1
|
Kos J, Langiu M, Hellyer SD, Gregory KJ. Pharmacology, Signaling and Therapeutic Potential of Metabotropic Glutamate Receptor 5 Negative Allosteric Modulators. ACS Pharmacol Transl Sci 2024; 7:3671-3690. [PMID: 39698283 PMCID: PMC11651194 DOI: 10.1021/acsptsci.4c00213] [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] [Received: 04/11/2024] [Revised: 06/18/2024] [Accepted: 07/01/2024] [Indexed: 12/20/2024]
Abstract
Metabotropic glutamate receptors are a family of eight class C G protein-coupled receptors regulating higher order brain functions including cognition and motion. Metabotropic glutamate receptors have thus been heavily investigated as potential drug targets for treating neurological disorders. Drug discovery efforts directed toward metabotropic glutamate receptor subtype 5 (mGlu5) have been particularly fruitful, with a wealth of drug candidates and pharmacological tools identified. mGlu5 negative allosteric modulators (NAMs) are promising novel therapeutics for developmental, neuropsychiatric and neurodegenerative disorders (e.g., Alzheimer's Disease, Huntington's Disease, Parkinson's Disease, amyotrophic lateral sclerosis, autism spectrum disorders, substance use disorders, stroke, anxiety and depression) and show promise in ameliorating adverse effects induced by other medications (e.g., L-dopa induced dyskinesia in Parkinson's Disease). However, despite preclinical success, mGlu5 NAMs are yet to reach the market due to poor safety and efficacy profiles in clinical trials. Herein, we review the physiology and signal transduction of mGlu5. We provide a comprehensive critique of therapeutic options with respect to mGlu5 inhibitors, spanning from orthosteric antagonists to NAMs. Finally, we address the challenges associated with drug development and highlight future directions to guide rational drug discovery of safe and effective novel therapeutics.
Collapse
Affiliation(s)
- Jackson
A. Kos
- Drug
Discovery Biology, Monash Institute of Pharmaceutical Sciences and
Department of Pharmacology, Monash University, Parkville, VIC 3052, Australia
| | - Monica Langiu
- Drug
Discovery Biology, Monash Institute of Pharmaceutical Sciences and
Department of Pharmacology, Monash University, Parkville, VIC 3052, Australia
| | - Shane D. Hellyer
- Drug
Discovery Biology, Monash Institute of Pharmaceutical Sciences and
Department of Pharmacology, Monash University, Parkville, VIC 3052, Australia
| | - Karen J. Gregory
- Drug
Discovery Biology, Monash Institute of Pharmaceutical Sciences and
Department of Pharmacology, Monash University, Parkville, VIC 3052, Australia
- ARC
Centre for Cryo-electron Microscopy of Membrane Proteins, Monash University, Parkville, VIC 3052, Australia
| |
Collapse
|
2
|
Grassi G, Scillitani E, Cecchelli C. New horizons for obsessive-compulsive disorder drug discovery: is targeting glutamate receptors the answer? Expert Opin Drug Discov 2024; 19:1235-1245. [PMID: 39105546 DOI: 10.1080/17460441.2024.2387127] [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: 04/06/2024] [Accepted: 07/29/2024] [Indexed: 08/07/2024]
Abstract
INTRODUCTION Over the past decade, glutamate has emerged as a prominent focus in the field of obsessive-compulsive disorder (OCD) pathophysiology. A convergence of evidence from genetic, preclinical, and clinical studies points to glutamatergic dysfunction as a key feature of this condition. In light of these findings, there has been a growing interest in exploring the potential of glutamatergic agents in the treatment of OCD. AREAS COVERED This paper reviews the literature on glutamate transmission in OCD. In addition, the authors examine the results of clinical trials investigating the efficacy of glutamatergic agents in the treatment of OCD patients. EXPERT OPINION Along with the recognition of neuroinflammation in the brain in OCD, the evidence of glutamate dysfunction represents one of the most promising recent discoveries for understanding the mechanisms involved in OCD. The importance of this discovery lies primarily in its pharmacological implications and has led to intense research activity in the field of glutamatergic agents. While this research has not yet had a substantial clinical impact, targeting glutamate receptors remains a promising horizon for the successful treatment of OCD patients.
Collapse
Affiliation(s)
- Giacomo Grassi
- Department of Psychiatry, Brain Center Firenze, Florence, Italy
| | | | | |
Collapse
|
3
|
Issa ASM, Scheins J, Tellmann L, Brambilla CR, Lohmann P, Rota-Kops E, Herzog H, Neuner I, Shah NJ, Lerche C. Impact of improved dead time correction on the quantification accuracy of a dedicated BrainPET scanner. PLoS One 2024; 19:e0296357. [PMID: 38578749 PMCID: PMC10997125 DOI: 10.1371/journal.pone.0296357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 12/11/2023] [Indexed: 04/07/2024] Open
Abstract
OBJECTIVE Quantitative values derived from PET brain images are of high interest for neuroscientific applications. Insufficient DT correction (DTC) can lead to a systematic bias of the output parameters obtained by a detailed analysis of the time activity curves (TACs). The DTC method currently used for the Siemens 3T MR BrainPET insert is global, i.e., differences in DT losses between detector blocks are not considered, leading to inaccurate DTC and, consequently, to inaccurate measurements masked by a bias. However, following careful evaluation with phantom measurements, a new block-pairwise DTC method has demonstrated a higher degree of accuracy compared to the global DTC method. APPROACH Differences between the global and the block-pairwise DTC method were studied in this work by applying several radioactive tracers. We evaluated the impact on [11C]ABP688, O-(2-[18F]fluoroethyl)-L-tyrosine (FET), and [15O]H2O TACs. RESULTS For [11C]ABP688, a relevant bias of between -0.0034 and -0.0053 ml/ (cm3 • min) was found in all studied brain regions for the volume of distribution (VT) when using the current global DTC method. For [18F]FET-PET, differences of up to 10% were observed in the tumor-to-brain ratio (TBRmax), these differences depend on the radial distance of the maximum from the PET isocenter. For [15O]H2O, differences between +4% and -7% were observed in the GM region. Average biases of -4.58%, -3.2%, and -1.2% for the regional cerebral blood flow (CBF (K1)), the rate constant k2, and the volume of distribution VT were observed, respectively. Conversely, in the white matter region, average biases of -4.9%, -7.0%, and 3.8% were observed for CBF (K1), k2, and VT, respectively. CONCLUSION The bias introduced by the global DTC method leads to an overestimation in the studied quantitative parameters for all applications compared to the block-pairwise method. SIGNIFICANCE The observed differences between the two DTC methods are particularly relevant for research applications in neuroscientific studies as they affect the accuracy of quantitative Brain PET images.
Collapse
Affiliation(s)
- Ahlam Said Mohamad Issa
- Institute of Neuroscience and Medicine 4, INM-4, Forschungszentrum Jülich, Jülich, Germany
- JARA, BRAIN, Translational Medicine, Aachen, Germany
- Department of Neurology, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Jürgen Scheins
- Institute of Neuroscience and Medicine 4, INM-4, Forschungszentrum Jülich, Jülich, Germany
| | - Lutz Tellmann
- Institute of Neuroscience and Medicine 4, INM-4, Forschungszentrum Jülich, Jülich, Germany
| | | | - Philipp Lohmann
- Institute of Neuroscience and Medicine 4, INM-4, Forschungszentrum Jülich, Jülich, Germany
| | - Elena Rota-Kops
- Institute of Neuroscience and Medicine 4, INM-4, Forschungszentrum Jülich, Jülich, Germany
| | - Hans Herzog
- Institute of Neuroscience and Medicine 4, INM-4, Forschungszentrum Jülich, Jülich, Germany
| | - Irene Neuner
- Institute of Neuroscience and Medicine 4, INM-4, Forschungszentrum Jülich, Jülich, Germany
- JARA, BRAIN, Translational Medicine, Aachen, Germany
- Department of Psychiatry, Psychotherapy and Psychosomatics, RWTH Aachen University, Aachen, Germany
| | - N. Jon Shah
- Institute of Neuroscience and Medicine 4, INM-4, Forschungszentrum Jülich, Jülich, Germany
- JARA, BRAIN, Translational Medicine, Aachen, Germany
- Department of Neurology, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
- Institute of Neuroscience and Medicine 11, INM-11, JARA, Forschungszentrum Jülich, Jülich, Germany
| | - Christoph Lerche
- Institute of Neuroscience and Medicine 4, INM-4, Forschungszentrum Jülich, Jülich, Germany
| |
Collapse
|
4
|
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
|
5
|
Witkin JM, Pandey KP, Smith JL. Clinical investigations of compounds targeting metabotropic glutamate receptors. Pharmacol Biochem Behav 2022; 219:173446. [PMID: 35987339 DOI: 10.1016/j.pbb.2022.173446] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 06/22/2022] [Accepted: 08/08/2022] [Indexed: 11/15/2022]
Abstract
Pharmacological modulation of glutamate has long been considered to be of immense therapeutic utility. The metabotropic glutamate receptors (mGluRs) are potential targets for safely altering glutamate-driven excitation. Data support the potential therapeutic use of mGluR modulators in the treatment of anxiety, depression, schizophrenia, and other psychiatric disorders, pain, epilepsy, as well as neurodegenerative and neurodevelopmental disorders. For each of the three mGluR groups, compounds have been constructed that produce either potentiation or functional blockade. PET ligands for mGlu5Rs have been studied in a range of patient populations and several mGlu5R antagonists have been tested for potential efficacy in patients including mavoglurant, diploglurant, basimglurant, GET 73, and ADX10059. Efficacy with mGlu5R antagonists has been reported in trials with patients with gastroesophageal reflux disease; data from patients with Parkinson's disease or Fragile X syndrome have not been as robust as hoped. Fenobam was approved for use as an anxiolytic prior to its recognition as an mGlu5R antagonist. mGlu2/3R agonists (pomaglumated methionil) and mGlu2R agonists (JNJ-40411813, AZD 8529, and LY2979165) have been studied in patients with schizophrenia with promising but mixed results. Antagonists of mGlu2/3Rs (decoglurant and TS-161) have been studied in depression where TS-161 has advanced into a planned Phase 2 study in treatment-resistant depression. The Group III mGluRs are the least developed of the mGluR receptor targets. The mGlu4R potentiator, foliglurax, did not meet its primary endpoint in patients with Parkinson's disease. Ongoing efforts to develop mGluR-targeted compounds continue to promise these glutamate modulators as medicines for psychiatric and neurological disorders.
Collapse
Affiliation(s)
- Jeffrey M Witkin
- Laboratory of Antiepileptic Drug Discovery, Ascension St. Vincent, Indianapolis, IN, USA; Department of Chemistry & Biochemistry, Milwaukee Institute of Drug Discovery, University of Wisconsin-Milwaukee, Milwaukee, WI, USA; RespireRx Pharmaceuticals Inc, Glen Rock, NJ, USA.
| | - Kamal P Pandey
- Department of Chemistry & Biochemistry, Milwaukee Institute of Drug Discovery, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - Jodi L Smith
- Laboratory of Antiepileptic Drug Discovery, Ascension St. Vincent, Indianapolis, IN, USA
| |
Collapse
|
6
|
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: 15] [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
|
7
|
Glorie D, Verhaeghe J, Miranda A, De Lombaerde S, Stroobants S, Staelens S. Quantification of Metabotropic Glutamate Receptor 5 Availability With Both [ 11C]ABP688 and [ 18F]FPEB Positron Emission Tomography in the Sapap3 Knockout Mouse Model for Obsessive-Compulsive-like Behavior. BIOLOGICAL PSYCHIATRY. COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2022; 7:607-615. [PMID: 34856382 DOI: 10.1016/j.bpsc.2021.11.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 11/17/2021] [Accepted: 11/20/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND This study provides a first direct comparison between positron emission tomography radioligands targeting the allosteric site of the metabotropic glutamate receptor 5 (mGluR5): [11C]ABP688 and [18F]FPEB. A blocking paradigm was set up to substantiate the common binding site of both radioligands. Second, both radioligands were applied in Sapap3 knockout (KO) mice showing compulsive-like behavior characterized by a lower in vivo mGluR5 availability. METHODS First, wild-type mice (n = 7) received four position emission tomography/computed tomography scans: a [11C]ABP688 scan, a [18F]FPEB scan, and two blocking scans using cold FPEB and cold ABP688, respectively. A second experiment compared both radioligands in wild-type (n = 7) and KO (n = 10) mice. The simplified reference tissue model was used to calculate the nondisplaceable binding potential representing the in vivo availability of mGluR5 in the brain. RESULTS Using cold FPEB as a blocking compound for [11C]ABP688 micro-positron emission tomography and vice versa, we observed averaged global reductions in mGluR5 availability of circa 98% for [11C]ABP688 and 82%-96% for [18F]FPEB. For KOs, the [11C]ABP688 nondisplaceable binding potential was on average 25% lower compared with wild-type control mice (p < .0001-.001), while this was about 17% for [18F]FPEB (p < .05). CONCLUSIONS The current findings substantiate a common binding site and suggest a strong relationship between mGluR5 availability levels measured with both radioligands. In Sapap3 KO mice, a reduced mGluR5 availability could therefore be demonstrated with both radioligands. With [11C]ABP688, higher significance levels were achieved in more brain regions. These findings suggest [11C]ABP688 as a preferable radiotracer to quantify mGluR5 availability, as exemplified here in a model for compulsive-like behavior.
