1
|
Legaki E, Dovrolis N, Moscholiou N, Koutromanos I, Vassilopoulos E, Dakanalis A, Gazouli M, Tzavellas E. Altered Expression of Neuroplasticity-Related Genes in Alcohol Addiction and Treatment. Int J Mol Sci 2024; 25:11349. [PMID: 39518903 PMCID: PMC11546795 DOI: 10.3390/ijms252111349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 10/16/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024] Open
Abstract
Alcohol use disorder's complexity arises from genetic and environmental factors, with alcohol metabolism genes and neurotransmitter pathways being critical. This study aims to analyze synaptic plasticity gene expression changes in individuals with AUD in order to study their contribution to AUD development and to identify potential biomarkers of treatment response. RNA was extracted from whole peripheral blood (20 patients, 10 healthy controls), before and after treatment (Qiagen AllPrep RNA/DNA Mini Kit), and the gene expression of 84 genes related to neuroplasticity was studied using the RT2 Profiler for Human Synaptic Plasticity RT-PCR Array (PAHS-126ZA, Qiagen), comparing AUD patients to control and responders to non-responders. The potential prognostic/predictive biomarkers were searched using machine learning models. A total of 35 dysregulated genes were found in AUD patients. EPHB2, EGR, and AKT1 were increased, while TIMP1, NCAM1, and GRM2 were decreased. Responders showed distinct gene expression profiles at baseline. After treatment, the expression of 57 genes was normalized, while NCAM1, GRM2, and BDNF showed the most significant recovery. EGR4, INHBA, and NCAM1 emerged as potential biomarkers to predict treatment success. These results indicate that gene profiles in peripheral blood can serve as prognostic markers for the prognosis and treatment of AUD, although further validation is required.
Collapse
Affiliation(s)
- Evangelia Legaki
- Laboratory of Biology, Department of Basic Biological Science, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (E.L.); (N.D.); (N.M.)
| | - Nikolas Dovrolis
- Laboratory of Biology, Department of Basic Biological Science, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (E.L.); (N.D.); (N.M.)
| | - Nikoletta Moscholiou
- Laboratory of Biology, Department of Basic Biological Science, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (E.L.); (N.D.); (N.M.)
| | - Ilias Koutromanos
- First Department of Psychiatry, “Aiginition” Hospital, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (I.K.); (E.V.)
- Department of Psychiatry and Psychotherapy, Psychiatric Services Aargou AG, 5210 Brugg-Windisch, Switzerland
| | - Efthimios Vassilopoulos
- First Department of Psychiatry, “Aiginition” Hospital, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (I.K.); (E.V.)
| | - Antonios Dakanalis
- Department of Medicine and Surgery, University of Milano Bicocca, Via Cadore 38, 20900 Monza, Italy;
| | - Maria Gazouli
- Laboratory of Biology, Department of Basic Biological Science, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (E.L.); (N.D.); (N.M.)
| | - Elias Tzavellas
- First Department of Psychiatry, “Aiginition” Hospital, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (I.K.); (E.V.)
| |
Collapse
|
2
|
Weiss ER, Davis MT, Asch RH, D'Souza DC, Cool R, Esterlis I. Metabotropic Glutamate Receptor 5 as a Potential Biomarker of the Intersection of Trauma and Cannabis Use. Int J Neuropsychopharmacol 2024; 27:pyae044. [PMID: 39320043 DOI: 10.1093/ijnp/pyae044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 09/23/2024] [Indexed: 09/26/2024] Open
Abstract
BACKGROUND Metabotropic glutamate receptor 5 (mGlu5) dysregulation has been implicated in the pathophysiology of trauma-related psychopathology, and there are direct interactions between the endocannabinoid and glutamatergic systems. However, relationships between cannabis use (CU) and mGlu5 have not been directly investigated in trauma-related psychopathology. METHODS Using positron emission tomography with [18F]FPEB, we examined relationships between CU status and mGlu5 availability in vivo in a cross-diagnostic sample of individuals with trauma-related psychopathology (n = 55). Specifically, we tested whether mGlu5 availability in frontolimbic regions of interest (ROIs; dorsolateral prefrontal cortex, orbitofrontal cortex, ventromedial prefrontal cortex, amygdala, hippocampus) differed as a function of CU status. RESULTS Past-year CU (n = 22) was associated with 18.62%-19.12% higher mGlu5 availability in frontal and 14.24%-16.55% higher mGlu5 in limbic ROIs relative to participants with no recent CU. Similarly, past-month or monthly CU (n = 16) was associated with higher mGlu5 availability in frontal (18.05%-20.62%) and limbic (15.53%-16.83%) ROIs. mGlu5 availability in the orbitofrontal cortex and amygdala was negatively associated with depressive symptoms in the past-year CU group. In both CU groups, exploratory analyses showed negative correlations between mGlu5 availability and sadness across all ROIs and with perceptions of worthlessness and past failures (r's = -.47 to .66, P's = .006-.033) in the ventromedial prefrontal cortex. Participants with CU reported lower mean depressive symptoms (P's = .006-.037) relative to those without CU. CONCLUSIONS These findings have substantial implications for our understanding of interactions between CU and glutamatergic neurotransmission in trauma-related psychopathology, underscoring the need for treatment development efforts to consider the effects of CU in this population.
Collapse
Affiliation(s)
- Emily R Weiss
- Departments of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA
| | - Margaret T Davis
- Psychiatry, Yale School of Medicine, New Haven, Connecticut, USA
- U.S. Department of Veteran Affairs National Center for Posttraumatic Stress Disorder, Clinical Neurosciences Division, VA Connecticut Healthcare System, West Haven, Connecticut, USA
- Department of Psychology, Yale University, New Haven, Connecticut, USA
| | - Ruth H Asch
- Psychiatry, Yale School of Medicine, New Haven, Connecticut, USA
| | - Deepak Cyril D'Souza
- Psychiatry, Yale School of Medicine, New Haven, Connecticut, USA
- U.S. Department of Veteran Affairs National Center for Posttraumatic Stress Disorder, Clinical Neurosciences Division, VA Connecticut Healthcare System, West Haven, Connecticut, USA
| | - Ryan Cool
- Psychiatry, Yale School of Medicine, New Haven, Connecticut, USA
| | - Irina Esterlis
- Departments of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA
- Psychiatry, Yale School of Medicine, New Haven, Connecticut, USA
- U.S. Department of Veteran Affairs National Center for Posttraumatic Stress Disorder, Clinical Neurosciences Division, VA Connecticut Healthcare System, West Haven, Connecticut, USA
- Department of Psychology, Yale University, New Haven, Connecticut, USA
| |
Collapse
|
3
|
Ramos-Prats A, Matulewicz P, Edenhofer ML, Wang KY, Yeh CW, Fajardo-Serrano A, Kress M, Kummer K, Lien CC, Ferraguti F. Loss of mGlu 5 receptors in somatostatin-expressing neurons alters negative emotional states. Mol Psychiatry 2024; 29:2774-2786. [PMID: 38575807 PMCID: PMC11420089 DOI: 10.1038/s41380-024-02541-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 03/15/2024] [Accepted: 03/19/2024] [Indexed: 04/06/2024]
Abstract
Subtype 5 metabotropic glutamate receptors (mGlu5) are known to play an important role in regulating cognitive, social and valence systems. However, it remains largely unknown at which circuits and neuronal types mGlu5 act to influence these behavioral domains. Altered tissue- or cell-specific expression or function of mGlu5 has been proposed to contribute to the exacerbation of neuropsychiatric disorders. Here, we examined how these receptors regulate the activity of somatostatin-expressing (SST+) neurons, as well as their influence on behavior and brain rhythmic activity. Loss of mGlu5 in SST+ neurons elicited excitatory synaptic dysfunction in a region and sex-specific manner together with a range of emotional imbalances including diminished social novelty preference, reduced anxiety-like behavior and decreased freezing during retrieval of fear memories. In addition, the absence of mGlu5 in SST+ neurons during fear processing impaired theta frequency oscillatory activity in the medial prefrontal cortex and ventral hippocampus. These findings reveal a critical role of mGlu5 in controlling SST+ neurons excitability necessary for regulating negative emotional states.
Collapse
Affiliation(s)
- Arnau Ramos-Prats
- Institute of Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Pawel Matulewicz
- Institute of Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | | | - Kai-Yi Wang
- Institute of Neuroscience, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chia-Wei Yeh
- Institute of Neuroscience, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ana Fajardo-Serrano
- Institute of Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Michaela Kress
- Institute of Physiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Kai Kummer
- Institute of Physiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Cheng-Chang Lien
- Institute of Neuroscience, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Francesco Ferraguti
- Institute of Pharmacology, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
4
|
Jahan M, Amir A, Das A, Kihlström J, Nag S. Automated radiosynthesis of mGluR5 PET tracer [ 18F]FPEB from aryl-chloro precursor and validation for clinical application. J Labelled Comp Radiopharm 2024; 67:155-164. [PMID: 38369901 DOI: 10.1002/jlcr.4088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 12/11/2023] [Accepted: 01/29/2024] [Indexed: 02/20/2024]
Abstract
The radioligand [18F]FPEB, used for PET imaging of the brain's metabotropic glutamate receptor subtype 5 (mGluR5), undergoes a thorough validation process to ensure its safety, efficacy, and quality for clinical use. The process starts by optimizing the synthesis of [18F]FPEB to achieve high radiochemical yield and purity. This study focuses on optimizing the radiolabeling process using an aryl-chloro precursor and validating the GMP production for clinical applications. Fully automated radiolabeling was achieved via one-step nucleophilic substitution reaction. [18F]FPEB was produced and isolated in high radioactivity and radiochemical purity. Throughout the validation process, thorough quality control measures are implemented. Radiopharmaceutical batch release criteria are established, including testing for physical appearance, filter integrity, pH, radiochemical purity, molar activity, radiochemical identity, chemical impurity, structural identity, stability, residual solvent, sterility, and endotoxin levels. In conclusion, the validation of [18F]FPEB involved a comprehensive process of synthesis optimization, quality control, which ensure the safety, efficacy, and quality of [18F]FPEB, enabling its reliable use in clinical PET. Here, we successfully radiolabeled and validated [18F]FPEB using aryl-chloro precursor according to GMP production for clinical application.
Collapse
Affiliation(s)
- Mahabuba Jahan
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | - Arsalan Amir
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | - Arindam Das
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | - Jacob Kihlström
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | - Sangram Nag
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| |
Collapse
|
5
|
Pepe M, Di Nicola M, Cocciolillo F, Chiappini S, Martinotti G, Calcagni ML, Sani G. 3-Methoxy-Phencyclidine Induced Psychotic Disorder: A Literature Review and an 18F-FDG PET/CT Case Report. Pharmaceuticals (Basel) 2024; 17:452. [PMID: 38675413 PMCID: PMC11053433 DOI: 10.3390/ph17040452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 03/23/2024] [Accepted: 03/28/2024] [Indexed: 04/28/2024] Open
Abstract
New Psychoactive Substances (NPS) are modifying the drug scenario worldwide and have become a public health concern because of their toxicological profiles and their harmful physical/psychological effects. 3-Methoxy-Phencyclidine (3-MeO-PCP), a non-competitive antagonist of glutamate N-methyl-D-aspartate (NMDA) receptors, belongs to the phencyclidine-like subfamily of arylcyclohexylamines and has gained attention for its toxic, sometimes fatal, effects. Despite several cases of intoxication and death reported in the literature, little is known about substance-induced psychotic disorders (SIP) and potential cognitive impairment following 3-MeO-PCP intake. This literature review aimed to summarize available evidence about 3-MeO-PCP mechanisms of action and physical and psychotropic effects and to spread preliminary findings about persistent psychotic symptoms and impaired cognitive functioning. Additionally, the case of an SIP is reported in a 29-year-old man with small oral intakes of 3-MeO-PCP over two weeks until a high dose ingestion. Psychometric and neuropsychological assessment and brain [18F]-fluorodeoxyglucose positron emission tomography integrated with computed tomography were used to support clinical description. Identifying and addressing the characteristic clinical features and neural substrates of NPS-induced psychoses might help clinicians with a more precise differentiation from other psychotic disorders. Although further studies are required, phenotyping the cognitive profile of NPS users might provide targets for tailored therapeutic approaches.
Collapse
Affiliation(s)
- Maria Pepe
- Department of Neuroscience, Section of Psychiatry, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168 Rome, Italy
- Department of Psychiatry, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy
| | - Marco Di Nicola
- Department of Neuroscience, Section of Psychiatry, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168 Rome, Italy
- Department of Psychiatry, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy
| | - Fabrizio Cocciolillo
- Nuclear Medicine Unit, Department of Diagnostic Imaging, Oncological Radiotherapy and Hematology, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy
| | - Stefania Chiappini
- Department of Neurosciences, Imaging and Clinical Sciences, Università degli Studi G. D’Annunzio, Chieti-Pescara, Via dei Vestini 31, 66013 Chieti, Italy
- School of Medical Sciences, UniCamillus International University of Medical Sciences, Via di S. Alessandro 8, 00131 Rome, Italy
| | - Giovanni Martinotti
- Department of Neurosciences, Imaging and Clinical Sciences, Università degli Studi G. D’Annunzio, Chieti-Pescara, Via dei Vestini 31, 66013 Chieti, Italy
- Psychopharmacology, Drug Misuse and Novel Psychoactive Substances Research Unit, School of Life and Medical Sciences, University of Hertfordshire, Hatfield AL10 9EU, UK
| | - Maria Lucia Calcagni
- Nuclear Medicine Unit, Department of Diagnostic Imaging, Oncological Radiotherapy and Hematology, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy
- Institute of Nuclear Medicine, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168 Rome, Italy
| | - Gabriele Sani
- Department of Neuroscience, Section of Psychiatry, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168 Rome, Italy
- Department of Psychiatry, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy
| |
Collapse
|
6
|
Dupont AC, Arlicot N, Vercouillie J, Serrière S, Maia S, Bonnet-Brilhault F, Santiago-Ribeiro MJ. Metabotropic Glutamate Receptor Subtype 5 Positron-Emission-Tomography Radioligands as a Tool for Central Nervous System Drug Development: Between Progress and Setbacks. Pharmaceuticals (Basel) 2023; 16:1127. [PMID: 37631042 PMCID: PMC10458693 DOI: 10.3390/ph16081127] [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: 07/04/2023] [Revised: 08/03/2023] [Accepted: 08/09/2023] [Indexed: 08/27/2023] Open
Abstract
The metabotropic glutamate receptor subtype 5 (mGluR5) is a class C G-protein-coupled receptor (GPCR) that has been implicated in various neuronal processes and, consequently, in several neuropsychiatric or neurodevelopmental disorders. Over the past few decades, mGluR5 has become a major focus for pharmaceutical companies, as an attractive target for drug development, particularly through the therapeutic potential of its modulators. In particular, allosteric binding sites have been targeted for better specificity and efficacy. In this context, Positron Emission Tomography (PET) appears as a useful tool for making decisions along a drug candidate's development process, saving time and money. Thus, PET provides quantitative information about a potential drug candidate and its target at the molecular level. However, in this area, particular attention has to be given to the interpretation of the PET signal and its conclusions. Indeed, the complex pharmacology of both mGluR5 and radioligands, allosterism, the influence of endogenous glutamate and the choice of pharmacokinetic model are all factors that may influence the PET signal. This review focuses on mGluR5 PET radioligands used at several stages of central nervous system drug development, highlighting advances and setbacks related to the complex pharmacology of these radiotracers.
