1
|
Fountoulakis KN, Tohen M, Zarate CA. Pharmacodynamic properties of lumateperone and its efficacy in acute bipolar depression: a mechanistic hypothesis based on data. Eur Neuropsychopharmacol 2024; 81:1-9. [PMID: 38310714 DOI: 10.1016/j.euroneuro.2024.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 01/07/2024] [Accepted: 01/10/2024] [Indexed: 02/06/2024]
Abstract
The treatment of bipolar depression is one of the most challenging needs in contemporary psychiatry. Currently, only quetiapine, olanzapine-fluoxetine combination, lurasidone, cariprazine, and recently lumateperone have been FDA-approved to treat this condition. The neurobiology of bipolar depression and the possible mechanistic targets of bipolar antidepressant therapy remain elusive. The current study investigated whether the pharmacodynamic properties of lumateperone fit into a previously developed model which was the first to be derived based on the strict combination of clinical and preclinical data. The authors performed a systematic review of the literature to identify the pharmacodynamic properties of lumateperone. The original model suggests that a constellation of effects on different receptors is necessary, but refinements, including the present study, suggest that the inhibition of the serotonin reuptake at the first level, the 5HT-2A blockade at the second level, and the norepinephrine alpha-1 receptors blockade at a third level in combination with D1 blockade contribute to the antidepressant effect in acute bipolar depression. The D2 blockade acts as a protective mechanism and reduces the risk of switching to mania/hypomania.
Collapse
Affiliation(s)
| | - Mauricio Tohen
- University Distinguished Professor and Chairman, Department of Psychiatry and Behavioral Sciences, University of New Mexico Health Sciences Center, 2400 Tucker Ave NE MSC09 5030, Albuquerque, NM 87131-0001, USA
| | - Carlos A Zarate
- Chief Experimental Therapeutics & Pathophysiology Branch, Division of Intramural Research Program, National Institute of Mental Health, Bethesda, MD 20892, USA
| |
Collapse
|
2
|
Tan Y, Cheng C, Zheng C, Zeng W, Yang X, Xu Y, Zhang Z, Ma Z, Xu Y, Cao X. Activation of mGlu 2/3 receptors in the striatum alleviates L-DOPA-induced dyskinesia and inhibits abnormal postsynaptic molecular expression. Pharmacol Biochem Behav 2023; 231:173637. [PMID: 37714223 DOI: 10.1016/j.pbb.2023.173637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 09/11/2023] [Accepted: 09/12/2023] [Indexed: 09/17/2023]
Abstract
Group II metabotropic glutamate receptors (mGlu2/3 receptors) have been regarded as promising candidates for the treatment of L-DOPA-induced dyskinesia (LID); however, confirmation is still lacking. As the hub of the basal ganglia circuit, the striatum plays a critical role in action control. Supersensitive responsiveness of glutamatergic corticostriatal input may be the key mechanism for the development of LID. In this study, we first examined the potency of LY354740 (12 mg/kg, i.p.) in modulating glutamate and dopamine release in lesioned striatum of stable LID rats. Then, we injected LY354740 (20nmoL or 40nmoL in 4 μL of sterile 0.9 % saline) directly into the lesioned striatum to verify its ability to reduce or attenuate L-DOPA-induced abnormal involuntary movements. In experiment conducted in established LID rats, after continuous injection for 4 days, we found that LY354740 significantly reduced the expression of dyskinesia. In another experiment conducted in parkinsonism rat models, we found that LY354740 attenuated the development of LID with an inverted-U dose-response curve. The role of LY354740 in modulating striatal expressions of LID-related molecular changes was also assessed after these behavioral experiments. We found that LY354740 significantly inhibited abnormal expressions of p-Fyn/p-NMDA/p-ERK1/2/p-HistoneH3/ΔFosB, which is in line with its ability to alleviate abnormal involuntary movements in both LID expression and induction phase. Our study indicates that activation of striatal mGlu2/3 receptors can attenuate the development of dyskinesia in parkinsonism rats and provide some functional improvements in LID rats by inhibiting LID-related molecular changes.
Collapse
Affiliation(s)
- Yang Tan
- Department of Neurology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China
| | - Chi Cheng
- Department of Neurology, Hanchuan People's Hospital, 432300, China
| | - Cong Zheng
- Institute of Neuroscience, Chinese Academy of Sciences, Shanghai 200000, China
| | - Weiqi Zeng
- Department of Neurology, The First People's Hospital of Foshan, Foshan 528000, China
| | - Xiaoman Yang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Yu Xu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Zhaoyuan Zhang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Zhuoran Ma
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Yan Xu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Xuebing Cao
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China.
| |
Collapse
|
3
|
Madhamanchi K, Madhamanchi P, Jayalakshmi S, Panigrahi M, Patil A, Phanithi PB. Dopamine and Glutamate Crosstalk Worsen the Seizure Outcome in TLE-HS Patients. Mol Neurobiol 2023; 60:4952-4965. [PMID: 37209264 DOI: 10.1007/s12035-023-03361-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 04/19/2023] [Indexed: 05/22/2023]
Abstract
Temporal lobe epilepsy (TLE), accompanied by hippocampal sclerosis (HS), is the most common form of drug-resistant epilepsy (DRE). Nearly 20% of the patients showed seizure recurrence even after surgery, and the reasons are yet to be understood. Dysregulation of neurotransmitters is evident during seizures, which can induce excitotoxicity. The present study focused on understanding the molecular changes associated with Dopamine (DA) and glutamate signaling and their possible impact on the persistence of excitotoxicity and seizure recurrence in patients with drug-resistant TLE-HS who underwent surgery. According to the International League against Epilepsy (ILAE) suggested classification for seizure outcomes, the patients (n = 26) were classified as class 1 (no seizures) and class 2 (persistent seizures) using the latest post-surgery follow-up data to understand the prevalent molecular changes in seizure-free and seizure-recurrence patient groups. Our study uses thioflavin T assay, western blot analysis, immunofluorescence assays, and fluorescence resonance energy transfer (FRET) assays. We have observed a substantial increase in the DA and glutamate receptors that promote excitotoxicity. Patients who had seizure recurrence showed a significant increase in (pNR2B, p < 0.009; and pGluR1, p < 0.01), protein phosphatase1γ (PP1γ; p < 0.009), protein kinase A (PKAc; p < 0.001) and dopamine-cAMP regulated phospho protein32 (pDARPP32T34; p < 0.009) which are critical for long-term potentiation (LTP), excitotoxicity compared to seizure-free patients and controls. A significant increase in D1R downstream kinases like PKA (p < 0.001), pCAMKII (p < 0.009), and Fyn (p < 0.001) was observed in patient samples compared to controls. Anti-epileptic DA receptor D2R was found to be decreased in ILAE class 2 (p < 0.02) compared to class 1. Since upregulation of DA and glutamate signaling supports LTP and excitotoxicity, we believe it could impact seizure recurrence. Further studies about the impact of DA and glutamate signaling on the distribution of PP1γ at postsynaptic density and synaptic strength could help us understand the seizure microenvironment in patients. Dopamine, Glutamate signal crosstalk. Diagram representing the PP1γ regulation by NMDAR negative feedback inhibition signaling (green circle-left) and D1R signal (red circle-middle) domination over PP1γ though increased PKA, pDARPP32T34, and supports pGluR1, pNR2B in seizure recurrent patients. D1R-D2R hetero dimer activation (red circle-right) increases cellular Ca2+ and pCAMKIIα activation. All these events lead to calcium overload in HS patients and excitotoxicity, particularly in patients experiencing recurrent seizures.
Collapse
Affiliation(s)
- Kishore Madhamanchi
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana, 500046, India
| | - Pradeep Madhamanchi
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana, 500046, India
- Govt. Degree College for Men's, Srikakulam District, Andhra Pradesh, 532001, India
| | - Sita Jayalakshmi
- Department of Neurology, Krishna Institute of Medical Sciences (KIMS), Secunderabad, Telangana, India
| | - Manas Panigrahi
- Department of Neurology, Krishna Institute of Medical Sciences (KIMS), Secunderabad, Telangana, India
| | - Anuja Patil
- Department of Neurology, Krishna Institute of Medical Sciences (KIMS), Secunderabad, Telangana, India
| | - Prakash Babu Phanithi
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana, 500046, India.
| |
Collapse
|
4
|
Rocha GS, Freire MAM, Britto AM, Paiva KM, Oliveira RF, Fonseca IAT, Araújo DP, Oliveira LC, Guzen FP, Morais PLAG, Cavalcanti JRLP. Basal ganglia for beginners: the basic concepts you need to know and their role in movement control. Front Syst Neurosci 2023; 17:1242929. [PMID: 37600831 PMCID: PMC10435282 DOI: 10.3389/fnsys.2023.1242929] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 07/21/2023] [Indexed: 08/22/2023] Open
Abstract
The basal ganglia are a subcortical collection of interacting clusters of cell bodies, and are involved in reward, emotional, and motor circuits. Within all the brain processing necessary to carry out voluntary movement, the basal nuclei are fundamental, as they modulate the activity of the motor regions of the cortex. Despite being much studied, the motor circuit of the basal ganglia is still difficult to understand for many people at all, especially undergraduate and graduate students. This review article seeks to bring the functioning of this circuit with a simple and objective approach, exploring the functional anatomy, neurochemistry, neuronal pathways, related diseases, and interactions with other brain regions to coordinate voluntary movement.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - José R. L. P. Cavalcanti
- Laboratory of Experimental Neurology, Department of Biomedical Sciences, State University of Rio Grande do Norte, Mossoró, Brazil
| |
Collapse
|
5
|
Olivero G, Grilli M, Marchi M, Pittaluga A. Metamodulation of presynaptic NMDA receptors: New perspectives for pharmacological interventions. Neuropharmacology 2023; 234:109570. [PMID: 37146939 DOI: 10.1016/j.neuropharm.2023.109570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 04/26/2023] [Accepted: 05/02/2023] [Indexed: 05/07/2023]
Abstract
Metamodulation shifted the scenario of the central neuromodulation from a simplified unimodal model to a multimodal one. It involves different receptors/membrane proteins physically associated or merely colocalized that act in concert to control the neuronal functions influencing each other. Defects or maladaptation of metamodulation would subserve neuropsychiatric disorders or even synaptic adaptations relevant to drug dependence. Therefore, this "vulnerability" represents a main issue to be deeply analyzed to predict its aetiopathogenesis, but also to propose targeted pharmaceutical interventions. The review focusses on presynaptic release-regulating NMDA receptors and on some of the mechanisms of their metamodulation described in the literature. Attention is paid to the interactors, including both ionotropic and metabotropic receptors, transporters and intracellular proteins, which metamodulate their responsiveness in physiological conditions but also undergo adaptation that are relevant to neurological dysfunctions. All these structures are attracting more and more the interest as promising druggable targets for the treatment of NMDAR-related central diseases: these substances would not exert on-off control of the colocalized NMDA receptors (as usually observed with NMDAR full agonists/antagonists), but rather modulate their functions, with the promise of limiting side effects that would favor their translation from preclinic to clinic.
Collapse
Affiliation(s)
- Guendalina Olivero
- Department of Pharmacy, University of Genoa, Viale Cembrano 4, 16148, Genoa, Italy
| | - Massimo Grilli
- Department of Pharmacy, University of Genoa, Viale Cembrano 4, 16148, Genoa, Italy; Inter-University Center for the Promotion of the 3Rs Principles in Teaching & Research (Centro 3R), 16148, Genoa, Italy.
| | - Mario Marchi
- Department of Pharmacy, University of Genoa, Viale Cembrano 4, 16148, Genoa, Italy
| | - Anna Pittaluga
- Department of Pharmacy, University of Genoa, Viale Cembrano 4, 16148, Genoa, Italy; Inter-University Center for the Promotion of the 3Rs Principles in Teaching & Research (Centro 3R), 16148, Genoa, Italy
| |
Collapse
|
6
|
Yin LT, Feng RR, Xie XY, Yang XR, Yang ZF, Hu JJ, Wu SF, Zhang C. Matrix metalloproteinase-9 overexpression in the hippocampus reduces alcohol-induced conditioned-place preference by regulating synaptic plasticity in mice. Behav Brain Res 2023; 442:114330. [PMID: 36746309 DOI: 10.1016/j.bbr.2023.114330] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 02/02/2023] [Accepted: 02/03/2023] [Indexed: 02/07/2023]
Abstract
Extracellular matrix proteins appear to be necessary for the synaptic plasticity that underlies addiction memory. In the brain, matrix metalloproteinases (MMPs), especially matrix metalloproteinase-9 (MMP-9), have been recently implicated in processes involving alcohol reward and memory. Here, we showed for the first time, the positive effects of MMP-9 on alcohol-induced conditioned place preference (CPP) behavior and hippocampal neuron plasticity in C57BL/6 mice. Using recombinant adeno-associated viruses to overexpress MMP-9 in the hippocampus, we investigated the NMDAR, PSD-95, and cellular cytoskeleton proteins F-actin/G-actin in the modulation of alcohol reward behavior in mice exposed to CPP. We found that hippocampal infusions of MMP-9 decreased alcohol-induced place preference suggesting a reduction in alcohol reward. Western blot analysis demonstrated that protein expression of NMDA receptors (GluN1, GluN2A and GluN2B) in the hippocampus of alcohol-exposed mice were higher than that of the saline group. Further, the expression of these proteins was decreased in MMP-9 overexpressing mice. MMP-9 also regulated the ratio of F-actin/G-actin (dendritic spines cytoskeleton proteins), which might be the key mediator for behavioral changes in mice. Consequently, our results highlight new evidence that MMP-9 may play an important role in the molecular mechanism underlying alcohol reward and preference.
