Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
Add more filters










Publication year range
1.
JMIR Public Health Surveill ; 10: e52691, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38701436

ABSTRACT

BACKGROUND: Structural racism produces mental health disparities. While studies have examined the impact of individual factors such as poverty and education, the collective contribution of these elements, as manifestations of structural racism, has been less explored. Milwaukee County, Wisconsin, with its racial and socioeconomic diversity, provides a unique context for this multifactorial investigation. OBJECTIVE: This research aimed to delineate the association between structural racism and mental health disparities in Milwaukee County, using a combination of geospatial and deep learning techniques. We used secondary data sets where all data were aggregated and anonymized before being released by federal agencies. METHODS: We compiled 217 georeferenced explanatory variables across domains, initially deliberately excluding race-based factors to focus on nonracial determinants. This approach was designed to reveal the underlying patterns of risk factors contributing to poor mental health, subsequently reintegrating race to assess the effects of racism quantitatively. The variable selection combined tree-based methods (random forest) and conventional techniques, supported by variance inflation factor and Pearson correlation analysis for multicollinearity mitigation. The geographically weighted random forest model was used to investigate spatial heterogeneity and dependence. Self-organizing maps, combined with K-means clustering, were used to analyze data from Milwaukee communities, focusing on quantifying the impact of structural racism on the prevalence of poor mental health. RESULTS: While 12 influential factors collectively accounted for 95.11% of the variability in mental health across communities, the top 6 factors-smoking, poverty, insufficient sleep, lack of health insurance, employment, and age-were particularly impactful. Predominantly, African American neighborhoods were disproportionately affected, which is 2.23 times more likely to encounter high-risk clusters for poor mental health. CONCLUSIONS: The findings demonstrate that structural racism shapes mental health disparities, with Black community members disproportionately impacted. The multifaceted methodological approach underscores the value of integrating geospatial analysis and deep learning to understand complex social determinants of mental health. These insights highlight the need for targeted interventions, addressing both individual and systemic factors to mitigate mental health disparities rooted in structural racism.


Subject(s)
Machine Learning , Humans , Wisconsin/epidemiology , Female , Male , Mental Health/statistics & numerical data , Health Status Disparities , Spatial Analysis , Adult , Systemic Racism/statistics & numerical data , Systemic Racism/psychology , Racism/statistics & numerical data , Racism/psychology , Middle Aged
2.
Mol Psychiatry ; 28(9): 3930-3942, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37845497

ABSTRACT

Chronic cocaine exposure induces enduring neuroadaptations that facilitate motivated drug taking. Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels are known to modulate neuronal firing and pacemaker activity in ventral tegmental area (VTA) dopamine neurons. However, it remained unknown whether cocaine self-administration affects HCN channel function and whether HCN channel activity modulates motivated drug taking. We report that rat VTA dopamine neurons predominantly express Hcn3-4 mRNA, while VTA GABA neurons express Hcn1-4 mRNA. Both neuronal types display similar hyperpolarization-activated currents (Ih), which are facilitated by acute increases in cAMP. Acute cocaine application decreases voltage-dependent activation of Ih in VTA dopamine neurons, but not in GABA neurons. Unexpectedly, chronic cocaine self-administration results in enhanced Ih selectively in VTA dopamine neurons. This differential modulation of Ih currents is likely mediated by a D2 autoreceptor-induced decrease in cAMP as D2 (Drd2) mRNA is predominantly expressed in dopamine neurons, whereas D1 (Drd1) mRNA is barely detectable in the VTA. Moreover, chronically decreased cAMP via Gi-DREADD stimulation leads to an increase in Ih in VTA dopamine neurons and enhanced binding of HCN3/HCN4 with tetratricopeptide repeat-containing Rab8b-interacting protein (TRIP8b), an auxiliary subunit that is known to facilitate HCN channel surface trafficking. Finally, we show that systemic injection and intra-VTA infusion of the HCN blocker ivabradine reduces cocaine self-administration under a progressive ratio schedule and produces a downward shift of the cocaine dose-response curve. Our results suggest that cocaine self-administration induces an upregulation of Ih in VTA dopamine neurons, while HCN inhibition reduces the motivation for cocaine intake.


Subject(s)
Cocaine , Dopaminergic Neurons , Rats , Animals , Dopaminergic Neurons/metabolism , Ventral Tegmental Area/metabolism , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/genetics , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Up-Regulation , Cocaine/pharmacology , RNA, Messenger
3.
Neuropsychopharmacology ; 48(8): 1121-1132, 2023 07.
Article in English | MEDLINE | ID: mdl-37188846

