1
|
Cai Y, Wu JC, Huang Y, Yu XF, Liu FH, Chen ZW, Gao DP. Impaired cognitive function and altered dendritic morphology of hippocampal neurons in a mouse model of fetal alcohol spectrum disorder. Behav Brain Res 2025; 490:115633. [PMID: 40345553 DOI: 10.1016/j.bbr.2025.115633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Revised: 05/01/2025] [Accepted: 05/05/2025] [Indexed: 05/11/2025]
Abstract
Prenatal ethanol exposure is a leading preventable cause of neurodevelopmental disability, clinically categorized under fetal alcohol spectrum disorders (FASD). This study explores how developmental alcohol exposure affects the dendritic morphology of hippocampal pyramidal neurons, focusing on the actin cytoskeleton's dynamics essential for neuronal structure and synaptic function. Within this context, we hypothesized that developmental alcohol exposure disrupts actin cytoskeleton dynamics, leading to cognitive deficits and dendritic remodeling in the hippocampus. Neonatal mice (C57BL/6 J) were administered ethanol (5.0 g/kg) intraperitoneally from postnatal day 2-8, establishing a third trimester-equivalent alcohol exposure FASD model. At postnatal day 28, cognitive performance was evaluated using novel location recognition (NLR), novel object recognition (NOR), and the Morris water maze (MWM). Golgi staining assessed dendritic morphology in the hippocampal CA1 region, and the ratio of polymerized (F-actin) to globular actin (G-actin) was measured using a biochemical assay. The results revealed that developmental alcohol exposure significantly impaired recognition and spatial memory, as evidenced by decreased performances in the NOR and MWM tests across both sexes. Golgi staining revealed reduced dendritic arborization complexity and spine density in the CA1 region of the hippocampal pyramidal neurons of both male and female juvenile mice. Biochemical analyses further revealed decresed hipocampal F-actin/G-actin ratios and decreased levels of polymerized F-actin in both sexes. These findings underscore the critical role of cytoskeletal integrity in cognitive development and highlight potential targets for therapeutic intervention in FASD.
Collapse
Affiliation(s)
- Yu Cai
- School of Pharmacy, Zhejiang Pharmaceutical University, 666 Siming Rd., Ningbo, Zhejiang 315500, PR China
| | - Jia-Chun Wu
- School of Pharmacy, Zhejiang Pharmaceutical University, 666 Siming Rd., Ningbo, Zhejiang 315500, PR China
| | - Ying Huang
- School of Pharmacy, Zhejiang Pharmaceutical University, 666 Siming Rd., Ningbo, Zhejiang 315500, PR China
| | - Xue-Feng Yu
- School of Pharmacy, Zhejiang Pharmaceutical University, 666 Siming Rd., Ningbo, Zhejiang 315500, PR China
| | - Fu-He Liu
- School of Pharmacy, Zhejiang Pharmaceutical University, 666 Siming Rd., Ningbo, Zhejiang 315500, PR China
| | - Zi-Wei Chen
- School of Pharmacy, Zhejiang Pharmaceutical University, 666 Siming Rd., Ningbo, Zhejiang 315500, PR China
| | - Da-Peng Gao
- Department of Neurology, The First Affiliated Hospital of Ningbo University, 247 Renmin Rd., Ningbo, Zhejiang 315020, PR China.
| |
Collapse
|
2
|
Hoisington ZW, Gangal H, Phamluong K, Shukla C, Ehinger Y, Moffat JJ, Homanics GE, Wang J, Ron D. Prosapip1 in the dorsal hippocampus mediates synaptic protein composition, long-term potentiation, and spatial memory. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.06.13.597459. [PMID: 38915579 PMCID: PMC11195216 DOI: 10.1101/2024.06.13.597459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Prosapip1 is a brain-specific protein localized to the postsynaptic density, where it promotes dendritic spine maturation in primary hippocampal neurons. However, nothing is known about the role of Prosapip1 in vivo. To examine this, we utilized the Cre-loxP system to develop a Prosapip1 neuronal knockout mouse. We found that Prosapip1 controls the synaptic localization of its binding partner SPAR, along with PSD-95 and the GluN2B subunit of the NMDA receptor (NMDAR) in the dorsal hippocampus (dHP). We next sought to identify the potential contribution of Prosapip1 to the activity and function of the NMDAR and found that Prosapip1 plays an important role in NMDAR-mediated transmission and long-term potentiation (LTP) in the CA1 region of the dHP. As LTP is the cellular hallmark of learning and memory, we examined the consequences of neuronal knockout of Prosapip1 on dHP-dependent memory. We found that global or dHP-specific neuronal knockout of Prosapip1 caused a deficit in learning and memory whereas developmental, locomotor, and anxiety phenotypes were normal. Taken together, Prosapip1 in the dHP promotes the proper localization of synaptic proteins which, in turn, facilitates LTP driving recognition, social, and spatial learning and memory.
Collapse
|
3
|
Zhao H, Li H, Meng L, Du P, Mo X, Gong M, Chen J, Liao Y. Disrupting heroin-associated memory reconsolidation through actin polymerization inhibition in the nucleus accumbens core. Int J Neuropsychopharmacol 2024; 28:pyae065. [PMID: 39716383 DOI: 10.1093/ijnp/pyae065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 12/23/2024] [Indexed: 12/25/2024] Open
Abstract
BACKGROUND Understanding drug addiction as a disorder of maladaptive learning, where drug-associated or environmental cues trigger drug cravings and seeking, is crucial for developing effective treatments. Actin polymerization, a biochemical process, plays a crucial role in drug-related memory formation, particularly evident in conditioned place preference paradigms involving drugs like morphine and methamphetamine. However, the role of actin polymerization in the reconsolidation of heroin-associated memories remains understudied. METHODS This study employed a rodent model of self-administered heroin to investigate the involvement of actin polymerization in the reconsolidation of heroin-associated memories. Rats underwent ten days of intravenous heroin self-administration paired with conditioned cues. Subsequently, a 10-day extinction phase aimed to reduce heroin-seeking behaviors. Following this, rats participated in a 15-minute retrieval trial with or without cues. Immediately post-retrieval, rats received bilateral injections of the actin polymerization inhibitor Latrunculin A (Lat A) into the nucleus accumbens core (NACc), a critical brain region for memory reconsolidation. RESULTS Immediate administration of Lat A into the NACc post-retrieval significantly reduced cue-induced and heroin-primed reinstatement of heroin-seeking behavior for at least 28 days. However, administering Lat A 6-hour post-retrieval or without a retrieval trial, as well as administering Jasplakionlide prior to memory reactivation did not affect heroin-seeking behaviors. CONCLUSIONS Inhibiting actin polymerization during the reconsolidation window disrupts heroin-associated memory reconsolidation, leading to decreased heroin-seeking behavior and prevention of relapse. These effects are contingent upon the presence of a retrieval trial and exhibit temporal specificity, shedding light on addiction mechanisms and potential therapeutic interventions.
Collapse
Affiliation(s)
- Haiting Zhao
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Haoyu Li
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Li Meng
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Peng Du
- Department of Neurosurgery, The Second Affiliated Hospital, Xinjiang Medical University, Urumqi, China
| | - Xin Mo
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Mengqi Gong
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jiaxin Chen
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yiwei Liao
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
4
|
Hoisington ZW, Salvi A, Laguesse S, Ehinger Y, Shukla C, Phamluong K, Ron D. The Small G-Protein Rac1 in the Dorsomedial Striatum Promotes Alcohol-Dependent Structural Plasticity and Goal-Directed Learning in Mice. J Neurosci 2024; 44:e1644232024. [PMID: 38886056 PMCID: PMC11255432 DOI: 10.1523/jneurosci.1644-23.2024] [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: 08/28/2023] [Revised: 04/04/2024] [Accepted: 04/10/2024] [Indexed: 06/20/2024] Open
Abstract
The small G-protein Ras-related C3 botulinum toxin substrate 1 (Rac1) promotes the formation of filamentous actin (F-actin). Actin is a major component of dendritic spines, and we previously found that alcohol alters actin composition and dendritic spine structure in the nucleus accumbens (NAc) and the dorsomedial striatum (DMS). To examine if Rac1 contributes to these alcohol-mediated adaptations, we measured the level of GTP-bound active Rac1 in the striatum of mice following 7 weeks of intermittent access to 20% alcohol. We found that chronic alcohol intake activates Rac1 in the DMS of male mice. In contrast, Rac1 is not activated by alcohol in the NAc and DLS of male mice or in the DMS of female mice. Similarly, closely related small G-proteins are not activated by alcohol in the DMS, and Rac1 activity is not increased in the DMS by moderate alcohol or natural reward. To determine the consequences of alcohol-dependent Rac1 activation in the DMS of male mice, we inhibited endogenous Rac1 by infecting the DMS of mice with an adeno-associated virus (AAV) expressing a dominant negative form of the small G-protein (Rac1-DN). We found that overexpression of AAV-Rac1-DN in the DMS inhibits alcohol-mediated Rac1 signaling and attenuates alcohol-mediated F-actin polymerization, which corresponded with a decrease in dendritic arborization and spine maturation. Finally, we provide evidence to suggest that Rac1 in the DMS plays a role in alcohol-associated goal-directed learning. Together, our data suggest that Rac1 in the DMS plays an important role in alcohol-dependent structural plasticity and aberrant learning.
Collapse
Affiliation(s)
- Zachary W Hoisington
- Alcohol and Addiction Research Group, Department of Neurology, University of California San Francisco, San Francisco, California 94107
| | - Alexandra Salvi
- Alcohol and Addiction Research Group, Department of Neurology, University of California San Francisco, San Francisco, California 94107
| | - Sophie Laguesse
- GIGA-Stem Cells and GIGA-Neurosciences, Interdisciplinary Cluster for Applied Genoproteomics (GIGA-R), University of Liège, Liège 4000, Belgium
| | - Yann Ehinger
- Alcohol and Addiction Research Group, Department of Neurology, University of California San Francisco, San Francisco, California 94107
| | - Chhavi Shukla
- Alcohol and Addiction Research Group, Department of Neurology, University of California San Francisco, San Francisco, California 94107
| | - Khanhky Phamluong
- Alcohol and Addiction Research Group, Department of Neurology, University of California San Francisco, San Francisco, California 94107
| | - Dorit Ron
- Alcohol and Addiction Research Group, Department of Neurology, University of California San Francisco, San Francisco, California 94107
| |
Collapse
|
5
|
Ehinger Y, Phamluong K, Ron D. Sex Differences In The Interaction Between Alcohol And mTORC1. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.04.560781. [PMID: 38712221 PMCID: PMC11071286 DOI: 10.1101/2023.10.04.560781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
The kinase mechanistic target of rapamycin complex 1 (mTORC1) plays an essential role in learning and memory by promoting mRNA to protein translation of a subset of synaptic proteins at dendrites. We generated a large body of data in male rodents indicating that mTORC1 is critically involved in mechanisms that promote numerous adverse behaviors associated with alcohol use disorder (AUD) including heavy alcohol use. For example, we found that mTORC1 is activated in the nucleus accumbens (NAc) and orbitofrontal cortex (OFC) of male mice and rats that were subjected to 7 weeks of intermittent access to 20% alcohol two-bottle choice (IA20%2BC). We further showed that systemic or intra-NAc administration of the selective mTORC1 inhibitor, rapamycin, decreases alcohol seeking and drinking, whereas intra-OFC administration of rapamycin reduces alcohol seeking and habit in male rats. This study aimed to assess mTORC1 activation in these corticostriatal regions of female mice and to determine whether the selective mTORC1 inhibitor, rapamycin, can be used to reduce heavy alcohol use in female mice. We found that mTORC1 is not activated by 7 weeks of intermittent 20% alcohol binge drinking and withdrawal in the NAc and OFC. Like in males, mTORC1 signaling was not activated by chronic alcohol intake and withdrawal in the medial prefrontal cortex (mPFC) of female mice. Interestingly, Pearson correlation comparisons revealed that the basal level of mTORC1 activation between the two prefrontal regions, OFC and mPFC were correlated and that the drinking profile predicts the level of mTORC1 activation in the mPFC after 4-hour binge drinking. Finally, we report that administration of rapamycin does not attenuate heavy alcohol drinking in female animals. Together, our results suggest a sex-dependent contribution of mTORC1 to the neuroadaptation that drives alcohol use and abuse.
Collapse
|
6
|
Pandey S, Miller CA. Targeting the cytoskeleton as a therapeutic approach to substance use disorders. Pharmacol Res 2024; 202:107143. [PMID: 38499081 PMCID: PMC11034636 DOI: 10.1016/j.phrs.2024.107143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 03/06/2024] [Accepted: 03/12/2024] [Indexed: 03/20/2024]
Abstract
Substance use disorders (SUD) are chronic relapsing disorders governed by continually shifting cycles of positive drug reward experiences and drug withdrawal-induced negative experiences. A large body of research points to plasticity within systems regulating emotional, motivational, and cognitive processes as drivers of continued compulsive pursuit and consumption of substances despite negative consequences. This plasticity is observed at all levels of analysis from molecules to networks, providing multiple avenues for intervention in SUD. The cytoskeleton and its regulatory proteins within neurons and glia are fundamental to the structural and functional integrity of brain processes and are potentially the major drivers of the morphological and behavioral plasticity associated with substance use. In this review, we discuss preclinical studies that provide support for targeting the brain cytoskeleton as a therapeutic approach to SUD. We focus on the interplay between actin cytoskeleton dynamics and exposure to cocaine, methamphetamine, alcohol, opioids, and nicotine and highlight preclinical studies pointing to a wide range of potential therapeutic targets, such as nonmuscle myosin II, Rac1, cofilin, prosapip 1, and drebrin. These studies broaden our understanding of substance-induced plasticity driving behaviors associated with SUD and provide new research directions for the development of SUD therapeutics.
