1
|
Kastner-Blasczyk AR, Hester SW, Reasons SE, Scofield MD, Woodward JJ. Effect of an astrocyte calcium exporter on orbitofrontal cortex neuron excitability, astrocyte-synaptic interaction, and alcohol consumption. Neuropharmacology 2025; 269:110365. [PMID: 39952350 PMCID: PMC11995387 DOI: 10.1016/j.neuropharm.2025.110365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/30/2025] [Accepted: 02/11/2025] [Indexed: 02/17/2025]
Abstract
Previous electrophysiology studies show that acute ethanol inhibits firing of orbitofrontal (OFC) cortex neurons while chronic intermittent ethanol (CIE) exposure increases firing accompanied by enhanced ethanol drinking. The acute ethanol inhibition of OFC neuronal firing is mediated by inhibitory glycine receptors and is reduced by expressing a plasma membrane calcium ATPase (PMCA) in OFC astrocytes. In this study, we tested the effects of astrocyte PMCA on CIE-induced increases in excitability and alcohol consumption and the physical interaction between OFC astrocytes and neurons. CIE increased neuronal firing in male mice as compared to Air controls while PMCA itself increased firing in Air control male mice. In contrast, PMCA reduced CIE-mediated hyperexcitability of firing in females. CIE did not affect OFC astrocyte size in control or PMCA male mice but increased astrocyte size in female mice. Similar to spiking, PMCA and CIE both increased the number of GluA1 containing synapses within the vicinity of virally labeled astrocytes in male mice but had differential effects in females. The astrocytic interaction with GluA1 labeled synapses was not affected by CIE treatment in male or female control mice, but there was a treatment-dependent effect of PMCA in male mice. CIE increased alcohol consumption in control but not PMCA male mice and had no effect on drinking in female mice. Lastly, OFC astrocyte PMCA expression had no effect on behavioral measures of locomotion, anxiety, spontaneous alternation, or spatial memory. These findings reveal important sex-dependent differences in the physiological, structural and behavioral actions of OFC astrocytes.
Collapse
Affiliation(s)
- A R Kastner-Blasczyk
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, United States
| | - S W Hester
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, United States
| | - S E Reasons
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, United States
| | - M D Scofield
- Department of Anesthesia and Perioperative Medicine, Medical University of South Carolina, Charleston, SC, United States
| | - J J Woodward
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, United States.
| |
Collapse
|
2
|
Tang S, Kim SW, Olsen-Dufour A, Pearson T, Freaney M, Singley E, Jenkins M, Burkard NJ, Wozniak A, Parcon P, Wu S, Morse CL, Jana S, Liow JS, Zoghbi SS, Vendruscolo JCM, Vendruscolo LF, Pike VW, Koob GF, Volkow ND, Innis RB. PET imaging in rat brain shows opposite effects of acute and chronic alcohol exposure on phosphodiesterase-4B, an indirect biomarker of cAMP activity. Neuropsychopharmacology 2024; 50:444-451. [PMID: 39285225 PMCID: PMC11632093 DOI: 10.1038/s41386-024-01988-y] [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: 05/10/2024] [Revised: 08/26/2024] [Accepted: 08/29/2024] [Indexed: 12/12/2024]
Abstract
The cyclic adenosine monophosphate (cAMP) cascade is thought to play an important role in regulating alcohol-dependent behaviors, with potentially opposite effects following acute versus chronic administration. Phosphodiesterase 4 (PDE4) is the primary brain enzyme that metabolizes cAMP, thereby terminating its signal. Radioligand binding to PDE4 serves as an indirect biomarker of cAMP activity, as cAMP-protein kinase A (PKA)-mediated phosphorylation of PDE4 increases its affinity for radioligand binding ~10-fold. Of the four PDE4 subtypes, PDE4B polymorphisms are known to be strongly associated with alcohol and substance use disorders. This study imaged rats with the PDE4B-preferring positron emission tomography (PET) radioligand [18F]PF-06445974 following acute and chronic ethanol administration, aiming to explore the potential of PDE4B PET imaging for future human studies. Compared to the control group treated with saline, acute alcohol administration (i.p. ethanol 0.5 g/kg) significantly increased whole brain uptake of [18F]PF-06445974 as early as 30 minutes post-exposure. This effect persisted at 2 hours, peaked at 4 hours, and diminished at 6 hours and 24 hours post-exposure. In contrast, in a rat model of alcohol dependence, [18F]PF-06445974 brain uptake was significantly reduced at 5 hours post-exposure and was normalized by 3 days. This reduction may reflect long-term adaptation to repeated alcohol-induced activation of cAMP signaling with chronic exposure. Taken together, the results suggest that PET imaging of PDE4B in individuals with alcohol use disorder (AUD) should be considered in conjunction with ongoing trials of PDE4 inhibitors to treat alcohol withdrawal and reduce alcohol consumption.
Collapse
Affiliation(s)
- Shiyu Tang
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, MD, USA
| | - Sung Won Kim
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, USA
| | - Amanda Olsen-Dufour
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, MD, USA
| | - Torben Pearson
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, USA
| | - Michael Freaney
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, USA
| | - Erick Singley
- Clinical Core Laboratory, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, USA
| | - Madeline Jenkins
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, MD, USA
| | - Nathaniel J Burkard
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, USA
| | - Aaron Wozniak
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, USA
| | - Paul Parcon
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, MD, USA
| | - Shawn Wu
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, MD, USA
| | - Cheryl L Morse
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, MD, USA
| | - Susovan Jana
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, MD, USA
| | - Jeih-San Liow
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, MD, USA
| | - Sami S Zoghbi
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, MD, USA
| | - Janaina C M Vendruscolo
- Neurobiology of Addiction Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Baltimore, MD, USA
| | - Leandro F Vendruscolo
- Stress and Addiction Neuroscience Unit, Integrative Neuroscience Research Branch, National Institute on Drug Abuse Intramural Research Program, National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, Baltimore, MD, USA
| | - Victor W Pike
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, MD, USA
| | - George F Koob
- Neurobiology of Addiction Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Baltimore, MD, USA
| | - Nora D Volkow
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, USA.
| | - Robert B Innis
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, MD, USA.
| |
Collapse
|
3
|
Koob GF, Vendruscolo L. Theoretical Frameworks and Mechanistic Aspects of Alcohol Addiction: Alcohol Addiction as a Reward Deficit/Stress Surfeit Disorder. Curr Top Behav Neurosci 2023. [PMID: 37421551 DOI: 10.1007/7854_2023_424] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/10/2023]
Abstract
Alcohol use disorder (AUD) can be defined by a compulsion to seek and take alcohol, the loss of control in limiting intake, and the emergence of a negative emotional state when access to alcohol is prevented. Alcohol use disorder impacts multiple motivational mechanisms and can be conceptualized as a disorder that includes a progression from impulsivity (positive reinforcement) to compulsivity (negative reinforcement). Compulsive drug seeking that is associated with AUD can be derived from multiple neuroadaptations, but the thesis argued herein is that a key component involves the construct of negative reinforcement. Negative reinforcement is defined as drug taking that alleviates a negative emotional state. The negative emotional state that drives such negative reinforcement is hypothesized to derive from the dysregulation of specific neurochemical elements that are involved in reward and stress within basal forebrain structures that involve the ventral striatum and extended amygdala, respectively. Specific neurochemical elements in these structures include decreases in reward neurotransmission (e.g., decreases in dopamine and opioid peptide function in the ventral striatum) and the recruitment of brain stress systems (e.g., corticotropin-releasing factor [CRF]) in the extended amygdala, which contributes to hyperkatifeia and greater alcohol intake that is associated with dependence. Glucocorticoids and mineralocorticoids may play a role in sensitizing the extended amygdala CRF system. Other components of brain stress systems in the extended amygdala that may contribute to the negative motivational state of withdrawal include norepinephrine in the bed nucleus of the stria terminalis, dynorphin in the nucleus accumbens, hypocretin and vasopressin in the central nucleus of the amygdala, and neuroimmune modulation. Decreases in the activity of neuropeptide Y, nociception, endocannabinoids, and oxytocin in the extended amygdala may also contribute to hyperkatifeia that is associated with alcohol withdrawal. Such dysregulation of emotional processing may also significantly contribute to pain that is associated with alcohol withdrawal and negative urgency (i.e., impulsivity that is associated with hyperkatifeia during hyperkatifeia). Thus, an overactive brain stress response system is hypothesized to be activated by acute excessive drug intake, to be sensitized during repeated withdrawal, to persist into protracted abstinence, and to contribute to the compulsivity of AUD. The combination of the loss of reward function and recruitment of brain stress systems provides a powerful neurochemical basis for a negative emotional state that is responsible for the negative reinforcement that at least partially drives the compulsivity of AUD.
Collapse
Affiliation(s)
- George F Koob
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, USA.
| | - Leandro Vendruscolo
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| |
Collapse
|
4
|
Koob GF. Drug Addiction: Hyperkatifeia/Negative Reinforcement as a Framework for Medications Development. Pharmacol Rev 2021; 73:163-201. [PMID: 33318153 PMCID: PMC7770492 DOI: 10.1124/pharmrev.120.000083] [Citation(s) in RCA: 195] [Impact Index Per Article: 48.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Compulsive drug seeking that is associated with addiction is hypothesized to follow a heuristic framework that involves three stages (binge/intoxication, withdrawal/negative affect, and preoccupation/anticipation) and three domains of dysfunction (incentive salience/pathologic habits, negative emotional states, and executive function, respectively) via changes in the basal ganglia, extended amygdala/habenula, and frontal cortex, respectively. This review focuses on neurochemical/neurocircuitry dysregulations that contribute to hyperkatifeia, defined as a greater intensity of negative emotional/motivational signs and symptoms during withdrawal from drugs of abuse in the withdrawal/negative affect stage of the addiction cycle. Hyperkatifeia provides an additional source of motivation for compulsive drug seeking via negative reinforcement. Negative reinforcement reflects an increase in the probability of a response to remove an aversive stimulus or drug seeking to remove hyperkatifeia that is augmented by genetic/epigenetic vulnerability, environmental trauma, and psychiatric comorbidity. Neurobiological targets for hyperkatifeia in addiction involve neurocircuitry of the extended amygdala and its connections via within-system neuroadaptations in dopamine, enkephalin/endorphin opioid peptide, and γ-aminobutyric acid/glutamate systems and between-system neuroadaptations in prostress corticotropin-releasing factor, norepinephrine, glucocorticoid, dynorphin, hypocretin, and neuroimmune systems and antistress neuropeptide Y, nociceptin, endocannabinoid, and oxytocin systems. Such neurochemical/neurocircuitry dysregulations are hypothesized to mediate a negative hedonic set point that gradually gains allostatic load and shifts from a homeostatic hedonic state to an allostatic hedonic state. Based on preclinical studies and translational studies to date, medications and behavioral therapies that reset brain stress, antistress, and emotional pain systems and return them to homeostasis would be promising new targets for medication development. SIGNIFICANCE STATEMENT: The focus of this review is on neurochemical/neurocircuitry dysregulations that contribute to hyperkatifeia, defined as a greater intensity of negative emotional/motivational signs and symptoms during withdrawal from drugs of abuse in the withdrawal/negative affect stage of the drug addiction cycle and a driving force for negative reinforcement in addiction. Medications and behavioral therapies that reverse hyperkatifeia by resetting brain stress, antistress, and emotional pain systems and returning them to homeostasis would be promising new targets for medication development.
