1
|
de León-López CAM, Carretero-Rey M, Khan ZU. AMPA Receptors in Synaptic Plasticity, Memory Function, and Brain Diseases. Cell Mol Neurobiol 2025; 45:14. [PMID: 39841263 PMCID: PMC11754374 DOI: 10.1007/s10571-024-01529-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 12/26/2024] [Indexed: 01/23/2025]
Abstract
Tetrameric AMPA-type ionotropic glutamate receptors are primary transducers of fast excitatory synaptic transmission in the central nervous system, and their properties and abundance at the synaptic surface are crucial determinants of synaptic efficacy in neuronal communication across the brain. The induction of long-term potentiation (LTP) leads to the insertion of GluA1-containing AMPA receptors at the synaptic surface, whereas during long-term depression (LTD), these receptors are internalized into the cytoplasm of the spine. Disruptions in the trafficking of AMPA receptors to and from the synaptic surface attenuate both forms of synaptic plasticity. Homeostatic scaling up and scaling down, which are additional types of plasticity similar to LTP and LTD, are also regulated by the insertion and removal of GluA1-containing AMPA receptors from the synaptic surface. The trafficking of AMPA receptors is an intricate process assisted by various proteins. Furthermore, AMPA receptors are critical for the formation and consolidation of various types of memory, and alterations in their function are intimately associated with cognitive dysfunction in aging and several neurological and psychiatric diseases. In this review, we will provide an overview of the current understanding of how AMPA receptors regulate various forms of synaptic plasticity, their contribution to memory functions, and their role in aging and brain diseases.
Collapse
Affiliation(s)
- Cristina A Muñoz de León-López
- Laboratory of Neurobiology, Centro de Investigaciones Medico Sanitarias (CIMES), University of Malaga, Calle Marqués de Beccaria, 3, Campus Teatinos s/n, 29010, Malaga, Spain
- Department of Medicine, Faculty of Medicine, University of Malaga, Campus Teatinos s/n, Malaga, Spain
| | - Marta Carretero-Rey
- Laboratory of Neurobiology, Centro de Investigaciones Medico Sanitarias (CIMES), University of Malaga, Calle Marqués de Beccaria, 3, Campus Teatinos s/n, 29010, Malaga, Spain
- Department of Medicine, Faculty of Medicine, University of Malaga, Campus Teatinos s/n, Malaga, Spain
| | - Zafar U Khan
- Laboratory of Neurobiology, Centro de Investigaciones Medico Sanitarias (CIMES), University of Malaga, Calle Marqués de Beccaria, 3, Campus Teatinos s/n, 29010, Malaga, Spain.
- Department of Medicine, Faculty of Medicine, University of Malaga, Campus Teatinos s/n, Malaga, Spain.
- CIBERNED, Institute of Health Carlos III, Madrid, Spain.
| |
Collapse
|
2
|
Zhang W, Zhang J, Zhang Z, Cha S, Li J, Chen L, Wu J, Teng J, Guo G, Zhang J. Effects of DeSUMOylated Spastin on AMPA Receptor Surface Delivery and Synaptic Function Are Enhanced by Phosphorylating at Ser210. Mol Neurobiol 2024; 61:6045-6059. [PMID: 38267753 DOI: 10.1007/s12035-024-03935-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 01/09/2024] [Indexed: 01/26/2024]
Abstract
Surface trafficking of AMPA receptors (AMPARs) is one of the important mechanisms mediating synaptic plasticity which is essential for cognitive functions such as learning and memory. Spastin, as a novel binding partner for the AMPAR, has been reported to regulate AMPAR surface expression and synaptic function. Additionally, Spastin undergoes two posttranslational modifications, phosphorylation and SUMOylation, both of which are crucial for synaptic function. However, gaps exist in our knowledge of how Spastin phosphorylation cross-talks with its SUMOylation in the regulation of AMPAR surface expression and synaptic function. Here, we reported that deSUMOylation of Spastin at Lys427 increased the surface level of AMPAR GluA2 subunit, the amplitude and frequency of miniature excitatory synaptic currents (mEPSC), and facilitated the morphological maturation of dendritic spines in cultured hippocampal neurons. Further studies demonstrated that Spastin phosphorylation at Ser210 further increased the enhancement of GluA2 surface expression and synaptic function by deSUMOylated Spastin, while dephosphorylation had the opposite effect. Simultaneously, deSUMOylation at Lys427 significantly increased the promoting effect of Spastin phosphorylation on synaptic function. In conclusion, our study suggests that cooperative interactions between phosphorylated and deSUMOylated Spastin are novel pathways to enhance synaptic function.
Collapse
Affiliation(s)
- Wenbin Zhang
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou, 510630, China
- Department of Surgery, The First Clinical Medical College, Jinan University, Guangzhou, 510630, China
| | - Jiaqi Zhang
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou, 510630, China
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Zhongqi Zhang
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou, 510630, China
- Department of Anesthesiology, The Affiliated Shunde Hospital of Jinan University, Foshan, 528305, China
| | - Shuhan Cha
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou, 510630, China
| | - Jiong Li
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou, 510630, China
| | - Li Chen
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou, 510630, China
| | - Jiaming Wu
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou, 510630, China
| | - Jijun Teng
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266011, China.
| | - Guoqing Guo
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou, 510630, China.
| | - Jifeng Zhang
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou, 510630, China.
| |
Collapse
|
3
|
Harris KM, Kuwajima M, Flores JC, Zito K. Synapse-specific structural plasticity that protects and refines local circuits during LTP and LTD. Philos Trans R Soc Lond B Biol Sci 2024; 379:20230224. [PMID: 38853547 PMCID: PMC11529630 DOI: 10.1098/rstb.2023.0224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 12/20/2023] [Accepted: 01/05/2024] [Indexed: 06/11/2024] Open
Abstract
Synapses form trillions of connections in the brain. Long-term potentiation (LTP) and long-term depression (LTD) are cellular mechanisms vital for learning that modify the strength and structure of synapses. Three-dimensional reconstruction from serial section electron microscopy reveals three distinct pre- to post-synaptic arrangements: strong active zones (AZs) with tightly docked vesicles, weak AZs with loose or non-docked vesicles, and nascent zones (NZs) with a postsynaptic density but no presynaptic vesicles. Importantly, LTP can be temporarily saturated preventing further increases in synaptic strength. At the onset of LTP, vesicles are recruited to NZs, converting them to AZs. During recovery of LTP from saturation (1-4 h), new NZs form, especially on spines where AZs are most enlarged by LTP. Sentinel spines contain smooth endoplasmic reticulum (SER), have the largest synapses and form clusters with smaller spines lacking SER after LTP recovers. We propose a model whereby NZ plasticity provides synapse-specific AZ expansion during LTP and loss of weak AZs that drive synapse shrinkage during LTD. Spine clusters become functionally engaged during LTP or disassembled during LTD. Saturation of LTP or LTD probably acts to protect recently formed memories from ongoing plasticity and may account for the advantage of spaced over massed learning. This article is part of a discussion meeting issue 'Long-term potentiation: 50 years on'.
Collapse
Affiliation(s)
- Kristen M. Harris
- Department of Neuroscience and Center for Learning and Memory, The University of Texas at Austin, Austin, TX78712, USA
| | - Masaaki Kuwajima
- Department of Neuroscience and Center for Learning and Memory, The University of Texas at Austin, Austin, TX78712, USA
| | - Juan C. Flores
- Center for Neuroscience, University of California, Davis, CA95618, USA
| | - Karen Zito
- Center for Neuroscience, University of California, Davis, CA95618, USA
| |
Collapse
|
4
|
Hepburn I, Lallouette J, Chen W, Gallimore AR, Nagasawa-Soeda SY, De Schutter E. Vesicle and reaction-diffusion hybrid modeling with STEPS. Commun Biol 2024; 7:573. [PMID: 38750123 PMCID: PMC11096338 DOI: 10.1038/s42003-024-06276-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 05/01/2024] [Indexed: 05/18/2024] Open
Abstract
Vesicles carry out many essential functions within cells through the processes of endocytosis, exocytosis, and passive and active transport. This includes transporting and delivering molecules between different parts of the cell, and storing and releasing neurotransmitters in neurons. To date, computational simulation of these key biological players has been rather limited and has not advanced at the same pace as other aspects of cell modeling, restricting the realism of computational models. We describe a general vesicle modeling tool that has been designed for wide application to a variety of cell models, implemented within our software STochastic Engine for Pathway Simulation (STEPS), a stochastic reaction-diffusion simulator that supports realistic reconstructions of cell tissue in tetrahedral meshes. The implementation is validated in an extensive test suite, parallel performance is demonstrated in a realistic synaptic bouton model, and example models are visualized in a Blender extension module.
Collapse
Affiliation(s)
- Iain Hepburn
- Computational Neuroscience Unit, Okinawa Institute of Science and Technology, 1919-1 Tancha, Onna-son, Okinawa, Japan
| | - Jules Lallouette
- Computational Neuroscience Unit, Okinawa Institute of Science and Technology, 1919-1 Tancha, Onna-son, Okinawa, Japan
| | - Weiliang Chen
- Computational Neuroscience Unit, Okinawa Institute of Science and Technology, 1919-1 Tancha, Onna-son, Okinawa, Japan
| | - Andrew R Gallimore
- Computational Neuroscience Unit, Okinawa Institute of Science and Technology, 1919-1 Tancha, Onna-son, Okinawa, Japan
| | - Sarah Y Nagasawa-Soeda
- Computational Neuroscience Unit, Okinawa Institute of Science and Technology, 1919-1 Tancha, Onna-son, Okinawa, Japan
| | - Erik De Schutter
- Computational Neuroscience Unit, Okinawa Institute of Science and Technology, 1919-1 Tancha, Onna-son, Okinawa, Japan.
| |
Collapse
|
5
|
Li G, Xu X, Yang L, Cai Y, Sun Y, Guo J, Lin Y, Hu Y, Chen M, Li H, Wu S. Exploring the association between circRNA expression and pediatric obesity based on a case-control study and related bioinformatics analysis. BMC Pediatr 2023; 23:561. [PMID: 37957626 PMCID: PMC10642011 DOI: 10.1186/s12887-023-04261-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 08/21/2023] [Indexed: 11/15/2023] Open
Abstract
OBJECTIVE Our present study utilized case-control research to explore the relationship between specific circRNAs and pediatric obesity through a literature review and bioinformatics and to predict their possible biological functions, providing ideas for epigenetic mechanism studies of pediatric obesity. METHODS CircRNAs related to pediatric obesity were preliminarily screened by a literature review and qRT-PCR. CircRNA expression in children with obesity (n = 75) and control individuals (n = 75) was confirmed with qRT-PCR in a case-control study. This was followed by bioinformatics analyses, such as GO analysis, KEGG pathway analysis, and ceRNA network construction. Multivariate logistic regression was utilized to analyze the effects of circRNAs on obesity. A receiver operating characteristic (ROC) curve was also drawn to explore the clinical application value of circRNAs in pediatric obesity. RESULTS Has_circ_0046367 and hsa_circ_0000284 were separately validated to be statistically downregulated and upregulated, respectively, in the peripheral blood mononuclear cells of children with obesity and revealed as independent indicators of increased CHD risk [hsa_circ_0046367 (OR = 0.681, 95% CI: 0.480 ~ 0.967) and hsa_circ_0000284 (OR = 1.218, 95% CI: 1.041 ~ 1.424)]. The area under the ROC curve in the combined analysis of hsa_circ_0046367 and hsa_circ_0000284 was 0.706 (95% CI: 0.623 ~ 0.789). Enrichment analyses revealed that these circRNAs were actively involved in neural plasticity mechanisms, cell secretion and signal regulation. CONCLUSION The present research revealed that low expression of hsa_circ_0046367 and high expression of hsa_circ_0000284 are risk factors for pediatric obesity and that neural plasticity mechanisms are closely related to obesity.
