1
|
Ganguly K, Adhikary K, Acharjee A, Acharjee P, Trigun SK, Mutlaq AS, Ashique S, Yasmin S, Alshahrani AM, Ansari MY. Biological significance and pathophysiological role of Matrix Metalloproteinases in the Central Nervous System. Int J Biol Macromol 2024; 280:135967. [PMID: 39322129 DOI: 10.1016/j.ijbiomac.2024.135967] [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: 06/22/2024] [Revised: 09/21/2024] [Accepted: 09/21/2024] [Indexed: 09/27/2024]
Abstract
Matrix Metalloproteinases (MMPs), which are endopeptidase reliant on zinc, are low in embryonic tissues but increases in response to a variety of physiological stimulus and pathological stresses. Neuro-glial cells, endothelial cells, fibroblasts, and leucocytes secrete MMPs, which cleave extracellular matrix proteins in a time-dependent manner. MMPs affect synaptic plasticity and the development of short-term memory by controlling the size, shape, and excitatory synapses' function through the lateral diffusion of receptors. In addition, MMPs influence the Extracellular Matrix proteins in the Peri-Neuronal Net at the Neuro-glial interface, which aids in the establishment of long-term memory. Through modulating neuronal, and glial cells migration, differentiation, Neurogenesis, and survival, MMPs impact brain development in mammals. In adult brains, MMPs play a beneficial role in physiological plasticity, which includes learning, memory consolidation, social interaction, and complex behaviors, by proteolytically altering a wide variety of factors, including growth factors, cytokines, receptors, DNA repair enzymes, and matrix proteins. Additionally, stress, depression, addiction, hepatic encephalopathy, and stroke may all have negative effects on MMPs. In addition to their role in glioblastoma development, MMPs influence neurological diseases such as epilepsy, schizophrenia, autism spectrum disorder, brain damage, pain, neurodegeneration, and Alzheimer's and Parkinson's. To help shed light on the potential of MMPs as a therapeutic target for neurodegenerative diseases, this review summarizes their regulation, mode of action, and participation in brain physiological plasticity and pathological damage. Finally, by employing different MMP-based nanotools and inhibitors, MMPs may also be utilized to map the anatomical and functional connectome of the brain, analyze its secretome, and treat neurodegenerative illnesses.
Collapse
Affiliation(s)
- Krishnendu Ganguly
- Department of Medical Lab Technology, Paramedical College Durgapur, Helen Keller Sarani, Durgapur 713212, West Bengal, India.
| | - Krishnendu Adhikary
- Department of Medical Lab Technology, Paramedical College Durgapur, Helen Keller Sarani, Durgapur 713212, West Bengal, India.
| | - Arup Acharjee
- Molecular Omics Laboratory, Department of Zoology, University of Allahabad, Allahabad, Uttar Pradesh, India.
| | - Papia Acharjee
- Biochemistry Section, Department of Zoology, Banaras Hindu University, Varanasi 221005, Uttar Pradesh, India.
| | - Surendra Kumar Trigun
- Biochemistry Section, Department of Zoology, Banaras Hindu University, Varanasi 221005, Uttar Pradesh, India.
| | | | - Sumel Ashique
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India; Department of Pharmaceutics, Bengal College of Pharmaceutical Sciences & Research, Durgapur 713212, West Bengal, India.
| | - Sabina Yasmin
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia.
| | - Asma M Alshahrani
- Department of Clinical Pharmacy, Faculty of Pharmacy, King Khalid University, Abha, Saudi Arabia; Department of Clinical Pharmacy, Shaqra University, Saudi Arabia.
| | - Mohammad Yousuf Ansari
- MM college of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, Haryana 133207, India.
| |
Collapse
|
2
|
Kaczmarek KT, Protokowicz K, Kaczmarek L. Matrix metalloproteinase-9: A magic drug target in neuropsychiatry? J Neurochem 2024; 168:1842-1853. [PMID: 37791997 DOI: 10.1111/jnc.15976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 09/12/2023] [Accepted: 09/15/2023] [Indexed: 10/05/2023]
Abstract
Neuropsychiatric conditions represent a major medical and societal challenge. The etiology of these conditions is very complex and combines genetic and environmental factors. The latter, for example, excessive maternal or early postnatal inflammation, as well as various forms of psychotrauma, often act as triggers leading to mental illness after a prolonged latent period (sometimes years). Matrix metalloproteinase-9 (MMP-9) is an extracellularly and extrasynaptic operating protease that is markedly activated in response to the aforementioned environmental insults. MMP-9 has also been shown to play a pivotal role in the plasticity of excitatory synapses, which, in its aberrant form, has repeatedly been implicated in the etiology of mental illness. In this conceptual review, we evaluate the experimental and clinical evidence supporting the claim that MMP-9 is uniquely positioned to be considered a drug target for ameliorating the adverse effects of environmental insults on the development of a variety of neuropsychiatric conditions, such as schizophrenia, bipolar disorder, major depression, autism spectrum disorders, addiction, and epilepsy. We also identify specific challenges and bottlenecks hampering the translation of knowledge on MMP-9 into new clinical treatments for the conditions above and suggest ways to overcome these barriers.
Collapse
|
3
|
Valeri J, Stiplosek C, O'Donovan SM, Sinclair D, Grant KA, Bollavarapu R, Platt DM, Stockmeier CA, Gisabella B, Pantazopoulos H. Extracellular matrix abnormalities in the hippocampus of subjects with substance use disorder. Transl Psychiatry 2024; 14:115. [PMID: 38402197 PMCID: PMC10894211 DOI: 10.1038/s41398-024-02833-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 02/07/2024] [Accepted: 02/13/2024] [Indexed: 02/26/2024] Open
Abstract
Contextual triggers are significant factors contributing to relapse in substance use disorders (SUD). Emerging evidence points to a critical role of extracellular matrix (ECM) molecules as mediators of reward memories. Chondroitin sulfate proteoglycans (CSPGs) are a subset of ECM molecules that form perineuronal nets (PNN) around inhibitory neurons. PNNs restrict synaptic connections and help maintain synapses. Rodent models suggest that modulation of PNNs may strengthen contextual reward memories in SUD. However, there is currently a lack of information regarding PNNs in the hippocampus of people with SUD as well as how comorbidity with major depressive disorder (MDD) may affect PNNs. We used postmortem hippocampal tissues from cohorts of human and nonhuman primates with or without chronic alcohol use to test the hypothesis that PNNs are increased in subjects with SUD. We used histochemical labeling and quantitative microscopy to examine PNNs, and qRT-PCR to examine gene expression for ECM molecules, synaptic markers and related markers. We identified increased densities of PNNs and CSPG-labeled glial cells in SUD, coinciding with decreased expression of the ECM protease matrix metalloproteinase 9 (Mmp9), and increased expression for the excitatory synaptic marker vesicle associated membrane protein 2 (Vamp2). Similar increases in PNNs were observed in monkeys with chronic alcohol self-administration. Subjects with MDD displayed changes opposite to SUD, and subjects with SUD and comorbid MDD had minimal changes in any of the outcome measures examined. Our findings demonstrate that PNNs are increased in SUD, possibly contributing to stabilizing contextual reward memories as suggested by preclinical studies. Our results also point to a previously unsuspected role for CSPG expression in glial cells in SUD. Evidence for increased hippocampal PNNs in SUD suggests that targeting PNNs to weaken contextual reward memories is a promising therapeutic approach for SUD, however comorbidity with MDD is a significant consideration.
Collapse
Affiliation(s)
- Jake Valeri
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, USA
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS, USA
| | - Charlotte Stiplosek
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, USA
| | | | - David Sinclair
- Department of Neuroscience, University of Toledo, Toledo, OH, USA
| | | | - Ratna Bollavarapu
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, USA
| | - Donna M Platt
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, USA
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS, USA
| | - Craig A Stockmeier
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, USA
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS, USA
| | - Barbara Gisabella
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, USA
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS, USA
| | - Harry Pantazopoulos
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, USA.
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS, USA.
| |
Collapse
|
4
|
Valeri J, Stiplosek C, O’Donovan SM, Sinclair D, Grant K, Bollavarapu R, Platt DM, Stockmeier CA, Gisabella B, Pantazopoulos H. Extracellular Matrix Abnormalities in the Hippocampus of Subjects with Substance Use Disorder. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2023.09.07.23295222. [PMID: 37732207 PMCID: PMC10508799 DOI: 10.1101/2023.09.07.23295222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
Contextual triggers are significant factors contributing to relapse in substance use disorders (SUD). Emerging evidence points to a critical role of extracellular matrix (ECM) molecules as mediators of reward memories. Chondroitin sulfate proteoglycans (CSPGs) are a subset of ECM molecules that form perineuronal nets (PNN) around inhibitory neurons. PNNs restrict synaptic connections and help maintain synapses. Rodent models suggest that modulation of PNNs may strengthen contextual reward memories in SUD. However, there is currently a lack of information regarding PNNs in the hippocampus of people with SUD as well as how comorbidity with major depressive disorder (MDD) may affect PNNs. We used postmortem hippocampal tissues from cohorts of human and nonhuman primates with or without chronic alcohol use to test the hypothesis that PNNs are increased in subjects with SUD. We used histochemical labeling and quantitative microscopy to examine PNNs, and qRT-PCR to examine gene expression for ECM molecules, synaptic markers and related markers. We identified increased densities of PNNs and CSPG-labeled glial cells in SUD, coinciding with decreased expression of the ECM protease matrix metalloproteinase 9 (Mmp9), and increased expression for the excitatory synaptic marker vesicle associated membrane protein 2 (Vamp2). Similar increases in PNNs were observed in monkeys with chronic alcohol self-administration. Subjects with MDD displayed changes opposite to SUD, and subjects with SUD and comorbid MDD had minimal changes in any of the outcome measures examined. Our findings demonstrate that PNNs are increased in SUD, possibly contributing to stabilizing contextual reward memories as suggested by preclinical studies. Our results also point to a previously unsuspected role for CSPG expression in glial cells in SUD. Evidence for increased hippocampal PNNs in SUD suggests that targeting PNNs to weaken contextual reward memories is a promising therapeutic approach for SUD, however comorbidity with MDD is a significant consideration.
Collapse
Affiliation(s)
- Jake Valeri
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS
| | - Charlotte Stiplosek
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS
| | | | - David Sinclair
- Department of Neuroscience, University of Toledo, Toledo, OH
| | | | - Ratna Bollavarapu
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS
| | - Donna M. Platt
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS
| | - Craig A. Stockmeier
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS
| | - Barbara Gisabella
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS
| | - Harry Pantazopoulos
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS
| |
Collapse
|
5
|
Valeri J, Gisabella B, Pantazopoulos H. Dynamic regulation of the extracellular matrix in reward memory processes: a question of time. Front Cell Neurosci 2023; 17:1208974. [PMID: 37396928 PMCID: PMC10311570 DOI: 10.3389/fncel.2023.1208974] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 05/30/2023] [Indexed: 07/04/2023] Open
Abstract
Substance use disorders are a global health problem with increasing prevalence resulting in significant socioeconomic burden and increased mortality. Converging lines of evidence point to a critical role of brain extracellular matrix (ECM) molecules in the pathophysiology of substance use disorders. An increasing number of preclinical studies highlight the ECM as a promising target for development of novel cessation pharmacotherapies. The brain ECM is dynamically regulated during learning and memory processes, thus the time course of ECM alterations in substance use disorders is a critical factor that may impact interpretation of the current studies and development of pharmacological therapies. This review highlights the evidence for the involvement of ECM molecules in reward learning, including drug reward and natural reward such as food, as well as evidence regarding the pathophysiological state of the brain's ECM in substance use disorders and metabolic disorders. We focus on the information regarding time-course and substance specific changes in ECM molecules and how this information can be leveraged for the development of therapeutic strategies.
Collapse
Affiliation(s)
- Jake Valeri
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, United States
- Graduate Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS, United States
| | - Barbara Gisabella
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, United States
- Graduate Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS, United States
| | - Harry Pantazopoulos
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, United States
- Graduate Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS, United States
| |
Collapse
|
6
|
Lepreux G, Shinn GE, Wei G, Suko A, Concepcion G, Sirohi S, Soon Go B, Bruchas MR, Walker BM. Recapitulating phenotypes of alcohol dependence via overexpression of Oprk1 in the ventral tegmental area of non-dependent TH::Cre rats. Neuropharmacology 2023; 228:109457. [PMID: 36764577 PMCID: PMC10034863 DOI: 10.1016/j.neuropharm.2023.109457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/31/2023] [Accepted: 02/07/2023] [Indexed: 02/11/2023]
Abstract
The dynorphin (DYN)/kappa-opioid receptor (KOR) system is involved in dysphoria and negative emotional states. Dysregulation of KOR function promotes maladaptive behavioral regulation during withdrawal associated with alcohol dependence. Mesolimbic dopaminergic (DA) projections from the ventral tegmental area (VTA) innervate the extended amygdala circuitry and presynaptic KORs attenuate DA in these regions leading to an excessive alcohol consumption and negative affective-like behavior, whereas mesocortical KOR-regulated DA projections have been implicated in executive function and decision-making. Thus, the neuroadaptations occurring in DYN/KOR systems are important aspects to consider for the development of personalized therapeutic solutions. Herein, we study the contribution of the VTA DA neuron Oprk1 (KOR gene) in excessive alcohol consumption, negative emotional state, and executive function. To do so, Oprk1 mRNA expression and KOR function were characterized to confirm alcohol dependence-induced dysregulation in the VTA. Then, a transgenic Cre-Lox rat model (male and female TH::Cre rats) was used to allow for conditional and inducible overexpression of Oprk1 in VTA DA neurons. The effect of this overexpression was evaluated on operant alcohol self-administration, negative emotional states, and executive function. We found that VTA Oprk1 overexpression recapitulates some phenotypes of alcohol dependence including escalated alcohol self-administration and depressive-like behavior. However, working memory performance was not impacted following VTA Oprk1 overexpression in TH::Cre rats. This supports the hypothesis that dysregulated KOR signaling within the mesolimbic DA system is an important contributor to symptoms of alcohol dependence and shows that understanding Oprk1-mediated contributions to alcohol use disorder (AUD) should be an important future goal.
