46 results on '"Gavériaux-Ruff, C"'
Search Results
2. Mu opioid receptors in the medial habenula contribute to naloxone aversion
- Author
-
Boulos, L. J., primary, Ben Hamida, S., additional, Bailly, J., additional, Maitra, M., additional, Ehrlich, A. T., additional, Gavériaux-Ruff, C., additional, Darcq, E., additional, and Kieffer, B. L., additional
- Published
- 2019
- Full Text
- View/download PDF
3. Opioid receptor genes inactivated in mice: the highlights
- Author
-
Gavériaux-Ruff, C. and Kieffer, B.L.
- Published
- 2002
- Full Text
- View/download PDF
4. S.1.01 Brain opioid receptor systems: molecular and functional aspects
- Author
-
Kieffer, B., Gavériaux-Ruff, C., Befort, K., Massotte, D., and Becker, J.
- Published
- 2009
- Full Text
- View/download PDF
5. Kappa opioid receptor antagonism and chronic antidepressant treatment have beneficial activities on social interactions and grooming deficits during heroin abstinence
- Author
-
Lalanne, L. (Laurence), Ayranci, G. (G), Filliol, D. (Dominique), Gavériaux-Ruff, C. (C), Befort, K. (Katia), Kieffer, B. (Brigitte) L. (L), and Lutz, P. (Pierre-Eric) E. (E)
- Subjects
mental disorders ,Aucun - Abstract
Addiction is a chronic brain disorder that progressively invades all aspects of personal life. Accordingly, addiction to opiates severely impairs interpersonal relationships, and the resulting social isolation strongly contributes to the severity and chronicity of the disease. Uncovering new therapeutic strategies that address this aspect of addiction is therefore of great clinical relevance. We recently established a mouse model of heroin addiction in which, following chronic heroin exposure, 'abstinent' mice progressively develop a strong and long-lasting social avoidance phenotype. Here, we explored and compared the efficacy of two pharmacological interventions in this mouse model. Because clinical studies indicate some efficacy of antidepressants on emotional dysfunction associated with addiction, we first used a chronic 4-week treatment with the serotonergic antidepressant fluoxetine, as a reference. In addition, considering prodepressant effects recently associated with kappa opioid receptor signaling, we also investigated the kappa opioid receptor antagonist norbinaltorphimine (norBNI). Finally, we assessed whether fluoxetine and norBNI could reverse abstinence-induced social avoidance after it has established. Altogether, our results show that two interspaced norBNI administrations are sufficient both to prevent and to reverse social impairment in heroin abstinent animals. Therefore, kappa opioid receptor antagonism may represent a useful approach to alleviate social dysfunction in addicted individuals.
- Published
- 2016
6. Mu and delta opioid receptor knockout mice show increased colonic sensitivity
- Author
-
Reiss, D., primary, Ceredig, R.A., additional, Secher, T., additional, Boué, J., additional, Barreau, F., additional, Dietrich, G., additional, and Gavériaux-Ruff, C., additional
- Published
- 2016
- Full Text
- View/download PDF
7. Kappa opioid receptor antagonism and chronic antidepressant treatment have beneficial activities on social interactions and grooming deficits during heroin abstinence
- Author
-
Lalanne, L., primary, Ayranci, G., additional, Filliol, D., additional, Gavériaux‐Ruff, C., additional, Befort, K., additional, Kieffer, B. L., additional, and Lutz, P‐E, additional
- Published
- 2016
- Full Text
- View/download PDF
8. Antibody response and allogeneic mixed lymphocyte reaction in mu-, delta-, and kappa-opioid receptor knockout mice
- Author
-
Gaveriaux-Ruff, C., Simonin, F., Filliol, D., and Kieffer, B.
- Published
- 2004
- Full Text
- View/download PDF
9. Kappa opioid receptor antagonism and chronic antidepressant treatment have beneficial activities on social interactions and grooming deficits during heroin abstinence.
- Author
-
Lalanne, L., Ayranci, G., Filliol, D., Gavériaux‐Ruff, C., Befort, K., Kieffer, B. L., and Lutz, P‐E
- Subjects
DRUG abstinence ,OPIOID receptors ,ANTIDEPRESSANTS ,SOCIAL isolation ,SEROTONIN ,HEROIN ,FLUOXETINE ,NARCOTIC antagonists ,SECOND-generation antidepressants ,ANIMAL behavior ,ANIMAL experimentation ,ANIMALS ,PERSONAL beauty ,BIOLOGICAL models ,CELL receptors ,MICE ,NALTREXONE ,RESEARCH funding ,SOCIAL skills ,SUBSTANCE abuse ,TIME ,TREATMENT effectiveness ,PHARMACODYNAMICS ,THERAPEUTICS - Abstract
Addiction is a chronic brain disorder that progressively invades all aspects of personal life. Accordingly, addiction to opiates severely impairs interpersonal relationships, and the resulting social isolation strongly contributes to the severity and chronicity of the disease. Uncovering new therapeutic strategies that address this aspect of addiction is therefore of great clinical relevance. We recently established a mouse model of heroin addiction in which, following chronic heroin exposure, 'abstinent' mice progressively develop a strong and long-lasting social avoidance phenotype. Here, we explored and compared the efficacy of two pharmacological interventions in this mouse model. Because clinical studies indicate some efficacy of antidepressants on emotional dysfunction associated with addiction, we first used a chronic 4-week treatment with the serotonergic antidepressant fluoxetine, as a reference. In addition, considering prodepressant effects recently associated with kappa opioid receptor signaling, we also investigated the kappa opioid receptor antagonist norbinaltorphimine (norBNI). Finally, we assessed whether fluoxetine and norBNI could reverse abstinence-induced social avoidance after it has established. Altogether, our results show that two interspaced norBNI administrations are sufficient both to prevent and to reverse social impairment in heroin abstinent animals. Therefore, kappa opioid receptor antagonism may represent a useful approach to alleviate social dysfunction in addicted individuals. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
10. Mu and delta opioid receptor knockout mice show increased colonic sensitivity.
- Author
-
Reiss, D., Ceredig, R.A., Secher, T., Boué, J., Barreau, F., Dietrich, G., and Gavériaux ‐ Ruff, C.
- Subjects
PAIN management ,ANALGESICS ,ANIMAL experimentation ,ANIMALS ,CELL receptors ,COLITIS ,DEXTRAN ,ENKEPHALINS ,INTERLEUKIN-1 ,MICE ,NARCOTICS ,OPIOID peptides ,PAIN ,PROTEIN precursors ,TUMOR necrosis factors ,PHARMACODYNAMICS - Abstract
Background: Opiates act through opioid receptors to diminish pain. Here, we investigated whether mu (MOR) and delta (DOR) receptor endogenous activity assessed in the whole mouse body or in particular at peripheral receptors on primary nociceptive neurons, control colonic pain.Methods: We compared global MOR and DOR receptor knockout (KO) mice, mice with a conditional deletion of MOR and DOR in Nav1.8-positive nociceptive primary afferent neurons (cKO), and control floxed mice of both genders for visceral sensitivity. Visceromotor responses to colorectal distension (CRD) and macroscopic colon scores were recorded on naïve mice and mice with acute colitis induced by 3% dextran sodium sulphate (DSS) for 5 days. Transcript expression for opioid genes and cytokines was measured by quantitative RT-PCR.Results: Naïve MOR and DOR global KO mice show increased visceral sensitivity that was not observed in cKO mice. MOR and preproenkephalin (Penk) were the most expressed opioid genes in colon. MOR KO mice had augmented kappa opioid receptor and Tumour-Necrosis-Factor-α and diminished Penk transcript levels while DOR, preprodynorphin and Interleukin-1β were unchanged. Global MOR KO females had a thicker colon than floxed females. No alteration was detected in DOR mutant animals. A 5-day DSS treatment led to comparable hypersensitivity in the different mouse lines.Conclusion: Our results suggest that mu and delta opioid receptor global endogenous activity but not activity at the peripheral Nav1.8 neurons contribute to visceral sensitivity in naïve mice, and that endogenous MOR and DOR tones were insufficient to elicit analgesia after 5-day DSS-induced colitis.Significance: Knockout mice for mu and delta opioid receptor have augmented colon sensitivity in the CRD assay. It shows endogenous mu and delta opioid analgesia that may be explored as potential targets for alleviating chronic intestinal pain. [ABSTRACT FROM AUTHOR]- Published
- 2017
- Full Text
- View/download PDF
11. kappa-Opioid receptor in humans: cDNA and genomic cloning, chromosomal assignment, functional expression, pharmacology, and expression pattern in the central nervous system.
- Author
-
Simonin, F, primary, Gavériaux-Ruff, C, additional, Befort, K, additional, Matthes, H, additional, Lannes, B, additional, Micheletti, G, additional, Mattéi, M G, additional, Charron, G, additional, Bloch, B, additional, and Kieffer, B, additional
- Published
- 1995
- Full Text
- View/download PDF
12. Molecular characterization of opioid receptors
- Author
-
Kieffer, B.L., primary, Befort, K., additional, Simonin, F., additional, Matthes, H., additional, Gavériaux-Ruff, C., additional, Mattéi, M.G., additional, and Hirth, C., additional
- Published
- 1994
- Full Text
- View/download PDF
13. O02 Récepteurs opioïdes mu/delta et analgésie
- Author
-
Gaveriaux-Ruff, C.
