1
|
Gao DP, Weng QY, Zhang YY, Ou YX, Niu YF, Lou Q, Xie DL, Cai Y, Yang JH. Memantine alleviates cognitive impairment and hippocampal morphology injury in a mouse model of chronic alcohol exposure. Pharmacol Biochem Behav 2024; 243:173827. [PMID: 39038728 DOI: 10.1016/j.pbb.2024.173827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 07/13/2024] [Accepted: 07/16/2024] [Indexed: 07/24/2024]
Abstract
Alcohol-related cognitive impairment (ARCI) is highly prevalent among patients with alcohol abuse and dependence. The pathophysiology of ARCI, pivotal for refined therapeutic approaches, is not fully elucidated, posing a risk of progression to severe neurological sequelae such as Korsakoff's syndrome (KS) and Alcohol-Related Dementia (ARD). This study ventures into the underlying mechanisms of chronic alcohol-induced neurotoxicity, notably glutamate excitotoxicity and cytoskeletal disruption, and explores the therapeutic potential of Memantine, a non-competitive antagonist of the N-methyl-d-aspartate (NMDA) receptor known for its neuroprotective effect against excitotoxicity. Our investigation centers on the efficacy of Memantine in mitigating chronic alcohol-induced cognitive and hippocampal damages in vivo. Male C57BL/6J mice were subjected to 30 % (v/v, 6.0 g/kg) ethanol via intragastric administration alongside Memantine co-treatment (10 mg/kg/day, intraperitoneally) for six weeks. The assessment involved Y maze, Morris water maze, and novel object recognition tests to evaluate spatial and recognition memory deficits. Histopathological evaluations of the hippocampus were conducted to examine the extent of alcohol-induced morphological changes and the potential protective effect of Memantine. The findings reveal that Memantine significantly improves chronic alcohol-compromised cognitive functions and mitigates hippocampal pathological changes, implicating a moderating effect on the disassembly of actin cytoskeleton and microtubules in the hippocampus, induced by chronic alcohol exposure. Our results underscore Memantine's capability to attenuate chronic alcohol-induced cognitive and hippocampal morphological harm may partly through regulating cytoskeleton dynamics, offering valuable insights into innovative therapeutic strategies for ARCI.
Collapse
Affiliation(s)
- Da-Peng Gao
- Department of Neurology, The First Affiliated Hospital of Ningbo University, 247 Renmin Rd, Ningbo, Zhejiang 315020, PR China
| | - Qiu-Yan Weng
- Department of Neurology, The First Affiliated Hospital of Ningbo University, 247 Renmin Rd, Ningbo, Zhejiang 315020, PR China
| | - Yun-Yun Zhang
- Department of Neurology, The First Affiliated Hospital of Ningbo University, 247 Renmin Rd, Ningbo, Zhejiang 315020, PR China
| | - Yang-Xin Ou
- Department of Neurology, The First Affiliated Hospital of Ningbo University, 247 Renmin Rd, Ningbo, Zhejiang 315020, PR China
| | - Yan-Fang Niu
- Department of Neurology, The First Affiliated Hospital of Ningbo University, 247 Renmin Rd, Ningbo, Zhejiang 315020, PR China
| | - Qiong Lou
- Department of Neurology, The First Affiliated Hospital of Ningbo University, 247 Renmin Rd, Ningbo, Zhejiang 315020, PR China
| | - Dong-Lin Xie
- Department of Neurology, The First Affiliated Hospital of Ningbo University, 247 Renmin Rd, Ningbo, Zhejiang 315020, PR China
| | - Yu Cai
- Department of Pharmacy, Zhejiang Pharmaceutical University, 666 Siming Rd, Ningbo, Zhejiang 315500, PR China.
| | - Jian-Hong Yang
- Department of Neurology, The First Affiliated Hospital of Ningbo University, 247 Renmin Rd, Ningbo, Zhejiang 315020, PR China.
| |
Collapse
|
2
|
Bhandari A, Seguin A, Rothenfluh A. Synaptic Mechanisms of Ethanol Tolerance and Neuroplasticity: Insights from Invertebrate Models. Int J Mol Sci 2024; 25:6838. [PMID: 38999947 PMCID: PMC11241699 DOI: 10.3390/ijms25136838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 06/09/2024] [Accepted: 06/10/2024] [Indexed: 07/14/2024] Open
Abstract
Alcohol tolerance is a neuroadaptive response that leads to a reduction in the effects of alcohol caused by previous exposure. Tolerance plays a critical role in the development of alcohol use disorder (AUD) because it leads to the escalation of drinking and dependence. Understanding the molecular mechanisms underlying alcohol tolerance is therefore important for the development of effective therapeutics and for understanding addiction in general. This review explores the molecular basis of alcohol tolerance in invertebrate models, Drosophila and C. elegans, focusing on synaptic transmission. Both organisms exhibit biphasic responses to ethanol and develop tolerance similar to that of mammals. Furthermore, the availability of several genetic tools makes them a great candidate to study the molecular basis of ethanol response. Studies in invertebrate models show that tolerance involves conserved changes in the neurotransmitter systems, ion channels, and synaptic proteins. These neuroadaptive changes lead to a change in neuronal excitability, most likely to compensate for the enhanced inhibition by ethanol.
Collapse
Affiliation(s)
- Aakriti Bhandari
- Department of Psychiatry, University of Utah, Salt Lake City, UT 84112, USA
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA
- Neuroscience Graduate Program, University of Utah, Salt Lake City, UT 84112, USA
| | - Alexandra Seguin
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA
| | - Adrian Rothenfluh
- Department of Psychiatry, University of Utah, Salt Lake City, UT 84112, USA
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA
- Neuroscience Graduate Program, University of Utah, Salt Lake City, UT 84112, USA
- Department of Neurobiology, University of Utah, Salt Lake City, UT 84112, USA
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
3
|
Pandey S, Miller CA. Targeting the cytoskeleton as a therapeutic approach to substance use disorders. Pharmacol Res 2024; 202:107143. [PMID: 38499081 PMCID: PMC11034636 DOI: 10.1016/j.phrs.2024.107143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 03/06/2024] [Accepted: 03/12/2024] [Indexed: 03/20/2024]
Abstract
Substance use disorders (SUD) are chronic relapsing disorders governed by continually shifting cycles of positive drug reward experiences and drug withdrawal-induced negative experiences. A large body of research points to plasticity within systems regulating emotional, motivational, and cognitive processes as drivers of continued compulsive pursuit and consumption of substances despite negative consequences. This plasticity is observed at all levels of analysis from molecules to networks, providing multiple avenues for intervention in SUD. The cytoskeleton and its regulatory proteins within neurons and glia are fundamental to the structural and functional integrity of brain processes and are potentially the major drivers of the morphological and behavioral plasticity associated with substance use. In this review, we discuss preclinical studies that provide support for targeting the brain cytoskeleton as a therapeutic approach to SUD. We focus on the interplay between actin cytoskeleton dynamics and exposure to cocaine, methamphetamine, alcohol, opioids, and nicotine and highlight preclinical studies pointing to a wide range of potential therapeutic targets, such as nonmuscle myosin II, Rac1, cofilin, prosapip 1, and drebrin. These studies broaden our understanding of substance-induced plasticity driving behaviors associated with SUD and provide new research directions for the development of SUD therapeutics.
Collapse
Affiliation(s)
- Surya Pandey
- Department of Molecular Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL 33458, United States; Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL 33458, United States
| | - Courtney A Miller
- Department of Molecular Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL 33458, United States; Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL 33458, United States.
| |
Collapse
|
4
|
Chvilicek MM, Seguin A, Lathen DR, Titos I, Cummins‐Beebee PN, Pabon MA, Miščević M, Nickel E, Merrill CB, Rodan AR, Rothenfluh A. Large analysis of genetic manipulations reveals an inverse correlation between initial alcohol resistance and rapid tolerance phenotypes. GENES, BRAIN, AND BEHAVIOR 2024; 23:e12884. [PMID: 38968320 PMCID: PMC10825885 DOI: 10.1111/gbb.12884] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 01/04/2024] [Accepted: 01/06/2024] [Indexed: 07/07/2024]
Abstract
Tolerance occurs when, following an initial experience with a substance, more of the substance is required subsequently to induce identical behavioral effects. Tolerance is not well-understood, and numerous researchers have turned to model organisms, particularly Drosophila melanogaster, to unravel its mechanisms. Flies have high translational relevance for human alcohol responses, and there is substantial overlap in disease-causing genes between flies and humans, including those associated with Alcohol Use Disorder. Numerous Drosophila tolerance mutants have been described; however, approaches used to identify and characterize these mutants have varied across time and labs and have mostly disregarded any impact of initial resistance/sensitivity to ethanol on subsequent tolerance development. Here, we analyzed our own, as well as data published by other labs to uncover an inverse correlation between initial ethanol resistance and tolerance phenotypes. This inverse correlation suggests that initial resistance phenotypes can explain many 'perceived' tolerance phenotypes, thus classifying such mutants as 'secondary' tolerance mutants. Additionally, we show that tolerance should be measured as a relative increase in time to sedation between an initial and second exposure rather than an absolute change in time to sedation. Finally, based on our analysis, we provide a method for using a linear regression equation to assess the residuals of potential tolerance mutants. These residuals provide predictive insight into the likelihood of a mutant being a 'primary' tolerance mutant, where a tolerance phenotype is not solely a consequence of initial resistance, and we offer a framework for understanding the relationship between initial resistance and tolerance.
Collapse
Affiliation(s)
- Maggie M. Chvilicek
- Department of Psychiatry, Huntsman Mental Health Institute, School of MedicineUniversity of UtahSalt Lake CityUtahUSA
- Neuroscience Graduate ProgramUniversity of UtahSalt Lake CityUtahUSA
| | - Alexandra Seguin
- Molecular Medicine Program, School of MedicineUniversity of UtahSalt Lake CityUtahUSA
| | - Daniel R. Lathen
- Department of Psychiatry, Huntsman Mental Health Institute, School of MedicineUniversity of UtahSalt Lake CityUtahUSA
- Neuroscience Graduate ProgramUniversity of UtahSalt Lake CityUtahUSA
| | - Iris Titos
- Molecular Medicine Program, School of MedicineUniversity of UtahSalt Lake CityUtahUSA
| | - Pearl N. Cummins‐Beebee
- Department of Psychiatry, Huntsman Mental Health Institute, School of MedicineUniversity of UtahSalt Lake CityUtahUSA
- Neuroscience Graduate ProgramUniversity of UtahSalt Lake CityUtahUSA
| | - Miguel A. Pabon
- Molecular Medicine Program, School of MedicineUniversity of UtahSalt Lake CityUtahUSA
| | - Maša Miščević
- Molecular Medicine Program, School of MedicineUniversity of UtahSalt Lake CityUtahUSA
- Present address:
Department of Neuroscience, Physiological Sciences Graduate Interdisciplinary ProgramUniversity of ArizonaTucsonArizonaUSA
| | - Emily Nickel
- Molecular Medicine Program, School of MedicineUniversity of UtahSalt Lake CityUtahUSA
| | - Collin B. Merrill
- Department of Psychiatry, Huntsman Mental Health Institute, School of MedicineUniversity of UtahSalt Lake CityUtahUSA
| | - Aylin R. Rodan
- Molecular Medicine Program, School of MedicineUniversity of UtahSalt Lake CityUtahUSA
- Division of Nephrology, Department of Internal Medicine, School of MedicineUniversity of UtahSalt Lake CityUtahUSA
- Medical ServiceVeterans Affairs Salt Lake City Health Care SystemSalt Lake CityUtahUSA
- Department of Human Genetics, School of MedicineUniversity of UtahSalt Lake CityUtahUSA
| | - Adrian Rothenfluh
- Department of Psychiatry, Huntsman Mental Health Institute, School of MedicineUniversity of UtahSalt Lake CityUtahUSA
- Neuroscience Graduate ProgramUniversity of UtahSalt Lake CityUtahUSA
- Molecular Medicine Program, School of MedicineUniversity of UtahSalt Lake CityUtahUSA
- Department of Human Genetics, School of MedicineUniversity of UtahSalt Lake CityUtahUSA
- Department of Neurobiology, School of MedicineUniversity of UtahSalt Lake CityUtahUSA
| |
Collapse
|
5
|
Chvilicek MM, Seguin A, Lathen DR, Titos I, Cummins-Beebe PN, Pabon MA, Miscevic M, Nickel EA, Merrill CB, Rodan AR, Rothenfluh A. Large genetic analysis of alcohol resistance and tolerance reveals an inverse correlation and suggests 'true' tolerance mutants. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.09.561599. [PMID: 37873285 PMCID: PMC10592763 DOI: 10.1101/2023.10.09.561599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Tolerance occurs when, following an initial experience with a substance, more of the substance is required subsequently to induce the same behavioral effects. Tolerance is historically not well-understood, and numerous researchers have turned to model organisms, particularly Drosophila melanogaster, to unravel its mechanisms. Flies have high translational relevance for human alcohol responses, and there is substantial overlap in disease-causing genes between flies and humans, including those associated with Alcohol Use Disorder. Numerous Drosophila tolerance mutants have been described; however, approaches used to identify and characterize these mutants have varied across time and between labs and have mostly disregarded any impact of initial resistance/sensitivity to ethanol on subsequent tolerance development. Here, we have analyzed a large amount of data - our own published and unpublished data and data published by other labs - to uncover an inverse correlation between initial ethanol resistance and tolerance phenotypes. This inverse correlation suggests that initial resistance phenotypes can explain many 'perceived' tolerance phenotypes. Additionally, we show that tolerance should be measured as a relative increase in time to sedation between an initial and second exposure rather than an absolute change in time to sedation. Finally, based on our analysis, we provide a method for using a linear regression equation to assess the residuals of potential tolerance mutants. We show that these residuals provide predictive insight into the likelihood of a mutant being a 'true' tolerance mutant, and we offer a framework for understanding the relationship between initial resistance and tolerance.
