Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
1.
Nature ; 598(7882): 646-651, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34646022

RESUMEN

µ-Opioid peptide receptor (MOPR) stimulation alters respiration, analgesia and reward behaviour, and can induce substance abuse and overdose1-3. Despite its evident importance, the endogenous mechanisms for MOPR regulation of consummatory behaviour have remained unknown4. Here we report that endogenous MOPR regulation of reward consumption in mice acts through a specific dorsal raphe to nucleus accumbens projection. MOPR-mediated inhibition of raphe terminals is necessary and sufficient to determine consummatory response, while select enkephalin-containing nucleus accumbens ensembles are engaged prior to reward consumption, suggesting that local enkephalin release is the source of the endogenous MOPR ligand. Selective modulation of nucleus accumbens enkephalin neurons and CRISPR-Cas9-mediated disruption of enkephalin substantiate this finding. These results isolate a fundamental endogenous opioid circuit for state-dependent consumptive behaviour and suggest alternative mechanisms for opiate modulation of reward.


Asunto(s)
Analgésicos Opioides/farmacología , Núcleo Accumbens/fisiología , Receptores Opioides mu/fisiología , Recompensa , Animales , Encefalinas , Femenino , Masculino , Ratones , Ratones Noqueados
2.
J Neurosci ; 38(11): 2780-2795, 2018 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-29459374

RESUMEN

A homozygous nonsense mutation in the cereblon (CRBN) gene results in autosomal recessive, nonsyndromic intellectual disability that is devoid of other phenotypic features, suggesting a critical role of CRBN in mediating learning and memory. In this study, we demonstrate that adult male Crbn knock-out (CrbnKO) mice exhibit deficits in hippocampal-dependent learning and memory tasks that are recapitulated by focal knock-out of Crbn in the adult dorsal hippocampus, with no changes in social or repetitive behavior. Cellular studies identify deficits in long-term potentiation at Schaffer collateral CA1 synapses. We further show that Crbn is robustly expressed in the mouse hippocampus and CrbnKO mice exhibit hyperphosphorylated levels of AMPKα (Thr172). Examination of processes downstream of AMP-activated protein kinase (AMPK) finds that CrbnKO mice have a selective impairment in mediators of the mTORC1 translation initiation pathway in parallel with lower protein levels of postsynaptic density glutamatergic proteins and higher levels of excitatory presynaptic markers in the hippocampus with no change in markers of the unfolded protein response or autophagy pathways. Acute pharmacological inhibition of AMPK activity in adult CrbnKO mice rescues learning and memory deficits and normalizes hippocampal mTORC1 activity and postsynaptic glutamatergic proteins without altering excitatory presynaptic markers. Thus, this study identifies that loss of Crbn results in learning, memory, and synaptic defects as a consequence of exaggerated AMPK activity, inhibition of mTORC1 signaling, and decreased glutamatergic synaptic proteins. Thus, CrbnKO mice serve as an ideal model of intellectual disability to further explore molecular mechanisms of learning and memory.SIGNIFICANCE STATEMENT Intellectual disability (ID) is one of the most common neurodevelopmental disorders. The cereblon (CRBN) gene has been linked to autosomal recessive, nonsyndromic ID, characterized by an intelligence quotient between 50 and 70 but devoid of other phenotypic features, making cereblon an ideal protein for the study of the fundamental aspects of learning and memory. Here, using the cereblon knock-out mouse model, we show that cereblon deficiency disrupts learning, memory, and synaptic function via AMP-activated protein kinase hyperactivity, downregulation of mTORC1, and dysregulation of excitatory synapses, with no changes in social or repetitive behaviors, consistent with findings in the human population. This establishes the cereblon knock-out mouse as a model of pure ID without the confounding behavioral phenotypes associated with other current models of ID.


Asunto(s)
Discapacidad Intelectual/genética , Discapacidad Intelectual/fisiopatología , Discapacidades para el Aprendizaje/genética , Discapacidades para el Aprendizaje/fisiopatología , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Trastornos de la Memoria/genética , Trastornos de la Memoria/fisiopatología , Proteínas del Tejido Nervioso/genética , Proteínas Adaptadoras Transductoras de Señales , Animales , Región CA1 Hipocampal/fisiopatología , Potenciales Postsinápticos Excitadores/genética , Hipocampo/metabolismo , Hipocampo/fisiopatología , Discapacidad Intelectual/tratamiento farmacológico , Discapacidades para el Aprendizaje/tratamiento farmacológico , Potenciación a Largo Plazo/genética , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina/biosíntesis , Trastornos de la Memoria/tratamiento farmacológico , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas del Tejido Nervioso/biosíntesis , Inhibidores de Proteínas Quinasas/uso terapéutico , Conducta Social
3.
Mol Cell Proteomics ; 14(10): 2564-76, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26149443

RESUMEN

Despite its efficacy, the use of morphine for the treatment of chronic pain remains limited because of the rapid development of tolerance, dependence and ultimately addiction. These undesired effects are thought to be because of alterations in synaptic transmission and neuroplasticity within the reward circuitry including the striatum. In this study we used subcellular fractionation and quantitative proteomics combined with computational approaches to investigate the morphine-induced protein profile changes at the striatal postsynaptic density. Over 2,600 proteins were identified by mass spectrometry analysis of subcellular fractions enriched in postsynaptic density associated proteins from saline or morphine-treated striata. Among these, the levels of 34 proteins were differentially altered in response to morphine. These include proteins involved in G-protein coupled receptor signaling, regulation of transcription and translation, chaperones, and protein degradation pathways. The altered expression levels of several of these proteins was validated by Western blotting analysis. Using Genes2Fans software suite we connected the differentially expressed proteins with proteins identified within the known background protein-protein interaction network. This led to the generation of a network consisting of 116 proteins with 40 significant intermediates. To validate this, we confirmed the presence of three proteins predicted to be significant intermediates: caspase-3, receptor-interacting serine/threonine protein kinase 3 and NEDD4 (an E3-ubiquitin ligase identified as a neural precursor cell expressed developmentally down-regulated protein 4). Because this morphine-regulated network predicted alterations in proteasomal degradation, we examined the global ubiquitination state of postsynaptic density proteins and found it to be substantially altered. Together, these findings suggest a role for protein degradation and for the ubiquitin/proteasomal system in the etiology of opiate dependence and addiction.


Asunto(s)
Analgésicos Opioides/farmacología , Morfina/farmacología , Proteínas del Tejido Nervioso/metabolismo , Animales , Caspasa 3/metabolismo , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Masculino , Ubiquitina-Proteína Ligasas Nedd4 , Proteolisis , Proteómica , Ratas Sprague-Dawley , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Transmisión Sináptica/efectos de los fármacos , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación/efectos de los fármacos
4.
J Neurosci ; 35(35): 12217-31, 2015 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-26338332

RESUMEN

Pain management in opioid abusers engenders ethical and practical difficulties for clinicians, often resulting in pain mismanagement. Although chronic opioid administration may alter pain states, the presence of pain itself may alter the propensity to self-administer opioids, and previous history of drug abuse comorbid with chronic pain promotes higher rates of opioid misuse. Here, we tested the hypothesis that inflammatory pain leads to increased heroin self-administration resulting from altered mu opioid receptor (MOR) regulation of mesolimbic dopamine (DA) transmission. To this end, the complete Freund's adjuvant (CFA) model of inflammation was used to assess the neurochemical and functional changes induced by inflammatory pain on MOR-mediated mesolimbic DA transmission and on rat intravenous heroin self-administration under fixed ratio (FR) and progressive ratio (PR) schedules of reinforcement. In the presence of inflammatory pain, heroin intake under an FR schedule was increased for high, but attenuated for low, heroin doses with concomitant alterations in mesolimbic MOR function suggested by DA microdialysis. Consistent with the reduction in low dose FR heroin self-administration, inflammatory pain reduced motivation for a low dose of heroin, as measured by responding under a PR schedule of reinforcement, an effect dissociable from high heroin dose PR responding. Together, these results identify a connection between inflammatory pain and loss of MOR function in the mesolimbic dopaminergic pathway that increases intake of high doses of heroin. These findings suggest that pain-induced loss of MOR function in the mesolimbic pathway may promote opioid dose escalation and contribute to opioid abuse-associated phenotypes. SIGNIFICANCE STATEMENT: This study provides critical new insights that show that inflammatory pain alters heroin intake through a desensitization of MORs located within the VTA. These findings expand our knowledge of the interactions between inflammatory pain and opioid abuse liability, and should help to facilitate the development of novel and safer opioid-based strategies for treating chronic pain.


Asunto(s)
Analgésicos Opioides/administración & dosificación , Encefalina Ala(2)-MeFe(4)-Gli(5)/administración & dosificación , Dolor , Receptores Opioides mu/metabolismo , Área Tegmental Ventral/metabolismo , Potenciales de Acción/efectos de los fármacos , Animales , Condicionamiento Operante/efectos de los fármacos , Modelos Animales de Enfermedad , Antagonistas de Aminoácidos Excitadores/farmacología , Glicinérgicos/farmacología , Heroína/administración & dosificación , Hiperalgesia/tratamiento farmacológico , Inflamación/inducido químicamente , Inflamación/complicaciones , Potenciales Postsinápticos Inhibidores/efectos de los fármacos , Masculino , Neuronas/efectos de los fármacos , Dolor/tratamiento farmacológico , Dolor/patología , Dolor/psicología , Umbral del Dolor/efectos de los fármacos , Quinoxalinas/farmacología , Ratas , Ratas Sprague-Dawley , Estricnina/farmacología , Sacarosa/administración & dosificación , Área Tegmental Ventral/efectos de los fármacos , Área Tegmental Ventral/patología
5.
J Neurosci ; 34(2): 527-38, 2014 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-24403152

RESUMEN

Learned associations between environmental cues and morphine use play an important role in the maintenance and/or relapse of opioid addiction. Although previous studies suggest that context-dependent morphine treatment alters glutamatergic transmission and synaptic plasticity in the hippocampus, their role in morphine conditioned place preference (CPP) and reinstatement remains unknown. We investigated changes in synaptic plasticity and NMDAR expression in the hippocampus after the expression, extinction, and reinstatement of morphine CPP. Here we report that morphine CPP is associated with increased basal synaptic transmission, impaired hippocampal long-term potentiation (LTP), and increased synaptic expression of the NR1 and NR2b NMDAR subunits. Changes in synaptic plasticity, synaptic NR1 and NR2b expression, and morphine CPP were absent when morphine was not paired with a specific context. Furthermore, hippocampal LTP was impaired and synaptic NR2b expression was increased after extinction of morphine CPP, indicating that these alterations in plasticity may be involved in the mechanisms underlying the learning of drug-environment associations. After extinction of morphine CPP, a priming dose of morphine was sufficient to reinstate morphine CPP and was associated with LTP that was indistinguishable from saline control groups. In contrast, morphine CPP extinguished mice that received a saline priming dose did not show CPP and had disrupted hippocampal LTP. Finally, we found that reinstatement of morphine CPP was prevented by the selective blockade of the NR2b subunit in the hippocampus. Together, these data suggest that alterations in synaptic plasticity and glutamatergic transmission play an important role in the reinstatement of morphine CPP.


Asunto(s)
Extinción Psicológica/fisiología , Hipocampo/fisiología , Potenciación a Largo Plazo/fisiología , Dependencia de Morfina/fisiopatología , Transmisión Sináptica/fisiología , Animales , Condicionamiento Clásico , Señales (Psicología) , Electrofisiología , Potenciales Postsinápticos Excitadores/fisiología , Immunoblotting , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores de N-Metil-D-Aspartato/metabolismo
6.
Neuropsychopharmacology ; 49(6): 915-923, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38374364

RESUMEN

Opioid use disorder is a chronic relapsing disorder encompassing misuse, dependence, and addiction to opioid drugs. Long term maintenance of associations between the reinforcing effects of the drug and the cues associated with its intake are a leading cause of relapse. Indeed, exposure to the salient drug-associated cues can lead to drug cravings and drug seeking behavior. The dorsal hippocampus (dHPC) and locus coeruleus (LC) have emerged as important structures for linking the subjective rewarding effects of opioids with environmental cues. However, their role in cue-induced reinstatement of opioid use remains to be further elucidated. In this study, we showed that chemogenetic inhibition of excitatory dHPC neurons during re-exposure to drug-associated cues significantly attenuates cue-induced reinstatement of morphine-seeking behavior. In addition, the same manipulation reduced reinstatement of sucrose-seeking behavior but failed to alter memory recall in the object location task. Finally, intact activity of tyrosine hydroxylase (TH) LC-dHPCTh afferents is necessary to drive cue induced reinstatement of morphine-seeking as inhibition of this pathway blunts cue-induced drug-seeking behavior. Altogether, these studies show an important role of the dHPC and LC-dHPCTh pathway in mediating cue-induced reinstatement of opioid seeking.


Asunto(s)
Señales (Psicología) , Comportamiento de Búsqueda de Drogas , Hipocampo , Locus Coeruleus , Autoadministración , Animales , Locus Coeruleus/efectos de los fármacos , Locus Coeruleus/metabolismo , Masculino , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Ratas , Femenino , Comportamiento de Búsqueda de Drogas/efectos de los fármacos , Comportamiento de Búsqueda de Drogas/fisiología , Morfina/farmacología , Morfina/administración & dosificación , Ratas Sprague-Dawley , Vías Nerviosas/efectos de los fármacos , Vías Nerviosas/fisiología , Analgésicos Opioides/farmacología , Analgésicos Opioides/administración & dosificación , Trastornos Relacionados con Opioides/fisiopatología , Extinción Psicológica/efectos de los fármacos , Extinción Psicológica/fisiología , Condicionamiento Operante/efectos de los fármacos , Condicionamiento Operante/fisiología
7.
Nat Commun ; 15(1): 750, 2024 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-38286800

RESUMEN

The hippocampus is pivotal in integrating emotional processing, learning, memory, and reward-related behaviors. The dorsal hippocampus (dHPC) is particularly crucial for episodic, spatial, and associative memory, and has been shown to be necessary for context- and cue-associated reward behaviors. The nucleus accumbens (NAc), a central structure in the mesolimbic reward pathway, integrates the salience of aversive and rewarding stimuli. Despite extensive research on dHPC→NAc direct projections, their sufficiency in driving reinforcement and reward-related behavior remains to be determined. Our study establishes that activating excitatory neurons in the dHPC is sufficient to induce reinforcing behaviors through its direct projections to the dorso-medial subregion of the NAc shell (dmNAcSh). Notably, dynorphin-containing neurons specifically contribute to dHPC-driven reinforcing behavior, even though both dmNAcSh dynorphin- and enkephalin-containing neurons are activated with dHPC stimulation. Our findings unveil a pathway governing reinforcement, advancing our understanding of the hippocampal circuity's role in reward-seeking behaviors.


Asunto(s)
Dinorfinas , Núcleo Accumbens , Éteres Fosfolípidos , Núcleo Accumbens/fisiología , Hipocampo/fisiología , Recompensa , Neuronas/fisiología
8.
Br J Pharmacol ; 180(7): 793-796, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36813266

RESUMEN

LINKED ARTICLES: This article is part of a themed issue on Advances in Opioid Pharmacology at the Time of the Opioid Epidemic. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v180.7/issuetoc.


Asunto(s)
Analgésicos Opioides , Trastornos Relacionados con Opioides , Humanos , Analgésicos Opioides/uso terapéutico , Trastornos Relacionados con Opioides/tratamiento farmacológico , Epidemia de Opioides
9.
Pain Rep ; 8(4): e1082, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37388406

RESUMEN

Introduction: Epidemiological studies have shown that there is a relation between pain and alcohol use disorder (AUD). Persistent pain is directly correlated with an increment in alcohol consumption and an increased risk of developing an AUD. Greater levels of pain intensity and unpleasantness are associated with higher levels of relapse, an increase in alcohol consumption, rates of hazardous drinking, and delay to seek for treatment. However, this interaction has not been deeply studied in the preclinical setting. Methods: Here, we aim to evaluate how inflammatory pain affects levels of alcohol drinking in male and female rats with a history of alcohol. For that, we used an intermittent access 2-bottle choice paradigm combined with the complete Freund Adjuvant (CFA) model of inflammatory pain. Results: Our results show that CFA-induced inflammatory pain does not alter total intake of 20% alcohol in male or female rats. Interestingly, in males, the presence of CFA-induced inflammatory pain blunts the decrease of alcohol intake when higher concentrations of alcohol are available, whereas it does not have an effect on intake at any concentration in female rats. Conclusion: Altogether, this study provides relevant data and constitutes an important contribution to the study of pain and AUD and it highlights the necessity to design better behavioral paradigms in animal models that are more translational and reflect current epidemiological findings.

10.
eNeuro ; 10(3)2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36849260

RESUMEN

In females, the hippocampus, a critical brain region for coordination of learning, memory, and behavior, displays altered physiology and behavioral output across the estrous or menstrual cycle. However, the molecular effectors and cell types underlying these observed cyclic changes have only been partially characterized to date. Recently, profiling of mice null for the AMPA receptor trafficking gene Cnih3 have demonstrated estrous-dependent phenotypes in dorsal hippocampal synaptic plasticity, composition, and learning/memory. We therefore profiled dorsal hippocampal transcriptomes from female mice in each estrous cycle stage, and contrasted it with that of males, across wild-type (WT) and Cnih3 mutants. In wild types, we identified only subtle differences in gene expression between the sexes, while comparing estrous stages to one another revealed up to >1000 differentially expressed genes (DEGs). These estrous-responsive genes are especially enriched in gene markers of oligodendrocytes and the dentate gyrus, and in functional gene sets relating to estrogen response, potassium channels, and synaptic gene splicing. Surprisingly, Cnih3 knock-outs (KOs) showed far broader transcriptomic differences between estrous cycle stages and males. Moreover, Cnih3 knock-out drove subtle but extensive expression changes accentuating sex differential expression at diestrus and estrus. Altogether, our profiling highlights cell types and molecular systems potentially impacted by estrous-specific gene expression patterns in the adult dorsal hippocampus, enabling mechanistic hypothesis generation for future studies of sex-differential neuropsychiatric function and dysfunction. Moreover, these findings suggest an unrecognized role of Cnih3 in buffering against transcriptional effects of estrous, providing a candidate molecular mechanism to explain estrous-dependent phenotypes observed with Cnih3 loss.