Collapse
Affiliation(s)
- Gaetan Lepreux
- Laboratory of Alcoholism and Addictions Neuroscience, Department of Psychiatry and Behavioral Neurosciences, Tampa, FL, USA
| | - Grace E Shinn
- College of Veterinary Medicine, Western University of Health Sciences, Pomona, CA, USA
| | - Gengze Wei
- Laboratory of Alcoholism and Addictions Neuroscience, Department of Psychiatry and Behavioral Neurosciences, Tampa, FL, USA
| | - Azra Suko
- Department of Anesthesiology and Pain Medicine, Seattle, WA, USA
| | - George Concepcion
- Laboratory of Alcoholism and Addictions Neuroscience, Department of Psychiatry and Behavioral Neurosciences, Tampa, FL, USA
| | - Sunil Sirohi
- Division of Basic Pharmaceutical Sciences, College of Pharmacy, Xavier University of Louisiana, New Orleans, LA, USA
| | - Bok Soon Go
- Department of Life Sciences, Korea University, Seoul, 02841, South Korea
| | - Michael R Bruchas
- Department of Anesthesiology and Pain Medicine, Seattle, WA, USA; Department of Pharmacology, Seattle, WA, USA; Center for the Neurobiology of Addiction, Pain and Emotion, University of Washington, Seattle, WA, USA
| | - Brendan M Walker
- Laboratory of Alcoholism and Addictions Neuroscience, Department of Psychiatry and Behavioral Neurosciences, Tampa, FL, USA; Department of Molecular Medicine, Tampa, FL, USA; USF Health Neuroscience Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, USA.
| |
Collapse
|
7
|
Yin LT, Feng RR, Xie XY, Yang XR, Yang ZF, Hu JJ, Wu SF, Zhang C. Matrix metalloproteinase-9 overexpression in the hippocampus reduces alcohol-induced conditioned-place preference by regulating synaptic plasticity in mice. Behav Brain Res 2023; 442:114330. [PMID: 36746309 DOI: 10.1016/j.bbr.2023.114330] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 02/02/2023] [Accepted: 02/03/2023] [Indexed: 02/07/2023]
Abstract
Extracellular matrix proteins appear to be necessary for the synaptic plasticity that underlies addiction memory. In the brain, matrix metalloproteinases (MMPs), especially matrix metalloproteinase-9 (MMP-9), have been recently implicated in processes involving alcohol reward and memory. Here, we showed for the first time, the positive effects of MMP-9 on alcohol-induced conditioned place preference (CPP) behavior and hippocampal neuron plasticity in C57BL/6 mice. Using recombinant adeno-associated viruses to overexpress MMP-9 in the hippocampus, we investigated the NMDAR, PSD-95, and cellular cytoskeleton proteins F-actin/G-actin in the modulation of alcohol reward behavior in mice exposed to CPP. We found that hippocampal infusions of MMP-9 decreased alcohol-induced place preference suggesting a reduction in alcohol reward. Western blot analysis demonstrated that protein expression of NMDA receptors (GluN1, GluN2A and GluN2B) in the hippocampus of alcohol-exposed mice were higher than that of the saline group. Further, the expression of these proteins was decreased in MMP-9 overexpressing mice. MMP-9 also regulated the ratio of F-actin/G-actin (dendritic spines cytoskeleton proteins), which might be the key mediator for behavioral changes in mice. Consequently, our results highlight new evidence that MMP-9 may play an important role in the molecular mechanism underlying alcohol reward and preference.
Collapse
Affiliation(s)
- Li-Tian Yin
- Key Laboratory for Cellular Physiology, Ministry of Education, Key Laboratory of Cellular Physiology in Shanxi Province, Department of Physiology, Shanxi Medical University, Taiyuan, Shanxi 030001, China.
| | - Rui-Rui Feng
- Key Laboratory for Cellular Physiology, Ministry of Education, Key Laboratory of Cellular Physiology in Shanxi Province, Department of Physiology, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Xiao-Yan Xie
- Key Laboratory for Cellular Physiology, Ministry of Education, Key Laboratory of Cellular Physiology in Shanxi Province, Department of Physiology, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Xiao-Rong Yang
- Key Laboratory for Cellular Physiology, Ministry of Education, Key Laboratory of Cellular Physiology in Shanxi Province, Department of Physiology, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Zhuan-Fang Yang
- Key Laboratory for Cellular Physiology, Ministry of Education, Key Laboratory of Cellular Physiology in Shanxi Province, Department of Physiology, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Jia-Jia Hu
- Key Laboratory for Cellular Physiology, Ministry of Education, Key Laboratory of Cellular Physiology in Shanxi Province, Department of Physiology, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Shu-Fen Wu
- Key Laboratory for Cellular Physiology, Ministry of Education, Key Laboratory of Cellular Physiology in Shanxi Province, Department of Physiology, Shanxi Medical University, Taiyuan, Shanxi 030001, China; Department of Pediatrics, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Ce Zhang
- Key Laboratory for Cellular Physiology, Ministry of Education, Key Laboratory of Cellular Physiology in Shanxi Province, Department of Physiology, Shanxi Medical University, Taiyuan, Shanxi 030001, China.
| |
Collapse
|
8
|
de Guglielmo G, Simpson S, Kimbrough A, Conlisk D, Baker R, Cantor M, Kallupi M, George O. Voluntary and forced exposure to ethanol vapor produces similar escalation of alcohol drinking but differential recruitment of brain regions related to stress, habit, and reward in male rats. Neuropharmacology 2023; 222:109309. [PMID: 36334765 PMCID: PMC10022477 DOI: 10.1016/j.neuropharm.2022.109309] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 10/13/2022] [Accepted: 10/24/2022] [Indexed: 11/06/2022]
Abstract
A major limitation of the most widely used current animal models of alcohol dependence is that they use forced exposure to ethanol including ethanol-containing liquid diet and chronic intermittent ethanol (CIE) vapor to produce clinically relevant blood alcohol levels (BAL) and addiction-like behaviors. We recently developed a novel animal model of voluntary induction of alcohol dependence using ethanol vapor self-administration (EVSA). However, it is unknown whether EVSA leads to an escalation of alcohol drinking per se, and whether such escalation is associated with neuroadaptations in brain regions related to stress, reward, and habit. To address these issues, we compared the levels of alcohol drinking during withdrawal between rats passively exposed to alcohol (CIE) or voluntarily exposed to EVSA and measured the number of Fos+ neurons during acute withdrawal (16 h) in key brain regions important for stress, reward, and habit-related processes. CIE and EVSA rats exhibited similar BAL and similar escalation of alcohol drinking and motivation for alcohol during withdrawal. Acute withdrawal from EVSA and CIE recruited a similar number of Fos+ neurons in the Central Amygdala (CeA), however, acute withdrawal from EVSA recruited a higher number of Fos+ neurons in every other brain region analyzed compared to acute withdrawal from CIE. In summary, while the behavioral measures of alcohol dependence between the voluntary (EVSA) and passive (CIE) model were similar, the recruitment of neuronal ensembles during acute withdrawal was very different. The EVSA model may be particularly useful to unveil the neuronal networks and pharmacology responsible for the voluntary induction and maintenance of alcohol dependence and may improve translational studies by providing preclinical researchers with an animal model that highlights the volitional aspects of alcohol use disorder.
Collapse
Affiliation(s)
| | - Sierra Simpson
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Adam Kimbrough
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, 47906, USA
| | - Dana Conlisk
- Univ. Bordeaux, INSERM, Neurocenter Magendie, Psychobiology of Drug Addiction Group, U1215, F-33000, Bordeaux, France
| | - Robert Baker
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Maxwell Cantor
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Marsida Kallupi
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Olivier George
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
9
|
Alteration of Ethanol Reward by Prior Mephedrone Exposure: The Role of Age and Matrix Metalloproteinase-9 (MMP-9). Int J Mol Sci 2022; 23:ijms23042122. [PMID: 35216236 PMCID: PMC8877998 DOI: 10.3390/ijms23042122] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 02/08/2022] [Accepted: 02/11/2022] [Indexed: 02/04/2023] Open
Abstract
Mephedrone, a synthetic cathinone, is widely abused by adolescents and young adults. The aim of this study was to determine: (i) whether prior mephedrone exposure would alter ethanol reward and (ii) whether age and matrix metalloproteinase-9 (MMP-9) are important in this regard. In our research, male Wistar rats at postnatal day 30 (PND30) received mephedrone at the dose of 10 mg/kg, i.p., 3 times a day for 7 days. To clarify the role of MMP-9 in the mephedrone effects, one mephedrone-treated group received minocycline, as an MMP-9 antagonist. Animals were then assigned to conditioned place preference (CPP) procedure at PND38 (adolescent) or at PND69 (adult). After the CPP test (PND48/79), expression of dopamine D1 receptors (D1R), Cav1.2 (a subtype of L-type calcium channels), and MMP-9 was quantified in the rat ventral striatum (vSTR). The influence of mephedrone administration on the N-methyl-D-aspartate glutamate receptors (NMDAR) subunits (GluN1, GluN2A, and GluN2B) was then assessed in the vSTR of adult rats (only). These results indicate that, in contrast with adolescent rats, adult rats with prior mephedrone administration appear to be more sensitive to the ethanol effect in the CPP test under the drug-free state. The mephedrone effect in adult rats was associated with upregulation of D1R, NMDAR/GluN2B, MMP-9, and Cav1.2 signaling. MMP-9 appears to contribute to these changes in proteins expression because minocycline pretreatment blocked mephedrone-evoked sensitivity to ethanol reward. Thus, our results suggest that prior mephedrone exposure differentially alters ethanol reward in adolescent and adult rats.
Collapse
|
10
|
Maiya R, Pomrenze MB, Tran T, Tiwari GR, Beckham A, Paul MT, Mayfield RD, Messing RO. Differential regulation of alcohol consumption and reward by the transcriptional cofactor LMO4. Mol Psychiatry 2021; 26:2175-2186. [PMID: 32144357 PMCID: PMC7558853 DOI: 10.1038/s41380-020-0706-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 02/20/2020] [Accepted: 02/25/2020] [Indexed: 01/04/2023]
Abstract
Repeated alcohol exposure leads to changes in gene expression that are thought to underlie the transition from moderate to excessive drinking. However, the mechanisms by which these changes are integrated into a maladaptive response that leads to alcohol dependence are not well understood. One mechanism could involve the recruitment of transcriptional co-regulators that bind and modulate the activity of transcription factors. Our results indicate that the transcriptional regulator LMO4 is one such candidate regulator. Lmo4-deficient mice (Lmo4gt/+) consumed significantly more and showed enhanced preference for alcohol in a 24 h intermittent access drinking procedure. shRNA-mediated knockdown of Lmo4 in the nucleus accumbens enhanced alcohol consumption, whereas knockdown in the basolateral amygdala (BLA) decreased alcohol consumption and reduced conditioned place preference for alcohol. To ascertain the molecular mechanisms that underlie these contrasting phenotypes, we carried out unbiased transcriptome profiling of these two brain regions in wild type and Lmo4gt/+ mice. Our results revealed that the transcriptional targets of LMO4 are vastly different between the two brain regions, which may explain the divergent phenotypes observed upon Lmo4 knockdown. Bioinformatic analyses revealed that Oprk1 and genes related to the extracellular matrix (ECM) are important transcriptional targets of LMO4 in the BLA. Chromatin immunoprecipitation revealed that LMO4 bound Oprk1 promoter elements. Consistent with these results, disruption of the ECM or infusion of norbinaltorphimine, a selective kappa opioid receptor antagonist, in the BLA reduced alcohol consumption. Hence our results indicate that an LMO4-regulated transcriptional network regulates alcohol consumption in the BLA.
Collapse
Affiliation(s)
- Rajani Maiya
- Department of Neuroscience, The University of Texas at Austin, Austin, TX, 78712, USA. .,Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX, 78712, USA. .,Department of Neurology, The University of Texas at Austin, Austin, TX, 78712, USA.
| | - Matthew B. Pomrenze
- Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712, USA,Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, USA
| | - Thi Tran
- Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712, USA
| | - Gayatri R. Tiwari
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, USA
| | - Andrea Beckham
- Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712, USA
| | - Madison T. Paul
- Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712, USA
| | - R. Dayne Mayfield
- Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712, USA,Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, USA
| | - Robert O. Messing
- Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712, USA,Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, USA,Department of Neurology, The University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
11
|
Gisabella B, Babu J, Valeri J, Rexrode L, Pantazopoulos H. Sleep and Memory Consolidation Dysfunction in Psychiatric Disorders: Evidence for the Involvement of Extracellular Matrix Molecules. Front Neurosci 2021; 15:646678. [PMID: 34054408 PMCID: PMC8160443 DOI: 10.3389/fnins.2021.646678] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Accepted: 04/22/2021] [Indexed: 12/13/2022] Open
Abstract
Sleep disturbances and memory dysfunction are key characteristics across psychiatric disorders. Recent advances have revealed insight into the role of sleep in memory consolidation, pointing to key overlap between memory consolidation processes and structural and molecular abnormalities in psychiatric disorders. Ongoing research regarding the molecular mechanisms involved in memory consolidation has the potential to identify therapeutic targets for memory dysfunction in psychiatric disorders and aging. Recent evidence from our group and others points to extracellular matrix molecules, including chondroitin sulfate proteoglycans and their endogenous proteases, as molecules that may underlie synaptic dysfunction in psychiatric disorders and memory consolidation during sleep. These molecules may provide a therapeutic targets for decreasing strength of reward memories in addiction and traumatic memories in PTSD, as well as restoring deficits in memory consolidation in schizophrenia and aging. We review the evidence for sleep and memory consolidation dysfunction in psychiatric disorders and aging in the context of current evidence pointing to the involvement of extracellular matrix molecules in these processes.
