1
|
Zhai S, Cui Q, Wokosin D, Sun L, Tkatch T, Crittenden JR, Graybiel AM, Surmeier DJ. State-dependent modulation of spiny projection neurons controls levodopa-induced dyskinesia in a mouse model of Parkinson's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.02.631090. [PMID: 39829758 PMCID: PMC11741361 DOI: 10.1101/2025.01.02.631090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
In the later stages of Parkinson's disease (PD), patients often manifest levodopa-induced dyskinesia (LID), compromising their quality of life. The pathophysiology underlying LID is poorly understood, and treatment options are limited. To move toward filling this treatment gap, the intrinsic and synaptic changes in striatal spiny projection neurons (SPNs) triggered by the sustained elevation of dopamine (DA) during dyskinesia were characterized using electrophysiological, pharmacological, molecular and behavioral approaches. Our studies revealed that the intrinsic excitability and functional corticostriatal connectivity of SPNs in dyskinetic mice oscillate between the on- and off-states of LID in a cell- and state-specific manner. Although triggered by levodopa, these rapid oscillations in SPN properties depended on both dopaminergic and cholinergic signaling. In a mouse PD model, disrupting M1 muscarinic receptor signaling specifically in iSPNs or deleting its downstream signaling partner CalDAG-GEFI blunted the levodopa-induced oscillation in functional connectivity, enhanced the beneficial effects of levodopa and attenuated LID severity.
Collapse
Affiliation(s)
- Shenyu Zhai
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Qiaoling Cui
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815 USA
| | - David Wokosin
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Linqing Sun
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Tatiana Tkatch
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815 USA
| | - Jill R. Crittenden
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, MIT, Cambridge, MA 02139, USA
| | - Ann M. Graybiel
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, MIT, Cambridge, MA 02139, USA
| | - D. James Surmeier
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815 USA
| |
Collapse
|
2
|
Gokalp D, Unal G. The role of mGluR5 on the therapeutic effects of ketamine in Wistar rats. Psychopharmacology (Berl) 2024; 241:1399-1415. [PMID: 38459971 PMCID: PMC11199271 DOI: 10.1007/s00213-024-06571-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 03/04/2024] [Indexed: 03/11/2024]
Abstract
RATIONALE Ketamine produces dissociative, psychomimetic, anxiolytic, antidepressant, and anesthetic effects in a dose dependent manner. It has a complex mechanism of action that involve alterations in other glutamate receptors. The metabotropic glutamate receptor 5 (mGluR5) has been investigated in relation to the psychotic and anesthetic properties of ketamine, while its role in mediating the therapeutic effects of ketamine remains unknown. OBJECTIVES We investigated the role of mGluR5 on the antidepressant, anxiolytic and fear memory-related effects of ketamine in adult male Wistar rats. METHODS Two sets of experiments were conducted. We first utilized the positive allosteric modulator CDPPB to investigate how acute mGluR5 activation regulates the therapeutic effects of ketamine (10 mg/kg). We then tested the synergistic antidepressant effect of mGluR5 antagonism and ketamine by combining MTEP with a sub-effective dose of ketamine (1 mg/kg). Behavioral despair, locomotor activity, anxiety-like behavior, and fear memory were respectively assessed in the forced swim test (FST), open field test (OFT), elevated plus maze (EPM), and auditory fear conditioning. RESULTS Enhancing mGluR5 activity via CDPPB occluded the antidepressant effect of ketamine without changing locomotor activity. Furthermore, concomitant administration of MTEP and ketamine exhibited a robust synergistic antidepressant effect. The MTEP + ketamine treatment, however, blocked the anxiolytic effect observed by sole administration of MTEP or the low dose ketamine. CONCLUSIONS These findings suggest that suppressed mGluR5 activity is required for the antidepressant effects of ketamine. Consequently, the antagonism of mGluR5 enhances the antidepressant effectiveness of low dose ketamine, but eliminates its anxiolytic effects.
Collapse
Affiliation(s)
- Dilan Gokalp
- Behavioral Neuroscience Laboratory, Department of Psychology, Boğaziçi University, 34342, Istanbul, Turkey
| | - Gunes Unal
- Behavioral Neuroscience Laboratory, Department of Psychology, Boğaziçi University, 34342, Istanbul, Turkey.
| |
Collapse
|
3
|
McClatchy DB, Powell SB, Yates JR. In vivo mapping of protein-protein interactions of schizophrenia risk factors generates an interconnected disease network. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.12.571320. [PMID: 38168169 PMCID: PMC10759996 DOI: 10.1101/2023.12.12.571320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Genetic analyses of Schizophrenia (SCZ) patients have identified thousands of risk factors. In silico protein-protein interaction (PPI) network analysis has provided strong evidence that disrupted PPI networks underlie SCZ pathogenesis. In this study, we performed in vivo PPI analysis of several SCZ risk factors in the rodent brain. Using endogenous antibody immunoprecipitations coupled to mass spectrometry (MS) analysis, we constructed a SCZ network comprising 1612 unique PPI with a 5% FDR. Over 90% of the PPI were novel, reflecting the lack of previous PPI MS studies in brain tissue. Our SCZ PPI network was enriched with known SCZ risk factors, which supports the hypothesis that an accumulation of disturbances in selected PPI networks underlies SCZ. We used Stable Isotope Labeling in Mammals (SILAM) to quantitate phencyclidine (PCP) perturbations in the SCZ network and found that PCP weakened most PPI but also led to some enhanced or new PPI. These findings demonstrate that quantitating PPI in perturbed biological states can reveal alterations to network biology.
Collapse
|
4
|
Bueschke N, Amaral-Silva L, Hu M, Alvarez A, Santin JM. Plasticity in the functional properties of NMDA receptors improves network stability during severe energy stress. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.19.524811. [PMID: 36711958 PMCID: PMC9882286 DOI: 10.1101/2023.01.19.524811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Brain energy stress leads to neuronal hyperexcitability followed by a rapid loss of function and cell death. In contrast, the frog brainstem switches into a state of extreme metabolic resilience that allows them to maintain motor function during hypoxia as they emerge from hibernation. NMDA receptors (NMDARs) are Ca2+-permeable glutamate receptors that contribute to the loss of homeostasis during hypoxia. Therefore, we hypothesized that hibernation leads to plasticity that reduces the role of NMDARs within neural networks to improve function during energy stress. To test this, we assessed a circuit with a large involvement of NMDAR synapses, the brainstem respiratory network of female bullfrogs, Lithobates catesbeianus. Contrary to our expectations, hibernation did not alter the role of NMDARs in generating network output, nor did it affect the amplitude, kinetics, and hypoxia sensitivity of NMDAR currents. Instead, hibernation strongly reduced NMDAR Ca2+ permeability and enhanced desensitization during repetitive stimulation. Under severe hypoxia, the normal NMDAR profile caused network hyperexcitability within minutes, which was mitigated by blocking NMDARs. After hibernation, the modified complement of NMDARs protected against hyperexcitability, as disordered output did not occur for at least one hour in hypoxia. These findings uncover state-dependence in the plasticity of NMDARs, whereby multiple changes to receptor function improve neural performance during energy stress without interfering with its normal role during healthy activity.
Collapse
Affiliation(s)
| | | | - Min Hu
- University of North Carolina-Greensboro, Greensboro, NC 27402
| | - Alvaro Alvarez
- University of North Carolina-Greensboro, Greensboro, NC 27402
| | | |
Collapse
|
5
|
Hoglund BK, Carfagno V, Olive MF, Leyrer-Jackson JM. Metabotropic glutamate receptors and cognition: From underlying plasticity and neuroprotection to cognitive disorders and therapeutic targets. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2023; 168:367-413. [PMID: 36868635 DOI: 10.1016/bs.irn.2022.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Metabotropic glutamate (mGlu) receptors are G protein-coupled receptors that play pivotal roles in mediating the activity of neurons and other cell types within the brain, communication between cell types, synaptic plasticity, and gene expression. As such, these receptors play an important role in a number of cognitive processes. In this chapter, we discuss the role of mGlu receptors in various forms of cognition and their underlying physiology, with an emphasis on cognitive dysfunction. Specifically, we highlight evidence that links mGlu physiology to cognitive dysfunction across brain disorders including Parkinson's disease, Alzheimer's disease, Fragile X syndrome, post-traumatic stress disorder, and schizophrenia. We also provide recent evidence demonstrating that mGlu receptors may elicit neuroprotective effects in particular disease states. Lastly, we discuss how mGlu receptors can be targeted utilizing positive and negative allosteric modulators as well as subtype specific agonists and antagonist to restore cognitive function across these disorders.
Collapse
Affiliation(s)
- Brandon K Hoglund
- Department of Medical Education, School of Medicine, Creighton University, Phoenix, AZ, United States
| | - Vincent Carfagno
- School of Medicine, Midwestern University, Glendale, AZ, United States
| | - M Foster Olive
- Department of Psychology, Arizona State University, Tempe, AZ, United States
| | - Jonna M Leyrer-Jackson
- Department of Medical Education, School of Medicine, Creighton University, Phoenix, AZ, United States.
| |
Collapse
|
6
|
Azam S, Jakaria M, Kim J, Ahn J, Kim IS, Choi DK. Group I mGluRs in Therapy and Diagnosis of Parkinson’s Disease: Focus on mGluR5 Subtype. Biomedicines 2022; 10:biomedicines10040864. [PMID: 35453614 PMCID: PMC9032558 DOI: 10.3390/biomedicines10040864] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/05/2022] [Accepted: 04/06/2022] [Indexed: 11/16/2022] Open
Abstract
Metabotropic glutamate receptors (mGluRs; members of class C G-protein-coupled receptors) have been shown to modulate excitatory neurotransmission, regulate presynaptic extracellular glutamate levels, and modulate postsynaptic ion channels on dendritic spines. mGluRs were found to activate myriad signalling pathways to regulate synapse formation, long-term potentiation, autophagy, apoptosis, necroptosis, and pro-inflammatory cytokines release. A notorious expression pattern of mGluRs has been evident in several neurodegenerative diseases, including Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, and schizophrenia. Among the several mGluRs, mGluR5 is one of the most investigated types of considered prospective therapeutic targets and potential diagnostic tools in neurodegenerative diseases and neuropsychiatric disorders. Recent research showed mGluR5 radioligands could be a potential tool to assess neurodegenerative disease progression and trace respective drugs’ kinetic properties. This article provides insight into the group I mGluRs, specifically mGluR5, in the progression and possible therapy for PD.
Collapse
Affiliation(s)
- Shofiul Azam
- Department of Applied Life Science, Graduate School, BK21 Program, Konkuk University, Chungju 27478, Korea; (S.A.); (M.J.); (J.K.); (J.A.)
| | - Md. Jakaria
- Department of Applied Life Science, Graduate School, BK21 Program, Konkuk University, Chungju 27478, Korea; (S.A.); (M.J.); (J.K.); (J.A.)
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC 3052, Australia
| | - JoonSoo Kim
- Department of Applied Life Science, Graduate School, BK21 Program, Konkuk University, Chungju 27478, Korea; (S.A.); (M.J.); (J.K.); (J.A.)
| | - Jaeyong Ahn
- Department of Applied Life Science, Graduate School, BK21 Program, Konkuk University, Chungju 27478, Korea; (S.A.); (M.J.); (J.K.); (J.A.)
| | - In-Su Kim
- Department of Biotechnology, College of Biomedical and Health Science, Research Institute of Inflammatory Disease (RID), Konkuk University, Chungju 27478, Korea
- Correspondence: (I.-S.K.); (D.-K.C.); Tel.: +82-43-840-3905 (I.-S.K.); +82-43-840-3610 (D.-K.C.); Fax: +82-43-840-3872 (D.-K.C.)
| | - Dong-Kug Choi
- Department of Applied Life Science, Graduate School, BK21 Program, Konkuk University, Chungju 27478, Korea; (S.A.); (M.J.); (J.K.); (J.A.)
