Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 23
Filtrer
2.
ACS Pharmacol Transl Sci ; 4(5): 1483-1498, 2021 Oct 08.
Article de Anglais | MEDLINE | ID: mdl-34661070

RÉSUMÉ

Prolonged exposure to opioid receptor agonists triggers adaptations in the adenylyl cyclase (AC) pathway that lead to enhanced production of cyclic adenosine monophosphate (cAMP) upon withdrawal. This cellular phenomenon contributes to withdrawal symptoms, hyperalgesia and analgesic tolerance that interfere with clinical management of chronic pain syndromes. Since δ-opioid receptors (DOPrs) are a promising target for chronic pain management, we were interested in finding out if cell-based signaling profiles as generated for drug discovery purposes could inform us of the ligand potential to induce sensitization of the cyclase path. For this purpose, signaling of DOPr agonists was monitored at multiple effectors. The resulting signaling profiles revealed marked functional selectivity, particularly for Met-enkephalin (Met-ENK) whose signaling bias profile differed from those of synthetic ligands like SNC-80 and ARM390. Signaling diversity among ligands was systematized by clustering agonists according to similarities in E max and Log(τ) values for the different responses. The classification process revealed that the similarity in Gα/Gßγ, but not in ß-arrestin (ßarr), responses was correlated with the potential of Met-ENK, deltorphin II, (d-penicillamine2,5)-enkephalin (DPDPE), ARM390, and SNC-80 to enhance cAMP production, all of which required Ca2+ mobilization to produce this response. Moreover, superactivation by Met-ENK, which was the most-effective Ca2+ mobilizing agonist, required Gαi/o activation, availability of Gßγ subunits at the membrane, and activation of Ca2+ effectors such as calmodulin and protein kinase C (PKC). In contrast, superactivation by (N-(l-tyrosyl)-(3S)-1,2,3,4-tetrahydroisoquinoline-3-carbonyl)-l-phenylalanyl-l-phenylalanine (TIPP), which was set in a distinct category through clustering, required activation of Gαi/o subunits but was independent of the Gßγ dimer and Ca2+ mobilization, relying instead on Src and Raf-1 to induce this cellular adaptation.

3.
Int J Mol Sci ; 22(15)2021 Jul 29.
Article de Anglais | MEDLINE | ID: mdl-34360884

RÉSUMÉ

Colorectal cancer (CRC) is the third most common cause of cancer-related deaths worldwide. Human papillomaviruses (HPVs) and Epstein-Barr virus (EBV) have been reported to be present in different types of human cancers, including CRCs, where they can play a key role in the onset and/or progression of these cancers. Thus, we herein explored the prevalence of high-risk HPVs and EBV in a cohort of 94 CRC tissue samples and 13 colorectal normal tissues from the Lebanese population using polymerase chain reaction, immunohistochemistry, and tissue microarray methodologies. We found that high-risk HPVs are present in 64%, while EBV is present in 29% of our CRC samples. Additionally, our data showed that high-risk HPV types (16, 18, 35, 58, 51, 45, 52, 31, and 33) are the most frequent in CRC in the Lebanese cohort, respectively. Our data point out that HPVs and EBV are copresent in 28% of the samples. Thus, this study clearly suggests that high-risk HPVs and EBV are present/copresent in CRCs, where they could play an important role in colorectal carcinogenesis. Nevertheless, further investigations using a larger cohort are needed to elucidate the possible cooperation between these oncoviruses in the development of CRC.


