38 results on '"Percherancier Y"'
Search Results
2. Real-time study of the effects of radiofrequency fields at the cellular and molecular levels: the results of the Geronimo project
- Author
-
Ruigrok, Hermanus, Poque-Haro, Emmanuelle, Arnaud-Cormos, D., Renom, Rémy, Hurtier, A., Poulletier De Gannes, F., Lagroye, I, Bernard, Veyret, Lévêque, Philippe, Percherancier, Y., Laboratoire de l'intégration, du matériau au système (IMS), Université Sciences et Technologies - Bordeaux 1-Institut Polytechnique de Bordeaux-Centre National de la Recherche Scientifique (CNRS), BIO-INGENIERIE (XLIM-BIO-INGENIERIE), XLIM (XLIM), Université de Limoges (UNILIM)-Centre National de la Recherche Scientifique (CNRS)-Université de Limoges (UNILIM)-Centre National de la Recherche Scientifique (CNRS), Institut Universitaire de France (IUF), Ministère de l'Education nationale, de l’Enseignement supérieur et de la Recherche (M.E.N.E.S.R.), and Leveque, Philippe
- Subjects
[SPI.ELEC]Engineering Sciences [physics]/Electromagnetism ,[SPI.ELEC] Engineering Sciences [physics]/Electromagnetism ,ComputingMilieux_MISCELLANEOUS - Abstract
International audience
- Published
- 2019
3. Delivery system setup and characterization for biological cells exposed to nanosecond pulsed electric field
- Author
-
Arnaud-Cormos, D., primary, Soueid, M., additional, O'Connor, R., additional, Percherancier, Y., additional, Lagroye, I., additional, Veyret, B., additional, and Leveque, P., additional
- Published
- 2015
- Full Text
- View/download PDF
4. Therapeutic applications of EMF and rTMS in cancer and pain management
- Author
-
Charlet De Sauvage, R., Percherancier, Y., Taxile, M., Haro, E., Hurtier, A., Lagroye, I., Veyret, B., Taxile, Murielle, Laboratoire de l'intégration, du matériau au système (IMS), Université Sciences et Technologies - Bordeaux 1-Institut Polytechnique de Bordeaux-Centre National de la Recherche Scientifique (CNRS), Laboratoire de Bioélectromagnétisme, École pratique des hautes études (EPHE), and Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-École Nationale Supérieure de Chimie et de Physique de Bordeaux (ENSCPB)
- Subjects
[SDV.BBM.BP]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Biophysics ,[SDV.TOX] Life Sciences [q-bio]/Toxicology ,[SDV.TOX]Life Sciences [q-bio]/Toxicology ,[SDV.BBM.BP] Life Sciences [q-bio]/Biochemistry, Molecular Biology/Biophysics ,ComputingMethodologies_GENERAL - Abstract
Poster
- Published
- 2010
5. Palmitoylation-dependent control of degradation, life span, and membrane expression of the CCR5 receptor.
- Author
-
Percherancier, Y, Planchenault, T, Valenzuela-Fernandez, A, Virelizier, J L, Arenzana-Seisdedos, F, and Bachelerie, F
- Abstract
We have shown that the chemokine and HIV receptor CCR5 is palmitoylated on a cluster of cysteine residues located at the boundary between the seventh transmembrane region and the cytoplasmic tail. Single or combined substitutions of the three cysteines (Cys-321, Cys-323, and Cys-324) or incubation of wild-type CCR5-transfected cells with the palmitic acid analog 2-bromopalmitate prevented palmitoylation of the receptor. Moreover, failure of CCR5 to be palmitoylated resulted in both accumulation in intracellular stores and a profound decrease of membrane expression of the receptor. Upon metabolic labeling, kinetic experiments showed that the half-life of palmitoylation-deficient CCR5 is profoundly decreased. Bafilomycin A1, but not a specific proteasome inhibitor, prevented early degradation of palmitoylation-deficient CCR5 and promoted its accumulation in lysosomal compartments. Although membrane expression of the CCR5 mutant was diminished, the molecules reaching the membrane were still able to interact efficiently with the chemokine ligand MIP1 beta and remained able to function as HIV co-receptors. Thus we conclude that palmitoylation controls CCR5 expression through regulation of the life span of this receptor.
- Published
- 2001
- Full Text
- View/download PDF
6. Inducible NF-kappaB activation is permitted by simultaneous degradation of nuclear IkappaBalpha.
- Author
-
Renard, P, Percherancier, Y, Kroll, M, Thomas, D, Virelizier, J L, Arenzana-Seisdedos, F, and Bachelerie, F
- Abstract
Signal-induced phosphorylation and ubiquitination of IkappaBalpha targets this inhibitor of NF-kappaB for proteasome-mediated degradation, thus permitting the release of active NF-kappaB. Upon cell stimulation, NF-kappaB activation results in neotranscription and neosynthesis of its own inhibitor, IkappaBalpha. As reported earlier, the neosynthesized inhibitor is then accumulated in the nucleus, where it rapidly binds to and terminates the function of nuclear NF-kappaB upon withdrawal of the stimulus. The present work was aimed at understanding how NF-kappaB activity is preserved while stimuli persist, despite intense, simultaneous IkappaBalpha neosynthesis, which would be expected to end NF-kappaB activity. We here show that incoming IkappaBalpha in the nucleus represents a target for resident nuclear proteasome complexes. Signal-induced, proteasome-dependent degradation of phosphorylated and ubiquitinated IkappaBalpha occurs in the nucleus, thus permitting the onset and persistence of NF-kappaB activity as long as stimulation is maintained. Our results suggest that intranuclear proteolysis of IkappaBalpha is necessarily required to avoid self-termination of NF-kappaB activity during cell activation.
- Published
- 2000
7. Monitoring protein-protein interactions in living cells by bioluminescence resonance energy transfer (BRET)
- Author
-
Hamdan, F. F., Percherancier, Y., Breton, B., and Michel Bouvier
8. Evaluation of mitochondrial stress following ultraviolet radiation and 5G radiofrequency field exposure in human skin cells.
- Author
-
Patrignoni L, Hurtier A, Orlacchio R, Joushomme A, Poulletier de Gannes F, Lévêque P, Arnaud-Cormos D, Revzani HR, Mahfouf W, Garenne A, Percherancier Y, and Lagroye I
- Subjects
- Humans, Reactive Oxygen Species metabolism, Radio Waves adverse effects, Keratinocytes metabolism, Electromagnetic Fields, Ultraviolet Rays adverse effects, Skin metabolism
- Abstract
Whether human cells are impacted by environmental electromagnetic fields (EMF) is still a matter of debate. With the deployment of the fifth generation (5G) of mobile communication technologies, the carrier frequency is increasing and the human skin becomes the main biological target. Here, we evaluated the impact of 5G-modulated 3.5 GHz radiofrequency (RF) EMF on mitochondrial stress in human fibroblasts and keratinocytes that were exposed for 24 h at specific absorption rate of 0.25, 1, and 4 W/kg. We assessed cell viability, mitochondrial reactive oxygen species (ROS) production, and membrane polarization. Knowing that human skin is the main target of environmental ultraviolet (UV), using the same read-out, we investigated whether subsequent exposure to 5G signal could alter the capacity of UV-B to damage skin cells. We found a statistically significant reduction in mitochondrial ROS concentration in fibroblasts exposed to 5G signal at 1 W/kg. On the contrary, the RF exposure slightly but statistically significantly enhanced the effects of UV-B radiation specifically in keratinocytes at 0.25 and 1 W/kg. No effect was found on mitochondrial membrane potential or apoptosis in any cell types or exposure conditions suggesting that the type and amplitude of the observed effects are very punctual., (© 2023 Bioelectromagnetics Society.)