Collapse
Affiliation(s)
- Li-Tian Yin
- Key Laboratory for Cellular Physiology, Ministry of Education, Key Laboratory of Cellular Physiology in Shanxi Province, Department of Physiology, Shanxi Medical University, Taiyuan, Shanxi 030001, China.
| | - Rui-Rui Feng
- Key Laboratory for Cellular Physiology, Ministry of Education, Key Laboratory of Cellular Physiology in Shanxi Province, Department of Physiology, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Xiao-Yan Xie
- Key Laboratory for Cellular Physiology, Ministry of Education, Key Laboratory of Cellular Physiology in Shanxi Province, Department of Physiology, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Xiao-Rong Yang
- Key Laboratory for Cellular Physiology, Ministry of Education, Key Laboratory of Cellular Physiology in Shanxi Province, Department of Physiology, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Zhuan-Fang Yang
- Key Laboratory for Cellular Physiology, Ministry of Education, Key Laboratory of Cellular Physiology in Shanxi Province, Department of Physiology, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Jia-Jia Hu
- Key Laboratory for Cellular Physiology, Ministry of Education, Key Laboratory of Cellular Physiology in Shanxi Province, Department of Physiology, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Shu-Fen Wu
- Key Laboratory for Cellular Physiology, Ministry of Education, Key Laboratory of Cellular Physiology in Shanxi Province, Department of Physiology, Shanxi Medical University, Taiyuan, Shanxi 030001, China; Department of Pediatrics, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Ce Zhang
- Key Laboratory for Cellular Physiology, Ministry of Education, Key Laboratory of Cellular Physiology in Shanxi Province, Department of Physiology, Shanxi Medical University, Taiyuan, Shanxi 030001, China.
| |
Collapse
|
7
|
Kennedy C, van Onselen R, Downing TG. β-N-methylamino-l-alanine is a non-competitive inhibitor of vesicular monoamine transporter 2. Toxicon 2023; 222:106978. [PMID: 36410456 DOI: 10.1016/j.toxicon.2022.106978] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/04/2022] [Accepted: 11/18/2022] [Indexed: 11/20/2022]
Abstract
The neurotoxic, non-proteinogenic amino acid β-N-methylamino-l-alanine (BMAA) has been implicated in the development of neurodegenerative diseases; however, the mechanism(s) and mode(s) of toxicity remain unclear. Similarities in the neuropathology and behavioural deficits of neonatal rats exposed to either BMAA or reserpine, a known vesicular monoamine transporter 2 (VMAT2) inhibitor, suggest a similar mode of action. The aims of this study were therefore to determine if BMAA could prevent the uptake of serotonin into dense granules via inhibition of VMAT2, and, if so, the type of inhibition caused by BMAA. Exposing platelet dense granules to BMAA resulted in a concentration-dependent reduction in serotonin uptake. The inhibition of VMAT2 was non-competitive. The findings from this study support previous reports that BMAA-associated neuropathologies in a neonatal rat model may be due to VMAT2 inhibition during critical periods of neurogenesis.
Collapse
Affiliation(s)
- Chanté Kennedy
- Department of Biochemistry and Microbiology, Nelson Mandela University, P.O. Box 77000, Port Elizabeth, 6021, South Africa.
| | - Rianita van Onselen
- Department of Biochemistry and Microbiology, Nelson Mandela University, P.O. Box 77000, Port Elizabeth, 6021, South Africa; Biomedical Research and Innovation Platform, South African Medical Research Council, Cape Town, South Africa.
| | - Tim G Downing
- Department of Biochemistry and Microbiology, Nelson Mandela University, P.O. Box 77000, Port Elizabeth, 6021, South Africa.
| |
Collapse
|
8
|
Chen LJ, Chen JR, Tseng GF. Modulation of striatal glutamatergic, dopaminergic and cholinergic neurotransmission pathways concomitant with motor disturbance in rats with kaolin-induced hydrocephalus. Fluids Barriers CNS 2022; 19:95. [PMID: 36437472 PMCID: PMC9701403 DOI: 10.1186/s12987-022-00393-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 11/15/2022] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Hydrocephalus is characterized by abnormal accumulation of cerebrospinal fluid in the cerebral ventricles and causes motor impairments. The mechanisms underlying the motor changes remain elusive. Enlargement of ventricles compresses the striatum of the basal ganglia, a group of nuclei involved in the subcortical motor circuit. Here, we used a kaolin-injection juvenile rat model to explore the effects of acute and chronic hydrocephalus, 1 and 5 weeks post-treatment, respectively on the three major neurotransmission pathways (glutamatergic, dopaminergic and cholinergic) in the striatum. METHODS Rats were evaluated for motor impairments. Expressions of presynaptic and postsynaptic protein markers related to the glutamatergic, dopaminergic, and cholinergic connections in the striatum were evaluated. Combined intracellular dye injection and substance P immunohistochemistry were used to distinguish between direct and indirect pathway striatal medium spiny neurons (d and i-MSNs) for the analysis of their dendritic spine density changes. RESULTS Hydrocephalic rats showed compromised open-field gait behavior. However, male but not female rats displayed stereotypic movements and compromised rotarod performance. Morphologically, the increase in lateral ventricle sizes was greater in the chronic than acute hydrocephalus conditions. Biochemically, hydrocephalic rats had significantly decreased striatal levels of synaptophysin, vesicular glutamate transporter 1, and glutamatergic postsynaptic density protein 95, suggesting a reduction of corticostriatal excitation. The expression of GluR2/3 was also reduced suggesting glutamate receptor compositional changes. The densities of dendritic spines, morphological correlates of excitatory synaptic foci, on both d and i-MSNs were also reduced. Hydrocephalus altered type 1 (DR1) and 2 (DR2) dopamine receptor expressions without affecting tyrosine hydroxylase level. DR1 was decreased in acute and chronic hydrocephalus, while DR2 only started to decrease later during chronic hydrocephalus. Since dopamine excites d-MSNs through DR1 and inhibits i-MSNs via DR2, our findings suggest that hydrocephalus downregulated the direct basal ganglia neural pathway persistently and disinhibited the indirect pathway late during chronic hydrocephalus. Hydrocephalus also persistently reduced the striatal choline acetyltransferase level, suggesting a reduction of cholinergic modulation. CONCLUSIONS Hydrocephalus altered striatal glutamatergic, dopaminergic, and cholinergic neurotransmission pathways and tipped the balance between the direct and indirect basal ganglia circuits, which could have contributed to the motor impairments in hydrocephalus.
Collapse
Affiliation(s)
- Li-Jin Chen
- grid.411824.a0000 0004 0622 7222Department of Anatomy, College of Medicine, Tzu Chi University, No. 701, Section 3, Jhongyang Rd., Hualien, 97004 Taiwan
| | - Jeng-Rung Chen
- grid.260542.70000 0004 0532 3749Department of Veterinary Medicine, College of Veterinary Medicine, National Chung-Hsing University, Taichung, Taiwan
| | - Guo-Fang Tseng
- grid.411824.a0000 0004 0622 7222Department of Anatomy, College of Medicine, Tzu Chi University, No. 701, Section 3, Jhongyang Rd., Hualien, 97004 Taiwan
| |
Collapse
|
9
|
Weber LA, Tomiello S, Schöbi D, Wellstein KV, Mueller D, Iglesias S, Stephan KE. Auditory mismatch responses are differentially sensitive to changes in muscarinic acetylcholine versus dopamine receptor function. eLife 2022; 11:74835. [PMID: 35502897 PMCID: PMC9098218 DOI: 10.7554/elife.74835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 04/26/2022] [Indexed: 11/13/2022] Open
Abstract
The auditory mismatch negativity (MMN) has been proposed as a biomarker of NMDA receptor (NMDAR) dysfunction in schizophrenia. Such dysfunction may be caused by aberrant interactions of different neuromodulators with NMDARs, which could explain clinical heterogeneity among patients. In two studies (N = 81 each), we used a double-blind placebo-controlled between-subject design to systematically test whether auditory mismatch responses under varying levels of environmental stability are sensitive to diminishing and enhancing cholinergic vs. dopaminergic function. We found a significant drug × mismatch interaction: while the muscarinic acetylcholine receptor antagonist biperiden delayed and topographically shifted mismatch responses, particularly during high stability, this effect could not be detected for amisulpride, a dopamine D2/D3 receptor antagonist. Neither galantamine nor levodopa, which elevate acetylcholine and dopamine levels, respectively, exerted significant effects on MMN. This differential MMN sensitivity to muscarinic versus dopaminergic receptor function may prove useful for developing tests that predict individual treatment responses in schizophrenia.
Collapse
Affiliation(s)
- Lilian Aline Weber
- Translational Neuroimaging Unit (TNU), Institute for Biomedical EngineeringInstitute for Biomedical Engineering, University of Zurich, Zurich, Switzerland
| | - Sara Tomiello
- Translational Neuroimaging Unit (TNU), Institute for Biomedical Engineering, University of Zurich, Zurich, Switzerland
| | - Dario Schöbi
- Translational Neuroimaging Unit (TNU), Institute for Biomedical Engineering, University of Zurich, Zurich, Switzerland
| | - Katharina V Wellstein
- Translational Neuroimaging Unit (TNU), Institute for Biomedical Engineering, University of Zurich, Zurich, Switzerland
| | - Daniel Mueller
- Institute for Clinical Chemistry, University Hospital of Zurich, Zurich, Switzerland
| | - Sandra Iglesias
- Translational Neuroimaging Unit (TNU), Institute for Biomedical Engineering, University of Zurich, Zurich, Switzerland
| | - Klaas Enno Stephan
- Translational Neuroimaging Unit (TNU), Institute for Biomedical Engineering, University of Zurich, Zurich, Switzerland
| |
Collapse
|
10
|
Puri NM, Romano GR, Lin TY, Mai QN, Irannejad R. The organic cation Transporter 2 regulates dopamine D1 receptor signaling at the Golgi apparatus. eLife 2022; 11:75468. [PMID: 35467530 PMCID: PMC9098220 DOI: 10.7554/elife.75468] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 04/22/2022] [Indexed: 11/13/2022] Open
Abstract
Dopamine is a key catecholamine in the brain and kidney, where it is involved in a number of physiological functions such as locomotion, cognition, emotion, endocrine regulation, and renal function. As a membrane-impermeant hormone and neurotransmitter, dopamine is thought to signal by binding and activating dopamine receptors, members of the G protein coupled receptor (GPCR) family, only on the plasma membrane. Here, using novel nanobody-based biosensors, we demonstrate for the first time that the dopamine D1 receptor (D1DR), the primary mediator of dopaminergic signaling in the brain and kidney, not only functions on the plasma membrane but becomes activated at the Golgi apparatus in the presence of its ligand. We present evidence that activation of the Golgi pool of D1DR is dependent on organic cation transporter 2 (OCT2), a dopamine transporter, providing an explanation for how the membrane-impermeant dopamine accesses subcellular pools of D1DR. We further demonstrate that dopamine activates Golgi-D1DR in murine striatal medium spiny neurons, and this activity depends on OCT2 function. We also introduce a new approach to selectively interrogate compartmentalized D1DR signaling by inhibiting Gαs coupling using a nanobody-based chemical recruitment system. Using this strategy, we show that Golgi-localized D1DRs regulate cAMP production and mediate local protein kinase A activation. Together, our data suggest that spatially compartmentalized signaling hubs are previously unappreciated regulatory aspects of D1DR signaling. Our data provide further evidence for the role of transporters in regulating subcellular GPCR activity.
Collapse
Affiliation(s)
- Natasha M Puri
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, United States
| | - Giovanna R Romano
- Biochemistry Department, Weill Cornell Medicine, New York, United States
| | - Ting-Yu Lin
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, United States
| | - Quynh N Mai
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, United States
| | - Roshanak Irannejad
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, United States
| |
Collapse
|
11
|
Abdullah M, Huang LC, Lin SH, Yang YK. Dopaminergic and glutamatergic biomarkers disruption in addiction and regulation by exercise: a mini review. Biomarkers 2022; 27:306-318. [PMID: 35236200 DOI: 10.1080/1354750x.2022.2049367] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Drug addiction is associated with disruption of a multitude of biomarkers in various brain regions, particularly in the reward center. The most pronounced are dopaminergic and glutamatergic biomarkers, which are affected at various levels. Neuropathological changes in biomarkers alter the homeostasis of the glutamatergic and dopaminergic nervous systems and promote addiction-associated characteristics such as repeated intake, maintenance, withdrawal, reinstatement, and relapse. Exercise has been shown to have a buffering effect on such biomarkers and reverse the effects of addictive substances. METHODS A review of the literature searched in PubMed, examining drug addiction and physical exercise in relation to dopaminergic and glutamatergic systems at any of the three biomarker levels (i.e., neurotransmitter, receptor, or transporter). RESULTS We review the collective impact of addictive substances on the dopaminergic and glutamatergic systems and the beneficial effect of exercise in terms of reversing the damage to these systems. We propose future directions, including implications of exercise as an add-on therapy, substance use disorder (SUD) prognosis and diagnosis and designing of optimized exercise and pharmaceutical regimens based on the aforementioned biomarkers. CONCLUSION Exercise is beneficial for all types of drug addiction at all stages, by reversing molecular damages caused to dopaminergic and glutamatergic systems.
