1
|
Erofeeva N, Galstyan DS, Yang L, Strekalova T, Lim LW, de Abreu MS, Golushko NI, Stewart AM, Kalueff AV. Developing zebrafish models of Notch-related CNS pathologies. Neurosci Biobehav Rev 2025; 170:106059. [PMID: 39929383 DOI: 10.1016/j.neubiorev.2025.106059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 02/01/2025] [Accepted: 02/06/2025] [Indexed: 02/20/2025]
Abstract
Notch signaling is an evolutionarily conserved cellular pathway that regulates various stem cell functions, including fate determination, differentiation, proliferation, and apoptosis. This crucial signaling mechanism also plays an important role in the brain, regulating neurogenesis, cell differentiation, and homeostasis, whereas disrupted Notch signaling is linked to various neurodegenerative diseases and brain cancers. Here, we review the central nervous system (CNS) pathologies associated with aberrant Notch signaling, and summarize the available experimental (animal) models used to study these pathologies, with a special focus on zebrafish (Danio rerio). As genetic, pharmacological, and behavioral models in zebrafish have significantly advanced our understanding of Notch-related CNS disorders, future research is expected to further link Notch signaling to brain disorders and, eventually, lead to their more specific and targeted therapeuties.
Collapse
Affiliation(s)
- Natalia Erofeeva
- St. Petersburg State University, St Petersburg, Russia; Institute of Translational Biomedicine, St. Petersburg State University, St Petersburg, Russia; Institute of Experimental Medicine, Almazov National Medical Research Center, Ministry of Healthcare of Russian Federation, St Petersburg, Russia; Neurobiology Program, Sirius University of Science and Technology, Sirius, Russia
| | - David S Galstyan
- Institute of Translational Biomedicine, St. Petersburg State University, St Petersburg, Russia; Institute of Experimental Medicine, Almazov National Medical Research Center, Ministry of Healthcare of Russian Federation, St Petersburg, Russia
| | - Longen Yang
- Department of Biosciences and Bioinformatics, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China
| | - Tatiana Strekalova
- Peoples Friendship University of Russia (RUDN University) and Department of Normal Physiology, Sechenov University, Moscow, Russia
| | - Lee Wei Lim
- Department of Biosciences and Bioinformatics, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China; Suzhou Municipal Key Laboratory on Neurobiology and Cell Signaling, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China
| | - Murilo S de Abreu
- Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre, Porto Alegre, Brazil; Western Caspian University, Baku, Azerbaijan.
| | - Nikita I Golushko
- Institute of Translational Biomedicine, St. Petersburg State University, St Petersburg, Russia; Institute of Experimental Medicine, Almazov National Medical Research Center, Ministry of Healthcare of Russian Federation, St Petersburg, Russia
| | - Adam Michael Stewart
- The International Zebrafish Neuroscience Research Consortium (ZNRC), New Orleans, USA
| | - Allan V Kalueff
- Institute of Translational Biomedicine, St. Petersburg State University, St Petersburg, Russia; Institute of Experimental Medicine, Almazov National Medical Research Center, Ministry of Healthcare of Russian Federation, St Petersburg, Russia; Department of Biosciences and Bioinformatics, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China; Suzhou Municipal Key Laboratory on Neurobiology and Cell Signaling, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China; Neurobiology Program, Sirius University of Science and Technology, Sirius, Russia.
| |
Collapse
|
2
|
Hiramatsu S, Saito K, Kondo S, Katow H, Yamagata N, Wu CF, Tanimoto H. Synaptic enrichment and dynamic regulation of the two opposing dopamine receptors within the same neurons. eLife 2025; 13:RP98358. [PMID: 39882849 PMCID: PMC11781798 DOI: 10.7554/elife.98358] [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] [Indexed: 01/31/2025] Open
Abstract
Dopamine can play opposing physiological roles depending on the receptor subtype. In the fruit fly Drosophila melanogaster, Dop1R1 and Dop2R encode the D1- and D2-like receptors, respectively, and are reported to oppositely regulate intracellular cAMP levels. Here, we profiled the expression and subcellular localization of endogenous Dop1R1 and Dop2R in specific cell types in the mushroom body circuit. For cell-type-specific visualization of endogenous proteins, we employed reconstitution of split-GFP tagged to the receptor proteins. We detected dopamine receptors at both presynaptic and postsynaptic sites in multiple cell types. Quantitative analysis revealed enrichment of both receptors at the presynaptic sites, with Dop2R showing a greater degree of localization than Dop1R1. The presynaptic localization of Dop1R1 and Dop2R in dopamine neurons suggests dual feedback regulation as autoreceptors. Furthermore, we discovered a starvation-dependent, bidirectional modulation of the presynaptic receptor expression in the protocerebral anterior medial (PAM) and posterior lateral 1 (PPL1) clusters, two distinct subsets of dopamine neurons, suggesting their roles in regulating appetitive behaviors. Our results highlight the significance of the co-expression of the two opposing dopamine receptors in the spatial and conditional regulation of dopamine responses in neurons.
Collapse
Affiliation(s)
- Shun Hiramatsu
- Graduate School of Life Sciences, Tohoku UniversitySendaiJapan
| | - Kokoro Saito
- Graduate School of Life Sciences, Tohoku UniversitySendaiJapan
| | - Shu Kondo
- Department of Biological Science and Technology, Faculty of Advanced Engineering, Tokyo University of ScienceTokyoJapan
| | - Hidetaka Katow
- Department of Cell Biology, New York UniversityNew YorkUnited States
| | - Nobuhiro Yamagata
- Faculty and Graduate School of Engineering Science, Akita UniversityAkitaJapan
| | - Chun-Fang Wu
- Department of Biology, University of IowaIowa CityUnited States
| | - Hiromu Tanimoto
- Graduate School of Life Sciences, Tohoku UniversitySendaiJapan
| |
Collapse
|
3
|
Suárez-Grimalt R, Grunwald Kadow IC, Scheunemann L. An integrative sensor of body states: how the mushroom body modulates behavior depending on physiological context. Learn Mem 2024; 31:a053918. [PMID: 38876486 PMCID: PMC11199956 DOI: 10.1101/lm.053918.124] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 04/08/2024] [Indexed: 06/16/2024]
Abstract
The brain constantly compares past and present experiences to predict the future, thereby enabling instantaneous and future behavioral adjustments. Integration of external information with the animal's current internal needs and behavioral state represents a key challenge of the nervous system. Recent advancements in dissecting the function of the Drosophila mushroom body (MB) at the single-cell level have uncovered its three-layered logic and parallel systems conveying positive and negative values during associative learning. This review explores a lesser-known role of the MB in detecting and integrating body states such as hunger, thirst, and sleep, ultimately modulating motivation and sensory-driven decisions based on the physiological state of the fly. State-dependent signals predominantly affect the activity of modulatory MB input neurons (dopaminergic, serotoninergic, and octopaminergic), but also induce plastic changes directly at the level of the MB intrinsic and output neurons. Thus, the MB emerges as a tightly regulated relay station in the insect brain, orchestrating neuroadaptations due to current internal and behavioral states leading to short- but also long-lasting changes in behavior. While these adaptations are crucial to ensure fitness and survival, recent findings also underscore how circuit motifs in the MB may reflect fundamental design principles that contribute to maladaptive behaviors such as addiction or depression-like symptoms.
