71 results on '"Kaplan GB"'
Search Results
2. Pharmacological Management of Sleep-Wake Disturbances in Delirium.
- Author
-
Levinsohn EA, Radhakrishnan V, Euting H, and Kaplan GB
- Abstract
Delirium is a heterogeneous syndrome primarily characterized by fluctuations in attention and awareness. Sleep-wake disturbances are a common and significant feature of delirium and can manifest as circadian rhythm inversion, sleep fragmentation, and reduced rapid eye movement (REM) and slow-wave sleep. Some literature suggests that the relationship between sleep disruption and delirium is reciprocal wherein the two reinforce one another and may share an underlying etiology. As there are no FDA-approved medications for delirium or delirium-related sleep disturbances, management is primarily focused on addressing underlying medical concerns and promoting physiologic circadian patterns with non-pharmacological behavioral interventions. In practice, however, medications are often used, albeit with limited evidence to support their use. This literature review explores the pharmacology and pharmacokinetics of several medications with literature investigating their use in delirium: melatonin, ramelteon, dual orexin receptor antagonists (DORAs), and dexmedetomidine. Current evidence suggests a possible benefit of ramelteon or melatonin, dexmedetomidine for patients in the ICU setting, and DORAs as therapeutic options for the re-regulation of sleep-wake cycle disruption in delirium. We discuss pertinent pharmacokinetic and pharmacodynamic factors that may influence clinical decision-making regarding these interventions., (Published 2024. This article is a U.S. Government work and is in the public domain in the USA.)
- Published
- 2024
- Full Text
- View/download PDF
3. Neuroplasticity of the extended amygdala in opioid withdrawal and prolonged opioid abstinence.
- Author
-
Kaplan GB and Thompson BL
- Abstract
Opioid use disorder is characterized by excessive use of opioids, inability to control its use, a withdrawal syndrome upon discontinuation of opioids, and long-term likelihood of relapse. The behavioral stages of opioid addiction correspond with affective experiences that characterize the opponent process view of motivation. In this framework, active involvement is accompanied by positive affective experiences which gives rise to "reward craving," whereas the opponent process, abstinence, is associated with the negative affective experiences that produce "relief craving." Relief craving develops along with a hypersensitization to the negatively reinforcing aspects of withdrawal during abstinence from opioids. These negative affective experiences are hypothesized to stem from neuroadaptations to a network of affective processing called the "extended amygdala." This negative valence network includes the three core structures of the central nucleus of the amygdala (CeA), the bed nucleus of the stria terminalis (BNST), and the nucleus accumbens shell (NAc shell), in addition to major inputs from the basolateral amygdala (BLA). To better understand the major components of this system, we have reviewed their functions, inputs and outputs, along with the associated neural plasticity in animal models of opioid withdrawal. These models demonstrate the somatic, motivational, affective, and learning related models of opioid withdrawal and abstinence. Neuroadaptations in these stress and motivational systems are accompanied by negative affective and aversive experiences that commonly give rise to relapse. CeA neuroplasticity accounts for many of the aversive and fear-related effects of opioid withdrawal via glutamatergic plasticity and changes to corticotrophin-releasing factor (CRF)-containing neurons. Neuroadaptations in BNST pre-and post-synaptic GABA-containing neurons, as well as their noradrenergic modulation, may be responsible for a variety of aversive affective experiences and maladaptive behaviors. Opioid withdrawal yields a hypodopaminergic and amotivational state and results in neuroadaptive increases in excitability of the NAc shell, both of which are associated with increased vulnerability to relapse. Finally, BLA transmission to hippocampal and cortical regions impacts the perception of conditioned aversive effects of opioid withdrawal by higher executive systems. The prevention or reversal of these varied neuroadaptations in the extended amygdala during opioid withdrawal could lead to promising new interventions for this life-threatening condition., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Kaplan and Thompson.)
- Published
- 2023
- Full Text
- View/download PDF
4. Estrous cycle dependent expression of oxycodone conditioned reward in rats.
- Author
-
Babb JA, Constantino NJ, Kaplan GB, and Chartoff EH
- Subjects
- Adult, Humans, Rats, Female, Male, Animals, Rats, Sprague-Dawley, Estrous Cycle, Reward, Oxycodone pharmacology, Analgesics, Opioid pharmacology
- Abstract
Oxycodone is one of the most widely prescribed and misused opioid painkillers in the United States. Evidence suggests that biological sex and hormonal status can impact drug reward in humans and rodents, but the extent to which these factors can influence the rewarding effects of oxycodone is unclear. The purpose of this study was to utilize place conditioning to determine the effects of sex and female hormonal status on the expression of oxycodone conditioned reward in rats. Gonadally intact adult Sprague-Dawley male and female rats were used to test: (1) whether both sexes express conditioned reward to oxycodone at similar doses, (2) the impact of conditioning session length on oxycodone conditioned reward expression in both sexes, and (3) the influence of female estrous cycle stage on oxycodone conditioned reward expression. Both sexes expressed conditioned reward at the same doses of oxycodone. Increasing the length of conditioning sessions did not reveal an effect of sex and resulted in lower magnitude conditioned reward expression. Importantly however, female stage of estrous cycle significantly influenced oxycodone conditioned reward expression. These results suggest that female hormonal status can impact the rewarding effects of opioids and thus have important implications for prescription opioid treatment practices., (© 2023. Springer Nature Limited.)
- Published
- 2023
- Full Text
- View/download PDF
5. Disturbances in fear extinction learning after mild traumatic brain injury in mice are accompanied by alterations in dendritic plasticity in the medial prefrontal cortex and basolateral nucleus of the amygdala.
- Author
-
Babb JA, Zuberer A, Heinrichs S, Rumbika KK, Alfiler L, Lakis GA, Leite-Morris KA, and Kaplan GB
- Subjects
- Mice, Male, Animals, Extinction, Psychological, Fear physiology, Mice, Inbred C57BL, Amygdala physiology, Prefrontal Cortex, Brain Concussion
- Abstract
Mild traumatic brain injury (mTBI) and post-traumatic stress disorder (PTSD) have emerged as the signature injuries of the U.S. veterans who served in Iraq and Afghanistan, and frequently co-occur in both military and civilian populations. To better understand how fear learning and underlying neural systems might be altered after mTBI, we examined the acquisition of cued fear conditioning and its extinction along with brain morphology and dendritic plasticity in a mouse model of mTBI. To induce mTBI in adult male C57BL/6J mice, a lateral fluid percussive injury (LFP 1.7) was produced using a fluid pulse of 1.7 atmosphere force to the right parietal lobe. Behavior in LFP 1.7 mice was compared to behavior in mice from two separate control groups: mice subjected to craniotomy without LFP injury (Sham) and mice that did not undergo surgery (Unoperated). Following behavioral testing, neural endpoints (dendritic structural plasticity and neuronal volume) were assessed in the basolateral nucleus of the amygdala (BLA), which plays a critical sensory role in fear learning, and medial prefrontal cortex (mPFC), responsible for executive functions and inhibition of fear behaviors. No gross motor abnormalities or increased anxiety-like behaviors were observed in LFP or Sham mice after surgery compared to Unoperated mice. We found that all mice acquired fear behavior, assessed as conditioned freezing to auditory cue in a single session of 6 trials, and acquisition was similar across treatment groups. Using a linear mixed effects analysis, we showed that fear behavior decreased overall over 6 days of extinction training with no effect of treatment group across extinction days. However, a significant interaction was demonstrated between the treatment groups during within-session freezing behavior (5 trials per day) during extinction training. Specifically, freezing behavior increased across within-session extinction trials in LFP 1.7 mice, whereas freezing behavior in control groups did not change on extinction test days, reflecting a dissociation between within-trial and between-trial fear extinction. Additionally, LFP mice demonstrated bilateral increases in dendritic spine density in the BLA and decreases in dendritic complexity in the PFC. The translational implications are that individuals with TBI undergoing fear extinction therapy may demonstrate within-session aberrant learning that could be targeted for more effective treatment interventions., (Published by Elsevier Inc.)
- Published
- 2023
- Full Text
- View/download PDF
6. Mitochondrial dysfunction in animal models of PTSD: Relationships between behavioral models, neural regions, and cellular maladaptation.
- Author
-
Kaplan GB, Dadhi NA, and Whitaker CS
- Abstract
Post-traumatic stress disorder (PTSD) is a trauma-related condition that produces distressing fear memory intrusions, avoidance behaviors, hyperarousal, stress responses, insomnia and other symptoms. This review of rodent models of PTSD examines trauma effects on fear-related learning, cognition, and avoidance, emotional and arousal behaviors and on mitochondrial dysfunction in relevant neural pathways. The review focuses on research that includes four elements: consensus PTSD rodent models, behavioral phenotyping, mitochondrial dysfunction within key neural regions. This approach allows for the integration of behavioral, neural and cellular findings in PTSD models. The PTSD models reviewed include fear conditioning, predator/social stress, chronic restraint stress, single prolonged stress, social isolation, chronic unpredictable stress and early life stress. These models produce a variety of PTSD-related behaviors that include associative and non-associative fear- and stress-related responses, hyperarousal, avoidance behaviors, cognitive disturbances, social withdrawal, compulsive behaviors, anhedonia-, anxiety- and depression-related behaviors. Neural regions included fear- and stress-related regions of the prefrontal cortex, hippocampal, amygdala, nucleus accumbens and hypothalamus. PTSD models produced mitochondrial dysfunction that includes dysregulation of oxidative phosphorylation and other metabolic pathways including β-oxidation of fatty acids and the tricarboxylic acid pathway. These models generated neural reactive oxygen species that damage DNA, proteins, and lipids. Trauma models further altered mitochondrial structure and replication and affected neuroinflammatory responses, signal transduction and apoptosis. Antidepressant medications used for the treatment of PTSD reversed stress-induced changes in some PTSD-like behaviors and many elements of brain mitochondrial dysfunction. Future studies can develop PTSD models which are ecologically valid and result in a broader manifestation of PTSD-related behaviors as it is clinically defined. This review highlights mitochondrial mechanisms associated with PTSD-like behaviors that have been produced in an array of consensus PTSD models and identifies putative circuit-based targets for more effective treatment for this debilitating disorder., (Copyright © 2023 Kaplan, Dadhi and Whitaker.)
- Published
- 2023
- Full Text
- View/download PDF
7. Introduction to special issue: Sleep and arousal in health and disease.
- Author
-
Brown RE and Kaplan GB
- Subjects
- Arousal, Sleep
- Published
- 2022
- Full Text
- View/download PDF
8. Sleep-wake and arousal dysfunctions in post-traumatic stress disorder: Role of orexin systems.
- Author
-
Kaplan GB, Lakis GA, and Zhoba H
- Subjects
- Animals, Arousal, Extinction, Psychological physiology, Fear physiology, Hypothalamo-Hypophyseal System metabolism, Mice, Orexin Receptors metabolism, Orexins metabolism, Pituitary-Adrenal System metabolism, Sleep physiology, Stress Disorders, Post-Traumatic
- Abstract
Post-traumatic stress disorder (PTSD) is a trauma-related condition that produces distressing fear memory intrusions, avoidance behaviors, hyperarousal/startle, stress responses and insomnia. This review focuses on the importance of the orexin neural system as a novel mechanism related to the pathophysiology of PTSD. Orexinergic neurons originate in the lateral hypothalamus and project widely to key neurotransmitter systems, autonomic neurons, the hypothalamic-pituitary-adrenal (HPA) axis, and fear-related neural circuits. After trauma or stress, the basolateral amygdala (BLA) transmits sensory information to the central nucleus of the amygdala (CeA) and in turn to the hypothalamus and other subcortical and brainstem regions to promote fear and threat behaviors. Orexin receptors have a prominent role in this circuit as fear conditioned orexin receptor knockout mice show decreased fear expression while dual orexin receptor antagonists (DORAs) inhibit fear acquisition and expression. Orexin activation of an infralimbic-amygdala circuit impedes fear extinction while DORA treatments enhance it. Increased orexin signaling to the amygdalo-cortical-hippocampal circuit promotes avoidance behaviors. Orexin has an important role in activating sympathetic nervous system (SNS) activity and HPA axis stress responses. Blockade of orexin receptors reduces fear-conditioned startle responses. In PTSD models, individuals demonstrate sleep disturbances such as increased sleep latency and more transitions to wakefulness. Increased orexin activity impairs sleep by promoting wakefulness and reducing total sleep time while DORA treatments enhance sleep onset and maintenance. The orexinergic neural system provides important mechanisms for understanding multiple PTSD behaviors and provides new medication targets to treat this often persistent and debilitating illness., (Published by Elsevier Inc.)