Collapse
Affiliation(s)
- Anne-Claire Dupont
- Radiopharmacie, CHRU de Tours, 37000 Tours, France
- UMR 1253, iBrain, Tours University, INSERM, 37000 Tours, France
| | - Nicolas Arlicot
- Radiopharmacie, CHRU de Tours, 37000 Tours, France
- UMR 1253, iBrain, Tours University, INSERM, 37000 Tours, France
- CIC 1415, Tours University, INSERM, 37000 Tours, France
| | | | - Sophie Serrière
- UMR 1253, iBrain, Tours University, INSERM, 37000 Tours, France
| | - Serge Maia
- Radiopharmacie, CHRU de Tours, 37000 Tours, France
- UMR 1253, iBrain, Tours University, INSERM, 37000 Tours, France
| | - Frédérique Bonnet-Brilhault
- UMR 1253, iBrain, Tours University, INSERM, 37000 Tours, France
- Excellence Center for Autism and Neurodevelopmental Disorders, CHRU de Tours, 37000 Tours, France
| | - Maria-Joao Santiago-Ribeiro
- UMR 1253, iBrain, Tours University, INSERM, 37000 Tours, France
- Nuclear Medicine Department, CHRU de Tours, 37000 Tours, France
| |
Collapse
|
7
|
Jong YJI, Izumi Y, Harmon SK, Zorumski CF, ÓMalley KL. Striatal mGlu 5-mediated synaptic plasticity is independently regulated by location-specific receptor pools and divergent signaling pathways. J Biol Chem 2023; 299:104949. [PMID: 37354970 PMCID: PMC10388212 DOI: 10.1016/j.jbc.2023.104949] [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: 02/13/2023] [Revised: 06/05/2023] [Accepted: 06/07/2023] [Indexed: 06/26/2023] Open
Abstract
Metabotropic glutamate receptor 5 (mGlu5) is widely expressed throughout the central nervous system and is involved in neuronal function, synaptic transmission, and a number of neuropsychiatric disorders such as depression, anxiety, and autism. Recent work from this lab showed that mGlu5 is one of a growing number of G protein-coupled receptors that can signal from intracellular membranes where it drives unique signaling pathways, including upregulation of extracellular signal-regulated kinase (ERK1/2), ETS transcription factor Elk-1, and activity-regulated cytoskeleton-associated protein (Arc). To determine the roles of cell surface mGlu5 as well as the intracellular receptor in a well-known mGlu5 synaptic plasticity model such as long-term depression, we used pharmacological isolation and genetic and physiological approaches to analyze spatially restricted pools of mGlu5 in striatal cultures and slice preparations. Here we show that both intracellular and cell surface receptors activate the phosphatidylinositol-3-kinase-protein kinase B-mammalian target of rapamycin (PI3K/AKT/mTOR) pathway, whereas only intracellular mGlu5 activates protein phosphatase 2 and leads to fragile X mental retardation protein degradation and de novo protein synthesis followed by a protein synthesis-dependent increase in Arc and post-synaptic density protein 95. However, both cell surface and intracellular mGlu5 activation lead to α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor GluA2 internalization and chemically induced long-term depression albeit via different signaling mechanisms. These data underscore the importance of intracellular mGlu5 in the cascade of events associated with sustained synaptic transmission in the striatum.
Collapse
Affiliation(s)
- Yuh-Jiin I Jong
- Department of Neuroscience, Washington University School of Medicine, St Louis, Missouri, USA
| | - Yukitoshi Izumi
- Department of Psychiatry, Washington University School of Medicine, St Louis, Missouri, USA; The Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St Louis, Missouri, USA
| | - Steven K Harmon
- Department of Neuroscience, Washington University School of Medicine, St Louis, Missouri, USA
| | - Charles F Zorumski
- Department of Neuroscience, Washington University School of Medicine, St Louis, Missouri, USA; Department of Psychiatry, Washington University School of Medicine, St Louis, Missouri, USA; The Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St Louis, Missouri, USA
| | - Karen L ÓMalley
- Department of Neuroscience, Washington University School of Medicine, St Louis, Missouri, USA.
| |
Collapse
|
8
|
Forgham H, Liu L, Zhu J, Javed I, Cai W, Qiao R, Davis TP. Vector enabled CRISPR gene editing - A revolutionary strategy for targeting the diversity of brain pathologies. Coord Chem Rev 2023; 487:215172. [PMID: 37305445 PMCID: PMC10249757 DOI: 10.1016/j.ccr.2023.215172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Brain pathologies are considered one of the greatest contributors of death and disability worldwide. Neurodegenerative Alzheimer's disease is the second leading cause of death in adults, whilst brain cancers including glioblastoma multiforme in adults, and pediatric-type high-grade gliomas in children remain largely untreatable. A further compounding issue for patients with brain pathologies is that of long-term neuropsychiatric sequela - as a symptom or arising from high dose therapeutic intervention. The major challenge to effective, low dose treatment is finding therapeutics that successfully cross the blood-brain barrier and target aberrant cellular processes, while having minimum effect on essential cellular processes, and healthy bystander cells. Following over 30 years of research, CRISPR technology has emerged as a biomedical tour de force with the potential to revolutionise the treatment of both neurological and cancer related brain pathologies. The aim of this review is to take stock of the progress made in CRISPR technology in relation to treating brain pathologies. Specifically, we will describe studies which look beyond design, synthesis, and theoretical application; and focus instead on in vivo studies with translation potential. Along with discussing the latest breakthrough techniques being applied within the CRISPR field, we aim to provide a prospective on the knowledge gaps that exist and challenges that still lay ahead for CRISPR technology prior to successful application in the brain disease treatment field.
Collapse
Affiliation(s)
- Helen Forgham
- Australian Institute of Bioengineering & Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Liwei Liu
- Australian Institute of Bioengineering & Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Jiayuan Zhu
- Australian Institute of Bioengineering & Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Ibrahim Javed
- Australian Institute of Bioengineering & Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Weibo Cai
- Departments of Radiology and Medical Physics, University of Wisconsin – Madison, Madison, WI, USA
| | - Ruirui Qiao
- Australian Institute of Bioengineering & Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Thomas P. Davis
- Australian Institute of Bioengineering & Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| |
Collapse
|
9
|
Holmes SE, Asch RH, Davis MT, DellaGioia N, Pashankar N, Gallezot JD, Nabulsi N, Matuskey D, Sanacora G, Carson RE, Blumberg HP, Esterlis I. Differences in Quantification of the Metabotropic Glutamate Receptor 5 Across Bipolar Disorder and Major Depressive Disorder. Biol Psychiatry 2023; 93:1099-1107. [PMID: 36764853 PMCID: PMC10164841 DOI: 10.1016/j.biopsych.2022.10.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 10/06/2022] [Accepted: 10/30/2022] [Indexed: 11/09/2022]
Abstract
BACKGROUND Understanding the neurobiology underlying bipolar disorder (BD) versus major depressive disorder (MDD) is crucial for accurate diagnosis and for driving the discovery of novel treatments. A promising target is the metabotropic glutamate receptor 5 (mGluR5), a modulator of glutamate transmission associated with synaptic plasticity. We measured mGluR5 availability in individuals with MDD and BD for the first time using positron emission tomography. METHODS Individuals with BD (n = 17 depressed; n = 10 euthymic) or MDD (n = 17) and healthy control (HC) individuals (n = 18) underwent imaging with [18F]FPEB positron emission tomography to quantify mGluR5 availability in regions of the prefrontal cortex, which was compared across groups and assessed in relation to depressive symptoms and cognitive function. RESULTS Prefrontal cortex mGluR5 availability was significantly different across groups (F6,116 = 2.18, p = .050). Specifically, mGluR5 was lower in BD versus MDD and HC groups, with no difference between MDD and HC groups. Furthermore, after dividing the BD group, mGluR5 was lower in both BD-depression and BD-euthymia groups versus both MDD and HC groups across regions of interest. Interestingly, lower dorsolateral prefrontal cortex mGluR5 was associated with worse depression in MDD (r = -0.67, p = .005) but not in BD. Significant negative correlations were observed between mGluR5 and working memory in MDD and BD-depression groups. CONCLUSIONS This work suggests that mGluR5 could be helpful in distinguishing BD and MDD as a possible treatment target for depressive symptoms in MDD and for cognitive alterations in both disorders. Further work is needed to confirm differentiating roles for mGluR5 in BD and MDD and to probe modulation of mGluR5 as a preventive/treatment strategy.
Collapse
Affiliation(s)
- Sophie E Holmes
- Department of Psychiatry, Yale School of Medicine, New Haven, Connecticut
| | - Ruth H Asch
- Department of Psychiatry, Yale School of Medicine, New Haven, Connecticut
| | - Margaret T Davis
- Department of Psychiatry, Yale School of Medicine, New Haven, Connecticut; Department of Psychology, Yale University, New Haven, Connecticut
| | - Nicole DellaGioia
- Department of Psychiatry, Yale School of Medicine, New Haven, Connecticut
| | - Neha Pashankar
- Department of Psychiatry, Yale School of Medicine, New Haven, Connecticut
| | - Jean-Dominique Gallezot
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut
| | - Nabeel Nabulsi
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut
| | - David Matuskey
- Department of Psychiatry, Yale School of Medicine, New Haven, Connecticut
| | - Gerard Sanacora
- Department of Psychiatry, Yale School of Medicine, New Haven, Connecticut
| | - Richard E Carson
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut
| | - Hilary P Blumberg
- Department of Psychiatry, Yale School of Medicine, New Haven, Connecticut; Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut; Child Study Center, Yale School of Medicine, New Haven, Connecticut
| | - Irina Esterlis
- Department of Psychiatry, Yale School of Medicine, New Haven, Connecticut; Child Study Center, Yale School of Medicine, New Haven, Connecticut; Clinical Neurosciences Division, U.S. Department of Veteran Affairs National Center for Posttraumatic Stress Disorder, VA Connecticut Healthcare System, West Haven, Connecticut.
| |
Collapse
|
10
|
Böttger SJ, Förstner BR, Szalek L, Koller-Schlaud K, Rapp MA, Tschorn M. Mood and anxiety disorders within the Research Domain Criteria framework of Positive and Negative Valence Systems: a scoping review. Front Hum Neurosci 2023; 17:1184978. [PMID: 37333832 PMCID: PMC10272468 DOI: 10.3389/fnhum.2023.1184978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 05/02/2023] [Indexed: 06/20/2023] Open
Abstract
Introduction While a growing body of research is adopting Research Domain Criteria (RDoC)-related methods and constructs, there is still a lack of comprehensive reviews on the state of published research on Positive Valence Systems (PVS) and Negative Valence Systems (NVS) in mood and anxiety disorders consistent with the RDoC framework. Methods Five electronic databases were searched to identify peer-reviewed publications covering research on "positive valence" and "negative valence" as well as "valence," "affect," and "emotion" for individuals with symptoms of mood and anxiety disorders. Data was extracted with a focus on disorder, domain, (sub-) constructs, units of analysis, key results, and study design. Findings are presented along four sections, distinguishing between primary articles and reviews each for PVS, NVS, and cross-domain PVS and NVS. Results A total of 231 abstracts were identified, and 43 met the inclusion criteria for this scoping review. Seventeen publications addressed research on PVS, seventeen on NVS, and nine covered cross-domain research on PVS and NVS. Psychological constructs were typically examined across different units of analysis, with the majority of publications incorporating two or more measures. Molecular, genetic, and physiological aspects were mainly investigated via review articles, primary articles focused on self-report, behavioral, and, to a lesser extent, physiological measures. Conclusions This present scoping review shows that mood and anxiety disorders were actively studied using a range of genetic, molecular, neuronal, physiological, behavioral, and self-report measures within the RDoC PVS and NVS. Results highlight the essential role of specific cortical frontal brain structures and of subcortical limbic structures in impaired emotional processing in mood and anxiety disorders. Findings also indicate overall limited research on NVS in bipolar disorders and PVS in anxiety disorders, a majority of self-report studies, and predominantly observational studies. Future research is needed to develop more RDoC-consistent advancements and intervention studies targeting neuroscience-driven PVS and NVS constructs.
Collapse
Affiliation(s)
- Sarah Jane Böttger
- Social and Preventive Medicine, Department of Sports and Health Sciences, University of Potsdam, Potsdam, Germany
- DZPG (German Center of Mental Health), partner site Berlin/Potsdam, Potsdam, Germany
| | - Bernd R. Förstner
- Social and Preventive Medicine, Department of Sports and Health Sciences, University of Potsdam, Potsdam, Germany
- DZPG (German Center of Mental Health), partner site Berlin/Potsdam, Potsdam, Germany
| | - Laura Szalek
- Social and Preventive Medicine, Department of Sports and Health Sciences, University of Potsdam, Potsdam, Germany
| | - Kristin Koller-Schlaud
- Department of Psychiatry, Psychotherapy and Psychosomatics, Brandenburg Medical School, University Hospital Ruppin-Brandenburg, Neuruppin, Germany
| | - Michael A. Rapp
- Social and Preventive Medicine, Department of Sports and Health Sciences, University of Potsdam, Potsdam, Germany
- DZPG (German Center of Mental Health), partner site Berlin/Potsdam, Potsdam, Germany
| | - Mira Tschorn
- Social and Preventive Medicine, Department of Sports and Health Sciences, University of Potsdam, Potsdam, Germany
- DZPG (German Center of Mental Health), partner site Berlin/Potsdam, Potsdam, Germany
| |
Collapse
|
11
|
Colyer-Patel K, Kuhns L, Weidema A, Lesscher H, Cousijn J. Age-dependent effects of tobacco smoke and nicotine on cognition and the brain: A systematic review of the human and animal literature comparing adolescents and adults. Neurosci Biobehav Rev 2023; 146:105038. [PMID: 36627063 DOI: 10.1016/j.neubiorev.2023.105038] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 10/21/2022] [Accepted: 01/05/2023] [Indexed: 01/09/2023]
Abstract
Cigarette smoking is often initiated during adolescence and an earlier age of onset is associated with worse health outcomes later in life. Paradoxically, the transition towards adulthood also marks the potential for recovery, as the majority of adolescents are able to quit smoking when adulthood emerges. This systematic review aimed to evaluate the evidence from both human and animal studies for the differential impact of adolescent versus adult repeated and long-term tobacco and nicotine exposure on cognitive and brain outcomes. The limited human studies and more extensive yet heterogeneous animal studies, provide preliminary evidence of heightened fear learning, anxiety-related behaviour, reward processing, nicotinic acetylcholinergic receptors expression, dopamine expression and serotonin functioning after adolescent compared to adult exposure. Effects of nicotine or tobacco use on impulsivity were comparable across age groups. These findings provide novel insights into the mechanisms underlying adolescents' vulnerability to tobacco and nicotine. Future research is needed to translate animal to human findings, with a focus on directly linking a broader spectrum of brain and behavioural outcomes.
