylinositol-4,5-bisphosphate in KCNQ1 channel regulation.
- Author
-
Fabien C Coyan, Fayal Abderemane-Ali, Mohamed Yassine Amarouch, Julien Piron, Jérôme Mordel, Céline S Nicolas, Marja Steenman, Jean Mérot, Céline Marionneau, Annick Thomas, Robert Brasseur, Isabelle Baró, and Gildas Loussouarn
- Subjects
Medicine ,Science - Abstract
INTRODUCTION:Phosphatidylinositol-4,5-bisphosphate (PIP2) is a cofactor necessary for the activity of KCNQ1 channels. Some Long QT mutations of KCNQ1, including R243H, R539W and R555C have been shown to decrease KCNQ1 interaction with PIP2. A previous study suggested that R539W is paradoxically less sensitive to intracellular magnesium inhibition than the WT channel, despite a decreased interaction with PIP2. In the present study, we confirm this peculiar behavior of R539W and suggest a molecular mechanism underlying it. METHODS AND RESULTS:COS-7 cells were transfected with WT or mutated KCNE1-KCNQ1 channel, and patch-clamp recordings were performed in giant-patch, permeabilized-patch or ruptured-patch configuration. Similar to other channels with a decreased PIP2 affinity, we observed that the R243H and R555C mutations lead to an accelerated current rundown when membrane PIP2 levels are decreasing. As opposed to R243H and R555C mutants, R539W is not more but rather less sensitive to PIP2 decrease than the WT channel. A molecular model of a fragment of the KCNQ1 C-terminus and the membrane bilayer suggested that a potential novel interaction of R539W with cholesterol stabilizes the channel opening and hence prevents rundown upon PIP2 depletion. We then carried out the same rundown experiments under cholesterol depletion and observed an accelerated R539W rundown that is consistent with this model. CONCLUSIONS:We show for the first time that a mutation may shift the channel interaction with PIP2 to a preference for cholesterol. This de novo interaction wanes the sensitivity to PIP2 variations, showing that a mutated channel with a decreased affinity to PIP2 could paradoxically present a slowed current rundown compared to the WT channel. This suggests that caution is required when using measurements of current rundown as an indicator to compare WT and mutant channel PIP2 sensitivity.
- Published
- 2014
- Full Text
- View/download PDF
21. 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
22. Computer modeling of whole-cell voltage-clamp analyses to delineate guidelines for good practice of manual and automated patch-clamp
- Author
-
Maxime Lorenzini, Michel De Waard, Adrien Lesage, Céline Marionneau, Eléonore Moreau, Jérôme Montnach, Isabelle Baró, Isabelle Simon, Gildas Loussouarn, Sébastien Nicolas, 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), Groupe de Reflexion sur la Recherche Cardiovasculaire-Societe Francaise de Cardiologie predoctoral fellowshipSFC/GRRC2018Federation Francaise de CardiologieLeducq Foundation'New Team' of the Region Pays de la LoireEuropean FEDER grant, ANR-15-CE14-0006,PhosphoNav,Phosphorylation des Canaux Nav1.5 et Régulation de l'Excitabilité Cardiaque Normale et Pathologique(2015), ANR-16-CE92-0013,Progress DHF,Mécanismes de la progression de la dysfonction diastolique vers l'insuffisance cardiaque(2016), ANR-11-LABX-0015,ICST,Canaux ioniques d'intérêt thérapeutique(2011), ANR-18-CE19-0024,OptChemCom,Technologies intégrées d' optique, photochimie et informatique pour étudier la synergie physiologique des canaux ioniques(2018), 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
Patch-Clamp Techniques ,Computer science ,Science ,[SDV]Life Sciences [q-bio] ,Voltage clamp ,Biophysics ,Models, Biological ,Ion Channels ,Article ,Membrane Potentials ,Mice ,Computational biophysics ,03 medical and health sciences ,0302 clinical medicine ,[SDV.MHEP.CSC]Life Sciences [q-bio]/Human health and pathology/Cardiology and cardiovascular system ,Automated patch clamp ,Electronic engineering ,Animals ,Myocytes, Cardiac ,Throughput (business) ,Cells, Cultured ,ComputingMilieux_MISCELLANEOUS ,Ion channel ,030304 developmental biology ,0303 health sciences ,Sodium channel ,Ion current ,Cardiovascular biology ,Potassium channel ,Amplitude ,Medicine ,Permeation and transport ,[SDV.NEU]Life Sciences [q-bio]/Neurons and Cognition [q-bio.NC] ,030217 neurology & neurosurgery - Abstract
The patch-clamp technique and more recently the high throughput patch-clamp technique have contributed to major advances in the characterization of ion channels. However, the whole-cell voltage-clamp technique presents certain limits that need to be considered for robust data generation. One major caveat is that increasing current amplitude profoundly impacts the accuracy of the biophysical analyses of macroscopic ion currents under study. Using mathematical kinetic models of a cardiac voltage-gated sodium channel and a cardiac voltage-gated potassium channel, we demonstrated how large current amplitude and series resistance artefacts induce an undetected alteration in the actual membrane potential and affect the characterization of voltage-dependent activation and inactivation processes. We also computed how dose–response curves are hindered by high current amplitudes. This is of high interest since stable cell lines frequently demonstrating high current amplitudes are used for safety pharmacology using the high throughput patch-clamp technique. It is therefore critical to set experimental limits for current amplitude recordings to prevent inaccuracy in the characterization of channel properties or drug activity, such limits being different from one channel type to another. Based on the predictions generated by the kinetic models, we draw simple guidelines for good practice of whole-cell voltage-clamp recordings.
