Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38766079

ABSTRACT

Converging findings have established that the endocannabinoid (eCB) system serves as a possible target for the development of new treatments for pain as a complement to opioid-based treatments. Here we show in male and female mice that enhancing levels of the eCB, 2-arachidonoylglycerol (2-AG), through pharmacological inhibition of its catabolic enzyme, monoacylglycerol lipase (MAGL), either systemically or in the ventral tegmental area (VTA) with JZL184, leads to a substantial attenuation of the rewarding effects of opioids in male and female mice using conditioned place preference and self-administration paradigms, without altering their analgesic properties. These effects are driven by CB1 receptors (CB1Rs) within the VTA as VTA CB1R conditional knockout, counteracts JZL184's effects. Conversely, pharmacologically enhancing the levels of the other eCB, anandamide (AEA), by inhibition of fatty acid amide hydrolase (FAAH) has no effect on opioid reward or analgesia. Using fiber photometry with fluorescent sensors for calcium and dopamine (DA), we find that enhancing 2-AG levels diminishes opioid reward-related nucleus accumbens (NAc) activity and DA neurotransmission. Together these findings reveal that 2-AG counteracts the rewarding properties of opioids and provides a potential adjunctive therapeutic strategy for opioid-related analgesic treatments.

2.
Biol Sex Differ ; 14(1): 52, 2023 08 18.
Article in English | MEDLINE | ID: mdl-37596655

ABSTRACT

BACKGROUND: Glutamate signaling within the nucleus accumbens underlies motivated behavior and is involved in psychiatric disease. Although behavioral sex differences in these processes are well-established, the neural mechanisms driving these differences are largely unexplored. In these studies, we examine potential sex differences in synaptic plasticity and excitatory transmission within the nucleus accumbens core. Further understanding of baseline sex differences in reward circuitry will shed light on potential mechanisms driving behavioral differences in motivated behavior and psychiatric disease. METHODS: Behaviorally naïve adult male and female Long-Evans rats, C57Bl/6J mice, and constitutive PKMζ knockout mice were killed and tissue containing the nucleus accumbens core was collected for ex vivo slice electrophysiology experiments. Electrophysiology recordings examined baseline sex differences in synaptic plasticity and transmission within this region and the potential role of PKMζ in long-term depression. RESULTS: Within the nucleus accumbens core, both female mice and rats exhibit higher AMPA/NMDA ratios compared to male animals. Further, female mice have a larger readily releasable pool of glutamate and lower release probability compared to male mice. No significant sex differences were detected in spontaneous excitatory postsynaptic current amplitude or frequency. Finally, the threshold for induction of long-term depression was lower for male animals than females, an effect that appears to be mediated, in part, by PKMζ. CONCLUSIONS: We conclude that there are baseline sex differences in synaptic plasticity and excitatory transmission in the nucleus accumbens core. Our data suggest there are sex differences at multiple levels in this region that should be considered in the development of pharmacotherapies to treat psychiatric illnesses such as depression and substance use disorder.


Understanding normal neural signaling within the nucleus accumbens, a key brain region involved in psychiatric disease including substance use disorder and depression, could provide insight into treatment options for these disorders. Although we know the behaviors regulated by the nucleus accumbens can differ between males and females, we do not understand the underlying differences in brain processing that could contribute to these behavioral differences. Further, even in cases when these behaviors are not different, the underlying brain signaling may exhibit sex-specific mechanisms. The current studies examined excitatory signaling with the nucleus accumbens in both rats and mice at the level of both individual cells and circuits. We found that female rodents (rats and mice) exhibit higher levels of excitatory signaling within the nucleus accumbens than male rodents. Further, procedures that can dampen neural transmission in males are not sufficient to do so in females, suggesting that excitatory signaling in the nucleus accumbens of females is less plastic. Finally, our last set of studies utilized mice missing the protein, PKMζ, and demonstrated that this reversed some of the sex differences seen in normal mice, pointing to a critical role for this protein in maintaining these differences. Our data suggest there are sex differences at multiple levels in this region that should be considered in the development of pharmacotherapies to treat psychiatric illnesses such as depression and substance use disorder.