Collapse
Affiliation(s)
- Karis Colyer-Patel
- Neuroscience of Addiction (NofA) Lab, Department of Psychology, Education & Child Studies, Erasmus University Rotterdam, the Netherlands.
| | - Lauren Kuhns
- Department of Psychology, University of Amsterdam, Amsterdam, the Netherlands
| | - Alix Weidema
- Neuroscience of Addiction (NofA) Lab, Department of Psychology, Education & Child Studies, Erasmus University Rotterdam, the Netherlands
| | - Heidi Lesscher
- Department Population Health Sciences, Animals in Science and Society, Division of Behavioural Neuroscience, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Janna Cousijn
- Neuroscience of Addiction (NofA) Lab, Department of Psychology, Education & Child Studies, Erasmus University Rotterdam, the Netherlands; Department of Psychology, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
12
|
Puglisi-Allegra S, Lazzeri G, Busceti CL, Giorgi FS, Biagioni F, Fornai F. Lithium engages autophagy for neuroprotection and neuroplasticity: translational evidence for therapy. Neurosci Biobehav Rev 2023; 148:105148. [PMID: 36996994 DOI: 10.1016/j.neubiorev.2023.105148] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/23/2023] [Accepted: 03/26/2023] [Indexed: 03/30/2023]
Abstract
Here an overview is provided on therapeutic/neuroprotective effects of Lithium (Li+) in neurodegenerative and psychiatric disorders focusing on the conspicuous action of Li+ through autophagy. The effects on the autophagy machinery remain the key molecular mechanisms to explain the protective effects of Li+ for neurodegenerative diseases, offering potential therapeutic strategies for the treatment of neuropsychiatric disorders and emphasizes a crossroad linking autophagy, neurodegenerative disorders, and mood stabilization. Sensitization by psychostimulants points to several mechanisms involved in psychopathology, most also crucial in neurodegenerative disorders. Evidence shows the involvement of autophagy and metabotropic Glutamate receptors-5 (mGluR5) in neurodegeneration due to methamphetamine neurotoxicity as well as in neuroprotection, both in vitro and in vivo models. More recently, Li+ was shown to modulate autophagy through its action on mGluR5, thus pointing to an additional way of autophagy engagement by Li+ and to a substantial role of mGluR5 in neuroprotection related to neural e neuropsychiatry diseases. We propose Li+ engagement of autophagy through the canonical mechanisms of autophagy machinery and through the intermediary of mGluR5.
Collapse
|
13
|
Krzyczmonik A, Grafinger KE, Keller T, Pfeifer L, Forsback S, Haaparanta-Solin M, Gouverneur V, López-Picón F, Solin O. Evaluation of [ 18F]FMTEB in Sprague Dawley rats as a PET tracer for metabotropic glutamate receptor 5. Nucl Med Biol 2023; 116-117:108309. [PMID: 36521341 DOI: 10.1016/j.nucmedbio.2022.108309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 10/31/2022] [Accepted: 11/17/2022] [Indexed: 11/27/2022]
Abstract
INTRODUCTION [18F]FMTEB, along with other tracers, was developed as a promising PET radioligand for imaging metabotropic glutamate receptor subtype 5 (mGluR5). Despite favorable preliminary results, it has not been used further for studies of mGluR5. This paper presents an in-depth preclinical evaluation of [18F]FMTEB in healthy Sprague Dawley rats. METHODS [18F]FMTEB was synthesized from a boronic ester precursor using copper-mediated fluorination. In vivo PET imaging was performed on six rats, of which three were pre-treated with a high affinity mGluR5 receptor antagonist. An additional 18 rats were used for ex vivo experiments for metabolite analyses in plasma, brain and urine, and for biodistribution and ex vivo brain autoradiography at different time points. RESULTS [18F]FMTEB was synthesized in adequate radiochemical yield and a molar activity of 154 ± 64 GBq/μmol. Both in vivo imaging and ex vivo brain autoradiography showed high specificity for mGluR5, and the blocking experiments showed a clear decrease in radioactivity in mGluR5-rich brain areas. Metabolite analyses confirmed fast metabolism of the tracer in plasma. The percentage of parent compound in brain tissue exceeded 90 % up to 90 min after injection. CONCLUSION [18F]FMTEB produced via copper-mediated 18F-fluorination fulfilled the requirements for preclinical evaluation in rats. The absence of specific uptake in cerebellum and absence of defluorination of the tracer allowed cerebellum to be used as a reference tissue. Due to the fast kinetics in rats, the region-to-cerebellum ratios equilibrated within 30 min. These results prove [18F]FMTEB to be a good candidate for mapping mGluR5 in rat brain and a suitable alternative to [18F]FPEB.
Collapse
Affiliation(s)
- Anna Krzyczmonik
- Turku PET Centre, Radiopharmaceutical Chemistry Laboratory, University of Turku, Turku, Finland
| | - Katharina E Grafinger
- Turku PET Centre, Preclinical Imaging, University of Turku, Turku, Finland; MediCity Research Laboratory, University of Turku, Turku, Finland
| | - Thomas Keller
- Turku PET Centre, Radiopharmaceutical Chemistry Laboratory, University of Turku, Turku, Finland
| | - Lukas Pfeifer
- University of Oxford, Chemistry Research Laboratory, Oxford, United Kingdom
| | - Sarita Forsback
- Turku PET Centre, Radiopharmaceutical Chemistry Laboratory, University of Turku, Turku, Finland; Department of Chemistry, University of Turku, Turku, Finland
| | - Merja Haaparanta-Solin
- Turku PET Centre, Preclinical Imaging, University of Turku, Turku, Finland; MediCity Research Laboratory, University of Turku, Turku, Finland
| | | | - Francisco López-Picón
- Turku PET Centre, Preclinical Imaging, University of Turku, Turku, Finland; MediCity Research Laboratory, University of Turku, Turku, Finland
| | - Olof Solin
- Turku PET Centre, Radiopharmaceutical Chemistry Laboratory, University of Turku, Turku, Finland; Department of Chemistry, University of Turku, Turku, Finland; Turku PET Centre, Accelerator Laboratory, Åbo Akademi University, Turku, Finland.
| |
Collapse
|
14
|
Asch RH, Hillmer AT, Baldassarri SR, Esterlis I. The metabotropic glutamate receptor 5 as a biomarker for psychiatric disorders. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2023; 168:265-310. [PMID: 36868631 DOI: 10.1016/bs.irn.2022.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The role of glutamate system in the etiology and pathophysiology of psychiatric disorders has gained considerable attention in the past two decades, including dysregulation of the metabotropic glutamatergic receptor subtype 5 (mGlu5). Thus, mGlu5 may represent a promising therapeutic target for psychiatric conditions, particularly stress-related disorders. Here, we describe mGlu5 findings in mood disorders, anxiety, and trauma disorders, as well as substance use (specifically nicotine, cannabis, and alcohol use). We highlight insights gained from positron emission tomography (PET) studies, where possible, and discuss findings from treatment trials, when available, to explore the role of mGlu5 in these psychiatric disorders. Through the research evidence reviewed in this chapter, we make the argument that, not only is dysregulation of mGlu5 evident in numerous psychiatric disorders, potentially functioning as a disease "biomarker," the normalization of glutamate neurotransmission via changes in mGlu5 expression and/or modulation of mGlu5 signaling may be a needed component in treating some psychiatric disorders or symptoms. Finally, we hope to demonstrate the utility of PET as an important tool for investigating mGlu5 in disease mechanisms and treatment response.
Collapse
Affiliation(s)
- Ruth H Asch
- Department of Psychiatry, Yale University, New Haven, CT, United States.
| | - Ansel T Hillmer
- Department of Psychiatry, Yale University, New Haven, CT, United States; Department of Radiology and Biomedical Imaging, New Haven, CT, United States
| | - Stephen R Baldassarri
- Yale Program in Addiction Medicine, Yale University, New Haven, CT, United States; Department of Internal Medicine, Yale University, New Haven, CT, United States
| | - Irina Esterlis
- Department of Psychiatry, Yale University, New Haven, CT, United States; Department of Psychology, Yale University, New Haven, CT, United States; Clinical Neurosciences Division, U.S. Department of Veterans Affairs National Center for Posttraumatic Stress Disorder, Veterans Affairs Connecticut Healthcare System, West Haven, CT, United States
| |
Collapse
|
15
|
Ray SK, Mukherjee S. Neuropharmacology of Alcohol Addiction with Special Emphasis on Proteomic Approaches for Identification of Novel Therapeutic Targets. Curr Neuropharmacol 2023; 21:119-132. [PMID: 35959616 PMCID: PMC10193758 DOI: 10.2174/1570159x20666220811092906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 07/01/2022] [Accepted: 07/10/2022] [Indexed: 02/04/2023] Open
Abstract
Alcohol is a generic pharmacological agent with only a few recognized primary targets. Nmethyl- D-aspartate, gamma-aminobutyric acid, glycine, 5-hydroxytryptamine 3 (serotonin), nicotinic acetylcholine receptors, and L-type Ca2+ channels and G-protein-activated inwardly rectifying K channels are all involved. Following the first hit of alcohol on specific brain targets, the second wave of indirect effects on various neurotransmitter/neuropeptide systems begins, leading to the typical acute behavioral effects of alcohol, which range from disinhibition to sedation and even hypnosis as alcohol concentrations rise. Recent research has revealed that gene regulation is significantly more complex than previously thought and does not fully explain changes in protein levels. As a result, studying the proteome directly, which differs from the genome/transcriptome in terms of complexity and dynamicity, has provided unique insights into extraordinary advances in proteomic techniques that have changed the way we can analyze the composition, regulation, and function of protein complexes and pathways underlying altered neurobiological conditions. Neuroproteomics has the potential to revolutionize alcohol research by allowing researchers to gain a better knowledge of how alcohol impacts protein structure, function, connections, and networks on a global scale. The amount of information collected from these breakthroughs can aid in identifying valuable biomarkers for early detection and improved prognosis of an alcohol use disorder and future pharmaceutical targets for the treatment of alcoholism.
Collapse
Affiliation(s)
- Suman Kumar Ray
- Independent Researcher, Bhopal, Madhya Pradesh 462020, India
| | - Sukhes Mukherjee
- Department of Biochemistry, All India Institute of Medical Science, Bhopal, Madhya Pradesh 462020, India
| |
Collapse
|
16
|
Baldassarri SR, Asch RH, Hillmer AT, Pietrzak RH, DellaGioia N, Esterlis I, Davis MT. Nicotine Use and Metabotropic Glutamate Receptor 5 in Individuals With Major Depressive and Posttraumatic Stress Disorders. CHRONIC STRESS (THOUSAND OAKS, CALIF.) 2023; 7:24705470231154842. [PMID: 36843572 PMCID: PMC9943964 DOI: 10.1177/24705470231154842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 01/18/2023] [Indexed: 02/12/2023]
Abstract
Metabotropic glutamate receptor 5 (mGluR5) dysregulation has been implicated in the pathophysiology of many psychiatric disorders, as well as nicotine use and dependence. We used positron emission tomography with [18F]FPEB to measure mGluR5 availability in vivo in 6 groups: (1) nicotine users (NUs) without other psychiatric comorbidities (n = 23); (2) comparison controls (CCs) without nicotine use or psychiatric comorbidities (n = 38); (3) major depressive disorder subjects with concurrent nicotine use (MDD-NU; n = 19); (4) MDD subjects without concurrent nicotine use (MDD-CC; n = 20); (5) posttraumatic stress disorder subjects with concurrent nicotine use (PTSD-NU; n = 17); and (6) PTSD subjects without concurrent nicotine use (PTSD-CC; n = 16). The goal of the study was to test the hypothesis that mGluR5 availability in key corticolimbic regions of interest (ROIs) is different in NU with versus without comorbid psychiatric disorders (ROI: dorsolateral prefrontal cortex [dlPFC], orbitofrontal cortex [OFC], ventromedial prefrontal cortex [vmPFC], anterior cingulate cortex [ACC], amygdala, hippocampus). We found that NU had 11%-13% lower mGluR5 availability in OFC, vmPFC, dlPFC, and ACC as compared with CC, while PTSD-NU had 9%-11% higher mGluR5 availability in OFC, dlPFC, and ACC compared with PTSD. Furthermore, relationships between mGluR5 availability and psychiatric symptoms varied as a function of psychiatric diagnosis among NUs. NU showed a negative correlation between mGluR5 and smoking cravings and urges (r's = -0.58 to -0.70, p's = 0.011 - 0.047), while PTSD-NU had the reverse relationship (r's = 0.60-0.71, p's = 0.013-0.035 in ACC, vmPFC, and dlPFC). These findings have substantial implications for our understanding of glutamate homeostasis in psychiatric subgroups and for identifying key neural phenotypes among NU. mGluR5 is a potential treatment target for precision medicine in individuals with nicotine use.
Collapse
Affiliation(s)
- Stephen R. Baldassarri
- Section of Pulmonary, Critical Care, & Sleep Medicine,
Department of Internal Medicine, Yale University School of
Medicine, New Haven, CT, USA
- Program in Addiction Medicine, Yale University School of
Medicine, New Haven, CT, USA
| | - Ruth H. Asch
- Departments of Psychiatry, Yale University School of
Medicine, New Haven, CT, USA
| | - Ansel T. Hillmer
- Departments of Psychiatry, Yale University School of
Medicine, New Haven, CT, USA
- Department of Radiology and Biomedical Imaging, Radiology, and
Biomedical Imaging, New Haven, CT, USA
| | - Robert H. Pietrzak
- Departments of Psychiatry, Yale University School of
Medicine, New Haven, CT, USA
- VA National Center for PTSD Clinical Neurosciences Division, New
Haven, CT, USA
| | - Nicole DellaGioia
- Departments of Psychiatry, Yale University School of
Medicine, New Haven, CT, USA
| | - Irina Esterlis
- Departments of Psychiatry, Yale University School of
Medicine, New Haven, CT, USA
- VA National Center for PTSD Clinical Neurosciences Division, New
Haven, CT, USA
| | - Margaret T. Davis
- Departments of Psychiatry, Yale University School of
Medicine, New Haven, CT, USA
| |
Collapse
|
17
|
The neuroprotective and neuroplastic potential of glutamatergic therapeutic drugs in bipolar disorder. Neurosci Biobehav Rev 2022; 142:104906. [DOI: 10.1016/j.neubiorev.2022.104906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 09/30/2022] [Accepted: 10/02/2022] [Indexed: 11/21/2022]
|
18
|
Postsynaptic Proteins at Excitatory Synapses in the Brain—Relationship with Depressive Disorders. Int J Mol Sci 2022; 23:ijms231911423. [PMID: 36232725 PMCID: PMC9569598 DOI: 10.3390/ijms231911423] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 09/21/2022] [Accepted: 09/22/2022] [Indexed: 11/17/2022] Open
Abstract
Depressive disorders (DDs) are an increasingly common health problem that affects all age groups. DDs pathogenesis is multifactorial. However, it was proven that stress is one of the most important environmental factors contributing to the development of these conditions. In recent years, there has been growing interest in the role of the glutamatergic system in the context of pharmacotherapy of DDs. Thus, it has become increasingly important to explore the functioning of excitatory synapses in pathogenesis and pharmacological treatment of psychiatric disorders (including DDs). This knowledge may lead to the description of new mechanisms of depression and indicate new potential targets for the pharmacotherapy of illness. An excitatory synapse is a highly complex and very dynamic structure, containing a vast number of proteins. This review aimed to discuss in detail the role of the key postsynaptic proteins (e.g., NMDAR, AMPAR, mGluR5, PSD-95, Homer, NOS etc.) in the excitatory synapse and to systematize the knowledge about changes that occur in the clinical course of depression and after antidepressant treatment. In addition, a discussion on the potential use of ligands and/or modulators of postsynaptic proteins at the excitatory synapse has been presented.
