1
|
Hedayati-Moghadam M, Razazpour F, Pourfridoni M, Mirzaee F, Baghcheghi Y. Ethanol's impact on the brain: a neurobiological perspective on the mechanisms of memory impairment. Mol Biol Rep 2024; 51:782. [PMID: 38918289 DOI: 10.1007/s11033-024-09748-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 06/21/2024] [Indexed: 06/27/2024]
Abstract
Alcohol consumption is known to have detrimental effects on memory function, with various studies implicating ethanol in the impairment of cognitive processes related to memory retention and retrieval. This review aims to elucidate the complex neurobiological mechanisms underlying ethanol-induced memory impairment. Through a thorough search of existing literature using electronic databases, relevant articles focusing on the neurobiological mechanisms of ethanol on memory were identified and critically evaluated. This review focuses on the molecular and neural pathways through which ethanol exerts its effects on memory formation, consolidation, and recall processes. Key findings from the included studies shed light on the impact of ethanol on neurotransmitter systems, synaptic plasticity, and neuroinflammation in relation to memory impairment. This review contributes to a better understanding of the intricate mechanisms by which alcohol impairs memory function, offering insights for future research directions and the development of targeted interventions to alleviate these cognitive impairments.
Collapse
Affiliation(s)
- Mahdiyeh Hedayati-Moghadam
- Department of Physiology, School of Medicine, Jiroft University of Medical Sciences, Jiroft, Iran
- Student Research Committee, Jiroft University of Medical Sciences, Jiroft, 7861755765, Iran
| | - Fateme Razazpour
- Student Research Committee, Jiroft University of Medical Sciences, Jiroft, 7861755765, Iran
| | - Mohammad Pourfridoni
- Student Research Committee, Jiroft University of Medical Sciences, Jiroft, 7861755765, Iran
| | - Faezeh Mirzaee
- Student Research Committee, Jiroft University of Medical Sciences, Jiroft, 7861755765, Iran
| | - Yousef Baghcheghi
- Student Research Committee, Jiroft University of Medical Sciences, Jiroft, 7861755765, Iran.
- Bio Environmental Health Hazards Research Center, Jiroft University of Medical Sciences, Jiroft, Iran.
| |
Collapse
|
2
|
Loetscher KB, Goldfarb EV. Integrating and fragmenting memories under stress and alcohol. Neurobiol Stress 2024; 30:100615. [PMID: 38375503 PMCID: PMC10874731 DOI: 10.1016/j.ynstr.2024.100615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 02/03/2024] [Accepted: 02/06/2024] [Indexed: 02/21/2024] Open
Abstract
Stress can powerfully influence the way we form memories, particularly the extent to which they are integrated or situated within an underlying spatiotemporal and broader knowledge architecture. These different representations in turn have significant consequences for the way we use these memories to guide later behavior. Puzzlingly, although stress has historically been argued to promote fragmentation, leading to disjoint memory representations, more recent work suggests that stress can also facilitate memory binding and integration. Understanding the circumstances under which stress fosters integration will be key to resolving this discrepancy and unpacking the mechanisms by which stress can shape later behavior. Here, we examine memory integration at multiple levels: linking together the content of an individual experience, threading associations between related but distinct events, and binding an experience into a pre-existing schema or sense of causal structure. We discuss neural and cognitive mechanisms underlying each form of integration as well as findings regarding how stress, aversive learning, and negative affect can modulate each. In this analysis, we uncover that stress can indeed promote each level of integration. We also show how memory integration may apply to understanding effects of alcohol, highlighting extant clinical and preclinical findings and opportunities for further investigation. Finally, we consider the implications of integration and fragmentation for later memory-guided behavior, and the importance of understanding which type of memory representation is potentiated in order to design appropriate interventions.
Collapse
Affiliation(s)
| | - Elizabeth V. Goldfarb
- Department of Psychiatry, Yale University, USA
- Department of Psychology, Yale University, USA
- Wu Tsai Institute, Yale University, USA
- National Center for PTSD, West Haven VA, USA
| |
Collapse
|
3
|
Lonnberg A, Logrip ML, Kuznetsov A. Mechanisms of alcohol influence on fear conditioning: a computational model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.30.573310. [PMID: 38260700 PMCID: PMC10802259 DOI: 10.1101/2023.12.30.573310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
A connection between stress-related illnesses and alcohol use disorders is extensively documented. Fear conditioning is a standard procedure used to study stress learning and links it to the activation of amygdala circuitry. However, the connection between the changes in amygdala circuit and function induced by alcohol and fear conditioning is not well established. We introduce a computational model to test the mechanistic relationship between amygdala functional and circuit adaptations during fear conditioning and the impact of acute vs. repeated alcohol exposure. In accordance with experiments, both acute and prior repeated alcohol decreases speed and robustness of fear extinction in our simulations. The model predicts that, first, the delay in fear extinction in alcohol is mostly induced by greater activation of the basolateral amygdala (BLA) after fear acquisition due to alcohol-induced modulation of synaptic weights. Second, both acute and prior repeated alcohol shifts the amygdala network away from the robust extinction regime by inhibiting the activity in the central amygdala (CeA). Third, our model predicts that fear memories formed in acute or after chronic alcohol are more connected to the context. Thus, the model suggests how circuit changes induced by alcohol may affect fear behaviors and provides a framework for investigating the involvement of multiple neuromodulators in this neuroadaptive process.
Collapse
Affiliation(s)
- Adam Lonnberg
- University of Evansville, Department of Mathematics, Indianapolis, Indiana, USA
| | - Marian L. Logrip
- Indiana University-Purdue University, Department of Psychology, Indianapolis, Indiana, USA
| | - Alexey Kuznetsov
- Indiana University-Purdue University, Department of Mathematical Sciences, Indianapolis, Indiana, USA
| |
Collapse
|
4
|
Seemiller LR, Garcia-Trevizo P, Novoa C, Goldberg LR, Murray S, Gould TJ. Adolescent intermittent alcohol exposure produces strain-specific cross-sensitization to nicotine and other behavioral adaptations in adulthood in C57BL/6J and DBA/2J mice. Pharmacol Biochem Behav 2023; 232:173655. [PMID: 37802393 PMCID: PMC10995114 DOI: 10.1016/j.pbb.2023.173655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/02/2023] [Accepted: 10/03/2023] [Indexed: 10/10/2023]
Abstract
Adolescent alcohol exposure is associated with lasting behavioral changes in humans and in mice. Prior work from our laboratory and others have demonstrated that C57BL/6J and DBA/2J mice differ in sensitivity to some effects of acute alcohol exposure during adolescence and adulthood. However, it is unknown if these strains differ in cognitive, anxiety-related, and addiction-related long-term consequences of adolescent intermittent alcohol exposure. This study examined the impact of a previously validated adolescent alcohol exposure paradigm (2-3 g/kg, i.p., every other day PND 30-44) in C57BL/6J and DBA/2J male and female mice on adult fear conditioning, anxiety-related behavior (elevated plus maze), and addiction-related phenotypes including nicotine sensitivity (hypothermia and locomotor depression) and alcohol sensitivity (loss of righting reflex; LORR). Both shared and strain-specific long-term consequences of adolescent alcohol exposure were found. Most notably, we found a strain-specific alcohol-induced increase in sensitivity to nicotine's hypothermic effects during adulthood in the DBA/2J strain but not in the C57BL/6J strain. Conversely, both strains demonstrated a robust increased latency to LORR during adulthood after adolescent alcohol exposure. Thus, we observed strain-dependent cross-sensitization to nicotine and strain-independent tolerance to alcohol due to adolescent alcohol exposure. Several strain and sex differences independent of adolescent alcohol treatment were also observed. These include increased sensitivity to nicotine-induced hypothermia in the C57BL/6J strain relative to the DBA/2J strain, in addition to DBA/2J mice showing more anxiety-like behaviors in the elevated plus maze relative to the C57BL/6J strain. Overall, these results suggest that adolescent alcohol exposure results in altered adult sensitivity to nicotine and alcohol with some phenotypes mediated by genetic background.
Collapse
Affiliation(s)
- Laurel R Seemiller
- Department of Biobehavioral Health, Penn State University, University Park, PA, USA
| | | | - Carlos Novoa
- Department of Biobehavioral Health, Penn State University, University Park, PA, USA
| | - Lisa R Goldberg
- Department of Biobehavioral Health, Penn State University, University Park, PA, USA
| | - Samantha Murray
- Department of Biobehavioral Health, Penn State University, University Park, PA, USA
| | - Thomas J Gould
- Department of Biobehavioral Health, Penn State University, University Park, PA, USA.
| |
Collapse
|
5
|
Seemiller LR, Goldberg LR, Garcia-Trevizo P, Gould TJ. Interstrain differences in adolescent fear conditioning after acute alcohol exposure. Brain Res Bull 2023; 194:35-44. [PMID: 36681252 PMCID: PMC10921434 DOI: 10.1016/j.brainresbull.2023.01.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 01/03/2023] [Accepted: 01/17/2023] [Indexed: 01/20/2023]
Abstract
Adolescent sensitivity to alcohol is a predictor of continued alcohol use and misuse later in life. Thus, it is important to understand the many factors that can impact alcohol sensitivity. Data from our laboratory suggested that susceptibility to alcohol-associated contextual fear learning deficits varied among adolescent and adult mice from two mouse strains. To investigate the extent of genetic background's influences on adolescent learning after alcohol exposure, we examined how 9 inbred mouse strains differed in vulnerability to alcohol-induced contextual and cued fear conditioning deficits. We demonstrated significant strain- and sex-dependent effects of acute alcohol exposure on adolescent fear learning, with alcohol having most pronounced effects on contextual fear learning. Female adolescents were more susceptible than males to alcohol-induced impairments in contextual, but not cued, fear learning, independent of genetic background. Heritability for contextual and cued fear learning after alcohol exposure was estimated to be 31 % and 18 %, respectively. Learning data were compared to Blood Ethanol Concentrations (BEC) to assess whether strain differences in alcohol metabolism contributed to strain differences in learning after alcohol exposure. There were no clear relationships between BEC and learning outcomes, suggesting that strains differed in learning outcomes for reasons other than strain differences in alcohol metabolism. Genetic analyses revealed polymorphisms across strains in notable genes, such as Chrna7, a promising genetic candidate for susceptibility to alcohol-induced fear conditioning deficits. These results are the first to demonstrate the impact of genetic background on alcohol-associated fear learning deficits during adolescence and suggest that the mechanisms underlying this sensitivity are distinct from alcohol metabolism.
Collapse
Affiliation(s)
- Laurel R Seemiller
- Department of Biobehavioral Health, Penn State University, University Park, PA, USA
| | - Lisa R Goldberg
- Department of Biobehavioral Health, Penn State University, University Park, PA, USA
| | | | - Thomas J Gould
- Department of Biobehavioral Health, Penn State University, University Park, PA, USA.
| |
Collapse
|
6
|
Marsland P, Parrella A, Vore AS, Barney TM, Varlinskaya EI, Deak T. Male, but not female, Sprague Dawley rats display enhanced fear learning following acute ethanol withdrawal (hangover). Pharmacol Biochem Behav 2021; 208:173229. [PMID: 34246729 PMCID: PMC9204503 DOI: 10.1016/j.pbb.2021.173229] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 07/06/2021] [Accepted: 07/07/2021] [Indexed: 12/13/2022]
Abstract
The present studies investigated the effects of withdrawal from a single binge-like dose of ethanol (hangover) on fear conditioning in male and female Sprague Dawley rats. In Experiment 1, males and females were given 0 or 3.5 g/kg ethanol intraperitoneally (i.p.) and then conditioned to contextual fear 24 h post injection. Withdrawal from acute ethanol enhanced expression of the conditioned freezing response in males, but not in females. Experiment 2 demonstrated that in males, withdrawal from acute ethanol administered 24 h prior to conditioning enhanced contextual fear conditioning, but not auditory-cued fear conditioning. In Experiment 3, male and female rats were given 3.5 g/kg ethanol, and blood ethanol concentrations (BECs) were assessed at various time points for determination of ethanol clearance. Female rats cleared ethanol at a higher rate than males, with 10 h required for females and 14 for males to eliminate ethanol from their systems. Because females cleared ethanol faster than males, in Experiment 4, females were conditioned 18 h after ethanol administration to keep the interval between ethanol clearance and fear conditioning similar to that of males. Withdrawal from acute ethanol given 18 h prior to conditioning did not affect both contextual and auditory-cued fear conditioning in females. In summary, these results highlight sex differences in the impact of withdrawal from acute ethanol (hangover) on fear learning; suggesting that males are more sensitive to hangover-associated enhancement of negative affect than females.