Collapse
Affiliation(s)
- Raquel Suárez-Grimalt
- Institute for Biology/Genetics, Freie Universität Berlin, 14195 Berlin, Germany
- Institut für Neurophysiologie and NeuroCure Cluster of Excellence, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
| | | | - Lisa Scheunemann
- Institute for Biology/Genetics, Freie Universität Berlin, 14195 Berlin, Germany
- Institut für Neurophysiologie and NeuroCure Cluster of Excellence, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
| |
Collapse
|
4
|
Carvalho L, Lasek AW. It is not just about transcription: involvement of brain RNA splicing in substance use disorders. J Neural Transm (Vienna) 2024; 131:495-503. [PMID: 38396082 PMCID: PMC11055753 DOI: 10.1007/s00702-024-02740-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Accepted: 01/04/2024] [Indexed: 02/25/2024]
Abstract
Alternative splicing is a co-transcriptional process that significantly contributes to the molecular landscape of the cell. It plays a multifaceted role in shaping gene transcription, protein diversity, and functional adaptability in response to environmental cues. Recent studies demonstrate that drugs of abuse have a profound impact on alternative splicing patterns within different brain regions. Drugs like alcohol and cocaine modify the expression of genes responsible for encoding splicing factors, thereby influencing alternative splicing of crucial genes involved in neurotransmission, neurogenesis, and neuroinflammation. Notable examples of these alterations include alcohol-induced changes in splicing factors such as HSPA6 and PCBP1, as well as cocaine's impact on PTBP1 and SRSF11. Beyond the immediate effects of drug exposure, recent research has shed light on the role of alternative splicing in contributing to the risk of substance use disorders (SUDs). This is exemplified by exon skipping events in key genes like ELOVL7, which can elevate the risk of alcohol use disorder. Lastly, drugs of abuse can induce splicing alterations through epigenetic modifications. For example, cocaine exposure leads to alterations in levels of trimethylated lysine 36 of histone H3, which exhibits a robust association with alternative splicing and serves as a reliable predictor for exon exclusion. In summary, alternative splicing has emerged as a critical player in the complex interplay between drugs of abuse and the brain, offering insights into the molecular underpinnings of SUDs.
Collapse
Affiliation(s)
- Luana Carvalho
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, 1220 E. Broad ST, Box 980613, Richmond, VA, 23298, USA.
| | - Amy W Lasek
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, 1220 E. Broad ST, Box 980613, Richmond, VA, 23298, USA
| |
Collapse
|
5
|
Larnerd C, Kachewar N, Wolf FW. Drosophila learning and memory centers and the actions of drugs of abuse. Learn Mem 2024; 31:a053815. [PMID: 38862166 PMCID: PMC11199947 DOI: 10.1101/lm.053815.123] [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] [Received: 02/12/2024] [Accepted: 03/27/2024] [Indexed: 06/13/2024]
Abstract
Drug addiction and the circuitry for learning and memory are intimately intertwined. Drugs of abuse create strong, inappropriate, and lasting memories that contribute to many of their destructive properties, such as continued use despite negative consequences and exceptionally high rates of relapse. Studies in Drosophila melanogaster are helping us understand how drugs of abuse, especially alcohol, create memories at the level of individual neurons and in the circuits where they function. Drosophila is a premier organism for identifying the mechanisms of learning and memory. Drosophila also respond to drugs of abuse in ways that remarkably parallel humans and rodent models. An emerging consensus is that, for alcohol, the mushroom bodies participate in the circuits that control acute drug sensitivity, not explicitly associative forms of plasticity such as tolerance, and classical associative memories of their rewarding and aversive properties. Moreover, it is becoming clear that drugs of abuse use the mushroom body circuitry differently from other behaviors, potentially providing a basis for their addictive properties.
Collapse
Affiliation(s)
- Caleb Larnerd
- Quantitative and Systems Biology Graduate Group, University of California, Merced, California 95343, USA
| | - Neha Kachewar
- Department of Molecular and Cell Biology, University of California, Merced, California 95343, USA
- Health Sciences Research Institute, University of California, Merced, California 95343, USA
| | - Fred W Wolf
- Quantitative and Systems Biology Graduate Group, University of California, Merced, California 95343, USA
- Department of Molecular and Cell Biology, University of California, Merced, California 95343, USA
| |
Collapse
|
6
|
Li L, Luo J, Zhu Z, Wang P, Xu Q, Chang B, Wang D, Yu L, Lu X, Zhou J, Chen Q, Zuo D. Macrophage-expressed SRA ameliorates alcohol-induced liver injury by suppressing S-glutathionylation of Notch1 via recruiting thioredoxin. J Leukoc Biol 2024; 115:322-333. [PMID: 37726110 DOI: 10.1093/jleuko/qiad110] [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: 08/27/2022] [Revised: 06/21/2023] [Accepted: 08/09/2023] [Indexed: 09/21/2023] Open
Abstract
Scavenger receptor A (SRA) is preferentially expressed in macrophages and implicated as a multifunctional pattern recognition receptor for innate immunity. Hepatic macrophages play a primary role in the pathogenesis of alcoholic liver disease. Herein, we observed that SRA expression was significantly increased in the liver tissues of mice with alcohol-related liver injury. SRA-deficient (SRA-/-) mice developed more severe alcohol-induced liver disease than wild-type mice. Enhanced liver inflammation existed in alcohol-challenged SRA-/- mice and was associated with increased Notch activation in hepatic macrophages compared with wild-type control animals. Mechanistically, SRA directly bound with Notch1 and suppressed its S-glutathionylation, thereby inhibiting Notch pathway activation. Further, we determined that the SRA interacted with thioredoxin-1 (Trx-1), a redox-active protein. SRA inhibited Trx-1 dimerization and facilitated the interaction of Trx-1 with Notch1. Application of a Trx-1-specific inhibitory agent during macrophage stimulation abolished SRA-mediated regulation of the Notch pathway and its downstream targets. In summary, our study revealed that SRA plays a critical role in macrophage inflammatory response by targeting Notch1 for its glutathionylation. SRA-mediated negative regulation of Notch activation might serve as a novel therapeutic strategy for alcohol-induced liver injury.