Collapse
|
19
|
From bench to bedside: The mGluR5 system in people with and without Autism Spectrum Disorder and animal model systems. Transl Psychiatry 2022; 12:395. [PMID: 36127322 PMCID: PMC9489881 DOI: 10.1038/s41398-022-02143-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 08/19/2022] [Accepted: 09/01/2022] [Indexed: 11/08/2022] Open
Abstract
The metabotropic glutamate receptor 5 (mGluR5) is a key regulator of excitatory (E) glutamate and inhibitory (I) γ-amino butyric acid (GABA) signalling in the brain. Despite the close functional ties between mGluR5 and E/I signalling, no-one has directly examined the relationship between mGluR5 and glutamate or GABA in vivo in the human brain of autistic individuals. We measured [18F] FPEB (18F-3-fluoro-5-[(pyridin-3-yl)ethynyl]benzonitrile) binding in 15 adults (6 with Autism Spectrum Disorder) using two regions of interest, the left dorsomedial prefrontal cortex and a region primarily composed of left striatum and thalamus. These two regions were mapped out using MEGA-PRESS voxels and then superimposed on reconstructed PET images. This allowed for direct comparison between mGluR5, GABA + and Glx. To better understand the molecular underpinnings of our results we used an autoradiography study of mGluR5 in three mouse models associated with ASD: Cntnap2 knockout, Shank3 knockout, and 16p11.2 deletion. Autistic individuals had significantly higher [18F] FPEB binding (t (13) = -2.86, p = 0.047) in the left striatum/thalamus region of interest as compared to controls. Within this region, there was a strong negative correlation between GABA + and mGluR5 density across the entire cohort (Pearson's correlation: r (14) = -0.763, p = 0.002). Cntnap2 KO mice had significantly higher mGlu5 receptor binding in the striatum (caudate-putamen) as compared to wild-type (WT) mice (n = 15, p = 0.03). There were no differences in mGluR5 binding for mice with the Shank3 knockout or 16p11.2 deletion. Given that Cntnap2 is associated with a specific striatal deficit of parvalbumin positive GABA interneurons and 'autistic' features, our findings suggest that an increase in mGluR5 in ASD may relate to GABAergic interneuron abnormalities.
Collapse
|
20
|
Hadizadeh H, Flores JM, Mayerson T, Worhunsky PD, Potenza MN, Angarita GA. Glutamatergic Agents for the Treatment of Cocaine Use Disorder. Curr Behav Neurosci Rep 2022. [DOI: 10.1007/s40473-022-00252-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
21
|
Kim J, Kang S, Choi TY, Chang KA, Koo JW. Metabotropic Glutamate Receptor 5 in Amygdala Target Neurons Regulates Susceptibility to Chronic Social Stress. Biol Psychiatry 2022; 92:104-115. [PMID: 35314057 DOI: 10.1016/j.biopsych.2022.01.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 01/10/2022] [Accepted: 01/13/2022] [Indexed: 12/25/2022]
Abstract
BACKGROUND Metabotropic glutamate receptor 5 (mGluR5) has been implicated in stress-related psychiatric disorders, particularly major depressive disorder. Although growing evidence supports the proresilient role of mGluR5 in corticolimbic circuitry in the depressive-like behaviors following chronic stress exposure, the underlying neural mechanisms, including circuits and molecules, remain unknown. METHODS We measured the c-Fos expression and probability of neurotransmitter release in and from basolateral amygdala (BLA) neurons projecting to the medial prefrontal cortex (mPFC) and to the ventral hippocampus (vHPC) after chronic social defeat stress. The role of BLA projections in depressive-like behaviors was assessed using optogenetic manipulations, and the underlying molecular mechanisms of mGluR5 and downstream signaling were investigated by Western blotting, viral-mediated gene transfer, and pharmacological manipulations. RESULTS Chronic social defeat stress disrupted neural activity and glutamatergic transmission in both BLA projections. Optogenetic activation of BLA projections reversed the detrimental effects of chronic social defeat stress on depressive-like behaviors and mGluR5 expression in the mPFC and vHPC. Conversely, inhibition of BLA projections of mice undergoing subthreshold social defeat stress induced a susceptible phenotype and mGluR5 reduction. These two BLA circuits appeared to act in an independent way. We demonstrate that mGluR5 overexpression in the mPFC or vHPC was proresilient while the mGluR5 knockdown was prosusceptible and that the proresilient effects of mGluR5 are mediated through distinctive downstream signaling pathways in the mPFC and vHPC. CONCLUSIONS These findings identify mGluR5 in the mPFC and vHPC that receive BLA inputs as a critical mediator of stress resilience, highlighting circuit-specific signaling for depressive-like behaviors.
Collapse
Affiliation(s)
- Jeongseop Kim
- Emotion, Cognition and Behavior Research Group, Korea Brain Research Institute, Daegu, Republic of Korea; Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea
| | - Shinwoo Kang
- Department of Pharmacology, College of Medicine, Gachon University, Incheon, Republic of Korea; Neuroscience Research Institute, Gachon University, Incheon, Republic of Korea; Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, Republic of Korea; Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
| | - Tae-Yong Choi
- Emotion, Cognition and Behavior Research Group, Korea Brain Research Institute, Daegu, Republic of Korea
| | - Keun-A Chang
- Department of Pharmacology, College of Medicine, Gachon University, Incheon, Republic of Korea; Neuroscience Research Institute, Gachon University, Incheon, Republic of Korea; Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, Republic of Korea.
| | - Ja Wook Koo
- Emotion, Cognition and Behavior Research Group, Korea Brain Research Institute, Daegu, Republic of Korea; Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea.
| |
Collapse
|
22
|
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
|
23
|
Maudes E, Mannara F, García-Serra A, Radosevic M, Mellado A, Serafim AB, Planagumà J, Sabater L, Dalmau J, Spatola M. Human mGluR5 antibodies alter receptor levels and behavior in mice. Ann Neurol 2022; 92:81-86. [PMID: 35373379 DOI: 10.1002/ana.26362] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 03/24/2022] [Accepted: 03/26/2022] [Indexed: 11/08/2022]
Abstract
The Ophelia syndrome or encephalitis with antibodies against the metabotropic glutamate receptor 5 (mGluR5) manifests with behavioral changes, memory deficits and anxiety. To study the antibody pathogenicity, mice received continuous cerebroventricular infusion of patients' or controls' IgG for 14 days, followed by a 15-day wash-out. The effects on hippocampal mGluR5 clusters were determined by confocal microscopy. Animals infused with patients' IgG, but not controls' IgG, showed memory impairment, increased anxiety, and decreased neuronal surface mGluR5 clusters. After antibody clearance, both behavioral and molecular changes reversed to baseline conditions. These findings support the pathogenicity of these antibodies in anti-mGluR5 encephalitis. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Estibaliz Maudes
- Neuroimmunology Program, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| | - Francesco Mannara
- Neuroimmunology Program, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| | - Anna García-Serra
- Neuroimmunology Program, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| | - Marija Radosevic
- Neuroimmunology Program, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| | - Araceli Mellado
- Neuroimmunology Program, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| | - Ana Beatriz Serafim
- Neuroimmunology Program, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| | - Jesús Planagumà
- Neuroimmunology Program, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| | - Lidia Sabater
- Neuroimmunology Program, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| | - Josep Dalmau
- Neuroimmunology Program, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Universitat de Barcelona, Barcelona, Spain.,Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA.,Centro de Investigación Biomédica en Red, Enfermedades Raras (CIBERER), Spain.,Catalan Institute for Research and Advanced Studies (ICREA), Barcelona, Spain
| | - Marianna Spatola
- Neuroimmunology Program, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Universitat de Barcelona, Barcelona, Spain.,Ragon Institute of MGH, MIT and Harvard Medical School, Cambridge, MA, USA
| |
Collapse
|
24
|
Kim JH, Joo YH, Son YD, Kim HK, Kim JH. Differences in mGluR5 Availability Depending on the Level of Social Avoidance in Drug-Naïve Young Patients with Major Depressive Disorder. Neuropsychiatr Dis Treat 2022; 18:2041-2053. [PMID: 36124236 PMCID: PMC9481450 DOI: 10.2147/ndt.s379395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 09/03/2022] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Previous research has shown that metabotropic glutamate receptor-5 (mGluR5) signaling is significantly involved in social avoidance. We investigated the relationship between levels of social avoidance and mGluR5 availability in drug-naïve young patients with major depressive disorder (MDD). METHODS Twenty non-smoking patients and eighteen matched non-smoking healthy controls underwent [11C]ABP688 positron emission tomography (PET) and magnetic resonance imaging scans. The binding potential (BPND) of [11C]ABP688 was obtained using the simplified reference tissue model. Patients' level of social avoidance was assessed using the Social Avoidance and Distress Scale (SADS). For [11C]ABP688 BPND, the region-of-interest (ROI)-based between-group comparisons and correlations with SADS scores were investigated. The frontal cortices were chosen as a priori ROIs based on previous PET investigations in MDD, and on literature underscoring the importance of the frontal cortex in social avoidance. RESULTS Independent samples t-tests revealed no significant differences in [11C]ABP688 BPND in the frontal cortices between the MDD patient group as a whole and healthy controls. One-way analysis of variance with post-hoc tests revealed significantly lower BPND in the bilateral superior frontal cortex (SFC) and left middle frontal cortex (MFC) in MDD patients with low levels of social avoidance (L-SADS) than in healthy controls. The L-SADS patients also had significantly lower BPND in the medial part of the right SFC than both MDD patients with high levels of social avoidance (H-SADS) and healthy controls. The L-SADS patients also showed significantly lower BPND in the orbital parts of the SFC, MFC, and inferior frontal cortex than H-SADS patients. No significant group differences were found between H-SADS patients and healthy controls. The ROI-based correlation analysis revealed significant positive correlations between social avoidance levels and frontal [11C]ABP688 BPND in the entire patients. CONCLUSION Our exploratory study shows significant differences in frontal mGluR5 availability depending on the level of social avoidance in drug-naïve non-smoking MDD patients, suggesting that social avoidance should be considered as one of the clinical factors involved in mGluR5 signaling changes in depression.
Collapse
Affiliation(s)
- Jeong-Hee Kim
- Neuroscience Research Institute, Gachon University, Incheon, Republic of Korea.,Department of Biomedical Engineering, College of Health Science, Gachon University, Incheon, Republic of Korea
| | - Yo-Han Joo
- Neuroscience Research Institute, Gachon University, Incheon, Republic of Korea
| | - Young-Don Son
- Neuroscience Research Institute, Gachon University, Incheon, Republic of Korea.,Department of Biomedical Engineering, College of Health Science, Gachon University, Incheon, Republic of Korea.,Gachon Advanced Institute for Health Sciences & Technology, Gachon University, Incheon, Republic of Korea
| | - Hang-Keun Kim
- Neuroscience Research Institute, Gachon University, Incheon, Republic of Korea.,Department of Biomedical Engineering, College of Health Science, Gachon University, Incheon, Republic of Korea.,Gachon Advanced Institute for Health Sciences & Technology, Gachon University, Incheon, Republic of Korea
| | - Jong-Hoon Kim
- Neuroscience Research Institute, Gachon University, Incheon, Republic of Korea.,Gachon Advanced Institute for Health Sciences & Technology, Gachon University, Incheon, Republic of Korea.,Department of Psychiatry, Gachon University College of Medicine, Gil Medical Center, Incheon, Republic of Korea
| |
Collapse
|
25
|
Stachowicz K, Bobula B, Kusek M, Lenda T, Tokarski K. Evidence for the interaction of COX-2 with mGluR5 in the regulation of EAAT1 and EAAT3 protein levels in the mouse hippocampus. The influence of oxidative stress mechanisms. Brain Res 2021; 1771:147660. [PMID: 34529964 DOI: 10.1016/j.brainres.2021.147660] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 08/17/2021] [Accepted: 09/07/2021] [Indexed: 12/29/2022]
Abstract
Since we found that inhibition of cyclooxygenase-2 (COX-2) with concomitant application of a metabotropic glutamate receptor subtype 5 (mGluR5) antagonist (MTEP) down-regulates mGluR7 in the hippocampus (HC) and changes behavior of mice, our team decided to investigate the mechanism responsible for the observed changes. The amino acid glutamate (Glu) is a major excitatory neurotransmitter in the brain. Glu uptake is regulated by excitatory amino acid transporters (EAAT). There are five transporters with documented expression in neurons and glia in the central nervous system (CNS). EAATs, maintain the correct transmission of the Glu signal and prevent its toxic accumulation by removing Glu from the synapse. It has been documented that the toxic level of Glu is one of the main causes of mental and cognitive abnormalities. Given the above mechanisms involved in the functioning of the Glu synapse, we hypothesized modification of Glu uptake, involving EAATs as the cause of the observed changes. This study investigated the level of selected EAATs in the HC after chronic treatment with mGluR5 antagonist MTEP, NS398, and their combination using Western blot. Concomitant MTEP treatment with NS398 or a single administration of the above causes changes in LTP and modulation of EAAT levels in mouse HC. As EAATs are cellular markers of oxidative stress mechanisms, the E. coli lipopolysaccharide (LPS) challenge was performed. The modified Barnes maze test (MBM) revealed alterations in the mouse spatial learning abilities. This study reports an interaction between the mGluR5 and COX-2 in the HC, with EAAT1 and EAAT3 involvement.
Collapse
Affiliation(s)
- Katarzyna Stachowicz
- Department of Neurobiology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland.
| | - Bartosz Bobula
- Department of Physiology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland
| | - Magdalena Kusek
- Department of Physiology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland
| | - Tomasz Lenda
- Department of Neuro- and Psychopharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland
| | - Krzysztof Tokarski
- Department of Physiology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland
| |
Collapse
|
26
|
Angarita GA, Hadizadeh H, Cerdena I, Potenza MN. Can pharmacotherapy improve treatment outcomes in people with co-occurring major depressive and cocaine use disorders? Expert Opin Pharmacother 2021; 22:1669-1683. [PMID: 34042556 DOI: 10.1080/14656566.2021.1931684] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Introduction: Major depressive disorder (MDD) and cocaine use disorder (CUD) are prevalent and frequently co-occur. When co-occurring, the presence of one disorder typically negatively impacts the prognosis for the other. Given the clinical relevance, we sought to examine pharmacotherapies for co-occurring CUD and MDD. While multiple treatment options have been examined in the treatment of each condition individually, studies exploring pharmacological options for their comorbidity are fewer and not conclusive.Areas Covered: For this review, the authors searched the literature in PubMed using clinical query options for therapies and keywords relating to each condition. Then, they described potentially promising pharmacologic therapeutic options based on shared mechanisms between the two conditions and/or results from individual clinical trials conducted to date.Expert opinion: Medications like stimulants, dopamine (D3) receptors partial agonists or antagonists, antagonists of kappa opioid receptors, topiramate, and ketamine could be promising as there is significant overlap relating to reward deficiency models, antireward pathways, and altered glutamatergic systems. However, the available clinical literature on any one of these types of agents is mixed. Additionally, for some agents there is possible concern related to abuse potential (e.g. ketamine and stimulants).