ABSTRACT

Stress is prevalent in the lives of those with substance use disorders (SUDs) and influences SUD outcomes. Understanding the neurobiological mechanisms through which stress promotes drug use is important for the development of effective SUD interventions. We have developed a model wherein exposure to a stressor, uncontrollable electric footshock, daily at the time of cocaine self-administration escalates intake in male rats. Here we test the hypothesis that stress-induced escalation of cocaine self-administration requires the CB1 cannabinoid receptor. Male Sprague-Dawley rats self-administered cocaine (0.5 mg/kg/inf, i.v.) during 2-h sessions comprised of four 30-min self-administration components separated by 5-min shock sequences or 5-min shock-free periods for 14 days. Footshock produced an escalation of cocaine self-administration that persisted following shock removal. Systemic administration of the cannabinoid receptor type 1 (CB1R) antagonist/inverse agonist, AM251, attenuated cocaine intake only in rats with a history of stress. This effect was localized to the mesolimbic system, as intra-nucleus accumbens (NAc) shell and intra-ventral tegmental area (VTA) micro-infusions of AM251 attenuated cocaine intake only in stress-escalated rats. Cocaine self-administration, regardless of stress history, increased CB1R binding site density in the VTA, but not NAc shell. Following extinction, cocaine-primed reinstatement (10 mg/kg, ip) was increased in rats with prior footshock during self-administration. AM251 attenuated reinstatement only in rats with a stress history. Altogether, these data demonstrate that mesolimbic CB1Rs are required to escalate intake and heighten relapse susceptibility and suggest that repeated stress at the time of cocaine use regulates mesolimbic CB1R activity through a currently unknown mechanism.


Subject(s)
Cocaine-Related Disorders , Cocaine , Rats , Male , Animals , Rats, Sprague-Dawley , Drug Inverse Agonism , Cocaine-Related Disorders/metabolism , Nucleus Accumbens/metabolism , Self Administration , Receptors, Cannabinoid/metabolism
4.
Drug Alcohol Depend ; 245: 109827, 2023 04 01.
Article in English | MEDLINE | ID: mdl-36868092

ABSTRACT

INTRODUCTION: Drug overdose deaths are often geographically discordant (the community in which the overdose death occurs is different from the community of residence). Thus, in many cases there is a journey to overdose. METHODS: We applied geospatial analysis to examine characteristics that define journeys to overdoses using Milwaukee, Wisconsin, a diverse and segregated metropolitan area in which 26.72 % of overdose deaths are geographically discordant, as a case study. First, we deployed spatial social network analysis to identify hubs (census tracts that are focal points of geographically discordant overdoses) and authorities (the communities of residence from which journeys to overdose commonly begin) for overdose deaths and characterized them according to key demographics. Second, we used temporal trend analysis to identify communities that were consistent, sporadic, and emergent hotspots for overdose deaths. Third, we identified characteristics that differentiated discordant versus non-discordant overdose deaths. RESULTS: Authority communities had lower housing stability and were younger, more impoverished, and less educated relative to hubs and county-wide numbers. White communities were more likely to be hubs, while Hispanic communities were more likely to be authorities. Geographically discordant deaths more commonly involved fentanyl, cocaine, and amphetamines and were more likely to be accidental. Non-discordant deaths more commonly involved opioids other than fentanyl or heroin and were more likely to be the result of suicide. CONCLUSION: This study is the first to examine the journey to overdose and demonstrates that such analysis can be applied in metropolitan areas to better understand and guide community responses.


Subject(s)
Drug Overdose , Social Network Analysis , Humans , Analgesics, Opioid , Heroin , Fentanyl
5.
Addict Neurosci ; 42022 Dec.
Article in English | MEDLINE | ID: mdl-36531188

ABSTRACT

The neuropeptide, corticotropin releasing factor (CRF), has been an enigmatic target for the development of medications aimed at treating stress-related disorders. Despite a large body of evidence from preclinical studies in rodents demonstrating that CRF receptor antagonists prevent stressor-induced drug seeking, medications targeting the CRF-R1 have failed in clinical trials. Here, we provide an overview of the abundant findings from preclinical rodent studies suggesting that CRF signaling is involved in stressor-induced relapse. The scientific literature that has defined the receptors, mechanisms and neurocircuits through which CRF contributes to stressor-induced reinstatement of drug seeking following self-administration and conditioned place preference in rodents is reviewed. Evidence that CRF signaling is recruited with repeated drug use in a manner that heightens susceptibility to stressor-induced drug seeking in rodents is presented. Factors that may determine the influence of CRF signaling in substance use disorders, including developmental windows, biological sex, and genetics are examined. Finally, we discuss the translational failure of medications targeting CRF signaling as interventions for substance use disorders and other stress-related conditions. We conclude that new perspectives and research directions are needed to unravel the mysterious role of CRF in substance use disorders.

