1
|
Titterness AK, Gräfe EL, Acosta C, Rodriguez C, Thomas JD, Christie BR. Developmental ethanol exposure produces deficits in long-term potentiation in vivo that persist following postnatal choline supplementation. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2024; 48:1483-1491. [PMID: 38850072 DOI: 10.1111/acer.15384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 04/18/2024] [Accepted: 05/20/2024] [Indexed: 06/09/2024]
Abstract
BACKGROUND Fetal alcohol spectrum disorder (FASD) is one of the leading causes of neurodevelopmental disorder for which there is a pressing need for an effective treatment. Recent studies have investigated the essential nutrient choline as a postnatal treatment option. Supplementation with choline has produced improvements in behavioral tasks related to learning and memory and reverted changes in methylation signature following third-trimester equivalent ethanol exposure. We examined whether there are related improvements in hippocampal synaptic plasticity in vivo. METHODS Sprague-Dawley offspring were administered binge-levels of ethanol from postnatal day (PND) 4 to 9, then treated with choline chloride (100 mg/kg/day) from PND 10 to 30. In vivo electrophysiology was performed on male and female offspring from PND 55 to 70. Long-term potentiation (LTP) was induced in the medial perforant pathway of the dentate gyrus using a theta-burst stimulation (TBS) protocol, and field-evoked postsynaptic potentials (EPSPs) were evoked for 60 min following the conditioning stimulus. RESULTS Developmental ethanol exposure caused long-lasting deficits in LTP of the slope of the evoked responses and in the amplitude of the population spike potentiation. Neither deficit was rescued by postnatal choline supplementation. CONCLUSIONS In contrast to our prior findings that choline can improve hippocampal plasticity (Nutrients, 2022, 14, 2004), here we found that deficits in hippocampal synaptic plasticity due to developmental ethanol exposure persisted into adulthood despite adolescent choline supplementation. Future research should examine more subtle changes in synaptic plasticity to identify synaptic changes that mirror behavioral improvements.
Collapse
Affiliation(s)
- A K Titterness
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - E L Gräfe
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - C Acosta
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - C Rodriguez
- Department of Psychology, San Diego State University, San Diego, California, USA
| | - J D Thomas
- Department of Psychology, San Diego State University, San Diego, California, USA
| | - B R Christie
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
- Department of Psychology, San Diego State University, San Diego, California, USA
- Island Medical Program and Department of Cellular and Physiological Sciences, University of British Columbia, Victoria, British Columbia, Canada
- Institute for Aging and Life-Long Health, University of Victoria, Victoria, British Columbia, Canada
| |
Collapse
|
2
|
Giri T, Maloney SE, Giri S, Goo YA, Song JH, Son M, Tycksen E, Conyers SB, Bice A, Ge X, Garbow JR, Quirk JD, Bauer AQ, Palanisamy A. Oxytocin-induced birth causes sex-specific behavioral and brain connectivity changes in developing rat offspring. iScience 2024; 27:108960. [PMID: 38327784 PMCID: PMC10847747 DOI: 10.1016/j.isci.2024.108960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 11/23/2023] [Accepted: 01/15/2024] [Indexed: 02/09/2024] Open
Abstract
Despite six decades of the use of exogenous oxytocin for management of labor, little is known about its effects on the developing brain. Motivated by controversial reports suggesting a link between oxytocin use during labor and autism spectrum disorders (ASDs), we employed our recently validated rat model for labor induction with oxytocin to address this important concern. Using a combination of molecular biological, behavioral, and neuroimaging assays, we show that induced birth with oxytocin leads to sex-specific disruption of oxytocinergic signaling in the developing brain, decreased communicative ability of pups, reduced empathy-like behaviors especially in male offspring, and widespread sex-dependent changes in functional cortical connectivity. Contrary to our hypothesis, social behavior, typically impaired in ASDs, was largely preserved. Collectively, our foundational studies provide nuanced insights into the neurodevelopmental impact of birth induction with oxytocin and set the stage for mechanistic investigations in animal models and prospective longitudinal clinical studies.
Collapse
Affiliation(s)
- Tusar Giri
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Susan E. Maloney
- Department of Psychiatry, Intellectual and Developmental Disabilities Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Saswat Giri
- Graduate Student, School of Public Health and Social Justice, St. Louis University, St. Louis, MO, USA
| | - Young Ah Goo
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, USA
- Mass Spectrometry Technology Access Center (MTAC), McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, USA
| | - Jong Hee Song
- Mass Spectrometry Technology Access Center (MTAC), McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, USA
| | - Minsoo Son
- Mass Spectrometry Technology Access Center (MTAC), McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, USA
| | - Eric Tycksen
- Genome Technology Access Center (GTAC), McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, USA
| | - Sara B. Conyers
- Department of Psychiatry, Intellectual and Developmental Disabilities Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Annie Bice
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Xia Ge
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Joel R. Garbow
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - James D. Quirk
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Adam Q. Bauer
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Arvind Palanisamy
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
3
|
Shah P, Kaneria A, Fleming G, Williams CRO, Sullivan RM, Lemon CH, Smiley J, Saito M, Wilson DA. Homeostatic NREM sleep and salience network function in adult mice exposed to ethanol during development. Front Neurosci 2023; 17:1267542. [PMID: 38033546 PMCID: PMC10682725 DOI: 10.3389/fnins.2023.1267542] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 10/30/2023] [Indexed: 12/02/2023] Open
Abstract
Developmental exposure to ethanol is a leading cause of cognitive, emotional and behavioral problems, with fetal alcohol spectrum disorder (FASD) affecting more than 1:100 children. Recently, comorbid sleep deficits have been highlighted in these disorders, with sleep repair a potential therapeutic target. Animal models of FASD have shown non-REM (NREM) sleep fragmentation and slow-wave oscillation impairments that predict cognitive performance. Here we use a mouse model of perinatal ethanol exposure to explore whether reduced sleep pressure may contribute to impaired NREM sleep, and compare the function of a brain network reported to be impacted by insomnia-the Salience network-in developmental ethanol-exposed mice with sleep-deprived, saline controls. Mice were exposed to ethanol or saline on postnatal day 7 (P7) and allowed to mature to adulthood for testing. At P90, telemetered cortical recordings were made for assessment of NREM sleep in home cage before and after 4 h of sleep deprivation to assess basal NREM sleep and homeostatic NREM sleep response. To assess Salience network functional connectivity, mice were exposed to the 4 h sleep deprivation period or left alone, then immediately sacrificed for immunohistochemical analysis of c-Fos expression. The results show that developmental ethanol severely impairs both normal rebound NREM sleep and sleep deprivation induced increases in slow-wave activity, consistent with reduced sleep pressure. Furthermore, the Salience network connectome in rested, ethanol-exposed mice was most similar to that of sleep-deprived, saline control mice, suggesting a sleep deprivation-like state of Salience network function after developmental ethanol even without sleep deprivation.
Collapse
Affiliation(s)
- Prachi Shah
- Emotional Brain Institute, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY,United States
| | - Aayush Kaneria
- Emotional Brain Institute, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY,United States
| | - Gloria Fleming
- Emotional Brain Institute, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY,United States
| | - Colin R. O. Williams
- Emotional Brain Institute, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY,United States
| | - Regina M. Sullivan
- Emotional Brain Institute, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY,United States
- School of Biological Sciences, University of Oklahoma, Norman, OK, United States
- Department of Child and Adolescent Psychiatry, NYU School of Medicine, New York, NY, United States
| | - Christian H. Lemon
- School of Biological Sciences, University of Oklahoma, Norman, OK, United States
| | - John Smiley
- Division of Neurochemistry, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY,United States
- Department of Psychiatry, New York University Medical Center, New York, NY,United States
| | - Mariko Saito
- Division of Neurochemistry, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY,United States
- Department of Psychiatry, New York University Medical Center, New York, NY,United States
| | - Donald A. Wilson
- Emotional Brain Institute, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY,United States
- School of Biological Sciences, University of Oklahoma, Norman, OK, United States
- Department of Child and Adolescent Psychiatry, NYU School of Medicine, New York, NY, United States
| |
Collapse
|
4
|
Basavarajappa BS, Subbanna S. Synaptic Plasticity Abnormalities in Fetal Alcohol Spectrum Disorders. Cells 2023; 12:442. [PMID: 36766783 PMCID: PMC9913617 DOI: 10.3390/cells12030442] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/10/2023] [Accepted: 01/27/2023] [Indexed: 01/31/2023] Open
Abstract
The brain's ability to strengthen or weaken synaptic connections is often termed synaptic plasticity. It has been shown to function in brain remodeling following different types of brain damage (e.g., drugs of abuse, alcohol use disorders, neurodegenerative diseases, and inflammatory conditions). Although synaptic plasticity mechanisms have been extensively studied, how neural plasticity can influence neurobehavioral abnormalities in alcohol use disorders (AUDs) is far from being completely understood. Alcohol use during pregnancy and its harmful effects on the developing offspring are major public health, social, and economic challenges. The significant attribute of prenatal alcohol exposure on offspring is damage to the central nervous system (CNS), causing a range of synaptic structural, functional, and behavioral impairments, collectively called fetal alcohol spectrum disorder (FASD). Although the synaptic mechanisms in FASD are limited, emerging evidence suggests that FASD pathogenesis involves altering a set of molecules involved in neurotransmission, myelination, and neuroinflammation. These studies identify several immediate and long-lasting changes using many molecular approaches that are essential for synaptic plasticity and cognitive function. Therefore, they can offer potential synaptic targets for the many neurobehavioral abnormalities observed in FASD. In this review, we discuss the substantial research progress in different aspects of synaptic and molecular changes that can shed light on the mechanism of synaptic dysfunction in FASD. Increasing our understanding of the synaptic changes in FASD will significantly advance our knowledge and could provide a basis for finding novel therapeutic targets and innovative treatment strategies.
Collapse
Affiliation(s)
- Balapal S. Basavarajappa
- Center for Dementia Research, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, NY 10032, USA
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY 10032, USA
- Department of Psychiatry, New York University Langone Medical Center, New York, NY 10016, USA
| | - Shivakumar Subbanna
- Center for Dementia Research, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA
| |
Collapse
|
5
|
Imbriani P, Sciamanna G, El Atiallah I, Cerri S, Hess EJ, Pisani A. Synaptic effects of ethanol on striatal circuitry: therapeutic implications for dystonia. FEBS J 2022; 289:5834-5849. [PMID: 34217152 PMCID: PMC9786552 DOI: 10.1111/febs.16106] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 05/21/2021] [Accepted: 07/02/2021] [Indexed: 12/30/2022]
Abstract
Alcohol consumption affects motor behavior and motor control. Both acute and chronic alcohol abuse have been extensively investigated; however, the therapeutic efficacy of alcohol on some movement disorders, such as myoclonus-dystonia or essential tremor, still does not have a plausible mechanistic explanation. Yet, there are surprisingly few systematic trials with known GABAergic drugs mimicking the effect of alcohol on neurotransmission. In this brief survey, we aim to summarize the effects of EtOH on striatal function, providing an overview of its cellular and synaptic actions in a 'circuit-centered' view. In addition, we will review both experimental and clinical evidence, in the attempt to provide a plausible mechanistic explanation for alcohol-responsive movement disorders, with particular emphasis on dystonia. Different hypotheses emerge, which may provide a rationale for the utilization of drugs that mimic alcohol effects, predicting potential drug repositioning.
Collapse
Affiliation(s)
- Paola Imbriani
- Department of Systems MedicineUniversity of Rome ‘Tor Vergata’Italy,IRCCS Fondazione Santa LuciaRomeItaly
| | - Giuseppe Sciamanna
- Department of Systems MedicineUniversity of Rome ‘Tor Vergata’Italy,IRCCS Fondazione Santa LuciaRomeItaly
| | - Ilham El Atiallah
- Department of Systems MedicineUniversity of Rome ‘Tor Vergata’Italy,IRCCS Fondazione Santa LuciaRomeItaly
| | | | - Ellen J. Hess
- Departments of Pharmacology and Chemical Biology and NeurologyEmory UniversityAtlantaGAUSA
| | - Antonio Pisani
- IRCCS Mondino FoundationPaviaItaly,Department of Brain and Behavioral SciencesUniversity of PaviaItaly
| |
Collapse
|
6
|
Deschamps C, Uyttersprot F, Debris M, Marié C, Fouquet G, Marcq I, Vilpoux C, Naassila M, Pierrefiche O. Anti-inflammatory drugs prevent memory and hippocampal plasticity deficits following initial binge-like alcohol exposure in adolescent male rats. Psychopharmacology (Berl) 2022; 239:2245-2262. [PMID: 35314896 DOI: 10.1007/s00213-022-06112-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 03/01/2022] [Indexed: 11/30/2022]
Abstract
RATIONALE Binge drinking during adolescence impairs learning and memory on the long term, and many studies suggest a role of neuroinflammation. However, whether neuroinflammation occurs after the very first exposures to alcohol remains unclear, while initial alcohol exposure impairs learning for several days in male rats. OBJECTIVES To investigate the role of neuroinflammation in the effects of only two binge-like episodes on learning and on neuronal plasticity in adolescent male rat hippocampus. METHODS Animals received two ethanol i.p. injections (3 g/kg) 9 h apart. Forty-eight hours later, we recorded long-term depression (LTD) and potentiation (LTP) in CA1 area of hippocampus slices. In isolated CA1, we measured immunolabelings for microglial activation and Toll-like receptor 4 (TLR4) and mRNA levels for several cytokines. RESULTS Forty-eight hours after the two binges, rats performed worse than control rats in novel object recognition, LTD was reduced, LTP was increased, and excitatory neurotransmission was more sensitive to an antagonist of the GluN2B subunit of the NMDA receptor. Exposure to ethanol with minocycline or indomethacin, two anti-inflammatory drugs, or with a TLR4 antagonist, prevented all effects of ethanol. Immunolabelings at 48 h showed a reduction of neuronal TLR4 that was prevented by minocycline pretreatment, while microglial reactivity was undetected and inflammatory cytokines mRNA levels were unchanged. CONCLUSION Two binge-like ethanol exposures during adolescence in rat involved neuroinflammation leading to changes in TLR4 expression and in GluN2B functioning inducing disturbances in synaptic plasticity and cognitive deficits. Anti-inflammatory drugs are good candidates to prevent brain function and memory deficits induced by few binge-drinking episodes.