- Published
- 2020
- Full Text
- View/download PDF
23. 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
24. 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
25. 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
26. Fast Track hERG phenotyping to evaluate the pathogenicity of KCNH2 genetic variants
- Author
-
Florian Lesage, Jean-Jacques Schott, Isabelle Denjoy, M. Menard, Vincent Probst, S. Nicolas, M. De Waard, Sylvain Feliciangeli, Jérôme Montnach, Isabelle Baró, B.B. Ribeiro de Oliveira Mendes, F. Kyndt, Franck C. Chatelain, Gildas Loussouarn, and B. Ollivier
- Subjects
congenital, hereditary, and neonatal diseases and abnormalities ,Gibson assembly ,biology ,business.industry ,In silico ,hERG ,Computational biology ,Pathogenicity ,Phenotype ,biology.protein ,Medicine ,Repolarization ,cardiovascular diseases ,Personalized medicine ,Cardiology and Cardiovascular Medicine ,business ,Fluorescent tag - Abstract
Introduction Mutations in KCNH2 (coding for hERG) cause long or short QT syndromes (LQTS or SQTS), predisposing to life-threatening arrhythmias. More than 1000 variations in hERG sequences have been described, most of them are to be characterized. Objective The objective is to standardize and accelerate the entire process necessary to phenotype hERG variants. An in silico evaluation was also included to characterize the structural impact of the variants. Methods We selected 12 variants from patients with LQTS, and 1 with SQTS. We optimized the protocol to efficiently introduce mutations in hERG cDNA despite GC-rich sequences, using the Gibson assembly strategy. A pH-sensitive fluorescent tag was fused to hERG for fast-track evaluation of hERG cell trafficking. An optimized patch-clamp protocol of 35 sec was developed to evaluate hERG channel activity in transfected cells. R software was used to speed up analyses. Results In the present work, we observed a good correlation between cell surface expression, assessed by the pH-sensitive tag, and current densities. Also, we showed that the new biophysical protocol allows a significant gain of time in recording ion channel properties and provides extensive information on WT and variant channel biophycical parameters, that can all be recapitulated in a single parameter defined herein as the repolarization power. The impact of the variants on channel structure were also reported where structural information was available. These three readouts (trafficking, repolarization power and structural impact) define 3 pathogenicity indexes that may help clinical diagnosis. Conclusion Fast track characterization of KCNH2 genetic variants shows its relevance to discriminate mutants that affect hERG channel activity from variants with undetectable effects. This information is meant to fill a patient database, as a basis for personalized medicine. The next steps will be to further accelerate the process using an automated patch-clamp system.
- Published
- 2021
- Full Text
- View/download PDF
27. Detergent-free solubilization of human Kv channels expressed in mammalian cells
- Author
-
Denis V. Abramochkin, Olga Sokolova, Konstantin V. Shaitan, Gildas Loussouarn, Heinz-Jürgen Steinhoff, A. Mulkidzhanyan, G. S. Gluhov, Olfat A. Malak, Natalia Voskoboynikova, M. G. Karlova, 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
Patch-Clamp Techniques ,Detergents ,CHO Cells ,02 engineering and technology ,[SDV.BC]Life Sciences [q-bio]/Cellular Biology ,Biochemistry ,law.invention ,03 medical and health sciences ,Cricetulus ,Affinity chromatography ,Dynamic light scattering ,law ,Cricetinae ,Chlorocebus aethiops ,Animals ,Humans ,Lipid bilayer ,Molecular Biology ,030304 developmental biology ,0303 health sciences ,Chemistry ,Organic Chemistry ,Cell Biology ,021001 nanoscience & nanotechnology ,SMA ,Negative stain ,Recombinant Proteins ,Transmembrane protein ,Microscopy, Electron ,Protein Subunits ,Solubility ,Membrane protein ,Potassium Channels, Voltage-Gated ,COS Cells ,Recombinant DNA ,Biophysics ,Polystyrenes ,0210 nano-technology - Abstract
International audience; Styrene-maleic acid (SMA) copolymers are used to extract lipid-encased membrane proteins from lipid bilayers in a detergent-free manner, yielding SMA lipid particles (SMALPs). SMALPs can serve as stable water-soluble nanocontainers for structural and functional studies of membrane proteins. Here, we used SMA copolymers to study full-length pore-forming α-subunits hKCNH5 and hKCNQ1 of human neuronal and cardiac voltage-gated potassium (Kv) channels, as well as the fusion construct comprising of an α-subunit hKCNQ1 and its regulatory transmembrane KCNE1 β-subunit (hKCNE1-hKCNQ1) with added affinity tags, expressed in mammalian COS-1 cells. All these recombinant proteins were shown to be functionally active. Treatment with the SMA copolymer, followed by purification on the affinity column, enabled extraction of all three channels. A DLS experiment demonstrated that Negative stain electron microscopy and single particle image analysis revealed a four-fold symmetry within channel-containing SMALPs, which indicates that purified hKCNH5 and hKCNQ1 channels, as well as the hKCNE1-hKCNQ1 fusion construct, retained their structural integrity as tetramers.
- Published
- 2019
- Full Text
- View/download PDF
28. 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
29. Optical control of hERG channel activity using a photosensitive Bekm-1 blocker
- Author
-
Gildas Loussouarn, Sébastien Nicolas, Virginie Forest, Jérôme Montnach, C. Jopling, Nathalie Gaborit, M. De Waard, E. Correia, B.B. Ribeiro De Oliveira Mendes, S. De Waard, and Patricia Lemarchand
- Subjects
congenital, hereditary, and neonatal diseases and abnormalities ,biology ,business.industry ,hERG ,Highly selective ,Cardiac repolarization ,Optical control ,biology.protein ,Biophysics ,Medicine ,Repolarization ,Stable cell line ,cardiovascular diseases ,Cardiology and Cardiovascular Medicine ,business - Abstract
Introduction Kv11.1 channel (hERG) and the corresponding IKr current is a major component of cardiac repolarization and sinoatrial pacemaking. Dysfunctions in hERG channels are associated with cardiac arrhythmias and remains a driving force to study the structure, biophysics and pharmacology of this channel. Several compounds that block hERG channels have been developed. In this study, we focused on BeKm-1, a toxin which is highly selective for the hERG channel. Objective To study hERG channels at high temporal and spatial resolution, we developed a photo-protected analog of BeKm-1 that enables control of hERG using light. Method We developed a photo-protected BeKm-1 analog (BeKm-1-Lys18Nvoc) which is cleaved by UV-light at 365 nm. Using a hERG stable cell line and high-throughput automated patch-clamp (Syncropatch 384, Nanion) we validated several properties of the BeKm-1 analog blockage. To demonstrate spatial control of cardiomyocyte function, hiPS-derived cardiomyocytes were plated on CardioExcyte96 to record extrafield potentials. Results We first validated physico-chemical properties of BeKm-1-Nvoc compared to BeKm-1 using HPLC analyses. Patch-clamp experiments displayed no inhibition of hERG current by BeKm-1-Nvoc at concentrations for which wild-type Bekm-1 is maximally inhibiting. Illumination of BeKm-1-Nvoc (100 nM) at 365 nm allows cleavage of the protecting group and strong inhibition of hERG current. The block of hERG is reversible after washes. Illumination of BeKm-1-Nvoc on hiPS-derived cardiomyocytes induces prolongation of extrafied potential which reflects an increase in repolarization time. Study of effects of photoactivation of BeKm-1 on mouse and zebrafish heart rates are ongoing. Conclusion We developed for the first time a photo-sensitive blocker of endogenous hERG channel which allows control of its activity with the spatiotemporal precision of light. Studies of implication of hERG on arrhythmias using this compound are ongoing.