6.
J Urban Health ; 99(2): 316-327, 2022 04.
Article in English | MEDLINE | ID: mdl-35181834

ABSTRACT

The effects of the opioid crisis have varied across diverse and socioeconomically defined urban communities, due in part to widening health disparities. The onset of the COVID-19 pandemic has coincided with a spike in drug overdose deaths in the USA. However, the extent to which the impact of the pandemic on overdose deaths has varied across different demographics in urban neighborhoods is unclear. We examine the influence of COVID-19 pandemic on opioid overdose deaths through spatiotemporal analysis techniques. Using Milwaukee County, Wisconsin as a study site, we used georeferenced opioid overdose data to examine the locational and demographic differences in overdose deaths over time (2017-2020). We find that the pandemic significantly increased the monthly overdose deaths. The worst effects were seen in the poor, urban neighborhoods, affecting Black and Hispanic communities. However, more affluent, suburban White communities also experienced a rise in overdose deaths. A better understanding of contributing factors is needed to guide interventions at the local, regional, and national scales.


Subject(s)
COVID-19 , Drug Overdose , Opiate Overdose , Analgesics, Opioid , Drug Overdose/epidemiology , Humans , Opiate Overdose/epidemiology , Pandemics , Spatio-Temporal Analysis
7.
J Urban Health ; 98(4): 551-562, 2021 08.
Article in English | MEDLINE | ID: mdl-34231120

ABSTRACT

To provide data that can guide community-targeted practices, policies, and interventions in urban metropolitan areas, we used geospatial analysis to examine the community-level opioid overdose death determinants and their spatial variation across a study area. We obtained spatial datasets containing multiple, high-quality measures of socioeconomic conditions, public health status, and demographics for analysis and visualization in geographic information systems. We employed a multiscale modeling approach (multiscale geographically weighted regression; MGWR) to provide a comprehensive and robust analysis of opioid overdose death determinants, explain how geospatial patterns vary across scales across Milwaukee County in 2019, and examine the differential influence of factors locally, regionally, and globally. We subsequently examined how associations varied with the racial/ethnic composition of communities by dividing Milwaukee County into White-majority, Black-majority, and Hispanic-majority regions according to census data and conducting separate, independent modeling processes. Overall, the multiscale model explained 83% of opioid overdose death variability across neighborhoods in Milwaukee County using 12 selected variables. Statistical analysis and geovisualization of patterns, trends, and clusters using MGWR unveiled dramatic racialized health disparities in Milwaukee, showing how factors that influenced opioid overdose deaths varied across diverse communities in Milwaukee. The observed geographic variation in relationships included the impact of naloxone availability and incarceration rates on overdose deaths with pronounced differences between White communities and communities of color. Understanding, community-level factors that contribute to overdose risk should guide targeted community-level solutions. Overall, our findings demonstrate the value of precision epidemiology using MGWR analysis for defining and guiding responses to public health challenges.


Subject(s)
Drug Overdose , Opiate Overdose , Analgesics, Opioid , Humans , Racial Groups , Spatial Regression
8.
J Neurosci ; 41(24): 5303-5314, 2021 06 16.
Article in English | MEDLINE | ID: mdl-33879537

ABSTRACT

Relapse susceptibility in women with substance use disorders (SUDs) has been linked to the estrogen, 17ß-estradiol (E2). Our previous findings in female rats suggest that the influence of E2 on cocaine seeking can be localized to the prelimbic prefrontal cortex (PrL-PFC). Here, we investigated the receptor mechanisms through which E2 regulates the reinstatement of extinguished cocaine seeking. Sexually mature female rats underwent intravenous cocaine self-administration (0.5 mg/inf; 14 × 2 h daily) and extinction, and then were ovariectomized before reinstatement testing. E2 (10 µg/kg, i.p.) alone did not reinstate cocaine seeking, but it potentiated reinstatement when combined with an otherwise subthreshold priming dose of cocaine. A similar effect was observed following intra-PrL-PFC microinfusions of E2 and by systemic or intra-PrL-PFC administration of the estrogen receptor (ER)ß agonist, DPN, but not agonists at ERα or the G-protein-coupled ER1 (GPER1). By contrast, E2-potentiated reinstatement was prevented by intra-PrL-PFC microinfusions of the ERß antagonist, MPP, or the GPER1 antagonist, G15, but not an ERα antagonist. Whole-cell recordings in PrL-PFC layer (L)5/6 pyramidal neurons revealed that E2 decreases the frequency, but not amplitude, of GABAA-dependent miniature IPSCs (mIPSC). As was the case with E2-potentiated reinstatement, E2 reductions in mIPSC frequency were prevented by ERß and GPER1, but not ERα, antagonists and mimicked by ERß, but not GPER1, agonists. Altogether, the findings suggest that E2 activates ERß and GPER1 in the PrL-PFC to attenuate the GABA-mediated constraint of key outputs that mediate cocaine seeking.