Collapse
Affiliation(s)
- Paige Marsland
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton, NY 13902-6000, United States
| | - Allissa Parrella
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton, NY 13902-6000, United States
| | - Andrew S Vore
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton, NY 13902-6000, United States
| | - Thaddeus M Barney
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton, NY 13902-6000, United States
| | - Elena I Varlinskaya
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton, NY 13902-6000, United States
| | - Terrence Deak
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton, NY 13902-6000, United States.
| |
Collapse
|
7
|
Seemiller LR, Gould TJ. Adult and adolescent C57BL/6J and DBA/2J mice are differentially susceptible to fear learning deficits after acute ethanol or MK-801 treatment. Behav Brain Res 2021; 410:113351. [PMID: 33974921 PMCID: PMC8403488 DOI: 10.1016/j.bbr.2021.113351] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 04/19/2021] [Accepted: 05/06/2021] [Indexed: 12/31/2022]
Abstract
Ethanol and other drugs of abuse disrupt learning and memory processes, creating problems associated with drug use and addiction. Understanding individual factors that determine susceptibility to drug-induced cognitive deficits, such as genetic background, age, and sex, is important for prevention and treatment. Comparison of adolescent and adult mice of both sexes across inbred mouse strains can reveal age, sex, and genetic contributions to phenotypes. We treated adolescent and adult, male and female, C57BL/6J and DBA/2J inbred mice with ethanol (1 g/kg or 1.5 g/kg) or MK-801 (0.05 mg/kg or 0.1 mg/kg), an NMDA receptor antagonist, prior to fear conditioning training. Contextual and cued fear retention were tested one day and eight or nine days after training. After ethanol exposure, adult C57BL/6J mice experienced greater deficits in contextual learning than adult DBA/2J mice. C57BL/6 J adolescents were less susceptible to ethanol-induced contextual learning disruptions than C57BL/6J adults, and adolescent males of both strains exhibited greater ethanol-induced contextual learning deficits than adolescent females. After MK-801 exposure, adolescent C57BL/6J mice experienced more severe contextual learning deficits than adolescent DBA/2J mice. Both ethanol and MK-801 had greater effects on contextual learning than cued learning. Collectively, we demonstrate that genetic background contributes to contextual and cued learning outcomes after ethanol or MK-801 exposure. Further, we report age-dependent drug sensitivities that are strain-, sex-, and drug-specific, suggesting that age, sex, and genetic background interact to determine contextual and cued learning impairments after ethanol or MK-801 exposure.
Collapse
Affiliation(s)
- L R Seemiller
- Department of Biobehavioral Health, Penn State University, 219 Biobehavioral Health Building, University Park, PA, 16801, United States
| | - T J Gould
- Department of Biobehavioral Health, Penn State University, 219 Biobehavioral Health Building, University Park, PA, 16801, United States.
| |
Collapse
|
8
|
López‐Gambero AJ, Rodríguez de Fonseca F, Suárez J. Energy sensors in drug addiction: A potential therapeutic target. Addict Biol 2021; 26:e12936. [PMID: 32638485 DOI: 10.1111/adb.12936] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 06/10/2020] [Accepted: 06/15/2020] [Indexed: 01/05/2023]
Abstract
Addiction is defined as the repeated exposure and compulsive seek of psychotropic drugs that, despite the harmful effects, generate relapse after the abstinence period. The psychophysiological processes associated with drug addiction (acquisition/expression, withdrawal, and relapse) imply important alterations in neurotransmission and changes in presynaptic and postsynaptic plasticity and cellular structure (neuroadaptations) in neurons of the reward circuits (dopaminergic neuronal activity) and other corticolimbic regions. These neuroadaptation mechanisms imply important changes in neuronal energy balance and protein synthesis machinery. Scientific literature links drug-induced stimulation of dopaminergic and glutamatergic pathways along with presence of neurotrophic factors with alterations in synaptic plasticity and membrane excitability driven by metabolic sensors. Here, we provide current knowledge of the role of molecular targets that constitute true metabolic/energy sensors such as AMPK, mTOR, ERK, or KATP in the development of the different phases of addiction standing out the main brain regions (ventral tegmental area, nucleus accumbens, hippocampus, and amygdala) constituting the hubs in the development of addiction. Because the available treatments show very limited effectiveness, evaluating the drug efficacy of AMPK and mTOR specific modulators opens up the possibility of testing novel pharmacotherapies for an individualized approach in drug abuse.
Collapse
Affiliation(s)
- Antonio Jesús López‐Gambero
- Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga Universidad de Málaga Málaga Spain
| | - Fernando Rodríguez de Fonseca
- Instituto de Investigación Biomédica de Málaga (IBIMA), UGC Salud Mental Hospital Regional Universitario de Málaga Málaga Spain
| | - Juan Suárez
- Instituto de Investigación Biomédica de Málaga (IBIMA), UGC Salud Mental Hospital Regional Universitario de Málaga Málaga Spain
| |
Collapse
|
9
|
Morningstar MD, Linsenbardt DN, Lapish CC. Ethanol Alters Variability, But Not Rate, of Firing in Medial Prefrontal Cortex Neurons of Awake-Behaving Rats. Alcohol Clin Exp Res 2020; 44:2225-2238. [PMID: 32966634 PMCID: PMC7680402 DOI: 10.1111/acer.14463] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 09/12/2020] [Indexed: 11/26/2022]
Abstract
BACKGROUND The medial prefrontal cortex (mPFC) is a brain region involved in the evaluation and selection of motivationally relevant outcomes. Neural activity in mPFC is altered following acute ethanol (EtOH) use and, in rodent models, doses as low as 0.75 g/kg yield cognitive deficits. Deficits in decision making following acute EtOH are thought to be mediated, at least in part, by decreases in mPFC firing rates (FRs). However, the data leading to this conclusion have been generated exclusively in anesthetized rodents. The present study characterizes the effects of acute EtOH injections on mPFC neural activity in awake-behaving rodents. METHODS Awake-behaving and anesthetized in vivo electrophysiological recordings were performed. We utilized 3 groups: the first received 2 saline injections, the second received a saline injection followed by 1.0 g/kg EtOH, and the last received saline followed by 2 g/kg EtOH. One week later, an anesthetized recording occurred where a saline injection was followed by an injection of 1.0 g/kg EtOH. RESULTS The anesthetized condition showed robust decreases in neural activity and differences in up-down states (UDS) dynamics. In the awake-behaving condition, FRs were grouped according to behavioral state: moving, not-moving, and sleep. The differences in median FRs were found for each treatment and behavioral state combination. A FR decrease was only found in the 2.0 g/kg EtOH treatment during not-moving states. However, robust decreases in FR variability were found across behavioral state in both the 1.0 and 2.0 g/kg EtOH treatment. Sleep was separately analyzed. EtOH modulated the UDS during sleep producing decreases in FRs. CONCLUSIONS In conclusion, the changes in neural activity following EtOH administration in anesthetized animals are not conserved in awake-behaving animals. The most prominent difference following EtOH was a decrease in FR variability suggesting that acute EtOH may be affecting decision making via this mechanism.
Collapse
|
10
|
Young CK, McNaughton N. Mixed Effects of Low-dose Ethanol on Cortical and Hippocampal Theta Oscillations. Neuroscience 2020; 429:213-224. [DOI: 10.1016/j.neuroscience.2020.01.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 01/02/2020] [Accepted: 01/06/2020] [Indexed: 12/14/2022]
|
11
|
Balachandran T, Cohen G, Le Foll B, Rehm J, Hassan AN. The effect of pre-existing alcohol use disorder on the risk of developing posttraumatic stress disorder: results from a longitudinal national representative sample. THE AMERICAN JOURNAL OF DRUG AND ALCOHOL ABUSE 2019; 46:232-240. [PMID: 31860361 DOI: 10.1080/00952990.2019.1690495] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Background: There is inconsistent evidence in the literature as to whether or not Alcohol Use Disorder (AUD) is a risk factor for Post-Traumatic Stress Disorder (PTSD).Objectives: We evaluated the risk of developing PTSD after trauma exposure in individuals with AUD. As a secondary analysis, we also tested if alcohol dependence or alcohol abuse separately increased the risk of PTSD development. We also explored the effect of AUD on exposure to various traumas.Methods: Longitudinal data was obtained from 30,180 individuals with and without AUD from National Epidemiologic Survey on Alcohol and Related Conditions (NESARC) waves I and II. Using propensity score methods, we matched individuals with AUD (alcohol abuse and/or dependence using DSM-IV criteria) to those without AUD at baseline on demographic, familial, and clinical factors to estimate the risk of PTSD development after trauma exposure. Data were adjusted for complex survey methods.Results: Individuals with AUD had an increased risk of being exposed to various traumas between wave I and II (60.6% vs. 48.3% of controls). Among individuals exposed to trauma between the two waves (N = 14,107), AUD had no effect on subsequent PTSD development after matching and controlling for covariates (OR: 1.00; 95%CI: 0.72-1.39; p = .99). However, those with alcohol dependence only did have an effect on subsequent PTSD development (OR: 1.76; 95%CI: 1.05-2.95; p = .03).Conclusion: In individuals with alcohol dependence the experience of trauma increases the risk of developing PTSD. These findings suggest that prevention methods from PTSD after trauma exposure for individuals with alcohol dependence are needed.
Collapse
Affiliation(s)
- Thaneson Balachandran
- Addictions Division, Centre for Addiction and Mental Health, Toronto, Ontario, Canada.,Department of Science, University of Toronto at Scarborough, Toronto, Canada
| | - Gregory Cohen
- Addictions Division, Centre for Addiction and Mental Health, Toronto, Ontario, Canada.,Department of Psychiatry, University of Toronto, Toronto, Canada
| | - Bernard Le Foll
- Addictions Division, Centre for Addiction and Mental Health, Toronto, Ontario, Canada.,Departments of Family and Community Medicine, Pharmacology and Toxicology, University of Toronto, Toronto, Canada.,Department of Psychiatry, Campbell Family Mental Health Research Institute, CAMH, Toronto, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Jürgen Rehm
- Department of Psychiatry, University of Toronto, Toronto, Canada.,Department of Psychiatry, Campbell Family Mental Health Research Institute, CAMH, Toronto, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada.,Centre for Addiction and Mental Health, Institute for Mental Health Policy Research, Toronto, Ontario, Canada.,Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Ahmed N Hassan
- Addictions Division, Centre for Addiction and Mental Health, Toronto, Ontario, Canada.,Department of Psychiatry, University of Toronto, Toronto, Canada.,Department of Psychiatry, King Abdul-Aziz University, Jeddah, Saudi Arabia
| |
Collapse
|
12
|
Van Skike CE, Goodlett C, Matthews DB. Acute alcohol and cognition: Remembering what it causes us to forget. Alcohol 2019; 79:105-125. [PMID: 30981807 DOI: 10.1016/j.alcohol.2019.03.006] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Revised: 03/14/2019] [Accepted: 03/18/2019] [Indexed: 12/17/2022]
Abstract
Addiction has been conceptualized as a specific form of memory that appropriates typically adaptive neural mechanisms of learning to produce the progressive spiral of drug-seeking and drug-taking behavior, perpetuating the path to addiction through aberrant processes of drug-related learning and memory. From that perspective, to understand the development of alcohol use disorders, it is critical to identify how a single exposure to alcohol enters into or alters the processes of learning and memory, so that involvement of and changes in neuroplasticity processes responsible for learning and memory can be identified early. This review characterizes the effects produced by acute alcohol intoxication as a function of brain region and memory neurocircuitry. In general, exposure to ethanol doses that produce intoxicating effects causes consistent impairments in learning and memory processes mediated by specific brain circuitry, whereas lower doses either have no effect or produce a facilitation of memory under certain task conditions. Therefore, acute ethanol does not produce a global impairment of learning and memory, and can actually facilitate particular types of memory, perhaps particular types of memory that facilitate the development of excessive alcohol use. In addition, the effects on cognition are dependent on brain region, task demands, dose received, pharmacokinetics, and tolerance. Additionally, we explore the underlying alterations in neurophysiology produced by acute alcohol exposure that help to explain these changes in cognition and highlight future directions for research. Through understanding the impact that acute alcohol intoxication has on cognition, the preliminary changes potentially causing a problematic addiction memory can better be identified.
