1
|
Santos TDS, Contrim IS, da Silva DF, Assumpção MEOD, de Paula-Lopes FF, Feitosa WB. Heat shock affects the Ca 2+/calmodulin-dependent protein kinase II dynamic during bovine sperm capacitation and acrosome reaction. Front Cell Dev Biol 2025; 13:1552282. [PMID: 40241796 PMCID: PMC12001034 DOI: 10.3389/fcell.2025.1552282] [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: 12/27/2024] [Accepted: 02/18/2025] [Indexed: 04/18/2025] Open
Abstract
Background Heat shock during sperm capacitation affects the spermatozoa quality, resulting in increased early acrosome reaction and consequently decreasing their fertilizing capacity. Although the mechanisms involved in the regulation of sperm capacitation and acrosome reaction are not fully understood, it has been reported that Ca2+/calmodulin-dependent protein kinase II (CaMKII) is an important regulator of these processes. Thus, the present aimed to evaluate the effect of heat shock in the CaMKII signaling during the bovine sperm capacitation and acrosome. Methods Bovine spermatozoa were in vitro capacitated for 4 hours. The acrosome reaction was induced by exposure to heparin and calcium ionophore A23187 for 1 hour. Heat shock was applied by incubating spermatozoa at 41 °C with 7% CO2, while the control group was maintained at 38.5 °C with 5% CO2. At the end of each treatment, the localization of total CaMKII and phosphorylated CaMKII (pCaMKII), as well as acrosomal membrane integrity, were evaluated by immunofluorescence. Results It was observed that CaMKII and not phosphorylated CaMKII (pCaMKII) localization at the acrosome region was affected by sperm capacitation. In contrast, the localization of both, CaMKII and its phosphorylated form was affected by the acrosome reaction (p < 0.05). The acrosome membrane integrity, as well as the pCamKII localization in bovine spermatozoa, was affected by incubation time. This effect of incubation time was stronger in heated shock sperm, although it was observed only after 2 h of incubation. Heat shock also affected the acrosomal localization of pCaMKII in the acrosomal region of spermatozoa with intact acrosome. Discussion Taken together, the data present here show that CaMKII and pCaMKII localization is dynamic during bovine sperm capacitation and acrosome reaction and that this pattern of localization is affected by heat shock, suggesting that failure in CaMKII signaling is probably involved in the early acrosome reaction observed in heated-shock spermatozoa.
Collapse
Affiliation(s)
- Thais de Sousa Santos
- Department of Biological Sciences, Federal University of Sao Paulo, Diadema, Sao Paulo, Brazil
| | | | | | | | | | - Weber Beringui Feitosa
- Department of Biological Sciences, Federal University of Sao Paulo, Diadema, Sao Paulo, Brazil
| |
Collapse
|
2
|
Patton MH, Thomas KT, Bayazitov IT, Newman KD, Kurtz NB, Robinson CG, Ramirez CA, Trevisan AJ, Bikoff JB, Peters ST, Pruett-Miller SM, Jiang Y, Schild AB, Nityanandam A, Zakharenko SS. Synaptic plasticity in human thalamocortical assembloids. Cell Rep 2024; 43:114503. [PMID: 39018245 PMCID: PMC11407288 DOI: 10.1016/j.celrep.2024.114503] [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: 03/11/2024] [Revised: 04/23/2024] [Accepted: 06/27/2024] [Indexed: 07/19/2024] Open
Abstract
Synaptic plasticities, such as long-term potentiation (LTP) and depression (LTD), tune synaptic efficacy and are essential for learning and memory. Current studies of synaptic plasticity in humans are limited by a lack of adequate human models. Here, we modeled the thalamocortical system by fusing human induced pluripotent stem cell-derived thalamic and cortical organoids. Single-nucleus RNA sequencing revealed that >80% of cells in thalamic organoids were glutamatergic neurons. When fused to form thalamocortical assembloids, thalamic and cortical organoids formed reciprocal long-range axonal projections and reciprocal synapses detectable by light and electron microscopy, respectively. Using whole-cell patch-clamp electrophysiology and two-photon imaging, we characterized glutamatergic synaptic transmission. Thalamocortical and corticothalamic synapses displayed short-term plasticity analogous to that in animal models. LTP and LTD were reliably induced at both synapses; however, their mechanisms differed from those previously described in rodents. Thus, thalamocortical assembloids provide a model system for exploring synaptic plasticity in human circuits.
Collapse
Affiliation(s)
- Mary H Patton
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Kristen T Thomas
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Ildar T Bayazitov
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Kyle D Newman
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Nathaniel B Kurtz
- Cell and Tissue Imaging Center, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Camenzind G Robinson
- Cell and Tissue Imaging Center, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Cody A Ramirez
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Alexandra J Trevisan
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Jay B Bikoff
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Samuel T Peters
- Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Shondra M Pruett-Miller
- Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Yanbo Jiang
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Andrew B Schild
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Anjana Nityanandam
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Stanislav S Zakharenko
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| |
Collapse
|
3
|
Boutonnet M, Carpena C, Bouquier N, Chastagnier Y, Font-Ingles J, Moutin E, Tricoire L, Chemin J, Perroy J. Voltage tunes mGlu 5 receptor function, impacting synaptic transmission. Br J Pharmacol 2024; 181:1793-1811. [PMID: 38369690 DOI: 10.1111/bph.16317] [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: 09/07/2023] [Revised: 12/08/2023] [Accepted: 12/29/2023] [Indexed: 02/20/2024] Open
Abstract
BACKGROUND AND PURPOSE Voltage sensitivity is a common feature of many membrane proteins, including some G-protein coupled receptors (GPCRs). However, the functional consequences of voltage sensitivity in GPCRs are not well understood. EXPERIMENTAL APPROACH In this study, we investigated the voltage sensitivity of the post-synaptic metabotropic glutamate receptor mGlu5 and its impact on synaptic transmission. Using biosensors and electrophysiological recordings in non-excitable HEK293T cells or neurons. KEY RESULTS We found that mGlu5 receptor function is optimal at resting membrane potentials. We observed that membrane depolarization significantly reduced mGlu5 receptor activation, Gq-PLC/PKC stimulation, Ca2+ release and mGlu5 receptor-gated currents through transient receptor potential canonical, TRPC6, channels or glutamate ionotropic NMDA receptors. Notably, we report a previously unknown activity of the NMDA receptor at the resting potential of neurons, enabled by mGlu5. CONCLUSIONS AND IMPLICATIONS Our findings suggest that mGlu5 receptor activity is directly regulated by membrane voltage which may have a significant impact on synaptic processes and pathophysiological functions.
Collapse
Affiliation(s)
- Marin Boutonnet
- IGF, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Camille Carpena
- IGF, University of Montpellier, CNRS, INSERM, Montpellier, France
| | | | - Yan Chastagnier
- IGF, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Joan Font-Ingles
- IGF, University of Montpellier, CNRS, INSERM, Montpellier, France
- SpliceBio, Barcelona, Spain
| | - Enora Moutin
- IGF, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Ludovic Tricoire
- Neuroscience Paris Seine, Institut de biologie Paris Seine, Sorbonne universite, CNRS, INSERM, Paris, France
| | - Jean Chemin
- IGF, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Julie Perroy
- IGF, University of Montpellier, CNRS, INSERM, Montpellier, France
| |
Collapse
|
4
|
Deep SN, Seelig S, Paul S, Poddar R. Homocysteine-induced sustained GluN2A NMDA receptor stimulation leads to mitochondrial ROS generation and neurotoxicity. J Biol Chem 2024; 300:107253. [PMID: 38569938 PMCID: PMC11081806 DOI: 10.1016/j.jbc.2024.107253] [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: 09/28/2023] [Revised: 03/11/2024] [Accepted: 03/24/2024] [Indexed: 04/05/2024] Open
Abstract
Homocysteine, a sulfur-containing amino acid derived from methionine metabolism, is a known agonist of N-methyl-D-aspartate receptor (NMDAR) and is involved in neurotoxicity. Our previous findings showed that neuronal exposure to elevated homocysteine levels leads to sustained low-level increase in intracellular Ca2+, which is dependent on GluN2A subunit-containing NMDAR (GluN2A-NMDAR) stimulation. These studies further showed a role of ERK MAPK in homocysteine-GluN2A-NMDAR-mediated neuronal death. However, the intracellular mechanisms associated with such sustained GluN2A-NMDAR stimulation and subsequent Ca2+ influx have remained unexplored. Using live-cell imaging with Fluo3-AM and biochemical approaches, we show that homocysteine-GluN2A NMDAR-induced initial Ca2+ influx triggers sequential phosphorylation and subsequent activation of the proline rich tyrosine kinase 2 (Pyk2) and Src family kinases, which in turn phosphorylates GluN2A-Tyr1325 residue of GluN2A-NMDARs to maintain channel activity. The continuity of this cycle of events leads to sustained Ca2+ influx through GluN2A-NMDAR. Our findings also show that lack of activation of the regulatory tyrosine phosphatase STEP, which can limit Pyk2 and Src family kinase activity further contributes to the maintenance of this cycle. Additional studies using live-cell imaging of neurons expressing a redox-sensitive GFP targeted to the mitochondrial matrix show that treatment with homocysteine leads to a progressive increase in mitochondrial reactive oxygen species generation, which is dependent on GluN2A-NMDAR-mediated sustained ERK MAPK activation. This later finding demonstrates a novel role of GluN2A-NMDAR in homocysteine-induced mitochondrial ROS generation and highlights the role of ERK MAPK as the intermediary signaling pathway between GluN2A-NMDAR stimulation and mitochondrial reactive oxygen species generation.
Collapse
Affiliation(s)
- Satya Narayan Deep
- Department of Neurology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA
| | - Sarah Seelig
- Department of Neurology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA
| | - Surojit Paul
- Department of Neurology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA
| | - Ranjana Poddar
- Department of Neurology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA.
| |
Collapse
|
5
|
Patton MH, Thomas KT, Bayazitov IT, Newman KD, Kurtz NB, Robinson CG, Ramirez CA, Trevisan AJ, Bikoff JB, Peters ST, Pruett-Miller SM, Jiang Y, Schild AB, Nityanandam A, Zakharenko SS. Synaptic plasticity in human thalamocortical assembloids. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.01.578421. [PMID: 38352415 PMCID: PMC10862901 DOI: 10.1101/2024.02.01.578421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/25/2024]
Abstract
Synaptic plasticities, such as long-term potentiation (LTP) and depression (LTD), tune synaptic efficacy and are essential for learning and memory. Current studies of synaptic plasticity in humans are limited by a lack of adequate human models. Here, we modeled the thalamocortical system by fusing human induced pluripotent stem cell-derived thalamic and cortical organoids. Single-nucleus RNA-sequencing revealed that most cells in mature thalamic organoids were glutamatergic neurons. When fused to form thalamocortical assembloids, thalamic and cortical organoids formed reciprocal long-range axonal projections and reciprocal synapses detectable by light and electron microscopy, respectively. Using whole-cell patch-clamp electrophysiology and two-photon imaging, we characterized glutamatergic synaptic transmission. Thalamocortical and corticothalamic synapses displayed short-term plasticity analogous to that in animal models. LTP and LTD were reliably induced at both synapses; however, their mechanisms differed from those previously described in rodents. Thus, thalamocortical assembloids provide a model system for exploring synaptic plasticity in human circuits.