Collapse
Affiliation(s)
- George F Koob
- National Institute on Alcohol Abuse and Alcoholism and National Institute on Drug Abuse, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
5
|
Alcohol. Alcohol 2021. [DOI: 10.1016/b978-0-12-816793-9.00001-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
6
|
Strong CE, Kabbaj M. Neural Mechanisms Underlying the Rewarding and Therapeutic Effects of Ketamine as a Treatment for Alcohol Use Disorder. Front Behav Neurosci 2020; 14:593860. [PMID: 33362485 PMCID: PMC7759199 DOI: 10.3389/fnbeh.2020.593860] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 11/06/2020] [Indexed: 11/13/2022] Open
Abstract
Alcohol use disorder (AUD) is the most prevalent substance use disorder and causes a significant global burden. Relapse rates remain incredibly high after decades of attempting to develop novel treatment options that have failed to produce increased rates of sobriety. Ketamine has emerged as a potential treatment for AUD following its success as a therapeutic agent for depression, demonstrated by several preclinical studies showing that acute administration reduced alcohol intake in rodents. As such, ketamine's therapeutic effects for AUD are now being investigated in clinical trials with the hope of it being efficacious in prolonging sobriety from alcohol in humans (ClinicalTrials.gov, Identifier: NCT01558063). Importantly, ketamine's antidepressant effects only last for about 1-week and because AUD is a lifelong disorder, repeated treatment regimens would be necessary to maintain sobriety. This raises questions regarding its safety for AUD treatment since ketamine itself has the potential for addiction. Therefore, this review aims to summarize the neuroadaptations related to alcohol's addictive properties as well as ketamine's therapeutic and addictive properties. To do this, the focus will be on reward-related brain regions such as the nucleus accumbens (NAc), dorsal striatum, prefrontal cortex (PFC), hippocampus, and ventral tegmental area (VTA) to understand how acute vs. chronic exposure will alter reward signaling over time. Additionally, evidence from these studies will be summarized in both male and female subjects. Accordingly, this review aims to address the safety of repeated ketamine infusions for the treatment of AUD. Although more work about the safety of ketamine to treat AUD is warranted, we hope this review sheds light on some answers about the safety of repeated ketamine infusions.
Collapse
Affiliation(s)
- Caroline E Strong
- Program in Neuroscience, Department of Biomedical Sciences, Florida State University, Tallahassee, FL, United States
| | - Mohamed Kabbaj
- Program in Neuroscience, Department of Biomedical Sciences, Florida State University, Tallahassee, FL, United States
| |
Collapse
|
7
|
Wen RT, Zhang FF, Zhang HT. Cyclic nucleotide phosphodiesterases: potential therapeutic targets for alcohol use disorder. Psychopharmacology (Berl) 2018; 235:1793-1805. [PMID: 29663017 PMCID: PMC5949271 DOI: 10.1007/s00213-018-4895-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 03/29/2018] [Indexed: 12/19/2022]
Abstract
Alcohol use disorder (AUD), which combines the criteria of both alcohol abuse and dependence, contributes as an important causal factor to multiple health and social problems. Given the limitation of current treatments, novel medications for AUD are needed to better control alcohol consumption and maintain abstinence. It has been well established that the intracellular signal transduction mediated by the second messengers cyclic AMP (cAMP) and cyclic GMP (cGMP) crucially underlies the genetic predisposition, rewarding properties, relapsing features, and systemic toxicity of compulsive alcohol consumption. On this basis, the upstream modulators phosphodiesterases (PDEs), which critically control intracellular levels of cyclic nucleotides by catalyzing their degradation, are proposed to play a role in modulating alcohol abuse and dependent process. Here, we highlight existing evidence that correlates cAMP and cGMP signal cascades with the regulation of alcohol-drinking behavior and discuss the possibility that PDEs may become a novel class of therapeutic targets for AUD.
Collapse
Affiliation(s)
- Rui-Ting Wen
- Department of Pharmacy, Peking University People's Hospital, Beijing, 100044, China
| | - Fang-Fang Zhang
- Institute of Pharmacology, Qilu Medical University, Taian, 271016, Shandong, China
| | - Han-Ting Zhang
- Institute of Pharmacology, Qilu Medical University, Taian, 271016, Shandong, China.
- Departments of Behavioral Medicine and Psychiatry and Physiology, Pharmacology and Neuroscience, Rockefeller Neurosciences Institute, West Virginia University Health Sciences Center, Morgantown, WV, 26506, USA.
| |
Collapse
|
8
|
Wen RT, Liang JH, Zhang HT. Targeting Phosphodiesterases in Pharmacotherapy for Substance Dependence. ADVANCES IN NEUROBIOLOGY 2018; 17:413-444. [PMID: 28956341 DOI: 10.1007/978-3-319-58811-7_15] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Substance dependence is a chronic relapsing brain disorder associated with adaptational changes in synaptic plasticity and neuronal functions. The high levels of substance consumption and relapse rate suggest more reliable medications are in need to better address the underlying causes of this disease. It has been well established that the intracellular second messengers cyclic AMP (cAMP) and cyclic GMP (cGMP) and their signaling systems play an important role in the molecular mechanisms of substance taking behaviors. On this basis, the phosphodiesterase (PDE) superfamily, which crucially controls cyclic nucleotide levels by catalyzing their hydrolysis, has been proposed as a novel class of therapeutic targets for substance use disorders. This chapter reviews the expression patterns of PDEs in the brain with regard to neural structures underlying the dependent process and highlights available evidence for a modulatory role of PDEs in substance dependence.
Collapse
Affiliation(s)
- Rui-Ting Wen
- Department of Pharmacy, Peking University People's Hospital, Beijing, 100044, China
| | - Jian-Hui Liang
- Department of Molecular and Cellular Pharmacology, Peking University School of Pharmaceutical Sciences, Beijing, 100191, China.
| | - Han-Ting Zhang
- Department of Behavioral Medicine and Psychiatry, West Virginia University Health Sciences Center, 1 Medical Center Drive, Morgantown, WV, 26506, USA. .,Department of Physiology and Pharmacology, West Virginia University Health Sciences Center, 1 Medical Center Drive, Morgantown, WV, 26506, USA. .,Institute of Pharmacology, Taishan Medical University, Taian, 271016, China.
| |
Collapse
|
9
|
Dar MS. Ethanol-Induced Cerebellar Ataxia: Cellular and Molecular Mechanisms. THE CEREBELLUM 2016; 14:447-65. [PMID: 25578036 DOI: 10.1007/s12311-014-0638-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The cerebellum is an important target of ethanol toxicity given that cerebellar ataxia is the most consistent physical manifestation of acute ethanol consumption. Despite the significance of the cerebellum in ethanol-induced cerebellar ataxia (EICA), the cellular and molecular mechanisms underlying EICA are incompletely understood. However, two important findings have shed greater light on this phenomenon. First, ethanol-induced blockade of cerebellar adenosine uptake in rodent models points to a role for adenosinergic A1 modulation of EICA. Second, the consistent observation that intracerebellar administration of nicotine in mice leads to antagonism of EICA provides evidence for a critical role of cerebellar nitric oxide (NO) in EICA reversal. Based on these two important findings, this review discusses the potential molecular events at two key synaptic sites (mossy fiber-granule cell-Golgi cell (MGG synaptic site) and granule cell parallel fiber-Purkinje cell (GPP synaptic site) that lead to EICA. Specifically, ethanol-induced neuronal NOS inhibition at the MGG synaptic site acts as a critical trigger for Golgi cell activation which leads to granule cell deafferentation. Concurrently, ethanol-induced inhibition of adenosine uptake at the GPP synaptic site produces adenosine accumulation which decreases glutamate release and leads to the profound activation of Purkinje cells (PCs). These molecular events at the MGG and GPP synaptic sites are mutually reinforcing and lead to cerebellar dysfunction, decreased excitatory output of deep cerebellar nuclei, and EICA. The critical importance of PCs as the sole output of the cerebellar cortex suggests normalization of PC function could have important therapeutic implications.
Collapse
Affiliation(s)
- M Saeed Dar
- Department of Pharmacology and Toxicology, Brody School of Medicine, East Carolina University, Greenville, NC, 27858, USA,
| |
Collapse
|
10
|
Logrip ML. Phosphodiesterase regulation of alcohol drinking in rodents. Alcohol 2015; 49:795-802. [PMID: 26095589 DOI: 10.1016/j.alcohol.2015.03.007] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Revised: 03/13/2015] [Accepted: 03/14/2015] [Indexed: 12/22/2022]
Abstract
Alcohol use disorders are chronically relapsing conditions characterized by persistent drinking despite the negative impact on one's life. The difficulty of achieving and maintaining sobriety suggests that current treatments fail to fully address the underlying causes of alcohol use disorders. Identifying additional pathways controlling alcohol consumption may uncover novel targets for medication development to improve treatment options. One family of proteins recently implicated in the regulation of alcohol consumption is the cyclic nucleotide phosphodiesterases (PDEs). As an integral component in the regulation of the second messengers cyclic AMP and cyclic GMP, and thus their cognate signaling pathways, PDEs present intriguing targets for pharmacotherapies to combat alcohol use disorders. As activation of cAMP/cGMP-dependent signaling cascades can dampen alcohol intake, PDE inhibitors may provide a novel target for reducing excessive alcohol consumption, as has been proposed for PDE4 and PDE10A. This review highlights preclinical literature demonstrating the involvement of cyclic nucleotide-dependent signaling in neuronal and behavioral responses to alcohol, as well as detailing the capacity of various PDE inhibitors to modulate alcohol intake. Together these data provide a framework for evaluating the potential utility of PDE inhibitors as novel treatments for alcohol use disorders.
Collapse
|
11
|
Rashid MA, Kim HY. N-Docosahexaenoylethanolamine ameliorates ethanol-induced impairment of neural stem cell neurogenic differentiation. Neuropharmacology 2015; 102:174-85. [PMID: 26586023 DOI: 10.1016/j.neuropharm.2015.11.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 10/08/2015] [Accepted: 11/06/2015] [Indexed: 11/16/2022]
Abstract
Previous studies demonstrated that prenatal exposure to ethanol interferes with embryonic and fetal development, and causes abnormal neurodevelopment. Docosahexaenoic acid (DHA), an omega-3 polyunsaturated fatty acid highly enriched in the brain, was shown to be essential for proper brain development and function. Recently, we found that N-docosahexenoyethanolamine (synaptamide), an endogenous metabolite of DHA, is a potent PKA-dependent neurogenic factor for neural stem cell (NSC) differentiation. In this study, we demonstrate that ethanol at pharmacologically relevant concentrations downregulates cAMP signaling in NSC and impairs neurogenic differentiation. In contrast, synaptamide reverses ethanol-impaired NSC neurogenic differentiation through counter-acting on the cAMP production system. NSC exposure to ethanol (25-50 mM) for 4 days dose-dependently decreased the number of Tuj-1 positive neurons and PKA/CREB phosphorylation with a concomitant reduction of cellular cAMP. Ethanol-induced cAMP reduction was accompanied by the inhibition of G-protein activation and expression of adenylyl cyclase (AC) 7 and AC8, as well as PDE4 upregulation. In contrast to ethanol, synaptamide increased cAMP production, GTPγS binding, and expression of AC7 and AC8 isoforms in a cAMP-dependent manner, offsetting the ethanol-induced impairment in neurogenic differentiation. These results indicate that synaptamide can reduce ethanol-induced impairment of neuronal differentiation by counter-affecting shared targets in G-protein coupled receptor (GPCR)/cAMP signaling. The synaptamide-mediated mechanism observed in this study may offer a possible avenue for ameliorating the adverse impact of fetal alcohol exposure on neurodevelopment.
