1
|
Peterson L, Coca R, Parikh S, McCarthy K, Man HY. ADAR2-mediated Q/R editing of GluA2 in homeostatic synaptic plasticity. Sci Signal 2025; 18:eadr1442. [PMID: 40359260 DOI: 10.1126/scisignal.adr1442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 06/24/2024] [Accepted: 04/22/2025] [Indexed: 05/15/2025]
Abstract
Homeostatic synaptic plasticity is a negative feedback mechanism through which neurons modify their synaptic strength to counteract chronic increases or decreases in activity. In response to activity deprivation, synaptic strength is enhanced by increasing the number of AMPA receptors (AMPARs), particularly Ca2+-permeable AMPARs, at the synapse. Here, we found that this increase in Ca2+-permeable AMPARs during homeostatic upscaling was mediated by decreased posttranscriptional editing of GRIA2 mRNA encoding the AMPAR subunit GluA2. In cultured neurons, activity deprivation resulted in increases in the amount of unedited GluA2, such that its ion channel pore contains a glutamine (Q) codon instead of arginine (R), and in the number of Ca2+-permeable AMPARs at the synapse. These effects were mediated by a splicing factor-dependent decrease in ADAR2 abundance and activity in the nucleus. Overexpression of ADAR2 or CRISPR-Cas13-directed editing of GluA2 transcripts blocked homeostatic upscaling in activity-deprived primary neurons. In mice, dark rearing resulted in decreased Q-to-R editing of GluA2-encoding transcripts in the primary visual cortex (V1), and viral overexpression of ADAR2 in the V1 blocked the induction of homeostatic synaptic plasticity. The findings indicate that activity-dependent regulation of GluA2 editing contributes to homeostatic synaptic plasticity.
Collapse
Affiliation(s)
- Lucy Peterson
- Department of Biology, Boston University, Boston, MA 02215, USA
- Department of Pharmacology, Physiology and Biophysics, Boston University School of Medicine, Boston, MA 02118, USA
| | - Richard Coca
- Department of Biology, Boston University, Boston, MA 02215, USA
- Graduate Program for Neuroscience, Boston University, Boston, MA 02215, USA
| | - Shreya Parikh
- Department of Biology, Boston University, Boston, MA 02215, USA
| | | | - Heng-Ye Man
- Department of Biology, Boston University, Boston, MA 02215, USA
- Department of Pharmacology, Physiology and Biophysics, Boston University School of Medicine, Boston, MA 02118, USA
- Graduate Program for Neuroscience, Boston University, Boston, MA 02215, USA
- Center for Systems Neuroscience, Boston University, Boston, MA 02215, USA
| |
Collapse
|
2
|
Jasińska M, Jasek-Gajda E, Ziaja M, Litwin JA, Lis GJ, Pyza E. Light-Modulated Circadian Synaptic Plasticity in the Somatosensory Cortex: Link to Locomotor Activity. Int J Mol Sci 2024; 25:12870. [PMID: 39684579 DOI: 10.3390/ijms252312870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 11/25/2024] [Accepted: 11/27/2024] [Indexed: 12/18/2024] Open
Abstract
The circadian clock controls various physiological processes, including synaptic function and neuronal activity, affecting the functioning of the entire organism. Light is an important external factor regulating the day-night cycle. This study examined the effects of the circadian clock and light on synaptic plasticity, and explored how locomotor activity contributes to these processes. We analyzed synaptic protein expression and excitatory synapse density in the somatosensory cortex of mice from four groups exposed to different lighting conditions (LD 12:12, DD, LD 16:8, and LL). Locomotor activity was assessed through individual wheel-running monitoring. To explore daily and circadian changes in synaptic proteins, we performed double-immunofluorescence labeling and laser scanning confocal microscopy imaging, targeting three pairs of presynaptic and postsynaptic proteins (Synaptophysin 1/PSD95, Piccolo/Homer 1, Neurexins/PICK1). Excitatory synapse density was evaluated by co-labeling presynaptic and postsynaptic markers. Our results demonstrated that all the analyzed synaptic proteins exhibited circadian regulation modulated by light. Under constant light conditions, only Piccolo and Homer 1 showed rhythmicity. Locomotor activity was also associated with the circadian clock's effects on synaptic proteins, showing a stronger connection to changes in postsynaptic protein levels. Excitatory synapse density peaked during the day/subjective day and exhibited an inverse relationship with locomotor activity. Continued light exposure disrupted cyclic changes in synapse density but kept it consistently elevated. These findings underscore the crucial roles of light and locomotor activity in regulating synaptic plasticity.
Collapse
Affiliation(s)
- Małgorzata Jasińska
- Department of Histology, Jagiellonian University Medical College, 31-034 Krakow, Poland
| | - Ewa Jasek-Gajda
- Department of Histology, Jagiellonian University Medical College, 31-034 Krakow, Poland
| | - Marek Ziaja
- Department of Histology, Jagiellonian University Medical College, 31-034 Krakow, Poland
| | - Jan A Litwin
- Department of Histology, Jagiellonian University Medical College, 31-034 Krakow, Poland
| | - Grzegorz J Lis
- Department of Histology, Jagiellonian University Medical College, 31-034 Krakow, Poland
| | - Elżbieta Pyza
- Department of Cell Biology and Imaging, Institute of Zoology and Biomedical Research, Jagiellonian University, 30-387 Krakow, Poland
| |
Collapse
|
3
|
Eltokhi A, Bertocchi I, Rozov A, Jensen V, Borchardt T, Taylor A, Proenca CC, Rawlins JNP, Bannerman DM, Sprengel R. Distinct effects of AMPAR subunit depletion on spatial memory. iScience 2023; 26:108116. [PMID: 37876813 PMCID: PMC10590979 DOI: 10.1016/j.isci.2023.108116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 07/01/2023] [Accepted: 09/29/2023] [Indexed: 10/26/2023] Open
Abstract
Pharmacological studies established a role for AMPARs in the mammalian forebrain in spatial memory performance. Here we generated global GluA1/3 double knockout mice (Gria1/3-/-) and conditional knockouts lacking GluA1 and GluA3 AMPAR subunits specifically from principal cells across the forebrain (Gria1/3ΔFb). In both models, loss of GluA1 and GluA3 resulted in reduced hippocampal GluA2 and increased levels of the NMDAR subunit GluN2A. Electrically-evoked AMPAR-mediated EPSPs were greatly diminished, and there was an absence of tetanus-induced LTP. Gria1/3-/- mice showed premature mortality. Gria1/3ΔFb mice were viable, and their memory performance could be analyzed. In the Morris water maze (MWM), Gria1/3ΔFb mice showed profound long-term memory deficits, in marked contrast to the normal MWM learning previously seen in single Gria1-/- and Gria3-/- knockout mice. Our results suggest a redundancy of function within the pool of available ionotropic glutamate receptors for long-term spatial memory performance.
Collapse
Affiliation(s)
- Ahmed Eltokhi
- Departments of Molecular Neurobiology and Physiology, Max Planck Institute for Medical Research, Heidelberg, Germany
- Department of Pharmacolog, University of Washington, Seattle, WA, USA
| | - Ilaria Bertocchi
- Departments of Molecular Neurobiology and Physiology, Max Planck Institute for Medical Research, Heidelberg, Germany
- Department of Neuroscience Rita Levi Montalcini, University of Turin, 10126 Turin, Italy
- Neuroscience Institute - Cavalieri-Ottolenghi Foundation (NICO), Laboratory of Neuropsychopharmacology, Regione Gonzole 10, Orbassano, 10043 Torino, Italy
| | - Andrei Rozov
- Departments of Molecular Neurobiology and Physiology, Max Planck Institute for Medical Research, Heidelberg, Germany
- Institute of Neuroscience, Lobachevsky State University of Nizhniy, 603022 Novgorod, Russia
- Federal Center of Brain Research and Neurotechnology, 117997 Moscow, Russia
| | - Vidar Jensen
- Department of Molecular Medicine, Division of Physiology, Institute of Basic Medical Sciences, University of Oslo, 0372 Oslo, Norway
| | - Thilo Borchardt
- Departments of Molecular Neurobiology and Physiology, Max Planck Institute for Medical Research, Heidelberg, Germany
| | - Amy Taylor
- Department of Experimental Psychology, University of Oxford, Oxford, UK
| | - Catia C. Proenca
- Departments of Molecular Neurobiology and Physiology, Max Planck Institute for Medical Research, Heidelberg, Germany
| | | | | | - Rolf Sprengel
- Departments of Molecular Neurobiology and Physiology, Max Planck Institute for Medical Research, Heidelberg, Germany
| |
Collapse
|
4
|
Chiu SL, Chen CM, Huganir RL. ICA69 regulates activity-dependent synaptic strengthening and learning and memory. Front Mol Neurosci 2023; 16:1171432. [PMID: 37251649 PMCID: PMC10213502 DOI: 10.3389/fnmol.2023.1171432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 04/13/2023] [Indexed: 05/31/2023] Open
Abstract
Long-term potentiation (LTP) is one of the major cellular mechanisms for learning and memory. Activity-dependent increases in surface AMPA receptors (AMPARs) are important for enhanced synaptic efficacy during LTP. Here, we report a novel function of a secretory trafficking protein, ICA69, in AMPAR trafficking, synaptic plasticity, and animal cognition. ICA69 is first identified as a diabetes-associated protein well characterized for its function in the biogenesis of secretory vesicles and trafficking of insulin from ER, Golgi to post-Golgi in pancreatic beta cells. In the brain, ICA69 is found in the AMPAR protein complex through its interaction with PICK1, which binds directly to GluA2 or GluA3 AMPAR subunits. Here, we showed that ICA69 regulates PICK1's distribution in neurons and stability in the mouse hippocampus, which in turn can impact AMPAR function in the brain. Biochemical analysis of postsynaptic density (PSD) proteins from hippocampi of mice lacking ICA69 (Ica1 knockout) and their wild-type littermates revealed comparable AMPAR protein levels. Electrophysiological recording and morphological analysis of CA1 pyramidal neurons from Ica1 knockout also showed normal AMPAR-mediated currents and dendrite architecture, indicating that ICA69 does not regulate synaptic AMPAR function and neuron morphology at the basal state. However, genetic deletion of ICA69 in mice selectively impairs NMDA receptor (NMDAR)-dependent LTP but not LTD at Schaffer collateral to CA1 synapses, which correlates with behavioral deficits in tests of spatial and associative learning and memory. Together, we identified a critical and selective role of ICA69 in LTP, linking ICA69-mediated synaptic strengthening to hippocampus-dependent learning and memory.