- Published
- 2007
- Full Text
- View/download PDF
14. The human delta-opioid receptor: genomic organization, cDNA cloning, functional expression, and distribution in human brain.
- Author
-
Simonin, F, Befort, K, Gavériaux-Ruff, C, Matthes, H, Nappey, V, Lannes, B, Micheletti, G, and Kieffer, B
- Abstract
We have used the mouse delta-opioid receptor (mDOR) cDNA to isolate the mDOR gene and its human homologue. In both species the coding region is interrupted by two introns with conserved exon-intron boundaries located after transmembrane domains 1 and 4. Using the polymerase chain reaction and primers based on the sequence of the cloned human delta-opioid receptor (hDOR) gene, we have obtained a full length cDNA encoding the hDOR from SH-SY5Y neuroblastoma cells. The cDNA sequence is 100% identical to the cloned human genomic sequence and 94% identical to the mouse sequence at the protein level. When expressed in COS cells, hDOR displays nanomolar affinities for delta-selective ligands, whereas the affinities for mu- and kappa-selective ligands are in the micromolar range. The delta agonists [D-Ala2, D-Leu5]enkephalin, cyclic [D-penicillamine2,D-penicillamine5]enkephalin, and BW373U86 efficiently decrease forskolin-induced cAMP levels in hDOR-expressing COS cells, indicating functional coupling of the receptor. The distribution of hDOR mRNA in human brain was investigated using delta-selective reverse transcription-polymerase chain reaction amplification, followed by Southern hybridization with a delta-specific probe. The transcript is found in cortical areas, including olfactory bulb, hippocampus, and amygdala, as well as in basal ganglia and hypothalamus. No expression is detected in internal globus pallidus, thalamus, any investigated brainstem structure, or pituitary gland. Taken together, our results indicate similar structural, pharmacological, functional, and anatomical properties for the hDOR and the mDOR and therefore support the use of rodent models for the study of these receptors in opioid function.
- Published
- 1994
15. Delta-opioid receptor gene expression in the mouse forebrain: Localization in cholinergic neurons of the striatum
- Author
-
Le Moine, C., Kieffer, B., Gaveriaux-Ruff, C., Befort, K., and Block, B.
- Published
- 1994
- Full Text
- View/download PDF
16. The delta-opioid receptor: isolation of a cDNA by expression cloning and pharmacological characterization.
- Author
-
Kieffer, B L, Befort, K, Gavériaux-Ruff, C, and Hirth, C G
- Published
- 1994
- Full Text
- View/download PDF
17. Expression of opioid receptors mRNAs in immune cells
- Author
-
Gavériaux-Ruff, C, Simonin, F, Peluso, J, Befort, K, and Kieffer, B
- Published
- 1994
- Full Text
- View/download PDF
18. Expression cloning of a murine delta opioid receptor
- Author
-
Kieffer, B.L., Befort, K., Gavériaux-Ruff, C, Mattéi, M.G., and Hirth, C
- Published
- 1994
- Full Text
- View/download PDF
19. Gdaphen, R pipeline to identify the most important qualitative and quantitative predictor variables from phenotypic data.
- Author
-
Muñiz Moreno MDM, Gavériaux-Ruff C, and Herault Y
- Subjects
- Animals, Genotype, Linear Models, Random Forest
- Abstract
Background: In individuals or animals suffering from genetic or acquired diseases, it is important to identify which clinical or phenotypic variables can be used to discriminate between disease and non-disease states, the response to treatments or sexual dimorphism. However, the data often suffers from low number of samples, high number of variables or unbalanced experimental designs. Moreover, several parameters can be recorded in the same test. Thus, correlations should be assessed, and a more complex statistical framework is necessary for the analysis. Packages already exist that provide analysis tools, but they are not found together, rendering the decision method and implementation difficult for non-statisticians., Result: We present Gdaphen, a fast joint-pipeline allowing the identification of most important qualitative and quantitative predictor variables to discriminate between genotypes, treatments, or sex. Gdaphen takes as input behavioral/clinical data and uses a Multiple Factor Analysis (MFA) to deal with groups of variables recorded from the same individuals or anonymize genotype-based recordings. Gdaphen uses as optimized input the non-correlated variables with 30% correlation or higher on the MFA-Principal Component Analysis (PCA), increasing the discriminative power and the classifier's predictive model efficiency. Gdaphen can determine the strongest variables that predict gene dosage effects thanks to the General Linear Model (GLM)-based classifiers or determine the most discriminative not linear distributed variables thanks to Random Forest (RF) implementation. Moreover, Gdaphen provides the efficacy of each classifier and several visualization options to fully understand and support the results as easily readable plots ready to be included in publications. We demonstrate Gdaphen capabilities on several datasets and provide easily followable vignettes., Conclusions: Gdaphen makes the analysis of phenotypic data much easier for medical or preclinical behavioral researchers, providing an integrated framework to perform: (1) pre-processing steps as data imputation or anonymization; (2) a full statistical assessment to identify which variables are the most important discriminators; and (3) state of the art visualizations ready for publication to support the conclusions of the analyses. Gdaphen is open-source and freely available at https://github.com/munizmom/gdaphen , together with vignettes, documentation for the functions and examples to guide you in each own implementation., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
20. μ-Opioid Receptors on Distinct Neuronal Populations Mediate Different Aspects of Opioid Reward-Related Behaviors.
- Author
-
Severino AL, Mittal N, Hakimian JK, Velarde N, Minasyan A, Albert R, Torres C, Romaneschi N, Johnston C, Tiwari S, Lee AS, Taylor AM, Gavériaux-Ruff C, Kieffer BL, Evans CJ, Cahill CM, and Walwyn WM
- Subjects
- Animals, Mice, Morphine, Neurons, Reward, Analgesics, Opioid pharmacology, Receptors, Opioid, mu genetics
- Abstract
μ-Opioid receptors (MORs) are densely expressed in different brain regions known to mediate reward. One such region is the striatum where MORs are densely expressed, yet the role of these MOR populations in modulating reward is relatively unknown. We have begun to address this question by using a series of genetically engineered mice based on the Cre recombinase/loxP system to selectively delete MORs from specific neurons enriched in the striatum: dopamine 1 (D1) receptors, D2 receptors, adenosine 2a (A2a) receptors, and choline acetyltransferase (ChAT). We first determined the effects of each deletion on opioid-induced locomotion, a striatal and dopamine-dependent behavior. We show that MOR deletion from D1 neurons reduced opioid (morphine and oxycodone)-induced hyperlocomotion, whereas deleting MORs from A2a neurons resulted in enhanced opioid-induced locomotion, and deleting MORs from D2 or ChAT neurons had no effect. We also present the effect of each deletion on opioid intravenous self-administration. We first assessed the acquisition of this behavior using remifentanil as the reinforcing opioid and found no effect of genotype. Mice were then transitioned to oxycodone as the reinforcer and maintained here for 9 d. Again, no genotype effect was found. However, when mice underwent 3 d of extinction training, during which the drug was not delivered, but all cues remained as during the maintenance phase, drug-seeking behavior was enhanced when MORs were deleted from A2a or ChAT neurons. These findings show that these selective MOR populations play specific roles in reward-associated behaviors., (Copyright © 2020 Severino et al.)
- Published
- 2020
- Full Text
- View/download PDF
21. Mu and delta opioid receptors play opposite nociceptive and behavioural roles on nerve-injured mice.
- Author
-
Martínez-Navarro M, Cabañero D, Wawrzczak-Bargiela A, Robe A, Gavériaux-Ruff C, Kieffer BL, Przewlocki R, Baños JE, and Maldonado R
- Subjects
- Animals, Hyperalgesia, Male, Mice, Mice, Neurologic Mutants, Receptors, Opioid, Receptors, Opioid, mu genetics, Nociception, Receptors, Opioid, delta genetics
- Abstract
Background and Purpose: Mu and delta opioid receptors(MOP, DOP) contribution to the manifestations of pathological pain is not understood. We used genetic approaches to investigate the opioid mechanisms modulating neuropathic pain and its comorbid manifestations., Experimental Approach: We generated conditional knockout mice with MOP or DOP deletion in sensoryNav1.8-positive neurons (Nav1.8), in GABAergic forebrain neurons (DLX5/6) orconstitutively (CMV). Mutant mice and wild-type littermates were subjected topartial sciatic nerve ligation (PSNL) or sham surgery and their nociception wascompared. Anxiety-, depressivelike behaviour and cognitive performance were also measured. Opioid receptor mRNA expression, microgliosis and astrocytosis were assessed in the dorsalroot ganglia (DRG) and/or the spinal cord (SC)., Key Results: Constitutive CMV-MOP knockouts after PSNL displayed reduced mechanical allodynia and enhanced heat hyperalgesia. This phenotype was accompanied by increased DOP expression in DRG and SC, and reduced microgliosis and astrocytosis in deep dorsal horn laminae. Conditional MOP knockouts and control mice developed similar hypersensitivity after PSNL, except for anenhanced heat hyperalgesia by DLX5/6-MOP male mice. Neuropathic pain-induced anxiety was aggravated in CMV-MOP and DLX5/6-MOP knockouts. Nerve-injured CMV-DOP mice showed increased mechanical allodynia, whereas Nav1.8-DOP and DLX5/8-DOP mice had partial nociceptive enhancement. CMV-DOP and DLX5/6-DOP mutants showed increased depressive-like behaviour after PSNL., Conclusions and Implications: MOP activity after nerve injury increased anxiety-like responses involving forebrain GABAergic neurons and enhanced mechanical pain sensitivity along with repression of DOP expression and spinal cord gliosis. In contrast, DOP shows a protective function limiting nociceptive and affective manifestations of neuropathic pain., (© 2019 The British Pharmacological Society.)