Collapse
Affiliation(s)
- Dorien Glorie
- Molecular Imaging Center Antwerp, University of Antwerp, Wilrijk, Belgium
| | - Jeroen Verhaeghe
- Molecular Imaging Center Antwerp, University of Antwerp, Wilrijk, Belgium
| | - Alan Miranda
- Molecular Imaging Center Antwerp, University of Antwerp, Wilrijk, Belgium
| | - Stef De Lombaerde
- Molecular Imaging Center Antwerp, University of Antwerp, Wilrijk, Belgium; Department of Nuclear Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Sigrid Stroobants
- Molecular Imaging Center Antwerp, University of Antwerp, Wilrijk, Belgium; Department of Nuclear Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Steven Staelens
- Molecular Imaging Center Antwerp, University of Antwerp, Wilrijk, Belgium.
| |
Collapse
|
8
|
mGluR5 binding changes during a mismatch negativity task in a multimodal protocol with [ 11C]ABP688 PET/MR-EEG. Transl Psychiatry 2022; 12:6. [PMID: 35013095 PMCID: PMC8748790 DOI: 10.1038/s41398-021-01763-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 11/22/2021] [Accepted: 11/30/2021] [Indexed: 02/08/2023] Open
Abstract
Currently, the metabotropic glutamate receptor 5 (mGluR5) is the subject of several lines of research in the context of neurology and is of high interest as a target for positron-emission tomography (PET). Here, we assessed the feasibility of using [11C]ABP688, a specific antagonist radiotracer for an allosteric site on the mGluR5, to evaluate changes in glutamatergic neurotransmission through a mismatch-negativity (MMN) task as a part of a simultaneous and synchronized multimodal PET/MR-EEG study. We analyzed the effect of MMN by comparing the changes in nondisplaceable binding potential (BPND) prior to (baseline) and during the task in 17 healthy subjects by applying a bolus/infusion protocol. Anatomical and functional regions were analyzed. A small change in BPND was observed in anatomical regions (posterior cingulate cortex and thalamus) and in a functional network (precuneus) after the start of the task. The effect size was quantified using Kendall's W value and was 0.3. The motor cortex was used as a control region for the task and did not show any significant BPND changes. There was a significant ΔBPND between acquisition conditions. On average, the reductions in binding across the regions were - 8.6 ± 3.2% in anatomical and - 6.4 ± 0.5% in the functional network (p ≤ 0.001). Correlations between ΔBPND and EEG latency for both anatomical (p = 0.008) and functional (p = 0.022) regions were found. Exploratory analyses suggest that the MMN task played a role in the glutamatergic neurotransmission, and mGluR5 may be indirectly modulated by these changes.
Collapse
|
9
|
Zangrandi L, Schmuckermair C, Ghareh H, Castaldi F, Heilbronn R, Zernig G, Ferraguti F, Ramos-Prats A. Loss of mGluR5 in D1 Receptor-Expressing Neurons Improves Stress Coping. Int J Mol Sci 2021; 22:ijms22157826. [PMID: 34360592 PMCID: PMC8346057 DOI: 10.3390/ijms22157826] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 07/11/2021] [Accepted: 07/19/2021] [Indexed: 11/16/2022] Open
Abstract
The metabotropic glutamate receptor type 5 (mGluR5) has been proposed to play a crucial role in the selection and regulation of cognitive, affective, and emotional behaviors. However, the mechanisms by which these receptors mediate these effects remain largely unexplored. Here, we studied the role of mGluR5 located in D1 receptor-expressing (D1) neurons in the manifestation of different behavioral expressions. Mice with conditional knockout (cKO) of mGluR5 in D1 neurons (mGluR5D1 cKO) and littermate controls displayed similar phenotypical profiles in relation to memory expression, anxiety, and social behaviors. However, mGluR5D1 cKO mice presented different coping mechanisms in response to acute escapable or inescapable stress. mGluR5D1 cKO mice adopted an enhanced active stress coping strategy upon exposure to escapable stress in the two-way active avoidance (TWA) task and a greater passive strategy upon exposure to inescapable stress in the forced swim test (FST). In summary, this work provides evidence for a functional integration of the dopaminergic and glutamatergic system to mediate control over internal states upon stress exposure and directly implicates D1 neurons and mGluR5 as crucial mediators of behavioral stress responses.
Collapse
Affiliation(s)
- Luca Zangrandi
- Department of Neurology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (L.Z.); (R.H.)
- Institute of Pharmacology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (C.S.); (F.C.); (F.F.)
| | - Claudia Schmuckermair
- Institute of Pharmacology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (C.S.); (F.C.); (F.F.)
| | - Hussein Ghareh
- Department of Psychiatry 1, Medical University of Innsbruck, 6020 Innsbruck, Austria; (H.G.); (G.Z.)
| | - Federico Castaldi
- Institute of Pharmacology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (C.S.); (F.C.); (F.F.)
| | - Regine Heilbronn
- Department of Neurology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (L.Z.); (R.H.)
| | - Gerald Zernig
- Department of Psychiatry 1, Medical University of Innsbruck, 6020 Innsbruck, Austria; (H.G.); (G.Z.)
| | - Francesco Ferraguti
- Institute of Pharmacology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (C.S.); (F.C.); (F.F.)
| | - Arnau Ramos-Prats
- Institute of Pharmacology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (C.S.); (F.C.); (F.F.)
- Correspondence:
| |
Collapse
|
10
|
Targeting metabotropic glutamate receptors for the treatment of depression and other stress-related disorders. Neuropharmacology 2021; 196:108687. [PMID: 34175327 DOI: 10.1016/j.neuropharm.2021.108687] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 06/11/2021] [Accepted: 06/18/2021] [Indexed: 12/15/2022]
Abstract
The discovery of robust antidepressant effects of ketamine in refractory patients has led to increasing focus on agents targeting glutamatergic signaling as potential novel antidepressant strategy. Among the agents targeting the glutamatergic system, compounds acting at metabotropic glutamate (mGlu) receptors are among the most promising agents under studies for depressive disorders. Further, the receptor diversity, distinct distribution in the CNS, and ability to modulate the glutamatergic neurotransmission in the brain areas implicated in mood disorders make them an exciting target for stress-related disorders. In preclinical models, antidepressant and anxiolytic effects of mGlu5 negative allosteric modulators (NAMs) have been reported. Interestingly, mGlu2/3 receptor antagonists show fast and sustained antidepressant-like effects similar to that of ketamine in rodents. Excitingly, they can also induce antidepressant effects in the animal models of treatment-resistant depression and are devoid of the side-effects associated with ketamine. Unfortunately, clinical trials of both mGlu5 and mGlu2/3 receptor NAMs have been inconclusive, and additional trials using other compounds with suitable preclinical and clinical properties are needed. Although group III mGlu receptors have gained less attention, mGlu7 receptor ligands have been shown to induce antidepressant-like effects in rodents. Collectively, compounds targeting mGlu receptors provide an alternative approach to fill the outstanding clinical need for safer and more efficacious antidepressants. This article is part of the special Issue on "Glutamate Receptors - mGluRs".
Collapse
|
11
|
Streffer J, Treyer V, Buck A, Ametamey SM, Blagoev M, Maguire RP, Gautier A, Auberson YP, Schmidt ME, Vranesic IT, Gomez-Mancilla B, Gasparini F. Regional brain mGlu5 receptor occupancy following single oral doses of mavoglurant as measured by [ 11C]-ABP688 PET imaging in healthy volunteers. Neuroimage 2021; 230:117785. [PMID: 33545349 DOI: 10.1016/j.neuroimage.2021.117785] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 11/17/2020] [Accepted: 01/13/2021] [Indexed: 11/16/2022] Open
Abstract
Mavoglurant binds to same allosteric site on metabotropic glutamate receptor 5 (mGluR5) as [11C]-ABP688, a radioligand. This open-label, single-center pilot study estimates extent of occupancy of mGluR5 receptors following single oral doses of mavoglurant, using [11C]-ABP688 positron emission tomography (PET) imaging, in six healthy males aged 20-40 years. This study comprised three periods and six subjects were divided into two cohorts. On Day 1 (Period 1), baseline clinical data and safety samples were obtained along with PET scan. During Period 2 (1-7 days after Period 1), cohort 1 and 2 received mavoglurant 25 mg and 100 mg, respectively. During Period 3 (7 days after Period 2), cohort 1 and 2 received mavoglurant 200 mg and 400 mg, respectively. Mavoglurant showed the highest distribution volumes in the cingulate region with lower uptake in cerebellum and white matter, possibly because myelinated axonal sheets maybe devoid of mGlu5 receptors. Maximum concentrations of mavoglurant were observed around 2-3.25 h post-dose. Mavoglurant passed the blood-brain barrier and induced dose- and exposure-dependent displacement of [11C]-ABP688 from the mGluR5 receptors, 3-4 h post-administration (27%, 59%, 74%, 85% receptor occupancy for mavoglurant 25 mg, 100 mg, 200 mg, 400 mg dose, respectively). There were no severe adverse effects or clinically significant changes in safety parameters. This is the first human receptor occupancy study completed with Mavoglurant. It served to guide the dosing of mavoglurant in the past and currently ongoing clinical studies. Furthermore, it confirms the utility of [11C]-ABP688 as a unique tool to study drug-induced occupancy of mGlu5 receptors in the living human brain.