Collapse
Affiliation(s)
- Chloé Deschamps
- UMR1247 INSERM, Groupe de Recherche Sur L'Alcool Et Les Pharmacodépendances, Université de Picardie Jules Verne, Centre Universitaire de Recherche en Santé, Chemin du Thil, 80025, Amiens, France
| | - Floriane Uyttersprot
- UMR1247 INSERM, Groupe de Recherche Sur L'Alcool Et Les Pharmacodépendances, Université de Picardie Jules Verne, Centre Universitaire de Recherche en Santé, Chemin du Thil, 80025, Amiens, France
| | - Margot Debris
- UMR1247 INSERM, Groupe de Recherche Sur L'Alcool Et Les Pharmacodépendances, Université de Picardie Jules Verne, Centre Universitaire de Recherche en Santé, Chemin du Thil, 80025, Amiens, France
| | - Constance Marié
- UMR1247 INSERM, Groupe de Recherche Sur L'Alcool Et Les Pharmacodépendances, Université de Picardie Jules Verne, Centre Universitaire de Recherche en Santé, Chemin du Thil, 80025, Amiens, France
| | - Grégory Fouquet
- UMR1247 INSERM, Groupe de Recherche Sur L'Alcool Et Les Pharmacodépendances, Université de Picardie Jules Verne, Centre Universitaire de Recherche en Santé, Chemin du Thil, 80025, Amiens, France
| | - Ingrid Marcq
- UMR1247 INSERM, Groupe de Recherche Sur L'Alcool Et Les Pharmacodépendances, Université de Picardie Jules Verne, Centre Universitaire de Recherche en Santé, Chemin du Thil, 80025, Amiens, France
| | - Catherine Vilpoux
- UMR1247 INSERM, Groupe de Recherche Sur L'Alcool Et Les Pharmacodépendances, Université de Picardie Jules Verne, Centre Universitaire de Recherche en Santé, Chemin du Thil, 80025, Amiens, France
| | - Mickael Naassila
- UMR1247 INSERM, Groupe de Recherche Sur L'Alcool Et Les Pharmacodépendances, Université de Picardie Jules Verne, Centre Universitaire de Recherche en Santé, Chemin du Thil, 80025, Amiens, France
| | - Olivier Pierrefiche
- UMR1247 INSERM, Groupe de Recherche Sur L'Alcool Et Les Pharmacodépendances, Université de Picardie Jules Verne, Centre Universitaire de Recherche en Santé, Chemin du Thil, 80025, Amiens, France.
| |
Collapse
|
7
|
Madaan P, Behl T, Sehgal A, Singh S, Sharma N, Yadav S, Kaur S, Bhatia S, Al-Harrasi A, Abdellatif AAH, Ashraf GM, Abdel-Daim MM, Dailah HG, Anwer MK, Bungau S. Exploring the Therapeutic Potential of Targeting Purinergic and Orexinergic Receptors in Alcoholic Neuropathy. Neurotox Res 2022; 40:646-669. [DOI: 10.1007/s12640-022-00477-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 01/13/2022] [Accepted: 01/19/2022] [Indexed: 12/11/2022]
|
8
|
Baradaran Z, Vakilian A, Zare M, Hashemzehi M, Hosseini M, Dinpanah H, Beheshti F. Metformin improved memory impairment caused by chronic ethanol consumption during adolescent to adult period of rats: Role of oxidative stress and neuroinflammation. Behav Brain Res 2021; 411:113399. [PMID: 34087254 DOI: 10.1016/j.bbr.2021.113399] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 05/22/2021] [Accepted: 05/30/2021] [Indexed: 01/06/2023]
Abstract
OBJECTIVE Adolescence is a crucial time for brain maturation. We investigated the protective effects of metformin (Met) on behavioral changes, oxidative stress, tumor necrosis factor alpha (TNF-α) and nitrite in adulthood induced by ethanol (Eth) consumption during adolescent to adult period of rats. MATERIALS AND METHODS The adolescence male rats (21 days old) were treated as: 1) Control, 2) Eth (Eth in drinking water (20 %)), 3-5) Eth-Met50, 100 and 150 mg/kg (Eth in drinking water and Met (50, 100, or 150 mg/kg). After 5 weeks treatment, Morris water maze (MMW) and passive avoidance (PA) tests were done. RESULTS The latency in the MWM test was higher and the latency to enter the dark chamber in the PA test was lower in the Eth group than in control. In Eth-Met100 and 150 groups, they were less than the Eth group. Malondialdehyde (MDA) and nitrite concentration in the hippocampus and cortex of the Eth group were higher than the control group. The thiol content and catalase and superoxide dismutase (SOD) activities in hippocampal and cortical tissues of the Eth group reduced compared to the control group. TNF-α was higher in hippocampal tissues of Eth group animals. Met reversed all of these effects. CONCLUSION Our findings showed that the protective effects of Met against chronic Eth consumption induced learning and memory impairment were accompanied by decreasing of TNF-a, nitrite and oxidative stress in adolescent rats.
Collapse
Affiliation(s)
- Zahra Baradaran
- Student Research Committee, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran; Neuroscience Research Center, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran; Departments of Physiology, School of Paramedical Sciences, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| | - Arefeh Vakilian
- Student Research Committee, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran; Neuroscience Research Center, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran; Departments of Physiology, School of Paramedical Sciences, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| | - Mostafa Zare
- Student Research Committee, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran; Neuroscience Research Center, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran; Departments of Physiology, School of Paramedical Sciences, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| | - Milad Hashemzehi
- Tropical and Communicable Diseases Research Centre, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | - Mahmoud Hosseini
- Division of Neurocognitive Sciences, Psychiatry and Behavioral Sciences Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Dinpanah
- Department of Emergency Medicine, 9-Day Hospital, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| | - Farimah Beheshti
- Neuroscience Research Center, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran; Departments of Physiology, School of Paramedical Sciences, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran.
| |
Collapse
|
9
|
Izumi Y, Mennerick SJ, Doherty JJ, Zorumski CF. Oxysterols Modulate the Acute Effects of Ethanol on Hippocampal N-Methyl-d-Aspartate Receptors, Long-Term Potentiation, and Learning. J Pharmacol Exp Ther 2021; 377:181-188. [PMID: 33441369 PMCID: PMC8051516 DOI: 10.1124/jpet.120.000376] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 01/08/2021] [Indexed: 12/29/2022] Open
Abstract
Ethanol is a noncompetitive inhibitor of N-methyl-d-aspartate receptors (NMDARs) and acutely disrupts hippocampal synaptic plasticity and learning. In the present study, we examined the effects of oxysterol positive allosteric modulators (PAMs) of NMDARs on ethanol-mediated inhibition of NMDARs, block of long-term potentiation (LTP) and long-term depression (LTD) in rat hippocampal slices, and defects in one-trial learning in vivo. We found that 24S-hydroxycholesterol and a synthetic oxysterol analog, SGE-301, overcame effects of ethanol on NMDAR-mediated synaptic responses in the CA1 region but did not alter acute effects of ethanol on LTD; the synthetic oxysterol, however, overcame acute inhibition of LTP. In addition, both oxysterols overcame persistent effects of ethanol on LTP in vitro, and the synthetic analog reversed defects in one-trial inhibitory avoidance learning in vivo. These results indicate that effects of ethanol on both LTP and LTD arise by complex mechanisms beyond NMDAR antagonism and that oxysterol NMDAR PAMS may represent a novel approach for preventing and reversing acute ethanol-mediated changes in cognition. SIGNIFICANCE STATEMENT: Ethanol acutely inhibits hippocampal NMDARs, LTP, and learning. This study found that certain oxysterols that are NMDAR-positive allosteric modulators can overcome the acute effects of ethanol on NMDARs, LTP, and learning. Oxysterols differ in their effects from agents that inhibit integrated cellular stress responses.
Collapse
Affiliation(s)
- Yukitoshi Izumi
- Department of Psychiatry and Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, Missouri (Y.I., S.J.M., C.F.Z.); and Sage Therapeutics, Cambridge, Massachusetts (J.J.D.)
| | - Steven J Mennerick
- Department of Psychiatry and Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, Missouri (Y.I., S.J.M., C.F.Z.); and Sage Therapeutics, Cambridge, Massachusetts (J.J.D.)
| | - James J Doherty
- Department of Psychiatry and Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, Missouri (Y.I., S.J.M., C.F.Z.); and Sage Therapeutics, Cambridge, Massachusetts (J.J.D.)
| | - Charles F Zorumski
- Department of Psychiatry and Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, Missouri (Y.I., S.J.M., C.F.Z.); and Sage Therapeutics, Cambridge, Massachusetts (J.J.D.)
| |
Collapse
|
10
|
Kazemi T, Huang S, Avci NG, Waits CMK, Akay YM, Akay M. Investigating the influence of perinatal nicotine and alcohol exposure on the genetic profiles of dopaminergic neurons in the VTA using miRNA-mRNA analysis. Sci Rep 2020; 10:15016. [PMID: 32929144 PMCID: PMC7490691 DOI: 10.1038/s41598-020-71875-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 08/04/2020] [Indexed: 12/11/2022] Open
Abstract
Nicotine and alcohol are two of the most commonly used and abused recreational drugs, are often used simultaneously, and have been linked to significant health hazards. Furthermore, patients diagnosed with dependence on one drug are highly likely to be dependent on the other. Several studies have shown the effects of each drug independently on gene expression within many brain regions, including the ventral tegmental area (VTA). Dopaminergic (DA) neurons of the dopamine reward pathway originate from the VTA, which is believed to be central to the mechanism of addiction and drug reinforcement. Using a well-established rat model for both nicotine and alcohol perinatal exposure, we investigated miRNA and mRNA expression of dopaminergic (DA) neurons of the VTA in rat pups following perinatal alcohol and joint nicotine-alcohol exposure. Microarray analysis was then used to profile the differential expression of both miRNAs and mRNAs from DA neurons of each treatment group to further explore the altered genes and related biological pathways modulated. Predicted and validated miRNA-gene target pairs were analyzed to further understand the roles of miRNAs within these networks following each treatment, along with their post transcription regulation points affecting gene expression throughout development. This study suggested that glutamatergic synapse and axon guidance pathways were specifically enriched and many miRNAs and genes were significantly altered following alcohol or nicotine-alcohol perinatal exposure when compared to saline control. These results provide more detailed insight into the cell proliferation, neuronal migration, neuronal axon guidance during the infancy in rats in response to perinatal alcohol/ or nicotine-alcohol exposure.
Collapse
Affiliation(s)
- Tina Kazemi
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | - Shuyan Huang
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | - Naze G Avci
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | - Charlotte Mae K Waits
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | - Yasemin M Akay
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | - Metin Akay
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA.
| |
Collapse
|
11
|
Boroda E, Krueger AM, Bansal P, Schumacher MJ, Roy AV, Boys CJ, Lim KO, Wozniak JR. A randomized controlled trial of transcranial direct-current stimulation and cognitive training in children with fetal alcohol spectrum disorder. Brain Stimul 2020; 13:1059-1068. [PMID: 32360392 DOI: 10.1016/j.brs.2020.04.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 04/20/2020] [Accepted: 04/22/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND This study was a randomized double-blind sham-controlled trial examining the effects of transcranial direct current stimulation (tDCS) augmented cognitive training (CT) in children with Fetal Alcohol Spectrum Disorders (FASD). Prenatal alcohol exposure has profound detrimental effects on brain development and individuals with FASD commonly present with deficits in executive functions including attention and working memory. The most commonly studied treatment for executive deficits is CT, which involves repeated drilling of exercises targeting the impaired functions. As currently implemented, CT requires many hours and the observed effect sizes are moderate. Neuromodulation via tDCS can enhance brain plasticity and prior studies demonstrate that combining tDCS with CT improves efficacy and functional outcomes. TDCS-augmented CT has not yet been tested in FASD, a condition in which there are known abnormalities in neuroplasticity and few interventions. METHODS This study examined the feasibility and efficacy of this approach in 44 children with FASD. Participants were randomized to receive five sessions of CT with either active or sham tDCS targeting the dorsolateral prefrontal cortex, a region of the brain that is heavily involved in executive functioning. RESULTS The intervention was feasible and well-tolerated in children with FASD. The tDCS group showed nominally significant improvement in attention on a continuous performance test compared to sham (p = .043). Group differences were observed at the third, fourth and fifth treatment sessions. There was no effect of tDCS on working memory (p = .911). Further, we found no group differences on a trail making task (p = .659) or on the verbal fluency test (p = .826). In the active tDCS group, a significant correlation was observed between improvement in attention scores and decrease in parent-reported attention deficits (p = .010). CONCLUSIONS These results demonstrate that tDCS-augmented CT is well tolerated in children with FASD and potentially offers benefits over and above CT alone.
