Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
eNeuro ; 11(2)2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-38164564

RESUMO

Opioid use disorder (OUD) is a public health crisis currently being exacerbated by increased rates of use and overdose of synthetic opioids, primarily fentanyl. Therefore, the identification of novel biomarkers and treatment strategies to reduce problematic fentanyl use and relapse to fentanyl taking is critical. In recent years, there has been a growing body of work demonstrating that the gut microbiome can serve as a potent modulator of the behavioral and transcriptional responses to both stimulants and opioids. Here, we advance this work to define how manipulations of the microbiome drive fentanyl intake and fentanyl-seeking in a translationally relevant drug self-administration model. Depletion of the microbiome of male rats with broad spectrum antibiotics leads to increased drug administration on increased fixed ratio, progressive ratio, and drug seeking after abstinence. Utilizing 16S  sequencing of microbiome contents from these animals, specific populations of bacteria from the gut microbiome correlate closely with levels of drug taking. Additionally, global proteomic analysis of the nucleus accumbens following microbiome manipulation and fentanyl administration to define how microbiome status alters the functional proteomic landscape in this key limbic substructure. These data demonstrate that an altered microbiome leads to marked changes in the synaptic proteome in response to repeated fentanyl treatment. Finally, behavioral effects of microbiome depletion are reversible by upplementation of the microbiome derived short-chain fatty acid metabolites. Taken together, these findings establish clear relevance for gut-brain signaling in models of OUD and lay foundations for further translational work in this space.


Assuntos
Microbioma Gastrointestinal , Transtornos Relacionados ao Uso de Opioides , Masculino , Ratos , Animais , Fentanila , Proteoma , Proteômica , Analgésicos Opioides , Transtornos Relacionados ao Uso de Opioides/tratamento farmacológico
2.
Biol Psychiatry ; 95(4): 329-338, 2024 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-37573004

RESUMO

Substance use disorders are a set of recalcitrant neuropsychiatric conditions that cause tremendous morbidity and mortality and are among the leading causes of loss of disability-adjusted life years worldwide. While each specific substance use disorder is driven by problematic use of a different substance, they all share a similar pattern of escalating and out-of-control substance use, continued use despite negative consequences, and a remitting/relapsing pattern over time. Despite significant advances in our understanding of the neurobiology of these conditions, current treatment options remain few and are ineffective for too many individuals. In recent years, there has been a rapidly growing body of literature demonstrating that the resident population of microbes in the gastrointestinal tract, collectively called the gut microbiome, plays an important role in modulating brain and behavior in preclinical and clinical studies of psychiatric disease. While these findings have not yet been translated into clinical practice, this remains an important and exciting avenue for translational research. In this review, we highlight the current state of microbiome-brain research within the substance use field with a focus on both clinical and preclinical studies. We also discuss potential neurobiological mechanisms underlying microbiome effects on models of substance use disorder and propose future directions to bring these findings from bench to bedside.


Assuntos
Microbioma Gastrointestinal , Transtornos Relacionados ao Uso de Substâncias , Humanos , Encéfalo , Trato Gastrointestinal
3.
Neuropsychopharmacology ; 49(2): 386-395, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37528220

RESUMO

Cocaine use disorder represents a public health crisis with no FDA-approved medications for its treatment. A growing body of research has detailed the important connections between the brain and the resident population of bacteria in the gut, the gut microbiome, in psychiatric disease models. Acute depletion of gut bacteria results in enhanced reward in a mouse cocaine place preference model, and repletion of bacterially-derived short-chain fatty acid (SCFA) metabolites reverses this effect. However, the role of the gut microbiome and its metabolites in modulating cocaine-seeking behavior after prolonged abstinence is unknown. Given that relapse prevention is the most clinically challenging issue in treating substance use disorders, studies examining the effects of microbiome manipulations in relapse-relevant models are critical. Here, male Sprague-Dawley rats received either untreated water or antibiotics to deplete the gut microbiome and its metabolites. Rats were trained to self-administer cocaine and subjected to either within-session threshold testing to evaluate motivation for cocaine or 21 days of abstinence followed by a cue-induced cocaine-seeking task to model relapse behavior. Microbiome depletion did not affect cocaine acquisition on an fixed-ratio 1 schedule. However, microbiome-depleted rats exhibited significantly enhanced motivation for low dose cocaine on a within-session threshold task. Similarly, microbiome depletion increased cue-induced cocaine-seeking following prolonged abstinence and altered transcriptional regulation in the nucleus accumbens. In the absence of a normal microbiome, repletion of bacterially-derived SCFA metabolites reversed the behavioral and transcriptional changes associated with microbiome depletion. These findings suggest that gut bacteria, via their metabolites, are key regulators of drug-seeking behaviors, positioning the microbiome as a potential translational research target.


