1
|
Tomczak J, Mackiewicz J, Lisek M, Kaluza A, Boczek T. Exploring AKAPs in visual signaling. Front Mol Neurosci 2024; 17:1412407. [PMID: 38813437 PMCID: PMC11133604 DOI: 10.3389/fnmol.2024.1412407] [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: 04/04/2024] [Accepted: 05/02/2024] [Indexed: 05/31/2024] Open
Abstract
The complex nature of the retina demands well-organized signaling to uphold signal accuracy and avoid interference, a critical aspect in handling a variety of visual stimuli. A-kinase anchoring proteins (AKAPs), known for binding protein kinase A (PKA), contribute to the specificity and efficiency of retinal signaling. They play multifaceted roles in various retinal cell types, influencing photoreceptor sensitivity, neurotransmitter release in bipolar cells, and the integration of visual information in ganglion cells. AKAPs like AKAP79/150 and AKAP95 exhibit distinct subcellular localizations, impacting synaptic transmission and receptor sensitivity in photoreceptors and bipolar cells. Furthermore, AKAPs are involved in neuroprotective mechanisms and axonal degeneration, particularly in retinal ganglion cells. In particular, AKAP6 coordinates stress-specific signaling and promotes neuroprotection following optic nerve injury. As our review underscores the therapeutic potential of targeting AKAP signaling complexes for retinal neuroprotection and enhancement, it acknowledges challenges in developing selective drugs that target complex protein-protein interactions. Overall, this exploration of AKAPs provides valuable insights into the intricacies of retinal signaling, offering a foundation for understanding and potentially addressing retinal disorders.
Collapse
Affiliation(s)
| | | | | | | | - Tomasz Boczek
- Department of Molecular Neurochemistry, Faculty of Health Sciences, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
2
|
Koster KP, Fyke Z, Nguyen TTA, Niqula A, Noriega-González LY, Woolfrey KM, Dell’Acqua ML, Cologna SM, Yoshii A. Akap5 links synaptic dysfunction to neuroinflammatory signaling in a mouse model of infantile neuronal ceroid lipofuscinosis. Front Synaptic Neurosci 2024; 16:1384625. [PMID: 38798824 PMCID: PMC11116793 DOI: 10.3389/fnsyn.2024.1384625] [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: 02/10/2024] [Accepted: 04/23/2024] [Indexed: 05/29/2024] Open
Abstract
Palmitoylation and depalmitoylation represent dichotomic processes by which a labile posttranslational lipid modification regulates protein trafficking and degradation. The depalmitoylating enzyme, palmitoyl-protein thioesterase 1 (PPT1), is associated with the devastating pediatric neurodegenerative condition, infantile neuronal ceroid lipofuscinosis (CLN1). CLN1 is characterized by the accumulation of autofluorescent lysosomal storage material (AFSM) in neurons and robust neuroinflammation. Converging lines of evidence suggest that in addition to cellular waste accumulation, the symptomology of CLN1 corresponds with disruption of synaptic processes. Indeed, loss of Ppt1 function in cortical neurons dysregulates the synaptic incorporation of the GluA1 AMPA receptor (AMPAR) subunit during a type of synaptic plasticity called synaptic scaling. However, the mechanisms causing this aberration are unknown. Here, we used the Ppt1-/- mouse model (both sexes) to further investigate how Ppt1 regulates synaptic plasticity and how its disruption affects downstream signaling pathways. To this end, we performed a palmitoyl-proteomic screen, which provoked the discovery that Akap5 is excessively palmitoylated at Ppt1-/- synapses. Extending our previous data, in vivo induction of synaptic scaling, which is regulated by Akap5, caused an excessive upregulation of GluA1 in Ppt1-/- mice. This synaptic change was associated with exacerbated disease pathology. Furthermore, the Akap5- and inflammation-associated transcriptional regulator, nuclear factor of activated T cells (NFAT), was sensitized in Ppt1-/- cortical neurons. Suppressing the upstream regulator of NFAT activation, calcineurin, with the FDA-approved therapeutic FK506 (Tacrolimus) modestly improved neuroinflammation in Ppt1-/- mice. These findings indicate that the absence of depalmitoylation stifles synaptic protein trafficking and contributes to neuroinflammation via an Akap5-associated mechanism.
Collapse
Affiliation(s)
- Kevin P. Koster
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Zach Fyke
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Thu T. A. Nguyen
- Department of Chemistry, University of Illinois at Chicago, Chicago, IL, United States
| | - Amanda Niqula
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | | | - Kevin M. Woolfrey
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Mark L. Dell’Acqua
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Stephanie M. Cologna
- Department of Chemistry, University of Illinois at Chicago, Chicago, IL, United States
| | - Akira Yoshii
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
- Department of Pediatrics, University of Illinois at Chicago, Chicago, IL, United States
- Department of Neurology, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
3
|
Kang M, Senatore AJ, Naughton H, McTigue M, Beltman RJ, Herppich AA, Pflum MKH, Howe AK. Protein kinase A is a functional component of focal adhesions. J Biol Chem 2024; 300:107234. [PMID: 38552737 PMCID: PMC11044056 DOI: 10.1016/j.jbc.2024.107234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 03/06/2024] [Accepted: 03/17/2024] [Indexed: 04/09/2024] Open
Abstract
Focal adhesions (FAs) form the junction between extracellular matrix (ECM)-bound integrins and the actin cytoskeleton and also transmit signals that regulate cell adhesion, cytoskeletal dynamics, and cell migration. While many of these signals are rooted in reversible tyrosine phosphorylation, phosphorylation of FA proteins on Ser/Thr residues is far more abundant yet its mechanisms and consequences are far less understood. The cAMP-dependent protein kinase (protein kinase A; PKA) has important roles in cell adhesion and cell migration and is both an effector and regulator of integrin-mediated adhesion to the ECM. Importantly, subcellular localization plays a critically important role in specifying PKA function. Here, we show that PKA is present in isolated FA-cytoskeleton complexes and active within FAs in live cells. Furthermore, using kinase-catalyzed biotinylation of isolated FA-cytoskeleton complexes, we identify 53 high-stringency candidate PKA substrates within FAs. From this list, we validate tensin-3 (Tns3)-a well-established molecular scaffold, regulator of cell migration, and a component of focal and fibrillar adhesions-as a novel direct substrate for PKA. These observations identify a new pathway for phospho-regulation of Tns3 and, importantly, establish a new and important niche for localized PKA signaling and thus provide a foundation for further investigation of the role of PKA in the regulation of FA dynamics and signaling.
Collapse
Affiliation(s)
- Mingu Kang
- Department of Pharmacology, Larner College of Medicine, University of Vermont Cancer Center, Burlington, Vermont, USA; Department of Molecular Physiology & Biophysics, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA
| | - Amanda J Senatore
- Department of Pharmacology, Larner College of Medicine, University of Vermont Cancer Center, Burlington, Vermont, USA; Department of Molecular Physiology & Biophysics, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA
| | - Hannah Naughton
- Department of Pharmacology, Larner College of Medicine, University of Vermont Cancer Center, Burlington, Vermont, USA; Department of Molecular Physiology & Biophysics, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA
| | - Madeline McTigue
- Department of Pharmacology, Larner College of Medicine, University of Vermont Cancer Center, Burlington, Vermont, USA; Department of Molecular Physiology & Biophysics, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA
| | - Rachel J Beltman
- Department of Chemistry, Wayne State University, Detroit, Michigan, USA
| | - Andrew A Herppich
- Department of Chemistry, Wayne State University, Detroit, Michigan, USA
| | - Mary Kay H Pflum
- Department of Chemistry, Wayne State University, Detroit, Michigan, USA
| | - Alan K Howe
- Department of Pharmacology, Larner College of Medicine, University of Vermont Cancer Center, Burlington, Vermont, USA; Department of Molecular Physiology & Biophysics, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA.
| |
Collapse
|
4
|
Kang M, Senatore AJ, Naughton H, McTigue M, Beltman RJ, Herppich AA, Pflum MKH, Howe AK. Protein Kinase A is a Functional Component of Focal Adhesions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.08.18.553932. [PMID: 37645771 PMCID: PMC10462105 DOI: 10.1101/2023.08.18.553932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Focal adhesions (FAs) form the junction between extracellular matrix (ECM)-bound integrins and the actin cytoskeleton and also transmit signals that regulate cell adhesion, cytoskeletal dynamics, and cell migration. While many of these signals are rooted in reversible tyrosine phosphorylation, phosphorylation of FA proteins on Ser/Thr residues is far more abundant yet its mechanisms and consequences are far less understood. The cAMP-dependent protein kinase (protein kinase A; PKA) has important roles in cell adhesion and cell migration and is both an effector and regulator of integrin-mediated adhesion to the ECM. Importantly, subcellular localization plays a critically important role in specifying PKA function. Here, we show that PKA is present in isolated FA-cytoskeleton complexes and active within FAs in live cells. Furthermore, using kinase-catalyzed biotinylation of isolated FA-cytoskeleton complexes, we identify fifty-three high-stringency candidate PKA substrates within FAs. From this list, we validate tensin-3 (Tns3) - a well-established molecular scaffold, regulator of cell migration, and component of focal and fibrillar adhesions - as a novel direct substrate for PKA. These observations identify a new pathway for phospho-regulation of Tns3 and, importantly, establish a new and important niche for localized PKA signaling and thus provide a foundation for further investigation of the role of PKA in the regulation of FA dynamics and signaling.
Collapse
|
5
|
Yook Y, Lee KY, Kim E, Lizarazo S, Yu X, Tsai NP. Hyperfunction of post-synaptic density protein 95 promotes seizure response in early-stage aβ pathology. EMBO Rep 2024; 25:1233-1255. [PMID: 38413732 PMCID: PMC10933348 DOI: 10.1038/s44319-024-00090-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 01/24/2024] [Accepted: 01/30/2024] [Indexed: 02/29/2024] Open
Abstract
Accumulation of amyloid-beta (Aβ) can lead to the formation of aggregates that contribute to neurodegeneration in Alzheimer's disease (AD). Despite globally reduced neural activity during AD onset, recent studies have suggested that Aβ induces hyperexcitability and seizure-like activity during the early stages of the disease that ultimately exacerbate cognitive decline. However, the underlying mechanism is unknown. Here, we reveal an Aβ-induced elevation of postsynaptic density protein 95 (PSD-95) in cultured neurons in vitro and in an in vivo AD model using APP/PS1 mice at 8 weeks of age. Elevation of PSD-95 occurs as a result of reduced ubiquitination caused by Akt-dependent phosphorylation of E3 ubiquitin ligase murine-double-minute 2 (Mdm2). The elevation of PSD-95 is consistent with the facilitation of excitatory synapses and the surface expression of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors induced by Aβ. Inhibition of PSD-95 corrects these Aβ-induced synaptic defects and reduces seizure activity in APP/PS1 mice. Our results demonstrate a mechanism underlying elevated seizure activity during early-stage Aβ pathology and suggest that PSD-95 could be an early biomarker and novel therapeutic target for AD.
Collapse
Affiliation(s)
- Yeeun Yook
- Department of Molecular and Integrative Physiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Kwan Young Lee
- Department of Molecular and Integrative Physiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Eunyoung Kim
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Simon Lizarazo
- Department of Molecular and Integrative Physiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Xinzhu Yu
- Department of Molecular and Integrative Physiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Nien-Pei Tsai
- Department of Molecular and Integrative Physiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
| |
Collapse
|
6
|
Amaya-Rodriguez CA, Carvajal-Zamorano K, Bustos D, Alegría-Arcos M, Castillo K. A journey from molecule to physiology and in silico tools for drug discovery targeting the transient receptor potential vanilloid type 1 (TRPV1) channel. Front Pharmacol 2024; 14:1251061. [PMID: 38328578 PMCID: PMC10847257 DOI: 10.3389/fphar.2023.1251061] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 12/14/2023] [Indexed: 02/09/2024] Open
Abstract
The heat and capsaicin receptor TRPV1 channel is widely expressed in nerve terminals of dorsal root ganglia (DRGs) and trigeminal ganglia innervating the body and face, respectively, as well as in other tissues and organs including central nervous system. The TRPV1 channel is a versatile receptor that detects harmful heat, pain, and various internal and external ligands. Hence, it operates as a polymodal sensory channel. Many pathological conditions including neuroinflammation, cancer, psychiatric disorders, and pathological pain, are linked to the abnormal functioning of the TRPV1 in peripheral tissues. Intense biomedical research is underway to discover compounds that can modulate the channel and provide pain relief. The molecular mechanisms underlying temperature sensing remain largely unknown, although they are closely linked to pain transduction. Prolonged exposure to capsaicin generates analgesia, hence numerous capsaicin analogs have been developed to discover efficient analgesics for pain relief. The emergence of in silico tools offered significant techniques for molecular modeling and machine learning algorithms to indentify druggable sites in the channel and for repositioning of current drugs aimed at TRPV1. Here we recapitulate the physiological and pathophysiological functions of the TRPV1 channel, including structural models obtained through cryo-EM, pharmacological compounds tested on TRPV1, and the in silico tools for drug discovery and repositioning.