- Department of Biotechnology, College of Biomedical and Health Science, Research Institute of Inflammatory Disease (RID), Konkuk University, Chungju 27478, Korea
- Correspondence: (I.-S.K.); (D.-K.C.); Tel.: +82-43-840-3905 (I.-S.K.); +82-43-840-3610 (D.-K.C.); Fax: +82-43-840-3872 (D.-K.C.)
| |
Collapse
|
7
|
NMDA receptors elicit flux-independent intracellular Ca 2+ signals via metabotropic glutamate receptors and flux-dependent nitric oxide release in human brain microvascular endothelial cells. Cell Calcium 2021; 99:102454. [PMID: 34454368 DOI: 10.1016/j.ceca.2021.102454] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 07/28/2021] [Accepted: 08/12/2021] [Indexed: 12/20/2022]
Abstract
The excitatory neurotransmitter glutamate gates post-synaptic N-methyl-d-aspartate (NMDA) receptors (NMDARs) to mediate extracellular Ca2+ entry and stimulate neuronal nitric oxide (NO) synthase to release NO and trigger neurovascular coupling (NVC). Neuronal and glial NMDARs may also operate in a flux-independent manner, although it is unclear whether their non-ionotropic mode of action is involved in NVC. Recently, endothelial NMDARs were found to trigger Ca2+-dependent NO production and induce NVC, but the underlying mode of signaling remains elusive. Herein, we report that GluN1 protein, as well as GluN2C and GluN3B transcripts and proteins, were expressed and that NMDA did not elicit inward currents, but induced a dose-dependent increase in intracellular Ca2+ concentration ([Ca2+]i) in the human brain microvascular endothelial cell line, hCMEC/D3. A multidisciplinary approach, including live cell imaging, whole-cell patch-clamp recordings, pharmacological manipulation and gene targeting, revealed that NMDARs increase the [Ca2+]i in a flux-independent manner in hCMEC/D3 cells. The Ca2+ response to NMDA was triggered by endogenous Ca2+ release from the endoplasmic reticulum and the lysosomal Ca2+ stores and sustained by store-operated Ca2+ entry. Unexpectedly, pharmacological and genetic blockade of mGluR1 and mGluR5 dramatically impaired NMDARs-mediated Ca2+ signals. These findings indicate that NMDARs may increase the endothelial [Ca2+]i in a flux-independent manner via group 1 mGluRs. However, imaging of DAF-FM fluorescence revealed that NMDARs may also induce Ca2+-dependent NO release by signaling in a flux-dependent manner. These findings, therefore, shed novel light on the mechanisms whereby brain microvascular endothelium decodes glutamatergic signaling and regulates NVC.
Collapse
|
8
|
Maternal stress programs a demasculinization of glutamatergic transmission in stress-related brain regions of aged rats. GeroScience 2021; 44:1047-1069. [PMID: 33983623 PMCID: PMC8116647 DOI: 10.1007/s11357-021-00375-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Accepted: 04/20/2021] [Indexed: 12/20/2022] Open
Abstract
Brain aging may be programmed by early-life stress. Aging affects males and females differently, but how perinatal stress (PRS) affects brain aging between sexes is unknown. We showed behavioral and neurobiological sex differences in non-stressed control rats that were strongly reduced or inverted in PRS rats. In particular, PRS decreased risk-taking behavior, spatial memory, exploratory behavior, and fine motor behavior in male aged rats. In contrast, female aged PRS rats displayed only increased risk-taking behavior and reduced exploratory behavior. PRS induced large reductions in the expression of glutamate receptors in the ventral and dorsal hippocampus and prefrontal cortex only in male rats. PRS also reduced the expression of synaptic vesicle-associated proteins, glucocorticoid receptors (GR), and mineralocorticoid receptors (MR) in the ventral hippocampus of aged male rats. In contrast, in female aged rats, PRS enhanced the expression of MRs and brain-derived neurotrophic factor (BDNF) in the ventral hippocampus and the expression of glial fibrillary acidic protein (GFAP) and BDNF in the prefrontal cortex. A common PRS effect in both sexes was a reduction in exploratory behavior and metabotropic glutamate (mGlu2/3) receptors in the ventral hippocampus and prefrontal cortex. A multidimensional analysis revealed that PRS induced a demasculinization profile in glutamate-related proteins in the ventral and dorsal hippocampus and prefrontal cortex, as well as a demasculinization profile of stress markers only in the dorsal hippocampus. In contrast, defeminization was observed only in the ventral hippocampus. Measurements of testosterone and 17-β-estradiol in the plasma and aromatase in the dorsal hippocampus were consistent with a demasculinizing action of PRS. These findings confirm that the brains of males and females differentially respond to PRS and aging suggesting that females might be more protected against early stress and age-related inflammation and neurodegeneration. Taken together, these results may contribute to understanding how early environmental factors shape vulnerability to brain aging in both sexes and may lay the groundwork for future studies aimed at identifying new treatment strategies to improve the quality of life of older individuals, which is of particular interest given that there is a high growth of aging in populations around the world.
Collapse
|
9
|
AlShimemeri S, Fox SH, Visanji NP. Emerging drugs for the treatment of L-DOPA-induced dyskinesia: an update. Expert Opin Emerg Drugs 2020; 25:131-144. [DOI: 10.1080/14728214.2020.1763954] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Sohaila AlShimemeri
- Edmond J Safra Program in Parkinson Disease & Morton and Gloria Shulman Movement Disorders Centre, Toronto Western Hospital, Toronto, ON, Canada
- Division of Neurology, Department of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Susan H Fox
- Edmond J Safra Program in Parkinson Disease & Morton and Gloria Shulman Movement Disorders Centre, Toronto Western Hospital, Toronto, ON, Canada
| | - Naomi P Visanji
- Edmond J Safra Program in Parkinson Disease & Morton and Gloria Shulman Movement Disorders Centre, Toronto Western Hospital, Toronto, ON, Canada
| |
Collapse
|
10
|
Wang HY, MacDonald ML, Borgmann-Winter KE, Banerjee A, Sleiman P, Tom A, Khan A, Lee KC, Roussos P, Siegel SJ, Hemby SE, Bilker WB, Gur RE, Hahn CG. mGluR5 hypofunction is integral to glutamatergic dysregulation in schizophrenia. Mol Psychiatry 2020; 25:750-760. [PMID: 30214040 PMCID: PMC7500805 DOI: 10.1038/s41380-018-0234-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 06/18/2018] [Accepted: 07/09/2018] [Indexed: 11/19/2022]
Abstract
Multiple lines of evidence point to glutamatergic signaling in the postsynaptic density (PSD) as a pathophysiologic mechanism in schizophrenia. Integral to PSD glutamatergic signaling is reciprocal interplay between GluN and mGluR5 signaling. We examined agonist-induced mGluR5 signaling in the postmortem dorsolateral prefrontal cortex (DLPFC) derived from 17 patients and age-matched and sex-matched controls. The patient group showed a striking reduction in mGluR5 signaling, manifested by decreases in Gq/11 coupling and association with PI3K and Homer compared to controls (p < 0.01 for all). This was accompanied by increases in serine and tyrosine phosphorylation of mGluR5, which can decrease mGluR5 activity via desensitization (p < 0.01). In addition, we find altered protein-protein interaction (PPI) of mGluR5 with RGS4, norbin, Preso 1 and tamalin, which can also attenuate mGluR5 activity. We previously reported molecular underpinnings of GluN hypofunction (decreased GluN2 phosphorylation) and here we show those of reduced mGluR5 signaling in schizophrenia. We find that reduced GluN2 phosphorylation can be precipitated by attenuated mGluR5 activity and that increased mGluR5 phosphorylation can result from decreased GluN function, suggesting a reciprocal interplay between the two pathways in schizophrenia. Interestingly, the patient group showed decreased mGluR5-GluN association (p < 0.01), a mechanistic basis for the reciprocal facilitation. In sum, we present the first direct evidence for mGluR5 hypoactivity, propose a reciprocal interplay between GluN and mGluR5 pathways as integral to glutamatergic dysregulation and suggest protein-protein interactions in mGluR5-GluN complexes as potential targets for intervention in schizophrenia.
Collapse
Affiliation(s)
- Hoau-Yan Wang
- Department of Physiology, Pharmacology and Neuroscience, City University of New York School of Medicine, New York, NY 10031,Department of Biology and Neuroscience, Graduate School of the City University of New York, NY 10016
| | - Mathew L. MacDonald
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104-3403
| | - Karin E. Borgmann-Winter
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104-3403,Department of Child and Adolescent Psychiatry and Behavioral Sciences, Children’s Hospital of Philadelphia, Philadelphia, PA 19104
| | - Anamika Banerjee
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104-3403
| | - Patrick Sleiman
- The Center for Applied Genomics, The Children’s Hospital of Philadelphia, and Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104
| | - Andrew Tom
- Department of Physiology, Pharmacology and Neuroscience, City University of New York School of Medicine, New York, NY 10031
| | - Amber Khan
- Department of Physiology, Pharmacology and Neuroscience, City University of New York School of Medicine, New York, NY 10031,Department of Biology and Neuroscience, Graduate School of the City University of New York, NY 10016
| | - Kuo-Chieh Lee
- Department of Physiology, Pharmacology and Neuroscience, City University of New York School of Medicine, New York, NY 10031
| | - Panos Roussos
- Department of Psychiatry, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029
| | - Steven J. Siegel
- Department of Psychiatry and the Behavioral Sciences, University of Southern California, Los Angeles, CA, 90007
| | - Scott E Hemby
- Department of Basic Pharmaceutical Sciences, High Point University, High Point, NC, 27106
| | - Warren B. Bilker
- Department of Biostatistics and Epidemiology, University of Pennsylvania, Philadelphia, PA 19104
| | - Raquel E. Gur
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104-3403
| | - Chang-Gyu Hahn
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, 19104-3403, USA.
| |
Collapse
|
11
|
Nam HW, Grant CA, Jorgensen AN, Holtz-Heppelmann CJ, Trutschl M, Cvek U. Neurogranin Regulates Alcohol Sensitivity through AKT Pathway in the Nucleus Accumbens. Proteomics 2019; 20:e1900266. [PMID: 31814311 DOI: 10.1002/pmic.201900266] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 11/19/2019] [Indexed: 01/03/2023]
Abstract
Dysfunction of glutamate neurotransmission in the nucleus accumbens (NAc) has been implicated in the pathophysiology of alcohol use disorders (AUD). Neurogranin (Ng) is exclusively expressed in the brain and mediates N-methyl-d-aspartate receptor (NMDAR) hypo-function by regulating the intracellular calcium-calmodulin (Ca2+ -CaM) pathway. Ng null mice (Ng-/- mice) demonstrate increased alcohol drinking compared to wild-type mice, while also showing less tolerance to the effect of alcohol. To identify the molecular mechanism related to alcohol seeking, both in vivo microdialysis and label-free quantification proteomics comparing Ng genotype and effects of alcohol treatment on the NAc are utilized. There is significant difference in glutamate and gamma-aminobutyric acid (GABA) neurotransmission between genotypes; however, alcohol administration normalizes both glutamate and GABA levels in the NAc. Using label-free proteomics, 427 protein expression changes are identified against alcohol treatment in the NAc among 4347 total proteins detected. Bioinformatics analyses reveal significant molecular differences in Ng null mice in response to acute alcohol treatment. Ingenuity pathway analysis found that the AKT network is altered significantly between genotypes, which may increase the sensitivity of alcohol in Ng null mice. The pharmacoproteomics results presented here illustrate a possible molecular basis of the alcohol sensitivity through Ng signaling in the NAc.
Collapse
Affiliation(s)
- Hyung W Nam
- Department of Pharmacology, Toxicology, and Neuroscience, Health Science Center, Louisiana State University, Shreveport, LA, 71130, USA
| | - Caleb A Grant
- Department of Pharmacology, Toxicology, and Neuroscience, Health Science Center, Louisiana State University, Shreveport, LA, 71130, USA
| | - Ashton N Jorgensen
- Department of Pharmacology, Toxicology, and Neuroscience, Health Science Center, Louisiana State University, Shreveport, LA, 71130, USA
| | | | - Marjan Trutschl
- Department of Computer Science, Louisiana State University-Shreveport, Shreveport, LA, 71115, USA
| | - Urska Cvek
- Department of Computer Science, Louisiana State University-Shreveport, Shreveport, LA, 71115, USA
| |
Collapse
|
12
|
Prenatal valproate in rodents as a tool to understand the neural underpinnings of social dysfunctions in autism spectrum disorder. Neuropharmacology 2019; 159:107477. [DOI: 10.1016/j.neuropharm.2018.12.024] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 12/11/2018] [Accepted: 12/20/2018] [Indexed: 12/22/2022]
|
13
|
Dal Prà I, Armato U, Chiarini A. Family C G-Protein-Coupled Receptors in Alzheimer's Disease and Therapeutic Implications. Front Pharmacol 2019; 10:1282. [PMID: 31719824 PMCID: PMC6826475 DOI: 10.3389/fphar.2019.01282] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 10/07/2019] [Indexed: 12/12/2022] Open
Abstract
Alzheimer’s disease (AD), particularly its sporadic or late-onset form (SAD/LOAD), is the most prevalent (96–98% of cases) neurodegenerative dementia in aged people. AD’s neuropathology hallmarks are intrabrain accumulation of amyloid-β peptides (Aβs) and of hyperphosphorylated Tau (p-Tau) proteins, diffuse neuroinflammation, and progressive death of neurons and oligodendrocytes. Mounting evidences suggest that family C G-protein-coupled receptors (GPCRs), which include γ-aminobutyric acid B receptors (GABABRs), metabotropic glutamate receptors (mGluR1-8), and the calcium-sensing receptor (CaSR), are involved in many neurotransmitter systems that dysfunction in AD. This review updates the available knowledge about the roles of GPCRs, particularly but not exclusively those expressed by brain astrocytes, in SAD/LOAD onset and progression, taking stock of their respective mechanisms of action and of their potential as anti-AD therapeutic targets. In particular, GABABRs prevent Aβs synthesis and neuronal hyperexcitability and group I mGluRs play important pathogenetic roles in transgenic AD-model animals. Moreover, the specific binding of Aβs to the CaSRs of human cortical astrocytes and neurons cultured in vitro engenders a pathological signaling that crucially promotes the surplus synthesis and release of Aβs and hyperphosphorylated Tau proteins, and also of nitric oxide, vascular endothelial growth factor-A, and proinflammatory agents. Concurrently, Aβs•CaSR signaling hinders the release of soluble (s)APP-α peptide, a neurotrophic agent and GABABR1a agonist. Altogether these effects progressively kill human cortical neurons in vitro and likely also in vivo. Several CaSR’s negative allosteric modulators suppress all the noxious effects elicited by Aβs•CaSR signaling in human cortical astrocytes and neurons thus safeguarding neurons’ viability in vitro and raising hopes about their potential therapeutic benefits in AD patients. Further basic and clinical investigations on these hot topics are needed taking always heed that activation of the several brain family C GPCRs may elicit divergent upshots according to the models studied.