Collapse
|
12
|
Inhibitors of cellular stress overcome acute effects of ethanol on hippocampal plasticity and learning. Neurobiol Dis 2020; 141:104875. [PMID: 32334031 DOI: 10.1016/j.nbd.2020.104875] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 04/03/2020] [Accepted: 04/20/2020] [Indexed: 01/04/2023] Open
Abstract
Ethanol intoxication can produce marked changes in cognitive function including states in which the ability to learn and remember new information is completely disrupted. These defects likely reflect changes in the synaptic plasticity thought to underlie memory formation. We have studied mechanisms contributing to the adverse effects of ethanol on hippocampal long-term potentiation (LTP) and provided evidence that ethanol-mediated LTP inhibition involves a form of metaplasticity resulting from local metabolism of ethanol to acetaldehyde and untimely activation of N-methyl-d-aspartate receptors (NMDARs), both of which are neuronal stressors. In the present studies, we sought to understand the role of cellular stress in LTP defects, and demonstrate that ethanol's effects on LTP in the CA1 hippocampal region are overcome by agents that inhibit cellular stress responses, including ISRIB, a specific inhibitor of integrated stress responses, and GW3965, an agonist that acts at liver X receptors (LXRs) and dampens cellular stress. The agents that alter LTP inhibition also prevent the adverse effects of acute ethanol on one trial inhibitory avoidance learning. Unexpectedly, we found that the LXR agonist but not ISRIB overcomes effects of ethanol on synaptic responses mediated by N-methyl-d-aspartate receptors (NMDARs). These results have implications for understanding the adverse effects of ethanol and possibly for identifying novel paths to treatments that can prevent or overcome ethanol-induced cognitive dysfunction.
Collapse
|
13
|
Plaza-Briceño W, Estay SF, de la Fuente-Ortega E, Gutiérrez C, Sánchez G, Hidalgo C, Chávez AE, Haeger PA. N-Methyl-d-Aspartate Receptor Modulation by Nicotinamide Adenine Dinucleotide Phosphate Oxidase Type 2 Drives Synaptic Plasticity and Spatial Memory Impairments in Rats Exposed Pre- and Postnatally to Ethanol. Antioxid Redox Signal 2020; 32:602-617. [PMID: 31880947 DOI: 10.1089/ars.2019.7787] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Aims: Pre- and/or early postnatal ethanol exposure (prenatal alcohol exposure [PAE]) impairs synaptic plasticity as well as memory formation, but the mechanisms underlying these effects remain unclear. Both long-term potentiation (LTP) and spatial memory formation in the hippocampus involve the nicotinamide adenine dinucleotide phosphate oxidase type 2 (NOX2) enzyme. Previous studies have reported that N-methyl-d-aspartate receptor (NMDAR) activation increases NOX2-mediated superoxide generation, resulting in inhibition of NMDAR function, but whether NOX2 impacts NMDAR function in PAE animals leading to impaired LTP and memory formation remains unknown. We aim to evaluate whether the NOX2-NMDAR complex is involved in the long-lasting deleterious effects of PAE on hippocampal LTP and memory formation. Results: Here we provide novel evidence that PAE animals display impaired NMDAR-dependent LTP in the cornus ammonis field 1 (CA1) and NMDAR-mediated LTP in the dentate gyrus (DG). Moreover, PAE rats displayed increased NMDAR-mediated transmission in both hippocampal areas. Interestingly, NOX2 pharmacological inhibition restored NMDAR-mediated transmission and LTP in the CA1, but not in the DG. PAE also induced overexpression of NOX2 and CaMKII isoforms, but did not modify the content or the redox state of the N-methyl-d-aspartate receptor subunit-1 (NR1) subunit of NMDAR in both areas of the hippocampus. In addition, adolescent PAE rats orally fed the antioxidant and free radical scavenger apocynin exhibited significantly improved spatial memory acquisition. Innovation and Conclusion: By showing in PAE animals NOX2 overexpression and increased NMDAR-mediated transmission, which might lead to impaired synaptic plasticity and memory formation in a region-specific manner, we provide an important advance to our current understanding of the cellular mechanisms underlying PAE-dependent defective hippocampal function.
Collapse
Affiliation(s)
- Wladimir Plaza-Briceño
- Departamento de Ciencias Biomédicas, Facultad De Medicina, Universidad Católica Del Norte, Coquimbo, Chile.,Programa de Magíster en Ciencias Biológicas mención Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaiso, Chile
| | - Sebastián F Estay
- Facultad de Ciencias, Centro Interdisciplinario de Neurociencias de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile.,Programa de Doctorado en Ciencias mención Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaiso, Chile
| | - Erwin de la Fuente-Ortega
- Departamento de Ciencias Biomédicas, Facultad De Medicina, Universidad Católica Del Norte, Coquimbo, Chile
| | - Camilo Gutiérrez
- Departamento de Ciencias Biomédicas, Facultad De Medicina, Universidad Católica Del Norte, Coquimbo, Chile
| | - Gina Sánchez
- Programa de Fisiopatología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile.,CEMC, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Cecilia Hidalgo
- CEMC, Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Biomedical Neuroscience Institute, Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Departamento de Neurociencia, Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Programa de Fisiología y Biofísica, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Andrés E Chávez
- Facultad de Ciencias, Centro Interdisciplinario de Neurociencias de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
| | - Paola A Haeger
- Departamento de Ciencias Biomédicas, Facultad De Medicina, Universidad Católica Del Norte, Coquimbo, Chile
| |
Collapse
|
14
|
Maloney SE, Creeley CE, Hartman RE, Yuede CM, Zorumski CF, Jevtovic-Todorovic V, Dikranian K, Noguchi KK, Farber NB, Wozniak DF. Using animal models to evaluate the functional consequences of anesthesia during early neurodevelopment. Neurobiol Learn Mem 2019; 165:106834. [PMID: 29550366 PMCID: PMC6179938 DOI: 10.1016/j.nlm.2018.03.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 02/16/2018] [Accepted: 03/13/2018] [Indexed: 12/15/2022]
Abstract
Fifteen years ago Olney and colleagues began using animal models to evaluate the effects of anesthetic and sedative agents (ASAs) on neurodevelopment. The results from ongoing studies indicate that, under certain conditions, exposure to these drugs during development induces an acute elevated apoptotic neurodegenerative response in the brain and long-term functional impairments. These animal models have played a significant role in bringing attention to the possible adverse effects of exposing the developing brain to ASAs when few concerns had been raised previously in the medical community. The apoptotic degenerative response resulting from neonatal exposure to ASAs has been replicated in many studies in both rodents and non-human primates, suggesting that a similar effect may occur in humans. In both rodents and non-human primates, significantly increased levels of apoptotic degeneration are often associated with functional impairments later in life. However, behavioral deficits following developmental ASA exposure have not been consistently reported even when significantly elevated levels of apoptotic degeneration have been documented in animal models. In the present work, we review this literature and propose a rodent model for assessing potential functional deficits following neonatal ASA exposure with special reference to experimental design and procedural issues. Our intent is to improve test sensitivity and replicability for detecting subtle behavioral effects, and thus enhance the translational significance of ASA models.
Collapse
Affiliation(s)
- Susan E Maloney
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA; Intellectual and Developmental Disabilities Research Center, Washington University, St. Louis, MO, USA
| | - Catherine E Creeley
- Department of Psychology, The State University of New York at Fredonia, Fredonia, NY 14063, USA
| | - Richard E Hartman
- Department of Psychology, Loma Linda University, 11130 Anderson St., Loma Linda, CA 92354, USA
| | - Carla M Yuede
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA; Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Charles F Zorumski
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA; Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Krikor Dikranian
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, USA
| | - Kevin K Noguchi
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA; Intellectual and Developmental Disabilities Research Center, Washington University, St. Louis, MO, USA
| | - Nuri B Farber
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA; Intellectual and Developmental Disabilities Research Center, Washington University, St. Louis, MO, USA
| | - David F Wozniak
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA; Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, MO, USA; Intellectual and Developmental Disabilities Research Center, Washington University, St. Louis, MO, USA.
| |
Collapse
|
15
|
Contreras A, Morales L, Del Olmo N. The intermittent administration of ethanol during the juvenile period produces changes in the expression of hippocampal genes and proteins and deterioration of spatial memory. Behav Brain Res 2019; 372:112033. [PMID: 31201872 DOI: 10.1016/j.bbr.2019.112033] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 06/10/2019] [Accepted: 06/11/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND Binge drinking is a pattern of alcohol intake characterized by excessive and intermittent alcohol consumption over a very short period of time that is more used during adolescence. We aim to compare the lasting effects of a chronic-moderate vs. this intermittent-excessive way of alcohol intake during adolescence in spatial memory and in the expression of glutamatergic receptors and GSK3β activity. METHODS Adolescent male Wistar rats were given ethanol/saline i.p. injections in four different groups: High-I (4 g/kg of a 25% (vol/vol) every 3 days), Low-I (1 g/kg of a 5% (vol/vol) every 3 days), M (0.3 g/kg of a 2.5% (vol/vol) daily) and Control (C, sterile isotonic saline daily). Rats received ethanol for up to five 3-day cycles. Spatial memory was measured by spontaneous alternation in the Y-Maze. Gene and protein expression of hippocampal proteins were also analysed. RESULTS Both high- and low-intermittent ethanol administration produced spatial memory impairment and changes in glutamatergic receptors gene expression were observed regardless of the pattern of exposure. High doses of intermittent alcohol administration produced an increase of phosphorylation of GSK3β Ser9. Moreover, moderate alcohol administration produced a down-regulation of the AMPAR 2/3 ratio despite lack of spatial memory deficits. CONCLUSIONS Excessive and intermittent ethanol exposure during adolescence impaired the spatial memory processes during adulthood regardless of the amount of alcohol administered. Moreover, chronic-moderate and intermittent pattern induced changes in the expression of glutamatergic receptors. In addition, high-intermittent ethanol exposure during adolescence inactivated GSK3β by increasing its phosphorylation in Ser9.
Collapse
Affiliation(s)
- Ana Contreras
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU-San Pablo, Spain
| | - Lidia Morales
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU-San Pablo, Spain
| | - Nuria Del Olmo
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU-San Pablo, Spain.
| |
Collapse
|
16
|
Razumkina EV, Anokhin PK, Proskuryakova TV, Shamakina IY. [Experimental approaches to the investigation of behavioral disorders associated with prenatal alcohol exposure]. Zh Nevrol Psikhiatr Im S S Korsakova 2019; 118:79-88. [PMID: 29658509 DOI: 10.17116/jnevro20181181279-88] [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: 11/18/2022]
Abstract
Fetal alcohol spectrum disorders (FASD) is an umbrella term which covers a wide range of deficits in prenatal and postnatal growth, anatomy and CNS functions produced by prenatal alcohol exposure. The most severe form of FASD is fetal alcohol syndrome (FAS) characterized by additional specific craniofacial and brain malformations. Despite a high prevalence and extensive clinical studies, the fundamental mechanisms of FASD are still poorly understood. Thereby, experimental models, which allow better control for both socio-environmental and genetic factors, are critical to our understanding of FASD. The review is focused on the effects of exposure to alcohol during the prenatal period in animal models. The authors outline that prenatally alcohol-induced changes in motor and executive functions, learning and memory, stress reactivity and affective state are remarkably parallel between animals and humans. Finally, the authors consider a potential impact of postnatal social and environmental factors on the outcome in experimental models of FASD.