Collapse
Affiliation(s)
- Cesar A. Amaya-Rodriguez
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
- Departamento de Fisiología y Comportamiento Animal, Facultad de Ciencias Naturales, Exactas y Tecnología, Universidad de Panamá, Ciudad de Panamá, Panamá
| | - Karina Carvajal-Zamorano
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Daniel Bustos
- Centro de Investigación de Estudios Avanzados del Maule (CIEAM), Vicerrectoría de Investigación y Postgrado Universidad Católica del Maule, Talca, Chile
- Laboratorio de Bioinformática y Química Computacional, Departamento de Medicina Traslacional, Facultad de Medicina, Universidad Católica del Maule, Talca, Chile
| | - Melissa Alegría-Arcos
- Núcleo de Investigación en Data Science, Facultad de Ingeniería y Negocios, Universidad de las Américas, Santiago, Chile
| | - Karen Castillo
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
- Centro de Investigación de Estudios Avanzados del Maule (CIEAM), Vicerrectoría de Investigación y Postgrado Universidad Católica del Maule, Talca, Chile
| |
Collapse
|
7
|
Collins KB, Scott JD. Phosphorylation, compartmentalization, and cardiac function. IUBMB Life 2023; 75:353-369. [PMID: 36177749 PMCID: PMC10049969 DOI: 10.1002/iub.2677] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 09/15/2022] [Indexed: 11/08/2022]
Abstract
Protein phosphorylation is a fundamental element of cell signaling. First discovered as a biochemical switch in glycogen metabolism, we now know that this posttranslational modification permeates all aspects of cellular behavior. In humans, over 540 protein kinases attach phosphate to acceptor amino acids, whereas around 160 phosphoprotein phosphatases remove phosphate to terminate signaling. Aberrant phosphorylation underlies disease, and kinase inhibitor drugs are increasingly used clinically as targeted therapies. Specificity in protein phosphorylation is achieved in part because kinases and phosphatases are spatially organized inside cells. A prototypic example is compartmentalization of the cyclic adenosine 3',5'-monophosphate (cAMP)-dependent protein kinase A through association with A-kinase anchoring proteins. This configuration creates autonomous signaling islands where the anchored kinase is constrained in proximity to activators, effectors, and selected substates. This article primarily focuses on A kinase anchoring protein (AKAP) signaling in the heart with an emphasis on anchoring proteins that spatiotemporally coordinate excitation-contraction coupling and hypertrophic responses.
Collapse
Affiliation(s)
- Kerrie B. Collins
- Department of Pharmacology, University of Washington, School of Medicine, 1959 NE Pacific Ave, Seattle WA, 98195
| | - John D. Scott
- Department of Pharmacology, University of Washington, School of Medicine, 1959 NE Pacific Ave, Seattle WA, 98195
| |
Collapse
|
8
|
Sumi T, Harada K. Muscarinic acetylcholine receptor-dependent and NMDA receptor-dependent LTP and LTD share the common AMPAR trafficking pathway. iScience 2023; 26:106133. [PMID: 36866246 PMCID: PMC9972575 DOI: 10.1016/j.isci.2023.106133] [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: 06/20/2022] [Revised: 10/30/2022] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
The forebrain cholinergic system promotes higher brain function in part by signaling through the M1 muscarinic acetylcholine receptor (mAChR). Long-term potentiation (LTP) and long-term depression (LTD) of excitatory synaptic transmission in the hippocampus are also induced by mAChR. An AMPA receptor (AMPAR) trafficking model for hippocampal neurons has been proposed to simulate N-methyl-D-aspartate receptor (NMDAR)-dependent synaptic plasticity in the early phase. In this study, we demonstrated the validity of the hypothesis that the mAChR-dependent LTP/LTD shares a common AMPAR trafficking pathway associated with NMDAR-dependent LTP/LTD. However, unlike NMDAR, Ca2+ influx into the spine cytosol occurs owing to the Ca2+ stored inside the ER and is induced via the activation of inositol 1,4,5-trisphosphate (IP3) receptors during M1 mAChR activation. Moreover, the AMPAR trafficking model implies that alterations in LTP and LTD observed in Alzheimer's disease could be attributed to age-dependent reductions in AMPAR expression levels.
Collapse
Affiliation(s)
- Tomonari Sumi
- Research Institute for Interdisciplinary Science, Okayama University, 3-1-1 Tsushima-Naka, Kita-ku, Okayama 700-8530, Japan
- Department of Chemistry, Faculty of Science, Okayama University, 3-1-1 Tsushima-Naka, Kita-ku, Okayama 700-8530, Japan
- Corresponding author
| | - Kouji Harada
- Department of Computer Science and Engineering, Toyohashi University of Technology, Tempaku-cho, Toyohashi 441-8580, Japan
- Center for IT-Based Education, Toyohashi University of Technology, Tempaku-cho, Toyohashi, 441-8580, Japan
| |
Collapse
|
9
|
Bahouth SW, Nooh MM, Mancarella S. Involvement of SAP97 anchored multiprotein complexes in regulating cardiorenal signaling and trafficking networks. Biochem Pharmacol 2023; 208:115406. [PMID: 36596415 DOI: 10.1016/j.bcp.2022.115406] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 12/26/2022] [Accepted: 12/28/2022] [Indexed: 01/02/2023]
Abstract
SAP97 is a member of the MAGUK family of proteins, but unlike other MAGUK proteins that are selectively expressed in the CNS, SAP97 is also expressed in peripheral organs, like the heart and kidneys. SAP97 has several protein binding cassettes, and this review will describe their involvement in creating SAP97-anchored multiprotein networks. SAP97-anchored networks localized at the inner leaflet of the cell membrane play a major role in trafficking and targeting of membrane G protein-coupled receptors (GPCR), channels, and structural proteins. SAP97 plays a major role in compartmentalizing voltage gated sodium and potassium channels to specific cellular compartments of heart cells. SAP97 undergoes extensive alternative splicing. These splice variants give rise to different SAP97 isoforms that alter its cellular localization, networking, signaling and trafficking effects. Regarding GPCR, SAP97 binds to the β1-adrenergic receptor and recruits AKAP5/PKA and PDE4D8 to create a multiprotein complex that regulates trafficking and signaling of cardiac β1-AR. In the kidneys, SAP97 anchored networks played a role in trafficking of aquaporin-2 water channels. Cardiac specific ablation of SAP97 (SAP97-cKO) resulted in cardiac hypertrophy and failure in aging mice. Similarly, instituting transverse aortic constriction (TAC) in young SAP97 c-KO mice exacerbated TAC-induced cardiac remodeling and dysfunction. These findings highlight a critical role for SAP97 in the pathophysiology of a number of cardiac and renal diseases, suggesting that SAP97 is a relevant target for drug discovery.
Collapse
Affiliation(s)
- Suleiman W Bahouth
- Department of Pharmacology, Addiction Science and Toxicology, The University of Tennessee-Health Sciences Center, Memphis, TN, United States.
| | - Mohammed M Nooh
- Department of Biochemistry, Faculty of Pharmacy Cairo University, Cairo, Egypt and Biochemistry Department, Faculty of Pharmacy, October 6 University, Giza, Egypt
| | - Salvatore Mancarella
- Department of Physiology, The University of Tennessee-Health Sciences Center, Memphis, TN, United States
| |
Collapse
|
10
|
Li JB, Hu XY, Chen MW, Xiong CH, Zhao N, Ge YH, Wang H, Gao XL, Xu NJ, Zhao LX, Yu ZH, Chen HZ, Qiu Y. p85S6K sustains synaptic GluA1 to ameliorate cognitive deficits in Alzheimer's disease. Transl Neurodegener 2023; 12:1. [PMID: 36624510 PMCID: PMC9827685 DOI: 10.1186/s40035-022-00334-w] [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: 07/03/2022] [Accepted: 12/26/2022] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Ribosomal protein S6 kinase 1 (S6K1) is a serine-threonine kinase that has two main isoforms: p70S6K (70-kDa isoform) and p85S6K (85-kDa isoform). p70S6K, with its upstream mammalian target of rapamycin (mTOR), has been shown to be involved in learning and memory and participate in the pathophysiology of Alzheimer's disease (AD). However, the function of p85S6K has long been neglected due to its high similarity to p70S6k. The role of p85S6K in learning and memory is still largely unknown. METHODS We fractionated the postsynaptic densities to illustrate the differential distribution of p85S6K and p70S6K. Coimmunoprecipitation was performed to unveil interactions between p85S6K and the GluA1 subunit of AMPA receptor. The roles of p85S6K in synaptic targeting of GluA1 and learning and memory were evaluated by specific knockdown or overexpression of p85S6K followed by a broad range of methodologies including immunofluorescence, Western blot, in situ proximity ligation assay, morphological staining and behavioral examination. Further, the expression level of p85S6K was measured in brains from AD patients and AD model mice. RESULTS p85S6K, but not p70S6K, was enriched in the postsynaptic densities. Moreover, knockdown of p85S6K resulted in defective spatial and recognition memory. In addition, p85S6K could interact with the GluA1 subunit of AMPA receptor through synapse-associated protein 97 and A-kinase anchoring protein 79/150. Mechanistic studies demonstrated that p85S6K could directly phosphorylate GluA1 at Ser845 and increase the amount of GluA1 in synapses, thus sustaining synaptic function and spine densities. Moreover, p85S6K was found to be specifically decreased in the synaptosomal compartment in the brains of AD patients and AD mice. Overexpression of p85S6K ameliorated the synaptic deficits and cognitive impairment in transgenic AD model mice. CONCLUSIONS These results strongly imply a significant role for p85S6K in maintaining synaptic and cognitive function by interacting with GluA1. The findings provide an insight into the rational targeting of p85S6K as a therapeutic potential for AD.
Collapse
Affiliation(s)
- Jia-Bing Li
- grid.16821.3c0000 0004 0368 8293Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Xiao-Yu Hu
- grid.16821.3c0000 0004 0368 8293Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Mu-Wen Chen
- grid.16821.3c0000 0004 0368 8293Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Cai-Hong Xiong
- grid.16821.3c0000 0004 0368 8293Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Na Zhao
- grid.16821.3c0000 0004 0368 8293Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Yan-Hui Ge
- grid.16821.3c0000 0004 0368 8293Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Hao Wang
- grid.16821.3c0000 0004 0368 8293Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Xiao-Ling Gao
- grid.16821.3c0000 0004 0368 8293Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Nan-Jie Xu
- grid.16821.3c0000 0004 0368 8293Collaborative Innovation Center for Brain Science, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Lan-Xue Zhao
- grid.16821.3c0000 0004 0368 8293Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Zhi-Hua Yu
- grid.16821.3c0000 0004 0368 8293Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Hong-Zhuan Chen
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China. .,Institute of Interdisciplinary Integrative Biomedical Research, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201210, China.