- Published
- 2020
- Full Text
- View/download PDF
30. Functional Characterization of KCNH2 genetic variants, encoding hERG potassium channel, as a clinically-relevant information for type 2 LQTS syndrome
- Author
-
Sylvain Feliciangeli, Vincent Probst, B.B. Ribeiro de Oliveira Mendes, Franck C. Chatelain, B. Ollivier, Jérôme Montnach, F. Kyndt, S. Nicolas, Florian Lesage, Gildas Loussouarn, Isabelle Denjoy, and M. De Waard
- Subjects
congenital, hereditary, and neonatal diseases and abnormalities ,biology ,business.industry ,High-throughput screening ,Electroporation ,hERG ,Transfection ,Computational biology ,Potassium channel ,Automated patch clamp ,FuGENE ,biology.protein ,Medicine ,cardiovascular diseases ,Patch clamp ,Cardiology and Cardiovascular Medicine ,business - Abstract
Introduction hERG potassium channels are essential for normal cardiac electrical activity. Mutations in the hERG gene (KCNH2) cause long QT syndrome, a disorder that predisposes individuals to life-threatening arrhythmias. More than 1000 mutations in hERG sequence are described in databases and most of them remains to be characterized. Objective The objective of the work is to determine the consequences of the variants identified in LQT2 patients on the expression and function of the hERG channel. Methods An optimized sequence hERG fused to pHluorin tag was synthesized. By amplifying mutated overlapping fragments, and using the Gibson assembly strategy, we constructed hERG-pHluorin plasmids with missense variations. CHO cells were transfected by electroporation with the Maxcyte system or Fugene method, and studied after 40 hours. Confocal images and flow cytometry analysis were used to evaluate the percentage of transfected cells and to quantify the membrane channel expression, through pH sensitive pHluorin tag. Conventional or automated patch clamp were used to evaluate the hERG activity. Results In the present work, we constructed hERG plasmids carrying 48 variations identified in patients in France. Higher efficiency of transfection was observed with electroporation as compared to the Fugene method. The activity of hERG-pHluorin was validated in manual patch-clamp, the pHluorin tag does not interfere with channel activity. Characterization of several mutations in conventional patch clamp showed difference in current amplitude. Automated patch-clamp data was compared to manual patch-clamp, as an initial validation for high throughput screening of all constructed mutations. Conclusion High-throughput characterization of KCNH2 genetic variants is relevant to discriminate mutants that affect hERG channel activity from variants with undetectable effects. This information will be indicated in a patient database, as a basis of personalized medicine.
- Published
- 2020
- Full Text
- View/download PDF
31. 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
32. Studying Kv Channels Function using Computational Methods
- Author
-
Audrey, Deyawe, Marina A, Kasimova, Lucie, Delemotte, Gildas, Loussouarn, Mounir, Tarek, Structure et Réactivité des Systèmes Moléculaires Complexes (SRSMC), Université de Lorraine (UL)-Centre National de la Recherche Scientifique (CNRS)-Institut de Chimie du CNRS (INC), 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
Models, Molecular ,Protein Stability ,Molecular dynamics simulations ,[SDV]Life Sciences [q-bio] ,Gating charge ,Computational Biology ,Homology modeling ,Lipid membranes ,Molecular Dynamics Simulation ,Crystallography, X-Ray ,Potassium Channels, Voltage-Gated ,Structural Homology, Protein ,Transmembrane potential ,Voltage-gated ion channels - Abstract
International audience; In recent years, molecular modeling techniques, combined with MD simulations, provided significant insights on voltage-gated (Kv) potassium channels intrinsic properties. Among the success stories are the highlight of molecular level details of the effects of mutations, the unraveling of several metastable intermediate states, and the influence of a particular lipid, PIP2, in the stability and the modulation of Kv channel function. These computational studies offered a detailed view that could not have been reached through experimental studies alone. With the increase of cross disciplinary studies, numerous experiments provided validation of these computational results, which endows an increase in the reliability of molecular modeling for the study of Kv channels. This chapter offers a description of the main techniques used to model Kv channels at the atomistic level.
- Published
- 2018
- Full Text
- View/download PDF
33. Building Atomic Models of the Ion Channels Based on Low Resolution Electron Microscopy Maps and Homology Modeling
- Author
-
Valery, Novoseletsky, Olfat A, Malak, Gildas, Loussouarn, Olga S, Sokolova, 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
Models, Molecular ,Microscopy, Electron ,Shab Potassium Channels ,Protein Conformation ,Structural Homology, Protein ,Single-particle electron microscopy ,[SDV]Life Sciences [q-bio] ,Kv2.1 ion channel ,Modeling ,Computational Biology ,Structure ,Homology - Abstract
International audience; Voltage-gated potassium channels play pivotal roles in excitable and non-excitable cells. For many decades, structural properties and molecular mechanisms of these channels were inferred from functional observations. At the turn of the twenty-first century, structural biology revealed major aspects in the structural basis of ion channel organization, permeation, and gating. Among the available tools, homology modeling associated with low resolution microscopy helps in delineating the different structural elements of voltage-gated channels. Here, we describe in detail the methodology of homology modeling, using the 3D structure of the Kv2.1ΔCTA ion channel as a reference.