- Published
- 2022
- Full Text
- View/download PDF
9. Translational approaches to influence sleep and arousal.
- Author
-
Brown RE, Spratt TJ, and Kaplan GB
- Subjects
- Arousal, Humans, Quality of Life, Sleep, Sleep Wake Disorders therapy, Veterans psychology
- Abstract
Sleep disorders are widespread in society and are prevalent in military personnel and in Veterans. Disturbances of sleep and arousal mechanisms are common in neuropsychiatric disorders such as schizophrenia, post-traumatic stress disorder, anxiety and affective disorders, traumatic brain injury, dementia, and substance use disorders. Sleep disturbances exacerbate suicidal ideation, a major concern for Veterans and in the general population. These disturbances impair quality of life, affect interpersonal relationships, reduce work productivity, exacerbate clinical features of other disorders, and impair recovery. Thus, approaches to improve sleep and modulate arousal are needed. Basic science research on the brain circuitry controlling sleep and arousal led to the recent approval of new drugs targeting the orexin/hypocretin and histamine systems, complementing existing drugs which affect GABA
A receptors and monoaminergic systems. Non-invasive brain stimulation techniques to modulate sleep and arousal are safe and show potential but require further development to be widely applicable. Invasive viral vector and deep brain stimulation approaches are also in their infancy but may be used to modulate sleep and arousal in severe neurological and psychiatric conditions. Behavioral, pharmacological, non-invasive brain stimulation and cell-specific invasive approaches covered here suggest the potential to selectively influence arousal, sleep initiation, sleep maintenance or sleep-stage specific phenomena such as sleep spindles or slow wave activity. These manipulations can positively impact the treatment of a wide range of neurological and psychiatric disorders by promoting the restorative effects of sleep on memory consolidation, clearance of toxic metabolites, metabolism, and immune function and by decreasing hyperarousal., (Published by Elsevier Inc.)- Published
- 2022
- Full Text
- View/download PDF
10. Suppression and acceptance in unipolar depression: Short-term and long-term effects on emotional responding.
- Author
-
Liverant GI, Gallagher MW, Arditte Hall KA, Rosebrock LE, Black SK, Kind S, Fava M, Kaplan GB, Kamholz BW, Pineles SL, and Sloan DM
- Subjects
- Affect, Emotions, Humans, Depressive Disorder, Emotional Regulation
- Abstract
Objectives: Transdiagnostic treatments increasingly include emotion regulation training focused on use of emotional suppression and acceptance. Despite the frequent use of these treatments in depression, little is known about the effects of these strategies in this population., Design: An experimental study., Methods: Eighty Veterans with unipolar depression participated in a study examining effects of these strategies on emotional responding (subjective, behavioural, and physiological). Physiological measures included: heart rate (HR), respiration (Resp), skin conductance (SC), and corrugator electromyography. On Day 1, participants were randomised to one of three conditions (acceptance, suppression, or control) and underwent an autobiographical sad mood induction. On Day 2, participants underwent a similar mood induction one week later., Results: The suppression group demonstrated reduced physiological reactivity (Resp and SC) on Day 1. However, the suppression group reported decreased positive affect on Day 2., Conclusions: Results support short-term effectiveness and longer term costs from suppression use among depressed individuals. Findings may inform application of transdiagnostic emotion regulation treatments and suggest suppression functions differently in depressed versus other clinical populations., (© 2021 British Psychological Society.)
- Published
- 2022
- Full Text
- View/download PDF
11. Opioid-induced structural and functional plasticity of medium-spiny neurons in the nucleus accumbens.
- Author
-
Thompson BL, Oscar-Berman M, and Kaplan GB
- Subjects
- Dopamine, Neuronal Plasticity, Neurons, Ventral Tegmental Area, Analgesics, Opioid, Nucleus Accumbens
- Abstract
Opioid Use Disorder (OUD) is a chronic relapsing clinical condition with tremendous morbidity and mortality that frequently persists, despite treatment, due to an individual's underlying psychological, neurobiological, and genetic vulnerabilities. Evidence suggests that these vulnerabilities may have neurochemical, cellular, and molecular bases. Key neuroplastic events within the mesocorticolimbic system that emerge through chronic exposure to opioids may have a determinative influence on behavioral symptoms associated with OUD. In particular, structural and functional alterations in the dendritic spines of medium spiny neurons (MSNs) within the nucleus accumbens (NAc) and its dopaminergic projections from the ventral tegmental area (VTA) are believed to facilitate these behavioral sequelae. Additionally, glutamatergic neurons from the prefrontal cortex, the basolateral amygdala, the hippocampus, and the thalamus project to these same MSNs, providing an enriched target for synaptic plasticity. Here, we review literature related to neuroadaptations in NAc MSNs from dopaminergic and glutamatergic pathways in OUD. We also describe new findings related to transcriptional, epigenetic, and molecular mechanisms in MSN plasticity in the different stages of OUD., (Copyright © 2020 Elsevier Ltd. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
12. Pathophysiological Bases of Comorbidity: Traumatic Brain Injury and Post-Traumatic Stress Disorder.
- Author
-
Kaplan GB, Leite-Morris KA, Wang L, Rumbika KK, Heinrichs SC, Zeng X, Wu L, Arena DT, and Teng YD
- Subjects
- Brain physiopathology, Comorbidity, Humans, Brain Injuries, Traumatic epidemiology, Brain Injuries, Traumatic physiopathology, Stress Disorders, Post-Traumatic epidemiology, Stress Disorders, Post-Traumatic physiopathology
- Abstract
The high rates of traumatic brain injury (TBI) and post-traumatic stress disorder (PTSD) diagnoses encountered in recent years by the United States Veterans Affairs Healthcare System have increased public awareness and research investigation into these conditions. In this review, we analyze the neural mechanisms underlying the TBI/PTSD comorbidity. TBI and PTSD present with common neuropsychiatric symptoms including anxiety, irritability, insomnia, personality changes, and memory problems, and this overlap complicates diagnostic differentiation. Interestingly, both TBI and PTSD can be produced by overlapping pathophysiological changes that disrupt neural connections termed the "connectome." The neural disruptions shared by PTSD and TBI and the comorbid condition include asymmetrical white matter tract abnormalities and gray matter changes in the basolateral amygdala, hippocampus, and prefrontal cortex. These neural circuitry dysfunctions result in behavioral changes that include executive function and memory impairments, fear retention, fear extinction deficiencies, and other disturbances. Pathophysiological etiologies can be identified using experimental models of TBI, such as fluid percussion or blast injuries, and for PTSD, using models of fear conditioning, retention, and extinction. In both TBI and PTSD, there are discernible signs of neuroinflammation, excitotoxicity, and oxidative damage. These disturbances produce neuronal death and degeneration, axonal injury, and dendritic spine dysregulation and changes in neuronal morphology. In laboratory studies, various forms of pharmacological or psychological treatments are capable of reversing these detrimental processes and promoting axonal repair, dendritic remodeling, and neurocircuitry reorganization, resulting in behavioral and cognitive functional enhancements. Based on these mechanisms, novel neurorestorative therapeutics using anti-inflammatory, antioxidant, and anticonvulsant agents may promote better outcomes for comorbid TBI and PTSD.
- Published
- 2018
- Full Text
- View/download PDF
13. Dopamine D1 receptor agonist treatment attenuates extinction of morphine conditioned place preference while increasing dendritic complexity in the nucleus accumbens core.
- Author
-
Kobrin KL, Arena DT, Heinrichs SC, Nguyen OH, and Kaplan GB
- Subjects
- Animals, Conditioning, Psychological physiology, Dendrites drug effects, Dendrites pathology, Dendrites physiology, Dose-Response Relationship, Drug, Extinction, Psychological drug effects, Extinction, Psychological physiology, Male, Mice, Inbred C57BL, Nucleus Accumbens pathology, Nucleus Accumbens physiopathology, Receptors, Dopamine D1 agonists, Receptors, Dopamine D1 metabolism, Reward, Spatial Behavior drug effects, Spatial Behavior physiology, Benzazepines pharmacology, Conditioning, Psychological drug effects, Dopamine Agonists pharmacology, Morphine pharmacology, Narcotics pharmacology, Nucleus Accumbens drug effects
- Abstract
The dopamine D1 receptor (D1R) has a role in opioid reward and conditioned place preference (CPP), but its role in CPP extinction is undetermined. We examined the effect of D1R agonist SKF81297 on the extinction of opioid CPP and associated dendritic morphology in the nucleus accumbens (NAc), a region involved with reward integration and its extinction. During the acquisition of morphine CPP, mice received morphine and saline on alternate days; injections were given immediately before each of eight daily conditioning sessions. Mice subsequently underwent six days of extinction training designed to diminish the previously learned association. Mice were treated with either 0.5mg/kg SKF81297, 0.8mg/kg SKF81297, or saline immediately after each extinction session. There was a dose-dependent effect, with the highest dose of SKF81297 attenuating extinction, as mice treated with this dose had significantly higher CPP scores than controls. Analysis of medium spiny neuron morphology revealed that in the NAc core, but not in the shell, dendritic arbors were significantly more complex in the morphine conditioned, SKF81297-treated mice compared to controls. In separate experiments using mice conditioned with only saline, SKF81297 administration after extinction sessions had no effect on CPP and produced differing effects on dendritic morphology. At the doses used in our experiments, SKF81297 appears to maintain previously learned opioid conditioned behavior, even in the face of new information. The D1R agonist's differential, rather than unidirectional, effects on dendritic morphology in the NAc core suggests that it may be involved in encoding reward information depending on previously learned behavior., (Published by Elsevier B.V.)
- Published
- 2017
- Full Text
- View/download PDF
14. Acquisition of morphine conditioned place preference increases the dendritic complexity of nucleus accumbens core neurons.
