1
|
Sex-Related Differences in Voluntary Alcohol Intake and mRNA Coding for Synucleins in the Brain of Adult Rats Prenatally Exposed to Alcohol. Biomedicines 2022; 10:biomedicines10092163. [PMID: 36140264 PMCID: PMC9496239 DOI: 10.3390/biomedicines10092163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 08/17/2022] [Accepted: 08/29/2022] [Indexed: 11/24/2022] Open
Abstract
Maternal alcohol consumption is one of the strong predictive factors of alcohol use and consequent abuse; however, investigations of sex differences in response to prenatal alcohol exposure (PAE) are limited. Here we compared the effects of PAE throughout gestation on alcohol preference, state anxiety and mRNA expression of presynaptic proteins α-, β- and γ-synucleins in the brain of adult (PND60) male and female Wistar rats. Total RNA was isolated from the hippocampus, midbrain and hypothalamus and mRNA levels were assessed with quantitative RT-PCR. Compared with naïve males, naïve female rats consumed more alcohol in “free choice” paradigm (10% ethanol vs. water). At the same time, PAE produced significant increase in alcohol consumption and preference in males but not in females compared to male and female naïve groups, correspondingly. We found significantly lower α-synuclein mRNA levels in the hippocampus and midbrain of females compared to males and significant decrease in α-synuclein mRNA in these brain areas in PAE males, but not in females compared to the same sex controls. These findings indicate that the impact of PAE on transcriptional regulation of synucleins may be sex-dependent, and in males’ disruption in α-synuclein mRNA expression may contribute to increased vulnerability to alcohol-associated behavior.
Collapse
|
2
|
Duttke SH, Montilla-Perez P, Chang MW, Li H, Chen H, Carrette LLG, de Guglielmo G, George O, Palmer AA, Benner C, Telese F. Glucocorticoid Receptor-Regulated Enhancers Play a Central Role in the Gene Regulatory Networks Underlying Drug Addiction. Front Neurosci 2022; 16:858427. [PMID: 35651629 PMCID: PMC9149415 DOI: 10.3389/fnins.2022.858427] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 04/25/2022] [Indexed: 01/07/2023] Open
Abstract
Substance abuse and addiction represent a significant public health problem that impacts multiple dimensions of society, including healthcare, the economy, and the workforce. In 2021, over 100,000 drug overdose deaths were reported in the US, with an alarming increase in fatalities related to opioids and psychostimulants. Understanding the fundamental gene regulatory mechanisms underlying addiction and related behaviors could facilitate more effective treatments. To explore how repeated drug exposure alters gene regulatory networks in the brain, we combined capped small (cs)RNA-seq, which accurately captures nascent-like initiating transcripts from total RNA, with Hi-C and single nuclei (sn)ATAC-seq. We profiled initiating transcripts in two addiction-related brain regions, the prefrontal cortex (PFC) and the nucleus accumbens (NAc), from rats that were never exposed to drugs or were subjected to prolonged abstinence after oxycodone or cocaine intravenous self-administration (IVSA). Interrogating over 100,000 active transcription start regions (TSRs) revealed that most TSRs had hallmarks of bonafide enhancers and highlighted the KLF/SP1, RFX, and AP1 transcription factors families as central to establishing brain-specific gene regulatory programs. Analysis of rats with addiction-like behaviors versus controls identified addiction-associated repression of transcription at regulatory enhancers recognized by nuclear receptor subfamily 3 group C (NR3C) factors, including glucocorticoid receptors. Cell-type deconvolution analysis using snATAC-seq uncovered a potential role of glial cells in driving the gene regulatory programs associated with addiction-related phenotypes. These findings highlight the power of advanced transcriptomics methods to provide insight into how addiction perturbs gene regulatory programs in the brain.
Collapse
Affiliation(s)
- Sascha H. Duttke
- Department of Medicine, University of California, San Diego, La Jolla, CA, United States
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA, United States
| | | | - Max W. Chang
- Department of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Hairi Li
- Department of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Hao Chen
- Department of Pharmacology, Addiction Science and Toxicology, University of Tennessee Health Science Center, Memphis, TN, United States
| | | | - Giordano de Guglielmo
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, United States
| | - Olivier George
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, United States
| | - Abraham A. Palmer
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, United States
- Institute for Genomic Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Christopher Benner
- Department of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Francesca Telese
- Department of Medicine, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
3
|
Cahill CM, Aleyadeh R, Gao J, Wang C, Rogers JT. Alpha-Synuclein in Alcohol Use Disorder, Connections with Parkinson's Disease and Potential Therapeutic Role of 5' Untranslated Region-Directed Small Molecules. Biomolecules 2020; 10:E1465. [PMID: 33096655 PMCID: PMC7589448 DOI: 10.3390/biom10101465] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/28/2020] [Accepted: 10/09/2020] [Indexed: 12/23/2022] Open
Abstract
Alpha-synuclein (α-Syn) is a 140-amino acid (aa) protein encoded by the Synuclein alpha SNCA gene. It is the synaptic protein associated with Parkinson's disease (PD) and is the most highly expressed protein in the Lewy bodies associated with PD and other alpha synucleopathies, including Lewy body dementia (LBD) and multiple system atrophy (MSA). Iron deposits are present in the core of Lewy bodies, and there are reports suggesting that divalent metal ions including Cu2+ and Fe2+ enhance the aggregation of α-Syn. Differential expression of α-Syn is associated with alcohol use disorder (AUD), and specific genetic variants contribute to the risk for alcoholism, including alcohol craving. Spliced variants of α-Syn, leading to the expression of several shorter forms which are more prone to aggregation, are associated with both PD and AUD, and common transcript variants may be able to predict at-risk populations for some movement disorders or subtypes of PD, including secondary Parkinsonism. Both PD and AUD are associated with liver and brain iron dyshomeostasis. Research over the past decade has shown that α-Syn has iron import functions with an ability to oxidize the Fe3+ form of iron to Fe2+ to facilitate its entry into cells. Our prior research has identified an iron-responsive element (IRE) in the 5' untranslated region (5'UTR) of α-Syn mRNA, and we have used the α-Syn 5'UTR to screen for small molecules that modulate its expression in the H4 neuronal cell line. These screens have led us to identify several interesting small molecules capable of both decreasing and increasing α-Syn expression and that may have the potential, together with the recently described mesenchymal stem cell therapies, to normalize α-Syn expression in different regions of the alcoholic and PD brain.