- Published
- 2018
- Full Text
- View/download PDF
34. Marine n-3 PUFAs modulate I-Ks gating, channel expression, and location in membrane microdomains
- Author
-
Carlotta Ronchi, Tomáš Starý, Carmen Valenzuela, Isabelle Baró, Alicia de la Cruz, Marcella Rocchetti, Sanjay Kharche, Antonio Zaza, Gildas Loussouarn, Stefano Severi, Cristina Moreno, Miriam Guizy, Antonio Felipe, Anna Oliveras, Núria Comes, Moreno, C, De La Cruz, A, Oliveras, A, Kharche, S, Guizy, M, Comes, N, Starý, T, Ronchi, C, Rocchetti, M, Baró, I, Loussouarn, G, Zaza, A, Severi, S, Felipe, A, Valenzuela, C, Consejo Superior de Investigaciones Científicas (España), Ministerio de Economía y Competitividad (España), Instituto de Salud Carlos III, Bedrijfsbureau CD, Cardiologie, RS: CARIM - R2 - Cardiac function and failure, Departament de Bioquímica i Biología Molecular, Universitat de Barcelona. Avda Diagonal 645, 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), Instituto de Investigaciones Biomedicas, Universidad Nacional Autónoma de México (UNAM), Moreno, Cristina, De La Cruz, Alicia, Oliveras, Anna, Kharche, Sanjay R., Guizy, Miriam, Comes, Nùria, Starý, Tomáš, Ronchi, Carlotta, Rocchetti, Marcella, Baró, Isabelle, Loussouarn, Gilda, Zaza, Antonio, Severi, Stefano, Felipe, Antonio, and Valenzuela, Carmen
- Subjects
Physiology ,[SDV]Life Sciences [q-bio] ,Action Potentials ,Cercopithecus aethiop ,Gating ,Pharmacology ,chemistry.chemical_compound ,Chlorocebus aethiops ,Myocytes, Cardiac ,Membrane Microdomain ,Lipid raft ,chemistry.chemical_classification ,Docosahexaenoic Acid ,Kv7.1 ,Medicine (all) ,Eicosapentaenoic acid ,3. Good health ,DHA ,Eicosapentaenoic Acid ,Docosahexaenoic acid ,Anti-Arrhythmia Agent ,Potassium Channels, Voltage-Gated ,COS Cells ,cardiovascular system ,Fatty Acids, Unsaturated ,KCNE1 ,lipids (amino acids, peptides, and proteins) ,Cardiology and Cardiovascular Medicine ,Anti-Arrhythmia Agents ,Ion Channel Gating ,Polyunsaturated fatty acid ,Human ,Docosahexaenoic Acids ,Biology ,Ventricular action potential ,Membrane Microdomains ,COS Cell ,Physiology (medical) ,parasitic diseases ,Animals ,Humans ,Action Potential ,Lipid rafts ,Cholesterol ,Animal ,I-Ks ,EPA ,IK ,Electrophysiology ,chemistry ,PUFAs ,K(v)7.1 ,PUFA - Abstract
et al., [Aims]: Polyunsaturated fatty n-3 acids (PUFAs) have been reported to exhibit antiarrhythmic properties. However, the mechanisms of action remain unclear. We studied the electrophysiological effects of eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) on IKs, and on the expression and location of Kv7.1 and KCNE1. [Methods and results]: Experiments were performed using patch-clamp, western blot, and sucrose gradient techniques in COS7 cells transfected with Kv7.1/KCNE1 channels. Acute perfusion with both PUFAs increased Kv7.1/KCNE1 current, this effect being greater for DHA than for EPA. Similar results were found in guinea pig cardiomyocytes. Acute perfusion of either PUFA slowed the activation kinetics and EPA shifted the activation curve to the left. Conversely, chronic EPA did not modify Kv7.1/KCNE1 current magnitude and shifted the activation curve to the right. Chronic PUFAs decreased the expression of Kv7.1, but not of KCNE1, and induced spatial redistribution of Kv7.1 over the cell membrane. Cholesterol depletion with methyl-β-cyclodextrin increased Kv7.1/KCNE1 current magnitude. Under these conditions, acute EPA produced similar effects than those induced in non-cholesterol-depleted cells. A ventricular action potential computational model suggested antiarrhythmic efficacy of acute PUFA application under IKr block. [Conclusions]: We provide evidence that acute application of PUFAs increases Kv7.1/KCNE1 through a probably direct effect, and shows antiarrhythmic efficacy under IKr block. Conversely, chronic EPA application modifies the channel activity through a change in the Kv7.1/KCNE1 voltage-dependence, correlated with a redistribution of Kv7.1 over the cell membrane. This loss of function may be pro-arrhythmic. This shed light on the controversial effects of PUFAs regarding arrhythmias., This work was supported by grants from CICYT (SAF2010-14916 and SAF2013-45800-R to C.V.; BFU2011-23268 and CSD2008-00005 to A.F.) and FIS (PI11/02459, RD06/0014/0006, and RD12/0042/0019 to C.V.). C.M. and M.G. hold FPI grants. N.C. and A.d.l.C. hold Juan de la Cierva and RIC contracts, respectively.
