Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 4100, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38773091

ABSTRACT

In most models of neuronal plasticity and memory, dopamine is thought to promote the long-term maintenance of Long-Term Potentiation (LTP) underlying memory processes, but not the initiation of plasticity or new information storage. Here, we used optogenetic manipulation of midbrain dopamine neurons in male DAT::Cre mice, and discovered that stimulating the Schaffer collaterals - the glutamatergic axons connecting CA3 and CA1 regions - of the dorsal hippocampus concomitantly with midbrain dopamine terminals within a 200 millisecond time-window triggers LTP at glutamatergic synapses. Moreover, we showed that the stimulation of this dopaminergic pathway facilitates contextual learning in awake behaving mice, while its inhibition hinders it. Thus, activation of midbrain dopamine can operate as a teaching signal that triggers NeoHebbian LTP and promotes supervised learning.


Subject(s)
Dopamine , Dopaminergic Neurons , Hippocampus , Learning , Long-Term Potentiation , Optogenetics , Ventral Tegmental Area , Animals , Long-Term Potentiation/physiology , Ventral Tegmental Area/physiology , Male , Dopamine/metabolism , Mice , Dopaminergic Neurons/physiology , Dopaminergic Neurons/metabolism , Hippocampus/physiology , Hippocampus/metabolism , Learning/physiology , Mice, Transgenic , CA1 Region, Hippocampal/physiology , CA1 Region, Hippocampal/cytology , Synapses/physiology , Synapses/metabolism , Mice, Inbred C57BL , Memory/physiology
2.
Mol Psychiatry ; 29(3): 718-729, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38123728

ABSTRACT

Chronic stress causes cognitive deficits, such as impairments in episodic-like hippocampus-dependent memory. Stress regulates an opioid-related neuropeptide named Nociceptin/Orphanin FQ (N/OFQ), the ligand of the G protein-coupled receptor NOP. Since this peptide has deleterious effects on memory, we hypothesized that the N/OFQ system could be a mediator of the negative effects of stress on memory. Chronic stress was mimicked by chronic exposure to corticosterone (CORT). The NOP receptor was either acutely blocked using selective antagonists, or knocked-down specifically in the hippocampus using genetic tools. Long-term memory was assessed in the object recognition (OR) and object location (OL) paradigms. Acute injection of NOP antagonists before learning had a negative impact on memory in naive mice whereas it restored memory performances in the chronic stress model. This rescue was associated with a normalization of neuronal cell activity in the CA3 part of the hippocampus. Chronic CORT induced an upregulation of the N/OFQ precursor in the hippocampus. Knock-down of the NOP receptor in the CA3/Dentate Gyrus region prevented memory deficits in the CORT model. These data demonstrate that blocking the N/OFQ system can be beneficial for long-term memory in a neuroendocrine model of chronic stress. We therefore suggest that NOP antagonists could be useful for the treatment of memory deficits in stress-related disorders.


Subject(s)
Corticosterone , Disease Models, Animal , Hippocampus , Memory, Long-Term , Nociceptin Receptor , Nociceptin , Opioid Peptides , Receptors, Opioid , Stress, Psychological , Animals , Receptors, Opioid/metabolism , Mice , Stress, Psychological/metabolism , Male , Hippocampus/metabolism , Hippocampus/drug effects , Opioid Peptides/metabolism , Memory, Long-Term/drug effects , Memory, Long-Term/physiology , Narcotic Antagonists/pharmacology , Mice, Inbred C57BL , Cognition/drug effects , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/drug therapy
3.
Commun Biol ; 6(1): 1168, 2023 11 16.
Article in English | MEDLINE | ID: mdl-37968381

ABSTRACT

Opioid-dependent immune-mediated analgesic effects have been broadly reported upon inflammation. In preclinical mouse models of intestinal inflammatory diseases, the local release of enkephalins (endogenous opioids) by colitogenic T lymphocytes alleviate inflammation-induced pain by down-modulating gut-innervating nociceptor activation in periphery. In this study, we wondered whether this immune cell-derived enkephalin-mediated regulation of the nociceptor activity also operates under steady state conditions. Here, we show that chimeric mice engrafted with enkephalin-deficient bone marrow cells exhibit not only visceral hypersensitivity but also an increase in both epithelial paracellular and transcellular permeability, an alteration of the microbial topography resulting in increased bacteria-epithelium interactions and a higher frequency of IgA-producing plasma cells in Peyer's patches. All these alterations of the intestinal homeostasis are associated with an anxiety-like behavior despite the absence of an overt inflammation as observed in patients with irritable bowel syndrome. Thus, our results show that immune cell-derived enkephalins play a pivotal role in maintaining gut homeostasis and normal behavior in mice. Because a defect in the mucosal opioid system remarkably mimics some major clinical symptoms of the irritable bowel syndrome, its identification might help to stratify subgroups of patients.