Subject(s)
Cocaine-Related Disorders/metabolism , Drug-Seeking Behavior/physiology , Estradiol/metabolism , Prefrontal Cortex/metabolism , Animals , Estrogen Receptor beta/metabolism , Extinction, Psychological/physiology , Female , Rats , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/metabolism
9.
J Neurochem ; 157(5): 1697-1713, 2021 06.
Article in English | MEDLINE | ID: mdl-33660857

ABSTRACT

In individuals with substance use disorders, stress is a critical determinant of relapse susceptibility. In some cases, stressors directly trigger cocaine use. In others, stressors interact with other stimuli to promote drug seeking, thereby setting the stage for relapse. Here, we review the mechanisms and neurocircuitry that mediate stress-triggered and stress-potentiated cocaine seeking. Stressors trigger cocaine seeking by activating noradrenergic projections originating in the lateral tegmentum that innervate the bed nucleus of the stria terminalis to produce beta adrenergic receptor-dependent regulation of neurons that release corticotropin releasing factor (CRF) into the ventral tegmental area (VTA). CRF promotes the activation of VTA dopamine neurons that innervate the prelimbic prefrontal cortex resulting in D1 receptor-dependent excitation of a pathway to the nucleus accumbens core that mediates cocaine seeking. The stage-setting effects of stress require glucocorticoids, which exert rapid non-canonical effects at several sites within the mesocorticolimbic system. In the nucleus accumbens, corticosterone attenuates dopamine clearance via the organic cation transporter 3 to promote dopamine signaling. In the prelimbic cortex, corticosterone mobilizes the endocannabinoid, 2-arachidonoylglycerol (2-AG), which produces CB1 receptor-dependent reductions in inhibitory transmission, thereby increasing excitability of neurons which comprise output pathways responsible for cocaine seeking. Factors that influence the role of stress in cocaine seeking, including prior history of drug use, biological sex, chronic stress/co-morbid stress-related disorders, adolescence, social variables, and genetics are discussed. Better understanding when and how stress contributes to drug seeking should guide the development of more effective interventions, particularly for those whose drug use is stress related.


Subject(s)
Brain Chemistry , Cocaine-Related Disorders/physiopathology , Drug-Seeking Behavior , Nerve Net/physiopathology , Stress, Psychological/physiopathology , Animals , Cocaine-Related Disorders/psychology , Humans , Stress, Psychological/psychology
10.
J Neurosci ; 41(11): 2428-2436, 2021 03 17.
Article in English | MEDLINE | ID: mdl-33622777

ABSTRACT

Chronic stress impairs the function of multiple brain regions and causes severe hedonic and motivational deficits. One brain region known to be susceptible to these effects is the PFC. Neurons in this region, specifically neuronal projections from the prelimbic region (PL) to the nucleus accumbens core (NAcC), have a significant role in promoting motivated approach. However, little is known about how activity in this pathway changes during associative learning to encode cues that promote approach. Less is known about how activity in this pathway may be altered by stress. In this study, an intersectional fiber photometry approach was used in male Sprague Dawley rats engaged in a Pavlovian autoshaping design to characterize the involvement of the PL-NAcC pathway in the typical acquisition of learned approach (directed at both the predictive cue and the goal), and its potential alteration by stress. Specifically, the hypothesis that neural activity in PL-NAcC would encode a Pavlovian approach cue and that prior exposure to chronic stress would disrupt both the nature of conditioned approach and the encoding of a cue that promotes approach was tested. Results of the study demonstrated that the rapid acquisition of conditioned approach was associated with cue-induced PL-NAcC activity. Prior stress both reduced cue-directed behavior and impaired the associated cortical activity. These findings demonstrate that prior stress diminishes the task-related activity of a brain pathway that regulates approach behavior. In addition, the results support the interpretation that stress disrupts reward processing by altering the incentive value of associated cues.SIGNIFICANCE STATEMENT Chronic stress causes hedonic and motivational deficits and disrupts the function of the PFC. A specific projection from the prelimbic region of the PFC to the nucleus accumbens core (PL-NAcC) promotes approach behavior and is a strong candidate for contributing to stress-induced disruptions in motivation. However, it is not known how activity in this pathway encodes cues that promote approach, and how this encoding may be altered by stress. Here we show that the rapid acquisition of conditioned approach is associated with cue-induced activity in the PL-NAcC pathway. Prior stress both reduces cue-directed behavior and impairs the associated cortical activity. These findings demonstrate that stress diminishes task-related activity in a brain pathway that regulates approach behavior.


Subject(s)
Brain/physiopathology , Conditioning, Classical/physiology , Neural Pathways/physiopathology , Stress, Psychological/physiopathology , Animals , Cues , Male , Rats , Rats, Sprague-Dawley
11.
Neuropsychopharmacology ; 45(12): 1974-1985, 2020 11.
Article in English | MEDLINE | ID: mdl-32303052