- Published
- 2015
- Full Text
- View/download PDF
35. 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
36. Phosphatidylinositol (4,5)-bisphosphate-mediated pathophysiological effect of HIV-1 Tat protein
- Author
-
Bruno Beaumelle, Petra Tóth, Nicolas Vitale, Christophe Chopard, Gildas Loussouarn, Olfat A. Malak, Centre d’études d’Agents Pathogènes et Biotechologies pour la Santé (CPBS), Université de Montpellier (UM)-Centre National de la Recherche Scientifique (CNRS), Institut de Recherche en Infectiologie de Montpellier (IRIM), Équipe 'Rythme, vie et mort de la rétine', Institut des Neurosciences Cellulaires et Intégratives (INCI), Université de Strasbourg (UNISTRA)-Centre National de la Recherche Scientifique (CNRS)-Université de Strasbourg (UNISTRA)-Centre National de la Recherche Scientifique (CNRS), 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), Département Neurotransmission et sécrétion neuroendocrine, 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)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
0301 basic medicine ,Phosphatidylinositol 4,5-Diphosphate ,Cell type ,5)P(2) ,Endosome ,[SDV.NEU.NB]Life Sciences [q-bio]/Neurons and Cognition [q-bio.NC]/Neurobiology ,5)P 2 ,Endosomes ,[SDV.BC]Life Sciences [q-bio]/Cellular Biology ,Biology ,Biochemistry ,Synaptic vesicle ,Exocytosis ,Potassium channels ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Phagocytosis ,[SDV.MHEP.CSC]Life Sciences [q-bio]/Human health and pathology/Cardiology and cardiovascular system ,Humans ,Phosphatidylinositol ,HIV-1 Tat ,PtdIns(4 ,ComputingMilieux_MISCELLANEOUS ,Neurosecretion ,General Medicine ,3. Good health ,Cell biology ,Potassium channel activity ,Cytosol ,030104 developmental biology ,Phosphatidylinositol 4,5-bisphosphate ,chemistry ,[SDV.MP.VIR]Life Sciences [q-bio]/Microbiology and Parasitology/Virology ,HIV-1 ,tat Gene Products, Human Immunodeficiency Virus ,030217 neurology & neurosurgery - Abstract
International audience; Human immunodeficiency virus (HIV)-infected cells actively release the transcriptional activator (Tat) viral protein that is required for efficient HIV gene transcription. Extracellular Tat is able to enter uninfected cells. We recently reported that internalized Tat escapes endosomes to reach the cytosol and is then recruited to the plasma membrane by phosphatidylinositol 4,5-bisphosphate (PtdIns(4,5)P2). As a consequence, Tat strongly impairs different critical cellular functions in several cell types. Here we will review recent evidences showing that Tat, by affecting the interaction of key cellular effectors with PtdIns(4,5)P2, blocks exocytosis from neuroendocrine cells, perturbs the synaptic vesicle exo-endocytosis cycle, prevents efficient phagocytosis by macrophages, and alters potassium channel activity in cardiac cells. Potential mechanistic aspects of Tat effects on these cellular processes will be discussed.
- Published
- 2017
- Full Text
- View/download PDF
37. 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
38. A novel method for measurement of submembrane ATP concentration
- Author
-
Fiona M. Gribble, Chao Zhao, Frances M. Ashcroft, Stephen J. Tucker, Gildas Loussouarn, and Colin G. Nichols
- Subjects
Cytoplasm ,Molar concentration ,Patch-Clamp Techniques ,Potassium Channels ,Xenopus ,Biosensing Techniques ,Biochemistry ,chemistry.chemical_compound ,Adenosine Triphosphate ,Animals ,Patch clamp ,Potassium Channels, Inwardly Rectifying ,Molecular Biology ,Sequence Deletion ,biology ,Chemistry ,Cell Membrane ,Electric Conductivity ,Cell Biology ,biology.organism_classification ,Potassium channel ,Cell Compartmentation ,Cytosol ,Membrane ,Mutation ,Biophysics ,Oocytes ,Adenosine triphosphate - Abstract
There has been considerable debate as to whether adenosine triphosphate (ATP) is compartmentalized within cells and, in particular, whether the ATP concentration directly beneath the plasma membrane, experienced by membrane proteins, is the same as that of the bulk cytoplasm. This issue has been difficult to address because there is no indicator of cytosolic ATP, such as those available for Ca(2+), capable of resolving the submembrane ATP concentration ([ATP](sm)) in real time within a single cell. We show here that mutant ATP-sensitive K(+) channels can be used to measure [ATP](sm) by comparing the increase in current amplitude on patch excision with the ATP dose-response curve. In Xenopus oocytes, [ATP](sm) was 4.6 +/- 0.3 mm (n = 29) under resting conditions, slightly higher than that measured for the bulk cytoplasm (2.3 mm). In mammalian (COSm6) cells, [ATP](sm) was slightly lower and averaged 1.4 +/- 0.1 mm (n = 66). Metabolic poisoning (10 min of 3 mm azide) produced a significant fall in [ATP](sm) in both types of cells: to 1.2 +/- 0.1 mm (n = 24) in oocytes and 0.8 +/- 0.11 mm for COSm6 cells. We conclude that [ATP](sm) lies in the low millimolar range and that there is no gradient between bulk cytosolic and submembrane [ATP].
- Published
- 2016
39. 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
40. hERG S4-S5 acts as a Voltage-Dependent Ligand Binding the Activation Gate and Locking it in a Closed State
- Author
-
Zeineb Es-Salah-Lamoureux, Gildas Loussouarn, and Olfat A. Malak
- Subjects
chemistry.chemical_classification ,biology ,Stereochemistry ,hERG ,Biophysics ,Peptide ,Endogeny ,Transfection ,Gating ,Ligand (biochemistry) ,chemistry ,Covalent bond ,biology.protein ,Cysteine - Abstract
Kv (voltage-gated potassium) channels are formed by a tetrameric pore (S5–S6) surrounded by four voltage sensor domains (S1–S4). Molecular mechanisms underlying gating remain poorly understood.Previously, we showed that peptides mimicking S4-S5L inhibit a cardiac Kv channel, KCNQ1 (Kv7.1). On the other hand, peptides mimicking S6T upregulate the channel activity by competing with the endogenous S6T for the endogenous S4-S5L (Choveau et al, 2010). Those observations suggested that S4-S5L acts as a voltage controled ligand that binds S6T and locks the channel in a closed state. However, given the low affinity between the peptide and the KCNQ1 channel, effects were moderate. In the present study, we asked whether covalently locking s4s5l onto s6t would lead to a permanent closure of the channel. To this end, we selected the human ether-a-go-go-related gene (hERG or Kv11.1) channel as a model since in this channel, a couple of cysteine in S4-S5L and S6T was introduced and led to channel closure (Ferrer et al, 2006). We used the whole-cell configuration of the patch-clamp technique in transfected COS-7 cells. We observed a complete inhibition of hERG in oxidative condition when coexpressing the S4-S5L peptide carrying an introduced cysteine (S4S5L-Cys-peptide) and the channel with a cysteine introduced in S6T (S6T-Cys-channel). Reciprocally, coexpressing S6TCyspeptide and the S4-S5L-Cys-channel led to a virtually voltage independent channel in oxidative condition. These results: 1) validate a model of gating in which S4-S5 acts as a voltage-dependent ligand binding the activation gate and locking it in a closed state and 2) suggest a generalization of this model.Choveau et al, J Biol Chem. 2011 286:707-16Ferrer et al, J Biol Chem. 2006 281:12858-64
- Published
- 2016
- Full Text
- View/download PDF
41. 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
42. 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
43. Delayed rectifier K+ currents and cardiac repolarization
- Author
-
Jean Mérot, Gildas Loussouarn, Isabelle Baró, and Flavien Charpentier
- Subjects
ERG1 Potassium Channel ,biology ,Chemistry ,Myocardium ,hERG ,Pharmacology ,Cardiac repolarization ,K currents ,Ether-A-Go-Go Potassium Channels ,Potassium channel ,Delayed rectifier ,Potassium Channels, Voltage-Gated ,KCNQ1 Potassium Channel ,Potassium ,biology.protein ,Humans ,Repolarization ,Action potential duration ,Cardiology and Cardiovascular Medicine ,Molecular Biology ,Neuroscience - Abstract
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.