- Published
- 2020
- Full Text
- View/download PDF
22. Mu Opioid Receptors in Gamma-Aminobutyric Acidergic Forebrain Neurons Moderate Motivation for Heroin and Palatable Food.
- Author
-
Charbogne P, Gardon O, Martín-García E, Keyworth HL, Matsui A, Mechling AE, Bienert T, Nasseef MT, Robé A, Moquin L, Darcq E, Ben Hamida S, Robledo P, Matifas A, Befort K, Gavériaux-Ruff C, Harsan LA, von Elverfeldt D, Hennig J, Gratton A, Kitchen I, Bailey A, Alvarez VA, Maldonado R, and Kieffer BL
- Subjects
- Animals, Conditioning, Classical drug effects, Conditioning, Classical physiology, Corpus Striatum drug effects, Corpus Striatum metabolism, Corpus Striatum physiology, Female, GABAergic Neurons metabolism, Inhibitory Postsynaptic Potentials drug effects, Male, Mice, Mice, Knockout, Morphine administration & dosage, Motivation drug effects, Neural Pathways physiology, Prosencephalon drug effects, Prosencephalon metabolism, Receptors, Opioid, mu genetics, Ventral Tegmental Area drug effects, Ventral Tegmental Area physiology, Feeding Behavior physiology, GABAergic Neurons physiology, Heroin administration & dosage, Motivation physiology, Narcotics administration & dosage, Prosencephalon physiology, Receptors, Opioid, mu physiology
- Abstract
Background: Mu opioid receptors (MORs) are central to pain control, drug reward, and addictive behaviors, but underlying circuit mechanisms have been poorly explored by genetic approaches. Here we investigate the contribution of MORs expressed in gamma-aminobutyric acidergic forebrain neurons to major biological effects of opiates, and also challenge the canonical disinhibition model of opiate reward., Methods: We used Dlx5/6-mediated recombination to create conditional Oprm1 mice in gamma-aminobutyric acidergic forebrain neurons. We characterized the genetic deletion by histology, electrophysiology, and microdialysis; probed neuronal activation by c-Fos immunohistochemistry and resting-state functional magnetic resonance imaging; and investigated main behavioral responses to opiates, including motivation to obtain heroin and palatable food., Results: Mutant mice showed MOR transcript deletion mainly in the striatum. In the ventral tegmental area, local MOR activity was intact, and reduced activity was only observed at the level of striatonigral afferents. Heroin-induced neuronal activation was modified at both sites, and whole-brain functional networks were altered in live animals. Morphine analgesia was not altered, and neither was physical dependence to chronic morphine. In contrast, locomotor effects of heroin were abolished, and heroin-induced catalepsy was increased. Place preference to heroin was not modified, but remarkably, motivation to obtain heroin and palatable food was enhanced in operant self-administration procedures., Conclusions: Our study reveals dissociable MOR functions across mesocorticolimbic networks. Thus, beyond a well-established role in reward processing, operating at the level of local ventral tegmental area neurons, MORs also moderate motivation for appetitive stimuli within forebrain circuits that drive motivated behaviors., (Copyright © 2017 Society of Biological Psychiatry. Published by Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
23. Opioid-induced hyperalgesia: Cellular and molecular mechanisms.
- Author
-
Roeckel LA, Le Coz GM, Gavériaux-Ruff C, and Simonin F
- Subjects
- Analgesics, Opioid pharmacology, Animals, Humans, Hyperalgesia genetics, Analgesics, Opioid adverse effects, Hyperalgesia chemically induced, Hyperalgesia metabolism
- Abstract
Opioids produce strong analgesia but their use is limited by a paradoxical hypersensitivity named opioid-induced hyperalgesia (OIH) that may be associated to analgesic tolerance. In the last decades, a significant number of preclinical studies have investigated the factors that modulate OIH development as well as the cellular and molecular mechanisms underlying OIH. Several factors have been shown to influence OIH including the genetic background and sex differences of experimental animals as well as the opioid regimen. Mu opioid receptor (MOR) variants and interactions of MOR with different proteins were shown important. Furthermore, at the cellular level, both neurons and glia play a major role in OIH development. Several neuronal processes contribute to OIH, like activation of neuroexcitatory mechanisms, long-term potentiation (LTP) and descending pain facilitation. Increased nociception is also mediated by neuroinflammation induced by the activation of microglia and astrocytes. Neurons and glial cells exert synergistic effects, which contribute to OIH. The molecular actors identified include the Toll-like receptor 4 and the anti-opioid systems as well as some other excitatory molecules, receptors, channels, chemokines, pro-inflammatory cytokines or lipids. This review summarizes the intracellular and intercellular pathways involved in OIH and highlights some mechanisms that may be challenged to limit OIH in the future., (Copyright © 2016 IBRO. Published by Elsevier Ltd. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
24. μ-Opioid receptor antibody reveals tissue-dependent specific staining and increased neuronal μ-receptor immunoreactivity at the injured nerve trunk in mice.
- Author
-
Schmidt Y, Gavériaux-Ruff C, and Machelska H
- Subjects
- Animals, Cell Line, Humans, Immunohistochemistry, Mice, Mice, Knockout, Pain metabolism, Peripheral Nervous System Diseases metabolism, Receptors, Opioid, mu genetics, Sciatic Nerve metabolism, Antibodies metabolism, Ganglia, Spinal metabolism, Receptors, Opioid, mu metabolism, Skin metabolism
- Abstract
Neuropathic pain is a debilitating chronic disease often resulting from damage to peripheral nerves. Activation of opioid receptors on peripheral sensory neurons can attenuate pain without central nervous system side effects. Here we aimed to analyze the distribution of neuronal μ-opioid receptors, the most relevant opioid receptors in the control of clinical pain, along the peripheral neuronal pathways in neuropathy. Hence, following a chronic constriction injury of the sciatic nerve in mice, we used immunohistochemistry to quantify the μ-receptor protein expression in the dorsal root ganglia (DRG), directly at the injured nerve trunk, and at its peripheral endings in the hind paw skin. We also thoroughly examined the μ-receptor antibody staining specificity. We found that the antibody specifically labeled μ-receptors in human embryonic kidney 293 cells as well as in neuronal processes of the sciatic nerve and hind paw skin dermis, but surprisingly not in the DRG, as judged by the use of μ/δ/κ-opioid receptor knockout mice. Therefore, a reliable quantitative analysis of μ-receptor expression in the DRG was not possible. However, we demonstrate that the μ-receptor immunoreactivity was strongly enhanced proximally to the injury at the nerve trunk, but was unaltered in paws, on days 2 and 14 following injury. Thus, μ-opioid receptors at the site of axonal damage might be a promising target for the control of painful neuropathies. Furthermore, our findings suggest a rigorous tissue-dependent characterization of antibodies' specificity, preferably using knockout animals.
- Published
- 2013
- Full Text
- View/download PDF
25. Opiate-induced analgesia: contributions from mu, delta and kappa opioid receptors mouse mutants.
- Author
-
Gavériaux-Ruff C
- Subjects
- Animals, Mice, Mice, Mutant Strains, Polymorphism, Genetic, Protein Transport, Receptors, Opioid genetics, Receptors, Opioid metabolism, Receptors, Opioid physiology, Analgesics, Opioid pharmacology
- Abstract
The three opioid receptors, mu, delta and kappa all mediate analgesia, and knockout mice with opioid receptor deletion have proven unique tools to investigate in vivo opioid pharmacology. Since a few years, a number of new mouse lines have been engineered, with several distinct mutations of the mu receptor, to assess the role of specific amino acid residues or peptidic sequences of this receptor in analgesia and tolerance. The analysis of analgesia in mice deleted in kappa receptor and triple mu/delta/kappa receptor knockout mice have provided advances in opioid-induced analgesia. Also, the global and conditional deletion of the delta receptor in peripheral nociceptive neurons has revealed the participation of the targeted receptors in opioid-induced analgesia. Another approach for the study of opioid receptors is the visualization of these receptors in vivo, by engineering of knock-in mice with fluorescently tagged receptors. A mouse line with a fluorescent delta receptor has allowed live imaging of this receptor in behavioral paradigms and first studies on ligand-biased agonism at this receptor in vivo. The studies with mutant mouse lines for opioid receptors, combined with novel molecular and pharmacological approaches, will allow to develop novel strategies for opioid-based analgesia. This review summarizes the different genetically modified mouse lines for opioid receptors as well as the data and concepts inferred from analgesia results on these mutant mouse lines.