- Author
-
Kobrin KL, Moody O, Arena DT, Moore CF, Heinrichs SC, and Kaplan GB
- Subjects
- Analysis of Variance, Animals, Conditioning, Psychological drug effects, Dendrites drug effects, Male, Mice, Inbred C57BL, Nucleus Accumbens anatomy & histology, Analgesics, Opioid pharmacology, Homing Behavior drug effects, Morphine pharmacology, Nucleus Accumbens drug effects
- Abstract
Contexts associated with opioid reward trigger craving and relapse in opioid addiction. Effects of reward-context associative learning on nucleus accumbens (NAc) dendritic morphology were studied using morphine conditioned place preference (CPP). Morphine-conditioned mice received saline and morphine 10 mg/kg subcutaneous (s.c.) on alternate days. Saline-conditioned mice received saline s.c. each day. Morphine-conditioned and saline-conditioned groups received injections immediately before each of eight daily conditioning sessions. Morphine homecage controls had no CPP training, but received saline and morphine in the homecage concomitantly with the morphine-conditioned group. Morphine conditioning produced greater place preference than saline conditioning. Mice were sacrificed 1 day after CPP expression. Dendritic changes were studied using Golgi-Cox staining and digital tracing of NAc core and shell neurons. In the NAc core, morphine homecage administration increased spine density, while morphine conditioning increased dendritic complexity, as defined by increased dendritic count, length and intersections. Place preference positively correlated with dendritic length and intersections in the NAc core. The core may mediate reward consolidation and determine how context-related signals from the shell lead to motor behavior. The combination of drug and conditioning in the morphine-conditioned group produced unique morphological effects different from the effects of drug or conditioning procedures by themselves. An additional study found no differences in neuron morphology between saline-conditioned mice, trained as described earlier, and mice that were not conditioned, but received saline in the homecage. The unique effect of morphine reward learning on NAc core dendrites reflects a brain substrate that could be targeted for therapeutic intervention in addiction., (Published 2015. This article is a U.S. Government work and is in the public domain in the USA.)
- Published
- 2016
- Full Text
- View/download PDF
15. Associations among smoking, anhedonia, and reward learning in depression.
- Author
-
Liverant GI, Sloan DM, Pizzagalli DA, Harte CB, Kamholz BW, Rosebrock LE, Cohen AL, Fava M, and Kaplan GB
- Subjects
- Adult, Aged, Aged, 80 and over, Depression diagnosis, Depression etiology, Female, Humans, Male, Middle Aged, Surveys and Questionnaires, Tobacco Use Disorder psychology, Tobacco Use Disorder therapy, Anhedonia, Depression psychology, Learning physiology, Reward, Smoking adverse effects, Smoking Cessation psychology, Tobacco Use Disorder complications
- Abstract
Depression and cigarette smoking co-occur at high rates. However, the etiological mechanisms that contribute to this relationship remain unclear. Anhedonia and associated impairments in reward learning are key features of depression, which also have been linked to the onset and maintenance of cigarette smoking. However, few studies have investigated differences in anhedonia and reward learning among depressed smokers and depressed nonsmokers. The goal of this study was to examine putative differences in anhedonia and reward learning in depressed smokers (n=36) and depressed nonsmokers (n=44). To this end, participants completed self-report measures of anhedonia and behavioral activation (BAS reward responsiveness scores) and as well as a probabilistic reward task rooted in signal detection theory, which measures reward learning (Pizzagalli, Jahn, & O'Shea, 2005). When considering self-report measures, depressed smokers reported higher trait anhedonia and reduced BAS reward responsiveness scores compared to depressed nonsmokers. In contrast to self-report measures, nicotine-satiated depressed smokers demonstrated greater acquisition of reward-based learning compared to depressed nonsmokers as indexed by the probabilistic reward task. Findings may point to a potential mechanism underlying the frequent co-occurrence of smoking and depression. These results highlight the importance of continued investigation of the role of anhedonia and reward system functioning in the co-occurrence of depression and nicotine abuse. Results also may support the use of treatments targeting reward learning (e.g., behavioral activation) to enhance smoking cessation among individuals with depression., (Copyright © 2014. Published by Elsevier Ltd.)
- Published
- 2014
- Full Text
- View/download PDF
16. Extinction of opiate reward reduces dendritic arborization and c-Fos expression in the nucleus accumbens core.
- Author
-
Leite-Morris KA, Kobrin KL, Guy MD, Young AJ, Heinrichs SC, and Kaplan GB
- Subjects
- Animals, Conditioning, Classical drug effects, Dendrites drug effects, Dendrites pathology, Dendritic Spines pathology, Dendritic Spines physiology, Dose-Response Relationship, Drug, Male, Mice, Mice, Inbred C57BL, Nucleus Accumbens drug effects, Nucleus Accumbens pathology, Opioid-Related Disorders pathology, Opioid-Related Disorders physiopathology, Reward, Space Perception drug effects, Substance Withdrawal Syndrome pathology, Substance Withdrawal Syndrome physiopathology, Dendrites physiology, Extinction, Psychological physiology, Morphine pharmacology, Narcotics pharmacology, Nucleus Accumbens physiopathology, Proto-Oncogene Proteins c-fos metabolism
- Abstract
Recurrent opiate use combined with environmental cues, in which the drug was administered, provokes cue-induced drug craving and conditioned drug reward. Drug abuse craving is frequently linked with stimuli from a prior drug-taking environment via classical conditioning and associative learning. We modeled the conditioned morphine reward process by using acquisition and extinction of conditioned place preference (CPP) in C57BL/6 mice. Mice were trained to associate a morphine injection with a drug context using a classical conditioning paradigm. In morphine conditioning (0, 0.25, 0.5, 1, 5, or 10 mg/kg) experimental mice acquired a morphine CPP dose response with 10mg/kg as most effective. During morphine CPP extinction experiments, mice were divided into three test groups: morphine CPP followed by extinction training, morphine CPP followed by sham extinction, and saline controls. Extinction of morphine CPP developed within one extinction experiment (4 days) that lasted over two more trials (another 8 days). However, the morphine CPP/sham extinction group retained a place preference that endured through all three extinction trials. Brains were harvested following CPP extinction and processed using Golgi-Cox impregnation. Changes in dendritic morphology and spine quantity were examined in the nucleus accumbens (NAc) Core and Shell neurons. In the NAcCore only, morphine CPP/extinguished mice produced less dendritic arborization, and a decrease in neuronal activity marker c-Fos compared to the morphine CPP/sham extinction group. Extinction of morphine CPP is associated with decreased structural complexity of dendrites in the NAcCore and may represent a substrate for learning induced structural plasticity relevant to addiction., (Published by Elsevier B.V.)
- Published
- 2014
- Full Text
- View/download PDF
17. Repeated valproate treatment facilitates fear extinction under specific stimulus conditions.
- Author
-
Heinrichs SC, Leite-Morris KA, Rasmusson AM, and Kaplan GB
- Subjects
- Animals, Butyric Acid pharmacology, Male, Mice, Time Factors, Conditioning, Classical drug effects, Extinction, Psychological drug effects, Fear drug effects, Histone Deacetylase Inhibitors pharmacology, Valproic Acid pharmacology
- Abstract
Single dose treatment with histone deacetylase inhibitor (HDACi) agents has been shown to enhance extinction learning in rodent models under certain conditions. The present novel studies were designed to examine the effects of repeated HDACi treatment, with valproate or sodium butyrate, on the extinction of conditioned fear. In Experiments 1 and 2, short duration CS exposure (30s) in combination with vehicle administration progressively attenuated conditioned fear responses over 40 or more sessions. This effective extinction training was not augmented by HDACi treatments. In Experiment 3, we used a long duration CS exposure (120 s) to weaken extinction training. With these extinction parameters, repeated valproate treatment substantially facilitated the acquisition and retention of fear extinction. Results of this study extend previous work suggesting that HDACi's have utility in augmenting the efficiency of fear extinction, although their apparent benefits are critically dependent upon specific parameters of extinction training., (Copyright © 2013. Published by Elsevier Ireland Ltd.)
- Published
- 2013
- Full Text
- View/download PDF
18. Association between smoking and heart rate variability among individuals with depression.
- Author
-
Harte CB, Liverant GI, Sloan DM, Kamholz BW, Rosebrock LE, Fava M, and Kaplan GB
- Subjects
- Adult, Aged, Female, Humans, Male, Middle Aged, Autonomic Nervous System physiopathology, Depressive Disorder physiopathology, Heart Rate physiology, Smoking physiopathology
- Abstract
Background: Both depression and smoking have been independently associated with lower heart rate variability (HRV), suggesting dysregulation of cardiac autonomic function. However, no studies have systematically explored the effects of smoking on HRV among depressed patients., Purpose: This study examined differences in HRV based on smoking status among depressed individuals., Methods: Electrophysiological data were examined among 77 adult outpatients without a history of myocardial infarction, who met criteria for major depressive disorder or dysthymia. Frequency domain [low frequency (LF), high frequency (HF), LF/HF ratio, respiratory sinus arrhythmia (RSA)] parameters of HRV, and heart rate and inter-beat interval (IBI) data were compared between depressed smokers (n = 34) and depressed nonsmokers (n = 44)., Results: After controlling for covariates, depressed smokers, compared to depressed nonsmokers, displayed significantly lower LF, HF, and RSA., Conclusions: Among depressed patients, smoking is associated with significantly lower HRV, indicating dysregulated autonomic modulation of the heart.
- Published
- 2013
- Full Text
- View/download PDF
19. Dendritic structural plasticity in the basolateral amygdala after fear conditioning and its extinction in mice.
- Author
-
Heinrichs SC, Leite-Morris KA, Guy MD, Goldberg LR, Young AJ, and Kaplan GB
- Subjects
- Amygdala pathology, Animals, Electroshock methods, Fear psychology, Male, Mice, Mice, Inbred C57BL, Neurons pathology, Neurons physiology, Amygdala physiology, Conditioning, Psychological physiology, Dendrites pathology, Extinction, Psychological physiology, Fear physiology, Neuronal Plasticity physiology
- Abstract
Previous research suggests that morphology and arborization of dendritic spines change as a result of fear conditioning in cortical and subcortical brain regions. This study uniquely aims to delineate these structural changes in the basolateral amygdala (BLA) after both fear conditioning and fear extinction. C57BL/6 mice acquired robust conditioned fear responses (70-80% cued freezing behavior) after six pairings with a tone cue associated with footshock in comparison to unshocked controls. During fear acquisition, freezing behavior was significantly affected by both shock exposure and trial number. For fear extinction, mice were exposed to the conditioned stimulus tone in the absence of shock administration and behavioral responses significantly varied by shock treatment. In the retention tests over 3 weeks, the percentage time spent freezing varied with the factor of extinction training. In all treatment groups, alterations in dendritic plasticity were analyzed using Golgi-Cox staining of dendrites in the BLA. Spine density differed between the fear conditioned group and both the fear extinction and control groups on third order dendrites. Spine density was significantly increased in the fear conditioned group compared to the fear extinction group and controls. Similarly in Sholl analyses, fear conditioning significantly increased BLA spine numbers and dendritic intersections while subsequent extinction training reversed these effects. In summary, fear extinction produced enduring behavioral plasticity that is associated with a reversal of alterations in BLA dendritic plasticity produced by fear conditioning. These neuroplasticity findings can inform our understanding of structural mechanisms underlying stress-related pathology can inform treatment research into these disorders., (Published by Elsevier B.V.)
- Published
- 2013
- Full Text
- View/download PDF
20. Alterations in expression and phosphorylation of GluA1 receptors following cocaine-cue extinction learning.
- Author
-
Nic Dhonnchadha BÁ, Lin A, Leite-Morris KA, Kaplan GB, Man HY, and Kantak KM
- Subjects
- Amygdala drug effects, Amygdala metabolism, Animals, Behavior, Addictive metabolism, Conditioning, Operant drug effects, Conditioning, Operant physiology, Cues, Extinction, Psychological physiology, Male, Nucleus Accumbens drug effects, Nucleus Accumbens metabolism, Phosphorylation, Prefrontal Cortex drug effects, Prefrontal Cortex metabolism, Rats, Self Administration, Cocaine administration & dosage, Dopamine Uptake Inhibitors administration & dosage, Extinction, Psychological drug effects, Receptors, AMPA metabolism
- Abstract
Brain regional analyses of total GluA1 and GluA1-pSer(845) were used to delineate plasticity of the AMPA receptor in conjunction with cocaine-cue extinction learning. Rats were trained to self-administer cocaine paired with a 2-s light cue and later underwent a single 2 h extinction session for which cocaine was withheld but response-contingent cues were presented. Control groups received yoked-saline sessions or received cocaine self-administration training without undergoing extinction training. Extinction-related increases and decreases, respectively, in total GluA1 were observed in the ventromedial prefrontal cortex (vmPFC) and basolateral amygdala (BLA). Phosphorylation of GluA1 at Ser(845) was increased in the vmPFC and nucleus accumbens (NAc). Though total GluA1 did not change in NAc, there was a positive association between the number of responses during extinction training and the magnitude of total GluA1 in NAc. No significant changes were evident in the dorsal hippocampus. We conclude that the BLA and vmPFC, in particular, appear to be loci for the inhibition of learned behavior induced via extinction training, but each site may have different signaling functions for cocaine-cue extinction learning., (Copyright © 2012 Elsevier B.V. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
21. Separate and combined effects of very low nicotine cigarettes and nicotine replacement in smokers with schizophrenia and controls.