Collapse
Affiliation(s)
| | | | | | | | - Harry Pantazopoulos
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, MS, United States
| |
Collapse
|
12
|
Hitzemann R, Phillips TJ, Lockwood DR, Darakjian P, Searles RP. Phenotypic and gene expression features associated with variation in chronic ethanol consumption in heterogeneous stock collaborative cross mice. Genomics 2020; 112:4516-4524. [PMID: 32771621 PMCID: PMC7749084 DOI: 10.1016/j.ygeno.2020.08.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 07/22/2020] [Accepted: 08/04/2020] [Indexed: 12/12/2022]
Abstract
Of the more than 100 studies that have examined relationships between excessive ethanol consumption and the brain transcriptome, few rodent studies have examined chronic consumption. Heterogeneous stock collaborative cross mice freely consumed ethanol vs. water for 3 months. Transcriptional differences were examined for the central nucleus of the amygdala, a brain region known to impact ethanol preference. Early preference was modestly predictive of final preference and there was significant escalation of preference in females only. Genes significantly correlated with female preference were enriched in annotations for the primary cilium and extracellular matrix. A single module in the gene co-expression network was enriched in genes with an astrocyte annotation. The key hub node was the master regulator, orthodenticle homeobox 2 (Otx2). These data support an important role for the extracellular matrix, primary cilium and astrocytes in ethanol preference and consumption differences among individual female mice of a genetically diverse population.
Collapse
Affiliation(s)
- Robert Hitzemann
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, USA; Portland Alcohol Research Center, Oregon Health & Science University, Portland, OR 97239, USA.
| | - Tamara J Phillips
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, USA; Portland Alcohol Research Center, Oregon Health & Science University, Portland, OR 97239, USA; Veterans Affairs Portland Health Care System, Portland, OR 97239, USA.
| | - Denesa R Lockwood
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, USA; Portland Alcohol Research Center, Oregon Health & Science University, Portland, OR 97239, USA.
| | - Priscila Darakjian
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, USA; Portland Alcohol Research Center, Oregon Health & Science University, Portland, OR 97239, USA.
| | - Robert P Searles
- Portland Alcohol Research Center, Oregon Health & Science University, Portland, OR 97239, USA; Integrated Genomics Laboratory, Oregon Health & Science University, Portland, OR 97239, USA.
| |
Collapse
|
13
|
Chondroitin sulfate proteoglycan-5 forms perisynaptic matrix assemblies in the adult rat cortex. Cell Signal 2020; 74:109710. [PMID: 32653642 DOI: 10.1016/j.cellsig.2020.109710] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 07/07/2020] [Indexed: 12/30/2022]
Abstract
Composition of the brain extracellular matrix changes in time as maturation proceeds. Chondroitin sulfate proteoglycan 5 (CSPG-5), also known as neuroglycan C, has been previously associated to differentiation since it shapes neurite growth and synapse forming. Here, we show that this proteoglycan persists in the postnatal rat brain, and its expression is higher in cortical regions with plastic properties, including hippocampus and the medial prefrontal cortex at the end of the second postnatal week. Progressively accumulating after birth, CSPG-5 typically concentrates around glutamatergic and GABAergic terminals in twelve-week old rat hippocampus. CSPG-5-containing perisynaptic matrix rings often appear at the peripheral margin of perineuronal nets. Electron microscopy and analysis of synaptosomal fraction showed that CSPG-5 accumulates around, and is associated to synapses, respectively. In vitro analyses suggest that neurons, but less so astrocytes, express CSPG-5 in rat primary neocortical cultures, and CSPG-5 produced by transfected neuroblastoma cells appear at endings and contact points of neurites. In human subjects, CSPG-5 expression shifts in brain areas of the default mode network of suicide victims, which may reflect an impact in the pathogenesis of psychiatric diseases or support diagnostic power.
Collapse
|
14
|
The Modulatory Effect of Metformin on Ethanol-Induced Anxiety, Redox Imbalance, and Extracellular Matrix Levels in the Brains of Wistar Rats. J Mol Neurosci 2020; 70:1943-1961. [PMID: 32621100 DOI: 10.1007/s12031-020-01593-w] [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: 03/08/2020] [Accepted: 05/13/2020] [Indexed: 01/14/2023]
Abstract
The study investigated the potential neuroprotective effects of metformin (MET) on alcohol-induced neurotoxicity in adult Wistar rats. The animals were randomized in four groups (n = 10): control, alcohol (ALC), ALC + MET, and MET. ALC (2 g/kg b.w.) and MET (200 mg/kg b.w.) were orally administered for 21 days, once daily. For the ALC + MET group, MET was administered 2 h after ALC treatment. On day 22, the open field test (OFT) and elevated plus maze (EPM) were performed. MET improved global activity and increased the time spent in unprotected open arms, decreased oxidative stress, both in the frontal lobe and in the hippocampus, and increased neuroglobin expression in the frontal cortex. Histopathologically, an increased neurosecretory activity in the frontal cortex in the ALC + MET group was noticed. Thus, our findings suggest that metformin has antioxidant and anxiolytic effects and may partially reverse the neurotoxic effects induced by ethanol.
Collapse
|
15
|
Matrix Metalloproteinase-9 Overexpression Regulates Hippocampal Synaptic Plasticity and Decreases Alcohol Consumption and Preference in Mice. Neurochem Res 2020; 45:1902-1912. [PMID: 32415404 DOI: 10.1007/s11064-020-03053-8] [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: 02/12/2020] [Revised: 04/30/2020] [Accepted: 05/07/2020] [Indexed: 02/06/2023]
Abstract
Brain matrix metalloproteinases (MMPs) have been recently implicated in alcohol addiction; however, the molecular mechanisms remain poorly understood. Matrix metalloproteinase-9 (MMP-9), an extrasynaptic protease, is the best described MMP that is thought to regulate addictive behavior. In the present study, the effect of MMP-9 overexpression on hippocampal neuron plasticity and alcoholic behavior was assessed in spontaneous alcohol drinking mice. Two-bottle choice model showed that the overexpression of MMP-9 in the hippocampus developed by adeno-associated virus (AAV) could decrease alcohol consumption and preference, but did not affect taste preference, which was tested using saccharin or quinine solutions. Dendritic spines number of hippocampal neurons was observed by Golgi staining. Compared with the alcohol treatment group, the density of dendritic spines in the hippocampus of alcohol drinking mice was decreased in alcohol + MMP-9 group. Western blot analysis indicated that GluN1 expression in the hippocampus of alcohol drinking group was lower than that in the control group, while the expression of GluN1 was increased in MMP-9 overexpressing mice. MMP-9 also regulated the depolymerization of actin filaments, which induced behavioral changes in mice. Taken together, overexpression of MMP-9 in the hippocampal neurons of mice resulted in decreased dendritic spine density and F-actin/G-actin ratio, which might be the crucial reason for the significant decrease in alcohol consumption in alcohol drinking mice. MMP-9 might be considered as a novel target studying the molecular mechanism of alcohol drinking.
Collapse
|
16
|
Go BS, Sirohi S, Walker BM. The role of matrix metalloproteinase-9 in negative reinforcement learning and plasticity in alcohol dependence. Addict Biol 2020; 25:e12715. [PMID: 30648329 DOI: 10.1111/adb.12715] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 11/21/2018] [Accepted: 12/17/2018] [Indexed: 01/08/2023]
Abstract
A role for matrix metalloproteinases (MMPs) in plasticity-dependent learning has been established. MMPs degrade the extracellular matrix (ECM) when synaptic reorganization is warranted. Previously, we showed that escalation of alcohol self-administration is a learned plasticity-dependent process that requires an intact MMP system. To identify the MMP subtypes within specific brain regions that are associated with plasticity underlying the negative reinforcing effects of alcohol (as measured by escalated alcohol self-administration) during acute withdrawal in alcohol dependence, male Wistar rats were trained to self-administer alcohol in an operant paradigm, subjected to one month of intermittent alcohol vapor exposure to induce alcohol dependence and then allowed to self-administer alcohol during repeated acute withdrawal self-administration sessions. Subsequently, rat brains were extracted after initial or stable escalated alcohol self-administration phases of acute withdrawal and analyzed by immunoblot to detect MMP-2, -3, and -9 levels in the anterior cingulate cortex (ACC), bed nucleus of the stria terminalis, central amygdala (CeA), hippocampus, and nucleus accumbens (NAc). The results showed that MMP-9 expression in the CeA and NAc of alcohol-dependent rats was increased, however, MMP-9 expression in the ACC was decreased during negative reinforcement learning. Subsequently, the importance of plasticity mediated by MMP-9 in escalated alcohol self-administration during acute withdrawal was functionally assessed through site-specific intra-CeA MMP-9 inhibition during repeated acute withdrawal self-administration sessions. MMP-9 inhibition prevented acute withdrawal-induced escalation of alcohol self-administration in a manner that was not confounded by locomotor effects or a permanent inability to learn about the negative reinforcing effects of alcohol.
Collapse
Affiliation(s)
- Bok Soon Go
- Laboratory of Alcoholism and Addictions Neuroscience, Department of Psychology, Alcohol and Drug Abuse Research Program, Translational Addictions Research CenterWashington State University Pullman Washington USA
| | - Sunil Sirohi
- Laboratory of Alcoholism and Addictions Neuroscience, Department of Psychology, Alcohol and Drug Abuse Research Program, Translational Addictions Research CenterWashington State University Pullman Washington USA
| | - Brendan M. Walker
- Laboratory of Alcoholism and Addictions Neuroscience, Department of Psychology, Alcohol and Drug Abuse Research Program, Translational Addictions Research CenterWashington State University Pullman Washington USA
| |
Collapse
|
17
|
Beroun A, Mitra S, Michaluk P, Pijet B, Stefaniuk M, Kaczmarek L. MMPs in learning and memory and neuropsychiatric disorders. Cell Mol Life Sci 2019; 76:3207-3228. [PMID: 31172215 PMCID: PMC6647627 DOI: 10.1007/s00018-019-03180-8] [Citation(s) in RCA: 125] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 05/27/2019] [Accepted: 05/29/2019] [Indexed: 12/20/2022]
Abstract
Matrix metalloproteinases (MMPs) are a group of over twenty proteases, operating chiefly extracellularly to cleave components of the extracellular matrix, cell adhesion molecules as well as cytokines and growth factors. By virtue of their expression and activity patterns in animal models and clinical investigations, as well as functional studies with gene knockouts and enzyme inhibitors, MMPs have been demonstrated to play a paramount role in many physiological and pathological processes in the brain. In particular, they have been shown to influence learning and memory processes, as well as major neuropsychiatric disorders such as schizophrenia, various kinds of addiction, epilepsy, fragile X syndrome, and depression. A possible link connecting all those conditions is either physiological or aberrant synaptic plasticity where some MMPs, e.g., MMP-9, have been demonstrated to contribute to the structural and functional reorganization of excitatory synapses that are located on dendritic spines. Another common theme linking the aforementioned pathological conditions is neuroinflammation and MMPs have also been shown to be important mediators of immune responses.
Collapse
Affiliation(s)
- Anna Beroun
- BRAINCITY, Nencki Institute, Pasteura 3, 02-093, Warsaw, Poland
| | | | - Piotr Michaluk
- BRAINCITY, Nencki Institute, Pasteura 3, 02-093, Warsaw, Poland
| | - Barbara Pijet
- BRAINCITY, Nencki Institute, Pasteura 3, 02-093, Warsaw, Poland
| | | | - Leszek Kaczmarek
- BRAINCITY, Nencki Institute, Pasteura 3, 02-093, Warsaw, Poland.
| |
Collapse
|
18
|
Farris SP, Riley BP, Williams RW, Mulligan MK, Miles MF, Lopez MF, Hitzemann R, Iancu OD, Colville A, Walter NAR, Darakjian P, Oberbeck DL, Daunais JB, Zheng CL, Searles RP, McWeeney SK, Grant KA, Mayfield RD. Cross-species molecular dissection across alcohol behavioral domains. Alcohol 2018; 72:19-31. [PMID: 30213503 PMCID: PMC6309876 DOI: 10.1016/j.alcohol.2017.11.036] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Revised: 11/17/2017] [Accepted: 11/28/2017] [Indexed: 12/14/2022]
Abstract
This review summarizes the proceedings of a symposium presented at the "Alcoholism and Stress: A Framework for Future Treatment Strategies" conference held in Volterra, Italy on May 9-12, 2017. Psychiatric diseases, including alcohol-use disorders (AUDs), are influenced through complex interactions of genes, neurobiological pathways, and environmental influences. A better understanding of the common neurobiological mechanisms underlying an AUD necessitates an integrative approach, involving a systematic assessment of diverse species and phenotype measures. As part of the World Congress on Stress and Alcoholism, this symposium provided a detailed account of current strategies to identify mechanisms underlying the development and progression of AUDs. Dr. Sean Farris discussed the integration and organization of transcriptome and postmortem human brain data to identify brain regional- and cell type-specific differences related to excessive alcohol consumption that are conserved across species. Dr. Brien Riley presented the results of a genome-wide association study of DSM-IV alcohol dependence; although replication of genetic associations with alcohol phenotypes in humans remains challenging, model organism studies show that COL6A3, KLF12, and RYR3 affect behavioral responses to ethanol, and provide substantial evidence for their role in human alcohol-related traits. Dr. Rob Williams expanded upon the systematic characterization of extensive genetic-genomic resources for quantifying and clarifying phenotypes across species that are relevant to precision medicine in human disease. The symposium concluded with Dr. Robert Hitzemann's description of transcriptome studies in a mouse model selectively bred for high alcohol ("binge-like") consumption and a non-human primate model of long-term alcohol consumption. Together, the different components of this session provided an overview of systems-based approaches that are pioneering the experimental prioritization and validation of novel genes and gene networks linked with a range of behavioral phenotypes associated with stress and AUDs.
