1
|
A L, Siddiqi MT, P M, Fp V. Neuroimmune regulation of the prefrontal cortex tetrapartite synapse. Neuropharmacology 2025:110335. [PMID: 39904409 DOI: 10.1016/j.neuropharm.2025.110335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 01/20/2025] [Accepted: 01/27/2025] [Indexed: 02/06/2025]
Abstract
The prefrontal cortex (PFC) is an essential driver of cognitive, affective, and motivational behavior. There is clear evidence that the neuroimmune system directly influences PFC synapses, in addition to its role as the first line of defense against toxins and pathogens. In this review, we first describe the core structures that form the tetrapartite PFC synapse, focusing on the signaling microdomain created by astrocytic cradling of the synapse as well as the emerging role of the extracellular matrix in synaptic organization and plasticity. Neuroimmune signals (e.g. pro-inflammatory interleukin 1β) can impact the function of each core structure within the tetrapartite synapse, as well as promote intra-synaptic crosstalk, and we will provide an overview of recent advances in this field. Finally, evidence from post mortem human brain tissue and preclinical studies indicate that inflammation may be a key contributor to PFC dysfunction. Therefore, we conclude with a mechanistic discussion of neuroimmune-mediated maladaptive plasticity in neuropsychiatric disorders, with a focus on alcohol use disorder (AUD). Growing recognition of the neuroimmune system's role as a critical regulator of the PFC tetrapartite synapse provides strong support for targeting the neuroimmune system to develop new pharmacotherapeutics.
Collapse
Affiliation(s)
- Liss A
- Developmental Exposure Alcohol Research Center and Behavioral Neuroscience Program, Department of Psychology, Binghamton University-SUNY, Binghamton, NY
| | - M T Siddiqi
- Developmental Exposure Alcohol Research Center and Behavioral Neuroscience Program, Department of Psychology, Binghamton University-SUNY, Binghamton, NY
| | - Marsland P
- Developmental Exposure Alcohol Research Center and Behavioral Neuroscience Program, Department of Psychology, Binghamton University-SUNY, Binghamton, NY
| | - Varodayan Fp
- Developmental Exposure Alcohol Research Center and Behavioral Neuroscience Program, Department of Psychology, Binghamton University-SUNY, Binghamton, NY.
| |
Collapse
|
2
|
Lapish CC. Understanding How Acute Alcohol Impacts Neural Encoding in the Rodent Brain. Curr Top Behav Neurosci 2024. [PMID: 38858298 DOI: 10.1007/7854_2024_479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2024]
Abstract
Alcohol impacts neural circuitry throughout the brain and has wide-ranging effects on the biophysical properties of neurons in these circuits. Articulating how these wide-ranging effects might eventually result in altered computational properties has the potential to provide a tractable working model of how alcohol alters neural encoding. This chapter reviews what is currently known about how acute alcohol influences neural activity in cortical, hippocampal, and dopaminergic circuits as these have been the primary focus of understanding how alcohol alters neural computation. While other neural systems have been the focus of exhaustive work on this topic, these brain regions are the ones where in vivo neural recordings are available, thus optimally suited to make the link between changes in neural activity and behavior. Rodent models have been key in developing an understanding of how alcohol impacts the function of these circuits, and this chapter therefore focuses on work from mice and rats. While progress has been made, it is critical to understand the challenges and caveats associated with experimental procedures, especially when performed in vivo, which are designed to answer this question and if/how to translate these data to humans. The hypothesis is discussed that alcohol impairs the ability of neural circuits to acquire states of neural activity that are transiently elevated and characterized by increased complexity. It is hypothesized that these changes are distinct from the traditional view of alcohol being a depressant of neural activity in the forebrain.
Collapse
Affiliation(s)
- Christopher C Lapish
- Department of Anatomy, Cell Biology, and Physiology, Stark Neuroscience Institute, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
3
|
Raffone N, Chistiakova M, Volgushev M. Ethanol Differentially Affects Excitatory and Inhibitory Synaptic Transmission in Visual Cortex of Wild-type and Adenosine A 1R Knock-out Mice. Neuroscience 2024; 540:117-127. [PMID: 38278472 DOI: 10.1016/j.neuroscience.2024.01.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 01/18/2024] [Accepted: 01/22/2024] [Indexed: 01/28/2024]
Abstract
Ethanol is one of the most commonly used and abused substances in the world. While the behavioral effects of ethanol are well characterized, mechanisms of its action on neurons and synapses remain elusive. Prior research suggested that ethanol could affect neurons by interfering with metabolism of biologically active molecules, such as adenosine. Here, we explored the involvement of adenosine A1 receptors (A1R) in mediating ethanol's effects on synaptic transmission to layer 2/3 pyramidal neurons of visual cortex using wild type (WT) and A1R knock-out (KO) mice. Ethanol differentially affected excitatory and inhibitory transmission in WT and KO mice. In slices from WT mice ethanol had heterogeneous effects on excitatory transmission (facilitation, suppression or no change), with no net change. Ethanol's effects remained heterogeneous during acute blockade of A1Rs with a selective antagonist DPCPX. However, in A1RKO mice ethanol consistently suppressed excitatory transmission, with no cases of enhancement observed. Inhibitory transmission was suppressed by ethanol in both WT and A1RKO mice. At both excitatory and inhibitory synapses, changes of response amplitude correlated with changes of paired-pulse ratio, suggesting involvement of presynaptic mechanisms. We conclude that A1Rs are not involved in mediating effects of ethanol on synaptic transmission in mouse visual cortex. However, A1Rs are necessary for development of mechanisms mediating facilitation at some excitatory synapses. Our results add evidence for the diversity of ethanol's effects and mechanisms of action on synaptic transmission in different brain structures, and even in the same brain area (visual cortex) in different species, rats vs mice.
Collapse
Affiliation(s)
- Noah Raffone
- Department of Psychological Sciences, University of Connecticut, Storrs, CT 06269, USA.
| | - Marina Chistiakova
- Department of Psychological Sciences, University of Connecticut, Storrs, CT 06269, USA.
| | - Maxim Volgushev
- Department of Psychological Sciences, University of Connecticut, Storrs, CT 06269, USA; The Institute for the Brain and Cognitive Sciences, University of Connecticut, Storrs, CT 06269 USA.
| |
Collapse
|
4
|
Siddiqi MT, Podder D, Pahng AR, Athanason AC, Nadav T, Cates-Gatto C, Kreifeldt M, Contet C, Roberts AJ, Edwards S, Roberto M, Varodayan FP. Prefrontal cortex glutamatergic adaptations in a mouse model of alcohol use disorder. ADDICTION NEUROSCIENCE 2023; 9:100137. [PMID: 38152067 PMCID: PMC10752437 DOI: 10.1016/j.addicn.2023.100137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
Alcohol use disorder (AUD) produces cognitive deficits, indicating a shift in prefrontal cortex (PFC) function. PFC glutamate neurotransmission is mostly mediated by α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid-type ionotropic receptors (AMPARs); however preclinical studies have mostly focused on other receptor subtypes. Here we examined the impact of early withdrawal from chronic ethanol on AMPAR function in the mouse medial PFC (mPFC). Dependent male C57BL/6J mice were generated using the chronic intermittent ethanol vapor-two bottle choice (CIE-2BC) paradigm. Non-dependent mice had access to water and ethanol bottles but did not receive ethanol vapor. Naïve mice had no ethanol exposure. We used patch-clamp electrophysiology to measure glutamate neurotransmission in layer 2/3 prelimbic mPFC pyramidal neurons. Since AMPAR function can be impacted by subunit composition or plasticity-related proteins, we probed their mPFC expression levels. Dependent mice had higher spontaneous excitatory postsynaptic current (sEPSC) amplitude and kinetics compared to the Naïve/Non-dependent mice. These effects were seen during intoxication and after 3-8 days withdrawal, and were action potential-independent, suggesting direct enhancement of AMPAR function. Surprisingly, 3 days withdrawal decreased expression of genes encoding AMPAR subunits (Gria1/2) and synaptic plasticity proteins (Dlg4 and Grip1) in Dependent mice. Further analysis within the Dependent group revealed a negative correlation between Gria1 mRNA levels and ethanol intake. Collectively, these data establish a role for mPFC AMPAR adaptations in the glutamatergic dysfunction associated with ethanol dependence. Future studies on the underlying AMPAR plasticity mechanisms that promote alcohol reinforcement, seeking, drinking and relapse behavior may help identify new targets for AUD treatment.
Collapse
Affiliation(s)
- Mahum T. Siddiqi
- Developmental Exposure Alcohol Research Center and Behavioral Neuroscience Program, Department of Psychology, Binghamton University-SUNY, 4400 Vestal Parkway East, Binghamton, NY, 13902, USA
| | - Dhruba Podder
- Developmental Exposure Alcohol Research Center and Behavioral Neuroscience Program, Department of Psychology, Binghamton University-SUNY, 4400 Vestal Parkway East, Binghamton, NY, 13902, USA
| | - Amanda R. Pahng
- Department of Physiology, Louisiana State University Health Sciences Center, 533 Bolivar Street, New Orleans, LA, 70112, USA
- Southeast Louisiana Veterans Health Care System, 2400 Canal Street, 11F, New Orleans, LA, 70119, USA
| | - Alexandria C. Athanason
- Developmental Exposure Alcohol Research Center and Behavioral Neuroscience Program, Department of Psychology, Binghamton University-SUNY, 4400 Vestal Parkway East, Binghamton, NY, 13902, USA
| | - Tali Nadav
- Animal Models Core Facility, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Chelsea Cates-Gatto
- Animal Models Core Facility, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Max Kreifeldt
- Department of Molecular Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Candice Contet
- Department of Molecular Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Amanda J. Roberts
- Animal Models Core Facility, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Scott Edwards
- Department of Physiology, Louisiana State University Health Sciences Center, 533 Bolivar Street, New Orleans, LA, 70112, USA
| | - Marisa Roberto
- Department of Molecular Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Florence P. Varodayan
- Developmental Exposure Alcohol Research Center and Behavioral Neuroscience Program, Department of Psychology, Binghamton University-SUNY, 4400 Vestal Parkway East, Binghamton, NY, 13902, USA
- Department of Molecular Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, 92037, USA
| |
Collapse
|
5
|
Honarvar F, Arfaie S, Edalati H, Ghasroddashti A, Solgi A, Mashayekhi MS, Mofatteh M, Ren LY, Kwan ATH, Keramatian K. Neuroanatomical predictors of problematic alcohol consumption in adolescents: a systematic review of longitudinal studies. Alcohol Alcohol 2023; 58:455-471. [PMID: 37553844 DOI: 10.1093/alcalc/agad049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 06/30/2023] [Accepted: 07/04/2023] [Indexed: 08/10/2023] Open
Abstract
AIMS This study aimed to systematically review the literature on neuroanatomical predictors of future problematic drinking in adolescents. METHODS Using PRISMA guidelines, a systematic review was conducted to evaluate neuroanatomical predictors of problematic alcohol consumption in adolescents. EMBASE, MEDLINE, and PsycINFO databases were searched from inception to 6 January 2023. Studies were included if they were original, had a prospective design, had a sample size of at least 12, had a follow-up period of at least 1 year, had at least one structural neuroimaging scan before 18 with no prior alcohol use, and had alcohol use as the primary outcome. Studies were excluded if they had animals only and were not in English. Risk of bias was conducted using the CASP tool. RESULTS Out of 1412 studies identified, 19 studies met the criteria, consisting of 11 gray matter (n = 4040), 5 white matter (n = 319), and 3 assessing both (n = 3608). Neuroanatomical predictors of future problematic drinking in adolescents were reported to be distributed across various brain regions such as the orbitofrontal cortex and paralimbic regions. However, the findings were largely heterogeneous. CONCLUSIONS This is the first systematic review to map out the existing literature on neuroanatomical predictors of problematic drinking in adolescents. Future research should focus on the aforementioned regions to determine their role in predicting future problematic drinking with more certainty.
Collapse
Affiliation(s)
- Faraz Honarvar
- School of Medicine, Queen's University, 80 Barrie Street, Kingston, Ontario K7L 3N6, Canada
- Temerty Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada
| | - Saman Arfaie
- Department of Neurology and Neurosurgery, McGill University, Irving Ludmer Building1033 Pine Avenue West, room 310, Montréal, Québec H3A 1A1, Canada
- Faculty of Medicine, McGill University, 3605 Rue de la Montagne, Montréal, Québec H3G 2M1, Canada
- Department of Molecular and Cell Biology, University of California Berkeley, Weill Hall, #3200, Berkeley, CA 94720, United States
| | - Hanie Edalati
- Institut national de psychiatrie légale Philippe-Pinel, Université de Montréal, 10905 Boulevard Henri-Bourassa E, Montreal, Québec, H1C 1H1, Canada
- Centre for Research on Children and Families, McGill University, Room 155, Suite 100, Tour Est550 Sherbrooke Ouest, Montreal, Québec H3A 1B9, Canada
| | - Arashk Ghasroddashti
- School of Medicine, Queen's University, 80 Barrie Street, Kingston, Ontario K7L 3N6, Canada
| | - Arad Solgi
- School of Kinesiology & Health Science, York University, Norman Bethune CollegeRoom 3414700 Keele St, Toronto, Ontario M3J 1P3, Canada
| | | | - Mohammad Mofatteh
- School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, 97 Lisburn Rd, Belfast BT9 7BL, United Kingdom
| | - Lily Yuxi Ren
- Lane Medical Library and Knowledge Management Center, Stanford Medicine, Stanford University, 300 Pasteur Dr, Stanford, CA 94305, United States
| | - Angela T H Kwan
- Faculty of Medicine, University of Ottawa, Roger Guindon Hall, 451 Smyth Rd #2044, Ottawa, Ontario K1H 8M5, Canada
| | - Kamyar Keramatian
- Department of Psychiatry, University of British Columbia, 2255 Wesbrook Mall, Vancouver, British Columbia V6T 2A1, Canada
| |
Collapse
|
6
|
Jangjou A, Moqadas M, Mohsenian L, Kamyab H, Chelliapan S, Alshehery S, Ali MA, Dehbozorgi F, Yadav KK, Khorami M, Zarei Jelyani N. Awareness raising and dealing with methanol poisoning based on effective strategies. ENVIRONMENTAL RESEARCH 2023; 228:115886. [PMID: 37072082 DOI: 10.1016/j.envres.2023.115886] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 04/04/2023] [Accepted: 04/10/2023] [Indexed: 05/16/2023]
Abstract
Intoxication with methanol most commonly occurs as a consequence of ingesting, inhaling, or coming into contact with formulations that include methanol as a base. Clinical manifestations of methanol poisoning include suppression of the central nervous system, gastrointestinal symptoms, and decompensated metabolic acidosis, which is associated with impaired vision and either early or late blindness within 0.5-4 h after ingestion. After ingestion, methanol concentrations in the blood that are greater than 50 mg/dl should raise some concern. Ingested methanol is typically digested by alcohol dehydrogenase (ADH), and it is subsequently redistributed to the body's water to attain a volume distribution that is about equivalent to 0.77 L/kg. Moreover, it is removed from the body as its natural, unchanged parent molecules. Due to the fact that methanol poisoning is relatively uncommon but frequently involves a large number of victims at the same time, this type of incident occupies a special position in the field of clinical toxicology. The beginning of the COVID-19 pandemic has resulted in an increase in erroneous assumptions regarding the preventative capability of methanol in comparison to viral infection. More than 1000 Iranians fell ill, and more than 300 of them passed away in March of this year after they consumed methanol in the expectation that it would protect them from a new coronavirus. The Atlanta epidemic, which involved 323 individuals and resulted in the deaths of 41, is one example of mass poisoning. Another example is the Kristiansand outbreak, which involved 70 people and resulted in the deaths of three. In 2003, the AAPCC received reports of more than one thousand pediatric exposures. Since methanol poisoning is associated with high mortality rates, it is vital that the condition be addressed seriously and managed as quickly as feasible. The objective of this review was to raise awareness about the mechanism and metabolism of methanol toxicity, the introduction of therapeutic interventions such as gastrointestinal decontamination and methanol metabolism inhibition, the correction of metabolic disturbances, and the establishment of novel diagnostic/screening nanoparticle-based strategies for methanol poisoning such as the discovery of ADH inhibitors as well as the detection of the adulteration of alcoholic drinks by nanoparticles in order to prevent methanol poisoning. In conclusion, increasing warnings and knowledge about clinical manifestations, medical interventions, and novel strategies for methanol poisoning probably results in a decrease in the death load.
