1
|
Certain N, Gan Q, Bennett J, Hsieh H, Wollmuth LP. Differential regulation of tetramerization of the AMPA receptor glutamate-gated ion channel by auxiliary subunits. J Biol Chem 2023; 299:105227. [PMID: 37673338 PMCID: PMC10558804 DOI: 10.1016/j.jbc.2023.105227] [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: 02/21/2023] [Revised: 08/21/2023] [Accepted: 08/24/2023] [Indexed: 09/08/2023] Open
Abstract
α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) auxiliary subunits are specialized, nontransient binding partners of AMPARs that modulate AMPAR channel gating properties and pharmacology, as well as their biogenesis and trafficking. The most well-characterized families of auxiliary subunits are transmembrane AMPAR regulatory proteins (TARPs), cornichon homologs (CNIHs), and the more recently discovered GSG1-L. These auxiliary subunits can promote or reduce surface expression of AMPARs (composed of GluA1-4 subunits) in neurons, thereby impacting their functional role in membrane signaling. Here, we show that CNIH-2 enhances the tetramerization of WT and mutant AMPARs, presumably by increasing the overall stability of the tetrameric complex, an effect that is mainly mediated by interactions with the transmembrane domain of the receptor. We also find CNIH-2 and CNIH-3 show receptor subunit-specific actions in this regard with CNIH-2 enhancing both GluA1 and GluA2 tetramerization, whereas CNIH-3 only weakly enhances GluA1 tetramerization. These results are consistent with the proposed role of CNIHs as endoplasmic reticulum cargo transporters for AMPARs. In contrast, TARP γ-2, TARP γ-8, and GSG1-L have no or negligible effect on AMPAR tetramerization. On the other hand, TARP γ-2 can enhance receptor tetramerization but only when directly fused with the receptor at a maximal stoichiometry. Notably, surface expression of functional AMPARs was enhanced by CNIH-2 to a greater extent than TARP γ-2, suggesting that this distinction aids in maturation and membrane expression. These experiments define a functional distinction between CNIHs and other auxiliary subunits in the regulation of AMPAR biogenesis.
Collapse
Affiliation(s)
- Noele Certain
- Graduate Program in Molecular and Cellular Pharmacology, Stony Brook University, Stony Brook, New York, USA
| | - Quan Gan
- Graduate Program in Neuroscience, Stony Brook University, Stony Brook, New York, USA
| | - Joseph Bennett
- Department of Neurobiology & Behavior, Stony Brook University, Stony Brook, New York, USA
| | - Helen Hsieh
- Department of Surgery, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York, USA; Center for Nervous System Disorders, Stony Brook University, Stony Brook, New York, USA
| | - Lonnie P Wollmuth
- Department of Neurobiology & Behavior, Stony Brook University, Stony Brook, New York, USA; Center for Nervous System Disorders, Stony Brook University, Stony Brook, New York, USA; Department of Biochemistry & Cell Biology, Stony Brook University, Stony Brook, New York, USA.
| |
Collapse
|
2
|
Larsen ME, Buonarati OR, Qian H, Hell JW, Bayer KU. Stimulating β-adrenergic receptors promotes synaptic potentiation by switching CaMKII movement from LTD to LTP mode. J Biol Chem 2023; 299:104706. [PMID: 37061000 PMCID: PMC10200978 DOI: 10.1016/j.jbc.2023.104706] [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: 09/21/2022] [Revised: 03/31/2023] [Accepted: 04/06/2023] [Indexed: 04/17/2023] Open
Abstract
Learning, memory, and cognition are thought to require synaptic plasticity, specifically including hippocampal long-term potentiation and depression (LTP and LTD). LTP versus LTD is induced by high-frequency stimulation versus low-frequency, but stimulating β-adrenergic receptors (βARs) enables LTP induction also by low-frequency stimulation (1 Hz) or theta frequencies (∼5 Hz) that do not cause plasticity by themselves. In contrast to high-frequency stimulation-LTP, such βAR-LTP requires Ca2+-flux through L-type voltage-gated Ca2+-channels, not N-methyl-D-aspartate-type glutamate receptors. Surprisingly, we found that βAR-LTP still required a nonionotropic scaffolding function of the N-methyl-D-aspartate-type glutamate receptor: the stimulus-induced binding of the Ca2+/calmodulin-dependent protein kinase II (CaMKII) to its GluN2B subunit that mediates CaMKII movement to excitatory synapses. In hippocampal neurons, β-adrenergic stimulation with isoproterenol (Iso) transformed LTD-type CaMKII movement to LTP-type movement, resulting in CaMKII movement to excitatory instead of inhibitory synapses. Additionally, Iso enabled induction of a major cell-biological feature of LTP in response to LTD stimuli: increased surface expression of GluA1 fused with super-ecliptic pHluorein. Like for βAR-LTP in hippocampal slices, the Iso effects on CaMKII movement and surface expression of GluA1 fused with super-ecliptic pHluorein involved L-type Ca2+-channels and specifically required β2-ARs. Taken together, these results indicate that Iso transforms LTD stimuli to LTP signals by switching CaMKII movement and GluN2B binding to LTP mode.
Collapse
Affiliation(s)
- Matthew E Larsen
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Program in Neuroscience, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Olivia R Buonarati
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Hai Qian
- Department of Pharmacology, University of Iowa, Iowa City, Iowa, USA
| | - Johannes W Hell
- Department of Pharmacology, University of Iowa, Iowa City, Iowa, USA; Department of Pharmacology, University of California at Davis, Davis, California, USA.
| | - K Ulrich Bayer
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Program in Neuroscience, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.
| |
Collapse
|
3
|
Rollenhagen A, Anstötz M, Zimmermann K, Kasugai Y, Sätzler K, Molnar E, Ferraguti F, Lübke JHR. Layer-specific distribution and expression pattern of AMPA- and NMDA-type glutamate receptors in the barrel field of the adult rat somatosensory cortex: a quantitative electron microscopic analysis. Cereb Cortex 2023; 33:2342-2360. [PMID: 35732315 PMCID: PMC9977369 DOI: 10.1093/cercor/bhac212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 05/05/2022] [Accepted: 05/06/2022] [Indexed: 11/14/2022] Open
Abstract
AMPA (α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid) and NMDA (N-methyl-d-aspartate) glutamate receptors are driving forces for synaptic transmission and plasticity at neocortical synapses. However, their distribution pattern in the adult rat neocortex is largely unknown and was quantified using freeze fracture replication combined with postimmunogold-labeling. Both receptors were co-localized at layer (L)4 and L5 postsynaptic densities (PSDs). At L4 dendritic shaft and spine PSDs, the number of gold grains detecting AMPA was similar, whereas at L5 shaft PSDs AMPA-receptors outnumbered those on spine PSDs. Their number was significantly higher at L5 vs. L4 PSDs. At L4 and L5 dendritic shaft PSDs, the number of gold grains detecting GluN1 was ~2-fold higher than at spine PSDs. The number of gold grains detecting the GluN1-subunit was higher for both shaft and spine PSDs in L5 vs. L4. Both receptors showed a large variability in L4 and L5. A high correlation between the number of gold grains and PSD size for both receptors and targets was observed. Both receptors were distributed over the entire PSD but showed a layer- and target-specific distribution pattern. The layer- and target-specific distribution of AMPA and GluN1 glutamate receptors partially contribute to the observed functional differences in synaptic transmission and plasticity in the neocortex.
Collapse
Affiliation(s)
- Astrid Rollenhagen
- Institute of Neuroscience and Medicine INM-10, Research Centre Jülich GmbH, Leo Brandt Str., Jülich 52425, Germany
| | - Max Anstötz
- Institute of Neuroscience and Medicine INM-10, Research Centre Jülich GmbH, Leo Brandt Str., Jülich 52425, Germany.,Institute of Anatomy II, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University, Universitätsstr. 1, Düsseldorf 40001, Germany
| | - Kerstin Zimmermann
- Institute of Neuroscience and Medicine INM-10, Research Centre Jülich GmbH, Leo Brandt Str., Jülich 52425, Germany
| | - Yu Kasugai
- Department of Pharmacology, Medical University of Innsbruck, Peter Mayr Strasse 1a, Innsbruck A-6020, Austria
| | - Kurt Sätzler
- School of Biomedical Sciences, University of Ulster, Cromore Rd., Londonderry BT52 1SA, United Kingdom
| | - Elek Molnar
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, University Walk, Bristol BS8 1TD, United Kingdom
| | - Francesco Ferraguti
- Department of Pharmacology, Medical University of Innsbruck, Peter Mayr Strasse 1a, Innsbruck A-6020, Austria
| | - Joachim H R Lübke
- Institute of Neuroscience and Medicine INM-10, Research Centre Jülich GmbH, Leo Brandt Str., Jülich 52425, Germany.,Department of Psychiatry, Psychotherapy and Psychosomatics, RWTH/Medical University Aachen, Pauwelstr. 30, Aachen 52074, Germany.,JARA Translational Medicine Jülich/Aachen, Germany
| |
Collapse
|
4
|
Certain N, Gan Q, Bennett J, Hsieh H, Wollmuth LP. Differential regulation of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid (AMPA) receptor tetramerization by auxiliary subunits. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.07.527516. [PMID: 36798164 PMCID: PMC9934675 DOI: 10.1101/2023.02.07.527516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
AMPA receptor (AMPAR) auxiliary subunits are specialized, non-transient binding partners of AMPARs that modulate their ion channel gating properties and pharmacology, as well as their biogenesis and trafficking. The most well characterized families of auxiliary subunits are transmembrane AMPAR regulatory proteins (TARPs) and cornichon homologs (CNIHs) and the more recently discovered GSG1-L. These auxiliary subunits can promote or reduce surface expression of AMPARs in neurons, thereby impacting their functional role in membrane signaling. Here, we show that CNIH-2 enhances the tetramerization of wild type and mutant AMPARs, possibly by increasing the overall stability of the tetrameric complex, an effect that is mainly mediated by interactions with the transmembrane domain of the receptor. We also find CNIH-2 and CNIH-3 show receptor subunit-specific actions in this regard with CNIH-2 enhancing both GluA1 and GluA2 tetramerization whereas CNIH-3 only weakly enhances GluA1 tetramerization. These results are consistent with the proposed role of CNIHs as endoplasmic reticulum cargo transporters for AMPARs. In contrast, TARP γ-2, TARP γ-8, and GSG1-L have no or negligible effect on AMPAR tetramerization. On the other hand, TARP γ-2 can enhance receptor tetramerization but only when directly fused with the receptor at a maximal stoichiometry. Notably, surface expression of functional AMPARs was enhanced by CNIH-2 to a greater extent than TARP γ-2 suggesting that this distinction aids in maturation and membrane expression. These experiments define a functional distinction between CNIHs and other auxiliary subunits in the regulation of AMPAR biogenesis.
Collapse
|
5
|
Chen L, Wang H, Cha S, Li J, Zhang J, Wu J, Guo G, Zhang J. Phosphorylation of Spastin Promotes the Surface Delivery and Synaptic Function of AMPA Receptors. Front Cell Neurosci 2022; 16:809934. [PMID: 35418834 PMCID: PMC8995424 DOI: 10.3389/fncel.2022.809934] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 03/07/2022] [Indexed: 12/14/2022] Open
Abstract
Synaptic plasticity is essential for cognitive functions such as learning and memory. One of the mechanisms involved in synaptic plasticity is the dynamic delivery of AMPA receptors (AMPARs) in and out of synapses. Mutations of SPAST, which encodes SPASTIN, a microtubule-severing protein, are considered the most common cause of hereditary spastic paraparesis (HSP). In some cases, patients with HSP also manifest cognitive impairment. In addition, mice with Spastin depletion exhibit working and associative memory deficits and reduced AMPAR levels. However, the exact effect and molecular mechanism of Spastin on AMPARs trafficking has remained unclear. Here, we report that Spastin interacts with AMPAR, and phosphorylation of Spastin enhances its interaction with AMPAR subunit GluA2. Further study shows that phosphorylation of Spastin can increase AMPAR GluA2 surface expression and the amplitude and frequency of miniature excitatory synaptic currents (mEPSC) in cultured hippocampal neurons. Moreover, phosphorylation of Spastin at Ser210 is crucial for GluA2 surface expression. Phosphorylation of Spastin K353A, which obliterates microtubule-severing activity, also promotes AMPAR GluA2 subunit trafficking to the surface and increases the amplitude and frequency of mEPSCs in cultured neurons. Taken together, our data demonstrate that Spastin phosphorylation promotes the surface delivery of the AMPAR GluA2 subunit independent of microtubule dynamics.
