Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 135
Filtrer
1.
Biophys J ; 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38970252

RÉSUMÉ

G-protein-coupled receptors (GPCRs) are integral membrane proteins that transduce chemical signals from the extracellular matrix into the cell. Traditional drug design has considered ligand-receptor interactions only under normal conditions. However, studies on opioids indicate that such interactions are very different in diseased tissues. In such microenvironments, protons play an important role in structural and functional alterations of both ligands and receptors. The pertinent literature strongly suggests that future drug design should take these aspects into account in order to reduce adverse side effects while preserving desired effects of novel compounds.

2.
Pain ; 164(11): 2501-2515, 2023 Nov 01.
Article de Anglais | MEDLINE | ID: mdl-37326658

RÉSUMÉ

ABSTRACT: Targeting the acidified inflammatory microenvironment with pH-sensitive opioids is a novel approach for managing visceral pain while mitigating side effects. The analgesic efficacy of pH-dependent opioids has not been studied during the evolution of inflammation, where fluctuating tissue pH and repeated therapeutic dosing could influence analgesia and side effects. Whether pH-dependent opioids can inhibit human nociceptors during extracellular acidification is unexplored. We studied the analgesic efficacy and side-effect profile of a pH-sensitive fentanyl analog, (±)- N -(3-fluoro-1-phenethylpiperidine-4-yl)- N -phenyl propionamide (NFEPP), during the evolution of colitis induced in mice with dextran sulphate sodium. Colitis was characterized by granulocyte infiltration, histological damage, and acidification of the mucosa and submucosa at sites of immune cell infiltration. Changes in nociception were determined by measuring visceromotor responses to noxious colorectal distension in conscious mice. Repeated doses of NFEPP inhibited nociception throughout the course of disease, with maximal efficacy at the peak of inflammation. Fentanyl was antinociceptive regardless of the stage of inflammation. Fentanyl inhibited gastrointestinal transit, blocked defaecation, and induced hypoxemia, whereas NFEPP had no such side effects. In proof-of-principle experiments, NFEPP inhibited mechanically provoked activation of human colonic nociceptors under acidic conditions mimicking the inflamed state. Thus, NFEPP provides analgesia throughout the evolution of colitis with maximal activity at peak inflammation. The actions of NFEPP are restricted to acidified layers of the colon, without common side effects in normal tissues. N -(3-fluoro-1-phenethylpiperidine-4-yl)- N -phenyl propionamide could provide safe and effective analgesia during acute colitis, such as flares of ulcerative colitis.


Sujet(s)
Colite , Douleur viscérale , Souris , Humains , Animaux , Analgésiques morphiniques/pharmacologie , Analgésiques morphiniques/usage thérapeutique , Colite/induit chimiquement , Colite/traitement médicamenteux , Côlon , Analgésiques/pharmacologie , Inflammation/anatomopathologie , Douleur viscérale/anatomopathologie , Fentanyl/pharmacologie , Fentanyl/usage thérapeutique , Concentration en ions d'hydrogène
3.
Front Mol Neurosci ; 16: 1171855, 2023.
Article de Anglais | MEDLINE | ID: mdl-37251645

RÉSUMÉ

N-(3-fluoro-1-phenethylpiperidine-4-yl)-N-phenyl propionamide is a newly-designed pain killer selectively activating G-protein-coupled mu-opioid receptors (MOR) in acidic injured tissues, and therefore devoid of central side effects which are typically elicited at normal pH values in healthy tissues. However, the neuronal mechanisms underlying NFEPP's antinociceptive effects were not examined in detail so far. Voltage-dependent Ca2+ channels (VDCCs) in nociceptive neurons play a major role in the generation and inhibition of pain. In this study, we focused on the effects of NFEPP on calcium currents in rat dorsal root ganglion (DRG) neurons. The inhibitory role of the G-protein subunits Gi/o and Gßγ on VDCCs was investigated using the blockers pertussis toxin and gallein, respectively. GTPγS binding, calcium signals and MOR phosphorylation were also investigated. All experiments were performed at acidic and normal pH values using NFEPP in comparison to the conventional opioid agonist fentanyl. At low pH, NFEPP produced more efficient G-protein activation in transfected HEK293 cells and significantly reduced VDCCs in depolarized DRG neurons. The latter effect was mediated by Gßγ subunits, and NFEPP-mediated MOR phosphorylation was pH-dependent. Fentanyl's responses were not affected by pH changes. Our data indicate that NFEPP-induced MOR signaling is more effective at low pH and that the inhibition of calcium channels in DRG neurons underlies NFEPP's antinociceptive actions.