Collapse
Affiliation(s)
- Surya Pandey
- Department of Molecular Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL 33458, United States; Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL 33458, United States
| | - Courtney A Miller
- Department of Molecular Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL 33458, United States; Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL 33458, United States.
| |
Collapse
|
7
|
Zhou M, Wu B, Jeong H, Burke DA, Namboodiri VMK. An open-source behavior controller for associative learning and memory (B-CALM). Behav Res Methods 2024; 56:2695-2710. [PMID: 37464151 PMCID: PMC10898869 DOI: 10.3758/s13428-023-02182-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/23/2023] [Indexed: 07/20/2023]
Abstract
Associative learning and memory, i.e., learning and remembering the associations between environmental stimuli, self-generated actions, and outcomes such as rewards or punishments, are critical for the well-being of animals. Hence, the neural mechanisms underlying these processes are extensively studied using behavioral tasks in laboratory animals. Traditionally, these tasks have been controlled using commercial hardware and software, which limits scalability and accessibility due to their cost. More recently, due to the revolution in microcontrollers or microcomputers, several general-purpose and open-source solutions have been advanced for controlling neuroscientific behavioral tasks. While these solutions have great strength due to their flexibility and general-purpose nature, for the same reasons, they suffer from some disadvantages including the need for considerable programming expertise, limited online visualization, or slower than optimal response latencies for any specific task. Here, to mitigate these concerns, we present an open-source behavior controller for associative learning and memory (B-CALM). B-CALM provides an integrated suite that can control a host of associative learning and memory behaviors. As proof of principle for its applicability, we show data from head-fixed mice learning Pavlovian conditioning, operant conditioning, discrimination learning, as well as a timing task and a choice task. These can be run directly from a user-friendly graphical user interface (GUI) written in MATLAB that controls many independently running Arduino Mega microcontrollers in parallel (one per behavior box). In sum, B-CALM will enable researchers to execute a wide variety of associative learning and memory tasks in a scalable, accurate, and user-friendly manner.
Collapse
Affiliation(s)
- Mingkang Zhou
- Department of Neurology, University of California, San Francisco, CA, USA
- Neuroscience Graduate Program, University of California, San Francisco, CA, USA
| | - Brenda Wu
- Department of Neurology, University of California, San Francisco, CA, USA
| | - Huijeong Jeong
- Department of Neurology, University of California, San Francisco, CA, USA
| | - Dennis A Burke
- Department of Neurology, University of California, San Francisco, CA, USA
| | - Vijay Mohan K Namboodiri
- Department of Neurology, University of California, San Francisco, CA, USA.
- Neuroscience Graduate Program, University of California, San Francisco, CA, USA.
- Weill Institute for Neuroscience, Kavli Institute for Fundamental Neuroscience, Center for Integrative Neuroscience, University of California, San Francisco, CA, USA.
| |
Collapse
|
8
|
Carrette L, Santos A, Brennan M, Othman D, Collazo A, George O. Antagonists of the stress and opioid systems restore the functional connectivity of the prefrontal cortex during alcohol withdrawal through divergent mechanisms. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.30.560339. [PMID: 37873478 PMCID: PMC10592857 DOI: 10.1101/2023.09.30.560339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Chronic alcohol consumption leads to dependence and withdrawal symptoms upon cessation, contributing to persistent use. However, the brain network mechanisms by which the brain orchestrates alcohol withdrawal and how these networks are affected by pharmacological treatments remain elusive. Recent work revealed that alcohol withdrawal produces a widespread increase in coordinated brain activity and a decrease in modularity of the whole-brain functional network using single-cell whole-brain imaging of immediate early genes. This decreased modularity and functional hyperconnectivity are hypothesized to be novel biomarkers of alcohol withdrawal in alcohol dependence, which could potentially be used to evaluate the efficacy of new medications for alcohol use disorder. However, there is no evidence that current FDA-approved medications or experimental treatments known to reduce alcohol drinking in animal models can normalize the changes in whole-brain functional connectivity. In this report, we tested the effect of R121919, a CRF1 antagonist, and naltrexone, an FDA-approved treatment for alcohol use disorder, on whole-brain functional connectivity using the cellular marker FOS combined with graph theory and advanced network analyses. Results show that both R121919 and naltrexone restored the functional connectivity of the prefrontal cortex during alcohol withdrawal, but through divergent mechanisms. Specifically, R121919 increased FOS activation in the prefrontal cortex, partially restored modularity, and normalized connectivity, particularly in CRF1-rich regions, including the prefrontal, pallidum, and extended amygdala circuits. On the other hand, naltrexone decreased FOS activation throughout the brain, decreased modularity, and increased connectivity overall except for the Mu opioid receptor-rich regions, including the thalamus. These results identify the brain networks underlying the pharmacological effects of R121919 and naltrexone and demonstrate that these drugs restored different aspects of functional connectivity of the prefrontal cortex, pallidum, amygdala, and thalamus during alcohol withdrawal. Notably, these effects were particularly prominent in CRF1- and Mu opioid receptors-rich regions highlighting the potential of whole-brain functional connectivity using FOS as a tool for identifying neuronal network mechanisms underlying the pharmacological effects of existing and new medications for alcohol use disorder.
Collapse
Affiliation(s)
- L.L.G. Carrette
- Department of Psychiatry, UC San Diego, La Jolla, CA, United States
| | - A. Santos
- Department of Psychiatry, UC San Diego, La Jolla, CA, United States
| | - M. Brennan
- Department of Psychiatry, UC San Diego, La Jolla, CA, United States
| | - D. Othman
- Department of Psychiatry, UC San Diego, La Jolla, CA, United States
| | - A. Collazo
- Beckman Institute, CalTech, Passadena, CA, United States
| | - O. George
- Department of Psychiatry, UC San Diego, La Jolla, CA, United States
| |
Collapse
|
9
|
Gu X, Chen S, Wang Z, Bu Q, An S. LZTS3/TAGLN Suppresses Cancer Progression in Human Colorectal Adenocarcinoma Through Regulating Cell Proliferation, Migration, and Actin Cytoskeleton. Arch Med Res 2023; 54:102894. [PMID: 37806182 DOI: 10.1016/j.arcmed.2023.102894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 08/23/2023] [Accepted: 09/28/2023] [Indexed: 10/10/2023]
Abstract
BACKGROUND Numerous studies have confirmed that the leucine zipper tumor suppressor (LZTS) gene family plays a vital role in modulating transcription and cell cycle control, especially in colorectal cancer. This study aimed to evaluate the potential of leucine zipper tumor suppressor family member 3 (LZTS3) as a marker for COAD. METHODS Bioinformatics, immunohistochemistry, and Western blotting were applied to assess the expression of LZTS3 in tissues. Gene overexpression or silencing was used to examine the biological roles of LZTS3 and validated using an in vivo nude mouse-human tumor model. RESULTS The results obtained in this study indicate that LZTS3 is highly expressed in COAD. RTCA, Transwell, actin stain, and in vitro transfection experiments confirmed that LZTS3 expression inhibits tumor cell proliferation and cell migration. The results obtained in the nude mouse-human tumor model are consistent with those obtained in vitro. In particular, LZTS3 may exert biological effects by targeting the NOTCH signaling pathway. Furthermore, TAGLN was demonstrated to be a downstream target of LZTS3. CONCLUSION This is the first study to demonstrate the important role of LZTS3 in the proliferation and migration of COAD and to shed light on the molecular mechanism underlying the tumor-suppressing role of LZTS3.
Collapse
Affiliation(s)
- Xinpei Gu
- Department of Human Anatomy, Shandong First Medical University and Shandong Academy of Medical Sciences, Tai'an, China; School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou, China
| | - Shuhui Chen
- Department of Gastrointestinal surgery, The Affiliated Tai'an City Central Hospital of Qingdao University, Tai'an, Shandong, China
| | - Zhaojin Wang
- Department of Human Anatomy, Shandong First Medical University and Shandong Academy of Medical Sciences, Tai'an, China
| | - Qianwen Bu
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China
| | - Shuhong An
- Department of Human Anatomy, Shandong First Medical University and Shandong Academy of Medical Sciences, Tai'an, China.
| |
Collapse
|
10
|
Pagano R, Salamian A, Skonieczna E, Wojtas B, Gielniewski B, Harda Z, Cały A, Havekes R, Abel T, Radwanska K. Molecular fingerprints in the hippocampus of alcohol seeking during withdrawal. RESEARCH SQUARE 2023:rs.3.rs-3337670. [PMID: 37841864 PMCID: PMC10571638 DOI: 10.21203/rs.3.rs-3337670/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/17/2023]
Abstract
Alcohol use disorder (AUD) is characterized by pathological motivation to consume alcohol and cognitive inflexibility, leading to excessive alcohol seeking and use. Due to limited understanding of the molecular basis of the disease, there are few pharmacological interventions available to combat AUD. In this study, we aimed to investigate the molecular correlates of impaired extinction of alcohol seeking during alcohol withdrawal using a mouse model of AUD implemented in the automated IntelliCage social system. This model enabled us to distinguish between animals exhibiting AUD-prone and AUD-resistant phenotypes, based on the presence of ≥ 2 or < 2 criteria of AUD, respectively. We utilized new generation RNA sequencing to identify genes that were differentially expressed in the hippocampus and amygdala of mice meeting ≥ 2 or < 2 criteria, as these brain regions are implicated in alcohol motivation, seeking, consumption and the cognitive inflexibility characteristic of AUD. To complement the sequencing studies, we conducted ex vivo electrophysiology experiments. Our findings revealed significant dysregulation of the hippocampal genes associated with the actin cytoskeleton and synaptic function, including actin binding molecule cofilin, during alcohol withdrawal in mice meeting ≥ 2 criteria compared to those meeting < 2 criteria. Moreover, this dysregulation was accompanied by impaired synaptic transmission in the molecular layer of the hippocampal dentate gyrus (ML-DG). Additionally, we demonstrated that overexpression of cofilin in the polymorphic layer of the hippocampal dentate gyrus (PoDG) inhibited ML-DG synapses, increased motivation to seek alcohol, impaired extinction of alcohol seeking and increased correlation between AUD behaviors, resembling the phenotype observed in mice meeting ≥ 2 criteria. Overall, our study uncovers a novel mechanism linking increased hippocampal cofilin expression with the AUD phenotype.
Collapse
Affiliation(s)
- Roberto Pagano
- Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur St., Warsaw 02-093, Poland
| | - Ahmad Salamian
- Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur St., Warsaw 02-093, Poland
| | - Edyta Skonieczna
- Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur St., Warsaw 02-093, Poland
| | - Bartosz Wojtas
- Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur St., Warsaw 02-093, Poland
| | - Bartek Gielniewski
- Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur St., Warsaw 02-093, Poland
| | - Zofia Harda
- Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur St., Warsaw 02-093, Poland
- current address: Department Molecular Neuropharmacology, Maj Institute of Pharmacology of Polish Academy of Sciences, Krakow, Poland
| | - Anna Cały
- Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur St., Warsaw 02-093, Poland
| | - Robbert Havekes
- Neurobiology expertise group, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, The Netherlands
| | - Ted Abel
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, IA, USA; Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Kasia Radwanska
- Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur St., Warsaw 02-093, Poland
| |
Collapse
|
11
|
Pagano R, Salamian A, Skonieczna E, Wojtas B, Gielniewski B, Harda Z, Cały A, Havekes R, Abel T, Radwanska K. Molecular fingerprints in the hippocampus of alcohol seeking during withdrawal. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.24.554622. [PMID: 37662388 PMCID: PMC10473700 DOI: 10.1101/2023.08.24.554622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Alcohol use disorder (AUD) is characterized by excessive alcohol seeking and use. Here, we investigated the molecular correlates of impaired extinction of alcohol seeking using a multidimentional mouse model of AUD. We distinguished AUD-prone and AUD-resistant mice, based on the presence of ≥ 2 or < 2 criteria of AUD and utilized RNA sequencing to identify genes that were differentially expressed in the hippocampus and amygdala of mice meeting ≥ 2 or < 2 criteria, as these brain regions are implicated in alcohol motivation, seeking, consumption and the cognitive inflexibility characteristic of AUD. Our findings revealed dysregulation of the genes associated with the actin cytoskeleton, including actin binding molecule cofilin, and impaired synaptic transmission in the hippocampi of mice meeting ≥ 2 criteria. Overexpression of cofilin in the polymorphic layer of the dentate gyrus (PoDG) inhibited ML-DG synapses, increased motivation to seek alcohol and impaired extinction of alcohol seeking, resembling the phenotype observed in mice meeting ≥ 2 criteria. Overall, our study uncovers a novel mechanism linking increased hippocampal cofilin expression with the AUD phenotype.