Collapse
Affiliation(s)
- Li Chen
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou, China
| | - Hanjie Wang
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou, China
| | - Shuhan Cha
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou, China
| | - Jiong Li
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou, China
| | - Jiaqi Zhang
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou, China
| | - Jiaming Wu
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou, China
- Department of Neurosurgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Guoqing Guo
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou, China
- *Correspondence: Guoqing Guo Jifeng Zhang
| | - Jifeng Zhang
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou, China
- *Correspondence: Guoqing Guo Jifeng Zhang
| |
Collapse
|
6
|
Royo M, Escolano BA, Madrigal MP, Jurado S. AMPA Receptor Function in Hypothalamic Synapses. Front Synaptic Neurosci 2022; 14:833449. [PMID: 35173598 PMCID: PMC8842481 DOI: 10.3389/fnsyn.2022.833449] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 01/03/2022] [Indexed: 12/15/2022] Open
Abstract
AMPA receptors (AMPARs) are critical for mediating glutamatergic synaptic transmission and plasticity, thus playing a major role in the molecular machinery underlying cellular substrates of memory and learning. Their expression pattern, transport and regulatory mechanisms have been extensively studied in the hippocampus, but their functional properties in other brain regions remain poorly understood. Interestingly, electrophysiological and molecular evidence has confirmed a prominent role of AMPARs in the regulation of hypothalamic function. This review summarizes the existing evidence on AMPAR-mediated transmission in the hypothalamus, where they are believed to orchestrate the role of glutamatergic transmission in autonomous, neuroendocrine function, body homeostasis, and social behavior.
Collapse
|
7
|
Stolz JR, Foote KM, Veenstra-Knol HE, Pfundt R, Ten Broeke SW, de Leeuw N, Roht L, Pajusalu S, Part R, Rebane I, Õunap K, Stark Z, Kirk EP, Lawson JA, Lunke S, Christodoulou J, Louie RJ, Rogers RC, Davis JM, Innes AM, Wei XC, Keren B, Mignot C, Lebel RR, Sperber SM, Sakonju A, Dosa N, Barge-Schaapveld DQCM, Peeters-Scholte CMPCD, Ruivenkamp CAL, van Bon BW, Kennedy J, Low KJ, Ellard S, Pang L, Junewick JJ, Mark PR, Carvill GL, Swanson GT. Clustered mutations in the GRIK2 kainate receptor subunit gene underlie diverse neurodevelopmental disorders. Am J Hum Genet 2021; 108:1692-1709. [PMID: 34375587 PMCID: PMC8456161 DOI: 10.1016/j.ajhg.2021.07.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 07/15/2021] [Indexed: 12/14/2022] Open
Abstract
Kainate receptors (KARs) are glutamate-gated cation channels with diverse roles in the central nervous system. Bi-allelic loss of function of the KAR-encoding gene GRIK2 causes a nonsyndromic neurodevelopmental disorder (NDD) with intellectual disability and developmental delay as core features. The extent to which mono-allelic variants in GRIK2 also underlie NDDs is less understood because only a single individual has been reported previously. Here, we describe an additional eleven individuals with heterozygous de novo variants in GRIK2 causative for neurodevelopmental deficits that include intellectual disability. Five children harbored recurrent de novo variants (three encoding p.Thr660Lys and two p.Thr660Arg), and four children and one adult were homozygous for a previously reported variant (c.1969G>A [p.Ala657Thr]). Individuals with shared variants had some overlapping behavioral and neurological dysfunction, suggesting that the GRIK2 variants are likely pathogenic. Analogous mutations introduced into recombinant GluK2 KAR subunits at sites within the M3 transmembrane domain (encoding p.Ala657Thr, p.Thr660Lys, and p.Thr660Arg) and the M3-S2 linker domain (encoding p.Ile668Thr) had complex effects on functional properties and membrane localization of homomeric and heteromeric KARs. Both p.Thr660Lys and p.Thr660Arg mutant KARs exhibited markedly slowed gating kinetics, similar to p.Ala657Thr-containing receptors. Moreover, we observed emerging genotype-phenotype correlations, including the presence of severe epilepsy in individuals with the p.Thr660Lys variant and hypomyelination in individuals with either the p.Thr660Lys or p.Thr660Arg variant. Collectively, these results demonstrate that human GRIK2 variants predicted to alter channel function are causative for early childhood development disorders and further emphasize the importance of clarifying the role of KARs in early nervous system development.
Collapse
Affiliation(s)
- Jacob R Stolz
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Kendall M Foote
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Hermine E Veenstra-Knol
- Department of Genetics, University Medical Center Groningen, Groningen 9700, the Netherlands
| | - Rolph Pfundt
- Department of Human Genetics, Radboud University Nijmegen Medical Centre, Nijmegen 6525, the Netherlands
| | - Sanne W Ten Broeke
- Department of Genetics, University Medical Center Groningen, Groningen 9700, the Netherlands
| | - Nicole de Leeuw
- Department of Human Genetics, Radboud University Nijmegen Medical Centre, Nijmegen 6525, the Netherlands
| | - Laura Roht
- Department of Clinical Genetics, Tartu University Hospital, Tartu 50406, Estonia; Department of Clinical Genetics, Institute of Clinical Medicine, Tartu University, Tartu 51003, Estonia
| | - Sander Pajusalu
- Department of Clinical Genetics, Tartu University Hospital, Tartu 50406, Estonia; Department of Clinical Genetics, Institute of Clinical Medicine, Tartu University, Tartu 51003, Estonia
| | - Reelika Part
- Department of Neonatal and Infant Medicine, Tallinn Children's Hospital, Tallinn 13419, Estonia
| | - Ionella Rebane
- Department of Neonatal and Infant Medicine, Tallinn Children's Hospital, Tallinn 13419, Estonia
| | - Katrin Õunap
- Department of Clinical Genetics, Tartu University Hospital, Tartu 50406, Estonia; Department of Clinical Genetics, Institute of Clinical Medicine, Tartu University, Tartu 51003, Estonia
| | - Zornitza Stark
- Murdoch Children's Research Institute, Melbourne, VIC 3052, Australia; Australian Genomics Health Alliance, Melbourne, VIC 3052, Australia; Department of Paediatrics, University of Melbourne, Melbourne, VIC 3052, Australia
| | - Edwin P Kirk
- School of Women's and Children's Health, UNSW Medicine, University of New South Wales, Randwick, NSW 2031, Australia; Centre for Clinical Genetics, Sydney Children's Hospital, Randwick, NSW 2031, Australia
| | - John A Lawson
- Department of Neurology, Sydney Children's Hospital, Randwick, NSW 2031, Australia
| | - Sebastian Lunke
- Murdoch Children's Research Institute, Melbourne, VIC 3052, Australia; Australian Genomics Health Alliance, Melbourne, VIC 3052, Australia
| | - John Christodoulou
- Murdoch Children's Research Institute, Melbourne, VIC 3052, Australia; Australian Genomics Health Alliance, Melbourne, VIC 3052, Australia; Department of Paediatrics, University of Melbourne, Melbourne, VIC 3052, Australia
| | | | | | | | - A Micheil Innes
- Departments of Medical Genetics and Pediatrics, Cumming School of Medicine, University of Calgary, Alberta T2N 4N1, Canada
| | - Xing-Chang Wei
- Department of Diagnostic Imaging, Cumming School of Medicine, University of Calgary, AB T2N 4N1, Canada
| | - Boris Keren
- Département de Génétique, Hôpital Pitié-Salpêtrière, Paris 75013, France
| | - Cyril Mignot
- Département de Génétique, Hôpital Pitié-Salpêtrière, Paris 75013, France
| | - Robert Roger Lebel
- Division of Development, Behavior, and Genetics, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Steven M Sperber
- Department of Pathology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Ai Sakonju
- Department of Neurology, Upstate Health Care Center, Syracuse, NY 13210, USA
| | - Nienke Dosa
- Division of Development, Behavior, and Genetics, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | | | | | - Claudia A L Ruivenkamp
- Department of Clinical Genetics, Leiden University Medical Center, 2333 Leiden, the Netherlands
| | - Bregje W van Bon
- Department of Human Genetics, Radboud University Nijmegen Medical Centre, Nijmegen 6525, the Netherlands
| | - Joanna Kennedy
- University Hospital Bristol, NHS Foundation Trust, Bristol BS1 3NU, UK
| | - Karen J Low
- University Hospital Bristol, NHS Foundation Trust, Bristol BS1 3NU, UK
| | - Sian Ellard
- Exeter Genomics Laboratory, Royal Devon and Exeter NHS Foundation Trust, Exeter EX2 5DW, UK
| | - Lewis Pang
- Exeter Genomics Laboratory, Royal Devon and Exeter NHS Foundation Trust, Exeter EX2 5DW, UK
| | - Joseph J Junewick
- Department of Radiology, Helen DeVos Children's Hospital, Grand Rapids, MI 49503, USA
| | - Paul R Mark
- Spectrum Health Medical Genetics, Grand Rapids, MI 49503, USA
| | - Gemma L Carvill
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Geoffrey T Swanson
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
8
|
Wu QL, Gao Y, Li JT, Ma WY, Chen NH. The Role of AMPARs Composition and Trafficking in Synaptic Plasticity and Diseases. Cell Mol Neurobiol 2021; 42:2489-2504. [PMID: 34436728 DOI: 10.1007/s10571-021-01141-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 08/11/2021] [Indexed: 11/28/2022]
Abstract
AMPA receptors are tetrameric ionic glutamate receptors, which mediate 90% fast excitatory synaptic transmission induced by excitatory glutamate in the mammalian central nervous system through the activation or inactivation of ion channels. The alternation of synaptic AMPA receptor number and subtype is thought to be one of the primary mechanisms that involve in synaptic plasticity regulation and affect the functions in learning, memory, and cognition. The increasing of surface AMPARs enhances synaptic strength during long-term potentiation, whereas the decreasing of AMPARs weakens synaptic strength during the long-term depression. It is closely related to the AMPA receptor as well as its subunits assembly, trafficking, and degradation. The dysfunction of any step in these precise regulatory processes is likely to induce the disorder of synaptic transmission and loss of neurons, or even cause neuropsychiatric diseases ultimately. Therefore, it is useful to understand how AMPARs regulate synaptic plasticity and its role in related neuropsychiatric diseases via comprehending architecture and trafficking of the receptors. Here, we reviewed the progress in structure, expression, trafficking, and relationship with synaptic plasticity of AMPA receptor, especially in anxiety, depression, neurodegenerative disorders, and cerebral ischemia.
Collapse
Affiliation(s)
- Qing-Lin Wu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.,State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Yan Gao
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Jun-Tong Li
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.,State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Wen-Yu Ma
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.,State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Nai-Hong Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China. .,State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| |
Collapse
|
9
|
Gutiérrez Y, López-García S, Lario A, Gutiérrez-Eisman S, Delevoye C, Esteban JA. KIF13A drives AMPA receptor synaptic delivery for long-term potentiation via endosomal remodeling. J Cell Biol 2021; 220:212112. [PMID: 33999113 PMCID: PMC8129809 DOI: 10.1083/jcb.202003183] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 02/16/2021] [Accepted: 03/31/2021] [Indexed: 02/06/2023] Open
Abstract
The regulated trafficking of AMPA-type glutamate receptors (AMPARs) from dendritic compartments to the synaptic membrane in response to neuronal activity is a core mechanism for long-term potentiation (LTP). However, the contribution of the microtubule cytoskeleton to this synaptic transport is still unknown. In this work, using electrophysiological, biochemical, and imaging techniques, we have found that one member of the kinesin-3 family of motor proteins, KIF13A, is specifically required for the delivery of AMPARs to the spine surface during LTP induction. Accordingly, KIF13A depletion from hippocampal slices abolishes LTP expression. We also identify the vesicular protein centaurin-α1 as part of a motor transport machinery that is engaged with KIF13A and AMPARs upon LTP induction. Finally, we determine that KIF13A is responsible for the remodeling of Rab11-FIP2 endosomal structures in the dendritic shaft during LTP. Overall, these results identify specific kinesin molecular motors and endosomal transport machinery that catalyzes the dendrite-to-synapse translocation of AMPA receptors during synaptic plasticity.
