1
|
Mielecki D, Bratek-Gerej E, Salińska E. Metabotropic glutamate receptors-guardians and gatekeepers in neonatal hypoxic-ischemic brain injury. Pharmacol Rep 2024:10.1007/s43440-024-00653-x. [PMID: 39289333 DOI: 10.1007/s43440-024-00653-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 09/03/2024] [Accepted: 09/11/2024] [Indexed: 09/19/2024]
Abstract
Injury to the developing central nervous system resulting from perinatal hypoxia-ischemia (HI) is still a clinical challenge. The only approach currently available in clinical practice for severe cases of HI is therapeutic hypothermia, initiated shortly after birth and supported by medications to regulate blood pressure, control epileptic seizures, and dialysis to support kidney function. However, these treatments are not effective enough to significantly improve infant survival or prevent brain damage. The need to create a new effective therapy has focused attention on metabotropic glutamate receptors (mGluR), which control signaling pathways involved in HI-induced neurodegeneration. The complexity of mGluR actions, considering their localization and developmental changes, and the functions of each subtype in HI-evoked brain damage, combined with difficulties in the availability of safe and effective modulators, raises the question whether modulation of mGluRs with subtype-selective ligands can become a new treatment in neonatal HI. Addressing this question, this review presents the available information concerning the role of each of the eight receptor subtypes of the three mGluR groups (group I, II, and III). Data obtained from experiments performed on in vitro and in vivo neonatal HI models show the neuroprotective potential of group I mGluR antagonists, as well as group II and III agonists. The information collected in this work indicates that the neuroprotective effects of manipulating mGluR in experimental HI models, despite the need to create more safe and selective ligands for particular receptors, provide a chance to create new therapies for the sensitive brains of infants at risk.
Collapse
Affiliation(s)
- Damian Mielecki
- Department of Neurochemistry, Mossakowski Medical Research Institute PAS, Pawińskiego 5, 02-106, Warsaw, Poland
| | - Ewelina Bratek-Gerej
- Department of Neurochemistry, Mossakowski Medical Research Institute PAS, Pawińskiego 5, 02-106, Warsaw, Poland
| | - Elżbieta Salińska
- Department of Neurochemistry, Mossakowski Medical Research Institute PAS, Pawińskiego 5, 02-106, Warsaw, Poland.
| |
Collapse
|
2
|
Mielecki D, Salińska E. Group III metabotropic glutamate receptors: guardians against excitotoxicity in ischemic brain injury, with implications for neonatal contexts. Pharmacol Rep 2024:10.1007/s43440-024-00651-z. [PMID: 39298028 DOI: 10.1007/s43440-024-00651-z] [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: 07/10/2024] [Revised: 09/06/2024] [Accepted: 09/09/2024] [Indexed: 09/21/2024]
Abstract
The group III metabotropic glutamate receptors (mGluRs), comprising mGluR4, mGluR6, mGluR7, and mGluR8, offer neuroprotective potential in mitigating excitotoxicity during ischemic brain injury, particularly in neonatal contexts. They are G-protein coupled receptors that inhibit adenylyl cyclase and reduce neurotransmitter release, mainly located presynaptically and acting as autoreceptors. This review aims to examine the differential expression and function of group III mGluRs across various brain regions such as the cortex, hippocampus, and cerebellum, with a special focus on the neonatal stage of development. Glutamate excitotoxicity plays a crucial role in the pathophysiology of brain ischemia in neonates. While ionotropic glutamate receptors are traditional targets for neuroprotection, their direct inhibition often leads to severe side effects due to their critical roles in normal neurotransmission and synaptic plasticity. Group III mGluRs provide a more nuanced and potentially safer approach by modulating rather than blocking glutamatergic transmission. Their downstream signaling cascade results in the regulation of intracellular calcium levels, neuronal hyperpolarization, and reduced neurotransmitter release, effectively decreasing excitotoxic signaling without completely suppressing essential glutamatergic functions. Importantly, the neuroprotective effects of group III mGluRs extend beyond direct modulation of glutamate release influencing glial cell function, neuroinflammation, and oxidative stress, all of which contribute to secondary injury cascades in brain ischemia. This comprehensive analysis of group III mGluRs multifaceted neuroprotective potential provides valuable insights for developing novel therapeutic strategies to combat excitotoxicity in neonatal ischemic brain injury.
Collapse
Affiliation(s)
- Damian Mielecki
- Department of Neurochemistry, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawińskiego 5, Warsaw, 02-106, Poland.
| | - Elżbieta Salińska
- Department of Neurochemistry, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawińskiego 5, Warsaw, 02-106, Poland
| |
Collapse
|
3
|
LaCrosse AL, May CE, Griffin WC, Olive MF. mGluR5 positive allosteric modulation prevents MK-801 induced increases in extracellular glutamate in the rat medial prefrontal cortex. Neuroscience 2024; 555:83-91. [PMID: 39019391 PMCID: PMC11344657 DOI: 10.1016/j.neuroscience.2024.06.016] [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: 01/29/2024] [Revised: 06/17/2024] [Accepted: 06/18/2024] [Indexed: 07/19/2024]
Abstract
Potentiation of metabotropic glutamate receptor subtype 5 (mGluR5) function produces antipsychotic-like and pro-cognitive effects in animal models of schizophrenia and can reverse cognitive deficits induced by N-methyl-D-aspartate type glutamate receptor (NMDAR) antagonists. However, it is currently unknown if mGluR5 positive allosteric modulators (PAMs) can modulate NMDAR antagonist-induced alterations in extracellular glutamate levels in regions underlying these cognitive and behavioral effects, such as the medial prefrontal cortex (mPFC). We therefore assessed the ability of the mGluR5 PAM, 3-cyano-N-(1,3-diphenyl-1H-pyrazol-5-yl) benzamide (CDPPB), to reduce elevated extracellular glutamate levels induced by the NMDAR antagonist, dizocilpine (MK-801), in the mPFC. Male Sprague-Dawley rats were implanted with a guide cannula aimed at the mPFC and treated for ten consecutive days with MK-801 and CDPPB or their corresponding vehicles. CDPPB or vehicle was administered thirty minutes before MK-801 or vehicle each day. On the final day of treatment, in vivo microdialysis was performed, and samples were collected every thirty minutes to analyze extracellular glutamate levels. Compared to animals receiving only vehicle, administration of MK-801 alone significantly increased extracellular levels of glutamate in the mPFC. This effect was not observed in animals administered CDPPB before MK-801, nor in those administered CDPPB alone, indicating that CDPPB decreased extracellular glutamate release stimulated by MK-801. Results indicate that CDPPB attenuates MK-801 induced elevations in extracellular glutamate in the mPFC. This effect of CDPPB may underlie neurochemical adaptations associated with the pro-cognitive effects of mGluR5 PAMs in rodent models of schizophrenia.
Collapse
Affiliation(s)
- Amber L LaCrosse
- Department of Psychology, Arizona State University, 950 S. McAllister Ave., Tempe, AZ 85281, United States; Department of Psychological Science, Northern Michigan University, 1401 Presque Isle Ave, Marquette, MI 49855, United States.
| | - Christina E May
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, MSC 861, 67 President Street, Charleston, SC 29425, United States; Department of Neuroscience and Physiology, New York University Grossman School of Medicine, 550 First Avenue, New York, NY 10016, United States
| | - William C Griffin
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, MSC 861, 67 President Street, Charleston, SC 29425, United States
| | - M Foster Olive
- Department of Psychology, Arizona State University, 950 S. McAllister Ave., Tempe, AZ 85281, United States
| |
Collapse
|
4
|
Mozafari R, Karimi-Haghighi S, Fattahi M, Kalivas P, Haghparast A. A review on the role of metabotropic glutamate receptors in neuroplasticity following psychostimulant use disorder. Prog Neuropsychopharmacol Biol Psychiatry 2023; 124:110735. [PMID: 36813105 DOI: 10.1016/j.pnpbp.2023.110735] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 02/03/2023] [Accepted: 02/16/2023] [Indexed: 02/22/2023]
Abstract
Psychostimulant Use Disorder (PUD) is a chronic relapsing disorder with high motivation for drug abuse. In addition to developing PUD, the use of psychostimulants is a growing public health concern because it is associated with several physical and mental health impairments. To date, there are no FDA-confirmed medicines for the treatment of psychostimulant abuse; therefore, clarification of the cellular and molecular alterations participating in PUD is crucial for developing beneficial medications. PUD causes extensive neuroadaptations in glutamatergic circuitry involved in reinforcement and reward processing. These adaptations include both transient and long-lasting changes in glutamate transmission and glutamate receptors, especially metabotropic glutamate receptors, that have been linked to developing and maintaining PUD. Here, we review the roles of all groups of mGluRs,including I,II, and III in synaptic plasticity within brain reward circuitry engaged by psychostimulants (cocaine, amphetamine, methamphetamine, and nicotine). The review concentrates on investigations of psychostimulant-induced behavioral and neurological plasticity, with an ultimate goal to explore circuit and molecular targets with the potential to contribute to the treatment of PUD.
Collapse
Affiliation(s)
- Roghayeh Mozafari
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saeideh Karimi-Haghighi
- Community Based Psychiatric Care Research Center, School of Nursing and Midwifery, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mojdeh Fattahi
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Peter Kalivas
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Abbas Haghparast
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; School of Cognitive Sciences, Institute for Research in Fundamental Sciences, Tehran, Iran; Department of Basic Sciences, Iranian Academy of Medical Sciences, Tehran, Iran.
| |
Collapse
|
5
|
Fan G, Liu M, Liu J, Huang Y. The initiator of neuroexcitotoxicity and ferroptosis in ischemic stroke: Glutamate accumulation. Front Mol Neurosci 2023; 16:1113081. [PMID: 37033381 PMCID: PMC10076579 DOI: 10.3389/fnmol.2023.1113081] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 03/06/2023] [Indexed: 04/11/2023] Open
Abstract
Glutamate plays an important role in excitotoxicity and ferroptosis. Excitotoxicity occurs through over-stimulation of glutamate receptors, specifically NMDAR, while in the non-receptor-mediated pathway, high glutamate concentrations reduce cystine uptake by inhibiting the System Xc-, leading to intracellular glutathione depletion and resulting in ROS accumulation, which contributes to increased lipid peroxidation, mitochondrial damage, and ultimately ferroptosis. Oxidative stress appears to crosstalk between excitotoxicity and ferroptosis, and it is essential to maintain glutamate homeostasis and inhibit oxidative stress responses in vivo. As researchers work to develop natural compounds to further investigate the complex mechanisms and regulatory functions of ferroptosis and excitotoxicity, new avenues will be available for the effective treatment of ischaemic stroke. Therefore, this paper provides a review of the molecular mechanisms and treatment of glutamate-mediated excitotoxicity and ferroptosis.
Collapse
Affiliation(s)
- Genhao Fan
- Graduate School, Tianjin University of Chinese Medicine, Tianjin, China
| | - Menglin Liu
- Graduate School, Tianjin University of Chinese Medicine, Tianjin, China
| | - Jia Liu
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Tianjin University of Chinese Medicine, Tianjin, China
| | - Yuhong Huang
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Tianjin University of Chinese Medicine, Tianjin, China
- *Correspondence: Yuhong Huang,
| |
Collapse
|
6
|
Jensen KL, Jensen SB, Madsen KL. A mechanistic overview of approaches for the treatment of psychostimulant dependence. Front Pharmacol 2022; 13:854176. [PMID: 36160447 PMCID: PMC9493975 DOI: 10.3389/fphar.2022.854176] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 07/21/2022] [Indexed: 11/13/2022] Open
Abstract
Psychostimulant use disorder is a major health issue around the world with enormous individual, family-related and societal consequences, yet there are no effective pharmacological treatments available. In this review, a target-based overview of pharmacological treatments toward psychostimulant addiction will be presented. We will go through therapeutic approaches targeting different aspects of psychostimulant addiction with focus on three major areas; 1) drugs targeting signalling, and metabolism of the dopamine system, 2) drugs targeting either AMPA receptors or metabotropic glutamate receptors of the glutamate system and 3) drugs targeting the severe side-effects of quitting long-term psychostimulant use. For each of these major modes of intervention, findings from pre-clinical studies in rodents to clinical trials in humans will be listed, and future perspectives of the different treatment strategies as well as their potential side-effects will be discussed. Pharmaceuticals modulating the dopamine system, such as antipsychotics, DAT-inhibitors, and disulfiram, have shown some promising results. Cognitive enhancers have been found to increase aspects of behavioural control, and drugs targeting the glutamate system such as modulators of metabotropic glutamate receptors and AMPA receptors have provided interesting changes in relapse behaviour. Furthermore, CRF-antagonists directed toward alleviating the symptoms of the withdrawal stage have been examined with interesting resulting changes in behaviour. There are promising results investigating therapeutics for psychostimulant addiction, but further preclinical work and additional human studies with a more stratified patient selection are needed to prove sufficient evidence of efficacy and tolerability.
Collapse
|
7
|
Luessen DJ, Conn PJ. Allosteric Modulators of Metabotropic Glutamate Receptors as Novel Therapeutics for Neuropsychiatric Disease. Pharmacol Rev 2022; 74:630-661. [PMID: 35710132 PMCID: PMC9553119 DOI: 10.1124/pharmrev.121.000540] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Metabotropic glutamate (mGlu) receptors, a family of G-protein-coupled receptors, have been identified as novel therapeutic targets based on extensive research supporting their diverse contributions to cell signaling and physiology throughout the nervous system and important roles in regulating complex behaviors, such as cognition, reward, and movement. Thus, targeting mGlu receptors may be a promising strategy for the treatment of several brain disorders. Ongoing advances in the discovery of subtype-selective allosteric modulators for mGlu receptors has provided an unprecedented opportunity for highly specific modulation of signaling by individual mGlu receptor subtypes in the brain by targeting sites distinct from orthosteric or endogenous ligand binding sites on mGlu receptors. These pharmacological agents provide the unparalleled opportunity to selectively regulate neuronal excitability, synaptic transmission, and subsequent behavioral output pertinent to many brain disorders. Here, we review preclinical and clinical evidence supporting the utility of mGlu receptor allosteric modulators as novel therapeutic approaches to treat neuropsychiatric diseases, such as schizophrenia, substance use disorders, and stress-related disorders.
Collapse
|
8
|
Nisar S, Bhat AA, Masoodi T, Hashem S, Akhtar S, Ali TA, Amjad S, Chawla S, Bagga P, Frenneaux MP, Reddy R, Fakhro K, Haris M. Genetics of glutamate and its receptors in autism spectrum disorder. Mol Psychiatry 2022; 27:2380-2392. [PMID: 35296811 PMCID: PMC9135628 DOI: 10.1038/s41380-022-01506-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 02/11/2022] [Accepted: 02/22/2022] [Indexed: 12/11/2022]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental impairment characterized by deficits in social interaction skills, impaired communication, and repetitive and restricted behaviors that are thought to be due to altered neurotransmission processes. The amino acid glutamate is an essential excitatory neurotransmitter in the human brain that regulates cognitive functions such as learning and memory, which are usually impaired in ASD. Over the last several years, increasing evidence from genetics, neuroimaging, protein expression, and animal model studies supporting the notion of altered glutamate metabolism has heightened the interest in evaluating glutamatergic dysfunction in ASD. Numerous pharmacological, behavioral, and imaging studies have demonstrated the imbalance in excitatory and inhibitory neurotransmitters, thus revealing the involvement of the glutamatergic system in ASD pathology. Here, we review the effects of genetic alterations on glutamate and its receptors in ASD and the role of non-invasive imaging modalities in detecting these changes. We also highlight the potential therapeutic targets associated with impaired glutamatergic pathways.