4.
Sci Rep ; 13(1): 607, 2023 01 12.
Article de Anglais | MEDLINE | ID: mdl-36635362

RÉSUMÉ

We previously reported the successful design, synthesis and testing of the prototype opioid painkiller NFEPP that does not elicit adverse side effects. The design process of NFEPP was based on mathematical modelling of extracellular interactions between G-protein coupled receptors (GPCRs) and ligands, recognizing that GPCRs function differently under pathological versus healthy conditions. We now present an additional and novel stochastic model of GPCR function that includes intracellular dissociation of G-protein subunits and modulation of plasma membrane calcium channels and their dependence on parameters of inflamed and healthy tissue (pH, radicals). The model is validated against in vitro experimental data for the ligands NFEPP and fentanyl at different pH values and radical concentrations. We observe markedly reduced binding affinity and calcium channel inhibition for NFEPP at normal pH compared to lower pH, in contrast to the effect of fentanyl. For increasing radical concentrations, we find enhanced constitutive G-protein activation but reduced ligand binding affinity. Assessing the different effects, the results suggest that, compared to radicals, low pH is a more important determinant of overall GPCR function in an inflamed environment. Future drug design efforts should take this into account.


Sujet(s)
Récepteurs couplés aux protéines G , Transduction du signal , Récepteurs couplés aux protéines G/métabolisme , Protéines G/métabolisme , Fentanyl/pharmacologie , Conception de médicament , Ligands
5.
Med Sci (Basel) ; 10(4)2022 11 17.
Article de Anglais | MEDLINE | ID: mdl-36412904

RÉSUMÉ

The sensory ion channel transient receptor potential vanilloid 1 (TRPV1) is mainly expressed in small to medium sized dorsal root ganglion neurons, which are involved in the transfer of acute noxious thermal and chemical stimuli. The Ankyrin-rich membrane spanning protein (ARMS) interaction with TRPV1 is modulated by protein kinase A (PKA) mediating sensitization. Here, we hypothesize that PKA phosphorylation sites of ARMS are crucial for the modulation of TRPV1 function, and that the phosphorylation of ARMS is facilitated by the A-kinase anchoring protein 79 (AKAP79). We used transfected HEK293 cells, immunoprecipitation, calcium flux, and patch clamp experiments to investigate potential PKA phosphorylation sites in ARMS and in ARMS-related peptides. Additionally, experiments were done to discriminate between PKA and protein kinase D (PKD) phosphorylation. We found different interaction ratios for TRPV1 and ARMS mutants lacking PKA phosphorylation sites. The degree of TRPV1 sensitization by ARMS mutants is independent on PKA phosphorylation. AKAP79 was also involved in the TRPV1/ARMS/PKA signaling complex. These data show that ARMS is a PKA substrate via AKAP79 in the TRPV1 signaling complex and that all four proteins interact physically, regulating TRPV1 sensitization in transfected HEK293 cells. To assess the physiological and/or therapeutic significance of these findings, similar investigations need to be performed in native neurons and/or in vivo.


Sujet(s)
Ankyrines , Protéines membranaires , Humains , Ankyrines/métabolisme , Cyclic AMP-Dependent Protein Kinases/métabolisme , Cellules HEK293 , Protéines membranaires/métabolisme , Phosphorylation , Canaux cationiques TRPV/génétique , Canaux cationiques TRPV/métabolisme
6.
Sci Rep ; 12(1): 15172, 2022 09 07.
Article de Anglais | MEDLINE | ID: mdl-36071140

RÉSUMÉ

Opioid agonists are powerful drugs for managing pain. However, their central side effects are limiting their use and drugs with similar potency, but a lower risk profile are needed. (±)-N-(3-fluoro-1-phenethylpiperidine-4-yl)-N-phenylpropionamide (NFEPP) is a novel opioid agonist that preferentially activates opioid receptors at acidic extracellular pH. NFEPP was designed to activate peripheral opioid receptors in injured tissue, therefore precluding side effects elicited at normal pH in brain or intestinal wall. Considering the common combination of opioids and nonsteroidal anti-inflammatory drugs (NSAIDs) in multimodal analgesia, we investigated the interaction between NFEPP and a widely prescribed prototypical NSAID, diclofenac (DCF), in a rat model of unilateral hindpaw inflammation induced by complete Freund's adjuvant. We evaluated the effects of systemically applied DCF on the paw tissue pH, on the expression of inflammatory mediators in immune cells from inflamed paws and on the expression of opioid receptors in dorsal root ganglia. Additionally, we investigated the antinociceptive efficacy of NFEPP injected into the inflamed paws after DCF treatment. We found that DCF reduced inflammation-induced nociceptive responses and tissue acidosis, but did not change the mRNA expression of IL-1ß, TNF-α, IL-6, IL-4, NGF, or of mu-, delta-, or kappa-opioid receptors. The treatment with DCF moderately reduced the antinociceptive efficacy of NFEPP, suggesting a correlation between an increase in local tissue pH and the decreased antinociceptive effect of this pH-sensitive opioid agonist.