Collapse
Affiliation(s)
- Ali Jangjou
- Department of Emergency Medicine, School of Medicine, Namazi Teaching Hospital, Shiraz University of Medical Sciences, Shiraz, Iran; Emergency Medicine Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mostafa Moqadas
- Department of Emergency Medicine, School of Medicine, Namazi Teaching Hospital, Shiraz University of Medical Sciences, Shiraz, Iran; Emergency Medicine Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Leila Mohsenian
- Department of Emergency Medicine, School of Medicine, Namazi Teaching Hospital, Shiraz University of Medical Sciences, Shiraz, Iran; Emergency Medicine Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hesam Kamyab
- Faculty of Architecture and Urbanism, UTE University, Calle Rumipamba S/N and Bourgeois, Quito, Ecuador; Department of Biomaterials, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai, 600 077, India; Process Systems Engineering Centre (PROSPECT), Faculty of Chemical and Energy Engineering, Faculty of Engineering, Universiti Teknologi Malaysia, Skudai, Johor, Malaysia.
| | - Shreeshivadasan Chelliapan
- Engineering Department, Razak Faculty of Technology and Informatics, Universiti Teknologi Malaysia, Jln Sultan Yahya Petra, 54100, Kuala Lumpur, Malaysia.
| | - Sultan Alshehery
- Department of Mechanical Engineering King Khalid University, zip code - 62217, Saudi Arabia
| | - Mohammed Azam Ali
- Department of Mechanical Engineering King Khalid University, zip code - 62217, Saudi Arabia
| | - Farbod Dehbozorgi
- Department of Emergency Medicine, School of Medicine, Namazi Teaching Hospital, Shiraz University of Medical Sciences, Shiraz, Iran; Emergency Medicine Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Krishna Kumar Yadav
- Faculty of Science and Technology, Madhyanchal Professional University, Ratibad, Bhopal, 462044, India; Environmental and Atmospheric Sciences Research Group, Scientific Research Center, Al-Ayen University, Thi-Qar, Nasiriyah, 64001, Iraq
| | - Masoud Khorami
- Department of Civil Engineering, Islamic Azad University, Central Tehran Branch, Tehran, Iran
| | - Najmeh Zarei Jelyani
- Department of Emergency Medicine, School of Medicine, Namazi Teaching Hospital, Shiraz University of Medical Sciences, Shiraz, Iran; Emergency Medicine Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
7
|
Foster TC. Animal models for studies of alcohol effects on the trajectory of age-related cognitive decline. Alcohol 2023; 107:4-11. [PMID: 35504438 DOI: 10.1016/j.alcohol.2022.04.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/18/2022] [Accepted: 04/19/2022] [Indexed: 02/04/2023]
Abstract
There is growing interest in understanding how ethanol use interacts with advancing age to influence the brain and cognition. Animal models are employed to investigate the cellular and molecular mechanisms of brain aging and age-related neurodegenerative diseases that underlie cognitive decline. However, all too often research on problems and diseases of the elderly are conducted in healthy young animals, providing little clinical relevance. The validity of animal models is discussed, and confounds due to age-related differences in anxiety, sensory-motor function, and procedural learning are highlighted in order to enhance the successful translation of preclinical results into clinical settings. The mechanism of action of ethanol on brain aging will depend on the dose, acute or chronic treatment, or withdrawal from treatment and the age examined. Due to the fact that humans experience alcohol use throughout life, important questions concern the effects of the dose and duration of ethanol treatment on the trajectory of cognitive function. Central to this research will be questions of the specificity of alcohol effects on cognitive functions and related brain regions that decline with age, as well as the interaction of alcohol with mechanisms or biomarkers of brain aging. Alternatively, moderate alcohol use may provide a source of reserve and resilience against brain aging. Longitudinal studies have the advantage of being sensitive to detecting the effects of treatment on the emergence of cognitive impairment in middle age and can minimize effects of stress/anxiety associated with the novelty of alcohol exposure and behavioral testing, which disproportionately influence aged animals. Finally, the effect of alcohol on senescent neurophysiology and biomarkers of brain aging are discussed. In particular, the interaction of age and effects of alcohol on inflammation, oxidative stress, N-methyl-d-aspartate receptor function, and the balance of excitatory and inhibitory synaptic transmission are highlighted.
Collapse
Affiliation(s)
- Thomas C Foster
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, United States.
| |
Collapse
|
8
|
Gimenez-Gomez P, Le T, Martin GE. Modulation of neuronal excitability by binge alcohol drinking. Front Mol Neurosci 2023; 16:1098211. [PMID: 36866357 PMCID: PMC9971943 DOI: 10.3389/fnmol.2023.1098211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 01/19/2023] [Indexed: 02/16/2023] Open
Abstract
Drug use poses a serious threat to health systems throughout the world. The number of consumers rises every year being alcohol the drug of abuse most consumed causing 3 million deaths (5.3% of all deaths) worldwide and 132.6 million disability-adjusted life years. In this review, we present an up-to-date summary about what is known regarding the global impact of binge alcohol drinking on brains and how it affects the development of cognitive functions, as well as the various preclinical models used to probe its effects on the neurobiology of the brain. This will be followed by a detailed report on the state of our current knowledge of the molecular and cellular mechanisms underlying the effects of binge drinking on neuronal excitability and synaptic plasticity, with an emphasis on brain regions of the meso-cortico limbic neurocircuitry.
Collapse
Affiliation(s)
- Pablo Gimenez-Gomez
- Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, MA, United States
- The Brudnick Neuropsychiatric Research Institute, Worcester, MA, United States
| | - Timmy Le
- Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, MA, United States
- The Brudnick Neuropsychiatric Research Institute, Worcester, MA, United States
- Graduate Program in Neuroscience, Morningside Graduate School of Biomedical Sciences, UMass Chan Medical School, Worcester, MA, United States
| | - Gilles E. Martin
- Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, MA, United States
- The Brudnick Neuropsychiatric Research Institute, Worcester, MA, United States
| |
Collapse
|
9
|
Fish KN, Joffe ME. Targeting prefrontal cortex GABAergic microcircuits for the treatment of alcohol use disorder. Front Synaptic Neurosci 2022; 14:936911. [PMID: 36105666 PMCID: PMC9465392 DOI: 10.3389/fnsyn.2022.936911] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 07/27/2022] [Indexed: 11/17/2022] Open
Abstract
Developing novel treatments for alcohol use disorders (AUDs) is of paramount importance for improving patient outcomes and alleviating the suffering related to the disease. A better understanding of the molecular and neurocircuit mechanisms through which alcohol alters brain function will be instrumental in the rational development of new efficacious treatments. Clinical studies have consistently associated the prefrontal cortex (PFC) function with symptoms of AUDs. Population-level analyses have linked the PFC structure and function with heavy drinking and/or AUD diagnosis. Thus, targeting specific PFC cell types and neural circuits holds promise for the development of new treatments. Here, we overview the tremendous diversity in the form and function of inhibitory neuron subtypes within PFC and describe their therapeutic potential. We then summarize AUD population genetics studies, clinical neurophysiology findings, and translational neuroscience discoveries. This study collectively suggests that changes in fast transmission through PFC inhibitory microcircuits are a central component of the neurobiological effects of ethanol and the core symptoms of AUDs. Finally, we submit that there is a significant and timely need to examine sex as a biological variable and human postmortem brain tissue to maximize the efforts in translating findings to new clinical treatments.
Collapse
Affiliation(s)
| | - Max E. Joffe
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
10
|
Melugin PR, Wu F, Munoz C, Phensy A, Pradhan G, Luo Y, Nofal A, Manepalli R, Kroener S. The effects of acamprosate on prefrontal cortical function are mimicked by CaCl2 and they are influenced by the history of alcohol exposure. Neuropharmacology 2022; 212:109062. [PMID: 35430241 PMCID: PMC10804777 DOI: 10.1016/j.neuropharm.2022.109062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 03/29/2022] [Accepted: 04/11/2022] [Indexed: 11/27/2022]
Abstract
Alcohol use disorder is associated with functional changes in the medial prefrontal cortex (mPFC), which include altered glutamatergic transmission and deficits in executive functions that contribute to relapse. Acamprosate (calcium-bis N-acetylhomotaurinate) reduces alcohol craving and relapse, effects that are thought to be mediated by acamprosate's ability to ameliorate alcohol-induced dysregulation of glutamatergic signaling. Treatment with acamprosate and its active moiety calcium (CaCl2) both improve deficits in cognitive flexibility in postdependent mice following chronic intermittent ethanol (CIE) exposure. Here, we show that mice that self-administered alcohol under goal-directed conditions (i.e., operant responding on a fixed-ratio schedule) also display similar deficits in cognitive flexibility and altered glutamatergic signaling in the mPFC, both of which were improved with acamprosate or CaCl2. However, under conditions shown to bias behavior towards habitual responding (operant self-administration after CIE exposure, or on a variable interval schedule), alcohol-induced changes to glutamatergic transmission were unaffected by either acamprosate or CaCl2 treatment. Together, these findings suggest that the variable effects of acamprosate on synaptic signaling may reflect a shift in mPFC networks related to the loss of behavioral control in habitual alcohol-seeking.
Collapse
Affiliation(s)
- Patrick R Melugin
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, TX, USA
| | - Fei Wu
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, TX, USA; Institute of Neurobiology, Jining Medical University, Jining, China
| | - Crystal Munoz
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, TX, USA
| | - Aarron Phensy
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, TX, USA
| | - Grishma Pradhan
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, TX, USA
| | - Yi Luo
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, TX, USA
| | - Abraham Nofal
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, TX, USA
| | - Rohan Manepalli
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, TX, USA
| | - Sven Kroener
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, TX, USA.
| |
Collapse
|
11
|
Plasma Amino Acid Concentrations in Patients with Alcohol and/or Cocaine Use Disorders and Their Association with Psychiatric Comorbidity and Sex. Biomedicines 2022; 10:biomedicines10051137. [PMID: 35625874 PMCID: PMC9138967 DOI: 10.3390/biomedicines10051137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 05/08/2022] [Accepted: 05/13/2022] [Indexed: 02/05/2023] Open
Abstract
(1) Background: Co-occurrence of mental and substance use disorders (SUD) is prevalent, but complicates their clinical courses, and specific biomarkers are required. Amino acids are altered in primary mental disorders; however, little is known about SUD and psychiatric comorbidity. Because most psychiatric disorders and biomarkers show sex differences, we investigated amino acids in men and women with alcohol and/or cocaine use disorders (AUD and/or CUD) and psychiatric comorbidity. (2) Methods: A cross-sectional study was conducted in 295 participants, who were divided into four groups (AUD, n = 60; CUD, n = 41; AUD + CUD, n = 64; and control, n = 130). Participants were clinically assessed, and plasma amino acid concentrations were analyzed in relation to sex, diagnosis of SUD and psychiatric comorbidity (3) Results: In the total sample, there were sex differences, and women showed lower Iso, Leu, Gln and Glu than men. While patients with CUD and AUD + CUD had higher Glu, Gly, Orn and Ser than controls, patients with AUD showed no differences. In SUD, patients with psychiatric comorbidity had lower Orn and higher Ala than non-comorbid patients in the AUD group. (4) Conclusions: There was a dysregulation of plasma amino acids in abstinent patients with SUD. However, our results suggest the importance of considering the clinical characteristics and sex in the validity of amino acids as potential biomarkers for SUD.
Collapse
|
12
|
León BE, Kang S, Franca-Solomon G, Shang P, Choi DS. Alcohol-Induced Neuroinflammatory Response and Mitochondrial Dysfunction on Aging and Alzheimer's Disease. Front Behav Neurosci 2022; 15:778456. [PMID: 35221939 PMCID: PMC8866940 DOI: 10.3389/fnbeh.2021.778456] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 12/07/2021] [Indexed: 12/27/2022] Open
Abstract
Mitochondria are essential organelles central to various cellular functions such as energy production, metabolic pathways, signaling transduction, lipid biogenesis, and apoptosis. In the central nervous system, neurons depend on mitochondria for energy homeostasis to maintain optimal synaptic transmission and integrity. Deficiencies in mitochondrial function, including perturbations in energy homeostasis and mitochondrial dynamics, contribute to aging, and Alzheimer's disease. Chronic and heavy alcohol use is associated with accelerated brain aging, and increased risk for dementia, especially Alzheimer's disease. Furthermore, through neuroimmune responses, including pro-inflammatory cytokines, excessive alcohol use induces mitochondrial dysfunction. The direct and indirect alcohol-induced neuroimmune responses, including pro-inflammatory cytokines, are critical for the relationship between alcohol-induced mitochondrial dysfunction. In the brain, alcohol activates microglia and increases inflammatory mediators that can impair mitochondrial energy production, dynamics, and initiate cell death pathways. Also, alcohol-induced cytokines in the peripheral organs indirectly, but synergistically exacerbate alcohol's effects on brain function. This review will provide recent and advanced findings focusing on how alcohol alters the aging process and aggravates Alzheimer's disease with a focus on mitochondrial function. Finally, we will contextualize these findings to inform clinical and therapeutic approaches towards Alzheimer's disease.