| | - Yu Qiu
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
11
|
Zhang Y, Jeske NA. A-kinase anchoring protein 79/150 coordinates α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor sensitization in sensory neurons. Mol Pain 2023; 19:17448069231222406. [PMID: 38073552 PMCID: PMC10722943 DOI: 10.1177/17448069231222406] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 11/30/2023] [Accepted: 11/30/2023] [Indexed: 12/17/2023] Open
Abstract
Changes in sensory afferent activity contribute to the transition from acute to chronic pain. However, it is unlikely that a single sensory receptor is entirely responsible for persistent pain. It is more probable that extended changes to multiple receptor proteins expressed by afferent neurons support persistent pain. A-Kinase Anchoring Protein 79/150 (AKAP) is an intracellular scaffolding protein expressed in sensory neurons that spatially and temporally coordinates signaling events. Since AKAP scaffolds biochemical modifications of multiple TRP receptors linked to pain phenotypes, we probed for other ionotropic receptors that may be mediated by AKAP and contribute to persistent pain. Here, we identify a role for AKAP modulation of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid Receptor (AMPA-R) functionality in sensory neurons. Pharmacological manipulation of distinct AMPA-R subunits significantly reduces persistent mechanical hypersensitivity observed during hyperalgesic priming. Stimulation of both protein kinases C and A (PKC, PKA, respectively) modulate AMPA-R subunit GluR1 and GluR2 phosphorylation and surface expression in an AKAP-dependent manner in primary cultures of DRG neurons. Furthermore, AKAP knock out reduces sensitized AMPA-R responsivity in DRG neurons. Collectively, these data indicate that AKAP scaffolds AMPA-R subunit organization in DRG neurons that may contribute to the transition from acute-to-chronic pain.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Oral and Maxillofacial Surgery, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Nathaniel A Jeske
- Department of Oral and Maxillofacial Surgery, University of Texas Health San Antonio, San Antonio, TX, USA
- Department of Pharmacology, University of Texas Health San Antonio, San Antonio, TX, USA
- Department of Physiology, University of Texas Health San Antonio, San Antonio, TX, USA
| |
Collapse
|
12
|
Chen X, Crosby KC, Feng A, Purkey AM, Aronova MA, Winters CA, Crocker VT, Leapman RD, Reese TS, Dell’Acqua ML. Palmitoylation of A-kinase anchoring protein 79/150 modulates its nanoscale organization, trafficking, and mobility in postsynaptic spines. Front Synaptic Neurosci 2022; 14:1004154. [PMID: 36186623 PMCID: PMC9521714 DOI: 10.3389/fnsyn.2022.1004154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 08/23/2022] [Indexed: 11/29/2022] Open
Abstract
A-kinase anchoring protein 79-human/150-rodent (AKAP79/150) organizes signaling proteins to control synaptic plasticity. AKAP79/150 associates with the plasma membrane and endosomes through its N-terminal domain that contains three polybasic regions and two Cys residues that are reversibly palmitoylated. Mutations abolishing palmitoylation (AKAP79/150 CS) reduce its endosomal localization and association with the postsynaptic density (PSD). Here we combined advanced light and electron microscopy (EM) to characterize the effects of AKAP79/150 palmitoylation on its postsynaptic nanoscale organization, trafficking, and mobility in hippocampal neurons. Immunogold EM revealed prominent extrasynaptic membrane AKAP150 labeling with less labeling at the PSD. The label was at greater distances from the spine membrane for AKAP150 CS than WT in the PSD but not in extra-synaptic locations. Immunogold EM of GFP-tagged AKAP79 WT showed that AKAP79 adopts a vertical, extended conformation at the PSD with its N-terminus at the membrane, in contrast to extrasynaptic locations where it adopts a compact or open configurations of its N- and C-termini with parallel orientation to the membrane. In contrast, GFP-tagged AKAP79 CS was displaced from the PSD coincident with disruption of its vertical orientation, while proximity and orientation with respect to the extra-synaptic membrane was less impacted. Single-molecule localization microscopy (SMLM) revealed a heterogeneous distribution of AKAP150 with distinct high-density, nano-scale regions (HDRs) overlapping the PSD but more prominently located in the extrasynaptic membrane for WT and the CS mutant. Thick section scanning transmission electron microscopy (STEM) tomography revealed AKAP150 immunogold clusters similar in size to HDRs seen by SMLM and more AKAP150 labeled endosomes in spines for WT than for CS, consistent with the requirement for AKAP palmitoylation in endosomal trafficking. Hidden Markov modeling of single molecule tracking data revealed a bound/immobile fraction and two mobile fractions for AKAP79 in spines, with the CS mutant having shorter dwell times and faster transition rates between states than WT, suggesting that palmitoylation stabilizes individual AKAP molecules in various spine subpopulations. These data demonstrate that palmitoylation fine tunes the nanoscale localization, mobility, and trafficking of AKAP79/150 in dendritic spines, which might have profound effects on its regulation of synaptic plasticity.
Collapse
Affiliation(s)
- Xiaobing Chen
- Laboratory of Neurobiology, National Institute of Neurological Diseases and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, MD, United States
- *Correspondence: Xiaobing Chen,
| | - Kevin C. Crosby
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, United States
| | - Austin Feng
- Laboratory of Neurobiology, National Institute of Neurological Diseases and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Alicia M. Purkey
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, United States
| | - Maria A. Aronova
- Laboratory of Cellular Imaging and Macromolecular Biophysics, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Christine A. Winters
- Laboratory of Neurobiology, National Institute of Neurological Diseases and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Virginia T. Crocker
- Laboratory of Neurobiology, National Institute of Neurological Diseases and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Richard D. Leapman
- Laboratory of Cellular Imaging and Macromolecular Biophysics, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Thomas S. Reese
- Laboratory of Neurobiology, National Institute of Neurological Diseases and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Mark L. Dell’Acqua
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, United States
- Mark L. Dell’Acqua,
| |
Collapse
|
13
|
Chapman CA, Nuwer JL, Jacob TC. The Yin and Yang of GABAergic and Glutamatergic Synaptic Plasticity: Opposites in Balance by Crosstalking Mechanisms. Front Synaptic Neurosci 2022; 14:911020. [PMID: 35663370 PMCID: PMC9160301 DOI: 10.3389/fnsyn.2022.911020] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 04/26/2022] [Indexed: 01/12/2023] Open
Abstract
Synaptic plasticity is a critical process that regulates neuronal activity by allowing neurons to adjust their synaptic strength in response to changes in activity. Despite the high proximity of excitatory glutamatergic and inhibitory GABAergic postsynaptic zones and their functional integration within dendritic regions, concurrent plasticity has historically been underassessed. Growing evidence for pathological disruptions in the excitation and inhibition (E/I) balance in neurological and neurodevelopmental disorders indicates the need for an improved, more "holistic" understanding of synaptic interplay. There continues to be a long-standing focus on the persistent strengthening of excitation (excitatory long-term potentiation; eLTP) and its role in learning and memory, although the importance of inhibitory long-term potentiation (iLTP) and depression (iLTD) has become increasingly apparent. Emerging evidence further points to a dynamic dialogue between excitatory and inhibitory synapses, but much remains to be understood regarding the mechanisms and extent of this exchange. In this mini-review, we explore the role calcium signaling and synaptic crosstalk play in regulating postsynaptic plasticity and neuronal excitability. We examine current knowledge on GABAergic and glutamatergic synapse responses to perturbances in activity, with a focus on postsynaptic plasticity induced by short-term pharmacological treatments which act to either enhance or reduce neuronal excitability via ionotropic receptor regulation in neuronal culture. To delve deeper into potential mechanisms of synaptic crosstalk, we discuss the influence of synaptic activity on key regulatory proteins, including kinases, phosphatases, and synaptic structural/scaffolding proteins. Finally, we briefly suggest avenues for future research to better understand the crosstalk between glutamatergic and GABAergic synapses.
Collapse
Affiliation(s)
| | | | - Tija C. Jacob
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
14
|
Neurodevelopmental Disorders Associated with PSD-95 and Its Interaction Partners. Int J Mol Sci 2022; 23:ijms23084390. [PMID: 35457207 PMCID: PMC9025546 DOI: 10.3390/ijms23084390] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/13/2022] [Accepted: 04/14/2022] [Indexed: 01/17/2023] Open
Abstract
The postsynaptic density (PSD) is a massive protein complex, critical for synaptic strength and plasticity in excitatory neurons. Here, the scaffolding protein PSD-95 plays a crucial role as it organizes key PSD components essential for synaptic signaling, development, and survival. Recently, variants in DLG4 encoding PSD-95 were found to cause a neurodevelopmental disorder with a variety of clinical features including intellectual disability, developmental delay, and epilepsy. Genetic variants in several of the interaction partners of PSD-95 are associated with similar phenotypes, suggesting that deficient PSD-95 may affect the interaction partners, explaining the overlapping symptoms. Here, we review the transmembrane interaction partners of PSD-95 and their association with neurodevelopmental disorders. We assess how the structural changes induced by DLG4 missense variants may disrupt or alter such protein-protein interactions, and we argue that the pathological effect of DLG4 variants is, at least partly, exerted indirectly through interaction partners of PSD-95. This review presents a direction for functional studies to elucidate the pathogenic mechanism of deficient PSD-95, providing clues for therapeutic strategies.
Collapse
|
15
|
Ferré S, Ciruela F, Dessauer CW, González-Maeso J, Hébert TE, Jockers R, Logothetis DE, Pardo L. G protein-coupled receptor-effector macromolecular membrane assemblies (GEMMAs). Pharmacol Ther 2022; 231:107977. [PMID: 34480967 PMCID: PMC9375844 DOI: 10.1016/j.pharmthera.2021.107977] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 08/11/2021] [Accepted: 08/17/2021] [Indexed: 12/12/2022]
Abstract
G protein-coupled receptors (GPCRs) are the largest group of receptors involved in cellular signaling across the plasma membrane and a major class of drug targets. The canonical model for GPCR signaling involves three components - the GPCR, a heterotrimeric G protein and a proximal plasma membrane effector - that have been generally thought to be freely mobile molecules able to interact by 'collision coupling'. Here, we synthesize evidence that supports the existence of GPCR-effector macromolecular membrane assemblies (GEMMAs) comprised of specific GPCRs, G proteins, plasma membrane effector molecules and other associated transmembrane proteins that are pre-assembled prior to receptor activation by agonists, which then leads to subsequent rearrangement of the GEMMA components. The GEMMA concept offers an alternative and complementary model to the canonical collision-coupling model, allowing more efficient interactions between specific signaling components, as well as the integration of the concept of GPCR oligomerization as well as GPCR interactions with orphan receptors, truncated GPCRs and other membrane-localized GPCR-associated proteins. Collision-coupling and pre-assembled mechanisms are not exclusive and likely both operate in the cell, providing a spectrum of signaling modalities which explains the differential properties of a multitude of GPCRs in their different cellular environments. Here, we explore the unique pharmacological characteristics of individual GEMMAs, which could provide new opportunities to therapeutically modulate GPCR signaling.
Collapse
Affiliation(s)
- Sergi Ferré
- Integrative Neurobiology Section, National Institute on Drug Addiction, Intramural Research Program, NIH, DHHS, Baltimore, MD, USA.
| | - Francisco Ciruela
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Institute of Neurosciences, IDIBELL, University of Barcelona, L’Hospitalet de Llobregat, Spain
| | - Carmen W. Dessauer
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Javier González-Maeso
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Terence E. Hébert
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec
| | - Ralf Jockers
- University of Paris, Institute Cochin, INSERM, CNRS, Paris, France
| | - Diomedes E. Logothetis
- Laboratory of Electrophysiology, Departments of Pharmaceutical Sciences, Chemistry and Chemical Biology and Center for Drug Discovery, School of Pharmacy at the Bouvé College of Health Sciences and College of Science, Northeastern University, Boston, Massachusetts, USA
| | - Leonardo Pardo
- Laboratory of Computational Medicine, Biostatistics Unit, Faculty of Medicine, Autonomous University of Barcelona, Bellaterra, Spain
| |
Collapse
|
16
|
Welch MA, Jansen LAR, Baro DJ. SUMOylation of the Kv4.2 Ternary Complex Increases Surface Expression and Current Amplitude by Reducing Internalization in HEK 293 Cells. Front Mol Neurosci 2021; 14:757278. [PMID: 34795560 PMCID: PMC8593141 DOI: 10.3389/fnmol.2021.757278] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 10/05/2021] [Indexed: 11/29/2022] Open
Abstract
Kv4 α-subunits exist as ternary complexes (TC) with potassium channel interacting proteins (KChIP) and dipeptidyl peptidase-like proteins (DPLP); multiple ancillary proteins also interact with the α-subunits throughout the channel’s lifetime. Dynamic regulation of Kv4.2 protein interactions adapts the transient potassium current, IA, mediated by Kv4 α-subunits. Small ubiquitin-like modifier (SUMO) is an 11 kD peptide post-translationally added to lysine (K) residues to regulate protein–protein interactions. We previously demonstrated that when expressed in human embryonic kidney (HEK) cells, Kv4.2 can be SUMOylated at two K residues, K437 and K579. SUMOylation at K437 increased surface expression of electrically silent channels while SUMOylation at K579 reduced IA maximal conductance (Gmax) without altering surface expression. KChIP and DPLP subunits are known to modify the pattern of Kv4.2 post-translational decorations and/or their effects. In this study, co-expressing Kv4.2 with KChIP2a and DPP10c altered the effects of enhanced Kv4.2 SUMOylation. First, the effect of enhanced SUMOylation was the same for a TC containing either the wild-type Kv4.2 or the mutant K437R Kv4.2, suggesting that either the experimental manipulation no longer enhanced K437 SUMOylation or K437 SUMOylation no longer influenced Kv4.2 surface expression. Second, instead of decreasing IA Gmax, enhanced SUMOylation at K579 now produced a significant ∼37–70% increase in IA maximum conductance (Gmax) and a significant ∼30–50% increase in Kv4.2g surface expression that was accompanied by a 65% reduction in TC internalization. Blocking clathrin-mediated endocytosis (CME) in HEK cells expressing the Kv4.2 TC mimicked and occluded the effect of SUMO on IA Gmax; however, the amount of Kv4.2 associated with the major adaptor for constitutive CME, adaptor protein 2 (AP2), was not SUMO dependent. Thus, SUMOylation reduced Kv4.2 internalization by acting downstream of Kv4.2 recruitment into clathrin-coated pits. In sum, the two major findings of this study are: SUMOylation of Kv4.2 at K579 regulates TC internalization most likely by promoting channel recycling. Additionally, there is a reciprocity between Kv4.2 SUMOylation and the Kv4.2 interactome such that SUMOylation regulates the interactome and the interactome influences the pattern and effect of SUMOylation.