Collapse
Affiliation(s)
- Muhammad Abdullah
- Taiwan International Graduate Program in Interdisciplinary Neuroscience, National Cheng Kung University and Academia Sinica, Taipei, Taiwan
| | - Li-Chung Huang
- Institute of Clinical Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Psychiatry, Chia-Yi Branch, Taichung Veterans General Hospital, Chia-Yi, Taiwan
| | - Shih-Hsien Lin
- Department of Psychiatry, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Institute of Behavioral Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yen Kuang Yang
- Department of Psychiatry, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Institute of Behavioral Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Psychiatry, Tainan Hospital, Ministry of Health and Welfare, Tainan, Taiwan
| |
Collapse
|
12
|
Li ML, Peng Y, An Y, Li GY, Lan Y. LY395756 promotes NR2B expression via activation of AKT/CREB signaling in the juvenile methylazoxymethanol mice model of schizophrenia. Brain Behav 2022; 12:e2466. [PMID: 35025141 PMCID: PMC8865150 DOI: 10.1002/brb3.2466] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 09/27/2021] [Accepted: 11/06/2021] [Indexed: 12/21/2022] Open
Abstract
INTRODUCTION Synaptic N-methyl-d-aspartate receptor subtype 2B(NR2B) is significantly reduced in prefrontal cortex (PFC) in the neurodevelopmental methylazoxymethanol (MAM) model of schizophrenia (SCZ). Recent research has shown that LY395756 can effectively restore NR2B levels and improve cognitive performance in juvenile MAM mice model. However, the underlying mechanisms of these beneficial effects remain unclear. MATERIALS AND METHODS Juvenile MAM mice model of SCZ is used in our study. Synaptic membrane protein levels were examined by western blotting under different treatment conditions. Interaction of cAMP-response element binding protein (CREB) and the promoter of NR2B was detected by the chromatin immunoprecipitation (ChIP) assay. Further examination of signaling pathway that mediates NR2B expression was also investigated by western blotting. RESULTS In the PFC of the juvenile MAM mice schizophrenia model, CREB was found to directly bind with the promoter of NR2B. LY395756 activated the phosphorylation of AKT. Phosphorylated AKT subsequently induced the phosphorylation of CREB, and the activated CREB promoted the expression of NR2B. Subsequent experiments showed that the dephosphorylation of CREB induced by protein phosphatase 1 (PP1) can inhibit NR2B levels. Taken together, these findings support that the AKT/CREB signaling pathway is essential for the promoting effect of LY395756 on synaptic NR2B in PFC in juvenile MAM mice SCZ model. CONCLUSIONS Our investigation has identified a novel mechanism by which LY395756 increases NR2B expression in juvenile MAM mice SCZ model. The AKT/CREB signaling pathway warrants further research as a potential direction for clinical treatment of SCZ.
Collapse
Affiliation(s)
- Meng-Lin Li
- Department of Rehabilitation, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Yuan Peng
- Department of Rehabilitation, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Ying An
- Department of Rehabilitation, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Guo-Yan Li
- Department of Rehabilitation, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Yue Lan
- Department of Rehabilitation, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| |
Collapse
|
13
|
Jones-Tabah J, Mohammad H, Paulus EG, Clarke PBS, Hébert TE. The Signaling and Pharmacology of the Dopamine D1 Receptor. Front Cell Neurosci 2022; 15:806618. [PMID: 35110997 PMCID: PMC8801442 DOI: 10.3389/fncel.2021.806618] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 12/23/2021] [Indexed: 12/30/2022] Open
Abstract
The dopamine D1 receptor (D1R) is a Gαs/olf-coupled GPCR that is expressed in the midbrain and forebrain, regulating motor behavior, reward, motivational states, and cognitive processes. Although the D1R was initially identified as a promising drug target almost 40 years ago, the development of clinically useful ligands has until recently been hampered by a lack of suitable candidate molecules. The emergence of new non-catechol D1R agonists, biased agonists, and allosteric modulators has renewed clinical interest in drugs targeting this receptor, specifically for the treatment of motor impairment in Parkinson's Disease, and cognitive impairment in neuropsychiatric disorders. To develop better therapeutics, advances in ligand chemistry must be matched by an expanded understanding of D1R signaling across cell populations in the brain, and in disease states. Depending on the brain region, the D1R couples primarily to either Gαs or Gαolf through which it activates a cAMP/PKA-dependent signaling cascade that can regulate neuronal excitability, stimulate gene expression, and facilitate synaptic plasticity. However, like many GPCRs, the D1R can signal through multiple downstream pathways, and specific signaling signatures may differ between cell types or be altered in disease. To guide development of improved D1R ligands, it is important to understand how signaling unfolds in specific target cells, and how this signaling affects circuit function and behavior. In this review, we provide a summary of D1R-directed signaling in various neuronal populations and describe how specific pathways have been linked to physiological and behavioral outcomes. In addition, we address the current state of D1R drug development, including the pharmacology of newly developed non-catecholamine ligands, and discuss the potential utility of D1R-agonists in Parkinson's Disease and cognitive impairment.
Collapse
|
14
|
Allichon MC, Ortiz V, Pousinha P, Andrianarivelo A, Petitbon A, Heck N, Trifilieff P, Barik J, Vanhoutte P. Cell-Type-Specific Adaptions in Striatal Medium-Sized Spiny Neurons and Their Roles in Behavioral Responses to Drugs of Abuse. Front Synaptic Neurosci 2022; 13:799274. [PMID: 34970134 PMCID: PMC8712310 DOI: 10.3389/fnsyn.2021.799274] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 11/26/2021] [Indexed: 12/21/2022] Open
Abstract
Drug addiction is defined as a compulsive pattern of drug-seeking- and taking- behavior, with recurrent episodes of abstinence and relapse, and a loss of control despite negative consequences. Addictive drugs promote reinforcement by increasing dopamine in the mesocorticolimbic system, which alters excitatory glutamate transmission within the reward circuitry, thereby hijacking reward processing. Within the reward circuitry, the striatum is a key target structure of drugs of abuse since it is at the crossroad of converging glutamate inputs from limbic, thalamic and cortical regions, encoding components of drug-associated stimuli and environment, and dopamine that mediates reward prediction error and incentive values. These signals are integrated by medium-sized spiny neurons (MSN), which receive glutamate and dopamine axons converging onto their dendritic spines. MSN primarily form two mostly distinct populations based on the expression of either DA-D1 (D1R) or DA-D2 (D2R) receptors. While a classical view is that the two MSN populations act in parallel, playing antagonistic functional roles, the picture seems much more complex. Herein, we review recent studies, based on the use of cell-type-specific manipulations, demonstrating that dopamine differentially modulates dendritic spine density and synapse formation, as well as glutamate transmission, at specific inputs projecting onto D1R-MSN and D2R-MSN to shape persistent pathological behavioral in response to drugs of abuse. We also discuss the identification of distinct molecular events underlying the detrimental interplay between dopamine and glutamate signaling in D1R-MSN and D2R-MSN and highlight the relevance of such cell-type-specific molecular studies for the development of innovative strategies with potential therapeutic value for addiction. Because drug addiction is highly prevalent in patients with other psychiatric disorders when compared to the general population, we last discuss the hypothesis that shared cellular and molecular adaptations within common circuits could explain the co-occurrence of addiction and depression. We will therefore conclude this review by examining how the nucleus accumbens (NAc) could constitute a key interface between addiction and depression.
Collapse
Affiliation(s)
- Marie-Charlotte Allichon
- CNRS, UMR 8246, Neuroscience Paris Seine, Paris, France.,INSERM, UMR-S 1130, Neuroscience Paris Seine, Institute of Biology Paris Seine, Paris, France.,Sorbonne Université, UPMC Université Paris 06, UM CR18, Neuroscience Paris Seine, Paris, France
| | - Vanesa Ortiz
- Université Côte d'Azur, Nice, France.,Institut de Pharmacologie Moléculaire et Cellulaire, CNRS UMR 7275, Valbonne, France
| | - Paula Pousinha
- Université Côte d'Azur, Nice, France.,Institut de Pharmacologie Moléculaire et Cellulaire, CNRS UMR 7275, Valbonne, France
| | - Andry Andrianarivelo
- CNRS, UMR 8246, Neuroscience Paris Seine, Paris, France.,INSERM, UMR-S 1130, Neuroscience Paris Seine, Institute of Biology Paris Seine, Paris, France.,Sorbonne Université, UPMC Université Paris 06, UM CR18, Neuroscience Paris Seine, Paris, France
| | - Anna Petitbon
- Université Bordeaux, INRAE, Bordeaux INP, NutriNeuro, Bordeaux, France
| | - Nicolas Heck
- CNRS, UMR 8246, Neuroscience Paris Seine, Paris, France.,INSERM, UMR-S 1130, Neuroscience Paris Seine, Institute of Biology Paris Seine, Paris, France.,Sorbonne Université, UPMC Université Paris 06, UM CR18, Neuroscience Paris Seine, Paris, France
| | - Pierre Trifilieff
- Université Bordeaux, INRAE, Bordeaux INP, NutriNeuro, Bordeaux, France
| | - Jacques Barik
- Université Côte d'Azur, Nice, France.,Institut de Pharmacologie Moléculaire et Cellulaire, CNRS UMR 7275, Valbonne, France
| | - Peter Vanhoutte
- CNRS, UMR 8246, Neuroscience Paris Seine, Paris, France.,INSERM, UMR-S 1130, Neuroscience Paris Seine, Institute of Biology Paris Seine, Paris, France.,Sorbonne Université, UPMC Université Paris 06, UM CR18, Neuroscience Paris Seine, Paris, France
| |
Collapse
|
15
|
Saitoe M, Naganos S, Miyashita T, Matsuno M, Ueno K. A non-canonical on-demand dopaminergic transmission underlying olfactory aversive learning. Neurosci Res 2021; 178:1-9. [PMID: 34973292 DOI: 10.1016/j.neures.2021.12.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 11/16/2021] [Accepted: 12/27/2021] [Indexed: 10/19/2022]
Abstract
Dopamine (DA) is involved in various brain functions including associative learning. However, it is unclear how a small number of DA neurons appropriately regulates various brain functions. DA neurons have a large number of release sites and release DA non-specifically to a large number of target neurons in the projection area in response to the activity of DA neurons. In contrast to this "broad transmission", recent studies in Drosophila ex vivo functional imaging studies have identified "on-demand transmission" that occurs independent on activity of DA neurons and releases DA specifically onto the target neurons that have produced carbon monoxide (CO) as a retrograde signal for DA release. Whereas broad transmission modulates the global function of the target area, on-demand transmission is suitable for modulating the function of specific circuits, neurons, or synapses. In Drosophila olfactory aversive conditioning, odor and shock information are associated in the brain region called mushroom body (MB) to form olfactory aversive memory. It has been suggested that DA neurons projecting to the MB mediate the transmission of shock information and reinforcement simultaneously. However, the circuit model based on on-demand transmission proposes that transmission of shock information and reinforcement are mediated by distinct neural mechanisms; while shock transmission is glutamatergic, DA neurons mediates reinforcement. On-demand transmission provides mechanical insights into how DA neurons regulate various brain functions.
Collapse
Affiliation(s)
- Minoru Saitoe
- Learning and Memory Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya, Tokyo, 156-8506, Japan.
| | - Shintaro Naganos
- Learning and Memory Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya, Tokyo, 156-8506, Japan
| | - Tomoyuki Miyashita
- Learning and Memory Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya, Tokyo, 156-8506, Japan
| | - Motomi Matsuno
- Learning and Memory Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya, Tokyo, 156-8506, Japan
| | - Kohei Ueno
- Learning and Memory Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya, Tokyo, 156-8506, Japan
| |
Collapse
|
16
|
Ferrari E, Cardinale A, Picconi B, Gardoni F. From cell lines to pluripotent stem cells for modelling Parkinson's Disease. J Neurosci Methods 2020; 340:108741. [PMID: 32311374 DOI: 10.1016/j.jneumeth.2020.108741] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 03/25/2020] [Accepted: 04/14/2020] [Indexed: 12/13/2022]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder characterized by loss of dopaminergic (DAergic) neurons in the substantia nigra (SN) that contributes to the main motor symptoms of the disease. At present, even if several advancements have been done in the last decades, the molecular and cellular mechanisms involved in the pathogenesis are far to be fully understood. Accordingly, the establishment of reliable in vitro experimental models to investigate the early events of the pathogenesis represents a key issue in the field. However, to mimic and reproduce in vitro the complex neuronal circuitry involved in PD-associated degeneration of DAergic neurons still remains a highly challenging issue. Here we will review the in vitro PD models used in the last 25 years of research, ranging from cell lines, primary rat or mice neuronal cultures to the more recent use of human induced pluripotent stem cells (hiPSCs) and, finally, the development of 3D midbrain organoids.