- Published
- 2024
- Full Text
- View/download PDF
9. In vitro exposure of neuronal networks to the 5G-3.5 GHz signal.
- Author
-
Canovi A, Orlacchio R, Poulletier de Gannes F, Lévêque P, Arnaud-Cormos D, Lagroye I, Garenne A, Percherancier Y, and Lewis N
- Subjects
- Heating, Neurons
- Abstract
Introduction: The current deployment of the fifth generation (5G) of wireless communications raises new questions about the potential health effects of exposure to radiofrequency (RF) fields. So far, most of the established biological effects of RF have been known to be caused by heating. We previously reported inhibition of the spontaneous electrical activity of neuronal networks in vitro when exposed to 1.8 GHz signals at specific absorption rates (SAR) well above the guidelines. The present study aimed to assess the effects of RF fields at 3.5 GHz, one of the frequencies related to 5G, on neuronal activity in-vitro. Potential differences in the effects elicited by continuous-wave (CW) and 5G-modulated signals were also investigated., Methods: Spontaneous activity of neuronal cultures from embryonic cortices was recorded using 60-electrode multi-electrode arrays (MEAs) between 17 and 27 days in vitro. The neuronal cultures were subjected to 15 min RF exposures at SAR of 1, 3, and 28 W/kg., Results: At SAR close to the guidelines (1 and 3 W/kg), we found no conclusive evidence that 3.5 GHz RF exposure impacts the activity of neurons in vitro. On the contrary, CW and 5G-modulated signals elicited a clear decrease in bursting and total firing rates during RF exposure at high SAR levels (28 W/kg). Our experimental findings extend our previous results, showing that RF, at 1.8 to 3.5 GHz, inhibits the electrical activity of neurons in vitro at levels above environmental standards., 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 © 2023 Canovi, Orlacchio, Poulletier de Gannes, Lévêque, Arnaud-Cormos, Lagroye, Garenne, Percherancier and Lewis.)
- Published
- 2023
- Full Text
- View/download PDF
10. Effects of 5G-modulated 3.5 GHz radiofrequency field exposures on HSF1, RAS, ERK, and PML activation in live fibroblasts and keratinocytes cells.
- Author
-
Joushomme A, Orlacchio R, Patrignoni L, Canovi A, Chappe YL, Poulletier De Gannes F, Hurtier A, Garenne A, Lagroye I, Moisan F, Cario M, Lévêque P, Arnaud-Cormos D, and Percherancier Y
- Subjects
- Humans, Electromagnetic Fields adverse effects, Extracellular Signal-Regulated MAP Kinases, Fibroblasts, Keratinocytes
- Abstract
The potential health risks of exposure to radiofrequency electromagnetic fields from mobile communications technologies have raised societal concerns. Guidelines have been set to protect the population (e.g. non-specific heating above 1 °C under exposure to radiofrequency fields), but questions remain regarding the potential biological effects of non-thermal exposures. With the advent of the fifth generation (5G) of mobile communication, assessing whether exposure to this new signal induces a cellular stress response is one of the mandatory steps on the roadmap for a safe deployment and health risk evaluation. Using the BRET (Bioluminescence Resonance Energy-Transfer) technique, we assessed whether continuous or intermittent (5 min ON/ 10 min OFF) exposure of live human keratinocytes and fibroblasts cells to 5G 3.5 GHz signals at specific absorption rate (SAR) up to 4 W/kg for 24 h impact basal or chemically-induced activity of Heat Shock Factor (HSF), RAt Sarcoma virus (RAS) and Extracellular signal-Regulated Kinases (ERK) kinases, and Promyelocytic Leukemia Protein (PML), that are all molecular pathways involved in environmental cell-stress responses. The main results are (i), a decrease of the HSF1 basal BRET signal when fibroblasts cells were exposed at the lower SARs tested (0.25 and 1 W/kg), but not at the highest one (4 W/kg), and (ii) a slight decrease of As
2 O3 maximal efficacy to trigger PML SUMOylation when fibroblasts cells, but not keratinocytes, were continuously exposed to the 5G RF-EMF signal. Nevertheless, given the inconsistency of these effects in terms of impacted cell type, effective SAR, exposure mode, and molecular cell stress response, we concluded that our study show no conclusive evidence that molecular effects can arise when skin cells are exposed to the 5G RF-EMF alone or with a chemical stressor., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
11. Genetically-encoded BRET probes shed light on ligand bias-induced variable ion selectivity in TRPV1 and P2X5/7.
- Author
-
Chappe YL, Pierredon S, Joushomme A, Molle P, Garenne A, Canovi A, Barbeau S, Poulletier De Gannes F, Hurtier A, Lagroye I, Ducret T, Quignard JF, Compan V, and Percherancier Y
- Subjects
- TRPV Cation Channels metabolism, Ligands, Capsaicin pharmacology, Energy Transfer, Bias, Transient Receptor Potential Channels metabolism
- Abstract
Whether ion channels experience ligand-dependent dynamic ion selectivity remains of critical importance since this could support ion channel functional bias. Tracking selective ion permeability through ion channels, however, remains challenging even with patch-clamp electrophysiology. In this study, we have developed highly sensitive bioluminescence resonance energy transfer (BRET) probes providing dynamic measurements of Ca
2+ and K+ concentrations and ionic strength in the nanoenvironment of Transient Receptor Potential Vanilloid-1 Channel (TRPV1) and P2X channel pores in real time and in live cells during drug challenges. Our results indicate that AMG517, BCTC, and AMG21629, three well-known TRPV1 inhibitors, more potently inhibit the capsaicin (CAPS)-induced Ca2+ influx than the CAPS-induced K+ efflux through TRPV1. Even more strikingly, we found that AMG517, when injected alone, is a partial agonist of the K+ efflux through TRPV1 and triggers TRPV1-dependent cell membrane hyperpolarization. In a further effort to exemplify ligand bias in other families of cationic channels, using the same BRET-based strategy, we also detected concentration- and time-dependent ligand biases in P2X7 and P2X5 cationic selectivity when activated by benzoyl-adenosine triphosphate (Bz-ATP). These custom-engineered BRET-based probes now open up avenues for adding value to ion-channel drug discovery platforms by taking ligand bias into account.- Published
- 2022
- Full Text
- View/download PDF
12. Comparative study between radiofrequency-induced and muscimol-induced inhibition of cultured networks of cortical neuron.
- Author
-
Lemercier CE, Garenne A, Poulletier de Gannes F, El Khoueiry C, Arnaud-Cormos D, Levêque P, Lagroye I, Percherancier Y, and Lewis N
- Subjects
- Action Potentials physiology, Muscimol pharmacology, Radio Waves, Neurons physiology, Synaptic Transmission physiology
- Abstract
Previous studies have shown that spontaneously active cultured networks of cortical neuron grown planar microelectrode arrays are sensitive to radiofrequency (RF) fields and exhibit an inhibitory response more pronounced as the exposure time and power increase. To better understand the mechanism behind the observed effects, we aimed at identifying similarities and differences between the inhibitory effect of RF fields (continuous wave, 1800 MHz) to the γ-aminobutyric acid type A (GABAA) receptor agonist muscimol (MU). Inhibition of the network bursting activity in response to RF exposure became apparent at an SAR level of 28.6 W/kg and co-occurred with an elevation of the culture medium temperature of ~1°C. Exposure to RF fields preferentially inhibits bursting over spiking activity and exerts fewer constraints on neural network bursting synchrony, differentiating it from a pharmacological inhibition with MU. Network rebound excitation, a phenomenon relying on the intrinsic properties of cortical neurons, was observed following the removal of tonic hyperpolarization after washout of MU but not in response to cessation of RF exposure. This implies that hyperpolarization is not the main driving force mediating the inhibitory effects of RF fields. At the level of single neurons, network inhibition induced by MU and RF fields occurred with reduced action potential (AP) half-width. As changes in AP waveform strongly influence efficacy of synaptic transmission, the narrowing effect on AP seen under RF exposure might contribute to reducing network bursting activity. By pointing only to a partial overlap between the inhibitory hallmarks of these two forms of inhibition, our data suggest that the inhibitory mechanisms of the action of RF fields differ from the ones mediated by the activation of GABAA receptors., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2022