Collapse
Affiliation(s)
- Meghyn A Welch
- Department of Biology, Georgia State University, Atlanta, GA, United States
| | | | - Deborah J Baro
- Department of Biology, Georgia State University, Atlanta, GA, United States.,Neuroscience Institute, Georgia State University, Atlanta, GA, United States
| |
Collapse
|
17
|
Chaklader M, Rothermel BA. Calcineurin in the heart: New horizons for an old friend. Cell Signal 2021; 87:110134. [PMID: 34454008 PMCID: PMC8908812 DOI: 10.1016/j.cellsig.2021.110134] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 08/10/2021] [Accepted: 08/23/2021] [Indexed: 01/20/2023]
Abstract
Calcineurin, also known as PP2B or PPP3, is a member of the PPP family of protein phosphatases that also includes PP1 and PP2A. Together these three phosphatases carryout the majority of dephosphorylation events in the heart. Calcineurin is distinct in that it is activated by the binding of calcium/calmodulin (Ca2+/CaM) and therefore acts as a node for integrating Ca2+ signals with changes in phosphorylation, two fundamental intracellular signaling cascades. In the heart, calcineurin is primarily thought of in the context of pathological cardiac remodeling, acting through the Nuclear Factor of Activated T-cell (NFAT) family of transcription factors. However, calcineurin activity is also essential for normal heart development and homeostasis in the adult heart. Furthermore, it is clear that NFAT-driven changes in transcription are not the only relevant processes initiated by calcineurin in the setting of pathological remodeling. There is a growing appreciation for the diversity of calcineurin substrates that can impact cardiac function as well as the diversity of mechanisms for targeting calcineurin to specific sub-cellular domains in cardiomyocytes and other cardiac cell types. Here, we will review the basics of calcineurin structure, regulation, and function in the context of cardiac biology. Particular attention will be given to: the development of improved tools to identify and validate new calcineurin substrates; recent studies identifying new calcineurin isoforms with unique properties and targeting mechanisms; and the role of calcineurin in cardiac development and regeneration.
Collapse
Affiliation(s)
- Malay Chaklader
- Departments of Internal Medicine (Division of Cardiology) and Molecular Biology, University of Texas Southwestern Medical Centre, Dallas, TX, USA
| | - Beverly A Rothermel
- Departments of Internal Medicine (Division of Cardiology) and Molecular Biology, University of Texas Southwestern Medical Centre, Dallas, TX, USA.
| |
Collapse
|
18
|
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: 1.0] [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
|
19
|
AKAP150 and its Palmitoylation Contributed to Pain Hypersensitivity Via Facilitating Synaptic Incorporation of GluA1-Containing AMPA Receptor in Spinal Dorsal Horn. Mol Neurobiol 2021; 58:6505-6519. [PMID: 34559357 DOI: 10.1007/s12035-021-02570-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 09/14/2021] [Indexed: 10/20/2022]
Abstract
The A-kinase anchoring protein 150 (AKAP150) organizes kinases and phosphatases to regulate AMPA receptors (AMPARs) that are pivotal for synaptic plasticity. AKAP150 itself undergoes S-palmitoylation. However, the roles of AKAP150 and its palmitoylation in spinal nociceptive processing remain unknown. In this study, we found that intraplantar injection of complete Freund's adjuvant (CFA) significantly increased the synaptic expression of AKAP150 and caused a reorganization of AKAP150 signaling complex in spinal dorsal horn. Knockdown of AKAP150 or interruption of its interactions with kinases effectively suppressed the CFA-induced synaptic expression of GluA1 subunit of AMPARs. Our data also showed that an upregulation of AKAP150 palmitoylation was involved in the synaptic redistribution of AKAP150. Inhibition of AKAP150 palmitoylation by expression of palmitoylation-defective mutant AKAP150 (C36, 123S) effectively repressed the CFA-induced phosphorylation and synaptic expression of GluA1 subunit, meanwhile, attenuated the development of mechanical allodynia and thermal hyperalgesia. Furthermore, we found that an increased expression of palmitoyl acyltransferase ZDHHC2 contributed to the upregulation of AKAP150 palmitoylation and GluA1 accumulation in inflamed mouse. These data indicated that AKAP150 and its palmitoylation were involved in AMPA receptor-dependent modification of nociceptive transmission, and the manipulations of AKAP150 signaling complex and palmitoylation might serve as potential therapeutic strategies for persistent pain after inflammation.
Collapse
|
20
|
Ge Y, Wang YT. GluA1-homomeric AMPA receptor in synaptic plasticity and neurological diseases. Neuropharmacology 2021; 197:108708. [PMID: 34274350 DOI: 10.1016/j.neuropharm.2021.108708] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/28/2021] [Accepted: 07/08/2021] [Indexed: 11/17/2022]
Abstract
Synaptic transmission is one of the fundamental processes that all brain functions are based on. Changes in the strength of synaptic transmission among neurons are crucial for information processing in the central nervous system. The α-amino-3-hydroxy-5-methylisoxazole-4-propionic acid subtype of ionotropic glutamate receptors (AMPARs) mediate the majority of the fast excitatory synaptic transmission in the mammalian brain. Rapid trafficking of AMPARs in and out of the postsynaptic membrane is proposed to be a major mechanism for synaptic plasticity, and learning and memory. Defects in the regulated AMPAR trafficking have been shown to be involved in the pathogenesis of certain psychiatric and neurodegenerative diseases. Studies accumulated in the past 30 years have provided a detailed molecular insight on how the trafficking of AMPARs is modulated in a subunit-specific manner. In particular, emerging evidence supports that the regulated expression and trafficking of Ca2+-permeable, GluA1-homomeric subtype of AMPARs mediates diverse types of synaptic plasticity, thereby playing critical roles in brain function and dysfunction. In this review, we will discuss the current knowledge of AMPAR subunit-specific trafficking, with a particular emphasis on the involvement of GluA1-homomeric receptor trafficking in synaptic plasticity and brain disorders.
Collapse
Affiliation(s)
- Yuan Ge
- Djavad Mowafaghian Centre for Brain Health and Department of Medicine, University of British Columbia, Vancouver, BC, V6T 2B5, Canada; Djavad Mowafaghian Centre for Brain Health and Department of Psychiatry, University of British Columbia, Vancouver, BC, V6T 2B5, Canada
| | - Yu Tian Wang
- Djavad Mowafaghian Centre for Brain Health and Department of Medicine, University of British Columbia, Vancouver, BC, V6T 2B5, Canada.
| |
Collapse
|
21
|
Matthews PM, Pinggera A, Kampjut D, Greger IH. Biology of AMPA receptor interacting proteins - From biogenesis to synaptic plasticity. Neuropharmacology 2021; 197:108709. [PMID: 34271020 DOI: 10.1016/j.neuropharm.2021.108709] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/19/2021] [Accepted: 07/08/2021] [Indexed: 12/19/2022]
Abstract
AMPA-type glutamate receptors mediate the majority of excitatory synaptic transmission in the central nervous system. Their signaling properties and abundance at synapses are both crucial determinants of synapse efficacy and plasticity, and are therefore under sophisticated control. Unique to this ionotropic glutamate receptor (iGluR) is the abundance of interacting proteins that contribute to its complex regulation. These include transient interactions with the receptor cytoplasmic tail as well as the N-terminal domain locating to the synaptic cleft, both of which are involved in AMPAR trafficking and receptor stabilization at the synapse. Moreover, an array of transmembrane proteins operate as auxiliary subunits that in addition to receptor trafficking and stabilization also substantially impact AMPAR gating and pharmacology. Here, we provide an overview of the catalogue of AMPAR interacting proteins, and how they contribute to the complex biology of this central glutamate receptor.
Collapse
Affiliation(s)
- Peter M Matthews
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Alexandra Pinggera
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Domen Kampjut
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Ingo H Greger
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, UK.
| |
Collapse
|
22
|
Asirvatham AL, Schworer CM, Stahl R, Heitzman D, Carey DJ. Role of A-kinase anchoring proteins in cyclic-AMP-mediated Schwann cell proliferation. Cell Signal 2021; 83:109977. [PMID: 33716104 DOI: 10.1016/j.cellsig.2021.109977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 03/05/2021] [Accepted: 03/08/2021] [Indexed: 10/21/2022]
Abstract
Proliferation of Schwann cells during peripheral nerve development is stimulated by the heregulin/neuregulin family of growth factors expressed by neurons. However, for neonatal rat Schwann cells growing in culture, heregulins produce only a weak mitogenic response. Supplementing heregulin with forskolin, an agent that elevates cyclic AMP levels, produces a dramatic increase in the proliferation of cultured Schwann cells. The mechanisms underlying this synergistic effect required for Schwann cell proliferation in vivo is not well established. Characterizing the A-kinase anchoring proteins (AKAPs) in Schwann cells might help identify substrates tethered to and phosphorylated by the cAMP-dependent protein kinase A (PKA). Using an RII overlay assay that detects AKAPs that are bound to the type II regulatory subunits of PKA, we identified AKAP150 in Schwann cells. Western blot analysis revealed that additional AKAPs, specifically AKAP95, and yotiao were also present. Disruption of PKA/AKAP interaction with Ht-31 peptide resulted in an increase in luciferase-conjugated cyclin D3 promoter activity. Transfection with sequence-specific AKAP siRNAs for AKAP150 and AKAP95 produced a marked reduction in cell proliferation. Immunoblot analysis revealed that knock down of AKAP95 protein caused a significant decrease in expression of the cell cycle regulatory proteins cyclin D2, cyclin D3 and the cell survival signal Akt/Protein Kinase B (Akt/PKB). Morphological characterization of Schwann cell AKAPs indicated the presence of nuclear (AKAP95), cytoplasm-associated (AKAP150) and perinuclear (yotiao) A-kinase anchoring proteins. These results indicate a role for AKAP95 and AKAP150 in the synergistic response of Schwann cells to treatment with heregulin and forskolin.
Collapse
Affiliation(s)
- Angela L Asirvatham
- Department of Biology, Misericordia University, 301 Lake Street Dallas, PA 18612, United States of America.
| | - Charles M Schworer
- Geisinger Medical Center Weis Center for Research, 100 N Academy Avenue, Danville, PA 17822, United States of America
| | - Rick Stahl
- Geisinger Medical Center Weis Center for Research, 100 N Academy Avenue, Danville, PA 17822, United States of America
| | - Deborah Heitzman
- Department of Biology, Bloomsburg University, 400 E. Second Street, Bloomsburg, PA 17815, United States of America
| | - David J Carey
- Geisinger Medical Center Weis Center for Research, 100 N Academy Avenue, Danville, PA 17822, United States of America
| |
Collapse
|
23
|
Di Benedetto G, Iannucci LF, Surdo NC, Zanin S, Conca F, Grisan F, Gerbino A, Lefkimmiatis K. Compartmentalized Signaling in Aging and Neurodegeneration. Cells 2021; 10:464. [PMID: 33671541 PMCID: PMC7926881 DOI: 10.3390/cells10020464] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/16/2021] [Accepted: 02/17/2021] [Indexed: 12/12/2022] Open
Abstract
The cyclic AMP (cAMP) signalling cascade is necessary for cell homeostasis and plays important roles in many processes. This is particularly relevant during ageing and age-related diseases, where drastic changes, generally decreases, in cAMP levels have been associated with the progressive decline in overall cell function and, eventually, the loss of cellular integrity. The functional relevance of reduced cAMP is clearly supported by the finding that increases in cAMP levels can reverse some of the effects of ageing. Nevertheless, despite these observations, the molecular mechanisms underlying the dysregulation of cAMP signalling in ageing are not well understood. Compartmentalization is widely accepted as the modality through which cAMP achieves its functional specificity; therefore, it is important to understand whether and how this mechanism is affected during ageing and to define which is its contribution to this process. Several animal models demonstrate the importance of specific cAMP signalling components in ageing, however, how age-related changes in each of these elements affect the compartmentalization of the cAMP pathway is largely unknown. In this review, we explore the connection of single components of the cAMP signalling cascade to ageing and age-related diseases whilst elaborating the literature in the context of cAMP signalling compartmentalization.
Collapse
Affiliation(s)
- Giulietta Di Benedetto
- Neuroscience Institute, National Research Council of Italy (CNR), 35121 Padova, Italy;
- Veneto Institute of Molecular Medicine, Foundation for Advanced Biomedical Research, 35129 Padova, Italy; (L.F.I.); (S.Z.); (F.C.); (F.G.)
| | - Liliana F. Iannucci
- Veneto Institute of Molecular Medicine, Foundation for Advanced Biomedical Research, 35129 Padova, Italy; (L.F.I.); (S.Z.); (F.C.); (F.G.)
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| | - Nicoletta C. Surdo
- Neuroscience Institute, National Research Council of Italy (CNR), 35121 Padova, Italy;
- Veneto Institute of Molecular Medicine, Foundation for Advanced Biomedical Research, 35129 Padova, Italy; (L.F.I.); (S.Z.); (F.C.); (F.G.)
| | - Sofia Zanin
- Veneto Institute of Molecular Medicine, Foundation for Advanced Biomedical Research, 35129 Padova, Italy; (L.F.I.); (S.Z.); (F.C.); (F.G.)
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| | - Filippo Conca
- Veneto Institute of Molecular Medicine, Foundation for Advanced Biomedical Research, 35129 Padova, Italy; (L.F.I.); (S.Z.); (F.C.); (F.G.)
- Department of Biology, University of Padova, 35122 Padova, Italy
| | - Francesca Grisan
- Veneto Institute of Molecular Medicine, Foundation for Advanced Biomedical Research, 35129 Padova, Italy; (L.F.I.); (S.Z.); (F.C.); (F.G.)
- Department of Biology, University of Padova, 35122 Padova, Italy
| | - Andrea Gerbino
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari, 70121 Bari, Italy;
| | - Konstantinos Lefkimmiatis
- Veneto Institute of Molecular Medicine, Foundation for Advanced Biomedical Research, 35129 Padova, Italy; (L.F.I.); (S.Z.); (F.C.); (F.G.)
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| |
Collapse
|
24
|
AKAP79/150 coordinates leptin-induced PKA signaling to regulate K ATP channel trafficking in pancreatic β-cells. J Biol Chem 2021; 296:100442. [PMID: 33617875 PMCID: PMC8010710 DOI: 10.1016/j.jbc.2021.100442] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 02/11/2021] [Accepted: 02/17/2021] [Indexed: 01/21/2023] Open
Abstract
The adipocyte hormone leptin regulates glucose homeostasis both centrally and peripherally. A key peripheral target is the pancreatic β-cell, which secretes insulin upon glucose stimulation. Leptin is known to suppress glucose-stimulated insulin secretion by promoting trafficking of KATP channels to the β-cell surface, which increases K+ conductance and causes β-cell hyperpolarization. We have previously shown that leptin-induced KATP channel trafficking requires protein kinase A (PKA)-dependent actin remodeling. However, whether PKA is a downstream effector of leptin signaling or PKA plays a permissive role is unknown. Using FRET-based reporters of PKA activity, we show that leptin increases PKA activity at the cell membrane and that this effect is dependent on N-methyl-D-aspartate receptors, CaMKKβ, and AMPK, which are known to be involved in the leptin signaling pathway. Genetic knockdown and rescue experiments reveal that the increased PKA activity upon leptin stimulation requires the membrane-targeted PKA-anchoring protein AKAP79/150, indicating that PKA activated by leptin is anchored to AKAP79/150. Interestingly, disrupting protein phosphatase 2B (PP2B) anchoring to AKAP79/150, known to elevate basal PKA signaling, leads to increased surface KATP channels even in the absence of leptin stimulation. Our findings uncover a novel role of AKAP79/150 in coordinating leptin and PKA signaling to regulate KATP channel trafficking in β-cells, hence insulin secretion. The study further advances our knowledge of the downstream signaling events that may be targeted to restore insulin secretion regulation in β-cells defective in leptin signaling, such as those from obese individuals with type 2 diabetes.
Collapse
|
25
|
Abstract
The field of cAMP signaling is witnessing exciting developments with the recognition that cAMP is compartmentalized and that spatial regulation of cAMP is critical for faithful signal coding. This realization has changed our understanding of cAMP signaling from a model in which cAMP connects a receptor at the plasma membrane to an intracellular effector in a linear pathway to a model in which cAMP signals propagate within a complex network of alternative branches and the specific functional outcome strictly depends on local regulation of cAMP levels and on selective activation of a limited number of branches within the network. In this review, we cover some of the early studies and summarize more recent evidence supporting the model of compartmentalized cAMP signaling, and we discuss how this knowledge is starting to provide original mechanistic insight into cell physiology and a novel framework for the identification of disease mechanisms that potentially opens new avenues for therapeutic interventions. SIGNIFICANCE STATEMENT: cAMP mediates the intracellular response to multiple hormones and neurotransmitters. Signal fidelity and accurate coordination of a plethora of different cellular functions is achieved via organization of multiprotein signalosomes and cAMP compartmentalization in subcellular nanodomains. Defining the organization and regulation of subcellular cAMP nanocompartments is necessary if we want to understand the complex functional ramifications of pharmacological treatments that target G protein-coupled receptors and for generating a blueprint that can be used to develop precision medicine interventions.
Collapse
Affiliation(s)
- Manuela Zaccolo
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Anna Zerio
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Miguel J Lobo
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
26
|
Raghuram V, Salhadar K, Limbutara K, Park E, Yang CR, Knepper MA. Protein kinase A catalytic-α and catalytic-β proteins have nonredundant regulatory functions. Am J Physiol Renal Physiol 2020; 319:F848-F862. [PMID: 33017189 PMCID: PMC7789987 DOI: 10.1152/ajprenal.00383.2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/28/2020] [Accepted: 09/25/2020] [Indexed: 01/08/2023] Open
Abstract
Vasopressin regulates osmotic water transport in the renal collecting duct by protein kinase A (PKA)-mediated control of the water channel aquaporin-2 (AQP2). Collecting duct principal cells express two seemingly redundant PKA catalytic subunits, PKA catalytic α (PKA-Cα) and PKA catalytic β (PKA-Cβ). To identify the roles of these two protein kinases, we carried out deep phosphoproteomic analysis in cultured mpkCCD cells in which either PKA-Cα or PKA-Cβ was deleted using CRISPR-Cas9-based genome editing. Controls were cells carried through the genome editing procedure but without deletion of PKA. TMT mass tagging was used for protein mass spectrometric quantification. Of the 4,635 phosphopeptides that were quantified, 67 phosphopeptides were significantly altered in abundance with PKA-Cα deletion, whereas 21 phosphopeptides were significantly altered in abundance with PKA-Cβ deletion. However, only four sites were changed in both. The target proteins identified in PKA-Cα-null cells were largely associated with cell membranes and membrane vesicles, whereas target proteins in PKA-Cβ-null cells were largely associated with the actin cytoskeleton and cell junctions. In contrast, in vitro incubation of mpkCCD proteins with recombinant PKA-Cα and PKA-Cβ resulted in virtually identical phosphorylation changes. In addition, analysis of total protein abundances in in vivo samples showed that PKA-Cα deletion resulted in a near disappearance of AQP2 protein, whereas PKA-Cβ deletion did not decrease AQP2 abundance. We conclude that PKA-Cα and PKA-Cβ serve substantially different regulatory functions in renal collecting duct cells and that differences in phosphorylation targets may be due to differences in protein interactions, e.g., mediated by A-kinase anchor proteins, C-kinase anchoring proteins, or PDZ binding.
Collapse
Affiliation(s)
- Viswanathan Raghuram
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Karim Salhadar
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Kavee Limbutara
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Euijung Park
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Chin-Rang Yang
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Mark A Knepper
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
27
|
Omar MH, Scott JD. AKAP Signaling Islands: Venues for Precision Pharmacology. Trends Pharmacol Sci 2020; 41:933-946. [PMID: 33082006 DOI: 10.1016/j.tips.2020.09.007] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 09/21/2020] [Accepted: 09/24/2020] [Indexed: 12/19/2022]
Abstract
Regulatory enzymes often have different roles in distinct subcellular compartments. Yet, most drugs indiscriminately saturate the cell. Thus, subcellular drug-delivery holds promise as a means to reduce off-target pharmacological effects. A-kinase anchoring proteins (AKAPs) sequester combinations of signaling enzymes within subcellular microdomains. Targeting drugs to these 'signaling islands' offers an opportunity for more precise delivery of therapeutics. Here, we review mechanisms that bestow protein kinase A (PKA) versatility inside the cell, appraise recent advances in exploiting AKAPs as platforms for precision pharmacology, and explore the impact of methodological innovations on AKAP research.
Collapse
Affiliation(s)
- Mitchell H Omar
- Department of Pharmacology, University of Washington, Seattle, WA, 98195, USA
| | - John D Scott
- Department of Pharmacology, University of Washington, Seattle, WA, 98195, USA.
| |
Collapse
|
28
|
Sumi T, Harada K. Mechanism underlying hippocampal long-term potentiation and depression based on competition between endocytosis and exocytosis of AMPA receptors. Sci Rep 2020; 10:14711. [PMID: 32895399 PMCID: PMC7477194 DOI: 10.1038/s41598-020-71528-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 08/17/2020] [Indexed: 02/08/2023] Open
Abstract
N-methyl-D-aspartate (NMDA) receptor-dependent long-term potentiation (LTP) and long-term depression (LTD) of signal transmission form neural circuits and thus are thought to underlie learning and memory. These mechanisms are mediated by AMPA receptor (AMPAR) trafficking in postsynaptic neurons. However, the regulatory mechanism of bidirectional plasticity at excitatory synapses remains unclear. We present a network model of AMPAR trafficking for adult hippocampal pyramidal neurons, which reproduces both LTP and LTD. We show that the induction of both LTP and LTD is regulated by the competition between exocytosis and endocytosis of AMPARs, which are mediated by the calcium-sensors synaptotagmin 1/7 (Syt1/7) and protein interacting with C-kinase 1 (PICK1), respectively. Our result indicates that recycling endosomes containing AMPAR are always ready for Syt1/7-dependent exocytosis of AMPAR at peri-synaptic/synaptic membranes. This is because molecular motor myosin Vb constitutively transports the recycling endosome toward the membrane in a Ca2+-independent manner.
Collapse
Affiliation(s)
- Tomonari Sumi
- Research Institute for Interdisciplinary Science, Okayama University, 3-1-1 Tsushima-Naka, Kita-ku, Okayama, 700-8530, Japan. .,Department of Chemistry, Faculty of Science, Okayama University, 3-1-1 Tsushima-Naka, Kita-ku, Okayama, 700-8530, Japan.
| | - Kouji Harada
- Department of Computer Science and Engineering, Toyohashi University of Technology, 1-1 Hibarigaoka, Tempaku-cho, Toyohashi, Aichi, 441-8580, Japan
| |
Collapse
|
29
|
Bucko PJ, Scott JD. Drugs That Regulate Local Cell Signaling: AKAP Targeting as a Therapeutic Option. Annu Rev Pharmacol Toxicol 2020; 61:361-379. [PMID: 32628872 DOI: 10.1146/annurev-pharmtox-022420-112134] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Cells respond to environmental cues by mobilizing signal transduction cascades that engage protein kinases and phosphoprotein phosphatases. Correct organization of these enzymes in space and time enables the efficient and precise transmission of chemical signals. The cyclic AMP-dependent protein kinase A is compartmentalized through its association with A-kinase anchoring proteins (AKAPs). AKAPs are a family of multivalent scaffolds that constrain signaling enzymes and effectors at subcellular locations to drive essential physiological events. More recently, it has been recognized that defective signaling in certain endocrine disorders and cancers proceeds through pathological AKAP complexes. Consequently, pharmacologically targeting these macromolecular complexes unlocks new therapeutic opportunities for a growing number of clinical indications. This review highlights recent findings on AKAP signaling in disease, particularly in certain cancers, and offers an overview of peptides and small molecules that locally regulate AKAP-binding partners.
Collapse
Affiliation(s)
- Paula J Bucko
- Department of Pharmacology, University of Washington, Seattle, Washington 98195, USA; ,
| | - John D Scott
- Department of Pharmacology, University of Washington, Seattle, Washington 98195, USA; ,
| |
Collapse
|
30
|
Warnet XL, Bakke Krog H, Sevillano-Quispe OG, Poulsen H, Kjaergaard M. The C-terminal domains of the NMDA receptor: How intrinsically disordered tails affect signalling, plasticity and disease. Eur J Neurosci 2020; 54:6713-6739. [PMID: 32464691 DOI: 10.1111/ejn.14842] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 04/16/2020] [Accepted: 05/18/2020] [Indexed: 01/14/2023]
Abstract
NMDA receptors are part of the ionotropic glutamate receptor family, and are crucial for neurotransmission and memory. At the cellular level, the effects of activating these receptors include long-term potentiation (LTP) or depression (LTD). The NMDA receptor is a stringently gated cation channel permeable to Ca2+ , and it shares the molecular architecture of a tetrameric ligand-gated ion channel with the other family members. Its subunits, however, have uniquely long cytoplasmic C-terminal domains (CTDs). While the molecular gymnastics of the extracellular domains have been described in exquisite detail, much less is known about the structure and function of these CTDs. The CTDs vary dramatically in length and sequence between receptor subunits, but they all have a composition characteristic of intrinsically disordered proteins. The CTDs affect channel properties, trafficking and downstream signalling output from the receptor, and these functions are regulated by alternative splicing, protein-protein interactions, and post-translational modifications such as phosphorylation and palmitoylation. Here, we review the roles of the CTDs in synaptic plasticity with a focus on biochemical mechanisms. In total, the CTDs play a multifaceted role as a modifier of channel function, a regulator of cellular location and abundance, and signalling scaffold control the downstream signalling output.
Collapse
Affiliation(s)
- Xavier L Warnet
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark.,The Danish Research Institute for Translational Neuroscience (DANDRITE), Aarhus University, Aarhus, Denmark.,The Center for Proteins in Memory (PROMEMO), Aarhus University, Aarhus, Denmark
| | - Helle Bakke Krog
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark.,The Danish Research Institute for Translational Neuroscience (DANDRITE), Aarhus University, Aarhus, Denmark.,The Center for Proteins in Memory (PROMEMO), Aarhus University, Aarhus, Denmark
| | - Oscar G Sevillano-Quispe
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark.,The Danish Research Institute for Translational Neuroscience (DANDRITE), Aarhus University, Aarhus, Denmark.,The Center for Proteins in Memory (PROMEMO), Aarhus University, Aarhus, Denmark
| | - Hanne Poulsen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark.,The Danish Research Institute for Translational Neuroscience (DANDRITE), Aarhus University, Aarhus, Denmark.,The Center for Proteins in Memory (PROMEMO), Aarhus University, Aarhus, Denmark
| | - Magnus Kjaergaard
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark.,The Danish Research Institute for Translational Neuroscience (DANDRITE), Aarhus University, Aarhus, Denmark.,The Center for Proteins in Memory (PROMEMO), Aarhus University, Aarhus, Denmark
| |
Collapse
|
31
|
Proteasomal-Mediated Degradation of AKAP150 Accompanies AMPAR Endocytosis during cLTD. eNeuro 2020; 7:ENEURO.0218-19.2020. [PMID: 32205379 PMCID: PMC7163082 DOI: 10.1523/eneuro.0218-19.2020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 02/17/2020] [Accepted: 03/02/2020] [Indexed: 12/26/2022] Open
Abstract
The number and function of synaptic AMPA receptors (AMPARs) tightly regulates excitatory synaptic transmission. Current evidence suggests that AMPARs are inserted into the postsynaptic membrane during long-term potentiation (LTP) and are removed from the membrane during long-term depression (LTD). Dephosphorylation of GluA1 at Ser-845 and enhanced endocytosis are critical events in the modulation of LTD. Moreover, changes in scaffold proteins from the postsynaptic density (PSD) could be also related to AMPAR regulation in LTD. In the present study we analyzed the effect of chemical LTD (cLTD) on A-kinase anchoring protein (AKAP)150 and AMPARs levels in mouse-cultured neurons. We show that cLTD induces AKAP150 protein degradation via proteasome, coinciding with GluA1 dephosphorylation at Ser-845 and endocytosis of GluA1-containing AMPARs. Pharmacological inhibition of proteasome activity, but not phosphatase calcineurin (CaN), reverted cLTD-induced AKAP150 protein degradation. Importantly, AKAP150 silencing induced dephosphorylation of GluA1 Ser-845 and GluA1-AMPARs endocytosis while AKAP150 overexpression blocked cLTD-mediated GluA1-AMPARs endocytosis. Our results provide direct evidence that cLTD-induced AKAP150 degradation by the proteasome contributes to synaptic AMPARs endocytosis.
Collapse
|
32
|
Purkey AM, Dell’Acqua ML. Phosphorylation-Dependent Regulation of Ca 2+-Permeable AMPA Receptors During Hippocampal Synaptic Plasticity. Front Synaptic Neurosci 2020; 12:8. [PMID: 32292336 PMCID: PMC7119613 DOI: 10.3389/fnsyn.2020.00008] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 02/18/2020] [Indexed: 01/28/2023] Open
Abstract
Experience-dependent learning and memory require multiple forms of plasticity at hippocampal and cortical synapses that are regulated by N-methyl-D-aspartate receptors (NMDA) and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)-type ionotropic glutamate receptors (NMDAR, AMPAR). These plasticity mechanisms include long-term potentiation (LTP) and depression (LTD), which are Hebbian input-specific mechanisms that rapidly increase or decrease AMPAR synaptic strength at specific inputs, and homeostatic plasticity that globally scales-up or -down AMPAR synaptic strength across many or even all inputs. Frequently, these changes in synaptic strength are also accompanied by a change in the subunit composition of AMPARs at the synapse due to the trafficking to and from the synapse of receptors lacking GluA2 subunits. These GluA2-lacking receptors are most often GluA1 homomeric receptors that exhibit higher single-channel conductance and are Ca2+-permeable (CP-AMPAR). This review article will focus on the role of protein phosphorylation in regulation of GluA1 CP-AMPAR recruitment and removal from hippocampal synapses during synaptic plasticity with an emphasis on the crucial role of local signaling by the cAMP-dependent protein kinase (PKA) and the Ca2+calmodulin-dependent protein phosphatase 2B/calcineurin (CaN) that is coordinated by the postsynaptic scaffold protein A-kinase anchoring protein 79/150 (AKAP79/150).
Collapse
Affiliation(s)
| | - Mark L. Dell’Acqua
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
33
|
Guerra DD, Bok R, Lorca RA, Hurt KJ. Protein kinase A facilitates relaxation of mouse ileum via phosphorylation of neuronal nitric oxide synthase. Br J Pharmacol 2020; 177:2765-2778. [PMID: 31975425 DOI: 10.1111/bph.15001] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 01/03/2020] [Accepted: 01/18/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND AND PURPOSE The enteric neurotransmitter nitric oxide (NO) regulates gastrointestinal motility by relaxing smooth muscle. Pharmacological cAMP induction also relaxes gastrointestinal smooth muscle, but it is uncertain whether cAMP augments or suppresses enteric NO signalling. In other organ systems, cAMP can increase neuronal NO production by stimulating protein kinase A (PKA) to phosphorylate neuronal NOS (nNOS) Serine-1412 (S1412). We hypothesized that cAMP also increases nNOS S1412 phosphorylation by PKA in enteric neurons to augment nitrergic relaxation of mouse ileum. EXPERIMENTAL APPROACH We measured contractile force and nNOS S1412 phosphorylation in ileal rings suspended in an organ bath. We used forskolin to induce cAMP-dependent relaxation of wild type, nNOSS1412A knock-in and nNOSα-null ileal rings in the presence or absence of PKA, protein kinase B (Akt) and NOS inhibitors. KEY RESULTS Forskolin stimulated phosphorylation of nNOS S1412 in mouse ileum. Forskolin relaxed nNOSα-null and nNOSS1412A ileal rings less than wild-type ileal rings. PKA inhibition blocked forskolin-induced nNOS phosphorylation and attenuated relaxation of wild type but not nNOSS1412A ileum. Akt inhibition did not alter nNOS phosphorylation with forskolin but did attenuate relaxation of wild type and nNOSS1412A . NOS inhibition with L-NAME eliminated the effects of PKA and Akt inhibitors on relaxation. CONCLUSION AND IMPLICATIONS PKA phosphorylation of nNOS S1412 augments forskolin-induced nitrergic ileal relaxation. The relationship between cAMP/PKA and NO is therefore synergistic in enteric nitrergic neurons. Because NO regulates gut motility, selective modulation of enteric neuronal cAMP synthesis may be useful for the treatment of gastrointestinal motility disorders.
Collapse
Affiliation(s)
- Damian D Guerra
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Rachael Bok
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Ramón A Lorca
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - K Joseph Hurt
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado.,Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| |
Collapse
|
34
|
Argyrousi EK, Heckman PRA, Prickaerts J. Role of cyclic nucleotides and their downstream signaling cascades in memory function: Being at the right time at the right spot. Neurosci Biobehav Rev 2020; 113:12-38. [PMID: 32044374 DOI: 10.1016/j.neubiorev.2020.02.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 01/23/2020] [Accepted: 02/03/2020] [Indexed: 01/23/2023]
Abstract
A plethora of studies indicate the important role of cAMP and cGMP cascades in neuronal plasticity and memory function. As a result, altered cyclic nucleotide signaling has been implicated in the pathophysiology of mnemonic dysfunction encountered in several diseases. In the present review we provide a wide overview of studies regarding the involvement of cyclic nucleotides, as well as their upstream and downstream molecules, in physiological and pathological mnemonic processes. Next, we discuss the regulation of the intracellular concentration of cyclic nucleotides via phosphodiesterases, the enzymes that degrade cAMP and/or cGMP, and via A-kinase-anchoring proteins that refine signal compartmentalization of cAMP signaling. We also provide an overview of the available data pointing to the existence of specific time windows in cyclic nucleotide signaling during neuroplasticity and memory formation and the significance to target these specific time phases for improving memory formation. Finally, we highlight the importance of emerging imaging tools like Förster resonance energy transfer imaging and optogenetics in detecting, measuring and manipulating the action of cyclic nucleotide signaling cascades.
Collapse
Affiliation(s)
- Elentina K Argyrousi
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, 6200 MD, the Netherlands
| | - Pim R A Heckman
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, 6200 MD, the Netherlands
| | - Jos Prickaerts
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, 6200 MD, the Netherlands.
| |
Collapse
|
35
|
Translating preclinical findings in clinically relevant new antipsychotic targets: focus on the glutamatergic postsynaptic density. Implications for treatment resistant schizophrenia. Neurosci Biobehav Rev 2019; 107:795-827. [DOI: 10.1016/j.neubiorev.2019.08.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Revised: 07/20/2019] [Accepted: 08/22/2019] [Indexed: 02/07/2023]
|
36
|
Man KNM, Navedo MF, Horne MC, Hell JW. β 2 Adrenergic Receptor Complexes with the L-Type Ca 2+ Channel Ca V1.2 and AMPA-Type Glutamate Receptors: Paradigms for Pharmacological Targeting of Protein Interactions. Annu Rev Pharmacol Toxicol 2019; 60:155-174. [PMID: 31561738 DOI: 10.1146/annurev-pharmtox-010919-023404] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Formation of signaling complexes is crucial for the orchestration of fast, efficient, and specific signal transduction. Pharmacological disruption of defined signaling complexes has the potential for specific intervention in selected regulatory pathways without affecting organism-wide disruption of parallel pathways. Signaling by epinephrine and norepinephrine through α and β adrenergic receptors acts on many signaling pathways in many cell types. Here, we initially provide an overview of the signaling complexes formed between the paradigmatic β2 adrenergic receptor and two of its most important targets, the L-type Ca2+ channel CaV1.2 and the AMPA-type glutamate receptor. Importantly, both complexes contain the trimeric Gs protein, adenylyl cyclase, and the cAMP-dependent protein kinase, PKA. We then discuss the functional implications of the formation of these complexes, how those complexes can be specifically disrupted, and how such disruption could be utilized in the pharmacological treatment of disease.
Collapse
Affiliation(s)
- Kwun Nok Mimi Man
- Department of Pharmacology, University of California, Davis, California 95616, USA;
| | - Manuel F Navedo
- Department of Pharmacology, University of California, Davis, California 95616, USA;
| | - Mary C Horne
- Department of Pharmacology, University of California, Davis, California 95616, USA;
| | - Johannes W Hell
- Department of Pharmacology, University of California, Davis, California 95616, USA;
| |
Collapse
|
37
|
Zhou HY, He JG, Hu ZL, Xue SG, Xu JF, Cui QQ, Gao SQ, Zhou B, Wu PF, Long LH, Wang F, Chen JG. A-Kinase Anchoring Protein 150 and Protein Kinase A Complex in the Basolateral Amygdala Contributes to Depressive-like Behaviors Induced by Chronic Restraint Stress. Biol Psychiatry 2019; 86:131-142. [PMID: 31076080 DOI: 10.1016/j.biopsych.2019.03.967] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 02/11/2019] [Accepted: 03/05/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND The basolateral amygdala (BLA) has been widely implicated in the pathophysiology of major depressive disorder. A-kinase anchoring protein 150 (AKAP150) directs kinases and phosphatases to synaptic glutamate receptors, controlling synaptic transmission and plasticity. However, the role of the AKAP150 in the BLA in major depressive disorder remains poorly understood. METHODS Depressive-like behaviors in C57BL/6J mice were developed by chronic restraint stress (CRS). Mice received either intra-BLA injection of lentivirus-expressing Akap5 short hairpin RNA or Ht-31, a peptide to disrupt the interaction of AKAP150 and protein kinase A (PKA), followed by depressive-like behavioral tests. Alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid glutamate receptor (AMPAR)-mediated miniature excitatory postsynaptic currents were recorded by whole-cell patch-clamp techniques. RESULTS Chronic stress exposure induced depressive-like behaviors, which were accompanied by an increase in total and synaptic AKAP150 expression in the BLA. Accordingly, CRS facilitated the association of AKAP150 with PKA, but not of calcineurin in the BLA. Intra-BLA infusion of lentivirus-expressing Akap5 short hairpin RNA or Ht-31 prevented depressive-like behaviors and normalized phosphorylation of serine 845 and surface expression of AMPAR subunit 1 (GluA1) in the BLA of CRS mice. Finally, blockage of AKAP150-PKA complex signaling rescued the changes in AMPAR-mediated miniature excitatory postsynaptic currents in depressive-like mice. CONCLUSIONS These results suggest that AKAP150-PKA directly modulates BLA neuronal synaptic strength, and that AKAP150-PKA-GluA1 streamline signaling complex is responsible for CRS-induced disruption of synaptic AMPAR-mediated transmission and depressive-like behaviors in mice.
Collapse
Affiliation(s)
- Hai-Yun Zhou
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jin-Gang He
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Research Center for Depression, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Laboratory of Neuropsychiatric Diseases, Institute of Brain Research, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory of Neurological Diseases, Huazhong University of Science and Technology, Ministry of Education of China, Wuhan, China
| | - Zhuang-Li Hu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Laboratory of Neuropsychiatric Diseases, Institute of Brain Research, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory of Neurological Diseases, Huazhong University of Science and Technology, Ministry of Education of China, Wuhan, China
| | - Shi-Ge Xue
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jun-Feng Xu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qian-Qian Cui
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuang-Qi Gao
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bin Zhou
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Peng-Fei Wu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Laboratory of Neuropsychiatric Diseases, Institute of Brain Research, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory of Neurological Diseases, Huazhong University of Science and Technology, Ministry of Education of China, Wuhan, China
| | - Li-Hong Long
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Laboratory of Neuropsychiatric Diseases, Institute of Brain Research, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory of Neurological Diseases, Huazhong University of Science and Technology, Ministry of Education of China, Wuhan, China
| | - Fang Wang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Research Center for Depression, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Laboratory of Neuropsychiatric Diseases, Institute of Brain Research, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory of Neurological Diseases, Huazhong University of Science and Technology, Ministry of Education of China, Wuhan, China.
| | - Jian-Guo Chen
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Research Center for Depression, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Laboratory of Neuropsychiatric Diseases, Institute of Brain Research, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory of Neurological Diseases, Huazhong University of Science and Technology, Ministry of Education of China, Wuhan, China.
| |
Collapse
|
38
|
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: 63] [Impact Index Per Article: 12.6] [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
|
39
|
Abstract
PSD-95 is a scaffolding protein that regulates the synaptic localization of many receptors, channels, and signaling proteins. The NLGN gene family encodes single-pass transmembrane postsynaptic cell adhesion molecules that are important for synapse assembly and function. At excitatory synapses, NLGN1 mediates transsynaptic binding with neurexin, a presynaptic cell adhesion molecule, and also binds to PSD-95, although the relevance of the PSD-95 interaction is not clear. We now show that disruption of the NLGN1 and PSD-95 interaction decreases surface expression of NLGN1 in cultured neurons. Furthermore, PKA phosphorylates NLGN1 on S839, near the PDZ ligand, and dynamically regulates PSD-95 binding. A phosphomimetic mutation of NLGN1 S839 significantly reduced PSD-95 binding. Impaired NLGN1/PSD-95 binding diminished synaptic NLGN1 expression and NLGN1-mediated synaptic enhancement. Our results establish a phosphorylation-dependent molecular mechanism that regulates NLGN1 and PSD-95 binding and provides insights into excitatory synaptic development and function.
Collapse
|
40
|
Hoffman JR, Brandwein NJ, Nguyen PV. Induction of β-adrenergic metaplasticity of LTP requires intact anchoring of PKA. ACTA ACUST UNITED AC 2019; 26:187-190. [PMID: 31109969 PMCID: PMC6529881 DOI: 10.1101/lm.049635.119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Accepted: 04/23/2019] [Indexed: 11/24/2022]
Abstract
Beta-adrenergic receptors (β-ARs) prime hippocampal synapses to stabilize long-term potentiation (LTP). This “metaplasticity” can persist for 1–2 h after pharmacologic activation of β-ARs. It requires activation of PKA (cAMP-dependent protein kinase) during β-AR priming. A-kinase anchoring proteins (AKAPs) tether PKA to downstream signaling proteins. We hypothesized that induction of this metaplasticity requires intact functioning of AKAPs. Acute application of stearated ht31, a membrane-permeant inhibitor of AKAPs, either during β-AR activation 30 min prior to LTP induction or during LTP induction, attenuated the persistence of LTP. A control, inactive ht31 peptide did not affect β-AR-mediated metaplasticity. These findings implicate PKA anchoring in the induction of β-adrenergic metaplasticity of LTP.
Collapse
Affiliation(s)
- Janlyn R Hoffman
- Department of Physiology, University of Alberta School of Medicine, Edmonton, Alberta T6G 2H7, Canada
| | - Nathan J Brandwein
- Department of Physiology, University of Alberta School of Medicine, Edmonton, Alberta T6G 2H7, Canada
| | - Peter V Nguyen
- Department of Physiology, University of Alberta School of Medicine, Edmonton, Alberta T6G 2H7, Canada
| |
Collapse
|
41
|
NPY 2 Receptors Reduce Tonic Action Potential-Independent GABA B Currents in the Basolateral Amygdala. J Neurosci 2019; 39:4909-4930. [PMID: 30971438 DOI: 10.1523/jneurosci.2226-18.2019] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 03/29/2019] [Accepted: 04/01/2019] [Indexed: 01/17/2023] Open
Abstract
Although NPY has potent anxiolytic actions within the BLA, selective activation of BLA NPY Y2 receptors (Y2Rs) acutely increases anxiety by an unknown mechanism. Using ex vivo male rat brain slice electrophysiology, we show that the selective Y2R agonist, [ahx5-24]NPY, reduced the frequency of GABAA-mediated mIPSCs in BLA principal neurons (PNs). [ahx5-24]NPY also reduced tonic activation of GABAB receptors (GABABR), which increased PN excitability through inhibition of a tonic, inwardly rectifying potassium current (KIR ). Surprisingly, Y2R-sensitive GABABR currents were action potential-independent, persisting after treatment with TTX. Additionally, the Ca2+-dependent, slow afterhyperpolarizing K+ current (IsAHP ) was enhanced in approximately half of the Y2R-sensitive PNs, possibly from enhanced Ca2+ influx, permitted by reduced GABABR tone. In male and female mice expressing tdTomato in Y2R-mRNA cells (tdT-Y2R mice), immunohistochemistry revealed that BLA somatostatin interneurons express Y2Rs, as do a significant subset of BLA PNs. In tdT-Y2R mice, [ahx5-24]NPY increased excitability and suppressed the KIR in nearly all BLA PNs independent of tdT-Y2R fluorescence, consistent with presynaptic Y2Rs on somatostatin interneurons mediating the above effects. However, only tdT-Y2R-expressing PNs responded to [ahx5-24]NPY with an enhancement of the IsAHP Ultimately, increased PN excitability via acute Y2R activation likely correlates with enhanced BLA output, consistent with reported Y2R-mediated anxiogenesis. Furthermore, we demonstrate the following: (1) a novel mechanism whereby activity-independent GABA release can powerfully dampen BLA neuronal excitability via postsynaptic GABABRs; and (2) that this tonic inhibition can be interrupted by neuromodulation, here by NPY via Y2Rs.SIGNIFICANCE STATEMENT Within the BLA, NPY is potently anxiolytic. However, selective activation of NPY2 receptors (Y2Rs) increases anxiety by an unknown mechanism. We show that activation of BLA Y2Rs decreases tonic GABA release onto BLA principal neurons, probably from Y2R-expressing somatostatin interneurons, some of which coexpress NPY. This increases principal neuron excitability by reducing GABAB receptor (GABABR)-mediated activation of G-protein-coupled, inwardly rectifying K+ currents. Tonic, Y2R-sensitive GABABR currents unexpectedly persisted in the absence of action potential firing, revealing, to our knowledge, the first report of substantial, activity-independent GABABR activation. Ultimately, we provide a plausible explanation for Y2R-mediated anxiogenesis in vivo and describe a novel and modulatable means of damping neuronal excitability.
Collapse
|
42
|
Summers KC, Bogard AS, Tavalin SJ. Preferential generation of Ca 2+-permeable AMPA receptors by AKAP79-anchored protein kinase C proceeds via GluA1 subunit phosphorylation at Ser-831. J Biol Chem 2019; 294:5521-5535. [PMID: 30737285 DOI: 10.1074/jbc.ra118.004340] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 02/06/2019] [Indexed: 01/01/2023] Open
Abstract
AMPA-type glutamate receptors (AMPARs) mediate fast excitatory neurotransmission in the mammalian central nervous system. Preferential AMPAR subunit assembly favors heteromeric GluA1/GluA2 complexes. The presence of the GluA2 subunit generates Ca2+-impermeable (CI) AMPARs that have linear current-voltage (I-V) relationships. However, diverse forms of synaptic plasticity and pathophysiological conditions are associated with shifts from CI to inwardly rectifying, GluA2-lacking, Ca2+-permeable (CP) AMPARs on time scales ranging from minutes to days. These shifts have been linked to GluA1 phosphorylation at Ser-845, a protein kinase A (PKA)-targeted site within its intracellular C-terminal tail, often in conjunction with protein kinase A anchoring protein 79 (AKAP79; AKAP150 in rodents), which targets PKA to GluA1. However, AKAP79 may impact GluA1 phosphorylation at other sites by interacting with other signaling enzymes. Here, we evaluated the ability of AKAP79, its signaling components, and GluA1 phosphorylation sites to induce CP-AMPARs under conditions in which CI-AMPARs normally predominate. We found that GluA1 phosphorylation at Ser-831 is sufficient for the appearance of CP-AMPARs and that AKAP79-anchored protein kinase C (PKC) primarily drives the appearance of these receptors via this site. In contrast, other AKAP79-signaling components and C-terminal tail GluA1 phosphorylation sites exhibited a permissive role, limiting the extent to which AKAP79 promotes CP-AMPARs. This may reflect the need for these sites to undergo active phosphorylation/dephosphorylation cycles that control their residency within distinct subcellular compartments. These findings suggest that AKAP79, by orchestrating phosphorylation, represents a key to a GluA1 phosphorylation passcode, which allows the GluA1 subunit to escape GluA2 dominance and promote the appearance of CP-AMPARs.
Collapse
Affiliation(s)
- Kyle C Summers
- From the Department of Pharmacology, University of Tennessee Health Science Center, Memphis, Tennessee 38103
| | - Amy S Bogard
- From the Department of Pharmacology, University of Tennessee Health Science Center, Memphis, Tennessee 38103
| | - Steven J Tavalin
- From the Department of Pharmacology, University of Tennessee Health Science Center, Memphis, Tennessee 38103
| |
Collapse
|
43
|
Cannizzaro C, Talani G, Brancato A, Mulas G, Spiga S, De Luca MA, Sanna A, Marino RAM, Biggio G, Sanna E, Diana M. Dopamine Restores Limbic Memory Loss, Dendritic Spine Structure, and NMDAR-Dependent LTD in the Nucleus Accumbens of Alcohol-Withdrawn Rats. J Neurosci 2019; 39:929-943. [PMID: 30446531 PMCID: PMC6382989 DOI: 10.1523/jneurosci.1377-18.2018] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 09/28/2018] [Accepted: 10/15/2018] [Indexed: 02/06/2023] Open
Abstract
Alcohol abuse leads to aberrant forms of emotionally salient memory, i.e., limbic memory, that promote escalated alcohol consumption and relapse. Accordingly, activity-dependent structural abnormalities are likely to contribute to synaptic dysfunctions that occur from suddenly ceasing chronic alcohol consumption. Here we show that alcohol-dependent male rats fail to perform an emotional-learning task during abstinence but recover their functioning by l-3,4-dihydroxyphenylalanin (l-DOPA) administration during early withdrawal. l-DOPA also reverses the selective loss of dendritic "long thin" spines observed in medium spiny neurons of the nucleus accumbens (NAc) shell of alcohol-dependent rats during abstinence, as well as the reduction in tyrosine hydroxylase immunostaining and postsynaptic density-95-positive elements. Patch-clamp experiments in NAc slices reveal that both in vivo systemic l-DOPA administration and in vitro exposure to dopamine can restore the loss of long-term depression (LTD) formation, counteract the reduction in NMDAR-mediated synaptic currents and rectify the altered NMDAR/AMPAR ratio observed in alcohol-withdrawn rats. Further, in vivo microdialysis experiments show that blunted dopaminergic signaling is revived after l-DOPA treatment during early withdrawal. These results suggest a key role of an efficient dopamine signaling for maintaining, and restore, neural trophism, NMDA-dependent LTD, and ultimately optimal learning.SIGNIFICANCE STATEMENT Blunted dopamine signaling and altered glutamate connectivity in the nucleus accumbens represent the neuroanatomical basis for the impairment in aversive limbic memory observed during withdrawal in alcohol dependence. Supplying l-DOPA during withdrawal re-establishes synaptic morphology and functional neuroadaptations, suggesting a complete recovery of nucleus accumbens glutamatergic synaptic plasticity when dopamine is revived. Importantly, restoring dopamine transmission allows those synapses to encode emotionally relevant information and rescue flexibility in the neuronal circuits that process limbic memory formation. Under these conditions, drugs capable of selectively boosting the dopaminergic function during the "fluid" and still responsive state of the early withdrawn maladaptive synapses may help in the treatment of alcohol addiction.
Collapse
Affiliation(s)
- Carla Cannizzaro
- Laboratory of Neuropsychopharmacology, Department ProSaMI G. D'Alessandro, University of Palermo, Via del Vespro 129 90127, Palermo, Italy
| | - Giuseppe Talani
- Institute of Neuroscience, National Research Council, 09042 Monserrato, Cagliari, Italy
| | - Anna Brancato
- Laboratory of Neuropsychopharmacology, Department ProSaMI G. D'Alessandro, University of Palermo, Via del Vespro 129 90127, Palermo, Italy
| | - Giovanna Mulas
- Department of Life and Environmental Sciences, Section of Neuroscience and Anthropology, University of Cagliari, 09042 Monserrato, Cagliari, Italy
| | - Saturnino Spiga
- Department of Life and Environmental Sciences, Section of Neuroscience and Anthropology, University of Cagliari, 09042 Monserrato, Cagliari, Italy
| | - Maria Antonietta De Luca
- Department of Biomedical Sciences, Section of Neuropsychopharmacology, University of Cagliari, Cittadella Universitaria-S.P., 09042 Monserrato, Cagliari, Italy
| | - Angela Sanna
- Department of Medical Science and Public Health, University of Cagliari, Cittadella Universitaria-S.P., 09042 Monserrato, Cagliari, Italy
| | - Rosa Anna Maria Marino
- Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland 21224, and
| | - Giovanni Biggio
- Institute of Neuroscience, National Research Council, 09042 Monserrato, Cagliari, Italy
- Department of Life and Environmental Sciences, Section of Neuroscience and Anthropology, University of Cagliari, 09042 Monserrato, Cagliari, Italy
| | - Enrico Sanna
- Institute of Neuroscience, National Research Council, 09042 Monserrato, Cagliari, Italy
- Department of Life and Environmental Sciences, Section of Neuroscience and Anthropology, University of Cagliari, 09042 Monserrato, Cagliari, Italy
| | - Marco Diana
- University of Sassari, G.Minardi Laboratory of Cognitive Neuroscience, Department of Chemistry and Pharmacy Via Muroni, 23 07100 Sassari, Italy
| |
Collapse
|
44
|
Rhee SW, Rusch NJ. Molecular determinants of beta-adrenergic signaling to voltage-gated K + channels in the cerebral circulation. Microcirculation 2018; 25. [PMID: 29072364 DOI: 10.1111/micc.12425] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 10/19/2017] [Indexed: 12/14/2022]
Abstract
Voltage-gated K+ (Kv ) channels are major determinants of membrane potential in vascular smooth muscle cells (VSMCs) and regulate the diameter of small cerebral arteries and arterioles. However, the intracellular structures that govern the expression and function of vascular Kv channels are poorly understood. Scaffolding proteins including postsynaptic density 95 (PSD95) recently were identified in rat cerebral VSMCs. Primarily characterized in neurons, the PSD95 scaffold has more than 50 known binding partners, and it can mediate macromolecular signaling between cell-surface receptors and ion channels. In cerebral arteries, Shaker-type Kv 1 channels appear to associate with the PSD95 molecular scaffold, and PSD95 is required for the normal expression and vasodilator influence of members of this K+ channel gene family. Furthermore, recent findings suggest that the β1-subtype adrenergic receptor is expressed in cerebral VSMCs and forms a functional vasodilator complex with Kv 1 channels on the PSD95 scaffold. Activation of β1-subtype adrenergic receptors in VSMCs enables protein kinase A-dependent phosphorylation and opening of Kv 1 channels in the PSD95 complex; the subsequent K+ efflux mediates membrane hyperpolarization and vasodilation of small cerebral arteries. Early evidence from other studies suggests that other families of Kv channels and scaffolding proteins are expressed in VSMCs. Future investigations into these macromolecular complexes that modulate the expression and function of Kv channels may reveal unknown signaling cascades that regulate VSMC excitability and provide novel targets for ion channel-based medications to optimize vascular tone.
Collapse
Affiliation(s)
- Sung W Rhee
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Nancy J Rusch
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|
45
|
Purkey AM, Woolfrey KM, Crosby KC, Stich DG, Chick WS, Aoto J, Dell'Acqua ML. AKAP150 Palmitoylation Regulates Synaptic Incorporation of Ca 2+-Permeable AMPA Receptors to Control LTP. Cell Rep 2018; 25:974-987.e4. [PMID: 30355502 PMCID: PMC6263960 DOI: 10.1016/j.celrep.2018.09.085] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 09/06/2018] [Accepted: 09/25/2018] [Indexed: 11/22/2022] Open
Abstract
Ca2+-permeable AMPA-type glutamate receptors (CP-AMPARs) containing GluA1 but lacking GluA2 subunits contribute to multiple forms of synaptic plasticity, including long-term potentiation (LTP), but mechanisms regulating CP-AMPARs are poorly understood. A-kinase anchoring protein (AKAP) 150 scaffolds kinases and phosphatases to regulate GluA1 phosphorylation and trafficking, and trafficking of AKAP150 itself is modulated by palmitoylation on two Cys residues. Here, we developed a palmitoylation-deficient knockin mouse to show that AKAP150 palmitoylation regulates CP-AMPAR incorporation at hippocampal synapses. Using biochemical, super-resolution imaging, and electrophysiological approaches, we found that palmitoylation promotes AKAP150 localization to recycling endosomes and the postsynaptic density (PSD) to limit CP-AMPAR basal synaptic incorporation. In addition, we found that AKAP150 palmitoylation is required for LTP induced by weaker stimulation that recruits CP-AMPARs to synapses but not stronger stimulation that recruits GluA2-containing AMPARs. Thus, AKAP150 palmitoylation controls its subcellular localization to maintain proper basal and activity-dependent regulation of synaptic AMPAR subunit composition.
Collapse
Affiliation(s)
- Alicia M Purkey
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Kevin M Woolfrey
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Kevin C Crosby
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Dominik G Stich
- Advanced Light Microscopy Core, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Wallace S Chick
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Jason Aoto
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Mark L Dell'Acqua
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045, USA; Advanced Light Microscopy Core, University of Colorado School of Medicine, Aurora, CO 80045, USA.
| |
Collapse
|
46
|
Patriarchi T, Buonarati OR, Hell JW. Postsynaptic localization and regulation of AMPA receptors and Cav1.2 by β2 adrenergic receptor/PKA and Ca 2+/CaMKII signaling. EMBO J 2018; 37:e99771. [PMID: 30249603 PMCID: PMC6187224 DOI: 10.15252/embj.201899771] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Revised: 07/25/2018] [Accepted: 08/17/2018] [Indexed: 11/09/2022] Open
Abstract
The synapse transmits, processes, and stores data within its tiny space. Effective and specific signaling requires precise alignment of the relevant components. This review examines current insights into mechanisms of AMPAR and NMDAR localization by PSD-95 and their spatial distribution at postsynaptic sites to illuminate the structural and functional framework of postsynaptic signaling. It subsequently delineates how β2 adrenergic receptor (β2 AR) signaling via adenylyl cyclase and the cAMP-dependent protein kinase PKA is organized within nanodomains. Here, we discuss targeting of β2 AR, adenylyl cyclase, and PKA to defined signaling complexes at postsynaptic sites, i.e., AMPARs and the L-type Ca2+ channel Cav1.2, and other subcellular surface localizations, the role of A kinase anchor proteins, the physiological relevance of the spatial restriction of corresponding signaling, and their interplay with signal transduction by the Ca2+- and calmodulin-dependent kinase CaMKII How localized and specific signaling by cAMP occurs is a central cellular question. The dendritic spine constitutes an ideal paradigm for elucidating the dimensions of spatially restricted signaling because of their small size and defined protein composition.
Collapse
MESH Headings
- Animals
- Calcium Channels, L-Type/genetics
- Calcium Channels, L-Type/metabolism
- Calcium Signaling/physiology
- Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics
- Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism
- Cyclic AMP-Dependent Protein Kinases/genetics
- Cyclic AMP-Dependent Protein Kinases/metabolism
- Humans
- Receptors, AMPA/genetics
- Receptors, AMPA/metabolism
- Receptors, Adrenergic, beta-2/genetics
- Receptors, Adrenergic, beta-2/metabolism
- Receptors, N-Methyl-D-Aspartate/genetics
- Receptors, N-Methyl-D-Aspartate/metabolism
- Synapses/genetics
- Synapses/metabolism
Collapse
Affiliation(s)
- Tommaso Patriarchi
- Department of Pharmacology, University of California, Davis, CA, USA
- Department of Biochemistry and Molecular Medicine, University of California, Davis, CA, USA
| | | | - Johannes W Hell
- Department of Pharmacology, University of California, Davis, CA, USA
| |
Collapse
|
47
|
Nguyen PV, Gelinas JN. Noradrenergic gating of long-lasting synaptic potentiation in the hippocampus: from neurobiology to translational biomedicine. J Neurogenet 2018; 32:171-182. [DOI: 10.1080/01677063.2018.1497630] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Peter V. Nguyen
- Department of Physiology and Institute of Neuroscience & Mental Health, University of Alberta School of Medicine, Edmonton, Canada
| | - Jennifer N. Gelinas
- Department of Neurology and Institute for Genomic Medicine, College of Physicians & Surgeons of Columbia University, New York, NY,USA
| |
Collapse
|
48
|
Penny CJ, Gold MG. Mechanisms for localising calcineurin and CaMKII in dendritic spines. Cell Signal 2018; 49:46-58. [DOI: 10.1016/j.cellsig.2018.05.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 05/23/2018] [Accepted: 05/24/2018] [Indexed: 10/14/2022]
|
49
|
Guercio LA, Hofmann ME, Swinford-Jackson SE, Sigman JS, Wimmer ME, Dell'Acqua ML, Schmidt HD, Pierce RC. A-Kinase Anchoring Protein 150 (AKAP150) Promotes Cocaine Reinstatement by Increasing AMPA Receptor Transmission in the Accumbens Shell. Neuropsychopharmacology 2018; 43:1395-1404. [PMID: 29317777 PMCID: PMC5916366 DOI: 10.1038/npp.2017.297] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 11/29/2017] [Accepted: 11/30/2017] [Indexed: 11/08/2022]
Abstract
Previous work indicated that activation of D1-like dopamine receptors (D1DRs) in the nucleus accumbens shell promoted cocaine seeking through a process involving the activation of PKA and GluA1-containing AMPA receptors (AMPARs). A-kinase anchoring proteins (AKAPs) localize PKA to AMPARs leading to enhanced phosphorylation of GluA1. AKAP150, the most well-characterized isoform, plays an important role in several forms of neuronal plasticity. However, its involvement in drug addiction has been minimally explored. Here we examine the role of AKAP150 in cocaine reinstatement, an animal model of relapse. We show that blockade of PKA binding to AKAPs in the nucleus accumbens shell of Sprague-Dawley rats attenuates reinstatement induced by either cocaine or a D1DR agonist. Moreover, this effect is specific to AKAP150, as viral overexpression of a PKA-binding deficient mutant of AKAP150 also impairs cocaine reinstatement. This viral-mediated attenuation of cocaine reinstatement was accompanied by decreased phosphorylation of GluA1-containing AMPARs and attenuated AMPAR eEPSCs. Collectively, these results suggest that AKAP150 facilitates the reinstatement of cocaine-seeking behavior by amplifying D1DR/PKA-dependent AMPA transmission in the nucleus accumbens.
Collapse
Affiliation(s)
- Leonardo A Guercio
- Neuroscience Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Neurobiology and Behavior, Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mackenzie E Hofmann
- Center for Neurobiology and Behavior, Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sarah E Swinford-Jackson
- Center for Neurobiology and Behavior, Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Julia S Sigman
- Center for Neurobiology and Behavior, Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mathieu E Wimmer
- Department of Psychology, Temple University, Philadelphia, PA, USA
| | - Mark L Dell'Acqua
- Department of Pharmacology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Heath D Schmidt
- Department for Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, PA, USA
| | - R Christopher Pierce
- Center for Neurobiology and Behavior, Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
50
|
Coley AA, Gao WJ. PSD95: A synaptic protein implicated in schizophrenia or autism? Prog Neuropsychopharmacol Biol Psychiatry 2018; 82:187-194. [PMID: 29169997 PMCID: PMC5801047 DOI: 10.1016/j.pnpbp.2017.11.016] [Citation(s) in RCA: 211] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 10/27/2017] [Accepted: 11/18/2017] [Indexed: 11/25/2022]
Abstract
The molecular components of the postsynaptic density (PSD) in excitatory synapses of the brain are currently being investigated as one of the major etiologies of neurodevelopmental disorders such as schizophrenia (SCZ) and autism. Postsynaptic density protein-95 (PSD-95) is a major regulator of synaptic maturation by interacting, stabilizing and trafficking N-methyl-d-aspartic acid receptors (NMDARs) and α-amino-3-hydroxy-5-methyl-4-isox-azoleproprionic acid receptors (AMPARs) to the postsynaptic membrane. Recently, there has been overwhelming evidence that associates PSD-95 disruption with cognitive and learning deficits observed in SCZ and autism. For instance, recent genomic and sequencing studies of psychiatric patients highlight the aberrations at the PSD of glutamatergic synapses that include PSD-95 dysfunction. In animal studies, PSD-95 deficiency shows alterations in NMDA and AMPA-receptor composition and function in specific brain regions that may contribute to phenotypes observed in neuropsychiatric pathologies. In this review, we describe the role of PSD-95 as an essential scaffolding protein during synaptogenesis and neurodevelopment. More specifically, we discuss its interactions with NMDA receptor subunits that potentially affect glutamate transmission, and the formation of silent synapses during critical time points of neurodevelopment. Furthermore, we describe how PSD-95 may alter dendritic spine morphologies, thus regulating synaptic function that influences behavioral phenotypes in SCZ versus autism. Understanding the role of PSD-95 in the neuropathologies of SCZ and autism will give an insight of the cellular and molecular attributes in the disorders, thus providing treatment options in patients affected.
Collapse
Affiliation(s)
| | - Wen-Jun Gao
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, United States.
| |
Collapse
|