Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Neuropsychopharmacology ; 49(6): 915-923, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38374364

RESUMO

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.


Assuntos
Sinais (Psicologia) , Comportamento de Procura de Droga , Hipocampo , Locus Cerúleo , Autoadministração , Animais , Locus Cerúleo/efeitos dos fármacos , Locus Cerúleo/metabolismo , Masculino , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Ratos , Feminino , Comportamento de Procura de Droga/efeitos dos fármacos , Comportamento de Procura de Droga/fisiologia , Morfina/farmacologia , Morfina/administração & dosagem , Ratos Sprague-Dawley , Vias Neurais/efeitos dos fármacos , Vias Neurais/fisiologia , Analgésicos Opioides/farmacologia , Analgésicos Opioides/administração & dosagem , Transtornos Relacionados ao Uso de Opioides/fisiopatologia , Extinção Psicológica/efeitos dos fármacos , Extinção Psicológica/fisiologia , Condicionamento Operante/efeitos dos fármacos , Condicionamento Operante/fisiologia
2.
Nat Commun ; 15(1): 750, 2024 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-38286800

RESUMO

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.


Assuntos
Dinorfinas , Núcleo Accumbens , Éteres Fosfolipídicos , Núcleo Accumbens/fisiologia , Hipocampo/fisiologia , Recompensa , Neurônios/fisiologia
3.
Pain Rep ; 8(4): e1082, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37388406

RESUMO

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.

4.
eNeuro ; 10(3)2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36849260

RESUMO

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.


Assuntos
Ciclo Estral , Hipocampo , Animais , Feminino , Masculino , Camundongos , Ciclo Estral/genética , Hipocampo/metabolismo , Aprendizagem , Plasticidade Neuronal , Transcriptoma
5.
Br J Pharmacol ; 180(7): 793-796, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36813266

RESUMO

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.


Assuntos
Analgésicos Opioides , Transtornos Relacionados ao Uso de Opioides , Humanos , Analgésicos Opioides/uso terapêutico , Transtornos Relacionados ao Uso de Opioides/tratamento farmacológico , Epidemia de Opioides
6.
Front Syst Neurosci ; 16: 1014768, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36341476

RESUMO

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.

7.
Pain ; 163(5): 809-819, 2022 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-34510137

RESUMO

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.


Assuntos
Atividade Motora , Roedores , Animais , Comportamento Animal , Modelos Animais de Doenças , Adjuvante de Freund/toxicidade , Dor/induzido quimicamente , Dor/psicologia , Sacarose/farmacologia
8.
Nat Neurosci ; 24(11): 1601-1613, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34663957

RESUMO

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.


Assuntos
Adaptação Fisiológica/fisiologia , Anedonia/fisiologia , Neurônios Dopaminérgicos/metabolismo , Dor/metabolismo , Área Tegmentar Ventral/metabolismo , Animais , Condicionamento Operante/fisiologia , Neurônios Dopaminérgicos/química , Feminino , Masculino , Optogenética/métodos , Dor/genética , Ratos , Ratos Long-Evans , Ratos Transgênicos , Área Tegmentar Ventral/química
9.
Nature ; 598(7882): 646-651, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34646022

RESUMO

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.


Assuntos
Analgésicos Opioides/farmacologia , Núcleo Accumbens/fisiologia , Receptores Opioides mu/fisiologia , Recompensa , Animais , Encefalinas , Feminino , Masculino , Camundongos , Camundongos Knockout
10.
Biol Psychiatry ; 90(11): 766-780, 2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34548146

RESUMO

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.


Assuntos
Caracteres Sexuais , Memória Espacial , Animais , Feminino , Hipocampo/metabolismo , Potenciação de Longa Duração , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Plasticidade Neuronal , Receptores de AMPA/genética , Receptores de AMPA/metabolismo , Sinapses/metabolismo , Transmissão Sináptica
11.
Int Rev Neurobiol ; 157: 31-68, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33648672

RESUMO

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.