Assuntos
Transtornos Relacionados ao Uso de Cocaína , Cocaína , Camundongos , Ratos , Masculino , Animais , Ratos Sprague-Dawley , Comportamento de Procura de Droga , Transtornos Relacionados ao Uso de Cocaína/metabolismo , Núcleo Accumbens , Recidiva , Autoadministração , Sinais (Psicologia) , Extinção Psicológica
4.
Sci Rep ; 13(1): 12223, 2023 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-37500710

RESUMO

Opioid Use Disorder (OUD) is associated with tremendous morbidity and mortality. Despite this burden, current pharmacotherapies for OUD are ineffective or intolerable for many patients. As such, interventions aimed at promoting resilience against OUD are of immense clinical interest. Treatment with a Bioactive Dietary Polyphenol Preparation (BDPP) promotes resilience and adaptive neuroplasticity in multiple models of neuropsychiatric disease. Here, we assessed effects of BDPP treatment on behavioral and molecular responses to repeated morphine treatment in male mice. BDPP pre-treatment alters responses for both locomotor sensitization and conditioned place preference. Most notably, polyphenol treatment consistently reduced formation of preference at low dose (5 mg/kg) morphine but enhanced it at high dose (15 mg/kg). In parallel, we performed transcriptomic profiling of the nucleus accumbens, which again showed a dose × polyphenol interaction. We also profiled microbiome composition and function, as polyphenols are metabolized by the microbiome and can act as prebiotics. The profile revealed polyphenol treatment markedly altered microbiome composition and function. Finally, we investigated involvement of the SIRT1 deacetylase, and the role of polyphenol metabolites in behavioral responses. These results demonstrate polyphenols have robust dose-dependent effects on behavioral and physiological responses to morphine and lay the foundation for future translational work.


Assuntos
Morfina , Núcleo Accumbens , Camundongos , Masculino , Animais , Núcleo Accumbens/metabolismo , Polifenóis/metabolismo
5.
Mol Cell Neurosci ; 126: 103874, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37315877

RESUMO

Opioid use disorder is a public health crisis that causes tremendous suffering for patients as well as substantial social and economic costs for society. There are currently available treatments for patients with opioid use disorder, but they remain intolerable or ineffective for many. Thus the need to develop new avenues for therapeutics development in this space is great. Substantial work in models of substance use disorders, including opioid use disorder, demonstrates that prolonged exposure to drugs of abuse leads to marked transcriptional and epigenetic dysregulation in limbic substructures. It is widely believed that these changes in gene regulation in response to drugs are a key driving factor in the perpetuation of drug taking and seeking behaviors. Thus, development of interventions that could shape transcriptional regulation in response to drugs of abuse would be of high value. Over the past decade there has been a surge in research demonstrating that the resident bacteria of the gastrointestinal tract, collectively the gut microbiome, can have tremendous influence on neurobiological and behavioral plasticity. Previous work from our group and others has demonstrated that alterations in the gut microbiome can alter behavioral responses to opioids in multiple paradigms. Additionally, we have previously reported that depletion of the gut microbiome with antibiotics markedly shifts the transcriptome of the nucleus accumbens following prolonged morphine exposure. In this manuscript we present a comprehensive analysis of the effects of the gut microbiome on transcriptional regulation of the nucleus accumbens following morphine by utilizing germ-free, antibiotic treated, and control mice. This allows for detailed understanding of the role of the microbiome in regulating baseline transcriptomic control, as well as response to morphine. We find that germ-free status leads to a marked gene dysregulation in a manner distinct to adult mice treated with antibiotics, and that altered gene pathways are highly related to cellular metabolic processes. These data provide additional insight into the role of the gut microbiome in modulating brain function and lay a foundation for further study in this area.