Collapse
Affiliation(s)
- Johannes Streffer
- Division of Psychiatric Research, University of Zurich, Zurich, Switzerland
| | - Valerie Treyer
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Alfred Buck
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Simon M Ametamey
- Radiopharmaceutical Sciences, Institute of Pharmaceutical Sciences, Zurich, Switzerland
| | - Milen Blagoev
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Ralph P Maguire
- Novartis Institutes for Biomedical Research, Novartis Pharma AG, Postfach, Basel CH-4002, Switzerland
| | - Aurélie Gautier
- Global Drug Development, Novartis Pharma AG, Basel, Switzerland
| | - Yves P Auberson
- Novartis Institutes for Biomedical Research, Novartis Pharma AG, Postfach, Basel CH-4002, Switzerland
| | - Mark E Schmidt
- Novartis Institutes for Biomedical Research, Novartis Pharma AG, Postfach, Basel CH-4002, Switzerland
| | - Ivan-Toma Vranesic
- Novartis Institutes for Biomedical Research, Novartis Pharma AG, Postfach, Basel CH-4002, Switzerland
| | - Baltazar Gomez-Mancilla
- Novartis Institutes for Biomedical Research, Novartis Pharma AG, Postfach, Basel CH-4002, Switzerland
| | - Fabrizio Gasparini
- Novartis Institutes for Biomedical Research, Novartis Pharma AG, Postfach, Basel CH-4002, Switzerland.
| |
Collapse
|
12
|
Brambilla CR, Scheins J, Issa A, Tellmann L, Herzog H, Rota Kops E, Shah NJ, Neuner I, Lerche CW. Bias evaluation and reduction in 3D OP-OSEM reconstruction in dynamic equilibrium PET studies with 11C-labeled for binding potential analysis. PLoS One 2021; 16:e0245580. [PMID: 33481896 PMCID: PMC7822533 DOI: 10.1371/journal.pone.0245580] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 01/05/2021] [Indexed: 11/26/2022] Open
Abstract
Iterative image reconstruction is widely used in positron emission tomography. However, it is known to contribute to quantitation bias and is particularly pronounced during dynamic studies with 11C-labeled radiotracers where count rates become low towards the end of the acquisition. As the strength of the quantitation bias depends on the counts in the reconstructed frame, it can differ from frame to frame of the acquisition. This is especially relevant in the case of neuro-receptor studies with simultaneous PET/MR when a bolus-infusion protocol is applied to allow the comparison of pre- and post-task effects. Here, count dependent changes in quantitation bias may interfere with task changes. We evaluated the impact of different framing schemes on quantitation bias and its propagation into binding potential (BP) using a phantom decay study with 11C and 3D OP-OSEM. Further, we propose a framing scheme that keeps the true counts per frame constant over the acquisition time as constant framing schemes and conventional increasing framing schemes are unlikely to achieve stable bias values during the acquisition time range. For a constant framing scheme with 5 minutes frames, the BP bias was 7.13±2.01% (10.8% to 3.8%) compared to 5.63±2.85% (7.8% to 4.0%) for conventional increasing framing schemes. Using the proposed constant true counts framing scheme, a stabilization of the BP bias was achieved at 2.56±3.92% (3.5% to 1.7%). The change in BP bias was further studied by evaluating the linear slope during the acquisition time interval. The lowest slope values were observed in the constant true counts framing scheme. The constant true counts framing scheme was effective for BP bias stabilization at relevant activity and time ranges. The mean BP bias under these conditions was 2.56±3.92%, which represents the lower limit for the detection of changes in BP during equilibrium and is especially important in the case of cognitive tasks where the expected changes are low.
Collapse
Affiliation(s)
- Cláudia Régio Brambilla
- Institute of Neuroscience and Medicine, INM-4, Forschungszentrum Jülich GmbH, Jülich, Germany
- Department of Psychiatry, Psychotherapy and Psychosomatics, RWTH Aachen University, Aachen, Germany
- * E-mail:
| | - Jürgen Scheins
- Institute of Neuroscience and Medicine, INM-4, Forschungszentrum Jülich GmbH, Jülich, Germany
| | - Ahlam Issa
- Institute of Neuroscience and Medicine, INM-4, Forschungszentrum Jülich GmbH, Jülich, Germany
| | - Lutz Tellmann
- Institute of Neuroscience and Medicine, INM-4, Forschungszentrum Jülich GmbH, Jülich, Germany
| | - Hans Herzog
- Institute of Neuroscience and Medicine, INM-4, Forschungszentrum Jülich GmbH, Jülich, Germany
| | - Elena Rota Kops
- Institute of Neuroscience and Medicine, INM-4, Forschungszentrum Jülich GmbH, Jülich, Germany
| | - N. Jon Shah
- Institute of Neuroscience and Medicine, INM-4, Forschungszentrum Jülich GmbH, Jülich, Germany
- Institute of Neuroscience and Medicine, INM-11, Forschungszentrum Jülich GmbH, Jülich, Germany
- JARA–BRAIN–Translational Medicine, RWTH Aachen University, Aachen, Germany
- Department of Neurology, RWTH Aachen University, Aachen, Germany
| | - Irene Neuner
- Institute of Neuroscience and Medicine, INM-4, Forschungszentrum Jülich GmbH, Jülich, Germany
- Department of Psychiatry, Psychotherapy and Psychosomatics, RWTH Aachen University, Aachen, Germany
- JARA–BRAIN–Translational Medicine, RWTH Aachen University, Aachen, Germany
| | - Christoph W. Lerche
- Institute of Neuroscience and Medicine, INM-4, Forschungszentrum Jülich GmbH, Jülich, Germany
| |
Collapse
|
13
|
Biria M, Cantonas LM, Banca P. Magnetic Resonance Spectroscopy (MRS) and Positron Emission Tomography (PET) Imaging in Obsessive-Compulsive Disorder. Curr Top Behav Neurosci 2021; 49:231-268. [PMID: 33751502 DOI: 10.1007/7854_2020_201] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Obsessive-compulsive disorder (OCD) is characterised by structural and functional deficits in the cortico-striato-thalamic-cortical (CSTC) circuitry and abnormal neurochemical changes are thought to modulate these deficits. The hypothesis that an imbalanced concentration of the brain neurotransmitters, in particular glutamate (Glu) and gamma-amino-butyric acid (GABA), could impair the normal functioning of the CSTC, thus leading to OCD symptoms, has been tested in humans using magnetic resonance spectroscopy (MRS) and positron emission tomography (PET). This chapter summarises these neurochemical findings and represents an attempt to condense such scattered literature. We also discuss potential challenges in the field that may explain the inconsistent findings and suggest ways to overcome them. There is some convergent research from MRS pointing towards abnormalities in the brain concentration of neurometabolite markers of neuronal integrity, such as N-acetylaspartate (NAA) and choline (Cho). Lower NAA levels have been found in dorsal and rostral ACC of OCD patients (as compared to healthy volunteers), which increase after CBT and SSRI treatment, and higher Cho concentration has been reported in the thalamus of the OCD brain. However, findings for other neurometabolites are very inconsistent. Studies have reported abnormalities in the concentrations of creatine (Cr), GABA, glutamate (Glu), glutamine (Gln), Ins (myo-inositol), and serotonin (5-HT), but most of the results were not replicated. The question remains whether the NAA and Cho findings are genuinely the only neurochemical abnormalities in OCD or whether the lack of consistent findings for the other neurometabolites is caused by the lower magnetic field (1-3 Tesla (T)) used by the studies conducted so far, their small sample sizes or a lack of proper control for medication effects. To answer these questions and to further inform the biological underpinning of the symptoms and the cognitive problems at the basis of OCD we need better controlled studies using clear medicated vs unmedicated groups, larger sample sizes, stronger magnetic fields (e.g. at 7 T), and more consistency in the definition of the regions of interest.
Collapse
Affiliation(s)
- Marjan Biria
- Department of Psychology, University of Cambridge, Cambridge, UK.
| | | | - Paula Banca
- Department of Psychology, University of Cambridge, Cambridge, UK
| |
Collapse
|
14
|
Abstract
Initial reports supporting the possibility of inflammation in the brain in obsessive-compulsive disorder (OCD) evolved from the models of Sydenham's Chorea, and Pediatric Autoimmune Neuropsychiatric Disorder Associated with Streptococcus (PANDAS), which implicated excessive autoimmune responses following exposure to group A B-hemolytic streptococcal infections. Subsequently, this model was expanded to Pediatric Autoimmune Neuropsychiatric Syndrome (PANS) which applied the same concept but included other infections. A critical shortcoming of this model was that it was attributable to a small minority of OCD cases. The relationship between inflammation and OCD was more broadly demonstrated through translocator protein (TSPO) positron emission tomography imaging, a method that detects gliosis, an important component of brain inflammation, in neuropsychiatric diseases, including morphological activation and proliferation of microglia and to some extent astroglia. This method identified greater TSPO binding in the cortico-striatal-thalamo-cortical circuit in OCD, providing a direct brain measure of an important component of inflammation. To identify OCD cases with prominent elevations in TSPO binding in clinical research settings with lower cost peripheral markers, a promising approach is to apply blood serum biomarkers of inflammatory molecules produced by activated microglia and astroglia (gliosis). Such measures may aid stratification in future clinical trials. Several inflammatory-modifying interventions, including celecoxib, minocycline, and n-acetylcysteine, have been tested as treatments in randomized double-blind placebo controlled clinical trials and there is a tendency toward positive results, although these medications are not optimized for brain penetration and sample sizes for most trials were small. Future clinical trials of medications that target gliosis in OCD should apply larger sample sizes, ideally incorporating stratification approaches to enrich samples for the presence of gliosis.
Collapse
|
15
|
Holmes SE, Gallezot JD, Davis MT, DellaGioia N, Matuskey D, Nabulsi N, Krystal JH, Javitch JA, DeLorenzo C, Carson RE, Esterlis I. Measuring the effects of ketamine on mGluR5 using [ 18F]FPEB and PET. J Cereb Blood Flow Metab 2020; 40:2254-2264. [PMID: 31744389 PMCID: PMC7585925 DOI: 10.1177/0271678x19886316] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 09/23/2019] [Accepted: 10/03/2019] [Indexed: 01/21/2023]
Abstract
The metabotropic glutamate receptor 5 (mGluR5) is a promising treatment target for psychiatric disorders due to its modulatory effects on glutamate transmission. Using [11C]ABP688, we previously showed that the rapidly acting antidepressant ketamine decreases mGluR5 availability. The mGluR5 radioligand [18F]FPEB offers key advantages over [11C]ABP688; however, its suitability for drug challenge studies is unknown. We evaluated whether [18F]FPEB can be used to capture ketamine-induced effects on mGluR5. Seven healthy subjects participated in three [18F]FPEB scans: a baseline, a same-day post-ketamine, and a 24-h post-ketamine scan. The outcome measure was VT/fP, obtained using a two-tissue compartment model and a metabolite-corrected arterial input function. Dissociative symptoms, heart rate and blood pressure increased following ketamine infusion. [18F]FPEB VT/fP decreased by 9% across the cortex after ketamine infusion, with minimal difference between baseline and 24-h scans. Compared to our previous work using [11C]ABP688, the magnitude of the ketamine-induced change in mGluR5 was smaller using [18F]FPEB; however, effect sizes were similar for the same-day post-ketamine vs. baseline scan (Cohen's d = 0.75 for [18F]FPEB and 0.88 for [11C]ABP688). [18F]FPEB is therefore able to capture some of the effects of ketamine on mGluR5, but [11C]ABP688 appears to be more suitable in drug challenge paradigms designed to probe glutamate transmission.
Collapse
Affiliation(s)
- Sophie E Holmes
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | | | - Margaret T Davis
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | - Nicole DellaGioia
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | - David Matuskey
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
| | - Nabeel Nabulsi
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | - John H Krystal
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- U.S. Department of Veteran Affairs National Center for Posttraumatic Stress Disorder, Clinical Neurosciences Division, VA Connecticut Healthcare System, West Haven, CT, USA
| | - Jonathan A Javitch
- Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, USA
- Departments of Psychiatry and Pharmacology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Christine DeLorenzo
- Department of Psychiatry and Behavioral Health, Stony Brook University, New York, NY, USA
| | - Richard E Carson
- Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
| | - Irina Esterlis
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, USA
| |
Collapse
|
16
|
Kim JH, Marton J, Ametamey SM, Cumming P. A Review of Molecular Imaging of Glutamate Receptors. Molecules 2020; 25:molecules25204749. [PMID: 33081223 PMCID: PMC7587586 DOI: 10.3390/molecules25204749] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 10/13/2020] [Accepted: 10/14/2020] [Indexed: 12/22/2022] Open
Abstract
Molecular imaging with positron emission tomography (PET) and single photon emission computed tomography (SPECT) is a well-established and important in vivo technique to evaluate fundamental biological processes and unravel the role of neurotransmitter receptors in various neuropsychiatric disorders. Specific ligands are available for PET/SPECT studies of dopamine, serotonin, and opiate receptors, but corresponding development of radiotracers for receptors of glutamate, the main excitatory neurotransmitter in mammalian brain, has lagged behind. This state of affairs has persisted despite the central importance of glutamate neurotransmission in brain physiology and in disorders such as stroke, epilepsy, schizophrenia, and neurodegenerative diseases. Recent years have seen extensive efforts to develop useful ligands for molecular imaging of subtypes of the ionotropic (N-methyl-D-aspartate (NMDA), kainate, and AMPA/quisqualate receptors) and metabotropic glutamate receptors (types I, II, and III mGluRs). We now review the state of development of radioligands for glutamate receptor imaging, placing main emphasis on the suitability of available ligands for reliable in vivo applications. We give a brief account of the radiosynthetic approach for selected molecules. In general, with the exception of ligands for the GluN2B subunit of NMDA receptors, there has been little success in developing radiotracers for imaging ionotropic glutamate receptors; failure of ligands for the PCP/MK801 binding site in vivo doubtless relates their dependence on the open, unblocked state of the ion channel. Many AMPA and kainite receptor ligands with good binding properties in vitro have failed to give measurable specific binding in the living brain. This may reflect the challenge of developing brain-penetrating ligands for amino acid receptors, compounded by conformational differences in vivo. The situation is better with respect to mGluR imaging, particularly for the mGluR5 subtype. Several successful PET ligands serve for investigations of mGluRs in conditions such as schizophrenia, depression, substance abuse and aging. Considering the centrality and diversity of glutamatergic signaling in brain function, we have relatively few selective and sensitive tools for molecular imaging of ionotropic and metabotropic glutamate receptors. Further radiopharmaceutical research targeting specific subtypes and subunits of the glutamate receptors may yet open up new investigational vistas with broad applications in basic and clinical research.