Collapse
Affiliation(s)
- Maggie M. Chvilicek
- Department of Psychiatry, Huntsman Mental Health Institute, School of Medicine, University of Utah, Salt Lake City, USA
- Neuroscience Graduate Program, University of Utah, Salt Lake City, USA
| | - Alexandra Seguin
- Molecular Medicine Program, School of Medicine, University of Utah, Salt Lake City, USA
| | - Daniel R. Lathen
- Department of Psychiatry, Huntsman Mental Health Institute, School of Medicine, University of Utah, Salt Lake City, USA
- Neuroscience Graduate Program, University of Utah, Salt Lake City, USA
| | - Iris Titos
- Department of Psychiatry, Huntsman Mental Health Institute, School of Medicine, University of Utah, Salt Lake City, USA
| | - Pearl N Cummins-Beebe
- Department of Psychiatry, Huntsman Mental Health Institute, School of Medicine, University of Utah, Salt Lake City, USA
- Neuroscience Graduate Program, University of Utah, Salt Lake City, USA
| | - Miguel A. Pabon
- Molecular Medicine Program, School of Medicine, University of Utah, Salt Lake City, USA
| | - Masa Miscevic
- Molecular Medicine Program, School of Medicine, University of Utah, Salt Lake City, USA
| | - Emily A. Nickel
- Molecular Medicine Program, School of Medicine, University of Utah, Salt Lake City, USA
| | - Collin B Merrill
- Department of Psychiatry, Huntsman Mental Health Institute, School of Medicine, University of Utah, Salt Lake City, USA
| | - Aylin R. Rodan
- Molecular Medicine Program, School of Medicine, University of Utah, Salt Lake City, USA
- Division of Nephrology, Department of Internal Medicine, School of Medicine, University of Utah, Salt Lake City, USA
- Medical Service, Veterans Affairs Salt Lake City Health Care System, Salt Lake City, USA
- Department of Human Genetics, School of Medicine, University of Utah, Salt Lake City, USA
| | - Adrian Rothenfluh
- Department of Psychiatry, Huntsman Mental Health Institute, School of Medicine, University of Utah, Salt Lake City, USA
- Neuroscience Graduate Program, University of Utah, Salt Lake City, USA
- Molecular Medicine Program, School of Medicine, University of Utah, Salt Lake City, USA
- Department of Human Genetics, School of Medicine, University of Utah, Salt Lake City, USA
- Department of Neurobiology, School of Medicine, University of Utah, Salt Lake City, USA
| |
Collapse
|
6
|
Nuñez KM, Catalano JL, Scaplen KM, Kaun KR. Ethanol Behavioral Responses in Drosophila. Cold Spring Harb Protoc 2023; 2023:719-24. [PMID: 37019606 PMCID: PMC10551053 DOI: 10.1101/pdb.top107887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2023]
Abstract
Drosophila melanogaster is a powerful genetic model for investigating the mechanisms underlying ethanol-induced behaviors, metabolism, and preference. Ethanol-induced locomotor activity is especially useful for understanding the mechanisms by which ethanol acutely affects the brain and behavior. Ethanol-induced locomotor activity is characterized by hyperlocomotion and subsequent sedation with increased exposure duration or concentration. Locomotor activity is an efficient, easy, robust, and reproducible behavioral screening tool for identifying underlying genes and neuronal circuits as well as investigating genetic and molecular pathways. We introduce a detailed protocol for performing experiments investigating how volatilized ethanol affects locomotor activity using the fly Group Activity Monitor (flyGrAM). We introduce installation, implementation, data collection, and subsequent data-analysis methods for investigating how volatilized stimuli affect activity. We also introduce a procedure for how to optogenetically probe neuronal activity to identify the neural mechanisms underlying locomotor activity.
Collapse
Affiliation(s)
- Kavin M Nuñez
- Molecular Pharmacology and Physiology Graduate Program, Brown University, Providence, Rhode Island 02912, USA
| | - Jamie L Catalano
- Molecular Pharmacology and Physiology Graduate Program, Brown University, Providence, Rhode Island 02912, USA
| | - Kristin M Scaplen
- Department of Psychology, Bryant University, Smithfield, Rhode Island 02917, USA
- Center for Health and Behavioral Sciences, Bryant University, Smithfield, Rhode Island 02917, USA
- Department of Neuroscience, Brown University, Providence, Rhode Island 02912, USA
| | - Karla R Kaun
- Department of Neuroscience, Brown University, Providence, Rhode Island 02912, USA
| |
Collapse
|
7
|
Korczyńska J, Szczuka A, Urzykowska J, Kochanowski M, Andrzejczyk NG, Piwowarek KJ, Godzińska EJ. The Effects of Ethanol and Acetic acid on Behaviour of Extranidal Workers of the Narrow-Headed Ant Formica exsecta (Hymenoptera, Formicidae) during a Field Experiment. Animals (Basel) 2023; 13:2734. [PMID: 37684998 PMCID: PMC10486794 DOI: 10.3390/ani13172734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 08/20/2023] [Accepted: 08/23/2023] [Indexed: 09/10/2023] Open
Abstract
Ethanol addiction belongs to the most important problems encountered in the domain of human mental health. The research on the behavioural effects of exposure to/consumption of ethanol are investigated largely with the help of animal models that also include insects, mainly fruit flies and honeybees. The effects of ethanol on ant behaviour remain, however, little known. In the present field study, we investigated the behaviour of workers of the narrow-headed ant (Formica exsecta) displayed in the vicinity of cotton pads soaked in water or in water solutions of ethanol or acetic acid during 5 min tests (n = 30 tests in each group). Both ethanol and acetic acid induced significant modifications of ant locomotion, exploratory behaviour, self-grooming behaviour, and aggressive social behaviour. We confirmed that acetic acid is aversive for the ants, but ethanol enhances their exploratory behaviour. We also found out that field studies may document more types of responses to experimental compounds than laboratory ones, as the tested animals may also escape from aversive substances. Our findings documented a wide spectrum of behavioural effects of exposure to ethanol and acetic acid in a highly social animal species and broadened the general knowledge about behavioural responses to these compounds encountered in animals.
Collapse
Affiliation(s)
- Julita Korczyńska
- Laboratory of Ethology, Nencki Institute of Experimental Biology PAS, Ludwika Pasteura 3, PL 02-093 Warsaw, Poland; (J.K.); (A.S.); (J.U.)
| | - Anna Szczuka
- Laboratory of Ethology, Nencki Institute of Experimental Biology PAS, Ludwika Pasteura 3, PL 02-093 Warsaw, Poland; (J.K.); (A.S.); (J.U.)
| | - Julia Urzykowska
- Laboratory of Ethology, Nencki Institute of Experimental Biology PAS, Ludwika Pasteura 3, PL 02-093 Warsaw, Poland; (J.K.); (A.S.); (J.U.)
- Faculty of Biology, University of Warsaw, Ilji Miecznikowa 1, PL 02-096 Warsaw, Poland
| | - Michał Kochanowski
- Botanic Garden, University of Warsaw, Aleje Ujazdowskie 4, PL 00-478 Warsaw, Poland;
| | - Neptun Gabriela Andrzejczyk
- Laboratory of Ethology, Nencki Institute of Experimental Biology PAS, Ludwika Pasteura 3, PL 02-093 Warsaw, Poland; (J.K.); (A.S.); (J.U.)
- Department of Animal Physiology, Institute of Functional Biology and Ecology, Faculty of Biology, University of Warsaw, Ilji Miecznikowa 1, PL 02-096 Warsaw, Poland; (N.G.A.); (K.J.P.)
| | - Kacper Jerzy Piwowarek
- Laboratory of Ethology, Nencki Institute of Experimental Biology PAS, Ludwika Pasteura 3, PL 02-093 Warsaw, Poland; (J.K.); (A.S.); (J.U.)
- Department of Animal Physiology, Institute of Functional Biology and Ecology, Faculty of Biology, University of Warsaw, Ilji Miecznikowa 1, PL 02-096 Warsaw, Poland; (N.G.A.); (K.J.P.)
| | - Ewa Joanna Godzińska
- Laboratory of Ethology, Nencki Institute of Experimental Biology PAS, Ludwika Pasteura 3, PL 02-093 Warsaw, Poland; (J.K.); (A.S.); (J.U.)
| |
Collapse
|
8
|
Pagano R, Salamian A, Skonieczna E, Wojtas B, Gielniewski B, Harda Z, Cały A, Havekes R, Abel T, Radwanska K. Molecular fingerprints in the hippocampus of alcohol seeking during withdrawal. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.24.554622. [PMID: 37662388 PMCID: PMC10473700 DOI: 10.1101/2023.08.24.554622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Alcohol use disorder (AUD) is characterized by excessive alcohol seeking and use. Here, we investigated the molecular correlates of impaired extinction of alcohol seeking using a multidimentional mouse model of AUD. We distinguished AUD-prone and AUD-resistant mice, based on the presence of ≥ 2 or < 2 criteria of AUD and utilized RNA sequencing to identify genes that were differentially expressed in the hippocampus and amygdala of mice meeting ≥ 2 or < 2 criteria, as these brain regions are implicated in alcohol motivation, seeking, consumption and the cognitive inflexibility characteristic of AUD. Our findings revealed dysregulation of the genes associated with the actin cytoskeleton, including actin binding molecule cofilin, and impaired synaptic transmission in the hippocampi of mice meeting ≥ 2 criteria. Overexpression of cofilin in the polymorphic layer of the dentate gyrus (PoDG) inhibited ML-DG synapses, increased motivation to seek alcohol and impaired extinction of alcohol seeking, resembling the phenotype observed in mice meeting ≥ 2 criteria. Overall, our study uncovers a novel mechanism linking increased hippocampal cofilin expression with the AUD phenotype.
Collapse
Affiliation(s)
- Roberto Pagano
- Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur St., Warsaw 02-093, Poland
| | - Ahmad Salamian
- Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur St., Warsaw 02-093, Poland
| | - Edyta Skonieczna
- Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur St., Warsaw 02-093, Poland
| | - Bartosz Wojtas
- Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur St., Warsaw 02-093, Poland
| | - Bartek Gielniewski
- Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur St., Warsaw 02-093, Poland
| | - Zofia Harda
- Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur St., Warsaw 02-093, Poland
- current address: Department Molecular Neuropharmacology, Maj Institute of Pharmacology of Polish Academy of Sciences, Krakow, Poland
| | - Anna Cały
- Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur St., Warsaw 02-093, Poland
| | - Robbert Havekes
- Neurobiology expertise group, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, The Netherlands
| | - Ted Abel
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, IA, USA; Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Kasia Radwanska
- Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur St., Warsaw 02-093, Poland
| |
Collapse
|
9
|
Cummins-Beebee PN, Chvilicek MM, Rothenfluh A. The Stage-Based Model of Addiction-Using Drosophila to Investigate Alcohol and Psychostimulant Responses. Int J Mol Sci 2023; 24:10909. [PMID: 37446084 PMCID: PMC10341944 DOI: 10.3390/ijms241310909] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 06/23/2023] [Accepted: 06/25/2023] [Indexed: 07/15/2023] Open
Abstract
Addiction is a progressive and complex disease that encompasses a wide range of disorders and symptoms, including substance use disorder (SUD), for which there are few therapeutic treatments. SUD is the uncontrolled and chronic use of substances despite the negative consequences resulting from this use. The progressive nature of addiction is organized into a testable framework, the neurobiological stage-based model, that includes three behavioral stages: (1) binge/intoxication, (2) withdrawal/negative affect, and (3) preoccupation/anticipation. Human studies offer limited opportunities for mechanistic insights into these; therefore, model organisms, like Drosophila melanogaster, are necessary for understanding SUD. Drosophila is a powerful model organism that displays a variety of SUD-like behaviors consistent with human and mammalian substance use, making flies a great candidate to study mechanisms of behavior. Additionally, there are an abundance of genetic tools like the GAL4/UAS and CRISPR/Cas9 systems that can be used to gain insight into the molecular mechanisms underlying the endophenotypes of the three-stage model. This review uses the three-stage framework and discusses how easily testable endophenotypes have been examined with experiments using Drosophila, and it outlines their potential for investigating other endophenotypes.
Collapse
Affiliation(s)
- Pearl N. Cummins-Beebee
- Department of Psychiatry, University of Utah, Salt Lake City, UT 84112, USA
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA
- Neuroscience Graduate Program, University of Utah, Salt Lake City, UT 84112, USA
| | - Maggie M. Chvilicek
- Department of Psychiatry, University of Utah, Salt Lake City, UT 84112, USA
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA
- Neuroscience Graduate Program, University of Utah, Salt Lake City, UT 84112, USA
| | - Adrian Rothenfluh
- Department of Psychiatry, University of Utah, Salt Lake City, UT 84112, USA
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA
- Neuroscience Graduate Program, University of Utah, Salt Lake City, UT 84112, USA
- Department of Neurobiology, University of Utah, Salt Lake City, UT 84112, USA
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
10
|
Ru Q, Wang Y, Zhou E, Chen L, Wu Y. The potential therapeutic roles of Rho GTPases in substance dependence. Front Mol Neurosci 2023; 16:1125277. [PMID: 37063367 PMCID: PMC10097952 DOI: 10.3389/fnmol.2023.1125277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 03/14/2023] [Indexed: 04/03/2023] Open
Abstract
Rho GTPases family are considered to be molecular switches that regulate various cellular processes, including cytoskeleton remodeling, cell polarity, synaptic development and maintenance. Accumulating evidence shows that Rho GTPases are involved in neuronal development and brain diseases, including substance dependence. However, the functions of Rho GTPases in substance dependence are divergent and cerebral nuclei-dependent. Thereby, comprehensive integration of their roles and correlated mechanisms are urgently needed. In this review, the molecular functions and regulatory mechanisms of Rho GTPases and their regulators such as GTPase-activating proteins (GAPs) and guanine nucleotide exchange factors (GEFs) in substance dependence have been reviewed, and this is of great significance for understanding their spatiotemporal roles in addictions induced by different addictive substances and in different stages of substance dependence.
Collapse
Affiliation(s)
| | | | | | - Lin Chen
- *Correspondence: Lin Chen, ; Yuxiang Wu,
| | - Yuxiang Wu
- *Correspondence: Lin Chen, ; Yuxiang Wu,
| |
Collapse
|
11
|
Wu X, Fan X, McMullen MR, Miyata T, Kim A, Pathak V, Wu J, Day LZ, Hardesty JE, Welch N, Dasarathy J, Allende DS, McCullough AJ, Jacobs JM, Rotroff DM, Dasarathy S, Nagy LE. Macrophage-derived MLKL in alcohol-associated liver disease: Regulation of phagocytosis. Hepatology 2023; 77:902-919. [PMID: 35689613 PMCID: PMC9741663 DOI: 10.1002/hep.32612] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 05/20/2022] [Accepted: 05/24/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND AIMS Mixed lineage kinase domain-like pseudokinase (MLKL), a key terminal effector of necroptosis, also plays a role in intracellular vesicle trafficking that is critical for regulating liver inflammation and injury in alcohol-associated liver disease (ALD). Although receptor interacting protein kinase 3 (Rip3)-/- mice are completely protected from ethanol-induced liver injury, Mlkl-/- mice are only partially protected. Therefore, we hypothesized that cell-specific functions of MLKL may contribute to ethanol-induced injury. APPROACH AND RESULTS Bone marrow transplants between Mlkl-/- mice and littermates were conducted to distinguish the role of myeloid versus nonmyeloid Mlkl in the Gao-binge model of ALD. Ethanol-induced hepatic injury, steatosis, and inflammation were exacerbated in Mlkl-/- →wild-type (WT) mice, whereas Mlkl deficiency in nonmyeloid cells (WT→ Mlkl-/- ) had no effect on Gao-binge ethanol-induced injury. Importantly, Mlkl deficiency in myeloid cells exacerbated ethanol-mediated bacterial burden and accumulation of immune cells in livers. Mechanistically, challenging macrophages with lipopolysaccharide (LPS) induced signal transducer and activator of transcription 1-mediated expression and phosphorylation of MLKL, as well as translocation and oligomerization of MLKL to intracellular compartments, including phagosomes and lysosomes but not plasma membrane. Importantly, pharmacological or genetic inhibition of MLKL suppressed the phagocytic capability of primary mouse Kupffer cells (KCs) at baseline and in response to LPS with/without ethanol as well as peripheral monocytes isolated from both healthy controls and patients with alcohol-associated hepatitis. Further, in vivo studies revealed that KCs of Mlkl-/- mice phagocytosed fewer bioparticles than KCs of WT mice. CONCLUSION Together, these data indicate that myeloid MLKL restricts ethanol-induced liver inflammation and injury by regulating hepatic immune cell homeostasis and macrophage phagocytosis.