Collapse
Affiliation(s)
- E V Razumkina
- Serbsky National Medical Research Center for Psychiatry and Narcology, Moscow, Russia
| | - P K Anokhin
- Serbsky National Medical Research Center for Psychiatry and Narcology, Moscow, Russia
| | - T V Proskuryakova
- Serbsky National Medical Research Center for Psychiatry and Narcology, Moscow, Russia
| | - I Yu Shamakina
- Serbsky National Medical Research Center for Psychiatry and Narcology, Moscow, Russia
| |
Collapse
|
17
|
Subbanna S, Nagre NN, Shivakumar M, Joshi V, Psychoyos D, Kutlar A, Umapathy NS, Basavarajappa BS. CB1R-Mediated Activation of Caspase-3 Causes Epigenetic and Neurobehavioral Abnormalities in Postnatal Ethanol-Exposed Mice. Front Mol Neurosci 2018; 11:45. [PMID: 29515368 PMCID: PMC5826222 DOI: 10.3389/fnmol.2018.00045] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 02/02/2018] [Indexed: 12/15/2022] Open
Abstract
Alcohol exposure can affect brain development, leading to long-lasting behavioral problems, including cognitive impairment, which together is defined as fetal alcohol spectrum disorder (FASD). However, the fundamental mechanisms through which this occurs are largely unknown. In this study, we report that the exposure of postnatal day 7 (P7) mice to ethanol activates caspase-3 via cannabinoid receptor type-1 (CB1R) in neonatal mice and causes a reduction in methylated DNA binding protein (MeCP2) levels. The developmental expression of MeCP2 in mice is closely correlated with synaptogenesis and neuronal maturation. It was shown that ethanol treatment of P7 mice enhanced Mecp2 mRNA levels but reduced protein levels. The genetic deletion of CB1R prevented, and administration of a CB1R antagonist before ethanol treatment of P7 mice inhibited caspase-3 activation. Additionally, it reversed the loss of MeCP2 protein, cAMP response element binding protein (CREB) activation, and activity-regulated cytoskeleton-associated protein (Arc) expression. The inhibition of caspase-3 activity prior to ethanol administration prevented ethanol-induced loss of MeCP2, CREB activation, epigenetic regulation of Arc expression, long-term potentiation (LTP), spatial memory deficits and activity-dependent impairment of several signaling molecules, including MeCP2, in adult mice. Collectively, these results reveal that the ethanol-induced CB1R-mediated activation of caspase-3 degrades the MeCP2 protein in the P7 mouse brain and causes long-lasting neurobehavioral deficits in adult mice. This CB1R-mediated instability of MeCP2 during active synaptic maturation may disrupt synaptic circuit maturation and lead to neurobehavioral abnormalities, as observed in this animal model of FASD.
Collapse
Affiliation(s)
- Shivakumar Subbanna
- Division of Analytical Psychopharmacology, Nathan Kline Institute for Psychiatric Research, New York, NY, United States
| | - Nagaraja N. Nagre
- Division of Analytical Psychopharmacology, Nathan Kline Institute for Psychiatric Research, New York, NY, United States
| | - Madhu Shivakumar
- Division of Analytical Psychopharmacology, Nathan Kline Institute for Psychiatric Research, New York, NY, United States
| | - Vikram Joshi
- Division of Analytical Psychopharmacology, Nathan Kline Institute for Psychiatric Research, New York, NY, United States
| | - Delphine Psychoyos
- Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, TX, United States
| | - Abdullah Kutlar
- Center for Blood Disorders, Augusta University, Augusta, GA, United States
| | | | - Balapal S. Basavarajappa
- Division of Analytical Psychopharmacology, Nathan Kline Institute for Psychiatric Research, New York, NY, United States
- New York State Psychiatric Institute, New York, NY, United States
- Department of Psychiatry, College of Physicians & Surgeons, Columbia University, New York, NY, United States
- Department of Psychiatry, New York University Langone Medical Center, New York, NY, United States
| |
Collapse
|
18
|
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: 282] [Impact Index Per Article: 35.3] [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
|
19
|
Long Term Depression in Rat Hippocampus and the Effect of Ethanol during Fetal Life. Brain Sci 2017; 7:brainsci7120157. [PMID: 29182556 PMCID: PMC5742760 DOI: 10.3390/brainsci7120157] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 11/20/2017] [Accepted: 11/27/2017] [Indexed: 12/11/2022] Open
Abstract
Alcohol (ethanol) disturbs cognitive functions including learning and memory in humans, non-human primates, and laboratory animals such as rodents. As studied in animals, cellular mechanisms for learning and memory include bidirectional synaptic plasticity, long-term potentiation (LTP), and long-term depression (LTD), primarily in the hippocampus. Most of the research in the field of alcohol has analyzed the effects of ethanol on LTP; however, with recent advances in the understanding of the physiological role of LTD in learning and memory, some authors have examined the effects of ethanol exposure on this particular signal. In the present review, I will focus on hippocampal LTD recorded in rodents and the effects of fetal alcohol exposure on this signal. A synthesis of the findings indicates that prenatal ethanol exposure disturbs LTD concurrently with LTP in offspring and that both glutamatergic and γ-aminobutyric acid (GABA) neurotransmissions are altered and contribute to LTD disturbances. Although the ultimate mode of action of ethanol on these two transmitter systems is not yet clear, novel suggestions have recently appeared in the literature.
Collapse
|
20
|
Lewin M, Ilina M, Betz J, Masiello K, Hui M, Wilson DA, Saito M. Developmental Ethanol-Induced Sleep Fragmentation, Behavioral Hyperactivity, Cognitive Impairment and Parvalbumin Cell Loss are Prevented by Lithium Co-treatment. Neuroscience 2017; 369:269-277. [PMID: 29183826 DOI: 10.1016/j.neuroscience.2017.11.033] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 11/16/2017] [Accepted: 11/17/2017] [Indexed: 02/08/2023]
Abstract
Developmental ethanol exposure is a well-known cause of lifelong cognitive deficits, behavioral hyperactivity, emotional dysregulation, and more. In healthy adults, sleep is thought to have a critical involvement in each of these processes. Our previous work has demonstrated that some aspects of cognitive impairment in adult mice exposed at postnatal day 7 (P7) to ethanol (EtOH) correlate with slow-wave sleep (SWS) fragmentation (Wilson et al., 2016). We and others have also previously demonstrated that co-treatment with LiCl on the day of EtOH exposure prevents many of the anatomical and physiological impairments observed in adults. Here we explored cognitive function, diurnal rhythms (activity, temperature), SWS, and parvalbumin (PV) and perineuronal net (PNN)-positive cell densities in adult mice that had received a single day of EtOH exposure on P7 and saline-treated littermate controls. Half of the animals also received a LiCl injection on P7. The results suggest that developmental EtOH resulted in adult behavioral hyperactivity, cognitive impairment, and reduced SWS compared to saline controls. Both of these effects were reduced by LiCl treatment on the day of EtOH exposure. Finally, developmental EtOH resulted in decreased PV/PNN-expressing cells in retrosplenial (RS) cortex and dorsal CA3 hippocampus at P90. As with sleep and behavioral activity, LiCl treatment reduced this decrease in PV expression. Together, these results further clarify the long-lasting effects of developmental EtOH on adult behavior, physiology, and anatomy. Furthermore, they demonstrate the neuroprotective effects of LiCl co-treatment on this wide range of developmental EtOH's long-lasting consequences.
Collapse
Affiliation(s)
- M Lewin
- Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, United States; Sackler Neuroscience Graduate Program, NYU School of Medicine, New York, NY, United States
| | - M Ilina
- Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, United States
| | - J Betz
- Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, United States
| | - K Masiello
- Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, United States
| | - M Hui
- Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, United States
| | - D A Wilson
- Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, United States; Department of Child and Adolescent Psychiatry, NYU School of Medicine, New York, NY, United States.
| | - M Saito
- Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, United States; Department of Psychiatry, NYU School of Medicine, New York, NY, United States
| |
Collapse
|
21
|
Paolozza A, Munoz DP, Brien D, Reynolds JN. Immediate Neural Plasticity Involving Reaction Time in a Saccadic Eye Movement Task is Intact in Children With Fetal Alcohol Spectrum Disorder. Alcohol Clin Exp Res 2016; 40:2351-2358. [DOI: 10.1111/acer.13224] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 08/19/2016] [Indexed: 11/30/2022]
Affiliation(s)
- Angelina Paolozza
- Centre for Neuroscience Studies; Queen's University; Kingston Ontario Canada
| | - Douglas P. Munoz
- Centre for Neuroscience Studies; Queen's University; Kingston Ontario Canada
- Department of Biomedical and Molecular Sciences; Queen's University; Kingston Ontario Canada
| | - Donald Brien
- Centre for Neuroscience Studies; Queen's University; Kingston Ontario Canada
| | - James N. Reynolds
- Centre for Neuroscience Studies; Queen's University; Kingston Ontario Canada
- Department of Biomedical and Molecular Sciences; Queen's University; Kingston Ontario Canada
| |
Collapse
|
22
|
Subbanna S, Nagre NN, Shivakumar M, Basavarajappa BS. A single day of 5-azacytidine exposure during development induces neurodegeneration in neonatal mice and neurobehavioral deficits in adult mice. Physiol Behav 2016; 167:16-27. [PMID: 27594097 DOI: 10.1016/j.physbeh.2016.08.036] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 08/30/2016] [Accepted: 08/31/2016] [Indexed: 12/13/2022]
Abstract
The present study was undertaken to evaluate the immediate and long-term effects of a single-day exposure to 5-Azacytidine (5-AzaC), a DNA methyltransferase inhibitor, on neurobehavioral abnormalities in mice. Our findings suggest that the 5-AzaC treatment significantly inhibited DNA methylation, impaired extracellular signal-regulated kinase (ERK1/2) activation and reduced expression of the activity-regulated cytoskeleton-associated protein (Arc). These events lead to the activation of caspase-3 (a marker for neurodegeneration) in several brain regions, including the hippocampus and cortex, two brain areas that are essential for memory formation and memory storage, respectively. 5-AzaC treatment of P7 mice induced significant deficits in spatial memory, social recognition, and object memory in adult mice and deficits in long-term potentiation (LTP) in adult hippocampal slices. Together, these data demonstrate that the inhibition of DNA methylation by 5-AzaC treatment in P7 mice causes neurodegeneration and impairs ERK1/2 activation and Arc protein expression in neonatal mice and induces behavioral abnormalities in adult mice. DNA methylation-mediated mechanisms appear to be necessary for the proper maturation of synaptic circuits during development, and disruption of this process by 5-AzaC could lead to abnormal cognitive function.
Collapse
Affiliation(s)
- Shivakumar Subbanna
- Division of Analytical Psychopharmacology, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA
| | - Nagaraja N Nagre
- Division of Analytical Psychopharmacology, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA
| | - Madhu Shivakumar
- Division of Analytical Psychopharmacology, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA
| | - Balapal S Basavarajappa
- Division of Analytical Psychopharmacology, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA; New York State Psychiatric Institute, New York, NY 10032, USA; Department of Psychiatry, College of Physicians & Surgeons, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
23
|
Amin FU, Shah SA, Kim MO. Glycine inhibits ethanol-induced oxidative stress, neuroinflammation and apoptotic neurodegeneration in postnatal rat brain. Neurochem Int 2016; 96:1-12. [DOI: 10.1016/j.neuint.2016.04.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 03/31/2016] [Accepted: 04/01/2016] [Indexed: 12/22/2022]
|
24
|
Patten AR, Sawchuk S, Wortman RC, Brocardo PS, Gil-Mohapel J, Christie BR. Prenatal ethanol exposure impairs temporal ordering behaviours in young adult rats. Behav Brain Res 2015; 299:81-9. [PMID: 26632335 DOI: 10.1016/j.bbr.2015.11.032] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Revised: 11/19/2015] [Accepted: 11/23/2015] [Indexed: 02/03/2023]
Abstract
Prenatal ethanol exposure (PNEE) causes significant deficits in functional (i.e., synaptic) plasticity in the dentate gyrus (DG) and cornu ammonis (CA) hippocampal sub-regions of young adult male rats. Previous research has shown that in the DG, these deficits are not apparent in age-matched PNEE females. This study aimed to expand these findings and determine if PNEE induces deficits in hippocampal-dependent behaviours in both male and female young adult rats (PND 60). The metric change behavioural test examines DG-dependent deficits by determining whether an animal can detect a metric change between two identical objects. The temporal order behavioural test is thought to rely in part on the CA sub-region of the hippocampus and determines whether an animal will spend more time exploring an object that it has not seen for a larger temporal window as compared to an object that it has seen more recently. Using the liquid diet model of FASD (where 6.6% (v/v) ethanol is provided through a liquid diet consumed ad libitum throughout the entire gestation), we found that PNEE causes a significant impairment in the temporal order task, while no deficits in the DG-dependent metric change task were observed. There were no significant differences between males and females for either task. These results indicate that behaviours relying partially on the CA-region may be more affected by PNEE than those that rely on the DG.