- Full Text
- View/download PDF
13. Cell Confluence Modulates TRPV4 Channel Activity in Response to Hypoxia.
- Author
-
Barbeau S, Joushomme A, Chappe Y, Cardouat G, Baudrimont I, Freund-Michel V, Guibert C, Marthan R, Berger P, Vacher P, Percherancier Y, Quignard JF, and Ducret T
- Subjects
- Calcium metabolism, HEK293 Cells, Humans, Hypoxia metabolism, Patch-Clamp Techniques, TRPV Cation Channels genetics, TRPV Cation Channels metabolism, Transient Receptor Potential Channels
- Abstract
Transient receptor potential vanilloid 4 (TRPV4) is a polymodal Ca
2+ -permeable channel involved in various hypoxia-sensitive pathophysiological phenomena. Different tools are available to study channel activity, requiring cells to be cultured at specific optimal densities. In the present study, we examined if cell density may influence the effect of hypoxia on TRPV4 activity. Transiently TRPV4-transfected HEK293T cells were seeded at low or high densities corresponding to non-confluent or confluent cells, respectively, on the day of experiments, and cultured under in vitro normoxia or hypoxia. TRPV4-mediated cytosolic Ca2+ responses, single-channel currents, and Ca2+ influx through the channel were measured using Ca2+ imaging/microspectrofluorimetric assay, patch-clamp, and Bioluminescence Resonance Energy Transfer (BRET), respectively. TRPV4 plasma membrane translocation was studied using confocal microscopy, biotinylation of cell surface proteins, and BRET. Our results show that hypoxia exposure has a differential effect on TRPV4 activation depending on cell confluence. At low confluence levels, TRPV4 response is increased in hypoxia, whereas at high confluence levels, TRPV4 response is strongly inhibited, due to channel internalization. Thus, cell density appears to be a crucial parameter for TRPV4 channel activity.- Published
- 2022
- Full Text
- View/download PDF
14. In Vivo Functional Ultrasound (fUS) Real-Time Imaging and Dosimetry of Mice Brain Under Radiofrequency Exposure.
- Author
-
Orlacchio R, Percherancier Y, Poulletier De Gannes F, Hurtier A, Lagroye I, Leveque P, and Arnaud-Cormos D
- Subjects
- Animals, Brain diagnostic imaging, Mice, Radiometry, Cell Phone, Radio Waves
- Abstract
This study aims to analyze in real-time the potential modifications induced by low-level continuous-wave and Global System for Mobile Communications radiofrequency (RF) exposure at 1.8 GHz on brain activation in anesthetized mice. A specific in vivo experimental setup consisting of a dipole antenna for the local exposure of the brain was fully characterized. A unique neuroimaging technique based on a functional ultrasound (fUS) probe was used to observe the areas of mice brain activation simultaneously to the RF exposure with unprecedented spatial and temporal resolution (~100 μm, 1 ms) following manual whisker stimulation using a brush. Numerical and experimental dosimetry was carried out to characterize the exposure and to guarantee the validity of the biological results. Our results show that the fUS probe can be efficiently used during in vivo exposure without interference with the dipole. In addition, we conclude that exposure to brain-averaged specific absorption rate levels of 2 and 6 W/kg does not introduce significant changes in the time course of the evoked fUS response in the left barrel field cortex. The proposed technique represents a valuable instrument for providing new insights into the possible effects induced on brain activation under RF exposure. For the first time, brain activity under mobile phone exposure was evaluated in vivo with fUS imaging, paving the way for more realistic exposure configurations, i.e. awake mice and new signals such as the 5 G networks. © 2022 Bioelectromagnetics Society., (© 2022 Bioelectromagnetics Society.)
- Published
- 2022
- Full Text
- View/download PDF
15. Label-Free Study of the Global Cell Behavior during Exposure to Environmental Radiofrequency Fields in the Presence or Absence of Pro-Apoptotic or Pro-Autophagic Treatments.
- Author
-
Joushomme A, Garenne A, Dufossée M, Renom R, Ruigrok HJ, Chappe YL, Canovi A, Patrignoni L, Hurtier A, Poulletier de Gannes F, Lagroye I, Lévêque P, Lewis N, Priault M, Arnaud-Cormos D, and Percherancier Y
- Subjects
- Arsenic Trioxide pharmacology, Astrocytes drug effects, Astrocytes pathology, Cell Line, Tumor, Culture Media, Serum-Free, Electric Impedance, Holography, Humans, Neurons drug effects, Neurons pathology, Time Factors, Apoptosis, Autophagy drug effects, Radio Waves, Staining and Labeling
- Abstract
It remains controversial whether exposure to environmental radiofrequency signals (RF) impacts cell status or response to cellular stress such as apoptosis or autophagy. We used two label-free techniques, cellular impedancemetry and Digital Holographic Microscopy (DHM), to assess the overall cellular response during RF exposure alone, or during co-exposure to RF and chemical treatments known to induce either apoptosis or autophagy. Two human cell lines (SH-SY5Y and HCT116) and two cultures of primary rat cortex cells (astrocytes and co-culture of neurons and glial cells) were exposed to RF using an 1800 MHz carrier wave modulated with various environmental signals (GSM: Global System for Mobile Communications, 2G signal), UMTS (Universal Mobile Telecommunications System, 3G signal), LTE (Long-Term Evolution, 4G signal, and Wi-Fi) or unmodulated RF (continuous wave, CW). The specific absorption rates (S.A.R.) used were 1.5 and 6 W/kg during DHM experiments and ranged from 5 to 24 W/kg during the recording of cellular impedance. Cells were continuously exposed for three to five consecutive days while the temporal phenotypic signature of cells behavior was recorded at constant temperature. Statistical analysis of the results does not indicate that RF-EMF exposure impacted the global behavior of healthy, apoptotic, or autophagic cells, even at S.A.R. levels higher than the guidelines, provided that the temperature was kept constant.
- Published
- 2022
- Full Text
- View/download PDF
16. High-Throughput Screening of Transient Receptor Potential Channel 1 Ligands in the Light of the Bioluminescence Resonance Energy Transfer Technique.
- Author
-
Chappe Y, Michel P, Joushomme A, Barbeau S, Pierredon S, Baron L, Garenne A, Poulletier De Gannes F, Hurtier A, Mayer S, Lagroye I, Quignard JF, Ducret T, Compan V, Franchet C, and Percherancier Y
- Subjects
- Biological Assay methods, Calcium chemistry, Calmodulin antagonists & inhibitors, HEK293 Cells, Humans, Ligands, Membrane Potentials drug effects, Patch-Clamp Techniques, Small Molecule Libraries, TRPV Cation Channels agonists, TRPV Cation Channels antagonists & inhibitors, Bioluminescence Resonance Energy Transfer Techniques methods, Drug Discovery methods, Drug Evaluation, Preclinical methods, High-Throughput Screening Assays methods, TRPV Cation Channels metabolism
- Abstract
Ion channels are attractive drug targets for many therapeutic applications. However, high-throughput screening (HTS) of drug candidates is difficult and remains very expensive. We thus assessed the suitability of the bioluminescence resonance energy transfer (BRET) technique as a new HTS method for ion-channel studies by taking advantage of our recently characterized intra- and intermolecular BRET probes targeting the transient receptor potential vanilloid type 1 (TRPV1) ion channel. These BRET probes monitor conformational changes during TRPV1 gating and subsequent coupling with calmodulin, two molecular events that are intractable using reference techniques such as automated calcium assay (ACA) and automated patch-clamp (APC). We screened the small-sized Prestwick chemical library, encompassing 1200 compounds with high structural diversity, using either intra- and intermolecular BRET probes or ACA. Secondary screening of the detected hits was done using APC. Multiparametric analysis of our results shed light on the capability of calmodulin inhibitors included in the Prestwick library to inhibit TRPV1 activation by capsaicin. BRET was the lead technique for this identification process. Finally, we present data exemplifying the use of intramolecular BRET probes to study other transient receptor potential (TRP) channels and non-TRPs ion channels. Knowing the ease of use of BRET biosensors and the low cost of the BRET technique, these assays may advantageously be included for extending ion-channel drug screening. SIGNIFICANCE STATEMENT: This study screened a chemical library against TRPV1 ion channel using bioluminescence resonance energy transfer (BRET) molecular probes and compared the results with the ones obtained using reference techniques such as automated calcium assay and automated patch-clamp. Multiparametric analysis of our results shed light on the capability of calmodulin antagonists to inhibit chemical activation of TRPV1 and indicates that BRET probes may advantageously be included in ion channel drug screening campaigns., Competing Interests: The authors declare that there is no conflict of interest., (Copyright © 2021 by The American Society for Pharmacology and Experimental Therapeutics.)