Collapse
Affiliation(s)
- Jong-Hoon Kim
- Neuroscience Research Institute, Gachon University, Incheon 21565, Korea
- Gachon Advanced Institute for Health Science and Technology, Graduate School, Incheon 21565, Korea
- Department of Psychiatry, Gil Medical Center, Gachon University College of Medicine, Gachon University, Incheon 21565, Korea
- Correspondence: (J.-H.K.); (P.C.); Tel.: +41-31-664-0498 (P.C.); Fax: +41-31-632-7663 (P.C.)
| | - János Marton
- ABX Advanced Biochemical Compounds, Biomedizinische Forschungsreagenzien GmbH, Heinrich-Glaeser-Strasse 10-14, D-1454 Radeberg, Germany;
| | - Simon Mensah Ametamey
- Centre for Radiopharmaceutical Sciences ETH-PSI-USZ, Institute of Pharmaceutical Sciences ETH, Vladimir-Prelog-Weg 4, CH-8093 Zürich, Switzerland;
| | - Paul Cumming
- Department of Nuclear Medicine, University of Bern, Inselspital, Freiburgstrasse 18, CH-3010 Bern, Switzerland
- School of Psychology and Counselling, Queensland University of Technology, Brisbane QLD 4059, Australia
- Correspondence: (J.-H.K.); (P.C.); Tel.: +41-31-664-0498 (P.C.); Fax: +41-31-632-7663 (P.C.)
| |
Collapse
|
17
|
Drăgoi AM, Pecie LG, Patrichi BE, Ladea M. Morphopathological changes in obsessive-compulsive disorder. ROMANIAN JOURNAL OF MORPHOLOGY AND EMBRYOLOGY 2020; 61:51-60. [PMID: 32747895 PMCID: PMC7728136 DOI: 10.47162/rjme.61.1.06] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
The pathophysiology of the obsessive-compulsive disorder (OCD) has been studied for many years using several structural magnetic resonance imaging, discovering that the anomalies of function and structure of the brain are widespread, they involve different areas, structures and circuits with a complex interconnectivity. More than that, these anomalies cover all the life of a patient, from early childhood, due to variations of developmental stages until adult life. The research is highly important also because OCD has a major hereditary factor, with the phenotype variance between 27–47% due to hereditary factors. Under this paper, that follows last 10 years studies in this area, we will find some relevant findings consisting on neuroanatomic changes, the morphology findings of striatum, globus pallidus and thalamus, the blood flow circuit changes in various regions of the brain, brain connectivity and various correlations of them. Not to forget that OCD must be understand as an emotional disorder but in the same time as a cognitive disorder too. This approach highlights the abnormalities that have been found in brain regions involved in the cognitive and emotional behavior, as for example: extended temporal, parietal, and occipital regions, anterior cingulate, frontal gyrus, amygdala.
Collapse
Affiliation(s)
- Ana Miruna Drăgoi
- Department of Psychiatry, Prof. Dr. Alexandru Obregia Clinical Hospital for Psychiatry, Bucharest, Romania;
| | | | | | | |
Collapse
|
18
|
Progression of obsessive compulsive disorder-like grooming in Sapap3 knockout mice: A longitudinal [ 11C]ABP688 PET study. Neuropharmacology 2020; 177:108160. [PMID: 32454126 DOI: 10.1016/j.neuropharm.2020.108160] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 03/18/2020] [Accepted: 05/21/2020] [Indexed: 01/23/2023]
Abstract
We aimed to evaluate [3-(6-methyl-pyridin-2-ylethynyl)-cyclohex-2-enone-0-11C-methyloxime] ([11C]ABP688) small animal positron emission tomography (μPET) as a biomarker to visualize possible longitudinal changes in metabotropic glutamate receptor 5 (mGluR5) availability in the brain of SAP90/PSD-95 associated protein 3 (Sapap3) knockout (ko) mice, showing obsessive compulsive disorder (OCD)-like behavior. METHODS Alongside the assessment of grooming, we performed [11C]ABP688 μPET/CT imaging in wildtype (wt; n=10) and ko (n=11) mice both at 3 and 9 months. Using the simplified reference tissue method (SRTM), the nondisplaceable binding potential (BPND) was calculated representing the in vivo availability of the metabotropic glutamate receptor 5 (mGluR5) in the brain with the cerebellum as a reference region. Longitudinal voxel-based statistical parametric mapping (SPM) was performed on BPND images. Results were verified using [11C]ABP688 ex vivo autoradiography, [3H]ABP688 in vitro autoradiography, and mGluR5 immunohistochemistry. RESULTS Cross-sectional comparisons revealed significantly increased grooming parameters in ko animals, at both time points. A significant longitudinal increase in % grooming duration (+268.25%; p<0.05) reflected aggravation of this behavior in ko mice. [11C]ABP688 μPET revealed significantly lower mGluR5 availability in the cortex, striatum, hippocampus, and amygdala of ko mice at both ages. A significant longitudinal BPND decline was present for ko mice (p<0.01: cortex -17.14%, striatum -19.82%, amygdala -23.57%; p<0.05: hippocampus -15.53%), which was confirmed by SPM (p<0.01). CONCLUSION Sapap3 ko mice show a decline in mGluR5 availability in OCD relevant brain regions parallel to the worsening of OCD-like behavior. This demonstrates a potential role for [11C]ABP688 PET as a biomarker to monitor disease progression in vivo.
Collapse
|
19
|
Abstract
Bulimia nervosa (BN) shares central features with substance-related and addictive disorders. The metabotropic glutamate receptor subtype 5 (mGlu5) plays an important role in addiction. Based on similarities between binge eating and substance-related and addictive disorders, we investigated mGlu5 in vivo in 15 female subjects with BN and 15 matched controls. We measured mGlu5 distribution volume ratio (DVR) with positron emission tomography (PET) using [11 C]ABP688. In BN mGlu5 DVR was higher in the anterior cingulate cortex (ACC), subgenual prefrontal cortex, and straight gyrus (p < 0.05). In BN, higher mGlu5 DVR in various brain regions, including ACC, pallidum, putamen, and caudate, positively correlated with "maturity fears" as assessed using the Eating Disorder Inventory-2 (p < 0.05). In BN and controls, smokers had globally decreased mGlu5 DVR. We present the first evidence for increased mGlu5 DVR in BN. Our findings suggest that pharmacological agents inhibiting mGlu5 might have a therapeutic potential in BN.
Collapse
|
20
|
Régio Brambilla C, Veselinović T, Rajkumar R, Mauler J, Orth L, Ruch A, Ramkiran S, Heekeren K, Kawohl W, Wyss C, Kops ER, Scheins J, Tellmann L, Boers F, Neumaier B, Ermert J, Herzog H, Langen K, Jon Shah N, Lerche C, Neuner I. mGluR5 receptor availability is associated with lower levels of negative symptoms and better cognition in male patients with chronic schizophrenia. Hum Brain Mapp 2020; 41:2762-2781. [PMID: 32150317 PMCID: PMC7294054 DOI: 10.1002/hbm.24976] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 02/25/2020] [Accepted: 02/25/2020] [Indexed: 12/29/2022] Open
Abstract
Consistent findings postulate disturbed glutamatergic function (more specifically a hypofunction of the ionotropic NMDA receptors) as an important pathophysiologic mechanism in schizophrenia. However, the role of the metabotropic glutamatergic receptors type 5 (mGluR5) in this disease remains unclear. In this study, we investigated their significance (using [11C]ABP688) for psychopathology and cognition in male patients with chronic schizophrenia and healthy controls. In the patient group, lower mGluR5 binding potential (BPND) values in the left temporal cortex and caudate were associated with higher general symptom levels (negative and depressive symptoms), lower levels of global functioning and worse cognitive performance. At the same time, in both groups, mGluR5 BPND were significantly lower in smokers (F[27,1] = 15.500; p = .001), but without significant differences between the groups. Our findings provide support for the concept that the impaired function of mGluR5 underlies the symptoms of schizophrenia. They further supply a new perspective on the complex relationship between tobacco addiction and schizophrenia by identifying glutamatergic neurotransmission—in particularly mGluR5—as a possible connection to a shared vulnerability.