Collapse
Affiliation(s)
- Anna R Patten
- Division of Medical Sciences, Island Medical Program, University of Victoria, Victoria, British Columbia, Canada.
| | - Scott Sawchuk
- Division of Medical Sciences, Island Medical Program, University of Victoria, Victoria, British Columbia, Canada
| | - Ryan C Wortman
- Division of Medical Sciences, Island Medical Program, University of Victoria, Victoria, British Columbia, Canada
| | - Patricia S Brocardo
- Division of Medical Sciences, Island Medical Program, University of Victoria, Victoria, British Columbia, Canada
| | - Joana Gil-Mohapel
- Division of Medical Sciences, Island Medical Program, University of Victoria, Victoria, British Columbia, Canada
| | - Brian R Christie
- Division of Medical Sciences, Island Medical Program, University of Victoria, Victoria, British Columbia, Canada; Brain Research Centre and Program in Neuroscience, University of British Columbia, Vancouver, British Columbia, Canada; Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
25
|
Fetal Alcohol Spectrum Disorder: Potential Role of Endocannabinoids Signaling. Brain Sci 2015; 5:456-93. [PMID: 26529026 PMCID: PMC4701023 DOI: 10.3390/brainsci5040456] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 10/19/2015] [Accepted: 10/19/2015] [Indexed: 12/15/2022] Open
Abstract
One of the unique features of prenatal alcohol exposure in humans is impaired cognitive and behavioral function resulting from damage to the central nervous system (CNS), which leads to a spectrum of impairments referred to as fetal alcohol spectrum disorder (FASD). Human FASD phenotypes can be reproduced in the rodent CNS following prenatal ethanol exposure. Several mechanisms are expected to contribute to the detrimental effects of prenatal alcohol exposure on the developing fetus, particularly in the developing CNS. These mechanisms may act simultaneously or consecutively and differ among a variety of cell types at specific developmental stages in particular brain regions. Studies have identified numerous potential mechanisms through which alcohol can act on the fetus. Among these mechanisms are increased oxidative stress, mitochondrial damage, interference with the activity of growth factors, glia cells, cell adhesion molecules, gene expression during CNS development and impaired function of signaling molecules involved in neuronal communication and circuit formation. These alcohol-induced deficits result in long-lasting abnormalities in neuronal plasticity and learning and memory and can explain many of the neurobehavioral abnormalities found in FASD. In this review, the author discusses the mechanisms that are associated with FASD and provides a current status on the endocannabinoid system in the development of FASD.
Collapse
|
26
|
Stein LR, Zorumski CF, Imai SI, Izumi Y. Nampt is required for long-term depression and the function of GluN2B subunit-containing NMDA receptors. Brain Res Bull 2015; 119:41-51. [PMID: 26481044 DOI: 10.1016/j.brainresbull.2015.10.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2015] [Revised: 09/13/2015] [Accepted: 10/12/2015] [Indexed: 01/01/2023]
Abstract
Nicotinamide adenine dinucleotide (NAD(+)) is an essential coenzyme/cosubstrate for many biological processes in cellular metabolism. The rate-limiting step in the major pathway of mammalian NAD(+) biosynthesis is mediated by nicotinamide phosphoribosyltransferase (Nampt). Previously, we showed that mice lacking Nampt in forebrain excitatory neurons (CamKIIαNampt(-/-) mice) exhibited hyperactivity, impaired learning and memory, and reduced anxiety-like behaviors. However, it remained unclear if these functional effects were accompanied by synaptic changes. Here, we show that CamKIIαNampt(-/-) mice have impaired induction of long-term depression (LTD) in the Schaffer collateral pathway, but normal induction of long-term potentiation (LTP), at postnatal day 30. Pharmacological assessments demonstrated that CamKIIαNampt(-/-) mice also display dysfunction of synaptic GluN2B (NR2B)-containing N-methyl-d-aspartate receptors (NMDARs) prior to changes in NMDAR subunit expression. These results support a novel, important role for Nampt-mediated NAD(+) biosynthesis in LTD and in the function of GluN2B-containing NMDARs.
Collapse
Affiliation(s)
- Liana Roberts Stein
- Department of Developmental Biology, Washington University School of Medicine, Campus Box 8103, 660 South Euclid Avenue, St. Louis, MO 63110, USA; Department of Psychiatry, The Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, Campus Box 8134, 660 South Euclid Avenue, St. Louis, MO 63110, USA.
| | - Charles F Zorumski
- Department of Psychiatry, The Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, Campus Box 8134, 660 South Euclid Avenue, St. Louis, MO 63110, USA; Department of Anatomy and Neurobiology, Washington University School of Medicine, Campus Box 8134, 660 South Euclid Avenue, St. Louis, MO 63110, USA.
| | - Shin-Ichiro Imai
- Department of Developmental Biology, Washington University School of Medicine, Campus Box 8103, 660 South Euclid Avenue, St. Louis, MO 63110, USA.
| | - Yukitoshi Izumi
- Department of Psychiatry, The Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, Campus Box 8134, 660 South Euclid Avenue, St. Louis, MO 63110, USA.
| |
Collapse
|
27
|
Silvestre de Ferron B, Bennouar KE, Kervern M, Alaux-Cantin S, Robert A, Rabiant K, Antol J, Naassila M, Pierrefiche O. Two Binges of Ethanol a Day Keep the Memory Away in Adolescent Rats: Key Role for GLUN2B Subunit. Int J Neuropsychopharmacol 2015; 19:pyv087. [PMID: 26254123 PMCID: PMC4772273 DOI: 10.1093/ijnp/pyv087] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 07/23/2015] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Binge drinking is common in adolescents, but the impact of only a few binges on learning and memory appears underestimated. Many studies have tested the effects of long and intermittent ethanol exposure on long-term synaptic potentiation, and whether long-term synaptic depression is affected remains unknown. METHODS We studied the effects of one (3 g/kg, i.p.; blood ethanol content of 197.5±19 mg/dL) or 2 alcohol intoxications (given 9 hours apart) on adolescent rat's memory and synaptic plasticity in hippocampus slice after different delay. RESULTS Animals treated with 2 ethanol intoxications 48 hours before training phase in the novel object recognition task failed during test phase. As learning is related to NMDA-dependent mechanisms, we tested ketamine and found the same effect as ethanol, whereas D-serine prevented learning deficit. In hippocampus slice, NMDA-dependent long-term synaptic depression was abolished 48 hours after ethanol or ketamine but prevented after D-serine or in a low-Mg(2+) recording medium. Long-term synaptic depression abolition was not observed 8 days after treatment. An i.p. treatment with MK-801, tetrahydroisoxazolopyridine, or muscimol was ineffective, and long-term synaptic potentiation, intrinsic excitability, and glutamate release remained unaffected. The input/ouput curve for NMDA-fEPSPs was shifted to the left 48 hours after the binges with a stronger contribution of GluN2B subunit, leading to a leftward shift of the Bienenstock-Cooper-Munro relationship. Interestingly, there were no cellular effects after only one ethanol injection. CONCLUSION Two ethanol "binges" in adolescent rats are sufficient to reversibly abolish long-term synaptic depression and to evoke cognitive deficits via a short-lasting, repeated blockade of NMDA receptors only, inducing a change in the receptor subunit composition. Furthermore, ethanol effects developed over a 48-hour period of abstinence, indicating an important role of intermittence during a repeated long-duration binge behavior.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Olivier Pierrefiche
- INSERM ERI-24, GRAP, Groupe de Recherche sur l'Alcool et les Pharmacodépendances, Université Picardie Jules Verne, Bât. CURS, CHU-Sud, Amiens, France (Mr Silvestre de Ferron, Bennouar PhD, Kervern PhD, Alaux-Cantin PhD, Mr Robert, Mr Rabiant, Mr Antol, Naassila PhD, and Pierrefiche PhD).
| |
Collapse
|
28
|
Lantz CL, Sipe GO, Wong EL, Majewska AK, Medina AE. Effects of Developmental Alcohol Exposure on Potentiation and Depression of Visual Cortex Responses. Alcohol Clin Exp Res 2015; 39:1434-42. [PMID: 26108422 PMCID: PMC4515209 DOI: 10.1111/acer.12775] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 05/07/2015] [Indexed: 11/29/2022]
Abstract
BACKGROUND Neuronal plasticity deficits are thought to underlie abnormal neurodevelopment in fetal alcohol spectrum disorders and in animal models of this condition. Previously, we found that alcohol exposure during a period that is similar to the last months of gestation in humans disrupts ocular dominance plasticity (ODP), as measured in superficial cortical layers. We hypothesize that exposure to alcohol can differentially affect the potentiation and depression of responses that are necessary for activity-dependent sprouting and pruning of neuronal networks. ODP is an established paradigm that allows the assessment of activity-dependent depression and potentiation of responses in vivo. METHODS Mouse pups were exposed to 3.6 to 5 g/kg of ethanol in saline daily or every other day between postnatal days 4 and 9. Visual cortex plasticity was then assessed during the critical period for ODP using 2 techniques that separately record in layers 4 (visually evoked potentials [VEPs]) and 2/3 (optical imaging of intrinsic signals [OI]). RESULTS We discovered a layer-specific effect of early alcohol exposure. Recording of VEPs from layer 4 showed that while the potentiation component of ODP was disrupted in animals treated with alcohol when compared with saline controls, the depression component of ODP (Dc-ODP) was unaltered. In contrast, OI from layers 2/3 showed that Dc-ODP was markedly disrupted in alcohol-treated animals when compared with controls. CONCLUSIONS Combined with our previous work, these findings strongly suggest that developmental alcohol exposure has a distinct and layer-specific effect on the potentiation and depression of cortical responses after monocular deprivation.
Collapse
Affiliation(s)
- Crystal L Lantz
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, Virginia
- Department of Biology, University of Maryland, College Park, Maryland
| | - Grayson O Sipe
- Department of Neurobiology and Anatomy, University of Rochester Medical Center, Rochester, New York
| | - Elissa L Wong
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York
| | - Ania K Majewska
- Department of Neurobiology and Anatomy, University of Rochester Medical Center, Rochester, New York
| | - Alexandre E Medina
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, Virginia
- Department of Pediatrics, School of Medicine, University of Maryland, Baltimore, Maryland
| |
Collapse
|
29
|
Izumi Y, Zorumski CF. Sensitivity of N-methyl-D-aspartate receptor-mediated excitatory postsynaptic potentials and synaptic plasticity to TCN 201 and TCN 213 in rat hippocampal slices. J Pharmacol Exp Ther 2015; 352:267-73. [PMID: 25413830 PMCID: PMC4293441 DOI: 10.1124/jpet.114.220582] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 11/18/2014] [Indexed: 01/07/2023] Open
Abstract
Whereas ifenprodil has been used as a selective GluN1/GluN2B (NR1/NR2B, B-type) receptor antagonist to distinguish between GluN2B (NR2B) and GluN2A (NR2A)-containing N-methyl-d-aspartate receptors (NMDARs), TCN 201 (3-chloro-4-fluoro-N-[4-[[2-(phenylcarbonyl)hydrazino]carbonyl]benzyl]benzenesulphonamide) and TCN 213 [N-(cyclohexylmethyl)-2-[{5-[(phenylmethyl)amino]-1,3,4-thiadiazol-2-yl}thio]acetamide] have been found to be selective GluN1/GluN2A (NR1/NR2A, A-type) antagonists. Based on the premise that A- and B-types are major synaptic NMDARs, we examined whether inhibition of NMDAR excitatory postsynaptic potentials (EPSPs) by the TCN compounds and ifenprodil are complementary. Contrary to this prediction, inhibition of NMDAR EPSPs by the TCN compounds and ifenprodil were largely overlapping in the CA1 region of hippocampal slices from 30-day-old rats. After partial inhibition by ifenprodil, TCN compounds produced little further suppression of NMDAR EPSPs. Similarly, after partial inhibition by TCN compounds ifenprodil failed to further suppress NMDAR EPSPs. However, low micromolar d-2-amino-5-phosphonovalerate, a competitive NMDAR antagonist, which alone only partially inhibits NMDAR EPSPs, markedly suppresses residual NMDAR responses in the presence of ifenprodil or the TCNs, suggesting that low 2-amino-5-phosphonovalerate antagonizes both ifenprodil- and TCN-insensitive synaptic NMDARs. These observations can be most readily interpreted if ifenprodil and TCNs act on a similar population of synaptic NMDARs. Recent lines of evidence suggest that the majority of hippocampal synaptic NMDARs are triheteromers. If so, modulation of GluN2A, and not just GluN2B NMDARs, could dampen long-term depression (LTD). Indeed, both TCNs, like ifenprodil, blocked LTD, suggesting the involvement of ifenprodil- and TCN-sensitive NMDARs in LTD induction. However, the TCNs plus ifenprodil failed to inhibit long-term potentiation (LTP), suggesting that neither ifenprodil- nor TCN-sensitive NMDARs are essential for LTP induction.
Collapse
Affiliation(s)
- Yukitoshi Izumi
- Department of Psychiatry and Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, Missouri
| | - Charles F Zorumski
- Department of Psychiatry and Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
30
|
Nagre NN, Subbanna S, Shivakumar M, Psychoyos D, Basavarajappa BS. CB1-receptor knockout neonatal mice are protected against ethanol-induced impairments of DNMT1, DNMT3A, and DNA methylation. J Neurochem 2015; 132:429-442. [PMID: 25487288 DOI: 10.1111/jnc.13006] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Revised: 11/25/2014] [Accepted: 11/26/2014] [Indexed: 12/21/2022]
Abstract
The significant consequences of ethanol use during pregnancy are neurobehavioral abnormalities involving hippocampal and neocortex malfunctions that cause learning and memory deficits collectively named fetal alcohol spectrum disorder. However, the molecular mechanisms underlying these abnormalities are still poorly understood and therefore warrant systematic research. Here, we document novel epigenetic abnormalities in the mouse model of fetal alcohol spectrum disorder. Ethanol treatment of P7 mice, which induces activation of caspase 3, impaired DNA methylation through reduced DNA methyltransferases (DNMT1 and DNMT3A) levels. Inhibition of caspase 3 activity, before ethanol treatment, rescued DNMT1, DNMT3A proteins as well as DNA methylation levels. Blockade of histone methyltransferase (G9a) activity or cannabinoid receptor type-1 (CB1R), prior to ethanol treatment, which, respectively, inhibits or prevents activation of caspase 3, rescued the DNMT1 and DNMT3A proteins and DNA methylation. No reduction of DNMT1 and DNMT3A proteins and DNA methylation was found in P7 CB1R null mice, which exhibit no ethanol-induced activation of caspase 3. Together, these data demonstrate that ethanol-induced activation of caspase 3 impairs DNA methylation through DNMT1 and DNMT3A in the neonatal mouse brain, and such impairments are absent in CB1R null mice. Epigenetic events mediated by DNA methylation may be one of the essential mechanisms of ethanol teratogenesis. Schematic mechanism of action by which ethanol impairs DNA methylation. Studies have demonstrated that ethanol has the capacity to bring epigenetic changes to contribute to the development of fetal alcohol spectrum disorder (FASD). However, the mechanisms are not well studied. P7 ethanol induces the activation of caspase 3 and impairs DNA methylation through reduced DNA methyltransferases (DNMT1 and DNMT3A) proteins (→). The inhibition or genetic ablation of cannabinoid receptor type-1 or inhibition of histone methyltransferase (G9a) by Bix (-----) or inhibition of caspase 3 activation by Q- quinoline-Val-Asp(Ome)-CH2-O-phenoxy (Q-VD-OPh) () rescue loss of DNMT1, DNMT3A as well as DNA methylation. Hence, the putative DNMT1/DNMT3A/DNA methylation mechanism may have a potential regulatory role in FASD.