Collapse
Affiliation(s)
- Guobo Li
- Department of Child Healthcare Centre, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, Fuzhou, 350001, China
| | - Xingyan Xu
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Minhou County, Fuzhou, 350122, China
| | - Le Yang
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Minhou County, Fuzhou, 350122, China
| | - Yingying Cai
- Department of Developmental and Behavioral Pediatrics, Fujian Children's Hospital, Fujian, 350014, China
| | - Yi Sun
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Minhou County, Fuzhou, 350122, China
| | - Jianhui Guo
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Minhou County, Fuzhou, 350122, China
| | - Yawen Lin
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Minhou County, Fuzhou, 350122, China
| | - Yuduan Hu
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Minhou County, Fuzhou, 350122, China
| | - Mingjun Chen
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Minhou County, Fuzhou, 350122, China
| | - Huangyuan Li
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Minhou County, Fuzhou, 350122, China.
| | - Siying Wu
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Minhou County, Fuzhou, 350122, China.
| |
Collapse
|
6
|
Georgiev SV, Rizzoli SO. The long-loop recycling (LLR) of synaptic components as a question of economics. Mol Cell Neurosci 2023; 126:103862. [PMID: 37236414 DOI: 10.1016/j.mcn.2023.103862] [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: 02/28/2023] [Revised: 05/15/2023] [Accepted: 05/19/2023] [Indexed: 05/28/2023] Open
Abstract
The pre- and post-synaptic compartments contain a variety of molecules that are known to recycle between the plasma membrane and intracellular organelles. The recycling steps have been amply described in functional terms, with, for example, synaptic vesicle recycling being essential for neurotransmitter release, and postsynaptic receptor recycling being a fundamental feature of synaptic plasticity. However, synaptic protein recycling may also serve a more prosaic role, simply ensuring the repeated use of specific components, thereby minimizing the energy expenditure on the synthesis of synaptic proteins. This type of process has been recently described for components of the extracellular matrix, which undergo long-loop recycling (LLR), to and from the cell body. Here we suggest that the energy-saving recycling of synaptic components may be more widespread than is generally acknowledged, potentially playing a role in both synaptic vesicle protein usage and postsynaptic receptor metabolism.
Collapse
Affiliation(s)
- Svilen Veselinov Georgiev
- University Medical Center Göttingen, Institute for Neuro- and Sensory Physiology, Germany; International Max Planck Research School for Neuroscience, Göttingen, Germany.
| | - Silvio O Rizzoli
- University Medical Center Göttingen, Institute for Neuro- and Sensory Physiology, Germany; Biostructural Imaging of Neurodegeneration (BIN) Center, Göttingen, Germany; Excellence Cluster Multiscale Bioimaging, Göttingen, Germany.
| |
Collapse
|
7
|
Guntupalli S, Park P, Han DH, Zhang L, Yong XLH, Ringuet M, Blackmore DG, Jhaveri DJ, Koentgen F, Widagdo J, Kaang BK, Anggono V. Ubiquitination of the GluA1 Subunit of AMPA Receptors Is Required for Synaptic Plasticity, Memory, and Cognitive Flexibility. J Neurosci 2023; 43:5448-5457. [PMID: 37419688 PMCID: PMC10376930 DOI: 10.1523/jneurosci.1542-22.2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 06/14/2023] [Accepted: 06/28/2023] [Indexed: 07/09/2023] Open
Abstract
Activity-dependent changes in the number of AMPA-type glutamate receptors (AMPARs) at the synapse underpin the expression of LTP and LTD, cellular correlates of learning and memory. Post-translational ubiquitination has emerged as a key regulator of the trafficking and surface expression of AMPARs, with ubiquitination of the GluA1 subunit at Lys-868 controlling the post-endocytic sorting of the receptors into the late endosome for degradation, thereby regulating their stability at synapses. However, the physiological significance of GluA1 ubiquitination remains unknown. In this study, we generated mice with a knock-in mutation in the major GluA1 ubiquitination site (K868R) to investigate the role of GluA1 ubiquitination in synaptic plasticity, learning, and memory. Our results reveal that these male mice have normal basal synaptic transmission but exhibit enhanced LTP and deficits in LTD. They also display deficits in short-term spatial memory and cognitive flexibility. These findings underscore the critical roles of GluA1 ubiquitination in bidirectional synaptic plasticity and cognition in male mice.SIGNIFICANCE STATEMENT Subcellular targeting and membrane trafficking determine the precise number of AMPA-type glutamate receptors at synapses, processes that are essential for synaptic plasticity, learning, and memory. Post-translational ubiquitination of the GluA1 subunit marks AMPARs for degradation, but its functional role in vivo remains unknown. Here we demonstrate that the GluA1 ubiquitin-deficient mice exhibit an altered threshold for synaptic plasticity accompanied by deficits in short-term memory and cognitive flexibility. Our findings suggest that activity-dependent ubiquitination of GluA1 fine-tunes the optimal number of synaptic AMPARs required for bidirectional synaptic plasticity and cognition in male mice. Given that increases in amyloid-β cause excessive ubiquitination of GluA1, inhibiting that GluA1 ubiquitination may have the potential to ameliorate amyloid-β-induced synaptic depression in Alzheimer's disease.
Collapse
Affiliation(s)
- Sumasri Guntupalli
- Clem Jones Centre for Ageing Dementia Research, University of Queensland, Brisbane, Queensland 4072, Australia
- Queensland Brain Institute, University of Queensland, Brisbane, Queensland 4072, Australia
| | - Pojeong Park
- School of Biological Sciences, Seoul National University, Seoul, 08826, Korea
| | - Dae Hee Han
- School of Biological Sciences, Seoul National University, Seoul, 08826, Korea
| | - Lingrui Zhang
- Clem Jones Centre for Ageing Dementia Research, University of Queensland, Brisbane, Queensland 4072, Australia
- Queensland Brain Institute, University of Queensland, Brisbane, Queensland 4072, Australia
| | - Xuan Ling Hilary Yong
- Clem Jones Centre for Ageing Dementia Research, University of Queensland, Brisbane, Queensland 4072, Australia
- Queensland Brain Institute, University of Queensland, Brisbane, Queensland 4072, Australia
| | - Mitchell Ringuet
- Clem Jones Centre for Ageing Dementia Research, University of Queensland, Brisbane, Queensland 4072, Australia
- Queensland Brain Institute, University of Queensland, Brisbane, Queensland 4072, Australia
| | - Daniel G Blackmore
- Clem Jones Centre for Ageing Dementia Research, University of Queensland, Brisbane, Queensland 4072, Australia
- Queensland Brain Institute, University of Queensland, Brisbane, Queensland 4072, Australia
| | - Dhanisha J Jhaveri
- Queensland Brain Institute, University of Queensland, Brisbane, Queensland 4072, Australia
- Mater Research Institute, University of Queensland, Brisbane, Queensland 4072, Australia
| | - Frank Koentgen
- Ozgene Pty Ltd, Bentley DC, Western Australia 6983, Australia
| | - Jocelyn Widagdo
- Clem Jones Centre for Ageing Dementia Research, University of Queensland, Brisbane, Queensland 4072, Australia
- Queensland Brain Institute, University of Queensland, Brisbane, Queensland 4072, Australia
| | - Bong-Kiun Kaang
- School of Biological Sciences, Seoul National University, Seoul, 08826, Korea
| | - Victor Anggono
- Clem Jones Centre for Ageing Dementia Research, University of Queensland, Brisbane, Queensland 4072, Australia
- Queensland Brain Institute, University of Queensland, Brisbane, Queensland 4072, Australia
| |
Collapse
|
8
|
Dysregulation of AMPA Receptor Trafficking and Intracellular Vesicular Sorting in the Prefrontal Cortex of Dopamine Transporter Knock-Out Rats. Biomolecules 2023; 13:biom13030516. [PMID: 36979451 PMCID: PMC10046215 DOI: 10.3390/biom13030516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/23/2023] [Accepted: 03/08/2023] [Indexed: 03/18/2023] Open
Abstract
Dopamine (DA) and glutamate interact, influencing neural excitability and promoting synaptic plasticity. However, little is known regarding the molecular mechanisms underlying this crosstalk. Since perturbation of DA-AMPA receptor interaction might sustain pathological conditions, the major aim of our work was to evaluate the effect of the hyperactive DA system on the AMPA subunit composition, trafficking, and membrane localization in the prefrontal cortex (PFC). Taking advantage of dopamine transporter knock-out (DAT−/−) rats, we found that DA overactivity reduced the translation of cortical AMPA receptors and their localization at both synaptic and extra-synaptic sites through, at least in part, altered intracellular vesicular sorting. Moreover, the reduced expression of AMPA receptor-specific anchoring proteins and structural markers, such as Neuroligin-1 and nCadherin, likely indicate a pattern of synaptic instability. Overall, these data reveal that a condition of hyperdopaminergia markedly alters the homeostatic plasticity of AMPA receptors, suggesting a general destabilization and depotentiation of the AMPA-mediated glutamatergic neurotransmission in the PFC. This effect might be functionally relevant for disorders characterized by elevated dopaminergic activity.
Collapse
|
9
|
Bencsik N, Oueslati Morales CO, Hausser A, Schlett K. Endocytosis of AMPA receptors: Role in neurological conditions. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 196:59-97. [PMID: 36813366 DOI: 10.1016/bs.pmbts.2022.09.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
AMPA receptors are glutamate-gated ion channels, present in a wide range of neuron types and in glial cells. Their main role is to mediate fast excitatory synaptic transmission, and therefore, they are critical for normal brain function. In neurons, AMPA receptors undergo constitutive and activity-dependent trafficking between the synaptic, extrasynaptic and intracellular pools. The kinetics of AMPA receptor trafficking is crucial for the precise functioning of both individual neurons and neural networks involved in information processing and learning. Many of the neurological diseases evoked by neurodevelopmental and neurodegenerative malfunctions or traumatic injuries are caused by impaired synaptic function in the central nervous system. For example, attention-deficit/hyperactivity disorder (ADHD), Alzheimer's disease (AD), tumors, seizures, ischemic strokes, and traumatic brain injury are all characterized by impaired glutamate homeostasis and associated neuronal death, typically caused by excitotoxicity. Given the important role of AMPA receptors in neuronal function, it is not surprising that perturbations in AMPA receptor trafficking are associated with these neurological disorders. In this book chapter, we will first introduce the structure, physiology and synthesis of AMPA receptors, followed by an in-depth description of the molecular mechanisms that control AMPA receptor endocytosis and surface levels under basal conditions or synaptic plasticity. Finally, we will discuss how impairments in AMPA receptor trafficking, particularly endocytosis, contribute to the pathophysiology of various neurological disorders and what efforts are being made to therapeutically target this process.