Collapse
Affiliation(s)
- Roberto Pagano
- Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur St., Warsaw 02-093, Poland
| | - Ahmad Salamian
- Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur St., Warsaw 02-093, Poland
| | - Edyta Skonieczna
- Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur St., Warsaw 02-093, Poland
| | - Bartosz Wojtas
- Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur St., Warsaw 02-093, Poland
| | - Bartek Gielniewski
- Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur St., Warsaw 02-093, Poland
| | - Zofia Harda
- Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur St., Warsaw 02-093, Poland
- current address: Department Molecular Neuropharmacology, Maj Institute of Pharmacology of Polish Academy of Sciences, Krakow, Poland
| | - Anna Cały
- Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur St., Warsaw 02-093, Poland
| | - Robbert Havekes
- Neurobiology expertise group, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, The Netherlands
| | - Ted Abel
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, IA, USA; Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Kasia Radwanska
- Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur St., Warsaw 02-093, Poland
| |
Collapse
|
12
|
Cały A, Ziółkowska M, Pagano R, Salamian A, Śliwińska MA, Sotoudeh N, Bernaś T, Radwanska K. Autophosphorylation of αCaMKII regulates alcohol consumption by controlling sedative effects of alcohol and alcohol-induced loss of excitatory synapses. Addict Biol 2023; 28:e13276. [PMID: 37186439 DOI: 10.1111/adb.13276] [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: 01/24/2023] [Revised: 03/07/2023] [Accepted: 03/20/2023] [Indexed: 05/17/2023]
Abstract
Calcium/calmodulin-dependent kinase II (CaMKII) is a key enzyme at the glutamatergic synapses. CAMK2A gene variants have been linked with alcohol use disorder (AUD) by an unknown mechanism. Here, we looked for the link between αCaMKII autophosphorylation and the AUD aetiology. Autophosphorylation-deficient heterozygous αCaMKII mutant mice (T286A+/- ) were trained in the IntelliCages to test the role of αCaMKII activity in AUD-related behaviours. The glutamatergic synapses morphology in CeA was studied in the animals drinking alcohol using 3D electron microscopy. We found that T286A+/- mutants consumed less alcohol and were more sensitive to sedating effects of alcohol, as compared to wild-type littermates (WT). After voluntary alcohol drinking, T286A+/- mice had less excitatory synapses in the CeA, as compared to alcohol-naive animals. This change correlated with alcohol consumption was not reversed after alcohol withdrawal and not observed in WT mice. Our study suggests that αCaMKII autophosphorylation affects alcohol consumption by controlling sedative effects of alcohol and preventing synaptic loss in the individuals drinking alcohol. This finding advances our understanding of the molecular processes that regulate alcohol dependence.
Collapse
Affiliation(s)
- Anna Cały
- Laboratory of Molecular Basis of Behavior, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Magdalena Ziółkowska
- Laboratory of Molecular Basis of Behavior, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Roberto Pagano
- Laboratory of Molecular Basis of Behavior, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
- Laboratory of Molecular and Cellular Neurobiology, International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Ahmad Salamian
- Laboratory of Molecular Basis of Behavior, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Małgorzata A Śliwińska
- Laboratory of Imaging Tissue Structure and Function, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Narges Sotoudeh
- Laboratory of Molecular Basis of Behavior, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Tytus Bernaś
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Kasia Radwanska
- Laboratory of Molecular Basis of Behavior, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
13
|
Moffat JJ, Sakhai SA, Hoisington ZW, Ehinger Y, Ron D. The BDNF Val68Met polymorphism causes a sex specific alcohol preference over social interaction and also acute tolerance to the anxiolytic effects of alcohol, a phenotype driven by malfunction of BDNF in the ventral hippocampus of male mice. Psychopharmacology (Berl) 2023; 240:303-317. [PMID: 36622381 PMCID: PMC9879818 DOI: 10.1007/s00213-022-06305-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 12/19/2022] [Indexed: 01/10/2023]
Abstract
BACKGROUND The brain-derived neurotrophic factor (BDNF) Valine 66 to Methionine human polymorphism results in impaired activity-dependent BDNF release and has been linked to psychiatric disorders including depression and anxiety. We previously showed that male knock-in mice carrying the mouse Methionine homolog (Met68BDNF) exhibit excessive and compulsive alcohol drinking behaviors as compared to the wild-type Val68BDNF mice. OBJECTIVE Here, we set out to determine the potential mechanism for the heightened and compulsive alcohol drinking phenotypes detected in Met68BDNF mice. RESULTS We found that male, but not female Met68BDNF mice exhibit social anxiety-like behaviors. We further show that male Met68BDNF mice exhibit a preference for alcohol over social interaction. In contrast, alcohol place preference without an alternative social reward, is similar in male Met68BDNF and Val68BDNF mice. Since the Met68BDNF mice show social anxiety phenotypes, we tested whether alcohol reliefs anxiety similarly in Met68BDNF and Val68BDNF mice and found that male, but not female Met68BDNF mice are insensitive to the acute anxiolytic action of alcohol. Finally, we show that this acute tolerance to alcohol-dependent anxiolysis can be restored by overexpressing wild-type Val68BDNF in the ventral hippocampus (vHC) of Met68BDNF mice. CONCLUSIONS Together, our results suggest that excessive alcohol drinking in the Met68BDNF may be attributed, in part, to heighted social anxiety and a lack of alcohol-dependent anxiolysis, a phenotype that is associated with malfunction of BDNF signaling in the vHC of male Met68BDNF mice.
Collapse
Affiliation(s)
- Jeffrey J Moffat
- Department of Neurology, University of California, 675 Nelson Rising Lane, BOX 0663, San Francisco, CA, 94143-0663, USA
| | - Samuel A Sakhai
- Department of Neurology, University of California, 675 Nelson Rising Lane, BOX 0663, San Francisco, CA, 94143-0663, USA
| | - Zachary W Hoisington
- Department of Neurology, University of California, 675 Nelson Rising Lane, BOX 0663, San Francisco, CA, 94143-0663, USA
| | - Yann Ehinger
- Department of Neurology, University of California, 675 Nelson Rising Lane, BOX 0663, San Francisco, CA, 94143-0663, USA
| | - Dorit Ron
- Department of Neurology, University of California, 675 Nelson Rising Lane, BOX 0663, San Francisco, CA, 94143-0663, USA.
| |
Collapse
|
14
|
Nuñez-delMoral A, Bianchi PC, Brocos-Mosquera I, Anesio A, Palombo P, Camarini R, Cruz FC, Callado LF, Vialou V, Erdozain AM. The Matricellular Protein Hevin Is Involved in Alcohol Use Disorder. Biomolecules 2023; 13:biom13020234. [PMID: 36830603 PMCID: PMC9953008 DOI: 10.3390/biom13020234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 01/16/2023] [Accepted: 01/21/2023] [Indexed: 01/27/2023] Open
Abstract
Astrocytic-secreted matricellular proteins have been shown to influence various aspects of synaptic function. More recently, they have been found altered in animal models of psychiatric disorders such as drug addiction. Hevin (also known as Sparc-like 1) is a matricellular protein highly expressed in the adult brain that has been implicated in resilience to stress, suggesting a role in motivated behaviors. To address the possible role of hevin in drug addiction, we quantified its expression in human postmortem brains and in animal models of alcohol abuse. Hevin mRNA and protein expression were analyzed in the postmortem human brain of subjects with an antemortem diagnosis of alcohol use disorder (AUD, n = 25) and controls (n = 25). All the studied brain regions (prefrontal cortex, hippocampus, caudate nucleus and cerebellum) in AUD subjects showed an increase in hevin levels either at mRNA or/and protein levels. To test if this alteration was the result of alcohol exposure or indicative of a susceptibility factor to alcohol consumption, mice were exposed to different regimens of intraperitoneal alcohol administration. Hevin protein expression was increased in the nucleus accumbens after withdrawal followed by a ethanol challenge. The role of hevin in AUD was determined using an RNA interference strategy to downregulate hevin expression in nucleus accumbens astrocytes, which led to increased ethanol consumption. Additionally, ethanol challenge after withdrawal increased hevin levels in blood plasma. Altogether, these results support a novel role for hevin in the neurobiology of AUD.
Collapse
Affiliation(s)
- Amaia Nuñez-delMoral
- Department of Pharmacology, University of the Basque Country, UPV/EHU, 48940 Leioa, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain
| | - Paula C. Bianchi
- Department of Pharmacology, Universidade Federal de São Paulo-UNIFESP, São Paulo 04023-062, Brazil
| | - Iria Brocos-Mosquera
- Department of Pharmacology, University of the Basque Country, UPV/EHU, 48940 Leioa, Spain
| | - Augusto Anesio
- Department of Pharmacology, Universidade Federal de São Paulo-UNIFESP, São Paulo 04023-062, Brazil
| | - Paola Palombo
- Department of Pharmacology, Universidade Federal de São Paulo-UNIFESP, São Paulo 04023-062, Brazil
| | - Rosana Camarini
- Department of Pharmacology, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-000, Brazil
| | - Fabio C. Cruz
- Department of Pharmacology, Universidade Federal de São Paulo-UNIFESP, São Paulo 04023-062, Brazil
| | - Luis F. Callado
- Department of Pharmacology, University of the Basque Country, UPV/EHU, 48940 Leioa, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain
- Biocruces-Bizkaia Health Research Institute, 48903 Barakaldo, Spain
| | - Vincent Vialou
- Institute of Biology Paris Seine, Neuroscience Paris Seine, CNRS UMR8246, INSERM U1130, Sorbonne Université, 75005 Paris, France
- Correspondence: (V.V.); (A.M.E.); Tel.: +33-1-44-27-60-98 (V.V.); +34-601-28-48 (A.M.E.)
| | - Amaia M. Erdozain
- Department of Pharmacology, University of the Basque Country, UPV/EHU, 48940 Leioa, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain
- Correspondence: (V.V.); (A.M.E.); Tel.: +33-1-44-27-60-98 (V.V.); +34-601-28-48 (A.M.E.)
| |
Collapse
|
15
|
Ben Hamida S, Carter M, Darcq E, Sourty M, Rahman MT, Decker AM, Jin C, Kieffer BL. The GPR88 agonist RTI-13951-33 reduces alcohol drinking and seeking in mice. Addict Biol 2022; 27:e13227. [PMID: 36301207 PMCID: PMC9539967 DOI: 10.1111/adb.13227] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 08/08/2022] [Accepted: 08/10/2022] [Indexed: 01/26/2023]
Abstract
GPR88 is an orphan G-protein-coupled receptor that is considered a potential target to treat neuropsychiatric disorders, including addiction. Most knowledge about GPR88 function stems from knockout mouse studies, and in vivo pharmacology is still scarce. Here we examine the effects of the novel brain-penetrant agonist RTI-13951-33 on several alcohol-related behaviours in the mouse. In the intermittent-access-two-bottle-choice paradigm, the compound reduced excessive voluntary alcohol drinking, while water drinking was intact. This was observed for C57BL/6 mice, as well as for control but not Gpr88 knockout mice, demonstrating efficacy and specificity of the drug in vivo. In the drinking-in-the-dark paradigm, RTI-13951-33 also reduced binge-like drinking behaviour for control but not Gpr88 knockout mice, confirming the alcohol consumption-reducing effect and in vivo specificity of the drug. When C57BL/6 mice were trained for alcohol self-administration, RTI-13951-33 decreased the number of nose-pokes over a 4-h session and reduced the number of licks and bursts of licks, suggesting reduced motivation to obtain alcohol. Finally, RTI-13951-33 did not induce any place preference or aversion but reduced the expression of conditioned place preference to alcohol, indicative of a reduction of alcohol-reward seeking. Altogether, data show that RTI-13951-33 limits alcohol intake under distinct conditions that require consummatory behaviour, operant response or association with contextual cues. RTI-13951-33 therefore is a promising lead compound to evaluate GPR88 as a therapeutic target for alcohol use disorders. More broadly, RTI-13951-33 represents a unique tool to better understand GPR88 function, disentangle receptor roles in development from those in the adult and perhaps address other neuropsychiatric disorders.
Collapse
Affiliation(s)
- Sami Ben Hamida
- Douglas Mental Health University InstituteMontrealQuebecCanada
- INSERM UMR 1247University of Picardie Jules VerneAmiensFrance
| | - Michelle Carter
- Douglas Mental Health University InstituteMontrealQuebecCanada
| | - Emmanuel Darcq
- Douglas Mental Health University InstituteMontrealQuebecCanada
- Department of PsychiatryMcGill UniversityMontrealQuebecCanada
- INSERM U1114University of StrasbourgStrasbourgFrance
| | - Marion Sourty
- INSERM U1114University of StrasbourgStrasbourgFrance
- Laboratory of Engineering, Informatics and Imaging (ICube), Integrative Multimodal Imaging in Healthcare Team (IMIS), CNRS UMR 7357University of StrasbourgStrasbourgFrance
| | - Md Toufiqur Rahman
- Center for Drug DiscoveryResearch Triangle InstituteResearch Triangle ParkNorth CarolinaUSA
| | - Ann M. Decker
- Center for Drug DiscoveryResearch Triangle InstituteResearch Triangle ParkNorth CarolinaUSA
| | - Chunyang Jin
- Center for Drug DiscoveryResearch Triangle InstituteResearch Triangle ParkNorth CarolinaUSA
| | - Brigitte L. Kieffer
- Douglas Mental Health University InstituteMontrealQuebecCanada
- Department of PsychiatryMcGill UniversityMontrealQuebecCanada
- INSERM U1114University of StrasbourgStrasbourgFrance
| |
Collapse
|
16
|
A Novel CaMKII Inhibitory Peptide Blocks Relapse to Morphine Seeking by Influencing Synaptic Plasticity in the Nucleus Accumbens Shell. Brain Sci 2022; 12:brainsci12080985. [PMID: 35892425 PMCID: PMC9394410 DOI: 10.3390/brainsci12080985] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/12/2022] [Accepted: 07/23/2022] [Indexed: 12/04/2022] Open
Abstract
Drugs of abuse cause enduring functional disorders in the brain reward circuits, leading to cravings and compulsive behavior. Although people may rehabilitate by detoxification, there is a high risk of relapse. Therefore, it is crucial to illuminate the mechanisms of relapse and explore the therapeutic strategies for prevention. In this research, by using an animal model of morphine self-administration in rats and a whole-cell patch–clamp in brain slices, we found changes in synaptic plasticity in the nucleus accumbens (NAc) shell were involved in the relapse to morphine-seeking behavior. Compared to the controls, the amplitude of long-term depression (LTD) induced in the medium spiny neurons increased after morphine self-administration was established, recovered after the behavior was extinguished, and increased again during the relapse induced by morphine priming. Intravenous injection of MA, a new peptide obtained by modifying Ca2+/calmodulin-dependent protein kinase II (CaMKII) inhibitor “myr-AIP”, decreased CaMKII activity in the NAc shell and blocked the reinstatement of morphine-seeking behavior without influence on the locomotor activity. Moreover, LTD was absent in the NAc shell of the MA-pretreated rats, whereas it was robust in the saline controls in which morphine-seeking behavior was reinstated. These results indicate that CaMKII regulates morphine-seeking behavior through its involvement in the change of synaptic plasticity in the NAc shell during the relapse, and MA may be of great value in the clinical treatment of relapse to opioid seeking.