Collapse
Affiliation(s)
- Nagaraja N Nagre
- Division of Analytical Psychopharmacology, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA
| | - Shivakumar Subbanna
- Division of Analytical Psychopharmacology, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA
| | - Madhu Shivakumar
- Division of Analytical Psychopharmacology, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA
| | - Delphine Psychoyos
- Institute of Biosciences and Technology, Houston, Texas A&M University Health Science Center, Houston, TX 77030, USA
| | - Balapal S Basavarajappa
- Division of Analytical Psychopharmacology, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA.,New York State Psychiatric Institute, College of Physicians & Surgeons, Columbia University, New York, NY 10032, USA.,Department of Psychiatry, College of Physicians & Surgeons, Columbia University, New York, NY 10032, USA
| |
Collapse
|
31
|
Subbanna S, Nagre NN, Umapathy NS, Pace BS, Basavarajappa BS. Ethanol exposure induces neonatal neurodegeneration by enhancing CB1R Exon1 histone H4K8 acetylation and up-regulating CB1R function causing neurobehavioral abnormalities in adult mice. Int J Neuropsychopharmacol 2014; 18:pyu028. [PMID: 25609594 PMCID: PMC4376538 DOI: 10.1093/ijnp/pyu028] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Ethanol exposure to rodents during postnatal day 7 (P7), which is comparable to the third trimester of human pregnancy, induces long-term potentiation and memory deficits. However, the molecular mechanisms underlying these deficits are still poorly understood. METHODS In the present study, we explored the potential role of epigenetic changes at cannabinoid type 1 (CB1R) exon1 and additional CB1R functions, which could promote memory deficits in animal models of fetal alcohol spectrum disorder. RESULTS We found that ethanol treatment of P7 mice enhances acetylation of H4 on lysine 8 (H4K8ace) at CB1R exon1, CB1R binding as well as the CB1R agonist-stimulated GTPγS binding in the hippocampus and neocortex, two brain regions that are vulnerable to ethanol at P7 and are important for memory formation and storage, respectively. We also found that ethanol inhibits cyclic adenosine monophosphate response element-binding protein (CREB) phosphorylation and activity-regulated cytoskeleton-associated protein (Arc) expression in neonatal and adult mice. The blockade or genetic deletion of CB1Rs prior to ethanol treatment at P7 rescued CREB phosphorylation and Arc expression. CB1R knockout mice exhibited neither ethanol-induced neurodegeneration nor inhibition of CREB phosphorylation or Arc expression. However, both neonatal and adult mice did exhibit enhanced CREB phosphorylation and Arc protein expression. P7 ethanol-treated adult mice exhibited impaired spatial and social recognition memory, which were prevented by the pharmacological blockade or deletion of CB1Rs at P7. CONCLUSIONS Together, these findings suggest that P7 ethanol treatment induces CB1R expression through epigenetic modification of the CB1R gene, and that the enhanced CB1R function induces pCREB, Arc, spatial, and social memory deficits in adult mice.
Collapse
MESH Headings
- AIDS-Related Complex/metabolism
- Acetylation/drug effects
- Age Factors
- Animals
- Animals, Newborn/metabolism
- Animals, Newborn/psychology
- CREB-Binding Protein/metabolism
- Central Nervous System Depressants/toxicity
- Epigenesis, Genetic/drug effects
- Ethanol/toxicity
- Exons/drug effects
- Female
- Gene Expression Regulation/drug effects
- Hippocampus/drug effects
- Hippocampus/metabolism
- Histones/genetics
- Male
- Memory Disorders/chemically induced
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Neocortex/drug effects
- Neocortex/metabolism
- Neurodegenerative Diseases/chemically induced
- Neurodegenerative Diseases/metabolism
- Neurodegenerative Diseases/psychology
- Phosphorylation/drug effects
- Receptor, Cannabinoid, CB1/deficiency
- Receptor, Cannabinoid, CB1/drug effects
- Receptor, Cannabinoid, CB1/genetics
- Receptor, Cannabinoid, CB1/metabolism
- Social Behavior
- Up-Regulation/drug effects
Collapse
Affiliation(s)
- Shivakumar Subbanna
- Division of Analytical Psychopharmacology, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY (Drs Subbanna, Nagre, and Basavarajappa); Vascular Biology Center, Georgia Regents University, Augusta, GA (Dr Umapathy); Department of Pediatrics, Georgia Regents University, Augusta, GA (Dr Pace); New York State Psychiatric Institute, New York, NY (Dr Basavarajappa); Department of Psychiatry, College of Physicians & Surgeons, Columbia University, New York, NY (Dr Basavarajappa)
| | - Nagaraja N Nagre
- Division of Analytical Psychopharmacology, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY (Drs Subbanna, Nagre, and Basavarajappa); Vascular Biology Center, Georgia Regents University, Augusta, GA (Dr Umapathy); Department of Pediatrics, Georgia Regents University, Augusta, GA (Dr Pace); New York State Psychiatric Institute, New York, NY (Dr Basavarajappa); Department of Psychiatry, College of Physicians & Surgeons, Columbia University, New York, NY (Dr Basavarajappa)
| | - Nagavedi S Umapathy
- Division of Analytical Psychopharmacology, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY (Drs Subbanna, Nagre, and Basavarajappa); Vascular Biology Center, Georgia Regents University, Augusta, GA (Dr Umapathy); Department of Pediatrics, Georgia Regents University, Augusta, GA (Dr Pace); New York State Psychiatric Institute, New York, NY (Dr Basavarajappa); Department of Psychiatry, College of Physicians & Surgeons, Columbia University, New York, NY (Dr Basavarajappa)
| | - Betty S Pace
- Division of Analytical Psychopharmacology, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY (Drs Subbanna, Nagre, and Basavarajappa); Vascular Biology Center, Georgia Regents University, Augusta, GA (Dr Umapathy); Department of Pediatrics, Georgia Regents University, Augusta, GA (Dr Pace); New York State Psychiatric Institute, New York, NY (Dr Basavarajappa); Department of Psychiatry, College of Physicians & Surgeons, Columbia University, New York, NY (Dr Basavarajappa)
| | - Balapal S Basavarajappa
- Division of Analytical Psychopharmacology, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY (Drs Subbanna, Nagre, and Basavarajappa); Vascular Biology Center, Georgia Regents University, Augusta, GA (Dr Umapathy); Department of Pediatrics, Georgia Regents University, Augusta, GA (Dr Pace); New York State Psychiatric Institute, New York, NY (Dr Basavarajappa); Department of Psychiatry, College of Physicians & Surgeons, Columbia University, New York, NY (Dr Basavarajappa).
| |
Collapse
|
32
|
Ramachandran B, Ahmed S, Zafar N, Dean C. Ethanol inhibits long-term potentiation in hippocampal CA1 neurons, irrespective of lamina and stimulus strength, through neurosteroidogenesis. Hippocampus 2014; 25:106-18. [PMID: 25155179 DOI: 10.1002/hipo.22356] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/19/2014] [Indexed: 11/09/2022]
Abstract
Ethanol inhibits memory encoding and the induction of long-term potentiation (LTP) in CA1 neurons of the hippocampus. Hippocampal LTP at Schaffer collateral synapses onto CA1 pyramidal neurons has been widely studied as a cellular model of learning and memory, but there is striking heterogeneity in the underlying molecular mechanisms in distinct regions and in response to distinct stimuli. Basal and apical dendrites differ in terms of innervation, input specificity, and molecular mechanisms of LTP induction and maintenance, and different stimuli determine distinct molecular pathways of potentiation. However, lamina or stimulus-dependent effects of ethanol on LTP have not been investigated. Here, we tested the effect of acute application of 60 mM ethanol on LTP induction in distinct dendritic compartments (apical versus basal) of CA1 neurons, and in response to distinct stimulation paradigms (single versus repeated, spaced high frequency stimulation). We found that ethanol completely blocks LTP in apical dendrites, whereas it reduces the magnitude of LTP in basal dendrites. Acute ethanol treatment for just 15 min altered pre- and post-synaptic protein expression. Interestingly, ethanol increases the neurosteroid allopregnanolone, which causes ethanol-dependent inhibition of LTP, more prominently in apical dendrites, where ethanol has greater effects on LTP. This suggests that ethanol has general effects on fundamental properties of synaptic plasticity, but the magnitude of its effect on LTP differs depending on hippocampal sub-region and stimulus strength.
Collapse
Affiliation(s)
- Binu Ramachandran
- Trans-Synaptic Signaling Group, European Neuroscience Institute (ENI), Grisebachstrasse 5, 37077, Goettingen, Germany
| | | | | | | |
Collapse
|
33
|
Sadrian B, Lopez-Guzman M, Wilson DA, Saito M. Distinct neurobehavioral dysfunction based on the timing of developmental binge-like alcohol exposure. Neuroscience 2014; 280:204-19. [PMID: 25241068 DOI: 10.1016/j.neuroscience.2014.09.008] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Revised: 09/03/2014] [Accepted: 09/05/2014] [Indexed: 12/24/2022]
Abstract
Gestational exposure to alcohol can result in long-lasting behavioral deficiencies generally described as fetal alcohol spectrum disorder (FASD). FASD-modeled rodent studies of acute ethanol exposure typically select one developmental window to simulate a specific context equivalent of human embryogenesis, and study consequences of ethanol exposure within that particular developmental epoch. Exposure timing is likely a large determinant in the neurobehavioral consequence of early ethanol exposure, as each brain region is variably susceptible to ethanol cytotoxicity and has unique sensitive periods in their development. We made a parallel comparison of the long-term effects of single-day binge ethanol at either embryonic day 8 (E8) or postnatal day 7 (P7) in male and female mice, and here demonstrate the differential long-term impacts on neuroanatomy, behavior and in vivo electrophysiology of two systems with very different developmental trajectories. The significant long-term differences in odor-evoked activity, local circuit inhibition, and spontaneous coherence between brain regions in the olfacto-hippocampal pathway that were found as a result of developmental ethanol exposure, varied based on insult timing. Long-term effects on cell proliferation and interneuron cell density were also found to vary by insult timing as well as by region. Finally, spatial memory performance and object exploration were affected in P7-exposed mice, but not E8-exposed mice. Our physiology and behavioral results are conceptually coherent with the neuroanatomical data attained from these same mice. Our results recognize both variable and shared effects of ethanol exposure timing on long-term circuit function and their supported behavior.
Collapse
Affiliation(s)
- B Sadrian
- Department of Child and Adolescent Psychiatry, NYU School of Medicine, New York, NY, United States; Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, United States.
| | - M Lopez-Guzman
- Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, United States
| | - D A Wilson
- Department of Child and Adolescent Psychiatry, NYU School of Medicine, New York, NY, United States; Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, United States
| | - M Saito
- Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, United States; Department of Psychiatry, NYU School of Medicine, New York, NY, United States
| |
Collapse
|
34
|
Subbanna S, Basavarajappa BS. Pre-administration of G9a/GLP inhibitor during synaptogenesis prevents postnatal ethanol-induced LTP deficits and neurobehavioral abnormalities in adult mice. Exp Neurol 2014; 261:34-43. [PMID: 25017367 DOI: 10.1016/j.expneurol.2014.07.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Revised: 06/10/2014] [Accepted: 07/02/2014] [Indexed: 01/01/2023]
Abstract
It has been widely accepted that deficits in neuronal plasticity underlie the cognitive abnormalities observed in fetal alcohol spectrum disorder (FASD). Exposure of rodents to acute ethanol on postnatal day 7 (P7), which is equivalent to the third trimester of fetal development in human, induces long-term potentiation (LTP) and memory deficits in adult animals. However, the molecular mechanisms underlying these deficits are not well understood. Recently, we found that histone H3 dimethylation (H3K9me2), which is mediated by G9a (lysine dimethyltransferase), is responsible for the neurodegeneration caused by ethanol exposure in P7 mice. In addition, pharmacological inhibition of G9a prior to ethanol treatment at P7 normalized H3K9me2 proteins to basal levels and prevented neurodegeneration in neonatal mice. Here, we tested the hypothesis that pre-administration of G9a/GLP inhibitor (Bix-01294, Bix) in conditions in which ethanol induces neurodegeneration would be neuroprotective against P7 ethanol-induced deficits in LTP, memory and social recognition behavior in adult mice. Ethanol treatment at P7 induces deficits in LTP, memory and social recognition in adult mice and these deficits were prevented by Bix pretreatment at P7. Together, these findings provide physiological and behavioral evidence that the long-term harmful consequences on brain function after ethanol exposure with a third trimester equivalent have an epigenetic origin.