- Published
- 2021
- Full Text
- View/download PDF
17. Effects of radiofrequency field exposure on proteotoxic-induced and heat-induced HSF1 response in live cells using the bioluminescence resonance energy transfer technique.
- Author
-
Poque E, Ruigrok HJ, Arnaud-Cormos D, Habauzit D, Chappe Y, Martin C, De Gannes FP, Hurtier A, Garenne A, Lagroye I, Le Dréan Y, Lévêque P, and Percherancier Y
- Subjects
- Energy Transfer, HEK293 Cells, Heat Shock Transcription Factors analysis, Humans, Luminescent Measurements, Heat Shock Transcription Factors metabolism, Heat-Shock Response, Radio Waves adverse effects
- Abstract
As of today, only acute effects of RF fields have been confirmed to represent a potential health hazard and they are attributed to non-specific heating (≥ 1 °C) under high-level exposure. Yet, the possibility that environmental RF impact living matter in the absence of temperature elevation needs further investigation. Since HSF1 is both a thermosensor and the master regulator of heat-shock stress response in eukaryotes, it remains to assess HSF1 activation in live cells under exposure to low-level RF signals. We thus measured basal, temperature-induced, and chemically induced HSF1 trimerization, a mandatory step on the cascade of HSF1 activation, under RF exposure to continuous wave (CW), Global System for Mobile (GSM), and Wi-Fi-modulated 1800 MHz signals, using a bioluminescence resonance energy transfer technique (BRET) probe. Our results show that, as expected, HSF1 is heat-activated by acute exposure of transiently transfected HEK293T cells to a CW RF field at a specific absorption rate of 24 W/kg for 30 min. However, we found no evidence of HSF1 activation under the same RF exposure condition when the cell culture medium temperature was fixed. We also found no experimental evidence that, at a fixed temperature, chronic RF exposure for 24 h at a SAR of 1.5 and 6 W/kg altered the potency or the maximal capability of the proteasome inhibitor MG132 to activate HSF1, whatever signal used. We only found that RF exposure to CW signals (1.5 and 6 W/kg) and GSM signals (1.5 W/kg) for 24 h marginally decreased basal HSF1 activity.
- Published
- 2021
- Full Text
- View/download PDF
18. Search for tumor-specific frequencies of amplitude-modulated 27 MHz electromagnetic fields in mice with hepatocarcinoma xenografted tumors: there are none so blind as those who will not read.
- Author
-
Veyret B, Arnaud-Cormos D, Poulletier de Gannes F, Lagroye I, Leveque P, and Percherancier Y
- Subjects
- Animals, Electromagnetic Fields, Mice, Carcinoma, Hepatocellular therapy, Liver Neoplasms therapy
- Published
- 2020
- Full Text
- View/download PDF
19. Effects of radiofrequency fields on RAS and ERK kinases activity in live cells using the bioluminescence resonance energy transfer technique.
- Author
-
Poque E, Arnaud-Cormos D, Patrignoni L, Ruigrok HJ, Poulletier De Gannes F, Hurtier A, Renom R, Garenne A, Lagroye I, Lévêque P, and Percherancier Y
- Subjects
- Cell Line, Tumor, Cell Survival radiation effects, Humans, Energy Transfer, Extracellular Signal-Regulated MAP Kinases metabolism, Luminescence, Radio Waves, ras Proteins metabolism
- Abstract
Purpose: The present study was conducted to re-evaluate the effect of low-level 1800 MHz RF signals on RAS/MAPK activation in live cells. Material and methods: Using Bioluminescence Resonance Energy Transfer technique (BRET), we assessed the effect of Continuous wave (CW) and Global System for Mobile (GSM)-modulated 1800 MHz signals (up to 2 W/kg) on ERK and RAS kinases' activity in live HuH7 cells. Results: We found that radiofrequency field (RF) exposure for 24 h altered neither basal level of RAS and ERK activation nor the potency of phorbol-12-myristate-13-acetate (PMA) to activate RAS and ERK kinases. However, we found that exposure to GSM-modulated 1800 MHz signals at 2 W/kg decreased the PMA maximal efficacy to activate both RAS and ERK kinases' activity. Exposure with CW 1800 MHz signal at 2 W/kg only decreased maximal efficacy of PMA to activate ERK but not RAS. No effects of RF exposure at 0.5 W/kg was observed on maximal efficacy of PMA to activate either RAS or ERK whatever the signal used. Conclusions: Our results indicate that RF exposure decreases the efficiency of the cascade of events, which, from the binding of PMA to its receptor(s), leads to the activation of RAS and ERK kinases.
- Published
- 2020
- Full Text
- View/download PDF
20. Activation of the TRPV1 Thermoreceptor Induced by Modulated or Unmodulated 1800 MHz Radiofrequency Field Exposure.
- Author
-
Ruigrok HJ, Arnaud-Cormos D, Hurtier A, Poque E, de Gannes FP, Ruffié G, Bonnaudin F, Lagroye I, Sojic N, Arbault S, Lévêque P, Veyret B, and Percherancier Y
- Subjects
- Calmodulin metabolism, Capsaicin pharmacology, HEK293 Cells, Humans, Thermoreceptors drug effects, Radio Waves adverse effects, TRPV Cation Channels metabolism, Thermoreceptors metabolism, Thermoreceptors radiation effects
- Abstract
The existence of effects of radiofrequency field exposure at environmental levels on living tissues and organisms remains controversial, in particular regarding potential "nonthermal" effects produced in the absence of temperature elevation. Therefore, we investigated whether TRPV1, one of the most studied thermosensitive channels, can be activated by the heat produced by radiofrequency fields and by some specific nonthermal interaction with the fields. We have recently shown that TRPV1 activation can be assessed in real-time on live cells using the bioluminescence resonance energy transfer technique. Taking advantage of this innovative assay, we monitored TRPV1 thermal and chemical modes of activation under radiofrequency exposure at 1800 MHz using different signals (CW, GSM, UMTS, LTE, Wi-Fi and WiMAX) at specific absorption rates between 8 and 32 W/kg. We showed that, as expected, TRPV1 channels were activated by the heat produced by radiofrequency field exposure of transiently-transfected HEK293T cells, but found no evidence of TRPV1 activation in the absence of temperature elevation under radiofrequency field exposure. There was no evidence either that, at fixed temperature, radiofrequency exposure altered the maximal efficacy of the agonist Capsaicin to activate TRPV1.
- Published
- 2018
- Full Text
- View/download PDF
21. Full-Spectral Multiplexing of Bioluminescence Resonance Energy Transfer in Three TRPV Channels.
- Author
-
Ruigrok HJ, Shahid G, Goudeau B, Poulletier de Gannes F, Poque-Haro E, Hurtier A, Lagroye I, Vacher P, Arbault S, Sojic N, Veyret B, and Percherancier Y
- Subjects
- Biosensing Techniques, Calmodulin metabolism, HEK293 Cells, Hot Temperature, Humans, Energy Transfer, Luminescent Measurements, TRPV Cation Channels chemistry, TRPV Cation Channels metabolism
- Abstract
Multiplexed bioluminescence resonance energy transfer (BRET) assays were developed to monitor the activation of several functional transient receptor potential (TRP) channels in live cells and in real time. We probed both TRPV1 intramolecular rearrangements and its interaction with Calmodulin (CaM) under activation by chemical agonists and temperature. Our BRET study also confirmed that: (1) capsaicin and heat promoted distinct transitions, independently coupled to channel gating, and that (2) TRPV1 and Ca
2+ -bound CaM but not Ca2+ -free CaM were preassociated in resting live cells, while capsaicin activation induced both the formation of more TRPV1/CaM complexes and conformational changes. The BRET assay, based on the interaction with Calmodulin, was successfully extended to TRPV3 and TRPV4 channels. We therefore developed a full-spectral three-color BRET assay for analyzing the specific activation of each of the three TRPV channels in a single sample. Such key improvement in BRET measurement paves the way for the simultaneous monitoring of independent biological pathways in live cells., (Copyright © 2017 Biophysical Society. Published by Elsevier Inc. All rights reserved.)- Published
- 2017
- Full Text
- View/download PDF
22. Dosimetric Characteristics of an EMF Delivery System Based on a Real-Time Impedance Measurement Device.
- Author
-
Garcia-Fernandez MA, Percherancier Y, Lagroye I, O'Connor RP, Veyret B, Arnaud-Cormos D, and Leveque P
- Subjects
- Equipment Design, Computer Simulation, Electric Impedance, Models, Biological, Radiometry instrumentation, Radiometry methods
- Abstract
In this paper, the dosimetric characterization of an EMF exposure setup compatible with real-time impedance measurements of adherent biological cells is proposed. The EMF are directly delivered to the 16-well format plate used by the commercial xCELLigence apparatus. Experiments and numerical simulations were carried out for the dosimetric analysis. The reflection coefficient was less than -10 dB up to 180 MHz and this exposure system can be matched at higher frequencies up to 900 and 1800 MHz. The specific absorption rate (SAR) distribution within the wells containing the biological medium was calculated by numerical finite-difference time domain simulations and results were verified by temperature measurements at 13.56 MHz. Numerical SAR values were obtained at the microelectrode level where the biological cells were exposed to EMF including 13.56, 900, and 1800 MHz. At 13.56 MHz, the SAR values, within the cell layer and the 270-μL volume of medium, are 1.9e3 and 3.5 W/kg/incident mW, respectively.