Assuntos
Morfina , Transtornos Relacionados ao Uso de Opioides , Camundongos , Animais , Morfina/efeitos adversos , Transcriptoma , Núcleo Accumbens , Antibacterianos
6.
Psychopharmacology (Berl) ; 239(4): 1009-1018, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33768375

RESUMO

RATIONALE: In rodents, environmental enrichment (EE) produces both preventive and curative effects on drug addiction, and this effect is believed to depend at least in part on EE's actions on the stress system. OBJECTIVES: This study investigated whether exposure to EE during abstinence reduces methamphetamine seeking after extended self-administration. In addition, we investigated whether these effects are associated with alterations in the levels of glucocorticoid receptors (GR) in the brain and whether administration of GR antagonists blocks methamphetamine relapse. METHODS: We allowed rats to self-administer methamphetamine for twenty 14-h sessions. After 3 weeks of abstinence either in standard (SE) or EE conditions, we measured methamphetamine seeking in a single 3-h session. Then, we used western blot techniques to measure GR levels in several brain areas. Finally, in an independent group of rats, after methamphetamine self-administration and abstinence in SE, we administered the GR antagonist mifepristone, and we investigated methamphetamine seeking. RESULTS: Exposure to EE reduced methamphetamine seeking and reversed methamphetamine-induced increases in GR levels in the ventral and dorsal hippocampus. In addition, EE decreased GR levels in the amygdala in drug-naive animals, but this effect was prevented by previous exposure to methamphetamine. Administration of mifepristone significantly decreased methamphetamine seeking. CONCLUSIONS: The anti-craving effects of EE are paralleled by restoration of methamphetamine-induced dysregulation of GR in the hippocampus. These results provide support for the hypothesis that the effect of EE on methamphetamine relapse is at least in part mediated by EE's action on the brain stress system.


Assuntos
Metanfetamina , Animais , Comportamento de Procura de Droga , Metanfetamina/farmacologia , Mifepristona/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores de Glucocorticoides , Recidiva , Autoadministração
7.
Neuropsychopharmacology ; 46(12): 2062-2072, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34127799

RESUMO

Recent evidence has demonstrated that the gut microbiome has marked effects on neuronal function and behavior. Disturbances to microbial populations within the gut have been linked to myriad models of neuropsychiatric disorders. However, the role of the microbiome in substance use disorders remains understudied. Here we show that male mice with their gut microbiome depleted by nonabsorbable antibiotics (Abx) exhibit decreased formation of morphine conditioned place preference across a range of doses (2.5-15 mg/kg), have decreased locomotor sensitization to morphine, and demonstrate marked changes in gene expression within the nucleus accumbens (NAc) in response to high-dose morphine (20 mg/kg × 7 days). Replacement of short-chain fatty acid (SCFA) metabolites, which are reduced by microbiome knockdown, reversed the behavioral and transcriptional effects of microbiome depletion. This identifies SCFA as the crucial mediators of microbiome-brain communication responsible for the effects on morphine reward caused by microbiome knockdown. These studies add important new behavioral, molecular, and mechanistic insight to the role of gut-brain signaling in substance use disorders.


Assuntos
Microbioma Gastrointestinal , Morfina , Animais , Condicionamento Clássico , Masculino , Camundongos , Morfina/farmacologia , Núcleo Accumbens , Recompensa
8.
Lab Anim (NY) ; 50(5): 125-126, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33782618
9.
J Neurosci ; 41(7): 1553-1565, 2021 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-33361463

RESUMO

Psychostimulant use disorder is a major public health issue, and despite the scope of the problem there are currently no Food and Drug Administration (FDA)-approved treatments. There would be tremendous utility in development of a treatment that could help patients both achieve and maintain abstinence. Previous work from our group has identified granulocyte-colony stimulating factor (G-CSF) as a neuroactive cytokine that alters behavioral response to cocaine, increases synaptic dopamine release, and enhances cognitive flexibility. Here, we investigate the role of G-CSF in affecting extinction and reinstatement of cocaine-seeking and perform detailed characterization of its proteomic effects in multiple limbic substructures. Male Sprague Dawley rats were injected with PBS or G-CSF during (1) extinction or (2) abstinence from cocaine self-administration, and drug seeking behavior was measured. Quantitative assessment of changes in the proteomic landscape in the nucleus accumbens (NAc) and medial prefrontal cortex (mPFC) were performed via data-independent acquisition (DIA) mass spectrometry analysis. Administration of G-CSF during extinction accelerated the rate of extinction, and administration during abstinence attenuated cue-induced cocaine-seeking. Analysis of global protein expression demonstrated that G-CSF regulated proteins primarily in mPFC that are critical to glutamate signaling and synapse maintenance. Taken together, these findings support G-CSF as a viable translational research target with the potential to reduce drug craving or seeking behaviors. Importantly, recombinant G-CSF exists as an FDA-approved medication which may facilitate rapid clinical translation. Additionally, using cutting-edge multiregion discovery proteomics analyses, these studies identify a novel mechanism underlying G-CSF effects on behavioral plasticity.SIGNIFICANCE STATEMENT Pharmacological treatments for psychostimulant use disorder are desperately needed, especially given the disease's chronic, relapsing nature. However, there are currently no Food and Drug Administration (FDA)-approved pharmacotherapies. Emerging evidence suggests that targeting the immune system may be a viable translational research strategy; preclinical studies have found that the neuroactive cytokine granulocyte-colony stimulating factor (G-CSF) alters cocaine reward and reinforcement and can enhance cognitive flexibility. Given this basis of evidence we studied the effects of G-CSF treatment on extinction and reinstatement of cocaine seeking. We find that administration of G-CSF accelerates extinction and reduces cue-induced drug seeking after cocaine self-administration. In addition, G-CSF leads to downregulation of synaptic glutamatergic proteins in medial prefrontal cortex (mPFC), suggesting that G-CSF influences drug seeking via glutamatergic mechanisms.