Collapse
Affiliation(s)
- Mary H. Patton
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital; Memphis, TN 38105, USA
| | - Kristen T. Thomas
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital; Memphis, TN 38105, USA
| | - Ildar T. Bayazitov
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital; Memphis, TN 38105, USA
| | - Kyle D. Newman
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital; Memphis, TN 38105, USA
| | - Nathaniel B. Kurtz
- Cell and Tissue Imaging Center, St. Jude Children’s Research Hospital; Memphis, TN 38105, USA
| | - Camenzind G. Robinson
- Cell and Tissue Imaging Center, St. Jude Children’s Research Hospital; Memphis, TN 38105, USA
| | - Cody A. Ramirez
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital; Memphis, TN 38105, USA
| | - Alexandra J. Trevisan
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital; Memphis, TN 38105, USA
| | - Jay B. Bikoff
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital; Memphis, TN 38105, USA
| | - Samuel T. Peters
- Center for Advanced Genome Engineering, St. Jude Children’s Research Hospital; Memphis, TN 38105, USA
| | - Shondra M. Pruett-Miller
- Center for Advanced Genome Engineering, St. Jude Children’s Research Hospital; Memphis, TN 38105, USA
- Department of Cell & Molecular Biology, St. Jude Children’s Research Hospital; Memphis, TN 38105, USA
| | - Yanbo Jiang
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital; Memphis, TN 38105, USA
| | - Andrew B. Schild
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital; Memphis, TN 38105, USA
| | - Anjana Nityanandam
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital; Memphis, TN 38105, USA
| | - Stanislav S. Zakharenko
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital; Memphis, TN 38105, USA
| |
Collapse
|
6
|
Purnell BS, Alves M, Boison D. Astrocyte-neuron circuits in epilepsy. Neurobiol Dis 2023; 179:106058. [PMID: 36868484 DOI: 10.1016/j.nbd.2023.106058] [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: 12/19/2022] [Revised: 02/20/2023] [Accepted: 02/27/2023] [Indexed: 03/05/2023] Open
Abstract
The epilepsies are a diverse spectrum of disease states characterized by spontaneous seizures and associated comorbidities. Neuron-focused perspectives have yielded an array of widely used anti-seizure medications and are able to explain some, but not all, of the imbalance of excitation and inhibition which manifests itself as spontaneous seizures. Furthermore, the rate of pharmacoresistant epilepsy remains high despite the regular approval of novel anti-seizure medications. Gaining a more complete understanding of the processes that turn a healthy brain into an epileptic brain (epileptogenesis) as well as the processes which generate individual seizures (ictogenesis) may necessitate broadening our focus to other cell types. As will be detailed in this review, astrocytes augment neuronal activity at the level of individual neurons in the form of gliotransmission and the tripartite synapse. Under normal conditions, astrocytes are essential to the maintenance of blood-brain barrier integrity and remediation of inflammation and oxidative stress, but in epilepsy these functions are impaired. Epilepsy results in disruptions in the way astrocytes relate to each other by gap junctions which has important implications for ion and water homeostasis. In their activated state, astrocytes contribute to imbalances in neuronal excitability due to their decreased capacity to take up and metabolize glutamate and an increased capacity to metabolize adenosine. Furthermore, due to their increased adenosine metabolism, activated astrocytes may contribute to DNA hypermethylation and other epigenetic changes that underly epileptogenesis. Lastly, we will explore the potential explanatory power of these changes in astrocyte function in detail in the specific context of the comorbid occurrence of epilepsy and Alzheimer's disease and the disruption in sleep-wake regulation associated with both conditions.
Collapse
Affiliation(s)
- Benton S Purnell
- Department of Neurosurgery, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, United States of America
| | - Mariana Alves
- Department of Neurosurgery, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, United States of America; Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin D02 YN77, Ireland
| | - Detlev Boison
- Department of Neurosurgery, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, United States of America; Brain Health Institute, Rutgers University, Piscataway, NJ, United States of America.
| |
Collapse
|
7
|
Azam S, Jakaria M, Kim J, Ahn J, Kim IS, Choi DK. Group I mGluRs in Therapy and Diagnosis of Parkinson’s Disease: Focus on mGluR5 Subtype. Biomedicines 2022; 10:biomedicines10040864. [PMID: 35453614 PMCID: PMC9032558 DOI: 10.3390/biomedicines10040864] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/05/2022] [Accepted: 04/06/2022] [Indexed: 11/16/2022] Open
Abstract
Metabotropic glutamate receptors (mGluRs; members of class C G-protein-coupled receptors) have been shown to modulate excitatory neurotransmission, regulate presynaptic extracellular glutamate levels, and modulate postsynaptic ion channels on dendritic spines. mGluRs were found to activate myriad signalling pathways to regulate synapse formation, long-term potentiation, autophagy, apoptosis, necroptosis, and pro-inflammatory cytokines release. A notorious expression pattern of mGluRs has been evident in several neurodegenerative diseases, including Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, and schizophrenia. Among the several mGluRs, mGluR5 is one of the most investigated types of considered prospective therapeutic targets and potential diagnostic tools in neurodegenerative diseases and neuropsychiatric disorders. Recent research showed mGluR5 radioligands could be a potential tool to assess neurodegenerative disease progression and trace respective drugs’ kinetic properties. This article provides insight into the group I mGluRs, specifically mGluR5, in the progression and possible therapy for PD.
Collapse
Affiliation(s)
- Shofiul Azam
- Department of Applied Life Science, Graduate School, BK21 Program, Konkuk University, Chungju 27478, Korea; (S.A.); (M.J.); (J.K.); (J.A.)
| | - Md. Jakaria
- Department of Applied Life Science, Graduate School, BK21 Program, Konkuk University, Chungju 27478, Korea; (S.A.); (M.J.); (J.K.); (J.A.)
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC 3052, Australia
| | - JoonSoo Kim
- Department of Applied Life Science, Graduate School, BK21 Program, Konkuk University, Chungju 27478, Korea; (S.A.); (M.J.); (J.K.); (J.A.)
| | - Jaeyong Ahn
- Department of Applied Life Science, Graduate School, BK21 Program, Konkuk University, Chungju 27478, Korea; (S.A.); (M.J.); (J.K.); (J.A.)
| | - In-Su Kim
- Department of Biotechnology, College of Biomedical and Health Science, Research Institute of Inflammatory Disease (RID), Konkuk University, Chungju 27478, Korea
- Correspondence: (I.-S.K.); (D.-K.C.); Tel.: +82-43-840-3905 (I.-S.K.); +82-43-840-3610 (D.-K.C.); Fax: +82-43-840-3872 (D.-K.C.)
| | - Dong-Kug Choi
- Department of Applied Life Science, Graduate School, BK21 Program, Konkuk University, Chungju 27478, Korea; (S.A.); (M.J.); (J.K.); (J.A.)
- Department of Biotechnology, College of Biomedical and Health Science, Research Institute of Inflammatory Disease (RID), Konkuk University, Chungju 27478, Korea
- Correspondence: (I.-S.K.); (D.-K.C.); Tel.: +82-43-840-3905 (I.-S.K.); +82-43-840-3610 (D.-K.C.); Fax: +82-43-840-3872 (D.-K.C.)
| |
Collapse
|
8
|
Mao LM, Demehri S, Wang JQ. Upregulation of Src Family Tyrosine Kinases in the Rat Striatum by Adenosine A 2A Receptors. J Mol Neurosci 2022; 72:802-811. [PMID: 35041190 PMCID: PMC8986616 DOI: 10.1007/s12031-021-01961-0] [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: 08/18/2021] [Accepted: 12/20/2021] [Indexed: 11/25/2022]
Abstract
Adenosine A2A receptors are Golf-coupled receptors and are predominantly expressed in the striatum of mammalian brains. As a mostly postsynaptic receptor, A2A receptors are implicated in the regulation of a variety of intracellular signaling pathways in striatopallidal output neurons and are linked to the pathogenesis of various neuropsychiatric and neurological disorders. This study investigated the possible role of A2A receptors in the modulation of the Src family kinase (SFK) in the adult rat striatum. In acutely prepared striatal slices, adding the A2A receptor agonist PSB-0777 induced a significant increase in phosphorylation of SFKs at a conserved autophosphorylation site (Y416) in the caudate putamen (CPu). This increase was also seen in the nucleus accumbens (NAc). Another A2A agonist CGS-21680 showed the similar ability to elevate SFK Y416 phosphorylation in the striatum. Treatment with the A2A receptor antagonist KW-6002 blocked the effect of PSB-0777 on SFK Y416 phosphorylation. In addition, PSB-0777 enhanced kinase activity of two key SFK members (Src and Fyn) immunoprecipitated from the striatum. These data demonstrate a positive linkage from A2A receptors to the SFK signaling pathway in striatal neurons. Activation of A2A receptors leads to the upregulation of phosphorylation of SFKs (Src and Fyn) at an activation-associated autophosphorylation site and kinase activity of these SFK members.
Collapse
Affiliation(s)
- Li-Min Mao
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City, Kansas City, MO, 64108, USA
| | - Shannon Demehri
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City, Kansas City, MO, 64108, USA
| | - John Q Wang
- Department of Biomedical Sciences, 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
|
9
|
A recurrent SHANK1 mutation implicated in autism spectrum disorder causes autistic-like core behaviors in mice via downregulation of mGluR1-IP3R1-calcium signaling. Mol Psychiatry 2022; 27:2985-2998. [PMID: 35388181 PMCID: PMC9205781 DOI: 10.1038/s41380-022-01539-1] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 03/09/2022] [Accepted: 03/21/2022] [Indexed: 12/27/2022]
Abstract
The genetic etiology and underlying mechanism of autism spectrum disorder (ASD) remain elusive. SHANK family genes (SHANK1/2/3) are well known ASD-related genes. However, little is known about how SHANK missense mutations contribute to ASD. Here, we aimed to clarify the molecular mechanism of and the multilevel neuropathological features induced by Shank1 mutations in knock-in (KI) mice. In this study, by sequencing the SHANK1 gene in a cohort of 615 ASD patients and 503 controls, we identified an ASD-specific recurrent missense mutation, c.2621 G > A (p.R874H). This mutation demonstrated strong pathogenic potential in in vitro experiments, and we generated the corresponding Shank1 R882H-KI mice. Shank1 R882H-KI mice displayed core symptoms of ASD, namely, social disability and repetitive behaviors, without confounding comorbidities of abnormal motor function and heightened anxiety. Brain structural changes in the frontal cortex, hippocampus and cerebellar cortex were observed in Shank1 R882H-KI mice via structural magnetic resonance imaging. These key brain regions also showed severe and consistent downregulation of mGluR1-IP3R1-calcium signaling, which subsequently affected the release of intracellular calcium. Corresponding cellular structural and functional changes were present in Shank1 R882H-KI mice, including decreased spine size, reduced spine density, abnormal morphology of postsynaptic densities, and impaired hippocampal long-term potentiation and basal excitatory transmission. These findings demonstrate the causative role of SHANK1 in ASD and elucidate the underlying biological mechanism of core symptoms of ASD. We also provide a reliable model of ASD with core symptoms for future studies, such as biomarker identification and therapeutic intervention studies.
Collapse
|
10
|
Gu L, Luo WY, Xia N, Zhang JN, Fan JK, Yang HM, Wang MC, Zhang H. Upregulated mGluR5 induces ER stress and DNA damage by regulating the NMDA receptor subunit NR2B. J Biochem 2021; 171:349-359. [PMID: 34908130 DOI: 10.1093/jb/mvab140] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 12/06/2021] [Indexed: 11/12/2022] Open
Abstract
Dysfunction caused by mGluR5 expression or activation is an important mechanism in the development of Parkinson's disease (PD). Early clinical studies on mGluR5 negative allosteric modulators have shown some limitations. It is therefore necessary to find a more specific approach to block mGluR5-mediated neurotoxicity. Here, we determined the role of NMDA receptor subunit NR2B in mGluR5-mediated ER stress and DNA damage. In vitro study, rotenone-induced ER stress and DNA damage were accompanied by an increase in mGluR5 expression, and overexpressed or activated mGluR5 with agonist CHPG induced ER stress and DNA damage, while blocking mGluR5 with antagonist MPEP alleviated the effect. Furthermore, the damage caused by CHPG was blocked by NMDA receptor antagonist MK-801. Additionally, rotenone or CHPG increased the p-Src and p-NR2B, which was inhibited by MPEP. Blocking p-Src or NR2B with PP2 or CP101,606 alleviated CHPG-induced ER stress and DNA damage. Overactivation of mGluR5 accompanied with the increase of p-Src and p-NR2B in the ER stress and DNA damage was found in rotenone-induced PD rat model. These findings suggest a new mechanism wherein mGluR5 induces ER stress and DNA damage through the NMDA receptor and propose NR2B as the molecular target for therapeutic strategy for PD.