Collapse
Affiliation(s)
- Candice E Van Skike
- Department of Cellular and Integrative Physiology and The Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78245, United States
| | - Charles Goodlett
- Department of Psychology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202, United States
| | - Douglas B Matthews
- Division of Psychology, University of Wisconsin - Eau Claire, Eau Claire, WI, 54702, United States.
| |
Collapse
|
13
|
Liu M, Yang S, Yang J, Lee Y, Kou J, Wang C. Neuroprotective and Memory-Enhancing Effects of Antioxidant Peptide From Walnut (Juglans regia L.) Protein Hydrolysates. Nat Prod Commun 2019. [DOI: 10.1177/1934578x19865838] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Peptides have been reported to possess interesting biological properties. The present study was designed to evaluate neuroprotective and memory-enhancing effects of antioxidant peptide from walnut ( Juglans regia L.) protein hydrolysates. The neuroprotective effect of walnut peptide (WP) against oxidative stress on PC12 cells was evaluated. And zebrafish was used as the model to corroborate the effect. Its effect on learning and memory of mice using the Morris water maze and the step-down passive avoidance tests were performed. Moreover, the acute toxicity of WP was carried out to assess its safety profile. It was found that WP was able to suppress H2O2-induced cell death in PC12 cells. In the zebrafish model, WP had an obvious neuroprotective effect, and the ratio reached 42% at 222 µg/mL. The mechanism study revealed that WP could inhibit the activity of caspases 3/7 and 8, reduce the mRNA expression levels of Bax and glial cell line-derived neurotrophic factor, and improve the mRNA expression level of brain-derived neurotrophic factor significantly. Besides, the treatment of mice with WP shortened the escape latency and exhibited much longer target time and more crossing times significantly, compared with untreated control groups in the Morris water maze test. Similarly, the step-down passive avoidance test showed that WP could ameliorate memory impairments. The administrated dose (20.1 g/kg body weight [BW]) did not produce mortality or treatment-related adverse effects with regard to BW, general behavior, or relative organ weights of the tested male and female mice. The current results indicated that WP could exert neuroprotective effect, and attenuated learning and memory impairments. These ameliorating effects of WP may be useful for treatment of memory impairment in Alzheimer’s and its related diseases.
Collapse
Affiliation(s)
- Mingchuan Liu
- R&D Center, Sinphar Tian-Li Pharmaceutical Co., Ltd., Hangzhou, China
| | - Shengjie Yang
- R&D Center, Sinphar Tian-Li Pharmaceutical Co., Ltd., Hangzhou, China
| | - Jinping Yang
- R&D Center, Sinphar Tian-Li Pharmaceutical Co., Ltd., Hangzhou, China
| | - Yita Lee
- R&D Center, Sinphar Pharmaceutical Co., Ltd., Ilan, Republic of China
| | - Junping Kou
- Department of Complex Prescription of TCM, China Pharmaceutical University, Nanjing, China
| | - Chaojih Wang
- R&D Center, Sinphar Tian-Li Pharmaceutical Co., Ltd., Hangzhou, China
- R&D Center, Sinphar Pharmaceutical Co., Ltd., Ilan, Republic of China
| |
Collapse
|
14
|
Williams AR, Lattal KM. Rapid reacquisition of contextual fear following extinction in mice: effects of amount of extinction, acute ethanol withdrawal, and ethanol intoxication. Psychopharmacology (Berl) 2019; 236:491-506. [PMID: 30338488 PMCID: PMC6374192 DOI: 10.1007/s00213-018-5057-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 09/28/2018] [Indexed: 12/16/2022]
Abstract
RATIONALE Many studies have found that ethanol intoxication and withdrawal impair initial acquisition or extinction of learned behaviors. Rapid reconditioning following extinction is a form of post-extinction re-emergence of conditioned behavior that has not been studied for its interaction with ethanol intoxication or withdrawal. OBJECTIVES The goals of this paper were to define the parameters that allow rapid post-extinction reacquisition of fear in mice and investigate the effect of acute ethanol withdrawal and intoxication on acquisition, extinction, and post-extinction reconditioning. METHODS We examined acquisition, extinction, and post-extinction reconditioning of contextual fear in male C57BL/6 mice. Acute ethanol withdrawal occurred 6 h following a 4 g/kg injection of 20% ethanol and acute ethanol intoxication occurred 5 min following a 1.5 g/kg injection of 20% ethanol. RESULTS A weak context-shock pairing caused rapid reacquisition of conditioned freezing following moderate, but not extensive extinction. Acute ethanol intoxication impaired initial conditioning and acute ethanol withdrawal impaired rapid reacquisition after extinction, but not reconditioning or extinction itself. CONCLUSIONS These findings show that rapid reconditioning occurs following moderate but not extensive extinction in C57BL/6J mice. Additionally, acute ethanol withdrawal and intoxication may differentially affect different phases of conditioning. Results are discussed in terms of current ideas about post-extinction behavior and ethanol's effects on memory.
Collapse
Affiliation(s)
- Amy R Williams
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97210, USA
| | - K Matthew Lattal
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97210, USA.
| |
Collapse
|
15
|
Hitchcock LN, Raybuck JD, Wood MA, Lattal KM. Effects of a histone deacetylase 3 inhibitor on extinction and reinstatement of cocaine self-administration in rats. Psychopharmacology (Berl) 2019; 236:517-529. [PMID: 30488346 PMCID: PMC6459190 DOI: 10.1007/s00213-018-5122-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 11/12/2018] [Indexed: 01/02/2023]
Abstract
RATIONALE A challenge in treating substance use disorder is that successful treatment often does not persist, resulting in relapse and continued drug seeking. One approach to persistently weaken drug-seeking behaviors is to pair exposure to drug-associated cues or behaviors with delivery of a compound that may strengthen the inhibition of the association between drug cues and behavior. OBJECTIVES We evaluated whether a selective histone deacetylase 3 (HDAC3) inhibitor could promote extinction and weaken contextual control of operant drug seeking after intravenous cocaine self-administration. METHODS Male Long-Evans rats received a systemic injection of the HDAC3 inhibitor RGFP966 either before or immediately after the first extinction session. Persistence of extinction was tested over subsequent extinction sessions, as well as tests of reinstatement that included cue-induced reinstatement, contextual renewal, and cocaine-primed reinstatement. Additional extinction sessions occurred between each reinstatement test. We also evaluated effects of RGFP966 on performance and motivation during stable fixed ratio operant responding for cocaine and during a progressive ratio of reinforcement. RESULTS RGFP966 administered before the first extinction session led to significantly less responding during subsequent extinction and reinstatement tests compared to vehicle-injected rats. Follow-up studies found that these effects were not likely due to a performance deficit or a change in motivation to self-administer cocaine, as injections of RGFP966 had no effect on stable responding during a fixed or progressive ratio schedule. In addition, RGFP966 administered just after the first extinction session had no effect during early extinction and reinstatement tests, but weakened long-term responding during later extinction sessions. CONCLUSIONS These results suggest that a systemic injection of a selective HDAC3 inhibitor can enhance extinction and suppress reinstatement after cocaine self-administration. The finding that behavioral and pharmacological manipulations can be combined to decrease drug seeking provides further potential for treatment by epigenetic modulation.
Collapse
Affiliation(s)
- Leah N. Hitchcock
- Department of Behavioral Neuroscience, Oregon Health & Science University
| | | | - Marcelo A. Wood
- Department of Neurobiology and Behavior, University of California, Irvine
| | - K. Matthew Lattal
- Department of Behavioral Neuroscience, Oregon Health & Science University
| |
Collapse
|
16
|
Sun W, Li X, Tang C, An L. Acute Low Alcohol Disrupts Hippocampus-Striatum Neural Correlate of Learning Strategy by Inhibition of PKA/CREB Pathway in Rats. Front Pharmacol 2018; 9:1439. [PMID: 30574089 PMCID: PMC6291496 DOI: 10.3389/fphar.2018.01439] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 11/22/2018] [Indexed: 11/30/2022] Open
Abstract
The hippocampus and striatum guide place-strategy and response-strategy learning, respectively, and they have dissociable roles in memory systems, which could compensate in case of temporary or permanent damage. Although acute alcohol (AA) treatment had been shown to have adverse effects on hippocampal function, whether it causes the functional compensation and the underlying mechanisms is unknown. In this study, rats treated with a low dose of AA avoided a hippocampus-dependent spatial strategy, instead preferring a striatum-dependent response strategy. Consistently, the learning-induced increase in hippocampal, but not striatal, pCREB was rendered less pronounced due to diminished activity of pPKA, but not pERK or pCaMKII. As rats approached the turn-decision area, Sp-cAMP, a PKA activator, was found to mitigate the inhibitory effect of AA on intra- and cross-structure synchronized neuronal oscillations, and rescue response-strategy bias and spatial learning deficits. Our study provides strong evidence of the critical link between neural couplings and strategy selection. Moreover, the PKA/CREB-signaling pathway is involved in the suppressive effect of AA on neural correlates of place-learning strategy. The novel important evidence provided here shows the functional couplings between the hippocampus and striatum in spatial learning processing and suggests possible avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Wei Sun
- Medical College of Acupuncture-Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaoliang Li
- Medical College of Acupuncture-Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chunzhi Tang
- Medical College of Acupuncture-Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lei An
- Medical College of Acupuncture-Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China.,College of Acupuncture-Moxibustion and Orthopedics, Guiyang University of Chinese Medicine, Guiyang, China.,Department of Physiology, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
17
|
Amorim RR, Silva PF, Luchiari AC. Effects of Alcohol on Inhibitory Avoidance Learning in Zebrafish (Danio rerio). Zebrafish 2017; 14:430-437. [DOI: 10.1089/zeb.2017.1438] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
| | | | - Ana Carolina Luchiari
- Departamento de Fisiologia, Universidade Federal do Rio Grande do Norte, Natal, Brazil
| |
Collapse
|
18
|
López-Cruz L, San-Miguel N, Bayarri P, Baqi Y, Müller CE, Salamone JD, Correa M. Ethanol and Caffeine Effects on Social Interaction and Recognition in Mice: Involvement of Adenosine A 2A and A 1 Receptors. Front Behav Neurosci 2016; 10:206. [PMID: 27853423 PMCID: PMC5090123 DOI: 10.3389/fnbeh.2016.00206] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 10/10/2016] [Indexed: 11/28/2022] Open
Abstract
Ethanol and caffeine are frequently consumed in combination and have opposite effects on the adenosine system: ethanol metabolism leads to an increase in adenosine levels, while caffeine is a non-selective adenosine A1/A2A receptor antagonist. These receptors are highly expressed in striatum and olfactory tubercle, brain areas involved in exploration and social interaction in rodents. Ethanol modulates social interaction processes, but the role of adenosine in social behavior is still poorly understood. The present work was undertaken to study the impact of ethanol, caffeine and their combination on social behavior, and to explore the involvement of A1 and A2A receptors on those actions. Male CD1 mice were evaluated in a social interaction three-chamber paradigm, for preference of conspecific vs. object, and also for long-term recognition memory of familiar vs. novel conspecific. Ethanol showed a biphasic effect, with low doses (0.25 g/kg) increasing social contact and higher doses (1.0-1.5 g/kg) reducing social interaction. However, no dose changed social preference; mice always spent more time sniffing the conspecific than the object, independently of the ethanol dose. Ethanol, even at doses that did not change social exploration, produced amnestic effects on social recognition the following day. Caffeine reduced social contact (15.0-60.0 mg/kg), and even blocked social preference at higher doses (30.0-60.0 mg/kg). The A1 antagonist Cyclopentyltheophylline (CPT; 3-9 mg/kg) did not modify social contact or preference on its own, and the A2A antagonist MSX-3 (1.5-6 mg/kg) increased social interaction at all doses. Ethanol at intermediate doses (0.5-1.0 g/kg) was able to reverse the reduction in social exploration induced by caffeine (15.0-30.0 mg/kg). Although there was no interaction between ethanol and CPT or MSX-3 on social exploration in the first day, MSX-3 blocked the amnestic effects of ethanol observed on the following day. Thus, ethanol impairs the formation of social memories, and A2A adenosine antagonists can prevent the amnestic effects of ethanol, so that animals can recognize familiar conspecifics. On the other hand, ethanol can counteract the social withdrawal induced by caffeine, a non-selective adenosine A1/A2A receptor antagonist. These results show the complex set of interactions between ethanol and caffeine, some of which could be the result of the opposing effects they have in modulating the adenosine system.
