1
|
Mews P, Mason AV, Kirchner EG, Estill M, Nestler EJ. Cocaine-induced gene regulation in D1 and D2 neuronal ensembles of the nucleus accumbens. Commun Biol 2025; 8:919. [PMID: 40506501 PMCID: PMC12163090 DOI: 10.1038/s42003-025-08327-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 06/02/2025] [Indexed: 06/16/2025] Open
Abstract
Cocaine use disorder is characterized by persistent drug-seeking behavior and a high risk of relapse, driven in part by lasting molecular and circuit adaptations in the nucleus accumbens. To explore the transcriptomic changes underlying these alterations, we employed fluorescence-activated nucleus sorting coupled with single-nucleus RNA sequencing to analyze D1 and D2 medium spiny neurons in this brain region of male mice subjected to acute cocaine exposure or to prolonged withdrawal from repeated cocaine exposure without or with an acute cocaine rechallenge. This approach allowed us to precisely delineate and contrast transcriptionally distinct neuronal subpopulations─or ensembles─across various treatment conditions. We identified significant heterogeneity within both D1 and D2 MSNs, revealing distinct clusters with unique transcriptional profiles. Notably, we identified a discrete D1 MSN population characterized by the upregulation of immediate early genes, as well as another group of D1 MSNs linked to prolonged withdrawal, uncovering novel regulators of withdrawal-related transcriptome dynamics. Our findings provide a high-resolution transcriptomic map of D1 and D2 MSNs, illustrating the dynamic changes induced by cocaine exposure and withdrawal. These insights into the molecular mechanisms underlying cocaine use disorder highlight potential targets for therapeutic intervention aimed at preventing relapse.
Collapse
Affiliation(s)
- Philipp Mews
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Pharmacology, Physiology, and Biophysics, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA.
| | - Autumn Va Mason
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pharmacology, Physiology, and Biophysics, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Emily G Kirchner
- Department of Pharmacology, Physiology, and Biophysics, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Molly Estill
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Eric J Nestler
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
2
|
Wang B, Wang J, Beacher NJ, Lin DT, Zhang Y. Cell-type specific epigenetic and transcriptional mechanisms in substance use disorder. Front Cell Neurosci 2025; 19:1552032. [PMID: 40226298 PMCID: PMC11985801 DOI: 10.3389/fncel.2025.1552032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Accepted: 03/13/2025] [Indexed: 04/15/2025] Open
Abstract
Substance use disorder (SUD) is a chronic and relapse-prone neuropsychiatric disease characterized by impaired brain circuitry within multiple cell types and neural circuits. Recent advancements in single-cell transcriptomics, epigenetics, and neural circuit research have unveiled molecular and cellular alterations associated with SUD. These studies have provided valuable insights into the transcriptional and epigenetic regulation of neuronal and non-neuronal cells, particularly in the context of drug exposure. Critical factors influencing the susceptibility of individuals to SUD include the regulation of gene expression during early developmental stages, neuroadaptive responses to psychoactive substances, and gene-environment interactions. Here we briefly review some of these mechanisms underlying SUD, with an emphasis on their crucial roles in in neural plasticity and maintenance of addiction and relapse in neuronal and non-neuronal cell-types. We foresee the possibility of integrating multi-omics technologies to devise targeted and personalized therapeutic strategies aimed at both the prevention and treatment of SUD. By utilizing these advanced methodologies, we can gain a deeper understanding of the fundamental biology of SUD, paving the way for more effective interventions.
Collapse
Affiliation(s)
- Bin Wang
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, China
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Jiale Wang
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, China
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Nicholas J. Beacher
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD, United States
| | - Da-Ting Lin
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD, United States
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Yan Zhang
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, China
| |
Collapse
|
3
|
Mews P, Van der Zee Y, Gurung A, Estill M, Futamura R, Kronman H, Ramakrishnan A, Ryan M, Reyes AA, Garcia BA, Browne CJ, Sidoli S, Shen L, Nestler EJ. Cell type-specific epigenetic priming of gene expression in nucleus accumbens by cocaine. SCIENCE ADVANCES 2024; 10:eado3514. [PMID: 39365860 PMCID: PMC11451531 DOI: 10.1126/sciadv.ado3514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 09/03/2024] [Indexed: 10/06/2024]
Abstract
A hallmark of addiction is the ability of drugs of abuse to trigger relapse after periods of prolonged abstinence. Here, we describe an epigenetic mechanism whereby chronic cocaine exposure causes lasting chromatin and downstream transcriptional modifications in the nucleus accumbens (NAc), a critical brain region controlling motivation. We link prolonged withdrawal from cocaine to the depletion of the histone variant H2A.Z, coupled with increased genome accessibility and latent priming of gene transcription, in D1 dopamine receptor-expressing medium spiny neurons (D1 MSNs) that relate to aberrant gene expression upon drug relapse. The histone chaperone ANP32E removes H2A.Z from chromatin, and we demonstrate that D1 MSN-selective Anp32e knockdown prevents cocaine-induced H2A.Z depletion and blocks cocaine's rewarding actions. By contrast, very different effects of cocaine exposure, withdrawal, and relapse were found for D2 MSNs. These findings establish histone variant exchange as an important mechanism and clinical target engaged by drugs of abuse to corrupt brain function and behavior.
Collapse
Affiliation(s)
- Philipp Mews
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yentl Van der Zee
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ashik Gurung
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Molly Estill
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Rita Futamura
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hope Kronman
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Aarthi Ramakrishnan
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Meagan Ryan
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Abner A. Reyes
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Benjamin A. Garcia
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, USA
| | - Caleb J. Browne
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Simone Sidoli
- Department of Biochemistry, Albert Einstein College of Medticine, New York, NY, USA
| | - Li Shen
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Eric J. Nestler
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
4
|
Faillace MP, Ortiz J, Rocco L, Bernabeu R. Histone Methyltransferase G9a Plays an Essential Role on Nicotine Preference in Zebrafish. Mol Neurobiol 2024; 61:6245-6263. [PMID: 38289455 DOI: 10.1007/s12035-024-03961-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 01/12/2024] [Indexed: 08/22/2024]
Abstract
Psychostimulants regulate behavioral responses in zebrafish via epigenetic mechanisms. We have previously shown that DNA methylation and histone deacetylase (HDAC) inhibition abolish nicotine-induced conditioned place preference (CPP) but little is known about the role of histone methylation in addictive-like behaviors. To assess the influence of histone methylation on nicotine-CPP, zebrafish were treated with a histone (H3) lysine-9 (K9) dimethyltransferase G9a/GLP inhibitor, BIX-01294 (BIX), which was administered before conditioning sessions. We observed a dual effect of the inhibitor BIX: at high doses inhibited while at low doses potentiated nicotine reward. Transcriptional expression of α6 and α7 subunits of the nicotinic acetylcholine receptor and of G9a, DNA methyl transferase-3, and HDAC-1 was upregulated in zebrafish with positive scores for nicotine-CPP. Changes in relative levels of these mRNA molecules reflected the effects of BIX on nicotine reward. BIX treatment per sé did not affect transcriptional levels of epigenetic enzymes that regulate trimethylation or demethylation of H3. BIX reduced H3K9me2 protein levels in a dose-dependent manner in key structures of the reward pathway. Thus, our findings indicated that different doses of BIX differentially affect nicotine CPP via strong or weak inhibition of G9a/GLP activity. Additionally, we found that the lysine demethylase inhibitor daminozide abolished nicotine-CPP and drug seeking. Our data demonstrate that H3 methylation catalyzed by G9a/GLP is involved in nicotine-CPP induction. Dimethylation of K9 at H3 is an important epigenetic modification that should be considered as a potential therapeutic target to treat nicotine reward and perhaps other drug addictions.
Collapse
Affiliation(s)
- Maria Paula Faillace
- Department of Physiology and Institute of Physiology and Biophysics (IFIBIO-Houssay, UBA-CONICET), School of Medicine, University of Buenos Aires, Paraguay 2155 7th floor (C1121ABG), Ciudad Autónoma de Buenos Aires, Argentina.
| | - Joaquin Ortiz
- Department of Physiology and Institute of Physiology and Biophysics (IFIBIO-Houssay, UBA-CONICET), School of Medicine, University of Buenos Aires, Paraguay 2155 7th floor (C1121ABG), Ciudad Autónoma de Buenos Aires, Argentina
| | - Leandro Rocco
- Department of Physiology and Institute of Physiology and Biophysics (IFIBIO-Houssay, UBA-CONICET), School of Medicine, University of Buenos Aires, Paraguay 2155 7th floor (C1121ABG), Ciudad Autónoma de Buenos Aires, Argentina
| | - Ramon Bernabeu
- Department of Physiology and Institute of Physiology and Biophysics (IFIBIO-Houssay, UBA-CONICET), School of Medicine, University of Buenos Aires, Paraguay 2155 7th floor (C1121ABG), Ciudad Autónoma de Buenos Aires, Argentina.
| |
Collapse
|
5
|
De Clerck M, Manguin M, Henkous N, d’Almeida MN, Beracochea D, Mons N. Chronic alcohol-induced long-lasting working memory deficits are associated with altered histone H3K9 dimethylation in the prefrontal cortex. Front Behav Neurosci 2024; 18:1354390. [PMID: 38495426 PMCID: PMC10941761 DOI: 10.3389/fnbeh.2024.1354390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 02/19/2024] [Indexed: 03/19/2024] Open
Abstract
Introduction Epigenetic modifications have emerged as key contributors to the enduring behavioral, molecular and epigenetic neuroadaptations during withdrawal from chronic alcohol exposure. The present study investigated the long-term consequences of chronic alcohol exposure on spatial working memory (WM) and associated changes of transcriptionally repressive histone H3 lysine 9 dimethylation (H3K9me2) in the prefrontal cortex (PFC). Methods Male C57BL/6 mice were allowed free access to either 12% (v/v) ethanol for 5 months followed by a 3-week abstinence period or water. Spatial WM was assessed through the spontaneous alternation T-maze test. Alcoholic and water mice received daily injections of GABAB agonist baclofen or saline during alcohol fading and early withdrawal. Global levels of histone modifications were determined by immunohistochemistry. Results Withdrawal mice displayed WM impairments along with reduced prefrontal H3K9me2 levels, compared to water-drinking mice. The withdrawal-induced decrease of H3K9me2 occurred concomitantly with increased level of permissive H3K9 acetylation (H3K9ac) in the PFC. Baclofen treatment rescued withdrawal-related WM deficits and fully restored prefrontal H3K9me2 and H3K9ac. Alcohol withdrawal induced brain region-specific changes of H3K9me2 and H3K9ac after testing, with significant decreases of both histone marks in the dorsal hippocampus and no changes in the amygdala and dorsal striatum. Furthermore, the magnitude of H3K9me2 in the PFC, but not the hippocampus, significantly and positively correlated with individual WM performances. No correlation was observed between H3K9ac and behavioral performance. Results also indicate that pre-testing intraperitoneal injection of UNC0642, a selective inhibitor of histone methyltransferase G9a responsible for H3K9me2, led to WM impairments in water-drinking and withdrawal-baclofen mice. Collectively, our results demonstrate that alcohol withdrawal induced brain-region specific alterations of H3K9me2 and H3K9ac, an effect that persisted for at least three weeks after cessation of chronic alcohol intake. Conclusion The findings suggest a role for long-lasting decreased H3K9me2 specifically in the PFC in the persistent WM impairments related to alcohol withdrawal.
Collapse
|
6
|
Anderson AG, Kulkarni A, Konopka G. A single-cell trajectory atlas of striatal development. Sci Rep 2023; 13:9031. [PMID: 37270616 PMCID: PMC10239471 DOI: 10.1038/s41598-023-36255-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 05/31/2023] [Indexed: 06/05/2023] Open
Abstract
The striatum integrates dense neuromodulatory inputs from many brain regions to coordinate complex behaviors. This integration relies on the coordinated responses from distinct striatal cell types. While previous studies have characterized the cellular and molecular composition of the striatum using single-cell RNA-sequencing at distinct developmental timepoints, the molecular changes spanning embryonic through postnatal development at the single-cell level have not been examined. Here, we combine published mouse striatal single-cell datasets from both embryonic and postnatal timepoints to analyze the developmental trajectory patterns and transcription factor regulatory networks within striatal cell types. Using this integrated dataset, we found that dopamine receptor-1 expressing spiny projection neurons have an extended period of transcriptional dynamics and greater transcriptional complexity over postnatal development compared to dopamine receptor-2 expressing neurons. Moreover, we found the transcription factor, FOXP1, exerts indirect changes to oligodendrocytes. These data can be accessed and further analyzed through an interactive website ( https://mouse-striatal-dev.cells.ucsc.edu ).
Collapse
Affiliation(s)
- Ashley G Anderson
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Ashwinikumar Kulkarni
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX, 75390-9111, USA
- Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Genevieve Konopka
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX, 75390-9111, USA.
- Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|
7
|
Choi EY, Franco D, Stapf CA, Gordin M, Chow A, Cover KK, Chandra R, Lobo MK. Inducible CRISPR Epigenome Systems Mimic Cocaine Induced Bidirectional Regulation of Nab2 and Egr3. J Neurosci 2023; 43:2242-2259. [PMID: 36849419 PMCID: PMC10072301 DOI: 10.1523/jneurosci.1802-22.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 12/06/2022] [Accepted: 12/22/2022] [Indexed: 03/01/2023] Open
Abstract
Substance use disorder is a chronic disease and a leading cause of disability around the world. The NAc is a major brain hub mediating reward behavior. Studies demonstrate exposure to cocaine is associated with molecular and functional imbalance in NAc medium spiny neuron subtypes (MSNs), dopamine receptor 1 and 2 enriched D1-MSNs and D2-MSNs. We previously reported repeated cocaine exposure induced transcription factor early growth response 3 (Egr3) mRNA in NAc D1-MSNs, and reduced it in D2-MSNs. Here, we report our findings of repeated cocaine exposure in male mice inducing MSN subtype-specific bidirectional expression of the Egr3 corepressor NGFI-A-binding protein 2 (Nab2). Using CRISPR activation and interference (CRISPRa and CRISPRi) tools combined with Nab2 or Egr3-targeted sgRNAs, we mimicked these bidirectional changes in Neuro2a cells. Furthermore, we investigated D1-MSN- and D2-MSN-specific expressional changes of histone lysine demethylases Kdm1a, Kdm6a, and Kdm5c in NAc after repeated cocaine exposure in male mice. Since Kdm1a showed bidirectional expression patterns in D1-MSNs and D2-MSNs, like Egr3, we developed a light-inducible Opto-CRISPR-KDM1a system. We were able to downregulate Egr3 and Nab2 transcripts in Neuro2A cells and cause similar bidirectional expression changes we observed in D1-MSNs and D2-MSNs of mouse repeated cocaine exposure model. Contrastingly, our Opto-CRISPR-p300 activation system induced the Egr3 and Nab2 transcripts and caused opposite bidirectional transcription regulations. Our study sheds light on the expression patterns of Nab2 and Egr3 in specific NAc MSNs in cocaine action and uses CRISPR tools to further mimic these expression patterns.SIGNIFICANCE STATEMENT Substance use disorder is a major societal issue. The lack of medication to treat cocaine addiction desperately calls for a treatment development based on precise understanding of molecular mechanisms underlying cocaine addiction. In this study, we show that Egr3 and Nab2 are bidirectionally regulated in mouse NAc D1-MSNs and D2-MSNs after repeated exposure to cocaine. Furthermore, histone lysine demethylations enzymes with putative EGR3 binding sites showed bidirectional regulation in D1- and D2-MSNs after repeated exposure to cocaine. Using Cre- and light-inducible CRISPR tools, we show that we can mimic this bidirectional regulation of Egr3 and Nab2 in Neuro2a cells.
Collapse
Affiliation(s)
- Eric Y Choi
- Department of Anatomy and Neurobiology
- Graduate Program in Life Sciences, Biochemistry and Molecular Biology
| | - Daniela Franco
- Department of Anatomy and Neurobiology
- Program in Neuroscience, Graduate Program in Life Sciences
| | - Catherine A Stapf
- Department of Anatomy and Neurobiology
- Program in Neuroscience, Graduate Program in Life Sciences
| | | | | | - Kara K Cover
- Department of Anatomy and Neurobiology
- Program in Neuroscience, Graduate Program in Life Sciences
| | - Ramesh Chandra
- Department of Anatomy and Neurobiology
- Center for Innovative Biomedical Resources, Virus Vector Core, University of Maryland School of Medicine Baltimore, Maryland, 21201
| | | |
Collapse
|
8
|
Safrole oxide induced 5-HT neuron-like cell differentiation of bone marrow mesenchymal stem cells by elevating G9a. In Vitro Cell Dev Biol Anim 2022; 58:513-520. [PMID: 35913528 DOI: 10.1007/s11626-021-00594-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 05/17/2021] [Indexed: 11/05/2022]
Abstract
In our previous study, we found that safrole oxide (SFO) could induce bone marrow mesenchymal stem cell differentiation into neuron-like cells. However, which kind of neuron cells was induced by SFO was unknown. Here, we found that SFO could induce BMSC differentiation into 5-hydroxytryptamine (5-HT) neuron-like cells. Microarray analysis of BMSCs treated with SFO for 6 h revealed a total of 35 genes changed more than twice. We selected G9a, a histone methyltransferase for further study. The upregulation of G9a was confirmed by RT-PCR and Western blot analysis. Small interfering RNA knockdown of G9a blocked SFO-induced BMSC differentiation. These results demonstrated that G9a was the pivotal factor in SFO-medicated 5-HT neuronal differentiation of BMSCs. Our findings provide a new clue for further investigating the gene control of BMSC differentiation into 5-HT neuron-like cells and provide a putative strategy for depression diseases therapies.
Collapse
|
9
|
Anderson EM, Taniguchi M. Epigenetic Effects of Addictive Drugs in the Nucleus Accumbens. Front Mol Neurosci 2022; 15:828055. [PMID: 35813068 PMCID: PMC9260254 DOI: 10.3389/fnmol.2022.828055] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 05/30/2022] [Indexed: 12/28/2022] Open
Abstract
Substance use induces long-lasting behavioral changes and drug craving. Increasing evidence suggests that epigenetic gene regulation contributes to the development and expression of these long-lasting behavioral alterations. Here we systematically review extensive evidence from rodent models of drug-induced changes in epigenetic regulation and epigenetic regulator proteins. We focus on histone acetylation and histone methylation in a brain region important for drug-related behaviors: the nucleus accumbens. We also discuss how experimentally altering these epigenetic regulators via systemically administered compounds or nucleus accumbens-specific manipulations demonstrate the importance of these proteins in the behavioral effects of drugs and suggest potential therapeutic value to treat people with substance use disorder. Finally, we discuss limitations and future directions for the field of epigenetic studies in the behavioral effects of addictive drugs and suggest how to use these insights to develop efficacious treatments.
Collapse
|
10
|
Establishment of H3K9-methylated heterochromatin and its functions in tissue differentiation and maintenance. Nat Rev Mol Cell Biol 2022; 23:623-640. [PMID: 35562425 PMCID: PMC9099300 DOI: 10.1038/s41580-022-00483-w] [Citation(s) in RCA: 236] [Impact Index Per Article: 78.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/30/2022] [Indexed: 12/14/2022]
Abstract
Heterochromatin is characterized by dimethylated or trimethylated histone H3 Lys9 (H3K9me2 or H3K9me3, respectively) and is found at transposable elements, satellite repeats and genes, where it ensures their transcriptional silencing. The histone methyltransferases (HMTs) that methylate H3K9 — in mammals Suppressor of variegation 3–9 homologue 1 (SUV39H1), SUV39H2, SET domain bifurcated 1 (SETDB1), SETDB2, G9A and G9A-like protein (GLP) — and the ‘readers’ of H3K9me2 or H3K9me3 are highly conserved and show considerable redundancy. Despite their redundancy, genetic ablation or mistargeting of an individual H3K9 methyltransferase can correlate with impaired cell differentiation, loss of tissue identity, premature aging and/or cancer. In this Review, we discuss recent advances in understanding the roles of the known H3K9-specific HMTs in ensuring transcriptional homeostasis during tissue differentiation in mammals. We examine the effects of H3K9-methylation-dependent gene repression in haematopoiesis, muscle differentiation and neurogenesis in mammals, and compare them with mechanistic insights obtained from the study of model organisms, notably Caenorhabditis elegans and Drosophila melanogaster. In all these organisms, H3K9-specific HMTs have both unique and redundant roles that ensure the maintenance of tissue integrity by restricting the binding of transcription factors to lineage-specific promoters and enhancer elements. Histone H3 Lys9 (H3K9)-methylated heterochromatin ensures transcriptional silencing of repetitive elements and genes, and its deregulation leads to impaired cell and tissue identity, premature aging and cancer. Recent studies in mammals clarified the roles H3K9-specific histone methyltransferases in ensuring transcriptional homeostasis during tissue differentiation.
Collapse
|
11
|
Anderson EM, Lopez MF, Kastner A, Mulholland PJ, Becker HC, Cowan CW. The histone methyltransferase G9a mediates stress-regulated alcohol drinking. Addict Biol 2022; 27:e13060. [PMID: 34013595 PMCID: PMC8602448 DOI: 10.1111/adb.13060] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 03/23/2021] [Accepted: 05/04/2021] [Indexed: 01/03/2023]
Abstract
The epigenetic enzyme G9a is a histone methyltransferase that dimethylates lysine 9 on histone H3 (H3K9me2), and in the adult nucleus accumbens (NAc), G9a regulates multiple behaviors associated with substance use disorder. We show here that chronic intermittent ethanol (CIE) exposure in male mice reduced both G9a and H3K9me2 levels in the adult NAc, but not dorsal striatum. Viral-mediated reduction of G9a in the NAc had no effects on baseline volitional ethanol drinking or escalated alcohol drinking produced by CIE exposure; however, NAc G9a was required for stress-regulated changes in ethanol drinking, including potentiated alcohol drinking produced by activation of the kappa-opioid receptor. In addition, we observed that chronic systemic administration of a G9a inhibitor, UNC0642, also blocked stress-potentiated alcohol drinking. Together, our findings suggest that chronic alcohol use, similar to other abused substances, produces a NAc-selective reduction in G9a levels that serves to limit stress-regulated alcohol drinking. Moreover, our findings suggest that pharmacological inhibition of G9a might provide a novel therapeutic approach to treat stress-induced alcohol drinking, which is a major trigger of relapse in individuals suffering from AUD.
Collapse
Affiliation(s)
- Ethan M. Anderson
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC
- Department of Psychiatry and Behavioral Sciences, Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC
| | - Marcelo F. Lopez
- Department of Psychiatry and Behavioral Sciences, Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC
| | - Abigail Kastner
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC
- Department of Psychiatry and Behavioral Sciences, Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC
| | - Patrick J. Mulholland
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC
- Department of Psychiatry and Behavioral Sciences, Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC
| | - Howard C. Becker
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC
- Department of Psychiatry and Behavioral Sciences, Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC
| | - Christopher W. Cowan
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC
- Department of Psychiatry and Behavioral Sciences, Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC
| |
Collapse
|
12
|
Poulard C, Noureddine LM, Pruvost L, Le Romancer M. Structure, Activity, and Function of the Protein Lysine Methyltransferase G9a. Life (Basel) 2021; 11:life11101082. [PMID: 34685453 PMCID: PMC8541646 DOI: 10.3390/life11101082] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 10/08/2021] [Accepted: 10/08/2021] [Indexed: 12/17/2022] Open
Abstract
G9a is a lysine methyltransferase catalyzing the majority of histone H3 mono- and dimethylation at Lys-9 (H3K9), responsible for transcriptional repression events in euchromatin. G9a has been shown to methylate various lysine residues of non-histone proteins and acts as a coactivator for several transcription factors. This review will provide an overview of the structural features of G9a and its paralog called G9a-like protein (GLP), explore the biochemical features of G9a, and describe its post-translational modifications and the specific inhibitors available to target its catalytic activity. Aside from its role on histone substrates, the review will highlight some non-histone targets of G9a, in order gain insight into their role in specific cellular mechanisms. Indeed, G9a was largely described to be involved in embryonic development, hypoxia, and DNA repair. Finally, the involvement of G9a in cancer biology will be presented.
Collapse
Affiliation(s)
- Coralie Poulard
- Cancer Research Cancer of Lyon, Université de Lyon, F-69000 Lyon, France; (L.M.N.); (L.P.); (M.L.R.)
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France
- Correspondence:
| | - Lara M. Noureddine
- Cancer Research Cancer of Lyon, Université de Lyon, F-69000 Lyon, France; (L.M.N.); (L.P.); (M.L.R.)
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France
- Laboratory of Cancer Biology and Molecular Immunology, Faculty of Sciences, Lebanese University, Hadat-Beirut 90565, Lebanon
| | - Ludivine Pruvost
- Cancer Research Cancer of Lyon, Université de Lyon, F-69000 Lyon, France; (L.M.N.); (L.P.); (M.L.R.)
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France
| | - Muriel Le Romancer
- Cancer Research Cancer of Lyon, Université de Lyon, F-69000 Lyon, France; (L.M.N.); (L.P.); (M.L.R.)
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France
| |
Collapse
|
13
|
McCullough KM, Missig G, Robbie MA, Foilb AR, Wells AM, Hartmann J, Anderson KJ, Neve RL, Nestler EJ, Ressler KJ, Carlezon WA. Nucleus Accumbens Medium Spiny Neuron Subtypes Differentially Regulate Stress-Associated Alterations in Sleep Architecture. Biol Psychiatry 2021; 89:1138-1149. [PMID: 33715826 PMCID: PMC8178228 DOI: 10.1016/j.biopsych.2020.12.030] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 12/17/2020] [Accepted: 12/20/2020] [Indexed: 01/04/2023]
Abstract
BACKGROUND Stress is implicated in the pathophysiology of major depression and posttraumatic stress disorder. These conditions share core features, including motivational deficits, heighted anxiety, and sleep dysregulation. Chronic stress produces these same features in rodents, with some individuals being susceptible or resilient, as seen in humans. While stress-induced neuroadaptations within the nucleus accumbens are implicated in susceptibility-related dysregulation of motivational and emotional behaviors, their effects on sleep are unclear. METHODS We used chemogenetics (DREADDs [designer receptors exclusively activated by designer drugs]) to examine the effects of selective alterations in activity of nucleus accumbens medium spiny neurons expressing dopamine D1 receptors (D1-MSNs) or dopamine D2 receptors (D2-MSNs) on sleep-related end points. Mice were implanted with wireless transmitters enabling continuous collection of data to quantify vigilance states over a 20-day test period. Parallel cohorts were examined in behavioral tests assessing stress susceptibility. RESULTS D1- and D2-MSNs play dissociable roles in sleep regulation. Stimulation of inhibitory or excitatory DREADDs expressed in D1-MSNs exclusively affects rapid eye movement sleep, whereas targeting D2-MSNs affects slow wave sleep. The combined effects of D1-MSN inhibition and D2-MSN activation on sleep resemble those of chronic social defeat stress. Alterations in D1-MSN function also affect stress susceptibility in social behavior tests. Elevation of CREB (cAMP response element-binding protein) within D1-MSNs is sufficient to produce stress-like effects on rapid eye movement sleep. CONCLUSIONS In addition to regulation of motivational and emotional behaviors, the nucleus accumbens also influences sleep, an end point with high translational relevance. These findings provide a neural basis for comorbidity in key features of stress-related illness.