Collapse
Affiliation(s)
- Brandon Emanuel León
- Regenerative Sciences Program, Center for Regenerative Medicine, Mayo Clinic, Rochester, MN, United States
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, United States
| | - Shinwoo Kang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, United States
| | - Gabriela Franca-Solomon
- Neuroscience Program, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
| | - Pei Shang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, United States
| | - Doo-Sup Choi
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, United States
- Neuroscience Program, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
- Department of Psychiatry and Psychology, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
| |
Collapse
|
13
|
Morningstar MD, Barnett WH, Goodlett CR, Kuznetsov A, Lapish CC. Understanding ethanol's acute effects on medial prefrontal cortex neural activity using state-space approaches. Neuropharmacology 2021; 198:108780. [PMID: 34480911 PMCID: PMC8488975 DOI: 10.1016/j.neuropharm.2021.108780] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 08/10/2021] [Accepted: 08/30/2021] [Indexed: 12/22/2022]
Abstract
Acute ethanol (EtOH) intoxication results in several maladaptive behaviors that may be attributable, in part, to the effects of EtOH on neural activity in medial prefrontal cortex (mPFC). The acute effects of EtOH on mPFC function have been largely described as inhibitory. However, translating these observations on function into a mechanism capable of delineating acute EtOH's effects on behavior has proven difficult. This review highlights the role of acute EtOH on electrophysiological measurements of mPFC function and proposes that interpreting these changes through the lens of dynamical systems theory is critical to understand the mechanisms that mediate the effects of EtOH intoxication on behavior. Specifically, the present review posits that the effects of EtOH on mPFC N-methyl-d-aspartate (NMDA) receptors are critical for the expression of impaired behavior following EtOH consumption. This hypothesis is based on the observation that recurrent activity in cortical networks is supported by NMDA receptors, and, when disrupted, may lead to impairments in cognitive function. To evaluate this hypothesis, we discuss the representation of mPFC neural activity in low-dimensional, dynamic state spaces. This approach has proven useful for identifying the underlying computations necessary for the production of behavior. Ultimately, we hypothesize that EtOH-related alterations to NMDA receptor function produces alterations that can be effectively conceptualized as impairments in attractor dynamics and provides insight into how acute EtOH disrupts forms of cognition that rely on mPFC function. This article is part of the special Issue on 'Neurocircuitry Modulating Drug and Alcohol Abuse'.
Collapse
Affiliation(s)
| | - William H Barnett
- Indiana University-Purdue University Indianapolis, Department of Psychology, USA
| | - Charles R Goodlett
- Indiana University-Purdue University Indianapolis, Department of Psychology, USA; Indiana University School of Medicine, Stark Neurosciences, USA
| | - Alexey Kuznetsov
- Indiana University-Purdue University Indianapolis, Department of Mathematics, USA; Indiana University School of Medicine, Stark Neurosciences, USA
| | - Christopher C Lapish
- Indiana University-Purdue University Indianapolis, Department of Psychology, USA; Indiana University School of Medicine, Stark Neurosciences, USA
| |
Collapse
|
14
|
Li M, Cabrera-Garcia D, Salling MC, Au E, Yang G, Harrison NL. Alcohol reduces the activity of somatostatin interneurons in the mouse prefrontal cortex: A neural basis for its disinhibitory effect? Neuropharmacology 2021; 188:108501. [PMID: 33636191 DOI: 10.1016/j.neuropharm.2021.108501] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 02/01/2021] [Accepted: 02/12/2021] [Indexed: 12/13/2022]
Abstract
The prefrontal cortex (PFC) is involved in executive ("top-down") control of behavior and its function is especially susceptible to the effects of alcohol, leading to behavioral disinhibition that is associated with alterations in decision making, response inhibition, social anxiety and working memory. The circuitry of the PFC involves a complex interplay between pyramidal neurons (PNs) and several subclasses of inhibitory interneurons (INs), including somatostatin (SST)-expressing INs. Using in vivo calcium imaging, we showed that alcohol dose-dependently altered network activity in layers 2/3 of the prelimbic subregion of the mouse PFC. Low doses of alcohol (1 g/kg, intraperitoneal, i.p.) caused moderate activation of SST INs and weak inhibition of PNs. At moderate to high doses, alcohol (2-3 g/kg) strongly inhibited the activity of SST INs in vivo, and this effect may result in disinhibition, as the activity of a subpopulation of PNs was simultaneously enhanced. In contrast, recordings in brain slices using ex vivo electrophysiology revealed no direct effect of alcohol on the excitability of either SST INs or PNs over a range of concentrations (20 and 50 mM) consistent with the blood alcohol levels reached in the in vivo experiments. This dose-dependent effect of alcohol on SST INs in vivo may reveal a neural basis for the disinhibitory effect of alcohol in the PFC mediated by other neurons within or external to the PFC circuitry.
Collapse
Affiliation(s)
- Miao Li
- Columbia University, Department of Anesthesiology, 630 West 168th Street, New York, NY, 10032, USA
| | - David Cabrera-Garcia
- Columbia University, Department of Anesthesiology, 630 West 168th Street, New York, NY, 10032, USA
| | - Michael C Salling
- Louisiana State University, Department of Anatomy, New Orleans, LA, 1901 Perdido Street, New Orleans, LA, 70112, USA
| | - Edmund Au
- Columbia University, Department of Pathology & Cell Biology and Rehabilitative Medicine and Regeneration, Columbia Translational Neuroscience Initiative Scholar, 630 West 168th Street, New York, NY, 10032, USA
| | - Guang Yang
- Columbia University, Department of Anesthesiology, 630 West 168th Street, New York, NY, 10032, USA.
| | - Neil L Harrison
- Columbia University, Department of Anesthesiology, 630 West 168th Street, New York, NY, 10032, USA; Columbia University, Department of Molecular Pharmacology and Therapeutics, 630 West 168th Street, New York, NY, 10032, USA.
| |
Collapse
|
15
|
Abstract
Alcohol is one of the oldest pharmacological agents used for its sedative/hypnotic effects, and alcohol abuse and alcohol use disorder (AUD) continues to be major public health issue. AUD is strongly indicated to be a brain disorder, and the molecular and cellular mechanism/s by which alcohol produces its effects in the brain are only now beginning to be understood. In the brain, synaptic plasticity or strengthening or weakening of synapses, can be enhanced or reduced by a variety of stimulation paradigms. Synaptic plasticity is thought to be responsible for important processes involved in the cellular mechanisms of learning and memory. Long-term potentiation (LTP) is a form of synaptic plasticity, and occurs via N-methyl-D-aspartate type glutamate receptor (NMDAR or GluN) dependent and independent mechanisms. In particular, NMDARs are a major target of alcohol, and are implicated in different types of learning and memory. Therefore, understanding the effect of alcohol on synaptic plasticity and transmission mediated by glutamatergic signaling is becoming important, and this will help us understand the significant contribution of the glutamatergic system in AUD. In the first part of this review, we will briefly discuss the mechanisms underlying long term synaptic plasticity in the dorsal striatum, neocortex and the hippocampus. In the second part we will discuss how alcohol (ethanol, EtOH) can modulate long term synaptic plasticity in these three brain regions, mainly from neurophysiological and electrophysiological studies. Taken together, understanding the mechanism(s) underlying alcohol induced changes in brain function may lead to the development of more effective therapeutic agents to reduce AUDs.
Collapse
Affiliation(s)
| | - Chitra D Mandyam
- VA San Diego Healthcare System, San Diego, CA, USA.,Department of Anesthesiology, University of California San Diego, San Diego, CA, USA
| |
Collapse
|
16
|
Morningstar MD, Linsenbardt DN, Lapish CC. Ethanol Alters Variability, But Not Rate, of Firing in Medial Prefrontal Cortex Neurons of Awake-Behaving Rats. Alcohol Clin Exp Res 2020; 44:2225-2238. [PMID: 32966634 PMCID: PMC7680402 DOI: 10.1111/acer.14463] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 09/12/2020] [Indexed: 11/26/2022]
Abstract
BACKGROUND The medial prefrontal cortex (mPFC) is a brain region involved in the evaluation and selection of motivationally relevant outcomes. Neural activity in mPFC is altered following acute ethanol (EtOH) use and, in rodent models, doses as low as 0.75 g/kg yield cognitive deficits. Deficits in decision making following acute EtOH are thought to be mediated, at least in part, by decreases in mPFC firing rates (FRs). However, the data leading to this conclusion have been generated exclusively in anesthetized rodents. The present study characterizes the effects of acute EtOH injections on mPFC neural activity in awake-behaving rodents. METHODS Awake-behaving and anesthetized in vivo electrophysiological recordings were performed. We utilized 3 groups: the first received 2 saline injections, the second received a saline injection followed by 1.0 g/kg EtOH, and the last received saline followed by 2 g/kg EtOH. One week later, an anesthetized recording occurred where a saline injection was followed by an injection of 1.0 g/kg EtOH. RESULTS The anesthetized condition showed robust decreases in neural activity and differences in up-down states (UDS) dynamics. In the awake-behaving condition, FRs were grouped according to behavioral state: moving, not-moving, and sleep. The differences in median FRs were found for each treatment and behavioral state combination. A FR decrease was only found in the 2.0 g/kg EtOH treatment during not-moving states. However, robust decreases in FR variability were found across behavioral state in both the 1.0 and 2.0 g/kg EtOH treatment. Sleep was separately analyzed. EtOH modulated the UDS during sleep producing decreases in FRs. CONCLUSIONS In conclusion, the changes in neural activity following EtOH administration in anesthetized animals are not conserved in awake-behaving animals. The most prominent difference following EtOH was a decrease in FR variability suggesting that acute EtOH may be affecting decision making via this mechanism.
Collapse
|
17
|
Shang P, Lindberg D, Starski P, Peyton L, Hong SI, Choi S, Choi DS. Chronic Alcohol Exposure Induces Aberrant Mitochondrial Morphology and Inhibits Respiratory Capacity in the Medial Prefrontal Cortex of Mice. Front Neurosci 2020; 14:561173. [PMID: 33192248 PMCID: PMC7646256 DOI: 10.3389/fnins.2020.561173] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 09/23/2020] [Indexed: 12/23/2022] Open
Abstract
Alcohol use disorder (AUD) is characterized as a chronic, relapsing disease with a pattern of excessive drinking despite negative consequences to an individual's life. Severe chronic alcohol use impairs the function of the medial prefrontal cortex (mPFC), which contributes to alcohol-induced cognitive and executive dysfunction. The mPFC contains more mitochondria compared to other cortical areas, which suggests mitochondrial damage may occur in AUD and trigger subsequent behavior change. Here, we identified morphological and functional changes in mitochondria in the mPFC in C57BL6/J mice after 8 h of withdrawal from chronic intermittent alcohol (CIA) exposure. Three-dimensional serial block-face scanning electron microscopy (SBFSEM) reconstruction revealed that CIA exposure elongated mPFC mitochondria and formed mitochondria-on-a-string (MOAS). Furthermore, alcohol significantly affected mitochondrial bioenergetics, including oxidative phosphorylation and electron transport, with inhibited aerobic respiration in mPFC mitochondria after CIA exposure. We also found decreased expression of fusion (mitofusin 2, Mfn2) and increased fission (mitochondrial fission 1 protein, Fis1) proteins in the mPFC of alcohol-treated mice. In sum, our study suggests that CIA exposure impairs mitochondrial dynamics and function in the mPFC.
Collapse
Affiliation(s)
- Pei Shang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
- Department of Neurology, First Hospital of Jilin University, Changchun, China
| | - Daniel Lindberg
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
| | - Phillip Starski
- Neuroscience Program, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
| | - Lee Peyton
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
| | - Sa-Ik Hong
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
| | - Sun Choi
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
| | - Doo-Sup Choi
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
- Neuroscience Program, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
- Department of Psychiatry and Psychology, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
| |
Collapse
|
18
|
García-Marchena N, Pizarro N, Pavón FJ, Martínez-Huélamo M, Flores-López M, Requena-Ocaña N, Araos P, Silva-Peña D, Suárez J, Santín LJ, de la Torre R, Rodríguez de Fonseca F, Serrano A. Potential association of plasma lysophosphatidic acid (LPA) species with cognitive impairment in abstinent alcohol use disorders outpatients. Sci Rep 2020; 10:17163. [PMID: 33051508 PMCID: PMC7555527 DOI: 10.1038/s41598-020-74155-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 09/28/2020] [Indexed: 12/17/2022] Open
Abstract
Lysophosphatidic acid (LPA) species are bioactive lipids participating in neurodevelopmental processes. The aim was to investigate whether the relevant species of LPA were associated with clinical features of alcohol addiction. A total of 55 abstinent alcohol use disorder (AUD) patients were compared with 34 age/sex/body mass index-matched controls. Concentrations of total LPA and 16:0-LPA, 18:0-LPA, 18:1-LPA, 18:2-LPA and 20:4-LPA species were quantified and correlated with neuroplasticity-associated growth factors including brain derived neurotrophic factor (BDNF), insulin-like growth factor-1 (IGF-1) and IGF-2, and neurotrophin-3 (NT-3). AUD patients showed dysexecutive syndrome (22.4%) and memory impairment (32.6%). Total LPA, 16:0-LPA, 18:0-LPA and 18:1-LPA concentrations, were decreased in the AUD group compared to control group. Total LPA, 16:0-LPA, 18:2-LPA and 20:4-LPA concentrations were decreased in men compared to women. Frontal lobe functions correlated with plasma LPA species. Alcohol-cognitive impairments could be related with the deregulation of the LPA species, especially in 16:0-LPA, 18:1-LPA and 20:4-LPA. Concentrations of BDNF correlated with total LPA, 18:2-LPA and 20:4-LPA species. The relation between LPA species and BDNF is interesting in plasticity and neurogenesis functions, their involvement in AUD might serve as a biomarker of cognitive impairment.