Sujet(s)
Alphapapillomavirus/génétique , Tumeurs colorectales/épidémiologie , Infections à virus Epstein-Barr/épidémiologie , Herpèsvirus humain de type 4/génétique , Infections à papillomavirus/épidémiologie , Adolescent , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Études cas-témoins , Enfant , Enfant d'âge préscolaire , Études de cohortes , Tumeurs colorectales/virologie , ADN viral/génétique , Infections à virus Epstein-Barr/virologie , Femelle , Humains , Immunohistochimie , Liban/épidémiologie , Mâle , Adulte d'âge moyen , Infections à papillomavirus/virologie , Réaction de polymérisation en chaîne , Prévalence , Facteurs de risque , Jeune adulte
4.
Cancer Cell Int ; 21(1): 308, 2021 Jun 10.
Article de Anglais | MEDLINE | ID: mdl-34112166

RÉSUMÉ

BACKGROUND: High-risk human papillomaviruses (HPVs) are present and can cooperate with Epstein-Barr virus (EBV) to initiate and/or enhance the progression of several types of human carcinomas including cervical as well as head and neck; in parallel, it has been recently pointed out that these oncoviruses can be detected in human breast cancers. Thus, we herein explored the presence/co-presence of high-risk HPVs and EBV in breast cancer in Lebanese women. METHODS: A cohort of 102 breast cancer samples and 14 normal breast tissues were assessed for the presence of HPVs and EBV. Polymerase chain reaction (PCR) and immunohistochemistry (IHC) analysis in addition to tissue microarray (TMA) platform were used in this study. RESULTS: We found the presence of HPV in 66/102 (65%) of our samples, while EBV is present in 41/102 (40%) of the cohort. Additionally, our data showed that high-risk HPV types (52, 35, 58, 45, 16 and 51) are the most frequent in breast cancer in Lebanese women. Meanwhile, we report that high-risk HPVs and EBV are co-present in 30/102 (29%) of the samples; more significantly, our results indicate that their co-presence is associated with tumor grade (p = 0.03). CONCLUSION: Our data revealed that HPVs and EBV are present/co-present in human breast cancer where they may play an important role in its development and/or progression; thus, we believe that further investigations are essential to confirm and elucidate the presence/co-presence of these oncoviruses and the underlying mechanisms of their interaction in breast carcinogenesis.

5.
Cell Signal ; 83: 109976, 2021 07.
Article de Anglais | MEDLINE | ID: mdl-33713808

RÉSUMÉ

Obesity is a global burden and a chronic ailment with damaging overall health effects. Ghrelin, an octanoylated 28 amino acid peptide hormone, is secreted from the oxyntic mucosa of the stomach. Ghrelin acts on regions of the hypothalamus to regulate feeding behavior and glucose homeostasis through its G protein-coupled receptor. Recently, several central pathways modulating the metabolic actions of ghrelin have been reported. While these signaling pathways can be inhibited or activated by antagonists or agonists, they can also be discriminatingly activated in a "biased" response to impart different degrees of activation in distinct pathways downstream of the receptor. Here, we review recent ghrelin biased signaling findings as well as characteristics of ghrelin hormone and its receptors pertinent for biased signaling. We then evaluate the feasibility for ghrelin receptor biased signaling as a strategy for the development of effective pharmacotherapy in obesity treatment.


Sujet(s)
Comportement alimentaire , Ghréline/métabolisme , Obésité/traitement médicamenteux , Obésité/métabolisme , Récepteurs à la ghréline/métabolisme , Transduction du signal , Animaux , Humains
6.
Cell Signal ; 83: 109981, 2021 07.
Article de Anglais | MEDLINE | ID: mdl-33744417

RÉSUMÉ

G protein-coupled receptors (GPCRs) represent the largest family of approved therapeutic targets. Ligands stimulating these receptors specifically activate multiple signalling pathways that induce not only the desired therapeutic response, but sometimes untolerated side effects that limit their clinical use. The diversity in signalling induced by each ligand could be considered a viable path for improving this situation. Biased agonism, which offers the promise of identifying pathway-selective drugs has been proposed as a means to exploit this opportunity. However, identifying biased agonists is not an easy process and quantifying ligand bias for a given signalling pathway requires careful consideration and control of several confounding factors. To date, the molecular mechanisms of biased signalling remain unclear and known theories that constitute our understanding of the mechanisms underlying therapeutic and side effects are still being challenged, making the strategy of selecting promising potential drugs more difficult. This special issue summarizes the latest advances in the discovery and optimization of biased ligands for different GPCRs. It also focuses on identifying novel insights into the field of biased agonism, while at the same time, highlighting the conceptual and experimental limitations of that concept for drug discovery. This aims to broaden our understanding of the signalling induced by the various identified biased agonists and provide perspectives that could straighten our path towards the development of more effective and tolerable therapeutics.