- Published
- 2010
- Full Text
- View/download PDF
44. LQT1-associated Mutations Increase KCNQ1 Proteasomal Degradation Independently of Derlin-1
- Author
-
Isabelle Baró, Gildas Loussouarn, Shehrazade Dahimène, David Peroz, and Jean Mérot
- Subjects
Proteasome Endopeptidase Complex ,Small interfering RNA ,endocrine system diseases ,Romano-Ward Syndrome ,Mutant ,Mutation, Missense ,Biology ,Endoplasmic Reticulum ,medicine.disease_cause ,Biochemistry ,Cell Line ,Western blot ,medicine ,Humans ,RNA, Small Interfering ,Molecular Biology ,Mutation ,medicine.diagnostic_test ,urogenital system ,Endoplasmic reticulum ,Ubiquitination ,Membrane Proteins ,RNA ,ER retention ,Cell Biology ,Molecular biology ,Amino Acid Substitution ,Gene Expression Regulation ,Proteasome ,KCNQ1 Potassium Channel ,Guanosine Triphosphate - Abstract
Mutations in the potassium channel KCNQ1 that determine retention of the mutated proteins in the endoplasmic reticulum (ER) are associated with the autosomal dominant negative Romano-Ward LQT1 cardiac syndrome. In the present study, we have analyzed the consequences and the potential molecular mechanisms involved in the ER retention of three Romano-Ward mutations located in KCNQ1 N terminus (Y111C, L114P, and P117L). We showed that the mutant KCNQ1 proteins exhibited reduced expression levels with respect to wild-type (WT)-KCNQ1. Radiolabeling pulse-chase experiments revealed that the lower expression levels did not result from reduced rate of synthesis. Instead, using a combination of Western blot and pulse-chase experiments, we showed that the mutant channel Y111C-KCNQ1, used as a model, was ubiquitinated and degraded in the proteasome more rapidly (t((1/2)) = 82 min) than WT-KCNQ1 channel (t((1/2)) = 113 min). On the other hand, KCNQ1 degradation did not appear to involve the GTP-dependent pathway. We also showed that KCNE1 stabilized both wild-type and Y111C proteins. To identify potential actors involved in KCNQ1 degradation, we studied the implication of the ER-resident protein Derlin-1 in KCNQ1 degradation. We showed that although KCNQ1 and Derlin-1 share the same molecular complex and co-immunoprecipitate when co-expressed in HEK293FT cells, Derlin-1 did not affect KCNQ1 steady state expression and degradation. These data were confirmed in T84 cells that express endogenous KCNQ1 and Derlin-1. Small interfering RNA knock-down of Derlin-1 did not modify KCNQ1 expression level, and no interaction between endogenous KCNQ1 and Derlin-1 could be detected.
- Published
- 2009
- Full Text
- View/download PDF
45. Ventricular fibrillation with prominent early repolarization associated with a rare variant of KCNJ8/KATP channel
- Author
-
Michel Haïssaguerre, Yoram Rudy, Rukshen Weerasooriya, Gildas Loussouarn, Frederic Sacher, Joseph C. Koster, Ruedige Liersch M.D., Vincent Probst, Arthur A.M. Wilde, Jean-Jacques Schott, Eric Schulze-Bahr, Marc Horlitz, Stéphanie Chatel, Stefan Kääb, Hervé Le Marec, ACS - Amsterdam Cardiovascular Sciences, and Cardiology
- Subjects
medicine.medical_specialty ,Potassium Channels ,Adolescent ,Benign early repolarization ,Amiodarone ,Polymorphism, Single Nucleotide ,Sudden death ,Sudden cardiac death ,Electrocardiography ,KATP Channels ,Physiology (medical) ,Internal medicine ,medicine ,Humans ,Repolarization ,Genetic Predisposition to Disease ,Potassium Channels, Inwardly Rectifying ,Flecainide ,J wave ,business.industry ,Genetic Variation ,medicine.disease ,Anesthesia ,Ventricular Fibrillation ,Ventricular fibrillation ,Cardiology ,Female ,Cardiology and Cardiovascular Medicine ,business ,medicine.drug - Abstract
Background: Early repolarization in the inferolateral leads has been recently recognized as a frequent syndrome associated with idiopathic ventricular fibrillation (VF). We report the case of a patient presenting dramatic changes in the ECG in association with recurrent VF in whom a novel genetic variant has been identified. Case Report: This young female (14 years) was resuscitated in 2001 following an episode of sudden death due to VF. All examinations including coronary angiogram with ergonovine injection, MRI, and flecainide or isoproterenol infusion were normal. The patient had multiple (>100) recurrences of VF unresponsive to beta-blockers, lidocaine/mexiletine, verapamil, and amiodarone. Recurrences of VF were associated with massive accentuation of the early repolarization pattern at times mimicking acute myocardial ischemia. Coronary angiography during an episode with 1.2 mV J/ST elevation was normal. Isoproterenol infusion acutely suppressed electrical storms, while quinidine eliminated all recurrences of VF and restored a normal ECG over a follow-up of 65 months. Genomic DNA sequencing of KATP channel genes showed missense variant in exon 3 (NC_000012) of the KCNJ8 gene, a subunit of the KATP channel, conferring predisposition to dramatic repolarization changes and ventricular vulnerability.
