Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Psychiatry ; 29(3): 718-729, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38123728

RESUMEN

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.


Asunto(s)
Corticosterona , Modelos Animales de Enfermedad , Hipocampo , Memoria a Largo Plazo , Receptor de Nociceptina , Nociceptina , Péptidos Opioides , Receptores Opioides , Estrés Psicológico , Animales , Receptores Opioides/metabolismo , Ratones , Estrés Psicológico/metabolismo , Masculino , Hipocampo/metabolismo , Hipocampo/efectos de los fármacos , Péptidos Opioides/metabolismo , Memoria a Largo Plazo/efectos de los fármacos , Memoria a Largo Plazo/fisiología , Antagonistas de Narcóticos/farmacología , Ratones Endogámicos C57BL , Cognición/efectos de los fármacos , Disfunción Cognitiva/metabolismo , Disfunción Cognitiva/tratamiento farmacológico
2.
Neurobiol Learn Mem ; 205: 107841, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37832816

RESUMEN

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.


Asunto(s)
Péptidos Opioides , Receptores Opioides , Femenino , Ratones , Masculino , Animales , Péptidos Opioides/farmacología , Receptor de Nociceptina , Aprendizaje , Memoria a Largo Plazo , Hipnóticos y Sedantes
3.
J Transl Med ; 17(1): 346, 2019 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-31640792

RESUMEN

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.


Asunto(s)
Dolor/fisiopatología , Receptor de Bradiquinina B1/fisiología , Receptor de Bradiquinina B2/fisiología , Fracturas de la Tibia/fisiopatología , Animales , Antagonistas del Receptor de Bradiquinina B1/farmacología , Antagonistas del Receptor de Bradiquinina B2/farmacología , Inhibidores de la Ciclooxigenasa/farmacología , Modelos Animales de Enfermedad , Inflamación/etiología , Inflamación/patología , Inflamación/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Dolor/tratamiento farmacológico , Dolor/prevención & control , Dimensión del Dolor , Proteínas Proto-Oncogénicas c-fos/biosíntesis , ARN Mensajero , Receptor de Bradiquinina B1/deficiencia , Receptor de Bradiquinina B1/genética , Receptor de Bradiquinina B2/deficiencia , Receptor de Bradiquinina B2/genética , Canales Catiónicos TRPV/antagonistas & inhibidores , Fracturas de la Tibia/complicaciones , Fracturas de la Tibia/patología , Investigación Biomédica Traslacional
4.
Handb Exp Pharmacol ; 254: 259-278, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30430261

RESUMEN

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.


Asunto(s)
Péptidos Opioides/metabolismo , Receptores Opioides/química , Aprendizaje , Memoria , Péptidos Opioides/química , Nociceptina
5.
J Biol Chem ; 289(49): 33754-66, 2014 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-25326382

RESUMEN

The neuropeptide FF2 (NPFF2) receptor belongs to the rhodopsin family of G protein-coupled receptors and mediates the effects of several related RFamide neuropeptides. One of the main pharmacological interests of this system resides in its ability to regulate endogenous opioid systems, making it a potential target to reduce the negative effects of chronic opioid use. Phosphorylation of intracellular residues is the most extensively studied post-translational modification regulating G protein-coupled receptor activity. However, until now, no information concerning NPFF2 receptor phosphorylation is available. In this study, we combined mass spectrometric analysis and site-directed mutagenesis to analyze for the first time the phosphorylation pattern of the NPFF2 receptor and the role of the various phosphorylation sites in receptor signaling, desensitization, and trafficking in a SH-SY5Y model cell line. We identified the major, likely GRK-dependent, phosphorylation cluster responsible for acute desensitization, (412)TNST(415) at the end of the C terminus of the receptor, and additional sites involved in desensitization ((372)TS(373)) and internalization (Ser(395)). We thus demonstrate the key role played by phosphorylation in the regulation of NPFF2 receptor activity and trafficking. Our data also provide additional evidence supporting the concept that desensitization and internalization are partially independent processes relying on distinct phosphorylation patterns.