- Published
- 2016
- Full Text
- View/download PDF
23. Effects of 50 Hz magnetic fields on gap junctional intercellular communication in NIH3T3 cells.
- Author
-
Percherancier Y, Goudeau B, Charlet de Sauvage R, de Gannes FP, Haro E, Hurtier A, Sojic N, Lagroye I, Arbault S, and Veyret B
- Subjects
- Animals, Fluorescent Dyes metabolism, Kinetics, Mice, NIH 3T3 Cells, Cell Communication, Gap Junctions, Magnetic Fields
- Abstract
The present study focused on gap junctional intercellular communication (GJIC) as a target for biological effects of extremely low-frequency (ELF) magnetic field (MF) exposure. Fluorescence recovery after photobleaching microscopy (FRAP) was used to visualize diffusion of a fluorescent dye between NIH3T3 fibroblasts through gap junctions. The direct effect of 24 h exposure to 50 Hz MF at 0.4 or 1 mT on GJIC function was assessed in one series of experiments. The potential synergism of MF with an inhibitor of GJIC, phorbol ester (TPA), was studied in another series by observing FRAP when NIH3T3 cells were incubated with TPA for 1 h following 24 h exposure to MF. In contrast to other reports of ELF-MF effects on GJIC, under our experimental conditions we observed neither direct inhibition of GJIC nor synergism with TPA-induced inhibition from 50 Hz MF exposures., (© 2015 Wiley Periodicals, Inc.)
- Published
- 2015
- Full Text
- View/download PDF
24. The chemokine CXC4 and CC2 receptors form homo- and heterooligomers that can engage their signaling G-protein effectors and βarrestin.
- Author
-
Armando S, Quoyer J, Lukashova V, Maiga A, Percherancier Y, Heveker N, Pin JP, Prézeau L, and Bouvier M
- Subjects
- HEK293 Cells, Humans, Protein Binding, Protein Multimerization, Receptors, CXCR4 genetics, beta-Arrestins, Arrestins metabolism, Chemokine CCL2 metabolism, GTP-Binding Protein alpha Subunits metabolism, Receptors, CCR2 metabolism, Receptors, CXCR4 metabolism, Signal Transduction
- Abstract
G-protein-coupled receptors have been shown to assemble at least as dimers early in the biosynthetic path, but some evidence suggests that they can also form larger oligomeric complexes. Using the human chemokine receptors CXCR4 and CCR2 as models, we directly probed the existence of higher order homo- and heterooligomers in human embryonic kidney cells. Combining bimolecular fluorescence and luminescence complementation (BiFC, BiLC) with bioluminescence resonance energy transfer (BRET) assays, we show that CXCR4 and CCR2 can assemble as homo- and heterooligomers, forming at least tetramers. Selective activation of CCR2 with the human monocyte chemotactic protein 1 (MCP-1) resulted in trans-conformational rearrangement of the CXCR4 dimer with an EC50 of 19.9 nM, compatible with a CCR2 action. Moreover, MCP-1 promoted the engagement of Gαi1, Gα13, Gαz, and βarrestin2 to the heterooligomer, resulting in calcium signaling that was synergistically potentiated on coactivation of CCR2 and CXCR4, demonstrating that complexes larger than dimers reach the cell surface as functional units. A mutation of CXCR4 (N119K), which prevents Gi activation, also affects the CCR2-promoted engagement of Gαi1 and βarrestin2 by the heterooligomer, supporting the occurrence of transprotomer regulation. Together, the results demonstrate that homo- and heteromultimeric CXCR4 and CCR2 can form functional signaling complexes that have unique properties., (© FASEB.)
- Published
- 2014
- Full Text
- View/download PDF
25. CNIH4 interacts with newly synthesized GPCR and controls their export from the endoplasmic reticulum.
- Author
-
Sauvageau E, Rochdi MD, Oueslati M, Hamdan FF, Percherancier Y, Simpson JC, Pepperkok R, and Bouvier M
- Subjects
- Animals, COS Cells, Chlorocebus aethiops, Gene Knockdown Techniques, HEK293 Cells, Humans, Proteasome Endopeptidase Complex metabolism, Protein Binding, Protein Transport, Receptors, Cytoplasmic and Nuclear genetics, Endoplasmic Reticulum metabolism, Receptors, Cytoplasmic and Nuclear metabolism, Receptors, G-Protein-Coupled metabolism
- Abstract
The molecular mechanisms regulating G protein-coupled receptors (GPCRs) trafficking from their site of synthesis in the endoplasmic reticulum (ER) to their site of function (the cell surface) remain poorly characterized. Using a bioluminescence resonance energy transfer-based proteomic screen, we identified a novel GPCR-interacting protein; the human cornichon homologue 4 (CNIH4). This previously uncharacterized protein is localized in the early secretory pathway where it interacts with members of the 3 family of GPCRs. Both overexpression and knockdown expression of CNIH4 caused the intracellular retention of GPCRs, indicating that this ER-resident protein plays an important role in GPCR export. Overexpression of CNIH4 at low levels rescued the maturation and cell surface expression of an intracellularly retained mutant form of the β2-adrenergic receptor, further demonstrating a positive role of CNIH4 in GPCR trafficking. Taken with the co-immunoprecipitation of CNIH4 with Sec23 and Sec24, components of the COPII coat complex responsible for ER export, these data suggest that CNIH4 acts as a cargo-sorting receptor, recruiting GPCRs into COPII vesicles., (© 2014 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd.)
- Published
- 2014
- Full Text
- View/download PDF
26. Rat fertility and embryo fetal development: influence of exposure to the Wi-Fi signal.
- Author
-
Poulletier de Gannes F, Billaudel B, Haro E, Taxile M, Le Montagner L, Hurtier A, Ait Aissa S, Masuda H, Percherancier Y, Ruffié G, Dufour P, Veyret B, and Lagroye I
- Subjects
- Animals, Dose-Response Relationship, Radiation, Embryo Implantation radiation effects, Embryo Loss etiology, Energy Intake radiation effects, Female, Genitalia, Male growth & development, Genitalia, Male immunology, Genitalia, Male radiation effects, Male, Maternal Exposure adverse effects, Organ Size radiation effects, Ovary growth & development, Ovary immunology, Ovary radiation effects, Paternal Exposure adverse effects, Random Allocation, Rats, Rats, Wistar, Embryonic Development radiation effects, Fetal Development radiation effects, Infertility, Female etiology, Infertility, Male etiology, Radio Waves adverse effects, Sexual Maturation radiation effects, Wireless Technology
- Abstract
In recent decades, concern has been growing about decreasing fecundity and fertility in the human population. Exposure to non-ionizing electromagnetic fields (EMF), especially radiofrequency (RF) fields used in wireless communications has been suggested as a potential risk factor. For the first time, we evaluated the effects of exposure to the 2450MHz Wi-Fi signal (1h/day, 6days/week) on the reproductive system of male and female Wistar rats, pre-exposed to Wi-Fi during sexual maturation. Exposure lasted 3 weeks (males) or 2 weeks (females), then animals were mated and couples exposed for 3 more weeks. On the day before delivery, the fetuses were observed for lethality, abnormalities, and clinical signs. In our experiment, no deleterious effects of Wi-Fi exposure on rat male and female reproductive organs and fertility were observed for 1h per days. No macroscopic abnormalities in fetuses were noted, even at the critical level of 4W/kg., (Copyright © 2012 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
