359 results on '"Nargeot, J."'
Search Results
102. Influence of metabolic inhibition by NaCN on electrical and mechanical activities of frog atrial fibers: Studies using current and voltage clamp
- Author
-
Nargeot, J, primary
- Published
- 1978
- Full Text
- View/download PDF
103. 040 - Cardiac mGluR1 metabotropic receptors protect against ischemiareperfusion injury in the mouse heart.
- Author
-
Vincent, A., Sportouch, C., Covinhes, A., Barrere, C., Gallot, L., Delgado-Betancourt, V., Lattuca, B., Solecki, K., Piot, C., Nargeot, J., and Barrere-Lemaire, S.
- Published
- 2017
- Full Text
- View/download PDF
104. P666 Heart rate control protects against ischemia-reperfusion injury.
- Author
-
Delgado Betancourt, V, Covihnes, A, Mesirca, P, Bidaud, I, Nargeot, J, Piot, C, Striessnig, J, Mangoni, ME, and Barrere-Lemaire, S
- Subjects
HEART beat ,REPERFUSION injury ,ISCHEMIA ,MYOCARDIAL infarction ,HEART cells ,IVABRADINE - Abstract
Acute myocardial infarction (AMI) is the major cause of cardiovascular mortality worldwide. Early reperfusion is the only treatment recommended to reduce infarct size (IS). However, reperfusion presents also deleterious effects such as ischemia-reperfusion (IR) injury due to irreversible apoptotic death of cardiomyocytes. Most ischemic episodes are triggered by an increase in heart rate (HR) that induces an imbalance between myocardial oxygen delivery and consumption. The BEAUTIFUL clinical trial has demonstrated that moderate HR reduction diminishes the frequency of AMI episodes in patients with stable coronary artery disease and increased HR at rest. The HCN-mediated If current and the Cav1.3-mediated L-type Ca2+ currents play important roles in the generation of automaticity and HR, therefore they are interesting targets for selective control of HR and cardioprotection during AMI.The aim of our study was to investigate if targeting Cav1.3 channels could be an efficient strategy to reduce IS. Cav1.3 -/- mice was used as a genetic model of Cav1.3 inhibition because of the lack of selective blocker. Ivabradine, the selective f-channel blocker, was used for pure HR reduction as a positive control. Results show that selective HR decrease (40%) in an in vivo mouse model of acute MI is associated with reduced IR injury. Ivabradine administration 30 minutes before ischemia significantly reduced IS (35%). Cav1.3 -/- mice presented reduced IS (30%) compared to WT mice. In addition, preliminary results show that Girk4 -/- mice, a genetic model of moderate tachycardia (10%) displayed increased IS (45%) compared to control mice. In conclusion, results suggest a direct relationship between HR and IR injury and that inhibition of Cav1.3 channels constitutes a promising strategy to reduce both HR and IS. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
105. Functional Coupling Between Voltage-Gated Calcium Channels, and Intracellular Ca2+ Transients in rat Aortic Myocytes.
- Author
-
Mangoni, M. E, Richard, S., Choby, C., Lacolley, P., Laurent, S., and Nargeot, J.
- Published
- 1999
106. Differential expression of voltage-gated Ca 2+-currents in cultivated aortic myocytes
- Author
-
Richard, S., Neveu, D., Carnac, G., Bodin, P., Travo, P., and Nargeot, J.
- Published
- 1992
- Full Text
- View/download PDF
107. Modulation of Ca currents in isolated frog atrial cells studied with photosensitive probes. Regulation by cAMP and Ca 2+: A common pathway?
- Author
-
Charnet, P., Richard, S., Gurney, A.M., Ouadid, H., Tiaho, F., and Nargeot, J.
- Published
- 1991
- Full Text
- View/download PDF
108. Kasai M Yoshioka T Suzuki H Biosignal Transduction Mechanisms 1989 Springer Verlag New York
- Author
-
Nargeot, J
- Published
- 1990
- Full Text
- View/download PDF
109. Potentiation of Ca current by 5-hydroxytryptamine in human atrial myocytes via newly described 5-HT 4 receptors
- Author
-
Ouadid, H., Séguin, J., Dumuis, A., Bockaert, J., and Nargeot, J.
- Published
- 1991
- Full Text
- View/download PDF
110. Mesenchymal stromal cells for improvement of cardiac function following acute myocardial infarction: a matter of timing.
- Author
-
Barrère-Lemaire S, Vincent A, Jorgensen C, Piot C, Nargeot J, and Djouad F
- Subjects
- Humans, Myocardium metabolism, Cardiovascular Physiological Phenomena, Myocardial Infarction therapy, Myocardial Infarction pathology, Heart Failure metabolism, Mesenchymal Stem Cells metabolism, Mesenchymal Stem Cells pathology
- Abstract
Acute myocardial infarction (AMI) is the leading cause of cardiovascular death and remains the most common cause of heart failure. Reopening of the occluded artery, i.e., reperfusion, is the only way to save the myocardium. However, the expected benefits of reducing infarct size are disappointing due to the reperfusion paradox, which also induces specific cell death. These ischemia-reperfusion (I/R) lesions can account for up to 50% of final infarct size, a major determinant for both mortality and the risk of heart failure (morbidity). In this review, we provide a detailed description of the cell death and inflammation mechanisms as features of I/R injury and cardioprotective strategies such as ischemic postconditioning as well as their underlying mechanisms. Due to their biological properties, the use of mesenchymal stromal/stem cells (MSCs) has been considered a potential therapeutic approach in AMI. Despite promising results and evidence of safety in preclinical studies using MSCs, the effects reported in clinical trials are not conclusive and even inconsistent. These discrepancies were attributed to many parameters such as donor age, in vitro culture, and storage time as well as injection time window after AMI, which alter MSC therapeutic properties. In the context of AMI, future directions will be to generate MSCs with enhanced properties to limit cell death in myocardial tissue and thereby reduce infarct size and improve the healing phase to increase postinfarct myocardial performance.
- Published
- 2024
- Full Text
- View/download PDF
111. Selective blockade of Ca v 1.2 (α1C) versus Ca v 1.3 (α1D) L-type calcium channels by the black mamba toxin calciseptine.
- Author
-
Mesirca P, Chemin J, Barrère C, Torre E, Gallot L, Monteil A, Bidaud I, Diochot S, Lazdunski M, Soong TW, Barrère-Lemaire S, Mangoni ME, and Nargeot J
- Subjects
- Animals, Myocytes, Cardiac metabolism, Protein Isoforms, Calcium metabolism, Calcium Channels, L-Type physiology, Dendroaspis metabolism
- Abstract
L-type voltage-gated calcium channels are involved in multiple physiological functions. Currently available antagonists do not discriminate between L-type channel isoforms. Importantly, no selective blocker is available to dissect the role of L-type isoforms Ca
v 1.2 and Cav 1.3 that are concomitantly co-expressed in the heart, neuroendocrine and neuronal cells. Here we show that calciseptine, a snake toxin purified from mamba venom, selectively blocks Cav 1.2 -mediated L-type calcium currents (ICaL ) at concentrations leaving Cav 1.3-mediated ICaL unaffected in both native cardiac myocytes and HEK-293T cells expressing recombinant Cav 1.2 and Cav 1.3 channels. Functionally, calciseptine potently inhibits cardiac contraction without altering the pacemaker activity in sino-atrial node cells, underscoring differential roles of Cav 1.2- and Cav 1.3 in cardiac contractility and automaticity. In summary, calciseptine is a selective L-type Cav 1.2 Ca2+ channel blocker and should be a valuable tool to dissect the role of these L-channel isoforms., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
112. Heart rate reduction after genetic ablation of L-type Ca v 1.3 channels induces cardioprotection against ischemia-reperfusion injury.
- Author
-
Delgado-Betancourt V, Chinda K, Mesirca P, Barrère C, Covinhes A, Gallot L, Vincent A, Bidaud I, Kumphune S, Nargeot J, Piot C, Wickman K, Mangoni ME, and Barrère-Lemaire S
- Abstract
Background: Acute myocardial infarction (AMI) is the major cause of cardiovascular mortality worldwide. Most ischemic episodes are triggered by an increase in heart rate, which induces an imbalance between myocardial oxygen delivery and consumption. Developing drugs that selectively reduce heart rate by inhibiting ion channels involved in heart rate control could provide more clinical benefits. The Ca
v 1.3-mediated L-type Ca2+ current ( ICav1.3 ) play important roles in the generation of heart rate. Therefore, they can constitute relevant targets for selective control of heart rate and cardioprotection during AMI., Objective: We aimed to investigate the relationship between heart rate and infarct size using mouse strains knockout for Cav 1.3 ( Cav 1.3-/- ) L-type calcium channel and of the cardiac G protein gated potassium channel ( Girk4-/- ) in association with the funny (f)-channel inhibitor ivabradine., Methods: Wild-type (WT), Cav 1.3+/- , Cav 1.3-/- and Girk4-/- mice were used as models of respectively normal heart rate, moderate heart rate reduction, bradycardia, and mild tachycardia, respectively. Mice underwent a surgical protocol of myocardial IR (40 min ischemia and 60 min reperfusion). Heart rate was recorded by one-lead surface ECG recording, and infarct size measured by triphenyl tetrazolium chloride staining. In addition, Cav 1.3-/- and WT hearts perfused on a Langendorff system were subjected to the same ischemia-reperfusion protocol ex vivo , without or with atrial pacing, and the coronary flow was recorded., Results: Cav 1.3-/- mice presented reduced infarct size (-29%), while Girk4-/- displayed increased infarct size (+30%) compared to WT mice. Consistently, heart rate reduction in Cav 1.3+/- or by the f-channel blocker ivabradine was associated with significant decrease in infarct size (-27% and -32%, respectively) in comparison to WT mice., Conclusion: Our results show that decreasing heart rate allows to protect the myocardium against IR injury in vivo and reveal a close relationship between basal heart rate and IR injury. In addition, this study suggests that targeting Cav 1.3 channels could constitute a relevant target for reducing infarct size, since maximal heart rate dependent cardioprotective effect is already observed in Cav 1.3+/- mice., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (© 2023 Delgado-Betancourt, Chinda, Mesirca, Barrere, Covinhes, Gallot, Vincent, Bidaud, Kumphune, Nargeot, Piot, Wickman, Mangoni and Barrère-Lemaire.)- Published
- 2023
- Full Text
- View/download PDF
113. Correction: PPARβ/δ priming enhances the anti-apoptotic and therapeutic properties of mesenchymal stromal cells in myocardial ischemia-reperfusion injury.
- Author
-
Sarre C, Contreras-Lopez R, Nernpermpisooth N, Barrere C, Bahraoui S, Terraza C, Tejedor G, Vincent A, Luz-Crawford P, Kongpol K, Kumphune S, Piot C, Nargeot J, Jorgensen C, Djouad F, and Barrere-Lemaire S
- Published
- 2022
- Full Text
- View/download PDF
114. PPARβ/δ priming enhances the anti-apoptotic and therapeutic properties of mesenchymal stromal cells in myocardial ischemia-reperfusion injury.