Collapse
Affiliation(s)
- Ilaria Dal Prà
- Human Histology and Embryology Unit, University of Verona Medical School, Verona, Italy
| | - Ubaldo Armato
- Human Histology and Embryology Unit, University of Verona Medical School, Verona, Italy
| | - Anna Chiarini
- Human Histology and Embryology Unit, University of Verona Medical School, Verona, Italy
| |
Collapse
|
14
|
Behavioral consequences of co-administration of MTEP and the COX-2 inhibitor NS398 in mice. Part 1. Behav Brain Res 2019; 370:111961. [DOI: 10.1016/j.bbr.2019.111961] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 05/16/2019] [Accepted: 05/17/2019] [Indexed: 01/01/2023]
|
15
|
Hellyer SD, Albold S, Sengmany K, Singh J, Leach K, Gregory KJ. Metabotropic glutamate receptor 5 (mGlu 5 )-positive allosteric modulators differentially induce or potentiate desensitization of mGlu 5 signaling in recombinant cells and neurons. J Neurochem 2019; 151:301-315. [PMID: 31376155 DOI: 10.1111/jnc.14844] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 07/17/2019] [Accepted: 07/31/2019] [Indexed: 12/14/2022]
Abstract
Allosteric modulators of metabotropic glutamate receptor 5 (mGlu5 ) are a promising therapeutic strategy for a number of neurological disorders. Multiple mGlu5 -positive allosteric modulator (PAM) chemotypes have been discovered that act as either pure PAMs or as PAM-agonists in recombinant and native cells. While these compounds have been tested in paradigms of receptor activation, their effects on receptor regulatory processes are largely unknown. In this study, acute desensitization of mGlu5 mediated intracellular calcium mobilization by structurally diverse mGlu5 orthosteric and allosteric ligands was assessed in human embryonic kidney 293 cells and primary murine neuronal cultures from both striatum and cortex. We aimed to determine the intrinsic efficacy and modulatory capacity of diverse mGlu5 PAMs [(R)-5-((3-fluorophenyl)ethynyl)-N-(3-hydroxy-3-methylbutan-2-yl)picolinamide (VU0424465), N-cyclobutyl-6-((3-fluorophenyl)ethynyl)picolinamide (VU0360172), 1-(4-(2,4-difluorophenyl)piperazin-1-yl)-2-((4-fluorobenzyl)oxy)ethanone (DPFE), ((4-fluorophenyl) (2-(phenoxymethyl)-6,7-dihydrooxazolo[5,4-c]pyridin-5(4H)-yl)methanone) (VU0409551), 3-Cyano-N-(1,3-diphenyl-1H-pyrazol-5-yl)benzamide (CDPPB)] on receptor desensitization and whether cellular context influences receptor regulatory processes. Only VU0424465 and VU0409551 induced desensitization alone in human embryonic kidney 293-mGlu5 cells, while all PAMs enhanced (S)-3,5-dihydroxyphenylglycine (DHPG)-induced desensitization. All mGlu5 PAMs induced receptor desensitization alone and enhanced DHPG-induced desensitization in striatal neurons. VU0424465 and VU0360172 were the only PAMs that induced desensitization alone in cortical neurons. With the exception of (CDPPB), PAMs enhanced DHPG-induced desensitization in cortical neurons. Moreover, differential apparent affinities, efficacies, and cooperativities with DHPG were observed for VU0360172, VU0409551, and VU0424465 when comparing receptor activation and desensitization in a cell type-dependent manner. These data indicate that biased mGlu5 allosteric modulator pharmacology extends to receptor regulatory processes in a tissue dependent manner, adding yet another layer of complexity to rational mGlu5 drug discovery.
Collapse
Affiliation(s)
- Shane D Hellyer
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria, Australia
| | - Sabine Albold
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria, Australia
| | - Kathy Sengmany
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria, Australia
| | - Junaid Singh
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria, Australia
| | - Katie Leach
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria, Australia
| | - Karen J Gregory
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria, Australia
| |
Collapse
|
16
|
Reiner A, Levitz J. Glutamatergic Signaling in the Central Nervous System: Ionotropic and Metabotropic Receptors in Concert. Neuron 2019; 98:1080-1098. [PMID: 29953871 DOI: 10.1016/j.neuron.2018.05.018] [Citation(s) in RCA: 376] [Impact Index Per Article: 62.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 04/19/2018] [Accepted: 05/10/2018] [Indexed: 12/28/2022]
Abstract
Glutamate serves as both the mammalian brain's primary excitatory neurotransmitter and as a key neuromodulator to control synapse and circuit function over a wide range of spatial and temporal scales. This functional diversity is decoded by two receptor families: ionotropic glutamate receptors (iGluRs) and metabotropic glutamate receptors (mGluRs). The challenges posed by the complexity and physiological importance of each of these subtypes has limited our appreciation and understanding of how these receptors work in concert. In this review, by comparing both receptor families with a focus on their crosstalk, we argue for a more holistic understanding of neural glutamate signaling.
Collapse
Affiliation(s)
- Andreas Reiner
- Department of Biology and Biotechnology, Ruhr University Bochum, Bochum, Germany.
| | - Joshua Levitz
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
17
|
Mahan VL. Neurointegrity and neurophysiology: astrocyte, glutamate, and carbon monoxide interactions. Med Gas Res 2019; 9:24-45. [PMID: 30950417 PMCID: PMC6463446 DOI: 10.4103/2045-9912.254639] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 02/15/2019] [Indexed: 12/27/2022] Open
Abstract
Astrocyte contributions to brain function and prevention of neuropathologies are as extensive as that of neurons. Astroglial regulation of glutamate, a primary neurotransmitter, is through uptake, release through vesicular and non-vesicular pathways, and catabolism to intermediates. Homeostasis by astrocytes is considered to be of primary importance in determining normal central nervous system health and central nervous system physiology - glutamate is central to dynamic physiologic changes and central nervous system stability. Gasotransmitters may affect diverse glutamate interactions positively or negatively. The effect of carbon monoxide, an intrinsic central nervous system gasotransmitter, in the complex astrocyte homeostasis of glutamate may offer insights to normal brain development, protection, and its use as a neuromodulator and neurotherapeutic. In this article, we will review the effects of carbon monoxide on astrocyte homeostasis of glutamate.
Collapse
Affiliation(s)
- Vicki L. Mahan
- Division of Pediatric Cardiothoracic Surgery in the Department of Surgery, St. Christopher's Hospital for Children/Drexel University College of Medicine, Philadelphia, PA, USA
| |
Collapse
|
18
|
Chang J, Zhang XL, Yu H, Chen J. Downregulation of RTN1-C attenuates MPP +-induced neuronal injury through inhibition of mGluR5 pathway in SN4741 cells. Brain Res Bull 2018; 146:1-6. [PMID: 30521940 DOI: 10.1016/j.brainresbull.2018.11.026] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Revised: 11/28/2018] [Accepted: 11/30/2018] [Indexed: 11/29/2022]
Abstract
Reticulons (RTNs) are a group of membrane-bound proteins that are dominantly localized to the endoplasmic reticulum (ER). RTN1-C, one isoform of RTNs highly expressed in the brain, has been shown to mediate neuronal injury in cerebral ischemia models. The aim of this study was to investigate the role of RTN1-C in an in vitro model of Parkinson's disease (PD) mimicked by 1-methyl-4-phenylpyridinium (MPP+) treatment in SN4741 cells. We found that MPP+ significantly increased the expression of RTN1-C, with no effect on RTN1-A and RTN1-B. Downregulation of RTN1-C using siRNA (Si-RTN1-C) markedly increased cell viability and inhibited apoptosis induced by MPP+ treatment. The results of western blot showed that downregulation of RTN1-C inhibited the surface expression of metabotropic glutamate receptor 5 (mGluR5) but had no effect on mGluR1. The protective effects of Si-RTN1-C were partially prevented by activating mGluR5, not mGluR1. In addition, the results of Ca2+ imaging showed that downregulation of RTN1-C attenuated intracellular Ca2+ release induced by MPP+, which could be nullified by activation of mGluR5 pathway. In conclusion, our data suggest that downregulation of RTN1-C protects SN4741 cells against MPP+ through mGluR5-mediated preservation of Ca2+ homeostasis. Therefore, RTN1-C might represent a therapeutic target for the treatment of neuronal injury in experimental PD models.
Collapse
Affiliation(s)
- Jiang Chang
- Department of Encephalopathy, Shaanxi Provincial Hospital of Traditional Chinese Medicine, Xi'an, Shaanxi, 710003, China
| | - Xiao-Le Zhang
- Department of Encephalopathy, Shaanxi Provincial Hospital of Traditional Chinese Medicine, Xi'an, Shaanxi, 710003, China
| | - Hua Yu
- Department of Encephalopathy, Shaanxi Provincial Hospital of Traditional Chinese Medicine, Xi'an, Shaanxi, 710003, China
| | - Jie Chen
- Department of Encephalopathy, Shaanxi Provincial Hospital of Traditional Chinese Medicine, Xi'an, Shaanxi, 710003, China.
| |
Collapse
|
19
|
Lundbye CJ, Toft AKH, Banke TG. Inhibition of GluN2A NMDA receptors ameliorates synaptic plasticity deficits in the Fmr1 -/y mouse model. J Physiol 2018; 596:5017-5031. [PMID: 30132892 DOI: 10.1113/jp276304] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Accepted: 08/20/2018] [Indexed: 02/06/2023] Open
Abstract
KEY POINTS Fragile X syndrome (FXS) is a genetic condition that is the most common form of inherited intellectual impairment and causes a range of neurodevelopmental complications including learning disabilities and intellectual disability and shares many characteristics with autism spectrum disorder (ASD). In the FXS mouse model, Fmr1-/y , impaired synaptic plasticity was restored by pharmacologically inhibiting GluN2A-containing NMDA receptors but not GluN2B-containing receptors. Similar results were obtained by crossing Fmr1-/y with GluN2A knock-out (Grin2A-/- ) mice. These results suggest that dampening the elevated levels of GluN2A-containing NMDA receptors in Fmr1-/y mice has the potential to restore hyperexcitability of the neural circuitry to (a more) normal-like level of brain activity. ABSTRACT NMDA receptors (NMDARs) play important roles in synaptic plasticity at central excitatory synapses, and dysregulation of their function may lead to severe disorders such Fragile X syndrome (FXS). FXS is caused by transcriptional silencing of the FMR1 gene followed by lack of the encoding protein. Here we examined the effects of pharmacological and genetic manipulation of hippocampal NMDAR functions in long-term potentiation (LTP) and depression (LTD). We found impaired NMDAR-dependent LTP in the Fmr1-deficient mice, which could be fully restored when GluN2A-containing NMDARs was pharmacological inhibited. Interestingly, similar LTP effects were observed when the GluN2A gene (Grin2a) was deleted in Fmr1-/y mice (Fmr1-/y /Grin2a-/- double knockout). In addition, GluN2A inhibition improved elevated mGluR5-dependent LTD to normal level in the Fmr1-/y mouse. These findings suggest that GluN2A is a promising target in FXS research that could help us better understand the disorder.