Sujet(s)
Découverte de médicament , Récepteurs couplés aux protéines G , Transduction du signal , Animaux , Humains , Récepteurs couplés aux protéines G/agonistes , Récepteurs couplés aux protéines G/génétique , Récepteurs couplés aux protéines G/métabolisme
7.
Cell Signal ; 80: 109906, 2021 04.
Article de Anglais | MEDLINE | ID: mdl-33383156

RÉSUMÉ

Opioid analgesics are elective for treating moderate to severe pain but their use is restricted by severe side effects. Signaling bias has been proposed as a viable means for improving this situation. To exploit this opportunity, continuous efforts are devoted to understand how ligand-specific modulations of receptor functions could mediate the different in vivo effects of opioids. Advances in the field have led to the development of biased agonists based on hypotheses that allocated desired and undesired effects to specific signaling pathways. However, the prevalent hypothesis associating ß-arrestin to opioid side effects was recently challenged and multiple of the newly developed biased drugs may not display the superior side effects profile that was sought. Moreover, biased agonism at opioid receptors is now known to be time- and cell-dependent, which adds a new layer of complexity for bias estimation. Here, we first review the signaling mechanisms underlying desired and undesired effects of opioids. We then describe biased agonism at opioid receptors and discuss the different perspectives that support the desired and undesired effects of opioids in view of exploiting biased signaling for therapeutic purposes. Finally, we explore how signaling kinetics and cellular background can influence the magnitude and directionality of bias at those receptors.


Sujet(s)
Protéines G/métabolisme , Récepteur delta/métabolisme , Récepteur mu/métabolisme , Transduction du signal , bêta-Arrestines/métabolisme , Analgésiques morphiniques/pharmacologie , Protéines G/agonistes , Humains , Cinétique , Ligands , Récepteur delta/agonistes , Récepteur mu/agonistes , Transduction du signal/effets des médicaments et des substances chimiques , bêta-Arrestines/agonistes
8.
Cell Signal ; 75: 109759, 2020 11.
Article de Anglais | MEDLINE | ID: mdl-32860951

RÉSUMÉ

Small ubiquitin like modifier (SUMO) conjugation or SUMOylation of ßarrestin2 promotes its association with the clathrin adaptor protein AP2 and facilitates rapid ß2 adrenergic receptor (ß2AR) internalization. However, disruption of the consensus SUMOylation site in ßarrestin2, did not prevent ßarrestin2's association with activated ß2ARs, dopamine D2 receptors (D2Rs), angiotensin type 1a receptors (AT1aRs) and V2 vasopressin receptors (V2Rs). To address the role of SUMOylation in the trafficking of ßarrestin and GPCR complexes, we generated and characterized a yellow fluorescent protein (YFP) tagged ßarrestin2-SUMO1 chimeric protein, which is resistant to de-SUMOylation. In HEK-293 cells, YFP-SUMO1 predominantly localized in the nucleus, whereas YFP-ßarrestin2 is cytoplasmic. YFP-ßarrestin2-SUMO1 in addition to being cytoplasmic, is localized at the nuclear membrane. Nonetheless, ßarrestin2-SUMO1 associated robustly with agonist-activated ß2ARs as evaluated by co-immunoprecipitation, confocal microscopy and bioluminescence resonance energy transfer (BRET). ßarrestin2-SUMO1 associated strongly with the D2R, which forms transient complexes with ßarrestin2. But, ßarrestin2-SUMO1 and ßarrestin2 showed equivalent binding with the V2R, which forms stable complexes with ßarrestin2. ßarrestin2 expression level directly correlated with the steady state levels of the unmodified form of RanGAP1, which upon SUMOylation associates with nuclear membrane. On the other hand, ßarrestin2-SUMO1 not only localized at the nuclear membrane, but also formed a macromolecular complex with RanGAP1. Taken together, our data suggest that SUMOylation of ßarrestin2 promotes its protein interactions at both cell and nuclear membranes. Furthermore, ßarrestin2-SUMO1 presents as a useful tool to characterize ßarrestin2 recruitment to GPCRs, which form transient and unstable complex with ßarrestin2.