Collapse
Affiliation(s)
- Gustavo A Angarita
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA.,Clinical Neuroscience Research Unit, Connecticut Mental Health Center, New Haven, CT, USA
| | - Hasti Hadizadeh
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA.,Clinical Neuroscience Research Unit, Connecticut Mental Health Center, New Haven, CT, USA
| | - Ignacio Cerdena
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA.,Connecticut Mental Health Center, New Haven, CT, USA
| | - Marc N Potenza
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA.,Connecticut Mental Health Center, New Haven, CT, USA.,Child Study Center, Yale University School of Medicine, New Haven, CT, USA.,Department of Neuroscience, Yale University, New Haven, CT, USA.,Connecticut Council on Problem Gambling, Wethersfield, CT, USA
| |
Collapse
|
27
|
Oliveira TPD, Gonçalves BDC, Oliveira BS, de Oliveira ACP, Reis HJ, Ferreira CN, Aguiar DC, de Miranda AS, Ribeiro FM, Vieira EML, Palotás A, Vieira LB. Negative Modulation of the Metabotropic Glutamate Receptor Type 5 as a Potential Therapeutic Strategy in Obesity and Binge-Like Eating Behavior. Front Neurosci 2021; 15:631311. [PMID: 33642987 PMCID: PMC7902877 DOI: 10.3389/fnins.2021.631311] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 01/06/2021] [Indexed: 01/22/2023] Open
Abstract
Obesity is a multifactorial disease, which in turn contributes to the onset of comorbidities, such as diabetes and atherosclerosis. Moreover, there are only few options available for treating obesity, and most current pharmacotherapy causes severe adverse effects, while offering minimal weight loss. Literature shows that metabotropic glutamate receptor 5 (mGluR5) modulates central reward pathways. Herein, we evaluated the effect of VU0409106, a negative allosteric modulator (NAM) of mGluR5 in regulating feeding and obesity parameters. Diet-induced obese C57BL/6 mice were treated for 14 days with VU0409106, and food intake, body weight, inflammatory/hormonal levels, and behavioral tests were performed. Our data suggest reduction of feeding, body weight, and adipose tissue inflammation in mice treated with high-fat diet (HFD) after chronic treatment with VU0409106. Furthermore, a negative modulation of mGluR5 also reduces binge-like eating, the most common type of eating disorder. Altogether, our results pointed out mGluR5 as a potential target for treating obesity, as well as related disorders.
Collapse
Affiliation(s)
- Tadeu P. D. Oliveira
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Bruno D. C. Gonçalves
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Bruna S. Oliveira
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Antonio Carlos P. de Oliveira
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Helton J. Reis
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Claudia N. Ferreira
- Colégio Técnico, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Daniele C. Aguiar
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Aline S. de Miranda
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Fabiola M. Ribeiro
- Departamento de Bioquimica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Erica M. L. Vieira
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - András Palotás
- Asklepios-Med (Private Medical Practice and Research Center), Szeged, Hungary
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Luciene B. Vieira
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
28
|
Crespo M, León-Navarro DA, Martín M. Glutamatergic System is Affected in Brain from an Hyperthermia-Induced Seizures Rat Model. Cell Mol Neurobiol 2021; 42:1501-1512. [PMID: 33492599 DOI: 10.1007/s10571-021-01041-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 01/11/2021] [Indexed: 12/22/2022]
Abstract
One of the most frequent neurological disorders in children is febrile seizures (FS), a risk for epilepsy in adults. Glutamate is the main excitatory neurotransmitter in CNS acting through ionotropic and metabotropic receptors. Excess of glutamate in the extracellular space elicits excitotoxicity and has been associated with neurological disorders, such as epilepsy. The removal of extracellular glutamate by excitatory amino acid transporters (EATT) plays an important neuroprotective role. GLT-1 is the main EAAT present in the cortex brain. On the other hand, an increase in metabotropic glutamate receptors 5 (mGlu5R) levels or their overstimulation have been related to the appearance of seizure events in different animal models and in temporal lobe epilepsy in humans. In this work, the status of several components of the glutamatergic system has been analysed in the cortex brain from an FS rat model at short (48 h) and long (20 days) term after hyperthermia-induced seizures. At the short term, we detected increased GLT-1 levels, reduced glutamate concentration, and unchanged mGlu5R levels, without neuronal loss. However, at the long term, an increase in mGlu5R levels together with a decrease in both GLT-1 and glutamate levels were observed. These changes were associated with the appearance of an anxious phenotype. These results suggest a neuroprotective role of the glutamatergic components mGlu5R and GLT-1 at the short term. However, this neuroprotective effect seems to be lost at the long term, leading to an anxious phenotype and suggesting an increased vulnerability and propensity to epileptic events in adults.
Collapse
Affiliation(s)
- M Crespo
- Department of Inorganic, Organic and Biochemistry, Faculty of Chemical and Technological Sciences, School of Medicine of Ciudad Real, Universidad de Castilla-La Mancha, Regional Centre of Biomedical Research (CRIB), Avenida Camilo José Cela, 10, 13071, Ciudad Real, Spain
| | - D A León-Navarro
- Department of Inorganic, Organic and Biochemistry, Faculty of Chemical and Technological Sciences, School of Medicine of Ciudad Real, Universidad de Castilla-La Mancha, Regional Centre of Biomedical Research (CRIB), Avenida Camilo José Cela, 10, 13071, Ciudad Real, Spain.
| | - M Martín
- Department of Inorganic, Organic and Biochemistry, Faculty of Chemical and Technological Sciences, School of Medicine of Ciudad Real, Universidad de Castilla-La Mancha, Regional Centre of Biomedical Research (CRIB), Avenida Camilo José Cela, 10, 13071, Ciudad Real, Spain
| |
Collapse
|
29
|
Study of influence of the glutamatergic concentration of [ 18F]FPEB binding to metabotropic glutamate receptor subtype 5 with N-acetylcysteine challenge in rats and SRM/PET study in human healthy volunteers. Transl Psychiatry 2021; 11:66. [PMID: 33473111 PMCID: PMC7817831 DOI: 10.1038/s41398-020-01152-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 12/01/2020] [Accepted: 12/04/2020] [Indexed: 11/08/2022] Open
Abstract
Altered glutamate signaling is thought to be involved in a myriad of psychiatric disorders. Positron emission tomography (PET) imaging with [18F]FPEB allows assessing dynamic changes in metabotropic glutamate receptor 5 (mGluR5) availability underlying neuropathological conditions. The influence of endogenous glutamatergic levels into receptor binding has not been well established yet. The purpose of this study was to explore the [18F]FPEB binding regarding to physiological fluctuations or acute changes of glutamate synaptic concentrations by a translational approach; a PET/MRS imaging study in 12 healthy human volunteers combined to a PET imaging after an N-acetylcysteine (NAc) pharmacological challenge in rodents. No significant differences were observed with small-animal PET in the test and retest conditions on the one hand and the NAc condition on the other hand for any regions. To test for an interaction of mGuR5 density and glutamatergic concentrations in healthy subjects, we correlated the [18F]FPEB BPND with Glu/Cr, Gln/Cr, Glx/Cr ratios in the anterior cingulate cortex VOI; respectively, no significance correlation has been revealed (Glu/Cr: r = 0.51, p = 0.09; Gln/Cr: r = -0.46, p = 0.13; Glx/Cr: r = -0.035, p = 0.92).These data suggest that the in vivo binding of [18F]FPEB to an allosteric site of the mGluR5 is not modulated by endogenous glutamate in vivo. Thus, [18F]FPEB appears unable to measure acute fluctuations in endogenous levels of glutamate.
Collapse
|
30
|
Gubert C, Kong G, Uzungil V, Zeleznikow-Johnston AM, Burrows EL, Renoir T, Hannan AJ. Microbiome Profiling Reveals Gut Dysbiosis in the Metabotropic Glutamate Receptor 5 Knockout Mouse Model of Schizophrenia. Front Cell Dev Biol 2020; 8:582320. [PMID: 33195226 PMCID: PMC7658610 DOI: 10.3389/fcell.2020.582320] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Accepted: 10/08/2020] [Indexed: 01/03/2023] Open
Abstract
Schizophrenia (SZ) is a psychiatric disorder that constitutes one of the top 10 global causes of disability. More recently, a potential pathogenic role for the gut microbial community (microbiota) has been highlighted, with numerous studies describing dysregulated microbial profiles in SZ patients when compared to healthy controls. However, no animal model of SZ has previously recapitulated the gut dysbiosis observed clinically. Since the metabotropic glutamate receptor 5 (mGlu5) knockout mice provide a preclinical model of SZ with strong face and predictive validity, in the present study we performed gut microbiome profiling of mGlu5 knockout (KO) and wild-type (WT) mice by 16S rRNA sequencing of bacterial genomic DNA from fecal samples, analyzing bacterial diversity and taxonomic composition, as well as gastrointestinal parameters as indicators of gut function. We found a significant genotype difference in microbial beta diversity. Analysis of composition of microbiomes (ANCOM) models were performed to evaluate microbiota compositions, which identified a decreased relative abundance of the Erysipelotrichaceae family and Allobaculum genus in this mouse model of SZ. We also identified a signature of bacteria discriminating between the genotypes (KO and WT), consisting of the Erysipelotrichales, Bacteroidales, and Clostridiales orders and macroscopic gut differences. We thus uncovered global differential community composition in the gut microbiota profile between mGlu5 KO and WT mice, outlining the first evidence for gut dysbiosis in a genetic animal model of SZ. Our findings suggest that this widely used preclinical model of SZ also has substantial utility for investigations of gut dysbiosis and associated signaling via the microbiota-gut-brain axis, as potential modulators of SZ pathogenesis. Our discovery opens up new avenues to explore gut dysbiosis and its proposed links to brain dysfunction in SZ, as well as novel therapeutic approaches to this devastating disorder.
Collapse
Affiliation(s)
- Carolina Gubert
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Geraldine Kong
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Volkan Uzungil
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | | | - Emma L. Burrows
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Thibault Renoir
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Anthony J. Hannan
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
- Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
31
|
Akkus F, Terbeck S, Haggarty CJ, Treyer V, Dietrich JJ, Hornschuh S, Hasler G. The role of the metabotropic glutamate receptor 5 in nicotine addiction. CNS Spectr 2020; 26:1-6. [PMID: 32713396 DOI: 10.1017/s1092852920001704] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
This review summarizes the evidence for the potential involvement of metabotropic glutamate receptor 5 (mGluR5) in the development of nicotine addiction. Nicotine is consumed worldwide and is highly addictive. Previous research has extensively investigated the role of dopamine in association with reward learning and addiction, which has provided strong evidence for the involvement of dopaminergic neuronal circuitry in nicotine addiction. More recently, researchers focused on glutamatergic transmission after nicotine abuse, and its involvement in the reinforcing and rewarding effects of nicotine addiction. A number of robust preclinical and clinical studies have shown mGluR5 signaling as a facilitating mechanism of nicotine addiction and nicotine withdrawal. Specifically, clinical studies have illustrated lower cortical mGluR5 density in smokers compared to nonsmokers in the human brain. In addition, mGluR5 might selectively regulate craving and withdrawal. This suggests that mGluR5 could be a key receptor in the development of nicotine addiction and therefore clinical trials to examine the therapeutic potential of mGluR5 agents could help to contribute to reduce nicotine addiction in society.
Collapse
Affiliation(s)
- Funda Akkus
- Department of Psychiatry, University of Fribourg, Fribourg, Switzerland
- Psychiatrie St. Gallen Nord, Wil, Switzerland
| | - Sylvia Terbeck
- School of Psychology, Liverpool John Moores University, LiverpoolUnited Kingdom
| | - Connor J Haggarty
- School of Psychology, Liverpool John Moores University, LiverpoolUnited Kingdom
| | - Valerie Treyer
- Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland
| | - Janan J Dietrich
- Perinatal HIV Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Stefanie Hornschuh
- Perinatal HIV Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Gregor Hasler
- Department of Psychiatry, University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
32
|
Nag S, Varnäs K, Arakawa R, Jahan M, Schou M, Farde L, Halldin C. Synthesis, Biodistribution, and Radiation Dosimetry of a Novel mGluR5 Radioligand: 18F-AZD9272. ACS Chem Neurosci 2020; 11:1048-1057. [PMID: 32167745 PMCID: PMC7309225 DOI: 10.1021/acschemneuro.9b00680] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
![]()
The metabotropic
glutamate receptor subtype mGluR5 has been proposed
as a potential drug target for CNS disorders such as anxiety, depression,
Parkinson’s disease, and epilepsy. The AstraZeneca compound
AZD9272 has previously been labeled with carbon-11 and used as a PET
radioligand for mGluR5 receptor binding. The molecular structure of
AZD9272 allows one to label the molecule with fluorine-18 without
altering the structure. The aim of this study was to develop a fluorine-18
analogue of AZD9272 and to examine its binding distribution in the
nonhuman primate brain in vivo as well as to obtain
whole body radiation dosimetry. 18F-AZD9272 was successfully
synthesized from a nitro precursor. The radioligand was stable, with
a radiochemical purity of >99% at 2 h after formulation in a sterile
phosphate buffered solution (pH = 7.4). After injection of 18F-AZD9272 in two cynomolgus monkeys, the maximum whole brain radioactivity
concentration was 4.9–6.7% of the injected dose (n = 2) and PET images showed a pattern of regional radioactivity consistent
with that previously obtained for 11C-AZD9272. The percentage
of parent radioligand in plasma was 59 and 64% (n = 2) at 120 min after injection of 18F-AZD9272, consistent
with high metabolic stability. Two whole body PET scans were performed
in nonhuman primates for a total of 231 min after injection of 18F-AZD9272. Highest uptakes were seen in liver and small intestine,
followed by brain and kidney. The estimated effective dose was around
0.017 mSv/MBq. 18F-AZD9272 shows suitable properties as
a PET radioligand for in vivo imaging of binding
in the primate brain. 18F-labeled AZD9272 offers advantages
over 11C-AZD9272 in terms of higher image resolution, combined
with a longer half-life. Moreover, based on the distribution and the
estimated radiation burden, imaging of 18F-AZD9272 could
be used as an improved tool for quantitative assessment and characterization
of AZD9272 binding sites in the human brain by using PET.