- Published
- 2013
- Full Text
- View/download PDF
26. δ-Opioid mechanisms for ADL5747 and ADL5859 effects in mice: analgesia, locomotion, and receptor internalization.
- Author
-
Nozaki C, Le Bourdonnec B, Reiss D, Windh RT, Little PJ, Dolle RE, Kieffer BL, and Gavériaux-Ruff C
- Subjects
- Analgesia methods, Analgesics, Opioid agonists, Animals, Disease Models, Animal, Gene Knock-In Techniques, Humans, Inflammation drug therapy, Inflammation genetics, Inflammation metabolism, Locomotion genetics, Locomotion physiology, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Mice, Transgenic, NAV1.8 Voltage-Gated Sodium Channel genetics, NAV1.8 Voltage-Gated Sodium Channel metabolism, Neuralgia genetics, Neuralgia metabolism, Neurons drug effects, Neurons metabolism, Neurons physiology, Pain Measurement methods, Piperazines pharmacology, Receptors, Opioid, delta genetics, Benzamides pharmacology, Benzopyrans pharmacology, Locomotion drug effects, Neuralgia drug therapy, Receptors, Opioid, delta metabolism, Spiro Compounds pharmacology
- Abstract
N,N-diethyl-4-(5-hydroxyspiro[chromene-2,4'-piperidine]-4-yl) benzamide (ADL5859) and N,N-diethyl-3-hydroxy-4-(spiro[chromene-2,4'-piperidine]-4-yl)benzamide (ADL5747) are novel δ-opioid agonists that show good oral bioavailability and analgesic and antidepressive effects in the rat and represent potential drugs for chronic pain treatment. Here, we used genetic approaches to investigate molecular mechanisms underlying their analgesic effects in the mouse. We tested analgesic effects of ADL5859 and ADL5747 in mice by using mechanical sensitivity measures in both complete Freund's adjuvant and sciatic nerve ligation pain models. We examined their analgesic effects in δ-opioid receptor constitutive knockout (KO) mice and mice with a conditional deletion of δ-receptor in peripheral voltage-gated sodium channel (Nav)1.8-expressing neurons (cKO mice). Both ADL5859 and ADL5747, and the prototypical δ agonist 4-[(R)-[(2S,5R)-4-allyl-2,5-dimethyl-piperazin-1-yl]-(3-methoxyphenyl)methyl]-N,N-diethyl-benzamide (SNC80) as a control, significantly reduced inflammatory and neuropathic pain. The antiallodynic effects of all three δ-opioid agonists were abolished in constitutive δ-receptor KO mice and strongly diminished in δ-receptor cKO mice. We also measured two other well described effects of δ agonists, increase in locomotor activity and agonist-induced receptor internalization by using knock-in mice expressing enhanced green fluorescence protein-tagged δ receptors. In contrast to SNC80, ADL5859 and ADL5747 did not induce either hyperlocomotion or receptor internalization in vivo. In conclusion, both ADL5859 and ADL5747 showed efficient pain-reducing properties in the two models of chronic pain. Their effects were mediated by δ-opioid receptors, with a main contribution of receptors expressed on peripheral Nav1.8-positive neurons. The lack of in vivo receptor internalization and locomotor activation, typically induced by SNC80, suggests agonist-biased activity at the receptor for the two drugs.
- Published
- 2012
- Full Text
- View/download PDF
27. Influence of endogenous opioid systems on T lymphocytes as assessed by the knockout of mu, delta and kappa opioid receptors.
- Author
-
Karaji AG, Reiss D, Matifas A, Kieffer BL, and Gavériaux-Ruff C
- Subjects
- Animals, Cytokines biosynthesis, Flow Cytometry, Mice, Mice, Inbred C57BL, Mice, Knockout, Receptors, Opioid, delta deficiency, Receptors, Opioid, delta genetics, Receptors, Opioid, kappa deficiency, Receptors, Opioid, kappa genetics, Receptors, Opioid, mu deficiency, Receptors, Opioid, mu genetics, Reverse Transcriptase Polymerase Chain Reaction, Spleen cytology, Spleen immunology, T-Lymphocyte Subsets immunology, T-Lymphocyte Subsets metabolism, Th1 Cells metabolism, Th2 Cells immunology, Th2 Cells metabolism, Thymus Gland cytology, Thymus Gland immunology, Opioid Peptides immunology, Receptors, Opioid, delta immunology, Receptors, Opioid, kappa immunology, Receptors, Opioid, mu immunology, Th1 Cells immunology
- Abstract
Here, we evaluated the influence of endogenous opioid activation on immune responses by examining consequences of all three opioid receptor gene (mu, delta and kappa) inactivation. In triple-opioid receptor knockout mice, splenocytes and thymocytes numbers, lymphocyte subsets as well as proliferation and cytokines induced by in vitro stimulation of T lymphocytes were measured. Compared with wild-type mice, similar lymphocyte distribution in thymus and spleen as well as comparable T lymphocyte proliferation were observed, while lower levels of IL-2 and IFNγ as well as higher levels of IL-4 and IL-10 were found in triple-opioid receptor knockout mice. Together, our results indicate a shift from TH1 to TH2 cytokines in triple-opioid receptor knockout animals, suggesting that global endogenous opioid tone drives T lymphocytes toward a TH1 profile under non-pathological conditions.
- Published
- 2011
- Full Text
- View/download PDF
28. The delta opioid receptor: an evolving target for the treatment of brain disorders.
- Author
-
Pradhan AA, Befort K, Nozaki C, Gavériaux-Ruff C, and Kieffer BL
- Subjects
- Animals, Brain Diseases metabolism, Clinical Trials as Topic, Drug Design, Drug Evaluation, Preclinical, Humans, Receptors, Opioid, delta metabolism, Brain Diseases drug therapy, Receptors, Opioid, delta agonists
- Abstract
Compared to the better-known mu opioid receptor, delta opioid receptors have been relatively understudied. However, the development of highly selective delta opioid agonists and the availability of genetic mouse models have extended our knowledge of delta opioid receptors in vivo. Here we review recent developments in the characterization of delta opioid receptor biology and aspects of delta opioid receptor function that have potential for therapeutic targeting. Preclinical data have confirmed that delta opioid receptor activation reduces persistent pain and improves negative emotional states; clinical trials have been initiated to assess the effectiveness of delta opioid agonists in chronic pain and depression. Furthermore, a possible role for these receptors in neuroprotection is being investigated. The usefulness of targeting delta opioid receptors in drug abuse remains open and a role for these receptors in impulse control disorders is emerging. Finally, the recent demonstration of biased agonism at the delta opioid receptor in vivo opens novel perspectives towards targeting specific therapeutic effects through drug design., (Copyright © 2011 Elsevier Ltd. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
29. Delta opioid receptor analgesia: recent contributions from pharmacology and molecular approaches.
- Author
-
Gavériaux-Ruff C and Kieffer BL
- Subjects
- Analgesics, Opioid pharmacology, Animals, Disease Models, Animal, Drug Delivery Systems, Drug Design, Humans, Mice, Mutation, Pain physiopathology, Protein Processing, Post-Translational, Receptors, Opioid, delta genetics, Receptors, Opioid, delta metabolism, Transcription, Genetic, Analgesics, Opioid therapeutic use, Pain drug therapy, Receptors, Opioid, delta agonists
- Abstract
Delta opioid receptors represent a promising target for the development of novel analgesics. A number of tools have been developed recently that have significantly improved our knowledge of δ receptor function in pain control. These include several novel δ agonists with potent analgesic properties, and genetic mouse models with targeted mutations in the δ opioid receptor gene. Also, recent findings have further documented the regulation of δ receptor function at cellular level, which impacts on the pain-reducing activity of the receptor. These regulatory mechanisms occur at transcriptional and post-translational levels, along agonist-induced receptor activation, signaling and trafficking, or in interaction with other receptors and neuromodulatory systems. All these tools for in-vivo research, and proposed mechanisms at molecular level, have tremendously increased our understanding of δ receptor physiology, and contribute to designing innovative strategies for the treatment of chronic pain and other diseases such as mood disorders.