Collapse
Affiliation(s)
- Shivakumar Subbanna
- Division of Analytical Psychopharmacology, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA
| | - Balapal S Basavarajappa
- Division of Analytical Psychopharmacology, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA; New York State Psychiatric Institute, New York, NY 10032, USA; Department of Psychiatry, College of Physicians & Surgeons, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
35
|
Protection of the Developing Brain with Anthocyanins Against Ethanol-Induced Oxidative Stress and Neurodegeneration. Mol Neurobiol 2014; 51:1278-91. [DOI: 10.1007/s12035-014-8805-7] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Accepted: 06/23/2014] [Indexed: 10/25/2022]
|
36
|
The impairment in spatial learning and hippocampal LTD induced through the PKA pathway in juvenile-onset diabetes rats are rescued by modulating NMDA receptor function. Neurosci Res 2014; 81-82:55-63. [DOI: 10.1016/j.neures.2014.02.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Revised: 01/22/2014] [Accepted: 02/03/2014] [Indexed: 12/30/2022]
|
37
|
Acute and chronic effects of ethanol on learning-related synaptic plasticity. Alcohol 2014; 48:1-17. [PMID: 24447472 DOI: 10.1016/j.alcohol.2013.09.045] [Citation(s) in RCA: 123] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Revised: 09/16/2013] [Accepted: 09/18/2013] [Indexed: 11/23/2022]
Abstract
Alcoholism is associated with acute and long-term cognitive dysfunction including memory impairment, resulting in substantial disability and cost to society. Thus, understanding how ethanol impairs cognition is essential for developing treatment strategies to dampen its adverse impact. Memory processing is thought to involve persistent, use-dependent changes in synaptic transmission, and ethanol alters the activity of multiple signaling molecules involved in synaptic processing, including modulation of the glutamate and gamma-aminobutyric acid (GABA) transmitter systems that mediate most fast excitatory and inhibitory transmission in the brain. Effects on glutamate and GABA receptors contribute to ethanol-induced changes in long-term potentiation (LTP) and long-term depression (LTD), forms of synaptic plasticity thought to underlie memory acquisition. In this paper, we review the effects of ethanol on learning-related forms of synaptic plasticity with emphasis on changes observed in the hippocampus, a brain region that is critical for encoding contextual and episodic memories. We also include studies in other brain regions as they pertain to altered cognitive and mental function. Comparison of effects in the hippocampus to other brain regions is instructive for understanding the complexities of ethanol's acute and long-term pharmacological consequences.
Collapse
|
38
|
Patten AR, Fontaine CJ, Christie BR. A comparison of the different animal models of fetal alcohol spectrum disorders and their use in studying complex behaviors. Front Pediatr 2014; 2:93. [PMID: 25232537 PMCID: PMC4153370 DOI: 10.3389/fped.2014.00093] [Citation(s) in RCA: 143] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 08/20/2014] [Indexed: 12/31/2022] Open
Abstract
Prenatal ethanol exposure (PNEE) has been linked to widespread impairments in brain structure and function. There are a number of animal models that are used to study the structural and functional deficits caused by PNEE, including, but not limited to invertebrates, fish, rodents, and non-human primates. Animal models enable a researcher to control important variables such as the route of ethanol administration, as well as the timing, frequency and amount of ethanol exposure. Each animal model and system of exposure has its place, depending on the research question being undertaken. In this review, we will examine the different routes of ethanol administration and the various animal models of fetal alcohol spectrum disorders (FASD) that are commonly used in research, emphasizing their strengths and limitations. We will also present an up-to-date summary on the effects of prenatal/neonatal ethanol exposure on behavior across the lifespan, focusing on learning and memory, olfaction, social, executive, and motor functions. Special emphasis will be placed where the various animal models best represent deficits observed in the human condition and offer a viable test bed to examine potential therapeutics for human beings with FASD.
Collapse
Affiliation(s)
- Anna R Patten
- Division of Medical Sciences, University of Victoria , Victoria, BC , Canada
| | | | - Brian R Christie
- Division of Medical Sciences, University of Victoria , Victoria, BC , Canada ; Department of Biology, University of Victoria , Victoria, BC , Canada ; Program in Neuroscience, The Brain Research Centre, University of British Columbia , Vancouver, BC , Canada ; Department of Cellular and Physiological Sciences, University of British Columbia , Vancouver, BC , Canada
| |
Collapse
|
39
|
Subbanna S, Nagre NN, Shivakumar M, Umapathy NS, Psychoyos D, Basavarajappa BS. Ethanol induced acetylation of histone at G9a exon1 and G9a-mediated histone H3 dimethylation leads to neurodegeneration in neonatal mice. Neuroscience 2013; 258:422-32. [PMID: 24300108 DOI: 10.1016/j.neuroscience.2013.11.043] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Revised: 11/19/2013] [Accepted: 11/20/2013] [Indexed: 11/16/2022]
Abstract
The transient exposure of immature rodents to ethanol during postnatal day 7 (P7), comparable to a time point within the third trimester of human pregnancy, induces neurodegeneration. However, the molecular mechanisms underlying the deleterious effects of ethanol on the developing brain are poorly understood. In our previous study, we showed that a high dose administration of ethanol at P7 enhances G9a and leads to caspase-3-mediated degradation of dimethylated H3 on lysine 9 (H3K9me2). In this study, we investigated the potential role of epigenetic changes at G9a exon1, G9a-mediated H3 dimethylation on neurodegeneration and G9a-associated proteins in the P7 brain following exposure to a low dose of ethanol. We found that a low dose of ethanol induces mild neurodegeneration in P7 mice, enhances specific acetylation of H3 on lysine 14 (H3K14ace) at G9a exon1, G9a protein levels, augments the dimethylation of H3K9 and H3 lysine 27 (H3K27me2). However, neither dimethylated H3K9 nor K27 underwent degradation. Pharmacological inhibition of G9a activity prior to ethanol treatment prevented H3 dimethylation and neurodegeneration. Further, our immunoprecipitation data suggest that G9a directly associates with DNA methyltransferase (DNMT3A) and methyl-CpG-binding protein 2 (MeCP2). In addition, DNMT3A and MeCP2 protein levels were enhanced by a low dose of ethanol that was shown to induce mild neurodegeneration. Collectively, these epigenetic alterations lead to association of G9a, DNMT3A and MeCP2 to form a larger repressive complex and have a significant role in low-dose ethanol-induced neurodegeneration in the developing brain.
Collapse
Affiliation(s)
- S Subbanna
- Division of Analytical Psychopharmacology, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA
| | - N N Nagre
- Division of Analytical Psychopharmacology, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA
| | - M Shivakumar
- Division of Analytical Psychopharmacology, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA
| | - N S Umapathy
- Vascular Biology Center, Georgia Regents University, Augusta, GA 30912, USA
| | - D Psychoyos
- Institute of Biosciences and Technology, Houston, Texas A&M University Health Science Center, Houston, TX 77030, USA
| | - B S Basavarajappa
- Division of Analytical Psychopharmacology, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA; New York State Psychiatric Institute, College of Physicians & Surgeons, Columbia University, New York, NY 10032, USA; Department of Psychiatry, College of Physicians & Surgeons, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
40
|
Anandamide-CB1 receptor signaling contributes to postnatal ethanol-induced neonatal neurodegeneration, adult synaptic, and memory deficits. J Neurosci 2013; 33:6350-66. [PMID: 23575834 DOI: 10.1523/jneurosci.3786-12.2013] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The transient exposure of immature rodents to ethanol during postnatal day 7 (P7), which is comparable with the third trimester in human pregnancy, induces synaptic dysfunctions. However, the molecular mechanisms underlying these dysfunctions are still poorly understood. Although the endocannabinoid system has been shown to be an important modulator of ethanol sensitivity in adult mice, its potential role in synaptic dysfunctions in mice exposed to ethanol during early brain development is not examined. In this study, we investigated the potential role of endocannabinoids and the cannabinoid receptor type 1 (CB1R) in neonatal neurodegeneration and adult synaptic dysfunctions in mice exposed to ethanol at P7. Ethanol treatment at P7, which induces neurodegeneration, increased anandamide (AEA) but not 2-arachidonylglycerol biosynthesis and CB1R protein expression in the hippocampus and cortex, two brain areas that are important for memory formation and storage, respectively. N-Arachidonoyl phosphatidylethanolamine-phospholipase D (NAPE-PLD), glycerophosphodiesterase (GDE1), and CB1R protein expression were enhanced by transcriptional activation of the genes encoding NAPE-PLD, GDE1, and CB1R proteins, respectively. In addition, ethanol inhibited ERK1/2 and AKT phosphorylation. The blockade of CB1Rs before ethanol treatment at P7 relieved ERK1/2 but not AKT phosphorylation and prevented neurodegeneration. CB1R knock-out mice exhibited no ethanol-induced neurodegeneration and inhibition of ERK1/2 phosphorylation. The protective effects of CB1R blockade through pharmacological or genetic deletion resulted in normal adult synaptic plasticity and novel object recognition memory in mice exposed to ethanol at P7. The AEA/CB1R/pERK1/2 signaling pathway may be directly responsible for the synaptic and memory deficits associated with fetal alcohol spectrum disorders.
Collapse
|
41
|
Sadrian B, Wilson DA, Saito M. Long-lasting neural circuit dysfunction following developmental ethanol exposure. Brain Sci 2013; 3:704-27. [PMID: 24027632 PMCID: PMC3767176 DOI: 10.3390/brainsci3020704] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Revised: 04/10/2013] [Accepted: 04/23/2013] [Indexed: 01/14/2023] Open
Abstract
Fetal Alcohol Spectrum Disorder (FASD) is a general diagnosis for those exhibiting long-lasting neurobehavioral and cognitive deficiencies as a result of fetal alcohol exposure. It is among the most common causes of mental deficits today. Those impacted are left to rely on advances in our understanding of the nature of early alcohol-induced disorders toward human therapies. Research findings over the last decade have developed a model where ethanol-induced neurodegeneration impacts early neural circuit development, thereby perpetuating subsequent integration and plasticity in vulnerable brain regions. Here we review our current knowledge of FASD neuropathology based on discoveries of long-lasting neurophysiological effects of acute developmental ethanol exposure in animal models. We discuss the important balance between synaptic excitation and inhibition in normal neural network function, and relate the significance of that balance to human FASD as well as related disease states. Finally, we postulate that excitation/inhibition imbalance caused by early ethanol-induced neurodegeneration results in perturbed local and regional network signaling and therefore neurobehavioral pathology.
Collapse
Affiliation(s)
- Benjamin Sadrian
- Department of Child and Adolescent Psychiatry, New York University Langone School of Medicine, One Park Avenue, Eighth Floor, New York, NY 10128, USA; E-Mail:
- Nathan Kline Institute, 140 Old Orangeburg Road, Orangeburg, NY 10962, USA; E-Mail:
| | - Donald A. Wilson
- Department of Child and Adolescent Psychiatry, New York University Langone School of Medicine, One Park Avenue, Eighth Floor, New York, NY 10128, USA; E-Mail:
- Nathan Kline Institute, 140 Old Orangeburg Road, Orangeburg, NY 10962, USA; E-Mail:
| | - Mariko Saito
- Nathan Kline Institute, 140 Old Orangeburg Road, Orangeburg, NY 10962, USA; E-Mail:
- Department of Psychiatry, New York University Langone School of Medicine, One Park Avenue, Eighth Floor, New York, NY 10128, USA
| |
Collapse
|
42
|
Tiwari V, Chopra K. Resveratrol abrogates alcohol-induced cognitive deficits by attenuating oxidative-nitrosative stress and inflammatory cascade in the adult rat brain. Neurochem Int 2013; 62:861-9. [PMID: 23422878 DOI: 10.1016/j.neuint.2013.02.012] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Revised: 02/03/2013] [Accepted: 02/07/2013] [Indexed: 10/27/2022]
Abstract
Chronic alcohol intake is known to induce permanent cognitive deficits along with enhanced oxidative-nitrosative stress and activation of neuroinflammatory cascade. In the present study, we investigated the protective effect of resveratrol, a natural polyphenolic phytoalexin against chronic alcohol-induced cognitive dysfunction and neuroiflammatory cascade in the brain of adult rats chronically administered ethanol. Male Wistar rats were adminstered ethanol (10g/kg; oral gavage) for ten weeks and treated with resveratrol (5, 10 and 20mg/kg) for the same duration. Ethanol-exposed rats showed impaired spatial navigation in the Morris water maze test and poor retention in the elevated plus maze task which was coupled with enhanced acetylcholinesterase activity, increased oxidative-nitrosative stress, cytokines (TNF-alpha and IL-1beta), NF-kappa β and caspase-3 levels in different brain regions (cerebral cortex and hippocampus) of ethanol-treated rats. Co-administration with resveratrol significantly and dose-dependently prevented all the behavioral, biochemical and molecular deficits. Correlatively, the results of the present study revealed that treatment with resveratrol significantly prevented cognitive deficits induced by chronic ethanol exposure not only by modulating oxido-nitrosative stress but also by attenuating the enhanced levels of pro-inflammatory cytokines (TNF-α and IL-1β), NF-kβ and caspase-3 in different brain regions of ethanol treated rats. Therefore, mechanism underlying the neuroprotective effects of resveratrol observed in our study may be due to its antioxidant, anti-inflammatory and neuromodulating activities.