Assuntos
Dor Aguda , Analgésicos Opioides , Manejo da Dor , Dor Aguda/tratamento farmacológico , Afeto , Analgésicos Opioides/efeitos adversos , Humanos , Transtornos Relacionados ao Uso de Opioides/prevenção & controle , Manejo da Dor/efeitos adversos , Transtornos Relacionados ao Uso de Substâncias/prevenção & controle
12.
Pain Rep ; 6(1): e897, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33693301

RESUMO

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.

13.
Pain ; 162(6): 1705-1721, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-33433146

RESUMO

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.


Assuntos
Comportamento Exploratório , Dor , Adaptação Psicológica , Animais , Ansiedade , Comportamento Animal , Depressão/etiologia , Modelos Animais de Doenças , Humanos , Masculino , Aprendizagem em Labirinto , Camundongos , Camundongos Endogâmicos C57BL , Dor/etiologia
14.
J Med Chem ; 63(22): 13618-13637, 2020 11 25.
Artigo em Inglês | MEDLINE | ID: mdl-33170687

RESUMO

In this work, we studied a series of carfentanyl amide-based opioid derivatives targeting the mu opioid receptor (µOR) and the delta opioid receptor (δOR) heteromer as a credible novel target in pain management therapy. We identified a lead compound named MP135 that exhibits high G-protein activity at µ-δ heteromers compared to the homomeric δOR or µOR and low ß-arrestin2 recruitment activity at all three. Furthermore, MP135 exhibits distinct signaling profile, as compared to the previously identified agonist targeting µ-δ heteromers, CYM51010. Pharmacological characterization of MP135 supports the utility of this compound as a molecule that could be developed as an antinociceptive agent similar to morphine in rodents. In vivo characterization reveals that MP135 maintains untoward side effects such as respiratory depression and reward behavior; together, these results suggest that optimization of MP135 is necessary for the development of therapeutics that suppress the classical side effects associated with conventional clinical opioids.


Assuntos
Fentanila/análogos & derivados , Receptores Opioides delta/agonistas , Analgésicos/síntese química , Analgésicos/farmacologia , Animais , Linhagem Celular , Fentanila/síntese química , Fentanila/farmacologia , Humanos , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Medição da Dor/efeitos dos fármacos , Medição da Dor/métodos , Ratos , Ratos Long-Evans , Receptores Opioides delta/metabolismo
15.
Artigo em Inglês | MEDLINE | ID: mdl-31978422

RESUMO

The neurobiological mechanisms underlying alcohol motivational properties are still not fully understood, however, the mu-opioid receptors (MORs) have been evidenced as central elements in the manifestation of the alcohol reinforcing properties. Drug-associated environmental stimuli can trigger alcohol relapse and promote alcohol consumption whereby N-methyl-d-aspartate (NMDA) receptors play a pivotal role. Here we sought to demonstrate, for the first time, that ethanol induces conditioned place preference or aversion (CPP or CPA) when administered locally into the ventral tegmental area (VTA) and the associated role of MORs. We further analyzed the changes in the expression and mRNA levels of GluN1 and GluN2A subunits in designated brain areas. The expression of CPP or CPA was characterized following intra-VTA ethanol administration and we showed that either reinforcing (CPP) or aversive (CPA) properties are dependent on the dose administered (ranging here from 35 to 300 nmol). Furthermore, the critical contribution of local MORs in the acquisition of CPP was revealed by a selective antagonist, namely ß-Funaltrexamine. Finally, modifications of the expression of NMDA receptor subunits in the Nucleus Accumbens (NAc) and Hippocampus after ethanol-induced CPP were analyzed at the proteomic and transcriptomic levels by western blot and In Situ Hybridation RNAscope techniques, respectively. Results showed that the mRNA levels of GluN2A but not GluN1 in NAc are higher after ethanol CPP. These novel results pave the way for further characterisation of the mechanisms by which ethanol motivational properties are associated with learned environmental cues.


Assuntos
Aprendizagem da Esquiva/fisiologia , Condicionamento Psicológico/fisiologia , Etanol/administração & dosagem , Receptores de N-Metil-D-Aspartato/metabolismo , Receptores Opioides mu/fisiologia , Área Tegmentar Ventral/metabolismo , Animais , Aprendizagem da Esquiva/efeitos dos fármacos , Condicionamento Psicológico/efeitos dos fármacos , Relação Dose-Resposta a Droga , Infusões Intraventriculares , Masculino , Microinjeções/métodos , Ratos , Ratos Wistar , Área Tegmentar Ventral/efeitos dos fármacos
16.
Psychol Sci Public Interest ; 20(2): 96-127, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31591935

RESUMO

Two major questions about addictive behaviors need to be explained by any worthwhile neurobiological theory. First, why do people seek drugs in the first place? Second, why do some people who use drugs seem to eventually become unable to resist drug temptation and so become "addicted"? We will review the theories of addiction that address negative-reinforcement views of drug use (i.e., taking opioids to alleviate distress or withdrawal), positive-reinforcement views (i.e., taking drugs for euphoria), habit views (i.e., growth of automatic drug-use routines), incentive-sensitization views (i.e., growth of excessive "wanting" to take drugs as a result of dopamine-related sensitization), and cognitive-dysfunction views (i.e., impaired prefrontal top-down control), including those involving competing neurobehavioral decision systems (CNDS), and the role of the insula in modulating addictive drug craving. In the special case of opioids, particular attention is paid to whether their analgesic effects overlap with their reinforcing effects and whether the perceived low risk of taking legal medicinal opioids, which are often prescribed by a health professional, could play a role in the decision to use. Specifically, we will address the issue of predisposition or vulnerability to becoming addicted to drugs (i.e., the question of why some people who experiment with drugs develop an addiction, while others do not). Finally, we review attempts to develop novel therapeutic strategies and policy ideas that could help prevent opioid and other substance abuse.


Assuntos
Comportamento Aditivo/psicologia , Epidemia de Opioides , Transtornos Relacionados ao Uso de Opioides/psicologia , Fissura , Tomada de Decisões , Humanos
17.
Curr Opin Behav Sci ; 26: 69-74, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30984806

RESUMO

Pain has a useful protective role; through avoidance learning, it helps to decrease the probability of engaging in tissue-damaging, or otherwise dangerous experiences. In our modern society, the experience of acute post-surgical pain and the development of chronic pain states represent an unnecessary negative outcome. This has become an important health issue as more than 30% of the US population reports experiencing "unnecessary" pain at any given time. Opioid therapies are often efficacious treatments for severe and acute pain; however, in addition to their powerful analgesic properties, opioids produce potent reinforcing properties and their inappropriate use has led to the current opioid overdose epidemic in North America. Dissecting the allostatic changes occurring in nociceptors and neuronal pathways in response to pain are the first and most important steps in understanding the physiologic changes underlying the opioid epidemic. Full characterization of these adaptations will provide novel targets for the development of safer pharmacotherapies. In this review, we highlight the current efforts toward safer opioid treatments and describe our current knowledge of the interaction between pain and opioid systems.

18.
Neuron ; 102(3): 564-573.e6, 2019 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-30878290

RESUMO

Negative affective states affect quality of life for patients suffering from pain. These maladaptive emotional states can lead to involuntary opioid overdose and many neuropsychiatric comorbidities. Uncovering the mechanisms responsible for pain-induced negative affect is critical in addressing these comorbid outcomes. The nucleus accumbens (NAc) shell, which integrates the aversive and rewarding valence of stimuli, exhibits plastic adaptations in the presence of pain. In discrete regions of the NAc, activation of the kappa opioid receptor (KOR) decreases the reinforcing properties of rewards and induces aversive behaviors. Using complementary techniques, we report that in vivo recruitment of NAc shell dynorphin neurons, acting through KOR, is necessary and sufficient to drive pain-induced negative affect. Taken together, our results provide evidence that pain-induced adaptations in the kappa opioid system within the NAc shell represent a functional target for therapeutic intervention that could circumvent pain-induced affective disorders. VIDEO ABSTRACT.