ABSTRACT

Clinical reports suggest that females diagnosed with substance use disorder experience enhanced relapse vulnerability compared with males, particularly during stress. We previously demonstrated that a stressor (footshock) can potentiate cocaine seeking in male rats via glucocorticoid-dependent cannabinoid type-1 receptor (CB1R)-mediated actions in the prelimbic prefrontal cortex (PrL-PFC). Here, we investigated the influence of biological sex on stress-potentiated cocaine seeking. Despite comparable self-administration and extinction, females displayed a lower threshold for cocaine-primed reinstatement than males. Unlike males, footshock, tested across a range of intensities, failed to potentiate cocaine-primed reinstatement in females. However, restraint potentiated reinstatement in both sexes. While sex differences in stressor-induced plasma corticosterone (CORT) elevations and defensive behaviors were not observed, differences were evident in footshock-elicited ultrasonic vocalizations. CORT administration, at a dose which recapitulates stressor-induced plasma levels, reproduced stress-potentiated cocaine-primed reinstatement in both sexes. In females, CORT effects varied across the estrous cycle; CORT-potentiated reinstatement was only observed during diestrus and proestrus. As in males, CORT-potentiated cocaine seeking in females was localized to the PrL-PFC and both CORT- and restraint-potentiated cocaine seeking required PrL-PFC CB1R activation. In addition, ex vivo whole-cell electrophysiological recordings from female layer V PrL-PFC pyramidal neurons revealed CB1R-dependent CORT-induced suppression of inhibitory synaptic activity, as previously observed in males. These findings demonstrate that, while stress potentiates cocaine seeking via PrL-PFC CB1R in both sexes, sensitivity to cocaine priming injections is greater in females, CORT-potentiating effects vary with the estrous cycle, and whether reactivity to specific stressors may manifest as drug seeking depends on biological sex.


Subject(s)
Cocaine , Animals , Drug-Seeking Behavior , Extinction, Psychological , Female , Male , Prefrontal Cortex , Rats , Rats, Sprague-Dawley , Self Administration
12.
Neurobiol Stress ; 9: 271-285, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30450391

ABSTRACT

Despite extensive research efforts, drug addiction persists as a largely unmet medical need. Perhaps the biggest challenge for treating addiction is the high rate of recidivism. While many factors can promote relapse in abstinent drug users, the contribution of stress is particularly problematic, as stress is uncontrollable and pervasive in the lives of those struggling with addiction. Thus, understanding the neurocircuitry that underlies the influence of stress on drug seeking is critical for guiding treatment. Preclinical research aimed at defining this neurocircuitry has, in part, relied upon the use of experimental approaches that allow visualization of cellular and circuit activity that corresponds to stressor-induced drug seeking in rodent relapse models. Much of what we have learned about the mechanisms that mediate stressor-induced relapse has been informed by studies that have used the expression of the immediate early gene, cfos, or its protein product, Fos, as post-mortem activity markers. In this review we provide an overview of the rodent models used to study stressor-induced relapse and briefly summarize what is known about the underlying neurocircuitry before describing the use of cfos/Fos-based approaches. In addition to reviewing findings obtained using this approach, its advantages and limitations are considered. Moreover, new techniques that leverage the expression profile of cfos to tag and manipulate cells based on their activity patterns are discussed. The intent of the review is to guide the interpretation of old and design of new studies that utilize cfos/Fos-based strategies to study the neurocircuitry that contributes to stress-related drug use.

13.
J Neurosci ; 38(50): 10657-10671, 2018 12 12.
Article in English | MEDLINE | ID: mdl-30355627

ABSTRACT

The ability of stress to trigger cocaine seeking in humans and rodents is variable and is determined by the amount and pattern of prior drug use. This study examined the role of a corticotropin releasing factor (CRF)-regulated dopaminergic projection from the ventral tegmental area (VTA) to the prelimbic cortex in shock-induced cocaine seeking and its recruitment under self-administration conditions that establish relapse vulnerability. Male rats with a history of daily long-access (LgA; 14 × 6 h/d) but not short-access (ShA; 14 × 2 h/d) self-administration showed robust shock-induced cocaine seeking. This was associated with a heightened shock-induced prelimbic cortex Fos response and activation of cholera toxin b retro-labeled VTA neurons that project to the prelimbic cortex. Chemogenetic inhibition of this pathway using a dual virus intersectional hM4Di DREADD (designer receptor exclusively activated by designer drug) based approach prevented shock-induced cocaine seeking. Both shock-induced reinstatement and the prelimbic cortex Fos response were prevented by bilateral intra-VTA injections of the CRF receptor 1 (CRFR1) antagonist, antalarmin. Moreover, pharmacological disconnection of the CRF-regulated dopaminergic projection to the prelimbic cortex by injection of antalarmin into the VTA in one hemisphere and the D1 receptor antagonist, SCH23390, into the prelimbic cortex of the contralateral hemisphere prevented shock-induced cocaine seeking. Finally, LgA, but not ShA, cocaine self-administration resulted in increased VTA CRFR1 mRNA levels as measured using in situ hybridization. Altogether, these findings suggest that excessive cocaine use may establish susceptibility to stress-induced relapse by recruiting CRF regulation of a stressor-responsive mesocortical dopaminergic pathway.SIGNIFICANCE STATEMENT Understanding the neural pathways and mechanisms through which stress triggers relapse to cocaine use is critical for the development of more effective treatment approaches. Prior work has demonstrated a critical role for the neuropeptide corticotropin releasing factor (CRF) in stress-induced cocaine seeking. Here we provide evidence that stress-induced reinstatement in a rat model of relapse is mediated by a CRF-regulated dopaminergic projection from the ventral tegmental area (VTA) that activates dopamine D1 receptors in the prelimbic cortex. Moreover, we report that this pathway may be recruited as a result of daily cocaine self-administration under conditions of extended drug access/heightened drug intake, likely as a result of increased CRFR1 expression in the VTA, thereby promoting susceptibility to stress-induced cocaine seeking.