Collapse
Affiliation(s)
- Li Gu
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Wen-Yuan Luo
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Ning Xia
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute for Brain Disorders, Capital Medical University, Beijing 100069, China.,Department of neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Jian-Nan Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Jing-Kai Fan
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Hui-Min Yang
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Meng-Chen Wang
- School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Hong Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute for Brain Disorders, Capital Medical University, Beijing 100069, China
| |
Collapse
|
11
|
Membrane trafficking and positioning of mGluRs at presynaptic and postsynaptic sites of excitatory synapses. Neuropharmacology 2021; 200:108799. [PMID: 34592242 DOI: 10.1016/j.neuropharm.2021.108799] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 08/31/2021] [Accepted: 09/17/2021] [Indexed: 01/21/2023]
Abstract
The plethora of functions of glutamate in the brain are mediated by the complementary actions of ionotropic and metabotropic glutamate receptors (mGluRs). The ionotropic glutamate receptors carry most of the fast excitatory transmission, while mGluRs modulate transmission on longer timescales by triggering multiple intracellular signaling pathways. As such, mGluRs mediate critical aspects of synaptic transmission and plasticity. Interestingly, at synapses, mGluRs operate at both sides of the cleft, and thus bidirectionally exert the effects of glutamate. At postsynaptic sites, group I mGluRs act to modulate excitability and plasticity. At presynaptic sites, group II and III mGluRs act as auto-receptors, modulating release properties in an activity-dependent manner. Thus, synaptic mGluRs are essential signal integrators that functionally couple presynaptic and postsynaptic mechanisms of transmission and plasticity. Understanding how these receptors reach the membrane and are positioned relative to the presynaptic glutamate release site are therefore important aspects of synapse biology. In this review, we will discuss the currently known mechanisms underlying the trafficking and positioning of mGluRs at and around synapses, and how these mechanisms contribute to synaptic functioning. We will highlight outstanding questions and present an outlook on how recent technological developments will move this exciting research field forward.
Collapse
|
12
|
de Pins B, Mendes T, Giralt A, Girault JA. The Non-receptor Tyrosine Kinase Pyk2 in Brain Function and Neurological and Psychiatric Diseases. Front Synaptic Neurosci 2021; 13:749001. [PMID: 34690733 PMCID: PMC8527176 DOI: 10.3389/fnsyn.2021.749001] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 09/14/2021] [Indexed: 12/28/2022] Open
Abstract
Pyk2 is a non-receptor tyrosine kinase highly enriched in forebrain neurons. Pyk2 is closely related to focal adhesion kinase (FAK), which plays an important role in sensing cell contacts with extracellular matrix and other extracellular signals controlling adhesion and survival. Pyk2 shares some of FAK’s characteristics including recruitment of Src-family kinases after autophosphorylation, scaffolding by interacting with multiple partners, and activation of downstream signaling pathways. Pyk2, however, has the unique property to respond to increases in intracellular free Ca2+, which triggers its autophosphorylation following stimulation of various receptors including glutamate NMDA receptors. Pyk2 is dephosphorylated by the striatal-enriched phosphatase (STEP) that is highly expressed in the same neuronal populations. Pyk2 localization in neurons is dynamic, and altered following stimulation, with post-synaptic and nuclear enrichment. As a signaling protein Pyk2 is involved in multiple pathways resulting in sometimes opposing functions depending on experimental models. Thus Pyk2 has a dual role on neurites and dendritic spines. With Src family kinases Pyk2 participates in postsynaptic regulations including of NMDA receptors and is necessary for specific types of synaptic plasticity and spatial memory tasks. The diverse functions of Pyk2 are also illustrated by its role in pathology. Pyk2 is activated following epileptic seizures or ischemia-reperfusion and may contribute to the consequences of these insults whereas Pyk2 deficit may contribute to the hippocampal phenotype of Huntington’s disease. Pyk2 gene, PTK2B, is associated with the risk for late-onset Alzheimer’s disease. Studies of underlying mechanisms indicate a complex contribution with involvement in amyloid toxicity and tauopathy, combined with possible functional deficits in neurons and contribution in microglia. A role of Pyk2 has also been proposed in stress-induced depression and cocaine addiction. Pyk2 is also important for the mobility of astrocytes and glioblastoma cells. The implication of Pyk2 in various pathological conditions supports its potential interest for therapeutic interventions. This is possible through molecules inhibiting its activity or increasing it through inhibition of STEP or other means, depending on a precise evaluation of the balance between positive and negative consequences of Pyk2 actions.
Collapse
Affiliation(s)
- Benoit de Pins
- Institut du Fer à Moulin, Paris, France.,Inserm UMR-S 1270, Paris, France.,Faculté des Sciences et Ingénierie, Sorbonne Université, Paris, France
| | - Tiago Mendes
- Institut du Fer à Moulin, Paris, France.,Inserm UMR-S 1270, Paris, France.,Faculté des Sciences et Ingénierie, Sorbonne Université, Paris, France
| | - Albert Giralt
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Production and Validation Center of Advanced Therapies (Creatio), Faculty of Medicine and Health Science, University of Barcelona, Barcelona, Spain
| | - Jean-Antoine Girault
- Institut du Fer à Moulin, Paris, France.,Inserm UMR-S 1270, Paris, France.,Faculté des Sciences et Ingénierie, Sorbonne Université, Paris, France
| |
Collapse
|
13
|
Zheng J, Suo L, Zhou Y, Jia L, Li J, Kuang Y, Cui D, Zhang X, Wu Q. Pyk2 suppresses contextual fear memory in an autophosphorylation-independent manner. J Mol Cell Biol 2021; 13:808-821. [PMID: 34529077 PMCID: PMC8782590 DOI: 10.1093/jmcb/mjab057] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 07/19/2021] [Indexed: 11/13/2022] Open
Abstract
Clustered protocadherins (Pcdhs) are a large family of cadherin-like cell adhesion proteins that are central for neurite self-avoidance and neuronal connectivity in the brain. Their downstream non-receptor tyrosine kinase Pyk2 (proline-rich tyrosine kinase 2, also known as Ptk2b, Cakb, Raftk, Fak2, and Cadtk) is predominantly expressed in the hippocampus. We constructed Pyk2 null mouse lines and found that these mutant mice showed enhancement in contextual fear memory, without any change in auditory-cued and spatial-referenced learning and memory. In addition, by preparing Y402F mutant mice, we observed that Pyk2 suppressed contextual fear memory in an autophosphorylation-independent manner. Moreover, using high-throughput RNA sequencing, we found that immediate early genes, such as Npas4, cFos, Zif268/Egr1, Arc, and Nr4a1, were enhanced in Pyk2 null mice. We further showed that Pyk2 disruption affected pyramidal neuronal complexity and spine dynamics. Thus, we demonstrated that Pyk2 is a novel fear memory suppressor molecule and Pyk2 null mice provide a model for understanding fear-related disorders. These findings have interesting implications regarding dysregulation of the Pcdh‒Pyk2 axis in neuropsychiatric disorders.
Collapse
Affiliation(s)
- Jin Zheng
- Center for Comparative Biomedicine, Ministry of Education Key Lab of Systems Biomedicine, State Key Laboratory of Oncogenes and Related Genes, Joint International Research Laboratory of Metabolic and Developmental Sciences, Institute of Systems Biomedicine, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China.,WLA Laboratories, Shanghai, China
| | - Lun Suo
- Center for Comparative Biomedicine, Ministry of Education Key Lab of Systems Biomedicine, State Key Laboratory of Oncogenes and Related Genes, Joint International Research Laboratory of Metabolic and Developmental Sciences, Institute of Systems Biomedicine, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China.,Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Yuxiao Zhou
- Center for Comparative Biomedicine, Ministry of Education Key Lab of Systems Biomedicine, State Key Laboratory of Oncogenes and Related Genes, Joint International Research Laboratory of Metabolic and Developmental Sciences, Institute of Systems Biomedicine, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China.,WLA Laboratories, Shanghai, China
| | - Liling Jia
- Center for Comparative Biomedicine, Ministry of Education Key Lab of Systems Biomedicine, State Key Laboratory of Oncogenes and Related Genes, Joint International Research Laboratory of Metabolic and Developmental Sciences, Institute of Systems Biomedicine, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China.,WLA Laboratories, Shanghai, China
| | - Jingwei Li
- Center for Comparative Biomedicine, Ministry of Education Key Lab of Systems Biomedicine, State Key Laboratory of Oncogenes and Related Genes, Joint International Research Laboratory of Metabolic and Developmental Sciences, Institute of Systems Biomedicine, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China.,WLA Laboratories, Shanghai, China
| | - Yanping Kuang
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Donghong Cui
- Shanghai Mental Health Center, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Xuehong Zhang
- Center for Comparative Biomedicine, Ministry of Education Key Lab of Systems Biomedicine, State Key Laboratory of Oncogenes and Related Genes, Joint International Research Laboratory of Metabolic and Developmental Sciences, Institute of Systems Biomedicine, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Qiang Wu
- Center for Comparative Biomedicine, Ministry of Education Key Lab of Systems Biomedicine, State Key Laboratory of Oncogenes and Related Genes, Joint International Research Laboratory of Metabolic and Developmental Sciences, Institute of Systems Biomedicine, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China.,WLA Laboratories, Shanghai, China
| |
Collapse
|
14
|
Pyk2 in dorsal hippocampus plays a selective role in spatial memory and synaptic plasticity. Sci Rep 2021; 11:16357. [PMID: 34381140 PMCID: PMC8358019 DOI: 10.1038/s41598-021-95813-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Accepted: 07/27/2021] [Indexed: 12/14/2022] Open
Abstract
Pyk2 is a Ca2+-activated non-receptor tyrosine kinase enriched in the forebrain, especially in pyramidal neurons of the hippocampus. Previous reports suggested its role in hippocampal synaptic plasticity and spatial memory but with contradictory findings possibly due to experimental conditions. Here we address this issue and show that novel object location, a simple test of spatial memory induced by a single training session, is altered in Pyk2 KO mice and that re-expression of Pyk2 in the dorsal hippocampus corrects this deficit. Bilateral targeted deletion of Pyk2 in dorsal hippocampus CA1 region also alters novel object location. Long term potentiation (LTP) in CA1 is impaired in Pyk2 KO mice using a high frequency stimulation induction protocol but not with a theta burst protocol, explaining differences between previous reports. The same selective LTP alteration is observed in mice with Pyk2 deletion in dorsal hippocampus CA1 region. Thus, our results establish the role of Pyk2 in specific aspects of spatial memory and synaptic plasticity and show the dependence of the phenotype on the type of experiments used to reveal it. In combination with other studies, we provide evidence for a selective role of non-receptor tyrosine kinases in specific aspects of hippocampal neurons synaptic plasticity.
Collapse
|
15
|
Burada AP, Vinnakota R, Lambolez B, Tricoire L, Kumar J. Structural biology of ionotropic glutamate delta receptors and their crosstalk with metabotropic glutamate receptors. Neuropharmacology 2021; 196:108683. [PMID: 34181979 DOI: 10.1016/j.neuropharm.2021.108683] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 05/31/2021] [Accepted: 06/17/2021] [Indexed: 10/21/2022]
Abstract
Enigmatic orphan glutamate delta receptors (GluD) are one of the four classes of the ionotropic glutamate receptors (iGluRs) that play key roles in synaptic transmission and plasticity. While members of other iGluR families viz AMPA, NMDA, and kainate receptors are gated by glutamate, the GluD receptors neither bind glutamate nor evoke ligand-induced currents upon binding of glycine and D-serine. Thus, the GluD receptors were considered to function as structural proteins that facilitate the formation, maturation, and maintenance of synapses in the hippocampus and cerebellum. Recent work has revealed that GluD receptors have extensive crosstalk with metabotropic glutamate receptors (mGlus) and are also gated by their activation. The latest development of a novel optopharamcological tool and the cryoEM structures of GluD receptors would help define the molecular and chemical basis of the GluD receptor's role in synaptic physiology. This article is part of the special Issue on "Glutamate Receptors - Orphan iGluRs".
Collapse
Affiliation(s)
- Ananth Prasad Burada
- Laboratory of Membrane Protein Biology, National Centre for Cell Science, NCCS Complex, S. P. Pune University, Pune, Maharashtra, 411007, India
| | - Rajesh Vinnakota
- Laboratory of Membrane Protein Biology, National Centre for Cell Science, NCCS Complex, S. P. Pune University, Pune, Maharashtra, 411007, India
| | - Bertrand Lambolez
- Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), CNRS, INSERM, Sorbonne Université, Paris, France
| | - Ludovic Tricoire
- Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), CNRS, INSERM, Sorbonne Université, Paris, France.
| | - Janesh Kumar
- Laboratory of Membrane Protein Biology, National Centre for Cell Science, NCCS Complex, S. P. Pune University, Pune, Maharashtra, 411007, India.
| |
Collapse
|
16
|
Su LD, Wang N, Han J, Shen Y. Group 1 Metabotropic Glutamate Receptors in Neurological and Psychiatric Diseases: Mechanisms and Prospective. Neuroscientist 2021; 28:453-468. [PMID: 34088252 PMCID: PMC9449437 DOI: 10.1177/10738584211021018] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Metabotropic glutamate receptors (mGluRs) are G-protein coupled receptors
that are activated by glutamate in the central nervous system (CNS).
