1
|
Iida I, Konno K, Natsume R, Abe M, Watanabe M, Sakimura K, Terunuma M. Behavioral analysis of kainate receptor KO mice and the role of GluK3 subunit in anxiety. Sci Rep 2024; 14:4521. [PMID: 38402313 PMCID: PMC10894277 DOI: 10.1038/s41598-024-55063-z] [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: 12/24/2023] [Accepted: 02/20/2024] [Indexed: 02/26/2024] Open
Abstract
Kainate receptors (KARs) are one of the ionotropic glutamate receptors in the central nervous system (CNS) comprised of five subunits, GluK1-GluK5. There is a growing interest in the association between KARs and psychiatric disorders, and there have been several studies investigating the behavioral phenotypes of KAR deficient mice, however, the difference in the genetic background has been found to affect phenotype in multiple mouse models of human diseases. Here, we examined GluK1-5 single KO mice in a pure C57BL/6N background and identified that GluK3 KO mice specifically express anxiolytic-like behavior with an alteration in dopamine D2 receptor (D2R)-induced anxiety, and reduced D2R expression in the striatum. Biochemical studies in the mouse cortex confirmed that GluK3 subunits do not assemble with GluK4 and GluK5 subunits, that can be activated by lower concentration of agonists. Overall, we found that GluK3-containing KARs function to express anxiety, which may represent promising anti-anxiety medication targets.
Collapse
Affiliation(s)
- Izumi Iida
- Division of Oral Biochemistry, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata University, Niigata, 951-8514, Japan
- Research Center for Advanced Oral Science, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata University, Niigata, 951-8514, Japan
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Meguro, Tokyo, 153-8902, Japan
| | - Kohtarou Konno
- Department of Anatomy, Faculty of Medicine, Hokkaido University, Sapporo, 060-8638, Japan
| | - Rie Natsume
- Department of Animal Model Development, Brain Research Institute, Niigata University, Niigata, 951-8585, Japan
| | - Manabu Abe
- Department of Animal Model Development, Brain Research Institute, Niigata University, Niigata, 951-8585, Japan
| | - Masahiko Watanabe
- Department of Anatomy, Faculty of Medicine, Hokkaido University, Sapporo, 060-8638, Japan
| | - Kenji Sakimura
- Department of Animal Model Development, Brain Research Institute, Niigata University, Niigata, 951-8585, Japan.
| | - Miho Terunuma
- Division of Oral Biochemistry, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata University, Niigata, 951-8514, Japan.
| |
Collapse
|
2
|
Yucel BP, Al Momany EM, Evans AJ, Seager R, Wilkinson KA, Henley JM. Coordinated interplay between palmitoylation, phosphorylation and SUMOylation regulates kainate receptor surface expression. Front Mol Neurosci 2023; 16:1270849. [PMID: 37868810 PMCID: PMC10585046 DOI: 10.3389/fnmol.2023.1270849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 09/11/2023] [Indexed: 10/24/2023] Open
Abstract
Kainate receptors (KARs) are key regulators of neuronal excitability and synaptic transmission. KAR surface expression is tightly controlled in part by post-translational modifications (PTMs) of the GluK2 subunit. We have shown previously that agonist activation of GluK2-containing KARs leads to phosphorylation of GluK2 at S868, which promotes subsequent SUMOylation at K886 and receptor endocytosis. Furthermore, GluK2 has been shown to be palmitoylated. However, how the interplay between palmitoylation, phosphorylation and SUMOylation orchestrate KAR trafficking remains unclear. Here, we used a library of site-specific GluK2 mutants to investigate the interrelationship between GluK2 PTMs, and their impact on KAR surface expression. We show that GluK2 is basally palmitoylated and that this is decreased by kainate (KA) stimulation. Moreover, a non-palmitoylatable GluK2 mutant (C858/C871A) shows enhanced S868 phosphorylation and K886 SUMOylation under basal conditions and is insensitive to KA-induced internalisation. These results indicate that GluK2 palmitoylation contributes to stabilising KAR surface expression and that dynamic depalmitoylation promotes downstream phosphorylation and SUMOylation to mediate activity-dependent KAR endocytosis.
Collapse
Affiliation(s)
| | | | | | | | - Kevin A. Wilkinson
- Centre for Synaptic Plasticity, School of Biochemistry, Biomedical Sciences Building, University of Bristol, Bristol, United Kingdom
| | - Jeremy M. Henley
- Centre for Synaptic Plasticity, School of Biochemistry, Biomedical Sciences Building, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
3
|
Identification of a Novel Idiopathic Epilepsy Risk Locus and a Variant in the CCDC85A Gene in the Dutch Partridge Dog. Animals (Basel) 2023; 13:ani13050810. [PMID: 36899667 PMCID: PMC10000155 DOI: 10.3390/ani13050810] [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: 01/18/2023] [Revised: 02/15/2023] [Accepted: 02/20/2023] [Indexed: 03/12/2023] Open
Abstract
(1) Idiopathic epilepsy (IE) is thought to have a genetic cause in several dog breeds. However, only two causal variants have been identified to date, and few risk loci are known. No genetic studies have been conducted on IE in the Dutch partridge dog (DPD), and little has been reported on the epileptic phenotype in this breed. (2) Owner-filled questionnaires and diagnostic investigations were used to characterize IE in the DPD. A genome-wide association study (GWAS) involving 16 cases and 43 controls was performed, followed by sequencing of the coding sequence and splice site regions of a candidate gene within the associated region. Subsequent whole-exome sequencing (WES) of one family (including one IE-affected dog, both parents, and an IE-free sibling) was performed. (3) IE in the DPD has a broad range in terms of age at onset, frequency, and duration of epileptic seizures. Most dogs showed focal epileptic seizures evolving into generalized seizures. A new risk locus on chromosome 12 (BICF2G630119560; praw = 4.4 × 10-7; padj = 0.043) was identified through GWAS. Sequencing of the GRIK2 candidate gene revealed no variants of interest. No WES variants were located within the associated GWAS region. However, a variant in CCDC85A (chromosome 10; XM_038680630.1: c.689C > T) was discovered, and dogs homozygous for the variant (T/T) had an increased risk of developing IE (OR: 6.0; 95% CI: 1.6-22.6). This variant was identified as likely pathogenic according to ACMG guidelines. (4) Further research is necessary before the risk locus or CCDC85A variant can be used for breeding decisions.
Collapse
|
4
|
Chałupnik P, Szymańska E. Kainate Receptor Antagonists: Recent Advances and Therapeutic Perspective. Int J Mol Sci 2023; 24:1908. [PMID: 36768227 PMCID: PMC9916396 DOI: 10.3390/ijms24031908] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/06/2023] [Accepted: 01/16/2023] [Indexed: 01/21/2023] Open
Abstract
Since the 1990s, ionotropic glutamate receptors have served as an outstanding target for drug discovery research aimed at the discovery of new neurotherapeutic agents. With the recent approval of perampanel, the first marketed non-competitive antagonist of AMPA receptors, particular interest has been directed toward 'non-NMDA' (AMPA and kainate) receptor inhibitors. Although the role of AMPA receptors in the development of neurological or psychiatric disorders has been well recognized and characterized, progress in understanding the function of kainate receptors (KARs) has been hampered, mainly due to the lack of specific and selective pharmacological tools. The latest findings in the biology of KA receptors indicate that they are involved in neurophysiological activity and play an important role in both health and disease, including conditions such as anxiety, schizophrenia, epilepsy, neuropathic pain, and migraine. Therefore, we reviewed recent advances in the field of competitive and non-competitive kainate receptor antagonists and their potential therapeutic applications. Due to the high level of structural divergence among the compounds described here, we decided to divide them into seven groups according to their overall structure, presenting a total of 72 active compounds.