Collapse
Affiliation(s)
- Cláudia Régio Brambilla
- INM‐4, Forschungszentrum Jülich GmbH, Wilhelm‐Johnen‐StraßeInstitute of Neuroscience and MedicineJülichGermany
- Department of Psychiatry, Psychotherapy and PsychosomaticsRWTH Aachen UniversityAachenGermany
| | - Tanja Veselinović
- Department of Psychiatry, Psychotherapy and PsychosomaticsRWTH Aachen UniversityAachenGermany
| | - Ravichandran Rajkumar
- INM‐4, Forschungszentrum Jülich GmbH, Wilhelm‐Johnen‐StraßeInstitute of Neuroscience and MedicineJülichGermany
- Department of Psychiatry, Psychotherapy and PsychosomaticsRWTH Aachen UniversityAachenGermany
- JARA – BRAIN – Translational MedicineAachenGermany
| | - Jörg Mauler
- INM‐4, Forschungszentrum Jülich GmbH, Wilhelm‐Johnen‐StraßeInstitute of Neuroscience and MedicineJülichGermany
| | - Linda Orth
- INM‐4, Forschungszentrum Jülich GmbH, Wilhelm‐Johnen‐StraßeInstitute of Neuroscience and MedicineJülichGermany
- Department of Psychiatry, Psychotherapy and PsychosomaticsRWTH Aachen UniversityAachenGermany
| | - Andrej Ruch
- Department of Psychiatry, Psychotherapy and PsychosomaticsRWTH Aachen UniversityAachenGermany
| | - Shukti Ramkiran
- INM‐4, Forschungszentrum Jülich GmbH, Wilhelm‐Johnen‐StraßeInstitute of Neuroscience and MedicineJülichGermany
- Department of Psychiatry, Psychotherapy and PsychosomaticsRWTH Aachen UniversityAachenGermany
| | - Karsten Heekeren
- Department of Psychiatry, Psychotherapy and PsychosomaticsUniversity Hospital of PsychiatryZürichSwitzerland
| | - Wolfram Kawohl
- Department of Psychiatry, Psychotherapy and PsychosomaticsUniversity Hospital of PsychiatryZürichSwitzerland
| | - Christine Wyss
- Department of Psychiatry, Psychotherapy and PsychosomaticsUniversity Hospital of PsychiatryZürichSwitzerland
| | - Elena Rota Kops
- INM‐4, Forschungszentrum Jülich GmbH, Wilhelm‐Johnen‐StraßeInstitute of Neuroscience and MedicineJülichGermany
| | - Jürgen Scheins
- INM‐4, Forschungszentrum Jülich GmbH, Wilhelm‐Johnen‐StraßeInstitute of Neuroscience and MedicineJülichGermany
| | - Lutz Tellmann
- INM‐4, Forschungszentrum Jülich GmbH, Wilhelm‐Johnen‐StraßeInstitute of Neuroscience and MedicineJülichGermany
| | - Frank Boers
- INM‐4, Forschungszentrum Jülich GmbH, Wilhelm‐Johnen‐StraßeInstitute of Neuroscience and MedicineJülichGermany
| | - Bernd Neumaier
- INM‐5, Forschungszentrum Jülich GmbH, Wilhelm‐Johnen‐StraßeInstitute of Neuroscience and MedicineJülichGermany
| | - Johannes Ermert
- INM‐5, Forschungszentrum Jülich GmbH, Wilhelm‐Johnen‐StraßeInstitute of Neuroscience and MedicineJülichGermany
| | - Hans Herzog
- INM‐4, Forschungszentrum Jülich GmbH, Wilhelm‐Johnen‐StraßeInstitute of Neuroscience and MedicineJülichGermany
| | - Karl‐Josef Langen
- INM‐4, Forschungszentrum Jülich GmbH, Wilhelm‐Johnen‐StraßeInstitute of Neuroscience and MedicineJülichGermany
- JARA – BRAIN – Translational MedicineAachenGermany
- Department of Nuclear MedicineRWTH Aachen UniversityAachenGermany
| | - N. Jon Shah
- INM‐4, Forschungszentrum Jülich GmbH, Wilhelm‐Johnen‐StraßeInstitute of Neuroscience and MedicineJülichGermany
- JARA – BRAIN – Translational MedicineAachenGermany
- INM‐11, Forschungszentrum Jülich GmbH, Wilhelm‐Johnen‐StraßeInstitute of Neuroscience and MedicineJülichGermany
- Department of NeurologyRWTH Aachen UniversityAachenGermany
| | - Christoph Lerche
- INM‐4, Forschungszentrum Jülich GmbH, Wilhelm‐Johnen‐StraßeInstitute of Neuroscience and MedicineJülichGermany
| | - Irene Neuner
- INM‐4, Forschungszentrum Jülich GmbH, Wilhelm‐Johnen‐StraßeInstitute of Neuroscience and MedicineJülichGermany
- Department of Psychiatry, Psychotherapy and PsychosomaticsRWTH Aachen UniversityAachenGermany
- JARA – BRAIN – Translational MedicineAachenGermany
| |
Collapse
|
21
|
Servaes S, Glorie D, Stroobants S, Staelens S. Neuroreceptor kinetics in rats repeatedly exposed to quinpirole as a model for OCD. PLoS One 2019; 14:e0213313. [PMID: 30845202 PMCID: PMC6405120 DOI: 10.1371/journal.pone.0213313] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Accepted: 02/19/2019] [Indexed: 11/24/2022] Open
Abstract
Background Obsessive-compulsive disorder (OCD) is a chronic, incapacitating, early onset psychiatric disorder that is characterized by obsessions and compulsions originating from a disturbance in the cortico-striato-thalamico-cortical circuit. We implemented the preclinical quinpirole (QP) rat model for compulsive checking in OCD to analyse the behaviour and visualize the D2R, mGluR5 and GLT1 density in order to contribute to the understanding of the neuroreceptor kinetics. Methods Animals (n = 14) were exposed to either saline (1 mL/kg) or QP (dopamine D2-agonist, 0.5 mg/kg) twice-weekly during 7 weeks. After each injection animals were placed on an open field test. After model setup, animals were placed in a behavioural cage equipped with tracking software and hardware in order to analyse the behaviour. Subsequently, sagittal slides were made of the CP in the right hemisphere and a staining was done with the D2R, mGluR5 and GLT-1 antibody to visualize the corresponding receptor. Results The QP animals displayed a strong increase in travelled distance (+596.70%) and in the number of homebase visits (+1222.90%) compared to the control animals. After chronic exposure to QP, animals had a significantly (p < 0.05) higher percentage of D2R density in the CP (7.92% ± 0.48%) versus 6.66% ± 0.28% in animals treated with saline. There were no differences for mGluR5 and GLT1 receptor density. Conclusions Chronic exposure to QP leads to hyperlocomotion and an increase in D2R density. Furthermore, as mGluR5 and GLT1 density did not seem to be directly affected, decreased levels of glutamate might have influenced the binding potential in earlier reports.
Collapse
Affiliation(s)
- Stijn Servaes
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Wilrijk, Antwerp, Belgium
| | - Dorien Glorie
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Wilrijk, Antwerp, Belgium
| | - Sigrid Stroobants
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Wilrijk, Antwerp, Belgium
- Department of Nuclear Medicine, University Hospital Antwerp, Edegem, Antwerp, Belgium
| | - Steven Staelens
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Wilrijk, Antwerp, Belgium
- * E-mail:
| |
Collapse
|
22
|
Xu Y, Li Z. Imaging metabotropic glutamate receptor system: Application of positron emission tomography technology in drug development. Med Res Rev 2019; 39:1892-1922. [DOI: 10.1002/med.21566] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 01/18/2019] [Accepted: 01/24/2019] [Indexed: 12/17/2022]
Affiliation(s)
- Youwen Xu
- Independent Consultant and Contractor, Radiopharmaceutical Development, Validation and Bio-Application; Philadelphia Pennsylvania
| | - Zizhong Li
- Pharmaceutical Research and Development, SOFIE Biosciences; Somerset New Jersey
| |
Collapse
|
23
|
Kosten L, Verhaeghe J, Wyffels L, Stroobants S, Staelens S. Acute Ketamine Infusion in Rat Does Not Affect In Vivo [ 11C]ABP688 Binding to Metabotropic Glutamate Receptor Subtype 5. Mol Imaging 2019; 17:1536012118788636. [PMID: 30213221 PMCID: PMC6144515 DOI: 10.1177/1536012118788636] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Detecting changes in metabotropic glutamate receptor 5 (mGluR5) availability through molecular imaging with the positron emission tomography (PET) tracer [11C]ABP688 is valuable for studying dysfunctional glutamate transmission associated with neuropsychiatric disorders. Using an infusion protocol in rats, we visualized the acute effect of subanesthetic doses of ketamine on mGluR5 in rat brain. Ketamine is an N-methyl-D-aspartate (NMDA) receptor antagonist known to increase glutamate release. Imaging was performed with a high-affinity PET ligand [11C]ABP688, a negative allosteric modulator of mGluR5. Binding did not change significantly from baseline to ketamine in any region, thereby confirming previous literature with other NMDA receptor antagonists in rodents. Hence, in rats, we could not reproduce the findings in a human setup showing significant decreases in the [11C]ABP688 binding after a ketamine bolus followed by ketamine infusion. Species differences may have contributed to the different findings in the present study of rats. In conclusion, we could not confirm in rats that endogenous glutamate increases by ketamine infusion are reflected in [11C]ABP688 binding decreases as was previously shown for humans.
Collapse
Affiliation(s)
- Lauren Kosten
- 1 Molecular Imaging Center Antwerp, University of Antwerp, Antwerp, Belgium
| | - Jeroen Verhaeghe
- 1 Molecular Imaging Center Antwerp, University of Antwerp, Antwerp, Belgium
| | - Leonie Wyffels
- 1 Molecular Imaging Center Antwerp, University of Antwerp, Antwerp, Belgium.,2 Department of Nuclear Medicine, University Hospital Antwerp, Antwerp, Belgium
| | - Sigrid Stroobants
- 1 Molecular Imaging Center Antwerp, University of Antwerp, Antwerp, Belgium.,2 Department of Nuclear Medicine, University Hospital Antwerp, Antwerp, Belgium
| | - Steven Staelens
- 1 Molecular Imaging Center Antwerp, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
24
|
O'Gorman Tuura R, Warnock G, Ametamey S, Treyer V, Noeske R, Buck A, Sommerauer M. Imaging glutamate redistribution after acute N-acetylcysteine administration: A simultaneous PET/MR study. Neuroimage 2018; 184:826-833. [PMID: 30296554 DOI: 10.1016/j.neuroimage.2018.10.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 08/31/2018] [Accepted: 10/04/2018] [Indexed: 12/25/2022] Open
Abstract
Glutamate is the most abundant excitatory neurotransmitter in the human brain, but in vivo imaging of acute fluctuations in glutamatergic levels has not been well established. The purpose of this study was to examine acute changes in glutamate after stimulation with N-acetylcysteine (NAC) using a simultaneous positron emission tomography/magnetic resonance spectroscopy (PET/MRS) approach. Ten healthy adult males were examined in two scanning sessions, and 5g NAC was administered 1 h prior to one of the scan sessions. Simultaneous PET/MR data were acquired using an integrated 3T PET/MR scanner. Glutamate (Glu), glutamine (Gln), and glutamate + glutamine (Glx) levels were assessed from MRS data collected from the basal ganglia with PRESS and from the left prefrontal cortex with PRESS and MEGAPRESS, and mGluR5 binding (BPND) was assessed from PET data collected with [18F]PSS232. NAC administration was associated with a significant reduction in Glx and Gln in the basal ganglia spectra, and in Glx in the frontal MEGAPRESS spectra (p < 0.05); no differences in [18F]PSS232 BPND were observed with NAC, although a correlation between pre-/post-treatment Glx and baseline BPnd was found. The MRS-visible Glx signal is sensitive to acute fluctuations in glutamate. The change in Glx was mostly driven by a change in Gln, lending weight to the notion that Gln can provide a proxy marker for neurotransmitter/synaptic glutamate. [18F]PSS232 binding is not sensitive to acute glutamate shifts independently, but was associated with the extent of glutamate liberation upon NAC stimulation.
Collapse
Affiliation(s)
- Ruth O'Gorman Tuura
- Center for MR Research, University Children's Hospital, Steinwiesstrasse 75, CH-8032, Zürich, Switzerland.
| | - Geoff Warnock
- Department of Nuclear Medicine, University Hospital Zürich, Rämistrasse 100, CH-8091, Zürich, Switzerland
| | - Simon Ametamey
- Center for Radiopharmaceutical Sciences, Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH), Vladimir-Prelog-Weg 4 4, CH-8093, Zürich, Switzerland
| | - Valerie Treyer
- Department of Nuclear Medicine, University Hospital Zürich, Rämistrasse 100, CH-8091, Zürich, Switzerland
| | | | - Alfred Buck
- Department of Nuclear Medicine, University Hospital Zürich, Rämistrasse 100, CH-8091, Zürich, Switzerland
| | - Michael Sommerauer
- Department of Nuclear Medicine, University Hospital Zürich, Rämistrasse 100, CH-8091, Zürich, Switzerland; Department of Neurology, University Hospital Cologne, Kerpener Str. 62, 50937, Cologne, Germany
| |
Collapse
|
25
|
Esterlis I, Holmes SE, Sharma P, Krystal JH, DeLorenzo C. Metabotropic Glutamatergic Receptor 5 and Stress Disorders: Knowledge Gained From Receptor Imaging Studies. Biol Psychiatry 2018; 84:95-105. [PMID: 29100629 PMCID: PMC5858955 DOI: 10.1016/j.biopsych.2017.08.025] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 08/24/2017] [Accepted: 08/28/2017] [Indexed: 12/28/2022]
Abstract
The metabotropic glutamatergic receptor subtype 5 (mGluR5) may represent a promising therapeutic target for stress-related psychiatric disorders. Here, we describe mGluR5 findings in stress disorders, particularly major depressive disorder (MDD), highlighting insights from positron emission tomography studies. Positron emission tomography studies report either no differences or lower mGluR5 in MDD, potentially reflecting MDD heterogeneity. Unlike the rapidly acting glutamatergic agent ketamine, mGluR5-specific modulation has not yet shown antidepressant efficacy in MDD and bipolar disorder. Although we recently showed that ketamine may work, in part, through significant mGluR5 modulation, the specific role of mGluR5 downregulation in ketamine's antidepressant response is unclear. In contrast to MDD, there has been much less investigation of mGluR5 in bipolar disorder, yet initial studies indicate that mGluR5-specific treatments may aid in both depressed and manic mood states. The direction of modulation needed may be state dependent, however, limiting clinical feasibility. There has been relatively little study of posttraumatic stress disorder or obsessive-compulsive disorder to date, although there is evidence for the upregulation of mGluR5 in these disorders. However, while antagonism of mGluR5 may reduce fear conditioning, it may also reduce fear extinction. Therefore, studies are needed to determine the role mGluR5 modulation might play in the treatment of these conditions. Further challenges in modulating this prevalent neurotransmitter system include potential induction of significant side effects. As such, more research is needed to identify level and type (positive/negative allosteric modulation or full antagonism) of mGluR5 modulation required to translate existing knowledge into improved therapies.