Collapse
Affiliation(s)
- Shu-Ling Chiu
- Institute of Cellular and Organismic Biology and Neuroscience Program of Academia Sinica (NPAS), Academia Sinica, Taipei, Taiwan
- Solomon H. Snyder Department of Neuroscience and Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Chih-Ming Chen
- Solomon H. Snyder Department of Neuroscience and Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Richard L. Huganir
- Solomon H. Snyder Department of Neuroscience and Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
5
|
Ghane MA, Wei W, Yakout DW, Allen ZD, Miller CL, Dong B, Yang JJ, Fang N, Mabb AM. Arc ubiquitination regulates endoplasmic reticulum-mediated Ca 2+ release and CaMKII signaling. Front Cell Neurosci 2023; 17:1091324. [PMID: 36998269 PMCID: PMC10043188 DOI: 10.3389/fncel.2023.1091324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 02/22/2023] [Indexed: 03/17/2023] Open
Abstract
Synaptic plasticity relies on rapid, yet spatially precise signaling to alter synaptic strength. Arc is a brain enriched protein that is rapidly expressed during learning-related behaviors and is essential for regulating metabotropic glutamate receptor-mediated long-term depression (mGluR-LTD). We previously showed that disrupting the ubiquitination capacity of Arc enhances mGluR-LTD; however, the consequences of Arc ubiquitination on other mGluR-mediated signaling events is poorly characterized. Here we find that pharmacological activation of Group I mGluRs with S-3,5-dihydroxyphenylglycine (DHPG) increases Ca2+ release from the endoplasmic reticulum (ER). Disrupting Arc ubiquitination on key amino acid residues enhances DHPG-induced ER-mediated Ca2+ release. These alterations were observed in all neuronal subregions except secondary branchpoints. Deficits in Arc ubiquitination altered Arc self-assembly and enhanced its interaction with calcium/calmodulin-dependent protein kinase IIb (CaMKIIb) and constitutively active forms of CaMKII in HEK293 cells. Colocalization of Arc and CaMKII was altered in cultured hippocampal neurons, with the notable exception of secondary branchpoints. Finally, disruptions in Arc ubiquitination were found to increase Arc interaction with the integral ER protein Calnexin. These results suggest a previously unknown role for Arc ubiquitination in the fine tuning of ER-mediated Ca2+ signaling that may support mGluR-LTD, which in turn, may regulate CaMKII and its interactions with Arc.
Collapse
Affiliation(s)
- Mohammad A. Ghane
- Neuroscience Institute, Georgia State University, Atlanta, GA, United States
- Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA, United States
| | - Wei Wei
- Neuroscience Institute, Georgia State University, Atlanta, GA, United States
- Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA, United States
| | - Dina W. Yakout
- Neuroscience Institute, Georgia State University, Atlanta, GA, United States
- Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA, United States
| | - Zachary D. Allen
- Neuroscience Institute, Georgia State University, Atlanta, GA, United States
- Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA, United States
| | - Cassandra L. Miller
- Department of Chemistry, Georgia State University, Atlanta, GA, United States
| | - Bin Dong
- Department of Chemistry, Georgia State University, Atlanta, GA, United States
| | - Jenny J. Yang
- Department of Chemistry, Georgia State University, Atlanta, GA, United States
- Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA, United States
| | - Ning Fang
- Department of Chemistry, Georgia State University, Atlanta, GA, United States
| | - Angela M. Mabb
- Neuroscience Institute, Georgia State University, Atlanta, GA, United States
- Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA, United States
| |
Collapse
|
6
|
Jain A, Woolley CS. Mechanisms That Underlie Expression of Estradiol-Induced Excitatory Synaptic Potentiation in the Hippocampus Differ between Males and Females. J Neurosci 2023; 43:1298-1309. [PMID: 36650060 PMCID: PMC9987570 DOI: 10.1523/jneurosci.2080-19.2023] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 01/08/2023] [Accepted: 01/09/2023] [Indexed: 01/19/2023] Open
Abstract
17β-estradiol (E2) is synthesized in the hippocampus of both sexes and acutely potentiates excitatory synapses in each sex. Previously, we found that the mechanisms for initiation of E2-induced synaptic potentiation differ between males and females, including in the molecular signaling involved. Here, we used electrical stimulation and two-photon glutamate uncaging in hippocampal slices from adult male and female rats to investigate whether the downstream consequences of distinct molecular signaling remain different between the sexes or converge to the same mechanism(s) of expression of potentiation. This showed that synaptic activity is necessary for expression of E2-induced potentiation in females but not males, which paralleled a sex-specific requirement in females for calcium-permeable AMPARs (cpAMPARs) to stabilize potentiation. Nonstationary fluctuation analysis of two-photon evoked unitary synaptic currents showed that the postsynaptic component of E2-induced potentiation occurs either through an increase in AMPAR conductance or in nonconductive properties of AMPARs (number of channels × open probability) and never both at the same synapse. In females, most synapses (76%) were potentiated via increased AMPAR conductance, whereas in males, more synapses (60%) were potentiated via an increase in nonconductive AMPAR properties. Inhibition of cpAMPARs eliminated E2-induced synaptic potentiation in females, whereas some synapses in males were unaffected by cpAMPAR inhibition; these synapses in males potentiated exclusively via increased AMPAR nonconductive properties. This sex bias in expression mechanisms of E2-induced synaptic potentiation underscores the concept of latent sex differences in mechanisms of synaptic plasticity in which the same outcome in each sex is achieved through distinct underlying mechanisms.SIGNIFICANCE STATEMENT Estrogens are synthesized in the brains of both sexes and potentiate excitatory synapses to the same degree in each sex. Despite this apparent similarity, the molecular signaling that initiates estrogen-induced synaptic potentiation differs between the sexes. Here we show that these differences extend to the mechanisms of expression of synaptic potentiation and result in distinct patterns of postsynaptic neurotransmitter receptor modulation in each sex. Such latent sex differences, in which the same outcome is achieved through distinct underlying mechanisms in males versus females, indicate that molecular mechanisms targeted for drug development may differ between the sexes even in the absence of an overt sex difference in behavior or disease.
Collapse
Affiliation(s)
- Anant Jain
- Department of Neurobiology, Northwestern University, Evanston, Illinois 60208
| | - Catherine S Woolley
- Department of Neurobiology, Northwestern University, Evanston, Illinois 60208
| |
Collapse
|
7
|
Huerta Sanchez LL, Sankaran M, Li TL, Doan H, Chiu A, Shulman E, Shab G, Kippin TE, Szumlinski KK. Profiling prefrontal cortex protein expression in rats exhibiting an incubation of cocaine craving following short-access self-administration procedures. Front Psychiatry 2023; 13:1031585. [PMID: 36684008 PMCID: PMC9846226 DOI: 10.3389/fpsyt.2022.1031585] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 11/25/2022] [Indexed: 01/05/2023] Open
Abstract
Introduction Incubation of drug-craving refers to a time-dependent increase in drug cue-elicited craving that occurs during protracted withdrawal. Historically, rat models of incubated cocaine craving employed extended-access (typically 6 h/day) intravenous drug self-administration (IV-SA) procedures, although incubated cocaine craving is reported to occur following shorter-access IV-SA paradigms. The notoriously low-throughput of extended-access IV-SA prompted us to determine whether two different short-access IV-SA procedures akin to those in the literature result in qualitatively similar changes in glutamate receptor expression and the activation of downstream signaling molecules within prefrontal cortex (PFC) subregions as those reported previously by our group under 6h-access conditions. Methods For this, adult, male Sprague-Dawley rats were trained to intravenously self-administer cocaine for 2 h/day for 10 consecutive days (2-h model) or for 6 h on day 1 and 2 h/day for the remaining 9 days of training (Mixed model). A sham control group was also included that did not self-administer cocaine. Results On withdrawal day 3 or 30, rats were subjected to a 2-h test of cue-reinforced responding in the absence of cocaine and a time-dependent increase in drug-seeking was observed under both IV-SA procedures. Immunoblotting of brain tissue collected immediately following the cue test session indicated elevated phospho-Akt1, phospho-CaMKII and Homer2a/b expression within the prelimbic subregion of the PFC of cocaine-incubated rats. However, we failed to detect incubation-related changes in Group 1 metabotropic glutamate receptor or ionotropic glutamate receptor subunit expression in either subregion. Discussion These results highlight further a role for Akt1-related signaling within the prelimbic cortex in driving incubated cocaine craving, and provide novel evidence supporting a potential role also for CaMKII-dependent signaling through glutamate receptors in this behavioral phenomenon.