Collapse
Affiliation(s)
- Lei Li
- Institute of Immunology, Guangdong Provincial Key Laboratory of Immune Regulation and Immunotherapy, School of Laboratory Medicine and Biotechnology, Southern Medical University, No.1023 South Shatai Road, Baiyun District, Guangzhou, Guangdong 510515, China
| | - Jialiang Luo
- Institute of Immunology, Guangdong Provincial Key Laboratory of Immune Regulation and Immunotherapy, School of Laboratory Medicine and Biotechnology, Southern Medical University, No.1023 South Shatai Road, Baiyun District, Guangzhou, Guangdong 510515, China
- Department of Dermatology, Fifth Hospital of Southern Medical University, Southern Medical University, No.566 Congcheng Avenue, Conghua District, Guangzhou, Guangdong 510515, China
| | - Zhengyumeng Zhu
- Institute of Immunology, Guangdong Provincial Key Laboratory of Immune Regulation and Immunotherapy, School of Laboratory Medicine and Biotechnology, Southern Medical University, No.1023 South Shatai Road, Baiyun District, Guangzhou, Guangdong 510515, China
| | - Ping Wang
- Department of Medical Research, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, No.106 Second Zhongshan Road, Yuexiu District, Guangzhou, Guangdong 510080, China
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, No.1023 South Shatai Road, Baiyun District, Guangzhou, Guangdong 510515, China
| | - Qishan Xu
- Institute of Immunology, Guangdong Provincial Key Laboratory of Immune Regulation and Immunotherapy, School of Laboratory Medicine and Biotechnology, Southern Medical University, No.1023 South Shatai Road, Baiyun District, Guangzhou, Guangdong 510515, China
| | - Bo Chang
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, No.1023 South Shatai Road, Baiyun District, Guangzhou, Guangdong 510515, China
| | - Di Wang
- Department of Dermatology, Dermatology Hospital of Southern Medical University, Southern Medical University, No.2 Lujing Road, Yuexiu District, Guangzhou, Guangdong 510091, China
| | - Lu Yu
- Institute of Immunology, Guangdong Provincial Key Laboratory of Immune Regulation and Immunotherapy, School of Laboratory Medicine and Biotechnology, Southern Medical University, No.1023 South Shatai Road, Baiyun District, Guangzhou, Guangdong 510515, China
| | - Xiao Lu
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, No.1023 South Shatai Road, Baiyun District, Guangzhou, Guangdong 510515, China
| | - Jia Zhou
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, No.1023 South Shatai Road, Baiyun District, Guangzhou, Guangdong 510515, China
| | - Qingyun Chen
- Department of Medical Research, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, No.106 Second Zhongshan Road, Yuexiu District, Guangzhou, Guangdong 510080, China
| | - Daming Zuo
- Institute of Immunology, Guangdong Provincial Key Laboratory of Immune Regulation and Immunotherapy, School of Laboratory Medicine and Biotechnology, Southern Medical University, No.1023 South Shatai Road, Baiyun District, Guangzhou, Guangdong 510515, China
| |
Collapse
|
7
|
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
|
8
|
Liu S, Zhang Z, Li L, Yao L, Ma Z, Li J. ADAM10- and γ-secretase-dependent cleavage of the transmembrane protein PTPRT attenuates neurodegeneration in the mouse model of Alzheimer's disease. FASEB J 2023; 37:e22734. [PMID: 36583697 DOI: 10.1096/fj.202201396r] [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: 08/25/2022] [Revised: 11/20/2022] [Accepted: 12/14/2022] [Indexed: 12/31/2022]
Abstract
PTPRT (receptor-type tyrosine-protein phosphatase T), a brain-specific type 1 transmembrane protein, plays an important role in neurodevelopment and synapse formation. However, whether abnormal PTPRT signaling is associated with Alzheimer's disease (AD) remains elusive. Here, we report that Ptprt mRNA expression is found to be downregulated in the brains of both human and mouse models of AD. We further identified that the PTPRT intracellular domain (PICD), which is released by ADAM10- and γ-secretase-dependent cleavage of PTPRT, efficiently translocates to the nucleus via a conserved nuclear localization signal (NLS). We show that inhibition of nuclear translocation of PICD leads to an accumulation of phosphorylated signal transducer and activator of transcription 3 (pSTAT3), a substrate of PTPRT-eventually resulting in neuronal cell death. Consistently, RNA sequencing reveals that overexpression of PICD leads to changes in the expression of genes that are functionally associated with synapse formation, cell adhesion, and protein dephosphorylation. Moreover, overexpression of PICD not only decreases the level of phospho-STAT3Y705 and amyloid β production in the hippocampus of APP/PS1 mice but also partially improves synaptic function and behavioral deficits in this mouse model of AD. These findings suggest that a novel role of the ADAM 10- and γ-secretase-dependent cleavage of PTPRT may alleviate the AD-like neurodegenerative processes.
Collapse
Affiliation(s)
- Siling Liu
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China
| | - Zhongyu Zhang
- National Institute on Drug Dependence, Peking University, Beijing, China
| | - Lianwei Li
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Li Yao
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China
| | - Zhanshan Ma
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Jiali Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,National Institute on Drug Dependence, Peking University, Beijing, China.,IDG/McGovern Institute for Brain Research, Peking University, Beijing, China.,Kunming Primate Research Center of the Chinese Academy of Sciences, Kunming, China.,National Research Facility for Phenotypic and Genetic Analysis of Model Animals, Kunming Institute of Zoology, the Chinese Academy of Sciences, Kunming, China
| |
Collapse
|
9
|
Wilson A, Periandri EM, Sievers M, Petruccelli E. Drosophila Stat92E Signaling Following Pre-exposure to Ethanol. Neurosci Insights 2023; 18:26331055221146755. [PMID: 36643884 PMCID: PMC9834942 DOI: 10.1177/26331055221146755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Accepted: 12/06/2022] [Indexed: 01/11/2023] Open
Abstract
Repeated exposure to alcohol alters neuromolecular signaling that influences acute and long-lasting behaviors underlying Alcohol Use Disorder (AUD). Recent animal model research has implicated changes in the conserved JAK/STAT pathway, a signaling pathway classically associated with development and the innate immune system. How ethanol exposure impacts STAT signaling within neural cells is currently unclear. Here, we investigated the role of Drosophila Stat92E in ethanol-induced locomotion, signaling activity, and downstream transcriptional responses. Findings suggest that expressing Stat92E-RNAi causes enhanced ethanol-induced hyperactivity in flies previously exposed to ethanol. Furthermore, alternative splicing of Stat92E itself was detected after repeated ethanol exposure, although no changes were found in downstream transcriptional activity. This work adds to our growing understanding of altered neuromolecular signaling following ethanol exposure and suggests that STAT signaling may be a relevant target to consider for AUD treatment.
Collapse
Affiliation(s)
| | | | | | - Emily Petruccelli
- Emily Petruccelli, Southern Illinois University Edwardsville College of Arts and Sciences, 44 Circle Dr, Edwardsville, IL 62026, USA.
| |
Collapse
|
10
|
Rose JK, Butterfield M, Liang J, Parvand M, Lin CHS, Rankin CH. Neuroligin Plays a Role in Ethanol-Induced Disruption of Memory and Corresponding Modulation of Glutamate Receptor Expression. Front Behav Neurosci 2022; 16:908630. [PMID: 35722190 PMCID: PMC9204643 DOI: 10.3389/fnbeh.2022.908630] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 05/04/2022] [Indexed: 11/13/2022] Open
Abstract
Exposure to alcohol causes deficits in long-term memory formation across species. Using a long-term habituation memory assay in Caenorhabditis elegans, the effects of ethanol on long-term memory (> 24 h) for habituation were investigated. An impairment in long-term memory was observed when animals were trained in the presence of ethanol. Cues of internal state or training context during testing did not restore memory. Ethanol exposure during training also interfered with the downregulation of AMPA/KA-type glutamate receptor subunit (GLR-1) punctal expression previously associated with long-term memory for habituation in C. elegans. Interestingly, ethanol exposure alone had the opposite effect, increasing GLR-1::GFP punctal expression. Worms with a mutation in the C. elegans ortholog of vertebrate neuroligins (nlg-1) were resistant to the effects of ethanol on memory, as they displayed both GLR-1::GFP downregulation and long-term memory for habituation after training in the presence of ethanol. These findings provide insights into the molecular mechanisms through which alcohol consumption impacts memory.