Collapse
Affiliation(s)
- Yolanda Gutiérrez
- Molecular Neuropathology Unit, Centro de Biología Molecular Severo Ochoa (Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid), Madrid, Spain
| | - Sergio López-García
- Molecular Neuropathology Unit, Centro de Biología Molecular Severo Ochoa (Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid), Madrid, Spain
| | - Argentina Lario
- Molecular Neuropathology Unit, Centro de Biología Molecular Severo Ochoa (Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid), Madrid, Spain
| | - Silvia Gutiérrez-Eisman
- Molecular Neuropathology Unit, Centro de Biología Molecular Severo Ochoa (Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid), Madrid, Spain
| | - Cédric Delevoye
- Institut Curie, Paris Sciences et Lettres Research University, Centre National de la Recherche Scientifique, UMR144, Structure and Membrane Compartments, Paris, France.,Institut Curie, Paris Sciences et Lettres Research University, Centre National de la Recherche Scientifique, UMR144, Cell and Tissue Imaging Facility, Paris, France
| | - José A Esteban
- Molecular Neuropathology Unit, Centro de Biología Molecular Severo Ochoa (Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid), Madrid, Spain
| |
Collapse
|
10
|
Inhibition of AKT/GSK3β/CREB Pathway Improves the Responsiveness to AMPA Receptor Antagonists by Regulating GRIA1 Surface Expression in Chronic Epilepsy Rats. Biomedicines 2021; 9:biomedicines9040425. [PMID: 33919872 PMCID: PMC8103519 DOI: 10.3390/biomedicines9040425] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 04/10/2021] [Accepted: 04/13/2021] [Indexed: 12/15/2022] Open
Abstract
α-Amino-3-hydroxy-5-methylisoxazole-4-propionic acid receptor (AMPAR) has been reported as one of the targets for treatment of epilepsy. Although maladaptive regulation of surface expression of glutamate ionotropic receptor AMPA type subunit 1 (GRIA1) subunit is relevant to the responsiveness to AMPAR antagonists (perampanel and GYKI 52466) in LiCl-pilocarpine-induced chronic epilepsy rats, the underlying mechanisms of refractory seizures to AMPAR antagonists have yet been unclear. In the present study, we found that both AMPAR antagonists restored the up-regulations of GRIA1 surface expression and Src family-mediated glycogen synthase kinase 3β (GSK3β)-Ca2+/cAMP response element-binding protein (CREB) phosphorylations to control levels in responders (whose seizure activities were responsive to AMPAR) but not non-responders (whose seizure activities were uncontrolled by AMPAR antagonists). In addition, 3-chloroacetyl indole (3CAI, an AKT inhibitor) co-treatment attenuated spontaneous seizure activities in non-responders, accompanied by reductions in AKT/GSK3β/CREB phosphorylations and GRIA1 surface expression. Although AMPAR antagonists reduced GRIA2 tyrosine (Y) phosphorylations in responders, they did not affect GRIA2 surface expression and protein interacting with C kinase 1 (PICK1) protein level in both responders and non-responders. Therefore, our findings suggest that dysregulation of AKT/GSK3β/CREB-mediated GRIA1 surface expression may be responsible for refractory seizures in non-responders, and that this pathway may be a potential target to improve the responsiveness to AMPAR antagonists.
Collapse
|
11
|
Cook SG, Buonarati OR, Coultrap SJ, Bayer KU. CaMKII holoenzyme mechanisms that govern the LTP versus LTD decision. SCIENCE ADVANCES 2021; 7:7/16/eabe2300. [PMID: 33853773 PMCID: PMC8046365 DOI: 10.1126/sciadv.abe2300] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 02/24/2021] [Indexed: 05/30/2023]
Abstract
Higher brain functions are thought to require synaptic frequency decoding that can lead to long-term potentiation (LTP) or depression (LTD). We show that the LTP versus LTD decision is determined by complex cross-regulation of T286 and T305/306 autophosphorylation within the 12meric CaMKII holoenzyme, which enabled molecular computation of stimulus frequency, amplitude, and duration. Both LTP and LTD require T286 phosphorylation, but T305/306 phosphorylation selectively promoted LTD. In response to excitatory LTP versus LTD stimuli, the differential T305/306 phosphorylation directed CaMKII movement to either excitatory or inhibitory synapses, thereby coordinating plasticity at both synapse types. Fast T305/306 phosphorylation required prior T286 phosphorylation and then curbed CaMKII activity by two mechanisms: (i) a cis-subunit reaction reduced both Ca2+ stimulation and autonomous activity and (ii) a trans-subunit reaction enabled complete activity shutdown and feed-forward inhibition of further T286 phosphorylation. These are fundamental additions to the long-studied CaMKII regulation and function in neuronal plasticity.
Collapse
Affiliation(s)
- Sarah G Cook
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Olivia R Buonarati
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Steven J Coultrap
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - K Ulrich Bayer
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.
| |
Collapse
|
12
|
Changes in Behavior and the Expression of Ionotropic Glutamate Receptor Genes in the Brains of Adult Rats after Neonatal Administration of Bacterial Lipopolysaccharide. ACTA ACUST UNITED AC 2020. [DOI: 10.1007/s11055-020-01025-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
13
|
Schwenk J, Fakler B. Building of AMPA‐type glutamate receptors in the endoplasmic reticulum and its implication for excitatory neurotransmission. J Physiol 2020; 599:2639-2653. [DOI: 10.1113/jp279025] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 07/21/2020] [Indexed: 11/08/2022] Open
Affiliation(s)
- Jochen Schwenk
- Institute of Physiology, Faculty of Medicine University of Freiburg Hermann‐Herder‐Str. 7 Freiburg 79104 Germany
| | - Bernd Fakler
- Institute of Physiology, Faculty of Medicine University of Freiburg Hermann‐Herder‐Str. 7 Freiburg 79104 Germany
- Signalling Research Centres BIOSS and CIBSS Schänzlestr. 18 Freiburg 79104 Germany
- Center for Basics in NeuroModulation Breisacherstr. 4 Freiburg 79106 Germany
| |
Collapse
|
14
|
Royo M, Gutiérrez Y, Fernández-Monreal M, Gutiérrez-Eisman S, Jiménez R, Jurado S, Esteban JA. A retention-release mechanism based on RAB11FIP2 for AMPA receptor synaptic delivery during long-term potentiation. J Cell Sci 2019; 132:jcs.234237. [PMID: 31757887 DOI: 10.1242/jcs.234237] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 11/15/2019] [Indexed: 02/05/2023] Open
Abstract
It is well--established that Rab11-dependent recycling endosomes drive the activity-dependent delivery of AMPA receptors (AMPARs) into synapses during long-term potentiation (LTP). Nevertheless, the molecular basis for this specialized function of recycling endosomes is still unknown. Here, we have investigated RAB11FIP2 (FIP2 hereafter) as a potential effector of Rab11-dependent trafficking during LTP in rat hippocampal slices. Surprisingly, we found that FIP2 operates independently from Rab11 proteins, and acts as a negative regulator of AMPAR synaptic trafficking. Under basal conditions, FIP2 associates with AMPARs at immobile compartments, separately from recycling endosomes. Using shRNA-mediated knockdown, we found that FIP2 prevents GluA1 (encoded by the Gria1 gene) AMPARs from reaching the surface of dendritic spines in the absence of neuronal stimulation. Upon induction of LTP, FIP2 is rapidly mobilized, dissociates from AMPARs and undergoes dephosphorylation. Interestingly, this dissociation of the FIP2-AMPAR complex, together with FIP2 dephosphorylation, is required for LTP, but the interaction between FIP2 and Rab11 proteins is not. Based on these results, we propose a retention-release mechanism, where FIP2 acts as a gate that restricts the trafficking of AMPARs, until LTP induction triggers their release and allows synaptic delivery.
Collapse
Affiliation(s)
- María Royo
- Department of Neurobiology, Centro de Biología Molecular 'Severo Ochoa' (CSIC-UAM), 28049 Madrid, Spain
| | - Yolanda Gutiérrez
- Department of Neurobiology, Centro de Biología Molecular 'Severo Ochoa' (CSIC-UAM), 28049 Madrid, Spain
| | - Mónica Fernández-Monreal
- Department of Neurobiology, Centro de Biología Molecular 'Severo Ochoa' (CSIC-UAM), 28049 Madrid, Spain
| | - Silvia Gutiérrez-Eisman
- Department of Neurobiology, Centro de Biología Molecular 'Severo Ochoa' (CSIC-UAM), 28049 Madrid, Spain
| | - Raquel Jiménez
- Department of Neurobiology, Centro de Biología Molecular 'Severo Ochoa' (CSIC-UAM), 28049 Madrid, Spain
| | - Sandra Jurado
- Instituto de Neurociencias CSIC-UMH, 03550 San Juan de Alicante, Spain
| | - José A Esteban
- Department of Neurobiology, Centro de Biología Molecular 'Severo Ochoa' (CSIC-UAM), 28049 Madrid, Spain
| |
Collapse
|
15
|
An ER Assembly Line of AMPA-Receptors Controls Excitatory Neurotransmission and Its Plasticity. Neuron 2019; 104:680-692.e9. [DOI: 10.1016/j.neuron.2019.08.033] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 06/28/2019] [Accepted: 08/20/2019] [Indexed: 11/15/2022]
|
16
|
Novel Positive Allosteric Modulators of AMPA Receptors Based on 3,7-Diazabicyclo[3.3.1]nonane Scaffold. Mol Neurobiol 2019; 57:191-199. [PMID: 31515692 DOI: 10.1007/s12035-019-01768-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 08/29/2019] [Indexed: 01/19/2023]
Abstract
A series of new positive allosteric modulators (PAMs) of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors based on 3,7-diazabicyclo[3.3.1]nonane scaffold have been designed, synthesized, and analyzed. In electrophysiological patch clamp studies, several compounds have demonstrated a sub-nanomolar potency. Compound 4 in in vivo tests showed anti-amnestic properties in the scopolamine-induced model of amnesia in the step-through passive avoidance or maximal electroshock experiments in rats at 0.01 mg/kg showing a significant "dose-response" advantage over memantine. Based on the analysis of the flexible docking results of PAMs, the cyclothiazide-like mechanism of binding mode was suggested as the major site for the interaction with AMPA receptors.
Collapse
|
17
|
Cao FL, Xu M, Gong K, Wang Y, Wang R, Chen X, Chen J. Imbalance Between Excitatory and Inhibitory Synaptic Transmission in the Primary Somatosensory Cortex Caused by Persistent Nociception in Rats. THE JOURNAL OF PAIN 2019; 20:917-931. [PMID: 30742914 DOI: 10.1016/j.jpain.2018.11.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 08/04/2018] [Accepted: 11/12/2018] [Indexed: 11/25/2022]
Abstract
There is substantial evidence supporting the notion that the primary somatosensory (S1) cortex is an important structure involved in the perceptional component of pain. However, investigations have mainly focused on other pain-related formations, and few reports have been provided to investigate the synaptic plasticity in the S1 cortex in response to persistent pain. In the present study, we report that bee venom (BV) injection triggered an imbalance between excitatory and inhibitory synaptic transmission in the S1 cortex in rats. Using a multi-electrode array recording, we found that BV-induced persistent inflammatory pain led to temporal and spatial enhancement of synaptic plasticity. Moreover, slice patch clamp recordings on identified pyramidal neurons demonstrated that BV injection increased presynaptic and postsynaptic transmission in excitatory synapses and decreased postsynaptic transmission in inhibitory synapses in the layer II/III neurons within the S1 cortex. In immunohistochemistry and Western blot sections, the distribution and expression of total AMPA receptor subunits and gamma-amino butyric acid-A (GABAA) were unaffected, although the membrane fractions of GluR2 and GABAA were decreased, and their cytosolic fractions were increased in contrast. The change of GluR1 was opposite to that of GluR2, and GluR3 did not change significantly. Our studies, therefore, provide direct evidence for both presynaptic and postsynaptic changes in synapses within the S1 cortex in persistent nociception, which are probably related to the membrane trafficking of GluR1, GluR2, and GABAA. Perspective: Increased synaptic plasticity was detected in S1 after peripheral nociception, with enhanced excitatory and decreased inhibitory synaptic transmissions. Increased GluR1, and decreased GABAAα1 and GluR2 membrane trafficking were detected. Therefore, the disrupted excitatory/inhibitory balance in transmissions is involved in nociception processing, and S1 can be a potential antinociceptive site.
Collapse
Affiliation(s)
- Fa-Le Cao
- The Department of Neurology, The 88th Hospital of PLA, Tai'an, PR China; Institute for Biomedical Sciences of Pain and Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an, PR China
| | - Min Xu
- The Department of Nephrology, The 88th Hospital of PLA, Tai'an, PR China
| | - Kerui Gong
- Department of Oral and Maxillofacial Surgery, University of California San Francisco, California
| | - Yan Wang
- Institute for Biomedical Sciences of Pain and Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an, PR China
| | - Ruirui Wang
- Institute for Biomedical Sciences of Pain and Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an, PR China
| | - Xuefeng Chen
- Institute for Biomedical Sciences of Pain and Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an, PR China
| | - Jun Chen
- Institute for Biomedical Sciences of Pain and Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an, PR China.
| |
Collapse
|
18
|
Mölders A, Koch A, Menke R, Klöcker N. Heterogeneity of the astrocytic AMPA-receptor transcriptome. Glia 2018; 66:2604-2616. [PMID: 30370555 DOI: 10.1002/glia.23514] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 07/01/2018] [Accepted: 07/18/2018] [Indexed: 11/06/2022]
Abstract
Astrocytes form the largest class of glial cells in the central nervous system. They serve plenty of diverse functions that range from supporting the formation and proper operation of synapses to controlling the blood-brain barrier. For many of them, the expression of ionotropic glutamate receptors of the AMPA subtype (AMPARs) in astrocytes is of key importance. AMPARs form as macromolecular protein complexes, whose composition of the pore-lining GluA subunits and of an extensive set of core and peripheral complex constituents defines both their trafficking and gating behavior. Although astrocytic AMPARs have been reported to exhibit heterogeneous properties, their molecular composition is largely unknown. In this study, we sought to quantify the astrocytic AMPAR transcriptome during brain development and with respect to selected brain regions. Whereas the early postnatal pattern of AMPAR mRNA expression showed minor variation over time, it did show significant heterogeneity in different brain regions. Cerebellar astrocytes express a combination of AMPAR complex constituents that is remarkably distinct from the one in neocortical or hippocampal astrocytes. Our study provides a workflow and a first reference for future investigations into the molecular and functional diversity of glial AMPARs.
Collapse
Affiliation(s)
- Andrea Mölders
- Institute of Neural and Sensory Physiology, Medical Faculty, University of Düsseldorf, Düsseldorf, Germany
| | - Angela Koch
- Institute of Neural and Sensory Physiology, Medical Faculty, University of Düsseldorf, Düsseldorf, Germany
| | - Raphael Menke
- Institute of Neural and Sensory Physiology, Medical Faculty, University of Düsseldorf, Düsseldorf, Germany
| | - Nikolaj Klöcker
- Institute of Neural and Sensory Physiology, Medical Faculty, University of Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
19
|
Drapier T, Geubelle P, Bouckaert C, Nielsen L, Laulumaa S, Goffin E, Dilly S, Francotte P, Hanson J, Pochet L, Kastrup JS, Pirotte B. Enhancing Action of Positive Allosteric Modulators through the Design of Dimeric Compounds. J Med Chem 2018; 61:5279-5291. [DOI: 10.1021/acs.jmedchem.8b00250] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Thomas Drapier
- Laboratory of Medicinal Chemistry, Center for Interdisciplinary Research on Medicines (CIRM), ULiège, Quartier Hôpital, Avenue Hippocrate, 15, B36, B-4000 Liège, Belgium
| | - Pierre Geubelle
- Laboratory of Medicinal Chemistry, Center for Interdisciplinary Research on Medicines (CIRM), ULiège, Quartier Hôpital, Avenue Hippocrate, 15, B36, B-4000 Liège, Belgium
- Laboratory of Molecular Pharmacology, GIGA-Molecular Biology of Diseases, ULiège, B34, Quartier Hôpital, Avenue de l’hôpital, 11, B-4000 Liège, Belgium
| | - Charlotte Bouckaert
- NAmur MEdicine & Drug Innovation Center (NAMEDIC), NARILIS, UNamur, rue de Bruxelles 61, B-5000 Namur, Belgium
| | - Lise Nielsen
- Biostructural Research, Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| | - Saara Laulumaa
- Biostructural Research, Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| | - Eric Goffin
- Laboratory of Medicinal Chemistry, Center for Interdisciplinary Research on Medicines (CIRM), ULiège, Quartier Hôpital, Avenue Hippocrate, 15, B36, B-4000 Liège, Belgium
| | - Sébastien Dilly
- Laboratory of Medicinal Chemistry, Center for Interdisciplinary Research on Medicines (CIRM), ULiège, Quartier Hôpital, Avenue Hippocrate, 15, B36, B-4000 Liège, Belgium
| | - Pierre Francotte
- Laboratory of Medicinal Chemistry, Center for Interdisciplinary Research on Medicines (CIRM), ULiège, Quartier Hôpital, Avenue Hippocrate, 15, B36, B-4000 Liège, Belgium
| | - Julien Hanson
- Laboratory of Medicinal Chemistry, Center for Interdisciplinary Research on Medicines (CIRM), ULiège, Quartier Hôpital, Avenue Hippocrate, 15, B36, B-4000 Liège, Belgium
- Laboratory of Molecular Pharmacology, GIGA-Molecular Biology of Diseases, ULiège, B34, Quartier Hôpital, Avenue de l’hôpital, 11, B-4000 Liège, Belgium
| | - Lionel Pochet
- NAmur MEdicine & Drug Innovation Center (NAMEDIC), NARILIS, UNamur, rue de Bruxelles 61, B-5000 Namur, Belgium
| | - Jette Sandholm Kastrup
- Biostructural Research, Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| | - Bernard Pirotte
- Laboratory of Medicinal Chemistry, Center for Interdisciplinary Research on Medicines (CIRM), ULiège, Quartier Hôpital, Avenue Hippocrate, 15, B36, B-4000 Liège, Belgium
| |
Collapse
|
20
|
Lavrov MI, Karlov DS, Palyulin VA, Grigoriev VV, Zamoyski VL, Brkich GE, Pyatigorskaya NV, Zapolskiy ME. Novel positive allosteric modulator of AMPA-receptors based on tricyclic scaffold. MENDELEEV COMMUNICATIONS 2018. [DOI: 10.1016/j.mencom.2018.05.028] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
21
|
Gallo A, Vukic D, Michalík D, O’Connell MA, Keegan LP. ADAR RNA editing in human disease; more to it than meets the I. Hum Genet 2017; 136:1265-1278. [DOI: 10.1007/s00439-017-1837-0] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 08/22/2017] [Indexed: 01/08/2023]
|
22
|
MAP1B Light Chain Modulates Synaptic Transmission via AMPA Receptor Intracellular Trapping. J Neurosci 2017; 37:9945-9963. [PMID: 28904092 DOI: 10.1523/jneurosci.0505-17.2017] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 08/17/2017] [Accepted: 08/18/2017] [Indexed: 12/19/2022] Open
Abstract
The regulated transport of AMPA-type glutamate receptors (AMPARs) to the synaptic membrane is a key mechanism to determine the strength of excitatory synaptic transmission in the brain. In this work, we uncovered a new role for the microtubule-associated protein MAP1B in modulating access of AMPARs to the postsynaptic membrane. Using mice and rats of either sex, we show that MAP1B light chain (LC) accumulates in the somatodendritic compartment of hippocampal neurons, where it forms immobile complexes on microtubules that limit vesicular transport. These complexes restrict AMPAR dendritic mobility, leading to the intracellular trapping of receptors and impairing their access to the dendritic surface and spines. Accordingly, increasing MAP1B-LC expression depresses AMPAR-mediated synaptic transmission. This effect is specific for the GluA2 subunit of the AMPAR and requires glutamate receptor interacting protein 1 (GRIP1) interaction with MAP1B-LC. Therefore, MAP1B-LC represents an alternative link between GRIP1-AMPARs and microtubules that does not result in productive transport, but rather limits AMPAR availability for synaptic insertion, with a direct impact on synaptic transmission.SIGNIFICANCE STATEMENT The ability of neurons to modify their synaptic connections, known as synaptic plasticity, is accepted as the cellular basis for learning and memory. One mechanism for synaptic plasticity is the regulated addition and removal of AMPA-type glutamate receptors (AMPARs) at excitatory synapses. In this study, we found that a microtubule-associated protein, MAP1B light chain (MAP1B-LC), participates in this process. MAP1B-LC forms immobile complexes along dendrites. These complexes limit intracellular vesicular trafficking and trap AMPARs inside the dendritic shaft. In this manner, MAP1B restricts the access of AMPARs to dendritic spines and the postsynaptic membrane, contributing to downregulating synaptic transmission.
Collapse
|
23
|
Lee SH, Jin C, Cai E, Ge P, Ishitsuka Y, Teng KW, de Thomaz AA, Nall D, Baday M, Jeyifous O, Demonte D, Dundas CM, Park S, Delgado JY, Green WN, Selvin PR. Super-resolution imaging of synaptic and Extra-synaptic AMPA receptors with different-sized fluorescent probes. eLife 2017; 6:27744. [PMID: 28749340 PMCID: PMC5779237 DOI: 10.7554/elife.27744] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 07/26/2017] [Indexed: 12/13/2022] Open
Abstract
Previous studies tracking AMPA receptor (AMPAR) diffusion at synapses observed a large mobile extrasynaptic AMPAR pool. Using super-resolution microscopy, we examined how fluorophore size and photostability affected AMPAR trafficking outside of, and within, post-synaptic densities (PSDs) from rats. Organic fluorescent dyes (≈4 nm), quantum dots, either small (≈10 nm diameter; sQDs) or big (>20 nm; bQDs), were coupled to AMPARs via different-sized linkers. We find that >90% of AMPARs labeled with fluorescent dyes or sQDs were diffusing in confined nanodomains in PSDs, which were stable for 15 min or longer. Less than 10% of sQD-AMPARs were extrasynaptic and highly mobile. In contrast, 5-10% of bQD-AMPARs were in PSDs and 90-95% were extrasynaptic as previously observed. Contrary to the hypothesis that AMPAR entry is limited by the occupancy of open PSD 'slots', our findings suggest that AMPARs rapidly enter stable 'nanodomains' in PSDs with lifetime >15 min, and do not accumulate in extrasynaptic membranes.
Collapse
Affiliation(s)
- Sang Hak Lee
- Department of Physics, Center for Biophysics, and Quantitative Biology, and Center for the Physics of Living Cells, University of Illinois, Urbana-Champaign, Champaign, United States
| | - Chaoyi Jin
- Department of Physics, Center for Biophysics, and Quantitative Biology, and Center for the Physics of Living Cells, University of Illinois, Urbana-Champaign, Champaign, United States
| | - En Cai
- Department of Physics, Center for Biophysics, and Quantitative Biology, and Center for the Physics of Living Cells, University of Illinois, Urbana-Champaign, Champaign, United States
| | - Pinghua Ge
- Department of Physics, Center for Biophysics, and Quantitative Biology, and Center for the Physics of Living Cells, University of Illinois, Urbana-Champaign, Champaign, United States
| | - Yuji Ishitsuka
- Department of Physics, Center for Biophysics, and Quantitative Biology, and Center for the Physics of Living Cells, University of Illinois, Urbana-Champaign, Champaign, United States
| | - Kai Wen Teng
- Department of Physics, Center for Biophysics, and Quantitative Biology, and Center for the Physics of Living Cells, University of Illinois, Urbana-Champaign, Champaign, United States
| | - Andre A de Thomaz
- Department of Physics, Center for Biophysics, and Quantitative Biology, and Center for the Physics of Living Cells, University of Illinois, Urbana-Champaign, Champaign, United States
| | - Duncan Nall
- Department of Physics, Center for Biophysics, and Quantitative Biology, and Center for the Physics of Living Cells, University of Illinois, Urbana-Champaign, Champaign, United States
| | - Murat Baday
- Department of Physics, Center for Biophysics, and Quantitative Biology, and Center for the Physics of Living Cells, University of Illinois, Urbana-Champaign, Champaign, United States
| | - Okunola Jeyifous
- Department of Neurobiology, University of Chicago and the Marine Biological Laboratory, Chicago, United States
| | - Daniel Demonte
- Department of Chemical and Biological Engineering, University at Buffalo, Buffalo, United States
| | - Christopher M Dundas
- Department of Chemical and Biological Engineering, University at Buffalo, Buffalo, United States
| | - Sheldon Park
- Department of Chemical and Biological Engineering, University at Buffalo, Buffalo, United States
| | - Jary Y Delgado
- Department of Neurobiology, University of Chicago and the Marine Biological Laboratory, Chicago, United States
| | - William N Green
- Department of Neurobiology, University of Chicago and the Marine Biological Laboratory, Chicago, United States
| | - Paul R Selvin
- Department of Physics, Center for Biophysics, and Quantitative Biology, and Center for the Physics of Living Cells, University of Illinois, Urbana-Champaign, Champaign, United States
| |
Collapse
|
24
|
Poddar R, Chen A, Winter L, Rajagopal S, Paul S. Role of AMPA receptors in homocysteine-NMDA receptor-induced crosstalk between ERK and p38 MAPK. J Neurochem 2017; 142:560-573. [PMID: 28543279 DOI: 10.1111/jnc.14078] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 04/28/2017] [Accepted: 05/09/2017] [Indexed: 01/12/2023]
Abstract
Homocysteine, a metabolite of the methionine cycle has been reported to play a role in neurotoxicity through activation of N-methyl-d-aspartate receptors (NMDAR)-mediated signaling pathway. The proposed mechanisms associated with homocysteine-NMDAR-induced neurotoxicity involve a unique signaling pathway that triggers a crosstalk between extracellular signal-regulated kinase (ERK) and p38 MAPKs, where activation of p38 MAPK is downstream of and dependent on ERK MAPK. However, the molecular basis of the ERK MAPK-mediated p38 MAPK activation is not understood. This study investigates whether α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPARs) play a role in facilitating the ERK MAPK-mediated p38 MAPK activation. Using surface biotinylation and immunoblotting approaches we show that treatment with homocysteine leads to a decrease in surface expression of GluA2-AMPAR subunit in neurons, but have no effect on the surface expression of GluA1-AMPAR subunit. Inhibition of NMDAR activation with D-AP5 or ERK MAPK phosphorylation with PD98059 attenuates homocysteine-induced decrease in surface expression of GluA2-AMPAR subunit. The decrease in surface expression of GluA2-AMPAR subunit is associated with p38 MAPK phosphorylation, which is inhibited by 1-napthyl acetyl spermine trihydrochloride (NASPM), a selective antagonist of GluA2-lacking Ca2+ -permeable AMPARs. These results suggest that homocysteine-NMDAR-mediated ERK MAPK phosphorylation leads to a decrease in surface expression of GluA2-AMPAR subunit resulting in Ca2+ influx through the GluA2-lacking Ca2+ -permeable AMPARs and p38 MAPK phosphorylation. Cell death assays further show that inhibition of AMPAR activity with 2,3-dioxo-6-nitro-1,2,3,4,tetrahydrobenzoquinoxaline-7-sulfonamide (NBQX)/6-cyano-7-nitroquinoxaline-2,3, -dione (CNQX) or GluA2-lacking Ca2+ -permeable AMPAR activity with NASPM attenuates homocysteine-induced neurotoxicity. We have identified an important mechanism involved in homocysteine-induced neurotoxicity that highlights the intermediary role of GluA2-lacking Ca2+ -permeable AMPARs in the crosstalk between ERK and p38 MAPKs.
Collapse
Affiliation(s)
- Ranjana Poddar
- Department of Neurology, University of New Mexico Health Sciences Center, University of New Mexico, Albuquerque, New Mexico, USA
| | - Alexandria Chen
- Department of Neurology, University of New Mexico Health Sciences Center, University of New Mexico, Albuquerque, New Mexico, USA
| | - Lucas Winter
- Department of Neurology, University of New Mexico Health Sciences Center, University of New Mexico, Albuquerque, New Mexico, USA
| | - Sathyanarayanan Rajagopal
- Department of Neurology, University of New Mexico Health Sciences Center, University of New Mexico, Albuquerque, New Mexico, USA
| | - Surojit Paul
- Department of Neurology, University of New Mexico Health Sciences Center, University of New Mexico, Albuquerque, New Mexico, USA
| |
Collapse
|
25
|
Evans AJ, Gurung S, Wilkinson KA, Stephens DJ, Henley JM. Assembly, Secretory Pathway Trafficking, and Surface Delivery of Kainate Receptors Is Regulated by Neuronal Activity. Cell Rep 2017; 19:2613-2626. [PMID: 28636947 PMCID: PMC5489663 DOI: 10.1016/j.celrep.2017.06.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 04/17/2017] [Accepted: 05/25/2017] [Indexed: 01/03/2023] Open
Abstract
Ionotropic glutamate receptor (iGluR) trafficking and function underpin excitatory synaptic transmission and plasticity and shape neuronal networks. It is well established that the transcription, translation, and endocytosis/recycling of iGluRs are all regulated by neuronal activity, but much less is known about the activity dependence of iGluR transport through the secretory pathway. Here, we use the kainate receptor subunit GluK2 as a model iGluR cargo to show that the assembly, early secretory pathway trafficking, and surface delivery of iGluRs are all controlled by neuronal activity. We show that the delivery of de novo kainate receptors is differentially regulated by modulation of GluK2 Q/R editing, PKC phosphorylation, and PDZ ligand interactions. These findings reveal that, in addition to short-term regulation of iGluRs by recycling/endocytosis and long-term modulation by altered transcription/translation, the trafficking of iGluRs through the secretory pathway is under tight activity-dependent control to determine the numbers and properties of surface-expressed iGluRs.
Collapse
Affiliation(s)
- Ashley J Evans
- School of Biochemistry, Centre for Synaptic Plasticity, Biomedical Sciences Building, University of Bristol, University Walk, Bristol BS8 1TD, UK
| | - Sonam Gurung
- School of Biochemistry, Centre for Synaptic Plasticity, Biomedical Sciences Building, University of Bristol, University Walk, Bristol BS8 1TD, UK
| | - Kevin A Wilkinson
- School of Biochemistry, Centre for Synaptic Plasticity, Biomedical Sciences Building, University of Bristol, University Walk, Bristol BS8 1TD, UK
| | - David J Stephens
- School of Biochemistry, Centre for Synaptic Plasticity, Biomedical Sciences Building, University of Bristol, University Walk, Bristol BS8 1TD, UK
| | - Jeremy M Henley
- School of Biochemistry, Centre for Synaptic Plasticity, Biomedical Sciences Building, University of Bristol, University Walk, Bristol BS8 1TD, UK.
| |
Collapse
|
26
|
Coleman SK, Hou Y, Willibald M, Semenov A, Möykkynen T, Keinänen K. Aggregation Limits Surface Expression of Homomeric GluA3 Receptors. J Biol Chem 2016; 291:8784-94. [PMID: 26912664 DOI: 10.1074/jbc.m115.689125] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Indexed: 11/06/2022] Open
Abstract
AMPA receptors are glutamate-gated cation channels assembled from GluA1-4 subunits and have properties that are strongly dependent on the subunit composition. The subunits have different propensities to form homomeric or various heteromeric receptors expressed on cell surface, but the underlying mechanisms are still poorly understood. Here, we examined the biochemical basis for the poor ability of GluA3 subunits to form homomeric receptors, linked previously to two amino acid residues, Tyr-454 and Arg-461, in its ligand binding domain (LBD). Surface expression of GluA3 was improved by co-assembly with GluA2 but not with stargazin, a trafficking chaperone and modulator of AMPA receptors. The secretion efficiency of GluA2 and GluA3 LBDs paralleled the transport difference between the respective full-length receptors and was similarly dependent on Tyr-454/Arg-461 but not on LBD stability. In comparison to GluA2, GluA3 homomeric receptors showed a strong and Tyr-454/Arg-461-dependent tendency to aggregate both in the macroscopic scale measured as lower solubility in nonionic detergent and in the microscopic scale evident as the preponderance of hydrodynamically large structures in density gradient centrifugation and native gel electrophoresis. We conclude that the impaired surface expression of homomeric GluA3 receptors is caused by nonproductive assembly and aggregation to which LBD residues Tyr-454 and Arg-461 strongly contribute. This aggregation inhibits the entry of newly synthesized GluA3 receptors to the secretory pathway.
Collapse
Affiliation(s)
- Sarah K Coleman
- From the Department of Biosciences, Division of Biochemistry and Biotechnology, University of Helsinki, Helsinki FI-00014, Finland
| | - Ying Hou
- From the Department of Biosciences, Division of Biochemistry and Biotechnology, University of Helsinki, Helsinki FI-00014, Finland
| | - Marina Willibald
- From the Department of Biosciences, Division of Biochemistry and Biotechnology, University of Helsinki, Helsinki FI-00014, Finland
| | - Artur Semenov
- From the Department of Biosciences, Division of Biochemistry and Biotechnology, University of Helsinki, Helsinki FI-00014, Finland
| | - Tommi Möykkynen
- From the Department of Biosciences, Division of Biochemistry and Biotechnology, University of Helsinki, Helsinki FI-00014, Finland
| | - Kari Keinänen
- From the Department of Biosciences, Division of Biochemistry and Biotechnology, University of Helsinki, Helsinki FI-00014, Finland
| |
Collapse
|
27
|
Joshi G, Bekier ME, Wang Y. Golgi fragmentation in Alzheimer's disease. Front Neurosci 2015; 9:340. [PMID: 26441511 PMCID: PMC4585163 DOI: 10.3389/fnins.2015.00340] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 09/08/2015] [Indexed: 11/24/2022] Open
Abstract
The Golgi apparatus is an essential cellular organelle for post-translational modifications, sorting, and trafficking of membrane and secretory proteins. Proper functionality of the Golgi requires the formation of its unique cisternal-stacking morphology. The Golgi structure is disrupted in a variety of neurodegenerative diseases, suggesting a common mechanism and contribution of Golgi defects in neurodegenerative disorders. A recent study on Alzheimer's disease (AD) revealed that phosphorylation of the Golgi stacking protein GRASP65 disrupts its function in Golgi structure formation, resulting in Golgi fragmentation. Inhibiting GRASP65 phosphorylation restores the Golgi morphology from Aβ-induced fragmentation and reduces Aβ production. Perturbing Golgi structure and function in neurons may directly impact trafficking, processing, and sorting of a variety of proteins essential for synaptic and dendritic integrity. Therefore, Golgi defects may ultimately promote the development of AD. In the current review, we focus on the cellular impact of impaired Golgi morphology and its potential relationship to AD disease development.
Collapse
Affiliation(s)
- Gunjan Joshi
- Department of Molecular, Cellular and Developmental Biology, University of Michigan Ann Arbor, MI, USA
| | - Michael E Bekier
- Department of Molecular, Cellular and Developmental Biology, University of Michigan Ann Arbor, MI, USA
| | - Yanzhuang Wang
- Department of Molecular, Cellular and Developmental Biology, University of Michigan Ann Arbor, MI, USA ; Department of Neurology, University of Michigan School of Medicine Ann Arbor, MI, USA
| |
Collapse
|
28
|
Castilho Á, Madsen E, Ambrósio AF, Veruki ML, Hartveit E. Diabetic hyperglycemia reduces Ca2+ permeability of extrasynaptic AMPA receptors in AII amacrine cells. J Neurophysiol 2015; 114:1545-53. [PMID: 26156384 DOI: 10.1152/jn.00295.2015] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 07/01/2015] [Indexed: 11/22/2022] Open
Abstract
There is increasing evidence that diabetic retinopathy is a primary neuropathological disorder that precedes the microvascular pathology associated with later stages of the disease. Recently, we found evidence for altered functional properties of synaptic α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors in A17, but not AII, amacrine cells in the mammalian retina, and the observed changes were consistent with an upregulation of the GluA2 subunit, a key determinant of functional properties of AMPA receptors, including Ca(2+) permeability and current-voltage (I-V) rectification properties. Here, we have investigated functional changes of extrasynaptic AMPA receptors in AII amacrine cells evoked by diabetes. With patch-clamp recording of nucleated patches from retinal slices, we measured Ca(2+) permeability and I-V rectification in rats with ∼3 wk of streptozotocin-induced diabetes and age-matched, noninjected controls. Under bi-ionic conditions (extracellular Ca(2+) concentration = 30 mM, intracellular Cs(+) concentration = 171 mM), the reversal potential (Erev) of AMPA-evoked currents indicated a significant reduction of Ca(2+) permeability in diabetic animals [Erev = -17.7 mV, relative permeability of Ca(2+) compared with Cs(+) (PCa/PCs) = 1.39] compared with normal animals (Erev = -7.7 mV, PCa/PCs = 2.35). Insulin treatment prevented the reduction of Ca(2+) permeability. I-V rectification was examined by calculating a rectification index (RI) as the ratio of the AMPA-evoked conductance at +40 and -60 mV. The degree of inward rectification in patches from diabetic animals (RI = 0.48) was significantly reduced compared with that in normal animals (RI = 0.30). These results suggest that diabetes evokes a change in the functional properties of extrasynaptic AMPA receptors of AII amacrine cells. These changes could be representative for extrasynaptic AMPA receptors elsewhere in AII amacrine cells and suggest that synaptic and extrasynaptic AMPA receptors are differentially regulated.
Collapse
Affiliation(s)
- Áurea Castilho
- Department of Biomedicine, University of Bergen, Bergen, Norway; Institute of Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Eirik Madsen
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - António F Ambrósio
- Institute of Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Coimbra, Portugal; Center for Neuroscience and Cell Biology, Institute of Biomedical Imaging and Life Sciences (CNC.IBILI) Consortium, University of Coimbra, Coimbra, Portugal; and Association for Innovation and Biomedical Research on Light and Image, Coimbra, Portugal
| | | | - Espen Hartveit
- Department of Biomedicine, University of Bergen, Bergen, Norway;
| |
Collapse
|
29
|
Chen N, Pandya NJ, Koopmans F, Castelo-Székelv V, van der Schors RC, Smit AB, Li KW. Interaction proteomics reveals brain region-specific AMPA receptor complexes. J Proteome Res 2014; 13:5695-706. [PMID: 25337787 DOI: 10.1021/pr500697b] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Fast excitatory synaptic transmission in the brain is mediated by glutamate acting on postsynaptic AMPA receptors. Recent studies have revealed a substantial number of AMPA receptor auxiliary proteins, which potentially contribute to the regulation of AMPA receptor trafficking, subcellular receptor localization, and receptor gating properties. Here we examined the AMPA receptor interactomes from cortex, hippocampus, and cerebellum by comprehensive interaction proteomics. The study reveals that AMPA receptor auxiliary proteins are engaged in distinct brain region-specific AMPA receptors subcomplexes, which might underlie brain region-specific differential regulation of AMPA receptor properties. Depending on the brain region, an interacting protein can be involved in an AMPA and a non-AMPA receptor complex.
Collapse
Affiliation(s)
- Ning Chen
- Department of Molecular and Cellular Neurobiology, and ‡Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, VU University , De Boelelaan 1085, 1081 HV Amsterdam, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
30
|
Francotte P, Nørholm AB, Deva T, Olsen L, Frydenvang K, Goffin E, Fraikin P, de Tullio P, Challal S, Thomas JY, Iop F, Louis C, Botez-Pop I, Lestage P, Danober L, Kastrup JS, Pirotte B. Positive Allosteric Modulators of 2-Amino-3-(3-hydroxy-5-methylisoxazol-4-yl)propionic Acid Receptors Belonging to 4-Cyclopropyl-3,4-dihydro-2H-1,2,4-pyridothiadiazine Dioxides and Diversely Chloro-Substituted 4-Cyclopropyl-3,4-dihydro-2H-1,2,4-benzothiadiazine 1,1-Dioxides. J Med Chem 2014; 57:9539-53. [DOI: 10.1021/jm501268r] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Pierre Francotte
- Department
of Medicinal Chemistry, Center for Interdisciplinary Research on Medicines
(CIRM), University of Liege, Avenue de l’Hôpital,
1, B36, B-4000 Liège, Belgium
| | - Ann-Beth Nørholm
- Department
of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken, 2, DK-2100 Copenhagen, Denmark
| | - Taru Deva
- Department
of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken, 2, DK-2100 Copenhagen, Denmark
| | - Lars Olsen
- Department
of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken, 2, DK-2100 Copenhagen, Denmark
| | - Karla Frydenvang
- Department
of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken, 2, DK-2100 Copenhagen, Denmark
| | - Eric Goffin
- Department
of Medicinal Chemistry, Center for Interdisciplinary Research on Medicines
(CIRM), University of Liege, Avenue de l’Hôpital,
1, B36, B-4000 Liège, Belgium
| | - Pierre Fraikin
- Department
of Medicinal Chemistry, Center for Interdisciplinary Research on Medicines
(CIRM), University of Liege, Avenue de l’Hôpital,
1, B36, B-4000 Liège, Belgium
| | - Pascal de Tullio
- Department
of Medicinal Chemistry, Center for Interdisciplinary Research on Medicines
(CIRM), University of Liege, Avenue de l’Hôpital,
1, B36, B-4000 Liège, Belgium
| | - Sylvie Challal
- Institut
de Recherches
Servier, 125 Chemin de Ronde, F-78290 Croissy-sur-Seine, France
| | - Jean-Yves Thomas
- Institut
de Recherches
Servier, 125 Chemin de Ronde, F-78290 Croissy-sur-Seine, France
| | - Fabrice Iop
- Institut
de Recherches
Servier, 125 Chemin de Ronde, F-78290 Croissy-sur-Seine, France
| | - Caroline Louis
- Institut
de Recherches
Servier, 125 Chemin de Ronde, F-78290 Croissy-sur-Seine, France
| | - Iuliana Botez-Pop
- Institut
de Recherches
Servier, 125 Chemin de Ronde, F-78290 Croissy-sur-Seine, France
| | - Pierre Lestage
- Institut
de Recherches
Servier, 125 Chemin de Ronde, F-78290 Croissy-sur-Seine, France
| | - Laurence Danober
- Institut
de Recherches
Servier, 125 Chemin de Ronde, F-78290 Croissy-sur-Seine, France
| | - Jette S. Kastrup
- Department
of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken, 2, DK-2100 Copenhagen, Denmark
| | - Bernard Pirotte
- Department
of Medicinal Chemistry, Center for Interdisciplinary Research on Medicines
(CIRM), University of Liege, Avenue de l’Hôpital,
1, B36, B-4000 Liège, Belgium
| |
Collapse
|
31
|
Zhang C, Li S, Sun Y, Dong W, Piao F, Piao Y, liu S, Guan H, Yu S. Arsenic downregulates gene expression at the postsynaptic density in mouse cerebellum, including genes responsible for long-term potentiation and depression. Toxicol Lett 2014; 228:260-9. [DOI: 10.1016/j.toxlet.2014.05.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Revised: 04/23/2014] [Accepted: 05/02/2014] [Indexed: 01/25/2023]
|
32
|
Rubio MD, Drummond JB, Meador-Woodruff JH. Glutamate receptor abnormalities in schizophrenia: implications for innovative treatments. Biomol Ther (Seoul) 2014; 20:1-18. [PMID: 24116269 PMCID: PMC3792192 DOI: 10.4062/biomolther.2012.20.1.001] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2011] [Accepted: 11/25/2011] [Indexed: 01/18/2023] Open
Abstract
Schizophrenia is a devastating psychiatric illness that afflicts 1% of the population worldwide, resulting in substantial impact to patients, their families, and health care delivery systems. For many years, schizophrenia has been felt to be associated with dysregulated dopaminergic neurotransmission as a key feature of the pathophysiology of the illness. Although numerous studies point to dopaminergic abnormalities in schizophrenia, dopamine dysfunction cannot completely account for all of the symptoms seen in schizophrenia, and dopamine-based treatments are often inadequate and can be associated with serious side effects. More recently, converging lines of evidence have suggested that there are abnormalities of glutamate transmission in schizophrenia. Glutamatergic neurotransmission involves numerous molecules that facilitate glutamate release, receptor activation, glutamate reuptake, and other synaptic activities. Evidence for glutamatergic abnormalities in schizophrenia primarily has implicated the NMDA and AMPA subtypes of the glutamate receptor. The expression of these receptors and other molecules associated with glutamate neurotransmission has been systematically studied in the brain in schizophrenia. These studies have generally revealed region- and molecule-specific changes in glutamate receptor transcript and protein expression in this illness. Given that glutamatergic neurotransmission has been implicated in the pathophysiology of schizophrenia, recent drug development efforts have targeted the glutamate system. Much effort to date has focused on modulation of the NMDA receptor, although more recently other glutamate receptors and transporters have been the targets of drug development. These efforts have been promising thus far, and ongoing efforts to develop additional drugs that modulate glutamatergic neurotransmission are underway that may hold the potential for novel classes of more effective treatments for this serious psychiatric illness.
Collapse
Affiliation(s)
- Maria D Rubio
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294-0021, USA
| | | | | |
Collapse
|
33
|
Möykkynen T, Coleman SK, Semenov A, Keinänen K. The N-terminal domain modulates α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor desensitization. J Biol Chem 2014; 289:13197-205. [PMID: 24652293 DOI: 10.1074/jbc.m113.526301] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
AMPA receptors are tetrameric glutamate-gated ion channels that mediate fast synaptic neurotransmission in mammalian brain. Their subunits contain a two-lobed N-terminal domain (NTD) that comprises over 40% of the mature polypeptide. The NTD is not obligatory for the assembly of tetrameric receptors, and its functional role is still unclear. By analyzing full-length and NTD-deleted GluA1-4 AMPA receptors expressed in HEK 293 cells, we found that the removal of the NTD leads to a significant reduction in receptor transport to the plasma membrane, a higher steady state-to-peak current ratio of glutamate responses, and strongly increased sensitivity to glutamate toxicity in cell culture. Further analyses showed that NTD-deleted receptors display both a slower onset of desensitization and a faster recovery from desensitization of agonist responses. Our results indicate that the NTD promotes the biosynthetic maturation of AMPA receptors and, for membrane-expressed channels, enhances the stability of the desensitized state. Moreover, these findings suggest that interactions of the NTD with extracellular/synaptic ligands may be able to fine-tune AMPA receptor-mediated responses, in analogy with the allosteric regulatory role demonstrated for the NTD of NMDA receptors.
Collapse
Affiliation(s)
- Tommi Möykkynen
- From the Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, 00014 Helsinki, Finland
| | | | | | | |
Collapse
|
34
|
Kinesin-1 regulates synaptic strength by mediating the delivery, removal, and redistribution of AMPA receptors. Neuron 2014; 80:1421-37. [PMID: 24360545 DOI: 10.1016/j.neuron.2013.10.050] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/18/2013] [Indexed: 01/22/2023]
Abstract
A primary determinant of the strength of neurotransmission is the number of AMPA-type glutamate receptors (AMPARs) at synapses. However, we still lack a mechanistic understanding of how the number of synaptic AMPARs is regulated. Here, we show that UNC-116, the C. elegans homolog of vertebrate kinesin-1 heavy chain (KIF5), modifies synaptic strength by mediating the rapid delivery, removal, and redistribution of synaptic AMPARs. Furthermore, by studying the real-time transport of C. elegans AMPAR subunits in vivo, we demonstrate that although homomeric GLR-1 AMPARs can diffuse to and accumulate at synapses in unc-116 mutants, glutamate-gated currents are diminished because heteromeric GLR-1/GLR-2 receptors do not reach synapses in the absence of UNC-116/KIF5-mediated transport. Our data support a model in which ongoing motor-driven delivery and removal of AMPARs controls not only the number but also the composition of synaptic AMPARs, and thus the strength of synaptic transmission.
Collapse
|
35
|
Abstract
Among the largest cells in the body, neurons possess an immense surface area and intricate geometry that poses many unique cell biological challenges. This morphological complexity is critical for neural circuit formation and enables neurons to compartmentalize cell-cell communication and local intracellular signalling to a degree that surpasses other cell types. The adaptive plastic properties of neurons, synapses and circuits have been classically studied by measurement of electrophysiological properties, ionic conductances and excitability. Over the last 15 years, the field of synaptic and neural electrophysiology has collided with neuronal cell biology to produce a more integrated understanding of how these remarkable highly differentiated cells utilize common eukaryotic cellular machinery to decode, integrate and propagate signals in the nervous system. The present article gives a very brief and personal overview of the organelles and trafficking machinery of neuronal dendrites and their role in dendritic and synaptic plasticity.
Collapse
Affiliation(s)
- Michael D Ehlers
- *Neuroscience Research Unit, Pfizer Worldwide Research and Development, 700 Main Street, Cambridge, MA 02139, U.S.A
| |
Collapse
|
36
|
Kim TK, Sul JY, Helmfors H, Langel U, Kim J, Eberwine J. Dendritic glutamate receptor mRNAs show contingent local hotspot-dependent translational dynamics. Cell Rep 2013; 5:114-25. [PMID: 24075992 DOI: 10.1016/j.celrep.2013.08.029] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Revised: 07/24/2013] [Accepted: 08/14/2013] [Indexed: 11/16/2022] Open
Abstract
Protein synthesis in neuronal dendrites underlies long-term memory formation in the brain. Local translation of reporter mRNAs has demonstrated translation in dendrites at focal points called translational hotspots. Various reports have shown that hundreds to thousands of mRNAs are localized to dendrites, yet the dynamics of translation of multiple dendritic mRNAs has remained elusive. Here, we show that the protein translational activities of two dendritically localized mRNAs are spatiotemporally complex but constrained by the translational hotspots in which they are colocalized. Cotransfection of glutamate receptor 2 (GluR2) and GluR4 mRNAs (engineered to encode different fluorescent proteins) into rat hippocampal neurons demonstrates a heterogeneous distribution of translational hotspots for the two mRNAs along dendrites. Stimulation with s-3,5-dihydroxy-phenylglycine modifies the translational dynamics of both of these RNAs in a complex saturable manner. These results suggest that the translational hotspot is a primary structural regulator of the simultaneous yet differential translation of multiple mRNAs in the neuronal dendrite.
Collapse
Affiliation(s)
- Tae Kyung Kim
- Department of Pharmacology, Perelman School of Medicine, Philadelphia, PA 19104-6084, USA
| | | | | | | | | | | |
Collapse
|
37
|
Kintz N, Petzinger GM, Akopian G, Ptasnik S, Williams C, Jakowec MW, Walsh JP. Exercise modifies α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor expression in striatopallidal neurons in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-lesioned mouse. J Neurosci Res 2013; 91:1492-507. [PMID: 23918451 DOI: 10.1002/jnr.23260] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Revised: 04/26/2013] [Accepted: 05/16/2013] [Indexed: 12/11/2022]
Abstract
The α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic-acid-type glutamate receptor (AMPAR) plays a critical role in modulating experience-dependent neuroplasticity, and alterations in AMPAR expression may underlie synaptic dysfunction and disease pathophysiology. Using the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of dopamine (DA) depletion, our previous work showed exercise increases total GluA2 subunit expression and the contribution of GluA2-containing channels in MPTP mice. The purpose of this study was to determine whether exercise-dependent changes in AMPAR expression after MPTP are specific to the striatopallidal (D2 R) or striatonigral (D1 R) medium spiny neuron (MSN) striatal projection pathways. Drd2 -eGFP-BAC transgenic mice were used to delineate differences in AMPAR expression between striatal D2 R-MSNs and D1 R-MSNs. Striatal AMPAR expression was assessed by immunohistochemical (IHC) staining, Western immunoblotting (WB) of preparations enriched for postsynaptic density (PSD), and alterations in the current-voltage relationship of MSNs. We found DA depletion results in the emergence of GluA2-lacking AMPARs selectively in striatopallidal D2 R-MSNs and that exercise reverses this effect in MPTP mice. Exercise-induced changes in AMPAR channels observed after DA depletion were associated with alterations in GluA1 and GluA2 subunit expression in postsynaptic protein, D2 R-MSN cell surface expression, and restoration of corticostriatal plasticity. Mechanisms regulating experience-dependent changes in AMPAR expression may provide innovative therapeutic targets to increase the efficacy of treatments for basal ganglia disorders, including Parkinson's disease.
Collapse
Affiliation(s)
- N Kintz
- The George and MaryLou Boone Center for Parkinson's Disease Research, Department of Neurology, University of Southern California, Los Angeles, California
| | | | | | | | | | | | | |
Collapse
|
38
|
Cioffi CL. Modulation of NMDA receptor function as a treatment for schizophrenia. Bioorg Med Chem Lett 2013; 23:5034-44. [PMID: 23916256 DOI: 10.1016/j.bmcl.2013.07.019] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Revised: 07/03/2013] [Accepted: 07/13/2013] [Indexed: 11/30/2022]
Abstract
Schizophrenia is a devastating mental illness that afflicts nearly 1% of the world's population. Currently available antipsychotics treat positive symptoms, but are largely ineffective at addressing negative symptoms and cognitive dysfunction. Thus, improved pharmacotherapies that treat all aspects of the disease remain a critical unmet need. There is mounting evidence that links NMDA receptor hypofunction and the expression of schizophrenia, and numerous drug discovery programs have developed agents that directly or indirectly potentiate NMDA receptor-mediated neurotransmission. Several compounds have emerged that show promise for treating all symptom sub-domains in both preclinical models and clinical studies, and we will review recent developments in many of these areas.
Collapse
|
39
|
Liu Y, Wang L, Long ZY, Wu YM, Wan Q, Jiang JX, Wang ZG. Inhibiting PTEN protects hippocampal neurons against stretch injury by decreasing membrane translocation of AMPA receptor GluR2 subunit. PLoS One 2013; 8:e65431. [PMID: 23799014 PMCID: PMC3684616 DOI: 10.1371/journal.pone.0065431] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2012] [Accepted: 04/29/2013] [Indexed: 11/18/2022] Open
Abstract
The AMPA type of glutamate receptors (AMPARs)-mediated excitotoxicity is involved in the secondary neuronal death following traumatic brain injury (TBI). But the underlying cellular and molecular mechanisms remain unclear. In this study, the role of phosphatase and tensin homolog deleted on chromosome 10 (PTEN) in GluR2-lacking AMPARs mediated neuronal death was investigated through an in vitro stretch injury model of neurons. It was indicated that both the mRNA and protein levels of PTEN were increased in cultured hippocampal neurons after stretch injury, which was associated with the decreasing expression of GluR2 subunits on the surface of neuronal membrane. Inhibition of PTEN activity by its inhibitor can promote the survival of neurons through preventing reduction of GluR2 on membrane. Moreover, the effect of inhibiting GluR2-lacking AMPARs was similar to PTEN suppression-mediated neuroprotective effect in stretch injury-induced neuronal death. Further evidence identified that the total GluR2 protein of neurons was not changed in all groups. So inhibition of PTEN or blockage of GluR2-lacking AMPARs may attenuate the death of hippocampal neurons post injury through decreasing the translocation of GluR2 subunit on the membrane effectively.
Collapse
Affiliation(s)
- Yuan Liu
- Department of Research Institute of Surgery, Daping Hospital, the Third Military Medical University, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing, China
| | - Li Wang
- Department of Research Institute of Surgery, Daping Hospital, the Third Military Medical University, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing, China
| | - Zai-yun Long
- Department of Research Institute of Surgery, Daping Hospital, the Third Military Medical University, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing, China
| | - Ya-min Wu
- Department of Research Institute of Surgery, Daping Hospital, the Third Military Medical University, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing, China
| | - Qi Wan
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada, United States of America
| | - Jian-xin Jiang
- Department of Research Institute of Surgery, Daping Hospital, the Third Military Medical University, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing, China
| | - Zheng-guo Wang
- Department of Research Institute of Surgery, Daping Hospital, the Third Military Medical University, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing, China
- * E-mail:
| |
Collapse
|
40
|
Kopach O, Voitenko N. Extrasynaptic AMPA receptors in the dorsal horn: Evidence and functional significance. Brain Res Bull 2013. [DOI: 10.1016/j.brainresbull.2012.11.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
41
|
Regulation of AMPA receptor surface trafficking and synaptic plasticity by a cognitive enhancer and antidepressant molecule. Mol Psychiatry 2013; 18:471-84. [PMID: 22733125 PMCID: PMC3606944 DOI: 10.1038/mp.2012.80] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The plasticity of excitatory synapses is an essential brain process involved in cognitive functions, and dysfunctions of such adaptations have been linked to psychiatric disorders such as depression. Although the intracellular cascades that are altered in models of depression and stress-related disorders have been under considerable scrutiny, the molecular interplay between antidepressants and glutamatergic signaling remains elusive. Using a combination of electrophysiological and single nanoparticle tracking approaches, we here report that the cognitive enhancer and antidepressant tianeptine (S 1574, [3-chloro-6-methyl-5,5-dioxo-6,11-dihydro-(c,f)-dibenzo-(1,2-thiazepine)-11-yl) amino]-7 heptanoic acid, sodium salt) favors synaptic plasticity in hippocampal neurons both under basal conditions and after acute stress. Strikingly, tianeptine rapidly reduces the surface diffusion of AMPA receptor (AMPAR) through a Ca(2+)/calmodulin-dependent protein kinase II (CaMKII)-dependent mechanism that enhances the binding of AMPAR auxiliary subunit stargazin with PSD-95. This prevents corticosterone-induced AMPAR surface dispersal and restores long-term potentiation of acutely stressed mice. Collectively, these data provide the first evidence that a therapeutically used drug targets the surface diffusion of AMPAR through a CaMKII-stargazin-PSD-95 pathway, to promote long-term synaptic plasticity.
Collapse
|
42
|
AMPA receptor/TARP stoichiometry visualized by single-molecule subunit counting. Proc Natl Acad Sci U S A 2013; 110:5163-8. [PMID: 23479622 DOI: 10.1073/pnas.1218765110] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Members of the transmembrane AMPA receptor-regulatory protein (TARP) family modulate AMPA receptor (AMPA-R) trafficking and function. AMPA-Rs consist of four pore-forming subunits. Previous studies show that TARPs are an integral part of the AMPA-R complex, acting as accessory subunits for mature receptors in vivo. The TARP/AMPA-R stoichiometry was previously measured indirectly and found to be variable and dependent on TARP expression level, with at most four TARPs associated with each AMPA-R complex. Here, we use a single-molecule technique in live cells that selectively images proteins located in the plasma membrane to directly count the number of TARPs associated with each AMPA-R complex. Although individual GFP-tagged TARP subunits are observed as freely diffusing fluorescent spots on the surface of Xenopus laevis oocytes when expressed alone, coexpression with AMPA-R-mCherry immobilizes the stargazin-GFP spots at sites of AMPA-R-mCherry, consistent with complex formation. We determined the number of TARP molecules associated with each AMPA-R by counting bleaching steps for three different TARP family members: γ-2, γ-3, and γ-4. We confirm that the TARP/AMPA-R stoichiometry depends on TARP expression level and discover that the maximum number of TARPs per AMPA-R complex falls into two categories: up to four γ-2 or γ-3 subunits, but rarely above two for γ-4 subunit. This unexpected AMPA-R/TARP stoichiometry difference has important implications for the assembly and function of TARP/AMPA-R complexes.
Collapse
|
43
|
Pirotte B, Francotte P, Goffin E, de Tullio P. AMPA receptor positive allosteric modulators: a patent review. Expert Opin Ther Pat 2013; 23:615-28. [DOI: 10.1517/13543776.2013.770840] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
44
|
Nair R, Lauks J, Jung S, Cooke NE, de Wit H, Brose N, Kilimann MW, Verhage M, Rhee J. Neurobeachin regulates neurotransmitter receptor trafficking to synapses. J Cell Biol 2013; 200:61-80. [PMID: 23277425 PMCID: PMC3542797 DOI: 10.1083/jcb.201207113] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2012] [Accepted: 10/26/2012] [Indexed: 12/12/2022] Open
Abstract
The surface density of neurotransmitter receptors at synapses is a key determinant of synaptic efficacy. Synaptic receptor accumulation is regulated by the transport, postsynaptic anchoring, and turnover of receptors, involving multiple trafficking, sorting, motor, and scaffold proteins. We found that neurons lacking the BEACH (beige-Chediak/Higashi) domain protein Neurobeachin (Nbea) had strongly reduced synaptic responses caused by a reduction in surface levels of glutamate and GABA(A) receptors. In the absence of Nbea, immature AMPA receptors accumulated early in the biosynthetic pathway, and mature N-methyl-d-aspartate, kainate, and GABA(A) receptors did not reach the synapse, whereas maturation and surface expression of other membrane proteins, synapse formation, and presynaptic function were unaffected. These data show that Nbea regulates synaptic transmission under basal conditions by targeting neurotransmitter receptors to synapses.
Collapse
Affiliation(s)
- Ramya Nair
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
| | - Juliane Lauks
- Department of Functional Genomics and Department of Clinical Genetics, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit and Vrije Universiteit Medical Center, 1081 HV Amsterdam, Netherlands
| | - SangYong Jung
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
| | - Nancy E. Cooke
- Department of Genetics and Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Heidi de Wit
- Department of Functional Genomics and Department of Clinical Genetics, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit and Vrije Universiteit Medical Center, 1081 HV Amsterdam, Netherlands
| | - Nils Brose
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
| | | | - Matthijs Verhage
- Department of Functional Genomics and Department of Clinical Genetics, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit and Vrije Universiteit Medical Center, 1081 HV Amsterdam, Netherlands
| | - JeongSeop Rhee
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
| |
Collapse
|
45
|
McCullumsmith RE, Hammond J, Funk A, Meador-Woodruff JH. Recent advances in targeting the ionotropic glutamate receptors in treating schizophrenia. Curr Pharm Biotechnol 2012; 13:1535-42. [PMID: 22283761 DOI: 10.2174/138920112800784899] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2010] [Revised: 05/08/2011] [Accepted: 05/24/2011] [Indexed: 12/28/2022]
Abstract
The treatment of schizophrenia has been focused on modulation of dopamine receptors for over 50 years. Recent developments have implicated other neurotransmitter systems in the pathophysiology of this illness. The discovery and characterization of glutamate receptors and their roles in the brain has lead to novel approaches for the treatment of schizophrenia. In this article, we review drugs that modulate ionotropic gluamate receptors and discuss their efficacy for the treatment of this often debilitating severe mental illness.
Collapse
Affiliation(s)
- Robert E McCullumsmith
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham School of Medicine, Birmingham, AL 35294, USA
| | | | | | | |
Collapse
|
46
|
Boudreau AC, Milovanovic M, Conrad KL, Nelson C, Ferrario CR, Wolf ME. A protein cross-linking assay for measuring cell surface expression of glutamate receptor subunits in the rodent brain after in vivo treatments. ACTA ACUST UNITED AC 2012; Chapter 5:Unit 5.30.1-19. [PMID: 22470150 DOI: 10.1002/0471142301.ns0530s59] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Trafficking of neurotransmitter receptors between intracellular and cell surface compartments is important for regulating neurotransmission. We developed a method for determining if an in vivo treatment has altered receptor distribution in a particular region of rodent brain. After the treatment, brain slices are rapidly prepared from the region of interest. Then, cell surface-expressed proteins are covalently cross-linked using the membrane-impermeable, bifunctional cross-linker bis(sulfosuccinimidyl)suberate (BS(3)). This increases the apparent molecular weight of surface receptors, while intracellular receptors are not modified. Thus, surface and intracellular receptor pools can be separated and quantified using SDS-PAGE and immunoblotting. This method is particularly useful for analyzing AMPA receptor subunits, offering advantages in accuracy, efficiency, and cost compared to biotinylation. A disadvantage is that some antibodies no longer recognize their target protein after cross-linking. We have used this method to quantify changes in receptor distribution after acute and chronic exposure to psychomotor stimulants.
Collapse
Affiliation(s)
- Amy C Boudreau
- Department of Neuroscience, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA
| | | | | | | | | | | |
Collapse
|
47
|
Mattison HA, Hayashi T, Barria A. Palmitoylation at two cysteine clusters on the C-terminus of GluN2A and GluN2B differentially control synaptic targeting of NMDA receptors. PLoS One 2012; 7:e49089. [PMID: 23166606 PMCID: PMC3499554 DOI: 10.1371/journal.pone.0049089] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Accepted: 10/09/2012] [Indexed: 11/19/2022] Open
Abstract
Palmitoylation of NMDARs occurs at two distinct cysteine clusters in the carboxyl-terminus of GluN2A and GluN2B subunits that differentially regulates retention in the Golgi apparatus and surface expression of NMDARs. Mutations of palmitoylatable cysteine residues in the membrane-proximal cluster to non-palmitoylatable serines leads to a reduction in the surface expression of recombinant NMDARs via enhanced internalization of the receptors. Mutations in a cluster of cysteines in the middle of the carboxyl-terminus of GluN2A and GluN2B, leads to an increase in the surface expression of NMDARs via an increase in post-Golgi trafficking. Using a quantitative electrophysiological assay, we investigated whether palmitoylation of GluN2 subunits and the differential regulation of surface expression affect functional synaptic incorporation of NMDARs. We show that a reduction in surface expression due to mutations in the membrane-proximal cluster translates to a reduction in synaptic expression of NMDARs. However, increased surface expression induced by mutations in the cluster of cysteines that regulates post-Golgi trafficking of NMDARs does not increase the synaptic pool of NMDA receptors, indicating that the number of synaptic receptors is tightly regulated.
Collapse
Affiliation(s)
- Hayley A. Mattison
- Department of Physiology and Biophysics, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Takashi Hayashi
- Department of Molecular Neurobiology and Pharmacology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Andres Barria
- Department of Physiology and Biophysics, University of Washington School of Medicine, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
48
|
Penn A, Balik A, Wozny C, Cais O, Greger I. Activity-mediated AMPA receptor remodeling, driven by alternative splicing in the ligand-binding domain. Neuron 2012; 76:503-10. [PMID: 23141062 PMCID: PMC3500689 DOI: 10.1016/j.neuron.2012.08.010] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/03/2012] [Indexed: 02/01/2023]
Abstract
The AMPA-type glutamate receptor (AMPAR) subunit composition shapes synaptic transmission and varies throughout development and in response to different input patterns. Here, we show that chronic activity deprivation gives rise to synaptic AMPAR responses with enhanced fidelity. Extrasynaptic AMPARs exhibited changes in kinetics and pharmacology associated with splicing of the alternative flip/flop exons. AMPAR mRNA indeed exhibited reprogramming of the flip/flop exons for GluA1 and GluA2 subunits in response to activity, selectively in the CA1 subfield. However, the functional changes did not directly correlate with the mRNA expression profiles but result from altered assembly of GluA1/GluA2 subunit splice variants, uncovering an additional regulatory role for flip/flop splicing in excitatory signaling. Our results suggest that activity-dependent AMPAR remodeling underlies changes in short-term synaptic plasticity and provides a mechanism for neuronal homeostasis.
Collapse
Affiliation(s)
- Andrew C. Penn
- Neurobiology Division, MRC Laboratory of Molecular Biology, Hills Road, Cambridge CB2 0QH, UK
| | - Ales Balik
- Neurobiology Division, MRC Laboratory of Molecular Biology, Hills Road, Cambridge CB2 0QH, UK
| | - Christian Wozny
- Neurobiology Division, MRC Laboratory of Molecular Biology, Hills Road, Cambridge CB2 0QH, UK
| | - Ondrej Cais
- Neurobiology Division, MRC Laboratory of Molecular Biology, Hills Road, Cambridge CB2 0QH, UK
| | - Ingo H. Greger
- Neurobiology Division, MRC Laboratory of Molecular Biology, Hills Road, Cambridge CB2 0QH, UK
| |
Collapse
|
49
|
Kuang YQ, Charette N, Frazer J, Holland PJ, Attwood KM, Dellaire G, Dupré DJ. Dopamine receptor-interacting protein 78 acts as a molecular chaperone for CCR5 chemokine receptor signaling complex organization. PLoS One 2012; 7:e40522. [PMID: 22815758 PMCID: PMC3398031 DOI: 10.1371/journal.pone.0040522] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2011] [Accepted: 06/11/2012] [Indexed: 11/18/2022] Open
Abstract
Chemokine receptors are members of the G protein-coupled receptor (GPCR) family. CCR5 and CXCR4 act as co-receptors for human immunodeficiency virus (HIV) and several efforts have been made to develop ligands to inhibit HIV infection by blocking those receptors. Removal of chemokine receptors from the cell surface using polymorphisms or other means confers some levels of immunity against HIV infection. Up to now, very limited success has been obtained using ligand therapies so we explored potential avenues to regulate chemokine receptor expression at the plasma membrane. We identified a molecular chaperone, DRiP78, that interacts with both CXCR4 and CCR5, but not the heterodimer formed by these receptors. We further characterized the effects of DRiP78 on CCR5 function. We show that the molecular chaperone inhibits CCR5 localization to the plasma membrane. We identified the interaction region on the receptor, the F(x)6LL motif, and show that upon mutation of this motif the chaperone cannot interact with the receptor. We also show that DRiP78 is involved in the assembly of CCR5 chemokine signaling complex as a homodimer, as well as with the Gαi protein. Finally, modulation of DRiP78 levels will affect receptor functions, such as cell migration in cells that endogenously express CCR5. Our results demonstrate that modulation of the functions of a chaperone can affect signal transduction at the cell surface.
Collapse
Affiliation(s)
- Yi-Qun Kuang
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Nicholle Charette
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Jennifer Frazer
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Patrick J. Holland
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Kathleen M. Attwood
- Department of Pathology, Biochemistry and Molecular Biology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Graham Dellaire
- Department of Pathology, Biochemistry and Molecular Biology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Denis J. Dupré
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
- * E-mail:
| |
Collapse
|
50
|
Hammond JC, Meador-Woodruff JH, Haroutunian V, McCullumsmith RE. AMPA receptor subunit expression in the endoplasmic reticulum in frontal cortex of elderly patients with schizophrenia. PLoS One 2012; 7:e39190. [PMID: 22720072 PMCID: PMC3375247 DOI: 10.1371/journal.pone.0039190] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2011] [Accepted: 05/21/2012] [Indexed: 12/11/2022] Open
Abstract
Several lines of evidence indicate altered trafficking of α-amino-3-hydroxyl-5-methyl-4-isoxazole-propionate (AMPA) receptors in schizophrenia. Previous reports have shown potential changes in the trafficking of AMPA receptors based on subunit expression of endosomes, subcellular organelles located near post-synaptic sites. We hypothesized that alterations in AMPA receptor trafficking through the endoplasmic reticulum (ER) may also be altered in schizophrenia. Accordingly, we developed a technique to isolate and measure content of the ER from postmortem brain tissue. We used Western blot and electron microscopy to show that we isolated an ER enriched fraction. We found no changes in the expression of the AMPA receptor subunits, GluR1–4, in the ER from the dorsolateral prefrontal cortex in schizophrenia. These data suggest that AMPA receptor trafficking through the ER is largely intact in schizophrenia.
Collapse
Affiliation(s)
- John C Hammond
- Department of Neurobiology, University of Alabama Birmingham, Birmingham, Alabama, United States of America.
| | | | | | | |
Collapse
|