- Author
-
Sarre C, Contreras-Lopez R, Nernpermpisooth N, Barrere C, Bahraoui S, Terraza C, Tejedor G, Vincent A, Luz-Crawford P, Kongpol K, Kumphune S, Piot C, Nargeot J, Jorgensen C, Djouad F, and Barrere-Lemaire S
- Subjects
- Animals, Endothelial Cells metabolism, Hydrogen Peroxide, Mice, Thiazoles, Mesenchymal Stem Cells metabolism, Myocardial Infarction metabolism, Myocardial Infarction therapy, Myocardial Reperfusion Injury metabolism, Myocardial Reperfusion Injury therapy, PPAR delta agonists, PPAR delta genetics, PPAR delta metabolism, PPAR-beta agonists, PPAR-beta genetics, PPAR-beta metabolism
- Abstract
Background: Mesenchymal Stromal Cells (MSC) have been widely used for their therapeutic properties in many clinical applications including myocardial infarction. Despite promising preclinical results and evidences of safety and efficacy in phases I/ II, inconsistencies in phase III trials have been reported. In a previous study, we have shown using MSC derived from the bone marrow of PPARβ/δ (Peroxisome proliferator-activated receptors β/δ) knockout mice that the acute cardioprotective properties of MSC during the first hour of reperfusion are PPARβ/δ-dependent but not related to the anti-inflammatory effect of MSC. However, the role of the modulation of PPARβ/δ expression on MSC cardioprotective and anti-apoptotic properties has never been investigated., Objectives: The aim of this study was to investigate the role of PPARβ/δ modulation (inhibition or activation) in MSC therapeutic properties in vitro and ex vivo in an experimental model of myocardial infarction., Methods and Results: Naïve MSC and MSC pharmacologically activated or inhibited for PPARβ/δ were challenged with H
2 O2 . Through specific DNA fragmentation quantification and qRT-PCR experiments, we evidenced in vitro an increased resistance to oxidative stress in MSC pre-treated by the PPARβ/δ agonist GW0742 versus naïve MSC. In addition, PPARβ/δ-priming allowed to reveal the anti-apoptotic effect of MSC on cardiomyocytes and endothelial cells in vitro. When injected during reperfusion, in an ex vivo heart model of myocardial infarction, 3.75 × 105 PPARβ/δ-primed MSC/heart provided the same cardioprotective efficiency than 7.5 × 105 naïve MSC, identified as the optimal dose in our experimental model. This enhanced short-term cardioprotective effect was associated with an increase in both anti-apoptotic effects and the number of MSC detected in the left ventricular wall at 1 h of reperfusion. By contrast, PPARβ/δ inhibition in MSC before their administration in post-ischemic hearts during reperfusion decreased their cardioprotective effects., Conclusion: Altogether these results revealed that PPARβ/δ-primed MSC exhibit an increased resistance to oxidative stress and enhanced anti-apoptotic properties on cardiac cells in vitro. PPARβ/δ-priming appears as an innovative strategy to enhance the cardioprotective effects of MSC and to decrease the therapeutic injected doses. These results could be of major interest to improve MSC efficacy for the cardioprotection of injured myocardium in AMI patients., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
115. Therapeutic Peptides to Treat Myocardial Ischemia-Reperfusion Injury.
- Author
-
Fernandez Rico C, Konate K, Josse E, Nargeot J, Barrère-Lemaire S, and Boisguérin P
- Abstract
Cardiovascular diseases (CVD) including acute myocardial infarction (AMI) rank first in worldwide mortality and according to the World Health Organization (WHO), they will stay at this rank until 2030. Prompt revascularization of the occluded artery to reperfuse the myocardium is the only recommended treatment (by angioplasty or thrombolysis) to decrease infarct size (IS). However, despite beneficial effects on ischemic lesions, reperfusion leads to ischemia-reperfusion (IR) injury related mainly to apoptosis. Improvement of revascularization techniques and patient care has decreased myocardial infarction (MI) mortality however heart failure (HF) morbidity is increasing, contributing to the cost-intense worldwide HF epidemic. Currently, there is no treatment for reperfusion injury despite promising results in animal models. There is now an obvious need to develop new cardioprotective strategies to decrease morbidity/mortality of CVD, which is increasing due to the aging of the population and the rising prevalence rates of diabetes and obesity. In this review, we will summarize the different therapeutic peptides developed or used focused on the treatment of myocardial IR injury (MIRI). Therapeutic peptides will be presented depending on their interacting mechanisms (apoptosis, necroptosis, and inflammation) reported as playing an important role in reperfusion injury following myocardial ischemia. The search and development of therapeutic peptides have become very active, with increasing numbers of candidates entering clinical trials. Their optimization and their potential application in the treatment of patients with AMI will be discussed., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Fernandez Rico, Konate, Josse, Nargeot, Barrère-Lemaire and Boisguérin.)
- Published
- 2022
- Full Text
- View/download PDF
116. PPARβ/δ Is Required for Mesenchymal Stem Cell Cardioprotective Effects Independently of Their Anti-inflammatory Properties in Myocardial Ischemia-Reperfusion Injury.
- Author
-
Nernpermpisooth N, Sarre C, Barrere C, Contreras R, Luz-Crawford P, Tejedor G, Vincent A, Piot C, Kumphune S, Nargeot J, Jorgensen C, Barrère-Lemaire S, and Djouad F
- Abstract
Myocardial infarction ranks first for the mortality worldwide. Because the adult heart is unable to regenerate, fibrosis develops to compensate for the loss of contractile tissue after infarction, leading to cardiac remodeling and heart failure. Adult mesenchymal stem cells (MSC) regenerative properties, as well as their safety and efficacy, have been demonstrated in preclinical models. However, in clinical trials, their beneficial effects are controversial. In an experimental model of arthritis, we have previously shown that PPARβ / δ deficiency enhanced the therapeutic effect of MSC. The aim of the present study was to compare the therapeutic effects of wild-type MSC (MSC) and MSC deficient for PPARβ / δ (KO MSC) perfused in an ex vivo mouse model of ischemia-reperfusion (IR) injury. For this purpose, hearts from C57BL/6J mice were subjected ex vivo to 30 min ischemia followed by 1-h reperfusion. MSC and KO MSC were injected into the Langendorff system during reperfusion. After 1 h of reperfusion, the TTC method was used to assess infarct size. Coronary effluents collected in basal condition (before ischemia) and after ischemia at 1 h of reperfusion were analyzed for their cytokine profiles. The dose-response curve for the cardioprotection was established ex vivo using different doses of MSC (3.10
5 , 6.105 , and 24.105 cells / heart) and the dose of 6.105 MSC was found to be the optimal concentration. We showed that the cardioprotective effect of MSC was PPARβ / δ-dependent since it was lost using KO MSC. Moreover, cytokine profiling of the coronary effluents collected in the eluates after 60 min of reperfusion revealed that MSC treatment decreases CXCL1 chemokine and interleukin-6 release compared with untreated hearts. This anti-inflammatory effect of MSC was also observed when hearts were treated with PPARβ / δ-deficient MSC. In conclusion, our study revealed that the acute cardioprotective properties of MSC in an ex vivo model of IR injury, assessed by a decreased infarct size at 1 h of reperfusion, are PPARβ / δ-dependent but not related to their anti-inflammatory effects., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Nernpermpisooth, Sarre, Barrere, Contreras, Luz-Crawford, Tejedor, Vincent, Piot, Kumphune, Nargeot, Jorgensen, Barrère-Lemaire and Djouad.)- Published
- 2021
- Full Text
- View/download PDF
117. Concomitant genetic ablation of L-type Ca v 1.3 (α 1D ) and T-type Ca v 3.1 (α 1G ) Ca 2+ channels disrupts heart automaticity.
- Author
-
Baudot M, Torre E, Bidaud I, Louradour J, Torrente AG, Fossier L, Talssi L, Nargeot J, Barrère-Lemaire S, Mesirca P, and Mangoni ME
- Subjects
- Animals, Bradycardia genetics, Bradycardia physiopathology, Calcium metabolism, Disease Models, Animal, Electrocardiography, Heart Rate, Mice, Mice, Knockout, Sarcoplasmic Reticulum metabolism, Atrioventricular Node physiopathology, Bradycardia diagnosis, Calcium Channels, L-Type genetics, Calcium Channels, T-Type genetics, Sinoatrial Node physiopathology
- Abstract
Cardiac automaticity is set by pacemaker activity of the sinus node (SAN). In addition to the ubiquitously expressed cardiac voltage-gated L-type Ca
v 1.2 Ca2+ channel isoform, pacemaker cells within the SAN and the atrioventricular node co-express voltage-gated L-type Cav 1.3 and T-type Cav 3.1 Ca2+ channels (SAN-VGCCs). The role of SAN-VGCCs in automaticity is incompletely understood. We used knockout mice carrying individual genetic ablation of Cav 1.3 (Cav 1.3-/- ) or Cav 3.1 (Cav 3.1-/- ) channels and double mutant Cav 1.3-/- /Cav 3.1-/- mice expressing only Cav 1.2 channels. We show that concomitant loss of SAN-VGCCs prevents physiological SAN automaticity, blocks impulse conduction and compromises ventricular rhythmicity. Coexpression of SAN-VGCCs is necessary for impulse formation in the central SAN. In mice lacking SAN-VGCCs, residual pacemaker activity is predominantly generated in peripheral nodal and extranodal sites by f-channels and TTX-sensitive Na+ channels. In beating SAN cells, ablation of SAN-VGCCs disrupted late diastolic local intracellular Ca2+ release, which demonstrates an important role for these channels in supporting the sarcoplasmic reticulum based "Ca2+ clock" mechanism during normal pacemaking. These data implicate an underappreciated role for co-expression of SAN-VGCCs in heart automaticity and define an integral role for these channels in mechanisms that control the heartbeat.- Published
- 2020
- Full Text
- View/download PDF
118. Anti-apoptotic peptide for long term cardioprotection in a mouse model of myocardial ischemia-reperfusion injury.
- Author
-
Covinhes A, Gallot L, Barrère C, Vincent A, Sportouch C, Piot C, Lebleu B, Nargeot J, Boisguérin P, and Barrère-Lemaire S
- Subjects
- Animals, Male, Mice, Mice, Inbred C57BL, Myocardial Infarction metabolism, Myocardial Infarction pathology, Myocardial Reperfusion Injury metabolism, Myocardial Reperfusion Injury pathology, Apoptosis drug effects, Disease Models, Animal, Myocardial Infarction prevention & control, Myocardial Reperfusion Injury prevention & control, Peptide Fragments pharmacology
- Abstract
Reperfusion therapy during myocardial infarction (MI) leads to side effects called ischemia-reperfusion (IR) injury for which no treatment exists. While most studies have targeted the intrinsic apoptotic pathway to prevent IR injury with no successful clinical translation, we evidenced recently the potent cardioprotective effect of the anti-apoptotic Tat-DAXXp (TD) peptide targeting the FAS-dependent extrinsic pathway. The aim of the present study was to evaluate TD long term cardioprotective effects against IR injury in a MI mouse model. TD peptide (1 mg/kg) was administered in mice subjected to MI (TD; n = 21), 5 min prior to reperfusion, and were clinically followed-up during 6 months after surgery. Plasma cTnI concentration evaluated 24 h post-MI was 70%-decreased in TD (n = 16) versus Ctrl (n = 20) mice (p***). Strain echocardiography highlighted a 24%-increase (p****) in the ejection fraction mean value in TD-treated (n = 12) versus Ctrl mice (n = 17) during the 6 month-period. Improved cardiac performance was associated to a 54%-decrease (p**) in left ventricular fibrosis at 6 months in TD (n = 16) versus Ctrl (n = 20). In conclusion, targeting the extrinsic pathway with TD peptide at the onset of reperfusion provided long-term cardioprotection in a mouse model of myocardial IR injury by improving post-MI cardiac performance and preventing cardiac remodeling.