Subject(s)
Irritable Bowel Syndrome , Humans , Animals , Mice , Analgesics, Opioid , Enkephalins/genetics , Inflammation , Pain
4.
eNeuro ; 10(11)2023 Nov.
Article in English | MEDLINE | ID: mdl-37863658

ABSTRACT

Mitochondria are integrative hubs central to cellular adaptive pathways. Such pathways are critical in highly differentiated postmitotic neurons, the plasticity of which sustains brain function. Consequently, defects in mitochondria and in their dynamics appear instrumental in neurodegenerative diseases and may also participate in cognitive impairments. To directly test this hypothesis, we analyzed cognitive performances in a mouse mitochondria-based disease model, because of haploinsufficiency in the mitochondrial optic atrophy type 1 (OPA1) protein involved in mitochondrial dynamics. In males, we evaluated adult hippocampal neurogenesis parameters using immunohistochemistry. We performed a battery of tests to assess basal behavioral characteristics and cognitive performances, and tested putative treatments. While in dominant optic atrophy (DOA) mouse models, the known main symptoms are late onset visual deficits, we discovered early impairments in hippocampus-dependent spatial memory attributable to defects in adult neurogenesis. Moreover, less connected adult-born hippocampal neurons showed a decrease in mitochondrial content. Remarkably, voluntary exercise or pharmacological treatment targeting mitochondrial dynamics restored spatial memory in DOA mice. Altogether, our study identifies a crucial role for OPA1-dependent mitochondrial functions in adult neurogenesis, and thus in hippocampal-dependent cognitive functions. More generally, our findings show that adult neurogenesis is highly sensitive to mild mitochondrial defects, generating impairments in spatial memory that can be detected at an early stage and counterbalanced by physical exercise and pharmacological targeting of mitochondrial dynamics. Thus, amplification of mitochondrial function at an early stage appears beneficial for late-onset neurodegenerative diseases.


Subject(s)
Neurodegenerative Diseases , Optic Atrophy, Autosomal Dominant , Male , Mice , Animals , Spatial Memory , Mitochondria/metabolism , Neurogenesis/physiology , Neurons/metabolism , Optic Atrophy, Autosomal Dominant/metabolism , Hippocampus/metabolism , Neurodegenerative Diseases/metabolism
5.
Neurobiol Learn Mem ; 205: 107841, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37832816

ABSTRACT

The Nociceptin/Orphanin FQ (N/OFQ) system has been shown to modulate various aspects of long-term memory. It is therefore important to study the effects on memory impairment by nociceptin receptor (NOP) agonists under preclinical development. In the present study, we investigated the effect of systemic injection of two small molecule selective NOP agonists, AT-202 and AT-524, in the object location memory task in male and female mice. Since high doses of NOP agonists have been shown to induce sedation, we first determined the sedative doses for the two compounds and found them to be higher in female than in male mice. We then observed that sub-sedative doses of NOP agonists administered before learning, induced memory impairment during a test session performed 24 h later. Again, female mice were less sensitive to the amnesic effects than males. On the contrary, in male mice, NOP agonists did not produce amnesia when they were injected after learning, suggesting that they do not affect the consolidation of object location memory. Finally, repeated administration of high doses of NOP agonists over 7 days did not impair long-term spatial memory. Together, our data show for the first time that NOP receptor agonists impair the acquisition of object location memory with sex-dependent potency but do not affect memory consolidation, and that repeated stimulation of the receptor does not compromise long-term episodic-like spatial memory.


Subject(s)
Opioid Peptides , Receptors, Opioid , Female , Mice , Male , Animals , Opioid Peptides/pharmacology , Nociceptin Receptor , Learning , Memory, Long-Term , Hypnotics and Sedatives
6.
Cells ; 11(24)2022 12 18.
Article in English | MEDLINE | ID: mdl-36552881