Sujet(s)
Acidose , Analgésiques morphiniques , Pipéridines , Acidose/induit chimiquement , Acidose/traitement médicamenteux , Analgésiques/pharmacologie , Analgésiques morphiniques/effets indésirables , Animaux , Concentration en ions d'hydrogène , Inflammation/induit chimiquement , Inflammation/traitement médicamenteux , Pipéridines/pharmacologie , Rats , Récepteurs aux opioïdes/métabolisme
7.
Gut ; 71(4): 695-704, 2022 04.
Article de Anglais | MEDLINE | ID: mdl-33785555

RÉSUMÉ

OBJECTIVE: The effectiveness of µ-opioid receptor (MOPr) agonists for treatment of visceral pain is compromised by constipation, respiratory depression, sedation and addiction. We investigated whether a fentanyl analogue, (±)-N-(3-fluoro-1-phenethylpiperidine-4-yl)-N-phenyl propionamide (NFEPP), which preferentially activates MOPr in acidified diseased tissues, would inhibit pain in a preclinical model of inflammatory bowel disease (IBD) without side effects in healthy tissues. DESIGN: Antinociceptive actions of NFEPP and fentanyl were compared in control mice and mice with dextran sodium sulfate colitis by measuring visceromotor responses to colorectal distension. Patch clamp and extracellular recordings were used to assess nociceptor activation. Defecation, respiration and locomotion were assessed. Colonic migrating motor complexes were assessed by spatiotemporal mapping of isolated tissue. NFEPP-induced MOPr signalling and trafficking were studied in human embryonic kidney 293 cells. RESULTS: NFEPP inhibited visceromotor responses to colorectal distension in mice with colitis but not in control mice, consistent with acidification of the inflamed colon. Fentanyl inhibited responses in both groups. NFEPP inhibited the excitability of dorsal root ganglion neurons and suppressed mechanical sensitivity of colonic afferent fibres in acidified but not physiological conditions. Whereas fentanyl decreased defecation and caused respiratory depression and hyperactivity in mice with colitis, NFEPP was devoid of these effects. NFEPP did not affect colonic migrating motor complexes at physiological pH. NFEPP preferentially activated MOPr in acidified extracellular conditions to inhibit cAMP formation, recruit ß-arrestins and evoke MOPr endocytosis. CONCLUSION: In a preclinical IBD model, NFEPP preferentially activates MOPr in acidified microenvironments of inflamed tissues to induce antinociception without causing respiratory depression, constipation and hyperactivity.


Sujet(s)
Colite , Tumeurs colorectales , Maladies inflammatoires intestinales , Insuffisance respiratoire , Douleur viscérale , Animaux , Colite/induit chimiquement , Côlon , Constipation , Fentanyl/effets indésirables , Humains , Maladies inflammatoires intestinales/complications , Souris , Récepteurs aux opioïdes , Microenvironnement tumoral
8.
Clin Pharmacol Ther ; 109(4): 1055-1062, 2021 04.
Article de Anglais | MEDLINE | ID: mdl-32770831