Collapse
Affiliation(s)
- Elena Ferrari
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | | | - Barbara Picconi
- Università Telematica San Raffaele, Rome, Italy; IRCCS San Raffaele Pisana, Rome, Italy.
| | - Fabrizio Gardoni
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy.
| |
Collapse
|
17
|
Kumar A. Calcium Signaling During Brain Aging and Its Influence on the Hippocampal Synaptic Plasticity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1131:985-1012. [PMID: 31646542 DOI: 10.1007/978-3-030-12457-1_39] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Calcium (Ca2+) ions are highly versatile intracellular signaling molecules and are universal second messenger for regulating a variety of cellular and physiological functions including synaptic plasticity. Ca2+ homeostasis in the central nervous system endures subtle dysregulation with advancing age. Research has provided abundant evidence that brain aging is associated with altered neuronal Ca2+ regulation and synaptic plasticity mechanisms. Much of the work has focused on the hippocampus, a brain region critically involved in learning and memory, which is particularly susceptible to dysfunction during aging. The current chapter takes a specific perspective, assessing various Ca2+ sources and the influence of aging on Ca2+ sources and synaptic plasticity in the hippocampus. Integrating the knowledge of the complexity of age-related alterations in neuronal Ca2+ signaling and synaptic plasticity mechanisms will positively shape the development of highly effective therapeutics to treat brain disorders including cognitive impairment associated with aging and neurodegenerative disease.
Collapse
Affiliation(s)
- Ashok Kumar
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
18
|
Goldsmith PJ. NMDAR PAMs: Multiple Chemotypes for Multiple Binding Sites. Curr Top Med Chem 2019; 19:2239-2253. [PMID: 31660834 DOI: 10.2174/1568026619666191011095341] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 08/30/2019] [Accepted: 09/06/2019] [Indexed: 12/16/2022]
Abstract
The N-methyl-D-aspartate receptor (NMDAR) is a member of the ionotropic glutamate receptor (iGluR) family that plays a crucial role in brain signalling and development. NMDARs are nonselective cation channels that are involved with the propagation of excitatory neurotransmission signals with important effects on synaptic plasticity. NMDARs are functionally and structurally complex receptors, they exist as a family of subtypes each with its own unique pharmacological properties. Their implication in a variety of neurological and psychiatric conditions means they have been a focus of research for many decades. Disruption of NMDAR-related signalling is known to adversely affect higherorder cognitive functions (e.g. learning and memory) and the search for molecules that can recover (or even enhance) receptor output is a current strategy for CNS drug discovery. A number of positive allosteric modulators (PAMs) that specifically attempt to overcome NMDAR hypofunction have been discovered. They include various chemotypes that have been found to bind to several different binding sites within the receptor. The heterogeneity of chemotype, binding site and NMDAR subtype provide a broad landscape of ongoing opportunities to uncover new features of NMDAR pharmacology. Research on NMDARs continues to provide novel mechanistic insights into receptor activation and this review will provide a high-level overview of the research area and discuss the various chemical classes of PAMs discovered so far.
Collapse
Affiliation(s)
- Paul J Goldsmith
- Eli Lilly and Co. Ltd, Lilly Research Centre, Erl Wood Manor, Windlesham, Surrey, GU20 6PH, United Kingdom
| |
Collapse
|
19
|
Differences in GluN2B-Containing NMDA Receptors Result in Distinct Long-Term Plasticity at Ipsilateral versus Contralateral Cortico-Striatal Synapses. eNeuro 2019; 6:ENEURO.0118-19.2019. [PMID: 31744842 PMCID: PMC6883172 DOI: 10.1523/eneuro.0118-19.2019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 10/14/2019] [Accepted: 11/06/2019] [Indexed: 12/24/2022] Open
Abstract
Excitatory neurons in the primary motor cortex project bilaterally to the striatum. However, whether synaptic structure and function in ipsilateral and contralateral cortico-striatal pathways is identical or different remains largely unknown. Here, we describe that excitatory synapses in the mouse contralateral pathway have higher levels of NMDA-type of glutamate receptors (NMDARs) than those in the ipsilateral pathway, although both synapses utilize the same presynaptic vesicular glutamate transporter (VGLUT). We also show that NMDARs containing the GluN2B subunit, but not GluN2A, contribute to this difference. The altered NMDAR subunit composition in these two pathways results in opposite synaptic plasticity induced by θ-burst stimulus: long-term depression in the ipsilateral pathway and long-term potentiation (LTP) in the contralateral pathway. The standard long-term depression (LTD)-inducing protocol using paired postsynaptic and presynaptic activity triggers synaptic depression at ipsilateral pathway synapses, but not at those of the contralateral pathway. Altogether, our results provide novel and unexpected evidence for the lack of bilaterality of NMDAR-mediated synaptic transmission at cortico-striatal pathways due to differences in the expression of GluN2B subunits, which results in differences in bidirectional synaptic plasticity.
Collapse
|
20
|
Koch ET, Raymond LA. Dysfunctional striatal dopamine signaling in Huntington's disease. J Neurosci Res 2019; 97:1636-1654. [PMID: 31304622 DOI: 10.1002/jnr.24495] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 06/06/2019] [Accepted: 06/26/2019] [Indexed: 12/17/2022]
Abstract
Dopamine signaling in the striatum is critical for a variety of behaviors including movement, behavioral flexibility, response to reward and many forms of learning. Alterations to dopamine transmission contribute to pathological features of many neurological diseases, including Huntington's disease (HD). HD is an autosomal dominant genetic disorder caused by a CAG repeat expansion in the Huntingtin gene. The striatum is preferentially degenerated in HD, and this region receives dopaminergic input from the substantia nigra. Studies of HD patients and genetic rodent models have shown changes to levels of dopamine and its receptors in the striatum, and alterations in dopamine receptor signaling and modulation of other neurotransmitters, notably glutamate. Throughout his career, Dr. Michael Levine's research has furthered our understanding of dopamine signaling in the striatum of healthy rodents and HD mouse models. This review will focus on the work of his group and others in elucidating alterations to striatal dopamine signaling that contribute to pathophysiology in HD mouse models, and how these findings relate to human HD studies. We will also discuss current and potential therapeutic interventions for HD that target the dopamine system, and future research directions for this field.
Collapse
Affiliation(s)
- Ellen T Koch
- Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada.,Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada.,Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada
| | - Lynn A Raymond
- Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada.,Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
21
|
Fartootzadeh R, Azizi F, Alaei H, Reisi P. Orexin type-2 receptor blockade prevents the nicotine-induced excitation of nucleus accumbens core neurons in rats: An electrophysiological perspective. Pharmacol Rep 2019; 71:361-366. [PMID: 30831442 DOI: 10.1016/j.pharep.2018.12.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Revised: 12/11/2018] [Accepted: 12/29/2018] [Indexed: 11/29/2022]
Abstract
BACKGROUND The nucleus accumbens core (NAcc) expresses both orexin and nicotinic acetylcholine receptors (nAChRs). Orexin is among important neurotransmitters, which regulates addictive properties of drugs of abuse including nicotine. The role of orexin-2 receptor (OX2R) in the regulation of NAcc neural activity in response to nicotine has not yet been studied. Hence, in this study, we examined whether the OX2R antagonist (TCS-OX2-29) can adjust the effects of nicotine on electrical activity of NAcc neurons, in urethane-anesthetized rats, using the single unit recording. METHODS Neuronal firing of NAcc was recorded for 15 min, then TCS-OX2-29 (OX2R-antagonist; 1, 3 and 10 ng/rat) or DMSO were microinjected into NAcc, just 5 min before subcutaneous (sc) administration of nicotine (0.5 mg/kg) or saline. The spontaneous firing activity was recorded for 70 min, after nicotine injection. RESULTS The results demonstrated that nicotine significantly excites the NAcc neurons and interestingly, the administration of TCS-OX2-29 (3 and 10 ng/rat) into the NAcc, inhibited nicotine-induced increases of NAcc neuronal responses. Furthermore, administration of TCS-OX2-29 (10 ng/rat), just 5 min before sc administration of saline instead of nicotine, did not significantly alter the neuronal responses, compared to the saline-control group. CONCLUSION Our results showed that, although OX2R blockade alone did not affect neuronal activity in the NAcc, it was able to prevent the exciting effects of nicotine on NAcc neuronal activity. Therefore, we proposed that orexin has a potential modulator effect, in response to nicotine.
Collapse
Affiliation(s)
- Reza Fartootzadeh
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Fatemeh Azizi
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hojjatallah Alaei
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Parham Reisi
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
22
|
Gallo EF. Disentangling the diverse roles of dopamine D2 receptors in striatal function and behavior. Neurochem Int 2019; 125:35-46. [PMID: 30716356 DOI: 10.1016/j.neuint.2019.01.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 01/21/2019] [Accepted: 01/27/2019] [Indexed: 02/07/2023]
Abstract
Dopamine D2 receptors (D2Rs) mediate many of the actions of dopamine in the striatum, ranging from movement to the effortful pursuit of reward. Yet despite significant advances in linking D2Rs to striatal functions with pharmacological and genetic strategies in animals, how dopamine orchestrates its myriad actions on different cell populations -each expressing D2Rs- remains unclear. Furthermore, brain imaging and genetic studies in humans have consistently associated striatal D2R alterations with various neurological and neuropsychiatric disorders, but how and which D2Rs are involved in each case is poorly understood. Therefore, a critical first step is to engage in a refined and systematic investigation of the impact of D2R function on specific striatal cells, circuits, and behaviors. Here, I will review recent efforts, primarily in animal models, aimed at unlocking the complex and heterogeneous roles of D2Rs in striatum.
Collapse
Affiliation(s)
- Eduardo F Gallo
- Department of Biological Sciences, Fordham University, Bronx, NY, USA.
| |
Collapse
|
23
|
Balance between dopamine and adenosine signals regulates the PKA/Rap1 pathway in striatal medium spiny neurons. Neurochem Int 2019; 122:8-18. [DOI: 10.1016/j.neuint.2018.10.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 10/02/2018] [Accepted: 10/03/2018] [Indexed: 12/19/2022]
|
24
|
Armañanzas R. Revealing post-transcriptional microRNA-mRNA regulations in Alzheimer's disease through ensemble graphs. BMC Genomics 2018; 19:668. [PMID: 30255799 PMCID: PMC6157163 DOI: 10.1186/s12864-018-5025-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND In silico investigations on the integration of multiple datasets are in need of higher statistical power methods to unveil secondary findings that were hidden from the initial analyses. We present here a novel method for the network analysis of messenger RNA post-translational regulation by microRNA molecules. The method integrates expression data and sequence binding predictions through a set of sound machine learning techniques, forwarding all results to an ensemble graph of regulations. RESULTS Bayesian network classifiers are induced based on a pool of ensemble graphs with ascending order of complexity. Individual goodness-of-fit and classification performances are evaluated for each learned model. As a testbed, four Alzheimer's disease datasets are integrated using the new approach, achieving top values of 0.9794 ± 0.01 for the area under the receiver operating characteristic curve and 0.9439 ± 0.0234 for the prediction accuracy. CONCLUSIONS Post-transcriptional regulations found by the optimal network classifier concur with previous literature findings. Furthermore, additional network structures suggest previously unreported regulations in the state of the art of Alzheimer's research. The quantitative performance as well as sound biological findings provide confidence in the ensemble approach and encourage similar integrative analyses for other conditions.
Collapse
Affiliation(s)
- Rubén Armañanzas
- Department of Bioengineering, Krasnow Institute for Advanced Study, George Mason University, 4400 University Dr, MS2A1, Fairfax, 22030, VA, USA.
| |
Collapse
|
25
|
Kumar A, Foster TC. Alteration in NMDA Receptor Mediated Glutamatergic Neurotransmission in the Hippocampus During Senescence. Neurochem Res 2018; 44:38-48. [PMID: 30209673 DOI: 10.1007/s11064-018-2634-4] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 09/07/2018] [Accepted: 09/08/2018] [Indexed: 12/17/2022]
Abstract
Glutamate is the primary excitatory neurotransmitter in neurons and glia. N-methyl-D-aspartate (NMDA), α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA), and kainate receptors are major ionotropic glutamate receptors. Glutamatergic neurotransmission is strongly linked with Ca2+ homeostasis. Research has provided ample evidence that brain aging is associated with altered glutamatergic neurotransmission and Ca2+ dysregulation. Much of the work has focused on the hippocampus, a brain region critically involved in learning and memory, which is particularly susceptible to dysfunction during senescence. The current review examines Ca2+ regulation with a focus on the NMDA receptors in the hippocampus. Integrating the knowledge of the complexity of age-related alterations in Ca2+ homeostasis and NMDA receptor-mediated glutamatergic neurotransmission will positively shape the development of highly effective therapeutics to treat brain disorders including cognitive impairment.
Collapse
Affiliation(s)
- Ashok Kumar
- Department of Neuroscience, McKnight Brain Institute, University of Florida, PO Box 100244, Gainesville, FL, 32610-0244, USA.
| | - Thomas C Foster
- Department of Neuroscience, McKnight Brain Institute, University of Florida, PO Box 100244, Gainesville, FL, 32610-0244, USA.