Collapse
Affiliation(s)
- Catherine M. Cahill
- Neurochemistry Laboratory, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA;
| | | | - Jin Gao
- Department of Clinical Psychology, Qilu Hospital of Shandong University, Qingdao 266011, China;
| | - Changning Wang
- Athinoula A. Martinos Center for Biomedical Imaging Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA;
| | - Jack T. Rogers
- Neurochemistry Laboratory, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA;
| |
Collapse
|
4
|
Martínez-Rodríguez TY, Rey-Buitrago M. Physiological, molecular and genetic aspects of alpha-synuclein and its correlation with high alcohol consumption. REVISTA DE LA FACULTAD DE MEDICINA 2019. [DOI: 10.15446/revfacmed.v67n3.69962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Introduction: Significant changes in the expression of α-synuclein (SNCA) can be seen in subjects with high alcohol consumption, altering neuroprotection and causing changes in the reward system.Objective: To present state-of-the-art studies on the physiological, molecular and genetic aspects of SNCA related to high alcohol consumption.Materials and methods: A search of records published from 2007 to 2017 was carried out in PUBMED, ScienceDirect and Cochrane with the terms alpha-synuclein, alcoholism, genetic polymorphism, gene expression, DNA methylation and molecular biology.Results: The search yielded 1 331 references, of which 51 full-texts were selected. The results describe the current evidence of the physiological and pathological aspects of α-synuclein (SNCA) and the genetic and epigenetic changes related to its expression in people with high alcohol consumption.Conclusions: The evidence suggests that a differential expression of α-synuclein (SNCA) is found in subjects with high alcohol consumption, as a result of modifications in the genetic and epigenetic mechanisms, leading to physipathological neuroadaptations. SNCA is a promising marker in the field of alcoholism research; therefore, more studies are required in this regard, taking into account the genetic heterogeneity of each population.
Collapse
|
5
|
Spence JP, Reiter JL, Qiu B, Gu H, Garcia DK, Zhang L, Graves T, Williams KE, Bice PJ, Zou Y, Lai Z, Yong W, Liang T. Estrogen-Dependent Upregulation of Adcyap1r1 Expression in Nucleus Accumbens Is Associated With Genetic Predisposition of Sex-Specific QTL for Alcohol Consumption on Rat Chromosome 4. Front Genet 2018; 9:513. [PMID: 30564267 PMCID: PMC6288178 DOI: 10.3389/fgene.2018.00513] [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: 03/07/2018] [Accepted: 10/12/2018] [Indexed: 12/15/2022] Open
Abstract
Humans show sex differences related to alcohol use disorders (AUD). Animal model research has the potential to provide important insight into how sex differences affect alcohol consumption, particularly because female animals frequently drink more than males. In previous work, inbred strains of the selectively bred alcohol-preferring (P) and non-preferring (NP) rat lines revealed a highly significant quantitative trait locus (QTL) on rat chromosome 4, with a logarithm of the odds score of 9.2 for alcohol consumption. Recently, interval-specific congenic strains (ISCS) were developed by backcrossing the congenic P.NP line to inbred P (iP) rats to further refine the chromosome 4 QTL region. Two ISCS sub-strains, ISCS-A and ISCS-B, were obtained with a narrowed QTL, where the smallest region of overlap consisted of 8.9 Mb in ISCS-B. Interestingly, we found that females from both ISCS lines consumed significantly less alcohol than female iP controls (p < 0.05), while no differences in alcohol consumption were observed between male ISCS and iP controls. RNA-sequencing was performed on the nucleus accumbens of alcohol-naïve female ISCS-B and iP rats, which revealed differentially expressed genes (DEG) with greater than 2-fold change and that were functionally relevant to behavior. These DEGs included down-regulation of Oxt, Asb4, Gabre, Gabrq, Chat, Slc5a7, Slc18a8, Slc10a4, and Ngfr, and up-regulation of Ttr, Msln, Mpzl2, Wnt6, Slc17a7, Aldh1a2, and Gstm2. Pathway analysis identified significant alterations in gene networks controlling nervous system development and function, as well as cell signaling, GABA and serotonin receptor signaling and G-protein coupled receptor signaling. In addition, β-estradiol was identified as the most significant upstream regulator. The expression levels of estrogen-responsive genes that mapped to the QTL interval and have been previously associated with alcohol consumption were measured using RT-qPCR. We found that expression of the Adcyap1r1 gene, encoding the pituitary adenylate cyclase-activating polypeptide type 1 (PAC1) receptor, was upregulated in female ISCS-B compared to female iP controls, while no differences were exhibited in males. In addition, sequence variants in the Adcyap1r1 promoter region showed a differential response to estrogen stimulation in vitro. These findings demonstrate that rat chromosome 4 QTL contains genetic variants that respond to estrogen and are associated with female alcohol consumption.
Collapse
Affiliation(s)
- John Paul Spence
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Jill L Reiter
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Bin Qiu
- Comparative Medical Center, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Hao Gu
- Comparative Medical Center, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Dawn K Garcia
- Greehey Children's Cancer Research Institute, UT Health San Antonio, San Antonio, TX, United States
| | - Lingling Zhang
- Comparative Medical Center, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Tamara Graves
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Kent E Williams
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Paula J Bice
- Department of Psychology, Southeast Missouri State University, Cape Girardeau, MO, United States
| | - Yi Zou
- Greehey Children's Cancer Research Institute, UT Health San Antonio, San Antonio, TX, United States
| | - Zhao Lai
- Department of Psychology, Southeast Missouri State University, Cape Girardeau, MO, United States
| | - Weidong Yong
- Comparative Medical Center, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Tiebing Liang
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
6
|
Persyn W, Houchi H, Papillon CA, Martinetti M, Antol J, Guillaumont C, Dervaux A, Naassila M. Ethanol (EtOH)-Related Behaviors in α-Synuclein Mutant Mice and Association of SNCA SNPs with Anxiety in EtOH-Dependent Patients. Alcohol Clin Exp Res 2018; 42:2172-2185. [PMID: 30120834 DOI: 10.1111/acer.13875] [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: 03/31/2018] [Accepted: 08/10/2018] [Indexed: 12/25/2022]
Abstract
BACKGROUND Data have shown a role of α-synuclein in anxiety and also in addiction, particularly in alcohol use disorders (AUD). Since the comorbidity between AUD and anxiety is very high and because anxiety is an important factor in ethanol (EtOH) relapse, the aim of the present study was to investigate the role of α-synuclein in moderating EtOH intake, the anxiolytic effects of EtOH, and EtOH withdrawal-induced anxiety and convulsions in mice. The study aimed to determine whether SNCA variants moderated anxiety in EtOH-dependent patients. METHODS We analyzed the moderator effect of 3 SNCA Tag-single nucleotide polymorphisms (Tag-SNPs) rs356200, rs356219, and rs2119787 on the anxiety symptoms in 128 EtOH-dependent patients. We used the C57BL/6JOlaHsd Snca mutant mice to assess EtOH intake; sensitivity to the anxiolytic effects of EtOH in a test battery comprising the open field, the light-dark box, and the elevated plus maze; and both anxiety and convulsions induced by EtOH withdrawal. RESULTS Our results demonstrated a reduction in both EtOH intake and preference and also a lack of sensitivity to the anxiolytic effects of EtOH in α-synuclein mutant mice. Results on anxiety-like behavior were mixed, but mutant mice displayed increased anxiety when exposed to a low anxiogenic environment. Mutant mice also displayed an increase in handling-induced convulsion scores during withdrawal after EtOH inhalation, but did not differ in terms of EtOH withdrawal-induced anxiety. In humans, we found a significant association of the rs356219 SNP with a high level of anxiety (Beck Anxiety Inventory score >15) and the rs356200 SNP with a positive familial history of AUD. CONCLUSIONS Our translational study highlights a significant role of α-synuclein in components of AUD.
Collapse
Affiliation(s)
- Wolfgang Persyn
- Research Group on Alcohol & Pharmacodependences (GRAP), Centre Universitaire de Recherche en Santé (CURS) , INSERM U1247, University of Picardie Jules Verne, Amiens, France.,Unité d'alcoologie SESAME , Centre hospitalier Psychiatrique Philippe Pinel, Amiens, France
| | - Hakim Houchi
- Research Group on Alcohol & Pharmacodependences (GRAP), Centre Universitaire de Recherche en Santé (CURS) , INSERM U1247, University of Picardie Jules Verne, Amiens, France
| | - Charles-Antoine Papillon
- Research Group on Alcohol & Pharmacodependences (GRAP), Centre Universitaire de Recherche en Santé (CURS) , INSERM U1247, University of Picardie Jules Verne, Amiens, France
| | - Margaret Martinetti
- Research Group on Alcohol & Pharmacodependences (GRAP), Centre Universitaire de Recherche en Santé (CURS) , INSERM U1247, University of Picardie Jules Verne, Amiens, France.,Department of Psychology , The College of New Jersey, Ewing, New Jersey
| | - Johann Antol
- Research Group on Alcohol & Pharmacodependences (GRAP), Centre Universitaire de Recherche en Santé (CURS) , INSERM U1247, University of Picardie Jules Verne, Amiens, France
| | - Cyrille Guillaumont
- Research Group on Alcohol & Pharmacodependences (GRAP), Centre Universitaire de Recherche en Santé (CURS) , INSERM U1247, University of Picardie Jules Verne, Amiens, France.,Unité d'alcoologie SESAME , Centre hospitalier Psychiatrique Philippe Pinel, Amiens, France
| | - Alain Dervaux
- Research Group on Alcohol & Pharmacodependences (GRAP), Centre Universitaire de Recherche en Santé (CURS) , INSERM U1247, University of Picardie Jules Verne, Amiens, France.,CHU Sud , Service de consultations de Psychiatrie et Addictologie, Amiens Cedex, France
| | - Mickael Naassila
- Research Group on Alcohol & Pharmacodependences (GRAP), Centre Universitaire de Recherche en Santé (CURS) , INSERM U1247, University of Picardie Jules Verne, Amiens, France
| |
Collapse
|
7
|
Rotermund C, Reolon GK, Leixner S, Boden C, Bilbao A, Kahle PJ. Enhanced motivation to alcohol in transgenic mice expressing human α-synuclein. J Neurochem 2017; 143:294-305. [PMID: 28833174 DOI: 10.1111/jnc.14151] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Revised: 07/20/2017] [Accepted: 08/14/2017] [Indexed: 12/17/2022]
Abstract
α-Synuclein (αSYN) is the neuropathological hallmark protein of Parkinson's disease (PD) and related neurodegenerative disorders. Moreover, the gene encoding αSYN (SNCA) is a major genetic contributor to PD. Interestingly, independent genome-wide association studies also identified SNCA as the most important candidate gene for alcoholism. Furthermore, single-nucleotide-polymorphisms have been associated with alcohol-craving behavior and alcohol-craving patients showed augmented αSYN expression in blood. To investigate the effect of αSYN on the addictive properties of chronic alcohol use, we examined consumption, motivation, and seeking responses induced by environmental stimuli and relapse behavior in transgenic mice expressing the human mutant [A30P]αSYN throughout the brain. The primary reinforcing effects of alcohol under operant self-administration conditions were increased, while consumption and the alcohol deprivation effect were not altered in the transgenic mice. The same mice were subjected to immunohistochemical measurements of immediate-early gene inductions in brain regions involved in addiction-related behaviors. Acute ethanol injection enhanced immunostaining for the phosphorylated form of cAMP response element binding protein in both amygdala and nucleus accumbens of αSYN transgenic mice, while in wild-type mice no effect was visible. However, at the same time, levels of cFos remain unchanged in both genotypes. These results provide experimental confirmation of SNCA as a candidate gene for alcoholism in addition to its known link to PD.
Collapse
Affiliation(s)
- Carola Rotermund
- Laboratory of Functional Neurogenetics, Department of Neurodegeneration, German Center of Neurodegenerative Diseases, Tübingen, Germany
| | - Gustavo K Reolon
- Laboratory of Functional Neurogenetics, Department of Neurodegeneration, Hertie Institute of Clinical Brain Research, Faculty of Medicine, University of Tübingen, Tübingen, Germany
| | - Sarah Leixner
- Behavioral Genetics Research Group, Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty of Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Cindy Boden
- Laboratory of Functional Neurogenetics, Department of Neurodegeneration, German Center of Neurodegenerative Diseases, Tübingen, Germany
| | - Ainhoa Bilbao
- Behavioral Genetics Research Group, Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty of Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Philipp J Kahle
- Laboratory of Functional Neurogenetics, Department of Neurodegeneration, German Center of Neurodegenerative Diseases, Tübingen, Germany.,Laboratory of Functional Neurogenetics, Department of Neurodegeneration, Hertie Institute of Clinical Brain Research, Faculty of Medicine, University of Tübingen, Tübingen, Germany
| |
Collapse
|
8
|
Walter NAR, Denmark DL, Kozell LB, Buck KJ. A Systems Approach Implicates a Brain Mitochondrial Oxidative Homeostasis Co-expression Network in Genetic Vulnerability to Alcohol Withdrawal. Front Genet 2017; 7:218. [PMID: 28096806 PMCID: PMC5206817 DOI: 10.3389/fgene.2016.00218] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 12/08/2016] [Indexed: 12/31/2022] Open
Abstract
Genetic factors significantly affect vulnerability to alcohol dependence (alcoholism). We previously identified quantitative trait loci on distal mouse chromosome 1 with large effects on predisposition to alcohol physiological dependence and associated withdrawal following both chronic and acute alcohol exposure in mice (Alcdp1 and Alcw1, respectively). We fine-mapped these loci to a 1.1–1.7 Mb interval syntenic with human 1q23.2-23.3. Alcw1/Alcdp1 interval genes show remarkable genetic variation among mice derived from the C57BL/6J and DBA/2J strains, the two most widely studied genetic animal models for alcohol-related traits. Here, we report the creation of a novel recombinant Alcw1/Alcdp1 congenic model (R2) in which the Alcw1/Alcdp1 interval from a donor C57BL/6J strain is introgressed onto a uniform, inbred DBA/2J genetic background. As expected, R2 mice demonstrate significantly less severe alcohol withdrawal compared to wild-type littermates. Additionally, comparing R2 and background strain animals, as well as reciprocal congenic (R8) and appropriate background strain animals, we assessed Alcw1/Alcdp1 dependent brain gene expression using microarray and quantitative PCR analyses. To our knowledge this includes the first Weighted Gene Co-expression Network Analysis using reciprocal congenic models. Importantly, this allows detection of co-expression patterns limited to one or common to both genetic backgrounds with high or low predisposition to alcohol withdrawal severity. The gene expression patterns (modules) in common contain genes related to oxidative phosphorylation, building upon human and animal model studies that implicate involvement of oxidative phosphorylation in alcohol use disorders (AUDs). Finally, we demonstrate that administration of N-acetylcysteine, an FDA-approved antioxidant, significantly reduces symptoms of alcohol withdrawal (convulsions) in mice, thus validating a phenotypic role for this network. Taken together, these studies support the importance of mitochondrial oxidative homeostasis in alcohol withdrawal and identify this network as a valuable therapeutic target in human AUDs.
Collapse
Affiliation(s)
- Nicole A R Walter
- Research and Development, Portland Veterans Affairs Medical Center, PortlandOR, USA; Department of Behavioral Neuroscience, School of Medicine, Oregon Health and Science University, PortlandOR, USA
| | - DeAunne L Denmark
- Research and Development, Portland Veterans Affairs Medical Center, PortlandOR, USA; Department of Behavioral Neuroscience, School of Medicine, Oregon Health and Science University, PortlandOR, USA
| | - Laura B Kozell
- Research and Development, Portland Veterans Affairs Medical Center, PortlandOR, USA; Department of Behavioral Neuroscience, School of Medicine, Oregon Health and Science University, PortlandOR, USA
| | - Kari J Buck
- Research and Development, Portland Veterans Affairs Medical Center, PortlandOR, USA; Department of Behavioral Neuroscience, School of Medicine, Oregon Health and Science University, PortlandOR, USA
| |
Collapse
|
9
|
Qiu B, Bell RL, Cao Y, Zhang L, Stewart RB, Graves T, Lumeng L, Yong W, Liang T. Npy deletion in an alcohol non-preferring rat model elicits differential effects on alcohol consumption and body weight. J Genet Genomics 2016; 43:421-30. [PMID: 27461754 PMCID: PMC5055068 DOI: 10.1016/j.jgg.2016.04.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2015] [Revised: 04/21/2016] [Accepted: 04/27/2016] [Indexed: 11/21/2022]
Abstract
Neuropeptide Y (NPY) is widely expressed in the central nervous system and influences many physiological processes. It is located within the rat quantitative trait locus (QTL) for alcohol preference on chromosome 4. Alcohol-nonpreferring (NP) rats consume very little alcohol, but have significantly higher NPY expression in the brain than alcohol-preferring (P) rats. We capitalized on this phenotypic difference by creating an Npy knockout (KO) rat using the inbred NP background to evaluate NPY effects on alcohol consumption. Zinc finger nuclease (ZNF) technology was applied, resulting in a 26-bp deletion in the Npy gene. RT-PCR, Western blotting and immunohistochemistry confirmed the absence of Npy mRNA and protein in KO rats. Alcohol consumption was increased in Npy(+/-) but not Npy(-/-) rats, while Npy(-/-) rats displayed significantly lower body weight when compared to Npy(+/+) rats. In whole brain tissue, expression levels of Npy-related and other alcohol-associated genes, Npy1r, Npy2r, Npy5r, Agrp, Mc3r, Mc4r, Crh and Crh1r, were significantly greater in Npy(-/-) rats, whereas Pomc and Crhr2 expressions were highest in Npy(+/-) rats. These findings suggest that the NPY-system works in close coordination with the melanocortin (MC) and corticotropin-releasing hormone (CRH) systems to modulate alcohol intake and body weight.
Collapse
Affiliation(s)
- Bin Qiu
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100021, China
| | - Richard L Bell
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Yong Cao
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100021, China; Experimental Medicine Center, The First Affiliated Hospital of Sichuan Medical University, Luzhou 646000, China
| | - Lingling Zhang
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100021, China
| | - Robert B Stewart
- Department of Psychology, Purdue School of Science, Indiana University-Purdue University of Indianapolis, Indianapolis, IN 46202, USA
| | - Tamara Graves
- Department of Gastroenterology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Lawrence Lumeng
- Department of Gastroenterology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Weidong Yong
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100021, China.
| | - Tiebing Liang
- Department of Gastroenterology, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
10
|
Gautam US, McGillivray A, Mehra S, Didier PJ, Midkiff CC, Kissee RS, Golden NA, Alvarez X, Niu T, Rengarajan J, Sherman DR, Kaushal D. DosS Is required for the complete virulence of mycobacterium tuberculosis in mice with classical granulomatous lesions. Am J Respir Cell Mol Biol 2015; 52:708-16. [PMID: 25322074 DOI: 10.1165/rcmb.2014-0230oc] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Mycobacterium tuberculosis (Mtb) must counter hypoxia within granulomas to persist. DosR, in concert with sensor kinases DosS and DosT, regulates the response to hypoxia. Yet Mtb lacking functional DosR colonize the lungs of C57Bl/6 mice, presumably owing to the lack of organized lesions with sufficient hypoxia in that model. We compared the phenotype of the Δ-dosR, Δ-dosS, and Δ-dosT mutants to Mtb using C3HeB/FeJ mice, an alternate mouse model where lesions develop hypoxia. C3HeB/FeJ mice were infected via aerosol. The progression of infection was analyzed by tissue bacterial burden and histopathology. A measure of the comparative global immune responses was also analyzed. Although Δ-dosR and Δ-dosT grew comparably to wild-type Mtb, Δ-dosS exhibited a significant defect in bacterial burden and pathology in vivo, accompanied by ablated proinflammatory response. Δ-dosS retained the ability to induce DosR. The Δ-dosS mutant was also attenuated in murine macrophages ex vivo, with evidence of reduced expression of the proinflammatory signature. Our results show that DosS, but not DosR and DosT, is required by Mtb to survive in C3HeB/FeJ mice. The attenuation of Δ-dosS is not due to its inability to induce the DosR regulon, nor is it a result of the accumulation of hypoxia. That the in vivo growth restriction of Δ-dosS could be mimicked ex vivo suggested sensitivity to macrophage oxidative burst. Anoxic caseous centers within tuberculosis lesions eventually progress to cavities. Our results provide greater insight into the molecular mechanisms of Mtb persistence within host lungs.
Collapse
|
11
|
An α-synuclein gene (SNCA) polymorphism moderates the association of PTSD symptomatology with hazardous alcohol use, but not with aggression-related measures. J Anxiety Disord 2015; 30:41-7. [PMID: 25594371 PMCID: PMC4355301 DOI: 10.1016/j.janxdis.2014.12.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Revised: 11/30/2014] [Accepted: 12/15/2014] [Indexed: 01/25/2023]
Abstract
Posttraumatic stress disorder (PTSD) often precedes comorbid substance use disorder and has been associated with aggression. Prior research has evidenced that alcohol use and other externalizing behaviors share genetic factors with PTSD; however, few studies have examined if specific genes are associated with externalizing behaviors in PTSD. The purpose of the current study was to investigate whether an α-synuclein gene polymorphism (SNCA rs356195) moderates the association of PTSD symptomatology with externalizing behaviors. We examined the separate and combined effects of PTSD symptomatology and SNCA rs356195 on alcohol- and aggression-related measures in nonclinical participants (N=138 European Americans; 15 diagnosed with probable PTSD). Probable PTSD status and SNCA were both associated with externalizing measures. SNCA also moderated the association of PTSD symptomatology with hazardous alcohol use, but not with aggression-related measures. Current findings suggest that variations in SNCA may increase the likelihood that PTSD symptomatology results in excessive alcohol use.
Collapse
|
12
|
Juraeva D, Treutlein J, Scholz H, Frank J, Degenhardt F, Cichon S, Ridinger M, Mattheisen M, Witt SH, Lang M, Sommer WH, Hoffmann P, Herms S, Wodarz N, Soyka M, Zill P, Maier W, Jünger E, Gaebel W, Dahmen N, Scherbaum N, Schmäl C, Steffens M, Lucae S, Ising M, Smolka MN, Zimmermann US, Müller-Myhsok B, Nöthen MM, Mann K, Kiefer F, Spanagel R, Brors B, Rietschel M. XRCC5 as a risk gene for alcohol dependence: evidence from a genome-wide gene-set-based analysis and follow-up studies in Drosophila and humans. Neuropsychopharmacology 2015; 40:361-71. [PMID: 25035082 PMCID: PMC4443948 DOI: 10.1038/npp.2014.178] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Revised: 06/06/2014] [Accepted: 06/08/2014] [Indexed: 12/15/2022]
Abstract
Genetic factors have as large role as environmental factors in the etiology of alcohol dependence (AD). Although genome-wide association studies (GWAS) enable systematic searches for loci not hitherto implicated in the etiology of AD, many true findings may be missed owing to correction for multiple testing. The aim of the present study was to circumvent this limitation by searching for biological system-level differences, and then following up these findings in humans and animals. Gene-set-based analysis of GWAS data from 1333 cases and 2168 controls identified 19 significantly associated gene-sets, of which 5 could be replicated in an independent sample. Clustered in these gene-sets were novel and previously identified susceptibility genes. The most frequently present gene, ie in 6 out of 19 gene-sets, was X-ray repair complementing defective repair in Chinese hamster cells 5 (XRCC5). Previous human and animal studies have implicated XRCC5 in alcohol sensitivity. This phenotype is inversely correlated with the development of AD, presumably as more alcohol is required to achieve the desired effects. In the present study, the functional role of XRCC5 in AD was further validated in animals and humans. Drosophila mutants with reduced function of Ku80-the homolog of mammalian XRCC5-due to RNAi silencing showed reduced sensitivity to ethanol. In humans with free access to intravenous ethanol self-administration in the laboratory, the maximum achieved blood alcohol concentration was influenced in an allele-dose-dependent manner by genetic variation in XRCC5. In conclusion, our convergent approach identified new candidates and generated independent evidence for the involvement of XRCC5 in alcohol dependence.
Collapse
Affiliation(s)
- Dilafruz Juraeva
- Division of Theoretical Bioinformatics, German Cancer Research Center, Heidelberg, Germany
| | - Jens Treutlein
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Henrike Scholz
- Department of Animal Physiology, University of Cologne, Cologne, Germany
| | - Josef Frank
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Franziska Degenhardt
- Institute of Human Genetics, University of Bonn, Bonn, Germany,Department of Genomics, Life & Brain Center, University of Bonn, Bonn, Germany
| | - Sven Cichon
- Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| | - Monika Ridinger
- Department of Psychiatry, University of Regensburg, Regensburg, Germany
| | | | - Stephanie H Witt
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Maren Lang
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Wolfgang H Sommer
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Per Hoffmann
- Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| | - Stefan Herms
- Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| | - Norbert Wodarz
- Department of Psychiatry, University of Regensburg, Regensburg, Germany
| | - Michael Soyka
- Private Hospital Meiringen, Meiringen, Switzerland,Department of Psychiatry, University of Munich, Munich, Germany
| | - Peter Zill
- Department of Psychiatry, University of Munich, Munich, Germany
| | - Wolfgang Maier
- Department of Psychiatry, University of Bonn, Bonn, Germany
| | - Elisabeth Jünger
- Department of Psychiatry and Psychotherapy, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden
| | - Wolfgang Gaebel
- Department of Psychiatry and Psychotherapy, University of Düsseldorf, Düsseldorf, Germany
| | - Norbert Dahmen
- Department of Psychiatry, University of Mainz, Mainz, Germany
| | - Norbert Scherbaum
- Addiction Research Group at the Department of Psychiatry and Psychotherapy, University of Duisburg-Essen, Essen, Germany
| | - Christine Schmäl
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Michael Steffens
- Division of Research, Federal Institute for Drugs and Medical Devices, Bonn, Germany
| | - Susanne Lucae
- Department of Psychiatric Pharmacogenetics, Max-Planck-Institute of Psychiatry, München, Germany
| | - Marcus Ising
- Department of Molecular Psychology, Max-Planck-Institute of Psychiatry, München, Germany
| | - Michael N Smolka
- Department of Psychiatry and Psychotherapy, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden
| | - Ulrich S Zimmermann
- Department of Psychiatry and Psychotherapy, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden
| | - Bertram Müller-Myhsok
- Department of Statistical Genetics, Max-Planck-Institute of Psychiatry, München, Germany,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany,Institute of Translational Medicine Liverpool, University of Liverpool, Liverpool, UK
| | - Markus M Nöthen
- Institute of Human Genetics, University of Bonn, Bonn, Germany,Department of Genomics, Life & Brain Center, University of Bonn, Bonn, Germany
| | - Karl Mann
- Department of Addictive Behavior and Addiction Medicine, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Falk Kiefer
- Department of Addictive Behavior and Addiction Medicine, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Rainer Spanagel
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Benedikt Brors
- Division of Theoretical Bioinformatics, German Cancer Research Center, Heidelberg, Germany
| | - Marcella Rietschel
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany,Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, University Medical Center Mannheim, University of Heidelberg, J5, Mannheim 68159, Germany, Tel: +49 621 1703 6051, Fax: +49 621 1703 6055, E-mail:
| |
Collapse
|
13
|
Vadnie CA, Park JH, Abdel Gawad N, Ho AMC, Hinton DJ, Choi DS. Gut-brain peptides in corticostriatal-limbic circuitry and alcohol use disorders. Front Neurosci 2014; 8:288. [PMID: 25278825 PMCID: PMC4166902 DOI: 10.3389/fnins.2014.00288] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 08/26/2014] [Indexed: 12/22/2022] Open
Abstract
Peptides synthesized in endocrine cells in the gastrointestinal tract and neurons are traditionally considered regulators of metabolism, energy intake, and appetite. However, recent work has demonstrated that many of these peptides act on corticostriatal-limbic circuitry and, in turn, regulate addictive behaviors. Given that alcohol is a source of energy and an addictive substance, it is not surprising that increasing evidence supports a role for gut-brain peptides specifically in alcohol use disorders (AUD). In this review, we discuss the effects of several gut-brain peptides on alcohol-related behaviors and the potential mechanisms by which these gut-brain peptides may interfere with alcohol-induced changes in corticostriatal-limbic circuitry. This review provides a summary of current knowledge on gut-brain peptides focusing on five peptides: neurotensin, glucagon-like peptide 1, ghrelin, substance P, and neuropeptide Y. Our review will be helpful to develop novel therapeutic targets for AUD.
Collapse
Affiliation(s)
- Chelsea A Vadnie
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine Rochester, MN, USA ; Neurobiology of Disease Program, Mayo Clinic College of Medicine Rochester, MN, USA
| | - Jun Hyun Park
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine Rochester, MN, USA ; Department of Psychiatry, Sanggye Paik Hospital, College of Medicine, InJe University Seoul, South Korea
| | - Noha Abdel Gawad
- Department of Psychiatry and Psychology, Mayo Clinic College of Medicine Rochester, MN, USA
| | - Ada Man Choi Ho
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine Rochester, MN, USA ; Department of Psychiatry and Psychology, Mayo Clinic College of Medicine Rochester, MN, USA
| | - David J Hinton
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine Rochester, MN, USA ; Neurobiology of Disease Program, Mayo Clinic College of Medicine Rochester, MN, USA
| | - Doo-Sup Choi
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine Rochester, MN, USA ; Neurobiology of Disease Program, Mayo Clinic College of Medicine Rochester, MN, USA ; Department of Psychiatry and Psychology, Mayo Clinic College of Medicine Rochester, MN, USA
| |
Collapse
|
14
|
Morozova TV, Mackay TFC, Anholt RRH. Genetics and genomics of alcohol sensitivity. Mol Genet Genomics 2014; 289:253-69. [PMID: 24395673 PMCID: PMC4037586 DOI: 10.1007/s00438-013-0808-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Accepted: 12/22/2013] [Indexed: 01/20/2023]
Abstract
Alcohol abuse and alcoholism incur a heavy socioeconomic cost in many countries. Both genetic and environmental factors contribute to variation in the inebriating effects of alcohol and alcohol addiction among individuals within and across populations. From a genetics perspective, alcohol sensitivity is a quantitative trait determined by the cumulative effects of multiple segregating genes and their interactions with the environment. This review summarizes insights from model organisms as well as human populations that represent our current understanding of the genetic and genomic underpinnings that govern alcohol metabolism and the sedative and addictive effects of alcohol on the nervous system.
Collapse
Affiliation(s)
- Tatiana V. Morozova
- Department of Biological Sciences and W. M. Keck Center for Behavioral Biology, North Carolina State University, Box 7617, Raleigh, NC 27695-7617 USA
| | - Trudy F. C. Mackay
- Department of Biological Sciences and W. M. Keck Center for Behavioral Biology, North Carolina State University, Box 7617, Raleigh, NC 27695-7617 USA
| | - Robert R. H. Anholt
- Department of Biological Sciences and W. M. Keck Center for Behavioral Biology, North Carolina State University, Box 7617, Raleigh, NC 27695-7617 USA
| |
Collapse
|
15
|
Janeczek P, MacKay RK, Lea RA, Dodd PR, Lewohl JM. Reduced expression of α-synuclein in alcoholic brain: influence of SNCA-Rep1 genotype. Addict Biol 2014; 19:509-15. [PMID: 22974310 DOI: 10.1111/j.1369-1600.2012.00495.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
α-Synuclein has recently been implicated in the pathophysiology of alcohol abuse due to its role in dopaminergic neurotransmission. In these studies, genetic variability in the α-synuclein gene influences its expression which may contribute to susceptibility to chronic alcohol abuse. Real-time PCR was used to quantify α-synuclein mRNA expression in autopsy samples of human dorsolateral prefrontal cortex. Because of the association between length of the α-synuclein-repeat 1 microsatellite marker and expression levels of the gene, this marker was genotyped in a Caucasian sample of 126 controls and 117 alcoholics using capillary gel electrophoresis. The allele and genotype frequencies of α-synuclein-repeat 1 marker differed significantly between alcoholics and controls. Alcoholics had greater frequencies of the shortest allele found (267 bp). The shortest allele of the α-synuclein-repeat 1 marker was associated with decreased expression of α-synuclein in prefrontal cortex. Individuals with at least one copy of the 267 bp allele were more likely to exhibit an alcohol abuse phenotype. These results suggest that individuals with the 267 bp allele may be at increased risk of developing alcoholism and that genetic variation at the α-synuclein-repeat 1 locus may influence α-synuclein expression in the prefrontal cortex.
Collapse
Affiliation(s)
- Paulina Janeczek
- Griffith Health Institute, School of Medical Sciences; Griffith University; Australia
| | - Rachel K. MacKay
- Griffith Health Institute, School of Medical Sciences; Griffith University; Australia
| | - Rodney A. Lea
- Griffith Health Institute, School of Medical Sciences; Griffith University; Australia
| | - Peter R. Dodd
- School of Chemistry and Molecular Biosciences; University of Queensland; Australia
| | - Joanne M. Lewohl
- Griffith Health Institute, School of Medical Sciences; Griffith University; Australia
| |
Collapse
|
16
|
Genetic risk prediction and neurobiological understanding of alcoholism. Transl Psychiatry 2014; 4:e391. [PMID: 24844177 PMCID: PMC4035721 DOI: 10.1038/tp.2014.29] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Accepted: 03/18/2014] [Indexed: 01/08/2023] Open
Abstract
We have used a translational Convergent Functional Genomics (CFG) approach to discover genes involved in alcoholism, by gene-level integration of genome-wide association study (GWAS) data from a German alcohol dependence cohort with other genetic and gene expression data, from human and animal model studies, similar to our previous work in bipolar disorder and schizophrenia. A panel of all the nominally significant P-value SNPs in the top candidate genes discovered by CFG (n=135 genes, 713 SNPs) was used to generate a genetic risk prediction score (GRPS), which showed a trend towards significance (P=0.053) in separating alcohol dependent individuals from controls in an independent German test cohort. We then validated and prioritized our top findings from this discovery work, and subsequently tested them in three independent cohorts, from two continents. A panel of all the nominally significant P-value single-nucleotide length polymorphisms (SNPs) in the top candidate genes discovered by CFG (n=135 genes, 713 SNPs) were used to generate a Genetic Risk Prediction Score (GRPS), which showed a trend towards significance (P=0.053) in separating alcohol-dependent individuals from controls in an independent German test cohort. In order to validate and prioritize the key genes that drive behavior without some of the pleiotropic environmental confounds present in humans, we used a stress-reactive animal model of alcoholism developed by our group, the D-box binding protein (DBP) knockout mouse, consistent with the surfeit of stress theory of addiction proposed by Koob and colleagues. A much smaller panel (n=11 genes, 66 SNPs) of the top CFG-discovered genes for alcoholism, cross-validated and prioritized by this stress-reactive animal model showed better predictive ability in the independent German test cohort (P=0.041). The top CFG scoring gene for alcoholism from the initial discovery step, synuclein alpha (SNCA) remained the top gene after the stress-reactive animal model cross-validation. We also tested this small panel of genes in two other independent test cohorts from the United States, one with alcohol dependence (P=0.00012) and one with alcohol abuse (a less severe form of alcoholism; P=0.0094). SNCA by itself was able to separate alcoholics from controls in the alcohol-dependent cohort (P=0.000013) and the alcohol abuse cohort (P=0.023). So did eight other genes from the panel of 11 genes taken individually, albeit to a lesser extent and/or less broadly across cohorts. SNCA, GRM3 and MBP survived strict Bonferroni correction for multiple comparisons. Taken together, these results suggest that our stress-reactive DBP animal model helped to validate and prioritize from the CFG-discovered genes some of the key behaviorally relevant genes for alcoholism. These genes fall into a series of biological pathways involved in signal transduction, transmission of nerve impulse (including myelination) and cocaine addiction. Overall, our work provides leads towards a better understanding of illness, diagnostics and therapeutics, including treatment with omega-3 fatty acids. We also examined the overlap between the top candidate genes for alcoholism from this work and the top candidate genes for bipolar disorder, schizophrenia, anxiety from previous CFG analyses conducted by us, as well as cross-tested genetic risk predictions. This revealed the significant genetic overlap with other major psychiatric disorder domains, providing a basis for comorbidity and dual diagnosis, and placing alcohol use in the broader context of modulating the mental landscape.
Collapse
|
17
|
Zhou Z, Enoch MA, Goldman D. Gene expression in the addicted brain. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2014; 116:251-73. [PMID: 25172478 DOI: 10.1016/b978-0-12-801105-8.00010-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Addiction is due to changes in the structure and function of the brain, including neuronal networks and the cells that comprise them. Within cells, gene expression changes can track and help explain their altered function. Transcriptional changes induced by addictive agents are dynamic and divergent and range from signal pathway-specific perturbations to widespread molecular and cellular dysregulation that can be measured by "omic" methods and that can be used to identify new pathways. The molecular effects of addiction depend on timing of exposure or withdrawal, the stage of adaptation, the brain region, and the behavioral model, there being many models of addiction. However, the molecular neural adaptations across different drug exposures, conditions, and regions are to some extent shared and can reflect common actions on pathways relevant to addiction. Epigenetic studies of DNA methylation and histone modifications and studies of regulatory RNA networks have been informative for elucidating the mechanisms of transcriptional change in the addicted brain.
Collapse
Affiliation(s)
- Zhifeng Zhou
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, USA.
| | - Mary-Anne Enoch
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, USA
| | - David Goldman
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
18
|
Abstract
Identification of genes influencing complex traits is hampered by genetic heterogeneity, the modest effect size of many alleles, and the likely involvement of rare and uncommon alleles. Etiologic complexity can be simplified in model organisms. By genomic sequencing, linkage analysis, and functional validation, we identified that genetic variation of Grm2, which encodes metabotropic glutamate receptor 2 (mGluR2), alters alcohol preference in animal models. Selectively bred alcohol-preferring (P) rats are homozygous for a Grm2 stop codon (Grm2 *407) that leads to largely uncompensated loss of mGluR2. mGluR2 receptor expression was absent, synaptic glutamate transmission was impaired, and expression of genes involved in synaptic function was altered. Grm2 *407 was linked to increased alcohol consumption and preference in F2 rats generated by intercrossing inbred P and nonpreferring rats. Pharmacologic blockade of mGluR2 escalated alcohol self-administration in Wistar rats, the parental strain of P and nonpreferring rats. The causal role of mGluR2 in altered alcohol preference was further supported by elevated alcohol consumption in Grm2 (-/-) mice. Together, these data point to mGluR2 as an origin of alcohol preference and a potential therapeutic target.
Collapse
|
19
|
Relationship between differential hepatic microRNA expression and decreased hepatic cytochrome P450 3A activity in cirrhosis. PLoS One 2013; 8:e74471. [PMID: 24058572 PMCID: PMC3772944 DOI: 10.1371/journal.pone.0074471] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Accepted: 07/31/2013] [Indexed: 12/17/2022] Open
Abstract
Background and Aim Liver cirrhosis is associated with decreased hepatic cytochrome P4503A (CYP3A) activity but the pathogenesis of this phenomenon is not well elucidated. In this study, we examined if certain microRNAs (miRNA) are associated with decreased hepatic CYP3A activity in cirrhosis. Methods Hepatic CYP3A activity and miRNA microarray expression profiles were measured in cirrhotic (n=28) and normal (n=12) liver tissue. Hepatic CYP3A activity was measured via midazolam hydroxylation in human liver microsomes. Additionally, hepatic CYP3A4 protein concentration and the expression of CYP3A4 mRNA were measured. Analyses were conducted to identify miRNAs which were differentially expressed between two groups but also were significantly associated with lower hepatic CYP3A activity. Results Hepatic CYP3A activity in cirrhotic livers was 1.7-fold lower than in the normal livers (0.28 ± 0.06 vs. 0.47 ± 0.07mL* min-1*mg protein-1 (mean ± SEM), P=0.02). Six microRNAs (miR-155, miR-454, miR-582-5p, let-7f-1*, miR-181d, and miR-500) had >1.2-fold increase in cirrhotic livers and also had significant negative correlation with hepatic CYP3A activity (range of r = -0.44 to -0.52, P <0.05). Notably, miR-155, a known regulator of liver inflammation, had the highest fold increase in cirrhotic livers (2.2-fold, P=4.16E-08) and significantly correlated with hepatic CYP3A activity (r=-0.50, P=0.017). The relative expression (2-ΔΔCt mean ± SEM) of hepatic CYP3A4 mRNA was significantly higher in cirrhotic livers (21.76 ± 2.65 vs. 5.91 ± 1.29, P=2.04E-07) but their levels did not significantly correlate with hepatic CYP3A activity (r=-0.43, P=0.08). Conclusion The strong association between certain miRNAs, notably miR-155, and lower hepatic CYP3A activity suggest that altered miRNA expression may regulate hepatic CYP3A activity.
Collapse
|
20
|
Whole brain and brain regional coexpression network interactions associated with predisposition to alcohol consumption. PLoS One 2013; 8:e68878. [PMID: 23894363 PMCID: PMC3720886 DOI: 10.1371/journal.pone.0068878] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Accepted: 06/01/2013] [Indexed: 01/02/2023] Open
Abstract
To identify brain transcriptional networks that may predispose an animal to consume alcohol, we used weighted gene coexpression network analysis (WGCNA). Candidate coexpression modules are those with an eigengene expression level that correlates significantly with the level of alcohol consumption across a panel of BXD recombinant inbred mouse strains, and that share a genomic region that regulates the module transcript expression levels (mQTL) with a genomic region that regulates alcohol consumption (bQTL). To address a controversy regarding utility of gene expression profiles from whole brain, vs specific brain regions, as indicators of the relationship of gene expression to phenotype, we compared candidate coexpression modules from whole brain gene expression data (gathered with Affymetrix 430 v2 arrays in the Colorado laboratories) and from gene expression data from 6 brain regions (nucleus accumbens (NA); prefrontal cortex (PFC); ventral tegmental area (VTA); striatum (ST); hippocampus (HP); cerebellum (CB)) available from GeneNetwork. The candidate modules were used to construct candidate eigengene networks across brain regions, resulting in three "meta-modules", composed of candidate modules from two or more brain regions (NA, PFC, ST, VTA) and whole brain. To mitigate the potential influence of chromosomal location of transcripts and cis-eQTLs in linkage disequilibrium, we calculated a semi-partial correlation of the transcripts in the meta-modules with alcohol consumption conditional on the transcripts' cis-eQTLs. The function of transcripts that retained the correlation with the phenotype after correction for the strong genetic influence, implicates processes of protein metabolism in the ER and Golgi as influencing susceptibility to variation in alcohol consumption. Integration of these data with human GWAS provides further information on the function of polymorphisms associated with alcohol-related traits.
Collapse
|
21
|
Janeczek P, Lewohl JM. The role of α-synuclein in the pathophysiology of alcoholism. Neurochem Int 2013; 63:154-62. [PMID: 23791711 DOI: 10.1016/j.neuint.2013.06.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Revised: 06/06/2013] [Accepted: 06/08/2013] [Indexed: 12/14/2022]
Abstract
Alcoholism has complex etiology and there is evidence for both genetic and environmental factors in its pathophysiology. Chronic, long-term alcohol abuse and alcohol dependence are associated with neuronal loss with the prefrontal cortex being particularly susceptible to neurotoxic damage. This brain region is involved in the development and persistence of alcohol addiction and neurotoxic damage is likely to exacerbate the reinforcing effects of alcohol and may hinder treatment. Understanding the mechanism of alcohol's neurotoxic effects on the brain and the genetic risk factors associated with alcohol abuse are the focus of current research. Because of its well-established role in neurodegenerative and neuropsychological disorders, and its emerging role in the pathophysiology of addiction, here we review the genetic and epigenetic factors involved in regulating α-synuclein expression and its potential role in the pathophysiology of chronic alcohol abuse. Elucidation of the mechanisms of α-synuclein regulation may prove beneficial in understanding the role of this key synaptic protein in disease and its potential for therapeutic modulation in the treatment of substance use disorders as well as other neurodegenerative diseases.
Collapse
Affiliation(s)
- Paulina Janeczek
- Griffith Health Institute, School of Medical Sciences, Griffith University, Gold Coast Campus, Southport, Australia
| | | |
Collapse
|
22
|
Spence JP, Lai D, Shekhar A, Carr LG, Foroud T, Liang T. Quantitative trait locus for body weight identified on rat chromosome 4 in inbred alcohol-preferring and -nonpreferring rats: potential implications for neuropeptide Y and corticotrophin releasing hormone 2. Alcohol 2013; 47:63-7. [PMID: 23312492 DOI: 10.1016/j.alcohol.2012.10.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2012] [Revised: 10/10/2012] [Accepted: 10/11/2012] [Indexed: 10/27/2022]
Abstract
The alcohol-preferring (P) and -nonpreferring (NP) rat lines were developed using bidirectional selective breeding for alcohol consumption (g/kg/day) and alcohol preference (water:ethanol ratio). During a preliminary study, we detected a difference in body weight between inbred P (iP) and inbred NP (iNP) rats that appeared to be associated with the transfer of the Chromosome 4 quantitative trait locus (QTL) seen in the P.NP and NP.P congenic strains. After the initial confirmation that iP rats displayed lower body weight when compared to iNP rats (data not shown), body weight and growth rates of each chromosome 4 reciprocal congenic rat strain (P.NP and NP.P) were measured, and their body weight was consistent with their respective donor strain phenotype, confirming that a quantitative trait locus for body weight mapped to the chromosome 4 interval. Utilizing the newly developed interval-specific congenic strains (ISCS-A and ISCS-B), the QTL interval was further narrowed identifying the following candidate genes of interest: neuropeptide Y (Npy), juxtaposed with another zinc finger gene 1 (Jazf1), corticotrophin releasing factor receptor 2 (Crfr2) and LanC lantibiotic synthetase component C-like 2 (Lancl2). These findings indicate that a biologically active variant(s) regulates body weight on rat chromosome 4 in iP and iNP rats. This QTL for body weight was successfully captured in the P.NP and NP.P congenic strains, and interval-specific congenic strains (ISCSs) were subsequently employed to fine-map the QTL interval identifying the following candidate genes of interest: Npy, Jazf1, Crfr2 and Lancl2. Both Npy and Crfr2 have been previously identified as candidate genes of interest underlying the chromosome 4 QTL for alcohol consumption in iP and iNP rats.
Collapse
|
23
|
Abstract
Animal models have been successfully developed to mimic and study alcoholism. These models have the unique feature of allowing the researcher to control for the genetic characteristics of the animal, alcohol exposure and environment. Moreover, these animal models allow pharmacological, neurochemical and behavioral manipulations otherwise impossible. Unquestionably, one of the major contributions to the understanding of the neurobiological basis of alcoholism comes from data that have been obtained from the study of genetically selected alcohol preferring rat lines and from the consequences that alcohol drinking and environmental manipulations, (i.e., protracted alcohol drinking, intoxication, exposure to stress, etc.) have on them. In fact, if on the one hand genetic factors may account for about 50-60% of the risk of developing alcohol dependence, on the other hand protracted alcohol exposure is a necessary precondition to actually develop the disease, while environmental vulnerability factors may be crucial for disease progression. The present article will offer an overview of the different genetically selected alcohol preferring rat lines developed and used to study alcoholism. The predictive, face and construct validity of these animal models and the translational significance of findings achieved through their use will be critically discussed.
Collapse
Affiliation(s)
- Roberto Ciccocioppo
- Pharmacology Unit, School of Pharmacy, University of Camerino, Madonna delle Carceri 9, 62032 Camerino, MC, Italy.
| |
Collapse
|
24
|
Zhao Z, Guo AY, van den Oord EJCG, Aliev F, Jia P, Edenberg HJ, Riley BP, Dick DM, Bettinger JC, Davies AG, Grotewiel MS, Schuckit MA, Agrawal A, Kramer J, Nurnberger JI, Kendler KS, Webb BT, Miles MF. Multi-species data integration and gene ranking enrich significant results in an alcoholism genome-wide association study. BMC Genomics 2012; 13 Suppl 8:S16. [PMID: 23282140 PMCID: PMC3535715 DOI: 10.1186/1471-2164-13-s8-s16] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND A variety of species and experimental designs have been used to study genetic influences on alcohol dependence, ethanol response, and related traits. Integration of these heterogeneous data can be used to produce a ranked target gene list for additional investigation. RESULTS In this study, we performed a unique multi-species evidence-based data integration using three microarray experiments in mice or humans that generated an initial alcohol dependence (AD) related genes list, human linkage and association results, and gene sets implicated in C. elegans and Drosophila. We then used permutation and false discovery rate (FDR) analyses on the genome-wide association studies (GWAS) dataset from the Collaborative Study on the Genetics of Alcoholism (COGA) to evaluate the ranking results and weighting matrices. We found one weighting score matrix could increase FDR based q-values for a list of 47 genes with a score greater than 2. Our follow up functional enrichment tests revealed these genes were primarily involved in brain responses to ethanol and neural adaptations occurring with alcoholism. CONCLUSIONS These results, along with our experimental validation of specific genes in mice, C. elegans and Drosophila, suggest that a cross-species evidence-based approach is useful to identify candidate genes contributing to alcoholism.
Collapse
Affiliation(s)
- Zhongming Zhao
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Björk K, Hansson AC, Sommer WH. Genetic Variation and Brain Gene Expression in Rodent Models of Alcoholism. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2010; 91:129-71. [DOI: 10.1016/s0074-7742(10)91005-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
26
|
Milner LC, Buck KJ. Identifying quantitative trait loci (QTLs) and genes (QTGs) for alcohol-related phenotypes in mice. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2010; 91:173-204. [PMID: 20813243 DOI: 10.1016/s0074-7742(10)91006-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Alcoholism is a complex clinical disorder with genetic and environmental contributions. Although no animal model duplicates alcoholism, models for specific factors, such as the withdrawal syndrome, are useful to identify potential genetic determinants of liability in humans. Murine models have been invaluable to identify quantitative trait loci (QTLs) that influence a variety of alcohol responses. However, the QTL regions are typically large, at least initially, and contain numerous genes, making identification of the causal quantitative trait gene(s) (QTGs) challenging. Here, we present QTG identification strategies currently used in the field of alcohol genetics and discuss relevance to alcoholic human populations.
Collapse
Affiliation(s)
- Lauren C Milner
- Department of Behavioral Neuroscience, VA Medical Center and Oregon Health & Science University, Portland, OR 97239, USA
| | | |
Collapse
|