Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

A single-cell genomic atlas for the effects of chronic ethanol exposure in the mouse dorsal striatum

Abstract

Alcohol use disorder (AUD) is characterized by compulsive drinking, which is thought to be mediated by effects of chronic intermittent ethanol exposure on the dorsal striatum, the input nucleus of the basal ganglia. Despite significant efforts to understand the impact of ethanol on the dorsal striatum, the rich diversity of striatal cell types and multitude of ethanol targets expressed by them necessitates an unbiased, discovery-based approach. In this study, we used single-nuclei RNA-sequencing (snRNA-seq; n = 86,715 cells) to examine the impact of chronic intermittent ethanol exposure on the dorsal striatum in C57BL/6 male and female mice. We detected 462 differentially expressed genes at FDR < 0.05, the majority of which were mapped to spiny projection neurons (SPNs), the most prominent cell type in the striatum. Gene co-expression network analysis and functional annotation of differentially expressed genes revealed down-regulation of postsynaptic intracellular signaling cascades in SPNs. Inflammation-related genes were down-regulated across many neuronal and non-neuronal cell types. Gene set enrichment analyses also pointed to altered states of rare cell types, including the induction of angiogenesis-related genes in vascular cells. A gene module down-regulated specifically in canonical SPNs was enriched for calcium-signaling genes and components of glutamatergic synapses, as well as for genes associated with genetic risk for AUD. Genetic perturbations of six of this module’s hub genes – Foxp1, Bcl11b, Pde10a, Rarb, Rgs9, and Itgr1 – had causal effects on its expression in the mouse striatum and/or on the broader set of differentially expressed genes in alcohol-exposed mice. These data provide important clues as to the impact of ethanol on striatal biology and provide a key resource for future investigation.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Single-nuclei RNA sequencing of the mouse dorsal striatum in the context of chronic intermittent ethanol exposure.
Fig. 2: Discovery and replication of differentially expressed genes in dorsal striatum cell types from chronic intermittent ethanol (CIE)-exposed vs. control mice.
Fig. 3: Gene sets enriched for differential gene expression in CIE vs. control mice.
Fig. 4: Cell type-specific vs. shared effects of chronic intermittent ethanol across striatal cell types.
Fig. 5: Gene co-expression networks dysregulated in spiny projection neurons after chronic intermittent ethanol.
Fig. 6: Causal effects of module M43 hub genes on striatal gene expression.

Similar content being viewed by others

Data availability

Sequencing data have been uploaded to the Gene Expression Omnibus (GSE292642, GSE292791). All other data are presented in the main text and supplementary figures and tables.

Code availability

Code used in the data analysis is available at www.github.com/seth-ament/cie-snrnaseq.

References

  1. Kranzler HR. Overview of alcohol use disorder. Am J Psychiatry. 2023;180:565–72.

    Article  PubMed  Google Scholar 

  2. Mason BJ, Heyser CJ. Alcohol use disorder: the role of medication in recovery. Alcohol Res. 2021;41:7.

    Article  Google Scholar 

  3. Koob GF, Volkow ND. Neurobiology of addiction: a neurocircuitry analysis. Lancet Psychiatry. 2016;3:760–73.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Mancini A, Ghiglieri V, Parnetti L, Calabresi P, Di Filippo M. Neuro-immune cross-talk in the striatum: from basal ganglia physiology to circuit dysfunction. Front Immunol. 2021;12:644294.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Gokce O, Stanley GM, Treutlein B, Neff NF, Camp JG, Malenka RC, et al. Cellular taxonomy of the mouse striatum as revealed by single-cell RNA-seq. Cell Rep. 2016;16:1126–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Saunders A, Macosko EZ, Wysoker A, Goldman M, Krienen FM, de Rivera H, et al. Molecular diversity and specializations among the cells of the adult mouse brain. Cell. 2018;174:1015–30.e16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Fieblinger T. Striatal control of movement: a role for new neuronal (sub-) populations? Front Hum Neurosci. 2021;15:697284.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Patton MS, Heckman M, Kim C, Mu C, Mathur BN. Compulsive alcohol consumption is regulated by dorsal striatum fast-spiking interneurons. Neuropsychopharmacology. 2021;46:351–9.

    Article  CAS  PubMed  Google Scholar 

  9. Egervari G, Siciliano CA, Whiteley EL, Ron D. Alcohol and the brain: from genes to circuits. Trends Neurosci. 2021;44:1004–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Wang J, Lanfranco MF, Gibb SL, Yowell QV, Carnicella S, Ron D. Long-lasting adaptations of the NR2B-containing NMDA receptors in the dorsomedial striatum play a crucial role in alcohol consumption and relapse. J Neurosci. 2010;30:10187–98.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Wang J, Hamida SB, Darcq E, Zhu W, Gibb SL, Lanfranco MF, et al. Ethanol-mediated facilitation of AMPA receptor function in the dorsomedial striatum: implications for alcohol drinking behavior. J Neurosci. 2012;32:15124–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Ron D, Wang J. The NMDA receptor and alcohol addiction. In: Van Dongen, AM, editors. Biology of the NMDA receptor. Boca Raton (FL): CRC Press/Taylor & Francis; 2009.

  13. Choi SJ, Kim KJ, Cho HS, Kim SY, Cho YJ, Hahn SJ, et al. Acute inhibition of corticostriatal synaptic transmission in the rat dorsal striatum by ethanol. Alcohol. 2006;40:95–101.

    Article  CAS  PubMed  Google Scholar 

  14. Haggerty DL, Munoz B, Pennington T, Di Prisco GV, Grecco GG, Atwood BK. The role of anterior insular cortex inputs to dorsolateral striatum in binge alcohol drinking. eLife. 2022;11:e77411.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Blomeley CP, Cains S, Smith R, Bracci E. Ethanol affects striatal interneurons directly and projection neurons through a reduction in cholinergic tone. Neuropsychopharmacology. 2011;36:1033–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Patton MH, Roberts BM, Lovinger DM, Mathur BN. Ethanol disinhibits dorsolateral striatal medium spiny neurons through activation of a presynaptic delta opioid receptor. Neuropsychopharmacology. 2016;41:1831–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Patton MS, Sheats SH, Siclair AN, Mathur BN. Alcohol potentiates multiple GABAergic inputs to dorsal striatum fast-spiking interneurons. Neuropharmacology. 2023;232:109527.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Banerjee N. Neurotransmitters in alcoholism: a review of neurobiological and genetic studies. Indian J Hum Genet. 2014;20:20–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Miller CN, Kamens HM. The role of nicotinic acetylcholine receptors in alcohol-related behaviors. Brain Res Bull. 2020;163:135–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Tupala E, Tiihonen J. Dopamine and alcoholism: neurobiological basis of ethanol abuse. Prog Neuropsychopharmacol Biol Psychiatry. 2004;28:1221–47.

    Article  CAS  PubMed  Google Scholar 

  21. Sarkisyan D, Hussain MZ, Watanabe H, Kononenko O, Bazov I, Zhou X, et al. Downregulation of the endogenous opioid peptides in the dorsal striatum of human alcoholics. Front Cell Neurosci. 2015;9:187.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Adermark L, Jonsson S, Ericson M, Söderpalm B. Intermittent ethanol consumption depresses endocannabinoid-signaling in the dorsolateral striatum of rat. Neuropharmacology. 2011;61:1160–5.

    Article  CAS  PubMed  Google Scholar 

  23. Jeanblanc J, He D-Y, Carnicella S, Kharazia V, Janak PH, Ron D. Endogenous BDNF in the dorsolateral striatum gates alcohol drinking. J Neurosci. 2009;29:13494–502.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Mews P, Cunningham AM, Scarpa J, Ramakrishnan A, Hicks EM, Bolnick S, et al. Convergent abnormalities in striatal gene networks in human cocaine use disorder and mouse cocaine administration models. Sci Adv. 2023;9:eadd8946.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Phan BN, Ray MH, Xue X, Fu C, Fenster RJ, Kohut SJ, et al. Single nuclei transcriptomics in human and non-human primate striatum in opioid use disorder. Nat Commun. 2024;15:878.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Sinirlioglu ZA, Coskunpinar E, Akbas F. miRNA and mRNA expression profiling in rat brain following alcohol dependence and withdrawal. Cell Mol Biol. 2017;63:49–56.

    Article  CAS  PubMed  Google Scholar 

  27. Darlington TM, McCarthy RD, Cox RJ, Miyamoto-Ditmon J, Gallego X, Ehringer MA. Voluntary wheel running reduces voluntary consumption of ethanol in mice: identification of candidate genes through striatal gene expression profiling. Genes Brain Behav. 2016;15:474–90.

    Article  CAS  PubMed  Google Scholar 

  28. Piechota M, Korostynski M, Solecki W, Gieryk A, Slezak M, Bilecki W, et al. The dissection of transcriptional modules regulated by various drugs of abuse in the mouse striatum. Genome Biol. 2010;11:R48.

    Article  PubMed  PubMed Central  Google Scholar 

  29. Brenner E, Tiwari GR, Kapoor M, Liu Y, Brock A, Mayfield RD. Single cell transcriptome profiling of the human alcohol-dependent brain. Hum Mol Genet. 2020;29:1144–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Erickson EK, Farris SP, Blednov YA, Mayfield RD, Harris RA. Astrocyte-specific transcriptome responses to chronic ethanol consumption. Pharmacogenomics J. 2018;18:578–89.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. McCarthy GM, Farris SP, Blednov YA, Harris RA, Mayfield RD. Microglial-specific transcriptome changes following chronic alcohol consumption. Neuropharmacology. 2018;128:416–24.

    Article  CAS  PubMed  Google Scholar 

  32. van den Oord EJ, Xie LY, Zhao M, Aberg KA, Clark SL. A single-nucleus transcriptomics study of alcohol use disorder in the nucleus accumbens. Addict Biol. 2023;28:e13250.

    Article  PubMed  Google Scholar 

  33. Becker HC, Lopez MF. Increased ethanol drinking after repeated chronic ethanol exposure and withdrawal experience in C57BL/6 mice. Alcohol Clin Exp Res. 2004;28:1829–38.

    Article  CAS  PubMed  Google Scholar 

  34. Malaiya S, Cortes-Gutierrez M, Herb BR, Coffey SR, Legg SRW, Cantle JP, et al. Single-nucleus RNA-seq reveals dysregulation of striatal cell identity due to Huntington’s disease mutations. J Neurosci. 2021;41:5534–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Kamath T, Abdulraouf A, Burris SJ, Langlieb J, Gazestani V, Nadaf NM, et al. Single-cell genomic profiling of human dopamine neurons identifies a population that selectively degenerates in Parkinson’s disease. Nat Neurosci. 2022;25:588–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Germain P-L, Lun A, Garcia Meixide C, Macnair W, Robinson MD. Doublet identification in single-cell sequencing data using scDblFinder. F1000Res. 2022;10:979.

    Article  PubMed Central  Google Scholar 

  37. Hao Y, Hao S, Andersen-Nissen E, Mauck WM, Zheng S, Butler A, et al. Integrated analysis of multimodal single-cell data. Cell. 2021;184:3573–87.e29.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Korsunsky I, Millard N, Fan J, Slowikowski K, Zhang F, Wei K, et al. Fast, sensitive and accurate integration of single-cell data with harmony. Nat Methods. 2019;16:1289–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Linderman GC, Zhao J, Roulis M, Bielecki P, Flavell RA, Nadler B, et al. Zero-preserving imputation of single-cell RNA-seq data. Nat Commun. 2022;13:192.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Robinson MD, McCarthy DJ, Smyth GK. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics. 2010;26:139–40.

    Article  CAS  PubMed  Google Scholar 

  41. Murphy AE, Skene NG. A balanced measure shows superior performance of pseudobulk methods in single-cell RNA-sequencing analysis. Nat Commun. 2022;13:7851.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Zhang Y, Parmigiani G, Johnson WE. ComBat-seq: batch effect adjustment for RNA-seq count data. NAR Genom Bioinform. 2020;2:lqaa078.

    Article  PubMed  PubMed Central  Google Scholar 

  43. Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W, et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 2015;43:e47.

    Article  PubMed  PubMed Central  Google Scholar 

  44. Ament SA, Cortes-Gutierrez M, Herb BR, Mocci E, Colantuoni C, McCarthy MM. A single-cell genomic atlas for maturation of the human cerebellum during early childhood. Sci Transl Med. 2023;15:eade1283.

    Article  CAS  PubMed  Google Scholar 

  45. Kember RL, Vickers-Smith R, Zhou H, Xu H, Jennings M, Dao C, et al. Genetic underpinnings of the transition from alcohol consumption to alcohol use disorder: shared and unique genetic architectures in a cross-ancestry sample. Am J Psychiatry. 2023;180:584–93.

    Article  PubMed  PubMed Central  Google Scholar 

  46. de Leeuw CA, Mooij JM, Heskes T, Posthuma D. MAGMA: generalized gene-set analysis of GWAS data. PLoS Comput Biol. 2015;11:e1004219.

    Article  PubMed  PubMed Central  Google Scholar 

  47. Wang N, Langfelder P, Stricos M, Ramanathan L, Richman JB, Vaca R, et al. Mapping brain gene coexpression in daytime transcriptomes unveils diurnal molecular networks and deciphers perturbation gene signatures. Neuron. 2022;110:3318–38.e9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Law CW, Chen Y, Shi W, Smyth GK. voom: Precision weights unlock linear model analysis tools for RNA-seq read counts. Genome Biol. 2014;15:R29.

    Article  PubMed  PubMed Central  Google Scholar 

  49. Lopez MF, Becker HC. Effect of pattern and number of chronic ethanol exposures on subsequent voluntary ethanol intake in C57BL/6J mice. Psychopharmacology. 2005;181:688–96.

    Article  CAS  PubMed  Google Scholar 

  50. Griffin WC, Lopez MF, Becker HC. Intensity and duration of chronic ethanol exposure is critical for subsequent escalation of voluntary ethanol drinking in mice. Alcohol Clin Exp Res. 2009;33:1893–900.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Plaisier SB, Taschereau R, Wong JA, Graeber TG. Rank–rank hypergeometric overlap: identification of statistically significant overlap between gene-expression signatures. Nucleic Acids Res. 2010;38:e169.

    Article  PubMed  PubMed Central  Google Scholar 

  52. Li J, Li C, Loreno EG, Miriyala S, Panchatcharam M, Lu X, et al. Chronic low-dose alcohol consumption promotes cerebral angiogenesis in mice. Front Cardiovasc Med. 2021;8:681627.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Morrow D, Cullen JP, Cahill PA, Redmond EM. Ethanol stimulates endothelial cell angiogenic activity via a Notch- and angiopoietin-1-dependent pathway. Cardiovasc Res. 2008;79:313–21.

    Article  CAS  PubMed  Google Scholar 

  54. Nakayama K, Hasegawa H. Blood vessels as a key mediator for ethanol toxicity: implication for neuronal damage. Life. 2022;12:1882.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Gaziano JM, Concato J, Brophy M, Fiore L, Pyarajan S, Breeling J, et al. Million veteran program: a mega-biobank to study genetic influences on health and disease. J Clin Epidemiol. 2016;70:214–23.

    Article  PubMed  Google Scholar 

  56. Morales-Garcia JA, Redondo M, Alonso-Gil S, Gil C, Perez C, Martinez A, et al. Phosphodiesterase 7 inhibition preserves dopaminergic neurons in cellular and rodent models of parkinson disease. PLoS ONE. 2011;6:e17240.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Morales-Garcia JA, Aguilar-Morante D, Hernandez-Encinas E, Alonso-Gil S, Gil C, Martinez A, et al. Silencing phosphodiesterase 7B gene by lentiviral-shRNA interference attenuates neurodegeneration and motor deficits in hemiparkinsonian mice. Neurobiol Aging. 2015;36:1160–73.

    Article  CAS  PubMed  Google Scholar 

  58. Erro R, Mencacci NE, Bhatia KP. The emerging role of phosphodiesterases in movement disorders. Mov Disord. 2021;36:2225–43.

    Article  PubMed  PubMed Central  Google Scholar 

  59. Logrip ML. Phosphodiesterase regulation of alcohol drinking in rodents. Alcohol. 2015;49:795–802.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Salinas AG, Mateo Y, Cuzon Carlson VC, Stinnett GS, Luo G, Seasholtz AF, et al. Long-term alcohol consumption alters dorsal striatal dopamine release and regulation by D2 dopamine receptors in rhesus macaques. Neuropsychopharmacology. 2021;46:1432–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Trantham-Davidson H, Chandler LJ. Alcohol-induced alterations in dopamine modulation of prefrontal activity. Alcohol. 2015;49:773–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Heinz A, Siessmeier T, Wrase J, Hermann D, Klein S, Grüsser SM, et al. Correlation between dopamine D2 receptors in the ventral striatum and central processing of alcohol cues and craving. Am J Psychiatry. 2004;161:1783–9.

    Article  PubMed  Google Scholar 

  63. Feltmann K, Borroto-Escuela DO, Rüegg J, Pinton L, de Oliveira Sergio T, Narváez M, et al. Effects of long-term alcohol drinking on the dopamine D2 receptor: gene expression and heteroreceptor complexes in the striatum in rats. Alcohol Clin Exp Res. 2018;42:338–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Hamada K, Lasek AW. Receptor tyrosine kinases as therapeutic targets for alcohol use disorder. Neurotherapeutics. 2020;17:4–16.

    Article  CAS  PubMed  Google Scholar 

  65. Deinhardt, K, Chao, MV. Trk receptors. In: Lewin, GR & Carter, BD, editors. Neurotrophic factors. Berlin, Heidelberg: Springer; 2014. pp. 103–19.

  66. Gupta VK, You Y, Gupta VB, Klistorner A, Graham SL. TrkB receptor signalling: implications in neurodegenerative, psychiatric and proliferative disorders. Int J Mol Sci. 2013;14:10122–42.

    Article  PubMed  PubMed Central  Google Scholar 

  67. Altar CA, Cai N, Bliven T, Juhasz M, Conner JM, Acheson AL, et al. Anterograde transport of brain-derived neurotrophic factor and its role in the brain. Nature. 1997;389:856–60.

    Article  CAS  PubMed  Google Scholar 

  68. Li Y, Yui D, Luikart BW, McKay RM, Li Y, Rubenstein JL, et al. Conditional ablation of brain-derived neurotrophic factor-TrkB signaling impairs striatal neuron development. Proc Natl Acad Sci. 2012;109:15491–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Logrip ML, Barak S, Warnault V, Ron D. Corticostriatal BDNF and alcohol addiction. Brain Res. 2015;1628:60–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Ghezzi, P, Bigini, P, Mengozzi, M. Role of Erythropoietin in Inflammatory Pathologies of the CNS. In: Höke, A, editors. Erythropoietin and the nervous system: novel therapeutic options for neuroprotection. Boston, MA: Springer US; 2006. pp. 191–209

  71. Rey F, Balsari A, Giallongo T, Ottolenghi S, Di Giulio AM, Samaja M, et al. Erythropoietin as a neuroprotective molecule: an overview of its therapeutic potential in neurodegenerative diseases. ASN Neuro. 2019;11:1759091419871420.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Hendriks JJA, Slaets H, Carmans S, de Vries HE, Dijkstra CD, Stinissenet P, et al. Leukemia inhibitory factor modulates production of inflammatory mediators and myelin phagocytosis by macrophages. J Neuroimmunol. 2008;204:52–57.

    Article  CAS  PubMed  Google Scholar 

  73. Luo Y, Ali T, Liu Z, Gao R, Li A, Yang C, et al. EPO prevents neuroinflammation and relieves depression via JAK/STAT signaling. Life Sci. 2023;333:122102.

    Article  CAS  PubMed  Google Scholar 

  74. Kim H, Seo JS, Lee S-Y, Ha K-T, Choi BT, Shin Y-I, et al. AIM2 inflammasome contributes to brain injury and chronic post-stroke cognitive impairment in mice. Brain Behav Immun. 2020;87:765–76.

    Article  CAS  PubMed  Google Scholar 

  75. Chai L, Dai L, Che Y, Xu J, Liu G, Zhang Z, et al. LRRC19, a novel member of the leucine-rich repeat protein family, activates NF-κB and induces expression of proinflammatory cytokines. Biochem Biophys Res Commun. 2009;388:543–8.

    Article  CAS  PubMed  Google Scholar 

  76. Lima IV, Bastos LF, Limborço-Filho M, Fiebich BL, de Oliveira AC. Role of prostaglandins in neuroinflammatory and neurodegenerative diseases. Mediators Inflamm. 2012;2012:946813.

    Article  PubMed  PubMed Central  Google Scholar 

  77. Bordon Y. Fine-tuning of inflammation by F-box proteins. Nat Rev Immunol. 2013;13:305–305.

    Google Scholar 

  78. Na YR, Jung D, Stakenborg M, Jang H, Gu GJ, Jeong MR, et al. Prostaglandin E2 receptor PTGER4-expressing macrophages promote intestinal epithelial barrier regeneration upon inflammation. Gut. 2021;70:2249–60.

    Article  CAS  PubMed  Google Scholar 

  79. Heger K, Wickliffe KE, Ndoja A, Zhang J, Murthy A, Dugger DL, et al. OTULIN limits cell death and inflammation by deubiquitinating LUBAC. Nature. 2018;559:120–4.

    Article  CAS  PubMed  Google Scholar 

  80. Ren Y, Jiang J, Jiang W, Zhou X, Lu W, Wang J, et al. Spata2 knockdown exacerbates brain inflammation via NF-κB/P38MAPK signaling and NLRP3 inflammasome activation in cerebral ischemia/reperfusion rats. Neurochem Res. 2021;46:2262–75.

    Article  CAS  PubMed  Google Scholar 

  81. Cardamone MD, Krones A, Tanasa B, Taylor H, Ricci L, Ohgi KA, et al. A protective strategy against hyperinflammatory responses requiring the nontranscriptional actions of GPS2. Mol Cell. 2012;46:91–104.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Achur RN, Freeman WM, Vrana KE. Circulating cytokines as biomarkers of alcohol abuse and alcoholism. J Neuroimmune Pharmacol. 2010;5:83–91.

    Article  PubMed  Google Scholar 

  83. Adams C, Perry N, Conigrave J, Hurzeler T, Stevens J, Dunbar KPY, et al. Central markers of neuroinflammation in alcohol use disorder: a meta-analysis of neuroimaging, cerebral spinal fluid, and postmortem studies. Alcohol Clin Exp Res. 2023;47:197–208.

    Article  CAS  Google Scholar 

  84. Erickson EK, Grantham EK, Warden AS, Harris RA. Neuroimmune signaling in alcohol use disorder. Pharmacol Biochem Behav. 2019;177:34–60.

    Article  CAS  PubMed  Google Scholar 

  85. Lékó AH, Ray LA, Leggio L. The vicious cycle between (neuro)inflammation and alcohol use disorder: an opportunity to develop new medications? Alcohol Clin Exp Res. 2023;47:843–7.

    Article  Google Scholar 

  86. Crews FT, Lawrimore CJ, Walter TJ, Coleman LG. The role of neuroimmune signaling in alcoholism. Neuropharmacology. 2017;122:56–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Kitagawa M, Hojo M, Imayoshi I, Goto M, Ando M, Ohtsuka T, et al. Hes1 and Hes5 regulate vascular remodeling and arterial specification of endothelial cells in brain vascular development. Mech Dev. 2013;130:458–66.

    Article  CAS  PubMed  Google Scholar 

  88. Ohtsuka T, Ishibashi M, Gradwohl G, Nakanishi S, Guillemot F, Kageyama R. Hes1 and Hes5 as Notch effectors in mammalian neuronal differentiation. EMBO J. 1999;18:2196–207.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Liu Z-J, Xiao M, Balint K, Soma A, Pinnix CC, Capobianco AJ, et al. Inhibition of endothelial cell proliferation by Notch1 signaling is mediated by repressing MAPK and PI3K/Akt pathways and requires MAML1. FASEB J. 2006;20:1009–11.

    Article  CAS  PubMed  Google Scholar 

  90. Quillard T, Coupel S, Coulon F, Fitau J, Chatelais M, Cuturi MC, et al. Impaired Notch4 activity elicits endothelial cell activation and apoptosis. Arterioscler Thromb Vasc Biol. 2008;28:2258–65.

    Article  CAS  PubMed  Google Scholar 

  91. Lebrin F, Deckers M, Bertolino P, ten Dijke P. TGF-β receptor function in the endothelium. Cardiovasc Res. 2005;65:599–608.

    Article  CAS  PubMed  Google Scholar 

  92. Jarad M, Kuczynski EA, Morrison J, Viloria-Petit AM, Coomber BL. Release of endothelial cell associated VEGFR2 during TGF-β modulated angiogenesis in vitro. BMC Cell Biol. 2017;18:10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Vore AS, Deak T. Alcohol, inflammation, and blood-brain barrier function in health and disease across development. Int Rev Neurobiol. 2022;161:209–49.

    CAS  PubMed  Google Scholar 

  94. Carrino D, Branca JJV, Becatti M, Paternostro F, Morucci G, Gulisano M, et al. Alcohol-induced blood-brain barrier impairment: an in vitro study. Int J Environ Res Public Health. 2021;18:2683.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Dilly GA, Kittleman CW, Kerr TM, Messing RO, Mayfield RD. Cell-type specific changes in PKC-delta neurons of the central amygdala during alcohol withdrawal. Transl Psychiatry. 2022;12:289.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Salem NA, Manzano L, Keist MW, Ponomareva O, Roberts AJ, Roberto M, et al. Cell-type brain-region specific changes in prefrontal cortex of a mouse model of alcohol dependence. Neurobiol Dis. 2024;190:106361.

    Article  CAS  PubMed  Google Scholar 

  97. Finan C, Gaulton A, Kruger FA, Lumbers RT, Shah T, Engmann J, et al. The druggable genome and support for target identification and validation in drug development. Sci Transl Med. 2017;9:eaag1166.

    Article  PubMed  PubMed Central  Google Scholar 

  98. Logrip ML, Vendruscolo LF, Schlosburg JE, Koob GF, Zorrilla EP. Phosphodiesterase 10A regulates alcohol and saccharin self-administration in rats. Neuropsychopharmacology. 2014;39:1722–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Menniti FS, Chappie TA, Schmidt CJ. PDE10A inhibitors-clinical failure or window into antipsychotic drug action? Front Neurosci. 2020;14:600178.

    Article  PubMed  Google Scholar 

  100. Cuzon Carlson VC, Seabold GK, Helms CM, Garg N, Odagiri M, Rau AR, et al. Synaptic and morphological neuroadaptations in the putamen associated with long-term, relapsing alcohol drinking in primates. Neuropsychopharmacology. 2011;36:2513–28.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Wilcox MV, Cuzon Carlson VC, Sherazee N, Sprow GM, Bock R, Thiele TE, et al. Repeated binge-like ethanol drinking alters ethanol drinking patterns and depresses striatal GABAergic transmission. Neuropsychopharmacology. 2014;39:579–94.

    Article  CAS  PubMed  Google Scholar 

  102. Cuzon Carlson VC, Grant KA, Lovinger DM. Synaptic adaptations to chronic ethanol intake in male rhesus monkey dorsal striatum depend on age of drinking onset. Neuropharmacology. 2018;131:128–42.

    Article  CAS  PubMed  Google Scholar 

  103. Becker, HC & Lopez, MF Animal models of excessive alcohol consumption in rodents. Curr Top Behav Neurosci. 2024;1–32 https://doi.org/10.1007/7854_2024_461.

Download references

Acknowledgements

This study was supported by grants from the National Institute on Alcohol Abuse and Alcoholism (R01 AA024845, BNM, PI) and the National Institute on Drug Abuse (U01 DA051947, SAA and MKL, PIs). This research is based in part on data from the Million Veteran Program (MVP), Office of Research and Development, Veterans Health Administration, and was supported by award #I01 BX004820 and the Veterans Integrated Service Network 4 Mental Illness Research, Education and Clinical Center. This publication does not represent the views of the Department of Veteran Affairs or the United States Government.

Author information

Authors and Affiliations

Authors

Contributions

BNM and SAA conceptualized the study. MSP, MC-G, and RRC performed experiments. EW, ZJ, BHG, and SAA performed formal analyses of the data. HRK, MKL, SAA, and BNM supervised the study and acquired funding. EW, SAA, and BNM wrote the original draft. All authors reviewed and edited the manuscript.

Corresponding authors

Correspondence to Seth A. Ament or Brian N. Mathur.

Ethics declarations

Competing interests

Dr. Kranzler is a member of advisory boards for Altimmune, Clearmind Medicine, Dicerna Pharmaceuticals, Enthion Pharmaceuticals, and Sophrosyne Pharmaceuticals; a consultant to Sobrera Pharmaceuticals and Altimmune; the recipient of research funding and medication supplies for an investigator-initiated study from Alkermes; a member of the American Society of Clinical Psychopharmacology’s Alcohol Clinical Trials Initiative, which was supported in the last three years by Alkermes, Dicerna, Ethypharm, Lundbeck, Mitsubishi, Otsuka, and Pear Therapeutics; and a holder of U.S. patent 10,900,082 titled: “Genotype-guided dosing of opioid agonists,” issued 26 January 2021. All other authors declare they have nothing to disclose.

Ethics approval and consent to participate

Animal studies were performed in accordance with NIH guidelines and were approved by the Institutional Animal Care and Use Committee of the University of Maryland Baltimore (Protocol # 0522009). The central Veterans Affairs (VA) institutional review board (IRB) approved the Million Veteran Program study. All relevant ethical regulations for work with human subjects were followed in the conduct of the study and informed consent was obtained from all participants.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wildermuth, E., Patton, M.S., Cortes-Gutierrez, M. et al. A single-cell genomic atlas for the effects of chronic ethanol exposure in the mouse dorsal striatum. Mol Psychiatry 30, 4320–4333 (2025). https://doi.org/10.1038/s41380-025-03014-z

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue date:

  • DOI: https://doi.org/10.1038/s41380-025-03014-z

Search

Quick links