Collapse
Affiliation(s)
- Sabah Nisar
- Laboratory of Molecular and Metabolic Imaging, Sidra Medicine, P.O. Box 26999, Doha, Qatar
| | - Ajaz A Bhat
- Laboratory of Molecular and Metabolic Imaging, Sidra Medicine, P.O. Box 26999, Doha, Qatar
| | - Tariq Masoodi
- Laboratory of Molecular and Metabolic Imaging, Sidra Medicine, P.O. Box 26999, Doha, Qatar
| | - Sheema Hashem
- Laboratory of Molecular and Metabolic Imaging, Sidra Medicine, P.O. Box 26999, Doha, Qatar
| | - Sabah Akhtar
- Laboratory of Molecular and Metabolic Imaging, Sidra Medicine, P.O. Box 26999, Doha, Qatar
| | - Tayyiba Akbar Ali
- Laboratory of Molecular and Metabolic Imaging, Sidra Medicine, P.O. Box 26999, Doha, Qatar
| | - Sara Amjad
- Shibli National College, Azamgarh, Uttar Pradesh, 276001, India
| | - Sanjeev Chawla
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Puneet Bagga
- Department of Diagnostic Imaging, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Michael P Frenneaux
- Academic Health System, Hamad Medical Corporation, P.O. Box 3050, Doha, Qatar
| | - Ravinder Reddy
- Center for Advanced Metabolic Imaging in Precision Medicine, Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Khalid Fakhro
- Department of Human Genetics, Sidra Medicine, P.O. Box 26999, Doha, Qatar
- Department of Genetic Medicine, Weill Cornell Medical College, P.O. Box 24144, Doha, Qatar
| | - Mohammad Haris
- Laboratory of Molecular and Metabolic Imaging, Sidra Medicine, P.O. Box 26999, Doha, Qatar.
- Center for Advanced Metabolic Imaging in Precision Medicine, Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Laboratory of Animal Research, Qatar University, P.O. Box 2713, Doha, Qatar.
| |
Collapse
|
9
|
An inpatient human laboratory study assessing the safety and tolerability, pharmacokinetics, and biobehavioral effect of GET 73 when co-administered with alcohol in individuals with alcohol use disorder. Psychopharmacology (Berl) 2022; 239:35-46. [PMID: 34731268 PMCID: PMC8865311 DOI: 10.1007/s00213-021-06008-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 10/08/2021] [Indexed: 12/17/2022]
Abstract
RATIONALE Previous work suggests that GET 73, a novel compound with putative activity on the metabotropic glutamate receptor subtype 5 (mGluR5), may represent a novel pharmacological treatment for alcohol use disorder (AUD). OBJECTIVE In this study, we investigated the safety, tolerability, pharmacokinetics, and biobehavioral effects of GET 73, when co-administered with alcohol, in individuals with alcohol dependence (AD). METHODS This was an inpatient, cross-over, randomized, double-blind, placebo-controlled, human laboratory study with non-treatment-seeking, alcohol-dependent individuals. The study used a within-subject design, with two counterbalanced stages, during which participants received GET 73 and then placebo, or vice versa. During each stage, participants underwent an alcohol interaction session and, on a separate day, an alcohol cue reactivity, followed by an alcohol self-administration session. RESULTS Safety outcomes of GET 73 were excellent with no serious adverse events, nor adverse events of severe grade. The co-administration of alcohol and GET 73 did not affect the pharmacokinetics of GET 73 or alcohol. GET 73, compared to placebo, did not affect the alcohol-related stimulation effects, but increased the subjective sedative effects of alcohol. GET 73 did not affect alcohol cue-induced craving, or alcohol self-administration in the laboratory. CONCLUSIONS The study confirms the safety and tolerability of GET 73 when co-administered with alcohol. Although, under this experimental condition, we did not detect an effect on alcohol craving and consumption in the laboratory, additional studies should be conducted administering GET 73 for an extended period in an outpatient setting.
Collapse
|
10
|
Kondra S, Chen F, Chen Y, Chen Y, Collette CJ, Xu W. A study of a hierarchical structure of proteins and ligand binding sites of receptors using the triangular spatial relationship-based structure comparison method and development of a size-filtering feature designed for comparing different sizes of protein structures. Proteins 2021; 90:239-257. [PMID: 34392570 DOI: 10.1002/prot.26215] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 07/28/2021] [Accepted: 08/09/2021] [Indexed: 12/15/2022]
Abstract
The presence of receptors and the specific binding of the ligands determine nearly all cellular responses. Binding of a ligand to its receptor causes conformational changes of the receptor that triggers the subsequent signaling cascade. Therefore, systematically studying structures of receptors will provide insight into their functions. We have developed the triangular spatial relationship (TSR)-based method where all possible triangles are constructed with Cα atoms of a protein as vertices. Every triangle is represented by an integer denoted as a "key" computed through the TSR algorithm. A structure is thereby represented by a vector of integers. In this study, we have first defined substructures using different types of keys. Second, using different types of keys represents a new way to interpret structure hierarchical relations and differences between structures and sequences. Third, we demonstrate the effects of sequence similarity as well as sample size on the structure-based classifications. Fourth, we show identification of structure motifs, and the motifs containing multiple triangles connected by either an edge or a vertex are mapped to the ligand binding sites of the receptors. The structure motifs are valuable resources for the researchers in the field of signal transduction. Next, we propose amino-acid scoring matrices that capture "evolutionary closeness" information based on BLOSUM62 matrix, and present the development of a new visualization method where keys are organized according to evolutionary closeness and shown in a 2D image. This new visualization opens a window for developing tools with the aim of identification of specific and common substructures by scanning pixels and neighboring pixels. Finally, we report a new algorithm called as size filtering that is designed to improve structure comparison of large proteins with small proteins. Collectively, we provide an in-depth interpretation of structure relations through the detailed analyses of different types of keys and their associated key occurrence frequencies, geometries, and labels. In summary, we consider this study as a new computational platform where keys are served as a bridge to connect sequence and structure as well as structure and function for a deep understanding of sequence, structure, and function relationships of the protein family.
Collapse
Affiliation(s)
- Sarika Kondra
- The Center for Advanced Computer Studies, University of Louisiana at Lafayette, Lafayette, Louisiana, USA
| | - Feng Chen
- High Performance Computing, 329 Frey Computing Services Center, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Yixin Chen
- Department of Computer and Information Science, The University of Mississippi, University, Mississippi, USA
| | - Yuwu Chen
- High Performance Computing, 329 Frey Computing Services Center, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Caleb J Collette
- Department of Chemistry, University of Louisiana at Lafayette, Lafayette, Louisiana, USA
| | - Wu Xu
- Department of Chemistry, University of Louisiana at Lafayette, Lafayette, Louisiana, USA
| |
Collapse
|
11
|
Chang CT, Hsieh PJ, Lee HC, Lo CH, Tam KW, Loh EW. Effectiveness of N-acetylcysteine in Treating Clinical Symptoms of Substance Abuse and Dependence: A Meta-analysis of Randomized Controlled Trials. CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE 2021; 19:282-293. [PMID: 33888657 PMCID: PMC8077050 DOI: 10.9758/cpn.2021.19.2.282] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 08/21/2020] [Accepted: 08/27/2020] [Indexed: 12/15/2022]
Abstract
Objective Treatment with N-acetylcysteine (NAC) is believed to reduce the clinical symptoms among individuals with substance abuse or dependence. We conducted a meta-analysis of randomized controlled trials to evaluate the effectiveness of NAC in treating substance abuse and dependence. Methods PubMed, EMBASE, ClinicalTrials.gov registry, and the Cochrane Library were searched for trials published before June 2020. Results A total of 16 trials were analyzed. The treatment effectiveness domains assessed in this study were craving and depressive symptoms, withdrawal syndrome, adverse events, and smoking frequency. Standardized mean difference (SMD), weighted mean difference (WMD), and odds ratio (OR) were used for evaluation where appropriate. A significant decrease in craving symptoms was observed in the NAC treatment group compared with the control group (SMD, −0.67; 95% confidence interval [CI], −1.21 to 0.21). When withdrawal and depressive symptoms were considered as a single domain, the NAC treatment group demonstrated a significantly higher overall improvement than the control group (SMD, −0.35; 95% CI, −0.64 to −0.06). No between-group differences in term of the OR of adverse events (OR, 1.18; 95% CI, 0.68 to 2.06) and a non-significant trend toward reduction in smoking frequency was observed in the NAC treatment group compared with the control group (WMD, −3.09; 95% CI, −6.50 to 0.32). Conclusion NAC provides certain noticeable benefits in attenuating substance craving and might help alleviate depressive symptoms and withdrawal syndrome. Precautious measures should be considered when using NAC although no difference in adverse effects was found between NAC treatment and control group.
Collapse
Affiliation(s)
- Chung-Ting Chang
- Department of Primary Medicine, Taipei Medical University Shuang Ho Hospital, New Taipei City, Taiwan
| | - Pei-Ju Hsieh
- Department of Medical Education, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan City, Taiwan
| | - Hsin-Chien Lee
- Department of Psychiatry, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Psychiatry, Taipei Medical University Hospital, Taipei, Taiwan
| | - Chun-Hong Lo
- Department of Primary Medicine, Taipei Medical University Shuang Ho Hospital, New Taipei City, Taiwan
| | - Ka-Wai Tam
- Center for Evidence-Based Health Care and Shared Decision Making Resource Center, Department of Medical Research, Taipei Medical University Shuang Ho Hospital, New Taipei City, Taiwan.,Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Cochrane Taiwan, Taipei Medical University, Taipei, Taiwan.,Division of General Surgery, Department of Surgery, Taipei Medical University Shuang Ho Hospital, New Taipei City, Taiwan.,Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - El-Wui Loh
- Center for Evidence-Based Health Care and Shared Decision Making Resource Center, Department of Medical Research, Taipei Medical University Shuang Ho Hospital, New Taipei City, Taiwan.,Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Cochrane Taiwan, Taipei Medical University, Taipei, Taiwan.,Department of Dentistry, Taipei Medical University Shuang Ho Hospital, New Taipei City, Taiwan
| |
Collapse
|
12
|
Cocaine use disorder: A look at metabotropic glutamate receptors and glutamate transporters. Pharmacol Ther 2021; 221:107797. [DOI: 10.1016/j.pharmthera.2020.107797] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 11/04/2020] [Indexed: 01/08/2023]
|
13
|
Huang G, Thompson SL, Taylor JR. MPEP Lowers Binge Drinking in Male and Female C57BL/6 Mice: Relationship with mGlu5/Homer2/Erk2 Signaling. Alcohol Clin Exp Res 2021; 45:732-742. [PMID: 33587295 PMCID: PMC8076072 DOI: 10.1111/acer.14576] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 02/10/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND Metabotropic glutamate receptor 5 (mGlu5) plays an important role in excessive alcohol use and the mGlu5/Homer2/Erk2 signaling pathway has been implicated in binge drinking. The mGlu5 negative allosteric modulator (NAM) 2-methyl-6-(phenylethynyl)pyridine hydrochloride (MPEP) has been shown to reduce binge drinking in male mice, but less is known about its effect on female mice. Here, we sought to determine whether sex differences exists in the effects of MPEP on binge drinking and whether they relate to changes in the MPEP mGlu5/Homer2/Erk2 signaling. METHODS We measured the dose-response effect of MPEP on alcohol consumption in male and female mice using the Drinking in the Dark (DID) paradigm to assess potential sex differences. To rule out possible confounds of MPEP on locomotion, we measured the effects of MPEP on locomotor activity and drinking simultaneously during DID. Lastly, to test whether MPEP-induced changes in alcohol consumption were related to changes in Homer2 or Erk2 expression, we performed qPCR using brain tissue acquired from mice that had undergone 7 days of DID. RESULTS 30 mg/kg MPEP reduced binge alcohol consumption across female and male mice, with no sex differences in the dose-response relationship. Locomotor activity did not mediate the effects of MPEP on alcohol intake, but activity correlated with alcohol intake independent of MPEP. MPEP did not change the expression of Homer2 and Erk2 mRNA in the bed nucleus of the stria terminalis (BNST) or nucleus accumbens in mice whose drinking was reduced by MPEP, relative to saline. There was a positive relationship between alcohol intake and Homer2 expression in the BNST. CONCLUSIONS MPEP reduced alcohol consumption during DID in male and female C57BL/6 mice but did not change Homer2/Erk2 expression. Locomotor activity did not mediate the effects of MPEP on alcohol intake, though it correlated with alcohol intake. Alcohol intake during DID predicted BNST Homer2 expression. These data provide support for the regulation of alcohol consumption by mGlu5 across sexes.
Collapse
Affiliation(s)
- Gan Huang
- Department of Psychiatry, Division of Molecular Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Summer L. Thompson
- Department of Psychiatry, Division of Molecular Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Jane R. Taylor
- Department of Psychiatry, Division of Molecular Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Department of Psychology, Yale University Graduate School of Arts and Sciences, New Haven, CT, USA
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
14
|
Bratek - Gerej E, Bronisz A, Ziembowicz A, Salinska E. Pretreatment with mGluR2 or mGluR3 Agonists Reduces Apoptosis Induced by Hypoxia-Ischemia in Neonatal Rat Brains. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8848015. [PMID: 33763176 PMCID: PMC7963909 DOI: 10.1155/2021/8848015] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 01/04/2021] [Accepted: 02/23/2021] [Indexed: 11/17/2022]
Abstract
Hypoxia-ischemia (HI) in an immature brain results in energy depletion and excessive glutamate release resulting in excitotoxicity and oxidative stress. An increase in reactive oxygen species (ROS) production induces apoptotic processes resulting in neuronal death. Activation of group II mGluR was shown to prevent neuronal damage after HI. The application of agonists of mGluR3 (N-acetylaspartylglutamate; NAAG) or mGluR2 (LY379268) inhibits the release of glutamate and reduces neurodegeneration in a neonatal rat model of HI, although the exact mechanism is not fully recognized. In the present study, the effects of NAAG (5 mg/kg) and LY379268 (5 mg/kg) application (24 h or 1 h before experimental birth asphyxia) on apoptotic processes as the potential mechanism of neuroprotection in 7-day-old rats were investigated. Intraperitoneal application of NAAG or LY379268 at either time point before HI significantly reduced the number of TUNEL-positive cells in the CA1 region of the ischemic brain hemisphere. Both agonists reduced expression of the proapoptotic Bax protein and increased expression of Bcl-2. Decreases in HI-induced caspase-9 and caspase-3 activity were also observed. Application of NAAG or LY379268 24 h or 1 h before HI reduced HIF-1α formation likely by reducing ROS levels. It was shown that LY379268 concentration remains at a level that is required for activation of mGluR2 for up to 24 h; however, NAAG is quickly metabolized by glutamate carboxypeptidase II (GCPII) into glutamate and N-acetyl-aspartate. The observed effect of LY379268 application 24 h or 1 h before HI is connected with direct activation of mGluR2 and inhibition of glutamate release. Based on the data presented in this study and on our previous findings, we conclude that the neuroprotective effect of NAAG applied 1 h before HI is most likely the result of a combination of mGluR3 and NMDA receptor activation, whereas the beneficial effects of NAAG pretreatment 24 h before HI can be explained by the activation of NMDA receptors and induction of the antioxidative/antiapoptotic defense system triggered by mild excitotoxicity in neurons. This response to NAAG pretreatment is consistent with the commonly accepted mechanism of preconditioning.