Collapse
Affiliation(s)
- Nuria García-Marchena
- Laboratorio de Medicina Regenerativa, Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Avda. Carlos Haya 82, sótano, 29010, Málaga, Spain. .,Institut D, Investigació en Ciències de la Salut Germans Trias i Pujol (IGTP), Unidad de Adicciones-Servicio de Medicina Interna, Campus Can Ruti, Carrer del Canyet s/n, 08916, Badalona, Spain.
| | - Nieves Pizarro
- Integrative Pharmacology and Systems Neurosciences Research Group, Programa de Investigación en Neurociencias, Institut Hospital del Mar d'Investigacions Mediques (IMIM), Dr. Aiguader 88, 08003, Barcelona, Spain
| | - Francisco J Pavón
- Laboratorio de Medicina Regenerativa, Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Avda. Carlos Haya 82, sótano, 29010, Málaga, Spain.,Unidad de Gestión Clínica del Corazón, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria de Málaga, Malaga, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Miriam Martínez-Huélamo
- Integrative Pharmacology and Systems Neurosciences Research Group, Programa de Investigación en Neurociencias, Institut Hospital del Mar d'Investigacions Mediques (IMIM), Dr. Aiguader 88, 08003, Barcelona, Spain.,Departamento de Nutrición, Ciencias de los Alimentos y Gastronomía, Facultad de Farmacia y Ciencias de los Alimentos, Universidad de Barcelona, Barcelona, Spain
| | - María Flores-López
- Laboratorio de Medicina Regenerativa, Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Avda. Carlos Haya 82, sótano, 29010, Málaga, Spain
| | - Nerea Requena-Ocaña
- Laboratorio de Medicina Regenerativa, Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Avda. Carlos Haya 82, sótano, 29010, Málaga, Spain
| | - Pedro Araos
- Laboratorio de Medicina Regenerativa, Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Avda. Carlos Haya 82, sótano, 29010, Málaga, Spain.,Departamento de Psicobiología y Metodología de las Ciencias del Comportamiento, Instituto de Investigación Biomédica de Málaga (IBIMA), Facultad de Psicología, Universidad de Málaga (UMA), Malaga, Spain
| | - Daniel Silva-Peña
- Laboratorio de Medicina Regenerativa, Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Avda. Carlos Haya 82, sótano, 29010, Málaga, Spain
| | - Juan Suárez
- Laboratorio de Medicina Regenerativa, Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Avda. Carlos Haya 82, sótano, 29010, Málaga, Spain
| | - Luis J Santín
- Departamento de Psicobiología y Metodología de las Ciencias del Comportamiento, Instituto de Investigación Biomédica de Málaga (IBIMA), Facultad de Psicología, Universidad de Málaga (UMA), Malaga, Spain
| | - Rafael de la Torre
- Integrative Pharmacology and Systems Neurosciences Research Group, Programa de Investigación en Neurociencias, Institut Hospital del Mar d'Investigacions Mediques (IMIM), Dr. Aiguader 88, 08003, Barcelona, Spain.
| | - Fernando Rodríguez de Fonseca
- Laboratorio de Medicina Regenerativa, Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Avda. Carlos Haya 82, sótano, 29010, Málaga, Spain.
| | - Antonia Serrano
- Laboratorio de Medicina Regenerativa, Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Avda. Carlos Haya 82, sótano, 29010, Málaga, Spain.
| |
Collapse
|
19
|
Smith ML, Lopez MF, Wolen AR, Becker HC, Miles MF. Brain regional gene expression network analysis identifies unique interactions between chronic ethanol exposure and consumption. PLoS One 2020; 15:e0233319. [PMID: 32469986 PMCID: PMC7259766 DOI: 10.1371/journal.pone.0233319] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 05/01/2020] [Indexed: 11/28/2022] Open
Abstract
Progressive increases in ethanol consumption is a hallmark of alcohol use disorder (AUD). Persistent changes in brain gene expression are hypothesized to underlie the altered neural signaling producing abusive consumption in AUD. To identify brain regional gene expression networks contributing to progressive ethanol consumption, we performed microarray and scale-free network analysis of expression responses in a C57BL/6J mouse model utilizing chronic intermittent ethanol by vapor chamber (CIE) in combination with limited access oral ethanol consumption. This model has previously been shown to produce long-lasting increased ethanol consumption, particularly when combining oral ethanol access with repeated cycles of intermittent vapor exposure. The interaction of CIE and oral consumption was studied by expression profiling and network analysis in medial prefrontal cortex, nucleus accumbens, hippocampus, bed nucleus of the stria terminalis, and central nucleus of the amygdala. Brain region expression networks were analyzed for ethanol-responsive gene expression, correlation with ethanol consumption and functional content using extensive bioinformatics studies. In all brain-regions studied the largest number of changes in gene expression were seen when comparing ethanol naïve mice to those exposed to CIE and drinking. In the prefrontal cortex, however, unique patterns of gene expression were seen compared to other brain-regions. Network analysis identified modules of co-expressed genes in all brain regions. The prefrontal cortex and nucleus accumbens showed the greatest number of modules with significant correlation to drinking behavior. Across brain-regions, however, many modules with strong correlations to drinking, both baseline intake and amount consumed after CIE, showed functional enrichment for synaptic transmission and synaptic plasticity.
Collapse
Affiliation(s)
- Maren L. Smith
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Marcelo F. Lopez
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Aaron R. Wolen
- Center for Clinical and Translational Research, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Howard C. Becker
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, South Carolina, United States of America
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina, United States of America
- RHJ Department of Veterans Affairs Medical Center, Charleston, South Carolina, United States of America
| | - Michael F. Miles
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, Virginia, United States of America
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia, United States of America
- VCU Alcohol Research Center, Virginia Commonwealth University, Richmond, Virginia, United States of America
| |
Collapse
|
20
|
Saganuwan SA. Chemistry and Effects of Brainstem Acting Drugs. Cent Nerv Syst Agents Med Chem 2020; 19:180-186. [PMID: 31223094 DOI: 10.2174/1871524919666190620164355] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 05/02/2019] [Accepted: 05/07/2019] [Indexed: 01/29/2023]
Abstract
BACKGROUND Brain is the most sensitive organ, whereas brainstem is the most important part of Central Nervous System (CNS). It connects the brain and the spinal cord. However, a myriad of drugs and chemicals affects CNS with severe resultant effects on the brainstem. METHODS In view of this, a number of literature were assessed for information on the most sensitive part of brain, drugs and chemicals that act on the brainstem and clinical benefit and risk assessment of such drugs and chemicals. RESULTS Findings have shown that brainstem regulates heartbeat, respiration and because it connects the brain and spinal cord, all the drugs that act on the spinal cord may overall affect the systems controlled by the spinal cord and brain. The message is sent and received by temporal lobe, occipital lobe, frontal lobe, parietal lobe and cerebellum. CONCLUSION Hence, the chemical functional groups of the brainstem and drugs acting on brainstem are complementary, and may produce either stimulation or depression of CNS.
Collapse
Affiliation(s)
- Saganuwan Alhaji Saganuwan
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Federal University of Agriculture, P.M.B. 2373, Makurdi, Benue State, Nigeria
| |
Collapse
|
21
|
Lai CC, Hsu JW, Cheng YS, Lin HH. Involvement of metabotropic glutamate receptor 5 in ethanol regulation of NMDA receptor activity in rat substantia gelatinosa neurons. Life Sci 2019; 233:116729. [PMID: 31386876 DOI: 10.1016/j.lfs.2019.116729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 06/28/2019] [Accepted: 08/02/2019] [Indexed: 11/17/2022]
Abstract
AIMS Glutamatergic receptors are important targets of ethanol. Intake of ethanol may produce analgesic effects. The present study examined the effects of ethanol on the activity of ionotropic glutamate receptors in spinal cord substantia gelatinosa (SG) neurons, critical neurons involved in nociceptive transmission. MAIN METHODS Whole-cell recordings were made from SG neurons of the lumbar spinal cord slices from 15 to 20-day-old rats. Ethanol and glutamate receptor agonists or antagonists were applied by superfusion. KEY FINDING Ethanol (50 and 100 mM) applied by superfusion for 5 min dose-dependently decreased the amplitude of evoked excitatory postsynaptic potential in SG neurons. Superfusion of ethanol (100 mM) for 15 min consistently inhibited NMDA- or AMPA-induced depolarizations in SG neurons. Ethanol (100 mM) also inhibited the depolarizations induced by glutamate. However, ethanol inhibition of glutamate-induced responses significantly decreased at 10-15 min following continuous superfusion, suggesting the development of acute tolerance to the inhibition during prolonged exposure. Application of MPEP hydrochloride (an antagonist of metabotropic glutamate receptor [mGluR] 5) or GF109203X (a protein kinase C [PKC] inhibitor), together with ethanol significantly blocked the tolerance. The inhibition by ethanol of the NMDA-induced, but not AMPA-induced, depolarizations significantly decreased at 15 min during continuous superfusion while ACPD (a mGluR agonist) was co-applied with ethanol. SIGNIFICANCE The results suggest that (1) ethanol exposure may inhibit ionotropic glutamate receptor-mediated neurotransmission; (2) regulation of NMDA receptor function by mGluR5/PKC pathways may be involved in the development of the tolerance to ethanol inhibition of glutamate-induced responses during prolonged exposure in SG neurons.
Collapse
Affiliation(s)
- Chih-Chia Lai
- Department of Pharmacology, School of Medicine, Tzu Chi University, Hualien, Taiwan 970; Master and Ph.D. Programs in Pharmacology and Toxicology, School of Medicine, Tzu Chi University, Hualien, Taiwan 970
| | - Jhih-Wei Hsu
- Master Program in Medical Physiology, School of Medicine, Tzu Chi University, Hualien, Taiwan 970
| | - Yi-Shan Cheng
- Master and Ph.D. Programs in Pharmacology and Toxicology, School of Medicine, Tzu Chi University, Hualien, Taiwan 970
| | - Hsun-Hsun Lin
- Master Program in Medical Physiology, School of Medicine, Tzu Chi University, Hualien, Taiwan 970; Department of Physiology, School of Medicine, Tzu Chi University, Hualien, Taiwan 970.
| |
Collapse
|
22
|
Shillinglaw JE, Morrisett RA, Mangieri RA. Ethanol Modulates Glutamatergic Transmission and NMDAR-Mediated Synaptic Plasticity in the Agranular Insular Cortex. Front Pharmacol 2018; 9:1458. [PMID: 30618752 PMCID: PMC6305468 DOI: 10.3389/fphar.2018.01458] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 11/29/2018] [Indexed: 12/17/2022] Open
Abstract
The agranular insular cortex (AIC) has recently been investigated by the alcohol field because of its connectivity to and modulatory control over limbic and brainstem regions implicated in alcohol use disorder (AUD), and because it has shown involvement in animal models of alcohol drinking. Despite evidence of AIC involvement in AUD, there has not yet been an examination of whether ethanol modulates glutamatergic and γ-amino-butyric acid (GABA)ergic synaptic transmission and plasticity in the AIC. Characterizing how the synaptic transmission and plasticity states of AIC cortical processing neurons are modulated by acute ethanol will likely reveal the molecular targets by which chronic ethanol alters AIC function as alcohol drinking transitions from controlled to problematic. Therefore, we collected brain slices from ethanol-naïve adult male mice, obtained whole-cell recording configuration in layer 2/3 AIC pyramidal neurons, and bath-applied ethanol at pharmacologically relevant concentrations during electrophysiological assays of glutamatergic and GABAergic synaptic transmission and plasticity. We found that ethanol inhibited electrically evoked N-methyl-D-aspartate receptor (NMDAR)-mediated excitatory post-synaptic currents (EPSCs) in a concentration-related fashion, and had little effect on evoked α-amino-3-hydrox-5-methylisoxazole-4-propionic acid-type receptor (AMPAR)-mediated EPSCs. Ethanol had no effect on spontaneous excitatory post-synaptic currents (sEPSCs) or inhibitory GABAAR-mediated post-synaptic currents (sIPSCs). We found that synaptic conditioning (low-frequency stimulation for 15 min at 1 Hz) induced a form of long-term depression (LTD) of evoked AMPAR-mediated EPSCs. The ability to induce LTD was inhibited by a non-selective NMDAR antagonist (DL-2-amino-5-phosphonovaleric acid), and also by acute, intoxicating concentrations of ethanol. Taken together these data suggest that the glutamate, but not GABA system in the AIC is uniquely sensitive to ethanol, and that in particular NMDAR-mediated processes in the AIC may be disrupted by pharmacologically relevant concentrations of ethanol.
Collapse
Affiliation(s)
- Joel E Shillinglaw
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX, United States
| | - Richard A Morrisett
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX, United States
| | - Regina A Mangieri
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX, United States
| |
Collapse
|
23
|
Yamada A, Koga K, Kume K, Ohsawa M, Furue H. Ethanol-induced enhancement of inhibitory synaptic transmission in the rat spinal substantia gelatinosa. Mol Pain 2018; 14:1744806918817969. [PMID: 30453825 PMCID: PMC6293375 DOI: 10.1177/1744806918817969] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Recent studies have shown that ethanol produces a widespread modulation
of neuronal activity in the central nervous system. It is not fully
understood, however, how ethanol changes nociceptive transmission. We
investigated acute effects of ethanol on synaptic transmission in the
substantia gelatinosa (lamina II of the spinal dorsal horn) and
mechanical responses in the spinal dorsal horn. In substantia
gelatinosa neurons, bath application of ethanol at low concentration
(10 mM) did not change the frequency and amplitude of spontaneous
inhibitory postsynaptic currents. At medium to high concentrations
(20–100 mM), however, ethanol elicited a barrage of large amplitude
spontaneous inhibitory postsynaptic currents. In the presence of
tetrodotoxin, such enhancement of spontaneous inhibitory postsynaptic
currents was not detected. In addition, ethanol (20–100 mM) increased
the frequency of spontaneous discharge of vesicular GABA
transporter-Venus-labeled neurons and suppressed the mechanical
nociceptive response in wide-dynamic range neurons in the spinal
dorsal horn. The present results suggest that ethanol may reduce
nociceptive information transfer in the spinal dorsal horn by
enhancement of inhibitory GABAergic and glycinergic synaptic
transmission.