27. ELF magnetic fields: animal studies, mechanisms of action.
- Author
-
Lagroye I, Percherancier Y, Juutilainen J, De Gannes FP, and Veyret B
- Subjects
- Animals, Biological Assay, Free Radicals metabolism, Humans, Neoplasms etiology, Teratology, Magnetic Fields adverse effects, Models, Animal
- Abstract
Animal studies can contribute to addressing the issue of possible greater health risk for children exposed to 50-60 Hz extremely low frequency (ELF) magnetic fields (MFs), mostly in terms of teratological effects and cancer. Teratology has been extensively studied in animals exposed to ELF MFs but experiments have not established adverse developmental effects. Childhood leukaemia has been the only cancer consistently reported in epidemiological studies as associated with exposure to ELF MFs. This association has been the basis for the classification as "possibly carcinogenic to humans" by the International Agency for Research on Cancer in 2002. Animal experiments have provided only limited support for these epidemiological findings. However, none but one study used an animal model for acute lymphoblastic leukaemia (ALL), the main form of childhood leukaemia, and exposures to ELF MFs were not carried out over the whole pregnancy period, when the first hit of ALL is assumed to occur. Moreover, there are no generally accepted biophysical mechanisms that could explain carcinogenic effects of low-level MFs. The radical pair mechanism and related cryptochromes (CRY) molecules have recently been identified in birds and other non-mammalian species, as a sensor of the geomagnetic field, involved in navigation. The hypothesis has to be tested in mammalian models. CRY, which is part of the molecular circadian clock machinery, is a ubiquitous protein likely to be involved in cancer cell growth and DNA repair. In summary, we now have some clues to test for a better characterization of the interaction between ALL and ELF MFs exposure., (Copyright © 2011 Elsevier Ltd. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
28. Role of SUMO in RNF4-mediated promyelocytic leukemia protein (PML) degradation: sumoylation of PML and phospho-switch control of its SUMO binding domain dissected in living cells.
- Author
-
Percherancier Y, Germain-Desprez D, Galisson F, Mascle XH, Dianoux L, Estephan P, Chelbi-Alix MK, and Aubry M
- Subjects
- Antineoplastic Agents pharmacology, Arsenic Trioxide, Arsenicals pharmacology, Bacterial Proteins genetics, Cell Line, Fluorescence Resonance Energy Transfer methods, Humans, In Vitro Techniques, Kidney cytology, Leukemia, Promyelocytic, Acute pathology, Luciferases, Renilla genetics, Luminescent Measurements, Luminescent Proteins genetics, Mutagenesis, Site-Directed, Nuclear Proteins chemistry, Nuclear Proteins genetics, Oxides pharmacology, Promyelocytic Leukemia Protein, Protein Binding drug effects, Protein Binding physiology, Protein Structure, Tertiary, SUMO-1 Protein genetics, Transcription Factors chemistry, Transcription Factors genetics, Transfection, Tumor Suppressor Proteins chemistry, Tumor Suppressor Proteins genetics, Leukemia, Promyelocytic, Acute metabolism, Nuclear Proteins metabolism, SUMO-1 Protein metabolism, Transcription Factors metabolism, Tumor Suppressor Proteins metabolism
- Abstract
Promyelocytic leukemia protein (PML) is a tumor suppressor acting as the organizer of subnuclear structures called PML nuclear bodies (NBs). Both covalent modification of PML by the small ubiquitin-like modifier (SUMO) and non-covalent binding of SUMO to the PML SUMO binding domain (SBD) are necessary for PML NB formation and maturation. PML sumoylation and proteasome-dependent degradation induced by the E3 ubiquitin ligase, RNF4, are enhanced by the acute promyelocytic leukemia therapeutic agent, arsenic trioxide (As2O3). Here, we established a novel bioluminescence resonance energy transfer (BRET) assay to dissect and monitor PML/SUMO interactions dynamically in living cells upon addition of therapeutic agents. Using this sensitive and quantitative SUMO BRET assay that distinguishes PML sumoylation from SBD-mediated PML/SUMO non-covalent interactions, we probed the respective roles of covalent and non-covalent PML/SUMO interactions in PML degradation and interaction with RNF4. We found that, although dispensable for As2O3-enhanced PML sumoylation and RNF4 interaction, PML SBD core sequence was required for As2O3- and RNF4-induced PML degradation. As confirmed with a phosphomimetic mutant, phosphorylation of a stretch of serine residues, contained within PML SBD was needed for PML interaction with SUMO-modified protein partners and thus for NB maturation. However, mutation of these serine residues did not impair As2O3- and RNF4-induced PML degradation, contrasting with the known role of these phosphoserine residues for casein kinase 2-promoted PML degradation. Altogether, these data suggest a model whereby sumoylation- and SBD-dependent PML oligomerization within NBs is sufficient for RNF4-mediated PML degradation and does not require the phosphorylation-dependent association of PML with other sumoylated partners.
- Published
- 2009
- Full Text
- View/download PDF
29. Cholesterol-dependent separation of the beta2-adrenergic receptor from its partners determines signaling efficacy: insight into nanoscale organization of signal transduction.
- Author
-
Pontier SM, Percherancier Y, Galandrin S, Breit A, Galés C, and Bouvier M
- Subjects
- Adenylyl Cyclases metabolism, Adrenergic Agonists pharmacology, Caveolins biosynthesis, Cell Line, Cyclic AMP-Dependent Protein Kinases metabolism, GTP-Binding Protein alpha Subunits metabolism, Humans, Phosphorylation drug effects, Signal Transduction drug effects, Cholesterol metabolism, Membrane Microdomains metabolism, Models, Biological, Receptors, Adrenergic, beta-2 metabolism, Signal Transduction physiology
- Abstract
Determining the role of lipid raft nanodomains in G protein-coupled receptor signaling remains fraught by the lack of assays directly monitoring rafts in native membranes. We thus combined extensive biochemical and pharmacological approaches to a nanoscale strategy based on bioluminescence resonance energy transfer (BRET) to assess the spatial and functional influence of cholesterol-rich liquid-ordered lipid nanodomains on beta2 adrenergic receptor (beta2AR) signaling. The data revealed that whereas beta2AR did not partition within liquid-ordered lipid phase, a pool of G protein and adenylyl cyclase (AC) were sequestered in these domains. Destabilization of the liquid-ordered phase by cholesterol depletion led to a lateral redistribution of Galphas and AC that favored interactions between the receptor and its signaling partners as assessed by BRET. This resulted in an increased basal and agonist-promoted beta2AR-stimulated cAMP production that was partially dampened as a result of constitutive protein kinase A-dependent phosphorylation and desensitization of the receptor. This restraining influence of nanodomains on beta2AR signaling was further substantiated by showing that liquid-ordered lipid phase stabilization using caveolin overexpression or increasing membrane cholesterol amount led to an inhibition of beta2AR-associated signaling. Given the emerging concept that clustering of receptors and effectors into signaling platforms contributes to the efficacy and selectivity of signal transduction, our results support a model whereby cholesterol-promoted liquid-ordered lipid phase-embedding Gs and AC allows their lateral separation from the receptor, thus restraining the basal activity and controlling responsiveness of beta2AR signaling machinery within larger signaling platforms.
- Published
- 2008
- Full Text
- View/download PDF
30. [Antiviral activities of interferon and PML pathway].