Collapse
Affiliation(s)
- Norbert Bencsik
- Neuronal Cell Biology Research Group, Department of Physiology and Neurobiology, Eötvös Loránd University, Budapest, Hungary
| | - Carlos Omar Oueslati Morales
- Membrane Trafficking and Signalling Group, Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Angelika Hausser
- Membrane Trafficking and Signalling Group, Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany; Stuttgart Research Center Systems Biology, University of Stuttgart, Stuttgart, Germany
| | - Katalin Schlett
- Neuronal Cell Biology Research Group, Department of Physiology and Neurobiology, Eötvös Loránd University, Budapest, Hungary.
| |
Collapse
|
10
|
Mendoza ML, Quigley LD, Dunham T, Volk LJ. KIBRA regulates activity-induced AMPA receptor expression and synaptic plasticity in an age-dependent manner. iScience 2022; 25:105623. [PMID: 36465112 PMCID: PMC9713372 DOI: 10.1016/j.isci.2022.105623] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 10/14/2022] [Accepted: 11/15/2022] [Indexed: 11/18/2022] Open
Abstract
A growing body of human literature implicates KIBRA in memory and neurodevelopmental disorders. Memory and the cellular substrates supporting adaptive cognition change across development. Using an inducible KIBRA knockout mouse, we demonstrate that adult-onset deletion of KIBRA in forebrain neurons impairs long-term spatial memory and long-term potentiation (LTP). These LTP deficits correlate with adult-selective decreases in extrasynaptic AMPA receptors under basal conditions, and we identify a role for KIBRA in LTP-induced AMPAR upregulation. In contrast, juvenile-onset deletion of KIBRA in forebrain neurons did not affect LTP and had minimal effects on basal AMPAR expression. LTP did not increase AMPAR protein expression in juvenile WT mice, providing a potential explanation for juvenile resilience to KIBRA deletion. These data suggest that KIBRA serves a unique role in adult hippocampal function through regulation of basal and activity-dependent AMPAR proteostasis that supports synaptic plasticity.
Collapse
Affiliation(s)
- Matthew L. Mendoza
- Neuroscience Graduate Program, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Lilyana D. Quigley
- Neuroscience Graduate Program, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Thomas Dunham
- Neuroscience Graduate Program, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Lenora J. Volk
- Neuroscience Graduate Program, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Peter O’ Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
11
|
Wu QL, Gao Y, Li JT, Ma WY, Chen NH. The Role of AMPARs Composition and Trafficking in Synaptic Plasticity and Diseases. Cell Mol Neurobiol 2022; 42:2489-2504. [PMID: 34436728 PMCID: PMC11421597 DOI: 10.1007/s10571-021-01141-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 08/11/2021] [Indexed: 11/28/2022]
Abstract
AMPA receptors are tetrameric ionic glutamate receptors, which mediate 90% fast excitatory synaptic transmission induced by excitatory glutamate in the mammalian central nervous system through the activation or inactivation of ion channels. The alternation of synaptic AMPA receptor number and subtype is thought to be one of the primary mechanisms that involve in synaptic plasticity regulation and affect the functions in learning, memory, and cognition. The increasing of surface AMPARs enhances synaptic strength during long-term potentiation, whereas the decreasing of AMPARs weakens synaptic strength during the long-term depression. It is closely related to the AMPA receptor as well as its subunits assembly, trafficking, and degradation. The dysfunction of any step in these precise regulatory processes is likely to induce the disorder of synaptic transmission and loss of neurons, or even cause neuropsychiatric diseases ultimately. Therefore, it is useful to understand how AMPARs regulate synaptic plasticity and its role in related neuropsychiatric diseases via comprehending architecture and trafficking of the receptors. Here, we reviewed the progress in structure, expression, trafficking, and relationship with synaptic plasticity of AMPA receptor, especially in anxiety, depression, neurodegenerative disorders, and cerebral ischemia.
Collapse
Affiliation(s)
- Qing-Lin Wu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Yan Gao
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Jun-Tong Li
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Wen-Yu Ma
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Nai-Hong Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| |
Collapse
|
12
|
Hayashi T. Membrane lipid rafts are required for AMPA receptor tyrosine phosphorylation. Front Synaptic Neurosci 2022; 14:921772. [PMID: 36387774 PMCID: PMC9662747 DOI: 10.3389/fnsyn.2022.921772] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 10/11/2022] [Indexed: 12/24/2023] Open
Abstract
Membrane lipid rafts are sphingolipids and cholesterol-enriched membrane microdomains, which form a center for the interaction or assembly of palmitoylated signaling molecules, including Src family non-receptor type protein tyrosine kinases. Lipid rafts abundantly exist in neurons and function in the maintenance of synapses. Excitatory synaptic strength is largely controlled by the surface expression of α-amino-3-hydroxy-5-methyl-4-isoxazole propionate (AMPA) receptors in the mammalian brain. AMPA receptor endocytosis from the synaptic surface is regulated by phosphorylation of the GluA2 subunit at tyrosine 876 by Src family kinases. Here, I revealed that tyrosine phosphorylated GluA2 is concentrated in the lipid rafts fraction. Furthermore, stimulation-induced upregulation of GluA2 tyrosine phosphorylation is disrupted by the treatment of neurons with a cholesterol-depleting compound, filipin III. These results indicate the importance of lipid rafts as enzymatic reactive sites for AMPA receptor tyrosine phosphorylation and subsequent AMPA receptor internalization from the synaptic surface.
Collapse
Affiliation(s)
- Takashi Hayashi
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology, Tsukuba, Ibaraki, Japan
- Department of Molecular Neurobiology and Pharmacology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
13
|
Stachowicz K. Is PSD-95 entangled in the side effects of antidepressants? Neurochem Int 2022; 159:105391. [PMID: 35817245 DOI: 10.1016/j.neuint.2022.105391] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 07/04/2022] [Accepted: 07/05/2022] [Indexed: 01/13/2023]
Abstract
PSD-95 is a component and a building block of an excitatory synapse. PSD-95 is a specialized protein that is part of a "combination lock" system responsible for plastic events at the synapse, such as receptor expression, which consequently induces changes in the PSD structure and thus affects synaptic plasticity. The possible involvement of PSD-95 in antidepressant side effects related to cognitive function and psychosis will be considered. An attempt will be made to trace the sequence of events in the proposed mechanism leading to these disorders, focusing mainly on NMDA receptors. Understanding the mechanisms of action of compounds with antidepressant potential may facilitate the design of safer drugs.
Collapse
Affiliation(s)
- Katarzyna Stachowicz
- Department of Neurobiology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna, 12, 31-343, Kraków, Poland.
| |
Collapse
|
14
|
Gao AYL, Lourdin-De Filippis E, Orlowski J, McKinney RA. Roles of Endomembrane Alkali Cation/Proton Exchangers in Synaptic Function and Neurodevelopmental Disorders. Front Physiol 2022; 13:892196. [PMID: 35547574 PMCID: PMC9081726 DOI: 10.3389/fphys.2022.892196] [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: 03/08/2022] [Accepted: 03/30/2022] [Indexed: 12/25/2022] Open
Abstract
Endomembrane alkali cation (Na+, K+)/proton (H+) exchangers (eNHEs) are increasingly associated with neurological disorders. These eNHEs play integral roles in regulating the luminal pH, processing, and trafficking of cargo along the secretory (Golgi and post-Golgi vesicles) and endocytic (early, recycling, and late endosomes) pathways, essential regulatory processes vital for neuronal development and plasticity. Given the complex morphology and compartmentalization of multipolar neurons, the contribution of eNHEs in maintaining optimal pH homeostasis and cargo trafficking is especially significant during periods of structural and functional development and remodeling. While the importance of eNHEs has been demonstrated in a variety of non-neuronal cell types, their involvement in neuronal function is less well understood. In this review, we will discuss their emerging roles in excitatory synaptic function, particularly as it pertains to cellular learning and remodeling. We will also explore their connections to neurodevelopmental conditions, including intellectual disability, autism, and attention deficit hyperactivity disorders.
Collapse
Affiliation(s)
- Andy Y L Gao
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada.,Department of Pharmacology & Therapeutics, McGill University, Montreal, QC, Canada
| | | | - John Orlowski
- Department of Physiology, McGill University, Montreal, QC, Canada
| | - R Anne McKinney
- Department of Pharmacology & Therapeutics, McGill University, Montreal, QC, Canada
| |
Collapse
|
15
|
Chen L, Wang H, Cha S, Li J, Zhang J, Wu J, Guo G, Zhang J. Phosphorylation of Spastin Promotes the Surface Delivery and Synaptic Function of AMPA Receptors. Front Cell Neurosci 2022; 16:809934. [PMID: 35418834 PMCID: PMC8995424 DOI: 10.3389/fncel.2022.809934] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 03/07/2022] [Indexed: 12/14/2022] Open
Abstract
Synaptic plasticity is essential for cognitive functions such as learning and memory. One of the mechanisms involved in synaptic plasticity is the dynamic delivery of AMPA receptors (AMPARs) in and out of synapses. Mutations of SPAST, which encodes SPASTIN, a microtubule-severing protein, are considered the most common cause of hereditary spastic paraparesis (HSP). In some cases, patients with HSP also manifest cognitive impairment. In addition, mice with Spastin depletion exhibit working and associative memory deficits and reduced AMPAR levels. However, the exact effect and molecular mechanism of Spastin on AMPARs trafficking has remained unclear. Here, we report that Spastin interacts with AMPAR, and phosphorylation of Spastin enhances its interaction with AMPAR subunit GluA2. Further study shows that phosphorylation of Spastin can increase AMPAR GluA2 surface expression and the amplitude and frequency of miniature excitatory synaptic currents (mEPSC) in cultured hippocampal neurons. Moreover, phosphorylation of Spastin at Ser210 is crucial for GluA2 surface expression. Phosphorylation of Spastin K353A, which obliterates microtubule-severing activity, also promotes AMPAR GluA2 subunit trafficking to the surface and increases the amplitude and frequency of mEPSCs in cultured neurons. Taken together, our data demonstrate that Spastin phosphorylation promotes the surface delivery of the AMPAR GluA2 subunit independent of microtubule dynamics.
Collapse
Affiliation(s)
- Li Chen
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou, China
| | - Hanjie Wang
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou, China
| | - Shuhan Cha
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou, China
| | - Jiong Li
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou, China
| | - Jiaqi Zhang
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou, China
| | - Jiaming Wu
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou, China
- Department of Neurosurgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Guoqing Guo
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou, China
- *Correspondence: Guoqing Guo Jifeng Zhang
| | - Jifeng Zhang
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou, China
- *Correspondence: Guoqing Guo Jifeng Zhang
| |
Collapse
|
16
|
Keifer J. Regulation of AMPAR trafficking in synaptic plasticity by BDNF and the impact of neurodegenerative disease. J Neurosci Res 2022; 100:979-991. [PMID: 35128708 DOI: 10.1002/jnr.25022] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 01/05/2022] [Accepted: 01/08/2022] [Indexed: 02/06/2023]
Abstract
Research demonstrates that the neural mechanisms underlying synaptic plasticity and learning and memory involve mobilization of AMPA-type neurotransmitter receptors at glutamatergic synaptic contacts, and that these mechanisms are targeted during neurodegenerative disease. Strengthening neural transmission occurs with insertion of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPARs) into synapses while weakening results from receptor withdrawal. A key player in the trafficking of AMPARs during plasticity and learning is the brain-derived neurotrophic factor (BDNF) signaling system. BDNF is a neurotrophic factor that supports neuronal growth and is required for learning and memory. Significantly, a primary feature of many neurodegenerative diseases is a reduction in BDNF protein as well as disrupted neuronal surface expression of synaptic AMPARs. The resulting weakening of synaptic contacts leads to synapse loss and neuronal degeneration that underlies the cognitive impairment and dementia observed in patients with progressive neurodegenerative disease such as Alzheimer's. In the face of these data, one therapeutic approach is to increase BDNF bioavailability in brain. While this has been met with significant challenges, the results of the research have been promising. In spite of this, there are currently no clinical trials to test many of these findings on patients. Here, research showing that BDNF drives AMPARs to synapses, AMPAR trafficking is essential for synaptic plasticity and learning, and that neurodegenerative disease results in a significant decline in BDNF will be reviewed. The aim is to draw attention to the need for increasing patient-directed clinical studies to test the possible benefits of increasing levels of neurotrophins, specifically BDNF, to treat brain disorders. Much is known about the cellular mechanisms that underlie learning and memory in brain. It can be concluded that signaling by neurotrophins like BDNF and AMPA-type glutamate receptor synaptic trafficking are fundamental to these processes. Data from animal models and patients reveal that these mechanisms are adversely targeted during neurodegenerative disease and results in memory loss and cognitive decline. A brief summary of our understanding of these mechanisms indicates that it is time to apply this knowledge base directly to development of therapeutic treatments that enhance neurotrophins for brain disorders in patient populations.