Collapse
|
11
|
Notch signaling pathway: architecture, disease, and therapeutics. Signal Transduct Target Ther 2022; 7:95. [PMID: 35332121 PMCID: PMC8948217 DOI: 10.1038/s41392-022-00934-y] [Citation(s) in RCA: 508] [Impact Index Per Article: 169.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 02/16/2022] [Accepted: 02/16/2022] [Indexed: 02/07/2023] Open
Abstract
The NOTCH gene was identified approximately 110 years ago. Classical studies have revealed that NOTCH signaling is an evolutionarily conserved pathway. NOTCH receptors undergo three cleavages and translocate into the nucleus to regulate the transcription of target genes. NOTCH signaling deeply participates in the development and homeostasis of multiple tissues and organs, the aberration of which results in cancerous and noncancerous diseases. However, recent studies indicate that the outcomes of NOTCH signaling are changeable and highly dependent on context. In terms of cancers, NOTCH signaling can both promote and inhibit tumor development in various types of cancer. The overall performance of NOTCH-targeted therapies in clinical trials has failed to meet expectations. Additionally, NOTCH mutation has been proposed as a predictive biomarker for immune checkpoint blockade therapy in many cancers. Collectively, the NOTCH pathway needs to be integrally assessed with new perspectives to inspire discoveries and applications. In this review, we focus on both classical and the latest findings related to NOTCH signaling to illustrate the history, architecture, regulatory mechanisms, contributions to physiological development, related diseases, and therapeutic applications of the NOTCH pathway. The contributions of NOTCH signaling to the tumor immune microenvironment and cancer immunotherapy are also highlighted. We hope this review will help not only beginners but also experts to systematically and thoroughly understand the NOTCH signaling pathway.
Collapse
|
12
|
Lacoste J, Soula H, Burg A, Audibert A, Darnat P, Gho M, Louvet-Vallée S. A neural progenitor mitotic wave is required for asynchronous axon outgrowth and morphology. eLife 2022; 11:75746. [PMID: 35254258 PMCID: PMC8933001 DOI: 10.7554/elife.75746] [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: 11/22/2021] [Accepted: 03/06/2022] [Indexed: 11/16/2022] Open
Abstract
Spatiotemporal mechanisms generating neural diversity are fundamental for understanding neural processes. Here, we investigated how neural diversity arises from neurons coming from identical progenitors. In the dorsal thorax of Drosophila, rows of mechanosensory organs originate from the division of sensory organ progenitor (SOPs). We show that in each row of the notum, an anteromedial located central SOP divides first, then neighbouring SOPs divide, and so on. This centrifugal wave of mitoses depends on cell-cell inhibitory interactions mediated by SOP cytoplasmic protrusions and Scabrous, a secreted protein interacting with the Delta/Notch complex. Furthermore, when this mitotic wave was reduced, axonal growth was more synchronous, axonal terminals had a complex branching pattern and fly behaviour was impaired. We show that the temporal order of progenitor divisions influences the birth order of sensory neurons, axon branching and impact on grooming behaviour. These data support the idea that developmental timing controls axon wiring neural diversity.
Collapse
Affiliation(s)
- Jérôme Lacoste
- UMR 7622 laboratory of Developmental Biology, CNRS Sorbonne-Université, Paris, France
| | - Hédi Soula
- NutriOmics Research Unit, Sorbonne Université, INSERM, Paris, France
| | - Angélique Burg
- UMR 7622 laboratory of Developmental Biology, CNRS Sorbonne-Université, Paris, France
| | - Agnès Audibert
- UMR 7622 laboratory of Developmental Biology, CNRS Sorbonne-Université, Paris, France
| | - Pénélope Darnat
- UMR 7622 laboratory of Developmental Biology, CNRS Sorbonne-Université, Paris, France
| | - Michel Gho
- UMR 7622 laboratory of Developmental Biology, CNRS Sorbonne-Université, Paris, France
| | - Sophie Louvet-Vallée
- UMR 7622 laboratory of Developmental Biology, CNRS Sorbonne-Université, Paris, France
| |
Collapse
|
13
|
Medial prefrontal cortex Notch1 signalling mediates methamphetamine-induced psychosis via Hes1-dependent suppression of GABA B1 receptor expression. Mol Psychiatry 2022; 27:4009-4022. [PMID: 35732696 PMCID: PMC9718672 DOI: 10.1038/s41380-022-01662-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 05/29/2022] [Accepted: 06/07/2022] [Indexed: 02/07/2023]
Abstract
Methamphetamine (METH), a widely abused stimulant drug, induces psychosis in approximately half of abusers; this effect is becoming a major concern for society. Although the Notch1 signalling pathway has been shown to play a part in the pathogenesis of some psychiatric disorders, its role in METH-induced psychosis (MIP) is still unknown. Here, the METH-induced locomotor sensitization model in rodents is considered to represent the underlying neurochemical changes driving psychoses. We found that the Notch1 signalling was downregulated in the medial prefrontal cortex (mPFC) in sensitized mice. Direct genetic and pharmacological manipulations of Notch1 signalling bidirectionally altered METH-induced locomotor sensitization and other MIP-related behaviours through governing neuronal activity in the mPFC. Moreover, Notch1 signalling negatively regulated GABAB1 receptor expression in the mPFC of METH-sensitized mice through Hes1, a transcriptional repressor in Notch1 signalling. Further, we show that Hes1 can directly bind to the GABAB1 receptor promoter. Notably, pharmacological regulation of the GABAB receptor in the mPFC reversed the changes in METH-induced locomotor sensitization caused by the dysfunction of Notch1 signalling. Together, our findings uncover a previously unrecognised Notch1-Hes1-GABAB1 receptor-dependent mechanism involved in regulating mPFC neuronal activity and behavioural phenotypes in MIP. Our work provides mechanistic insight into the aetiology and pathophysiology of MIP.
Collapse
|
14
|
Santalla M, Gómez IM, Valverde CA, Ferrero P. A low-cost Portable Device to Deliver Smoke, Volatile or Vaporized Substances to Drosophila melanogaster, Useful for Research and/or Educational Assays. Bio Protoc 2021; 11:e4244. [PMID: 35005089 PMCID: PMC8678554 DOI: 10.21769/bioprotoc.4244] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 09/01/2021] [Accepted: 09/06/2021] [Indexed: 11/25/2023] Open
Abstract
Drosophila melanogaster has been used to test drugs of abuse, substances with potential benefits for medical purposes, as well as contaminants and hazardous volatile compounds. This model has also been used for the characterization of behavioral changes, physiopathological consequences, and subcellular mechanisms of the use of cocaine, methamphetamines, ethanol, nicotine, cannabinoids, toluene, and other airborne volatile organic compounds. When testing these substances, routes of administration are important to define. Admixing the test compounds with water or food is one suitable option in many cases, but the inhalation route is especially suitable when the administration of one or more volatile compounds is desired. One advantage of the administration of substances via the inhalation route is its rapid exchange and distribution throughout the cuticle and the tracheal system. In addition, this route allows treating a large group of individuals simultaneously. Moreover, the inhalation route is frequently used to administer different drugs to humans. A good model system shares physiology and molecular pathways with humans, and D. melanogaster possesses almost 75% homologous genes associated with human diseases. Methodologies to deliver the abovementioned substances usually include customized devices. Herein, we focus on the development of a low-cost customized device useful to deliver smoke or vaporizable compounds to D. melanogaster. This approach might be applied for acute or chronic exposure to vaporized substances. In particular, our device was utilized for testing cigarette smoke and vaporized cannabis extract on cardiac performance of adult individuals during chronic treatment. We are describing how to set up this low-cost portable device, useful for research and/or educational assays, taking advantage of the amenability of D. melanogaster to test different compounds in relatively short periods, and especially including a large number of individuals at the same time. Graphic abstract: Custom-made device useful for inhalation pathway assays in Drosophila melanogaster.