Collapse
Affiliation(s)
- Ewelina Bratek - Gerej
- Department of Neurochemistry, Mossakowski Medical Research Institute Polish Academy of Sciences, Warsaw, Poland
| | - Agnieszka Bronisz
- Tumor Microenvironment Laboratory, Mossakowski Medical Research Institute Polish Academy of Sciences, Warsaw, Poland
| | - Apolonia Ziembowicz
- Department of Neurochemistry, Mossakowski Medical Research Institute Polish Academy of Sciences, Warsaw, Poland
| | - Elzbieta Salinska
- Department of Neurochemistry, Mossakowski Medical Research Institute Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
15
|
Szpręgiel I, Wrońska D, Kmiecik M, Pałka S, Kania BF. Glutamic Acid Decarboxylase Concentration Changes in Response to Stress and Altered Availability of Glutamic Acid in Rabbit ( Oryctolagus cuniculus) Brain Limbic Structures. Animals (Basel) 2021; 11:455. [PMID: 33572286 PMCID: PMC7915518 DOI: 10.3390/ani11020455] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/31/2021] [Accepted: 02/04/2021] [Indexed: 12/18/2022] Open
Abstract
Glutamic acid decarboxylase (GAD) is an enzyme that catalyses the formation of γ-aminobutyric acid (GABA), the most important inhibitory neurotransmitter, from glutamic acid (Glu), which is considered the most important excitatory transmitter in the central and peripheral nervous systems. GAD is a key enzyme that provides a balance between Glu and GABA concentration. Hence, it can be assumed that if the GAD executes the synthesis of GABA from Glu, it is important in the stress response, and thus also in triggering the emotional states of the body that accompany stress. The aim of the study was to investigate the concentration of the GAD in motivational structures in the brain of the rabbit (Oryctolagus cuniculus) under altered homeostatic conditions caused by stress and variable availability of Glu. Summarising, the experimental results clearly showed variable concentrations of GAD in the motivational structures of the rabbit brain. The highest concentration of GAD was found in the hypothalamus, which suggests a strong effect of Glu and GABA on the activity of this brain structure. The GAD concentrations in individual experimental groups depended to a greater extent on blocking the activity of glutamate receptors than on the effects of a single stress exposure. The results obtained clearly support the possibility that a rapid change in the concentration of GAD could shift bodily responses to quickly achieve homeostasis, especially in this species. Further studies are necessary to reveal the role of the Glu-GAD-GABA system in the modulation of stress situations as well as in body homeostasis.
Collapse
Affiliation(s)
- Izabela Szpręgiel
- Department of Animal Physiology and Endocrinology, Faculty of Animal Sciences, University of Agriculture in Krakow, Al. Mickiewicza 24/28, 30-059 Kraków, Poland;
| | - Danuta Wrońska
- Department of Animal Physiology and Endocrinology, Faculty of Animal Sciences, University of Agriculture in Krakow, Al. Mickiewicza 24/28, 30-059 Kraków, Poland;
| | - Michał Kmiecik
- Department of Genetics, Animal Breeding and Ethology, Faculty of Animal Sciences, University of Agriculture in Krakow, Al. Mickiewicza 24/28, 30-059 Kraków, Poland; (M.K.); (S.P.)
| | - Sylwia Pałka
- Department of Genetics, Animal Breeding and Ethology, Faculty of Animal Sciences, University of Agriculture in Krakow, Al. Mickiewicza 24/28, 30-059 Kraków, Poland; (M.K.); (S.P.)
| | - Bogdan F. Kania
- University Centre of Veterinary Medicine JU-AU, University of Agriculture in Kraków, Mickiewicza 24/28, 30-059 Kraków, Poland;
| |
Collapse
|
16
|
Salisbury AJ, Blackwood CA, Cadet JL. Prolonged Withdrawal From Escalated Oxycodone Is Associated With Increased Expression of Glutamate Receptors in the Rat Hippocampus. Front Neurosci 2021; 14:617973. [PMID: 33536871 PMCID: PMC7848144 DOI: 10.3389/fnins.2020.617973] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 12/30/2020] [Indexed: 11/30/2022] Open
Abstract
People suffering from opioid use disorder (OUD) exhibit cognitive dysfunctions. Here, we investigated potential changes in the expression of glutamate receptors in rat hippocampi at 2 h and 31 days after the last session of oxycodone self-administration (SA). RNA extracted from the hippocampus was used in quantitative polymerase chain reaction analyses. Rats, given long-access (9 h per day) to oxycodone (LgA), took significantly more drug than rats exposed to short-access (3 h per day) (ShA). In addition, LgA rats could be further divided into higher oxycodone taking (LgA-H) or lower oxycodone taking (LgA-L) groups, based on a cut-off of 50 infusions per day. LgA rats, but not ShA, rats exhibited incubation of oxycodone craving. In addition, LgA rats showed increased mRNA expression of GluA1-3 and GluN2a-c subunits as well as Grm3, Grm5, Grm6, and Grm8 subtypes of glutamate receptors after 31 days but not after 2 h of stopping the SA experiment. Changes in GluA1-3, Grm6, and Grm8 mRNA levels also correlated with increased lever pressing (incubation) after long periods of withdrawal from oxycodone. More studies are needed to elucidate the molecular mechanisms involved in altering the expression of these receptors during withdrawal from oxycodone and/or incubation of drug seeking.
Collapse
Affiliation(s)
| | | | - Jean Lud Cadet
- National Institute on Drug Abuse, Molecular Neuropsychiatry Branch, National Institutes of Health, Baltimore, MD, United States
| |
Collapse
|
17
|
Murray CH, Christian DT, Milovanovic M, Loweth JA, Hwang EK, Caccamise AJ, Funke JR, Wolf ME. mGlu5 function in the nucleus accumbens core during the incubation of methamphetamine craving. Neuropharmacology 2021; 186:108452. [PMID: 33444640 DOI: 10.1016/j.neuropharm.2021.108452] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 01/02/2021] [Accepted: 01/06/2021] [Indexed: 12/11/2022]
Abstract
Many studies have demonstrated that negative allosteric modulators (NAM) of metabotropic glutamate receptor 5 (mGlu5) reduce cocaine and methamphetamine seeking in extinction-reinstatement animal models of addiction. Less is known about effects of mGlu5 NAMs in abstinence models, particularly for methamphetamine. We used the incubation of drug craving model, in which cue-induced craving progressively intensifies after withdrawal from drug self-administration, to conduct the first studies of the following aspects of mGlu5 function in the rat nucleus accumbens (NAc) core during abstinence from methamphetamine self-administration: 1) functionality of the major form of synaptic depression in NAc medium spiny neurons, which is induced postsynaptically via mGlu5 and expressed presynaptically via cannabinoid type 1 receptors (CB1Rs), 2) mGlu5 surface expression and physical associations between mGlu5, Homer proteins, and diacylglycerol lipase-α, and 3) the effect of systemic and intra-NAc core administration of the mGlu5 NAM 3-((2-methyl-4-)ethynyl)pyridine (MTEP) on expression of incubated methamphetamine craving. We found that mGlu5/CB1R-dependent synaptic depression was lost during the rising phase of methamphetamine incubation but then recovered, in contrast to its persistent impairment during the plateau phase of incubation of cocaine craving. Furthermore, whereas the cocaine-induced impairment was accompanied by reduced mGlu5 levels and mGlu5-Homer associations, this was not the case for methamphetamine. Systemic MTEP reduced incubated methamphetamine seeking, but also reduced inactive hole nose-pokes and locomotion, while intra-NAc core MTEP had no significant effects. These findings provide the first insight into the role of mGlu5 in the incubation of methamphetamine craving and reveal differences from incubation of cocaine craving.
Collapse
Affiliation(s)
- Conor H Murray
- Department of Neuroscience, The Chicago Medical School at Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL, 60064, USA; Department of Behavioral Neuroscience, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA.
| | - Daniel T Christian
- Department of Neuroscience, The Chicago Medical School at Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL, 60064, USA.
| | - Mike Milovanovic
- Department of Neuroscience, The Chicago Medical School at Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL, 60064, USA.
| | - Jessica A Loweth
- Department of Neuroscience, The Chicago Medical School at Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL, 60064, USA.
| | - Eun-Kyung Hwang
- Department of Behavioral Neuroscience, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA.
| | - Aaron J Caccamise
- Department of Neuroscience, The Chicago Medical School at Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL, 60064, USA.
| | - Jonathan R Funke
- Department of Behavioral Neuroscience, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA.
| | - Marina E Wolf
- Department of Neuroscience, The Chicago Medical School at Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL, 60064, USA; Department of Behavioral Neuroscience, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA.
| |
Collapse
|
18
|
Smart K, Nagano-Saito A, Milella MS, Sakae DY, Favier M, Vigneault E, Louie L, Hamilton A, Ferguson SSG, Rosa-Neto P, Narayanan S, El Mestikawy S, Leyton M, Benkelfat C. Metabotropic glutamate type 5 receptor binding availability during dextroamphetamine sensitization in mice and humans. J Psychiatry Neurosci 2021; 46:E1-E13. [PMID: 32559027 PMCID: PMC7955855 DOI: 10.1503/jpn.190162] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Glutamate transmission is implicated in drug-induced behavioural sensitization and the associated long-lasting increases in mesolimbic output. Metabotropic glutamate type 5 (mGlu5) receptors might be particularly important, but most details are poorly understood. METHODS We first assessed in mice (n = 51, all male) the effects of repeated dextroamphetamine administration (2.0 mg/kg, i.p.) on locomotor activity and binding of the mGlu5 ligand [3H]ABP688. In a parallel study, in 19 stimulant-drug-naïve healthy human volunteers (14 female) we administered 3 doses of dextroamphetamine (0.3 mg/kg, p.o.) or placebo, followed by a fourth dose 2 weeks later. We measured [11C]ABP688 binding using positron emission tomography before and after the induction phase. We assessed psychomotor and behavioural sensitization using speech rate, eye blink rate and self-report. We measured the localization of mGlu5 relative to synaptic markers in mouse striatum using immunofluorescence. RESULTS We observed amphetamine-induced psychomotor sensitization in mice and humans. We did not see group differences in mGlu5 availability following 3 pre-challenge amphetamine doses, but group differences did develop in mice administered 5 doses. In mice and humans, individual differences in mGlu5 binding after repeated amphetamine administration were negatively correlated with the extent of behavioural sensitization. In drug-naïve mice, mGlu5 was expressed at 67% of excitatory synapses on dendrites of striatal medium spiny neur. LIMITATIONS Correlational results should be interpreted as suggestive because of the limited sample size. We did not assess sex differences. CONCLUSION Together, these results suggest that changes in mGlu5 availability are not part of the earliest neural adaptations in stimulant-induced behavioural sensitization, but low mGlu5 binding might identify a higher propensity for sensitization.
Collapse
Affiliation(s)
- Kelly Smart
- From the Department of Psychiatry, McGill University, Montreal, Que. (Smart, Nagano-Saito, Milella, Sakae, Favier, Vigneault, Louie, Rosa-Neto, El Mestikawy, Leyton, Benkelfat); the Douglas Mental Health University Institute, McGill University, Montreal, Que. (Smart, Sakae, Favier, Vigneault, Rosa-Neto, El Mestikawy); the Department of Cellular and Molecular Medicine, University of Ottawa, Ont. (Hamilton, Ferguson); the McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Que. (Rosa-Neto, Narayanan, Leyton, Benkelfat); the Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Que. (Rosa-Neto, Narayanan, Leyton, Benkelfat); and the Center for Studies in Behavioral Neurobiology, Concordia University, Montreal, Que. (Leyton)
| | - Atsuko Nagano-Saito
- From the Department of Psychiatry, McGill University, Montreal, Que. (Smart, Nagano-Saito, Milella, Sakae, Favier, Vigneault, Louie, Rosa-Neto, El Mestikawy, Leyton, Benkelfat); the Douglas Mental Health University Institute, McGill University, Montreal, Que. (Smart, Sakae, Favier, Vigneault, Rosa-Neto, El Mestikawy); the Department of Cellular and Molecular Medicine, University of Ottawa, Ont. (Hamilton, Ferguson); the McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Que. (Rosa-Neto, Narayanan, Leyton, Benkelfat); the Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Que. (Rosa-Neto, Narayanan, Leyton, Benkelfat); and the Center for Studies in Behavioral Neurobiology, Concordia University, Montreal, Que. (Leyton)
| | - Michele S Milella
- From the Department of Psychiatry, McGill University, Montreal, Que. (Smart, Nagano-Saito, Milella, Sakae, Favier, Vigneault, Louie, Rosa-Neto, El Mestikawy, Leyton, Benkelfat); the Douglas Mental Health University Institute, McGill University, Montreal, Que. (Smart, Sakae, Favier, Vigneault, Rosa-Neto, El Mestikawy); the Department of Cellular and Molecular Medicine, University of Ottawa, Ont. (Hamilton, Ferguson); the McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Que. (Rosa-Neto, Narayanan, Leyton, Benkelfat); the Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Que. (Rosa-Neto, Narayanan, Leyton, Benkelfat); and the Center for Studies in Behavioral Neurobiology, Concordia University, Montreal, Que. (Leyton)
| | - Diana Yae Sakae
- From the Department of Psychiatry, McGill University, Montreal, Que. (Smart, Nagano-Saito, Milella, Sakae, Favier, Vigneault, Louie, Rosa-Neto, El Mestikawy, Leyton, Benkelfat); the Douglas Mental Health University Institute, McGill University, Montreal, Que. (Smart, Sakae, Favier, Vigneault, Rosa-Neto, El Mestikawy); the Department of Cellular and Molecular Medicine, University of Ottawa, Ont. (Hamilton, Ferguson); the McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Que. (Rosa-Neto, Narayanan, Leyton, Benkelfat); the Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Que. (Rosa-Neto, Narayanan, Leyton, Benkelfat); and the Center for Studies in Behavioral Neurobiology, Concordia University, Montreal, Que. (Leyton)
| | - Mathieu Favier
- From the Department of Psychiatry, McGill University, Montreal, Que. (Smart, Nagano-Saito, Milella, Sakae, Favier, Vigneault, Louie, Rosa-Neto, El Mestikawy, Leyton, Benkelfat); the Douglas Mental Health University Institute, McGill University, Montreal, Que. (Smart, Sakae, Favier, Vigneault, Rosa-Neto, El Mestikawy); the Department of Cellular and Molecular Medicine, University of Ottawa, Ont. (Hamilton, Ferguson); the McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Que. (Rosa-Neto, Narayanan, Leyton, Benkelfat); the Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Que. (Rosa-Neto, Narayanan, Leyton, Benkelfat); and the Center for Studies in Behavioral Neurobiology, Concordia University, Montreal, Que. (Leyton)
| | - Erika Vigneault
- From the Department of Psychiatry, McGill University, Montreal, Que. (Smart, Nagano-Saito, Milella, Sakae, Favier, Vigneault, Louie, Rosa-Neto, El Mestikawy, Leyton, Benkelfat); the Douglas Mental Health University Institute, McGill University, Montreal, Que. (Smart, Sakae, Favier, Vigneault, Rosa-Neto, El Mestikawy); the Department of Cellular and Molecular Medicine, University of Ottawa, Ont. (Hamilton, Ferguson); the McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Que. (Rosa-Neto, Narayanan, Leyton, Benkelfat); the Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Que. (Rosa-Neto, Narayanan, Leyton, Benkelfat); and the Center for Studies in Behavioral Neurobiology, Concordia University, Montreal, Que. (Leyton)
| | - Leanne Louie
- From the Department of Psychiatry, McGill University, Montreal, Que. (Smart, Nagano-Saito, Milella, Sakae, Favier, Vigneault, Louie, Rosa-Neto, El Mestikawy, Leyton, Benkelfat); the Douglas Mental Health University Institute, McGill University, Montreal, Que. (Smart, Sakae, Favier, Vigneault, Rosa-Neto, El Mestikawy); the Department of Cellular and Molecular Medicine, University of Ottawa, Ont. (Hamilton, Ferguson); the McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Que. (Rosa-Neto, Narayanan, Leyton, Benkelfat); the Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Que. (Rosa-Neto, Narayanan, Leyton, Benkelfat); and the Center for Studies in Behavioral Neurobiology, Concordia University, Montreal, Que. (Leyton)
| | - Alison Hamilton