- Published
- 2011
- Full Text
- View/download PDF
30. Mu-opioid receptors are not necessary for nortriptyline treatment of neuropathic allodynia.
- Author
-
Bohren Y, Karavelic D, Tessier LH, Yalcin I, Gavériaux-Ruff C, Kieffer BL, Freund-Mercier MJ, and Barrot M
- Subjects
- Adrenergic Uptake Inhibitors therapeutic use, Analysis of Variance, Animals, Male, Mice, Mice, Knockout, Morphine therapeutic use, Naltrexone analogs & derivatives, Naltrexone pharmacology, Narcotic Antagonists pharmacology, Neuralgia drug therapy, Neuralgia genetics, Pain Measurement drug effects, Receptors, Opioid, mu genetics, Neuralgia metabolism, Nortriptyline therapeutic use, Receptors, Opioid, mu metabolism
- Abstract
Tricyclic antidepressants (TCAs) are among the first line treatments clinically recommended against neuropathic pain. However, the mechanism by which they alleviate pain is still unclear. Pharmacological and genetic approaches evidenced a critical role of delta-opioid receptors (DORs) in the therapeutic action of chronic TCA treatment. It is however unclear whether mu-opioid receptors (MORs) are also necessary to the pain-relieving action of TCAs. The lack of highly selective MOR antagonists makes difficult to conclude based on pharmacological studies. In the present work, we thus used a genetic approach and compared mutant mice lacking MORs and their wild-type littermates. The neuropathy was induced by unilateral sciatic nerve cuffing. The threshold for mechanical response was evaluated using von Frey filaments. MOR-deficient mice displayed the same baseline for mechanical sensitivity as their wild-type littermates. After sciatic nerve cuffing, both wild-type and MOR-deficient mice displayed an ipsilateral mechanical allodynia. After about 10 days of treatment, nortriptyline suppressed this allodynia in both wild-type and MOR-deficient mice. MORs are thus not critical for nortriptyline action against neuropathic pain. An acute injection of the DOR antagonist naltrindole induced a relapse of neuropathic allodynia in both wild-type and MOR-deficient mice, thus confirming the critical role of DORs in nortriptyline action. Moreover, morphine induced an acute analgesia in control and in neuropathic wild-type mice, but was without effect in MOR-deficient mice. While MORs are crucial for morphine action, they are not critical for nortriptyline action. Our results highlight the functional difference between DORs and MORs in mechanisms of pain relief., (Copyright (c) 2009 European Federation of International Association for the Study of Pain Chapters. Published by Elsevier Ltd. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
31. In vivo delta opioid receptor internalization controls behavioral effects of agonists.
- Author
-
Pradhan AA, Becker JA, Scherrer G, Tryoen-Toth P, Filliol D, Matifas A, Massotte D, Gavériaux-Ruff C, and Kieffer BL
- Subjects
- Animals, Benzamides pharmacology, Biological Transport, Active drug effects, Cell Membrane metabolism, Cells, Cultured, Green Fluorescent Proteins chemistry, Green Fluorescent Proteins genetics, Green Fluorescent Proteins metabolism, In Vitro Techniques, Ligands, Mice, Mice, Transgenic, Neurons drug effects, Neurons physiology, Pain drug therapy, Pain physiopathology, Phosphorylation, Piperazines pharmacology, Piperidines pharmacology, Protein Conformation, Receptors, Opioid, delta chemistry, Receptors, Opioid, delta genetics, Recombinant Fusion Proteins chemistry, Recombinant Fusion Proteins genetics, Recombinant Fusion Proteins metabolism, Behavior, Animal drug effects, Behavior, Animal physiology, Receptors, Opioid, delta agonists, Receptors, Opioid, delta physiology
- Abstract
Background: GPCRs regulate a remarkable diversity of biological functions, and are thus often targeted for drug therapies. Stimulation of a GPCR by an extracellular ligand triggers receptor signaling via G proteins, and this process is highly regulated. Receptor activation is typically accompanied by desensitization of receptor signaling, a complex feedback regulatory process of which receptor internalization is postulated as a key event. The in vivo significance of GPCR internalization is poorly understood. In fact, the majority of studies have been performed in transfected cell systems, which do not adequately model physiological environments and the complexity of integrated responses observed in the whole animal., Methods and Findings: In this study, we used knock-in mice expressing functional fluorescent delta opioid receptors (DOR-eGFP) in place of the native receptor to correlate receptor localization in neurons with behavioral responses. We analyzed the pain-relieving effects of two delta receptor agonists with similar signaling potencies and efficacies, but distinct internalizing properties. An initial treatment with the high (SNC80) or low (AR-M100390) internalizing agonist equally reduced CFA-induced inflammatory pain. However, subsequent drug treatment produced highly distinct responses. Animals initially treated with SNC80 showed no analgesic response to a second dose of either delta receptor agonist. Concomitant receptor internalization and G-protein uncoupling were observed throughout the nervous system. This loss of function was temporary, since full DOR-eGFP receptor responses were restored 24 hours after SNC80 administration. In contrast, treatment with AR-M100390 resulted in retained analgesic response to a subsequent agonist injection, and ex vivo analysis showed that DOR-eGFP receptor remained G protein-coupled on the cell surface. Finally SNC80 but not AR-M100390 produced DOR-eGFP phosphorylation, suggesting that the two agonists produce distinct active receptor conformations in vivo which likely lead to differential receptor trafficking., Conclusions: Together our data show that delta agonists retain full analgesic efficacy when receptors remain on the cell surface. In contrast, delta agonist-induced analgesia is abolished following receptor internalization, and complete behavioral desensitization is observed. Overall these results establish that, in the context of pain control, receptor localization fully controls receptor function in vivo. This finding has both fundamental and therapeutic implications for slow-recycling GPCRs.
- Published
- 2009
- Full Text
- View/download PDF
32. Inflammatory pain is enhanced in delta opioid receptor-knockout mice.
- Author
-
Gavériaux-Ruff C, Karchewski LA, Hever X, Matifas A, and Kieffer BL
- Subjects
- Analgesics, Opioid pharmacology, Animals, Benzamides pharmacology, Brain physiopathology, Female, Freund's Adjuvant, Hyperalgesia chemically induced, Hyperalgesia genetics, Hyperalgesia metabolism, Inflammation genetics, Inflammation physiopathology, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Mutation genetics, Pain genetics, Pain physiopathology, Piperazines pharmacology, Receptors, Opioid, delta metabolism, Brain metabolism, Inflammation metabolism, Nociceptors metabolism, Pain metabolism, Receptors, Opioid, delta genetics
- Abstract
To examine the involvement of opioid receptors in inflammatory pain, we compared Complete Freund's Adjuvant-induced hyperalgesia in mice lacking mu, delta or kappa receptors under the same experimental conditions. Mechanical allodynia and thermal hyperalgesia were measured using von Frey filaments and the plantar test, respectively. All three receptor-knockout mice, as well as wild-type animals, developed inflammatory hyperalgesia following Complete Freund's Adjuvant administration. Mu-receptor mutants showed similar hyperalgesia to wild-types in the two tests. Kappa-receptor mutants exhibited enhanced mechanical allodynia compared with wild-type mice but similar thermal hyperalgesia. In contrast, mechanical allodynia and thermal hyperalgesia were both markedly augmented in delta-receptor mutants, indicating a role for an endogenous delta-receptor tone in the control of inflammatory pain. Treatment with the delta-selective agonist SNC80 produced antihyperalgesia, and this effect was abolished in the delta-receptor knockout mice. Altogether, these data demonstrate that delta receptors inhibit inflammatory pain when activated either endogenously or exogenously. We have previously shown enhanced neuropathic pain in delta-receptor knockout mice. The delta receptor definitely represents a promising target for treating chronic pain conditions.
- Published
- 2008
- Full Text
- View/download PDF
33. Delta-opioid receptors are critical for tricyclic antidepressant treatment of neuropathic allodynia.
- Author
-
Benbouzid M, Gavériaux-Ruff C, Yalcin I, Waltisperger E, Tessier LH, Muller A, Kieffer BL, Freund-Mercier MJ, and Barrot M
- Subjects
- Animals, Dose-Response Relationship, Drug, Mice, Mice, Inbred C57BL, Mice, Transgenic, Sciatica drug therapy, Amitriptyline pharmacology, Antidepressive Agents, Tricyclic pharmacology, Disease Models, Animal, Neuralgia drug therapy, Nortriptyline pharmacology, Receptors, Opioid, delta drug effects, Receptors, Opioid, delta genetics
- Abstract
Background: The therapeutic effect of antidepressant drugs against depression usually necessitates a chronic treatment. A large body of clinical evidence indicates that antidepressant drugs can also be highly effective against chronic neuropathic pain. However, the mechanism by which these drugs alleviate pain is still unclear., Methods: We used a murine model of neuropathic pain induced by sciatic nerve constriction to study the antiallodynic properties of a chronic treatment with the tricyclic antidepressants nortriptyline and amitriptyline. Using knockout and pharmacological approaches in mice, we determined the influence of delta-opioid receptors in the therapeutic action of chronic antidepressant treatment., Results: In our model, a chronic treatment by tricyclic antidepressant drugs totally suppresses the mechanical allodynia in neuropathic C57Bl/6J mice. This therapeutic effect can be acutely reversed by an injection of the delta-opioid receptor antagonist naltrindole. Moreover, the antiallodynic property of antidepressant treatment is absent in mice deficient for the delta-opioid receptor gene., Conclusions: The antiallodynic effect of chronic antidepressant treatment is mediated by a recruitment of the endogenous opioid system acting through delta-opioid receptors.