Collapse
Affiliation(s)
- Camilla J Lundbye
- Institute of Biomedicine - Physiology, Aarhus University, DK-8000, Aarhus, Denmark
| | - Anna Karina H Toft
- Institute of Biomedicine - Physiology, Aarhus University, DK-8000, Aarhus, Denmark
| | - Tue G Banke
- Institute of Biomedicine - Physiology, Aarhus University, DK-8000, Aarhus, Denmark
| |
Collapse
|
20
|
Ketamine and Ceftriaxone-Induced Alterations in Glutamate Levels Do Not Impact the Specific Binding of Metabotropic Glutamate Receptor Subtype 5 Radioligand [ 18F]PSS232 in the Rat Brain. Pharmaceuticals (Basel) 2018; 11:ph11030083. [PMID: 30158438 PMCID: PMC6161118 DOI: 10.3390/ph11030083] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 08/23/2018] [Accepted: 08/25/2018] [Indexed: 01/21/2023] Open
Abstract
Several studies showed that [11C]ABP688 binding is altered following drug-induced perturbation of glutamate levels in brains of humans, non-human primates and rats. We evaluated whether the fluorinated derivative [18F]PSS232 can be used to assess metabotropic glutamate receptor 5 (mGluR5) availability in rats after pharmacological challenge with ketamine, known to increase glutamate, or ceftriaxone, known to decrease glutamate. In vitro autoradiography was performed on rat brain slices with [18F]PSS232 to prove direct competition of the drugs for mGluR5. One group of rats were challenged with a bolus injection of either vehicle, racemic ketamine, S-ketamine or ceftriaxone followed by positron emission tomography PET imaging with [18F]PSS232. The other group received an infusion of the drugs during the PET scan. Distribution volume ratios (DVRs) were calculated using a reference tissue model. In vitro autoradiography showed no direct competition of the drugs with [18F]PSS232 for the allosteric binding site of mGluR5. DVRs of [18F]PSS232 binding in vivo did not change in any brain region neither after bolus injection nor after infusion. We conclude that [18F]PSS232 has utility for measuring mGluR5 density or occupancy of the allosteric site in vivo, but it cannot be used to measure in vivo fluctuations of glutamate levels in the rat brain.
Collapse
|
21
|
Masilamoni GJ, Smith Y. Metabotropic glutamate receptors: targets for neuroprotective therapies in Parkinson disease. Curr Opin Pharmacol 2018; 38:72-80. [PMID: 29605730 DOI: 10.1016/j.coph.2018.03.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 03/12/2018] [Indexed: 01/22/2023]
Abstract
Metabotropic glutamate receptors (mGluRs) are heavily expressed throughout the basal ganglia (BG), where they modulate neuronal excitability, transmitter release and long term synaptic plasticity. Therefore, targeting specific mGluR subtypes by means of selective drugs could be a possible strategy for restoring normal synaptic function and neuronal activity of the BG in Parkinson disease (PD). Preclinical studies have revealed that specific mGluR subtypes mediate significant neuroprotective effects that reduce toxin-induced midbrain dopaminergic neuronal death in animal models of PD. Although the underlying mechanisms of these effects must be further studied, there is evidence that intracellular calcium regulation, anti-inflammatory effects, and glutamatergic network modulation contribute to some of these neuroprotective properties. It is noteworthy that these protective effects extend beyond midbrain dopaminergic neurons to include other monoaminergic cell groups for some mGluRs. In this review, we discuss evidence for mGluR-mediated neuroprotection in PD and highlight the challenges to translate these findings into human trials.
Collapse
Affiliation(s)
- Gunasingh J Masilamoni
- Yerkes National Primate Research Center, Emory University, 954, Gatewood Rd NE, Atlanta, GA 30322, USA; Udall Center of Excellence for Parkinson's Disease, Emory University, 954, Gatewood Rd NE, Atlanta, GA 30322, USA
| | - Yoland Smith
- Yerkes National Primate Research Center, Emory University, 954, Gatewood Rd NE, Atlanta, GA 30322, USA; Department of Neurology, Emory University, 954, Gatewood Rd NE, Atlanta, GA 30322, USA; Udall Center of Excellence for Parkinson's Disease, Emory University, 954, Gatewood Rd NE, Atlanta, GA 30322, USA.
| |
Collapse
|
22
|
Involvement of mGluR I in EphB/ephrinB reverse signaling activation induced retinal ganglion cell apoptosis in a rat chronic hypertension model. Brain Res 2018; 1683:27-35. [PMID: 29366625 DOI: 10.1016/j.brainres.2018.01.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 01/05/2018] [Accepted: 01/17/2018] [Indexed: 02/08/2023]
Abstract
EphB/ephrinB reverse signaling is involved in retinal ganglion cell (RGC) apoptosis in experimental glaucoma. Here, we further investigated the mechanisms underlying EphB/ephrinB reverse signaling activation induced RGC apoptosis in a rat chronic ocular hypertension (COH) model, using patch-clamp techniques in retinal slices. In COH retinas, RGCs showed higher spontaneous firing frequency and much more depolarized membrane potential as compared to control, which was mimicked by intravitreally injection of EphB2-Fc, an activator of ephrinB2. The changes in RGC spontaneous firing and membrane potential could be reversed by the tyrosine kinase inhibitor PP2, suggesting that EphB/ephrinB reverse signaling activation induced RGC hyperexcitability. Intravitreal pre-injection of either LY367385 or MPEP, selective mGluR1 and mGluR5 antagonists, also blocked the changes in RGC spontaneous firing and membrane potential. Co-immunoprecipitation experiments showed an interaction between ephrinB2 and group I metabotropic glutamate receptor (mGluR I) (mGluR1/mGluR5). Furthermore, intravitreal pre-injection of the mixture of L-NAME (an NO synthase inhibitor) and XPro1595 (a selective inhibitor of soluble TNF-α) could reduce the EphB2-Fc injection induced increase in RGC firing, suggesting that Müller cells might be involved in EphB/ephrinB reverse signaling activation induced change in RGC hyperexcitability. In addition, LY367385/MPEP reduced the numbers of TUNEL-positive RGCs both in EphB2-Fc injected and COH retinas. All results suggest that activation of EphB/ephrinB reverse signaling induces RGC hyperexcitability and apoptosis by interacting with mGluR I in COH rats. Appropriate reduction of EphB/ephrinB reverse signaling could alleviate the loss of RGCs in glaucoma.
Collapse
|
23
|
Nicolini C, Fahnestock M. The valproic acid-induced rodent model of autism. Exp Neurol 2018; 299:217-227. [DOI: 10.1016/j.expneurol.2017.04.017] [Citation(s) in RCA: 235] [Impact Index Per Article: 33.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 04/26/2017] [Accepted: 04/28/2017] [Indexed: 01/12/2023]
|
24
|
Reker AN, Oliveros A, Sullivan JM, Nahar L, Hinton DJ, Kim T, Bruner RC, Choi DS, Goeders NE, Nam HW. Neurogranin in the nucleus accumbens regulates NMDA receptor tolerance and motivation for ethanol seeking. Neuropharmacology 2017; 131:58-67. [PMID: 29225043 DOI: 10.1016/j.neuropharm.2017.12.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2017] [Revised: 11/08/2017] [Accepted: 12/05/2017] [Indexed: 01/15/2023]
Abstract
Dysfunction of N-methyl-d-aspartate receptor (NMDAR) signaling in the nucleus accumbens (NAc) has been implicated in the pathophysiology of alcohol use disorders (AUD). Neurogranin (Ng), a calmodulin-binding protein, is exclusively expressed in the post-synapse, and mediates NMDAR driven synaptic plasticity by regulating the calcium-calmodulin (Ca2+-CaM) pathway. To study the functional role of Ng in AUD, we administrated behavior tests including Pavlovian instrument transfer (PIT), operant conditioning, and rotarod test using Ng null mice (Ng-/- mice). We used adeno-associated virus (AAV)-mediated Ng expression and pharmacological manipulation to validate behavioral responses in Ng-/- mice. The results from our multidisciplinary approaches demonstrated that deficit of Ng increases tolerance to NMDAR inhibition and elicit faster cue reactivity during PIT without changes in ethanol reward. Operant conditioning results demonstrated that Ng-/- mice self-administered significantly more ethanol and displayed reduced sensitivity to aversive motivation. We identified that ethanol exposure decreases mGluR5 (metabotropic glutamate receptor 5) expression in the NAc of Ng-/- mice and pharmacological inhibition of mGluR5 reverses NMDAR desensitization in Ng-/- mice. Together these findings specifically suggest that accumbal Ng plays an essential role in the counterbalance between NMDAR and mGluR5 signaling; which alters NMDAR resistance, and thereby altering aversive motivation for ethanol and may ultimately contribute to susceptibility for alcohol addiction.
Collapse
Affiliation(s)
- Ashlie N Reker
- Department of Pharmacology, Toxicology, and Neuroscience, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA
| | - Alfredo Oliveros
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - John M Sullivan
- Department of Pharmacology, Toxicology, and Neuroscience, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA
| | - Lailun Nahar
- Department of Pharmacology, Toxicology, and Neuroscience, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA
| | - David J Hinton
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Taehyun Kim
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Robert C Bruner
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Doo-Sup Choi
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Nicholas E Goeders
- Department of Pharmacology, Toxicology, and Neuroscience, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA
| | - Hyung W Nam
- Department of Pharmacology, Toxicology, and Neuroscience, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA.
| |
Collapse
|
25
|
Di Menna L, Joffe ME, Iacovelli L, Orlando R, Lindsley CW, Mairesse J, Gressèns P, Cannella M, Caraci F, Copani A, Bruno V, Battaglia G, Conn PJ, Nicoletti F. Functional partnership between mGlu3 and mGlu5 metabotropic glutamate receptors in the central nervous system. Neuropharmacology 2017; 128:301-313. [PMID: 29079293 DOI: 10.1016/j.neuropharm.2017.10.026] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 10/09/2017] [Accepted: 10/21/2017] [Indexed: 12/19/2022]
Abstract
mGlu5 receptors are involved in mechanisms of activity-dependent synaptic plasticity, and are targeted by drugs developed for the treatment of CNS disorders. We report that mGlu3 receptors, which are traditionally linked to the control of neurotransmitter release, support mGlu5 receptor signaling in neurons and largely contribute to the robust mGlu5 receptor-mediated polyphosphoinositide hydrolysis in the early postnatal life. In cortical pyramidal neurons, mGlu3 receptor activation potentiated mGlu5 receptor-mediated somatic Ca2+ mobilization, and mGlu3 receptor-mediated long-term depression in the prefrontal cortex required the endogenous activation of mGlu5 receptors. The interaction between mGlu3 and mGlu5 receptors was also relevant to mechanisms of neuronal toxicity, with mGlu3 receptors shaping the influence of mGlu5 receptors on excitotoxic neuronal death. These findings shed new light into the complex role played by mGlu receptors in physiology and pathology, and suggest reconsideration of some of the current dogmas in the mGlu receptor field.