Collapse
|
17
|
Van Hees L, Didone V, Charlet‐Briart M, Van Ingelgom T, Alexandre A, Quertemont E, Nguyen L, Laguesse S. Voluntary alcohol binge-drinking in adolescent C57Bl6 mice induces delayed appearance of behavioural defects in both males and females. Addict Biol 2022; 27:e13102. [PMID: 34611982 PMCID: PMC9285796 DOI: 10.1111/adb.13102] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 07/27/2021] [Accepted: 09/13/2021] [Indexed: 12/20/2022]
Abstract
Adolescence is a developmental period characterized by significant changes in brain architecture and behaviour. The immaturity of the adolescent brain is associated with heightened vulnerability to exogenous agents, including alcohol. Alcohol is the most consumed drug among teenagers, and binge‐drinking during adolescence is a major public health concern. Studies have suggested that adolescent alcohol exposure may interfere with the maturation of frontal brain regions and lead to long‐lasting behavioural consequences. In this study, by using a slightly modified version of the Drinking in the Dark paradigm, adolescent C57Bl6 mice reach high blood alcohol concentration after voluntary binge‐drinking. In order to assess short‐ and long‐term consequences of adolescent alcohol exposure (AAE), a battery of behavioural tests was performed during late adolescence and during adulthood. We showed that AAE had no short‐term effect on young mice behaviour but rather increased anxiety‐ and depressive‐like behaviours, as well as alcohol consumption during adulthood. Moreover, alcohol binge‐drinking during adolescence dramatically decreased recognition memory performances and behavioural flexibility in both adult males and females. Furthermore, we showed that voluntary consumption of alcohol during adolescence did not trigger any major activation of the innate immune system in the prefrontal cortex. Together, our data suggest that voluntary alcohol binge‐drinking in adolescent mice induces a delayed appearance of behavioural impairments in adulthood.
Collapse
Affiliation(s)
- Laura Van Hees
- GIGA‐Stem Cells, Interdisciplinary Cluster for Applied Genoproteomics (GIGA‐R) University of Liège Liège Belgium
| | - Vincent Didone
- Psychology and Neuroscience of Cognition Research Unit (PsyNCog), Animal models of cognition University of Liège Liège Belgium
| | - Manon Charlet‐Briart
- GIGA‐Stem Cells, Interdisciplinary Cluster for Applied Genoproteomics (GIGA‐R) University of Liège Liège Belgium
| | - Théo Van Ingelgom
- Psychology and Neuroscience of Cognition Research Unit (PsyNCog), Animal models of cognition University of Liège Liège Belgium
| | - Alysson Alexandre
- GIGA‐Stem Cells, Interdisciplinary Cluster for Applied Genoproteomics (GIGA‐R) University of Liège Liège Belgium
| | - Etienne Quertemont
- Psychology and Neuroscience of Cognition Research Unit (PsyNCog), Animal models of cognition University of Liège Liège Belgium
| | - Laurent Nguyen
- GIGA‐Stem Cells, Interdisciplinary Cluster for Applied Genoproteomics (GIGA‐R) University of Liège Liège Belgium
| | - Sophie Laguesse
- GIGA‐Stem Cells, Interdisciplinary Cluster for Applied Genoproteomics (GIGA‐R) University of Liège Liège Belgium
| |
Collapse
|
18
|
Egervari G, Siciliano CA, Whiteley EL, Ron D. Alcohol and the brain: from genes to circuits. Trends Neurosci 2021; 44:1004-1015. [PMID: 34702580 PMCID: PMC8616825 DOI: 10.1016/j.tins.2021.09.006] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 09/08/2021] [Accepted: 09/30/2021] [Indexed: 01/27/2023]
Abstract
Alcohol use produces wide-ranging and diverse effects on the central nervous system. It influences intracellular signaling mechanisms, leading to changes in gene expression, chromatin remodeling, and translation. As a result of these molecular alterations, alcohol affects the activity of neuronal circuits. Together, these mechanisms produce long-lasting cellular adaptations in the brain that in turn can drive the development and maintenance of alcohol use disorder (AUD). We provide an update on alcohol research, focusing on multiple levels of alcohol-induced adaptations, from intracellular changes to changes in neural circuits. A better understanding of how alcohol affects these diverse and interlinked mechanisms may lead to the identification of novel therapeutic targets and to the development of much-needed novel and efficacious treatment options.
Collapse
Affiliation(s)
- Gabor Egervari
- Department of Cell and Developmental Biology, Epigenetics Institute, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Cody A Siciliano
- Department of Pharmacology, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN 37203, USA.
| | - Ellanor L Whiteley
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Dorit Ron
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
19
|
ATP-citrate lyase promotes axonal transport across species. Nat Commun 2021; 12:5878. [PMID: 34620845 PMCID: PMC8497606 DOI: 10.1038/s41467-021-25786-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 08/24/2021] [Indexed: 01/22/2023] Open
Abstract
Microtubule (MT)-based transport is an evolutionary conserved process finely tuned by posttranslational modifications. Among them, α-tubulin acetylation, primarily catalyzed by a vesicular pool of α-tubulin N-acetyltransferase 1 (Atat1), promotes the recruitment and processivity of molecular motors along MT tracks. However, the mechanism that controls Atat1 activity remains poorly understood. Here, we show that ATP-citrate lyase (Acly) is enriched in vesicles and provide Acetyl-Coenzyme-A (Acetyl-CoA) to Atat1. In addition, we showed that Acly expression is reduced upon loss of Elongator activity, further connecting Elongator to Atat1 in a pathway regulating α-tubulin acetylation and MT-dependent transport in projection neurons, across species. Remarkably, comparable defects occur in fibroblasts from Familial Dysautonomia (FD) patients bearing an autosomal recessive mutation in the gene coding for the Elongator subunit ELP1. Our data may thus shine light on the pathophysiological mechanisms underlying FD. Microtubule tracks are important for the transport of molecules within axons. Here, the authors show that ATAT1, the enzyme responsible for acetylating a-tubulin, receives acetyl groups from ATP citrate lyase whose stability is regulated by Elongator, a protein mutated in the neuronal disease Familial dysautonomia.
Collapse
|
20
|
Faccidomo S, Cogan ES, Hon OJ, Hoffman JL, Saunders BL, Eastman VR, Kim M, Taylor SM, McElligott ZA, Hodge CW. Calcium-permeable AMPA receptor activity and GluA1 trafficking in the basolateral amygdala regulate operant alcohol self-administration. Addict Biol 2021; 26:e13049. [PMID: 33955100 DOI: 10.1111/adb.13049] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 04/16/2021] [Indexed: 12/22/2022]
Abstract
Addiction is viewed as maladaptive glutamate-mediated neuroplasticity that is regulated, in part, by calcium-permeable AMPA receptor (CP-AMPAR) activity. However, the contribution of CP-AMPARs to alcohol-seeking behavior remains to be elucidated. We evaluated CP-AMPAR activity in the basolateral amygdala (BLA) as a potential target of alcohol that also regulates alcohol self-administration in C57BL/6J mice. Operant self-administration of sweetened alcohol increased spontaneous EPSC frequency in BLA neurons that project to the nucleus accumbens as compared with behavior-matched sucrose controls indicating an alcohol-specific upregulation of synaptic activity. Bath application of the CP-AMPAR antagonist NASPM decreased evoked EPSC amplitude only in alcohol self-administering mice indicating alcohol-induced synaptic insertion of CP-AMPARs in BLA projection neurons. Moreover, NASPM infusion in the BLA dose-dependently decreased the rate of operant alcohol self-administration providing direct evidence for CP-AMPAR regulation of alcohol reinforcement. As most CP-AMPARs are GluA1-containing, we asked if alcohol alters the activation state of GluA1-containing AMPARs. Immunocytochemistry results showed elevated GluA1-S831 phosphorylation in the BLA of alcohol as compared with sucrose mice. To investigate mechanistic regulation of alcohol self-administration by GluA1-containing AMPARs, we evaluated the necessity of GluA1 trafficking using a TET-ON AAV encoding a dominant-negative GluA1 c-terminus (GluA1ct) that blocks activity-dependent synaptic delivery of native GluA1-containing AMPARs. GluA1ct expression in the BLA reduced alcohol self-administration with no effect on sucrose controls. These results show that CP-AMPAR activity and GluA1 trafficking in the BLA mechanistically regulate the reinforcing effects of sweetened alcohol. Pharmacotherapeutic targeting these mechanisms of maladaptive neuroplasticity may aid medical management of alcohol use disorder.
Collapse
Affiliation(s)
- Sara Faccidomo
- Bowles Center for Alcohol Studies The University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
- Department of Psychiatry The University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| | - Elizabeth S. Cogan
- Bowles Center for Alcohol Studies The University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| | - Olivia J. Hon
- Bowles Center for Alcohol Studies The University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
- Neuroscience Curriculum The University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| | - Jessica L. Hoffman
- Bowles Center for Alcohol Studies The University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| | - Briana L. Saunders
- Bowles Center for Alcohol Studies The University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| | - Vallari R. Eastman
- Bowles Center for Alcohol Studies The University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| | - Michelle Kim
- Bowles Center for Alcohol Studies The University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| | - Seth M. Taylor
- Bowles Center for Alcohol Studies The University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| | - Zoe A. McElligott
- Bowles Center for Alcohol Studies The University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
- Department of Psychiatry The University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
- Department of Pharmacology The University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| | - Clyde W. Hodge
- Bowles Center for Alcohol Studies The University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
- Department of Psychiatry The University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
- Department of Pharmacology The University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| |
Collapse
|
21
|
Ehinger Y, Zhang Z, Phamluong K, Soneja D, Shokat KM, Ron D. Brain-specific inhibition of mTORC1 eliminates side effects resulting from mTORC1 blockade in the periphery and reduces alcohol intake in mice. Nat Commun 2021; 12:4407. [PMID: 34315870 PMCID: PMC8316332 DOI: 10.1038/s41467-021-24567-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 06/23/2021] [Indexed: 12/11/2022] Open
Abstract
Alcohol Use Disorder (AUD) affects a large portion of the population. Unfortunately, efficacious medications to treat the disease are limited. Studies in rodents suggest that mTORC1 plays a crucial role in mechanisms underlying phenotypes such as heavy alcohol intake, habit, and relapse. Thus, mTORC1 inhibitors, which are used in the clinic, are promising therapeutic agents to treat AUD. However, chronic inhibition of mTORC1 in the periphery produces undesirable side effects, which limit their potential use for the treatment of AUD. To overcome these limitations, we designed a binary drug strategy in which male mice were treated with the mTORC1 inhibitor RapaLink-1 together with a small molecule (RapaBlock) to protect mTORC1 activity in the periphery. We show that whereas RapaLink-1 administration blocked mTORC1 activation in the liver, RapaBlock abolished the inhibitory action of Rapalink-1. RapaBlock also prevented the adverse side effects produced by chronic inhibition of mTORC1. Importantly, co-administration of RapaLink-1 and RapaBlock inhibited alcohol-dependent mTORC1 activation in the nucleus accumbens and attenuated alcohol seeking and drinking.
Collapse
Affiliation(s)
- Yann Ehinger
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Ziyang Zhang
- Department of Cellular and Molecular Pharmacology and Howard Hughes Medical Institute, University of California, San Francisco, CA, USA
| | - Khanhky Phamluong
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Drishti Soneja
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Kevan M Shokat
- Department of Cellular and Molecular Pharmacology and Howard Hughes Medical Institute, University of California, San Francisco, CA, USA
| | - Dorit Ron
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
22
|
Xing L, Kubik-Zahorodna A, Namba T, Pinson A, Florio M, Prochazka J, Sarov M, Sedlacek R, Huttner WB. Expression of human-specific ARHGAP11B in mice leads to neocortex expansion and increased memory flexibility. EMBO J 2021; 40:e107093. [PMID: 33938018 PMCID: PMC8246068 DOI: 10.15252/embj.2020107093] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 03/20/2021] [Accepted: 03/25/2021] [Indexed: 12/14/2022] Open
Abstract
Neocortex expansion during human evolution provides a basis for our enhanced cognitive abilities. Yet, which genes implicated in neocortex expansion are actually responsible for higher cognitive abilities is unknown. The expression of human-specific ARHGAP11B in embryonic/foetal mouse, ferret and marmoset neocortex was previously found to promote basal progenitor proliferation, upper-layer neuron generation and neocortex expansion during development, features commonly thought to contribute to increased cognitive abilities. However, a key question is whether this phenotype persists into adulthood and if so, whether cognitive abilities are indeed increased. Here, we generated a transgenic mouse line with physiological ARHGAP11B expression that exhibits increased neocortical size and upper-layer neuron numbers persisting into adulthood. Adult ARHGAP11B-transgenic mice showed altered neurobehaviour, notably increased memory flexibility and a reduced anxiety level. Our data are consistent with the notion that neocortex expansion by ARHGAP11B, a gene implicated in human evolution, underlies some of the altered neurobehavioural features observed in the transgenic mice, such as the increased memory flexibility, a neocortex-associated trait, with implications for the increase in cognitive abilities during human evolution.