Collapse
Affiliation(s)
| | - Ewa Szymańska
- Department of Technology and Biotechnology of Drugs, Jagiellonian University Medical College in Kraków, PL 30-688 Kraków, Poland
| |
Collapse
|
5
|
Glutamate Signaling and Filopodiagenesis of Astrocytoma Cells in Brain Cancers: Survey and Questions. Cells 2022; 11:cells11172657. [PMID: 36078065 PMCID: PMC9454653 DOI: 10.3390/cells11172657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 08/19/2022] [Accepted: 08/24/2022] [Indexed: 11/22/2022] Open
Abstract
Astrocytes are non-excitable cells in the CNS that can cause life-threatening astrocytoma tumors when they transform to cancerous cells. Perturbed homeostasis of the neurotransmitter glutamate is associated with astrocytoma tumor onset and progression, but the factors that govern this phenomenon are less known. Herein, we review possible mechanisms by which glutamate may act in facilitating the growth of projections in astrocytic cells. This review discusses the similarities and differences between the morphology of astrocytes and astrocytoma cells, and the role that dysregulation in glutamate and calcium signaling plays in the aberrant morphology of astrocytoma cells. Converging reports suggest that ionotropic glutamate receptors and voltage-gated calcium channels expressed in astrocytes may be responsible for the abnormal filopodiagenesis or process extension leading to astrocytoma cells’ infiltration throughout the brain.
Collapse
|
6
|
Abstract
Neural communication and modulation are complex processes. Ionotropic glutamate receptors (iGluRs) significantly contribute to mediating the fast-excitatory branch of neurotransmission in the mammalian brain. Kainate receptors (KARs), a subfamily of the iGluRs, act as modulators of the neuronal circuitry by playing important roles at both the post- and presynaptic sites of specific neurons. The functional tetrameric receptors are formed by two different gene families, low agonist affinity (GluK1-GluK3) and high agonist affinity (GluK4-GluK5) subunits. These receptors garnered attention in the past three decades, and since then, much work has been done to understand their localization, interactome, physiological functions, and regulation. Cloning of the receptor subunits (GluK1-GluK5) in the early 1990s led to recombinant expression of kainate receptors in heterologous systems. This facilitated understanding of the functional differences between subunit combinations, splice variants, trafficking, and drug discovery. Structural studies of individual domains and recent full-length homomeric and heteromeric kainate receptors have revealed unique functional mechanisms, which have answered several long-standing questions in the field of kainate receptor biology. In this chapter, we review the current understanding of kainate receptors and associated disorders.
Collapse
Affiliation(s)
- Surbhi Dhingra
- Laboratory of Membrane Protein Biology, National Centre for Cell Science, NCCS Complex, S. P. Pune University, Pune, Maharashtra, India
| | - Juhi Yadav
- Laboratory of Membrane Protein Biology, National Centre for Cell Science, NCCS Complex, S. P. Pune University, Pune, Maharashtra, India
| | - Janesh Kumar
- Laboratory of Membrane Protein Biology, National Centre for Cell Science, NCCS Complex, S. P. Pune University, Pune, Maharashtra, India.
| |
Collapse
|
7
|
Long-term effect of neonatal antagonism of ionotropic glutamate receptors on dendritic spines and cognitive function in rats. J Chem Neuroanat 2021; 119:102054. [PMID: 34839003 DOI: 10.1016/j.jchemneu.2021.102054] [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: 09/05/2021] [Revised: 11/04/2021] [Accepted: 11/23/2021] [Indexed: 12/11/2022]
Abstract
Glutamate is the most abundant excitatory neurotransmitter in the hippocampus where mediates its actions by activating glutamate receptors. The activation of these receptors is essential for the maintenance and dynamics of dendritic spines and plasticity that correlate with learning and memory processes during neurodevelopment and adulthood. We studied in adults the effect of blocking ionotropic glutamate receptors (NMDAR, AMPAR, and KAR) functions at neonatal age (PD1-PD15) with their respective antagonists D-AP5, GYKI-53655 and UBP-302. We first evaluated memory using a new object recognition test in adults. Second, we evaluated the levels of glial fibrillary acidic protein, synaptophysin and actin with immunohistochemistry in the CA1, CA3, and dentate gyrus regions of the hippocampus and, finally, the number of dendritic spines and their dynamics using Golgi-Cox staining. We found that ionotropic glutamate receptor function blockade at neonatal age causes a reduction in short and long-term memory in adulthood and a reduction in the expression of synaptophysin and actin protein levels in the hippocampus regions studied. This blockade also reduced the number of dendritic spines and modified dendritic dynamics in the CA1 region. The antagonism of the three types of ionotropic glutamate receptors reduced the mushrooms and bifurcated types of spines and increased the thin spines. The number of stubby spines was reduced by D-AP5, increased by UPB-302, and not affected by GYKI-53655. Our results indicate that the blockade of neonatal ionotropic glutamate receptors produces alterations that persist until adulthood.
Collapse
|
8
|
Henley JM, Nair JD, Seager R, Yucel BP, Woodhall G, Henley BS, Talandyte K, Needs HI, Wilkinson KA. Kainate and AMPA receptors in epilepsy: Cell biology, signalling pathways and possible crosstalk. Neuropharmacology 2021; 195:108569. [PMID: 33915142 DOI: 10.1016/j.neuropharm.2021.108569] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 03/13/2021] [Accepted: 04/09/2021] [Indexed: 02/06/2023]
Abstract
Epilepsy is caused when rhythmic neuronal network activity escapes normal control mechanisms, resulting in seizures. There is an extensive and growing body of evidence that the onset and maintenance of epilepsy involves alterations in the trafficking, synaptic surface expression and signalling of kainate and AMPA receptors (KARs and AMPARs). The KAR subunit GluK2 and AMPAR subunit GluA2 are key determinants of the properties of their respective assembled receptors. Both subunits are subject to extensive protein interactions, RNA editing and post-translational modifications. In this review we focus on the cell biology of GluK2-containing KARs and GluA2-containing AMPARs and outline how their regulation and dysregulation is implicated in, and affected by, seizure activity. Further, we discuss role of KARs in regulating AMPAR surface expression and plasticity, and the relevance of this to epilepsy. This article is part of the special issue on 'Glutamate Receptors - Kainate receptors'.
Collapse
Affiliation(s)
- Jeremy M Henley
- School of Biochemistry, Centre for Synaptic Plasticity, Biomedical Sciences Building, University of Bristol, University Walk, Bristol, BS8 1TD, UK; Centre for Neuroscience and Regenerative Medicine, Faculty of Science, University of Technology Sydney, Ultimo, NSW, Australia.
| | - Jithin D Nair
- School of Biochemistry, Centre for Synaptic Plasticity, Biomedical Sciences Building, University of Bristol, University Walk, Bristol, BS8 1TD, UK
| | - Richard Seager
- School of Biochemistry, Centre for Synaptic Plasticity, Biomedical Sciences Building, University of Bristol, University Walk, Bristol, BS8 1TD, UK
| | - Busra P Yucel
- School of Biochemistry, Centre for Synaptic Plasticity, Biomedical Sciences Building, University of Bristol, University Walk, Bristol, BS8 1TD, UK
| | - Gavin Woodhall
- School of Life and Health Sciences, Aston University, Aston Triangle, Birmingham, B4 7ET, UK
| | - Benjamin S Henley
- Faculty of Medical Sciences, The Medical School, Newcastle University, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK
| | - Karolina Talandyte
- School of Biochemistry, Centre for Synaptic Plasticity, Biomedical Sciences Building, University of Bristol, University Walk, Bristol, BS8 1TD, UK
| | - Hope I Needs
- School of Biochemistry, Centre for Synaptic Plasticity, Biomedical Sciences Building, University of Bristol, University Walk, Bristol, BS8 1TD, UK
| | - Kevin A Wilkinson
- School of Biochemistry, Centre for Synaptic Plasticity, Biomedical Sciences Building, University of Bristol, University Walk, Bristol, BS8 1TD, UK.
| |
Collapse
|
9
|
Iida I, Konno K, Natsume R, Abe M, Watanabe M, Sakimura K, Terunuma M. A comparative analysis of kainate receptor GluK2 and GluK5 knockout mice in a pure genetic background. Behav Brain Res 2021; 405:113194. [PMID: 33631192 DOI: 10.1016/j.bbr.2021.113194] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 01/21/2021] [Accepted: 02/15/2021] [Indexed: 11/21/2022]
Abstract
Kainate receptors (KARs) are members of the glutamate receptor family that regulate synaptic function in the brain. Although they are known to be associated with psychiatric disorders, how they are involved in these disorders remains unclear. KARs are tetrameric channels assembled from a combination of GluK1-5 subunits. Among these, GluK2 and GluK5 subunits are the major heteromeric subunits in the brain. To determine the functional similarities and differences between GluK2 and GluK5 subunits, we generated GluK2 KO and GluK5 KO mice on a C57BL/6N background, a well-characterized inbred strain, and compared their behavioral phenotypes. We found that GluK2 KO and GluK5 KO mice exhibited the same phenotypes in many tests, such as reduced locomotor activity, impaired motor function, and enhanced depressive-like behavior. No change was observed in motor learning, anxiety-like behavior, or sociability. Additionally, we identified subunit-specific phenotypes, such as reduced motivation toward their environment in GluK2 KO mice and an enhancement in the contextual memory in GluK5 KO mice. These results revealed that GluK2 and GluK5 subunits not only function in a coordinated manner but also have a subunit-specific role in regulating behavior. To summarize, we demonstrated subunit-specific and common behavioral effects of GluK2 and GluK5 subunits for the first time. Moreover, to the best of our knowledge, this is the first evidence of the involvement of the GluK5 subunit in the expression of depressive-like behavior and contextual memory, which strongly indicates its role in psychiatric disorders.