- Author
-
Dianoux L, Bougrini JE, Galisson F, Percherancier Y, and Chelbi-Alix MK
- Abstract
Discovered in 1957 for their antiviral properties, interferons (IFNs) are a growing cytokine family with diverse biological activities including antitumor and immunoregulatory activities. IFN are classified in three types I, II and III. They bind to different specific cell receptors and induce via the Jak/Stat pathway the expression of more than 300 genes, the products of which are believed to mediate their biological effects. Several proteins have been implicated in resistance to viral infection in IFN-treated cells, i.e. the dsRNAdependent protein kinase PKR, the 2'5' oligoadenylate synthetase/RNaseL and Mx proteins. However, it was demonstrated that cells from triple knockout mice lacking PKR, RNase L and Mx are still sensitive to the IFN-induced antiviral state, indicating that other pathways exist. One of these pathways implicates promyelocytic leukemia (PML) protein. This article reviews the potential antiviral activities of the different IFN-induced mediators focusing onPMLpathway and how viruses from different families overcome this defence.
- Published
- 2008
- Full Text
- View/download PDF
31. Direct assessment of CXCR4 mutant conformations reveals complex link between receptor structure and G(alpha)(i) activation.
- Author
-
Berchiche YA, Chow KY, Lagane B, Leduc M, Percherancier Y, Fujii N, Tamamura H, Bachelerie F, and Heveker N
- Subjects
- Animals, Cell Line, Humans, Protein Conformation, Receptors, CXCR4 genetics, Signal Transduction genetics, Amino Acid Substitution, GTP-Binding Protein alpha Subunits metabolism, Models, Biological, Receptors, CXCR4 metabolism
- Abstract
Ligand binding to G protein-coupled receptors (GPCRs) is thought to induce changes in receptor conformation that translate into activation of downstream effectors. The link between receptor conformation and activity is still insufficiently understood, as current models of GPCR activation fail to take an increasing amount of experimental data into account. To elucidate structure-function relationships in GPCR activation, we used bioluminescence resonance energy transfer to directly assess the conformation of mutants of the chemokine receptor CXCR4. We analyzed substitutions in the arginine cage DRY motif and in the conserved asparagine N(3.35)119, which are pivotal molecular switches for receptor conformation and activation. G(alpha)(i) activation of the mutants was either similar to wild-type CXCR4 (D133N, Y135A, and N119D) or resulted in loss of activity (R134A and N119K). Mutant N119S was constitutively active but further activated by agonist. Bioluminescence resonance energy transfer analysis suggested no simple correlation between conformational changes in response to ligand binding and activation of G(alpha)(i) by the mutants. Different conformations of active receptors were detected (for wild-type CXCR4, D133N, and N119S), suggesting that different receptor conformations are able to trigger G(alpha)(i) activity. Several conformations were also found for inactive mutants. These data provide biophysical evidence for different receptor conformations being active with respect to a single readout. They support models of GPCR structure-activity relationships that take this conformational flexibility of active receptors into account.
- Published
- 2007
- Full Text
- View/download PDF
32. Probing the activation-promoted structural rearrangements in preassembled receptor-G protein complexes.
- Author
-
Galés C, Van Durm JJ, Schaak S, Pontier S, Percherancier Y, Audet M, Paris H, and Bouvier M
- Subjects
- Cell Survival, Cells, Cultured, Energy Transfer, Humans, Ligands, Models, Molecular, Protein Binding, Protein Structure, Secondary, Protein Subunits chemistry, Protein Subunits metabolism, Spectrometry, Fluorescence, Heterotrimeric GTP-Binding Proteins chemistry, Heterotrimeric GTP-Binding Proteins metabolism, Receptors, G-Protein-Coupled chemistry, Receptors, G-Protein-Coupled metabolism
- Abstract
Activation of heterotrimeric G proteins by their cognate seven transmembrane domain receptors is believed to involve conformational changes propagated from the receptor to the G proteins. However, the nature of these changes remains unknown. We monitored the conformational rearrangements at the interfaces between receptors and G proteins and between G protein subunits by measuring bioluminescence resonance energy transfer between probes inserted at multiple sites in receptor-G protein complexes. Using the data obtained for the alpha(2A)AR-G alpha(i1) beta1gamma2 complex and the available crystal structures of G alpha(i1) beta1gamma2, we propose a model wherein agonist binding induces conformational reorganization of a preexisting receptor-G protein complex, leading the G alpha-G betagamma interface to open but not dissociate. This conformational change may represent the movement required to allow nucleotide exit from the G alpha subunit, thus reflecting the initial activation event.
- Published
- 2006
- Full Text
- View/download PDF
33. Cross talk between PML and p53 during poliovirus infection: implications for antiviral defense.
- Author
-
Pampin M, Simonin Y, Blondel B, Percherancier Y, and Chelbi-Alix MK
- Subjects
- Apoptosis, Cell Line, Tumor, Cell Nucleus Structures metabolism, Humans, Organelles metabolism, Phosphorylation, Promyelocytic Leukemia Protein, RNA, Small Interfering genetics, RNA, Small Interfering metabolism, Small Ubiquitin-Related Modifier Proteins metabolism, Tumor Suppressor Protein p53 genetics, Virus Replication, Neoplasm Proteins metabolism, Nuclear Proteins metabolism, Poliovirus pathogenicity, Transcription Factors metabolism, Tumor Suppressor Protein p53 metabolism, Tumor Suppressor Proteins metabolism
- Abstract
PML nuclear bodies (NBs) are dynamic intranuclear structures harboring numerous transiently or permanently localized proteins. PML, the NBs' organizer, is directly induced by interferon, and its expression is critical for antiviral host defense. We describe herein the molecular events following poliovirus infection that lead to PML-dependent p53 activation and protection against virus infection. Poliovirus infection induces PML phosphorylation through the extracellular signal-regulated kinase pathway, increases PML SUMOylation, and induces its transfer from the nucleoplasm to the nuclear matrix. These events result in the recruitment of p53 to PML NBs, p53 phosphorylation on Ser15, and activation of p53 target genes leading to the induction of apoptosis. Moreover, the knock-down of p53 by small interfering RNA results in higher poliovirus replication, suggesting that p53 participates in antiviral defense. This effect, which requires the presence of PML, is transient since poliovirus targets p53 by inducing its degradation in a proteasome- and MDM2-dependent manner. Our results provide evidence of how poliovirus counteracts p53 antiviral activity by regulating PML and NBs, thus leading to p53 degradation.
- Published
- 2006
- Full Text
- View/download PDF
34. Monitoring protein-protein interactions in living cells by bioluminescence resonance energy transfer (BRET).
- Author
-
Hamdan FF, Percherancier Y, Breton B, and Bouvier M
- Subjects
- Animals, Cell Line, Transformed, Gene Expression physiology, Humans, Luminescent Proteins genetics, Microscopy, Fluorescence instrumentation, Proteins chemistry, Proteins genetics, Transfection methods, Fluorescence Resonance Energy Transfer methods, Luminescent Proteins metabolism, Microscopy, Fluorescence methods, Proteins metabolism
- Abstract
Bioluminescence resonance energy transfer (BRET) allows monitoring of protein-protein interactions in real time in living cells. One candidate interacting protein is fused to a luminescent energy donor, such as Renilla luciferase, and the other to a fluorescent energy acceptor, such the green fluorescent protein (GFP), and the two are then coexpressed in the same cells. If the two proteins interact, their close proximity allows nonradiative energy transfer (BRET) between the luciferase and the GFP. BRET does not occur if the two proteins are separated by more than 100 A, making the technique ideal for monitoring protein-protein interactions in biological systems. This unit describes the use of BRET to study constitutive and agonist-promoted interactions among signaling molecules, as illustrated by the homodimerization of the CXCR4 receptor and the recruitment of beta-arrestin2 to agonist-activated G-protein-coupled receptors. This noninvasive and homogeneous assay provides a robust and sensitive proteomic platform with applications for basic science research and drug discovery.
- Published
- 2006
- Full Text
- View/download PDF
35. Mutation of the DRY motif reveals different structural requirements for the CC chemokine receptor 5-mediated signaling and receptor endocytosis.