Collapse
Affiliation(s)
- Sangram Nag
- Department of Clinical Neuroscience, Center for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm 17176, Sweden
| | - Katarina Varnäs
- Department of Clinical Neuroscience, Center for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm 17176, Sweden
| | - Ryosuke Arakawa
- Department of Clinical Neuroscience, Center for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm 17176, Sweden
| | - Mahabuba Jahan
- Department of Medicinal Chemistry, Uppsala University, Uppsala 751 05, Sweden
| | - Magnus Schou
- Department of Clinical Neuroscience, Center for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm 17176, Sweden
- PET Science Centre, Precision Medicine, Oncology R&D, AstraZeneca, Stockholm 17176, Sweden
| | - Lars Farde
- Department of Clinical Neuroscience, Center for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm 17176, Sweden
| | - Christer Halldin
- Department of Clinical Neuroscience, Center for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm 17176, Sweden
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 639798
| |
Collapse
|
33
|
Johnson KA, Lovinger DM. Allosteric modulation of metabotropic glutamate receptors in alcohol use disorder: Insights from preclinical investigations. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2020; 88:193-232. [PMID: 32416868 DOI: 10.1016/bs.apha.2020.02.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Metabotropic glutamate (mGlu) receptors are family C G protein-coupled receptors (GPCRs) that modulate neuronal excitability and synaptic transmission throughout the nervous system. Owing to recent advances in development of subtype-selective allosteric modulators of mGlu receptors, individual members of the mGlu receptor family have been proposed as targets for treating a variety of neurological and psychiatric disorders, including substance use disorders. In this chapter, we highlight preclinical evidence that allosteric modulators of mGlu receptors could be useful for reducing alcohol consumption and preventing relapse in alcohol use disorder (AUD). We begin with an overview of the preclinical models that are used to study mGlu receptor involvement in alcohol-related behaviors. Alcohol exposure causes adaptations in both expression and function of various mGlu receptor subtypes, and pharmacotherapies aimed at reversing these adaptations have the potential to reduce alcohol consumption and seeking. Positive allosteric modulators (PAMs) of mGlu2 and negative allosteric modulators of mGlu5 show particular promise for reducing alcohol intake and/or preventing relapse. Finally, this chapter discusses important considerations for translating preclinical findings toward the development of clinically useful drugs, including the potential for PAMs to avoid tolerance issues that are frequently observed with repeated administration of GPCR agonists.
Collapse
Affiliation(s)
- Kari A Johnson
- Department of Pharmacology, Uniformed Services University of the Health Sciences, Bethesda, MD, United States.
| | - David M Lovinger
- Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, US National Institutes of Health, Rockville, MD, United States
| |
Collapse
|
34
|
Varlow C, Murrell E, Holland JP, Kassenbrock A, Shannon W, Liang SH, Vasdev N, Stephenson NA. Revisiting the Radiosynthesis of [ 18F]FPEB and Preliminary PET Imaging in a Mouse Model of Alzheimer's Disease. Molecules 2020; 25:molecules25040982. [PMID: 32098347 PMCID: PMC7070414 DOI: 10.3390/molecules25040982] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 02/19/2020] [Accepted: 02/20/2020] [Indexed: 11/24/2022] Open
Abstract
[18F]FPEB is a positron emission tomography (PET) radiopharmaceutical used for imaging the abundance and distribution of mGluR5 in the central nervous system (CNS). Efficient radiolabeling of the aromatic ring of [18F]FPEB has been an ongoing challenge. Herein, five metal-free precursors for the radiofluorination of [18F]FPEB were compared, namely, a chloro-, nitro-, sulfonium salt, and two spirocyclic iodonium ylide (SCIDY) precursors bearing a cyclopentyl (SPI5) and a new adamantyl (SPIAd) auxiliary. The chloro- and nitro-precursors resulted in a low radiochemical yield (<10% RCY), whereas both SCIDY precursors and the sulfonium salt precursor produced [18F]FPEB in the highest RCYs of 25% and 36%, respectively. Preliminary PET/CT imaging studies with [18F]FPEB were conducted in a transgenic model of Alzheimer’s Disease (AD) using B6C3-Tg(APPswe,PSEN1dE9)85Dbo/J (APP/PS1) mice, and data were compared with age-matched wild-type (WT) B6C3F1/J control mice. In APP/PS1 mice, whole brain distribution at 5 min post-injection showed a slightly higher uptake (SUV = 4.8 ± 0.4) than in age-matched controls (SUV = 4.0 ± 0.2). Further studies to explore mGluR5 as an early biomarker for AD are underway.
Collapse
Affiliation(s)
- Cassis Varlow
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada; (C.V.); (E.M.); (W.S.)
- Institute of Medical Science, University of Toronto, Toronto, ON M5S1A8, Canada
| | - Emily Murrell
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada; (C.V.); (E.M.); (W.S.)
| | - Jason P. Holland
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA 02114, USA; (J.P.H.); (A.K.); (S.H.L.)
- Department of Chemistry, University of Zurich, 8057 Zurich, Switzerland
| | - Alina Kassenbrock
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA 02114, USA; (J.P.H.); (A.K.); (S.H.L.)
| | - Whitney Shannon
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada; (C.V.); (E.M.); (W.S.)
- Department of Chemistry, University of Saskatchewan, Saskatoon, SK S7N OX2, Canada
| | - Steven H. Liang
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA 02114, USA; (J.P.H.); (A.K.); (S.H.L.)
| | - Neil Vasdev
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada; (C.V.); (E.M.); (W.S.)
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA 02114, USA; (J.P.H.); (A.K.); (S.H.L.)
- Department of Psychiatry, University of Toronto, Toronto, ON M5T-1R8, Canada
- Correspondence: (N.V.); (N.A.S.); Tel.: +416-535-8501 (ext. 30988) (N.V.); +1-876-927-1910 (N.A.S.)
| | - Nickeisha A. Stephenson
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada; (C.V.); (E.M.); (W.S.)
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA 02114, USA; (J.P.H.); (A.K.); (S.H.L.)
- Department of Chemistry, The University of West Indies at Mona, Kingston 7, Jamaica
- Correspondence: (N.V.); (N.A.S.); Tel.: +416-535-8501 (ext. 30988) (N.V.); +1-876-927-1910 (N.A.S.)
| |
Collapse
|
35
|
Zhao Y, Chen J, Liu Q, Li Y. Profiling the Structural Determinants of Aryl Benzamide Derivatives as Negative Allosteric Modulators of mGluR5 by In Silico Study. Molecules 2020; 25:molecules25020406. [PMID: 31963723 PMCID: PMC7024197 DOI: 10.3390/molecules25020406] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Revised: 01/09/2020] [Accepted: 01/16/2020] [Indexed: 11/16/2022] Open
Abstract
Glutamate plays a crucial role in the treatment of depression by interacting with the metabotropic glutamate receptor subtype 5 (mGluR5), whose negative allosteric modulators (NAMs) are thus promising antidepressants. At present, to explore the structural features of 106 newly synthesized aryl benzamide series molecules as mGluR5 NAMs, a set of ligand-based three-dimensional quantitative structure-activity relationship (3D-QSAR) analyses were firstly carried out applying comparative molecular field analysis (CoMFA) and comparative molecular similarity indices analysis (CoMSIA) methods. In addition, receptor-based analysis, namely molecular docking and molecular dynamics (MD) simulations, were performed to further elucidate the binding modes of mGluR5 NAMs. As a result, the optimal CoMSIA model obtained shows that cross-validated correlation coefficient Q2 = 0.70, non-cross-validated correlation coefficient R2ncv = 0.89, predicted correlation coefficient R2pre = 0.87. Moreover, we found that aryl benzamide series molecules bind as mGluR5 NAMs at Site 1, which consists of amino acids Pro655, Tyr659, Ile625, Ile651, Ile944, Ser658, Ser654, Ser969, Ser965, Ala970, Ala973, Trp945, Phe948, Pro903, Asn907, Val966, Leu904, and Met962. This site is the same as that of other types of NAMs; mGluR5 NAMs are stabilized in the "linear" and "arc" configurations mainly through the H-bonds interactions, π-π stacking interaction with Trp945, and hydrophobic contacts. We hope that the models and information obtained will help understand the interaction mechanism of NAMs and design and optimize NAMs as new types of antidepressants.
Collapse
Affiliation(s)
- Yujing Zhao
- Key Laboratory of Industrial Ecology and Environmental Engineering, Faculty of Chemical, Environmental and Biological Science and Technology, Dalian University of Technology, Dalian 116024, China; (Y.Z.); (J.C.)
| | - Jiabin Chen
- Key Laboratory of Industrial Ecology and Environmental Engineering, Faculty of Chemical, Environmental and Biological Science and Technology, Dalian University of Technology, Dalian 116024, China; (Y.Z.); (J.C.)
| | - Qilei Liu
- Institute of Chemical Process Systems Engineering, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, China;
| | - Yan Li
- Key Laboratory of Industrial Ecology and Environmental Engineering, Faculty of Chemical, Environmental and Biological Science and Technology, Dalian University of Technology, Dalian 116024, China; (Y.Z.); (J.C.)
- Correspondence: ; Tel.: +86-15640888728
| |
Collapse
|
36
|
Translating preclinical findings in clinically relevant new antipsychotic targets: focus on the glutamatergic postsynaptic density. Implications for treatment resistant schizophrenia. Neurosci Biobehav Rev 2019; 107:795-827. [DOI: 10.1016/j.neubiorev.2019.08.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Revised: 07/20/2019] [Accepted: 08/22/2019] [Indexed: 02/07/2023]
|
37
|
Laukkanen V, Kärkkäinen O, Kautiainen H, Tiihonen J, Storvik M. Increased [³H]quisqualic acid binding density in the dorsal striatum and anterior insula of alcoholics: A post-mortem whole-hemisphere autoradiography study. Psychiatry Res Neuroimaging 2019; 287:63-69. [PMID: 30991250 DOI: 10.1016/j.pscychresns.2019.04.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 04/05/2019] [Accepted: 04/07/2019] [Indexed: 01/28/2023]
Abstract
The function of group I metabotropic glutamate receptors mGluR1 and mGluR5 is involved in the hyperglutamatergic state caused by chronic alcohol. Preclinical studies suggest that group I mGluR modulation could serve as a novel treatment of alcoholism. Considering the wide role of glutamatergic neurochemistry in addiction, group I mGluR binding was studied in brain areas involved in decision-making, learning and memory. Post-mortem whole hemisphere autoradiography was used to study the binding density of [³H]quisqualic acid, a potent group I mGluR agonist, in 9 Cloninger type 1 alcoholics, 8 Cloninger type 2 alcoholics and 10 controls. Binding was studied in the dorsal striatum, hippocampus and cortex. Alcoholics displayed a trend towards increased [³H]quisqualic acid binding in all brain areas. The most robust findings were in the putamen (p = 0.006) and anterior insula (p = 0.005), where both alcoholic subtypes displayed increased binding compared to the controls. These findings suggest altered group I mGluR function in alcoholic subjects in the dorsal striatum, which is involved in habitual learning, and in the anterior insula, which has a pivotal role in the perception of bodily sensations. Increased [³H]quisqualic acid binding might suggest a beneficial impact of mGluR1/5 modulators in the treatment of alcoholism.
Collapse
Affiliation(s)
- Virpi Laukkanen
- Department of Forensic Psychiatry, University of Eastern Finland, Niuvanniemi Hospital, Niuvankuja 65, FI-70240 Kuopio, Finland; Department of Psychiatry, Kuopio University Hospital, P.O. Box 100, FI-70029 Kuopio, Finland.
| | - Olli Kärkkäinen
- Department of Pharmacology and Toxicology, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Hannu Kautiainen
- Unit of Primary Health Care, Helsinki University Central Hospital, P.O. Box 705, FI-00029 HUS, Helsinki, Finland; Department of General Practice, Helsinki University, P.O. Box 20, FI-00014 Helsinki, Finland
| | - Jari Tiihonen
- Department of Forensic Psychiatry, University of Eastern Finland, Niuvanniemi Hospital, Niuvankuja 65, FI-70240 Kuopio, Finland; Department of Clinical Neuroscience, Karolinska Institutet, Karolinska Hospital, 17176 Stockholm, Sweden
| | - Markus Storvik
- Department of Psychiatry, Kuopio University Hospital, P.O. Box 100, FI-70029 Kuopio, Finland
| |
Collapse
|
38
|
Yohn SE, Galbraith J, Calipari ES, Conn PJ. Shared Behavioral and Neurocircuitry Disruptions in Drug Addiction, Obesity, and Binge Eating Disorder: Focus on Group I mGluRs in the Mesolimbic Dopamine Pathway. ACS Chem Neurosci 2019; 10:2125-2143. [PMID: 30933466 PMCID: PMC7898461 DOI: 10.1021/acschemneuro.8b00601] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Accumulated data from clinical and preclinical studies suggest that, in drug addiction and states of overeating, such as obesity and binge eating disorder (BED), there is an imbalance in circuits that are critical for motivation, reward saliency, executive function, and self-control. Central to these pathologies and the extensive topic of this Review are the aberrations in dopamine (DA) and glutamate (Glu) within the mesolimbic pathway. Group I metabotropic glutamate receptors (mGlus) are highly expressed in the mesolimbic pathway and are poised in key positions to modulate disruptions in synaptic plasticity and neurotransmitter release observed in drug addiction, obesity, and BED. The use of allosteric modulators of group I mGlus has been studied in drug addiction, as they offer several advantages over traditional orthosteric agents. However, they have yet to be studied in obesity or BED. With the substantial overlap between the neurocircuitry involved in drug addiction and eating disorders, group I mGlus may also provide novel targets for obesity and BED.
Collapse
Affiliation(s)
- Samantha E. Yohn
- Department of Pharmacology, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, 37232, United States
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, 37232, United States
| | - Jordan Galbraith
- Department of Pharmacology, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, 37232, United States
| | - Erin S. Calipari
- Department of Pharmacology, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, 37232, United States
| | - P. Jeffrey Conn
- Department of Pharmacology, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, 37232, United States
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, 37232, United States
| |
Collapse
|
39
|
Moccia L, Tofani A, Mazza M, Covino M, Martinotti G, Schifano F, Janiri L, Di Nicola M. Dorsolateral Prefrontal Cortex Impairment in Methoxetamine-Induced Psychosis: An 18F-FDG PET/CT Case Study. J Psychoactive Drugs 2019; 51:254-259. [PMID: 30741111 DOI: 10.1080/02791072.2019.1578444] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Novel psychoactive substances (NPSs) have currently become a major public health concern because of relatively easy accessibility to these compounds and difficulty in identifying them with routine laboratory techniques. Here, we report the 18F-fluorodeoxyglucose positron emission tomography/computerized tomography (18F-FDG PET/CT) case study of a 23-year-old man who developed a substance-induced psychotic disorder after having intravenously injected himself with an unspecified amount of methoxetamine (MXE), a ketamine derivative hallucinogen. From a clinical perspective, a blunted affective responsiveness with diminished social drive and sense of purpose, along with a profound detachment from the environment, was observed. Psychometric and neuropsychological assessments highlighted severe dissociative symptoms and lack of motivation, along with a mild impairment of verbal fluency, working memory, and attention. Patient's 18F-FDG PET/CT scans displayed a significant bilateral deficit of tracer uptake within the dorsolateral prefrontal cortex (DLPFC). DLPFC activity is critical to goal-oriented cognitive functions, including working memory and sustained attention. DLPFC is also involved in both the temporal integration across multiple sensory modes and in the volitional control of actions, leading to the possibility to construct logically coherent temporal configurations of thought, speech, and behavior. This report highlights that a single acute MXE intoxication may produce severe brain impairment.