- Published
- 2008
- Full Text
- View/download PDF
34. Conditional gene targeting in the mouse nervous system: Insights into brain function and diseases.
- Author
-
Gavériaux-Ruff C and Kieffer BL
- Subjects
- Animals, Behavior, Animal, Gene Expression Regulation physiology, Integrases genetics, Integrases metabolism, Mice, Mice, Transgenic, Models, Animal, Phenotype, Brain metabolism, Brain Diseases genetics, Gene Targeting methods, Mice, Knockout, Nervous System chemistry, Nervous System Diseases physiopathology
- Abstract
Conditional gene knockout represents an extremely powerful approach to study the function of single genes in the nervous system. The Cre-LoxP system is the most advanced technology for spatial and temporal control of genetic inactivation, and there is rapid progress using this methodology in neuroscience research. In this approach, mice with LoxP sites flanking the gene of interest (floxed mice) are bred with transgenic mice expressing Cre recombinase under the control of a selected promoter (Cre mice). This promoter is critical in that it determines the time and site of Cre expression. Cre enzyme, in turn, recombines the floxed gene and produces gene knockout. Here we review Cre mouse lines that have been developed to target either the entire brain, selected brain areas, or specific neuronal populations. We then summarize phenotypic consequences of conditional gene targeting in the brain for more than 40 genes, as reported to date. For many broadly expressed genes, brain-restricted knockout has overcome lethality of conventional knockout (KO) and has highlighted a specific role of the encoded protein in some aspect of brain function. In the case of neural genes, data from null mutants in specific brain sites or neurons has refined our understanding of the role of individual molecules that regulate complex behaviors or synaptic plasticity within neural circuits. Among the many developing functional genomic approaches, conditional gene targeting in the mouse has become an excellent tool to elucidate the function of the approximately 5000 known or unknown genes that operate in the nervous system.
- Published
- 2007
- Full Text
- View/download PDF
35. Dissociation of analgesic and hormonal responses to forced swim stress using opioid receptor knockout mice.
- Author
-
Contet C, Gavériaux-Ruff C, Matifas A, Caradec C, Champy MF, and Kieffer BL
- Subjects
- Animals, Female, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Pain Measurement methods, Reaction Time physiology, Stress, Physiological psychology, Adrenocorticotropic Hormone metabolism, Analgesia methods, Corticosterone metabolism, Receptors, Opioid deficiency, Receptors, Opioid metabolism, Stress, Physiological metabolism, Swimming psychology
- Abstract
Exposure to stress triggers hormonal and behavioral responses. It has been shown that the endogenous opioid system plays a role in some physiological reactions to stress. The opioid system was described to mediate analgesia induced by mild stressors and to modulate the activation of the hypothalamic-pituitary-adrenal axis. Our study assessed the contribution of opioid receptors in stress-induced analgesia and adrenocorticotropic hormone (ACTH) and corticosterone release by a genetic approach. We performed a parallel analysis of mice deficient in mu, delta, or kappa opioid receptors, as well as of triple opioid receptor knockout mice, following exposure to a mild stress (3-min swim at 32 degrees C). In wild-type mice, stress elicited an increase in jumping latency on the hot plate, which was influenced by gender and genetic background. This analgesic response was reversed both by naloxone and by the triple mutation, and decreased in mu and delta opioid receptor knockout females. In wild-type females, stress also delayed front- and hindpaw behaviors in the hot plate test and increased tail-flick latency in the tail immersion test. Opioid receptor deletion however did not affect these stress responses. In addition, stress produced an increase in ACTH and corticosterone plasma levels. This endocrine response remained unchanged in all mutant strains. Therefore our data indicate that, under our stress conditions, the endogenous opioid system is recruited to produce some analgesia whereas it does not influence hypothalamic-pituitary-adrenal axis activity. This implies that brain circuits mediating analgesic and hormonal responses to stress can be dissociated.
- Published
- 2006
- Full Text
- View/download PDF
36. Knockin mice expressing fluorescent delta-opioid receptors uncover G protein-coupled receptor dynamics in vivo.
- Author
-
Scherrer G, Tryoen-Tóth P, Filliol D, Matifas A, Laustriat D, Cao YQ, Basbaum AI, Dierich A, Vonesh JL, Gavériaux-Ruff C, and Kieffer BL
- Subjects
- Animals, Brain metabolism, Endocytosis, Gene Expression, Green Fluorescent Proteins genetics, Kinetics, Mice, Mice, Transgenic, Transgenes genetics, Green Fluorescent Proteins metabolism, Receptors, Opioid, delta genetics, Receptors, Opioid, delta metabolism
- Abstract
The combination of fluorescent genetically encoded proteins with mouse engineering provides a fascinating means to study dynamic biological processes in mammals. At present, green fluorescent protein (GFP) mice were mainly developed to study gene expression patterns or cell morphology and migration. Here we used enhanced GFP (EGFP) to achieve functional imaging of a G protein-coupled receptor (GPCR) in vivo. We created mice where the delta-opioid receptor (DOR) is replaced by an active DOR-EGFP fusion. Confocal imaging revealed detailed receptor neuroanatomy throughout the nervous system of knock-in mice. Real-time imaging in primary neurons allowed dynamic visualization of drug-induced receptor trafficking. In DOR-EGFP animals, drug treatment triggered receptor endocytosis that correlated with the behavioral response. Mice with internalized receptors were insensitive to subsequent agonist administration, providing evidence that receptor sequestration limits drug efficacy in vivo. Direct receptor visualization in mice is a unique approach to receptor biology and drug design.
- Published
- 2006
- Full Text
- View/download PDF
37. Enhanced humoral response in kappa-opioid receptor knockout mice.
- Author
-
Gavériaux-Ruff C, Simonin F, Filliol D, and Kieffer BL
- Subjects
- Animals, B-Lymphocytes immunology, CD4-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes immunology, Cell Division drug effects, Cell Division immunology, Concanavalin A immunology, Concanavalin A pharmacology, Immunization, Immunoglobulin G blood, Immunoglobulin G genetics, Immunoglobulin G immunology, Immunoglobulin M blood, Immunoglobulin M genetics, Immunoglobulin M immunology, Killer Cells, Natural immunology, Lymphocyte Count, Mice, Mice, Knockout, Receptors, Opioid, kappa genetics, Spleen cytology, Spleen metabolism, Thymus Gland cytology, Thymus Gland metabolism, Antibody Formation genetics, Neuroimmunomodulation genetics, Receptors, Opioid, kappa deficiency, Spleen immunology, Thymus Gland immunology
- Abstract
Opiates are major analgesics and addictive drugs described also as immunomodulators. Here, we investigated the contribution of kappa-opioid receptor (KOR) activity in immunity in vivo by studying immune responses in KOR knockout mice. These animals displayed a modest reduction in thymus cellularity and CD4(+) cell ratio, parallel to a slight increase in immature CD4(+)CD8(+) lymphocytes. In spleen, KOR null animals showed augmented cell number with no change in cell distribution. T and B lymphocyte proliferative capabilities in vitro, Natural Killer activity and steady-state Ig levels were unchanged in KOR-/- mice. We immunized the mice with the antigen keyhole limpet hemocyanin (KLH). Compared to wild-type (WT) mice, KOR-/- animals produced significant higher levels of antigen-specific total Ig, IgM, IgG1 and IgG2a antibodies. This enhancement of humoral activity was not observed in mu-opioid receptor and delta-opioid receptor knockout animals. These results show that endogenous activation of kappa-opioid receptors may exert a tonic inhibition of antibody (Ab) response.
- Published
- 2003
- Full Text
- View/download PDF
38. Exploring the opioid system by gene knockout.
- Author
-
Kieffer BL and Gavériaux-Ruff C
- Subjects
- Animals, Mice, Mice, Knockout, Brain Chemistry physiology, Opioid Peptides genetics, Opioid Peptides metabolism, Receptors, Opioid genetics, Receptors, Opioid metabolism
- Abstract
The endogenous opioid system consists of three opioid peptide precursor genes encoding enkephalins (preproenkephalin, Penk), dynorphins (preprodynorphin, Pdyn) and beta-endorphin (betaend), proopiomelanocortin (POMC) and three receptor genes encoding mu-opiod receptor (MOR), delta-opiod receptor (DOR) and kappa-opiod receptor (KOR). In the past years, all six genes have been inactivated in mice by homologous recombination. The analysis of spontaneous behavior in mutant mice has demonstrated significant and distinct roles of each gene in modulating locomotion, pain perception and emotional behaviors. The observation of opposing phenotypes of MOR- and DOR-deficient mice in several behaviors highlights unexpected roles for DOR to be further explored genetically and using more specific delta compounds. The analysis of responses of mutant mice to exogenous opiates has definitely clarified the essential role of MOR in both morphine analgesia and addiction, and demonstrated that DOR and KOR remain promising targets for pain treatment. These studies also show that prototypic DOR agonists partially require MOR for their biological activity and provide some support for the postulated mu-delta interactions in vivo. Finally, data confirm and define a role for several genes of the opioid system in responses to other drugs of abuse, and the triple opioid receptor knockout mutant allows exploring non-classical opioid pharmacology. In summary, the study of null mutant mice has extended our previous knowledge of the opioid system by identifying the molecular players in opioid pharmacology and physiology. Future studies should involve parallel behavioral analysis of mice lacking receptors and peptides and will benefit from more sophisticated gene targeting approaches, including site-directed and anatomically-restricted mutations.