Sujet(s)
Protéines d'activation de la GTPase/métabolisme , Pore nucléaire/métabolisme , Protéine SUMO-1/métabolisme , bêta-Arrestine 2/métabolisme , Cellules HEK293 , Humains , Liaison aux protéines , Transport des protéines , Sumoylation
9.
Front Immunol ; 11: 820, 2020.
Article de Anglais | MEDLINE | ID: mdl-32547536

RÉSUMÉ

About 50 million of the U.S. adult population suffer from chronic pain. It is a complex disease in its own right for which currently available analgesics have been deemed woefully inadequate since ~20% of the sufferers derive no benefit. Vitamin D, known for its role in calcium homeostasis and bone metabolism, is thought to be of clinical benefit in treating chronic pain without the side-effects of currently available analgesics. A strong correlation between hypovitaminosis D and incidence of bone pain is known. However, the potential underlying mechanisms by which vitamin D might exert its analgesic effects are poorly understood. In this review, we discuss pathways involved in pain sensing and processing primarily at the level of dorsal root ganglion (DRG) neurons and the potential interplay between vitamin D, its receptor (VDR) and known specific pain signaling pathways including nerve growth factor (NGF), glial-derived neurotrophic factor (GDNF), epidermal growth factor receptor (EGFR), and opioid receptors. We also discuss how vitamin D/VDR might influence immune cells and pain sensitization as well as review the increasingly important topic of vitamin D toxicity. Further in vitro and in vivo experimental studies will be required to study these potential interactions specifically in pain models. Such studies could highlight the potential usefulness of vitamin D either alone or in combination with existing analgesics to better treat chronic pain.


Sujet(s)
Analgésiques/métabolisme , Analgésiques/pharmacologie , Douleur/métabolisme , Vitamine D/métabolisme , Vitamine D/pharmacologie , Analgésiques/effets indésirables , Animaux , Récepteurs ErbB/métabolisme , Ganglions sensitifs des nerfs spinaux/métabolisme , Facteur neurotrophique dérivé des cellules gliales/métabolisme , Humains , Facteur de croissance nerveuse/métabolisme , Neurones/métabolisme , Nociception , Récepteur calcitriol/métabolisme , Récepteurs aux opioïdes/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Vitamine D/effets indésirables
11.
Nat Commun ; 10(1): 4075, 2019 09 09.
Article de Anglais | MEDLINE | ID: mdl-31501422

RÉSUMÉ

Signaling diversity of G protein-coupled (GPCR) ligands provides novel opportunities to develop more effective, better-tolerated therapeutics. Taking advantage of these opportunities requires identifying which effectors should be specifically activated or avoided so as to promote desired clinical responses and avoid side effects. However, identifying signaling profiles that support desired clinical outcomes remains challenging. This study describes signaling diversity of mu opioid receptor (MOR) ligands in terms of logistic and operational parameters for ten different in vitro readouts. It then uses unsupervised clustering of curve parameters to: classify MOR ligands according to similarities in type and magnitude of response, associate resulting ligand categories with frequency of undesired events reported to the pharmacovigilance program of the Food and Drug Administration and associate signals to side effects. The ability of the classification method to associate specific in vitro signaling profiles to clinically relevant responses was corroborated using ß2-adrenergic receptor ligands.