Collapse
Affiliation(s)
- Laura López-Cruz
- Àrea de Psicobiologia, Campus de Riu Sec, Universitat Jaume ICastelló, Spain
| | - Noemí San-Miguel
- Àrea de Psicobiologia, Campus de Riu Sec, Universitat Jaume ICastelló, Spain
| | - Pilar Bayarri
- Àrea de Psicobiologia, Campus de Riu Sec, Universitat Jaume ICastelló, Spain
| | - Younis Baqi
- Pharma-Zentrum Bonn, Pharmazeutisches Institut, Pharmazeutische Chemie, Universität BonnBonn, Germany
| | - Christa E. Müller
- Pharma-Zentrum Bonn, Pharmazeutisches Institut, Pharmazeutische Chemie, Universität BonnBonn, Germany
| | - John D. Salamone
- Department of Psychological Sciences, University of ConnecticutStorrs, CT, USA
| | - Mercé Correa
- Àrea de Psicobiologia, Campus de Riu Sec, Universitat Jaume ICastelló, Spain
- Department of Psychological Sciences, University of ConnecticutStorrs, CT, USA
| |
Collapse
|
19
|
Kutlu MG, Gould TJ. Effects of drugs of abuse on hippocampal plasticity and hippocampus-dependent learning and memory: contributions to development and maintenance of addiction. Learn Mem 2016; 23:515-33. [PMID: 27634143 PMCID: PMC5026208 DOI: 10.1101/lm.042192.116] [Citation(s) in RCA: 202] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 07/19/2016] [Indexed: 11/25/2022]
Abstract
It has long been hypothesized that conditioning mechanisms play major roles in addiction. Specifically, the associations between rewarding properties of drugs of abuse and the drug context can contribute to future use and facilitate the transition from initial drug use into drug dependency. On the other hand, the self-medication hypothesis of drug abuse suggests that negative consequences of drug withdrawal result in relapse to drug use as an attempt to alleviate the negative symptoms. In this review, we explored these hypotheses and the involvement of the hippocampus in the development and maintenance of addiction to widely abused drugs such as cocaine, amphetamine, nicotine, alcohol, opiates, and cannabis. Studies suggest that initial exposure to stimulants (i.e., cocaine, nicotine, and amphetamine) and alcohol may enhance hippocampal function and, therefore, the formation of augmented drug-context associations that contribute to the development of addiction. In line with the self-medication hypothesis, withdrawal from stimulants, ethanol, and cannabis results in hippocampus-dependent learning and memory deficits, which suggest that an attempt to alleviate these deficits may contribute to relapse to drug use and maintenance of addiction. Interestingly, opiate withdrawal leads to enhancement of hippocampus-dependent learning and memory. Given that a conditioned aversion to drug context develops during opiate withdrawal, the cognitive enhancement in this case may result in the formation of an augmented association between withdrawal-induced aversion and withdrawal context. Therefore, individuals with opiate addiction may return to opiate use to avoid aversive symptoms triggered by the withdrawal context. Overall, the systematic examination of the role of the hippocampus in drug addiction may help to formulate a better understanding of addiction and underlying neural substrates.
Collapse
Affiliation(s)
- Munir Gunes Kutlu
- Department of Biobehavioral Health, Penn State University, University Park, Pennsylvania 16802, USA
| | - Thomas J Gould
- Department of Biobehavioral Health, Penn State University, University Park, Pennsylvania 16802, USA
| |
Collapse
|
20
|
Tipps ME, Raybuck JD, Kozell LB, Lattal KM, Buck KJ. G Protein-Gated Inwardly Rectifying Potassium Channel Subunit 3 Knock-Out Mice Show Enhanced Ethanol Reward. Alcohol Clin Exp Res 2016; 40:857-64. [PMID: 27012303 PMCID: PMC4820358 DOI: 10.1111/acer.13012] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 01/07/2016] [Indexed: 12/17/2022]
Abstract
BACKGROUND G protein-gated inwardly rectifying potassium (GIRK) channels contribute to the effects of a number of drugs of abuse, including ethanol. However, the roles of individual subunits in the rewarding effects of ethanol are poorly understood. METHODS We compare conditioned place preference (CPP) in GIRK3 subunit knock-out (GIRK3(-/-)), heterozygote (GIRK3(+/-)), and wild-type (WT) mice. In addition, the development of locomotor tolerance/sensitization and the effects of EtOH intoxication on associative learning (fear conditioning) are also assessed. RESULTS Our data show significant EtOH CPP in GIRK3(-/-) and GIRK3(+/-) mice, but not in the WT littermates. In addition, we demonstrate that these effects are not due to differences in EtOH metabolism, the development of EtOH tolerance/sensitivity, or associative learning abilities. While there were no consistent genotype differences in the fear conditioning assay, our data do show a selective sensitization of the impairing effects of EtOH intoxication on contextual learning, but no effect on cued learning. CONCLUSIONS These findings suggest that GIRK3 plays a role in EtOH reward. Furthermore, the selectivity of this effect suggests that GIRK channels could be an effective therapeutic target for the prevention and/or treatment of alcoholism.
Collapse
Affiliation(s)
- Megan E. Tipps
- Portland Alcohol Research Center; Portland VA Medical Center, 3710 SW US Veterans Hospital Rd., Bldg 104, Portland, OR 97239-3098, USA
- Department of Behavioral Neuroscience; Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Portland, OR 97239-3098, USA
| | - Jonathan D. Raybuck
- Department of Behavioral Neuroscience; Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Portland, OR 97239-3098, USA
| | - Laura B. Kozell
- Portland Alcohol Research Center; Portland VA Medical Center, 3710 SW US Veterans Hospital Rd., Bldg 104, Portland, OR 97239-3098, USA
- Department of Behavioral Neuroscience; Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Portland, OR 97239-3098, USA
| | - K. Matthew Lattal
- Department of Behavioral Neuroscience; Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Portland, OR 97239-3098, USA
| | - Kari J. Buck
- Portland Alcohol Research Center; Portland VA Medical Center, 3710 SW US Veterans Hospital Rd., Bldg 104, Portland, OR 97239-3098, USA
- Department of Behavioral Neuroscience; Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Portland, OR 97239-3098, USA
| |
Collapse
|
21
|
Bisby JA, King JA, Sulpizio V, Degeilh F, Valerie Curran H, Burgess N. Extinction learning is slower, weaker and less context specific after alcohol. Neurobiol Learn Mem 2015; 125:55-62. [PMID: 26234587 PMCID: PMC4655873 DOI: 10.1016/j.nlm.2015.07.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 07/24/2015] [Indexed: 11/29/2022]
Abstract
Alcohol is frequently involved in psychological trauma and often used by individuals to reduce fear and anxiety. We examined the effects of alcohol on fear acquisition and extinction within a virtual environment. Healthy volunteers were administered alcohol (0.4 g/kg) or placebo and underwent acquisition and extinction from different viewpoints of a virtual courtyard, in which the conditioned stimulus, paired with a mild electric shock, was centrally located. Participants returned the following day to test fear recall from both viewpoints of the courtyard. Skin conductance responses were recorded as an index of conditioned fear. Successful fear acquisition under alcohol contrasted with impaired extinction learning evidenced by persistent conditioned responses (Experiment 1). Participants’ impairments in extinction under alcohol correlated with impairments in remembering object-locations in the courtyard seen from one viewpoint when tested from the other viewpoint. Alcohol-induced extinction impairments were overcome by increasing the number of extinction trials (Experiment 2). However, a test of fear recall the next day showed persistent fear in the alcohol group across both viewpoints. Thus, alcohol impaired extinction rather than acquisition of fear, suggesting that extinction is more dependent than acquisition on alcohol-sensitive representations of spatial context. Overall, extinction learning under alcohol was slower, weaker and less context-specific, resulting in persistent fear at test that generalized to the extinction viewpoint. The selective effect on extinction suggests an effect of alcohol on prefrontal involvement, while the reduced context-specificity implicates the hippocampus. These findings have important implications for the use of alcohol by individuals with clinical anxiety disorders.
Collapse
Affiliation(s)
- James A Bisby
- Institute of Cognitive Neuroscience, University College London, 17 Queen Square, London, WC1N 3AR, UK; Institute of Neurology, University College London, Queen Square, London, WC1 3BG, UK.
| | - John A King
- Institute of Cognitive Neuroscience, University College London, 17 Queen Square, London, WC1N 3AR, UK; Clinical, Education and Health Psychology, University College London, London, UK
| | - Valentina Sulpizio
- Department of Psychology, Sapienza University, Rome, Italy; Laboratory of Neuropsychology, Fondazione Santa Lucia IRCCS, Roma, Italy
| | - Fanny Degeilh
- Inserm-EPHE-UCBN, Unité U1077, Boulevard Becquerel, 14000 Caen, France
| | - H Valerie Curran
- Clinical Psychopharmacology Unit, University College London, London, UK; Clinical, Education and Health Psychology, University College London, London, UK
| | - Neil Burgess
- Institute of Cognitive Neuroscience, University College London, 17 Queen Square, London, WC1N 3AR, UK; Institute of Neurology, University College London, Queen Square, London, WC1 3BG, UK.
| |
Collapse
|
22
|
Tipps ME, Raybuck JD, Buck KJ, Lattal KM. Acute ethanol withdrawal impairs contextual learning and enhances cued learning. Alcohol Clin Exp Res 2015; 39:282-90. [PMID: 25684050 PMCID: PMC4331355 DOI: 10.1111/acer.12614] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Accepted: 10/24/2014] [Indexed: 11/29/2022]
Abstract
BACKGROUND Alcohol affects many of the brain regions and neural processes that support learning and memory, and these effects are thought to underlie, at least in part, the development of addiction. Although much work has been done regarding the effects of alcohol intoxication on learning and memory, little is known about the effects of acute withdrawal from a single alcohol exposure. METHODS We assess the effects of acute ethanol withdrawal (6 hours postinjection with 4 g/kg ethanol) on 2 forms of fear conditioning (delay and trace fear conditioning) in C57BL/6J and DBA/2J mice. The influence of a number of experimental parameters (pre- and post training withdrawal exposure; foreground/background processing; training strength; and nonassociative effects) is also investigated. RESULTS Acute ethanol withdrawal during training had a bidirectional effect on fear-conditioned responses, decreasing contextual responses and increasing cued responses. These effects were apparent for both trace and delay conditioning in DBA/2J mice and for trace conditioning in C57BL/6J mice; however, C57BL/6J mice were selectively resistant to the effects of acute withdrawal on delay cued responses. CONCLUSIONS Our results show that acute withdrawal from a single, initial ethanol exposure is sufficient to alter long-term learning in mice. In addition, the differences between the strains and conditioning paradigms used suggest that specific learning processes can be differentially affected by acute withdrawal in a manner that is distinct from the reported effects of both alcohol intoxication and withdrawal following chronic alcohol exposure. Thus, our results suggest a unique effect of acute alcohol withdrawal on learning and memory processes.
Collapse
Affiliation(s)
- Megan E. Tipps
- Portland Alcohol Research Center; Portland VA Medical Center 3710 SW US Veterans Hospital Rd., Bld 104 Portland, OR 97239-3098, USA
- Department of Behavioral Neuroscience; Oregon Health & Science University 3181 SW Sam Jackson Park Rd. Portland, OR 97239-3098, USA
| | - Jonathan D. Raybuck
- Department of Behavioral Neuroscience; Oregon Health & Science University 3181 SW Sam Jackson Park Rd. Portland, OR 97239-3098, USA
| | - Kari J. Buck
- Portland Alcohol Research Center; Portland VA Medical Center 3710 SW US Veterans Hospital Rd., Bld 104 Portland, OR 97239-3098, USA
- Department of Behavioral Neuroscience; Oregon Health & Science University 3181 SW Sam Jackson Park Rd. Portland, OR 97239-3098, USA
| | - K. Matthew Lattal
- Department of Behavioral Neuroscience; Oregon Health & Science University 3181 SW Sam Jackson Park Rd. Portland, OR 97239-3098, USA
| |
Collapse
|
23
|
Giannoni-Guzmán MA, Giray T, Agosto-Rivera JL, Stevison BK, Freeman B, Ricci P, Brown EA, Abramson CI. Ethanol-induced effects on sting extension response and punishment learning in the western honey bee (Apis mellifera). PLoS One 2014; 9:e100894. [PMID: 24988309 PMCID: PMC4079248 DOI: 10.1371/journal.pone.0100894] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Accepted: 05/30/2014] [Indexed: 11/19/2022] Open
Abstract
Acute ethanol administration is associated with sedation and analgesia as well as behavioral disinhibition and memory loss but the mechanisms underlying these effects remain to be elucidated. During the past decade, insects have emerged as important model systems to understand the neural and genetic bases of alcohol effects. However, novel assays to assess ethanol's effects on complex behaviors in social or isolated contexts are necessary. Here we used the honey bee as an especially relevant model system since bees are typically exposed to ethanol in nature when collecting standing nectar crop of flowers, and there is recent evidence for independent biological significance of this exposure for social behavior. Bee's inhibitory control of the sting extension response (SER) and a conditioned-place aversion assay were used to study ethanol effects on analgesia, behavioral disinhibition, and associative learning. Our findings indicate that although ethanol, in a dose-dependent manner, increases SER thresholds (analgesic effects), it disrupts the ability of honey bees to inhibit SER and to associate aversive stimuli with their environment. These results suggest that ethanol's effects on analgesia, behavioral disinhibition and associative learning are common across vertebrates and invertebrates. These results add to the use of honey bees as an ethanol model to understand ethanol's effects on complex, socially relevant behaviors.
Collapse
Affiliation(s)
| | - Tugrul Giray
- Department of Biology, University of Puerto Rico Rio Piedras, San Juan, Puerto Rico
| | | | - Blake K. Stevison
- Laboratory of Behavioral Biology and Comparative Psychology, Oklahoma State University, Stillwater, Oklahoma, United States of America
| | - Brett Freeman
- Laboratory of Behavioral Biology and Comparative Psychology, Oklahoma State University, Stillwater, Oklahoma, United States of America
| | - Paige Ricci
- Laboratory of Behavioral Biology and Comparative Psychology, Oklahoma State University, Stillwater, Oklahoma, United States of America
| | - Erika A. Brown
- Laboratory of Behavioral Biology and Comparative Psychology, Oklahoma State University, Stillwater, Oklahoma, United States of America
| | - Charles I. Abramson
- Laboratory of Behavioral Biology and Comparative Psychology, Oklahoma State University, Stillwater, Oklahoma, United States of America
| |
Collapse
|
24
|
Juran SA, Johanson G, Ernstgård L, Iregren A, van Thriel C. Neurobehavioral performance in volunteers after inhalation of white spirits with high and low aromatic content. Arch Toxicol 2014; 88:1127-40. [DOI: 10.1007/s00204-014-1236-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Accepted: 03/26/2014] [Indexed: 11/30/2022]
|
25
|
Sircar R, Ishiwari K. Systemic Administration of γ-Hydroxybutyric Acid in Adolescent Rat Impairs Contextual Fear Conditioning, But Not Cued Conditioning. ACTA ACUST UNITED AC 2014. [DOI: 10.4303/jdar/235801] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
26
|
Tipps ME, Raybuck JD, Lattal KM. Substance abuse, memory, and post-traumatic stress disorder. Neurobiol Learn Mem 2013; 112:87-100. [PMID: 24345414 DOI: 10.1016/j.nlm.2013.12.002] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Revised: 11/23/2013] [Accepted: 12/03/2013] [Indexed: 12/20/2022]
Abstract
A large body of literature demonstrates the effects of abused substances on memory. These effects differ depending on the drug, the pattern of delivery (acute or chronic), and the drug state at the time of learning or assessment. Substance use disorders involving these drugs are often comorbid with anxiety disorders, such as post-traumatic stress disorder (PTSD). When the cognitive effects of these drugs are considered in the context of the treatment of these disorders, it becomes clear that these drugs may play a deleterious role in the development, maintenance, and treatment of PTSD. In this review, we examine the literature evaluating the cognitive effects of three commonly abused drugs: nicotine, cocaine, and alcohol. These three drugs operate through both common and distinct neurobiological mechanisms and alter learning and memory in multiple ways. We consider how the cognitive and affective effects of these drugs interact with the acquisition, consolidation, and extinction of learned fear, and we discuss the potential impediments that substance abuse creates for the treatment of PTSD.
Collapse
Affiliation(s)
- Megan E Tipps
- Department of Behavioral Neuroscience, Oregon Health & Science University, United States.
| | - Jonathan D Raybuck
- Department of Behavioral Neuroscience, Oregon Health & Science University, United States.
| | - K Matthew Lattal
- Department of Behavioral Neuroscience, Oregon Health & Science University, United States.
| |
Collapse
|
27
|
Bray JG, Reyes KC, Roberts AJ, Ransohoff RM, Gruol DL. Synaptic plasticity in the hippocampus shows resistance to acute ethanol exposure in transgenic mice with astrocyte-targeted enhanced CCL2 expression. Neuropharmacology 2012; 67:115-25. [PMID: 23164616 DOI: 10.1016/j.neuropharm.2012.11.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Revised: 10/31/2012] [Accepted: 11/02/2012] [Indexed: 01/22/2023]
Abstract
It has been shown that ethanol exposure can activate astrocytes and microglia resulting in the production of neuroimmune factors, including the chemokine CCL2. The role of these neuroimmune factors in the effects of ethanol on the central nervous system has yet to be elucidated. To address this question, we investigated the effects of ethanol on synaptic transmission and plasticity in the hippocampus from mice that express elevated levels of CCL2 in the brain and their non-transgenic littermate controls. The brains of the transgenic mice simulate one aspect of the alcoholic brain, chronically increased levels of CCL2. We used extracellular field potential recordings in acutely isolated hippocampal slices to identify neuroadaptive changes produced by elevated levels of CCL2 and how these neuroadaptive changes affect the actions of acute ethanol. Results showed that synaptic transmission and the effects of ethanol on synaptic transmission were similar in the CCL2-transgenic and non-transgenic hippocampus. However, long-term potentiation (LTP), a cellular mechanism thought to underlie learning and memory, in the CCL2-transgenic hippocampus was resistant to the ethanol-induced depression of LTP observed in the non-transgenic hippocampus. Consistent with these results, ethanol pretreatment significantly impaired cued and contextual fear conditioning in non-transgenic mice, but had no effect in CCL2-transgenic mice. These data show that chronically elevated levels of CCL2 in the hippocampus produce neuroadaptive changes that block the depressing effects of ethanol on hippocampal synaptic plasticity and support the hypothesis that CCL2 may provide a neuroprotective effect against the devastating actions of ethanol on hippocampal function.
Collapse
Affiliation(s)
- Jennifer G Bray
- Molecular and Integrative Neuroscience Department, SP30-1522, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | | | | | | | | |
Collapse
|
28
|
RETRACTED: Acute ethanol administration affects memory reactivation: A look at the neuronal density and apoptosis in the rat hippocampus. Pharmacol Biochem Behav 2012; 102:321-8. [DOI: 10.1016/j.pbb.2012.04.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2011] [Revised: 04/10/2012] [Accepted: 04/21/2012] [Indexed: 11/16/2022]
|
29
|
Sanday L, Patti CL, Zanin KA, Fernandes-Santos L, Oliveira LC, Kameda SR, Tufik S, Frussa-Filho R. Ethanol-Induced Memory Impairment in a Discriminative Avoidance Task is State-Dependent. Alcohol Clin Exp Res 2012; 37 Suppl 1:E30-9. [DOI: 10.1111/j.1530-0277.2012.01905.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2011] [Accepted: 06/06/2012] [Indexed: 11/27/2022]
Affiliation(s)
- Leandro Sanday
- Departamento de Farmacologia; Universidade Federal de São Paulo; São Paulo; SP; Brasil
| | | | | | | | - Larissa C. Oliveira
- Departamento de Farmacologia; Universidade Federal de São Paulo; São Paulo; SP; Brasil
| | | | - Sergio Tufik
- Departamento de Psicobiologia; Universidade Federal de São Paulo; São Paulo; SP; Brasil
| | | |
Collapse
|
30
|
Burns BE, Proctor WR. Cigarette smoke exposure greatly increases alcohol consumption in adolescent C57BL/6 mice. Alcohol Clin Exp Res 2012; 37 Suppl 1:E364-72. [PMID: 22827559 DOI: 10.1111/j.1530-0277.2012.01911.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Accepted: 05/30/2012] [Indexed: 11/28/2022]
Abstract
BACKGROUND Alcohol and tobacco are often used together, and alcoholism is much more common among smokers compared with nonsmokers. Studies in humans suggest that nicotine (an active ingredient in cigarette smoke) can increase the consumption of alcohol. Research on rats and mice demonstrated mixed results; some studies report that nicotine increases alcohol consumption, while others show a decrease in drinking. Because cigarette smoke includes many other chemicals, these also may play a significant role in alcohol consumption. For example, 2 of these other constituents, monoamine oxidase inhibitors and acetaldehyde, increase alcohol tolerance and/or alcohol consumption in rodents. This study was designed to investigate how cigarette smoke from tobacco may modify self-administration of alcohol in adolescent C57BL/6 mice, a critical time when adolescent humans begin abusing drugs. METHODS C57BL/6 male mice (4 to 5 weeks old) were acclimated for 3 weeks to consume a 10% (w/v) alcohol solution during a 2-hour daily access in the dark. Subsequently, half the animals were exposed to cigarette smoke for 6 h/d for 16 days. The remaining animals (control) were placed in a smoke-free adjacent chamber. Immediately following the 6-hour period in the chambers, the control and smoke-exposed mice were given access to the 10% alcohol solution for 2 hours. RESULTS Animals exposed to cigarette smoke for 6 h/d consumed approximately 3- to 5-fold more alcohol than the mice in the control group throughout the 16-day study. The mice in the smoke group had a blood alcohol concentration that was nearly 4-fold that of the control mice. CONCLUSIONS Cigarette smoke increases alcohol consumption several fold higher than reported studies using nicotine treatment alone in adolescent rodents. Thus, this model should be useful to determine the roles of other bioactive components in cigarette smoke that may be important in the high co-abuse of smoking and alcohol consumption.
Collapse
Affiliation(s)
- Benjamin E Burns
- University of Colorado School of Medicine, Aurora, Colorado 80045, USA
| | | |
Collapse
|
31
|
Alijan-pour J, Abrari K, Lashkar bluki T, Ghorbanian MT, Goudarzi I, Elahdadi Salmani M. Ethanol disrupts reactivated contextual conditioned fear memory: behavioral and histological perspectives. CELL JOURNAL 2012; 13:265-74. [PMID: 23507995 PMCID: PMC3584476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2010] [Accepted: 08/13/2011] [Indexed: 11/27/2022]
Abstract
OBJECTIVE This research study is an attempt to examine whether the administration of ethanol after memory reactivation would modulate subsequent expression of memory in rats. Additionally, we examined whether this administration alters the density of Cornu Ammonis (CA)1 and CA3 pyramidal and dentate gyrus (DG) granule cells. MATERIALS AND METHODS In this experimental study, adult male Wistar rats (200-300 g) were trained in a fear conditioning system using two 1 second, 0.6 mA shocks with an interval of 180 seconds. Twenty four hours later rats were returned to the chamber for 120 seconds. Immediately after reactivation they were injected with ethanol (0.5, 1, 1.5 mg/ kg) or saline. 1, 7 and 14 days after reactivation, rats were returned to the context for 5 minutes. Seconds of freezing (absence of all movement except respiration) were scored. In the second experiment (described in the previous paragraph), after test 1, animals were anesthetized with sodium pentobarbital and perfused transcardially with phosphate buffer (10 minutes) and 4% paraformaldehyde (15 minutes). The brains were postfixed in phosphate-buffered 4% paraformaldehyde (24 hours) and 30% sucrose. 10-µm sections were stained with cresyl violet. Data were analyzed by 1-and 2-way ANOVA for repeated measurements by means of SPSS 16.0. Tukey's post hoc test was performed to determine the source of detected significant differences. P <0 .05 were considered significant. Data are presented as mean ± SEM. RESULTS Findings from the first experiment indicated that ethanol at a dose of 1.5 mg/kg significantly impaired recall of memory only in the first test. The density of CA1 and CA3 pyramidal and DG granule cells in the ethanol group was decreased (p< 0.01) compared with control group respectively 43.7%, 35.8%, and 37.8. CONCLUSION The data demonstrate that ethanol exposure impairs post retrieval processes. Moreover, ethanol decreases the density of CA1, CA3 and DG cells. Presumably it would be a correlation between our behavioral and histological results.
Collapse
Affiliation(s)
| | - Kataneh Abrari
- * Corresponding Address:
P.O.Box: 36715-364School of BiologyDamghan UniversityDamghanIran
| | | | | | | | | |
Collapse
|
32
|
Wang Y, Tang L, Feng X, Du W, Liu BF. Ethanol interferes with gustatory plasticity in Caenorhabditis elegans. Neurosci Res 2011; 71:341-7. [DOI: 10.1016/j.neures.2011.08.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2011] [Revised: 07/25/2011] [Accepted: 08/18/2011] [Indexed: 12/01/2022]
|
33
|
Barrenha GD, Coon LE, Chester JA. Effects of alcohol on the acquisition and expression of fear-potentiated startle in mouse lines selectively bred for high and low alcohol preference. Psychopharmacology (Berl) 2011; 218:191-201. [PMID: 21487654 PMCID: PMC3160503 DOI: 10.1007/s00213-011-2285-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2010] [Accepted: 03/25/2011] [Indexed: 11/26/2022]
Abstract
RATIONALE Anxiety disorders and alcohol use disorders frequently co-occur in humans perhaps because alcohol relieves anxiety. Studies in humans and rats indicate that alcohol may have greater anxiolytic effects in organisms with increased genetic propensity for high alcohol consumption. OBJECTIVES AND METHODS The purpose of this study was to investigate the effects of moderate doses of alcohol (0.5, 1.0, 1.5 g/kg) on the acquisition and expression of anxiety-related behavior using a fear-potentiated startle (FPS) procedure. Experiments were conducted in two replicate pairs of mouse lines selectively bred for high- (HAP1 and HAP2) and low- (LAP1 and LAP2) alcohol preference; these lines have previously shown a genetic correlation between alcohol preference and FPS (HAP > LAP; Barrenha and Chester, Alcohol Clin Exp Res 31:1081-1088, 2007). In a control experiment, the effect of diazepam (4.0 mg/kg) on the expression of FPS was tested in HAP2 and LAP2 mice. RESULTS The 1.5 g/kg alcohol dose moderately decreased the expression of FPS in both HAP lines but not LAP lines. Alcohol had no effect on the acquisition of FPS in any line. Diazepam reduced FPS to a similar extent in both HAP2 and LAP2 mice. CONCLUSIONS HAP mice may be more sensitive to the anxiolytic effects of alcohol than LAP mice when alcohol is given prior to the expression of FPS. These data collected in two pairs of HAP/LAP mouse lines suggest that the anxiolytic response to alcohol in HAP mice may be genetically correlated with their propensity toward high alcohol preference and robust FPS.
Collapse
Affiliation(s)
- Gustavo D Barrenha
- Department of Psychological Sciences, Purdue University, 703 Third Street, West Lafayette, IN 47907-2081, USA
| | | | | |
Collapse
|
34
|
Effect of acute ethanol and acute allopregnanolone on spatial memory in adolescent and adult rats. Alcohol 2011; 45:473-83. [PMID: 21600728 DOI: 10.1016/j.alcohol.2011.03.001] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2010] [Revised: 02/17/2011] [Accepted: 03/02/2011] [Indexed: 11/20/2022]
Abstract
The effects of ethanol differ in adolescent and adult rats on a number of measures. The evidence of the effects of ethanol on spatial memory in adolescents and adults is equivocal. Whether adolescents are more or less sensitive to ethanol-induced impairment of spatial memory acquisition remains unclear; with regard to the effects of acute ethanol on spatial memory retrieval there is almost no research looking into any age difference. Thus, we examined the effects of acute ethanol on spatial memory in the Morris Watermaze in adolescents and adults. Allopregnanolone (ALLO) is a modulator of the GABA(A) receptor and has similar behavioral effects as ethanol. We sought to also determine the effects of allopreganolone on spatial memory in adolescent and adults. Male adolescent (post natal [PN]28-30) and adult (PN70-72) rats were trained in the Morris Watermaze for 6 days and acute doses of ethanol (saline, 1.5 and 2.0 g/kg) or ALLO (vehicle, 9 and 18 mg/kg) were administered on Day 7. A probe trial followed on Day 8. As expected, there were dose effects; higher doses of both ethanol and ALLO impaired spatial memory. However, in both the ethanol and ALLO conditions adolescents and adults had similar spatial memory impairments. The current results suggest that ethanol and ALLO both impair hippocampal-dependent spatial memory regardless of age in that once learning has occurred, ethanol or ALLO does not differentially impair the retrieval of spatial memory in adolescents and adults. Given the mixed results on the effect of ethanol on cognition in adolescent rats, additional research is needed to ascertain the factors critical for the reported differential results.
Collapse
|
35
|
Yamada M, Hayashida M, Zhao Q, Shibahara N, Tanaka K, Miyata T, Matsumoto K. Ameliorative effects of yokukansan on learning and memory deficits in olfactory bulbectomized mice. JOURNAL OF ETHNOPHARMACOLOGY 2011; 135:737-746. [PMID: 21513784 DOI: 10.1016/j.jep.2011.04.010] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2010] [Revised: 03/18/2011] [Accepted: 04/05/2011] [Indexed: 05/30/2023]
Abstract
AIM OF THE STUDY Yokukansan (YKS) is a Japanese traditional herbal medicine and has been used for the treatment of the behavioral and psychological symptoms of dementia (BPSD). The present study aimed to clarify the effects of YKS on learning and memory impairments, and its mechanisms of action in olfactory bulbectomized (OBX) mice, one of the animal models of Alzheimer's disease (AD). MATERIALS AND METHODS OBX or sham-operated ddY mice were treated with YKS or donepezil (DPZ), a reference drug, and their cognitive performances were tested by the modified Y-maze test, novel object recognition test, and fear conditioning test to elucidate the spatial working memory, non-spatial short-term memory, and long-term memory, respectively. After completing the behavioral experiments, the expression level of cholinergic marker proteins and the activity of acetylcholinesterase (AChE) in the brain were analyzed by western blotting and Ellman's method, respectively. RESULTS OBX caused spatial working memory and non-spatial working memory impairments that were reversed by YKS and also by DPZ; however, YKS failed to affect the long-term memory deficits. Amelioration of the spatial working memory by YKS was reversible by scopolamine, a muscarinic receptor antagonist. YKS treatment reversed OBX-induced down-regulation of choline acetyltransferase and muscarinic muscarinic M₁ receptor expression without affecting muscarinic M₃ receptor expression or AChE activity. CONCLUSION These results demonstrate that YKS improves short-term memory deficit caused by OBX and that the effect is at least partly mediated by muscarinic receptor stimulation and the normalization of central cholinergic systems. The present findings also suggest that YKS has a therapeutic effect not only on BPSD, but also on memory impairment of AD.
Collapse
Affiliation(s)
- Marina Yamada
- Institute of Natural Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | | | | | | | | | | | | |
Collapse
|
36
|
Cognitive enhancers for facilitating drug cue extinction: insights from animal models. Pharmacol Biochem Behav 2011; 99:229-44. [PMID: 21295059 DOI: 10.1016/j.pbb.2011.01.018] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2010] [Revised: 12/23/2010] [Accepted: 01/24/2011] [Indexed: 12/30/2022]
Abstract
Given the success of cue exposure (extinction) therapy combined with a cognitive enhancer for reducing anxiety, it is anticipated that this approach will prove more efficacious than exposure therapy alone in preventing relapse in individuals with substance use disorders. Several factors may undermine the efficacy of exposure therapy for substance use disorders, but we suspect that neurocognitive impairments associated with chronic drug use are an important contributing factor. Numerous insights on these issues are gained from research using animal models of addiction. In this review, the relationship between brain sites whose learning, memory and executive functions are impaired by chronic drug use and brain sites that are important for effective drug cue extinction learning is explored first. This is followed by an overview of animal research showing improved treatment outcome for drug addiction (e.g. alcohol, amphetamine, cocaine, heroin) when explicit extinction training is conducted in combination with acute dosing of a cognitive-enhancing drug. The mechanism by which cognitive enhancers are thought to exert their benefits is by facilitating consolidation of drug cue extinction memory after activation of glutamatergic receptors. Based on the encouraging work in animals, factors that may be important for the treatment of drug addiction are considered.
Collapse
|
37
|
Lansbergen MM, Dumont GJH, van Gerven JMA, Buitelaar JK, Verkes RJ. Acute effects of MDMA (3,4-methylenedioxymethamphetamine) on EEG oscillations: alone and in combination with ethanol or THC (delta-9-tetrahydrocannabinol). Psychopharmacology (Berl) 2011; 213:745-56. [PMID: 20924751 PMCID: PMC3033515 DOI: 10.1007/s00213-010-2031-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2010] [Accepted: 09/20/2010] [Indexed: 01/09/2023]
Abstract
RATIONALE Typical users of 3,4-methylenedioxymethamphetamine (MDMA or "ecstasy") are polydrug users, combining MDMA with alcohol or cannabis [most active compound: delta-9-tetrahydrocannabinol (THC)]. OBJECTIVES The aim of the present study was to investigate whether co-administration of alcohol or THC with MDMA differentially affects ongoing electroencephalogram (EEG) oscillations compared to the administration of each drug alone. METHODS In two separate experiments, 16 volunteers received four different drug conditions: (1) MDMA (100 mg); (2) alcohol clamp (blood alcohol concentration = 0.6‰) or THC (inhalation of 4, 6 and 6 mg, interval of 1.5 h); (3) MDMA in combination with alcohol or THC; and (4) placebo. Before and after drug administration, electroencephalography was recorded during an eyes closed resting state. RESULTS Theta and alpha power increased after alcohol intake compared to placebo and reduced after MDMA intake. No interaction between alcohol and MDMA was found. Significant MDMA x THC effects for theta and lower-1-alpha power indicated that the power attenuation after the combined intake of MDMA and THC was less than the sum of each drug alone. For the lower-2-alpha band, the intake of MDMA or THC alone did not significantly affect power, but the intake of combined MDMA and THC significantly decreased lower-2-alpha power. CONCLUSIONS The present findings indicate that the combined intake of MDMA and THC, but not of MDMA and alcohol, affects ongoing EEG oscillations differently than the sum of either one drug alone. Changes in ongoing EEG oscillations may be related to the impaired task performance that has often been reported after drug intake.
Collapse
Affiliation(s)
- Marieke M. Lansbergen
- Department of Psychiatry, Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands
| | - Glenn J. H. Dumont
- Department of Psychiatry, Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands ,Moleman Psychopharmacology, Amerongen, the Netherlands
| | | | - Jan K. Buitelaar
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands ,Karakter Child and Adolescent Psychiatry University Centre, Nijmegen, the Netherlands
| | - Robbert-Jan Verkes
- Department of Psychiatry, Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands
| |
Collapse
|
38
|
Gould TJ. Addiction and cognition. Addict Sci Clin Pract 2010; 5:4-14. [PMID: 22002448 PMCID: PMC3120118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The brain regions and neural processes that underlie addiction overlap extensively with those that support cognitive functions, including learning, memory, and reasoning. Drug activity in these regions and processes during early stages of abuse foster strong maladaptive associations between drug use and environmental stimuli that may underlie future cravings and drug-seeking behaviors. With continued drug use, cognitive deficits ensue that exacerbate the difficulty of establishing sustained abstinence. The developing brain is particularly susceptible to the effects of drugs of abuse; prenatal, childhood, and adolescent exposures produce long-lasting changes in cognition. Patients with mental illness are at high risk for substance abuse, and the adverse impact on cognition may be particularly deleterious in combination with cognitive problems related to their mental disorders.
Collapse
Affiliation(s)
- Thomas J. Gould
- CORRESPONDENCE: Thomas J. Gould, Department of Psychology, Weiss Hall, Temple University, Philadelphia, PA 19122; e-mail:
| |
Collapse
|
39
|
Treweek JB, Roberts AJ, Janda KD. Superadditive effects of ethanol and flunitrazepam: implications of using immunopharmacotherapy as a therapeutic. Mol Pharm 2010; 7:2056-68. [PMID: 20849117 DOI: 10.1021/mp900293a] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
While benzodiazepine intoxication alone may elicit sedative and antianxiety effects, alcohol coingestion greatly amplifies this central nervous system depression. As a result, this drug combination gained notoriety for its role in cases of facilitated sexual assault and fatal overdose. We previously validated the ability of the novel antiflunitrazepam monoclonal antibody (mAb) RCA3A3 to bind flunitrazepam (FLU) in vivo and block FLU-induced impairment of locomotion and memory. A therapeutically relevant application of this high affinity mAb (K(d,app) = 200 nM), however, is to the more tenuous indication of flunitrazepam (FLU) and alcohol cointoxication. Employing a murine behavioral model, passive immunization with mAb RCA3A3 before injection of ethanol (EtOH: low-dose, 1 g/kg, or high-dose, 1.5 g/kg), FLU (0.06 mg/kg), or a cocktail of both drugs offered partial to full restoration of motor activity levels in co-drug treated and FLU-treated mouse groups (n = 12), respectively. Whereas all drug treatments left contextual learning intact, auditory cued learning was severely disrupted. Prophylactic administration of mAb RCA3A3 prevented this deficit in cued learning in FLU-treated mice but not in the FLU- and EtOH-treated mice, in which co-drug exposure exacerbated the impairment in cued fear conditioning. To substantiate this finding, a dose-response study was performed, and the changes in locomotor activity incurred by different FLU (low-dose, 0.06 mg/kg, or high-dose, 0.09 mg/kg), EtOH (1.0 g/kg, 1.5 g/kg), and mAb RCA3A3 (14.5 mg/kg, 21.8 mg/kg) dose combinations illustrated the potentiation in motor effects by concomitant exposure to FLU and EtOH. Thus, motor activity and fear conditioning results demonstrated that both the amount of FLU left unbound by antibody and the pharmacological additivity between FLU and EtOH, a GABA mimetic, were limiting factors in the therapeutic efficacy of mAb RCA3A3. In sum, our study highlights the complex nature of psychomotor impairment upon co-drug versus singular drug exposure, which may pose a unique challenge to therapeutic treatment.
Collapse
Affiliation(s)
- Jennifer B Treweek
- Department of Chemistry and Immunology of The Skaggs Institute for Chemical Biology, and Worm Institute for Research and Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, USA
| | | | | |
Collapse
|
40
|
Chin VS, Van Skike CE, Matthews DB. Effects of ethanol on hippocampal function during adolescence: a look at the past and thoughts on the future. Alcohol 2010; 44:3-14. [PMID: 20113870 DOI: 10.1016/j.alcohol.2009.10.015] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2009] [Revised: 09/14/2009] [Accepted: 10/22/2009] [Indexed: 12/20/2022]
Abstract
It has been demonstrated by several laboratories that ethanol, both acute and chronic, produces effects that are age dependent. Specifically, adolescent rats are less sensitive to the hypnotic and motor-impairing effects of ethanol but are more sensitive to the hypothermic effects of the drug. However, the results on hippocampal function are not as clear. For example, there have been mixed findings regarding adolescent sensitivity of hippocampal-dependent (spatial) memory in response to ethanol. The current review explores the present state of the field as it relates to ethanol's effects in the hippocampus, particularly as it relates to spatial memory. In addition, we review potential neurobiological mechanisms that might underlie the age-dependent effects of ethanol in the hippocampus. Finally, future directions are proposed that will advance the state of the field as it relates to ethanol's effect during this developmental period.
Collapse
Affiliation(s)
- Vivien S Chin
- Department of Psychology, Baylor University Additions Research Consortium, Waco, TX, USA
| | | | | |
Collapse
|
41
|
Dumont GJH, Schoemaker RC, Touw DJ, Sweep FCGJ, Buitelaar JK, van Gerven JMA, Verkes RJ. Acute psychomotor effects of MDMA and ethanol (co-) administration over time in healthy volunteers. J Psychopharmacol 2010; 24:155-64. [PMID: 19164498 DOI: 10.1177/0269881108099214] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
In Western societies, a considerable percentage of young people use 3,4-methylenedioxymethamphetamine (MDMA or 'ecstasy'). The use of alcohol (ethanol) in combination with ecstasy is common. The aim of the present study was to assess the acute psychomotor and subjective effects of (co-) administration of MDMA and ethanol over time and in relation to the pharmacokinetics. We performed a four-way, double blind, randomized, crossover, placebo-controlled study in 16 healthy volunteers (nine men, seven women) between the ages of 18 and 29. MDMA (100 mg) was given orally while blood alcohol concentration was maintained at pseudo-steady state levels of approximately 0.6 per thousand for 3 h by a 10% intravenous ethanol clamp. MDMA significantly increased psychomotor speed but did not affect psychomotor accuracy and induced subjective arousal. Ethanol impaired both psychomotor speed and accuracy and induced sedation. Coadministration of ethanol and MDMA improved psychomotor speed but impaired psychomotor accuracy compared with placebo and reversed ethanol-induced sedation. Pharmacokinetics and pharmacodynamics showed maximal effects at 90-150 min after MDMA administration after which drug effects declined in spite of persisting MDMA plasma concentration, with the exception of ethanol-induced sedation, which manifested itself fully only after the infusion was stopped. In conclusion, results show that subjects were more aroused when intoxicated with both substances combined compared with placebo, but psychomotor accuracy was significantly impaired. These findings may have implications for general neuropsychological functioning as this may provide a sense of adequate performance that does not agree with a significant reduction in psychomotor accuracy.
Collapse
Affiliation(s)
- G J H Dumont
- Department of Psychiatry, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
42
|
Molnár M, Boha R, Czigler B, Gaál ZA. The Acute Effect of Alcohol on Various Memory Processes. J PSYCHOPHYSIOL 2010. [DOI: 10.1027/0269-8803/a000038] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
This review surveys relevant and recent data of the pertinent literature regarding the acute effect of alcohol on various kinds of memory processes with special emphasis on working memory. The characteristics of different types of long-term memory (LTM) and short-term memory (STM) processes are summarized with an attempt to relate these to various structures in the brain. LTM is typically impaired by chronic alcohol intake but according to some data a single dose of ethanol may have long lasting effects if administered at a critically important age. The most commonly seen deleterious acute effect of alcohol to STM appears following large doses of ethanol in conditions of “binge drinking” causing the “blackout” phenomenon. However, with the application of various techniques and well-structured behavioral paradigms it is possible to detect, albeit occasionally, subtle changes of cognitive processes even as a result of a low dose of alcohol. These data may be important for the consideration of legal consequences of low-dose ethanol intake in conditions such as driving, etc.
Collapse
Affiliation(s)
- Márk Molnár
- Institute for Psychology, Hungarian Academy of Sciences, Budapest, Hungary
| | - Roland Boha
- Institute for Psychology, Hungarian Academy of Sciences, Budapest, Hungary
| | - Balázs Czigler
- Institute for Psychology, Hungarian Academy of Sciences, Budapest, Hungary
| | - Zsófia Anna Gaál
- Institute for Psychology, Hungarian Academy of Sciences, Budapest, Hungary
| |
Collapse
|
43
|
Acute alcohol decreases performance of an instrumental response to avoid aversive consequences in social drinkers. Psychopharmacology (Berl) 2009; 205:577-87. [PMID: 19468716 DOI: 10.1007/s00213-009-1565-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2009] [Accepted: 05/05/2009] [Indexed: 10/20/2022]
Abstract
BACKGROUND Recent studies demonstrated that alcohol impairs inhibitory control of behavioural responses. AIMS We questioned whether alcohol via its disinhibiting effects would also impair the inhibition of an instrumental avoidance response in the presence of a safety signal. DESIGN Thirty-six moderate social drinkers were randomly allocated to receiving either alcohol (0.8 g/kg) or placebo before performing an instrumental avoidance procedure. White noise of 102 db was used as aversive outcome presented at a variable interval schedule in S+ trials, while no noise was presented in S- trials. An instrumental response (repeated space bar presses to avoid the noise presented at a variable interval) abolished the noise. The Stop Signal task and the affective Go/No-Go task were administered as inhibitory control tasks. RESULTS Alcohol did not change the avoidance response rate in the presence of S- (safety signal). However, participants under alcohol performed the avoidance response to a lower extent than placebo subjects in S+ trials. Alcohol impaired performance in the Stop Signal task and increased the number of commission errors in the affective Go/No-Go task. Conditioned attentional and emotional responses to the S+ as well as knowledge of stimulus-response outcome contingencies were not affected by alcohol. CONCLUSIONS Acute alcohol may decrease the motivation to avoid negative consequences and thus might contribute to risky behaviour and binge drinking.
Collapse
|
44
|
The hippocampus and cingulate cortex differentially mediate the effects of nicotine on learning versus on ethanol-induced learning deficits through different effects at nicotinic receptors. Neuropsychopharmacology 2009; 34:2167-79. [PMID: 19404242 PMCID: PMC2770339 DOI: 10.1038/npp.2009.45] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The current study examined the effects of nicotine infusion into the dorsal hippocampus or anterior cingulate on fear conditioning and on ethanol-induced deficits in fear conditioning, and whether these effects involved receptor activation or inactivation. Conditioning consisted of two white noise (30 s, 85 dB)-foot-shock (2 s, 0.57 mA) pairings. Saline or ethanol was administered to C57BL/6 mice 15 min before training and saline or nicotine was administered 5 min before training or before training and testing. The ability of the high-affinity nicotinic acetylcholinergic receptor (nAChR) antagonist dihydro-beta-erythroidine (DHbetaE) to modulate the effects of ethanol and nicotine was also tested; saline or DHbetaE was administered 25 (injection) or 15 (infusion) minutes before training or before training and testing. Infusion of nicotine into the hippocampus enhanced contextual fear conditioning but had no effect on ethanol-induced learning deficits. Infusion of nicotine into the anterior cingulate ameliorated ethanol-induced deficits in contextual and cued fear conditioning but had no effect on learning in ethanol-naive mice. DHbetaE blocked the effects of nicotine on ethanol-induced deficits; interestingly, DHbetaE alone and co-administration of subthreshold doses of DHbetaE and nicotine also ameliorated ethanol-induced deficits but failed to enhance learning. Finally, DHbetaE failed to ameliorate ethanol-induced deficits in beta2 nAChR subunit knockout mice. These results suggest that nicotine acts in the hippocampus to enhance contextual learning, but acts in the cingulate to ameliorate ethanol-induced learning deficits through inactivation of high-affinity beta2 subunit-containing nAChRs.
Collapse
|
45
|
Extinction learning of stimulus reward contingencies: The acute effects of alcohol. Drug Alcohol Depend 2009; 102:56-62. [PMID: 19278796 DOI: 10.1016/j.drugalcdep.2009.01.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2008] [Revised: 01/05/2009] [Accepted: 01/06/2009] [Indexed: 11/22/2022]
Abstract
BACKGROUND Recent theories suggest that extinction is, at least partly, new learning suppressing original associations between a conditioned stimulus and a conditioned response without severing those associations. During extinction alcohol via its effects on inhibitory control may reduce the ability to suppress the original associations between a conditioned stimulus and a conditioned response leading to an impairment of extinction learning. Thus, the present study is set out to examine the effects of alcohol on extinction learning to enhance current knowledge on mechanisms of extinction and conditions that might hamper extinction, which is an important aspect for the treatment of alcohol-dependent patients. METHODS Light to moderate social drinkers (N=32) acquired an instrumental reward seeking response. Extinction training of the reward seeking response was performed after administration of a dose of 0.8 g/kg alcohol resulting in a peak blood alcohol concentration ranging from 112 to 184 mg/dL. In addition, we assessed subjective alcohol effects and administered a Stop-Signal task which measures the ability to inhibit a pre-potent motor response. RESULTS Alcohol influenced subjective ratings of light-headedness and increased the Stop-Signal reaction time indicating disinhibiting effects. However, our results did not show any impairment of learning of extinction after the administration of alcohol. Behavioural as well as attentional responses indicated extinction of conditioned responses for both experimental groups. CONCLUSIONS These findings suggest that alcohol at a dose that impairs performance in a task of inhibitory control does not impair learning of extinction.
Collapse
|
46
|
Hunt PS, Levillain ME, Spector BM, Kostelnik LA. Post-training ethanol disrupts trace conditioned fear in rats: effects of timing of ethanol, dose and trace interval duration. Neurobiol Learn Mem 2008; 91:73-80. [PMID: 18952186 DOI: 10.1016/j.nlm.2008.10.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2008] [Revised: 09/27/2008] [Accepted: 10/07/2008] [Indexed: 10/21/2022]
Abstract
Ethanol has complex effects on memory performance, although hippocampus-dependent memory may be especially vulnerable to disruption by acute ethanol intoxication occurring during or shortly after a training episode. In the present experiments, the effects of post-training ethanol on delay and trace fear conditioning were examined in adolescent rats. In Experiment 1, 30-day-old Sprague-Dawley rats were given delay or trace conditioning trials in which a 10s flashing light CS was paired with a 0.5 mA shock US. For trace groups, the trace interval was 10 s. On days 31-33, animals were administered ethanol once daily (0.0 or 2.5 g/kg via intragastric intubation), and on day 34 animals were tested for CS-elicited freezing. Results showed that post-training ethanol affected the expression of trace, but had no effect on delay conditioned fear. Experiment 2 revealed that this effect was dose-dependent; doses lower than 2.5 g/kg were without effect. Experiment 3 evaluated whether proximity of ethanol to the time of training or testing was critical. Results show that ethanol administration beginning 24h after training was more detrimental to trace conditioned freezing than administration that was delayed by 48 h. Finally, in Experiment 4 animals were trained with one of three different trace intervals: 1, 3 or 10s. Results indicate that post-training administration of 2.5 g/kg ethanol disrupted trace conditioned fear in subjects trained with a 10s, but not with a 1 or 3s, trace interval. Collectively the results suggest that ethanol administration impairs post-acquisition memory processing of hippocampus-dependent trace fear conditioning.
Collapse
Affiliation(s)
- Pamela S Hunt
- Department of Psychology, College of William and Mary, Williamsburg, VA 23187-8795, USA.
| | | | | | | |
Collapse
|
47
|
Basavarajappa BS, Ninan I, Arancio O. Acute ethanol suppresses glutamatergic neurotransmission through endocannabinoids in hippocampal neurons. J Neurochem 2008; 107:1001-13. [PMID: 18796007 DOI: 10.1111/j.1471-4159.2008.05685.x] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Ethanol exposure during fetal development is a leading cause of long-term cognitive impairments. Studies suggest that ethanol exposure have deleterious effects on the hippocampus, a brain region that is important for learning and memory. Ethanol exerts its effects, in part, via alterations in glutamatergic neurotransmission, which is critical for the maturation of neuronal circuits during development. The current literature strongly supports the growing evidence that ethanol inhibits glutamate release in the neonatal CA1 hippocampal region. However, the exact molecular mechanism responsible for this effect is not well understood. In this study, we show that ethanol enhances endocannabinoid (EC) levels in cultured hippocampal neurons, possibly through calcium pathways. Acute ethanol depresses miniature post-synaptic current (mEPSC) frequencies without affecting their amplitude. This suggests that ethanol inhibits glutamate release. The CB1 receptors (CB1Rs) present on pre-synaptic neurons are not altered by acute ethanol. The CB1R antagonist SR 141716A reverses ethanol-induced depression of mEPSC frequency. Drugs that are known to enhance the in vivo function of ECs occlude ethanol effects on mEPSC frequency. Chelation of post-synaptic calcium by EGTA antagonizes ethanol-induced depression of mEPSC frequency. The activation of CB1R with the selective agonist WIN55,212-2 also suppresses the mEPSC frequency. This WIN55,212-2 effect is similar to the ethanol effects and is reversed by SR141716A. In addition, tetani-induced excitatory post-synaptic currents (EPSCs) are depressed by acute ethanol. SR141716A significantly reverses ethanol effects on evoked EPSC amplitude in a dual recording preparation. These observations, taken together, suggest the participation of ECs as retrograde messengers in the ethanol-induced depression of synaptic activities.
Collapse
Affiliation(s)
- Balapal S Basavarajappa
- Division of Analytical Psychopharmacology, Nathan Kline Institute for Psychiatric Research, Orangeburg, New York, USA.
| | | | | |
Collapse
|
48
|
Mustard JA, Edgar EA, Mazade RE, Wu C, Lillvis JL, Wright GA. Acute ethanol ingestion impairs appetitive olfactory learning and odor discrimination in the honey bee. Neurobiol Learn Mem 2008; 90:633-43. [PMID: 18723103 DOI: 10.1016/j.nlm.2008.07.017] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2008] [Revised: 07/29/2008] [Accepted: 07/29/2008] [Indexed: 11/30/2022]
Abstract
Invertebrates are valuable models for increasing our understanding of the effects of ethanol on the nervous system, but most studies on invertebrates and ethanol have focused on the effects of ethanol on locomotor behavior. In this work we investigate the influence of an acute dose of ethanol on appetitive olfactory learning in the honey bee (Apis mellifera), a model system for learning and memory. Adult worker honey bees were fed a range of doses (2.5%, 5%, 10%, or 25%) of ethanol and then conditioned to associate an odor with a sucrose reward using either a simple or differential conditioning paradigm. Consumption of ethanol before conditioning significantly reduced both the rate of acquisition and the asymptotic strength of the association. Honey bees also exhibited a dose dependent reduction in arousal/attention during conditioning. Consumption of ethanol after conditioning did not affect recall 24h later. The observed deficits in acquisition were not due to the affect of ethanol on gustatory sensitivity or motor function. However, honey bees given higher doses of ethanol had difficulty discriminating amongst different odors suggesting that ethanol consumption influences olfactory processing. Taken together, these results demonstrate that an acute dose of ethanol affects appetitive learning and olfactory perception in the honey bee.
Collapse
Affiliation(s)
- Julie A Mustard
- School of Life Sciences, Arizona State University, PO Box 874501, Tempe, AZ 85287, USA.
| | | | | | | | | | | |
Collapse
|
49
|
Gulick D, Gould TJ. Varenicline ameliorates ethanol-induced deficits in learning in C57BL/6 mice. Neurobiol Learn Mem 2008; 90:230-6. [PMID: 18411066 DOI: 10.1016/j.nlm.2008.03.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2008] [Revised: 03/06/2008] [Accepted: 03/06/2008] [Indexed: 11/29/2022]
Abstract
Ethanol is a frequently abused drug that impairs cognitive processes such as learning. Varenicline, an alpha4beta2 nicotinic receptor partial agonist and alpha7 nicotinic receptor full agonist prescribed for smoking cessation, has been shown to decrease ethanol consumption. The current study investigated whether varenicline could ameliorate ethanol-induced deficits in learning and whether varenicline alters blood alcohol concentration in C57BL/6 mice. Conditioning consisted of two auditory conditioned stimulus (CS; 30s, 85dB white noise)-foot shock unconditioned stimulus (US; 2s, 0.57mA) pairings. For all studies, saline or ethanol (1.0, 1.5, 2.0g/kg i.p.) was administered 15min before training, and saline or varenicline (0.05, 0.1, 0.2mg/kg i.p.) was administered 60min before either training or testing. For blood alcohol analysis, saline or varenicline (0.1mg/kg) was administered 60min before collection, and saline or ethanol (1.0, 1.5, 2.0g/kg) was administered 15min before collection. Varenicline dose-dependently ameliorated ethanol-induced conditioning deficits for all three doses of ethanol when administered before training but not when administered 24h later, before testing. In addition, varenicline did not alter blood alcohol concentration. The smoking cessation aid varenicline may have therapeutic uses for treating ethanol-associated disruptions in cognitive processes.
Collapse
Affiliation(s)
- Danielle Gulick
- Department of Psychology, Neuroscience Program, Temple University, Weiss Hall, Rm 865, 1701 N. 13th Street, Philadelphia, PA 19122, USA
| | | |
Collapse
|
50
|
Interactive effects of ethanol and nicotine on learning in C57BL/6J mice depend on both dose and duration of treatment. Psychopharmacology (Berl) 2008; 196:483-95. [PMID: 17968532 PMCID: PMC2722442 DOI: 10.1007/s00213-007-0982-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2007] [Accepted: 10/11/2007] [Indexed: 10/22/2022]
Abstract
OBJECTIVE AND RATIONALE Alcohol and nicotine are commonly co-abused; one possible explanation for co-abuse is that each drug ameliorates the aversive effects of the other. Both drugs have dose-dependent effects on learning and memory. Thus, this study examined the interactive effects of acute ethanol and acute, chronic, or withdrawal from chronic nicotine on fear conditioning in C57BL/6J mice. MATERIALS AND METHODS Conditioning consisted of auditory conditioned stimulus-foot-shock unconditioned stimulus pairings. For acute studies, saline or ethanol, then saline or nicotine was administered before training, and saline or nicotine was also administered before testing. For chronic and withdrawal studies, saline or nicotine was administered chronically, and ethanol or saline was administered before training. RESULTS Acute nicotine (0.09 mg/kg) reversed ethanol-induced deficits (1.0 and 1.5 g/kg) in contextual and cued fear conditioning, whereas a low dose of ethanol (0.25 g/kg) reversed nicotine (6.3 mg kg(-1) day(-1)) withdrawal-induced deficits in contextual conditioning. Tolerance developed for the effects of nicotine on ethanol-induced deficits in conditioning and cross-tolerance between chronic nicotine and acute ethanol was seen for the enhancing effects of ethanol on conditioning. CONCLUSIONS The complex and sometimes polar actions of ethanol and nicotine on behavior may contribute to co-abuse of these drugs. Specifically, smoking may initially reduce the aversive effects of ethanol, but tolerance develops for this effect. In addition, low doses of alcohol may lessen nicotine withdrawal symptoms.
Collapse
|