RÉSUMÉ

HIV remains a major burden to the health care system and neuropathic pain is the most common neurological complication of HIV infection. Because current treatment strategies often lack satisfying pain relief, cannabinoids (CBs) are discussed as a new option. We investigated cannabidivarin (CBDV) as treatment for HIV-associated neuropathic pain. We conducted a randomized, double-blind, placebo-controlled crossover study. Patients underwent two successive treatment phases (4 weeks each) and were treated with CBDV (400 mg/day) or placebo in a randomized order. A 3-week washout phase was designed to eliminate potential carry-over effects. Patients were followed up for 3 weeks after the end of the second treatment phase. The primary end point was pain intensity on an 11-point numeric rating scale, recorded in a diary. Secondary end points were additional pain medication, pain characteristics, and quality of life. We included 32 patients. The mean pain intensity under CBDV was 0.62 points higher compared with placebo (P = 0.16, 95% confidence interval -0.27 to 1.51). CBDV did not influence the amount of additional pain medication, pain characteristics, or quality of life. The incidence of adverse events was similar during both treatments. No suspected unexpected adverse reactions occurred during either treatment. CBDV was safe but failed to reduce neuropathic pain in patients with HIV. This may be explained by a lack of CB receptor activation, as indicated by preclinical experiments. Although a larger patient number might be desirable, we would not expect a change in the conclusions because the present differences are far from statistical significance. Therefore, we would currently not consider CBDV as a clinically meaningful treatment option for neuropathic pain.


Sujet(s)
Cannabinoïdes/usage thérapeutique , Infections à VIH/complications , Névralgie/traitement médicamenteux , Névralgie/étiologie , Adulte , Sujet âgé , Analgésiques , Cannabinoïdes/administration et posologie , Cannabinoïdes/effets indésirables , Études croisées , Méthode en double aveugle , Femelle , Humains , Mâle , Adulte d'âge moyen , Mesure de la douleur , Qualité de vie
9.
PLoS One ; 15(12): e0243286, 2020.
Article de Anglais | MEDLINE | ID: mdl-33270761

RÉSUMÉ

In vitro and ex vivo development of novel therapeutic agents requires reliable and accurate analyses of the cell conditions they were preclinical tested for, such as apoptosis. The detection of apoptotic cells by annexin V (AV) coupled to fluorophores has often shown limitations in the choice of the dye due to interference with other fluorescent-labeled cell markers. The SNAP-tag technology is an easy, rapid and versatile method for functionalization of proteins and was therefore used for labeling AV with various fluorophores. We generated the fusion protein AV-SNAP and analyzed its capacity for the specific display of apoptotic cells in various assays with therapeutic agents. AV-SNAP showed an efficient coupling reaction with five different fluorescent dyes. Two selected fluorophores were tested with suspension, adherent and peripheral blood cells, treated by heat-shock or apoptosis-inducing therapeutic agents. Flow cytometry analysis of apoptotic cells revealed a strong visualization using AV-SNAP coupled to these two fluorophores exemplary, which was comparable to a commercial AV-Assay-kit. The combination of the apoptosis-specific binding protein AV with the SNAP-tag provides a novel solid method to facilitate protein labeling using several, easy to change, fluorescent dyes at once. It avoids high costs and allows an ordinary exchange of dyes and easier use of other fluorescent-labeled cell markers, which is of high interest for the preclinical testing of therapeutic agents in e.g. cancer research.


Sujet(s)
Apoptose/physiologie , Colorants fluorescents/composition chimique , Coloration et marquage/méthodes , Marqueurs d'affinité/composition chimique , Annexine A5/composition chimique , Annexine A5/métabolisme , Cellules sanguines/métabolisme , Lignée cellulaire tumorale , Cytométrie en flux/méthodes , Humains , Tumeurs/métabolisme , Protéines/composition chimique , Protéines/métabolisme , Technologie
10.
Sci Rep ; 10(1): 18599, 2020 10 29.
Article de Anglais | MEDLINE | ID: mdl-33122720

RÉSUMÉ

The newly designed fentanyl derivative [( ±)-N-(3-fluoro-1-phenethylpiperidine-4-yl)-N-phenyl propionamide] (NFEPP) was recently shown to produce analgesia selectively via peripheral mu-opioid receptors (MOR) at acidic pH in rat inflamed tissues. Here, we examined the pH-dependency of NFEPP binding to brain MOR and its effects on bone cancer-induced pain in mice. The IC50 of NFEPP to displace bound [3H]-DAMGO was significantly higher compared to fentanyl at pH 7.4, but no differences were observed at pH 5.5 or 6.5. Intravenous NFEPP (30-100 nmol/kg) or fentanyl (17-30 nmol/kg) inhibited heat hyperalgesia in mice inoculated with B16-F10 melanoma cells. The peripherally-restricted opioid receptor antagonist naloxone-methiodide reversed the effect of NFEPP (100 nmol/kg), but not of fentanyl (30 nmol/kg). The antihyperalgesic effect of NFEPP was abolished by a selective MOR- (cyprodime), but not delta- (naltrindole) or kappa- (nor-binaltorphimine) receptor antagonists. Ten-fold higher doses of NFEPP than fentanyl induced maximal antinociception in mice without tumors, which was reversed by the non-restricted antagonist naloxone, but not by naloxone-methiodide. NFEPP also reduced heat hyperalgesia produced by fibrosarcoma- (NCTC 2472) or prostate cancer-derived (RM1) cells. These data demonstrate the increased affinity of NFEPP for murine MOR at low pH, and its ability to inhibit bone cancer-induced hyperalgesia through peripheral MOR. In mice, central opioid receptors may be activated by ten-fold higher doses of NFEPP.