Collapse
Affiliation(s)
- Izumi Iida
- Division of Oral Biochemistry, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan; Research Center for Advanced Oral Science, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan
| | - Kohtarou Konno
- Department of Anatomy, Faculty of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| | - Rie Natsume
- Department of Animal Model Development, Brain Research Institute, Niigata University, Niigata 951-8585, Japan
| | - Manabu Abe
- Department of Animal Model Development, Brain Research Institute, Niigata University, Niigata 951-8585, Japan
| | - Masahiko Watanabe
- Department of Anatomy, Faculty of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| | - Kenji Sakimura
- Department of Animal Model Development, Brain Research Institute, Niigata University, Niigata 951-8585, Japan.
| | - Miho Terunuma
- Division of Oral Biochemistry, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan.
| |
Collapse
|
10
|
Mprah R, Adzika GK, Gyasi YI, Ndzie Noah ML, Adu-Amankwaah J, Adekunle AO, Duah M, Wowui PI, Weili Q. Glutaminolysis: A Driver of Vascular and Cardiac Remodeling in Pulmonary Arterial Hypertension. Front Cardiovasc Med 2021; 8:667446. [PMID: 33996951 PMCID: PMC8113389 DOI: 10.3389/fcvm.2021.667446] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Accepted: 03/08/2021] [Indexed: 12/30/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a decimating ailment described by chronic precapillary pulmonary hypertension, an elevated mean pulmonary arterial pressure with a normal pulmonary capillary wedge pressure, and a raised pulmonary vascular resistance resulting in increased right ventricular afterload culminating in heart failure and death. Current PAH treatments regulate the vasodilatory/vasoconstrictory balance of pulmonary vessels. However, these treatment options are unable to stop the progression of, or reverse, an already established disease. Recent studies have advanced a metabolic dysregulation, featuring increased glutamine metabolism, as a mechanism driving PAH progression. Metabolic dysregulation in PAH leads to increased glutaminolysis to produce substrate to meet the high-energy requirement by hyperproliferative and apoptosis-resistant pulmonary vascular cells. This article explores the role of glutamate metabolism in PAH and how it could be targeted as an anti-remodeling therapeutic strategy.
Collapse
Affiliation(s)
- Richard Mprah
- Department of Physiology, Xuzhou Medical University, Xuzhou, China
| | | | - Yusif I. Gyasi
- Department of Chemistry & Biochemistry, Central Michigan University, Mount Pleasant, TX, United States
| | | | | | | | - Maxwell Duah
- Haematology Department, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | | | - Qiao Weili
- Department of Physiology, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
11
|
Kainate receptors in the developing neuronal networks. Neuropharmacology 2021; 195:108585. [PMID: 33910033 DOI: 10.1016/j.neuropharm.2021.108585] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 04/15/2021] [Accepted: 04/16/2021] [Indexed: 12/14/2022]
Abstract
Kainate receptors (KARs) are highly expressed in the immature brain and have unique developmentally regulated functions that may be important in linking neuronal activity to morphogenesis during activity-dependent fine-tuning of the synaptic connectivity. Altered expression of KARs in the developing neural network leads to changes in glutamatergic connectivity and network excitability, which may lead to long-lasting changes in behaviorally relevant circuitries in the brain. Here, we summarize the current knowledge on physiological and morphogenic functions described for different types of KARs at immature neural circuitries, focusing on their roles in modulating synaptic transmission and plasticity as well as circuit maturation in the rodent hippocampus and amygdala. Finally, we discuss the emerging evidence suggesting that malfunction of KARs in the immature brain may contribute to the pathophysiology underlying developmentally originating neurological disorders.
Collapse
|
12
|
Li H, Li J, Guan Y, Wang Y. The emerging role of kainate receptor functional dysregulation in pain. Mol Pain 2021; 17:1744806921990944. [PMID: 33567997 PMCID: PMC7883153 DOI: 10.1177/1744806921990944] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Pain is a serious clinical challenge, and is associated with a significant reduction in quality of life and high financial costs for affected patients. Research efforts have been made to explore the etiological basis of pain to guide the future treatment of patients suffering from pain conditions. Findings from studies using KA (kainate) receptor agonist, antagonists and receptor knockout mice suggested that KA receptor dysregulation and dysfunction may govern both peripheral and central sensitization in the context of pain. Additional evidence showed that KA receptor dysfunction may disrupt the finely-tuned process of glutamic acid transmission, thereby contributing to the onset of a range of pathological contexts. In the present review, we summarized major findings in recent studies which examined the roles of KA receptor dysregulation in nociceptive transmission and in pain. This timely overview of current knowledge will help to provide a framework for future developing novel therapeutic strategies to manage pain.
Collapse
Affiliation(s)
- Huili Li
- Department of Anesthesiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Junfa Li
- Department of Neurobiology, Capital Medical University, Beijing, China
| | - Yun Guan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yun Wang
- Department of Anesthesiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
13
|
Tubbs JD, Ding J, Baum L, Sham PC. Systemic neuro-dysregulation in depression: Evidence from genome-wide association. Eur Neuropsychopharmacol 2020; 39:1-18. [PMID: 32896454 DOI: 10.1016/j.euroneuro.2020.08.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 06/10/2020] [Accepted: 08/17/2020] [Indexed: 12/16/2022]
Abstract
Depression is the world's leading cause of disability. Greater understanding of the neurobiological basis of depression is necessary for developing novel treatments with improved efficacy and acceptance. Recently, major advances have been made in the search for genetic variants associated with depression which may help to elucidate etiological mechanisms. The present review has two major objectives. First, we offer a brief review of two major biological systems with strong evidence for involvement in depression pathology: neurotransmitter systems and the stress response. Secondly, we provide a synthesis of the functions of the 269 genes implicated by the most recent genome-wide meta-analysis, supporting the importance of these systems in depression and providing insights into other possible mechanisms involving neurodevelopment, neurogenesis, and neurodegeneration. Our goal is to undertake a broad, preliminary stock-taking of the most recent hypothesis-free findings and examine the weight of the evidence supporting these existing theories and highlighting novel directions. This qualitative review and accompanying gene function table provides a valuable resource and guide for basic and translational researchers, with suggestions for future mechanistic research, leveraging genetics to prioritize studies on the neurobiological processes involved in depression etiology and treatment.