Collapse
Affiliation(s)
- Sean P Farris
- University of Texas at Austin, Austin, TX, United States
| | - Brien P Riley
- Virginia Commonwealth University, Richmond, VA, United States
| | - Robert W Williams
- University of Tennessee Health Science Center, Memphis, TN, United States
| | - Megan K Mulligan
- University of Tennessee Health Science Center, Memphis, TN, United States
| | - Michael F Miles
- University of Tennessee Health Science Center, Memphis, TN, United States
| | - Marcelo F Lopez
- University of Tennessee Health Science Center, Memphis, TN, United States
| | - Robert Hitzemann
- Oregon Health and Science University, Portland, OR, United States
| | - Ovidiu D Iancu
- Oregon Health and Science University, Portland, OR, United States
| | | | | | | | | | - James B Daunais
- Wake Forest School of Medicine, Winston-Salem, NC, United States
| | | | - Robert P Searles
- Oregon Health and Science University, Portland, OR, United States
| | | | - Kathleen A Grant
- Oregon Health and Science University, Portland, OR, United States
| | | |
Collapse
|
19
|
Erikson CM, Wei G, Walker BM. Maladaptive behavioral regulation in alcohol dependence: Role of kappa-opioid receptors in the bed nucleus of the stria terminalis. Neuropharmacology 2018; 140:162-173. [PMID: 30075159 DOI: 10.1016/j.neuropharm.2018.07.034] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 07/27/2018] [Accepted: 07/28/2018] [Indexed: 12/15/2022]
Abstract
There is an important emerging role for the endogenous opioid dynorphin (DYN) and the kappa-opioid receptor (KOR) in the treatment of alcohol dependence. Evidence suggests that the DYN/KOR system in the bed nucleus of the stria terminalis (BNST) contributes to maladaptive behavioral regulation during withdrawal in alcohol dependence. The current experiments were designed to assess dysregulation of the BNST DYN/KOR system by evaluating alcohol dependence-induced changes in DYN/KOR gene expression (Pdyn and Oprk1, respectively), and the sensitivity of alcohol self-administration, negative affective-like behavior and physiological withdrawal to intra-BNST KOR antagonism during acute withdrawal. Wistar rats trained to self-administer alcohol, or not trained, were subjected to an alcohol dependence induction procedure (14 h alcohol vapor/10 h air) or air-exposure. BNST micropunches from air- and vapor-exposed animals were analyzed using RT-qPCR to quantify dependence-induced changes in Pdyn and Oprk1 mRNA expression. In addition, vapor- and air-exposed groups received an intra-BNST infusion of a KOR antagonist or vehicle prior to measurement of alcohol self-administration. A separate cohort of vapor-exposed rats was assessed for physiological withdrawal and negative affective-like behavior signs following intra-BNST KOR antagonism. During acute withdrawal, following alcohol dependence induction, there was an upregulation in Oprk1 mRNA expression in alcohol self-administering animals, but not non-alcohol self-administering animals, that confirmed dysregulation of the KOR/DYN system within the BNST. Furthermore, intra-BNST KOR antagonism attenuated escalated alcohol self-administration and negative affective-like behavior during acute withdrawal without reliably impacting physiological symptoms of withdrawal. The results confirm KOR system dysregulation in the BNST in alcohol dependence, illustrating the therapeutic potential of targeting the KOR to treat alcohol dependence.
Collapse
Affiliation(s)
- Chloe M Erikson
- Laboratory of Alcoholism and Addictions Neuroscience, Department of Psychology, Alcohol and Drug Abuse Research Program, Translational Addiction Research Center, Washington State University, Pullman, WA, 99164-4820, USA
| | - Gengze Wei
- Laboratory of Alcoholism and Addictions Neuroscience, Department of Psychology, Alcohol and Drug Abuse Research Program, Translational Addiction Research Center, Washington State University, Pullman, WA, 99164-4820, USA
| | - Brendan M Walker
- Laboratory of Alcoholism and Addictions Neuroscience, Department of Psychology, Alcohol and Drug Abuse Research Program, Translational Addiction Research Center, Washington State University, Pullman, WA, 99164-4820, USA.
| |
Collapse
|
20
|
Lasek AW, Chen H, Chen WY. Releasing Addiction Memories Trapped in Perineuronal Nets. Trends Genet 2017; 34:197-208. [PMID: 29289347 DOI: 10.1016/j.tig.2017.12.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Revised: 11/28/2017] [Accepted: 12/06/2017] [Indexed: 12/20/2022]
Abstract
Drug addiction can be conceptualized at a basic level as maladaptive learning and memory. Addictive substances elicit changes in brain circuitry involved in reward, cognition, and emotional state, leading to the formation and persistence of strong drug-associated memories that lead to craving and relapse. Recently, perineuronal nets (PNNs), extracellular matrix (ECM) structures surrounding neurons, have emerged as regulators of learning, memory, and addiction behaviors. PNNs do not merely provide structural support to neurons but are dynamically remodeled in an experience-dependent manner by metalloproteinases. They function in various brain regions through constituent proteins such as brevican that are implicated in neural plasticity. Understanding the function of PNN components in memory processes may lead to new therapeutic approaches to treating addiction.
Collapse
Affiliation(s)
- Amy W Lasek
- Center for Alcohol Research in Epigenetics and Department of Psychiatry, University of Illinois at Chicago, Chicago, IL 60612, USA.
| | - Hu Chen
- Center for Alcohol Research in Epigenetics and Department of Psychiatry, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Wei-Yang Chen
- Center for Alcohol Research in Epigenetics and Department of Psychiatry, University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
21
|
Bobadilla AC, Heinsbroek JA, Gipson CD, Griffin WC, Fowler CD, Kenny PJ, Kalivas PW. Corticostriatal plasticity, neuronal ensembles, and regulation of drug-seeking behavior. PROGRESS IN BRAIN RESEARCH 2017; 235:93-112. [PMID: 29054293 DOI: 10.1016/bs.pbr.2017.07.013] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The idea that interconnected neuronal ensembles code for specific behaviors has been around for decades; however, recent technical improvements allow studying these networks and their causal role in initiating and maintaining behavior. In particular, the role of ensembles in drug-seeking behaviors in the context of addiction is being actively investigated. Concurrent with breakthroughs in quantifying ensembles, research has identified a role for synaptic glutamate spillover during relapse. In particular, the transient relapse-associated changes in glutamatergic synapses on accumbens neurons, as well as in adjacent astroglia and extracellular matrix, are key elements of the synaptic plasticity encoded by drug use and the metaplasticity induced by drug-associated cues that precipitate drug-seeking behaviors. Here, we briefly review the recent discoveries related to ensembles in the addiction field and then endeavor to link these discoveries with drug-induced striatal plasticity and cue-induced metaplasticity toward deeper neurobiological understandings of drug seeking.
Collapse
Affiliation(s)
| | | | | | | | | | - Paul J Kenny
- Icahn School of Medicine at Mount Sinai, Icahn, New York, NY, United States
| | - Peter W Kalivas
- Medical University of South Carolina, Charleston, SC, United States.
| |
Collapse
|
22
|
Mason BJ. Emerging pharmacotherapies for alcohol use disorder. Neuropharmacology 2017; 122:244-253. [PMID: 28454983 PMCID: PMC5643030 DOI: 10.1016/j.neuropharm.2017.04.032] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Revised: 04/21/2017] [Accepted: 04/23/2017] [Indexed: 01/29/2023]
Abstract
The identification of different stages within the alcohol use disorder (AUD) cycle that are linked to neurocircuitry changes in pathophysiology associated with the negative emotional states of abstinence has provided a view of medication development for AUD that emphasizes changes in the brain reward and stress systems. Alcohol use disorder can be defined as a chronic relapsing disorder that involves compulsive alcohol seeking and taking, loss of control over alcohol intake, and emergence of a negative emotional state during abstinence. The focus of early medications development was to block the motivation to seek alcohol in the binge/intoxication stage. More recent work has focused on reversing the motivational dysregulations associated with the withdrawal/negative affect and preoccupation/anticipation stages during protracted abstinence. Advances in our understanding of the neurocircuitry and neuropharmacological mechanisms that are involved in the development and maintenance of the withdrawal/negative affect stage using validated animal models have provided viable targets for future medications. Another major advance has been proof-of-concept testing of potential therapeutics and clinical validation of relevant pharmacological targets using human laboratory models of protracted abstinence. This review focuses on future targets for medication development associated with reversal of the loss of reward function and gain in brain stress function that drive negative reinforcement in the withdrawal/negative affect stage of addiction. Basic research has identified novel neurobiological targets associated with the withdrawal/negative affect stage and preoccupation/anticipation stage, with a focus on neuroadaptive changes within the extended amygdala that account for the transition to dependence and vulnerability to relapse. This article is part of the Special Issue entitled "Alcoholism".
Collapse
Affiliation(s)
- Barbara J Mason
- The Pearson Center on Alcoholism and Addiction Research, Department of Neuroscience, The Scripps Research Institute, 10550 North Torrey Pines Road, TPC-5 La Jolla, CA 92037 USA.
| |
Collapse
|
23
|
Stefaniuk M, Beroun A, Lebitko T, Markina O, Leski S, Meyza K, Grzywacz A, Samochowiec J, Samochowiec A, Radwanska K, Kaczmarek L. Matrix Metalloproteinase-9 and Synaptic Plasticity in the Central Amygdala in Control of Alcohol-Seeking Behavior. Biol Psychiatry 2017; 81:907-917. [PMID: 28190519 DOI: 10.1016/j.biopsych.2016.12.026] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 12/01/2016] [Accepted: 12/16/2016] [Indexed: 01/08/2023]
Abstract
BACKGROUND Dysfunction of the glutamatergic system has been implicated in alcohol addiction; however, the molecular underpinnings of this phenomenon are still poorly understood. In the current study we have investigated the possible function of matrix metalloproteinase-9 (MMP-9) in alcohol addiction because this protein has recently emerged as an important regulator of excitatory synaptic plasticity. METHODS For long-term studies of alcohol drinking in mice we used IntelliCages. Dendritic spines were analyzed using Diolistic staining with DiI. Whole-cell patch clamp was used to assess silent synapses. Motivation for alcohol in human subjects was assessed on the basis of a Semi-Structured Assessment for the Genetics of Alcoholism interview. RESULTS Mice devoid of MMP-9 (MMP-9 knockout) drank as much alcohol as wild-type animals; however, they were impaired in alcohol seeking during the motivation test and withdrawal. The deficit could be rescued by overexpression of exogenous MMP-9 in the central nucleus of the amygdala (CeA). Furthermore, the impaired alcohol seeking was associated with structural alterations of dendritic spines in the CeA and, moreover, whole-cell patch clamp analysis of the basal amygdala to CeA projections showed that alcohol consumption and withdrawal were associated with generation of silent synapses. These plastic changes were impaired in MMP-9 knockout mice. Finally, C/T polymorphism of MMP-9 gene at position -1562, which upregulates MMP-9 expression, correlated with increased motivation for alcohol in alcoholics. CONCLUSIONS In aggregate, our results indicate a novel mechanism of alcohol craving that involves MMP-9-dependent synaptic plasticity in CeA.