Collapse
Affiliation(s)
- Lei Xing
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Agnieszka Kubik-Zahorodna
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Vestec, Czech Republic
| | - Takashi Namba
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Anneline Pinson
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Marta Florio
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Jan Prochazka
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Vestec, Czech Republic
| | - Mihail Sarov
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Radislav Sedlacek
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Vestec, Czech Republic
| | - Wieland B Huttner
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| |
Collapse
|
23
|
Radke AK, Sneddon EA, Frasier RM, Hopf FW. Recent Perspectives on Sex Differences in Compulsion-Like and Binge Alcohol Drinking. Int J Mol Sci 2021; 22:ijms22073788. [PMID: 33917517 PMCID: PMC8038761 DOI: 10.3390/ijms22073788] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/31/2021] [Accepted: 04/02/2021] [Indexed: 12/29/2022] Open
Abstract
Alcohol use disorder remains a substantial social, health, and economic problem and problem drinking levels in women have been increasing in recent years. Understanding whether and how the underlying mechanisms that drive drinking vary by sex is critical and could provide novel, more targeted therapeutic treatments. Here, we examine recent results from our laboratories and others which we believe provide useful insights into similarities and differences in alcohol drinking patterns across the sexes. Findings for binge intake and aversion-resistant, compulsion-like alcohol drinking are considered, since both are likely significant contributors to alcohol problems in humans. We also describe studies regarding mechanisms that may underlie sex differences in maladaptive alcohol drinking, with some focus on the importance of nucleus accumbens (NAcb) core and shell regions, several receptor types (dopamine, orexin, AMPA-type glutamate), and possible contributions of sex hormones. Finally, we discuss how stressors such as early life stress and anxiety-like states may interact with sex differences to contribute to alcohol drinking. Together, these findings underscore the importance and critical relevance of studying female and male mechanisms for alcohol and co-morbid conditions to gain a true and clinically useful understanding of addiction and neuropsychiatric mechanisms and treatment.
Collapse
Affiliation(s)
- Anna K. Radke
- Department of Psychology and Center for Neuroscience and Behavior, Miami University, Oxford, OH 45040, USA;
- Correspondence:
| | - Elizabeth A. Sneddon
- Department of Psychology and Center for Neuroscience and Behavior, Miami University, Oxford, OH 45040, USA;
| | - Raizel M. Frasier
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (R.M.F.); (F.W.H.)
| | - Frederic W. Hopf
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (R.M.F.); (F.W.H.)
| |
Collapse
|
24
|
Ouyang J, Chen X, Su S, Li X, Xu X, Yu X, Ke C, Zhu X. Neuroligin1 Contributes to Neuropathic Pain by Promoting Phosphorylation of Cofilin in Excitatory Neurons. Front Mol Neurosci 2021; 14:640533. [PMID: 33716669 PMCID: PMC7947913 DOI: 10.3389/fnmol.2021.640533] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 02/05/2021] [Indexed: 11/13/2022] Open
Abstract
Neuropathic pain is a kind of chronic pain that remains difficult to treat due to its complicated underlying mechanisms. Accumulating evidence has indicated that enhanced synaptic plasticity of nociceptive interneurons in the superficial spinal dorsal horn contributes to the development of neuropathic pain. Neuroligin1 (NL1) is a type of excitatory postsynaptic adhesion molecule, which can mediate excitatory synaptic activity, hence promoting neuronal activation. Vglut2 is the most common marker of excitatory glutamatergic neurons. To explore the role of NL1 in excitatory neurons in nociceptive regulation, we used transgenic mice with cre recombinase expression driven by the Vglut2 promoter combined with viral vectors to knockdown the expression of NL1 in excitatory neurons in the spinal dorsal horn. We found that NL1 was upregulated in the L4-L6 spinal dorsal horn in Vglut2-cre+/- mouse subjected to spared nerve injury (SNI). Meanwhile, the expression of phosphorylated cofilin (p-cofilin) and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor subunit 1 (GluR1) was also increased. Spinal microinjection of a cre-dependent NL1-targeting RNAi in Vglut2-cre+/- mouse alleviated the neuropathic pain-induced mechanical hypersensitivity and reduced the increase in p-cofilin and GluR1 caused by SNI. Taken together, NL1 in excitatory neurons regulates neuropathic pain by promoting the SNI-dependent increase in p-cofilin and GluR1 in the spinal dorsal horn. Our study provides a better understanding of the role of NL1 in excitatory neurons, which might represent a possible therapeutic target for alleviating neuropathic pain.
Collapse
Affiliation(s)
- Junlin Ouyang
- Department of Orthopedic Rehabilitation, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Xiaping Chen
- Department of Scientific Research, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Shanchun Su
- Department of Anesthesiology, Institute of Anesthesiology and Pain (IAP), Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Xiaohui Li
- Department of Anesthesiology, Institute of Anesthesiology and Pain (IAP), Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Xueqin Xu
- Department of Anesthesiology, Institute of Anesthesiology and Pain (IAP), Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Xinhua Yu
- Department of Orthopedic Rehabilitation, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Changbin Ke
- Department of Anesthesiology, Institute of Anesthesiology and Pain (IAP), Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Xiaohu Zhu
- Department of Orthopedic Rehabilitation, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| |
Collapse
|
25
|
Differential importance of nucleus accumbens Ox1Rs and AMPARs for female and male mouse binge alcohol drinking. Sci Rep 2021; 11:231. [PMID: 33420199 PMCID: PMC7794293 DOI: 10.1038/s41598-020-79935-2] [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: 10/02/2020] [Accepted: 12/15/2020] [Indexed: 02/06/2023] Open
Abstract
Alcohol use disorder exhausts substantial social and economic costs, with recent dramatic increases in female problem drinking. Thus, it is critically important to understand signaling differences underlying alcohol consumption across the sexes. Orexin-1 receptors (Ox1Rs) can strongly promote motivated behavior, and we previously identified Ox1Rs within nucleus accumbens shell (shell) as crucial for driving binge intake in higher-drinking male mice. Here, shell Ox1R inhibition did not alter female mouse alcohol drinking, unlike in males. Also, lower dose systemic Ox1R inhibition reduced compulsion-like alcohol intake in both sexes, indicating that female Ox1Rs can drive some aspects of pathological consumption, and higher doses of systemic Ox1R inhibition (which might have more off-target effects) reduced binge drinking in both sexes. In contrast to shell Ox1Rs, inhibiting shell calcium-permeable AMPA receptors (CP-AMPARs) strongly reduced alcohol drinking in both sexes, which was specific to alcohol since this did not reduce saccharin intake in either sex. Our results together suggest that the shell critically regulates binge drinking in both sexes, with shell CP-AMPARs supporting intake in both sexes, while shell Ox1Rs drove drinking only in males. Our findings provide important new information about sex-specific and -general mechanisms that promote binge alcohol intake and possible targeted therapeutic interventions.
Collapse
|
26
|
Ehinger Y, Morisot N, Phamluong K, Sakhai SA, Soneja D, Adrover MF, Alvarez VA, Ron D. cAMP-Fyn signaling in the dorsomedial striatum direct pathway drives excessive alcohol use. Neuropsychopharmacology 2021; 46:334-342. [PMID: 32417851 PMCID: PMC7852539 DOI: 10.1038/s41386-020-0712-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 05/11/2020] [Indexed: 12/19/2022]
Abstract
Fyn kinase in the dorsomedial striatum (DMS) of rodents plays a central role in mechanisms underlying excessive alcohol intake. The DMS is comprised of medium spiny neurons (MSNs) that project directly (dMSNs) or indirectly (iMSNs) to the substantia nigra. Here, we examined the cell-type specificity of Fyn's actions in alcohol use. First, we knocked down Fyn selectively in DMS dMSNs or iMSNs of mice and measured the level of alcohol consumption. We found that downregulation of Fyn in dMSNs, but not in iMSNs, reduces excessive alcohol but not saccharin intake. D1Rs are coupled to Gαs/olf, which activate cAMP signaling. To examine whether Fyn's actions are mediated through cAMP signaling, DMS dMSNs were infected with GαsDREADD, and the activation of Fyn signaling was measured following CNO treatment. We found that remote stimulation of cAMP signaling in DMS dMSNs activates Fyn and promotes the phosphorylation of the Fyn substrate, GluN2B. In contract, remote activation of GαsDREADD in DLS dMSNs did not alter Fyn signaling. We then tested whether activation of GαsDREADD in DMS dMSNs or iMSNs alters alcohol intake and observed that CNO-dependent activation of GαsDREADD in DMS dMSNs but not iMSNs increases alcohol but not saccharin intake. Finally, we examined the contribution of Fyn to GαsDREADD-dependent increase in alcohol intake, and found that systemic administration of the Fyn inhibitor, AZD0503 blocks GαsDREADD-dependent increase in alcohol consumption. Our results suggest that the cAMP-Fyn axis in the DMS dMSNs is a molecular transducer of mechanisms underlying the development of excessive alcohol consumption.
Collapse
Affiliation(s)
- Yann Ehinger
- Department of Neurology, University of California San Francisco (UCSF), San Francisco, CA, 94143, USA
| | - Nadege Morisot
- Department of Neurology, University of California San Francisco (UCSF), San Francisco, CA, 94143, USA
- Nkarta Therapeutics, San Francisco, CA, USA
| | - Khanhky Phamluong
- Department of Neurology, University of California San Francisco (UCSF), San Francisco, CA, 94143, USA
| | - Samuel A Sakhai
- Department of Neurology, University of California San Francisco (UCSF), San Francisco, CA, 94143, USA
- Sage Therapeutics, San Francisco, CA, USA
| | - Drishti Soneja
- Department of Neurology, University of California San Francisco (UCSF), San Francisco, CA, 94143, USA
| | - Martin F Adrover
- National Institutes of Alcohol Abuse and Alcoholism, National Institute of Health, Bethesda, MD, 20892, USA
- INGEBI, CONICET, Buenos Aires, Argentina
| | - Veronica A Alvarez
- National Institutes of Alcohol Abuse and Alcoholism, National Institute of Health, Bethesda, MD, 20892, USA
- Center on Compulsive Behaviors, Intramural Research Program, National Institute of Health, Bethesda, MD, 20892, USA
| | - Dorit Ron
- Department of Neurology, University of California San Francisco (UCSF), San Francisco, CA, 94143, USA.
| |
Collapse
|
27
|
Strong CE, Kabbaj M. Neural Mechanisms Underlying the Rewarding and Therapeutic Effects of Ketamine as a Treatment for Alcohol Use Disorder. Front Behav Neurosci 2020; 14:593860. [PMID: 33362485 PMCID: PMC7759199 DOI: 10.3389/fnbeh.2020.593860] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 11/06/2020] [Indexed: 11/13/2022] Open
Abstract
Alcohol use disorder (AUD) is the most prevalent substance use disorder and causes a significant global burden. Relapse rates remain incredibly high after decades of attempting to develop novel treatment options that have failed to produce increased rates of sobriety. Ketamine has emerged as a potential treatment for AUD following its success as a therapeutic agent for depression, demonstrated by several preclinical studies showing that acute administration reduced alcohol intake in rodents. As such, ketamine's therapeutic effects for AUD are now being investigated in clinical trials with the hope of it being efficacious in prolonging sobriety from alcohol in humans (ClinicalTrials.gov, Identifier: NCT01558063). Importantly, ketamine's antidepressant effects only last for about 1-week and because AUD is a lifelong disorder, repeated treatment regimens would be necessary to maintain sobriety. This raises questions regarding its safety for AUD treatment since ketamine itself has the potential for addiction. Therefore, this review aims to summarize the neuroadaptations related to alcohol's addictive properties as well as ketamine's therapeutic and addictive properties. To do this, the focus will be on reward-related brain regions such as the nucleus accumbens (NAc), dorsal striatum, prefrontal cortex (PFC), hippocampus, and ventral tegmental area (VTA) to understand how acute vs. chronic exposure will alter reward signaling over time. Additionally, evidence from these studies will be summarized in both male and female subjects. Accordingly, this review aims to address the safety of repeated ketamine infusions for the treatment of AUD. Although more work about the safety of ketamine to treat AUD is warranted, we hope this review sheds light on some answers about the safety of repeated ketamine infusions.