- From the Department of Psychiatry, McGill University, Montreal, Que. (Smart, Nagano-Saito, Milella, Sakae, Favier, Vigneault, Louie, Rosa-Neto, El Mestikawy, Leyton, Benkelfat); the Douglas Mental Health University Institute, McGill University, Montreal, Que. (Smart, Sakae, Favier, Vigneault, Rosa-Neto, El Mestikawy); the Department of Cellular and Molecular Medicine, University of Ottawa, Ont. (Hamilton, Ferguson); the McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Que. (Rosa-Neto, Narayanan, Leyton, Benkelfat); the Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Que. (Rosa-Neto, Narayanan, Leyton, Benkelfat); and the Center for Studies in Behavioral Neurobiology, Concordia University, Montreal, Que. (Leyton)
| | - Stephen S G Ferguson
- From the Department of Psychiatry, McGill University, Montreal, Que. (Smart, Nagano-Saito, Milella, Sakae, Favier, Vigneault, Louie, Rosa-Neto, El Mestikawy, Leyton, Benkelfat); the Douglas Mental Health University Institute, McGill University, Montreal, Que. (Smart, Sakae, Favier, Vigneault, Rosa-Neto, El Mestikawy); the Department of Cellular and Molecular Medicine, University of Ottawa, Ont. (Hamilton, Ferguson); the McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Que. (Rosa-Neto, Narayanan, Leyton, Benkelfat); the Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Que. (Rosa-Neto, Narayanan, Leyton, Benkelfat); and the Center for Studies in Behavioral Neurobiology, Concordia University, Montreal, Que. (Leyton)
| | - Pedro Rosa-Neto
- From the Department of Psychiatry, McGill University, Montreal, Que. (Smart, Nagano-Saito, Milella, Sakae, Favier, Vigneault, Louie, Rosa-Neto, El Mestikawy, Leyton, Benkelfat); the Douglas Mental Health University Institute, McGill University, Montreal, Que. (Smart, Sakae, Favier, Vigneault, Rosa-Neto, El Mestikawy); the Department of Cellular and Molecular Medicine, University of Ottawa, Ont. (Hamilton, Ferguson); the McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Que. (Rosa-Neto, Narayanan, Leyton, Benkelfat); the Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Que. (Rosa-Neto, Narayanan, Leyton, Benkelfat); and the Center for Studies in Behavioral Neurobiology, Concordia University, Montreal, Que. (Leyton)
| | - Sridar Narayanan
- From the Department of Psychiatry, McGill University, Montreal, Que. (Smart, Nagano-Saito, Milella, Sakae, Favier, Vigneault, Louie, Rosa-Neto, El Mestikawy, Leyton, Benkelfat); the Douglas Mental Health University Institute, McGill University, Montreal, Que. (Smart, Sakae, Favier, Vigneault, Rosa-Neto, El Mestikawy); the Department of Cellular and Molecular Medicine, University of Ottawa, Ont. (Hamilton, Ferguson); the McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Que. (Rosa-Neto, Narayanan, Leyton, Benkelfat); the Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Que. (Rosa-Neto, Narayanan, Leyton, Benkelfat); and the Center for Studies in Behavioral Neurobiology, Concordia University, Montreal, Que. (Leyton)
| | - Salah El Mestikawy
- From the Department of Psychiatry, McGill University, Montreal, Que. (Smart, Nagano-Saito, Milella, Sakae, Favier, Vigneault, Louie, Rosa-Neto, El Mestikawy, Leyton, Benkelfat); the Douglas Mental Health University Institute, McGill University, Montreal, Que. (Smart, Sakae, Favier, Vigneault, Rosa-Neto, El Mestikawy); the Department of Cellular and Molecular Medicine, University of Ottawa, Ont. (Hamilton, Ferguson); the McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Que. (Rosa-Neto, Narayanan, Leyton, Benkelfat); the Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Que. (Rosa-Neto, Narayanan, Leyton, Benkelfat); and the Center for Studies in Behavioral Neurobiology, Concordia University, Montreal, Que. (Leyton)
| | - Marco Leyton
- From the Department of Psychiatry, McGill University, Montreal, Que. (Smart, Nagano-Saito, Milella, Sakae, Favier, Vigneault, Louie, Rosa-Neto, El Mestikawy, Leyton, Benkelfat); the Douglas Mental Health University Institute, McGill University, Montreal, Que. (Smart, Sakae, Favier, Vigneault, Rosa-Neto, El Mestikawy); the Department of Cellular and Molecular Medicine, University of Ottawa, Ont. (Hamilton, Ferguson); the McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Que. (Rosa-Neto, Narayanan, Leyton, Benkelfat); the Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Que. (Rosa-Neto, Narayanan, Leyton, Benkelfat); and the Center for Studies in Behavioral Neurobiology, Concordia University, Montreal, Que. (Leyton)
| | - Chawki Benkelfat
- From the Department of Psychiatry, McGill University, Montreal, Que. (Smart, Nagano-Saito, Milella, Sakae, Favier, Vigneault, Louie, Rosa-Neto, El Mestikawy, Leyton, Benkelfat); the Douglas Mental Health University Institute, McGill University, Montreal, Que. (Smart, Sakae, Favier, Vigneault, Rosa-Neto, El Mestikawy); the Department of Cellular and Molecular Medicine, University of Ottawa, Ont. (Hamilton, Ferguson); the McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Que. (Rosa-Neto, Narayanan, Leyton, Benkelfat); the Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Que. (Rosa-Neto, Narayanan, Leyton, Benkelfat); and the Center for Studies in Behavioral Neurobiology, Concordia University, Montreal, Que. (Leyton)
| |
Collapse
|
19
|
Thompson SL, Gianessi CA, O'Malley SS, Cavallo DA, Shi JM, Tetrault JM, DeMartini KS, Gueorguieva R, Pittman B, Krystal JH, Taylor JR, Krishnan-Sarin S. Saracatinib Fails to Reduce Alcohol-Seeking and Consumption in Mice and Human Participants. Front Psychiatry 2021; 12:709559. [PMID: 34531767 PMCID: PMC8438169 DOI: 10.3389/fpsyt.2021.709559] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 08/03/2021] [Indexed: 11/13/2022] Open
Abstract
More effective treatments to reduce pathological alcohol drinking are needed. The glutamatergic system and the NMDA receptor (NMDAR), in particular, are implicated in behavioral and molecular consequences of chronic alcohol use, making the NMDAR a promising target for novel pharmacotherapeutics. Ethanol exposure upregulates Fyn, a protein tyrosine kinase that indirectly modulates NMDAR signaling by phosphorylating the NR2B subunit. The Src/Fyn kinase inhibitor saracatinib (AZD0530) reduces ethanol self-administration and enhances extinction of goal-directed ethanol-seeking in mice. However, less is known regarding how saracatinib affects habitual ethanol-seeking. Moreover, no prior studies have assessed the effects of Src/Fyn kinase inhibitors on alcohol-seeking or consumption in human participants. Here, we tested the effects of saracatinib on alcohol consumption and craving/seeking in two species, including the first trial of an Src/Fyn kinase inhibitor to reduce drinking in humans. Eighteen male C57BL/6NCrl mice underwent operant conditioning on a variable interval schedule to induce habitual responding for 10% ethanol/0.1% saccharin. Next, mice received 5 mg/kg saracatinib or vehicle 2 h or 30 min prior to contingency degradation to measure habitual responding. In the human study, 50 non-treatment seeking human participants who drank heavily and met DSM-IV criteria for alcohol abuse or dependence were randomized to receive 125 mg/day saracatinib (n = 33) or placebo (n = 17). Alcohol Drinking Paradigms (ADP) were completed in a controlled research setting: before and after 7-8 days of treatment. Each ADP involved consumption of a priming drink of alcohol (0.03 mg%) followed by ad libitum access (3 h) to 12 additional drinks (0.015 g%); the number of drinks consumed and craving (Alcohol Urge Questionnaire) were recorded. In mice, saracatinib did not affect habitual ethanol seeking or consumption at either time point. In human participants, no significant effects of saracatinib on alcohol craving or consumption were identified. These results in mice and humans suggest that Fyn kinase inhibition using saracatinib, at the doses tested here, may not reduce alcohol consumption or craving/seeking among those habitually consuming alcohol, in contrast to reports of positive effects of saracatinib in individuals that seek ethanol in a goal-directed manner. Nevertheless, future studies should confirm these negative findings using additional doses and schedules of saracatinib administration.
Collapse
Affiliation(s)
- Summer L Thompson
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
| | - Carol A Gianessi
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States.,Interdepartmental Neuroscience Program, Yale University Graduate School of Arts and Sciences, New Haven, CT, United States
| | - Stephanie S O'Malley
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
| | - Dana A Cavallo
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
| | - Julia M Shi
- Program in Addiction Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Jeanette M Tetrault
- Program in Addiction Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Kelly S DeMartini
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
| | - Ralitza Gueorguieva
- Department of Biostatistics, Yale University School of Public Health, New Haven, CT, United States
| | - Brian Pittman
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
| | - John H Krystal
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States.,Department of Neuroscience, Yale University School of Medicine, New Haven, CT, United States
| | - Jane R Taylor
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States.,Department of Neuroscience, Yale University School of Medicine, New Haven, CT, United States.,Department of Psychology, Yale University, New Haven, CT, United States
| | | |
Collapse
|
20
|
Groman SM, Hillmer AT, Heather L, Fowles K, Holden D, Morris ED, Lee D, Taylor JR. Dysregulation of Decision Making Related to Metabotropic Glutamate 5, but Not Midbrain D 3, Receptor Availability Following Cocaine Self-administration in Rats. Biol Psychiatry 2020; 88:777-787. [PMID: 32826065 PMCID: PMC8935943 DOI: 10.1016/j.biopsych.2020.06.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/05/2020] [Accepted: 06/19/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND Compulsive patterns of drug use are thought to be the consequence of drug-induced adaptations in the neural mechanisms that enable behavior to be flexible. Neuroimaging studies have found evidence of robust alterations in glutamate and dopamine receptors within brain regions that are known to be critical for decision-making processes in cocaine-dependent individuals, and these changes have been argued to be the consequence of persistent drug use. The causal relationships among drug-induced alterations, cocaine taking, and maladaptive decision-making processes, however, are difficult to establish in humans. METHODS We assessed decision making in adult male rats using a probabilistic reversal learning task and used positron emission tomography with the [11C]-(+)-PHNO and [18F]FPEB radioligands to quantify regional dopamine D2/3 and metabotropic glutamate 5 (mGlu5) receptor availability, respectively, before and after 21 days of cocaine or saline self-administration. Tests of motivation and relapse-like behaviors were also conducted. RESULTS We found that self-administration of cocaine, but not of saline, disrupted behavior in the probabilistic reversal learning task measured by selective impairments in negative-outcome updating and also increased cortical mGlu5 receptor availability following 2 weeks of forced abstinence. D2/3 and, importantly, midbrain D3 receptor availability was not altered following 2 weeks of abstinence from cocaine. Notably, the degree of the cocaine-induced increase in cortical mGlu5 receptor availability was related to the degree of disruption in negative-outcome updating. CONCLUSIONS These findings suggest that cocaine-induced changes in mGlu5 signaling may be a mechanism by which disruptions in negative-outcome updating emerge in cocaine-dependent individuals.
Collapse
Affiliation(s)
- Stephanie M. Groman
- Department of Psychiatry Yale University,Correspondence should be addressed to: Stephanie M. Groman, Ph.D. (), Jane R. Taylor, Ph.D. (), 34 Park Street, New Haven CT 06515
| | - Ansel T. Hillmer
- Department of Psychiatry Yale University,Department of Radiology and Biomedical Imaging Yale University,Department of Yale Positron Emission Tomography Center Yale University
| | - Liu Heather
- Department of Radiology and Biomedical Imaging Yale University
| | - Krista Fowles
- Department of Yale Positron Emission Tomography Center Yale University
| | - Daniel Holden
- Department of Yale Positron Emission Tomography Center Yale University
| | - Evan D. Morris
- Department of Radiology and Biomedical Imaging Yale University,Department of Yale Positron Emission Tomography Center Yale University,Invicro LLC
| | - Daeyeol Lee
- The Zanvyl Krieger Mind/Brain Institute, The Solomon H Snyder Department of Neuroscience, Department of Psychological and Brain Sciences, Johns Hopkins University
| | - Jane R. Taylor
- Department of Psychiatry Yale University,Department of Neuroscience Yale University,Correspondence should be addressed to: Stephanie M. Groman, Ph.D. (), Jane R. Taylor, Ph.D. (), 34 Park Street, New Haven CT 06515
| |
Collapse
|
21
|
Spanagel R. Cannabinoids and the endocannabinoid system in reward processing and addiction: from mechanisms to interventions
. DIALOGUES IN CLINICAL NEUROSCIENCE 2020; 22:241-250. [PMID: 33162767 PMCID: PMC7605022 DOI: 10.31887/dcns.2020.22.3/rspanagel] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The last decades have seen a major gain in understanding the action of
cannabinoids and the endocannabinoid system in reward processing and the development of
addictive behavior. Cannabis-derived psychoactive compounds such as
Δ9-tetrahydrocannabinol and synthetic cannabinoids directly interact with the reward
system and thereby have addictive properties. Cannabinoids induce their reinforcing
properties by an increase in tonic dopamine levels through a cannabinoid type 1 (CB1)
receptor–dependent mechanism within the ventral tegmental area. Cues that are
conditioned to cannabis smoking can induce drug-seeking responses (ie, craving) by
eliciting phasic dopamine events. A dopamine-independent mechanism involved in
drug-seeking responses involves an endocannabinoid/glutamate interaction within the
corticostriatal part of the reward system. In conclusion, pharmacological blockade of
endocannabinoid signaling should lead to a reduction in drug craving and subsequently
should reduce relapse behavior in addicted individuals. Indeed, there is increasing
preclinical evidence that targeting the endocannabinoid system reduces craving and
relapse, and allosteric modulators at CB1 receptors and fatty acid amide hydrolase
inhibitors are in clinical development for cannabis use disorder. Cannabidiol, which
mainly acts on CB1 and CB2 receptors, is currently being tested in patients with alcohol
use disorder and opioid use disorder.
Collapse
Affiliation(s)
- Rainer Spanagel
- Institute of Psychopharmacology, Central Institute of Mental Health, Faculty of Medicine Mannheim, University of Heidelberg, Germany
| |
Collapse
|
22
|
Onaolapo AY, Onaolapo OJ. Dietary glutamate and the brain: In the footprints of a Jekyll and Hyde molecule. Neurotoxicology 2020; 80:93-104. [PMID: 32687843 DOI: 10.1016/j.neuro.2020.07.001] [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: 05/14/2020] [Revised: 06/29/2020] [Accepted: 07/14/2020] [Indexed: 12/15/2022]
Abstract
Glutamate is a crucial neurotransmitter of the mammalian central nervous system, a molecular component of our diet, and a popular food-additive. However, for decades, concerns have been raised about the issue of glutamate's safety as a food additive; especially, with regards to its ability (or otherwise) to cross the blood-brain barrier, cause excitotoxicity, or lead to neuron death. Results of animal studies following glutamate administration via different routes suggest that an array of effects can be observed. While some of the changes appear deleterious, some are not fully-understood, and the impact of others might even be beneficial. These observations suggest that with regards to the mammalian brain, exogenous glutamate might exert a double-sided effect, and in essence be a two-faced molecule whose effects may be dependent on several factors. This review draws from the research experiences of the authors and other researchers regarding the effects of exogenous glutamate on the brain of rodents. We also highlight the possible implications of such effects on the brain, in health and disease. Finally, we deduce that beyond the culinary effects of exogenous glutamate, there is the possibility of a beneficial role in the understanding and management of brain disorders.