- Published
- 2020
- Full Text
- View/download PDF
119. A novel therapeutic peptide targeting myocardial reperfusion injury.
- Author
-
Boisguérin P, Covinhes A, Gallot L, Barrère C, Vincent A, Busson M, Piot C, Nargeot J, Lebleu B, and Barrère-Lemaire S
- Subjects
- Animals, Cell Line, Cell Survival drug effects, Co-Repressor Proteins metabolism, Disease Models, Animal, Male, Mice, Inbred C57BL, Molecular Chaperones metabolism, Myocardial Infarction metabolism, Myocardial Infarction pathology, Myocardial Infarction physiopathology, Myocardial Reperfusion Injury metabolism, Myocardial Reperfusion Injury pathology, Myocardial Reperfusion Injury physiopathology, Myocytes, Cardiac metabolism, Myocytes, Cardiac pathology, Recovery of Function drug effects, Signal Transduction, fas Receptor metabolism, Apoptosis drug effects, Cell-Penetrating Peptides pharmacology, Co-Repressor Proteins antagonists & inhibitors, Molecular Chaperones antagonists & inhibitors, Myocardial Infarction prevention & control, Myocardial Reperfusion Injury prevention & control, Myocytes, Cardiac drug effects
- Abstract
Aims: Regulated cell death is a main contributor of myocardial ischaemia-reperfusion (IR) injury during acute myocardial infarction. In this context, targeting apoptosis could be a potent therapeutical strategy. In a previous study, we showed that DAXX (death-associated protein) was essential for transducing the FAS-dependent apoptotic signal during IR injury. The present study aims at evaluating the cardioprotective effects of a synthetic peptide inhibiting FAS:DAXX interaction., Methods and Results: An interfering peptide was engineered and then coupled to the Tat cell penetrating peptide (Tat-DAXXp). Its internalization and anti-apoptotic properties were demonstrated in primary cardiomyocytes. Importantly, an intravenous bolus injection of Tat-DAXXp (1 mg/kg) 5 min before reperfusion in a murine myocardial IR model decreased infarct size by 48% after 24 h of reperfusion. In addition, Tat-DAXXp was still efficient after a 30-min delayed administration, and was completely degraded and eliminated within 24 h thereby reducing risks of potential side effects. Importantly, Tat-DAXXp reduced mouse early post-infarction mortality by 67%. Mechanistically, cardioprotection was supported by both anti-apoptotic and pro-survival effects, and an improvement of myocardial functional recovery as evidenced in ex vivo experiments., Conclusions: Our study demonstrates that a single dose of Tat-DAXXp injected intravenously at the onset of reperfusion leads to a strong cardioprotection in vivo by inhibiting IR injury validating Tat-DAXXp as a promising candidate for therapeutic application., (Published on behalf of the European Society of Cardiology. All rights reserved. © The Author(s) 2019. For permissions, please email: journals.permissions@oup.com.)
- Published
- 2020
- Full Text
- View/download PDF
120. The cardioprotective effects of secretory leukocyte protease inhibitor against myocardial ischemia/reperfusion injury.
- Author
-
Prompunt E, Sanit J, Barrère-Lemaire S, Nargeot J, Noordali H, Madhani M, and Kumphune S
- Abstract
Protease enzymes generated from injured cells and leukocytes are the primary cause of myocardial cell damage following ischemia/reperfusion (I/R). The inhibition of protease enzyme activity via the administration of particular drugs may reduce injury and potentially save patients' lives. The aim of the current study was to investigate the cardioprotective effects of treatment with recombinant human secretory leukocyte protease inhibitor (rhSLPI) on in vitro and ex vivo models of myocardial I/R injury. rhSLPI was applied to isolated adult rat ventricular myocytes (ARVMs) subjected to simulated I/R and to ex vivo murine hearts prior to I/R injury. Cellular injury, cell viability, reactive oxygen species (ROS) levels, and levels of associated proteins were assessed. The results demonstrated that administration of rhSLPI prior to or during sI/R significantly reduced the death and injury of ARVMs and significantly reduced intracellular ROS levels in ARVMs during H
2 O2 stimulation. In addition, treatment of ARVMs with rhSLPI significantly attenuated p38 mitogen-activated protein kinase (MAPK) activation and increased the activation of Akt. Furthermore, pretreatment of ex vivo murine hearts with rhSLPI prior to I/R significantly decreased infarct size, attenuated p38 MAPK activation and increased Akt phosphorylation. The results of the current study demonstrated that treatment with rhSLPI induced a cardioprotective effect and reduced ARVM injury and death, intracellular ROS levels and infarct size. rhSLPI also attenuated p38 MAPK phosphorylation and activated Akt phosphorylation. These results suggest that rhSLPI may be developed as a novel therapeutic strategy of treating ischemic heart disease.- Published
- 2018
- Full Text
- View/download PDF
121. Acute and long-term cardioprotective effects of the Traditional Chinese Medicine MLC901 against myocardial ischemia-reperfusion injury in mice.
- Author
-
Vincent A, Covinhes A, Barrère C, Gallot L, Thoumala S, Piot C, Heurteaux C, Lazdunski M, Nargeot J, and Barrère-Lemaire S
- Subjects
- Animals, Disease Models, Animal, Extracellular Signal-Regulated MAP Kinases metabolism, Fibrosis drug therapy, Humans, Male, Mice, Mice, Inbred C57BL, Oncogene Protein v-akt metabolism, Regional Blood Flow drug effects, Signal Transduction, Troponin I blood, Drugs, Chinese Herbal therapeutic use, Heart drug effects, Medicine, Chinese Traditional, Myocardial Infarction drug therapy, Myocardium pathology, Reperfusion Injury drug therapy
- Abstract
MLC901, a traditional Chinese medicine containing a cocktail of active molecules, both reduces cerebral infarction and improves recovery in patients with ischemic stroke. The aim of this study was to evaluate the acute and long-term benefits of MLC901 in ischemic and reperfused mouse hearts. Ex vivo, under physiological conditions, MLC901 did not show any modification in heart rate and contraction amplitude. However, upon an ischemic insult, MLC901 administration during reperfusion, improved coronary flow in perfused hearts. In vivo, MLC901 (4 µg/kg) intravenous injection 5 minutes before reperfusion provided a decrease in both infarct size (49.8%) and apoptosis (49.9%) after 1 hour of reperfusion. Akt and ERK1/2 survival pathways were significantly activated in the myocardium of those mice. In the 4-month clinical follow-up upon an additional continuous per os administration, MLC901 treatment decreased cardiac injury as revealed by a 45%-decrease in cTnI plasmatic concentrations and an improved cardiac performance assessed by echocardiography. A histological analysis revealed a 64%-decreased residual scar fibrosis and a 44%-increased vascular density in the infarct region. This paper demonstrates that MLC901 treatment was able to provide acute and long-term cardioprotective effects in a murine model of myocardial ischemia-reperfusion injury in vivo.
- Published
- 2017
- Full Text
- View/download PDF
122. Cardiac mGluR1 metabotropic receptors in cardioprotection.
- Author
-
Vincent A, Sportouch C, Covinhes A, Barrère C, Gallot L, Delgado-Betancourt V, Lattuca B, Solecki K, Boisguérin P, Piot C, Nargeot J, and Barrère-Lemaire S
- Subjects
- Animals, Disease Models, Animal, Excitatory Amino Acid Antagonists pharmacology, Genetic Predisposition to Disease, Mice, Inbred C57BL, Mice, Knockout, Myocardial Infarction metabolism, Myocardial Infarction pathology, Myocardial Infarction physiopathology, Myocardial Reperfusion Injury metabolism, Myocardial Reperfusion Injury pathology, Myocardial Reperfusion Injury physiopathology, Myocardium pathology, Phenotype, Phosphatidylinositol 3-Kinase metabolism, Proto-Oncogene Proteins c-akt metabolism, Receptors, Metabotropic Glutamate deficiency, Receptors, Metabotropic Glutamate genetics, Signal Transduction, Time Factors, Ventricular Function, Left drug effects, Excitatory Amino Acid Agonists administration & dosage, Glutamine administration & dosage, Myocardial Infarction prevention & control, Myocardial Reperfusion Injury prevention & control, Myocardium metabolism, Receptors, Metabotropic Glutamate agonists
- Abstract
Aims: In a previous study using a genome-wide microarray strategy, we identified metabotropic glutamate receptor 1 (mGluR1) as a putative cardioprotective candidate in ischaemic postconditioning (PostC). In the present study, we investigated the role of cardiac mGluR1 receptors during cardioprotection against myocardial ischaemia-reperfusion injury in the mouse myocardium., Methods and Results: mGluR1 activation by glutamate administered 5 min before reperfusion in C57Bl/6 mice subjected to a myocardial ischaemia protocol strongly decreased both infarct size and DNA fragmentation measured at 24 h reperfusion. This cardioprotective effect was mimicked by the mGluR1 agonist, DHPG (10 μM), and abolished when glutamate was coinjected with the mGluR1 antagonist YM298198 (100 nM). Wortmannin (100 nM), an inhibitor of PI3-kinase, was able to prevent glutamate-induced cardioprotection. A glutamate bolus at the onset of reperfusion failed to protect the heart of mGluR1 knockout mice subjected to a myocardial ischaemia-reperfusion protocol, although PostC still protected the mGluR1 KO mice. Glutamate-treatment improved post-infarction functional recovery as evidenced by an echocardiographic study performed 15 days after treatment and by a histological evaluation of fibrosis 21 days post-treatment. Interestingly, restoration of functional mGluR1s by a PostC stimulus was evidenced at the transcriptional level. Since mGluR1s were localized at the surface membrane of cardiomyocytes, they might contribute to the cardioprotective effect of ischaemic PostC as other Gq-coupled receptors., Conclusion: This study provides the first demonstration that mGluR1 activation at the onset of reperfusion induces cardioprotection and might represent a putative strategy to prevent ischaemia-reperfusion injury., (Published on behalf of the European Society of Cardiology. All rights reserved. © The Author 2017. For permissions, please email: journals.permissions@oup.com.)
- Published
- 2017
- Full Text
- View/download PDF
123. L-type Cav1.3 channels regulate ryanodine receptor-dependent Ca2+ release during sino-atrial node pacemaker activity.