Subject(s)
Glutamic Acid , Nucleus Accumbens , Female , Male , Mice , Rats , Animals , Rats, Long-Evans , Sex Characteristics , Excitatory Postsynaptic Potentials , Mice, Inbred C57BL , Mice, Knockout
3.
Horm Behav ; 155: 105408, 2023 09.
Article in English | MEDLINE | ID: mdl-37541099

ABSTRACT

Protein interacting with C kinase 1 (PICK1) is an AMPA receptor binding protein that works in conjunction with glutamate receptor interacting protein (GRIP) to balance the number of GluA2-containing AMPARs in the synapse. In male mice, disrupting PICK1 in the medial prefrontal cortex (mPFC) leads to a decrease in cue-induced cocaine seeking and disrupting GRIP in the mPFC has the opposing effect, consistent with other evidence that removal of GluA2-containing AMPARs potentiates reinstatement. However, PICK1 does not seem to play the same role in female mice, as knockdown of either PICK1 or GRIP in the mPFC leads to similar increases in cue-induced cocaine seeking. These previous findings indicate that the role of PICK1 in the prefrontal cortex is sex specific. The goal of the current study was to examine whether ovarian hormones contribute to the effect of prefrontal PICK1 knockdown on reinstatement of cocaine seeking. While we replicated the increased cue-induced cocaine seeking in prefrontal PICK1 knockdown sham mice, we did not see any difference between the GFP control mice and PICK1 knockdowns following ovariectomy. However, this effect was driven primarily by an increase in cocaine seeking in ovariectomized GFP control mice while there was no effect ovariectomy in PICK1 knockdown mice. Taken together, these findings suggest that circulating ovarian hormones interact with the effects of PICK1 on cue-induced reinstatement.


Subject(s)
Cocaine , Mice , Animals , Male , Female , Cocaine/pharmacology , Nucleus Accumbens/metabolism , Synapses , Prefrontal Cortex , Hormones/metabolism , Self Administration , Extinction, Psychological
4.
Biol Sex Differ ; 13(1): 66, 2022 11 08.
Article in English | MEDLINE | ID: mdl-36348414

ABSTRACT

BACKGROUND: Dysregulation in the prefrontal cortex underlies a variety of psychiatric illnesses, including substance use disorder, depression, and anxiety. Despite the established sex differences in prevalence and presentation of these illnesses, the neural mechanisms driving these differences are largely unexplored. Here, we investigate potential sex differences in glutamatergic transmission within the medial prefrontal cortex (mPFC). The goal of these experiments was to determine if there are baseline sex differences in transmission within this region that may underlie sex differences in diseases that involve dysregulation in the prefrontal cortex. METHODS: Adult male and female C57Bl/6J mice were used for all experiments. Mice were killed and bilateral tissue samples were taken from the medial prefrontal cortex for western blotting. Both synaptosomal and total GluA1 and GluA2 levels were measured. In a second set of experiments, mice were killed and ex vivo slice electrophysiology was performed on prepared tissue from the medial prefrontal cortex. Spontaneous excitatory postsynaptic currents and rectification indices were measured. RESULTS: Females exhibit higher levels of synaptosomal GluA1 and GluA2 in the mPFC compared to males. Despite similar trends, no statistically significant differences are seen in total levels of GluA1 and GluA2. Females also exhibit both a higher amplitude and higher frequency of spontaneous excitatory postsynaptic currents and greater inward rectification in the mPFC compared to males. CONCLUSIONS: Overall, we conclude that there are sex differences in glutamatergic transmission in the mPFC. Our data suggest that females have higher levels of glutamatergic transmission in this region. This provides evidence that the development of sex-specific pharmacotherapies for various psychiatric diseases is important to create more effective treatments.


Subject(s)
Glutamic Acid , Sex Characteristics , Female , Male , Mice , Animals , Prefrontal Cortex/physiology , Excitatory Postsynaptic Potentials , Mice, Inbred C57BL
5.
Addict Biol ; 27(1): e13077, 2022 01.
Article in English | MEDLINE | ID: mdl-34278652

ABSTRACT

Exposure to adversity during early childhood and adolescence increases an individual's vulnerability to developing substance use disorder. Despite the knowledge of this vulnerability, the mechanisms underlying it are still poorly understood. Excitatory afferents to the nucleus accumbens (NAc) mediate responses to both stressful and rewarding stimuli. Understanding how adolescent social isolation alters these afferents could inform the development of targeted interventions both before and after drug use. Here, we used social isolation rearing as a model of early life adversity which we have previously demonstrated increases vulnerability to cocaine addiction-like behaviour. The current study examined the effect of social isolation rearing on presynaptic glutamatergic transmission in NAc medium spiny neurons in both male and female mice. We show that social isolation rearing alters presynaptic plasticity in the NAc by decreasing the paired-pulse ratio and the size of the readily releasable pool of glutamate. Optogenetically activating the glutamatergic input from the ventral hippocampus to the NAc is sufficient to recapitulate the decreases in paired-pulse ratio and readily releasable pool size seen following electrical stimulation of all NAc afferents. Further, optogenetically inhibiting the ventral hippocampal afferent during electrical stimulation eliminates the effect of early life adversity on the paired-pulse ratio or readily releasable pool size. In summary, we demonstrate that social isolation rearing leads to alterations in glutamate transmission driven by projections from the ventral hippocampus. These data suggest that targeting the circuit from the ventral hippocampus to the nucleus accumbens could provide a means to reverse stress-induced plasticity.