Collapse
Affiliation(s)
- Manuela Santalla
- Departamento de Ciencias Básicas y Experimentales, Universidad Nacional del Noroeste (UNNOBA), Pergamino, Argentina
- Centro de Investigaciones Cardiovasculares “Dr. Horacio E. Cingolani”, Facultad de Ciencias Médicas-UNLP CONICET, La Plata, Argentina
| | - Ivana M. Gómez
- Centro de Investigaciones Cardiovasculares “Dr. Horacio E. Cingolani”, Facultad de Ciencias Médicas-UNLP CONICET, La Plata, Argentina
| | - Carlos A. Valverde
- Centro de Investigaciones Cardiovasculares “Dr. Horacio E. Cingolani”, Facultad de Ciencias Médicas-UNLP CONICET, La Plata, Argentina
| | - Paola Ferrero
- Departamento de Ciencias Básicas y Experimentales, Universidad Nacional del Noroeste (UNNOBA), Pergamino, Argentina
- Centro de Investigaciones Cardiovasculares “Dr. Horacio E. Cingolani”, Facultad de Ciencias Médicas-UNLP CONICET, La Plata, Argentina
| |
Collapse
|
15
|
Egervari G, Siciliano CA, Whiteley EL, Ron D. Alcohol and the brain: from genes to circuits. Trends Neurosci 2021; 44:1004-1015. [PMID: 34702580 PMCID: PMC8616825 DOI: 10.1016/j.tins.2021.09.006] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 09/08/2021] [Accepted: 09/30/2021] [Indexed: 01/27/2023]
Abstract
Alcohol use produces wide-ranging and diverse effects on the central nervous system. It influences intracellular signaling mechanisms, leading to changes in gene expression, chromatin remodeling, and translation. As a result of these molecular alterations, alcohol affects the activity of neuronal circuits. Together, these mechanisms produce long-lasting cellular adaptations in the brain that in turn can drive the development and maintenance of alcohol use disorder (AUD). We provide an update on alcohol research, focusing on multiple levels of alcohol-induced adaptations, from intracellular changes to changes in neural circuits. A better understanding of how alcohol affects these diverse and interlinked mechanisms may lead to the identification of novel therapeutic targets and to the development of much-needed novel and efficacious treatment options.
Collapse
Affiliation(s)
- Gabor Egervari
- Department of Cell and Developmental Biology, Epigenetics Institute, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Cody A Siciliano
- Department of Pharmacology, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN 37203, USA.
| | - Ellanor L Whiteley
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Dorit Ron
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
16
|
Atkinson NS. Alcohol-induced Aggression. Neurosci Insights 2021; 16:26331055211061145. [PMID: 34841248 PMCID: PMC8611288 DOI: 10.1177/26331055211061145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 11/02/2021] [Indexed: 11/16/2022] Open
Abstract
Intraspecies aggression is commonly focused on securing reproductive resources such as food, territory, and mates, and it is often males who do the fighting. In humans, individual acts of overt physical aggression seem maladaptive and probably represent dysregulation of the pathways underlying aggression. Such acts are often associated with ethanol consumption. The Drosophila melanogaster model system, which has long been used to study how ethanol affects the nervous system and behavior, has also been used to study the molecular origins of aggression. In addition, ethanol-induced aggression has been demonstrated in flies. Recent publications show that ethanol stimulates Drosophila aggression in 2 ways: the odor of ethanol and the consumption of ethanol both make males more aggressive. These ethanol effects occur at concentrations that flies likely experience in the wild. A picture emerges of males arriving on their preferred reproductive site-fermenting plant matter-and being stimulated by ethanol to fight harder to secure the site for their own use. Fly fighting assays appear to be a suitable bioassay for studying how low doses of ethanol reshape neural signaling.
Collapse
Affiliation(s)
- Nigel S Atkinson
- Department of Neuroscience and The Waggoner
Center for Alcohol and Addiction Research, The University of Texas at
Austin, Austin, TX, USA
| |
Collapse
|
17
|
Fathi D, Abulsoud AI, Saad MA, Nassar NN, Maksimos MM, Rizk SM, Senousy MA. Agomelatine attenuates alcohol craving and withdrawal symptoms by modulating the Notch1 signaling pathway in rats. Life Sci 2021; 284:119904. [PMID: 34453945 DOI: 10.1016/j.lfs.2021.119904] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 08/11/2021] [Accepted: 08/12/2021] [Indexed: 12/14/2022]
Abstract
AIM Alcohol abuse is a significant causative factor of death worldwide. The Notch1 signaling pathway is involved in alcohol tolerance, withdrawal and dependence. Agomelatine is a known antidepressant acting as a melatonin receptor (MT1/2) agonist and a 5-hydroxytryptamine receptor-2C antagonist. However, its effects on alcohol cravings and alcohol withdrawal symptoms have not been investigated. In this study, we assessed the possibility of using agomelatine for the treatment of these symptoms in a rat model of alcoholism and the possible role of Notch1 signaling. MAIN METHODS We induced alcoholism in rats using a free-choice drinking model for 60 days. From day 61, free-choice was continued until day 82 for the craving model, whereas only water was offered in the withdrawal model. Meanwhile, the treated groups for both models received agomelatine (50 mg/kg/day) orally from day 61 to 82, followed by behavioral, histopathological and biochemical assessment. KEY FINDINGS Agomelatine treatment caused significant decrease in alcohol consumption with a positive effect on anxiety-like behavior in the open field, memory in the Morris water maze and immobility in the forced swim test. Moreover, agomelatine induced the expression of Notch1 pathway markers, including Notch1, NICD, CREB, CCNE-2, Hes-1, both total and phosphorylated ERK1/2, MMP9, Per2and RGS-2 in the hippocampal formation. By contrast, NMDAR expression was reduced. Furthermore, agomelatine normalized the serum levels of BDNF, cortisol, dopamine and glutamate which were disrupted by alcohol consumption. SIGNIFICANCE Based on these findings, agomelatine reversed alcohol cravings and withdrawal symptoms associated with alcohol dependence by modulating the Notch1 signaling pathway.
Collapse
Affiliation(s)
- Doaa Fathi
- Department of Biochemistry, Faculty of Pharmacy, Heliopolis University, Cairo, Egypt
| | - Ahmed I Abulsoud
- Department of Biochemistry, Faculty of Pharmacy, Heliopolis University, Cairo, Egypt; Department of Biochemistry, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Muhammed A Saad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt; School of Pharmacy, Newgiza University, Cairo, Egypt
| | - Noha N Nassar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Mina M Maksimos
- Department of Microbiology and Immunology, Faculty of Pharmacy, Heliopolis University, Cairo, Egypt; Institute for Microbiology, Faculty of Life Sciences, Friedrich Schiller University of Jena, Jena, Germany
| | - Sherine M Rizk
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| | - Mahmoud A Senousy
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
18
|
Oepen AS, Catalano JL, Azanchi R, Kaun KR. The foraging gene affects alcohol sensitivity, metabolism and memory in Drosophila. J Neurogenet 2021; 35:236-248. [PMID: 34092172 PMCID: PMC9215342 DOI: 10.1080/01677063.2021.1931178] [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: 11/02/2020] [Accepted: 05/13/2021] [Indexed: 10/21/2022]
Abstract
The genetic basis of alcohol use disorder (AUD) is complex. Understanding how natural genetic variation contributes to alcohol phenotypes can help us identify and understand the genetic basis of AUD. Recently, a single nucleotide polymorphism in the human foraging (for) gene ortholog, Protein Kinase cGMP-Dependent 1 (PRKG1), was found to be associated with stress-induced risk for alcohol abuse. However, the mechanistic role that PRKG1 plays in AUD is not well understood. We use natural variation in the Drosophila for gene to describe how variation of cGMP-dependent protein kinase (PKG) activity modifies ethanol-induced phenotypes. We found that variation in for affects ethanol-induced increases in locomotion and memory of the appetitive properties of ethanol intoxication. Further, these differences may stem from the ability to metabolize ethanol. Together, this data suggests that natural variation in PKG modulates cue reactivity for alcohol, and thus could influence alcohol cravings by differentially modulating metabolic and behavioral sensitivities to alcohol.