- Author
-
Torrente AG, Mesirca P, Neco P, Rizzetto R, Dubel S, Barrere C, Sinegger-Brauns M, Striessnig J, Richard S, Nargeot J, Gomez AM, and Mangoni ME
- Subjects
- Action Potentials physiology, Animals, Calcium Channels, L-Type genetics, Mice, Inbred C57BL, Mice, Knockout, Pacemaker, Artificial, Ryanodine Receptor Calcium Release Channel genetics, Sinoatrial Node metabolism, Calcium metabolism, Calcium Channels, L-Type metabolism, Sarcoplasmic Reticulum metabolism
- Abstract
Aims: Sino-atrial node (SAN) automaticity is an essential mechanism of heart rate generation that is still not completely understood. Recent studies highlighted the importance of intracellular Ca(2+) ([Ca(2+)]i) dynamics during SAN pacemaker activity. Nevertheless, the functional role of voltage-dependent L-type Ca(2+) channels in controlling SAN [Ca(2+)]i release is largely unexplored. Since Cav1.3 is the predominant L-type Ca(2+) channel isoform in SAN cells, we studied [Ca(2+)]i dynamics in isolated cells and ex vivo SAN preparations explanted from wild-type (WT) and Cav1.3 knockout (KO) mice (Cav1.3(-/-))., Methods and Results: We found that Cav1.3 deficiency strongly impaired [Ca(2+)]i dynamics, reducing the frequency of local [Ca(2+)]i release events and preventing their synchronization. This impairment inhibited the generation of Ca(2+) transients and delayed spontaneous activity. We also used action potentials recorded in WT SAN cells as voltage-clamp commands for Cav1.3(-/-) cells. Although these experiments showed abolished Ca(2+) entry through L-type Ca(2+) channels in the diastolic depolarization range of KO SAN cells, their sarcoplasmic reticulum Ca(2+) load remained normal. β-Adrenergic stimulation enhanced pacemaking of both genotypes, though, Cav1.3(-/-) SAN cells remained slower than WT. Conversely, we rescued pacemaker activity in Cav1.3(-/-) SAN cells and intact tissues through caffeine-mediated stimulation of Ca(2+)-induced Ca(2+) release., Conclusions: Cav1.3 channels play a critical role in the regulation of [Ca(2+)]i dynamics, providing an unanticipated mechanism for triggering local [Ca(2+)]i releases and thereby controlling pacemaker activity. Our study also provides an additional pathophysiological mechanism for congenital SAN dysfunction and heart block linked to Cav1.3 loss of function in humans., (Published on behalf of the European Society of Cardiology. All rights reserved. © The Author 2016. For permissions please email: journals.permissions@oup.com.)
- Published
- 2016
- Full Text
- View/download PDF
124. G protein-gated IKACh channels as therapeutic targets for treatment of sick sinus syndrome and heart block.
- Author
-
Mesirca P, Bidaud I, Briec F, Evain S, Torrente AG, Le Quang K, Leoni AL, Baudot M, Marger L, Chung You Chong A, Nargeot J, Striessnig J, Wickman K, Charpentier F, and Mangoni ME
- Subjects
- Animals, Calcium Channels, L-Type genetics, Calcium Channels, L-Type physiology, Humans, Mice, Mice, Knockout, Calcium Channels, L-Type drug effects, GTP-Binding Proteins physiology, Heart Block drug therapy, Ion Channel Gating physiology, Sick Sinus Syndrome drug therapy
- Abstract
Dysfunction of pacemaker activity in the sinoatrial node (SAN) underlies "sick sinus" syndrome (SSS), a common clinical condition characterized by abnormally low heart rate (bradycardia). If untreated, SSS carries potentially life-threatening symptoms, such as syncope and end-stage organ hypoperfusion. The only currently available therapy for SSS consists of electronic pacemaker implantation. Mice lacking L-type Cav1.3 Ca(2+) channels (Cav1.3(-/-)) recapitulate several symptoms of SSS in humans, including bradycardia and atrioventricular (AV) dysfunction (heart block). Here, we tested whether genetic ablation or pharmacological inhibition of the muscarinic-gated K(+) channel (IKACh) could rescue SSS and heart block in Cav1.3(-/-) mice. We found that genetic inactivation of IKACh abolished SSS symptoms in Cav1.3(-/-) mice without reducing the relative degree of heart rate regulation. Rescuing of SAN and AV dysfunction could be obtained also by pharmacological inhibition of IKACh either in Cav1.3(-/-) mice or following selective inhibition of Cav1.3-mediated L-type Ca(2+) (ICa,L) current in vivo. Ablation of IKACh prevented dysfunction of SAN pacemaker activity by allowing net inward current to flow during the diastolic depolarization phase under cholinergic activation. Our data suggest that patients affected by SSS and heart block may benefit from IKACh suppression achieved by gene therapy or selective pharmacological inhibition.
- Published
- 2016
- Full Text
- View/download PDF
125. Comment on: 'Homozygous knockout of the piezo1 gene in the zebrafish is not associated with anemia'.
- Author
-
Faucherre A, Kissa K, Nargeot J, Mangoni ME, and Jopling C
- Subjects
- Anemia, Animals, Homozygote, Humans, Anemia, Hemolytic, Congenital genetics, Zebrafish
- Published
- 2016
- Full Text
- View/download PDF
126. The Low-Threshold Calcium Channel Cav3.2 Determines Low-Threshold Mechanoreceptor Function.
- Author
-
François A, Schüetter N, Laffray S, Sanguesa J, Pizzoccaro A, Dubel S, Mantilleri A, Nargeot J, Noël J, Wood JN, Moqrich A, Pongs O, and Bourinet E
- Abstract
The T-type calcium channel Cav3.2 emerges as a key regulator of sensory functions, but its expression pattern within primary afferent neurons and its contribution to modality-specific signaling remain obscure. Here, we elucidate this issue using a unique knockin/flox mouse strain wherein Cav3.2 is replaced by a functional Cav3.2-surface-ecliptic GFP fusion. We demonstrate that Cav3.2 is a selective marker of two major low-threshold mechanoreceptors (LTMRs), Aδ- and C-LTMRs, innervating the most abundant skin hair follicles. The presence of Cav3.2 along LTMR-fiber trajectories is consistent with critical roles at multiple sites, setting their strong excitability. Strikingly, the C-LTMR-specific knockout uncovers that Cav3.2 regulates light-touch perception and noxious mechanical cold and chemical sensations and is essential to build up that debilitates allodynic symptoms of neuropathic pain, a mechanism thought to be entirely A-LTMR specific. Collectively, our findings support a fundamental role for Cav3.2 in touch/pain pathophysiology, validating their critic pharmacological relevance to relieve mechanical and cold allodynia., (Copyright © 2015 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
127. [The imminent peril in the law of July the fifth 2011, two years later: the impact on health?].
- Author
-
Mondoloni A, Buard M, Nargeot J, and Vacheron MN
- Subjects
- Emergency Services, Psychiatric legislation & jurisprudence, France, Hospitals, Psychiatric legislation & jurisprudence, Humans, Length of Stay legislation & jurisprudence, Mental Disorders diagnosis, Commitment of Mentally Ill legislation & jurisprudence, Dangerous Behavior, Mental Disorders therapy, Patient Admission legislation & jurisprudence, Patient Rights legislation & jurisprudence
- Abstract
In 1938, the French government decided to enact a first legislation to enforce admission of the mentally ill to hospitals. Later in 1990, the law took into consideration the evolution of practices with an increase of free admissions and the right to maintain the mentally ill in cities. Three types of psychiatric hospitalization were defined: free, on third party request and for involuntary confinement. A review had theoretically to be conducted every 5 years. In practice this was not the case, probably due to the balance between individual freedom, patient care and public safety always hard to find. However, considering the imperative European harmonization and the fact the Constitutional Council declared a double unconstitutionality of the law, the Act of July 5th was enacted in a hurry during the summer 2011. The Act defines the "rights and the protection of people subject to psychiatric care and methods of coverage". In this document, we will briefly review the context of this law. We will also explore the clinical implications of the very innovative measure: the "péril imminent". We will use the admissions at the Sainte-Anne hospital in Paris in 2010 to 2012. Three major key points were introduced in the law: a judge controls an agreeable release after 15 days and 6 months of continuous hospitalization. The law let the new possibility to provide ambulatory cares under constraints, and these to make an involuntary confinement without a third party request, using the "imminent peril". This law implies the involvement of the judge and the lawyer. This one has to defend a client who needs care, he controls the formal validity of decisions concerning the patient. To provide treatment without consent in "imminent peril" to someone, conditions are requested: these mental disorders make his consent impossible and his mental state requires immediate care with immediate care of constant medical monitoring justifying a full hospitalization or regular medical monitoring for support under another form of full hospitalization (Article L.3212. 1 of the Code of Public Health). Moreover, a demand for care by a third party has also to be impossible to obtain and an imminent peril to the person's health has to exist, supported by a medical certificate from a doctor who does not belong to the patient's psychiatric hospital. The imminent peril would be an immediate danger to the health or life of the patient. What has been the impact of this law adopted in emergency at Sainte-Anne hospital? This psychiatric hospital is in charge of the population in southern Paris, where reside about 655,000 people. This work observes the evolution of the type of hospitalization and care before and after the adoption of the law. We can observe an overall increase in entries under constraints. There is a decrease in admissions for involuntary confinement for the benefit of imminent peril. This imminent peril corresponds to only a small proportion of hospitalizations without consent but are rising between 2011 and 2012, perhaps in part due to a better understanding of the law. But this progression is to monitor to ensure compliance with the restrictive conditions laid down by this law. Also note that the imminent peril may be used at the refusal of the family or entourage to make the demand for care. The number of hospitalizations at the request of a third party with two certificates is down, which is probably due to a change in status of the CPOA, emergency structure within Sainte-Anne, which is no longer seen as extraterritorial. The imminent peril has advantages: it allows access to the care of people isolated and desocialized, of people whose identity is unknown, of pathological travellers. It avoids hospitalization at the request of the representative of the State for social reasons and not for risks to the safety of persons, even when this type of hospitalization is more stigmatizing and often more difficult to remove. It protects the entourage sometimes, when the family is ambivalent or hostile to care, or has been designated as a persecutor. The imminent peril also has disadvantages. One of them is the risk of its misuse to allow rapid hospitalization without taking the time to seek a third party. The imminent danger made when there is an entourage but which refuses to request care can undermine the development work on information about the disease, the need for care and treatment and the importance of the involvement of the entourage in the care plan. The alliance with the patient may be compromised. In some cases, a decision of care by the request of the representative of the State is more appropriate than the "imminent peril". The "imminent peril" may be preferred because of the administrative burden of prefectural measures when patient presents clinical improvement and we would go up to the ambulatory care in a care program. Yet, the use of a symbolic third, carrying authority, can avoid the too direct confrontation with the patient. Do not use it can complicate the management of the patient. Finally, with desocialized patients, imminent peril can facilitate access to care, but not continuity of care. Indeed, for the care program it is necessary to have an address for the patient. Once the crisis is not to develop a plan of care. Finally in some situations of desocialized patients, the imminent peril can promote access to care but not the continuity of care as to the care program it is necessary to have an address for the patient. Once the crisis is past, it is impossible to implement a program of care. The Law of 5 July 2011 marks a change in the practice of psychiatrists. Take into account the fundamental rights of the patient and to harmonize legislation at EU level was necessary. Some measures are designed to promote access to care as the "imminent peril", we now need to be vigilant to ensure that it is not diverted to promote an increase in care under constraints and that psychiatrists remain in an obligation of means and not of result., (Copyright © 2014 L’Encéphale, Paris. Published by Elsevier Masson SAS. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
128. Cardiac arrhythmia induced by genetic silencing of 'funny' (f) channels is rescued by GIRK4 inactivation.
- Author
-
Mesirca P, Alig J, Torrente AG, Müller JC, Marger L, Rollin A, Marquilly C, Vincent A, Dubel S, Bidaud I, Fernandez A, Seniuk A, Engeland B, Singh J, Miquerol L, Ehmke H, Eschenhagen T, Nargeot J, Wickman K, Isbrandt D, and Mangoni ME
- Subjects
- Animals, Arrhythmias, Cardiac drug therapy, Benzazepines pharmacology, Calcium Signaling genetics, Disease Models, Animal, Female, G Protein-Coupled Inwardly-Rectifying Potassium Channels genetics, Heart Rate drug effects, Humans, Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels metabolism, Ivabradine, Male, Mice, Mice, Inbred C57BL, Mice, Transgenic, Muscle Proteins metabolism, Myocytes, Cardiac metabolism, Myocytes, Cardiac pathology, Oocytes physiology, Patch-Clamp Techniques, Potassium Channels metabolism, Pregnancy, Xenopus, Arrhythmias, Cardiac genetics, Arrhythmias, Cardiac physiopathology, G Protein-Coupled Inwardly-Rectifying Potassium Channels metabolism, Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels genetics, Muscle Proteins genetics, Potassium Channels genetics
- Abstract
The mechanisms underlying cardiac automaticity are still incompletely understood and controversial. Here we report the complete conditional and time-controlled silencing of the 'funny' current (If) by expression of a dominant-negative, non-conductive HCN4-channel subunit (hHCN4-AYA). Heart-specific If silencing caused altered [Ca(2+)]i release and Ca(2+) handling in the sinoatrial node, impaired pacemaker activity and symptoms reminiscent of severe human disease of pacemaking. The effects of If silencing critically depended on the activity of the autonomic nervous system. We were able to rescue the failure of impulse generation and conduction by additional genetic deletion of cardiac muscarinic G-protein-activated (GIRK4) channels in If-deficient mice without impairing heartbeat regulation. Our study establishes the role of f-channels in cardiac automaticity and indicates that arrhythmia related to HCN loss-of-function may be managed by pharmacological or genetic inhibition of GIRK4 channels, thus offering a new therapeutic strategy for the treatment of heart rhythm diseases.