Subject(s)
Cocaine/administration & dosage , Drug-Seeking Behavior/physiology , Prefrontal Cortex/physiology , Receptors, Corticotropin-Releasing Hormone/biosynthesis , Stress, Psychological/metabolism , Ventral Tegmental Area/physiology , Animals , Conditioning, Operant/physiology , Drug-Seeking Behavior/drug effects , Male , Neural Pathways/chemistry , Neural Pathways/physiology , Prefrontal Cortex/chemistry , Rats , Rats, Sprague-Dawley , Receptors, Corticotropin-Releasing Hormone/genetics , Self Administration , Stress, Psychological/psychology , Ventral Tegmental Area/chemistry
15.
Neuropsychopharmacology ; 43(4): 781-790, 2018 03.
Article in English | MEDLINE | ID: mdl-28825421

ABSTRACT

Clinical observations imply that female cocaine addicts experience enhanced relapse vulnerability compared with males, an effect tied to elevated estrogen phases of the ovarian hormone cycle. Although estrogens can enhance drug-seeking behavior, they do not directly induce reinstatement on their own. To model this phenomenon, we tested whether an estrogen could augment drug-seeking behavior in response to an ordinarily subthreshold reinstatement trigger. Following cocaine self-administration and extinction, female rats were ovariectomized to isolate estrogen effects on reinstatement. Although neither peak proestrus levels of the primary estrogen 17ß-estradiol (E2; 10 µg/kg, i.p., 1-h pretreatment) nor a subthreshold cocaine dose (1.25 mg/kg, i.p.) alone were sufficient to reinstate drug-seeking behavior, pretreatment with E2 potentiated reinstatement to the ordinarily subthreshold cocaine dose. Furthermore, E2 microinfusions revealed that E2 (5 µg/0.3 µl, 15-min pretreatment) acts directly within the prelimbic prefrontal cortex (PrL-PFC) to potentiate reinstatement. As E2 has been implicated in endocannabinoid mobilization, which can disinhibit PrL-PFC projection neurons, we investigated whether cannabinoid type-1 receptor (CB1R) activation is necessary for E2 to potentiate reinstatement. The CB1R antagonist AM251 (1 or 3 mg/kg, i.p., 30-min pretreatment) administered prior to E2 and cocaine suppressed reinstatement in a dose-dependent manner. Finally, PrL-PFC AM251 microinfusions (300 ng/side, 15-min pretreatment) also suppressed E2-potentiated reinstatement. Together, these results suggest that E2 can augment reactivity to an ordinarily subthreshold relapse trigger in a PrL-PFC CB1R activation-dependent manner.


Subject(s)
Cocaine/administration & dosage , Drug-Seeking Behavior/drug effects , Estradiol/metabolism , Estrogens/metabolism , Prefrontal Cortex/metabolism , Receptor, Cannabinoid, CB1/metabolism , Animals , Dopamine Uptake Inhibitors/administration & dosage , Dose-Response Relationship, Drug , Drug-Seeking Behavior/physiology , Estradiol/pharmacology , Estrogens/pharmacology , Female , Piperidines/pharmacology , Prefrontal Cortex/drug effects , Pyrazoles/pharmacology , Rats , Rats, Sprague-Dawley , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Self Administration , Sex Factors
16.
Biol Psychiatry ; 84(2): 85-94, 2018 07 15.
Article in English | MEDLINE | ID: mdl-29100630

ABSTRACT

BACKGROUND: Clinical reports suggest that rather than directly driving cocaine use, stress may create a biological context within which other triggers for drug use become more potent. We hypothesize that stress-induced increases in corticosterone "set the stage" for relapse by promoting endocannabinoid-induced attenuation of inhibitory transmission in the prelimbic cortex (PL). METHODS: We have established a rat model for these stage-setting effects of stress. In this model, neither a stressor (electric footshock) nor stress-level corticosterone treatment alone reinstates cocaine seeking following self-administration and extinction, but each treatment potentiates reinstatement in response to an otherwise subthreshold cocaine priming dose (2.5 mg/kg, intraperitoneal). The contributions of endocannabinoid signaling in the PL to the effects of stress-level corticosterone on PL neurotransmission and cocaine seeking were determined using intra-PL microinfusions. Endocannabinoid-dependent effects of corticosterone on inhibitory synaptic transmission in the rat PL were determined using whole-cell recordings in layer V pyramidal neurons. RESULTS: Corticosterone application attenuated inhibitory synaptic transmission in the PL via cannabinoid receptor type 1 (CB1R)- and 2-arachidonoylglycerol-dependent inhibition of gamma-aminobutyric acid release without altering postsynaptic responses. The ability of systemic stress-level corticosterone treatment to potentiate cocaine-primed reinstatement was recapitulated by intra-PL injection of corticosterone, the CB1R agonist WIN 55,212-2, or the monoacylglycerol lipase inhibitor URB602. Corticosterone effects on reinstatement were attenuated by intra-PL injections of either the CB1R antagonist, AM251, or the diacylglycerol lipase inhibitor, DO34. CONCLUSIONS: These findings suggest that stress-induced increases in corticosterone promote cocaine seeking by mobilizing 2-arachidonoylglycerol in the PL, resulting in CB1R-mediated attenuation of inhibitory transmission in this brain region.