Sujet(s)
Analgésiques morphiniques/pharmacologie , Douleur cancéreuse/traitement médicamenteux , Pipéridines/pharmacologie , Récepteur mu/métabolisme , Animaux , Tumeurs osseuses/complications , Tumeurs osseuses/métabolisme , Douleur cancéreuse/métabolisme , Lignée cellulaire tumorale , Fentanyl/pharmacologie , Concentration en ions d'hydrogène , Hyperalgésie/traitement médicamenteux , Hyperalgésie/métabolisme , Ligands , Mâle , Mélanome expérimental/complications , Mélanome expérimental/métabolisme , Souris , Souris de lignée C3H , Souris de lignée C57BL , Morphinanes/pharmacologie , Naloxone/pharmacologie , Naltrexone/analogues et dérivés , Naltrexone/pharmacologie , Antagonistes narcotiques/pharmacologie
11.
Pain ; 161(12): 2798-2804, 2020 12.
Article de Anglais | MEDLINE | ID: mdl-32639370

RÉSUMÉ

Currently, opioids targeting mu-opioid receptors are the most potent drugs for acute and cancer pain. However, opioids produce adverse side effects such as constipation, respiratory depression, or addiction potential. We recently developed (±)-N-(3-fluoro-1-phenethylpiperidine-4-yl)-N-phenyl propionamide (NFEPP), a compound that does not evoke central or intestinal side effects due to its selective activation of mu-opioid receptors at low pH in peripheral injured tissues. Although we demonstrated that NFEPP effectively abolishes injury-induced pain, hyperalgesia, and allodynia in rodents, the efficacy of NFEPP in nonevoked ongoing pain remains to be established. Here, we examined reward, locomotor activity, and defecation in rats with complete Freund's adjuvant-induced paw inflammation to compare fentanyl's and NFEPP's potentials to induce side effects and to inhibit spontaneous pain. We demonstrate that low, but not higher, doses of NFEPP produce conditioned place preference but not constipation or motor disturbance, in contrast to fentanyl. Using a peripherally restricted antagonist, we provide evidence that NFEPP-induced place preference is mediated by peripheral opioid receptors. Our results indicate that a low dose of NFEPP produces reward by abolishing spontaneous inflammatory pain.


Sujet(s)
Analgésiques morphiniques , Douleur , Analgésiques morphiniques/usage thérapeutique , Animaux , Concentration en ions d'hydrogène , Hyperalgésie/traitement médicamenteux , Douleur/traitement médicamenteux , Rats , Récepteurs aux opioïdes , Récepteur mu/génétique
12.
J Chem Inf Model ; 60(7): 3566-3576, 2020 07 27.
Article de Anglais | MEDLINE | ID: mdl-32491854

RÉSUMÉ

Opioids are molecules whose binding to specialized G-Protein Coupled Receptors (GPCRs) triggers a signaling cascade that leads to the downregulation of pain pathways. Binding of an opioid to the membrane-embedded GPCR occurs when the opioid molecule is protonated, which provides a potential strategy to design nontoxic opioids that are protonated and bind to the GPCR only at the low pH of injured or inflamed tissue. Excellent model systems to study protonation-dependent binding of opioids to GPCRs are fentanyl, which is protonated and binds to the GPCR at both physiological and low pH, and the fluorinated fentanyl derivative NFEPP, which is protonated and binds to the GPCR only at low pH. The molecular mechanisms of fentanyl and NFEPP binding to the GPCR are largely unknown. To enable atomistic studies of opioid binding to GPCRs, we have carried out extensive quantum mechanical and classical mechanical computations to derive a potential energy function for fentanyl and NFEPP and present force field parameters for both opioid molecules. We find that fluorination alters the electronic ground state properties of fentanyl. As a consequence, fentanyl and NFEPP have distinct torsional and electrostatic properties likely to impact how they bind to receptors.


Sujet(s)
Analgésiques morphiniques , Fentanyl , Analgésiques , Fentanyl/usage thérapeutique , Humains , Douleur/traitement médicamenteux , Récepteur mu
13.
Curr Opin Support Palliat Care ; 14(2): 112-117, 2020 06.
Article de Anglais | MEDLINE | ID: mdl-32304400

RÉSUMÉ

PURPOSE OF REVIEW: Opioids are potent drugs for the treatment of severe pain, but they are burdened by detrimental side-effects, such as respiratory depression, addiction, sedation and constipation. Their clinical application is undisputed in acute (e.g. perioperative) and cancer pain, but their use in chronic nonmalignant pain has met increasing scrutiny and has contributed to the opioid crisis. Thus, novel analgesics with reduced side-effects are badly needed. RECENT FINDINGS: Current research topics include enkephalinase inhibitors, allosteric and multivalent ligands, biased opioid receptor signaling and selective activation of peripheral opioid receptors in injured tissues. SUMMARY: Opioids still appear to be most promising among current approaches in the development of analgesics. Basic knowledge about pathophysiology of clinical pain and novel insights in pharmacology suggest that the most interesting perspectives are augmenting endogenous opioid actions and selectively targeting peripheral opioid receptors. The latter approach is additionally supported by evidence from clinical studies. Some biased, multivalent and peripherally selective agonists have advanced to phase III trials, but novel drugs have not become available for clinical application. Future strategies in analgesic drug development might include public-private partnerships and nonprofit pharmaceutical companies, as exemplified by the AIDS crisis and proposals to combat antibiotic resistance.


Sujet(s)
Analgésiques morphiniques/usage thérapeutique , Douleur chronique/traitement médicamenteux , Douleur chronique/physiopathologie , Formulations dissuadant les abus , Analgésiques morphiniques/administration et posologie , Analgésiques morphiniques/effets indésirables , Tolérance aux médicaments/physiologie , Humains , Néprilysine/antagonistes et inhibiteurs , Troubles liés aux opiacés/physiopathologie , Troubles liés aux opiacés/prévention et contrôle , Récepteurs aux opioïdes/métabolisme , Transduction du signal/physiologie
14.
Sci Rep ; 10(1): 4366, 2020 Mar 04.
Article de Anglais | MEDLINE | ID: mdl-32127599

RÉSUMÉ

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

15.
Sci Rep ; 9(1): 19344, 2019 12 18.
Article de Anglais | MEDLINE | ID: mdl-31852967

RÉSUMÉ

The non-selective activation of central and peripheral opioid receptors is a major shortcoming of currently available opioids. Targeting peripheral opioid receptors is a promising strategy to preclude side effects. Recently, we showed that fentanyl-derived µ-opioid receptor (MOR) agonists with reduced acid dissociation constants (pKa) due to introducing single fluorine atoms produced injury-restricted antinociception in rat models of inflammatory, postoperative and neuropathic pain. Here, we report that a new double-fluorinated compound (FF6) and fentanyl show similar pKa, MOR affinity and [35S]-GTPγS binding at low and physiological pH values. In vivo, FF6 produced antinociception in injured and non-injured tissue, and induced sedation and constipation. The comparison of several fentanyl derivatives revealed a correlation between pKa values and pH-dependent MOR activation, antinociception and side effects. An opioid ligand's pKa value may be used as discriminating factor to design safer analgesics.


Sujet(s)
Analgésiques morphiniques/effets indésirables , Analgésiques morphiniques/synthèse chimique , Analgésiques morphiniques/composition chimique , Animaux , Conception de médicament , Cellules HEK293 , Humains , Concentration en ions d'hydrogène , Ligands , Mâle , Nociception/effets des médicaments et des substances chimiques , Pipéridines/effets indésirables , Pipéridines/synthèse chimique , Pipéridines/composition chimique , Rat Wistar , Récepteur mu/métabolisme
16.
Best Pract Res Clin Rheumatol ; 33(3): 101420, 2019 06.
Article de Anglais | MEDLINE | ID: mdl-31703793

RÉSUMÉ

This article reviews the role of analgesic drugs with a particular emphasis on opioids. Opioids are the oldest and most potent drugs for the treatment of severe pain, but they are burdened by detrimental side effects such as respiratory depression, addiction, sedation, nausea, and constipation. Their clinical application is undisputed in acute (e.g., perioperative) and cancer pain, but their long-term use in chronic pain has met increasing scrutiny and has contributed to the current opioid crisis. We discuss epidemiological data, pharmacological principles, clinical applications, and research strategies aiming at novel opioids with reduced side effects.