Basically, mGluRs contribute to fine-tuning of synaptic efficacy and
control the accuracy and sharpness of neurotransmission. Among eight
subtypes, mGluR1 and mGluR5 belong to group 1 (Gp1) family, and are
implicated in multiple CNS disorders, such as Alzheimer’s disease,
autism, Parkinson’s disease, and so on. In the present review, we
systematically discussed underlying mechanisms and prospective of Gp1
mGluRs in a group of neurological and psychiatric diseases, including
Alzheimer’s disease, Parkinson’s disease, autism spectrum disorder,
epilepsy, Huntington’s disease, intellectual disability, Down’s
syndrome, Rett syndrome, attention-deficit hyperactivity disorder,
addiction, anxiety, nociception, schizophrenia, and depression, in
order to provide more insights into the therapeutic potential of Gp1
mGluRs.
Collapse
Affiliation(s)
- Li-Da Su
- Neuroscience Care Unit, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Na Wang
- School of Medicine, Zhejiang University City College, Hangzhou, China
| | - Junhai Han
- School of Life Science and Technology, the Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Ying Shen
- Department of Physiology, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
17
|
Rajani V, Sengar AS, Salter MW. Src and Fyn regulation of NMDA receptors in health and disease. Neuropharmacology 2021; 193:108615. [PMID: 34051267 DOI: 10.1016/j.neuropharm.2021.108615] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 05/10/2021] [Accepted: 05/12/2021] [Indexed: 11/28/2022]
Abstract
The Src family kinases (SFKs) are cytoplasmic non-receptor tyrosine kinases involved in multiple signalling pathways. In the central nervous system (CNS), SFKs are key regulators of N-methyl-d-aspartate receptor (NMDAR) function and major points of convergence for neuronal transduction pathways. Physiological upregulation of NMDAR activity by members of the SFKs, namely Src and Fyn, is crucial for induction of plasticity at Schaffer collateral-CA1 synapses of the hippocampus. Aberrant SFK regulation of NMDARs is implicated in several pathological conditions in the CNS including schizophrenia and pain hypersensitivity. Here, evidence is presented to highlight the current understanding of the intermolecular interactions of SFKs within the NMDAR macromolecular complex, the upstream regulators of SFK activity on NMDAR function and the role Src and Fyn have in synaptic plasticity and metaplasticity. The targeting of SFK protein-protein interactions is discussed as a potential therapeutic strategy to restore signalling activity underlying glutamatergic dysregulation in CNS disease pathophysiology.
Collapse
Affiliation(s)
- Vishaal Rajani
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, A1B 3V6, Canada; Program in Neurosciences & Mental Health, Hospital for Sick Children, Toronto, Ontario, M5G 1X8, Canada
| | - Ameet S Sengar
- Program in Neurosciences & Mental Health, Hospital for Sick Children, Toronto, Ontario, M5G 1X8, Canada
| | - Michael W Salter
- Program in Neurosciences & Mental Health, Hospital for Sick Children, Toronto, Ontario, M5G 1X8, Canada; Department of Physiology, University of Toronto, Toronto, Ontario, M5S 1A8, Canada.
| |
Collapse
|
18
|
Alcoreza OB, Patel DC, Tewari BP, Sontheimer H. Dysregulation of Ambient Glutamate and Glutamate Receptors in Epilepsy: An Astrocytic Perspective. Front Neurol 2021; 12:652159. [PMID: 33828523 PMCID: PMC8019783 DOI: 10.3389/fneur.2021.652159] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 02/24/2021] [Indexed: 12/16/2022] Open
Abstract
Given the important functions that glutamate serves in excitatory neurotransmission, understanding the regulation of glutamate in physiological and pathological states is critical to devising novel therapies to treat epilepsy. Exclusive expression of pyruvate carboxylase and glutamine synthetase in astrocytes positions astrocytes as essential regulators of glutamate in the central nervous system (CNS). Additionally, astrocytes can significantly alter the volume of the extracellular space (ECS) in the CNS due to their expression of the bi-directional water channel, aquaporin-4, which are enriched at perivascular endfeet. Rapid ECS shrinkage has been observed following epileptiform activity and can inherently concentrate ions and neurotransmitters including glutamate. This review highlights our emerging knowledge on the various potential contributions of astrocytes to epilepsy, particularly supporting the notion that astrocytes may be involved in seizure initiation via failure of homeostatic responses that lead to increased ambient glutamate. We also review the mechanisms whereby ambient glutamate can influence neuronal excitability, including via generation of the glutamate receptor subunit GluN2B-mediated slow inward currents, as well as indirectly affect neuronal excitability via actions on metabotropic glutamate receptors that can potentiate GluN2B currents and influence neuronal glutamate release probabilities. Additionally, we discuss evidence for upregulation of System x c - , a cystine/glutamate antiporter expressed on astrocytes, in epileptic tissue and changes in expression patterns of glutamate receptors.
Collapse
Affiliation(s)
- Oscar B Alcoreza
- Glial Biology in Health, Disease, and Cancer Center, Fralin Biomedical Research Institute, Virginia Tech Carilion, Roanoke, VA, United States.,School of Medicine, Virginia Tech Carilion, Roanoke, VA, United States.,Translational Biology, Medicine and Health, Virginia Tech, Blacksburg, VA, United States
| | - Dipan C Patel
- Glial Biology in Health, Disease, and Cancer Center, Fralin Biomedical Research Institute, Virginia Tech Carilion, Roanoke, VA, United States
| | - Bhanu P Tewari
- Glial Biology in Health, Disease, and Cancer Center, Fralin Biomedical Research Institute, Virginia Tech Carilion, Roanoke, VA, United States
| | - Harald Sontheimer
- Glial Biology in Health, Disease, and Cancer Center, Fralin Biomedical Research Institute, Virginia Tech Carilion, Roanoke, VA, United States
| |
Collapse
|
19
|
Ibrahim KS, Abd-Elrahman KS, El Mestikawy S, Ferguson SSG. Targeting Vesicular Glutamate Transporter Machinery: Implications on Metabotropic Glutamate Receptor 5 Signaling and Behavior. Mol Pharmacol 2020; 98:314-327. [PMID: 32873747 DOI: 10.1124/molpharm.120.000089] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 07/10/2020] [Indexed: 11/22/2022] Open
Abstract
Cross talk between both pre- and postsynaptic components of glutamatergic neurotransmission plays a crucial role in orchestrating a multitude of brain functions, including synaptic plasticity and motor planning. Metabotropic glutamate receptor (mGluR) 5 exhibits promising therapeutic potential for many neurodevelopmental and neurodegenerative disorders as a consequence of its modulatory control over diverse neuronal networks required for memory, motor coordination, neuronal survival, and differentiation. Given these crucial roles, mGluR5 signaling is under the tight control of glutamate release machinery mediated through vesicular glutamate transporters (VGLUTs) that ultimately dictate glutamatergic output. A particular VGLUT isoform, VGLUT3, exhibits an overlapping, but unique, distribution with mGluR5, and the dynamic cross talk between mGluR5 and VGLUT3 is key for the function of specific neuronal networks involved in motor coordination, emotions, and cognition. Thus, aberrant signaling of the VGLUT3-mGluR5 axis is linked to various pathologies including, but not limited to, Parkinson disease, anxiety disorders, and drug addiction. We argue that a comprehensive profiling of how coordinated VGLUT3-mGluR5 signaling influences overall glutamatergic neurotransmission is warranted. SIGNIFICANCE STATEMENT: Vesicular glutamate receptor (VGLUT) 3 machinery orchestrates glutamate release, and its distribution overlaps with metabotropic glutamate receptor (mGluR) 5 in regional brain circuitries, including striatum, hippocampus, and raphe nucleus. Therefore, VGLUT3-mGluR5 cross talk can significantly influence both physiologic and pathophysiologic glutamatergic neurotransmission. Pathological signaling of the VGLUT3-mGluR5 axis is linked to Parkinson disease, anxiety disorders, and drug addiction. However, it is also predicted to contribute to other motor and cognitive disorders.
Collapse
Affiliation(s)
- Karim S Ibrahim
- University of Ottawa Brain and Mind Institute (K.S.I., K.S.A.-E., S.S.G.F.) and Department of Cellular and Molecular Medicine (K.S.I., K.S.A.-E., S.S.G.F.), University of Ottawa, Ottawa, Ontario, Canada; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt (K.S.I., K.S.A.-E.); Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS-IBPS) INSERM, CNRS, Sorbonne Université, Paris, France (S.E.M.); and Department of Psychiatry, Douglas Hospital Research Center, McGill University, Verdun, Quebec, Canada (S.E.M.)
| | - Khaled S Abd-Elrahman
- University of Ottawa Brain and Mind Institute (K.S.I., K.S.A.-E., S.S.G.F.) and Department of Cellular and Molecular Medicine (K.S.I., K.S.A.-E., S.S.G.F.), University of Ottawa, Ottawa, Ontario, Canada; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt (K.S.I., K.S.A.-E.); Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS-IBPS) INSERM, CNRS, Sorbonne Université, Paris, France (S.E.M.); and Department of Psychiatry, Douglas Hospital Research Center, McGill University, Verdun, Quebec, Canada (S.E.M.)
| | - Salah El Mestikawy
- University of Ottawa Brain and Mind Institute (K.S.I., K.S.A.-E., S.S.G.F.) and Department of Cellular and Molecular Medicine (K.S.I., K.S.A.-E., S.S.G.F.), University of Ottawa, Ottawa, Ontario, Canada; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt (K.S.I., K.S.A.-E.); Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS-IBPS) INSERM, CNRS, Sorbonne Université, Paris, France (S.E.M.); and Department of Psychiatry, Douglas Hospital Research Center, McGill University, Verdun, Quebec, Canada (S.E.M.)
| | - Stephen S G Ferguson
- University of Ottawa Brain and Mind Institute (K.S.I., K.S.A.-E., S.S.G.F.) and Department of Cellular and Molecular Medicine (K.S.I., K.S.A.-E., S.S.G.F.), University of Ottawa, Ottawa, Ontario, Canada; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt (K.S.I., K.S.A.-E.); Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS-IBPS) INSERM, CNRS, Sorbonne Université, Paris, France (S.E.M.); and Department of Psychiatry, Douglas Hospital Research Center, McGill University, Verdun, Quebec, Canada (S.E.M.)
| |
Collapse
|
20
|
Dai WL, Bao YN, Fan JF, Ma B, Li SS, Zhao WL, Yu BY, Liu JH. Blockade of spinal dopamine D1/D2 receptor suppresses activation of NMDA receptor through Gαq and Src kinase to attenuate chronic bone cancer pain. J Adv Res 2020; 28:139-148. [PMID: 33364051 PMCID: PMC7753228 DOI: 10.1016/j.jare.2020.08.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 07/28/2020] [Accepted: 08/10/2020] [Indexed: 12/23/2022] Open
Abstract
Introduction Spinal N-methyl-D-aspartate receptor (NMDAR) is vital in chronic pain, while NMDAR antagonists have severe side effects. NMDAR has been reported to be controlled by G protein coupled receptors (GPCRs), which might present new therapeutic targets to attenuate chronic pain. Dopamine receptors which belong to GPCRs have been reported could modulate the NMDA-mediated currents, while their exact effects on NMDAR in chronic bone cancer pain have not been elucidated. Objectives This study was aim to explore the effects and mechanisms of dopamine D1 receptor (D1DR) and D2 receptor (D2DR) on NMDAR in chronic bone cancer pain. Methods A model for bone cancer pain was established using intra-tibia bone cavity tumor cell implantation (TCI) of Walker 256 in rats. The nociception was assessed by Von Frey assay. A range of techniques including the fluorescent imaging plate reader, western blotting, and immunofluorescence were used to detect cell signaling pathways. Primary cultures of spinal neurons were used for in vitro evaluation. Results Both D1DR and D2DR antagonists decreased NMDA-induced upregulation of Ca2+ oscillations in primary culture spinal neurons. Additionally, D1DR/D2DR antagonists inhibited spinal Calcitonin Gene-Related Peptide (CGRP) and c-Fos expression and alleviated bone cancer pain induced by TCI which could both be reversed by NMDA. And D1DR/D2DR antagonists decreased p-NR1, p-NR2B, and Gαq protein, p-Src expression. Both Gαq protein and Src inhibitors attenuated TCI-induced bone cancer pain, which also be reversed by NMDA. The Gαq protein inhibitor decreased p-Src expression. In addition, D1DR/D2DR antagonists, Src, and Gαq inhibitors inhibited spinal mitogen-activated protein kinase (MAPK) expression in TCI rats, which could be reversed by NMDA. Conclusions Spinal D1DR/D2DR inhibition eliminated NMDAR-mediated spinal neuron activation through Src kinase in a Gαq-protein-dependent manner to attenuate TCI-induced bone cancer pain, which might present a new therapeutic strategy for bone cancer pain.
Collapse
Affiliation(s)
- Wen-Ling Dai
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Yi-Ni Bao
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Ji-Fa Fan
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Bin Ma
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Shan-Shan Li
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Wan-Li Zhao
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Bo-Yang Yu
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China.,State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| | - Ji-Hua Liu
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China.,State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| |
Collapse
|
21
|
Didikoglu A, Maharani A, Canal MM, Pendleton N, Payton A. Interactions between season of birth, chronological age and genetic polymorphisms in determining later-life chronotype. Mech Ageing Dev 2020; 188:111253. [PMID: 32371234 DOI: 10.1016/j.mad.2020.111253] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 03/30/2020] [Accepted: 04/17/2020] [Indexed: 01/19/2023]
Abstract
Human chronotype, the temporal pattern of daily behaviors, is influenced by postnatal seasonal programming and ageing. The aim of this study was to investigate genetic variants that are associated with season of birth programming and longitudinal chronotype change. Longitudinal sleep timing and genotype data from 1449 participants were collected for up to 27 years. Gene-environment interaction analysis was performed for 445 candidate single nucleotide polymorphisms that have previously been associated with chronotype. Associations were tested using linear mixed model. We identified 67 suggestively significant genomic loci that have genotype-ageing interaction and 25 genomic loci that may have genotype-season of birth interaction in determining chronotype. We attempted to replicate the results using longitudinal data of the UK Biobank from approximately 30,000 participants. Biological functions of these genes suggest that epigenetic regulation of gene expression and neural development may have roles in these processes. The strongest associated gene for sleep trajectories was ALKBH5, which has functions of DNA repair and epigenetic regulation. A potential candidate gene for postnatal seasonal programming was SIRT1, which has previously been implicated in postnatal programming, ageing and longevity. Genetic diversity may explain the heterogeneity in ageing-related change of sleep timing and postnatal environmental programming of later-life chronotype.
Collapse
Affiliation(s)
- Altug Didikoglu
- Division of Neuroscience & Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, UK(2).
| | - Asri Maharani
- Division of Nursing, Midwifery & Social Work, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, UK
| | - Maria Mercè Canal
- Division of Neuroscience & Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, UK(2)
| | - Neil Pendleton
- Division of Neuroscience & Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, UK(2)
| | - Antony Payton
- Division of Informatics, Imaging & Data Sciences, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, UK
| |
Collapse
|
22
|
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: 14] [Impact Index Per Article: 2.8] [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
|
23
|
Muscarinic receptor stimulation induces TASK1 channel endocytosis through a PKC-Pyk2-Src pathway in PC12 cells. Cell Signal 2020; 65:109434. [DOI: 10.1016/j.cellsig.2019.109434] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 10/01/2019] [Accepted: 10/01/2019] [Indexed: 11/21/2022]
|
24
|
Dal Prà I, Armato U, Chiarini A. Family C G-Protein-Coupled Receptors in Alzheimer's Disease and Therapeutic Implications. Front Pharmacol 2019; 10:1282. [PMID: 31719824 PMCID: PMC6826475 DOI: 10.3389/fphar.2019.01282] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 10/07/2019] [Indexed: 12/12/2022] Open
Abstract
Alzheimer’s disease (AD), particularly its sporadic or late-onset form (SAD/LOAD), is the most prevalent (96–98% of cases) neurodegenerative dementia in aged people. AD’s neuropathology hallmarks are intrabrain accumulation of amyloid-β peptides (Aβs) and of hyperphosphorylated Tau (p-Tau) proteins, diffuse neuroinflammation, and progressive death of neurons and oligodendrocytes. Mounting evidences suggest that family C G-protein-coupled receptors (GPCRs), which include γ-aminobutyric acid B receptors (GABABRs), metabotropic glutamate receptors (mGluR1-8), and the calcium-sensing receptor (CaSR), are involved in many neurotransmitter systems that dysfunction in AD. This review updates the available knowledge about the roles of GPCRs, particularly but not exclusively those expressed by brain astrocytes, in SAD/LOAD onset and progression, taking stock of their respective mechanisms of action and of their potential as anti-AD therapeutic targets. In particular, GABABRs prevent Aβs synthesis and neuronal hyperexcitability and group I mGluRs play important pathogenetic roles in transgenic AD-model animals. Moreover, the specific binding of Aβs to the CaSRs of human cortical astrocytes and neurons cultured in vitro engenders a pathological signaling that crucially promotes the surplus synthesis and release of Aβs and hyperphosphorylated Tau proteins, and also of nitric oxide, vascular endothelial growth factor-A, and proinflammatory agents. Concurrently, Aβs•CaSR signaling hinders the release of soluble (s)APP-α peptide, a neurotrophic agent and GABABR1a agonist. Altogether these effects progressively kill human cortical neurons in vitro and likely also in vivo. Several CaSR’s negative allosteric modulators suppress all the noxious effects elicited by Aβs•CaSR signaling in human cortical astrocytes and neurons thus safeguarding neurons’ viability in vitro and raising hopes about their potential therapeutic benefits in AD patients. Further basic and clinical investigations on these hot topics are needed taking always heed that activation of the several brain family C GPCRs may elicit divergent upshots according to the models studied.
Collapse
Affiliation(s)
- Ilaria Dal Prà
- Human Histology and Embryology Unit, University of Verona Medical School, Verona, Italy
| | - Ubaldo Armato
- Human Histology and Embryology Unit, University of Verona Medical School, Verona, Italy
| | - Anna Chiarini
- Human Histology and Embryology Unit, University of Verona Medical School, Verona, Italy
| |
Collapse
|
25
|
Dubovyk V, Manahan-Vaughan D. Distinct Time-Course of Alterations of Groups I and II Metabotropic Glutamate Receptor and GABAergic Receptor Expression Along the Dorsoventral Hippocampal Axis in an Animal Model of Psychosis. Front Behav Neurosci 2019; 13:98. [PMID: 31139061 PMCID: PMC6519509 DOI: 10.3389/fnbeh.2019.00098] [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] [Received: 12/21/2018] [Accepted: 04/23/2019] [Indexed: 01/13/2023] Open
Abstract
Psychosis is a clinical state that encompasses a range of abnormal conditions, including distortions in sensory information processing and the resultant delusional thinking, emotional discordance and cognitive impairments. Upon developing this condition, the rate at which cognitive and behavioral deteriorations progress steadily increases suggesting an active contribution of the first psychotic event to the progression of structural and functional abnormalities and disease establishment in diagnosed patients. Changes in GABAergic and glutamatergic function, or expression, in the hippocampus have been proposed as a key factor in the pathophysiology of psychosis. However, little is known as to the time-point of onset of putative changes, to what extent they are progressive, and their relation to disease stabilization. Here, we characterized the expression and distribution patterns of groups I and II metabotropic glutamate (mGlu) receptors and GABA receptors 1 week and 3 months after systemic treatment with an N-methyl-D-aspartate receptor (NMDAR) antagonist (MK801) that is used to model a psychosis-like state in adult rats. We found an early alteration in the expression of mGlu1, mGlu2/3, and GABAB receptors across the hippocampal dorsoventral and transverse axes. This expanded to include an up-regulation of mGlu5 levels across the entire CA1 region and a reduction in GABAB expression, as well as GAD67-positive interneurons particularly in the dorsal hippocampus that appeared 3 months after treatment. Our findings indicate that a reduction of excitability may occur in the hippocampus soon after first-episode psychosis. This changes, over time, into increased excitability. These hippocampus-specific alterations are likely to contribute to the pathophysiology and stabilization of psychosis.
Collapse
Affiliation(s)
- Valentyna Dubovyk
- Department of Neurophysiology, Medical Faculty, Ruhr-University Bochum, Bochum, Germany.,International Graduate School of Neuroscience, Ruhr-University Bochum, Bochum, Germany
| | | |
Collapse
|
26
|
Neyman S, Braunewell KH, O'Connell KE, Dev KK, Manahan-Vaughan D. Inhibition of the Interaction Between Group I Metabotropic Glutamate Receptors and PDZ-Domain Proteins Prevents Hippocampal Long-Term Depression, but Not Long-Term Potentiation. Front Synaptic Neurosci 2019; 11:13. [PMID: 31057390 PMCID: PMC6482240 DOI: 10.3389/fnsyn.2019.00013] [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: 01/21/2019] [Accepted: 04/04/2019] [Indexed: 01/07/2023] Open
Abstract
The group I metabotropic glutamate (mGlu) receptor subtypes, mGlu1 and mGlu5, strongly regulate hippocampal synaptic plasticity. Both harbor PSD-95/discs-large/ZO-1 (PDZ) motifs at their extreme carboxyl terminals, which allow interaction with the PDZ domain of Tamalin, regulate the cell surface expression of group I mGlu receptors, and may modulate their coupling to signaling proteins. We investigated the functional role of this interaction in hippocampal long-term depression (LTD). Acute intracerebral treatment of adult rats with a cell-permeable PDZ-blocking peptide (pep-mGluR-STL), designed to competitively inhibit the interaction between Tamalin and group 1 mGlu receptors, prevented expression of LTD in the hippocampal CA1 region without affecting long-term potentiation (LTP) or basal synaptic transmission. Pep-mGluR-STL prevented facilitation by the group I mGlu receptor agonist, (S)-3,5-Dihydroxyphenylglycine (DHPG), and the mGlu5 agonist, (R,S)-2-chloro-5-Hydroxyphenylglycine (CHPG), of short-term depression (STD) into LTD, suggesting that Tamalin preferentially acts by mediating signaling through mGlu5. These data support that Tamalin is essential for the persistent expression of LTD and that it subserves the effective signaling of group 1 mGlu receptors.
Collapse
Affiliation(s)
- Sergey Neyman
- Department of Neurophysiology, Medical Faculty, Ruhr University Bochum, Bochum, Germany
| | - Karl-Heinz Braunewell
- Department of Neurophysiology, Medical Faculty, Ruhr University Bochum, Bochum, Germany
| | - Kara E O'Connell
- Drug Development, School of Medicine, Faculty of Health Sciences, Trinity College Dublin, Dublin, Ireland
| | - Kumlesh K Dev
- Drug Development, School of Medicine, Faculty of Health Sciences, Trinity College Dublin, Dublin, Ireland
| | | |
Collapse
|
27
|
Dantsuji M, Nakamura S, Nakayama K, Mochizuki A, Park SK, Bae YC, Ozeki M, Inoue T. 5-HT 2A receptor activation enhances NMDA receptor-mediated glutamate responses through Src kinase in the dendrites of rat jaw-closing motoneurons. J Physiol 2019; 597:2565-2589. [PMID: 30919966 DOI: 10.1113/jp275440] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 03/26/2019] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS 5-HT increases the excitability of brainstem and spinal motoneurons, including the jaw-closing motoneurons, by depolarizing the membrane potential and decreasing the medium-duration afterhyperpolarization. In this study, we focused on how 5-HT enhances postsynaptic glutamatergic responses in the dendrites of the jaw-closing motoneurons. We demonstrate that 5-HT augments glutamatergic signalling by enhancing the function of the GluN2A-containing NMDA receptor (NMDAR) through the activation of 5-HT2A receptors (5-HT2A Rs) and Src kinase. To enhance glutamatergic responses, activation of the 5-HT2A Rs must occur within ∼60 μm of the location of the glutamate responses. 5-HT inputs to the jaw-closing motoneurons can significantly vary their input-output relationship, which may contribute to wide-range regulation of contractile forces of the jaw-closing muscles. ABSTRACT Various motor behaviours are modulated by 5-HT. Although the masseter (jaw-closing) motoneurons receive both glutamatergic and serotonergic inputs, it remains unclear how 5-HT affects the glutamatergic inputs to the motoneuronal dendrites. We examined the effects of 5-HT on postsynaptic responses evoked by single- or two-photon uncaging of caged glutamate (glutamate responses) to the dendrites of masseter motoneurons in postnatal day 2-5 rats of either sex. Application of 5-HT induced membrane depolarization and enhanced the glutamate-response amplitude. This enhancement was mimicked by the 5-HT2A receptor (5-HT2A R) agonist and was blocked by the 5-HT2A/2C R antagonist. However, neither the 5-HT2B R nor the 5-HT2C R agonists altered glutamate responses. Blockade of the NMDA receptors (NMDARs), but not AMPA receptors, abolished the 5-HT-induced enhancement. Furthermore, the selective antagonist for the GluN2A subunit abolished the 5-HT-induced enhancement. 5-HT increased GluN2A phosphorylation, while the Src kinase inhibitor reduced the 5-HT-induced enhancement and GluN2A phosphorylation. When exposure to the 5-HT2A R agonist was targeted to the dendrites, the enhancement of glutamate responses was restricted to the loci of the dendrites near the puff loci. Electron microscopic immunohistochemistry revealed that both the NMDARs and the 5-HT2A Rs were close to each other in the same dendrite. These results suggest that activation of dendritic 5-HT2A Rs enhances the function of local GluN2A-containing NMDARs through Src kinase. Such enhancement of the glutamate responses by 5-HT may contribute to wide-range regulation of contractile forces of the jaw-closing muscles.
Collapse
Affiliation(s)
- Masanori Dantsuji
- Department of Oral Physiology, Showa University School of Dentistry, Shinagawa-ku, Tokyo, 142-8555, Japan.,Department of Implant Dentistry, Showa University School of Dentistry, Oota-ku, Tokyo, 145-8515, Japan
| | - Shiro Nakamura
- Department of Oral Physiology, Showa University School of Dentistry, Shinagawa-ku, Tokyo, 142-8555, Japan
| | - Kiyomi Nakayama
- Department of Oral Physiology, Showa University School of Dentistry, Shinagawa-ku, Tokyo, 142-8555, Japan
| | - Ayako Mochizuki
- Department of Oral Physiology, Showa University School of Dentistry, Shinagawa-ku, Tokyo, 142-8555, Japan
| | - Sook Kyung Park
- Department of Oral Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, Daegu, 700-412, Republic of Korea
| | - Yong Chul Bae
- Department of Oral Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, Daegu, 700-412, Republic of Korea
| | - Masahiko Ozeki
- Department of Implant Dentistry, Showa University School of Dentistry, Oota-ku, Tokyo, 145-8515, Japan
| | - Tomio Inoue
- Department of Oral Physiology, Showa University School of Dentistry, Shinagawa-ku, Tokyo, 142-8555, Japan
| |
Collapse
|
28
|
Zoicas I, Kornhuber J. The Role of Metabotropic Glutamate Receptors in Social Behavior in Rodents. Int J Mol Sci 2019; 20:ijms20061412. [PMID: 30897826 PMCID: PMC6470515 DOI: 10.3390/ijms20061412] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 03/15/2019] [Accepted: 03/20/2019] [Indexed: 01/04/2023] Open
Abstract
The appropriate display of social behavior is critical for the well-being and survival of an individual. In many psychiatric disorders, including social anxiety disorder, autism spectrum disorders, depression and schizophrenia social behavior is severely impaired. Selective targeting of metabotropic glutamate receptors (mGluRs) has emerged as a novel treatment strategy for these disorders. In this review, we describe some of the behavioral paradigms used to assess different types of social behavior, such as social interaction, social memory, aggressive behavior and sexual behavior. We then focus on the effects of pharmacological modulation of mGluR1-8 on these types of social behavior. Indeed, accumulating evidence indicates beneficial effects of selective ligands of specific mGluRs in ameliorating innate or pharmacologically-induced deficits in social interaction and social memory as well as in reducing aggression in rodents. We emphasize the importance of future studies investigating the role of selective mGluR ligands on different types of social behavior to provide a better understanding of the neural mechanisms involved which, in turn, might promote the development of selective mGluR-targeted tools for the improved treatment of psychiatric disorders associated with social deficits.
Collapse
Affiliation(s)
- Iulia Zoicas
- Department of Psychiatry and Psychotherapy, University Hospital, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen 91054, Germany.
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, University Hospital, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen 91054, Germany.
| |
Collapse
|
29
|
Mao LM, Wang JQ. Amphetamine-induced Conditioned Place Preference and Changes in mGlu1/5 Receptor Expression and Signaling in the Rat Medial Prefrontal Cortex. Neuroscience 2018; 400:110-119. [PMID: 30599269 DOI: 10.1016/j.neuroscience.2018.12.041] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 11/27/2018] [Accepted: 12/21/2018] [Indexed: 12/27/2022]
Abstract
The medial prefrontal cortex (mPFC) is implicated in the rewarding effect of psychostimulants, although molecular mechanisms underlying the rewarding properties of stimulants in this region are poorly understood. Group I metabotropic glutamate (mGlu) receptors (mGlu1/5 subtypes) are believed to be critical in this event. We thus in this study investigated changes in mGlu1/5 receptor expression and function in the rat mPFC in response to conditioned place preference (CPP) induced by amphetamine. Repeated amphetamine administration (2.5 mg/kg, once daily on alternate days for 10 days) induced reliable CPP. In the mPFC, surface expression of mGlu5 receptors was elevated in rats after amphetamine conditioning. mGlu5 receptors were also increased at synaptic and extrasynaptic sites in amphetamine-conditioned rats. Expression of mGlu1 receptors was stable in surface and synaptic compartments, while it was elevated in the extrasynaptic location. In mPFC neurons, the mGlu1/5 agonist-stimulated phosphoinositide signaling pathway was upregulated in its efficacy following amphetamine conditioning. The mGlu1/5 agonist-stimulated Src kinase phosphorylation was also augmented in rats treated with amphetamine. These results demonstrate the sensitivity of mPFC mGlu1/5 receptors to amphetamine-induced CPP. Amphetamine conditioning results in the upregulation of mGlu1/5 receptor expression at subcellular and/or subsynaptic levels and mGlu1/5-mediated postreceptor signaling in mPFC neurons.
Collapse
Affiliation(s)
- Li-Min Mao
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City, Kansas City, MO 64108, USA
| | - John Q Wang
- Department of Biomedical Sciences, 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
|
30
|
Joffe ME, Centanni SW, Jaramillo AA, Winder DG, Conn PJ. Metabotropic Glutamate Receptors in Alcohol Use Disorder: Physiology, Plasticity, and Promising Pharmacotherapies. ACS Chem Neurosci 2018; 9:2188-2204. [PMID: 29792024 PMCID: PMC6192262 DOI: 10.1021/acschemneuro.8b00200] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Developing efficacious treatments for alcohol use disorder (AUD) has proven difficult. The insidious nature of the disease necessitates a deep understanding of its underlying biology as well as innovative approaches to ameliorate ethanol-related pathophysiology. Excessive ethanol seeking and relapse are generated by long-term changes to membrane properties, synaptic physiology, and plasticity throughout the limbic system and associated brain structures. Each of these factors can be modulated by metabotropic glutamate (mGlu) receptors, a diverse set of G protein-coupled receptors highly expressed throughout the central nervous system. Here, we discuss how different components of the mGlu receptor family modulate neurotransmission in the limbic system and other brain regions involved in AUD etiology. We then describe how these processes are dysregulated following ethanol exposure and speculate about how mGlu receptor modulation might restore such pathophysiological changes. To that end, we detail the current understanding of the behavioral pharmacology of mGlu receptor-directed drug-like molecules in animal models of AUD. Together, this review highlights the prominent position of the mGlu receptor system in the pathophysiology of AUD and provides encouragement that several classes of mGlu receptor modulators may be translated as viable treatment options.
Collapse
Affiliation(s)
- Max E. Joffe
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232-0697, United States
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232-0697, United States
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, Tennessee 37232-0697, United States
| | - Samuel W. Centanni
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, Tennessee 37232-0697, United States
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee 37212, United States
| | - Anel A. Jaramillo
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, Tennessee 37232-0697, United States
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee 37212, United States
| | - Danny G. Winder
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232-0697, United States
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, Tennessee 37232-0697, United States
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee 37212, United States
| | - P. Jeffrey Conn
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232-0697, United States
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232-0697, United States
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, Tennessee 37232-0697, United States
| |
Collapse
|
31
|
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: 46] [Impact Index Per Article: 6.6] [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
|
32
|
Scheefhals N, MacGillavry HD. Functional organization of postsynaptic glutamate receptors. Mol Cell Neurosci 2018; 91:82-94. [PMID: 29777761 PMCID: PMC6276983 DOI: 10.1016/j.mcn.2018.05.002] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 04/16/2018] [Accepted: 05/07/2018] [Indexed: 01/28/2023] Open
Abstract
Glutamate receptors are the most abundant excitatory neurotransmitter receptors in the brain, responsible for mediating the vast majority of excitatory transmission in neuronal networks. The AMPA- and NMDA-type ionotropic glutamate receptors (iGluRs) are ligand-gated ion channels that mediate the fast synaptic responses, while metabotropic glutamate receptors (mGluRs) are coupled to downstream signaling cascades that act on much slower timescales. These functionally distinct receptor sub-types are co-expressed at individual synapses, allowing for the precise temporal modulation of postsynaptic excitability and plasticity. Intriguingly, these receptors are differentially distributed with respect to the presynaptic release site. While iGluRs are enriched in the core of the synapse directly opposing the release site, mGluRs reside preferentially at the border of the synapse. As such, to understand the differential contribution of these receptors to synaptic transmission, it is important to not only consider their signaling properties, but also the mechanisms that control the spatial segregation of these receptor types within synapses. In this review, we will focus on the mechanisms that control the organization of glutamate receptors at the postsynaptic membrane with respect to the release site, and discuss how this organization could regulate synapse physiology.
Collapse
Affiliation(s)
- Nicky Scheefhals
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, 3584 CH Utrecht, The Netherlands
| | - Harold D MacGillavry
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, 3584 CH Utrecht, The Netherlands.
| |
Collapse
|
33
|
Silent Allosteric Modulation of mGluR5 Maintains Glutamate Signaling while Rescuing Alzheimer's Mouse Phenotypes. Cell Rep 2018; 20:76-88. [PMID: 28683325 DOI: 10.1016/j.celrep.2017.06.023] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 06/05/2017] [Accepted: 06/06/2017] [Indexed: 12/22/2022] Open
Abstract
Metabotropic glutamate receptor 5 (mGluR5) has been implicated in Alzheimer's disease (AD) pathology. We sought to understand whether mGluR5's role in AD requires glutamate signaling. We used a potent mGluR5 silent allosteric modulator (SAM, BMS-984923) to separate its well-known physiological role in glutamate signaling from a pathological role in mediating amyloid-β oligomer (Aβo) action. Binding of the SAM to mGluR5 does not change glutamate signaling but strongly reduces mGluR5 interaction with cellular prion protein (PrPC) bound to Aβo. The SAM compound prevents Aβo-induced signal transduction in brain slices and in an AD transgenic mouse model, the APPswe/PS1ΔE9 strain. Critically, 4 weeks of SAM treatment rescues memory deficits and synaptic depletion in the APPswe/PS1ΔE9 transgenic mouse brain. Our data show that mGluR5's role in Aβo-dependent AD phenotypes is separate from its role in glutamate signaling and silent allosteric modulation of mGluR5 has promise as a disease-modifying AD intervention with a broad therapeutic window.
Collapse
|
34
|
Sun Y, Chen Y, Zhan L, Zhang L, Hu J, Gao Z. The role of non-receptor protein tyrosine kinases in the excitotoxicity induced by the overactivation of NMDA receptors. Rev Neurosci 2018; 27:283-9. [PMID: 26540220 DOI: 10.1515/revneuro-2015-0037] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 09/17/2015] [Indexed: 11/15/2022]
Abstract
Protein tyrosine phosphorylation is one of the primary modes of regulation of N-methyl-d-aspartate (NMDA) receptors. The non-receptor tyrosine kinases are one of the two types of protein tyrosine kinases that are involved in this process. The overactivation of NMDA receptors is a primary reason for neuron death following cerebral ischemia. Many studies have illustrated the important role of non-receptor tyrosine kinases in ischemia insults. This review introduces the roles of Src, Fyn, focal adhesion kinase, and proline-rich tyrosine kinase 2 in the excitotoxicity induced by the overactivation of NMDA receptors following cerebral ischemia.
Collapse
|
35
|
Mao LM, Geosling R, Penman B, Wang JQ. Local substrates of non-receptor tyrosine kinases at synaptic sites in neurons. SHENG LI XUE BAO : [ACTA PHYSIOLOGICA SINICA] 2017; 69:657-665. [PMID: 29063113 PMCID: PMC5672811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Several non-receptor tyrosine kinase (nRTK) members are expressed in neurons of mammalian brains. Among these neuron-enriched nRTKs, two Src family kinase members (Src and Fyn) are particularly abundant at synaptic sites and have been most extensively studied for their roles in the regulation of synaptic activity and plasticity. Increasing evidence shows that the synaptic subpool of nRTKs interacts with a number of local substrates, including glutamate receptors (both ionotropic and metabotropic glutamate receptors), postsynaptic scaffold proteins, presynaptic proteins, and synapse-enriched enzymes. By phosphorylating specific tyrosine residues in the intracellular domains of these synaptic proteins either constitutively or in an activity-dependent manner, nRTKs regulate these substrates in trafficking, surface expression, and function. Given the high sensitivity of nRTKs to changing synaptic input, nRTKs are considered to act as a critical regulator in the determination of the strength and efficacy of synaptic transmission.
Collapse
Affiliation(s)
- Li-Min Mao
- Department of Basic Medical Science, School of Medicine, University of Missouri-Kansas City, Kansas City, MO 64108, USA
| | - Ryan Geosling
- Department of Anesthesiology, School of Medicine, University of Missouri-Kansas City, Kansas City, MO 64108, USA
| | - Brian Penman
- Department of Anesthesiology, 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
|
36
|
Brody AH, Strittmatter SM. Synaptotoxic Signaling by Amyloid Beta Oligomers in Alzheimer's Disease Through Prion Protein and mGluR5. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2017; 82:293-323. [PMID: 29413525 PMCID: PMC5835229 DOI: 10.1016/bs.apha.2017.09.007] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Alzheimer's disease (AD) represents an impending global health crisis, yet the complexity of AD pathophysiology has so far precluded the development of any interventions to successfully slow or halt AD progression. It is clear that accumulation of Amyloid-beta (Aβ) peptide triggers progressive synapse loss to cause AD symptoms. Once initiated by Aβ, disease progression is complicated and accelerated by inflammation and by tau pathology. The recognition that Aβ peptide assumes multiple distinct states and that soluble oligomeric species (Aβo) are critical for synaptic damage is central to molecular understanding of AD. This knowledge has led to the identification of specific Aβo receptors, such as cellular prion protein (PrPC), mediating synaptic toxicity and neuronal dysfunction. The identification of PrPC as an Aβo receptor has illuminated an Aβo-induced signaling cascade involving mGluR5, Fyn, and Pyk2 that links Aβ and tau pathologies. This pathway provides novel potential therapeutic targets for disease-modifying AD therapy. Here, we discuss the methods by which several putative Aβo receptors were identified. We also offer an in-depth examination of the known molecular mechanisms believed to mediate Aβo-induced synaptic dysfunction, toxicity, and memory dysfunction.
Collapse
Affiliation(s)
- A Harrison Brody
- Program in Cellular Neuroscience, Neurodegeneration & Repair, Yale University School of Medicine, New Haven, CT, United States; Yale University, New Haven, CT, United States
| | - Stephen M Strittmatter
- Program in Cellular Neuroscience, Neurodegeneration & Repair, Yale University School of Medicine, New Haven, CT, United States; Yale University, New Haven, CT, United States.
| |
Collapse
|
37
|
Oliveira JM, da Cruz e Silva CB, Müller T, Martins TS, Cova M, da Cruz e Silva OAB, Henriques AG. Toward Neuroproteomics in Biological Psychiatry: A Systems Approach Unravels Okadaic Acid-Induced Alterations in the Neuronal Phosphoproteome. ACTA ACUST UNITED AC 2017; 21:550-563. [DOI: 10.1089/omi.2017.0108] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Joana Machado Oliveira
- Neurosciences and Signalling Laboratory, Department of Medical Sciences and Institute of Biomedicine (iBiMED), University of Aveiro, Aveiro, Portugal
| | | | - Thorsten Müller
- Cell Signaling, Biochemistry II—Molecular Biochemistry, Ruhr-University Bochum, Bochum, Germany
| | - Tânia Soares Martins
- Neurosciences and Signalling Laboratory, Department of Medical Sciences and Institute of Biomedicine (iBiMED), University of Aveiro, Aveiro, Portugal
| | - Marta Cova
- Neurosciences and Signalling Laboratory, Department of Medical Sciences and Institute of Biomedicine (iBiMED), University of Aveiro, Aveiro, Portugal
| | - Odete A. B. da Cruz e Silva
- Neurosciences and Signalling Laboratory, Department of Medical Sciences and Institute of Biomedicine (iBiMED), University of Aveiro, Aveiro, Portugal
| | - Ana Gabriela Henriques
- Neurosciences and Signalling Laboratory, Department of Medical Sciences and Institute of Biomedicine (iBiMED), University of Aveiro, Aveiro, Portugal
| |
Collapse
|
38
|
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: 79] [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
|
39
|
Foster DJ, Conn PJ. Allosteric Modulation of GPCRs: New Insights and Potential Utility for Treatment of Schizophrenia and Other CNS Disorders. Neuron 2017; 94:431-446. [PMID: 28472649 PMCID: PMC5482176 DOI: 10.1016/j.neuron.2017.03.016] [Citation(s) in RCA: 185] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 03/02/2017] [Accepted: 03/09/2017] [Indexed: 01/11/2023]
Abstract
G-protein-coupled receptors (GPCRs) play critical roles in regulating brain function. Recent advances have greatly expanded our understanding of these receptors as complex signaling machines that can adopt numerous conformations and modulate multiple downstream signaling pathways. While agonists and antagonists have traditionally been pursued to target GPCRs, allosteric modulators provide several mechanistic advantages, including the ability to distinguish between closely related receptor subtypes. Recently, the discovery of allosteric ligands that confer bias and modulate some, but not all, of a given receptor's downstream signaling pathways can provide pharmacological modulation of brain circuitry with remarkable precision. In addition, allosteric modulators with unprecedented specificity have been developed that can differentiate between subpopulations of a given receptor subtype based on the receptor's dimerization state. These advances are not only providing insight into the biological roles of specific receptor populations, but hold great promise for treating numerous CNS disorders.
Collapse
Affiliation(s)
- Daniel J Foster
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - P Jeffrey Conn
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA.
| |
Collapse
|
40
|
Maubert ME, Wigdahl B, Nonnemacher MR. Opinion: Inhibition of Blood-Brain Barrier Repair as a Mechanism in HIV-1 Disease. Front Neurosci 2017; 11:228. [PMID: 28491017 PMCID: PMC5405129 DOI: 10.3389/fnins.2017.00228] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 04/05/2017] [Indexed: 12/22/2022] Open
Affiliation(s)
- Monique E Maubert
- Department of Microbiology and Immunology, and Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of MedicinePhiladelphia, PA, USA
| | - Brian Wigdahl
- Department of Microbiology and Immunology, and Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of MedicinePhiladelphia, PA, USA.,Sidney Kimmel Cancer Center, Thomas Jefferson UniversityPhiladelphia, PA, USA
| | - Michael R Nonnemacher
- Department of Microbiology and Immunology, and Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of MedicinePhiladelphia, PA, USA
| |
Collapse
|
41
|
Metabotropic glutamate receptors and neurodegenerative diseases. Pharmacol Res 2017; 115:179-191. [DOI: 10.1016/j.phrs.2016.11.013] [Citation(s) in RCA: 145] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 11/11/2016] [Accepted: 11/15/2016] [Indexed: 12/21/2022]
|
42
|
Anguita E, Villalobo A. Src-family tyrosine kinases and the Ca 2+ signal. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1864:915-932. [PMID: 27818271 DOI: 10.1016/j.bbamcr.2016.10.022] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 10/25/2016] [Accepted: 10/30/2016] [Indexed: 01/08/2023]
Abstract
In this review, we shall describe the rich crosstalk between non-receptor Src-family kinases (SFKs) and the Ca2+ transient generated in activated cells by a variety of extracellular and intracellular stimuli, resulting in diverse signaling events. The exchange of information between SFKs and Ca2+ is reciprocal, as it flows in both directions. These kinases are main actors in pathways leading to the generation of the Ca2+ signal, and reciprocally, the Ca2+ signal modulates SFKs activity and functions. We will cover how SFKs participate in the generation of the cytosolic Ca2+ rise upon activation of a series of receptors and the mechanism of clearance of this Ca2+ signal. The role of SFKs modulating Ca2+-translocating channels participating in these events will be amply discussed. Finally, the role of the Ca2+ sensor protein calmodulin on the activity of c-Src, and potentially on other SFKs, will be outlined as well. This article is part of a Special Issue entitled: ECS Meeting edited by Claus Heizmann, Joachim Krebs and Jacques Haiech.
Collapse
Affiliation(s)
- Estefanía Anguita
- Department of Cancer Biology, Instituto de Investigaciones Biomédicas, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, c/ Arturo Duperier 4, E-28029 Madrid, Spain
| | - Antonio Villalobo
- Department of Cancer Biology, Instituto de Investigaciones Biomédicas, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, c/ Arturo Duperier 4, E-28029 Madrid, Spain.
| |
Collapse
|
43
|
Synaptic ERK2 Phosphorylates and Regulates Metabotropic Glutamate Receptor 1 In Vitro and in Neurons. Mol Neurobiol 2016; 54:7156-7170. [PMID: 27796752 DOI: 10.1007/s12035-016-0225-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 10/13/2016] [Indexed: 12/21/2022]
Abstract
A synaptic pool of extracellular signal-regulated kinases (ERK) controls synaptic transmission, although little is known about its underlying signaling mechanisms. Here, we found that synaptic ERK2 directly binds to postsynaptic metabotropic glutamate receptor 1a (mGluR1a). This binding is direct and the ERK-binding site is located in the intracellular C-terminus (CT) of mGluR1a. Parallel with this binding, ERK2 phosphorylates mGluR1a at a cluster of serine residues in the distal part of mGluR1a-CT. In rat cerebellar neurons, ERK2 interacts with mGluR1a at synaptic sites, and active ERK constitutively phosphorylates mGluR1a under normal conditions. This basal phosphorylation is critical for maintaining adequate surface expression of mGluR1a. ERK is also essential for controlling mGluR1a signaling in triggering distinct postreceptor signaling transduction pathways. In summary, we have demonstrated that mGluR1a is a sufficient substrate of ERK2. ERK that interacts with and phosphorylates mGluR1a is involved in the regulation of the trafficking and signaling of mGluR1.
Collapse
|
44
|
Lum JS, Fernandez F, Matosin N, Andrews JL, Huang XF, Ooi L, Newell KA. Neurodevelopmental Expression Profile of Dimeric and Monomeric Group 1 mGluRs: Relevance to Schizophrenia Pathogenesis and Treatment. Sci Rep 2016; 6:34391. [PMID: 27721389 PMCID: PMC5056358 DOI: 10.1038/srep34391] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 09/08/2016] [Indexed: 12/18/2022] Open
Abstract
Group 1 metabotropic glutamate receptors (mGluR1/mGluR5) play an integral role in neurodevelopment and are implicated in psychiatric disorders, such as schizophrenia. mGluR1 and mGluR5 are expressed as homodimers, which is important for their functionality and pharmacology. We examined the protein expression of dimeric and monomeric mGluR1α and mGluR5 in the prefrontal cortex (PFC) and hippocampus throughout development (juvenile/adolescence/adulthood) and in the perinatal phencyclidine (PCP) model of schizophrenia. Under control conditions, mGluR1α dimer expression increased between juvenile and adolescence (209-328%), while monomeric levels remained consistent. Dimeric mGluR5 was steadily expressed across all time points; monomeric mGluR5 was present in juveniles, dramatically declining at adolescence and adulthood (-97-99%). The mGluR regulators, Homer 1b/c and Norbin, significantly increased with age in the PFC and hippocampus. Perinatal PCP treatment significantly increased juvenile dimeric mGluR5 levels in the PFC and hippocampus (37-50%) but decreased hippocampal mGluR1α (-50-56%). Perinatal PCP treatment also reduced mGluR1α dimer levels in the PFC at adulthood (-31%). These results suggest that Group 1 mGluRs have distinct dimeric and monomeric neurodevelopmental patterns, which may impact their pharmacological profiles at specific ages. Perinatal PCP treatment disrupted the early expression of Group 1 mGluRs which may underlie neurodevelopmental alterations observed in this model.
Collapse
Affiliation(s)
- Jeremy S. Lum
- Illawarra Health and Medical Research Institute, Wollongong, New South Wales 2522 Australia
- School of Medicine, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW 2522 Australia
- Schizophrenia Research Institute, Sydney, NSW 2010 Australia
| | - Francesca Fernandez
- Illawarra Health and Medical Research Institute, Wollongong, New South Wales 2522 Australia
- School of Medicine, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW 2522 Australia
- Schizophrenia Research Institute, Sydney, NSW 2010 Australia
- School of Psychology, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Natalie Matosin
- Illawarra Health and Medical Research Institute, Wollongong, New South Wales 2522 Australia
- School of Medicine, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW 2522 Australia
- Schizophrenia Research Institute, Sydney, NSW 2010 Australia
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Kraepelinstrasse 2-10 Munich Germany
| | - Jessica L. Andrews
- Illawarra Health and Medical Research Institute, Wollongong, New South Wales 2522 Australia
- School of Medicine, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW 2522 Australia
- Schizophrenia Research Institute, Sydney, NSW 2010 Australia
| | - Xu-Feng Huang
- Illawarra Health and Medical Research Institute, Wollongong, New South Wales 2522 Australia
- School of Medicine, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW 2522 Australia
- Schizophrenia Research Institute, Sydney, NSW 2010 Australia
| | - Lezanne Ooi
- Illawarra Health and Medical Research Institute, Wollongong, New South Wales 2522 Australia
- School of Biological Sciences, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW 2522 Australia
| | - Kelly A. Newell
- Illawarra Health and Medical Research Institute, Wollongong, New South Wales 2522 Australia
- School of Medicine, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW 2522 Australia
- Schizophrenia Research Institute, Sydney, NSW 2010 Australia
| |
Collapse
|
45
|
Khariv V, Ni L, Ratnayake A, Sampath S, Lutz BM, Tao XX, Heary RF, Elkabes S. Impaired sensitivity to pain stimuli in plasma membrane calcium ATPase 2 (PMCA2) heterozygous mice: a possible modality- and sex-specific role for PMCA2 in nociception. FASEB J 2016; 31:224-237. [PMID: 27702770 DOI: 10.1096/fj.201600541r] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 09/22/2016] [Indexed: 11/11/2022]
Abstract
Plasma membrane calcium ATPase 2 (PMCA2) is a calcium pump that plays important roles in neuronal function. Although it is expressed in pain-associated regions of the CNS, including in the dorsal horn (DH), its contribution to pain remains undefined. The present study assessed the role of PMCA2 in pain responsiveness and the link between PMCA2 and glutamate receptors, GABA receptors (GABARs), and glutamate transporters that have been implicated in pain processing in the DH of adult female and male PMCA2+/+ and PMCA2+/- mice. Behavioral assays evaluated mechanical and thermal pain responsiveness. Mechanical sensitivity was significantly increased by 52% and heat sensitivity was reduced by 29% in female, but not male, PMCA2+/- mice compared with PMCA2+/+ controls. There were female-specific changes in metabotropic glutamate receptor 1, NMDA receptor 2A, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor subunit GluR1, GABABR1, and GABABR2 levels, whereas metabotropic glutamate receptor 5, NMDA receptor 2B, GluR2, and GABAARα2 levels were not altered. Glutamate aspartate transporter levels were higher and glial glutamate transporter 1 levels were lower in the DH of female, but not male, PMCA2+/- mice. These findings indicate a novel role for PMCA2 in modality- and sex-dependent pain responsiveness. Female-specific molecular changes potentially account for the altered pain responses.-Khariv, V., Ni, L., Ratnayake, A., Sampath, S., Lutz, B. M., Tao, X.-X., Heary, R. F., Elkabes, S. Impaired sensitivity to pain stimuli in plasma membrane calcium ATPase 2 (PMCA2) heterozygous mice: a possible modality- and sex-specific role for PMCA2 in nociception.
Collapse
Affiliation(s)
- Veronika Khariv
- Department of Neurological Surgery, The Reynolds Family Spine Laboratory, New Jersey Medical School-Rutgers, The State University of New Jersey, Newark, New Jersey, USA.,Graduate School of Biomedical Sciences, New Jersey Medical School-Rutgers, The State University of New Jersey, Newark, New Jersey, USA
| | - Li Ni
- Department of Neurological Surgery, The Reynolds Family Spine Laboratory, New Jersey Medical School-Rutgers, The State University of New Jersey, Newark, New Jersey, USA
| | - Ayomi Ratnayake
- Department of Neurological Surgery, The Reynolds Family Spine Laboratory, New Jersey Medical School-Rutgers, The State University of New Jersey, Newark, New Jersey, USA
| | - Sujitha Sampath
- Department of Neurological Surgery, The Reynolds Family Spine Laboratory, New Jersey Medical School-Rutgers, The State University of New Jersey, Newark, New Jersey, USA
| | - Brianna M Lutz
- Graduate School of Biomedical Sciences, New Jersey Medical School-Rutgers, The State University of New Jersey, Newark, New Jersey, USA.,Department of Anesthesiology, New Jersey Medical School-Rutgers, The State University of New Jersey, Newark, New Jersey, USA; and
| | - Xuan-Xiang Tao
- Department of Anesthesiology, New Jersey Medical School-Rutgers, The State University of New Jersey, Newark, New Jersey, USA; and
| | - Robert F Heary
- Department of Neurological Surgery, The Reynolds Family Spine Laboratory, New Jersey Medical School-Rutgers, The State University of New Jersey, Newark, New Jersey, USA
| | - Stella Elkabes
- Department of Neurological Surgery, The Reynolds Family Spine Laboratory, New Jersey Medical School-Rutgers, The State University of New Jersey, Newark, New Jersey, USA;
| |
Collapse
|
46
|
Bostrom C, Yau SY, Majaess N, Vetrici M, Gil-Mohapel J, Christie BR. Hippocampal dysfunction and cognitive impairment in Fragile-X Syndrome. Neurosci Biobehav Rev 2016; 68:563-574. [DOI: 10.1016/j.neubiorev.2016.06.033] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 06/21/2016] [Accepted: 06/22/2016] [Indexed: 01/03/2023]
|
47
|
Haas LT, Strittmatter SM. Oligomers of Amyloid β Prevent Physiological Activation of the Cellular Prion Protein-Metabotropic Glutamate Receptor 5 Complex by Glutamate in Alzheimer Disease. J Biol Chem 2016; 291:17112-21. [PMID: 27325698 DOI: 10.1074/jbc.m116.720664] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Indexed: 12/11/2022] Open
Abstract
The dysfunction and loss of synapses in Alzheimer disease are central to dementia symptoms. We have recently demonstrated that pathological Amyloid β oligomer (Aβo) regulates the association between intracellular protein mediators and the synaptic receptor complex composed of cellular prion protein (PrP(C)) and metabotropic glutamate receptor 5 (mGluR5). Here we sought to determine whether Aβo alters the physiological signaling of the PrP(C)-mGluR5 complex upon glutamate activation. We provide evidence that acute exposure to Aβo as well as chronic expression of familial Alzheimer disease mutant transgenes in model mice prevents protein-protein interaction changes of the complex induced by the glutamate analog 3,5-dihydroxyphenylglycine. We further show that 3,5-dihydroxyphenylglycine triggers the phosphorylation and activation of protein-tyrosine kinase 2-β (PTK2B, also referred to as Pyk2) and of calcium/calmodulin-dependent protein kinase II in wild-type brain slices but not in Alzheimer disease transgenic brain slices or wild-type slices incubated with Aβo. This study further distinguishes two separate Aβo-dependent signaling cascades, one dependent on extracellular Ca(2+) and Fyn kinase activation and the other dependent on the release of Ca(2+) from intracellular stores. Thus, Aβo triggers multiple distinct PrP(C)-mGluR5-dependent events implicated in neurodegeneration and dementia. We propose that targeting the PrP(C)-mGluR5 complex will reverse aberrant Aβo-triggered states of the complex to allow physiological fluctuations of glutamate signaling.
Collapse
Affiliation(s)
- Laura T Haas
- From the Cellular Neuroscience, Neurodegeneration, and Repair Program, Departments of Neurology and Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06536 and the Graduate School of Cellular and Molecular Neuroscience, University of Tübingen, 72074 Tübingen, Germany
| | - Stephen M Strittmatter
- From the Cellular Neuroscience, Neurodegeneration, and Repair Program, Departments of Neurology and Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06536 and
| |
Collapse
|
48
|
Li YQ, Tan MS, Wang HF, Tan CC, Zhang W, Zheng ZJ, Kong LL, Wang ZX, Tan L, Jiang T, Tan L, Yu JT. Common variant in PTK2B is associated with late-onset Alzheimer’s disease: A replication study and meta-analyses. Neurosci Lett 2016; 621:83-87. [DOI: 10.1016/j.neulet.2016.04.020] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 03/20/2016] [Accepted: 04/08/2016] [Indexed: 10/22/2022]
|
49
|
Bhattacharyya S. Inside story of Group I Metabotropic Glutamate Receptors (mGluRs). Int J Biochem Cell Biol 2016; 77:205-12. [PMID: 26987586 DOI: 10.1016/j.biocel.2016.03.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 03/10/2016] [Accepted: 03/11/2016] [Indexed: 12/31/2022]
Abstract
Metabotropic glutamate receptors (mGluRs) are G-protein coupled receptors (GPCRs) that are activated by the neurotransmitter glutamate in the central nervous system. Among the eight subtypes, mGluR1 and mGluR5 belong to the group I family. These receptors play important roles in the brain and are believed to be involved in multiple forms of experience dependent synaptic plasticity including learning and memory. In addition, group I mGluRs also have been implicated in various neuropsychiatric disorders like Fragile X syndrome, autism etc. The normal signaling depends on the precise location of these receptors in specific region of the neuron and the process of receptor trafficking plays a crucial role in controlling this localization. Intracellular trafficking could also regulate the desensitization, resensitization, down-regulation and intracellular signaling of these receptors. In this review I focus on the current understanding of group I mGluR regulation in the central nervous system and also their role in neuropsychiatric disorders.
Collapse
Affiliation(s)
- Samarjit Bhattacharyya
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Mohali, Knowledge city, Sector-81, SAS Nagar, PO: 140306, Punjab, India.
| |
Collapse
|
50
|
Src Family Kinases in Brain Edema After Acute Brain Injury. ACTA NEUROCHIRURGICA. SUPPLEMENT 2016; 121:185-90. [PMID: 26463946 DOI: 10.1007/978-3-319-18497-5_33] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Brain edema, the first stage of intracranial hypertension, has been associated with poor prognosis and increased mortality after acute brain injury such as ischemic stroke, intracranial hemorrhage (ICH), and traumatic brain injury (TBI). Acute brain injury often initiates release of many molecules, including glutamate, adenosine, thrombin, oxyhemoglobin, cytokines, reactive oxygen species (ROS), damage-associated molecular pattern molecules (DAMPs), and others. Most of these molecules activate Src family kinases (SFKs), a family of proto-oncogenic non-receptor tyrosine kinases, resulting in blood-brain barrier (BBB) disruption and brain edema at the acute stage after brain injury. However, SFKs also contribute to BBB self-repair and brain edema resolution in the chronic stage that follows brain injury. In this review, we summarize possible pathways through which SFKs are implicated in both brain edema formation and its eventual resolution.
Collapse
|