Collapse
Affiliation(s)
- Adejoke Y Onaolapo
- Behavioural Neuroscience/Neurobiology Unit, Department of Anatomy, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria.
| | - Olakunle J Onaolapo
- Behavioural Neuroscience/Neuropharmacology Unit, Department of Pharmacology, Ladoke Akintola University of Technology, Osogbo, Osun State, Nigeria.
| |
Collapse
|
23
|
Alasmari F, Alhaddad H, Wong W, Bell RL, Sari Y. Ampicillin/Sulbactam Treatment Modulates NMDA Receptor NR2B Subunit and Attenuates Neuroinflammation and Alcohol Intake in Male High Alcohol Drinking Rats. Biomolecules 2020; 10:biom10071030. [PMID: 32664441 PMCID: PMC7407831 DOI: 10.3390/biom10071030] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 07/06/2020] [Accepted: 07/09/2020] [Indexed: 12/21/2022] Open
Abstract
Exposure to ethanol commonly manifests neuroinflammation. Beta (β)-lactam antibiotics attenuate ethanol drinking through upregulation of astroglial glutamate transporters, especially glutamate transporter-1 (GLT-1), in the mesocorticolimbic brain regions, including the nucleus accumbens (Acb). However, the effect of β-lactam antibiotics on neuroinflammation in animals chronically exposed to ethanol has not been fully investigated. In this study, we evaluated the effects of ampicillin/sulbactam (AMP/SUL, 100 and 200 mg/kg, i.p.) on ethanol consumption in high alcohol drinking (HAD1) rats. Additionally, we investigated the effects of AMP/SUL on GLT-1 and N-methyl-d-aspartate (NMDA) receptor subtypes (NR2A and NR2B) in the Acb core (AcbCo) and Acb shell (AcbSh). We found that AMP/SUL at both doses attenuated ethanol consumption and restored ethanol-decreased GLT-1 and NR2B expression in the AcbSh and AcbCo, respectively. Moreover, AMP/SUL (200 mg/kg, i.p.) reduced ethanol-increased high mobility group box 1 (HMGB1) and receptor for advanced glycation end-products (RAGE) expression in the AcbSh. Moreover, both doses of AMP/SUL attenuated ethanol-elevated tumor necrosis factor-alpha (TNF-α) in the AcbSh. Our results suggest that AMP/SUL attenuates ethanol drinking and modulates NMDA receptor NR2B subunits and HMGB1-associated pathways.
Collapse
Affiliation(s)
- Fawaz Alasmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia;
- Department of Pharmacology and Experimental Therapeutics, University of Toledo, College of Pharmacy and Pharmaceutical Sciences, Toledo, OH 43614, USA; (H.A.); (W.W.)
| | - Hasan Alhaddad
- Department of Pharmacology and Experimental Therapeutics, University of Toledo, College of Pharmacy and Pharmaceutical Sciences, Toledo, OH 43614, USA; (H.A.); (W.W.)
| | - Woonyen Wong
- Department of Pharmacology and Experimental Therapeutics, University of Toledo, College of Pharmacy and Pharmaceutical Sciences, Toledo, OH 43614, USA; (H.A.); (W.W.)
| | - Richard L. Bell
- Department of Psychiatry and Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Correspondence: (R.L.B.); (Y.S.); Tel.: +317-278-8407 (R.L.B.); +419-383-1507 (Y.S.)
| | - Youssef Sari
- Department of Pharmacology and Experimental Therapeutics, University of Toledo, College of Pharmacy and Pharmaceutical Sciences, Toledo, OH 43614, USA; (H.A.); (W.W.)
- Correspondence: (R.L.B.); (Y.S.); Tel.: +317-278-8407 (R.L.B.); +419-383-1507 (Y.S.)
| |
Collapse
|
24
|
Kasten CR, Carzoli KL, Sharfman NM, Henderson T, Holmgren EB, Lerner MR, Miller MC, Wills TA. Adolescent alcohol exposure produces sex differences in negative affect-like behavior and group I mGluR BNST plasticity. Neuropsychopharmacology 2020; 45:1306-1315. [PMID: 32268346 PMCID: PMC7297734 DOI: 10.1038/s41386-020-0670-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 03/27/2020] [Accepted: 03/31/2020] [Indexed: 12/13/2022]
Abstract
Adolescent alcohol exposure increases the risk of developing alcohol use disorders (AUDs), yet the mechanisms responsible for this vulnerability remain largely unknown. One potential target for alcohol-induced changes is the circuitry that modulates negative affect and stress, two sexually dependent drivers of alcohol relapse. The bed nucleus of the stria terminalis (BNST) is a sexually dimorphic region that critically regulates negative affective- and stress-induced relapse. Group I metabotropic glutamate receptors (mGluR) are a target of interest due to their regulation of stress, anxiety behaviors, and BNST plasticity. The current studies investigate sex-dependent sensitivity to the effects of adolescent intermittent ethanol vapor exposure (AIE) on negative affect during acute and protracted alcohol withdrawal and following stress in adulthood. This work also assessed whether BNST group I mGluR-mediated long-term depression (LTD) was disrupted at these timepoints. During acute withdrawal, AIE altered LTD induced by the group I mGluR antagonist DHPG in females, but not males. During adulthood, stress unmasked persistent changes in DHPG-induced LTD and behavior that were not present under basal conditions. Females with an AIE history demonstrated enhanced negative affective-like behavior in the novelty-induced hypophagia test following restraint stress-a phenotype that could be blocked with systemic mGluR5 allosteric antagonism via MTEP. Conversely, males with an AIE history demonstrated elevated freezing in a contextual fear conditioning paradigm. These studies demonstrate long-lasting, sex-dependent phenotypes produced by AIE and suggest pharmaceutical interventions for alcohol use and comorbid disorders may be more effective if designed with sex differences in mind.
Collapse
Affiliation(s)
- C R Kasten
- Department of Cell Biology and Anatomy, LSU Health Sciences Center New Orleans, New Orleans, LA, USA
| | - K L Carzoli
- Department of Cell Biology and Anatomy, LSU Health Sciences Center New Orleans, New Orleans, LA, USA
| | - N M Sharfman
- Department of Cell Biology and Anatomy, LSU Health Sciences Center New Orleans, New Orleans, LA, USA
| | - T Henderson
- Department of Cell Biology and Anatomy, LSU Health Sciences Center New Orleans, New Orleans, LA, USA
| | - E B Holmgren
- Department of Cell Biology and Anatomy, LSU Health Sciences Center New Orleans, New Orleans, LA, USA
| | - M R Lerner
- Department of Cell Biology and Anatomy, LSU Health Sciences Center New Orleans, New Orleans, LA, USA
| | - M C Miller
- Department of Cell Biology and Anatomy, LSU Health Sciences Center New Orleans, New Orleans, LA, USA
| | - T A Wills
- Department of Cell Biology and Anatomy, LSU Health Sciences Center New Orleans, New Orleans, LA, USA.
- Neuroscience Center of Excellence, LSU Health Sciences Center New Orleans, New Orleans, LA, USA.
| |
Collapse
|
25
|
Abstract
Bulimia nervosa (BN) shares central features with substance-related and addictive disorders. The metabotropic glutamate receptor subtype 5 (mGlu5) plays an important role in addiction. Based on similarities between binge eating and substance-related and addictive disorders, we investigated mGlu5 in vivo in 15 female subjects with BN and 15 matched controls. We measured mGlu5 distribution volume ratio (DVR) with positron emission tomography (PET) using [11 C]ABP688. In BN mGlu5 DVR was higher in the anterior cingulate cortex (ACC), subgenual prefrontal cortex, and straight gyrus (p < 0.05). In BN, higher mGlu5 DVR in various brain regions, including ACC, pallidum, putamen, and caudate, positively correlated with "maturity fears" as assessed using the Eating Disorder Inventory-2 (p < 0.05). In BN and controls, smokers had globally decreased mGlu5 DVR. We present the first evidence for increased mGlu5 DVR in BN. Our findings suggest that pharmacological agents inhibiting mGlu5 might have a therapeutic potential in BN.
Collapse
|
26
|
Bilbao A, Neuhofer D, Sepers M, Wei SP, Eisenhardt M, Hertle S, Lassalle O, Ramos-Uriarte A, Puente N, Lerner R, Thomazeau A, Grandes P, Lutz B, Manzoni OJ, Spanagel R. Endocannabinoid LTD in Accumbal D1 Neurons Mediates Reward-Seeking Behavior. iScience 2020; 23:100951. [PMID: 32179475 PMCID: PMC7068121 DOI: 10.1016/j.isci.2020.100951] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 08/15/2019] [Accepted: 02/24/2020] [Indexed: 11/30/2022] Open
Abstract
The nucleus accumbens (NAc) plays a key role in drug-related behavior and natural reward learning. Synaptic plasticity in dopamine D1 and D2 receptor medium spiny neurons (MSNs) of the NAc and the endogenous cannabinoid (eCB) system have been implicated in reward seeking. However, the precise molecular and physiological basis of reward-seeking behavior remains unknown. We found that the specific deletion of metabotropic glutamate receptor 5 (mGluR5) in D1-expressing MSNs (D1miRmGluR5 mice) abolishes eCB-mediated long-term depression (LTD) and prevents the expression of drug (cocaine and ethanol), natural reward (saccharin), and brain-stimulation-seeking behavior. In vivo enhancement of 2-arachidonoylglycerol (2-AG) eCB signaling within the NAc core restores both eCB-LTD and reward-seeking behavior in D1miRmGluR5 mice. The data suggest a model where the eCB and glutamatergic systems of the NAc act in concert to mediate reward-seeking responses. mGluR5-D1-CB1-induced eCB-LTD mediates drugs of abuse and natural reward seeking eCB-LTD in D2-MSNs plays no important role in processing of reward-seeking responses Loss of eCB-LTD is a consequence of higher MAGL activity and lower CB1R expression Acute drug administration stops craving for alternative rewards on following days
Collapse
Affiliation(s)
- Ainhoa Bilbao
- Behavioral Genetics Research Group, Heidelberg University, Medical Faculty Mannheim, 68159 Mannheim, Germany; Institute of Psychopharmacology, Central Institute of Mental Health, Heidelberg University, Medical Faculty Mannheim, 68159 Mannheim, Germany.
| | - Daniela Neuhofer
- INSERM U1249, Parc Scientifique de Luminy - BP 13 - 13273, Marseille Cedex 09, France; Aix-Marseille University, Jardindu Pharo, 58 Boulevard Charles Livon, Marseille, 13007, France
| | - Marja Sepers
- INSERM U1249, Parc Scientifique de Luminy - BP 13 - 13273, Marseille Cedex 09, France; Aix-Marseille University, Jardindu Pharo, 58 Boulevard Charles Livon, Marseille, 13007, France
| | - Shou-Peng Wei
- Behavioral Genetics Research Group, Heidelberg University, Medical Faculty Mannheim, 68159 Mannheim, Germany; Institute of Psychopharmacology, Central Institute of Mental Health, Heidelberg University, Medical Faculty Mannheim, 68159 Mannheim, Germany
| | - Manuela Eisenhardt
- Behavioral Genetics Research Group, Heidelberg University, Medical Faculty Mannheim, 68159 Mannheim, Germany; Institute of Psychopharmacology, Central Institute of Mental Health, Heidelberg University, Medical Faculty Mannheim, 68159 Mannheim, Germany
| | - Sarah Hertle
- Behavioral Genetics Research Group, Heidelberg University, Medical Faculty Mannheim, 68159 Mannheim, Germany; Institute of Psychopharmacology, Central Institute of Mental Health, Heidelberg University, Medical Faculty Mannheim, 68159 Mannheim, Germany
| | - Olivier Lassalle
- INSERM U1249, Parc Scientifique de Luminy - BP 13 - 13273, Marseille Cedex 09, France; Aix-Marseille University, Jardindu Pharo, 58 Boulevard Charles Livon, Marseille, 13007, France; Cannalab, Cannabinoids Neuroscience Research International Associated Laboratory, INSERM-Indiana University, 107 S Indiana Avenue, Bloomington, IN 47405, USA
| | - Almudena Ramos-Uriarte
- Department of Neurosciences, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, Barrio Sarriena s/n, 48940 Leioa, Spain; Achucarro Basque Center for Neuroscience, Science Park of the UPV/EHU, Barrio Sarriena s/n, 48940 Leioa, Spain
| | - Nagore Puente
- Department of Neurosciences, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, Barrio Sarriena s/n, 48940 Leioa, Spain; Achucarro Basque Center for Neuroscience, Science Park of the UPV/EHU, Barrio Sarriena s/n, 48940 Leioa, Spain
| | - Raissa Lerner
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg, University Mainz, Duesbergweg 6, 55099 Mainz, Germany
| | - Aurore Thomazeau
- INSERM U1249, Parc Scientifique de Luminy - BP 13 - 13273, Marseille Cedex 09, France; Aix-Marseille University, Jardindu Pharo, 58 Boulevard Charles Livon, Marseille, 13007, France
| | - Pedro Grandes
- Department of Neurosciences, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, Barrio Sarriena s/n, 48940 Leioa, Spain; Achucarro Basque Center for Neuroscience, Science Park of the UPV/EHU, Barrio Sarriena s/n, 48940 Leioa, Spain
| | - Beat Lutz
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg, University Mainz, Duesbergweg 6, 55099 Mainz, Germany
| | - Olivier J Manzoni
- INSERM U1249, Parc Scientifique de Luminy - BP 13 - 13273, Marseille Cedex 09, France; Aix-Marseille University, Jardindu Pharo, 58 Boulevard Charles Livon, Marseille, 13007, France; Cannalab, Cannabinoids Neuroscience Research International Associated Laboratory, INSERM-Indiana University, 107 S Indiana Avenue, Bloomington, IN 47405, USA.
| | - Rainer Spanagel
- Institute of Psychopharmacology, Central Institute of Mental Health, Heidelberg University, Medical Faculty Mannheim, 68159 Mannheim, Germany.
| |
Collapse
|
27
|
Gobin C, Schwendt M. The cognitive cost of reducing relapse to cocaine-seeking with mGlu5 allosteric modulators. Psychopharmacology (Berl) 2020; 237:115-125. [PMID: 31446451 DOI: 10.1007/s00213-019-05351-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 08/09/2019] [Indexed: 12/24/2022]
Abstract
RATIONALE Cocaine use disorder (CUD) remains difficult to treat with no FDA-approved medications to reduce relapse. Antagonism of metabotropic glutamate receptor 5 (mGlu5) has been demonstrated to decrease cocaine-seeking but may also further compromise cognitive function in long-term cocaine users. OBJECTIVES Here we assessed the effect of repeated administration of negative or positive allosteric modulators (NAM or PAM) of mGlu5 on both cognitive performance and (context+cue)-primed cocaine-seeking after prolonged abstinence (≥ 45 days). METHODS Adult male Sprague-Dawley rats underwent 6 days of short-access (1 h/day) and 12 days of long-access (6 h/day) cocaine self-administration. Rats were then trained and tested in a delayed match-to-sample (DMS) task to establish baseline working memory performance over a 5-day block of testing. Next, rats received daily systemic administration of the mGlu5 NAM 3-((2-methyl-1,3-thiazol-4-yl)ethynyl)pyridine hydrochloride (MTEP; 3 mg/kg), the mGlu5 PAM 3-cyano-N-(1,3-diphenyl-1H-pyrazol-5-yl)benzamide (CDPPB; 30 mg/kg) or vehicle prior to DMS testing during a block of 5 days, followed by a 5-day washout DMS testing block. RESULTS MTEP and CDPPB decreased drug-seeking in response to cocaine-associated cues after prolonged abstinence. However, repeated treatment with MTEP impaired working memory, while CDPPB had no effects on performance. CONCLUSIONS These results emphasize the relevance of evaluating cognitive function within the context of investigating pharmacotherapies to treat CUD. Further research is needed to determine how two mechanistically different pharmacological compounds can exert the same behavioral effects to reduce cocaine-seeking.
Collapse
Affiliation(s)
- Christina Gobin
- Psychology Department, University of Florida, 114 Psychology Building, 945 Center Drive, Gainesville, FL, 32611-2250, USA
- Center for Addiction Research and Education (CARE), University of Florida, Gainesville, FL, USA
| | - Marek Schwendt
- Psychology Department, University of Florida, 114 Psychology Building, 945 Center Drive, Gainesville, FL, 32611-2250, USA.