Collapse
Affiliation(s)
- Akihiro Yamada
- Department of Neurophysiology, Hyogo College of
Medicine, Nishinomiya, Japan
- Department of Neuropharmacology, Graduate School of
Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
- Department of Information Physiology, National
Institute for Physiological Sciences, Okazaki, Japan
| | - Kohei Koga
- Department of Neurophysiology, Hyogo College of
Medicine, Nishinomiya, Japan
| | - Kazuhiko Kume
- Department of Neuropharmacology, Graduate School of
Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | - Masahiro Ohsawa
- Department of Neuropharmacology, Graduate School of
Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | - Hidemasa Furue
- Department of Neurophysiology, Hyogo College of
Medicine, Nishinomiya, Japan
- Department of Information Physiology, National
Institute for Physiological Sciences, Okazaki, Japan
- School of Life Science, Graduate University for
Advanced Studies, Okazaki, Japan
- Hidemasa Furue, Department of
Neurophysiology 663–8131, Hyogo College of Medicine, Nishinomiya,
Japan.
| |
Collapse
|
24
|
Role of glutamatergic system and mesocorticolimbic circuits in alcohol dependence. Prog Neurobiol 2018; 171:32-49. [PMID: 30316901 DOI: 10.1016/j.pneurobio.2018.10.001] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 09/08/2018] [Accepted: 10/08/2018] [Indexed: 02/06/2023]
Abstract
Emerging evidence demonstrates that alcohol dependence is associated with dysregulation of several neurotransmitters. Alterations in dopamine, glutamate and gamma-aminobutyric acid release are linked to chronic alcohol exposure. The effects of alcohol on the glutamatergic system in the mesocorticolimbic areas have been investigated extensively. Several studies have demonstrated dysregulation in the glutamatergic systems in animal models exposed to alcohol. Alcohol exposure can lead to an increase in extracellular glutamate concentrations in mesocorticolimbic brain regions. In addition, alcohol exposure affects the expression and functions of several glutamate receptors and glutamate transporters in these brain regions. In this review, we discussed the effects of alcohol exposure on glutamate receptors, glutamate transporters and glutamate homeostasis in each area of the mesocorticolimbic system. In addition, we discussed the genetic aspect of alcohol associated with glutamate and reward circuitry. We also discussed the potential therapeutic role of glutamate receptors and glutamate transporters in each brain region for the treatment of alcohol dependence. Finally, we provided some limitations on targeting the glutamatergic system for potential therapeutic options for the treatment alcohol use disorders.
Collapse
|
25
|
Alcohol Consumption during Adolescence in a Mouse Model of Binge Drinking Alters the Intrinsic Excitability and Function of the Prefrontal Cortex through a Reduction in the Hyperpolarization-Activated Cation Current. J Neurosci 2018; 38:6207-6222. [PMID: 29915134 DOI: 10.1523/jneurosci.0550-18.2018] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 05/24/2018] [Accepted: 05/26/2018] [Indexed: 12/20/2022] Open
Abstract
Periodic episodes of excessive alcohol consumption ("binge drinking") occur frequently among adolescents, and early binge drinking is associated with an increased risk of alcohol use disorders later in life. The PFC undergoes significant development during adolescence and hence may be especially susceptible to the effects of binge drinking. In humans and in animal models, adolescent alcohol exposure is known to alter PFC neuronal activity and produce deficits in PFC-dependent behaviors, such as decision making, response inhibition, and working memory. Using a voluntary intermittent access to alcohol (IA EtOH) procedure in male mice, we demonstrate that binge-level alcohol consumption during adolescence leads to altered drinking patterns and working memory deficits in young adulthood, two outcomes that suggest medial PFC dysfunction. We recorded from pyramidal neurons (PNs) in the prelimbic subregion of the medial PFC in slices obtained from mice that had IA EtOH and found that they display altered excitability, including a hyperpolarization of the resting membrane potential and reductions in the hyperpolarization-activated cation current (Ih) and in intrinsic persistent activity (a mode of neuronal firing that is dependent on Ih). Many of these effects on intrinsic excitability were sustained following abstinence and observed in mice that showed working memory deficits. In addition, we found that resting membrane potential and the Ih-dependent voltage "sag" in prelimbic PFC PNs are developmentally regulated during adolescence, suggesting that adolescent alcohol exposure may compromise PFC function by arresting the normal developmental trajectory of PN intrinsic excitability.SIGNIFICANCE STATEMENT Binge alcohol drinking during adolescence has negative consequences for the function of the developing PFC. Using a mouse model of voluntary binge drinking during adolescence, we found that this behavior leads to working memory deficits and altered drinking behavior in adulthood. In addition, we found that adolescent drinking is associated with specific changes to the intrinsic excitability of pyramidal neurons in the PFC, reducing the ability of these neurons to generate intrinsic persistent activity, a phenomenon thought to be important for working memory. These findings may help explain why human adolescent binge drinkers show performance deficits on tasks mediated by the PFC.
Collapse
|
26
|
Klenowski PM. Emerging role for the medial prefrontal cortex in alcohol-seeking behaviors. Addict Behav 2018; 77:102-106. [PMID: 28992574 DOI: 10.1016/j.addbeh.2017.09.024] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 09/01/2017] [Accepted: 09/27/2017] [Indexed: 01/01/2023]
Abstract
The medial prefrontal cortex (mPFC) plays an important role in high-order executive processes and sends highly organized projections to sub-cortical regions controlling mood, motivation and impulsivity. Recent preclinical and clinical studies have demonstrated alcohol-induced effects on the activity and composition of the PFC which are implicated in associative learning processes and may disrupt executive control over impulsivity, leading to an inability to self-limit alcohol intake. Animal studies have begun to dissect the role of the mPFC circuitry in alcohol-seeking behavior and withdrawal, and have identified a key role for projections to sub-cortical sites including the extended amygdala and the nucleus accumbens (NAc). Importantly, these studies have highlighted that alcohol can have contrasting effects on the mPFC compared to other addictive substances and also produce differential effects on the structure and activity of the mPFC following short-term versus long-term consumption. Because of these differences, how the mPFC influences the initial aspects of alcohol-seeking behavior and how we can better understand the long-term effects of alcohol use on the activity and connectivity of the mPFC need to be considered. Given the lack of preclinical data from long-term drinking models, an increased focus should be directed towards identifying how long-term alcohol use changes the mPFC, in order to provide new insights into the mechanisms underlying the transition to dependence.
Collapse
|
27
|
The Cerebellar GABA AR System as a Potential Target for Treating Alcohol Use Disorder. Handb Exp Pharmacol 2018; 248:113-156. [PMID: 29736774 DOI: 10.1007/164_2018_109] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In the brain, fast inhibitory neurotransmission is mediated primarily by the ionotropic subtype of the gamma-aminobutyric acid (GABA) receptor subtype A (GABAAR). It is well established that the brain's GABAAR system mediates many aspects of neurobehavioral responses to alcohol (ethanol; EtOH). Accordingly, in both preclinical studies and some clinical scenarios, pharmacologically targeting the GABAAR system can alter neurobehavioral responses to acute and chronic EtOH consumption. However, many of the well-established interactions of EtOH and the GABAAR system have been identified at concentrations of EtOH ([EtOH]) that would only occur during abusive consumption of EtOH (≥40 mM), and there are still inadequate treatment options for prevention of or recovery from alcohol use disorder (AUD, including abuse and dependence). Accordingly, there is a general acknowledgement that more research is needed to identify and characterize: (1) neurobehavioral targets of lower [EtOH] and (2) associated brain structures that would involve such targets in a manner that may influence the development and maintenance of AUDs.Nearly 15 years ago it was discovered that the GABAAR system of the cerebellum is highly sensitive to EtOH, responding to concentrations as low as 10 mM (as would occur in the blood of a typical adult human after consuming 1-2 standard units of EtOH). This high sensitivity to EtOH, which likely mediates the well-known motor impairing effects of EtOH, combined with recent advances in our understanding of the role of the cerebellum in non-motor, cognitive/emotive/reward processes has renewed interest in this system in the specific context of AUD. In this chapter we will describe recent advances in our understanding of cerebellar processing, actions of EtOH on the cerebellar GABAAR system, and the potential relationship of such actions to the development of AUD. We will finish with speculation about how cerebellar specific GABAAR ligands might be effective pharmacological agents for treating aspects of AUD.
Collapse
|
28
|
Abrahao KP, Salinas AG, Lovinger DM. Alcohol and the Brain: Neuronal Molecular Targets, Synapses, and Circuits. Neuron 2017; 96:1223-1238. [PMID: 29268093 PMCID: PMC6566861 DOI: 10.1016/j.neuron.2017.10.032] [Citation(s) in RCA: 268] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 09/30/2017] [Accepted: 10/27/2017] [Indexed: 12/13/2022]
Abstract
Ethanol is one of the most commonly abused drugs. Although environmental and genetic factors contribute to the etiology of alcohol use disorders, it is ethanol's actions in the brain that explain (1) acute ethanol-related behavioral changes, such as stimulant followed by depressant effects, and (2) chronic changes in behavior, including escalated use, tolerance, compulsive seeking, and dependence. Our knowledge of ethanol use and abuse thus relies on understanding its effects on the brain. Scientists have employed both bottom-up and top-down approaches, building from molecular targets to behavioral analyses and vice versa, respectively. This review highlights current progress in the field, focusing on recent and emerging molecular, cellular, and circuit effects of the drug that impact ethanol-related behaviors. The focus of the field is now on pinpointing which molecular effects in specific neurons within a brain region contribute to behavioral changes across the course of acute and chronic ethanol exposure.
Collapse
Affiliation(s)
- Karina P Abrahao
- Laboratory for Integrative Neuroscience, Division of Intramural Clinical and Biological Research, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD 20892, USA
| | - Armando G Salinas
- Laboratory for Integrative Neuroscience, Division of Intramural Clinical and Biological Research, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD 20892, USA
| | - David M Lovinger
- Laboratory for Integrative Neuroscience, Division of Intramural Clinical and Biological Research, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD 20892, USA.
| |
Collapse
|
29
|
Harrison NL, Skelly MJ, Grosserode EK, Lowes DC, Zeric T, Phister S, Salling MC. Effects of acute alcohol on excitability in the CNS. Neuropharmacology 2017; 122:36-45. [PMID: 28479395 DOI: 10.1016/j.neuropharm.2017.04.007] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 04/04/2017] [Accepted: 04/06/2017] [Indexed: 01/23/2023]
Abstract
Alcohol has many effects on brain function and hence on human behavior, ranging from anxiolytic and mild disinhibitory effects, sedation and motor incoordination, amnesia, emesis, hypnosis and eventually unconsciousness. In recent years a variety of studies have shown that acute and chronic exposure to alcohol can modulate ion channels that regulate excitability. Modulation of intrinsic excitability provides another way in which alcohol can influence neuronal network activity, in addition to its actions on synaptic inputs. In this review, we review "low dose" effects [between 2 and 20 mM EtOH], and "medium dose"; effects [between 20 and 50 mM], by considering in turn each of the many networks and brain regions affected by alcohol, and thereby attempt to integrate in vitro physiological studies in specific brain regions (e.g. amygdala, ventral tegmental area, prefrontal cortex, thalamus, cerebellum etc.) within the context of alcohol's behavioral actions in vivo (e.g. anxiolysis, euphoria, sedation, motor incoordination). This article is part of the Special Issue entitled "Alcoholism".
Collapse
Affiliation(s)
- Neil L Harrison
- Departments of Anesthesiology and Pharmacology, College of Physicians and Surgeons, Columbia University, New York, N.Y., 10032, United States.
| | - Mary Jane Skelly
- Departments of Anesthesiology and Pharmacology, College of Physicians and Surgeons, Columbia University, New York, N.Y., 10032, United States
| | - Emma K Grosserode
- Departments of Anesthesiology and Pharmacology, College of Physicians and Surgeons, Columbia University, New York, N.Y., 10032, United States
| | - Daniel C Lowes
- Departments of Anesthesiology and Pharmacology, College of Physicians and Surgeons, Columbia University, New York, N.Y., 10032, United States
| | - Tamara Zeric
- Departments of Anesthesiology and Pharmacology, College of Physicians and Surgeons, Columbia University, New York, N.Y., 10032, United States
| | - Sara Phister
- Departments of Anesthesiology and Pharmacology, College of Physicians and Surgeons, Columbia University, New York, N.Y., 10032, United States
| | - Michael C Salling
- Departments of Anesthesiology and Pharmacology, College of Physicians and Surgeons, Columbia University, New York, N.Y., 10032, United States
| |
Collapse
|
30
|
Centanni SW, Burnett EJ, Trantham-Davidson H, Chandler LJ. Loss of δ-GABA A receptor-mediated tonic currents in the adult prelimbic cortex following adolescent alcohol exposure. Addict Biol 2017; 22:616-628. [PMID: 26804056 DOI: 10.1111/adb.12353] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Revised: 10/29/2015] [Accepted: 11/24/2015] [Indexed: 11/30/2022]
Abstract
Delayed maturation of the adolescent prefrontal cortex may render it particularly vulnerable to insults, including those associated with drugs of abuse. Using a rat model of binge alcohol exposure, the present study examined the effect of adolescent intermittent ethanol (AIE) exposure during postnatal days 28-42 on γ-aminobutyric acid (GABA)ergic neurotransmission in the prelimbic cortex. In control rats, patch-clamp electrophysiology in acute slices obtained at different postnatal ages revealed a developmental increase in the GABAA receptor-mediated tonic current in layer V pyramidal neurons but no change in layers II/III when measured in the adult. In slices from AIE-exposed rats, the amplitude of the tonic current was significantly reduced compared with controls when tested at postnatal days 45, 60 and 90-120. This AIE-induced reduction in tonic current was found to reflect attenuation of currents mediated by δ-subunit containing receptors. Consistent with this, facilitation of the tonic current by bath application of either ethanol or allopregnanolone was attenuated in slices from AIE-exposed adult rats compared with control rats. However, expression of this facilitation as a percent of the amplitude of the total current mediated by δ-GABAA receptors revealed that AIE did not alter their sensitivity to either agonist. Lastly, immunohistochemistry and Western blot analysis revealed no change in the expression of δ-GABAA subunits or their surface expression. Taken together, these studies reveal that AIE exposure results in persistent deficits in δ-GABAA tonic currents in the adult prelimbic cortex that may contribute to deficits in decision-making and behavioral control in adulthood.