Collapse
Affiliation(s)
- Kenneth M. McCullough
- Basic Neuroscience Division, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA
| | - Galen Missig
- Basic Neuroscience Division, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA
| | - Mykel A. Robbie
- Basic Neuroscience Division, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA
| | - Allison R. Foilb
- Basic Neuroscience Division, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA
| | - Audrey M. Wells
- Basic Neuroscience Division, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA
| | - Jakob Hartmann
- Basic Neuroscience Division, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA
| | - Kasey J. Anderson
- Basic Neuroscience Division, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA
| | - Rachael L. Neve
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Eric J. Nestler
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai. New York, NY 10029, USA
| | - Kerry J. Ressler
- Basic Neuroscience Division, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA
| | - William A. Carlezon
- Basic Neuroscience Division, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA,Corresponding Author: William A. Carlezon, Jr., Ph.D., Department of Psychiatry, McLean Hospital, 115 Mill Street, Belmont, MA 02478, USA,
| |
Collapse
|
14
|
Anderson AG, Kulkarni A, Harper M, Konopka G. Single-Cell Analysis of Foxp1-Driven Mechanisms Essential for Striatal Development. Cell Rep 2021; 30:3051-3066.e7. [PMID: 32130906 PMCID: PMC7137930 DOI: 10.1016/j.celrep.2020.02.030] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 01/16/2020] [Accepted: 02/07/2020] [Indexed: 12/30/2022] Open
Abstract
The striatum is a critical forebrain structure integrating cognitive, sensory, and motor information from diverse brain regions into meaningful behavioral output. However, the transcriptional mechanisms underlying striatal development at single-cell resolution remain unknown. Using single-cell RNA sequencing (RNA-seq), we examine the cellular diversity of the early postnatal striatum and show that Foxp1, a transcription factor strongly linked to autism and intellectual disability, regulates the cellular composition, neurochemical architecture, and connectivity of the striatum in a cell-type-dependent fashion. We also identify Foxp1-regulated target genes within distinct cell types and connect these molecular changes to functional and behavioral deficits relevant to phenotypes described in patients with FOXP1 loss-of-function mutations. Using this approach, we could also examine the non-cell-autonomous effects produced by disrupting one cell type and the molecular compensation that occurs in other populations. These data reveal the cell-type-specific transcriptional mechanisms regulated by Foxp1 that underlie distinct features of striatal circuitry.
Collapse
Affiliation(s)
- Ashley G Anderson
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390-9111, USA
| | - Ashwinikumar Kulkarni
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390-9111, USA
| | - Matthew Harper
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390-9111, USA
| | - Genevieve Konopka
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390-9111, USA.
| |
Collapse
|
15
|
HDAC3 Activity within the Nucleus Accumbens Regulates Cocaine-Induced Plasticity and Behavior in a Cell-Type-Specific Manner. J Neurosci 2021; 41:2814-2827. [PMID: 33602824 PMCID: PMC8018887 DOI: 10.1523/jneurosci.2829-20.2021] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 01/04/2021] [Accepted: 02/03/2021] [Indexed: 01/19/2023] Open
Abstract
Epigenetic mechanisms regulate processes of neuroplasticity critical to cocaine-induced behaviors. This includes the Class I histone deacetylase (HDAC) HDAC3, known to act as a negative regulator of cocaine-associated memory formation within the nucleus accumbens (NAc). Despite this, it remains unknown how cocaine alters HDAC3-dependent mechanisms. Here, we profiled HDAC3 expression and activity in total NAc mouse tissue following cocaine exposure. Although chronic cocaine did not affect expression of Hdac3 within the NAc, chronic cocaine did affect promoter-specific changes in HDAC3 and H4K8Ac occupancy. These changes in promoter occupancy correlated with cocaine-induced changes in expression of plasticity-related genes. To causally determine whether cocaine-induced plasticity is mediated by HDAC3's deacetylase activity, we overexpressed a deacetylase-dead HDAC3 point mutant (HDAC3-Y298H-v5) within the NAc of adult male mice. We found that disrupting HDAC3's enzymatic activity altered selective changes in gene expression and synaptic plasticity following cocaine exposure, despite having no effects on cocaine-induced behaviors. In further assessing HDAC3's role within the NAc, we observed that chronic cocaine increases Hdac3 expression in Drd1 but not Drd2-cells of the NAc. Moreover, we discovered that HDAC3 acts selectively within D1R cell-types to regulate cocaine-associated memory formation and cocaine-seeking. Overall, these results suggest that cocaine induces cell-type-specific changes in epigenetic mechanisms to promote plasticity important for driving cocaine-related behaviors. SIGNIFICANCE STATEMENT Drugs of abuse alter molecular mechanisms throughout the reward circuitry that can lead to persistent drug-associated behaviors. Epigenetic regulators are critical drivers of drug-induced changes in gene expression. Here, we demonstrate that the activity of an epigenetic enzyme promotes neuroplasticity within the nucleus accumbens (NAc) critical to cocaine action. In addition, we demonstrate that these changes in epigenetic activity drive cocaine-seeking behaviors in a cell-type-specific manner. These findings are key in understanding and targeting cocaine's impact of neural circuitry and behavior.
Collapse
|
16
|
Mews P, Calipari ES, Day J, Lobo MK, Bredy T, Abel T. From Circuits to Chromatin: The Emerging Role of Epigenetics in Mental Health. J Neurosci 2021; 41:873-882. [PMID: 33446519 PMCID: PMC7880276 DOI: 10.1523/jneurosci.1649-20.2020] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 12/19/2020] [Accepted: 12/22/2020] [Indexed: 02/01/2023] Open
Abstract
A central goal of neuroscience research is to understand how experiences modify brain circuits to guide future adaptive behavior. In response to environmental stimuli, neural circuit activity engages gene regulatory mechanisms within each cell. This activity-dependent gene expression is governed, in part, by epigenetic processes that can produce persistent changes in both neural circuits and the epigenome itself. The complex interplay between circuit activity and neuronal gene regulation is vital to learning and memory, and, when disrupted, is linked to debilitating psychiatric conditions, such as substance use disorder. To develop clinical treatments, it is paramount to advance our understanding of how neural circuits and the epigenome cooperate to produce behavioral adaptation. Here, we discuss how new genetic tools, used to manipulate neural circuits and chromatin, have enabled the discovery of epigenetic processes that bring about long-lasting changes in behavior relevant to mental health and disease.
Collapse
Affiliation(s)
- Philipp Mews
- Friedman Brain Institute, Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York 10129
| | - Erin S Calipari
- Departments of Pharmacology, Molecular Physiology and Biophysics, Psychiatry and Behavioral Sciences; Vanderbilt Center for Addiction Research; Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee 37323
| | - Jeremy Day
- Department of Neurobiology, McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Mary Kay Lobo
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Timothy Bredy
- Queensland Brain Institute, University of Queensland, Brisbane, 4072, Australia
| | - Ted Abel
- Department of Neuroscience and Pharmacology, Iowa Neuroscience Institute, University of Iowa, Iowa City, Iowa 52242
| |
Collapse
|
17
|
FOXP1 negatively regulates intrinsic excitability in D2 striatal projection neurons by promoting inwardly rectifying and leak potassium currents. Mol Psychiatry 2021; 26:1761-1774. [PMID: 33402705 PMCID: PMC8255328 DOI: 10.1038/s41380-020-00995-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 11/18/2020] [Accepted: 12/09/2020] [Indexed: 01/13/2023]
Abstract
Heterozygous loss-of-function mutations in the transcription factor FOXP1 are strongly associated with autism. Dopamine receptor 2 expressing (D2) striatal projection neurons (SPNs) in heterozygous Foxp1 (Foxp1+/-) mice have higher intrinsic excitability. To understand the mechanisms underlying this alteration, we examined SPNs with cell-type specific homozygous Foxp1 deletion to study cell-autonomous regulation by Foxp1. As in Foxp1+/- mice, D2 SPNs had increased intrinsic excitability with homozygous Foxp1 deletion. This effect involved postnatal mechanisms. The hyperexcitability was mainly due to down-regulation of two classes of potassium currents: inwardly rectifying (KIR) and leak (KLeak). Single-cell RNA sequencing data from D2 SPNs with Foxp1 deletion indicated the down-regulation of transcripts of candidate ion channels that may underlie these currents: Kcnj2 and Kcnj4 for KIR and Kcnk2 for KLeak. This Foxp1-dependent regulation was neuron-type specific since these same currents and transcripts were either unchanged, or very little changed, in D1 SPNs with cell-specific Foxp1 deletion. Our data are consistent with a model where FOXP1 negatively regulates the excitability of D2 SPNs through KIR and KLeak by transcriptionally activating their corresponding transcripts. This, in turn, provides a novel example of how a transcription factor may regulate multiple genes to impact neuronal electrophysiological function that depends on the integration of multiple current types - and do this in a cell-specific fashion. Our findings provide initial clues to altered neuronal function and possible therapeutic strategies not only for FOXP1-associated autism but also for other autism forms associated with transcription factor dysfunction.
Collapse
|
18
|
Jan S, Dar MI, Wani R, Sandey J, Mushtaq I, Lateef S, Syed SH. Targeting EHMT2/ G9a for cancer therapy: Progress and perspective. Eur J Pharmacol 2020; 893:173827. [PMID: 33347828 DOI: 10.1016/j.ejphar.2020.173827] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 12/12/2020] [Accepted: 12/16/2020] [Indexed: 12/11/2022]
Abstract
Euchromatic histone lysine methyltransferase-2, also known as G9a, is a ubiquitously expressed SET domain-containing histone lysine methyltransferase linked with both facultative and constitutive heterochromatin formation and transcriptional repression. It is an essential developmental gene and reported to play role in embryonic development, establishment of proviral silencing in ES cells, tumor cell growth, metastasis, T-cell immune response, cocaine induced neural plasticity and cognition and adaptive behavior. It is mainly responsible for carrying out mono, di and tri methylation of histone H3K9 in euchromatin. G9a levels are elevated in many cancers and its selective inhibition is known to reduce the cell growth and induce autophagy, apoptosis and senescence. We carried out a thorough search of online literature databases including Pubmed, Scopus, Journal websites, Clinical trials etc to gather the maximum possible information related to the G9a. The main messages from the cited papers are presented in a systematic manner. Chemical structures were drawn by Chemdraw software. In this review, we shed light on current understanding of structure and biological activity of G9a, the molecular events directing its targeting to genomic regions and its post-translational modification. Finally, we discuss the current strategies to target G9a in different cancers and evaluate the available compounds and agents used to inhibit G9a functions. The review provides the present status and future directions of research in targeting G9a and provides the basis to persuade the development of novel strategies to target G9a -related effects in cancer cells.
Collapse
Affiliation(s)
- Suraya Jan
- CSIR, Indian Institute of Integrative Medicine, Sanatnagar, 190005, Srinagar, Kashmir, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Mohd Ishaq Dar
- CSIR, Indian Institute of Integrative Medicine, Sanatnagar, 190005, Srinagar, Kashmir, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Rubiada Wani
- CSIR, Indian Institute of Integrative Medicine, Sanatnagar, 190005, Srinagar, Kashmir, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Jagjeet Sandey
- CSIR, Indian Institute of Integrative Medicine, Sanatnagar, 190005, Srinagar, Kashmir, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Iqra Mushtaq
- CSIR, Indian Institute of Integrative Medicine, Sanatnagar, 190005, Srinagar, Kashmir, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Sammar Lateef
- CSIR, Indian Institute of Integrative Medicine, Sanatnagar, 190005, Srinagar, Kashmir, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Sajad Hussain Syed
- CSIR, Indian Institute of Integrative Medicine, Sanatnagar, 190005, Srinagar, Kashmir, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
19
|
Blum K, Giordano J, Baron D, McLaughlin T, Badgaiyan RD. Proposing FDA consideration for the treatment and prophylaxis of opioid and psychostimulant abuse to incorporate the induction of DNA guided dopamine homeostasis: Anti-reward deficiency restoration solution (ARDS). JOURNAL OF SYSTEMS AND INTEGRATIVE NEUROSCIENCE 2020; 8:10.15761/JSIN.1000253. [PMID: 36407844 PMCID: PMC9670280 DOI: 10.15761/jsin.1000253] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
BACKGROUND In face of an American opioid/psychostimulant crisis with overdose fatalities, due, in part, to the COVOD 19 pandemic, we are proposing a paradigm shift in response. Currently, The FDA has approved pharmaceuticals for the treatment of opioids, alcohol, and nicotine but not for psychostimulants or even cannabis. PROPOSITION To respond to the deadly overdose issue globally, we are proposing that the FDA embrace, for the treatment and prophylaxis of opioid and psychostimulant abuse, induction of DNA-guided, dopamine homeostasis. We refer to this novel therapeutic target as the Anti-Reward Deficiency Restoration Solution (ARDS). EXPERT OPINION This futuristic proposal regarding the FDA will provide important information that may ultimately lead to significant improvement in the recovery of individuals with opioid/psychostimulant and polydrug abuse issues, especially, those with genetically-induced dopamine deficiency. CONCLUSION With large populations supporting these initial results, and possibly even additional candidate genes and single nucleotide polymorphisms, the neuroscience and neurological community may eventually have the clinical ability to classify addiction severity, according to genotype and possession of risk alleles. A promising goal is the identification of high risk vulnerability, along with the provision of a safe, non-addicting ARDS natural nutrigenomic, involving a therapeutic model that potentially up-regulates instead of down-regulates dopaminergic receptors, preferably, the D2 subtype, is one laudable goal.