- Published
- 2014
- Full Text
- View/download PDF
129. [Antipsychotics-induced acute necrotizing enterocolitis: a case report].
- Author
-
Nargeot J, Langlet C, Merlot A, Buard M, Mondoloni A, Viala A, and Vacheron MN
- Subjects
- Adult, Alprazolam administration & dosage, Alprazolam adverse effects, Antipsychotic Agents administration & dosage, Delirium drug therapy, Dibenzothiazepines administration & dosage, Dibenzothiazepines adverse effects, Dissociative Disorders drug therapy, Estazolam administration & dosage, Estazolam adverse effects, Humans, Male, Phenothiazines administration & dosage, Phenothiazines adverse effects, Quetiapine Fumarate, Antipsychotic Agents adverse effects, Enterocolitis, Necrotizing chemically induced
- Abstract
The acute necrotizing enterocolitis (ANE) is a partial or total necrosis of the small and large intestine. This is a case report of an antipsychotic induced ANE., (© 2014 Société Française de Pharmacologie et de Thérapeutique.)
- Published
- 2014
- Full Text
- View/download PDF
130. Piezo1 plays a role in erythrocyte volume homeostasis.
- Author
-
Faucherre A, Kissa K, Nargeot J, Mangoni ME, and Jopling C
- Subjects
- Animals, Erythropoiesis genetics, Gene Expression Regulation, Developmental, Gene Knockdown Techniques, Ion Channels metabolism, Zebrafish embryology, Zebrafish Proteins metabolism, Erythrocyte Volume genetics, Homeostasis genetics, Ion Channels genetics, Zebrafish blood, Zebrafish genetics, Zebrafish Proteins genetics
- Abstract
Mechanosensitivity is an inherent property of virtually all cell types, allowing them to sense and respond to physical environmental stimuli. Stretch-activated ion channels represent a class of mechanosensitive proteins which allow cells to respond rapidly to changes in membrane tension; however their identity has remained elusive. The piezo genes have recently been identified as a family of stretch-activated mechanosensitive ion channels. We set out to determine the role of piezo1 during zebrafish development. Here we report that morpholino-mediated knockdown of piezo1 impairs erythrocyte survival without affecting hematopoiesis or differentiation. Our results demonstrate that piezo1 is involved in erythrocyte volume homeostasis, disruption of which results in swelling/lysis of red blood cells and consequent anemia.
- Published
- 2014
- Full Text
- View/download PDF
131. piezo2b regulates vertebrate light touch response.
- Author
-
Faucherre A, Nargeot J, Mangoni ME, and Jopling C
- Subjects
- Animals, Ion Channels antagonists & inhibitors, Ion Channels genetics, Morpholinos pharmacology, Neurons metabolism, Neurons physiology, Zebrafish, Zebrafish Proteins antagonists & inhibitors, Zebrafish Proteins genetics, Ion Channels metabolism, Touch, Zebrafish Proteins metabolism
- Abstract
The sense of touch allows an organism to detect and respond to physical environmental stimuli. Mechanosensitive proteins play a crucial role in this process by converting the mechanical cue into a biological response. Recently, the Piezo family of stretch-activated ion channels has been identified as genuine mechanosensitive proteins. We set out to determine whether any of these genes are involved in touch response during zebrafish development. In situ hybridization indicates that piezo2b is specifically expressed in a subset of neurons (Rohon-Beard cells) responsible for detecting light touch. Using morpholino-mediated knockdown, we specifically targeted piezo2b and determined that it is involved in mediating touch-evoked response.
- Published
- 2013
- Full Text
- View/download PDF
132. The G-protein-gated K+ channel, IKACh, is required for regulation of pacemaker activity and recovery of resting heart rate after sympathetic stimulation.
- Author
-
Mesirca P, Marger L, Toyoda F, Rizzetto R, Audoubert M, Dubel S, Torrente AG, Difrancesco ML, Muller JC, Leoni AL, Couette B, Nargeot J, Clapham DE, Wickman K, and Mangoni ME
- Subjects
- Action Potentials, Animals, Electrocardiography, G Protein-Coupled Inwardly-Rectifying Potassium Channels genetics, Mice, Mice, Inbred C57BL, Mice, Knockout, Myocardial Reperfusion, Myocytes, Cardiac drug effects, Myocytes, Cardiac metabolism, Myocytes, Cardiac physiology, Physical Exertion, Protein Subunits genetics, Protein Subunits metabolism, Sinoatrial Node cytology, Sinoatrial Node drug effects, Sinoatrial Node innervation, Stress, Physiological, Sympathetic Nervous System physiology, Acetylcholine pharmacology, G Protein-Coupled Inwardly-Rectifying Potassium Channels metabolism, Heart Rate, Sinoatrial Node physiology
- Abstract
Parasympathetic regulation of sinoatrial node (SAN) pacemaker activity modulates multiple ion channels to temper heart rate. The functional role of the G-protein-activated K(+) current (IKACh) in the control of SAN pacemaking and heart rate is not completely understood. We have investigated the functional consequences of loss of IKACh in cholinergic regulation of pacemaker activity of SAN cells and in heart rate control under physiological situations mimicking the fight or flight response. We used knockout mice with loss of function of the Girk4 (Kir3.4) gene (Girk4(-/-) mice), which codes for an integral subunit of the cardiac IKACh channel. SAN pacemaker cells from Girk4(-/-) mice completely lacked IKACh. Loss of IKACh strongly reduced cholinergic regulation of pacemaker activity of SAN cells and isolated intact hearts. Telemetric recordings of electrocardiograms of freely moving mice showed that heart rate measured over a 24-h recording period was moderately increased (10%) in Girk4(-/-) animals. Although the relative extent of heart rate regulation of Girk4(-/-) mice was similar to that of wild-type animals, recovery of resting heart rate after stress, physical exercise, or pharmacological β-adrenergic stimulation of SAN pacemaking was significantly delayed in Girk4(-/-) animals. We conclude that IKACh plays a critical role in the kinetics of heart rate recovery to resting levels after sympathetic stimulation or after direct β-adrenergic stimulation of pacemaker activity. Our study thus uncovers a novel role for IKACh in SAN physiology and heart rate regulation.
- Published
- 2013
- Full Text
- View/download PDF
133. Delayed postconditioning: not too late?
- Author
-
Barrère-Lemaire S, Nargeot J, and Piot C
- Subjects
- Animals, Humans, Mice, Time Factors, Ischemic Postconditioning, Myocardial Infarction therapy, Myocardial Reperfusion
- Abstract
Ischemic postconditioning applied at the onset of reperfusion reduces myocardial infarction in both animals and humans. Our recent study on the mouse myocardium showed for the first time that delayed postconditioning (applied up to 30 min after the onset of reperfusion) can decrease infarct size. The existence of a longer cardioprotection window is conceptually relevant for clinical application and also in the case of a pharmacological strategy., (Copyright © 2012 Elsevier Inc. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
134. Down-regulation of the transcription factor ZAC1 upon pre- and postconditioning protects against I/R injury in the mouse myocardium.
- Author
-
Vincent A, Gahide G, Sportouch-Dukhan C, Covinhes A, Franck-Miclo A, Roubille F, Barrère C, Adda J, Dantec C, Redt-Clouet C, Piot C, Nargeot J, and Barrère-Lemaire S
- Subjects
- Animals, Apoptosis, Cell Cycle Proteins genetics, Down-Regulation, Echocardiography, Genes, Tumor Suppressor, Mice, Mice, Inbred C57BL, Mice, Knockout, Myocardial Infarction genetics, Myocardial Reperfusion Injury genetics, Myocardial Reperfusion Injury prevention & control, Myocardium pathology, Oligonucleotide Array Sequence Analysis, RNA, Messenger metabolism, Real-Time Polymerase Chain Reaction, Transcription Factors genetics, Cell Cycle Proteins metabolism, Ischemic Postconditioning methods, Ischemic Preconditioning, Myocardial methods, Myocardial Infarction metabolism, Myocardial Reperfusion Injury metabolism, Myocardium metabolism, Transcription Factors metabolism
- Abstract
Aims: Myocardial infarction leads to heart failure and death. Ischaemic preconditioning (PreC) and postconditioning (PostC) reduce infarct size in animal models and human. Zac1 was identified as the only gene related to apoptosis and jointly down-regulated upon PreC and PostC. The aim of our study was to investigate the role of Zac1 down-regulation during ischaemia-reperfusion (I/R) in vivo., Methods and Results: C57BL/6 mice were submitted to myocardial I/R injury, PreC, or PostC protocols. QPCR and immunochemistry showed that Zac1 expression was down-regulated both at the transcriptional and the protein levels upon PreC and PostC. Zac1(-/-) Knockout mice (n = 7) developed smaller infarcts (54%) than Zac1(+/+) littermates (n = 8) and decreased apoptosis (61.7%) in the ischaemic part of the left ventricle during I/R (Zac1(-/-), n = 6 vs. Zac1(+/+), n = 7; P = 0.0012). Mutants showed under control conditions a decrease of 53.9% in mRNA of Daxx, a pro-apoptotic protein playing a key role in I/R injuries (4.81 ± 0.77, n = 4 Zac1(-/-) mice vs. 10.44 ± 3.5, n = 7 Zac1(+/+) mice; P = 0.0121)., Conclusion: Our study shows for the first time that Zac1 is down-regulated both at the transcriptional and protein levels upon PreC and PostC in wild-type mice. Moreover, inactivation of Zac1 in vivo is associated with a decreased amount of Daxx transcripts and, upon I/R injury, decreased infarct size and apoptosis. Altogether, our results show that Zac1 down-regulation plays a key role during cardioprotection against I/R injury and support the concept that cardioprotection regulates a network of interacting pro-apoptotic genes including Zac1 and Daxx.