Sujet(s)
Récepteurs couplés aux protéines G/métabolisme , Transduction du signal , Analgésiques morphiniques/métabolisme , Animaux , Analyse de regroupements , Protéines G/métabolisme , Cochons d'Inde , Cellules HEK293 , Humains , Ligands , Récepteurs bêta-2 adrénergiques/métabolisme , Récepteur mu/métabolisme , bêta-Arrestines/métabolisme
12.
Vitam Horm ; 111: 49-90, 2019.
Article de Anglais | MEDLINE | ID: mdl-31421707

RÉSUMÉ

The delta opioid receptor (DOP) belongs to the Class A, rhodopsin-like family of G protein-coupled receptors. Although this receptor has a high level of similarity with the other opioid receptors, it displays unique aspects and functions. Indeed, as opposed to most membrane receptors, DOP is poorly addressed to the plasma membrane. In this chapter, we first review the molecular and cellular mechanisms regulating the expression and the cellular trafficking/sorting of DOP. We then summarize the structural insights of this receptor through the analysis of the existing crystal structures, with a particular focus on the role of the sodium binding site. Finally, we review the current signaling mechanisms mediating receptor function and desensitization.


Sujet(s)
Récepteur delta , Animaux , Sites de fixation , Membrane cellulaire/métabolisme , Séquence conservée , Cristallisation , Régulation de l'expression des gènes , Humains , Modèles moléculaires , Structure moléculaire , Phosphotransferases/métabolisme , Récepteur delta/composition chimique , Récepteur delta/génétique , Récepteur delta/physiologie , Transduction du signal/physiologie , Sodium/métabolisme
13.
J Drug Target ; 27(10): 1118-1126, 2019 12.
Article de Anglais | MEDLINE | ID: mdl-30995134

RÉSUMÉ

Diabetic peripheral neuropathy (DPN) is the most incapacitating complication of diabetes mellitus. Up to 50% of patients with DPN develop peripheral neuropathic pain (PNP). The underlying ionic and molecular mechanisms of diabetic PNP (DPNP) are poorly understood. However, voltage gated potassium (Kv7) channels which have been implicated in the pathogenesis of other types of PNP are likely to be involved. Here we examined, in the streptozotocin (STZ) rat model of DPNP, whether activating the Kv7 channels with a potent activator retigabine (ezogabine) would reverse/attenuate behavioural signs of DPNP. STZ rats exhibited behavioural indices of mechanical and heat hypersensitivity, but not cold hypersensitivity or spontaneous pain, 35 days after STZ injection. Retigabine given at a dose of 15 mg/kg (but not at 7.5 mg/kg, i.p.) significantly attenuated mechanical, but not heat hypersensitivity in DPNP rats, and was as effective as the positive control gabapentin. This analgesic effect of retigabine was completely reversed by the Kv7/M channel blocker XE991 (3 mg/kg, i.p.) indicating that the anti-allodynic effects of retigabine were mediated by Kv7 channels. In conclusion, the findings suggest that Kv7 channels are involved in DPNP pathogenesis, and that strategies that target their activation may prove to be effective in treating DPNP.


Sujet(s)
Anticonvulsivants/pharmacologie , Carbamates/pharmacologie , Neuropathies diabétiques/induit chimiquement , Neuropathies diabétiques/traitement médicamenteux , Canal potassique KCNQ1/métabolisme , Névralgie/traitement médicamenteux , Phénylènediamines/pharmacologie , Streptozocine/pharmacologie , Animaux , Neuropathies diabétiques/métabolisme , Modèles animaux de maladie humaine , Mâle , Névralgie/métabolisme , Rats , Rat Sprague-Dawley
14.
Methods Mol Biol ; 1957: 93-104, 2019.
Article de Anglais | MEDLINE | ID: mdl-30919349