Collapse
Affiliation(s)
- Irina Esterlis
- Department of Psychiatry, Yale University, New Haven, Connecticut; US Department of Veterans Affairs National Center for Posttraumatic Stress Disorder, Clinical Neurosciences Division, Veteran's Affairs Connecticut Healthcare System, West Haven, Connecticut.
| | | | - Priya Sharma
- Department of Psychiatry, Schulich School of Medicine and Dentistry; Western University- London, Ontario, Canada; London Health Sciences Centre- Victoria Hospital
| | - John H. Krystal
- Yale University, Department of Psychiatry,Yale University, Department of Neuroscience,U.S. Department of Veterans Affairs National Center for Posttraumatic Stress Disorder, Clinical Neurosciences Division, VA Connecticut Healthcare System
| | - Christine DeLorenzo
- Stony Brook University, Department of Psychiatry,Stony Brook University, Department of Biomedical Engineering
| |
Collapse
|
26
|
Ferraguti F. Metabotropic glutamate receptors as targets for novel anxiolytics. Curr Opin Pharmacol 2018; 38:37-42. [PMID: 29494817 DOI: 10.1016/j.coph.2018.02.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 02/12/2018] [Indexed: 12/12/2022]
Abstract
Anxiety disorders are highly prevalent psychiatric illnesses posing an important social and economic burden. Their current pharmacotherapy shows short term efficacy, though nearly one third of patients do not achieve sustained remission. There is, therefore, a strong medical need for new therapeutic agents acting through novel mechanisms of action. Considerable work has focused on metabotropic glutamate (mGlu) receptors as potential targets for novel anxiolytics. Ligands acting at mGlu receptors showed promising results in preclinical studies, whereas their efficacy was dubious in clinical trials. Recent preclinical and clinical studies have opened new prospects for targeting mGlu receptors to treat anxiety disorders. This review provides an outlook on these progresses.
Collapse
Affiliation(s)
- Francesco Ferraguti
- Department of Pharmacology, Medical University of Innsbruck, Peter Mayr Strasse 1A, 6020 Innsbruck, Austria.
| |
Collapse
|
27
|
Akkus F, Mihov Y, Treyer V, Ametamey SM, Johayem A, Senn S, Rösner S, Buck A, Hasler G. Metabotropic glutamate receptor 5 binding in male patients with alcohol use disorder. Transl Psychiatry 2018; 8:17. [PMID: 29317611 PMCID: PMC5802584 DOI: 10.1038/s41398-017-0066-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 09/13/2017] [Accepted: 10/26/2017] [Indexed: 01/30/2023] Open
Abstract
Glutamate signaling plays a major role in addiction. Preclinical research strongly suggests an implication of G-protein-coupled metabotropic glutamate receptor subtype 5 (mGluR5) in nicotine addiction and alcohol use disorder. In humans, smoking is related to a global reduction in mGluR5 availability. In the present study, we investigated mGluR5 in vivo in patients with alcohol use disorder without the confounding effects of smoking. A total of 14 male subjects with alcohol use disorder and at least a 25-day abstinence and 14 matched male non-smoking healthy controls were included in the study. We employed positron emission tomography (PET) with the mGluR5-specific radiotracer [11C]ABP688, using a bolus/infusion protocol. We found increased mGluR5 DVR in several regions within the temporal lobe in patients, as compared to controls. The largest between-group difference was in the amygdala. There was a marked positive relation between mGluR5 DVR in the anterior cingulate and mGluR5 DVR in the orbitofrontal cortex in patients, but not in controls. In patients, lower temptation to drink was related to higher amygdala mGluR5 DVR. We did not find altered mGluR5 DVR in the basal ganglia of subjects recovering from alcohol use disorder. In conclusion, our study provides clinical evidence for altered mGluR5 signaling in the amygdala in alcohol use disorder. This alteration was associated with the temptation to drink. In addition, this study suggests abnormal mGluR5 signaling in a network underlying reward-related behavioral flexibility. These findings strengthen the case for pharmacological agents acting on mGluR5 as promising candidates for the treatment of alcohol use disorder.
Collapse
Affiliation(s)
- Funda Akkus
- Division of Molecular Psychiatry, Translational Research Center, University Hospital of Psychiatry, University of Bern, 3000, Bern 60, Switzerland
| | - Yoan Mihov
- Division of Molecular Psychiatry, Translational Research Center, University Hospital of Psychiatry, University of Bern, 3000, Bern 60, Switzerland
| | - Valerie Treyer
- PET Center, Division of Nuclear Medicine, University Hospital, 8091, Zurich, Switzerland
| | - Simon M Ametamey
- Center for Radiopharmaceutical Science of ETH, PSI, and USZ, Department of Chemistry and Applied Biosciences of ETH, 8093, Zurich, Switzerland
| | - Anass Johayem
- PET Center, Division of Nuclear Medicine, University Hospital, 8091, Zurich, Switzerland
| | - Smeralda Senn
- Forel Clinic, Addiction Treatment Center, 8548, Ellikon an der Thur, Switzerland
| | - Susanne Rösner
- Forel Clinic, Addiction Treatment Center, 8548, Ellikon an der Thur, Switzerland
| | - Alfred Buck
- PET Center, Division of Nuclear Medicine, University Hospital, 8091, Zurich, Switzerland
| | - Gregor Hasler
- Division of Molecular Psychiatry, Translational Research Center, University Hospital of Psychiatry, University of Bern, 3000, Bern 60, Switzerland.
| |
Collapse
|
28
|
Neuronal Glutamate Transporters Control Dopaminergic Signaling and Compulsive Behaviors. J Neurosci 2017; 38:937-961. [PMID: 29229708 DOI: 10.1523/jneurosci.1906-17.2017] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 11/24/2017] [Accepted: 12/01/2017] [Indexed: 12/21/2022] Open
Abstract
There is an ongoing debate on the contribution of the neuronal glutamate transporter EAAC1 to the onset of compulsive behaviors. Here, we used behavioral, electrophysiological, molecular, and viral approaches in male and female mice to identify the molecular and cellular mechanisms by which EAAC1 controls the execution of repeated motor behaviors. Our findings show that, in the striatum, a brain region implicated with movement execution, EAAC1 limits group I metabotropic glutamate receptor (mGluRI) activation, facilitates D1 dopamine receptor (D1R) expression, and ensures long-term synaptic plasticity. Blocking mGluRI in slices from mice lacking EAAC1 restores D1R expression and synaptic plasticity. Conversely, activation of intracellular signaling pathways coupled to mGluRI in D1R-containing striatal neurons of mice expressing EAAC1 leads to reduced D1R protein level and increased stereotyped movement execution. These findings identify new molecular mechanisms by which EAAC1 can shape glutamatergic and dopaminergic signals and control repeated movement execution.SIGNIFICANCE STATEMENT Genetic studies implicate Slc1a1, a gene encoding the neuronal glutamate transporter EAAC1, with obsessive-compulsive disorder (OCD). EAAC1 is abundantly expressed in the striatum, a brain region that is hyperactive in OCD. What remains unknown is how EAAC1 shapes synaptic function in the striatum. Our findings show that EAAC1 limits activation of metabotropic glutamate receptors (mGluRIs) in the striatum and, by doing so, promotes D1 dopamine receptor (D1R) expression. Targeted activation of signaling cascades coupled to mGluRIs in mice expressing EAAC1 reduces D1R expression and triggers repeated motor behaviors. These findings provide new information on the molecular basis of OCD and suggest new avenues for its treatment.
Collapse
|
29
|
Imaging the glutamate receptor subtypes-Much achieved, and still much to do. DRUG DISCOVERY TODAY. TECHNOLOGIES 2017; 25:27-36. [PMID: 29233264 DOI: 10.1016/j.ddtec.2017.10.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 10/17/2017] [Accepted: 10/24/2017] [Indexed: 11/20/2022]
Abstract
Functional imaging of glutamate receptors using PET imaging modality can be used to study numerous CNS disorders and also to select appropriate doses of clinically relevant glutamate-receptor-targeting candidate drugs. Great strides have been made in developing PET imaging probes for the non-invasive detection of glutamate receptors in the brain. This review highlights recent progress made towards the development of glutamatergic PET imaging agents. Focus is placed on PET imaging probes that have been labelled with either carbon-11 or fluorine-18.
Collapse
|
30
|
Attwells S, Setiawan E, Wilson AA, Rusjan PM, Mizrahi R, Miler L, Xu C, Richter MA, Kahn A, Kish SJ, Houle S, Ravindran L, Meyer JH. Inflammation in the Neurocircuitry of Obsessive-Compulsive Disorder. JAMA Psychiatry 2017; 74. [PMID: 28636705 PMCID: PMC5710556 DOI: 10.1001/jamapsychiatry.2017.1567] [Citation(s) in RCA: 122] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
IMPORTANCE For a small percentage of obsessive-compulsive disorder (OCD) cases exhibiting additional neuropsychiatric symptoms, it was proposed that neuroinflammation occurs in the basal ganglia as an autoimmune response to infections. However, it is possible that elevated neuroinflammation, inducible by a diverse range of mechanisms, is important throughout the cortico-striato-thalamo-cortical circuit of OCD. Identifying brain inflammation is possible with the recent advance in positron emission tomography (PET) radioligands that bind to the translocator protein (TSPO). Translocator protein density increases when microglia are activated during neuroinflammation and the TSPO distribution volume (VT) is an index of TSPO density. OBJECTIVE To determine whether TSPO VT is elevated in the dorsal caudate, orbitofrontal cortex, thalamus, ventral striatum, dorsal putamen, and anterior cingulate cortex in OCD. DESIGN, SETTING, AND PARTICIPANTS This case-control study was conducted at a tertiary care psychiatric hospital from May 1, 2010, to November 30, 2016. Participants with OCD (n = 20) and age-matched healthy control individuals (n = 20) underwent a fluorine F 18-labeled N-(2-(2-fluoroethoxy)benzyl)-N-(4-phenoxypyridin-3-yl)acetamide PET scan. It is a high-quality second-generation TSPO-binding PET radiotracer. All participants were drug and medication free, nonsmoking, and otherwise healthy. MAIN OUTCOMES AND MEASURES The TSPO VT was measured in the dorsal caudate, orbitofrontal cortex, thalamus, ventral striatum, dorsal putamen, and anterior cingulate cortex. Compulsions were assessed with the Yale-Brown Obsessive Compulsive Scale. RESULTS In the OCD and healthy groups, the mean (SD) ages were 27.4 (7.1) years and 27.6 (6.6) years, respectively, and 11 (55%) and 8 (40%) were women, respectively. In OCD, TSPO VT was significantly elevated in these brain regions (mean, 32%; range, 31%-36% except anterior cingulate cortex, 24%; analysis of variance, effect of diagnosis: P < .001 to P = .004). Slightly lower elevations in TSPO VT (22%-29%) were present in other gray matter regions. The Yale-Brown Obsessive Compulsive Scale measure of distress associated with preventing compulsive behaviors significantly correlated with TSPO VT in the orbitofrontal cortex (uncorrected Pearson correlation r = 0.62; P = .005). CONCLUSIONS AND RELEVANCE To our knowledge, this is the first study demonstrating inflammation within the neurocircuitry of OCD. The regional distribution of elevated TSPO VT argues that the autoimmune/neuroinflammatory theories of OCD should extend beyond the basal ganglia to include the cortico-striato-thalamo-cortical circuit. Immunomodulatory therapies should be investigated in adult OCD, rather than solely childhood OCD, particularly in cases with prominent distress when preventing compulsions.