Collapse
Affiliation(s)
- Samuel W. Centanni
- Department of Neuroscience; Medical University of South Carolina; Charleston SC USA
| | - Elizabeth J. Burnett
- Department of Neuroscience; Medical University of South Carolina; Charleston SC USA
| | | | - L. Judson Chandler
- Department of Neuroscience; Medical University of South Carolina; Charleston SC USA
| |
Collapse
|
31
|
Zamroziewicz M, Raskin SA, Tennen H, Austad CS, Wood RM, Fallahi CR, Dager AD, Sawyer B, Leen S, Pearlson GD. Effects of drinking patterns on prospective memory performance in college students. Neuropsychology 2017; 31:191-199. [PMID: 27841457 PMCID: PMC5280574 DOI: 10.1037/neu0000313] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
OBJECTIVE Traditional college students are at a critical juncture in the development of prospective memory (PM). Their brains are vulnerable to the effects of alcohol. METHOD There were 123 third and fourth year college students, 19-23 years old, who completed the Self-Rating Effects of Alcohol (SREA), Modified Timeline Follow-back (TFLB), Brief Young Adult Alcohol Consequences Scale (BYAACS), and Alcohol Effects Questionnaire (AEQ) once per month on a secure online database, as reported elsewhere (Dager et al., 2013). Data from the 6 months immediately before memory testing were averaged. In a single testing session participants were administered the Mini International Neuropsychiatric Interview-Diagnostic and Statistical Manual for Mental Disorders-Fourth Edition-Text Revision (MINI-DSM-IV-TR), measures of PM (event-based and time-based), and retrospective memory (RM). Based on the average score of six consecutive monthly responses to the SREA, TLFB, and AEQ, students were classified as nondrinkers, light drinkers, or heavy drinkers (as defined previously; Dager et al., 2013). Alcohol-induced amnesia (blackout) was measured with the BYAACS. RESULTS We found a relationship between these alcohol use classifications and time-based PM, such that participants who were classified as heavier drinkers were more likely to forget to perform the time-based PM task. We also found that self-reported alcohol-induced amnesia (blackouts) during the month immediately preceding memory testing was associated with lower performance on the event-based PM task. Participants' ability to recall the RM tasks suggested the PM items were successfully encoded even when they were not carried out, and we observed no relationship between alcohol use and RM performance. CONCLUSION Heavy alcohol use in college students may be related to impairments in PM. (PsycINFO Database Record
Collapse
Affiliation(s)
| | | | - Howard Tennen
- Department of Community Medicine and Health Care, University of Connecticut School of Medicine
| | - Carol S Austad
- Department of Psychology, Central Connecticut State University
| | - Rebecca M Wood
- Department of Psychology, Central Connecticut State University
| | | | | | | | | | | |
Collapse
|
32
|
Klenowski PM, Fogarty MJ, Shariff M, Belmer A, Bellingham MC, Bartlett SE. Increased Synaptic Excitation and Abnormal Dendritic Structure of Prefrontal Cortex Layer V Pyramidal Neurons following Prolonged Binge-Like Consumption of Ethanol. eNeuro 2016; 3:ENEURO.0248-16.2016. [PMID: 28032119 PMCID: PMC5179982 DOI: 10.1523/eneuro.0248-16.2016] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 10/26/2016] [Accepted: 11/17/2016] [Indexed: 01/23/2023] Open
Abstract
Long-term alcohol use causes a multitude of neurochemical changes in cortical regions that facilitate the transition to dependence. Therefore, we used a model of long-term, binge-like ethanol consumption in rats to determine the effects on morphology and synaptic physiology of medial prefrontal cortex (mPFC) layer V pyramidal neurons. Following 10 weeks of ethanol consumption, we recorded synaptic currents from mPFC neurons and used neurobiotin filling to analyze their morphology. We then compared these data to measurements obtained from age-matched, water-drinking control rats. We found that long-term ethanol consumption caused a significant increase in total dendrite arbor length of mPFC layer V pyramidal neurons. Dendritic restructuring was primarily observed in basal dendrite arbors, with mPFC neurons from animals engaged in long-term ethanol drinking having significantly larger and more complex basal arbors compared with controls. These changes were accompanied by significantly increased total spine densities and spontaneous postsynaptic excitatory current frequency, suggesting that long-term binge-like ethanol consumption enhances basal excitatory synaptic transmission in mPFC layer V pyramidal neurons. Our results provide insights into the morphological and functional changes in mPFC layer V pyramidal neuronal physiology following prolonged exposure to ethanol and support changes in mPFC activity during the development of alcohol dependence.
Collapse
Affiliation(s)
- Paul M. Klenowski
- Translational Research Institute and Institute for Health and Biomedical Innovation, Queensland University of Technology, Brisbane 4000, Queensland, Australia
| | - Matthew J. Fogarty
- School of Biomedical Sciences, University of Queensland, Brisbane 4072, Queensland, Australia
| | - Masroor Shariff
- Translational Research Institute and Institute for Health and Biomedical Innovation, Queensland University of Technology, Brisbane 4000, Queensland, Australia
| | - Arnauld Belmer
- Translational Research Institute and Institute for Health and Biomedical Innovation, Queensland University of Technology, Brisbane 4000, Queensland, Australia
| | - Mark C. Bellingham
- School of Biomedical Sciences, University of Queensland, Brisbane 4072, Queensland, Australia
| | - Selena E. Bartlett
- Translational Research Institute and Institute for Health and Biomedical Innovation, Queensland University of Technology, Brisbane 4000, Queensland, Australia
| |
Collapse
|
33
|
Castilla-Ortega E, Pavón FJ, Sánchez-Marín L, Estivill-Torrús G, Pedraza C, Blanco E, Suárez J, Santín L, Rodríguez de Fonseca F, Serrano A. Both genetic deletion and pharmacological blockade of lysophosphatidic acid LPA1 receptor results in increased alcohol consumption. Neuropharmacology 2016; 103:92-103. [DOI: 10.1016/j.neuropharm.2015.12.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2015] [Revised: 11/25/2015] [Accepted: 12/11/2015] [Indexed: 12/21/2022]
|
34
|
den Hartog C, Zamudio-Bulcock P, Nimitvilai S, Gilstrap M, Eaton B, Fedarovich H, Motts A, Woodward JJ. Inactivation of the lateral orbitofrontal cortex increases drinking in ethanol-dependent but not non-dependent mice. Neuropharmacology 2016; 107:451-459. [PMID: 27016020 DOI: 10.1016/j.neuropharm.2016.03.031] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 02/05/2016] [Accepted: 03/21/2016] [Indexed: 12/01/2022]
Abstract
Long-term consumption of ethanol affects cortical areas that are important for learning and memory, cognition, and decision-making. Deficits in cortical function may contribute to alcohol-abuse disorders by impeding an individual's ability to control drinking. Previous studies from this laboratory show that acute ethanol reduces activity of lateral orbitofrontal cortex (LOFC) neurons while chronic exposure impairs LOFC-dependent reversal learning and induces changes in LOFC excitability. Despite these findings, the role of LOFC neurons in ethanol consumption is unknown. To address this issue, we examined ethanol drinking in adult C57Bl/6J mice that received an excitotoxic lesion or viral injection of the inhibitory DREADD (designer receptor exclusively activated by designer drug) into the LOFC. No differences in ethanol consumption were observed between sham and lesioned mice during access to increasing concentrations of ethanol (3-40%) every other day for 7 weeks. Adulterating the ethanol solution with saccharin (0.2%) or quinine (0.06 mM) enhanced or inhibited, respectively, consumption of the 40% ethanol solution similarly in both groups. Using a chronic intermittent ethanol (CIE) vapor exposure model that produces dependence, we found no difference in baseline drinking between sham and lesioned mice prior to vapor treatments. CIE enhanced drinking in both groups as compared to air-treated animals and CIE treated lesioned mice showed an additional increase in ethanol drinking as compared to CIE sham controls. This effect persisted during the first week when quinine was added to the ethanol solution but consumption decreased to control levels in CIE lesioned mice in the following 2 weeks. In viral injected mice, baseline drinking was not altered by expression of the inhibitory DREADD receptor and repeated cycles of CIE exposure enhanced drinking in DREADD and virus control groups. Consistent with the lesion study, treatment with clozapine-N-oxide (CNO) further enhanced consumption only in CIE exposed DREADD mice with no change in air-treated mice. These results suggest that the LOFC is not critical for the initiation and maintenance of ethanol drinking in non-dependent mice, but may regulate the escalated drinking observed during dependence.
Collapse
Affiliation(s)
- Carolina den Hartog
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA; Addiction Sciences Division, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Paula Zamudio-Bulcock
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA; Addiction Sciences Division, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Sudarat Nimitvilai
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA; Addiction Sciences Division, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Meghin Gilstrap
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA; Addiction Sciences Division, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Bethany Eaton
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA; Addiction Sciences Division, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Hleb Fedarovich
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA; Addiction Sciences Division, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Andrew Motts
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA; Addiction Sciences Division, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - John J Woodward
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA; Addiction Sciences Division, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC 29425, USA.
| |
Collapse
|
35
|
Bell RL, Hauser S, Rodd ZA, Liang T, Sari Y, McClintick J, Rahman S, Engleman EA. A Genetic Animal Model of Alcoholism for Screening Medications to Treat Addiction. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2016; 126:179-261. [PMID: 27055615 PMCID: PMC4851471 DOI: 10.1016/bs.irn.2016.02.017] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The purpose of this review is to present up-to-date pharmacological, genetic, and behavioral findings from the alcohol-preferring P rat and summarize similar past work. Behaviorally, the focus will be on how the P rat meets criteria put forth for a valid animal model of alcoholism with a highlight on its use as an animal model of polysubstance abuse, including alcohol, nicotine, and psychostimulants. Pharmacologically and genetically, the focus will be on the neurotransmitter and neuropeptide systems that have received the most attention: cholinergic, dopaminergic, GABAergic, glutamatergic, serotonergic, noradrenergic, corticotrophin releasing hormone, opioid, and neuropeptide Y. Herein, we sought to place the P rat's behavioral and neurochemical phenotypes, and to some extent its genotype, in the context of the clinical literature. After reviewing the findings thus far, this chapter discusses future directions for expanding the use of this genetic animal model of alcoholism to identify molecular targets for treating drug addiction in general.
Collapse
Affiliation(s)
- R L Bell
- Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN, United States.
| | - S Hauser
- Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Z A Rodd
- Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN, United States
| | - T Liang
- Indiana University School of Medicine, Indianapolis, IN, United States
| | - Y Sari
- University of Toledo, Toledo, OH, United States
| | - J McClintick
- Center for Medical Genomics, Indiana University School of Medicine, Indianapolis, IN, United States
| | - S Rahman
- Department of Pharmaceutical Sciences, South Dakota State University, Brookings, SD, United States
| | - E A Engleman
- Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
36
|
McKim TH, Shnitko TA, Robinson DL, Boettiger CA. Translational Research on Habit and Alcohol. CURRENT ADDICTION REPORTS 2016; 3:37-49. [PMID: 26925365 DOI: 10.1007/s40429-016-0089-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Habitual actions enable efficient daily living, but they can also contribute to pathological behaviors that resistant change, such as alcoholism. Habitual behaviors are learned actions that appear goal-directed but are in fact no longer under the control of the action's outcome. Instead, these actions are triggered by stimuli, which may be exogenous or interoceptive, discrete or contextual. A major hallmark characteristic of alcoholism is continued alcohol use despite serious negative consequences. In essence, although the outcome of alcohol seeking and drinking is dramatically devalued, these actions persist, often triggered by environmental cues associated with alcohol use. Thus, alcoholism meets the definition of an initially goal-directed behavior that converts to a habit-based process. Habit and alcohol have been well investigated in rodent models, with comparatively less research in non-human primates and people. This review focuses on translational research on habit and alcohol with an emphasis on cross-species methodology and neural circuitry.
Collapse
Affiliation(s)
- Theresa H McKim
- University of North Carolina at Chapel Hill, Department of Psychology and Neuroscience, Davie Hall, CB #3270, Chapel Hill, NC 27599
| | - Tatiana A Shnitko
- University of North Carolina at Chapel Hill, Bowles Center for Alcohol Studies, CB #7178, Chapel Hill, NC 27599
| | - Donita L Robinson
- University of North Carolina at Chapel Hill, Department of Psychiatry, Bowles Center for Alcohol Studies, CB #7178, Chapel Hill, NC 27599
| | - Charlotte A Boettiger
- Biomedical Research Imaging Center, Bowles Center for Alcohol Studies, Davie Hall, CB #3270, Chapel Hill, NC 27599
| |
Collapse
|
37
|
Time-Course Analysis of Brain Regional Expression Network Responses to Chronic Intermittent Ethanol and Withdrawal: Implications for Mechanisms Underlying Excessive Ethanol Consumption. PLoS One 2016; 11:e0146257. [PMID: 26730594 PMCID: PMC4701666 DOI: 10.1371/journal.pone.0146257] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 12/15/2015] [Indexed: 01/17/2023] Open
Abstract
Long lasting abusive consumption, dependence, and withdrawal are characteristic features of alcohol use disorders (AUD). Mechanistically, persistent changes in gene expression are hypothesized to contribute to brain adaptations leading to ethanol toxicity and AUD. We employed repeated chronic intermittent ethanol (CIE) exposure by vapor chamber as a mouse model to simulate the cycles of ethanol exposure and withdrawal commonly seen with AUD. This model has been shown to induce progressive ethanol consumption in rodents. Brain CIE-responsive expression networks were identified by microarray analysis across five regions of the mesolimbic dopamine system and extended amygdala with tissue harvested from 0-hours to 7-days following CIE. Weighted Gene Correlated Network Analysis (WGCNA) was used to identify gene networks over-represented for CIE-induced temporal expression changes across brain regions. Differential gene expression analysis showed that long-lasting gene regulation occurred 7-days after the final cycle of ethanol exposure only in prefrontal cortex (PFC) and hippocampus. Across all brain regions, however, ethanol-responsive expression changes occurred mainly within the first 8-hours after removal from ethanol. Bioinformatics analysis showed that neuroinflammatory responses were seen across multiple brain regions at early time-points, whereas co-expression modules related to neuroplasticity, chromatin remodeling, and neurodevelopment were seen at later time-points and in specific brain regions (PFC or HPC). In PFC a module containing Bdnf was identified as highly CIE responsive in a biphasic manner, with peak changes at 0 hours and 5 days following CIE, suggesting a possible role in mechanisms underlying long-term molecular and behavioral response to CIE. Bioinformatics analysis of this network and several other modules identified Let-7 family microRNAs as potential regulators of gene expression changes induced by CIE. Our results suggest a complex temporal and regional pattern of widespread gene network responses involving neuroinflammatory and neuroplasticity related genes as contributing to physiological and behavioral responses to chronic ethanol.