Collapse
Affiliation(s)
- Joyce Keifer
- Neuroscience Group, Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, South Dakota, USA
| |
Collapse
|
17
|
Royo M, Escolano BA, Madrigal MP, Jurado S. AMPA Receptor Function in Hypothalamic Synapses. Front Synaptic Neurosci 2022; 14:833449. [PMID: 35173598 PMCID: PMC8842481 DOI: 10.3389/fnsyn.2022.833449] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 01/03/2022] [Indexed: 12/15/2022] Open
Abstract
AMPA receptors (AMPARs) are critical for mediating glutamatergic synaptic transmission and plasticity, thus playing a major role in the molecular machinery underlying cellular substrates of memory and learning. Their expression pattern, transport and regulatory mechanisms have been extensively studied in the hippocampus, but their functional properties in other brain regions remain poorly understood. Interestingly, electrophysiological and molecular evidence has confirmed a prominent role of AMPARs in the regulation of hypothalamic function. This review summarizes the existing evidence on AMPAR-mediated transmission in the hypothalamus, where they are believed to orchestrate the role of glutamatergic transmission in autonomous, neuroendocrine function, body homeostasis, and social behavior.
Collapse
|
18
|
Zbinden JC, Mirhaidari GJM, Blum KM, Musgrave AJ, Reinhardt JW, Breuer CK, Barker JC. The lysosomal trafficking regulator is necessary for normal wound healing. Wound Repair Regen 2021; 30:82-99. [PMID: 34837653 DOI: 10.1111/wrr.12984] [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: 06/07/2021] [Revised: 11/01/2021] [Accepted: 11/09/2021] [Indexed: 11/29/2022]
Abstract
Non-healing wounds are a major threat to public health throughout the United States. Tissue healing is complex multifactorial process that requires synchronicity of several cell types. Endolysosomal trafficking, which contributes to various cell functions from protein degradation to plasma membrane repair, is an understudied process in the context of wound healing. The lysosomal trafficking regulator protein (LYST) is an essential protein of the endolysosomal system through an indeterminate mechanism. In this study, we examine the impact of impaired LYST function both in vitro with primary LYST mutant fibroblasts as well as in vivo with an excisional wound model. The wound model shows that LYST mutant mice have impaired wound healing in the form of delayed epithelialization and collagen deposition, independent of macrophage infiltration and polarisation. We show that LYST mutation confers a deficit in MCP-1, IGF-1, and IGFBP-2 secretion in beige fibroblasts, which are critical factors in normal wound healing. Identifying the mechanism of LYST function is important for understanding normal wound biology, which may facilitate the development of strategies to address problem wound healing.
Collapse
Affiliation(s)
- Jacob C Zbinden
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA.,Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, USA
| | - Gabriel J M Mirhaidari
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA.,Biomedical Sciences Graduate Program, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Kevin M Blum
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA.,Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, USA
| | - Andrew J Musgrave
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
| | - James W Reinhardt
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Christopher K Breuer
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Jenny C Barker
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA.,Department of Plastic and Reconstructive Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| |
Collapse
|
19
|
Choudhry H, Aggarwal M, Pan PY. Mini-review: Synaptojanin 1 and its implications in membrane trafficking. Neurosci Lett 2021; 765:136288. [PMID: 34637856 PMCID: PMC8572151 DOI: 10.1016/j.neulet.2021.136288] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 10/03/2021] [Accepted: 10/05/2021] [Indexed: 12/04/2022]
Abstract
This mini-review aims to summarize a growing body of literature on synaptojanin 1 (Synj1), a phosphoinositide phosphatase that was initially known to have a prominent role in synaptic vesicle recycling. Synj1 is coded by the SYNJ1 gene, whose mutations and variants are associated with an increasing number of neurological disorders. To better understand the mechanistic role of Synj1 in disease pathogenesis, we review details of phosphoinositide signaling pathways and the reported involvement of Synj1 in membrane trafficking with a specific focus on Parkinson’s disease (PD). Recent studies have tremendously advanced our understanding of Synj1 protein structure and function while broadening our view of how Synj1 regulates synaptic membrane trafficking and endosomal trafficking in various organisms and cell types. A growing body of evidence points to inefficient membrane trafficking as key pathogenic mechanisms in neurodegenerative diseases associated with abnormal Synj1 expression. Despite significant progress made in the field, the mechanism by which Synj1 connects to trafficking, signaling, and pathogenesis is lacking and remains to be addressed.
Collapse
Affiliation(s)
- Hassaam Choudhry
- Dept. of Neuroscience and Cell Biology, Rutgers, Robert Wood Johnson Medical School, 675 Hoes Lane West, Piscataway, NJ 08854, USA
| | - Meha Aggarwal
- Dept. of Neuroscience and Cell Biology, Rutgers, Robert Wood Johnson Medical School, 675 Hoes Lane West, Piscataway, NJ 08854, USA
| | - Ping-Yue Pan
- Dept. of Neuroscience and Cell Biology, Rutgers, Robert Wood Johnson Medical School, 675 Hoes Lane West, Piscataway, NJ 08854, USA.
| |
Collapse
|
20
|
Cheng L, Chen K, Li J, Wu J, Zhang J, Chen L, Guo G, Zhang J. Phosphorylation of CRMP2 by Cdk5 Negatively Regulates the Surface Delivery and Synaptic Function of AMPA Receptors. Mol Neurobiol 2021; 59:762-777. [PMID: 34773219 DOI: 10.1007/s12035-021-02581-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 09/24/2021] [Indexed: 11/28/2022]
Abstract
AMPA receptor mediate most fast excitatory synaptic transmission and play a key role in synaptic plasticity in the central nervous system (CNS) by trafficking and targeting of its subunits to individual postsynaptic membrane. Collapsing response mediator protein 2 (CRMP2), an intracellular phospho-protein, has been reported to promote the maturation of the dendritic spine and transfer AMPA receptors to the membrane. However, our knowledge about the molecular mechanisms of CRMP2 regulating AMPA receptors trafficking is limited. Here, we reported that CRMP2 promoted the surface expression of AMPA receptor GluA1 subunit in cultured hippocampal neurons and in HEK293T cells expressing GluA1 subunits. Furthermore, we found that CRMP2 interacted with GluA1, and their interaction was inhibited by CRMP2 phosphorylation at ser522. Moreover, our results showed that phosphorylation of CRMP2 at ser522 by cyclin-dependent kinase 5 (Cdk5) decreased the fluorescence intensity of surface GluA1 and the amplitude and frequency of miniature excitatory synaptic currents (mEPSCs) in cultured hippocampal neurons, indicating a reduction levels and synaptic function of AMPA receptors. Taken together, our data demonstrated that phosphorylation of CRMP2 by Cdk5 is important for AMPA receptor surface delivery in hippocampal neurons.
Collapse
Affiliation(s)
- Longfei Cheng
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou, 510630, China.,Department of Neurosurgery, The First Affiliated Hospital of Jinan University Guangzhou, Guangzhou, 510630, China
| | - Keen Chen
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou, 510630, China.,Department of Neurosurgery, The First Affiliated Hospital of Jinan University Guangzhou, Guangzhou, 510630, China
| | - Jiong Li
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou, 510630, China
| | - Jiaming Wu
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou, 510630, China.,Department of Neurosurgery, The First Affiliated Hospital of Jinan University Guangzhou, Guangzhou, 510630, China
| | - Jiaqi Zhang
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou, 510630, China
| | - Li Chen
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou, 510630, China
| | - Guoqing Guo
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou, 510630, China.
| | - Jifeng Zhang
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou, 510630, China.
| |
Collapse
|
21
|
Vadakkan KI. Framework for internal sensation of pleasure using constraints from disparate findings in nucleus accumbens. World J Psychiatry 2021; 11:681-695. [PMID: 34733636 PMCID: PMC8546768 DOI: 10.5498/wjp.v11.i10.681] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/27/2021] [Accepted: 09/02/2021] [Indexed: 02/06/2023] Open
Abstract
It is necessary to find a mechanism that generates first-person inner sensation of pleasure to understand what causes addiction and associated behaviour by drugs of abuse. The actual mechanism is expected to explain several disparate findings in nucleus accumbens (NAc), a brain region associated with pleasure, in an interconnected manner. Previously, it was possible to derive a mechanism for natural learning and explain: (1) Generation of inner sensation of memory using changes generated by learning; and (2) Long-term potentiation as an experimental delayed scaled-up change by the same mechanism that occur during natural learning. By extending these findings and by using disparate third person observations in NAc from several studies, present work provides a framework of a mechanism that generates internal sensation of pleasure that can provide interconnected explanations for: (1) Ability to induce robust long-term depression (LTD) in NAc from naïve animals; (2) Impaired ability to induce LTD in “addicted” state; (3) Attenuation of postsynaptic potentials by cocaine; and (4) Reduced firing of medium spiny neurons in response to cocaine or dopamine. Findings made by this work are testable.
Collapse
|
22
|
Cruz Del Angel Y, Orfila JE, Herson PS, Brooks-Kayal A, González MI. Down-regulation of AMPA receptors and long-term potentiation during early epileptogenesis. Epilepsy Behav 2021; 124:108320. [PMID: 34592633 DOI: 10.1016/j.yebeh.2021.108320] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 08/24/2021] [Accepted: 08/31/2021] [Indexed: 10/20/2022]
Abstract
Epilepsy is a brain disorder characterized by the occurrence of recurrent spontaneous seizures. Behavioral disorders and altered cognition are frequent comorbidities affecting the quality of life of people with epilepsy. These impairments are undoubtedly multifactorial and the specific mechanisms underlying these comorbidities are largely unknown. Long-lasting alterations in synaptic strength due to changes in expression, phosphorylation, or function of α-amino-3-hydroxy-5-methylisoxazole-4-propionic acid receptors (AMPARs) have been associated with alterations in neuronal synaptic plasticity. In particular, alterations in hippocampal long-term potentiation (LTP), a well-accepted model of learning and memory, have been associated with altered cognition in epilepsy. Here, we analyzed the effects of pilocarpine-induced status epilepticus (SE) on AMPARs to determine if alterations in AMPAR signaling might be one of the mechanisms contributing to altered cognition during epilepsy. We found alterations in the phosphorylation and plasma membrane expression of AMPARs. In addition, we detected altered expression of GRIP, a key scaffolding protein involved in the proper distribution of AMPARs at the neuronal cell surface. Interestingly, a functional analysis revealed that these molecular changes are linked to impaired LTP. Together, these observations suggest that seizure-induced alterations in the molecular machinery regulating AMPARs likely impact the neuron's ability to support synaptic plasticity that is required for learning and memory.