Collapse
Affiliation(s)
| | - Anna Beroun
- Laboratory of Neurobiology, the Nencki Institute, Warsaw
| | - Tomasz Lebitko
- Laboratory of Neurobiology, the Nencki Institute, Warsaw
| | - Olga Markina
- Laboratory of Neurobiology, the Nencki Institute, Warsaw
| | - Szymon Leski
- Laboratory of Neuroinformatics, the Nencki Institute, Warsaw
| | - Ksenia Meyza
- Laboratory of Emotions Neurobiology, the Nencki Institute, Warsaw
| | - Anna Grzywacz
- Department of Psychiatry, Pomeranian Medical University, University of Szczecin, Szczecin, Poland
| | - Jerzy Samochowiec
- Department of Psychiatry, Pomeranian Medical University, University of Szczecin, Szczecin, Poland
| | - Agnieszka Samochowiec
- Department of Clinical Psychology, Institute of Psychology, University of Szczecin, Szczecin, Poland
| | - Kasia Radwanska
- Laboratory of Molecular Basis of Behavior, the Nencki Institute, Warsaw
| | | |
Collapse
|
24
|
Adkins AE, Hack LM, Bigdeli TB, Williamson VS, McMichael GO, Mamdani M, Edwards A, Aliev F, Chan RF, Bhandari P, Raabe RC, Alaimo JT, Blackwell GG, Moscati AA, Poland RS, Rood B, Patterson DG, Walsh D, Whitfield JB, Zhu G, Montgomery GW, Henders AK, Martin NG, Heath AC, Madden PA, Frank J, Ridinger M, Wodarz N, Soyka M, Zill P, Ising M, Nöthen MM, Kiefer F, Rietschel M, Gelernter J, Sherva R, Koesterer R, Almasy L, Zhao H, Kranzler HR, Farrer LA, Maher BS, Prescott CA, Dick DM, Bacanu SA, Mathies LD, Davies AG, Vladimirov VI, Grotewiel M, Bowers MS, Bettinger JC, Webb BT, Miles MF, Kendler KS, Riley BP. Genomewide Association Study of Alcohol Dependence Identifies Risk Loci Altering Ethanol-Response Behaviors in Model Organisms. Alcohol Clin Exp Res 2017; 41:911-928. [PMID: 28226201 PMCID: PMC5404949 DOI: 10.1111/acer.13362] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Accepted: 02/16/2017] [Indexed: 01/23/2023]
Abstract
BACKGROUND Alcohol dependence (AD) shows evidence for genetic liability, but genes influencing risk remain largely unidentified. METHODS We conducted a genomewide association study in 706 related AD cases and 1,748 unscreened population controls from Ireland. We sought replication in 15,496 samples of European descent. We used model organisms (MOs) to assess the role of orthologous genes in ethanol (EtOH)-response behaviors. We tested 1 primate-specific gene for expression differences in case/control postmortem brain tissue. RESULTS We detected significant association in COL6A3 and suggestive association in 2 previously implicated loci, KLF12 and RYR3. None of these signals are significant in replication. A suggestive signal in the long noncoding RNA LOC339975 is significant in case:control meta-analysis, but not in a population sample. Knockdown of a COL6A3 ortholog in Caenorhabditis elegans reduced EtOH sensitivity. Col6a3 expression correlated with handling-induced convulsions in mice. Loss of function of the KLF12 ortholog in C. elegans impaired development of acute functional tolerance (AFT). Klf12 expression correlated with locomotor activation following EtOH injection in mice. Loss of function of the RYR3 ortholog reduced EtOH sensitivity in C. elegans and rapid tolerance in Drosophila. The ryanodine receptor antagonist dantrolene reduced motivation to self-administer EtOH in rats. Expression of LOC339975 does not differ between cases and controls but is reduced in carriers of the associated rs11726136 allele in nucleus accumbens (NAc). CONCLUSIONS We detect association between AD and COL6A3, KLF12, RYR3, and LOC339975. Despite nonreplication of COL6A3, KLF12, and RYR3 signals, orthologs of these genes influence behavioral response to EtOH in MOs, suggesting potential involvement in human EtOH response and AD liability. The associated LOC339975 allele may influence gene expression in human NAc. Although the functions of long noncoding RNAs are poorly understood, there is mounting evidence implicating these genes in multiple brain functions and disorders.
Collapse
Affiliation(s)
- Amy E. Adkins
- Virginia Commonwealth University Alcohol Research Center, PO Box
980424, Virginia Commonwealth University, Richmond, VA, 23298-0424, USA
- Department of Psychiatry, PO Box 980424, Virginia Commonwealth
University, Richmond, VA, 23298-0424, USA
| | - Laura M. Hack
- Virginia Commonwealth University Alcohol Research Center, PO Box
980424, Virginia Commonwealth University, Richmond, VA, 23298-0424, USA
- Department of Psychiatry, PO Box 980424, Virginia Commonwealth
University, Richmond, VA, 23298-0424, USA
| | - Tim B. Bigdeli
- Department of Psychiatry, PO Box 980424, Virginia Commonwealth
University, Richmond, VA, 23298-0424, USA
| | - Vernell S. Williamson
- Virginia Commonwealth University Alcohol Research Center, PO Box
980424, Virginia Commonwealth University, Richmond, VA, 23298-0424, USA
- Department of Psychiatry, PO Box 980424, Virginia Commonwealth
University, Richmond, VA, 23298-0424, USA
| | - G. Omari McMichael
- Virginia Commonwealth University Alcohol Research Center, PO Box
980424, Virginia Commonwealth University, Richmond, VA, 23298-0424, USA
- Department of Psychiatry, PO Box 980424, Virginia Commonwealth
University, Richmond, VA, 23298-0424, USA
| | - Mohammed Mamdani
- Virginia Commonwealth University Alcohol Research Center, PO Box
980424, Virginia Commonwealth University, Richmond, VA, 23298-0424, USA
- Department of Psychiatry, PO Box 980424, Virginia Commonwealth
University, Richmond, VA, 23298-0424, USA
| | - Alexis Edwards
- Virginia Commonwealth University Alcohol Research Center, PO Box
980424, Virginia Commonwealth University, Richmond, VA, 23298-0424, USA
- Department of Psychiatry, PO Box 980424, Virginia Commonwealth
University, Richmond, VA, 23298-0424, USA
| | - Fazil Aliev
- Virginia Commonwealth University Alcohol Research Center, PO Box
980424, Virginia Commonwealth University, Richmond, VA, 23298-0424, USA
- Department of Psychiatry, PO Box 980424, Virginia Commonwealth
University, Richmond, VA, 23298-0424, USA
| | - Robin F. Chan
- Virginia Commonwealth University Alcohol Research Center, PO Box
980424, Virginia Commonwealth University, Richmond, VA, 23298-0424, USA
- Department of Human & Molecular Genetics, PO Box 980424,
Virginia Commonwealth University, Richmond, VA, 23298-0424, USA
| | - Poonam Bhandari
- Department of Human & Molecular Genetics, PO Box 980424,
Virginia Commonwealth University, Richmond, VA, 23298-0424, USA
| | - Richard C. Raabe
- Department of Pharmacology & Toxicology, PO Box 980424,
Virginia Commonwealth University, Richmond, VA, 23298-0424, USA
| | - Joseph T. Alaimo
- Department of Pharmacology & Toxicology, PO Box 980424,
Virginia Commonwealth University, Richmond, VA, 23298-0424, USA
| | - GinaMari G. Blackwell
- Department of Pharmacology & Toxicology, PO Box 980424,
Virginia Commonwealth University, Richmond, VA, 23298-0424, USA
| | - Arden A. Moscati
- Virginia Commonwealth University Alcohol Research Center, PO Box
980424, Virginia Commonwealth University, Richmond, VA, 23298-0424, USA
- Department of Psychiatry, PO Box 980424, Virginia Commonwealth
University, Richmond, VA, 23298-0424, USA
| | - Ryan S. Poland
- Department of Pharmacology & Toxicology, PO Box 980424,
Virginia Commonwealth University, Richmond, VA, 23298-0424, USA
| | - Benjamin Rood
- Department of Pharmacology & Toxicology, PO Box 980424,
Virginia Commonwealth University, Richmond, VA, 23298-0424, USA
| | - Diana G. Patterson
- Shaftesbury Square Hospital, 116-120 Great Victoria Street, Belfast,
BT2 7BG, United Kingdom
| | - Dermot Walsh
- Health Research Board, 67-72 Lower Mount Street, Dublin 2,
Ireland
| | | | - John B. Whitfield
- Genetic Epidemiology, QIMR Berghofer Medical Research Institute,
Royal Brisbane and Women’s Hospital, 300 Herston Road, Brisbane, QLD 4006,
Australia
| | - Gu Zhu
- Genetic Epidemiology, QIMR Berghofer Medical Research Institute,
Royal Brisbane and Women’s Hospital, 300 Herston Road, Brisbane, QLD 4006,
Australia
| | - Grant W. Montgomery
- Genetic Epidemiology, QIMR Berghofer Medical Research Institute,
Royal Brisbane and Women’s Hospital, 300 Herston Road, Brisbane, QLD 4006,
Australia
| | - Anjali K. Henders
- Genetic Epidemiology, QIMR Berghofer Medical Research Institute,
Royal Brisbane and Women’s Hospital, 300 Herston Road, Brisbane, QLD 4006,
Australia
| | - Nicholas G. Martin
- Genetic Epidemiology, QIMR Berghofer Medical Research Institute,
Royal Brisbane and Women’s Hospital, 300 Herston Road, Brisbane, QLD 4006,
Australia
| | - Andrew C. Heath
- Department of Psychiatry, Washington University School of Medicine,
4560 Clayton Ave., Suite 1000, St. Louis, MO, 63110, USA
| | - Pamela A.F. Madden
- Department of Psychiatry, Washington University School of Medicine,
4560 Clayton Ave., Suite 1000, St. Louis, MO, 63110, USA
| | - Josef Frank
- Department of Genetic Epidemiology in Psychiatry, Central Institute
of Mental Health, Medical Faculty Mannheim/Heidelberg University, J 5, 68159
Mannheim, Germany
| | - Monika Ridinger
- Department of Psychiatry, University Hospital Regensburg,
University of Regensburg, 93042 Regensburg, Germany
| | - Norbert Wodarz
- Department of Psychiatry, University Hospital Regensburg,
University of Regensburg, 93042 Regensburg, Germany
| | - Michael Soyka
- Privatklinik Meiringen, Willigen, 3860 Meiringen, Switzerland
- Department of Psychiatry and Psychotherapy, University of Munich,
Nussbaumstrasse 7, 80336 Munich, Germany
| | - Peter Zill
- Department of Psychiatry and Psychotherapy, University of Munich,
Nussbaumstrasse 7, 80336 Munich, Germany
| | - Marcus Ising
- Department of Molecular Psychology, Max-Planck-Institute of
Psychiatry, Kraepelinstrasse 2–10, 80804 Munich, Germany
| | - Markus M Nöthen
- Department of Genomics, Life & Brain Center, University of
Bonn, Sigmund-Freud-Strasse 25, D-53127 Bonn, Germany
- Department of Institute of Human Genetics, University of Bonn,
Sigmund-Freud-Strasse 25, D-53127 Bonn, Germany
- German Center for Neurodegenerative Diseases (DZNE), University of
Bonn, Sigmund-Freud-Strasse 25, D-53127 Bonn, Germany
| | - Falk Kiefer
- Department of Addictive Behavior and Addiction Medicine, Central
Institute of Mental Health, Medical Faculty Mannheim/Heidelberg University, J 5,
68159 Mannheim, Germany
| | - Marcella Rietschel
- Department of Genetic Epidemiology in Psychiatry, Central Institute
of Mental Health, Medical Faculty Mannheim/Heidelberg University, J 5, 68159
Mannheim, Germany
| | | | - Joel Gelernter
- Department of Psychiatry, Yale University School of Medicine, 333
Cedar Street, New Haven, CT, 06510, USA
- Department of Neurobiology, Yale University School of Medicine, 333
Cedar Street, New Haven, CT, 06510, USA
- Department of Genetics, Yale University School of Medicine, 333
Cedar Street, New Haven, CT, 06510, USA
- Department of Psychiatry, VA CT Healthcare Center, 950 Campbell
Avenue, West Haven, CT, 06516, USA
| | - Richard Sherva
- Department of Medicine (Biomedical Genetics), Boston University
School of Medicine, 72 East Concord Street, Boston, MA, 02118, USA
| | - Ryan Koesterer
- Department of Medicine (Biomedical Genetics), Boston University
School of Medicine, 72 East Concord Street, Boston, MA, 02118, USA
| | - Laura Almasy
- Texas Biomedical Research Institute, Department of Genetics, P.O.