Collapse
Affiliation(s)
- Caroline E Strong
- Program in Neuroscience, Department of Biomedical Sciences, Florida State University, Tallahassee, FL, United States
| | - Mohamed Kabbaj
- Program in Neuroscience, Department of Biomedical Sciences, Florida State University, Tallahassee, FL, United States
| |
Collapse
|
28
|
Ucha M, Roura-Martínez D, Ambrosio E, Higuera-Matas A. The role of the mTOR pathway in models of drug-induced reward and the behavioural constituents of addiction. J Psychopharmacol 2020; 34:1176-1199. [PMID: 32854585 DOI: 10.1177/0269881120944159] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Exposure to drugs of abuse induces neuroadaptations in critical nodes of the so-called reward systems that are thought to mediate the transition from controlled drug use to the compulsive drug-seeking that characterizes addictive disorders. These neural adaptations are likely to require protein synthesis, which is regulated, among others, by the mechanistic target of the rapamycin kinase (mTOR) signalling cascade. METHODS We have performed a narrative review of the literature available in PubMed about the involvement of the mTOR pathway in drug-reward and addiction-related phenomena. AIMS The aim of this study was to review the underlying architecture of this complex intracellular network and to discuss the alterations of its components that are evident after exposure to drugs of abuse. The aim was also to delineate the effects that manipulations of the mTOR network have on models of drug reward and on paradigms that recapitulate some of the psychological components of addiction. RESULTS There is evidence for the involvement of the mTOR pathway in the acute and rewarding effects of drugs of abuse, especially psychostimulants. However, the data regarding opiates are scarce. There is a need to use sophisticated animal models of addiction to ascertain the real role of the mTOR pathway in this pathology and not just in drug-mediated reward. The involvement of this pathway in behavioural addictions and impulsivity should also be studied in detail in the future. CONCLUSIONS Although there is a plethora of data about the modulation of mTOR by drugs of abuse, the involvement of this signalling pathway in addictive disorders requires further research.
Collapse
Affiliation(s)
- Marcos Ucha
- Department of Psychobiology, National University for Distance Learning (UNED), Madrid, Spain
| | - David Roura-Martínez
- Department of Psychobiology, National University for Distance Learning (UNED), Madrid, Spain
| | - Emilio Ambrosio
- Department of Psychobiology, National University for Distance Learning (UNED), Madrid, Spain
| | - Alejandro Higuera-Matas
- Department of Psychobiology, National University for Distance Learning (UNED), Madrid, Spain
| |
Collapse
|
29
|
Kamal H, Tan GC, Ibrahim SF, Shaikh MF, Mohamed IN, Mohamed RMP, Hamid AA, Ugusman A, Kumar J. Alcohol Use Disorder, Neurodegeneration, Alzheimer's and Parkinson's Disease: Interplay Between Oxidative Stress, Neuroimmune Response and Excitotoxicity. Front Cell Neurosci 2020; 14:282. [PMID: 33061892 PMCID: PMC7488355 DOI: 10.3389/fncel.2020.00282] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 08/10/2020] [Indexed: 12/12/2022] Open
Abstract
Alcohol use disorder (AUD) has been associated with neurodegenerative diseases such as Alzheimer's and Parkinson's disease. Prolonged excessive alcohol intake contributes to increased production of reactive oxygen species that triggers neuroimmune response and cellular apoptosis and necrosis via lipid peroxidation, mitochondrial, protein or DNA damage. Long term binge alcohol consumption also upregulates glutamate receptors, glucocorticoids and reduces reuptake of glutamate in the central nervous system, resulting in glutamate excitotoxicity, and eventually mitochondrial injury and cell death. In this review, we delineate the following principles in alcohol-induced neurodegeneration: (1) alcohol-induced oxidative stress, (2) neuroimmune response toward increased oxidants and lipopolysaccharide, (3) glutamate excitotoxicity and cell injury, and (4) interplay between oxidative stress, neuroimmune response and excitotoxicity leading to neurodegeneration and (5) potential chronic alcohol intake-induced development of neurodegenerative diseases, including Alzheimer's and Parkinson's disease.
Collapse
Affiliation(s)
- Haziq Kamal
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Geok Chin Tan
- Department of Pathology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Siti Fatimah Ibrahim
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Mohd. Farooq Shaikh
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Selangor, Malaysia
| | - Isa Naina Mohamed
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Rashidi M. Pakri Mohamed
- Department of Family Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Adila A. Hamid
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Azizah Ugusman
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Jaya Kumar
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
30
|
Halladay LR, Kocharian A, Piantadosi PT, Authement ME, Lieberman AG, Spitz NA, Coden K, Glover LR, Costa VD, Alvarez VA, Holmes A. Prefrontal Regulation of Punished Ethanol Self-administration. Biol Psychiatry 2020; 87:967-978. [PMID: 31937415 PMCID: PMC7217757 DOI: 10.1016/j.biopsych.2019.10.030] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 10/08/2019] [Accepted: 10/25/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND A clinical hallmark of alcohol use disorder is persistent drinking despite potential adverse consequences. The ventromedial prefrontal cortex (vmPFC) and dorsomedial prefrontal cortex (dmPFC) are positioned to exert top-down control over subcortical regions, such as the nucleus accumbens shell (NAcS) and basolateral amygdala, which encode positive and negative valence of ethanol (EtOH)-related stimuli. Prior rodent studies have implicated these regions in regulation of punished EtOH self-administration (EtOH-SA). METHODS We conducted in vivo electrophysiological recordings in mouse vmPFC and dmPFC to obtain neuronal correlates of footshock-punished EtOH-SA. Ex vivo recordings were performed in NAcS D1 receptor-expressing medium spiny neurons receiving vmPFC input to examine punishment-related plasticity in this pathway. Optogenetic photosilencing was employed to assess the functional contribution of the vmPFC, dmPFC, vmPFC projections to NAcS, or vmPFC projections to basolateral amygdala, to punished EtOH-SA. RESULTS Punishment reduced EtOH lever pressing and elicited aborted presses (lever approach followed by rapid retraction). Neurons in the vmPFC and dmPFC exhibited phasic firing to EtOH lever presses and aborts, but only in the vmPFC was there a population-level shift in coding from lever presses to aborts with punishment. Closed-loop vmPFC, but not dmPFC, photosilencing on a postpunishment probe test negated the reduction in EtOH lever presses but not in aborts. Punishment was associated with altered plasticity at vmPFC inputs to D1 receptor-expressing medium spiny neurons in the NAcS. Photosilencing vmPFC projections to the NAcS, but not to the basolateral amygdala, partially reversed suppression of EtOH lever presses on probe testing. CONCLUSIONS These findings demonstrate a key role for the vmPFC in regulating EtOH-SA after punishment, with implications for understanding the neural basis of compulsive drinking in alcohol use disorder.
Collapse
Affiliation(s)
- Lindsay R Halladay
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland; Department of Psychology, Santa Clara University, Santa Clara, California.
| | - Adrina Kocharian
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Patrick T Piantadosi
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland; Center on Compulsive Behaviors, Intramural Research Program, National Institutes of Health, Bethesda, Maryland
| | - Michael E Authement
- Laboratory on Neurobiology of Compulsive Behaviors, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland; Center on Compulsive Behaviors, Intramural Research Program, National Institutes of Health, Bethesda, Maryland
| | - Abby G Lieberman
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Nathen A Spitz
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Kendall Coden
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Lucas R Glover
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Vincent D Costa
- Department of Behavioral Neuroscience, Oregon Health Sciences University, Portland, Oregon
| | - Veronica A Alvarez
- Laboratory on Neurobiology of Compulsive Behaviors, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland; Center on Compulsive Behaviors, Intramural Research Program, National Institutes of Health, Bethesda, Maryland
| | - Andrew Holmes
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
31
|
Zhou Y, Liang Y, Kreek MJ. mTORC1 pathway is involved in the kappa opioid receptor activation-induced increase in excessive alcohol drinking in mice. Pharmacol Biochem Behav 2020; 195:172954. [PMID: 32470351 DOI: 10.1016/j.pbb.2020.172954] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 05/22/2020] [Accepted: 05/25/2020] [Indexed: 12/18/2022]
Abstract
KOP-r agonist U50,488H produces strong aversion and anxiety/depression-like behaviors that enhance alcohol intake and promote alcohol seeking and relapse-like drinking in rodents. Mammalian target of rapamycin complex 1 (mTORC1) pathway in mouse striatum is highly involved in excessive alcohol intake and seeking, and in the U50,488H-induced conditioned place aversion. Therefore, we hypothesized that KOP-r activation increases alcohol consumption through the mTORC1 activation. This study focuses on: (1) how chronic excessive alcohol drinking (4-day drinking-in-the-dark paradigm followed by 3-week chronic intermittent access drinking paradigm [two-bottle choice, 24-h access every other day]) affected nuclear transcript levels of the mTORC1 pathway genes in mouse nucleus accumbens shell (NAcs), using transcriptome-wide RNA sequencing analysis; and (2) whether selective mTORC1 inhibitor rapamycin could alter excessive alcohol drinking and prevent U50,488H-promoted alcohol intake. Thirteen nuclear transcripts of mTORC1 pathway genes showed significant up-regulation in the NAcs, with two genes down-regulated, after excessive alcohol drinking, suggesting the mTORC1 pathway was profoundly disrupted. Single administration of rapamycin decreased alcohol drinking in a dose-dependent manner. U50,488H increased alcohol drinking, and pretreatment with rapamycin, at a dose lower than effective doses, blocked the U50,488H-promoted alcohol intake in a dose-dependent manner, indicating a mTORC1-mediated mechanism. Our results provide supportive and direct evidence relevant to the transcriptional profiling of the critical mTORC1 genes in mouse NAc shell: with functional and pharmacological effects of rapamycin, altered nuclear transcripts in the mTORC1 signaling pathway after excessive alcohol drinking may contribute to increased alcohol intake triggered by KOP-r activation.
Collapse
Affiliation(s)
- Yan Zhou
- Laboratory of the Biology of Addictive Diseases, The Rockefeller University, NY, USA.
| | - Yupu Liang
- Research Bioinformatics, CCTS, The Rockefeller University, NY, USA
| | - Mary Jeanne Kreek
- Laboratory of the Biology of Addictive Diseases, The Rockefeller University, NY, USA
| |
Collapse
|
32
|
Karkhanis AN, Al-Hasani R. Dynorphin and its role in alcohol use disorder. Brain Res 2020; 1735:146742. [PMID: 32114059 DOI: 10.1016/j.brainres.2020.146742] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 02/04/2020] [Accepted: 02/25/2020] [Indexed: 02/07/2023]
Abstract
The dynorphin / kappa opioid receptor (KOR) system has been implicated in many aspects that influence neuropsychiatric disorders. Namely, this system modulates neural circuits that primarily regulate reward seeking, motivation processing, stress responsivity, and pain sensitivity, thus affecting the development of substance and alcohol use disorder (AUD). The effects of this system are often bidirectional and depend on projection targets. To date, a majority of the studies focusing on this system have examined the KOR function using agonists and antagonists. Indeed, there are studies that have examined prodynorphin and dynorphin levels by measuring mRNA and tissue content levels; however, static levels of the neuropeptide and its precursor do not explain complete and online function of the peptide as would be explained by measuring dynorphin transmission in real time. New and exciting methods using optogenetics, chemogenetics, genetic sensors, fast scan cyclic voltammetry are now being developed to detect various neuropeptides with a focus on opioid peptides, including dynorphin. In this review we discuss studies that examine dynorphin projections in areas involved in AUD, its functional involvement in AUD and vulnerability to develop AUD at various ages. Moreover, we discuss dynorphin's role in promoting AUD by dysregulation motivation circuits and how advancements in opioid peptide detection will further our understanding.
Collapse
Affiliation(s)
- Anushree N Karkhanis
- Department of Psychology, Developmental Exposure Alcohol Research Center, Center for Developmental and Behavioral Neuroscience, Binghamton University - SUNY, 4400 Vestal Parkway East, Binghamton, NY 13902, USA.
| | - Ream Al-Hasani
- Department of Pharmaceutical and Administrative Sciences, St. Louis College of Pharmacy, Department of Anesthesiology Washington University in St. Louis, Center for Clinical Pharmacology, Washington University School of Medicine & St. Louis College of Pharmacy 660 S.Euclid, Box 8054, St. Louis, MO 63110, USA.
| |
Collapse
|
33
|
Morisot N, Phamluong K, Ehinger Y, Berger AL, Moffat JJ, Ron D. mTORC1 in the orbitofrontal cortex promotes habitual alcohol seeking. eLife 2019; 8:51333. [PMID: 31820733 PMCID: PMC6959998 DOI: 10.7554/elife.51333] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Accepted: 12/09/2019] [Indexed: 12/12/2022] Open
Abstract
The mechanistic target of rapamycin complex 1 (mTORC1) plays an important role in dendritic translation and in learning and memory. We previously showed that heavy alcohol use activates mTORC1 in the orbitofrontal cortex (OFC) of rodents (Laguesse et al., 2017a). Here, we set out to determine the consequences of alcohol-dependent mTORC1 activation in the OFC. We found that inhibition of mTORC1 activity in the OFC attenuates alcohol seeking and restores sensitivity to outcome devaluation in rats that habitually seek alcohol. In contrast, habitual responding for sucrose was unaltered by mTORC1 inhibition, suggesting that mTORC1’s role in habitual behavior is specific to alcohol. We further show that inhibition of GluN2B in the OFC attenuates alcohol-dependent mTORC1 activation, alcohol seeking and habitual responding for alcohol. Together, these data suggest that the GluN2B/mTORC1 axis in the OFC drives alcohol seeking and habit.