ABSTRACT

The PSEN1 ΔE9 mutation causes a familial form of Alzheimer's disease (AD) by shifting the processing of amyloid precursor protein (APP) towards the generation of highly amyloidogenic Aß42 peptide. We have previously shown that the PSEN1 ΔE9 mutation in human-induced pluripotent stem cell (iPSC)-derived astrocytes increases Aß42 production and impairs cellular responses. Here, we injected PSEN1 ΔE9 mutant astrosphere-derived glial progenitors into newborn mice and investigated mouse behavior at the ages of 8, 12, and 16 months. While we did not find significant behavioral changes in younger mice, spatial learning and memory were paradoxically improved in 16-month-old PSEN1 ΔE9 glia-transplanted male mice as compared to age-matched isogenic control-transplanted animals. Memory improvement was associated with lower levels of soluble, but not insoluble, human Aß42 in the mouse brain. We also found a decreased engraftment of PSEN1 ΔE9 mutant cells in the cingulate cortex and significant transcriptional changes in both human and mouse genes in the hippocampus, including the extracellular matrix-related genes. Overall, the presence of PSEN1 ΔE9 mutant glia exerted a more beneficial effect on aged mouse brain than the isogenic control human cells likely as a combination of several factors.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Animals , Humans , Male , Mice , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Astrocytes/metabolism , Presenilin-1/genetics , Presenilin-1/metabolism , Spatial Learning , Aging
7.
Neuropharmacology ; 212: 109077, 2022 07 01.
Article in English | MEDLINE | ID: mdl-35513173

ABSTRACT

Nociceptin/orphanin FQ (N/OFQ) is the endogenous ligand of an inhibitory G protein coupled receptor named N/OFQ peptide receptor (NOP). Clinical and preclinical findings suggest that the blockade of the NOP signaling induces antidepressant-like effects. Additionally, the blockade of the NOP receptor during inescapable stress exposure prevented the acquisition of the helplessness phenotype, suggesting that NOP antagonists are able to increase stress resilience. BTRX-246040 (aka LY2940094) is a NOP receptor antagonist with high affinity, potency and selectivity for the NOP over classical opioid receptors. BTRX-246040 is under development for the treatment of depression, eating disorders and alcohol abuse and it already entered clinical trials. In the present study, the antidepressant effects of BTRX-246040 were evaluated in mice subjected to the forced swimming test and to the learned helplessness model of depression. Additionally, the ability of BTRX-246040 to prevent the development of the helpless behavior and to modulate adult hippocampal neurogenesis has been investigated. BTRX-246040 (30 mg/kg, i.p.) produced antidepressant-like effects in the forced swimming test and in the learned helplessness model. More interestingly, when given before the stress induction sessions it was able to prevent the development of the helplessness behavior. Under these experimental conditions, BTRX-246040 did not modulate adult hippocampal neurogenesis, neither in naive nor in stressed mice. This study, performed with a clinically viable ligand, further corroborates growing evidence indicating that the blockade of the NOP signaling may provide an innovative strategy for the treatment of stress related psychopathologies.


Subject(s)
Opioid Peptides , Receptors, Opioid , Animals , Antidepressive Agents/pharmacology , Hippocampus/metabolism , Ligands , Mice , Neurogenesis , Opioid Peptides/metabolism , Receptors, Opioid/metabolism
8.
NPJ Regen Med ; 6(1): 63, 2021 Oct 14.
Article in English | MEDLINE | ID: mdl-34650070

ABSTRACT

Tissue repair after injury in adult mammals, usually results in scarring and loss of function in contrast to lower vertebrates such as the newt and zebrafish that regenerate. Understanding the regulatory processes that guide the outcome of tissue repair is therefore a concerning challenge for regenerative medicine. In multiple regenerative animal species, the nerve dependence of regeneration is well established, but the nature of the innervation required for tissue regeneration remains largely undefined. Using our model of induced adipose tissue regeneration in adult mice, we demonstrate here that nociceptive nerves promote regeneration and their removal impairs tissue regeneration. We also show that blocking the receptor for the nociceptive neuropeptide calcitonin gene-related peptide (CGRP) inhibits regeneration, whereas CGRP administration induces regeneration. These findings reveal that peptidergic nociceptive neurons are required for adult mice tissue regeneration.

9.
Commun Biol ; 4(1): 1070, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34522000

ABSTRACT

G protein-coupled receptors (GPCRs) are notoriously difficult to detect in native tissues. In an effort to resolve this problem, we have developed a novel mouse model by fusing the hemagglutinin (HA)-epitope tag sequence to the amino-terminus of the µ-opioid receptor (MOP). Although HA-MOP knock-in mice exhibit reduced receptor expression, we found that this approach allowed for highly efficient immunodetection of low abundant GPCR targets. We also show that the HA-tag facilitates both high-resolution imaging and immunoisolation of MOP. Mass spectrometry (MS) confirmed post-translational modifications, most notably agonist-selective phosphorylation of carboxyl-terminal serine and threonine residues. MS also unequivocally identified the carboxyl-terminal 387LENLEAETAPLP398 motif, which is part of the canonical MOP sequence. Unexpectedly, MS analysis of brain lysates failed to detect any of the 15 MOP isoforms that have been proposed to arise from alternative splicing of the MOP carboxyl-terminus. For quantitative analysis, we performed multiple successive rounds of immunodepletion using the well-characterized rabbit monoclonal antibody UMB-3 that selectively detects the 387LENLEAETAPLP398 motif. We found that >98% of HA-tagged MOP contain the UMB-3 epitope indicating that virtually all MOP expressed in the mouse brain exhibit the canonical amino acid sequence.