Collapse
Affiliation(s)
| | - Max E Joffe
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN 37232-0697, USA
| | - Luisa Iacovelli
- Department of Physiology and Pharmacology, University Sapienza of Roma, 00185 Roma, Italy
| | - Rosamaria Orlando
- Department of Physiology and Pharmacology, University Sapienza of Roma, 00185 Roma, Italy
| | - Craig W Lindsley
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN 37232-0697, USA
| | - Jèrome Mairesse
- PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, 1141 Paris, France
| | - Pierre Gressèns
- PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, 1141 Paris, France; Centre for the Developing Brain, Department of Perinatal Health and Imaging, Division of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners, St. Thomas' Hospital, London SE1 7EH, United Kingdom
| | | | - Filippo Caraci
- Department of Drug Sciences, University of Catania, 95125 Catania, Italy; I.R.C.C.S. Oasi Maria SS, 94018 Troina, Italy
| | - Agata Copani
- Department of Drug Sciences, University of Catania, 95125 Catania, Italy; Institute of Biostructure and Bioimaging, National Research Council, 95126 Catania, Italy
| | - Valeria Bruno
- I.R.C.C.S. Neuromed, 86077 Pozzilli, Italy; Department of Physiology and Pharmacology, University Sapienza of Roma, 00185 Roma, Italy
| | | | - P Jeffrey Conn
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN 37232-0697, USA
| | - Ferdinando Nicoletti
- I.R.C.C.S. Neuromed, 86077 Pozzilli, Italy; Department of Physiology and Pharmacology, University Sapienza of Roma, 00185 Roma, Italy.
| |
Collapse
|
26
|
Lin S, Li X, Chen YH, Gao F, Chen H, Hu NY, Huang L, Luo ZY, Liu JH, You QL, Yin YN, Li ZL, Li XW, Du ZJ, Yang JM, Gao TM. Social Isolation During Adolescence Induces Anxiety Behaviors and Enhances Firing Activity in BLA Pyramidal Neurons via mGluR5 Upregulation. Mol Neurobiol 2017; 55:5310-5320. [PMID: 28914419 DOI: 10.1007/s12035-017-0766-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 09/06/2017] [Indexed: 11/28/2022]
Abstract
Social isolation during the vulnerable period of adolescence contributes to the occurrence of psychiatric disorders and profoundly affects brain development and adult behavior. Although the impact of social isolation during adolescence on anxiety behaviors has been well studied, much less is known about the onset and underlying mechanisms of these behaviors. We observed that following 2 weeks, but not 1 week, of social isolation, adolescent mice exhibited anxiety behaviors. Strikingly, the mGluR5 protein levels in the amygdala increased concomitantly with anxiety behaviors, and both intraperitoneal administration and intra-basolateral amygdala (BLA) infusion of MPEP, a metabotropic glutamate receptor 5 antagonist, normalized anxiety behaviors. Furthermore, electrophysiological studies showed that 2 weeks of social isolation during adolescence facilitated pyramidal neuronal excitability in the BLA, which could be normalized by MPEP. Together, these results reveal a critical period in adolescence during which social isolation can induce anxiety behaviors and facilitate BLA pyramidal neuronal excitability, both of which are mediated by mGluR5, thus providing mechanistic insights into the onset of anxiety behaviors after social isolation during adolescence.
Collapse
Affiliation(s)
- Song Lin
- State Key Laboratory of Organ Failure Research, Key Laboratory of Psychiatric Disorders of Guangdong Province, Collaborative Innovation Center for Brain Science, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, 1023S Shatai Road, Guangzhou, 510515, China
| | - Xin Li
- State Key Laboratory of Organ Failure Research, Key Laboratory of Psychiatric Disorders of Guangdong Province, Collaborative Innovation Center for Brain Science, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, 1023S Shatai Road, Guangzhou, 510515, China
| | - Yi-Hua Chen
- State Key Laboratory of Organ Failure Research, Key Laboratory of Psychiatric Disorders of Guangdong Province, Collaborative Innovation Center for Brain Science, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, 1023S Shatai Road, Guangzhou, 510515, China
| | - Feng Gao
- State Key Laboratory of Organ Failure Research, Key Laboratory of Psychiatric Disorders of Guangdong Province, Collaborative Innovation Center for Brain Science, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, 1023S Shatai Road, Guangzhou, 510515, China
| | - Hao Chen
- State Key Laboratory of Organ Failure Research, Key Laboratory of Psychiatric Disorders of Guangdong Province, Collaborative Innovation Center for Brain Science, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, 1023S Shatai Road, Guangzhou, 510515, China
| | - Neng-Yuan Hu
- State Key Laboratory of Organ Failure Research, Key Laboratory of Psychiatric Disorders of Guangdong Province, Collaborative Innovation Center for Brain Science, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, 1023S Shatai Road, Guangzhou, 510515, China
| | - Lang Huang
- State Key Laboratory of Organ Failure Research, Key Laboratory of Psychiatric Disorders of Guangdong Province, Collaborative Innovation Center for Brain Science, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, 1023S Shatai Road, Guangzhou, 510515, China
| | - Zheng-Yi Luo
- State Key Laboratory of Organ Failure Research, Key Laboratory of Psychiatric Disorders of Guangdong Province, Collaborative Innovation Center for Brain Science, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, 1023S Shatai Road, Guangzhou, 510515, China
| | - Ji-Hong Liu
- State Key Laboratory of Organ Failure Research, Key Laboratory of Psychiatric Disorders of Guangdong Province, Collaborative Innovation Center for Brain Science, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, 1023S Shatai Road, Guangzhou, 510515, China
| | - Qiang-Long You
- State Key Laboratory of Organ Failure Research, Key Laboratory of Psychiatric Disorders of Guangdong Province, Collaborative Innovation Center for Brain Science, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, 1023S Shatai Road, Guangzhou, 510515, China
| | - Ya-Nan Yin
- State Key Laboratory of Organ Failure Research, Key Laboratory of Psychiatric Disorders of Guangdong Province, Collaborative Innovation Center for Brain Science, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, 1023S Shatai Road, Guangzhou, 510515, China
| | - Ze-Lin Li
- State Key Laboratory of Organ Failure Research, Key Laboratory of Psychiatric Disorders of Guangdong Province, Collaborative Innovation Center for Brain Science, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, 1023S Shatai Road, Guangzhou, 510515, China
| | - Xiao-Wen Li
- State Key Laboratory of Organ Failure Research, Key Laboratory of Psychiatric Disorders of Guangdong Province, Collaborative Innovation Center for Brain Science, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, 1023S Shatai Road, Guangzhou, 510515, China
| | - Zhuo-Jun Du
- State Key Laboratory of Organ Failure Research, Key Laboratory of Psychiatric Disorders of Guangdong Province, Collaborative Innovation Center for Brain Science, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, 1023S Shatai Road, Guangzhou, 510515, China
| | - Jian-Ming Yang
- State Key Laboratory of Organ Failure Research, Key Laboratory of Psychiatric Disorders of Guangdong Province, Collaborative Innovation Center for Brain Science, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, 1023S Shatai Road, Guangzhou, 510515, China.
| | - Tian-Ming Gao
- State Key Laboratory of Organ Failure Research, Key Laboratory of Psychiatric Disorders of Guangdong Province, Collaborative Innovation Center for Brain Science, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, 1023S Shatai Road, Guangzhou, 510515, China.
| |
Collapse
|
27
|
Dysregulated NMDA-Receptor Signaling Inhibits Long-Term Depression in a Mouse Model of Fragile X Syndrome. J Neurosci 2017; 36:9817-27. [PMID: 27656021 DOI: 10.1523/jneurosci.3038-15.2016] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 08/05/2016] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED Fragile X syndrome (FXS) is a neurodevelopmental disease. It is one of the leading monogenic causes of intellectual disability among boys with most also displaying autism spectrum disorder traits. Here we investigated the role of NMDA receptors on mGluR-dependent long-term depression (mGluR-LTD), a key biomarker in the disease, at four different developmental stages. First, we applied the mGluR agonist 3,5-dihydroxyphenylglycine in the absence or presence of the NMDAR blocker, APV, hereby unmasking the NMDAR component in this process. As expected, in the presence of APV, we found more LTD in the mouse KO than in WT. This, however, was only observed in the p30-60 age group. At all other age groups tested, mGluR-LTD was almost identical between KO and WT. Interestingly, at p60, in the absence of APV, no or very little LTD was found in KO that was completely restored by application of APV. This suggests that the underlying cause of the enhanced mGluR-LTD in KO (at p30) is caused by dysregulated NMDAR signaling. To investigate this further, we next used NMDAR-subunit-specific antagonists. Inhibition of GluN2B, but not GluN2A, blocked mGluR-LTD only in WT. This was in contrast in the KO where blocking GluN2B rescued mGluR-LTD, suggesting GluN2B-containing NMDARs in the KO are hyperactive. Thus, these findings suggest strong involvement of GluN2B-containing-NMDARs in the pathophysiology of FXS and highlight a potential path for treatment for the disease. SIGNIFICANCE STATEMENT There is currently no cure for fragile X, although medications targeting specific FXS symptoms do exist. The FXS animal model, the Fmr1 knock-out mouse, has demonstrated an increased mGluR5-mediated long-term depression (LTD) leading to several clinical trials of mGluR5 inhibitors/modulators, yet all have failed. In addition, surprisingly little information exists about the possible role of other ion channels/receptors, including NMDA receptors (NMDAR), in mGluR-LTD. Here we focus on NMDARs and their regulation of mGluR-mediated LTD at different developmental stages using several different NMDAR blockers/antagonists. Our findings suggest dysregulated NMDARs in the pathophysiology of FXS leading to altered mGluR-mediated LTD. Together, these data will help to develop new drug candidates that could lead to reversal of the FXS phenotype.
Collapse
|
28
|
Lee E, Lee J, Kim E. Excitation/Inhibition Imbalance in Animal Models of Autism Spectrum Disorders. Biol Psychiatry 2017; 81:838-847. [PMID: 27450033 DOI: 10.1016/j.biopsych.2016.05.011] [Citation(s) in RCA: 326] [Impact Index Per Article: 40.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 05/02/2016] [Accepted: 05/16/2016] [Indexed: 12/19/2022]
Abstract
Imbalances between excitation and inhibition in synaptic transmission and neural circuits have been implicated in autism spectrum disorders. Excitation and inhibition imbalances are frequently observed in animal models of autism spectrum disorders, and their correction normalizes key autistic-like phenotypes in these animals. These results suggest that excitation and inhibition imbalances may contribute to the development and maintenance of autism spectrum disorders and represent an important therapeutic target.
Collapse
Affiliation(s)
- Eunee Lee
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science, Daejeon, Korea
| | - Jiseok Lee
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Korea
| | - Eunjoon Kim
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science, Daejeon, Korea; Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Korea.
| |
Collapse
|
29
|
Huang Y, Todd N, Thathiah A. The role of GPCRs in neurodegenerative diseases: avenues for therapeutic intervention. Curr Opin Pharmacol 2017; 32:96-110. [DOI: 10.1016/j.coph.2017.02.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 02/08/2017] [Accepted: 02/09/2017] [Indexed: 12/20/2022]
|
30
|
Metabotropic glutamate receptors and neurodegenerative diseases. Pharmacol Res 2017; 115:179-191. [DOI: 10.1016/j.phrs.2016.11.013] [Citation(s) in RCA: 145] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 11/11/2016] [Accepted: 11/15/2016] [Indexed: 12/21/2022]
|
31
|
Bostrom C, Yau SY, Majaess N, Vetrici M, Gil-Mohapel J, Christie BR. Hippocampal dysfunction and cognitive impairment in Fragile-X Syndrome. Neurosci Biobehav Rev 2016; 68:563-574. [DOI: 10.1016/j.neubiorev.2016.06.033] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 06/21/2016] [Accepted: 06/22/2016] [Indexed: 01/03/2023]
|
32
|
Sun Y, Lipton JO, Boyle LM, Madsen JR, Goldenberg MC, Pascual-Leone A, Sahin M, Rotenberg A. Direct current stimulation induces mGluR5-dependent neocortical plasticity. Ann Neurol 2016; 80:233-46. [PMID: 27315032 DOI: 10.1002/ana.24708] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 06/07/2016] [Accepted: 06/08/2016] [Indexed: 01/07/2023]
Abstract
OBJECTIVE To obtain insights into mechanisms mediating changes in cortical excitability induced by cathodal transcranial direct current stimulation (tDCS). METHODS Neocortical slices were exposed to direct current stimulation (DCS) delivered through Ag/AgCl electrodes over a range of current orientations, magnitudes, and durations. DCS-induced cortical plasticity and its receptor dependency were measured as the change in layer II/III field excitatory postsynaptic potentials by a multielectrode array, both with and without neurotransmitter receptor blockers or allosteric modulators. In vivo, tDCS was delivered to intact mice scalp via surface electrodes. Molecular consequences of DCS in vitro or tDCS in vivo were tested by immunoblot of protein extracted from stimulated slices or the neocortex harvested from stimulated intact mice. RESULTS Cathodal DCS in vitro induces a long-term depression (DCS-LTD) of excitatory synaptic strength in both human and mouse neocortical slices. DCS-LTD is abolished with an mGluR5 negative allosteric modulator, mechanistic target of rapamycin (mTOR) inhibitor, and inhibitor of protein synthesis. However, DCS-LTD persists despite either γ-aminobutyric acid type A receptor or N-methyl-D-aspartate receptor inhibition. An mGluR5-positive allosteric modulator, in contrast, transformed transient synaptic depression resultant from brief DCS application into durable DCS-LTD. INTERPRETATION We identify a novel molecular pathway by which tDCS modulates cortical excitability, and indicate a capacity for synergistic interaction between tDCS and pharmacologic mGluR5 facilitation. The findings support exploration of cathodal tDCS as a treatment of neurologic conditions characterized by aberrant regional cortical excitability referable to mGluR5-mTOR signaling. Ann Neurol 2016;80:233-246.