- Published
- 2002
- Full Text
- View/download PDF
39. Immunosuppression by delta-opioid antagonist naltrindole: delta- and triple mu/delta/kappa-opioid receptor knockout mice reveal a nonopioid activity.
- Author
-
Gavériaux-Ruff C, Filliol D, Simonin F, Matthes HW, and Kieffer BL
- Subjects
- Animals, Benzylidene Compounds pharmacology, Cell Division drug effects, Concanavalin A pharmacology, Dipeptides pharmacology, Interleukin-2 biosynthesis, Lymphocyte Culture Test, Mixed, Lymphocytes drug effects, Lymphocytes immunology, Mice, Mice, Knockout, Receptors, sigma genetics, T-Lymphocytes drug effects, T-Lymphocytes immunology, Immunosuppressive Agents pharmacology, Naltrexone analogs & derivatives, Naltrexone pharmacology, Narcotic Antagonists pharmacology, Receptors, Opioid, delta genetics, Receptors, Opioid, kappa genetics, Receptors, sigma antagonists & inhibitors, Tetrahydroisoquinolines
- Abstract
The delta-opioid antagonist naltrindole has been shown to inhibit graft rejection in vivo and suppress allogeneic mixed lymphocyte reaction (MLR) in vitro, similarly to cyclosporin A. We investigated whether this action is mediated by delta-opioid receptors using both genetic and pharmacological tools. Naltrindole and two related compounds, 7-benzylidene-7-dehydronaltrexone and naltriben, inhibited MLR performed with lymphocytes from wild-type and delta-opioid receptor knockout mice, with comparable potency. Furthermore, these compounds suppressed the proliferation of spleen cells from triple delta/mu/kappa-opioid receptor-deficient animals as well. Finally, the highly delta-selective, but structurally distinct, antagonist N,N-dimethyl-Dmt-Tic-OH and the general opioid antagonist naltrexone were inactive in the MLR assay. In conclusion, we demonstrate for the first time that the immunosuppressive activity of naltrindole and close derivatives is not mediated by any of the three cloned opioid receptors. Therefore, the postulated inhibitory activity of naltrindole in the graft rejection process is mediated by a target, which remains to be discovered.
- Published
- 2001
40. Orphanin FQ/nociceptin binds to functionally coupled ORL1 receptors on human immune cell lines and alters peripheral blood mononuclear cell proliferation.
- Author
-
Peluso J, Gavériaux-Ruff C, Matthes HW, Filliol D, and Kieffer BL
- Subjects
- Adult, Animals, Binding Sites drug effects, Binding Sites physiology, CHO Cells drug effects, CHO Cells metabolism, Cell Division drug effects, Cell Division physiology, Cell Line cytology, Cell Line drug effects, Cell Membrane drug effects, Cell Membrane metabolism, Cricetinae, Guanosine 5'-O-(3-Thiotriphosphate) pharmacokinetics, Humans, Immune System cytology, Immune System drug effects, Leukocytes, Mononuclear cytology, Leukocytes, Mononuclear drug effects, Neuroblastoma, Opioid Peptides immunology, Opioid Peptides pharmacology, Phytohemagglutinins pharmacology, Radioligand Assay, Receptors, Opioid drug effects, Receptors, Opioid immunology, Sulfur Radioisotopes pharmacology, Tumor Cells, Cultured drug effects, Tumor Cells, Cultured metabolism, Nociceptin Receptor, Nociceptin, Cell Line metabolism, Immune System metabolism, Leukocytes, Mononuclear metabolism, Opioid Peptides metabolism, Receptors, Opioid metabolism
- Abstract
Orphanin FQ/nociceptin (OFQ/N) has been shown to modulate nociception, responses to stress and anxiety. We investigated OFQ/N function in human immune cells. We find that monocytic U937, T lymphocytic CEM, and MOLT-4 cell lines express OFQ/N binding sites at levels comparable to that of human SH-SY5Y neuroblastoma cells. We show that OFQ/N receptors are functionally coupled to G proteins in these cells. Finally OFQ/N decreases proliferation of phytohemagglutinin-stimulated peripheral blood mononuclear cells in vitro at doses ranging from 10(-13) to 10(-8) M. Thus, our data suggest that OFQ/N and OFQ/N receptor may act as an immunomodulatory system.
- Published
- 2001
- Full Text
- View/download PDF
41. Mice deficient for delta- and mu-opioid receptors exhibit opposing alterations of emotional responses.
- Author
-
Filliol D, Ghozland S, Chluba J, Martin M, Matthes HW, Simonin F, Befort K, Gavériaux-Ruff C, Dierich A, LeMeur M, Valverde O, Maldonado R, and Kieffer BL
- Subjects
- Animals, Anxiety genetics, Binding Sites, Darkness, Depression genetics, Electroshock, Female, Light, Male, Mice, Mice, Knockout, Motor Activity drug effects, Naloxone pharmacology, Naltrexone analogs & derivatives, Naltrexone metabolism, Naltrexone pharmacology, Narcotic Antagonists metabolism, Narcotic Antagonists pharmacology, Pain Threshold drug effects, Phenotype, Receptors, Opioid, delta deficiency, Receptors, Opioid, delta genetics, Receptors, Opioid, kappa deficiency, Receptors, Opioid, kappa genetics, Receptors, Opioid, kappa metabolism, Receptors, Opioid, mu deficiency, Receptors, Opioid, mu genetics, Sex Characteristics, Swimming, Anxiety metabolism, Depression metabolism, Gene Deletion, Receptors, Opioid, delta metabolism, Receptors, Opioid, mu metabolism
- Abstract
The role of the opioid system in controlling pain, reward and addiction is well established, but its role in regulating other emotional responses is poorly documented in pharmacology. The mu-, delta- and kappa- opioid receptors (encoded by Oprm, Oprd1 and Oprk1, respectively) mediate the biological activity of opioids. We have generated Oprd1-deficient mice and compared the behavioural responses of mice lacking Oprd1, Oprm (ref. 6) and Oprk1 (ref. 7) in several models of anxiety and depression. Our data show no detectable phenotype in Oprk1-/- mutants, suggesting that kappa-receptors do not have a role in this aspect of opioid function; opposing phenotypes in Oprm-/- and Oprd1-/- mutants which contrasts with the classical notion of similar activities of mu- and delta-receptors; and consistent anxiogenic- and depressive-like responses in Oprd1-/- mice, indicating that delta-receptor activity contributes to improvement of mood states. We conclude that the Oprd1-encoded receptor, which has been proposed to be a promising target for the clinical management of pain, should also be considered in the treatment of drug addiction and other mood-related disorders.
- Published
- 2000
- Full Text
- View/download PDF
42. Down-regulation of mu-opioid receptor expression in rat oligodendrocytes during their development in vitro.
- Author
-
Tryoen-Toth P, Gavériaux-Ruff C, and Labourdette G
- Subjects
- Animals, Animals, Newborn, Blotting, Northern, Blotting, Southern, Cells, Cultured, Down-Regulation, Fibroblast Growth Factor 2 pharmacology, Gene Expression Regulation, Developmental, RNA, Messenger metabolism, Rats, Rats, Wistar, Receptors, Opioid, mu genetics, Tretinoin pharmacology, Oligodendroglia metabolism, Receptors, Opioid, mu metabolism
- Abstract
In the central nervous system, opioid receptors are found in neurons and also in glial cells. To gain more information on their presence and possibly on their function, we investigated the expression of mu-opioid receptors (MOR) during oligodendroglial cell development in two culture systems. In these models, during the first days, the cells are O-2A bipotential progenitor cells (also called OPCs; oligodendrocyte precursor cells), and then they differentiate into oligodendrocytes, which mature. In the first system, oligodendroglial cells, derived from newborn rat brain hemispheres, are grown in primary culture in the presence of a confluent layer of astrocytes, and they differentiate slowly. In the second, cells are specifically detached from the mixed cultures of the first system and are grown thereafter alone in secondary culture, a condition allowing a rapid cell differentiation. Under both conditions OPCs and immature oligodendrocytes were found to express a high level of MOR mRNA, whereas mature oligodendrocytes did not express it at all. The decrease of MOR expression during oligodendrocyte maturation was progressive, suggesting that it was not a primary effect of differentiation but an indirect secondary effect. Our study also shows that basic fibroblast growth factor (bFGF), which has been claimed by some authors to induce a dedifferentiation of the mature oligodendrocytes, and retinoic acid (RA), which had not been tested before, were not able to restore MOR expression in mature oligodendrocytes. These results indicate that bFGF and RA neither reverse the maturation process nor dedifferentiate the cells. However, RA was found to inhibit almost completely the expression of the myelin basic protein. The main result of this study is that MOR is expressed in progenitors and in immature oligodendrocytes, but not in mature oligodendrocytes. This suggests that MOR could be involved in some developmental process of the cells of the oligodendroglial lineage., (Copyright 2000 Wiley-Liss, Inc.)