Subject(s)
Hemagglutinins/genetics , Receptors, Opioid, mu/genetics , Amino Acid Sequence , Animals , Female , Hemagglutinins/metabolism , Male , Mice , Phosphorylation , Protein Isoforms , Receptors, Opioid, mu/chemistry , Receptors, Opioid, mu/metabolism
10.
Peptides ; 134: 170406, 2020 12.
Article in English | MEDLINE | ID: mdl-32920044

ABSTRACT

The neuropeptide FF2 (NPFF2) receptor, predominantly expressed in the central nervous system, plays an important role in the modulation of sensory input and opioid analgesia, as well as in locomotion, feeding, intestinal motility, reward, and the control of obesity. The NPFF2 receptor belongs to the RFamide peptide receptor family and to the G protein coupled receptor (GPCR) super family, but contrary to many other class A GPCRs, no 3D structure has been solved. Thus, it is essential to perform mutagenesis to gain information on the fine functioning of the NPFF2 receptor. In this study, we examined the role of aspartic acid (D) from the "D/ERY/F" motif found in the second intracellular loop (ICL2) and the role of the C-terminal end of the receptor in ligand binding and signal transduction. We found that mutation D3.49A does not impair binding capacities but inhibits G protein activation as well as adenylyl cyclase regulation. Truncation of the C terminal part of the receptor has different effects depending on the position of truncation. When truncation was realized downstream of the putative acylation site, ligand binding and signal transduction capabilities were not lost, contrary to total deletion of the C terminus, which totally impairs the activity of the receptor.


Subject(s)
Analgesics, Opioid/pharmacology , Neuropeptides/pharmacology , Receptors, Neuropeptide/chemistry , Receptors, Neuropeptide/metabolism , Amino Acid Sequence , Animals , Cells, Cultured , Cricetinae , Humans , Mutagenesis , Receptors, Neuropeptide/genetics , Signal Transduction/drug effects , Structure-Activity Relationship
11.
Eur J Pharmacol ; 875: 172884, 2020 May 15.
Article in English | MEDLINE | ID: mdl-31870829

ABSTRACT

Managing severe acute nociceptive pain in buprenorphine-maintained individuals for opioid use disorder management is challenging owing to the high affinity and very slow dissociation of buprenorphine from µ-opioid receptors that hinders the use of full agonist opioid analgesics. In a translational approach, the aim of this study was to use an animal setting to investigate the effects of a chronic high dose of buprenorphine treatment on nociceptive thresholds before and after applying a severe acute nociceptive traumatic surgery stimulus and to screen postoperative pharmacological analgesic strategies. A chronic treatment of mice with a high dose of buprenorphine (BUP HD, 2 × 200 µg/kg/day; i.p.) revealed significant mechanical allodynia. One and two days after having discontinued buprenorphine administration and having induced a severe nociceptive acute pain by a closed tibial fracture, acute administration of morphine at a dose which has analgesic effects in absence of pretreatment (4.5 mg/kg; i.p.), was ineffective to reduce pain in the BUP HD group. However, mimicking multimodal analgesia strategy used in human postoperative context, the combination of morphine (administered at the same dose) with a NMDA receptor antagonist (ketamine) or an NSAID (ketoprofen) produced antinociceptive responses in these animals. The mouse model of closed tibial fracture could be useful to identify analgesic strategies of postoperative pain for patients with chronic exposure to opioids and suffering from hyperalgesia.