Asunto(s)
Neuronas/metabolismo , Oligopéptidos/metabolismo , Procesamiento Proteico-Postraduccional , Receptores de Neuropéptido/química , Secuencia de Aminoácidos , Línea Celular Tumoral , Humanos , Cinética , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Neuronas/citología , Oligopéptidos/química , Mapeo Peptídico , Fosforilación , Transporte de Proteínas , Receptores de Neuropéptido/genética , Receptores de Neuropéptido/metabolismo , Alineación de Secuencia , Transducción de Señal
6.
J Biol Chem ; 289(41): 28697-706, 2014 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-25183007

RESUMEN

The dynamic organization of G protein-coupled receptors in the plasma membrane is suspected of playing a role in their function. The regulation of the diffusion mode of the mu-opioid (MOP) receptor was previously shown to be agonist-specific. Here we investigate the regulation of MOP receptor diffusion by heterologous activation of other G protein-coupled receptors and characterize the dynamic properties of the MOP receptor within the heterodimer MOP/neuropeptide FF (NPFF2) receptor. The data show that the dynamics and signaling of the MOP receptor in SH-SY5Y cells are modified by the activation of α2-adrenergic and NPFF2 receptors, but not by the activation of receptors not described to interact with the opioid receptor. By combining, for the first time, fluorescence recovery after photobleaching at variable radius experiments with bimolecular fluorescence complementation, we show that the MOP/NPFF2 heterodimer adopts a specific diffusion behavior that corresponds to a mix of the dynamic properties of both MOP and NPFF2 receptors. Altogether, the data suggest that heterologous regulation is accompanied by a specific organization of receptors in the membrane.


Asunto(s)
Analgésicos Opioides/farmacología , Transporte de Proteínas/efectos de los fármacos , Receptor Cross-Talk/efectos de los fármacos , Receptores Adrenérgicos alfa 2/metabolismo , Receptores de Neuropéptido/metabolismo , Receptores Opioides mu/metabolismo , Línea Celular Tumoral , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Clonidina/farmacología , Difusión , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacología , Recuperación de Fluorescencia tras Fotoblanqueo , Colorantes Fluorescentes , Regulación de la Expresión Génica , Humanos , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuropéptido Y/farmacología , Oligopéptidos/farmacología , Multimerización de Proteína , Receptores Adrenérgicos alfa 2/genética , Receptores de Neuropéptido/genética , Receptores Opioides mu/genética , Transducción de Señal
7.
J Transl Med ; 13: 208, 2015 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-26136113

RESUMEN

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.


Asunto(s)
Analgésicos Opioides/efectos adversos , Anestesia de Conducción , Bupivacaína/análogos & derivados , Diabetes Mellitus Experimental/complicaciones , Hiperalgesia/inducido químicamente , Hiperalgesia/tratamiento farmacológico , Complicaciones Posoperatorias/tratamiento farmacológico , Animales , Bupivacaína/farmacología , Bupivacaína/uso terapéutico , Diabetes Mellitus Experimental/patología , Hiperalgesia/patología , Inflamación/patología , Levobupivacaína , Masculino , Ratones Endogámicos C57BL , Bloqueo Nervioso , Nervio Ciático/efectos de los fármacos , Nervio Ciático/patología , Piel/patología , Cicatrización de Heridas/efectos de los fármacos
8.
Anal Biochem ; 453: 50-4, 2014 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-24613258

RESUMEN

A new radioiodinated photoaffinity compound, [(125)I]YE(Bpa)WSLAAPQRFNH2, derived from a peptide present in the rat neuropeptide FF (NPFF) precursor was synthesized, and its binding characteristics were investigated on a neuroblastoma clone, SH-SY5Y, stably expressing rat NPFF2 receptors tagged with the T7 epitope. The binding of the probe was saturable and revealed a high-affinity interaction (KD=0.24nM) with a single class of binding sites. It was also able to affinity label NPFF2 receptor in a specific and efficient manner given that 38% of the bound radioligand at saturating concentration formed a wash-resistant binding after ultraviolet (UV) irradiation. Photoaffinity labeling with [(125)I]YE(Bpa)WSLAAPQRFamide showed two molecular forms of NPFF2 receptor with apparent molecular weights of 140 and 95kDa in a 2:1 ratio. The comparison of the results between photoaffinity labeling and Western blot analysis suggests that all receptor forms bind the probe irreversibly with the same efficiency. On membranes of mouse olfactory bulb, only the high molecular weight form of NPFF2 receptor is observed. [(125)I]YE(Bpa)WSLAAPQRFamide is an excellent radioiodinated peptidic ligand for direct and selective labeling of NPFF2 receptors in vitro.