Collapse
Affiliation(s)
- Yasmin Cruz Del Angel
- Department of Pediatrics, Division of Neurology and Translational Epilepsy Research Program, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - James E Orfila
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Paco S Herson
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Neuroscience Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Amy Brooks-Kayal
- Department of Pediatrics, Division of Neurology and Translational Epilepsy Research Program, University of Colorado School of Medicine, Aurora, CO 80045, USA; Neuroscience Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Children's Hospital Colorado, Aurora, CO 80045, USA
| | - Marco I González
- Department of Pediatrics, Division of Neurology and Translational Epilepsy Research Program, University of Colorado School of Medicine, Aurora, CO 80045, USA; Neuroscience Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
23
|
Wang D, Liu S, Wang G. Establishment of an Endocytosis-Related Prognostic Signature for Patients With Low-Grade Glioma. Front Genet 2021; 12:709666. [PMID: 34552618 PMCID: PMC8450508 DOI: 10.3389/fgene.2021.709666] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 08/09/2021] [Indexed: 12/15/2022] Open
Abstract
Background Low-grade glioma (LGG) is a heterogeneous tumor that might develop into high-grade malignant glioma, which markedly reduces patient survival time. Endocytosis is a cellular process responsible for the internalization of cell surface proteins or external materials into the cytosol. Dysregulated endocytic pathways have been linked to all steps of oncogenesis, from initial transformation to late invasion and metastasis. However, endocytosis-related gene (ERG) signatures have not been used to study the correlations between endocytosis and prognosis in cancer. Therefore, it is essential to develop a prognostic model for LGG based on the expression profiles of ERGs. Methods The Cancer Genome Atlas and the Genotype-Tissue Expression database were used to identify differentially expressed ERGs in LGG patients. Gene ontology, Kyoto Encyclopedia of Genes and Genomes, and Gene set enrichment analysis methodologies were adopted for functional analysis. A protein-protein interaction (PPI) network was constructed and hub genes were identified based on the Search Tool for the Retrieval of Interacting Proteins database. Univariate and multivariate Cox regression analyses were used to develop an ERG signature to predict the overall survival (OS) of LGG patients. Finally, the association between the ERG signature and gene mutation status was further analyzed. Results Sixty-two ERGs showed distinct mRNA expression patterns between normal brain tissues and LGG tissues. Functional analysis indicated that these ERGs were strikingly enriched in endosomal trafficking pathways. The PPI network indicated that EGFR was the most central protein. We then built a 29-gene signature, dividing patients into high-risk and low-risk groups with significantly different OS times. The prognostic performance of the 29-gene signature was validated in another LGG cohort. Additionally, we found that the mutation scores calculated based on the TTN, PIK3CA, NF1, and IDH1 mutation status were significantly correlated with the endocytosis-related prognostic signature. Finally, a clinical nomogram with a concordance index of 0.881 predicted the survival probability of LGG patients by integrating clinicopathologic features and ERG signatures. Conclusion Our ERG-based prediction models could serve as an independent prognostic tool to accurately predict the outcomes of LGG.
Collapse
Affiliation(s)
- Dawei Wang
- Shandong Academy of Clinical Medicine, Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.,Shandong Academy of Clinical Medicine, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Shiguang Liu
- Research Center of Translational Medicine, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Guangxin Wang
- Research Center of Translational Medicine, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Shandong Innovation Center of Intelligent Diagnosis, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
24
|
Harb A, Vogel N, Shaib A, Becherer U, Bruns D, Mohrmann R. Auxiliary Subunits Regulate the Dendritic Turnover of AMPA Receptors in Mouse Hippocampal Neurons. Front Mol Neurosci 2021; 14:728498. [PMID: 34497491 PMCID: PMC8419334 DOI: 10.3389/fnmol.2021.728498] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 08/02/2021] [Indexed: 12/30/2022] Open
Abstract
Different families of auxiliary subunits regulate the function and trafficking of native α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors in the central nervous system. While a facilitatory role of auxiliary subunits in ER export and forward trafficking of newly synthesized AMPA receptors is firmly established, it is unclear whether auxiliary subunits also control endosomal receptor turnover in dendrites. Here, we manipulated the composition of AMPA receptor complexes in cultured hippocampal neurons by overexpression of two auxiliary subunits, transmembrane AMPAR regulatory protein (TARP) γ-8 or cysteine knot AMPAR-modulating protein (CKAMP) 44a, and monitored dendritic receptor cycling in live-cell imaging experiments. Receptor surface delivery was assayed using a modified AMPA receptor subunit carrying the pH-dependent fluorophore superecliptic pHluorin (SEP-GluA1), which regains its fluorescence during receptor exocytosis, when transiting from the acidic lumen of transport organelles to the neutral extracellular medium. Strikingly, we observed a dramatic reduction in the spontaneous fusion rate of AMPA receptor-containing organelles in neurons overexpressing either type of auxiliary subunit. An analysis of intracellular receptor distribution also revealed a decreased receptor pool in dendritic recycling endosomes, suggesting that incorporation of TARPγ-8 or CKAMP44a in receptor complexes generally diminishes cycling through the endosomal compartment. To directly analyze dendritic receptor turnover, we also generated a new reporter by N-terminal fusion of a self-labeling HaloTag to an AMPA receptor subunit (HaloTag-GluA1), which allows for selective, irreversible staining of surface receptors. Pulse chase-experiments with HaloTag-GluA1 indeed demonstrated that overexpression of TARPγ-8 or CKAMP44a reduces the constitutive internalization rate of surface receptors at extrasynaptic but not synaptic sites. Thus, our data point to a yet unrecognized regulatory function of TARPγ-8 and CKAMP44a, by which these structurally unrelated auxiliary subunits delay local recycling and increase surface lifetime of extrasynaptic AMPA receptors.
Collapse
Affiliation(s)
- Ali Harb
- Zentrum für Human- und Molekularbiologie, Saarland University, Homburg, Germany.,Department of Anaesthesiology, University Medical Center Göttingen, Göttingen, Germany
| | - Nils Vogel
- Institute for Physiology, Otto-von-Guericke University, Magdeburg, Germany
| | - Ali Shaib
- Institute of Neurophysiology, University Medical Center Göttingen, Göttingen, Germany
| | - Ute Becherer
- Institute for Physiology, Center for Integrative Physiology and Molecular Medicine, Saarland University, Homburg, Germany
| | - Dieter Bruns
- Institute for Physiology, Center for Integrative Physiology and Molecular Medicine, Saarland University, Homburg, Germany
| | - Ralf Mohrmann
- Institute for Physiology, Otto-von-Guericke University, Magdeburg, Germany.,Center for Behavioral Brain Science, Otto-von-Guericke University, Magdeburg, Germany
| |
Collapse
|
25
|
Sathler MF, Khatri L, Roberts JP, Schmidt IG, Zaytseva A, Kubrusly RCC, Ziff EB, Kim S. Phosphorylation of AMPA receptor subunit GluA1 regulates clathrin-mediated receptor internalization. J Cell Sci 2021; 134:272078. [PMID: 34369573 DOI: 10.1242/jcs.257972] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 07/29/2021] [Indexed: 11/20/2022] Open
Abstract
Synaptic strength is altered during synaptic plasticity by controlling the number of AMPA receptors (AMPARs) at excitatory synapses. During long-term potentiation and synaptic up-scaling, AMPARs are accumulated at synapses to increase synaptic strength. Neuronal activity leads to phosphorylation of AMPAR subunit GluA1 and subsequent elevation of GluA1 surface expression, either by an increase in receptor forward trafficking to the synaptic membrane or a decrease in receptor internalization. However, the molecular pathways underlying GluA1 phosphorylation-induced elevation of surface AMPAR expression are not completely understood. Here, we employ fluorescence recovery after photobleaching (FRAP) to reveal that phosphorylation of GluA1 Serine 845 (S845) predominantly plays a role in receptor internalization than forward trafficking during synaptic plasticity. Notably, internalization of AMPARs depends upon the clathrin adaptor, AP2, which recruits cargo proteins into endocytic clathrin coated pits. In fact, we further reveal that an increase in GluA1 S845 phosphorylation by two distinct forms of synaptic plasticity diminishes the binding of the AP2 adaptor, reducing internalization, and resulting in elevation of GluA1 surface expression. We thus demonstrate a mechanism of GluA1 phosphorylation-regulated clathrin-mediated internalization of AMPARs.
Collapse
Affiliation(s)
- Matheus F Sathler
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, 540 First Avenue, New York, NY 10016, USA.,Department of Biomedical Sciences, 1617 Campus Delivery, Colorado State University, Fort Collins, CO, 80525, USA.,Neuroscience Program, Department of Physiology and Pharmacology, Rua São João Batista, 187, sala 428, Fluminense Federal University, Niterói, RJ, 24020-005, Brazil
| | - Latika Khatri
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, 540 First Avenue, New York, NY 10016, USA
| | | | | | | | - Regina C C Kubrusly
- Neuroscience Program, Department of Physiology and Pharmacology, Rua São João Batista, 187, sala 428, Fluminense Federal University, Niterói, RJ, 24020-005, Brazil
| | - Edward B Ziff
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, 540 First Avenue, New York, NY 10016, USA
| | - Seonil Kim
- Department of Biomedical Sciences, 1617 Campus Delivery, Colorado State University, Fort Collins, CO, 80525, USA.,Molecular, Cellular and Integrative Neurosciences Program
| |
Collapse
|
26
|
Rajgor D, Welle TM, Smith KR. The Coordination of Local Translation, Membranous Organelle Trafficking, and Synaptic Plasticity in Neurons. Front Cell Dev Biol 2021; 9:711446. [PMID: 34336865 PMCID: PMC8317219 DOI: 10.3389/fcell.2021.711446] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 06/14/2021] [Indexed: 12/16/2022] Open
Abstract
Neurons are highly complex polarized cells, displaying an extraordinary degree of spatial compartmentalization. At presynaptic and postsynaptic sites, far from the cell body, local protein synthesis is utilized to continually modify the synaptic proteome, enabling rapid changes in protein production to support synaptic function. Synapses undergo diverse forms of plasticity, resulting in long-term, persistent changes in synapse strength, which are paramount for learning, memory, and cognition. It is now well-established that local translation of numerous synaptic proteins is essential for many forms of synaptic plasticity, and much work has gone into deciphering the strategies that neurons use to regulate activity-dependent protein synthesis. Recent studies have pointed to a coordination of the local mRNA translation required for synaptic plasticity and the trafficking of membranous organelles in neurons. This includes the co-trafficking of RNAs to their site of action using endosome/lysosome “transports,” the regulation of activity-dependent translation at synapses, and the role of mitochondria in fueling synaptic translation. Here, we review our current understanding of these mechanisms that impact local translation during synaptic plasticity, providing an overview of these novel and nuanced regulatory processes involving membranous organelles in neurons.
Collapse
Affiliation(s)
- Dipen Rajgor
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Theresa M Welle
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Katharine R Smith
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, United States
| |
Collapse
|
27
|
Camblor-Perujo S, Kononenko NL. Brain-specific functions of the endocytic machinery. FEBS J 2021; 289:2219-2246. [PMID: 33896112 DOI: 10.1111/febs.15897] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 03/29/2021] [Indexed: 12/12/2022]
Abstract
Endocytosis is an essential cellular process required for multiple physiological functions, including communication with the extracellular environment, nutrient uptake, and signaling by the cell surface receptors. In a broad sense, endocytosis is accomplished through either constitutive or ligand-induced invagination of the plasma membrane, which results in the formation of the plasma membrane-retrieved endocytic vesicles, which can either be sent for degradation to the lysosomes or recycled back to the PM. This additional function of endocytosis in membrane retrieval has been adopted by excitable cells, such as neurons, for membrane equilibrium maintenance at synapses. The last two decades were especially productive with respect to the identification of brain-specific functions of the endocytic machinery, which additionally include but not limited to regulation of neuronal differentiation and migration, maintenance of neuron morphology and synaptic plasticity, and prevention of neurotoxic aggregates spreading. In this review, we highlight the current knowledge of brain-specific functions of endocytic machinery with a specific focus on three brain cell types, neuronal progenitor cells, neurons, and glial cells.
Collapse
Affiliation(s)
| | - Natalia L Kononenko
- CECAD Cluster of Excellence, University of Cologne, Germany.,Center for Physiology & Pathophysiology, Medical Faculty, University of Cologne, Germany
| |
Collapse
|
28
|
Long HZ, Cheng Y, Zhou ZW, Luo HY, Wen DD, Gao LC. PI3K/AKT Signal Pathway: A Target of Natural Products in the Prevention and Treatment of Alzheimer's Disease and Parkinson's Disease. Front Pharmacol 2021; 12:648636. [PMID: 33935751 PMCID: PMC8082498 DOI: 10.3389/fphar.2021.648636] [Citation(s) in RCA: 249] [Impact Index Per Article: 62.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Accepted: 03/08/2021] [Indexed: 12/12/2022] Open
Abstract
Alzheimer’s disease (AD) and Parkinson’s disease (PD) are two typical neurodegenerative diseases that increased with aging. With the emergence of aging population, the health problem and economic burden caused by the two diseases also increase. Phosphatidylinositol 3-kinases/protein kinase B (PI3K/AKT) signaling pathway regulates signal transduction and biological processes such as cell proliferation, apoptosis and metabolism. According to reports, it regulates neurotoxicity and mediates the survival of neurons through different substrates such as forkhead box protein Os (FoxOs), glycogen synthase kinase-3β (GSK-3β), and caspase-9. Accumulating evidences indicate that some natural products can play a neuroprotective role by activating PI3K/AKT pathway, providing an effective resource for the discovery of potential therapeutic drugs. This article reviews the relationship between AKT signaling pathway and AD and PD, and discusses the potential natural products based on the PI3K/AKT signaling pathway to treat two diseases in recent years, hoping to provide guidance and reference for this field. Further development of Chinese herbal medicine is needed to treat these two diseases.
Collapse
Affiliation(s)
- Hui-Zhi Long
- Department of Pharmacy, Cancer Institute, Phase I Clinical Trial Centre, Changsha Central Hospital Affiliated to University of South China, School of Pharmacy, University of South China, Changsha, China.,Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang, China
| | - Yan Cheng
- Department of Pharmacy, Cancer Institute, Phase I Clinical Trial Centre, Changsha Central Hospital Affiliated to University of South China, School of Pharmacy, University of South China, Changsha, China.,Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang, China
| | - Zi-Wei Zhou
- Department of Pharmacy, Cancer Institute, Phase I Clinical Trial Centre, Changsha Central Hospital Affiliated to University of South China, School of Pharmacy, University of South China, Changsha, China.,Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang, China
| | - Hong-Yu Luo
- Department of Pharmacy, Cancer Institute, Phase I Clinical Trial Centre, Changsha Central Hospital Affiliated to University of South China, School of Pharmacy, University of South China, Changsha, China.,Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang, China
| | - Dan-Dan Wen
- Department of Pharmacy, Cancer Institute, Phase I Clinical Trial Centre, Changsha Central Hospital Affiliated to University of South China, School of Pharmacy, University of South China, Changsha, China
| | - Li-Chen Gao
- Department of Pharmacy, Cancer Institute, Phase I Clinical Trial Centre, Changsha Central Hospital Affiliated to University of South China, School of Pharmacy, University of South China, Changsha, China.,Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang, China
| |
Collapse
|
29
|
Inhibition of AKT/GSK3β/CREB Pathway Improves the Responsiveness to AMPA Receptor Antagonists by Regulating GRIA1 Surface Expression in Chronic Epilepsy Rats. Biomedicines 2021; 9:biomedicines9040425. [PMID: 33919872 PMCID: PMC8103519 DOI: 10.3390/biomedicines9040425] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 04/10/2021] [Accepted: 04/13/2021] [Indexed: 12/15/2022] Open
Abstract
α-Amino-3-hydroxy-5-methylisoxazole-4-propionic acid receptor (AMPAR) has been reported as one of the targets for treatment of epilepsy. Although maladaptive regulation of surface expression of glutamate ionotropic receptor AMPA type subunit 1 (GRIA1) subunit is relevant to the responsiveness to AMPAR antagonists (perampanel and GYKI 52466) in LiCl-pilocarpine-induced chronic epilepsy rats, the underlying mechanisms of refractory seizures to AMPAR antagonists have yet been unclear. In the present study, we found that both AMPAR antagonists restored the up-regulations of GRIA1 surface expression and Src family-mediated glycogen synthase kinase 3β (GSK3β)-Ca2+/cAMP response element-binding protein (CREB) phosphorylations to control levels in responders (whose seizure activities were responsive to AMPAR) but not non-responders (whose seizure activities were uncontrolled by AMPAR antagonists). In addition, 3-chloroacetyl indole (3CAI, an AKT inhibitor) co-treatment attenuated spontaneous seizure activities in non-responders, accompanied by reductions in AKT/GSK3β/CREB phosphorylations and GRIA1 surface expression. Although AMPAR antagonists reduced GRIA2 tyrosine (Y) phosphorylations in responders, they did not affect GRIA2 surface expression and protein interacting with C kinase 1 (PICK1) protein level in both responders and non-responders. Therefore, our findings suggest that dysregulation of AKT/GSK3β/CREB-mediated GRIA1 surface expression may be responsible for refractory seizures in non-responders, and that this pathway may be a potential target to improve the responsiveness to AMPAR antagonists.
Collapse
|
30
|
Soykan T, Haucke V, Kuijpers M. Mechanism of synaptic protein turnover and its regulation by neuronal activity. Curr Opin Neurobiol 2021; 69:76-83. [PMID: 33744822 DOI: 10.1016/j.conb.2021.02.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/11/2021] [Accepted: 02/12/2021] [Indexed: 12/23/2022]
Abstract
Neurons are long-lived cells with a complex architecture, in which synapses may be located far away from the cell body and are subject to plastic changes, thereby posing special challenges to the systems that maintain and dynamically regulate the synaptic proteome. These mechanisms include neuronal autophagy and the endolysosome pathway, as well as the ubiquitin/proteasome system, which cooperate in the constitutive and regulated turnover of presynaptic and postsynaptic proteins. Here, we summarize the pathways involved in synaptic protein degradation and the mechanisms underlying their regulation, for example, by neuronal activity, with an emphasis on the presynaptic compartment and outline perspectives for future research. Keywords: Synapse, Synaptic vesicle, Autophagy, Endolysosome, Proteasome, Protein turnover, Protein degradation, Endosome, Lysosome.
Collapse
Affiliation(s)
- Tolga Soykan
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Robert-Rössle-Straße 10, 13125 Berlin, Germany
| | - Volker Haucke
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Robert-Rössle-Straße 10, 13125 Berlin, Germany; Freie Universität Berlin, Faculty of Biology, Chemistry, Pharmacy, 14195, Berlin, Germany.
| | - Marijn Kuijpers
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Robert-Rössle-Straße 10, 13125 Berlin, Germany.
| |
Collapse
|
31
|
Rebiai R, Givogri MI, Gowrishankar S, Cologna SM, Alford ST, Bongarzone ER. Synaptic Function and Dysfunction in Lysosomal Storage Diseases. Front Cell Neurosci 2021; 15:619777. [PMID: 33746713 PMCID: PMC7978225 DOI: 10.3389/fncel.2021.619777] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 02/12/2021] [Indexed: 11/13/2022] Open
Abstract
Lysosomal storage diseases (LSDs) with neurological involvement are inherited genetic diseases of the metabolism characterized by lysosomal dysfunction and the accumulation of undegraded substrates altering glial and neuronal function. Often, patients with neurological manifestations present with damage to the gray and white matter and irreversible neuronal decline. The use of animal models of LSDs has greatly facilitated studying and identifying potential mechanisms of neuronal dysfunction, including alterations in availability and function of synaptic proteins, modifications of membrane structure, deficits in docking, exocytosis, recycling of synaptic vesicles, and inflammation-mediated remodeling of synapses. Although some extrapolations from findings in adult-onset conditions such as Alzheimer's disease or Parkinson's disease have been reported, the pathogenetic mechanisms underpinning cognitive deficits in LSDs are still largely unclear. Without being fully inclusive, the goal of this mini-review is to present a discussion on possible mechanisms leading to synaptic dysfunction in LSDs.
Collapse
Affiliation(s)
- Rima Rebiai
- Department of Anatomy and Cell Biology, College of Medicine, The University of Illinois at Chicago, Chicago, IL, United States
| | - Maria I. Givogri
- Department of Anatomy and Cell Biology, College of Medicine, The University of Illinois at Chicago, Chicago, IL, United States
| | - Swetha Gowrishankar
- Department of Anatomy and Cell Biology, College of Medicine, The University of Illinois at Chicago, Chicago, IL, United States
| | - Stephania M. Cologna
- Department of Chemistry, College of Liberal Arts and Sciences, The University of Illinois at Chicago, Chicago, IL, United States
| | - Simon T. Alford
- Department of Anatomy and Cell Biology, College of Medicine, The University of Illinois at Chicago, Chicago, IL, United States
| | - Ernesto R. Bongarzone
- Department of Anatomy and Cell Biology, College of Medicine, The University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
32
|
Ji B, Skup M. Roles of palmitoylation in structural long-term synaptic plasticity. Mol Brain 2021; 14:8. [PMID: 33430908 PMCID: PMC7802216 DOI: 10.1186/s13041-020-00717-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 12/15/2020] [Indexed: 11/30/2022] Open
Abstract
Long-term potentiation (LTP) and long-term depression (LTD) are important cellular mechanisms underlying learning and memory processes. N-Methyl-d-aspartate receptor (NMDAR)-dependent LTP and LTD play especially crucial roles in these functions, and their expression depends on changes in the number and single channel conductance of the major ionotropic glutamate receptor α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) located on the postsynaptic membrane. Structural changes in dendritic spines comprise the morphological platform and support for molecular changes in the execution of synaptic plasticity and memory storage. At the molecular level, spine morphology is directly determined by actin cytoskeleton organization within the spine and indirectly stabilized and consolidated by scaffold proteins at the spine head. Palmitoylation, as a uniquely reversible lipid modification with the ability to regulate protein membrane localization and trafficking, plays significant roles in the structural and functional regulation of LTP and LTD. Altered structural plasticity of dendritic spines is also considered a hallmark of neurodevelopmental disorders, while genetic evidence strongly links abnormal brain function to impaired palmitoylation. Numerous studies have indicated that palmitoylation contributes to morphological spine modifications. In this review, we have gathered data showing that the regulatory proteins that modulate the actin network and scaffold proteins related to AMPAR-mediated neurotransmission also undergo palmitoylation and play roles in modifying spine architecture during structural plasticity.
Collapse
Affiliation(s)
- Benjun Ji
- Nencki Institute of Experimental Biology, 02-093, Warsaw, Poland.
| | - Małgorzata Skup
- Nencki Institute of Experimental Biology, 02-093, Warsaw, Poland.
| |
Collapse
|
33
|
Lessons learned from CHMP2B, implications for frontotemporal dementia and amyotrophic lateral sclerosis. Neurobiol Dis 2020; 147:105144. [PMID: 33144171 DOI: 10.1016/j.nbd.2020.105144] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 10/16/2020] [Accepted: 10/23/2020] [Indexed: 12/12/2022] Open
Abstract
Frontotemporal dementia (FTD) and Amyotrophic Lateral Sclerosis (ALS) are two neurodegenerative diseases with clinical, genetic and pathological overlap. As such, they are commonly regarded as a single spectrum disorder, with pure FTD and pure ALS representing distinct ends of a continuum. Dysfunctional endo-lysosomal and autophagic trafficking, leading to impaired proteostasis is common across the FTD-ALS spectrum. These pathways are, in part, mediated by CHMP2B, a protein that coordinates membrane scission events as a core component of the ESCRT machinery. Here we review how ALS and FTD disease causing mutations in CHMP2B have greatly contributed to our understanding of how endosomal-lysosomal and autophagic dysfunction contribute to neurodegeneration, and how in vitro and in vivo models have helped elucidate novel candidates for potential therapeutic intervention with implications across the FTD-ALS spectrum.
Collapse
|
34
|
O'Connor M, Shentu YP, Wang G, Hu WT, Xu ZD, Wang XC, Liu R, Man HY. Acetylation of AMPA Receptors Regulates Receptor Trafficking and Rescues Memory Deficits in Alzheimer's Disease. iScience 2020; 23:101465. [PMID: 32861999 PMCID: PMC7476873 DOI: 10.1016/j.isci.2020.101465] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 07/21/2020] [Accepted: 08/13/2020] [Indexed: 12/26/2022] Open
Abstract
In Alzheimer's disease (AD), decreases in the amount and synaptic localization of AMPA receptors (AMPARs) result in weakened synaptic activity and dysfunction in synaptic plasticity, leading to impairments in cognitive functions. We have previously found that AMPARs are subject to lysine acetylation, resulting in higher AMPAR stability and protein accumulation. Here we report that AMPAR acetylation was significantly reduced in AD and neurons with Aβ incubation. We identified p300 as the acetyltransferase responsible for AMPAR acetylation and found that enhancing GluA1 acetylation ameliorated Aβ-induced reductions in total and cell-surface AMPARs. Importantly, expression of acetylation mimetic GluA1 (GluA1-4KQ) in APP/PS1 mice rescued impairments in synaptic plasticity and memory. These findings indicate that Aβ-induced reduction in AMPAR acetylation and stability contributes to synaptopathy and memory deficiency in AD, suggesting that AMPAR acetylation may be an effective molecular target for AD therapeutics.
Collapse
Affiliation(s)
- Margaret O'Connor
- Department of Biology, Boston University, 5 Cummington Mall, Boston, MA 02215, USA
| | - Yang-Ping Shentu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Department of Pathology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Guan Wang
- Department of Biology, Boston University, 5 Cummington Mall, Boston, MA 02215, USA
| | - Wen-Ting Hu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhen-Dong Xu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiao-Chuan Wang
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Rong Liu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Heng-Ye Man
- Department of Biology, Boston University, 5 Cummington Mall, Boston, MA 02215, USA
- Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, 72 East Concord St., L-603, Boston, MA 02118, USA
- Center for Systems Neuroscience, Boston University, 610 Commonwealth Avenue, Boston, MA, USA
| |
Collapse
|
35
|
Mallmann RT, Klugbauer N. Genetic Inactivation of Two-Pore Channel 1 Impairs Spatial Learning and Memory. Behav Genet 2020; 50:401-410. [PMID: 32889694 PMCID: PMC7581579 DOI: 10.1007/s10519-020-10011-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 07/28/2020] [Indexed: 12/16/2022]
Abstract
Two-pore channels (TPCs) constitute a small family of cation channels that are localized in membranes of endosomal and lysosomal compartments. Although their roles for vesicular fusion and endolysosomal trafficking have been investigated, our knowledge on their expression pattern and higher order functions in the murine brain is still limited. Western blot analysis indicated a broad expression of TPC1 in the neocortex, cerebellum and hippocampus. In order to investigate the consequences of the genetic inactivation of TPC1, we performed a set of behavioural studies with TPC1−/− mice. TPC1−/− mice were analysed for an altered motor coordination and grip-strength, exploratory drive and anxiety as well as learning and memory. TPC1−/− mice did not show any differences in their exploratory drive or in their anxiety levels. There were also no differences in spontaneous activity or motor performance. However, the Morris water maze test uncovered a deficit in spatial learning and memory in TPC1−/− mice.
Collapse
Affiliation(s)
- Robert Theodor Mallmann
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Medizinische Fakultät, Albert-Ludwigs-Universität, Freiburg, Germany
| | - Norbert Klugbauer
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Medizinische Fakultät, Albert-Ludwigs-Universität, Freiburg, Germany. .,Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Medizinische Fakultät, Universität Freiburg, Albertstr. 25, 79104, Freiburg, Germany.
| |
Collapse
|
36
|
Zhang J, Li J, Yin Y, Li X, Jiang Y, Wang Y, Cha C, Guo G. Collapsin Response Mediator Protein 2 and Endophilin2 Coordinate Regulation of AMPA Receptor GluA1 Subunit Recycling. Front Mol Neurosci 2020; 13:128. [PMID: 32848595 PMCID: PMC7417516 DOI: 10.3389/fnmol.2020.00128] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 06/26/2020] [Indexed: 11/13/2022] Open
Abstract
The dynamic trafficking of AMPA receptors (AMPARs), which enables the endocytosis, recycling, and exocytosis of AMPARs, is crucial for synaptic plasticity. Endophilin2, which directly interacts with the GluA1 subunit of AMPARs, plays an important role in AMPAR endocytosis. Collapsin response mediator protein 2 (CRMP2) promotes the maturation of the dendritic spine and can transfer AMPARs to the membrane. Although the mechanisms of AMPAR endocytosis and exocytosis are well known, the exact molecular mechanisms underlying AMPAR recycling remain unclear. Here, we report a unique interaction between CRMP2 and endophilin2. Our results showed that overexpression of CRMP2 and endophilin2 increased the amplitude and frequency of miniature excitatory synaptic currents (mEPSCs) and modestly enhanced AMPAR levels in hippocampal neurons. Furthermore, the CRMP2 and endophilin2 overexpression phenotype failed to occur when the interaction between these two proteins was inhibited. Further analysis revealed that this interaction was regulated by CRMP2 phosphorylation. The phosphorylation of CRMP2 inhibited its interaction with endophilin2; this was mainly affected by the phosphorylation of Thr514 and Ser518 by glycogen synthase kinase (GSK) 3β. CRMP2 phosphorylation increased degradation and inhibited the surface expression of AMPAR GluA1 subunits in cultured hippocampal neurons. However, the dephosphorylation of CRMP2 inhibited degradation and promoted the surface expression of AMPAR GluA1 subunits in cultured hippocampal neurons. Taken together, our data demonstrated that the interaction between CRMP2 and endophilin2 was conductive to the recycling of AMPA receptor GluA1 subunits in hippocampal neurons.
Collapse
Affiliation(s)
- Jifeng Zhang
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou, China
| | - Jiong Li
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou, China
| | - Yichen Yin
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou, China.,Department of Neurology, Guangzhou Red Cross Hospital, Medical College, Jinan University, Guangzhou, China
| | - Xueling Li
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou, China
| | - Yuxin Jiang
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou, China
| | - Yong Wang
- Department of Forensic Science, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Caihui Cha
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou, China.,Department of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Guoqing Guo
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou, China
| |
Collapse
|
37
|
Germann UA, Alam JJ. P38α MAPK Signaling-A Robust Therapeutic Target for Rab5-Mediated Neurodegenerative Disease. Int J Mol Sci 2020; 21:E5485. [PMID: 32751991 PMCID: PMC7432772 DOI: 10.3390/ijms21155485] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 07/25/2020] [Accepted: 07/30/2020] [Indexed: 12/11/2022] Open
Abstract
Multifactorial pathologies, involving one or more aggregated protein(s) and neuroinflammation are common in major neurodegenerative diseases, such as Alzheimer's disease and dementia with Lewy bodies. This complexity of multiple pathogenic drivers is one potential explanation for the lack of success or, at best, the partial therapeutic effects, respectively, with approaches that have targeted one specific driver, e.g., amyloid-beta, in Alzheimer's disease. Since the endosome-associated protein Rab5 appears to be a convergence point for many, if not all the most prominent pathogenic drivers, it has emerged as a major therapeutic target for neurodegenerative disease. Further, since the alpha isoform of p38 mitogen-activated protein kinase (p38α) is a major regulator of Rab5 activity and its effectors, a biology that is distinct from the classical nuclear targets of p38 signaling, brain-penetrant selective p38α kinase inhibitors provide the opportunity for significant therapeutic advances in neurogenerative disease through normalizing dysregulated Rab5 activity. In this review, we provide a brief summary of the role of Rab5 in the cell and its association with neurodegenerative disease pathogenesis. We then discuss the connection between Rab5 and p38α and summarize the evidence that through modulating Rab5 activity there are therapeutic opportunities in neurodegenerative diseases for p38α kinase inhibitors.
Collapse
|
38
|
Deyts C, Clutter M, Pierce N, Chakrabarty P, Ladd TB, Goddi A, Rosario AM, Cruz P, Vetrivel K, Wagner SL, Thinakaran G, Golde TE, Parent AT. APP-Mediated Signaling Prevents Memory Decline in Alzheimer's Disease Mouse Model. Cell Rep 2020; 27:1345-1355.e6. [PMID: 31042463 DOI: 10.1016/j.celrep.2019.03.087] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 02/11/2019] [Accepted: 03/22/2019] [Indexed: 01/04/2023] Open
Abstract
Amyloid precursor protein (APP) and its metabolites play key roles in Alzheimer's disease (AD) pathophysiology. Whereas short amyloid-β (Aβ) peptides derived from APP are pathogenic, the APP holoprotein serves multiple purposes in the nervous system through its cell adhesion and receptor-like properties. Our studies focused on the signaling mediated by the APP cytoplasmic tail. We investigated whether sustained APP signaling during brain development might favor neuronal plasticity and memory process through a direct interaction with the heterotrimeric G-protein subunit GαS (stimulatory G-protein alpha subunit). Our results reveal that APP possesses autonomous regulatory capacity within its intracellular domain that promotes APP cell surface residence, precludes Aβ production, facilitates axodendritic development, and preserves cellular substrates of memory. Altogether, these events contribute to strengthening cognitive functions and are sufficient to modify the course of AD pathology.
Collapse
Affiliation(s)
- Carole Deyts
- Department of Neurobiology, The University of Chicago, 924 East 57th Street, Chicago, IL 60637, USA
| | - Mary Clutter
- Department of Neurobiology, The University of Chicago, 924 East 57th Street, Chicago, IL 60637, USA
| | - Nicholas Pierce
- Department of Neurobiology, The University of Chicago, 924 East 57th Street, Chicago, IL 60637, USA
| | - Paramita Chakrabarty
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, and McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Thomas B Ladd
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, and McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Anna Goddi
- Department of Neurobiology, The University of Chicago, 924 East 57th Street, Chicago, IL 60637, USA
| | - Awilda M Rosario
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, and McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Pedro Cruz
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, and McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Kulandaivelu Vetrivel
- Department of Neurobiology, The University of Chicago, 924 East 57th Street, Chicago, IL 60637, USA
| | - Steven L Wagner
- Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, USA; Veterans Affairs San Diego Healthcare System, La Jolla, CA 92161, USA
| | - Gopal Thinakaran
- Department of Neurobiology, The University of Chicago, 924 East 57th Street, Chicago, IL 60637, USA
| | - Todd E Golde
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, and McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Angèle T Parent
- Department of Neurobiology, The University of Chicago, 924 East 57th Street, Chicago, IL 60637, USA.
| |
Collapse
|
39
|
Hayashi T. Post-translational palmitoylation of ionotropic glutamate receptors in excitatory synaptic functions. Br J Pharmacol 2020; 178:784-797. [PMID: 32159240 DOI: 10.1111/bph.15050] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 02/07/2020] [Accepted: 03/05/2020] [Indexed: 12/17/2022] Open
Abstract
In the mammalian CNS, glutamate is the major excitatory neurotransmitter. Ionotropic glutamate receptors (iGluRs) are responsible for the glutamate-mediated postsynaptic excitation of neurons. Regulation of glutamatergic synapses is critical for higher brain functions including neural communication, memory formation, learning, emotion, and behaviour. Many previous studies have shown that post-translational protein S-palmitoylation, the only reversible covalent attachment of lipid to protein, regulates synaptic expression, intracellular localization, and membrane trafficking of iGluRs and their scaffolding proteins in neurons. This modification mechanism is extremely conserved in the vertebrate lineages. The failure of appropriate palmitoylation-dependent regulation of iGluRs leads to hyperexcitability that reduces the maintenance of network stability, resulting in brain disorders, such as epileptic seizures. This review summarizes advances in the study of palmitoylation of iGluRs, especially AMPA receptors and NMDA receptors, and describes the current understanding of palmitoylation-dependent regulation of excitatory glutamatergic synapses. LINKED ARTICLES: This article is part of a themed issue on Neurochemistry in Japan. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.4/issuetoc.
Collapse
Affiliation(s)
- Takashi Hayashi
- Section of Cellular Biochemistry, Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Kodaira, Tokyo, Japan
| |
Collapse
|
40
|
Yan Y, Yang H, Xie Y, Ding Y, Kong D, Yu H. Research Progress on Alzheimer's Disease and Resveratrol. Neurochem Res 2020; 45:989-1006. [PMID: 32162143 DOI: 10.1007/s11064-020-03007-0] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 02/27/2020] [Accepted: 03/03/2020] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD), a common irreversible neurodegenerative disease characterized by amyloid-β plaques, neurofibrillary tangles, and changes in tau phosphorylation, is accompanied by memory loss and symptoms of cognitive dysfunction. Increases in disease incidence due to the ageing of the population have placed a great burden on society. To date, the mechanism of AD and the identities of adequate drugs for AD prevention and treatment have eluded the medical community. It has been confirmed that phytochemicals have certain neuroprotective effects against AD. For example, some progress has been made in research on the use of resveratrol, a natural polyphenolic phytochemical, for the prevention and treatment of AD in recent years. Elucidation of the pathogenesis of AD will create a solid foundation for drug treatment. In addition, research on resveratrol, including its mechanism of action, the roles of signalling pathways and its therapeutic targets, will provide new ideas for AD treatment, which is of great significance. In this review, we discuss the possible relationships between AD and the following factors: synapses, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)-type glutamate receptors (AMPARs), silent information regulator 1 (SIRT1), and estrogens. We also discuss the findings of previous studies regarding these relationships in the context of AD treatment and further summarize research progress related to resveratrol treatment.
Collapse
Affiliation(s)
- Yan Yan
- The Department of Epidemiology and Health Statistics, Public Health School of Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Huihuang Yang
- The Department of Epidemiology and Health Statistics, Public Health School of Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Yuxun Xie
- The Department of Epidemiology and Health Statistics, Public Health School of Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Yuanlin Ding
- The Department of Epidemiology and Health Statistics, Public Health School of Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Danli Kong
- The Department of Epidemiology and Health Statistics, Public Health School of Guangdong Medical University, Dongguan, 523808, Guangdong, China.
| | - Haibing Yu
- The Department of Epidemiology and Health Statistics, Public Health School of Guangdong Medical University, Dongguan, 523808, Guangdong, China.
| |
Collapse
|
41
|
Guo H, Yuan K, Zhang Z, Xue Y, Yan W, Meng S, Zhu W, Wu P, Bao Y, Shi J, Zhang W, Lu L, Han Y. Pi4KIIα Regulates Unconditioned Stimulus-Retrieval-Induced Fear Memory Reconsolidation through Endosomal Trafficking of AMPA Receptors. iScience 2020; 23:100895. [PMID: 32088394 PMCID: PMC7038502 DOI: 10.1016/j.isci.2020.100895] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Revised: 01/06/2020] [Accepted: 02/04/2020] [Indexed: 11/30/2022] Open
Abstract
Targeting memory reconsolidation is an effective intervention for treating posttraumatic stress disorder (PTSD). Disrupting unconditioned stimulus (US)-retrieval-induced fear memory reconsolidation has become an effective therapeutic approach to attenuate fear memory, but the underlying molecular mechanisms remain unknown. Here, we report that US-retrieval-dependent increase in phosphatidylinositol 4-kinase IIα (Pi4KIIα) promotes early endosomal trafficking of AMPA receptors, leading to the enhancement of synaptic efficacy in basolateral amygdala (BLA) neurons. The inhibition of Pi4KIIα by an inhibitor or short hairpin RNA impaired contextual fear memory reconsolidation. This disruptive effect persisted for at least 2 weeks, which was restored by Pi4KIIα overexpression with TAT-Pi4KIIα. Furthermore, the blockade of early endosomal trafficking following US retrieval reduced synaptosomal membrane GluA1 levels and decreased subsequent fear expression. These data demonstrate that Pi4KIIα in the BLA is crucial for US-retrieval-induced fear memory reconsolidation, the inhibition of which might be an effective therapeutic strategy for treating PTSD. Unconditioned stimulus (US) retrieval induces a transient increase in Pi4KIIα expression Pi4KIIα regulates early endosomal trafficking of AMPARs during memory reconsolidation Pi4KIIα contributes to US-retrieval-induced synaptic enhancement in rat BLA Pi4KIIα inhibition after US retrieval impairs fear expression and shows long-term effects
Collapse
Affiliation(s)
- Hongling Guo
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), 51 Huayuan Bei Road, Haidian District, Beijing 100191, China
| | - Kai Yuan
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), 51 Huayuan Bei Road, Haidian District, Beijing 100191, China
| | - Zhongyu Zhang
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, 38 Xueyuan Road, Haidian District, Beijing 100191, China
| | - Yanxue Xue
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, 38 Xueyuan Road, Haidian District, Beijing 100191, China
| | - Wei Yan
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), 51 Huayuan Bei Road, Haidian District, Beijing 100191, China
| | - Shiqiu Meng
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, 38 Xueyuan Road, Haidian District, Beijing 100191, China
| | - Weili Zhu
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, 38 Xueyuan Road, Haidian District, Beijing 100191, China
| | - Ping Wu
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, 38 Xueyuan Road, Haidian District, Beijing 100191, China
| | - Yanping Bao
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, 38 Xueyuan Road, Haidian District, Beijing 100191, China
| | - Jie Shi
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, 38 Xueyuan Road, Haidian District, Beijing 100191, China
| | - Wen Zhang
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, 38 Xueyuan Road, Haidian District, Beijing 100191, China.
| | - Lin Lu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), 51 Huayuan Bei Road, Haidian District, Beijing 100191, China; Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China.
| | - Ying Han
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, 38 Xueyuan Road, Haidian District, Beijing 100191, China.
| |
Collapse
|
42
|
Ämmälä AJ, Urrila AS, Lahtinen A, Santangeli O, Hakkarainen A, Kantojärvi K, Castaneda AE, Lundbom N, Marttunen M, Paunio T. Epigenetic dysregulation of genes related to synaptic long-term depression among adolescents with depressive disorder and sleep symptoms. Sleep Med 2019; 61:95-103. [DOI: 10.1016/j.sleep.2019.01.050] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 01/18/2019] [Accepted: 01/23/2019] [Indexed: 12/14/2022]
|
43
|
Rogers EJ, Jada R, Schragenheim-Rozales K, Sah M, Cortes M, Florence M, Levy NS, Moss R, Walikonis RS, Palty R, Shalgi R, Lichtman D, Kavushansky A, Gerges NZ, Kahn I, Umanah GKE, Levy AP. An IQSEC2 Mutation Associated With Intellectual Disability and Autism Results in Decreased Surface AMPA Receptors. Front Mol Neurosci 2019; 12:43. [PMID: 30842726 PMCID: PMC6391579 DOI: 10.3389/fnmol.2019.00043] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 02/01/2019] [Indexed: 12/30/2022] Open
Abstract
We have recently described an A350V mutation in IQSEC2 associated with intellectual disability, autism and epilepsy. We sought to understand the molecular pathophysiology of this mutation with the goal of developing targets for drug intervention. We demonstrate here that the A350V mutation results in interference with the binding of apocalmodulin to the IQ domain of IQSEC2. We further demonstrate that this mutation results in constitutive activation of the guanine nucleotide exchange factor (GEF) activity of IQSEC2 resulting in increased production of the active form of Arf6. In a CRISPR generated mouse model of the A350V IQSEC2 mutation, we demonstrate that the surface expression of GluA2 AMPA receptors in mouse hippocampal tissue was significantly reduced in A350V IQSEC2 mutant mice compared to wild type IQSEC2 mice and that there is a significant reduction in basal synaptic transmission in the hippocampus of A350V IQSEC2 mice compared to wild type IQSEC2 mice. Finally, the A350V IQSEC2 mice demonstrated increased activity, abnormal social behavior and learning as compared to wild type IQSEC2 mice. These findings suggest a model of how the A350V mutation in IQSEC2 may mediate disease with implications for targets for drug therapy. These studies provide a paradigm for a personalized approach to precision therapy for a disease that heretofore has no therapy.
Collapse
Affiliation(s)
- Eli J Rogers
- Technion Faculty of Medicine, Technion Israel Institute of Technology, Haifa, Israel
| | - Reem Jada
- Technion Faculty of Medicine, Technion Israel Institute of Technology, Haifa, Israel
| | | | - Megha Sah
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, United States
| | - Marisol Cortes
- Department of Neurology, Johns Hopkins University, Baltimore, MD, United States
| | - Matthew Florence
- Department of Biopharmaceutical Sciences and Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Nina S Levy
- Technion Faculty of Medicine, Technion Israel Institute of Technology, Haifa, Israel
| | - Rachel Moss
- Technion Faculty of Medicine, Technion Israel Institute of Technology, Haifa, Israel
| | - Randall S Walikonis
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, United States
| | - Raz Palty
- Technion Faculty of Medicine, Technion Israel Institute of Technology, Haifa, Israel
| | - Reut Shalgi
- Technion Faculty of Medicine, Technion Israel Institute of Technology, Haifa, Israel
| | - Daniela Lichtman
- Technion Faculty of Medicine, Technion Israel Institute of Technology, Haifa, Israel
| | - Alexandra Kavushansky
- Technion Faculty of Medicine, Technion Israel Institute of Technology, Haifa, Israel
| | - Nashaat Z Gerges
- Department of Biopharmaceutical Sciences and Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Itamar Kahn
- Technion Faculty of Medicine, Technion Israel Institute of Technology, Haifa, Israel
| | - George K E Umanah
- Department of Neurology, Johns Hopkins University, Baltimore, MD, United States
| | - Andrew P Levy
- Technion Faculty of Medicine, Technion Israel Institute of Technology, Haifa, Israel
| |
Collapse
|