- Center for Addiction Research and Education (CARE), University of Florida, Gainesville, FL, USA.
| |
Collapse
|
28
|
Giacometti LL, Barker JM. Comorbid HIV infection and alcohol use disorders: Converging glutamatergic and dopaminergic mechanisms underlying neurocognitive dysfunction. Brain Res 2019; 1723:146390. [PMID: 31421128 PMCID: PMC6766419 DOI: 10.1016/j.brainres.2019.146390] [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: 06/10/2019] [Revised: 08/02/2019] [Accepted: 08/13/2019] [Indexed: 10/26/2022]
Abstract
Alcohol use disorders (AUDs) are highly comorbid with human immunodeficiency virus (HIV) infection, occurring at nearly twice the rate in HIV positive individuals as in the general population. Individuals with HIV who consume alcohol show worse long-term prognoses and may be at elevated risk for the development of HIV-associated neurocognitive disorders. The direction of this relationship is unclear, and likely multifactorial. Chronic alcohol exposure and HIV infection independently promote cognitive dysfunction and further may interact to exacerbate neurocognitive deficits through effects on common targets, including corticostriatal glutamate and dopamine neurotransmission. Additionally, drug and alcohol use is likely to reduce treatment adherence, potentially resulting in accelerated disease progression and subsequent neurocognitive impairment. The development of neurocognitive impairments may further reduce cognitive control over behavior, resulting in escalating alcohol use. This review will examine the complex relationship between HIV infection and alcohol use, highlighting impacts on dopamine and glutamate systems by which alcohol use and HIV act independently and in tandem to alter corticostriatal circuit structure and function to dysregulate cognitive function.
Collapse
Affiliation(s)
- Laura L Giacometti
- Department of Pharmacology and Physiology, Drexel University College of Medicine, United States
| | - Jacqueline M Barker
- Department of Pharmacology and Physiology, Drexel University College of Medicine, United States.
| |
Collapse
|
29
|
Müller Herde A, Mihov Y, Krämer SD, Mu L, Adamantidis A, Ametamey SM, Hasler G. Chronic Nicotine Exposure Alters Metabotropic Glutamate Receptor 5: Longitudinal PET Study and Behavioural Assessment in Rats. Neurotox Res 2019; 36:806-816. [PMID: 31119680 DOI: 10.1007/s12640-019-00055-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 03/22/2019] [Accepted: 04/26/2019] [Indexed: 12/13/2022]
Abstract
Using positron emission tomography (PET), a profound alteration of the metabotropic glutamate receptor 5 (mGluR5) was found in human smoking addiction and abstinence. As human PET data either reflect the impact of chronic nicotine exposure or a pre-existing vulnerability to nicotine addiction, we designed a preclinical, longitudinal study to investigate the effect of chronic nicotine exposure on mGluR5 with the novel radiotracer [18F]PSS232 using PET. Twelve male dark Agouti rats at the age of 6 weeks were assigned randomly to three groups. From day 0 to day 250 the groups received 0 mg/L, 4 mg/L, or 8 mg/L nicotine solution in the drinking water. From day 250 to 320 all groups received nicotine-free drinking water. PET scans with [18F]PSS232 were performed in all animals on days 0, 250, and 320. To assess locomotion, seven tests in square open field arenas were carried out 72 days after the last PET scan. During the first four tests, rats received 0 mg/L nicotine and for the last three tests 4 mg/L nicotine in the drinking water. After 250 days of nicotine consumption [18F]PSS232 binding was reduced in the striatum, hippocampus, thalamus, and midbrain. At day 320, after nicotine withdrawal, [18F]PSS232 binding increased. These effects were more pronounced in the 4 mg/L nicotine group. Chronic administration of nicotine through the drinking water reduced exploratory behaviour. This preliminary longitudinal PET study demonstrates that chronic nicotine administration alters behaviour and mGluR5 availability. Chronic nicotine administration leads to decreased [18F]PSS232 binding which normalizes after prolonged nicotine withdrawal.
Collapse
Affiliation(s)
- Adrienne Müller Herde
- Center for Radiopharmaceutical Sciences of ETH, PSI, and USZ, Department of Chemistry and Applied Biosciences of ETH, 8093, Zurich, Switzerland
| | - Yoan Mihov
- Translational Research Center, University Hospital of Psychiatry, University of Bern, 3000, Bern 60, Switzerland
| | - Stefanie D Krämer
- Center for Radiopharmaceutical Sciences of ETH, PSI, and USZ, Department of Chemistry and Applied Biosciences of ETH, 8093, Zurich, Switzerland
| | - Linjing Mu
- Department of Nuclear Medicine, University Hospital Zurich, Center for Radiopharmaceutical Sciences of ETH, PSI, and USZ, 8091, Zurich, Switzerland
| | - Antoine Adamantidis
- Department of Biomedical Research, Inselspital University Hospital, University of Bern, 3000, Bern, Switzerland
- Centre for Experimental Neurology, Department of Neurology, Inselspital University Hospital, University of Bern, Bern, Switzerland
| | - Simon M Ametamey
- Center for Radiopharmaceutical Sciences of ETH, PSI, and USZ, Department of Chemistry and Applied Biosciences of ETH, 8093, Zurich, Switzerland
| | - Gregor Hasler
- Psychiatry Research Unit, University of Fribourg, Chemin du Cardinal-Journet 3, Villars-sur-Glâne, 1752, Fribourg, Switzerland.
| |
Collapse
|
30
|
Kasten CR, Holmgren EB, Wills TA. Metabotropic Glutamate Receptor Subtype 5 in Alcohol-Induced Negative Affect. Brain Sci 2019; 9:E183. [PMID: 31366097 PMCID: PMC6721373 DOI: 10.3390/brainsci9080183] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 07/26/2019] [Accepted: 07/28/2019] [Indexed: 02/07/2023] Open
Abstract
Allosteric modulators of metabotropic glutamate 5 receptors (mGlu5 receptors) have been identified as a promising treatment to independently alleviate both negative affective states and ethanol-seeking and intake. However, these conditions are often comorbid and might precipitate one another. Acute and protracted ethanol withdrawal can lead to negative affective states. In turn, these states are primary drivers of alcohol relapse, particularly among women. The current review synthesizes preclinical studies that have observed the role of mGlu5 receptor modulation in negative affective states following ethanol exposure. The primary behavioral assays discussed are ethanol-seeking and intake, development and extinction of ethanol-associated cues and contexts, behavioral despair, and anxiety-like activity. The work done to-date supports mGlu5 receptor modulation as a promising target for mediating negative affective states to reduce ethanol intake or prevent relapse. Limitations in interpreting these data include the lack of models that use alcohol-dependent animals, limited use of adolescent and female subjects, and a lack of comprehensive evaluations of negative affective-like behavior.
Collapse
Affiliation(s)
- Chelsea R Kasten
- LSU Health Sciences Center-New Orleans, Department of Cell Biology and Anatomy, Medical Education Building, 1901 Perdido Street, Room 6103, New Orleans, LA 70112, USA
| | - Eleanor B Holmgren
- LSU Health Sciences Center-New Orleans, Department of Cell Biology and Anatomy, Medical Education Building, 1901 Perdido Street, Room 6103, New Orleans, LA 70112, USA
| | - Tiffany A Wills
- LSU Health Sciences Center-New Orleans, Department of Cell Biology and Anatomy, Medical Education Building, 1901 Perdido Street, Room 6103, New Orleans, LA 70112, USA.
| |
Collapse
|
31
|
Morató X, Luján R, Gonçalves N, Watanabe M, Altafaj X, Carvalho AL, Fernández-Dueñas V, Cunha RA, Ciruela F. Metabotropic glutamate type 5 receptor requires contactin-associated protein 1 to control memory formation. Hum Mol Genet 2019; 27:3528-3541. [PMID: 30010864 DOI: 10.1093/hmg/ddy264] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 07/09/2018] [Indexed: 12/31/2022] Open
Abstract
The hippocampus is a key brain region for memory formation. Metabotropic glutamate type 5 receptors (mGlu5R) are strongly expressed in CA1 pyramidal neurons and fine-tune synaptic plasticity. Accordingly, mGlu5R pharmacological manipulation may represent an attractive therapeutic strategy to manage hippocampal-related neurological disorders. Here, by means of a membrane yeast two-hybrid screening, we identified contactin-associated protein 1 (Caspr1), a type I transmembrane protein member of the neurexin family, as a new mGlu5R partner. We report that mGlu5R and Caspr1 co-distribute and co-assemble both in heterologous expression systems and in rat brain. Furthermore, downregulation of Caspr1 in rat hippocampal primary cultures decreased mGlu5R-mediated signaling. Finally, silencing Caspr1 expression in the hippocampus impaired the impact of mGlu5R on spatial memory. Our results indicate that Caspr1 plays a pivotal role controlling mGlu5R function in hippocampus-dependent memory formation. Hence, this new protein-protein interaction may represent novel target for neurological disorders affecting hippocampal glutamatergic neurotransmission.
Collapse
Affiliation(s)
- Xavier Morató
- Unitat de Farmacologia, Departament Patologia i Terapèutica Experimental, Facultat de Medicina, IDIBELL, Universitat de Barcelona, L'Hospitalet de Llobregat, Spain.,Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
| | - Rafael Luján
- IDINE, Departamento de Ciencias Médicas, Facultad de Medicina, Universidad Castilla-La Mancha, Albacete, Spain
| | - Nélio Gonçalves
- CNC-Center for Neurosciences and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Masahiko Watanabe
- Department of Anatomy, Hokkaido University School of Medicine, Sapporo, Japan
| | - Xavier Altafaj
- Unitat de Farmacologia, Departament Patologia i Terapèutica Experimental, Facultat de Medicina, IDIBELL, Universitat de Barcelona, L'Hospitalet de Llobregat, Spain
| | - Ana Luísa Carvalho
- CNC-Center for Neurosciences and Cell Biology, University of Coimbra, Coimbra, Portugal.,Department of Life Sciences, University of Coimbra, Coimbra, Portugal
| | - Víctor Fernández-Dueñas
- Unitat de Farmacologia, Departament Patologia i Terapèutica Experimental, Facultat de Medicina, IDIBELL, Universitat de Barcelona, L'Hospitalet de Llobregat, Spain.,Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
| | - Rodrigo A Cunha
- CNC-Center for Neurosciences and Cell Biology, University of Coimbra, Coimbra, Portugal.,Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Francisco Ciruela
- Unitat de Farmacologia, Departament Patologia i Terapèutica Experimental, Facultat de Medicina, IDIBELL, Universitat de Barcelona, L'Hospitalet de Llobregat, Spain.,Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
32
|
Drug addiction: a curable mental disorder? Acta Pharmacol Sin 2018; 39:1823-1829. [PMID: 30382181 DOI: 10.1038/s41401-018-0180-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 09/29/2018] [Indexed: 02/06/2023] Open
Abstract
Drug addiction is a chronic, relapsing brain disorder. Multiple neural networks in the brain including the reward system (e.g., the mesocorticolimbic system), the anti-reward/stress system (e.g., the extended amygdala), and the central immune system, are involved in the development of drug addiction and relapse after withdrawal from drugs of abuse. Preclinical and clinical studies have demonstrated that it is promising to control drug addiction by pharmacologically targeting the addiction-related systems in the brain. Here we review the pharmacological targets within the dopamine system, glutamate system, trace amine system, anti-reward system, and central immune system, which are of clinical interests. Furthermore, we discuss other potential therapies, e.g., brain stimulation, behavioral treatments, and therapeutic gene modulation, which could be effective to treat drug addiction. We conclude that, although drug addiction is a complex disorder that involves complicated neural mechanisms and psychological processes, this mental disorder is treatable and may be curable by therapies such as gene modulation in the future.
Collapse
|
33
|
Gross KS, Moore KM, Meisel RL, Mermelstein PG. mGluR5 Mediates Dihydrotestosterone-Induced Nucleus Accumbens Structural Plasticity, but Not Conditioned Reward. Front Neurosci 2018; 12:855. [PMID: 30515075 PMCID: PMC6255826 DOI: 10.3389/fnins.2018.00855] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 11/01/2018] [Indexed: 12/12/2022] Open
Abstract
Gonadal hormones play a vital role in driving motivated behavior. They not only modulate responses to naturally rewarding stimuli, but also influence responses to drugs of abuse. A commonality between gonadal hormones and drugs of abuse is that they both impact the neurocircuitry of reward, including the regulation of structural plasticity in the nucleus accumbens (NAc). Previous hormonal studies have focused on the mechanisms and behavioral correlates of estradiol-induced dendritic spine changes in the female NAc. Here we sought to determine the effects of androgens on medium spiny neuron (MSN) spine plasticity in the male NAc. Following treatment with the androgen receptor agonist dihydrotestosterone (DHT), MSNs in castrated male rats exhibited a significant decrease in dendritic spine density. This effect was isolated to the shell subregion of the NAc. The effect of DHT was dependent on mGluR5 activity, and local mGluR5 activation and subsequent endocannabinoid signaling produce an analogous NAc shell spine decrease. Somewhat surprisingly, DHT-induced conditioned place preference remained intact following systemic inhibition of mGluR5. These findings indicate that androgens can utilize mGluR signaling, similar to estrogens, to mediate changes in NAc dendritic structure. In addition, there are notable differences in the direction of spine changes, and site specificity of estrogen and androgen action, suggesting sex differences in the hormonal regulation of motivated behaviors.
Collapse
Affiliation(s)
- Kellie S Gross
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, United States.,Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | - Kelsey M Moore
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, United States.,Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | - Robert L Meisel
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, United States.,Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | - Paul G Mermelstein
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, United States.,Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
34
|
Simmons SJ, Leyrer-Jackson JM, Oliver CF, Hicks C, Muschamp JW, Rawls SM, Olive MF. DARK Classics in Chemical Neuroscience: Cathinone-Derived Psychostimulants. ACS Chem Neurosci 2018; 9:2379-2394. [PMID: 29714473 DOI: 10.1021/acschemneuro.8b00147] [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] [Indexed: 02/08/2023] Open
Abstract
Cathinone is a plant alkaloid found in khat leaves of perennial shrubs grown in East Africa. Similar to cocaine, cathinone elicits psychostimulant effects which are in part attributed to its amphetamine-like structure. Around 2010, home laboratories began altering the parent structure of cathinone to synthesize derivatives with mechanisms of action, potencies, and pharmacokinetics permitting high abuse potential and toxicity. These "synthetic cathinones" include 4-methylmethcathinone (mephedrone), 3,4-methylenedioxypyrovalerone (MDPV), and the empathogenic agent 3,4-methylenedioxymethcathinone (methylone) which collectively gained international popularity following aggressive online marketing as well as availability in various retail outlets. Case reports made clear the health risks associated with these agents and, in 2012, the Drug Enforcement Agency of the United States placed a series of synthetic cathinones on Schedule I under emergency order. Mechanistically, cathinone and synthetic derivatives work by augmenting monoamine transmission through release facilitation and/or presynaptic transport inhibition. Animal studies confirm the rewarding and reinforcing properties of synthetic cathinones by utilizing self-administration, place conditioning, and intracranial self-stimulation assays and additionally show persistent neuropathological features which demonstrate a clear need to better understand this class of drugs. This Review will thus detail (i) historical context of cathinone use and the rise of "dark" synthetic derivatives, (ii) structural features and mechanisms of synthetic cathinones, (iii) behavioral effects observed clinically and in animals under controlled laboratory conditions, and (iv) neurotransmitters and circuits that may be targeted to manage synthetic cathinone abuse in humans.