Assuntos
Transtornos Relacionados ao Uso de Cocaína/tratamento farmacológico , Comportamento de Procura de Droga/efeitos dos fármacos , Glutamatos/fisiologia , Fator Estimulador de Colônias de Granulócitos/farmacologia , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/metabolismo , Sinapses/efeitos dos fármacos , Sinapses/metabolismo , Animais , Transtornos Relacionados ao Uso de Cocaína/psicologia , Fissura/efeitos dos fármacos , Sinais (Psicologia) , Extinção Psicológica/efeitos dos fármacos , Sistema Límbico/efeitos dos fármacos , Masculino , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , Proteômica , Ratos , Ratos Sprague-Dawley , Síndrome de Abstinência a Substâncias/tratamento farmacológico , Síndrome de Abstinência a Substâncias/psicologia
10.
Neuropharmacology ; 184: 108441, 2021 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-33340530

RESUMO

Differences in overall cocaine intake can directly affect neuroadaptations, and this relationship can make it difficult to interpret neurobiological changes seen in drug-choice studies, since drug intake varies between subjects. Herein, a choice procedure that controls for cocaine intake was utilized to explore if neuronal activity, measured as cFos expression in the orbitofrontal cortex (OFC) and nucleus accumbens (NAc), was reflective of preference. Results demonstrated that cFos expression, in both the OFC and NAc, was independent of cocaine preference when cocaine intake was kept constant across individuals. However, when cocaine intake was systematically varied, the expression of cFos associated with cocaine preference was related to overall cocaine intake in the OFC, but not the NAc. Altogether, these results demonstrate that cocaine intake during choice can affect neurobiological outcome measures; thus, the neurobehavioral mechanisms underlying cocaine preference may be better isolated when controlling for cocaine frequency and intake. In all, some caution is warranted when interpreting results from choice studies evaluating the neurobehavioral mechanisms that underlie drug preference when drug frequency and intake are uncontrolled, and future research is needed to determine the role of drug frequency and intake on neurobiological measures associated with drug choice.


Assuntos
Encéfalo/efeitos dos fármacos , Comportamento de Escolha/efeitos dos fármacos , Cocaína/administração & dosagem , Neurônios/efeitos dos fármacos , Reforço Psicológico , Animais , Encéfalo/metabolismo , Comportamento de Escolha/fisiologia , Condicionamento Operante/efeitos dos fármacos , Condicionamento Operante/fisiologia , Masculino , Neurônios/metabolismo , Ratos , Ratos Sprague-Dawley , Autoadministração
11.
Drug Alcohol Depend ; 213: 108125, 2020 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-32590212

RESUMO

BACKGROUND: Social peers influence human drug use at every stage of addiction. Using a dual-compartment apparatus that allows for limited social contact, recent work has shown that cocaine self-administration is enhanced when two rats are trained to self-administer at the same time compared to rats trained alone or trained in the presence of a saline self-administration control peer. The current study measured social influence on self-administration of the short-acting opioid remifentanil using a dual-compartment operant conditioning chamber. METHODS: Adult male rats were placed in one of five groups: (1) REMI-REMI group, in which both rats self-administered remifentanil; (2) REMI-SAL group, in which rats self-administered remifentanil in the presence of a peer that self-administered saline; (3) SAL-REMI group, in which rats self-administered saline in the presence of a peer that self-administered remifentanil; and (4) REMI ALONE and (5) SAL ALONE groups, in which rats administered their respective drugs alone (no peer). Self-administration was measured using a 2-lever procedure during acquisition, maintenance, increasing fixed-ratio, and dose-response phases. RESULTS: The presence of a social peer enhanced drug intake during acquisition, regardless of the drug exposure of their peer. Additionally, active lever position significantly affected remifentanil intake during acquisition and maintenance, with the greatest influence occurring when the active lever was close to the peer. CONCLUSION: The presence of a social peer in the drug-taking context potentiates remifentanil self-administration, regardless of the peer's drug access. Future studies utilizing the dual-compartment apparatus will help elucidate the neural mechanisms underlying social influence on opioid abuse.