Subject(s)
Cocaine-Related Disorders/physiopathology , Cocaine/administration & dosage , Drug-Seeking Behavior , Prefrontal Cortex/drug effects , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Stress, Psychological/physiopathology , Animals , Arachidonic Acids/pharmacology , Cocaine-Related Disorders/psychology , Endocannabinoids/pharmacology , Extinction, Psychological , Glucocorticoids/pharmacology , Glycerides/pharmacology , Male , Piperidines/pharmacology , Pyrazoles/pharmacology , Rats , Rats, Sprague-Dawley , Self Administration
17.
Eur J Neurosci ; 46(10): 2638-2646, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28965353

ABSTRACT

Stressful and aversive events promote maladaptive reward-seeking behaviors such as drug addiction by acting, in part, on the mesolimbic dopamine system. Using animal models, data from our laboratory and others show that stress and cocaine can interact to produce a synergistic effect on reward circuitry. This effect is also observed when the stress hormone corticosterone is administered directly into the nucleus accumbens (NAc), indicating that glucocorticoids act locally in dopamine terminal regions to enhance cocaine's effects on dopamine signaling. However, prior studies in behaving animals have not provided mechanistic insight. Using fast-scan cyclic voltammetry, we examined the effect of systemic corticosterone on spontaneous dopamine release events (transients) in the NAc core and shell in behaving rats. A physiologically relevant systemic injection of corticosterone (2 mg/kg i.p.) induced an increase in dopamine transient amplitude and duration (both voltammetric measures sensitive to decreases in dopamine clearance), but had no effect on the frequency of transient release events. This effect was compounded by cocaine (2.5 mg/kg i.p.). However, a second experiment indicated that the same injection of corticosterone had no detectable effect on the dopaminergic encoding of a palatable natural reward (saccharin). Taken together, these results suggest that corticosterone interferes with naturally occurring dopamine uptake locally, and this effect is a critical determinant of dopamine concentration specifically in situations in which the dopamine transporter is pharmacologically blocked by cocaine.


Subject(s)
Cocaine/administration & dosage , Corticosterone/metabolism , Dopamine Uptake Inhibitors/administration & dosage , Dopamine/metabolism , Nucleus Accumbens/metabolism , Animals , Corticosterone/administration & dosage , Male , Rats, Sprague-Dawley , Reward , Signal Transduction
18.
Neuropsychopharmacology ; 42(3): 757-765, 2017 02.
Article in English | MEDLINE | ID: mdl-27604564

ABSTRACT

The mechanisms by which stressful life events increase the risk of relapse in recovering cocaine addicts are not well understood. We previously reported that stress, via elevated corticosterone, potentiates cocaine-primed reinstatement of cocaine seeking following self-administration in rats and that this potentiation appears to involve corticosterone-induced blockade of dopamine clearance via the organic cation transporter 3 (OCT3). In the present study, we use a conditioned place preference/reinstatement paradigm in mice to directly test the hypothesis that corticosterone potentiates cocaine-primed reinstatement by blockade of OCT3. Consistent with our findings following self-administration in rats, pretreatment of male C57/BL6 mice with corticosterone (using a dose that reproduced stress-level plasma concentrations) potentiated cocaine-primed reinstatement of extinguished cocaine-induced conditioned place preference. Corticosterone failed to re-establish extinguished preference alone but produced a leftward shift in the dose-response curve for cocaine-primed reinstatement. A similar potentiating effect was observed upon pretreatment of mice with the non-glucocorticoid OCT3 blocker, normetanephrine. To determine the role of OCT3 blockade in these effects, we examined the abilities of corticosterone and normetanephrine to potentiate cocaine-primed reinstatement in OCT3-deficient and wild-type mice. Conditioned place preference, extinction and reinstatement of extinguished preference in response to low-dose cocaine administration did not differ between genotypes. However, corticosterone and normetanephrine failed to potentiate cocaine-primed reinstatement in OCT3-deficient mice. Together, these data provide the first direct evidence that the interaction of corticosterone with OCT3 mediates corticosterone effects on drug-seeking behavior and establish OCT3 function as an important determinant of susceptibility to cocaine use.