Collapse
Affiliation(s)
- Nadege Morisot
- Department of Neurology, University of California, San Francisco, San Francisco, United States
| | - Khanhky Phamluong
- Department of Neurology, University of California, San Francisco, San Francisco, United States
| | - Yann Ehinger
- Department of Neurology, University of California, San Francisco, San Francisco, United States
| | - Anthony L Berger
- Department of Neurology, University of California, San Francisco, San Francisco, United States
| | - Jeffrey J Moffat
- Department of Neurology, University of California, San Francisco, San Francisco, United States
| | - Dorit Ron
- Department of Neurology, University of California, San Francisco, San Francisco, United States
| |
Collapse
|
34
|
Sex and Individual Differences in Alcohol Intake Are Associated with Differences in Ketamine Self-Administration Behaviors and Nucleus Accumbens Dendritic Spine Density. eNeuro 2019; 6:ENEURO.0221-19.2019. [PMID: 31740575 PMCID: PMC6893233 DOI: 10.1523/eneuro.0221-19.2019] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 10/14/2019] [Accepted: 10/22/2019] [Indexed: 01/03/2023] Open
Abstract
Clinical and preclinical studies have shown that ketamine, an NMDA receptor antagonist, has promising therapeutic value for the treatment of alcohol use disorder (AUD). However, the maintenance of remission will ultimately require repeated infusions of ketamine, which may lead to abuse potential and may hinder its therapeutic benefits. It is therefore crucial to assess the effects of repeated treatments with ketamine on alcohol intake. Accordingly, this study aimed to examine in both sexes how individual differences in alcohol intake alter ketamine self-administration and how ketamine self-administration will alter subsequent alcohol-drinking behaviors. Male and female rats intermittently drank alcohol or water for 10 weeks and were divided into high- or low-alcohol intake groups prior to ketamine self-administration. Rats self-administered ketamine under fixed and progressive ratio schedules of reinforcement from week 4 to 7, and the incubation of ketamine craving was examined from week 8 to 10. To investigate structural plasticity in a brain region involved in reward, nucleus accumbens dendritic spine morphology was examined. Our results show that high alcohol intake in male rats attenuated ketamine self-administration, whereas in female rats high alcohol intake enhanced motivation to self-administer ketamine. Ketamine reduced alcohol intake in high-alcohol male rats but increased it in low-alcohol female rats. Incubation of ketamine craving developed in all groups except low-alcohol females. Three weeks of abstinence from ketamine was associated with increased mushroom spines in all groups except the high-alcohol male group. Overall, these data suggest that ketamine as a treatment for AUD may benefit male subjects, but not female subjects, and warrants further investigation before use as a therapeutic agent.
Collapse
|
35
|
Altered Actin Filament Dynamics in the Drosophila Mushroom Bodies Lead to Fast Acquisition of Alcohol Consumption Preference. J Neurosci 2019; 39:8877-8884. [PMID: 31558618 DOI: 10.1523/jneurosci.0973-19.2019] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 08/22/2019] [Accepted: 09/13/2019] [Indexed: 01/12/2023] Open
Abstract
Alcohol use is highly prevalent in the United States and across the world, and every year millions of people suffer from alcohol use disorders (AUDs). Although the genetic contribution to developing AUDs is estimated to be 50-60%, many of the underlying molecular mechanisms remain unclear. Previous studies from our laboratory revealed that Drosophila melanogaster lacking RhoGAP18B and Ras Suppressor 1 (Rsu1) display reduced sensitivity to ethanol-induced sedation. Both Rsu1 and RhoGAP18B are negative regulators of the small Rho-family GTPase, Rac1, a modulator of actin dynamics. Here we investigate the role of Rac1 and its downstream target, the actin-severing protein cofilin, in alcohol consumption preference. We show that these two regulators of actin dynamics can alter male experience-dependent alcohol preference in a bidirectional manner: expressing either activated Rac1 or dominant-negative cofilin in the mushroom bodies (MBs) abolishes experience-dependent alcohol preference. Conversely, dominant-negative Rac1 or activated cofilin MB expression lead to faster acquisition of alcohol preference. Our data show that Rac1 and cofilin activity are key to determining the rate of acquisition of alcohol preference, revealing a critical role of actin dynamics regulation in the development of voluntary self-administration in Drosophila SIGNIFICANCE STATEMENT The risks for developing an alcohol use disorder (AUD) are strongly determined by genetic factors. Understanding the genes and molecular mechanisms that contribute to that risk is therefore a necessary first step for the development of targeted therapeutic intervention. Here we show that regulators of actin cytoskeleton dynamics can bidirectionally determine the acquisition rate of alcohol self-administration, highlighting this process as a key mechanism contributing to the risk of AUD development.
Collapse
|
36
|
Ben Hamida S, Laguesse S, Morisot N, Park JH, Phuamluong K, Berger AL, Park KD, Ron D. Mammalian target of rapamycin complex 1 and its downstream effector collapsin response mediator protein-2 drive reinstatement of alcohol reward seeking. Addict Biol 2019; 24:908-920. [PMID: 30022576 DOI: 10.1111/adb.12653] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Revised: 05/09/2018] [Accepted: 05/22/2018] [Indexed: 01/01/2023]
Abstract
Alcohol use disorder is a chronic relapsing disease. Maintaining abstinence represents a major challenge for alcohol-dependent patients. Yet the molecular underpinnings of alcohol relapse remain poorly understood. In the present study, we investigated the potential role of the mammalian target of rapamycin complex 1 (mTORC1) in relapse to alcohol-seeking behavior by using the reinstatement of a previously extinguished alcohol conditioned place preference (CPP) response as a surrogate relapse paradigm. We found that mTORC1 is activated in the nucleus accumbens shell following alcohol priming-induced reinstatement of alcohol place preference. We further report that the selective mTORC1 inhibitor, rapamycin, abolishes reinstatement of alcohol place preference. Activation of mTORC1 initiates the translation of synaptic proteins, and we observed that reinstatement of alcohol CPP is associated with increased protein levels of one of mTORC1's downstream targets, collapsin response mediator protein-2 (CRMP2), in the nucleus accumbens. Importantly, the level of mTORC1 activation and CRMP2 expression positively correlate with the CPP score during reinstatement. Finally, we found that systemic administration of the CRMP2 inhibitor, lacosamide, attenuates alcohol priming-induced reinstatement of CPP. Together, our results reveal that mTORC1 and its downstream target, CRMP2, contribute to mechanisms underlying reinstatement of alcohol reward seeking. Our results could have important implications for the treatment of relapse to alcohol use and position the Food and Drug Administration approved drugs, rapamycin and lacosamide, for the treatment of alcohol use disorder.
Collapse
Affiliation(s)
- Sami Ben Hamida
- Department of Neurology; University of California; San Francisco CA USA
| | - Sophie Laguesse
- Department of Neurology; University of California; San Francisco CA USA
| | - Nadege Morisot
- Department of Neurology; University of California; San Francisco CA USA
| | - Jong-Hyun Park
- Convergence Research Center for Diagnosis, Treatment and Care System of Dementia; Korea Institute of Science and Technology (KIST); Republic of Korea
- Division of Bio-Medical Science &Technology, KIST School; Korea University of Science and Technology; Republic of Korea
| | | | - Anthony L. Berger
- Department of Neurology; University of California; San Francisco CA USA
| | - Ki Duk Park
- Convergence Research Center for Diagnosis, Treatment and Care System of Dementia; Korea Institute of Science and Technology (KIST); Republic of Korea
- Division of Bio-Medical Science &Technology, KIST School; Korea University of Science and Technology; Republic of Korea
| | - Dorit Ron
- Department of Neurology; University of California; San Francisco CA USA
| |
Collapse
|
37
|
Indirect Medium Spiny Neurons in the Dorsomedial Striatum Regulate Ethanol-Containing Conditioned Reward Seeking. J Neurosci 2019; 39:7206-7217. [PMID: 31315945 DOI: 10.1523/jneurosci.0876-19.2019] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 07/01/2019] [Accepted: 07/03/2019] [Indexed: 11/21/2022] Open
Abstract
Adenosine 2A receptor (A2AR)-containing indirect medium spiny neurons (iMSNs) in the dorsomedial striatum (DMS) contribute to reward-seeking behaviors. However, those roles for ethanol-seeking behaviors remain unknown. To investigate ethanol-seeking behaviors, we used an ethanol-containing reward (10% ethanol and 10% sucrose solution; 10E10S). Upon conditioning with 10E10S, mice that initially only preferred 10% sucrose, not 10E10S, showed a stronger preference for 10E10S. Then, we investigated whether the manipulation of the DMS-external globus pallidus (GPe) iMSNs circuit alters the ethanol-containing reward (10E10S) seeking behaviors using the combination of pharmacologic and optogenetic approaches. DMS A2AR activation dampened operant conditioning-induced ethanol-containing reward, whereas A2AR antagonist abolished the effects of the A2AR agonist and restored ethanol-containing reward-seeking. Moreover, pre-ethanol exposure potentiated the A2AR-dependent reward-seeking. Interestingly, mice exhibiting ethanol-containing reward-seeking showed the reduction of the DMS iMSNs activity, suggesting that disinhibiting iMSNs decreases reward-seeking behaviors. In addition, we found that A2AR activation reversed iMSNs neural activity in the DMS. Similarly, optogenetic stimulation of the DMS-GPe iMSNs reduced ethanol-containing reward-seeking, whereas optogenetic inhibition of the DMS-GPe iMSNs reversed this change. Together, our study demonstrates that DMS A2AR and iMSNs regulate ethanol-containing reward-seeking behaviors.SIGNIFICANCE STATEMENT Our findings highlight the mechanisms of how operant conditioning develops the preference of ethanol-containing conditioned reward. Mice exhibiting ethanol-containing reward-seeking showed a reduction of the indirect medium spiny neuronal activity in the dorsomedial striatum. Pharmacological activation of adenosine A2A receptor (A2AR) or optogenetic activation of indirect medium spiny neurons dampened operant conditioned ethanol-containing reward-seeking, whereas inhibiting this neuronal activity restored ethanol-containing reward-seeking. Furthermore, repeated intermittent ethanol exposure potentiated A2AR-dependent reward-seeking. Therefore, our finding suggests that A2AR-containing indirect medium spiny neuronal activation reduces ethanol-containing reward-seeking, which may provide a potential therapeutic target for alcohol use disorder.
Collapse
|
38
|
Abstract
Although historically research has focused on transcription as the central governor of protein expression, protein translation is now increasingly being recognized as a major factor for determining protein levels within cells. The central nervous system relies on efficient updating of the protein landscape. Thus, coordinated regulation of mRNA localization, initiation, or termination of translation is essential for proper brain function. In particular, dendritic protein synthesis plays a key role in synaptic plasticity underlying learning and memory as well as cognitive processes. Increasing evidence suggests that impaired mRNA translation is a common feature found in numerous psychiatric disorders. In this review, we describe how malfunction of translation contributes to development of psychiatric diseases, including schizophrenia, major depression, bipolar disorder, and addiction.
Collapse
Affiliation(s)
- Sophie Laguesse
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA.,GIGA-Neurosciences, GIGA-Stem Cells, University of Liège, Liège, Belgium
| | - Dorit Ron
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
39
|
Parkhurst SJ, Adhikari P, Navarrete JS, Legendre A, Manansala M, Wolf FW. Perineurial Barrier Glia Physically Respond to Alcohol in an Akap200-Dependent Manner to Promote Tolerance. Cell Rep 2019; 22:1647-1656. [PMID: 29444420 PMCID: PMC5831198 DOI: 10.1016/j.celrep.2018.01.049] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2017] [Revised: 12/04/2017] [Accepted: 01/16/2018] [Indexed: 12/22/2022] Open
Abstract
Ethanol is the most common drug of abuse. It exerts its behavioral effects by acting on widespread neural circuits; however, its impact on glial cells is less understood. We show that Drosophila perineurial glia are critical for ethanol tolerance, a simple form of behavioral plasticity. The perineurial glia form the continuous outer cellular layer of the blood-brain barrier and are the interface between the brain and the circulation. Ethanol tolerance development requires the A kinase anchoring protein Akap200 specifically in perineurial glia. Akap200 tightly coordinates protein kinase A, actin, and calcium signaling at the membrane to control tolerance. Furthermore, ethanol causes a structural remodeling of the actin cytoskeleton and perineurial membrane topology in an Akap200-dependent manner, without disrupting classical barrier functions. Our findings reveal an active molecular signaling process in the cells at the blood-brain interface that permits a form of behavioral plasticity induced by ethanol.
Collapse
Affiliation(s)
- Sarah J Parkhurst
- Quantitative and Systems Biology Graduate Program, University of California, Merced, CA 95343, USA
| | - Pratik Adhikari
- Quantitative and Systems Biology Graduate Program, University of California, Merced, CA 95343, USA
| | - Jovana S Navarrete
- Molecular Cell Biology, School of Natural Sciences, University of California, Merced, CA 95343, USA
| | - Arièle Legendre
- Molecular Cell Biology, School of Natural Sciences, University of California, Merced, CA 95343, USA
| | - Miguel Manansala
- Molecular Cell Biology, School of Natural Sciences, University of California, Merced, CA 95343, USA
| | - Fred W Wolf
- Quantitative and Systems Biology Graduate Program, University of California, Merced, CA 95343, USA; Molecular Cell Biology, School of Natural Sciences, University of California, Merced, CA 95343, USA.
| |
Collapse
|
40
|
Yin L, Li F, Li J, Yang X, Xie X, Xue L, Li Y, Zhang C. Chronic Intermittent Ethanol Exposure Induces Upregulation of Matrix Metalloproteinase-9 in the Rat Medial Prefrontal Cortex and Hippocampus. Neurochem Res 2019; 44:1593-1601. [PMID: 30915602 DOI: 10.1007/s11064-019-02783-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 03/15/2019] [Accepted: 03/18/2019] [Indexed: 12/17/2022]
Abstract
Matrix metalloproteinase-9 (MMP-9, Gelatinase B), an extracellular-acting Zn2+-dependent endopeptidase, are involved in brain pathologies including ischemia, glioma, and epilepsy. Recent studies suggested that MMP-9 plays an important role in neuronal plasticity, specifically in learning and memory. To determine whether and how MMP-9 plays role in alcohol-related behaviors, male Sprague-Dawley (SD) rats were subjected to chronic intermittent ethanol (CIE) exposure for 4 weeks, following which we collected tissue samples from the hippocampus, medial prefrontal cortex (mPFC), and amygdala at different stages (acute and chronic exposure) during alcohol exposure. Real-time PCR and western blot assays were used to detect changes in the mRNA and protein expression of MMP-9. Our results indicated that both acute and chronic alcohol exposure induced up-regulation of MMP-9 mRNA levels in the hippocampus and mPFC, but not in the amygdala. Furthermore, acute and chronic alcohol exposure up regulated the expression of total MMP-9 and active MMP-9 in these two brain regions. Moreover, the increase of active MMP-9 expression was larger than those in total MMP-9 expression. Immunoprecipitation analyses identified potential MMP-9-interacting proteins, including Itgb1, Src, Eef1a2, tubulin, actin, and histone H2B. These results demonstrate that both acute and CIE exposure induced increases in MMP-9 expression in the mPFC and hippocampus, suggesting that MMP-9 plays a key role in chronic alcohol exposure and dependence.