Collapse
|
38
|
Fuhl A, Müller-Dahlhaus F, Lücke C, Toennes SW, Ziemann U. Low Doses of Ethanol Enhance LTD-like Plasticity in Human Motor Cortex. Neuropsychopharmacology 2015; 40:2969-80. [PMID: 26038159 PMCID: PMC4864632 DOI: 10.1038/npp.2015.151] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2015] [Revised: 05/25/2015] [Accepted: 05/25/2015] [Indexed: 12/26/2022]
Abstract
Humans liberally use ethanol for its facilitating effects on social interactions but its effects on central nervous system function remain underexplored. We have recently described that very low doses of ethanol abolish long-term potentiation (LTP)-like plasticity in human cortex, most likely through enhancement of tonic inhibition [Lücke et al, 2014, Neuropsychopharmacology 39:1508-18]. Here, we studied the effects of low-dose ethanol on long-term depression (LTD)-like plasticity. LTD-like plasticity was induced in human motor cortex by paired associative transcranial magnetic stimulation (PASLTD), and measured as decreases of motor evoked potential input-output curve (IO-curve). In addition, sedation was measured by decreases in saccade peak velocity (SPV). Ethanol in two low doses (EtOH<10mM, EtOH<20mM) was compared to single oral doses of alprazolam (APZ, 1mg) a classical benzodiazepine, and zolpidem (ZLP, 10 mg), a non-benzodiazepine hypnotic, in a double-blinded randomized placebo-controlled crossover design in ten healthy human subjects. EtOH<10mM and EtOH<20mM but not APZ or ZLP enhanced the PASLTD-induced LTD-like plasticity, while APZ and ZLP but not EtOH<10mM or EtOH<20mM decreased SPV. Non-sedating low doses of ethanol, easily reached during social drinking, enhance LTD-like plasticity in human cortex. This effect is most likely explained by the activation of extrasynaptic α4-subunit containing gamma-aminobutyric type A receptors by low-dose EtOH, resulting in increased tonic inhibition. Findings may stimulate cellular research on the role of tonic inhibition in regulating excitability and plasticity of cortical neuronal networks.
Collapse
Affiliation(s)
- Anna Fuhl
- Department of Neurology, Goethe-University, Frankfurt am Main, Germany
| | - Florian Müller-Dahlhaus
- Department of Neurology, Goethe-University, Frankfurt am Main, Germany,Department of Neurology & Stroke, and Hertie Institute for Clinical Brain Research, Eberhard Karls University, Tübingen, Germany
| | - Caroline Lücke
- Department of Neurology, Goethe-University, Frankfurt am Main, Germany,Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Goethe-University, Frankfurt am Main, Germany
| | - Stefan W Toennes
- Department of Forensic Toxicology, Goethe-University, Frankfurt am Main, Germany
| | - Ulf Ziemann
- Department of Neurology, Goethe-University, Frankfurt am Main, Germany,Department of Neurology & Stroke, and Hertie Institute for Clinical Brain Research, Eberhard Karls University, Tübingen, Germany,Department of Neurology & Stroke, and Hertie Institute for Clinical Brain Research, Eberhard Karls University Tübingen, Hoppe-Seyler-Str. 3, Tübingen, D-72076, Germany, Tel: +49 7071 2982049, Fax: +49 7071 295260, E-mail:
| |
Collapse
|
39
|
Hu W, Morris B, Carrasco A, Kroener S. Effects of acamprosate on attentional set-shifting and cellular function in the prefrontal cortex of chronic alcohol-exposed mice. Alcohol Clin Exp Res 2015; 39:953-61. [PMID: 25903298 PMCID: PMC10782929 DOI: 10.1111/acer.12722] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 03/05/2015] [Indexed: 12/01/2022]
Abstract
BACKGROUND The medial prefrontal cortex (mPFC) inhibits impulsive and compulsive behaviors that characterize drug abuse and dependence. Acamprosate is the leading medication approved for the maintenance of abstinence, shown to reduce craving and relapse in animal models and human alcoholics. Whether acamprosate can modulate executive functions that are impaired by chronic ethanol (EtOH) exposure is unknown. Here we explored the effects of acamprosate on an attentional set-shifting task and tested whether these behavioral effects are correlated with modulation of glutamatergic synaptic transmission and intrinsic excitability of mPFC neurons. METHODS We induced alcohol dependence in mice via chronic intermittent EtOH (CIE) exposure in vapor chambers and measured changes in alcohol consumption in a limited access 2-bottle choice paradigm. Impairments of executive function were assessed in an attentional set-shifting task. Acamprosate was applied subchronically for 2 days during withdrawal before the final behavioral test. Alcohol-induced changes in cellular function of layer 5/6 pyramidal neurons, and the potential modulation of these changes by acamprosate, were measured using patch clamp recordings in brain slices. RESULTS Chronic EtOH exposure impaired cognitive flexibility in the attentional set-shifting task. Acamprosate improved overall performance and reduced perseveration. Recordings of mPFC neurons showed that chronic EtOH exposure increased use-dependent presynaptic transmitter release and enhanced postsynaptic N-methyl-D-aspartate receptor function. Moreover, CIE treatment lowered input resistance, and decreased the threshold and the after hyperpolarization of action potentials, suggesting chronic EtOH exposure also impacted membrane excitability of mPFC neurons. However, acamprosate treatment did not reverse these EtOH-induced changes cellular function. CONCLUSIONS Acamprosate improved attentional control of EtOH exposed animals, but did not alter the concurrent changes in synaptic transmission or membrane excitability of mPFC neurons, indicating that these changes are not the pharmacological targets of acamprosate in the recovery of mPFC functions affected by chronic EtOH exposure.
Collapse
Affiliation(s)
- Wei Hu
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, Texas
| | - Brett Morris
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, Texas
| | - Angelique Carrasco
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, Texas
| | - Sven Kroener
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, Texas
| |
Collapse
|
40
|
Mishra D, Harrison NR, Gonzales CB, Schilström B, Konradsson-Geuken Å. Effects of age and acute ethanol on glutamatergic neurotransmission in the medial prefrontal cortex of freely moving rats using enzyme-based microelectrode amperometry. PLoS One 2015; 10:e0125567. [PMID: 25927237 PMCID: PMC4416039 DOI: 10.1371/journal.pone.0125567] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 03/24/2015] [Indexed: 01/04/2023] Open
Abstract
Ethanol abuse during adolescence may significantly alter development of the prefrontal cortex which continues to undergo structural remodeling into adulthood. Glutamatergic neurotransmission plays an important role during these brain maturation processes and is modulated by ethanol. In this study, we investigated glutamate dynamics in the medial prefrontal cortex of freely moving rats, using enzyme-based microelectrode amperometry. We analyzed the effects of an intraperitoneal ethanol injection (1 g/kg) on cortical glutamate levels in adolescent and adult rats. Notably, basal glutamate levels decreased with age and these levels were found to be significantly different between postnatal day (PND) 28-38 vs PND 44-55 (p<0.05) and PND 28-38 vs adult animals (p<0.001). We also observed spontaneous glutamate release (transients) throughout the recordings. The frequency of transients (per hour) was significantly higher in adolescent rats (PND 28-38 and PND 44-55) compared to those of adults. In adolescent rats, post-ethanol injection, the frequency of glutamate transients decreased within the first hour (p<0.05), it recovered slowly and in the third hour there was a significant rebound increase of the frequency (p<0.05). Our data demonstrate age-dependent differences in extracellular glutamate levels in the medial prefrontal cortex and suggest that acute ethanol injections have both inhibitory and excitatory effects in adolescent rats. These effects of ethanol on the prefrontal cortex may disturb its maturation and possibly limiting individuals´ control over addictive behaviors.
Collapse
Affiliation(s)
- Devesh Mishra
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Nicholas R. Harrison
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Carolina B. Gonzales
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Björn Schilström
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
- Department of Neuroscience and Physiology, Section of Neuropsychiatry, Gothenburg University, Gothenburg, Sweden
| | - Åsa Konradsson-Geuken
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
41
|
Navarro AI, Mandyam CD. Protracted abstinence from chronic ethanol exposure alters the structure of neurons and expression of oligodendrocytes and myelin in the medial prefrontal cortex. Neuroscience 2015; 293:35-44. [PMID: 25732140 DOI: 10.1016/j.neuroscience.2015.02.043] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 02/05/2015] [Accepted: 02/21/2015] [Indexed: 10/23/2022]
Abstract
In rodents, chronic intermittent ethanol vapor exposure (CIE) produces alcohol dependence, alters the structure and activity of pyramidal neurons and decreases the number of oligodendroglial progenitors in the medial prefrontal cortex (mPFC). In this study, adult Wistar rats were exposed to seven weeks of CIE and were withdrawn from CIE for 21 days (protracted abstinence; PA). Tissue enriched in the mPFC was processed for Western blot analysis and Golgi-Cox staining to investigate the long-lasting effects of CIE on the structure of mPFC neurons and the levels of myelin-associated proteins. PA increased dendritic arborization within apical dendrites of pyramidal neurons. These changes occurred concurrently with hypophosphorylation of the N-methyl-d-aspartate (NMDA) receptor 2B (NR2B) at Tyr-1472. PA increased myelin basic protein (MBP) levels which occurred concurrently with hypophosphorylation of the premyelinating oligodendrocyte bHLH transcription factor Olig2 in the mPFC. Given that PA is associated with increased sensitivity to stress and hypothalamic-pituitary-adrenal (HPA) axis dysregulation, and stress alters oligodendrocyte expression as a function of glucocorticoid receptor (GR) activation, the levels of total GR and phosphorylated GR were also evaluated. PA produced hypophosphorylation of the GR at Ser-232 without affecting expression of total protein. These findings demonstrate persistent and compensatory effects of ethanol in the mPFC long after cessation of CIE, including enhanced myelin production and impaired GR function. Collectively, these results suggest a novel relationship between oligodendrocytes and GR in the mPFC, in which stress may alter frontal cortex function in alcohol dependent subjects by promoting hypermyelination, thereby altering the cellular composition and white matter structure in the mPFC.
Collapse
Affiliation(s)
- A I Navarro
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
| | - C D Mandyam
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA.
| |
Collapse
|
42
|
Hillemacher T, Leggio L, Heberlein A. Investigational therapies for the pharmacological treatment of alcoholism. Expert Opin Investig Drugs 2014; 24:17-30. [PMID: 25164385 DOI: 10.1517/13543784.2014.954037] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Introduction: Alcohol dependence is one of the most important psychiatric disorders leading to enormous harm in individuals and indeed within society. Yet, although alcohol dependence is a disease of significant importance, the availability of efficacious pharmacological treatment is still limited. Areas covered: The current review focuses on neurobiological pathways that are the rationale for recent preclinical and clinical studies testing novel compounds that could be used as treatments for alcohol dependence. These neurobiological mechanisms include the: glutamatergic, dopaminergic and GABA mediated pathways as well as neuroendocrine systems. There is also an interest in the approaches for influencing chromatin structure. Expert opinion: There are several compounds in Phase I and Phase II clinical studies that have produced potentially useful results for the treating alcoholism. Further evaluation is still necessary, and the implementation of Phase III studies will help to elucidate the usefulness of these compounds. It is important that personalized approaches (e.g., pharmacogenomics) are investigated in these later studies, as the efficacy of different compounds may vary substantially between subgroups of patients.
Collapse
Affiliation(s)
- Thomas Hillemacher
- Hannover Medical School, Center for Addiction Research (CARe), Department of Psychiatry, Social Psychiatry and Psychotherapy , Carl-Neuberg-Str. 1, 30625 Hannover , Germany +49 511 532 2427 ; +49 511 532 2415 ;
| | | | | |
Collapse
|
43
|
Salling MC, Harrison NL. Strychnine-sensitive glycine receptors on pyramidal neurons in layers II/III of the mouse prefrontal cortex are tonically activated. J Neurophysiol 2014; 112:1169-78. [PMID: 24872538 DOI: 10.1152/jn.00714.2013] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Processing of signals within the cerebral cortex requires integration of synaptic inputs and a coordination between excitatory and inhibitory neurotransmission. In addition to the classic form of synaptic inhibition, another important mechanism that can regulate neuronal excitability is tonic inhibition via sustained activation of receptors by ambient levels of inhibitory neurotransmitter, usually GABA. The purpose of this study was to determine whether this occurs in layer II/III pyramidal neurons (PNs) in the prelimbic region of the mouse medial prefrontal cortex (mPFC). We found that these neurons respond to exogenous GABA and to the α4δ-containing GABAA receptor (GABA(A)R)-selective agonist gaboxadol, consistent with the presence of extrasynaptic GABA(A)R populations. Spontaneous and miniature synaptic currents were blocked by the GABA(A)R antagonist gabazine and had fast decay kinetics, consistent with typical synaptic GABA(A)Rs. Very few layer II/III neurons showed a baseline current shift in response to gabazine, but almost all showed a current shift (15-25 pA) in response to picrotoxin. In addition to being a noncompetitive antagonist at GABA(A)Rs, picrotoxin also blocks homomeric glycine receptors (GlyRs). Application of the GlyR antagonist strychnine caused a modest but consistent shift (∼15 pA) in membrane current, without affecting spontaneous synaptic events, consistent with the tonic activation of GlyRs. Further investigation showed that these neurons respond in a concentration-dependent manner to glycine and taurine. Inhibition of glycine transporter 1 (GlyT1) with sarcosine resulted in an inward current and an increase of the strychnine-sensitive current. Our data demonstrate the existence of functional GlyRs in layer II/III of the mPFC and a role for these receptors in tonic inhibition that can have an important influence on mPFC excitability and signal processing.