Collapse
Affiliation(s)
- Justin D Tubbs
- Department of Psychiatry, The University of Hong Kong, Hong Kong
| | - Jiahong Ding
- Department of Psychiatry, The University of Hong Kong, Hong Kong
| | - Larry Baum
- Department of Psychiatry, The University of Hong Kong, Hong Kong; State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong
| | - Pak C Sham
- Department of Psychiatry, The University of Hong Kong, Hong Kong; State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong; Centre of PanorOmic Sciences, The University of Hong Kong, Hong Kong.
| |
Collapse
|
14
|
Kainate receptors have different modulatory effect in seizure-like events and slow rhythmic activity in entorhinal cortex ex vivo. Brain Res Bull 2019; 153:279-288. [PMID: 31550521 DOI: 10.1016/j.brainresbull.2019.09.009] [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: 03/15/2019] [Revised: 09/16/2019] [Accepted: 09/18/2019] [Indexed: 11/22/2022]
Abstract
In the neocortex, neurons form functional networks, the members of which exhibit a variable degree of synchronization. Slow rhythmic activity may be regarded as a balanced interplay of excitatory and inhibitory neuronal network activity, which is essential in learning and memory consolidation. On the other hand, seizures may be considered as hypersynchronized network states occurring in epileptic diseases. The brain slice method and multi-electrode array (MEA) systems offer a good opportunity for the modelling of cortical spontaneous activities by examining their initiation and propagation. Our main goals were to characterise and compare spontaneous activities developing in different conditions and cortical network states. The role of kainate receptors in these processes was also tested. According to our results, there are demonstrable dissimilarities between slow rhythmic activities vs. seizure-like events developing in the rat entorhinal cortex ex vivo in normal vs. epileptic conditions. Propagation velocity, time scale, activity pattern and pharmacological sensitivity are all different. Kainate receptors play a role in network activity in entorhinal cortex, they are capable to prolong the duration of the events of epileptiform activity. Their regulatory effect is more prominent under epileptic than under normal conditions.
Collapse
|
15
|
Chogii I, Das P, Njardarson JT. Efforts Toward a Unified Kainoid Family Synthesis Approach: Unexpected Sulfinamide‐Directed Conjugate Addition Results. ASIAN J ORG CHEM 2019. [DOI: 10.1002/ajoc.201800728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Isaac Chogii
- Department of Chemistry and BiochemistryUniversity of Arizona 1306 E. University Blvd. Tucson AZ 85721 USA
| | - Pradipta Das
- Department of Chemistry and BiochemistryUniversity of Arizona 1306 E. University Blvd. Tucson AZ 85721 USA
| | - Jon T. Njardarson
- Department of Chemistry and BiochemistryUniversity of Arizona 1306 E. University Blvd. Tucson AZ 85721 USA
| |
Collapse
|
16
|
Poulie CBM, Alcaide A, Krell-Jørgensen M, Larsen Y, Astier E, Bjørn-Yoshimoto WE, Yi F, Syrenne JT, Storgaard M, Nielsen B, Frydenvang KA, Jensen AA, Hansen KB, Pickering DS, Bunch L. Design and Synthesis of 2,3- trans-Proline Analogues as Ligands for Ionotropic Glutamate Receptors and Excitatory Amino Acid Transporters. ACS Chem Neurosci 2019; 10:2989-3007. [PMID: 31124660 DOI: 10.1021/acschemneuro.9b00205] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Development of pharmacological tools for the ionotropic glutamate receptors (iGluRs) is imperative for the study and understanding of the role and function of these receptors in the central nervous system. We report the synthesis of 18 analogues of (2 S,3 R)-2-carboxy-3-pyrrolidine acetic acid (3a), which explores the effect of introducing a substituent on the ε-carbon (3c-q). A new synthetic method was developed for the efficient synthesis of racemic 3a and applied to give expedited access to 13 racemic analogues of 3a. Pharmacological characterization was carried out at native iGluRs, cloned homomeric kainate receptors (GluK1-3), NMDA receptors (GluN1/GluN2A-D), and excitatory amino acid transporters (EAAT1-3). From the structure-activity relationship studies, several new ligands emerged, exemplified by triazole 3p-d1, GluK3-preferring (GluK1/GluK3 Ki ratio of 15), and the structurally closely related tetrazole 3q-s3-4 that displayed 4.4-100-fold preference as an antagonist for the GluN1/GluN2A receptor ( Ki = 0.61 μM) over GluN1/GluN2B-D ( Ki = 2.7-62 μM).
Collapse
Affiliation(s)
- Christian B. M. Poulie
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2100, Denmark
| | - Anna Alcaide
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2100, Denmark
| | - Mikkel Krell-Jørgensen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2100, Denmark
| | - Younes Larsen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2100, Denmark
| | - Eloi Astier
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2100, Denmark
| | - Walden E. Bjørn-Yoshimoto
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2100, Denmark
| | - Feng Yi
- Department of Biomedical and Pharmaceutical Sciences, Center for Structural and Functional Neuroscience, and Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, Montana 59812, United States
| | - Jed T. Syrenne
- Department of Biomedical and Pharmaceutical Sciences, Center for Structural and Functional Neuroscience, and Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, Montana 59812, United States
| | - Morten Storgaard
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2100, Denmark
| | - Birgitte Nielsen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2100, Denmark
| | - Karla A. Frydenvang
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2100, Denmark
| | - Anders A. Jensen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2100, Denmark
| | - Kasper B. Hansen
- Department of Biomedical and Pharmaceutical Sciences, Center for Structural and Functional Neuroscience, and Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, Montana 59812, United States
| | - Darryl S. Pickering
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2100, Denmark
| | - Lennart Bunch
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2100, Denmark
| |
Collapse
|
17
|
Fu H, Chen Z, Josephson L, Li Z, Liang SH. Positron Emission Tomography (PET) Ligand Development for Ionotropic Glutamate Receptors: Challenges and Opportunities for Radiotracer Targeting N-Methyl-d-aspartate (NMDA), α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid (AMPA), and Kainate Receptors. J Med Chem 2019; 62:403-419. [PMID: 30110164 PMCID: PMC6393217 DOI: 10.1021/acs.jmedchem.8b00714] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Ionotropic glutamate receptors (iGluRs) mediate excitatory neurotransmission within the mammalian central nervous system. iGluRs exist as three main groups: N-methyl-d-aspartate receptors (NMDARs), α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPARs), and kainate receptors. The past decades have witnessed a remarkable development of PET tracers targeting different iGluRs including NMDARs and AMPARs, and several of the tracers have advanced to clinical imaging studies. Here, we assess the recent development of iGluR PET probes, focusing on tracer design, brain kinetics, and performance in PET imaging studies. Furthermore, this review will not only present challenges in the tracer development but also provide novel approaches in conjunction with most recent drug discovery efforts on these iGluRs, including subtype-selective NMDAR and transmembrane AMPAR regulatory protein modulators and positive allosteric modulators (PAMs) of AMPARs. These approaches, if successful as PET tracers, may provide fundamental knowledge to understand the roles of iGluR receptors under physiological and pathological conditions.
Collapse
Affiliation(s)
- Hualong Fu
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, 55 Fruit St., Boston, MA 02114 USA
| | - Zhen Chen
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, 55 Fruit St., Boston, MA 02114 USA
| | - Lee Josephson
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, 55 Fruit St., Boston, MA 02114 USA
| | - Zijing Li
- State Key Laboratory of Molecular Vaccinology, Molecular Diagnosis & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, P. R. China
| | - Steven H. Liang
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, 55 Fruit St., Boston, MA 02114 USA
| |
Collapse
|
18
|
Scholefield CL, Atlason PT, Jane DE, Molnár E. Assembly and Trafficking of Homomeric and Heteromeric Kainate Receptors with Impaired Ligand Binding Sites. Neurochem Res 2018; 44:585-599. [PMID: 30302614 PMCID: PMC6420462 DOI: 10.1007/s11064-018-2654-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 09/27/2018] [Accepted: 10/01/2018] [Indexed: 10/28/2022]
Abstract
Kainate receptors (KARs) are a subfamily of ionotropic glutamate receptors (iGluRs) mediating excitatory synaptic transmission. Cell surface expressed KARs modulate the excitability of neuronal networks. The transfer of iGluRs from the endoplasmic reticulum (ER) to the cell surface requires occupation of the agonist binding sites. Here we used molecular modelling to produce a range of ligand binding domain (LBD) point mutants of GluK1-3 KAR subunits with and without altered agonist efficacy to further investigate the role of glutamate binding in surface trafficking and activation of homomeric and heteromeric KARs using endoglycosidase digestion, cell surface biotinylation and imaging of changes in intracellular Ca2+ concentration [Ca2+]i. Mutations of conserved amino acid residues in the LBD that disrupt agonist binding to GluK1-3 (GluK1-T675V, GluK2-A487L, GluK2-T659V and GluK3-T661V) reduced both the total expression levels and cell surface delivery of all of these mutant subunits compared to the corresponding wild type in transiently transfected human embryonic kidney 293 (HEK293) cells. In contrast, the exchange of non-conserved residues in the LBD that convert antagonist selectivity of GluK1-3 (GluK1-T503A, GluK2-A487T, GluK3-T489A, GluK1-N705S/S706N, GluK2-S689N/N690S, GluK3-N691S) did not alter the biosynthesis and trafficking of subunit proteins. Co-assembly of mutant GluK2 with an impaired LBD and wild type GluK5 subunits enables the cell surface expression of both subunits. However, [Ca2+]i imaging indicates that the occupancy of both GluK2 and GluK5 LBDs is required for the full activation of GluK2/GluK5 heteromeric KAR channels.