Box 760549, San Antonio, TX, 78245-0549, USA
| | - Hongyu Zhao
- Department of Genetics, Yale University School of Medicine, 333
Cedar Street, New Haven, CT, 06510, USA
- Department of Biostatistics, Yale University School of Medicine,
333 Cedar Street, New Haven, CT, 06510, USA
| | - Henry R. Kranzler
- Department of Psychiatry, University of Pennsylvania Perelman
School of Medicine, Treatment Research Center, 3900 Chestnut Street, Philadelphia,
PA 19104, USA
- VISN 4 MIRECC, Philadelphia VA Medical Center, 3900 Woodland
Avenue, Philadelphia, PA, 19104, USA
| | - Lindsay A. Farrer
- Department of Psychiatry, VA CT Healthcare Center, 950 Campbell
Avenue, West Haven, CT, 06516, USA
- Department of Neurology, Boston University School of Medicine, 72
East Concord Street, Boston, MA, 02118, USA
- Department of Ophthalmology, Boston University School of Medicine,
72 East Concord Street, Boston, MA, 02118, USA
- Department of Genetics and Genomics, Boston University School of
Medicine, 72 East Concord Street, Boston, MA, 02118, USA
- Department of Epidemiology and Biostatistics, Boston University
School of Public Health, 715 Albany Street, Boston, MA, 02118, USA
| | - Brion S. Maher
- Department of Mental Health, Johns Hopkins Bloomberg School of
Public Health, 624 N. Broadway, 8th Floor, Baltimore, MD, 21205, USA
| | - Carol A. Prescott
- Department of Psychology, University of Southern California, SGM
501, 3620 South McClintock Ave., Los Angeles, CA, 90089-1061, USA
| | - Danielle M. Dick
- Virginia Commonwealth University Alcohol Research Center, PO Box
980424, Virginia Commonwealth University, Richmond, VA, 23298-0424, USA
- Department of Psychiatry, PO Box 980424, Virginia Commonwealth
University, Richmond, VA, 23298-0424, USA
- Department of Human & Molecular Genetics, PO Box 980424,
Virginia Commonwealth University, Richmond, VA, 23298-0424, USA
| | - Silviu A. Bacanu
- Virginia Commonwealth University Alcohol Research Center, PO Box
980424, Virginia Commonwealth University, Richmond, VA, 23298-0424, USA
- Department of Psychiatry, PO Box 980424, Virginia Commonwealth
University, Richmond, VA, 23298-0424, USA
| | - Laura D. Mathies
- Department of Pharmacology & Toxicology, PO Box 980424,
Virginia Commonwealth University, Richmond, VA, 23298-0424, USA
| | - Andrew G. Davies
- Virginia Commonwealth University Alcohol Research Center, PO Box
980424, Virginia Commonwealth University, Richmond, VA, 23298-0424, USA
- Department of Pharmacology & Toxicology, PO Box 980424,
Virginia Commonwealth University, Richmond, VA, 23298-0424, USA
| | - Vladimir I. Vladimirov
- Virginia Commonwealth University Alcohol Research Center, PO Box
980424, Virginia Commonwealth University, Richmond, VA, 23298-0424, USA
- Department of Psychiatry, PO Box 980424, Virginia Commonwealth
University, Richmond, VA, 23298-0424, USA
- Lieber Institute for Brain Development, Johns Hopkins University,
855 North Wolfe Street Suite 300, Baltimore, MD, 21205, USA
- Center for Biomarker Research and Personalized Medicine, School of
Pharmacy, PO Box 980533, Virginia Commonwealth University, Richmond, VA 23298-0533,
USA
| | - Mike Grotewiel
- Virginia Commonwealth University Alcohol Research Center, PO Box
980424, Virginia Commonwealth University, Richmond, VA, 23298-0424, USA
- Department of Human & Molecular Genetics, PO Box 980424,
Virginia Commonwealth University, Richmond, VA, 23298-0424, USA
| | - M. Scott Bowers
- Virginia Commonwealth University Alcohol Research Center, PO Box
980424, Virginia Commonwealth University, Richmond, VA, 23298-0424, USA
- Department of Psychiatry, PO Box 980424, Virginia Commonwealth
University, Richmond, VA, 23298-0424, USA
- Department of Pharmacology & Toxicology, PO Box 980424,
Virginia Commonwealth University, Richmond, VA, 23298-0424, USA
| | - Jill C. Bettinger
- Virginia Commonwealth University Alcohol Research Center, PO Box
980424, Virginia Commonwealth University, Richmond, VA, 23298-0424, USA
- Department of Pharmacology & Toxicology, PO Box 980424,
Virginia Commonwealth University, Richmond, VA, 23298-0424, USA
| | - Bradley T. Webb
- Virginia Commonwealth University Alcohol Research Center, PO Box
980424, Virginia Commonwealth University, Richmond, VA, 23298-0424, USA
- Department of Psychiatry, PO Box 980424, Virginia Commonwealth
University, Richmond, VA, 23298-0424, USA
| | - Michael F. Miles
- Virginia Commonwealth University Alcohol Research Center, PO Box
980424, Virginia Commonwealth University, Richmond, VA, 23298-0424, USA
- Department of Human & Molecular Genetics, PO Box 980424,
Virginia Commonwealth University, Richmond, VA, 23298-0424, USA
- Department of Pharmacology & Toxicology, PO Box 980424,
Virginia Commonwealth University, Richmond, VA, 23298-0424, USA
| | - Kenneth S. Kendler
- Virginia Commonwealth University Alcohol Research Center, PO Box
980424, Virginia Commonwealth University, Richmond, VA, 23298-0424, USA
- Department of Psychiatry, PO Box 980424, Virginia Commonwealth
University, Richmond, VA, 23298-0424, USA
- Department of Human & Molecular Genetics, PO Box 980424,
Virginia Commonwealth University, Richmond, VA, 23298-0424, USA
| | - Brien P. Riley
- Virginia Commonwealth University Alcohol Research Center, PO Box
980424, Virginia Commonwealth University, Richmond, VA, 23298-0424, USA
- Department of Psychiatry, PO Box 980424, Virginia Commonwealth
University, Richmond, VA, 23298-0424, USA
- Department of Human & Molecular Genetics, PO Box 980424,
Virginia Commonwealth University, Richmond, VA, 23298-0424, USA
| |
Collapse
|
25
|
Scofield MD, Heinsbroek JA, Gipson CD, Kupchik YM, Spencer S, Smith ACW, Roberts-Wolfe D, Kalivas PW. The Nucleus Accumbens: Mechanisms of Addiction across Drug Classes Reflect the Importance of Glutamate Homeostasis. Pharmacol Rev 2017; 68:816-71. [PMID: 27363441 DOI: 10.1124/pr.116.012484] [Citation(s) in RCA: 379] [Impact Index Per Article: 54.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The nucleus accumbens is a major input structure of the basal ganglia and integrates information from cortical and limbic structures to mediate goal-directed behaviors. Chronic exposure to several classes of drugs of abuse disrupts plasticity in this region, allowing drug-associated cues to engender a pathologic motivation for drug seeking. A number of alterations in glutamatergic transmission occur within the nucleus accumbens after withdrawal from chronic drug exposure. These drug-induced neuroadaptations serve as the molecular basis for relapse vulnerability. In this review, we focus on the role that glutamate signal transduction in the nucleus accumbens plays in addiction-related behaviors. First, we explore the nucleus accumbens, including the cell types and neuronal populations present as well as afferent and efferent connections. Next we discuss rodent models of addiction and assess the viability of these models for testing candidate pharmacotherapies for the prevention of relapse. Then we provide a review of the literature describing how synaptic plasticity in the accumbens is altered after exposure to drugs of abuse and withdrawal and also how pharmacological manipulation of glutamate systems in the accumbens can inhibit drug seeking in the laboratory setting. Finally, we examine results from clinical trials in which pharmacotherapies designed to manipulate glutamate systems have been effective in treating relapse in human patients. Further elucidation of how drugs of abuse alter glutamatergic plasticity within the accumbens will be necessary for the development of new therapeutics for the treatment of addiction across all classes of addictive substances.
Collapse
Affiliation(s)
- M D Scofield
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina (M.D.S., J.A.H., S.S., D.R.-W., P.W.K.); Department of Psychology, Arizona State University, Tempe, Arizona (C.D.G.); Department of Neuroscience, Hebrew University, Jerusalem, Israel (Y.M.K.); and Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, New York (A.C.W.S.)
| | - J A Heinsbroek
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina (M.D.S., J.A.H., S.S., D.R.-W., P.W.K.); Department of Psychology, Arizona State University, Tempe, Arizona (C.D.G.); Department of Neuroscience, Hebrew University, Jerusalem, Israel (Y.M.K.); and Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, New York (A.C.W.S.)
| | - C D Gipson
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina (M.D.S., J.A.H., S.S., D.R.-W., P.W.K.); Department of Psychology, Arizona State University, Tempe, Arizona (C.D.G.); Department of Neuroscience, Hebrew University, Jerusalem, Israel (Y.M.K.); and Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, New York (A.C.W.S.)
| | - Y M Kupchik
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina (M.D.S., J.A.H., S.S., D.R.-W., P.W.K.); Department of Psychology, Arizona State University, Tempe, Arizona (C.D.G.); Department of Neuroscience, Hebrew University, Jerusalem, Israel (Y.M.K.); and Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, New York (A.C.W.S.)
| | - S Spencer
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina (M.D.S., J.A.H., S.S., D.R.-W., P.W.K.); Department of Psychology, Arizona State University, Tempe, Arizona (C.D.G.); Department of Neuroscience, Hebrew University, Jerusalem, Israel (Y.M.K.); and Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, New York (A.C.W.S.)
| | - A C W Smith
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina (M.D.S., J.A.H., S.S., D.R.-W., P.W.K.); Department of Psychology, Arizona State University, Tempe, Arizona (C.D.G.); Department of Neuroscience, Hebrew University, Jerusalem, Israel (Y.M.K.); and Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, New York (A.C.W.S.)
| | - D Roberts-Wolfe
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina (M.D.S., J.A.H., S.S., D.R.-W., P.W.K.); Department of Psychology, Arizona State University, Tempe, Arizona (C.D.G.); Department of Neuroscience, Hebrew University, Jerusalem, Israel (Y.M.K.); and Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, New York (A.C.W.S.)
| | - P W Kalivas
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina (M.D.S., J.A.H., S.S., D.R.-W., P.W.K.); Department of Psychology, Arizona State University, Tempe, Arizona (C.D.G.); Department of Neuroscience, Hebrew University, Jerusalem, Israel (Y.M.K.); and Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, New York (A.C.W.S.)
| |
Collapse
|
26
|
Smith ACW, Scofield MD, Heinsbroek JA, Gipson CD, Neuhofer D, Roberts-Wolfe DJ, Spencer S, Garcia-Keller C, Stankeviciute NM, Smith RJ, Allen NP, Lorang MR, Griffin WC, Boger HA, Kalivas PW. Accumbens nNOS Interneurons Regulate Cocaine Relapse. J Neurosci 2017; 37:742-756. [PMID: 28123012 PMCID: PMC5296777 DOI: 10.1523/jneurosci.2673-16.2016] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 11/16/2016] [Accepted: 11/30/2016] [Indexed: 12/31/2022] Open
Abstract
Relapse to drug use can be initiated by drug-associated cues. The intensity of cue-induced relapse is correlated with the induction of transient synaptic potentiation (t-SP) at glutamatergic synapses on medium spiny neurons (MSNs) in the nucleus accumbens core (NAcore) and requires spillover of glutamate from prefrontal cortical afferents. We used a rodent self-administration/reinstatement model of relapse to show that cue-induced t-SP and reinstated cocaine seeking result from glutamate spillover, initiating a metabotropic glutamate receptor 5 (mGluR5)-dependent increase in nitric oxide (NO) production. Pharmacological stimulation of mGluR5 in NAcore recapitulated cue-induced reinstatement in the absence of drug-associated cues. Using NO-sensitive electrodes, mGluR5 activation by glutamate was shown to stimulate NO production that depended on activation of neuronal nitric oxide synthase (nNOS). nNOS is expressed in ∼1% of NAcore neurons. Using a transgene strategy to express and stimulate designer receptors that mimicked mGluR5 signaling through Gq in nNOS interneurons, we recapitulated cue-induced reinstatement in the absence of cues. Conversely, using a transgenic caspase strategy, the intensity of cue-induced reinstatement was correlated with the extent of selective elimination of nNOS interneurons. The induction of t-SP during cued reinstatement depends on activating matrix metalloproteinases (MMPs) and selective chemogenetic stimulation of nNOS interneurons recapitulated MMP activation and t-SP induction (increase in AMPA currents in MSNs). These data demonstrate critical involvement of a sparse population of nNOS-expressing interneurons in cue-induced cocaine seeking, revealing a bottleneck in brain processing of drug-associated cues where therapeutic interventions could be effective in treating drug addiction. SIGNIFICANCE STATEMENT Relapse to cocaine use in a rat model is associated with transient increases in synaptic strength at prefrontal cortex synapses in the nucleus accumbens. We demonstrate the sequence of events that mediates synaptic potentiation and reinstated cocaine seeking induced by cocaine-conditioned cues. Activation of prefrontal inputs to the accumbens by cues initiates spillover of synaptic glutamate, which stimulates metabotropic glutamate receptor 5 (mGluR5) on a small population of interneurons (∼1%) expressing neuronal nitric oxide synthase. Stimulating these glutamate receptors increases nitric oxide (NO) production, which stimulates matrix metalloprotease-2 (MMP-2) and MMP-9 activity in the extracellular space. Manipulating the interaction between mGluR5, NO production, or MMP-2 and MMP-9 pharmacologically or genetically is sufficient to recapitulate transient synaptic potentiation and reinstate cocaine seeking.
Collapse
Affiliation(s)
- Alexander C W Smith
- Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Michael D Scofield
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Jasper A Heinsbroek
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Cassandra D Gipson
- Department of Psychology, Arizona State University, Tempe, Arizona 85287
| | - Daniela Neuhofer
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Doug J Roberts-Wolfe
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Sade Spencer
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Constanza Garcia-Keller
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Neringa M Stankeviciute
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Rachel J Smith
- Department of Psychology, Institute for Neuroscience, Texas A&M University, College Station, Texas 77843, and
| | - Nicholas P Allen
- Department of Biology, College of Charleston, Charleston, South Carolina 29401
| | - Melissa R Lorang
- Department of Biology, College of Charleston, Charleston, South Carolina 29401
| | - William C Griffin
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Heather A Boger
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Peter W Kalivas
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina 29425,
| |
Collapse
|
27
|
Lasek AW. Effects of Ethanol on Brain Extracellular Matrix: Implications for Alcohol Use Disorder. Alcohol Clin Exp Res 2016; 40:2030-2042. [PMID: 27581478 DOI: 10.1111/acer.13200] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 07/24/2016] [Indexed: 12/20/2022]
Abstract
The brain extracellular matrix (ECM) occupies the space between cells and is involved in cell-matrix and cell-cell adhesion. However, in addition to providing structural support to brain tissue, the ECM activates cell signaling and controls synaptic transmission. The expression and activity of brain ECM components are regulated by alcohol exposure. This review will discuss what is currently known about the effects of alcohol on the activity and expression of brain ECM components. An interpretation of how these changes might promote alcohol use disorder (AUD) will be also provided. Ethanol (EtOH) exposure decreases levels of structural proteins involved in the interstitial matrix and basement membrane, with a concomitant increase in proteolytic enzymes that degrade these components. In contrast, EtOH exposure generally increases perineuronal net components. Because the ECM has been shown to regulate both synaptic plasticity and behavioral responses to drugs of abuse, regulation of the brain ECM by alcohol may be relevant to the development of alcoholism. Although investigation of the function of brain ECM in alcohol abuse is still in early stages, a greater understanding of the interplay between ECM and alcohol might lead to novel therapeutic strategies for treating AUD.