- Published
- 2012
- Full Text
- View/download PDF
135. AKAP79 modulation of L-type channels involves disruption of intramolecular interactions in the CaV1.2 subunit.
- Author
-
Altier C, Dubel SJ, Barrere C, Jarvis SE, Stotz SC, Scott JD, Nargeot J, Zamponi GW, and Bourinet E
- Subjects
- Amino Acid Sequence, Animals, Calcium Channels, L-Type genetics, Cell Line, Transformed, Gene Deletion, Humans, Mice, Molecular Sequence Data, Mutation, Missense, Oocytes, Patch-Clamp Techniques, Proline metabolism, Proline-Rich Protein Domains, Protein Interaction Domains and Motifs, Protein Subunits genetics, Protein Transport genetics, Xenopus, A Kinase Anchor Proteins metabolism, Calcium Channels, L-Type chemistry, Calcium Channels, L-Type metabolism, Protein Subunits chemistry, Protein Subunits metabolism
- Abstract
L-type voltage gated calcium channels (VGCCs) interact with a variety of proteins that modulate both their function and localization. A-Kinase Anchoring Proteins (AKAPs) facilitate L-type calcium channel phosphorylation through β adrenergic stimulation. Our previous work indicated a role of neuronal AKAP79/150 in the membrane targeting of Ca(V)1.2 L-type calcium channels, which involved a proline rich domain (PRD) in the intracellular II-III loop of the channel.(1) Here, we show that mutation of proline 857 to alanine (P857A) into the PRD does not disrupt the AKAP79-induced increase in Ca(v)1.2 membrane expression. Furthermore, deletion of two other PRDs into the carboxy terminal domain of Ca(V)1.2 did not alter the targeting role of AKAP79. In contrast, the distal carboxy terminus region of the channel directly interacts with AKAP79. This protein-protein interaction competes with a direct association of the channel II-III linker on the carboxy terminal tail and modulates membrane targeting of Ca(V)1.2. Thus, our results suggest that the effects of AKAP79 occur through relief of an autoinhibitory mechanism mediated by intramolecular interactions of Ca(v)1.2 intracellular regions.
- Published
- 2012
- Full Text
- View/download PDF
136. The case for the funny current and the calcium clock.
- Author
-
Rosen MR, Nargeot J, and Salama G
- Subjects
- Humans, Calcium Channel Blockers therapeutic use, Calcium Channels drug effects, Cardiac Pacing, Artificial, Cyclic Nucleotide-Gated Cation Channels, Sinoatrial Node
- Published
- 2012
- Full Text
- View/download PDF
137. Systemic delivery of BH4 anti-apoptotic peptide using CPPs prevents cardiac ischemia-reperfusion injuries in vivo.
- Author
-
Boisguerin P, Redt-Clouet C, Franck-Miclo A, Licheheb S, Nargeot J, Barrère-Lemaire S, and Lebleu B
- Subjects
- Amino Acid Sequence, Animals, Cell Line, Cell-Penetrating Peptides chemistry, Cells, Cultured, Heart drug effects, Mice, Mice, Inbred C57BL, Molecular Sequence Data, Myocardial Reperfusion Injury pathology, Myocardium pathology, Myocytes, Cardiac drug effects, Myocytes, Cardiac pathology, Apoptosis drug effects, Myocardial Reperfusion Injury prevention & control, Peptides chemistry, Peptides therapeutic use, bcl-X Protein chemistry, bcl-X Protein therapeutic use
- Abstract
There is an obvious need to develop pharmacological strategies to protect the heart in patients suffering from acute myocardial infarction. Apoptosis was evidenced as a main contributor of myocardial ischemia-reperfusion (IR) injury. Our cardioprotective strategy was based on the use of four cell penetrating peptides (CPP: Tat, (RXR)4, Bpep and Pip2b) which were conjugated to the BH4-peptide, derived from the BH4 domain of the Bcl-xL anti-apoptotic protein. These CPP-BH4 conjugates were able to reduce staurosporine-induced apoptosis in primary cardiomyocytes in vitro. Although Pip2b-BH4 was more efficient in terms of cellular uptake, it was as efficient as Tat-BH4 for its anti-apoptotic activity. As required for potential therapeutic application their cardioprotective effects were evaluated in an in vivo mouse model of myocardial IR injury. Our results clearly show that a single low dose (1 mg/kg) injection of Tat-BH4 and Pip2b-BH4 administered intravenously 5 min before reperfusion was able to drastically reduce infarct size (~47%) and to inhibit apoptosis (~60%) in the left ventricle of treated mice. Importantly, these effects are not observed following the injection of CPP alone or scrambled version of BH4. This study evidences that the Pip2b CPP, designed for oligonucleotides translocation, as well as the widely used natural Tat CPP exhibit similar efficacy in vivo to deliver BH4 anti-apoptotic peptide to the reperfused myocardium and may thus become useful therapeutic tools to treat acute myocardial infarction in the clinical setting., (Copyright © 2011 Elsevier B.V. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
138. Delayed postconditioning in the mouse heart in vivo.
- Author
-
Roubille F, Franck-Miclo A, Covinhes A, Lafont C, Cransac F, Combes S, Vincent A, Fontanaud P, Sportouch-Dukhan C, Redt-Clouet C, Nargeot J, Piot C, and Barrère-Lemaire S
- Subjects
- Animals, Disease Models, Animal, Male, Mice, Mice, Inbred C57BL, Models, Biological, Myocardial Infarction pathology, Myocardial Infarction therapy, Myocardial Reperfusion, Myocardial Reperfusion Injury pathology, Myocardial Reperfusion Injury therapy, Myocardium pathology, Time Factors, Ischemic Postconditioning methods, Ischemic Preconditioning, Myocardial methods, Myocardial Infarction prevention & control, Myocardial Reperfusion Injury prevention & control
- Abstract
Background: Reperfusion during acute myocardial infarction remains the best treatment for reducing infarct size. Postconditioning, applied at the onset of reperfusion, reduces myocardial infarction both in animals and humans. The objective of this study was to identify the time delay to apply postconditioning at reperfusion, allowing preservation of cardioprotection in the mouse myocardium. This is a major issue in the management of acute myocardial infarction patients., Methods and Results: Mice were subjected to 40 minutes of ischemia and 60 minutes of reperfusion (IR(60')). Postconditioning protocols corresponding to repetitive ischemia (3 cycles of 1 minute of ischemia and 1 minute of reperfusion) were applied during early reperfusion at various time durations (Δt) after reopening of the coronary artery (Δt=10 seconds, 1, 5, 10, 15, 20, 30, and 45 minutes; PostC(Δt)). Infarct size/area at risk was reduced by 71% in PostC(Δ1) compared with IR(60') mice (P=5×10(-6)). There was a linear correlation (r(2)=0.91) between infarct size and Δt, indicating that the cardioprotective effect of delayed postconditioning was progressively attenuated when Δt time increased. The protective effect of PostC(Δ1) and PostC(Δ15) was still effective when the duration of reperfusion was prolonged to 24 hours (IR(24 hours); PostC(Δ1) and PostC(Δ15) versus IR(24 hours), P=0.001). Similar results were obtained for internucleosomal DNA fragmentation and lactate dehydrogenase release., Conclusions: This study in our in vivo mouse model of myocardial IR shows for the first time that delaying the intervention of postconditioning to 30 minutes does not abrogate the cardioprotective effect of postconditioning. This finding provides evidence that the time window of protection afforded by postconditioning may be larger than initially reported.
- Published
- 2011
- Full Text
- View/download PDF
139. T-type calcium channels contribute to colonic hypersensitivity in a rat model of irritable bowel syndrome.
- Author
-
Marger F, Gelot A, Alloui A, Matricon J, Ferrer JF, Barrère C, Pizzoccaro A, Muller E, Nargeot J, Snutch TP, Eschalier A, Bourinet E, and Ardid D
- Subjects
- Animals, Base Sequence, Butyrates toxicity, Calcium Channel Blockers pharmacology, Calcium Channels, T-Type deficiency, Calcium Channels, T-Type genetics, Disease Models, Animal, Electrophysiological Phenomena, Gene Knockdown Techniques, Irritable Bowel Syndrome chemically induced, Irritable Bowel Syndrome drug therapy, Male, Neuralgia drug therapy, Neuralgia physiopathology, Nociceptors physiology, Pain Perception physiology, RNA, Small Interfering genetics, Rats, Rats, Sprague-Dawley, Calcium Channels, T-Type physiology, Colon innervation, Colon physiopathology, Irritable Bowel Syndrome physiopathology
- Abstract
The symptoms of irritable bowel syndrome (IBS) include significant abdominal pain and bloating. Current treatments are empirical and often poorly efficacious, and there is a need for the development of new and efficient analgesics aimed at IBS patients. T-type calcium channels have previously been validated as a potential target to treat certain neuropathic pain pathologies. Here we report that T-type calcium channels encoded by the Ca(V)3.2 isoform are expressed in colonic nociceptive primary afferent neurons and that they contribute to the exaggerated pain perception in a butyrate-mediated rodent model of IBS. Both the selective genetic inhibition of Ca(V)3.2 channels and pharmacological blockade with calcium channel antagonists attenuates IBS-like painful symptoms. Mechanistically, butyrate acts to promote the increased insertion of Ca(V)3.2 channels into primary sensory neuron membranes, likely via a posttranslational effect. The butyrate-mediated regulation can be recapitulated with recombinant Ca(V)3.2 channels expressed in HEK cells and may provide a convenient in vitro screening system for the identification of T-type channel blockers relevant to visceral pain. These results implicate T-type calcium channels in the pathophysiology of chronic visceral pain and suggest Ca(V)3.2 as a promising target for the development of efficient analgesics for the visceral discomfort and pain associated with IBS.
- Published
- 2011
- Full Text
- View/download PDF
140. Functional roles of Ca(v)1.3, Ca(v)3.1 and HCN channels in automaticity of mouse atrioventricular cells: insights into the atrioventricular pacemaker mechanism.
- Author
-
Marger L, Mesirca P, Alig J, Torrente A, Dubel S, Engeland B, Kanani S, Fontanaud P, Striessnig J, Shin HS, Isbrandt D, Ehmke H, Nargeot J, and Mangoni ME
- Subjects
- Adrenergic beta-Agonists pharmacology, Animals, Arrhythmia, Sinus genetics, Arrhythmia, Sinus metabolism, Atrioventricular Node cytology, Biological Clocks drug effects, Calcium Channels, L-Type genetics, Calcium Channels, T-Type genetics, Cells, Cultured, Cyclic AMP genetics, Cyclic AMP metabolism, Cyclic Nucleotide-Gated Cation Channels genetics, Cyclic Nucleotide-Gated Cation Channels metabolism, Humans, Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels, Isoproterenol pharmacology, Mice, Mice, Knockout, Atrioventricular Node metabolism, Biological Clocks physiology, Calcium Channels, L-Type metabolism, Calcium Channels, T-Type metabolism
- Abstract
The atrioventricular node controls cardiac impulse conduction and generates pacemaker activity in case of failure of the sino-atrial node. Understanding the mechanisms of atrioventricular automaticity is important for managing human pathologies of heart rate and conduction. However, the physiology of atrioventricular automaticity is still poorly understood. We have investigated the role of three key ion channel-mediated pacemaker mechanisms namely, Ca(v)1.3, Ca(v)3.1 and HCN channels in automaticity of atrioventricular node cells (AVNCs). We studied atrioventricular conduction and pacemaking of AVNCs in wild-type mice and mice lacking Ca(v)3.1 (Ca(v)3.1(-/-)), Ca(v)1.3 (Ca(v)1.3(-/-)), channels or both (Ca(v)1.3(-/-)/Ca(v)3.1(-/-)). The role of HCN channels in the modulation of atrioventricular cells pacemaking was studied by conditional expression of dominant-negative HCN4 channels lacking cAMP sensitivity. Inactivation of Ca(v)3.1 channels impaired AVNCs pacemaker activity by favoring sporadic block of automaticity leading to cellular arrhythmia. Furthermore, Ca(v)3.1 channels were critical for AVNCs to reach high pacemaking rates under isoproterenol. Unexpectedly, Ca(v)1.3 channels were required for spontaneous automaticity, because Ca(v)1.3(-/-) and Ca(v)1.3(-/-)/Ca(v)3.1(-/-) AVNCs were completely silent under physiological conditions. Abolition of the cAMP sensitivity of HCN channels reduced automaticity under basal conditions, but maximal rates of AVNCs could be restored to that of control mice by isoproterenol. In conclusion, while Ca(v)1.3 channels are required for automaticity, Ca(v)3.1 channels are important for maximal pacing rates of mouse AVNCs. HCN channels are important for basal AVNCs automaticity but do not appear to be determinant for β-adrenergic regulation.
- Published
- 2011
- Full Text
- View/download PDF
141. Pacemaker activity and ionic currents in mouse atrioventricular node cells.
- Author
-
Marger L, Mesirca P, Alig J, Torrente A, Dubel S, Engeland B, Kanani S, Fontanaud P, Striessnig J, Shin HS, Isbrandt D, Ehmke H, Nargeot J, and Mangoni ME
- Subjects
- Animals, Atrioventricular Node cytology, Biological Clocks drug effects, Calcium Channel Blockers pharmacology, Cardiovascular Agents pharmacology, Drug Resistance drug effects, Drug Resistance physiology, Ion Transport drug effects, Ion Transport physiology, Isradipine pharmacology, Membrane Potentials drug effects, Mice, Mice, Transgenic, Myocardial Contraction drug effects, Myocytes, Cardiac cytology, Potassium Channels, Tandem Pore Domain antagonists & inhibitors, Potassium Channels, Tandem Pore Domain genetics, Potassium Channels, Tandem Pore Domain metabolism, Pyrimidines pharmacology, Sinoatrial Node cytology, Sinoatrial Node metabolism, Sodium Channel Blockers pharmacology, Tetrodotoxin pharmacology, Atrioventricular Node metabolism, Biological Clocks physiology, Membrane Potentials physiology, Myocardial Contraction physiology, Myocytes, Cardiac metabolism
- Abstract
It is well established that Pacemaker activity of the sino-atrial node (SAN) initiates the heartbeat. However, the atrioventricular node (AVN) can generate viable pacemaker activity in case of SAN failure, but we have limited knowledge of the ionic bases of AVN automaticity. We characterized pacemaker activity and ionic currents in automatic myocytes of the mouse AVN. Pacemaking of AVN cells (AVNCs) was lower than that of SAN pacemaker cells (SANCs), both in control conditions and upon perfusion of isoproterenol (ISO). Block of I(Na) by tetrodotoxin (TTX) or of I(Ca,L) by isradipine abolished AVNCs pacemaker activity. TTX-resistant (I(Nar)) and TTX-sensitive (I(Nas)) Na(+) currents were recorded in mouse AVNCs, as well as T-(I(Ca,T)) and L-type (I(Ca,L)) Ca(2+) currents I(Ca,L) density was lower than in SANCs (51%). The density of the hyperpolarization-activated current, (I(f)) and that of the fast component of the delayed rectifier current (I(Kr)) were, respectively, lower (52%) and higher (53%) in AVNCs than in SANCs. Pharmacological inhibition of I(f) by 3 µM ZD-7228 reduced pacemaker activity by 16%, suggesting a relevant role for I(f) in AVNCs automaticity. Some AVNCs expressed also moderate densities of the transient outward K(+) current (I(to)). In contrast, no detectable slow component of the delayed rectifier current (I(Ks)) could be recorded in AVNCs. The lower densities of I(f) and I(Ca,L), as well as higher expression of I(Kr) in AVNCs than in SANCs may contribute to the intrinsically slower AVNCs pacemaking than that of SANCs.
- Published
- 2011
- Full Text
- View/download PDF
142. Connexin 30 is expressed in the mouse sino-atrial node and modulates heart rate.
- Author
-
Gros D, Théveniau-Ruissy M, Bernard M, Calmels T, Kober F, Söhl G, Willecke K, Nargeot J, Jongsma HJ, and Mangoni ME
- Subjects
- Animals, Connexin 30, Connexins deficiency, Connexins genetics, Electrocardiography, Female, Male, Mice, Mice, Inbred C57BL, Myocardium metabolism, Rats, Rats, Wistar, Ventricular Remodeling, Connexins physiology, Heart Rate, Sinoatrial Node physiology
- Abstract
Aims: This study aimed at characterizing expression and the functional role of the Gjb6 gene, encoding for connexin 30 (Cx30) protein, in the adult mouse heart., Methods and Results: The expression of the Gjb6 gene in the mouse heart was investigated by RT-PCR and sequencing of amplified cDNA fragments. The sites of Gjb6 expression were identified in the adult heart using transgenic mice with reporter genes (Cx30(LacZ/LacZ) and Cx30(LacZ/LacZ)/Cx40(EGFP/EGFP) mice), as well as anti-HCN4 (hyperpolarization activated cyclic nucleotide-gated potassium channel 4) or anti-connexin antibodies. Cine-magnetic resonance imaging and telemetric ECG recordings were used to evaluate the impact of Cx30 deficiency on cardiac physiology. Gjb6 was shown to be expressed in the sinoatrial (SA) node of the adult mouse heart. Eighty from 100 nuclei on average were LacZ-positive in the SA node of Cx30(LacZ/LacZ) mice. No significant LacZ expression was seen in other cardiac tissues. Cx30 protein was identified in low abundance in the SA node of wild-type mice, as indicated by immunofluorescence experiments. Telemetric ECG recordings indicated that Cx30-deficient mice displayed a mean daily heart rate (HR) that was 9% faster than that measured in control mice (572 +/- 38 b.p.m. vs. 524 +/- 23, P < 0.05). This moderate tachycardia was still observed after inhibition of the autonomic nervous system, demonstrating that Cx30 deficiency resulted in changes in the intrinsic electrical properties of the SA node. Consistent with this hypothesis, Cx30(LacZ/LacZ) displayed a significant reduction of SDNN (standard deviation of the interbeat interval) compared with control mice. Increase of both the cardiac index (20%) and the end-diastolic volume to body weight ratio (16%) with no deficiency in ejection fraction or stroke volume were observed in mutant mice. An increase in cardiac index was interpreted as being a direct consequence of high HR, whereas large end-diastolic volume may be an indirect consequence of prolonged high HR., Conclusion: Cx30 is functionally expressed, in low abundance, in the SA node of the adult mouse heart where it participates in HR regulation.
- Published
- 2010
- Full Text
- View/download PDF
143. The NALCN ion channel is a new actor in pancreatic β-cell physiology.
- Author
-
Swayne LA, Mezghrani A, Lory P, Nargeot J, and Monteil A
- Subjects
- Animals, Cell Line, Humans, Insulin-Secreting Cells metabolism, Ion Channels metabolism, Membrane Proteins, Mice, Nerve Tissue Proteins metabolism, Rodentia, Insulin-Secreting Cells physiology, Ion Channels physiology, Nerve Tissue Proteins physiology
- Abstract
Ion channels are critical components of cell excitability involved in many physiological processes, including hormone secretion, and are thought be targets of choice in a pathological context. In the present paper, we summarize and discuss our recent findings on a four domain cation channel named NALCN which has been previously described as mediating a TTX-resistant leak sodium current in neurons. We recently reported that NALCN is also expressed in rodent islets of Langerhans as well as in the mouse MIN6 pancreatic β-cell line. This pancreatic NALCN channel encodes for a cation current triggered by acetylcholine activation of M3 muscarinic receptors. Importantly, the activation mechanism is independent of G protein action, but is dependent on a SFK-pathway, and involves the co-inclusion of M3 muscarinic receptors and NALCN in the same complex. Although additional work is now needed, considering the importance of the cholinergic control on the pancreatic β-cell function, this study has unravelled the molecular identity of a new actor in pancreatic β-cell excitability that could be a major target for new compounds modulating insulin secretion.
- Published
- 2010
- Full Text
- View/download PDF
144. T-type calcium channel inhibition underlies the analgesic effects of the endogenous lipoamino acids.
- Author
-
Barbara G, Alloui A, Nargeot J, Lory P, Eschalier A, Bourinet E, and Chemin J
- Subjects
- Animals, Behavior, Animal physiology, Calcium metabolism, Calcium Channel Blockers pharmacology, Calcium Channels, L-Type genetics, Calcium Channels, T-Type classification, Calcium Channels, T-Type genetics, Cells, Cultured, Disease Models, Animal, Electric Stimulation methods, Ganglia, Spinal cytology, Glycine pharmacology, Green Fluorescent Proteins genetics, Humans, Hyperalgesia drug therapy, Hyperalgesia genetics, Male, Membrane Potentials drug effects, Membrane Potentials genetics, Mice, Mice, Inbred C57BL, Mice, Knockout, Morphine pharmacology, NAV1.7 Voltage-Gated Sodium Channel, Nerve Tissue Proteins genetics, Neuroblastoma, Patch-Clamp Techniques methods, Potassium Channels, Tandem Pore Domain genetics, Sensory Receptor Cells, Sodium Channels genetics, TRPV Cation Channels genetics, Transfection, gamma-Aminobutyric Acid pharmacology, Analgesics pharmacology, Arachidonic Acids pharmacology, Calcium Channels, T-Type metabolism, Glycine analogs & derivatives, gamma-Aminobutyric Acid analogs & derivatives
- Abstract
Lipoamino acids are anandamide-related endogenous molecules that induce analgesia via unresolved mechanisms. Here, we provide evidence that the T-type/Cav3 calcium channels are important pharmacological targets underlying their physiological effects. Various lipoamino acids, including N-arachidonoyl glycine (NAGly), reversibly inhibited Cav3.1, Cav3.2, and Cav3.3 currents, with potent effects on Cav3.2 [EC(50) approximately 200 nm for N-arachidonoyl 3-OH-gamma-aminobutyric acid (NAGABA-OH)]. This inhibition involved a large shift in the Cav3.2 steady-state inactivation and persisted during fatty acid amide hydrolase (FAAH) inhibition as well as in cell-free outside-out patch. In contrast, lipoamino acids had weak effects on high-voltage-activated (HVA) Cav1.2 and Cav2.2 calcium currents, on Nav1.7 and Nav1.8 sodium currents, and on anandamide-sensitive TRPV1 and TASK1 currents. Accordingly, lipoamino acids strongly inhibited native Cav3.2 currents in sensory neurons with small effects on sodium and HVA calcium currents. In addition, we demonstrate here that lipoamino acids NAGly and NAGABA-OH produced a strong thermal analgesia and that these effects (but not those of morphine) were abolished in Cav3.2 knock-out mice. Collectively, our data revealed lipoamino acids as a family of endogenous T-type channel inhibitors, suggesting that these ligands can modulate multiple cell functions via this newly evidenced regulation.