Collapse
Affiliation(s)
- Caroline L Scholefield
- Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Biomedical Sciences Building, University Walk, Bristol, BS8 1TD, UK
| | - Palmi T Atlason
- Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Biomedical Sciences Building, University Walk, Bristol, BS8 1TD, UK
| | - David E Jane
- Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Biomedical Sciences Building, University Walk, Bristol, BS8 1TD, UK
| | - Elek Molnár
- Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Biomedical Sciences Building, University Walk, Bristol, BS8 1TD, UK.
| |
Collapse
|
19
|
Brogi S, Brindisi M, Butini S, Kshirsagar GU, Maramai S, Chemi G, Gemma S, Campiani G, Novellino E, Fiorenzani P, Pinassi J, Aloisi AM, Gynther M, Venskutonytė R, Han L, Frydenvang K, Kastrup JS, Pickering DS. ( S)-2-Amino-3-(5-methyl-3-hydroxyisoxazol-4-yl)propanoic Acid (AMPA) and Kainate Receptor Ligands: Further Exploration of Bioisosteric Replacements and Structural and Biological Investigation. J Med Chem 2018; 61:2124-2130. [PMID: 29451794 DOI: 10.1021/acs.jmedchem.8b00099] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Starting from 1-4 and 7 structural templates, analogues based on bioisosteric replacements (5a-c vs 1, 2 and 6 vs 7) were synthesized for completing the SAR analysis. Interesting binding properties at GluA2, GluK1, and GluK3 receptors were discovered. The requirements for GluK3 interaction were elucidated by determining the X-ray structures of the GluK3-LBD with 2 and 5c and by computational studies. Antinociceptive potential was demonstrated for GluK1 partial agonist 3 and antagonist 7 (2 mg/kg ip).
Collapse
Affiliation(s)
- Simone Brogi
- Department of Biotechnology, Chemistry and Pharmacy, (DoE 2018-2022) NatSynDrugs , University of Siena , Via A. Moro 2 , 53100 Siena , Italy
| | - Margherita Brindisi
- Department of Biotechnology, Chemistry and Pharmacy, (DoE 2018-2022) NatSynDrugs , University of Siena , Via A. Moro 2 , 53100 Siena , Italy
| | - Stefania Butini
- Department of Biotechnology, Chemistry and Pharmacy, (DoE 2018-2022) NatSynDrugs , University of Siena , Via A. Moro 2 , 53100 Siena , Italy
| | - Giridhar U Kshirsagar
- Department of Biotechnology, Chemistry and Pharmacy, (DoE 2018-2022) NatSynDrugs , University of Siena , Via A. Moro 2 , 53100 Siena , Italy
| | - Samuele Maramai
- Department of Biotechnology, Chemistry and Pharmacy, (DoE 2018-2022) NatSynDrugs , University of Siena , Via A. Moro 2 , 53100 Siena , Italy
| | - Giulia Chemi
- Department of Biotechnology, Chemistry and Pharmacy, (DoE 2018-2022) NatSynDrugs , University of Siena , Via A. Moro 2 , 53100 Siena , Italy
| | - Sandra Gemma
- Department of Biotechnology, Chemistry and Pharmacy, (DoE 2018-2022) NatSynDrugs , University of Siena , Via A. Moro 2 , 53100 Siena , Italy
| | - Giuseppe Campiani
- Department of Biotechnology, Chemistry and Pharmacy, (DoE 2018-2022) NatSynDrugs , University of Siena , Via A. Moro 2 , 53100 Siena , Italy
| | - Ettore Novellino
- Department of Pharmacy , University of Napoli Federico II , Via D. Montesano 49 , 80131 Napoli , Italy
| | - Paolo Fiorenzani
- Department of Medicine, Surgery and Neuroscience , University of Siena , Viale M. Bracci 16 , 53100 Siena , Italy
| | - Jessica Pinassi
- Department of Medicine, Surgery and Neuroscience , University of Siena , Viale M. Bracci 16 , 53100 Siena , Italy
| | - Anna Maria Aloisi
- Department of Medicine, Surgery and Neuroscience , University of Siena , Viale M. Bracci 16 , 53100 Siena , Italy
| | - Mikko Gynther
- School of Pharmacy, Faculty of Health Sciences , University of Eastern Finland , 70211 Kuopio , Finland
| | - Raminta Venskutonytė
- Department of Drug Design and Pharmacology , University of Copenhagen , Jagtvej 162 , DK-2100 Copenhagen , Denmark
| | - Liwei Han
- Department of Drug Design and Pharmacology , University of Copenhagen , Jagtvej 162 , DK-2100 Copenhagen , Denmark
| | - Karla Frydenvang
- Department of Drug Design and Pharmacology , University of Copenhagen , Jagtvej 162 , DK-2100 Copenhagen , Denmark
| | - Jette Sandholm Kastrup
- Department of Drug Design and Pharmacology , University of Copenhagen , Jagtvej 162 , DK-2100 Copenhagen , Denmark
| | - Darryl S Pickering
- Department of Drug Design and Pharmacology , University of Copenhagen , Jagtvej 162 , DK-2100 Copenhagen , Denmark
| |
Collapse
|
20
|
Exciting Times: New Advances Towards Understanding the Regulation and Roles of Kainate Receptors. Neurochem Res 2017; 44:572-584. [PMID: 29270706 PMCID: PMC6420428 DOI: 10.1007/s11064-017-2450-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 11/27/2017] [Accepted: 12/07/2017] [Indexed: 12/11/2022]
Abstract
Kainate receptors (KARs) are glutamate-gated ion channels that play fundamental roles in regulating neuronal excitability and network function in the brain. After being cloned in the 1990s, important progress has been made in understanding the mechanisms controlling the molecular and cellular properties of KARs, and the nature and extent of their regulation of wider neuronal activity. However, there have been significant recent advances towards understanding KAR trafficking through the secretory pathway, their precise synaptic positioning, and their roles in synaptic plasticity and disease. Here we provide an overview highlighting these new findings about the mechanisms controlling KARs and how KARs, in turn, regulate other proteins and pathways to influence synaptic function.
Collapse
|
21
|
Li B, Rex E, Wang H, Qian Y, Ogden AM, Bleakman D, Johnson KW. Pharmacological Modulation of GluK1 and GluK2 by NETO1, NETO2, and PSD95. Assay Drug Dev Technol 2016; 14:131-43. [PMID: 26991362 DOI: 10.1089/adt.2015.689] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
The association between the kainate receptors (KARs) GluK1 and GluK2 and the modifying proteins neuropilin- and tolloid-like 1 (NETO1), neuropilin- and tolloid-like 2 (NETO2), and postsynaptic density protein 95 (PSD95) is likely to produce distinct GluK1 and GluK2 pharmacology in postsynaptic neurons. However, little is known about their corresponding modulatory effects on GluK1 and GluK2 activity in high-throughput assays for cell-based drug discovery. Using heterologous cells that potentially mimic the response in native cells in a fluorescence imaging plate reader (FLIPR) assay, we have investigated assays that incorporate (1) coexpression of GluK1 or GluK2 with their modulatory proteins (NETO1, NETO2, PSD95) and/or (2) enablement of assays with physiological concentration of native GluK1 and GluK2 agonist (glutamate) in the absence of an artificial potentiator (e.g., concanavalin A [Con A]). We found that in the absence of Con A, both NETO1 and NETO2 accessory proteins are able to potentiate kainate- and glutamate-evoked GluK1-mediated Ca(2+) influx. We also noted the striking ability of PSD95 to enhance glutamate-stimulated potentiation effects of NETO2 on GluK1 without the need for Con A and with a robust signal that could be utilized for high-throughput FLIPR assays. These experiments demonstrate the utility of heterologous cells coexpressing PSD95/NETO2 with GluK1 or GluK2 in native cell-mimicking heterologous cell systems for high-throughput assays and represent new avenues into the discovery of KAR modulating therapies.