Subject(s)
Acute Pain/drug therapy , Analgesics/pharmacology , Buprenorphine/adverse effects , Hyperalgesia/drug therapy , Narcotic Antagonists/adverse effects , Nociceptive Pain/drug therapy , Acute Pain/diagnosis , Acute Pain/etiology , Analgesics/therapeutic use , Animals , Buprenorphine/administration & dosage , Dose-Response Relationship, Drug , Drug Therapy, Combination/methods , Humans , Hyperalgesia/chemically induced , Hyperalgesia/diagnosis , Ketamine/pharmacology , Ketamine/therapeutic use , Ketoprofen/pharmacology , Ketoprofen/therapeutic use , Male , Mice , Morphine/pharmacology , Morphine/therapeutic use , Narcotic Antagonists/administration & dosage , Nociception/drug effects , Nociceptive Pain/diagnosis , Nociceptive Pain/etiology , Opioid-Related Disorders/drug therapy , Pain Management/methods , Pain Measurement , Pain Threshold/drug effects , Tibial Fractures/complications
12.
J Transl Med ; 17(1): 346, 2019 10 22.
Article in English | MEDLINE | ID: mdl-31640792

ABSTRACT

BACKGROUND: Tibial fracture is associated with inflammatory reaction leading to severe pain syndrome. Bradykinin receptor activation is involved in inflammatory reactions, but has never been investigated in fracture pain. METHODS: This study aims at defining the role of B1 and B2-kinin receptors (B1R and B2R) in a closed tibial fracture pain model by using knockout mice for B1R (B1KO) or B2R (B2KO) and wild-type (WT) mice treated with antagonists for B1R (SSR 240612 and R954) and B2R (HOE140) or vehicle. A cyclooxygenase (COX) inhibitor (ketoprofen) and an antagonist (SB366791) of Transient Receptor Potential Vaniloid1 (TRPV1) were also investigated since these pathways are associated with BK-induced pain in other models. The impact on mechanical and thermal hyperalgesia and locomotion was assessed by behavior tests. Gene expression of B1R and B2R and spinal cord expression of c-Fos were measured by RT-PCR and immunohistochemistry, respectively. RESULTS: B1KO and B2KO mice demonstrated a reduction in post-fracture pain sensitivity compared to WT mice that was associated with decreased c-Fos expression in the ipsilateral spinal dorsal horn in B2KO. B1R and B2R mRNA and protein levels were markedly enhanced at the fracture site. B1R and B2R antagonists and inhibition of COX and TRPV1 pathways reduced pain in WT. However, the analgesic effect of the COX-1/COX-2 inhibitor disappeared in B1KO and B2KO. In contrast, the analgesic effect of the TRPV1 antagonist persisted after gene deletion of either receptor. CONCLUSIONS: It is suggested that B1R and B2R activation contributes significantly to tibial fracture pain through COX. Hence, B1R and B2R antagonists appear potential therapeutic agents to manage post fracture pain.


Subject(s)
Pain/physiopathology , Receptor, Bradykinin B1/physiology , Receptor, Bradykinin B2/physiology , Tibial Fractures/physiopathology , Animals , Bradykinin B1 Receptor Antagonists/pharmacology , Bradykinin B2 Receptor Antagonists/pharmacology , Cyclooxygenase Inhibitors/pharmacology , Disease Models, Animal , Inflammation/etiology , Inflammation/pathology , Inflammation/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pain/drug therapy , Pain/prevention & control , Pain Measurement , Proto-Oncogene Proteins c-fos/biosynthesis , RNA, Messenger , Receptor, Bradykinin B1/deficiency , Receptor, Bradykinin B1/genetics , Receptor, Bradykinin B2/deficiency , Receptor, Bradykinin B2/genetics , TRPV Cation Channels/antagonists & inhibitors , Tibial Fractures/complications , Tibial Fractures/pathology , Translational Research, Biomedical
13.
Psychopharmacology (Berl) ; 236(7): 2069-2082, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30879119

ABSTRACT

RATIONALE: Pregabalin is a psychoactive drug indicated in the treatment of epilepsy, neuropathic pain, and generalized anxiety disorders. Pregabalin acts on different neurotransmission systems by inactivating the alpha2-delta subunit of voltage-gated calcium channels. In light of this pharmacological property, the hypothesis has been raised that pregabalin may regulate the mesolimbic dopamine pathway and thereby display a potential for misuse or abuse as recently observed in humans. Although some preclinical data support this possibility, the rewarding properties of gabapentinoid are still a matter for debate. OBJECTIVE: The aim of this work was to evaluate the rewarding properties of pregabalin and to determine its putative mechanism of action in healthy mice. RESULTS: Pregabalin alone (60 mg/kg; s.c.) produced a rewarding effect in the conditioned place preference (CPP) test albeit to a lower extent than cocaine (30 mg/kg; s.c.). Interestingly, when assessing locomotor activity in the CPP, the PGB60 group, similarly to the cocaine group, showed an increased locomotor activity. In vivo single unit extracellular recording showed that pregabalin had mixed effects on dopamine (DA) neuronal activity in the ventral tegmental area since it decreased the activity of 50% of neurons and increased 28.5% of them. In contrast, cocaine decreased 75% of VTA DA neuronal activity whereas none of the neurons were activated. Intracerebal microdialysis was then conducted in awake freely mice to determine to what extent such electrophysiological parameters influence the extracellular DA concentrations ([DA]ext) in the nucleus accumbens. Although pregabalin failed to modify this parameter, cocaine produced a robust increase (800%) in [DA]ext. CONCLUSIONS: Collectively, these electrophysiological and neurochemical experiments suggest that the rewarding properties of pregabalin result from a different mode of action than that observed with cocaine. Further experiments are warranted to determine whether such undesirable effects can be potentiated under pathological conditions such as neuropathic pain, mood disorders, or addiction and to identify the key neurotransmitter system involved.