Collapse
Affiliation(s)
- Amy W Lasek
- Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois.
| |
Collapse
|
28
|
Mulholland PJ, Chandler LJ, Kalivas PW. Signals from the Fourth Dimension Regulate Drug Relapse. Trends Neurosci 2016; 39:472-485. [PMID: 27173064 PMCID: PMC4930682 DOI: 10.1016/j.tins.2016.04.007] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Revised: 04/12/2016] [Accepted: 04/14/2016] [Indexed: 12/21/2022]
Abstract
Despite the enormous societal burden of alcohol and drug addiction and abundant research describing drug-induced maladaptive synaptic plasticity, there are few effective strategies for treating substance use disorders. Recent awareness that synaptic plasticity involves astroglia and the extracellular matrix is revealing new possibilities for understanding and treating addiction. We first review constitutive corticostriatal adaptations that are elicited by and shared between all abused drugs from the perspective of tetrapartite synapses, and integrate recent discoveries regarding cell type-specificity in striatal neurons. Next, we describe recent discoveries that drug-seeking is associated with transient synaptic plasticity that requires all four synaptic elements and is shared across drug classes. Finally, we prognosticate how considering tetrapartite synapses can provide new treatment strategies for addiction.
Collapse
Affiliation(s)
- Patrick J Mulholland
- Department of Neuroscience, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA; Department of Psychiatry and Behavioral Sciences, Addiction Sciences Division, Medical University of South Carolina, 67 President Street, Charleston, SC, 29425, USA.
| | - L Judson Chandler
- Department of Neuroscience, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA; Department of Psychiatry and Behavioral Sciences, Addiction Sciences Division, Medical University of South Carolina, 67 President Street, Charleston, SC, 29425, USA
| | - Peter W Kalivas
- Department of Neuroscience, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA.
| |
Collapse
|
29
|
Vafadari B, Salamian A, Kaczmarek L. MMP-9 in translation: from molecule to brain physiology, pathology, and therapy. J Neurochem 2016; 139 Suppl 2:91-114. [PMID: 26525923 DOI: 10.1111/jnc.13415] [Citation(s) in RCA: 265] [Impact Index Per Article: 33.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 10/13/2015] [Accepted: 10/19/2015] [Indexed: 12/11/2022]
Abstract
Matrix metalloproteinase-9 (MMP-9) is a member of the metzincin family of mostly extracellularly operating proteases. Despite the fact that all of these enzymes might be target promiscuous, with largely overlapping catalogs of potential substrates, MMP-9 has recently emerged as a major and apparently unique player in brain physiology and pathology. The specificity of MMP-9 may arise from its very local and time-restricted actions, even when released in the brain from cells of various types, including neurons, glia, and leukocytes. In fact, the quantity of MMP-9 is very low in the naive brain, but it is markedly activated at the levels of enzymatic activity, protein abundance, and gene expression following various physiological stimuli and pathological insults. Neuronal MMP-9 participates in synaptic plasticity by controlling the shape of dendritic spines and function of excitatory synapses, thus playing a pivotal role in learning, memory, and cortical plasticity. When improperly unleashed, MMP-9 contributes to a large variety of brain disorders, including epilepsy, schizophrenia, autism spectrum disorder, brain injury, stroke, neurodegeneration, pain, brain tumors, etc. The foremost mechanism of action of MMP-9 in brain disorders appears to be its involvement in immune/inflammation responses that are related to the enzyme's ability to process and activate various cytokines and chemokines, as well as its contribution to blood-brain barrier disruption, facilitating the extravasation of leukocytes into brain parenchyma. However, another emerging possibility (i.e., the control of MMP-9 over synaptic plasticity) should not be neglected. The translational potential of MMP-9 has already been recognized in both the diagnosis and treatment domains. The most striking translational aspect may be the discovery of MMP-9 up-regulation in a mouse model of Fragile X syndrome, quickly followed by human studies and promising clinical trials that have sought to inhibit MMP-9. With regard to diagnosis, suggestions have been made to use MMP-9 alone or combined with tissue inhibitor of matrix metalloproteinase-1 or brain-derived neurotrophic factor as disease biomarkers. MMP-9, through cleavage of specific target proteins, plays a major role in synaptic plasticity and neuroinflammation, and by those virtues contributes to brain physiology and a host of neurological and psychiatric disorders. This article is part of the 60th Anniversary special issue.
Collapse
|
30
|
Becker HC, Lopez MF. An Animal Model of Alcohol Dependence to Screen Medications for Treating Alcoholism. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2016; 126:157-77. [PMID: 27055614 DOI: 10.1016/bs.irn.2016.02.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Despite the high prevalence of alcohol use disorders in the United States, only a relatively small percentage of those afflicted seek treatment. This is further compounded by the fact that there are too few medications available to effectively treat this significant public health problem. The need for identifying and evaluating more effective treatments that aid in preventing relapse and/or tempering risky and harmful alcohol consumption cannot be overstated. Use of animal models represents a critical step in the process of screening, identifying, and informing plans for prioritizing the most promising candidate medications that can be advanced to the next stage of evaluation (clinical laboratory paradigms and controlled clinical trials). Numerous animal models have been developed to study excessive levels of alcohol self-administration. In recent years, a large literature has amassed of studies in which rodent models of dependence have been linked with alcohol self-administration procedures. This chapter focuses on studies employing a dependence model that involves chronic exposure to alcohol vapor by inhalation, which yields in both mice and rats significant escalation of voluntary alcohol consumption. These animal models of dependence and alcohol self-administration have revealed valuable insights about underlying mechanisms that drive excessive drinking. Additionally, this preclinical approach is useful in evaluating the effects of medications on escalated drinking associated with dependence vs more stable levels displayed by nondependent animals.
Collapse
Affiliation(s)
- H C Becker
- Charleston Alcohol Research Center, Charleston, SC, United States; Medical University of South Carolina, Charleston, SC, United States; RHJ Department of Veterans Affairs Medical Center, Charleston, SC, United States.
| | - M F Lopez
- Charleston Alcohol Research Center, Charleston, SC, United States
| |
Collapse
|
31
|
Caught in the Net: Perineuronal Nets and Addiction. Neural Plast 2016; 2016:7538208. [PMID: 26904301 PMCID: PMC4745418 DOI: 10.1155/2016/7538208] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 11/23/2015] [Indexed: 12/14/2022] Open
Abstract
Exposure to drugs of abuse induces plasticity in the brain and creates persistent drug-related memories. These changes in plasticity and persistent drug memories are believed to produce aberrant motivation and reinforcement contributing to addiction. Most studies have explored the effect drugs of abuse have on pre- and postsynaptic cells and astrocytes; however, more recently, attention has shifted to explore the effect these drugs have on the extracellular matrix (ECM). Within the ECM are unique structures arranged in a net-like manner, surrounding a subset of neurons called perineuronal nets (PNNs). This review focuses on drug-induced changes in PNNs, the molecules that regulate PNNs, and the expression of PNNs within brain circuitry mediating motivation, reward, and reinforcement as it pertains to addiction.
Collapse
|
32
|
Koob GF, Mason BJ. Existing and Future Drugs for the Treatment of the Dark Side of Addiction. Annu Rev Pharmacol Toxicol 2015; 56:299-322. [PMID: 26514207 DOI: 10.1146/annurev-pharmtox-010715-103143] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The identification of a heuristic framework for the stages of the addiction cycle that are linked to neurocircuitry changes in pathophysiology includes the binge/intoxication stage, the withdrawal/negative affect stage, and the preoccupation/anticipation (craving) stage, which represent neuroadaptations in three neurocircuits (basal ganglia, extended amygdala, and frontal cortex, respectively). The identification of excellent and validated animal models, the development of human laboratory models, and an enormous surge in our understanding of neurocircuitry and neuropharmacological mechanisms have provided a revisionist view of addiction that emphasizes the loss of brain reward function and gain of stress function that drive negative reinforcement (the dark side of addiction) as a key to compulsive drug seeking. Reversing the dark side of addiction not only explains much of the existing successful pharmacotherapies for addiction but also points to vast new opportunities for future medications to alleviate this major source of human suffering.
Collapse
Affiliation(s)
- George F Koob
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, California 92037; ,
| | - Barbara J Mason
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, California 92037; ,
| |
Collapse
|
33
|
Costa PA, Poli JHZ, Sperotto NDM, Moura DJ, Saffi J, Nin MS, Barros HMT. Brain DNA damage and behavioral changes after repeated intermittent acute ethanol withdrawal by young rats. Psychopharmacology (Berl) 2015; 232:3623-36. [PMID: 26231496 DOI: 10.1007/s00213-015-4015-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2015] [Accepted: 06/30/2015] [Indexed: 01/10/2023]
Abstract
RATIONALE Alcohol addiction causes severe problems, and its deprivation may potentiate symptoms such as anxiety. Furthermore, ethanol is a neurotoxic agent that induces degeneration and the consequences underlying alcohol-mediated brain damage remain unclear. OBJECTIVES This study assessed the behavioral changes during acute ethanol withdrawal periods and determined the levels of DNA damage and reactive oxygen species (ROS) in multiple brain areas. METHODS Male Wistar rats were subjected to an oral ethanol self-administration procedure with a forced diet where they were offered 8% (v/v) ethanol solution for 21 days followed by five repeated 24-h cycles alternating between ethanol withdrawal and re-exposure. Control animals received an isocaloric control diet without ethanol. Behavioral changes were analyzed on ethanol withdrawal days in the open-field (OF) and elevated plus-maze (EPM) tests within the first 6 h of ethanol deprivation. The pre-frontal cortex, hypothalamus, striatum, hippocampus, and cerebellum were dissected for alkaline and neutral comet assays and for dichlorofluorescein ROS testing. RESULTS The repeated intermittent ethanol access enhanced solution intake and alcohol-seeking behavior. Decreased exploratory activity was observed in the OF test, and the animals stretched less in the EPM test. DNA single-strand breaks and ROS production were significantly higher in all structures evaluated in the ethanol-treated rats compared with controls. CONCLUSIONS The animal model of repeated intermittent ethanol access induced behavioral changes in rats, and this ethanol exposure model induced an increase in DNA single-strand breaks and ROS production in all brain areas. Our results suggest that these brain damages may influence future behaviors.
Collapse
Affiliation(s)
- Priscila A Costa
- Laboratory of Neuropsycopharmacology, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
| | | | | | | | | | | | | |
Collapse
|
34
|
The tetrapartite synapse: Extracellular matrix remodeling contributes to corticoaccumbens plasticity underlying drug addiction. Brain Res 2015; 1628:29-39. [PMID: 25838241 DOI: 10.1016/j.brainres.2015.03.027] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 02/10/2015] [Accepted: 03/13/2015] [Indexed: 12/29/2022]
Abstract
Synaptic plasticity has long been known to involve three key elements of neuropil, the presynapse, the postsynapse and adjacent glia. Here we review the role of the extracellular matrix in synaptic plasticity as a necessary component forming the tetrapartite synapse. We describe the role of matrix metalloproteinases as enzymes sculpting extracellular proteins and thereby creating an extracellular signaling domain required for synaptic plasticity. Specifically we focus on the role of the tetrapartite synapse in mediating the effects of addictive drugs at cortico-striatal synapses, and conclude that the extracellular signaling domain and its regulation by matrix metalloproteinases is critical for developing and expressing drug seeking behaviors.
Collapse
|
35
|
Vendruscolo LF, Roberts AJ. Operant alcohol self-administration in dependent rats: focus on the vapor model. Alcohol 2014; 48:277-86. [PMID: 24290310 DOI: 10.1016/j.alcohol.2013.08.006] [Citation(s) in RCA: 124] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Revised: 08/02/2013] [Accepted: 08/05/2013] [Indexed: 12/28/2022]
Abstract
Alcoholism (alcohol dependence) is characterized by a compulsion to seek and ingest alcohol (ethanol), loss of control over intake, and the emergence of a negative emotional state during withdrawal. Animal models are critical in promoting our knowledge of the neurobiological mechanisms underlying alcohol dependence. Here, we review the studies involving operant alcohol self-administration in rat models of alcohol dependence and withdrawal with the focus on the alcohol vapor model. In 1996, the first articles were published reporting that rats made dependent on alcohol by exposure to alcohol vapors displayed increased operant alcohol self-administration during acute withdrawal compared with nondependent rats (i.e., not exposed to alcohol vapors). Since then, it has been repeatedly demonstrated that this model reliably produces physical and motivational symptoms of alcohol dependence. The functional roles of various systems implicated in stress and reward, including opioids, dopamine, corticotropin-releasing factor (CRF), glucocorticoids, neuropeptide Y (NPY), γ-aminobutyric acid (GABA), norepinephrine, and cannabinoids, have been investigated in the context of alcohol dependence. The combination of models of alcohol withdrawal and dependence with operant self-administration constitutes an excellent tool to investigate the neurobiology of alcoholism. In fact, this work has helped lay the groundwork for several ongoing clinical trials for alcohol dependence. Advantages and limitations of this model are discussed, with an emphasis on what future directions of great importance could be.