RÉSUMÉ

Ubiquitination of G protein-coupled receptors (GPCRs) is an important dynamic posttranslational modification that has been linked to the intracellular trafficking of internalized GPCRs to lysosomes. Ubiquitination of GPCRs is mediated by specific E3 ubiquitin ligases that are scaffolded by the adaptor proteins called ß-arrestins. Traditionally, detection of GPCR ubiquitination is achieved by using ubiquitin antibodies to Western blot immunoprecipitates of detergent-solubilized GPCRs expressed in heterologous cells. However, studies have also shown that bioluminescence resonance energy transfer (BRET)-based techniques can reveal ubiquitination of GPCRs in intact cells and in real time. This chapter describes a step-by-step protocol to evaluate ubiquitination of GPCRs using the BRET methodology.


Sujet(s)
Techniques de transfert d'énergie par résonance de bioluminescence/méthodes , Récepteurs couplés aux protéines G/métabolisme , Ubiquitination , bêta-Arrestines/métabolisme , Analyse de données , Cellules HEK293 , Humains , Liaison aux protéines , Ubiquitine/métabolisme
15.
ACS Pharmacol Transl Sci ; 2(4): 230-246, 2019 Aug 09.
Article de Anglais | MEDLINE | ID: mdl-32259059

RÉSUMÉ

G-protein-coupled receptors (GPCRs) can bias signaling through distinct biochemical pathways that originate from G-protein/receptor and ß-arrestin/receptor complexes. Receptor conformations supporting ß-arrestin engagement depend on multiple receptor determinants. Using ghrelin receptor GHR1a, we demonstrate by bioluminescence resonance energy transfer and fluorescence microscopy a critical role for its second intracellular loop 2 (ICL2) domain in stabilizing ß-arrestin/GHSR1a core interactions and determining receptor trafficking fate. We validate our findings in ICL2 gain- and loss-of-function experiments assessing ß-arrestin and ubiquitin-dependent internalization of the CC chemokine receptor, CCR1. Like all CC and CXC subfamily chemokine receptors, CCR1 lacks a critical proline residue found in the ICL2 consensus domain of rhodopsin-family GPCRs. Our study indicates that ICL2, C-tail determinants, and the orthosteric binding pocket that regulates ß-arrestin/receptor complex stability are sufficient to encode a broad repertoire of the trafficking fates observed for rhodopsin-family GPCRs, suggesting they provide the essential elements for regulating a large fraction of ß-arrestin signaling bias.

16.
Cardiovasc Res ; 113(13): 1551-1559, 2017 Nov 01.
Article de Anglais | MEDLINE | ID: mdl-29048463

RÉSUMÉ

AIMS: Chronic kidney disease (CKD) is a powerful independent risk factor for cardiovascular events, including vein graft failure. Because CKD impairs the clearance of small proteins, we tested the hypothesis that CKD exacerbates vein graft disease by elevating serum levels of critical cytokines that promote vein graft neointimal hyperplasia. METHODS AND RESULTS: We modelled CKD in C57BL/6 mice with 5/6ths nephrectomy, which reduced glomerular filtration rate by 60%, and we modelled vein grafting with inferior-vena-cava-to-carotid interposition grafting. CKD increased vein graft neointimal hyperplasia four-fold, decreased vein graft re-endothelialization two-fold, and increased serum levels of interleukin-9 (IL-9) five-fold. By quantitative immunofluorescence and histochemical staining, vein grafts from CKD mice demonstrated a ∼two-fold higher prevalence of mast cells, and a six-fold higher prevalence of activated mast cells. Concordantly, vein grafts from CKD mice showed higher levels of TNF and NFκB activation, as judged by phosphorylation of NFκB p65 on Ser536 and by expression of VCAM-1. Arteriovenous fistula veins from humans with CKD also showed up-regulation of mast cells and IL-9. Treating CKD mice with IL-9-neutralizing IgG reduced vein graft neointimal area four-fold, increased vein graft re-endothelialization ∼two-fold, and reduced vein graft total and activated mast cell levels two- and four-fold, respectively. Treating CKD mice with the mast cell stabilizer cromolyn reduced neointimal hyperplasia and increased re-endothelialization in vein grafts. In vitro, IL-9 promoted endothelial cell apoptosis but had no effect on smooth muscle cell proliferation. CONCLUSION: CKD aggravates vein graft disease through mechanisms involving IL-9 and mast cell activation.