- Genetics and Genomics Program, University of Florida, Gainesville, FL, 32611, USA.
| |
Collapse
|
26
|
Jakaria M, Park SY, Haque ME, Karthivashan G, Kim IS, Ganesan P, Choi DK. Neurotoxic Agent-Induced Injury in Neurodegenerative Disease Model: Focus on Involvement of Glutamate Receptors. Front Mol Neurosci 2018; 11:307. [PMID: 30210294 PMCID: PMC6123546 DOI: 10.3389/fnmol.2018.00307] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 08/13/2018] [Indexed: 12/13/2022] Open
Abstract
Glutamate receptors play a crucial role in the central nervous system and are implicated in different brain disorders. They play a significant role in the pathogenesis of neurodegenerative diseases (NDDs) such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis. Although many studies on NDDs have been conducted, their exact pathophysiological characteristics are still not fully understood. In in vivo and in vitro models of neurotoxic-induced NDDs, neurotoxic agents are used to induce several neuronal injuries for the purpose of correlating them with the pathological characteristics of NDDs. Moreover, therapeutic drugs might be discovered based on the studies employing these models. In NDD models, different neurotoxic agents, namely, kainic acid, domoic acid, glutamate, β-N-Methylamino-L-alanine, amyloid beta, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, 1-methyl-4-phenylpyridinium, rotenone, 3-Nitropropionic acid and methamphetamine can potently impair both ionotropic and metabotropic glutamate receptors, leading to the progression of toxicity. Many other neurotoxic agents mainly affect the functions of ionotropic glutamate receptors. We discuss particular neurotoxic agents that can act upon glutamate receptors so as to effectively mimic NDDs. The correlation of neurotoxic agent-induced disease characteristics with glutamate receptors would aid the discovery and development of therapeutic drugs for NDDs.
Collapse
Affiliation(s)
- Md. Jakaria
- Department of Applied Life Sciences, Graduate School, Konkuk University, Chungju, South Korea
| | - Shin-Young Park
- Department of Applied Life Sciences, Graduate School, Konkuk University, Chungju, South Korea
| | - Md. Ezazul Haque
- Department of Applied Life Sciences, Graduate School, Konkuk University, Chungju, South Korea
| | - Govindarajan Karthivashan
- Department of Integrated Bioscience and Biotechnology, College of Biomedical and Health Sciences, Research Institute of Inflammatory Diseases (RID), Konkuk University, Chungju, South Korea
| | - In-Su Kim
- Department of Integrated Bioscience and Biotechnology, College of Biomedical and Health Sciences, Research Institute of Inflammatory Diseases (RID), Konkuk University, Chungju, South Korea
| | - Palanivel Ganesan
- Department of Integrated Bioscience and Biotechnology, College of Biomedical and Health Sciences, Research Institute of Inflammatory Diseases (RID), Konkuk University, Chungju, South Korea
- Nanotechnology Research Center, Konkuk University, Chungju, South Korea
| | - Dong-Kug Choi
- Department of Applied Life Sciences, Graduate School, Konkuk University, Chungju, South Korea
- Department of Integrated Bioscience and Biotechnology, College of Biomedical and Health Sciences, Research Institute of Inflammatory Diseases (RID), Konkuk University, Chungju, South Korea
- Nanotechnology Research Center, Konkuk University, Chungju, South Korea
| |
Collapse
|
27
|
You H, Mariani LL, Mangone G, Le Febvre de Nailly D, Charbonnier-Beaupel F, Corvol JC. Molecular basis of dopamine replacement therapy and its side effects in Parkinson's disease. Cell Tissue Res 2018. [PMID: 29516217 DOI: 10.1007/s00441-018-2813-2] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
There is currently no cure for Parkinson's disease. The symptomatic therapeutic strategy essentially relies on dopamine replacement whose efficacy was demonstrated more than 50 years ago following the introduction of the dopamine precursor, levodopa. The spectacular antiparkinsonian effect of levodopa is, however, balanced by major limitations including the occurrence of motor complications related to its particular pharmacokinetic and pharmacodynamic properties. Other therapeutic strategies have thus been developed to overcome these problems such as the use of dopamine receptor agonists, dopamine metabolism inhibitors and non-dopaminergic drugs. Here we review the pharmacology and molecular mechanisms of dopamine replacement therapy in Parkinson's disease, both at the presynaptic and postsynaptic levels. The perspectives in terms of novel drug development and prediction of drug response for a more personalised medicine will be discussed.
Collapse
Affiliation(s)
- Hana You
- Sorbonne Université, UPMC Univ Paris 06, UMR S 1127, ICM, Hôpital Pitié-Salpêtrière, Paris, France.,INSERM, Unit 1127, CIC 1422, NS-PARK/FCRIN, Hôpital Pitié-Salpêtrière, Paris, France.,CNRS, Unit 7225, Hôpital Pitié-Salpêtrière, Paris, France.,Assistance Publique Hôpitaux de Paris, Department of Neurology, Hôpital Pitié-Salpêtrière, Paris, France.,Department of Neurology, University Hospital (Inselspital) and University of Bern, Freiburgstrasse 18, 3010, Bern, Switzerland
| | - Louise-Laure Mariani
- Sorbonne Université, UPMC Univ Paris 06, UMR S 1127, ICM, Hôpital Pitié-Salpêtrière, Paris, France.,INSERM, Unit 1127, CIC 1422, NS-PARK/FCRIN, Hôpital Pitié-Salpêtrière, Paris, France.,CNRS, Unit 7225, Hôpital Pitié-Salpêtrière, Paris, France.,Assistance Publique Hôpitaux de Paris, Department of Neurology, Hôpital Pitié-Salpêtrière, Paris, France
| | - Graziella Mangone
- Sorbonne Université, UPMC Univ Paris 06, UMR S 1127, ICM, Hôpital Pitié-Salpêtrière, Paris, France.,INSERM, Unit 1127, CIC 1422, NS-PARK/FCRIN, Hôpital Pitié-Salpêtrière, Paris, France.,CNRS, Unit 7225, Hôpital Pitié-Salpêtrière, Paris, France.,Assistance Publique Hôpitaux de Paris, Department of Neurology, Hôpital Pitié-Salpêtrière, Paris, France
| | - Delphine Le Febvre de Nailly
- INSERM, Unit 1127, CIC 1422, NS-PARK/FCRIN, Hôpital Pitié-Salpêtrière, Paris, France.,Assistance Publique Hôpitaux de Paris, Department of Pharmacy, Hôpital Pitié-Salpêtrière, Paris, France
| | - Fanny Charbonnier-Beaupel
- Assistance Publique Hôpitaux de Paris, Department of Pharmacy, Hôpital Pitié-Salpêtrière, Paris, France
| | - Jean-Christophe Corvol
- Sorbonne Université, UPMC Univ Paris 06, UMR S 1127, ICM, Hôpital Pitié-Salpêtrière, Paris, France. .,INSERM, Unit 1127, CIC 1422, NS-PARK/FCRIN, Hôpital Pitié-Salpêtrière, Paris, France. .,CNRS, Unit 7225, Hôpital Pitié-Salpêtrière, Paris, France. .,Assistance Publique Hôpitaux de Paris, Department of Neurology, Hôpital Pitié-Salpêtrière, Paris, France. .,CIC Neurosciences, ICM building, Hôpital Pitié-Salpêtrière, 47/83 Boulevard de l'Hôpital, 75013, Paris, France.
| |
Collapse
|
28
|
Lindroos R, Dorst MC, Du K, Filipović M, Keller D, Ketzef M, Kozlov AK, Kumar A, Lindahl M, Nair AG, Pérez-Fernández J, Grillner S, Silberberg G, Hellgren Kotaleski J. Basal Ganglia Neuromodulation Over Multiple Temporal and Structural Scales-Simulations of Direct Pathway MSNs Investigate the Fast Onset of Dopaminergic Effects and Predict the Role of Kv4.2. Front Neural Circuits 2018; 12:3. [PMID: 29467627 PMCID: PMC5808142 DOI: 10.3389/fncir.2018.00003] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 01/09/2018] [Indexed: 12/16/2022] Open
Abstract
The basal ganglia are involved in the motivational and habitual control of motor and cognitive behaviors. Striatum, the largest basal ganglia input stage, integrates cortical and thalamic inputs in functionally segregated cortico-basal ganglia-thalamic loops, and in addition the basal ganglia output nuclei control targets in the brainstem. Striatal function depends on the balance between the direct pathway medium spiny neurons (D1-MSNs) that express D1 dopamine receptors and the indirect pathway MSNs that express D2 dopamine receptors. The striatal microstructure is also divided into striosomes and matrix compartments, based on the differential expression of several proteins. Dopaminergic afferents from the midbrain and local cholinergic interneurons play crucial roles for basal ganglia function, and striatal signaling via the striosomes in turn regulates the midbrain dopaminergic system directly and via the lateral habenula. Consequently, abnormal functions of the basal ganglia neuromodulatory system underlie many neurological and psychiatric disorders. Neuromodulation acts on multiple structural levels, ranging from the subcellular level to behavior, both in health and disease. For example, neuromodulation affects membrane excitability and controls synaptic plasticity and thus learning in the basal ganglia. However, it is not clear on what time scales these different effects are implemented. Phosphorylation of ion channels and the resulting membrane effects are typically studied over minutes while it has been shown that neuromodulation can affect behavior within a few hundred milliseconds. So how do these seemingly contradictory effects fit together? Here we first briefly review neuromodulation of the basal ganglia, with a focus on dopamine. We furthermore use biophysically detailed multi-compartmental models to integrate experimental data regarding dopaminergic effects on individual membrane conductances with the aim to explain the resulting cellular level dopaminergic effects. In particular we predict dopaminergic effects on Kv4.2 in D1-MSNs. Finally, we also explore dynamical aspects of the onset of neuromodulation effects in multi-scale computational models combining biochemical signaling cascades and multi-compartmental neuron models.
Collapse
Affiliation(s)
- Robert Lindroos
- Department of Neuroscience, Nobel Institute for Neurophysiology, Stockholm, Sweden
| | - Matthijs C. Dorst
- Department of Neuroscience, Nobel Institute for Neurophysiology, Stockholm, Sweden
| | - Kai Du
- Department of Neuroscience, Nobel Institute for Neurophysiology, Stockholm, Sweden
| | - Marko Filipović
- Bernstein Center Freiburg, University of Freiburg, Freiburg im Breisgau, Germany
| | - Daniel Keller
- Blue Brain Project, Ecole Polytechnique Fédérale de Lausanne, Geneva, Switzerland
| | - Maya Ketzef
- Department of Neuroscience, Nobel Institute for Neurophysiology, Stockholm, Sweden
| | - Alexander K. Kozlov
- Science for Life Laboratory, School of Electrical Engineering and Computer Science, KTH Royal Institute of Technology, Solna, Sweden
| | - Arvind Kumar
- Bernstein Center Freiburg, University of Freiburg, Freiburg im Breisgau, Germany
- Department Computational Science and Technology, School of Electrical Engineering and Computer Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Mikael Lindahl
- Science for Life Laboratory, School of Electrical Engineering and Computer Science, KTH Royal Institute of Technology, Solna, Sweden
| | - Anu G. Nair
- Science for Life Laboratory, School of Electrical Engineering and Computer Science, KTH Royal Institute of Technology, Solna, Sweden
| | - Juan Pérez-Fernández
- Department of Neuroscience, Nobel Institute for Neurophysiology, Stockholm, Sweden
| | - Sten Grillner
- Department of Neuroscience, Nobel Institute for Neurophysiology, Stockholm, Sweden
| | - Gilad Silberberg
- Department of Neuroscience, Nobel Institute for Neurophysiology, Stockholm, Sweden
| | - Jeanette Hellgren Kotaleski
- Department of Neuroscience, Nobel Institute for Neurophysiology, Stockholm, Sweden
- Science for Life Laboratory, School of Electrical Engineering and Computer Science, KTH Royal Institute of Technology, Solna, Sweden
| |
Collapse
|
29
|
Rizzi G, Tan KR. Dopamine and Acetylcholine, a Circuit Point of View in Parkinson's Disease. Front Neural Circuits 2017; 11:110. [PMID: 29311846 PMCID: PMC5744635 DOI: 10.3389/fncir.2017.00110] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Accepted: 12/14/2017] [Indexed: 12/30/2022] Open
Abstract
Data from the World Health Organization (National Institute on Aging, 2011) and the National Institutes of Health (He et al., 2016) predicts that while today the worldwide population over 65 years of age is estimated around 8.5%, this number will reach an astounding 17% by 2050. In this framework, solving current neurodegenerative diseases primarily associated with aging becomes more pressing than ever. In 2017, we celebrate a grim 200th anniversary since the very first description of Parkinson’s disease (PD) and its related symptomatology. Two centuries after this debilitating disease was first identified, finding a cure remains a hopeful goal rather than an attainable objective on the horizon. Tireless work has provided insight into the characterization and progression of the disease down to a molecular level. We now know that the main motor deficits associated with PD arise from the almost total loss of dopaminergic cells in the substantia nigra pars compacta. A concomitant loss of cholinergic cells entails a cognitive decline in these patients, and current therapies are only partially effective, often inducing side-effects after a prolonged treatment. This review covers some of the recent developments in the field of Basal Ganglia (BG) function in physiology and pathology, with a particular focus on the two main neuromodulatory systems known to be severely affected in PD, highlighting some of the remaining open question from three main stand points: - Heterogeneity of midbrain dopamine neurons. - Pairing of dopamine (DA) sub-circuits. - Dopamine-Acetylcholine (ACh) interaction. A vast amount of knowledge has been accumulated over the years from experimental conditions, but very little of it is reflected or used at a translational or clinical level. An initiative to implement the knowledge that is emerging from circuit-based approaches to tackle neurodegenerative disorders like PD will certainly be tremendously beneficial.
Collapse
Affiliation(s)
| | - Kelly R Tan
- Biozentrum, University of Basel, Basel, Switzerland
| |
Collapse
|
30
|
Phamluong K, Darcq E, Wu S, Sakhai SA, Ron D. Fyn Signaling Is Compartmentalized to Dopamine D1 Receptor Expressing Neurons in the Dorsal Medial Striatum. Front Mol Neurosci 2017; 10:273. [PMID: 28912680 PMCID: PMC5583218 DOI: 10.3389/fnmol.2017.00273] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 08/11/2017] [Indexed: 01/24/2023] Open
Abstract
The tyrosine kinase Fyn plays an important role in synaptic plasticity, learning, and memory. Here we report that Fyn is activated in response to 15 min D1 receptor (D1R) but not D2 receptor (D2R) stimulation specifically in the dorsomedial striatum (DMS) of mice but not in the other substriatal regions, the dorsolateral striatum (DLS), and the nucleus accumbens (NAc). Once activated Fyn phosphorylates its substrate GluN2B, and we show that GluN2B is phosphorylated only in the DMS but not in the other striatal regions. Striatal neurons are divided into D1R expressing medium spiny neurons (MSNs) and D2R expressing MSNs. Thus, to explore the cell-type specificity of this signaling pathway in the DMS, we developed a Cre-dependent Flip Excision (FLEX) approach to knockdown Fyn in D1R MSNs or D2R MSNs, and proved that the D1R-dependent Fyn activation is localized to DMS D1R MSNs. Importantly, we provide evidence to suggest that the differential association of Fyn and GluN2B with the scaffolding RACK1 is due to the differential localization of Fyn in lipid rafts.Our data further suggest that the differential cholesterol content in the three striatal regions may determine the region specificity of this signaling pathway. Together, our data show that the D1R-dependent Fyn/GluN2B pathway is selectively activated in D1R expressing MSNs in the DMS, and that the brain region specificity of pathway depends on the molecular and cellular compartmentalization of Fyn and GluN2B.
Collapse
Affiliation(s)
- Khanhky Phamluong
- Department of Neurology, University of California San FranciscoSan Francisco, CA, United States
| | - Emmanuel Darcq
- Department of Neurology, University of California San FranciscoSan Francisco, CA, United States
| | - Su Wu
- Department of Neurology, University of California San FranciscoSan Francisco, CA, United States
| | - Samuel A Sakhai
- Department of Neurology, University of California San FranciscoSan Francisco, CA, United States
| | - Dorit Ron
- Department of Neurology, University of California San FranciscoSan Francisco, CA, United States
| |
Collapse
|
31
|
Pyk2 modulates hippocampal excitatory synapses and contributes to cognitive deficits in a Huntington's disease model. Nat Commun 2017; 8:15592. [PMID: 28555636 PMCID: PMC5459995 DOI: 10.1038/ncomms15592] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 04/11/2017] [Indexed: 12/11/2022] Open
Abstract
The structure and function of spines and excitatory synapses are under the dynamic control of multiple signalling networks. Although tyrosine phosphorylation is involved, its regulation and importance are not well understood. Here we study the role of Pyk2, a non-receptor calcium-dependent protein-tyrosine kinase highly expressed in the hippocampus. Hippocampal-related learning and CA1 long-term potentiation are severely impaired in Pyk2-deficient mice and are associated with alterations in NMDA receptors, PSD-95 and dendritic spines. In cultured hippocampal neurons, Pyk2 has autophosphorylation-dependent and -independent roles in determining PSD-95 enrichment and spines density. Pyk2 levels are decreased in the hippocampus of individuals with Huntington and in the R6/1 mouse model of the disease. Normalizing Pyk2 levels in the hippocampus of R6/1 mice rescues memory deficits, spines pathology and PSD-95 localization. Our results reveal a role for Pyk2 in spine structure and synaptic function, and suggest that its deficit contributes to Huntington's disease cognitive impairments.
Collapse
|
32
|
Rodrigues S, Salum C, Ferreira TL. Dorsal striatum D1-expressing neurons are involved with sensorimotor gating on prepulse inhibition test. J Psychopharmacol 2017; 31:505-513. [PMID: 28114835 DOI: 10.1177/0269881116686879] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Prepulse inhibition (PPI) is a behavioral test in which the startle reflex response to a high-intensity stimulus (pulse) is inhibited by the prior presentation of a weak stimulus (prepulse). The classic neural circuitry that mediates startle response is localized in the brainstem; however, recent studies point to the contribution of structures involved in higher cognitive functions in regulating the sensorimotor gating, particularly forebrain regions innervated by dopaminergic nuclei. The aim of the present study was to verify the role of dorsal striatum (DS) and dopaminergic transmitting mediated by D1 and D2 receptors on PPI test in rats. DS inactivation induced by muscimol injection did not affect PPI (%PPI and startle response), although it impaired the locomotor activity and caused catalepsy. Infusion of D1-like antagonist SCH23390 impaired %PPI but did not disturb the startle response and locomotor activity evaluated immediately after PPI test. D2 antagonist microinjection (sulpiride) did not affect %PPI and startle response, but impaired motor activity. These results point to an important role of DS, probably mediated by direct basal ganglia pathway, on modulation of sensorimotor gating, in accordance with clinical studies showing PPI deficits in schizophrenia, Tourette syndrome, and compulsive disorders - pathologies related to basal ganglia dysfunctions.
Collapse
Affiliation(s)
- Samanta Rodrigues
- Centro de Matemática Computação e Cognição, Universidade Federal do ABC, São Bernardo do Campo, Brazil
| | - Cristiane Salum
- Centro de Matemática Computação e Cognição, Universidade Federal do ABC, São Bernardo do Campo, Brazil
| | - Tatiana L Ferreira
- Centro de Matemática Computação e Cognição, Universidade Federal do ABC, São Bernardo do Campo, Brazil
| |
Collapse
|
33
|
Sun Y, Zhan L, Cheng X, Zhang L, Hu J, Gao Z. The Regulation of GluN2A by Endogenous and Exogenous Regulators in the Central Nervous System. Cell Mol Neurobiol 2017; 37:389-403. [PMID: 27255970 DOI: 10.1007/s10571-016-0388-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Accepted: 05/25/2016] [Indexed: 12/25/2022]
Abstract
The NMDA receptor is the most widely studied ionotropic glutamate receptor, and it is central to many physiological and pathophysiological processes in the central nervous system. GluN2A is one of the two main types of GluN2 NMDA receptor subunits in the forebrain. The proper activity of GluN2A is important to brain function, as the abnormal regulation of GluN2A may induce some neuropsychiatric disorders. This review will examine the regulation of GluN2A by endogenous and exogenous regulators in the central nervous system.
Collapse
Affiliation(s)
- Yongjun Sun
- Department of Pharmacy, Hebei University of Science and Technology, Yuhua East Road 70, Shijiazhuang, 050018, People's Republic of China
- Hebei Research Center of Pharmaceutical and Chemical Engineering, Hebei University of Science and Technology, Shijiazhuang, 050018, People's Republic of China
| | - Liying Zhan
- Department of Pharmacy, Hebei University of Science and Technology, Yuhua East Road 70, Shijiazhuang, 050018, People's Republic of China
| | - Xiaokun Cheng
- North China Pharmaceutical Group New Drug Research and Development Co., Ltd, Shijiazhuang, 050015, People's Republic of China
| | - Linan Zhang
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, 050017, People's Republic of China
| | - Jie Hu
- School of Nursing, Hebei Medical University, Shijiazhuang, 050017, People's Republic of China
| | - Zibin Gao
- Department of Pharmacy, Hebei University of Science and Technology, Yuhua East Road 70, Shijiazhuang, 050018, People's Republic of China.
- Hebei Research Center of Pharmaceutical and Chemical Engineering, Hebei University of Science and Technology, Shijiazhuang, 050018, People's Republic of China.
- State Key Laboratory Breeding Base-Hebei Province Key Laboratory of Molecular Chemistry for Drug, Shijiazhuang, 050018, People's Republic of China.
| |
Collapse
|
34
|
Li YJ, Ping XJ, Qi C, Shen F, Sun LL, Sun XW, Ge FF, Xing GG, Cui CL. Re-exposure to morphine-associated context facilitated long-term potentiation in the vSUB-NAc glutamatergic pathway via GluN2B-containing receptor activation. Addict Biol 2017; 22:435-445. [PMID: 26692025 DOI: 10.1111/adb.12343] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 09/29/2015] [Accepted: 11/04/2015] [Indexed: 12/28/2022]
Abstract
The glutamatergic projection from the ventral subiculum of the hippocampus (vSUB) to the nucleus accumbens (NAc) shell has been reported to play a key role in drug-related behavior. The GluN2B subunit of N-methyl-D-aspartate receptors (NMDARs) in the NAc can be selectively elevated after the retrieval of drug-conditioned memory. However, whether the increased GluN2B-containing NMDARs (GluN2B-NMDARs) are able to alter the synaptic plasticity of the vSUB-NAc glutamatergic pathway remains unclear. Here, we found that the long-term potentiation (LTP) in the vSUB-NAc pathway was facilitated and the GluN2B subunit protein level was elevated in synaptoneurosomes of the NAc shell, but not in the core, following morphine-induced conditioned place preference (CPP) expression in rats. The facilitated LTP was prevented by the GluN2B-NMDAR antagonist RO25-6981. Also, a neurochemical disconnection following microinjection of RO25-6981 into the NAc shell, plus microinfusion of GABA agonist baclofen and muscimol into the contralateral vSUB prevented the expression of morphine-induced CPP. These findings suggest that the retrieval of drug-associated memory potentiated synaptic plasticity in the vSUB-NAc pathway, which was dependent on GluN2B-NMDAR activation in the NAc shell. These findings provide a new explanation for the mechanisms that underlie the morphine-associated-context memory. The GluN2B-NMDARs may be regarded as a potential target for erasing morphine-related memory.
Collapse
Affiliation(s)
- Yi-Jing Li
- Neuroscience Research Institute; Peking University; Beijing China
- Department of Neurobiology, School of Basic Medical Sciences; Peking University; Beijing China
- Key Laboratory for Neuroscience; Ministry of Education; Beijing China
- Key Laboratory for Neuroscience; Ministry of National Health and Family Planning Commission; Beijing China
| | - Xing-Jie Ping
- Neuroscience Research Institute; Peking University; Beijing China
- Department of Neurobiology, School of Basic Medical Sciences; Peking University; Beijing China
- Key Laboratory for Neuroscience; Ministry of Education; Beijing China
- Key Laboratory for Neuroscience; Ministry of National Health and Family Planning Commission; Beijing China
| | - Chong Qi
- Neuroscience Research Institute; Peking University; Beijing China
- Department of Neurobiology, School of Basic Medical Sciences; Peking University; Beijing China
- Key Laboratory for Neuroscience; Ministry of Education; Beijing China
- Key Laboratory for Neuroscience; Ministry of National Health and Family Planning Commission; Beijing China
| | - Fang Shen
- Neuroscience Research Institute; Peking University; Beijing China
- Department of Neurobiology, School of Basic Medical Sciences; Peking University; Beijing China
- Key Laboratory for Neuroscience; Ministry of Education; Beijing China
- Key Laboratory for Neuroscience; Ministry of National Health and Family Planning Commission; Beijing China
| | - Lin-Lin Sun
- Neuroscience Research Institute; Peking University; Beijing China
- Department of Neurobiology, School of Basic Medical Sciences; Peking University; Beijing China
- Key Laboratory for Neuroscience; Ministry of Education; Beijing China
- Key Laboratory for Neuroscience; Ministry of National Health and Family Planning Commission; Beijing China
| | - Xiao-Wei Sun
- Neuroscience Research Institute; Peking University; Beijing China
- Department of Neurobiology, School of Basic Medical Sciences; Peking University; Beijing China
- Key Laboratory for Neuroscience; Ministry of Education; Beijing China
- Key Laboratory for Neuroscience; Ministry of National Health and Family Planning Commission; Beijing China
| | - Fei-Fei Ge
- Neuroscience Research Institute; Peking University; Beijing China
- Department of Neurobiology, School of Basic Medical Sciences; Peking University; Beijing China
- Key Laboratory for Neuroscience; Ministry of Education; Beijing China
- Key Laboratory for Neuroscience; Ministry of National Health and Family Planning Commission; Beijing China
| | - Guo-Gang Xing
- Neuroscience Research Institute; Peking University; Beijing China
- Department of Neurobiology, School of Basic Medical Sciences; Peking University; Beijing China
- Key Laboratory for Neuroscience; Ministry of Education; Beijing China
- Key Laboratory for Neuroscience; Ministry of National Health and Family Planning Commission; Beijing China
| | - Cai-Lian Cui
- Neuroscience Research Institute; Peking University; Beijing China
- Department of Neurobiology, School of Basic Medical Sciences; Peking University; Beijing China
- Key Laboratory for Neuroscience; Ministry of Education; Beijing China
- Key Laboratory for Neuroscience; Ministry of National Health and Family Planning Commission; Beijing China
| |
Collapse
|
35
|
Li ML, Gulchina Y, Monaco SA, Xing B, Ferguson BR, Li YC, Li F, Hu XQ, Gao WJ. Juvenile treatment with a novel mGluR2 agonist/mGluR3 antagonist compound, LY395756, reverses learning deficits and cognitive flexibility impairments in adults in a neurodevelopmental model of schizophrenia. Neurobiol Learn Mem 2017; 140:52-61. [PMID: 28213064 DOI: 10.1016/j.nlm.2017.02.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 02/03/2017] [Accepted: 02/09/2017] [Indexed: 01/23/2023]
Abstract
Schizophrenia (SCZ) is a neurodevelopmental psychiatric disorder, in which cognitive function becomes disrupted at early stages of the disease. Although the mechanisms underlying cognitive impairments remain unclear, N-methyl-D-aspartate receptors (NMDAR) hypofunctioning in the prefrontal cortex (PFC) has been implicated. Moreover, cognitive symptoms in SCZ are usually unresponsive to treatment with current antipsychotics and by onset, disruption of the dopamine system, not NMDAR hypofunctioning, dominates the symptoms. Therefore, treating cognitive deficits at an early stage is a realistic approach. In this study, we tested whether an early treatment targeting mGluR2 would be effective in ameliorating cognitive impairments in the methylazoxymethanol acetate (MAM) model of SCZ. We investigated the effects of an mGluR2 agonist/mGluR3 antagonist, LY395756 (LY39), on the NMDAR expression and function in juveniles, as well as cognitive deficits in adult rats after juvenile treatment. We found that gestational MAM exposure induced a significant decrease in total protein levels of the NMDAR subunit, NR2B, and a significant increase of pNR2BTyr1472 in the juvenile rat PFC. Treatment with LY39 in juvenile MAM-exposed rats effectively recovered the disrupted NMDAR expression. Furthermore, a subchronic LY39 treatment in juvenile MAM-exposed rats also alleviated the learning deficits and cognitive flexibility impairments when tested with a cross-maze based set-shifting task in adults. Therefore, our study demonstrates that targeting dysfunctional NMDARs with an mGluR2 agonist during the early stage of SCZ could be an effective strategy in preventing the development and progression in addition to ameliorating cognitive impairments of SCZ.
Collapse
Affiliation(s)
- Meng-Lin Li
- Department of Rehabilitation, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, China; Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA; Department of Rehabilitation, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou 510180, China
| | - Yelena Gulchina
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Sarah A Monaco
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Bo Xing
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Brielle R Ferguson
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Yan-Chun Li
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Feng Li
- Department of Neurobiology and Anatomy, Zhongshan School of Medicine, Sun Yat-Sen University, 510275 Guangzhou, China.
| | - Xi-Quan Hu
- Department of Rehabilitation, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, China.
| | - Wen-Jun Gao
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA.
| |
Collapse
|
36
|
Dopamine promotes NMDA receptor hypofunction in the retina through D 1 receptor-mediated Csk activation, Src inhibition and decrease of GluN2B phosphorylation. Sci Rep 2017; 7:40912. [PMID: 28098256 PMCID: PMC5241882 DOI: 10.1038/srep40912] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 12/12/2016] [Indexed: 11/21/2022] Open
Abstract
Dopamine and glutamate are critical neurotransmitters involved in light-induced synaptic activity in the retina. In brain neurons, dopamine D1 receptors (D1Rs) and the cytosolic protein tyrosine kinase Src can, independently, modulate the behavior of NMDA-type glutamate receptors (NMDARs). Here we studied the interplay between D1Rs, Src and NMDARs in retinal neurons. We reveal that dopamine-mediated D1R stimulation provoked NMDAR hypofunction in retinal neurons by attenuating NMDA-gated currents, by preventing NMDA-elicited calcium mobilization and by decreasing the phosphorylation of NMDAR subunit GluN2B. This dopamine effect was dependent on upregulation of the canonical D1R/adenylyl cyclase/cAMP/PKA pathway, of PKA-induced activation of C-terminal Src kinase (Csk) and of Src inhibition. Accordingly, knocking down Csk or overexpressing a Csk phosphoresistant Src mutant abrogated the dopamine-induced NMDAR hypofunction. Overall, the interplay between dopamine and NMDAR hypofunction, through the D1R/Csk/Src/GluN2B pathway, might impact on light-regulated synaptic activity in retinal neurons.
Collapse
|
37
|
Bergado Acosta JR, Kahl E, Kogias G, Uzuneser TC, Fendt M. Relief learning requires a coincident activation of dopamine D1 and NMDA receptors within the nucleus accumbens. Neuropharmacology 2016; 114:58-66. [PMID: 27894877 DOI: 10.1016/j.neuropharm.2016.11.022] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 11/03/2016] [Accepted: 11/24/2016] [Indexed: 01/28/2023]
Abstract
Relief learning is the association of a stimulus with the offset of an aversive event. Later, the now conditioned relief stimulus induces appetitive-like behavioral changes. We previously demonstrated that the NMDA receptors within the nucleus accumbens (NAC) are involved in relief learning. The NAC is also important for reward learning and it has been shown that reward learning is mediated by an interaction of accumbal dopamine and NMDA glutamate receptors. Since conditioned relief has reward-like properties, we hypothesized that (a) acquisition of relief learning requires the activation of dopamine D1 receptors in the NAC, and (b) if D1 receptors are involved in this process as expected, a concurrent dopamine D1 and NMDA receptor activation may mediate this learning. The present study tested these hypotheses. Therefore, rats received intra-NAC injections of the dopamine D1 receptor antagonist SCH23390 and the NMDA antagonist AP5, either separately or together, at different time points of a relief conditioning procedure. First, we showed that SCH23390 dose-dependently blocked acquisition and the expression of conditioned relief. Next, we demonstrated that co-injections of SCH23390 and AP5 into the NAC, at doses that were ineffective when applied separately, blocked acquisition but not consolidation or expression of relief learning. Notably, neither of the injections affected the locomotor response of the animals to the aversive stimuli suggesting that their perception is not changed. This data indicates that a co-activation of dopamine D1 and NMDA receptors in the NAC is required for acquisition of relief learning.
Collapse
Affiliation(s)
- Jorge R Bergado Acosta
- Institute for Pharmacology and Toxicology, Otto-von-Guericke University Magdeburg, Germany
| | - Evelyn Kahl
- Institute for Pharmacology and Toxicology, Otto-von-Guericke University Magdeburg, Germany
| | - Georgios Kogias
- Institute for Pharmacology and Toxicology, Otto-von-Guericke University Magdeburg, Germany; Integrative Neuroscience Program, Otto-von-Guericke University Magdeburg, Germany
| | - Taygun C Uzuneser
- Institute for Pharmacology and Toxicology, Otto-von-Guericke University Magdeburg, Germany; Integrative Neuroscience Program, Otto-von-Guericke University Magdeburg, Germany
| | - Markus Fendt
- Institute for Pharmacology and Toxicology, Otto-von-Guericke University Magdeburg, Germany; Center of Behavioral Brain Sciences, Otto-von-Guericke University Magdeburg, Germany.
| |
Collapse
|
38
|
Nagai T, Yoshimoto J, Kannon T, Kuroda K, Kaibuchi K. Phosphorylation Signals in Striatal Medium Spiny Neurons. Trends Pharmacol Sci 2016; 37:858-871. [DOI: 10.1016/j.tips.2016.07.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 07/18/2016] [Accepted: 07/21/2016] [Indexed: 12/21/2022]
|
39
|
Li Y, Ping X, Yu P, Liang J, Shen F, Han J, Cui C. Over-expression of the GluN2B subunit in the forebrain facilitates the acquisition of morphine-related positive and aversive memory in rats. Behav Brain Res 2016; 311:416-424. [DOI: 10.1016/j.bbr.2016.05.039] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2016] [Revised: 05/14/2016] [Accepted: 05/19/2016] [Indexed: 12/18/2022]
|
40
|
Reappraising striatal D1- and D2-neurons in reward and aversion. Neurosci Biobehav Rev 2016; 68:370-386. [PMID: 27235078 DOI: 10.1016/j.neubiorev.2016.05.021] [Citation(s) in RCA: 115] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 05/16/2016] [Accepted: 05/22/2016] [Indexed: 12/31/2022]
Abstract
The striatum has been involved in complex behaviors such as motor control, learning, decision-making, reward and aversion. The striatum is mainly composed of medium spiny neurons (MSNs), typically divided into those expressing dopamine receptor D1, forming the so-called direct pathway, and those expressing D2 receptor (indirect pathway). For decades it has been proposed that these two populations exhibit opposing control over motor output, and recently, the same dichotomy has been proposed for valenced behaviors. Whereas D1-MSNs mediate reinforcement and reward, D2-MSNs have been associated with punishment and aversion. In this review we will discuss pharmacological, genetic and optogenetic studies that indicate that there is still controversy to what concerns the role of striatal D1- and D2-MSNs in this type of behaviors, highlighting the need to reconsider the early view that they mediate solely opposing aspects of valenced behaviour.
Collapse
|
41
|
Di Maio V, Ventriglia F, Santillo S. A model of dopamine regulation of glutamatergic synapse on medium size spiny neurons. Biosystems 2016; 142-143:25-31. [PMID: 26957078 DOI: 10.1016/j.biosystems.2016.03.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Revised: 02/10/2016] [Accepted: 03/01/2016] [Indexed: 11/24/2022]
Abstract
Spiny neurons of striatum receive glutamatergic synapses on dendritic spines on the neck of which project dopaminergic synapses. Dopamine modulates, by D1 type receptors, the glutamatergic synapses by inducing the phosphorylation of AMPA and NMDA receptors which produces an increased amplitude response. Herein we present a model where, in addition to phosphorylation, the direct modulation by dopamine of the spine resistance can cooperate in producing the observed effect on some of these synapses.
Collapse
Affiliation(s)
- Vito Di Maio
- Istituto di Scienze Applicate e Sistemi Intelligenti del CNR, Italy.
| | | | - Silvia Santillo
- Istituto di Scienze Applicate e Sistemi Intelligenti del CNR, Italy.
| |
Collapse
|
42
|
Nagai T, Nakamuta S, Kuroda K, Nakauchi S, Nishioka T, Takano T, Zhang X, Tsuboi D, Funahashi Y, Nakano T, Yoshimoto J, Kobayashi K, Uchigashima M, Watanabe M, Miura M, Nishi A, Kobayashi K, Yamada K, Amano M, Kaibuchi K. Phosphoproteomics of the Dopamine Pathway Enables Discovery of Rap1 Activation as a Reward Signal In Vivo. Neuron 2016; 89:550-65. [DOI: 10.1016/j.neuron.2015.12.019] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 10/17/2015] [Accepted: 12/10/2015] [Indexed: 12/21/2022]
|
43
|
Singh AM, Neva JL, Staines WR. Aerobic exercise enhances neural correlates of motor skill learning. Behav Brain Res 2015; 301:19-26. [PMID: 26706889 DOI: 10.1016/j.bbr.2015.12.020] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2015] [Revised: 12/10/2015] [Accepted: 12/13/2015] [Indexed: 10/22/2022]
Abstract
INTRODUCTION Repetitive, in-phase bimanual motor training tasks can expand the excitable cortical area of the trained muscles. Recent evidence suggests that an acute bout of moderate-intensity aerobic exercise can enhance the induction of rapid motor plasticity at the motor hotspot. However, these changes have not been investigated throughout the entire cortical representation. Furthermore, it is unclear how exercise-induced changes in excitability may relate to motor performance. We investigated whether aerobic exercise could enhance the neural correlates of motor learning. We hypothesized that the combination of exercise and training would increase the excitable cortical area to a greater extent than either exercise or training alone, and that the addition of exercise would enhance performance on a motor training task. METHODS 25 young, healthy, right-handed individuals were recruited and divided into two groups and three experimental conditions. The exercise group performed exercise alone (EX) and exercise followed by training (EXTR) while the training group performed training alone (TR). RESULTS The combination of exercise and training increased excitability within the cortical map of the trained muscle to a greater extent than training alone. However, there was no difference in performance between the two groups. These results indicate that exercise may enhance the cortical adaptations to motor skill learning.
Collapse
Affiliation(s)
- Amaya M Singh
- Department of Kinesiology, University of Waterloo, 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada.
| | - Jason L Neva
- Department of Kinesiology, University of Waterloo, 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada
| | - W Richard Staines
- Department of Kinesiology, University of Waterloo, 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada
| |
Collapse
|
44
|
Kumar A. NMDA Receptor Function During Senescence: Implication on Cognitive Performance. Front Neurosci 2015; 9:473. [PMID: 26732087 PMCID: PMC4679982 DOI: 10.3389/fnins.2015.00473] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2015] [Accepted: 11/25/2015] [Indexed: 12/13/2022] Open
Abstract
N-methyl-D-aspartate (NMDA) receptors, a family of L-glutamate receptors, play an important role in learning and memory, and are critical for spatial memory. These receptors are tetrameric ion channels composed of a family of related subunits. One of the hallmarks of the aging human population is a decline in cognitive function; studies in the past couple of years have demonstrated deterioration in NMDA receptor subunit expression and function with advancing age. However, a direct relationship between impaired memory function and a decline in NMDA receptors is still ambiguous. Recent studies indicate a link between an age-associated NMDA receptor hypofunction and memory impairment and provide evidence that age-associated enhanced oxidative stress might be contributing to the alterations associated with senescence. However, clear evidence is still deficient in demonstrating the underlying mechanisms and a relationship between age-associated impaired cognitive faculties and NMDA receptor hypofunction. The current review intends to present an overview of the research findings regarding changes in expression of various NMDA receptor subunits and deficits in NMDA receptor function during senescence and its implication in age-associated impaired hippocampal-dependent memory function.
Collapse
Affiliation(s)
- Ashok Kumar
- Department of Neuroscience, Evelyn F. and William L. McKnight Brain Institute, University of Florida Gainesville, FL, USA
| |
Collapse
|
45
|
Köles L, Kató E, Hanuska A, Zádori ZS, Al-Khrasani M, Zelles T, Rubini P, Illes P. Modulation of excitatory neurotransmission by neuronal/glial signalling molecules: interplay between purinergic and glutamatergic systems. Purinergic Signal 2015; 12:1-24. [PMID: 26542977 DOI: 10.1007/s11302-015-9480-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 10/26/2015] [Indexed: 12/29/2022] Open
Abstract
Glutamate is the main excitatory neurotransmitter of the central nervous system (CNS), released both from neurons and glial cells. Acting via ionotropic (NMDA, AMPA, kainate) and metabotropic glutamate receptors, it is critically involved in essential regulatory functions. Disturbances of glutamatergic neurotransmission can be detected in cognitive and neurodegenerative disorders. This paper summarizes the present knowledge on the modulation of glutamate-mediated responses in the CNS. Emphasis will be put on NMDA receptor channels, which are essential executive and integrative elements of the glutamatergic system. This receptor is crucial for proper functioning of neuronal circuits; its hypofunction or overactivation can result in neuronal disturbances and neurotoxicity. Somewhat surprisingly, NMDA receptors are not widely targeted by pharmacotherapy in clinics; their robust activation or inhibition seems to be desirable only in exceptional cases. However, their fine-tuning might provide a promising manipulation to optimize the activity of the glutamatergic system and to restore proper CNS function. This orchestration utilizes several neuromodulators. Besides the classical ones such as dopamine, novel candidates emerged in the last two decades. The purinergic system is a promising possibility to optimize the activity of the glutamatergic system. It exerts not only direct and indirect influences on NMDA receptors but, by modulating glutamatergic transmission, also plays an important role in glia-neuron communication. These purinergic functions will be illustrated mostly by depicting the modulatory role of the purinergic system on glutamatergic transmission in the prefrontal cortex, a CNS area important for attention, memory and learning.
Collapse
Affiliation(s)
- László Köles
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvárad tér 4, Budapest, 1089, Hungary.
| | - Erzsébet Kató
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvárad tér 4, Budapest, 1089, Hungary
| | - Adrienn Hanuska
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvárad tér 4, Budapest, 1089, Hungary
| | - Zoltán S Zádori
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvárad tér 4, Budapest, 1089, Hungary
| | - Mahmoud Al-Khrasani
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvárad tér 4, Budapest, 1089, Hungary
| | - Tibor Zelles
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvárad tér 4, Budapest, 1089, Hungary
| | - Patrizia Rubini
- Rudolf-Boehm-Institute of Pharmacology and Toxicology, University of Leipzig, 04107, Leipzig, Germany
| | - Peter Illes
- Rudolf-Boehm-Institute of Pharmacology and Toxicology, University of Leipzig, 04107, Leipzig, Germany.
| |
Collapse
|
46
|
Mao LM, Wang JQ. Dopaminergic and cholinergic regulation of Fyn tyrosine kinase phosphorylation in the rat striatum in vivo. Neuropharmacology 2015; 99:491-9. [PMID: 26277342 DOI: 10.1016/j.neuropharm.2015.08.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 07/27/2015] [Accepted: 08/11/2015] [Indexed: 12/21/2022]
Abstract
Src and Fyn are two Src family kinase (SFK) members that are expressed in mammalian brains and play important roles in the regulation of a variety of neuronal and synaptic substrates. Here we investigated the responsiveness of these SFKs to changing dopamine receptor signals in dopamine responsive regions of adult rat brains in vivo. Pharmacological activation of dopamine D1 receptors (D1Rs) by a systemic injection of the selective agonist SKF81297 increased phosphorylation of SFKs at a conserved and activation-associated autophosphorylation site (Y416) in the striatum, indicating activation of SFKs following SKF81297 injection. The dopamine D2 receptor (D2R) agonist quinpirole had no effect. Blockade of D1Rs with an antagonist SCH23390 did not alter striatal Y416 phosphorylation, while the D2R antagonist eticlopride elevated it. Between Src and Fyn, SKF81297 seemed to preferentially facilitate Fyn phosphorylation. Activation of muscarinic acetylcholine M4 receptors (M4Rs) with a positive allosteric modulator VU0152100 suppressed SFK Y416 responses to SKF81297. Additionally, SKF81297 induced a correlated increase in phosphorylation of N-methyl-D-aspartate (NMDA) receptor GluN2B subunits at a Fyn site (Y1472), which was attenuated by VU0152100. SKF81297 also enhanced synaptic recruitments of active Fyn and GluN1/GluN2B-containing NMDA receptors. These data demonstrate that D1Rs regulate Fyn and downstream NMDA receptors in striatal neurons in vivo. Acetylcholine through activating M4Rs inhibits Fyn and NMDA receptors in their sensitivity to D1R signaling.
Collapse
Affiliation(s)
- Li-Min Mao
- Department of Basic Medical Science, School of Medicine, University of Missouri-Kansas City, Kansas City, MO 64108, USA
| | - John Q Wang
- Department of Basic Medical Science, School of Medicine, University of Missouri-Kansas City, Kansas City, MO 64108, USA; Department of Anesthesiology, School of Medicine, University of Missouri-Kansas City, Kansas City, MO 64108, USA; Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China.
| |
Collapse
|
47
|
Jin DZ, Xue B, Mao LM, Wang JQ. Metabotropic glutamate receptor 5 upregulates surface NMDA receptor expression in striatal neurons via CaMKII. Brain Res 2015; 1624:414-423. [PMID: 26256252 DOI: 10.1016/j.brainres.2015.07.053] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Revised: 07/23/2015] [Accepted: 07/30/2015] [Indexed: 12/29/2022]
Abstract
Metabotropic and ionotropic glutamate receptors are closely clustered in postsynaptic membranes and are believed to interact actively with each other to control excitatory synaptic transmission. Metabotropic glutamate receptor 5 (mGluR5), for example, has been well documented to potentiate ionotropic NMDA receptor activity, although underlying mechanisms are poorly understood. In this study, we investigated the role of mGluR5 in regulating trafficking and subcellular distribution of NMDA receptors in adult rat striatal neurons. We found that the mGluR1/5 agonist DHPG concentration-dependently increased NMDA receptor GluN1 and GluN2B subunit expression in the surface membrane. Meanwhile, DHPG reduced GluN1 and GluN2B levels in the intracellular compartment. The effect of DHPG was blocked by an mGluR5 selective antagonist MTEP but not by an mGluR1 selective antagonist 3-MATIDA. Pretreatment with an inhibitor or a specific inhibitory peptide for synapse-enriched Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) also blocked the DHPG-stimulated redistribution of GluN1 and GluN2B. In addition, DHPG enhanced CaMKIIα activity and elevated GluN2B phosphorylation at a CaMKII-sensitive site (serine 1303). These results demonstrate that mGluR5 regulates trafficking of NMDA receptors in striatal neurons. Activation of mGluR5 appears to induce rapid trafficking of GluN1 and GluN2B to surface membranes through a signaling pathway involving CaMKII.
Collapse
Affiliation(s)
- Dao-Zhong Jin
- Department of Basic Medical Science, School of Medicine, University of Missouri-Kansas City, Kansas City, MO 64108, USA
| | - Bing Xue
- Department of Basic Medical Science, School of Medicine, University of Missouri-Kansas City, Kansas City, MO 64108, USA
| | - Li-Min Mao
- Department of Basic Medical Science, School of Medicine, University of Missouri-Kansas City, Kansas City, MO 64108, USA
| | - John Q Wang
- Department of Basic Medical Science, School of Medicine, University of Missouri-Kansas City, Kansas City, MO 64108, USA; Department of Anesthesiology, School of Medicine, University of Missouri-Kansas City, Kansas City, MO 64108, USA.
| |
Collapse
|
48
|
Cone JJ, Roitman JD, Roitman MF. Ghrelin regulates phasic dopamine and nucleus accumbens signaling evoked by food-predictive stimuli. J Neurochem 2015; 133:844-56. [PMID: 25708523 PMCID: PMC4464979 DOI: 10.1111/jnc.13080] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Revised: 02/16/2015] [Accepted: 02/17/2015] [Indexed: 12/24/2022]
Abstract
Environmental stimuli that signal food availability hold powerful sway over motivated behavior and promote feeding, in part, by activating the mesolimbic system. These food-predictive cues evoke brief (phasic) changes in nucleus accumbens (NAc) dopamine concentration and in the activity of individual NAc neurons. Phasic fluctuations in mesolimbic signaling have been directly linked to goal-directed behaviors, including behaviors elicited by food-predictive cues. Food-seeking behavior is also strongly influenced by physiological state (i.e., hunger vs. satiety). Ghrelin, a stomach hormone that crosses the blood-brain barrier, is linked to the perception of hunger and drives food intake, including intake potentiated by environmental cues. Notwithstanding, whether ghrelin regulates phasic mesolimbic signaling evoked by food-predictive stimuli is unknown. Here, rats underwent Pavlovian conditioning in which one cue predicted the delivery of rewarding food (CS+) and a second cue predicted nothing (CS-). After training, we measured the effect of ghrelin infused into the lateral ventricle (LV) on sub-second fluctuations in NAc dopamine using fast-scan cyclic voltammetry and individual NAc neuron activity using in vivo electrophysiology in separate groups of rats. LV ghrelin augmented both phasic dopamine and phasic increases in the activity of NAc neurons evoked by the CS+. Importantly, ghrelin did not affect the dopamine nor NAc neuron response to the CS-, suggesting that ghrelin selectively modulated mesolimbic signaling evoked by motivationally significant stimuli. These data demonstrate that ghrelin, a hunger signal linked to physiological state, can regulate cue-evoked mesolimbic signals that underlie food-directed behaviors. Cues that predict food availability powerfully regulate food-seeking behavior. Here we show that cue-evoked changes in both nucleus accumbens (NAc) dopamine (DA) and NAc cell activity are modulated by intra-cranial infusions of the stomach hormone ghrelin--a hormone known to act centrally to promote food intake. These data demonstrate that hormones associated with physiological state (i.e., hunger) can affect encoding of food-predictive cues in brain regions that drive food-motivated behavior.
Collapse
Affiliation(s)
- Jackson J. Cone
- Graduate Program in Neuroscience, University of Illinois at Chicago, 60612
| | - Jamie D. Roitman
- Department of Psychology, University of Illinois at Chicago, 60607
| | - Mitchell F. Roitman
- Graduate Program in Neuroscience, University of Illinois at Chicago, 60612
- Department of Psychology, University of Illinois at Chicago, 60607
| |
Collapse
|
49
|
Gardoni F, Bellone C. Modulation of the glutamatergic transmission by Dopamine: a focus on Parkinson, Huntington and Addiction diseases. Front Cell Neurosci 2015; 9:25. [PMID: 25784855 PMCID: PMC4345909 DOI: 10.3389/fncel.2015.00025] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 01/14/2015] [Indexed: 01/11/2023] Open
Abstract
Dopamine (DA) plays a major role in motor and cognitive functions as well as in reward processing by regulating glutamatergic inputs. In particular in the striatum the release of DA rapidly influences synaptic transmission modulating both AMPA and NMDA receptors. Several neurodegenerative and neuropsychiatric disorders, including Parkinson, Huntington and addiction-related diseases, manifest a dysregulation of glutamate and DA signaling. Here, we will focus our attention on the mechanisms underlying the modulation of the glutamatergic transmission by DA in striatal circuits.
Collapse
Affiliation(s)
- Fabrizio Gardoni
- Department of Pharmacological and Biomolecular Sciences, University of Milano Milano, Italy
| | - Camilla Bellone
- Department of Fundamental Neuroscience, University of Lausanne Lausanne, Switzerland
| |
Collapse
|
50
|
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: 110] [Impact Index Per Article: 12.2] [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
|