Collapse
Affiliation(s)
- Kenneth Blum
- Western University Health Sciences, Graduate College, Pomona CA, USA
- The Kenneth Blum Behavioral & Neurogenetic Institute (Division of Ivitalize) Austin, TX, USA
| | - John Giordano
- The Kenneth Blum Behavioral & Neurogenetic Institute (Division of Ivitalize) Austin, TX, USA
| | - David Baron
- Western University Health Sciences, Graduate College, Pomona CA, USA
| | - Thomas McLaughlin
- The Kenneth Blum Behavioral & Neurogenetic Institute (Division of Ivitalize) Austin, TX, USA
| | - Rajendra D Badgaiyan
- Department of Psychiatry, South Texas Veteran Health Care System, Audie L. Murphy Memorial VA Hospital, San Antonio, TX, Long School of Medicine, University of Texas Medical Center, San Antonio, TX, USA
| |
Collapse
|
20
|
Sharma M, Sajikumar S. G9a/GLP Complex Acts as a Bidirectional Switch to Regulate Metabotropic Glutamate Receptor-Dependent Plasticity in Hippocampal CA1 Pyramidal Neurons. Cereb Cortex 2020; 29:2932-2946. [PMID: 29982412 DOI: 10.1093/cercor/bhy161] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 06/14/2018] [Accepted: 06/17/2018] [Indexed: 02/01/2023] Open
Abstract
Metabotropic glutamate receptor-dependent long-term depression (mGluR-LTD) is conventionally considered to be solely dependent on local protein synthesis. Given the impact of epigenetics on memory, the intriguing question is whether epigenetic regulation influences mGluR-LTD as well. G9a/GLP histone lysine methyltransferase complex is crucial for brain development and goal-directed learning as well as for drug-addiction. In this study, we analyzed whether the epigenetic regulation by G9a/GLP complex affects mGluR-LTD in CA1 hippocampal pyramidal neurons of 5-7 weeks old male Wistar rats. In hippocampal slices with intact CA1 dendritic regions, inhibition of G9a/GLP activity abolished mGluR-LTD. The inhibition of this complex upregulated the expression of plasticity proteins like PKMζ, which mediated the prevention of mGluR-LTD expression by regulating the NSF-GluA2-mediated trafficking of AMPA receptors towards the postsynaptic site. G9a/GLP inhibition during the induction of mGluR-LTD also downregulated the protein levels of phosphorylated-GluA2 and Arc. Interestingly, G9a/GLP inhibition could not impede the mGluR-LTD when the cell-body was severed. Our study highlights the role of G9a/GLP complex in intact neuronal network as a bidirectional switch; when turned on, it facilitates the expression of mGluR-LTD, and when turned off, it promotes the expression of long-term potentiation.
Collapse
Affiliation(s)
- Mahima Sharma
- Department of Physiology, National University of Singapore, 2 Medical Drive, MD9, Singapore, Singapore.,Neurobiology/Aging Programme, Life Sciences Institute, Centre for Life Sciences, 28 Medical Drive, Singapore, Singapore
| | - Sreedharan Sajikumar
- Department of Physiology, National University of Singapore, 2 Medical Drive, MD9, Singapore, Singapore.,Neurobiology/Aging Programme, Life Sciences Institute, Centre for Life Sciences, 28 Medical Drive, Singapore, Singapore
| |
Collapse
|
21
|
Li Q, Xiang YH, Liang XJ, Zhang Y, Zhao PP, Wang M, Bao XM, Zhu XB, Deng AC. Expression of G9a in Auditory Cortex Is Downregulated in a Rat Model of Age-Related Hearing Loss. J Mol Neurosci 2020; 71:409-418. [PMID: 32671696 DOI: 10.1007/s12031-020-01663-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 07/08/2020] [Indexed: 01/11/2023]
Abstract
G9a is essential for dendritic plasticity and is associated with neurological disorders. The possible relationship between age-related hearing loss and G9a expression in the auditory cortex has not been fully explored. This study aimed to understand the expression patterns of G9a-mediated histone methylations in the auditory cortex during aging. Using immunofluorescence and western blotting, we demonstrated that a significant reduction in G9a expression observed in the auditory cortex of 24-month-old rats compared to 3-month-old rats, was associated with remarkable hearing threshold elevation and hair cell loss. Correspondingly, histone H3 lysine 9 (H3K9) mono- and dimethylation (marked by H3K9me1 and H3K9me2, respectively), which were regulated by G9a activity, also evidently decreased during aging. These findings, which merit further investigation, suggest a possible association between G9a-mediated histone methylations and central age-related hearing disorders.
Collapse
Affiliation(s)
- Qian Li
- Department of Otolaryngology Head and Neck Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Yang-Hong Xiang
- Department of Otolaryngology Head and Neck Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Xiao-Jun Liang
- Department of Otolaryngology Head and Neck Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Yun Zhang
- Department of Otolaryngology Head and Neck Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Peng-Peng Zhao
- Department of Otolaryngology Head and Neck Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Min Wang
- Department of Otolaryngology Head and Neck Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Xiao-Min Bao
- Department of Otolaryngology Head and Neck Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Xian-Bai Zhu
- Department of Otolaryngology Head and Neck Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - An-Chun Deng
- Department of Otolaryngology Head and Neck Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China.
| |
Collapse
|
22
|
Soleilhavoup C, Travaglio M, Patrick K, Garção P, Boobalan E, Adolfs Y, Spriggs RV, Moles-Garcia E, Dhiraj D, Oosterveen T, Ferri SL, Abel T, Brodkin ES, Pasterkamp RJ, Brooks BP, Panman L. Nolz1 expression is required in dopaminergic axon guidance and striatal innervation. Nat Commun 2020; 11:3111. [PMID: 32561725 PMCID: PMC7305235 DOI: 10.1038/s41467-020-16947-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 05/29/2020] [Indexed: 11/24/2022] Open
Abstract
Midbrain dopaminergic (DA) axons make long longitudinal projections towards the striatum. Despite the importance of DA striatal innervation, processes involved in establishment of DA axonal connectivity remain largely unknown. Here we demonstrate a striatal-specific requirement of transcriptional regulator Nolz1 in establishing DA circuitry formation. DA projections are misguided and fail to innervate the striatum in both constitutive and striatal-specific Nolz1 mutant embryos. The lack of striatal Nolz1 expression results in nigral to pallidal lineage conversion of striatal projection neuron subtypes. This lineage switch alters the composition of secreted factors influencing DA axonal tract formation and renders the striatum non-permissive for dopaminergic and other forebrain tracts. Furthermore, transcriptomic analysis of wild-type and Nolz1-/- mutant striatal tissue led to the identification of several secreted factors that underlie the observed guidance defects and proteins that promote DA axonal outgrowth. Together, our data demonstrate the involvement of the striatum in orchestrating dopaminergic circuitry formation.
Collapse
Affiliation(s)
- Clement Soleilhavoup
- MRC Toxicology Unit, University of Cambridge, Hodgkin Building, Lancaster Road, Leicester, LE1 9HN, UK
| | - Marco Travaglio
- MRC Toxicology Unit, University of Cambridge, Hodgkin Building, Lancaster Road, Leicester, LE1 9HN, UK
| | - Kieran Patrick
- MRC Toxicology Unit, University of Cambridge, Hodgkin Building, Lancaster Road, Leicester, LE1 9HN, UK
| | - Pedro Garção
- MRC Toxicology Unit, University of Cambridge, Hodgkin Building, Lancaster Road, Leicester, LE1 9HN, UK
| | - Elangovan Boobalan
- Ophthalmic Genetics & Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Youri Adolfs
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Ruth V Spriggs
- MRC Toxicology Unit, University of Cambridge, Hodgkin Building, Lancaster Road, Leicester, LE1 9HN, UK
| | - Emma Moles-Garcia
- MRC Toxicology Unit, University of Cambridge, Hodgkin Building, Lancaster Road, Leicester, LE1 9HN, UK
| | - Dalbir Dhiraj
- MRC Toxicology Unit, University of Cambridge, Hodgkin Building, Lancaster Road, Leicester, LE1 9HN, UK
| | - Tony Oosterveen
- MRC Toxicology Unit, University of Cambridge, Hodgkin Building, Lancaster Road, Leicester, LE1 9HN, UK
| | - Sarah L Ferri
- Department of Neuroscience and Pharmacology, Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, 52242, USA
| | - Ted Abel
- Department of Neuroscience and Pharmacology, Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, 52242, USA
| | - Edward S Brodkin
- Center for Neurobiology and Behavior, Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104-3403, USA
| | - R Jeroen Pasterkamp
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Brian P Brooks
- Ophthalmic Genetics & Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Lia Panman
- MRC Toxicology Unit, University of Cambridge, Hodgkin Building, Lancaster Road, Leicester, LE1 9HN, UK.
| |
Collapse
|
23
|
Green TA, Bardo MT. Opposite regulation of conditioned place preference and intravenous drug self-administration in rodent models: Motivational and non-motivational examples. Neurosci Biobehav Rev 2020; 116:89-98. [PMID: 32534899 DOI: 10.1016/j.neubiorev.2020.06.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 06/01/2020] [Accepted: 06/03/2020] [Indexed: 12/13/2022]
Abstract
Although developed from a common antecedent, conditioned place preference (CPP) and intravenous drug self-administration (SA) represent different behavioral paradigms, each with strong face validity. The field has treated results from these studies largely interchangeably; however, there is considerable evidence of opposite modulation of CPP vs. SA. This review outlines four manipulations that differentially affect CPP and SA based on alterations of motivation. These examples are contrasted with one example of differential CPP and SA results that can be explained by simple parallel shifts in dose-response functions. The final two examples have yet to be classified as motivation-based or parallel shifts. Important aspects, including motivation, volitional control of drug administration, reward, and the role of cues are discussed. One major conclusion of this paper is that explanations for apparent discrepancies between CPP and SA require full dose effect functions and assessment of PR breakpoints. Overall, this manuscript offers a more nuanced insight into how CPP and SA can be used to study different aspects of substance use disorders.
Collapse
Affiliation(s)
- Thomas A Green
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, United States.
| | - Michael T Bardo
- Department of Psychology, University of Kentucky, United States.
| |
Collapse
|
24
|
Salery M, Trifilieff P, Caboche J, Vanhoutte P. From Signaling Molecules to Circuits and Behaviors: Cell-Type-Specific Adaptations to Psychostimulant Exposure in the Striatum. Biol Psychiatry 2020; 87:944-953. [PMID: 31928716 DOI: 10.1016/j.biopsych.2019.11.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 10/30/2019] [Accepted: 11/01/2019] [Indexed: 12/17/2022]
Abstract
Addiction is characterized by a compulsive pattern of drug seeking and consumption and a high risk of relapse after withdrawal that are thought to result from persistent adaptations within brain reward circuits. Drugs of abuse increase dopamine (DA) concentration in these brain areas, including the striatum, which shapes an abnormal memory trace of drug consumption that virtually highjacks reward processing. Long-term neuronal adaptations of gamma-aminobutyric acidergic striatal projection neurons (SPNs) evoked by drugs of abuse are critical for the development of addiction. These neurons form two mostly segregated populations, depending on the DA receptor they express and their output projections, constituting the so-called direct (D1 receptor) and indirect (D2 receptor) SPN pathways. Both SPN subtypes receive converging glutamate inputs from limbic and cortical regions, encoding contextual and emotional information, together with DA, which mediates reward prediction and incentive values. DA differentially modulates the efficacy of glutamate synapses onto direct and indirect SPN pathways by recruiting distinct striatal signaling pathways, epigenetic and genetic responses likely involved in the transition from casual drug use to addiction. Herein we focus on recent studies that have assessed psychostimulant-induced alterations in a cell-type-specific manner, from remodeling of input projections to the characterization of specific molecular events in each SPN subtype and their impact on long-lasting behavioral adaptations. We discuss recent evidence revealing the complex and concerted action of both SPN populations on drug-induced behavioral responses, as these studies can contribute to the design of future strategies to alleviate specific behavioral components of addiction.
Collapse
Affiliation(s)
- Marine Salery
- Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Pierre Trifilieff
- NutriNeuro, Unité Mixte de Recherche (UMR) 1286, Institut National de la Recherche Agronomique, Bordeaux Institut Polytechnique, University of Bordeaux, Bordeaux, France
| | - Jocelyne Caboche
- Neuroscience Paris Seine, Institut de Biologie Paris-Seine, Sorbonne Université, Faculty of Sciences, Paris, France; Centre National de la Recherche Scientifique, UMR8246, Paris, France; Institut National de la Santé et de la Recherche Médicale, U1130, Paris France.
| | - Peter Vanhoutte
- Neuroscience Paris Seine, Institut de Biologie Paris-Seine, Sorbonne Université, Faculty of Sciences, Paris, France; Centre National de la Recherche Scientifique, UMR8246, Paris, France; Institut National de la Santé et de la Recherche Médicale, U1130, Paris France
| |
Collapse
|
25
|
Prodromidou K, Matsas R. Species-Specific miRNAs in Human Brain Development and Disease. Front Cell Neurosci 2019; 13:559. [PMID: 31920559 PMCID: PMC6930153 DOI: 10.3389/fncel.2019.00559] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Accepted: 12/04/2019] [Indexed: 12/20/2022] Open
Abstract
Identification of the unique features of human brain development and function can be critical towards the elucidation of intricate processes such as higher cognitive functions and human-specific pathologies like neuropsychiatric and behavioral disorders. The developing primate and human central nervous system (CNS) are distinguished by expanded progenitor zones and a protracted time course of neurogenesis, leading to the expansion in brain size, prominent gyral anatomy, distinctive synaptic properties, and complex neural circuits. Comparative genomic studies have revealed that adaptations of brain capacities may be partly explained by human-specific genetic changes that impact the function of proteins associated with neocortical expansion, synaptic function, and language development. However, the formation of complex gene networks may be most relevant for brain evolution. Indeed, recent studies identified distinct human-specific gene expression patterns across developmental time occurring in brain regions linked to cognition. Interestingly, such modules show species-specific divergence and are enriched in genes associated with neuronal development and synapse formation whilst also being implicated in neuropsychiatric diseases. microRNAs represent a powerful component of gene-regulatory networks by promoting spatiotemporal post-transcriptional control of gene expression in the human and primate brain. It has also been suggested that the divergence in miRNA expression plays an important role in shaping gene expression divergence among species. Primate-specific and human-specific miRNAs are principally involved in progenitor proliferation and neurogenic processes but also associate with human cognition, and neurological disorders. Human embryonic or induced pluripotent stem cells and brain organoids, permitting experimental access to neural cells and differentiation stages that are otherwise difficult or impossible to reach in humans, are an essential means for studying species-specific brain miRNAs. Single-cell sequencing approaches can further decode refined miRNA-mRNA interactions during developmental transitions. Elucidating species-specific miRNA regulation will shed new light into the mechanisms that control spatiotemporal events during human brain development and disease, an important step towards fostering novel, holistic and effective therapeutic approaches for neural disorders. In this review, we discuss species-specific regulation of miRNA function, its contribution to the evolving features of the human brain and in neurological disease, with respect also to future therapeutic approaches.
Collapse
Affiliation(s)
- Kanella Prodromidou
- Laboratory of Cellular and Molecular Neurobiology-Stem Cells, Department of Neurobiology, Hellenic Pasteur Institute, Athens, Greece
| | - Rebecca Matsas
- Laboratory of Cellular and Molecular Neurobiology-Stem Cells, Department of Neurobiology, Hellenic Pasteur Institute, Athens, Greece
| |
Collapse
|
26
|
Camilo C, Maschietto M, Vieira HC, Tahira AC, Gouveia GR, Feio dos Santos AC, Negrão AB, Ribeiro M, Laranjeira R, Vallada H, Brentani H. Genome-wide DNA methylation profile in the peripheral blood of cocaine and crack dependents. REVISTA BRASILEIRA DE PSIQUIATRIA (SAO PAULO, BRAZIL : 1999) 2019; 41:485-493. [PMID: 31116258 PMCID: PMC6899365 DOI: 10.1590/1516-4446-2018-0092] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 10/23/2018] [Indexed: 12/30/2022]
Abstract
OBJECTIVE Cocaine use disorders (CUDs) represent a major public health problem in many countries. To better understand the interaction between the environmental modulations and phenotype, the aim of the present study was to investigate the DNA methylation pattern of CUD patients, who had concomitant cocaine and crack dependence, and healthy controls. METHODS We studied DNA methylation profiles in the peripheral blood of 23 CUD patients and 24 healthy control subjects using the Illumina Infinium HumanMethylation450 BeadChip arrays. RESULTS Comparison between CUD patients and controls revealed 186 differentially methylated positions (DMPs; adjusted p-value [adjP] < 10-5) related to 152 genes, with a subset of CpGs confirmed by pyrosequencing. DNA methylation patterns discriminated CUD patients and control groups. A gene network approach showed that the EHMT1, EHMT2, MAPK1, MAPK3, MAP2K1, and HDAC5 genes, which are involved in transcription and chromatin regulation cellular signaling pathways, were also associated with cocaine dependence. CONCLUSION The investigation of DNA methylation patterns may contribute to a better understanding of the biological mechanisms involved in CUD.
Collapse
Affiliation(s)
- Caroline Camilo
- Departamento e Instituto de Psiquiatria, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, SP, Brazil
| | - Mariana Maschietto
- Laboratório Nacional de Biociências (LNBio), Centro Nacional de Pesquisa em Energia e Materiais, Campinas, SP, Brazil
| | - Henrique C. Vieira
- Departamento e Instituto de Psiquiatria, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, SP, Brazil
| | - Ana C. Tahira
- Departamento e Instituto de Psiquiatria, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, SP, Brazil
| | - Gisele R. Gouveia
- Departamento e Instituto de Psiquiatria, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, SP, Brazil
| | - Ana C. Feio dos Santos
- Departamento e Instituto de Psiquiatria, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, SP, Brazil
| | - André B. Negrão
- Departamento e Instituto de Psiquiatria, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, SP, Brazil
| | - Marcelo Ribeiro
- Unidade de Pesquisa em Álcool e Drogas (UNIAD), Departamento de Psiquiatria, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Ronaldo Laranjeira
- Unidade de Pesquisa em Álcool e Drogas (UNIAD), Departamento de Psiquiatria, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Homero Vallada
- Departamento e Instituto de Psiquiatria, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, SP, Brazil
| | - Helena Brentani
- Departamento e Instituto de Psiquiatria, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, SP, Brazil
| |
Collapse
|
27
|
Anderson EM, Penrod RD, Barry SM, Hughes BW, Taniguchi M, Cowan CW. It is a complex issue: emerging connections between epigenetic regulators in drug addiction. Eur J Neurosci 2019; 50:2477-2491. [PMID: 30251397 DOI: 10.1111/ejn.14170] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 09/04/2018] [Accepted: 09/11/2018] [Indexed: 02/06/2023]
Abstract
Drug use leads to addiction in some individuals, but the underlying brain mechanisms that control the transition from casual drug use to an intractable substance use disorder (SUD) are not well understood. Gene x environment interactions such as the frequency of drug use and the type of substance used likely to promote maladaptive plastic changes in brain regions that are critical for controlling addiction-related behavior. Epigenetics encompasses a broad spectrum of mechanisms important for regulating gene transcription that are not dependent on changes in DNA base pair sequences. This review focuses on the proteins and complexes contributing to epigenetic modifications in the nucleus accumbens (NAc) following drug experience. We discuss in detail the three major mechanisms: histone acetylation and deacetylation, histone methylation, and DNA methylation. We discuss how drug use alters the regulation of the associated proteins regulating these processes and highlight how experimental manipulations of these proteins in the NAc can alter drug-related behaviors. Finally, we discuss the ways that histone modifications and DNA methylation coordinate actions by recruiting large epigenetic enzyme complexes to aid in transcriptional repression. Targeting these multiprotein epigenetic enzyme complexes - and the individual proteins that comprise them - might lead to effective therapeutics to reverse or treat SUDs in patients.
Collapse
Affiliation(s)
- Ethan M Anderson
- Departments of Neuroscience and Psychiatry and Behavioral Sciences, Medical University of South Carolina, 173 Ashley Ave, MSC 510, Charleston, SC, 29425-2030, USA
| | - Rachel D Penrod
- Departments of Neuroscience and Psychiatry and Behavioral Sciences, Medical University of South Carolina, 173 Ashley Ave, MSC 510, Charleston, SC, 29425-2030, USA
| | - Sarah M Barry
- Departments of Neuroscience and Psychiatry and Behavioral Sciences, Medical University of South Carolina, 173 Ashley Ave, MSC 510, Charleston, SC, 29425-2030, USA
| | - Brandon W Hughes
- Departments of Neuroscience and Psychiatry and Behavioral Sciences, Medical University of South Carolina, 173 Ashley Ave, MSC 510, Charleston, SC, 29425-2030, USA
| | - Makoto Taniguchi
- Departments of Neuroscience and Psychiatry and Behavioral Sciences, Medical University of South Carolina, 173 Ashley Ave, MSC 510, Charleston, SC, 29425-2030, USA
| | - Christopher W Cowan
- Departments of Neuroscience and Psychiatry and Behavioral Sciences, Medical University of South Carolina, 173 Ashley Ave, MSC 510, Charleston, SC, 29425-2030, USA
| |
Collapse
|
28
|
Knockdown of the histone di-methyltransferase G9a in nucleus accumbens shell decreases cocaine self-administration, stress-induced reinstatement, and anxiety. Neuropsychopharmacology 2019; 44:1370-1376. [PMID: 30587852 PMCID: PMC6785019 DOI: 10.1038/s41386-018-0305-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 12/06/2018] [Accepted: 12/16/2018] [Indexed: 12/18/2022]
Abstract
Comorbid neuropsychiatric disorders such as addiction and anxiety could involve common underlying mechanisms. One potential mechanism involves epigenetic regulation of histone 3 dimethylation at lysine 9 residues (H3K9me2) by the histone dimethyltransferase G9a. Here we provide evidence that local AAV-RNAi-mediated knockdown of G9a expression in nucleus accumbens shell (NAcSh) of male rats reduces both addictive-related and anxiety-related behaviors. Specifically, G9a knockdown reduces sensitivity to low dose cocaine reinforcement when cocaine is freely available (fixed ratio schedule). Similarly, G9a knockdown reduces motivation for cocaine under higher effort demands (progressive ratio schedule). Following several weeks of forced abstinence, G9a knockdown attenuates extinction responding and reinstatement triggered by either cocaine-priming injections or footshock stress. This decrease in addictive behavior is associated with a long-term reduction in anxiety-like behavior as measured by the elevated plus maze (EPM). G9a knockdown also reduces basal anxiety-like behavior in EPM and marble burying tests in drug-naïve rats. These results complement our previous work showing that increased G9a expression in NAcSh enhances addictive-related and anxiety-related behaviors, indicating that G9a bi-directionally controls these responses. These results also suggest that regulation of G9a-influenced gene expression could be a common epigenetic mechanism for co-morbid anxiety and psychostimulant addiction.
Collapse
|
29
|
Bavley CC, Rajadhyaksha AM. Anxiety, the chicken or the egg of addiction: targeting G9a for the treatment of comorbid anxiety and cocaine addiction. Neuropsychopharmacology 2019; 44:1345-1346. [PMID: 30760826 PMCID: PMC6785016 DOI: 10.1038/s41386-019-0329-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 01/22/2019] [Indexed: 12/11/2022]
Abstract
Treating cocaine addiction is a major challenge and currently no FDA approved pharmacotherapies exist. One complicating factor is a high rate of comorbidity between cocaine and neuropsychiatric conditions such as anxiety. The relationship between anxiety symptoms and cocaine addiction is complicated; anxiety can be both a predisposing factor and a consequence of cocaine use as anxiety symptoms often emerge during drug use and withdrawal. Identifying and understanding the shared biological mechanisms that lead to comorbid anxiety and cocaine addiction, irrespective of which comes first, is critical for the identification of new treatments.
Collapse
Affiliation(s)
- Charlotte C. Bavley
- 000000041936877Xgrid.5386.8Pediatric Neurology, Pediatrics, Weill Cornell Medicine, New York, NY 10065 USA ,000000041936877Xgrid.5386.8Feil Family Brain and Mind and Research Institute, Weill Cornell Medicine, New York, NY 10065 USA
| | - Anjali M. Rajadhyaksha
- 000000041936877Xgrid.5386.8Pediatric Neurology, Pediatrics, Weill Cornell Medicine, New York, NY 10065 USA ,000000041936877Xgrid.5386.8Feil Family Brain and Mind and Research Institute, Weill Cornell Medicine, New York, NY 10065 USA
| |
Collapse
|
30
|
Ruud J, Alber J, Tokarska A, Engström Ruud L, Nolte H, Biglari N, Lippert R, Lautenschlager Ä, Cieślak PE, Szumiec Ł, Hess ME, Brönneke HS, Krüger M, Nissbrandt H, Korotkova T, Silberberg G, Rodriguez Parkitna J, Brüning JC. The Fat Mass and Obesity-Associated Protein (FTO) Regulates Locomotor Responses to Novelty via D2R Medium Spiny Neurons. Cell Rep 2019; 27:3182-3198.e9. [DOI: 10.1016/j.celrep.2019.05.037] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 12/14/2018] [Accepted: 05/09/2019] [Indexed: 12/17/2022] Open
|
31
|
7,8-Dihydroxyflavone blocks the development of behavioral sensitization to MDPV, but not to cocaine: Differential role of the BDNF-TrkB pathway. Biochem Pharmacol 2019; 163:84-93. [PMID: 30738029 DOI: 10.1016/j.bcp.2019.02.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 02/05/2019] [Indexed: 02/05/2023]
Abstract
3,4-Methylenedioxypyrovalerone (MDPV) acts as a dopamine transporter blocker and exerts powerful psychostimulant effects. In this study we aimed to investigate the bidirectional cross-sensitization between MDPV and cocaine, as well as to evaluate the role of the BDNF-TrkB signaling pathway in the development of locomotor sensitization to both drugs. Mice were treated with MDPV (1.5 mg/kg) or cocaine (10 or 15 mg/kg) once daily for 5 days. After withdrawal (10 days), animals were challenged with cocaine (8 mg/kg) or MDPV (1 mg/kg). For biochemical determinations, MDPV (1.5 mg/kg) or cocaine (15 mg/kg) were administered acutely or repeatedly, and BDNF, D3R and G9a transcription levels as well as pro- and mature BDNF protein levels were determined. Our results demonstrate that repeated administration of MDPV or cocaine sensitizes to cocaine and MDPV locomotor effects. After an acute or a repeated exposure to MDPV, cortical mRNA BDNF levels were increased, while a decrease in mBDNF protein levels in the nucleus accumbens 2 h after repeated exposure was evidenced. Interestingly, such decline was involved in the development of locomotor sensitization, thus the pretreatment with 7,8-dihydroxyflavone (10 mg/kg), a TrkB agonist, blocked the development of sensitization to MDPV but not to cocaine, for which no changes in the BDNF-TrkB signaling pathway were observed at early withdrawal. In conclusion, a bidirectional cross-sensitization between MDPV and cocaine was evidenced. Our findings suggest that decreased BDNF-TrkB signaling has an important role in the behavioral sensitization to MDPV, pointing TrkB modulation as a target to prevent MDPV sensitization.
Collapse
|
32
|
Pang KKL, Sharma M, Sajikumar S. Epigenetics and memory: Emerging role of histone lysine methyltransferase G9a/GLP complex as bidirectional regulator of synaptic plasticity. Neurobiol Learn Mem 2019; 159:1-5. [PMID: 30703547 DOI: 10.1016/j.nlm.2019.01.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 12/13/2018] [Accepted: 01/24/2019] [Indexed: 12/20/2022]
Abstract
Various epigenetic modifications, including histone lysine methylation, play an integral role in learning and memory. The importance of the histone lysine methyltransferase complex G9a/GLP and its associated histone H3 lysine K9 dimethylation in memory formation and cognition, has garnered the attention of researchers in the past decade. Recent studies feature G9a/GLP as the 'bidirectional regulator of synaptic plasticity', the neural correlate of memory. As the 'title' suggests, G9a/GLP participates in the maintenance of both long-term potentiation (LTP) and long-term depression (LTD). This complex is demonstrated to mostly suppress LTP-related plasticity-related products (PRPs). Notably, our recent paper also shows that G9a/GLP facilitates LTD maintenance in intact hippocampal slices - shedding light on the overlooked influence of epigenetics on LTD. Although the exact mechanisms of G9a/GLP activity regulation in cognition remain elusive, pharmacological inhibition of G9a/GLP presents a new avenue of therapeutic intervention in epigenetic dysfunction-related cognitive deficits.
Collapse
Affiliation(s)
- Karen Ka Lam Pang
- Department of Physiology, 2 Medical Drive, MD9, National University of Singapore, Singapore 117593, Singapore; Neurobiology/Aging Programme, Life Sciences Institute, Centre for Life Sciences, 28 Medical Drive, Singapore 117456, Singapore
| | - Mahima Sharma
- Department of Physiology, 2 Medical Drive, MD9, National University of Singapore, Singapore 117593, Singapore; Neurobiology/Aging Programme, Life Sciences Institute, Centre for Life Sciences, 28 Medical Drive, Singapore 117456, Singapore; Department of Biomedical Engineering, City College of New York, 160 Convent Ave, New York, NY 10031, United States
| | - Sreedharan Sajikumar
- Department of Physiology, 2 Medical Drive, MD9, National University of Singapore, Singapore 117593, Singapore; Neurobiology/Aging Programme, Life Sciences Institute, Centre for Life Sciences, 28 Medical Drive, Singapore 117456, Singapore.
| |
Collapse
|
33
|
De Sa Nogueira D, Merienne K, Befort K. Neuroepigenetics and addictive behaviors: Where do we stand? Neurosci Biobehav Rev 2018; 106:58-72. [PMID: 30205119 DOI: 10.1016/j.neubiorev.2018.08.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 07/28/2018] [Accepted: 08/29/2018] [Indexed: 12/21/2022]
Abstract
Substance use disorders involve long-term changes in the brain that lead to compulsive drug seeking, craving, and a high probability of relapse. Recent findings have highlighted the role of epigenetic regulations in controlling chromatin access and regulation of gene expression following exposure to drugs of abuse. In the present review, we focus on data investigating genome-wide epigenetic modifications in the brain of addicted patients or in rodent models exposed to drugs of abuse, with a particular focus on DNA methylation and histone modifications associated with transcriptional studies. We highlight critical factors for epigenomic studies in addiction. We discuss new findings related to psychostimulants, alcohol, opiate, nicotine and cannabinoids. We examine the possible transmission of these changes across generations. We highlight developing tools, specifically those that allow investigation of structural reorganization of the chromatin. These have the potential to increase our understanding of alteration of chromatin architecture at gene regulatory regions. Neuroepigenetic mechanisms involved in addictive behaviors could explain persistent phenotypic effects of drugs and, in particular, vulnerability to relapse.
Collapse
Affiliation(s)
- David De Sa Nogueira
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), UMR 7364, CNRS, Université de Strasbourg, Team 3 « Abuse of Drugs and Neuroadaptations », Faculté de Psychologie, 12 rue Goethe, F-67000, France
| | - Karine Merienne
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), UMR 7364, CNRS, Université de Strasbourg, Team 1 « Dynamics of Memory and Epigenetics », Faculté de Psychologie, 12 rue Goethe, F-67000, France
| | - Katia Befort
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), UMR 7364, CNRS, Université de Strasbourg, Team 3 « Abuse of Drugs and Neuroadaptations », Faculté de Psychologie, 12 rue Goethe, F-67000, France.
| |
Collapse
|
34
|
McCullough KM, Daskalakis NP, Gafford G, Morrison FG, Ressler KJ. Cell-type-specific interrogation of CeA Drd2 neurons to identify targets for pharmacological modulation of fear extinction. Transl Psychiatry 2018; 8:164. [PMID: 30135420 PMCID: PMC6105686 DOI: 10.1038/s41398-018-0190-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 04/23/2018] [Accepted: 06/05/2018] [Indexed: 12/15/2022] Open
Abstract
Behavioral and molecular characterization of cell-type-specific populations governing fear learning and behavior is a promising avenue for the rational identification of potential therapeutics for fear-related disorders. Examining cell-type-specific changes in neuronal translation following fear learning allows for targeted pharmacological intervention during fear extinction learning, mirroring possible treatment strategies in humans. Here we identify the central amygdala (CeA) Drd2-expressing population as a novel fear-supporting neuronal population that is molecularly distinct from other, previously identified, fear-supporting CeA populations. Sequencing of actively translating transcripts of Drd2 neurons using translating ribosome affinity purification (TRAP) technology identifies mRNAs that are differentially regulated following fear learning. Differentially expressed transcripts with potentially targetable gene products include Npy5r, Rxrg, Adora2a, Sst5r, Fgf3, Erbb4, Fkbp14, Dlk1, and Ssh3. Direct pharmacological manipulation of NPY5R, RXR, and ADORA2A confirms the importance of this cell population and these cell-type-specific receptors in fear behavior. Furthermore, these findings validate the use of functionally identified specific cell populations to predict novel pharmacological targets for the modulation of emotional learning.
Collapse
Affiliation(s)
- Kenneth M McCullough
- Division of Depression and Anxiety Disorders, McLean Hospital, Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Department of Psychiatry, and Behavioral Sciences, Behavioral Neuroscience, Emory University, Atlanta, GA, USA
| | - Nikolaos P Daskalakis
- Division of Depression and Anxiety Disorders, McLean Hospital, Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Georgette Gafford
- Department of Psychiatry, and Behavioral Sciences, Behavioral Neuroscience, Emory University, Atlanta, GA, USA
| | - Filomene G Morrison
- Department of Psychiatry, and Behavioral Sciences, Behavioral Neuroscience, Emory University, Atlanta, GA, USA
- VA Boston Healthcare System, Boston, MA, USA
- Behavioral Science Division, National Center for PTSD, Boston, MA, USA
- Department of Psychiatry, Boston University School of Medicine, Boston, MA, USA
| | - Kerry J Ressler
- Division of Depression and Anxiety Disorders, McLean Hospital, Department of Psychiatry, Harvard Medical School, Boston, MA, USA.
- Department of Psychiatry, and Behavioral Sciences, Behavioral Neuroscience, Emory University, Atlanta, GA, USA.
| |
Collapse
|
35
|
Early alteration of epigenetic-related transcription in Huntington's disease mouse models. Sci Rep 2018; 8:9925. [PMID: 29967375 PMCID: PMC6028428 DOI: 10.1038/s41598-018-28185-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 06/18/2018] [Indexed: 12/18/2022] Open
Abstract
Transcriptional dysregulation in Huntington’s disease (HD) affects the expression of genes involved in survival and neuronal functions throughout the progression of the pathology. In recent years, extensive research has focused on epigenetic and chromatin-modifying factors as a causative explanation for such dysregulation, offering attractive targets for pharmacological therapies. In this work, we extensively examined the gene expression profiles in the cortex, striatum, hippocampus and cerebellum of juvenile R6/1 and N171-82Q mice, models of rapidly progressive HD, to retrieve the early transcriptional signatures associated with this pathology. These profiles were largely consistent across HD datasets, contained tissular and neuronal-specific genes and showed significant correspondence with the transcriptional changes in mouse strains deficient for epigenetic regulatory genes. The most prominent cases were the conditional knockout of the lysine acetyltransferase CBP in post-mitotic forebrain neurons, the double knockout of the histone methyltransferases Ezh1 and Ezh2, components of the polycomb repressor complex 2 (PRC2), and the conditional mutants of the histone methyltransferases G9a (Ehmt2) and GLP (Ehmt1). Based on these observations, we propose that the neuronal epigenetic status is compromised in the prodromal stages of HD, leading to an altered transcriptional programme that is prominently involved in neuronal identity.
Collapse
|
36
|
Pierce RC, Fant B, Swinford-Jackson SE, Heller EA, Berrettini WH, Wimmer ME. Environmental, genetic and epigenetic contributions to cocaine addiction. Neuropsychopharmacology 2018; 43:1471-1480. [PMID: 29453446 PMCID: PMC5983541 DOI: 10.1038/s41386-018-0008-x] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 12/21/2017] [Accepted: 12/30/2017] [Indexed: 12/13/2022]
Abstract
Decades of research on cocaine has produced volumes of data that have answered many important questions about the nature of this highly addictive drug. Sadly, none of this information has translated into the development of effective therapies for the treatment of cocaine addiction. This review endeavors to assess the current state of cocaine research in an attempt to identify novel pathways for therapeutic development. For example, risk of cocaine addiction is highly heritable but genome-wide analyses comparing cocaine-dependent individuals to controls have not resulted in promising targets for drug development. Is this because the genetics of addiction is too complex or because the existing research methodologies are inadequate? Likewise, animal studies have revealed dozens of enduring changes in gene expression following prolonged exposure to cocaine, none of which have translated into therapeutics either because the resulting compounds were ineffective or produced intolerable side-effects. Recently, attention has focused on epigenetic modifications resulting from repeated cocaine intake, some of which appear to be heritable through changes in the germline. While epigenetic changes represent new vistas for therapeutic development, selective manipulation of epigenetic marks is currently challenging even in animals such that translational potential is a distant prospect. This review will reveal that despite the enormous progress made in understanding the molecular and physiological bases of cocaine addiction, there is much that remains a mystery. Continued advances in genetics and molecular biology hold potential for revealing multiple pathways toward the development of treatments for the continuing scourge of cocaine addiction.
Collapse
Affiliation(s)
- R. Christopher Pierce
- 0000 0004 1936 8972grid.25879.31Department of Psychiatry, Center for Neurobiology and Behavior, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Bruno Fant
- 0000 0004 1936 8972grid.25879.31Department of Psychiatry, Center for Neurobiology and Behavior, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Sarah E. Swinford-Jackson
- 0000 0004 1936 8972grid.25879.31Department of Psychiatry, Center for Neurobiology and Behavior, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Elizabeth A. Heller
- 0000 0004 1936 8972grid.25879.31Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Wade H. Berrettini
- 0000 0004 1936 8972grid.25879.31Department of Psychiatry, Center for Neurobiology and Behavior, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Mathieu E. Wimmer
- 0000 0001 2248 3398grid.264727.2Department of Psychology, Temple University, Philadelphia, PA 19122 USA
| |
Collapse
|
37
|
Mostafavi S, Gaiteri C, Sullivan SE, White CC, Tasaki S, Xu J, Taga M, Klein HU, Patrick E, Komashko V, McCabe C, Smith R, Bradshaw EM, Root DE, Regev A, Yu L, Chibnik LB, Schneider JA, Young-Pearse TL, Bennett DA, De Jager PL. A molecular network of the aging human brain provides insights into the pathology and cognitive decline of Alzheimer's disease. Nat Neurosci 2018; 21:811-819. [PMID: 29802388 PMCID: PMC6599633 DOI: 10.1038/s41593-018-0154-9] [Citation(s) in RCA: 359] [Impact Index Per Article: 51.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 04/20/2018] [Indexed: 02/07/2023]
Abstract
There is a need for new therapeutic targets with which to prevent Alzheimer’s disease (AD), a major contributor to aging-related cognitive decline. Here, we report the construction and validation of a molecular network of the aging human frontal cortex. Using RNA sequence data from 478 individuals, we first build a molecular network using modules of coexpressed genes and then relate these modules to AD and its neuropathologic and cognitive endophenotypes. We confirm these associations in two independent AD datasets as well as in epigenomic data. We also illustrate the use of the network in prioritizing amyloid-associated genes for in vitro validation in human neurons and astrocytes. These analyses based on unique cohorts enable us to resolve the role of distinct cortical modules that have a direct effect on the accumulation of AD pathology from those that have a direct effect on cognitive decline, exemplifying a network approach to complex diseases. Systems biology analysis of RNA sequencing data from the aging human cortex identifies a molecular network which prioritizes groups of genes that influence cognitive decline or neuropathology in Alzheimer’s disease.
Collapse
Affiliation(s)
- Sara Mostafavi
- Department of Statistics, Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada.,Canadian Institute for Advanced Research, Toronto, ON, Canada
| | - Chris Gaiteri
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Sarah E Sullivan
- Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
| | | | - Shinya Tasaki
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Jishu Xu
- Broad Institute, Cambridge, MA, USA
| | - Mariko Taga
- Broad Institute, Cambridge, MA, USA.,Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Medical Center, New York, NY, USA
| | - Hans-Ulrich Klein
- Broad Institute, Cambridge, MA, USA.,Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Medical Center, New York, NY, USA
| | | | - Vitalina Komashko
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | | | - Robert Smith
- Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
| | - Elizabeth M Bradshaw
- Broad Institute, Cambridge, MA, USA.,Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Medical Center, New York, NY, USA
| | | | | | - Lei Yu
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Lori B Chibnik
- Broad Institute, Cambridge, MA, USA.,Harvard Medical School, Boston, MA, USA.,Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Julie A Schneider
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Tracy L Young-Pearse
- Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA.
| | - Philip L De Jager
- Broad Institute, Cambridge, MA, USA. .,Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Medical Center, New York, NY, USA.
| |
Collapse
|
38
|
Epigenetic mechanisms associated with addiction-related behavioural effects of nicotine and/or cocaine: implication of the endocannabinoid system. Behav Pharmacol 2018; 28:493-511. [PMID: 28704272 DOI: 10.1097/fbp.0000000000000326] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The addictive use of nicotine (NC) and cocaine (COC) continues to be a major public health problem, and their combined use has been reported, particularly during adolescence. In neural plasticity, commonly induced by NC and COC, as well as behavioural plasticity related to the use of these two drugs, the involvement of epigenetic mechanisms, in which the reversible regulation of gene expression occurs independently of the DNA sequence, has recently been reported. Furthermore, on the basis of intense interactions with the target neurotransmitter systems, the endocannabinoid (ECB) system has been considered pivotal for eliciting the effects of NC or COC. The combined use of marijuana with NC and/or COC has also been reported. This article presents the addiction-related behavioural effects of NC and/or COC, based on the common behavioural/neural plasticity and combined use of NC/COC, and reviews the interacting role of the ECB system. The epigenetic processes inseparable from the effects of NC and/or COC (i.e. DNA methylation, histone modifications and alterations in microRNAs) and the putative therapeutic involvement of the ECB system at the epigenetic level are also discussed.
Collapse
|
39
|
Sharma M, Razali NB, Sajikumar S. Inhibition of G9a/GLP Complex Promotes Long-Term Potentiation and Synaptic Tagging/Capture in Hippocampal CA1 Pyramidal Neurons. Cereb Cortex 2018; 27:3161-3171. [PMID: 27252354 DOI: 10.1093/cercor/bhw170] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Epigenetic regulations play an important role in regulating the learning and memory processes. G9a/G9a-like protein (GLP) lysine dimethyltransferase complex controls a prominent histone H3 lysine9 dimethylation (H3K9me2) that results in transcriptional silencing of the chromatin. Here, we report that the inhibition of G9a/GLP complex by either of the substrate competitive inhibitors UNC 0638 or BIX 01294 reinforces protein synthesis-independent long-term potentiation (early-LTP) to protein synthesis-dependent long-term potentiation (late-LTP). The reinforcement effect was observed if the inhibitors were present during the induction of early-LTP and in addition when G9a/GLP complex inhibition was carried out by priming of synapses within an interval of 30 min before or after the induction of early-LTP. Surprisingly, the reinforced LTP by G9a/GLP complex inhibition was able to associate with a weak plasticity event from nearby independent synaptic populations, resulting in synaptic tagging/capture (STC). We have identified brain-derived neurotrophic factor (BDNF) as a critical plasticity protein that maintains G9a/GLP complex inhibition-mediated LTP facilitation and its STC. Our study reveals an epigenetic mechanism for promoting plasticity and associativity by G9a/GLP complex inhibition, and it may engender a promising epigenetic target for enhancing memory in neural networks.
Collapse
Affiliation(s)
- Mahima Sharma
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117 597, Singapore.,Neurobiology/Aging Program, #04-44, 28 Medical Drive, Life Sciences Institute (LSI), National University of Singapore, Singapore 117 456, Singapore
| | - Nuralyah Bte Razali
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117 597, Singapore.,Neurobiology/Aging Program, #04-44, 28 Medical Drive, Life Sciences Institute (LSI), National University of Singapore, Singapore 117 456, Singapore
| | - Sreedharan Sajikumar
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117 597, Singapore.,Neurobiology/Aging Program, #04-44, 28 Medical Drive, Life Sciences Institute (LSI), National University of Singapore, Singapore 117 456, Singapore
| |
Collapse
|
40
|
Cell-Type-Specific Epigenetic Editing at the Fosb Gene Controls Susceptibility to Social Defeat Stress. Neuropsychopharmacology 2018; 43:272-284. [PMID: 28462942 PMCID: PMC5729576 DOI: 10.1038/npp.2017.88] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 04/12/2017] [Accepted: 04/25/2017] [Indexed: 12/12/2022]
Abstract
Chronic social defeat stress regulates the expression of Fosb in the nucleus accumbens (NAc) to promote the cell-type-specific accumulation of ΔFosB in the two medium spiny neuron (MSN) subtypes in this region. ΔFosB is selectively induced in D1-MSNs in the NAc of resilient mice, and in D2-MSNs of susceptible mice. However, little is known about the consequences of such selective induction, particularly in D2-MSNs. This study examined how cell-type-specific control of the endogenous Fosb gene in NAc regulates susceptibility to social defeat stress. Histone post-translational modifications (HPTMs) were targeted specifically to Fosb using engineered zinc-finger proteins (ZFPs). Fosb-ZFPs were fused to either the transcriptional repressor, G9a, which promotes histone methylation or the transcriptional activator, p65, which promotes histone acetylation. These ZFPs were expressed in D1- vs D2-MSNs using Cre-dependent viral expression in the NAc of mice transgenic for Cre recombinase in these MSN subtypes. We found that stress susceptibility is oppositely regulated by the specific cell type and HPTM targeted. We report that Fosb-targeted histone acetylation in D2-MSNs or histone methylation in D1-MSNs promotes a stress-susceptible, depressive-like phenotype, while histone methylation in D2-MSNs or histone acetylation in D1-MSNs increases resilience to social stress as quantified by social interaction behavior and sucrose preference. This work presents the first demonstration of cell- and gene-specific targeting of histone modifications, which model naturally occurring transcriptional phenomena that control social defeat stress behavior. This epigenetic-editing approach, which recapitulates physiological changes in gene expression, reveals clear differences in the social defeat phenotype induced by Fosb gene manipulation in MSN subtypes.
Collapse
|
41
|
Mancarci BO, Toker L, Tripathy SJ, Li B, Rocco B, Sibille E, Pavlidis P. Cross-Laboratory Analysis of Brain Cell Type Transcriptomes with Applications to Interpretation of Bulk Tissue Data. eNeuro 2017; 4:ENEURO.0212-17.2017. [PMID: 29204516 PMCID: PMC5707795 DOI: 10.1523/eneuro.0212-17.2017] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 10/25/2017] [Accepted: 10/31/2017] [Indexed: 12/13/2022] Open
Abstract
Establishing the molecular diversity of cell types is crucial for the study of the nervous system. We compiled a cross-laboratory database of mouse brain cell type-specific transcriptomes from 36 major cell types from across the mammalian brain using rigorously curated published data from pooled cell type microarray and single-cell RNA-sequencing (RNA-seq) studies. We used these data to identify cell type-specific marker genes, discovering a substantial number of novel markers, many of which we validated using computational and experimental approaches. We further demonstrate that summarized expression of marker gene sets (MGSs) in bulk tissue data can be used to estimate the relative cell type abundance across samples. To facilitate use of this expanding resource, we provide a user-friendly web interface at www.neuroexpresso.org.
Collapse
Affiliation(s)
- B. Ogan Mancarci
- Graduate Program in Bioinformatics, University of British Columbia, Vancouver V6T 1Z4, Canada
- Department of Psychiatry, University of British Columbia, Vancouver V6T 2A1, Canada
- Michael Smith Laboratories, University of British Columbia, Vancouver V6T 1Z4, Canada
| | - Lilah Toker
- Department of Psychiatry, University of British Columbia, Vancouver V6T 2A1, Canada
- Michael Smith Laboratories, University of British Columbia, Vancouver V6T 1Z4, Canada
| | - Shreejoy J. Tripathy
- Department of Psychiatry, University of British Columbia, Vancouver V6T 2A1, Canada
- Michael Smith Laboratories, University of British Columbia, Vancouver V6T 1Z4, Canada
| | - Brenna Li
- Department of Psychiatry, University of British Columbia, Vancouver V6T 2A1, Canada
- Michael Smith Laboratories, University of British Columbia, Vancouver V6T 1Z4, Canada
| | - Brad Rocco
- Campbell Family Mental Health Research Institute of CAMH
- Department of Psychiatry and the Department of Pharmacology and Toxicology, University of Toronto, Vancouver M5S 1A8, Canada
| | - Etienne Sibille
- Campbell Family Mental Health Research Institute of CAMH
- Department of Psychiatry and the Department of Pharmacology and Toxicology, University of Toronto, Vancouver M5S 1A8, Canada
| | - Paul Pavlidis
- Department of Psychiatry, University of British Columbia, Vancouver V6T 2A1, Canada
- Michael Smith Laboratories, University of British Columbia, Vancouver V6T 1Z4, Canada
| |
Collapse
|
42
|
Pinzón JH, Reed AR, Shalaby NA, Buszczak M, Rodan AR, Rothenfluh A. Alcohol-Induced Behaviors Require a Subset of Drosophila JmjC-Domain Histone Demethylases in the Nervous System. Alcohol Clin Exp Res 2017; 41:2015-2024. [PMID: 28940624 DOI: 10.1111/acer.13508] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 09/15/2017] [Indexed: 12/18/2022]
Abstract
BACKGROUND Long-lasting transcriptional changes underlie a number of adaptations that contribute to alcohol use disorders (AUD). Chromatin remodeling, including histone methylation, can confer distinct, long-lasting transcriptional changes, and histone methylases are known to play a role in the development of addiction. Conversely, little is known about the relevance of Jumonji (JmjC) domain-containing demethylases in AUDs. We systematically surveyed the alcohol-induced phenotypes of null mutations in all 13 Drosophila JmjC genes. METHODS We used a collection of JmjC mutants, the majority of which we generated by homologous recombination, and assayed them in the Booze-o-mat to determine their naïve sensitivity to sedation and their tolerance (change in sensitivity upon repeat exposure). Mutants with reproducible phenotypes had their phenotypes rescued with tagged genomic transgenes, and/or phenocopied by nervous system-specific knockdown using RNA interference (RNAi). RESULTS Four of the 13 JmjC genes (KDM3, lid, NO66, and HSPBAP1) showed reproducible ethanol (EtOH) sensitivity phenotypes. Some of the phenotypes were observed across doses, for example, the enhanced EtOH sensitivity of KDM3KO and NO66KO , but others were dose dependent, such as the reduced EtOH sensitivity of HSPBAP1KO , or the enhanced EtOH tolerance of NO66KO . These phenotypes were rescued by their respective genomic transgenes in KDM3KO and NO66KO mutants. While we were unable to rescue lidk mutants, knockdown of lid in the nervous system recapitulated the lidk phenotype, as was observed for KDM3KO and NO66KO RNAi-mediated knockdown. CONCLUSIONS Our study reveals that the Drosophila JmjC-domain histone demethylases Lid, KDM3, NO66, and HSPBAP1 are required for normal EtOH-induced sedation and tolerance. Three of 3 tested of those 4 JmjC genes are required in the nervous system for normal alcohol-induced behavioral responses, suggesting that this gene family is an intriguing avenue for future research.
Collapse
Affiliation(s)
- Jorge H Pinzón
- Department of Psychiatry, Southwestern Medical Center, University of Texas, Dallas, Texas.,Molecular Biology, Southwestern Medical Center, University of Texas, Dallas, Texas
| | - Addison R Reed
- Department of Psychiatry, University of Utah, Salt Lake City, Utah
| | - Nevine A Shalaby
- Molecular Biology, Southwestern Medical Center, University of Texas, Dallas, Texas
| | - Michael Buszczak
- Molecular Biology, Southwestern Medical Center, University of Texas, Dallas, Texas
| | - Aylin R Rodan
- Departments of Internal Medicine/Division of Nephrology, Molecular Medicine Program, University of Utah, Salt Lake City, Utah
| | - Adrian Rothenfluh
- Department of Psychiatry, Southwestern Medical Center, University of Texas, Dallas, Texas.,Department of Psychiatry, University of Utah, Salt Lake City, Utah
| |
Collapse
|
43
|
Kim HD, Call T, Magazu S, Ferguson D. Drug Addiction and Histone Code Alterations. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 978:127-143. [PMID: 28523544 DOI: 10.1007/978-3-319-53889-1_7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Acute and prolonged exposure to drugs of abuse induces changes in gene expression, synaptic function, and neural plasticity in brain regions involved in reward. Numerous genes are involved in this process, and persistent changes in gene expression coincide with epigenetic histone modifications and DNA methylation. Histone modifications are attractive regulatory mechanisms, which can encode complex environmental signals in the genome of postmitotic cells, like neurons. Recently, it has been demonstrated that specific histone modifications are involved in addiction-related gene regulatory mechanisms, by a diverse set of histone-modifying enzymes and readers. These histone modifiers and readers may prove to be valuable pharmacological targets for effective treatments for drug addiction.
Collapse
Affiliation(s)
- Hee-Dae Kim
- Department of Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, 85004, USA
| | - Tanessa Call
- Department of Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, 85004, USA
| | - Samantha Magazu
- Department of Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, 85004, USA
| | - Deveroux Ferguson
- Department of Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, 85004, USA.
| |
Collapse
|
44
|
Chandra R, Lobo MK. Beyond Neuronal Activity Markers: Select Immediate Early Genes in Striatal Neuron Subtypes Functionally Mediate Psychostimulant Addiction. Front Behav Neurosci 2017. [PMID: 28642692 PMCID: PMC5462953 DOI: 10.3389/fnbeh.2017.00112] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Immediate early genes (IEGs) were traditionally used as markers of neuronal activity in striatum in response to stimuli including drugs of abuse such as psychostimulants. Early studies using these neuronal activity markers led to important insights in striatal neuron subtype responsiveness to psychostimulants. Such studies have helped identify striatum as a critical brain center for motivational, reinforcement and habitual behaviors in psychostimulant addiction. While the use of IEGs as neuronal activity markers in response to psychostimulants and other stimuli persists today, the functional role and implications of these IEGs has often been neglected. Nonetheless, there is a subset of research that investigates the functional role of IEGs in molecular, cellular and behavioral alterations by psychostimulants through striatal medium spiny neuron (MSN) subtypes, the two projection neuron subtypes in striatum. This review article will address and highlight the studies that provide a functional mechanism by which IEGs mediate psychostimulant molecular, cellular and behavioral plasticity through MSN subtypes. Insight into the functional role of IEGs in striatal MSN subtypes could provide improved understanding into addiction and neuropsychiatric diseases affecting striatum, such as affective disorders and compulsive disorders characterized by dysfunctional motivation and habitual behavior.
Collapse
Affiliation(s)
- Ramesh Chandra
- Department of Anatomy and Neurobiology, University of Maryland School of MedicineBaltimore, MD, United States
| | - Mary Kay Lobo
- Department of Anatomy and Neurobiology, University of Maryland School of MedicineBaltimore, MD, United States
| |
Collapse
|
45
|
Liang L, Gu X, Zhao JY, Wu S, Miao X, Xiao J, Mo K, Zhang J, Lutz BM, Bekker A, Tao YX. G9a participates in nerve injury-induced Kcna2 downregulation in primary sensory neurons. Sci Rep 2016; 6:37704. [PMID: 27874088 PMCID: PMC5118693 DOI: 10.1038/srep37704] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 11/01/2016] [Indexed: 12/25/2022] Open
Abstract
Nerve injury-induced downregulation of voltage-gated potassium channel subunit Kcna2 in the dorsal root ganglion (DRG) is critical for DRG neuronal excitability and neuropathic pain genesis. However, how nerve injury causes this downregulation is still elusive. Euchromatic histone-lysine N-methyltransferase 2, also known as G9a, methylates histone H3 on lysine residue 9 to predominantly produce a dynamic histone dimethylation, resulting in condensed chromatin and gene transcriptional repression. We showed here that blocking nerve injury-induced increase in G9a rescued Kcna2 mRNA and protein expression in the axotomized DRG and attenuated the development of nerve injury-induced pain hypersensitivity. Mimicking this increase decreased Kcna2 mRNA and protein expression, reduced Kv current, and increased excitability in the DRG neurons and led to spinal cord central sensitization and neuropathic pain-like symptoms. G9a mRNA is co-localized with Kcna2 mRNA in the DRG neurons. These findings indicate that G9a contributes to neuropathic pain development through epigenetic silencing of Kcna2 in the axotomized DRG.
Collapse
Affiliation(s)
- Lingli Liang
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, USA
| | - Xiyao Gu
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, USA
| | - Jian-Yuan Zhao
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, USA.,State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Shaogen Wu
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, USA
| | - Xuerong Miao
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, USA
| | - Jifang Xiao
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, USA
| | - Kai Mo
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, USA
| | - Jun Zhang
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, USA
| | - Brianna Marie Lutz
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, USA
| | - Alex Bekker
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, USA
| | - Yuan-Xiang Tao
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, USA.,Departments of Cell Biology &Molecular Medicine and Physiology, Pharmacology &Neuroscience, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, USA
| |
Collapse
|
46
|
Aberrant H3.3 dynamics in NAc promote vulnerability to depressive-like behavior. Proc Natl Acad Sci U S A 2016; 113:12562-12567. [PMID: 27791098 DOI: 10.1073/pnas.1608270113] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Human major depressive disorder (MDD), along with related mood disorders, is among the world's greatest public health concerns; however, its pathophysiology remains poorly understood. Persistent changes in gene expression are known to promote physiological aberrations implicated in MDD. More recently, histone mechanisms affecting cell type- and regional-specific chromatin structures have also been shown to contribute to transcriptional programs related to depressive behaviors, as well as responses to antidepressants. Although much emphasis has been placed in recent years on roles for histone posttranslational modifications and chromatin-remodeling events in the etiology of MDD, it has become increasingly clear that replication-independent histone variants (e.g., H3.3), which differ in primary amino acid sequence from their canonical counterparts, similarly play critical roles in the regulation of activity-dependent neuronal transcription, synaptic connectivity, and behavioral plasticity. Here, we demonstrate a role for increased H3.3 dynamics in the nucleus accumbens (NAc)-a key limbic brain reward region-in the regulation of aberrant social stress-mediated gene expression and the precipitation of depressive-like behaviors in mice. We find that molecular blockade of these dynamics promotes resilience to chronic social stress and results in a partial renormalization of stress-associated transcriptional patterns in the NAc. In sum, our findings establish H3.3 dynamics as a critical, and previously undocumented, regulator of mood and suggest that future therapies aimed at modulating striatal histone dynamics may potentiate beneficial behavioral adaptations to negative emotional stimuli.
Collapse
|
47
|
Wang N, Shen X, Bao S, Feng SW, Wang W, Liu Y, Wang Y, Wang X, Guo X, Shen R, Wu H, Lei L, Xu S, Wang F. Dopaminergic inhibition by G9a/Glp complex on tyrosine hydroxylase in nerve injury-induced hypersensitivity. Mol Pain 2016; 12:12/0/1744806916663731. [PMID: 27562335 PMCID: PMC5006299 DOI: 10.1177/1744806916663731] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 07/18/2016] [Indexed: 12/24/2022] Open
Abstract
The neural balance between facilitation and inhibition determines the final tendency of central sensitization. Nerve injury-induced hypersensitivity was considered as the results from the enhanced ascending facilitation and the diminished descending inhibition. The role of dopaminergic transmission in the descending inhibition has been well documented, but its underlying molecular mechanisms are unclear. Previous studies demonstrated that the lysine dimethyltransferase G9a/G9a-like protein (Glp) complex plays a critical role in cocaine-induced central plasticity, and given cocaine’s role in the nerve system is relied on its function on dopamine system, we herein proposed that the reduced inhibition of dopaminergic transmission was from the downregulation of tyrosine hydroxylase expression by G9a/Glp complex through methylating its gene Th. After approval by the Animal Care and Use Committee, C57BL/6 mice were used for pain behavior using von Frey after spared nerve injury, and Th CpG islands methylation was measured using bisulfite sequencing at different nerve areas. The inhibitor of G9a/Glp, BIX 01294, was administered intraventricularly daily with bolus injection. The protein levels of G9a, Glp, and tyrosine hydroxylase were measured with immunoblotting. Dopamine levels were detected using high-performance liquid chromatography. The expression of G9a but not Glp was upregulated in ventral tegmental area at post-injury day 4 till day 49 (the last day of the behavioral test). Correspondingly, the Th CpG methylation is increased, but the tyrosine hydroxylase expression was downregulated and the dopamine level was decreased. After the intracerebroventriclar injection of BIX 01294 since the post-injury days 7 and 14 for consecutive three days, three weeks, and six weeks, the expression of tyrosine hydroxylase was upregulated with a significant decrease in Th methylation and increase in dopamine level. Moreover, the pain after G9a/Glp inhibitor was attenuated significantly. In sum, methytransferase G9a/Glp complex partially controls dopaminergic transmission by methylating Th in peripheral nerve injury-induced neuropathic pain.
Collapse
Affiliation(s)
- Nan Wang
- Department of Anesthesiology, Affiliated Hospital of Obstetrics and Gynecology, Nanjing Medical University, China
| | - Xiaofeng Shen
- Department of Anesthesiology, Affiliated Hospital of Obstetrics and Gynecology, Nanjing Medical University, China
| | - Senzhu Bao
- Department of Stomatology, Affiliated Hospital of Qinghai University, China
| | - Shan-Wu Feng
- Department of Anesthesiology, Affiliated Hospital of Obstetrics and Gynecology, Nanjing Medical University, China
| | - Wei Wang
- Department of Anesthesiology, Affiliated Hospital of Obstetrics and Gynecology, Nanjing Medical University, China
| | - Yusheng Liu
- Department of Anesthesiology, Affiliated Hospital of Obstetrics and Gynecology, Nanjing Medical University, China
| | - Yiquan Wang
- Internal Medicine, Shanghai Municipal Hospital of TCM, Shanghai TCM University, China
| | - Xian Wang
- Department of Anesthesiology, Affiliated Hospital of Obstetrics and Gynecology, Nanjing Medical University, China
| | - Xirong Guo
- Institute of Pediatrics, Affiliated Hospital of Obstetrics and Gynecology, Nanjing Medical University, China
| | - Rong Shen
- Institute of Pediatrics, Affiliated Hospital of Obstetrics and Gynecology, Nanjing Medical University, China
| | - Haibo Wu
- Department of Anesthesiology, Affiliated Hospital of Obstetrics and Gynecology, Nanjing Medical University, China
| | - Liming Lei
- Department of Anesthesiology, Affiliated Hospital of Obstetrics and Gynecology, Nanjing Medical University, China
| | - Shiqin Xu
- Department of Anesthesiology, Affiliated Hospital of Obstetrics and Gynecology, Nanjing Medical University, China
| | - Fuzhou Wang
- Department of Anesthesiology, Affiliated Hospital of Obstetrics and Gynecology, Nanjing Medical University, China Division of Neuroscience, The Bonoi Academy of Science & Education, USA
| |
Collapse
|
48
|
von Schimmelmann M, Feinberg PA, Sullivan JM, Ku SM, Badimon A, Duff MK, Wang Z, Lachmann A, Dewell S, Ma'ayan A, Han MH, Tarakhovsky A, Schaefer A. Polycomb repressive complex 2 (PRC2) silences genes responsible for neurodegeneration. Nat Neurosci 2016; 19:1321-30. [PMID: 27526204 PMCID: PMC5088783 DOI: 10.1038/nn.4360] [Citation(s) in RCA: 161] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 07/14/2016] [Indexed: 12/13/2022]
Abstract
Normal brain function depends on the interaction between highly specialized neurons that operate within anatomically and functionally distinct brain regions. Neuronal specification is driven by transcriptional programs that are established during early neuronal development and remain in place in the adult brain. The fidelity of neuronal specification depends on the robustness of the transcriptional program that supports the neuron type-specific gene expression patterns. Here we show that polycomb repressive complex 2 (PRC2), which supports neuron specification during differentiation, contributes to the suppression of a transcriptional program that is detrimental to adult neuron function and survival. We show that PRC2 deficiency in striatal neurons leads to the de-repression of selected, predominantly bivalent PRC2 target genes that are dominated by self-regulating transcription factors normally suppressed in these neurons. The transcriptional changes in PRC2-deficient neurons lead to progressive and fatal neurodegeneration in mice. Our results point to a key role of PRC2 in protecting neurons against degeneration.
Collapse
Affiliation(s)
- Melanie von Schimmelmann
- Friedman Brain Institute, Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Philip A Feinberg
- Friedman Brain Institute, Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Josefa M Sullivan
- Friedman Brain Institute, Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Stacy M Ku
- Friedman Brain Institute, Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Ana Badimon
- Friedman Brain Institute, Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Mary Kaye Duff
- Friedman Brain Institute, Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Zichen Wang
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,BD2K-LINCS Data Coordination and Integration Center, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Alexander Lachmann
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,BD2K-LINCS Data Coordination and Integration Center, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Scott Dewell
- Genomics Resource Center, The Rockefeller University, New York, New York, USA
| | - Avi Ma'ayan
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,BD2K-LINCS Data Coordination and Integration Center, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Ming-Hu Han
- Friedman Brain Institute, Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Alexander Tarakhovsky
- Laboratory of Immune Cell Epigenetics and Signaling, The Rockefeller University, New York, New York, USA
| | - Anne Schaefer
- Friedman Brain Institute, Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
49
|
Fiszbein A, Kornblihtt AR. Histone methylation, alternative splicing and neuronal differentiation. NEUROGENESIS 2016; 3:e1204844. [PMID: 27606339 DOI: 10.1080/23262133.2016.1204844] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 06/14/2016] [Accepted: 06/17/2016] [Indexed: 12/31/2022]
Abstract
Alternative splicing, as well as chromatin structure, greatly contributes to specific transcriptional programs that promote neuronal differentiation. The activity of G9a, the enzyme responsible for mono- and di-methylation of lysine 9 on histone H3 (H3K9me1 and H3K9me2) in mammalian euchromatin, has been widely implicated in the differentiation of a variety of cell types and tissues. In a recent work from our group (Fiszbein et al., 2016) we have shown that alternative splicing of G9a regulates its nuclear localization and, therefore, the efficiency of H3K9 methylation, which promotes neuronal differentiation. We discuss here our results in the light of a report from other group (Laurent et al. 2015) demonstrating a key role for the alternative splicing of the histone demethylase LSD1 in controlling specific gene expression in neurons. All together, these results illustrate the importance of alternative splicing in the generation of a proper equilibrium between methylation and demethylation of histones for the regulation of neuron-specific transcriptional programs.
Collapse
Affiliation(s)
- Ana Fiszbein
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE-CONICET) and Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires , Buenos Aires, Argentina
| | - Alberto R Kornblihtt
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE-CONICET) and Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires , Buenos Aires, Argentina
| |
Collapse
|
50
|
Fiszbein A, Giono LE, Quaglino A, Berardino BG, Sigaut L, von Bilderling C, Schor IE, Enriqué Steinberg JH, Rossi M, Pietrasanta LI, Caramelo JJ, Srebrow A, Kornblihtt AR. Alternative Splicing of G9a Regulates Neuronal Differentiation. Cell Rep 2016; 14:2797-808. [PMID: 26997278 DOI: 10.1016/j.celrep.2016.02.063] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 01/25/2016] [Accepted: 02/12/2016] [Indexed: 01/08/2023] Open
Abstract
Chromatin modifications are critical for the establishment and maintenance of differentiation programs. G9a, the enzyme responsible for histone H3 lysine 9 dimethylation in mammalian euchromatin, exists as two isoforms with differential inclusion of exon 10 (E10) through alternative splicing. We find that the G9a methyltransferase is required for differentiation of the mouse neuronal cell line N2a and that E10 inclusion increases during neuronal differentiation of cultured cells, as well as in the developing mouse brain. Although E10 inclusion greatly stimulates overall H3K9me2 levels, it does not affect G9a catalytic activity. Instead, E10 increases G9a nuclear localization. We show that the G9a E10(+) isoform is necessary for neuron differentiation and regulates the alternative splicing pattern of its own pre-mRNA, enhancing E10 inclusion. Overall, our findings indicate that by regulating its own alternative splicing, G9a promotes neuron differentiation and creates a positive feedback loop that reinforces cellular commitment to differentiation.
Collapse
Affiliation(s)
- Ana Fiszbein
- Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires and Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE-CONICET), Ciudad Universitaria Pabellón II, C1428EHA Buenos Aires, Argentina
| | - Luciana E Giono
- Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires and Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE-CONICET), Ciudad Universitaria Pabellón II, C1428EHA Buenos Aires, Argentina
| | - Ana Quaglino
- Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires and Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE-CONICET), Ciudad Universitaria Pabellón II, C1428EHA Buenos Aires, Argentina
| | - Bruno G Berardino
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria Pabellón II, C1428EHA Buenos Aires, Argentina
| | - Lorena Sigaut
- Departamento de Física, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires and IFIBA-CONICET, Cuidad Universitaria Pabellón I, C1428EHA Buenos Aires, Argentina
| | - Catalina von Bilderling
- Departamento de Física, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires and IFIBA-CONICET, Cuidad Universitaria Pabellón I, C1428EHA Buenos Aires, Argentina
| | - Ignacio E Schor
- Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires and Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE-CONICET), Ciudad Universitaria Pabellón II, C1428EHA Buenos Aires, Argentina
| | - Juliana H Enriqué Steinberg
- Instituto de Investigación en Biomedicina de Buenos Aires CONICET, Partner Institute of the Max Planck Society, C1425FQD Buenos Aires, Argentina
| | - Mario Rossi
- Instituto de Investigación en Biomedicina de Buenos Aires CONICET, Partner Institute of the Max Planck Society, C1425FQD Buenos Aires, Argentina
| | - Lía I Pietrasanta
- Departamento de Física, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires and IFIBA-CONICET, Cuidad Universitaria Pabellón I, C1428EHA Buenos Aires, Argentina; Centro de Microscopías Avanzadas, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Cuidad Universitaria, C1428EHA Buenos Aires, Argentina
| | - Julio J Caramelo
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria Pabellón II, C1428EHA Buenos Aires, Argentina; Fundación Instituto Leloir, C1405BWE Buenos Aires, Argentina
| | - Anabella Srebrow
- Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires and Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE-CONICET), Ciudad Universitaria Pabellón II, C1428EHA Buenos Aires, Argentina
| | - Alberto R Kornblihtt
- Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires and Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE-CONICET), Ciudad Universitaria Pabellón II, C1428EHA Buenos Aires, Argentina.
| |
Collapse
|