Collapse
Affiliation(s)
- Lorenzo Moccia
- a Institute of Psychiatry and Psychology, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Università Cattolica del Sacro Cuore , Rome , Italy
| | - Anna Tofani
- b Unit of Nuclear Medicine, Department of Surgical and Medical Sciences and Translational Medicine, Sant'Andrea Hospital, "Sapienza" University of Rome , Rome , Italy
| | - Marianna Mazza
- a Institute of Psychiatry and Psychology, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Università Cattolica del Sacro Cuore , Rome , Italy
| | - Marcello Covino
- c Emergency Medicine, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Università Cattolica del Sacro Cuore , Rome , Italy
| | - Giovanni Martinotti
- d Department of Neuroscience and Imaging, Institute of Psychiatry, "G. D'Annunzio" University of Chieti-Pescara , Chieti , Italy
| | - Fabrizio Schifano
- e Psychopharmacology, Drug Misuse and Novel Psychoactive Substances Research Unit, University of Hertfordshire , Hatfield , UK
| | - Luigi Janiri
- a Institute of Psychiatry and Psychology, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Università Cattolica del Sacro Cuore , Rome , Italy
| | - Marco Di Nicola
- a Institute of Psychiatry and Psychology, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Università Cattolica del Sacro Cuore , Rome , Italy
| |
Collapse
|
40
|
Lum JS, Millard SJ, Frank E, Matosin N, Huang XF, Ooi L, Newell KA. Chronic Adolescent CDPPB Treatment Alters Short-Term, but not Long-Term, Glutamatergic Receptor Expression. Neurochem Res 2018; 43:1683-1691. [PMID: 29936568 DOI: 10.1007/s11064-018-2584-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 05/30/2018] [Accepted: 06/20/2018] [Indexed: 12/21/2022]
Abstract
Dysfunction of the glutamatergic system is believed to underlie many neurodevelopmental disorders including autism, Rett syndrome and schizophrenia. Metabotropic glutamate receptor (mGluR5) positive allosteric modulators (PAM) potentiate glutamatergic signaling, particularly indirectly via the NMDA receptor. Preclinical studies report mGluR5 PAMs can improve schizophrenia-relevant behaviours. Furthermore, adolescent administration has shown to prevent cognitive induced deficits in adult rodents. However, there is limited understanding of the short- and long-term neurochemical effects of mGluR5 PAMs, which may underlie their therapeutic effects. We examined the effect of 7-day adolescent (PN28-34) treatment with the mGluR5 PAM, CDDPB (30 mg/kg), on glutamatergic receptor expression at adolescence (PN35) and adulthood (PN96). Immunoblot analysis revealed that 7-day adolescent CDPPB treatment increased protein expression of glutamatergic receptors including the NMDA receptor subunits, NR1 and NR2A and the AMPA subunits (GluA1 and GluA2) in the adolescent hippocampus, changes that did not extend to adulthood. In contrast, there were no changes in the adolescent frontal cortex, however elevated mGluR5 protein expression was observed at adulthood following adolescent CDPPB treatment. The present study indicates adolescent CDPPB treatment may cause brain region dependent effects on the glutamatergic system, which do not persist into adulthood. These findings may have implications for the preclinical development of mGluR5 PAMs for the treatment of neurodevelopmental disorders.
Collapse
Affiliation(s)
- Jeremy S Lum
- School of Medicine, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW, 2522, Australia.
- Illawarra Health and Medical Research Institute, Wollongong, NSW, 2522, Australia.
- Schizophrenia Research Institute, 405 Liverpool Street, Darlinghurst, NSW, 2010, Australia.
| | - Samuel J Millard
- School of Medicine, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW, 2522, Australia
- Illawarra Health and Medical Research Institute, Wollongong, NSW, 2522, Australia
| | - Elisabeth Frank
- Illawarra Health and Medical Research Institute, Wollongong, NSW, 2522, Australia
- Schizophrenia Research Institute, 405 Liverpool Street, Darlinghurst, NSW, 2010, Australia
| | - Natalie Matosin
- School of Medicine, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW, 2522, Australia
- Illawarra Health and Medical Research Institute, Wollongong, NSW, 2522, Australia
- Schizophrenia Research Institute, 405 Liverpool Street, Darlinghurst, NSW, 2010, Australia
| | - Xu-Feng Huang
- School of Medicine, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW, 2522, Australia
- Illawarra Health and Medical Research Institute, Wollongong, NSW, 2522, Australia
| | - Lezanne Ooi
- Illawarra Health and Medical Research Institute, Wollongong, NSW, 2522, Australia
- School of Biological Sciences, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW, 2522, Australia
| | - Kelly A Newell
- School of Medicine, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW, 2522, Australia
- Illawarra Health and Medical Research Institute, Wollongong, NSW, 2522, Australia
| |
Collapse
|
41
|
Barnes SA, Sheffler DJ, Semenova S, Cosford NDP, Bespalov A. Metabotropic Glutamate Receptor 5 as a Target for the Treatment of Depression and Smoking: Robust Preclinical Data but Inconclusive Clinical Efficacy. Biol Psychiatry 2018; 83:955-962. [PMID: 29628194 PMCID: PMC5953810 DOI: 10.1016/j.biopsych.2018.03.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 03/02/2018] [Accepted: 03/05/2018] [Indexed: 12/11/2022]
Abstract
The ability of novel pharmacological compounds to improve outcomes in preclinical models is often not translated into clinical efficacy. Psychiatric disorders do not have biological boundaries, and identifying mechanisms to improve the translational bottleneck between preclinical and clinical research domains is an important and challenging task. Glutamate transmission is disrupted in several neuropsychiatric disorders. Metabotropic glutamate (mGlu) receptors represent a diverse class of receptors that contribute to excitatory neurotransmission. Given the wide, yet region-specific manner of expression, developing pharmacological compounds to modulate mGlu receptor activity provides an opportunity to subtly and selectively modulate excitatory neurotransmission. This review focuses on the potential involvement of mGlu5 receptor disruption in major depressive disorder and substance and/or alcohol use disorders. We provide an overview of the justification of targeting mGlu5 receptors in the treatment of these disorders, summarize the preclinical evidence for negatively modulating mGlu5 receptors as a therapeutic target for major depressive disorders and nicotine dependence, and highlight the outcomes of recent clinical trials. While the evidence of mGlu5 receptor negative allosteric modulation has been promising in preclinical investigations, these beneficial effects have not translated into clinical efficacy. In this review, we identify key challenges that may contribute to poor clinical translation and provide suggested approaches moving forward to potentially improve the translation from preclinical to clinical domains. Such approaches may increase the success of clinical trials and may reduce the translational bottleneck that exists in drug discovery for psychiatric disorders.
Collapse
Affiliation(s)
- Samuel A. Barnes
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive MC 0603, La Jolla, CA 92093, USA
| | - Douglas J. Sheffler
- NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Svetlana Semenova
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive MC 0603, La Jolla, CA 92093, USA,PAREXEL International, 1560 E Chevy Chase Dr, suite 140, Glendale, CA 91206, USA
| | - Nicholas D. P. Cosford
- NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Anton Bespalov
- EXCIVA, Heidelberg, Germany; Valdman Institute of Pharmacology, Pavlov Medical University, St. Petersburg, Russia.
| |
Collapse
|
42
|
Vijaya Prabhu S, Singh SK. Atom-based 3D-QSAR, induced fit docking, and molecular dynamics simulations study of thieno[2,3-b]pyridines negative allosteric modulators of mGluR5. J Recept Signal Transduct Res 2018; 38:225-239. [PMID: 29806525 DOI: 10.1080/10799893.2018.1476542] [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: 01/01/2023]
Abstract
Atom-based three dimensional-quantitative structure-activity relationship (3D-QSAR) model was developed on the basis of 5-point pharmacophore hypothesis (AARRR) with two hydrogen bond acceptors (A) and three aromatic rings for the derivatives of thieno[2,3-b]pyridine, which modulates the activity to inhibit the mGluR5 receptor. Generation of a highly predictive 3D-QSAR model was performed using the alignment of predicted pharmacophore hypothesis for the training set (R2 = 0.84, SD = 0.26, F = 45.8, N = 29) and test set (Q2 = 0.74, RMSE = 0.235, Pearson-R = 0.94, N = 9). The best pharmacophore hypothesis AARRR was selected, and developed three dimensional-quantitative structure activity relationship (3D-QSAR) model also supported the outcome of this study by means of favorable and unfavorable electron withdrawing group and hydrophobic regions of most active compound 42d and least active compound 18b. Following, induced fit docking and binding free energy calculations reveals the reliable binding orientation of the compounds. Finally, molecular dynamics simulations for 100 ns were performed to depict the protein-ligand stability. We anticipate that the resulted outcome could be supportive to discover potent negative allosteric modulators for metabotropic glutamate receptor 5 (mGluR5).
Collapse
Affiliation(s)
- Sitrarasu Vijaya Prabhu
- a Department of Bioinformatics, Computer Aided Drug Design and Molecular Modeling Lab , Alagappa University , Karaikudi , India
| | - Sanjeev Kumar Singh
- a Department of Bioinformatics, Computer Aided Drug Design and Molecular Modeling Lab , Alagappa University , Karaikudi , India
| |
Collapse
|
43
|
Faye C, McGowan JC, Denny CA, David DJ. Neurobiological Mechanisms of Stress Resilience and Implications for the Aged Population. Curr Neuropharmacol 2018; 16:234-270. [PMID: 28820053 PMCID: PMC5843978 DOI: 10.2174/1570159x15666170818095105] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 06/25/2017] [Accepted: 07/27/2017] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Stress is a common reaction to an environmental adversity, but a dysregulation of the stress response can lead to psychiatric illnesses such as major depressive disorder (MDD), post-traumatic stress disorder (PTSD), and anxiety disorders. Yet, not all individuals exposed to stress will develop psychiatric disorders; those with enhanced stress resilience mechanisms have the ability to adapt successfully to stress without developing persistent psychopathology. Notably, the potential to enhance stress resilience in at-risk populations may prevent the onset of stress-induced psychiatric disorders. This novel idea has prompted a number of studies probing the mechanisms of stress resilience and how it can be manipulated. METHODS Here, we review the neurobiological factors underlying stress resilience, with particular focus on the serotoninergic (5-HT), glutamatergic, and γ-Aminobutyric acid (GABA) systems, as well as the hypothalamic-pituitary axis (HPA) in rodents and in humans. Finally, we discuss stress resiliency in the context of aging, as the likelihood of mood disorders increases in older adults. RESULTS Interestingly, increased resiliency has been shown to slow aging and improved overall health and quality of life. Research in the neurobiology of stress resilience, particularly throughout the aging process, is a nascent, yet, burgeoning field. CONCLUSION Overall, we consider the possible methods that may be used to induce resilient phenotypes, prophylactically in at-risk populations, such as in military personnel or in older MDD patients. Research in the mechanisms of stress resilience may not only elucidate novel targets for antidepressant treatments, but also provide novel insight about how to prevent these debilitating disorders from developing.
Collapse
Affiliation(s)
- Charlène Faye
- CESP/UMR-S 1178, Univ. Paris-Sud, Fac Pharmacie, Inserm, Université Paris-Saclay, 92296 Chatenay-Malabry, France
| | - Josephine C. McGowan
- Doctoral Program in Neurobiology and Behavior, Columbia University, New York, NY, USA
| | - Christine A. Denny
- Department of Psychiatry, Columbia University, New York, NY, USA
- Division of Integrative Neuroscience, New York State Psychiatric Institute/Research Foundation for Mental Hygiene, Inc., New York, NY, USA
| | - Denis J. David
- CESP/UMR-S 1178, Univ. Paris-Sud, Fac Pharmacie, Inserm, Université Paris-Saclay, 92296 Chatenay-Malabry, France
| |
Collapse
|
44
|
Park JY, Son J, Yun M, Ametamey SM, Chun JH. Automated cGMP-compliant radiosynthesis of [ 18 F]-(E)-PSS232 for brain PET imaging of metabotropic glutamate receptor subtype 5. J Labelled Comp Radiopharm 2017; 61:30-37. [PMID: 28948638 DOI: 10.1002/jlcr.3566] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Revised: 09/12/2017] [Accepted: 09/15/2017] [Indexed: 11/10/2022]
Abstract
(E)-3-(Pyridin-2-yl ethynyl)cyclohex-2-enone O-(3-(2-[18 F]-fluoroethoxy)propyl) oxime ([18 F]-(E)-PSS232, [18 F]2a) is a recently developed radiotracer that can be used to visualize metabotropic glutamate receptor subtype 5 (mGlu5 ) in vivo. The mGlu5 has become an attractive therapeutic and diagnostic target owing to its role in many neuropsychiatric disorders. Several carbon-11-labeled and fluorine-18-labeled radiotracers have been developed to measure mGlu5 receptor occupancy in the human brain. The radiotracer [18 F]2a, which is used as an analogue for [11 C]ABP688 ([11 C]1) and has a longer physical half-life, is a selective radiotracer that exhibits high binding affinity for mGlu5 . Herein, we report the fully automated radiosynthesis of [18 F]2a using a commercial GE TRACERlab™ FX-FN synthesizer for routine production and distribution to nearby satellite clinics. Nucleophilic substitution of the corresponding mesylate precursor with cyclotron-produced [18 F]fluoride ion at 100°C in dimethyl sulfoxide (DMSO), followed by high-performance liquid chromatography (HPLC) purification and formulation, readily provided [18 F]2a with a radiochemical yield of 40 ± 2% (decay corrected, n = 5) at the end of synthesis. Radiochemical purity for the [18 F]-(E)-conformer was greater than 95%. Molar activity was determined to be 63.6 ± 9.6 GBq/μmol (n = 5), and the overall synthesis time was 70 minutes.
Collapse
Affiliation(s)
- Jun Young Park
- Department of Nuclear Medicine, Severance Hospital, Yonsei University Health System, Seoul, Republic of Korea
| | - Jeongmin Son
- Department of Nuclear Medicine, Severance Hospital, Yonsei University Health System, Seoul, Republic of Korea
| | - Mijin Yun
- Department of Nuclear Medicine, Severance Hospital, Yonsei University Health System, Seoul, Republic of Korea.,Department of Nuclear Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Simon M Ametamey
- Department of Applied Biosciences of ETH Zurich, Center for Radiopharmaceutical Sciences of ETH, PSI, and USZ, Zurich, Switzerland
| | - Joong-Hyun Chun
- Department of Nuclear Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
45
|
Galambos J, Bielik A, Wágner G, Domány G, Kóti J, Béni Z, Szigetvári Á, Sánta Z, Orgován Z, Bobok A, Kiss B, Mikó-Bakk ML, Vastag M, Sághy K, Krasavin M, Gál K, Greiner I, Szombathelyi Z, Keserű GM. Discovery of 4-amino-3-arylsulfoquinolines, a novel non-acetylenic chemotype of metabotropic glutamate 5 (mGlu 5 ) receptor negative allosteric modulators. Eur J Med Chem 2017; 133:240-254. [DOI: 10.1016/j.ejmech.2017.03.071] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2017] [Revised: 03/09/2017] [Accepted: 03/28/2017] [Indexed: 10/19/2022]
|
46
|
Hoffmann HM, Crouzin N, Moreno E, Raivio N, Fuentes S, McCormick PJ, Ortiz J, Vignes M. Long-Lasting Impairment of mGluR5-Activated Intracellular Pathways in the Striatum After Withdrawal of Cocaine Self-Administration. Int J Neuropsychopharmacol 2016; 20:72-82. [PMID: 27744406 PMCID: PMC5412585 DOI: 10.1093/ijnp/pyw086] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 09/22/2016] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND Cocaine addiction continues to be a major heath concern, and despite public health intervention there is a lack of efficient pharmacological treatment options. A newly identified potential target are the group I metabotropic glutamate receptors, with allosteric modulators showing particular promise. METHODS We evaluated the capacity of group I metabotropic glutamate receptors to induce functional responses in ex vivo striatal slices from rats with (1) acute cocaine self-administration, (2) chronic cocaine self-administration, and (3) 60 days cocaine self-administration withdrawal by Western blot and extracellular recordings of synaptic transmission. RESULTS We found that striatal group I metabotropic glutamate receptors are the principal mediator of the mGluR1/5 agonist (RS)-3,5-dihydroxyphenylglycine-induced cAMP responsive-element binding protein phosphorylation. Both acute and chronic cocaine self-administration blunted group I metabotropic glutamate receptor effects on cAMP responsive-element binding protein phosphorylation in the striatum, which correlated with the capacity to induce long-term depression, an effect that was maintained 60 days after chronic cocaine self-administration withdrawal. In the nucleus accumbens, the principal brain region mediating the rewarding effects of drugs, chronic cocaine self-administration blunted group I metabotropic glutamate receptor stimulation of extracellular signal-regulated protein kinases 1/2 and cAMP responsive-element binding protein. Interestingly, the group I metabotropic glutamate receptor antagonist/inverse-agonist, 2-methyl-6-(phenylethynyl)pyridine hydrochloride, led to a specific increase in cAMP responsive-element binding protein phosphorylation after chronic cocaine self-administration, specifically in the nucleus accumbens, but not in the striatum. CONCLUSIONS Prolonged cocaine self-administration, through withdrawal, leads to a blunting of group I metabotropic glutamate receptor responses in the striatum. In addition, specifically in the accumbens, group I metabotropic glutamate receptor signaling to cAMP responsive-element binding protein shifts from an agonist-induced to an antagonist-induced cAMP responsive-element binding protein phosphorylation.