Collapse
Affiliation(s)
- Leandro F Vendruscolo
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Amanda J Roberts
- Molecular and Cellular Neuroscience Department, Mouse Behavioral Assessment Core, The Scripps Research Institute, 10550 North Torrey Pines Road, MB18, La Jolla, CA 92037, USA.
| |
Collapse
|
36
|
Abstract
The extracellular matrix (ECM) has a prominent role in brain development, maturation of neural circuits, and adult neuroplasticity. This multifactorial role of the ECM suggests that processes that affect composition or turnover of ECM in the brain could lead to altered brain function, possibly underlying conditions of impaired mental health, such as neuropsychiatric or neurodegenerative disease. In support of this, in the last two decades, clinical and preclinical research provided evidence of correlations and to some degree causal links, between aberrant ECM function and neuropsychiatric disorders, the most prominent being addiction and schizophrenia. Based on these initial observations of involvement of different classes of ECM molecules (laminin, reelin, and their integrin receptors, as well as tenascins and chondroitin sulfate proteoglycans), ECM targets have been suggested as a novel entry point in the treatment of neuropsychiatric disorders. Hence, understanding how ECM molecules contribute to proper neuronal functioning and how this is dysregulated in conditions of mental illness is of pivotal importance. In this chapter, we will review available literature that implicates the different classes of brain ECM molecules in psychiatric disorders, with a primary focus on addiction (opiates, psychostimulants, and alcohol), and we will compare these ECM adaptations with those implicated in schizophrenia and mood disorders.
Collapse
|
37
|
Natarajan R, Harding JW, Wright JW. A role for matrix metalloproteinases in nicotine-induced conditioned place preference and relapse in adolescent female rats. J Exp Neurosci 2013; 7:1-14. [PMID: 25157203 PMCID: PMC4089657 DOI: 10.4137/jen.s11381] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Reconfiguration of extracellular matrix proteins appears to be necessary for the synaptic plasticity that underlies memory consolidation. The primary candidates involved in controlling this process are a family of endopeptidases called matrix metalloproteinases (MMPs); however, the potential role of MMPs in nicotine addiction-related memories has not been adequately tested. Present results indicate transient changes in hippocampal MMP-2, -3, and -9 expression following context dependent learning of nicotine-induced conditioned place preference (CPP). Members of a CPP procedural control group also indicated similar MMP changes, suggesting that memory activation occurred in these animals as well. However, hippocampal MMP-9 expression was differentially elevated in members of the nicotine-induced CPP group on days 4 and 5 of training. Inhibition of MMPs using a broad spectrum MMP inhibitor (FN439) during nicotine-induced CPP training blocked the acquisition of CPP. Elevations in hippocampal and prefrontal cortex MMP-3 expression-but not MMP-2 and -9-accompanied reactivation of a previously learned drug related memory. Decreases in the actin regulatory cytoskeletal protein cortactin were measured in the HIP and PFC during the initial two days of acquisition of CPP; however, no changes were seen following re-exposure to the drug related environment. These results suggest that MMP-9 may be involved in facilitating the intracellular and extracellular events required for the synaptic plasticity underlying the acquisition of nicotine-induced CPP. Furthermore, MMP-3 appears to be important during re-exposure to the drug associated environment. However, rats introduced into the CPP apparatus and given injections of vehicle rather than nicotine during training also revealed a pattern of MMP expression similar to nicotine-induced CPP animals.
Collapse
Affiliation(s)
- Reka Natarajan
- Department of Neurosciences, University of Toledo College of Medicine, Toledo, OH, USA
| | - Joseph W Harding
- Departments of Psychology, and Veterinary and Comparative Anatomy, Pharmacology, and Physiology, Washington State University, Pullman, WA, USA
| | - John W Wright
- Departments of Psychology, and Veterinary and Comparative Anatomy, Pharmacology, and Physiology, Washington State University, Pullman, WA, USA
| |
Collapse
|
38
|
Williams AM, Reis DJ, Powell AS, Neira LJ, Nealey KA, Ziegler CE, Kloss N, Bilimoria JL, Smith CE, Walker BM. The effect of intermittent alcohol vapor or pulsatile heroin on somatic and negative affective indices during spontaneous withdrawal in Wistar rats. Psychopharmacology (Berl) 2012; 223:75-88. [PMID: 22461104 PMCID: PMC3419345 DOI: 10.1007/s00213-012-2691-3] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Accepted: 03/09/2012] [Indexed: 12/31/2022]
Abstract
RATIONALE Once dependent on alcohol or opioids, negative affect may accompany withdrawal. Dependent individuals are hypothesized to "self-medicate" in order to cope with withdrawal, which promotes escalated alcohol and drug use. OBJECTIVES The current study aimed to develop a reliable animal model to assess symptoms that occur during spontaneous alcohol and opioid withdrawal. METHODS Dependence was induced using intermittent alcohol exposure or pulsatile heroin delivery and assessed for the presence of withdrawal symptoms during acute withdrawal by measuring somatic signs, behavior in the forced swim test (FST), and air-puff-induced 22-kHz ultrasonic vocalizations (USVs). Additional animals subjected to 8 weeks of alcohol vapor exposure were evaluated for altered somatic signs, operant alcohol self-administration, and 22-kHz USV production, as well as performance in the elevated plus maze (EPM). RESULTS During spontaneous withdrawal from pulsatile heroin or intermittent alcohol vapor, animals displayed increased somatic withdrawal signs, FST immobility, and 22-kHz USV production but did not show any behavioral change in the EPM unless the duration of alcohol exposure was extended to 4 weeks. Following 8 weeks of alcohol vapor exposure, animals displayed somatic withdrawal signs, escalated alcohol self-administration, and increased 22-kHz USVs. CONCLUSIONS These paradigms provide consistent methods to evaluate the behavioral ramifications, and neurobiological substrates, of alcohol and opioid dependence during spontaneous withdrawal. As immobility in the FST and percent open-arm time in the EPM were dissociable, with 22-kHz USVs paralleling immobility in the FST, assessment of air-puff-induced 22-kHz USVs could provide an ethologically valid alternative to the FST.
Collapse
Affiliation(s)
- Angela M. Williams
- Laboratory of Alcoholism and Addictions Neuroscience, Department of Psychology, Washington State University, Pullman, WA
| | - Daniel J. Reis
- Laboratory of Alcoholism and Addictions Neuroscience, Department of Psychology, Washington State University, Pullman, WA
| | - Alexa S. Powell
- Laboratory of Alcoholism and Addictions Neuroscience, Department of Psychology, Washington State University, Pullman, WA
| | - Louis J. Neira
- Laboratory of Alcoholism and Addictions Neuroscience, Department of Psychology, Washington State University, Pullman, WA
| | - Kathryn A. Nealey
- Laboratory of Alcoholism and Addictions Neuroscience, Department of Psychology, Washington State University, Pullman, WA
| | - Cole E. Ziegler
- Laboratory of Alcoholism and Addictions Neuroscience, Department of Psychology, Washington State University, Pullman, WA
| | - Nina Kloss
- Laboratory of Alcoholism and Addictions Neuroscience, Department of Psychology, Washington State University, Pullman, WA
| | - Jessica L. Bilimoria
- Laboratory of Alcoholism and Addictions Neuroscience, Department of Psychology, Washington State University, Pullman, WA
| | - Chelsea E. Smith
- Laboratory of Alcoholism and Addictions Neuroscience, Department of Psychology, Washington State University, Pullman, WA
| | - Brendan M. Walker
- Laboratory of Alcoholism and Addictions Neuroscience, Department of Psychology, Washington State University, Pullman, WA,Graduate Program in Neuroscience, Washington State University, Pullman, WA,Alcohol and Drug Abuse Research Program, Washington State University, Pullman, WA,Translational Addiction Research Center, Washington State University, Pullman, WA,Corresponding Author: Dr. Brendan M. Walker, Laboratory of Alcoholism and Addictions Neuroscience, Department of Psychology, Graduate Program in Neuroscience, Alcohol and Drug Abuse Research Program, Translational Addiction Research Center, 100 Dairy Road, Mail Code: 644820, Washington State University, Pullman, WA 99164-4820, 509-335-8526 (phone), 509-335-5043 (fax),
| |
Collapse
|
39
|
Wright JW, Wilson WL, Wakeling V, Boydstun AS, Jensen A, Kawas L, Harding JW. The Hepatocyte Growth Factor/c-Met Antagonist, Divalinal-Angiotensin IV, Blocks the Acquisition of Methamphetamine Dependent Conditioned Place Preference in Rats. Brain Sci 2012; 2:298-318. [PMID: 24961196 PMCID: PMC4061800 DOI: 10.3390/brainsci2030298] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2012] [Revised: 07/06/2012] [Accepted: 08/10/2012] [Indexed: 11/16/2022] Open
Abstract
The use of methamphetamine (MA) is increasing in the U.S. and elsewhere around the world. MA's capacity to cause addiction significantly exceeds other psychostimulant drugs, and its use negatively impacts learning and memory. Recently, attempts have been made to interfere with the presumed mechanism(s) underlying the establishment of drug-induced memory consolidation. The majority of these studies have employed matrix metalloproteinase (MMP) inhibitors to disrupt MMP-induced extracellular matrix molecule dependent synaptic reconfiguration, or GABA receptor agonists. The present investigation utilized an angiotensin IV (AngIV) analogue, Divalinal-AngIV (divalinal), to disrupt acquisition of MA-induced dependence in rats as measured using the conditioned place preference paradigm. Results indicate that both acute and chronic intracerebroventricular infusion of divalinal prior to each daily subcutaneous injection of MA prevented acquisition. However, divalinal was unable to prevent MA-induced reinstatement after prior acquisition followed by extinction trials. These results indicate that prevention of MA dependence can be accomplished by blockade of the brain AT4 receptor subtype. On the other hand, once MA-induced memory consolidation is in place divalinal appears to be ineffective. Mechanistic studies indicated that divalinal is a potent inhibitor of the hepatocyte growth factor (HGF)/c-Met receptor system, and thus it appears that a functional HGF/c-Met system is required for the acquisition of MA-mediated conditioned place preference.
Collapse
Affiliation(s)
- John W Wright
- Department of Psychology, Washington State University, Pullman, WA 99164-4820, USA.
| | - Wendy L Wilson
- Department of Psychology, Dickinson State University, Dickinson, ND 58601, USA.
| | - Vanessa Wakeling
- Department of Psychology, Washington State University, Pullman, WA 99164-4820, USA.
| | - Alan S Boydstun
- L-3 Communications, Link Simulation and Training, Wright Patterson Air Force Base, OH 45433-7955, USA.
| | - Audrey Jensen
- Department of Psychology, Washington State University, Pullman, WA 99164-4820, USA.
| | - Leen Kawas
- Program in Pharmacology and Toxicology, Washington State University, Pullman, WA 99164-6510, USA.
| | - Joseph W Harding
- Department of Psychology, Washington State University, Pullman, WA 99164-4820, USA.
| |
Collapse
|
40
|
Conceptualizing withdrawal-induced escalation of alcohol self-administration as a learned, plasticity-dependent process. Alcohol 2012; 46:339-48. [PMID: 22459874 DOI: 10.1016/j.alcohol.2012.01.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2011] [Revised: 01/11/2012] [Accepted: 01/24/2012] [Indexed: 11/24/2022]
Abstract
This article represents one of five contributions focusing on the topic "Plasticity and neuroadaptive responses within the extended amygdala in response to chronic or excessive alcohol exposure" that were developed by awardees participating in the Young Investigator Award Symposium at the "Alcoholism and Stress: A Framework for Future Treatment Strategies" conference in Volterra, Italy on May 3-6, 2011 that was organized/chaired by Drs. Antonio Noronha and Fulton Crews and sponsored by the National Institute on Alcohol Abuse and Alcoholism. This review discusses the dependence-induced neuroadaptations in affective systems that provide a basis for negative reinforcement learning and presents evidence demonstrating that escalated alcohol consumption during withdrawal is a learned, plasticity-dependent process. The review concludes by identifying changes within extended amygdala dynorphin/kappa-opioid receptor systems that could serve as the foundation for the occurrence of negative reinforcement processes. While some evidence contained herein may be specific to alcohol dependence-related learning and plasticity, much of the information will be of relevance to any addictive disorder involving negative reinforcement mechanisms. Collectively, the information presented within this review provides a framework to assess the negative reinforcing effects of alcohol in a manner that distinguishes neuroadaptations produced by chronic alcohol exposure from the actual plasticity that is associated with negative reinforcement learning in dependent organisms.
Collapse
|
41
|
Matrix metalloproteinases contribute to neuronal dysfunction in animal models of drug dependence, Alzheimer's disease, and epilepsy. Biochem Res Int 2011; 2011:681385. [PMID: 22235372 PMCID: PMC3253438 DOI: 10.1155/2011/681385] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2011] [Accepted: 11/17/2011] [Indexed: 01/07/2023] Open
Abstract
Matrix metalloproteinases (MMPs) and tissue inhibitors of metalloproteinases (TIMPs) remodel the pericellular environment by regulating the cleavage of extracellular matrix proteins, cell surface components, neurotransmitter receptors, and growth factors that mediate cell adhesion, synaptogenesis, synaptic plasticity, and long-term potentiation. Interestingly, increased MMP activity and dysregulation of the balance between MMPs and TIMPs have also been implicated in various pathologic conditions. In this paper, we discuss various animal models that suggest that the activation of the gelatinases MMP-2 and MMP-9 is involved in pathogenesis of drug dependence, Alzheimer's disease, and epilepsy.
Collapse
|