- Author
-
Tidey JW, Rohsenow DJ, Kaplan GB, Swift RM, and Ahnallen CG
- Subjects
- Administration, Cutaneous, Adult, Female, Humans, Male, Middle Aged, Smoking Cessation methods, Substance Withdrawal Syndrome psychology, Tobacco Use Cessation Devices, Treatment Outcome, Schizophrenic Psychology, Smoking psychology, Smoking Cessation psychology, Tobacco Products
- Abstract
Introduction: The prevalence of smoking among people with schizophrenia in the United States is about 3 times that of the general population. Novel approaches are needed to reduce rates of smoking-related morbidity and mortality among these smokers., Methods: This study used a within-subjects design to investigate the separate and combined effects of sensorimotor replacement for smoking (very low nicotine content [VLNC] cigarettes vs. no cigarettes) and transdermal nicotine replacement (42 mg nicotine [NIC] vs. placebo [PLA] patches) in smokers with schizophrenia (SS; n = 30) and control smokers without psychiatric illness (CS; n = 26). Each session contained a 5-hr controlled administration period in which participants underwent the following conditions, in counterbalanced order: VLNC + NIC, VLNC + PLA, no cigarettes + NIC, no cigarettes + PLA, usual-brand cigarettes + no patches. Next, participants completed measures of cigarette craving, nicotine withdrawal, smoking habit withdrawal, and cigarette subjective effects, followed by a 90-min period of ad libitum usual-brand smoking., Results: Smoking VLNC cigarettes during the controlled administration periods reduced cigarette craving, nicotine withdrawal symptoms, habit withdrawal symptoms, and usual-brand smoking in SS and CS relative to the no cigarette conditions. VLNC cigarettes were well accepted by both groups and did not affect psychiatric symptom levels in SS. Transdermal nicotine significantly reduced cigarette craving but did not affect usual-brand smoking., Conclusions: These findings suggest that reducing the nicotine content of cigarettes to nonaddictive levels may be a promising approach for reducing nicotine dependence among people with schizophrenia.
- Published
- 2013
- Full Text
- View/download PDF
22. Changes in expression of c-Fos protein following cocaine-cue extinction learning.
- Author
-
Nic Dhonnchadha BÁ, Lovascio BF, Shrestha N, Lin A, Leite-Morris KA, Man HY, Kaplan GB, and Kantak KM
- Subjects
- Animals, Behavior, Addictive, Behavior, Animal drug effects, Brain metabolism, Male, Rats, Rats, Wistar, Receptors, AMPA metabolism, Brain drug effects, Cocaine pharmacology, Extinction, Psychological drug effects, Learning drug effects, Proto-Oncogene Proteins c-fos metabolism
- Abstract
Extinguishing abnormally strengthened learned responses to cues associated with drugs of abuse remains a key tactic for alleviating addiction. To assist in developing pharmacotherapies to augment exposure therapy for relapse prevention, investigation into neurobiological underpinnings of drug-cue extinction learning is needed. We used regional analyses of c-Fos and GluR2 protein expression to delineate neural activity and plasticity that may be associated with cocaine-cue extinction learning. Rats were trained to self-administer cocaine paired with a light cue, and later underwent a single 2h extinction session for which cocaine was withheld but response-contingent cues were presented (cocaine-cue extinction). Control groups consisted of rats yoked to animals self-administering cocaine and receiving saline non-contingently followed by an extinction session, or rats trained to self-administer cocaine followed by a no-extinction session for which levers were retracted, and cocaine and cues were withheld. Among 11 brain sites examined, extinction training increased c-Fos expression in basolateral amygdala and prelimbic prefrontal cortex of cocaine-cue extinguished rats relative to both control conditions. In dorsal subiculum and infralimbic prefrontal cortex, extinction training increased c-Fos expression in both cocaine-cue and saline-cue extinguished rats relative to the no-extinction control condition. GluR2 protein expression was not altered in any site examined after extinction or control training. Findings suggest that basolateral amygdala and prelimbic prefrontal cortex neurons are activated during acquisition of cocaine-cue extinction learning, a process that is independent of changes in GluR2 abundance. Other sites are implicated in processing the significance of cues that are present early in extinction training., (Copyright © 2012 Elsevier B.V. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
23. The relationship between reward-based learning and nicotine dependence in smokers with schizophrenia.
- Author
-
Ahnallen CG, Liverant GI, Gregor KL, Kamholz BW, Levitt JJ, Gulliver SB, Pizzagalli DA, Koneru VK, and Kaplan GB
- Subjects
- Anhedonia, Behavior, Addictive psychology, Case-Control Studies, Female, Humans, Male, Middle Aged, Psychomotor Performance, Psychotic Disorders complications, Psychotic Disorders psychology, Reaction Time, Signal Detection, Psychological, Tobacco Use Disorder complications, Learning, Reward, Schizophrenia complications, Schizophrenic Psychology, Smoking psychology, Tobacco Use Disorder psychology
- Abstract
Cigarette smoking rates remain remarkably high in schizophrenia relative to smoking in other psychiatric groups. Impairments in the reward system may be related to elevated rates of nicotine dependence and lower cessation rates in this psychiatric group. Smokers with schizophrenia and schizoaffective disorder (SWS; n=15; M(age)=54.87, S.D.=6.51, 100% male) and a non-psychiatric control group of smokers (NCL; n=16; M(age)=50.38, S.D.=11.52; 93.8% male) were administered a computerized signal detection task to measure reward-based learning. Performance on the signal detection task was assessed by response bias, discriminability, reaction time, and hit rate. Clinician-assessed and self-reported measures of smoking and psychiatric symptoms were completed. SWS exhibited similar patterns of reward-based learning compared to control smokers. However, decreased reward-based learning was associated with increased levels of nicotine dependence in SWS, but not among control smokers. Nicotine withdrawal and urge to smoke were correlated with anhedonia within the SWS group. Among SWS, reduced reward responsiveness and increased anhedonia were associated with and may contribute to greater co-occurring nicotine dependence. These findings emphasize the importance of targeting reward system functioning in smoking cessation treatment for individuals with schizophrenia., (Published by Elsevier Ireland Ltd.)
- Published
- 2012
- Full Text
- View/download PDF
24. Effects of contingency management and bupropion on cigarette smoking in smokers with schizophrenia.
- Author
-
Tidey JW, Rohsenow DJ, Kaplan GB, Swift RM, and Reid N
- Subjects
- Adult, Behavior, Addictive drug therapy, Behavior, Addictive psychology, Breath Tests, Bupropion administration & dosage, Carbon Monoxide analysis, Combined Modality Therapy, Cotinine urine, Diagnosis, Dual (Psychiatry), Double-Blind Method, Female, Humans, Male, Patient Dropouts psychology, Reinforcement, Psychology, Schizophrenic Psychology, Smoking psychology, Smoking Cessation psychology, Substance Withdrawal Syndrome psychology, Behavior Therapy methods, Bupropion therapeutic use, Schizophrenia diagnosis, Smoking drug therapy, Smoking Cessation methods, Substance Withdrawal Syndrome prevention & control
- Abstract
Rationale: Individuals with schizophrenia have high smoking-related morbidity and mortality rates and need powerful and innovative smoking cessation interventions., Objectives: This proof-of-concept study investigated the feasibility and initial efficacy of combining a contingency management intervention with bupropion to reduce smoking in people with schizophrenia., Methods: Using a double-blind, placebo-controlled, between-groups design, 57 non-treatment-seeking participants were randomized to receive 300 mg/day bupropion or placebo. One week later, participants were randomized to a contingency management (CM) intervention in which reductions in urinary cotinine levels were reinforced, or a non-contingent reinforcement (NR) condition in which session attendance was reinforced, regardless of cotinine level. Over the 22-day study period, participants visited the laboratory approximately three times per week to provide urine samples for analysis of cotinine levels, to give breath samples for analysis of carbon monoxide (CO) levels, and to report number of cigarettes smoked per day, nicotine withdrawal symptoms, cigarette craving, and psychiatric symptoms., Results: Cotinine and CO levels significantly decreased during the study period in participants randomized to the CM condition, but not the NR condition. Bupropion did not reduce cotinine levels or increase the efficacy of CM. Cigarette craving and psychiatric symptom levels significantly decreased during the study in all groups., Conclusions: The results of this study indicate the efficacy and feasibility of this CM intervention for reducing smoking in individuals with schizophrenia.
- Published
- 2011
- Full Text
- View/download PDF
25. The use of cognitive enhancers in animal models of fear extinction.
- Author
-
Kaplan GB and Moore KA
- Subjects
- Adrenergic Agents pharmacology, Animals, Brain drug effects, Brain physiology, Cannabinoid Receptor Modulators pharmacology, Cholinergic Agents pharmacology, Conditioning, Classical, Dopamine Agents pharmacology, Epigenesis, Genetic drug effects, Excitatory Amino Acid Agents pharmacology, Extinction, Psychological drug effects, Extinction, Psychological physiology, Fear physiology, GABA Agents pharmacology, Humans, Memory drug effects, Memory physiology, Models, Animal, Fear drug effects, Nootropic Agents pharmacology
- Abstract
In anxiety disorders, such as posttraumatic stress disorders and phobias, classical conditioning pairs natural (unconditioned) fear-eliciting stimuli with contextual or discrete cues resulting in enduring fear responses to multiple stimuli. Extinction is an active learning process that results in a reduction of conditioned fear responses after conditioned stimuli are no longer paired with unconditioned stimuli. Fear extinction often produces incomplete effects and this highlights the relative permanence of bonds between conditioned stimuli and conditioned fear responses. The animal research literature is rich in its demonstration of cognitive enhancing agents that alter fear extinction. This review specifically examines the fear extinguishing effects of cognitive enhancers that act on gamma-aminobutyric acid (GABA), glutamatergic, cholinergic, adrenergic, dopaminergic, and cannabinoid signaling pathways. It also examines the effects of compounds that alter epigenetic and neurotrophic mechanisms in fear extinction. Of these cognitive enhancers, glutamatergic N-methyl d-aspartate (NMDA) receptor agonists, such as D-cycloserine, have enhanced fear extinction in a context-, dose- and time-dependent manner. Agents that function as glutamatergic α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor agonists, alpha2-adrenergic receptor antagonists (such as yohimbine), neurotrophic factors (brain derived neurotrophic factor or BDNF) and histone deacetylase inhibitors (valproate and sodium butyrate) also improve fear extinction in animals. However, some have anxiogenic effects and their contextual and temporal effects need to be more reliably demonstrated. Various cognitive enhancers produce changes in cortico-amygdala synaptic plasticity through multiple mechanisms and these neural changes enhance fear extinction. We need to better define the changes in neural plasticity produced by these agents in order to develop more effective compounds. In the clinical setting, such use of effective cognitive enhancers with cue exposure therapy, using compounds derived from animal model studies, provides great hope for the future treatment of anxiety disorders., (Published by Elsevier Inc.)
- Published
- 2011
- Full Text
- View/download PDF
26. Treatment of addiction and anxiety using extinction approaches: neural mechanisms and their treatment implications.
- Author
-
Kaplan GB, Heinrichs SC, and Carey RJ
- Subjects
- Animals, Anxiety psychology, Conditioning, Classical, Fear, Humans, Motivation, Substance-Related Disorders psychology, Anxiety therapy, Substance-Related Disorders therapy
- Abstract
Clinical interventions which produce cue and contextual extinction learning can reduce craving and relapse in substance abuse and inhibit conditioned fear responses in anxiety disorders. In both types of disorders, classical conditioning links unconditioned drug or fear responses to associated contextual cues and result in enduring pathological responses to multiple stimuli. Extinction therapy countermeasures seek to reduce conditioned responses using a set of techniques in which patients are repeatedly exposed to conditioned appetitive or aversive stimuli using imaginal imagery, in vivo exposure, or written scripts. Such interventions allow patients to rehearse more adaptive responses to conditioned stimuli. The ultimate goal of these interventions, extinction of the original conditioned response, is a new learning process that results in a decrease in frequency or intensity of conditioned responses to drug or fear cues. This review explores extinction approaches in conditioned drug reward and fear responses. The behavioral, neuroanatomical and neurochemical mechanisms of conditioned reward and fear responses and their extinction are derived from our understanding of the animal literature. Extensive neuroscience research shows that even though many mechanisms differ in conditioned fear and reward, converging prefrontal cortical glutamatergic pathways underlie extinction learning. Efficacy of pharmacological and behavioral treatment approaches in addiction and anxiety disorders may be optimized by enhancing extinction and weakening the bond between the original conditioned stimuli and conditioned responses. Adjunctive pharmacotherapy approaches using agents which alter glutamate or γ-aminobutyric acid signaling or epigenetic mechanisms in prefrontal cortical pathways can enhance extinction learning. A comparative study of extinction processes and its neural mechanisms can be translated into more effective behavioral and pharmacological treatment approaches in substance abuse and anxiety., (Published by Elsevier Inc.)
- Published
- 2011
- Full Text
- View/download PDF
27. Opiate sensitization induces FosB/ΔFosB expression in prefrontal cortical, striatal and amygdala brain regions.
- Author
-
Kaplan GB, Leite-Morris KA, Fan W, Young AJ, and Guy MD
- Subjects
- Amygdala drug effects, Animals, Immunohistochemistry, Male, Mice, Mice, Inbred C57BL, Morphine administration & dosage, Motor Activity drug effects, Mutation genetics, Neostriatum drug effects, Prefrontal Cortex drug effects, Amygdala metabolism, Morphine pharmacology, Neostriatum metabolism, Prefrontal Cortex metabolism, Proto-Oncogene Proteins c-fos metabolism
- Abstract
Sensitization to the effects of drugs of abuse and associated stimuli contributes to drug craving, compulsive drug use, and relapse in addiction. Repeated opiate exposure produces behavioral sensitization that is hypothesized to result from neural plasticity in specific limbic, striatal and cortical systems. ΔFosB and FosB are members of the Fos family of transcription factors that are implicated in neural plasticity in addiction. This study examined the effects of intermittent morphine treatment, associated with motor sensitization, on FosB/ΔFosB levels using quantitative immunohistochemistry. Motor sensitization was tested in C57BL/6 mice that received six intermittent pre-treatments (on days 1, 3, 5, 8, 10, 12) with either subcutaneous morphine (10 mg/kg) or saline followed by a challenge injection of morphine or saline on day 16. Mice receiving repeated morphine injections demonstrated significant increases in locomotor activity on days 8, 10, and 12 of treatment (vs. day 1), consistent with development of locomotor sensitization. A morphine challenge on day 16 significantly increased locomotor activity of saline pre-treated mice and produced even larger increases in motor activity in the morphine pre-treated mice, consistent with the expression of opiate sensitization. Intermittent morphine pre-treatment on these six pre-treatment days produced a significant induction of FosB/ΔFosB, measured on day 16, in multiple brain regions including prelimbic (PL) and infralimbic (IL) cortex, nucleus accumbens (NAc) core, dorsomedial caudate-putamen (CPU), basolateral amygdala (BLA) and central nucleus of the amygdala (CNA) but not in a motor cortex control region. Opiate induced sensitization may develop via Fos/ΔFosB plasticity in motivational pathways (NAc), motor outputs (CPU), and associative learning (PL, IL, BLA) and stress pathways (CNA).
- Published
- 2011
- Full Text
- View/download PDF
28. Brain-derived neurotrophic factor in traumatic brain injury, post-traumatic stress disorder, and their comorbid conditions: role in pathogenesis and treatment.
- Author
-
Kaplan GB, Vasterling JJ, and Vedak PC
- Subjects
- Animals, Brain Injuries complications, Brain Injuries therapy, Combat Disorders physiopathology, Combat Disorders therapy, Humans, Military Personnel, Neuronal Plasticity, Signal Transduction, Stress Disorders, Post-Traumatic complications, Stress Disorders, Post-Traumatic therapy, United States epidemiology, Veterans, Brain Injuries physiopathology, Brain-Derived Neurotrophic Factor metabolism, Stress Disorders, Post-Traumatic physiopathology
- Abstract
As US military service members return from the wars in Iraq and Afghanistan with elevated rates of traumatic brain injury (TBI) and post-traumatic stress disorder (PTSD), attention has been increasingly focused on TBI/PTSD comorbidity, its neurobiological mechanisms, and novel and effective treatment approaches. TBI and PTSD, and their comorbid conditions, present with a spectrum of common clinical features such as sleep disturbance, depression, anxiety, irritability, difficulty in concentrating, fatigue, suicidality, chronic pain, and alterations in arousal. These TBI and PTSD disorders are also thought to be characterized by overlapping neural mechanisms. Both conditions are associated with changes in hippocampal, prefrontal cortical, and limbic region function because of alterations in synaptogenesis, dendritic remodeling, and neurogenesis. Neural changes in TBI and PTSD result from pathophysiological disturbances in metabolic, cytotoxic, inflammatory, and apoptic processes, amongst other mechanisms. Neurotrophins have well-established actions in regulating cell growth and survival, differentiation, apoptosis, and cytoskeleton restructuring. A body of research indicates that dysregulation of neural brain-derived neurotrophic factor (BDNF) is found in conditions of TBI and PTSD. Induction of BDNF and activation of its intracellular receptors can produce neural regeneration, reconnection, and dendritic sprouting, and can improve synaptic efficacy. In this review, we consider treatment approaches that enhance BDNF-related signaling and have the potential to restore neural connectivity. Such treatment approaches could facilitate neuroplastic changes that lead to adaptive neural repair and reverse cognitive and emotional deficits in both TBI and PTSD.
- Published
- 2010
- Full Text
- View/download PDF
29. Intolerance for Smoking Abstinence Questionnaire: psychometric properties and relationship to tobacco dependence and abstinence.
- Author
-
Sirota AD, Rohsenow DJ, Mackinnon SV, Martin RA, Eaton CA, Kaplan GB, Monti PM, Tidey JW, and Swift RM
- Subjects
- Adult, Aged, Female, Humans, Male, Middle Aged, Psychometrics, Time Factors, Young Adult, Behavior, Addictive psychology, Smoking psychology, Smoking Cessation psychology, Stress, Psychological psychology, Substance Withdrawal Syndrome psychology, Surveys and Questionnaires, Tobacco Use Disorder psychology
- Abstract
Unlabelled: While smokers' ability to tolerate emotional or physical distress has been associated with length of smoking cessation, there is no measure of ability to tolerate smoking abstinence discomfort specifically, which may be more heuristic than a measure of tolerance of general emotional stress or physical discomfort., Methods: Questionnaires completed by 300 smokers assessed inability to tolerate smoking abstinence discomfort (IDQ-S), general physical discomfort (IDQ-P), and general emotional discomfort (IDQ-E), so that shared variance among these measures could be assessed., Results: The IDQ-S has three reliable components: withdrawal Intolerance, Lack of Cognitive Coping, and Pain Intolerance. The 14-item IDQ-P and 9-item IDQ-E each consist of one reliable component. Intercorrelations suggest only modest shared variance. Support for construct and discriminant validity was seen. Two scales of the IDQ-S showed excellent convergent validity, correlating with smoking use, dependence, motivation, and length of past smoking cessation, while IDQ-P and IDQ-E correlated with few indices of use or dependence and not with smoking cessation., Conclusions: The final 17-item IDQ-S with two scales is reliable and valid, and more heuristic than measures of general physical or emotional discomfort intolerance as a correlate of motivation and past success with smoking cessation., (Published by Elsevier Ltd.)
- Published
- 2010
- Full Text
- View/download PDF
30. Baclofen enhances extinction of opiate conditioned place preference.
- Author
-
Heinrichs SC, Leite-Morris KA, Carey RJ, and Kaplan GB
- Subjects
- Animals, Baclofen administration & dosage, Dose-Response Relationship, Drug, GABA Agonists administration & dosage, Mice, Mice, Inbred C57BL, Motor Activity drug effects, Space Perception drug effects, Time Factors, Analgesics, Opioid pharmacology, Baclofen pharmacology, Conditioning, Classical drug effects, Extinction, Psychological drug effects, GABA Agonists pharmacology, Morphine pharmacology
- Abstract
Conditioned opiate reward (COR) is rapidly acquired and slowly extinguished. The slow rate of extinction of the salience of drug-related cues contributes to drug craving and relapse. The gamma-aminobutyric acid receptor type B (GABA(B)) agonist, baclofen, attenuates the unconditioned rewarding actions of several drugs of abuse and was investigated for effects on the extinction of COR. C57BL/6 mice were utilized in an unbiased conditioned place preference (CPP) protocol using morphine (10mg/kg, s.c.) and saline. CPP was measured by increases in time spent in the morphine-associated (CS+) vs. the saline-associated (CS-) chamber in a 15-min test after four morphine and four saline alternated conditioning sessions. CPP and locomotor sensitization were produced to the CS+ chamber. Subsequently, sixteen daily extinction sessions were conducted with either vehicle or baclofen (1 or 2.5mg/kg, s.c.) treatment given either before or after the sessions. This design was used to create the baclofen drug state before or after the activation of the CPP memory trace in the extinction protocol. After morphine CPP development, its extinction was significantly facilitated in a dose-dependent manner by post-session, but not by pre-session, baclofen treatments. No significant sedative effects of baclofen were detected during any extinction training or testing phase. Baclofen treatment facilitated the extinction of COR and reduced conditioned sensitization during extinction when given after, but not before, the activation of the CPP memory trace. Baclofen appears to have disrupted reconsolidation of conditioned reward memory during extinction training and might similarly facilitate extinction learning in human opiate addiction., (Copyright 2009 Elsevier B.V. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
31. Intra-VTA adenosine A1 receptor activation blocks morphine stimulation of motor behavior and cortical and limbic Fos immunoreactivity.
- Author
-
Kaplan GB, Leite-Morris KA, Klufas MA, and Fan W
- Subjects
- Adenosine analogs & derivatives, Adenosine pharmacology, Animals, Biomarkers metabolism, Cerebral Cortex immunology, Cerebral Cortex metabolism, Dose-Response Relationship, Drug, Gene Expression Regulation drug effects, Limbic System immunology, Limbic System metabolism, Male, Mice, Mice, Inbred C57BL, Morphine pharmacology, Neurons drug effects, Neurons metabolism, Nucleus Accumbens drug effects, Nucleus Accumbens immunology, Nucleus Accumbens physiology, Proto-Oncogene Proteins c-fos immunology, Theophylline analogs & derivatives, Theophylline pharmacology, Ventral Tegmental Area cytology, Ventral Tegmental Area drug effects, Adenosine A1 Receptor Agonists, Cerebral Cortex drug effects, Limbic System drug effects, Morphine antagonists & inhibitors, Motor Activity drug effects, Proto-Oncogene Proteins c-fos metabolism, Ventral Tegmental Area metabolism
- Abstract
Drugs of abuse produce psychomotor stimulation as one of their characteristic behavioral effects. Single administration of opiates stimulates motor activity via effects on gamma-aminobutyric acid (GABA) and dopamine transmission in the ventral tegmental area (VTA). Adenosine A(1) receptor agonists inhibit VTA GABAergic and dopaminergic transmission and are predicted to alter the behavioral effects of opiates. This study examined the effects of intra-VTA administration of selective adenosine A(1) receptor agonist N(6)-cyclopentyladenosine (CPA) and antagonist 8-cyclopentyl-1, 3-dimethylxanthine (CPT) on morphine-induced motor stimulation in C57BL/6 mice. It also examined the effects of CPA on morphine's activation of VTA neurons projecting to limbic and cortical regions including the nucleus accumbens (NAc), anterior cingulate cortex (ACg) and prelimbic cortex (PrL) via quantitation of immediate-early gene c-Fos protein in these regions. Mice received subcutaneous morphine and intra-VTA administration of CPA and then automated motor activity was measured. Morphine treatment induced both motor activity and Fos immunoreactivity in the NAc, ACg and PrL suggesting that behavioral stimulation is produced by neural activation in these regions. Intra-VTA CPA administration produced a dose-dependent inhibition of morphine-induced motor stimulation and blocked c-Fos induction in all three regions. Intra-VTA CPT treatment had no effects on motor activity or on morphine-induced motor stimulation. VTA adenosine A(1) agonist inhibition of morphine's effects on motor activity and of neural activation of VTA projections suggests that these neurons and their regulation are critical to morphine's stimulant effects. Adenosine A(1) receptor agonists and purinergic modulators may represent useful treatment approaches for blocking the behavioral effects of opiates.
- Published
- 2009
- Full Text
- View/download PDF
32. Effects of smoking abstinence, smoking cues and nicotine replacement in smokers with schizophrenia and controls.
- Author
-
Tidey JW, Rohsenow DJ, Kaplan GB, Swift RM, and Adolfo AB
- Subjects
- Administration, Cutaneous, Adult, Carbon Monoxide metabolism, Female, Humans, Male, Nicotine administration & dosage, Schizophrenic Psychology, Cues, Schizophrenia complications, Smoking Cessation methods, Smoking Prevention, Tobacco Use Disorder complications, Tobacco Use Disorder rehabilitation
- Abstract
The mechanisms underlying the low smoking cessation rates among smokers with schizophrenia (SS) are unknown. In this laboratory study, we compared the responses of 21 SS and 21 non-psychiatric controls (CS) to manipulations of 5-hour smoking abstinence, transdermal nicotine replacement (0 mg, 21 mg and 42 mg), and in vivo smoking cues. Results indicate that SS were more sensitive than CS to the effects of acute abstinence on carbon monoxide (CO) boost, but not more sensitive to the effects of abstinence on urge levels or withdrawal symptoms. SS and CS did not differ in urge response to in vivo smoking cues, but SS were less consistent in their reactions. These findings suggest that heightened sensitivity to the effects of abstinence on smoke intake may partially account for the low cessation rates experienced by SS, but other potential mechanisms should be explored using behavioral laboratory models.
- Published
- 2008
- Full Text
- View/download PDF
33. High-dose transdermal nicotine and naltrexone: effects on nicotine withdrawal, urges, smoking, and effects of smoking.
- Author
-
Rohsenow DJ, Monti PM, Hutchison KE, Swift RM, MacKinnon SV, Sirota AD, and Kaplan GB
- Subjects
- Administration, Cutaneous, Administration, Oral, Adult, Blood Pressure drug effects, Breath Tests, Capsules, Carbon Monoxide metabolism, Demography, Disruptive, Impulse Control, and Conduct Disorders prevention & control, Dose-Response Relationship, Drug, Female, Heart Rate drug effects, Humans, Male, Middle Aged, Naltrexone administration & dosage, Narcotic Antagonists administration & dosage, Narcotic Antagonists pharmacology, Nicotine administration & dosage, Nicotine adverse effects, Nicotinic Agonists administration & dosage, Nicotinic Agonists adverse effects, Nicotinic Agonists pharmacology, Smoking adverse effects, Substance Withdrawal Syndrome prevention & control, Surveys and Questionnaires, Tablets, Disruptive, Impulse Control, and Conduct Disorders diagnosis, Naltrexone pharmacology, Nicotine pharmacology, Smoking psychology, Substance Withdrawal Syndrome diagnosis
- Abstract
Although treatment with transdermal nicotine replacement (TNR) has improved smoking abstinence rates, higher doses of TNR could improve effects on urge to smoke, nicotine withdrawal, and reinforcement from smoking, and naltrexone might further reduce reinforcement and urges. A laboratory investigation with 134 smokers using a 3 x 2 parallel-group design evaluated the effects of TNR (42-mg, 21-mg, or 0-mg patch) as crossed with a single dose of naltrexone (50 mg) versus placebo on urge to smoke, withdrawal, and responses to an opportunity to smoke (intake, subjective effects) after 10 hr of deprivation. Urge and withdrawal were assessed both prior to and after cigarette cue exposure. Only 42 mg TNR, not 21 mg, prevented urge to smoke, heart rate change, and cue-elicited increase in withdrawal. Both 21 and 42 mg TNR blocked cue-elicited drop in heart rate and arterial pressure. Naltrexone reduced cue-elicited withdrawal symptoms but not urges to smoke or deprivation-induced withdrawal prior to cue exposure. Neither medication significantly affected carbon monoxide intake or subjective effects of smoking except that 42 mg TNR resulted in lower subjective physiological activation. No interaction effects were found, and no results differed by gender. Results suggest that starting smokers with 42 mg TNR may increase comfort during initial abstinence, but limited support is seen for naltrexone during smoking abstinence.
- Published
- 2007
- Full Text
- View/download PDF
34. A composite neural network model for perseveration and distractibility in the Wisconsin card sorting test.
- Author
-
Kaplan GB, Sengör NS, Gürvit H, Genç I, and Güzeliş C
- Subjects
- Brain Diseases complications, Brain Diseases physiopathology, Cognition Disorders physiopathology, Color Perception, Computer Simulation, Humans, Photic Stimulation methods, Attention physiology, Neural Networks, Computer, Neuropsychological Tests, Problem Solving physiology
- Abstract
A composite artificial neural network model is proposed to simulate the performance of the Wisconsin Card Sorting Test. The Wisconsin Card Sorting Test is a test of executive functions where prefrontal deficits are matched to some quantitative measures such as percentage of perseverative errors and number of failures to maintain set. In this work, the proposed model is used to simulate the performances of healthy subjects and patients with prefrontal involvement particularly on these measures. The model is designed in such a way that one of the subsystems, namely, the Hopfield network, serves as the working memory and the other, the Hamming block, as the hypothesis generator. The results show that the proposed relatively simple model is capable of simulating the wide range of the performances of both normal subjects and prefrontal patients on the Wisconsin Card Sorting Test. While lowering the Hamming distance in the Hamming block gave rise to progressively more perseverative responses, changing the threshold vector of the Hopfield network resulted in more set maintenance failures. The former manipulation disrupts the abstraction or mental flexibility and the latter sustained attention or perseverance both of which are the major functions of the prefrontal system.
- Published
- 2006
- Full Text
- View/download PDF
35. Assessment of insight into delusional beliefs in schizophrenia using the Brown Assessment of Beliefs Scale.
- Author
-
Kaplan GB, Phillips KA, Vaccaro A, Eisen JL, Posternak MA, and MacAskill HS
- Subjects
- Adult, Female, Humans, Male, Middle Aged, Pilot Projects, Psychometrics statistics & numerical data, Psychopathology, Reproducibility of Results, Statistics as Topic, Awareness, Culture, Delusions psychology, Psychiatric Status Rating Scales statistics & numerical data, Schizophrenia diagnosis, Schizophrenic Psychology
- Published
- 2006
- Full Text
- View/download PDF
36. Speech production in schizophrenia: preliminary data regarding voice onset time.
- Author
-
Epstein-Lubow G, Hochstadt J, Lieberman P, and Kaplan GB
- Subjects
- Adult, Antipsychotic Agents adverse effects, Antipsychotic Agents therapeutic use, Articulation Disorders physiopathology, Cerebral Cortex physiopathology, Corpus Striatum physiopathology, Diagnosis, Differential, Female, Humans, Male, Middle Aged, Nerve Net physiopathology, Phonetics, Pilot Projects, Psychiatric Status Rating Scales, Psychotic Disorders drug therapy, Psychotic Disorders physiopathology, Schizophrenia drug therapy, Schizophrenia physiopathology, Veterans psychology, Articulation Disorders diagnosis, Psychotic Disorders diagnosis, Schizophrenia diagnosis, Schizophrenic Language, Speech Acoustics, Speech Production Measurement
- Published
- 2006
- Full Text
- View/download PDF
37. Cigarette smoking topography in smokers with schizophrenia and matched non-psychiatric controls.
- Author
-
Tidey JW, Rohsenow DJ, Kaplan GB, and Swift RM
- Subjects
- Adolescent, Adult, Comorbidity, Female, Humans, Male, Prevalence, Psychotic Disorders diagnosis, Psychotic Disorders epidemiology, Schizophrenia diagnosis, Surveys and Questionnaires, Tobacco Use Disorder diagnosis, Schizophrenia epidemiology, Smoking epidemiology, Tobacco Use Disorder epidemiology
- Abstract
Smoking is highly prevalent among people with schizophrenia, and little is known about factors that affect smoking in these patients. One basic question is whether smoking behavior differs for smokers with schizophrenia compared to equally nicotine-dependent smokers who do not have a major mental illness. In this study, 20 smokers with schizophrenia or schizoaffective disorder (SCZ) and 20 non-psychiatric smokers (CON) underwent smoking topography assessments. The groups were matched on age, gender, daily smoking rate, years of regular smoking and nicotine dependence rating. Results indicate that, compared to the CON participants, the SCZ participants smoked significantly more total puffs (SCZ: 58.5 +/- 48.3; CON: 21.3 +/- 9.4) and puffs per cigarette (SCZ: 12.3 +/- 6.0; CON: 8.9 +/- 2.3) and had shorter inter-puff intervals (SCZ: 21.9 +/- 9.7 s; CON: 42.0 +/- 21.5 s), larger total cigarette puff volumes (SCZ: 583 +/- 169 ml; CON: 429 +/- 159 ml) and higher carbon monoxide boosts (SCZ: 3.8+/-5.4 ppm; CON: 1.0 +/- 2.5 ppm). Test-retest reliabilities were good to excellent for most smoking measures in both groups. These findings suggest that smokers with schizophrenia smoke more intensely than matched non-psychiatric smokers and that their smoking behavior is reliable when assessed under laboratory conditions.