Collapse
Affiliation(s)
- Hanne Mette Hoffmann
- Oxidative Stress and Neuroprotection, IBMM, CNRS UMR-5247, University of Montpellier II, Montpellier, France (Drs Hoffmann, Crouzin, and Vignes); Neuroscience Institute and Department of Biochemistry and Molecular Biology, School of Medicine, Universitat Autonoma de Barcelona, Bellaterra, Spain (Dr Hoffmann, Ms Raivio, Dr Fuentes, and Dr Ortiz); Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas. Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Barcelona, Spain (Drs Moreno and McCormick); University of East Anglia, School of Pharmacy, NR4 7TJ, Norwich, United Kingdom (Dr McCormick)
| | - Nadine Crouzin
- Oxidative Stress and Neuroprotection, IBMM, CNRS UMR-5247, University of Montpellier II, Montpellier, France (Drs Hoffmann, Crouzin, and Vignes); Neuroscience Institute and Department of Biochemistry and Molecular Biology, School of Medicine, Universitat Autonoma de Barcelona, Bellaterra, Spain (Dr Hoffmann, Ms Raivio, Dr Fuentes, and Dr Ortiz); Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas. Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Barcelona, Spain (Drs Moreno and McCormick); University of East Anglia, School of Pharmacy, NR4 7TJ, Norwich, United Kingdom (Dr McCormick)
| | - Estefanía Moreno
- Oxidative Stress and Neuroprotection, IBMM, CNRS UMR-5247, University of Montpellier II, Montpellier, France (Drs Hoffmann, Crouzin, and Vignes); Neuroscience Institute and Department of Biochemistry and Molecular Biology, School of Medicine, Universitat Autonoma de Barcelona, Bellaterra, Spain (Dr Hoffmann, Ms Raivio, Dr Fuentes, and Dr Ortiz); Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas. Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Barcelona, Spain (Drs Moreno and McCormick); University of East Anglia, School of Pharmacy, NR4 7TJ, Norwich, United Kingdom (Dr McCormick)
| | - Noora Raivio
- Oxidative Stress and Neuroprotection, IBMM, CNRS UMR-5247, University of Montpellier II, Montpellier, France (Drs Hoffmann, Crouzin, and Vignes); Neuroscience Institute and Department of Biochemistry and Molecular Biology, School of Medicine, Universitat Autonoma de Barcelona, Bellaterra, Spain (Dr Hoffmann, Ms Raivio, Dr Fuentes, and Dr Ortiz); Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas. Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Barcelona, Spain (Drs Moreno and McCormick); University of East Anglia, School of Pharmacy, NR4 7TJ, Norwich, United Kingdom (Dr McCormick)
| | - Silvia Fuentes
- Oxidative Stress and Neuroprotection, IBMM, CNRS UMR-5247, University of Montpellier II, Montpellier, France (Drs Hoffmann, Crouzin, and Vignes); Neuroscience Institute and Department of Biochemistry and Molecular Biology, School of Medicine, Universitat Autonoma de Barcelona, Bellaterra, Spain (Dr Hoffmann, Ms Raivio, Dr Fuentes, and Dr Ortiz); Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas. Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Barcelona, Spain (Drs Moreno and McCormick); University of East Anglia, School of Pharmacy, NR4 7TJ, Norwich, United Kingdom (Dr McCormick)
| | - Peter J. McCormick
- Oxidative Stress and Neuroprotection, IBMM, CNRS UMR-5247, University of Montpellier II, Montpellier, France (Drs Hoffmann, Crouzin, and Vignes); Neuroscience Institute and Department of Biochemistry and Molecular Biology, School of Medicine, Universitat Autonoma de Barcelona, Bellaterra, Spain (Dr Hoffmann, Ms Raivio, Dr Fuentes, and Dr Ortiz); Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas. Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Barcelona, Spain (Drs Moreno and McCormick); University of East Anglia, School of Pharmacy, NR4 7TJ, Norwich, United Kingdom (Dr McCormick)
| | - Jordi Ortiz
- Present address (H.M.H.): Department of Reproductive Medicine, 349 Leichtag Biomedical Research Building, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0674
| | | |
Collapse
|
47
|
Gould RW, Amato RJ, Bubser M, Joffe ME, Nedelcovych MT, Thompson AD, Nickols HH, Yuh JP, Zhan X, Felts AS, Rodriguez AL, Morrison RD, Byers FW, Rook JM, Daniels JS, Niswender CM, Conn PJ, Emmitte KA, Lindsley CW, Jones CK. Partial mGlu₅ Negative Allosteric Modulators Attenuate Cocaine-Mediated Behaviors and Lack Psychotomimetic-Like Effects. Neuropsychopharmacology 2016; 41:1166-78. [PMID: 26315507 PMCID: PMC4748441 DOI: 10.1038/npp.2015.265] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 08/17/2015] [Accepted: 08/21/2015] [Indexed: 11/09/2022]
Abstract
Cocaine abuse remains a public health concern for which pharmacotherapies are largely ineffective. Comorbidities between cocaine abuse, depression, and anxiety support the development of novel treatments targeting multiple symptom clusters. Selective negative allosteric modulators (NAMs) targeting the metabotropic glutamate receptor 5 (mGlu5) subtype are currently in clinical trials for the treatment of multiple neuropsychiatric disorders and have shown promise in preclinical models of substance abuse. However, complete blockade or inverse agonist activity by some full mGlu5 NAM chemotypes demonstrated adverse effects, including psychosis in humans and psychotomimetic-like effects in animals, suggesting a narrow therapeutic window. Development of partial mGlu5 NAMs, characterized by their submaximal but saturable levels of blockade, may represent a novel approach to broaden the therapeutic window. To understand potential therapeutic vs adverse effects in preclinical behavioral assays, we examined the partial mGlu5 NAMs, M-5MPEP and Br-5MPEPy, in comparison with the full mGlu5 NAM MTEP across models of addiction and psychotomimetic-like activity. M-5MPEP, Br-5MPEPy, and MTEP dose-dependently decreased cocaine self-administration and attenuated the discriminative stimulus effects of cocaine. M-5MPEP and Br-5MPEPy also demonstrated antidepressant- and anxiolytic-like activity. Dose-dependent effects of partial and full mGlu5 NAMs in these assays corresponded with increasing in vivo mGlu5 occupancy, demonstrating an orderly occupancy-to-efficacy relationship. PCP-induced hyperlocomotion was potentiated by MTEP, but not by M-5MPEP and Br-5MPEPy. Further, MTEP, but not M-5MPEP, potentiated the discriminative-stimulus effects of PCP. The present data suggest that partial mGlu5 NAM activity is sufficient to produce therapeutic effects similar to full mGlu5 NAMs, but with a broader therapeutic index.
Collapse
Affiliation(s)
- Robert W Gould
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Russell J Amato
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Michael Bubser
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Max E Joffe
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Michael T Nedelcovych
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Analisa D Thompson
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Hilary H Nickols
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN, USA.,Department of Pathology, Microbiology and Immunology, Division of Neuropathology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Johannes P Yuh
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Xiaoyan Zhan
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Andrew S Felts
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN, USA.,Department of Chemistry, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alice L Rodriguez
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ryan D Morrison
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Frank W Byers
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jerri M Rook
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - John S Daniels
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Colleen M Niswender
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - P Jeffrey Conn
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Kyle A Emmitte
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN, USA.,Department of Chemistry, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Craig W Lindsley
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN, USA.,Department of Chemistry, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Carrie K Jones
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
48
|
Leurquin-Sterk G, Postnov A, de Laat B, Casteels C, Celen S, Crunelle CL, Bormans G, Koole M, Van Laere K. Kinetic modeling and long-term test-retest reproducibility of the mGluR5 PET tracer18F-FPEB in human brain. Synapse 2016; 70:153-62. [DOI: 10.1002/syn.21890] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 01/15/2016] [Accepted: 01/15/2016] [Indexed: 12/12/2022]
Affiliation(s)
- Gil Leurquin-Sterk
- Division of Nuclear Medicine; KU Leuven - University of Leuven and University Hospital Leuven; Herestraat 49 Leuven 3000 Belgium
| | - Andrey Postnov
- Division of Nuclear Medicine; KU Leuven - University of Leuven and University Hospital Leuven; Herestraat 49 Leuven 3000 Belgium
| | - Bart de Laat
- Division of Nuclear Medicine; KU Leuven - University of Leuven and University Hospital Leuven; Herestraat 49 Leuven 3000 Belgium
- MoSAIC, Molecular Small Animal Imaging Center, KU Leuven - University of Leuven; Leuven Belgium
| | - Cindy Casteels
- Division of Nuclear Medicine; KU Leuven - University of Leuven and University Hospital Leuven; Herestraat 49 Leuven 3000 Belgium
- MoSAIC, Molecular Small Animal Imaging Center, KU Leuven - University of Leuven; Leuven Belgium
| | - Sofie Celen
- Laboratory for Radiopharmacy, KU Leuven - University of Leuven; Leuven Belgium
| | | | - Guy Bormans
- Laboratory for Radiopharmacy, KU Leuven - University of Leuven; Leuven Belgium
| | - Michel Koole
- Division of Nuclear Medicine; KU Leuven - University of Leuven and University Hospital Leuven; Herestraat 49 Leuven 3000 Belgium
- MoSAIC, Molecular Small Animal Imaging Center, KU Leuven - University of Leuven; Leuven Belgium
| | - Koen Van Laere
- Division of Nuclear Medicine; KU Leuven - University of Leuven and University Hospital Leuven; Herestraat 49 Leuven 3000 Belgium
- MoSAIC, Molecular Small Animal Imaging Center, KU Leuven - University of Leuven; Leuven Belgium
| |
Collapse
|
49
|
Moeller SJ, London ED, Northoff G. Neuroimaging markers of glutamatergic and GABAergic systems in drug addiction: Relationships to resting-state functional connectivity. Neurosci Biobehav Rev 2016; 61:35-52. [PMID: 26657968 PMCID: PMC4731270 DOI: 10.1016/j.neubiorev.2015.11.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 11/05/2015] [Accepted: 11/21/2015] [Indexed: 12/29/2022]
Abstract
Drug addiction is characterized by widespread abnormalities in brain function and neurochemistry, including drug-associated effects on concentrations of the excitatory and inhibitory neurotransmitters glutamate and gamma-aminobutyric acid (GABA), respectively. In healthy individuals, these neurotransmitters drive the resting state, a default condition of brain function also disrupted in addiction. Here, our primary goal was to review in vivo magnetic resonance spectroscopy and positron emission tomography studies that examined markers of glutamate and GABA abnormalities in human drug addiction. Addicted individuals tended to show decreases in these markers compared with healthy controls, but findings also varied by individual characteristics (e.g., abstinence length). Interestingly, select corticolimbic brain regions showing glutamatergic and/or GABAergic abnormalities have been similarly implicated in resting-state functional connectivity deficits in drug addiction. Thus, our secondary goals were to provide a brief review of this resting-state literature, and an initial rationale for the hypothesis that abnormalities in glutamatergic and/or GABAergic neurotransmission may underlie resting-state functional deficits in drug addiction. In doing so, we suggest future research directions and possible treatment implications.
Collapse
Affiliation(s)
- Scott J Moeller
- Departments of Psychiatry and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Edythe D London
- Departments of Psychiatry and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Departments of Psychiatry and Biobehavioral Sciences, and Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA, USA
| | - Georg Northoff
- Brain Imaging and Neuroethics Research Unit, Institute of Mental Health Research, Ottawa, Canada.
| |
Collapse
|
50
|
Mihov Y, Hasler G. Negative Allosteric Modulators of Metabotropic Glutamate Receptors Subtype 5 in Addiction: a Therapeutic Window. Int J Neuropsychopharmacol 2016; 19:pyw002. [PMID: 26802568 PMCID: PMC4966271 DOI: 10.1093/ijnp/pyw002] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 01/08/2016] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Abundant evidence at the anatomical, electrophysiological, and molecular levels implicates metabotropic glutamate receptor subtype 5 (mGluR5) in addiction. Consistently, the effects of a wide range of doses of different mGluR5 negative allosteric modulators (NAMs) have been tested in various animal models of addiction. Here, these studies were subjected to a systematic review to find out if mGluR5 NAMs have a therapeutic potential that can be translated to the clinic. METHODS Literature on consumption/self-administration and reinstatement of drug seeking as outcomes of interest published up to April 2015 was retrieved via PubMed. The review focused on the effects of systemic (i.p., i.v., s.c.) administration of the mGluR5 NAMs 3-((2-Methyl-4-thiazolyl)ethynyl)pyridine (MTEP) and 2-Methyl-6-(phenylethynyl)pyridine (MPEP) on paradigms with cocaine, ethanol, nicotine, and food in rats. RESULTS MTEP and MPEP were found to reduce self-administration of cocaine, ethanol, and nicotine at doses ≥1mg/kg and 2.5mg/kg, respectively. Dose-response relationship resembled a sigmoidal curve, with low doses not reaching statistical significance and high doses reliably inhibiting self-administration of drugs of abuse. Importantly, self-administration of cocaine, ethanol, and nicotine, but not food, was reduced by MTEP and MPEP in the dose range of 1 to 2mg/kg and 2.5 to 3.2mg/kg, respectively. This dose range corresponds to approximately 50% to 80% mGluR5 occupancy. Interestingly, the limited data found in mice and monkeys showed a similar therapeutic window. CONCLUSION Altogether, this review suggests a therapeutic window for mGluR5 NAMs that can be translated to the treatment of substance-related and addictive disorders.
Collapse
Affiliation(s)
- Yoan Mihov
- Division of Molecular Psychiatry, Translational Research Center, Psychiatric University Hospital, University of Bern, Switzerland
| | - Gregor Hasler
- Division of Molecular Psychiatry, Translational Research Center, Psychiatric University Hospital, University of Bern, Switzerland
| |
Collapse
|