Assuntos
Afeto/fisiologia , Dinorfinas/metabolismo , Inflamação/metabolismo , Transtornos do Humor/metabolismo , Neurônios/metabolismo , Núcleo Accumbens/metabolismo , Dor/metabolismo , Receptores Opioides kappa/metabolismo , Animais , Inflamação/complicações , Inflamação/psicologia , Camundongos , Transtornos do Humor/etiologia , Transtornos do Humor/psicologia , Inibição Neural , Plasticidade Neuronal , Núcleo Accumbens/citologia , Dor/complicações , Dor/psicologia , Ratos
19.
Exp Neurol ; 314: 58-66, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30660616

RESUMO

Acute inflammation induces sensitization of nociceptive neurons and triggers the accumulation of calcium permeable (CP) α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptors (AMPARs) in the dorsal horn of the spinal cord. This coincides with behavioral signs of acute inflammatory pain, but whether CP-AMPARs contribute to chronic pain remains unclear. To evaluate this question, we first constructed current-voltage (IV) curves of C-fiber stimulus-evoked, AMPAR-mediated EPSCs in lamina II to test for inward rectification, a key characteristic of CP-AMPARs. We found that the intraplantar injection of complete Freund's adjuvant (CFA) induced an inward rectification at 3 d that persisted to 21 d after injury. Furthermore, the CP- AMPAR antagonist IEM-1460 (50 µM) inhibited AMPAR-evoked Ca2+ transients 21d after injury but had no effect in uninflamed mice. We then used a model of long-lasting vulnerability for chronic pain that is determined by the balance between latent central sensitization (LCS) and mu opioid receptor constitutive activity (MORCA). When administered 21 d after the intraplantar injection of CFA, intrathecal administration of the MORCA inverse agonist naltrexone (NTX, 1 µg, i.t.) reinstated mechanical hypersensitivity, and superfusion of spinal cord slices with NTX (10 µM) increased the peak amplitude of AMPAR-evoked Ca2+ transients in lamina II neurons. The CP-AMPAR antagonist naspm (0-10 nmol, i.t.) inhibited these NTX-induced increases in mechanical hypersensitivity. NTX had no effect in uninflamed mice. Subsequent western blot analysis of the postsynaptic density membrane fraction from lumbar dorsal horn revealed that CFA increased GluA1 expression at 2 d and GluA4 expression at both 2 and 21 d post-injury, indicating that not just the GluA1 subunit, but also the GluA4 subunit, contributes to the expression of CP-AMPARs and synaptic strength during hyperalgesia. GluA2 expression increased at 21 d, an unexpected result that requires further study. We conclude that after tissue injury, dorsal horn AMPARs retain a Ca2+ permeability that underlies LCS. Because of their effectiveness in reducing naltrexone-induced reinstatement of hyperalgesia and potentiation of AMPAR-evoked Ca2+ signals, CP-AMPAR inhibitors are a promising class of agents for the treatment of chronic inflammatory pain.


Assuntos
Cálcio/metabolismo , Dor Crônica/fisiopatologia , Receptores de AMPA/metabolismo , Receptores Opioides/metabolismo , Medula Espinal/metabolismo , Medula Espinal/fisiopatologia , Adamantano/análogos & derivados , Adamantano/farmacologia , Animais , Dor Crônica/induzido quimicamente , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Adjuvante de Freund , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Naltrexona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Fibras Nervosas Amielínicas , Nociceptividade , Células do Corno Posterior/efeitos dos fármacos , Receptores de AMPA/antagonistas & inibidores , Receptores de Glutamato/metabolismo , Sinapses/efeitos dos fármacos
20.
J Neurosci ; 38(11): 2780-2795, 2018 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-29459374

RESUMO

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.


Assuntos
Deficiência Intelectual/genética , Deficiência Intelectual/fisiopatologia , Deficiências da Aprendizagem/genética , Deficiências da Aprendizagem/fisiopatologia , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Transtornos da Memória/genética , Transtornos da Memória/fisiopatologia , Proteínas do Tecido Nervoso/genética , Proteínas Adaptadoras de Transdução de Sinal , Animais , Região CA1 Hipocampal/fisiopatologia , Potenciais Pós-Sinápticos Excitadores/genética , Hipocampo/metabolismo , Hipocampo/fisiopatologia , Deficiência Intelectual/tratamento farmacológico , Deficiências da Aprendizagem/tratamento farmacológico , Potenciação de Longa Duração/genética , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina/biossíntese , Transtornos da Memória/tratamento farmacológico , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas do Tecido Nervoso/biossíntese , Inibidores de Proteínas Quinases/uso terapêutico , Comportamento Social
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...