12.
Drug Alcohol Depend ; 207: 107804, 2020 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-31862556

RESUMO

BACKGROUND: Individuals with opioid use disorder (OUD) exhibit high levels of economic demand for opioids, with high levels of consumption and relative insensitivity to changes in price. Because the medications used to treat OUD in medication-assisted therapy (MAT) act as antagonists or agonists at µ opioid receptors, they may alter the relationship between price and opioid intake. METHODS: This study examined demand for a commonly abused synthetic prescription opioid, fentanyl, in male rats following s.c. pre-treatment with naltrexone (0.1-1.0 mg/kg), morphine (0.3-3.0 mg/kg) or buprenorphine (0.3-3.0 mg/kg). We normalized demand curves to intake at the lowest price and estimated effects on elasticity (sensitivity to changes in price). Rats were first trained to earn fentanyl (5 µg/kg/infusion) on a fixed ratio schedule, then they underwent daily training under a threshold procedure designed to produce within-session demand curve estimates. Rats received 14 threshold sessions before undergoing a series of tests encompassing each drug, at each dose. RESULTS: Elasticity was increased by pretreatment with naltrexone, morphine or buprenorphine. Morphine also decreased initial intake, when the price for fentanyl was lowest. In contrast, initial intake was increased by naltrexone (according to an inverted-U shaped curve). The effects of naltrexone did not persist after the test session, but morphine and buprenorphine continued affecting demand elasticity 24 h or 48 h after the test, respectively. CONCLUSIONS: These results indicate that fentanyl demand is sensitive to blockade or activation of opioid receptors by the drug classes used for MAT in humans.


Assuntos
Buprenorfina/farmacologia , Fentanila/farmacologia , Morfina/farmacologia , Naltrexona/farmacologia , Animais , Relação Dose-Resposta a Droga , Interações Medicamentosas , Masculino , Ratos , Receptores Opioides , Receptores Opioides mu/antagonistas & inibidores , Esquema de Reforço , Autoadministração
13.
ACS Chem Neurosci ; 10(10): 4213-4220, 2019 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-31479229

RESUMO

Addiction to psychostimulants is a major public health crisis that leads to significant morbidity and mortality, for which there are currently no FDA-approved pharmacotherapies. Female subjects have increased propensity to develop pathological substance use disorders after initial use, suggesting the possibility of different pathophysiological mechanisms between males and females. Recently, we identified the neuroactive cytokine granulocyte-colony stimulating factor (G-CSF) as a key mediator of neuronal and behavioral plasticity in response to cocaine in male mice. Here, we found that G-CSF potentiated the rewarding effects of cocaine in female mice as well; however, the dopaminergic mechanism linked to these effects was highly dependent on the ovarian hormone cycle. G-CSF treatment enhanced the ability of cocaine to inhibit dopamine clearance; however, this effect was observed specifically during pro/estrus, when circulating ovarian hormone levels were high. These findings demonstrate important sex differences in the synaptic effects of this translationally relevant neuroimmune modulator.


Assuntos
Cocaína/farmacologia , Condicionamento Operante/efeitos dos fármacos , Inibidores da Captação de Dopamina/farmacologia , Ciclo Estral/fisiologia , Fator Estimulador de Colônias de Granulócitos/farmacologia , Animais , Feminino , Fator Estimulador de Colônias de Granulócitos/sangue , Camundongos , Núcleo Accumbens/efeitos dos fármacos , Recompensa
14.
Eur J Neurosci ; 50(3): 2562-2573, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30179286

RESUMO

Substance use disorders are global health problems with few effective treatment options. Unfortunately, most potential pharmacological treatments are hindered by abuse potential of their own, limited efficacy, or adverse side effects. As a consequence, there is a pressing need for the development of addiction treatments with limited abuse potential and fewer off target effects. Given the difficulties in developing new pharmacotherapies for substance use disorders, there has been growing interest in medications that act on non-traditional targets. Recent evidence suggests a role for dysregulated immune signaling in the pathophysiology of multiple psychiatric diseases. While there is evidence that immune responses in the periphery and the central nervous system are altered by exposure to drugs of abuse, the contributions of neuroimmune interactions to addictive behaviors are just beginning to be appreciated. In this review, we discuss the data on immunological changes seen in clinical populations with substance use disorders, as well as in translational animal models of addiction. Importantly, we highlight those mechanistic findings showing causal roles for central or peripheral immune mediators in substance use disorder and appropriate animal models. Based on the literature reviewed here, it is clear that brain-immune system interactions in substance use disorders are much more complex and important than previously understood. While much work remains to be done, there are tremendous potential therapeutic implications for immunomodulatory treatments in substance use disorders.