Collapse
Affiliation(s)
- Michael C Salling
- Department of Anesthesiology, Columbia University Medical Center, New York, New York;
| | - Neil L Harrison
- Department of Anesthesiology, Columbia University Medical Center, New York, New York; Department of Pharmacology, Columbia University, New York, New York
| |
Collapse
|
44
|
Deleterious effects of a low amount of ethanol on LTP-like plasticity in human cortex. Neuropsychopharmacology 2014; 39:1508-18. [PMID: 24385131 PMCID: PMC3988555 DOI: 10.1038/npp.2013.350] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2013] [Revised: 12/28/2013] [Accepted: 12/28/2013] [Indexed: 11/08/2022]
Abstract
Ingesting ethanol (EtOH) at low doses during social drinking is a common human behavior for its facilitating effects on social interactions. However, low-dose EtOH may have also detrimental effects that so far are underexplored. Here we sought to test the effects of low-dose EtOH on long-term potentiation (LTP)-like plasticity in human motor cortex. Previous cellular experiments showed that low-dose EtOH potentiates extrasynaptic GABAAR and reduces NMDAR-mediated currents, processes that would limit the expression of LTP. Paired associative transcranial magnetic stimulation (PASLTP) was employed in nine healthy subjects for induction of LTP-like plasticity, indexed by a long-term increase in motor-evoked potential input-output curves. Synaptic α1-GABAAR function was measured by saccadic peak velocity (SPV). Very low doses of EtOH (resulting in blood concentrations of <5 mM) suppressed LTP-like plasticity but did not affect SPV when compared with a placebo condition. In contrast, 1 mg of alprazolam, a classical benzodiazepine, or 10 mg of zolpidem, a non-benzodiazepine hypnotic, decreased SPV but did not significantly affect LTP-like plasticity when compared with placebo. This double dissociation of low-dose EtOH vs alprazolam/zolpidem effects is best explained by the putatively high affinity of EtOH but not alprazolam/zolpidem to extrasynaptic GABAARs and to NMDARs. Findings suggest that enhancement of extrasynaptic GABAAR-mediated tonic inhibition and/or reduction of NMDAR-mediated neurotransmission by EtOH blocks LTP-like plasticity in human cortex at very low doses that are easily reached during social drinking. Therefore, low-dose EtOH may jeopardize LTP-dependent processes, such as learning and memory formation.
Collapse
|
45
|
Tritsch NX, Oh WJ, Gu C, Sabatini BL. Midbrain dopamine neurons sustain inhibitory transmission using plasma membrane uptake of GABA, not synthesis. eLife 2014; 3:e01936. [PMID: 24843012 PMCID: PMC4001323 DOI: 10.7554/elife.01936] [Citation(s) in RCA: 138] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Synaptic transmission between midbrain dopamine neurons and target neurons in the striatum is essential for the selection and reinforcement of movements. Recent evidence indicates that nigrostriatal dopamine neurons inhibit striatal projection neurons by releasing a neurotransmitter that activates GABAA receptors. Here, we demonstrate that this phenomenon extends to mesolimbic afferents, and confirm that the released neurotransmitter is GABA. However, the GABA synthetic enzymes GAD65 and GAD67 are not detected in midbrain dopamine neurons. Instead, these cells express the membrane GABA transporters mGAT1 (Slc6a1) and mGAT4 (Slc6a11) and inhibition of these transporters prevents GABA co-release. These findings therefore indicate that GABA co-release is a general feature of midbrain dopaminergic neurons that relies on GABA uptake from the extracellular milieu as opposed to de novo synthesis. This atypical mechanism may confer dopaminergic neurons the flexibility to differentially control GABAergic transmission in a target-dependent manner across their extensive axonal arbors. DOI:http://dx.doi.org/10.7554/eLife.01936.001 The electrical signals that are fired along neurons cannot be transmitted across the small gaps, called synapses that are found between neurons. Instead, the neuron sending the signal releases chemicals called neurotransmitters into the synapse. These neurotransmitters bind to receptor proteins on the surface of the second neuron and control how it fires. A neurotransmitter called dopamine plays a key role in the circuits of the brain that control how we learn certain tasks involving movement. In particular, two populations of neurons from the midbrain that release dopamine target the striatum, an area of the brain that is responsible for motor control. These neurons also release other neurotransmitters, but the identity of these other chemicals is not known, and the details of the interaction between the neurons and the striatum are poorly understood. Previous research showed that some of the midbrain neurons activate receptors that normally respond to a neurotransmitter called gamma-aminobutyric acid (GABA). However, several different chemicals can trigger this receptor. Using a range of techniques, Tritsch et al. now confirm that dopamine neurons release GABA alongside dopamine, and that this applies to both sets of the dopamine-producing neurons that feed into the striatum. Some neurons can manufacture GABA from amino acids found in their internal fluid. However, Tritsch et al. could not detect the enzymes needed for this reaction in dopamine-producing neurons. Instead, these neurons contain proteins that can transport GABA across the cell membrane, which suggests that the neurons collect GABA from the extracellular fluid that surrounds them. DOI:http://dx.doi.org/10.7554/eLife.01936.002
Collapse
Affiliation(s)
- Nicolas X Tritsch
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, United States
| | - Won-Jong Oh
- Department of Neurobiology, Harvard Medical School, Boston, United States
| | - Chenghua Gu
- Department of Neurobiology, Harvard Medical School, Boston, United States
| | - Bernardo L Sabatini
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, United States
| |
Collapse
|
46
|
Maguire EP, Mitchell EA, Greig SJ, Corteen N, Balfour DJK, Swinny JD, Lambert JJ, Belelli D. Extrasynaptic glycine receptors of rodent dorsal raphe serotonergic neurons: a sensitive target for ethanol. Neuropsychopharmacology 2014; 39:1232-44. [PMID: 24264816 PMCID: PMC3957119 DOI: 10.1038/npp.2013.326] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Revised: 10/17/2013] [Accepted: 11/12/2013] [Indexed: 11/08/2022]
Abstract
Alcohol abuse is a significant medical and social problem. Several neurotransmitter systems are implicated in ethanol's actions, with certain receptors and ion channels emerging as putative targets. The dorsal raphe (DR) nucleus is associated with the behavioral actions of alcohol, but ethanol actions on these neurons are not well understood. Here, using immunohistochemistry and electrophysiology we characterize DR inhibitory transmission and its sensitivity to ethanol. DR neurons exhibit inhibitory 'phasic' post-synaptic currents mediated primarily by synaptic GABAA receptors (GABAAR) and, to a lesser extent, by synaptic glycine receptors (GlyR). In addition to such phasic transmission mediated by the vesicular release of neurotransmitter, the activity of certain neurons may be governed by a 'tonic' conductance resulting from ambient GABA activating extrasynaptic GABAARs. However, for DR neurons extrasynaptic GABAARs exert only a limited influence. By contrast, we report that unusually the GlyR antagonist strychnine reveals a large tonic conductance mediated by extrasynaptic GlyRs, which dominates DR inhibition. In agreement, for DR neurons strychnine increases their input resistance, induces membrane depolarization, and consequently augments their excitability. Importantly, this glycinergic conductance is greatly enhanced in a strychnine-sensitive fashion, by behaviorally relevant ethanol concentrations, by drugs used for the treatment of alcohol withdrawal, and by taurine, an ingredient of certain 'energy drinks' often imbibed with ethanol. These findings identify extrasynaptic GlyRs as critical regulators of DR excitability and a novel molecular target for ethanol.
Collapse
Affiliation(s)
- Edward P Maguire
- Division of Neuroscience, Medical Research Institute, Ninewells Hospital and Medical School, Dundee University, Dundee, UK
| | - Elizabeth A Mitchell
- Division of Neuroscience, Medical Research Institute, Ninewells Hospital and Medical School, Dundee University, Dundee, UK
| | - Scott J Greig
- Division of Neuroscience, Medical Research Institute, Ninewells Hospital and Medical School, Dundee University, Dundee, UK
| | - Nicole Corteen
- Institute for Biomedical and Biomolecular Sciences, School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK
| | - David J K Balfour
- Division of Neuroscience, Medical Research Institute, Ninewells Hospital and Medical School, Dundee University, Dundee, UK
| | - Jerome D Swinny
- Institute for Biomedical and Biomolecular Sciences, School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK
| | - Jeremy J Lambert
- Division of Neuroscience, Medical Research Institute, Ninewells Hospital and Medical School, Dundee University, Dundee, UK
| | - Delia Belelli
- Division of Neuroscience, Medical Research Institute, Ninewells Hospital and Medical School, Dundee University, Dundee, UK
| |
Collapse
|
47
|
Kim A, Zamora-Martinez ER, Edwards S, Mandyam CD. Structural reorganization of pyramidal neurons in the medial prefrontal cortex of alcohol dependent rats is associated with altered glial plasticity. Brain Struct Funct 2014; 220:1705-20. [PMID: 24667898 PMCID: PMC4177030 DOI: 10.1007/s00429-014-0755-3] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Accepted: 03/10/2014] [Indexed: 12/22/2022]
Abstract
In rodents, chronic intermittent ethanol vapor exposure (CIE) produces alcohol dependence, alters the activity of pyramidal neurons and decreases the number of glial progenitors in the medial prefrontal cortex (mPFC). Adult male Wistar rats were exposed to CIE and were injected with mitotic markers to label and phenotype proliferating cells to test the hypothesis that CIE produces concurrent alterations in the structure of pyramidal neurons and the cell cycle kinetics and developmental stages of glial progenitors in the mPFC. Medial prefrontal cortical tissue was processed for Golgi-Cox staining, immunohistochemistry and Western blotting analysis. CIE increased dendritic arborization and spine densities within basal and apical dendrites of pyramidal neurons via aberrant reorganization of actin cytoskeleton-associated molecules. CIE concomitantly increased the expression of total NR2B subunits without affecting phosphorylation of NR2B at Tyr-1472 or levels of PSD-95. CIE reduced the length of S-phase of the cell cycle of glial progenitors and reduced proliferation and differentiation of progenitors into bHLH transcription factor Olig2-expressing premyelinating oligodendrocyte progenitor cells (OPCs). CIE also produced a corresponding hyperphosphorylation of Olig2, and reduced expression of myelin basic protein. Our findings demonstrate that CIE-induced alterations in OPCs and myelin-related proteins are associated with profound alterations in the structure of pyramidal neurons. In sum, our results not only provide evidence that alcohol dependence leads to pathological changes in the mPFC, which may in part define a cellular basis for cognitive impairments associated with alcoholism, but also show dependence-associated morphological changes in the PFC at the single neuron level.
Collapse
Affiliation(s)
- Airee Kim
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
| | - Eva R. Zamora-Martinez
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
| | - Scott Edwards
- Department of Physiology, Alcohol & Drug Abuse Center of Excellence, LSU Health Sciences Center, New Orleans, LA, USA
| | - Chitra D. Mandyam
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
- Skaggs School of Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
48
|
Ethanol reduces neuronal excitability of lateral orbitofrontal cortex neurons via a glycine receptor dependent mechanism. Neuropsychopharmacology 2013; 38:1176-88. [PMID: 23314219 PMCID: PMC3656360 DOI: 10.1038/npp.2013.12] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Trauma-induced damage to the orbitofrontal cortex (OFC) often results in behavioral inflexibility and impaired judgment. Human alcoholics exhibit similar cognitive deficits suggesting that OFC neurons are susceptible to alcohol-induced dysfunction. A previous study from this laboratory examined OFC mediated cognitive behaviors in mice and showed that behavioral flexibility during a reversal learning discrimination task was reduced in alcohol-dependent mice. Despite these intriguing findings, the actions of alcohol on OFC neuron function are unknown. To address this issue, slices containing the lateral OFC (lOFC) were prepared from adult C57BL/6J mice and whole-cell patch clamp electrophysiology was used to characterize the effects of ethanol (EtOH) on neuronal function. EtOH (66 mM) had no effect on AMPA-mediated EPSCs but decreased those mediated by NMDA receptors. EtOH (11-66 mM) also decreased current-evoked spike firing and this was accompanied by a decrease in input resistance and a modest hyperpolarization. EtOH inhibition of spike firing was prevented by the GABAA antagonist picrotoxin, but EtOH had no effect on evoked or spontaneous GABA IPSCs. EtOH increased the holding current of voltage-clamped neurons and this action was blocked by picrotoxin but not the more selective GABAA antagonist biccuculine. The glycine receptor antagonist strychnine also prevented EtOH's effect on holding current and spike firing, and western blotting revealed the presence of glycine receptors in lOFC. Overall, these results suggest that acutely, EtOH may reduce lOFC function via a glycine receptor dependent process and this may trigger neuroadaptive mechanisms that contribute to the impairment of OFC-dependent behaviors in alcohol-dependent subjects.
Collapse
|
49
|
Maternal separation and proclivity for ethanol intake: A potential role of the endocannabinoid system in rats. Neuroscience 2012; 223:296-304. [DOI: 10.1016/j.neuroscience.2012.07.071] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Revised: 07/30/2012] [Accepted: 07/31/2012] [Indexed: 12/31/2022]
|
50
|
Badanich KA, Becker HC, Woodward JJ. Effects of chronic intermittent ethanol exposure on orbitofrontal and medial prefrontal cortex-dependent behaviors in mice. Behav Neurosci 2012; 125:879-91. [PMID: 22122149 DOI: 10.1037/a0025922] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
In humans, stroke or trauma-induced damage to the orbitofrontal cortex (OFC) or medial prefrontal cortex (mPFC) results in impaired cognitive flexibility. Alcoholics also exhibit similar deficits in cognitive flexibility, suggesting that the OFC and mPFC are susceptible to alcohol-induced dysfunction. The present experiments investigated this issue using an attention set-shifting assay in ethanol dependent adult male C57BL/6J mice. Ethanol dependence was induced by exposing mice to repeated cycles of chronic intermittent ethanol (CIE) vapor inhalation. Behavioral testing was conducted 72 hours or 10 days following CIE exposure to determine whether ethanol-induced changes in OFC-dependent (reversal learning) and mPFC-dependent (set-shifting) behaviors are long lasting. During early ethanol abstinence (72 hrs), CIE mice showed reduced reversal learning performance as compared to controls. Reversal learning deficits were revealed as greater number of trials to criterion, more errors made, and a greater difficulty in performing a reversal learning task relative to baseline performance. Furthermore, the magnitude of the impairment was greater during reversal of a simple discrimination rather than reversal of an intra-dimensional shift. Reversal learning deficits were no longer present when mice were tested 10 days after CIE exposure, suggesting that ethanol-induced changes in OFC function can recover. Unexpectedly, performance on the set-shifting task was not impaired during abstinence from ethanol. These data suggest reversal learning, but not attention set-shifting, is transiently disrupted during short-term abstinence from CIE. Given that reversal learning requires an intact OFC, these findings support the idea that the OFC may be vulnerable to the cognitive impairing actions of ethanol.
Collapse
Affiliation(s)
- Kimberly A Badanich
- Center for Drug and Alcohol Programs, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC 29425, USA.
| | | | | |
Collapse
|