Collapse
Affiliation(s)
- Xiaoqin Wu
- Northern Ohio Alcohol Center, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA
| | - Xiude Fan
- Northern Ohio Alcohol Center, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA
| | - Megan R. McMullen
- Northern Ohio Alcohol Center, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA
| | - Tatsunori Miyata
- Northern Ohio Alcohol Center, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA
| | - Adam Kim
- Northern Ohio Alcohol Center, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA
| | - Vai Pathak
- Northern Ohio Alcohol Center, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA
| | - Jianguo Wu
- Northern Ohio Alcohol Center, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Molecular Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Le Z. Day
- Biological Sciences Division and Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Josiah E. Hardesty
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Louisville, Louisville, Kentucky, USA
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Nicole Welch
- Northern Ohio Alcohol Center, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Gastroenterology and Hepatology, Cleveland Clinic, Cleveland, Ohio, USA
| | - Jaividhya Dasarathy
- Department of Family Medicine, Metro Health Medical Center, Case Western Reserve University, Cleveland, Ohio, USA
| | | | - Arthur J. McCullough
- Department of Gastroenterology and Hepatology, Cleveland Clinic, Cleveland, Ohio, USA
| | - Jon M. Jacobs
- Biological Sciences Division and Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Daniel M. Rotroff
- Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio, USA
- Endocrine and Metabolism Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Srinivasan Dasarathy
- Northern Ohio Alcohol Center, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Molecular Medicine, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Gastroenterology and Hepatology, Cleveland Clinic, Cleveland, Ohio, USA
| | - Laura E. Nagy
- Northern Ohio Alcohol Center, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Molecular Medicine, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Gastroenterology and Hepatology, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
12
|
Lei F, Xu X, Huang J, Su D, Wan P. Drosophila RhoGAP18B regulates actin cytoskeleton during border cell migration. PLoS One 2023; 18:e0280652. [PMID: 36662713 PMCID: PMC9858088 DOI: 10.1371/journal.pone.0280652] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 01/04/2023] [Indexed: 01/21/2023] Open
Abstract
Drosophila RhoGAP18B was identified as a negative regulator of small GTPase in the behavioral response to ethanol. However, the effect of RhoGAP18B on cell migration is unknown. Here, we report that RhoGAP18B regulates the migration of border cells in Drosophila ovary. The RhoGAP18B gene produces four transcripts and encodes three translation isoforms. We use different RNAi lines to knockdown each RhoGAP18B isoform, and find that knockdown of RhoGAP18B-PA, but not PC or PD isoform, blocks border cell migration. Knockdown of RhoGAP18B-PA disrupts the asymmetric distribution of F-actin in border cell cluster and increases F-actin level. Furthermore, RhoGAP18B-PA may act on Rac to regulate F-actin organization. Our data indicate that RhoGAP18B shows isoform-specific regulation of border cell migration.
Collapse
Affiliation(s)
- Fengyun Lei
- Laboratory of Molecular Biology, School of Life Science, Jiangxi Science and Technology Normal University, Nanchang, China
| | - Xiaoqing Xu
- Laboratory of Molecular Biology, School of Life Science, Jiangxi Science and Technology Normal University, Nanchang, China
| | - Jianhua Huang
- Laboratory of Molecular Biology, School of Life Science, Jiangxi Science and Technology Normal University, Nanchang, China
| | - Dan Su
- Key Laboratory of Animal Model of TCM Syndromes of Depression, Jiangxi Administration of traditional Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Ping Wan
- Laboratory of Molecular Biology, School of Life Science, Jiangxi Science and Technology Normal University, Nanchang, China
| |
Collapse
|
13
|
Liu D, Liu S, Li J, Liu X, Wu X, Peng Y, Shen Q. Proteome-Wide Analysis of the Hippocampus in Adult Mice with Learning and Memory Impairment Caused by Chronic Ethanol Exposure. Neurobiol Learn Mem 2022; 194:107661. [PMID: 35878712 DOI: 10.1016/j.nlm.2022.107661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 07/06/2022] [Accepted: 07/17/2022] [Indexed: 11/30/2022]
Abstract
Alcohol consumption may cause various impairments in the brain. The hippocampus is particularly vulnerable to alcohol exposure, which may cause learning and memory deficits. Recently, proteomics analysis has become a popular approach to explore the pathogenesis of various diseases. The present study was conducted to investigate protein expression alteration in the hippocampus and to identify the molecular mechanisms underlying ethanol-induced learning and memory impairments. Mouse models of chronic ethanol intoxication were established by intragastrical administration for 28 consecutive days, and hippocampal neuronal damage was assessed by Nissl staining. Recognition memory was evaluated by Novel object recognition and Morris water maze tests, and hippocampus tissues were collected for label-free quantitative proteomics and analyzed using bioinformatics methods. Our study showed that chronic ethanol exposure prompted marked changes in protein expression in the hippocampus. We identified 32 differentially expressed proteins, of which 21 were upregulated and 11 downregulated. Gene Ontology analysis suggested that the identified differentially proteins were mainly involved in cytoskeleton and signal transduction mechanisms. Further verification using Western blotting and real-time quantitative PCR revealed that the hippocampal CTSL (cathepsin L), and PVALB (Parvalbumin) showed strongest expression changes, the latter being specifically expressed in GABAergic interneurons. These two proteins might serve as candidate protein biomarkers, providing new prospects for the diagnosis and treatment of ethanol-induced learning and memory disorders.
Collapse
Affiliation(s)
- Dandan Liu
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shuqiong Liu
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jiande Li
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaohuan Liu
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaoxuan Wu
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ying Peng
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Qingyu Shen
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; The Fourth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
14
|
Philyaw TJ, Rothenfluh A, Titos I. The Use of Drosophila to Understand Psychostimulant Responses. Biomedicines 2022; 10:119. [PMID: 35052798 PMCID: PMC8773124 DOI: 10.3390/biomedicines10010119] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 12/31/2021] [Accepted: 12/31/2021] [Indexed: 01/27/2023] Open
Abstract
The addictive properties of psychostimulants such as cocaine, amphetamine, methamphetamine, and methylphenidate are based on their ability to increase dopaminergic neurotransmission in the reward system. While cocaine and methamphetamine are predominately used recreationally, amphetamine and methylphenidate also work as effective therapeutics to treat symptoms of disorders including attention deficit and hyperactivity disorder (ADHD) and autism spectrum disorder (ASD). Although both the addictive properties of psychostimulant drugs and their therapeutic efficacy are influenced by genetic variation, very few genes that regulate these processes in humans have been identified. This is largely due to population heterogeneity which entails a requirement for large samples. Drosophila melanogaster exhibits similar psychostimulant responses to humans, a high degree of gene conservation, and allow performance of behavioral assays in a large population. Additionally, amphetamine and methylphenidate reduce impairments in fly models of ADHD-like behavior. Therefore, Drosophila represents an ideal translational model organism to tackle the genetic components underlying the effects of psychostimulants. Here, we break down the many assays that reliably quantify the effects of cocaine, amphetamine, methamphetamine, and methylphenidate in Drosophila. We also discuss how Drosophila is an efficient and cost-effective model organism for identifying novel candidate genes and molecular mechanisms involved in the behavioral responses to psychostimulant drugs.
Collapse
Affiliation(s)
- Travis James Philyaw
- Molecular Biology Graduate Program, University of Utah, Salt Lake City, UT 84112, USA;
| | - Adrian Rothenfluh
- Department of Psychiatry, Huntsman Mental Health Institute, University of Utah, Salt Lake City, UT 84108, USA
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA
- Department of Neurobiology, University of Utah, Salt Lake City, UT 84132, USA
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Iris Titos
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
15
|
Scaplen KM, Petruccelli E. Receptors and Channels Associated with Alcohol Use: Contributions from Drosophila. Neurosci Insights 2021; 16:26331055211007441. [PMID: 33870197 PMCID: PMC8020223 DOI: 10.1177/26331055211007441] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 03/03/2021] [Indexed: 12/14/2022] Open
Abstract
Alcohol Use Disorder (AUD) is a debilitating disorder that manifests as problematic patterns of alcohol use. At the core of AUD's behavioral manifestations are the profound structural, physiological, cellular, and molecular effects of alcohol on the brain. While the field has made considerable progress in understanding the neuromolecular targets of alcohol we still lack a comprehensive understanding of alcohol's actions and effective treatment strategies. Drosophila melanogaster is a powerful model for investigating the neuromolecular targets of alcohol because flies model many of the core behavioral elements of AUD and offer a rich genetic toolkit to precisely reveal the in vivo molecular actions of alcohol. In this review, we focus on receptors and channels that are often targeted by alcohol within the brain. We discuss the general roles of these proteins, their role in alcohol-associated behaviors across species, and propose ways in which Drosophila models can help advance the field.
Collapse
Affiliation(s)
- Kristin M Scaplen
- Department of Psychology, Bryant University, Smithfield, RI, USA
- Center for Health and Behavioral Studies, Bryant University, Smithfield, RI, USA
- Department of Neuroscience, Brown University, Providence, RI, USA
| | - Emily Petruccelli
- Department of Biological Sciences, Southern Illinois University Edwardsville, Edwardsville, IL, USA
| |
Collapse
|
16
|
Chakraborty M, Chang CH, Khost DE, Vedanayagam J, Adrion JR, Liao Y, Montooth KL, Meiklejohn CD, Larracuente AM, Emerson JJ. Evolution of genome structure in the Drosophila simulans species complex. Genome Res 2021; 31:380-396. [PMID: 33563718 PMCID: PMC7919458 DOI: 10.1101/gr.263442.120] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 12/28/2020] [Indexed: 12/25/2022]
Abstract
The rapid evolution of repetitive DNA sequences, including satellite DNA, tandem duplications, and transposable elements, underlies phenotypic evolution and contributes to hybrid incompatibilities between species. However, repetitive genomic regions are fragmented and misassembled in most contemporary genome assemblies. We generated highly contiguous de novo reference genomes for the Drosophila simulans species complex (D. simulans, D. mauritiana, and D. sechellia), which speciated ∼250,000 yr ago. Our assemblies are comparable in contiguity and accuracy to the current D. melanogaster genome, allowing us to directly compare repetitive sequences between these four species. We find that at least 15% of the D. simulans complex species genomes fail to align uniquely to D. melanogaster owing to structural divergence-twice the number of single-nucleotide substitutions. We also find rapid turnover of satellite DNA and extensive structural divergence in heterochromatic regions, whereas the euchromatic gene content is mostly conserved. Despite the overall preservation of gene synteny, euchromatin in each species has been shaped by clade- and species-specific inversions, transposable elements, expansions and contractions of satellite and tRNA tandem arrays, and gene duplications. We also find rapid divergence among Y-linked genes, including copy number variation and recent gene duplications from autosomes. Our assemblies provide a valuable resource for studying genome evolution and its consequences for phenotypic evolution in these genetic model species.
Collapse
Affiliation(s)
- Mahul Chakraborty
- Department of Ecology and Evolutionary Biology, University of California Irvine, Irvine, California 92697, USA
| | - Ching-Ho Chang
- Department of Biology, University of Rochester, Rochester, New York 14627, USA
| | - Danielle E Khost
- Department of Biology, University of Rochester, Rochester, New York 14627, USA
- FAS Informatics and Scientific Applications, Harvard University, Cambridge, Massachusetts 02138, USA
| | - Jeffrey Vedanayagam
- Department of Developmental Biology, Memorial Sloan-Kettering Cancer Center, New York, New York 10065, USA
| | - Jeffrey R Adrion
- Institute of Ecology and Evolution, University of Oregon, Eugene, Oregon 97403, USA
| | - Yi Liao
- Department of Ecology and Evolutionary Biology, University of California Irvine, Irvine, California 92697, USA
| | - Kristi L Montooth
- School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska 68502, USA
| | - Colin D Meiklejohn
- School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska 68502, USA
| | | | - J J Emerson
- Department of Ecology and Evolutionary Biology, University of California Irvine, Irvine, California 92697, USA
| |
Collapse
|
17
|
El-Merhie N, Krüger A, Uliczka K, Papenmeier S, Roeder T, Rabe KF, Wagner C, Angstmann H, Krauss-Etschmann S. Sex dependent effect of maternal e-nicotine on F1 Drosophila development and airways. Sci Rep 2021; 11:4441. [PMID: 33627715 PMCID: PMC7904947 DOI: 10.1038/s41598-021-81607-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 01/01/2021] [Indexed: 01/03/2023] Open
Abstract
E-cigarettes are heavily advertised as healthier alternative to common tobacco cigarettes, leading more and more women to switch from regular cigarettes to ENDS (electronic nicotine delivery system) during pregnancy. While the noxious consequences of tobacco smoking during pregnancy on the offspring health are well-described, information on the long-term consequences due to maternal use of e-cigarettes do not exist so far. Therefore, we aimed to investigate how maternal e-nicotine influences offspring development from earliest life until adulthood. To this end, virgin female Drosophila melanogaster flies were exposed to nicotine vapor (8 µg nicotine) once per hour for a total of eight times. Following the last exposure, e-nicotine or sham exposed females were mated with non-exposed males. The F1-generation was then analyzed for viability, growth and airway structure. We demonstrate that maternal exposure to e-nicotine not only leads to reduced maternal fertility, but also negatively affects size and weight, as well as tracheal development of the F1-generation, lasting from embryonic stage until adulthood. These results not only underline the need for studies investigating the effects of maternal vaping on offspring health, but also propose our established model for analyzing molecular mechanisms and signaling pathways mediating these intergenerational changes.
Collapse
Affiliation(s)
- Natalia El-Merhie
- Division of Experimental Asthma Research, Early Life Origins of Chronic Lung Disease, Research Center Borstel, Leibniz Lung Center, German Center for Lung Research (DZL) and the Airway Research Center North (ARCN), Borstel, Germany
| | - Arne Krüger
- Division of Experimental Asthma Research, Early Life Origins of Chronic Lung Disease, Research Center Borstel, Leibniz Lung Center, German Center for Lung Research (DZL) and the Airway Research Center North (ARCN), Borstel, Germany
| | - Karin Uliczka
- Division of Experimental Asthma Research, Early Life Origins of Chronic Lung Disease, Research Center Borstel, Leibniz Lung Center, German Center for Lung Research (DZL) and the Airway Research Center North (ARCN), Borstel, Germany
- Invertebrate Models, Priority Area Asthma & Allergy, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Stephanie Papenmeier
- Division of Experimental Asthma Research, Early Life Origins of Chronic Lung Disease, Research Center Borstel, Leibniz Lung Center, German Center for Lung Research (DZL) and the Airway Research Center North (ARCN), Borstel, Germany
- Invertebrate Models, Priority Area Asthma & Allergy, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Thomas Roeder
- Department of Molecular Physiology and Zoology, Christian Albrechts University, German Center for Lung Research (DZL) and the Airway Research Center North (ARCN), Kiel, Germany
| | - Klaus F Rabe
- Department of Pneumology, LungenClinic, German Center for Lung Research (DZL) and the Airway Research Center North (ARCN), Grosshansdorf, Germany
- Department of Medicine, Christian Albrechts University, German Center for Lung Research (DZL) and the Airway Research Center North (ARCN), Kiel, Germany
| | - Christina Wagner
- Invertebrate Models, Priority Area Asthma & Allergy, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Hanna Angstmann
- Division of Experimental Asthma Research, Early Life Origins of Chronic Lung Disease, Research Center Borstel, Leibniz Lung Center, German Center for Lung Research (DZL) and the Airway Research Center North (ARCN), Borstel, Germany
| | - Susanne Krauss-Etschmann
- Division of Experimental Asthma Research, Early Life Origins of Chronic Lung Disease, Research Center Borstel, Leibniz Lung Center, German Center for Lung Research (DZL) and the Airway Research Center North (ARCN), Borstel, Germany.