Collapse
Affiliation(s)
- Laura L. Huerta Sanchez
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, Santa Barbara, CA, United States
| | - Mathangi Sankaran
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, Santa Barbara, CA, United States
| | - Taylor L. Li
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, Santa Barbara, CA, United States
| | - Hoa Doan
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, Santa Barbara, CA, United States
| | - Alvin Chiu
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, Santa Barbara, CA, United States
| | - Eleanora Shulman
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, Santa Barbara, CA, United States
| | - Gabriella Shab
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, Santa Barbara, CA, United States
| | - Tod E. Kippin
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, Santa Barbara, CA, United States
- Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA, United States
- Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, CA, United States
| | - Karen K. Szumlinski
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, Santa Barbara, CA, United States
- Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA, United States
- Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, CA, United States
| |
Collapse
|
8
|
Sørensen AT, Rombach J, Gether U, Madsen KL. The Scaffold Protein PICK1 as a Target in Chronic Pain. Cells 2022; 11:1255. [PMID: 35455935 PMCID: PMC9031029 DOI: 10.3390/cells11081255] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 03/23/2022] [Accepted: 03/30/2022] [Indexed: 02/05/2023] Open
Abstract
Well-tolerated and effective drugs for treating chronic pain conditions are urgently needed. Most chronic pain patients are not effectively relieved from their pain and suffer from debilitating drug side effects. This has not only drastic negative consequences for the patients' quality of life, but also constitute an enormous burden on society. It is therefore of great interest to explore new potent targets for effective pain treatment with fewer side effects and without addiction liability. A critical component of chronic pain conditions is central sensitization, which involves the reorganization and strengthening of synaptic transmission within nociceptive pathways. Such changes are considered as maladaptive and depend on changes in the surface expression and signaling of AMPA-type glutamate receptors (AMPARs). The PDZ-domain scaffold protein PICK1 binds the AMPARs and has been suggested to play a key role in these maladaptive changes. In the present paper, we review the regulation of AMPARs by PICK1 and its relation to pain pathology. Moreover, we highlight other pain-relevant PICK1 interactions, and we evaluate various compounds that target PICK1 and have been successfully tested in pain models. Finally, we evaluate the potential on-target side effects of interfering with the action of PICK1 action in CNS and beyond. We conclude that PICK1 constitutes a valid drug target for the treatment of inflammatory and neuropathic pain conditions without the side effects and abuse liability associated with current pain medication.
Collapse
Affiliation(s)
| | | | | | - Kenneth Lindegaard Madsen
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark; (A.T.S.); (J.R.); (U.G.)
| |
Collapse
|
9
|
Zhang X, Wang J, Ran R, Peng Y, Xiao Y. FSC231 alleviates paclitaxel-induced neuralgia by inhibiting the interactions between PICK1 and GluA2 and activates GSK-3β and ERK1/2. Brain Behav 2021; 11:e2380. [PMID: 34582111 PMCID: PMC8613442 DOI: 10.1002/brb3.2380] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 07/27/2021] [Accepted: 08/31/2021] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND FSC231, a PSD-95/DLG/ZO-1 (PDZ) domain inhibitor of protein kinase Cα interacting protein 1 (PICK1), has analgesic effects, but the mechanism remains unclear. METHODS The expression level of PICK1 in dorsal root ganglion (DRG) of rats was changed by vector plasmid, and the effect of PICK1 on paclitaxel (PTL)-induced neuralgia of rats was observed in collaboration with FSC231 treatment. The possible molecular mechanisms were explored by quantitative real-time polymerase chain reaction (qRT-PCR), Western Blot and co-immunoprecipitation (Co-IP) techniques. RESULTS PTL treatment can significantly reduce mechanical withdrawal threshold (MWT), shorten thermal withdrawal latency (TWL), promote DRG inflammation and release of substance P (SP), stimulate PICK1 expression, decrease α-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid receptor 2 (AMPAR, GluA2) level and increase glycogen synthase kinase-3β (GSK-3β) and extracellular regulated protein kinases1/2 (ERK1/2) phosphorylation in rats, while FSC231 treatment can alleviate the above effects induced by PTL. Overexpression of PICK1 can counteract reduced PICK1 level, increased GluA2 level and decreased GSK-3β and ERK1/2 phosphorylation levels caused by FSC231 treatment. The results of Co-IP confirmed the interactions between PICK1 and GluA2. Both FSC231 treatment and silent PICK1 improved PTL-induced MWT reduction, TWL shortening, inflammation, SP release and related gene expression changes, with cumulative effect. CONCLUSION FSC231 activates GSK-3β/ERK1/2 by inhibiting the interaction between PICK1 and GluA2 and alleviates PTL-induced DRG neuralgia in rats.
Collapse
Affiliation(s)
- Xi Zhang
- Department of Anesthesiology, Renmin Hospital, Hubei University of Medicine, Shiyan, P. R. China
| | - Jiagao Wang
- Department of Anesthesiology, Renmin Hospital, Hubei University of Medicine, Shiyan, P. R. China
| | - Ran Ran
- Department of Anesthesiology, Renmin Hospital, Hubei University of Medicine, Shiyan, P. R. China
| | - Yuchuan Peng
- Department of Anesthesiology, Renmin Hospital, Hubei University of Medicine, Shiyan, P. R. China
| | - Yun Xiao
- Department of Anesthesiology, Renmin Hospital, Hubei University of Medicine, Shiyan, P. R. China
| |
Collapse
|
10
|
Dutta P, Bharti P, Kumar J, Maiti S. Role of actin cytoskeleton in the organization and function of ionotropic glutamate receptors. Curr Res Struct Biol 2021; 3:277-289. [PMID: 34766008 PMCID: PMC8569634 DOI: 10.1016/j.crstbi.2021.10.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 09/04/2021] [Accepted: 10/09/2021] [Indexed: 12/22/2022] Open
Abstract
Neural networks with precise connection are compulsory for learning and memory. Various cellular events occur during the genesis of dendritic spines to their maturation, synapse formation, stabilization of the synapse, and proper signal transmission. The cortical actin cytoskeleton and its multiple regulatory proteins are crucial for the above cellular events. The different types of ionotropic glutamate receptors (iGluRs) present on the postsynaptic density (PSD) are also essential for learning and memory. Interaction of the iGluRs in association of their auxiliary proteins with actin cytoskeleton regulated by actin-binding proteins (ABPs) are required for precise long-term potentiation (LTP) and long-term depression (LTD). There has been a quest to understand the mechanistic detail of synapse function involving these receptors with dynamic actin cytoskeleton. A major, emerging area of investigation is the relationship between ABPs and iGluRs in synapse development. In this review we have summarized the current understanding of iGluRs functioning with respect to the actin cytoskeleton, scaffolding proteins, and their regulators. The AMPA, NMDA, Delta and Kainate receptors need the stable underlying actin cytoskeleton to anchor through synaptic proteins for precise synapse formation. The different types of ABPs present in neurons play a critical role in dynamizing/stabilizing the actin cytoskeleton needed for iGluRs function.
Collapse
Affiliation(s)
- Priyanka Dutta
- National Centre for Cell Science, Pune, Maharashtra, 411007, India
| | - Pratibha Bharti
- National Centre for Cell Science, Pune, Maharashtra, 411007, India
| | - Janesh Kumar
- National Centre for Cell Science, Pune, Maharashtra, 411007, India
| | - Sankar Maiti
- Indian Institute of Science Education and Research, Kolkata, 741246, India
| |
Collapse
|
11
|
Sathler MF, Khatri L, Roberts JP, Schmidt IG, Zaytseva A, Kubrusly RCC, Ziff EB, Kim S. Phosphorylation of AMPA receptor subunit GluA1 regulates clathrin-mediated receptor internalization. J Cell Sci 2021; 134:272078. [PMID: 34369573 DOI: 10.1242/jcs.257972] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 07/29/2021] [Indexed: 11/20/2022] Open
Abstract
Synaptic strength is altered during synaptic plasticity by controlling the number of AMPA receptors (AMPARs) at excitatory synapses. During long-term potentiation and synaptic up-scaling, AMPARs are accumulated at synapses to increase synaptic strength. Neuronal activity leads to phosphorylation of AMPAR subunit GluA1 and subsequent elevation of GluA1 surface expression, either by an increase in receptor forward trafficking to the synaptic membrane or a decrease in receptor internalization. However, the molecular pathways underlying GluA1 phosphorylation-induced elevation of surface AMPAR expression are not completely understood. Here, we employ fluorescence recovery after photobleaching (FRAP) to reveal that phosphorylation of GluA1 Serine 845 (S845) predominantly plays a role in receptor internalization than forward trafficking during synaptic plasticity. Notably, internalization of AMPARs depends upon the clathrin adaptor, AP2, which recruits cargo proteins into endocytic clathrin coated pits. In fact, we further reveal that an increase in GluA1 S845 phosphorylation by two distinct forms of synaptic plasticity diminishes the binding of the AP2 adaptor, reducing internalization, and resulting in elevation of GluA1 surface expression. We thus demonstrate a mechanism of GluA1 phosphorylation-regulated clathrin-mediated internalization of AMPARs.