Collapse
Affiliation(s)
- Vinod Tiwari
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences, UGC Center of Advanced Study, Panjab University, Chandigarh 160 014, India
| | | |
Collapse
|
43
|
Subbanna S, Shivakumar M, Umapathy NS, Saito M, Mohan PS, Kumar A, Nixon RA, Verin AD, Psychoyos D, Basavarajappa BS. G9a-mediated histone methylation regulates ethanol-induced neurodegeneration in the neonatal mouse brain. Neurobiol Dis 2013; 54:475-85. [PMID: 23396011 DOI: 10.1016/j.nbd.2013.01.022] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Revised: 01/10/2013] [Accepted: 01/30/2013] [Indexed: 10/27/2022] Open
Abstract
Rodent exposure to binge-like ethanol during postnatal day 7 (P7), which is comparable to the third trimester of human pregnancy, induces neuronal cell loss. However, the molecular mechanisms underlying these neuronal losses are still poorly understood. Here, we tested the possibility of histone methylation mediated by G9a (lysine dimethyltransferase) in regulating neuronal apoptosis in P7 mice exposed to ethanol. G9a protein expression, which is higher during embryogenesis and synaptogenic period compared to adult brain, is entirely confined to the cell nuclei in the developing brain. We found that ethanol treatment at P7, which induces apoptotic neurodegeneration in neonatal mice, enhanced G9a activity followed by increased histone H3 lysine 9 (H3K9me2) and 27 (H3K27me2) dimethylation. In addition, it appears that increased dimethylation of H3K9 makes it susceptible to proteolytic degradation by caspase-3 in conditions in which ethanol induces neurodegeneration. Further, pharmacological inhibition of G9a activity prior to ethanol treatment at P7 normalized H3K9me2, H3K27me2 and total H3 proteins to basal levels and prevented neurodegeneration in neonatal mice. Together, these data demonstrate that G9a mediated histone H3K9 and K27 dimethylation critically regulates ethanol-induced neurodegeneration in the developing brain. Furthermore, these findings reveal a novel link between G9a and neurodegeneration in the developing brain exposed to postnatal ethanol and may have a role in fetal alcohol spectrum disorders.
Collapse
Affiliation(s)
- Shivakumar Subbanna
- Division of Analytical Psychopharmacology, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Tiwari V, Chopra K. Attenuation of oxidative stress, neuroinflammation, and apoptosis by curcumin prevents cognitive deficits in rats postnatally exposed to ethanol. Psychopharmacology (Berl) 2012; 224:519-35. [PMID: 22790976 DOI: 10.1007/s00213-012-2779-9] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2012] [Accepted: 06/13/2012] [Indexed: 10/28/2022]
Abstract
RATIONALE Clinical and experimental evidence have demonstrated that alcohol consumption during pregnancy can disrupt brain development, leading to a variety of behavioral alterations including hyperactivity, motor dysfunction, and cognitive deficits in offsprings. Alcohol-induced neurocognitive deficits are associated with activation of oxidative-inflammatory cascade coupled with extensive apoptotic neurodegeneration in different brain regions. OBJECTIVES The present study was designed with an aim to investigate the protective effect of curcumin, a principal curcuminoid present in the Indian spice turmeric, against alcohol-induced cognitive deficits, neuroinflammation, and neuronal apoptosis in rat pups postnatally exposed to ethanol. METHODS AND RESULTS Male Wistar rat pups were administered ethanol (5 g/kg, 12 % v/v) by intragastric intubation on postnatal days (PD) 7, 8, and 9 and were treated with curcumin (30 and 60 mg/kg) from PD 6 to 28. Performance of ethanol-exposed pups that did not receive curcumin was significantly impaired as evaluated in both Morris water maze and elevated plus maze tasks recorded by using computer tracking. Cognitive deficit was associated with enhanced acetylcholinesterase activity, increased neuroinflammation (oxidative-nitrosative stress, TNF-α, IL-1β, and TGF-β1), and neuronal apoptosis (NF-κβ and caspase 3) in both cerebral cortex and hippocampus of ethanol-exposed pups. Chronic treatment with curcumin significantly ameliorated all the behavioral, biochemical, and molecular alterations in different brain regions of ethanol-exposed pups. CONCLUSIONS The current study demonstrates the possible involvement of oxidative-inflammatory cascade-mediated apoptotic signaling in cognitive deficits associated with postnatal ethanol exposure and points towards the neuroprotective potential of curcumin in mitigating alcohol-induced behavioral, biochemical, and molecular deficits.
Collapse
Affiliation(s)
- Vinod Tiwari
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences, UGC Center of Advanced Study, Panjab University, Chandigarh 160014, India
| | | |
Collapse
|
45
|
Attenuation of NF-κβ mediated apoptotic signaling by tocotrienol ameliorates cognitive deficits in rats postnatally exposed to ethanol. Neurochem Int 2012; 61:310-20. [DOI: 10.1016/j.neuint.2012.05.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2012] [Revised: 05/02/2012] [Accepted: 05/09/2012] [Indexed: 01/27/2023]
|
46
|
Chopra K, Tiwari V. Alcoholic neuropathy: possible mechanisms and future treatment possibilities. Br J Clin Pharmacol 2012; 73:348-62. [PMID: 21988193 DOI: 10.1111/j.1365-2125.2011.04111.x] [Citation(s) in RCA: 145] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Chronic alcohol consumption produces painful peripheral neuropathy for which there is no reliable successful therapy, mainly due to lack of understanding of its pathobiology. Alcoholic neuropathy involves coasting caused by damage to nerves that results from long term excessive drinking of alcohol and is characterized by spontaneous burning pain, hyperalgesia and allodynia. The mechanism behind alcoholic neuropathy is not well understood, but several explanations have been proposed. These include activation of spinal cord microglia after chronic alcohol consumption, oxidative stress leading to free radical damage to nerves, activation of mGlu5 receptors in the spinal cord and activation of the sympathoadrenal and hypothalamo-pituitary-adrenal (HPA) axis. Nutritional deficiency (especially thiamine deficiency) and/or the direct toxic effect of alcohol or both have also been implicated in alcohol-induced neuropathic pain. Treatment is directed towards halting further damage to the peripheral nerves and restoring their normal functioning. This can be achieved by alcohol abstinence and a nutritionally balanced diet supplemented by all B vitamins. However, in the setting of ongoing alcohol use, vitamin supplementation alone has not been convincingly shown to be sufficient for improvement in most patients. The present review is focused around the multiple pathways involved in the development of peripheral neuropathy associated with chronic alcohol intake and the different therapeutic agents which may find a place in the therapeutic armamentarium for both prevention and management of alcoholic neuropathy.
Collapse
Affiliation(s)
- Kanwaljit Chopra
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences, UGC Center of Advanced Study, Panjab University, Chandigarh-160 014, India.
| | | |
Collapse
|
47
|
Sadrian B, Subbanna S, Wilson DA, Basavarajappa BS, Saito M. Lithium prevents long-term neural and behavioral pathology induced by early alcohol exposure. Neuroscience 2012; 206:122-35. [PMID: 22266347 DOI: 10.1016/j.neuroscience.2011.12.059] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2011] [Revised: 12/29/2011] [Accepted: 12/30/2011] [Indexed: 11/30/2022]
Abstract
Fetal alcohol exposure can cause developmental defects in offspring known as fetal alcohol spectrum disorder (FASD). FASD symptoms range from obvious facial deformities to changes in neuroanatomy and neurophysiology that disrupt normal brain function and behavior. Ethanol exposure at postnatal day 7 in C57BL/6 mice induces neuronal cell death and long-lasting neurobehavioral dysfunction. Previous work has demonstrated that early ethanol exposure impairs spatial memory task performance into adulthood and perturbs local and interregional brain circuit integrity in the olfacto-hippocampal pathway. Here we pursue these findings to examine whether lithium prevents anatomical, neurophysiological, and behavioral pathologies that result from early ethanol exposure. Lithium has neuroprotective properties that have been shown to prevent ethanol-induced apoptosis. Here we show that mice co-treated with lithium on the same day as ethanol exposure exhibit dramatically reduced acute neurodegeneration in the hippocampus and retain hippocampal-dependent spatial memory as adults. Lithium co-treatment also blocked ethanol-induced disruption in synaptic plasticity in slice recordings of hippocampal CA1 in the adult mouse brain. Moreover, long-lasting dysfunctions caused by ethanol in olfacto-hippocampal networks, including sensory-evoked oscillations and resting state coherence, were prevented in mice co-treated with lithium. Together, these results provide behavioral and physiological evidence that lithium is capable of preventing or reducing immediate and long-term deleterious consequences of early ethanol exposure on brain function.
Collapse
Affiliation(s)
- B Sadrian
- Emotional Brain Institute, Nathan S. Kline Institute for Psychiatric Research, 140 Old Orangeburg Road, Orangeburg, NY 10962, USA.
| | | | | | | | | |
Collapse
|
48
|
Zorumski CF, Izumi Y. NMDA receptors and metaplasticity: mechanisms and possible roles in neuropsychiatric disorders. Neurosci Biobehav Rev 2012; 36:989-1000. [PMID: 22230702 DOI: 10.1016/j.neubiorev.2011.12.011] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2011] [Revised: 11/14/2011] [Accepted: 12/22/2011] [Indexed: 02/08/2023]
Abstract
N-methyl-D-aspartate receptors (NMDARs) are key components of neural signaling, playing roles in synaptic transmission and in the synaptic plasticity thought to underlie learning and memory. NMDAR activation can also have neurotoxic consequences contributing to several forms of neurodegeneration. Additionally, NMDARs can modulate neuronal function and regulate the ability of synapses to undergo synaptic plasticity. Evidence gathered over the past 20 years strongly supports the idea that untimely activation of NMDARs impairs the induction of long-term potentiation (LTP) by a form of metaplasticity. This metaplasticity can be triggered by multiple stimuli including physiological receptor activation, and metabolic and behavioral stressors. These latter findings raise the possibility that NMDARs contribute to cognitive dysfunction associated with neuropsychiatric disorders. This paper examines NMDAR metaplasticity and its potential role in cognition. Recent studies using NMDAR antagonists for therapeutic purposes also raise the possibility that metaplasticity may contribute to clinical effects of certain drugs.
Collapse
Affiliation(s)
- Charles F Zorumski
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | | |
Collapse
|
49
|
Rubio J, Yucra S, Gasco M, Gonzales GF. Dose-response effect of black maca (Lepidium meyenii) in mice with memory impairment induced by ethanol. Toxicol Mech Methods 2011; 21:628-34. [PMID: 21780878 DOI: 10.3109/15376516.2011.583294] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Previous studies have shown that black variety of maca has beneficial effects on learning and memory in experimental animal models. The present study aimed to determine whether the hydroalcoholic extract of black maca (BM) showed a dose-response effect in mice treated with ethanol 20% (EtOH) as a model of memory impairment. Mice were divided in the following groups: control, EtOH, ascorbic acid (AA) and 0.125, 0.25, 0.50 and 1.00 g/kg of BM plus EtOH. All treatments were orally administered for 28 days. Open field test was performed to determine locomotor activity and water Morris maze was done to determine spatial memory. Also, total polyphenol content in the hydroalcoholic extract of BM was determined (0.65 g pyrogallol/100 g). Mice treated with EtOH took more time to find the hidden platform than control during escape acquisition trials; meanwhile, AA and BM reversed the effect of EtOH. In addition, AA and BM ameliorated the deleterious effect of EtOH during the probe trial. Correlation analyses showed that the effect of BM a dose-dependent behavior. Finally, BM improved experimental memory impairment induced by ethanol in a dose-response manner due, in part, to its content of polyphenolic compounds.
Collapse
Affiliation(s)
- Julio Rubio
- Department of Biological and Physiological Sciences, Faculty of Sciences and Philosophy and Instituto de Investigaciones de la Altura, Universidad Peruana Cayetano Heredia , Lima , Peru.
| | | | | | | |
Collapse
|
50
|
Zink M, Ferbert T, Frank ST, Seufert P, Gebicke-Haerter PJ, Spanagel R. Perinatal exposure to alcohol disturbs spatial learning and glutamate transmission-related gene expression in the adult hippocampus. Eur J Neurosci 2011; 34:457-68. [DOI: 10.1111/j.1460-9568.2011.07776.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|