Subject(s)
Cocaine/pharmacology , Conditioning, Classical/drug effects , Corticosterone/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Normetanephrine/pharmacology , Octamer Transcription Factor-3/metabolism , Animals , Cocaine/administration & dosage , Corticosterone/administration & dosage , Dopamine Uptake Inhibitors/administration & dosage , Male , Mice , Mice, Inbred C57BL , Normetanephrine/administration & dosage , Octamer Transcription Factor-3/antagonists & inhibitors , Octamer Transcription Factor-3/deficiency
19.
J Neurochem ; 137(3): 384-93, 2016 May.
Article in English | MEDLINE | ID: mdl-26851652

ABSTRACT

Glutamate signaling is achieved by an elaborate network involving neurons and astrocytes. Hence, it is critical to better understand how neurons and astrocytes interact to coordinate the cellular regulation of glutamate signaling. In these studies, we used rat cortical cell cultures to examine whether neurons or releasable neuronal factors were capable of regulating system xc (-) (Sxc), a glutamate-releasing mechanism that is expressed primarily by astrocytes and has been shown to regulate synaptic transmission. We found that astrocytes cultured with neurons or exposed to neuronal-conditioned media displayed significantly higher levels of Sxc activity. Next, we demonstrated that the pituitary adenylate cyclase-activating polypeptide (PACAP) may be a neuronal factor capable of regulating astrocytes. In support, we found that PACAP expression was restricted to neurons, and that PACAP receptors were expressed in astrocytes. Interestingly, blockade of PACAP receptors in cultures comprised of astrocytes and neurons significantly decreased Sxc activity to the level observed in purified astrocytes, whereas application of PACAP to purified astrocytes increased Sxc activity to the level observed in cultures comprised of neurons and astrocytes. Collectively, these data reveal that neurons coordinate the actions of glutamate-related mechanisms expressed by astrocytes, such as Sxc, a process that likely involves PACAP. A critical gap in modeling excitatory signaling is how distinct components of the glutamate system expressed by neurons and astrocytes are coordinated. In these studies, we found that system xc (-) (Sxc), a glutamate release mechanism expressed by astrocytes, is regulated by releasable neuronal factors including PACAP. This represents a novel form of neuron-astrocyte communication, and highlights the possibility that pathological changes involving astrocytic Sxc may stem from altered neuronal activity.


Subject(s)
Astrocytes/metabolism , Glutamic Acid/metabolism , Neurons/physiology , Pituitary Adenylate Cyclase-Activating Polypeptide/physiology , Amino Acid Transport Systems, Acidic , Animals , Cystine/metabolism , Female , Neurons/metabolism , Pituitary Adenylate Cyclase-Activating Polypeptide/biosynthesis , Pregnancy , Rats , Rats, Sprague-Dawley , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/biosynthesis , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , Synaptic Transmission/physiology , Up-Regulation/genetics
20.
Neuropharmacology ; 102: 197-206, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26596556

ABSTRACT

Stress-induced reinstatement of cocaine seeking requires corticotropin releasing factor (CRF) actions in the ventral tegmental area (VTA). However the mechanisms through which CRF regulates VTA function to promote cocaine use are not fully understood. Here we examined the role of GABAergic neurotransmission in the VTA mediated by GABA-A or GABA-B receptors in the reinstatement of extinguished cocaine seeking by a stressor, uncontrollable intermittent footshock, or bilateral intra-VTA administration of CRF. Rats underwent repeated daily cocaine self-administration (1.0 mg/kg/ing; 14 × 6 h/day) and extinction and were tested for reinstatement in response to footshock (0.5 mA, 0.5" duration, average every 40 s; range 10-70 s) or intra-VTA CRF delivery (500 ng/side) following intra-VTA pretreatment with the GABA-A antagonist, bicuculline, the GABA-B antagonist, 2-hydroxysaclofen or vehicle. Intra-VTA bicuculline (1, 10 or 20 ng/side) failed to block footshock- or CRF-induced cocaine seeking at either dose tested. By contrast, 2-hydroxysaclofen (0.2 or 2 µg/side) prevented reinstatement by both footshock and intra-VTA CRF at a concentration that failed to attenuate food-reinforced lever pressing (45 mg sucrose-sweetened pellets; FR4 schedule) in a separate group of rats. These data suggest that GABA-B receptor-dependent CRF actions in the VTA mediate stress-induced cocaine seeking and that GABA-B receptor antagonists may have utility for the management of stress-induced relapse in cocaine addicts.


Subject(s)
Cocaine/administration & dosage , Corticotropin-Releasing Hormone/pharmacology , Drug-Seeking Behavior/drug effects , Extinction, Psychological/drug effects , GABA-A Receptor Antagonists/pharmacology , GABA-B Receptor Antagonists/pharmacology , Ventral Tegmental Area/drug effects , Animals , Baclofen/analogs & derivatives , Baclofen/pharmacology , Bicuculline/pharmacology , Male , Rats , Rats, Sprague-Dawley , Self Administration
SELECTION OF CITATIONS
SEARCH DETAIL
...