Assuntos
Encéfalo/imunologia , Estimulantes do Sistema Nervoso Central/efeitos adversos , Microbioma Gastrointestinal/imunologia , Neuroimunomodulação/imunologia , Transtornos Relacionados ao Uso de Opioides/imunologia , Animais , Encéfalo/efeitos dos fármacos , Microbioma Gastrointestinal/efeitos dos fármacos , Humanos , Fatores Imunológicos/farmacologia , Fatores Imunológicos/uso terapêutico , Mediadores da Inflamação/imunologia , Neuroimunomodulação/efeitos dos fármacos , Transtornos Relacionados ao Uso de Opioides/tratamento farmacológico
15.
Proteomes ; 6(4)2018 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-30249060

RESUMO

Cocaine addiction is characterized by aberrant plasticity of the mesolimbic dopamine circuit, leading to dysregulation of motivation to seek and take drug. Despite the significant toll that cocaine use disorder exacts on society, there are currently no available pharmacotherapies. We have recently identified granulocyte-colony stimulating factor (G-CSF) as a soluble cytokine that alters the behavioral response to cocaine and which increases dopamine release from the ventral tegmental area (VTA). Despite these known effects on behavior and neurophysiology, the molecular mechanisms by which G-CSF affects brain function are unclear. In this study mice were treated with repeated injections of G-CSF, cocaine or a combination and changes in protein expression in the VTA were examined using an unbiased proteomics approach. Repeated G-CSF treatment resulted in alterations in multiple signaling pathways related to synaptic plasticity and neuronal morphology. While the treatment groups had marked overlap in their effect, injections of cocaine and the combination of cocaine and G-CSF lead to distinct patterns of significantly regulated proteins. These experiments provide valuable information as to the molecular pathways that G-CSF activates in an important limbic brain region and will help to guide further characterization of G-CSF function and evaluation as a possible translational target.

16.
J Neurosci ; 38(41): 8845-8859, 2018 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-30150359

RESUMO

Deficits in motivation and cognition are hallmark symptoms of multiple psychiatric diseases. These symptoms are disruptive to quality of life and often do not improve with available medications. In recent years there has been increased interest in the role of the immune system in neuropsychiatric illness, but to date no immune-related treatment strategies have come to fruition. The cytokine granulocyte-colony stimulating factor (G-CSF) is known to have trophic and neuroprotective properties in the brain, and we recently identified it as a modulator of neuronal and behavioral plasticity. By combining operant tasks that assess discrete aspects of motivated behavior and decision-making in male mice and rats with subsecond dopamine monitoring via fast-scan cyclic voltammetry, we defined the role of G-CSF in these processes as well as the neural mechanism by which it modulates dopamine function to exert these effects. G-CSF enhanced motivation for sucrose as well as cognitive flexibility as measured by reversal learning. These behavioral outcomes were driven by mesolimbic dopamine system plasticity, as systemically administered G-CSF increased evoked dopamine release in the nucleus accumbens independent of clearance mechanisms. Importantly, sustained increases in G-CSF were required for these effects as acute exposure did not enhance behavioral outcomes and decreased dopamine release. These effects seem to be a result of the ability of G-CSF to alter local inflammatory signaling cascades, particularly tumor necrosis factor α. Together, these data show G-CSF as a potent modulator of the mesolimbic dopamine circuit and its ability to appropriately attend to salient stimuli.SIGNIFICANCE STATEMENT Emerging evidence has highlighted the importance of the immune system in psychiatric diseases states. However, the effects of peripheral cytokines on motivation and cognitive function are largely unknown. Here, we report that granulocyte-colony stimulating factor (G-CSF), a pleiotropic cytokine with known trophic and neuroprotective properties in the brain, acts directly on dopaminergic circuits to enhance their function. These changes in dopaminergic dynamics enhance reward learning and motivation for natural stimuli. Together, these results suggest that targeting immune factors may provide a new avenue for therapeutic intervention in the multiple psychiatric disorders that are characterized by motivational and cognitive deficits.