Collapse
Affiliation(s)
- Steven J. Simmons
- Center for Substance Abuse Research (CSAR), Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania 19140, United States
| | | | - Chicora F. Oliver
- Center for Substance Abuse Research (CSAR), Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania 19140, United States
| | - Callum Hicks
- Center for Substance Abuse Research (CSAR), Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania 19140, United States
| | - John W. Muschamp
- Center for Substance Abuse Research (CSAR), Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania 19140, United States
| | - Scott M. Rawls
- Center for Substance Abuse Research (CSAR), Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania 19140, United States
| | - M. Foster Olive
- Department of Psychology, Arizona State University, Tempe, Arizona 85281, United States
| |
Collapse
|
35
|
Loweth JA, Reimers JM, Caccamise A, Stefanik MT, Woo KKY, Chauhan NM, Werner CT, Wolf ME. mGlu1 tonically regulates levels of calcium-permeable AMPA receptors in cultured nucleus accumbens neurons through retinoic acid signaling and protein translation. Eur J Neurosci 2018; 50:2590-2601. [PMID: 30222904 DOI: 10.1111/ejn.14151] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 08/22/2018] [Accepted: 08/28/2018] [Indexed: 12/24/2022]
Abstract
In several brain regions, ongoing metabotropic glutamate receptor 1 (mGlu1) transmission has been shown to tonically suppress synaptic levels of Ca2+ -permeable AMPA receptors (CP-AMPARs) while pharmacological activation of mGlu1 removes CP-AMPARs from these synapses. Consistent with this, we previously showed in nucleus accumbens (NAc) medium spiny neurons (MSNs) that reduced mGlu1 tone enables and mGlu1 positive allosteric modulation reverses the elevation of CP-AMPAR levels in the NAc that underlies enhanced cocaine craving in the "incubation of craving" rat model of addiction. To better understand mGlu1/CP-AMPAR interactions, we used a NAc/prefrontal cortex co-culture system in which NAc MSNs express high CP-AMPAR levels, providing an in vitro model for NAc MSNs after the incubation of cocaine craving. The non-specific group I orthosteric agonist dihydroxyphenylglycine (10 min) decreased cell surface GluA1 but not GluA2, indicating CP-AMPAR internalization. This was prevented by mGlu1 (LY367385) or mGlu5 (MTEP) blockade. However, a selective role for mGlu1 emerged in studies of long-term antagonist treatment. Thus, LY367385 (24 hr) increased surface GluA1 without affecting GluA2, whereas MTEP (24 hr) had no effect. In hippocampal neurons, scaling up of CP-AMPARs can occur through a mechanism requiring retinoic acid (RA) signaling and new GluA1 synthesis. Consistent with this, the LY367385-induced increase in surface GluA1 was blocked by anisomycin (translation inhibitor) or 4-(diethylamino)-benzaldehyde (RA synthesis inhibitor). Thus, mGlu1 transmission tonically suppresses cell surface CP-AMPAR levels, and decreasing mGlu1 tone increases surface CP-AMPARs via RA signaling and protein translation. These results identify a novel mechanism for homeostatic plasticity in NAc MSNs.
Collapse
Affiliation(s)
- Jessica A Loweth
- Department of Neuroscience, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois
| | - Jeremy M Reimers
- Department of Neuroscience, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois
| | - Aaron Caccamise
- Department of Neuroscience, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois
| | - Michael T Stefanik
- Department of Neuroscience, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois
| | - Kenneth Kin Yan Woo
- Department of Neuroscience, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois
| | - Nirav M Chauhan
- Department of Neuroscience, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois
| | - Craig T Werner
- Department of Neuroscience, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois
| | - Marina E Wolf
- Department of Neuroscience, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois
| |
Collapse
|
36
|
Phosphoproteomic Analysis of the Amygdala Response to Adolescent Glucocorticoid Exposure Reveals G-Protein Coupled Receptor Kinase 2 as a Target for Reducing Motivation for Alcohol. Proteomes 2018; 6:proteomes6040041. [PMID: 30322021 PMCID: PMC6313880 DOI: 10.3390/proteomes6040041] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 09/28/2018] [Accepted: 10/09/2018] [Indexed: 01/19/2023] Open
Abstract
Early life stress is associated with risk for developing alcohol use disorders (AUDs) in adulthood. Though the neurobiological mechanisms underlying this vulnerability are not well understood, evidence suggests that aberrant glucocorticoid and noradrenergic system functioning play a role. The present study investigated the long-term consequences of chronic exposure to elevated glucocorticoids during adolescence on the risk of increased alcohol-motivated behavior, and on amygdalar function in adulthood. A discovery-based analysis of the amygdalar phosphoproteome using mass spectrometry was employed, to identify changes in function. Adolescent corticosterone (CORT) exposure increased alcohol, but not sucrose, self-administration, and enhanced stress-induced reinstatement with yohimbine in adulthood. Phosphoproteomic analysis indicated that the amygdala phosphoproteome was significantly altered by adolescent CORT exposure, generating a list of potential novel mechanisms involved in the risk of alcohol drinking. In particular, increased phosphorylation at serines 296–299 on the α2A adrenergic receptor (α2AAR), mediated by the G-protein coupled receptor kinase 2 (GRK2), was evident after adolescent CORT exposure. We found that intra-amygdala infusion of a peptidergic GRK2 inhibitor reduced alcohol seeking, as measured by progressive ratio and stress reinstatement tests, and induced by the α2AAR antagonist yohimbine. These results suggest that GRK2 represents a novel target for treating stress-induced motivation for alcohol which may counteract alterations in brain function induced by adolescent stress exposure.
Collapse
|
37
|
Tonn Eisinger KR, Gross KS, Head BP, Mermelstein PG. Interactions between estrogen receptors and metabotropic glutamate receptors and their impact on drug addiction in females. Horm Behav 2018; 104:130-137. [PMID: 29505763 PMCID: PMC6131090 DOI: 10.1016/j.yhbeh.2018.03.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 02/28/2018] [Accepted: 03/01/2018] [Indexed: 02/07/2023]
Abstract
Contribution to Special Issue on Fast effects of steroids. Estrogen receptors α and β (ERα and ERβ) have a unique relationship with metabotropic glutamate receptors (mGluRs) in the female rodent brain such that estradiol is able to recruit intracellular G-protein signaling cascades to influence neuronal physiology, structure, and ultimately behavior. While this association between ERs and mGluRs exists in many cell types and brain regions, its effects are perhaps most striking in the nucleus accumbens (NAc). This review will discuss the original characterization of ER/mGluR signaling and how estradiol activity in the NAc confers increased sensitivity to drugs of abuse in females through this mechanism.
Collapse
Affiliation(s)
- Katherine R Tonn Eisinger
- Department of Neuroscience and Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Kellie S Gross
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, USA
| | - Brian P Head
- Department of Anesthesiology, University of California-San Diego, La Jolla, CA 92093, USA
| | - Paul G Mermelstein
- Department of Neuroscience and Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
38
|
Caprioli D, Justinova Z, Venniro M, Shaham Y. Effect of Novel Allosteric Modulators of Metabotropic Glutamate Receptors on Drug Self-administration and Relapse: A Review of Preclinical Studies and Their Clinical Implications. Biol Psychiatry 2018; 84:180-192. [PMID: 29102027 PMCID: PMC5837933 DOI: 10.1016/j.biopsych.2017.08.018] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 08/23/2017] [Accepted: 08/28/2017] [Indexed: 12/31/2022]
Abstract
Results from preclinical rodent studies during the last 20 years implicated glutamate neurotransmission in different brain regions in drug self-administration and rodent models of relapse. These results, along with evidence for drug-induced neuroadaptations in glutamatergic neurons and receptors, suggested that addiction might be treatable by medications that inhibit glutamatergic responses to drugs of abuse, drug-associated cues, and stressors. This idea is supported by findings in rodent and primate models that drug self-administration and relapse are reduced by systemic injections of antagonists of ionotropic glutamate receptors or metabotropic glutamate receptors (mGluRs) or orthosteric agonists of mGluR2/3. However, these compounds have not advanced to clinical use because of potential side effects and other factors. This state of affairs has led to the development of positive allosteric modulators (PAMs) and negative allosteric modulators (NAMs) of mGluRs. PAMs and NAMs of mGluRs, either of which can inhibit evoked glutamate release, may be suitable for testing in humans. We reviewed results from recent studies of systemically injected PAMs and NAMs of mGluRs in rodents and monkeys, focusing on whether they reduce drug self-administration, reinstatement of drug seeking, and incubation of drug craving. We also review results from rat studies in which PAMs or NAMs of mGluRs were injected intracranially to reduce drug self-administration and reinstatement. We conclude that PAMs and NAMs of mGluRs should be considered for clinical trials.
Collapse
Affiliation(s)
- Daniele Caprioli
- Department of Physiology and Pharmacology "Vittorio Erspamer", Sapienza University of Rome, Rome, Italy.
| | - Zuzana Justinova
- Behavioral Neuroscience Research Branch, Intramural Research Program, NIDA, NIH, DHHS, Baltimore, MD, USA
| | - Marco Venniro
- Behavioral Neuroscience Research Branch, Intramural Research Program, NIDA, NIH, DHHS, Baltimore, MD, USA
| | - Yavin Shaham
- Behavioral Neuroscience Research Branch, Intramural Research Program, NIDA, NIH, DHHS, Baltimore, MD, USA
| |
Collapse
|
39
|
Aboutalebi F, Alaei H, Oryan S, Radahmadi M. Blockade of prelimbic glutamate receptor reduces the reinforcing effect of morphine. Can J Physiol Pharmacol 2018; 96:815-822. [PMID: 29947552 DOI: 10.1139/cjpp-2017-0758] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The prelimbic cortex (PrL) as a part of the medial prefrontal cortex (mPFC) plays a crucial role in drug addiction. Previous studies have shown that glutamatergic transmission through the NMDA and AMPA receptors plays an important role in morphine rewarding properties. In this study, we evaluated the effect of glutamate receptors blockade within the PrL on morphine self-administration. Male Wistar rats were randomly selected and divided into 7 groups. Trained rats were placed in self-administration apparatus, where they pressed an active lever for receiving morphine (5 mg/mL) in test groups and saline in saline group during 11 consecutive days for 2 h per session. The effects of intra-prelimbic AMPA receptor antagonist (CNQX; 0.5 and 2.5 μg/0.5 μL) and the NMDA antagonist (AP5; 0.1 and 1 μg/0.5 μL) on self-administration were tested. Our results demonstrated that intra-prelimbic injection of different doses of CNQX and AP5, and co-administration of these 2 drugs before self-administration significantly decreased active lever pressing compared with morphine group (p < 0.001). Also, the number of self-infusion significantly decreased in test groups compared with morphine group (p < 0.001). These findings suggest that a reduction in PrL glutamatergic output can modulate morphine reinforcement.
Collapse
Affiliation(s)
| | - Hojjatallah Alaei
- b Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Shahrbanoo Oryan
- a Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Maryam Radahmadi
- b Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
40
|
Cross AJ, Anthenelli R, Li X. Metabotropic Glutamate Receptors 2 and 3 as Targets for Treating Nicotine Addiction. Biol Psychiatry 2018; 83:947-954. [PMID: 29301614 PMCID: PMC5953779 DOI: 10.1016/j.biopsych.2017.11.021] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2017] [Revised: 11/02/2017] [Accepted: 11/14/2017] [Indexed: 12/27/2022]
Abstract
Tobacco smoking, driven by the addictive properties of nicotine, continues to be a worldwide health problem. Based on the well-established role of glutamatergic neurotransmission in drug addiction, novel medication development strategies seek to halt nicotine consumption and prevent relapse to tobacco smoking by modulating glutamate transmission. The presynaptic inhibitory metabotropic glutamate receptors 2 and 3 (mGluR2/3) are key autoreceptors on glutamatergic terminals that maintain glutamate homeostasis. Accumulating evidence suggests the critical role of mGluR2/3 in different aspects of nicotine addiction, including acquisition and maintenance of nicotine taking, nicotine withdrawal, and persistent nicotine seeking even after prolonged abstinence. The involvement of mGluR2/3 in other neuropsychiatric conditions, such as anxiety, depression, schizophrenia, Alzheimer's disease, Parkinson's disease, and pain, provides convincing evidence suggesting that mGluR2/3 may provide an effective therapeutic approach for comorbidity of smoking and these conditions. This focused review article highlights that mGluR2/3 provide a promising target in the search for smoking cessation medication with novel mechanisms of actions that differ from those of currently U.S. Food and Drug Administration-approved pharmacotherapies.
Collapse
Affiliation(s)
- Alan J Cross
- AstraZeneca Neuroscience Innovative Medicines, Cambridge, Massachusetts
| | - Robert Anthenelli
- Department of Psychiatry, School of Medicine, University of California San Diego, La Jolla, California
| | - Xia Li
- Department of Psychiatry, School of Medicine, University of California San Diego, La Jolla, California.
| |
Collapse
|
41
|
Beggiato S, Borelli AC, Tomasini MC, Castelli MP, Pintori N, Cacciaglia R, Loche A, Ferraro L. In Vitro Functional Characterization of GET73 as Possible Negative Allosteric Modulator of Metabotropic Glutamate Receptor 5. Front Pharmacol 2018; 9:327. [PMID: 29674969 PMCID: PMC5895880 DOI: 10.3389/fphar.2018.00327] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 03/21/2018] [Indexed: 01/08/2023] Open
Abstract
The present study was aimed to further characterize the pharmacological profile of N-[4-(trifluoromethyl) benzyl]-4-methoxybutyramide (GET73), a putative negative allosteric modulator (NAM) of metabotropic glutamate subtype 5 receptor (mGluR5) under development as a novel medication for the treatment of alcohol dependence. This aim has been accomplished by means of a series of in vitro functional assays. These assays include the measure of several down-stream signaling [intracellular Ca++ levels, inositol phosphate (IP) formation and CREB phosphorylation (pCREB)] which are generally affected by mGluR5 ligands. In particular, GET73 (0.1 nM-10 μM) was explored for its ability to displace the concentration-response curve of some mGluR5 agonists/probes (glutamate, L-quisqualate, CHPG) in different native preparations. GET73 produced a rightward shift of concentration-response curves of glutamate- and CHPG-induced intracellular Ca++ levels in primary cultures of rat cortical astrocytes. The compound also induced a rightward shift of concentration response curve of glutamate- and L-quisqualate-induced increase in IP turnover in rat hippocampus slices, along with a reduction of CHPG (10 mM)-induced increase in IP formation. Moreover, GET73 produced a rightward shift of concentration-response curve of glutamate-, CHPG- and L-quisqualate-induced pCREB levels in rat cerebral cortex neurons. Although the engagement of other targets cannot be definitively ruled out, these data support the view that GET73 acts as an mGluR5 NAM and support the significance of further investigating the possible mechanism of action of the compound.
Collapse
Affiliation(s)
- Sarah Beggiato
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy.,IRET Foundation, Bologna, Italy
| | - Andrea C Borelli
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy.,IRET Foundation, Bologna, Italy
| | - Maria C Tomasini
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy.,IRET Foundation, Bologna, Italy
| | - M Paola Castelli
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy.,Center of Excellence "Neurobiology of Addiction", University of Cagliari, Cagliari, Italy
| | - Nicholas Pintori
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | | | | | - Luca Ferraro
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy.,IRET Foundation, Bologna, Italy.,LTTA Centre, University of Ferrara, Ferrara, Italy
| |
Collapse
|
42
|
Tang ZQ, Lu Y. Anatomy and Physiology of Metabotropic Glutamate Receptors in Mammalian and Avian Auditory System. ACTA ACUST UNITED AC 2018; 1. [PMID: 30854519 DOI: 10.24966/tap-7752/100001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Glutamate, as the major excitatory neurotransmitter used in the vertebrate brain, activates ionotropic and metabotropic glutamate receptors (iGluRs and mGluRs), which mediate fast and slow neuronal actions, respectively. mGluRs play important modulatory roles in many brain areas, forming potential targets for drugs developed to treat brain disorders. Here, we review studies on mGluRs in the mammalian and avian auditory system. Although anatomical expression of mGluRs in the cochlear nucleus has been well characterized, data for other auditory nuclei await more systematic investigations especially at the electron microscopy level. The physiology of mGluRs has been extensively studied using in vitro brain slice preparations, with a focus on the auditory circuitry in the brainstem. These in vitro physiological studies have demonstrated that mGluRs participate in synaptic transmission, regulate ionic homeostasis, induce synaptic plasticity, and maintain the balance between Excitation and Inhibition (E/I) in a variety of auditory structures. However, the modulatory roles of mGluRs in auditory processing remain largely unclear at the system and behavioral levels, and the functions of mGluRs in auditory disorders remain entirely unknown.