- Author
-
Lagane B, Ballet S, Planchenault T, Balabanian K, Le Poul E, Blanpain C, Percherancier Y, Staropoli I, Vassart G, Oppermann M, Parmentier M, and Bachelerie F
- Subjects
- Amino Acid Motifs, Animals, Arrestins metabolism, Arrestins pharmacology, Cell Line, Cricetinae, Dose-Response Relationship, Drug, Endocytosis drug effects, Humans, Molecular Structure, Protein Binding drug effects, Protein Binding physiology, Receptors, CCR5 chemistry, Signal Transduction drug effects, beta-Arrestins, Endocytosis physiology, Mutation, Receptors, CCR5 genetics, Receptors, CCR5 physiology, Signal Transduction physiology
- Abstract
CC chemokine receptor 5 (CCR5) is a G protein-coupled receptor that governs migration of leukocytes and serves as a coreceptor for the R5 tropic strains of human immunodeficiency virus (HIV). CCR5-mediated signaling in response to CC chemokines relies on G protein activation. Desensitization, which rapidly turns off G protein-dependent signaling, involves phosphorylation of CCR5 that promotes interaction of the receptor with beta-arrestins for endocytosis. Whether coupling to G proteins, desensitization, and endocytosis of CCR5 require the same structural determinants remains a matter of investigation. Here, we show that CCR5 displayed agonist-independent coupling to G proteins. This constitutive activity of the receptor was abrogated by TAK779 (N,N-dimethyl-N-[4-[[[2-(4-methylphenyl)-6,7-dihydro-5H-benzocyclohepten-8-yl]carbonyl]amino]benzyl]tetrahydro-2H-pyran-4-aminium chloride), a nonpeptidic CCR5 ligand that inhibits HIV infection and was found to depend on the integrity of the Asp-Arg-Tyr (DRY) motif. Changing Arg-126 by the neutral residue Asn (R126N-CCR5 mutant) abolished CCR5-mediated activation of G proteins, either constitutively or in response to agonists. In contrast, R126N-CCR5 not only retained agonist-promoted phosphorylation and beta-arrestin-dependent endocytosis but also displayed a higher basal phosphorylation than wild-type CCR5. Expression of beta-arrestin in R126N-CCR5-expressing cells resulted in receptor down-regulation, thereby suggesting that R126N-CCR5 spontaneously interacts with beta-arrestins. However, although expression of beta-arrestin favored wild-type CCR5-mediated chemotaxis, it failed to promote migration of cells expressing R126N-CCR5. Overall, these data indicate that structural requirements for CCR5-mediated activation of G proteins, albeit not involved in receptor desensitization and internalization, are needed for beta-arrestin-mediated chemotaxis. These results have implications for how distinct biological responses of CCR5 might rely on a different set of receptor conformations.
- Published
- 2005
- Full Text
- View/download PDF
36. Bioluminescence resonance energy transfer reveals ligand-induced conformational changes in CXCR4 homo- and heterodimers.
- Author
-
Percherancier Y, Berchiche YA, Slight I, Volkmer-Engert R, Tamamura H, Fujii N, Bouvier M, and Heveker N
- Subjects
- Bacterial Proteins metabolism, Binding Sites, Cell Line, Chemokine CCL2 metabolism, Cyclic AMP metabolism, Dimerization, Dose-Response Relationship, Drug, Flow Cytometry, Humans, Kinetics, Ligands, Luminescent Proteins metabolism, Peptides chemistry, Plasmids metabolism, Protein Binding, Protein Conformation, Protein Structure, Tertiary, Temperature, Time Factors, Transfection, Fluorescence Resonance Energy Transfer methods, Receptors, CXCR4 chemistry
- Abstract
Homo- and heterodimerization have emerged as prominent features of G-protein-coupled receptors with possible impact on the regulation of their activity. Using a sensitive bioluminescence resonance energy transfer system, we investigated the formation of CXCR4 and CCR2 chemokine receptor dimers. We found that both receptors exist as constitutive homo- and heterodimers and that ligands induce conformational changes within the pre-formed dimers without promoting receptor dimer formation or disassembly. Ligands with different intrinsic efficacies yielded distinct bioluminescence resonance energy transfer modulations, indicating the stabilization of distinct receptor conformations. We also found that peptides derived from the transmembrane domains of CXCR4 inhibited activation of this receptor by blocking the ligand-induced conformational transitions of the dimer. Taken together, our data support a model in which chemokine receptor homo- and heterodimers form spontaneously and respond to ligand binding as units that undergo conformational changes involving both protomers even when only one of the two ligand binding sites is occupied.
- Published
- 2005
- Full Text
- View/download PDF
37. The effects of HIV-1 Nef on CD4 surface expression and viral infectivity in lymphoid cells are independent of rafts.
- Author
-
Sol-Foulon N, Esnault C, Percherancier Y, Porrot F, Metais-Cunha P, Bachelerie F, and Schwartz O
- Subjects
- CD4-Positive T-Lymphocytes immunology, CD4-Positive T-Lymphocytes virology, Cell Line, Cells, Cultured, Gene Deletion, Genes, nef, HIV-1 genetics, HIV-1 physiology, Histocompatibility Antigens Class I metabolism, Humans, Jurkat Cells, Membrane Microdomains immunology, Membrane Microdomains virology, Mutation, Palmitic Acid metabolism, Virulence, nef Gene Products, Human Immunodeficiency Virus, CD4 Antigens metabolism, Gene Products, nef physiology, HIV-1 pathogenicity
- Abstract
The HIV-1 Nef protein is a critical virulence factor that exerts multiple effects during viral replication. Nef modulates surface expression of various cellular proteins including CD4 and MHC-I, enhances viral infectivity, and affects signal transduction pathways. Nef has been shown to partially associate with rafts, where it can prime T cells for activation. The contribution of rafts during Nef-induced CD4 down-regulation and enhancement of viral replication remains poorly understood. We show here that Nef does not modify the palmitoylation state of CD4 or its partition within rafts. Moreover, CD4 mutants lacking palmitoylation or unable to associate with rafts are efficiently down-regulated by Nef. In HIV-infected cells, viral assembly and budding occurs from rafts, and Nef has been suggested to increase this process. However, using T cells acutely infected with wild-type or nef-deleted HIV, we did not observe any impact of Nef on raft segregation of viral structural proteins. We have also designed a palmitoylated mutant of Nef (NefG3C), which significantly accumulates in rafts. Interestingly, the efficiency of NefG3C to down-regulate CD4 and MHC-I, and to promote viral replication was not increased when compared with the wild-type protein. Altogether, these results strongly suggest that rafts are not a key element involved in the effects of Nef on trafficking of cellular proteins and on viral replication.
- Published
- 2004
- Full Text
- View/download PDF
38. HIV-1 entry into T-cells is not dependent on CD4 and CCR5 localization to sphingolipid-enriched, detergent-resistant, raft membrane domains.
- Author
-
Percherancier Y, Lagane B, Planchenault T, Staropoli I, Altmeyer R, Virelizier JL, Arenzana-Seisdedos F, Hoessli DC, and Bachelerie F
- Subjects
- Base Sequence, Cell Line, Cell Membrane drug effects, Cell Membrane virology, Cholera Toxin pharmacology, Cholesterol pharmacology, DNA Primers, HIV-1 drug effects, Humans, Membrane Microdomains drug effects, Receptors, HIV immunology, CD4 Antigens immunology, HIV-1 physiology, Membrane Microdomains virology, Receptors, CCR5 immunology, Receptors, HIV physiology, Sphingolipids pharmacology, T-Lymphocytes immunology
- Abstract
The contribution of raft domains to human immunodeficiency virus (HIV) 1 entry was assessed. In particular, we asked whether the CD4 and CCR5 HIV-1 receptors need to associate with sphingolipid-enriched, detergent-resistant membrane domains (rafts) to allow viral entry into primary and T-cell lines. Based on Triton X-100 solubilization and confocal microscopy, CD4 was shown to distribute partially to rafts. In contrast, CCR5 did not associate with rafts and localized in nonraft plasma membrane domains. HIV-1-receptor partitioning remained unchanged upon viral adsorption, suggesting that viral entry probably takes place outside rafts. To directly investigate this possibility, we targeted CD4 to nonraft domains of the membrane by preventing CD4 palmitoylation and interaction with p56(lck). Directed mutagenesis of both targeting signals significantly prevented association of CD4 with rafts, but did not suppress the HIV-1 receptor function of CD4. Collectively, these results strongly suggest that the presence of HIV-1 receptors in rafts is not required for viral infection. We show, however, that depleting plasma membrane cholesterol inhibits HIV-1 entry. We therefore propose that cholesterol modulates the HIV-1 entry process independently of its ability to promote raft formation.
- Published
- 2003
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.