Collapse
Affiliation(s)
- Baolin Li
- 1 Neuroscience Discovery, Lilly Corporate Center , Eli Lilly and Company, Indianapolis, Indiana
| | - Elizabeth Rex
- 1 Neuroscience Discovery, Lilly Corporate Center , Eli Lilly and Company, Indianapolis, Indiana
| | - He Wang
- 2 TTx-Reagents-Proteins, Lilly Corporate Center , Eli Lilly and Company, Indianapolis, Indiana
| | - Yuewei Qian
- 2 TTx-Reagents-Proteins, Lilly Corporate Center , Eli Lilly and Company, Indianapolis, Indiana
| | - Ann Marie Ogden
- 1 Neuroscience Discovery, Lilly Corporate Center , Eli Lilly and Company, Indianapolis, Indiana
| | - David Bleakman
- 1 Neuroscience Discovery, Lilly Corporate Center , Eli Lilly and Company, Indianapolis, Indiana
| | - Kirk W Johnson
- 1 Neuroscience Discovery, Lilly Corporate Center , Eli Lilly and Company, Indianapolis, Indiana
| |
Collapse
|
22
|
Exploring Genetic Factors Involved in Huntington Disease Age of Onset: E2F2 as a New Potential Modifier Gene. PLoS One 2015; 10:e0131573. [PMID: 26148071 PMCID: PMC4493078 DOI: 10.1371/journal.pone.0131573] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 06/03/2015] [Indexed: 01/09/2023] Open
Abstract
Age of onset (AO) of Huntington disease (HD) is mainly determined by the length of the CAG repeat expansion (CAGexp) in exon 1 of the HTT gene. Additional genetic variation has been suggested to contribute to AO, although the mechanism by which it could affect AO is presently unknown. The aim of this study is to explore the contribution of candidate genetic factors to HD AO in order to gain insight into the pathogenic mechanisms underlying this disorder. For that purpose, two AO definitions were used: the earliest age with unequivocal signs of HD (earliest AO or eAO), and the first motor symptoms age (motor AO or mAO). Multiple linear regression analyses were performed between genetic variation within 20 candidate genes and eAO or mAO, using DNA and clinical information of 253 HD patients from REGISTRY project. Gene expression analyses were carried out by RT-qPCR with an independent sample of 35 HD patients from Basque Country Hospitals. We found suggestive association signals between HD eAO and/or mAO and genetic variation within the E2F2, ATF7IP, GRIN2A, GRIN2B, LINC01559, HIP1 and GRIK2 genes. Among them, the most significant was the association between eAO and rs2742976, mapping to the promoter region of E2F2 transcription factor. Furthermore, rs2742976 T allele patient carriers exhibited significantly lower lymphocyte E2F2 gene expression, suggesting a possible implication of E2F2-dependent transcriptional activity in HD pathogenesis. Thus, E2F2 emerges as a new potential HD AO modifier factor.
Collapse
|
23
|
Demmer CS, Møller C, Brown PMGE, Han L, Pickering DS, Nielsen B, Bowie D, Frydenvang K, Kastrup JS, Bunch L. Binding mode of an α-amino acid-linked quinoxaline-2,3-dione analogue at glutamate receptor subtype GluK1. ACS Chem Neurosci 2015; 6:845-54. [PMID: 25856736 DOI: 10.1021/acschemneuro.5b00038] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Two α-amino acid-functionalized quinoxalines, 1a (CNG-10301) and 1b (CNG-10300), of a quinoxaline moiety coupled to an amino acid moiety were designed, synthesized, and characterized pharmacologically. While 1a displayed low affinity at native AMPA, KA, and NMDA receptors, and at homomeric GluK1,3 receptors, the affinity for GluK2 was in the midmicromolar range (Ki = 136 μM), 1b displayed low to midmicromolar range binding affinity at all the iGluRs (Ki = 9-126 μM). In functional experiments (outside-out patches excised from transfected HEK293T cells), 100 μM 1a partially blocked GluK1 (33% peak response), while GluK2 was unaffected (96% peak response). Furthermore, 1a was shown not to be an agonist at GluK1 and GluK2 at 100 μM. On the other hand, 100 μM 1b fully antagonized GluK1 (8% peak response) but only partially blocked GluK2 (33% peak response). An X-ray structure at 2.3 Å resolution of 1b in the GluK1-LBD (ligand-binding domain) disclosed an unexpected binding mode compared to the predictions made during the design phase; the quinoxaline moiety remains to act as an amino acid bioisostere, but the amino acid moiety is oriented into a new area within the GluK1 receptor. The structure of the GluK1-LBD with 1b showed a large variation in domain openings of the three molecules from 25° to 49°, demonstrating that the GluK1-LBD is capable of undergoing major domain movements.
Collapse
|
24
|
Hill M, Dušková M, Stárka L. Dehydroepiandrosterone, its metabolites and ion channels. J Steroid Biochem Mol Biol 2015; 145:293-314. [PMID: 24846830 DOI: 10.1016/j.jsbmb.2014.05.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2014] [Revised: 05/06/2014] [Accepted: 05/11/2014] [Indexed: 11/20/2022]
Abstract
This review is focused on the physiological and pathophysiological relevance of steroids influencing the activities of the central and peripheral nervous systems with regard to their concentrations in body fluids and tissues in various stages of human life like the fetal development or pregnancy. The data summarized in this review shows that DHEA and its unconjugated and sulfated metabolites are physiologically and pathophysiologically relevant in modulating numerous ion channels and participate in vital functions of the human organism. DHEA and its unconjugated and sulfated metabolites including 5α/β-reduced androstane steroids participate in various physiological and pathophysiological processes like the management of GnRH cyclic release, regulation of glandular and neurotransmitter secretions, maintenance of glucose homeostasis on one hand and insulin insensitivity on the other hand, control of skeletal muscle and smooth muscle activities including vasoregulation, promotion of tolerance to ischemia and other neuroprotective effects. In respect of prevalence of steroid sulfates over unconjugated steroids in the periphery and the opposite situation in the CNS, the sulfated androgens and androgen metabolites reach relevance in peripheral organs. The unconjugated androgens and estrogens are relevant in periphery and so much the more in the CNS due to higher concentrations of most unconjugated steroids in the CNS tissues than in circulation and peripheral organs. This article is part of a Special Issue entitled "Essential role of DHEA".
Collapse
Affiliation(s)
- M Hill
- Steroid Hormone Unit, Institute of Endocrinology, Národní třída 8, Prague 116 94, Praha 1, CZ 116 94, Czech Republic.
| | - M Dušková
- Steroid Hormone Unit, Institute of Endocrinology, Národní třída 8, Prague 116 94, Praha 1, CZ 116 94, Czech Republic.
| | - L Stárka
- Steroid Hormone Unit, Institute of Endocrinology, Národní třída 8, Prague 116 94, Praha 1, CZ 116 94, Czech Republic.
| |
Collapse
|
25
|
Venskutonytė R, Larsen AP, Frydenvang K, Gajhede M, Sagot E, Assaf Z, Gefflaut T, Pickering DS, Bunch L, Kastrup JS. Molecular recognition of two 2,4-syn-functionalized (S)-glutamate analogues by the kainate receptor GluK3 ligand binding domain. ChemMedChem 2014; 9:2254-9. [PMID: 25044437 DOI: 10.1002/cmdc.201402204] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Indexed: 11/11/2022]
Abstract
The kainate receptors are the least studied subfamily of ionotropic glutamate receptors. These receptors are thought to have a neuromodulatory role and have been associated with a variety of disorders in the central nervous system. This makes kainate receptors interesting potential drug targets. Today, structures of the ligand binding domain (LBD) of the kainate receptor GluK3 are only known in complex with the endogenous agonist glutamate, the natural product kainate, and two synthetic agonists. Herein we report structures of GluK3 LBD in complex with two 2,4-syn-functionalized (S)-glutamate analogues to investigate their structural potential as chemical scaffolds. Similar binding affinities at GluK3 were determined for the 2-(methylcarbamoyl)ethyl analogue (Ki =4.0 μM) and the 2-(methoxycarbonyl)ethyl analogue (Ki =1.7 μM), in agreement with the similar positioning of the compounds within the binding pocket. As the binding affinity is similar to that of glutamate, this type of Cγ substituent could be used as a scaffold for introduction of even larger substituents reaching into unexplored binding site regions to achieve subtype selectivity.