Subject(s)
Dopamine/metabolism , Dopaminergic Neurons/metabolism , Nucleus Accumbens/metabolism , Pregabalin/pharmacology , Reward , Ventral Tegmental Area/metabolism , Analgesics/pharmacology , Animals , Cocaine/pharmacology , Conditioning, Classical/drug effects , Conditioning, Classical/physiology , Dopaminergic Neurons/drug effects , Male , Mice , Mice, Inbred C57BL , Microdialysis/methods , Motor Activity/drug effects , Motor Activity/physiology , Nucleus Accumbens/drug effects , Ventral Tegmental Area/drug effects
14.
PLoS One ; 14(3): e0213203, 2019.
Article in English | MEDLINE | ID: mdl-30845158

ABSTRACT

Although sphingosine-1-phosphate receptor 1 (S1P1) has been shown to trigger several S1P targeted functions such as immune cell trafficking, cell proliferation, migration, or angiogenesis, tools that allow the accurate detection of endogenous S1P1 localization and trafficking remain to be obtained and validated. In this study, we developed and characterized a novel monoclonal S1P1 antibody. Mice were immunized with S1P1 produced in the yeast Pichia pastoris and nine hybridoma clones producing monoclonal antibodies were created. Using different technical approaches including Western blot, immunoprecipitation and immunocytochemistry, we show that a selected clone, hereinafter referred to as 2B9, recognizes human and mouse S1P1 in various cell lineages. The interaction between 2B9 and S1P1 is specific over receptor subtypes, as the antibody does not binds to S1P2 or S1P5 receptors. Using cell-imaging methods, we demonstrate that 2B9 binds to an epitope located at the intracellular domain of S1P1; reveals cytosolic and membrane localization of the endogenous S1P1; and receptor internalization upon S1P or FTY720-P stimulation. Finally, loss of 2B9 signal upon knockdown of endogenous S1P1 by specific small interference RNAs further confirms its specificity. 2B9 was also able to detect S1P1 in human kidney and spinal cord tissue by immunohistochemistry. Altogether, our results suggest that 2B9 could be a useful tool to detect, quantify or localize low amounts of endogenous S1P1 in various physiological and pathological processes.


Subject(s)
Antibodies, Monoclonal/immunology , Breast Neoplasms/metabolism , Fibroblasts/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Image Processing, Computer-Assisted/methods , Receptors, Lysosphingolipid/immunology , Receptors, Lysosphingolipid/metabolism , Animals , Breast Neoplasms/pathology , Cells, Cultured , Female , Fibroblasts/cytology , Humans , Immunization , Kidney/metabolism , Mice , Microscopy, Fluorescence , Sphingosine-1-Phosphate Receptors , Spinal Cord/metabolism
15.
Sci Signal ; 12(574)2019 03 26.
Article in English | MEDLINE | ID: mdl-30914485

ABSTRACT

Agonists of the nociceptin/orphanin FQ opioid peptide (NOP) receptor, a member of the opioid receptor family, are under active investigation as novel analgesics, but their modes of signaling are less well characterized than those of other members of the opioid receptor family. Therefore, we investigated whether different NOP receptor ligands showed differential signaling or functional selectivity at the NOP receptor. Using newly developed phosphosite-specific antibodies to the NOP receptor, we found that agonist-induced NOP receptor phosphorylation occurred primarily at four carboxyl-terminal serine (Ser) and threonine (Thr) residues, namely, Ser346, Ser351, Thr362, and Ser363, and proceeded with a temporal hierarchy, with Ser346 as the first site of phosphorylation. G protein-coupled receptor kinases 2 and 3 (GRK2/3) cooperated during agonist-induced phosphorylation, which, in turn, facilitated NOP receptor desensitization and internalization. A comparison of structurally distinct NOP receptor agonists revealed dissociation in functional efficacies between G protein-dependent signaling and receptor phosphorylation. Furthermore, in NOP-eGFP and NOP-eYFP mice, NOP receptor agonists induced multisite phosphorylation and internalization in a dose-dependent and agonist-selective manner that could be blocked by specific antagonists. Our study provides new tools to study ligand-activated NOP receptor signaling in vitro and in vivo. Differential agonist-selective NOP receptor phosphorylation by chemically diverse NOP receptor agonists suggests that differential signaling by NOP receptor agonists may play a role in NOP receptor ligand pharmacology.