- Published
- 2009
- Full Text
- View/download PDF
46. Kv7.1 (KCNQ1) properties and channelopathies
- Author
-
Isabelle Baró, Nicolas Rodriguez, Gildas Loussouarn, David Peroz, Frank S. Choveau, and Jean Mérot
- Subjects
Genetics ,Mutation ,Channel complex ,Physiology ,business.industry ,KCNQ1 Potassium Channel ,Long QT syndrome ,Cardiac metabolism ,Short QT syndrome ,medicine.disease ,medicine.disease_cause ,Medicine ,In patient ,business - Abstract
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.
- Published
- 2008
- Full Text
- View/download PDF
47. HIV-Tat induces a decrease in I
- Author
-
Zeineb, Es-Salah-Lamoureux, Mariam, Jouni, Olfat A, Malak, Nadjet, Belbachir, Zeina Reda, Al Sayed, Marine, Gandon-Renard, Guillaume, Lamirault, Chantal, Gauthier, Isabelle, Baró, Flavien, Charpentier, Kazem, Zibara, Patricia, Lemarchand, Bruno, Beaumelle, Nathalie, Gaborit, and Gildas, Loussouarn
- Subjects
Phosphatidylinositol 4,5-Diphosphate ,ERG1 Potassium Channel ,Induced Pluripotent Stem Cells ,Action Potentials ,Gene Expression ,Cell Differentiation ,Transfection ,Cell Line ,Electrophysiological Phenomena ,HEK293 Cells ,Potassium Channels, Voltage-Gated ,COS Cells ,KCNQ1 Potassium Channel ,Animals ,Humans ,Myocytes, Cardiac ,tat Gene Products, Human Immunodeficiency Virus ,RNA, Messenger - Abstract
Patients with HIV present with a higher prevalence of QT prolongation, of which molecular bases are still not clear. Among HIV proteins, Tat serves as a transactivator that stimulates viral genes expression and is required for efficient HIV replication. Tat is actively secreted into the blood by infected T-cells and affects organs such as the heart. Tat has been shown to alter cardiac repolarization in animal models but how this is mediated and whether this is also the case in human cells is unknown. In the present study, we show that Tat transfection in heterologous expression systems led to a decrease in hERG (underlying cardiac I
- Published
- 2015
48. Toward Personalized Medicine: Using Cardiomyocytes Differentiated From Urine-Derived Pluripotent Stem Cells to Recapitulate Electrophysiological Characteristics of Type 2 Long QT Syndrome
- Author
-
Kazem Zibara, Patricia Lemarchand, Amandine Caillaud, Mariam Jouni, Anais Rungoat, Flavien Charpentier, Isabelle Baró, Gildas Loussouarn, Nathalie Gaborit, Benoite Champon, Karim Si-Tayeb, Zeineb Es-Salah-Lamoureux, Xenia Latypova, 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), Laboratory of Stem Cells (ER045-PRASE), Lebanese University [Beirut] (LU), This work was funded by grants from the Lefoulon DelalandeFoundation, the Federation Francaise de Cardiologie, Genavieand the Marie Curie European Actions (PIIF-GA-2012-331436)to Dr Gaborit, by grants from the FP7 Marie Curie IRG277188/IPSMILD, the Leducq Foundation and the GenavieFoundation to Dr Si-Tayeb, by a grant from the VaCarMEproject funded by the Region Pays de la Loire to Dr Si-Tayeb, B. Champon and Dr Caillaud, and by a grant from theLebanese University to Dr Zibara. M. Jouni was awarded ascholarship from Association of Scientific Orientation andSpecialization (ASOS) and X. Latypova from Pasteur MutualiteFoundation. Dr Z. Es-Salah-Lamoureux was supported bygrants from the Lefoulon Delalande Foundation, the FrenchFoundation for Medical Research (FRM) and Genavie Foundation.Finally, this work was also supported by the Region Paysde Loire (Projet Devips)., European Project: IRG 277188,IPSMILD, European Project: 277188,EC:FP7:PEOPLE,FP7-PEOPLE-2010-RG,IPSMILD(2011), Lemarchand, Patricia, FP7 Marie Curie IRG 277188/IPSMILD - IPSMILD - IRG 277188 - INCOMING, HUMAN INDUCED PLURIPOTENT STEM CELLS AS A MODEL TO STUDY METABOLIC INHERITED LIVER DISEASES - IPSMILD - - EC:FP7:PEOPLE2011-12-01 - 2014-11-30 - 277188 - VALID, 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
Male ,urine-derived induced pluripotent stem cells ,ERG1 Potassium Channel ,Patch-Clamp Techniques ,Cellular differentiation ,Cell Culture Techniques ,Action Potentials ,cardiomyocytes ,Gene mutation ,Urine ,medicine.disease_cause ,Electrocardiography ,Cellular Reprogramming Techniques ,Myocytes, Cardiac ,Precision Medicine ,Induced pluripotent stem cell ,Cells, Cultured ,health care economics and organizations ,Original Research ,Mutation ,biology ,HERG gene ,Cell Differentiation ,Middle Aged ,3. Good health ,Phenotype ,Female ,Cardiology and Cardiovascular Medicine ,Pluripotent Stem Cells ,medicine.medical_specialty ,Long QT syndrome ,hERG ,Mutation, Missense ,Heterologous ,[SDV.BC]Life Sciences [q-bio]/Cellular Biology ,arrhythmia ,Young Adult ,Internal medicine ,medicine ,long QT syndrome ,Humans ,Genetic Predisposition to Disease ,[SDV.BC] Life Sciences [q-bio]/Cellular Biology ,business.industry ,medicine.disease ,Ether-A-Go-Go Potassium Channels ,High-Throughput Screening Assays ,Endocrinology ,Cancer research ,biology.protein ,business - Abstract
Background Human genetically inherited cardiac diseases have been studied mainly in heterologous systems or animal models, independent of patients' genetic backgrounds. Because sources of human cardiomyocytes ( CM s) are extremely limited, the use of urine samples to generate induced pluripotent stem cell–derived CM s would be a noninvasive method to identify cardiac dysfunctions that lead to pathologies within patients' specific genetic backgrounds. The objective was to validate the use of CMs differentiated from urine‐derived human induced pluripotent stem (UhiPS) cells as a new cellular model for studying patients' specific arrhythmia mechanisms. Methods and Results Cells obtained from urine samples of a patient with long QT syndrome who harbored the HERG A561P gene mutation and his asymptomatic noncarrier mother were reprogrammed using the episomal‐based method. Uhi PS cells were then differentiated into CM s using the matrix sandwich method. Uhi PS ‐ CM s showed proper expression of atrial and ventricular myofilament proteins and ion channels. They were electrically functional, with nodal‐, atrial‐ and ventricular‐like action potentials recorded using high‐throughput optical and patch‐clamp techniques. Comparison of HERG expression from the patient's Uhi PS ‐ CM s to the mother's Uhi PS ‐ CM s showed that the mutation led to a trafficking defect that resulted in reduced delayed rectifier K + current (I Kr ). This phenotype gave rise to action potential prolongation and arrhythmias. Conclusions UhiPS cells from patients carrying ion channel mutations can be used as novel tools to differentiate functional CMs that recapitulate cardiac arrhythmia phenotypes.
- Published
- 2015
- Full Text
- View/download PDF
49. 0441 : Electrophysiological characterization of a novel SCN5A mutation causing Brugada syndrome, using cardiomyocytes differentiated from hiPSCs
- Author
-
Kazem Zibara, Mariam Jouni, Gildas Loussouarn, Flavien Charpentier, Benoite Champon, Laurent David, Anais Rungoat, Nathalie Gaborit, Aude Derevier, Isabelle Baró, Patricia Lemarchanda, and Xenia Martin Latypova
- Subjects
congenital, hereditary, and neonatal diseases and abnormalities ,Mutation ,medicine.medical_specialty ,business.industry ,Mutant ,Context (language use) ,Gene mutation ,medicine.disease_cause ,medicine.disease ,Sudden death ,Cell biology ,Endocrinology ,Internal medicine ,medicine ,cardiovascular diseases ,Patch clamp ,Cellular model ,business ,Cardiology and Cardiovascular Medicine ,Brugada syndrome - Abstract
Rationale Brugada syndrome is a human hereditary cardiac disease known to cause ventricular tachyarrhythmias (torsade de pointes) that can lead to sudden death. In about 20% of the Brugada affected patients, mutations have been identified in the gene encoding the Na+ ion channel, SCN5A. Up to now, genotype-phenotype studies have been performed using heterologous expression systems that lack the correct cellular context of a cardiomyocyte. Human Induced Pluripotent stem cells (hiPSCs) offer a now new paradigm for gene mutation characterization. Objective In this study, using cardiomyocytes differentiated from hiPSCs, we have electrophysiologically characterized a previously undescribed mutation in SCN5A gene, carried by a Brugada affected patient. Methods and results hiPSCs from a Brugada affected patient carrying the N1722D mutation in SCN5A have been generated and validated. hiPSCs from a healthy subject were used as control. Using patch clamp techniques, the biophysical properties of the Na+ channel and action potential characteristics were evaluated in both cardiomyocytes differentiated from these hiPSCs and in a mammalian expression system expressing the mutant channel. Preliminary data from both cellular models suggest a three-times reduction in Na+ current. The hiPSCs-derived cardiomyocytes revealed a specific action potential phenotype which is still under investigation Conclusion Brugada syndrome modeling using hiPSCs-derived cardiomyocytes suggests that this cellular model recapitulates the characteristics of a loss-of-function Na+ channel mutation and that hiPSCs-derived cardiomyocytes can be used as an accurate model for cardiac Na+ channel disease.
- Published
- 2015
- Full Text
- View/download PDF
50. Molecular Basis of Inward Rectification: Structural Features of the Blocker Defined by Extended Polyamine Analogs
- Author
-
Laurence J. Marton, Gildas Loussouarn, and Colin G. Nichols
- Subjects
Pharmacology ,chemistry.chemical_classification ,COS cells ,Alkene ,Stereochemistry ,Mutant ,Block (permutation group theory) ,Spermine ,Potassium channel blocker ,chemistry.chemical_compound ,chemistry ,Cytoplasm ,COS Cells ,Chlorocebus aethiops ,Polyamines ,Potassium Channel Blockers ,medicine ,Animals ,Molecular Medicine ,Potassium Channels, Inwardly Rectifying ,Polyamine ,medicine.drug - Abstract
Polyamines cause inward rectification of Kir K(+) channels by blocking deep within the channel pore. We investigated structural constraints of polyamine block of strongly rectifying mutant K(ATP) channels (Kir6.2[L164C,N160D,C166S] + SUR1). We studied three groups of polyamine analogs: 1) conformationally restricted linear tetra-amines with a cycloalkyl or alkene group between the second and third amines (CGC-11047, CGC-11093, CGC-11099, and CGC-11098), 2) conformationally restricted linear deca-amines with a cycloalkyl or alkene group between the fifth and sixth amines (CGC-11150, CGC-11179, and CGC-11241), and 3) cyclic tetra-amines (CGC-11174, CGC-11197, CGC-11199, and CGC-11254). All linear analogs cause a voltage-dependent block similar to that of spermine, but slightly weaker (at 1 microM, V(1/2) for spermine block = -10 +/- 1 mV, Z = 2.9 +/- 0.1, n = 19; V(1/2) for analogs varies from polyamine -7 to +10 mV, Z = 2.6-3.9). These data indicate tolerance for conformational restriction and an upper limit to the voltage dependence of the blocking process. There was no voltage-dependent block by the cyclic compounds; instead, they induce irreversible rundown of the current. Structural models of Kir channels suggest that a narrow entry at the top of the cytoplasmic pore may exclude cyclic analogs from the inner cavity, thereby explaining the structure-activity relationship that we observe.
- Published
- 2005
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.