Collapse
Affiliation(s)
- Anne S. Oepen
- Department of Neuroscience, Brown University, Providence,
RI, USA
- Masters Program in Developmental, Neuronal and Behavioral
Biology, Georg-August-University, Göttingen, Germany
| | - Jamie L. Catalano
- Department of Neuroscience, Brown University, Providence,
RI, USA
- Molecular Pharmacology and Physiology Graduate Program,
Brown University, Providence, RI, USA
| | - Reza Azanchi
- Department of Neuroscience, Brown University, Providence,
RI, USA
| | - Karla R. Kaun
- Department of Neuroscience, Brown University, Providence,
RI, USA
| |
Collapse
|
19
|
Park A, Tran T, Gutierrez L, Stojanik CJ, Plyler J, Thompson GA, Bohm RA, Scheuerman EA, Smith DP, Atkinson NS. Alcohol-induced aggression in Drosophila. Addict Biol 2021; 26:e13045. [PMID: 34044470 DOI: 10.1111/adb.13045] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 03/16/2021] [Accepted: 03/31/2021] [Indexed: 11/28/2022]
Abstract
Alcohol-induced aggression is a destructive and widespread phenomenon associated with violence and sexual assault. However, little is understood concerning its mechanistic origin. We have developed a Drosophila melanogaster model to genetically dissect and understand the phenomenon of sexually dimorphic alcohol-induced aggression. Males with blood alcohol levels of 0.04-mg/ml BAC were less aggressive than alcohol-naive males, but when the BAC had dropped to ~0.015 mg/ml, the alcohol-treated males showed an increase in aggression toward other males. This aggression-promoting treatment is referred to as the post-ethanol aggression (PEA) treatment. Females do not show increased aggression after the same treatment. PEA-treated males also spend less time courting and attempt to copulate earlier than alcohol-naive flies. PEA treatment induces expression of the FruM transcription factor (encoded by a male-specific transcript from the fruitless gene), whereas sedating doses of alcohol reduce FruM expression and reduce male aggression. Transgenic suppression of FruM induction also prevents alcohol-induced aggression. In male flies, alcohol-induced aggression is dependent on the male isoform of the fruitless transcription factor (FruM). Low-dose alcohol induces FruM expression and promotes aggression, whereas higher doses of alcohol suppress FruM and suppress aggression.
Collapse
Affiliation(s)
- Annie Park
- Department of Neuroscience and Waggoner Center for Alcohol and Addiction Research The University of Texas at Austin Austin Texas USA
- Centre for Neural Circuits and Behaviour The University of Oxford Oxford UK
| | - Tracy Tran
- Department of Neuroscience and Waggoner Center for Alcohol and Addiction Research The University of Texas at Austin Austin Texas USA
| | - Linda Gutierrez
- Department of Neuroscience and Waggoner Center for Alcohol and Addiction Research The University of Texas at Austin Austin Texas USA
| | - Christopher J. Stojanik
- Department of Neuroscience and Waggoner Center for Alcohol and Addiction Research The University of Texas at Austin Austin Texas USA
| | - Julian Plyler
- Department of Neuroscience and Waggoner Center for Alcohol and Addiction Research The University of Texas at Austin Austin Texas USA
| | - Grace A. Thompson
- Department of Neuroscience and Waggoner Center for Alcohol and Addiction Research The University of Texas at Austin Austin Texas USA
| | - Rudolf A. Bohm
- Department of Biological and Health Sciences Texas A&M University‐Kingsville Kingsville Texas USA
| | - Elizabeth A. Scheuerman
- Department of Pharmacology and Neuroscience University of Texas Southwestern Medical Center Dallas Texas USA
| | - Dean P. Smith
- Department of Pharmacology and Neuroscience University of Texas Southwestern Medical Center Dallas Texas USA
| | - Nigel S. Atkinson
- Department of Neuroscience and Waggoner Center for Alcohol and Addiction Research The University of Texas at Austin Austin Texas USA
| |
Collapse
|
20
|
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
|
21
|
Fang X, Deza-Araujo YI, Petzold J, Spreer M, Riedel P, Marxen M, O'Connor SJ, Zimmermann US, Smolka MN. Effects of moderate alcohol levels on default mode network connectivity in heavy drinkers. Alcohol Clin Exp Res 2021; 45:1039-1050. [PMID: 33742481 DOI: 10.1111/acer.14602] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 03/08/2021] [Accepted: 03/10/2021] [Indexed: 11/28/2022]
Abstract
BACKGROUND It is well established that even moderate levels of alcohol affect cognitive functions such as memory, self-related information processing, and response inhibition. Nevertheless, the neural mechanisms underlying these alcohol-induced changes are still unclear, especially on the network level. The default mode network (DMN) plays an important role in memory and self-initiated mental activities; hence, studying functional interactions of the DMN may provide new insights into the neural mechanisms underlying alcohol-related changes. METHODS We investigated resting-state functional connectivity (rsFC) of the DMN in a cohort of 37 heavy drinkers at a breath alcohol concentration of 0.8 g/kg. Alcohol and saline were infused in a single-blind crossover design. RESULTS Intranetwork connectivity analyses revealed that participants showed significantly decreased rsFC of the right hippocampus and right middle temporal gyrus during acute alcohol exposure. Moreover, follow-up analyses revealed that these rsFC decreases were more pronounced in participants who reported stronger craving for alcohol. Exploratory internetwork connectivity analyses of the DMN with other resting-state networks showed no significant alcohol-induced changes, but suffered from low statistical power. CONCLUSIONS Our results indicate that acute alcohol exposure affects rsFC within the DMN. Functionally, this finding may be associated with impairments in memory encoding and self-referential processes commonly observed during alcohol intoxication. Future resting-state functional magnetic resonance imaging studies might therefore also investigate memory function and test whether DMN-related connectivity changes are associated with alcohol-induced impairments or craving.
Collapse
Affiliation(s)
- Xiaojing Fang
- Department of Psychiatry and Neuroimaging Center, Technische Universität Dresden, Dresden, Germany
| | - Yacila I Deza-Araujo
- Department of Psychiatry and Neuroimaging Center, Technische Universität Dresden, Dresden, Germany
| | - Johannes Petzold
- Department of Psychiatry and Neuroimaging Center, Technische Universität Dresden, Dresden, Germany
| | - Maik Spreer
- Department of Psychiatry and Neuroimaging Center, Technische Universität Dresden, Dresden, Germany
| | - Philipp Riedel
- Department of Psychiatry and Neuroimaging Center, Technische Universität Dresden, Dresden, Germany
| | - Michael Marxen
- Department of Psychiatry and Neuroimaging Center, Technische Universität Dresden, Dresden, Germany
| | - Sean J O'Connor
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Ulrich S Zimmermann
- Department of Psychiatry and Neuroimaging Center, Technische Universität Dresden, Dresden, Germany.,Department of Addiction Medicine and Psychotherapy, Isar-Amper-Klinikum München-Ost, Haar, Germany
| | - Michael N Smolka
- Department of Psychiatry and Neuroimaging Center, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
22
|
Kanno M, Hiramatsu S, Kondo S, Tanimoto H, Ichinose T. Voluntary intake of psychoactive substances is regulated by the dopamine receptor Dop1R1 in Drosophila. Sci Rep 2021; 11:3432. [PMID: 33564023 PMCID: PMC7873259 DOI: 10.1038/s41598-021-82813-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 01/21/2021] [Indexed: 11/09/2022] Open
Abstract
Dysregulated motivation to consume psychoactive substances leads to addictive behaviors that often result in serious health consequences. Understanding the neuronal mechanisms that drive drug consumption is crucial for developing new therapeutic strategies. The fruit fly Drosophila melanogaster offers a unique opportunity to approach this problem with a battery of sophisticated neurogenetic tools available, but how they consume these drugs remains largely unknown. Here, we examined drug self-administration behavior of Drosophila and the underlying neuronal mechanisms. We measured the preference of flies for five different psychoactive substances using a two-choice feeding assay and monitored its long-term changes. We found that flies show acute preference for ethanol and methamphetamine, but not for cocaine, caffeine or morphine. Repeated intake of ethanol, but not methamphetamine, increased over time. Preference for methamphetamine and the long-term escalation of ethanol preference required the dopamine receptor Dop1R1 in the mushroom body. The protein level of Dop1R1 increased after repeated intake of ethanol, but not methamphetamine, which correlates with the acquired preference. Genetic overexpression of Dop1R1 enhanced ethanol preference. These results reveal a striking diversity of response to individual drugs in the fly and the role of dopamine signaling and its plastic changes in controlling voluntary intake of drugs.
Collapse
Affiliation(s)
- Mai Kanno
- Graduate School of Life Sciences, Tohoku University, Sendai, 980-8577, Japan
| | - Shun Hiramatsu
- Graduate School of Life Sciences, Tohoku University, Sendai, 980-8577, Japan
| | - Shu Kondo
- Invertebrate Genetics Laboratory, National Institute of Genetics, Mishima, 411-8540, Japan
| | - Hiromu Tanimoto
- Graduate School of Life Sciences, Tohoku University, Sendai, 980-8577, Japan
| | - Toshiharu Ichinose
- Graduate School of Life Sciences, Tohoku University, Sendai, 980-8577, Japan. .,Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Sendai, 980-8578, Japan. .,Center for Transdisciplinary Research, Niigata University, Niigata, 950-2181, Japan. .,Department of Neuropharmacology, Nagoya City University, Nagoya, 467-8603, Japan.
| |
Collapse
|
23
|
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
|
24
|
Salazar JL, Yang SA, Yamamoto S. Post-Developmental Roles of Notch Signaling in the Nervous System. Biomolecules 2020; 10:biom10070985. [PMID: 32630239 PMCID: PMC7408554 DOI: 10.3390/biom10070985] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 06/25/2020] [Accepted: 06/26/2020] [Indexed: 12/14/2022] Open
Abstract
Since its discovery in Drosophila, the Notch signaling pathway has been studied in numerous developmental contexts in diverse multicellular organisms. The role of Notch signaling in nervous system development has been extensively investigated by numerous scientists, partially because many of the core Notch signaling components were initially identified through their dramatic ‘neurogenic’ phenotype of developing fruit fly embryos. Components of the Notch signaling pathway continue to be expressed in mature neurons and glia cells, which is suggestive of a role in the post-developmental nervous system. The Notch pathway has been, so far, implicated in learning and memory, social behavior, addiction, and other complex behaviors using genetic model organisms including Drosophila and mice. Additionally, Notch signaling has been shown to play a modulatory role in several neurodegenerative disease model animals and in mediating neural toxicity of several environmental factors. In this paper, we summarize the knowledge pertaining to the post-developmental roles of Notch signaling in the nervous system with a focus on discoveries made using the fruit fly as a model system as well as relevant studies in C elegans, mouse, rat, and cellular models. Since components of this pathway have been implicated in the pathogenesis of numerous psychiatric and neurodegenerative disorders in human, understanding the role of Notch signaling in the mature brain using model organisms will likely provide novel insights into the mechanisms underlying these diseases.
Collapse
Affiliation(s)
- Jose L. Salazar
- Department of Molecular and Human Genetics, Baylor College of Medicine (BCM), Houston, TX 77030, USA; (J.L.S.); (S.-A.Y.)
| | - Sheng-An Yang
- Department of Molecular and Human Genetics, Baylor College of Medicine (BCM), Houston, TX 77030, USA; (J.L.S.); (S.-A.Y.)
| | - Shinya Yamamoto
- Department of Molecular and Human Genetics, Baylor College of Medicine (BCM), Houston, TX 77030, USA; (J.L.S.); (S.-A.Y.)
- Department of Neuroscience, BCM, Houston, TX 77030, USA
- Program in Developmental Biology, BCM, Houston, TX 77030, USA
- Development, Disease Models & Therapeutics Graduate Program, BCM, Houston, TX 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX 77030, USA
- Correspondence: ; Tel.: +1-832-824-8119
| |
Collapse
|
25
|
Scaplen KM, Talay M, Nunez KM, Salamon S, Waterman AG, Gang S, Song SL, Barnea G, Kaun KR. Circuits that encode and guide alcohol-associated preference. eLife 2020; 9:48730. [PMID: 32497004 PMCID: PMC7272191 DOI: 10.7554/elife.48730] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 05/18/2020] [Indexed: 12/21/2022] Open
Abstract
A powerful feature of adaptive memory is its inherent flexibility. Alcohol and other addictive substances can remold neural circuits important for memory to reduce this flexibility. However, the mechanism through which pertinent circuits are selected and shaped remains unclear. We show that circuits required for alcohol-associated preference shift from population level dopaminergic activation to select dopamine neurons that predict behavioral choice in Drosophila melanogaster. During memory expression, subsets of dopamine neurons directly and indirectly modulate the activity of interconnected glutamatergic and cholinergic mushroom body output neurons (MBON). Transsynaptic tracing of neurons important for memory expression revealed a convergent center of memory consolidation within the mushroom body (MB) implicated in arousal, and a structure outside the MB implicated in integration of naïve and learned responses. These findings provide a circuit framework through which dopamine neuronal activation shifts from reward delivery to cue onset, and provide insight into the maladaptive nature of memory.