Assuntos
Condicionamento Operante/fisiologia , Dopamina/fisiologia , Fator Estimulador de Colônias de Granulócitos/fisiologia , Motivação/fisiologia , Núcleo Accumbens/fisiologia , Recompensa , Animais , Tomada de Decisões/fisiologia , Fator Estimulador de Colônias de Granulócitos/administração & dosagem , Masculino , Camundongos Endogâmicos C57BL , Núcleo Accumbens/imunologia , Ratos Sprague-Dawley , Reversão de Aprendizagem/fisiologia , Sacarose/administração & dosagem
17.
Psychopharmacology (Berl) ; 235(5): 1427-1437, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29455291

RESUMO

RATIONALE: Laboratory experiments often model risk through a choice between a large, uncertain (LU) reward against a small, certain (SC) reward as an index of an individual's risk tolerance. An important factor generally lacking from these procedures are reward-associated cues that may modulate risk preferences. OBJECTIVE: We tested whether the addition of cues signaling 'jackpot' wins to LU choices would modulate risk preferences and if these cue effects were mediated by dopaminergic signaling. METHODS: Three groups of rats chose between LU and SC rewards for which the LU probability of reward decreased across blocks. The unsignaled group received a non-informative stimulus of trial outcome. The signaled group received a jackpot signal prior to reward delivery and blackout on losses. The signaled-light group received a similar jackpot for wins, but a salient loss signal distinct from the win signal. RESULTS: Presenting win signals decreased the discounting of LU value for both signaled groups regardless of loss signal, while the unsignaled group showed discounting similar to previous research without cues. Pharmacological challenges with D1/D2 agonists and antagonists revealed that D1 antagonism increased and decreased sensitives to the relative probability of reward for unsignaled and signaled groups, respectively, while D2 agonists decreased sensitivities to the relative magnitude of reward. CONCLUSION: The results highlight how signals predictive of wins can promote maladaptive risk taking in individuals, while loss signals have reduced effect. Additionally, the presence of reward-predictive cues may change the underlying neurobehavioral mechanisms mediating decision-making under risk.


Assuntos
Tomada de Decisões/efeitos dos fármacos , Agonistas de Dopamina/farmacologia , Antagonistas de Dopamina/farmacologia , Receptores de Dopamina D1/antagonistas & inibidores , Receptores de Dopamina D2/agonistas , Recompensa , Animais , Comportamento de Escolha/efeitos dos fármacos , Comportamento de Escolha/fisiologia , Sinais (Psicologia) , Tomada de Decisões/fisiologia , Dopamina/farmacologia , Relação Dose-Resposta a Droga , Masculino , Ratos , Ratos Sprague-Dawley , Receptores de Dopamina D1/fisiologia , Receptores de Dopamina D2/fisiologia , Assunção de Riscos
18.
Behav Brain Res ; 338: 143-152, 2018 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-29061385

RESUMO

Until recently, there were few rodent models available to study the interaction of post-traumatic stress disorder (PTSD) and drug taking. Like PTSD, single prolonged stress (SPS) produces hypothalamic-pituitary-adrenal (HPA) axis dysfunction and alters psychostimulant self-administration. Other stressors, such as isolation stress, also alter psychostimulant self-administration. However, it is currently unknown if isolation housing combined with SPS can alter the acquisition or maintenance of cocaine self-administration. The current study applied modified SPS (modSPS; two hours restraint immediately followed by cold swim stress) to rats raised in an isolation condition (Iso), enrichment condition (Enr), or standard condition (Std) to measure changes in cocaine self-administration and HPA markers. Regardless of rearing condition, rats exposed to modSPS had greater corticosterone (CORT) release and reduced cocaine self-administration during initial acquisition compared to non-stressed controls. In addition, during initial acquisition, rats that received both Iso rearing and modSPS showed a more rapid increase in cocaine self-administration across sessions compared to Enr and Std rats exposed to modSPS. Following initial acquisition, a dose response analysis showed that Iso rats were overall most sensitive to changes in cocaine unit dose; however, modSPS had no effect on the cocaine dose response curve. Further, there was no effect of either modSPS or differential rearing on expression of glucocorticoid receptor (GR) in hypothalamus, medial prefrontal cortex, amygdala, or nucleus accumbens. By using modSPS in combination with Iso housing, this study identified unique contributions of each stressor to acquisition of cocaine self-administration.