Sujet(s)
Anastomose chirurgicale artérioveineuse , Artère carotide commune/chirurgie , Interleukine-9/métabolisme , Mastocytes/métabolisme , Insuffisance rénale chronique/complications , Maladies vasculaires/complications , Veine cave inférieure/transplantation , Animaux , Apoptose , Artère carotide commune/immunologie , Artère carotide commune/métabolisme , Artère carotide commune/anatomopathologie , Cellules cultivées , Modèles animaux de maladie humaine , Cellules endothéliales/métabolisme , Cellules endothéliales/anatomopathologie , Humains , Hyperplasie , Interleukine-9/immunologie , Mastocytes/immunologie , Souris de lignée C57BL , Néointima , Phosphorylation , Insuffisance rénale chronique/immunologie , Insuffisance rénale chronique/métabolisme , Transduction du signal , Facteurs temps , Facteur de transcription RelA/métabolisme , Facteur de nécrose tumorale alpha/métabolisme , Molécule-1 d'adhérence des cellules vasculaires/métabolisme , Maladies vasculaires/immunologie , Maladies vasculaires/métabolisme , Maladies vasculaires/anatomopathologie , Veine cave inférieure/immunologie , Veine cave inférieure/métabolisme , Veine cave inférieure/anatomopathologie
17.
Methods ; 92: 78-86, 2016 Jan 01.
Article de Anglais | MEDLINE | ID: mdl-26364590

RÉSUMÉ

Signaling bias makes reference to the capacity of G-protein coupled receptor (GPCR) ligands to direct pharmacological stimuli to a subset of effectors among all of those controlled by the receptor. This new signaling modality has added texture to the classical notion of efficacy. In doing so, it has opened new avenues for the development of therapeutic GPCR ligands that specifically modulate signals underlying desired effects while sparing those that support undesired drug actions. Essential to taking advantage of this texture is the ability to identify, quantify and represent bias in a reliable and intuitive manner that ensures comparison among ligands. Here, we present a practical guide on how the operational model may be used to evaluate ligand efficiency to induce different responses, how differences in response may be used to estimate bias and how quantitative information derived from this analysis may be graphically represented to recreate a drug's unique signaling footprint. The approach used is discussed in terms of data interpretation and limitations that may influence the conclusions drawn from the analysis.


Sujet(s)
Biologie informatique/méthodes , Découverte de médicament/méthodes , Récepteurs couplés aux protéines G/métabolisme , Transduction du signal/physiologie , Relation dose-effet des médicaments , Humains , Ligands , Préparations pharmaceutiques/administration et posologie , Préparations pharmaceutiques/métabolisme , Liaison aux protéines/physiologie , Transduction du signal/effets des médicaments et des substances chimiques
18.
Methods ; 92: 19-35, 2016 Jan 01.
Article de Anglais | MEDLINE | ID: mdl-26210401