Collapse
Affiliation(s)
- Litian Yin
- Key Laboratory for Cellular Physiology of Ministry of Education, Department of Physiology, National Key Disciplines, Shanxi Medical University, Taiyuan, 030001, Shanxi, China.
| | - Fengqing Li
- Key Laboratory for Cellular Physiology of Ministry of Education, Department of Physiology, National Key Disciplines, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Jue Li
- School of Clinic, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Xiaorong Yang
- Key Laboratory for Cellular Physiology of Ministry of Education, Department of Physiology, National Key Disciplines, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Xiaoyan Xie
- Key Laboratory for Cellular Physiology of Ministry of Education, Department of Physiology, National Key Disciplines, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Linyuan Xue
- Key Laboratory for Cellular Physiology of Ministry of Education, Department of Physiology, National Key Disciplines, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Yanli Li
- Key Laboratory for Cellular Physiology of Ministry of Education, Department of Physiology, National Key Disciplines, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Ce Zhang
- Key Laboratory for Cellular Physiology of Ministry of Education, Department of Physiology, National Key Disciplines, Shanxi Medical University, Taiyuan, 030001, Shanxi, China.
| |
Collapse
|
41
|
Logrip ML. Molecular tools to elucidate factors regulating alcohol use. Alcohol 2019; 74:3-9. [PMID: 30033149 DOI: 10.1016/j.alcohol.2018.03.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 03/15/2018] [Accepted: 03/15/2018] [Indexed: 12/20/2022]
Abstract
Alcohol use disorder (AUD) is a pervasive societal problem, marked by high levels of alcohol intake and recidivism. Despite these common disease traits, individuals diagnosed with AUD display a range of disordered drinking and alcohol-related behaviors. The diversity in disease presentation, as well as the established polygenic nature of the disorder and complex neurocircuitry, speaks to the variety of neurochemical changes resulting from alcohol intake that may differentially regulate alcohol-related behaviors. Investigations into the molecular adaptations responsible for maladaptive alcohol-related behavioral outcomes require an ever-evolving set of molecular tools to elucidate with increasing precision how alcohol alters behavior through neurochemical changes. This review highlights recent advances in molecular methodology, addressing how incorporation of these cutting-edge techniques not only may enhance current knowledge of the molecular bases of AUD, but also may facilitate identification of improved treatment targets that may be therapeutic in specific subpopulations of AUD individuals.
Collapse
|
42
|
Wang H, Li X, Xie X, Zhao H, Gao Y, Li Y, Xu X, Zhang X, Ke C, Liu J. Promotion of bone cancer pain development by decorin is accompanied by modification of excitatory synaptic molecules in the spinal cord. Mol Pain 2019; 15:1744806919864253. [PMID: 31258052 PMCID: PMC6659184 DOI: 10.1177/1744806919864253] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2018] [Revised: 06/16/2019] [Accepted: 06/18/2019] [Indexed: 01/25/2023] Open
Abstract
Bone cancer pain is refractory to currently available clinical treatment owing to its complicated underlying mechanisms. Studies found that extracellular matrix molecules can participate in the regulation of chronic pain. Decorin is one of the most abundant extracellular matrix molecules, and the present study evaluated the effect of decorin on the development of bone cancer pain. We found that decorin was upregulated in the L4–L6 spinal dorsal horn of the bone cancer pain rats. Spinal microinjection of a decorin-targeting RNAi lentivirus alleviated bone cancer pain-induced mechanical allodynia and reduced the expression of pGluR1-Ser831 in the bone cancer pain rats. Meanwhile, decorin knockdown impaired the excitatory synaptogenesis in cultured neurons and prevented the clustering and insertion of pGluR1-Ser831 into postsynaptic membranes. Taken together, the results of our study suggested that decorin contributes to the development of bone cancer pain possibly by regulating the activity of excitatory synaptic molecules in the spinal cord. Our findings provide a better understanding of the function of decorin as a possible therapeutic target for alleviating bone cancer pain.
Collapse
Affiliation(s)
- Huan Wang
- Department of Anesthesiology, Institute of Anesthesiology &
Pain, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Xiaohui Li
- Department of Anesthesiology, Institute of Anesthesiology &
Pain, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Xianqiao Xie
- Department of Anesthesiology, Institute of Anesthesiology &
Pain, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Haiwen Zhao
- Department of Anesthesiology, Institute of Anesthesiology &
Pain, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Yan Gao
- Department of PET Center, Taihe Hospital, Hubei University of
Medicine, Shiyan, Hubei, China
| | - Yang Li
- Department of Anesthesiology, Institute of Anesthesiology &
Pain, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Xueqin Xu
- Department of Anesthesiology, Institute of Anesthesiology &
Pain, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Xiaofei Zhang
- Department of Anesthesiology, Institute of Anesthesiology &
Pain, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Changbin Ke
- Department of Anesthesiology, Institute of Anesthesiology &
Pain, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Juying Liu
- Department of Anesthesiology, Institute of Anesthesiology &
Pain, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| |
Collapse
|
43
|
Lovinger DM, Abrahao KP. Synaptic plasticity mechanisms common to learning and alcohol use disorder. ACTA ACUST UNITED AC 2018; 25:425-434. [PMID: 30115764 PMCID: PMC6097767 DOI: 10.1101/lm.046722.117] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 06/21/2018] [Indexed: 11/24/2022]
Abstract
Alcohol use disorders include drinking problems that span a range from binge drinking to alcohol abuse and dependence. Plastic changes in synaptic efficacy, such as long-term depression and long-term potentiation are widely recognized as mechanisms involved in learning and memory, responses to drugs of abuse, and addiction. In this review, we focus on the effects of chronic ethanol (EtOH) exposure on the induction of synaptic plasticity in different brain regions. We also review findings indicating that synaptic plasticity occurs in vivo during EtOH exposure, with a focus on ex vivo electrophysiological indices of plasticity. Evidence for effects of EtOH-induced or altered synaptic plasticity on learning and memory and EtOH-related behaviors is also reviewed. As this review indicates, there is much work needed to provide more information about the molecular, cellular, circuit, and behavioral consequences of EtOH interactions with synaptic plasticity mechanisms.
Collapse
Affiliation(s)
- David M Lovinger
- Laboratory for Integrative Neuroscience, Division of Intramural Clinical and Biological Research, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland 20892, USA
| | - Karina P Abrahao
- Laboratory for Integrative Neuroscience, Division of Intramural Clinical and Biological Research, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland 20892, USA
| |
Collapse
|
44
|
Laguesse S, Morisot N, Phamluong K, Sakhai SA, Ron D. mTORC2 in the dorsomedial striatum of mice contributes to alcohol-dependent F-Actin polymerization, structural modifications, and consumption. Neuropsychopharmacology 2018; 43:1539-1547. [PMID: 29497165 PMCID: PMC5983552 DOI: 10.1038/s41386-018-0012-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 12/22/2017] [Accepted: 01/05/2018] [Indexed: 01/21/2023]
Abstract
Actin is highly enriched at dendritic spines, and actin remodeling plays an essential role in structural plasticity. The mammalian target of rapamycin complex 2 (mTORC2) is a regulator of actin polymerization. Here, we report that alcohol consumption increases F-actin content in the dorsomedial striatum (DMS) of mice, thereby altering dendritic spine morphology in a mechanism that requires mTORC2. Specifically, we found that excessive alcohol consumption increases mTORC2 activity in the DMS, and that knockdown of Rictor, an essential component of mTORC2 signaling, reduces actin polymerization, and attenuates the alcohol-dependent alterations in spine head size and the number of mushroom spines. Finally, we show that knockdown of Rictor in the DMS reduces alcohol consumption, whereas intra-DMS infusion of the mTORC2 activator, A-443654, increases alcohol intake. Together, these results suggest that mTORC2 in the DMS facilitates the formation of F-actin, which in turn induces changes in spine structure to promote and/or maintain excessive alcohol intake.
Collapse
Affiliation(s)
- Sophie Laguesse
- 0000 0001 2297 6811grid.266102.1Department of Neurology, University of California, San Francisco, San Francisco, CA 94143 USA ,Present Address: N.M. Brains On-line LLC, South San Francisco, CA USA
| | - Nadege Morisot
- 0000 0001 2297 6811grid.266102.1Department of Neurology, University of California, San Francisco, San Francisco, CA 94143 USA ,0000 0001 0805 7253grid.4861.bPresent Address: S.L. GIGA-Neurosciences, University of Liège, Liège, Belgium
| | - Khanhky Phamluong
- 0000 0001 2297 6811grid.266102.1Department of Neurology, University of California, San Francisco, San Francisco, CA 94143 USA
| | - Samuel A. Sakhai
- 0000 0001 2297 6811grid.266102.1Department of Neurology, University of California, San Francisco, San Francisco, CA 94143 USA
| | - Dorit Ron
- Department of Neurology, University of California, San Francisco, San Francisco, CA, 94143, USA.
| |
Collapse
|
45
|
Varodayan FP, Sidhu H, Kreifeldt M, Roberto M, Contet C. Morphological and functional evidence of increased excitatory signaling in the prelimbic cortex during ethanol withdrawal. Neuropharmacology 2018; 133:470-480. [PMID: 29471053 PMCID: PMC5865397 DOI: 10.1016/j.neuropharm.2018.02.014] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 01/26/2018] [Accepted: 02/16/2018] [Indexed: 02/06/2023]
Abstract
Excessive alcohol consumption in humans induces deficits in decision making and emotional processing, which indicates a dysfunction of the prefrontal cortex (PFC). The present study aimed to determine the impact of chronic intermittent ethanol (CIE) inhalation on mouse medial PFC pyramidal neurons. Data were collected 6-8 days into withdrawal from 7 weeks of CIE exposure, a time point when mice exhibit behavioral symptoms of withdrawal. We found that spine maturity in prelimbic (PL) layer 2/3 neurons was increased, while dendritic spines in PL layer 5 neurons or infralimbic (IL) neurons were not affected. Corroborating these morphological observations, CIE enhanced glutamatergic transmission in PL layer 2/3 pyramidal neurons, but not IL layer 2/3 neurons. Contrary to our predictions, these cellular alterations were associated with improved, rather than impaired, performance in reversal learning and strategy switching tasks in the Barnes maze at an earlier stage of chronic ethanol exposure (5-7 days withdrawal from 3 to 4 weeks of CIE), which could result from the anxiety-like behavior associated with ethanol withdrawal. Altogether, this study adds to a growing body of literature indicating that glutamatergic activity in the PFC is upregulated following chronic ethanol exposure, and identifies PL layer 2/3 pyramidal neurons as a sensitive target of synaptic remodeling. It also indicates that the Barnes maze is not suitable to detect deficits in cognitive flexibility in CIE-withdrawn mice.
Collapse
Affiliation(s)
| | - Harpreet Sidhu
- The Scripps Research Institute, Department of Neuroscience, La Jolla, CA, USA
| | - Max Kreifeldt
- The Scripps Research Institute, Department of Neuroscience, La Jolla, CA, USA
| | - Marisa Roberto
- The Scripps Research Institute, Department of Neuroscience, La Jolla, CA, USA
| | - Candice Contet
- The Scripps Research Institute, Department of Neuroscience, La Jolla, CA, USA.
| |
Collapse
|
46
|
Targeting the intracellular signaling "STOP" and "GO" pathways for the treatment of alcohol use disorders. Psychopharmacology (Berl) 2018; 235:1727-1743. [PMID: 29654346 PMCID: PMC5949137 DOI: 10.1007/s00213-018-4882-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 03/12/2018] [Indexed: 12/12/2022]
Abstract
In recent years, research has identified the molecular and neural substrates underlying the transition of moderate "social" consumption of alcohol to the characteristic alcohol use disorder (AUD) phenotypes including excessive and compulsive alcohol use which we define in the review as the GO signaling pathways. In addition, growing evidence points to the existence of molecular mechanisms that keep alcohol consumption in check and that confer resilience for the development of AUD which we define herein as the STOP signaling pathways. In this review, we focus on examples of the GO and the STOP intracellular signaling pathways and discuss our current knowledge of how manipulations of these pathways may be used for the treatment of AUD.
Collapse
|
47
|
mTORC and ProSAPiP1: How Alcohol Changes Synapses of Reward Circuitry. Neuron 2017; 96:6-8. [PMID: 28957677 DOI: 10.1016/j.neuron.2017.09.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Alcohol addiction is characterized by broad and persistent changes in brain function, but the underlying neural adaptations remain largely unknown. In this issue of Neuron, Laguesse et al. (2017) describe a neural mechanism through which long-term alcohol exposure induces structural and synaptic adaptations that promote excessive alcohol use.
Collapse
|