- Published
- 2000
- Full Text
- View/download PDF
43. Abolition of morphine-immunosuppression in mice lacking the mu-opioid receptor gene.
- Author
-
Gavériaux-Ruff C, Matthes HW, Peluso J, and Kieffer BL
- Subjects
- Animals, Gene Expression Regulation immunology, Mice, Mice, Knockout, Morphine pharmacology, Narcotics pharmacology, Immunosuppression Therapy, Morphine immunology, Narcotics immunology, Receptors, Opioid, mu deficiency, Receptors, Opioid, mu genetics, Receptors, Opioid, mu immunology
- Abstract
Opiates are potent analgesic and addictive compounds. They also act on immune responses, and morphine, the prototypic opiate, has been repeatedly described as an immunosuppressive drug. Pharmacological studies have suggested that the inhibitory action of opiates on immunity is mediated by multiple opioid receptor sites but molecular evidence has remained elusive. Recently, three genes encoding mu- (MOR), delta-, and kappa-opioid receptors have been cloned. To investigate whether the mu-opioid receptor is functionally implicated in morphine immunosuppression in vivo, we have examined immune responses of mice with a genetic disruption of the MOR gene. In the absence of drug, there was no difference between wild-type and mutant mice with regard to a large number of immunological endpoints, suggesting that the lack of MOR-encoded protein has little consequence on immune status. Chronic morphine administration induced lymphoid organ atrophy, diminished the ratio of CD4(+)CD8(+) cells in the thymus and strongly reduced natural killer activity in wild-type mice. None of these effects was observed in MOR-deficient mice after morphine treatment. This demonstrates that the MOR gene product represents a major molecular target for morphine action on the immune system. Because our previous studies of MOR-deficient mice have shown that this receptor protein is also responsible for morphine analgesia, reward, and physical dependence, the present results imply that MOR-targeted therapeutic drugs that are developed for the treatment of pain or opiate addiction may concomitantly influence immune responses.
- Published
- 1998
- Full Text
- View/download PDF
44. Regulation of kappa-opioid receptor mRNA level by cyclic AMP and growth factors in cultured rat glial cells.
- Author
-
Tryoen-Toth P, Gavériaux-Ruff C, Maderspach K, and Labourdette G
- Subjects
- Animals, Animals, Newborn, Astrocytes drug effects, Astrocytes metabolism, Becaplermin, Blotting, Southern, Brain cytology, Cell Communication, Cells, Cultured, Colforsin pharmacology, Cyclic AMP analogs & derivatives, Cyclic AMP pharmacology, Leukemia Inhibitory Factor, Lipopolysaccharides pharmacology, Microglia drug effects, Microglia metabolism, Nerve Tissue Proteins biosynthesis, Neuroglia metabolism, Oligodendroglia drug effects, Oligodendroglia metabolism, Opioid Peptides physiology, Polymerase Chain Reaction, Proto-Oncogene Proteins c-sis, RNA, Messenger genetics, Rats, Rats, Wistar, Receptors, Opioid, kappa biosynthesis, Stem Cells drug effects, Stem Cells metabolism, Thionucleotides pharmacology, Cyclic AMP physiology, Fibroblast Growth Factor 2 pharmacology, Gene Expression Regulation drug effects, Growth Inhibitors pharmacology, Interleukin-6, Lymphokines pharmacology, Nerve Tissue Proteins genetics, Neuroglia drug effects, Platelet-Derived Growth Factor pharmacology, RNA, Messenger biosynthesis, Receptors, Opioid, kappa genetics
- Abstract
The mRNA of the kappa-opioid receptor (KOR) has been found recently in cultured astrocytes and in microglia. By using RT-PCR and Southern hybridization, we confirmed these observations and, in addition, we observed that KOR mRNA was expressed in oligodendrocytes and in the precursors of astrocytes and oligodendrocytes. KOR mRNA level was the highest in the immature astrocytes and decreased with their maturation. Very few data are available on the regulation of KOR level by extracellular signals. Therefore, we examined the effect of three growth factors known to be present in the adult brain, basic fibroblast growth factor (bFGF), platelet-derived growth factor (PDGF-BB) and leukemia inhibitory factor (LIF) and of two cyclic AMP (cAMP) generating systems, the cAMP analog, 8-(4-chlorophenylthio)-cAMP and forskolin, on this level. It was found that in astrocytes, KOR mRNA level decreased dramatically under the effect of cAMP and less under the effect of bFGF while it did not change significantly after LIF treatment. In oligodendrocytes, it also decreased with cAMP, but increased under the effect of bFGF and PDGF-BB. In microglia, a decrease was observed with cAMP and lipopolysaccharides (LPS), the most used activators of macrophages. These results shed new evidence on the expression of opioid receptor mRNA in the glial cells of the rat CNS. The regulation of KOR mRNA level under the effect of extracellular signals suggests that opioids take part in dynamic processes in glial cells, possibly related to glial-neuron communication.
- Published
- 1998
- Full Text
- View/download PDF
45. Distribution of nociceptin/orphanin FQ receptor transcript in human central nervous system and immune cells.
- Author
-
Peluso J, LaForge KS, Matthes HW, Kreek MJ, Kieffer BL, and Gavériaux-Ruff C
- Subjects
- Adult, Amino Acid Sequence, Base Sequence, Brain Chemistry, Cell Line, Cloning, Molecular, Female, Humans, Male, Middle Aged, Molecular Sequence Data, Nerve Tissue Proteins genetics, Neuroimmunomodulation, Opioid Peptides genetics, Organ Specificity, Polymerase Chain Reaction, RNA Splicing, RNA, Messenger analysis, Sequence Alignment, Sequence Homology, Nociceptin, Nerve Tissue Proteins analysis, Opioid Peptides analysis
- Abstract
We have examined the distribution of the opioid receptor-like-1 (ORL-1) transcript in the human CNS as well as human immune cells by RT-PCR and RNAse protection. The hORL-1 mRNA was distributed throughout the brain and particularly abundant in cortical areas, striatum, thalamus and hypothalamus. In the immune system, gene transcription was observed in normal circulating lymphocytes and monocytes as well as in T, B and monocytic cell lines. A splice variant, lacking 15 nucleotides at the junction between exon 1 and exon 2, showed a distribution similar to the already known ORL-1 transcript. Altogether these results show comparable expression levels of the hORL-1 gene in both nervous and immune systems, suggesting that the ORL-1-encoded receptor may participate to neuronal and non-neuronal physiological functions in humans.
- Published
- 1998
- Full Text
- View/download PDF
46. Detection of opioid receptor mRNA by RT-PCR reveals alternative splicing for the delta- and kappa-opioid receptors.
- Author
-
Gavériaux-Ruff C, Peluso J, Befort K, Simonin F, Zilliox C, and Kieffer BL
- Subjects
- Amino Acid Sequence, Animals, Base Sequence, Cell Line, DNA, Complementary isolation & purification, Exons, Genome, Humans, Mice, Mice, Inbred C57BL, Molecular Sequence Data, Transcription, Genetic, Alternative Splicing, Polymerase Chain Reaction methods, RNA, Messenger analysis, Receptors, Opioid, delta genetics, Receptors, Opioid, kappa genetics
- Abstract
The three mu-, delta- and kappa-opioid receptors have recently been cloned and characterized at the molecular level. Our analysis of opioid receptor transcripts by RT-PCR revealed two PCR products derived from delta and kappa mRNAs with size higher than expected from the known cDNA sequences. DNA sequencing showed additional nucleotides inserted between the known splice sites, indicating the possible existence of alternative splicing pathways for delta and kappa receptors. The novel delta-opioid receptor transcript is expressed in mouse brain and contains a 243 bp insertion. This additional sequence is located at the splice junction between the first and second coding exons and is encoded by a single exon located 9 kb upstream exon 2 in the mDOR gene. The other alternative transcript occurs in human monocytic and T lymphocytic cell lines and encodes a novel form of the kappa-opioid receptor. The PCR product presents a 23 bp deletion at the 3' end of exon 2 followed by a 246 bp insertion found between exons 2 and 3. In the hKOR gene, this insertion is encoded by two DNA segments. One of them is located 0.4 kb downstream exon 2 while the second is flanking exon 3 on the 5' side. Both novel putative delta and kappa exons present in-frame stop codons that would lead to truncated receptor proteins. A possible functional or regulatory role of these shorter proteins in opioid function remains to be determined.
- Published
- 1997
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.