RÉSUMÉ

Ion channels play a vital role in numerous physiological functions and drugs that target them are actively pursued for development of novel therapeutic agents. Here we report a means for monitoring in real time the conformational changes undergone by channel proteins upon exposure to pharmacological stimuli. The approach relies on tracking structural rearrangements by monitoring changes in bioluminescence energy transfer (BRET). To provide proof of principle we have worked with Kir3 neuronal channels producing 10 different constructs which were combined into 17 donor-acceptor BRET pairs. Among these combinations, pairs bearing the donor Nano-Luc (NLuc) at the C-terminal end of Kir3.2 subunits and the FlAsH acceptor at the N-terminal end (NT) or the interfacial helix (N70) of Kir3.1 subunits were identified as potential tools. These pairs displayed significant changes in energy transfer upon activation with direct channel ligands or via stimulation of G protein-coupled receptors. Conformational changes associated with channel activation followed similar kinetics as channel currents. Dose response curves generated by different agonists in FlAsH-BRET assays displayed similar rank order of potency as those obtained with conventional BRET readouts of G protein activation and ion flux assays. Conformational biosensors as the ones reported herein should prove a valuable complement to other methodologies currently used in channel drug discovery.


Sujet(s)
Techniques de transfert d'énergie par résonance de bioluminescence/méthodes , Techniques de biocapteur/méthodes , Conception de médicament , Fluorescéine/synthèse chimique , Fluorescéine/métabolisme , Canaux potassiques rectifiants entrants couplés aux protéines G/métabolisme , Analgésiques morphiniques/synthèse chimique , Analgésiques morphiniques/métabolisme , Analgésiques morphiniques/pharmacologie , Animaux , Relation dose-effet des médicaments , Canaux potassiques rectifiants entrants couplés aux protéines G/composition chimique , Cellules HEK293 , Humains , Ouverture et fermeture des portes des canaux ioniques/effets des médicaments et des substances chimiques , Ouverture et fermeture des portes des canaux ioniques/physiologie , Souris , Conformation des protéines
19.
Cell Mol Life Sci ; 72(18): 3543-57, 2015 Sep.
Article de Anglais | MEDLINE | ID: mdl-25900661

RÉSUMÉ

Kir3 channels control excitability in the nervous system and the heart. Their surface expression is strictly regulated, but mechanisms responsible for channel removal from the membrane remain incompletely understood. Using transfected cells, we show that Kir3.1/3.2 channels and delta opioid receptors (DORs) associate in a complex which persists during receptor activation, behaving as a scaffold that allows beta-arrestin (ßarr) to interact with both signaling partners. This organization favored co-internalization of DORs and Kir3 channels in a ßarr-dependent manner via a clathrin/dynamin-mediated endocytic path. Taken together, these findings identify a new way of modulating Kir3 channel availability at the membrane and assign a putatively novel role for ßarrs in regulating canonical effectors for G protein-coupled receptors.


Sujet(s)
Arrestines/métabolisme , Canaux potassiques rectifiants entrants couplés aux protéines G/métabolisme , Récepteur delta/métabolisme , Animaux , Membrane cellulaire/métabolisme , Cellules cultivées , Clathrine/métabolisme , Dynamines/métabolisme , Endocytose/physiologie , Cellules HEK293 , Humains , Rats , Transduction du signal/physiologie , bêta-Arrestines
20.
Front Cell Neurosci ; 8: 186, 2014.
Article de Anglais | MEDLINE | ID: mdl-25071446

RÉSUMÉ

Opioids are among the most effective drugs to treat severe pain. They produce their analgesic actions by specifically activating opioid receptors located along the pain perception pathway where they inhibit the flow of nociceptive information. This inhibition is partly accomplished by activation of hyperpolarizing G protein-coupled inwardly-rectifying potassium (GIRK or Kir3) channels. Kir3 channels control cellular excitability in the central nervous system and in the heart and, because of their ubiquitous distribution, they mediate the effects of a large range of hormones and neurotransmitters which, upon activation of corresponding G protein-coupled receptors (GPCRs) lead to channel opening. Here we analyze GPCR signaling via these effectors in reference to precoupling and collision models. Existing knowledge on signaling bias is discussed in relation to these models as a means of developing strategies to produce novel opioid analgesics with an improved side effects profile.

SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...