Collapse
Affiliation(s)
- Raminta Venskutonytė
- Department of Drug Design & Pharmacology, Faculty of Health & Medical Sciences, University of Copenhagen, 2100 Copenhagen (Denmark)
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Lee S, Hong Y, Park S, Lee SR, Chang KT, Hong Y. Comparison of surgical methods of transient middle cerebral artery occlusion between rats and mice. J Vet Med Sci 2014; 76:1555-61. [PMID: 25649935 PMCID: PMC4300368 DOI: 10.1292/jvms.14-0258] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Rodent models of focal cerebral ischemia that do not require craniotomy have been
developed by intraluminal suture middle cerebral artery occlusion (MCAo). Mouse MCAo
models have been widely used and extended to genetic studies of cell death or recovery
mechanisms. Therefore, we compared surgery-related parameters and techniques between such
rats and mice. In rodent MCAo models, has to be considered body temperature during the
operative period, as well as the need for the use of a standardized tip in terms of the
outer diameter of probes. Induction of focal cerebral ischemia was measured by
neurological dysfunction parameters. Our methods could induce stable moderate-severity
ischemic brain injury models and histological alteration at 24 hr after MCAo surgery.
Moreover approximately 80% (rats) and 85% (mice) survival ratios were shown indicating
with model engineering success. Finally, we described and compared major parameters
between rats and mice, including probe size, thread insert length, operation and occlusion
periods, and differences in the procedures.
Collapse
Affiliation(s)
- Seunghoon Lee
- Biohealth Products Research Center (BPRC), Inje University, Gimhae, Korea
| | | | | | | | | | | |
Collapse
|
27
|
Bolea I, Colivicchi MA, Ballini C, Marco-Contelles J, Tipton KF, Unzeta M, Della Corte L. Neuroprotective effects of the MAO-B inhibitor, PF9601N, in an in vivo model of excitotoxicity. CNS Neurosci Ther 2014; 20:641-50. [PMID: 24767579 DOI: 10.1111/cns.12271] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Revised: 02/18/2014] [Accepted: 03/22/2014] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND PF9601N [N-(2-propynyl)-2-(5-benzyloxy-indolyl) methylamine] is an inhibitor of monoamine oxidase B (MAO-B), which has shown to possess neuroprotective properties in several in vitro and in vivo models of Parkinson's disease (PD). As there is evidence that excitotoxicity may be implicated in the pathophysiology of several neurodegenerative diseases, the aim of the present work was to investigate the effects of PF9601N in an acute in vivo model of excitotoxicity induced by the local administration of kainic acid during striatal microdialysis in adult rats. METHODS The basal and evoked release of neurotransmitters was monitored by HPLC analysis of microdialysate samples and tissue damage was evaluated histologically "ex vivo." RESULTS PF9601N (40 mg/kg, single i.p. administration) reduced the kainate-evoked release of glutamate and aspartate and increased taurine release, but it had no effect on the release of dopamine, DOPAC, and HVA. PF9601N pretreatment also resulted in a significant reduction in the kainate-induced astrocytosis, microgliosis, and apoptosis. CONCLUSIONS The results suggest PF9601N to be a good candidate for the treatment of neurodegenerative diseases mediated by excitotoxicity.
Collapse
Affiliation(s)
- Irene Bolea
- Departament de Bioquimica i Biologia Molecular, Facultat de Medicina, Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain; Dipartimento di Neuroscienze, Psicologia Area del Farmaco e Salute del Bambino (NEUROFARBA), Università degli Studi di Firenze, Firenze, Italy
| | | | | | | | | | | | | |
Collapse
|
28
|
Kaczor AA, Karczmarzyk Z, Fruziński A, Pihlaja K, Sinkkonen J, Wiinämaki K, Kronbach C, Unverferth K, Poso A, Matosiuk D. Structural studies, homology modeling and molecular docking of novel non-competitive antagonists of GluK1/GluK2 receptors. Bioorg Med Chem 2014; 22:787-95. [PMID: 24368028 DOI: 10.1016/j.bmc.2013.12.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Revised: 12/02/2013] [Accepted: 12/05/2013] [Indexed: 01/19/2023]
Abstract
Non-competitive ligands of kainate receptors have focused significant attention as medicinal compounds because they seem to be better tolerated than competitive antagonists and uncompetitive blocker of these receptors. Here we present structural studies (X-ray structure determination, NMR and MS spectra) of novel indole-derived non-competitive antagonists of GluK1/GluK2 receptors, homology models of GluK1 and GluK2 receptors based on novel AMPA receptor template as well as molecular docking of ligands to their molecular targets. We find that the allosteric site is in the receptor transduction domain, in one receptor subunit, not between the two subunits as it was indicated by our earlier studies.
Collapse
Affiliation(s)
- Agnieszka A Kaczor
- Department of Synthesis and Chemical Technology of Pharmaceutical Substances with Computer Modeling Lab, Faculty of Pharmacy with Division of Medical Analytics, 4A Chodźki St., PL-20093 Lublin, Poland; School of Pharmacy, University of Eastern Finland, Yliopistonranta 1, PO Box 1627, FI-70211 Kuopio, Finland.
| | - Zbigniew Karczmarzyk
- Department of Chemistry, Siedlce University, 3 Maja 54 St., PL-08110 Siedlce, Poland
| | - Andrzej Fruziński
- Institute of General and Ecological Chemistry, Technical University, Żeromskiego115 St.,PL-90924 Łódź, Poland
| | - Kalevi Pihlaja
- Department of Chemistry, University of Turku, Vatselankatu 2, FI-20014 Turku, Finland
| | - Jari Sinkkonen
- Department of Chemistry, University of Turku, Vatselankatu 2, FI-20014 Turku, Finland
| | - Kirsti Wiinämaki
- Department of Chemistry, University of Turku, Vatselankatu 2, FI-20014 Turku, Finland
| | | | - Klaus Unverferth
- Biotie Therapie GmbH, Meissner Str. 191, DE-01445 Radebul, Germany
| | - Antti Poso
- School of Pharmacy, University of Eastern Finland, Yliopistonranta 1, PO Box 1627, FI-70211 Kuopio, Finland
| | - Dariusz Matosiuk
- Department of Synthesis and Chemical Technology of Pharmaceutical Substances with Computer Modeling Lab, Faculty of Pharmacy with Division of Medical Analytics, 4A Chodźki St., PL-20093 Lublin, Poland.
| |
Collapse
|
29
|
Abstract
Our understanding of the molecular properties of kainate receptors and their involvement in synaptic physiology has progressed significantly over the last 30 years. A plethora of studies indicate that kainate receptors are important mediators of the pre- and postsynaptic actions of glutamate, although the mechanisms underlying such effects are still often a topic for discussion. Three clear fields related to their behavior have emerged: there are a number of interacting proteins that pace the properties of kainate receptors; their activity is unconventional since they can also signal through G proteins, behaving like metabotropic receptors; they seem to be linked to some devastating brain diseases. Despite the significant progress in their importance in brain function, kainate receptors remain somewhat puzzling. Here we examine discoveries linking these receptors to physiology and their probable implications in disease, in particular mood disorders, and propose some ideas to obtain a deeper understanding of these intriguing proteins.
Collapse
|
30
|
Cavaliere F, Benito-Muñoz M, Panicker M, Matute C. NMDA modulates oligodendrocyte differentiation of subventricular zone cells through PKC activation. Front Cell Neurosci 2013; 7:261. [PMID: 24391542 PMCID: PMC3866621 DOI: 10.3389/fncel.2013.00261] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Accepted: 12/01/2013] [Indexed: 12/21/2022] Open
Abstract
Multipotent cells from the juvenile subventricular zone (SVZ) possess the ability to differentiate into new neural cells. Depending on local signals, SVZ can generate new neurons, astrocytes, or oligodendrocytes. We previously demonstrated that activation of NMDA receptors in SVZ progenitors increases the rate of oligodendrocyte differentiation. Here we investigated the mechanisms involved in NMDA receptor-dependent differentiation. Using functional studies performed with the reporter gene luciferase we found that activation of NMDA receptor stimulates PKC. In turn, stimulation of PKC precedes the activation of NADPH oxidase (NOX) as demonstrated by translocation of the p67phox subunit to the cellular membrane. We propose that NOX2 is involved in the transduction of the signal from NMDA receptors through PKC activation as the inhibitor gp91 reduced their pro-differentiation effect. In addition, our data and that from other groups suggest that signaling through the NMDA receptor/PKC/NOX2 cascade generates ROS that activate the PI3/mTOR pathway and finally leads to the generation of new oligodendrocytes.
Collapse
Affiliation(s)
- Fabio Cavaliere
- Departamento de Neurociencias, Universidad del País Vasco (UPV/EHU) Leioa, Spain ; Achucarro Basque Center for Neuroscience, Universidad del País Vasco (UPV/EHU) Zamudio, Spain ; Instituto de Salud Carlos III, Centro Investigación Biomédica en Red Enfermedades Neurodegenerativas Leioa, Spain
| | - Monica Benito-Muñoz
- Departamento de Neurociencias, Universidad del País Vasco (UPV/EHU) Leioa, Spain ; Achucarro Basque Center for Neuroscience, Universidad del País Vasco (UPV/EHU) Zamudio, Spain ; Instituto de Salud Carlos III, Centro Investigación Biomédica en Red Enfermedades Neurodegenerativas Leioa, Spain
| | - Mitradas Panicker
- National Centre for Biological Sciences, UAS-GKVK Campus Bangalore, India
| | - Carlos Matute
- Departamento de Neurociencias, Universidad del País Vasco (UPV/EHU) Leioa, Spain ; Achucarro Basque Center for Neuroscience, Universidad del País Vasco (UPV/EHU) Zamudio, Spain ; Instituto de Salud Carlos III, Centro Investigación Biomédica en Red Enfermedades Neurodegenerativas Leioa, Spain
| |
Collapse
|
31
|
Szaroma W, Dziubek K, Greń A, Kreczmer B, Kapusta E. Influence of the kainic acid on antioxidant status in the brain, liver and kidneys of the mouse. ACTA ACUST UNITED AC 2012; 99:447-59. [DOI: 10.1556/aphysiol.99.2012.4.9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
32
|
Majo VJ, Prabhakaran J, Mann JJ, Kumar JSD. PET and SPECT tracers for glutamate receptors. Drug Discov Today 2012; 18:173-84. [PMID: 23092894 DOI: 10.1016/j.drudis.2012.10.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2012] [Revised: 07/10/2012] [Accepted: 10/15/2012] [Indexed: 12/19/2022]
Abstract
Radioligands for PET imaging of glutamate receptors will have the potential for studying neurological and neuropsychiatric disorders and their diagnosis and therapeutic intervention. Glutamate is the major excitatory neurotransmitter in the brain and is implicated in the pathophysiology of many neurodegenerative and neuropsychiatric disorders. Glutamate and its receptors are potential targets in the treatment of these disorders. Glutamate signaling is mediated through ionotropic and metabotropic receptors. The abundant concentration of these receptors can facilitate their in vivo quantification using positron emission tomography (PET). Glutamate receptors are a potentially important set of targets for monitoring disease progression, for evaluating the effect of therapy and for new treatment development based on the quantification of receptor occupancy. Here, we review the PET and single-photon emission computed tomography (SPECT) radioligands that have been developed for imaging glutamate receptors in living brain.
Collapse
Affiliation(s)
- Vattoly J Majo
- Division of Molecular Imaging and Neuropathology, Department of Psychiatry, Columbia University, New York, NY 10032, USA
| | | | | | | |
Collapse
|
33
|
Zhang HJ, Li C, Zhang GY. ATPA induced GluR5-containing kainite receptor S-nitrosylation via activation of GluR5-Gq-PLC-IP(3)R pathway and signalling module GluR5·PSD-95·nNOS. Int J Biochem Cell Biol 2012; 44:2261-71. [PMID: 23000395 DOI: 10.1016/j.biocel.2012.09.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Revised: 08/17/2012] [Accepted: 09/14/2012] [Indexed: 11/19/2022]
Abstract
GluR5-containing kainite receptor (GluR5-KAR) plays an important role in the pathophysiology of nervous system diseases, while S-nitrosylation exerts a variety of effects on biological systems. However, the mechanism of GluR5-KAR S-nitrosylation is still unclear up to now. Here our researches found that GluR5-KAR selective agonist ATPA stimulation activated the nonclassical GluR5-KAR-Gq-PLC-IP(3)R pathway and the signalling module GluR5·PSD-95·nNOS (the former is more important), led to Ca(2+) release from intracellular calcium stores endoplasmic reticulum (ER) to cytoplasm and extracellular calcium indrawal, respectively, which further resulted in nNOS activation and GluR5-KAR S-nitrosylation, and then inhibited GluR5-mediated whole-cell current attenuation and induced apoptosis in primary cultured hippocampal neurons. Clarification of the primary mechanisms of GluR5-KAR S-nitrosylation induced by ATPA and identification of critical cysteine for GluR5-2a S-nitrosylation (Cys231 and Cys804) open up a brand-new field for revealing downstream signalling pathway of GluR5-KAR and its molecular characteristics, exploring the pathogenesis of neurological diseases and searching for promising therapies.
Collapse
Affiliation(s)
- Hai-Jian Zhang
- Research Center of Biochemistry and Molecular Biology and Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical College, Xuzhou, China
| | | | | |
Collapse
|
34
|
Catches JS, Xu J, Contractor A. Genetic ablation of the GluK4 kainate receptor subunit causes anxiolytic and antidepressant-like behavior in mice. Behav Brain Res 2011; 228:406-14. [PMID: 22203159 DOI: 10.1016/j.bbr.2011.12.026] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2011] [Revised: 12/12/2011] [Accepted: 12/14/2011] [Indexed: 11/30/2022]
Abstract
There is a clear link between dysregulation of glutamatergic signaling and mood disorders. Genetic variants in the glutamate receptor gene GRIK4, which encodes the kainate receptor subunit GluK4, alter the susceptibility for depression, bipolar disorder and schizophrenia. Here we demonstrate that Grik4(-/-) mice have reduced anxiety and an antidepressant-like phenotype. In the elevated zero-maze, a test for anxiety and risk taking behavior, Grik4(-/-) mice spent significantly more time exploring the open areas of the maze. In anxiogenic tests of marble-burying and novelty-induced suppression of feeding, anxiety-like behavior was consistently reduced in knockout animals. In the forced swim test, a test of learned helplessness that is used to determine depression-like behavior, knockout mice demonstrated significantly less immobility suggesting that Grik4 ablation has an antidepressant-like effect. Finally, in the sucrose preference test, a test for anhedonia in rodents, Grik4(-/-) mice demonstrated increased sucrose preference. Expression of the GluK4 receptor subunit in the forebrain is restricted to the CA3 region of the hippocampus and dentate gyrus regions where KARs are known to modulate synaptic plasticity. We tested whether Grik4 ablation had effects on mossy fiber (MF) plasticity and found there to be a significant impairment in LTP likely through a loss of KAR modulation of excitability of the presynaptic MF axons. These studies demonstrate a clear anxiolytic and antidepressant phenotype associated with ablation of Grik4 and a parallel disruption in hippocampal plasticity, providing support for the importance of this receptor subunit in mood disorders.
Collapse
Affiliation(s)
- Justin S Catches
- Department of Physiology, Northwestern University, Feinberg School of Medicine, 303 E Chicago Avenue, Chicago, IL 60611, USA.
| | | | | |
Collapse
|
35
|
González-González IM, Konopacki FA, Rocca DL, Doherty AJ, Jaafari N, Wilkinson KA, Henley JM. Kainate receptor trafficking. ACTA ACUST UNITED AC 2011. [DOI: 10.1002/wmts.23] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|