Subject(s)
Glutamic Acid/metabolism , Nucleus Accumbens/metabolism , Social Isolation , Animals , Female , Hippocampus/metabolism , Male , Mice , Optogenetics , Receptors, Dopamine D1 , Synaptic Transmission
6.
Brain Res ; 1777: 147755, 2022 02 15.
Article in English | MEDLINE | ID: mdl-34932973

ABSTRACT

Post-weaning social isolation stress has been shown to increase addiction-like behavior in adulthood. These long-term behavioral alterations may be due to long lasting isolation-induced structural changes to neurons in brain regions involved in reward processing. Previous studies have shown that various stressors alter dendritic spine density in the prefrontal cortex (PFC) and the nucleus accumbens, though many of these studies examine the short-term effects of stress, and are primarily conducted in males. There is mounting evidence that males and females exhibit differences in their stress responses, with some studies showing sex differences in stress-induced plasticity. To determine the long-lasting, sex-specific alterations in spine density following post-weaning social isolation, male and female mice were either isolated or group housed at weaning and spine density was measured once they reached adulthood. Post-weaning isolation increased spine density in the PFC of both the males and females, although the effects in the infralimbic cortex were more pronounced in the females. In the nucleus accumbens, adolescent isolation increased spine density in males only in the core and shell. Females also had higher baseline spine density than males in the nucleus accumbens core. Together these data suggest that adolescent social isolation causes long-term, sex-specific alterations to the prefrontal cortex and the nucleus accumbens.


Subject(s)
Dendritic Spines/physiology , Nucleus Accumbens/physiology , Prefrontal Cortex/physiology , Social Isolation , Stress, Psychological , Weaning , Animals , Animals, Newborn , Brain/physiology , Cerebral Cortex/physiology , Female , Hippocampus/physiology , Male , Mice , Neurons , Reward , Sex Characteristics
7.
Neurosci Biobehav Rev ; 128: 358-366, 2021 09.
Article in English | MEDLINE | ID: mdl-34214512

ABSTRACT

Females are more vulnerable than males to many aspects of cocaine use disorder. This vulnerability also translates to opioid use disorder, with females exhibiting stronger behavioral responses than males to drugs such as heroin and morphine. While there is evidence for many overlapping neural mechanisms underlying cocaine and opioid abuse, there is also a breadth of evidence indicating divergent effects of the drugs on synaptic plasticity. This makes it unclear whether the behavioral sex differences seen in substance use disorder across different drugs of abuse rely on the same mechanisms. Ovarian hormones have consistently been implicated as drivers of the behavioral sex differences in cocaine taking and seeking. While there are far fewer studies on the role of ovarian hormones in opioid use disorder, the existing data suggest that ovarian hormones may not drive these behavioral effects in the same manner as in cocaine use disorder. This review highlights evidence that behavioral sex differences in substance use disorder might be driven by different mechanisms depending on drug class.


Subject(s)
Cocaine , Opioid-Related Disorders , Substance-Related Disorders , Female , Gonadal Hormones , Hormones , Humans , Male , Sex Characteristics
8.
Addict Biol ; 26(5): e13051, 2021 09.
Article in English | MEDLINE | ID: mdl-34110073

ABSTRACT

Disruption of prefrontal glutamate receptor interacting protein (GRIP), which anchors GluA2-containing AMPA receptors (AMPARs) into the synaptic membrane, potentiates cue-induced cocaine seeking in both males and females. Protein interacting with C kinase 1 (PICK1) plays an opposing role to that of GRIP, removing AMPARs from the synapse. Consistent with our hypothesis that disruption of PICK1 in the mPFC would lead to a decrease in addiction-like behaviour, we found that conditional deletion of PICK1 in the mPFC attenuates cue-induced cocaine seeking in male mice. However, prefrontal PICK1 deletion had the opposite effect in females, leading to an increase in cue-induced reinstatement of cocaine seeking. We did not see any effects of PICK1 knockdown on sucrose taking or seeking, suggesting the sex-specific effects do not generalise to natural reinforcers. These findings suggest the role of PICK1 in the prefrontal cortex of females may not be consistent with its accepted role in males. To determine whether these sex differences were influenced by gonadal hormones, we gonadectomised a cohort of males and found that removal of circulating androgens eliminated the effect of prefrontal PICK1 knockdown. As there was no effect of gonadectomy on its own on any of the behavioural measures collected, our results suggest that androgens may be involved in compensatory downstream effects of PICK1 knockdown. Taken together, these results highlight the need for consideration of sex as a biological variable when examining mechanisms underlying all behaviours, as convergent sex differences can reveal different mechanisms where behavioural sex differences do not exist.


Subject(s)
Cell Cycle Proteins/metabolism , Cocaine/pharmacology , Drug-Seeking Behavior/drug effects , Prefrontal Cortex/metabolism , Animals , Cocaine-Related Disorders , Conditioning, Operant , Female , Glutamic Acid/metabolism , Male , Mice , Nucleus Accumbens/drug effects , Receptors, AMPA/metabolism , Self Administration , Sex Characteristics , Sucrose/administration & dosage , Synapses/metabolism
9.
Proc Natl Acad Sci U S A ; 118(8)2021 02 23.
Article in English | MEDLINE | ID: mdl-33593913

ABSTRACT

Experiencing some early life adversity can have an "inoculating" effect that promotes resilience in adulthood. However, the mechanisms underlying stress inoculation are unknown, and animal models are lacking. Here we used the limited bedding and nesting (LBN) model of adversity to evaluate stress inoculation of addiction-related phenotypes. In LBN, pups from postnatal days 2 to 9 and their dams were exposed to a low-resource environment. In adulthood, they were tested for addiction-like phenotypes and compared to rats raised in standard housing conditions. High levels of impulsivity are associated with substance abuse, but in males, LBN reduced impulsive choice compared to controls. LBN males also self-administered less morphine and had a lower breakpoint on a progressive ratio reinforcement schedule than controls. These effects of LBN on addiction-related behaviors were not found in females. Because the nucleus accumbens (NAc) mediates these behaviors, we tested whether LBN altered NAc physiology in drug-naïve and morphine-exposed rats. LBN reduced the frequency of spontaneous excitatory postsynaptic currents in males, but a similar effect was not observed in females. Only in males did LBN prevent a morphine-induced increase in the AMPA/NMDA ratio. RNA sequencing was performed to delineate the molecular signature in the NAc associated with LBN-derived phenotypes. LBN produced sex-specific changes in transcription, including in genes related to glutamate transmission. Collectively, these studies reveal that LBN causes a male-specific stress inoculation effect against addiction-related phenotypes. Identifying factors that promote resilience to addiction may reveal novel treatment options for patients.


Subject(s)
Behavior, Animal , Nucleus Accumbens/physiopathology , Opioid-Related Disorders/prevention & control , Resilience, Psychological , Stress, Psychological , Transcriptome , Animals , Animals, Newborn , Female , Gene Expression Regulation , Male , Nucleus Accumbens/drug effects , Opioid-Related Disorders/genetics , Opioid-Related Disorders/metabolism , Phenotype , Rats , Rats, Long-Evans , Receptors, AMPA/genetics , Receptors, AMPA/metabolism , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Sex Factors
10.
Cell Stem Cell ; 26(5): 766-781.e9, 2020 05 07.
Article in English | MEDLINE | ID: mdl-32142682

ABSTRACT

Human brain organoids provide unique platforms for modeling development and diseases by recapitulating the architecture of the embryonic brain. However, current organoid methods are limited by interior hypoxia and cell death due to insufficient surface diffusion, preventing generation of architecture resembling late developmental stages. Here, we report the sliced neocortical organoid (SNO) system, which bypasses the diffusion limit to prevent cell death over long-term cultures. This method leads to sustained neurogenesis and formation of an expanded cortical plate that establishes distinct upper and deep cortical layers for neurons and astrocytes, resembling the third trimester embryonic human neocortex. Using the SNO system, we further identify a critical role of WNT/ß-catenin signaling in regulating human cortical neuron subtype fate specification, which is disrupted by a psychiatric-disorder-associated genetic mutation in patient induced pluripotent stem cell (iPSC)-derived SNOs. These results demonstrate the utility of SNOs for investigating previously inaccessible human-specific, late-stage cortical development and disease-relevant mechanisms.


Subject(s)
Induced Pluripotent Stem Cells , Neocortex , Humans , Neurogenesis , Neurons , Organoids
11.
Neurosci Biobehav Rev ; 107: 360-369, 2019 12.
Article in English | MEDLINE | ID: mdl-31550452

ABSTRACT

Stress is an important risk factor for the development of substance use disorder (SUD). Exposure to both stress and drugs abuse lead to changes in synaptic plasticity and stress-induced alterations in synaptic plasticity may contribute to later vulnerability to SUD. Recent developmental neuroscience studies have identified microglia as regulators of synaptic plasticity. As both stress and drugs of abuse lead to microglial activation, we propose this as a potential mechanism underlying their ability to change synaptic plasticity. This review focuses on three components of synaptic plasticity: spine density, brain-derived neurotrophic factor (BDNF) and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor expression. Their roles in addiction, stress, and development will be reviewed, as well as possible mechanisms by which microglia could regulate their function. Potential links between stress, vulnerability to addiction, and microglial activity will be explored.


Subject(s)
Microglia/metabolism , Neuronal Plasticity/physiology , Nucleus Accumbens/physiology , Substance-Related Disorders/physiopathology , Animals , Cocaine/pharmacology , Glutamic Acid/metabolism , Humans , Microglia/drug effects , Neuronal Plasticity/drug effects , Nucleus Accumbens/drug effects
12.
J Neurosci ; 39(39): 7801-7809, 2019 09 25.
Article in English | MEDLINE | ID: mdl-31409665

ABSTRACT

Cocaine-induced plasticity persists during abstinence and is thought to underlie cue-evoked craving. Reversing this plasticity could provide an opportunity for therapeutic intervention. Converging evidence suggest that zeta inhibitory peptide (ZIP) eliminates memories for experience-dependent behaviors, including conditioned drug associations. However, the effect of ZIP on reward seeking and drug-induced plasticity is unknown. The current study examined the effect of ZIP administration in the nucleus accumbens on reinstatement (RI) of cocaine seeking, a rodent model of relapse. We demonstrate that intra-accumbal ZIP administration blocks cocaine-primed RI in rats when administered 24 h or 1 week before testing. These effects of ZIP on drug seeking are specific, as we did not see any effect of ZIP on RI of sucrose seeking. ZIP is a synthetic compound designed to inhibit the atypical PKC, PKMζ, a protein implicated in learning and memory. However, recent evidence from PKMζ-knock-out (KO) mice suggests that ZIP may function through alternative mechanisms. In support of this, we found that ZIP was able to block cue-induced RI in PKMζ-KO mice. One possible mechanism underlying addictive phenotypes is the ability of cocaine to block further plasticity. We hypothesized that ZIP may be working to reverse this anaplasticity. Although ZIP has no effect on accumbal LTD in slices from naive or yoked saline mice, it is able to restore both NMDA-dependent and mGluR5-dependent LTD in animals after cocaine self-administration and withdrawal. These findings demonstrate that intra-accumbal ZIP persistently reverses cocaine-induced behavioral and synaptic plasticity in male and female rodents.SIGNIFICANCE STATEMENT Zeta-inhibitory peptide (ZIP) has been shown to disrupt memory maintenance for experience-dependent behaviors. We examined the effect of ZIP infused into the nucleus accumbens on the reinstatement (RI) of cocaine seeking. We found that intra-accumbal ZIP blocked RI of cocaine seeking 24 h and 1 week later. This effect was specific to RI of cocaine seeking as ZIP did not disrupt RI of food seeking. In conjunction with these behavioral studies we examined the ability of ZIP to reverse cocaine-induced deficits in LTD. We found that ZIP was able to rescue two forms of LTD in cocaine-experienced mice. These studies demonstrate that ZIP is able to reverse cocaine-induced behavioral and synaptic plasticity in a persistent manner.


Subject(s)
Cell-Penetrating Peptides/pharmacology , Cocaine/pharmacology , Drug-Seeking Behavior/physiology , Lipopeptides/pharmacology , Long-Term Synaptic Depression/drug effects , Nucleus Accumbens/drug effects , Protein Kinase Inhibitors/pharmacology , Animals , Cocaine-Related Disorders/physiopathology , Extinction, Psychological/drug effects , Long-Term Synaptic Depression/physiology , Male , Mice , Mice, Inbred C57BL , Nucleus Accumbens/physiology , Peptides/pharmacology , Rats , Rats, Sprague-Dawley
13.
Neuropharmacology ; 157: 107672, 2019 10.
Article in English | MEDLINE | ID: mdl-31233823

ABSTRACT

Glutamate receptor interacting protein (GRIP) is a neuronal scaffolding protein that anchors GluA2-containing AMPA receptors to the cell membrane. GRIP plays a critical role in activity-dependent synaptic plasticity, including that which occurs after drug exposure. Given that cocaine administration alters glutamate receptor trafficking within the prefrontal cortex (PFC), a better understanding of the role of receptor trafficking proteins could lead to a more complete understanding of addictive phenotypes. AMPA receptor trafficking in general, and GRIP specifically, is known to play a role in cocaine seeking and conditioned reward in the nucleus accumbens, but its role in the PFC has not been characterized. The current study demonstrates that conditional deletion of GRIP1 in the medial prefrontal cortex increases the motivation for cocaine and potentiates cue-induced reinstatement of cocaine seeking in male and female mice. As no effects of PFC GRIP1 deletion were seen in reinstatement of food seeking, strategy set-shifting, or reversal learning the effects on cocaine seeking are not related to generalized alterations in cognitive function. While disrupting GRIP1 might be expected to lead to decreased AMPA transmission, our electrophysiological data indicate an increase in sEPSC amplitude in the prefrontal cortex and a corresponding decrease in paired pulse facilitation in the nucleus accumbens. Taken together this suggests a strengthening of the PFC to NAc input following prefrontal GRIP1 deletion that may mediate the enhanced drug seeking behavior.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Drug-Seeking Behavior/physiology , Excitatory Postsynaptic Potentials/physiology , Nerve Tissue Proteins/physiology , Prefrontal Cortex/physiology , Reversal Learning/physiology , Adaptor Proteins, Signal Transducing/biosynthesis , Adaptor Proteins, Signal Transducing/genetics , Animals , Cocaine/pharmacology , Conditioning, Operant/drug effects , Cues , Extinction, Psychological/physiology , Female , Male , Mice , Mice, Knockout , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Nucleus Accumbens/physiology , Reinforcement Schedule , Self Administration , Sucrose/pharmacology
14.
Behav Brain Res ; 359: 589-596, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30296530

ABSTRACT

Childhood and adolescent adversity are associated with a wide range of psychiatric disorders, including an increased risk for substance abuse. Despite this, the mechanisms underlying the ability of chronic stress during adolescence to alter reward signaling remains largely unexplored. Understanding how adolescent stress increases addiction-like phenotypes could inform the development of targeted interventions both before and after drug use. The current study examined how prolonged isolation stress, beginning during adolescence, affected behavioral and neuronal underpinnings to the response to cocaine in male and female mice. Adolescent-onset social isolation did not alter the ability of mice to learn an operant response for food, nor influence food self-administration or motivation for food on a progressive ratio schedule. However, male and female social isolation mice exhibited an increase in motivation for cocaine and cocaine seeking during a cue-induced reinstatement session. Additionally, we demonstrated that adolescent-onset social isolation increased cocaine-induced neuronal activation, as assessed by c-Fos expression, within the nucleus accumbens core and shell, ventral pallidum, dorsal bed nucleus of the stria terminalis, lateral septum and basolateral amygdala. Taken together, the present studies demonstrate that social isolation stress during adolescence augments the behavioral responses to cocaine during adulthood and alters the responsiveness of reward-related brain circuitry.


Subject(s)
Brain/growth & development , Cocaine-Related Disorders/psychology , Drug-Seeking Behavior , Social Isolation , Animals , Behavior, Addictive/metabolism , Behavior, Addictive/psychology , Brain/drug effects , Brain/metabolism , Cocaine/administration & dosage , Cocaine-Related Disorders/metabolism , Conditioning, Operant/physiology , Dopamine Uptake Inhibitors/administration & dosage , Drug-Seeking Behavior/physiology , Eating/physiology , Eating/psychology , Female , Male , Mice, Inbred C57BL , Motivation/physiology , Proto-Oncogene Proteins c-fos/metabolism , Random Allocation , Self Administration , Sexual Maturation
15.
Neuropsychopharmacology ; 43(12): 2390-2398, 2018 11.
Article in English | MEDLINE | ID: mdl-30111812

ABSTRACT

The constitutively active, atypical protein kinase C, protein kinase M-ζ (PKMζ), is exclusively expressed in the brain and its expression increases following exposure to drugs of abuse. However, the limitations of currently available tools have made it difficult to examine the role of PKMζ in cocaine self-administration and relapse. The current study demonstrates that constitutive deletion of PKMζ potentiates cue-induced reinstatement of cocaine seeking and increases both food and cocaine self-administration, without affecting cue-driven food seeking in both male and female mice. Conditional deletion of PKMζ within the nucleus accumbens recapitulated the increase in cocaine taking and seeking seen in the constitutive knockout mice, but only in male animals. Site-specific knockdown of PKMζ in the nucleus accumbens had no effect on cocaine taking or seeking in female mice. Additionally, neither male nor female mice exhibited any alterations in food self-administration or cue-induced reinstatement of food seeking following accumbal deletion of PKMζ. Taken together these results indicate that PKMζ may act to dampen cocaine taking and seeking. Furthermore, these results indicate that PKMζ is playing divergent roles in reward seeking in males and females.


Subject(s)
Cocaine/administration & dosage , Gene Deletion , Nucleus Accumbens/drug effects , Nucleus Accumbens/enzymology , Protein Kinase C/deficiency , Animals , Dopamine Uptake Inhibitors/administration & dosage , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Kinase C/genetics
16.
Front Mol Neurosci ; 11: 197, 2018.
Article in English | MEDLINE | ID: mdl-29922129

ABSTRACT

Alterations in glutamate, the primary excitatory neurotransmitter in the brain, are implicated in several psychiatric diseases. Many of these psychiatric diseases display epidemiological sex differences, with either males or females exhibiting different symptoms or disease prevalence. However, little work has considered the interaction of disrupted glutamatergic transmission and sex on disease states. This review describes the clinical and preclinical evidence for these sex differences with a focus on two conditions that are more prevalent in women: Alzheimer's disease and major depressive disorder, and three conditions that are more prevalent in men: schizophrenia, autism spectrum disorder, and attention deficit hyperactivity disorder. These studies reveal sex differences at multiple levels in the glutamate system including metabolic markers, receptor levels, genetic interactions, and therapeutic responses to glutamatergic drugs. Our survey of the current literature revealed a considerable need for more evaluations of sex differences in future studies examining the role of the glutamate system in psychiatric disease. Gaining a more thorough understanding of how sex differences in the glutamate system contribute to psychiatric disease could provide novel avenues for the development of sex-specific pharmacotherapies.

17.
Behav Brain Res ; 333: 83-89, 2017 08 30.
Article in English | MEDLINE | ID: mdl-28668281

ABSTRACT

Cocaine addiction is characterized by persistent craving and addicts frequently relapse even after long periods of abstinence. Exposure to stress can precipitate relapse in humans and rodents. Stress and drug use can lead to common alterations in synaptic plasticity and these commonalities may contribute to the ability of stress to elicit relapse. These common changes in synaptic plasticity are mediated, in part, by alterations in the trafficking and stabilization of AMPA receptors. Exposure to both cocaine and stress can lead to alterations in protein kinase C-mediated phosphorylation of GluA2 AMPA subunits and thus alter the trafficking of GluA2-containing AMPARs. However, it is not clear what role AMPAR trafficking plays in the interactions between stress and cocaine. The current study utilized a mouse with a point mutation within the GluA2 subunit c-terminus resulting in a disruption of PKC-mediated GluA2 phosphorylation to examine stress responsivity. Although no differences were seen in the response to a forced swim stress in naïve mice, GluA2 K882A knock-in mice exhibited an increased stress response following cocaine self-administration. Furthermore, we demonstrated that disrupting GluA2 phosphorylation increases vulnerability to stress-induced reinstatement of both cocaine seeking and cocaine-conditioned reward. Finally, GluA2 K882A knock-in mice exhibit an increased vulnerability to social defeat as indicated by increased social avoidance. Taken together these results indicate that disrupting GluA2 phosphorylation leads to increased responsivity to acute stress following cocaine exposure and increased vulnerability to chronic stress. These results highlight the GluA2 phosphorylation site as a novel target for the stress-related disorders.


Subject(s)
Receptors, AMPA/metabolism , Stress, Psychological/metabolism , Alanine/genetics , Animals , Cocaine/toxicity , Conditioning, Operant/drug effects , Conditioning, Operant/physiology , Disease Models, Animal , Dopamine Uptake Inhibitors/toxicity , Drug-Seeking Behavior/drug effects , Drug-Seeking Behavior/physiology , Extinction, Psychological/drug effects , Extinction, Psychological/physiology , Female , Gene Expression Regulation/genetics , Gene Expression Regulation/physiology , Lysine/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Phosphorylation/physiology , Protein Kinase C/metabolism , Receptors, AMPA/genetics , Swimming/psychology
18.
Am J Pathol ; 187(1): 91-109, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27993242

ABSTRACT

Mounting evidence implicates antiretroviral (ARV) drugs as potential contributors to the persistence and evolution of clinical and pathological presentation of HIV-associated neurocognitive disorders in the post-ARV era. Based on their ability to induce endoplasmic reticulum (ER) stress in various cell types, we hypothesized that ARV-mediated ER stress in the central nervous system resulted in chronic dysregulation of the unfolded protein response and altered amyloid precursor protein (APP) processing. We used in vitro and in vivo models to show that HIV protease inhibitor (PI) class ARVs induced neuronal damage and ER stress, leading to PKR-like ER kinase-dependent phosphorylation of the eukaryotic translation initiation factor 2α and enhanced translation of ß-site APP cleaving enzyme-1 (BACE1). In addition, PIs induced ß-amyloid production, indicative of increased BACE1-mediated APP processing, in rodent neuroglial cultures and human APP-expressing Chinese hamster ovary cells. Inhibition of BACE1 activity protected against neuronal damage. Finally, ARVs administered to mice and SIV-infected macaques resulted in neuronal damage and BACE1 up-regulation in the central nervous system. These findings implicate a subset of PIs as potential mediators of neurodegeneration in HIV-associated neurocognitive disorders.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/metabolism , HIV Protease Inhibitors/pharmacology , Protein Biosynthesis/drug effects , Protein Processing, Post-Translational/drug effects , Up-Regulation/drug effects , Animals , Axons/drug effects , Axons/metabolism , Axons/pathology , Cells, Cultured , Macaca , Male , Mice, Inbred C57BL , Microtubule-Associated Proteins/metabolism , Neuroglia/drug effects , Neuroglia/metabolism , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Protein Stability/drug effects , Rats , Ritonavir/pharmacology , Unfolded Protein Response/drug effects , eIF-2 Kinase/metabolism
19.
Neuropharmacology ; 111: 231-241, 2016 12.
Article in English | MEDLINE | ID: mdl-27622930

ABSTRACT

Addiction is associated with changes in synaptic plasticity mediated, in part, by alterations in the trafficking and stabilization of AMPA receptors at synapses within the nucleus accumbens. Exposure to cocaine can lead to protein kinase C-mediated phosphorylation of GluA2 AMPA subunits and this phosphorylation event leads to the internalization of GluA2-containing AMPARs, which are calcium-impermeable. However, it is not clear whether this internalization is necessary for the expression of addictive phenotypes. Utilizing a mouse with a point mutation within the GluA2 subunit c-terminus, the current study demonstrates that disrupting PKC-mediated GluA2 phosphorylation potentiates reinstatement of both cue-induced cocaine seeking and cocaine conditioned reward without affecting operant learning, food self-administration or cocaine sensitization. Electrophysiological recordings revealed increased GluA2-mediated AMPA transmission as evidenced by increased sEPSC amplitude without any changes in sEPSC frequency or rectification. In support of this increase in GluA2 activity mediating the augmented cocaine reinstatement, we found that accumbal overexpression of GluA2 recapitulated this behavioral effect in wildtype mice while not altering reinstatement behavior in the GluA2 K882A knock-in mice. In addition, disrupting GluA2 phosphorylation was associated with blunted long-term depression in the nucleus accumbens, mimicking the anaplasticity seen following cocaine self-administration. Taken together these results indicate that disrupting GluA2 phosphorylation and increasing GluA2-mediated transmission in the nucleus accumbens leads to increased vulnerability to cocaine relapse. Further, these results indicate that modulating GluA2-containing AMPAR trafficking can contribute to addictive phenotypes in the absence of alterations in GluA2-lacking receptors. These results highlight the GluA2 phosphorylation site as a novel target for the development of cocaine addiction therapeutics.


Subject(s)
Cocaine/metabolism , Drug-Seeking Behavior/physiology , Nucleus Accumbens/drug effects , Nucleus Accumbens/physiology , Receptors, AMPA/metabolism , Animals , Conditioning, Operant/drug effects , Excitatory Postsynaptic Potentials , Gene Knock-In Techniques , Long-Term Synaptic Depression , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/drug effects , Nucleus Accumbens/metabolism , Phosphorylation , Protein Kinase C/metabolism , Receptors, AMPA/genetics , Receptors, AMPA/physiology
20.
Physiol Behav ; 166: 32-42, 2016 Nov 01.
Article in English | MEDLINE | ID: mdl-26907955

ABSTRACT

Drug addiction is a major public health concern in the United States costing taxpayers billions in health care costs, lost productivity and law enforcement. However, the availability of effective treatment options remains limited. The development of novel therapeutics will not be possible without a better understanding of the addicted brain. Studies in both clinical and preclinical models indicate that chronic drug use leads to alterations in the body and brain's response to stress. Dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis may shed light on the ability of stress to increase vulnerability to relapse. Further, within both the HPA axis and limbic brain regions, corticotropin-releasing factor (CRF) is critically involved in the brain's response to stress. Alterations in both central and peripheral CRF activity seen following chronic drug use provide a mechanism by which substance use can alter stress reactivity, thus mediating addictive phenotypes. While many reviews have focused on how stress alters drug-mediated changes in physiology and behavior, the goal of this review is to focus on how substance use alters responses to stress.


Subject(s)
Hypothalamo-Hypophyseal System/physiopathology , Pituitary-Adrenal System/physiopathology , Stress, Psychological/etiology , Substance-Related Disorders/physiopathology , Substance-Related Disorders/psychology , Animals , Corticotropin-Releasing Hormone/metabolism , Humans , Hypothalamo-Hypophyseal System/metabolism , Pituitary-Adrenal System/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...