Asunto(s)
Marcadores de Afinidad/química , Oligopéptidos/química , Fenilalanina/análogos & derivados , Ensayo de Unión Radioligante/métodos , Receptores de Neuropéptido/análisis , Marcadores de Afinidad/síntesis química , Secuencia de Aminoácidos , Animales , Humanos , Radioisótopos de Yodo , Ligandos , Ratones , Ratones Endogámicos C57BL , Peso Molecular , Neuroblastoma/metabolismo , Bulbo Olfatorio/metabolismo , Oligopéptidos/análisis , Fenilalanina/química , Fotólisis , Ratas , Receptores de Neuropéptido/química , Receptores de Neuropéptido/metabolismo , Células Tumorales Cultivadas
9.
Nat Commun ; 15(1): 4100, 2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38773091

RESUMEN

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.


Asunto(s)
Dopamina , Neuronas Dopaminérgicas , Hipocampo , Aprendizaje , Potenciación a Largo Plazo , Optogenética , Área Tegmental Ventral , Animales , Potenciación a Largo Plazo/fisiología , Área Tegmental Ventral/fisiología , Masculino , Dopamina/metabolismo , Ratones , Neuronas Dopaminérgicas/fisiología , Neuronas Dopaminérgicas/metabolismo , Hipocampo/fisiología , Hipocampo/metabolismo , Aprendizaje/fisiología , Ratones Transgénicos , Región CA1 Hipocampal/fisiología , Región CA1 Hipocampal/citología , Sinapsis/fisiología , Sinapsis/metabolismo , Ratones Endogámicos C57BL , Memoria/fisiología
10.
J Biol Chem ; 287(16): 12736-49, 2012 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-22375000

RESUMEN

Neuropeptide FF (NPFF) interacts with specific receptors to modulate opioid functions in the central nervous system. On dissociated neurons and neuroblastoma cells (SH-SY5Y) transfected with NPFF receptors, NPFF acts as a functional antagonist of µ-opioid (MOP) receptors by attenuating the opioid-induced inhibition of calcium conductance. In the SH-SY5Y model, MOP and NPFF(2) receptors have been shown to heteromerize. To understand the molecular mechanism involved in the anti-opioid activity of NPFF, we have investigated the phosphorylation status of the MOP receptor using phospho-specific antibody and mass spectrometry. Similarly to direct opioid receptor stimulation, activation of the NPFF(2) receptor by [D-Tyr-1-(NMe)Phe-3]NPFF (1DMe), an analog of NPFF, induced the phosphorylation of Ser-377 of the human MOP receptor. This heterologous phosphorylation was unaffected by inhibition of second messenger-dependent kinases and, contrarily to homologous phosphorylation, was prevented by inactivation of G(i/o) proteins by pertussis toxin. Using siRNA knockdown we could demonstrate that 1DMe-induced Ser-377 cross-phosphorylation and MOP receptor loss of function were mediated by the G protein receptor kinase GRK2. In addition, mass spectrometric analysis revealed that the phosphorylation pattern of MOP receptors was qualitatively similar after treatment with the MOP agonist Tyr-D-Ala-Gly (NMe)-Phe-Gly-ol (DAMGO) or after treatment with the NPFF agonist 1DMe, but the level of multiple phosphorylation was more intense after DAMGO. Finally, NPFF(2) receptor activation was sufficient to recruit ß-arrestin2 to the MOP receptor but not to induce its internalization. These data show that NPFF-induced heterologous desensitization of MOP receptor signaling is mediated by GRK2 and could involve transphosphorylation within the heteromeric receptor complex.


Asunto(s)
Quinasa 2 del Receptor Acoplado a Proteína-G/metabolismo , Receptores de Neuropéptido/metabolismo , Receptores Opioides mu/metabolismo , Secuencia de Aminoácidos , Analgésicos Opioides/farmacología , Arrestinas/metabolismo , Línea Celular Tumoral , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacología , Quinasa 2 del Receptor Acoplado a Proteína-G/genética , Técnicas de Silenciamiento del Gen , Proteínas de Unión al GTP Heterotriméricas/metabolismo , Humanos , Datos de Secuencia Molecular , Neuroblastoma , Fosforilación/fisiología , Receptores Acoplados a Proteínas G/metabolismo , Receptores Opioides mu/agonistas , Sistemas de Mensajero Secundario/efectos de los fármacos , Sistemas de Mensajero Secundario/fisiología , Serina/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , beta-Arrestinas
11.
Biol Chem ; 394(3): 361-8, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23324378

RESUMEN

Postoperative pain management is a clinical challenge that can be complicated by opioid-induced hyperalgesia (OIH). Kinin receptors could mediate both the acute and chronic phases of inflammation and pain. A few recent studies suggest that dynorphin A could maintain neuropathic pain by activating the bradykinin (BK) receptor. Thus, the effect of a single administration of sufentanil (a µ-opioid receptor agonist) was investigated in a model of carrageenan-induced inflammatory pain using three strains of mice, i.e., knockout mice for one kinin receptor, B1R or B2R (B1KO, B2KO), and wild-type C57/BL6J mice (WT) treated with either a B1R (R954) or a B2R antagonist (HOE140) or a KKS inhibitor (aprotinin). Pain was assessed and compared between the different groups using two behavioral tests exploring mechanical (von Frey filaments) and thermal (Hargreaves test) sensitivity. Pretreatment with sufentanil induced a sustained increase in pain sensitivity with a delayed return to baseline values characterizing an OIH in carrageenan-injected mice only. Sufentanil-induced OIH was not observed in B2KO but persisted in B1KO and was blunted by aprotinin and the B2R antagonist only. Collectively, our data indicate that the B2R receptor and BK synthesis or availability are essential peripheral steps in the mechanism leading to OIH in a pain context.


Asunto(s)
Analgésicos Opioides/efectos adversos , Hiperalgesia/inducido químicamente , Hiperalgesia/fisiopatología , Inflamación/inducido químicamente , Dolor/fisiopatología , Receptor de Bradiquinina B2/metabolismo , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor de Bradiquinina B1/metabolismo , Sufentanilo/efectos adversos
12.
eNeuro ; 10(11)2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37863658

RESUMEN

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.


Asunto(s)
Enfermedades Neurodegenerativas , Atrofia Óptica Autosómica Dominante , Masculino , Ratones , Animales , Memoria Espacial , Mitocondrias/metabolismo , Neurogénesis/fisiología , Neuronas/metabolismo , Atrofia Óptica Autosómica Dominante/metabolismo , Hipocampo/metabolismo , Enfermedades Neurodegenerativas/metabolismo
13.
Commun Biol ; 6(1): 1168, 2023 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-37968381

RESUMEN

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.


Asunto(s)
Síndrome del Colon Irritable , Humanos , Animales , Ratones , Analgésicos Opioides , Encefalinas/genética , Inflamación , Dolor
14.
Neuropharmacology ; 212: 109077, 2022 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-35513173

RESUMEN

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.


Asunto(s)
Péptidos Opioides , Receptores Opioides , Animales , Antidepresivos/farmacología , Hipocampo/metabolismo , Ligandos , Ratones , Neurogénesis , Péptidos Opioides/metabolismo , Receptores Opioides/metabolismo
15.
Cells ; 11(24)2022 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-36552881

RESUMEN

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.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides , Animales , Humanos , Masculino , Ratones , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Astrocitos/metabolismo , Presenilina-1/genética , Presenilina-1/metabolismo , Aprendizaje Espacial , Envejecimiento
16.
NPJ Regen Med ; 6(1): 63, 2021 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-34650070

RESUMEN

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.

17.
Commun Biol ; 4(1): 1070, 2021 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-34522000

RESUMEN

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.


Asunto(s)
Hemaglutininas/genética , Receptores Opioides mu/genética , Secuencia de Aminoácidos , Animales , Femenino , Hemaglutininas/metabolismo , Masculino , Ratones , Fosforilación , Isoformas de Proteínas , Receptores Opioides mu/química , Receptores Opioides mu/metabolismo
18.
Synapse ; 64(9): 672-81, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20336629

RESUMEN

Neuropeptide FF (NPFF) is a neurotransmitter known to modulate opioid-induced analgesia, sensitization, and reward. The expression of the immediate early gene c-Fos was analyzed to map the distribution of neurons whose activity is regulated by central administration of the NPFF(2)-selective agonist dNPA in naive mice and in animals who had received a systemic injection of morphine. The number of c-Fos positive nuclei was quantified in 28 brain regions. Intracerebro-ventricular injection of 1 nmol dNPA alone produced an overall inhibition of basal c-Fos expression in the brain with a statistically significant decrease in the lateral ventral part of the bed nucleus of the stria terminalis, the medial preoptic area, and the medial parvicellular part of the paraventricular nucleus of the hypothalamus. In contrast, intraperitoneal injection of morphine 5 mg.kg(-1) induced a statistically significant increase in c-Fos expression in the prelimbic cortex, the nucleus accumbens core and shell, the ventral pallidum, the lateral hypothalamus, and the nucleus of the tractus solitarius. dNPA counteracted morphine effect only in the nucleus accumbens shell and the ventral pallidum. The inhibitory effects of a low dose of dNPA in the hypothalamus and its afferents suggest that NPFF(2) receptors negatively regulate the hypothalamic-pituitary-adrenal axis in mouse. Moreover, our study identified the nucleus accumbens shell and ventral pallidum as putative sites of interaction between NPFF and opioid systems in relation with the modulation of acute morphine rewarding and locomotor effects.


Asunto(s)
Analgésicos Opioides/farmacología , Química Encefálica/efectos de los fármacos , Morfina/farmacología , Neuronas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-fos/metabolismo , Receptores de Neuropéptido/agonistas , Animales , Encéfalo/anatomía & histología , Femenino , Inmunohistoquímica , Inyecciones Intraventriculares , Ratones , Ratones Endogámicos C57BL
19.
Peptides ; 134: 170406, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32920044

RESUMEN

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.


Asunto(s)
Analgésicos Opioides/farmacología , Neuropéptidos/farmacología , Receptores de Neuropéptido/química , Receptores de Neuropéptido/metabolismo , Secuencia de Aminoácidos , Animales , Células Cultivadas , Cricetinae , Humanos , Mutagénesis , Receptores de Neuropéptido/genética , Transducción de Señal/efectos de los fármacos , Relación Estructura-Actividad
20.
Eur J Pharmacol ; 875: 172884, 2020 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-31870829

RESUMEN

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.


Asunto(s)
Dolor Agudo/tratamiento farmacológico , Analgésicos/farmacología , Buprenorfina/efectos adversos , Hiperalgesia/tratamiento farmacológico , Antagonistas de Narcóticos/efectos adversos , Dolor Nociceptivo/tratamiento farmacológico , Dolor Agudo/diagnóstico , Dolor Agudo/etiología , Analgésicos/uso terapéutico , Animales , Buprenorfina/administración & dosificación , Relación Dosis-Respuesta a Droga , Quimioterapia Combinada/métodos , Humanos , Hiperalgesia/inducido químicamente , Hiperalgesia/diagnóstico , Ketamina/farmacología , Ketamina/uso terapéutico , Cetoprofeno/farmacología , Cetoprofeno/uso terapéutico , Masculino , Ratones , Morfina/farmacología , Morfina/uso terapéutico , Antagonistas de Narcóticos/administración & dosificación , Nocicepción/efectos de los fármacos , Dolor Nociceptivo/diagnóstico , Dolor Nociceptivo/etiología , Trastornos Relacionados con Opioides/tratamiento farmacológico , Manejo del Dolor/métodos , Dimensión del Dolor , Umbral del Dolor/efectos de los fármacos , Fracturas de la Tibia/complicaciones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA