1
|
Iyer ES, Vitaro P, Wu S, Muir J, Tse YC, Cvetkovska V, Bagot RC. Reward integration in prefrontal-cortical and ventral-hippocampal nucleus accumbens inputs cooperatively modulates engagement. Nat Commun 2025; 16:3573. [PMID: 40234437 PMCID: PMC12000462 DOI: 10.1038/s41467-025-58858-4] [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: 06/18/2024] [Accepted: 04/03/2025] [Indexed: 04/17/2025] Open
Abstract
The nucleus accumbens, a highly integrative brain region controlling motivated behavior, receives various glutamatergic inputs, yet the relative functional specialization of these inputs is unclear. While circuit neuroscience commonly seeks specificity, redundancy can be highly adaptive and is a critical motif in circuit organization. Using dual-site fiber photometry in an operant reward task in mice, we simultaneously recorded from two accumbal glutamatergic afferents to assess circuit specialization. We identify a common neural motif integrating reward history in medial prefrontal cortex and ventral hippocampus inputs. By systematically degrading task complexity, dissociating reward from choice and action, we identify circuit-specificity in the behavioral conditions that recruit encoding. While input from the prefrontal cortex invariantly encodes reward, encoding in ventral hippocampal input is uniquely anchored to unrewarded outcomes. Optogenetic stimulation demonstrates that both inputs co-operatively modulate task engagement. We illustrate how similar encoding, differentially gated by behavioral state, supports state-sensitive tuning of reward-motivated behavior.
Collapse
Affiliation(s)
- Eshaan S Iyer
- Integrated Program in Neuroscience, McGill University, Montréal, QC, Canada
| | - Peter Vitaro
- Department of Psychology, McGill University, Montréal, QC, Canada
| | - Serena Wu
- Integrated Program in Neuroscience, McGill University, Montréal, QC, Canada
| | - Jessie Muir
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
- Omenn-Darling Bioengineering Institute, Princeton University, Princeton, NJ, USA
| | - Yiu Chung Tse
- Department of Psychology, McGill University, Montréal, QC, Canada
| | | | - Rosemary C Bagot
- Department of Psychology, McGill University, Montréal, QC, Canada.
- Ludmer Centre for Neuroinformatics and Mental Health, Montréal, QC, Canada.
| |
Collapse
|
2
|
Choi YJ, Rosa BA, Fernandez-Baca MV, Ore RA, Martin J, Ortiz P, Hoban C, Cabada MM, Mitreva M. Independent origins and non-parallel selection signatures of triclabendazole resistance in Fasciola hepatica. Nat Commun 2025; 16:2996. [PMID: 40148292 PMCID: PMC11950404 DOI: 10.1038/s41467-025-57796-5] [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: 05/14/2024] [Accepted: 03/04/2025] [Indexed: 03/29/2025] Open
Abstract
Triclabendazole (TCBZ) is the primary treatment for fascioliasis, a global foodborne zoonosis caused by Fasciola hepatica. Widespread resistance to TCBZ (TCBZ-R) in livestock and a rapid rise in resistant human infections are significant concerns. To understand the genetic basis of TCBZ-R, we sequenced the genomes of 99 TCBZ-sensitive (TCBZ-S) and 210 TCBZ-R adult flukes from 146 bovine livers in Cusco, Peru. We identify genomic regions of high differentiation (FST outliers above the 99.9th percentile) that encod genes involved in the EGFR-PI3K-mTOR-S6K pathway and microtubule function. Transcript expression differences are observed in microtubule-related genes between TCBZ-S and -R flukes, both without drug treatment and in response to treatment. Using only 30 SNPs, it is possible to differentiate between TCBZ-S and -R parasites with ≥75% accuracy. Our outlier loci are distinct from the previously reported TCBZ-R-associated QTLs in the UK, suggesting an independent evolution of resistance alleles. Effective genetics-based TCBZ-R surveillance must consider the heterogeneity of loci under selection across diverse geographical populations.
Collapse
Affiliation(s)
- Young-Jun Choi
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Bruce A Rosa
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Martha V Fernandez-Baca
- Sede Cusco, Instituto de Medicina Tropical "Alexander von Humboldt", Universidad Peruana Cayetano Heredia, Cusco, Peru
| | - Rodrigo A Ore
- Sede Cusco, Instituto de Medicina Tropical "Alexander von Humboldt", Universidad Peruana Cayetano Heredia, Cusco, Peru
| | - John Martin
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Pedro Ortiz
- Laboratorio de Inmunología, Facultad de Ciencias Veterinarias, Universidad Nacional de Cajamarca, Cajamarca, Peru
| | - Cristian Hoban
- Laboratorio de Inmunología, Facultad de Ciencias Veterinarias, Universidad Nacional de Cajamarca, Cajamarca, Peru
| | - Miguel M Cabada
- Sede Cusco, Instituto de Medicina Tropical "Alexander von Humboldt", Universidad Peruana Cayetano Heredia, Cusco, Peru.
- Division of Infectious Diseases, Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, USA.
| | - Makedonka Mitreva
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
3
|
Liu X, Huang Y, Mu L, Friedman V, Kelly TJ, Hu Y, Yuan D, Liu QS. Epac2-mediated synaptic insertion of Ca 2+-permeable AMPARs in the nucleus accumbens contributes to incubation of cocaine craving. Neuropsychopharmacology 2025; 50:620-629. [PMID: 39702576 PMCID: PMC11845495 DOI: 10.1038/s41386-024-02030-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 11/04/2024] [Accepted: 11/12/2024] [Indexed: 12/21/2024]
Abstract
The accumulation of GluA2-lacking Ca2+-permeable AMPARs (CP-AMPARs) in the medium spiny neurons (MSNs) of the nucleus accumbens (NAc) is required for the expression of incubation of cocaine craving. The exchange protein directly activated by cAMP (Epac) is an intracellular effector of cAMP and a guanine nucleotide exchange factor for the small GTPase Rap1. Epac2 has been implicated in the trafficking of AMPA receptors at central synapses. We tested the hypothesis that Epac2 activation contributes to the accumulation of CP-AMPARs in NAc MSNs and incubation of cocaine craving. Here we demonstrate that the selective Epac2 agonist S-220 facilitated the synaptic insertion of GluA2-lacking CP-AMPARs at excitatory synapses onto NAc MSNs. In addition, prolonged abstinence from cocaine self-administration in rats resulted in elevated Rap1-GTP levels in the NAc, implying that Epac2 is activated during incubation. Importantly, we show that AAV-mediated shRNA knockdown of Epac2 in the NAc core attenuated the accumulation of CP-AMPARs and cue-induced drug-seeking behavior after prolonged abstinence from cocaine self-administration. In contrast, acute pharmacological inhibition of Epac2 with the selective Epac2 inhibitor ESI-05 did not alter CP-AMPARs that had already accumulated during incubation, and intra-NAc application of ESI-05 did not significantly affect cue-induced drug seeking following prolonged abstinence. Taken together, these results suggest that Epac2 activation during the period of incubation, but not during cue-induced drug seeking, leads to the accumulation of CP-AMPARs in NAc MSNs, which in turn contributes to incubation of cocaine craving.
Collapse
Affiliation(s)
- Xiaojie Liu
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA.
| | - Yao Huang
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Lianwei Mu
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Vladislav Friedman
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Thomas J Kelly
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Ying Hu
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Dong Yuan
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Qing-Song Liu
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA.
| |
Collapse
|
4
|
Wang H, Li Y, Li X, Sun Z, Yu F, Pashang A, Kulasiri D, Li HW, Chen H, Hou H, Zhang Y. The Primary Cilia are Associated with the Axon Initial Segment in Neurons. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2407405. [PMID: 39804991 PMCID: PMC11884599 DOI: 10.1002/advs.202407405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 12/16/2024] [Indexed: 01/16/2025]
Abstract
The primary cilia serve as pivotal mediators of environmental signals and play crucial roles in neuronal responses. Disruption of ciliary function has been implicated in neuronal circuit disorders and aberrant neuronal excitability. However, the precise mechanisms remain elusive. To study the link between the primary cilia and neuronal excitability, manipulation of somatostatin receptor 3 (SSTR3) is investigated, as an example of how alterations in ciliary signaling may affect neuronal activity. It is found that aberrant SSTR3 expression perturbed not only ciliary morphology but also disrupted ciliary signaling cascades. Genetic deletion of SSTR3 resulted in perturbed spatial memory and synaptic plasticity. The axon initial segment (AIS) is a specialized region in the axon where action potentials are initiated. Interestingly, loss of ciliary SSTR3 led to decrease of Akt-dependent cyclic AMP-response element binding protein (CREB)-mediated transcription at the AIS, specifically downregulating AIS master organizer adaptor protein ankyrin G (AnkG) expression. In addition, alterations of other ciliary proteins serotonin 6 receptor (5-HT6R)and intraflagellar transport protein 88 (IFT88) also induced length changes of the AIS. The findings elucidate a specific interaction between the primary cilia and AIS, providing insight into the impact of the primary cilia on neuronal excitability and circuit integrity.
Collapse
Affiliation(s)
- Han Wang
- State Key Laboratory of Membrane BiologySchool of Life SciencesPeking UniversityBeijing100871China
| | - Yu Li
- State Key Laboratory of Membrane BiologySchool of Life SciencesPeking UniversityBeijing100871China
| | - Xin Li
- Beijing Life Science AcademyBeijing102200China
| | - Zehui Sun
- State Key Laboratory of Membrane BiologySchool of Life SciencesPeking UniversityBeijing100871China
| | - Fengdan Yu
- State Key Laboratory of Membrane BiologySchool of Life SciencesPeking UniversityBeijing100871China
| | - Abolghasem Pashang
- Centre for Advanced Computational Solutions (C‐fACS)AGLS facultyLincoln UniversityCanterbury7647New Zealand
| | - Don Kulasiri
- Centre for Advanced Computational Solutions (C‐fACS)AGLS facultyLincoln UniversityCanterbury7647New Zealand
| | - Hung Wing Li
- Department of ChemistryThe Chinese University of Hong KongHong Kong999077China
| | - Huan Chen
- Beijing Life Science AcademyBeijing102200China
| | - Hongwei Hou
- Beijing Life Science AcademyBeijing102200China
| | - Yan Zhang
- State Key Laboratory of Membrane BiologySchool of Life SciencesPeking UniversityBeijing100871China
| |
Collapse
|
5
|
Nestler EJ. The biology of addiction. Sci Signal 2025; 18:eadq0031. [PMID: 39903810 DOI: 10.1126/scisignal.adq0031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Accepted: 12/10/2024] [Indexed: 02/06/2025]
Abstract
The tools of modern genetics and neurobiology have propelled a renaissance of research that has advanced our understanding of the pathophysiology of drug addiction. We know that an individual's risk for addiction is determined by interactions between genetics and environment and that only a minute fraction of chemical agents share the ability to act on this vulnerability to induce a state of addiction. Repeated exposure to these drugs causes addiction through repeated activation of dopaminergic transmission (and many other actions) in the brain, inducing changes at the molecular, cellular, and synaptic levels that, over time, rewire the circuitry throughout the limbic system. In this Review, I discuss how we are gaining a clearer picture of this drug-induced plasticity-some of which is shared by all addictive drugs, whereas other aspects are specific to certain drug classes-and of the ways in which these adaptations mediate the range of behavioral abnormalities that define the addicted state. Despite the challenges, there is reason for optimism in translating this rich biological understanding of addiction into improved treatments for the many individuals burdened by this illness around the world.
Collapse
Affiliation(s)
- Eric J Nestler
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
6
|
Bjornson KJ, Kermath BA, Cahill ME. Identification of ARHGEF11 (PDZ-RhoGEF) as an in vivo regulator of synapses and cognition. Proc Natl Acad Sci U S A 2025; 122:e2415316122. [PMID: 39835891 PMCID: PMC11789018 DOI: 10.1073/pnas.2415316122] [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: 07/30/2024] [Accepted: 12/11/2024] [Indexed: 01/22/2025] Open
Abstract
Given the influence of cognitive abilities on life outcomes, there is inherent value in identifying genes involved in controlling learning and memory. Further, cognitive dysfunction is a core feature of many neuropsychiatric disorders. Here, we use a combinatory in silico approach to identify human gene targets that will have an especially high likelihood of individually and directly impacting cognition. This broad and unbiased screen led to the specific identification of ARHGEF11, which encodes PDZ-RhoGEF. PDZ-RhoGEF is a largely RhoA-specific activator that is highly enriched in dendritic spines, and recent work identified hyperexpression of PDZ-RhoGEF in the prefrontal cortex of bipolar disorder subjects, a disease characterized by an early emergence and persistence of broad scope cognitive dysfunction. Here, we characterize the effects of PDZ-RhoGEF on synaptic and behavioral phenotypes, and we identify molecular and biochemical mechanisms that control PDZ-RhoGEF's expression, synaptic spatial localization, and enzymatic activity. Importantly, our identified direct regulators of PDZ-RhoGEF (miR-132 and DISC1) have themselves been repeatedly implicated in controlling cognitive phenotypes in humans, including those caused by several neuropsychiatric disorders. Taken together, our findings indicate that PDZ-RhoGEF is a key convergence point among multiple synaptic and cognition-relevant signaling cascades with potential translational significance.
Collapse
Affiliation(s)
- Kathryn J. Bjornson
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI53706
| | - Bailey A. Kermath
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI53706
| | - Michael E. Cahill
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI53706
| |
Collapse
|
7
|
Kim YJ, Kim K, Lee Y, Min HW, Ko YH, Lee BR, Hur KH, Kim SK, Lee SY, Jang CG. The mutated cytoplasmic fragile X messenger ribonucleoprotein 1 (FMR1)-interacting protein 2 (CYFIP2 S968F) regulates cocaine-induced reward behaviour and plasticity in the nucleus accumbens. Br J Pharmacol 2024; 181:3327-3345. [PMID: 38751203 DOI: 10.1111/bph.16427] [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: 12/21/2023] [Revised: 04/02/2024] [Accepted: 04/25/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND AND PURPOSE Cytoplasmic fragile X messenger ribonucleoprotein 1 (FMR1)-interacting protein 2 (CYFIP2), as a component of the Wiskott-Aldrich syndrome protein family verprolin-homologous protein (WAVE) regulatory complex, is involved in actin polymerization, contributing to neuronal development and structural plasticity. Mutating serine-968 to phenylalanine (S968F) in CYFIP2 causes an altered cocaine response in mice. The neuronal mechanisms underlying this response remain unknown. EXPERIMENTAL APPROACH We performed cocaine reward-related behavioural tests and examined changes in synaptic protein phenotypes and neuronal morphology in the nucleus accumbens (NAc), using CYFIP2 S968F knock-in mice to investigate the role of CYFIP2 in regulating cocaine reward. KEY RESULTS CYFIP2 S968F mutation attenuated cocaine-induced behavioural sensitization and conditioned place preference. Cocaine-induced c-Fos was not observed in the NAc of CYFIP2 S968F knock-in mice. However, c-Fos induction was still evident in the medial prefrontal cortex (mPFC). CYFIP2 S968F mutation altered cocaine-associated CYFIP2 signalling, glutamatergic protein expression and synaptic density in the NAc following cocaine exposure. To further determine the role of CYFIP2 in NAc neuronal activity and the mPFC projecting to the NAc activity-mediating reward response, we used optogenetic tools to stimulate the NAc or mPFC-NAc pathway and observed that optogenetic activation of the NAc or mPFC-NAc pathway induced reward-related behaviours. This effect was not observed in the S968F mutation in CYFIP2. CONCLUSION AND IMPLICATIONS These results suggest that CYFIP2 plays a role in controlling cocaine-mediated neuronal function and structural plasticity in the NAc, and that CYFIP2 could serve as a target for regulating cocaine reward.
Collapse
Affiliation(s)
- Young-Jung Kim
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Kyungin Kim
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Youyoung Lee
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Hee-Won Min
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Yong-Hyun Ko
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Bo-Ram Lee
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Kwang-Hyun Hur
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Seon-Kyung Kim
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Seok-Yong Lee
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Choon-Gon Jang
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| |
Collapse
|
8
|
Kietzman HW, Trinoskey-Rice G, Seo EH, Guo J, Gourley SL. Neuronal Ensembles in the Amygdala Allow Social Information to Motivate Later Decisions. J Neurosci 2024; 44:e1848232024. [PMID: 38499360 PMCID: PMC11026342 DOI: 10.1523/jneurosci.1848-23.2024] [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: 09/28/2023] [Revised: 02/02/2024] [Accepted: 02/19/2024] [Indexed: 03/20/2024] Open
Abstract
Social experiences carry tremendous weight in our decision-making, even when social partners are not present. To determine mechanisms, we trained female mice to respond for two food reinforcers. Then, one food was paired with a novel conspecific. Mice later favored the conspecific-associated food, even in the absence of the conspecific. Chemogenetically silencing projections from the prelimbic subregion (PL) of the medial prefrontal cortex to the basolateral amygdala (BLA) obstructed this preference while leaving social discrimination intact, indicating that these projections are necessary for socially driven choice. Further, mice that performed the task had greater densities of dendritic spines on excitatory BLA neurons relative to mice that did not. We next induced chemogenetic receptors in cells active during social interactions-when mice were encoding information that impacted later behavior. BLA neurons stimulated by social experience were necessary for mice to later favor rewards associated with social conspecifics but not make other choices. This profile contrasted with that of PL neurons stimulated by social experience, which were necessary for choice behavior in social and nonsocial contexts alike. The PL may convey a generalized signal allowing mice to favor particular rewards, while units in the BLA process more specialized information, together supporting choice motivated by social information.
Collapse
Affiliation(s)
- Henry W Kietzman
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut 06510
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia 30322
- Department of Psychiatry, Emory University School of Medicine, Atlanta, Georgia 30322
- Graduate Program in Neuroscience, Emory University, Atlanta, Georgia 30322
- Emory National Primate Research Center, Emory University, Atlanta, Georgia 30329
| | - Gracy Trinoskey-Rice
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia 30322
- Department of Psychiatry, Emory University School of Medicine, Atlanta, Georgia 30322
- Emory National Primate Research Center, Emory University, Atlanta, Georgia 30329
| | - Esther H Seo
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia 30322
- Department of Psychiatry, Emory University School of Medicine, Atlanta, Georgia 30322
- Emory National Primate Research Center, Emory University, Atlanta, Georgia 30329
| | - Jidong Guo
- Emory National Primate Research Center, Emory University, Atlanta, Georgia 30329
| | - Shannon L Gourley
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia 30322
- Department of Psychiatry, Emory University School of Medicine, Atlanta, Georgia 30322
- Graduate Program in Neuroscience, Emory University, Atlanta, Georgia 30322
- Emory National Primate Research Center, Emory University, Atlanta, Georgia 30329
- Children's Healthcare of Atlanta, Atlanta, Georgia 30322
| |
Collapse
|
9
|
Mews P, Sosnick L, Gurung A, Sidoli S, Nestler EJ. Decoding cocaine-induced proteomic adaptations in the mouse nucleus accumbens. Sci Signal 2024; 17:eadl4738. [PMID: 38626009 PMCID: PMC11170322 DOI: 10.1126/scisignal.adl4738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Accepted: 03/28/2024] [Indexed: 04/18/2024]
Abstract
Cocaine use disorder (CUD) is a chronic neuropsychiatric condition that results from enduring cellular and molecular adaptations. Among substance use disorders, CUD is notable for its rising prevalence and the lack of approved pharmacotherapies. The nucleus accumbens (NAc), a region that is integral to the brain's reward circuitry, plays a crucial role in the initiation and continuation of maladaptive behaviors that are intrinsic to CUD. Leveraging advancements in neuroproteomics, we undertook a proteomic analysis that spanned membrane, cytosolic, nuclear, and chromatin compartments of the NAc in a mouse model. The results unveiled immediate and sustained proteomic modifications after cocaine exposure and during prolonged withdrawal. We identified congruent protein regulatory patterns during initial cocaine exposure and reexposure after withdrawal, which contrasted with distinct patterns during withdrawal. Pronounced proteomic shifts within the membrane compartment indicated adaptive and long-lasting molecular responses prompted by cocaine withdrawal. In addition, we identified potential protein translocation events between soluble-nuclear and chromatin-bound compartments, thus providing insight into intracellular protein dynamics after cocaine exposure. Together, our findings illuminate the intricate proteomic landscape that is altered in the NAc by cocaine use and provide a dataset for future research toward potential therapeutics.
Collapse
Affiliation(s)
- Philipp Mews
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Lucas Sosnick
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ashik Gurung
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Simone Sidoli
- Department of Biochemistry, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Eric J. Nestler
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
10
|
Bjornson KJ, Vanderplow AM, Yang Y, Anderson DR, Kermath BA, Cahill ME. Stress-mediated dysregulation of the Rap1 small GTPase impairs hippocampal structure and function. iScience 2023; 26:107566. [PMID: 37664580 PMCID: PMC10470260 DOI: 10.1016/j.isci.2023.107566] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 05/15/2023] [Accepted: 08/02/2023] [Indexed: 09/05/2023] Open
Abstract
The effects of repeated stress on cognitive impairment are thought to be mediated, at least in part, by reductions in the stability of dendritic spines in brain regions critical for proper learning and memory, including the hippocampus. Small GTPases are particularly potent regulators of dendritic spine formation, stability, and morphology in hippocampal neurons. Through the use of small GTPase protein profiling in mice, we identify increased levels of synaptic Rap1 in the hippocampal CA3 region in response to escalating, intermittent stress. We then demonstrate that increased Rap1 in the CA3 is sufficient in and of itself to produce stress-relevant dendritic spine and cognitive phenotypes. Further, using super-resolution imaging, we investigate how the pattern of Rap1 trafficking to synapses likely underlies its effects on the stability of select dendritic spine subtypes. These findings illuminate the involvement of aberrant Rap1 regulation in the hippocampus in contributing to the psychobiological effects of stress.
Collapse
Affiliation(s)
- Kathryn J. Bjornson
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Amanda M. Vanderplow
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Yezi Yang
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Danielle R. Anderson
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Bailey A. Kermath
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Michael E. Cahill
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| |
Collapse
|
11
|
Zanella D, Smith NK, Hardaway JA, Buchanan AM, Mullins CH, Galli A, Carter AM. Bile acids modulate reinstatement of cocaine conditioned place preference and accumbal dopamine dynamics without compromising appetitive learning. Sci Rep 2023; 13:13359. [PMID: 37591972 PMCID: PMC10435481 DOI: 10.1038/s41598-023-40456-3] [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: 06/08/2023] [Accepted: 08/10/2023] [Indexed: 08/19/2023] Open
Abstract
Psychostimulants target the dopamine transporter (DAT) to elicit their psychomotor actions. Bile acids (BAs) can also bind to DAT and reduce behavioral responses to cocaine, suggesting a potential therapeutic application of BAs in psychostimulant use disorder. Here, we investigate the potential of BAs to decrease drug-primed reinstatement when administered during an abstinence phase. To do this, after successful development of cocaine-associated contextual place preference (cocaine CPP), cocaine administration was terminated, and animals treated with vehicle or obeticholic acid (OCA). When preference for the cocaine-associated context was extinguished, mice were challenged with a single priming dose of cocaine, and reinstatement of cocaine-associated contextual preference was measured. Animals treated with OCA demonstrate a significantly lower reinstatement for cocaine CPP. OCA also impairs the ability of cocaine to reduce the clearance rate of electrically stimulated dopamine release and diminishes the area under the curve (AUC) observed with amperometry. Furthermore, the AUC of the amperometric signal positively correlates with the reinstatement index. Using operant feeding devices, we demonstrate that OCA has no effect on contextual learning or motivation for natural rewards. These data highlight OCA as a potential therapeutic for cocaine use disorder.
Collapse
Affiliation(s)
- Daniele Zanella
- Department of Surgery, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, USA
| | - Nicholas K Smith
- Department of Biology, University of Pennsylvania, Philadelphia, USA
| | - J Andrew Hardaway
- Department of Psychiatry and Behavioral Neurobiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, USA
| | - Anna Marie Buchanan
- Department of Surgery, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, USA
| | - Clarence H Mullins
- Department of Surgery, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, USA
| | - Aurelio Galli
- Department of Surgery, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, USA
- Center for Inter-Systemic Networks and Enteric Medical Advances (UAB CINEMA), Birmingham, USA
| | - Angela M Carter
- Department of Surgery, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, USA.
- Center for Inter-Systemic Networks and Enteric Medical Advances (UAB CINEMA), Birmingham, USA.
| |
Collapse
|
12
|
Ru Q, Wang Y, Zhou E, Chen L, Wu Y. The potential therapeutic roles of Rho GTPases in substance dependence. Front Mol Neurosci 2023; 16:1125277. [PMID: 37063367 PMCID: PMC10097952 DOI: 10.3389/fnmol.2023.1125277] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 03/14/2023] [Indexed: 04/03/2023] Open
Abstract
Rho GTPases family are considered to be molecular switches that regulate various cellular processes, including cytoskeleton remodeling, cell polarity, synaptic development and maintenance. Accumulating evidence shows that Rho GTPases are involved in neuronal development and brain diseases, including substance dependence. However, the functions of Rho GTPases in substance dependence are divergent and cerebral nuclei-dependent. Thereby, comprehensive integration of their roles and correlated mechanisms are urgently needed. In this review, the molecular functions and regulatory mechanisms of Rho GTPases and their regulators such as GTPase-activating proteins (GAPs) and guanine nucleotide exchange factors (GEFs) in substance dependence have been reviewed, and this is of great significance for understanding their spatiotemporal roles in addictions induced by different addictive substances and in different stages of substance dependence.
Collapse
Affiliation(s)
| | | | | | - Lin Chen
- *Correspondence: Lin Chen, ; Yuxiang Wu,
| | - Yuxiang Wu
- *Correspondence: Lin Chen, ; Yuxiang Wu,
| |
Collapse
|
13
|
Engeln M, Fox ME, Chandra R, Choi EY, Nam H, Qadir H, Thomas SS, Rhodes VM, Turner MD, Herman RJ, Calarco CA, Lobo MK. Transcriptome profiling of the ventral pallidum reveals a role for pallido-thalamic neurons in cocaine reward. Mol Psychiatry 2022; 27:3980-3991. [PMID: 35764708 PMCID: PMC9722585 DOI: 10.1038/s41380-022-01668-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 05/28/2022] [Accepted: 06/08/2022] [Indexed: 02/06/2023]
Abstract
Psychostimulant exposure alters the activity of ventral pallidum (VP) projection neurons. However, the molecular underpinnings of these circuit dysfunctions are unclear. We used RNA-sequencing to reveal alterations in the transcriptional landscape of the VP that are induced by cocaine self-administration in mice. We then probed gene expression in select VP neuronal subpopulations to isolate a circuit associated with cocaine intake. Finally, we used both overexpression and CRISPR-mediated knockdown to test the role of a gene target on cocaine-mediated behaviors as well as dendritic spine density. Our results showed that a large proportion (55%) of genes associated with structural plasticity were changed 24 h following cocaine intake. Among them, the transcription factor Nr4a1 (Nuclear receptor subfamily 4, group A, member 1, or Nur77) showed high expression levels. We found that the VP to mediodorsal thalamus (VP → MDT) projection neurons specifically were recapitulating this increase in Nr4a1 expression. Overexpressing Nr4a1 in VP → MDT neurons enhanced drug-seeking and drug-induced reinstatement, while Nr4a1 knockdown prevented self-administration acquisition and subsequent cocaine-mediated behaviors. Moreover, we showed that Nr4a1 negatively regulated spine dynamics in this specific cell subpopulation. Together, our study identifies for the first time the transcriptional mechanisms occurring in VP in drug exposure. Our study provides further understanding on the role of Nr4a1 in cocaine-related behaviors and identifies the crucial role of the VP → MDT circuit in drug intake and relapse-like behaviors.
Collapse
Affiliation(s)
- Michel Engeln
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA.
- Univ. Bordeaux, CNRS, INCIA, UMR 5287, F-33000, Bordeaux, France.
| | - Megan E Fox
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Anesthesiology & Perioperative Medicine, Penn State College of Medicine, Hershey, PA, USA
| | - Ramesh Chandra
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Eric Y Choi
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Hyungwoo Nam
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Houman Qadir
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Shavin S Thomas
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Victoria M Rhodes
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Makeda D Turner
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Rae J Herman
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Cali A Calarco
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Mary Kay Lobo
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
14
|
Wang Y, Spolitu S, Zadroga JA, Sarecha AK, Ozcan L. Hepatocyte Rap1a contributes to obesity- and statin-associated hyperglycemia. Cell Rep 2022; 40:111259. [PMID: 36001955 PMCID: PMC9446800 DOI: 10.1016/j.celrep.2022.111259] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 05/26/2022] [Accepted: 08/03/2022] [Indexed: 12/28/2022] Open
Abstract
Excessive hepatic glucose production contributes to the development of hyperglycemia and is a key feature of type 2 diabetes. Here, we report that activation of hepatocyte Rap1a suppresses gluconeogenic gene expression and glucose production, whereas Rap1a silencing stimulates them. Rap1a activation is suppressed in obese mouse liver, and restoring its activity improves glucose intolerance. As Rap1a′s membrane localization and activation depends on its geranylgeranylation, which is inhibited by statins, we show that statin-treated hepatocytes and the human liver have lower active-Rap1a levels. Similar to Rap1a inhibition, statins stimulate hepatic gluconeogenesis and increase fasting blood glucose in obese mice. Geranylgeraniol treatment, which acts as the precursor for geranylgeranyl isoprenoids, restores Rap1a activity and improves statin-mediated glucose intolerance. Mechanistically, Rap1a activation induces actin polymerization, which suppresses gluconeogenesis by Akt-mediated FoxO1 inhibition. Thus, Rap1a regulates hepatic glucose homeostasis, and blocking its activity, via lowering geranylgeranyl isoprenoids, contributes to statin-induced glucose intolerance. Wang et al. show that activation of hepatic Rap1a suppresses gluconeogenic gene expression and improves glucose intolerance via Akt-mediated FoxO1 inhibition. Statins lower intracellular isoprenoid levels and inhibit Rap1a activation, which contributes to their hyperglycemic effect. These findings identify a role of hepatic Rap1a in obesity- and statin-associated glucose homeostasis.
Collapse
Affiliation(s)
- Yating Wang
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Stefano Spolitu
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - John A Zadroga
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Amesh K Sarecha
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Lale Ozcan
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA.
| |
Collapse
|
15
|
Liu X, Vickstrom CR, Yu H, Liu S, Snarrenberg ST, Friedman V, Mu L, Chen B, Kelly TJ, Baker DA, Liu QS. Epac2 in midbrain dopamine neurons contributes to cocaine reinforcement via enhancement of dopamine release. eLife 2022; 11:e80747. [PMID: 35993549 PMCID: PMC9436413 DOI: 10.7554/elife.80747] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 08/21/2022] [Indexed: 11/21/2022] Open
Abstract
Repeated exposure to drugs of abuse results in an upregulation of cAMP signaling in the mesolimbic dopamine system, a molecular adaptation thought to be critically involved in the development of drug dependence. Exchange protein directly activated by cAMP (Epac2) is a major cAMP effector abundantly expressed in the brain. However, it remains unknown whether Epac2 contributes to cocaine reinforcement. Here, we report that Epac2 in the mesolimbic dopamine system promotes cocaine reinforcement via enhancement of dopamine release. Conditional knockout of Epac2 from midbrain dopamine neurons (Epac2-cKO) and the selective Epac2 inhibitor ESI-05 decreased cocaine self-administration in mice under both fixed-ratio and progressive-ratio reinforcement schedules and across a broad range of cocaine doses. In addition, Epac2-cKO led to reduced evoked dopamine release, whereas Epac2 agonism robustly enhanced dopamine release in the nucleus accumbens in vitro. This mechanism is central to the behavioral effects of Epac2 disruption, as chemogenetic stimulation of ventral tegmental area (VTA) dopamine neurons via deschloroclozapine (DCZ)-induced activation of Gs-DREADD increased dopamine release and reversed the impairment of cocaine self-administration in Epac2-cKO mice. Conversely, chemogenetic inhibition of VTA dopamine neurons with Gi-DREADD reduced dopamine release and cocaine self-administration in wild-type mice. Epac2-mediated enhancement of dopamine release may therefore represent a novel and powerful mechanism that contributes to cocaine reinforcement.
Collapse
Affiliation(s)
- Xiaojie Liu
- Department of Pharmacology and Toxicology, Medical College of WisconsinMilwaukeeUnited States
| | - Casey R Vickstrom
- Department of Pharmacology and Toxicology, Medical College of WisconsinMilwaukeeUnited States
| | - Hao Yu
- Department of Pharmacology and Toxicology, Medical College of WisconsinMilwaukeeUnited States
| | - Shuai Liu
- Department of Pharmacology and Toxicology, Medical College of WisconsinMilwaukeeUnited States
| | - Shana Terai Snarrenberg
- Department of Pharmacology and Toxicology, Medical College of WisconsinMilwaukeeUnited States
| | - Vladislav Friedman
- Department of Pharmacology and Toxicology, Medical College of WisconsinMilwaukeeUnited States
| | - Lianwei Mu
- Department of Pharmacology and Toxicology, Medical College of WisconsinMilwaukeeUnited States
| | - Bixuan Chen
- Department of Pharmacology and Toxicology, Medical College of WisconsinMilwaukeeUnited States
| | - Thomas J Kelly
- Department of Pharmacology and Toxicology, Medical College of WisconsinMilwaukeeUnited States
| | - David A Baker
- Department of Biomedical Sciences, Marquette UniversityMilwaukeeUnited States
| | - Qing-song Liu
- Department of Pharmacology and Toxicology, Medical College of WisconsinMilwaukeeUnited States
| |
Collapse
|
16
|
Li H, Zhou X, Chen R, Xiao Y, Zhou T. The Src-Kinase Fyn is Required for Cocaine-Associated Memory Through Regulation of Tau. Front Pharmacol 2022; 13:769827. [PMID: 35185557 PMCID: PMC8850722 DOI: 10.3389/fphar.2022.769827] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 01/10/2022] [Indexed: 11/24/2022] Open
Abstract
Drug-associated context-induced relapse of cocaine-seeking behaviour requires the retrieval of drug-associated memory. Studies exploring the underlying neurobiological mechanism of drug memory formation will likely contribute to the development of treatments for drug addiction and the prevention of relapse. In our study, we applied a cocaine-conditioned place preference (CPP) paradigm and a self-administration paradigm (two drug-associated memory formation model) to confirm the hypothesis that the Src kinase Fyn critically regulates cocaine-associated memory formation in the hippocampus. For this experiment, we administered the Src kinase inhibitor PP2 into the bilateral hippocampus before cocaine-CPP and self-administration training, and the results showed that pharmacological manipulation of the Src kinase Fyn activity significantly attenuated the response to cocaine-paired cues in the cocaine-CPP and self-administration paradigms, indicating that hippocampal Fyn activity contributes to cocaine-associated memory formation. In addition, the regulation of cocaine-associated memory formation by Fyn depends on Tau expression, as restoring Tau to normal levels disrupted cocaine memory formation. Together, these results indicate that hippocampal Fyn activity plays a key role in the formation of cocaine-associated memory, which underlies cocaine-associated contextual stimulus-mediated regulation of cocaine-seeking behaviour, suggesting that Fyn represents a promising therapeutic target for weakening cocaine-related memory and treating cocaine addiction.
Collapse
Affiliation(s)
- Hongchun Li
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Hongchun Li,
| | - Xinglong Zhou
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, West China Hospital, Sichuan University, Chengdu, China
| | - Rong Chen
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yuzhou Xiao
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Tao Zhou
- Department of Drug and Equipment, China Rongtong Bayi Orthopaedic Hospital, Chengdu, China
| |
Collapse
|
17
|
Vanderplow AM, Kermath BA, Bernhardt CR, Gums KT, Seablom EN, Radcliff AB, Ewald AC, Jones MV, Baker TL, Watters JJ, Cahill ME. A feature of maternal sleep apnea during gestation causes autism-relevant neuronal and behavioral phenotypes in offspring. PLoS Biol 2022; 20:e3001502. [PMID: 35113852 PMCID: PMC8812875 DOI: 10.1371/journal.pbio.3001502] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 11/29/2021] [Indexed: 12/26/2022] Open
Abstract
Mounting epidemiologic and scientific evidence indicates that many psychiatric disorders originate from a complex interplay between genetics and early life experiences, particularly in the womb. Despite decades of research, our understanding of the precise prenatal and perinatal experiences that increase susceptibility to neurodevelopmental disorders remains incomplete. Sleep apnea (SA) is increasingly common during pregnancy and is characterized by recurrent partial or complete cessations in breathing during sleep. SA causes pathological drops in blood oxygen levels (intermittent hypoxia, IH), often hundreds of times each night. Although SA is known to cause adverse pregnancy and neonatal outcomes, the long-term consequences of maternal SA during pregnancy on brain-based behavioral outcomes and associated neuronal functioning in the offspring remain unknown. We developed a rat model of maternal SA during pregnancy by exposing dams to IH, a hallmark feature of SA, during gestational days 10 to 21 and investigated the consequences on the offspring's forebrain synaptic structure, synaptic function, and behavioral phenotypes across multiples stages of development. Our findings represent a rare example of prenatal factors causing sexually dimorphic behavioral phenotypes associated with excessive (rather than reduced) synapse numbers and implicate hyperactivity of the mammalian target of rapamycin (mTOR) pathway in contributing to the behavioral aberrations. These findings have implications for neuropsychiatric disorders typified by superfluous synapse maintenance that are believed to result, at least in part, from largely unknown insults to the maternal environment.
Collapse
Affiliation(s)
- Amanda M. Vanderplow
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Bailey A. Kermath
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Cassandra R. Bernhardt
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Kimberly T. Gums
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Erin N. Seablom
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Abigail B. Radcliff
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Andrea C. Ewald
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Mathew V. Jones
- Department of Neuroscience, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Tracy L. Baker
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Jyoti J. Watters
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Michael E. Cahill
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| |
Collapse
|
18
|
Allichon MC, Ortiz V, Pousinha P, Andrianarivelo A, Petitbon A, Heck N, Trifilieff P, Barik J, Vanhoutte P. Cell-Type-Specific Adaptions in Striatal Medium-Sized Spiny Neurons and Their Roles in Behavioral Responses to Drugs of Abuse. Front Synaptic Neurosci 2022; 13:799274. [PMID: 34970134 PMCID: PMC8712310 DOI: 10.3389/fnsyn.2021.799274] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 11/26/2021] [Indexed: 12/21/2022] Open
Abstract
Drug addiction is defined as a compulsive pattern of drug-seeking- and taking- behavior, with recurrent episodes of abstinence and relapse, and a loss of control despite negative consequences. Addictive drugs promote reinforcement by increasing dopamine in the mesocorticolimbic system, which alters excitatory glutamate transmission within the reward circuitry, thereby hijacking reward processing. Within the reward circuitry, the striatum is a key target structure of drugs of abuse since it is at the crossroad of converging glutamate inputs from limbic, thalamic and cortical regions, encoding components of drug-associated stimuli and environment, and dopamine that mediates reward prediction error and incentive values. These signals are integrated by medium-sized spiny neurons (MSN), which receive glutamate and dopamine axons converging onto their dendritic spines. MSN primarily form two mostly distinct populations based on the expression of either DA-D1 (D1R) or DA-D2 (D2R) receptors. While a classical view is that the two MSN populations act in parallel, playing antagonistic functional roles, the picture seems much more complex. Herein, we review recent studies, based on the use of cell-type-specific manipulations, demonstrating that dopamine differentially modulates dendritic spine density and synapse formation, as well as glutamate transmission, at specific inputs projecting onto D1R-MSN and D2R-MSN to shape persistent pathological behavioral in response to drugs of abuse. We also discuss the identification of distinct molecular events underlying the detrimental interplay between dopamine and glutamate signaling in D1R-MSN and D2R-MSN and highlight the relevance of such cell-type-specific molecular studies for the development of innovative strategies with potential therapeutic value for addiction. Because drug addiction is highly prevalent in patients with other psychiatric disorders when compared to the general population, we last discuss the hypothesis that shared cellular and molecular adaptations within common circuits could explain the co-occurrence of addiction and depression. We will therefore conclude this review by examining how the nucleus accumbens (NAc) could constitute a key interface between addiction and depression.
Collapse
Affiliation(s)
- Marie-Charlotte Allichon
- CNRS, UMR 8246, Neuroscience Paris Seine, Paris, France.,INSERM, UMR-S 1130, Neuroscience Paris Seine, Institute of Biology Paris Seine, Paris, France.,Sorbonne Université, UPMC Université Paris 06, UM CR18, Neuroscience Paris Seine, Paris, France
| | - Vanesa Ortiz
- Université Côte d'Azur, Nice, France.,Institut de Pharmacologie Moléculaire et Cellulaire, CNRS UMR 7275, Valbonne, France
| | - Paula Pousinha
- Université Côte d'Azur, Nice, France.,Institut de Pharmacologie Moléculaire et Cellulaire, CNRS UMR 7275, Valbonne, France
| | - Andry Andrianarivelo
- CNRS, UMR 8246, Neuroscience Paris Seine, Paris, France.,INSERM, UMR-S 1130, Neuroscience Paris Seine, Institute of Biology Paris Seine, Paris, France.,Sorbonne Université, UPMC Université Paris 06, UM CR18, Neuroscience Paris Seine, Paris, France
| | - Anna Petitbon
- Université Bordeaux, INRAE, Bordeaux INP, NutriNeuro, Bordeaux, France
| | - Nicolas Heck
- CNRS, UMR 8246, Neuroscience Paris Seine, Paris, France.,INSERM, UMR-S 1130, Neuroscience Paris Seine, Institute of Biology Paris Seine, Paris, France.,Sorbonne Université, UPMC Université Paris 06, UM CR18, Neuroscience Paris Seine, Paris, France
| | - Pierre Trifilieff
- Université Bordeaux, INRAE, Bordeaux INP, NutriNeuro, Bordeaux, France
| | - Jacques Barik
- Université Côte d'Azur, Nice, France.,Institut de Pharmacologie Moléculaire et Cellulaire, CNRS UMR 7275, Valbonne, France
| | - Peter Vanhoutte
- CNRS, UMR 8246, Neuroscience Paris Seine, Paris, France.,INSERM, UMR-S 1130, Neuroscience Paris Seine, Institute of Biology Paris Seine, Paris, France.,Sorbonne Université, UPMC Université Paris 06, UM CR18, Neuroscience Paris Seine, Paris, France
| |
Collapse
|
19
|
Neuroplasticity and Multilevel System of Connections Determine the Integrative Role of Nucleus Accumbens in the Brain Reward System. Int J Mol Sci 2021; 22:ijms22189806. [PMID: 34575969 PMCID: PMC8471564 DOI: 10.3390/ijms22189806] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 09/06/2021] [Accepted: 09/08/2021] [Indexed: 12/27/2022] Open
Abstract
A growing body of evidence suggests that nucleus accumbens (NAc) plays a significant role not only in the physiological processes associated with reward and satisfaction but also in many diseases of the central nervous system. Summary of the current state of knowledge on the morphological and functional basis of such a diverse function of this structure may be a good starting point for further basic and clinical research. The NAc is a part of the brain reward system (BRS) characterized by multilevel organization, extensive connections, and several neurotransmitter systems. The unique role of NAc in the BRS is a result of: (1) hierarchical connections with the other brain areas, (2) a well-developed morphological and functional plasticity regulating short- and long-term synaptic potentiation and signalling pathways, (3) cooperation among several neurotransmitter systems, and (4) a supportive role of neuroglia involved in both physiological and pathological processes. Understanding the complex function of NAc is possible by combining the results of morphological studies with molecular, genetic, and behavioral data. In this review, we present the current views on the NAc function in physiological conditions, emphasizing the role of its connections, neuroplasticity processes, and neurotransmitter systems.
Collapse
|
20
|
Zhu H, Zhuang D, Lou Z, Lai M, Fu D, Hong Q, Liu H, Zhou W. Akt and its phosphorylation in nucleus accumbens mediate heroin-seeking behavior induced by cues in rats. Addict Biol 2021; 26:e13013. [PMID: 33619816 PMCID: PMC8459226 DOI: 10.1111/adb.13013] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 01/07/2021] [Accepted: 01/14/2021] [Indexed: 01/14/2023]
Abstract
Akt is initially identified as one of the downstream targets of phosphatidylinositol-3 kinase (PI3K) and is involved in morphine reward and tolerance. However, whether phospholyration of Akt (p-Akt) mediates heroin relapse remains unclear. Here, we aimed to explore the role of p-Akt in the nucleus accumbens (NAc) in cue-induced heroin-seeking behaviors after withdrawal. First, rats were trained to self-administer heroin for 14 days, after which we assessed heroin-seeking behaviors induced by a context cue (CC) or by discrete conditioned cues (CS) after 1 day or 14 days of withdrawal. We found that the active responses induced by CC or CS after 14 days of withdrawal were higher than those after 1 day of withdrawal. Meanwhile, the expression of p-Akt in the NAc was also greatest when rats were exposed to the CS after 14 days of withdrawal. Additionally, a microinjection of LY294002, an inhibitor of PI3K, into the NAc inhibited the CS-induced heroin-seeking behaviors after 14 days of withdrawal, paralleling the decreased levels of p-Akt in the NAc. Finally, Akt1 or β-arrestin 2 was downregulated via a lentiviral injection to assess the effect on heroin seeking after 14 days of withdrawal. CS-induced heroin-seeking behavior was inhibited by downregulation of Akt1, but not β-arrestin 2, in the NAc. These data demonstrate that Akt phosphorylation in the NAc may play an important role in the incubation of heroin-seeking behavior, suggesting that the PI3K/Akt pathways may be involved in the process of heroin relapse and addiction.
Collapse
Affiliation(s)
- Huaqiang Zhu
- Zhejiang Provincial Key Laboratory of Addiction Research, Ningbo Kangning Hospital, School of Medicine Ningbo University China
| | - Dingding Zhuang
- Zhejiang Provincial Key Laboratory of Addiction Research, Ningbo Kangning Hospital, School of Medicine Ningbo University China
| | - Zhongze Lou
- Department of Psychosomatic Medicine, Ningbo First Hospital Ningbo Hospital of Zhejiang University China
| | - Miaojun Lai
- Zhejiang Provincial Key Laboratory of Addiction Research, Ningbo Kangning Hospital, School of Medicine Ningbo University China
| | - Dan Fu
- Zhejiang Provincial Key Laboratory of Addiction Research, Ningbo Kangning Hospital, School of Medicine Ningbo University China
| | - Qingxiao Hong
- Zhejiang Provincial Key Laboratory of Addiction Research, Ningbo Kangning Hospital, School of Medicine Ningbo University China
| | - Huifen Liu
- Zhejiang Provincial Key Laboratory of Addiction Research, Ningbo Kangning Hospital, School of Medicine Ningbo University China
| | - Wenhua Zhou
- Zhejiang Provincial Key Laboratory of Addiction Research, Ningbo Kangning Hospital, School of Medicine Ningbo University China
| |
Collapse
|
21
|
Spatiotemporal expression of Rap1 and Ras mediates the acquisition and reinstatement of methamphetamine-induced conditioned place preference in mice via extracellular signal-regulated kinase activation. Neuroreport 2021; 32:1035-1040. [PMID: 34232127 DOI: 10.1097/wnr.0000000000001686] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Drug addiction is a chronic recurrent brain disease characterized by compulsive drug use and a high tendency to relapse. We previously reported that the Ras-extracellular signal-regulated kinase (ERK)-ΔFosB pathway in the caudate putamen (CPu) was involved in methamphetamine-induced behavioral sensitization. Rap1, as an antagonist of Ras originally, was found to participate in neuronal synaptic plasticity recently, but the role of Rap1 in methamphetamine addiction is unclear. First, in this study, we constructed the acquisition, extinction and reinstatement of methamphetamine-induced conditioned place preference (CPP) in mice, respectively. Then, protein levels of Rap1, Ras and pERK/ERK in the prefrontal cortex (PFc), CPu and hippocampus of CPP mice on three phases were detected. We found that protein levels of Rap1, Ras and pERK/ERK in the CPu were significantly increased after repeated methamphetamine administration, as well as Rap1 and pERK/ERK in the hippocampus. However, protein levels of Rap1 and pERK/ERK in the CPu were decreased on the reinstatement of CPP mice. Therefore, Rap1 and Ras in the CPu and Rap1 in the hippocampus may participate in the regulation of the acquisition of methamphetamine-induced CPP in mice by activating ERK. Moreover, Rap1-ERK cascade in the CPu contributes to both the acquisition and reinstatement of methamphetamine-induced CPP in mice.
Collapse
|
22
|
Piva A, Caffino L, Mottarlini F, Pintori N, Castillo Díaz F, Fumagalli F, Chiamulera C. Metaplastic Effects of Ketamine and MK-801 on Glutamate Receptors Expression in Rat Medial Prefrontal Cortex and Hippocampus. Mol Neurobiol 2021; 58:3443-3456. [PMID: 33723767 PMCID: PMC8257545 DOI: 10.1007/s12035-021-02352-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 03/05/2021] [Indexed: 12/16/2022]
Abstract
Ketamine and MK-801 by blocking NMDA receptors may induce reinforcing effects as well as schizophrenia-like symptoms. Recent results showed that ketamine can also effectively reverse depressive signs in patients' refractory to standard therapies. This evidence clearly points to the need of characterization of effects of these NMDARs antagonists on relevant brain areas for mood disorders. The aim of the present study was to investigate the molecular changes occurring at glutamatergic synapses 24 h after ketamine or MK-801 treatment in the rat medial prefrontal cortex (mPFC) and hippocampus (Hipp). In particular, we analyzed the levels of the glutamate transporter-1 (GLT-1), NMDA receptors, AMPA receptors subunits, and related scaffolding proteins. In the homogenate, we found a general decrease of protein levels, whereas their changes in the post-synaptic density were more complex. In fact, ketamine in the mPFC decreased the level of GLT-1 and increased the level of GluN2B, GluA1, GluA2, and scaffolding proteins, likely indicating a pattern of enhanced excitability. On the other hand, MK-801 only induced sparse changes with apparently no correlation to functional modification. Differently from mPFC, in Hipp, both substances reduced or caused no changes of glutamate receptors and scaffolding proteins expression. Ketamine decreased NMDA receptors while increased AMPA receptors subunit ratios, an effect indicative of permissive metaplastic modulation; conversely, MK-801 only decreased the latter, possibly representing a blockade of further synaptic plasticity. Taken together, these findings indicate a fine tuning of glutamatergic synapses by ketamine compared to MK-801 both in the mPFC and Hipp.
Collapse
Affiliation(s)
- Alessandro Piva
- Neuropsychopharmacology Lab, Section Pharmacology, Department Diagnostic & Public Health, University of Verona, Policlinico GB Rossi, P.le Scuro 10, 37134, Verona, Italy.
| | - Lucia Caffino
- Department of Pharmacological and Biomolecular Sciences, University of Milano, Via Balzaretti 9, 20133, Milano, Italy
| | - Francesca Mottarlini
- Department of Pharmacological and Biomolecular Sciences, University of Milano, Via Balzaretti 9, 20133, Milano, Italy
| | - Nicholas Pintori
- Neuropsychopharmacology Lab, Section Pharmacology, Department Diagnostic & Public Health, University of Verona, Policlinico GB Rossi, P.le Scuro 10, 37134, Verona, Italy
| | - Fernando Castillo Díaz
- Department of Pharmacological and Biomolecular Sciences, University of Milano, Via Balzaretti 9, 20133, Milano, Italy
| | - Fabio Fumagalli
- Department of Pharmacological and Biomolecular Sciences, University of Milano, Via Balzaretti 9, 20133, Milano, Italy
| | - Cristiano Chiamulera
- Neuropsychopharmacology Lab, Section Pharmacology, Department Diagnostic & Public Health, University of Verona, Policlinico GB Rossi, P.le Scuro 10, 37134, Verona, Italy
| |
Collapse
|
23
|
The Rap1 small GTPase is a critical mediator of the effects of stress on prefrontal cortical dysfunction. Mol Psychiatry 2021; 26:3223-3239. [PMID: 32651478 DOI: 10.1038/s41380-020-0835-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 06/23/2020] [Accepted: 06/30/2020] [Indexed: 02/06/2023]
Abstract
The neural molecular and biochemical response to stress is a distinct physiological process, and multiple lines of evidence indicate that the prefrontal cortex (PFC) is particularly sensitive to, and afflicted by, exposure to stress. Largely through this PFC dysfunction, stress has a characterized role in facilitating cognitive impairment, which is often dissociable from its effects on non-cognitive behaviors. The Rap1 small GTPase pathway has emerged as a commonly disrupted intracellular target in neuropsychiatric conditions, whether it be via alterations in Rap1 expression or through alterations in the expression of direct and specific upstream Rap1 activators and inhibitors. Here we demonstrate that escalating, intermittent stress increases Rap1 in mouse PFC synapses, results in cognitive impairments, and reduces the preponderance of mature dendritic spines in PFC neurons. Using viral-mediated gene transfer, we reveal that the hyper-induction of Rap1 in the PFC is sufficient to drive stress-relevant cognitive and synaptic phenotypes. These findings point to Rap1 as a critical mediator of stress-driven neuronal and behavioral pathology and highlight a previously unrecognized involvement for Rap1 in novelty-driven PFC engagement.
Collapse
|
24
|
Vanderplow AM, Eagle AL, Kermath BA, Bjornson KJ, Robison AJ, Cahill ME. Akt-mTOR hypoactivity in bipolar disorder gives rise to cognitive impairments associated with altered neuronal structure and function. Neuron 2021; 109:1479-1496.e6. [PMID: 33765445 PMCID: PMC8105282 DOI: 10.1016/j.neuron.2021.03.008] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 01/20/2021] [Accepted: 03/04/2021] [Indexed: 12/22/2022]
Abstract
The Akt family of kinases exerts many of its cellular effects via the activation of the mammalian target of rapamycin (mTOR) kinase through a series of intermediary proteins. Multiple lines of evidence have identified Akt-family kinases as candidate schizophrenia and bipolar disorder genes. Although dysfunction of the prefrontal cortex (PFC) is a key feature of both schizophrenia and bipolar disorder, no studies have comprehensively assessed potential alterations in Akt-mTOR pathway activity in the PFC of either disorder. Here, we examined the activity and expression profile of key proteins in the Akt-mTOR pathway in bipolar disorder and schizophrenia homogenates from two different PFC subregions. Our findings identify reduced Akt-mTOR PFC signaling in a subset of bipolar disorder subjects. Using a reverse-translational approach, we demonstrated that Akt hypofunction in the PFC is sufficient to give rise to key cognitive phenotypes that are paralleled by alterations in synaptic connectivity and function.
Collapse
Affiliation(s)
- Amanda M Vanderplow
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Andrew L Eagle
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
| | - Bailey A Kermath
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Kathryn J Bjornson
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Alfred J Robison
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
| | - Michael E Cahill
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA.
| |
Collapse
|
25
|
Kermath BA, Vanderplow AM, Cahill ME. Dysregulated Prefrontal Cortical RhoA Signal Transduction in Bipolar Disorder with Psychosis: New Implications for Disease Pathophysiology. Cereb Cortex 2021; 30:59-71. [PMID: 31220216 DOI: 10.1093/cercor/bhz070] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
While research has identified alterations in dorsolateral prefrontal cortical function as a key factor to the etiology of bipolar disorder, few studies have uncovered robust changes in protein signal transduction pathways in this disorder. Given the direct relevance of protein-based expressional alterations to cellular functions and because many of the key regulatory mechanisms for the disease pathogenesis likely include alterations in protein activity rather than changes in expression alone, the identification of alterations in discrete signal transduction pathways in bipolar disorder would have broad implications for understanding the disease pathophysiology. As prior microarray data point to a previously unrecognized involvement of the RhoA network in bipolar disorder, here we investigate the protein expression and activity of key components of a RhoA signal transduction pathway in dorsolateral prefrontal cortical homogenates from subjects with bipolar disorder. The results of this investigation implicate overactivation of prefrontal cortical RhoA signaling in specific subtypes of bipolar disorder. The specificity of these findings is demonstrated by a lack of comparable changes in schizophrenia; however, our findings do identify convergence between both disorders at the level of activity-mediated actin cytoskeletal regulation. These findings have implications for understanding the altered cortical synaptic connectivity of bipolar disorder.
Collapse
Affiliation(s)
- Bailey A Kermath
- Department of Comparative Biosciences, University of Wisconsin at Madison, Madison, WI, USA
| | - Amanda M Vanderplow
- Department of Comparative Biosciences, University of Wisconsin at Madison, Madison, WI, USA
| | - Michael E Cahill
- Department of Comparative Biosciences, University of Wisconsin at Madison, Madison, WI, USA
| |
Collapse
|
26
|
Expression of a novel brain specific isoform of C3G is regulated during development. Sci Rep 2020; 10:18838. [PMID: 33139841 PMCID: PMC7606606 DOI: 10.1038/s41598-020-75813-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 10/01/2020] [Indexed: 12/13/2022] Open
Abstract
Mice lacking C3G (RapGEF1), a ubiquitously expressed protein essential for neuronal differentiation, show multiple defects in brain development. Function of C3G in neurogenesis is poorly defined. Here, we identify brain specific expression of a novel C3G isoform in mice and humans. This isoform has an insert in the Crk-binding region, generating a polypeptide of 175 kDa, unlike the previously known 140 kDa form expressed in all other tissues. In the adult mouse brain, C3G expression is seen in neurons, but was not detectable in GFAP-positive cells. C3G levels were high in the CA3 region of hippocampus and in mitral cells of olfactory bulb. Neural progenitor cells positive for Doublecortin and Nestin, show expression of C3G. During development, C3G is expressed in precursor cells prior to their differentiation into mature neurons or astrocytes. The 175 kDa as well as 140 kDa forms are seen in embryonic mouse brain, while only the 175 kDa variant is seen in post-natal brain. Human cerebral organoids generated from induced pluripotent stem cells predominantly expressed the 140 kDa polypeptides, and the 175 kDa isoform appeared upon maturation. This study describes developmental regulation and neuronal expression of a brain specific isoform of C3G, a molecule essential for normal development of the mammalian brain.
Collapse
|
27
|
ARHGEF11 promotes proliferation and epithelial-mesenchymal transition of hepatocellular carcinoma through activation of β-catenin pathway. Aging (Albany NY) 2020; 12:20235-20253. [PMID: 33122451 PMCID: PMC7655160 DOI: 10.18632/aging.103772] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 06/29/2020] [Indexed: 02/05/2023]
Abstract
Rho guanine nucleotide exchange factor 11 (ARHGEF11) has been proved to promote tumor metastasis in glioblastoma and ovarian carcinoma. However, the role of ARHGEF11 in hepatocellular carcinoma (HCC) progression is largely unknown. Here, we found that ARHGEF11 was upregulated in HCC samples and highly metastatic hepatoma cell lines. Knockdown of ARHGEFF11 inhibited the cell proliferation and invasion in both HCCLM3 and SKHEP1 cell lines. Subsequent mechanistic investigation showed that downregulation of ARHGEF11 significantly attenuated β-catenin nuclear translocation, thereafter repressed the expression of ZEB1 and cyclinD1, finally contributing to inhibition of epithelial-mesenchymal transition (EMT) and cell cycle arrest. Moreover, high levels of ARHGEF11 were found to be associated with shorter disease free and overall survival. A prognostic nomogram model that integrated ARHGEF11, tumor size and BCLC classification showed good performance in predicting clinical outcomes of HCC patients. Overall, this study demonstrated that ARHGEF11 could promote proliferation and metastasis of HCC via activating β-catenin pathway, suggesting that ARHGEF11 might serve as a potential prognostic biomarker for HCC.
Collapse
|
28
|
Chiamulera C, Piva A, Abraham WC. Glutamate receptors and metaplasticity in addiction. Curr Opin Pharmacol 2020; 56:39-45. [PMID: 33128937 DOI: 10.1016/j.coph.2020.09.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 09/10/2020] [Accepted: 09/10/2020] [Indexed: 12/12/2022]
Abstract
Chronic drug use is a neuroadaptive disorder characterized by strong and persistent plasticity in the mesocorticolimbic reward system. Long-lasting effects of drugs of abuse rely on their ability to hijack glutamate receptor activity and long-term synaptic plasticity processes like long-term potentiation and depression. Importantly, metaplasticity-based modulation of synaptic plasticity contributes to durable neurotransmission changes in mesocorticolimbic pathways including the ventral tegmental area and the nucleus accumbens, causing 'maladaptive' drug memory and higher risk for drug-seeking relapse. On the other hand, drug-induced metaplasticity can make appetitive memories more malleable to modification, offering a potential target mechanism for intervention. Here we review the literature on the role of glutamate receptors in addiction-related metaplasticity phenomena.
Collapse
Affiliation(s)
- Cristiano Chiamulera
- Neuropsychopharmacology Lab, Section Pharmacology, Department Diagnostic & Public Health, University of Verona, Verona, Italy.
| | - Alessandro Piva
- Neuropsychopharmacology Lab, Section Pharmacology, Department Diagnostic & Public Health, University of Verona, Verona, Italy
| | - Wickliffe C Abraham
- Department of Psychology, Brain Health Research Centre, Brain Research New Zealand, University of Otago, Dunedin, New Zealand
| |
Collapse
|
29
|
Chen WB, Pan HQ, He Y, Wang XH, Zhang WH, Pan BX. Rap1b but not Rap1a in the forebrain is required for learned fear. Cell Biosci 2020; 10:107. [PMID: 32944221 PMCID: PMC7488763 DOI: 10.1186/s13578-020-00469-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 09/04/2020] [Indexed: 11/10/2022] Open
Abstract
Background Fear is an adaptive response across species in the face of threatening cues. It can be either innate or learned through postnatal experience. We have previously shown that genetic deletion of both Rap1a and Rap1b, two isoforms of small GTPase Rap1 in forebrain, causes impairment in auditory fear conditioning. However, the specific roles of these two isoforms are not yet known. Results In the present study, employing mice with forebrain-restricted deletion of Rap1a or Rap1b, we found that they are both dispensable for normal acquisition of fear learning. However, Rap1b but not Rap1a knockout (KO) mice displayed impairment in the retrieval of learned fear. Subsequently, we found that the expression of c-Fos, a marker of neuronal activity, is specifically decreased in prelimbic cortex (PL) of Rap1b KO mice after auditory fear conditioning, while remained unaltered in the amygdala and infralimbic cortex (IL). On the other hand, neither Rap1a nor Rap1b knockout altered the innate fear of mice in response to their predator odor, 2,5-Dihydro-2,4,5-Trimethylthiazoline (TMT). Conclusion Thus, our results indicate that it is Rap1b but not Rap1a involved in the retrieval process of fear learning, and the learned but not innate fear requires Rap1 signaling in forebrain.
Collapse
Affiliation(s)
- Wen-Bing Chen
- School of Life Sciences, Nanchang University, Nanchang, 330031 China.,Institute of Life Science, Nanchang University, Nanchang, 330031 China
| | - Han-Qing Pan
- School of Life Sciences, Nanchang University, Nanchang, 330031 China.,Institute of Life Science, Nanchang University, Nanchang, 330031 China
| | - Ye He
- Institute of Life Science, Nanchang University, Nanchang, 330031 China.,Center for Basic Medical Experiment, Nanchang University, Nanchang, 330031 China
| | - Xue-Hui Wang
- School of Life Sciences, Nanchang University, Nanchang, 330031 China.,Institute of Life Science, Nanchang University, Nanchang, 330031 China
| | - Wen-Hua Zhang
- Institute of Life Science, Nanchang University, Nanchang, 330031 China
| | - Bing-Xing Pan
- School of Life Sciences, Nanchang University, Nanchang, 330031 China.,Institute of Life Science, Nanchang University, Nanchang, 330031 China.,Department of Ophthalmology, The 2nd Affiliated Hospital of Nanchang University, Nanchang, 330006 China
| |
Collapse
|
30
|
Huggett SB, Stallings MC. Genetic Architecture and Molecular Neuropathology of Human Cocaine Addiction. J Neurosci 2020; 40:5300-5313. [PMID: 32457073 PMCID: PMC7329314 DOI: 10.1523/jneurosci.2879-19.2020] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 05/04/2020] [Accepted: 05/10/2020] [Indexed: 01/12/2023] Open
Abstract
We integrated genomic and bioinformatic analyses, using data from the largest genome-wide association study of cocaine dependence (CD; n = 6546; 82.37% with CD; 57.39% male) and the largest postmortem gene-expression sample of individuals with cocaine use disorder (CUD; n = 36; 51.35% with CUD; 100% male). Our genome-wide analyses identified one novel gene (NDUFB9) associated with the genetic predisposition to CD in African-Americans. The genetic architecture of CD was similar across ancestries. Individual genes associated with CD demonstrated modest overlap across European-Americans and African-Americans, but the genetic liability for CD converged on many similar tissue types (brain, heart, blood, liver) across ancestries. In a separate sample, we investigated the neuronal gene expression associated with CUD by using RNA sequencing of dorsal-lateral prefrontal cortex neurons. We identified 133 genes differentially expressed between CUD case patients and cocaine-free control subjects, including previously implicated candidates for cocaine use/addiction (FOSB, ARC, KCNJ9/GIRK3, NR4A2, JUNB, and MECP2). Differential expression analyses significantly correlated across European-Americans and African-Americans. While genes significantly associated with CD via genome-wide methods were not differentially expressed, two of these genes (NDUFB9 and C1qL2) were part of a robust gene coexpression network associated with CUD involved in neurotransmission (GABA, acetylcholine, serotonin, and dopamine) and drug addiction. We then used a "guilt-by-association" approach to unravel the biological relevance of NDUFB9 and C1qL2 in the context of CD. In sum, our study furthers the understanding of the genetic architecture and molecular neuropathology of human cocaine addiction and provides a framework for translating biological meaning into otherwise obscure genome-wide associations.SIGNIFICANCE STATEMENT Our study further clarifies the genetic and neurobiological contributions to cocaine addiction, provides a rapid approach for generating testable hypotheses for specific candidates identified by genome-wide research, and investigates the cross-ancestral biological contributions to cocaine use disorder/dependence for individuals of European-American and African-American ancestries.
Collapse
Affiliation(s)
- Spencer B Huggett
- Department of Psychology and Neuroscience, University of Colorado, Boulder, Colorado 80309-0345
- Institute for Behavioral Genetics, University of Colorado, Boulder, Colorado 80309-0447
| | - Michael C Stallings
- Department of Psychology and Neuroscience, University of Colorado, Boulder, Colorado 80309-0345
- Institute for Behavioral Genetics, University of Colorado, Boulder, Colorado 80309-0447
| |
Collapse
|
31
|
Abstract
Substance and alcohol use disorders impose large health and economic burdens on individuals, families, communities, and society. Neither prevention nor treatment efforts are effective in all individuals. Results are often modest. Advances in neuroscience and addiction research have helped to describe the neurobiological changes that occur when a person transitions from recreational substance use to a substance use disorder or addiction. Understanding both the drivers and consequences of substance use in vulnerable populations, including those whose brains are still maturing, has revealed behavioral and biological characteristics that can increase risks of addiction. These findings are particularly timely, as law- and policymakers are tasked to reverse the ongoing opioid epidemic, as more states legalize marijuana, as new products including electronic cigarettes and newly designed abused substances enter the legal and illegal markets, and as "deaths of despair" from alcohol and drug misuse continue.
Collapse
Affiliation(s)
- George R. Uhl
- New Mexico VA Healthcare SystemAlbuquerqueNew Mexico
| | | | | |
Collapse
|
32
|
Kiernan EA, Wang T, Vanderplow AM, Cherukuri S, Cahill ME, Watters JJ. Neonatal Intermittent Hypoxia Induces Lasting Sex-Specific Augmentation of Rat Microglial Cytokine Expression. Front Immunol 2019; 10:1479. [PMID: 31333645 PMCID: PMC6615134 DOI: 10.3389/fimmu.2019.01479] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 06/13/2019] [Indexed: 01/08/2023] Open
Abstract
Sleep disordered breathing (SDB) affects 3-5% of the pediatric population, including neonates who are highly susceptible due to an underdeveloped ventilatory control system, and REM-dominated sleep. Although pediatric SDB is associated with poor cognitive outcomes, very little research has focused on models of pediatric SDB, particularly in neonates. In adults and neonates, intermittent hypoxia (IH), a hallmark of SDB, recapitulates multiple physiological aspects of severe SDB, including neuronal apoptosis, sex-specific cognitive deficits, and neuroinflammation. Microglia, resident CNS immune cells, are important mediators of neurodevelopment and neuroinflammation, but to date, no studies have examined the molecular properties of microglia in the context of neonatal IH. Here, we tested the hypothesis that neonatal IH will enhance microglial inflammation and sex-specifically lead to long-term changes in working memory. To test this hypothesis, we exposed post-natal day (P1) neonates with dams to an established adult model of pathological IH consisting of 2 min cycles of 10.5% O2 followed by 21% O2, 8 h/day for 8 days. We then challenged the offspring with bacterial lipopolysaccharide (LPS) at P9 or at 6-8 weeks of age and immunomagnetically isolated microglia for gene expression analyses and RNA-sequencing. We also characterized neonatal CNS myeloid cell populations by flow cytometry analyses. Lastly, we examined working memory performance using a Y-maze in the young adults. Contrary to our hypothesis, we found that neonatal IH acutely augmented basal levels of microglial anti-inflammatory cytokines, attenuated microglial responses to LPS, and sex-specifically altered CNS myeloid populations. We identified multiple sex differences in basal neonatal microglial expression of genes related to chemotaxis, cognition, and aging. Lastly, we found that basal, but not LPS-induced, anti-inflammatory cytokines were augmented sex-specifically in the young adults, and that there was a significant interaction between sex and IH on basal working memory. Our results support the idea that neonates may be able to adapt to IH exposures that are pathological in adults. Further, they suggest that male and female microglial responses to IH are sex-specific, and that these sex differences in basal microglial gene expression may contribute to sexual dimorphisms in vulnerability to IH-induced cognitive disruption.
Collapse
Affiliation(s)
- Elizabeth A Kiernan
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, United States
| | - Tao Wang
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, United States
| | - Amanda M Vanderplow
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, United States
| | - Sneha Cherukuri
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, United States
| | - Michael E Cahill
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, United States.,Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, United States
| | - Jyoti J Watters
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, United States.,Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
33
|
Synaptic Microtubule-Associated Protein EB3 and SRC Phosphorylation Mediate Structural and Behavioral Adaptations During Withdrawal From Cocaine Self-Administration. J Neurosci 2019; 39:5634-5646. [PMID: 31092585 DOI: 10.1523/jneurosci.0024-19.2019] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 04/16/2019] [Accepted: 05/08/2019] [Indexed: 12/21/2022] Open
Abstract
Addictive behaviors, including relapse, are thought to depend in part on long-lasting drug-induced adaptations in dendritic spine signaling and morphology in the nucleus accumbens (NAc). While the influence of activity-dependent actin remodeling in these phenomena has been studied extensively, the role of microtubules and associated proteins remains poorly understood. We report that pharmacological inhibition of microtubule polymerization in the NAc inhibited locomotor sensitization to cocaine and contextual reward learning. We then investigated the roles of microtubule end-binding protein 3 (EB3) and SRC kinase in the neuronal and behavioral responses to volitionally administered cocaine. In synaptoneurosomal fractions from the NAc of self-administering male rats, the phosphorylation of SRC at an activating site was induced after 1 d of withdrawal, while EB3 levels were increased only after 30 d of withdrawal. Blocking SRC phosphorylation during early withdrawal by virally overexpressing SRCIN1, a negative regulator of SRC activity known to interact with EB3, abolished the incubation of cocaine craving in both male and female rats. Conversely, mimicking the EB3 increase observed after prolonged withdrawal increased the motivation to consume cocaine in male rats. In mice, the overexpression of either EB3 or SRCIN1 increased dendritic spine density and altered the spine morphology of NAc medium spiny neurons. Finally, a cocaine challenge after prolonged withdrawal recapitulated most of the synaptic protein expression profiles observed at early withdrawal. These findings suggest that microtubule-associated signaling proteins such as EB3 cooperate with actin remodeling pathways, notably SRC kinase activity, to establish and maintain long-lasting cellular and behavioral alterations following cocaine self-administration.SIGNIFICANCE STATEMENT Drug-induced morphological restructuring of dendritic spines of nucleus accumbens neurons is thought to be one of the cellular substrates of long-lasting drug-associated memories. The molecular basis of these persistent changes has remained incompletely understood. Here we implicate for the first time microtubule function in this process, together with key players such as microtubule-bound protein EB3 and synaptic SRC phosphorylation. We propose that microtubule and actin remodeling cooperate during withdrawal to maintain the plastic structural changes initially established by cocaine self-administration. This work opens new translational avenues for further characterization of microtubule-associated regulatory molecules as putative drug targets to tackle relapse to drug taking.
Collapse
|
34
|
Xu SJ, Heller EA. Recent advances in neuroepigenetic editing. Curr Opin Neurobiol 2019; 59:26-33. [PMID: 31015104 DOI: 10.1016/j.conb.2019.03.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 02/28/2019] [Accepted: 03/18/2019] [Indexed: 02/09/2023]
Abstract
A wealth of studies in the mammalian nervous system indicate the role of epigenetic gene regulation in both basic neurobiological function and disease. However, the relationship between epigenetic regulation and neuropathology is largely correlational due to the presence of mixed cell populations within brain regions and the genome-wide effects of classical approaches to manipulate the epigenome. Locus-specific epigenetic editing allows direct epigenetic regulation of specific genes to elucidate the direct causal relationship between epigenetic modifications and transcription. This review discusses some of the latest innovations in the efficacy and flexibility in this approach that hold promise for neurobiological application.
Collapse
Affiliation(s)
- Song-Jun Xu
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA, USA
| | - Elizabeth A Heller
- Department of Systems Pharmacology and Translational Therapeutics and Penn Epigenetics Institute, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
35
|
Yaw AM, Prosser RA, Jones PC, Garcia BJ, Jacobson DA, Glass JD. Epigenetic effects of paternal cocaine on reward stimulus behavior and accumbens gene expression in mice. Behav Brain Res 2019; 367:68-81. [PMID: 30910707 DOI: 10.1016/j.bbr.2019.02.043] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 02/08/2019] [Accepted: 02/25/2019] [Indexed: 12/23/2022]
Abstract
Paternal cocaine use causes phenotypic alterations in offspring behavior and associated neural processing. In rodents, changes in first generation (F1) offspring include drug reward behavior, circadian timing, and anxiety responses. This study, utilizing a murine (C57BL/6J) oral cocaine model, examines the effects of paternal cocaine exposure on fundamental characteristics of offspring reward responses, including: 1) the extent of cocaine-induced effects after different durations of sire drug withdrawal; 2) sex- and drug-dependent differences in F1 reward preference; 3) effects on second generation (F2) cocaine preference; and 4) corresponding changes in reward area (nucleus accumbens) mRNA expression. We demonstrate that paternal cocaine intake over a single ˜40-day spermatogenic cycle significantly decreased cocaine (but not ethanol or sucrose) preference in a sex-specific manner in F1 mice from sires mated 24 h after drug withdrawal. However, F1 offspring of sires bred 4 months after withdrawal did not exhibit altered cocaine preference. Altered cocaine preference also was not observed in F2's. RNASeq analyses of F1 accumbens tissue revealed changes in gene expression in male offspring of cocaine-exposed sires, including many genes not previously linked to cocaine addiction. Enrichment analyses highlight genes linked to CNS development, synaptic signaling, extracellular matrix, and immune function. Expression correlation analyses identified a novel target, Fam19a4, that may negatively regulate many genes in the accumbens, including genes already identified in addiction. Collectively, these results reveal that paternal cocaine effects in F1 offspring may involve temporally limited epigenetic germline effects and identify new genetic targets for addiction research.
Collapse
Affiliation(s)
- Alexandra M Yaw
- School of Biomedical Sciences, Kent State Univ., Kent, OH, 44242, United States
| | - Rebecca A Prosser
- Dept. of Biochemistry & Cellular and Molecular Biology, University of Tennessee, Knoxville, TN, 37996, United States; NeuroNET Research Center, University of Tennessee, Knoxville, TN, 37996, United States
| | - Piet C Jones
- Biosciences Division, Oak Ridge National Laboratory, Oak Ridge, TN, 37830, United States; Bredesen Center for Interdisciplinary Research, University of Tennessee, Knoxville, TN, 37996, United States
| | - Benjamin J Garcia
- Biosciences Division, Oak Ridge National Laboratory, Oak Ridge, TN, 37830, United States
| | - Daniel A Jacobson
- NeuroNET Research Center, University of Tennessee, Knoxville, TN, 37996, United States; Biosciences Division, Oak Ridge National Laboratory, Oak Ridge, TN, 37830, United States; Bredesen Center for Interdisciplinary Research, University of Tennessee, Knoxville, TN, 37996, United States; Department of Psychology, University of Tennessee, Knoxville, TN, 37996, United States
| | - J David Glass
- School of Biomedical Sciences, Kent State Univ., Kent, OH, 44242, United States.
| |
Collapse
|
36
|
Pei F, Li H, Liu B, Bahar I. Quantitative Systems Pharmacological Analysis of Drugs of Abuse Reveals the Pleiotropy of Their Targets and the Effector Role of mTORC1. Front Pharmacol 2019; 10:191. [PMID: 30906261 PMCID: PMC6418047 DOI: 10.3389/fphar.2019.00191] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 02/14/2019] [Indexed: 12/14/2022] Open
Abstract
Existing treatments against drug addiction are often ineffective due to the complexity of the networks of protein-drug and protein-protein interactions (PPIs) that mediate the development of drug addiction and related neurobiological disorders. There is an urgent need for understanding the molecular mechanisms that underlie drug addiction toward designing novel preventive or therapeutic strategies. The rapidly accumulating data on addictive drugs and their targets as well as advances in machine learning methods and computing technology now present an opportunity to systematically mine existing data and draw inferences on potential new strategies. To this aim, we carried out a comprehensive analysis of cellular pathways implicated in a diverse set of 50 drugs of abuse using quantitative systems pharmacology methods. The analysis of the drug/ligand-target interactions compiled in DrugBank and STITCH databases revealed 142 known and 48 newly predicted targets, which have been further analyzed to identify the KEGG pathways enriched at different stages of drug addiction cycle, as well as those implicated in cell signaling and regulation events associated with drug abuse. Apart from synaptic neurotransmission pathways detected as upstream signaling modules that “sense” the early effects of drugs of abuse, pathways involved in neuroplasticity are distinguished as determinants of neuronal morphological changes. Notably, many signaling pathways converge on important targets such as mTORC1. The latter emerges as a universal effector of the persistent restructuring of neurons in response to continued use of drugs of abuse.
Collapse
Affiliation(s)
- Fen Pei
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Hongchun Li
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Bing Liu
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Ivet Bahar
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
37
|
Zhang JY, Liu TH, He Y, Pan HQ, Zhang WH, Yin XP, Tian XL, Li BM, Wang XD, Holmes A, Yuan TF, Pan BX. Chronic Stress Remodels Synapses in an Amygdala Circuit-Specific Manner. Biol Psychiatry 2019; 85:189-201. [PMID: 30060908 PMCID: PMC6747699 DOI: 10.1016/j.biopsych.2018.06.019] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 06/20/2018] [Accepted: 06/22/2018] [Indexed: 12/20/2022]
Abstract
BACKGROUND Chronic stress exposure increases the risk of developing various neuropsychiatric illnesses. The behavioral sequelae of stress correlate with dendritic hypertrophy and glutamate-related synaptic remodeling at basolateral amygdala projection neurons (BLA PNs). Yet, though BLA PNs are functionally heterogeneous with diverse corticolimbic targets, it remains unclear whether stress differentially impacts specific output circuits. METHODS Confocal imaging was used to reconstruct the morphology of mouse BLA PNs with the aid of retrograde tracing and biocytin staining. The synaptic activity in these neurons was measured with in vitro electrophysiology, and anxiety-like behavior of the mice was assessed with the elevated plus maze and open field test. RESULTS Chronic restraint stress (CRS) produced dendritic hypertrophy across mouse BLA PNs, regardless of whether they did (BLA→dorsomedial prefrontal cortex [dmPFC]) or did not (BLA↛dmPFC) target dmPFC. However, CRS increased the size of dendritic spine heads and the number of mature, mushroom-shaped spines only in BLA↛dmPFC PNs, sparing neighboring BLA→dmPFC PNs. Moreover, the excitatory glutamatergic transmission was also selectively increased in BLA↛dmPFC PNs, and this effect correlated with CRS-induced increases in anxiety-like behavior. Segregating BLA↛dmPFC PNs based on their targeting of ventral hippocampus (BLA→ventral hippocampus) or nucleus accumbens (BLA→nucleus accumbens) revealed that CRS increased spine density and glutamatergic signaling in BLA→ventral hippocampus PNs in a manner that correlated with anxiety-like behavior. CONCLUSIONS Chronic stress caused BLA PN neuronal remodeling with a previously unrecognized degree of circuit specificity, offering new insight into the pathophysiological basis of depression, anxiety disorders, and other stress-related conditions.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Bing-Xing Pan
- Laboratory of Fear and Anxiety Disorders, Institute of Life Science, Nanchang University, Nanchang, China; Department of Neurology, the 2nd Affiliated Hospital, Nanchang University, Nanchang, China; Human Aging Research Institute, School of Life Science, Nanchang University, Nanchang, China.
| |
Collapse
|
38
|
Walker DM, Cates HM, Loh YHE, Purushothaman I, Ramakrishnan A, Cahill KM, Lardner CK, Godino A, Kronman HG, Rabkin J, Lorsch ZS, Mews P, Doyle MA, Feng J, Labonté B, Koo JW, Bagot RC, Logan RW, Seney ML, Calipari ES, Shen L, Nestler EJ. Cocaine Self-administration Alters Transcriptome-wide Responses in the Brain's Reward Circuitry. Biol Psychiatry 2018; 84:867-880. [PMID: 29861096 PMCID: PMC6202276 DOI: 10.1016/j.biopsych.2018.04.009] [Citation(s) in RCA: 137] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 03/15/2018] [Accepted: 04/17/2018] [Indexed: 11/29/2022]
Abstract
BACKGROUND Global changes in gene expression underlying circuit and behavioral dysregulation associated with cocaine addiction remain incompletely understood. Here, we show how a history of cocaine self-administration (SA) reprograms transcriptome-wide responses throughout the brain's reward circuitry at baseline and in response to context and/or cocaine re-exposure after prolonged withdrawal (WD). METHODS We assigned male mice to one of six groups: saline/cocaine SA + 24-hour WD or saline/cocaine SA + 30-day WD + an acute saline/cocaine challenge within the previous drug-paired context. RNA sequencing was conducted on six interconnected brain reward regions. Using pattern analysis of gene expression and factor analysis of behavior, we identified genes that are strongly associated with addiction-related behaviors and uniquely altered by a history of cocaine SA. We then identified potential upstream regulators of these genes. RESULTS We focused on three patterns of gene expression that reflect responses to 1) acute cocaine, 2) context re-exposure, and 3) drug + context re-exposure. These patterns revealed region-specific regulation of gene expression. Further analysis revealed that each of these gene expression patterns correlated with an addiction index-a composite score of several addiction-like behaviors during cocaine SA-in a region-specific manner. Cyclic adenosine monophosphate response element binding protein and nuclear receptor families were identified as key upstream regulators of genes associated with such behaviors. CONCLUSIONS This comprehensive picture of transcriptome-wide regulation in the brain's reward circuitry by cocaine SA and prolonged WD provides new insight into the molecular basis of cocaine addiction, which will guide future studies of the key molecular pathways involved.
Collapse
Affiliation(s)
- Deena M Walker
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Hannah M Cates
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Yong-Hwee E Loh
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Immanuel Purushothaman
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Aarthi Ramakrishnan
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Kelly M Cahill
- Department of Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Casey K Lardner
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Arthur Godino
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Hope G Kronman
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Jacqui Rabkin
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Zachary S Lorsch
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Philipp Mews
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Marie A Doyle
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Jian Feng
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Benoit Labonté
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Ja Wook Koo
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Rosemary C Bagot
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Ryan W Logan
- Department of Psychiatry, University of Pittsburgh Medical School, Pittsburgh, Pennsylvania; Translational Neuroscience Program, University of Pittsburgh Medical School, Pittsburgh, Pennsylvania; Center for Systems Neurogenetics of Addiction, The Jackson Laboratory, Bar Harbor, Maine
| | - Marianne L Seney
- Department of Psychiatry, University of Pittsburgh Medical School, Pittsburgh, Pennsylvania; Translational Neuroscience Program, University of Pittsburgh Medical School, Pittsburgh, Pennsylvania
| | - Erin S Calipari
- Department of Pharmacology, Vanderbilt Center for Addiction Research, Vanderbilt University School of Medicine, Nashville, Tennessee.
| | - Li Shen
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Eric J Nestler
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York.
| |
Collapse
|
39
|
Seelke AMH, Bond JM, Simmons TC, Joshi N, Settles ML, Stolzenberg D, Rhemtulla M, Bales KL. Fatherhood alters gene expression within the MPOA. ENVIRONMENTAL EPIGENETICS 2018; 4:dvy026. [PMID: 30568805 PMCID: PMC6305489 DOI: 10.1093/eep/dvy026] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 10/03/2018] [Accepted: 10/19/2018] [Indexed: 06/09/2023]
Abstract
Female parenting is obligate in mammals, but fathering behavior among mammals is rare. Only 3-5% of mammalian species exhibit biparental care, including humans, and mechanisms of fathering behavior remain sparsely studied. However, in species where it does exist, paternal care is often crucial to the survivorship of offspring. The present study is the first to identify new gene targets linked to the experience of fathering behavior in a biparental species using RNA sequencing. In order to determine the pattern of gene expression within the medial preoptic area that is specifically associated with fathering behavior, we identified genes in male prairie voles (Microtus ochrogaster) that experienced one of three social conditions: virgin males, pair bonded males, and males with fathering experience. A list of genes exhibiting different expression patterns in each comparison (i.e. Virgin vs Paired, Virgin vs Fathers, and Paired vs Fathers) was evaluated using the gene ontology enrichment analysis, and Kyoto Encyclopedia of Genes and Genomes pathways analysis to reveal metabolic pathways associated with specific genes. Using these tools, we generated a filtered list of genes that exhibited altered patterns of expression in voles with different amounts of social experience. Finally, we used NanoString to quantify differences in the expression of these selected genes. These genes are involved in a variety of processes, with enrichment in genes associated with immune function, metabolism, synaptic plasticity, and the remodeling of dendritic spines. The identification of these genes and processes will lead to novel insights into the biological basis of fathering behavior.
Collapse
Affiliation(s)
- Adele M H Seelke
- Department of Psychology, University of California, Davis, Davis, USA
| | - Jessica M Bond
- Department of Psychology, University of California, Davis, Davis, USA
| | - Trent C Simmons
- Department of Psychology, University of California, Davis, Davis, USA
| | - Nikhil Joshi
- Bioinformatics Core Facility, University of California, Davis, Davis, USA
| | - Matthew L Settles
- Bioinformatics Core Facility, University of California, Davis, Davis, USA
| | | | - Mijke Rhemtulla
- Department of Psychology, University of California, Davis, Davis, USA
| | - Karen L Bales
- Department of Psychology, University of California, Davis, Davis, USA
- California National Primate Research Center, University of California, Davis, Davis, USA
| |
Collapse
|
40
|
Xu SJ, Heller EA. Single sample sequencing (S3EQ) of epigenome and transcriptome in nucleus accumbens. J Neurosci Methods 2018; 308:62-73. [PMID: 30031009 PMCID: PMC6296235 DOI: 10.1016/j.jneumeth.2018.07.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 06/19/2018] [Accepted: 07/06/2018] [Indexed: 12/30/2022]
Abstract
BACKGROUND High-throughput sequencing has been widely applied to uncover the molecular mechanisms underlying neurological and psychiatric disorders. The large body of data support the role of epigenetic mechanisms in neurological function of both human and animals. Yet, the existing data is limited by the fact that epigenetic and transcriptomic changes have only been measured in separate cohorts. This has limited precise correlation of epigenetic changes in gene expression. NEW METHOD Single Sample Sequencing (S3EQ) is an innovative approach to analyze both epigenetic and transcriptomic regulation within a single neuronal sample. Using this method, we analyzed chromatin immunoprecipitation (ChIP)- and RNA-sequencing data from the nucleus accumbens (NAc) of the same animal. RESULTS ChIP-S3EQ of neuronal nuclei reliably identified hPTM enrichment in the adult mouse NAc with high precision. Comparing cellular compartments, we found that the spliceosome of whole cell RNA-seq was more closely recapitulated by cytosolic RNA-S3EQ than nuclear RNA-seq. Finally, S3EQ showed increased sensitivity for correlating chromatin modifications with gene expression, especially for lowly expressed transcripts. COMPARISON WITH EXISTING METHODS S3EQ accurately generates both RNA- and ChIP-seq from a single sample, providing a clear advantage over existing methods which require two samples. ChIP-S3EQ performance was comparable to ChIP-seq, while RNA-S3EQ generated an almost identical expression profile to nuclear-enriched and whole cell RNA-seq. Finally, we directly compared RNA-seq by cellular compartments, addressing a limitation of RNA-seq studies limited to neuronal nuclei. CONCLUSION The S3EQ method can be applied to improve the correlative power of transcriptomic and epigenomic studies in neuronal tissue.
Collapse
Affiliation(s)
- S J Xu
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA, USA
| | - E A Heller
- Department of Systems Pharmacology and Translational Therapeutics and Penn Epigenetics Institute, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
41
|
Cahill ME, Browne CJ, Wang J, Hamilton PJ, Dong Y, Nestler EJ. Withdrawal from repeated morphine administration augments expression of the RhoA network in the nucleus accumbens to control synaptic structure. J Neurochem 2018; 147:84-98. [PMID: 30071134 DOI: 10.1111/jnc.14563] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 07/26/2018] [Accepted: 07/27/2018] [Indexed: 12/18/2022]
Abstract
The nucleus accumbens (NAc) is a critical brain reward region that mediates the rewarding effects of drugs of abuse, including those of morphine and other opiates. Drugs of abuse induce widespread alterations in gene transcription and dendritic spine morphology in medium spiny neurons (MSNs) of the NAc that ultimately influence NAc excitability and hence reward-related behavioral responses. Growing evidence indicates that within the NAc small GTPases are common intracellular targets of drugs of abuse where these molecules regulate drug-mediated transcriptional and spine morphogenic effects. The RhoA small GTPase is among the most well-characterized members of the Ras superfamily of small GTPases, and recent work highlights an important role for hippocampal RhoA in morphine-facilitated reward behavior. Despite this, it remains unclear how RhoA pathway signaling in the NAc is affected by withdrawal from morphine. To investigate this question, using subcellular fractionation and subsequent protein profiling we examined the expression of key components of the RhoA pathway in NAc nuclear, cytoplasmic, and synaptosomal compartments during multiple withdrawal periods from repeated morphine administration. Furthermore, using in vivo viral-mediated gene transfer, we determined the consequences of revealed RhoA pathway alterations on NAc MSN dendritic spine morphology. Our findings reveal an important role for RhoA signaling cascades in mediating the effects of long-term morphine withdrawal on NAc MSN dendritic spine elimination. OPEN PRACTICES Open Science: This manuscript was awarded with the Open Materials Badge. For more information see: https://cos.io/our-services/open-science-badges/.
Collapse
Affiliation(s)
- Michael E Cahill
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Caleb J Browne
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Junshi Wang
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Peter J Hamilton
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Yan Dong
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Eric J Nestler
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
42
|
Mervosh NL, Wilson R, Rauniyar N, Hofford RS, Kutlu MG, Calipari ES, Lam TT, Kiraly DD. Granulocyte-Colony-Stimulating Factor Alters the Proteomic Landscape of the Ventral Tegmental Area. Proteomes 2018; 6:proteomes6040035. [PMID: 30249060 PMCID: PMC6313867 DOI: 10.3390/proteomes6040035] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 09/17/2018] [Accepted: 09/20/2018] [Indexed: 12/12/2022] Open
Abstract
Cocaine addiction is characterized by aberrant plasticity of the mesolimbic dopamine circuit, leading to dysregulation of motivation to seek and take drug. Despite the significant toll that cocaine use disorder exacts on society, there are currently no available pharmacotherapies. We have recently identified granulocyte-colony stimulating factor (G-CSF) as a soluble cytokine that alters the behavioral response to cocaine and which increases dopamine release from the ventral tegmental area (VTA). Despite these known effects on behavior and neurophysiology, the molecular mechanisms by which G-CSF affects brain function are unclear. In this study mice were treated with repeated injections of G-CSF, cocaine or a combination and changes in protein expression in the VTA were examined using an unbiased proteomics approach. Repeated G-CSF treatment resulted in alterations in multiple signaling pathways related to synaptic plasticity and neuronal morphology. While the treatment groups had marked overlap in their effect, injections of cocaine and the combination of cocaine and G-CSF lead to distinct patterns of significantly regulated proteins. These experiments provide valuable information as to the molecular pathways that G-CSF activates in an important limbic brain region and will help to guide further characterization of G-CSF function and evaluation as a possible translational target.
Collapse
Affiliation(s)
- Nicholas L Mervosh
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
- Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Rashaun Wilson
- Yale/NIDA Neuroproteomics Center, New Haven, CT 06511, USA.
| | - Navin Rauniyar
- Yale/NIDA Neuroproteomics Center, New Haven, CT 06511, USA.
| | - Rebecca S Hofford
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
- Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Munir Gunes Kutlu
- Department of Pharmacology, Vanderbilt Center for Addiction Research, Vanderbilt University School of Medicine, Nashville, TN 37232, USA.
| | - Erin S Calipari
- Department of Pharmacology, Vanderbilt Center for Addiction Research, Vanderbilt University School of Medicine, Nashville, TN 37232, USA.
| | - TuKiet T Lam
- Yale/NIDA Neuroproteomics Center, New Haven, CT 06511, USA.
- Department of Molecular Biophysics & Biochemistry, New Haven, CT 06510, USA.
- Yale MS & Proteomics Resource, New Haven, CT 06510, USA.
| | - Drew D Kiraly
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
- Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
43
|
Wu JH, Li Y, Zhou YF, Haslam J, Elvis ON, Mao L, Xia YP, Hu B. Semaphorin-3E attenuates neointimal formation via suppressing VSMCs migration and proliferation. Cardiovasc Res 2018; 113:1763-1775. [PMID: 29016743 DOI: 10.1093/cvr/cvx190] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 09/14/2017] [Indexed: 02/06/2023] Open
Abstract
Aims The migration and proliferation of vascular smooth muscle cells (VSMCs) are crucial events in the neointimal formation, a hallmark of atherosclerosis and restenosis. Semaphorin3E (Sema3E) has been found to be a critical regulator of cell migration and proliferation in many scenarios. However, its role on VSMCs migration and proliferation is unclear. This study aimed to investigate the effect of Sema3E on VSMCs migration, proliferation and neointimal formation, and explore possible mechanisms. Methods and results We found that the expression of Sema3E was progressively decreased during neointimal formation in a carotid ligation model. H&E-staining showed lentivirus-mediated overexpression of Sema3E in carotid ligation area attenuated neointimal formation. Immunofluorescence staining showed that the receptor (PlexinD1) of Sema3E was expressed in vascular walls. In cultured mouse VSMCs, Sema3E inhibited VSMCs migration and proliferation via plexinD1 receptor. The inhibitory effect was mediated, at least in part, by inactivating Rap1-AKT signalling pathways in VSMCs. Moreover, we found that PDGFBB down-regulated the expression of Sema3E in VSMCs and Sema3E notably inhibited the expression of PDGFB in endothelial cells. In addition, the number of Sema3E-positive VSMCs was diminished in plaques of atherosclerotic patients. Results from a public GEO microarray database showed a negative correlation between Sema3E and PDGFB transcriptional levels in the human plaques examined. Conclusion Our study demonstrates that Sema3E/plexinD1 inhibits proliferation and migration of VSMCs via inactivation of Rap1-AKT signalling pathways. The mutual inhibition between PDGF-BB and Sema3E after vascular injury plays a critical role in the process of neointimal formation.
Collapse
Affiliation(s)
- Jie-Hong Wu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yanan Li
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yi-Fan Zhou
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - James Haslam
- Swansea College of Medicine, Swansea University, Singleton Park, Swansea, Wales SA2 8PP, UK
| | - Opoku Nana Elvis
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ling Mao
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yuan-Peng Xia
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Bo Hu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
44
|
Dopamine D1 receptor subtype mediates acute stress-induced dendritic growth in excitatory neurons of the medial prefrontal cortex and contributes to suppression of stress susceptibility in mice. Mol Psychiatry 2018; 23:1717-1730. [PMID: 28924188 DOI: 10.1038/mp.2017.177] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 06/18/2017] [Accepted: 07/28/2017] [Indexed: 12/12/2022]
Abstract
Dopamine in prefrontal cortices is implicated in cognitive and emotional functions, and the dysfunction of prefrontal dopamine has been associated with cognitive and emotional deficits in mental illnesses. These findings have led to clinical trials of dopamine-targeting drugs and brain imaging of dopamine receptors in patients with mental illnesses. Rodent studies have suggested that dopaminergic pathway projecting to the medial prefrontal cortex (mPFC) suppresses stress susceptibility. Although various types of mPFC neurons express several dopamine receptor subtypes, previous studies neither isolated a role of dopamine receptor subtype nor identified the site of its action in mPFC. Using social defeat stress (SDS) in mice, here we identified a role of dopamine D1 receptor subtype in mPFC excitatory neurons in suppressing stress susceptibility. Repeated social defeat stress (R-SDS) reduces the expression of D1 receptor subtype in mPFC of mice susceptible to R-SDS. Knockdown of D1 receptor subtype in whole neuronal populations or excitatory neurons in mPFC facilitates the induction of social avoidance by SDS. Single social defeat stress (S-SDS) induces D1 receptor-mediated extracellular signal-regulated kinase phosphorylation and c-Fos expression in mPFC neurons. Whereas R-SDS reduces dendritic lengths of mPFC layer II/III pyramidal neurons, S-SDS increases arborization and spines of apical dendrites of these neurons in a D1 receptor-dependent manner. Collectively, our findings show that D1 receptor subtype and related signaling in mPFC excitatory neurons mediate acute stress-induced dendritic growth of these neurons and contribute to suppression of stress susceptibility. Therefore, we propose that D1 receptor-mediated dendritic growth in mPFC excitatory neurons suppresses stress susceptibility.
Collapse
|
45
|
Cates HM, Heller EA, Lardner CK, Purushothaman I, Peña CJ, Walker DM, Cahill M, Neve RL, Shen L, Bagot RC, Nestler EJ. Transcription Factor E2F3a in Nucleus Accumbens Affects Cocaine Action via Transcription and Alternative Splicing. Biol Psychiatry 2018; 84:167-179. [PMID: 29397901 PMCID: PMC5988910 DOI: 10.1016/j.biopsych.2017.11.027] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 11/09/2017] [Accepted: 11/16/2017] [Indexed: 11/25/2022]
Abstract
BACKGROUND Lasting changes in gene expression in brain reward regions, including nucleus accumbens (NAc), contribute to persistent functional changes in the addicted brain. We and others have demonstrated that altered expression of several candidate transcription factors in NAc regulates drug responses. A recent large-scale genome-wide study from our group predicted transcription factor E2F3 (E2F3) as a prominent upstream regulator of cocaine-induced changes in gene expression and alternative splicing. METHODS We studied expression of two E2F3 isoforms-E2F3a and E2F3b-in mouse NAc after repeated cocaine administration and assayed the effects of overexpression or depletion of E2f3 isoforms in NAc on cocaine behavioral responses. We then performed RNA sequencing to investigate the effect of E2f3a overexpression in this region on gene expression and alternative splicing and performed quantitative chromatin immunoprecipitation at downstream targets in NAc following E2f3a overexpression or repeated cocaine exposure. Sample sizes varied between experiments and are noted in the text. RESULTS We showed that E2f3a, but not E2f3b, overexpression or knockdown in mouse NAc regulates cocaine-induced locomotor and place conditioning behavior. Furthermore, we demonstrated that E2f3a overexpression substantially recapitulates genome-wide transcriptional profiles and alternative splicing induced by cocaine. We further validated direct binding of E2F3a at key target genes following cocaine exposure. CONCLUSIONS This study establishes E2F3a as a novel transcriptional regulator of cocaine action in NAc. The findings reveal a crucial role for E2F3a in the regulation of cocaine-elicited behavioral states. Moreover, the importance of this role is bolstered by the extensive recapitulation of cocaine's transcriptional effects in NAc by overexpression of E2f3a.
Collapse
Affiliation(s)
- Hannah M. Cates
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Elizabeth A. Heller
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Casey K. Lardner
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Immanuel Purushothaman
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Catherine J. Peña
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Deena M. Walker
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Michael Cahill
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Rachael L. Neve
- Department of Neurology, Massachusetts General Hospital, Boston, MA
| | - Li Shen
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Rosemary C. Bagot
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Eric J. Nestler
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, Corresponding Author, Lead Contact,
| |
Collapse
|
46
|
Mul JD, Soto M, Cahill ME, Ryan RE, Takahashi H, So K, Zheng J, Croote DE, Hirshman MF, la Fleur SE, Nestler EJ, Goodyear LJ. Voluntary wheel running promotes resilience to chronic social defeat stress in mice: a role for nucleus accumbens ΔFosB. Neuropsychopharmacology 2018; 43:1934-1942. [PMID: 29875450 PMCID: PMC6046059 DOI: 10.1038/s41386-018-0103-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 04/26/2018] [Accepted: 05/16/2018] [Indexed: 11/09/2022]
Abstract
Elucidating mechanisms by which physical exercise promotes resilience, the brain's ability to cope with prolonged stress exposure while maintaining normal psychological functioning, is a major research challenge given the high prevalence of stress-related mental disorders, including major depressive disorder. Chronic voluntary wheel running (VWR), a rodent model that mimics aspects of human physical exercise, induces the transcription factor ΔFosB in the nucleus accumbens (NAc), a key reward-related brain area. ΔFosB expression in NAc modulates stress susceptibility. Here, we explored whether VWR induction of NAc ΔFosB promotes resilience to chronic social defeat stress (CSDS). Male young-adult C57BL/6J mice were single housed for up to 21 d with or without running wheels and then subjected to 10 d of CSDS. Stress-exposed sedentary mice developed a depressive-like state, characterized by anhedonia and social avoidance, whereas stress-exposed mice that had been wheel running showed resilience. Functional inhibition of NAc ΔFosB during VWR, by viral-mediated overexpression of a transcriptionally inactive JunD mutant, reinstated susceptibility to CSDS. Within the NAc, VWR induction of ΔFosB was CREB-dependent, associated with altered dendritic morphology, and medium spiny neuron (MSN) subtype specific in the NAc core and shell subregions. Finally, when mice performed VWR following the onset of CSDS-induced social avoidance, VWR normalized such behavior. These data indicate that VWR promoted resilience to CSDS, and suggest that sustained induction of ΔFosB in the NAc underlies, at least in part, the stress resilience mediated by VWR. These findings provide a potential framework for the development of treatments for stress-associated mental illnesses based on physical exercise.
Collapse
Affiliation(s)
- Joram D Mul
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, 02215, USA.
- Department of Endocrinology and Metabolism, Academic Medical Center, University of Amsterdam, 1105 AZ, Amsterdam, The Netherlands.
- Metabolism and Reward Group, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, 1105 BA, Amsterdam, Amsterdam, The Netherlands.
| | - Marion Soto
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Michael E Cahill
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Rebecca E Ryan
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Hirokazu Takahashi
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Kawai So
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Jia Zheng
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Denise E Croote
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Michael F Hirshman
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Susanne E la Fleur
- Department of Endocrinology and Metabolism, Academic Medical Center, University of Amsterdam, Amsterdam Movement Sciences, 1105 AZ, Amsterdam, The Netherlands
- Metabolism and Reward Group, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, 1105 BA, Amsterdam, The Netherlands
| | - Eric J Nestler
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Laurie J Goodyear
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, 02215, USA
- Department of Medicine, Brigham, and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| |
Collapse
|
47
|
Cahill ME, Walker DM, Gancarz AM, Wang ZJ, Lardner CK, Bagot RC, Neve RL, Dietz DM, Nestler EJ. The dendritic spine morphogenic effects of repeated cocaine use occur through the regulation of serum response factor signaling. Mol Psychiatry 2018; 23:1474-1486. [PMID: 28555077 PMCID: PMC5709273 DOI: 10.1038/mp.2017.116] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 03/23/2017] [Accepted: 04/12/2017] [Indexed: 12/28/2022]
Abstract
The nucleus accumbens (NAc) is a primary brain reward region composed predominantly of medium spiny neurons (MSNs). In response to early withdrawal from repeated cocaine administration, de novo dendritic spine formation occurs in NAc MSNs. Much evidence indicates that this new spine formation facilitates the rewarding properties of cocaine. Early withdrawal from repeated cocaine also produces dramatic alterations in the transcriptome of NAc MSNs, but how such alterations influence cocaine's effects on dendritic spine formation remain unclear. Studies in non-neuronal cells indicate that actin cytoskeletal regulatory pathways in nuclei have a direct role in the regulation of gene transcription in part by controlling the access of co-activators to their transcription factor partners. In particular, actin state dictates the interaction between the serum response factor (SRF) transcription factor and one of its principal co-activators, MAL. Here we show that cocaine induces alterations in nuclear F-actin signaling pathways in the NAc with associated changes in the nuclear subcellular localization of SRF and MAL. Using in vivo optogenetics, the brain region-specific inputs to the NAc that mediate these nuclear changes are investigated. Finally, we demonstrate that regulated SRF expression, in turn, is critical for the effects of cocaine on dendritic spine formation and for cocaine-mediated behavioral sensitization. Collectively, these findings reveal a mechanism by which nuclear-based changes influence the structure of NAc MSNs in response to cocaine.
Collapse
Affiliation(s)
- ME Cahill
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - DM Walker
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - AM Gancarz
- Department of Pharmacology and Toxicology, Research Institute on Addictions, Program in Neuroscience, State University at Buffalo, Buffalo, NY, USA
| | - ZJ Wang
- Department of Pharmacology and Toxicology, Research Institute on Addictions, Program in Neuroscience, State University at Buffalo, Buffalo, NY, USA
| | - CK Lardner
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - RC Bagot
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA,Department of Psychology, McGill University, Montréal, QC, Canada
| | - RL Neve
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - DM Dietz
- Department of Pharmacology and Toxicology, Research Institute on Addictions, Program in Neuroscience, State University at Buffalo, Buffalo, NY, USA
| | - EJ Nestler
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
48
|
Peña CJ, Kronman HG, Walker DM, Cates HM, Bagot RC, Purushothaman I, Issler O, Loh YHE, Leong T, Kiraly DD, Goodman E, Neve RL, Shen L, Nestler EJ. Early life stress confers lifelong stress susceptibility in mice via ventral tegmental area OTX2. Science 2018; 356:1185-1188. [PMID: 28619944 DOI: 10.1126/science.aan4491] [Citation(s) in RCA: 260] [Impact Index Per Article: 37.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2017] [Accepted: 05/22/2017] [Indexed: 12/23/2022]
Abstract
Early life stress increases risk for depression. Here we establish a "two-hit" stress model in mice wherein stress at a specific postnatal period increases susceptibility to adult social defeat stress and causes long-lasting transcriptional alterations that prime the ventral tegmental area (VTA)-a brain reward region-to be in a depression-like state. We identify a role for the developmental transcription factor orthodenticle homeobox 2 (Otx2) as an upstream mediator of these enduring effects. Transient juvenile-but not adult-knockdown of Otx2 in VTA mimics early life stress by increasing stress susceptibility, whereas its overexpression reverses the effects of early life stress. This work establishes a mechanism by which early life stress encodes lifelong susceptibility to stress via long-lasting transcriptional programming in VTA mediated by Otx2.
Collapse
Affiliation(s)
- Catherine J Peña
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Hope G Kronman
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Deena M Walker
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Hannah M Cates
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Rosemary C Bagot
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Immanuel Purushothaman
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Orna Issler
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Yong-Hwee Eddie Loh
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Tin Leong
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Drew D Kiraly
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Department of Psychiatry, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Emma Goodman
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Rachael L Neve
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Li Shen
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Eric J Nestler
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
49
|
Beloate LN, Coolen LM. Effects of Sexual Experience on Psychostimulant- and Opiate-Induced Behavior and Neural Plasticity in the Mesocorticolimbic Pathway. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2018; 140:249-270. [DOI: 10.1016/bs.irn.2018.07.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
50
|
Dos Santos M, Salery M, Forget B, Garcia Perez MA, Betuing S, Boudier T, Vanhoutte P, Caboche J, Heck N. Rapid Synaptogenesis in the Nucleus Accumbens Is Induced by a Single Cocaine Administration and Stabilized by Mitogen-Activated Protein Kinase Interacting Kinase-1 Activity. Biol Psychiatry 2017; 82:806-818. [PMID: 28545678 DOI: 10.1016/j.biopsych.2017.03.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 03/10/2017] [Accepted: 03/14/2017] [Indexed: 12/21/2022]
Abstract
BACKGROUND Repeated cocaine exposure produces new spine formation in striatal projection neurons (SPNs) of the nucleus accumbens. However, an acute exposure to cocaine can trigger long-lasting synaptic plasticity in SPNs leading to behavioral alterations. This raises the intriguing question as to whether a single administration of cocaine could enduringly modify striatal connectivity. METHODS A three-dimensional morphometric analysis of presynaptic glutamatergic boutons and dendritic spines was performed on SPNs 1 hour and 1 week after a single cocaine administration. Time-lapse two-photon microscopy in adult slices was used to determine the precise molecular-events sequence responsible for the rapid spine formation. RESULTS A single injection triggered a rapid synaptogenesis and persistent increase in glutamatergic connectivity in SPNs from the shell part of the nucleus accumbens, specifically. Synapse formation occurred through clustered growth of active spines contacting pre-existing axonal boutons. Spine growth required extracellular signal-regulated kinase activation, while spine stabilization involved transcription-independent protein synthesis driven by mitogen-activated protein kinase interacting kinase-1, downstream from extracellular signal-regulated kinase. The maintenance of new spines driven by mitogen-activated protein kinase interacting kinase-1 was essential for long-term connectivity changes induced by cocaine in vivo. CONCLUSIONS Our study originally demonstrates that a single administration of cocaine is able to induce stable synaptic rewiring in the nucleus accumbens, which will likely influence responses to subsequent drug exposure. It also unravels a new functional role for cocaine-induced extracellular signal-regulated kinase pathway independently of nuclear targets. Finally, it reveals that mitogen-activated protein kinase interacting kinase-1 has a pivotal role in cocaine-induced connectivity.
Collapse
Affiliation(s)
- Marc Dos Santos
- Neurosciences Paris Seine, Institut de Biologie Paris Seine, University Pierre and Marie Curie University of Paris 06, Sorbonne Universités, Centre National pour la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Paris, France
| | - Marine Salery
- Neurosciences Paris Seine, Institut de Biologie Paris Seine, University Pierre and Marie Curie University of Paris 06, Sorbonne Universités, Centre National pour la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Paris, France
| | - Benoit Forget
- Neurosciences Paris Seine, Institut de Biologie Paris Seine, University Pierre and Marie Curie University of Paris 06, Sorbonne Universités, Centre National pour la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Paris, France
| | - Maria Alexandra Garcia Perez
- Neurosciences Paris Seine, Institut de Biologie Paris Seine, University Pierre and Marie Curie University of Paris 06, Sorbonne Universités, Centre National pour la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Paris, France
| | - Sandrine Betuing
- Neurosciences Paris Seine, Institut de Biologie Paris Seine, University Pierre and Marie Curie University of Paris 06, Sorbonne Universités, Centre National pour la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Paris, France
| | - Thomas Boudier
- Neurosciences Paris Seine, Institut de Biologie Paris Seine, University Pierre and Marie Curie University of Paris 06, Sorbonne Universités, Centre National pour la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Paris, France; Bioinformatics Institute, Agency for Science, Technology, and Research, Singapore
| | - Peter Vanhoutte
- Neurosciences Paris Seine, Institut de Biologie Paris Seine, University Pierre and Marie Curie University of Paris 06, Sorbonne Universités, Centre National pour la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Paris, France
| | - Jocelyne Caboche
- Neurosciences Paris Seine, Institut de Biologie Paris Seine, University Pierre and Marie Curie University of Paris 06, Sorbonne Universités, Centre National pour la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Paris, France.
| | - Nicolas Heck
- Neurosciences Paris Seine, Institut de Biologie Paris Seine, University Pierre and Marie Curie University of Paris 06, Sorbonne Universités, Centre National pour la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Paris, France
| |
Collapse
|