Subject(s)
Receptors, Opioid/agonists , Amino Acid Sequence , Animals , Antibody Specificity , Dose-Response Relationship, Drug , G-Protein-Coupled Receptor Kinase 2/metabolism , G-Protein-Coupled Receptor Kinase 3/metabolism , Genes, Reporter , HEK293 Cells , Humans , Ligands , Mice , Models, Molecular , Phosphorylation , Phosphoserine/analysis , Phosphothreonine/analysis , Protein Processing, Post-Translational , Receptors, Opioid/immunology , Receptors, Opioid/metabolism , Recombinant Proteins/metabolism , Structure-Activity Relationship , Nociceptin Receptor
16.
Neurotox Res ; 36(2): 257-267, 2019 Aug.
Article in English | MEDLINE | ID: mdl-30215161

ABSTRACT

Generation of new neurons is a tightly regulated process that involves several intrinsic and extrinsic factors. Among them, a metabolic switch from glycolysis to oxidative phosphorylation, together with mitochondrial remodeling, has emerged as crucial actors of neurogenesis. However, although accumulating data raise the importance of mitochondrial morphology and function in neural stem cell proliferation and differentiation during development, information regarding the contribution of mitochondria to adult neurogenesis processes remains limited. In the present review, we discuss recent evidence covering the importance of mitochondrial morphology, function, and energy metabolism in the regulation of neuronal development and adult neurogenesis, and their impact on memory processes.


Subject(s)
Mitochondria/physiology , Neural Stem Cells/physiology , Neurogenesis/physiology , Neurons/physiology , Adult , Animals , Cell Differentiation/physiology , Humans
17.
Handb Exp Pharmacol ; 254: 259-278, 2019.
Article in English | MEDLINE | ID: mdl-30430261

ABSTRACT

Nociceptin/orphanin FQ (N/OFQ) is an endogenous neuropeptide of 17 amino acids, related to opioid peptides but with its own receptor, distinct from conventional opioid receptors, the ORL1 or NOP receptor. The NOP receptor is a G protein-coupled receptor which activates Gi/o proteins and thus induces an inhibition of neuronal activity. The peptide and its receptor are widely expressed in the central nervous system with a high density of receptors in regions involved in learning and memory. This review describes the consequences of the pharmacological manipulation of the N/OFQ system by NOP receptor ligands on learning processes and on the consolidation of various types of long-term memory. We also discuss the role of endogenous N/OFQ release in the modulation of learning and memory. Finally we propose several putative neuronal mechanisms taking place at the level of the hippocampus and amygdala and possibly underlying the behavioral amnestic or promnesic effects of NOP ligands.


Subject(s)
Opioid Peptides/metabolism , Receptors, Opioid/chemistry , Learning , Memory , Opioid Peptides/chemistry , Nociceptin
18.
Neuropharmacology ; 125: 39-49, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28705439

ABSTRACT

Several neuropeptidergic systems act as modulators of cognitive performances. Among them, nociceptin, an opioid-like peptide also known as orphanin FQ (N/OFQ), has recently gained attention. Stimulation of its receptor, the N/OFQ opioid receptor (NOP), which is expressed in brain regions involved in emotion, memory and stress response, has inhibitory effects on the acquisition and/or consolidation of spatial and emotional memory in rodents. Recently, N/OFQ was also proposed to be linked to the pathogenesis of Post-Traumatic Stress Disorder in humans. However, until now the effect of the activation of the N/OFQ-NOP system on already consolidated memory, such as during retrieval and reconsolidation phases, has never been explored. In the present study, we investigated the consequences of systemic injection of NOP agonists or i.c.v. injection of the N/OFQ peptide on the retrieval and the reconsolidation of contextual fear memory in mice. We demonstrate that the activation of the N/OFQ system impairs the reconsolidation of context-dependent but not cue-dependent aversive memories. We also show that this amnestic effect is associated with decreased c-Fos expression in the hippocampus and amygdala. Our data thus provide the first evidence that the NOP receptor could be targeted during the reconsolidation process to weaken maladaptive memories. The N/OFQ-NOP system might constitute in the future an interesting pharmacological target for interfering with so-called "pathological memories", in particular those involving maladaptive contextual memories.