Collapse
Affiliation(s)
- Mohammad Abdur Rashid
- Laboratory of Molecular Signaling, DICBR, NIAAA, NIH, 5625 Fishers Lane, Bethesda, MD 20892-9410, USA
| | - Hee-Yong Kim
- Laboratory of Molecular Signaling, DICBR, NIAAA, NIH, 5625 Fishers Lane, Bethesda, MD 20892-9410, USA.
| |
Collapse
|
12
|
Bakoyiannis I, Gkioka E, Pergialiotis V, Mastroleon I, Prodromidou A, Vlachos GD, Perrea D. Fetal alcohol spectrum disorders and cognitive functions of young children. Rev Neurosci 2014; 25:631-9. [PMID: 24978898 DOI: 10.1515/revneuro-2014-0029] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Accepted: 05/29/2014] [Indexed: 12/18/2022]
Abstract
Fetal alcohol spectrum disorder (FASD) is one of the main causes of mental retardation worldwide. Nearly 1% of children in North America are affected from antenatal exposure to ethanol. Its economic burden in industrialized countries is increasing. It is estimated that, in the United States, 4.0 billion dollars are annually expended in the treatment and rehabilitation of these patients. As a pathologic entity, they present with a broad symptomatology. Fetal alcohol syndrome (FAS) is the most readily recognized clinical manifestation of these disorders. Various factors seem to contribute in the pathogenesis of FASD-related cognitive disorders. During the last 20 years, several potential pretranslational and posttranslational factors have been extensively studied in various experimental animal models. Research has specifically focused on several neurotransmitters, insulin resistance, alterations of the hypothalamic-pituitary-adrenal (HPA) axis, abnormal glycosylation of several proteins, oxidative stress, nutritional antioxidants, and various epigenetic factors. The purpose of the present review is to summarize the clinical manifestations of this disorder during childhood and adolescence and to summarize the possible pathophysiologic and epigenetic pathways that have been implicated in the pathophysiology of FASD.
Collapse
|
13
|
Marie N, Noble F. Dépendance aux drogues : avancées de la neurobiologie et perspectives thérapeutiques. Presse Med 2012; 41:1259-70. [DOI: 10.1016/j.lpm.2012.07.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Accepted: 07/31/2012] [Indexed: 02/08/2023] Open
|
14
|
Akula KK, Kulkarni SK. Adenosinergic system: an assorted approach to therapeutics for drug addiction. FUTURE NEUROLOGY 2012. [DOI: 10.2217/fnl.12.19] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Adenosine is an endogenous purine nucleoside and it is extensively present in the brain. It exerts several metabolic and neuromodulatory roles in the body. Adenosine also acts as an important messenger molecule for extracellular signaling and shows a homeostatic neuromodulatory function at the synaptic level. Extracellular adenosine exerts a wide variety of biological actions through four cell surface G-protein-coupled receptor subtypes, namely A1, A2A, A2B and A3 adenosine receptors. The extracellular levels of adenosine have been found to be enhanced in several neuropathological conditions, including drug addiction, and thus a neuroprotective role of adenosine was perceived by various experimental studies. The aversive withdrawal symptoms emanating from drug discontinuation provokes rebound drug intake patterns. In addition, alteration of neurotransmitter(s) release and changes in receptor expression contribute to the behavioral changes of drug withdrawal. Furthermore, the abuse of major drugs such as alcohol and opioids are reported to modulate extracellular adenosine levels. In this context, the neuromodulatory functions of adenosine would be valuable if projected to the clinical applications and thus, an increasing attention is currently given to the functional role of adenosine in human addictive disorders. This review will focus on recent clinical and experimental studies that reveal the actions of adenosine and related ligands in drug addiction and various drug-withdrawal syndromes. The evidence and reports provided in this review highlight the looming therapeutic potential of purinergic drugs, with a hope that new therapeutic interventions based on the adenosinergic concept will emerge in the coming years for the management of drug withdrawal syndrome.
Collapse
Affiliation(s)
- Kiran Kumar Akula
- R.S. Dow Neurobiology Laboratories, Legacy Research, 1225 NE 2nd Avenue, Portland, OR 97232, USA
| | - SK Kulkarni
- Pharmacology Division, University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh-160014, India
| |
Collapse
|
15
|
Kim JH, Karpyak VM, Biernacka JM, Nam HW, Lee MR, Preuss UW, Zill P, Yoon G, Colby C, Mrazek DA, Choi DS. Functional role of the polymorphic 647 T/C variant of ENT1 (SLC29A1) and its association with alcohol withdrawal seizures. PLoS One 2011; 6:e16331. [PMID: 21283641 PMCID: PMC3026043 DOI: 10.1371/journal.pone.0016331] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2010] [Accepted: 12/11/2010] [Indexed: 01/10/2023] Open
Abstract
Background Adenosine is involved in several neurological and behavioral disorders including alcoholism. In cultured cell and animal studies, type 1 equilibrative nucleoside transporter (ENT1, slc29a1), which regulates adenosine levels, is known to regulate ethanol sensitivity and preference. Interestingly, in humans, the ENT1 (SLC29A1) gene contains a non-synonymous single nucleotide polymorphism (647 T/C; rs45573936) that might be involved in the functional change of ENT1. Principal Findings Our functional analysis showed that prolonged ethanol exposure increased adenosine uptake activity of mutant cells (ENT1-216Thr) compared to wild-type (ENT1-216Ile) transfected cells, which might result in reduced extracellular adenosine levels. We found that mice lacking ENT1 displayed increased propensity to ethanol withdrawal seizures compared to wild-type littermates. We further investigated a possible association of the 647C variant with alcoholism and the history of alcohol withdrawal seizures in subjects of European ancestry recruited from two independent sites. Analyses of the combined data set showed an association of the 647C variant and alcohol dependence with withdrawal seizures at the nominally significant level. Conclusions Together with the functional data, our findings suggest a potential contribution of a genetic variant of ENT1 to the development of alcoholism with increased risk of alcohol withdrawal-induced seizures in humans.
Collapse
Affiliation(s)
- Jeong-Hyun Kim
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, Minnesota, United States of America
| | - Victor M. Karpyak
- Department of Psychiatry, Mayo Clinic College of Medicine, Rochester, Minnesota, United States of America
| | - Joanna M. Biernacka
- Department of Psychiatry, Mayo Clinic College of Medicine, Rochester, Minnesota, United States of America
- Department of Health Sciences Research, Mayo Clinic College of Medicine, Rochester, Minnesota, United States of America
| | - Hyung Wook Nam
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, Minnesota, United States of America
| | - Moonnoh R. Lee
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, Minnesota, United States of America
| | - Ulrich W. Preuss
- Department of Psychiatry, Psychotherapy and Psychosomatics, Martin-Luther-University, Halle/Saale, Germany
| | - Peter Zill
- Section Psychiatric Genetics and Neurochemistry, Ludwig-Maximilians University of Munich, Munich, Germany
| | - Gihyun Yoon
- Department of Psychiatry, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Colin Colby
- Department of Health Sciences Research, Mayo Clinic College of Medicine, Rochester, Minnesota, United States of America
| | - David A. Mrazek
- Department of Psychiatry, Mayo Clinic College of Medicine, Rochester, Minnesota, United States of America
| | - Doo-Sup Choi
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, Minnesota, United States of America
- Department of Psychiatry, Mayo Clinic College of Medicine, Rochester, Minnesota, United States of America
- * E-mail:
| |
Collapse
|
16
|
Asatryan L, Nam HW, Lee MR, Thakkar MM, Saeed Dar M, Davies DL, Choi DS. Implication of the purinergic system in alcohol use disorders. Alcohol Clin Exp Res 2011; 35:584-94. [PMID: 21223299 DOI: 10.1111/j.1530-0277.2010.01379.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
In the central nervous system, adenosine and adenosine 5'-triphosphate (ATP) play an important role in regulating neuronal activity as well as controlling other neurotransmitter systems, such as, GABA, glutamate, and dopamine. Ethanol increases extracellular adenosine levels that regulate the ataxic and hypnotic/sedative effects of ethanol. Interestingly, ethanol is known to increase adenosine levels by inhibiting an ethanol-sensitive adenosine transporter, equilibrative nucleoside transporter type 1 (ENT1). Ethanol is also known to inhibit ATP-specific P2X receptors, which might result in such similar effects as those caused by an increase in adenosine. Adenosine and ATP exert their functions through P1 (metabotropic) and P2 (P2X-ionotropic and P2Y-metabotropic) receptors, respectively. Purinergic signaling in cortex-striatum-ventral tegmental area (VTA) has been implicated in regulating cortical glutamate signaling as well as VTA dopaminergic signaling, which regulates the motivational effect of ethanol. Moreover, several nucleoside transporters and receptors have been identified in astrocytes, which regulate not only adenosine-ATP neurotransmission, but also homeostasis of major inhibitory-excitatory neurotransmission (i.e., GABA or glutamate) through neuron-glial interactions. This review will present novel findings on the implications of adenosine and ATP neurotransmission in alcohol use disorders.
Collapse
Affiliation(s)
- Liana Asatryan
- Department of Clinical Pharmacy and Pharmaceutical Economics and Policy, University of Southern California, Los Angeles, Los Angeles, California, USA
| | | | | | | | | | | | | |
Collapse
|
17
|
de la Monte SM, Wands JR. Role of central nervous system insulin resistance in fetal alcohol spectrum disorders. JOURNAL OF POPULATION THERAPEUTICS AND CLINICAL PHARMACOLOGY = JOURNAL DE LA THERAPEUTIQUE DES POPULATIONS ET DE LA PHARMACOLOGIE CLINIQUE 2010; 17:e390-e404. [PMID: 21063035 PMCID: PMC3113413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Fetal alcohol spectrum disorder (FASD) is the most common preventable cause of mental retardation in the USA. Ethanol impairs neuronal survival and function by two major mechanisms: 1) it inhibits insulin signaling required for viability, metabolism, synapse formation, and acetylcholine production; and 2) it functions as a neurotoxicant, causing oxidative stress, DNA damage and mitochondrial dysfunction. Ethanol inhibition of insulin signaling is mediated at the insulin receptor (IR) level and caused by both impaired receptor binding and increased activation of phosphatases that reverse IR tyrosine kinase activity. As a result, insulin activation of PI3K-Akt, which mediates neuronal survival, motility, energy metabolism, and plasticity, is impaired. The neurotoxicant effects of ethanol promote DNA damage, which could contribute to mitochondrial dysfunction and oxidative stress. Therefore, chronic in utero ethanol exposure produces a dual state of CNS insulin resistance and oxidative stress, which we postulate plays a major role in ethanol neurobehavioral teratogenesis. We propose that many of the prominent adverse effects of chronic prenatal exposure to ethanol on CNS development and function may be prevented or reduced by treatment with peroxisome-proliferated activated receptor (PPAR) agonists which enhance insulin sensitivity by increasing expression and function of insulin-responsive genes, and reducing cellular oxidative stress.
Collapse
Affiliation(s)
- Suzanne M de la Monte
- Department of Pathology, Rhode Island Hospital and the Warren Alpert School of Medicine at Brown University, Providence, RI, USA.
| | | |
Collapse
|
18
|
Abstract
Abstract
The specific events between initial presumably manageable drug intake and the development of a drug- addicted state are not yet known. Drugs of abuse have varying mechanisms of action that create a complex pattern of behaviour related to drug consumption, drug-seeking, withdrawal and relapse. The neuromodulator adenosine has been shown to play a role in reward-related behaviour, both as an independent mediator and via interactions of adenosine receptors with other receptors. Adenosine levels are elevated upon exposure to drugs of abuse and adenosine A2A receptors are enriched in brain nuclei known for their involvement in the processing of drug-related reinforcement processing. A2A receptors are found in receptor clusters with dopamine and glutamate receptors. A2A receptors are thus ideally situated to influence the signalling of neurotransmitters relevant in the neuronal responses and plasticity that underlie the development of drug taking and drug-seeking behaviour. In this review, we present evidence for the role of adenosine and A2A receptors in drug addiction, thereby providing support for current efforts aimed at developing drug therapies to combat substance abuse that target adenosine signalling via A2A receptors.
Collapse
Affiliation(s)
- Robyn M Brown
- Medicinal Chemistry and Drug Action, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), 381 Royal Parade, Parkville, 3052, Australia
| | - Jennifer L Short
- Medicinal Chemistry and Drug Action, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), 381 Royal Parade, Parkville, 3052, Australia
| |
Collapse
|
19
|
Kalant H. Alcohol Tolerance, Dependence and Withdrawal: an Overview of Current Issues. ACTA ACUST UNITED AC 2009. [DOI: 10.1080/09595238880000081] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
20
|
Federici M, Nisticò R, Giustizieri M, Bernardi G, Mercuri NB. Ethanol enhances GABAB-mediated inhibitory postsynaptic transmission on rat midbrain dopaminergic neurons by facilitating GIRK currents. Eur J Neurosci 2009; 29:1369-77. [DOI: 10.1111/j.1460-9568.2009.06700.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
21
|
Allen-Gipson DS, Jarrell JC, Bailey KL, Robinson JE, Kharbanda KK, Sisson JH, Wyatt TA. Ethanol blocks adenosine uptake via inhibiting the nucleoside transport system in bronchial epithelial cells. Alcohol Clin Exp Res 2009; 33:791-8. [PMID: 19298329 DOI: 10.1111/j.1530-0277.2009.00897.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND Adenosine uptake into cells by nucleoside transporters plays a significant role in governing extracellular adenosine concentration. Extracellular adenosine is an important signaling molecule that modulates many cellular functions via 4 G-protein-coupled receptor subtypes (A(1), A(2A), A(2B), and A(3)). Previously, we demonstrated that adenosine is critical in maintaining airway homeostasis and airway repair and that airway host defenses are impaired by alcohol. Taken together, we hypothesized that ethanol impairs adenosine uptake via the nucleoside transport system. METHODS To examine ethanol-induced alteration on adenosine transport, we used a human bronchial epithelial cell line (BEAS-2B). Cells were preincubated for 10 minutes in the presence and absence of varying concentrations of ethanol (EtOH). In addition, some cells were pretreated with S-(4-Nitrobenzyl)-6-thioinosine (100 microM: NBT), a potent adenosine uptake inhibitor. Uptake was then determined by addition of [(3)H]-adenosine at various time intervals. RESULTS Increasing EtOH concentrations resulted in increasing inhibition of adenosine uptake when measured at 1 minute. Cells pretreated with NBT effectively blocked adenosine uptake. In addition, short-term EtOH revealed increased extracellular adenosine concentration. Conversely, adenosine transport became desensitized in cells exposed to EtOH (100 mM) for 24 hours. To determine the mechanism of EtOH-induced desensitization of adenosine transport, cAMP activity was assessed in response to EtOH. Short-term EtOH exposure (10 minutes) had little or no effect on adenosine-mediated cAMP activation, whereas long-term EtOH exposure (24 hours) blocked adenosine-mediated cAMP activation. Western blot analysis of lysates from unstimulated BEAS-2B cells detected a single 55 kDa band indicating the presence of hENT1 and hENT2, respectively. Real-time RT-PCR of RNA from BEAS-2B revealed transcriptional expression of ENT1 and ENT2. CONCLUSIONS Collectively, these data reveal that acute exposure of cells to EtOH inhibits adenosine uptake via a nucleoside transporter, and chronic exposure of cells to EtOH desensitizes the adenosine transporter to these inhibitory effects of ethanol. Furthermore, our data suggest that inhibition of adenosine uptake by EtOH leads to an increased extracellular adenosine accumulation, influencing the effect of adenosine at the epithelial cell surface, which may alter airway homeostasis.
Collapse
Affiliation(s)
- Diane S Allen-Gipson
- Department of Internal Medicine, Pulmonary, Critical Care, Sleep and Allergy Section, 985815 Nebraska Medical Center, Omaha, NE 68198-5815, USA.
| | | | | | | | | | | | | |
Collapse
|
22
|
Marczak ED, Jinsmaa Y, Li T, Bryant SD, Tsuda Y, Okada Y, Lazarus LH. [N-allyl-Dmt1]-endomorphins are micro-opioid receptor antagonists lacking inverse agonist properties. J Pharmacol Exp Ther 2007; 323:374-80. [PMID: 17626793 DOI: 10.1124/jpet.107.125807] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
[N-allyl-Dmt1]-endomorphin-1 and -2 ([N-allyl-Dmt1]-EM-1 and -2) are new selective micro-opioid receptor antagonists obtained by N-alkylation with an allyl group on the amino terminus of 2',6'-dimethyl-L-tyrosine (Dmt) derivatives. To further characterize properties of these compounds, their intrinsic activities were assessed by functional guanosine 5'-O-(3-[35S]thiotriphosphate) binding assays and forskolin-stimulated cyclic AMP accumulation in cell membranes obtained from vehicle, morphine, and ethanol-treated SK-N-SH cells and brain membranes isolated from naive and morphine-dependent mice; their mode of action was compared with naloxone or naltrexone, which both are standard nonspecific opioid-receptor antagonists. [N-allyl-Dmt1]-EM-1 and -2 were neutral antagonists under all of the experimental conditions examined, in contrast to naloxone and naltrexone, which behave as neutral antagonists only in membranes from vehicle-treated cells and mice but act as inverse agonists in membranes from morphine- and ethanol-treated cells as well as morphine-treated mice. Both endomorphin analogs inhibited the naloxone- and naltrexone-elicited withdrawal syndromes from acute morphine dependence in mice. This suggests their potential therapeutic application in the treatment of drug addiction and alcohol abuse without the adverse effects observed with inverse agonist alkaloid-derived compounds that produce severe withdrawal symptoms.
Collapse
Affiliation(s)
- Ewa D Marczak
- Medicinal Chemistry Group, Laboratory of Pharmacology and Chemistry, National Institute of Environmental Health Sciences, P.O. Box 12233, MD C304, Research Triangle Park, NC 27709, USA.
| | | | | | | | | | | | | |
Collapse
|
23
|
Pal-Bhadra M, Bhadra U, Jackson DE, Mamatha L, Park PH, Shukla SD. Distinct methylation patterns in histone H3 at Lys-4 and Lys-9 correlate with up- & down-regulation of genes by ethanol in hepatocytes. Life Sci 2007; 81:979-87. [PMID: 17826801 PMCID: PMC2706023 DOI: 10.1016/j.lfs.2007.07.030] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2007] [Revised: 07/03/2007] [Accepted: 07/18/2007] [Indexed: 10/22/2022]
Abstract
Ethanol induced liver injury is associated with a global change in gene expression but its mechanisms are not known. We studied whether alcohol-induced gene expression is associated with post-translational methylations of histone H3. Primary culture of rat hepatocytes was treated with ethanol (50 or 100 mM) for 24 h and the status of methylation of H3 at lys 4 (H3dimeK4) or lys 9 (H3dimeK9) was monitored by Western blotting using antibodies to dimethylated histone H3 at lys 4 or lys 9. The cells exposed to ethanol showed strikingly opposing behaviors in methylation patterns; H3dimeK9 methylation was decreased whereas H3dimeK4 increased. Similar results were obtained in the interphase nuclei. Their binding on the metaphase chromosomes exhibits distinct site specific pattern of accumulation. Next, chromatin immunoprecipitation of the ethanol treated samples with antibodies for methylated lys 4 or lys 9 histone H3 followed by amplification of the immunoprecipitated DNA, was used to determine their association with the promoters of genes up- or downregulated by ethanol. Lys4 methylation was associated with ethanol upregulated genes (Adh, GST-yc2) whereas lys 9 methylation with downregulated genes (Lsdh, cytP4502c11) demonstrating a difference between these two methylations. These results suggest that exposure of hepatocytes to ethanol changes the expression of several susceptible genes which are associated with site specific modification of dimethylated forms of histone H3 amino termini at their regulatory regions.
Collapse
Affiliation(s)
- Manika Pal-Bhadra
- Department of Chemical Biology, Indian Institute of Chemical Technology, Hyderabad-500007, India
| | - Utpal Bhadra
- Functional Genomics & Gene Silencing Group, Centre for Cellular & Molecular Biology, Hyderabad-500007, India
| | - Daniel E. Jackson
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri-Columbia, MO-65212 USA
| | - Linga Mamatha
- Functional Genomics & Gene Silencing Group, Centre for Cellular & Molecular Biology, Hyderabad-500007, India
- Department of Chemical Biology, Indian Institute of Chemical Technology, Hyderabad-500007, India
| | - Pil-Hoon Park
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri-Columbia, MO-65212 USA
| | - Shivendra D. Shukla
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri-Columbia, MO-65212 USA
| |
Collapse
|
24
|
Basavarajappa BS. The endocannabinoid signaling system: a potential target for next-generation therapeutics for alcoholism. Mini Rev Med Chem 2007; 7:769-79. [PMID: 17692039 PMCID: PMC1975858 DOI: 10.2174/138955707781387920] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Research into the endocannabinoid signaling system has grown exponentially in recent years following the discovery of cannabinoid receptors (CB) and their endogenous ligands, such as anandamide (AEA) and 2-arachidonoylglycerol (2-AG). Important advances have been made in our understanding of the endocannabinoid signaling system in various aspects of alcoholism, including alcohol-seeking behavior. Alcohol increases the synthesis or impairs the degradation of endocannabinoids, leading to a locally elevated endocannabinoid tone within the brain. Elevated endocannabinoid tone might be expected to result in compensatory down-regulation of CB1 receptors or dampened signal transduction. Following release, endocannabinoids diffuse back to the presynaptic neuron where they act as short-range modulators of synaptic activity by altering neurotransmitter release and synaptic plasticity. Mice treated with the CB1 receptor antagonist SR141716A (rimonabant) or homozygous for a deletion of the CB1 receptor gene exhibit reduced voluntary alcohol intake. CB1 knockout mice also show increased alcohol sensitivity, withdrawal, and reduced conditioned place preference. Conversely, activation of CB1 receptor promotes alcohol intake. Recent studies also suggest that elevated endocannabinoid tone due to impaired degradation contributes to high alcohol preference and self-administration. These effects are reversed by local administration of rimonabant, suggesting the participation of the endocannabinoid signaling system in high alcohol preference and self-administration. These recent advances will be reviewed with an emphasis on the endocannabinoid signaling system for possible therapeutic interventions of alcoholism.
Collapse
Affiliation(s)
- Balapal S Basavarajappa
- Division of Analytical Psychopharmacology, New York State Psychiatric Institute, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
25
|
Thorsell A, Johnson J, Heilig M. Effect of the adenosine A2a receptor antagonist 3,7-dimethyl-propargylxanthine on anxiety-like and depression-like behavior and alcohol consumption in Wistar Rats. Alcohol Clin Exp Res 2007; 31:1302-7. [PMID: 17550371 DOI: 10.1111/j.1530-0277.2007.00425.x] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND It has been suggested that the reinforcing properties of ethanol are in part mediated via an A2 activation of cAMP/PKA signaling in the nucleus accumbens, predicting that administration of an A2a antagonist might reduce ethanol reward and consumption. We therefore examined the effect of the adenosine A2a receptor antagonist 3,7-dimethylpropargylxanthine (DMPX, 3, and 10 mg/kg intraperitoneal) on alcohol reinforcement, anxiety-related, depression, and rewarding behaviors in nonselected Wistar rats. METHODS Operant ethanol self-administration was used for examining alcohol intake, elevated plus-maze and Vogel conflict test for anxiety-related behavior, Porsolt swim test for depression-like behavior, and conditioned place preference for examination of the rewarding properties of the drug. RESULTS 3,7-Dimethylpropargylxanthine decreased lever-pressing for ethanol in a dose-dependent manner. When analyzed as percentage of pretreatment baseline, maximum suppression was approximately 60% (39+/-7.5 vs 98+/-12%, mean+/-SEM, p=0.017). This effect was behaviorally specific, as no effect was found on the water lever. In agreement with previously published data, stimulation of locomotion was found (beam-breaks: 3590+/-540 vs 2475+/-240, 10 mg/kg vs saline, p=0.048). No anxiety-modulating effects were seen in either the elevated plus-maze or the Vogel conflict test. 3,7-Dimethylpropargylxanthine was not found to have intrinsic rewarding properties in the conditioned place preference model. CONCLUSIONS In summary, DMPX produced a robust and behaviorally selective reduction of ethanol reinforcement, while anxiety-modulating effects were less consistent. These results bring further support to a role for adenosine in the regulation of ethanol consumption and possibly alcohol addiction/abuse, and the A2a receptor as a potential target for the treatment of alcoholism and alcohol abuse.
Collapse
Affiliation(s)
- Annika Thorsell
- Laboratory of Clinical and Translational Studies, NIAAA, NIH, Bethesda, Maryland 20892-1108, USA.
| | | | | |
Collapse
|
26
|
Schoenfeld H, Franke R, Von Heymann C, Doepfmer UR, Blaicher AM, Ziemer S, Spies C. Peri-operative decreased cAMP levels in long-term alcoholic patients. J Int Med Res 2007; 34:445-55. [PMID: 17133773 DOI: 10.1177/147323000603400501] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Long-term alcoholic patients have a five-fold higher risk of post-operative bleeding complications compared with nonalcoholic individuals. Serotonin increases and cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP) decrease platelet aggregation. We examined the platelet-rich plasma levels of these substances and agonist-induced platelet aggregation in long-term alcoholic patients before and after surgery. Thirty-three consecutive patients (13 long-term alcoholics and 20 non-alcoholics) scheduled for tumour resections of the upper digestive tract were included in the study. The levels of cAMP were significantly decreased before and after surgery in long-term alcoholic patients, but there were no significant differences in cGMP and serotonin levels in alcoholic compared with non-alcoholic patients. In contrast to previous studies, no significantly altered aggregation responses in long-term alcoholics were found. A possible explanation is decreased inhibition through diminished cAMP levels; cGMP and serotonin do not seem to influence peri-operative haemostasis.
Collapse
Affiliation(s)
- H Schoenfeld
- Department of Anaesthesiology and Intensive Care Medicine, Charité-Universitätsmedizin Berlin, Berlin, Germany.
| | | | | | | | | | | | | |
Collapse
|
27
|
Park PH, Lim RW, Shukla SD. Involvement of histone acetyltransferase (HAT) in ethanol-induced acetylation of histone H3 in hepatocytes: potential mechanism for gene expression. Am J Physiol Gastrointest Liver Physiol 2005; 289:G1124-36. [PMID: 16081763 DOI: 10.1152/ajpgi.00091.2005] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Ethanol treatment increases gene expression in the liver through mechanisms that are not clearly understood. Histone acetylation has been shown to induce transcriptional activation. We have investigated the characteristics and mechanisms of ethanol-induced histone H3 acetylation in rat hepatocytes. Immunocytochemical and immunoblot analysis revealed that ethanol treatment significantly increased H3 acetylation at Lys9 with negligible effects at Lys14, -18, and -23. Acute in vivo administration of alcohol in rats produced the same results as in vitro observations. Nuclear extracts from ethanol-treated hepatocytes increased acetylation in H3 peptide to a greater extent than extracts from untreated cells, suggesting that ethanol either increased the expression level or the specific activity of histone acetyltransferases (HAT). Use of different H3 peptides indicated that ethanol selectively modulated HAT(s) targeting H3-Lys9. Treatment with acetate, an ethanol metabolite, also increased acetylation of H3-Lys9 and modulated HAT(s) in the same manner as ethanol, suggesting that acetate mediates the ethanol-induced effect on HAT. Inhibitors of MEK (U0126) and JNK (SP600125), but not p38 MAPK inhibitor (SB203580), suppressed ethanol-induced H3 acetylation. However, U0126 and SP600125 did not significantly affect ethanol-induced effect on HAT, suggesting that ERK and JNK regulate histone acetylation through a separate pathway(s) that does not involve modulation of HAT. Chromatin immunoprecipitation assay demonstrated that ethanol treatment increased the association of the class I alcohol dehydrogenase (ADH I) gene with acetylated H3-Lys9. These data provide first evidence that ethanol increases acetylation of H3-Lys9 through modulation of HAT(s) and that histone acetylation may underlie the mechanism for ethanol-induced ADH I gene expression.
Collapse
Affiliation(s)
- Pil-Hoon Park
- Dept. of Medical Pharmacology and Physiology, School of Medicine, University of Missouri-Columbia, MO 65212, USA
| | | | | |
Collapse
|
28
|
Li Z, Kang SS, Lee S, Rivier C. Effect of ethanol on the regulation of corticotropin-releasing factor (CRF) gene expression. Mol Cell Neurosci 2005; 29:345-54. [PMID: 15914027 DOI: 10.1016/j.mcn.2005.04.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2004] [Revised: 02/09/2005] [Accepted: 04/06/2005] [Indexed: 11/20/2022] Open
Abstract
Ethanol stimulates hypothalamic-pituitary-adrenal axis activity in vivo. To determine the cellular and molecular mechanisms through which ethanol regulates corticotropin-releasing factor (CRF) gene expression, we compared the effect of ethanol and forskolin on CRF peptide secretion and messenger RNA levels in hypothalamic primary cell cultures, and on CRF promoter activity in the NG108-15 cell line. CRF secretion, mRNA levels, and gene transcription significantly increased in response to ethanol or forskolin. Mutation of the cAMP-response element (CRE) reduced luciferase activity under basal conditions as well as in response to forskolin or ethanol. On the other hand, plasmid with five CRE repeats yielded dramatically elevated basal luciferase activity and significantly increased upregulation by ethanol. Inclusion of adenosine deaminase reduced the promoter response to ethanol. Finally a PKA inhibitor and a cAMP antagonist both decreased ethanol-induced CRF peptide secretion, gene expression, and transcription. These results suggest that ethanol upregulates CRF expression through cAMP/PKA-dependent pathways.
Collapse
Affiliation(s)
- Zhongqi Li
- The Clayton Foundation Laboratories for Peptide Biology, The Salk Institute, 10010 N. Torrey Pines Road, La Jolla, CA 92037, USA
| | | | | | | |
Collapse
|
29
|
Poplawski MM, Boyadjieva N, Sarkar DK. Vasoactive intestinal peptide and corticotropin-releasing hormone increase beta-endorphin release and proopiomelanocortin messenger RNA levels in primary cultures of hypothalamic cells: effects of acute and chronic ethanol treatment. Alcohol Clin Exp Res 2005; 29:648-55. [PMID: 15834231 DOI: 10.1097/01.alc.0000158834.11252.2e] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND beta-Endorphin (beta-EP) neurons are involved in ethanol's action on a variety of brain functions, including positive reinforcement. These neurons are innervated by vasoactive intestinal peptide (VIP)-containing and corticotropin-releasing hormone (CRH)-containing neurons in the hypothalamus. Whether these neuropeptides affect beta-EP neuronal function in the presence or absence of ethanol has not previously been determined. METHODS The authors determined the effects of VIP and CRH on gene expression and peptide release from beta-EP neurons in primary cultures of mediobasal hypothalamic cells. The effects of receptor antagonists on VIP- and CRH-induced beta-EP release was determined. Furthermore, the authors studied the effects of acute and chronic treatment with ethanol on the response of beta-EP neurons to VIP and CRH. Real-time reverse-transcription polymerase chain reaction was used for messenger RNA (mRNA) detection, and radioimmunoassay was used for hormone measurements. RESULTS We show that beta-EP neurons responded concentration dependently to VIP and CRH treatments by increasing both beta-EP release and proopiomelanocortin mRNA expression. Simultaneous treatment with a nonspecific receptor antagonist reduced the ability of CRH or VIP to induce beta-EP release from mediobasal hypothalamic cells. Acute treatment with ethanol increased beta-EP neuronal gene expression and the secretory response to CRH and VIP. However, previous exposure to chronic ethanol reduced the CRH and VIP responses of these neurons. CONCLUSIONS These results indicate that VIP and CRH stimulate beta-EP release from hypothalamic cells in primary cultures and that the stimulatory and adaptive responses of beta-EP neurons to ethanol may involve alteration in the responsiveness of beta-EP-secreting neurons to CRH and VIP.
Collapse
Affiliation(s)
- Michael M Poplawski
- Endocrinology Program, Center of Alcohol Studies and Department of Animal Sciences, Rutgers, The State University of New Jersey, New Brunswick, New Jersey 08901-8525, USA
| | | | | |
Collapse
|
30
|
Kralic JE, Criswell HE, Osterman JL, O'Buckley TK, Wilkie ME, Matthews DB, Hamre K, Breese GR, Homanics GE, Morrow AL. Genetic essential tremor in gamma-aminobutyric acidA receptor alpha1 subunit knockout mice. J Clin Invest 2005; 115:774-9. [PMID: 15765150 PMCID: PMC1052003 DOI: 10.1172/jci23625] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2004] [Accepted: 12/21/2004] [Indexed: 12/16/2022] Open
Abstract
Essential tremor is the most common movement disorder and has an unknown etiology. Here we report that gamma-aminobutyric acidA (GABA(A)) receptor alpha1-/- mice exhibit postural and kinetic tremor and motor incoordination that is characteristic of essential tremor disease. We tested mice with essential-like tremor using current drug therapies that alleviate symptoms in essential tremor patients (primidone, propranolol, and gabapentin) and several candidates hypothesized to reduce tremor, including ethanol; the noncompetitive N-methyl-D-aspartate receptor antagonist MK-801; the adenosine A1 receptor agonist 2-chloro-N6-cyclopentyladenosine (CCPA); the GABA(A) receptor modulators diazepam, allopregnanolone, and Ro15-4513; and the L-type Ca2+ channel antagonist nitrendipine. Primidone, propranolol, and gabapentin reduced the amplitude (power) of the pathologic tremor. Nonsedative doses of ethanol eliminated tremor in mice. Diazepam, allopregnanolone, Ro15-4513, and nitrendipine had no effect or enhanced tremor, whereas MK-801 and CCPA reduced tremor. To understand the etiology of tremor in these mice, we studied the electrophysiological properties of cerebellar Purkinje cells. Cerebellar Purkinje cells in GABA(A) receptor alpha1-/- mice exhibited a profound loss of all responses to synaptic or exogenous GABA, but no differences in abundance, gross morphology, or spontaneous synaptic activity were observed. This genetic animal model elucidates a mechanism of GABAergic dysfunction in the major motor pathway and potential targets for pharmacotherapy of essential tremor.
Collapse
Affiliation(s)
- Jason E Kralic
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, North Carolina 27599-7178, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Kralic JE, Criswell HE, Osterman JL, O’Buckley TK, Wilkie ME, Matthews DB, Hamre K, Breese GR, Homanics GE, Morrow AL. Genetic essential tremor in γ-aminobutyric acidA receptor α1 subunit knockout mice. J Clin Invest 2005. [DOI: 10.1172/jci200523625] [Citation(s) in RCA: 135] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
32
|
Arolfo MP, Yao L, Gordon AS, Diamond I, Janak PH. Ethanol operant self-administration in rats is regulated by adenosine A2 receptors. Alcohol Clin Exp Res 2004; 28:1308-16. [PMID: 15365300 DOI: 10.1097/01.alc.0000139821.38167.20] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Recent findings suggest that adenosine is involved in the neural and behavioral effects of ethanol (EtOH). Studies in neural cell culture show that EtOH, via activation of adenosine A2 receptors, triggers cyclic adenosine monophosphate/protein kinase A (cAMP/PKA) signaling and CRE (cAMP regulatory element)-mediated gene expression and that this effect is blocked by inhibiting G-protein betagamma subunits. Recently, we reported that expression of a betagamma inhibitor in the nucleus accumbens (NAc) reduces EtOH drinking in rats. The NAc expresses high levels of the adenosine A2A receptor in GABAergic medium spiny neurons. If the reinforcing effects of EtOH are mediated through an A2 activation of cAMP/PKA signaling via betagamma, then A2 receptor blockade should attenuate EtOH consumption. Here we tested this hypothesis. Because adenosine A2 and dopamine D2 receptors are coexpressed in neurons of the NAc, we compared the effects of A2 blockade with those of D2 receptor blockade. METHODS Male Long-Evans rats were trained to self-administer 10% EtOH in daily 30-min sessions with an active and an inactive lever. Separate groups of rats were given the D2 antagonist eticlopride (0.005, 0.007, and 0.01 mg/kg), the A2 antagonist 3,7-dimethyl-1-propargylxanthine (DMPX; 1, 3, 5, 7, 10, and 20 mg/kg), and the A1 antagonist 8-cyclopentyl-1,3-dipropylxanthine (DPCPX; 0.125, 0.25, and 0.5 mg/kg) by systemic injection. RESULTS Eticlopride dose-dependently reduced EtOH drinking. DMPX showed a bimodal effect: 10 and 20 mg/kg decreased, but 1 mg/kg increased, EtOH consumption. DPCPX was without effect. CONCLUSIONS In support of our hypothesis, the A2 antagonist DMPX attenuated EtOH self-administration. Low doses of the A2 antagonist enhanced EtOH drinking, consistent with the possibility that rats increase EtOH self-administration to overcome partial A2 blockade. The D2 antagonist eticlopride also decreased EtOH self-administration. These data provide the first evidence that pharmacological modulation of adenosine A2 receptors can regulate EtOH consumption in rats.
Collapse
Affiliation(s)
- Maria Pia Arolfo
- Ernest Gallo Clinic and Research Center, Department of Neurology, University of California, San Francisco, Emeryville, California 94608, USA.
| | | | | | | | | |
Collapse
|
33
|
Basavarajappa BS, Hungund BL. ROLE OF THE ENDOCANNABINOID SYSTEM IN THE DEVELOPMENT OF TOLERANCE TO ALCOHOL. Alcohol Alcohol 2004; 40:15-24. [PMID: 15550443 DOI: 10.1093/alcalc/agh111] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The present review evaluates the evidence that the endocannabinoid system plays in the development of tolerance to alcohol. The identification of a G-protein-coupled receptor, namely, the cannabinoid receptor (CB(1) receptor), which was activated by Delta(9)-tetrahydrocannabinol (Delta(9)-THC), the major psychoactive component of marijuana, led to the discovery of endogenous cannabinoid agonists. Until now, four fatty acid derivatives identified to be arachidonylethanolamide (AEA), 2-arachidonylglycerol (2-AG), 2-arachidonylglycerol ether (noladin ether) and virodhamine have been isolated from both nervous and peripheral tissues. Both AEA and 2-AG have been shown to mimic the pharmacological and behavioural effects of Delta(9)-THC. The role of the endocannabinoid system in the development of tolerance to alcohol was not known until recently. Recent studies from our laboratory have implicated for the first time a role for the endocannabinoid system in development of tolerance to alcohol. Chronic alcohol treatment has been shown to down-regulate CB(1) receptors and its signal transduction. The observed downregulation of CB(1) receptor function results from the persistent stimulation of the receptors by AEA and 2-AG, the synthesis of which has been shown to be increased by chronic alcohol treatment. The enhanced formation of endocannabinoids may subsequently influence the release of neurotransmitters. It was found that the DBA/2 mice, known to avoid alcohol intake, have significantly reduced CB(1) receptor function in the brain, consistent with other studies in which the CB(1) receptor antagonist SR 141716A has been shown to block voluntary alcohol intake in rodents. Similarly, activation of the CB(1) receptor system promoted alcohol craving, suggesting a role for the CB(1) receptor gene in excessive alcohol drinking behaviour and development of alcoholism. Ongoing investigations may lead to a better understanding of the mechanisms underlying the development of tolerance to alcohol and to develop therapeutic strategies to treat alcoholism.
Collapse
Affiliation(s)
- Balapal S Basavarajappa
- Nathan Kline Institute for Psychiatric Research, 140 Old Orangeburg Road, Orangeburg, NY 10962, USA.
| | | |
Collapse
|
34
|
Vagts AJ, He DY, Yaka R, Ron D. Cellular adaptation to chronic ethanol results in altered compartmentalization and function of the scaffolding protein RACK1. Alcohol Clin Exp Res 2004; 27:1599-605. [PMID: 14574230 DOI: 10.1097/01.alc.0000089957.63597.a4] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Previously, we found that acute ethanol induces the translocation of the scaffolding protein RACK1 to the nucleus. Recently, we found that nuclear RACK1 mediates acute ethanol induction of immediate early gene c-fos expression. Alterations in gene expression are thought to lead to long-term changes that ultimately contribute to the development of alcohol addiction and toxicity. Therefore, we sought to determine the effects of chronic exposure of cells to ethanol on the cellular compartmentalization of RACK1 and on c-fos messenger RNA (mRNA) and protein expression. METHODS Rat C6 glioma cells were used as the cell culture model. Immunohistochemistry was implemented to visualize the localization of RACK1 and to monitor the protein level of c-fos. Reverse-transcription polymerase chain reaction was used to measure c-fos mRNA levels. The Tat-protein transduction method was used to transduce recombinant Tat-RACK1 into cells as previously described. RESULTS Chronic exposure of cells to 200 mM ethanol for 24 and 48 hr resulted in the gradual re-distribution of RACK1 out of the nucleus. It is interesting to note that acute ethanol re-challenge immediately after chronic treatment did not result in RACK1 translocation to the nucleus, and nuclear compartmentalization of RACK1 in response to acute ethanol was detected only after 24 hr of withdrawal. Similar patterns were obtained for c-fos expression. Chronic exposure to ethanol did not result in an increase in mRNA or protein levels of c-fos. Furthermore, acute ethanol exposure did not increase c-fos protein levels in cells that were first treated chronically with ethanol. However, transduction of exogenous RACK1 expressed as a Tat-fusion protein was able to rescue c-fos mRNA expression after chronic ethanol exposure. CONCLUSIONS Our data suggest that RACK1 nuclear compartmentalization and ethanol-induced c-fos expression are transient and are desensitized to ethanol during prolonged exposure to high concentrations. The desensitization is temporary, and RACK1 can respond to acute ethanol treatment after a 24-hr withdrawal period. Our data further suggest that the altered compartmentalization of RACK1 leads to differences in c-fos expression upon acute or chronic exposure to ethanol. In summary, RACK1 is an important molecular mediator of the acute and chronic actions of ethanol on the expression of c-fos. These findings could have implications for the molecular signaling pathways leading to pathologic states associated with alcoholism, including toxicity.
Collapse
Affiliation(s)
- Alicia J Vagts
- Ernest Gallo Clinic and Research Center, Department of Neurology, University of California, San Francisco, Emeryville, California, USA
| | | | | | | |
Collapse
|
35
|
Abstract
Neuronal responses to alcohol involve several hormone- and neurotransmitter-activated signal transduction pathways. Recent studies suggest that the adenosine A2 receptor (A2) mediates important actions of alcohol. Ethanol inhibits adenosine reuptake, increases extracellular adenosine, and promotes activation of A2. This leads to enhanced cAMP/protein kinase A (PKA) signaling ranging from increases in cAMP to stimulation of cAMP-dependent cAMP response element (CRE)-mediated gene expression. Medium spiny neurons in the striatum/nucleus accumbens (NAc) express A2 and dopamine D2 receptor (D2) on the same cells. Studies in model neuronal cell lines and primary neurons in culture expressing A2 and D2 provide evidence for synergy between ethanol/A2 and D2. Subthreshold concentrations of ethanol or a D2 agonist, without effect separately, synergistically activate cAMP/PKA signaling. Thus, neurons expressing A2 and D2 on the same cells, like in the NAc, are characterized by hypersensitivity to ethanol with a simultaneous activation of dopaminergic signaling. Synergy requires adenosine and appears to be mediated by the release of free betagamma dimers from G(i/o) via D2 activation. The release of free betagamma has pathophysiological significance in the drinking animal because specific blockade of betagamma signaling in the NAc strikingly reduces voluntary alcohol consumption. These findings suggest that signaling pathways, which regulate synergy between A2 and D2, might contain molecular targets for the prevention and treatment of alcoholism and alcohol abuse.
Collapse
Affiliation(s)
- William S Mailliard
- Ernest Gallo Clinic and Research Center, University of California, San Francisco, 5858 Horton Street, Suite 200, , Emeryville, CA 94608, USA
| | | |
Collapse
|
36
|
Deaciuc IV, Peng X, D'Souza NB, Shedlofsky SI, Burikhanov R, Voskresensky IV, de Villiers WJS. Microarray gene analysis of the liver in a rat model of chronic, voluntary alcohol intake. Alcohol 2004; 32:113-27. [PMID: 15163562 DOI: 10.1016/j.alcohol.2003.12.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2003] [Revised: 12/04/2003] [Accepted: 12/11/2003] [Indexed: 12/12/2022]
Abstract
The mechanisms underlying alcoholic liver disease are not fully understood. It has been established that alcohol interferes with transcriptional and translational regulatory steps of cell function. To understand such an effect, assessment of alcohol-induced changes in the simultaneous expression of a large number of genes may prove very useful. The purpose of the current study was to test a large number of genes ( approximately 8700) for possible changes in expression induced by alcohol alone or in addition to treatment with lipopolysaccharide (LPS), a putative mediator of alcohol effects on the liver. Male rats were fed an alcohol-containing liquid diet (Lieber-DeCarli) for 14-15 weeks, injected with Escherichia coli LPS (0.8 mg x kg(-1)), and killed 24 h later. Blood samples were taken for determination of plasma liver enzyme activity, and liver samples were obtained for histologic evaluation and total RNA extraction. Total RNA was analyzed for gene expression (Rat Toxicology U34 Array; Affymetrix, Santa Clara, CA). Of 8740 genes on the microchip, 2259 were expressed in the liver. Seven hundred ninety-eight genes underwent significant changes induced by either alcohol or LPS, but listed in this article are only those that significantly increased or decreased expression twofold or more. The genes were assigned to functional groups and reviewed. Gene changes were discussed from two viewpoints: relevance to established hypotheses of alcohol and LPS mechanisms of action and revealing of novel mechanisms of alcohol-induced liver injury. Application of DNA microarray technology to the study of alcohol-induced liver injury generated novel theoretical and experimental approaches to alcohol-induced liver injury.
Collapse
Affiliation(s)
- Ion V Deaciuc
- College of Medicine, Department of Internal Medicine, 800 Rose Street, MN649A-0298, University of Kentucky, Lexington, KY 40536, USA.
| | | | | | | | | | | | | |
Collapse
|
37
|
Deaciuc IV, Doherty DE, Burikhanov R, Lee EY, Stromberg AJ, Peng X, de Villiers WJS. Large-scale gene profiling of the liver in a mouse model of chronic, intragastric ethanol infusion. J Hepatol 2004; 40:219-27. [PMID: 14739091 DOI: 10.1016/j.jhep.2003.10.021] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND/AIMS The mechanisms underlying alcohol-induced liver injury are not fully elucidated. An approach in this direction would consist of an all-inclusive assessment of gene expression in the liver. The purpose of this study was to perform a comprehensive analysis of gene expression in the livers of mice treated with ethanol by means of intragastric infusion. METHODS An ethanol- or glucose-enriched liquid diet was fed to animals for 4 weeks via a long-term gastrostomy catheter. The animals were killed and plasma alanine:2-oxoglutarate aminotransferase (ALT) assay, liver histology and total RNA analysis by microarray gene technology were performed. RESULTS Alcohol increased ALT, induced steatosis, necrosis and inflammation. A total of 12,423 genes were analyzed for expression out of which 4867 were expressed by the liver. Alcohol repressed expression of 11 genes, induced expression of 13 genes, and up- or down-regulated expression of 44 and 42 genes >2-fold, respectively. Gene expression analysis identified several genes that have not previously been tested for alcohol effects. CONCLUSIONS This study: (i) expands the knowledge of mechanism(s) of action of ethanol; (ii) indicates novel pathways of ethanol action on the liver, and (iii) illustrates the utility of microarray gene analysis in hepatology research.
Collapse
Affiliation(s)
- Ion V Deaciuc
- Division of Gastroentrology/Hepatology, Department of Medicine, University of Louisville School of Medicine, Louisville, KY 40292, USA.
| | | | | | | | | | | | | |
Collapse
|
38
|
Yao L, Fan P, Jiang Z, Mailliard WS, Gordon AS, Diamond I. Addicting drugs utilize a synergistic molecular mechanism in common requiring adenosine and Gi-beta gamma dimers. Proc Natl Acad Sci U S A 2003; 100:14379-84. [PMID: 14605213 PMCID: PMC283600 DOI: 10.1073/pnas.2336093100] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2003] [Indexed: 11/18/2022] Open
Abstract
The mesolimbic dopamine system and cAMP-dependent/protein kinase A (PKA) pathways are strongly implicated in addictive behaviors. Here we determine the role of dopamine D2 receptors (D2) in PKA signaling responses to delta-opioid (DOR) and cannabinoid (CB1) receptors. We find in NG108-15/D2 cells and in cultured primary neurons that a brief exposure to saturating concentrations of DOR and CB1 agonists increases cAMP, promotes PKA C alpha translocation and increases cAMP-dependent gene expression. Activation of PKA signaling is mediated by Gi-beta gamma dimers. Importantly, subthreshold concentrations of DOR or CB1 agonists with D2 agonists, which are without effect when added separately, together activate cAMP/PKA signaling synergistically. There is also synergy between DOR or CB1 with ethanol, another addicting agent. In all instances, synergy requires adenosine activation of adenosine A2 receptors and is mediated by beta gamma dimers. Synergy by this molecular mechanism appears to confer hypersensitivity to opioids and cannabinoids while simultaneously increasing the sensitivity of D2 signaling when receptors are expressed on the same cells. This mechanism may account, in part, for drug-induced activation of medium spiny neurons in the nucleus accumbens.
Collapse
MESH Headings
- Adenosine/metabolism
- Animals
- Arachidonic Acids/pharmacology
- Cell Line
- Cells, Cultured
- Cyclic AMP/metabolism
- Cyclic AMP-Dependent Protein Kinase Catalytic Subunits
- Cyclic AMP-Dependent Protein Kinases/metabolism
- Dimerization
- Dopamine Agonists/pharmacology
- Drug Synergism
- Enkephalin, Leucine-2-Alanine/pharmacology
- Ethanol/pharmacology
- GTP-Binding Protein beta Subunits/chemistry
- GTP-Binding Protein beta Subunits/metabolism
- Isoenzymes/metabolism
- Models, Neurological
- Neurons/drug effects
- Neurons/metabolism
- Rats
- Receptor, Cannabinoid, CB1/agonists
- Receptor, Cannabinoid, CB1/metabolism
- Receptors, Adenosine A2/metabolism
- Receptors, Opioid, delta/agonists
- Receptors, Opioid, delta/metabolism
- Substance-Related Disorders/metabolism
Collapse
Affiliation(s)
- Lina Yao
- Ernest Gallo Clinic and Research Center, 5858 Horton Street, Suite 200, Emeryville, CA 94608, USA.
| | | | | | | | | | | |
Collapse
|
39
|
Hassan S, Duong B, Kim KS, Miles MF. Pharmacogenomic analysis of mechanisms mediating ethanol regulation of dopamine beta-hydroxylase. J Biol Chem 2003; 278:38860-9. [PMID: 12842874 DOI: 10.1074/jbc.m305040200] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
We previously showed that ethanol regulates dopamine beta-hydroxylase (DBH) mRNA and protein levels in human neuroblastoma cells (Thibault, C., Lai, C., Wilke, N., Duong, B., Olive, M. F., Rahman, S., Dong, H., Hodge, C. W., Lockhart, D. J., and Miles, M. F. (2000) Mol. Pharmacol. 58, 1593-1600). DBH catalyzes norepinephrine synthesis, and several studies have suggested a role for norepinephrine in ethanol-mediated behaviors. Here, we performed a detailed analysis of mechanism(s) underlying ethanol regulation of DBH expression in SH-SY5Y cells. Transient transfection analysis showed that ethanol (25-200 mM) caused concentration- and time-dependent increases in DBH gene transcription. Progressive deletions identified ethanol-responsive sequences in the -262 to -142 bp region of the DBH gene promoter. Mutagenesis of cAMP-response element (CRE) sequences in this region abolished ethanol responsiveness while maintaining responsiveness to phorbol esters. Coexpression of dominant-negative CRE-binding protein greatly reduced ethanol induction of DBH. Inhibitors of protein kinase A, casein kinase II, and MAPK reduced ethanol induction of DBH promoter activity. Pharmacogenomic studies with microarrays showed that protein kinase A, MEK, and casein kinase II inhibitors blocked induction of DBH and a large subset of ethanol-responsive genes. These genes had diverse functional groupings, including multiple members of the MAPK and phosphatidylinositol signaling cascades. Real-time PCR analysis validated select microarray results. Taken together, these results suggest that ethanol regulation of DBH requires a functional CRE and its binding protein and may require interaction of multiple kinase pathways. This mechanism may also mediate ethanol responsiveness of a complex subset of genes in neural cells. These studies may have implications for behavioral responses to ethanol or mechanisms underlying ethanol-related neurological disease.
Collapse
Affiliation(s)
- Sajida Hassan
- Departments of Pharmacology Toxicology and Neurology and the Center for Study of Biological Complexity, Virginia Commonwealth University, Richmond, Virginia 23298, USA
| | | | | | | |
Collapse
|
40
|
Abstract
This article provides a detailed assessment of the toxicological and pharmacological literature concerning alcohol-induced biphasic dose-response relationships. The assessment reveals that alcohol-induced hormetic-like dose-response relationships are commonly observed, highly generalizeable according to model and endpoint and quantitative feature of the dose response. These findings have important implications affecting study design, animal model, and endpoint selection as well as clinical applications.
Collapse
Affiliation(s)
- Edward J Calabrese
- Department of Environmental Health Sciences, Morrill Science Center, University of Massachusetts, Amherst, MA 01003, USA.
| | | |
Collapse
|
41
|
Liu X, Zha J, Nishitani J, Chen H, Zack JA. HIV-1 infection in peripheral blood lymphocytes (PBLs) exposed to alcohol. Virology 2003; 307:37-44. [PMID: 12667812 DOI: 10.1016/s0042-6822(02)00031-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Epidemiological and in vitro studies have implied that heavy alcohol consumption may increase an individual's risk of HIV-1 infection. To examine the role of alcohol in direct infection of T-cells, viral reverse transcripts and HIV-1 receptor expression were examined in infected peripheral blood lymphocytes (PBLs) pretreated with alcohol. PCR results showed that alcohol increased HIV-1 DNA in PBLs by at least 10-fold. Alcohol enhanced the expression of the CXCR4 chemokine co-receptor but not the major HIV-1 CD4 receptor. Pretreatment with alcohol was also associated with increased intracellular cAMP. Thus, alcohol may facilitate enhanced viral infection by increasing the availability of HIV-1 co-receptor. This effect is associated with increases in intracellular cAMP.
Collapse
Affiliation(s)
- Xuan Liu
- Department of Oral & Maxillofacial Surgery, Charles R. Drew University of Medicine & Science, Hawkins Building, Room 3067, 1731 East 120th Street, Los Angeles, CA 90059, USA.
| | | | | | | | | |
Collapse
|
42
|
Yao L, Arolfo MP, Dohrman DP, Jiang Z, Fan P, Fuchs S, Janak PH, Gordon AS, Diamond I. betagamma Dimers mediate synergy of dopamine D2 and adenosine A2 receptor-stimulated PKA signaling and regulate ethanol consumption. Cell 2002; 109:733-43. [PMID: 12086672 DOI: 10.1016/s0092-8674(02)00763-8] [Citation(s) in RCA: 111] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Dopamine release is activated by ethanol and addicting drugs, but molecular mechanisms linking dopaminergic signaling to neuronal responses and drinking behavior are poorly understood. We report that dopamine-D2 receptors induce PKA Calpha translocation and increase CRE-regulated gene expression. Ethanol also activates PKA signaling. Subthreshold concentrations of the D2 agonist NPA and ethanol, without effect alone, together cause synergistic PKA translocation and CRE-mediated gene transcription. D2 or adenosine A2 receptor blockade, pertussis toxin, Rp-cAMPS, or overexpression of dominant-negative peptides that sequester betagamma dimers prevent synergy. Importantly, overexpression of a betagamma inhibitor peptide in the nucleus accumbens strikingly reduces sustained alcohol consumption. We propose that synergy of D2 and A2 confers ethanol hypersensitivity and that betagamma dimers are required for voluntary drinking.
Collapse
MESH Headings
- Adenoviridae/genetics
- Alcohol Drinking
- Animals
- Animals, Newborn
- Apomorphine/analogs & derivatives
- Apomorphine/pharmacology
- Blotting, Western
- Cyclic AMP/metabolism
- Cyclic AMP-Dependent Protein Kinases/metabolism
- Dimerization
- Dopamine Agonists/pharmacology
- Dose-Response Relationship, Drug
- Ethanol/pharmacology
- G1 Phase
- Gene Expression Regulation
- Genes, Reporter
- Hippocampus/cytology
- Immunohistochemistry
- Integrases/metabolism
- Isoenzymes/metabolism
- Luciferases/metabolism
- Microscopy, Confocal
- Models, Biological
- Peptides/chemistry
- Pertussis Toxin
- Protein Binding
- Protein Kinase C/metabolism
- Protein Kinase C-alpha
- Protein Structure, Tertiary
- Protein Transport
- Rats
- Rats, Sprague-Dawley
- Receptors, Dopamine D2/chemistry
- Receptors, Dopamine D2/metabolism
- Receptors, Purinergic P1/chemistry
- Receptors, Purinergic P1/metabolism
- Signal Transduction
- Subcellular Fractions
- Time Factors
- Transcription, Genetic
- Transfection
- Viral Proteins/metabolism
- Virulence Factors, Bordetella/pharmacology
Collapse
Affiliation(s)
- Lina Yao
- Ernest Gallo Clinic and Research Center, San Francisco, CA 94110, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Constantinescu A, Gordon AS, Diamond I. cAMP-dependent protein kinase types I and II differentially regulate cAMP response element-mediated gene expression: implications for neuronal responses to ethanol. J Biol Chem 2002; 277:18810-6. [PMID: 11886856 DOI: 10.1074/jbc.m112107200] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We have shown that ethanol induces translocation of cAMP-dependent protein kinase (PKA) to the nucleus, cAMP response element-binding protein (CREB) phosphorylation, and cAMP response element-mediated gene transcription in NG108-15 cells. However, little is known about which PKA types regulate this process. We show here that under basal conditions NG108-15 cells contain type I PKA (CbetaRIbeta) primarily in cytosol and type II PKA (CalphaRIIbeta) in the particulate and nuclear fractions. Antagonists of both type I and type II PKA inhibit forskolin- and ethanol-induced cAMP response element-mediated gene transcription. However, only the type II PKA antagonist inhibits forskolin-induced Calpha and ethanol-induced Calpha and RIIbeta translocation to the nucleus and CREB phosphorylation; the type I antagonist is without effect. Our data suggest that forskolin- and ethanol-induced CREB phosphorylation and gene activation are differentially mediated by the two types of PKA. We propose that type II PKA is translocated and activated in the nucleus and induces CREB phosphorylation that is necessary but not sufficient for gene transcription. By contrast, type I PKA is activated in the cytoplasm, turning on a downstream pathway that activates other transcription cofactors that interact with phosphorylated CREB to induce gene transcription.
Collapse
Affiliation(s)
- Anastasia Constantinescu
- Ernest Gallo Clinic and Research Center, Department of Neurology, University of California at San Francisco, Emeryville, California 94608, USA.
| | | | | |
Collapse
|
44
|
Asher O, Cunningham TD, Yao L, Gordon AS, Diamond I. Ethanol stimulates cAMP-responsive element (CRE)-mediated transcription via CRE-binding protein and cAMP-dependent protein kinase. J Pharmacol Exp Ther 2002; 301:66-70. [PMID: 11907158 DOI: 10.1124/jpet.301.1.66] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Alcoholism is characterized by tolerance, dependence, and unrestrained craving for alcohol. Adaptive responses, including changes in gene expression in neurons, are thought to account for some of these complex behavioral abnormalities. We have shown in the NG108-15 neuroblastoma x glioma hybrid cell line that ethanol increases cellular cAMP levels via activation of adenosine A(2) receptors, leading to phosphorylation of the cAMP response element-binding protein (CREB). However, phosphorylation of CREB is not sufficient to activate cAMP response element (CRE)-mediated gene expression. Here we investigate whether ethanol increases CRE-mediated gene expression via endogenous CREB using a CRE-regulated luciferase reporter construct, transfected into NG108-15 cells. We find increased luciferase activity as a function of time of exposure to ethanol. Coexpression of a dominant-negative CREB construct blocked ethanol-stimulated CRE-luciferase expression, further suggesting that CREB is required for this response. We also determined whether ethanol-induced increases in gene expression are mediated by ethanol-induced increases in extracellular adenosine. We found that CRE-mediated gene expression induced by ethanol occurs in two phases: an early phase (4 h), in which adenosine receptor blockade prevents ethanol-induced gene expression, and a later phase (14 h), which is not blocked by an adenosine receptor antagonist. In both phases, inhibition of cAMP-dependent protein kinase A (PKA) activity prevented ethanol-induced CRE-mediated luciferase expression. Our data suggest that ethanol induces cAMP-dependent gene expression regulated by CREB and PKA and that this signaling pathway may mediate some of the addictive behaviors underlying alcoholism.
Collapse
Affiliation(s)
- Orna Asher
- Ernest Gallo Clinic and Research Center and Department of Neurology, University of California, San Francisco, Emeryville, California 94608, USA
| | | | | | | | | |
Collapse
|
45
|
Ethanol-Induced Translocation of Protein Kinase A Occurs in Two Phases: Control by Different Molecular Mechanisms. Alcohol Clin Exp Res 2002. [DOI: 10.1097/00000374-200203000-00016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
46
|
Dohrman DP, Chen HM, Gordon AS, Diamond I. Ethanol-Induced Translocation of Protein Kinase A Occurs in Two Phases: Control by Different Molecular Mechanisms. Alcohol Clin Exp Res 2002. [DOI: 10.1111/j.1530-0277.2002.tb02553.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
47
|
Basavarajappa BS, Hungund BL. Neuromodulatory role of the endocannabinoid signaling system in alcoholism: an overview. Prostaglandins Leukot Essent Fatty Acids 2002; 66:287-99. [PMID: 12052043 DOI: 10.1054/plef.2001.0352] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The current review evaluates the evidence that some of the pharmacological and behavioral effects of ethanol (EtOH), including EtOH-preferring behavior, may be mediated through the endocannabinoid signaling system. The recent advances in the understanding of the neurobiological basis of alcoholism suggest that the pharmacological and behavioral effects of EtOH are mediated through its action on neuronal signal transduction pathways and ligand-gated ion channels, receptor systems, and receptors that are coupled to G-proteins. The identification of a G-protein-coupled receptor, namely, the cannabinoid receptor (CB1 receptor) that was activated by Delta(9)-tetrahydrocannabinol (Delta(9)-THC), the major psychoactive component of marijuana, led to the discovery of endogenous cannabinoid agonists. To date, two fatty acid derivatives identified to be arachidonylethanolamide (AEA) and 2-arachidonylglycerol (2-AG) have been isolated from both nervous and peripheral tissues. Both these compounds have been shown to mimic the pharmacological and behavioral effects of Delta(9)-THC. The involvement of the endocannabinoid signaling system in the development of tolerance to the drugs of abuse including EtOH has not been known until recently. Recent studies from our laboratory have demonstrated for the first time the down-regulation of CB1 receptor function and its signal transduction by chronic EtOH. The observed down-regulation of CB1 receptor binding and its signal transduction results from the persistent stimulation of the receptors by the endogenous CB1 receptor agonists, AEA and 2-AG, the synthesis of which has been found to be increased by chronic EtOH treatment. This enhanced formation of endocannabinoids may subsequently influence the release of neurotransmitters. It was found that the DBA/2 mice, known to avoid EtOH intake, have significantly reduced brain-CB1-receptor function consistent with other studies, where the CB1 receptor antagonist SR141716A has been shown to block voluntary EtOH intake in rodents. Similarly, activation of the CB1 receptor system promoted alcohol craving, suggesting a role for the CB1 receptor gene in excessive EtOH drinking behavior and development of alcoholism. Ongoing investigations may lead to the development of potential therapeutic strategies for the treatment of alcoholism.
Collapse
Affiliation(s)
- B S Basavarajappa
- Division of Analytical Psychopharmacology, New York State Psychiatric Institute, New York, NY, USA.
| | | |
Collapse
|
48
|
Basavarajappa BS, Hungund BL. Cannabinoid receptor agonist-stimulated [35S]guanosine triphosphate gammaS binding in the brain of C57BL/6 and DBA/2 mice. J Neurosci Res 2001; 64:429-36. [PMID: 11340650 DOI: 10.1002/jnr.1094] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The two inbred strains of mice C57BL/6 (alcohol-preferring) and DBA/2 (alcohol-avoiding) mice have been shown to differ significantly in their preference for alcohol (EtOH). We have previously demonstrated the differences in the density and the affinity of cannabinoid (CB1) receptors in the brains of the two inbred C57BL/6 and DBA/2 mouse strains. In the present study, we investigated the CB1 receptor agonist-stimulated guanosine-5'-O-(3-[(35)S]thio)-triphosphate ([(35)S]GTPgammaS) binding in plasma membranes (PM) from C57BL/6 and DBA/2 mice. The results indicate that the net CP55,940-stimulated [(35)S]GTPgammaS binding was increased with increasing concentrations of CB1 receptor agonists and GDP. The net CB1 receptor agonist (WIN55,212-2 or HU-210 or CP55,940)-stimulated [(35)S]GTPgammaS binding was reduced significantly (-10% to -12%, P < 0.05) in PM from DBA/2 mice; no significant differences were observed in basal [(35)S]GTPgammaS binding among these strains. Nonlinear regression analysis of net CP55,940-stimulated [(35)S]GTPgammaS binding showed that the B(max) of cannabinoid agonist-stimulated binding was significantly reduced (-24%) in DBA/2 mice (B(max) = 12.43 +/- 0.64 for C57BL/6 and 9.46 +/- 0.98 pmol/mg protein for DBA/2; P < 0.05) without any significant changes in the G protein affinity. The pharmacological specificity of CP55,940-stimulated [(35)S]GTPgammaS binding was examined with CB1 receptor antagonist SR141716A, and these studies indicated that CP55,940-stimulated [(35)S]GTPgammaS binding was blocked by SR141716A, with a decrease in the IC(50) values in the PM from the DBA/2 mouse strain. These results suggest that a signal transduction pathway(s) downstream from the CB1 receptor system may play an important role in controlling the voluntary EtOH consumption by these strains of mice.
Collapse
Affiliation(s)
- B S Basavarajappa
- New York State Psychiatric Institute at NKI, Orangeburg, New York 10962, USA
| | | |
Collapse
|
49
|
Lewohl JM, Dodd PR, Mayfield RD, Harris RA. Application of DNA microarrays to study human alcoholism. J Biomed Sci 2001; 8:28-36. [PMID: 11173973 DOI: 10.1007/bf02255968] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
An emerging idea is that long-term alcohol abuse results in changes in gene expression in the brain and that these changes are responsible at least partly for alcohol tolerance, dependence and neurotoxicity. The overall goal of our research is to identify genes which are differentially expressed in the brains of well-characterized human alcoholics as compared with non-alcoholics. This should identify as-yet-unknown alcohol-responsive genes, and may well confirm changes in the expression of genes which have been delineated in animal models of alcohol abuse. Cases were carefully selected and samples pooled on the basis of relevant criteria; differential expression was monitored by microarray hybridization. The inherent diversity of human alcoholics can be exploited to identify genes associated with specific pathological processes, as well as to assess the effects of concomitant disease, severity of brain damage, drinking behavior, and factors such as gender and smoking history. Initial results show selective changes in gene expression in alcoholics; of particular importance is a coordinated reduction in genes coding for myelin components.
Collapse
Affiliation(s)
- J M Lewohl
- Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, TX 78712, USA
| | | | | | | |
Collapse
|
50
|
Greer F, Friars D, Graham TE. Comparison of caffeine and theophylline ingestion: exercise metabolism and endurance. J Appl Physiol (1985) 2000; 89:1837-44. [PMID: 11053334 DOI: 10.1152/jappl.2000.89.5.1837] [Citation(s) in RCA: 97] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
This two-part investigation compared the ergogenic and metabolic effects of theophylline and caffeine. Initially (part A), the ergogenic potential of theophylline on endurance exercise was investigated. Eight men cycled at 80% maximum O(2) consumption to exhaustion 90 min after ingesting either placebo (dextrose), caffeine (6 mg/kg; Caff), or theophylline (4.5 mg/kg Theolair; Theo). There was a significant increase in time to exhaustion in both the Caff (41.2+/-4.8 min) and Theo (37.4+/-5.0 min) trials compared with placebo (32.6+/-3.4 min) (P<0.05). In part B, the effects of Theo on muscle metabolism were investigated and compared with Caff. Seven men cycled for 45 min at 70% maximum O(2) consumption (identical treatment protocol as in part A). Neither methylxanthines (MX) affected muscle glycogen utilization (P>0.05). Only Caff increased plasma epinephrine (P<0.05), but both MX increased blood glycerol levels (P<0.05). Muscle cAMP was increased (P<0.05) by both MX at 15 min and remained elevated at 45 min with Theo. This demonstrates that both MX are ergogenic and that this can be independent of muscle glycogen.
Collapse
Affiliation(s)
- F Greer
- Department of Human Biology and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada N1G 2W1
| | | | | |
Collapse
|