- Institute for Experimental Medicine, Christian Albrechts University, German Center for Lung Research (DZL) and the Airway Research Center North (ARCN), Kiel, Germany.
| |
Collapse
|
18
|
Karnib N, van Staaden MJ. The Deep Roots of Addiction: A Comparative Perspective. BRAIN, BEHAVIOR AND EVOLUTION 2021; 95:222-229. [PMID: 33567426 DOI: 10.1159/000514180] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Accepted: 12/31/2020] [Indexed: 11/19/2022]
Abstract
Addiction is a debilitating condition that extracts enormous social and economic tolls. Despite several decades of research, our knowledge of its etiology, preventive measures, and treatments is limited. A relatively recent research field with the potential to provide a more holistic understanding, and subsequently treatments, takes a phylogenetic view of addiction. This perspective is based on deep homologies at the genetic, proteomic, and behavioral levels, which are shared across all metazoan life; particularly those organisms faced with plant secondary metabolites as defensive compounds against insect herbivory. These addictive alkaloids, such as nicotine, cocaine, or cathinone, are commonly referred to as "human drugs of abuse" even though humans had little to no role in the co-evolutionary processes that determined their initial emergence or continued selection. This commentary discusses the overwhelming homologies of addictive alkaloid effects on neural systems across a wide range of taxa, as we aim to develop a broader comparative view of the "addicted brain." Taking nicotine as an example, homologous physiological responses to this compound identify common underlying cellular and molecular mechanisms that advocate for the adoption of a phylogenetic view of addiction.
Collapse
Affiliation(s)
- Nabil Karnib
- Department of Biological Sciences, JP Scott Center for Neuroscience, Mind and Behavior, Bowling Green State University, Bowling Green, Ohio, USA
| | - Moira J van Staaden
- Department of Biological Sciences, JP Scott Center for Neuroscience, Mind and Behavior, Bowling Green State University, Bowling Green, Ohio, USA,
| |
Collapse
|
19
|
Identification of Genes Involved in the Differentiation of R7y and R7p Photoreceptor Cells in Drosophila. G3-GENES GENOMES GENETICS 2020; 10:3949-3958. [PMID: 32972998 PMCID: PMC7642934 DOI: 10.1534/g3.120.401370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The R7 and R8 photoreceptor cells of the Drosophila compound eye mediate color vision. Throughout the majority of the eye, these cells occur in two principal types of ommatidia. Approximately 35% of ommatidia are of the pale type and express Rh3 in R7 cells and Rh5 in R8 cells. The remaining 65% are of the yellow type and express Rh4 in R7 cells and Rh6 in R8 cells. The specification of an R8 cell in a pale or yellow ommatidium depends on the fate of the adjacent R7 cell. However, pale and yellow R7 cells are specified by a stochastic process that requires the genes spineless, tango and klumpfuss. To identify additional genes involved in this process we performed genetic screens using a collection of 480 P{EP} transposon insertion strains. We identified genes in gain of function and loss of function screens that significantly altered the percentage of Rh3 expressing R7 cells (Rh3%) from wild-type. 36 strains resulted in altered Rh3% in the gain of function screen where the P{EP} insertion strains were crossed to a sevEP-GAL4 driver line. 53 strains resulted in altered Rh3% in the heterozygous loss of function screen. 4 strains showed effects that differed between the two screens, suggesting that the effect found in the gain of function screen was either larger than, or potentially masked by, the P{EP} insertion alone. Analyses of homozygotes validated many of the candidates identified. These results suggest that R7 cell fate specification is sensitive to perturbations in mRNA transcription, splicing and localization, growth inhibition, post-translational protein modification, cleavage and secretion, hedgehog signaling, ubiquitin protease activity, GTPase activation, actin and cytoskeletal regulation, and Ser/Thr kinase activity, among other diverse signaling and cell biological processes.
Collapse
|
20
|
Witt SH, Frank J, Frischknecht U, Treutlein J, Streit F, Foo JC, Sirignano L, Dukal H, Degenhardt F, Koopmann A, Hoffmann S, Koller G, Pogarell O, Preuss UW, Zill P, Adorjan K, Schulze TG, Nöthen M, Spanagel R, Kiefer F, Rietschel M. Acute alcohol withdrawal and recovery in men lead to profound changes in DNA methylation profiles: a longitudinal clinical study. Addiction 2020; 115:2034-2044. [PMID: 32080920 DOI: 10.1111/add.15020] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 10/24/2019] [Accepted: 02/14/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIMS Withdrawal is a serious and sometimes life-threatening event in alcohol-dependent individuals. It has been suggested that epigenetic processes may play a role in this context. This study aimed to identify genes and pathways involved in such processes which hint to relevant mechanisms underlying withdrawal. DESIGN Cross-sectional case-control study and longitudinal within-cases study during alcohol withdrawal and after 2 weeks of recovery SETTING: Addiction medicine departments in two university hospitals in southern Germany. PARTICIPANTS/CASES Ninety-nine alcohol-dependent male patients receiving in-patient treatment and suffering from severe withdrawal symptoms during detoxification and 95 age-matched male controls. MEASUREMENTS Epigenome-wide methylation patterns were analyzed in patients during acute alcohol withdrawal and after 2 weeks of recovery, as well as in age-matched controls using Illumina EPIC bead chips. Methylation levels of patients and controls were tested for association with withdrawal status. Tests were adjusted for technical and batch effects, age, smoking and cell type distribution. Single-site analysis, as well as an analysis of differentially methylated regions and gene ontology analysis, were performed. FINDINGS We found pronounced epigenome-wide significant [false discovery rate (FDR) < 0.05] differences between patients during withdrawal and after 2 weeks [2876 cytosine-phosphate-guanine (CpG) sites], as well as between patients and controls (9845 and 6094 CpG sites comparing patients at time-point 1 and patients at time-point 2 versus controls, respectively). Analysis of differentially methylated regions and involved pathways revealed an over-representation of gene ontology terms related to the immune system response. Differences between patients and controls diminished after recovery (> 800 CpG sites less), suggesting a partial reversibility of alcohol- and withdrawal-related methylation. CONCLUSIONS Acute alcohol withdrawal in severely dependent male patients appears to be associated with extensive changes in epigenome-wide methylation patterns. In particular, genes involved in immune system response seem to be affected by this condition.
Collapse
Affiliation(s)
- Stephanie H Witt
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany
| | - Josef Frank
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany
| | - Ulrich Frischknecht
- Department of Addictive Behaviour and Addiction Medicine, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany
| | - Jens Treutlein
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany
| | - Fabian Streit
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany
| | - Jerome C Foo
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany
| | - Lea Sirignano
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany
| | - Helene Dukal
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany
| | - Franziska Degenhardt
- Institute of Human Genetics, University of Bonn, Bonn, Germany.,Department of Genomics, Life and Brain Center, University of Bonn, Bonn, Germany
| | - Anne Koopmann
- Department of Addictive Behaviour and Addiction Medicine, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany
| | - Sabine Hoffmann
- Department of Addictive Behaviour and Addiction Medicine, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany
| | - Gabi Koller
- Department of Psychiatry and Psychotherapy, University Hospital, Ludwig Maximilian University (LMU) Munich, Munich, Germany
| | - Oliver Pogarell
- Department of Psychiatry and Psychotherapy, University Hospital, Ludwig Maximilian University (LMU) Munich, Munich, Germany
| | - Ulrich W Preuss
- Department of Psychiatry, Psychotherapy, Psychosomatics, Martin-Luther-University (MLU), Halle/Saale, Germany
| | - Peter Zill
- Department of Psychiatry and Psychotherapy, University Hospital, Ludwig Maximilian University (LMU) Munich, Munich, Germany
| | - Kristina Adorjan
- Department of Psychiatry and Psychotherapy, University Hospital, Ludwig Maximilian University (LMU) Munich, Munich, Germany.,Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, Ludwig Maximilian University (LMU) Munich, Munich, Germany
| | - Thomas G Schulze
- Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, Ludwig Maximilian University (LMU) Munich, Munich, Germany
| | - Markus Nöthen
- Institute of Human Genetics, University of Bonn, Bonn, Germany.,Department of Genomics, Life and Brain Center, University of Bonn, Bonn, Germany
| | - Rainer Spanagel
- Institute of Psychopharmacology, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany
| | - Falk Kiefer
- Department of Addictive Behaviour and Addiction Medicine, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany
| | - Marcella Rietschel
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany
| |
Collapse
|
21
|
Lathen DR, Merrill CB, Rothenfluh A. Flying Together: Drosophila as a Tool to Understand the Genetics of Human Alcoholism. Int J Mol Sci 2020; 21:E6649. [PMID: 32932795 PMCID: PMC7555299 DOI: 10.3390/ijms21186649] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/07/2020] [Accepted: 09/08/2020] [Indexed: 12/14/2022] Open
Abstract
Alcohol use disorder (AUD) exacts an immense toll on individuals, families, and society. Genetic factors determine up to 60% of an individual's risk of developing problematic alcohol habits. Effective AUD prevention and treatment requires knowledge of the genes that predispose people to alcoholism, play a role in alcohol responses, and/or contribute to the development of addiction. As a highly tractable and translatable genetic and behavioral model organism, Drosophila melanogaster has proven valuable to uncover important genes and mechanistic pathways that have obvious orthologs in humans and that help explain the complexities of addiction. Vinegar flies exhibit remarkably strong face and mechanistic validity as a model for AUDs, permitting many advancements in the quest to understand human genetic involvement in this disease. These advancements occur via approaches that essentially fall into one of two categories: (1) discovering candidate genes via human genome-wide association studies (GWAS), transcriptomics on post-mortem tissue from AUD patients, or relevant physiological connections, then using reverse genetics in flies to validate candidate genes' roles and investigate their molecular function in the context of alcohol. (2) Utilizing flies to discover candidate genes through unbiased screens, GWAS, quantitative trait locus analyses, transcriptomics, or single-gene studies, then validating their translational role in human genetic surveys. In this review, we highlight the utility of Drosophila as a model for alcoholism by surveying recent advances in our understanding of human AUDs that resulted from these various approaches. We summarize the genes that are conserved in alcohol-related function between humans and flies. We also provide insight into some advantages and limitations of these approaches. Overall, this review demonstrates how Drosophila have and can be used to answer important genetic questions about alcohol addiction.
Collapse
Affiliation(s)
- Daniel R. Lathen
- Department of Psychiatry and Neuroscience Ph.D. Program, University of Utah, Salt Lake City, UT 84108, USA;
| | - Collin B. Merrill
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA;
| | - Adrian Rothenfluh
- Department of Psychiatry and Neuroscience Ph.D. Program, University of Utah, Salt Lake City, UT 84108, USA;
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA;
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT 84132, USA
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
22
|
Wang XC, Liu Z, Jin LH. Drosophila jumu modulates apoptosis via a JNK-dependent pathway and is required for other processes in wing development. Apoptosis 2020; 24:465-477. [PMID: 30796611 DOI: 10.1007/s10495-019-01527-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Previous studies in several model organisms have revealed that members of the Forkhead (Fkh) transcription factor family have multiple functions. Drosophila Jumeau (Jumu), a member of this family, participates in cardiogenesis, hematopoiesis and immune system homeostasis. Here, we show that loss of jumu function positively regulates or triggers apoptosis via a JNK-dependent pathway in wing development. jumu mutants showed reduced wing size and increased apoptosis. Moreover, we observed a loss of the anterior cross vein (ACV) phenotype that was similar to that observed in wings in which JNK signaling has been ectopically activated. The JNK signaling markers puckered (puc) and p-JNK were also significantly increased in the wing discs of jumu mutants. In addition, apoptosis induced by the loss of jumu was rescued by knocking down JNK, indicating a role for JNK in reducing jumu-induced apoptosis. Jumu could also control wing margin development via the positive regulation of cut expression, and the observed wing margin defect did not result from a loss of jumu-induced apoptosis. Further, jumu deficiency in the pupal wing could induce multiple wing hairs via a Rho1-mediated planar cell polarity pathway, but abnormal Rho1 expression was not why jumu loss induced apoptosis via a JNK-dependent pathway in wing discs.
Collapse
Affiliation(s)
- Xiao Chun Wang
- Department of Genetics, College of Life Sciences, Northeast Forestry University, Harbin, 150040, China
| | - Ziguang Liu
- Heilongjiang Academy of Agricultural Sciences, Harbin, 150040, China
| | - Li Hua Jin
- Department of Genetics, College of Life Sciences, Northeast Forestry University, Harbin, 150040, China.
| |
Collapse
|
23
|
Yang S, Zhao Y, Chen X, Lu X, Chen Y, Zhao X, Zhu L, Fang Z, Zhao H, Yao Y, Liu C, Shen C. The ACTB Variants and Alcohol Drinking Confer Joint Effect to Ischemic Stroke in Chinese Han Population. J Atheroscler Thromb 2020; 27:226-244. [PMID: 31327802 PMCID: PMC7113141 DOI: 10.5551/jat.49536] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 05/28/2019] [Indexed: 12/11/2022] Open
Abstract
AIM β-actin (ACTB) participates in the vascular remodeling and contributes to the cardiovascular diseases. Herein, we investigated the associations of ACTB with hypertension and stroke. METHODS Three single-nucleotide polymorphisms in ACTB were selected for genotyping in 2,012 hypertension cases and 2,210 controls. The associations of ACTB with hypertension and stroke were examined in another follow-up study. Logistic and Cox regression were performed in a case-control study and a follow-up study, respectively. Additive scale interaction was examined by calculating the relative excess risk due to interaction (RERI), attributable proportion due to interaction (AP) and synergy index (SI). The multiplicative interaction hazard ratio was calculated by fitting the Cox regression model. ACTB mRNA in peripheral blood mononuclear cells was measured in ischemic stroke (IS) cases and in controls. RESULTS The associations of rs852426 with hypertension and stroke had statistical significance in drinkers but not after Bonferroni correction. An additive interaction of rs852426 and drinking was observed for stroke incidence, the adjusted RERI was -0.907 (p=4.108×10-4), and the multiplicative interaction was still sound (HR=0.541, p=0.048). Furthermore, the significant interaction was further replicated in a nested case-control study. In the drinking population, the relative expression of ACTB mRNA in IS was lower (0.99±0.26) than that in controls (1.13±0.20), with a p value of 0.026. CONCLUSIONS ACTB rs852426 was significantly associated with alcohol consumption on stroke risk, and the expression of ACTB mRNA in IS who had a drinking habit was significantly down-regulated. This finding will provide a novel insight into the prevention of stroke.
Collapse
Affiliation(s)
- Song Yang
- Department of Cardiology, Affiliated Yixing People's Hospital of Jiangsu University, People's Hospital of Yixing City, Yixing, China
| | - Yanping Zhao
- Department of Neurology, Affiliated Yixing People's Hospital of Jiangsu University, People's Hospital of Yixing City, Yixing, China
| | - Xiaotian Chen
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Xiangfeng Lu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yanchun Chen
- Department of Cardiology, Affiliated Yixing People's Hospital of Jiangsu University, People's Hospital of Yixing City, Yixing, China
| | - Xianghai Zhao
- Department of Cardiology, Affiliated Yixing People's Hospital of Jiangsu University, People's Hospital of Yixing City, Yixing, China
| | - Lijun Zhu
- Department of Epidemiology and Biostatistics, School of Public Health, Wannan Medical College, Wuhu, China
| | - Zhengmei Fang
- Department of Epidemiology and Biostatistics, School of Public Health, Wannan Medical College, Wuhu, China
| | - Hailong Zhao
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Yingshui Yao
- Department of Epidemiology and Biostatistics, School of Public Health, Wannan Medical College, Wuhu, China
| | - Chunlan Liu
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Chong Shen
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, China
| |
Collapse
|
24
|
Scholz H. Unraveling the Mechanisms of Behaviors Associated With AUDs Using Flies and Worms. Alcohol Clin Exp Res 2019; 43:2274-2284. [PMID: 31529787 DOI: 10.1111/acer.14199] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 09/11/2019] [Indexed: 12/11/2022]
Abstract
Alcohol use disorders (AUDs) are very common worldwide and negatively affect both individuals and societies. To understand how normal behavior turns into uncontrollable use of alcohol, several approaches have been utilized in the last decades. However, we still do not completely understand how AUDs evolve or how they are maintained in the brains of affected individuals. In addition, efficient and effective treatment is still in need of development. This review focuses on alternative approaches developed over the last 20 years using Drosophila melanogaster (Drosophila) and Caenorhabditis elegans (C. elegans) as genetic model systems to determine the mechanisms underlying the action of ethanol (EtOH) and behaviors associated with AUDs. All the results and insights of studies over the last 20 years cannot be comprehensively summarized. Thus, a few prominent examples are provided highlighting the principles of the genes and mechanisms that have been uncovered and are involved in the action of EtOH at the cellular level. In addition, examples are provided of the genes and mechanisms that regulate behaviors relevant to acquiring and maintaining excessive alcohol intake, such as decision making, reward and withdrawal, and/or relapse regulation. How the insight gained from the results of Drosophila and C. elegans models can be translated to higher organisms, such as rodents and/or humans, is discussed, as well as whether these insights have any relevance or impact on our understanding of the mechanisms underlying AUDs in humans. Finally, future directions are presented that might facilitate the identification of drugs to treat AUDs.
Collapse
Affiliation(s)
- Henrike Scholz
- From the, Department of Biology, Institute for Zoology, Albertus-Magnus University of Cologne, Cologne, Germany
| |
Collapse
|
25
|
Altered Actin Filament Dynamics in the Drosophila Mushroom Bodies Lead to Fast Acquisition of Alcohol Consumption Preference. J Neurosci 2019; 39:8877-8884. [PMID: 31558618 DOI: 10.1523/jneurosci.0973-19.2019] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 08/22/2019] [Accepted: 09/13/2019] [Indexed: 01/12/2023] Open
Abstract
Alcohol use is highly prevalent in the United States and across the world, and every year millions of people suffer from alcohol use disorders (AUDs). Although the genetic contribution to developing AUDs is estimated to be 50-60%, many of the underlying molecular mechanisms remain unclear. Previous studies from our laboratory revealed that Drosophila melanogaster lacking RhoGAP18B and Ras Suppressor 1 (Rsu1) display reduced sensitivity to ethanol-induced sedation. Both Rsu1 and RhoGAP18B are negative regulators of the small Rho-family GTPase, Rac1, a modulator of actin dynamics. Here we investigate the role of Rac1 and its downstream target, the actin-severing protein cofilin, in alcohol consumption preference. We show that these two regulators of actin dynamics can alter male experience-dependent alcohol preference in a bidirectional manner: expressing either activated Rac1 or dominant-negative cofilin in the mushroom bodies (MBs) abolishes experience-dependent alcohol preference. Conversely, dominant-negative Rac1 or activated cofilin MB expression lead to faster acquisition of alcohol preference. Our data show that Rac1 and cofilin activity are key to determining the rate of acquisition of alcohol preference, revealing a critical role of actin dynamics regulation in the development of voluntary self-administration in Drosophila SIGNIFICANCE STATEMENT The risks for developing an alcohol use disorder (AUD) are strongly determined by genetic factors. Understanding the genes and molecular mechanisms that contribute to that risk is therefore a necessary first step for the development of targeted therapeutic intervention. Here we show that regulators of actin cytoskeleton dynamics can bidirectionally determine the acquisition rate of alcohol self-administration, highlighting this process as a key mechanism contributing to the risk of AUD development.
Collapse
|
26
|
Zhao Y, Ge Y, Zheng ZL. Brain Imaging-Guided Analysis Reveals DNA Methylation Profiles Correlated with Insular Surface Area and Alcohol Use Disorder. Alcohol Clin Exp Res 2019; 43:628-639. [PMID: 30830696 PMCID: PMC6443499 DOI: 10.1111/acer.13971] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 01/26/2019] [Indexed: 12/26/2022]
Abstract
BACKGROUND Alcohol use disorder (AUD) is a wide-spread, heritable brain disease, but few studies have linked genetic variants or epigenetic factors to brain structures related to AUD in humans, due to many factors including the high-dimensional nature of imaging and genomic data. METHODS To provide potential insights into the links among epigenetic regulation, brain structure, and AUD, we have performed an integrative analysis of brain structural imaging and blood DNA methylome data from 52 AUD and 58 healthy control (HC) subjects collected in the Nathan Kline Institute-Rockland Sample. RESULTS We first found that AUD subjects had significantly larger insular surface area than HC in both left and right hemispheres. We then found that 7,827 DNA methylation probes on the HumanMethylation450K BeadChip had significant correlations with the right insular surface area (false discovery rate [FDR] < 0.05). Furthermore, we showed that 44 of the insular surface area-correlated methylation probes were also strongly correlated with AUD status (FDR < 0.05). These AUD-correlated probes are annotated to 36 protein-coding genes, with 16 genes (44%) having been reported by others to be related to AUD or alcohol response, including TAS2R16 and PER2. The remaining 20 genes, in particular ARHGAP22, might represent novel genes involved in AUD or responsive to alcohol. CONCLUSIONS We have identified 36 insular surface area- and AUD-correlated protein-coding genes that are either known to be AUD- or alcohol-related or not yet reported by prior studies. Therefore, our study suggests that the brain imaging-guided epigenetic analysis has a potential of identifying possible epigenetic mechanisms involved in AUD.
Collapse
Affiliation(s)
- Yihong Zhao
- Department of Child and Adolescent Psychiatry, NYU Langone Medical Center, New York, NY 10016, USA
- Center for Behavioral Science Research, Department of Health Policy & Health Services Research, Boston University, Boston, MA 02118, USA
| | - Yongchao Ge
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Zhi-Liang Zheng
- Department of Biological Sciences, Lehman College, City University of New York, Bronx, NY 10468, USA
| |
Collapse
|
27
|
Signor S, Nuzhdin S. Dynamic changes in gene expression and alternative splicing mediate the response to acute alcohol exposure in Drosophila melanogaster. Heredity (Edinb) 2018; 121:342-360. [PMID: 30143789 PMCID: PMC6133934 DOI: 10.1038/s41437-018-0136-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 06/21/2018] [Accepted: 07/19/2018] [Indexed: 12/18/2022] Open
Abstract
Environmental changes typically cause rapid gene expression responses in the exposed organisms, including changes in the representation of gene isoforms with different functions or properties. Identifying the genes that respond to environmental change, including in genotype-specific ways, is an important step in treating the undesirable physiological effects of stress, such as exposure to toxins or ethanol. Ethanol is a unique environmental stress in that chronic exposure results in permanent physiological changes and the development of alcohol use disorders. Drosophila is a classic model for deciphering the mechanisms of the response to alcohol exposure, as it meets the criteria for the development of alcohol use disorders, and has similar physiological underpinnings with vertebrates. Because many studies on the response to ethanol have relied on a priori candidate genes, broad surveys of gene expression and splicing are required and have been investigated here. Further, we expose Drosophila to ethanol in an environment that is genetically, socially, and ecologically relevant. Both expression and splicing differences, inasmuch as they can be decomposed, contribute to the response to ethanol in Drosophila melanogaster. However, we find that while D. melanogaster responds to ethanol, there is very little genetic variation in how it responds to ethanol. In addition, the response to alcohol over time is dynamic, suggesting that incorporating time into studies on the response to the environment is important.
Collapse
Affiliation(s)
- Sarah Signor
- Department of Molecular and Computational Biology, University of Southern California, Los Angeles, CA, USA.
| | - Sergey Nuzhdin
- Department of Molecular and Computational Biology, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
28
|
Zhang P, Rhodes JS, Garland T, Perez SD, Southey BR, Rodriguez-Zas SL. Brain region-dependent gene networks associated with selective breeding for increased voluntary wheel-running behavior. PLoS One 2018; 13:e0201773. [PMID: 30071007 PMCID: PMC6072066 DOI: 10.1371/journal.pone.0201773] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 07/20/2018] [Indexed: 12/14/2022] Open
Abstract
Mouse lines selectively bred for high voluntary wheel-running behavior are helpful models for uncovering gene networks associated with increased motivation for physical activity and other reward-dependent behaviors. The fact that multiple brain regions are hypothesized to contribute to distinct behavior components necessitates the simultaneous study of these regions. The goals of this study were to identify brain-region dependent and independent gene expression patterns, regulators, and networks associated with increased voluntary wheel-running behavior. The cerebellum and striatum from a high voluntary running line and a non-selected control line were compared. Neuropeptide genes annotated to reward-dependent processes including neuropeptide S receptor 1 (Npsr1), neuropeptide Y (Npy), and proprotein convertase subtilisin/kexin type 9 (Pcsk9), and genes implicated in motor coordination including vitamin D receptor (Vdr) and keratin, type I cytoskeletal 25 (Krt25) were among the genes exhibiting activity line-by-region interaction effects. Genes annotated to the Parkinson pathway presented consistent line patterns, albeit at different orders of magnitude between brain regions, suggesting some parallel events in response to selection for high voluntary activity. The comparison of gene networks between brain regions highlighted genes including transcription factor AP-2-delta (Tfap2d), distal-less homeobox 5 gene (Dlx5) and sine oculis homeobox homolog 3 (Six3) that exhibited line differential expression in one brain region and are associated with reward-dependent behaviors. Transcription factors including En2, Stat6 and Eomes predominated among regulators of genes that differed in expression between lines. Results from the simultaneous study of striatum and cerebellum confirm the necessity to study molecular mechanisms associated with voluntary activity and reward-dependent behaviors in consideration of brain region dependencies.
Collapse
Affiliation(s)
- Pan Zhang
- Illinois Informatics Institute, University of Illinois at Urbana-Champaign, Urbana, IL, United States of America
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States of America
| | - Justin S. Rhodes
- Beckman Institute for Advanced Science and Technology, Urbana, IL, United States of America
- Center for Nutrition, Learning and Memory, University of Illinois at Urbana-Champaign, Urbana, IL, United States of America
| | - Theodore Garland
- Department of Evolution, Ecology, and Organismal Biology, University of California, Riverside, CA, United States of America
| | - Sam D. Perez
- Beckman Institute for Advanced Science and Technology, Urbana, IL, United States of America
| | - Bruce R. Southey
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States of America
| | - Sandra L. Rodriguez-Zas
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States of America
- Department of Statistics, University of Illinois at Urbana-Champaign, Urbana, IL, United States of America
- Carle Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, United States of America
| |
Collapse
|
29
|
Morris M, Shaw A, Lambert M, Perry HH, Lowenstein E, Valenzuela D, Velazquez-Ulloa NA. Developmental nicotine exposure affects larval brain size and the adult dopaminergic system of Drosophila melanogaster. BMC DEVELOPMENTAL BIOLOGY 2018; 18:13. [PMID: 29898654 PMCID: PMC6001141 DOI: 10.1186/s12861-018-0172-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 05/21/2018] [Indexed: 01/15/2023]
Abstract
BACKGROUND Pregnant women may be exposed to nicotine if they smoke or use tobacco products, nicotine replacement therapy, or via e-cigarettes. Prenatal nicotine exposure has been shown to have deleterious effects on the nervous system in mammals including changes in brain size and in the dopaminergic system. The genetic and molecular mechanisms for these changes are not well understood. A Drosophila melanogaster model for these effects of nicotine exposure could contribute to faster identification of genes and molecular pathways underlying these effects. The purpose of this study was to determine if developmental nicotine exposure affects the nervous system of Drosophila melanogaster, focusing on changes to brain size and the dopaminergic system at two developmental stages. RESULTS We reared flies on control or nicotine food from egg to 3rd instar larvae or from egg to adult and determined effectiveness of the nicotine treatment. We used immunohistochemistry to visualize the whole brain and dopaminergic neurons, using tyrosine hydroxylase as the marker. We measured brain area, tyrosine hydroxylase fluorescence, and counted the number of dopaminergic neurons in brain clusters. We detected an increase in larval brain hemisphere area, a decrease in tyrosine hydroxylase fluorescence in adult central brains, and a decrease in the number of neurons in the PPM3 adult dopaminergic cluster. We tested involvement of Dα7, one of the nicotinic acetylcholine receptor subunits, and found it was involved in eclosion, as previously described, but not involved in brain size. CONCLUSIONS We conclude that developmental nicotine exposure in Drosophila melanogaster affects brain size and the dopaminergic system. Prenatal nicotine exposure in mammals has also been shown to have effects on brain size and in the dopaminergic system. This study further establishes Drosophila melanogaster as model organism to study the effects of developmental nicotine exposure. The genetic and molecular tools available for Drosophila research will allow elucidation of the mechanisms underlying the effects of nicotine exposure during development.
Collapse
Affiliation(s)
- Melanie Morris
- School of Medicine, University of Washington, Seattle, USA
| | - Ariel Shaw
- Biochemistry, Cell and Molecular Biology Program, Lewis & Clark College, Portland, USA
| | | | | | - Eve Lowenstein
- Biology Department, Lewis & Clark College, Portland, USA
| | | | | |
Collapse
|
30
|
Lowenstein EG, Velazquez-Ulloa NA. A Fly's Eye View of Natural and Drug Reward. Front Physiol 2018; 9:407. [PMID: 29720947 PMCID: PMC5915475 DOI: 10.3389/fphys.2018.00407] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 04/04/2018] [Indexed: 12/18/2022] Open
Abstract
Animals encounter multiple stimuli each day. Some of these stimuli are innately appetitive or aversive, while others are assigned valence based on experience. Drugs like ethanol can elicit aversion in the short term and attraction in the long term. The reward system encodes the predictive value for different stimuli, mediating anticipation for attractive or punishing stimuli and driving animal behavior to approach or avoid conditioned stimuli. The neurochemistry and neurocircuitry of the reward system is partly evolutionarily conserved. In both vertebrates and invertebrates, including Drosophila melanogaster, dopamine is at the center of a network of neurotransmitters and neuromodulators acting in concert to encode rewards. Behavioral assays in D. melanogaster have become increasingly sophisticated, allowing more direct comparison with mammalian research. Moreover, recent evidence has established the functional modularity of the reward neural circuits in Drosophila. This functional modularity resembles the organization of reward circuits in mammals. The powerful genetic and molecular tools for D. melanogaster allow characterization and manipulation at the single-cell level. These tools are being used to construct a detailed map of the neural circuits mediating specific rewarding stimuli and have allowed for the identification of multiple genes and molecular pathways that mediate the effects of reinforcing stimuli, including their rewarding effects. This report provides an overview of the research on natural and drug reward in D. melanogaster, including natural rewards such as sugar and other food nutrients, and drug rewards including ethanol, cocaine, amphetamine, methamphetamine, and nicotine. We focused mainly on the known genetic and neural mechanisms underlying appetitive reward for sugar and reward for ethanol. We also include genes, molecular pathways, and neural circuits that have been identified using assays that test the palatability of the rewarding stimulus, the preference for the rewarding stimulus, or other effects of the stimulus that indicate how it can modify behavior. Commonalities between mechanisms of natural and drug reward are highlighted and future directions are presented, putting forward questions best suited for research using D. melanogaster as a model organism.
Collapse
Affiliation(s)
- Eve G Lowenstein
- Department of Biology, Lewis & Clark College, Portland, OR, United States
| | | |
Collapse
|
31
|
Gonzalez DA, Jia T, Pinzón JH, Acevedo SF, Ojelade SA, Xu B, Tay N, Desrivières S, Hernandez JL, Banaschewski T, Büchel C, Bokde AL, Conrod PJ, Flor H, Frouin V, Gallinat J, Garavan H, Gowland PA, Heinz A, Ittermann B, Lathrop M, Martinot JL, Paus T, Smolka MN, Rodan AR, Schumann G, Rothenfluh A. The Arf6 activator Efa6/PSD3 confers regional specificity and modulates ethanol consumption in Drosophila and humans. Mol Psychiatry 2018; 23:621-628. [PMID: 28607459 PMCID: PMC5729071 DOI: 10.1038/mp.2017.112] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 03/21/2017] [Accepted: 04/11/2017] [Indexed: 12/19/2022]
Abstract
Ubiquitously expressed genes have been implicated in a variety of specific behaviors, including responses to ethanol. However, the mechanisms that confer this behavioral specificity have remained elusive. Previously, we showed that the ubiquitously expressed small GTPase Arf6 is required for normal ethanol-induced sedation in adult Drosophila. Here, we show that this behavioral response also requires Efa6, one of (at least) three Drosophila Arf6 guanine exchange factors. Ethanol-naive Arf6 and Efa6 mutants were sensitive to ethanol-induced sedation and lacked rapid tolerance upon re-exposure to ethanol, when compared with wild-type flies. In contrast to wild-type flies, both Arf6 and Efa6 mutants preferred alcohol-containing food without prior ethanol experience. An analysis of the human ortholog of Arf6 and orthologs of Efa6 (PSD1-4) revealed that the minor G allele of single nucleotide polymorphism (SNP) rs13265422 in PSD3, as well as a haplotype containing rs13265422, was associated with an increased frequency of drinking and binge drinking episodes in adolescents. The same haplotype was also associated with increased alcohol dependence in an independent European cohort. Unlike the ubiquitously expressed human Arf6 GTPase, PSD3 localization is restricted to the brain, particularly the prefrontal cortex (PFC). Functional magnetic resonance imaging revealed that the same PSD3 haplotype was also associated with a differential functional magnetic resonance imaging signal in the PFC during a Go/No-Go task, which engages PFC-mediated executive control. Our translational analysis, therefore, suggests that PSD3 confers regional specificity to ubiquitous Arf6 in the PFC to modulate human alcohol-drinking behaviors.
Collapse
Affiliation(s)
- Dante A. Gonzalez
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX,Program in Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX
| | - Tianye Jia
- Institute of Psychiatry, King’s College London, United Kingdom,MRC Social, Genetic and Developmental Psychiatry (SGDP) Centre, London, United Kingdom
| | - Jorge H. Pinzón
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX
| | - Summer F. Acevedo
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX
| | - Shamsideen A. Ojelade
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX,Program in Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX
| | - Bing Xu
- Institute of Psychiatry, King’s College London, United Kingdom,MRC Social, Genetic and Developmental Psychiatry (SGDP) Centre, London, United Kingdom
| | - Nicole Tay
- Institute of Psychiatry, King’s College London, United Kingdom,MRC Social, Genetic and Developmental Psychiatry (SGDP) Centre, London, United Kingdom
| | - Sylvane Desrivières
- Institute of Psychiatry, King’s College London, United Kingdom,MRC Social, Genetic and Developmental Psychiatry (SGDP) Centre, London, United Kingdom
| | - Jeannie L. Hernandez
- Department of Psychiatry, Molecular Medicine Program, University of Utah, Salt Lake City
| | - Tobias Banaschewski
- Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Germany
| | | | - Arun L.W. Bokde
- Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Patricia J. Conrod
- MRC Social, Genetic and Developmental Psychiatry (SGDP) Centre, London, United Kingdom,Department of Psychiatry, Université de Montreal, CHU Ste Justine Hospital, Canada
| | - Herta Flor
- Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Germany
| | - Vincent Frouin
- Neurospin, Commissariat à l’Energie Atomique, Gif-sur-Yvette, France
| | - Jürgen Gallinat
- Department of Psychiatry and Psychotherapy, Campus Charité Mitte, Charité – Universitätsmedizin Berlin, Germany
| | - Hugh Garavan
- Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland,Departments of Psychiatry and Psychology, University of Vermont, Burlington, USA
| | - Penny A. Gowland
- Physikalisch-Technische Bundesanstalt (PTB), Braunschweig und Berlin, Germany
| | - Andreas Heinz
- Department of Psychiatry and Psychotherapy, Campus Charité Mitte, Charité – Universitätsmedizin Berlin, Germany
| | - Bernd Ittermann
- Physikalisch-Technische Bundesanstalt (PTB), Braunschweig und Berlin, Germany
| | - Mark Lathrop
- McGill University and Genome Quebec Innovation Centre, Ontario, Canada
| | - Jean-Luc Martinot
- Institut National de la Santé et de la Recherche Médicale, INSERM CEA Unit 1000 “Imaging & Psychiatry”, University Paris Sud, Orsay, and AP-HP Department of Adolescent Psychopathology and Medicine, Maison de Solenn, University Paris Descartes, Paris, France
| | - Tomás Paus
- School of Psychology, University of Nottingham, United Kingdom,Rotman Research Institute, University of Toronto, Toronto, Canada,Montreal Neurological Institute, McGill University, Canada
| | - Michael N. Smolka
- Department of Psychiatry and Psychotherapy, Technische Universität Dresden, Germany,Neuroimaging Center, Department of Psychology, Technische Universität Dresden, Germany
| | | | - Aylin R. Rodan
- Department of Internal Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, TX,Department of Internal Medicine, Division of Nephrology, Molecular Medicine Program, University of Utah, Salt Lake City
| | - Gunter Schumann
- Institute of Psychiatry, King’s College London, United Kingdom,MRC Social, Genetic and Developmental Psychiatry (SGDP) Centre, London, United Kingdom
| | - Adrian Rothenfluh
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX,Program in Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX,Department of Psychiatry, Molecular Medicine Program, University of Utah, Salt Lake City
| |
Collapse
|
32
|
De Nobrega AK, Lyons LC. Drosophila: An Emergent Model for Delineating Interactions between the Circadian Clock and Drugs of Abuse. Neural Plast 2017; 2017:4723836. [PMID: 29391952 PMCID: PMC5748135 DOI: 10.1155/2017/4723836] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 08/13/2017] [Indexed: 01/12/2023] Open
Abstract
Endogenous circadian oscillators orchestrate rhythms at the cellular, physiological, and behavioral levels across species to coordinate activity, for example, sleep/wake cycles, metabolism, and learning and memory, with predictable environmental cycles. The 21st century has seen a dramatic rise in the incidence of circadian and sleep disorders with globalization, technological advances, and the use of personal electronics. The circadian clock modulates alcohol- and drug-induced behaviors with circadian misalignment contributing to increased substance use and abuse. Invertebrate models, such as Drosophila melanogaster, have proven invaluable for the identification of genetic and molecular mechanisms underlying highly conserved processes including the circadian clock, drug tolerance, and reward systems. In this review, we highlight the contributions of Drosophila as a model system for understanding the bidirectional interactions between the circadian system and the drugs of abuse, alcohol and cocaine, and illustrate the highly conserved nature of these interactions between Drosophila and mammalian systems. Research in Drosophila provides mechanistic insights into the corresponding behaviors in higher organisms and can be used as a guide for targeted inquiries in mammals.
Collapse
Affiliation(s)
- Aliza K. De Nobrega
- Department of Biological Science, Program in Neuroscience, Florida State University, Tallahassee, FL 32306, USA
| | - Lisa C. Lyons
- Department of Biological Science, Program in Neuroscience, Florida State University, Tallahassee, FL 32306, USA
| |
Collapse
|
33
|
Das R, Bhattacharjee S, Patel AA, Harris JM, Bhattacharya S, Letcher JM, Clark SG, Nanda S, Iyer EPR, Ascoli GA, Cox DN. Dendritic Cytoskeletal Architecture Is Modulated by Combinatorial Transcriptional Regulation in Drosophila melanogaster. Genetics 2017; 207:1401-1421. [PMID: 29025914 PMCID: PMC5714456 DOI: 10.1534/genetics.117.300393] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 10/04/2017] [Indexed: 01/08/2023] Open
Abstract
Transcription factors (TFs) have emerged as essential cell autonomous mediators of subtype specific dendritogenesis; however, the downstream effectors of these TFs remain largely unknown, as are the cellular events that TFs control to direct morphological change. As dendritic morphology is largely dictated by the organization of the actin and microtubule (MT) cytoskeletons, elucidating TF-mediated cytoskeletal regulatory programs is key to understanding molecular control of diverse dendritic morphologies. Previous studies in Drosophila melanogaster have demonstrated that the conserved TFs Cut and Knot exert combinatorial control over aspects of dendritic cytoskeleton development, promoting actin and MT-based arbor morphology, respectively. To investigate transcriptional targets of Cut and/or Knot regulation, we conducted systematic neurogenomic studies, coupled with in vivo genetic screens utilizing multi-fluor cytoskeletal and membrane marker reporters. These analyses identified a host of putative Cut and/or Knot effector molecules, and a subset of these putative TF targets converge on modulating dendritic cytoskeletal architecture, which are grouped into three major phenotypic categories, based upon neuromorphometric analyses: complexity enhancer, complexity shifter, and complexity suppressor. Complexity enhancer genes normally function to promote higher order dendritic growth and branching with variable effects on MT stabilization and F-actin organization, whereas complexity shifter and complexity suppressor genes normally function in regulating proximal-distal branching distribution or in restricting higher order branching complexity, respectively, with spatially restricted impacts on the dendritic cytoskeleton. Collectively, we implicate novel genes and cellular programs by which TFs distinctly and combinatorially govern dendritogenesis via cytoskeletal modulation.
Collapse
Affiliation(s)
- Ravi Das
- Neuroscience Institute, Georgia State University, Atlanta, Georgia 30302
| | | | - Atit A Patel
- Neuroscience Institute, Georgia State University, Atlanta, Georgia 30302
| | - Jenna M Harris
- Neuroscience Institute, Georgia State University, Atlanta, Georgia 30302
| | | | - Jamin M Letcher
- Neuroscience Institute, Georgia State University, Atlanta, Georgia 30302
| | - Sarah G Clark
- Neuroscience Institute, Georgia State University, Atlanta, Georgia 30302
| | - Sumit Nanda
- Krasnow Institute for Advanced Study, George Mason University, Fairfax, Virginia 22030
| | | | - Giorgio A Ascoli
- Krasnow Institute for Advanced Study, George Mason University, Fairfax, Virginia 22030
| | - Daniel N Cox
- Neuroscience Institute, Georgia State University, Atlanta, Georgia 30302
| |
Collapse
|
34
|
Pinzón JH, Reed AR, Shalaby NA, Buszczak M, Rodan AR, Rothenfluh A. Alcohol-Induced Behaviors Require a Subset of Drosophila JmjC-Domain Histone Demethylases in the Nervous System. Alcohol Clin Exp Res 2017; 41:2015-2024. [PMID: 28940624 DOI: 10.1111/acer.13508] [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: 04/03/2017] [Accepted: 09/15/2017] [Indexed: 12/18/2022]
Abstract
BACKGROUND Long-lasting transcriptional changes underlie a number of adaptations that contribute to alcohol use disorders (AUD). Chromatin remodeling, including histone methylation, can confer distinct, long-lasting transcriptional changes, and histone methylases are known to play a role in the development of addiction. Conversely, little is known about the relevance of Jumonji (JmjC) domain-containing demethylases in AUDs. We systematically surveyed the alcohol-induced phenotypes of null mutations in all 13 Drosophila JmjC genes. METHODS We used a collection of JmjC mutants, the majority of which we generated by homologous recombination, and assayed them in the Booze-o-mat to determine their naïve sensitivity to sedation and their tolerance (change in sensitivity upon repeat exposure). Mutants with reproducible phenotypes had their phenotypes rescued with tagged genomic transgenes, and/or phenocopied by nervous system-specific knockdown using RNA interference (RNAi). RESULTS Four of the 13 JmjC genes (KDM3, lid, NO66, and HSPBAP1) showed reproducible ethanol (EtOH) sensitivity phenotypes. Some of the phenotypes were observed across doses, for example, the enhanced EtOH sensitivity of KDM3KO and NO66KO , but others were dose dependent, such as the reduced EtOH sensitivity of HSPBAP1KO , or the enhanced EtOH tolerance of NO66KO . These phenotypes were rescued by their respective genomic transgenes in KDM3KO and NO66KO mutants. While we were unable to rescue lidk mutants, knockdown of lid in the nervous system recapitulated the lidk phenotype, as was observed for KDM3KO and NO66KO RNAi-mediated knockdown. CONCLUSIONS Our study reveals that the Drosophila JmjC-domain histone demethylases Lid, KDM3, NO66, and HSPBAP1 are required for normal EtOH-induced sedation and tolerance. Three of 3 tested of those 4 JmjC genes are required in the nervous system for normal alcohol-induced behavioral responses, suggesting that this gene family is an intriguing avenue for future research.
Collapse
Affiliation(s)
- Jorge H Pinzón
- Department of Psychiatry, Southwestern Medical Center, University of Texas, Dallas, Texas.,Molecular Biology, Southwestern Medical Center, University of Texas, Dallas, Texas
| | - Addison R Reed
- Department of Psychiatry, University of Utah, Salt Lake City, Utah
| | - Nevine A Shalaby
- Molecular Biology, Southwestern Medical Center, University of Texas, Dallas, Texas
| | - Michael Buszczak
- Molecular Biology, Southwestern Medical Center, University of Texas, Dallas, Texas
| | - Aylin R Rodan
- Departments of Internal Medicine/Division of Nephrology, Molecular Medicine Program, University of Utah, Salt Lake City, Utah
| | - Adrian Rothenfluh
- Department of Psychiatry, Southwestern Medical Center, University of Texas, Dallas, Texas.,Department of Psychiatry, University of Utah, Salt Lake City, Utah
| |
Collapse
|
35
|
Iacobucci GJ, Gunawardena S. Ethanol stimulates the in vivo axonal movement of neuropeptide dense-core vesicles in Drosophila motor neurons. J Neurochem 2017; 144:466-482. [PMID: 28960313 DOI: 10.1111/jnc.14230] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Revised: 09/10/2017] [Accepted: 09/18/2017] [Indexed: 01/01/2023]
Abstract
Proper neuronal function requires essential biological cargoes to be packaged within membranous vesicles and transported, intracellularly, through the extensive outgrowth of axonal and dendritic fibers. The precise spatiotemporal movement of these cargoes is vital for neuronal survival and, thus, is highly regulated. In this study we test how the axonal movement of a neuropeptide-containing dense-core vesicle (DCV) responds to alcohol stressors. We found that ethanol induces a strong anterograde bias in vesicle movement. Low doses of ethanol stimulate the anterograde movement of neuropeptide-DCV while high doses inhibit bi-directional movement. This process required the presence of functional kinesin-1 motors as reduction in kinesin prevented the ethanol-induced stimulation of the anterograde movement of neuropeptide-DCV. Furthermore, expression of inactive glycogen synthase kinase 3 (GSK-3β) also prevented ethanol-induced stimulation of neuropeptide-DCV movement, similar to pharmacological inhibition of GSK-3β with lithium. Conversely, inhibition of PI3K/AKT signaling with wortmannin led to a partial prevention of ethanol-stimulated transport of neuropeptide-DCV. Taken together, we conclude that GSK-3β signaling mediates the stimulatory effects of ethanol. Therefore, our study provides new insight into the physiological response of the axonal movement of neuropeptide-DCV to exogenous stressors. Cover Image for this Issue: doi: 10.1111/jnc.14165.
Collapse
Affiliation(s)
- Gary J Iacobucci
- Department of Biological Sciences, the State University of New York at Buffalo, Buffalo, New York, USA
| | - Shermali Gunawardena
- Department of Biological Sciences, the State University of New York at Buffalo, Buffalo, New York, USA
| |
Collapse
|
36
|
|
37
|
Velazquez-Ulloa NA. A Drosophila model for developmental nicotine exposure. PLoS One 2017; 12:e0177710. [PMID: 28498868 PMCID: PMC5428972 DOI: 10.1371/journal.pone.0177710] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 05/02/2017] [Indexed: 01/08/2023] Open
Abstract
Despite the known health risks of tobacco smoking, many people including pregnant women continue smoking. The effects of developmental nicotine exposure are known, but the underlying mechanisms are not well understood. Drosophila melanogaster is a model organism that can be used for uncovering genetic and molecular mechanisms for drugs of abuse. Here I show that Drosophila can be a model to elucidate the mechanisms for nicotine’s effects on a developing organism. Drosophila reared on nicotine food display developmental and behavioral effects similar to those in mammals including decreased survival and weight, increased developmental time, and decreased sensitivity to acute nicotine and ethanol. The Drosophila nicotinic acetylcholine receptor subunit alpha 7 (Dα7) mediates some of these effects. A novel role for Dα7 on ethanol sedation in Drosophila is also shown. Future research taking advantage of the genetic and molecular tools for Drosophila will allow additional discovery of the mechanisms behind the effects of nicotine during development.
Collapse
|
38
|
Colville AM, Iancu OD, Oberbeck DL, Darakjian P, Zheng CL, Walter NAR, Harrington CA, Searles RP, McWeeney S, Hitzemann RJ. Effects of selection for ethanol preference on gene expression in the nucleus accumbens of HS-CC mice. GENES BRAIN AND BEHAVIOR 2017; 16:462-471. [PMID: 28058793 DOI: 10.1111/gbb.12367] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 12/16/2016] [Accepted: 01/03/2017] [Indexed: 12/15/2022]
Abstract
Previous studies on changes in murine brain gene expression associated with the selection for ethanol preference have used F2 intercross or heterogeneous stock (HS) founders, derived from standard laboratory strains. However, these populations represent only a small proportion of the genetic variance available in Mus musculus. To investigate a wider range of genetic diversity, we selected mice for ethanol preference using an HS derived from the eight strains of the collaborative cross. These HS mice were selectively bred (four generations) for high and low ethanol preference. The nucleus accumbens shell of naive S4 mice was interrogated using RNA sequencing (RNA-Seq). Gene networks were constructed using the weighted gene coexpression network analysis assessing both coexpression and cosplicing. Selection targeted one of the network coexpression modules (greenyellow) that was significantly enriched in genes associated with receptor signaling activity including Chrna7, Grin2a, Htr2a and Oprd1. Connectivity in the module as measured by changes in the hub nodes was significantly reduced in the low preference line. Of particular interest was the observation that selection had marked effects on a large number of cell adhesion molecules, including cadherins and protocadherins. In addition, the coexpression data showed that selection had marked effects on long non-coding RNA hub nodes. Analysis of the cosplicing network data showed a significant effect of selection on a large cluster of Ras GTPase-binding genes including Cdkl5, Cyfip1, Ndrg1, Sod1 and Stxbp5. These data in part support the earlier observation that preference is linked to Ras/Mapk pathways.
Collapse
Affiliation(s)
- A M Colville
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA
| | - O D Iancu
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA
| | - D L Oberbeck
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA
| | - P Darakjian
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA
| | - C L Zheng
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA
| | - N A R Walter
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA
| | - C A Harrington
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA
| | - R P Searles
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA
| | - S McWeeney
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA
| | - R J Hitzemann
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA.,Research Service, Portland Veterans Affairs Medical Center, Portland, OR, USA
| |
Collapse
|
39
|
Park A, Ghezzi A, Wijesekera TP, Atkinson NS. Genetics and genomics of alcohol responses in Drosophila. Neuropharmacology 2017; 122:22-35. [PMID: 28161376 DOI: 10.1016/j.neuropharm.2017.01.032] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2016] [Revised: 01/24/2017] [Accepted: 01/29/2017] [Indexed: 02/07/2023]
Abstract
Drosophila melanogaster has become a significant model organism for alcohol research. In flies, a rich variety of behaviors can be leveraged for identifying genes affecting alcohol responses and adaptations. Furthermore, almost all genes can be easily genetically manipulated. Despite the great evolutionary distance between flies and mammals, many of the same genes have been implicated in strikingly similar alcohol-induced behaviors. A major problem in medical research today is that it is difficult to extrapolate from any single model system to humans. Strong evolutionary conservation of a mechanistic response between distantly related organisms, such as flies and mammals, is a powerful predictor that conservation will continue all the way to humans. This review describes the state of the Drosophila alcohol research field. It describes common alcohol behavioral assays, the independent origins of resistance and tolerance, the results of classical genetic screens and candidate gene analysis, and the outcomes of recent genomics studies employing GWAS, transcriptome, miRNA, and genome-wide histone acetylation surveys. This article is part of the Special Issue entitled "Alcoholism".
Collapse
Affiliation(s)
- Annie Park
- Department of Neuroscience and The Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX, United States
| | - Alfredo Ghezzi
- Department of Biology, University of Puerto Rico, Rio Piedras. San Juan, PR, United States
| | - Thilini P Wijesekera
- Department of Neuroscience and The Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX, United States
| | - Nigel S Atkinson
- Department of Neuroscience and The Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX, United States.
| |
Collapse
|
40
|
Cerebellum Transcriptome of Mice Bred for High Voluntary Activity Offers Insights into Locomotor Control and Reward-Dependent Behaviors. PLoS One 2016; 11:e0167095. [PMID: 27893846 PMCID: PMC5125674 DOI: 10.1371/journal.pone.0167095] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 11/07/2016] [Indexed: 12/19/2022] Open
Abstract
The role of the cerebellum in motivation and addictive behaviors is less understood than that in control and coordination of movements. High running can be a self-rewarding behavior exhibiting addictive properties. Changes in the cerebellum transcriptional networks of mice from a line selectively bred for High voluntary running (H) were profiled relative to an unselected Control (C) line. The environmental modulation of these changes was assessed both in activity environments corresponding to 7 days of Free (F) access to running wheel and to Blocked (B) access on day 7. Overall, 457 genes exhibited a significant (FDR-adjusted P-value < 0.05) genotype-by-environment interaction effect, indicating that activity genotype differences in gene expression depend on environmental access to running. Among these genes, network analysis highlighted 6 genes (Nrgn, Drd2, Rxrg, Gda, Adora2a, and Rab40b) connected by their products that displayed opposite expression patterns in the activity genotype contrast within the B and F environments. The comparison of network expression topologies suggests that selection for high voluntary running is linked to a predominant dysregulation of hub genes in the F environment that enables running whereas a dysregulation of ancillary genes is favored in the B environment that blocks running. Genes associated with locomotor regulation, signaling pathways, reward-processing, goal-focused, and reward-dependent behaviors exhibited significant genotype-by-environment interaction (e.g. Pak6, Adora2a, Drd2, and Arhgap8). Neuropeptide genes including Adcyap1, Cck, Sst, Vgf, Npy, Nts, Penk, and Tac2 and related receptor genes also exhibited significant genotype-by-environment interaction. The majority of the 183 differentially expressed genes between activity genotypes (e.g. Drd1) were under-expressed in C relative to H genotypes and were also under-expressed in B relative to F environments. Our findings indicate that the high voluntary running mouse line studied is a helpful model for understanding the molecular mechanisms in the cerebellum that influence locomotor control and reward-dependent behaviors.
Collapse
|
41
|
I Believe I Can Fly!: Use of Drosophila as a Model Organism in Neuropsychopharmacology Research. Neuropsychopharmacology 2016; 41:1439-46. [PMID: 26576740 PMCID: PMC4832023 DOI: 10.1038/npp.2015.322] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Revised: 10/14/2015] [Accepted: 10/16/2015] [Indexed: 01/03/2023]
Abstract
Neuropsychiatric disorders are of complex etiology, often including a large genetic component. In order to help identify and study the molecular and physiological mechanisms that such genes participate in, numerous animal models have been established in a variety of species. Over the past decade, this has increasingly included the vinegar fly, Drosophila melanogaster. Here, we outline why we study an invertebrate organism in the context of neuropsychiatric disorders, and we discuss how we can gain insight from studies in Drosophila. We focus on a few disorders and findings to make the larger point that modeling these diseases in flies can have both mechanistic and predictive validity. Highlighting some translational examples, we underline the fact that their brains works more like ours than one would have anticipated.
Collapse
|
42
|
Johnson JR, Rajamanoharan D, McCue HV, Rankin K, Barclay JW. Small Heat Shock Proteins Are Novel Common Determinants of Alcohol and Nicotine Sensitivity in Caenorhabditis elegans. Genetics 2016; 202:1013-27. [PMID: 26773049 PMCID: PMC4788107 DOI: 10.1534/genetics.115.185025] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 01/11/2016] [Indexed: 12/26/2022] Open
Abstract
Addiction to drugs is strongly determined by multiple genetic factors. Alcohol and nicotine produce distinct pharmacological effects within the nervous system through discrete molecular targets; yet, data from family and twin analyses support the existence of common genetic factors for addiction in general. The mechanisms underlying addiction, however, are poorly described and common genetic factors for alcohol and nicotine remain unidentified. We investigated the role that the heat shock transcription factor, HSF-1, and its downstream effectors played as common genetic modulators of sensitivity to addictive substances. Using Caenorhabditis elegans, an exemplary model organism with substance dose-dependent responses similar to mammals, we demonstrate that HSF-1 altered sensitivity to both alcohol and nicotine. Using a combination of a targeted RNAi screen of downstream factors and transgenic approaches we identified that these effects were contingent upon the constitutive neuronal expression of HSP-16.48, a small heat shock protein (HSP) homolog of human α-crystallin. Furthermore we demonstrated that the function of HSP-16.48 in drug sensitivity surprisingly was independent of chaperone activity during the heat shock stress response. Instead we identified a distinct domain within the N-terminal region of the HSP-16.48 protein that specified its function in comparison to related small HSPs. Our findings establish and characterize a novel genetic determinant underlying sensitivity to diverse addictive substances.
Collapse
Affiliation(s)
- James R Johnson
- The Physiological Laboratory, Institute of Translational Medicine, University of Liverpool, Liverpool L69 3BX, United Kingdom
| | - Dayani Rajamanoharan
- The Physiological Laboratory, Institute of Translational Medicine, University of Liverpool, Liverpool L69 3BX, United Kingdom
| | - Hannah V McCue
- The Physiological Laboratory, Institute of Translational Medicine, University of Liverpool, Liverpool L69 3BX, United Kingdom
| | - Kim Rankin
- The Physiological Laboratory, Institute of Translational Medicine, University of Liverpool, Liverpool L69 3BX, United Kingdom
| | - Jeff W Barclay
- The Physiological Laboratory, Institute of Translational Medicine, University of Liverpool, Liverpool L69 3BX, United Kingdom
| |
Collapse
|
43
|
Abstract
UNLABELLED Alcohol use disorders (AUDs) affect people at great individual and societal cost. Individuals at risk for AUDs are sensitive to alcohol's rewarding effects and/or resistant to its aversive and sedating effects. The molecular basis for these traits is poorly understood. Here, we show that p70 S6 kinase (S6k), acting downstream of the insulin receptor (InR) and the small GTPase Arf6, is a key mediator of ethanol-induced sedation in Drosophila. S6k signaling in the adult nervous system determines flies' sensitivity to sedation. Furthermore, S6k activity, measured via levels of phosphorylation (P-S6k), is a molecular marker for sedation and overall neuronal activity: P-S6k levels are decreased when neurons are silenced, as well as after acute ethanol sedation. Conversely, P-S6k levels rebound upon recovery from sedation and are increased when neuronal activity is enhanced. Reducing neural activity increases sensitivity to ethanol-induced sedation, whereas neuronal activation decreases ethanol sensitivity. These data suggest that ethanol has acute silencing effects on adult neuronal activity, which suppresses InR/Arf6/S6k signaling and results in behavioral sedation. In addition, we show that activity of InR/Arf6/S6k signaling determines flies' behavioral sensitivity to ethanol-induced sedation, highlighting this pathway in acute responses to ethanol. SIGNIFICANCE STATEMENT Genetic factors play a major role in the development of addiction. Identifying these genes and understanding their molecular mechanisms is a necessary first step in the development of targeted therapeutic intervention. Here, we show that signaling from the insulin receptor in Drosophila neurons determines flies' sensitivity to ethanol-induced sedation. We show that this signaling cascade includes the small GTPase Arf6 and S6 kinase (S6k). In addition, activity of S6k is regulated by acute ethanol exposure and by neuronal activity. S6k activity is therefore both an acute target of ethanol exposure and a regulator of ethanol's effects on behavior.