Collapse
Affiliation(s)
- Sophia Attwells
- Research Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada,Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Elaine Setiawan
- Research Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Alan A. Wilson
- Research Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada,Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Pablo M. Rusjan
- Research Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Romina Mizrahi
- Research Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada,Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada,Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Laura Miler
- Research Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Cynthia Xu
- Research Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Margaret Anne Richter
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada,Frederick W. Thompson Anxiety Disorders Centre, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Alan Kahn
- Research Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada,Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Stephen J. Kish
- Research Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada,Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada,Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Sylvain Houle
- Research Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada,Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Lakshmi Ravindran
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Jeffrey H. Meyer
- Research Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada,Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada,Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
31
|
Altered metabotropic glutamate receptor 5 markers in PTSD: In vivo and postmortem evidence. Proc Natl Acad Sci U S A 2017; 114:8390-8395. [PMID: 28716937 DOI: 10.1073/pnas.1701749114] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Posttraumatic stress disorder (PTSD) is a prevalent and highly disabling disorder, but there is currently no targeted pharmacological treatment for it. Dysfunction of the glutamate system has been implicated in trauma and stress psychopathology, resulting in a growing interest in modulation of the glutamate system for the treatment of PTSD. Specifically, the metabotropic glutamate receptor 5 (mGluR5) represents a promising treatment target. We used [18F]FPEB, a radioligand that binds to the mGluR5, and positron emission tomography (PET) to quantify in vivo mGluR5 availability in human PTSD vs. healthy control (HCs) subjects. In an independent sample of human postmortem tissue, we investigated expression of proteins that have a functional relationship with mGluR5 and glucocorticoids in PTSD. We observed significantly higher cortical mGluR5 availability in PTSD in vivo and positive correlations between mGluR5 availability and avoidance symptoms. In the postmortem sample, we observed up-regulation of SHANK1, a protein that anchors mGluR5 to the cell surface, as well as decreased expression of FKBP5, implicating aberrant glucocorticoid functioning in PTSD. Results of this study provide insight into molecular mechanisms underlying PTSD and suggest that mGluR5 may be a promising target for mechanism-based treatments aimed at mitigating this disorder.
Collapse
|
32
|
Servaes S, Glorie D, Verhaeghe J, Stroobants S, Staelens S. Preclinical molecular imaging of glutamatergic and dopaminergic neuroreceptor kinetics in obsessive compulsive disorder. Prog Neuropsychopharmacol Biol Psychiatry 2017; 77:90-98. [PMID: 28365375 DOI: 10.1016/j.pnpbp.2017.02.027] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Revised: 02/06/2017] [Accepted: 02/27/2017] [Indexed: 10/19/2022]
Abstract
BACKGROUND Molecular neuroimaging was applied in the quinpirole rat model for compulsive checking in OCD to visualize the D2- and mGluR5-receptor occupancy with Raclopride and ABP-688 microPET/CT. METHODS Animals (n=48) were exposed to either saline (CTRL; 1mL/kg) or quinpirole (QP; dopamine D2-agonist, 0.5mg/kg) in a single injection (RAC and ABP acute groups) or twice-weekly during 7weeks (chronic group). Animals underwent PET/CT after the 1st injection (acute) or before initial exposure and following the 10th injection in week 5 (chronic). For the latter, each injection was paired with an open field test and video tracking. RESULTS The QP animals displayed a strong increase in visiting frequency (checking) in the chronic group (+699.29%) compared to the control animals. Acute administration of the drug caused significant (p<0.01) decreases in D2R occupancy in the CP (-42.03%±4.01%). Chronical exposure resulted in significantly stronger decreases in the CP (-52.29%±3.79%). Furthermore significant increases in mGluR5 occupancy were found in the CP (10.36%±4.09%), anterior cingulate cortex (13.26%±4.01%), amygdala (24.36%±6.86%), entorhinal cortex (18.49%±5.14%) and nucleus accumbens (13.8%±4.87%) of the chronic group, not present after acute exposure. CONCLUSIONS Compared to acute exposure, sensitisation to QP as a model for OCD differs both on a dopaminergic and glutamateric level, indicating involvement of processes such as receptor internalization and changes in extracellular availability of both neurotransmitters.
Collapse
Affiliation(s)
- S Servaes
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Antwerp, Belgium.
| | - D Glorie
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Antwerp, Belgium.
| | - J Verhaeghe
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Antwerp, Belgium.
| | - S Stroobants
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Antwerp, Belgium; Department of Nuclear Medicine, University Hospital Antwerp, Wilrijkstraat 10, 2650, Edegem, Antwerp, Belgium.
| | - S Staelens
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Antwerp, Belgium.
| |
Collapse
|
33
|
Akkus F, Treyer V, Ametamey SM, Johayem A, Buck A, Hasler G. Metabotropic glutamate receptor 5 neuroimaging in schizophrenia. Schizophr Res 2017; 183:95-101. [PMID: 27847228 DOI: 10.1016/j.schres.2016.11.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 10/20/2016] [Accepted: 11/09/2016] [Indexed: 11/29/2022]
Abstract
The metabotropic glutamate receptor 5 (mGluR5) is a promising drug target for the treatment of schizophrenia. In this study, we compared mGluR5 distribution volume ration (DVR) in subjects with schizophrenia and healthy controls. Given our previous findings, we matched samples for gender, age, and smoking status. Binding to mGluR5 was determined using positron emission tomography and [11C]ABP688, which binds to an allosteric site with high selectivity. DVR in the 15 individuals with schizophrenia did not differ from that of the 15 controls. In both groups, smoking was associated with marked global reductions in mGluR5 availability (on average 23.8%). In nonsmoking subjects with schizophrenia, there was a positive correlation between mGluR5 DVR in the medial orbitofrontal cortex and the use of antipsychotic drugs (r=0.9, p=0.019). Because antipsychotic drugs such as clozapine appeared to have indirect effects on mGluR5 signaling, our findings may be clinically relevant. They also provide promising leads for elucidating the high comorbidity between schizophrenia and tobacco addiction. Low mGluR5 DVR in smokers my represent a risk factor for schizophrenia. Alternatively, smoking may counteract the potential upregulation of mGluR5 by antipsychotic drugs.
Collapse
Affiliation(s)
- Funda Akkus
- Psychiatric University Hospital, University of Bern, 3000 Bern 60, Switzerland
| | - Valerie Treyer
- PET Center, Division of Nuclear Medicine, University Hospital, 8091 Zurich, Switzerland
| | - Simon M Ametamey
- Center for Radiopharmaceutical Science of ETH, PSI, and USZ, Department of Chemistry and Applied Biosciences of ETH, 8093 Zurich, Switzerland
| | - Anass Johayem
- PET Center, Division of Nuclear Medicine, University Hospital, 8091 Zurich, Switzerland
| | - Alfred Buck
- PET Center, Division of Nuclear Medicine, University Hospital, 8091 Zurich, Switzerland
| | - Gregor Hasler
- Psychiatric University Hospital, University of Bern, 3000 Bern 60, Switzerland.
| |
Collapse
|
34
|
Hu X, Du M, Chen L, Li L, Zhou M, Zhang L, Liu Q, Lu L, Mreedha K, Huang X, Gong Q. Meta-analytic investigations of common and distinct grey matter alterations in youths and adults with obsessive-compulsive disorder. Neurosci Biobehav Rev 2017; 78:91-103. [PMID: 28442404 DOI: 10.1016/j.neubiorev.2017.04.012] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 04/15/2017] [Accepted: 04/15/2017] [Indexed: 02/05/2023]
Abstract
Obsessive-compulsive disorder (OCD) is a disabling illness with onset generally in childhood. OCD-youths differ from OCD-adults with regard to gender distribution, comorbidity patterns and treatment options. However, little is known about the neural correlate differences underpin those two populations. The current meta-analysis summarizes voxel based morphometry findings to elucidate whether differences of neural correlates exist between these two populations. Both OCD-youths and OCD-adults demonstrated greater striatal volume and smaller prefrontal grey matter volume (GMV). However, smaller GMV in left visual cortex was observed in OCD-youths only, while smaller GMV in anterior cingulate gyrus and greater GMV in cerebellum were demonstrated only in OCD-adults. Meta-regression showed greater GMV in left putamen was most prominent in samples with higher percentages of medicated OCD-adults. Our findings confirmed the most consistent GMV alterations in OCD were in prefrontal-striatal circuitry. Besides, other regions may involve at different developmental stages including deficits of visual cortex in OCD-youths and abnormalities of limbic-cerebellar circuit in OCD-adults. Medication effect may be more pronounced in the striatum, especially the putamen.
Collapse
Affiliation(s)
- Xinyu Hu
- Huaxi MR Research Centre(HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, China
| | - Mingying Du
- Department of Radiology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Lizhou Chen
- Huaxi MR Research Centre(HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, China
| | - Lei Li
- Huaxi MR Research Centre(HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, China
| | - Ming Zhou
- Huaxi MR Research Centre(HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, China
| | - Lianqing Zhang
- Huaxi MR Research Centre(HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, China
| | - Qi Liu
- Huaxi MR Research Centre(HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, China
| | - Lu Lu
- Huaxi MR Research Centre(HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, China
| | - Kunal Mreedha
- Huaxi MR Research Centre(HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, China
| | - Xiaoqi Huang
- Huaxi MR Research Centre(HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, China.
| | - Qiyong Gong
- Huaxi MR Research Centre(HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, China.
| |
Collapse
|
35
|
Rutrick D, Stein DJ, Subramanian G, Smith B, Fava M, Hasler G, Cha JH, Gasparini F, Donchev T, Ocwieja M, Johns D, Gomez-Mancilla B. Mavoglurant Augmentation in OCD Patients Resistant to Selective Serotonin Reuptake Inhibitors: A Proof-of-Concept, Randomized, Placebo-Controlled, Phase 2 Study. Adv Ther 2017; 34:524-541. [PMID: 28044255 DOI: 10.1007/s12325-016-0468-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Indexed: 11/28/2022]
Abstract
INTRODUCTION To determine if mavoglurant (modified release) as an augmentation therapy to selective serotonin reuptake inhibitors (SSRIs) could have beneficial effects reducing Yale-Brown Obsessive Compulsive Scale (Y-BOCS) total score in patients with obsessive-compulsive disorder (OCD) resistant to SSRI treatment. METHODS This was a multicenter, randomized, double-blind, placebo-controlled, parallel-group, phase 2 study. Patients remained on their SSRI treatment and mavoglurant or placebo was added on. Non-smoking men and women aged 18-65 years primarily diagnosed with OCD according to Diagnostic and Statistical Manual of Mental Disorders (4th ed., text rev.; DSM-IV-TR) criteria were randomized (1:1) to mavoglurant or placebo groups. After 50 patients were randomized, an interim analysis was conducted to determine whether the study should be continued. The primary outcome measure was absolute change in Y-BOCS from baseline at week 17. Safety was assessed by recording adverse events (AEs) and serious adverse events (SAEs). RESULTS Interim analysis led to a decision to terminate the study. In total 38 (76.0%) participants completed 17 weeks of treatment and 37 (74.0%) completed the study. There was no significant difference in least squares (LS) mean change from baseline at week 17 in Y-BOCS total score for mavoglurant compared with placebo groups [-6.9 (1.75) vs. -8.0 (1.78), respectively; LS mean difference 1.1; 95% CI -3.9, 6.2; p = 0.671]. The incidence of AEs was higher in the mavoglurant compared with the placebo group (80.8% vs. 70.8%, respectively). CONCLUSION This study of mavoglurant in OCD was terminated because of the lack of efficacy at interim analysis. The study did not support the use of an antagonist of mGluR5 receptors for OCD treatment. TRIAL REGISTRATION The study was registered with ClinicalTrials.gov: NCT01813019. FUNDING This study was sponsored by Novartis Pharma AG, Basel, Switzerland.