Collapse
Affiliation(s)
- Kristin M Scaplen
- Department of Neuroscience, Brown University, Providence, United States
| | - Mustafa Talay
- Department of Neuroscience, Brown University, Providence, United States
| | - Kavin M Nunez
- Department of Molecular Pharmacology and Physiology, Brown University, Providence, United States
| | - Sarah Salamon
- Department of Pharmacology, University of Cologne, Cologne, Germany
| | - Amanda G Waterman
- Department of Neuroscience, Brown University, Providence, United States
| | - Sydney Gang
- Department of Biochemistry, Brown University, Providence, United States
| | - Sophia L Song
- Department of Neuroscience, Brown University, Providence, United States
| | - Gilad Barnea
- Department of Neuroscience, Brown University, Providence, United States
| | - Karla R Kaun
- Department of Neuroscience, Brown University, Providence, United States
| |
Collapse
|
26
|
Petruccelli E, Brown T, Waterman A, Ledru N, Kaun KR. Alcohol Causes Lasting Differential Transcription in Drosophila Mushroom Body Neurons. Genetics 2020; 215:103-116. [PMID: 32132098 PMCID: PMC7198272 DOI: 10.1534/genetics.120.303101] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 02/26/2020] [Indexed: 12/12/2022] Open
Abstract
Repeated alcohol experiences can produce long-lasting memories for sensory cues associated with intoxication. These memories can problematically trigger relapse in individuals recovering from alcohol use disorder (AUD). The molecular mechanisms by which ethanol changes memories to become long-lasting and inflexible remain unclear. New methods to analyze gene expression within precise neuronal cell types can provide further insight toward AUD prevention and treatment. Here, we used genetic tools in Drosophila melanogaster to investigate the lasting consequences of ethanol on transcription in memory-encoding neurons. Drosophila rely on mushroom body (MB) neurons to make associative memories, including memories of ethanol-associated sensory cues. Differential expression analyses revealed that distinct transcripts, but not genes, in the MB were associated with experiencing ethanol alone compared to forming a memory of an odor cue associated with ethanol. Adult MB-specific knockdown of spliceosome-associated proteins demonstrated the necessity of RNA-processing in ethanol memory formation. These findings highlight the dynamic, context-specific regulation of transcription in cue-encoding neurons, and the lasting effect of ethanol on transcript usage during memory formation.
Collapse
Affiliation(s)
- Emily Petruccelli
- Department of Neuroscience, Brown University, Providence, Rhode Island 02912
- Department of Biological Sciences, Southern Illinois University Edwardsville, Illinois 62026
| | - Tariq Brown
- Department of Neuroscience, Brown University, Providence, Rhode Island 02912
| | - Amanda Waterman
- Department of Neuroscience, Brown University, Providence, Rhode Island 02912
| | - Nicolas Ledru
- Department of Neuroscience, Brown University, Providence, Rhode Island 02912
| | - Karla R Kaun
- Department of Neuroscience, Brown University, Providence, Rhode Island 02912
| |
Collapse
|
27
|
Ho DM, Artavanis-Tsakonas S, Louvi A. The Notch pathway in CNS homeostasis and neurodegeneration. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2019; 9:e358. [PMID: 31502763 DOI: 10.1002/wdev.358] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 06/19/2019] [Accepted: 06/23/2019] [Indexed: 12/19/2022]
Abstract
The role of the Notch signaling pathway in neural development has been well established over many years. More recent studies, however, have demonstrated that Notch continues to be expressed and active throughout adulthood in many areas of the central nervous system. Notch signals have been implicated in adult neurogenesis, memory formation, and synaptic plasticity in the adult organism, as well as linked to acute brain trauma and chronic neurodegenerative conditions. NOTCH3 mutations are responsible for the most common form of hereditary stroke, the progressive disorder cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy. Notch has also been associated with several progressive neurodegenerative diseases, including Alzheimer's disease, multiple sclerosis, and amyotrophic lateral sclerosis. Although numerous studies link Notch activity with CNS homeostasis and neurodegenerative diseases, the data thus far are primarily correlative, rather than functional. Nevertheless, the evidence for Notch pathway activity in specific neural cellular contexts is strong, and certainly intriguing, and points to the possibility that the pathway carries therapeutic promise. This article is categorized under: Nervous System Development > Flies Signaling Pathways > Cell Fate Signaling Nervous System Development > Vertebrates: General Principles.
Collapse
Affiliation(s)
- Diana M Ho
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts
| | | | - Angeliki Louvi
- Departments of Neurosurgery and Neuroscience and Program on Neurogenetics, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
28
|
Karam CS, Jones SK, Javitch JA. Come Fly with Me: An overview of dopamine receptors in Drosophila melanogaster. Basic Clin Pharmacol Toxicol 2019; 126 Suppl 6:56-65. [PMID: 31219669 DOI: 10.1111/bcpt.13277] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 06/17/2019] [Indexed: 12/23/2022]
Abstract
Dopamine (DA) receptors play critical roles in a wide range of behaviours, including sensory processing, motor function, reward and arousal. As such, aberrant DA signalling is associated with numerous neurological and psychiatric disorders. Therefore, understanding the mechanisms by which DA neurotransmission drives intracellular signalling pathways that modulate behaviour can provide critical insights to guide the development of targeted therapeutics. Drosophila melanogaster has emerged as a powerful model with unique advantages to study the mechanisms underlying DA neurotransmission and associated behaviours in a controlled and systematic manner. Many regions in the fly brain innervated by dopaminergic neurons have been mapped and linked to specific behaviours, including associative learning and arousal. Here, we provide an overview of the homology between human and Drosophila dopaminergic systems and review the current literature on the pharmacology, molecular signalling mechanisms and behavioural outcome of DA receptor activation in the Drosophila brain.
Collapse
Affiliation(s)
- Caline S Karam
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York City, New York, USA.,Division of Molecular Therapeutics, New York State Psychiatric Institute, New York City, New York, USA
| | - Sandra K Jones
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York City, New York, USA.,Division of Molecular Therapeutics, New York State Psychiatric Institute, New York City, New York, USA
| | - Jonathan A Javitch
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York City, New York, USA.,Division of Molecular Therapeutics, New York State Psychiatric Institute, New York City, New York, USA.,Department of Pharmacology, Columbia University Vagelos College of Physicians and Surgeons, New York City, New York, USA
| |
Collapse
|
29
|
Engel GL, Taber K, Vinton E, Crocker AJ. Studying alcohol use disorder using Drosophila melanogaster in the era of 'Big Data'. BEHAVIORAL AND BRAIN FUNCTIONS : BBF 2019; 15:7. [PMID: 30992041 PMCID: PMC6469124 DOI: 10.1186/s12993-019-0159-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 04/04/2019] [Indexed: 02/08/2023]
Abstract
Our understanding of the networks of genes and protein functions involved in Alcohol Use Disorder (AUD) remains incomplete, as do the mechanisms by which these networks lead to AUD phenotypes. The fruit fly (Drosophila melanogaster) is an efficient model for functional and mechanistic characterization of the genes involved in alcohol behavior. The fly offers many advantages as a model organism for investigating the molecular and cellular mechanisms of alcohol-related behaviors, and for understanding the underlying neural circuitry driving behaviors, such as locomotor stimulation, sedation, tolerance, and appetitive (reward) learning and memory. Fly researchers are able to use an extensive variety of tools for functional characterization of gene products. To understand how the fly can guide our understanding of AUD in the era of Big Data we will explore these tools, and review some of the gene networks identified in the fly through their use, including chromatin-remodeling, glial, cellular stress, and innate immunity genes. These networks hold great potential as translational drug targets, making it prudent to conduct further research into how these gene mechanisms are involved in alcohol behavior.
Collapse
Affiliation(s)
- Gregory L. Engel
- Department of Psychological Sciences, Castleton University, Castleton, VT 05735 USA
| | - Kreager Taber
- Program in Neuroscience, Middlebury College, Middlebury, VT 05753 USA
| | - Elizabeth Vinton
- Program in Neuroscience, Middlebury College, Middlebury, VT 05753 USA
| | - Amanda J. Crocker
- Program in Neuroscience, Middlebury College, Middlebury, VT 05753 USA
| |
Collapse
|