Asunto(s)
Ciclo Estral , Hipocampo , Animales , Femenino , Masculino , Ratones , Ciclo Estral/genética , Hipocampo/metabolismo , Aprendizaje , Plasticidad Neuronal , Transcriptoma
11.
J Neurosci ; 31(45): 16279-91, 2011 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-22072679

RESUMEN

Glutamatergic systems, including AMPA receptors (AMPARs), are involved in opiate-induced neuronal and behavioral plasticity, although the mechanisms underlying these effects are not fully understood. In the present study, we investigated the effects of repeated morphine administration on AMPAR expression, synaptic plasticity, and context-dependent behavioral sensitization to morphine. We found that morphine treatment produced changes of synaptic AMPAR expression in the hippocampus, a brain area that is critically involved in learning and memory. These changes could be observed 1 week after the treatment, but only when mice developed context-dependent behavioral sensitization to morphine in which morphine treatment was associated with drug administration environment. Context-dependent behavioral sensitization to morphine was also associated with increased basal synaptic transmission and disrupted hippocampal long-term potentiation (LTP), whereas these effects were less robust when morphine administration was not paired with the drug administration environment. Interestingly, some effects may be related to the prior history of morphine exposure in the drug-associated environment, since alterations of AMPAR expression, basal synaptic transmission, and LTP were observed in mice that received a saline challenge 1 week after discontinuation of morphine treatment. Furthermore, we demonstrated that phosphorylation of GluA1 AMPAR subunit plays a critical role in the acquisition and expression of context-dependent behavioral sensitization, as this behavior is blocked by a viral vector that disrupts GluA1 phosphorylation. These data provide evidence that glutamatergic signaling in the hippocampus plays an important role in context-dependent sensitization to morphine and supports further investigation of glutamate-based strategies for treating opiate addiction.


Asunto(s)
Hipocampo/efectos de los fármacos , Dependencia de Morfina/metabolismo , Morfina/farmacología , Actividad Motora/efectos de los fármacos , Narcóticos/farmacología , Receptores AMPA/metabolismo , Alanina/genética , Análisis de Varianza , Animales , Conducta Animal/efectos de los fármacos , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular , Homólogo 4 de la Proteína Discs Large , Relación Dosis-Respuesta a Droga , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Técnicas de Transferencia de Gen , Proteínas Fluorescentes Verdes/genética , Guanilato-Quinasas/metabolismo , Etiquetado Corte-Fin in Situ/métodos , Potenciación a Largo Plazo/efectos de los fármacos , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Mutación/genética , Proteínas Nucleares/metabolismo , Fosforilación/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Distribución Aleatoria , Receptores AMPA/genética , Serina/genética , Fracciones Subcelulares/efectos de los fármacos , Fracciones Subcelulares/metabolismo
12.
Front Syst Neurosci ; 16: 1014768, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36341476

RESUMEN

Decades of research advances have established a central role for endogenous opioid systems in regulating reward processing, mood, motivation, learning and memory, gastrointestinal function, and pain relief. Endogenous opioid systems are present ubiquitously throughout the central and peripheral nervous system. They are composed of four families, namely the µ (MOPR), κ (KOPR), δ (DOPR), and nociceptin/orphanin FQ (NOPR) opioid receptors systems. These receptors signal through the action of their endogenous opioid peptides ß-endorphins, dynorphins, enkephalins, and nociceptins, respectfully, to maintain homeostasis under normal physiological states. Due to their prominent role in pain regulation, exogenous opioids-primarily targeting the MOPR, have been historically used in medicine as analgesics, but their ability to produce euphoric effects also present high risks for abuse. The ability of pain and opioid use to perturb endogenous opioid system function, particularly within the central nervous system, may increase the likelihood of developing opioid use disorder (OUD). Today, the opioid crisis represents a major social, economic, and public health concern. In this review, we summarize the current state of the literature on the function, expression, pharmacology, and regulation of endogenous opioid systems in pain. Additionally, we discuss the adaptations in the endogenous opioid systems upon use of exogenous opioids which contribute to the development of OUD. Finally, we describe the intricate relationship between pain, endogenous opioid systems, and the proclivity for opioid misuse, as well as potential advances in generating safer and more efficient pain therapies.

13.
Pain ; 163(5): 809-819, 2022 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-34510137

RESUMEN

ABSTRACT: Many analgesics inadequately address the psychiatric comorbidities of chronic and persistent pain, but there is no standard preclinical model of pain-altered behavior to support the development of new therapies. To explore this conflicting and inconclusive literature, we conducted a focused systematic review and meta-analysis on the effect of complete Freund adjuvant-induced (CFA) rodent hind paw inflammation on multiple classical indicators of exploratory behavior, stress coping, and naturalistic behavior. Our primary objective was to define CFA's effect on assays including, but not limited to, the elevated plus maze and forced swim test. Our secondary objective was to discover how variables such as species and strain may influence outcomes in such assays. We searched Ovid MEDLINE, Embase, Scopus, and Web of Science in April and October 2020 for studies with adult rodents injected with CFA into the hind paw and subsequently tested for aspects of "anxiety-like" or "depressive-like" behaviors. Forty-four studies evaluated performance in the elevated plus or zero maze, open field test, light-dark box, place escape and avoidance paradigm, forced swim test, tail suspension test, sucrose preference test, wheel running, and burrowing assay. Complete Freund adjuvant modestly but significantly decreased exploratory behavior, significantly increased passive stress coping in the tail suspension test but not the forced swim test, and significantly decreased preference for sucrose and naturally rewarding activity. Subgroup analyses revealed significant differences between species and animal sourcing. Based on the evidence provided here, we conclude future studies should focus on CFA's effect on natural rewards and naturalistic behaviors.


Asunto(s)
Actividad Motora , Roedores , Animales , Conducta Animal , Modelos Animales de Enfermedad , Adyuvante de Freund/toxicidad , Dolor/inducido químicamente , Dolor/psicología , Sacarosa/farmacología
14.
Pain ; 162(6): 1705-1721, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33433146

RESUMEN

ABSTRACT: Pain puts patients at risk for developing psychiatric conditions such as anxiety and depression. Preclinical mouse models of pain-induced affective behavior vary widely in methodology and results, impairing progress towards improved therapeutics. To systematically investigate the effect of long-term inflammatory pain on exploratory behavior and stress coping strategy, we assessed male C57BL/6J mice in the forced swim test (FST), elevated zero maze, and open field test at 4 and 6 weeks postinjection of Complete Freund's Adjuvant, while controlling for testing order and combination. Inflammatory pain did not induce a passive stress coping strategy in the FST and did not reduce exploratory behavior in the elevated zero maze or the open field test. Using systematic correlational analysis and composite behavioral scores, we found no consistent association among measures for mice with or without inflammatory pain. A meta-analysis of similar studies indicated a modest, significant effect of Complete Freund's Adjuvant on exploratory behavior, but not immobility in the FST, and high heterogeneity among effect sizes in all 3 paradigms. Given the urgency for understanding the mechanisms of pain comorbidities and identifying novel therapies, these findings support the reallocation of our limited resources away from such unreliable assays and toward motivated and naturalistic behaviors. Future studies in pain and psychiatric translational research may benefit by considering outcomes beyond binary categorization, quantifying the associations between multiple measured behaviors, and agnostically identifying subtle yet meaningful patterns in behaviors.


Asunto(s)
Conducta Exploratoria , Dolor , Adaptación Psicológica , Animales , Ansiedad , Conducta Animal , Depresión/etiología , Modelos Animales de Enfermedad , Humanos , Masculino , Aprendizaje por Laberinto , Ratones , Ratones Endogámicos C57BL , Dolor/etiología
15.
Int Rev Neurobiol ; 157: 31-68, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33648672

RESUMEN

Across centuries and civilizations opioids have been used to relieve pain. In our modern societies, opioid-based analgesics remain one of the most efficient treatments for acute pain. However, the long-term use of opioids can lead to the development of analgesic tolerance, opioid-induced hyperalgesia, opioid use disorders, and overdose, which can ultimately produce respiratory depressant effects with fatal consequences. In addition to the nociceptive sensory component of pain, negative affective states arising from persistent pain represent a risk factor for developing an opioid use disorder. Several studies have indicated that the increase in prescribed opioid analgesics since the 1990s represents the root of our current opioid epidemic. In this review, we will present our current knowledge on the endogenous opioid system within the pain neuroaxis and the plastic changes occurring in this system that may underlie the occurrence of pain-induced negative affect leading to misuse and abuse of opioid medications. Dissecting the allostatic neuronal changes occurring during pain is the most promising avenue to uncover novel targets for the development of safer pain medications. We will discuss this along with current and potential approaches to treat pain-induced negative affective states that lead to drug misuse. Moreover, this chapter will provide a discussion on potential avenues to reduce the abuse potential of new analgesic drugs and highlight a basis for future research and drug development based on recent advances in this field.


Asunto(s)
Dolor Agudo , Analgésicos Opioides , Manejo del Dolor , Dolor Agudo/tratamiento farmacológico , Afecto , Analgésicos Opioides/efectos adversos , Humanos , Trastornos Relacionados con Opioides/prevención & control , Manejo del Dolor/efectos adversos , Trastornos Relacionados con Sustancias/prevención & control
16.
Pain Rep ; 6(1): e897, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33693301

RESUMEN

INTRODUCTION: The periaqueductal gray (PAG) mediates the antinociceptive properties of analgesics, including opioids and cannabinoids. Administration of either opioids or cannabinoids into the PAG induces antinociception. However, most studies characterizing the antinociceptive properties of cannabinoids in the PAG have been conducted in naive animals. Few studies have reported on the role of CB1 receptors in the PAG during conditions which would prompt the administration of analgesics, namely, during pain states. OBJECTIVES: To examine inflammatory pain-induced changes in CB1 receptor expression and function in the midbrain periaqueductal gray. METHODS: In this study, we used the Complete Freund Adjuvant model to characterize CB1 receptor expression and G-protein coupling during persistent inflammatory pain. RESULTS: Inflammatory pain induced an upregulation in the expression of synaptic CB1 receptors in the PAG. Despite this pain-induced change in CB1 expression, there was no corresponding upregulation of CB1 mRNA after the induction of inflammatory pain, suggesting a pain-induced recruitment of CB1 receptors to the synaptic sites within PAG neurons or increased coupling efficiency between the receptor and effector systems. Inflammatory pain also enhanced ventrolateral PAG CB1 receptor activity, as there was an increase in CP55,940-stimulated G-protein activation compared with pain-naïve control animals. CONCLUSION: These findings complement a growing body of evidence which demonstrate pain-induced changes in brain regions that are responsible for both the analgesic and rewarding properties of analgesic pharmacotherapies. Because much of our understanding of the pharmacology of cannabinoids is based on studies which use largely pain-naïve male animals, this work fills in important gaps in the knowledge base by incorporating pain-induced adaptations and cannabinoid pharmacology in females.

17.
Nat Neurosci ; 24(11): 1601-1613, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34663957

RESUMEN

The persistence of negative affect in pain leads to co-morbid symptoms such as anhedonia and depression-major health issues in the United States. The neuronal circuitry and contribution of specific cellular populations underlying these behavioral adaptations remains unknown. A common characteristic of negative affect is a decrease in motivation to initiate and complete goal-directed behavior, known as anhedonia. We report that in rodents, inflammatory pain decreased the activity of ventral tegmental area (VTA) dopamine (DA) neurons, which are critical mediators of motivational states. Pain increased rostromedial tegmental nucleus inhibitory tone onto VTA DA neurons, making them less excitable. Furthermore, the decreased activity of DA neurons was associated with reduced motivation for natural rewards, consistent with anhedonia-like behavior. Selective activation of VTA DA neurons was sufficient to restore baseline motivation and hedonic responses to natural rewards. These findings reveal pain-induced adaptations within VTA DA neurons that underlie anhedonia-like behavior.


Asunto(s)
Adaptación Fisiológica/fisiología , Anhedonia/fisiología , Neuronas Dopaminérgicas/metabolismo , Dolor/metabolismo , Área Tegmental Ventral/metabolismo , Animales , Condicionamiento Operante/fisiología , Neuronas Dopaminérgicas/química , Femenino , Masculino , Optogenética/métodos , Dolor/genética , Ratas , Ratas Long-Evans , Ratas Transgénicas , Área Tegmental Ventral/química
18.
Biol Psychiatry ; 90(11): 766-780, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34548146

RESUMEN

BACKGROUND: CNIH3 is an AMPA receptor (AMPAR) auxiliary protein prominently expressed in the dorsal hippocampus (dHPC), a region that plays a critical role in spatial memory and synaptic plasticity. However, the effects of CNIH3 on AMPAR-dependent synaptic function and behavior have not been investigated. METHODS: We assessed a gain-of-function model of Cnih3 overexpression in the dHPC and generated and characterized a line of Cnih3-/- C57BL/6 mice. We assessed spatial memory through behavioral assays, protein levels of AMPAR subunits and synaptic proteins by immunoblotting, and long-term potentiation in electrophysiological recordings. We also utilized a super-resolution imaging workflow, SEQUIN (Synaptic Evaluation and Quantification by Imaging of Nanostructure), for analysis of nanoscale synaptic connectivity in the dHPC. RESULTS: Overexpression of Cnih3 in the dHPC improved short-term spatial memory in female mice but not in male mice. Cnih3-/- female mice exhibited weakened short-term spatial memory, reduced dHPC synapse density, enhanced expression of calcium-impermeable AMPAR (GluA2-containing) subunits in synaptosomes, and attenuated long-term potentiation maintenance compared with Cnih3+/+ control mice; Cnih3-/- males were unaffected. Further investigation revealed that deficiencies in spatial memory and changes in AMPAR composition and synaptic plasticity were most pronounced during the metestrus phase of the estrous cycle in female Cnih3-/- mice. CONCLUSIONS: This study identified a novel effect of sex and estrous on CNIH3's role in spatial memory and synaptic plasticity. Manipulation of CNIH3 unmasked sexually dimorphic effects on spatial memory, synaptic function, AMPAR composition, and hippocampal plasticity. These findings reinforce the importance of considering sex as a biological variable in studies of memory and hippocampal synaptic function.


Asunto(s)
Caracteres Sexuales , Memoria Espacial , Animales , Femenino , Hipocampo/metabolismo , Potenciación a Largo Plazo , Masculino , Ratones , Ratones Endogámicos C57BL , Plasticidad Neuronal , Receptores AMPA/genética , Receptores AMPA/metabolismo , Sinapsis/metabolismo , Transmisión Sináptica
19.
Mol Pharmacol ; 77(5): 874-83, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20159947

RESUMEN

Evidence suggests that the long-term adaptations in the hippocampus after repeated drug treatment may parallel its role during memory formation. The neuroplasticity that subserves learning and memory is also believed to underlie addictive processes. We have reported previously that repeated morphine administration alters local distribution of endocytic proteins at hippocampal synapses, which could in turn affect expression of glutamate receptors. Glutamatergic systems, including alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptors (AMPARs), are believed to be involved in opiate-induced neuronal and behavioral plasticity, although the mechanisms underlying these effects are only beginning to be understood. The present study further examines the effects of repeated morphine administration on the expression and composition of AMPARs and the functional ramifications. Twelve hours after the last morphine injection, we observed an increased expression of AMPARs lacking glutamate receptor (GluR) 2 in hippocampal synaptic fractions. Immunoblotting studies show that 12 h after morphine treatment, GluR1 subunits are increased at the postsynaptic density (PSD) and at extrasynaptic sites, whereas GluR3 subunits are only increased at the PSD, and they show how this alters receptor subunit composition. In addition, we provide electrophysiological evidence that AMPARs are switched to Ca(2+)-permeable (GluR2-lacking) at the synapse 12 h after repeated morphine treatment, affecting the magnitude of long-term depression at hippocampal neurons. We propose that morphine-induced changes in glutamatergic synaptic transmission in the hippocampus may play an important role in the neuroadaptations induced by repeated morphine administration.


Asunto(s)
Morfina/farmacología , Receptores AMPA/biosíntesis , Animales , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Antagonistas del GABA/farmacología , Potenciación a Largo Plazo/efectos de los fármacos , Potenciación a Largo Plazo/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ácidos Fosfínicos/farmacología , Fosforilación , Picrotoxina/farmacología , Propanolaminas/farmacología , Subunidades de Proteína/efectos de los fármacos , Subunidades de Proteína/genética , Receptores AMPA/deficiencia , Receptores AMPA/efectos de los fármacos , Receptores AMPA/genética , Receptores AMPA/fisiología , Sinapsis/efectos de los fármacos , Sinapsis/fisiología , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/fisiología
20.
Eur J Neurosci ; 32(4): 625-31, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20626460

RESUMEN

The addictive properties of morphine limit its clinical use. Learned associations that develop between the abused opiate and the environment in which it is consumed are engendered through Pavlovian conditioning processes. Disruption of the learned associations between the opiate and environmental cues may be a therapeutic approach to prevent morphine dependence. Although a role for the delta-opioid receptor in the regulation of the rewarding properties of morphine has already been shown, in this study we further characterized the role of the delta-opioid receptor in morphine-induced conditioned responses by examining the effect of a selective delta2-opioid receptor antagonist (naltriben), using a conditioned place preference paradigm in rats. Additionally, we used a subcellular fractionation technique to analyze the synaptic localization of mu-opioid and delta-opioid receptors in the hippocampus, in order to examine the molecular mechanisms that may underlie this morphine-induced conditioned behavior. Our data show that the administration of 1 mg/kg naltriben (but not 0.1 mg/kg) prior to morphine was able to block morphine-induced conditioned place preference. Interestingly, this naltriben-induced disruption of morphine conditioned place preference was associated with a significant increase in the expression of the delta-opioid receptor dimer at the postsynaptic density. In addition, we also observed that morphine conditioned place preference was associated with an increase in the expression of the mu-opoid receptor in the total homogenate. Overall, these results suggest that modulation of the delta-opioid receptor expression and its synaptic localization may constitute a viable therapeutic approach to disrupt morphine-induced conditioned responses.


Asunto(s)
Conducta Animal/efectos de los fármacos , Condicionamiento Clásico/efectos de los fármacos , Morfina/farmacología , Antagonistas de Narcóticos/farmacología , Receptores Opioides delta , Receptores Opioides mu/metabolismo , Sinapsis/metabolismo , Analgésicos Opioides/química , Analgésicos Opioides/farmacología , Animales , Masculino , Morfina/química , Naltrexona/análogos & derivados , Naltrexona/farmacología , Ratas , Ratas Sprague-Dawley , Receptores Opioides delta/antagonistas & inhibidores , Receptores Opioides delta/metabolismo , Fracciones Subcelulares/química , Sinapsis/química , Sinapsis/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA