1
|
Mignogna KM, Tatom Z, Macleod L, Sergi Z, Nguyen A, Michenkova M, Smith ML, Miles MF. Identification of novel genetic loci and candidate genes for progressive ethanol consumption in diversity outbred mice. Neuropsychopharmacology 2024; 49:1892-1904. [PMID: 38951586 PMCID: PMC11473901 DOI: 10.1038/s41386-024-01902-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 04/26/2024] [Accepted: 06/05/2024] [Indexed: 07/03/2024]
Abstract
Mouse behavioral genetic mapping studies can identify genomic intervals modulating complex traits under well-controlled environmental conditions and have been used to study ethanol behaviors to aid in understanding genetic risk and the neurobiology of alcohol use disorder (AUD). However, historically such studies have produced large confidence intervals, thus complicating identification of potential causal candidate genes. Diversity Outbred (DO) mice offer the ability to perform high-resolution quantitative trait loci (QTL) mapping on a very genetically diverse background, thus facilitating identification of candidate genes. Here, we studied a population of 636 male DO mice with four weeks of intermittent ethanol access via a three-bottle choice procedure, producing a progressive ethanol consumption phenotype. QTL analysis identified 3 significant (Chrs 3, 4, and 12) and 13 suggestive loci for ethanol-drinking behaviors with narrow confidence intervals (1-4 Mbp for significant QTLs). Results suggested that genetic influences on initial versus progressive ethanol consumption were localized to different genomic intervals. A defined set of positional candidate genes were prioritized using haplotype analysis, identified coding polymorphisms, prefrontal cortex transcriptomics data, human GWAS data and prior rodent gene set data for ethanol or other misused substances. These candidates included Car8, the lone gene with a significant cis-eQTL within a Chr 4 QTL for week four ethanol consumption. These results represent the highest-resolution genetic mapping of ethanol consumption behaviors in mice to date, providing identification of novel loci and candidate genes for study in relation to the neurobiology of AUD.
Collapse
Affiliation(s)
- Kristin M Mignogna
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA
| | - Zachary Tatom
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA
| | - Lorna Macleod
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| | - Zachary Sergi
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| | - Angel Nguyen
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| | - Marie Michenkova
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| | - Maren L Smith
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| | - Michael F Miles
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA.
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA.
- Department of Neurology, Virginia Commonwealth University, Richmond, VA, USA.
- VCU Alcohol Research Center, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
2
|
Veerappa A, Guda C. Coordination among frequent genetic variants imparts substance use susceptibility and pathogenesis. Front Neurosci 2024; 18:1332419. [PMID: 38660223 PMCID: PMC11041639 DOI: 10.3389/fnins.2024.1332419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 04/02/2024] [Indexed: 04/26/2024] Open
Abstract
Determining the key genetic variants is a crucial step to comprehensively understand substance use disorders (SUDs). In this study, utilizing whole exome sequences of five multi-generational pedigrees with SUDs, we used an integrative omics-based approach to uncover candidate genetic variants that impart susceptibility to SUDs and influence addition traits. We identified several SNPs and rare, protein-function altering variants in genes, GRIA3, NCOR1, and SHANK1; compound heterozygous variants in LNPEP, LRP1, and TBX2, that play a significant role in the neurotransmitter-neuropeptide axis, specifically in the dopaminergic circuits. We also noted a greater frequency of heterozygous and recessive variants in genes involved in the structural and functional integrity of synapse receptors, CHRNA4, CNR2, GABBR1, DRD4, NPAS4, ADH1B, ADH1C, OPRM1, and GABBR2. Variant analysis in upstream promoter regions revealed regulatory variants in NEK9, PRRX1, PRPF4B, CELA2A, RABGEF1, and CRBN, crucial for dopamine regulation. Using family-and pedigree-based data, we identified heterozygous recessive alleles in LNPEP, LRP1 (4 frameshift deletions), and TBX2 (2 frameshift deletions) linked to SUDs. GWAS overlap identified several SNPs associated with SUD susceptibility, including rs324420 and rs1229984. Furthermore, miRNA variant analysis revealed notable variants in mir-548 U and mir-532. Pathway studies identified the presence of extensive coordination among these genetic variants to impart substance use susceptibility and pathogenesis. This study identified variants that were found to be overrepresented among genes of dopaminergic circuits participating in the neurotransmitter-neuropeptide axis, suggesting pleiotropic influences in the development and sustenance of chronic substance use. The presence of a diverse set of haploinsufficient variants in varying frequencies demonstrates the existence of extraordinary coordination among them in attributing risk and modulating severity to SUDs.
Collapse
Affiliation(s)
- Avinash Veerappa
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, United States
| | - Chittibabu Guda
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, United States
- Center for Biomedical Informatics Research and Innovation, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
3
|
Anjos PAR, Marchette RCN, Kremer R, Granzotto N, Alves TM, Fadanni GP, Mazur FG, Anton EL, da Silva-Santos JE, Linder ÁE, Izídio GS. The influence of chromosome 4 on high ethanol consumption and blood pressure. Alcohol 2022; 102:1-10. [PMID: 35500756 DOI: 10.1016/j.alcohol.2022.04.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 04/06/2022] [Accepted: 04/18/2022] [Indexed: 11/16/2022]
Abstract
The Spontaneously Hypertensive Rats (SHR) strain was developed through selective breeding for high systolic blood pressure. In our laboratory, we established a congenic rat strain named SHR.Lewis-Anxrr16 (SLA16). The SLA16 rat strain is genetically identical to the SHR except for the inserted Anxrr16 region in chromosome 4. Our objective was to evaluate the influence of this genomic region on ethanol consumption and blood pressure. First, we exposed SHR and SLA16 male and female rats to ethanol consumption. Results showed that, regardless of strain, females consumed more ethanol than males during forced (10% v/v) and spontaneous ethanol consumption (SEC; 2.5-20% v/v). Then, females from both strains were used to evaluate sensitivity to ethanol. No strain differences in the loss of righting reflex were observed after ethanol treatment (3 g/kg, 20% w/v, intraperitoneal [i.p.]). But, in the triple test, female SHR rats presented lower sensitivity to the ethanol (1.2 g/kg, 14% w/v, i.p.). Surprisingly, female SHR rats also presented higher blood pressure after SEC (10% v/v). Finally, losartan treatment was effective in decreasing the blood pressure of female rats of both strains, but had specific effects on SHR ethanol consumption. Our data suggest that SLA16 female rats consume less ethanol (10%), are more sensitive to its effects, and present lower blood pressure than SHR female rats. We demonstrated that the Anxrr16 locus in chromosome 4 is a genetic candidate to explain high ethanol consumption and blood pressure, at least in females.
Collapse
Affiliation(s)
| | - Renata Cristina Nunes Marchette
- Department of Pharmacology - Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil; Neurobiology of Addiction Section, Integrative Neuroscience Branch, National Institute on Drug Abuse Intramural Program, Baltimore, MD, United States
| | - Rafael Kremer
- Department of Cellular Biology, Embryology, and Genetics - Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil; Department of Medicine - Federal University of Fronteira Sul, Passo Fundo, Rio Grande do Sul, Brazil
| | - Natalli Granzotto
- Department of Pharmacology - Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Thalita Mello Alves
- Department of Cellular Biology, Embryology, and Genetics - Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Guilherme Pasetto Fadanni
- Department of Pharmacology - Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil; Department of Cellular Biology, Embryology, and Genetics - Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Fernando Gabriel Mazur
- Department of Cellular Biology, Embryology, and Genetics - Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Elaine Leocádia Anton
- Department of Pharmacology - Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | | | - Áurea Elizabeth Linder
- Department of Pharmacology - Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Geison Souza Izídio
- Department of Pharmacology - Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil; Department of Cellular Biology, Embryology, and Genetics - Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil.
| |
Collapse
|
4
|
Chen K, Palagashvili T, Hsu W, Chen Y, Tabakoff B, Hong F, Shih AT, Shih JC. Brain injury and inflammation genes common to a number of neurological diseases and the genes involved in the genesis of GABAnergic neurons are altered in monoamine oxidase B knockout mice. Brain Res 2022; 1774:147724. [PMID: 34780749 PMCID: PMC8638699 DOI: 10.1016/j.brainres.2021.147724] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 11/03/2021] [Accepted: 11/08/2021] [Indexed: 01/03/2023]
Abstract
Monoamine oxidase B (MAO B) oxidizes trace amine phenylethylamine (PEA), and neurotransmitters serotonin and dopamine in the brain. We reported previously that PEA levels increased significantly in all brain regions, but serotonin and dopamine levels were unchanged in MAO B knockout (KO) mice. PEA and dopamine are both synthesized from phenylalanine by aromatic L-amino acid decarboxylase in dopaminergic neurons in the striatum. A high concentration of PEA in the striatum may cause dopaminergic neuronal death in the absence of MAO B. We isolated the RNA from brain tissue of MAO B KO mice (2-month old) and age-matched wild type (WT) male mice and analyzed the altered genes by Affymetrix microarray. Differentially expressed genes (DEGs) in MAO B KO compared to WT mice were analyzed by Partek Genomics Suite, followed by Ingenuity Pathway Analysis (IPA) to assess their functional relationships. DEGs in MAO B KO mice are involved in brain inflammation and the genesis of GABAnergic neurons. The significant DEGs include four brain injury or inflammation genes (upregulated: Ido1, TSPO, AVP, Tdo2), five gamma-aminobutyric acid (GABA) receptors (down-regulated: GABRA2, GABRA3, GABRB1, GABRB3, GABRG3), five transcription factors related to adult neurogenesis (upregulated: Wnt7b, Hes5; down-regulated: Pax6, Tcf4, Dtna). Altered brain injury and inflammation genes in MAO B knockout mice are involved in various neurological disorders: attention deficit hyperactive disorder, panic disorder, obsessive compulsive disorder, autism, amyotrophic lateral sclerosis, Parkinson's diseases, Alzheimer's disease, bipolar affective disorder. Many were commonly involved in these disorders, indicating that there are overlapping molecular pathways.
Collapse
Affiliation(s)
- Kevin Chen
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, 1985 Zonal Ave., Los Angeles, CA, USA
| | - Tamara Palagashvili
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, 1985 Zonal Ave., Los Angeles, CA, USA
| | - W Hsu
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, 1985 Zonal Ave., Los Angeles, CA, USA
| | - Yibu Chen
- Norris Medical Library, University of Southern California, Los Angeles, CA, USA
| | - Boris Tabakoff
- University of Colorado Health Science Center, Denver, CO, USA
| | - Frank Hong
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, 1985 Zonal Ave., Los Angeles, CA, USA
| | - Abigail T Shih
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, 1985 Zonal Ave., Los Angeles, CA, USA
| | - Jean C Shih
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, 1985 Zonal Ave., Los Angeles, CA, USA; Department of Integrative Anatomical Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; USC-Taiwan Center for Translational Research, University of Southern California, Los Angeles CA, USA.
| |
Collapse
|
5
|
Walter N, Cervera-Juanes R, Zheng C, Darakjian P, Lockwood D, Cuzon-Carlson V, Ray K, Fei S, Conrad D, Searles R, Grant K, Hitzemann R. Effect of chronic ethanol consumption in rhesus macaques on the nucleus accumbens core transcriptome. Addict Biol 2021; 26:e13021. [PMID: 33942443 PMCID: PMC8588809 DOI: 10.1111/adb.13021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 01/15/2021] [Accepted: 01/19/2021] [Indexed: 12/13/2022]
Abstract
The nucleus accumbens core (NAcc) has been repeatedly demonstrated to be a key component of the circuitry associated with excessive ethanol consumption. Previous studies have illustrated that in a nonhuman primate (NHP) model of chronic ethanol consumption, there is significant epigenetic remodeling of the NAcc. In the current study, RNA-Seq was used to examine genome-wide gene expression in eight each of control, low/binge (LD*), and high/very high (HD*) rhesus macaque drinkers. Using an FDR < 0.05, zero genes were significantly differentially expressed (DE) between LD* and controls, six genes between HD* and LD*, and 734 genes between HD* and controls. Focusing on HD* versus control DE genes, the upregulated genes (N = 366) were enriched in genes with annotations associated with signal recognition particle (SRP)-dependent co-translational protein targeting to membrane (FDR < 3 × 10-59 ), structural constituent of ribosome (FDR < 3 × 10-47 ), and ribosomal subunit (FDR < 5 × 10-48 ). Downregulated genes (N = 363) were enriched in annotations associated with behavior (FDR < 2 × 10-4 ), membrane organization (FDR < 1 × 10-4 ), inorganic cation transmembrane transporter activity (FDR < 2 × 10-3 ), synapse part (FDR < 4 × 10-10 ), glutamatergic synapse (FDR < 1 × 10-6 ), and GABAergic synapse (FDR < 6 × 10-4 ). Ingenuity Pathway Analysis (IPA) revealed that EIF2 signaling and mTOR pathways were significantly upregulated in HD* animals (FDR < 3 × 10-33 and <2 × 10-16 , respectively). Overall, the data supported our working hypothesis; excessive consumption would be associated with transcriptional differences in GABA/glutamate-related genes.
Collapse
Affiliation(s)
- Nicole Walter
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Portland, Oregon, USA
| | - Rita Cervera-Juanes
- Division of Genetics, Oregon National Primate Research Center, Oregon Health & Science University, Portland, Oregon, USA
| | - Christina Zheng
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Priscila Darakjian
- Division of Genetics, Oregon National Primate Research Center, Oregon Health & Science University, Portland, Oregon, USA
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon, USA
| | - Denesa Lockwood
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon, USA
| | - Verginia Cuzon-Carlson
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Portland, Oregon, USA
| | - Karina Ray
- Division of Genetics, Oregon National Primate Research Center, Oregon Health & Science University, Portland, Oregon, USA
| | - Suzanne Fei
- Division of Genetics, Oregon National Primate Research Center, Oregon Health & Science University, Portland, Oregon, USA
| | - Don Conrad
- Division of Genetics, Oregon National Primate Research Center, Oregon Health & Science University, Portland, Oregon, USA
| | - Robert Searles
- Integrated Genomics Laboratory, Oregon Health & Science University, Portland, Oregon, USA
| | - Kathleen Grant
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Portland, Oregon, USA
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon, USA
| | - Robert Hitzemann
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
6
|
Van der Veen DR, Laing EE, Bae SE, Johnston JD, Dijk DJ, Archer SN. A Topological Cluster of Differentially Regulated Genes in Mice Lacking PER3. Front Mol Neurosci 2020; 13:15. [PMID: 32116548 PMCID: PMC7031657 DOI: 10.3389/fnmol.2020.00015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 01/20/2020] [Indexed: 11/13/2022] Open
Abstract
Polymorphisms in the human circadian clock gene PERIOD3 (PER3) are associated with a wide variety of phenotypes such as diurnal preference, delayed sleep phase disorder, sleep homeostasis, cognitive performance, bipolar disorder, type 2 diabetes, cardiac regulation, cancer, light sensitivity, hormone and cytokine secretion, and addiction. However, the molecular mechanisms underlying these phenotypic associations remain unknown. Per3 knockout mice (Per3-/- ) have phenotypes related to activity, sleep homeostasis, anhedonia, metabolism, and behavioral responses to light. Using a protocol that induces behavioral differences in response to light in wild type and Per3-/- mice, we compared genome-wide expression in the eye and hypothalamus in the two genotypes. Differentially expressed transcripts were related to inflammation, taste, olfactory and melatonin receptors, lipid metabolism, cell cycle, ubiquitination, and hormones, as well as receptors and channels related to sleep regulation. Differentially expressed transcripts in both tissues co-localized with Per3 on an ∼8Mbp region of distal chromosome 4. The most down-regulated transcript is Prdm16, which is involved in adipocyte differentiation and may mediate altered body mass accumulation in Per3-/- mice. eQTL analysis with BXD mouse strains showed that the expression of some of these transcripts and also others co-localized at distal chromosome 4, is correlated with brain tissue expression levels of Per3 with a highly significant linkage to genetic variation in that region. These data identify a cluster of transcripts on mouse distal chromosome 4 that are co-regulated with Per3 and whose expression levels correlate with those of Per3. This locus lies within a topologically associating domain island that contains many genes with functional links to several of the diverse non-circadian phenotypes associated with polymorphisms in human PER3.
Collapse
Affiliation(s)
- Daan R Van der Veen
- School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | - Emma E Laing
- School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | - Sung-Eun Bae
- School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | - Jonathan D Johnston
- School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | - Derk-Jan Dijk
- School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom.,UK Dementia Research Institute, London, United Kingdom
| | - Simon N Archer
- School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| |
Collapse
|
7
|
Tankyrase inhibition preserves osteoarthritic cartilage by coordinating cartilage matrix anabolism via effects on SOX9 PARylation. Nat Commun 2019; 10:4898. [PMID: 31653858 PMCID: PMC6814715 DOI: 10.1038/s41467-019-12910-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Accepted: 10/07/2019] [Indexed: 01/31/2023] Open
Abstract
Osteoarthritis (OA) is a prevalent degenerative disease, which involves progressive and irreversible destruction of cartilage matrix. Despite efforts to reconstruct cartilage matrix in osteoarthritic joints, it has been a difficult task as adult cartilage exhibits marginal repair capacity. Here we report the identification of tankyrase as a regulator of the cartilage anabolism axis based on systems-level factor analysis of mouse reference populations. Tankyrase inhibition drives the expression of a cartilage-signature matrisome and elicits a transcriptomic pattern that is inversely correlated with OA progression. Furthermore, tankyrase inhibitors ameliorate surgically induced OA in mice, and stem cell transplantation coupled with tankyrase knockdown results in superior regeneration of cartilage lesions. Mechanistically, the pro-regenerative features of tankyrase inhibition are mainly triggered by uncoupling SOX9 from a poly(ADP-ribosyl)ation (PARylation)-dependent protein degradation pathway. Our findings provide insights into the development of future OA therapies aimed at reconstruction of articular cartilage. Osteoarthritis results from the progressive destruction of cartilage matrix. Here, Kim et al. identify tankyrase as a regulator of cartilage matrix anabolism, and find that tankyrase inhibition, by preventing SOX9 PARylation, protects from cartilage destruction in a mouse model of osteoarthritis.
Collapse
|
8
|
Bogenpohl JW, Smith ML, Farris SP, Dumur CI, Lopez MF, Becker HC, Grant KA, Miles MF. Cross-Species Co-analysis of Prefrontal Cortex Chronic Ethanol Transcriptome Responses in Mice and Monkeys. Front Mol Neurosci 2019; 12:197. [PMID: 31456662 PMCID: PMC6701453 DOI: 10.3389/fnmol.2019.00197] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 07/30/2019] [Indexed: 12/20/2022] Open
Abstract
Despite recent extensive genomic and genetic studies on behavioral responses to ethanol, relatively few new therapeutic targets for the treatment of alcohol use disorder have been validated. Here, we describe a cross-species genomic approach focused on identifying gene networks associated with chronic ethanol consumption. To identify brain mechanisms underlying a chronic ethanol consumption phenotype highly relevant to human alcohol use disorder, and to elucidate potential future therapeutic targets, we conducted a genomic study in a non-human primate model of chronic open-access ethanol consumption. Microarray analysis of RNA expression in anterior cingulate and subgenual cortices from rhesus macaques was performed across multiple cohorts of animals. Gene networks correlating with ethanol consumption or showing enrichment for ethanol-regulated genes were identified, as were major ethanol-related hub genes within these networks. A subsequent consensus module analysis was used to co-analyze monkey data with expression data from a chronic intermittent ethanol vapor-exposure and consumption model in C57BL/6J mice. Ethanol-related gene networks conserved between primates and rodents were enriched for genes involved in discrete biological functions, including; myelination, synaptic transmission, chromatin modification, Golgi apparatus function, translation, cellular respiration, and RNA processing. The myelin-related network, in particular, showed strong correlations with ethanol consumption behavior and displayed marked network reorganization between control and ethanol-drinking animals. Further bioinformatics analysis revealed that these networks also showed highly significant overlap with other ethanol-regulated gene sets. Altogether, these studies provide robust primate and rodent cross-species validation of gene networks associated with chronic ethanol consumption. Our results also suggest potential novel focal points for future therapeutic interventions in alcohol use disorder.
Collapse
Affiliation(s)
- James W Bogenpohl
- Department of Molecular Biology and Chemistry, Christopher Newport University, Newport News, VA, United States
| | - Maren L Smith
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, United States
| | - Sean P Farris
- Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, TX, United States
| | - Catherine I Dumur
- Aurora Diagnostics-Sonic Healthcare, Bernhardt Laboratories, Jacksonville, FL, United States
| | - Marcelo F Lopez
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, United States
| | - Howard C Becker
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, United States
| | - Kathleen A Grant
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR, United States.,Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, United States
| | - Michael F Miles
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, United States.,Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, United States.,Department of Neurology, Virginia Commonwealth University, Richmond, VA, United States.,VCU Alcohol Research Center, Virginia Commonwealth University, Richmond, VA, United States
| |
Collapse
|
9
|
Zhao Y, Ge Y, Zheng ZL. Brain Imaging-Guided Analysis Reveals DNA Methylation Profiles Correlated with Insular Surface Area and Alcohol Use Disorder. Alcohol Clin Exp Res 2019; 43:628-639. [PMID: 30830696 PMCID: PMC6443499 DOI: 10.1111/acer.13971] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 01/26/2019] [Indexed: 12/26/2022]
Abstract
BACKGROUND Alcohol use disorder (AUD) is a wide-spread, heritable brain disease, but few studies have linked genetic variants or epigenetic factors to brain structures related to AUD in humans, due to many factors including the high-dimensional nature of imaging and genomic data. METHODS To provide potential insights into the links among epigenetic regulation, brain structure, and AUD, we have performed an integrative analysis of brain structural imaging and blood DNA methylome data from 52 AUD and 58 healthy control (HC) subjects collected in the Nathan Kline Institute-Rockland Sample. RESULTS We first found that AUD subjects had significantly larger insular surface area than HC in both left and right hemispheres. We then found that 7,827 DNA methylation probes on the HumanMethylation450K BeadChip had significant correlations with the right insular surface area (false discovery rate [FDR] < 0.05). Furthermore, we showed that 44 of the insular surface area-correlated methylation probes were also strongly correlated with AUD status (FDR < 0.05). These AUD-correlated probes are annotated to 36 protein-coding genes, with 16 genes (44%) having been reported by others to be related to AUD or alcohol response, including TAS2R16 and PER2. The remaining 20 genes, in particular ARHGAP22, might represent novel genes involved in AUD or responsive to alcohol. CONCLUSIONS We have identified 36 insular surface area- and AUD-correlated protein-coding genes that are either known to be AUD- or alcohol-related or not yet reported by prior studies. Therefore, our study suggests that the brain imaging-guided epigenetic analysis has a potential of identifying possible epigenetic mechanisms involved in AUD.
Collapse
Affiliation(s)
- Yihong Zhao
- Department of Child and Adolescent Psychiatry, NYU Langone Medical Center, New York, NY 10016, USA
- Center for Behavioral Science Research, Department of Health Policy & Health Services Research, Boston University, Boston, MA 02118, USA
| | - Yongchao Ge
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Zhi-Liang Zheng
- Department of Biological Sciences, Lehman College, City University of New York, Bronx, NY 10468, USA
| |
Collapse
|
10
|
Iancu OD, Colville A, Walter NA, Darakjian P, Oberbeck DL, Daunais JB, Zheng CL, Searles RP, McWeeney SK, Grant KA, Hitzemann R. On the relationships in rhesus macaques between chronic ethanol consumption and the brain transcriptome. Addict Biol 2018; 23:196-205. [PMID: 28247455 DOI: 10.1111/adb.12501] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 02/01/2017] [Accepted: 02/03/2017] [Indexed: 12/19/2022]
Abstract
This is the first description of the relationship between chronic ethanol self-administration and the brain transcriptome in a non-human primate (rhesus macaque). Thirty-one male animals self-administered ethanol on a daily basis for over 12 months. Gene transcription was quantified with RNA-Seq in the central nucleus of the amygdala (CeA) and cortical Area 32. We constructed coexpression and cosplicing networks, and we identified areas of preservation and areas of differentiation between regions and network types. Correlations between intake and transcription included largely distinct gene sets and annotation categories across brain regions and between expression and splicing; positive and negative correlations were also associated with distinct annotation groups. Membrane, synaptic and splicing annotation categories were over-represented in the modules (gene clusters) enriched in positive correlations (CeA); our cosplicing analysis further identified the genes affected only at the exon inclusion level. In the CeA coexpression network, we identified Rab6b, Cdk18 and Igsf21 among the intake-correlated hubs, while in the Area 32, we identified a distinct hub set that included Ppp3r1 and Myeov2. Overall, the data illustrate that excessive ethanol self-administration is associated with broad expression and splicing mechanisms that involve membrane and synapse genes.
Collapse
|
11
|
Chen K, Kardys A, Chen Y, Flink S, Tabakoff B, Shih JC. Altered gene expression in early postnatal monoamine oxidase A knockout mice. Brain Res 2017; 1669:18-26. [PMID: 28535982 PMCID: PMC5531263 DOI: 10.1016/j.brainres.2017.05.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 04/10/2017] [Accepted: 05/16/2017] [Indexed: 11/19/2022]
Abstract
We reported previously that monoamine oxidase (MAO) A knockout (KO) mice show increased serotonin (5-hydroxytryptamine, 5-HT) levels and autistic-like behaviors characterized by repetitive behaviors, and anti-social behaviors. We showed that administration of the serotonin synthesis inhibitor para-chlorophenylalanine (pCPA) from post-natal day 1 (P1) through 7 (P7) in MAO A KO mice reduced the serotonin level to normal and reverses the repetitive behavior. These results suggested that the altered gene expression at P1 and P7 may be important for the autistic-like behaviors seen in MAO A KO mice and was studied here. In this study, Affymetrix mRNA array data for P1 and P7 MAO A KO mice were analyzed using Partek Genomics Suite and Ingenuity Pathways Analysis to identify genes differentially expressed versus wild-type and assess their functions and relationships. The number of significant differentially expressed genes (DEGs) varied with age: P1 (664) and P7 (3307) [false discovery rate (FDR) <0.05, fold-change (FC) >1.5 for autism-linked genes and >2.0 for functionally categorized genes]. Eight autism-linked genes were differentially expressed in P1 (upregulated: NLGN3, SLC6A2; down-regulated: HTR2C, MET, ADSL, MECP2, ALDH5A1, GRIN3B) while four autism-linked genes were differentially expressed at P7 (upregulated: HTR2B; downregulated: GRIN2D, GRIN2B, CHRNA4). Many other genes involved in neurodevelopment, apoptosis, neurotransmission, and cognitive function were differentially expressed at P7 in MAO A KO mice. This result suggests that modulation of these genes by the increased serotonin may lead to neurodevelopmental alteration in MAO A KO mice and results in autistic-like behaviors.
Collapse
Affiliation(s)
- Kevin Chen
- Dept. of Pharmacology & Pharmaceutical Science, School of Pharmacy, Los Angeles, CA 90089, United States
| | - Abbey Kardys
- Dept. of Pharmacology & Pharmaceutical Science, School of Pharmacy, Los Angeles, CA 90089, United States
| | - Yibu Chen
- Norris Medical Library, University of Southern California, Los Angeles, CA 90089, United States
| | - Stephen Flink
- University of Colorado Health Science Center, Denver, CO 80262, United States
| | - Boris Tabakoff
- University of Colorado Health Science Center, Denver, CO 80262, United States
| | - Jean C Shih
- Dept. of Pharmacology & Pharmaceutical Science, School of Pharmacy, Los Angeles, CA 90089, United States; USC-Taiwan Center for Translational Research, University of Southern California, Los Angeles, CA 90089, United States; Dept. of Cell & Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, United States.
| |
Collapse
|
12
|
A role for the peripheral immune system in the development of alcohol use disorders? Neuropharmacology 2017; 122:148-160. [DOI: 10.1016/j.neuropharm.2017.04.013] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 04/06/2017] [Accepted: 04/07/2017] [Indexed: 02/07/2023]
|
13
|
Wang L, Jiao Y, Sun S, Jarrett HW, Sun D, Gu W. Gene network of a phosphoglycerate mutase in muscle wasting in mice. Cell Biol Int 2015; 39:666-77. [PMID: 25644094 DOI: 10.1002/cbin.10437] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 01/09/2015] [Indexed: 12/29/2022]
Abstract
We previously identified the insertion of an intracisternal A-particle retrotransposons (IAPs) sequence in a gene, 9630033F20Rik, that contains domains involved in glycolysis from a mouse model called lethal wasting (lew). However, because both IAP insertion and the muation of vesicle-associated membrane protein 1 (VAMP1) were discovered from lew, the impact of the IAP insertion and Vamp1 on the lew mouse phenotype needs further investigation. In this study, the effect of the 9630033F20Rik and Vamp1 on glycolysis and muscle-wasting genes in heart, muscle, and brain tissues was further investigated using data of gene expression profiles in these tissues. Our data indicated that the expression levels of 9630033F20Rik and Vamp1 are not associated with each other. While 9630033F20Rik affects the expression of several key genes in pathways of glycolysis and muscle wasting, Vamp1 affects a different set of genes, with fewer numbers. In situ hybridization indicated that the expression of 9630033F20Rik is different in musculoskeletal tissues between the muscle-wasting mouse model and the wild-type model. Our data indicated that 9630033F20Rik may play an important role in muscle wasting and that it has a distinguished characterization of gene network. Our data also suggest that both 9630033F20Rik and Vamp1 play functional roles in muscle development and lead to the muscle-wasting phenotype when they are mutated.
Collapse
Affiliation(s)
- Lishi Wang
- Department of Orthopedic Surgery and BME, Campbell-Clinic, University of Tennessee Health Science Center, Memphis, TN, 38163, USA.,Department of Basic Medical Research, Inner Mongolia Medical University, Inner Mongolia, 010110, PR China
| | - Yan Jiao
- Department of Orthopedic Surgery and BME, Campbell-Clinic, University of Tennessee Health Science Center, Memphis, TN, 38163, USA.,Mudanjiang Medical College, Mudanjiang, 157011, PR China
| | - Shuqiu Sun
- National Center for Endemic Disease Control, Harbin Medical University, Harbin, 150081, PR China
| | - Harry W Jarrett
- Department of Chemistry, University of Texas at San Antonio, San Antonio, TX, 78249, USA
| | - Dianjun Sun
- National Center for Endemic Disease Control, Harbin Medical University, Harbin, 150081, PR China
| | - Weikuan Gu
- Department of Orthopedic Surgery and BME, Campbell-Clinic, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| |
Collapse
|
14
|
Gene expression profile analysis of rat cerebellum under acute alcohol intoxication. Gene 2014; 557:188-94. [PMID: 25527120 DOI: 10.1016/j.gene.2014.12.032] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Revised: 11/20/2014] [Accepted: 12/14/2014] [Indexed: 11/22/2022]
Abstract
Acute alcohol intoxication, a common disease causing damage to the central nervous system (CNS) has been primarily studied on the aspects of alcohol addiction and chronic alcohol exposure. The understanding of gene expression change in the CNS during acute alcohol intoxication is still lacking. We established a model for acute alcohol intoxication in SD rats by oral gavage. A rat cDNA microarray was used to profile mRNA expression in the cerebella of alcohol-intoxicated rats (experimental group) and saline-treated rats (control group). A total of 251 differentially expressed genes were identified in response to acute alcohol intoxication, in which 208 of them were up-regulated and 43 were down-regulated. Gene ontology (GO) term enrichment analysis and pathway analysis revealed that the genes involved in the biological processes of immune response and endothelial integrity are among the most severely affected in response to acute alcohol intoxication. We discovered five transcription factors whose consensus binding motifs are overrepresented in the promoter region of differentially expressed genes. Additionally, we identified 20 highly connected hub genes by co-expression analysis, and validated the differential expression of these genes by real-time quantitative PCR. By determining novel biological pathways and transcription factors that have functional implication to acute alcohol intoxication, our study substantially contributes to the understanding of the molecular mechanism underlying the pathology of acute alcoholism.
Collapse
|
15
|
Hoffman PL, Saba LM, Flink S, Grahame NJ, Kechris K, Tabakoff B. Genetics of gene expression characterizes response to selective breeding for alcohol preference. GENES, BRAIN, AND BEHAVIOR 2014; 13:743-57. [PMID: 25160899 PMCID: PMC4241152 DOI: 10.1111/gbb.12175] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Revised: 08/18/2014] [Accepted: 08/24/2014] [Indexed: 01/30/2023]
Abstract
Numerous selective breeding experiments have been performed with rodents, in an attempt to understand the genetic basis for innate differences in preference for alcohol consumption. Quantitative trait locus (QTL) analysis has been used to determine regions of the genome that are associated with the behavioral difference in alcohol preference/consumption. Recent work suggests that differences in gene expression represent a major genetic basis for complex traits. Therefore, the QTLs are likely to harbor regulatory regions (eQTLs) for the differentially expressed genes that are associated with the trait. In this study, we examined brain gene expression differences over generations of selection of the third replicate lines of high and low alcohol-preferring (HAP3 and LAP3) mice, and determined regions of the genome that control the expression of these differentially expressed genes (de eQTLs). We also determined eQTL regions (rv eQTLs) for genes that showed a decrease in variance of expression levels over the course of selection. We postulated that de eQTLs that overlap with rv eQTLs, and also with phenotypic QTLs, represent genomic regions that are affected by the process of selection. These overlapping regions controlled the expression of candidate genes (that displayed differential expression and reduced variance of expression) for the predisposition to differences in alcohol consumption by the HAP3/LAP3 mice.
Collapse
Affiliation(s)
- Paula L. Hoffman
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045
| | - Laura M. Saba
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045
| | - Stephen Flink
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045
| | - Nicholas J. Grahame
- Department of Psychology, Indiana University Purdue University, Indianapolis, IN 46202
| | - Katerina Kechris
- Department of Biostatistics and Informatics, University of Colorado School of Public Health, Aurora, CO 80045
| | - Boris Tabakoff
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045
| |
Collapse
|
16
|
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
|
17
|
Abstract
Acute alcohol intoxication causes cellular changes in the brain that last for hours, while chronic alcohol use induces widespread neuroadaptations in the nervous system that can last a lifetime. Chronic alcohol use and the progression into dependence involve the remodeling of synapses caused by changes in gene expression produced by alcohol. The progression of alcohol use, abuse, and dependence can be divided into stages, which include intoxication, withdrawal, and craving. Each stage is associated with specific changes in gene expression, cellular function, brain circuits, and ultimately behavior. What are the molecular mechanisms underlying the transition from recreational use (acute) to dependence (chronic)? What cellular adaptations result in drug memory retention, leading to the persistence of addictive behaviors, even after prolonged drug abstinence? Research into the neurobiology of alcoholism aims to answer these questions. This chapter will describe the molecular adaptations caused by alcohol use and dependence, and will outline key neurochemical participants in alcoholism at the molecular level, which are also potential targets for therapy.
Collapse
Affiliation(s)
- Dana Most
- Waggoner Center for Alcohol and Addiction Research, University of Texas, Austin, TX, USA
| | - Laura Ferguson
- Waggoner Center for Alcohol and Addiction Research, University of Texas, Austin, TX, USA
| | - R Adron Harris
- Waggoner Center for Alcohol and Addiction Research, University of Texas, Austin, TX, USA.
| |
Collapse
|
18
|
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
|
19
|
FADS2 Genetic Variance in Combination with Fatty Acid Intake Might Alter Composition of the Fatty Acids in Brain. PLoS One 2013; 8:e68000. [PMID: 23826354 PMCID: PMC3694926 DOI: 10.1371/journal.pone.0068000] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Accepted: 05/29/2013] [Indexed: 02/01/2023] Open
Abstract
Multiple lines of evidence suggest that fatty acids (FA) play an important role in cognitive function. However, little is known about the functional genetic pathways involved in cognition. The main goals of this study were to replicate previously reported interaction effects between breast feeding (BF) and FA desaturase (FADS) genetic variation on IQ and to investigate the possible mechanisms by which these variants might moderate BF effect, focusing on brain expression. Using a sample of 534 twins, we observed a trend in the moderation of BF effects on IQ by FADS2 variation. In addition, we made use of publicly available gene expression databases from both humans (193) and mice (93) and showed that FADS2 variants also correlate with FADS1 brain expression (P-value<1.1E-03). Our results provide novel clues for the understanding of the genetic mechanisms regulating FA brain expression and improve the current knowledge of the FADS moderation effect on cognition.
Collapse
|
20
|
Logan RW, Robledo RF, Recla JM, Philip VM, Bubier JA, Jay JJ, Harwood C, Wilcox T, Gatti DM, Bult CJ, Churchill GA, Chesler EJ. High-precision genetic mapping of behavioral traits in the diversity outbred mouse population. GENES BRAIN AND BEHAVIOR 2013; 12:424-37. [PMID: 23433259 PMCID: PMC3709837 DOI: 10.1111/gbb.12029] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2012] [Revised: 01/14/2013] [Accepted: 02/17/2013] [Indexed: 12/11/2022]
Abstract
Historically our ability to identify genetic variants underlying complex behavioral traits in mice has been limited by low mapping resolution of conventional mouse crosses. The newly developed Diversity Outbred (DO) population promises to deliver improved resolution that will circumvent costly fine-mapping studies. The DO is derived from the same founder strains as the Collaborative Cross (CC), including three wild-derived strains. Thus the DO provides more allelic diversity and greater potential for discovery compared to crosses involving standard mouse strains. We have characterized 283 male and female DO mice using open-field, light–dark box, tail-suspension and visual-cliff avoidance tests to generate 38 behavioral measures. We identified several quantitative trait loci (QTL) for these traits with support intervals ranging from 1 to 3 Mb in size. These intervals contain relatively few genes (ranging from 5 to 96). For a majority of QTL, using the founder allelic effects together with whole genome sequence data, we could further narrow the positional candidates. Several QTL replicate previously published loci. Novel loci were also identified for anxiety- and activity-related traits. Half of the QTLs are associated with wild-derived alleles, confirming the value to behavioral genetics of added genetic diversity in the DO. In the presence of wild-alleles we sometimes observe behaviors that are qualitatively different from the expected response. Our results demonstrate that high-precision mapping of behavioral traits can be achieved with moderate numbers of DO animals, representing a significant advance in our ability to leverage the mouse as a tool for behavioral genetics
Collapse
Affiliation(s)
- R W Logan
- The Jackson Laboratory, Bar Harbor, ME 04609, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Boughter JD, Mulligan MK, St John SJ, Tokita K, Lu L, Heck DH, Williams RW. Genetic control of a central pattern generator: rhythmic oromotor movement in mice is controlled by a major locus near Atp1a2. PLoS One 2012; 7:e38169. [PMID: 22675444 PMCID: PMC3364982 DOI: 10.1371/journal.pone.0038169] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2012] [Accepted: 05/04/2012] [Indexed: 12/21/2022] Open
Abstract
Fluid licking in mice is a rhythmic behavior that is controlled by a central pattern generator (CPG) located in a complex of brainstem nuclei. C57BL/6J (B6) and DBA/2J (D2) strains differ significantly in water-restricted licking, with a highly heritable difference in rates (h(2)≥0.62) and a corresponding 20% difference in interlick interval (mean ± SEM = 116.3±1 vs 95.4±1.1 ms). We systematically quantified motor output in these strains, their F(1) hybrids, and a set of 64 BXD progeny strains. The mean primary interlick interval (MPI) varied continuously among progeny strains. We detected a significant quantitative trait locus (QTL) for a CPG controlling lick rate on Chr 1 (Lick1), and a suggestive locus on Chr 10 (Lick10). Linkage was verified by testing of B6.D2-1D congenic stock in which a segment of Chr 1 of the D2 strain was introgressed onto the B6 parent. The Lick1 interval on distal Chr 1 contains several strong candidate genes. One of these is a sodium/potassium pump subunit (Atp1a2) with widespread expression in astrocytes, as well as in a restricted population of neurons. Both this subunit and the entire Na(+)/K(+)-ATPase molecule have been implicated in rhythmogenesis for respiration and locomotion. Sequence variants in or near Apt1a2 strongly modulate expression of the cognate mRNA in multiple brain regions. This gene region has recently been sequenced exhaustively and we have cataloged over 300 non-coding and synonymous mutations segregating among BXD strains, one or more of which is likely to contribute to differences in central pattern generator tempo.
Collapse
Affiliation(s)
- John D Boughter
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America.
| | | | | | | | | | | | | |
Collapse
|
22
|
Mulligan MK, Wang X, Adler AL, Mozhui K, Lu L, Williams RW. Complex control of GABA(A) receptor subunit mRNA expression: variation, covariation, and genetic regulation. PLoS One 2012; 7:e34586. [PMID: 22506031 PMCID: PMC3323555 DOI: 10.1371/journal.pone.0034586] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2012] [Accepted: 03/06/2012] [Indexed: 12/13/2022] Open
Abstract
GABA type-A receptors are essential for fast inhibitory neurotransmission and are critical in brain function. Surprisingly, expression of receptor subunits is highly variable among individuals, but the cause and impact of this fluctuation remains unknown. We have studied sources of variation for all 19 receptor subunits using massive expression data sets collected across multiple brain regions and platforms in mice and humans. Expression of Gabra1, Gabra2, Gabrb2, Gabrb3, and Gabrg2 is highly variable and heritable among the large cohort of BXD strains derived from crosses of fully sequenced parents—C57BL/6J and DBA/2J. Genetic control of these subunits is complex and highly dependent on tissue and mRNA region. Remarkably, this high variation is generally not linked to phenotypic differences. The single exception is Gabrb3, a locus that is linked to anxiety. We identified upstream genetic loci that influence subunit expression, including three unlinked regions of chromosome 5 that modulate the expression of nine subunits in hippocampus, and that are also associated with multiple phenotypes. Candidate genes within these loci include, Naaa, Nos1, and Zkscan1. We confirmed a high level of coexpression for subunits comprising the major channel—Gabra1, Gabrb2, and Gabrg2—and identified conserved members of this expression network in mice and humans. Gucy1a3, Gucy1b3, and Lis1 are novel and conserved associates of multiple subunits that are involved in inhibitory signaling. Finally, proximal and distal regions of the 3′ UTRs of single subunits have remarkably independent expression patterns in both species. However, corresponding regions of different subunits often show congruent genetic control and coexpression (proximal-to-proximal or distal-to-distal), even in the absence of sequence homology. Our findings identify novel sources of variation that modulate subunit expression and highlight the extraordinary capacity of biological networks to buffer 4–100 fold differences in mRNA levels.
Collapse
Affiliation(s)
- Megan K Mulligan
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America.
| | | | | | | | | | | |
Collapse
|
23
|
Giardino WJ, Cote DM, Li J, Ryabinin AE. Characterization of Genetic Differences within the Centrally Projecting Edinger-Westphal Nucleus of C57BL/6J and DBA/2J Mice by Expression Profiling. Front Neuroanat 2012; 6:22. [PMID: 22347848 PMCID: PMC3278674 DOI: 10.3389/fnana.2012.00005] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2012] [Accepted: 06/05/2012] [Indexed: 11/13/2022] Open
Abstract
Detailed examination of the midbrain Edinger–Westphal (EW) nucleus revealed the existence of two distinct nuclei. One population of EW preganglionic (EWpg) neurons was found to control oculomotor functions, and a separate population of EW centrally projecting (EWcp) neurons was found to contain stress- and feeding-related neuropeptides. Although it has been shown that EWcp neurons are highly responsive to drugs of abuse and behavioral stress, a genetic characterization of the EWcp was needed. To identify genetic differences in the EWcp of inbred mouse strains that differ in behaviors relevant to EWcp function, we used publicly available tools from the Allen Brain Atlas to identify 68 transcripts that were selectively expressed in the EWcp, and examined their expression within tissue punch microdissection samples containing the EWcp of adult male C57BL/6J (B6) and DBA/2J (D2) mice. Using 96-well quantitative real-time PCR (qPCR) arrays that included the EWcp-specific genes, several other genes of interest, and five housekeeping genes, we identified strain differences in expression of 11 EWcp-specific genes (BC023892, Btg3, Bves, Cart, Cck, Ghsr, Neto1, Postn, Ptprn, Rcn1, and Ucn), two immediate early genes (Egr1 and Fos), and one dopamine-related gene (Drd5). All significant expression differences were greater in B6 vs. D2 mice, and several of these were verified either at the protein level using immunohistochemistry (IHC) or in silico using microarray data sets from whole brain and other brain areas. These results demonstrate a significant advance in our understanding of the EWcp on three levels. First, we generated a list of EWcp-specific genes (most of which had not yet been reported within the EWcp in the literature) that will be informative for future studies of EWcp function. Second, due to similarity in results from qPCR and IHC, we revealed that strain differences in basal EWcp neuropeptide content are accounted for by differential transcription and number of peptidergic neurons, rather than by differential rates of peptide release. And third, our identification of differentially expressed EWcp-specific genes between B6 and D2 mice may hold powerful insight into the neurogenetic contributions of the EWcp to stress- and addiction-related behaviors.
Collapse
Affiliation(s)
- William J Giardino
- Department of Behavioral Neuroscience, Oregon Health and Science University Portland, OR, USA
| | | | | | | |
Collapse
|
24
|
Using genome-wide expression profiling to define gene networks relevant to the study of complex traits: from RNA integrity to network topology. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2012. [PMID: 23195313 DOI: 10.1016/b978-0-12-398323-7.00005-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Postgenomic studies of the function of genes and their role in disease have now become an area of intense study since efforts to define the raw sequence material of the genome have largely been completed. The use of whole-genome approaches such as microarray expression profiling and, more recently, RNA-sequence analysis of transcript abundance has allowed an unprecedented look at the workings of the genome. However, the accurate derivation of such high-throughput data and their analysis in terms of biological function has been critical to truly leveraging the postgenomic revolution. This chapter will describe an approach that focuses on the use of gene networks to both organize and interpret genomic expression data. Such networks, derived from statistical analysis of large genomic datasets and the application of multiple bioinformatics data resources, potentially allow the identification of key control elements for networks associated with human disease, and thus may lead to derivation of novel therapeutic approaches. However, as discussed in this chapter, the leveraging of such networks cannot occur without a thorough understanding of the technical and statistical factors influencing the derivation of genomic expression data. Thus, while the catch phrase may be "it's the network … stupid," the understanding of factors extending from RNA isolation to genomic profiling technique, multivariate statistics, and bioinformatics are all critical to defining fully useful gene networks for study of complex biology.
Collapse
|
25
|
Blednov YA, Ponomarev I, Geil C, Bergeson S, Koob GF, Harris RA. Neuroimmune regulation of alcohol consumption: behavioral validation of genes obtained from genomic studies. Addict Biol 2012; 17:108-20. [PMID: 21309947 DOI: 10.1111/j.1369-1600.2010.00284.x] [Citation(s) in RCA: 197] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Analysis of mouse brain gene expression, using strains that differ in alcohol consumption, provided a number of novel candidate genes that potentially regulate alcohol consumption. We selected six genes [beta-2-microglobulin (B2m), cathepsin S (Ctss), cathepsin F (Ctsf), interleukin 1 receptor antagonist (Il1rn), CD14 molecule (Cd14) and interleukin 6 (Il6)] for behavioral validation using null mutant mice. These genes are known to be important for immune responses but were not specifically linked to alcohol consumption by previous research. Null mutant mice were tested for ethanol intake in three tests: 24-hour two-bottle choice, limited access two-bottle choice and limited access to one bottle of ethanol. Ethanol consumption and preference were reduced in all the null mutant mice in the 24-hour two-bottle choice test, the test that was the basis for selection of these genes. No major differences were observed in consumption of saccharin or quinine in the null mutant mice. Deletion of B2m, Ctss, Il1rn, Cd14 and Il6 also reduced ethanol consumption in the limited access two bottle choice test for ethanol intake; with the Il1rn and Ctss null mutants showing reduced intake in all three tests (with some variation between males and females). These results provide the most compelling evidence to date that global gene expression analysis can identify novel genetic determinants of complex behavioral traits. Specifically, they suggest a novel role for neuroimmune signaling in regulation of alcohol consumption.
Collapse
Affiliation(s)
- Yuri A Blednov
- Waggoner Center for Alcohol and Addiction Research, Section of Neurobiology and Department Pharmacology/Toxicology, University of Texas, Austin, USA.
| | | | | | | | | | | |
Collapse
|
26
|
Maurage P, Callot C, Philippot P, Rombaux P, de Timary P. Chemosensory event-related potentials in alcoholism: A specific impairment for olfactory function. Biol Psychol 2011; 88:28-36. [DOI: 10.1016/j.biopsycho.2011.06.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2010] [Revised: 03/05/2011] [Accepted: 06/13/2011] [Indexed: 11/24/2022]
|
27
|
Bennett B, Saba LM, Hornbaker CK, Kechris KJ, Hoffman P, Tabakoff B. Genetical genomic analysis of complex phenotypes using the PhenoGen website. Behav Genet 2011; 41:625-8. [PMID: 21184165 PMCID: PMC3121939 DOI: 10.1007/s10519-010-9427-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2010] [Accepted: 12/03/2010] [Indexed: 11/17/2022]
Abstract
Our laboratory has developed an online interactive resource called PhenoGen ( http://phenogen.ucdenver.edu ) which provides an archive of brain and other organ gene expression data from a panel of 20 common inbred mouse strains, and three recombinant inbred (RI) panels (two mouse and one rat). DNA microarray data can also be uploaded to the site where numerous analytical tools can be implemented. An important advantage to the archived data is that each array represents data from a single animal and each strain was sampled 4-7 times, providing an estimate of genetic variance (heritability) of individual transcript levels. These panels also allow genetic mapping of expression QTLs. Overlap of eQTLs with phenotypic QTLs provides a powerful approach to candidate gene identification. These methods are briefly described here and we encourage the use of our site for both scientific discovery and as a teaching tool in quantitative genetics.
Collapse
Affiliation(s)
- Beth Bennett
- Department of Pharmacology, University of Colorado Denver School of Medicine, Aurora, CO 80045-0511, USA.
| | | | | | | | | | | |
Collapse
|
28
|
Hoffman PL, Bennett B, Saba LM, Bhave SV, Carosone-Link PJ, Hornbaker CK, Kechris KJ, Williams RW, Tabakoff B. Using the Phenogen website for 'in silico' analysis of morphine-induced analgesia: identifying candidate genes. Addict Biol 2011; 16:393-404. [PMID: 21054686 PMCID: PMC3115429 DOI: 10.1111/j.1369-1600.2010.00254.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The identification of genes that contribute to polygenic (complex) behavioral phenotypes is a key goal of current genetic research. One approach to this goal is to combine gene expression information with genetic information, i.e. to map chromosomal regions that regulate gene expression levels. This approach has been termed 'genetical genomics', and, when used in conjunction with the identification of genomic regions (QTLs) that regulate the complex physiological trait under investigation, provides a strong basis for candidate gene discovery. In this paper, we describe the implementation of the genetical genomic/phenotypic approach to identify candidate genes for sensitivity to the analgesic effect of morphine in BXD recombinant inbred mice. Our analysis was performed 'in silico', using an online interactive resource called PhenoGen (http://phenogen.ucdenver.edu). We describe in detail the use of this resource, which identified a set of candidate genes, some of whose products regulate the cellular localization and activity of the mu opiate receptor. The results demonstrate how PhenoGen can be used to identify a novel set of genes that can be further investigated for their potential role in pain, morphine analgesia and/or morphine tolerance.
Collapse
Affiliation(s)
- Paula L. Hoffman
- University of Colorado Denver, School of Medicine, Department of Pharmacology, Aurora CO USA 80045
| | - Beth Bennett
- University of Colorado Denver, School of Medicine, Department of Pharmacology, Aurora CO USA 80045
| | - Laura M. Saba
- University of Colorado Denver, School of Medicine, Department of Pharmacology, Aurora CO USA 80045
| | - Sanjiv V. Bhave
- University of Colorado Denver, School of Medicine, Department of Pharmacology, Aurora CO USA 80045
| | - Phyllis J. Carosone-Link
- University of Colorado Denver, School of Medicine, Department of Pharmacology, Aurora CO USA 80045
| | - Cheryl K. Hornbaker
- University of Colorado Denver, School of Medicine, Department of Pharmacology, Aurora CO USA 80045
| | - Katerina J. Kechris
- University of Colorado Denver, School of Medicine, Department of Pharmacology, Aurora CO USA 80045
| | - Robert W. Williams
- University of Tennessee Health Science Center, Department of Anatomy and Neurobiology, Memphis, TN 38163
| | - Boris Tabakoff
- University of Colorado Denver, School of Medicine, Department of Pharmacology, Aurora CO USA 80045
| |
Collapse
|
29
|
Blednov Y, Benavidez J, Geil C, Perra S, Morikawa H, Harris R. Activation of inflammatory signaling by lipopolysaccharide produces a prolonged increase of voluntary alcohol intake in mice. Brain Behav Immun 2011; 25 Suppl 1:S92-S105. [PMID: 21266194 PMCID: PMC3098320 DOI: 10.1016/j.bbi.2011.01.008] [Citation(s) in RCA: 185] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2010] [Revised: 01/17/2011] [Accepted: 01/17/2011] [Indexed: 02/03/2023] Open
Abstract
Previous studies showed that mice with genetic predisposition for high alcohol consumption as well as human alcoholics show changes in brain expression of genes related to immune signaling. In addition, mutant mice lacking genes related to immune function show decreased alcohol consumption (Blednov et al., 2011), suggesting that immune signaling promotes alcohol consumption. To test the possibility that activation of immune signaling will increase alcohol consumption, we treated mice with lipopolysaccaride (LPS; 1mg/kg, i.p.) and tested alcohol consumption in the continuous two-bottle choice test. To take advantage of the long-lasting activation of brain immune signaling by LPS, we measured drinking beginning one week or one month after LPS treatment and continued the studies for several months. LPS produced persistent increases in alcohol consumption in C57BL/6J (B6) inbred mice, FVBxB6F1 and B6xNZBF1 hybrid mice, but not in FVB inbred mice. To determine if this effect of LPS is mediated through binding to TLR4, we tested mice lacking CD14, a key component of TLR4 signaling. These null mutants showed no increase of alcohol intake after treatment with LPS. LPS treatment decreased ethanol-conditioned taste aversion but did not alter ethanol-conditioned place preference (B6xNZBF1 mice). Electrophysiological studies of dopamine neurons in the ventral tegmental area showed that pretreatment of mice with LPS decreased the neuronal firing rate. These results suggest that activation of immune signaling promotes alcohol consumption and alters certain aspects of alcohol reward/aversion.
Collapse
Affiliation(s)
- Y.A. Blednov
- Corresponding author. Address: Waggoner Center for Alcohol and Addiction Research, 1 University Station A4800, Austin, TX 78712-0159, USA. Fax: +1 512 232 2525., (Y.A. Blednov)
| | | | | | | | | | | |
Collapse
|
30
|
Saba LM, Bennett B, Hoffman PL, Barcomb K, Ishii T, Kechris K, Tabakoff B. A systems genetic analysis of alcohol drinking by mice, rats and men: influence of brain GABAergic transmission. Neuropharmacology 2011; 60:1269-80. [PMID: 21185315 PMCID: PMC3079014 DOI: 10.1016/j.neuropharm.2010.12.019] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2010] [Revised: 12/01/2010] [Accepted: 12/15/2010] [Indexed: 11/19/2022]
Abstract
Genetic influences on the predisposition to complex behavioral or physiological traits can reflect genetic polymorphisms that lead to altered gene product function, and/or variations in gene expression levels. We have explored quantitative variations in an animal's alcohol consumption, using a genetical genomic/phenomic approach. In our studies, gene expression is correlated with amount of alcohol consumed, and genomic regions that regulate the alcohol consumption behavior and the quantitative levels of gene expression (behavioral and expression quantitative trait loci [QTL]) are determined and used as a filter to identify candidate genes predisposing the behavior. We determined QTLs for alcohol consumption using the LXS panel of recombinant inbred mice. We then identified genes that were: 1) differentially expressed between five high and five low alcohol-consuming lines or strains of mice; and 2) were physically located in, or had an expression QTL (eQTL) within the alcohol consumption QTLs. Comparison of mRNA and protein levels in brains of high and low alcohol consuming mice led us to a bioinformatic examination of potential regulation by microRNAs of an identified candidate transcript, Gnb1 (G protein beta subunit 1). We combined our current analysis with our earlier work identifying candidate genes for the alcohol consumption trait in mice, rats and humans. Our overall analysis leads us to postulate that the activity of the GABAergic system, and in particular GABA release and GABA receptor trafficking and signaling, which involves G protein function, contributes significantly to genetic variation in the predisposition to varying levels of alcohol consumption. This article is part of a Special Issue entitled 'Trends in neuropharmacology: in memory of Erminio Costa'.
Collapse
Affiliation(s)
- Laura M. Saba
- University of Colorado Denver School of Medicine, PO Box 6511, MS 8303, Aurora, CO 80045 USA; , , , , ,
| | - Beth Bennett
- University of Colorado Denver School of Medicine, PO Box 6511, MS 8303, Aurora, CO 80045 USA; , , , , ,
| | - Paula L. Hoffman
- University of Colorado Denver School of Medicine, PO Box 6511, MS 8303, Aurora, CO 80045 USA; , , , , ,
| | - Kelsey Barcomb
- University of Colorado Denver School of Medicine, PO Box 6511, MS 8303, Aurora, CO 80045 USA; , , , , ,
| | - Takao Ishii
- University of Colorado Denver School of Medicine, PO Box 6511, MS 8303, Aurora, CO 80045 USA; , , , , ,
| | - Katerina Kechris
- Colorado School of Public Health, Campus Box B119, Aurora, CO 80045 USA,
| | - Boris Tabakoff
- University of Colorado Denver School of Medicine, PO Box 6511, MS 8303, Aurora, CO 80045 USA; , , , , ,
| |
Collapse
|
31
|
Ethanol-induced changes in the expression of proteins related to neurotransmission and metabolism in different regions of the rat brain. Pharmacol Biochem Behav 2011; 99:428-36. [PMID: 21397625 DOI: 10.1016/j.pbb.2011.03.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2010] [Revised: 03/01/2011] [Accepted: 03/04/2011] [Indexed: 01/06/2023]
Abstract
Despite extensive description of the damaging effects of chronic alcohol exposure on brain structure, mechanistic explanations for the observed changes are just emerging. To investigate regional brain changes in protein expression levels following chronic ethanol treatment, one rat per sibling pair of male Wistar rats was exposed to intermittent (14 h/day) vaporized ethanol, the other to air for 26 weeks. At the end of 24 weeks of vapor exposure, the ethanol group had blood ethanol levels averaging 450 mg%, had not experienced a protracted (> 16 h) withdrawal from ethanol, and revealed only mild evidence of hepatic steatosis. Extracted brains were micro-dissected to isolate the prefrontal cortex (PFC), dorsal striatum (STR), corpus callosum genu (CCg), CC body (CCb), anterior vermis (AV), and anterior dorsal lateral cerebellum (ADLC) for protein analysis with two-dimensional gel electrophoresis. Expression levels for 54 protein spots were significantly different between the ethanol- and air-treated groups. Of these 54 proteins, tandem mass spectroscopy successfully identified 39 unique proteins, the levels of which were modified by ethanol treatment: 13 in the PFC, 7 in the STR, 2 in the CCg, 7 in the CCb, 7 in the AV, and 5 in the ADLC. The functions of the proteins altered by chronic ethanol exposure were predominantly associated with neurotransmitter systems in the PFC and cell metabolism in the STR. Stress response proteins were elevated only in the PFC, AV, and ADLC perhaps supporting a role for frontocerebellar circuitry disruption in alcoholism. Of the remaining proteins, some had functions associated with cytoskeletal physiology (e.g., in the CCb) and others with transcription/translation (e.g., in the ADLC). Considered collectively, all but 4 of the 39 proteins identified in the present study have been previously identified in ethanol gene- and/or protein-expression studies lending support for their role in ethanol-related brain alterations.
Collapse
|
32
|
Sari Y, Sakai M, Weedman JM, Rebec GV, Bell RL. Ceftriaxone, a beta-lactam antibiotic, reduces ethanol consumption in alcohol-preferring rats. Alcohol Alcohol 2011; 46:239-46. [PMID: 21422004 DOI: 10.1093/alcalc/agr023] [Citation(s) in RCA: 117] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
AIMS Changes in glutamatergic transmission affect many aspects of neuroplasticity associated with ethanol and drug addiction. For instance, ethanol- and drug-seeking behavior is promoted by increased glutamate transmission in key regions of the motive circuit. We hypothesized that because glutamate transporter 1 (GLT1) is responsible for the removal of most extracellular glutamate, up-regulation or activation of GLT1 would attenuate ethanol consumption. METHODS Alcohol-preferring (P) rats were given 24 h/day concurrent access to 15 and 30% ethanol, water and food for 7 weeks. During Week 6, P rats received either 25, 50, 100 or 200 mg/kg ceftriaxone (CEF, i.p.), a β-lactam antibiotic known to elevate GLT1 expression, or a saline vehicle for five consecutive days. Water intake, ethanol consumption and body weight were measured daily for 15 days starting on Day 1 of injections. We also tested the effects of CEF (100 and 200 mg/kg, i.p.) on daily sucrose (10%) consumption as a control for motivated behavioral drinking. RESULTS Statistical analyses revealed a significant reduction in daily ethanol, but not sucrose, consumption following CEF treatment. During the post treatment period, there was a recovery of ethanol intake across days. Dose-dependent increases in water intake were manifest concurrent with the CEF-induced decreases in ethanol intake. Nevertheless, CEF did not affect body weight. An examination of a subset of the CEF-treated ethanol-drinking rats, on the third day post CEF treatment, revealed increases in GTL1 expression levels within the prefrontal cortex and nucleus accumbens. CONCLUSIONS These results indicate that CEF effectively reduces ethanol intake, possibly through activation of GLT1, and may be a potential therapeutic drug for alcohol addiction treatment.
Collapse
Affiliation(s)
- Youssef Sari
- Department of Pharmacology, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Health Science Campus, 3000 Arlington Avenue, Toledo, OH 43614, USA.
| | | | | | | | | |
Collapse
|
33
|
Molecular targets of alcohol action: Translational research for pharmacotherapy development and screening. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2011; 98:293-347. [PMID: 21199775 DOI: 10.1016/b978-0-12-385506-0.00007-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Alcohol abuse and dependence are multifaceted disorders with neurobiological, psychological, and environmental components. Research on other complex neuropsychiatric diseases suggests that genetically influenced intermediate characteristics affect the risk for heavy alcohol consumption and its consequences. Diverse therapeutic interventions can be developed through identification of reliable biomarkers for this disorder and new pharmacological targets for its treatment. Advances in the fields of genomics and proteomics offer a number of possible targets for the development of new therapeutic approaches. This brain-focused review highlights studies identifying neurobiological systems associated with these targets and possible pharmacotherapies, summarizing evidence from clinically relevant animal and human studies, as well as sketching improvements and challenges facing the fields of proteomics and genomics. Concluding thoughts on using results from these profiling technologies for medication development are also presented.
Collapse
|
34
|
Oneda B, Preisig M, Dobrinas M, Eap CB. Lack of association between genetic polymorphisms of ARRB2 and alcohol dependence in a Caucasian population. Alcohol Alcohol 2010; 45:590-1. [PMID: 20864483 DOI: 10.1093/alcalc/agq064] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
35
|
Pronko SP, Saba LM, Hoffman PL, Tabakoff B. Type 7 adenylyl cyclase-mediated hypothalamic-pituitary-adrenal axis responsiveness: influence of ethanol and sex. J Pharmacol Exp Ther 2010; 334:44-52. [PMID: 20363852 PMCID: PMC2912051 DOI: 10.1124/jpet.110.166793] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2010] [Accepted: 04/01/2010] [Indexed: 11/22/2022] Open
Abstract
Although ethanol has been considered to be an anxiolytic agent, consumption of ethanol has also been shown to increase plasma adrenocorticotropin and glucocorticoids. The corticotrophin-releasing factor (CRF) receptor 1alpha (CRF-R1) is a G protein-coupled receptor that activates adenylyl cyclase (AC), leading to adrenocorticotropin (and subsequently glucocorticoid) release into the circulation. There are nine members of the membrane-bound AC family, and the type 7 AC (AC7) is most sensitive to ethanol, which enhances the responsiveness of AC7 to G protein-coupled receptor activation. We determined the time course of ethanol's effect on plasma adrenocorticotropin and corticosterone levels in male and female AC7 transgenic (Adcy7(huTG)) mice (in which AC7 is overexpressed in neural tissue) and AC7 heterozygous knockdown [Adcy7(+/-)] mice (in which AC7 is underexpressed in neural tissue), and their respective littermate controls [wild type (WT)]. CRF-R1 mRNA and mRNA and protein for different forms of ACs were measured by using gene expression arrays, quantitative reverse transcription-polymerase chain reaction, and immunoblotting in pituitaries of all animals. Our results demonstrated increased levels of AC7 in pituitary of Adcy7(huTG) mice and decreased levels in pituitary of Adcy7(+/-) mice compared with WT animals. Male and female Adcy7(huTG) mice displayed higher plasma adrenocorticotropin and corticosterone levels than WT and/or Adcy7(+/-) mice after ethanol injection. Female mice displayed higher adrenocorticotropin and corticosterone levels after ethanol injection than males, regardless of genotype. The data provide evidence for an integral role of AC7 in the increase of plasma adrenocorticotropin and corticosterone levels during alcohol intoxication.
Collapse
Affiliation(s)
- Sergey P Pronko
- Department of Pharmacology, University of Colorado Denver, School of Medicine, Aurora, CO 80045, USA
| | | | | | | |
Collapse
|
36
|
Quantitative trait loci contributing to physiological and behavioural ethanol responses after acute and chronic treatment. Int J Neuropsychopharmacol 2010; 13:155-69. [PMID: 19691874 DOI: 10.1017/s1461145709990447] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The aim of the present study was the identification of gene loci that contribute to the development and manifestation of behaviours related to acute and chronic alcohol exposure, as well as to alcohol withdrawal. For this purpose, we performed a serial behavioural phenotyping of 534 animals from the second filial (F2) generation of a C57BL/6J and C3H/HeJ mice intercross in paradigms with relevance to alcohol dependence. First, ethanol-induced hypothermia was determined in ethanol-naive animals. The mice then received an ethanol solution for several weeks as their only fluid source. Ethanol tolerance, locomotor activity and anxiety-related behaviours were evaluated. The ethanol was next withdrawn and the withdrawal severity was assessed. The ethanol-experienced animals were finally analysed in a two-bottle choice paradigm to determine ethanol preference and stress-induced changes in ethanol preference. The genotypes of these mice were subsequently assessed by microsatellite marker mapping. We genotyped 264 markers with an average marker distance of 5.56 cM, which represents a high-density whole genome coverage. Quantitative trait loci (QTL) were subsequently identified using univariate analysis performed with the R/qtl tool, which is an extensible, interactive environment for mapping QTL in experimental crosses. We found QTL that have already been published, thus validating the serial phenotyping protocol, and identified several novel loci. Our analysis demonstrates that the various responses to ethanol are regulated by independent groups of genes.
Collapse
|
37
|
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]
|
38
|
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
|
39
|
Spanagel R. Alcoholism: A Systems Approach From Molecular Physiology to Addictive Behavior. Physiol Rev 2009; 89:649-705. [DOI: 10.1152/physrev.00013.2008] [Citation(s) in RCA: 491] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Alcohol consumption is an integral part of daily life in many societies. The benefits associated with the production, sale, and use of alcoholic beverages come at an enormous cost to these societies. The World Health Organization ranks alcohol as one of the primary causes of the global burden of disease in industrialized countries. Alcohol-related diseases, especially alcoholism, are the result of cumulative responses to alcohol exposure, the genetic make-up of an individual, and the environmental perturbations over time. This complex gene × environment interaction, which has to be seen in a life-span perspective, leads to a large heterogeneity among alcohol-dependent patients, in terms of both the symptom dimensions and the severity of this disorder. Therefore, a reductionistic approach is not very practical if a better understanding of the pathological processes leading to an addictive behavior is to be achieved. Instead, a systems-oriented perspective in which the interactions and dynamics of all endogenous and environmental factors involved are centrally integrated, will lead to further progress in alcohol research. This review adheres to a systems biology perspective such that the interaction of alcohol with primary and secondary targets within the brain is described in relation to the behavioral consequences. As a result of the interaction of alcohol with these targets, alterations in gene expression and synaptic plasticity take place that lead to long-lasting alteration in neuronal network activity. As a subsequent consequence, alcohol-seeking responses ensue that can finally lead via complex environmental interactions to an addictive behavior.
Collapse
|
40
|
Hu W, Saba L, Kechris K, Bhave SV, Hoffman PL, Tabakoff B. Genomic insights into acute alcohol tolerance. J Pharmacol Exp Ther 2008; 326:792-800. [PMID: 18550690 PMCID: PMC2574863 DOI: 10.1124/jpet.108.137521] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Alcohol "sensitivity" has been proposed as a predictive factor for development of alcohol dependence (Schuckit et al., 2005). Most measures of alcohol sensitivity in humans and animals include a component that can be ascribed to acute functional tolerance (AFT). AFT is a form of tolerance that develops within a single period of alcohol exposure and has a genetic component. We used microarray technology as well as quantitative trait locus analysis of phenotypic and gene expression data across 30 BXD recombinant inbred strains of mice, 20 inbred strains of mice, and two replicate lines of mice selectively bred for differences in AFT, to identify differentially expressed candidate genes that contribute to predisposition to AFT. Eight candidate genes were identified by our statistical and filtering methods. The location of brain expression of these genes was mapped using the Allen Brain Atlas (http://www.brain-map.org), and the transcript location and molecular pathway analysis indicated that brain structures and biochemical pathways implicated in long-term potentiation and memory might also participate in the generation of acute functional alcohol tolerance.
Collapse
Affiliation(s)
- Wei Hu
- Department of Pharmacology, University of Colorado School of Medicine, Mail Stop 8303, P.O. Box 6511, Aurora, CO 80045-0511, USA
| | | | | | | | | | | |
Collapse
|
41
|
The genomic determinants of alcohol preference in mice. Mamm Genome 2008; 19:352-65. [PMID: 18563486 DOI: 10.1007/s00335-008-9115-z] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2008] [Accepted: 05/14/2008] [Indexed: 02/06/2023]
Abstract
Searches for the identity of genes that influence the levels of alcohol consumption by humans and other animals have often been driven by presupposition of the importance of particular gene products in determining positively or negatively reinforcing effects of ethanol. We have taken an unbiased approach and performed a meta-analysis across three types of mouse populations to correlate brain gene expression with levels of alcohol intake. Our studies, using filtering procedures based on QTL analysis, produced a list of eight candidate genes with highly heritable expression, which could explain a significant amount of the variance in alcohol preference in mice. Using the Allen Brain Atlas for gene expression, we noted that the candidate genes' expression was localized to the olfactory and limbic areas as well as to the orbitofrontal cortex. Informatics techniques and pathway analysis illustrated the role of the candidate genes in neuronal migration, differentiation, and synaptic remodeling. The importance of olfactory cues, learning and memory formation (Pavlovian conditioning), and cortical executive function, for regulating alcohol intake by animals (including humans), is discussed.
Collapse
|
42
|
Sommer WH, Saavedra JM. Targeting brain angiotensin and corticotrophin-releasing hormone systems interaction for the treatment of mood and alcohol use disorders. J Mol Med (Berl) 2008; 86:723-8. [PMID: 18449521 DOI: 10.1007/s00109-008-0333-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2008] [Revised: 02/15/2008] [Accepted: 02/20/2008] [Indexed: 01/16/2023]
Abstract
The brain renin-angiotensin system (RAS) participates importantly in the regulation of endocrine, autonomic, and behavioral response to stress. Recent data indicate that central action of AT(1) receptor antagonists can reduce anxiety symptoms in experimental animals. Furthermore, central inhibition of RAS activity decreases ethanol intake in an animal model of alcoholism. Pathological anxiety responses and the development of substance dependence are both critically mediated through corticotrophin-releasing hormone (CRH) systems, and the RAS is positioned to interact both with hypothalamic as well as extrahypothalamic CRH systems. The thesis of this paper is that the RAS is part of the neurochemical dysregulation underlying negative affective states, anxiety disorders, and ethanol dependence and that medications targeting the RAS should be considered to augment the treatment of these disorders.
Collapse
Affiliation(s)
- Wolfgang H Sommer
- Laboratory of Clinical and Translational Studies, National Institute of Alcohol Abuse and Alcoholism, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892-1108, USA.
| | | |
Collapse
|
43
|
Björk K, Rimondini R, Hansson AC, Terasmaa A, Hyytiä P, Heilig M, Sommer WH. Modulation of voluntary ethanol consumption by beta-arrestin 2. FASEB J 2008; 22:2552-60. [PMID: 18367649 DOI: 10.1096/fj.07-102442] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Beta-arrestin 2 is a multifunctional key component of the G protein-coupled receptor complex and is involved in mu-opiate and dopamine D2 receptor signaling, both of which are thought to mediate the rewarding effects of ethanol consumption. We identified elevated expression of the beta-arrestin 2 gene (Arrb2) in the striatum and the hippocampus of ethanol-preferring AA rats compared to their nonpreferring counterpart ANA line. Differential mRNA expression was accompanied by different levels of Arrb2 protein. The elevated expression was associated with a 7-marker haplotype in complete linkage disequilibrium, which segregated fully between the lines, and was unique to the preferring line. Furthermore, a single, distinct, and highly significant quantitative trait locus for Arrb2 expression in hippocampus and striatum was identified at the locus of this gene, providing evidence that genetic variation may affect a cis-regulatory mechanism for expression and regional control of Arrb2. These findings were functionally validated using mice lacking Arrb2, which displayed both reduced voluntary ethanol consumption and ethanol-induced psychomotor stimulation. Our results demonstrate that beta-arrestin 2 modulates acute responses to ethanol and is an important mediator of ethanol reward.
Collapse
Affiliation(s)
- K Björk
- Laboratory of Clinical and Translational Studies, NIAAA/NIH, 10 Center Dr., Bethesda, MD 20892-1108, USA
| | | | | | | | | | | | | |
Collapse
|
44
|
Mayfield RD, Harris RA, Schuckit MA. Genetic factors influencing alcohol dependence. Br J Pharmacol 2008; 154:275-87. [PMID: 18362899 DOI: 10.1038/bjp.2008.88] [Citation(s) in RCA: 127] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Plentiful data from both animal and human studies support the importance of genetic influences in substance abuse and dependence (Bierut et al., 1998; Tsuang et al., 1998; Kendler et al., 2003). This review summarizes the evidence supporting such genetic influences, places them into perspective regarding animal and human studies, discusses the importance of both genes and environment, and highlights some specific genes of interest regarding the vulnerabilities for problems associated with alcohol use disorders. A long history of repetitive heavy use of alcohol exists across generations as well as the high prevalence of alcohol-related problems in Western societies. Moreover, the information offered here addresses the importance of more general issues regarding genetics and gene expression related to alcohol abuse and dependence.
Collapse
Affiliation(s)
- R D Mayfield
- Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, TX 78712, USA
| | | | | |
Collapse
|
45
|
Hofstetter JR, Svihla-Jones DA, Mayeda AR. A QTL on mouse chromosome 12 for the genetic variance in free-running circadian period between inbred strains of mice. J Circadian Rhythms 2007; 5:7. [PMID: 17974007 PMCID: PMC2174920 DOI: 10.1186/1740-3391-5-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2007] [Accepted: 10/31/2007] [Indexed: 11/21/2022] Open
Abstract
Background Many genes control circadian period in mice. Prior studies suggested a quantitative trait locus (QTL) on proximal mouse chromosome 12 for interstrain differences in circadian period. Since the B6.D2NAhrd/J strain has DBA/2 alleles for a portion of proximal chromosome 12 introgressed onto its C57BL/6J background, we hypothesized that these mice would have a shorter circadian period than C57BL/6J mice. Methods We compared circadian phenotypes of B6.D2NAhrd/J and C57BL/6 mice: period of general locomotor activity in constant dark and rest/activity pattern in alternating light and dark. We genotyped the B6.D2NAhrd/J mice to characterize the size of the genomic insert. To aid in identifying candidate quantitative trait genes we queried databases about the resident SNPs, whole brain gene expression in C57BL/6J versus DBA/2J mice, and circadian patterns of gene expression. Results The B6.D2NAhrd/J inbred mice have a shorter circadian period of locomotor activity than the C57BL/6J strain. Furthermore, the genomic insert is associated with another phenotype: the mean phase of activity minimum in the dark part of a light-dark lighting cycle. It was one hour later than in the background strain. The B6.D2NAhrd/J mice have a DBA/2J genomic insert spanning 35.4 to 41.0 megabase pairs on Chromosome 12. The insert contains 15 genes and 12 predicted genes. In this region Ahr (arylhydrocarbon receptor) and Zfp277 (zinc finger protein 277) both contain non-synonymous SNPs. Zfp277 also showed differential expression in whole brain and was cis-regulated. Three genes and one predicted gene showed a circadian pattern of expression in liver, including Zfp277. Conclusion We not only fine-mapped the QTL for circadian period on chromosome 12 but found a new QTL there as well: an association with the timing of the nocturnal activity-minimum. Candidate quantitative trait genes in this QTL are zinc finger protein 277 and arylhydrocarbon receptor. Arylhydrocarbon receptor is structurally related to Bmal1, a canonical clock gene.
Collapse
Affiliation(s)
- John R Hofstetter
- Department of Psychiatry, Richard L, Roudebush Veterans Administration Medical Center (VAMC), Indianapolis, IN 46202, USA.
| | | | | |
Collapse
|
46
|
Sommer WH, Rimondini R, Marquitz M, Lidström J, Siems WE, Bader M, Heilig M. Plasticity and impact of the central renin–angiotensin system during development of ethanol dependence. J Mol Med (Berl) 2007; 85:1089-97. [PMID: 17823780 DOI: 10.1007/s00109-007-0255-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2007] [Revised: 05/22/2007] [Accepted: 07/02/2007] [Indexed: 10/22/2022]
Abstract
Pharmacological and genetic interference with the renin-angiotensin system (RAS) seems to alter voluntary ethanol consumption. However, understanding the influence of the RAS on ethanol dependence and its treatment requires modeling the neuroadaptations that occur with prolonged exposure to ethanol. Increased ethanol consumption was induced in rats through repeated cycles of intoxication and withdrawal. Expression of angiotensinogen, angiotensin-converting enzyme, and the angiotensin II receptor, AT1a, was examined by quantitative reverse transcription polymerase chain reaction. Increased ethanol consumption after a history of dependence was associated with increased angiotensinogen expression in medial prefrontal cortex but not in nucleus accumbens or amygdala. Increased angiotensinogen expression also demonstrates that the astroglia is an integral part of the plasticity underlying the development of dependence. The effects of low central RAS activity on increased ethanol consumption were investigated using either spirapril, a blood-brain barrier-penetrating inhibitor of angiotensin-converting enzyme, or transgenic rats (TGR(ASrAOGEN)680) with reduced central angiotensinogen expression. Spirapril reduced ethanol intake in dependent rats compared to controls. After induction of dependence, TGR(ASrAOGEN)680 rats had increased ethanol consumption but to a lesser degree than Wistar rats with the same history of dependence. These data suggest that the central RAS is sensitized in its modulatory control of ethanol consumption in the dependent state, but pharmacological or genetic blockade of the system appears to be insufficient to halt the progression of dependence.
Collapse
Affiliation(s)
- W H Sommer
- Laboratory of Clinical and Translational Studies, NIAAA, NIH, Bldg 10, CRC, Bethesda, MD, 20892-1108, USA.
| | | | | | | | | | | | | |
Collapse
|
47
|
Bhave SV, Hornbaker C, Phang TL, Saba L, Lapadat R, Kechris K, Gaydos J, McGoldrick D, Dolbey A, Leach S, Soriano B, Ellington A, Ellington E, Jones K, Mangion J, Belknap JK, Williams RW, Hunter LE, Hoffman PL, Tabakoff B. The PhenoGen informatics website: tools for analyses of complex traits. BMC Genet 2007; 8:59. [PMID: 17760997 PMCID: PMC2034588 DOI: 10.1186/1471-2156-8-59] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2006] [Accepted: 08/30/2007] [Indexed: 11/26/2022] Open
Abstract
Background With the advent of "omics" (e.g. genomics, transcriptomics, proteomics and phenomics), studies can produce enormous amounts of data. Managing this diverse data and integrating with other biological data are major challenges for the bioinformatics community. Comprehensive new tools are needed to store, integrate and analyze the data efficiently. Description The PhenoGen Informatics website is a comprehensive toolbox for storing, analyzing and integrating microarray data and related genotype and phenotype data. The site is particularly suited for combining QTL and microarray data to search for "candidate" genes contributing to complex traits. In addition, the site allows, if desired by the investigators, sharing of the data. Investigators can conduct "in-silico" microarray experiments using their own and/or "shared" data. Conclusion The PhenoGen website provides access to tools that can be used for high-throughput data storage, analyses and interpretation of the results. Some of the advantages of the architecture of the website are that, in the future, the present set of tools can be adapted for the analyses of any type of high-throughput "omics" data, and that access to new tools, available in the public domain or developed at PhenoGen, can be easily provided.
Collapse
Affiliation(s)
- Sanjiv V Bhave
- Department of Pharmacology, University of Colorado at Denver and Health Sciences Center, Aurora, CO 80045, USA
| | - Cheryl Hornbaker
- Department of Pharmacology, University of Colorado at Denver and Health Sciences Center, Aurora, CO 80045, USA
| | - Tzu L Phang
- Department of Pharmacology, University of Colorado at Denver and Health Sciences Center, Aurora, CO 80045, USA
| | - Laura Saba
- Department of Pharmacology, University of Colorado at Denver and Health Sciences Center, Aurora, CO 80045, USA
| | - Razvan Lapadat
- Department of Pharmacology, University of Colorado at Denver and Health Sciences Center, Aurora, CO 80045, USA
| | - Katherina Kechris
- Department of Pharmacology, University of Colorado at Denver and Health Sciences Center, Aurora, CO 80045, USA
- Department of Preventive Medicine and Biometrics, University of Colorado at Denver and Health Sciences Center, Aurora, CO 80045, USA
| | - Jeanette Gaydos
- Department of Pharmacology, University of Colorado at Denver and Health Sciences Center, Aurora, CO 80045, USA
| | - Daniel McGoldrick
- Department of Pharmacology, University of Colorado at Denver and Health Sciences Center, Aurora, CO 80045, USA
| | - Andrew Dolbey
- Department of Pharmacology, University of Colorado at Denver and Health Sciences Center, Aurora, CO 80045, USA
| | - Sonia Leach
- Department of Pharmacology, University of Colorado at Denver and Health Sciences Center, Aurora, CO 80045, USA
| | - Brian Soriano
- Department of Pharmacology, University of Colorado at Denver and Health Sciences Center, Aurora, CO 80045, USA
| | - Allison Ellington
- Department of Pharmacology, University of Colorado at Denver and Health Sciences Center, Aurora, CO 80045, USA
| | - Eric Ellington
- Department of Pharmacology, University of Colorado at Denver and Health Sciences Center, Aurora, CO 80045, USA
| | - Kendra Jones
- Department of Pharmacology, University of Colorado at Denver and Health Sciences Center, Aurora, CO 80045, USA
| | - Jonathan Mangion
- MRC Clinical Sciences Centre, Faculty of Medicine, Imperial College, London W12 0NN, UK
| | - John K Belknap
- US Department of Veterans Affairs Medical Center, Portland, Oregon 97239, USA
| | - Robert W Williams
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, 855 Monroe Avenue, Memphis, TN 38163, USA
| | - Lawrence E Hunter
- Department of Pharmacology, University of Colorado at Denver and Health Sciences Center, Aurora, CO 80045, USA
| | - Paula L Hoffman
- Department of Pharmacology, University of Colorado at Denver and Health Sciences Center, Aurora, CO 80045, USA
| | - Boris Tabakoff
- Department of Pharmacology, University of Colorado at Denver and Health Sciences Center, Aurora, CO 80045, USA
| |
Collapse
|
48
|
Hofstetter JR, Hitzemann RJ, Belknap JK, Walter NAR, McWeeney SK, Mayeda AR. Characterization of the quantitative trait locus for haloperidol-induced catalepsy on distal mouse chromosome 1. GENES BRAIN AND BEHAVIOR 2007; 7:214-23. [PMID: 17696997 DOI: 10.1111/j.1601-183x.2007.00340.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
We report here the confirmation of the quantitative trait locus for haloperidol-induced catalepsy on distal chromosome (Chr) 1. We determined that this quantitative trait locus was captured in the B6.D2-Mtv7a/Ty congenic mouse strain, whose introgressed genomic interval extends from approximately 169.1 to 191.3 Mb. We then constructed a group of overlapping interval-specific congenic strains to further break up the interval and remapped the locus between 177.5 and 183.4 Mb. We next queried single nucleotide polymorphism (SNP) data sets and identified three genes with nonsynonymous coding SNPs in the quantitative trait locus. We also queried two brain gene expression data sets and found five known genes in this 5.9-Mb interval that are differentially expressed in both whole brain and striatum. Three of the candidate quantitative trait genes were differentially expressed using quantitative real-time polymerase chain reaction analyses. Overall, the current study illustrates how multiple approaches, including congenic fine mapping, SNP analysis and microarray gene expression screens, can be integrated both to reduce the quantitative trait locus interval significantly and to detect promising candidate quantitative trait genes.
Collapse
Affiliation(s)
- J R Hofstetter
- Department of Veterans Affairs, Richard L. Roudebush Medical Center, Indianapolis, IN 46202, USA.
| | | | | | | | | | | |
Collapse
|
49
|
Hines LM, Hoffman PL, Bhave S, Saba L, Kaiser A, Snell L, Goncharov I, LeGault L, Dongier M, Grant B, Pronko S, Martinez L, Yoshimura M, Tabakoff B. A sex-specific role of type VII adenylyl cyclase in depression. J Neurosci 2006; 26:12609-19. [PMID: 17135423 PMCID: PMC6674903 DOI: 10.1523/jneurosci.1040-06.2006] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Major depression represents a complex mental disorder. The identification of biological markers that define subtypes of major depressive disorder would greatly facilitate appropriate medical treatments, as well as provide insight into etiology. Reduced activity of the cAMP signaling system has been implicated in the etiology of major depression. Previous work has shown low adenylyl cyclase activity in platelets and postmortem brain tissue of depressed individuals. Here, we investigate the role of the brain type VII isoform of adenylyl cyclase (AC7) in the manifestation of depressive symptoms in genetically modified animals, using a combination of in vivo behavioral experiments, gene expression profiling, and bioinformatics. We also completed studies with humans on the association of polymorphisms in the AC7 gene with major depressive illness (unipolar depression) based on Diagnostic and Statistical Manual of Mental Disorders IV criteria. Collectively, our results demonstrate a sex-specific influence of the AC7 gene on a heritable form of depressive illness.
Collapse
Affiliation(s)
- Lisa M. Hines
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Paula L. Hoffman
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Sanjiv Bhave
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Laura Saba
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Alan Kaiser
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Larry Snell
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Igor Goncharov
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Lucie LeGault
- Department of Psychiatry, McGill University, Montreal, Quebec, Canada H4H 1R3
| | - Maurice Dongier
- Department of Psychiatry, McGill University, Montreal, Quebec, Canada H4H 1R3
| | - Bridget Grant
- Division of Epidemiology, National Institute on Alcohol Abuse and Alcoholism, Rockville, Maryland 20852, and
| | - Sergey Pronko
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Larry Martinez
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Masami Yoshimura
- Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana 70803
| | - Boris Tabakoff
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, Colorado 80045
| |
Collapse
|
50
|
Björk K, Saarikoski ST, Arlinde C, Kovanen L, Osei-Hyiaman D, Ubaldi M, Reimers M, Hyytiä P, Heilig M, Sommer WH. Glutathione-S-transferase expression in the brain: possible role in ethanol preference and longevity. FASEB J 2006; 20:1826-35. [PMID: 16940154 DOI: 10.1096/fj.06-5896com] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Identification of genes that are differentially expressed in rats bidirectionally selected for alcohol preference might reveal biological mechanisms underlying alcoholism or related phenotypes. Microarray analysis from medial prefrontal cortex (mPFC), a key brain region for drug reward, indicated increased expression of glutathione-S-transferases of the alpha (Gsta4) and mu (Gstm1-5) classes in ethanol-preferring AA rats compared with nonpreferring ANA rats. Real-time RT polymerase chain reaction (RT-PCR) analysis demonstrated approximately 2-fold higher Gsta4 transcript levels in several brain regions of ethanol-naive AA compared with ANA rats. Differences in mRNA levels were accompanied by differential levels of GSTA4 protein. We identified a novel haplotype variant in the rat Gsta4 gene, defined here as var3. Allele frequencies of var3 were markedly different between AA and ANA rats, 52% and 100%, respectively. Gsta4 expression was strongly correlated with the gene dose of var3, with approximately 60% of the variance in expression accounted for by genotype at this locus. The contribution of glutathione S-transferase expression to the ethanol-preferring phenotype is presently unclear. It could, however, underlie observed differences in life span between AA and ANA lines, prompting a utility of this animal model in aging research.
Collapse
Affiliation(s)
- K Björk
- Laboratory of Clinical and Translational Studies, NCI, National Institutes of Health, Bethesda, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|