Sujet(s)
Analgésiques morphiniques , Douleur chronique/traitement médicamenteux , Gestion de la douleur/méthodes , Gestion de la douleur/tendances , Analgésiques morphiniques/effets indésirables , Humains
17.
Brain Res Bull ; 152: 134-142, 2019 10.
Article de Anglais | MEDLINE | ID: mdl-31323279

RÉSUMÉ

We evaluated acetylcholine release by microdialysis in 10 month old control and JNPL3 mice which carry a mutant tau gene (P301 L). Three brain regions were compared: hippocampus and thalamus which receive cholinergic input from the basal forebrain, and the red nucleus which receives cholinergic projections from brain stem nuclei. Cognitive and motor functions of the mice were largely normal. In microdialysis experiments, we found significant reductions in basal ACh levels in hippocampus and thalamus, but not in the red nucleus. ACh release was impaired most strongly (by 50%) when a physiological stimulus was applied, i.e. exploration of a novel environment, whereas most mice responded adequately with an increase of ACh release upon infusion of scopolamine. A strong reduction of scopolamine-mediated ACh release was seen after amyloid Aß42 peptide was administered into the hippocampus of tau-transgenic mice. Choline acetyltransferase activities were unchanged in tau-transgenic mice but acetylcholinesterase activities were increased in thalamus. Lactate and choline levels were increased in tau-transgenic mice but high-affinity choline uptake was slightly reduced. Our data suggest that even mild to moderate tau pathology in JNPL3 mice is able to depress cholinergic transmission in brain regions that receive input from the basal forebrain via long projection neurons. This impairment may be reinforced by amyloid peptide formation.


Sujet(s)
Acétylcholine/métabolisme , Tauopathies/métabolisme , Protéines tau/génétique , Acétylcholine/physiologie , Acetylcholinesterase/métabolisme , Peptides bêta-amyloïdes/métabolisme , Animaux , Encéphale/métabolisme , Choline , Choline O-acetyltransferase/métabolisme , Femelle , Hippocampe/métabolisme , Hippocampe/physiologie , Interneurones/métabolisme , Mâle , Souris , Lignées consanguines de souris , Souris transgéniques , Microdialyse , Noyau rouge/métabolisme , Noyau rouge/physiologie , Scopolamine/pharmacologie , Lobe temporal/métabolisme , Thalamus/métabolisme , Thalamus/physiologie , Protéines tau/métabolisme
18.
Br J Pharmacol ; 176(23): 4510-4520, 2019 12.
Article de Anglais | MEDLINE | ID: mdl-31355457

RÉSUMÉ

BACKGROUND AND PURPOSE: Adverse side effects of conventional opioids can be avoided if ligands selectively activate peripheral opioid receptors in injured tissue. Injury and inflammation are typically accompanied by acidification. In this study, we examined influences of low pH and mutation of the ionizable amino acid residue H2976.52 on µ-opioid receptor binding and signalling induced by the µ-opioid receptor ligands fentanyl, DAMGO, and naloxone. EXPERIMENTAL APPROACH: HEK 293 cells stably transfected with µ-opioid receptors were used to study opioid ligand binding, [35 S]-GTPγS binding, and cAMP reduction at physiological and acidic pH. We used µ-opioid receptors mutated at H2976.52 to A (MOR-H2976.52 A) to delineate ligand-specific interactions with H2976.52 . KEY RESULTS: Low pH and the mutant receptor MOR-H2976.52 A impaired naloxone binding and antagonism of cAMP reduction. In addition, DAMGO binding and G-protein activation were decreased under these conditions. Fentanyl-induced signalling was not influenced by pH and largely independent of H2976.52 . CONCLUSIONS AND IMPLICATIONS: Our investigations indicate that low pH selectively impairs µ-opioid receptor signalling modulated by ligands capable of forming hydrogen bonds with H2976.52 . We propose that protonation of H2976.52 at acidic pH reduces binding and subsequent signalling of such ligands. Novel agonists targeting opioid receptors in injured tissue might benefit from lack of hydrogen bond formation with H2976.52 .