Collapse
|
44
|
De Nobrega AK, Lyons LC. Circadian Modulation of Alcohol-Induced Sedation and Recovery in Male and Female Drosophila. J Biol Rhythms 2016; 31:142-60. [PMID: 26833081 DOI: 10.1177/0748730415627067] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Delineating the factors that affect behavioral and neurological responses to alcohol is critical to facilitate measures for preventing or treating alcohol abuse. The high degree of conserved molecular and physiological processes makes Drosophila melanogaster a valuable model for investigating circadian interactions with alcohol-induced behaviors and examining sex-specific differences in alcohol sensitivity. We found that wild-type Drosophila exhibited rhythms in alcohol-induced sedation under light-dark and constant dark conditions with considerably greater alcohol exposure necessary to induce sedation during the late (subjective) day and peak sensitivity to alcohol occurring during the late (subjective) night. The circadian clock also modulated the recovery from alcohol-induced sedation with flies regaining motor control significantly faster during the late (subjective) day. As predicted, the circadian rhythms in sedation and recovery were absent in flies with a mutation in the circadian gene period or arrhythmic flies housed in constant light conditions. Flies lacking a functional circadian clock were more sensitive to the effects of alcohol with significantly longer recovery times. Similar to other animals and humans, Drosophila exhibit sex-specific differences in alcohol sensitivity. We investigated whether the circadian clock modulated the rhythms in the loss-of-righting reflex, alcohol-induced sedation, and recovery differently in males and females. We found that both sexes demonstrated circadian rhythms in the loss-of-righting reflex and sedation with the differences in alcohol sensitivity between males and females most pronounced during the late subjective day. Recovery of motor reflexes following alcohol sedation also exhibited circadian modulation in male and female flies, although the circadian clock did not modulate the difference in recovery times between the sexes. These studies provide a framework outlining how the circadian clock modulates alcohol-induced behaviors in Drosophila and identifies sexual dimorphisms in the circadian modulation of alcohol behaviors.
Collapse
Affiliation(s)
- Aliza K De Nobrega
- Department of Biological Science, Program in Neuroscience, Florida State University, Tallahassee, FL
| | - Lisa C Lyons
- Department of Biological Science, Program in Neuroscience, Florida State University, Tallahassee, FL
| |
Collapse
|
45
|
Morozova TV, Huang W, Pray VA, Whitham T, Anholt RRH, Mackay TFC. Polymorphisms in early neurodevelopmental genes affect natural variation in alcohol sensitivity in adult drosophila. BMC Genomics 2015; 16:865. [PMID: 26503115 PMCID: PMC4624176 DOI: 10.1186/s12864-015-2064-5] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 10/13/2015] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Alcohol abuse and alcoholism are significant public health problems, but the genetic basis for individual variation in alcohol sensitivity remains poorly understood. Drosophila melanogaster presents a powerful model system for dissecting the genetic underpinnings that determine individual variation in alcohol-related phenotypes. We performed genome wide association analyses for alcohol sensitivity using the sequenced, inbred lines of the D. melanogaster Genetic Reference Panel (DGRP) together with extreme QTL mapping in an advanced intercross population derived from sensitive and resistant DGRP lines. RESULTS The DGRP harbors substantial genetic variation for alcohol sensitivity and tolerance. We identified 247 candidate genes affecting alcohol sensitivity in the DGRP or the DGRP-derived advanced intercross population, some of which met a Bonferroni-corrected significance threshold, while others occurred among the top candidate genes associated with variation in alcohol sensitivity in multiple analyses. Among these were candidate genes associated with development and function of the nervous system, including several genes in the Dopamine decarboxylase (Ddc) cluster involved in catecholamine synthesis. We found that 58 of these genes formed a genetic interaction network. We verified candidate genes using mutational analysis, targeted gene disruption through RNAi knock-down and transcriptional profiling. Two-thirds of the candidate genes have been implicated in previous Drosophila, mouse and human studies of alcohol-related phenotypes. CONCLUSIONS Individual variation in alcohol sensitivity in Drosophila is highly polygenic and in part determined by variation in evolutionarily conserved signaling pathways that are associated with catecholamine neurotransmitter biosynthesis and early development of the nervous system.
Collapse
Affiliation(s)
- Tatiana V Morozova
- Department of Biological Sciences, W. M. Keck Center for Behavioral Biology and Program in Genetics, North Carolina State University, Box 7614, Raleigh, NC, 27695, USA
| | - Wen Huang
- Department of Biological Sciences, W. M. Keck Center for Behavioral Biology and Program in Genetics, North Carolina State University, Box 7614, Raleigh, NC, 27695, USA
| | - Victoria A Pray
- Department of Biological Sciences, W. M. Keck Center for Behavioral Biology and Program in Genetics, North Carolina State University, Box 7614, Raleigh, NC, 27695, USA
| | - Thomas Whitham
- Department of Biological Sciences, W. M. Keck Center for Behavioral Biology and Program in Genetics, North Carolina State University, Box 7614, Raleigh, NC, 27695, USA
- Department of Biochemistry and Physiology, School of Bioscience and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, GU2 7XH, UK
| | - Robert R H Anholt
- Department of Biological Sciences, W. M. Keck Center for Behavioral Biology and Program in Genetics, North Carolina State University, Box 7614, Raleigh, NC, 27695, USA
| | - Trudy F C Mackay
- Department of Biological Sciences, W. M. Keck Center for Behavioral Biology and Program in Genetics, North Carolina State University, Box 7614, Raleigh, NC, 27695, USA.
| |
Collapse
|
46
|
Ojelade SA, Acevedo SF, Kalahasti G, Rodan AR, Rothenfluh A. RhoGAP18B Isoforms Act on Distinct Rho-Family GTPases and Regulate Behavioral Responses to Alcohol via Cofilin. PLoS One 2015; 10:e0137465. [PMID: 26366560 PMCID: PMC4569326 DOI: 10.1371/journal.pone.0137465] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2015] [Accepted: 08/17/2015] [Indexed: 11/18/2022] Open
Abstract
Responses to the effects of ethanol are highly conserved across organisms, with reduced responses to the sedating effects of ethanol being predictive of increased risk for human alcohol dependence. Previously, we described that regulators of actin dynamics, such as the Rho-family GTPases Rac1, Rho1, and Cdc42, alter Drosophila's sensitivity to ethanol-induced sedation. The GTPase activating protein RhoGAP18B also affects sensitivity to ethanol. To better understand how different RhoGAP18B isoforms affect ethanol sedation, we examined them for their effects on cell shape, GTP-loading of Rho-family GTPase, activation of the actin-severing cofilin, and actin filamentation. Our results suggest that the RhoGAP18B-PA isoform acts on Cdc42, while PC and PD act via Rac1 and Rho1 to activate cofilin. In vivo, a loss-of-function mutation in the cofilin-encoding gene twinstar leads to reduced ethanol-sensitivity and acts in concert with RhoGAP18B. Different RhoGAP18B isoforms, therefore, act on distinct subsets of Rho-family GTPases to modulate cofilin activity, actin dynamics, and ethanol-induced behaviors.
Collapse
Affiliation(s)
- Shamsideen A. Ojelade
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, TX, United States of America
- Program in Neuroscience, UT Southwestern Medical Center, Dallas, TX, United States of America
| | - Summer F. Acevedo
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, TX, United States of America
| | - Geetha Kalahasti
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, TX, United States of America
| | - Aylin R. Rodan
- Division of Nephrology, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, United States of America
| | - Adrian Rothenfluh
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, TX, United States of America
- Program in Neuroscience, UT Southwestern Medical Center, Dallas, TX, United States of America
- * E-mail:
| |
Collapse
|
47
|
Grotewiel M, Bettinger JC. Drosophila and Caenorhabditis elegans as Discovery Platforms for Genes Involved in Human Alcohol Use Disorder. Alcohol Clin Exp Res 2015; 39:1292-311. [PMID: 26173477 PMCID: PMC4656040 DOI: 10.1111/acer.12785] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Accepted: 05/18/2015] [Indexed: 01/08/2023]
Abstract
BACKGROUND Despite the profound clinical significance and strong heritability of alcohol use disorder (AUD), we do not yet have a comprehensive understanding of the naturally occurring genetic variance within the human genome that drives its development. This lack of understanding is likely to be due in part to the large phenotypic and genetic heterogeneities that underlie human AUD. As a complement to genetic studies in humans, many laboratories are using the invertebrate model organisms (iMOs) Drosophila melanogaster (fruit fly) and Caenorhabditis elegans (nematode worm) to identify genetic mechanisms that influence the effects of alcohol (ethanol) on behavior. While these extremely powerful models have identified many genes that influence the behavioral responses to alcohol, in most cases it has remained unclear whether results from behavioral-genetic studies in iMOs are directly applicable to understanding the genetic basis of human AUD. METHODS In this review, we critically evaluate the utility of the fly and worm models for identifying genes that influence AUD in humans. RESULTS Based on results published through early 2015, studies in flies and worms have identified 91 and 50 genes, respectively, that influence 1 or more aspects of behavioral responses to alcohol. Collectively, these fly and worm genes correspond to 293 orthologous genes in humans. Intriguingly, 51 of these 293 human genes have been implicated in AUD by at least 1 study in human populations. CONCLUSIONS Our analyses strongly suggest that the Drosophila and C. elegans models have considerable utility for identifying orthologs of genes that influence human AUD.
Collapse
Affiliation(s)
- Mike Grotewiel
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, Virginia
- Virginia Commonwealth University Alcohol Research Center, Richmond, Virginia
| | - Jill C Bettinger
- Department of Pharmacology and Toxicology , Virginia Commonwealth University, Richmond, Virginia
- Virginia Commonwealth University Alcohol Research Center, Richmond, Virginia
| |
Collapse
|
48
|
Abstract
Alcohol abuse is highly prevalent, but little is understood about the molecular causes. Here, we report that Ras suppressor 1 (Rsu1) affects ethanol consumption in flies and humans. Drosophila lacking Rsu1 show reduced sensitivity to ethanol-induced sedation. We show that Rsu1 is required in the adult nervous system for normal sensitivity and that it acts downstream of the integrin cell adhesion molecule and upstream of the Ras-related C3 botulinum toxin substrate 1 (Rac1) GTPase to regulate the actin cytoskeleton. In an ethanol preference assay, global loss of Rsu1 causes high naïve preference. In contrast, flies lacking Rsu1 only in the mushroom bodies of the brain show normal naïve preference but then fail to acquire ethanol preference like normal flies. Rsu1 is, thus, required in distinct neurons to modulate naïve and acquired ethanol preference. In humans, we find that polymorphisms in RSU1 are associated with brain activation in the ventral striatum during reward anticipation in adolescents and alcohol consumption in both adolescents and adults. Together, these data suggest a conserved role for integrin/Rsu1/Rac1/actin signaling in modulating reward-related phenotypes, including ethanol consumption, across phyla.
Collapse
|
49
|
Wetherill L, Agrawal A, Kapoor M, Bertelsen S, Bierut LJ, Brooks A, Dick D, Hesselbrock M, Hesselbrock V, Koller DL, Le N, Nurnberger JI, Salvatore JE, Schuckit M, Tischfield JA, Wang JC, Xuei X, Edenberg HJ, Porjesz B, Bucholz K, Goate AM, Foroud T. Association of substance dependence phenotypes in the COGA sample. Addict Biol 2015; 20:617-27. [PMID: 24832863 PMCID: PMC4233207 DOI: 10.1111/adb.12153] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Alcohol and drug use disorders are individually heritable (50%). Twin studies indicate that alcohol and substance use disorders share common genetic influences, and therefore may represent a more heritable form of addiction and thus be more powerful for genetic studies. This study utilized data from 2322 subjects from 118 European-American families in the Collaborative Study on the Genetics of Alcoholism sample to conduct genome-wide association analysis of a binary and a continuous index of general substance dependence liability. The binary phenotype (ANYDEP) was based on meeting lifetime criteria for any DSM-IV dependence on alcohol, cannabis, cocaine or opioids. The quantitative trait (QUANTDEP) was constructed from factor analysis based on endorsement across the seven DSM-IV criteria for each of the four substances. Heritability was estimated to be 54% for ANYDEP and 86% for QUANTDEP. One single-nucleotide polymorphism (SNP), rs2952621 in the uncharacterized gene LOC151121 on chromosome 2, was associated with ANYDEP (P = 1.8 × 10(-8) ), with support from surrounding imputed SNPs and replication in an independent sample [Study of Addiction: Genetics and Environment (SAGE); P = 0.02]. One SNP, rs2567261 in ARHGAP28 (Rho GTPase-activating protein 28), was associated with QUANTDEP (P = 3.8 × 10(-8) ), and supported by imputed SNPs in the region, but did not replicate in an independent sample (SAGE; P = 0.29). The results of this study provide evidence that there are common variants that contribute to the risk for a general liability to substance dependence.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Nhung Le
- Washington University School of Medicine
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Sandhu S, Kollah AP, Lewellyn L, Chan RF, Grotewiel M. An inexpensive, scalable behavioral assay for measuring ethanol sedation sensitivity and rapid tolerance in Drosophila. J Vis Exp 2015:52676. [PMID: 25939022 PMCID: PMC4423423 DOI: 10.3791/52676] [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] [Indexed: 01/19/2023] Open
Abstract
Alcohol use disorder (AUD) is a serious health challenge. Despite a large hereditary component to AUD, few genes have been unambiguously implicated in their etiology. The fruit fly, Drosophila melanogaster, is a powerful model for exploring molecular-genetic mechanisms underlying alcohol-related behaviors and therefore holds great promise for identifying and understanding the function of genes that influence AUD. The use of the Drosophila model for these types of studies depends on the availability of assays that reliably measure behavioral responses to ethanol. This report describes an assay suitable for assessing ethanol sensitivity and rapid tolerance in flies. Ethanol sensitivity measured in this assay is influenced by the volume and concentration of ethanol used, a variety of previously reported genetic manipulations, and also the length of time the flies are housed without food immediately prior to testing. In contrast, ethanol sensitivity measured in this assay is not affected by the vigor of fly handling, sex of the flies, and supplementation of growth medium with antibiotics or live yeast. Three different methods for quantitating ethanol sensitivity are described, all leading to essentially indistinguishable ethanol sensitivity results. The scalable nature of this assay, combined with its overall simplicity to set-up and relatively low expense, make it suitable for small and large scale genetic analysis of ethanol sensitivity and rapid tolerance in Drosophila.
Collapse
Affiliation(s)
- Simran Sandhu
- Department of Human and Molecular Genetics, Virginia Commonwealth University
| | - Arnavaz P Kollah
- Department of Human and Molecular Genetics, Virginia Commonwealth University
| | - Lara Lewellyn
- Department of Human and Molecular Genetics, Virginia Commonwealth University
| | - Robin F Chan
- Department of Human and Molecular Genetics, Virginia Commonwealth University
| | - Mike Grotewiel
- Department of Human and Molecular Genetics, Virginia Commonwealth University; VCU Alcohol Research Center, Virginia Commonwealth University;
| |
Collapse
|