Collapse
Affiliation(s)
- Matheus F Sathler
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, 540 First Avenue, New York, NY 10016, USA.,Department of Biomedical Sciences, 1617 Campus Delivery, Colorado State University, Fort Collins, CO, 80525, USA.,Neuroscience Program, Department of Physiology and Pharmacology, Rua São João Batista, 187, sala 428, Fluminense Federal University, Niterói, RJ, 24020-005, Brazil
| | - Latika Khatri
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, 540 First Avenue, New York, NY 10016, USA
| | | | | | | | - Regina C C Kubrusly
- Neuroscience Program, Department of Physiology and Pharmacology, Rua São João Batista, 187, sala 428, Fluminense Federal University, Niterói, RJ, 24020-005, Brazil
| | - Edward B Ziff
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, 540 First Avenue, New York, NY 10016, USA
| | - Seonil Kim
- Department of Biomedical Sciences, 1617 Campus Delivery, Colorado State University, Fort Collins, CO, 80525, USA.,Molecular, Cellular and Integrative Neurosciences Program
| |
Collapse
|
12
|
Yeung JHY, Walby JL, Palpagama TH, Turner C, Waldvogel HJ, Faull RLM, Kwakowsky A. Glutamatergic receptor expression changes in the Alzheimer's disease hippocampus and entorhinal cortex. Brain Pathol 2021; 31:e13005. [PMID: 34269494 PMCID: PMC8549033 DOI: 10.1111/bpa.13005] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 05/25/2021] [Accepted: 06/21/2021] [Indexed: 11/29/2022] Open
Abstract
Alzheimer's Disease (AD) is the leading form of dementia worldwide. Currently, the pathological mechanisms underlying AD are not well understood. Although the glutamatergic system is extensively implicated in its pathophysiology, there is a gap in knowledge regarding the expression of glutamate receptors in the AD brain. This study aimed to characterize the expression of specific glutamate receptor subunits in post‐mortem human brain tissue using immunohistochemistry and confocal microscopy. Free‐floating immunohistochemistry and confocal laser scanning microscopy were used to quantify the density of glutamate receptor subunits GluA2, GluN1, and GluN2A in specific cell layers of the hippocampal sub‐regions, subiculum, entorhinal cortex, and superior temporal gyrus. Quantification of GluA2 expression in human post‐mortem hippocampus revealed a significant increase in the stratum (str.) moleculare of the dentate gyrus (DG) in AD compared with control. Increased GluN1 receptor expression was found in the str. moleculare and hilus of the DG, str. oriens of the CA2 and CA3, str. pyramidale of the CA2, and str. radiatum of the CA1, CA2, and CA3 subregions and the entorhinal cortex. GluN2A expression was significantly increased in AD compared with control in the str. oriens, str. pyramidale, and str. radiatum of the CA1 subregion. These findings indicate that the expression of glutamatergic receptor subunits shows brain region‐specific changes in AD, suggesting possible pathological receptor functioning. These results provide evidence of specific glutamatergic receptor subunit changes in the AD hippocampus and entorhinal cortex, indicating the requirement for further research to elucidate the pathophysiological mechanisms it entails, and further highlight the potential of glutamatergic receptor subunits as therapeutic targets.
Collapse
Affiliation(s)
- Jason H Y Yeung
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Joshua L Walby
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Thulani H Palpagama
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Clinton Turner
- Department of Anatomical Pathology, LabPlus, Auckland City Hospital, Auckland, New Zealand
| | - Henry J Waldvogel
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Richard L M Faull
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Andrea Kwakowsky
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
13
|
Electroacupuncture Attenuates CFA-Induced Inflammatory Pain by Regulating CaMKII. Neural Plast 2020; 2020:8861994. [PMID: 33488694 PMCID: PMC7790579 DOI: 10.1155/2020/8861994] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 12/05/2020] [Accepted: 12/17/2020] [Indexed: 01/17/2023] Open
Abstract
Ca2+/calmodulin-dependent protein kinase II (CaMKII) is a multifunctional serine/threonine kinase that is ubiquitously distributed in the central and peripheral nervous systems. Moreover, its phosphorylated protein (P-CaMKII) is involved in memory, mood, and pain regulation in the anterior cingulate cortex (ACC). Electroacupuncture (EA) is a traditional Chinese therapeutic technique that can effectively treat chronic inflammatory pain. However, the CaMKII-GluA1 role in EA analgesia in the ACC remains unclear. This study investigated the role of P-CaMKII and P-GluA1 in a mouse model of inflammatory pain induced by complete Freund's adjuvant (CFA). There were increased P-CaMKII and P-GluA1 levels in the ACC. We found that intracerebroventricular injection of KN93, a CaMKII inhibitor, as well as EA stimulation, attenuated complete Freund's adjuvant-induced pain behavior. Further, EA increased pCaMKII-PICK1 complex (abbreviated as C-P complex) levels. Our findings demonstrate that EA inhibits inflammatory pain by inhibiting CaMKII-GluA1 phosphorylation. P-CaMKII is involved in EA analgesia as the pCaMKII-PICK1 complex.
Collapse
|
14
|
Glutamatergic Receptor Trafficking and Delivery: Role of the Exocyst Complex. Cells 2020; 9:cells9112402. [PMID: 33153008 PMCID: PMC7693776 DOI: 10.3390/cells9112402] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/21/2020] [Accepted: 10/23/2020] [Indexed: 12/29/2022] Open
Abstract
Cells comprise several intracellular membrane compartments that allow them to function properly. One of these functions is cargo movement, typically proteins and membranes within cells. These cargoes ride microtubules through vesicles from Golgi and recycling endosomes to the plasma membrane in order to be delivered and exocytosed. In neurons, synaptic functions employ this cargo trafficking to maintain inter-neuronal communication optimally. One of the complexes that oversee vesicle trafficking and tethering is the exocyst. The exocyst is a protein complex containing eight subunits first identified in yeast and then characterized in multicellular organisms. This complex is related to several cellular processes, including cellular growth, division, migration, and morphogenesis, among others. It has been associated with glutamatergic receptor trafficking and tethering into the synapse, providing the molecular machinery to deliver receptor-containing vesicles into the plasma membrane in a constitutive manner. In this review, we discuss the evidence so far published regarding receptor trafficking and the exocyst complex in both basal and stimulated levels, comparing constitutive trafficking and long-term potentiation-related trafficking.
Collapse
|
15
|
Yan Y, Yang H, Xie Y, Ding Y, Kong D, Yu H. Research Progress on Alzheimer's Disease and Resveratrol. Neurochem Res 2020; 45:989-1006. [PMID: 32162143 DOI: 10.1007/s11064-020-03007-0] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 02/27/2020] [Accepted: 03/03/2020] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD), a common irreversible neurodegenerative disease characterized by amyloid-β plaques, neurofibrillary tangles, and changes in tau phosphorylation, is accompanied by memory loss and symptoms of cognitive dysfunction. Increases in disease incidence due to the ageing of the population have placed a great burden on society. To date, the mechanism of AD and the identities of adequate drugs for AD prevention and treatment have eluded the medical community. It has been confirmed that phytochemicals have certain neuroprotective effects against AD. For example, some progress has been made in research on the use of resveratrol, a natural polyphenolic phytochemical, for the prevention and treatment of AD in recent years. Elucidation of the pathogenesis of AD will create a solid foundation for drug treatment. In addition, research on resveratrol, including its mechanism of action, the roles of signalling pathways and its therapeutic targets, will provide new ideas for AD treatment, which is of great significance. In this review, we discuss the possible relationships between AD and the following factors: synapses, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)-type glutamate receptors (AMPARs), silent information regulator 1 (SIRT1), and estrogens. We also discuss the findings of previous studies regarding these relationships in the context of AD treatment and further summarize research progress related to resveratrol treatment.
Collapse
Affiliation(s)
- Yan Yan
- The Department of Epidemiology and Health Statistics, Public Health School of Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Huihuang Yang
- The Department of Epidemiology and Health Statistics, Public Health School of Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Yuxun Xie
- The Department of Epidemiology and Health Statistics, Public Health School of Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Yuanlin Ding
- The Department of Epidemiology and Health Statistics, Public Health School of Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Danli Kong
- The Department of Epidemiology and Health Statistics, Public Health School of Guangdong Medical University, Dongguan, 523808, Guangdong, China.
| | - Haibing Yu
- The Department of Epidemiology and Health Statistics, Public Health School of Guangdong Medical University, Dongguan, 523808, Guangdong, China.
| |
Collapse
|
16
|
Ibrahim RR, Amer RA, Abozeid AA, Elsharaby RM, Shafik NM. Micro RNA 146a gene variant / TNF-α / IL-6 / IL-1 β; A cross-link axis inbetween oxidative stress, endothelial dysfunction and neuro-inflammation in acute ischemic stroke and chronic schizophrenic patients. Arch Biochem Biophys 2020; 679:108193. [DOI: 10.1016/j.abb.2019.108193] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 11/09/2019] [Accepted: 11/12/2019] [Indexed: 12/18/2022]
|
17
|
Simunovic M, Evergren E, Callan-Jones A, Bassereau P. Curving Cells Inside and Out: Roles of BAR Domain Proteins in Membrane Shaping and Its Cellular Implications. Annu Rev Cell Dev Biol 2019; 35:111-129. [DOI: 10.1146/annurev-cellbio-100617-060558] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Many cellular processes rely on precise and timely deformation of the cell membrane. While many proteins participate in membrane reshaping and scission, usually in highly specialized ways, Bin/amphiphysin/Rvs (BAR) domain proteins play a pervasive role, as they not only participate in many aspects of cell trafficking but also are highly versatile membrane remodelers. Subtle changes in the shape and size of the BAR domain can greatly impact the way in which BAR domain proteins interact with the membrane. Furthermore, the activity of BAR domain proteins can be tuned by external physical parameters, and so they behave differently depending on protein surface density, membrane tension, or membrane shape. These proteins can form 3D structures that mold the membrane and alter its liquid properties, even promoting scission under various circumstances.As such, BAR domain proteins have numerous roles within the cell. Endocytosis is among the most highly studied processes in which BAR domain proteins take on important roles. Over the years, a more complete picture has emerged in which BAR domain proteins are tied to almost all intracellular compartments; examples include endosomal sorting and tubular networks in the endoplasmic reticulum and T-tubules. These proteins also have a role in autophagy, and their activity has been linked with cancer. Here, we briefly review the history of BAR domain protein discovery, discuss the mechanisms by which BAR domain proteins induce curvature, and attempt to settle important controversies in the field. Finally, we review BAR domain proteins in the context of a cell, highlighting their emerging roles in cell signaling and organelle shaping.
Collapse
Affiliation(s)
- Mijo Simunovic
- Center for Studies in Physics and Biology, The Rockefeller University, New York, NY 10065, USA
| | - Emma Evergren
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast BT9 7BL, United Kingdom
| | - Andrew Callan-Jones
- Laboratoire Matière et Systèmes Complexes, CNRS UMR 7057, 75205 Paris, France
| | - Patricia Bassereau
- Laboratoire Physico Chimie Curie, CNRS UMR 168, Institut Curie, PSL Research University, 75005 Paris, France
- Sorbonne Université, 75005 Paris, France
| |
Collapse
|
18
|
Reiner A, Levitz J. Glutamatergic Signaling in the Central Nervous System: Ionotropic and Metabotropic Receptors in Concert. Neuron 2019; 98:1080-1098. [PMID: 29953871 DOI: 10.1016/j.neuron.2018.05.018] [Citation(s) in RCA: 407] [Impact Index Per Article: 67.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 04/19/2018] [Accepted: 05/10/2018] [Indexed: 12/28/2022]
Abstract
Glutamate serves as both the mammalian brain's primary excitatory neurotransmitter and as a key neuromodulator to control synapse and circuit function over a wide range of spatial and temporal scales. This functional diversity is decoded by two receptor families: ionotropic glutamate receptors (iGluRs) and metabotropic glutamate receptors (mGluRs). The challenges posed by the complexity and physiological importance of each of these subtypes has limited our appreciation and understanding of how these receptors work in concert. In this review, by comparing both receptor families with a focus on their crosstalk, we argue for a more holistic understanding of neural glutamate signaling.
Collapse
Affiliation(s)
- Andreas Reiner
- Department of Biology and Biotechnology, Ruhr University Bochum, Bochum, Germany.
| | - Joshua Levitz
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
19
|
Christensen NR, Čalyševa J, Fernandes EFA, Lüchow S, Clemmensen LS, Haugaard‐Kedström LM, Strømgaard K. PDZ Domains as Drug Targets. ADVANCED THERAPEUTICS 2019; 2:1800143. [PMID: 32313833 PMCID: PMC7161847 DOI: 10.1002/adtp.201800143] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 03/25/2019] [Indexed: 12/14/2022]
Abstract
Protein-protein interactions within protein networks shape the human interactome, which often is promoted by specialized protein interaction modules, such as the postsynaptic density-95 (PSD-95), discs-large, zona occludens 1 (ZO-1) (PDZ) domains. PDZ domains play a role in several cellular functions, from cell-cell communication and polarization, to regulation of protein transport and protein metabolism. PDZ domain proteins are also crucial in the formation and stability of protein complexes, establishing an important bridge between extracellular stimuli detected by transmembrane receptors and intracellular responses. PDZ domains have been suggested as promising drug targets in several diseases, ranging from neurological and oncological disorders to viral infections. In this review, the authors describe structural and genetic aspects of PDZ-containing proteins and discuss the current status of the development of small-molecule and peptide modulators of PDZ domains. An overview of potential new therapeutic interventions in PDZ-mediated protein networks is also provided.
Collapse
Affiliation(s)
- Nikolaj R. Christensen
- Center for BiopharmaceuticalsDepartment of Drug Design and PharmacologyUniversity of CopenhagenUniversitetsparken 22100CopenhagenDenmark
| | - Jelena Čalyševa
- European Molecular Biology Laboratory (EMBL)Structural and Computational Biology UnitMeyerhofstraße 169117HeidelbergGermany
- EMBL International PhD ProgrammeFaculty of BiosciencesEMBL–Heidelberg UniversityGermany
| | - Eduardo F. A. Fernandes
- Center for BiopharmaceuticalsDepartment of Drug Design and PharmacologyUniversity of CopenhagenUniversitetsparken 22100CopenhagenDenmark
| | - Susanne Lüchow
- Department of Chemistry – BMCUppsala UniversityBox 576SE75123UppsalaSweden
| | - Louise S. Clemmensen
- Center for BiopharmaceuticalsDepartment of Drug Design and PharmacologyUniversity of CopenhagenUniversitetsparken 22100CopenhagenDenmark
| | - Linda M. Haugaard‐Kedström
- Center for BiopharmaceuticalsDepartment of Drug Design and PharmacologyUniversity of CopenhagenUniversitetsparken 22100CopenhagenDenmark
| | - Kristian Strømgaard
- Center for BiopharmaceuticalsDepartment of Drug Design and PharmacologyUniversity of CopenhagenUniversitetsparken 22100CopenhagenDenmark
| |
Collapse
|
20
|
Bissen D, Foss F, Acker-Palmer A. AMPA receptors and their minions: auxiliary proteins in AMPA receptor trafficking. Cell Mol Life Sci 2019; 76:2133-2169. [PMID: 30937469 PMCID: PMC6502786 DOI: 10.1007/s00018-019-03068-7] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 02/12/2019] [Accepted: 03/07/2019] [Indexed: 12/12/2022]
Abstract
To correctly transfer information, neuronal networks need to continuously adjust their synaptic strength to extrinsic stimuli. This ability, termed synaptic plasticity, is at the heart of their function and is, thus, tightly regulated. In glutamatergic neurons, synaptic strength is controlled by the number and function of AMPA receptors at the postsynapse, which mediate most of the fast excitatory transmission in the central nervous system. Their trafficking to, at, and from the synapse, is, therefore, a key mechanism underlying synaptic plasticity. Intensive research over the last 20 years has revealed the increasing importance of interacting proteins, which accompany AMPA receptors throughout their lifetime and help to refine the temporal and spatial modulation of their trafficking and function. In this review, we discuss the current knowledge about the roles of key partners in regulating AMPA receptor trafficking and focus especially on the movement between the intracellular, extrasynaptic, and synaptic pools. We examine their involvement not only in basal synaptic function, but also in Hebbian and homeostatic plasticity. Included in our review are well-established AMPA receptor interactants such as GRIP1 and PICK1, the classical auxiliary subunits TARP and CNIH, and the newest additions to AMPA receptor native complexes.
Collapse
Affiliation(s)
- Diane Bissen
- Institute of Cell Biology and Neuroscience and Buchmann Institute for Molecular Life Sciences (BMLS), University of Frankfurt, Max-von-Laue-Str. 15, 60438, Frankfurt am Main, Germany
- Max Planck Institute for Brain Research, Max von Laue Str. 4, 60438, Frankfurt am Main, Germany
| | - Franziska Foss
- Institute of Cell Biology and Neuroscience and Buchmann Institute for Molecular Life Sciences (BMLS), University of Frankfurt, Max-von-Laue-Str. 15, 60438, Frankfurt am Main, Germany
| | - Amparo Acker-Palmer
- Institute of Cell Biology and Neuroscience and Buchmann Institute for Molecular Life Sciences (BMLS), University of Frankfurt, Max-von-Laue-Str. 15, 60438, Frankfurt am Main, Germany.
- Max Planck Institute for Brain Research, Max von Laue Str. 4, 60438, Frankfurt am Main, Germany.