Assuntos
Cocaína/administração & dosagem , Corticosterona/sangue , Inibidores da Captação de Dopamina/administração & dosagem , Receptores de Glucocorticoides/metabolismo , Isolamento Social , Estresse Psicológico/sangue , Tonsila do Cerebelo/efeitos dos fármacos , Tonsila do Cerebelo/metabolismo , Animais , Abrigo para Animais , Masculino , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/metabolismo , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/metabolismo , Ratos , Ratos Sprague-Dawley , Restrição Física , Autoadministração
19.
Psychopharmacology (Berl) ; 234(23-24): 3499-3506, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28916995

RESUMO

BACKGROUND: Opioid abuse is a major problem around the world. Identifying environmental factors that contribute to opioid abuse and addiction is necessary for decreasing this epidemic. In rodents, environmental enrichment protects against the development of low dose stimulant self-administration, but studies examining the effect of enrichment and isolation (compared to standard housing) on the development of intravenous opioid self-administration have not been conducted. The present study investigated the role of environmental enrichment on self-administration of the short-acting µ-opioid remifentanil. METHODS: Rats were raised in an enriched condition (Enr), standard condition (Std), or isolated condition (Iso) beginning at 21 days of age and were trained to lever press for 1 or 3 µg/kg/infusion remifentanil in young adulthood. Acquisition of self-administration and responding during increasing fixed ratio requirements were assessed, and a dose-response curve was generated. RESULTS: In all phases, Enr rats lever pressed significantly less than Std and Iso rats, with Enr rats pressing between 9 and 40% the amount of Iso rats. Enr rats did not acquire remifentanil self-administration when trained with 1 µg/kg/infusion, did not increase responding over increasing FR when trained at either dose, and their dose-response curves were flattened compared to Std and Iso rats. When expressed as economic demand curves, Enr rats displayed a decrease in both essential value (higher α) and reinforcer intensity (Q 0) compared to Std and Iso rats at the 1 µg/kg/infusion training dose. CONCLUSION: Environmental enrichment reduced remifentanil intake, suggesting that social and environmental novelty may protect against opioid abuse.


Assuntos
Analgésicos Opioides/administração & dosagem , Comportamento Aditivo/psicologia , Meio Ambiente , Transtornos Relacionados ao Uso de Opioides/psicologia , Piperidinas/administração & dosagem , Animais , Comportamento Aditivo/prevenção & controle , Relação Dose-Resposta a Droga , Infusões Intravenosas , Masculino , Transtornos Relacionados ao Uso de Opioides/prevenção & controle , Ratos , Ratos Sprague-Dawley , Remifentanil , Autoadministração , Resultado do Tratamento
20.
Drug Alcohol Depend ; 167: 89-94, 2016 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-27511893

RESUMO

BACKGROUND: Research has shown that previous experiences during development, especially if stressful, can alter an organism's response to opioids later in life. Given the previous literature on opioid modulation of cocaine self-administration, the current study raised rats in either an enriched condition (EC) or isolated condition (IC) and employed behavioral economics to study the effects of naltrexone and morphine on cocaine self-administration. METHODS: EC and IC rats were trained to lever press for cocaine using a within-session demand procedure. This procedure measured cocaine consumption under changing cocaine price by decreasing the dose of cocaine earned throughout a session. Rats were able to self-administer cocaine on a FR1; every 10min the cocaine dose was systematically decreased (0.75-0.003mg/kg/infusion cocaine). After reaching stability on this procedure, rats were randomly pretreated with 0, 0.3, 1, or 3mg/kg naltrexone once every 3days, followed by random pretreatments of 0, 0.3, 1, or 3mg/kg morphine once every 3days. Economic demand functions were fit to each rat's cocaine consumption from each pretreatment, and appropriate mathematical parameters were extracted and analyzed. RESULTS: Naltrexone decreased the essential value of cocaine in IC rats only. However, morphine decreased the essential value of cocaine and the consumption of cocaine at zero price in both EC and IC rats. CONCLUSION: These results indicate that environmental experiences during development should be considered when determining the efficacy of opioid drugs, especially for the treatment of substance abuse.


Assuntos
Analgésicos Opioides/farmacologia , Comportamento Animal/efeitos dos fármacos , Cocaína/administração & dosagem , Morfina/farmacologia , Naltrexona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Animais , Economia Comportamental , Meio Ambiente , Masculino , Ratos , Ratos Sprague-Dawley , Autoadministração , Transtornos Relacionados ao Uso de Substâncias/psicologia , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...