Collapse
Affiliation(s)
| | - Dan J Stein
- MRC Unit on Anxiety and Stress Disorders, Department of Psychiatry, Groote Schuur Hospital, University of Cape Town, Cape Town, 7925, South Africa
| | | | - Brian Smith
- Novartis Pharmaceutical Corporation, Cambridge, MA, 02139, USA
| | - Maurizio Fava
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, 02115, USA
| | - Gregor Hasler
- Division of Molecular Psychiatry, Psychiatric University Hospital, University of Bern, Bern 60, 3000, Bern, Switzerland
| | - Jang-Ho Cha
- Novartis Pharmaceutical Corporation, Cambridge, MA, 02139, USA
| | - Fabrizio Gasparini
- Novartis Institutes for Biomedical Research, Novartis Pharma AG, 4056, Basel, Switzerland
| | | | - Magdalena Ocwieja
- Novartis Institutes for Biomedical Research, Novartis Pharma AG, 4056, Basel, Switzerland
| | - Donald Johns
- Novartis Institutes for Biomedical Research, Novartis Pharma AG, 4056, Basel, Switzerland
- Biogen, 300 Binney Street, Cambridge, MA, 02142, USA
| | - Baltazar Gomez-Mancilla
- Novartis Institutes for Biomedical Research, Novartis Pharma AG, 4056, Basel, Switzerland.
- Department Neurology and Neurosurgery, McGill University, Montreal, QC, Canada.
| |
Collapse
|
36
|
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: 0.7] [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
|
37
|
Ade KK, Wan Y, Hamann HC, O’Hare JK, Guo W, Quian A, Kumar S, Bhagat S, Rodriguiz RM, Wetsel WC, Conn PJ, Dzirasa K, Huber KM, Calakos N. Increased Metabotropic Glutamate Receptor 5 Signaling Underlies Obsessive-Compulsive Disorder-like Behavioral and Striatal Circuit Abnormalities in Mice. Biol Psychiatry 2016; 80:522-33. [PMID: 27436084 PMCID: PMC5536332 DOI: 10.1016/j.biopsych.2016.04.023] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 04/29/2016] [Accepted: 04/29/2016] [Indexed: 01/15/2023]
Abstract
BACKGROUND Development of treatments for obsessive-compulsive disorder (OCD) is hampered by a lack of mechanistic understanding about this prevalent neuropsychiatric condition. Although circuit changes such as elevated frontostriatal activity are linked to OCD, the underlying molecular signaling that drives OCD-related behaviors remains largely unknown. Here, we examine the significance of type 5 metabotropic glutamate receptors (mGluR5s) for behavioral and circuit abnormalities relevant to OCD. METHODS Sapap3 knockout (KO) mice treated acutely with an mGluR5 antagonist were evaluated for OCD-relevant phenotypes of self-grooming, anxiety-like behaviors, and increased striatal activity. The role of mGluR5 in the striatal circuit abnormalities of Sapap3 KO mice was further explored using two-photon calcium imaging to monitor striatal output from the direct and indirect pathways. A contribution of constitutive signaling to increased striatal mGluR5 activity in Sapap3 KO mice was investigated using pharmacologic and biochemical approaches. Finally, sufficiency of mGluR5 to drive OCD-like behavior in wild-type mice was tested by potentiating mGluR5 with a positive allosteric modulator. RESULTS Excessive mGluR5 signaling underlies OCD-like behaviors and striatal circuit abnormalities in Sapap3 KO mice. Accordingly, enhancing mGluR5 activity acutely recapitulates these behavioral phenotypes in wild-type mice. In Sapap3 KO mice, elevated mGluR5 signaling is associated with constitutively active receptors and increased and imbalanced striatal output that is acutely corrected by antagonizing striatal mGluR5. CONCLUSIONS These findings demonstrate a causal role for increased mGluR5 signaling in driving striatal output abnormalities and behaviors with relevance to OCD and show the tractability of acute mGluR5 inhibition to remedy circuit and behavioral abnormalities.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Nicole Calakos
- Departments of Neurology, Duke University Medical Center, Durham, North Carolina; Neurobiology, Duke University Medical Center, Durham, North Carolina.
| |
Collapse
|
38
|
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: 42] [Impact Index Per Article: 4.7] [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
|
39
|
Metabotropic glutamate receptor 5 – a promising target in drug development and neuroimaging. Eur J Nucl Med Mol Imaging 2016; 43:1151-70. [DOI: 10.1007/s00259-015-3301-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2015] [Accepted: 12/22/2015] [Indexed: 10/22/2022]
|
40
|
DeLorenzo C, Sovago J, Gardus J, Xu J, Yang J, Behrje R, Kumar JSD, Devanand DP, Pelton GH, Mathis CA, Mason NS, Gomez-Mancilla B, Aizenstein H, Mann JJ, Parsey RV. Characterization of brain mGluR5 binding in a pilot study of late-life major depressive disorder using positron emission tomography and [¹¹C]ABP688. Transl Psychiatry 2015; 5:e693. [PMID: 26645628 PMCID: PMC5068588 DOI: 10.1038/tp.2015.189] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 10/05/2015] [Accepted: 10/26/2015] [Indexed: 01/06/2023] Open
Abstract
The metabotropic glutamate receptor subtype 5 (mGluR5) has been implicated in the pathophysiology of mood and anxiety disorders and is a potential treatment target in major depressive disorder (MDD). This study compared brain mGluR5 binding in elderly patients suffering from MDD with that in elderly healthy volunteers using positron emission tomography (PET) and [(11)C]ABP688. Twenty elderly (mean age: 63.0 ± 6.3) subjects with MDD and twenty-two healthy volunteers in the same age range (mean age: 66.4 ± 7.3) were examined with PET after a single bolus injection of [(11)C]ABP688, with many receiving arterial sampling. PET images were analyzed on a region of interest and a voxel level to compare mGluR5 binding in the brain between the two groups. Differences in [(11)C]ABP688 binding between patients with early- and late-onset depression were also assessed. In contrast to a previously published report in a younger cohort, no significant difference in [(11)C]ABP688 binding was observed between elderly subjects with MDD and healthy volunteers. [(11)C]ABP688 binding was also similar between subgroups with early- or late-onset depression. We believe this is the first study to examine mGluR5 expression in depression in the elderly. Although future work is required, results suggest potential differences in the pathophysiology of elderly depression versus depression earlier in life.
Collapse
Affiliation(s)
- C DeLorenzo
- Department of Psychiatry, Columbia University, New York, NY, USA,Department of Psychiatry, Stony Brook University, Stony Brook, NY, USA,Department of Psychiatry, Stony Brook University, HSC-T-10, Room 40D, Stony Brook, NY 11794, USA. E-mail:
| | - J Sovago
- Novartis Institute for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - J Gardus
- Department of Psychiatry, Stony Brook University, Stony Brook, NY, USA
| | - J Xu
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY, USA
| | - J Yang
- Department of Preventive Medicine, Stony Brook University, Stony Brook, NY, USA
| | - R Behrje
- Novartis Pharmaceuticals Corporations, East Hanover, NJ, USA
| | - J S D Kumar
- Department of Psychiatry, Columbia University, New York, NY, USA
| | - D P Devanand
- Department of Psychiatry, Columbia University, New York, NY, USA
| | - G H Pelton
- Department of Psychiatry, Columbia University, New York, NY, USA
| | - C A Mathis
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - N S Mason
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - B Gomez-Mancilla
- Novartis Institute for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - H Aizenstein
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - J J Mann
- Department of Psychiatry, Columbia University, New York, NY, USA
| | - R V Parsey
- Department of Psychiatry, Stony Brook University, Stony Brook, NY, USA
| |
Collapse
|
41
|
Terbeck S, Akkus F, Chesterman LP, Hasler G. The role of metabotropic glutamate receptor 5 in the pathogenesis of mood disorders and addiction: combining preclinical evidence with human Positron Emission Tomography (PET) studies. Front Neurosci 2015; 9:86. [PMID: 25852460 PMCID: PMC4364244 DOI: 10.3389/fnins.2015.00086] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 02/27/2015] [Indexed: 12/30/2022] Open
Abstract
In the present review, we deliver an overview of the involvement of metabotropic glutamate receptor 5 (mGluR5) activity and density in pathological anxiety, mood disorders and addiction. Specifically, we will describe mGluR5 studies in humans that employed Positron Emission Tomography (PET) and combined the findings with preclinical animal research. This combined view of different methodological approaches—from basic neurobiological approaches to human studies—might give a more comprehensive and clinically relevant view of mGluR5 function in mental health than the view on preclinical data alone. We will also review the current research data on mGluR5 along the Research Domain Criteria (RDoC). Firstly, we found evidence of abnormal glutamate activity related to the positive and negative valence systems, which would suggest that antagonistic mGluR5 intervention has prominent anti-addictive, anti-depressive and anxiolytic effects. Secondly, there is evidence that mGluR5 plays an important role in systems for social functioning and the response to social stress. Finally, mGluR5's important role in sleep homeostasis suggests that this glutamate receptor may play an important role in RDoC's arousal and modulatory systems domain. Glutamate was previously mostly investigated in non-human studies, however initial human clinical PET research now also supports the hypothesis that, by mediating brain excitability, neuroplasticity and social cognition, abnormal metabotropic glutamate activity might predispose individuals to a broad range of psychiatric problems.
Collapse
Affiliation(s)
- Sylvia Terbeck
- School of Psychology, Faculty of Health and Human Sciences, University of Plymouth Plymouth, UK
| | - Funda Akkus
- Division of Molecular Psychiatry, Translational Research Center, Psychiatric University Hospital, University of Bern Bern, Switzerland
| | | | - Gregor Hasler
- Division of Molecular Psychiatry, Translational Research Center, Psychiatric University Hospital, University of Bern Bern, Switzerland
| |
Collapse
|
42
|
Contoreggi C. Corticotropin releasing hormone and imaging, rethinking the stress axis. Nucl Med Biol 2014; 42:323-39. [PMID: 25573209 DOI: 10.1016/j.nucmedbio.2014.11.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Revised: 11/07/2014] [Accepted: 11/19/2014] [Indexed: 11/25/2022]
Abstract
The stress system provides integration of both neurochemical and somatic physiologic functions within organisms as an adaptive mechanism to changing environmental conditions throughout evolution. In mammals and primates the complexity and sophistication of these systems have surpassed other species in triaging neurochemical and physiologic signaling to maximize chances of survival. Corticotropin releasing hormone (CRH) and its related peptides and receptors have been identified over the last three decades and are fundamental molecular initiators of the stress response. They are crucial in the top down regulatory cascade over a myriad of neurochemical, neuroendocrine and sympathetic nervous system events. From neuroscience, we've seen that stress activation impacts behavior, endocrine and somatic physiology and influences neurochemical events that one can capture in real time with current imaging technologies. To delineate these effects one can demonstrate how the CRH neuronal networks infiltrate critical cognitive, emotive and autonomic regions of the central nervous system (CNS) with somatic effects. Abundant preclinical and clinical studies show inter-regulatory actions of CRH with multiple neurotransmitters/peptides. Stress, both acute and chronic has epigenetic effects which magnify genetic susceptibilities to alter neurochemistry; stress system activation can add critical variables in design and interpretation of basic and clinical neuroscience and related research. This review will attempt to provide an overview of the spectrum of known functions and speculative actions of CRH and stress responses in light of imaging technology and its interpretation. Metabolic and neuroreceptor positron emission/single photon tomography (PET/SPECT), functional magnetic resonance imaging (fMRI), anatomic MRI, diffusion tensor imaging (DTI), and proton magnetic resonance spectroscopy (pMRS) are technologies that can delineate basic mechanisms of neurophysiology and pharmacology. Stress modulates the myriad of neurochemical and networks within and controlled through the central and peripheral nervous system and the effects of stress activation on imaging will be highlighted.
Collapse
Affiliation(s)
- Carlo Contoreggi
- Intramural Research Program (IRP), National Institute on Drug Abuse (NIDA), National Institutes of Health (NIH), Baltimore, MD, 21224.
| |
Collapse
|