- Cardio-Pulmonary Institute (CPI), Max-von-Laue-Str. 15, 60438, Frankfurt am Main, Germany.
| |
Collapse
|
21
|
Parkinson GT, Hanley JG. Mechanisms of AMPA Receptor Endosomal Sorting. Front Mol Neurosci 2018; 11:440. [PMID: 30568574 PMCID: PMC6289981 DOI: 10.3389/fnmol.2018.00440] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 11/13/2018] [Indexed: 12/21/2022] Open
Abstract
The regulation of synaptic AMPA receptors (AMPARs) is critical for excitatory synaptic transmission, synaptic plasticity and the consequent formation of neural circuits during brain development and their modification during learning and memory processes. The number of synaptic AMPARs is regulated through endocytosis, exocytosis and endosomal sorting that results in recycling back to the plasma membrane or degradation in the lysosome. Hence, endo-lysosomal sorting is vitally important in maintaining AMPAR expression at the synapse, and the dynamic regulation of these trafficking events is a key component of synaptic plasticity. A reduction in synaptic strength such as in long-term depression (LTD) involves AMPAR sorting to lysosomes to reduce synaptic AMPAR number, whereas long-term potentiation (LTP) involves an increase in AMPAR recycling to increase the number of AMPARs at synapses. Here, we review our current understanding of the endosomal trafficking routes taken by AMPARs, and the mechanisms involved in AMPAR endosomal sorting, focussing on the numerous AMPAR associated proteins that have been implicated in this complex process. We also discuss how these events are dysregulated in brain disorders.
Collapse
Affiliation(s)
- Gabrielle T Parkinson
- Centre for Synaptic Plasticity and School of Biochemistry, University of Bristol, Bristol, United Kingdom
| | - Jonathan G Hanley
- Centre for Synaptic Plasticity and School of Biochemistry, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
22
|
Mansilla A, Jordán-Álvarez S, Santana E, Jarabo P, Casas-Tintó S, Ferrús A. Molecular mechanisms that change synapse number. J Neurogenet 2018; 32:155-170. [DOI: 10.1080/01677063.2018.1506781] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
23
|
Choi B, Lee HW, Mo S, Kim JY, Kim HW, Rhyu IJ, Hong E, Lee YK, Choi JS, Kim CH, Kim H. Inositol 1,4,5-trisphosphate 3-kinase A overexpressed in mouse forebrain modulates synaptic transmission and mGluR-LTD of CA1 pyramidal neurons. PLoS One 2018; 13:e0193859. [PMID: 29617377 PMCID: PMC5884490 DOI: 10.1371/journal.pone.0193859] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 02/20/2018] [Indexed: 11/18/2022] Open
Abstract
Inositol 1,4,5-trisphosphate 3-kinase A (IP3K-A) regulates the level of the inositol polyphosphates, inositol trisphosphate (IP3) and inositol tetrakisphosphate to modulate cellular signaling and intracellular calcium homeostasis in the central nervous system. IP3K-A binds to F-actin in an activity-dependent manner and accumulates in dendritic spines, where it is involved in the regulation of synaptic plasticity. IP3K-A knockout mice exhibit deficits in some forms of hippocampus-dependent learning and synaptic plasticity, such as long-term potentiation in the dentate gyrus synapses of the hippocampus. In the present study, to further elucidate the role of IP3K-A in the brain, we developed a transgenic (Tg) mouse line in which IP3K-A is conditionally overexpressed approximately 3-fold in the excitatory neurons of forebrain regions, including the hippocampus. The Tg mice showed an increase in both presynaptic release probability of evoked responses, along with bigger synaptic vesicle pools, and miniature excitatory postsynaptic current amplitude, although the spine density or the expression levels of the postsynaptic density-related proteins NR2B, synaptotagmin 1, and PSD-95 were not affected. Hippocampal-dependent learning and memory tasks, including novel object recognition and radial arm maze tasks, were partially impaired in Tg mice. Furthermore, (R,S)-3,5-dihydroxyphenylglycine-induced metabotropic glutamate receptor long-term depression was inhibited in Tg mice and this inhibition was dependent on protein kinase C but not on the IP3 receptor. Long-term potentiation and depression dependent on N-methyl-d-aspartate receptor were marginally affected in Tg mice. In summary, this study shows that overexpressed IP3K-A plays a role in some forms of hippocampus-dependent learning and memory tasks as well as in synaptic transmission and plasticity by regulating both presynaptic and postsynaptic functions.
Collapse
Affiliation(s)
- Byungil Choi
- Department of Anatomy, College of Medicine, Korea University, Brain Korea, Seoul, Korea
| | - Hyun Woo Lee
- Department of Anatomy, College of Medicine, Korea University, Brain Korea, Seoul, Korea
| | - Seojung Mo
- Department of Anatomy, College of Medicine, Korea University, Brain Korea, Seoul, Korea
| | - Jin Yong Kim
- Department of Anatomy, College of Medicine, Korea University, Brain Korea, Seoul, Korea
| | - Hyun Wook Kim
- Department of Anatomy, College of Medicine, Korea University, Brain Korea, Seoul, Korea
| | - Im Joo Rhyu
- Department of Anatomy, College of Medicine, Korea University, Brain Korea, Seoul, Korea
| | - Eunhwa Hong
- Department of Psychology, Korea University, Seoul, Korea
| | - Yeon Kyung Lee
- Department of Psychology, Korea University, Seoul, Korea
| | - June-Seek Choi
- Department of Psychology, Korea University, Seoul, Korea
| | - Chong-Hyun Kim
- Center for Neuroscience, Brain Science Institute, Korea Institute of Science and Technology and Neuroscience Program, Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology, Seoul, Korea
- * E-mail: (C-HK); (HK)
| | - Hyun Kim
- Department of Anatomy, College of Medicine, Korea University, Brain Korea, Seoul, Korea
- * E-mail: (C-HK); (HK)
| |
Collapse
|
24
|
Regulation of AMPA receptor trafficking and exit from the endoplasmic reticulum. Mol Cell Neurosci 2018; 91:3-9. [PMID: 29545119 DOI: 10.1016/j.mcn.2018.03.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 03/09/2018] [Accepted: 03/12/2018] [Indexed: 02/06/2023] Open
Abstract
A fundamental property of the brain is its ability to modify its function in response to its own activity. This ability for self-modification depends to a large extent on synaptic plasticity. It is now appreciated that for excitatory synapses, a significant part of synaptic plasticity depends upon changes in the post synaptic response to glutamate released from nerve terminals. Modification of the post synaptic response depends, in turn, on changes in the abundances of AMPA receptors in the post synaptic membrane. In this review, we consider mechanisms of trafficking of AMPA receptors to and from synapses that take place in the early trafficking stages, starting in the endoplasmic reticulum (ER) and continuing into the secretory pathway. We consider mechanisms of AMPA receptor assembly in the ER, highlighting the role of protein synthesis and the selective properties of specific AMPA receptor subunits, as well as regulation of ER exit, including the roles of chaperones and accessory proteins and the incorporation of AMPA receptors into COPII vesicles. We consider these processes in the context of the mechanism of mGluR LTD and discuss a compelling role for the dendritic ER membrane that is found proximal to synapses. The review illustrates the important, yet little studied, contribution of the early stages of AMPA receptor trafficking to synaptic plasticity.
Collapse
|
25
|
PICK1 facilitates lasting reduction in GluA2 concentration in the hippocampus during chronic epilepsy. Epilepsy Res 2017; 137:25-32. [DOI: 10.1016/j.eplepsyres.2017.08.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 07/05/2017] [Accepted: 08/21/2017] [Indexed: 11/18/2022]
|
26
|
Kunde SA, Rademacher N, Zieger H, Shoichet SA. Protein kinase C regulates AMPA receptor auxiliary protein Shisa9/CKAMP44 through interactions with neuronal scaffold PICK1. FEBS Open Bio 2017; 7:1234-1245. [PMID: 28904854 PMCID: PMC5586339 DOI: 10.1002/2211-5463.12261] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 05/19/2017] [Accepted: 06/08/2017] [Indexed: 12/22/2022] Open
Abstract
Synaptic α‐amino‐3‐hydroxyl‐5‐methyl‐4‐isoxazole‐propionate (AMPA) receptors are essential mediators of neurotransmission in the central nervous system. Shisa9/cysteine‐knot AMPAR modulating protein 44 (CKAMP44) is a transmembrane protein recently found to be present in AMPA receptor‐associated protein complexes. Here, we show that the cytosolic tail of Shisa9/CKAMP44 interacts with multiple scaffold proteins that are important for regulating synaptic plasticity in central neurons. We focussed on the interaction with the scaffold protein PICK1, which facilitates the formation of a tripartite complex with the protein kinase C (PKC) and thereby regulates phosphorylation of Shisa9/CKAMP44 C‐terminal residues. This work has implications for our understanding of how PICK1 modulates AMPAR‐mediated transmission and plasticity and also highlights a novel function of PKC.
Collapse
Affiliation(s)
- Stella-Amrei Kunde
- Neuroscience Research Center/Institute of Biochemistry Charité - Universitätsmedizin Berlin Germany
| | - Nils Rademacher
- Neuroscience Research Center/Institute of Biochemistry Charité - Universitätsmedizin Berlin Germany
| | - Hanna Zieger
- Neuroscience Research Center/Institute of Biochemistry Charité - Universitätsmedizin Berlin Germany
| | - Sarah A Shoichet
- Neuroscience Research Center/Institute of Biochemistry Charité - Universitätsmedizin Berlin Germany
| |
Collapse
|
27
|
Pick JE, Khatri L, Sathler MF, Ziff EB. mGluR long-term depression regulates GluA2 association with COPII vesicles and exit from the endoplasmic reticulum. EMBO J 2016; 36:232-244. [PMID: 27856517 DOI: 10.15252/embj.201694526] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 10/09/2016] [Accepted: 10/11/2016] [Indexed: 01/05/2023] Open
Abstract
mGluR long-term depression (mGluR-LTD) is a form of synaptic plasticity induced at excitatory synapses by metabotropic glutamate receptors (mGluRs). mGluR-LTD reduces synaptic strength and is relevant to learning and memory, autism, and sensitization to cocaine; however, the mechanism is not known. Here we show that activation of Group I mGluRs in medium spiny neurons induces trafficking of GluA2 from the endoplasmic reticulum (ER) to the synapse by enhancing GluA2 binding to essential COPII vesicle proteins, Sec23 and Sec13. GluA2 exit from the ER further depends on IP3 and Ryanodine receptor-controlled Ca2+ release as well as active translation. Synaptic insertion of GluA2 is coupled to removal of high-conducting Ca2+-permeable AMPA receptors from synapses, resulting in synaptic depression. This work demonstrates a novel mechanism in which mGluR signals release AMPA receptors rapidly from the ER and couple ER release to GluA2 synaptic insertion and GluA1 removal.
Collapse
Affiliation(s)
- Joseph E Pick
- Department of Biochemistry and Molecular Pharmacology, New York University Langone Medical Center, New York, NY, USA
| | - Latika Khatri
- Department of Biochemistry and Molecular Pharmacology, New York University Langone Medical Center, New York, NY, USA
| | - Matheus F Sathler
- Department of Biochemistry and Molecular Pharmacology, New York University Langone Medical Center, New York, NY, USA.,Department of Pharmacology and Physiology, Fluminense Federal University, Niteroi, Brazil
| | - Edward B Ziff
- Department of Biochemistry and Molecular Pharmacology, New York University Langone Medical Center, New York, NY, USA
| |
Collapse
|
28
|
Multiple faces of protein interacting with C kinase 1 (PICK1): Structure, function, and diseases. Neurochem Int 2016; 98:115-21. [DOI: 10.1016/j.neuint.2016.03.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 03/02/2016] [Accepted: 03/02/2016] [Indexed: 11/19/2022]
|
29
|
Toussaint F, Charbel C, Allen BG, Ledoux J. Vascular CaMKII: heart and brain in your arteries. Am J Physiol Cell Physiol 2016; 311:C462-78. [PMID: 27306369 DOI: 10.1152/ajpcell.00341.2015] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 06/14/2016] [Indexed: 01/02/2023]
Abstract
First characterized in neuronal tissues, the multifunctional calcium/calmodulin-dependent protein kinase II (CaMKII) is a key signaling component in several mammalian biological systems. Its unique capacity to integrate various Ca(2+) signals into different specific outcomes is a precious asset to excitable and nonexcitable cells. Numerous studies have reported roles and mechanisms involving CaMKII in brain and heart tissues. However, corresponding functions in vascular cell types (endothelium and vascular smooth muscle cells) remained largely unexplored until recently. Investigation of the intracellular Ca(2+) dynamics, their impact on vascular cell function, the regulatory processes involved and more recently the spatially restricted oscillatory Ca(2+) signals and microdomains triggered significant interest towards proteins like CaMKII. Heteromultimerization of CaMKII isoforms (four isoforms and several splice variants) expands this kinase's peculiar capacity to decipher Ca(2+) signals and initiate specific signaling processes, and thus controlling cellular functions. The physiological functions that rely on CaMKII are unsurprisingly diverse, ranging from regulating contractile state and cellular proliferation to Ca(2+) homeostasis and cellular permeability. This review will focus on emerging evidence of CaMKII as an essential component of the vascular system, with a focus on the kinase isoform/splice variants and cellular system studied.
Collapse
Affiliation(s)
- Fanny Toussaint
- Research Center, Montreal Heart Institute, Montreal, Quebec, Canada; Department of Molecular and Integrative Physiology, Université de Montréal, Montreal Quebec, Canada
| | - Chimène Charbel
- Research Center, Montreal Heart Institute, Montreal, Quebec, Canada; Department of Pharmacology, Université de Montréal, Montreal Quebec, Canada
| | - Bruce G Allen
- Research Center, Montreal Heart Institute, Montreal, Quebec, Canada; Department of Medicine, Université de Montréal, Montreal Quebec, Canada; and Department of Biochemistry and Molecular Medicine, Université de Montréal, Montreal Quebec, Canada
| | - Jonathan Ledoux
- Research Center, Montreal Heart Institute, Montreal, Quebec, Canada; Department of Medicine, Université de Montréal, Montreal Quebec, Canada; and
| |
Collapse
|
30
|
Sathler MF, Stutz B, Martins RS, Dos Santos Pereira M, Pecinalli NR, Santos LE, Taveira-da-Silva R, Lowe J, de Freitas IG, de Melo Reis RA, Manhães AC, Kubrusly RCC. Single exposure to cocaine impairs aspartate uptake in the pre-frontal cortex via dopamine D1-receptor dependent mechanisms. Neuroscience 2016; 329:326-36. [PMID: 27208619 DOI: 10.1016/j.neuroscience.2016.05.022] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 05/10/2016] [Accepted: 05/12/2016] [Indexed: 11/16/2022]
Abstract
Dopamine and glutamate play critical roles in the reinforcing effects of cocaine. We demonstrated that a single intraperitoneal administration of cocaine induces a significant decrease in [(3)H]-d-aspartate uptake in the pre-frontal cortex (PFC). This decrease is associated with elevated dopamine levels, and requires dopamine D1-receptor signaling (D1R) and adenylyl cyclase activation. The effect was observed within 10min of cocaine administration and lasted for up to 30min. This rapid response is related to D1R-mediated cAMP-mediated activation of PKA and phosphorylation of the excitatory amino acid transporters EAAT1, EAAT2 and EAAT3. We also demonstrated that cocaine exposure increases extracellular d-aspartate, l-glutamate and d-serine in the PFC. Our data suggest that cocaine activates dopamine D1 receptor signaling and PKA pathway to regulate EAATs function and extracellular EAA level in the PFC.
Collapse
Affiliation(s)
- Matheus Figueiredo Sathler
- Laboratório de Neurofarmacologia, Departamento de Fisiologia e Farmacologia, Universidade Federal Fluminense, Niterói, Brazil.
| | - Bernardo Stutz
- Laboratório de Neuroquímica, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil.
| | - Robertta Silva Martins
- Laboratório de Neurofarmacologia, Departamento de Fisiologia e Farmacologia, Universidade Federal Fluminense, Niterói, Brazil.
| | - Maurício Dos Santos Pereira
- Laboratório de Neurofarmacologia, Departamento de Fisiologia e Farmacologia, Universidade Federal Fluminense, Niterói, Brazil; Laboratório de Neurofisiologia Molecular, Departamento de Fisiologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil.
| | - Ney Roner Pecinalli
- Laboratório de Neurofarmacologia, Departamento de Fisiologia e Farmacologia, Universidade Federal Fluminense, Niterói, Brazil.
| | - Luis E Santos
- Laboratório de Neuroquímica, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil.
| | - Rosilane Taveira-da-Silva
- Laboratório de Fisico-Química Biológica Aída Hassón-Voloch Instituto de Biofísica Carlos Chagas Filho Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| | - Jennifer Lowe
- Laboratório de Fisico-Química Biológica Aída Hassón-Voloch Instituto de Biofísica Carlos Chagas Filho Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| | - Isis Grigorio de Freitas
- Laboratório de Neurofarmacologia, Departamento de Fisiologia e Farmacologia, Universidade Federal Fluminense, Niterói, Brazil.
| | - Ricardo Augusto de Melo Reis
- Laboratório de Neuroquímica, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil.
| | - Alex C Manhães
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Centro Biomédico, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| | - Regina C C Kubrusly
- Laboratório de Neurofarmacologia, Departamento de Fisiologia e Farmacologia, Universidade Federal Fluminense, Niterói, Brazil.
| |
Collapse
|
31
|
Abstract
AMPA receptors (AMPARs) are assemblies of four core subunits, GluA1-4, that mediate most fast excitatory neurotransmission. The component subunits determine the functional properties of AMPARs, and the prevailing view is that the subunit composition also determines AMPAR trafficking, which is dynamically regulated during development, synaptic plasticity and in response to neuronal stress in disease. Recently, the subunit dependence of AMPAR trafficking has been questioned, leading to a reappraisal of this field. In this Review, we discuss what is known, uncertain, conjectured and unknown about the roles of the individual subunits, and how they affect AMPAR assembly, trafficking and function under both normal and pathological conditions.
Collapse
|
32
|
Takeuchi Y, Morise J, Morita I, Takematsu H, Oka S. Role of Site-Specific N-Glycans Expressed on GluA2 in the Regulation of Cell Surface Expression of AMPA-Type Glutamate Receptors. PLoS One 2015; 10:e0135644. [PMID: 26271046 PMCID: PMC4535760 DOI: 10.1371/journal.pone.0135644] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 07/23/2015] [Indexed: 12/28/2022] Open
Abstract
The AMPA-type glutamate receptor (AMPAR), which is a tetrameric complex composed of four subunits (GluA1-4) with several combinations, mediates the majority of rapid excitatory synaptic transmissions in the nervous system. Cell surface expression levels of AMPAR modulate synaptic plasticity, which is considered one of the molecular bases for learning and memory formation. To date, a unique trisaccharide (HSO3-3GlcAβ1-3Galβ1-4GlcNAc), human natural killer-1 (HNK-1) carbohydrate, was found expressed specifically on N-linked glycans of GluA2 and regulated the cell surface expression of AMPAR and the spine maturation process. However, evidence that the HNK-1 epitope on N-glycans of GluA2 directly affects these phenomena is lacking. Moreover, it is thought that other N-glycans on GluA2 also have potential roles in the regulation of AMPAR functions. In the present study, using a series of mutants lacking potential N-glycosylation sites (N256, N370, N406, and N413) within GluA2, we demonstrated that the mutant lacking the N-glycan at N370 strongly suppressed the intracellular trafficking of GluA2 from the endoplasmic reticulum (ER) in HEK293 cells. Cell surface expression of GluA1, which is a major subunit of AMPAR in neurons, was also suppressed by co-expression of the GluA2 N370S mutant. The N370S mutant and wild-type GluA2 were co-immunoprecipitated with GluA1, suggesting that N370S was properly associated with GluA1. Moreover, we found that N413 was the main potential site of the HNK-1 epitope that promoted the interaction of GluA2 with N-cadherin, resulting in enhanced cell surface expression of GluA2. The HNK-1 epitope on N-glycan at the N413 of GluA2 was also involved in the cell surface expression of GluA1. Thus, our data suggested that site-specific N-glycans on GluA2 regulate the intracellular trafficking and cell surface expression of AMPAR.
Collapse
Affiliation(s)
- Yusuke Takeuchi
- Department of Biological Chemistry, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Jyoji Morise
- Department of Biological Chemistry, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Ippei Morita
- Department of Biological Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Hiromu Takematsu
- Department of Biological Chemistry, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shogo Oka
- Department of Biological Chemistry, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- * E-mail:
| |
Collapse
|
33
|
Abstract
In the CNS, synapse formation and maturation play crucial roles in the construction and consolidation of neuronal circuits. Neurexin and neuroligin localize on the opposite sides of synaptic membrane and interact with each other to promote the assembly and specialization of synapses. However, the excitatory synapses induced by the neurexin-neuroligin complex are initially immature synapses that lack AMPA receptors. Previously, PICK1 (protein interacting with C kinase 1) was shown to cluster and regulate the synaptic localization of AMPA receptors. Here, we report that during synaptogenesis induced by neurexin in cultured neurons from rat hippocampus, PICK1 recruited AMPA receptors to immature postsynaptic sites. This synaptic recruitment of AMPA receptors depended on the interaction between GluA2 and PICK1, and on the lipid-binding ability of PICK1, but not the interaction between PICK1 and neuroligin. Last, our results demonstrated that the recruitment of GluA2 to synapses could be prevented by ICA69 (islet cell autoantigen 69 kDa), a key binding partner of PICK1. Our study showed that PICK1, being negatively regulated by ICA69, could facilitate synapse maturation.
Collapse
|
34
|
Garcia-Alvarez G, Lu B, Yap KAF, Wong LC, Thevathasan JV, Lim L, Ji F, Tan KW, Mancuso JJ, Tang W, Poon SY, Augustine GJ, Fivaz M. STIM2 regulates PKA-dependent phosphorylation and trafficking of AMPARs. Mol Biol Cell 2015; 26:1141-59. [PMID: 25609091 PMCID: PMC4357513 DOI: 10.1091/mbc.e14-07-1222] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
STIMs (STIM1 and STIM2 in mammals) are transmembrane proteins that reside in the endoplasmic reticulum and regulate store-operated Ca2+ entry. STIM2 mediates cAMP/PKA-dependent phosphorylation of the AMPA receptor subunit GluA1 in excitatory neurons. In addition, STIM2 promotes cAMP-dependent surface delivery of GluA1. STIMs (STIM1 and STIM2 in mammals) are transmembrane proteins that reside in the endoplasmic reticulum (ER) and regulate store-operated Ca2+ entry (SOCE). The function of STIMs in the brain is only beginning to be explored, and the relevance of SOCE in nerve cells is being debated. Here we identify STIM2 as a central organizer of excitatory synapses. STIM2, but not its paralogue STIM1, influences the formation of dendritic spines and shapes basal synaptic transmission in excitatory neurons. We further demonstrate that STIM2 is essential for cAMP/PKA-dependent phosphorylation of the AMPA receptor (AMPAR) subunit GluA1. cAMP triggers rapid migration of STIM2 to ER–plasma membrane (PM) contact sites, enhances recruitment of GluA1 to these ER-PM junctions, and promotes localization of STIM2 in dendritic spines. Both biochemical and imaging data suggest that STIM2 regulates GluA1 phosphorylation by coupling PKA to the AMPAR in a SOCE-independent manner. Consistent with a central role of STIM2 in regulating AMPAR phosphorylation, STIM2 promotes cAMP-dependent surface delivery of GluA1 through combined effects on exocytosis and endocytosis. Collectively our results point to a unique mechanism of synaptic plasticity driven by dynamic assembly of a STIM2 signaling complex at ER-PM contact sites.
Collapse
Affiliation(s)
- Gisela Garcia-Alvarez
- Program in Neuroscience and Behavioral Disorders, DUKE-NUS Graduate Medical School, Singapore 169857
| | - Bo Lu
- Program in Neuroscience and Behavioral Disorders, DUKE-NUS Graduate Medical School, Singapore 169857
| | - Kenrick An Fu Yap
- Program in Neuroscience and Behavioral Disorders, DUKE-NUS Graduate Medical School, Singapore 169857
| | - Loo Chin Wong
- Program in Neuroscience and Behavioral Disorders, DUKE-NUS Graduate Medical School, Singapore 169857
| | - Jervis Vermal Thevathasan
- Program in Neuroscience and Behavioral Disorders, DUKE-NUS Graduate Medical School, Singapore 169857
| | - Lynette Lim
- Program in Neuroscience and Behavioral Disorders, DUKE-NUS Graduate Medical School, Singapore 169857
| | - Fang Ji
- Program in Neuroscience and Behavioral Disorders, DUKE-NUS Graduate Medical School, Singapore 169857
| | - Kia Wee Tan
- Program in Neuroscience and Behavioral Disorders, DUKE-NUS Graduate Medical School, Singapore 169857
| | - James J Mancuso
- Program in Neuroscience and Behavioral Disorders, DUKE-NUS Graduate Medical School, Singapore 169857
| | - Willcyn Tang
- Program in Neuroscience and Behavioral Disorders, DUKE-NUS Graduate Medical School, Singapore 169857
| | - Shou Yu Poon
- Program in Neuroscience and Behavioral Disorders, DUKE-NUS Graduate Medical School, Singapore 169857
| | - George J Augustine
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 637553 Center for Functional Connectomics, Korea Institute of Science and Technology, Seoul 136-791, Republic of Korea
| | - Marc Fivaz
- Program in Neuroscience and Behavioral Disorders, DUKE-NUS Graduate Medical School, Singapore 169857 Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597
| |
Collapse
|
35
|
Gan Q, Salussolia CL, Wollmuth LP. Assembly of AMPA receptors: mechanisms and regulation. J Physiol 2014; 593:39-48. [PMID: 25556786 DOI: 10.1113/jphysiol.2014.273755] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 07/01/2014] [Indexed: 12/16/2022] Open
Abstract
AMPA receptors (AMPARs) play a critical role in excitatory glutamatergic neurotransmission. The number and subunit composition of AMPARs at synapses determines the dynamics of fast glutamatergic signalling. Functional AMPARs on the cell surface are tetramers. Thus tetrameric assembly of AMPARs represents a promising target for modulating AMPAR-mediated signalling in health and disease. Multiple structural domains within the receptor influence AMPAR assembly. In a proposed model for AMPAR assembly, the amino-terminal domain underlies the formation of a dimer pool. The transmembrane domain facilitates the formation and enhances the stability of the tetramer. The ligand-binding domain influences assembly through a process referred to as 'domain swapping'. We propose that this core AMPAR assembly process could be regulated by neuronal signals and speculate on possible mechanisms for such regulation.
Collapse
Affiliation(s)
- Quan Gan
- Graduate Program in Neuroscience, Stony Brook University, Stony Brook, NY, USA; Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY, USA; Department of Neurobiology and Behaviour, Stony Brook University, Stony Brook, NY, USA
| | | | | |
Collapse
|