- Published
- 2005
- Full Text
- View/download PDF
38. Subjective and physiological responses to smoking cues in smokers with schizophrenia.
- Author
-
Tidey JW, Rohsenow DJ, Kaplan GB, and Swift RM
- Subjects
- Blood Pressure, Female, Heart Rate, Humans, Male, Schizophrenia complications, Substance Withdrawal Syndrome physiopathology, Schizophrenia physiopathology, Smoking physiopathology
- Abstract
The prevalence of smoking is high among people with schizophrenia. Although several research groups are developing smoking treatments for these smokers, abstinence rates to date have been modest. Methodological tools such as cue exposure are useful in clinical research with smokers in general, but the value of these paradigms with smokers with schizophrenia has yet to be established. The aim of the present study was to determine the subjective and physiological effects of exposure to in vivo smoking cues in smokers with schizophrenia. A total of 25 heavy smokers with schizophrenia or schizoaffective disorder were assessed while nonabstinent and after 2-hr smoking abstinence. Urge to smoke, mood, nicotine withdrawal symptoms, heart rate, and blood pressure were measured during a precue relaxation period, after exposure to neutral cues, and after exposure to smoking cues. Results indicate that both exposure to smoking cues and brief abstinence increased urge levels, nicotine withdrawal symptom levels, and negative affect. Abstinence did not amplify the effects of cues on urges or other cue reactivity measures. These results indicate that smoking cue reactivity laboratory models may be useful for investigating potential smoking treatments for, or neurobiological contributions to, smoking behavior in smokers with schizophrenia.
- Published
- 2005
- Full Text
- View/download PDF
39. PROSSA challenge.
- Author
-
Kaplan GB
- Subjects
- Humans, South Africa, Prosthodontics organization & administration, Societies, Dental
- Published
- 2005
40. Baclofen as adjunctive treatment for a patient with cocaine dependence and schizoaffective disorder.
- Author
-
Kaplan GB, McRoberts RL 3rd, and Smokler HJ
- Subjects
- Antidepressive Agents, Second-Generation therapeutic use, Antipsychotic Agents therapeutic use, Comorbidity, Dose-Response Relationship, Drug, Drug Administration Schedule, Drug Therapy, Combination, Fluoxetine therapeutic use, Follow-Up Studies, Humans, Long-Term Care, Male, Marijuana Abuse rehabilitation, Middle Aged, Risperidone therapeutic use, Secondary Prevention, Alcoholism rehabilitation, Baclofen therapeutic use, Bipolar Disorder rehabilitation, Cocaine-Related Disorders rehabilitation, GABA Agonists therapeutic use
- Published
- 2004
- Full Text
- View/download PDF
41. Antipsychotics regulate cyclic AMP-dependent protein kinase and phosphorylated cyclic AMP response element-binding protein in striatal and cortical brain regions in mice.
- Author
-
Turalba AV, Leite-Morris KA, and Kaplan GB
- Subjects
- Animals, Benzodiazepines pharmacology, Cerebral Cortex enzymology, Corpus Striatum enzymology, Cyclic AMP metabolism, Cyclic AMP Response Element-Binding Protein drug effects, Cyclic AMP-Dependent Protein Kinases drug effects, Dose-Response Relationship, Drug, Down-Regulation drug effects, Down-Regulation physiology, Haloperidol pharmacology, Male, Mice, Nucleus Accumbens drug effects, Nucleus Accumbens metabolism, Olanzapine, Phosphorylation drug effects, Signal Transduction drug effects, Signal Transduction physiology, Antipsychotic Agents pharmacology, Cerebral Cortex drug effects, Corpus Striatum drug effects, Cyclic AMP Response Element-Binding Protein metabolism, Cyclic AMP-Dependent Protein Kinases metabolism
- Abstract
Adenosine 3',5'-monophosphate (cAMP) and cAMP-dependent protein kinase (PKA) signaling have been implicated in antipsychotic drug action. This study examines the effects of acute antipsychotic treatment using typical (haloperidol) and atypical (olanzapine) agents on cAMP signaling in dorsal striatum, nucleus accumbens and medial prefrontal cortex in mice. PKA catalytic subunit (PKA-c) and phosphorylated cAMP response element-binding protein (pCREB) levels were measured to evaluate antipsychotic drug effects. Nuclear PKA-c levels increased in the dorsal striatum after haloperidol and olanzapine treatment. In medial prefrontal cortex, olanzapine produced dose-dependent decreases in PKA-c and pCREB levels. The differential effects of typical versus atypical antipsychotic agents on PKA and pCREB in striatal and cortical regions illustrate the diverging actions of these agents on cAMP pathways.
- Published
- 2004
- Full Text
- View/download PDF
42. GABA(B) receptor activation in the ventral tegmental area inhibits the acquisition and expression of opiate-induced motor sensitization.
- Author
-
Leite-Morris KA, Fukudome EY, Shoeb MH, and Kaplan GB
- Subjects
- Animals, Baclofen pharmacology, Drug Administration Routes, Drug Combinations, Drug Interactions, GABA Agonists pharmacology, GABA Antagonists pharmacology, GABA-B Receptor Agonists, GABA-B Receptor Antagonists, Male, Mice, Mice, Inbred C57BL, Morphine administration & dosage, Morphine pharmacology, Motor Activity physiology, Narcotics pharmacology, Proto-Oncogene Proteins c-fos metabolism, Motor Activity drug effects, Receptors, GABA-B physiology, Ventral Tegmental Area metabolism
- Abstract
Opiate-induced motor sensitization refers to the progressive and enduring motor response that develops after intermittent drug administration, and results from neuroadaptive changes in ventral tegmental area (VTA) and nucleus accumbens (NAc) neurons. Repeated activation of mu-opioid receptors localized on gamma-aminobutyric acid (GABA) neurons in the VTA enhances dopaminergic cell activity and stimulates dopamine release in the nucleus accumbens. We hypothesize that GABA(B) receptor agonist treatment in the VTA blocks morphine-induced motor stimulation, motor sensitization, and accumbal Fos immunoreactivity by inhibiting the activation of dopaminergic neurons. First, C57BL/6 mice were coadministered a single subcutaneous injection of morphine with intra-VTA baclofen, a GABA(B) receptor agonist. Baclofen produced a dose-dependent inhibition of opiate-induced motor stimulation that was attenuated by 2-hydroxysaclofen, a GABA(B) receptor antagonist. Next, morphine was administered on days 1, 3, 5, and 9 and mice demonstrated sensitization to its motor stimulant effects and concomitant induction of Fos immunoreactivity in the NAc shell (NAcS) but not NAc core. Intra-VTA baclofen administered during morphine pretreatment blocked the acquisition of morphine-induced motor sensitization and Fos activation in the NAcS. Intra-VTA baclofen administered only on day 9 blocked the expression of morphine-induced motor sensitization and Fos activation in the NAcS. A linear relationship was found between morphine-induced motor activity and accumbal Fos in single- and repeated-dose treatment groups. In conclusion, GABA(B) receptor stimulation in the VTA blocked opiate-induced motor stimulation and motor sensitization by inhibiting the activation of NAcS neurons. GABA(B) receptor agonists may be useful pharmacological treatments in altering the behavioral effects of opiates.
- Published
- 2004
- Full Text
- View/download PDF
43. Baclofen inhibits opiate-induced conditioned place preference and associated induction of Fos in cortical and limbic regions.
- Author
-
Kaplan GB, Leite-Morris KA, Joshi M, Shoeb MH, and Carey RJ
- Subjects
- Animals, Behavior, Animal drug effects, Brain metabolism, Gyrus Cinguli drug effects, Immunohistochemistry, Limbic System drug effects, Male, Mice, Mice, Inbred C57BL, Nucleus Accumbens drug effects, Prosencephalon drug effects, Proto-Oncogene Proteins c-fos drug effects, Reward, Analgesics, Opioid pharmacology, Baclofen pharmacology, Brain drug effects, Conditioning, Operant drug effects, GABA Agonists pharmacology, Morphine pharmacology, Proto-Oncogene Proteins c-fos metabolism, Receptors, GABA-B metabolism
- Abstract
In C57BL/6 mice, pretreatment with GABA(B) receptor agonist baclofen blocked the rewarding effects of morphine as measured by acquisition of conditioned place preference. Fos immunoreactivity, a neuronal activity marker, was induced in opiate conditioned mice in several forebrain regions including the nucleus accumbens core and shell, anterior cingulate cortex, and prelimbic cortex. Baclofen pretreatment blocked the induction of Fos in opiate conditioned subjects. These result suggest that GABA(B) receptor transmission has a role in reversing morphine-induced activation of motivational circuitry and conditioned reward.
- Published
- 2003
- Full Text
- View/download PDF
44. Opiate-induced motor stimulation is regulated by gamma-aminobutyric acid type B receptors found in the ventral tegmental area in mice.
- Author
-
Leite-Morris KA, Fukudome EY, and Kaplan GB
- Subjects
- Animals, Baclofen pharmacology, Dopamine metabolism, Drug Interactions physiology, Feedback drug effects, Feedback physiology, GABA Agonists pharmacology, GABA-B Receptor Agonists, Immunohistochemistry, Male, Mice, Mice, Inbred C57BL, Morphine pharmacology, Motor Activity physiology, Neural Inhibition drug effects, Neural Inhibition physiology, Neural Pathways cytology, Neural Pathways drug effects, Neurons cytology, Neurons drug effects, Nucleus Accumbens cytology, Nucleus Accumbens drug effects, Proto-Oncogene Proteins c-fos drug effects, Proto-Oncogene Proteins c-fos metabolism, Synaptic Transmission drug effects, Synaptic Transmission physiology, Ventral Tegmental Area cytology, Ventral Tegmental Area drug effects, gamma-Aminobutyric Acid metabolism, Motor Activity drug effects, Narcotics pharmacology, Neural Pathways metabolism, Neurons metabolism, Nucleus Accumbens metabolism, Receptors, GABA-B metabolism, Ventral Tegmental Area metabolism
- Abstract
Recent studies suggest that gamma-aminobutyric acid type B (GABA(B)) receptors located on dopaminergic cells in the ventral tegmental area (VTA) regulate mesolimbic dopaminergic (A10) activity. In the current study, we identified GABA(B) receptor subtypes in the area of the VTA and examined their role in modulating acute opiate actions. We studied the effects of intra-VTA infusions of the selective GABA(B) agonist baclofen on morphine-induced locomotor stimulation and A10 neuronal activation. Drug treatments were followed by ambulatory activity monitoring for 180 min. Intra-VTA baclofen treatment produced a 70% inhibition of morphine-stimulated locomotor activity. Furthermore, functional activation of A10 neurons was assessed by immunohistochemical staining of c-Fos in the nucleus accumbens (NAc), where A10 neurons terminate. We found that morphine treatment increased the levels of Fos-positive nuclei in the NAc, while intra-VTA baclofen treatment reversed morphine's effects. Finally, GABA(B) receptor subtypes and isoforms were identified in the ventromedial mesencephalon using immunoblotting. We demonstrated the presence of GABA(B)R1a (130 kDa), GABA(B)R1b (100 kDa), and GABA(B)R2 (120 kDa) receptor subtypes in this region. These results suggest that GABA(B) receptor isoforms are found in the VTA and their activation results in the blockade of behavioral effects of opiates via inhibition of dopaminergic neurotransmission.
- Published
- 2002
- Full Text
- View/download PDF
45. Naltrexone's effects on reactivity to alcohol cues among alcoholic men.
- Author
-
Rohsenow DJ, Monti PM, Hutchison KE, Swift RM, Colby SM, and Kaplan GB
- Subjects
- Adult, Alcohol Drinking adverse effects, Alcohol Drinking psychology, Alcoholism psychology, Arousal drug effects, Attention drug effects, Cues, Humans, Male, Middle Aged, Substance Withdrawal Syndrome psychology, Taste drug effects, Alcoholism rehabilitation, Motivation, Naltrexone therapeutic use, Substance Withdrawal Syndrome diagnosis
- Abstract
The mechanisms of naltrexone's effects on urges to drink during abstinence are unclear. Naltrexone may suppress either urges to drink specifically or appetitive responses in general. The effects of naltrexone on cue reactivity to alcoholic and sweet nonalcoholic beverages were investigated. Alcohol-dependent men (N = 53) in treatment received naltrexone (50 mg) or placebo. Four hours later, they received baseline assessment, exposure to fruit juice, and exposure to their usual alcoholic beverage in 3-min trials. Naltrexone reduced urge to drink and self-reported attention to the alcohol cues, not at the initial exposure but after repeated exposures to alcohol cues. Naltrexone reduced negative affect across baseline and alcohol trials. No effects of naltrexone on responses to the nonalcoholic appetitive beverage cues were found, suggesting that general appetite suppression does not mediate the effects of naltrexone on urges.
- Published
- 2000
46. Differences in pharmacodynamics but not pharmacokinetics between subjects with panic disorder and healthy subjects after treatment with a single dose of alprazolam.
- Author
-
Kaplan GB, Greenblatt DJ, Ehrenberg BL, Goddard JE, Harmatz JS, and Shader RI
- Subjects
- Adult, Alprazolam therapeutic use, Anti-Anxiety Agents therapeutic use, Cross-Over Studies, Double-Blind Method, Electroencephalography drug effects, Female, Half-Life, Humans, Male, Panic Disorder psychology, Psychiatric Status Rating Scales, Time Factors, Alprazolam pharmacokinetics, Alprazolam pharmacology, Anti-Anxiety Agents pharmacokinetics, Anti-Anxiety Agents pharmacology, Panic Disorder drug therapy
- Abstract
The pharmacokinetics and pharmacodynamics of the benzodiazepine alprazolam (1 mg, administered orally) were compared between eight patients with panic disorder and eight age- and sex-matched healthy volunteers. Subjects received orally administered placebo and alprazolam in a randomized, double-blind, single-dose crossover study. The elimination half-life, time of maximum plasma concentration, maximum concentration, volume of distribution, and clearance of alprazolam were similar for both groups. For each cohort, alprazolam treatment (vs. placebo) produced significant changes in typical benzodiazepine agonist effects, such as increased sedation and impaired cognitive performance on the digit-symbol substitution test. For the panic disorder group only, there was a significant increase in the subjective rating of"contented" and a reduction in the rating of "easily irritated." For the healthy volunteer group, alprazolam produced increases in ratings of "fatigued" and "slowed thinking," but also increases in ratings of "relaxed." In each group, alprazolam significantly increased the electroencephalographic (EEG) measure of relative beta amplitude (range, 13-30 Hz) compared with placebo. Concentration-EEG response curves fit a sigmoid E(max) model, and there was greater sensitivity to EEG effects, as measured by a 28% reduction in the EC50 value, in the panic disorder group compared with healthy control subjects. After alprazolam treatment, there was increased sensitivity to EEG and mood effects and fewer aversive effects in the panic disorder group compared with healthy subjects. There were no differences in the pharmacodynamic measures of sedation and cognition or differences in pharmacokinetics between the two groups.
- Published
- 2000
- Full Text
- View/download PDF
47. Differential effects of treatment with typical and atypical antipsychotic drugs on adenylyl cyclase and G proteins.
- Author
-
Kaplan GB, Leite-Morris KA, and Keith DJ
- Subjects
- Adenylyl Cyclases metabolism, Animals, Benzodiazepines, Cerebral Cortex drug effects, Cerebral Cortex metabolism, Corpus Striatum drug effects, Corpus Striatum metabolism, GTP-Binding Proteins metabolism, Male, Mice, Olanzapine, Pirenzepine pharmacology, Adenylyl Cyclases drug effects, Antipsychotic Agents pharmacology, GTP-Binding Proteins drug effects, Guanosine 5'-O-(3-Thiotriphosphate) metabolism, Haloperidol pharmacology, Pirenzepine analogs & derivatives
- Abstract
We examined the effects of chronic in vivo antipsychotic drug treatments on G protein function and regulation. Mice were treated with typical antipsychotic haloperidol (6 mg/kg per day) and atypical agent olanzapine (20 mg/kg per day) for 14 days via mini-osmotic pumps. G protein-activated adenylyl cyclase activity in brain tissues was measured in the presence of guanine nucleotide analogue guanosine-5'-O(3-thiotriphosphate) tetralithium salt, or GTPgammaS. In frontal cortex, haloperidol treatment produced 21% increases in the GTPgammaS -mediated adenylyl cyclase Emax value (vs. vehicle controls) while olanzapine produced 20% reductions in this value (vs. controls); these effects were significant. In striatum, olanzapine treatment produced significant 31 and 27% decreases in Emax values compared with vehicle and haloperidol treatment, respectively. Chronic haloperidol treatment produced significant 24% reductions in the immunoreactivity of cortical, but not striatal, Gialpha1,2 subunits. There were no effects of chronic olanzapine treatment on G(i)alpha1,2 levels and no effects of either antipsychotic on G(s)alpha, levels. Chronic haloperidol and olanzapine treatments differentially regulate G protein-mediated adenylyl cyclase responses in brain regions possibly relating to their unique effects on G protein-coupled receptors.
- Published
- 1999
- Full Text
- View/download PDF
48. Role of adenosine A1 and A2A receptors in the alcohol withdrawal syndrome.
- Author
-
Kaplan GB, Bharmal NH, Leite-Morris KA, and Adams WR
- Subjects
- Adenosine therapeutic use, Animals, Brain drug effects, Brain metabolism, Central Nervous System Depressants administration & dosage, Ethanol administration & dosage, Male, Mice, Receptor, Adenosine A2A, Receptors, Purinergic P1 physiology, Substance Withdrawal Syndrome psychology, Thioinosine analogs & derivatives, Thioinosine metabolism, Adenosine analogs & derivatives, Central Nervous System Depressants adverse effects, Ethanol adverse effects, Phenethylamines therapeutic use, Purinergic P1 Receptor Agonists, Receptors, Purinergic P1 drug effects, Substance Withdrawal Syndrome drug therapy
- Abstract
The role of adenosine receptor-mediated signaling was examined in the alcohol withdrawal syndrome. CD-1 mice received a liquid diet containing ethanol (6.7%, v/v) or a control liquid diet that were abruptly discontinued after 14 days of treatment. Mice consuming ethanol showed a progressive increase in signs of intoxication throughout the drinking period. Following abrupt discontinuation of ethanol diet, mice demonstrated reversible signs of handling-induced hyperexcitability that were maximal between 5-8 h. Withdrawing mice received treatment with adenosine receptor agonists at the onset of peak withdrawal (5.5 h) and withdrawal signs were blindly rated (during withdrawal hours 6 and 7). Adenosine A1-receptor agonist R-N6(phenylisopropyl)adenosine (0.15 and 0.3 mg/ kg) reduced withdrawal signs 0.5 and 1.5 h after drug administration in a dose-dependent fashion. Adenosine A2A-selective agonist 2-p-(2-carboxyethyl)phenylethyl-amino-5'-N-ethylcarboxamidoadenosine (0.3 mg/kg) reduced withdrawal signs at both time points. In ethanol-withdrawing mice, there were significant decreases in adenosine transporter sites in striatum without changes in cortex or cerebellum. In ethanol-withdrawing mice, there were no changes in adenosine A1 and A2A receptor concentrations in cortex, striatum, or cerebellum. There appears to be a role for adenosine A1 and A2A receptors in the treatment of the ethanol withdrawal syndrome. Published by Elsevier Science Inc.
- Published
- 1999
- Full Text
- View/download PDF
49. Adenosine receptor antagonists inhibit the development of morphine sensitization in the C57BL/6 mouse.
- Author
-
Weisberg SP and Kaplan GB
- Subjects
- Adenosine antagonists & inhibitors, Animals, Caffeine pharmacology, Drug Combinations, Drug Resistance, Injections, Intraperitoneal, Male, Mice, Mice, Inbred C57BL, Morphine antagonists & inhibitors, Motor Activity drug effects, Xanthines pharmacology, Morphine pharmacology, Narcotics pharmacology, Purinergic P1 Receptor Antagonists
- Abstract
We examined the effects of adenosine antagonists on the development of morphine sensitization in C57BL/6 mice. Adenosine antagonists or vehicle were repeatedly co-administered intraperitoneally with morphine (10 mg/kg, s.c.) to mice once every other day for 9 days. Two days later, a 10 mg/kg morphine-only challenge was administered to each group. Consistent with sensitization, mice receiving morphine alone developed enhanced ambulatory activity responses to subsequent morphine administrations and, upon morphine-only challenge, had a significantly greater response to morphine than vehicle pretreated animals. The nonselective adenosine antagonist, caffeine, at 10 and 20 mg/kg but not at 5 mg/kg, attenuated the development of sensitization during co-administration with morphine and also following morphine-only challenge. The adenosine A1 selective antagonist 1,3-dipropyl-8-(2-amino-4-chlorophenyl)-xanthine (PACPX), at 0.001 and 0.002 mg/kg but not at 0.2 mg/kg, similarly attenuated the development of morphine sensitization. We propose a mechanism which involves an adenosine receptor role in the mesolimbic dopamine system.
- Published
- 1999
- Full Text
- View/download PDF
50. Adenosine kinase inhibitors attenuate opiate withdrawal via adenosine receptor activation.
- Author
-
Kaplan GB and Coyle TS
- Subjects
- 4-(3-Butoxy-4-methoxybenzyl)-2-imidazolidinone pharmacology, Animals, Body Weight drug effects, Caffeine pharmacology, Deoxyadenosines pharmacology, Dose-Response Relationship, Drug, Enzyme Inhibitors pharmacology, Male, Mice, Adenosine Kinase antagonists & inhibitors, Morphine pharmacology, Receptors, Purinergic P1 metabolism, Substance Withdrawal Syndrome prevention & control
- Abstract
Previous studies have demonstrated a role for adenosine in mediating opiate effects. This study examines the effects of indirect activation of adenosine receptors, via treatment with adenosine kinase inhibitors, on the expression of opiate withdrawal in mice. Mice receive chronic morphine treatment via implantation of subcutaneous morphine pellets (75 mg) for 72 h. Mice then receive parenteral treatment with adenosine kinase inhibitors, either 5'-amino-5'-deoxyadenosine (2, 5, 20, 40 mg/kg, intraperitoneal or i.p.) or iodotubericidin (1, 2, 5 mg/kg, i.p.), followed by naloxone injection and opiate withdrawal signs are measured over 20 min. Both adenosine kinase inhibitors significantly reduce the following opiate withdrawal signs in a dose-dependent manner compared to vehicle: withdrawal jumps, teeth chattering, forepaw tremors, and forepaw treads. Additionally, 5'-amino-5'-deoxyadenosine significantly reduces withdrawal-induced diarrhea and weight loss. Effects of 5'-amino-5'-deoxyadenosine (40 mg/kg) on opiate withdrawal signs appear to be mediated via adenosine receptor activation as they are reversed by pretreatment by adenosine receptor antagonist caffeine (20 mg, i.p.) but not by selective phosphodiesterase inhibitor Ro 20-1724 (10 mg/kg, i.p.). Adenosine receptor activation via adenosine kinase inhibitor treatment attenuates opiate withdrawal and these agents may be generally useful in the treatment of drug withdrawal syndromes.
- Published
- 1998
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.