- Published
- 2009
- Full Text
- View/download PDF
145. The NALCN ion channel is activated by M3 muscarinic receptors in a pancreatic beta-cell line.
- Author
-
Swayne LA, Mezghrani A, Varrault A, Chemin J, Bertrand G, Dalle S, Bourinet E, Lory P, Miller RJ, Nargeot J, and Monteil A
- Subjects
- Blotting, Western, Cell Line, Electrophysiology, Enzyme Inhibitors pharmacology, Humans, Insulin-Secreting Cells drug effects, Ion Channels genetics, Patch-Clamp Techniques, Pyrazoles pharmacology, Pyrimidines pharmacology, RNA Interference, RNA, Small Interfering, Receptor, Muscarinic M3 genetics, Reverse Transcriptase Polymerase Chain Reaction, Insulin-Secreting Cells metabolism, Ion Channels metabolism, Receptor, Muscarinic M3 metabolism
- Abstract
A previously uncharacterized putative ion channel, NALCN (sodium leak channel, non-selective), has been recently shown to be responsible for the tetrodotoxin (TTX)-resistant sodium leak current implicated in the regulation of neuronal excitability. Here, we show that NALCN encodes a current that is activated by M3 muscarinic receptors (M3R) in a pancreatic beta-cell line. This current is primarily permeant to sodium ions, independent of intracellular calcium stores and G proteins but dependent on Src activation, and resistant to TTX. The current is recapitulated by co-expression of NALCN and M3R in human embryonic kidney-293 cells and in Xenopus oocytes. We also show that NALCN and M3R belong to the same protein complex, involving the intracellular I-II loop of NALCN and the intracellular i3 loop of M3R. Taken together, our data show the molecular basis of a muscarinic-activated inward sodium current that is independent of G-protein activation, and provide new insights into the properties of NALCN channels.
- Published
- 2009
- Full Text
- View/download PDF
146. Identification of potential pharmacological targets by analysis of the comprehensive G protein-coupled receptor repertoire in the four cardiac chambers.
- Author
-
Moore-Morris T, Varrault A, Mangoni ME, Le Digarcher A, Negre V, Dantec C, Journot L, Nargeot J, and Couette B
- Subjects
- Animals, Blotting, Western, Calcium metabolism, Gene Expression Profiling, Glycine analogs & derivatives, Glycine pharmacology, Male, Mice, Mice, Inbred C57BL, RNA, Messenger analysis, Receptors, Metabotropic Glutamate genetics, Resorcinols pharmacology, Reverse Transcriptase Polymerase Chain Reaction, Myocardium metabolism, Receptors, G-Protein-Coupled genetics
- Abstract
Cardiac function is regulated by many hormones and neurotransmitters that exert their physiological effects through the activation of G protein-coupled receptors (GPCRs). Identification of new GPCRs that might display a specific pattern of expression within the heart and differentially regulate specific cardiac functions represents an important issue for the development of new drugs. Indeed, highly targeted receptors represent only a small percentage of known GPCRs. Here, we quantified the expression of 395 endoGPCRs (all GPCRs excluding taste and odorant receptors) in male mouse right and left atria and ventricles by using high-throughput real-time reverse-transcriptase polymerase chain reaction (RT-PCR) and focused on the 135 most highly expressed transcripts. No cardiac functional data are available for almost half of these receptors; therefore, linking GPCR expression patterns to cardiac function has allowed us to provide new insights into the possible function of some of these receptors. Indeed, ventricles and atria are both contractile; however, the latter, and especially the right atrium, are central to the generation and regulation of cardiac rhythm. Accordingly, the right atrium exhibited the most specific signature, whereas the majority of GPCRs found in ventricles were evenly expressed in both the right and left chambers. RT-PCR data were confirmed at the protein level for six selected transcripts. Furthermore, we provide new data showing that, as suggested by our repertoire, the metabotropic glutamate receptor 1b is expressed and is functional in ventricular cardiac myocytes. This is the first report describing GPCRs in the four cardiac chambers and may assist in the identification of therapeutic targets.
- Published
- 2009
- Full Text
- View/download PDF
147. Genesis and regulation of the heart automaticity.
- Author
-
Mangoni ME and Nargeot J
- Subjects
- Action Potentials, Animals, Arrhythmias, Cardiac metabolism, Arrhythmias, Cardiac physiopathology, Arrhythmias, Cardiac therapy, Calcium Signaling, Gene Expression Regulation, Humans, Ion Channels genetics, Mice, Models, Cardiovascular, Autonomic Nervous System metabolism, Heart innervation, Heart Conduction System metabolism, Heart Rate genetics, Ion Channels metabolism, Periodicity
- Abstract
The heart automaticity is a fundamental physiological function in higher organisms. The spontaneous activity is initiated by specialized populations of cardiac cells generating periodical electrical oscillations. The exact cascade of steps initiating the pacemaker cycle in automatic cells has not yet been entirely elucidated. Nevertheless, ion channels and intracellular Ca(2+) signaling are necessary for the proper setting of the pacemaker mechanism. Here, we review the current knowledge on the cellular mechanisms underlying the generation and regulation of cardiac automaticity. We discuss evidence on the functional role of different families of ion channels in cardiac pacemaking and review recent results obtained on genetically engineered mouse strains displaying dysfunction in heart automaticity. Beside ion channels, intracellular Ca(2+) release has been indicated as an important mechanism for promoting automaticity at rest as well as for acceleration of the heart rate under sympathetic nerve input. The potential links between the activity of ion channels and Ca(2+) release will be discussed with the aim to propose an integrated framework of the mechanism of automaticity.
- Published
- 2008
- Full Text
- View/download PDF
148. Myocardial expression of a dominant-negative form of Daxx decreases infarct size and attenuates apoptosis in an in vivo mouse model of ischemia/reperfusion injury.
- Author
-
Roubille F, Combes S, Leal-Sanchez J, Barrère C, Cransac F, Sportouch-Dukhan C, Gahide G, Serre I, Kupfer E, Richard S, Hueber AO, Nargeot J, Piot C, and Barrère-Lemaire S
- Subjects
- Acute Disease, Animals, Carrier Proteins genetics, Caspase 3 metabolism, Caspase 8 metabolism, Chronic Disease, Co-Repressor Proteins, Disease Models, Animal, Heart Failure genetics, Heart Failure metabolism, Heart Failure pathology, Intracellular Signaling Peptides and Proteins genetics, Mice, Mice, Transgenic, Mitogen-Activated Protein Kinase 1 metabolism, Mitogen-Activated Protein Kinase 3 metabolism, Molecular Chaperones, Myocardial Infarction genetics, Myocardial Infarction pathology, Myocardial Reperfusion Injury genetics, Myocardial Reperfusion Injury pathology, Nuclear Proteins genetics, Apoptosis, Carrier Proteins metabolism, Genes, Dominant, Intracellular Signaling Peptides and Proteins metabolism, Myocardial Infarction metabolism, Myocardial Reperfusion Injury metabolism, Nuclear Proteins metabolism, Signal Transduction
- Abstract
Background: Apoptosis has been described extensively in acute myocardial infarction and chronic heart failure. Because Daxx (death-associated protein) appears to be essential for stress-induced cell death and acts as an antisurvival molecule, we tested the hypothesis that Daxx is involved in myocardial ischemia/reperfusion-induced cell death in vivo., Methods and Results: Transgenic mice overexpressing a dominant-negative form of Daxx (Daxx-DN) under the control of the beta-actin promoter and control wild-type mice underwent an ischemia/reperfusion protocol: 40 minutes of left coronary artery occlusion and 60 minutes of reperfusion. Area at risk and infarct size were measured after dual staining by triphenyltetrazolium chloride and phthalocyanine blue dye. Apoptosis was measured in the ischemic versus the nonischemic part of the left ventricle by terminal deoxynucleotidyl transferase-mediated dUTP biotin nick end labeling staining, enzyme-linked immunosorbent assay, and Western blotting of caspase-3, caspase-8, and poly(ADP-ribose) polymerase. The mitogen-activated protein kinase status was investigated by Western blot analysis. Comparison between groups was assessed by ANOVA or Student t test (statistical significance: P<0.05). Left ventricle tissues from transgenic mice expressed Daxx-DN at the protein level. Area at risk/left ventricle values were comparable among groups. Infarct size/area at risk was 45% reduced in Daxx-DN versus wild-type mice (P<0.001). This cardioprotection was maintained for a 4-hour reperfusion. Ischemia/reperfusion-induced apoptosis was significantly decreased and ERK1/2 prosurvival pathway was activated in ischemic Daxx-DN hearts., Conclusions: Our study clearly indicates that Daxx participates in myocardial ischemia/reperfusion proapoptotic signaling in vivo.
- Published
- 2007
- Full Text
- View/download PDF
149. Temperature-dependent modulation of CaV3 T-type calcium channels by protein kinases C and A in mammalian cells.
- Author
-
Chemin J, Mezghrani A, Bidaud I, Dupasquier S, Marger F, Barrère C, Nargeot J, and Lory P
- Subjects
- Animals, Calcium Channels, T-Type genetics, Cell Line, Cricetinae, Electrophysiology, Patch-Clamp Techniques, Protein Transport, Tetradecanoylphorbol Acetate analogs & derivatives, Tetradecanoylphorbol Acetate pharmacology, Calcium Channels, T-Type metabolism, Cyclic AMP-Dependent Protein Kinases metabolism, Protein Kinase C metabolism, Temperature
- Abstract
Modulation of low voltage-activated Ca(V)3 T-type calcium channels remains poorly characterized compared with high voltage-activated Ca(V)1 and Ca(V)2 calcium channels. Notably, it is yet unresolved whether Ca(V)3 channels are modulated by protein kinases in mammalian cells. In this study, we demonstrate that protein kinase A (PKA) and PKC (but not PKG) activation induces a potent increase in Ca(V)3.1, Ca(V)3.2, and Ca(V)3.3 currents in various mammalian cell lines. Notably, we show that protein kinase effects occur at physiological temperature ( approximately 30-37 degrees C) but not at room temperature ( approximately 22-27 degrees C). This temperature dependence could involve kinase translocation, which is impaired at room temperature. A similar temperature dependence was observed for PKC-mediated increase in high voltage-activated Ca(V)2.3 currents. We also report that neither Ca(V)3 surface expression nor T-current macroscopic properties are modified upon kinase activation. In addition, we provide evidence for the direct phosphorylation of Ca(V)3.2 channels by PKA in in vitro assays. Overall, our results clearly establish the role of PKA and PKC in the modulation of Ca(V)3 T-channels and further highlight the key role of the physiological temperature in the effects described.
- Published
- 2007
- Full Text
- View/download PDF
150. Ligand-based virtual screening to identify new T-type calcium channel blockers.
- Author
-
Ijjaali I, Barrere C, Nargeot J, Petitet F, and Bourinet E
- Subjects
- Animals, Calcium Channels, T-Type chemistry, Calcium Channels, T-Type genetics, Calcium Channels, T-Type metabolism, Calcium Channels, T-Type pharmacology, Cell Line, Databases, Factual, Humans, Ligands, Membrane Potentials, Molecular Structure, Neurons metabolism, Reproducibility of Results, Structure-Activity Relationship, Transfection, Calcium Channels, T-Type drug effects, Computer-Aided Design, Drug Design, Neurons drug effects, User-Computer Interface
- Abstract
T-type calcium channels are involved in the generation of rhythmical firing patterns in the mammalian central nervous system and in various pathological alterations of neuronal excitability such as in epilepsy or neuropathic pain. In the search for new T-type calcium channel blockers that would help to treat these disorders, we have followed a bi-dimensional pharmacophore-based virtual screening approach to identify new inhibitors. Nineteen molecules extracted from AurSCOPE Ion Channels knowledgebase were used as query molecules to screen an external database. This in silico approach was then validated using electrophysiology. Interestingly, 16 compounds out of 38 distinct molecules tested showed more than 50% blockade of the Ca(V)3.2 mediated T-type current. Two series of compounds show chemical originality compared with known T-type calcium channel blockers.
- Published
- 2007
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.