Collapse
Affiliation(s)
- Yan Sun
- Department of Neurology and F. M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA.,Neuromodulation Program, Boston Children's Hospital, Boston, MA.,Program in Neuroscience, Harvard Medical School, Boston, MA
| | - Jonathan O Lipton
- Department of Neurology and F. M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA.,Division of Sleep Medicine, Harvard Medical School, Boston, MA
| | - Lara M Boyle
- Department of Neurology and F. M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA
| | - Joseph R Madsen
- Department of Neurosurgery, Boston Children's Hospital, Boston, MA
| | - Marti C Goldenberg
- Department of Neurology and F. M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA
| | - Alvaro Pascual-Leone
- Berenson-Allen Center for Noninvasive Brain Stimulation, Division of Cognitive Neurology, Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Mustafa Sahin
- Department of Neurology and F. M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA
| | - Alexander Rotenberg
- Department of Neurology and F. M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA.,Neuromodulation Program, Boston Children's Hospital, Boston, MA.,Berenson-Allen Center for Noninvasive Brain Stimulation, Division of Cognitive Neurology, Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| |
Collapse
|
33
|
Agnati LF, Marcoli M, Maura G, Fuxe K, Guidolin D. The multi-facet aspects of cell sentience and their relevance for the integrative brain actions: role of membrane protein energy landscape. Rev Neurosci 2016; 27:347-63. [DOI: 10.1515/revneuro-2015-0049] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 11/11/2015] [Indexed: 12/14/2022]
Abstract
AbstractSeveral ion channels can be randomly and spontaneously in an open state, allowing the exchange of ion fluxes between extracellular and intracellular environments. We propose that the random changes in the state of ion channels could be also due to proteins exploring their energy landscapes. Indeed, proteins can modify their steric conformation under the effects of the physicochemical parameters of the environments with which they are in contact, namely, the extracellular, intramembrane and intracellular environments. In particular, it is proposed that the random walk of proteins in their energy landscape is towards attractors that can favor the open or close condition of the ion channels and/or intrinsic activity of G-protein-coupled receptors. The main aspect of the present proposal is that some relevant physicochemical parameters of the environments (e.g. molecular composition, temperature, electrical fields) with which some signaling-involved plasma membrane proteins are in contact alter their conformations. In turn, these changes can modify their information handling via a modulatory action on their random walk towards suitable attractors of their energy landscape. Thus, spontaneous and/or signal-triggered electrical activities of neurons occur that can have emergent properties capable of influencing the integrative actions of brain networks. Against this background, Cook’s hypothesis on ‘cell sentience’ is developed by proposing that physicochemical parameters of the environments with which the plasma-membrane proteins of complex cellular networks are in contact fulfill a fundamental role in their spontaneous and/or signal-triggered activity. Furthermore, it is proposed that a specialized organelle, the primary cilium, which is present in most cells (also neurons and astrocytes), could be of peculiar importance to pick up chemical signals such as ions and transmitters and to detect physical signals such as pressure waves, thermal gradients, and local field potentials.
Collapse
Affiliation(s)
| | - Manuela Marcoli
- 3University of Genova, Department of Pharmacy and Center of Excellence for Biomedical Research, Viale Cembrano 4, I-16148 Genova, Italy
| | - Guido Maura
- 3University of Genova, Department of Pharmacy and Center of Excellence for Biomedical Research, Viale Cembrano 4, I-16148 Genova, Italy
| | - Kjell Fuxe
- 2Karolinska Institutet, Department of Neuroscience, S-17177 Stockholm, Sweden
| | - Diego Guidolin
- 4University of Padova, Department of Molecular Medicine, I-35122 Padova, Italy
| |
Collapse
|
34
|
Shimoda Y, Yamasaki T, Fujinaga M, Ogawa M, Kurihara Y, Nengaki N, Kumata K, Yui J, Hatori A, Xie L, Zhang Y, Kawamura K, Zhang MR. Synthesis and Evaluation of Novel Radioligands Based on 3-[5-(Pyridin-2-yl)-2H-tetrazol-2-yl]benzonitrile for Positron Emission Tomography Imaging of Metabotropic Glutamate Receptor Subtype 5. J Med Chem 2016; 59:3980-90. [DOI: 10.1021/acs.jmedchem.6b00209] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Yoko Shimoda
- Department
of Radiopharmaceuticals Development, Radiological Science Research
and Development Directorate, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Tomoteru Yamasaki
- Department
of Radiopharmaceuticals Development, Radiological Science Research
and Development Directorate, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Masayuki Fujinaga
- Department
of Radiopharmaceuticals Development, Radiological Science Research
and Development Directorate, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Masanao Ogawa
- Department
of Radiopharmaceuticals Development, Radiological Science Research
and Development Directorate, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
- SHI Accelerator
Service Co. Ltd., 1-17-6 Osaki, Shinagawa-ku, Tokyo 141-0032, Japan
| | - Yusuke Kurihara
- Department
of Radiopharmaceuticals Development, Radiological Science Research
and Development Directorate, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
- SHI Accelerator
Service Co. Ltd., 1-17-6 Osaki, Shinagawa-ku, Tokyo 141-0032, Japan
| | - Nobuki Nengaki
- Department
of Radiopharmaceuticals Development, Radiological Science Research
and Development Directorate, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
- SHI Accelerator
Service Co. Ltd., 1-17-6 Osaki, Shinagawa-ku, Tokyo 141-0032, Japan
| | - Katsushi Kumata
- Department
of Radiopharmaceuticals Development, Radiological Science Research
and Development Directorate, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Joji Yui
- Department
of Radiopharmaceuticals Development, Radiological Science Research
and Development Directorate, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Akiko Hatori
- Department
of Radiopharmaceuticals Development, Radiological Science Research
and Development Directorate, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Lin Xie
- Department
of Radiopharmaceuticals Development, Radiological Science Research
and Development Directorate, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Yiding Zhang
- Department
of Radiopharmaceuticals Development, Radiological Science Research
and Development Directorate, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Kazunori Kawamura
- Department
of Radiopharmaceuticals Development, Radiological Science Research
and Development Directorate, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Ming-Rong Zhang
- Department
of Radiopharmaceuticals Development, Radiological Science Research
and Development Directorate, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| |
Collapse
|
35
|
Borroto-Escuela DO, Pintsuk J, Schäfer T, Friedland K, Ferraro L, Tanganelli S, Liu F, Fuxe K. Multiple D2 heteroreceptor complexes: new targets for treatment of schizophrenia. Ther Adv Psychopharmacol 2016; 6:77-94. [PMID: 27141290 PMCID: PMC4837969 DOI: 10.1177/2045125316637570] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The dopamine (DA) neuron system most relevant for schizophrenia is the meso-limbic-cortical DA system inter alia densely innervating subcortical limbic regions. The field of dopamine D2 receptors and schizophrenia changed markedly with the discovery of many types of D2 heteroreceptor complexes in subcortical limbic areas as well as the dorsal striatum. The results indicate that the D2 is a hub receptor which interacts not only with many other G protein-coupled receptors (GPCRs) including DA isoreceptors but also with ion-channel receptors, receptor tyrosine kinases, scaffolding proteins and DA transporters. Disturbances in several of these D2 heteroreceptor complexes may contribute to the development of schizophrenia through changes in the balance of diverse D2 homo- and heteroreceptor complexes mediating the DA signal, especially to the ventral striato-pallidal γ-aminobutyric acid (GABA) pathway. This will have consequences for the control of this pathway of the glutamate drive to the prefrontal cortex via the mediodorsal thalamic nucleus which can contribute to psychotic processes. Agonist activation of the A2A protomer in the A2A-D2 heteroreceptor complex inhibits D2 Gi/o mediated signaling but increases the D2 β-arrestin2 mediated signaling. Through this allosteric receptor-receptor interaction, the A2A agonist becomes a biased inhibitory modulator of the Gi/o mediated D2 signaling, which may the main mechanism for its atypical antipsychotic properties especially linked to the limbic A2A-D2 heterocomplexes. The DA and glutamate hypotheses of schizophrenia come together in the signal integration in D2-N-methyl-d-aspartate (NMDA) and A2A-D2-metabotropic glutamate receptor 5 (mGlu5) heteroreceptor complexes, especially in the ventral striatum. 5-Hydroxytryptamine 2A (5-HT2A)-D2 heteroreceptor complexes are special targets for atypical antipsychotics with high potency to block their 5-HT2A protomer signaling in view of the potential development of pathological allosteric facilitatory 5-HT2A-D2 interaction increasing D2 protomer signaling. Neurotensin (NTS1)-D2 heterocomplexes also exist in the ventral and dorsal striatum, and likely also in midbrain DA nerve cells as NTS1-D2 autoreceptor complexes where neurotensin produces antipsychotic and propsychotic actions, respectively.
Collapse
Affiliation(s)
- Dasiel O. Borroto-Escuela
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden Department of Biomolecular Science, Section of Physiology, University of Urbino, Italy
| | - Julia Pintsuk
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden Institute of Biomedicine and Translational Medicine, University of Tartu, Estonia
| | - Thorsten Schäfer
- Clinical and Molecular Pharmacy, Department of Chemistry and Pharmacy, Friedrich Alexander University, Erlangen-Nürnberg, Germany
| | - Kristina Friedland
- Clinical and Molecular Pharmacy, Department of Chemistry and Pharmacy, Friedrich Alexander University, Erlangen-Nürnberg, Germany
| | - Luca Ferraro
- Department of Life Sciences and Biotechnology, University of Ferrara, Italy
| | - Sergio Tanganelli
- Department of Life Sciences and Biotechnology, University of Ferrara, Italy Department of Medical Sciences, University of Ferrara, Italy
| | - Fang Liu
- Campbell Research Institute, Centre for Addiction and Mental Health, University of Toronto, Toronto, Ontario, Canada
| | | |
Collapse
|
36
|
Guntupalli S, Widagdo J, Anggono V. Amyloid-β-Induced Dysregulation of AMPA Receptor Trafficking. Neural Plast 2016; 2016:3204519. [PMID: 27073700 PMCID: PMC4814684 DOI: 10.1155/2016/3204519] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 02/28/2016] [Indexed: 01/22/2023] Open
Abstract
Evidence from neuropathological, genetic, animal model, and biochemical studies has indicated that the accumulation of amyloid-beta (Aβ) is associated with, and probably induces, profound neuronal changes in brain regions critical for memory and cognition in the development of Alzheimer's disease (AD). There is considerable evidence that synapses are particularly vulnerable to AD, establishing synaptic dysfunction as one of the earliest events in pathogenesis, prior to neuronal loss. It is clear that excessive Aβ levels can disrupt excitatory synaptic transmission and plasticity, mainly due to dysregulation of the AMPA and NMDA glutamate receptors in the brain. Importantly, AMPA receptors are the principal glutamate receptors that mediate fast excitatory neurotransmission. This is essential for synaptic plasticity, a cellular correlate of learning and memory, which are the cognitive functions that are most disrupted in AD. Here we review recent advances in the field and provide insights into the molecular mechanisms that underlie Aβ-induced dysfunction of AMPA receptor trafficking. This review focuses primarily on NMDA receptor- and metabotropic glutamate receptor-mediated signaling. In particular, we highlight several mechanisms that underlie synaptic long-term depression as common signaling pathways that are hijacked by the neurotoxic effects of Aβ.
Collapse
Affiliation(s)
- Sumasri Guntupalli
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Jocelyn Widagdo
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Victor Anggono
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| |
Collapse
|
37
|
Zolkowska D, Kondrat-Wrobel MW, Florek-Luszczki M, Luszczki JJ. Influence of MPEP (a selective mGluR5 antagonist) on the anticonvulsant action of novel antiepileptic drugs against maximal electroshock-induced seizures in mice. Prog Neuropsychopharmacol Biol Psychiatry 2016; 65:172-8. [PMID: 26478256 DOI: 10.1016/j.pnpbp.2015.10.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 10/12/2015] [Accepted: 10/13/2015] [Indexed: 11/25/2022]
Abstract
The aim of this study was to determine the effects of 2-methyl-6-(phenylethynyl)pyridine (MPEP - a selective antagonist for the glutamate metabotropic receptor subtype mGluR5) on the protective action of some novel antiepileptic drugs (lamotrigine, oxcarbazepine, pregabalin and topiramate) against maximal electroshock-induced seizures in mice. Brain concentrations of antiepileptic drugs were measured to determine whether MPEP altered pharmacokinetics of antiepileptic drugs. Intraperitoneal injection of 1.5 and 2mg/kg of MPEP significantly elevated the threshold for electroconvulsions in mice, whereas MPEP at a dose of 1mg/kg considerably enhanced the anticonvulsant activity of pregabalin and topiramate, but not that of lamotrigine or oxcarbazepine in the maximal electroshock-induced seizures in mice. Pharmacokinetic results revealed that MPEP (1mg/kg) did not alter total brain concentrations of pregabalin and topiramate, and the observed effect in the mouse maximal electroshock seizure model was pharmacodynamic in nature. Collectively, our preclinical data suggest that MPEP may be a safe and beneficial adjunct to the therapeutic effects of antiepileptic drugs in human patients.
Collapse
Affiliation(s)
- Dorota Zolkowska
- Department of Neurology, School of Medicine, University of California-Davis, Sacramento, CA, USA
| | | | | | - Jarogniew J Luszczki
- Department of Pathophysiology, Medical University of Lublin, Lublin, Poland; Isobolographic Analysis Laboratory, Institute of Rural Health, Lublin, Poland.
| |
Collapse
|
38
|
Wierońska JM, Zorn SH, Doller D, Pilc A. Metabotropic glutamate receptors as targets for new antipsychotic drugs: Historical perspective and critical comparative assessment. Pharmacol Ther 2015; 157:10-27. [PMID: 26549541 DOI: 10.1016/j.pharmthera.2015.10.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
In this review, we aim to present, discuss and clarify our current understanding regarding the prediction of possible antipsychotic effects of metabotropic glutamate (mGlu) receptor ligands. The number of preclinical trials clearly indicates, that this group of compounds constitutes an excellent alternative to presently used antipsychotic therapy, being effective not only to positive, but also negative and cognitive symptoms of schizophrenia. Although the results of clinical trials that were performed for the group of mGlu2/3 agonists were not so enthusiastic as in animal studies, they still showed that mGlu ligands do not induced variety of side effects typical for presently used antipsychotics, and were generally well tolerated. The lack of satisfactory effectiveness towards schizophrenia symptoms of mGlu2/3 activators in humans could be a result of variety of uncontrolled factors and unidentified biomarkers different for each schizophrenia patient, that should be taken into consideration in the future set of clinical trials. The subject is still open for further research, and the novel classes of mGlu5 or mGlu2/3 agonists/PAMs were recently introduced, including the large group of compounds from the third group of mGlu receptors, especially of mGlu4 subtype. Finally, more precise treatment based on simultaneous administration of minimal doses of the ligands for two or more receptors, seems to be promising in the context of symptoms-specific schizophrenia treatment.
Collapse
Affiliation(s)
- Joanna M Wierońska
- Institute of Pharmacology, Polish Academy of Sciences, 31-343 Krakow, Poland
| | | | | | - Andrzej Pilc
- Institute of Pharmacology, Polish Academy of Sciences, 31-343 Krakow, Poland.
| |
Collapse
|
39
|
Ellard JM, Madin A, Philps O, Hopkin M, Henderson S, Birch L, O'Connor D, Arai T, Takase K, Morgan L, Reynolds D, Talma S, Howley E, Powney B, Payne AH, Hall A, Gartlon JE, Dawson LA, Castro L, Atkinson PJ. Identification and optimisation of a series of tetrahydrobenzotriazoles as metabotropic glutamate receptor 5-selective positive allosteric modulators that improve performance in a preclinical model of cognition. Bioorg Med Chem Lett 2015; 25:5792-6. [PMID: 26531152 DOI: 10.1016/j.bmcl.2015.10.050] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Revised: 10/14/2015] [Accepted: 10/16/2015] [Indexed: 11/17/2022]
Abstract
Herein we describe a series of tetrahydrobenzotriazoles as novel, potent metabotropic glutamate receptor subtype 5 (mGlu5) positive allosteric modulators (PAMs). Exploration of the SAR surrounding the tetrahydrobenzotriazole core ultimately led to the identification of 29 as a potent mGlu5 PAM with a low maximal glutamate potency fold shift, acceptable in vitro DMPK parameters and in vivo PK profile and efficacy in the rat novel object recognition (NOR) assay. As a result 29 was identified as a suitable compound for progression to in vivo safety evaluation.
Collapse
Affiliation(s)
- John M Ellard
- Medicinal Chemistry, Neuroscience Product Creation Unit, Eisai Limited, European Knowledge Centre, Mosquito Way, Hatfield, Hertfordshire AL10 9SN, UK
| | - Andrew Madin
- Medicinal Chemistry, Neuroscience Product Creation Unit, Eisai Limited, European Knowledge Centre, Mosquito Way, Hatfield, Hertfordshire AL10 9SN, UK
| | - Oliver Philps
- Medicinal Chemistry, Neuroscience Product Creation Unit, Eisai Limited, European Knowledge Centre, Mosquito Way, Hatfield, Hertfordshire AL10 9SN, UK
| | - Mark Hopkin
- Medicinal Chemistry, Neuroscience Product Creation Unit, Eisai Limited, European Knowledge Centre, Mosquito Way, Hatfield, Hertfordshire AL10 9SN, UK
| | - Scott Henderson
- Medicinal Chemistry, Neuroscience Product Creation Unit, Eisai Limited, European Knowledge Centre, Mosquito Way, Hatfield, Hertfordshire AL10 9SN, UK
| | - Louise Birch
- Medicinal Chemistry, Neuroscience Product Creation Unit, Eisai Limited, European Knowledge Centre, Mosquito Way, Hatfield, Hertfordshire AL10 9SN, UK
| | - Desmond O'Connor
- DMPK, Neuroscience Product Creation Unit, Eisai Limited, European Knowledge Centre, Mosquito Way, Hatfield, Hertfordshire AL10 9SN, UK
| | - Tohru Arai
- Next Generation Systems Core Function Unit, Eisai Product Creation Systems, Eisai Co., Ltd, 5-1-3 Tokodai, Tsukuba, Ibaraki 300-2635 Japan
| | - Kazuma Takase
- Biomarker and Personalized Medicine Core Function Unit, Eisai Product Creation Systems, Eisai Co., Ltd, 5-1-3 Tokodai, Tsukuba, Ibaraki 300-2635 Japan
| | - Louise Morgan
- Pharmacology, Neuroscience Product Creation Unit, Eisai Limited, European Knowledge Centre, Mosquito Way, Hatfield, Hertfordshire AL10 9SN, UK
| | - David Reynolds
- Pharmacology, Neuroscience Product Creation Unit, Eisai Limited, European Knowledge Centre, Mosquito Way, Hatfield, Hertfordshire AL10 9SN, UK
| | - Sonia Talma
- Pharmacology, Neuroscience Product Creation Unit, Eisai Limited, European Knowledge Centre, Mosquito Way, Hatfield, Hertfordshire AL10 9SN, UK
| | - Eimear Howley
- Pharmacology, Neuroscience Product Creation Unit, Eisai Limited, European Knowledge Centre, Mosquito Way, Hatfield, Hertfordshire AL10 9SN, UK
| | - Ben Powney
- Pharmacology, Neuroscience Product Creation Unit, Eisai Limited, European Knowledge Centre, Mosquito Way, Hatfield, Hertfordshire AL10 9SN, UK
| | - Andrew H Payne
- Medicinal Chemistry, Neuroscience Product Creation Unit, Eisai Limited, European Knowledge Centre, Mosquito Way, Hatfield, Hertfordshire AL10 9SN, UK
| | - Adrian Hall
- Medicinal Chemistry, Neuroscience Product Creation Unit, Eisai Limited, European Knowledge Centre, Mosquito Way, Hatfield, Hertfordshire AL10 9SN, UK
| | - Jane E Gartlon
- Pharmacology, Neuroscience Product Creation Unit, Eisai Limited, European Knowledge Centre, Mosquito Way, Hatfield, Hertfordshire AL10 9SN, UK
| | - Lee A Dawson
- Pharmacology, Neuroscience Product Creation Unit, Eisai Limited, European Knowledge Centre, Mosquito Way, Hatfield, Hertfordshire AL10 9SN, UK
| | - Luis Castro
- Medicinal Chemistry, Neuroscience Product Creation Unit, Eisai Limited, European Knowledge Centre, Mosquito Way, Hatfield, Hertfordshire AL10 9SN, UK
| | - Peter J Atkinson
- Pharmacology, Neuroscience Product Creation Unit, Eisai Limited, European Knowledge Centre, Mosquito Way, Hatfield, Hertfordshire AL10 9SN, UK.
| |
Collapse
|
40
|
Abstract
All currently approved antidepressant medications for major depressive disorder (MDD) and bipolar disorder act primarily on the monoaminergic system and have varying affinities for serotonergic, norepinephrine-ergic, and/or dopaminergic receptors. Unfortunately, these drugs are only effective in approximately two-thirds of patients. Glutamate is the major excitatory neurotransmitter in the central nervous system, and the glutamatergic system has been implicated in the pathophysiology of MDD. Here, we review the putative involvement of the glutamate receptor subtypes-N-methyl-D-aspartate (NMDA), α-amino-3-hydroxyl-5-methyl-4-isoxazoleproprionic acid (AMPA), kainate, and the group I, II, and III metabotropic glutamate receptors (mGluRs)-in the development of novel and more effective treatments for MDD as well as preclinical and clinical trials of drugs targeting these receptors. The rapid and robust antidepressant effects of ketamine-an NMDA receptor antagonist-have been consistently replicated in multiple trials. Other glutamatergic drugs have been investigated with varying success. Here, we highlight some of the most interesting results, including: 1) repeated oral, intramuscular, and sublingual ketamine appears to be less robustly effective than intravenous ketamine, but also causes fewer significant adverse effects; 2) the glycine partial agonist GLYX-13 appears to be effective both as monotherapy and adjunctive treatment in the treatment of MDD. An oral analogue, NRX-1074, is currently under investigation; and 3) mGluR modulators targeting mGluR5 have demonstrated convincing preclinical results.
Collapse
|
41
|
Sarantis K, Tsiamaki E, Kouvaros S, Papatheodoropoulos C, Angelatou F. Adenosine A₂A receptors permit mGluR5-evoked tyrosine phosphorylation of NR2B (Tyr1472) in rat hippocampus: a possible key mechanism in NMDA receptor modulation. J Neurochem 2015; 135:714-26. [PMID: 26303340 DOI: 10.1111/jnc.13291] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 07/23/2015] [Accepted: 08/07/2015] [Indexed: 12/30/2022]
Abstract
A great body of evidence points toward a functional interaction between metabotropic glutamate 5 receptors (mGluR5) and NMDA receptors (NMDAR) that enhances synaptic plasticity and cognition. However, the molecular mechanism underlying this interaction remains unclear. Here, we show that co-activation of mGluR5 and NMDAR in hippocampal slices synergistically leads to a robust phosphorylation of NR2B (Tyr1472), which is Src kinase dependent and is enabled by endogenous adenosine acting on A2A receptors. As it is well known, NR2B (Tyr1472) phosphorylation anchors NR2B-containing NMDARs to the surface of post-synaptic membranes, preventing their internalization. This is supported by our electrophysiological experiments showing that co-activation of mGluR5 and NMDARs robustly enhances NMDAR-dependent neuronal excitability recorded in CA1 hippocampal region, which temporally coincides with the robust increase in NR2B (Tyr1472) phosphorylation, depends on Src kinases and is also permitted by A2A receptors. Thus, we strongly suggest that NR2B (Tyr1472) phosphorylation constitutes, at least to some extent, the molecular mechanism underlying the mGluR5-mediated enhancement of NMDAR-dependent responses, which is modulated by A2A receptors. A better understanding of the molecular basis of mGluR5/NMDAR interaction would elucidate their role in synaptic plasticity processes as well as in pathological conditions. We propose the following molecular mechanism by which metabotropic Glutamate Receptor 5 (mGluR5) potentiate ionotropic Glutamate N-Methyl-D-Aspartate Receptor (NMDAR) responses in rat hippocampus. Co-activation of mGLUR5/NMDAR activates Src kinases, leading to NR2B(Tyr1472) phosphorylation, which anchors NR2B-containing NMDAR to the plasma membrane, thus inducing a robust increase in the NMDA-dependent excitability. Interestingly, adenosine A2A receptors license the mGluR5-induced NR2B(Tyr1472) phosphorylation.
Collapse
Affiliation(s)
| | - Eirini Tsiamaki
- Physiology Department, Medical School, University of Patras, Patras, Greece
| | - Stylianos Kouvaros
- Physiology Department, Medical School, University of Patras, Patras, Greece
| | | | - Fevronia Angelatou
- Physiology Department, Medical School, University of Patras, Patras, Greece
| |
Collapse
|
42
|
Kang J, Park H, Kim E. IRSp53/BAIAP2 in dendritic spine development, NMDA receptor regulation, and psychiatric disorders. Neuropharmacology 2015; 100:27-39. [PMID: 26275848 DOI: 10.1016/j.neuropharm.2015.06.019] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 06/26/2015] [Accepted: 06/28/2015] [Indexed: 01/08/2023]
Abstract
IRSp53 (also known as BAIAP2) is a multi-domain scaffolding and adaptor protein that has been implicated in the regulation of membrane and actin dynamics at subcellular structures, including filopodia and lamellipodia. Accumulating evidence indicates that IRSp53 is an abundant component of the postsynaptic density at excitatory synapses and an important regulator of actin-rich dendritic spines. In addition, IRSp53 has been implicated in diverse psychiatric disorders, including autism spectrum disorders, schizophrenia, and attention deficit/hyperactivity disorder. Mice lacking IRSp53 display enhanced NMDA (N-methyl-d-aspartate) receptor function accompanied by social and cognitive deficits, which are reversed by pharmacological suppression of NMDA receptor function. These results suggest the hypothesis that defective actin/membrane modulation in IRSp53-deficient dendritic spines may lead to social and cognitive deficits through NMDA receptor dysfunction. This article is part of the Special Issue entitled 'Synaptopathy--from Biology to Therapy'.
Collapse
Affiliation(s)
- Jaeseung Kang
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, South Korea
| | - Haram Park
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, South Korea
| | - Eunjoon Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, South Korea; Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon 305-701, South Korea.
| |
Collapse
|
43
|
Kang J, Kim E. Suppression of NMDA receptor function in mice prenatally exposed to valproic acid improves social deficits and repetitive behaviors. Front Mol Neurosci 2015; 8:17. [PMID: 26074764 PMCID: PMC4444740 DOI: 10.3389/fnmol.2015.00017] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 05/09/2015] [Indexed: 12/12/2022] Open
Abstract
Animals prenatally exposed to valproic acid (VPA), an antiepileptic agent, have been used as a model for autism spectrum disorders (ASDs). Previous studies have identified enhanced NMDA receptor (NMDAR) function in the brain of VPA rats, and demonstrated that pharmacological suppression of NMDAR function normalizes social deficits in these animals. However, whether repetitive behavior, another key feature of ASDs, can be rescued by NMDAR inhibition remains unknown. We report here that memantine, an NMDAR antagonist, administered to VPA mice rescues both social deficits and repetitive behaviors such as self-grooming and jumping. These results suggest that suppression of elevated NMDAR function in VPA animals normalizes repetitive behaviors in addition to social deficits.
Collapse
Affiliation(s)
- Jaeseung Kang
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology Daejeon, Korea
| | - Eunjoon Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology Daejeon, Korea ; Center for Synaptic Brain Dysfunctions, Institute for Basic Science Daejeon, Korea
| |
Collapse
|
44
|
Rudy CC, Hunsberger HC, Weitzner DS, Reed MN. The role of the tripartite glutamatergic synapse in the pathophysiology of Alzheimer's disease. Aging Dis 2015; 6:131-48. [PMID: 25821641 PMCID: PMC4365957 DOI: 10.14336/ad.2014.0423] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Accepted: 04/23/2014] [Indexed: 12/26/2022] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia in individuals over 65 years of age and is characterized by accumulation of beta-amyloid (Aβ) and tau. Both Aβ and tau alter synaptic plasticity, leading to synapse loss, neural network dysfunction, and eventually neuron loss. However, the exact mechanism by which these proteins cause neurodegeneration is still not clear. A growing body of evidence suggests perturbations in the glutamatergic tripartite synapse, comprised of a presynaptic terminal, a postsynaptic spine, and an astrocytic process, may underlie the pathogenic mechanisms of AD. Glutamate is the primary excitatory neurotransmitter in the brain and plays an important role in learning and memory, but alterations in glutamatergic signaling can lead to excitotoxicity. This review discusses the ways in which both beta-amyloid (Aβ) and tau act alone and in concert to perturb synaptic functioning of the tripartite synapse, including alterations in glutamate release, astrocytic uptake, and receptor signaling. Particular emphasis is given to the role of N-methyl-D-aspartate (NMDA) as a possible convergence point for Aβ and tau toxicity.
Collapse
Affiliation(s)
- Carolyn C. Rudy
- Behavioral Neuroscience, Department of Psychology, West Virginia University, Morgantown, WV, 26506, USA
| | - Holly C. Hunsberger
- Behavioral Neuroscience, Department of Psychology, West Virginia University, Morgantown, WV, 26506, USA
| | - Daniel S. Weitzner
- Behavioral Neuroscience, Department of Psychology, West Virginia University, Morgantown, WV, 26506, USA
| | - Miranda N. Reed
- Behavioral Neuroscience, Department of Psychology, West Virginia University, Morgantown, WV, 26506, USA
- Center for Neuroscience, West Virginia University, Morgantown, WV, 26506, USA
- Center for Basic and Translational Stroke Research, West Virginia University, Morgantown, WV, 26506, USA
| |
Collapse
|
45
|
Social deficits in IRSp53 mutant mice improved by NMDAR and mGluR5 suppression. Nat Neurosci 2015; 18:435-43. [DOI: 10.1038/nn.3927] [Citation(s) in RCA: 124] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 12/16/2014] [Indexed: 02/07/2023]
|
46
|
The effect of the mGlu5 negative allosteric modulator MTEP and NMDA receptor partial agonist D-cycloserine on Pavlovian conditioned fear. Int J Neuropsychopharmacol 2014; 17:1521-32. [PMID: 24674862 DOI: 10.1017/s1461145714000303] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
The metabotropic glutamate receptor 5 (mGlu5) and N-methyl-D-aspartate (NMDA) receptor are critical for processes underlying synaptic plasticity, such as long-term potentiation. mGlu5 signaling increases neuronal excitability and potentiates NMDA receptor currents in the amygdala and the hippocampus. The present study examined the involvement of mGlu5 in the acquisition and consolidation of conditioned fear to a tone and context in mice, and explored the functional relationship between mGlu5 and NMDA receptors in this regard. Experiment 1 showed that systemic administration of the mGlu5 negative allosteric modulator 3-[(2-methyl-1,3-thiazol-4-yl)ethynyl]pyridine (MTEP) prior to conditioning significantly attenuated cue-elicited freezing during fear conditioning, which suggests that mGlu5 is necessary for the formation of a tone-shock association. This effect was dose-related (Experiment 2) and not due to any effects of MTEP on shock sensitivity or state-dependency (Experiment 3). Post-conditioning injection of MTEP had no effects (Experiment 4). Although post-conditioning injection of the NMDA receptor partial agonist D-cycloserine (DCS) alone facilitated consolidation of conditioned fear (Experiment 6), it was not able to rescue the acquisition deficit caused by MTEP (Experiment 5). Taken together, these findings indicate a crucial role for mGlu5 signaling in acquisition and NMDA receptor signaling in consolidation of conditioned fear.
Collapse
|
47
|
Ribeiro FM, Doria JG, Ferguson SSG. mGluR5: a potential target for the treatment of Huntington's disease. FUTURE NEUROLOGY 2014. [DOI: 10.2217/fnl.14.14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Affiliation(s)
- Fabiola M Ribeiro
- Departamento de Bioquimica & Imunologia, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Juliana G Doria
- Departamento de Bioquimica & Imunologia, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Stephen SG Ferguson
- J Allyn Taylor Centre for Cell Biology, University of Western Ontario, London, ON, Canada
| |
Collapse
|
48
|
Partridge JG, Lewin AE, Yasko JR, Vicini S. Contrasting actions of group I metabotropic glutamate receptors in distinct mouse striatal neurones. J Physiol 2014; 592:2721-33. [PMID: 24710062 DOI: 10.1113/jphysiol.2014.272773] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
In mouse striatum, metabotropic glutamate receptor (mGluR) activation leads to several modulatory effects in synaptic transmission. These effects range from dampening of glutamate release from excitatory terminals to depolarization of divergent classes of interneurones. We compared the action of group I mGluR activation on several populations of striatal neurones using a combination of genetic identification, electrophysiology, and Ca(2+) imaging techniques. Patch-clamp recordings from spiny projection neurones (SPNs) and various interneurone populations demonstrated that the group I mGluR agonist (RS)-3,5-dihydroxyphenylglycine (DHPG) robustly depolarizes several interneurone classes that form GABAergic synapses onto SPNs. We further utilized the genetic reporter mouse strain Ai38, which expresses the calcium indicator protein GCaMP3 in a Cre-dependent manner. Breeding Ai38 mice with various neurone selective, promoter-driven Cre recombinase mice resulted in GCaMP3 expression in defined cell populations in striatum. Consistent with our electrophysiological findings, group I agonist applications increased intracellular levels of calcium ([Ca(2+)]i) in all interneurone populations tested. We also found that acute DHPG application evoked a transient, rapid increase in [Ca(2+)]i from only a small percentage of identifiable SPNs. Surprisingly, this fast [Ca(2+)]i response exhibited a robust enhancement or sensitization, in a calcium-dependent fashion. Following several procedures to increase [Ca(2+)]i, the vast majority of SPNs responded with rapid changes in [Ca(2+)]i to mGluR agonists in a time-dependent fashion. These findings extend our understanding on group I mGluR influence of striatal output via powerful, local GABAergic connections in addition to [Ca(2+)]i dynamics that impact on activity or spike-timing-dependent forms of synaptic plasticity.
Collapse
Affiliation(s)
- John G Partridge
- Department of Pharmacology and Physiology, Georgetown University School of Medicine, Washington, DC, 20007, USA Interdisciplinary Program in Neuroscience, Georgetown University School of Medicine, Washington, DC, 20007, USA
| | - Amanda E Lewin
- Department of Pharmacology and Physiology, Georgetown University School of Medicine, Washington, DC, 20007, USA
| | - Jessica R Yasko
- Department of Pharmacology and Physiology, Georgetown University School of Medicine, Washington, DC, 20007, USA
| | - Stefano Vicini
- Department of Pharmacology and Physiology, Georgetown University School of Medicine, Washington, DC, 20007, USA Interdisciplinary Program in Neuroscience, Georgetown University School of Medicine, Washington, DC, 20007, USA
| |
Collapse
|
49
|
D'Antoni S, Spatuzza M, Bonaccorso CM, Musumeci SA, Ciranna L, Nicoletti F, Huber KM, Catania MV. Dysregulation of group-I metabotropic glutamate (mGlu) receptor mediated signalling in disorders associated with Intellectual Disability and Autism. Neurosci Biobehav Rev 2014; 46 Pt 2:228-41. [PMID: 24548786 DOI: 10.1016/j.neubiorev.2014.02.003] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Revised: 01/13/2014] [Accepted: 02/06/2014] [Indexed: 12/11/2022]
Abstract
Activation of group-I metabotropic glutamate receptors, mGlu1 and mGlu5, triggers a variety of signalling pathways in neurons and glial cells, which are differently implicated in synaptic plasticity. The earliest and much of key studies discovered abnormal mGlu5 receptor function in Fragile X syndrome (FXS) mouse models which then motivated more recent work that finds mGlu5 receptor dysfunction in related disorders such as intellectual disability (ID), obsessive-compulsive disorder (OCD) and autism. Therefore, mGlu1/5 receptor dysfunction may represent a common aetiology of these complex diseases. Furthermore, many studies have focused on dysregulation of mGlu5 signalling to synaptic protein synthesis. However, emerging evidence finds abnormal mGlu5 receptor interactions with its scaffolding proteins in FXS which results in mGlu5 receptor dysfunction and phenotypes independent of signalling to protein synthesis. Finally, both an increased and reduced mGlu5 functioning seem to be associated with ID and autism spectrum disorders, with important consequences for potential treatment of these developmental disorders.
Collapse
Affiliation(s)
- Simona D'Antoni
- Institute of Neurological Sciences, the National Research Council of Italy (CNR), Catania, Italy
| | - Michela Spatuzza
- Institute of Neurological Sciences, the National Research Council of Italy (CNR), Catania, Italy
| | | | | | - Lucia Ciranna
- Department of Biomedical Sciences, section of Physiology, University of Catania, Italy
| | - Ferdinando Nicoletti
- IRCCS Neuromed, Pozzilli (IS), Italy; University of Rome La Sapienza, Rome, Italy
| | - Kimberly M Huber
- University of Texas Southwestern Medical Center, Department of Neuroscience, Dallas, TX 75390-9111, USA
| | - Maria Vincenza Catania
- Institute of Neurological Sciences, the National Research Council of Italy (CNR), Catania, Italy; IRCCS Oasi Maria SS, Troina (EN), Italy.
| |
Collapse
|
50
|
Ambrosi G, Cerri S, Blandini F. A further update on the role of excitotoxicity in the pathogenesis of Parkinson’s disease. J Neural Transm (Vienna) 2014; 121:849-59. [DOI: 10.1007/s00702-013-1149-z] [Citation(s) in RCA: 131] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Accepted: 12/19/2013] [Indexed: 11/30/2022]
|