Collapse
Affiliation(s)
- Zheng-Quan Tang
- Oregon Hearing Research Center, Vollum Institute, Oregon Health and Science University, Oregon, USA
| | - Yong Lu
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Ohio, USA
| |
Collapse
|
43
|
Haass-Koffler CL, Goodyear K, Loche A, Long VM, Lobina C, Tran HH, Cacciaglia R, Swift RM, Colombo G, Leggio L. Administration of the metabotropic glutamate receptor subtype 5 allosteric modulator GET 73 with alcohol: A translational study in rats and humans. J Psychopharmacol 2018; 32:163-173. [PMID: 29361897 PMCID: PMC7014573 DOI: 10.1177/0269881117746904] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Preclinical work suggests that GET 73 (N-[4-(trifluoromethyl)benzyl]-4-methoxybutyramide), a novel metabotropic glutamate receptor subtype 5 negative allosteric modulator, may represent a novel pharmacological treatment for alcohol use disorder. Two independent experiments evaluated the effect of acutely administered GET 73 (0, 30, and 100 mg/kg, intragastrically) on alcohol-induced hypolocomotion ( n=72) and sedation/hypnosis ( n=36) in rats. In healthy male volunteers ( n=14), an open-label, randomised, crossover study was conducted to compare adverse events and pharmacokinetic parameters, in two experiments in which 300 mg GET 73 was administered, with and without alcohol, once and thrice. In rats, when administered with alcohol-vehicle, 100 mg/kg, but not 30 mg/kg, GET 73 reduced spontaneous locomotor activity. When administered with alcohol, no dose of GET 73 altered either alcohol-induced hypolocomotion or sedation/hypnosis. In humans, both single and thrice 300 mg GET 73 administration were well tolerated, in the presence and absence of alcohol, with no differences in adverse events. There were no significant differences in relative bioavailability between administering 300 mg GET 73 in the presence or absence of alcohol.
Collapse
Affiliation(s)
- Carolina L Haass-Koffler
- Center for Alcohol and Addiction Studies, Department of Psychiatry and Human Behavior, Brown University, Providence, RI, USA,Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, Brown University, Providence, RI, USA,Corresponding author: Carolina L Haass-Koffler, Center for Alcohol and Addiction Studies, Department Psychiatry and Human Behavior, Brown University, 121 South Main Street, Providence, RI 02912, USA;
| | - Kimberly Goodyear
- Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, Brown University, Providence, RI, USA
| | | | - Victoria M Long
- Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, Brown University, Providence, RI, USA
| | - Carla Lobina
- Neuroscience Institute, Section of Cagliari, National Research Council of Italy, Cagliari, Italy
| | - Harrison H Tran
- Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, Brown University, Providence, RI, USA
| | | | - Robert M Swift
- Center for Alcohol and Addiction Studies, Department of Psychiatry and Human Behavior, Brown University, Providence, RI, USA
| | - Giancarlo Colombo
- Neuroscience Institute, Section of Cagliari, National Research Council of Italy, Cagliari, Italy
| | - Lorenzo Leggio
- Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, Brown University, Providence, RI, USA
| |
Collapse
|
44
|
Hearing M, Graziane N, Dong Y, Thomas MJ. Opioid and Psychostimulant Plasticity: Targeting Overlap in Nucleus Accumbens Glutamate Signaling. Trends Pharmacol Sci 2018; 39:276-294. [PMID: 29338873 DOI: 10.1016/j.tips.2017.12.004] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Revised: 11/11/2017] [Accepted: 12/13/2017] [Indexed: 12/11/2022]
Abstract
Commonalities in addictive behavior, such as craving, stimuli-driven drug seeking, and a high propensity for relapse following abstinence, have pushed for a unified theory of addiction that encompasses most abused substances. This unitary theory has recently been challenged - citing distinctions in structural neural plasticity, biochemical signaling, and neural circuitry to argue that addiction to opioids and psychostimulants is behaviorally and neurobiologically distinct. Recent more selective examination of drug-induced plasticity has highlighted that these two drug classes promote an overall reward circuitry signaling overlap through modifying excitatory synapses in the nucleus accumbens - a key constituent of the reward system. We discuss adaptations in presynaptic/postsynaptic and extrasynaptic glutamate signaling produced by opioids and psychostimulants, and their relevance to circuit remodeling and addiction-related behavior - arguing that these core neural adaptations are important targets for developing pharmacotherapies to treat addiction to multiple drugs.
Collapse
Affiliation(s)
- Matthew Hearing
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI 53233, USA.
| | - Nicholas Graziane
- Department of Anesthesiology and Perioperative Medicine, Penn State College of Medicine, Hershey, PA 17033, USA; Departments of Neuroscience and Psychiatry, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Yan Dong
- Departments of Neuroscience and Psychiatry, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Mark J Thomas
- Department of Psychology, University of Minnesota, Minneapolis, MN 55455, USA; Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
45
|
Akkus F, Mihov Y, Treyer V, Ametamey SM, Johayem A, Senn S, Rösner S, Buck A, Hasler G. Metabotropic glutamate receptor 5 binding in male patients with alcohol use disorder. Transl Psychiatry 2018; 8:17. [PMID: 29317611 PMCID: PMC5802584 DOI: 10.1038/s41398-017-0066-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 09/13/2017] [Accepted: 10/26/2017] [Indexed: 01/30/2023] Open
Abstract
Glutamate signaling plays a major role in addiction. Preclinical research strongly suggests an implication of G-protein-coupled metabotropic glutamate receptor subtype 5 (mGluR5) in nicotine addiction and alcohol use disorder. In humans, smoking is related to a global reduction in mGluR5 availability. In the present study, we investigated mGluR5 in vivo in patients with alcohol use disorder without the confounding effects of smoking. A total of 14 male subjects with alcohol use disorder and at least a 25-day abstinence and 14 matched male non-smoking healthy controls were included in the study. We employed positron emission tomography (PET) with the mGluR5-specific radiotracer [11C]ABP688, using a bolus/infusion protocol. We found increased mGluR5 DVR in several regions within the temporal lobe in patients, as compared to controls. The largest between-group difference was in the amygdala. There was a marked positive relation between mGluR5 DVR in the anterior cingulate and mGluR5 DVR in the orbitofrontal cortex in patients, but not in controls. In patients, lower temptation to drink was related to higher amygdala mGluR5 DVR. We did not find altered mGluR5 DVR in the basal ganglia of subjects recovering from alcohol use disorder. In conclusion, our study provides clinical evidence for altered mGluR5 signaling in the amygdala in alcohol use disorder. This alteration was associated with the temptation to drink. In addition, this study suggests abnormal mGluR5 signaling in a network underlying reward-related behavioral flexibility. These findings strengthen the case for pharmacological agents acting on mGluR5 as promising candidates for the treatment of alcohol use disorder.
Collapse
Affiliation(s)
- Funda Akkus
- Division of Molecular Psychiatry, Translational Research Center, University Hospital of Psychiatry, University of Bern, 3000, Bern 60, Switzerland
| | - Yoan Mihov
- Division of Molecular Psychiatry, Translational Research Center, University Hospital of Psychiatry, University of Bern, 3000, Bern 60, Switzerland
| | - Valerie Treyer
- PET Center, Division of Nuclear Medicine, University Hospital, 8091, Zurich, Switzerland
| | - Simon M Ametamey
- Center for Radiopharmaceutical Science of ETH, PSI, and USZ, Department of Chemistry and Applied Biosciences of ETH, 8093, Zurich, Switzerland
| | - Anass Johayem
- PET Center, Division of Nuclear Medicine, University Hospital, 8091, Zurich, Switzerland
| | - Smeralda Senn
- Forel Clinic, Addiction Treatment Center, 8548, Ellikon an der Thur, Switzerland
| | - Susanne Rösner
- Forel Clinic, Addiction Treatment Center, 8548, Ellikon an der Thur, Switzerland
| | - Alfred Buck
- PET Center, Division of Nuclear Medicine, University Hospital, 8091, Zurich, Switzerland
| | - Gregor Hasler
- Division of Molecular Psychiatry, Translational Research Center, University Hospital of Psychiatry, University of Bern, 3000, Bern 60, Switzerland.
| |
Collapse
|
46
|
Cuzon Carlson VC. GABA and Glutamate Synaptic Coadaptations to Chronic Ethanol in the Striatum. Handb Exp Pharmacol 2018; 248:79-112. [PMID: 29460153 DOI: 10.1007/164_2018_98] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Alcohol (ethanol) is a widely used and abused drug with approximately 90% of adults over the age of 18 consuming alcohol at some point in their lifetime. Alcohol exerts its actions through multiple neurotransmitter systems within the brain, most notably the GABAergic and glutamatergic systems. Alcohol's actions on GABAergic and glutamatergic neurotransmission have been suggested to underlie the acute behavioral effects of ethanol. The striatum is the primary input nucleus of the basal ganglia that plays a role in motor and reward systems. The effect of ethanol on GABAergic and glutamatergic neurotransmission within striatal circuitry has been thought to underlie ethanol taking, seeking, withdrawal and relapse. This chapter reviews the effects of ethanol on GABAergic and glutamatergic transmission, highlighting the dynamic changes in striatal circuitry from acute to chronic exposure and withdrawal.
Collapse
|
47
|
Accumbens Mechanisms for Cued Sucrose Seeking. Neuropsychopharmacology 2017; 42:2377-2386. [PMID: 28726801 PMCID: PMC5645741 DOI: 10.1038/npp.2017.153] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 07/10/2017] [Accepted: 07/11/2017] [Indexed: 12/29/2022]
Abstract
Many studies support a perspective that addictive drugs usurp brain circuits used by natural rewards, especially for the dopamine-dependent reinforcing qualities of both drugs and natural rewards. Reinstated drug seeking in animal models of relapse relies on glutamate spillover from cortical terminals synapsing in the nucleus accumbens core (NAcore) to stimulate metabotropic glutamate receptor5 (mGluR5) on neuronal nitric oxide synthase (nNOS) interneurons. Contrasting the release of dopamine that is shared by sucrose and drugs of abuse, reinstated sucrose seeking does not induce glutamate spillover. We hypothesized that pharmacologically promoting glutamate spillover in the NAcore would mimic cocaine-induced adaptations and potentiate cued reinstatement of sucrose seeking. Inducing glutamate spillover by blocking astroglial glutamate transporters (GLT-1) had no effect on reinstated sucrose seeking. However, glutamate release probability is negatively regulated by presynaptic mGluR2/3, and sucrose reinstatement was potentiated following mGluR2/3 blockade. Potentiated sucrose reinstatement by mGluR2/3 blockade was reversed by antagonizing mGluR5, but reinstated sucrose seeking in the absence of mGluR2/3 blockade was not affected by blocking mGluR5. In cocaine-trained rodents mGluR5 stimulation reinstates drug seeking by activating nNOS, but activating mGluR5 did not promote reinstated sucrose seeking, nor was potentiated reinstatement after mGluR2/3 blockade reduced by blocking nNOS. However, chemogenetic activation of nNOS interneurons in the NAcore reinstated sucrose seeking. These data indicate that dysregulated presynaptic mGluR2/3 signaling is a possible site of shared signaling in drug seeking and potentiated reinstated sucrose seeking, but that downregulated glutamate transport and subsequent activation of nNOS by synaptic glutamate spillover is not shared.
Collapse
|
48
|
Ding ZM, Ingraham CM, Hauser SR, Lasek AW, Bell RL, McBride WJ. Reduced Levels of mGlu2 Receptors within the Prelimbic Cortex Are Not Associated with Elevated Glutamate Transmission or High Alcohol Drinking. Alcohol Clin Exp Res 2017; 41:1896-1906. [PMID: 28858384 PMCID: PMC5659915 DOI: 10.1111/acer.13488] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 08/23/2017] [Indexed: 12/23/2022]
Abstract
BACKGROUND A Grm2 cys407* stop codon mutation, which results in a loss of the metabotropic glutamate 2 (mGlu2) receptor protein, was identified as being associated with high alcohol drinking by alcohol-preferring (P) rats. The objectives of the current study were to characterize the effects of reduced levels of mGlu2 receptors on glutamate transmission and alcohol drinking. METHODS Quantitative no-net-flux microdialysis was used to test the hypothesis that basal extracellular glutamate levels in the prelimbic (PL) cortex and nucleus accumbens shell (NACsh) will be higher in P than Wistar rats. A lentiviral-delivered short-hairpin RNA (shRNA)-mediated knockdown was used to test the hypothesis that reduced levels of mGlu2 receptors within the PL cortex will increase voluntary alcohol drinking by Wistar rats. A linear regression analysis was used to test the hypothesis that there will be a significant correlation between the Grm2 cys407* mutation and level of alcohol intake. RESULTS Extracellular glutamate concentrations within the PL cortex (3.6 ± 0.6 vs. 6.4 ± 0.6 μM) and NACsh (3.2 ± 0.4 vs. 6.6 ± 0.6 μM) were significantly lower in female P than female Wistar rats. Western blot detected the presence of mGlu2 receptors in these regions of female Wistar rats, but not female P rats. Micro-infusion of shRNAs into the PL cortex significantly reduced local mGlu2 receptor levels (by 40%), but did not alter voluntary alcohol drinking in male Wistar rats. In addition, there was no significant correlation between the Grm2 mutation and alcohol intake in 36 rodent lines (r = 0.29, p > 0.05). CONCLUSIONS Collectively, these results suggest a lack of association between the loss of mGlu2 receptors and glutamate transmission in the NACsh and PL cortex of female P rats, and between the level of mGlu2 receptors in the PL cortex and alcohol drinking of male Wistar rats.
Collapse
Affiliation(s)
- Zheng-Ming Ding
- Institute of Psychiatric Research, Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Cynthia M. Ingraham
- Institute of Psychiatric Research, Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Sheketha R. Hauser
- Institute of Psychiatric Research, Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Amy W. Lasek
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL 60612
| | - Richard L. Bell
- Institute of Psychiatric Research, Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN 46202
| | - William J. McBride
- Institute of Psychiatric Research, Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN 46202
| |
Collapse
|
49
|
Knezevic NN, Yekkirala A, Yaksh TL. Basic/Translational Development of Forthcoming Opioid- and Nonopioid-Targeted Pain Therapeutics. Anesth Analg 2017; 125:1714-1732. [PMID: 29049116 PMCID: PMC5679134 DOI: 10.1213/ane.0000000000002442] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Opioids represent an efficacious therapeutic modality for some, but not all pain states. Singular reliance on opioid therapy for pain management has limitations, and abuse potential has deleterious consequences for patient and society. Our understanding of pain biology has yielded insights and opportunities for alternatives to conventional opioid agonists. The aim is to have efficacious therapies, with acceptable side effect profiles and minimal abuse potential, which is to say an absence of reinforcing activity in the absence of a pain state. The present work provides a nonexclusive overview of current drug targets and potential future directions of research and development. We discuss channel activators and blockers, including sodium channel blockers, potassium channel activators, and calcium channel blockers; glutamate receptor-targeted agents, including N-methyl-D-aspartate, α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid, and metabotropic receptors. Furthermore, we discuss therapeutics targeted at γ-aminobutyric acid, α2-adrenergic, and opioid receptors. We also considered antagonists of angiotensin 2 and Toll receptors and agonists/antagonists of adenosine, purine receptors, and cannabinoids. Novel targets considered are those focusing on lipid mediators and anti-inflammatory cytokines. Of interest is development of novel targeting strategies, which produce long-term alterations in pain signaling, including viral transfection and toxins. We consider issues in the development of druggable molecules, including preclinical screening. While there are examples of successful translation, mechanistically promising preclinical candidates may unexpectedly fail during clinical trials because the preclinical models may not recapitulate the particular human pain condition being addressed. Molecular target characterization can diminish the disconnect between preclinical and humans' targets, which should assist in developing nonaddictive analgesics.
Collapse
Affiliation(s)
- Nebojsa Nick Knezevic
- From the *Department of Anesthesiology, Advocate Illinois Masonic Medical Center Chicago, Illinois; Departments of †Anesthesiology and ‡Surgery, University of Illinois, Chicago, Illinois; §Department of Neurobiology, Harvard Medical School, and Boston Children's Hospital, Boston, Massachusetts; ‖Blue Therapeutics, Harvard Innovation Launch Lab, Allston, Massachusetts; and Departments of ¶Anesthesiology and #Pharmacology, University of California, San Diego, La Jolla, California
| | | | | |
Collapse
|
50
|
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
|