Sujet(s)
2-Alanine-5-glycine-4-méthylphénylalanine-enképhaline/pharmacologie , Fentanyl/pharmacologie , Histamine/métabolisme , Naloxone/pharmacologie , Récepteur mu/antagonistes et inhibiteurs , Cellules cultivées , Relation dose-effet des médicaments , 2-Alanine-5-glycine-4-méthylphénylalanine-enképhaline/composition chimique , Fentanyl/composition chimique , Cellules HEK293 , Histamine/génétique , Humains , Concentration en ions d'hydrogène , Ligands , Structure moléculaire , Mutation , Naloxone/composition chimique , Récepteur mu/génétique , Récepteur mu/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Relation structure-activité
19.
Expert Opin Investig Drugs ; 27(10): 765-775, 2018 10.
Article de Anglais | MEDLINE | ID: mdl-30148648

RÉSUMÉ

INTRODUCTION: Opioids are the oldest and most potent drugs for the treatment of severe pain, but they are burdened by detrimental side effects such as respiratory depression, addiction, sedation, nausea, and constipation. Their clinical application is undisputed in acute (e.g. perioperative) and cancer pain, but their long-term use in chronic pain has met increasing scrutiny and has contributed to the current 'opioid crisis.' AREAS COVERED: This article reviews pharmacological principles and research strategies aiming at novel opioids with reduced side effects. Basic mechanisms underlying pain, opioid analgesia, and other opioid actions are outlined. To illustrate the clinical situation and medical needs, plasticity of opioid receptors, intracellular signaling pathways, endogenous and exogenous opioid receptor ligands, central and peripheral sites of analgesic, and side effects are discussed. EXPERT OPINION: The epidemic of opioid misuse has taught us that there is a lack of fundamental knowledge about the characteristics and management of chronic pain, that conflicts of interest and validity of models must be considered in the context of drug development, and that novel analgesics with less abuse liability are badly needed. Currently, the most promising perspectives appear to be augmenting endogenous opioid actions and selectively targeting pathological conformations of peripheral opioid receptors.


Sujet(s)
Analgésiques morphiniques/usage thérapeutique , Douleur chronique/traitement médicamenteux , Récepteurs aux opioïdes/effets des médicaments et des substances chimiques , Analgésiques morphiniques/effets indésirables , Analgésiques morphiniques/pharmacologie , Animaux , Douleur chronique/physiopathologie , Conception de médicament , Humains , Troubles liés aux opiacés/épidémiologie , Troubles liés aux opiacés/prévention et contrôle , Transduction du signal/effets des médicaments et des substances chimiques
20.
Pain ; 159(11): 2277-2284, 2018 Nov.
Article de Anglais | MEDLINE | ID: mdl-29994988

RÉSUMÉ

Recently, (±)-N-(3-fluoro-1-phenethylpiperidine-4-yl)-N-phenyl propionamide (NFEPP), a newly designed µ-opioid receptor (MOR) agonist with a low pKa, has been shown to produce injury-restricted analgesia in models of inflammatory and postoperative pain, without exhibiting typical opioid side effects. Here, we investigated MOR binding of NFEPP in brain and dorsal root ganglia, pH in injured tissues, and the analgesic efficacy of NFEPP compared with fentanyl in a chronic constriction injury model of neuropathic pain, and in the acetic acid-induced abdominal writhing assay in rats. Binding experiments revealed significantly lower affinity of NFEPP compared with fentanyl at pH 7.4. In vivo, pH significantly dropped both at injured nerves after chronic constriction injury and in the abdominal cavity after acetic acid administration. Intravenous NFEPP as well as fentanyl dose-dependently diminished neuropathy-induced mechanical and heat hypersensitivity, and acetic acid-induced abdominal constrictions. In both models, NFEPP-induced analgesia was fully reversed by naloxone methiodide, a peripherally restricted opioid receptor antagonist, injected at the nerve injury site or into the abdominal cavity. Our results indicate that NFEPP exerts peripheral opioid receptor-mediated analgesia exclusively in damaged tissue in models of neuropathic and abdominal pain.


Sujet(s)
Douleur abdominale/traitement médicamenteux , Névralgie/traitement médicamenteux , Pipéridines/usage thérapeutique , Récepteur mu/agonistes , Animaux , Encéphale/cytologie , Membrane cellulaire/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Ganglions sensitifs des nerfs spinaux/cytologie , Concentration en ions d'hydrogène , Hyperalgésie/traitement médicamenteux , Mâle , Mesure de la douleur/effets des médicaments et des substances chimiques , Seuil nociceptif/effets des médicaments et des substances chimiques , Liaison aux protéines/effets des médicaments et des substances chimiques , Rats , Rat Wistar , Récepteur mu/métabolisme , Statistique non paramétrique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...