Subject(s)
Fear/physiology , Memory Consolidation/physiology , Receptors, Opioid/metabolism , Amygdala/drug effects , Amygdala/metabolism , Analgesics, Opioid/pharmacology , Animals , Conditioning, Psychological/drug effects , Conditioning, Psychological/physiology , Cues , Dose-Response Relationship, Drug , Fear/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , Imidazoles/pharmacology , Male , Memory Consolidation/drug effects , Mice, Inbred C57BL , Neurons/drug effects , Neurons/metabolism , Opioid Peptides/pharmacology , Proto-Oncogene Proteins c-fos/metabolism , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, Opioid/agonists , Spiro Compounds/pharmacology , Time Factors , Nociceptin Receptor , Nociceptin
19.
J Transl Med ; 13: 208, 2015 Jul 02.
Article in English | MEDLINE | ID: mdl-26136113

ABSTRACT

BACKGROUND: Diabetic neuropathy is one of the most common complications of diabetes and causes various problems in daily life. The aim of this study was to assess the effect of regional anaesthesia on post surgery opioid induced hyperalgesia in diabetic and non-diabetic mice. METHODS: Diabetic and non-diabetic mice underwent plantar surgery. Levobupivacaine and sufentanil were used before surgery, for sciatic nerve block (regional anaesthesia) and analgesia, respectively. Diabetic and non-diabetic groups were each randomly assigned to three subgroups: control, no sufentanil and no levobupivacaine; sufentanil and no levobupivacaine; sufentanil and levobupivacaine. Three tests were used to assess pain behaviour: mechanical nociception; thermal nociception and guarding behaviours using a pain scale. RESULTS: Sufentanil, alone or in combination with levobupivacaine, produced antinociceptive effects shortly after administration. Subsequently, sufentanil induced hyperalgesia in diabetic and non-diabetic mice. Opioid-induced hyperalgesia was enhanced in diabetic mice. Levobupivacaine associated to sufentanil completely prevented hyperalgesia in both groups of mice. CONCLUSION: The results suggest that regional anaesthesia can decrease opioid-induced hyperalgesia in diabetic as well as in non-diabetic mice. These observations may be clinically relevant for the management of diabetic patients.


Subject(s)
Analgesics, Opioid/adverse effects , Anesthesia, Conduction , Bupivacaine/analogs & derivatives , Diabetes Mellitus, Experimental/complications , Hyperalgesia/chemically induced , Hyperalgesia/drug therapy , Postoperative Complications/drug therapy , Animals , Bupivacaine/pharmacology , Bupivacaine/therapeutic use , Diabetes Mellitus, Experimental/pathology , Hyperalgesia/pathology , Inflammation/pathology , Levobupivacaine , Male , Mice, Inbred C57BL , Nerve Block , Sciatic Nerve/drug effects , Sciatic Nerve/pathology , Skin/pathology , Wound Healing/drug effects
20.
FEBS Lett ; 589(18): 2401-8, 2015 Aug 19.
Article in English | MEDLINE | ID: mdl-26226422

ABSTRACT

Many in vitro data have shown that the efficacy of several opioid drugs is correlated with differential mu-opioid (MOP) receptor phosphorylation. Label-free semiquantitative on-line nanoflow liquid chromatography-tandem mass spectrometry (nanoLC-MS/MS) analyses were performed to compare the endogenous MOP receptor phosphorylation patterns of mice administered with morphine, etonitazene and fentanyl. The analysis identified S363, T370 and S375 as phosphorylated residues in the carboxy-terminus. Only T370 and S375 were regulated by agonists, with a higher propensity to promote double phosphorylation for high efficacy agonists. Our study provides confirmation that differential agonist-driven multi-site phosphorylation of MOP receptor occurs in vivo and validate the use of MS to study endogenous GPCR phosphorylation.


Subject(s)
Brain/metabolism , Phosphoproteins/metabolism , Proteomics , Receptors, Opioid, mu/metabolism , Amino Acid Sequence , Analgesics, Opioid/pharmacology , Animals , Brain/drug effects , Mass Spectrometry , Mice , Molecular Sequence Data , Phosphoproteins/agonists , Phosphoproteins/chemistry , Phosphorylation/drug effects , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL